Archives

Type 2 Diabetes In Children and Adolescents – A Focus On Diagnosis and Treatment

ABSTRACT

Nearly three decades have passed since the first publications on type 2 diabetes (T2D) in children and adolescents, and it is now well established as a global problem. As the prevalence of obesity continues to increase within the general population, diagnosing T2D in adolescents presents a multifaceted challenge. In this chapter, we shall delve into the epidemiological aspects, as well as the distinct characteristics inherent to various types of diabetes. Cohort studies with long-term follow-ups have illuminated our understanding of the alarming incidence of complications in adolescents diagnosed with T2D. Recent approvals of novel pharmaceutical interventions for teenagers have ushered in a new era of hope. Hopefully, in the forthcoming decade, we anticipate a decline in the prevalence of these complications. However, in the interim, a proactive and assertive approach remains essential for addressing the complications associated with T2D in adolescents.

 

SHORT HISTORY

Youth-onset type 2 diabetes (T2D) was initially described in Pima Indian children and adolescents (1). This tribe, also known as “the pathfinders,” has a notable history of obesity and early-onset diabetes. In the mid-nineties, studies from various clinics in the United States reported cases of T2D in children from diverse ethnic backgrounds, primarily non-Hispanic Blacks and Hispanics (2-4). While these reports were initially met with suspicion, it soon became clear that a new disease had emerged among children. Subsequently, similar reports emerged from various parts of the world. In 2000, the SEARCH for Diabetes Study was launched in the US. This prospective nationwide, multi-center study aimed at understanding the epidemiology of both type 1 diabetes (T1D) and T2D in children. A few years later, in 2004, the Treatment Options for Type 2 Diabetes in Adolescents and Youth (TODAY) study was initiated. The TODAY study was a prospective, randomized treatment trial, designed to explore treatment regimens and the clinical course of T2D in youth. Registries from Germany, Diabetes Prospective Follow-up Registry (DPV) (5), Hong Kong (6), China (7), India (Registry of People with Diabetes with Youth Age at Onset (YDR)) (8) and Israel (9), contributed to the current knowledge. Importantly, while the US is leading in the scope of the T2D epidemic, similar trends have been observed worldwide.

 

EPIDEMIOLOGY

 

Epidemiology in the USA

 

According to the most recent data from the SEARCH study published in 2023, the adjusted incidence of T2DM among children and adolescents nearly doubled, from 9.0 to 17.9 cases per 100,000 persons per year, from 2002-03 to 2017-18 (10).

 

EPIDEMIOLOGY ACCORDING TO ETHNICITY

 

The incidence of T2D varies largely based on ethnicity, the highest incidence of T2D, 50.1 per 100,000 children age 10-20 years, was recorded in non-Hispanic Blacks, followed by Pima Indians (46), Hispanics (25.8), and Asian/Pacific Islanders (16.6). Among non-Hispanic Whites, the incidence is only 5.5 per 100,000. An annual increase was observed in all groups, but the highest increase was observed in Asian/Pacific Islanders (8.92%), followed by 7.17% in Hispanics and 5.99% in non-Hispanic Blacks, compared to 1.83% in non-Hispanic Whites.

 

EPIDEMIOLOGY ACCORDING TO GENDER AND AGE

The current incidence in females is 21.6 per 100,000 and in males 14.2; the temporal increase was similar in both genders. The incidence increases with age. One of the most remarkable findings in the SEARCH study is that among individuals 15-19 years of age, the incidence of T2D in 2017-2018 exceeded that of T1D (19.7 vs. 14.6 per 100,000). This is the first time the incidence of T2D surpassed that of T1D among youth, sounding alarming for other countries, which may have a lower incidence than the USA, but are showing steady increases.

Epidemiology in Europe

Although the overall incidence of T2D in Europe is much lower than in the US, a similar change over time has been reported. In Germany, a three-fold increase in the prevalence of T2D was reported for 10- to 19-year-olds between 2002 and 2020 (3.4 to 10.8 per 100,000) (5). Similar to the US, the estimated standardized prevalence of T2D was 1.4 times higher among girls (12.8) than boys (9.0). In the UK, the number of children registered as having T2D and being treated in pediatric diabetes units has risen by more than 50% in the past five years (11).

Epidemiology in Asia

Data retrieved from the Hong Kong Childhood Diabetes Registry revealed a threefold increase in T2D in children, from 1.27 per 100,000 in 1997-2007 to 3.42 per 100,000 in 2008-2018 (6). Data from China demonstrated an average annual increase of 26.6% in youth aged 10-19 years (12). There was no statistically significant difference in incidence between boys and girls. However, the risk of T2D was 1.49 times higher in urban areas than in rural areas.

 

RISK FACTORS FOR T2D IN CHILDREN

 

The increased risk of developing T2D in children is associated with genetics, an obesogenic environment, and obesity, along with the interactions among these factors (Figure 1). This visual representation emphasizes the multifaceted nature of risk factors for T2D in children and adolescents, illustrating the intricate connections between genetics, environmental factors, obesity, and the eventual development of T2D.

 

Figure 1. Risk Factors for T2D in Children and Adolescents. Risk factors for T2D in children and adolescents can be visualized as a series of interconnected circles, illustrating the complex interplay of various influences:
Environment (Outer Circle): The outermost circle represents the broader environmental factors that encompass everything from the intrauterine environment to residential neighborhood.
Genetics (Second Circle): Within the environment circle, genetics forms an important inner circle. It signifies the genetic predisposition that some individuals may have towards developing obesity and, consequently, T2D.
Obesity (Third Circle): Nested within the genetics circle is the obesity circle. It is influenced by multiple factors such as immigration, sedentary lifestyle, diet, microbiome, medication use, depression, and abuse. Of note, not everyone exposed to the obesogenic environment will become obese, indicating the influence of other factors.
Type 2 Diabetes (Innermost Circle): The innermost circle represents the development of T2D. Importantly, not all obese individuals will go on to develop early-onset T2D, highlighting the complexity of these interrelated risk factors.

Obesity

 

Severe obesity stands out as one of the most significant risk factors for youth-onset of T2D. Obesity is closely linked to the worsening of insulin resistance, a primary feature of the pathophysiology of T2D, along with progressive β-cell and α-cell dysfunction (13). In a recent meta-analysis comprising data from 30 studies involving 4688 children and adolescents, the prevalence of obesity at the time of T2D diagnosis was found to be 77 % (95% CI, 71%-83%) (14). Male participants exhibited higher odds of obesity than their female counterparts (odds ratio, 2.10; 95% CI, 1.3-3.3). These findings were sustained after several sensitivity analyses that excluded studies with uncertain or unspecified T2D diagnostic criteria, studies involving individuals with positive pancreatic autoantibodies, individuals presenting with weight loss, or those with genetically proven monogenic diabetes. While acknowledging the limitations of using BMI-based measures as a surrogate for obesity and considering the retrospective nature of the included studies, these data suggest that approximately 20-25% of adolescents with diabetes but without obesity may still have T2D. Furthermore, the definition of obesity needs to consider body composition variations among certain populations, such as East Asian and South Asian populations.

 

Family History of T2D

 

Most adolescents with T2D have a family member with the disease. In the TODAY study, 59.6% of adolescents with T2D had a first-degree family member with a history of diabetes, and 89% had a grandparent affected with diabetes (15). This increased risk of T2D associated with family history reflects genetic influences, the impact of the environment, and the intrauterine environment.

Maternal Obesity and Maternal T2D During Pregnancy

 

Exposure to maternal obesity has been linked to an increased risk of childhood obesity in offspring (16) and 2.8-fold higher odds of T2D. Exposure to maternal gestational diabetes was associated with 5.7-fold higher odds of T2D (17). Moreover, individuals exposed to maternal diabetes during pregnancy were diagnosed with diabetes at a younger age and exhibited worse β-cell function. Exposure in utero to maternal diabetes and obesity accounted for 47% of the T2D risk in this population (17,18), and, importantly, it sets up a vicious cycle for future generations. Suggested mechanisms affecting the developmental programming of offspring towards T2D include epigenetic modifications, alterations in stem cell differentiation, variation in the metabolome and microbiome, and immune dysregulation (19). Of note, assistedreproductive technology was not found to be a risk factor for early-onset T2D (20).

Pregnancies complicated by pre-existing diabetes are associated with extreme birth weights. In a large study, small for gestational age (SGA) babies, defined as below the 10th percentile for birth weight, were reported in 14.1% of pregnancies of women with T2D, while 26.2% had newborns with a large birth weight, defined as above the 90th percentile (21). Both SGA and high birth weight are associated with an increased risk of a history of early-onset T2D among adults. Of note, a lower birthweight was associated with an increased risk of developing T2D independently of adult BMI and the genetic risk of T2D (22).

 

Pregnancies complicated by pre-existing diabetes are associated with preterm delivery and birth weight extremes. In a national cohort study involving over 4 million singletons born in Sweden from 1973 to 2014, preterm birth (occurring before 37 weeks) was associated with a 1.3-fold increased risk of developing T2D before age 18 years (23). Notably, the association between preterm birth and T2D was significantly stronger among females (23).

 

Mental Health and Treatment

 

The association between psychiatric disorders in adolescents and T2D is bidirectional, complex, and not well studied in the pediatric population (24). On the one hand, youth with chronic illnesses such as obesity are more likely to develop depression, depressive symptoms, and anxiety than those without chronic illnesses. On the other hand, youth who have mental health morbidities are at increased risk for isolation, sedentary lifestyle, weight gain, and the development of T2D. Furthermore, some psychotropic medications, particularly atypical antipsychotics, are associated with weight gain and increased risk for T2D in adults. These associations have not been systematically studied in youth, but clinical experience suggests a contribution.

 

Immigration

 

Globally, immigration is on the rise. Studies conducted among adults have indicated that immigrants, when compared to non-immigrants, exhibit higher rates of obesity, insulin-resistance, and hypertension. In a recent review (25) focusing on European countries, non-European migrant children face a greater risk of being overweight or obese compared to their native counterparts; the prevalence of obesity in migrant and native children ranged from 1.2 to 15.4% and from 0.6 to 11.6%, respectively. The increased rates of overweight and obesity among migrating children and adolescents render them more susceptible to developing T2D and other metabolic abnormalities. An Israeli study found that among adolescents of Ethiopian origin, the prevalence of overweight and obesity increased two-and-a-half fold and fourfold in males and females, respectively, during the study period, compared to a 1.5-fold increase among native Israeli-bornmales and females (26).

 

Socioeconomic Status

 

A discernible disparity exists in the socioeconomic status (SES) of children and adolescents with T2D. In developed countries, such as the United States, low SES is a recognized risk factor for the development of obesity and T2D. This is attributed to factors such as limited access to healthy foods and reduced physical inactivity. However, in developing countries, higher SES groups tend to have greater levels of physical inactivity and consume higher quantities of fat, salt, and processed foods compared to their lower SES counterparts (Figure 2). When data from 384 youth with T2D who completed a baseline research visit as part of the SEARCH study and 227 youth with T2D from the Registry of People with Diabetes with Youth Age at Onset (YDR) in India who completed a baseline visit were compared, only 23.6% of SEARCH youth belonged to a high SES group, in contrast to 88.5% of YDR youths (P < .001) (8).

Figure 2. Global Disparities in Type 2 Diabetes Risk Factors. This illustrative figure offers insights into the diversity in risk factors for T2D among children across the world. It highlights how these risk factors vary across different regions and populations.
Gender Disparities (Upper Part of Figure): In many Western countries, the risk of developing T2D in children is higher in girls compared to boys. However, this pattern is not consistent globally. In other parts of the world, an equal or even higher risk of T2D has been documented in males. The upper portion of the figure visually represents these variations in risk, depicting the female-to-male ratios in different regions.
Socioeconomic Status (SES) Variation: (Lower Part of Figure) SES plays a significant role in T2D risk. In Western countries, lower SES is associated with an increased risk of T2D in children. Conversely, in developing countries, a contrary trend is observed, with a higher risk of T2D observed in populations with higher SES. These complex socioeconomic variations are highlighted in the figure, showcasing how T2D risk is influenced by economic factors on a global scale.

 

Data from 747 children and youths with T2D under 19 years of age, collected between 2009 and 2016 (from the population-based National Pediatric Diabetes Audit, covering over >95% of diabetes cases in England and Wales), revealed that under half of those with T2D reside in the most disadvantaged areas of England and Wales, and 41% fall into the most disadvantaged SES quintile (27). In contrast, in China, in Zhejiang, one of the most economically prosperous coastal provinces marked by industrialization and urbanization, changes in diet and decreased physical activity have led to a higher incidence of pediatric obesity. The mean annual incidence was 2.32 per 100,000 person-years in urban area compared with 1.44 in urban and rural area (12).


COVID-19 Pandemic


In a study conducted in the United States, the incidence of T2DM at 24 diabetes centers during the first year of the COVID-19 pandemic was examined (28). The average number of new diagnoses per year in the two years before the pandemic was 825 but rose to 1463 during the first pandemic year. This increase of 77% is significantly higher than the 5% expected annual increase in incidence observed in the two previous years. Similarly, a retrospective cross-sectional review of youths (age ≤ 21) diagnosed with T2D during the COVID-19 pandemic (from May 1, 2020, to April 30, 2021) and the five years preceding it (from May 1, 2015, to April 30, 2020) at a tertiary diabetes center revealed an increase of 293% (29).  Similar trends were reported from Germany (30). Data on T2D in adolescents during 2 years of the COVID-19 pandemic (2020–2021) were compared with the control period 2011–2019 in children aged 6 to <18 years were obtained from the DPV (German Diabetes Prospective Follow-up) Registry. The incidence of youth-onset T2D increased from 0.75 per 100,000 patient-years in 2011 to 1.25 per 100,000 in 2019, an annual increase of 6.8%. However, in 2021, the observed incidence was 1.95, significantly higher than expected. Of note, the observed incidence was significantly higher in boys (2.16), leading to a reversal of the sex ratio of pediatric T2D incidence (30).  The surge in T2D incidence during the pandemic can be attributed to several factors. First, there was an increase in obesity among young people during the COVID-19 pandemic, accompanied by increased consumption of processed foods and reduced physical activity, both of which contribute to the risk of developing T2D (31). In addition, there was increased psychosocial stress, which is emerging as an important contributor to the risk of T2D. Furthermore, viral mediated non-autoimmune β-cell destruction, resulting in reduced β-cell function in predisposed adolescents, has been suggested as a contributing factor (28).    

 

Genetics


The genetics of T2D in children and adolescents have largely remained unexplored. ProDiGY is a multiethnic collaboration encompassing three studies (TODAY, SEARCH, and T2D-GENES) involving 3,006 youth subjects with T2D, diagnosed at a mean age of 15.1±2.9 years, along with 6,061 diabetes-free adult control subjects. Association analyses were conducted on approximately 10 million imputed variants, employing a generalized linear mixed model incorporating a genetic relationship matrix to account for population structure. The analysis was further adjusted for sex. This comprehensive study identified seven genome-wide significant loci that included a novel locus in PHF2, along with TCF7L2, MC4R, CDC123, KCNQ1, IGF2BP2, and SLC16A11. A secondary analysis involving 856 diabetes-free youth subjects uncovered an additional locus in CPEB2, and consistent directional effects for diabetes risk were observed (32). 

CLINICAL PRESENTATION

Diabetes Related Signs

The classical signs at the onset of diabetes - polydipsia and polyuria - are observed in about two-thirds of youth at the diagnosis of T2D. Only about one-third are diagnosed through routine screening of asymptomatic youth with obesity. Furthermore, the prevalence of diabetic ketoacidosis (DKA) among youth with T2D in the SEARCH study and the YDR in India was 5.5% and 6.6%, respectively. Importantly, the incidence of DKA at the onset of T2D was higher during the SARS-CoV-2 pandemic (33). Hyperglycemic hyperosmolar state is present at diagnosis in 2% of youth with T2D (34).  

 

Seasonal Diagnosis of T2D

 

There is significant seasonal variation in the onset of T2D in children and young people in the United States; diagnoses increased in August. Possible explanations for an August peak in diagnoses include weight gain during the summer vacation and an increase in physical exams for school athletic programs that may detect asymptomatic hyperglycemia. The greater proportion of diagnoses made during routine health visits rather than following symptoms is consistent with this latter possibility. Similar seasonal variations in incidence are beginning to emerge in other countries.

 

Age of Onset of T2D

 

The incidence of T2D rises gradually during puberty, primarily due to the physiological insulin resistance characteristic of this period. However, illustrating the heterogeneity of T2D in children and adolescents, there was a significant difference between the mean age of onset of youth in the United States (Pediatric Diabetes Consortium, PDC) databases who were diagnosed at the mean age of 12 years compared to the mean age in Germany of 13 years (35).

 

PREPUBERTAL T2D

With the worldwide epidemic of childhood obesity on the rise, there are increasing reports of T2D occurring in prepubertal children. Initially, there were case reports on prepubertal children with T2D, including cases from Nigeria (36), and a 5-year-old Indigenous girl from Australia (37). Lately, several reports have described the onset of T2D in prepubertal children. A total of 42 children ≤ 10 years with T2DM were reported from Alabama (38), 12 from Australia (39), 35 prepubertal children from Houston, Texas (40), from San Antonio, Texas (41) and India (42). The common denominator of all reports of prepubertal children is extreme obesity, with a disproportionate impact on females and ethnic minorities as shown in Table 1. Additionally, there is a high prevalence of comorbidities.

Table 1. Characteristics of Prepubertal Children Diagnosed with T2D

 

San Antonio, Texas

N=20

Alabama, US

N=42

Australia

N=12

Houston, Texas.

N=35

Chennai, Tamil Nadu India

N=4

Age, years

Range

8.1

4-9

 

 

7-10

10.6± 2.5

 

5-8

10

8

 

 

 

9-10

10

34

 

 

 

Ethnicity

Hispanic 80%

African American 88(%)

 Caucasian 12%

Aboriginal Australians 92

Maori 8

 Hispanic 71

Black 23

Non Hispanic White 3%

India

Females (%)

75

88

75

51

100

BMI Z score

2.72

(1.7-5.0)

2.5 ± 0.4

2.38± 0.64

2.4±0.4

>97%ile

Hypertension (%)

NA

21

25

14

 

Dyslipidemia

 

NA

58

58%

100

 

NAFLD

NA

 

 

20

 

Microalbuminuria

NA

 

16.6

5

 

 

These reports suggest that there are unique features and pathophysiologic drivers distinct from the influence of pubertal hormones, which are often implicated in the mechanism underlying T2D in older children and adolescents. Furthermore, the emergence of T2D in prepubertal children and the high prevalence of comorbidities among them serve as a warning for an impending public health challenge.

Sex

 

In the Western world, youth-onset T2D is almost twice as common in girls as in boys, whereas Asian countries report no differences in incidence by sex. For T2D, SEARCH had a higher proportion of females, while studies from China show no difference between females and males, and there are reports of increased prevalence in males in the Middle East; in the UAE (43), the female to male ratio was 0.5:1, in Iraq (44) 0.6:1, and Kuwait (45) 0.8:1 (Figure 2). This may reflect the higher prevalence of obesity in males in these countries (46). Indeed, in the UAE, among children aged 11 to 14 years, the prevalence of obesity was reported as 24.3% and extreme obesity (BMI ≥99th percentile) as 5.7%; the rate of extreme obesity was 9.6-fold higher in boys than girls (47). In Kuwait, 41.4% of boys were classified as obese, compared to 28.9% of girls of the same age (46).

 

DIAGNOSIS

 

Early diagnosis of T2D is likely to bring several benefits, enabling prompt multifactorial treatment and management of cardiovascular risk factors. Considering the increasing prevalence of T2D among children and adolescents, clinical symptoms have become less distinct, and age, sex, and weight are no longer definitive criteria for a clear diagnosis. Therefore, the most important diagnostic feature is the presence or absence of pancreatic autoantibodies in any child diagnosed with hyperglycemia; positive antibodies indicate T1D even if the child is overweight or has a family history of T2D. If the antibodies are negative, a genetic test for monogenic diabetes (previously referred to as Maturity-Onset Diabetes of the Young (MODY) should be considered if available.

TREATMENT

 

Initial treatment for T2D includes a focus on lifestyle changes, including healthy diet, increasing regular physical activity, achieving weight loss, and receiving emotional support. While lifestyle change is recommended as a practical approach,there is limited evidence that lifestyle intervention alone has a sustained impact on glycemic control in youth with T2D (48).

 

The Impact of Lifestyle Changes

 

The impact of lifestyle intervention, as determined by changes in diet and cardiovascular fitness, on glycemic control in youth with T2D was assessed in the TODAY clinical trial cohort spanning 15 centers across the United States. The study included 699 youth aged 10 to 17 years with T2D for less than 2 years. Those participants randomly assigned to the lifestyle intervention participated in a family-based behavioral program aimed at promoting weight loss. Each family was assigned a dedicated leader responsible for guiding them through their journey of physical activity and nutrition improvement. The intervention was structured across three stages, initially there were weekly meetings for 6 to 8 months, focusing on physical activity, setting individual calorie intake goals, self-monitoring, and problem-solving. In the subsequent stage, biweekly meetings were held for 12 to 16 months. Lastly, meetings were held monthly for 24 to 28 months, concentrating on maintaining a healthy lifestyle. Dietary data were collected through an interviewer-administered food frequency questionnaire, and cardiovascular fitness was assessed using a submaximal cycle ergometer test at baseline, 6 months, and 24 months (49). At 6 months, approximately 25% of females and 33% of males improved cardiovascular fitness. The authors concluded that a minority of youth improved fitness and/or diet over time, although those who did showed a beneficial impact on glycemic outcomes. However, the positive lifestyle behavior changes did not persist after 24 months, providing a sobering perspective on the challenges of implementing lasting lifestyle changes in this population.

 

Pharmacotherapy

 

The beginning of the third decade of the 21st century will be marked by the introduction of novel medications for children with T2D. There are several groups of medications for T2D, biguanides, sulfonylureas, GLP1 receptor agonists, SGLT2 inhibitors, dipeptidyl peptidase inhibitors, and combinations of the dual incretin analogs and the new triple G agents (dual incretin and glucagon). However, only biguanides, GLP1 receptor agonists, and SGLT2 inhibitors are currently FDA-approved for children. Doses and modes of administration are presented in Table 2.

 

Table 2. FDA-Approved Medications for Treating Type 2 Diabetes in Children Aged 10 Years and Older

Biguanide

Metformin

Glucophage

p.o daily

850, 1000. up to 3000 day

GLP receptor agonist

Liraglutide

Victoza

s.c. daily

0.6mg
1.2mg
 1.8mg

Exenatide

Bydureon

s.c weekly

2 mg

Dulaglutide

Trulicity

s.c weekly

0.75mg/0.5mL

1.5mg/0.5mL

3mg/0.5mL

4.5mg/0.5mL

SGLT2 inhibitor

Empagliflozin

Jardiance

p.o daily

10 mg

25 mg

Dapagliflozin

Forxiga

p.o daily

5mg
10 mg

 

Five randomized control studies assessed the impact of these drugs in children with T2D. The inclusion criteria were consistent across all studies and included an age above 10 years, a BMI above the 85th percentile, and minor variations in HbA1c levels (between 7.0 and 11.0%, or 6.5% and 10%). The characteristics of the participants and outcomes are detailed in Table 3. Figure 3 depicts HbA1c results vs. placebo for each medication, and Figure 4 depicts the percent of individuals with HbA1c less than 7% at the end of each study.

 

Table 3. Effects of Various Medications on Glycemic and Anthropometric Parameters in Adolescents with Type 2 Diabetes After 26 Weeks of Treatment

 

Liraglutide

 

Exenatide

Dulaglutide
0.75 mg

Dulaglutide

1.5 mg

Empagliflozin

Dapagliflozin

Linagliptin

Sitagliptin

 

Tx

Pl

Tx

Pl

Tx

PL

Tx

PL

Tx

PL

Tx

PL

Tx

Pl

Tx

Pl

Number

66

68

59

24

52

51

52

51

52

5

39

33

53

53

95

95

Age (years)

14.6

14.6

15

16

14.7

14.2

14.7

14.2

14.6

14.4

16.1

14.4

14.6

14.4

14.3

13.7

Baseline HbA1c )%(

7.9

7.7

8.1

8.1

7.9

8.1

8.2

8.1

8.0

8.05

7.95

7.85

8.05

8.05

7.4

7.6

HbA1c change

-0.64

0.46

-0.36

0.49

-0.6

0.6

-0.9

0.6

-0.17

0.68

-0.25

0.5

0.33

0.68

-0.01

0.18

HbA1c ETD 

1.06

0.85

1.2

1.5

0.84

0.75

0.35

0.19

% HbA1c <7%

63.7

36.5

48

35.1

55

14

48

14

34.6

24.5

25

4

26.5

24.5

49.5

36.8

Baseline BMI (kg/m2)

34.6

33.3

36.9

35.4

33.6

34.3

34.3

34.3

35.5

36.1

31.3

33.6

36.5

36.1

0.0

-0.7

BMI ETD

-0.25

-0.21

ND

ND

-0.2

0

-0.1

0

ND

ND

-0.08

-0.11

ND

ND

-0.7

Weight change (kg)

-2.3

-0.99

-0.59

0.63

0.3

0.1

0.2

0.1

-0.79

-0.04

ND

ND

1.42

-0.04

-1.1

0.06

Weight ETD

-1.3

-1.2

0.2

0.1

-0.75

 

1.46

1.04

Systolic BP ETD

0.03

-2.9

3.8

2.5

-1.42

1.9

0.91

 

Diastolic BP

ETD

-1.08

ND

-0.8

-2.6

0.02

-0.5

1.5

 

Pl=placebo, Tx= treatment, ETD = estimated treatment difference, ND= no data, placebo group for Empagliflozin and Linagliptin was the same.

 

Figure 3. Change in HbA1c percentage points in adolescents with T2D under medication treatment compared with untreated control group. The differences in HbA1c percentage points from the baseline for adolescents diagnosed with T2D who received medication treatment (depicted in black) compared to control groups (shown in white with data points). In the medication-treated group, there is a significant decrease in HbA1c levels, denoted by the downward trend in the black bars. Conversely, in the control groups of all the studies, HbA1c levels increased, as indicated by the upward trend in the white bars with data points.

Figure 4. HbA1c Levels below 7% in adolescents with T2D. The percentage of adolescents who achieved HbA1c levels below 7% at the end of the follow-up period. A comparison is drawn between those treated with medication (depicted in black) and the control group (depicted in white).

 

METFORMIN

 

Metformin is a biguanide antihyperglycemic used in conjunction with diet and exercise to control glycemia. Metformin is considered the preferred first-line agent for treating T2D in pediatric patients 10 or older (50). Its onset of action is around 1.5 hours, and its total duration of action is 16-20 hours. Metformin exerts its glucose-lowering effect through various mechanisms, including inhibiting hepatic gluconeogenesis, increasing glucose uptake in skeletal muscles, reducing adipogenesis, and activating brown adipose tissue, leading to enhanced thermogenesis. Additionally, metformin decreases the absorption of glucose from the intestine and downregulates inflammation.

Although it is the first line of treatment for children and adolescents with T2D, prospective, randomized, controlled, long-term studies are limited. In a randomized control study involving 82 children and adolescents, 42 received metformin at doses up to 2000 mg/day, and 40 received placebo (51). After 16 weeks, mean HbA1c levels were lower in the metformin group compared to the placebo (7.5% and 8.6%, respectively). The metformin group also experienced a mean weight decrease of -1.5 kg compared with a mean increase of 0.9 kg in the placebo group. The mean BMI changed by -0.5 units in the treatment group vs. -0.4 units in the placebo group. In another study, metformin (500–1000 mg twice daily) was compared to glimepiride, a sulfonylurea (1–8 mg once daily), for 24 weeks (52). Both metformin (-0.71%) and glimepiride (-0.54%) groups showed significant reductions from baseline HbA1c levels. A total of 48.1% of metformin-treated and 42.4% of glimepride-treated participants achieved HbA1c levels below 7.0% at week 24. While the incidence of hypoglycemia was similar in both groups, metformin resulted in less weight gain compared to glimepiride. In a large observational study (53), 927 youth aged 13.7±2.0 years old, who had been diagnosed with T2D for a median of 2 months and had a baseline HbA1c of 7.7±2.2%, were treated with metformin. After a median of 71 days, the mean HbA1c change was -1.33, and the mean weight change -0.43 kg (53,54).

 

Metformin-related side effects are gastrointestinal, including diarrhea, abdominal pain, bloating, nausea, and decreased appetite, which occur in about 50% of users. Gastrointestinal side effects are usually transient and improve over time. Another potential side effect of metformin is vitamin B12 deficiency, and regular monitoring is indicated, though no cases of vitamin B12 deficiency were identified in the TODAY study. Lactic acidosis is a concerning side effect associated with metformin use in adults. It occurs in the presence of hypoperfusion or hypoxemia; however, it has rarely been reported in children and adolescents.

GLUCAGON-LIKE-PEPTIDE 1 (GLP1) 

 

Glucagon-like peptide-1 (GLP-1) is a peptide hormone primarily produced by intestinal enteroendocrine cells at low basal levels that rapidly increases within minutes of food consumption. GLP-1 plays a vital role in regulating meal-related glycemic excursions by enhancing insulin secretion and inhibiting glucagon secretion. Additionally, GLP-1 slows gastric emptying and reduces food intake through a central effect on appetite, which contributes to weight loss. However, GLP-1 has a short half-life of approximately 2 minutes, as it is rapidly degraded by dipeptidyl peptidase-4 (DPP-4). To enhance GLP-1 activity, medications such as DPP-4 inhibitors and GLP-1 receptor agonists have been developed.


The story of the development of GLP1 agonists is truly fascinating. It begins with the venomous Gila monster (Heloderma suspectum), native to New Mexico and Arizona. H. suspectum, a long-lived and reclusive species, spends a significant portion of its life underground. Although its venomous bite causes pain and weakness, it is rarely fatal to adult humans. In 1990, endocrinologist Dr. John Eng, while analyzing the venom to identify new hormones, identified a peptide named exendin-4. He discovered that exendin-4 had a remarkable ability to stimulate the synthesis and release of insulin from β-cells in the pancreas. Interestingly, exendin-4 closely resembled GLP-1. However, while GLP-1 remains active for about two minutes, the effect of exendin-4 persists for several hours. Preclinical studies demonstrated that a single daily injection of exendin-4 normalized blood glucose concentrations in mice with diabetes. Following extensive clinical testing, exenatide, an analog of exendin-4, was found to be safe and effective, leading to its FDA approval for adults in 2005.

Of note, in a 2-year rat carcinogenicity study involving prolonged-release exenatide, an increased incidence of thyroid adenoma and C-cell carcinoma was observed at doses ≥2-fold the human systemic exposure. The clinical relevance of these adverse findings is currently unknown. However, GLP-1 analogs are contraindicated in patients with a personal or family history of medullary thyroid carcinoma (MTC) or in patients with multiple endocrine neoplasia type 2 (MEN 2), as well as in patients with a serious hypersensitivity reaction. Patients should be counseled about the potential risk of MTC associated with the GLP1 receptor agonists, and it is important to inform them about the symptoms that may indicate thyroid tumors, such as a neck mass, dysphagia, dyspnea, and persistent hoarseness. Routine monitoring of serum calcitonin levels or use of thyroid ultrasound for the detection of MTC is of uncertain value.

 

Side effects include serious hypersensitivity reactions, including anaphylactic reactions and angioedema. Additionally, acute pancreatitis, including fatal and nonfatal hemorrhagic or necrotizing pancreatitis, and the acute onset of gallbladder disease, such as cholelithiasis or cholecystitis, have been reported in GLP-1 receptor agonists trials and post-marketing surveys (55).

 

Liraglutide-Victoza® 

 

Children aged 10 to less than 17 years with T2D were randomly assigned 1:1 to receive subcutaneous liraglutide (up to 1.8 mg per day) or placebo for a 26-week double-blind period, followed by a 26-week open-label extension (56). At the 26-week visit, the mean HbA1c had decreased by 0.64 percentage points with liraglutide and increased by 0.42 percentage points with placebo, resulting in an estimated treatment difference of -1.06 percentage points. By 52 weeks, the difference had increased to -1.30 percentage points (Table 3). In the liraglutide group, 63.7% of patients achieved HbA1c levels of less than 7.0%, compared with 36.5% in the placebo group. On the other hand, there was no statistical benefit of liraglutide over placebo in lowering BMI or blood pressure (Table 3). The overall rate of adverse events, including gastrointestinal adverse events, was higher among patients receiving liraglutide. The U.S. Food and Drug Administration approved liraglutide injection for the treatment of pediatric patients 10 years of age or older with T2D. Liraglutide was the first non-insulin drug approved to treat T2D in pediatric patients since metformin was approved for pediatric use in 2000.

Mode of administration: The recommended initial dose is 0.6 mg subcutaneously once daily for the first week, followed by an increase to 1.2 mg once daily. It’s important to note that the initial dose of 0.6 mg once daily is primarily intended to mitigate gastrointestinal adverse effects and does not provide glycemic control. If adequate glycemic control is not achieved, the dose can be further increased to 1.8 mg once daily.

 

Exenatide-Bydureon  

 

The effectiveness of once-weekly exenatide 2 mg (Bydureon AstraZeneca) in youth with suboptimally controlled T2D was evaluated (57). At 24 weeks, exenatide demonstrated superiority over the placebo in reducing HbA1c levels (a decrease of -0.36% with exenatide compared to +0.49% with placebo), resulting in a significant between-group difference of 0.85 percentage points. Notably, no major hypoglycemic events were reported. and the most frequently reported adverse events included gastrointestinal symptoms such as nausea, diarrhea, and vomiting, along with injection site reactions such as pruritus, erythema, and nodules. Clinically meaningful differences were not observed in change from baseline in BMI Z score, body weight, or waist circumference, nor were there significant differences in blood pressure. The prolonged-release suspension of exenatide administered via injection with a pre-filled pen has received approval from the European Union and the FDA for the treatment of T2DM in children and adolescents aged 10 years and older.

Mode of administration: Bydureon (2 mg per dose) should be administered once every 7 days. The dose can be administered at any time of day, with or without meals. Bydureon is not recommended as first-line therapy for patients with inadequate glycemic control on diet and exercise because of the uncertain relevance of the rat thyroid C-cell tumor findings to humans.

Dulaglutide-Trulicity

 

Dulaglutide (Trulicity) is a GLP-1 receptor agonist administered via subcutaneous injections once a week. In a randomized, double-blind, phase 3 trial involving youth with inadequately controlled T2DM, the efficacy and safety of dulaglutide treatment in reducing HbA1c levels at 26 weeks were assessed (58). Remarkably, there was 99% adherence to treatment in this trial. Treatment resulted in a significant reduction in HbA1c relative to placebo; at week 26, HbA1c decreased by 0.8 percentage points but increased by 0.6 percentage points in the placebo group (1.4 percentage point difference). Gastrointestinal symptoms were among the most common adverse events, but they were primarily mild and were most likely to occur soon after the initiation of therapy. There were no clinically meaningful differences in the incidence or annual rate of hypoglycemia between the dulaglutide groups and the placebo group. Body weight did not decrease significantly following treatment.

Mode of administration: The recommended initiating dose of Trulicity is 0.75 mg once weekly, administered subcutaneously. The dose may be increased to 1.5 mg once a week for additional glycemic control. Trulicity can be administered any time of day, with or without meals, and should be injected subcutaneously in the abdomen, thigh, or upper arm. The day of weekly administration can be changed, if necessary, as long as the last dose was administered 3 days (72 hours) or more before. Trulicity and insulin should not be mixed in the same syringe and must be administered as two separate injections at two different injection sites.

 

SODIUM-GLUCOSE TRANSPORT PROTEIN 2 - SGLT2 INHIBITORS

 

Sodium-glucose transport protein 2 (SGLT2) inhibitors belong to a class of medications that modulate sodium-glucose transport proteins in the nephron. These inhibitors primarily target the SGLT2 proteins, expressed in the renal proximal convoluted tubules, to reduce the reabsorption of filtered glucose and sodium (59). Apart from their role in controlling glucose concentrations, SGLT2 inhibitors have demonstrated substantial cardiovascular benefits, particularly reducing heart failure events, for adults with T2D. In adults, the FDA-approved indications for SGLT2 inhibitors include the reduction of major adverse cardiovascular events in individuals with T2D and established cardiovascular disease, as well as the reduction of the risk of eGFR decline in patients with chronic kidney disease at risk of progression. The most common reported adverse events associated with SGLT2 inhibitors include female genital mycotic infections, urinary tract infections (including urosepsis and pyelonephritis), as well as nausea and constipation. Most importantly, SGLT2 inhibitors are associated with an almost three-fold increased risk for DKA in adults. The risk for DKA is highest for canagliflozin, followed by empagliflozin and dapagliflozin. Furthermore, individuals may develop euglycemic DKA, i.e., a triad of increased anion gap acidosis, ketosis, and a serum glucose level below 250 mg/dL. The association between SGLT-2 inhibitors and euglycemic DKA appears to be secondary to their noninsulin-dependent glucose clearance, compensatory hyperglucagonemia, and volume depletion. Currently, euglycemic DKA has not been reported among youth with T2D taking SLGT2 inhibitors.

 

Empagliflozin - Jardiance® 

 

A total of 105 children and adolescents were randomly assigned 1:1 to receive empagliflozin 10 mg or placebo (60). The adjusted mean change in HbA1c from baseline at week 26 was significantly greater in the empagliflozin group (–0·84%). Changes in body weight and blood pressure did not reach statistical significance. Adverse events occurred in 64% of participants in the placebo group and 77% in the empagliflozin group up to week 26, including severe adverse events in 4% of participants in the placebo group and 2% in the empagliflozin group. Mild and moderate hypoglycemia were the most frequently reported adverse events, with higher rates for those on active drug treatment compared with placebo; no severe hypoglycemic events were reported, and there were no episodes of DKA or necrotizing fasciitis associated with empagliflozin treatment. An increased incidence of bone fractures has been reported in adults, leading the American Diabetes Association to recommend avoiding SGLT-2 inhibitors in patients with a risk factor for fractures. Furthermore, SGLT2 inhibitors are not recommended for glycemic lowering in patients with T2D with an eGFR less than 30 mL/min/1.73 m2.

Mode of administration: Empagliflozin is an oral medication dosed at either 10 mg daily or 25 mg daily. The recommended dose is 10 mg once daily in the morning, taken with or without food. If tolerated initially, dosing may increase up to 25 mg. 

 

Dapagliflozin - Farxiga®

 

Participants aged 10–24 years with T2D and HbA1c concentrations ranging from 6.5 to 11% were given oral dapagliflozin 10 mg or placebo during a 24-week double-blind period (61); 39 participants were assigned to dapagliflozin and 33 were assigned to placebo. After 24 weeks, the mean change in HbA1c concentration was −0·25% for dapagliflozin and 0·50% for the placebo group; the between-group difference was −0·75%, which did not reach statistical significance. However, in a sub-analysis, excluding participants with poor compliance, the HbA1c change was -0.51%, while it was 0.62% for the placebo group, resulting in a between group significant difference of 1.13%.

Adverse events occurred in 69% of participants assigned to dapagliflozin and 58% of those assigned to placebo over a 24-week period. Over 52 weeks, adverse events occurred in 74% of participants who received dapagliflozin. Hypoglycemia was noted in 28% of participants assigned to dapagliflozin and in 18% of those assigned to placebo, none were considered serious adverse events. Notably, there were no reported episodes of DKA.

Mode of administration: To improve glycemic control the recommended starting dose is 5 mg once daily, taken in the morning. Increase dose to 10 mg once daily in patients tolerating 5 mg who require additional glycemic control. Dapagliflozin is not recommended for use in patients receiving loop diuretics or who are volume depleted, e.g., due to acute illness (such as gastrointestinal illness).

 

DIPEPTIDYL PEPTIDASE 4 (DPP 4) INHIBITORS

 

DPP-4 is a ubiquitous enzyme that degrades GLP-1 and GIP (gastric inhibitory peptide), resulting in a short half-life. By inhibiting the DPP-4 enzyme, DPP-4 inhibitors increase the levels of GLP-1 and GIP. This, in turn, enhances beta-cell insulin secretion, thus reducing postprandial and fasting hyperglycemia (62).


Linagliptin (Tradjenta)

 

105 children and adolescents were randomly assigned 1:1 to oral linagliptin (5mg daily) or placebo (60).  The adjusted mean HbA1c change from baseline at week 26 was 0.33% for linagliptin and 0·68% for the placebo group; the difference was not statistically or clinically significant. None of the adverse events of special interest occurred with DPP-4 inhibitors, such as hypersensitivity reactions, angioedema, angioedema-like events and anaphylaxis, skin lesions, pancreatitis, or hepatic injury, were present. Linagliptin is not approved for adolescents with T2D (60).

 

Sitagliptin (Januvia)

 

A total of 190 children and adolescents were randomly assigned 1:1 to oral sitagliptin 100 mg daily or placebo (63). The adjusted mean change in HbA1c from baseline at week 20 was -0.01% for sitagliptin and 0·18% for the placebo group, resulting in a between-group difference of 0.19%, which was not significant. There was no significant difference in the percentage of participants with HbA1c below 7%, with 49.5% in the sitagliptin group and 36.8% in the placebo group. Notably, after 54 weeks, the HbA1c in the sitagliptin group decreased from 7.4% at baseline to 7.1%. Sitagliptin is not approved for adolescents with T2D (50).

 

ANTI-OBESITY PHARMACOTHERAPY FOR TREATMENT OF PEDIATRIC TYPE 2 DIABETES

 

As obesity is a significant risk factor for the development of T2D, the American Diabetes Association (ADA) recommends the use of anti-obesity medications as adjuvant therapy for adults with both T2D and overweight/obesity. In adults, adding anti-obesity medications to a diabetes regimen can improve glycemic control, reduce weight, and decrease the use of anti-diabetes medication (64). Semaglutide and tirzepatide have shown promise in reducing weight in adults with obesity and T2DM compared to liraglutide (65). Tirzepatide is a novel dual glucose-dependent insulinotropic polypeptide (GIP) and GLP-1 receptor agonist. A study to evaluate tirzepatide in pediatric and adolescent participants with T2D, who are inadequately controlled with metformin, or basal Insulin, or both (SURPASS-PEDS) is currently ongoing.

Liraglutide 3.0mg/day 

 

A higher dose of liraglutide of 3.0 mg/day (Saxenda®) is FDA-approved for obesity in youth aged 12 years and older who weigh ≥ 60kg or have an initial BMI ≥ 30kg/m2. The Satiety and Clinical Adipose-Liraglutide Evidence Trial in Adolescents with and without T2D (SCALE-Teens) was a double-blind, placebo-controlled RCT that randomized 251 adolescents to either liraglutide 3.0 mg/day or placebo. However, only two adolescents with T2D were enrolled. Consequently, no conclusive findings can be drawn regarding the impact of high-dose liraglutide on reducing BMI and improving HbA1c in young individuals with T2D. Since almost all adolescents with T2DM have overweight or obesity and could potentially meet the prescribing criteria, it is reasonable to speculate that the results might resemble those observed in adult trials for T2D, potentially leading to reductions in weight and BMI as well as improvements in hyperglycemia.

 

Semaglutide 2.4mg Weekly

 

Semaglutide in its once-weekly injectable GLP-1RA form at doses up to 2.4 mg weekly (Wegovy®) has shown significant weight loss in obese children and has now been approved for treatment of obesity in adolescents aged 12 years and older (66). The mean change in BMI from baseline to week 68 was −16.1% with semaglutide and 0.6% with placebo. Of note, HbA1c decreased 0.4 percentage points in the treatment group compared with 0.1 in the placebo. However, the impact on youth with T2D has not yet been studied.

 

Semaglutide injection up to 2.0 mg (Ozempic®) and oral semaglutide up to 14 mg (Rybelsus®) are approved for adults with T2D, but studies in youth with T2D have not yet been reported.

SUMMARY: WHICH TREATMENT DO I RECOMMEND TO MY PATIENT?

 

The answer to this pivotal question hinges on the art of personalized medicine. First, it is crucial to recognize that 50% of youth with T2D can maintain good glycemic control on metformin monotherapy, making this agent still an excellent choice for initial therapy. However, several measures can predict the likelihood of glycemic deterioration, including decreased insulin secretion, impaired insulin processing (elevated proinsulin/insulin ratio), and HbA1c concentration after a few months on monotherapy. Longitudinally, a rising proinsulin and an increase in HbA1c of more than 0.5 percentage points over any 6-month period are predictive of loss of glycemic control on monotherapy (67).

 

If these parameters suggest a risk for deterioration, the next step is to consider the therapeutic objective(s). Is it primarily elevated HbA1c? If so, the best approach may involve selecting a medication known for its efficacy in reducing HbA1c. Is obesity the central concern for the patient? In such cases, exploring pharmaceutical interventions tailored for weight management may be prudent. Does the individual have concerns about needle-based treatments? In such instances, favoring orally administered medications could offer a more suitable solution. If there is an elevated risk of cardiac or renal complications, it would be wise to consider options from the SGLT2 inhibitor group or GLP1 agonist group. In essence, treatment needs to be tailored to the unique needs and circumstances of each individual, ensuring that treatment decisions align with their specific health challenges and objectives.

 

BARIATRIC SURGERY

 

Weight reduction plays a crucial role in the management of T2D because weight loss is associated with improved insulin resistance and glycemic status. Considering the limited efficacy of lifestyle and pharmacologic interventions in treating severe obesity and obesity-related T2D, along with the remarkable weight reduction and remission of T2D observed in youth who undergo bariatric surgery the ADA recommends considering bariatric surgery for adolescents with T2D who have a BMI >35 kg/m2 (68). The two most common bariatric surgeries performed in adolescents with T2D are laparoscopic sleeve gastrectomy (LSG) and laparoscopic Roux-en-Y gastric bypass (LRYGB).

 

Impact of Bariatric Surgery on Glycemic Control

 

Studies have demonstrated improvement in HbA1c across all age groups. In one study, sixty-four pediatric patients diagnosed with morbid obesity and T2D underwent LSG surgery. The patients’ ages ranged from 5 to 14 years old. Their BMI decreased from 44.6±9.3 to 34.8 ± 9.6 kg/m2.Their HbA1c decreased from 6.0±0.8% prior to LSG surgery to 5.4±0.4%, 12 months post-surgery, a change of 10.9% (p = 0.001) (69). There was no significant difference in post-operative HbA1c between the age groups.

 

Impact of Bariatric Surgery on T2D Remission

 

Bariatric surgery in adolescents with T2D has a better outcome compared to medical treatment with metformin alone or in combination with rosiglitazone or intensive lifestyle intervention with insulin therapy given for glycemic progression (70). In a multicenter, nonrandomized, retrospective study of 202 obese adolescents with T2D (before the approval of new drugs for adolescents with T2D), 109 adolescents underwent surgery, and 93 adolescents received nonsurgical treatment (71(. In the surgery group, the remission rate for diabetes was 76%, compared to 6.5% in the medical treatment group. The remission was sustained for the two years of follow-up. Of note, LRYGB had better effects on weight loss and glycemic control than LSG.


Impact of Bariatric Surgery on Kidney Outcomes

 

In a three-year longitudinal study, kidney outcomes in youth following bariatric surgery were assessed. Improvement in albuminuria and GFR was observed in those with pre-operative kidney impairment. Among participants with a preoperative eGFR < 90 ml/min/1/73m2, eGFR improved from a mean of 76 ml/min/1.73m2 to 102 ml/min/1.73m2. Similarly, in those with an elevated albumin-to-creatinine ratio at baseline (ACR ≥30mg/g), the median ACR decreased from 74 mg/g to 17 mg/g. Estimated GFR and albuminuria remained stable in those without any evidence of pre-operative impairment. The effect of bariatric surgery on diabetic kidney disease was also investigated in adolescents with severe obesity and T2D five years after surgery compared to adolescents receiving medical management in the TODAY study (72). Elevated albuminuria was present in 21% of those receiving medical management at baseline, and it increased to 43% at 5 years. Conversely, albuminuria decreased from 27% prior to surgery to 5% at the 5-year follow-up. In adjusted analyses, the medical group had 27-fold higher odds of diabetic kidney disease at 5 years compared to Teen-LABS participants (72-73).


Impact of Bariatric Surgery on CVD Events

 

In another analysis, the risk for CVD events in two cohorts of adolescents with T2D and severe obesity undergoing medical or surgical treatment of T2D was assessed. Thirty adolescents who underwent bariatric surgery were matched to 63 who had received metformin alone or in combination with rosiglitazone or an intensive lifestyle intervention, with insulin therapy given for glycemic progression. The BMI of those who underwent bariatric surgery was 54.4±9.5 kg/m2, and that of the medical group was BMI 40.5±4.9 kg/m2. Although the baseline likelihood of CVD events was higher in the group that underwent bariatric surgery, one year after the surgery, the event risk was significantly lower and sustained at 5-year follow-up, whereas medical therapy was associated with an increase in risk among adolescents with T2D and severe obesity (74).

 

Adverse Effects of Bariatric Surgery

Bariatric surgery in adolescents can lead to various potential complications beyond the first postoperative month. The most commonly reported include gastroesophageal reflux, incisional hernia, treatment failure requiring operative revision, and nutritional deficits (50). Less frequent complications include stomach cancer, liver necrosis, gallbladder disorders, pancreatic disorders, acute kidney failure, neuromuscular complications, skin complications, and rarely mortality (51).

 

Summary

 

In summary, bariatric surgery currently outperforms medical management of T2D in adolescents and is the most efficacious treatment for youth-onset T2D, albeit with significant risks and economic implications. However, given the rapidly emerging medical therapies for weight reduction in adults and youth, many of which (e.g., dual-incretin analogs, triple-G agonists (75)) are now achieving degrees of weight loss approaching those seen following surgery, the individual roles of surgical and medical treatment for youth-onset T2D will need continued reassessment in the coming years.

 

COMPLICATIONS AND COMORBIDITIES AMONG ADOLESCENTS WITH T2D

 

 Large cohort studies investigating the incidence of complications in children and teenagers with T2D unequivocally demonstrated a higher incidence of complications, both compared to the incidence of complications in T2D in adults and in T1D in children. In a multicenter observational study conducted from 2011 to 2020, the cumulative incidence of diabetic complications was assessed in 500 adolescents who had participated in the TODAY study (76). After only 13 years from diagnosis, higher complication rates were observed compared to those reported for pediatric patients with T1DM or for adults with T2D. The first striking finding from this report is the participants’ poor glycemic control despite participation in a well-resourced clinical trial. While glycemic control in adolescents with T1DM tends to improve with age, a gradual deterioration occurred in adolescents with T2D throughout the follow-up period. Approximately 45.0% had HbA1c of at least 10%, and an additional 20% were in the range of 8-10%. Their BMI remained consistently in the range of 35.0 to 37.5 kg/m2. It is important to note that poor glycemic control and obesity are associated with early complications. The complications associated with T2D in children affect various systems (Figure 5).

 

Figure 5. Comprehensive Overview of Type 2 Diabetes (T2D) Complications in Adolescents. A comprehensive visual representation of the diverse range of complications affecting major physiological systems that can arise in adolescents with T2D.

 

On average, thirteen years after the initial diagnosis of T2D, incidence rates for hypertension, dyslipidemia, diabetic kidney disease, and neuropathy were 67.5%, 51.6%, 54.8%, and 32.4%, respectively. Additionally, retinopathy was present in 51% of the participants. One-fifth of the cohort (21.3%) had two complications, and 7.1% had three. There were seventeen serious cardiovascular events, including myocardial infarctions, six cases of congestive heart failure, three cases of coronary artery disease, and four stroke events. Additionally, six deaths were reported. This rapid development of complications was associated with severe insulin resistance and poor socioeconomic circumstances.

Hypertension

 

Hypertension, defined as blood pressure ≥ 95th percentile for age, sex, and height or systolic BP ≥ 130/80 mm Hg, is often present at the time of diagnosis in adolescents with T2D. In a study of 391 children from Hong Kong under 18 years of age, 22.5% had hypertension at diagnosis. In the TODAY study, 11.6% were hypertensive at the time of diagnosis (77) and the cumulative incidence of hypertension was 67.5% at a mean age of 26.4±2.8 years and 13.3±1.8 years from diagnosis (78). A systematic review and meta-analysis of 31 international studies on 4363 children with T2D aged 6.5 to 21 years at diagnosis found a pooled prevalence of hypertension of 25.33% (95% CI, 19.57%-31.53%) (79).Male participants had a higher hypertension risk than female participants (odds ratio 1.42 [95% CI, 1.10-1.83]).

 

Initial treatment involves lifestyle modification, aiming to not only enhance glycemic control but also incorporate a diet that is low in sodium and abundant in fruits, vegetables, whole grains, low-fat dairy products, and lean proteins. The Dietary Approaches to Stop Hypertension (DASH diet) is highly recommended for its effectiveness in reducing high blood pressure and supporting weight loss. In addition to lifestyle modification, the ADA recommends starting Angiotensin-Converting Enzyme inhibitors (ACEis) or Angiotensin Receptor Blockers (ARBs). Due to the potential teratogenic effects, the use of reliable contraception should be encouraged in individuals of childbearing age (68). There are no agents specifically approved for use in youth with T2D, however, a once-daily medication is recommended for better compliance (80,81).   

 

Nephropathy

 

The primary microvascular diabetic complication in adolescents with T2D is diabetic kidney disease (DKD), which develops in 25–40% of T2D patients and is associated with rapid progression and poor prognosis. The severity and risk of rapid deterioration in young people with T2D are reflected in the fact that microalbuminuria is reported at diagnosis among adolescents with T2D. In a recent systematic review and meta-analysis (79) that included 14 studies of 2250 children and adolescents with T2D, the prevalence of albuminuria was 22.17% (95%CI, 17.34%-27.38%), and the pooled prevalence of macroalbuminuria among 730 children and adolescents was 3.85% (79). In the SEARCH study (82) after a duration of 8 years, the prevalence was 19.9% among adolescents with T2D compared with 5.8% in those with T1D.

The accelerated development of kidney damage is attributed to inadequate glycemic control and the presence of various risk factors, including obesity, dyslipidemia, insulin resistance, hypertension, elevated serum uric acid, female sex, and the presence of chronic inflammation (83).

While albuminuria is the earliest clinical sign of diabetic kidney disease (DKD), the natural history of DKD begins with hyperfiltration, characterized by an increase in glomerular filtration rate (GFR) >120 mL/min/1.73 m² as a consequence of obesity and impaired glucose tolerance (84).  This increase predicts deterioration before other clinical signs appear. The second stage in the evolution of kidney dysfunction, still without clinical manifestations, is a mild reduction in GFR (60-89 mL/min/1.73 m²) (Figure 6). Structural changes of the kidney are typical at this stage, yet they are often reversible, making this a critical time for risk factor reduction (84).

 

Figure 6. Heat map depicting diabetes kidney disease (DKD) staging by GFR and albuminuria and suggested treatments. Adapted from Naaman SC, Bakris GLJDC. Diabetic Nephropathy: Update on Pillars of Therapy Slowing Progression. 2023;46:1574-86.

 

Treatment in these early stages involves achieving adequate glycemic control along with lifestyle modification, including increasing physical activity, improving sleep quality, and quitting smoking. Additionally, a healthy diet with lower salt and potassium intake, along with a low protein intake of 0.8 g/kg/day, is recommended (68). Pharmacological treatment includes control of blood pressure and managing dyslipidemia. Theoretically, medications that improve glycemic control would also provide benefits for preventing and/or treating DKD. GLP1 receptor agonists are reported to have a renal protective effect in adults with T2D. A recent meta-analysis of approximately 60,000 adults with T2DM treated with GLP-1 receptor agonist found a significant reduction in the composite kidney outcome (development of macroalbuminuria, doubling of serum creatinine, 40% or greater decline in eGFR, kidney replacement therapy, or death from kidney disease) when compared to placebo, as well as a trend towards a reduction in worsening kidney function (84). Similarly, the hemodynamic and natriuretic effects of SGLT2 inhibition are also postulated as protective mechanisms for DKD (85).However, studies in adolescents with T2D did not show an effect of systolic or diastolic blood pressure (Table 3). Of note, these studies involved relatively small groups of youth, and were short-term. Larger, long-term studies are needed, as well as additional therapeutic options for reducing the risk of DKD, such as mineralocorticoid receptor antagonists (MRAs), and endothelin receptor antagonists (ERAs).

Dyslipidemia

 

Screening for dyslipidemia should be conducted once optimal glycemic control has been achieved or within 3 months after T2D diagnosis. Initial cholesterol screening can be performed non-fasting. The recommended target values are triglycerides <150 mg/dL (1.7 mmol/L), HDL cholesterol >35 mg/dL (0.91 mmol/L), and LDL cholesterol <100 mg/dL (2.6 mmol/L). If these levels are outside the normal range, medical nutritional therapy (MNT) should be initiated, including restricting caloric intake from fat to 20–30% and daily cholesterol intake to <200 mg/day, avoiding trans fatty acids, limiting saturated fats to <7%, and controlling mono and poly unsaturated fats to 10-15%. If, after 6 months of dietary intervention, the LDL-C remains >130 mg/dL, statin therapy should be initiated, with a goal of achieving LDL-C <100 mg/dL. Currently, there are 7 approved statins for use in children and adolescents: Lovastatin, Simvastatin, Atorvastatin, and Fluvastatin [children ≥10 years old], Pitavastatin and Pravastatin [> 8 years], and Rosuvastatin [>6 years old]. It is important to note that due to potential teratogenic effects, individuals of childbearing age should receive reproductive counseling, and statins should be avoided in individuals of childbearing age who are not using reliable contraception. If triglyceride levels are greater than 400 mg/dL (4.7 mmol/L) in a fasting state or exceed 1,000 mg/dL (11.6 mmol/L) in a non-fasting state, it is imperative to optimize glycemic control and initiate fibrate therapy. This is essential for reducing the risk of pancreatitis.

 

In a study assessing whether clinicians are sufficiently aggressive in treating diabetes-related hyperlipidemia in youth, among 278 youth with T2D <10 years, 57% had LDL-C exceeding 100 mg/dL, 24% had LDL-C at or above 130 mg/dL, and 9% had LDL-C surpassing 160 mg/dL (86). Additionally, 29% had hypertriglyceridemia, and 44% had HDL-C levels below 40 mg/dL. However, only 5% of these youth were prescribed lipid-lowering medications.

 

Polycystic Ovarian Syndrome (PCOS)

 

There is a strong association between T2D in adolescent girls and polycystic ovary syndrome (PCOS). A systematic review and meta-analysis comprising 6 studies that involved 470 girls diagnosed with T2D, with an age range at diagnosis between 12.9 and 16.1 years, revealed that the prevalence of PCOS was estimated at 19.58% (87). PCOS is associated with a range of metabolic diseases, including hypertension and dyslipidemia. Furthermore, it is associated with a higher prevalence of cardiovascular risk factors, such as higher carotid intima thickness, β stiffness index, and reduced arterial compliance. Additionally, adolescent girls with PCOS have an increased likelihood of experiencing psychiatric comorbidities, such as anxiety and depression, which can negatively impact their health-related quality of life. A menstrual history should be taken on every girl with T2D at the diagnosis and at every follow-up visit. Metformin, in addition to lifestyle modification, is likely to improve menstrual cyclicity and hyperandrogenism in female individuals with T2D.

 

Pregnancy and Contraception

 

In the TODAY study, the maternal and offspring outcomes of young women with youth-onset T2D who experienced one or more pregnancies were evaluated (88). Over a span of up to 15 years, a total of 260 pregnancies were reported by 141 women. Their average age was 21.5±3.2 years, a mean BMI of 35.6±7.2 kg/m2, and an average diabetes duration of 8.1±3.2 years (88). Complications during pregnancy were identified in 65% of the women. Elevated HbA1c levels of ≥8% were observed in 31.9% of the pregnancies, and chronic hypertension complicated 35% of them. Pregnancy loss was observed in 25.3% and preterm birth occurred in 32.6% of pregnancies. Among the offspring, 7.8% of the babies were classified as small for gestational age, 26.8% as large for gestational age, and 17.9% fell into the macrosomic range. Other complications and congenital anomalies were noted in 10% of infants, including anencephaly, renal anomalies, and complications related to prematurity. The rate of known miscarriage for the entire study was 12.3%. The rate of known stillbirths in the cohort was 3%, which is more than triple the reported national rates.

 

The impact of T2D during pregnancy extends beyond the immediate prenatal and birth periods, exerting long-term effects on offsprings. Data pertaining to the diagnosis of diabetes in biological mothers were available for 621 participants in the TODAY study, of whom 301 had never been diagnosed with diabetes, 218 were diagnosed either before or during pregnancy, and 102 were diagnosed after pregnancy (89). For biological fathers, the data were available for 519 participants, with 352 having no history of diabetes and 167 having paternal diabetes. Notably, it was found that maternal diabetes, but not paternal diabetes, was associated with lower beta-cell function and deterioration of glycemic control over time. These effects remained significant even after accounting for variables such as age, sex, race/ethnicity, and household income (89).

 

Contraception is indicated in adolescent girls with T2D for several compelling reasons. Firstly, it is a recommendedtreatment for managing the symptoms of PCOS. Secondly, these young women are at increased risk of unintended pregnancy, which can lead to poor outcomes. Lastly, it is imperative for those treated with potentially teratogenic medications such as ACEis, ARBs, and statins. Unfortunately, despite the evident need, preconception counseling was reported only in 16.3% of women prior to their first pregnancy and only 14.9% used any method of contraception prior to the first pregnancy (88). This gap in reproductive health care highlights the importance of healthcare providers and support staff addressing this issue promptly and effectively.

 

Although in theory, adolescent girls with T2D without severe obesity, micro- or macrovascular disease, or other cardiovascular risk factors can consider a wide range of contraceptive methods, the practical reality often differs. A significant proportion of these individuals are afflicted by severe obesity, and a considerable number also exhibit elevated blood pressure. Consequently, when morbid obesity, severe hypertension, micro- or macrovascular disease, or multiple cardiovascular risk factors are present, it is advisable to prioritize nonhormonal or progestin-only contraceptive methods (Figure 7) (90).

 

Figure 7. Heat map of contraceptive treatment options for adolescent girls with T2D based on disease duration and complications. Adapted from Merino PM, Codner E. Contraception for Adolescents and Young Women with Type 2 Diabetes-Specific Considerations. Current diabetes reports 2022;22:77-84.

 

Diabetic Retinopathy 

 

Diabetic retinopathy (DR) is one of the most important causes of visual loss worldwide. DR is often asymptomatic until the very late stages. Given the potential for a rapid rate of progression and the effectiveness of new therapies in slowing deterioration, it is imperative to routinely screen individuals with diabetes for the early detection of retinal changes. It is important to note that direct fundoscopy may be less sensitive than 7-field stereoscopic fundus photography for detecting retinopathy (91). Diabetic retinopathy (DR) is divided into two major forms: non-proliferative and proliferative, according to the presence of abnormal new blood vessels emanating from the retina. Non-proliferative DR consists of microvascular abnormalities (including microaneurysms, occluded vessels, and dilated or tortuous vessels) primarily in the macula and posterior retina, intraretinal hemorrhages, hard exudates; and the variable presence of nerve-fiber layer infarcts (cotton wool spots). Visual loss is mainly due to the development of macular edema. Proliferative DR is marked by the presence of neovascularization arising from the disc and/or retinal vessels, resulting in preretinal and vitreous hemorrhage, subsequent fibrosis, and traction retinal detachment resulting in visual loss.

 

In a meta-analysis that included 27 studies involving 5924 children and adolescents (91) with T2D duration between 6.5-21.0 years, 1.11% had DR less than 2.5 years after T2D diagnosis. The prevalence of DR increased over time, with rates of 9.04% at 2.5 to 5.0 years after T2D diagnosis, and 28.14% more than 5 years after T2D diagnosis. The global prevalence of diabetic retinopathy in pediatric T2D was found to be 6.99%. Optical coherence tomography (OCT)performed in adolescents with T2D in the TODAY study revealed that changes in retinal thickness correlated with HbA1c, fasting glucose, and blood pressure, illustrating the importance of intensive control of hyperglycemia, hypertension, and hyperlipidemia.(92)

 

Among the systemic strategies for the prevention and treatment of diabetic retinopathy, two randomized clinical trials, "The Fenofibrate Intervention and Event Lowering in Diabetes (FIELD) Study and the Action to Control Cardiovascular Risk in Diabetes (ACCORD)-Eye Study, documented a reduction in the risk of non-proliferative DR progressing in individuals using fenofibrate (93). The protective effects of fenofibrate have been attributed to its antioxidative, anti-inflammatory, anti-apoptotic, and anti-angiogenic properties. Other treatments include pan-retinal photocoagulation, intravitreal anti-vascular endothelial growth factor (VEGF) therapy, integrin antagonists, and anti-inflammatory agents. Corticosteroids remain important second-line therapies for patients with macular edema.

 

Diabetic Neuropathy

 

DIABETIC PERIPHERAL NEUROPATHY

 

Diabetic peripheral neuropathy (DPN) is a cause of significant disability and poor quality of life. Signs and symptoms include sensory loss, paresthesia, and pain. However, subclinical signs of DPN may precede the development of frank neuropathic symptoms, and systematic screening is required to identify DPN in its earliest stages. Studies from USA (78,94), Canada (95), and India (96) reported the prevalence of DPN in adolescents with T2D assessed by examination for foot abnormalities, distal vibration perception, and ankle reflexes (97). In the SEARCH study, the prevalence of peripheral neuropathy assessed after 8 years in 258 adolescents with T2D was 22%, compared with 7% in T1DM (98). Data from the TODAY Study Group revealed that at baseline, 1.0% of the participants had nerve disease, and the cumulative incidence at 15 years was 32.4% (78). The risk factors associated with peripheral neuropathy in youth are longer duration of diabetes, older age, male sex, and smoking.  

Calcium channel a2δ ligands (gabapentin, pregabalin= Lyrica), serotonin- norepinephrine reuptake inhibitors, and tricyclic antidepressants are the most widely used medications for painful DN in adults. These medications are used to treat various presentations of neuropathic pain in children, but there have been no clinical studies devoted to painful DPN and no agents are licensed specifically for painful DN in childhood and youth (99). “Fortunately, painful DPN is relatively rare in children and youth.

AUTONOMIC NEUROPATHY 

 

Diabetic autonomic neuropathy (DAN) is a common form of neuropathy in individuals with diabetes mellitus characterized by dysfunction due to impairment of peripheral autonomic nerves, with a wide spectrum of manifestations (100). DAN that involves autonomic fibers of the enteric nervous system may cause upper and lower gastrointestinal symptoms, including gastroesophageal reflux disease, gastroparesis, and constipation. Additional manifestations include bladder and sexual dysfunction. DAN also involves the cardiovascular system. Cardiovascular autonomic neuropathy (CAN), secondary to the pathology of the autonomic nerve fibers that innervate the heart and blood vessels, results in resting tachycardia, exercise intolerance, orthostatic hypotension, syncope, and silent myocardial infarction and ischemia(101). A reduction in heart rate variability is an early sign of CAN and was reported in 47% of youth with T2D after a mean disease duration of only 1.7 years (102). In the SEARCH study, the prevalence of CAN assessed after duration of 8 years in 252 adolescents with T2D was 17% compared with 12% in T1DM (103). CAN was associated with elevated triglycerides and increased urinary albumin excretion.

 

Cardiovascular Complications 

 

T2D is a major risk factor for cardiovascular disease (CVD), including myocardial infarction and stroke. Markers for early CVD include increased arterial stiffness, carotid intima-media thickness, an alteration in the vascular endothelium, and cardiac autonomic neuropathy.
Arterial stiffness prevalence was significantly higher among youth with T2D than those with T1D (104). Arterial stiffness (measured by pulse wave velocity from carotid-femoral, femoral-foot, and carotid-radial), was measured in 388 youth with T2D at a mean age of 21 years and a diabetes duration of 7.7±1.5 years (105). Higher arterial stiffness was associated with an adverse change in left ventricular diastolic function. Similarly, cardiac parameters were assessed in 177 individuals with T2D, with a mean age of 24.6±4.2 years and a disease duration of 10.3±3.5 years (106). Of these, 72% were females, and their HbA1c levels averaged 8.9±1.9. Their parameters were compared to those of individuals with T1D. The prevalence of increased atrial stiffness was significantly higher in those with T2D, at 75.7%, compared to only 23.4% in individuals with T1D. In addition, the prevalence of cardiac autonomic neuropathy was significantly higher in those with T2D, at 16.9%, compared to 9.0% in individuals with T1D. Participants with T2D exhibited a higher left ventricular mass index, lower systolic function, and lower diastolic function compared to those with T1D.

Adolescents with T2DM have increased carotid intima-media thickness (cIMT) compared to obese and normal weight groups; the group difference was detected early at 14 years of age (107).  

Vascular endothelial function was lower in adolescents with T2D compared to normal weight and compared to adolescents with T1D (108).  These changes are of importance as the rate of all adjudicated heart, vascular, and cerebrovascular events was 3.73 per 1000 person-years (78). There were 17 serious cardiovascular events, myocardial infarction [4 events], congestive heart failure [6 events], coronary artery disease [3 events], and stroke [4 events].

 

Decreased Bone Mineral Density (BMD) 

 

In a study involving 17 adolescents newly diagnosed with T2D and 59 age, sex, and BMI-matched controls, bone mineral density (BMD) was assessed at multiple sites while accounting for potential confounding factors such as age, sex, Tanner stage, and BMI (109). The results revealed that BMD Z-scores for the femoral neck and bone mineral apparent density Z-scores of the lumbar spine were notably lower in individuals with T2D compared to their healthy counterparts.In another cross-sectional study of BMD in 180 youths aged 10 to 23 years, the participants were categorized into three groups: those with T2D were compared to those with obesity (n=226) and those with a healthy weight (BMI <85th percentile; n=238) (110). An age-dependent pattern emerged, wherein the BMD Z-score in children was higher in the T2D group compared to the obese group. However, in adolescents and young adults, the BMD Z-scores were lower in the T2D group when compared to the obese group. These findings suggest that T2D may have a detrimental impact on bone density, particularly around the age when individuals reach peak bone mass. Considering the elevated fracture risk observed in adults with T2D, the decrease in bone density at a young age raises concerns about potential long-term morbidity and fracture risk in adulthood.

Mental Health Comorbidity

 

DEPRESSION  

 

As part of routine diabetes care, a total of 197 adolescents and youth diagnosed with T2D, with a mean age of 16.9 years and 57% male, completed the Patient Health Questionnaire (PHQ) (111). 19.3% reported elevated depressive symptoms (PHQ score ≥10). Furthermore, in a sample of 53, 18.9% acknowledged thoughts of self-harm. Despite the prevalence of depressive symptoms, only 50.0% of those with depressive symptoms had a documented referral for mental health treatment in the electronic health record after the positive screening outcome. Older age, shorter diabetes duration, a higher HbA1c level, more blood glucose checks per day, and being prescribed oral medications were significantly associated with more depressive symptoms. Results from the TODAY study revealed that older teen girls had the highest rates of clinically significant depressive symptoms (112). Increased number of stressful life events were associated with elevated depressive symptoms. Depressive symptoms were correlated with low adherence to diabetes treatment, lower psychosocial functioning, and impaired health-related quality of life (QOL). In another study comparing PHQ between adolescents with T2D and T1D, the median PHQ-9 score in females with T2D was significantly greater than in females with T1D, but did not differ between males with T2D and T1D (113). The association between depression and T2D has been reviewed elsewhere (114).


DISORDERED EATING BEHAVIORS

 

In the TODAY study, out of 678 adolescents with T2D at a mean age of 14.0 years, 6% had clinical and 20% had subclinical levels of binge eating (115). Moreover, 50.3% of adolescents with T2D who are receiving insulin therapy had disordered eating behaviors (DEB) compared with 21.2% of those with T1DM (116). Disordered eating behavior is associated with poorer clinical outcomes and psychosocial well-being.

 

Mortality

 

In the SEARCH study, over a median follow-up of 5.3 years, the mortality rate among individuals with T1D was 70.6 deaths per 100,000 patient-years, compared with 185.6 deaths per 100,000 patients-years for those with T2D (117)..When compared to the general populations of US states, it was observed that the mortality rate for individuals with T2D was significantly higher than expected while this was not the case for individuals with T1DM. Females had higher mortality rates than males. Among adolescents with T2D, the leading underlying cause of death was attributed to transport/motor vehicle accidents followed by accidental poisoning, and intentional self-harm. According to a life expectancy model, it was predicted that youth with T2DM lose approximately 15 years (118).

 

CONCLUSION

 

A summary of the key points is presented in Table 4.

 

Table 4. Key Points

•           The prevalence of type 2 diabetes (T2D) varies across different countries and regions globally, but there is a consistent upward trend in its occurrence. Large-scale national studies in countries like Sweden, Germany, and India have reported similar increasing trends.

•           Contrary to common perception, not all adolescents with T2D are severely obese. Only 70-80% fall into this category.

•           Socioeconomic status plays a significant role in the prevalence of T2D, with lower socioeconomic status being associated with higher incidence in Western countries, while the opposite is observed in low-income countries.

•           Although adolescents are at higher risk of developing T2D, there are growing reports of T2D cases in children younger than 10 years old.

•           The gender distribution of T2D varies, with a higher proportion of cases in females in Western countries but a higher proportion in males in the Middle East and China.

•           Promisingly, there are new medications designed for weight loss, which hold the potential to reduce the high rates of complications associated with T2D in children.

•           There are low prescription rates for statins and contraception in the management of T2D in adolescents, highlighting potential gaps in medical care.

•           Many complications of T2D can potentially be reversed if detected and treated aggressively in their early stages.

 

 

ACKNOWLEGEMENTS

 

The authors extend their heartfelt gratitude to the gifted illustrator, Mrs Noah Levek, for her exceptional eye-opening illustrations.

 

REFERENCES

 

  1. Knowler WC, Pettitt DJ, Savage PJ, Bennett PH. Diabetes incidence in Pima indians: contributions of obesity and parental diabetes. Am J Epidemiol 1981; 113:144-156
  2. Pinhas-Hamiel O, Dolan LM, Daniels SR, Standiford D, Khoury PR, Zeitler P. Increased incidence of non-insulin-dependent diabetes mellitus among adolescents. J Pediatr 1996; 128:608-615
  3. Glaser NS, Jones KL. Non-insulin dependent diabetes mellitus in Mexican-American children. West J Med 1998; 168:11-16
  4. Neufeld ND, Raffel LJ, Landon C, Chen YD, Vadheim CM. Early presentation of type 2 diabetes in Mexican-American youth. Diabetes Care 1998; 21:80-86
  5. Stahl-Pehe A, Kamrath C, Prinz N, Kapellen T, Menzel U, Kordonouri O, Schwab KO, Bechtold-Dalla Pozza S, Rosenbauer J, Holl RW. Prevalence of type 1 and type 2 diabetes in children and adolescents in Germany from 2002 to 2020: A study based on electronic health record data from the DPV registry. J Diabetes 2022; 14:840-850
  6. Tung JY, Kwan EY, But BW, Wong WH, Fu AC, Pang G, Tsang JW, Yau HC, Belaramani K, Wong LM, Wong SM, Lo P, Ng KL, Yeung WK, Chan KT, Chan AM, Wong SW, Tay MK, Chung J, Lee CY, Lam YY, Cheung PT. Incidence and clinical characteristics of pediatric-onset type 2 diabetes in Hong Kong: The Hong Kong childhood diabetes registry 2008 to 2017. Pediatr Diabetes 2022; 23:556-561
  7. Urakami T, Miyata M, Yoshida K, Mine Y, Kuwabara R, Aoki M, Suzuki J. Changes in annual incidence of school children with type 2 diabetes in the Tokyo Metropolitan Area during 1975-2015. Pediatr Diabetes 2018; 19:1385-1392
  8. Hockett CW, Praveen PA, Ong TC, Amutha A, Isom SP, Jensen ET, D'Agostino RB, Jr., Hamman RF, Mayer-Davis EJ, Lawrence JM, Pihoker C, Kahn MG, Mohan V, Tandon N, Dabelea D. Clinical profile at diagnosis with youth-onset type 1 and type 2 diabetes in two pediatric diabetes registries: SEARCH (United States) and YDR (India). Pediatr Diabetes 2021; 22:22-30
  9. Zuckerman Levin N, Cohen M, Phillip M, Tenenbaum A, Koren I, Tenenbaum-Rakover Y, Admoni O, Hershkovitz E, Haim A, Mazor Aronovitch K, Zangen D, Strich D, Brener A, Yeshayahu Y, Schon Y, Rachmiel M, Ben-Ari T, Levy-Khademi F, Tibi R, Weiss R, Lebenthal Y, Pinhas-Hamiel O, Shehadeh N. Youth-onset type 2 diabetes in Israel: A national cohort. Pediatr Diabetes 2022; 23:649-659
  10. Wagenknecht LE, Lawrence JM, Isom S, Jensen ET, Dabelea D, Liese AD, Dolan LM, Shah AS, Bellatorre A, Sauder K, Marcovina S, Reynolds K, Pihoker C, Imperatore G, Divers J, study SfDiY. Trends in incidence of youth-onset type 1 and type 2 diabetes in the USA, 2002-18: results from the population-based SEARCH for Diabetes in Youth study. The lancet Diabetes & endocrinology 2023; 11:242-250
  11. Wise J. Type 2 diabetes: Charity warns of "perfect storm" putting more children at risk. BMJ (Clinical research ed)2022; 377:o1451
  12. Wu H, Zhong J, Yu M, Wang H, Gong W, Pan J, Fei F, Wang M, Yang L, Hu R. Incidence and time trends of type 2 diabetes mellitus in youth aged 5-19 years: a population-based registry in Zhejiang, China, 2007 to 2013. BMC pediatrics 2017; 17:85
  13. Biondi G, Marrano N, Borrelli A, Rella M, Palma G, Calderoni I, Siciliano E, Lops P, Giorgino F, Natalicchio A. Adipose Tissue Secretion Pattern Influences &beta;-Cell Wellness in the Transition from Obesity to Type 2 Diabetes. 2022; 23:5522
  14. Cioana M, Deng J, Nadarajah A, Hou M, Qiu Y, Chen SSJ, Rivas A, Banfield L, Toor PP, Zhou F, Guven A, Alfaraidi H, Alotaibi A, Thabane L, Samaan MC. The Prevalence of Obesity Among Children With Type 2 Diabetes: A Systematic Review and Meta-analysis. JAMA Network Open 2022; 5:e2247186-e2247186
  15. Copeland KC, Zeitler P, Geffner M, Guandalini C, Higgins J, Hirst K, Kaufman FR, Linder B, Marcovina S, McGuigan P, Pyle L, Tamborlane W, Willi S. Characteristics of Adolescents and Youth with Recent-Onset Type 2 Diabetes: The TODAY Cohort at Baseline. The Journal of Clinical Endocrinology & Metabolism 2011; 96:159-167
  16. Lowe WL, Jr., Lowe LP, Kuang A, Catalano PM, Nodzenski M, Talbot O, Tam WH, Sacks DA, McCance D, Linder B, Lebenthal Y, Lawrence JM, Lashley M, Josefson JL, Hamilton J, Deerochanawong C, Clayton P, Brickman WJ, Dyer AR, Scholtens DM, Metzger BE. Maternal glucose levels during pregnancy and childhood adiposity in the Hyperglycemia and Adverse Pregnancy Outcome Follow-up Study. Diabetologia 2019; 62:598-610
  17. Dabelea D, Mayer-Davis EJ, Lamichhane AP, D'Agostino RB, Jr., Liese AD, Vehik KS, Narayan KM, Zeitler P, Hamman RF. Association of intrauterine exposure to maternal diabetes and obesity with type 2 diabetes in youth: the SEARCH Case-Control Study. Diabetes Care 2008; 31:1422-1426
  18. Shah AS, Nadeau KJ, Dabelea D, Redondo MJ. Spectrum of Phenotypes and Causes of Type 2 Diabetes in Children. Annual review of medicine 2022; 73:501-515
  19. Rughani A, Friedman JE, Tryggestad JB. Type 2 Diabetes in Youth: the Role of Early Life Exposures. Current diabetes reports 2020; 20:45
  20. Norrman E, Petzold M, Gissler M, Spangmose AL, Opdahl S, Henningsen AK, Pinborg A, Tiitinen A, Rosengren A, Romundstad LB, Wennerholm UB, Bergh C. Cardiovascular disease, obesity, and type 2 diabetes in children born after assisted reproductive technology: A population-based cohort study. PLoS medicine 2021; 18:e1003723
  21. Murphy HR, Howgate C, O'Keefe J, Myers J, Morgan M, Coleman MA, Jolly M, Valabhji J, Scott EM, Knighton P, Young B, Lewis-Barned N. Characteristics and outcomes of pregnant women with type 1 or type 2 diabetes: a 5-year national population-based cohort study. The lancet Diabetes & endocrinology 2021; 9:153-164
  22. Wibaek R, Andersen GS, Linneberg A, Hansen T, Grarup N, Thuesen ACB, Jensen RT, Wells JCK, Pilgaard KA, Brøns C, Vistisen D, Vaag AA. Low birthweight is associated with a higher incidence of type 2 diabetes over two decades independent of adult BMI and genetic predisposition. Diabetologia 2023;
  23. Crump C, Sundquist J, Sundquist K. Preterm birth and risk of type 1 and type 2 diabetes: a national cohort study. Diabetologia 2020; 63:508-518
  24. Nouwen A, Adriaanse MC, van Dam K, Iversen MM, Viechtbauer W, Peyrot M, Caramlau I, Kokoszka A, Kanc K, de Groot M, Nefs G, Pouwer F. Longitudinal associations between depression and diabetes complications: a systematic review and meta-analysis. Diabet Med 2019; 36:1562-1572
  25. Omer Gilon M, Balmakov Y, Gelman S, Twig G. Adolescent Immigration and Type-2 Diabetes. Current diabetes reports 2021; 21:60
  26. Hamiel U, Pinhas-Hamiel O, Vivante A, Bendor C, Bardugo A, Afek A, Beer Z, Derazne E, Tzur D, Behar D, Itzhak A, Skorecki K, Tirosh A, Grossman E, Twig G. Impact of Immigration on Body Mass Index and Blood Pressure Among Adolescent Males and Females: A Nationwide Study. Hypertension 2019; 74:1316-1323
  27. Khanolkar AR, Amin R, Taylor-Robinson D, Viner RM, Warner J, Stephenson T. Inequalities in glycemic control in childhood onset type 2 diabetes in England and Wales-A national population-based longitudinal study. Pediatr Diabetes 2019; 20:821-831
  28. Magge SN, Wolf RM, Pyle L, Brown EA, Benavides VC, Bianco ME, Chao LC, Cymbaluk A, Balikcioglu PG, Halpin K, Hsia DS, Huerta-Saenz L, Kim JJ, Kumar S, Levitt Katz LE, Marks BE, Neyman A, O'Sullivan KL, Pillai SS, Shah AS, Shoemaker AH, Siddiqui JAW, Srinivasan S, Thomas IH, Tryggestad JB, Yousif MF, Kelsey MM. The Coronavirus Disease 2019 Pandemic is Associated with a Substantial Rise in Frequency and Severity of Presentation of Youth-Onset Type 2 Diabetes. J Pediatr 2022; 251:51-59.e52
  29. DeLacey S, Arzu J, Levin L, Ranganna A, Swamy A, Bianco ME. Impact of SARS-CoV2 on youth onset type 2 diabetes new diagnoses and severity. J Diabetes 2022; 14:532-540
  30. Denzer C, Rosenbauer J, Klose D, Körner A, Reinehr T, Baechle C, Schröder C, Wiegand S, Holl RW, Prinz N. Is COVID-19 to Blame? Trends of Incidence and Sex Ratio in Youth-Onset Type 2 Diabetes in Germany. Diabetes Care 2023; 46:1379-1387
  31. Chang TH, Chen YC, Chen WY, Chen CY, Hsu WY, Chou Y, Chang YH. Weight Gain Associated with COVID-19 Lockdown in Children and Adolescents: A Systematic Review and Meta-Analysis. Nutrients 2021; 13
  32. Srinivasan S, Chen L, Todd J, Divers J, Gidding S, Chernausek S, Gubitosi-Klug RA, Kelsey MM, Shah R, Black MH, Wagenknecht LE, Manning A, Flannick J, Imperatore G, Mercader JM, Dabelea D, Florez JC. The First Genome-Wide Association Study for Type 2 Diabetes in Youth: The Progress in Diabetes Genetics in Youth (ProDiGY) Consortium. Diabetes 2021; 70:996-1005
  33. Hao S, Cossen K, Westbrook AL, Umpierrez GE, Vellanki P. Diabetic Ketoacidosis and Long-term Insulin Requirements in Youths with Newly Diagnosed Type 2 Diabetes During the SARS-CoV-2 Pandemic. Endocrine practice : official journal of the American College of Endocrinology and the American Association of Clinical Endocrinologists 2023; 29:754-761
  34. Klingensmith GJ, Connor CG, Ruedy KJ, Beck RW, Kollman C, Haro H, Wood JR, Lee JM, Willi SM, Cengiz E, Tamborlane WV. Presentation of youth with type 2 diabetes in the Pediatric Diabetes Consortium. Pediatr Diabetes 2016; 17:266-273
  35. Klingensmith GJ, Lanzinger S, Tamborlane WV, Hofer SE, Cheng P, de Beaufort C, Gal RL, Reinehr T, Kollman C, Holl RW. Adolescent type 2 diabetes: Comparing the Pediatric Diabetes Consortium and Germany/Austria/Luxemburg Pediatric Diabetes Prospective registries. Pediatr Diabetes 2018; 19:1156-1163
  36. Otaigbe BE, Imafidon EE. Type 2 diabetes mellitus in a Nigerian child: a case report. African health sciences2011; 11:454-456
  37. Kevat D, Wilson D, Sinha A. A 5-year-old girl with type 2 diabetes. Lancet 2014; 383:1268
  38. Greenup E, Sunil B, Barr MM, Ashraf AP. Glycaemic control and outcomes in children with type 2 diabetes diagnosed at or before 10 years of age. Endocrinology, diabetes & metabolism 2021; 4:e00192
  39. Curran JA, Haynes A, Davis EA. Clinical characteristics of Western Australian children diagnosed with type 2 diabetes before 10 years of age. The Medical journal of Australia 2020; 212:95-95.e91
  40. Astudillo M, Tosur M, Castillo B, Rafaey A, Siller AF, Nieto J, Sisley S, McKay S, Nella AA, Balasubramanyam A, Bacha F, Redondo MJ. Type 2 diabetes in prepubertal children. Pediatr Diabetes 2021; 22:946-950
  41. Hutchins J, Barajas RA, Hale D, Escaname E, Lynch J. Type 2 diabetes in a 5-year-old and single center experience of type 2 diabetes in youth under 10. Pediatr Diabetes 2017; 18:674-677
  42. Amutha A, Unnikrishnan R, Anjana RM, Mohan V. Prepubertal Childhood Onset Type 2 Diabetes Mellitus: Four Case Reports. The Journal of the Association of Physicians of India 2017; 65:43-46
  43. Al Amiri E, Abdullatif M, Abdulle A, Al Bitar N, Afandi EZ, Parish M, Darwiche G. The prevalence, risk factors, and screening measure for prediabetes and diabetes among Emirati overweight/obese children and adolescents. BMC Public Health 2015; 15:1298-1298
  44. Alsaffar Y, Hussain AM, Selman NAJAAVdFyT. Prevalence of type 2 diabetes in pediatrics and adolescents newly diagnosed with diabetes in Babylon Governorate, Iraq. 2020; 39
  45. Moussa MA, Alsaeid M, Abdella N, Refai TM, Al-Sheikh N, Gomez JE. Prevalence of type 2 diabetes mellitus among Kuwaiti children and adolescents. Medical principles and practice : international journal of the Kuwait University, Health Science Centre 2008; 17:270-275
  46. Elkum N, Al-Arouj M, Sharifi M, Shaltout A, Bennakhi A. Prevalence of childhood obesity in the state of Kuwait. Pediatr Obes 2016; 11:e30-e34
  47. AlBlooshi A, Shaban S, AlTunaiji M, Fares N, AlShehhi L, AlShehhi H, AlMazrouei A, Souid AK. Increasing obesity rates in school children in United Arab Emirates. Obesity science & practice 2016; 2:196-202
  48. Pearson ER. Treating type 2 diabetes in youth: a depressing picture. The journal of the Royal College of Physicians of Edinburgh 2012; 42:228
  49. Kriska A, El Ghormli L, Copeland KC, Higgins J, Ievers-Landis CE, Levitt Katz LE, Trief PM, Wauters AD, Yasuda PM, Delahanty LM. Impact of lifestyle behavior change on glycemic control in youth with type 2 diabetes. Pediatr Diabetes 2018; 19:36-44
  50. Alfaraidi H, Samaan MC. Metformin therapy in pediatric type 2 diabetes mellitus and its comorbidities: A review. Frontiers in endocrinology 2022; 13:1072879
  51. Jones KL, Arslanian S, Peterokova VA, Park JS, Tomlinson MJ. Effect of metformin in pediatric patients with type 2 diabetes: a randomized controlled trial. Diabetes Care 2002; 25:89-94
  52. Gottschalk M, Danne T, Vlajnic A, Cara JF. Glimepiride versus metformin as monotherapy in pediatric patients with type 2 diabetes: a randomized, single-blind comparative study. Diabetes Care 2007; 30:790-794
  53. Laffel L, Chang N, Grey M, Hale D, Higgins L, Hirst K, Izquierdo R, Larkin M, Macha C, Pham T, Wauters A, Weinstock RS. Metformin monotherapy in youth with recent onset type 2 diabetes: experience from the prerandomization run-in phase of the TODAY study. Pediatr Diabetes 2012; 13:369-375
  54. Kelsey MM, Geffner ME, Guandalini C, Pyle L, Tamborlane WV, Zeitler PS, White NH. Presentation and effectiveness of early treatment of type 2 diabetes in youth: lessons from the TODAY study. Pediatr Diabetes2016; 17:212-221
  55. Filippatos TD, Panagiotopoulou TV, Elisaf MS. Adverse Effects of GLP-1 Receptor Agonists. The review of diabetic studies : RDS 2014; 11:202-230
  56. Tamborlane WV, Barrientos-Pérez M, Fainberg U, Frimer-Larsen H, Hafez M, Hale PM, Jalaludin MY, Kovarenko M, Libman I, Lynch JL, Rao P, Shehadeh N, Turan S, Weghuber D, Barrett T. Liraglutide in Children and Adolescents with Type 2 Diabetes. N Engl J Med 2019; 381:637-646
  57. Tamborlane WV, Bishai R, Geller D, Shehadeh N, Al-Abdulrazzaq D, Vazquez EM, Karoly E, Troja T, Doehring O, Carter D, Monyak J, Sjostrom CD. Once-Weekly Exenatide in Youth With Type 2 Diabetes. Diabetes Care 2022; 45:1833-1840
  58. Arslanian SA, Hannon T, Zeitler P, Chao LC, Boucher-Berry C, Barrientos-Perez M, Bismuth E, Dib S, Cho JI, Cox D, Investigators A-P. Once-Weekly Dulaglutide for the Treatment of Youths with Type 2 Diabetes. N Engl J Med 2022; 387:433-443
  59. Padda IS, Mahtani AU, Parmar M. Sodium-Glucose Transport Protein 2 (SGLT2) Inhibitors. StatPearls. Treasure Island (FL) ineligible companies. Disclosure: Arun Mahtani declares no relevant financial relationships with ineligible companies. Disclosure: Mayur Parmar declares no relevant financial relationships with ineligible companies.: StatPearls Publishing, Copyright © 2023, StatPearls Publishing LLC.; 2023.
  60. Laffel LM, Danne T, Klingensmith GJ, Tamborlane WV, Willi S, Zeitler P, Neubacher D, Marquard J. Efficacy and safety of the SGLT2 inhibitor empagliflozin versus placebo and the DPP-4 inhibitor linagliptin versus placebo in young people with type 2 diabetes (DINAMO): a multicentre, randomised, double-blind, parallel group, phase 3 trial. The lancet Diabetes & endocrinology 2023; 11:169-181
  61. Tamborlane WV, Laffel LM, Shehadeh N, Isganaitis E, Van Name M, Ratnayake J, Karlsson C, Norjavaara E. Efficacy and safety of dapagliflozin in children and young adults with type 2 diabetes: a prospective, multicentre, randomised, parallel group, phase 3 study. The lancet Diabetes & endocrinology 2022; 10:341-350
  62. Mulvihill EE, Drucker DJ. Pharmacology, physiology, and mechanisms of action of dipeptidyl peptidase-4 inhibitors. Endocrine reviews 2014; 35:992-1019
  63. Shankar RR, Zeitler P, Deeb A, Jalaludin MY, Garcia R, Newfield RS, Samoilova Y, Rosario CA, Shehadeh N, Saha CK, Zhang Y, Zilli M, Scherer LW, Lam RLH, Golm GT, Engel SS, Kaufman KD. A randomized clinical trial of the efficacy and safety of sitagliptin as initial oral therapy in youth with type 2 diabetes. Pediatr Diabetes 2022; 23:173-182
  64. Bensignor MO, Kelly AS, Arslanian S. Anti-obesity pharmacotherapy for treatment of pediatric type 2 diabetes: Review of the literature and lessons learned from adults. Frontiers in endocrinology 2022; 13:1043650
  65. Stretton B, Kovoor J, Bacchi S, Chang S, Ngoi B, Murray T, Bristow TC, Heng J, Gupta A, Ovenden C, Maddern G, Thompson CH, Heilbronn L, Boyd M, Rayner C, Talley NJ, Horowtiz M. Weight loss with subcutaneous semaglutide versus other glucagon-like peptide 1 receptor agonists in type 2 diabetes: a systematic review. Internal medicine journal 2023; 53:1311-1320
  66. Weghuber D, Barrett T, Barrientos-Pérez M, Gies I, Hesse D, Jeppesen OK, Kelly AS, Mastrandrea LD, Sørrig R, Arslanian S. Once-Weekly Semaglutide in Adolescents with Obesity. N Engl J Med 2022; 387:2245-2257
  67. Zeitler P, El Ghormli L, Arslanian S, Caprio S, Isganaitis E, Kelsey MK, Weinstock RS, White NH, Drews K. Deterioration of Glycemic Control in Youth-Onset Type 2 Diabetes: What Are the Early and Late Predictors? J Clin Endocrinol Metab 2022; 107:e3384-e3394
  68. ElSayed NA, Aleppo G, Aroda VR, Bannuru RR, Brown FM, Bruemmer D, Collins BS, Hilliard ME, Isaacs D, Johnson EL, Kahan S, Khunti K, Leon J, Lyons SK, Perry ML, Prahalad P, Pratley RE, Seley JJ, Stanton RC, Gabbay RA, on behalf of the American Diabetes A. 14. Children and Adolescents: Standards of Care in Diabetes-2023. Diabetes Care 2023; 46:S230-S253
  69. Asiri A, Alzahrani F, Alghamdi H, Alamri Z. Effect of Laparoscopic Sleeve Gastrectomy on HbA1C Level in Children with Type 2 Diabetes Mellitus. Medicina (Kaunas, Lithuania) 2022; 58
  70. Inge TH, Laffel LM, Jenkins TM, Marcus MD, Leibel NI, Brandt ML, Haymond M, Urbina EM, Dolan LM, Zeitler PS. Comparison of Surgical and Medical Therapy for Type 2 Diabetes in Severely Obese Adolescents. JAMA pediatrics 2018; 172:452-460
  71. Zhu X, Zhou G, Gu X, Jiang X, Huang H, You S, Zhang G. Comparison of bariatric surgery and medical therapy for obese adolescents with type 2 diabetes. Asian journal of surgery 2022;
  72. Bjornstad P, Hughan K, Kelsey MM, Shah AS, Lynch J, Nehus E, Mitsnefes M, Jenkins T, Xu P, Xie C, Inge T, Nadeau K. Effect of Surgical Versus Medical Therapy on Diabetic Kidney Disease Over 5 Years in Severely Obese Adolescents With Type 2 Diabetes. Diabetes Care 2020; 43:187-195
  73. Bjornstad P, Nehus E, van Raalte D. Bariatric surgery and kidney disease outcomes in severely obese youth. Seminars in pediatric surgery 2020; 29:150883
  74. Ryder JR, Xu P, Nadeau KJ, Kelsey MM, Xie C, Jenkins T, Inge TH, Bjornstad P. Effect of surgical versus medical therapy on estimated cardiovascular event risk among adolescents with type 2 diabetes and severe obesity. Surgery for obesity and related diseases : official journal of the American Society for Bariatric Surgery 2021; 17:23-33
  75. Jastreboff AM, Kaplan LM, Frías JP, Wu Q, Du Y, Gurbuz S, Coskun T, Haupt A, Milicevic Z, Hartman ML. Triple-Hormone-Receptor Agonist Retatrutide for Obesity - A Phase 2 Trial. N Engl J Med 2023; 389:514-526
  76. Medicine TSGJNEJo. Long-term complications in youth-onset type 2 diabetes. 2021; 385:416-426
  77. Rapid rise in hypertension and nephropathy in youth with type 2 diabetes: the TODAY clinical trial. Diabetes Care2013; 36:1735-1741
  78. Bjornstad P, Drews KL, Caprio S, Gubitosi-Klug R, Nathan DM, Tesfaldet B, Tryggestad J, White NH, Zeitler P. Long-Term Complications in Youth-Onset Type 2 Diabetes. N Engl J Med 2021; 385:416-426
  79. Cioana M, Deng J, Hou M, Nadarajah A, Qiu Y, Chen SSJ, Rivas A, Banfield L, Chanchlani R, Dart A, Wicklow B, Alfaraidi H, Alotaibi A, Thabane L, Samaan MC. Prevalence of Hypertension and Albuminuria in Pediatric Type 2 Diabetes: A Systematic Review and Meta-analysis. JAMA Netw Open 2021; 4:e216069
  80. Chu PY, Campbell MJ, Miller SG, Hill KD. Anti-hypertensive drugs in children and adolescents. World journal of cardiology 2014; 6:234-244
  81. Siddiqi N, Shatat IF. Antihypertensive agents: a long way to safe drug prescribing in children. Pediatric nephrology (Berlin, Germany) 2020; 35:2049-2065
  82. Dabelea D, Sauder KA, Jensen ET, Mottl AK, Huang A, Pihoker C, Hamman RF, Lawrence J, Dolan LM, Agostino R, Jr., Wagenknecht L, Mayer-Davis EJ, Marcovina SM. Twenty years of pediatric diabetes surveillance: what do we know and why it matters. Ann N Y Acad Sci 2021;
  83. Amatruda M, Gembillo G, Giuffrida AE, Santoro D, Conti G. The Aggressive Diabetic Kidney Disease in Youth-Onset Type 2 Diabetes: Pathogenetic Mechanisms and Potential Therapies. Medicina (Kaunas, Lithuania) 2021; 57
  84. Naaman SC, Bakris GL. Diabetic Nephropathy: Update on Pillars of Therapy Slowing Progression. Diabetes Care 2023; 46:1574-1586
  85. Blazek O, Bakris GL. Slowing the Progression of Diabetic Kidney Disease. Cells 2023; 12
  86. Jackson S, Creo A, Kumar S. Are Clinicians Aggressive Enough in Treating Diabetes-Related Hyperlipidemia in Youth? Current atherosclerosis reports 2022; 24:471-481
  87. Cioana M, Deng J, Nadarajah A, Hou M, Qiu Y, Chen SSJ, Rivas A, Banfield L, Alfaraidi H, Alotaibi A, Thabane L, Samaan MC. Prevalence of Polycystic Ovary Syndrome in Patients With Pediatric Type 2 Diabetes: A Systematic Review and Meta-analysis. JAMA Network Open 2022; 5:e2147454-e2147454
  88. Pregnancy Outcomes in Young Women With Youth-Onset Type 2 Diabetes Followed in the TODAY Study. Diabetes Care 2021; 45:1038-1045
  89. Shah RD, Chernausek SD, El Ghormli L, Geffner ME, Keady J, Kelsey MM, Farrell R, Tesfaldet B, Tryggestad JB, Van Name M, Isganaitis E. Maternal Diabetes in Youth-Onset Type 2 Diabetes Is Associated With Progressive Dysglycemia and Risk of Complications. J Clin Endocrinol Metab 2023; 108:1120-1131
  90. Merino PM, Codner E. Contraception for Adolescents and Young Women with Type 2 Diabetes-Specific Considerations. Current diabetes reports 2022; 22:77-84
  91. Cioana M, Deng J, Nadarajah A, Hou M, Qiu Y, Chen SSJ, Rivas A, Toor PP, Banfield L, Thabane L, Chaudhary V, Samaan MC. Global Prevalence of Diabetic Retinopathy in Pediatric Type 2 Diabetes: A Systematic Review and Meta-analysis. JAMA Netw Open 2023; 6:e231887
  92. Mititelu M, Uschner D, Doherty L, Bjornstad P, Domalpally A, Drews KL, Gubitosi-Klug R, Levitsky LL, Pak JW, White NH, Blodi BA. Retinal Thickness and Morphology Changes on OCT in Youth with Type 2 Diabetes: Findings from the TODAY Study. Ophthalmol Sci 2022; 2:100191
  93. Wright AD, Dodson PM. Medical management of diabetic retinopathy: fenofibrate and ACCORD Eye studies. Eye (London, England) 2011; 25:843-849
  94. Dabelea D, Stafford JM, Mayer-Davis EJ, D'Agostino R, Jr., Dolan L, Imperatore G, Linder B, Lawrence JM, Marcovina SM, Mottl AK, Black MH, Pop-Busui R, Saydah S, Hamman RF, Pihoker C. Association of Type 1 Diabetes vs Type 2 Diabetes Diagnosed During Childhood and Adolescence With Complications During Teenage Years and Young Adulthood. Jama 2017; 317:825-835
  95. Dart AB, Martens PJ, Rigatto C, Brownell MD, Dean HJ, Sellers EA. Earlier Onset of Complications in Youth With Type 2 Diabetes. Diabetes Care 2014; 37:436-443
  96. Amutha A, Datta M, Unnikrishnan R, Anjana RM, Mohan V. Clinical profile and complications of childhood- and adolescent-onset type 2 diabetes seen at a diabetes center in south India. Diabetes Technol Ther 2012; 14:497-504
  97. Jaiswal M, Lauer A, Martin CL, Bell RA, Divers J, Dabelea D, Pettitt DJ, Saydah S, Pihoker C, Standiford DA, Rodriguez BL, Pop-Busui R, Feldman EL. Peripheral neuropathy in adolescents and young adults with type 1 and type 2 diabetes from the SEARCH for Diabetes in Youth follow-up cohort: a pilot study. Diabetes Care 2013; 36:3903-3908
  98. Jaiswal M, Divers J, Dabelea D, Isom S, Bell RA, Martin CL, Pettitt DJ, Saydah S, Pihoker C, Standiford DA, Dolan LM, Marcovina S, Linder B, Liese AD, Pop-Busui R, Feldman EL. Prevalence of and Risk Factors for Diabetic Peripheral Neuropathy in Youth With Type 1 and Type 2 Diabetes: SEARCH for Diabetes in Youth Study. Diabetes Care 2017; 40:1226-1232
  99. Akinci G, Savelieff MG, Gallagher G, Callaghan BC, Feldman EL. Diabetic neuropathy in children and youth: New and emerging risk factors. Pediatr Diabetes 2021; 22:132-147
  100. Vinik AI, Maser RE, Mitchell BD, Freeman R. Diabetic autonomic neuropathy. Diabetes Care 2003; 26:1553-1579
  101. Duque A, Mediano MFF, De Lorenzo A, Rodrigues LF, Jr. Cardiovascular autonomic neuropathy in diabetes: Pathophysiology, clinical assessment and implications. World J Diabetes 2021; 12:855-867
  102. Varley BJ, Gow ML, Cho YH, Benitez-Aguirre P, Cusumano J, Pryke A, Chan A, Velayutham V, Donaghue KC, Craig ME. Higher frequency of cardiovascular autonomic neuropathy in youth with type 2 compared to type 1 diabetes: Role of cardiometabolic risk factors. Pediatr Diabetes 2022; 23:1073-1079
  103. Jaiswal M, Divers J, Urbina EM, Dabelea D, Bell RA, Pettitt DJ, Imperatore G, Pihoker C, Dolan LM, Liese AD, Marcovina S, Linder B, Feldman EL, Pop-Busui R. Cardiovascular autonomic neuropathy in adolescents and young adults with type 1 and type 2 diabetes: The SEARCH for Diabetes in Youth Cohort Study. Pediatr Diabetes2018; 19:680-689
  104. Wadwa RP, Urbina EM, Anderson AM, Hamman RF, Dolan LM, Rodriguez BL, Daniels SR, Dabelea D, Group ftSS. Measures of Arterial Stiffness in Youth With Type 1 and Type 2 Diabetes: The SEARCH for Diabetes in Youth study. Diabetes Care 2010; 33:881-886
  105. Shah AS, Gidding SS, El Ghormli L, Tryggestad JB, Nadeau KJ, Bacha F, Levitt Katz LE, Willi SM, Lima J, Urbina EM. Relationship between Arterial Stiffness and Subsequent Cardiac Structure and Function in Young Adults with Youth-Onset Type 2 Diabetes: Results from the TODAY Study. Journal of the American Society of Echocardiography : official publication of the American Society of Echocardiography 2022; 35:620-628.e624
  106. Urbina EM, Isom S, Dabelea D, D'Agostino R, Daniels SR, Dolan LM, Imperatore G, Lustigova E, Marcovina S, Mottl A, Pihoker C, Shah AS. Association of Elevated Arterial Stiffness With Cardiac Target Organ Damage and Cardiac Autonomic Neuropathy in Young Adults With Diabetes: The SEARCH for Diabetes in Youth Study. Diabetes Care 2023; 46:786-793
  107. Urbina EM, Kimball TR, McCoy CE, Khoury PR, Daniels SR, Dolan LM. Youth with obesity and obesity-related type 2 diabetes mellitus demonstrate abnormalities in carotid structure and function. Circulation 2009; 119:2913-2919
  108. Shah AS, Urbina EM. Vascular and Endothelial Function in Youth with Type 2 Diabetes Mellitus. Current diabetes reports 2017; 17:36-36
  109. Lee HS, Yoon JS, Park KJ, Lim JS, Hwang JS. The Relationship Between Bone Mineral Density and Type 2 Diabetes in Obese Children and Adolescents at the Time of Initial Diagnosis. Hormone and metabolic research = Hormon- und Stoffwechselforschung = Hormones et metabolisme 2019; 51:42-46
  110. Kindler JM, Kelly A, Khoury PR, Levitt Katz LE, Urbina EM, Zemel BS. Bone Mass and Density in Youth With Type 2 Diabetes, Obesity, and Healthy Weight. Diabetes Care 2020; 43:2544-2552
  111. Monaghan M, Mara CA, Kichler JC, Westen SC, Rawlinson A, Jacobsen LM, Adams RN, Stone JY, Hood KK, Mulvaney SA. Multisite Examination of Depression Screening Scores and Correlates Among Adolescents and Young Adults With Type 2 Diabetes. Can J Diabetes 2021; 45:411-416
  112. McVoy M, Hardin H, Fulchiero E, Caforio K, Briggs F, Neudecker M, Sajatovic M. Mental health comorbidity and youth onset type 2 diabetes: A systematic review of the literature. International journal of psychiatry in medicine2023; 58:37-55
  113. Hoffman RP, Damilano CP, Hong KMC, Glick BA, Kamboj MK. Glycemic control, depression, diabetes distress among adolescents with type 2 diabetes: effects of sex, race, insurance, and obesity. Acta Diabetol 2022; 59:1083-1089
  114. Gulley LD, Shomaker LB. Depression in Youth-Onset Type 2 Diabetes. Current diabetes reports 2020; 20:51
  115. Wilfley D, Berkowitz R, Goebel-Fabbri A, Hirst K, Ievers-Landis C, Lipman TH, Marcus M, Ng D, Pham T, Saletsky R, Schanuel J, Van Buren D. Binge eating, mood, and quality of life in youth with type 2 diabetes: baseline data from the today study. Diabetes Care 2011; 34:858-860
  116. Nip ASY, Reboussin BA, Dabelea D, Bellatorre A, Mayer-Davis EJ, Kahkoska AR, Lawrence JM, Peterson CM, Dolan L, Pihoker C. Disordered Eating Behaviors in Youth and Young Adults With Type 1 or Type 2 Diabetes Receiving Insulin Therapy: The SEARCH for Diabetes in Youth Study. Diabetes Care 2019; 42:859-866
  117. Reynolds K, Saydah SH, Isom S, Divers J, Lawrence JM, Dabelea D, Mayer-Davis EJ, Imperatore G, Bell RA, Hamman RF. Mortality in youth-onset type 1 and type 2 diabetes: The SEARCH for Diabetes in Youth study. Journal of diabetes and its complications 2018; 32:545-549
  118. Rhodes ET, Prosser LA, Hoerger TJ, Lieu T, Ludwig DS, Laffel LM. Estimated morbidity and mortality in adolescents and young adults diagnosed with Type 2 diabetes mellitus. Diabetic medicine : a journal of the British Diabetic Association 2012; 29:453-463

Diabetes in People Living with HIV

ABSTRACT

 

People living with HIV (PLWH) are living longer and also have unique risk factors for developing several metabolic diseases, including diabetes. This has been observed in both high income and low to middle income countries. Risk factors for diabetes in PLWH consist of specific antiretroviral therapies (ART), including older generation protease inhibitors and nucleoside reverse transcriptase inhibitors, lipodystrophy, and hepatitis C co-infection. In addition, obese and overweight states are common in PLWH, and an increased risk of incident diabetes has been noted with weight gain after ART initiation in PLWH, compared to individuals without HIV. Inflammation associated with HIV has also been linked to incident diabetes. This chapter reviews points to consider in the diagnosis and monitoring of diabetes in PLWH and discusses interactions that may occur between specific ART agents and glucose-lowering medications. Moreover, PLWH have risk factors for complications involving organ systems that are also affected by microvascular disease in diabetes. Because PLWH have a greater risk for cardiovascular disease (CVD) than individuals without HIV, modifiable risk factors of CVD should be addressed in the care of PLWH, considering that dyslipidemia, hypertension, and cigarette smoking are all highly prevalent in PLWH.

 

BACKGROUND

           

HIV infection is prevalent in 38.4 million people worldwide as of 2021 (1) and 1.2 million people in the United States as of 2019. The incidence of HIV infection has decreased from 2015 to 2019 in the United States but has remained stable in some demographic groups, including African-Americans and Latinos (2). Untreated, HIV infection can lead to opportunistic infections including cytomegalovirus disease and pneumocystosis (3) associated with AIDS, which is defined as a CD4+ cell count < 200 cells/mL. With advances in antiretroviral therapy (ART), however, HIV infection has been transformed from a disease strongly linked to AIDS and opportunistic infections into a chronic disease that is associated with several cardiometabolic consequences, including diabetes (4), heart disease (5), and other non-AIDS related illnesses such as osteoporosis (6). In part, this phenomenon is secondary to the fact that people living with HIV (PLWH) are living longer and are more susceptible to diseases of aging.

 

An important aspect to consider in the understanding of HIV associated cardiometabolic diseases is their growing global presence. The same factors that are associated with cardiometabolic disease prevalence in PLWH in high-income countries are also present in low-income countries, in addition to unique factors including industrialization, with resultant decreased physical activity and increased energy consumption (7) (8). As such, the care of PLWH needs to focus not only on ART but also on the management of chronic co-morbidities. This chapter will focus primarily on diabetes in PLWH.

 

EPIDEMIOLOGY AND RISK FACTORS FOR DIABETES IN PLWH

 

Epidemiology           

 

PLWH have a unique set of risk factors that may increase their likelihood of developing diabetes. Illustrating the greater burden of diabetes in PLWH on ART, a study from 2005 on the Multicenter AIDS Cohort Study (MACS) of gay and bisexual men with (MWH) and without HIV (MWoH) found that the incidence of diabetes was found to be more than fourfold higher in MWH and that the prevalence of diabetes was 14% in MWH on ART and 7% in MWH not on ART, compared to 5% in MWoH. These differences were significant even after adjusting for age and body mass index (BMI). Of note, the majority of MWH on ART in the study were on first generation protease inhibitor (PI) therapy (discussed further in the section Protease Inhibitors) (9). Other studies have described incidence rates of diabetes in PLWH of 4.4 cases per 1000 person-years of follow-up in the Swiss HIV Cohort Study (10) and 5.72 cases per 1000 person-years of follow-up in the Data Collection on Adverse Events of Anti-HIV Drugs (D:A:D) Study of participants from Europe, the US, Australia, and Argentina (11).

 

The effect of HIV disease on developing diabetes is also growing in low- and middle-income countries (LMIC), where the majority of the people with diabetes live (12). Prevalence of estimates of diabetes in PLWH in LMIC range from 6.8% in Chile (13) to 26% in Cameroon (14). PLWH and diabetes in LMIC are younger than those in high income countries (15). The wide range of prevalence estimates of diabetes in PLWH in LMIC may be in part because of differences in the definitions used to identifying diabetes. In addition to factors common to PLWH globally, urbanization may be a contributing factor to the development of diabetes in PLWH in LMIC (16).

 

Antiretroviral Therapy (ART)

 

PROTEASE INHIBITORS (PI)

 

One risk factor for developing diabetes in PLWH is PI use. In 1995, saquinavir became the first FDA approved PI (17). Incidence rates of hyperglycemia as high as fivefold have been reported in the setting of PI use (18). In addition, PIs, including ritonavir, have been associated with hypertriglyceridemia (19,20) and lipodystrophy (21). Mechanisms that may account for these effects are multifold. In one study, insulin sensitivity, as measured by glucose infusion rate during hyperglycemic clamp, decreased significantly after 12 weeks of PI therapy compared to baseline. A defect in beta cell function, in particular, a decrease in the disposition index, was also observed after PI therapy (22). An in vitro study demonstrated that indinavir may lower the function of the glucose transporter GLUT4 (23). Additional mechanisms may include changes in the hormone adiponectin, which is associated with improved insulin sensitivity, although in vivo and in vitro effects of PIs on adiponectin have differed (24,25).

 

The prescribing patterns of PI use have evolved. One study found that in the Veterans Affairs system, the prevalence of PI based regimen use decreased from 1997 to         2004 (26). In addition, the effects of all PIs, namely older generation versus newer generation PIs, including atazanavir and darunavir, on glucose homeostasis are not equal. In an in vitro study, atazanavir, which received FDA approval in 2003 (27), did not inhibit either GLUT1 or GLUT4 glucose transporters (28). In a clinical study of HIV- participants, atazanavir in combination with ritonavir had a significantly lower effect on insulin sensitivity compared to lopinavir/ritonavir, as measured by glucose disposal rate during hyperinsulinemic euglycemic clamp. (29). Moreover, the newer generation PIs have not been associated with an increased incidence of diabetes, with one study showing a lower risk of diabetes in individuals on these medications (30).

 

NUCLEOSIDE REVERSE TRANSCRIPTASE INHIBITORS (NRTIs)

 

In addition to PIs, certain nucleoside reverse transcriptase inhibitors (NRTIs) have been associated with diabetes. In the D:A:D study, incident diabetes was associated with exposure to the NRTIs stavudine and zidovudine, both thymidine analogs, and didanosine, after adjusting for age, sex, race, and BMI (11). NRTIs have also been implicated in mitochondrial dysfunction, which may be another mechanism by which NRTIs are associated with diabetes. One study found that one month of treatment with stavudine was associated with significant decreases in mitochondrial DNA from muscle biopsies and insulin sensitivity, as measured by the glucose infusion rate during a hyperinsulinemic euglycemic clamp (31). Because of serious adverse effects linked to the use of these NRTIs, stavudine, zidovudine, and didanosine are not advised for use in the United States (32).

 

Similar to the newer generation of PIs, the use of newer NRTIs, including emtricitabine, abacavir, and tenofovir, have been associated with a lower risk of diabetes (30). Supporting this finding, tenofovir disoproxil fumarate (TDF), in a study of participants without HIV or diabetes, did not significantly affect insulin sensitivity, as measured by hyperinsulinemic euglycemic clamp (33).

 

Tenofovir alafenamide (TAF) is a newer prodrug of tenofovir and is associated with less renal and bone toxicity than TDF (33a). However, TAF has been associated with weight gain in PLWH, compared with TDF (33b). Weight gain leading to an overweight state or obesity is a risk factor for the development of type 2 diabetes. In a retrospective study of the ADVANCE trial, which included ART naïve PLWH randomized to 3 treatment arms, 2 of which included TDF and 1 of which included TAF, an increase in the 10-year predicted risk of type 2 diabetes was observed for participants in the TAF arm, compared to participants in one of the TDF arms, although the risk calculator that was used had not been validated for the population in the study (33c). On the other hand, in a cohort study of more than 4,000 PWH on a TDF-based ART regimen for at least 6 months, of whom about 80% switched to a TAF-based ART regimen and about 20% continued on a TDF-based ART regimen, TAF was associated with a significant weight gain, compared to TDF, after 18 months, but it was not associated with incident diabetes.

 

INTEGRASE STRAND TRANSFER INHIBITORS (INSTIs)

           

Integrase strand transfer inhibitor (INSTI) based regimens are among the recommended initial ART regimens for PLWH (32) (34). Reasons for this include the effectiveness of and fewer adverse effects associated with INSTIs in studies of treatment-naïve patients (34-37). However, studies have discovered some metabolic effects of INSTIs. These include weight gain on average of 2.9 kilograms over 18 months, as observed in a retrospective study of patients who switched to an INSTI based regimen from previously being on a non-NRTI (NNRTI)-based regimen. This was in contrast to an average 0.7 kg weight gain in those participants who switched to a PI-based regimen and 0.9 kg weight gain in those who continued on a NNRTI-based regimen (38). In addition to weight gain, case reports of new onset diabetes have been reported in conjunction with INSTI use (39,40). One clinical trial compared the INSTI raltegravir to 2 boosted PI regimens and found that the increases in homeostasis model assessment-insulin resistance (HOMA-IR) in all 3 arms were not significantly different from one another, were noted by 4 weeks, and appeared to be independent of changes in visceral adipose tissue (41). However, other studies have not demonstrated an association between diabetes and INSTI use (41a) or have found that the association between INSTI use and incident diabetes varies by individual INSTI (41b).

 

The exact mechanisms by which INSTIs may be related to weight gain and diabetes are not known. One proposed mechanism by which INSTIs may cause hyperglycemia is their effect on magnesium, which is a necessary cation for insulin action (40). In mice, INSTI use leads to an increase in fat mass and decreases brown and beige adipocyte differentiation, leading to lower energy expenditure and weight gain (41c).

 

Lipodystrophy

 

Lipodystrophy associated with ART is characterized by either lipoatrophy in the face, arms, and legs, or lipoaccumulation that can lead to gynecomastia, dorsocervical fat tissue, and increased intra-abdominal fat,  (42,43), or mixed lipodystrophy, in which lipoatrophy and lipoaccumulation occur together. As noted above, lipodystrophy has been associated with PI use (21) and with older generation NRTI use, including stavudine (44), but less commonly with the newer generation NRTI tenofovir (45). In a cross-sectional study, HIV+ men with lipodystrophy had a greater insulin resistance than men without lipodystrophy (21). Lipodystrophy has also been associated with incident diabetes (46,47).

 

In one study that measured body fat changes longitudinally using dual-energy x-ray absorptiometry (DXA) in PLWH on ART (specifically zidovudine and lamivudine or didanosine and stavudine in combination with nelfinavir, efavirenz, or both), 32% of participants had discordant changes in trunk and limb fat (48).

 

Lipodystrophy may also be persistent in patients with exposure to thymidine analogs. Although improvements in limb fat mass were reported in patients who switched from taking the thymidine analog zidovudine to the NRTI abacavir, no significant improvement was noted in self-assessment of dorsocervical fat (49).

 

Hepatitis C Co-Infection

 

25% of PLWH in the U.S. are co-infected with hepatitis C (50). Hepatitis C infection is associated with a higher risk of developing diabetes. Treatments for hepatitis C, including direct-acting antiviral treatment and pegylated interferon/ribivirin, have been found to lower the incidence of diabetes, with a larger effect in those patients with advanced fibrosis/cirrhosis. In addition among individuals who received treatment for hepatitis C, those with a sustained virologic response were less likely to develop diabetes than those without a sustained virologic response (51).

 

Weight Gain, Overweight, and Obesity

 

Weight gain that is observed after the initiation of ART in those patients with wasting associated with untreated HIV has been characterized as a “return to health” phenomenon. However, because of an emphasis on early ART initiation, the wasting that was previously seen with advanced HIV infection is less common in countries with access to ART (52). In one 2012 study conducted in Alabama, more than 40% of patients were overweight or obese at the time of starting ART. After 2 years on ART, the percentages of underweight and normal weight participants decreased significantly, and significant increases in the percentages of overweight and obese participants were observed. Having a lower baseline CD4 count and the use of a PI as a third drug were risk factors associated with a greater increase in BMI (53).

 

The consequences of weight gain in PLWH may be different from those in the general population. In a study of U.S. Veterans, the risk of incident diabetes with 10 or more pounds of weight gain during the first year after ART initiation in HIV+ Veterans was significantly greater than in HIV- Veterans (54).

 

Among cohorts of PLWH from different countries, prevalence estimates of an overweight state or obesity range from 25% to 68% (55,56). In one study of PLWH, risk factors for being overweight or obese in PLWH included increasing age and either no evidence of hepatitis C infection or evidence of cleared hepatitis C infection (57).

 

Inflammation

 

In addition to the risk factors for diabetes in PLWH outlined above, inflammation may play a role in the development of diabetes in PLWH. Systemic inflammation results from several factors: viral replication, immune activation, which leads to T cell depletion, as well as T cell loss specifically in the gastrointestinal tract, with associated translocation of microbial factors including lipopolysaccharide (58). Co-infection with viruses, including hepatitis C virus (as discussed above), hepatitis B virus, Epstein-Barr virus, and cytomegalovirus, perpetuate systemic inflammation (59).

 

Although ART reduces some inflammatory biomarkers in PLWH (60), there is evidence of residual inflammation, with levels of other inflammatory biomarkers in PLWH on ART that do not decrease to levels seen in HIV- individuals (61). Persistent inflammation measured months after ART initiation has been associated with incident diabetes in PLWH (62).

 

DIAGNOSIS OF AND MONOTORING DIABETES IN PLWH

 

The American Diabetes Association (ADA) recommends the use of a fasting plasma glucose, hemoglobin A1c, or a 2-hour plasma glucose after a 75-gram oral glucose tolerance test to establish the diagnosis of diabetes in the general population. However, there is a caveat that for certain populations of patients, including PLWH, fasting plasma glucose is preferable to hemoglobin A1c (63). Hemoglobin A1c has been found to underestimate glycemia in PLWH (64,65). One reason for this is the use of medications that cause hemolysis, including dapsone and trimethoprim-sulfamethoxazole, which are used for prophylaxis of Pneumocystis jiroveci, an opportunistic infection (65). However, another reason for the discrepancy between glucose levels and hemoglobin A1c is NRTI use. NRTIs, especially the thymidine analogs, are associated with an increased risk of macrocytosis (66), which can lower hemoglobin A1c. Finally, lower CD4 cell count (< 500 cells/mm3) was associated with a significant discordance between expected and measured hemoglobin A1c in MWoH in the MACS (67). As such, self-monitoring of blood glucose in PLWH may be preferable to using hemoglobin A1c for monitoring glycemic control (64).

 

The most recent Infectious Diseases Society of America guidelines on the primary care of PLWH recommends obtaining either a fasting plasma glucose and/or hemoglobin A1c at baseline and at 1 to 3 months after starting ART. In addition, hemoglobin A1c is preferred to fasting glucose for diagnosing diabetes because of the relative ease of obtaining a hemoglobin A1c over a fasting glucose. However, a hemoglobin A1c level ≥ 5.8% can be considered to diagnose diabetes in PLWH, instead of the ADA recommendation of ≥ 6.5%, as this improves the sensitivity of the test (68,69). Moreover, a hemoglobin A1c is recommended to be obtained every 6 months in PLWH and diabetes, with a goal hemoglobin A1c of < 7% (68).

 

TREATMENT OF DIABETES IN PLWH

           

Special considerations should be taken into account in the treatment of diabetes in PLWH. PLWH may not respond to treatment for diabetes in the same manner as HIV- individuals. Part of this observation may be because treatment responses were measured using hemoglobin A1c, which may be an inaccurate indicator of glycemia in PLWH (70), as noted above.

Interactions between several diabetes medications and ART are known. In addition, some diabetes medications may present advantages or disadvantages in PLWH. These conditions are described in further detail below and are organized by diabetes medication.

 

Lifestyle Interventions

 

Both the 2023 ADA Standards of Care on Pharmacologic Approaches to Glycemic Treatment and the 2023 updated American Association of Clinical Endocrinology algorithm of the management of type 2 diabetes highlight lifestyle interventions as part of the recommended treatment of type 2 diabetes (70a, 70b). Lifestyle interventions include not only a healthy diet and exercise but also smoking cessation and moderating alcohol intake (70b).

 

Physical activity has benefits for PLWH, including decreasing waist circumference (70c) and improvements in glucose and high-density lipoprotein cholesterol levels (70d). However, low physical activity has been reported in some studies of PLWH (70e, 70f).

 

Metformin

 

The 2023 ADA Standards of Care on Pharmacologic Approaches to Glycemic Treatment includes metformin as one first-line treatment to consider in patients in the general population with type 2 diabetes (70a) (71).

 

In PLWH with lipodystrophy and insulin resistance but without diabetes, 3 months of metformin 1000 mg twice daily was found to significantly decrease insulin area under the curve after an oral glucose tolerance test. Although no increased incidence in lactic acidosis was noted in the participants who received metformin, the study was not powered for this outcome, and liver and kidney dysfunction were exclusion criteria (72).

 

The commonly used integrase transfer strand inhibitor, dolutegravir, increases plasma levels of metformin, and thus adjusting the dose of metformin upon dolutegravir initiation has been recommended (73,74). However, one retrospective study found that there was no significant difference in glycemic control before and after starting dolutegravir in PLWH and diabetes on metformin (75).

 

In summary, there are no guidelines to suggest that metformin should not be a possible first-line treatment of type 2 diabetes in PLWH, after consideration of liver and kidney function and dolutegravir treatment.

 

Sulfonylureas

 

Sulfonylureas are a substrate of the CYP2C9 enzyme. The PIs ritonavir and nelfinavir are CYP2C9 inducers and can decrease sulfonylurea levels (76,77). In comparison with initial use of metformin in PLWH, no significant difference in glycemia after one year of therapy was noted with initial use of a sulfonylurea (78).

 

The 2023 ADA guidelines note that sulfonylureas are a high efficacy drug class for glucose management and are less cost-prohibitive compared to many other drug classes (70a) (71). However, adverse effects of sulfonylureas include hypoglycemia and weight gain. In PLWH, consideration should be made if a patient is on ritonavir or nelfinavir and the possible loss of efficacy of sulfonylurea treatment.

 

Thiazolidinediones

 

The effect of thiazolidinediones on glycemic control in PLWH with diabetes specifically has not been studied, although some trials found that rosiglitazone lowered serum insulin levels in PLWH without diabetes (79) and improved insulin sensitivity in PLWH with hyperinsulinemia (80). Several studies have focused on the effect of thiazolidinediones (TZDs) on body fat in PLWH with lipodystrophy, with equivocal findings. One 24-week study on the effect of rosiglitazone on PLWH, all of whom were on a PI, found no significant difference in arm fat between the treatment and placebo groups (81). However, the study did not have enough power to detect a difference in this outcome (82). On the other hand, other studies demonstrated that rosiglitazone increased visceral and subcutaneous abdominal fat in MWH with lipodystrophy over 6 months (83) and subcutaneous leg fat in PLWH with lipodystrophy over 3 months (80).

 

Adverse side effects of TZDs should be considered and discussed prior to initiation. These include fluid retention, osteoporosis, edema, and potential liver injury (84).

 

Dipeptidyl Peptidase 4 Inhibitors

 

Some clinical studies have demonstrated that dipeptidyl peptidase 4 (DPP4) inhibitors exert an anti-inflammatory effect in patients with type 2 diabetes (85,86). This idea was further examined in PLWH in a pilot study of 20 PLWH on ART randomized to either the DPP4 inhibitor sitagliptin or placebo for 24 weeks. A significant decrease was noted in the chemokine SDF-1α in the treatment group. In addition, an improvement in glucose tolerance was noted at week 8 in the treatment group, but the difference in glucose tolerance between the two groups was no longer significant at the end of the study (87). In another study of PLWH with impaired glucose tolerance, sitagliptin resulted in a significant improvement in glucose tolerance and decreases in the inflammatory markers hsCRP and CXCL10 from baseline after 8 weeks of treatment, compared to placebo (88). Similarly, in a larger study of 84 PLWH on ART with viral suppression and without diabetes who were randomized to 16 weeks of sitagliptin versus placebo, a significant decrease from baseline was seen at week 15 in CXCL10 in participants in the treatment group (89).

 

Glucagon-Like Peptide-1 Receptor Agonists

           

Several glucagon-like peptide-1 (GLP-1) receptor agonists have been shown to improve weight and cardiovascular outcomes, albeit in patient populations not specific to PLWH (90,91). Liraglutide, dulaglutide, and semaglutide have been shown to lower the risk of a composite outcome of nonfatal stroke, nonfatal myocardial infarction, and cardiovascular death in patients with type 2 diabetes (90,91). Liraglutide, semaglutide, and dulaglutide have indications from the Food and Drug Administration for lowering cardiovascular event risk in patients with cardiovascular disease and type 2 diabetes, and dulaglutide is also approved for lowering cardiovascular event risk in patients with multiple cardiovascular risk factors and type 2 diabetes. The 2023 ADA guidelines recommend the use of a GLP-1 receptor agonist as a first-line treatment in patients with cardiovascular disease and type 2 diabetes (70a).

 

One case report described a single PLWH who was able to discontinue insulin treatment (insulin glargine 60 units daily) and who experienced improvements in weight and hemoglobin A1c after starting liraglutide therapy (92). Another case report described improvements in hemoglobin A1c and fasting glucose on dulaglutide in a PLWH. Relatively scant literature exists on the effects of GLP-1 receptors agonists in PLWH and diabetes (93,94).

 

Sodium-Glucose Co-Transporter-2 Inhibitors

           

Similar to the GLP-1 receptor agonists, sodium-glucose co-transporter-2 (SGLT2) inhibitors have demonstrated reductions in adverse cardiovascular outcomes, including cardiovascular death and hospitalization for heart failure in general population studies (95,96). In addition, both dapagliflozin and canagliflozin have an FDA indication to reduce the risk of end-stage renal disease in patients with type 2 diabetes and diabetic nephropathy with albuminuria (96a). As such, the 2023 ADA guidelines on pharmacologic treatment of diabetes recommend that in those patients with type 2 diabetes and chronic kidney disease, a SGLT2 inhibitor that has been shown to reduce risk of renal disease progression should be used (70a).

 

SGLT2 inhibitors are also approved for reduction of adverse cardiovascular events and heart failure hospitalization in people with diabetes and have cardiovascular benefits for patients with either systolic or diastolic heart failure (96a). Diastolic dysfunction is more common in PLWH compared with people without HIV. As such, the 2023 ADA guidelines recommend the use of an SGLT2 inhibitor with demonstrated benefit in patients with type 2 diabetes and heart failure (70a).

 

One small trial studied canagliflozin for 24 weeks in 8 obese PLWH with type 2 diabetes and hemoglobin A1c > 7% and observed improvements in weight and hemoglobin A1c at the end of the study compared to baseline (97).

 

Adverse side effects of SGLT2 inhibitors include genital mycotic and urinary tract infections. In addition, 55 post-marketing cases of Fournier’s gangrene have been reported between March 2013 to January 2019 (98). A risk factor for Fournier’s gangrene is HIV infection (99-101), especially a CD4 cell count < 200 cells/μL (102).

 

Insulin

 

The 2023 ADA guidelines recommend considering insulin initiation in the patient 1) with a hemoglobin A1c > 11%, 2) with signs and symptoms of catabolism, 3) who has a presentation concerning for type 1 diabetes, and/or 4) whose hemoglobin A1c is not at goal despite taking 2 to 3 medications for diabetes, in addition to a GLP-1 receptor agonist.

 

As with all patients with diabetes, side effects of insulin therapy to consider in a discussion with a patient with HIV disease and diabetes include the risks of hypoglycemia and weight gain.

 

Sequence of Initiating Diabetes Medications in PLWH

 

There are no specific guidelines for the treatment of diabetes in PLWH. The 2023 ADA guidelines recommend the treatment of type 2 diabetes be tailored to the individual patient and consider factors including cost of medications, weight loss goals, and comorbidities including chronic kidney disease, atherosclerotic cardiovascular disease, and heart failure. Healthy lifestyle interventions should be incorporated alongside the pharmacologic treatment of type 2 diabetes.

 

For those patients with type 2 diabetes and ASCVD or who are at high risk for ASCVD, treatment with either a GLP-1 receptor agonist or SGLT2 inhibitor is recommended. If A1c is not at goal, then adding an additional agent, 1) such as a SGLT2 inhibitor, if the patient is already on a GLP-1 receptor agonist, or a GLP-1 receptor agonist, if the patient is already on a SGLT2 inhibitor, or 2) a thiazolidinedione, is recommended (70a). For patients with type 2 diabetes and heart failure, a SGLT2 inhibitor is recommended, as noted above. For patients with type 2 diabetes and chronic kidney disease, a SGLT2 inhibitor, or if contraindicated, a GLP-1 receptor agonist is recommended. Finally, for those patients with type 2 diabetes for whom weight management is a priority, a medication with weight loss or weight neutral effects can be considered. With regards to weight loss, semaglutide and tirzepatide have the most effect, whereas metformin and DPP4 inhibitors are weight neutral. Dulaglutide and liraglutide have a high weight loss efficacy, and SGLT2 inhibitors have intermediate weight loss efficacy (70a).

In PLWH, an additional factor to consider is the possible interaction of a diabetes medication with specific ART.

 

MICROVASCULAR COMPLICATIONS ASSOCIATED WITH DIABETES IN PLWH

 

Peripheral Neuropathy

 

HIV is associated with multiple peripheral neuropathies (PNs), including a distal, symmetric polyneuropathy (DSPN) (103). The prevalence of HIV-associated PN among PLWH varies < 10% to upwards of 50%, with the wide variability in prevalence estimates secondary in part to differences in methods used to assess PN (104). Risk factors for HIV-associated DSPN include use of the NRTIs zalcitabine, didanosine, and stavudine (105). In addition, HIV-associated DSPN has been observed in PLWH within 1 year of HIV transmission and is significantly associated with evidence of immune activation present in the central nervous system (106).

 

HIV-associated PN, in addition to the risk of developing PN secondary to diabetes, may place PLWH and diabetes at increased risk of sequelae including falls, particularly in those PLWH with a detectable viral load (107), foot ulceration, and amputations.

 

In summary, in addition to considering the risk of diabetic peripheral neuropathy, the risk of HIV-associated PNs should be kept in mind in the assessment and treatment of PLWH with diabetes.

 

Nephropathy

 

The prevalence estimates of chronic kidney disease (CKD) in PLWH vary depending on geographic region, with 7.9% of PLWH affected in Africa (with the highest prevalence among African regions in West Africa at 22%), compared to 3.7% in Europe. In addition, prevalence estimates of CKD in PLWH are significantly greater among those individuals with co-morbid diabetes (108). In the MACS, two independent risk factors associated with greater odds of proteinuria were HIV+ serostatus with ART use, compared to HIV- serostatus, and a history of diabetes (109). Moreover, glomerular hyperfiltration, or a supranormal estimated glomerular filtration rate, is more prevalent among HIV+ men without CKD than HIV- men without CKD (110). Glomerular hyperfiltration has been reported to be an initial state of dysfunction seen in patients with diabetic kidney disease and proteinuria (111).

 

Other risk factors for CKD in PLWH include the following: recurrent acute kidney injury, African-American race, in part because of risk variants of the APOL1 gene, and persistent inflammation, even in the setting of ART (112-114). In addition, an HIV-associated nephropathy (HIVAN), which is characterized by several insults to the kidney, including focal and segmental glomerulosclerosis, exists. Certain ART drugs within the NRTI, NNRTI, and PI classes are also associated with renal injury (114). Among these are the NRTI tenofovir, which is associated with adverse kidney disease outcomes independent of diabetes (115), and the PI indinavir (116).

           

Retinopathy

 

Ocular opportunistic infections in PLWH include CMV retinitis and ischemic HIV retinopathy (117). In addition, patients with a history of CMV retinitis should be monitored periodically for retinitis recurrence by an ophthalmologist (118). However, these ocular opportunistic infections are less common in the setting of ART. However, retinal disease has been noted in PLWH on ART as part of a larger syndrome of the HIV-associated neuroretinal disorder (117), which has an incidence of more than 50% at 20 years after a diagnosis of AIDS (119). One study found that among HIV+ men with a median duration of ART use of 12 years and suppressed viremia did have a significant difference in total peripheral retinal thickness from HIV- men, although the long-term clinical relevance of this is unknown (117).

 

There is limited literature on the effect of co-morbid diabetes on HIV-associated retinal disease.

 

MODIFIABLE FACTORS OF MACROVASCULAR COMPLICATIONS ASSOCIATED WITH DIABETES IN PLWH

 

PLWH are at increased risk of developing atherosclerotic cardiovascular disease (ASCVD) compared to individuals without HIV, despite controlling for traditional cardiovascular (CV) risk factors such as diabetes (5,120). In addition, PLWH have a greater burden of traditional CV risk factors (120). Other non-traditional risk factors include some ART such as older generation PIs (121) and inflammation (122). As such, calculators developed for the general population to calculate ASCVD risk may not accurately capture risk in PLWH (123).

 

Aspirin

 

Recent American College of Cardiology/American Heart Association (ACC/AHA) recommend the use of aspirin for primary prevention of ASCVD in general population patients age 40 to 70 years with high ASCVD risk (124), and similarly the American Diabetes Association (ADA) guidelines also recommend aspirin use in select patients with diabetes < 70 years of age with high ASCVD risk and low bleeding risk (125). However, evidence has shown that aspirin use is lower in PLWH with CV risk factors than in HIV- individuals (126), and PLWH are less likely to be prescribed aspirin for primary prevention than HIV- individuals (127).

 

Blood Pressure

 

The prevalence of hypertension globally in PLWH is substantial. PLWH have several risk factors for developing hypertension, including a greater prevalence of smoking (as noted below) (128,129) and ART use (130). The relationship of ART to hypertension is thought to be in part from its association to weight gain. Moreover, in the MACS, ART use was associated with greater systolic hypertension but not diastolic hypertension. A mechanism for this could be a change in arterial compliance as a consequence of ART use (131).

 

The 2023 ADA guidelines recommend a goal blood pressure of < 130/80 (132).

 

Cholesterol

 

Dyslipidemia is seen in both untreated PLWH and PLWH on ART (133,134). The decision to initiate a statin in a patient living with HIV for primary prevention should take into account the patient’s HIV serostatus, especially in those patients with a 10-year ASCVD risk of 5 to < 20% (124).

 

The primary prevention of atherosclerotic cardiovascular disease is a clinically important topic. The Evaluating the Use of Pitavastatin to Reduce Cardiovascular Disease in HIV-Infected Adults (REPRIEVE) study is a multicenter, international, randomized clinical trial on PWH on ART, in which participants with a low to moderate risk of ASCVD were randomized to either pitavastatin daily or placebo (134a). In March 2023, the Data Safety and Monitoring Board recommended premature closure of the study based on the observed efficacy of the study treatment to reduce the primary endpoint of major adverse cardiovascular events (MACE) by 35% relative to the placebo. The incidence of a major adverse cardiovascular event was 4.81 per 1000 person-years in the pitavastatin group and 7.32 per 1000 person-years in the placebo group (hazard ratio, 0.65; 95% confidence interval, 0.48 to 0.90; P = 0.002). Of note, similar to other studies the risk of diabetes was increased in the pitavastatin group (diabetes mellitus occurred in 5.3% in pitavastatin group and 4.0% in the control group. These results suggest that many PLWH should be on statin therapy.

 

Cigarette Smoking

 

Cigarette smoking is more prevalent in PLWH than in individuals without HIV (129), and life expectancy is estimated to be lower in HIV+ men and women who are current smokers at the time of HIV care initiation than in HIV+ men and women who are former or never smokers, with a 2.9 year gain in life expectancy at 10 years after HIV care initiation (135). The ADA guidelines recommend that people with diabetes not smoke cigarettes and that smoking cessation treatment be offered for those patients with diabetes who do smoke (136).

 

SUMMARY

 

In summary, PLWH have unique risk factors that increase their risk of diabetes. Factors to take into consideration in the treatment of PLWH include the following: type of ART, evidence of lipodystrophy, co-infection with other viruses, and overweight or obese state. A caveat is that HbA1c may be inaccurate in diagnosing and monitoring diabetes in PLWH. PLWH have additional risk factors for developing microvascular complications of diabetes. Some glycemic agents may interact with ART, and other glycemic agents may have unwanted effects, including weight gain, that should be addressed in a patient-provider discussion. Finally, additional modifiable cardiovascular risk factors, including hypertension and smoking, should be addressed in the comprehensive treatment of diabetes in PLWH.

           

REFERENCES

 

  1. World Health Organization. https://www.who.int/hiv/data/en/. Accessed July 3, 2019.
  2. Centers for Disease Control and Prevention. https://www.cdc.gov/nchhstp/newsroom/docs/factsheets/HIV-Incidence-Fact-Sheet_508.pdf. Accessed July 3, 2019.
  3. Chu C, Selwyn PA. An epidemic in evolution: the need for new models of HIV care in the chronic disease era. J Urban Health. 2011;88(3):556-566.
  4. Duncan AD, Goff LM, Peters BS. Type 2 diabetes prevalence and its risk factors in HIV: A cross-sectional study. PLoS One. 2018;13(3):e0194199.
  5. Freiberg MS, Chang CC, Kuller LH, Skanderson M, Lowy E, Kraemer KL, et al. HIV infection and the risk of acute myocardial infarction. JAMA Intern Med. 2013;173(8):614-622.
  6. Mallon PW. Aging with HIV: osteoporosis and fractures. Curr Opin HIV AIDS. 2014;9(4):428-435.
  7. Mohan V DM. The metabolic syndrome in developing countries. Diabetes Voice. 2006;51:15-17.
  8. Naidu S, Ponnampalvanar S, Kamaruzzaman SB, Kamarulzaman A. Prevalence of Metabolic Syndrome Among People Living with HIV in Developing Countries: A Systematic Review. AIDS Patient Care STDS. 2017;31(1):1-13.
  9. Brown TT, Cole SR, Li X, Kingsley LA, Palella FJ, Riddler SA, et al. Antiretroviral therapy and the prevalence and incidence of diabetes mellitus in the multicenter AIDS cohort study. Arch Intern Med. 2005;165(10):1179-1184.
  10. Ledergerber B, Furrer H, Rickenbach M, Lehmann R, Elzi L, Hirschel B, et al. Factors associated with the incidence of type 2 diabetes mellitus in HIV-infected participants in the Swiss HIV Cohort Study. Clin Infect Dis. 2007;45(1):111-119.
  11. De Wit S, Sabin CA, Weber R, Worm SW, Reiss P, Cazanave C, et al. Incidence and risk factors for new-onset diabetes in HIV-infected patients: the Data Collection on Adverse Events of Anti-HIV Drugs (D:A:D) study. Diabetes Care. 2008;31(6):1224-1229.
  12. Cho NH, Shaw JE, Karuranga S, Huang Y, da Rocha Fernandes JD, Ohlrogge AW, et al. IDF Diabetes Atlas: Global estimates of diabetes prevalence for 2017 and projections for 2045. Diabetes Res Clin Pract. 2018;138:271-281.
  13. Cahn P, Leite O, Rosales A, Cabello R, Alvarez CA, Seas C, et al. Metabolic profile and cardiovascular risk factors among Latin American HIV-infected patients receiving HAART. Braz J Infect Dis. 2010;14(2):158-166.
  14. Ngatchou W, Lemogoum D, Ndobo P, Yagnigni E, Tiogou E, Nga E, et al. Increased burden and severity of metabolic syndrome and arterial stiffness in treatment-naive HIV+ patients from Cameroon. Vasc Health Risk Manag. 2013;9:509-516.
  15. Ali MK, Magee MJ, Dave JA, Ofotokun I, Tungsiripat M, Jones TK, et al. HIV and metabolic, body, and bone disorders: what we know from low- and middle-income countries. J Acquir Immune Defic Syndr. 2014;67 Suppl 1:S27-39.
  16. Patel P, Rose CE, Collins PY, Nuche-Berenguer B, Sahasrabuddhe VV, Peprah E, et al. Noncommunicable diseases among HIV-infected persons in low-income and middle-income countries: a systematic review and meta-analysis. AIDS. 2018;32 Suppl 1:S5-S20.
  17. HIV/AIDS Historical Time Line 1995-1999. https://www.fda.gov/patients/hiv-timeline-and-history-approvals/hivaids-historical-time-line-1995-1999. Accessed July 5, 2019.
  18. Tsiodras S, Mantzoros C, Hammer S, Samore M. Effects of protease inhibitors on hyperglycemia, hyperlipidemia, and lipodystrophy: a 5-year cohort study. Arch Intern Med. 2000;160(13):2050-2056.
  19. Markowitz M, Saag M, Powderly WG, Hurley AM, Hsu A, Valdes JM, et al. A preliminary study of ritonavir, an inhibitor of HIV-1 protease, to treat HIV-1 infection. N Engl J Med. 1995;333(23):1534-1539.
  20. Cameron DW, Heath-Chiozzi M, Danner S, Cohen C, Kravcik S, Maurath C, et al. Randomised placebo-controlled trial of ritonavir in advanced HIV-1 disease. The Advanced HIV Disease Ritonavir Study Group. Lancet. 1998;351(9102):543-549.
  21. Carr A, Samaras K, Burton S, Law M, Freund J, Chisholm DJ, et al. A syndrome of peripheral lipodystrophy, hyperlipidaemia and insulin resistance in patients receiving HIV protease inhibitors. AIDS. 1998;12(7):F51-58.
  22. Woerle HJ, Mariuz PR, Meyer C, Reichman RC, Popa EM, Dostou JM, et al. Mechanisms for the deterioration in glucose tolerance associated with HIV protease inhibitor regimens. Diabetes. 2003;52(4):918-925.
  23. Hertel J, Struthers H, Horj CB, Hruz PW. A structural basis for the acute effects of HIV protease inhibitors on GLUT4 intrinsic activity. J Biol Chem. 2004;279(53):55147-55152.
  24. Lee GA, Mafong DD, Noor MA, Lo JC, Mulligan K, Schwarz JM, et al. HIV protease inhibitors increase adiponectin levels in HIV-negative men. J Acquir Immune Defic Syndr. 2004;36(1):645-647.
  25. Xu A, Yin S, Wong L, Chan KW, Lam KS. Adiponectin ameliorates dyslipidemia induced by the human immunodeficiency virus protease inhibitor ritonavir in mice. Endocrinology. 2004;145(2):487-494.
  26. Holodniy M, Hornberger J, Rapoport D, Robertus K, MaCurdy TE, Lopez J, et al. Relationship between antiretroviral prescribing patterns and treatment guidelines in treatment-naive HIV-1-infected US veterans (1992-2004). J Acquir Immune Defic Syndr. 2007;44(1):20-29.
  27. Research CfDEa. https://www.accessdata.fda.gov/drugsatfda_docs/nda/2003/21-567_Reyataz_Approv.pdf. Accessed July 5, 2019.
  28. Hresko RC, Hruz PW. HIV protease inhibitors act as competitive inhibitors of the cytoplasmic glucose binding site of GLUTs with differing affinities for GLUT1 and GLUT4. PLoS One. 2011;6(9):e25237.
  29. Noor MA, Flint OP, Maa JF, Parker RA. Effects of atazanavir/ritonavir and lopinavir/ritonavir on glucose uptake and insulin sensitivity: demonstrable differences in vitro and clinically. AIDS. 2006;20(14):1813-1821.
  30. Spagnuolo V, Galli L, Poli A, Salpietro S, Gianotti N, Piatti P, et al. Associations of statins and antiretroviral drugs with the onset of type 2 diabetes among HIV-1-infected patients. BMC Infect Dis. 2017;17(1):43.
  31. Fleischman A, Johnsen S, Systrom DM, Hrovat M, Farrar CT, Frontera W, et al. Effects of a nucleoside reverse transcriptase inhibitor, stavudine, on glucose disposal and mitochondrial function in muscle of healthy adults. Am J Physiol Endocrinol Metab. 2007;292(6):E1666-1673.
  32. Guidelines for the Use of Antiretroviral Agents in Adults and Adolescents with HIV. https://aidsinfo.nih.gov/guidelines/html/1/adult-and-adolescent-arv-guidelines/11/what-to-start. Accessed July 5, 2019.
  33. Randell PA, Jackson AG, Zhong L, Yale K, Moyle GJ. The effect of tenofovir disoproxil fumarate on whole-body insulin sensitivity, lipids and adipokines in healthy volunteers. Antivir Ther. 2010;15(2):227-233.

33A.  Wang H, Lu X, Yang X, Xu N. The efficacy and safety of tenofovir alafenamide versus tenofovir disoproxil fumarate in antiretroviral regimens for HIV-1 therapy: Meta-analysis. Medicine (Baltimore). 2016 Oct;95(41):e5146.

33B.  Sax, P. E. , Erlandson, K. M. , Lake, J. E. , Mccomsey, G. A. , Orkin, C. , Esser, S. , Brown, T. T. , Rockstroh, J. K. , Wei, X. , Carter, C. C. , Zhong, L. , Brainard, D. M. , Melbourne, K. , Das, M. , Stellbrink, H. , Post, F. A. , Waters, L. & Koethe, J. R. 2020. Weight Gain Following Initiation of Antiretroviral Therapy. Clinical Infectious Diseases, 71 (6), 1379-1389.

33C.  McCann K, Shah S, Hindley L, Hill A, Qavi A, Simmons B, Serenata C, Sokhela S, Venter WDF. Implications of weight gain with newer anti-retrovirals: 10-year predictions of cardiovascular disease and diabetes. AIDS. 2021 Aug 1;35(10):1657-1665.

  1. Hulgan T. Factors Associated With Insulin Resistance in Adults With HIV Receiving Contemporary Antiretroviral Therapy: a Brief Update. Curr HIV/AIDS Rep. 2018;15(3):223-232.
  2. Lennox JL, DeJesus E, Lazzarin A, Pollard RB, Madruga JV, Berger DS, et al. Safety and efficacy of raltegravir-based versus efavirenz-based combination therapy in treatment-naive patients with HIV-1 infection: a multicentre, double-blind randomised controlled trial. Lancet. 2009;374(9692):796-806.
  3. DeJesus E, Rockstroh JK, Henry K, Molina JM, Gathe J, Ramanathan S, et al. Co-formulated elvitegravir, cobicistat, emtricitabine, and tenofovir disoproxil fumarate versus ritonavir-boosted atazanavir plus co-formulated emtricitabine and tenofovir disoproxil fumarate for initial treatment of HIV-1 infection: a randomised, double-blind, phase 3, non-inferiority trial. Lancet. 2012;379(9835):2429-2438.
  4. Raffi F, Rachlis A, Stellbrink HJ, Hardy WD, Torti C, Orkin C, et al. Once-daily dolutegravir versus raltegravir in antiretroviral-naive adults with HIV-1 infection: 48 week results from the randomised, double-blind, non-inferiority SPRING-2 study. Lancet. 2013;381(9868):735-743.
  5. Norwood J, Turner M, Bofill C, Rebeiro P, Shepherd B, Bebawy S, et al. Brief Report: Weight Gain in Persons With HIV Switched From Efavirenz-Based to Integrase Strand Transfer Inhibitor-Based Regimens. J Acquir Immune Defic Syndr. 2017;76(5):527-531.
  6. McLaughlin M, Walsh S, Galvin S. Dolutegravir-induced hyperglycaemia in a patient living with HIV. J Antimicrob Chemother. 2018;73(1):258-260.
  7. Fong PS, Flynn DM, Evans CD, Korthuis PT. Integrase strand transfer inhibitor-associated diabetes mellitus: A case report. Int J STD AIDS. 2017;28(6):626-628.
  8. Dirajlal-Fargo S, Moser C, Brown TT, Kelesidis T, Dube MP, Stein JH, et al. Changes in Insulin Resistance After Initiation of Raltegravir or Protease Inhibitors With Tenofovir-Emtricitabine: AIDS Clinical Trials Group A5260s. Open Forum Infect Dis. 2016;3(3):ofw174.

41A.  Calmy A, Tovar Sanchez T, Kouanfack C, Mpoudi-Etame M, Leroy S, Perrineau S, Lantche Wandji M, Tetsa Tata D, Omgba Bassega P, Abong Bwenda T, Varloteaux M, Tongo M, Mpoudi-Ngolé E, Montoyo A, Mercier N, LeMoing V, Peeters M, Reynes J, Delaporte E; Dolutegravir-based and low-dose efavirenz-based regimen for the initial treatment of HIV-1 infection (NAMSAL): week 96 results from a two-group, multicentre, randomised, open label, phase 3 non-inferiority trial in Cameroon. New Antiretroviral and Monitoring Strategies in HIV-infected Adults in Low-Income Countries (NAMSAL) ANRS 12313 Study Group. Lancet HIV. 2020 Oct;7(10):e677-e687.

41B.  Rebeiro PF, Jenkins CA, Bian A, Lake JE, Bourgi K, Moore RD, Horberg MA, Matthews WC, Silverberg MJ, Thorne J, Mayor AM, Lima VD, Palella FJ, Saag MS, Althoff KN, Gill MJ, Wong C, Klein MB, Crane HM, Marconi VC, Shepherd BE, Sterling TR, Koethe JR. Risk of Incident Diabetes Mellitus, Weight Gain, and Their Relationships With Integrase Inhibitor-Based Initial Antiretroviral Therapy Among Persons With Human Immunodeficiency Virus in the United States and Canada. Clin Infect Dis. 2021 Oct 5;73(7):e2234-e2242.

41C.  Jung I, Tu-Sekine B, Jin S, Anokye-Danso F, Ahima RS, Brown TT, Kim SF. Dolutegravir Suppresses Thermogenesis via Disrupting Uncoupling Protein 1 Expression and Mitochondrial Function in Brown/Beige Adipocytes in Preclinical Models. J Infect Dis. 2022 Nov 1;226(9):1626-1636.

  1. Baril JG, Junod P, Leblanc R, Dion H, Therrien R, Laplante F, et al. HIV-associated lipodystrophy syndrome: A review of clinical aspects. Can J Infect Dis Med Microbiol. 2005;16(4):233-243.
  2. Carr A. HIV protease inhibitor-related lipodystrophy syndrome. Clin Infect Dis. 2000;30 Suppl 2:S135-142.
  3. Mallal SA, John M, Moore CB, James IR, McKinnon EJ. Contribution of nucleoside analogue reverse transcriptase inhibitors to subcutaneous fat wasting in patients with HIV infection. AIDS. 2000;14(10):1309-1316.
  4. Gallant JE, Staszewski S, Pozniak AL, DeJesus E, Suleiman JM, Miller MD, et al. Efficacy and safety of tenofovir DF vs stavudine in combination therapy in antiretroviral-naive patients: a 3-year randomized trial. JAMA. 2004;292(2):191-201.
  5. Nansseu JR, Bigna JJ, Kaze AD, Noubiap JJ. Incidence and Risk Factors for Prediabetes and Diabetes Mellitus Among HIV-infected Adults on Antiretroviral Therapy: A Systematic Review and Meta-analysis. Epidemiology. 2018;29(3):431-441.
  6. Putcharoen O, Wattanachanya L, Sophonphan J, Siwamogsatham S, Sapsirisavat V, Gatechompol S, et al. New-onset diabetes in HIV-treated adults: predictors, long-term renal and cardiovascular outcomes. AIDS. 2017;31(11):1535-1543.
  7. Mulligan K, Parker RA, Komarow L, Grinspoon SK, Tebas P, Robbins GK, et al. Mixed patterns of changes in central and peripheral fat following initiation of antiretroviral therapy in a randomized trial. J Acquir Immune Defic Syndr. 2006;41(5):590-597.
  8. Martin A, Smith DE, Carr A, Ringland C, Amin J, Emery S, et al. Reversibility of lipoatrophy in HIV-infected patients 2 years after switching from a thymidine analogue to abacavir: the MITOX Extension Study. AIDS. 2004;18(7):1029-1036.
  9. Prevention CfDCa. Coinfection with HIV and Viral Hepatitis. https://www.cdc.gov/hepatitis/hiv-hepatitis-coinfection.htm. Accessed July 12, 2019.
  10. Butt AA, Aslam S, Yan P, Shaikh OS, Abou-Samra A-B. Incident Diabetes and Glucose Control after HCV Treatment with DAAs in Erchives. Paper presented at: Conference on Retroviruses and Opportunistic Infections 2019; Seattle, Washington.
  11. Kumar S, Samaras K. The Impact of Weight Gain During HIV Treatment on Risk of Pre-diabetes, Diabetes Mellitus, Cardiovascular Disease, and Mortality. Front Endocrinol (Lausanne). 2018;9:705.
  12. Tate T, Willig AL, Willig JH, Raper JL, Moneyham L, Kempf MC, et al. HIV infection and obesity: where did all the wasting go? Antivir Ther. 2012;17(7):1281-1289.
  13. Herrin M, Tate JP, Akgun KM, Butt AA, Crothers K, Freiberg MS, et al. Weight Gain and Incident Diabetes Among HIV-Infected Veterans Initiating Antiretroviral Therapy Compared With Uninfected Individuals. J Acquir Immune Defic Syndr. 2016;73(2):228-236.
  14. Levy ME, Greenberg AE, Hart R, Powers Happ L, Hadigan C, Castel A, et al. High burden of metabolic comorbidities in a citywide cohort of HIV outpatients: evolving health care needs of people aging with HIV in Washington, DC. HIV Med. 2017;18(10):724-735.
  15. Semu H, Zack RM, Liu E, Hertzmark E, Spiegelman D, Sztam K, et al. Prevalence and Risk Factors for Overweight and Obesity among HIV-Infected Adults in Dar es Salaam, Tanzania. J Int Assoc Provid AIDS Care. 2016;15(6):512-521.
  16. Obry-Roguet V, Bregigeon S, Cano CE, Lions C, Zaegel-Faucher O, Laroche H, et al. Risk factors associated with overweight and obesity in HIV-infected people: Aging, behavioral factors but not cART in a cross-sectional study. Medicine (Baltimore). 2018;97(23):e10956.
  17. Douek DC, Roederer M, Koup RA. Emerging concepts in the immunopathogenesis of AIDS. Annu Rev Med. 2009;60:471-484.
  18. Zicari S, Sessa L, Cotugno N, Ruggiero A, Morrocchi E, Concato C, et al. Immune Activation, Inflammation, and Non-AIDS Co-Morbidities in HIV-Infected Patients under Long-Term ART. Viruses. 2019;11(3).
  19. Wada NI, Jacobson LP, Margolick JB, Breen EC, Macatangay B, Penugonda S, et al. The effect of HAART-induced HIV suppression on circulating markers of inflammation and immune activation. AIDS. 2015;29(4):463-471.
  20. Neuhaus J, Jacobs DR, Jr., Baker JV, Calmy A, Duprez D, La Rosa A, et al. Markers of inflammation, coagulation, and renal function are elevated in adults with HIV infection. J Infect Dis. 2010;201(12):1788-1795.
  21. Brown TT, Tassiopoulos K, Bosch RJ, Shikuma C, McComsey GA. Association between systemic inflammation and incident diabetes in HIV-infected patients after initiation of antiretroviral therapy. Diabetes Care. 2010;33(10):2244-2249.
  22. American Diabetes A. 2. Classification and Diagnosis of Diabetes: Standards of Medical Care in Diabetes-2019. Diabetes Care. 2019;42(Suppl 1):S13-S28.
  23. Diop ME, Bastard JP, Meunier N, Thevenet S, Maachi M, Capeau J, et al. Inappropriately low glycated hemoglobin values and hemolysis in HIV-infected patients. AIDS Res Hum Retroviruses. 2006;22(12):1242-1247.
  24. Polgreen PM, Putz D, Stapleton JT. Inaccurate glycosylated hemoglobin A1C measurements in human immunodeficiency virus-positive patients with diabetes mellitus. Clin Infect Dis. 2003;37(4):e53-56.
  25. Geene D, Sudre P, Anwar D, Goehring C, Saaidia A, Hirschel B. Causes of macrocytosis in HIV-infected patients not treated with zidovudine. Swiss HIV Cohort Study. J Infect. 2000;40(2):160-163.
  26. Slama L, Palella FJ, Jr., Abraham AG, Li X, Vigouroux C, Pialoux G, et al. Inaccuracy of haemoglobin A1c among HIV-infected men: effects of CD4 cell count, antiretroviral therapies and haematological parameters. J Antimicrob Chemother. 2014;69(12):3360-3367.
  27. Aberg JA, Gallant JE, Ghanem KG, Emmanuel P, Zingman BS, Horberg MA, et al. Primary care guidelines for the management of persons infected with HIV: 2013 update by the HIV Medicine Association of the Infectious Diseases Society of America. Clin Infect Dis. 2014;58(1):1-10.
  28. Eckhardt BJ, Holzman RS, Kwan CK, Baghdadi J, Aberg JA. Glycated Hemoglobin A(1c) as screening for diabetes mellitus in HIV-infected individuals. AIDS Patient Care STDS. 2012;26(4):197-201.
  29. Han JH, Crane HM, Bellamy SL, Frank I, Cardillo S, Bisson GP, et al. HIV infection and glycemic response to newly initiated diabetic medical therapy. AIDS. 2012;26(16):2087-2095.

70A.  ElSayed NA, Aleppo G, Aroda VR, Bannuru RR, Brown FM, Bruemmer D, Collins BS, Hilliard ME, Isaacs D, Johnson EL, Kahan S, Khunti K, Leon J, Lyons SK, Perry ML, Prahalad P, Pratley RE, Seley JJ, Stanton RC, Gabbay RA, on behalf of the American Diabetes Association. 9. Pharmacologic Approaches to Glycemic Treatment: Standards of Care in Diabetes-2023. Diabetes Care. 2023 Jan 1;46(Suppl 1):S140-S157.

70B.  Samson SL, Vellanki P, Blonde L, Christofides EA, Galindo RJ, Hirsch IB, Isaacs SD, Izuora KE, Low Wang CC, Twining CL, Umpierrez GE, Valencia WM. American Association of Clinical Endocrinology Consensus Statement: Comprehensive Type 2 Diabetes Management Algorithm - 2023 Update. Endocr Pract. 2023 May;29(5):305-340.

70C.  Jaggers JR, Prasad VK, Dudgeon WD, Blair SN, Sui X, Burgess S, Hand GA. Associations between physical activity and sedentary time on components of metabolic syndrome among adults with HIV. AIDS Care. 2014;26(11):1387-92.

70D.  Roos R, Myezwa H, van Aswegen H, Musenge E. Effects of an education and home-based pedometer walking program on ischemic heart disease risk factors in people infected with HIV: a randomized trial. J Acquir Immune Defic Syndr. 2014 Nov 1;67(3):268-76.

70E.  Rehm KE, Konkle-Parker D. Physical activity levels and perceived benefits and barriers to physical activity in HIV-infected women living in the deep south of the United States. AIDS Care. 2016 Sep;28(9):1205-10.

70F.  Vancampfort D, Mugisha J, Richards J, De Hert M, Lazzarotto AR, Schuch FB, Probst M, Stubbs B. Dropout from physical activity interventions in people living with HIV: a systematic review and meta-analysis. AIDS Care. 2017 May;29(5):636-643.

  1. American Diabetes Association. 9. Pharmacologic Approaches to Glycemic Treatment: Standards of Medical Care in Diabetes-2019. Diabetes Care. 2019;42(Suppl 1):S90-S102.
  2. Hadigan C, Corcoran C, Basgoz N, Davis B, Sax P, Grinspoon S. Metformin in the treatment of HIV lipodystrophy syndrome: A randomized controlled trial. JAMA. 2000;284(4):472-477.
  3. Song IH, Zong J, Borland J, Jerva F, Wynne B, Zamek-Gliszczynski MJ, et al. The Effect of Dolutegravir on the Pharmacokinetics of Metformin in Healthy Subjects. J Acquir Immune Defic Syndr. 2016;72(4):400-407.
  4. https://www.accessdata.fda.gov/drugsatfda_docs/label/2013/204790lbl.pdf. Tdpi. Accessed July 8, 2019.
  5. Gervasoni C, Minisci D, Clementi E, Rizzardini G, Cattaneo D. How Relevant is the Interaction Between Dolutegravir and Metformin in Real Life? J Acquir Immune Defic Syndr. 2017;75(1):e24-e26.
  6. May M, Schindler C. Clinically and pharmacologically relevant interactions of antidiabetic drugs. Ther Adv Endocrinol Metab. 2016;7(2):69-83.
  7. Fichtenbaum CJ, Gerber JG. Interactions between antiretroviral drugs and drugs used for the therapy of the metabolic complications encountered during HIV infection. Clin Pharmacokinet. 2002;41(14):1195-1211.
  8. Han JH, Gordon K, Womack JA, Gibert CL, Leaf DA, Rimland D, et al. Comparative Effectiveness of Diabetic Oral Medications Among HIV-Infected and HIV-Uninfected Veterans. Diabetes Care. 2017;40(2):218-225.
  9. Sutinen J, Hakkinen AM, Westerbacka J, Seppala-Lindroos A, Vehkavaara S, Halavaara J, et al. Rosiglitazone in the treatment of HAART-associated lipodystrophy--a randomized double-blind placebo-controlled study. Antivir Ther. 2003;8(3):199-207.
  10. Hadigan C, Yawetz S, Thomas A, Havers F, Sax PE, Grinspoon S. Metabolic effects of rosiglitazone in HIV lipodystrophy: a randomized, controlled trial. Ann Intern Med. 2004;140(10):786-794.
  11. Cavalcanti RB, Raboud J, Shen S, Kain KC, Cheung A, Walmsley S. A randomized, placebo-controlled trial of rosiglitazone for HIV-related lipoatrophy. J Infect Dis. 2007;195(12):1754-1761.
  12. Grinspoon S. Use of thiazolidinediones in HIV-infected patients: what have we learned? J Infect Dis. 2007;195(12):1731-1733.
  13. van Wijk JP, de Koning EJ, Cabezas MC, op't Roodt J, Joven J, Rabelink TJ, et al. Comparison of rosiglitazone and metformin for treating HIV lipodystrophy: a randomized trial. Ann Intern Med. 2005;143(5):337-346.
  14. Pioglitazone - FDA label. https://wwwaccessdatafdagov/drugsatfda_docs/label/2011/021073s043s044lblpdf. Accessed July 10, 2019.
  15. Makdissi A, Ghanim H, Vora M, Green K, Abuaysheh S, Chaudhuri A, et al. Sitagliptin exerts an antinflammatory action. J Clin Endocrinol Metab. 2012;97(9):3333-3341.
  16. Tremblay AJ, Lamarche B, Deacon CF, Weisnagel SJ, Couture P. Effects of sitagliptin therapy on markers of low-grade inflammation and cell adhesion molecules in patients with type 2 diabetes. Metabolism. 2014;63(9):1141-1148.
  17. Goodwin SR, Reeds DN, Royal M, Struthers H, Laciny E, Yarasheski KE. Dipeptidyl peptidase IV inhibition does not adversely affect immune or virological status in HIV infected men and women: a pilot safety study. J Clin Endocrinol Metab. 2013;98(2):743-751.
  18. Best C, Struthers H, Laciny E, Royal M, Reeds DN, Yarasheski KE. Sitagliptin Reduces Inflammation and Chronic Immune Cell Activation in HIV+ Adults With Impaired Glucose Tolerance. J Clin Endocrinol Metab. 2015;100(7):2621-2629.
  19. Dube MP, Chan ES, Lake JE, Williams B, Kinslow J, Landay A, et al. A Randomized, Double-Blinded, Placebo-Controlled Trial of Sitagliptin for Reducing Inflammation and Immune Activation in Treated and Suppressed HIV Infection. Clin Infect Dis. 2018.
  20. Marso SP, Daniels GH, Brown-Frandsen K, Kristensen P, Mann JF, Nauck MA, et al. Liraglutide and Cardiovascular Outcomes in Type 2 Diabetes. N Engl J Med. 2016;375(4):311-322.
  21. Marso SP, Bain SC, Consoli A, Eliaschewitz FG, Jodar E, Leiter LA, et al. Semaglutide and Cardiovascular Outcomes in Patients with Type 2 Diabetes. N Engl J Med. 2016;375(19):1834-1844.
  22. Diamant M, van Agtmael M. Liraglutide treatment in a patient with HIV and uncontrolled insulin-treated type 2 diabetes. Diabetes Care. 2012;35(5):e34.
  23. Gutierrez AD, Balasubramanyam A. Dysregulation of glucose metabolism in HIV patients: epidemiology, mechanisms, and management. Endocrine. 2012;41(1):1-10.
  24. Paik IJ, Kotler DP. The prevalence and pathogenesis of diabetes mellitus in treated HIV-infection. Best Pract Res Clin Endocrinol Metab. 2011;25(3):469-478.
  25. Wiviott SD, Raz I, Bonaca MP, Mosenzon O, Kato ET, Cahn A, et al. Dapagliflozin and Cardiovascular Outcomes in Type 2 Diabetes. N Engl J Med. 2019;380(4):347-357.
  26. Zinman B, Wanner C, Lachin JM, Fitchett D, Bluhmki E, Hantel S, et al. Empagliflozin, Cardiovascular Outcomes, and Mortality in Type 2 Diabetes. N Engl J Med. 2015;373(22):2117-2128.

96A.  Feingold KR. Oral and Injectable (Non-Insulin) Pharmacological Agents for the Treatment of Type 2 Diabetes. 2022 Aug 26. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrère B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Shah AS, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000–97.  

  1. Garcia de Lucas MD, Olalla J. Experience of using ISGTL-2 in patients with DM2 and HIV infection. Eur J Intern Med. 2017;41:e29.
  2. Bersoff-Matcha SJ, Chamberlain C, Cao C, Kortepeter C, Chong WH. Fournier Gangrene Associated With Sodium-Glucose Cotransporter-2 Inhibitors: A Review of Spontaneous Postmarketing Cases. Ann Intern Med. 2019.
  3. Elem B, Ranjan P. Impact of immunodeficiency virus (HIV) on Fournier's gangrene: observations in Zambia. Ann R Coll Surg Engl. 1995;77(4):283-286.
  4. Ngugi P, Magoha G, Nyaga P. Fournier's ganrene in the HIV era. Afr Health Sci. 2014;14(4):1063-1068.
  5. Chazan B, Chen Y, Raz R, Kopelman D, Katz Z, Kniznik D, et al. Fournier's gangrene as the initial presentation of HIV infection. Int J Infect Dis. 2007;11(2):184-185.
  6. Chalya PL, Igenge JZ, Mabula JB, Simbila S. Fournier's gangrene at a tertiary health facility in northwestern Tanzania: a single centre experiences with 84 patients. BMC Res Notes. 2015;8:481.
  7. Saylor D. Neurologic Complications of Human Immunodeficiency Virus Infection. Continuum (Minneap Minn). 2018;24(5, Neuroinfectious Disease):1397-1421.
  8. Ghosh S, Chandran A, Jansen JP. Epidemiology of HIV-related neuropathy: a systematic literature review. AIDS Res Hum Retroviruses. 2012;28(1):36-48.
  9. Simpson DM, Tagliati M. Nucleoside analogue-associated peripheral neuropathy in human immunodeficiency virus infection. J Acquir Immune Defic Syndr Hum Retrovirol. 1995;9(2):153-161.
  10. Wang SX, Ho EL, Grill M, Lee E, Peterson J, Robertson K, et al. Peripheral neuropathy in primary HIV infection associates with systemic and central nervous system immune activation. J Acquir Immune Defic Syndr. 2014;66(3):303-310.
  11. Erlandson KM, Zhang L, Ng DK, Althoff KN, Palella FJ, Jr., Kingsley LA, et al. Risk Factors for Falls, Falls With Injury, and Falls With Fracture Among Older Men With or at Risk of HIV Infection. J Acquir Immune Defic Syndr. 2019;81(4):e117-e126.
  12. Ekrikpo UE, Kengne AP, Bello AK, Effa EE, Noubiap JJ, Salako BL, et al. Chronic kidney disease in the global adult HIV-infected population: A systematic review and meta-analysis. PLoS One. 2018;13(4):e0195443.
  13. Palella FJ, Jr., Li X, Gupta SK, Estrella MM, Phair JP, Margolick JB, et al. Long-term kidney function, proteinuria, and associated risks among HIV-infected and uninfected men. AIDS. 2018;32(10):1247-1256.
  14. Ng DK, Jacobson LP, Brown TT, Palella FJ, Jr., Martinson JJ, Bolan R, et al. HIV therapy, metabolic and cardiovascular health are associated with glomerular hyperfiltration among men with and without HIV infection. AIDS. 2014;28(3):377-386.
  15. Tonneijck L, Muskiet MH, Smits MM, van Bommel EJ, Heerspink HJ, van Raalte DH, et al. Glomerular Hyperfiltration in Diabetes: Mechanisms, Clinical Significance, and Treatment. J Am Soc Nephrol. 2017;28(4):1023-1039.
  16. Wyatt CM, Arons RR, Klotman PE, Klotman ME. Acute renal failure in hospitalized patients with HIV: risk factors and impact on in-hospital mortality. AIDS. 2006;20(4):561-565.
  17. Naicker S, Aboud O, Gharbi MB. Epidemiology of acute kidney injury in Africa. Semin Nephrol. 2008;28(4):348-353.
  18. Hou J, Nast CC. Changing concepts of HIV infection and renal disease. Curr Opin Nephrol Hypertens. 2018;27(3):144-152.
  19. Scherzer R, Estrella M, Li Y, Choi AI, Deeks SG, Grunfeld C, et al. Association of tenofovir exposure with kidney disease risk in HIV infection. AIDS. 2012;26(7):867-875.
  20. Mocroft A, Kirk O, Gatell J, Reiss P, Gargalianos P, Zilmer K, et al. Chronic renal failure among HIV-1-infected patients. AIDS. 2007;21(9):1119-1127.
  21. Demirkaya N, Wit FW, van Den Berg TJ, Kooij KW, Prins M, Schlingemann RO, et al. HIV-Associated Neuroretinal Disorder in Patients With Well-Suppressed HIV-Infection: A Comparative Cohort Study. Invest Ophthalmol Vis Sci. 2016;57(3):1388-1397.
  22. Masur H, Brooks JT, Benson CA, Holmes KK, Pau AK, Kaplan JE, et al. Prevention and treatment of opportunistic infections in HIV-infected adults and adolescents: Updated Guidelines from the Centers for Disease Control and Prevention, National Institutes of Health, and HIV Medicine Association of the Infectious Diseases Society of America. Clin Infect Dis. 2014;58(9):1308-1311.
  23. Jabs DA, Drye L, Van Natta ML, Thorne JE, Holland GN, Studies of the Ocular Complications of ARG. Incidence and long-term outcomes of the human immunodeficiency virus neuroretinal disorder in patients with AIDS. Ophthalmology. 2015;122(4):760-768.
  24. Triant VA, Lee H, Hadigan C, Grinspoon SK. Increased acute myocardial infarction rates and cardiovascular risk factors among patients with human immunodeficiency virus disease. J Clin Endocrinol Metab. 2007;92(7):2506-2512.
  25. Worm SW, Sabin C, Weber R, Reiss P, El-Sadr W, Dabis F, et al. Risk of myocardial infarction in patients with HIV infection exposed to specific individual antiretroviral drugs from the 3 major drug classes: the data collection on adverse events of anti-HIV drugs (D:A:D) study. J Infect Dis. 2010;201(3):318-330.
  26. Duprez DA, Neuhaus J, Kuller LH, Tracy R, Belloso W, De Wit S, et al. Inflammation, coagulation and cardiovascular disease in HIV-infected individuals. PLoS One. 2012;7(9):e44454.
  27. Thompson-Paul AM, Lichtenstein KA, Armon C, Palella FJ, Jr., Skarbinski J, Chmiel JS, et al. Cardiovascular Disease Risk Prediction in the HIV Outpatient Study. Clin Infect Dis. 2016;63(11):1508-1516.
  28. Arnett DK, Blumenthal RS, Albert MA, Buroker AB, Goldberger ZD, Hahn EJ, et al. 2019 ACC/AHA Guideline on the Primary Prevention of Cardiovascular Disease: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. J Am Coll Cardiol. 2019.
  29. American Diabetes A. 9. Cardiovascular Disease and Risk Management: Standards of Medical Care in Diabetes-2018. Diabetes Care. 2018;41(Suppl 1):S86-S104.
  30. Suchindran S, Regan S, Meigs JB, Grinspoon SK, Triant VA. Aspirin Use for Primary and Secondary Prevention in Human Immunodeficiency Virus (HIV)-Infected and HIV-Uninfected Patients. Open Forum Infect Dis. 2014;1(3):ofu076.
  31. Ladapo JA, Richards AK, DeWitt CM, Harawa NT, Shoptaw S, Cunningham WE, et al. Disparities in the Quality of Cardiovascular Care Between HIV-Infected Versus HIV-Uninfected Adults in the United States: A Cross-Sectional Study. J Am Heart Assoc. 2017;6(11).
  32. Elf JL, Variava E, Chon S, Lebina L, Motlhaoleng K, Gupte N, et al. Prevalence and Correlates of Smoking Among People Living With HIV in South Africa. Nicotine Tob Res. 2018;20(9):1124-1131.
  33. Mdodo R, Frazier EL, Dube SR, Mattson CL, Sutton MY, Brooks JT, et al. Cigarette smoking prevalence among adults with HIV compared with the general adult population in the United States: cross-sectional surveys. Ann Intern Med. 2015;162(5):335-344.
  34. Xu Y, Chen X, Wang K. Global prevalence of hypertension among people living with HIV: a systematic review and meta-analysis. J Am Soc Hypertens. 2017;11(8):530-540.
  35. Seaberg EC, Munoz A, Lu M, Detels R, Margolick JB, Riddler SA, et al. Association between highly active antiretroviral therapy and hypertension in a large cohort of men followed from 1984 to 2003. AIDS. 2005;19(9):953-960.
  36. American Diabetes A. 10. Cardiovascular Disease and Risk Management: Standards of Medical Care in Diabetes-2019. Diabetes care. 2019;42(Suppl 1):S103-S123.
  37. Grunfeld C, Pang M, Doerrler W, Shigenaga JK, Jensen P, Feingold KR. Lipids, lipoproteins, triglyceride clearance, and cytokines in human immunodeficiency virus infection and the acquired immunodeficiency syndrome. J Clin Endocrinol Metab. 1992;74(5):1045-1052.
  38. Riddler SA, Smit E, Cole SR, Li R, Chmiel JS, Dobs A, et al. Impact of HIV infection and HAART on serum lipids in men. JAMA. 2003;289(22):2978-2982.

134A. Grinspoon SK, Fitch KV, Zanni MV, Fichtenbaum CJ, Umbleja T, Aberg JA, Overton ET, Malvestutto CD, Bloomfield GS, Currier JS, Martinez E, Roa JC, Diggs MR, Fulda ES, Paradis K, Wiviott SD, Foldyna B, Looby SE, Desvigne-Nickens P, Alston-Smith B, Leon-Cruz J, McCallum S, Hoffmann U, Lu MT, Ribaudo HJ, Douglas PS; REPRIEVE Investigators. Pitavastatin to Prevent Cardiovascular Disease in HIV Infection. N Engl J Med. 2023 Aug 24;389(8):687-699.

  1. Reddy KP, Parker RA, Losina E, Baggett TP, Paltiel AD, Rigotti NA, et al. Impact of Cigarette Smoking and Smoking Cessation on Life Expectancy Among People With HIV: A US-Based Modeling Study. J Infect Dis. 2016;214(11):1672-1681.
  2. American Diabetes A. 5. Lifestyle Management: Standards of Medical Care in Diabetes-2019. Diabetes Care. 2019;42(Suppl 1):S46-S60.

 

Dyslipidemia in Patients With Diabetes

ABSTRACT

Atherosclerotic cardiovascular disease (ASCVD) is a major cause of morbidity and mortality in both men and women with T1DM and T2DM. In patients with T1DM in good glycemic control, the lipid profile is very similar to the general population. In contrast, in patients with T2DM, even with good glycemic control, there are frequently lipid abnormalities (elevated TG and non-HDL-C, decreased HDL-C, and an increase in small dense LDL). In both T1DM and T2DM, poor glycemic control increases TG levels and decreases HDL-C levels with modest effects on LDL-C levels. Extensive studies have demonstrated that statins decrease ASCVD in patients with diabetes. Treatment with high doses of potent statins reduces ASCVD events to a greater extent than low dose statin therapy. Adding fibrates or niacin to statin therapy has not been shown to further decrease ASCVD events. In contrast, studies have shown that the combination of a statin and ezetimibe or a statin and a PCSK9 inhibitor result in a greater decrease in ASCVD events than statins alone. Studies have suggested that EPA, an omega-3-fatty acid, when added to statins also reduces ASCVD events but this result is controversial. In statin intolerant patients with T2DM bempedoic acid decreases ASCVD events. Current recommendations state that most patients with diabetes should be on statin therapy. In certain patients with diabetes ezetimibe, PCSK9 inhibitors, and bempedoic acid can play a role in reducing ASCVD.  

 

INTRODUCTION

 

Atherosclerotic cardiovascular disease (ASCVD) is a major cause of morbidity and mortality in both men and women with diabetes (1-5). In addition to coronary disease, ASCVD includes stroke and peripheral arterial disease (PAD).  PAD is common in diabetes, may be the first presentation of ASCVD and should be recognized as needing aggressive treatment of risk factors. The risk of ASCVD is increased approximately 2-fold in men and 3-4-fold in women (2-4,6,7). In the Framingham study, the annual rate of ASCVD was similar in men and women with diabetes, emphasizing that woman with diabetes need as aggressive preventive treatment as men with diabetes (2,6). In addition, several but not all studies, have shown that patients with diabetes with no history of ASCVD have a similar risk of having a myocardial infarction as non-diabetic patients who have a history of ASCVD, i.e., diabetes is an equivalent risk factor as a history of a previous cardiovascular event (8,9). The duration of diabetes and the presence of other risk factors or complications of diabetes likely determine whether a patient with diabetes has a risk equivalent to patients with a history of previous ASCVD events (10,11). In one study patients with T2DM who had the following risk factors within the target range, HbA1c, LDL-C, albuminuria, smoking, and blood pressure, the risk of an acute myocardial infarction or stroke was similar to individuals without diabetics (12). Moreover, numerous studies have shown that patients with diabetes who have ASCVD are at a very high risk of having another event, indicating that this population of patient’s needs especially aggressive preventive measures (1,8). This increased risk for the development of ASCVD in patients with diabetes is seen both in populations where the prevalence of ASCVD is high (Western societies) and low (for example, Japan) (2). However, in societies where the prevalence of ASCVD is low, the contribution of ASCVD as a cause of morbidity and mortality in patients with diabetes is relatively low compared to Western societies (2).

 

While the database is not as robust, the evidence indicates that patients with T1DM are also at high risk for the development of ASCVD (1,13-15). Interestingly, women with T1DM have twice the excess risk of fatal and nonfatal vascular events compared to men with T1DM (16,17). Additionally, developing T1DM at a young age increases the risk of ASCVD to a greater degree than late onset T1DM (17). Approximately 50% of patients with T1DM are obese or overweight and between 8% and 40% meet the criteria for the metabolic syndrome, which increases their risk of developing ASCVD (18).

 

While the development of diabetes at a young age increases the risk of ASCVD in patients with both T1DM and T2DM the deleterious impact is greater in patients with T2DM (19). Lastly, in patients with both T1DM and T2DM the presence of renal disease increases the risk of ASCVD (4,14). Of note is that the risk of developing ASCVD events in patients with diabetes has decreased recently, most likely due to better lipid and blood pressure control, which again reinforces the need to aggressively treat these risk factors in patients with diabetes (5,7,20). 

 

ROLE OF RISK FACTORS IN ASCVD

 

Numerous studies have demonstrated that the traditional risk factors for ASCVD play an important role in patients with diabetes (2,4,5,110). Patients with diabetes without other risk factors have a relatively low risk of ASCVD (in most studies higher than similar non-diabetic patients), whereas the increasing prevalence of other risk factors markedly increases the risk of developing ASCVD (2). The major reversible traditional risk factors are hypertension, cigarette smoking, and lipid abnormalities (2,4,5,14,111). Other risk factors include obesity (particularly visceral obesity), insulin resistance, small dense LDL, elevated TG, low HDL-C, procoagulant state (increased PAI-1, fibrinogen), family history of early ASCVD, homocysteine, Lp (a), renal disease, albuminuria, and inflammation (C-reactive protein, SAA, cytokines) (2,4,5,110,111). In the last decade, it has become clear that to reduce the risk of ASCVD in patients with diabetes, one will not only need to improve glycemic control but also address these other cardiovascular risk factors. In the remainder of this chapter, I will focus on the dyslipidemia that occurs in patients with diabetes.

 

ROLE OF LIPIDS IN ASCVD

 

As in non-diabetic populations, epidemiological studies have shown that increased LDL-C and non-HDL-C levels and decreased HDL-C levels are associated with an increased risk of ASCVD in patients with diabetes (2,4,110,111). In the UKPDS cohort LDL-C levels were the strongest predictor of coronary artery disease (112). While it is universally accepted that elevated levels of LDL-C and non-HDL-C cause atherosclerosis and ASCVD the role of HDL-C is uncertain. Genetic studies and studies of drugs that raise HDL-C have not supported low HDL-C levels as a causative factor for atherosclerosis (113). Rather it is currently thought that HDL function is associated with atherosclerosis risk and that this does not precisely correlate with HDL-C levels (113). In patients with diabetes, elevations in serum triglyceride (TG) levels also are associated with an increased risk of ASCVD (4,111,114). With regard to TG, it is not clear whether they are a causative factor for ASCVD or whether the elevation in TG is a marker for other abnormalities (4,111,114,115). Recent Mendelian randomization studies have provided support for the hypothesis that elevated TG levels play a causal role in atherosclerosis (115,116). Unfortunately, as will be discussed later in this chapter lowering TG levels in patients on statin therapy has not decreased cardiovascular events.

 

LIPID ABNORMALITIES IN PATIENTS WITH DIABETES

 

In patients with T1DM in good glycemic control, the lipid profile is very similar to lipid profiles in the general population (110). In some studies HDL-C levels are modestly increased in patients with T1DM (117). In contrast, in patients with T2DM, even when in good glycemic control, there are abnormalities in lipid levels (118-121). It is estimated that 30-60% of patients with T2DM have dyslipidemia (5,122). Specifically, patients with T2DM often have an increase in serum TG levels, increased VLDL and IDL, and decreased HDL-C levels. Non-HDL-C levels are increased due to the increase in VLDL and IDL. LDL-C levels are typically not markedly different than in normal subjects but there is an increase in small dense LDL, a lipoprotein particle that may be particularly pro-atherogenic (123). As a consequence, there are more LDL particles, which coupled with the increases in VLDL and IDL, leads to an increase in apolipoprotein B levels (118-121). Additionally, the postprandial increase in serum TG is accentuated and elevations in postprandial lipids may increase the risk of ASCVD (118-121).

 

It should be recognized that the lipid changes in patients with T2D are characteristic of the alterations in lipid profile seen in obesity and the metabolic syndrome (insulin resistance syndrome) (124). Since a high percentage of patients with T2DM are obese, insulin resistant, and have the metabolic syndrome, it is not surprising that the prevalence of increased TG and small dense LDL and decreased HDL-C is common in patients with T2DM even when these patients are in good glycemic control. Obesity is also accompanied by increased systemic inflammation. The increasing prevalence of obesity/overweight in patients with T1D will likely result in an increased prevalence of dyslipidemia in this population.

 

Studies have shown that the anti-oxidant and anti-inflammatory functions of HDL isolated from patients with T1DM and T2DM are reduced (117,125). Additionally, the ability of HDL to facilitate cholesterol efflux is reduced in patients with T1DM and T2DM (126,127). Together these findings indicate that HDL-C levels per se may not fully reflect risk of ASCVD in patients with diabetes and that HDL function is perturbed in patients with diabetes.

 

In both T1DM and T2DM, poor glycemic control increases serum TG levels, VLDL, and IDL, and decreases HDL-C levels (119). Poor glycemic control can also result in a modest increase in LDL-C, which because of the elevation in TG is often in the small dense LDL subfraction. It is therefore important to optimize glycemic control in patients with diabetes because this will have secondary beneficial effects on lipid levels.

 

Lp(a) levels are usually within the normal range in patients with T1DM and T2DM (128). Some studies have observed no impact of diabetes mellitus on Lp(a) concentrations while other studies reported an elevation or a decrease in Lp(a) concentrations (128). The development of microalbuminuria and the onset of renal disease are associated with an increase in Lp (a) levels (129). Of note low Lp(a) levels are associated with an increased risk of developing T2DM (128). A recent very large case control study found that an Lp(a) concentration in the bottom 10% increases T2DM risk (130).

 

Table 1. Lipid Abnormalities in Patients with Diabetes

T1DM

Lipid profile is similar to controls if glycemic control is good

T2DM

Increased TG, VLDL, IDL, and non-HDL-C. Decreased HDL-C. Normal LDL-C but increase in small dense LDL, LDL particle number, and apolipoprotein B.

Poor glycemic control

Increased TG, VLDL, IDL, and non-HDL-C.  Decreased HDL-C. Modest increase in LDL-C with increase in small dense LDL, LDL particle number, and apolipoprotein B.

 

EFFECT OF GLUCOSE LOWERING DRUGS ON LIPIDS

 

Some therapies used to improve glycemic control may have an impact on lipid levels above and beyond their effects on glucose metabolism. In reviewing the literature, it is often very difficult to separate improvements in glycemic control vs. direct effects of drugs. Additionally, many of the changes induced by drug therapy result in only small changes in LDL-C, HDL-C, and TG levels, are variable from study to study, and are of questionable clinical significance. Insulin, sulfonylureas, meglinitides, DPP4 inhibitors, and alpha-glucosidase inhibitors do not appear to markedly alter fasting lipid profiles other than by improving glucose control (there are data indicating that DPP4 inhibitors and acarbose decrease postprandial triglyceride excursions, but they do not markedly alter fasting lipid levels) (131). In contrast, metformin, thiazolidinediones, GLP1 receptor agonists, bromocriptine-QR, and SGLT2 inhibitors have effects independent of glycemic control on serum lipid levels (table 2).

 

Metformin may decrease serum TG levels and LDL-C levels without altering HDL-C levels (131). In a meta-analysis of 37 trials with 2,891 patients, metformin decreased TG by 11.4mg/dL when compared with control treatment (p=0.003) (132). In an analysis of 24 trials with 1,867 patients, metformin decreased LDL-C by 8.4mg/dL compared to control treatment (p<0.001) (132). In contrast, metformin did not significantly alter HDL-C levels (132). It should be noted that in the Diabetes Prevention Program 3,234 individuals with impaired glucose metabolism were randomized to placebo, intensive lifestyle, or metformin therapy. In the metformin therapy group no significant changes were noted in TG, LDL-C, or HDL-C levels compared to the placebo group (133). Thus, metformin may have small effects on lipid levels.    

 

The effect of thiazolidinediones depends on which agent is used. Rosiglitazone increases serum LDL-C levels, increases HDL-C levels, and only decreases serum TG if the baseline TG levels are high (131). In contrast, pioglitazone has less impact on LDL-C levels, but increases HDL-C levels, and decreases TG (131). In the PROactive study, a large randomized cardiovascular outcome study, pioglitazone decreased TG levels by approximately 10%, increased HDL-C levels by approximately 10%, and increased LDL-C by 1-4% (134). It should be noted that reductions in the small dense LDL subfraction and an increase in the large buoyant LDL subfraction are seen with both thiazolidinediones (131). In a randomized head-to-head trial, it was shown that pioglitazone decreased TG levels and increased serum HDL-C levels to a greater degree than rosiglitazone treatment (135,136). Additionally, pioglitazone increased LDL-C levels less than rosiglitazone. In contrast to the differences in lipid parameters, both rosiglitazone and pioglitazone decreased A1c and C-reactive protein to a similar extent. The mechanism by which pioglitazone induces more favorable changes in lipid levels than rosiglitazone despite similar changes in glucose levels is unclear, but differential actions of ligands for nuclear hormone receptors are well described.

 

Treatment with SGLT2 inhibitors results in a small increase in LDL-C and HDL-C levels (131). In a meta-analysis of 48 randomized controlled trials SGLT2 inhibitors significantly increased LDL-C (3.8mg/dL, p < 0.00001), HDL-C (2.3mg/dL, p < 0.00001), and decreased TG levels (8.8mg/dL, p < 0.00001) (137). The mechanism for these increases in LDL and HDL cholesterol is unknown but could be due to a decrease in plasma volume. The decrease in TG levels could be secondary to weight loss.

 

Bromocriptine-QR (Cycloset) treatment decreases TG levels but has no significant effect on LDL-C or HDL-C levels (138,139). The decrease in TG levels is thought to be due to a decrease in hepatic TG synthesis, likely due to a decrease in adipose tissue lipolysis resulting in decreased blood free fatty acid levels and reduced delivery of fatty acids to the liver for TG synthesis (140).

 

Colesevelam, a bile acid sequestrant that is approved for glucose lowering, lowers LDL-C levels by 15-20% and has only a modest effect on HDL-C levels (101,141). The effect of bile acid sequestrants on TG levels varies (141). In patients with normal TG levels, bile acid sequestrants increase TG levels by a small amount. However, as baseline TG levels increase, the effect of bile acid sequestrants on TG levels becomes greater, and can result in substantial increases in TG levels (141). In patients with TG > 500mg/dL the use of bile acid sequestrants is contraindicated (141).

 

Finally, GLP-1 receptor agonists can favorably affect the lipid profile by inducing weight loss (decreasing TG and very modestly decreasing LDL-C levels) (131). In a review by Nauck and colleagues it was noted that GLP-1 receptor agonists lowered TG levels by 18 to 62mg/dL depending upon the specific GLP-1 receptor agonist while decreasing LDL-C by 3-8mg/dL and increasing HDL-C by less than 1mg/dL (142). Additionally, GLP-1 receptor agonists reduce postprandial TG by reducing circulating chylomicrons by decreasing intestinal lipoprotein production (131,142). DPP4 inhibitors have a similar effect on postprandial TG levels as GLP-1 receptor agonists while having minimal effects on fasting lipid levels (142).

 

In the SURPASS trials, tirzepatide studies TG levels were consistently decreased by 13-25% (83,143). In most studies with the exception of SURPASS 5, HDL cholesterol levels increased by 3-11% (83,143). Total cholesterol and LDL cholesterol levels were modestly decreased in most studies (83,143). Not unexpectedly given the decrease in TG levels small LDL particles were decreased. For details see the Endotext chapter Oral and Injectable (Non-Insulin) Pharmacological Agents for the Treatment of Type 2 Diabetes (83).

 

Table 2. Effect of Glucose Lowering Drugs on Lipid Levels

Metformin

Modestly decrease TG and LDL-C

Sulfonylureas

No effect

DPP4 inhibitors

Decrease postprandial TG

GLP1 analogues

Decrease fasting and postprandial TG, modestly decrease LDL-C

Tirzepatide

Decrease TG, modestly decrease LDL-C, increase HDL-C

Acarbose

Decrease postprandial TG

Pioglitazone

Rosiglitazone

Decrease TG and increase HDL-C. Small increase LDL-C but a decrease in small dense LDL

SGLT2 inhibitors

Small increase in LDL-C and HDL-C

Colesevelam

Decrease LDL-C. May increase TG

Bromocriptine-QR

Decrease TG

Insulin

No effect

 

PATHOPHYSIOLOGY OF THE DYSLIPIDEMIA OF DIABETES

 

Figure 1. Pathophysiology of the Dyslipidemia of Diabetes

 

Multiple mechanisms account for the dyslipidemia seen in patients with T2DM, which are affected both by the level of glucose control and by factors such as obesity and inflammation that also contribute to dyslipidemia.

 

Increase in TG

 

There are a number of different abnormalities that contribute to the dyslipidemia seen in patients with T2DM and obesity (figure 1) (119-122,144-146).

 

OVERPRODUCTION OF VLDL BY THE LIVER

 

A key abnormality is the overproduction of VLDL by the liver, which is a major contributor to the elevations in serum TG levels. The rate of secretion of VLDL is highly dependent on TG availability, which is determined by the levels of fatty acids available for the synthesis of TG in the liver. An abundance of TG prevents the intra-hepatic degradation of Apo B-100 allowing for increased VLDL formation and secretion. There are three major sources of fatty acids in the liver all of which may be altered in patients with T2DM. First, the flux of fatty acids from adipose tissue to the liver is increased. An increased mass of adipose tissue, particularly visceral stores, results in increased fatty acid delivery to the liver. Additionally, insulin suppresses the lipolysis of TG to free fatty acids in adipose tissue; thus, in patients with either poorly controlled diabetes due to a decrease in insulin or a decrease in insulin activity due to insulin resistance, the inhibition of TG lipolysis is blunted and there is increased TG breakdown leading to increased fatty acid deliver to the liver. A second source of fatty acids in the liver is de novo fatty acid synthesis. Numerous studies have shown that fatty acid synthesis is increased in the liver in patients with T2DM. This increase may be mediated by the hyperinsulinemia seen in patients with insulin resistance. While the liver is resistant to the effects of insulin on carbohydrate metabolism, the liver remains sensitive to the effects of insulin stimulating lipid synthesis. Specifically, insulin stimulates the activity of SREBP-1c, a transcription factor that increases the expression of the enzymes required for the synthesis of fatty acids. Thus, while the liver is resistant to the effects of insulin on carbohydrate metabolism the liver remains sensitive to the effects of insulin stimulating lipid synthesis. Additionally, in the presence of hyperglycemia, glucose can induce another transcription factor, carbohydrate responsive element binding protein (ChREBP), which also stimulates the transcription of the enzymes required for fatty acid synthesis. The third source of fatty acids is the uptake of TG rich lipoproteins by the liver. Studies have shown an increase in intestinal fatty acid synthesis and the enhanced secretion of chylomicrons in animal models of T2DM. This increase in chylomicrons leads to the increased delivery of fatty acids to the liver. The increase in hepatic fatty acids produced by these three pathways results in an increase in the synthesis of TG in the liver and the protection of Apo B-100 from degradation resulting in the increased formation and secretion of VLDL. Finally, insulin stimulates the post translational degradation of Apo B-100 in the liver and a decrease in insulin activity in patients with T2DM also allows for the enhanced survival of Apo B-100 promoting increased VLDL formation.

 

DECREASED DEGRADATION OF TRIGLYCERIDE RICH LIPOPROTEINS

 

While the overproduction of triglyceride rich lipoproteins by the liver and intestine are important contributors to the elevations in serum TG levels in patients with T2DM, there are also abnormalities in the metabolism of these TG rich lipoproteins. First, there is a modest decrease in lipoprotein lipase activity, the key enzyme that metabolizes TG rich lipoproteins. The expression of lipoprotein lipase is stimulated by insulin and decreased insulin activity in patients with T2DM results in a decrease in lipoprotein lipase, which plays a key role in the hydrolysis of the TG carried in chylomicrons and VLDL. Additionally, patients with T2DM have an increase in Apo C-III levels, a key regulator of TG rich lipoprotein clearance. Glucose stimulates and insulin suppresses Apo C-III expression; thus, diabetes with hyperglycemia and either insulin deficiency or insulin resistance contribute to an increase in Apo C-III. Apo C-III is an inhibitor of lipoprotein lipase activity and thereby reduces the clearance of TG rich lipoproteins. In addition, Apo C-III also inhibits the cellular uptake of lipoproteins. Studies have shown that loss of function mutations in Apo C-III lead to lower serum TG levels and a reduced risk of ASCVD (147,148). Interestingly, inhibition of Apo C-III expression results in a decrease in serum TG levels even in patients deficient in lipoprotein lipase, indicating that the ability of Apo C-III to modulate serum TG levels is not dependent solely on regulating lipoprotein lipase activity (149). Lastly, insulin resistance is associated with an increase in Angptl3, an inhibitor of LPL (150). Thus, in patients with diabetes, a decrease in clearance of TG rich lipoproteins also contributes to the elevation in serum triglyceride levels.  

 

Mechanism for the Increase in Small Dense LDL and Decrease in HDL

 

The elevation in TG rich lipoproteins in turn has effects on other lipoproteins. Specifically, cholesterol ester transfer protein (CETP) mediates the exchange of TG from TG rich VLDL and chylomicrons to LDL and HDL. The increase in TG rich lipoproteins per se leads to an increase in CETP mediated exchange, increasing the TG content of both LDL and HDL. The TG on LDL and HDL is then hydrolyzed by hepatic lipase and lipoprotein lipase leading to the production of small dense LDL and small HDL. Notably hepatic lipase activity is increased in patients with T2DM, which will also facilitate the removal of TG from LDL and HDL resulting in small lipoprotein particles. The affinity of Apo A-I for small HDL particles is reduced, leading to the disassociation of Apo A-I, which in turn leads to the accelerated clearance and breakdown of Apo A-I by the kidneys. Additionally, the production of Apo A-I may be reduced in patients with diabetes. High glucose levels can activate ChREBP and this transcription factor inhibits Apo A-I expression. Furthermore, insulin stimulates Apo A-I expression and a reduction in insulin activity due to insulin resistance or decreased insulin levels may also lead to a decrease in Apo A-I expression. The net result is lower levels of Apo A-I and HDL-C levels in patients with T2DM.

 

Role of Poor Glycemic Control

 

The above-described changes lead to the typical dyslipidemia observed in patients with T2DM (increased TG, decreased HDL-C, and an abundance of small dense LDL and small HDL). In patients with both Type 1 and T2DM, poor glycemic control can further adversely affect lipid and lipoprotein metabolism. As noted above the expression of lipoprotein lipase is stimulated by insulin. If insulin activity is very low the expression of lipoprotein lipase is severely suppressed and the metabolism of TG rich lipoproteins is markedly impaired. This leads to the delayed clearance of both chylomicrons and VLDL and elevations of TG rich lipoproteins. Additionally, insulinopenia results in a marked increase in lipolysis in adipose tissue, leading to the release of free fatty acids into the circulation. This increase in serum fatty acids results in the increased delivery of fatty acids to the liver, enhanced TG synthesis in the liver, and the increased production and secretion of VLDL. Whereas patients with T1DM who are well controlled and not obese or overweight typically have normal serum lipid profiles, if their control deteriorates, they will develop hypertriglyceridemia. In patients with T2DM deterioration of glycemic control will further exacerbate their underlying dyslipidemia resulting in greater increases in TG levels. If the synthesis of new VLDL is increased sufficiently this can result in an increase in LDL-C levels. HDL-C levels may decrease due to the formation of small HDL that are more susceptible to accelerated clearance. Improvements in glycemic control can markedly lower TG levels and may increase serum HDL-C levels. In patients with poorly controlled diabetes improvements in glycemic control may also lower LDL-C levels.

 

Role of Obesity and Inflammation

 

Most patients with T2DM and many patients with T1D are obese or overweight. Obesity is a pro-inflammatory state due to the macrophages that infiltrate adipose tissue. The cytokines produced by these macrophages and the adipokines that are produced by fat cells also alter lipid metabolism (151,152). The pro-inflammatory cytokines, TNF and IL-1, decrease the expression of lipoprotein lipase and increase the expression of angiopoietin like protein 4, an inhibitor of lipoprotein lipase. Together these changes decrease lipoprotein lipase activity, thereby delaying the clearance of TG rich lipoproteins. In addition, pro-inflammatory cytokines stimulate lipolysis in adipocytes increasing circulating free fatty acid levels, which will provide substrate for hepatic TG synthesis. In the liver, pro-inflammatory cytokines stimulate de novo fatty acid and TG synthesis. These alterations will lead to the increased production and secretion of VLDL. Thus, increases in the levels of pro-inflammatory cytokines will stimulate the production of TG rich lipoproteins and delay the clearance of TG rich lipoproteins, which together will contribute to the increase in serum TG that occurs in obese patients.

 

Obesity and the increase in pro-inflammatory cytokines may also affect HDL-C levels (153-155). First, pro-inflammatory cytokines inhibit the production of Apo A-I, the main protein constituent of HDL. Second, in many tissues pro-inflammatory cytokines decrease the expression of ABCA1 and ABCG1, which will lead to a decrease in the efflux of phospholipids and cholesterol from the cell to HDL decreasing the formation of mature HDL. Third, pro-inflammatory cytokines inhibit the production and activity of LCAT, which will limit the conversion of cholesterol to cholesterol esters in HDL. This conversion step is required for the formation of a normal spherical HDL particle and is crucial for the ability of HDL to increase the efflux of cholesterol from cells (including macrophages). Together these effects may lead to a decrease in HDL-C levels and a decrease in reverse cholesterol transport. Reverse cholesterol transport plays an important role in preventing cholesterol accumulation in macrophages and thereby reduces atherosclerosis.

 

Inflammation also decreases other important functions of HDL, such as its ability to prevent LDL oxidation (156). This reduction in the ability of HDL to protect from oxidation may be mediated in part by inflammation inducing lower levels of the enzyme paraoxonase, which is commonly seen in patients with diabetes (151,157). In parallel inflammation increases the oxidation of LDL and the amount of small dense LDL that is more susceptible to oxidation.

 

Role of Adipokines

 

Adipokines, such as leptin, adiponectin, and resistin, regulate lipid metabolism and the levels are altered in obese patients. Obesity increases serum leptin levels and leptin stimulates lipolysis in adipocytes which will increase serum free fatty acid levels (158). The circulating levels of adiponectin are decreased in subjects who are obese (159). Decreased adiponectin levels are associated with elevations in serum TG levels and decreases in HDL-C levels (159). This association is thought to be causal as studies in mice have shown that overexpressing adiponectin (transgenic mice) decreases TG and increases HDL-C levels while conversely, adiponectin knock-out mice have increased TG and decreased HDL-C levels (159). The adiponectin induced decrease in TG levels is mediated by an increased catabolism of TG rich lipoproteins due to an increase in lipoprotein lipase activity and a decrease Apo C-III, an inhibitor of lipoprotein lipase (159). The increase in HDL-C levels induced by adiponectin is mediated by an increase in hepatic Apo A-I and ABCA1, which results in the increased production of HDL particles (159).

 

Resistin is increased in subjects who are obese and the levels of resistin directly correlate with plasma TG levels (160). Moreover, resistin has been shown to stimulate hepatic VLDL production and secretion due to an increase in the synthesis of Apo B, TG, and cholesterol (160,161). Finally, resistin is associated with a decrease in HDL-C and Apo A-I levels (160).

 

EFFECT OF LIPID LOWERING ON ASCVD EVENTS IN PATIENTS WITH DIABETES

 

Monotherapy Studies

 

STATINS

 

The Cholesterol Treatment Trialists analyzed data from 18,686 subjects with diabetes (mostly T2DM) from 14 randomized trials (162). In the statin treated group there was a 9% decrease in all-cause mortality, a 13% decrease in vascular mortality, and a 21% decrease in major vascular events per 39mg/dL (1mmol/L) reduction in LDL-C. The beneficial effect of statin therapy was seen in both primary and secondary prevention patients. The effect of statin treatment on cardiovascular events in patients with diabetes was similar to that seen in non-diabetic subjects. Thus, these studies indicate that statins are beneficial in reducing ASCVD in patients with diabetes. Because of the large number of patients with diabetes included in the Heart Protection Study (HPS) and CARDS these two studies will be discussed in greater depth.

 

The HPS was a double-blind randomized trial that focused on patients at high risk for the development of cardiovascular events, including patients with a history of MIs, other atherosclerotic lesions, diabetes, and/or hypertension (163,164). Patients were between 40 and 80 years of age and had to have total serum cholesterol levels greater than 135mg/dL (thus very few patients were excluded because they did not have a high enough cholesterol level). The major strength of this trial was the large number of patients studied (>20,000). The diabetes subgroup included 5,963 subjects and thus was as large as many other prevention trials. The study was a 2x2 study design comparing simvastatin 40mg a day vs. placebo and anti-oxidant vitamins (vitamin E 600mg, vitamin C 250mg, and beta-carotene 20mg) vs. placebo and lasted approximately 5 years. Analysis of the group randomized to the anti-oxidant vitamins revealed no beneficial or harmful effects. In contrast, simvastatin therapy (40mg per day) reduced cardiovascular events, including MIs and strokes, by approximately 25% in all participants and to a similar degree in the diabetic subjects (total ASCVD reduced 27%, coronary mortality 20%, MI 37%, stroke 24%). Further analysis of the subjects with diabetes revealed that the reduction in cardiovascular events with statin therapy was similar in individuals with diabetes diagnosed for a short duration (<6 years) and for a long duration (>13 years). Similarly, subjects with diabetes in good control (HbA1c <7%) and those not in ideal control (HbA1c >7%) also benefited to a similar degree with statin therapy. Moreover, both T1DM and T2DM patients had a comparable reduction in ASCVD with simvastatin therapy. The decrease in cardiovascular events in patients with T1DM was not statistically significant because of the small number of subjects. Nevertheless, this is the only trial that included patients with T1DM and suggests that patients with T1DM will benefit from statin therapy similar to T2DM. In general, statin therapy reduced ASCVD in all subgroups of subjects with diabetes (females, males, older age, renal disease, hypertension, high TG, low HDL, ASA therapy, etc.) i.e., statin therapy benefits all patients with diabetes (note this study did not include patients with end stage renal disease but other studies have failed to show benefits of statin therapy in patients with diabetes and end stage renal disease (165)).

 

The CARDS trial specifically focused on subjects with diabetes (166). The subjects in this trial were males and females with T2DM between the ages of 40 to 75 years of age who were at high risk of developing ASCVD based on the presence of hypertension, retinopathy, renal disease, or current smoking. Of particular note, the subjects did not have any evidence of clinical atherosclerosis (myocardial disease, stroke, peripheral vascular disease) at entry and hence this study is a primary prevention trial. Inclusion criteria included LDL-C levels less than 160mg/dL and TG levels less than 600mg/dL. It is important to recognize that the average LDL-C in this trial was approximately 118mg/dL, indicating relatively low LDL-C levels. A total of 2,838 T2DM subjects were randomized to either placebo or atorvastatin 10mg a day. Atorvastatin therapy resulted in a 40% decrease in LDL-C levels with over 80% of patients achieving LDL-C levels less than 100mg/dL. Most importantly, atorvastatin therapy resulted in a 37% reduction in cardiovascular events. In addition, strokes were reduced by 48% and coronary revascularization by 31%. As seen in the HPS, subjects with relatively low LDL-C levels (LDL <120mg/dL) benefited to a similar extent as subjects with higher LDL-C levels (>120mg/dL).

 

HPS and CARDS, in combination with the other statin trials, provide conclusive evidence that statin therapy will reduce cardiovascular events in patients with diabetes. Importantly, the benefits of statin therapy are seen in patients with diabetes in both primary and secondary prevention trials. 

 

Effect of Aggressive LDL-C Lowering with Statins

 

Studies have compared reductions of LDL-C to approximately 100mg/dL to more aggressive reductions in LDL-C on atheroma volume. The Reversal Trial studied 502 symptomatic coronary artery disease patients with an average LDL-C of 150mg/dL (167). Approximately 19% of the patients in this trial had diabetes. Patients were randomized to moderate LDL lowering therapy with pravastatin 40mg per day or to aggressive lipid lowering with atorvastatin 80mg per day. As expected, LDL-C levels were considerably lower in the atorvastatin treated group (pravastatin LDL= 110mg/dL vs. atorvastatin LDL= 79mg/dL). Most importantly, when one analyzed the change in atheroma volume determined after 18 months of therapy using intravascular ultrasound, the group treated aggressively with atorvastatin had a much lower progression rate than the group treated with pravastatin. Compared with baseline values, patients treated with atorvastatin had no change in atheroma burden (there was a very slight regression of lesions), whereas patients treated with pravastatin showed progression of lesions. When one compares the extent of the reduction in LDL-C to the change in atheroma volume, a 50% reduction in LDL (LDL-C levels of approximately 75mg/dL) resulted in the absence of lesion progression. This study suggests that lowering the LDL-C to levels well below 100mg/dL is required to prevent disease progression as measured by intravascular ultrasound. Other studies, such as Asteroid, have shown that marked reductions in LDL-C (in Asteroid the mean LDL-C levels were 61mg/dL) can also result in the regression of coronary artery atherosclerosis determined by intravascular ultrasound measurements (168). Additionally, the Saturn trial demonstrated that aggressive lipid lowering with either atorvastatin 80mg or rosuvastatin 40mg would induce regression of coronary artery atherosclerosis to a similar degree in patients with and without diabetes if the LDL-C levels were reduced to less than 70mg/dL (169). Together these trials indicate that aggressive lowering of LDL-C levels to below 70mg/dL can induce regression of atherosclerotic lesions.

 

The Prove-It trial determined in patients recently hospitalized for an acute coronary syndrome whether aggressively lowering of LDL-C with atorvastatin 80mg per day vs. moderate LDL-C lowering with pravastatin 40mg per day would have a similar effect on cardiovascular end points such as death, MI, documented unstable angina requiring hospitalization, revascularization, or stroke (170,171). In this trial, approximately 18% of the patients were diabetic. As expected, the on-treatment LDL-C levels were significantly lower in patients aggressively treated with atorvastatin compared to the moderate treated pravastatin group (atorvastatin LDL-C = approximately 62 vs. pravastatin LDL-C = approximately 95mg/dL). Of great significance, death or major cardiovascular events was reduced by 16% over the two years of the study in the group aggressively treated with atorvastatin. Moreover, the risk reduction in the patients with diabetes in the aggressive treatment group was similar to that observed in non-diabetics.

 

In the treating to new targets trial (TNT) patients with stable coronary heart disease and LDL-C levels less than 130mg/dL were randomized to either 10mg or 80mg atorvastatin and followed for an average of 4.9 years (172,173). Approximately 15% of the patients had diabetes. As expected, LDL-C levels were lowered to a greater extent in the patients treated with 80mg atorvastatin than with 10mg atorvastatin (77mg/dL vs. 101mg/dL). Impressively, the occurrence of major cardiovascular events was reduced by 22% in the group treated with atorvastatin 80mg (p<0.001). In the patients with diabetes events were reduced by 25% in the high dose statin group.

 

Finally, the IDEAL trial was a randomized study that compared atorvastatin 80mg vs. simvastatin 20-40mg in 8,888 patients with a history of ASCVD (174). Approximately 12% of the patients had diabetes. As expected, LDL-C levels were reduced to a greater extent in the atorvastatin treated group than the simvastatin treated group (approximately 81mg/dL vs. 104mg/dL). Once again, the greater reduction in LDL-C levels was associated with a greater reduction in cardiovascular events. Specifically, major coronary events defined as coronary death, nonfatal MI, or cardiac arrest was reduced by 11% (p=0.07), while nonfatal acute MI were reduced by 17% (p=0.02).

 

Combining the results of the Heart Protection Study, CARDS, Reversal, Saturn, Asteroid, Prove-It, TNT, and IDEAL leads one to the conclusion that aggressive lowering of LDL-C with statin therapy will be beneficial and suggests that in high-risk patients lowering the LDL to levels well below 100mg/dL is desirable. Moreover, the Cholesterol Treatment Trialists reviewed five trials with 39,612 subjects that were designed to determine the effect of usual vs. aggressive reductions in LDL-C (175). They reported that intensive control (approximately a 19mg/dL difference in LDL-C) resulted in a 15% decrease in major vascular events, a 13% reduction in coronary death or non-fatal MI, a 19% decrease in coronary revascularization, and a 16% decrease in strokes. As will be discussed below treatment guidelines reflect the results of these studies. Additionally, as described in detail below, studies of the addition of either ezetimibe or PCSK9 inhibitors to statins further demonstrates that aggressive lowering of LDL-C levels further reduces cardiovascular events

 

FIBRATES

 

The beneficial effect of monotherapy with fibrates (e.g., gemfibrozil, fenofibrate) on ASCVD in patients with diabetes is shown in Table 3. The results of these randomized trials suggest that monotherapy with this class of drug might reduce cardiovascular events in patients with diabetes, but the data is not very robust. The largest trial was the Field Trial (176). In this trial, 9,795 patients with T2DM between the ages of 50 and 75 not taking statin therapy were randomized to fenofibrate or placebo and followed for approximately 5 years. Fenofibrate therapy resulted in a 12% decrease in LDL-C, a 29% decrease in TG, and a 5% increase in HDL-C levels. The primary outcome was coronary events (coronary heart disease death and non-fatal MI), which were reduced by 11% in the fenofibrate group but did not reach statistical significance (p= 0.16). However, there was a 24% decrease in non-fatal MI in the fenofibrate treated group (p=0.01) and a non-significant increase in coronary heart disease mortality. Total ASCVD events (coronary events plus stroke and coronary or carotid revascularization) were reduced 11% (p=0.035). These beneficial effects of fenofibrate therapy on ASCVD were observed in patients without a previous history of ASCVD. In patients with a previous history of ASCVD no benefits were observed. Additionally, the beneficial effect of fenofibrate therapy was seen only in those subjects less than 65 years of age. The beneficial effects of fenofibrate in this study may have been muted by the increased use of statins in the placebo group, which reduced the differences in lipid levels between the placebo and fenofibrate groups. If one adjusted for the addition of lipid-lowering therapy, fenofibrate reduced the risk of coronary heart disease events by 19% (p=0.01) and of total ASCVD events by 15% (p=0.004). Thus, while the results of this large trial are intriguing they do not clearly show a benefit of fibrate therapy reducing ASCVD events. The number of patients with diabetes in the other fibrate trials are relatively small (table 3).

 

While the results of the monotherapy fibrate trials have been very heterogeneous it should be noted that fibrate trials in patients with elevated TG levels have reported a greater reduction of cardiovascular events (177). Additionally, subgroup analysis of several fibrate trials has also suggested that the benefit of fibrates was greatest in patients with elevated TG levels (177,178).

 

The mechanism by which fibrates may reduce cardiovascular events is unclear. These drugs lower serum TG levels and increase HDL-C, but it should be recognized that the beneficial effects of fibrates could be due to other actions of these drugs. Specifically, these drugs activate the nuclear hormone receptor PPAR alpha, which is present in the cells that comprise the atherosclerotic lesions, and it is possible that these compounds directly affect lesion formation and development. In addition, fibrates are anti-inflammatory. In fact, analysis of the VA-HIT study suggested that much of the benefit of fibrate therapy was not due to changes in serum lipoprotein levels (179,180).

 

To summarize, while in general the studies suggest that monotherapy with fibrates may reduce ASCVD in patients with diabetes, the results are not very robust or consistent as seen in the statin trials. Of note fibrate therapy appeared to be most effective in patients with increased TG levels and decreased HDL levels, a lipid profile typically seen in patients with T2DM. However, as will be presented in detail below (combination therapy section) the addition of fibrates to statins does not reduce ASCVD.

 

Table 3. Effect of Fibrate Monotherapy on Cardiovascular Outcomes

Study

Drug

#Diabetic subjects

%Decrease controls

% Decrease diabetics

Helsinki Heart Study (181)

Gemfibrozil

135

34

60*

VA-HIT (180)

Gemfibrozil

620

24

24

DIAS (182)

Fenofibrate

418

-

23*

Sendcap (183)

Bezafibrate

164

-

70

Field (176)

Fenofibrate

9795

-

11*

* Not statistically significant

 

NIACIN

 

A single randomized trial, the Coronary Drug Project, has examined the effect of niacin monotherapy on cardiovascular outcomes (184). This trial was carried out from 1966 to 1974 (before the introduction of statin therapy) in men with a history of a prior MI and demonstrated that niacin therapy reduced cardiovascular events. The results of this study were re-analyzed to determine the effect of niacin therapy in subjects with varying baseline fasting and 1-hour post meal glucose levels (185). It was noted that 6 years of niacin therapy reduced the risk of coronary heart disease death or nonfatal MI by approximately 15-25% regardless of baseline fasting or 1-hour post glucose challenge glucose levels. Particularly notable is that reductions in events were seen in the subjects who had a fasting glucose level >126mg/dL or 1-hour glucose levels >220mg/dL (i.e., patients with diabetes). Thus, based on this single study, niacin monotherapy reduces cardiovascular events both in normal subjects and patients with diabetes. However, as will be presented in detail below (combination therapy section) the addition of niacin to statins does not reduce ASCVD.

 

EZETIMIBE

 

A multicenter, randomized trial in Japan examined the efficacy of ezetimibe in patients aged ≥75 years with elevated LDL-C (≥140 mg/dL) without a history of coronary artery disease who were not taking lipid lowering drugs (186). Patients were randomized to ezetimibe (n=1716) or usual care (n=1695) and followed for 4.1 years. The primary outcome was a composite of sudden cardiac death, MI, coronary revascularization, or stroke. In the ezetimibe group LDL-C was decreased by 25.9% and non-HDL-C by 23.1% while in the usual care group LDL-C was decreased by 18.5% and non-HDL-C by 16.5% (p<0.001 for both lipid parameters). By the end of the trial 9.6% of the patients in the usual care group and 2.1% of the ezetimibe group were taking statins. Ezetimibe reduced the incidence of the primary outcome by 34% (HR 0.66; P=0.002). Additionally, composite cardiac events were reduced by 60% (HR 0.60; P=0.039) and coronary revascularization by 62% (HR 0.38; P=0.007) in the ezetimibe group vs. the control group. There was no difference in the incidence of stroke or all-cause mortality between the groups. Approximately 25% of the patients in this trial had diabetes and the beneficial effects were similar in the diabetic and non-diabetic subjects. It should be noted that the reduction in cardiovascular events was much greater than one would expect based on the absolute difference in LDL-C levels (121mg/dL in ezetimibe group vs. 132mg/dL). As stated by the authors “Given the open-label nature of the trial, its premature termination, and issues with follow-up, the magnitude of benefit observed should be interpreted with caution.” Nevertheless, this study provides suggestive evidence that ezetimibe monotherapy may reduce cardiovascular events in patients with diabetes.

 

BEMPEDOIC ACID

 

A multicenter study of bempedoic acid in statin intolerant patients with ASCVD or at high risk for ASCVD was recently reported (187). Patients were randomized to bempedoic acid, 180 mg daily (n=6992), or placebo (n=6978) and the primary end point was death from cardiovascular causes, nonfatal MI, nonfatal stroke, or coronary revascularization. Bempedoic acid therapy reduced LDL-C and hsCRP levels by approximately 22% compared to the placebo group. The primary composite endpoint was reduced by 13% in the bempedoic acid group (HR 0.87; 95% CI, 0.79 to 0.96; P = 0.004). The four individual components of the primary endpoint were also significantly reduced in the bempedoic acid treatment group. In this trial approximately 45% of the patients had diabetes. In an analysis of the patients without clinical ASCVD, (i.e., primary prevention) (bempedoic acid n = 2100 or placebo n = 2106), there was a 30% decrease in cardiovascular events (HR 0.70: 95% CI, 0.55-0.89; P = .002) (188). In this subgroup analysis 66% of the patients had diabetes. This study clearly indicates that monotherapy with bempedoic acid will reduce cardiovascular events.

 

OTHER DRUGS

 

With regard to PCSK9 inhibitors and bile acid sequestrants there have been no randomized monotherapy studies that have examined the effect of these drugs on cardiovascular end points in subjects with diabetes. In non-diabetic subjects, monotherapy with bile acid sequestrants have reduced cardiovascular events (102,103). Since bile acid sequestrants have a similar beneficial impact on serum lipid levels in diabetic and non-diabetic subjects one would anticipate that these drugs would also result in a reduction in events in the diabetic population. Additionally, bile acid sequestrants improve glycemic control (101). However, bile acid sequestrants can raise TG levels and therefore must be used with caution in hypertriglyceridemic patients. There are no outcome studies with PCSK9 inhibitor monotherapy in patients with diabetes but given that these drugs reduce LDL-C levels and in combination with statins reduce cardiovascular events one would anticipate that PCSK9 inhibitor monotherapy will also reduce cardiovascular events.

 

Combination Therapy

 

The studies with statins have been so impressive that most patients with diabetes over the age of 40 are routinely treated with statin therapy and younger patients with diabetes at high risk for ASCVD are also typically on statin therapy (see Current Guidelines Section). Therefore, a key issue is whether the addition of other lipid lowering drugs to statins will result in a further reduction in cardiovascular events. A difficulty with such studies is that the reduction in cardiovascular events induced by statin therapy is so robust that very large trials may be required to see additional benefit.

 

STATINS + FIBRATES

 

The ACCORD-LIPID trial was designed to determine if the addition of fenofibrate to aggressive statin therapy would result in a further reduction in ASCVD in patients with T2DM (189). In this trial, 5,518 patients on statin therapy were randomized to placebo or fenofibrate therapy. The patients had diabetes for approximately 10 years and either had pre-existing ASCVD or were at high risk for developing ASCVD. During the trial, LDL-C levels were approximately 80mg/dL in both groups. There was only a small difference in HDL-C with the fenofibrate groups having a mean HDL-C of 41.2mg/dL while the control group had an HDL-C of 40.5mg/dL. Differences in TG levels were somewhat more impressive with the fenofibrate group having a mean TG level of 122mg/dL while the control group had a TG level of 144mg/dL. First occurrence of nonfatal MI, nonfatal stroke, or death from cardiovascular causes was the primary outcome and there was no statistical difference between the fenofibrate treated group and the placebo group. Additionally, there were also no statistically significant differences between the groups with regards to any of the secondary outcome measures of ASCVD. Of note, the addition of fenofibrate to statin therapy did not result in an increase in either muscle or liver side effects. On further analysis, there was a possible benefit of fenofibrate therapy in the patients in whom the baseline TG levels were elevated (>204mg/dL) and HDL-C levels decreased (<34mg/dL). Finally, similar to what has been reported in other trials, fenofibrate had beneficial effects on the progression of microvascular disease (190,191).

 

The PROMINENT trial studied the effect of pemafibrate, a new selective PPAR-alpha activator, in reducing cardiovascular outcomes in 10,497 patients (66.9% with previous ASCVD) with diabetes (192). This was a double-blind, randomized, controlled trial, in patients with T2DM, with mild-to-moderate hypertriglyceridemia (TG level, 200 to 499 mg/dL), LDL-C < 100mg/dL, and HDL-C levels < 40 mg/dL) who received either pemafibrate (0.2-mg tablets twice daily) or placebo in addition to statin therapy (96% on statins). The primary end point was a composite of nonfatal MI, ischemic stroke, coronary revascularization, or death from cardiovascular causes. Baseline fasting TG was 271 mg/dL, HDL-C 33 mg/dL, and LDL-C 78 mg/dL. Compared with placebo, pemafibrate decreased TG by 26.2%, while HDL-C increased 5.1% and LDL-C increased 12.3%. Notably non-HDL-C levels were unchanged and Apo B levels increased 4.8%. The primary endpoint was similar in the pemafibrate and placebo group (HR 1.03; 95% CI 0.91 to 1.15). The increase in LDL-C and Apo B levels likely account for the failure to reduce cardiovascular events.

 

Taken together the ACCORD study and the PROINENT trial indicate that the addition of fibrate therapy to statin therapy will not result in a reduction in cardiovascular events in patients with diabetes.

 

STATIN + NIACIN

 

The AIM-HIGH trial was designed to determine if the addition of Niaspan to aggressive statin therapy would result in a further reduction in cardiovascular events in patients with pre-existing ASCVD (193). In this trial 3,314 patients were randomized to Niaspan vs. placebo. Approximately 33% of the patients had diabetes. On trial, LDL-C levels were in the 60-70mg/dL range in both groups. As expected, HDL-C levels were increased in the Niaspan treated group (approximately 44mg/dL vs. 38mg/dL), while TG were decreased (approximately 121mg/dL vs. 155mg/dL). However, there were no differences in the primary endpoint between the control and Niaspan treated groups (Primary endpoint consisted of the first event of death from coronary heart disease, nonfatal MI, ischemic stroke, hospitalization for an acute coronary syndrome, or symptom-driven coronary or cerebral revascularization). There were also no differences in secondary endpoints except for a possible increase in strokes in the Niaspan treated group. The addition of Niaspan to statin therapy did not result in a significant increase in either muscle or liver toxicity. Thus, this study does not provide support for the addition of niacin to statins. However, it should be recognized that this was a relatively small study and a considerable number of patients stopped taking the Niaspan during the course of the study (25.4% of patients discontinued Niaspan therapy). In addition, most of the patients included in this study did not have a lipid profile that one would typically consider treating with niacin therapy. In the subset of patients with TG > 198mg/dL and HDL-C < 33mg/dL niacin showed a trend towards benefit (hazard ratio 0.74; p=0.073), suggesting that if the appropriate patient population was studied the results may have been positive (194).

 

HPS 2 Thrive also studied the effect of niacin added to statin therapy (195). This trial utilized extended-release niacin combined with laropiprant, a prostaglandin D2 receptor antagonist that reduces the flushing side effect of niacin treatment. HPS 2 Thrive was a very large trial with over 25,000 patients randomized to either niacin therapy or placebo. Approximately 32% of the patients in this trial had diabetes. The LDL-C level was 63mg/dL, the HDL-C 44mg/dL, and the TG 125mg/dL at baseline. As expected, niacin therapy resulted in a modest reduction in LDL-C (10mg/dL), a modest increase in HDL-C (6mg/dL), and a larger reduction in TG (33mg/dL). However, despite these lipid changes there were no significant differences in major cardiovascular events between the niacin and control group (risk ratio 0.96 CI 0.90- 1.03). It is unknown whether laropiprant, the prostaglandin D2 receptor antagonist, might have effects that worsen atherosclerosis and increase event rates. Similar to the AIM-HIGH study, the group of patients included in the HPS 2 Thrive trial were not the ideal patient population to test for the beneficial effects of niacin treatment added to statin therapy. Ideally, patients with high TG and high non-HDL-C levels coupled with low HDL-C levels should be studied. Nevertheless, the results of the AIM-HIGH and HPS 2 Thrive trials do not provide support for the addition of niacin to statin therapy in patients with diabetes.

 

STATIN + EZETIMIBE

 

The IMPROVE-IT trial tested whether the addition of ezetimibe to statin therapy would provide an additional beneficial effect in patients with the acute coronary syndrome (196). This was a large trial with over 18,000 patients randomized to statin therapy vs. statin therapy + ezetimibe. Approximately 27% of the patients in this trial had diabetes. On treatment LDL-C levels were 70mg/dL in the statin alone group vs. 53mg/dL in the statin + ezetimibe group. There was a small but significant 6.4% decrease in major cardiovascular events (Cardiovascular death, MI, documented unstable angina requiring re-hospitalization, coronary revascularization, or stroke) in the statin + ezetimibe group (HR 0.936 CI (0.887, 0.988) p=0.016). Cardiovascular death, non-fatal MI, or non-fatal stroke were reduced by 10% (HR 0.90 CI (0.84, 0.97) p=0.003). These beneficial effects were particularly pronounced in the patients with diabetes (Primary endpoint hazard ratio, 0.85; 95% confidence interval, 0.78-0.94) (197,198). This trial provides evidence that the combination of a statin + ezetimibe that results in a greater reduction in LDL-C levels will lead to a larger decrease in cardiovascular events than statin alone. It should be noted that the observed reduction in events was in the range expected based on the decrease in LDL-C levels.

 

The RACING trial compared rosuvastatin 10 mg plus ezetimibe 10 mg (combination therapy) vs. rosuvastatin 20mg in 3,780 patients (1,398 patients (37.0%) with diabetes) at 26 centers in South Korea (199). In the patients with diabetes the baseline LDL-C levels was 74mg/dL and during the study the median LDL-C was 53mg/dL in the combination therapy group and 61mg/dL in the high-intensity statin group (P < 0.001). After a median follow-up of 3 years the rate of cardiovascular events in patients with diabetes was 10.0% in the combination therapy group and 11.3% in the high-intensity statin group (HR: 0.89; 95% CI: 0.64–1.22; P = 0.460). Interestingly the rate of discontinuation or dose reduction of the study drug due to intolerance was lower in the combination therapy group than in the high-intensity statin group (5.2 vs. 8.7%; P = 0.014). This study demonstrates that cardiovascular outcomes were comparable between those receiving combination therapy vs. high-intensity statin monotherapy and that combination therapy significantly reduced the rate of drug discontinuation or dose reduction due to intolerance.

 

STATIN + PCSK9 INHIBITORS

 

The FOURIER trial was a randomized, double-blind, placebo-controlled trial of evolocumab vs. placebo in 27,564 patients with atherosclerotic ASCVD and an LDL-C level of 70 mg/dL or higher who were on statin therapy (200). Approximately 40% of the patients had diabetes (201). The primary end point was cardiovascular death, MI, stroke, hospitalization for unstable angina, or coronary revascularization and the key secondary end point was cardiovascular death, MI, or stroke. The median duration of follow-up was 2.2 years. Baseline LDL-C levels were 92mg/dL and evolocumab resulted in a 59% decrease in LDL-C levels (LDL-C level on treatment approximately 30mg/dL). Evolocumab treatment significantly reduced the risk of the primary end point (HR 0.85; 95% CI 0.79 to 0.92; P<0.001) and the key secondary end point (HR 0.80; 95% CI 0.73 to 0.88; P<0.001). The results were consistent across key subgroups, including the subgroup of patients in the lowest quartile for baseline LDL-C levels (median, 74 mg/dL). Of note, a similar decrease in cardiovascular events occurred in patients with diabetes treated with evolocumab and glycemic control was not altered (202). Further analysis has shown that in the small number of patients with a baseline LDL-C level less than 70mg/dL, evolocumab reduced cardiovascular events to a similar degree as in the patients with an LDL-C greater than 70mg/dL (203). Finally, the lower the on-treatment LDL-C levels (down to levels below 20mg/dL), the lower the cardiovascular event rate, suggesting that greater reductions in LDL-C levels will result in greater reductions in ASCVD (204).

 

The ODYSSEY trial was a multicenter, randomized, double-blind, placebo-controlled trial involving 18,924 patients who had an acute coronary syndrome 1 to 12 months earlier, an LDL-C level of at least 70 mg/dL, a non-HDL-C level of at least 100 mg/dL, or an Apo B level of at least 80 mg/dL while on high intensity statin therapy or the maximum tolerated statin dose (205). Approximately 29% of the patients had diabetes. Patients were randomly assigned to receive alirocumab 75 mg every 2 weeks or matching placebo. The dose of alirocumab was adjusted to target an LDL-C level of 25 to 50 mg/dL. The primary end point was a composite of death from coronary heart disease, nonfatal MI, fatal or nonfatal ischemic stroke, or unstable angina requiring hospitalization. During the trial LDL-C levels in the placebo group was 93-103mg/dL while in the alirocumab group LDL-C levels were 40mg/dL at 4 months, 48mg/dL at 12 months, and 66mg/dL at 48 months (the increase with time was due to discontinuation of alirocumab or a decrease in dose). The primary endpoint was reduced by 15% in the alirocumab group (HR 0.85; 95% CI 0.78 to 0.93; P<0.001). In addition, total mortality was reduced by 15% in the alirocumab group (HR 0.85; 95% CI 0.73 to 0.98). The absolute benefit of alirocumab was greatest in patients with a baseline LDL-C level > than 100mg/dL. In patients with an LDL-C level > than 100mg/dL the number needed to treat with alirocumab to prevent an event was only 16. It should be noted that similar to the FOURIER trial the duration of this trial was very short (median follow-up 2.8 years) which may have minimized the beneficial effects. Additionally, because alirocumab 75mg every 2 weeks was stopped if the LDL-C level was < 15mg/dL on two consecutive measurements the beneficial effects may have been blunted (7.7% of patients randomized to alirocumab were switched to placebo).

 

It should be noted that that the duration of the PCSK9 outcome trials were relatively short and it is well recognized from previous statin trials that the beneficial effects of lowering LDL-C levels takes time with only modest effects observed during the first year of treatment. In the FOURIER trial the reduction of cardiovascular death, MI, or stroke was 16% during the first year but was 25% beyond 12 months. In the ODYSSEY trial the occurrence of cardiovascular events was similar in the alirocumab and placebo group during the first year of the study with benefits of alirocumab appearing after year one. Thus, the long-term benefits of treatment with a PCSK9 inhibitor may be greater than that observed during these relatively short-term studies.

 

Additional support for the benefits of further lowering of LDL-C levels with a PCSK9 inhibitor added to statin therapy is seen in the GLAGOV trial (206). This trial was a double-blind, placebo-controlled, randomized trial of evolocumab vs. placebo in 968 patients presenting for coronary angiography. Approximately 20-21% of the patients had diabetes. The primary efficacy measure was the change in percent atheroma volume (PAV) from baseline to week 78, measured by serial intravascular ultrasonography (IVUS) imaging. Secondary efficacy measures included change in normalized total atheroma volume (TAV) and percentage of patients demonstrating plaque regression. As expected, there was a marked decrease in LDL-C levels in the evolocumab group (Placebo 93mg/dL vs. evolocumab 37mg/dL; p<0.001). PAV increased 0.05% with placebo and decreased 0.95% with evolocumab (P < .001) while TAV decreased 0.9 mm3 with placebo and 5.8 mm3 with evolocumab (P < .001). There was a linear relationship between achieved LDL-C and change in PAV (i.e., the lower the LDL-C the greater the regression in atheroma volume down to an LDL-C of 20mg/dL). Additionally, evolocumab induced plaque regression in a greater percentage of patients than placebo (64.3% vs 47.3%; P < .001 for PAV and 61.5% vs 48.9%; P < .001 for TAV). The results in the patients with diabetes were similar to the non-diabetic patients.

 

Taken together these trials demonstrate that further lowering LDL-C levels with PCSK9 monoclonal antibodies in patients taking statins will have beneficial effects on ASCVD outcomes. A study of the effect of inclisiran on ASCVD endpoints is currently in progress.

 

The results of the ezetimibe and PCSK9 trials have several important implications. First, it demonstrates that combination therapy may have benefits above and beyond statin therapy alone. Second, it provides further support for the hypothesis that lowering LDL per se will reduce cardiovascular events. Third, it suggests that lowering LDL levels to much lower levels than usual will have significant benefits. These results have implications for determining goals of therapy.

 

STATINS + LOW DOSE OMEGA-3-FATTY ACIDS

 

ORIGIN was a double-blind study in 12,536 patients at high risk for ASCVD who had impaired fasting glucose, impaired glucose tolerance, or diabetes (207).  Patients were randomized to receive a 1-gram capsule containing at least 900mg of ethyl esters of omega-3 fatty acids (EPA 465mg and DHA 375mg) or placebo for approximately 6 years. Greater than 50% of the patients were on statin therapy. The primary outcome was death from cardiovascular causes. TG levels were reduced by 14.5mg/dL in the group receiving omega-3-fatty acids compared to the placebo group (P<0.001), without a significant effect on other lipids. The incidence of the primary outcome was not significantly decreased among patients receiving omega-3-fatty acids as compared with those receiving placebo. The use of omega-3-fatty acids also had no significant effect on the rates of major vascular events, death from any cause, or death from arrhythmia.

 

A Study of Cardiovascular Events in Diabetes (ASCEND) was a randomized, placebo controlled, double blind trial of 1-gram omega-3-fattys acids (400mg EPA and 300mg DHA ethyl esters) vs. olive oil placebo in 15,480 patients with diabetes without a history of ASCVD (primary prevention trial) (208). Approximately 75% of patients were on statin therapy. The primary end point was serious vascular events (non-fatal MI, non-fatal stroke, transient ischemic attack, or vascular death). Total cholesterol, HDL-C, and non-HDL-C levels were not significantly altered by omega-3-fatty acid treatment (changes in TG levels were not reported). After a mean follow-up of 7.4 years the composite outcome of a serious vascular event or revascularization occurred in 882 patients (11.4%) on omega-3-fatty acids and 887 patients (11.5%) on placebo (rate ratio, 1.00; 95% CI, 0.91 to 1.09). Serious adverse events were similar in placebo and omega-3-fatty acid treated groups.

 

Taken together these studies indicate that low dose omega-3-fatty acids do not reduce cardiovascular events in patients with diabetes. Studies in non-diabetics have also found little effect of low dose omega-3-fatty acids on ASCVD (209).

 

STATINS + HIGH DOSE OMEGA-3-FATTY ACIDS

 

The Japan EPA Lipid Intervention Study (JELIS) was an open label non-placebo controlled study in patients on statin therapy with total cholesterol levels > 254mg/dL with (n= 3664) or without ASCVD (n=14,981) who were randomly assigned to be treated with 1800 mg of EPA (Vascepa) + statin (n=9326) or statin alone (n= 9319) with a 5 year follow-up (210). Approximately 16% of the patients had diabetes. The mean baseline TG level was 153mg/dL. The primary endpoint was any major coronary event, including sudden cardiac death, fatal and non-fatal MI, and other non-fatal events including unstable angina pectoris, angioplasty, stenting, or coronary artery bypass grafting. On treatment total cholesterol, LDL-C, and HDL-C levels were similar in the two groups but plasma TG were modestly decreased in the EPA treated group (5% decrease in EPA group compared to controls; p = 0.0001). In the EPA + statin group the primary endpoint occurred in 2.8% of the patients vs. 3.5% of the patients in the statin alone group (19% decrease; p = 0.011). Unstable angina and non-fatal coronary events were also significantly reduced in the EPA group but in this study sudden cardiac death and coronary death did not differ between groups. Unstable angina was the main component contributing to the primary endpoint and this is a more subjective endpoint than other endpoints such as a MI, stroke, or cardiovascular death. A subjective endpoint has the potential to be an unreliable endpoint in an open label study and is a major limitation of the JELIS Study. The reduction in events was similar in the subgroup of patients with diabetes. In patients with TG levels >150mg/dL and HDL-C levels < 40mg/dL there was a 53% decrease in events (211). In the EPA group, small increases in the occurrence of bleeding (1.1% vs. 0.6%, p=0.0006), gastrointestinal disturbance (3.8%% vs. 1.7%, p<0.0001) and skin abnormalities (1.7 vs. 0.7%, p<0.0001) were seen. 

 

The Reduction of Cardiovascular Events with EPA – Intervention Trial (REDUCE-IT) was a randomized, double blind trial of 2 grams twice per day of EPA ethyl ester (icosapent ethyl) (Vascepa) vs. placebo (mineral oil) in 8,179 patients with hypertriglyceridemia (135mg/dL to 499mg/dL) and established ASCVD or high ASCVD risk (diabetes plus one risk factor) who were on stable statin therapy (212). Approximately 60% of the patients in this trial had diabetes. The primary end point was a composite of cardiovascular death, nonfatal MI, nonfatal stroke, coronary revascularization, or unstable angina. At baseline, the median LDL-C level was 75.0 mg/dL, HDL-C level was 40.0 mg/dL, and TG level was 216.0 mg/dL. The median change in TG level from baseline to 1 year was a decrease of 18.3% (−39.0 mg/dL) in the EPA group and an increase of 2.2% (4.5 mg/dL) in the placebo group. After a median of 4.9 years the primary end-point occurred in 17.2% of the patients in the EPA group vs. 22.0% of the patients in the placebo group (HR 0.75; 95% CI 0.68 to 0.83; P<0.001), indicating a 25% decrease in events. The beneficial effects were similar in patients with and without diabetes. The number needed to treat to avoid one primary end-point event was 21. The reduction in cardiovascular events was noted after approximately 2 years of EPA treatment. Additionally, the risk of cardiovascular death was decreased by 20% in the EPA group (HR 0.80; 95% CI, 0.66 to 0.98; P=0.03). The cardiovascular benefits of EPA were similar across baseline levels of TG (<150, ≥150 to <200, and ≥200 mg/dL). Moreover, the cardiovascular benefits of EPA appeared to occur irrespective of the attained TG level at 1 year (≥150 or <150 mg/dL), suggesting that the cardiovascular risk reduction was not associated with attainment of a normal TG levels. An increase in hospitalization for atrial fibrillation or flutter (3.1% vs. 2.1%, P=0.004) occurred in the EPA group. In addition, serious bleeding events occurred in 2.7% of the patients in the EPA group and in 2.1% in the placebo group (P=0.06). There were no fatal bleeding events in either group and the rates of hemorrhagic stroke, serious central nervous system bleeding, and serious gastrointestinal bleeding were not significantly higher in the EPA group.

 

These results demonstrate that EPA treatment reduces ASCVD events. Of note the reduction in TG levels is relatively modest and would not be expected to result in the magnitude of the decrease in ASCVD observed in the JELIS and REDUCE-IT trials. Other actions of EPA, such as decreasing platelet function, anti-inflammation, decreasing lipid oxidation, stabilizing membranes, etc. could account for or contribute to the reduction in cardiovascular events (213). It is likely that the beneficial effects of EPA seen in the JELIS and REDUCE-IT trials are multifactorial.

 

The Statin Residual Risk Reduction with Epanova in High Risk Patients with Hypertriglyceridemia (STRENGTH) trial was a randomized, placebo controlled, double blind trial of 4 grams per day of omega-3-fatty acids (Epanova) (carboxylic acid formulation of EPA and DHA) vs. placebo (corn oil) in 13,000 patients on statins with hypertriglyceridemia (180-500mg/dL), optimal LDL-C levels (< 100mg/dL or on maximal statin therapy), low HDL-C (<42mg/dL in men and < 47mg/dL in women), and either ASCVD or high risk for ASCVD (214). The primary outcome was major atherosclerotic cardiovascular events (cardiovascular death, MI, stroke, coronary revascularization or hospitalization for unstable angina). The primary end point occurred in 785 patients (12.0%) treated with omega-3 CA vs 795 (12.2%) treated with corn oil (HR, 0.99: [95% CI, 0.90-1.09]; P = .84) (215). Thus, in contrast to EPA alone this omega-3-fatty acid formulation failed to show benefits despite reducing TG levels (18% decrease) to a similar degree as in the REDUCE-IT trial.

 

Whether EPA has special properties that resulted in the reduction in cardiovascular events in the REDUCE-IT trial or there were flaws in the trial design (the use of mineral oil as the placebo) is uncertain and debated. It should be noted that in the REDUCE-IT trial LDL-C and non-HDL-C levels were increased by approximately 10% (LDL-C by approximately 9mg/dL and non-HDL-C by approximately 10mg/dL) in the mineral oil placebo group (212). Additionally, Apo B levels were increased by 7% (6mg/dL) by mineral oil (212). Finally, an increase in hsCRP (20-30%) and other biomarkers of atherosclerosis (oxidized LDL-C, IL-6, IL-1 beta, and lipoprotein-associated phospholipase A2) were noted in the mineral oil group (212,216). In the STRENGTH trial there were no differences in LDL-C, Non-HDL-C, HDL-C, Apo B, or hsCRP levels between the treated vs. placebo groups (215). Whether EPA has special properties compared to DHA leading to a reduction in cardiovascular events or the mineral oil placebo resulted in adverse changes increasing ASCVD in the placebo resulting in an artifactual decrease in the EPA group is debated (217,218). Ideally, another large randomized cardiovascular trial with EPA ethyl ester (icosapent ethyl) (Vascepa) using a placebo other than mineral oil would resolve this controversy.

 

CURRENT GUIDELINES FOR SERUM LIPIDS

 

There are several different guidelines for treating lipids in patients with diabetes. While they all focus on lowering LDL-C there are differences between the various guidelines.

 

American Diabetes Association Guidelines

 

The 2023 American Diabetes Association (ADA) recommends that adult patients with diabetes have their lipid profile determined at the time of diabetes diagnosis and at least every 5 years thereafter or more frequently if indicated (219). This profile includes total cholesterol, HDL-C, TG, and calculated LDL-C. A lipid panel should be obtained immediately prior to initiating statin therapy. Once a patient is on statin therapy testing should be carried out 4-12 weeks after initiating therapy and annually thereafter to monitor adherence and efficacy. Lifestyle modifications including a reduction in saturated fat, trans fat, and cholesterol intake, weight loss if indicated, an increase in omega-3-fatty acids, viscous fiber, and plant stanols /sterol intake, and increased physical activity is indicated in all patients with diabetes. A focus on a Mediterranean style diet or Dietary Approaches to Stop Hypertension (DASH) diet should be encouraged. In patients with elevated TG levels glycemic control is beneficial and dietary changes and lifestyle changes including weight loss and abstinence from alcohol should be undertaken. Secondary disorders and medications that raise TG levels should be evaluated. Optimize glycemic control to improve TG and HDL-C levels. The recommendations for lipid lowering therapy are shown in table 4. If one follows these recommendations almost all patients with diabetes over the age of 40 will be on statin therapy and many under the age of 40 will also be treated with statins. The addition of ezetimibe should be considered to further lower LDL-C levels in high-risk primary prevention patients. In very high-risk patients with ASCVD if the LDL-C level on statin therapy is greater than 70mg/dL the use of ezetimibe or a PCSK9 inhibitor should be considered. The use of fibrates or niacin with statins were generally not recommended as there is no evidence of benefit. However, in patients with ASCVD or other cardiovascular risk factors on a statin with controlled LDL-C but elevated TG levels (135-499mg/dL) the addition of icosapent ethyl can be considered. Finally, in patients with fasting TG levels greater than 500mg/dL an evaluation for secondary causes of hypertriglyceridemia should be initiated and consideration of drug therapy to reduce the risk of pancreatitis.

 

Table 4. ADA Recommendations for Lipid Lowering Therapy

Primary Prevention

Age 20-39: With additional risk factors may be reasonable to initiate statin therapy

Age 40-75: Use moderate-intensity statin therapy* in addition to lifestyle therapy

Age 40-75: If at higher cardiovascular risk, including those with one or more ASCVD risk factors, it is recommended to use high intensity statin therapy to reduce LDL cholesterol by >50% and to target an LDL-C <70 mg/dL

Age 40-75: If at higher cardiovascular risk, especially those with multiple ASCVD risk factors and an LDL-C >70 mg/dL, it may be reasonable to add ezetimibe or a PCSK9 inhibitor to maximum tolerated statin therapy

Age > 75: Initiating moderate intensity statin therapy is reasonable after discussion and in patient already on statin therapy it is reasonable to continue statin therapy

Secondary Prevention

All ages: High intensity statin therapy**/maximally tolerated stain

For people with diabetes and ASCVD, treatment with high intensity statin therapy is recommended to target an LDL-C reduction of >50% and an LDL-C l goal of <55 mg/dL. Addition of ezetimibe or a PCSK9 inhibitor is recommended if this goal is not achieved

*Moderate intensity statin- atorvastatin 10-20mg, rosuvastatin 5-10mg, simvastatin 20-40mg, pravastatin 40-80mg, lovastatin 40mg, Fluvastatin XL 80mg, pitavastatin 3-4mg.

**High Intensity statin- atorvastatin 40-80mg, rosuvastatin 20-40mg.

 

American College of Cardiology and American Heart Association Guidelines

 

The 2018 American College of Cardiology and American Heart Association (ACC/AHA) guidelines recommend the following (220). “In patients 40 to 75 years of age with diabetes mellitus and LDL-C ≥70 mg/dL (≥1.8 mmol/L), start moderate-intensity statin therapy without calculating 10-year ASCVD risk. In patients with diabetes mellitus at higher risk, especially those with multiple risk factors or those 50 to 75 years of age, it is reasonable to use a high-intensity statin to reduce the LDL-C level by ≥50%.” In patients with diabetes and ASCVD they recommend “In patients with clinical ASCVD, reduce LDL-C with high-intensity statin therapy or maximally tolerated statin therapy. The more LDL-C is reduced on statin therapy, the greater will be subsequent risk reduction. Use a maximally tolerated statin to lower LDLC levels by ≥50%. In very high-risk ASCVD, use an LDL-C threshold of 70 mg/dL (1.8 mmol/L) to consider addition of non-statins to statin therapy. Very high-risk includes a history of multiple major ASCVD events or 1 major ASCVD event and multiple high-risk conditions. In very high-risk ASCVD patients, it is reasonable to add ezetimibe to maximally tolerated statin therapy when the LDL-C level remains ≥70 mg/dL (≥1.8 mmol/L). In patients at very high risk whose LDL-C level remains ≥70 mg/dL (≥1.8 mmol/L) on maximally tolerated statin and ezetimibe therapy, adding a PCSK9 inhibitor is reasonable, although the long-term safety (>3 years) is uncertain and cost effectiveness is low at mid-2018 list prices.” With regards to testing they recommend “Assess adherence and percentage response to LDL-C–lowering medications and lifestyle changes with repeat lipid measurement 4 to 12 weeks after statin initiation or dose adjustment, repeated every 3 to 12 months as needed”. Finally, there are several diabetes specific risk enhancers that are independent of other risk factors that should be considered in deciding the risk of cardiovascular events in a patient with diabetes (Table 5).

 

Table 5. Diabetes Specific Risk Enhancers That are Independent of Other Risk Factors in Diabetes

Long duration (≥10 years for type 2 diabetes mellitus or ≥20 years for type 1 diabetes mellitus

Albuminuria ≥30 mcg of albumin/mg creatinine

eGFR <60 mL/min/1.73 m2

Retinopathy

Neuropathy

ABI <0.9

ABI indicates ankle-brachial index

 

American Association of Clinical Endocrinologists/American College of Endocrinology Guidelines

 

The American Association of Clinical Endocrinologists and American College of Endocrinology guidelines consider individuals with T2DM to be at high, very high, or extreme risk for ASCVD (221,222). Patients with T1DM and a duration of diabetes of more than 15 years or two or more risk factors, poorly controlled A1c, or insulin resistance with metabolic syndrome should be considered to have an equivalent risk to patients with T2DM (221). The recommended treatment goals are shown in Table 6.

 

Table 6. ASCVD Risk Categories and Treatment Goals

Risk Category

Risk Factors/10-year risk

LDL-C mg/dL

Non-HDL-C mg/dL

Apo B mg/dL

TG

mg/dL

Extreme Risk

Diabetes and clinical ASCVD

<55

<80

<70

<150

Very High Risk

Diabetes with one or more risk factors

<70

<100

<80

<150

High Risk

Diabetes and no other risk factors

<100

<130

<90

<150

 

European Society of Cardiology and European Atherosclerosis Society Guidelines

 

The European Society of Cardiology (ESC) and European Atherosclerosis Society (EAS) has 2019 guidelines for the treatment of lipids in patients with diabetes (223). These guidelines classify patients with diabetes as very high risk, high risk, or moderate risk (table 7). The recommended goals of therapy based on risk classification are shown in table 8. As with other guidelines intensification of statin therapy should be considered before the introduction of combination therapy. If the goal is not reached, statin combination with ezetimibe should be considered next.

 

Table 7. ESC/EAS Classification of Risk in Patients with Diabetes

Very High Risk- target organ damage, or at least three major risk factors, or early onset of T1DM of long duration (>20 years)

High Risk- without target organ damage, with DM duration >10 years or another additional risk factor

Moderate Risk- Young patients (T1DM <35 years; T2DM <50 years) with DM duration <10 years, without other risk factors. Calculated SCORE >1 % and <5% for 10-year risk of fatal CVD

 

Table 8. ESC/EAS Goals of Therapy in Patients with Diabetes

 

LDL-C

Non-HDL-C

Apo B

Very High Risk

>50% reduction and <55mg/dL (<1.4mmol/L)

<85mg/d;

<65mg/dL

High Risk

>50% reduction and <70mg/dL (<1.8mmol/L)

<100mg/dL

<80mg/dL

Moderate Risk

<100mg/dL

<130mg/dL

<100mg/dL

 

European Society of Cardiology Guidelines

 

The ESC has updated their guidelines in 2023 (224). An important new recommendation is that in patients with T2DM without symptomatic ASCVD or severe target organ damage, it is recommended to estimate 10-year CVD risk using SCORE2-Diabetes (225). This has resulted in a new classification of risk in patients with T2DM (table 9). The LDL-C goals for each category are shown in table 9.

 

Table 9. Cardiovascular Risk Categories and Goals in Patients with Type 2 Diabetes

Very high risk

Clinically established ASCVD or

Severe target organ damage or

10-year risk of CVD> 20% using SCORE2-Diabetes

LDL-C < 55mg/dl

Non-HDL-C <85mg/dL

High risk*

10-year risk of CVD 10% to < 20% using SCORE2-Diabetes

LDL-C < 70mg/dL

Non-HDL-C <100mg/dL

Moderate risk*

10-year risk of CVD 5% to <10% using SCORE2-Diabetes

LDL-C < 100mg/dL

Low Risk*

10-year risk of CVD <5% using SCORE2-Diabetes

no recommendations

*Patients not meeting the very high-risk category.

Severe target organ damage is defined as eGFR <45 mL/min/1.73 m2 irrespective of albuminuria; or eGFR 45–59 mL/min/1.73 m2 and microalbuminuria (UACR 30–300 mg/g; stage A2); or proteinuria (UACR>300 mg/g; stage A3), or presence of microvascular disease in at least three different sites [e.g., microalbuminuria (stage A2) plus retinopathy plus neuropathy].

 

My Guideline Recommendations

 

Thus, different organizations have proposed somewhat different recommendations for the treatment of lipids in patients with diabetes. Despite these differences it is clear that the vast majority of patients with diabetes will need to be treated with statins regardless of which guidelines one elects to follow.

 

The approach I use is to combine these recommendations (Tables 10 and 11). In patients with diabetes who have pre-existing ASCVD I initiate intensive statin therapy. I prefer LDL or non-HDL-C goals over percent reduction goals. Given the extensive data showing that the lower the LDL-C the greater the reduction in cardiovascular events most secondary prevention patients would benefit from the addition of ezetimibe to maximize LDL-C lowering without markedly increasing costs (226). In patients with diabetes 40-75 years of age without pre-existing ASCVD I calculate the 10-year risk of developing ASCVD (http://www.cvriskcalculator.com/) and identify risk enhancing factors (Table 5) and other factors that increase risk that are not included in the calculator (for example family history, inflammatory disorders, etc.). I initiate intensive statin therapy if the 10-year risk is > 7.5% or if there are multiple risk factors/risk enhancers. I initiate moderate statin therapy if the risk is < 7.5% without multiple risk factors/enhancers. Four to twelve weeks after initiating statin therapy I obtain a lipid panel to determine if the LDL and non-HDL-C levels are at goal. In patients with pre-existing ASCVD or multiple risk factors/risk enhancers (i.e., very high-risk patients) my goal is an LDL-C < 55mg/dL and a non-HDL-C < 80mg/dL. In patients that are at high-risk the goal my goal is an LDL-C < 70mg/dL and a non-HDL-C < 100mg/dL. In patients with moderate risk an LDL-C goal of < 100mg/dL and a non-HDL c < 130mg/dL is appropriate. If the levels are not at goal, I first adjust the statin dose until the patient is taking the maximally tolerated statin dose and then consider adding additional medications.  In patients with diabetes who are less than 40 years of age I initiate statin therapy if the patient has overt ASCVD, long standing diabetes, or risk factors/risk enhancers for ASCVD and the LDL and non-HDL-C levels are not at goal. In these younger patients I also calculate the life time risk of ASCVD events to start a discussion of beginning early therapy given the abundance of data indicating that initiating LDL-C lowering therapy early has great potential in markedly lowering ASCVD risk (226,227). In patients over 75 years of age with a reasonable life expectancy I begin moderate statin therapy and adjust based on response. When there is difficulty classifying a patient’s risk, I will obtain a coronary calcium score and use the score to help stratify the patient’s risk. In all cases the benefits and risks of lipid lowering therapy needs to be discussed with patients and the patient’s personnel preferences taken into account.

 

Table 10. ASCVD Risk Categories and Treatment Goals

Risk Category

Risk Factors/10-year risk

LDL-C mg/dL

Non-HDL-C mg/dL

Very High Risk

Diabetes and clinical ASCVD, multiple risk factors, 10-year risk > 20%

<55

<85

High Risk

Diabetes with one or more risk factors, 10-year risk >7.5% to <20%

<70

<100

Moderate Risk

Diabetes and no other risk factors. 10-year risk <7.5%

<100

<130

 

Table 11. Drug Therapy According to Risk Category that is Typically Required

Very High Risk

Intensive statin therapy + ezetimibe. Add PCSK9 if not close to goal

High Risk

Intensive statin therapy. Add ezetimibe if not at goal

Moderate Risk

Moderate statin therapy. Increase to intensive statin therapy if not at goal

 

TREATMENT OF LIPID ABNORMALITIES IN PATIENT WITH DIABETES

 

Life Style Changes and Weight Loss

 

Initial treatment of lipid disorders should focus on lifestyle changes. There is little debate that exercise is beneficial and that all patients with diabetes should, if possible, exercise for at least 150 minutes per week (for example 30 minutes 5 times per week). Exercise will decrease serum TG levels and increase HDL-C levels (an increase in HDL-C requires vigorous exercise) (124). Exercise increases fitness and improves insulin resistance even with limited weight loss; reductions in obesity are even more beneficial. It should be noted that many patients with diabetes may have substantial barriers to participating in exercise programs, such as comorbidities that limit exercise tolerance, risk of hypoglycemia, and presence of microvascular complications (visual impairment, neuropathy) that make exercise difficult.

 

Diet is debated to a greater extent and for detailed information on nutrition therapy for adults with diabetes see the consensus report by the American Diabetes Association (228). Everyone agrees that weight loss in obese patients is essential (124). But how this can be achieved is hotly debated with many different "experts" advocating different dietary approaches. The wide diversity of approach is likely due to the failure of any approach to be effective in the long term for the majority of obese patients with diabetes. If successful, weight loss will decrease serum TG levels, increase HDL-C levels, and modestly reduce LDL-C (124,229). To reduce LDL-C levels, it is important that the diet decrease saturated fat, trans fatty acids, and cholesterol intake. Increasing soluble fiber is also helpful.

 

It is debated whether a low fat, high complex carbohydrate diets vs. a high monounsaturated fat  diet is ideal for obese patients with diabetes (124). One can find "experts" in favor of either of these approaches and there are pros and cons to each approach. It is essential to recognize that both approaches reduce simple sugars, saturated fat, trans fatty acids, and cholesterol intake. The high complex carbohydrate diet will increase serum TG levels in some patients and if the amount of fat in the diet is markedly reduced serum HDL-C levels may decrease. In obese patients, it has been postulated that a diet high in monounsaturated fats, because of the increase in caloric density, will lead to an increase in weight gain. Both diets reduce saturated fat and cholesterol intake that will result in reductions in LDL-C levels. Additionally, both diets also reduce trans-fatty acid intake, which will have a beneficial effect on LDL and HDL-C levels and simple sugars, which will have a beneficial effect on TG levels. Very high levels of TG (>1000mg/dL), require diets that are very low in fat.

 

The available data do not indicate that any particular diet is best for inducing weight loss and it is essential to adapt the diet to fit the food preferences of the patient. Ultimately no weight loss diet will be successful if the patient cannot follow the diet for the long term and therefore the diet needs to be tailored to the specific preferences of the patient. For more detailed information on the effect of diet on lipid and lipoprotein levels and cardiovascular disease see the Endotext chapter “The Effect of Diet on Cardiovascular Disease and Lipid and Lipoprotein Levels” (229).

 

While it is widely accepted that lifestyle changes will decrease ASCVD events it should be recognized that the Look Ahead trial failed to demonstrate a reduction in ASCVD events (230). In this trial, over 5,000 overweight or obese patients with T2DM were randomized to either an intensive lifestyle intervention group that promoted weight loss through decreased caloric intake and increased physical activity or to a group that received diabetes support and education (control group). After a median follow-up of 9.6 years there was no difference in cardiovascular events (hazard ratio in the intervention group, 0.95; 95% CI 0.83 to 1.09; P=0.51). A major limitation of this study was that while the weight difference between groups was impressive during the first year of the trial, over time the differences greatly narrowed such that at the end of the trial the intensive group had a 6.0% weight loss while the control group had a 3.5% weight loss. This very modest weight difference demonstrates the difficulty in sustaining long term lifestyle changes. Thus, while weight loss and diet therapy are likely to be beneficial in reducing cardiovascular events, in clinical practice they are seldom sufficient because long-term life style changes are very difficult for most patients to maintain.

 

In contrast to the failure of lifestyle therapy in the Look Ahead trial to reduce cardiovascular events, the PREDIMED trial employing a Mediterranean diet (increased monounsaturated fats) did reduce the incidence of major ASCVD events (231,232). In this multicenter trial center trial, carried out in Spain, over 7,000 patients at high risk for developing ASCVD were randomized to three diets (primary prevention trial). A Mediterranean diet supplemented with extra-virgin olive oil, a Mediterranean diet supplemented with mixed nuts, or a control diet. Approximately 50% of the patients in this trial had T2DM. In the patients assigned to the Mediterranean diets there was 29% decrease in the primary end point (MI, stroke, and death from ASCVD). Subgroup analysis demonstrated that the Mediterranean diet was equally beneficial in patients with and without diabetes. The Mediterranean diet resulted in only a small but significant increase in HDL-C levels and a small decrease in both LDL-C and TG levels, suggesting that the beneficial effects were not mediated by changes in lipids (233).

 

The CORDIOPREV study was a single center randomized trial that compared a Mediterranean diet to a low-fat diet in 1,002 patients with ASCVD (234). Approximately 50% of the patients had diabetes. The Mediterranean diet contained a minimum of 35% of the calories as fat (22% monounsaturated fatty acids, 6% polyunsaturated fatty acids, and <10% saturated fat), 15% proteins, and a maximum of 50% carbohydrates while the low-fat diet contained less than 30% of total fat (<10% saturated fat, 12–14% monounsaturated fatty acids, and 6–8% polyunsaturated fatty acids), 15% protein, and a minimum of 55% carbohydrates. The risk of an ASCVD event was reduced by approximately 25-30% in the Mediterranean diet group. Whether these diets differed in their effects on fasting lipid levels is unknown.

 

Finally, another secondary prevention trial of a Mediterranean diet has also demonstrated a reduction in cardiovascular events. The Lyon Diet Heart Study randomized 584 patients who had a MI within 6 months to a Mediterranean type diet vs usual diet (235,236). There was a marked reduction in events in the group of patients randomized to the Mediterranean diet (cardiac death and nonfatal MI rate was 4.07 per 100 patient years in the control diet vs. 1.24 in the Mediterranean diet; p<0.0001). Unfortunately, there is no indication of the number of patients with diabetes in the Lyon Diet Heart Study or whether patients with diabetes responded similar to the entire group. Lipid levels were similar in both groups in this trial (235).

 

The results of these three randomized trials indicate that we should be encouraging our patients to follow a Mediterranean type diet. It is likely that the beneficial effects of the Mediterranean diet on ASCVD is mediated by multiple mechanisms with alterations in lipid levels making only a minor contribution.

 

For additional information on the effect of diets on lipid levels and ASCVD see the chapter entitled “The Effect of Diet on Cardiovascular Disease and Lipid and Lipoprotein Levels” in the Lipids and Lipoproteins section of Endotext (229).

 

Bariatric surgery can have profound effects on weight and can result in marked improvements in lipid profiles with a decrease in TG and LDL-C and an increase in HDL-C (124,229) Additionally, observational studies have shown a decrease in cardiovascular events following bariatric surgery in patients with and without diabetes (237-241). For additional information see the chapter entitled “Obesity and Dyslipidemia” (124).

 

Ethanol and simple sugars, in particular fructose, increase serum TG levels in susceptible patients. In patients with hypertriglyceridemia efforts should be made to reduce the intake of ethanol, simple sugars, and fructose (229).

 

Lastly, in the past some "experts" advocated the addition of fish oil supplements to reduce cardiovascular events. However, both the Origin Trial and the ASCEND Trial did not demonstrate that fish oil supplements were beneficial in patients with T2DM or patients at high risk for the development of T2DM (207,208). It should be recognized that higher doses of fish oil are required to lower serum triglyceride levels (~ 3-4 grams of DHA/EPA per day) and are useful in treating patients with high TG levels (242). Most studies of fish oil supplements in patients with diabetes have demonstrated that this is a safe approach and that worsening of glycemic control does not occur in patients with diabetes treated with fish oil supplements (242). Additionally, in some patient's high dose fish oil increases LDL-C levels, particularly when serum TG levels are very high (242). For additional information on fish oil see the chapter on Triglyceride Lowering Drugs (209).

 

Drug Therapy

 

The effect of statins, fibrates, niacin, ezetimibe, omega-3-fatty acids, bile acid sequestrants, bempedoic acid, and PCSK9 inhibitors on lipid levels in patients with diabetes is virtually identical to that seen in non-diabetic patients (Table 12). Below we will highlight issues particularly relevant to the use of these drugs in patients with diabetes. For detailed information on lipid lowering drugs see the chapters on Triglyceride Lowering Drugs and Cholesterol Lowering Drugs (141,209).

 

STATINS

 

Statins are easy to use and generally well tolerated by patients with diabetes. However, statins can adversely affect glucose homeostasis. In patients without diabetes the risk of developing diabetes is increased by approximately 10% with higher doses of statin causing a greater risk than more moderate doses (243,244). The mechanism for this adverse effect is unknown but older, obese patients with higher baseline glucose levels are at greatest risk. In patients with diabetes, an analysis of 9 studies with over 9,000 patients with diabetes reported that the patients randomized to statin therapy had a 0.12% higher HbA1c than the placebo group indicating that statin therapy is associated with only a very small increase in HbA1c levels in patients with diabetes, which is unlikely to be clinically significant (245). Individual studies such as CARDS and the Heart Protection Study have also shown only a very modest effect of statins on HbA1c levels in patients with diabetes (163,166,246). Muscle symptoms occur in patients with diabetes similar to what is observed in patients without diabetes.

 

EZETIMIBE

 

Ezetimibe is easy to use and generally well tolerated by patients with diabetes. Ezetimibe does not appear to increase the risk of new onset diabetes (199,247,248).

 

FIBRATES

 

Fibrates are easy to use and generally well tolerated by patients with diabetes. When combining fibrates with statin therapy it is best to use fenofibrate as the risk of inducing myositis is much less than when statins are used in combination with gemfibrozil, which can inhibit statin metabolism (249). In the ACCORD-LIPID Trial the incidence of muscle disorders was not increased in the statin + fenofibrate group compared to statin alone (189). The dose of fenofibrate needs to be adjusted in patients with renal disease and fenofibrate itself can induce a reversible increase in serum creatinine levels. It should be noted that marked reductions in HDL-C levels can occur in some patients treated with both fenofibrate and a TZD (250).

 

Diabetic Retinopathy

 

Fenofibrate has been shown to have beneficial effects on diabetic eye disease. The FIELD study, described earlier, was a randomized trial of fenofibrate vs. placebo in patients with T2DM. Laser treatment for retinopathy was significantly lower in the fenofibrate group than in the placebo group (3.4% patients on fenofibrate vs 4.9% on placebo; p=0.0002) (191). Fenofibrate therapy reduced the need for laser therapy to a similar extent for maculopathy (31% decrease) and for proliferative retinopathy (30% decrease). In the ophthalmology sub-study (n=1012), the primary endpoint of 2-step progression of retinopathy grade did not differ significantly between the fenofibrate and control groups (9.6% patients on fenofibrate vs 12.3% on placebo; p=0.19). In patients without pre-existing retinopathy there was no difference in progression (11.4% vs 11.7%; p=0.87). However, in patients with pre-existing retinopathy, significantly fewer patients on fenofibrate had a 2-step progression than did those on placebo (3.1% patients vs 14.6%; p=0.004). A composite endpoint of 2-step progression of retinopathy grade, macular edema, or laser treatments was significantly reduced in the fenofibrate group (HR 0.66, 95% CI 0.47-0.94; p=0.022).

 

In the ACCORD Study a subgroup of participants was evaluated for the progression of diabetic retinopathy by 3 or more steps on the Early Treatment Diabetic Retinopathy Study Severity Scale or the development of diabetic retinopathy necessitating laser photocoagulation or vitrectomy over a four-year period (190). At 4 years, the rates of progression of diabetic retinopathy were 6.5% with fenofibrate therapy (n=806) vs. 10.2% with placebo (n=787) (adjusted odds ratio, 0.60; 95% CI, 0.42 to 0.87; P = 0.006). Of note, this reduction in the progression of diabetic retinopathy was of a similar magnitude as intensive glycemic treatment vs. standard therapy.

 

Taken together these results indicate that fibrates have beneficial effects on the progression of diabetic retinopathy. The mechanisms by which fibrates decrease diabetic retinopathy are unknown.

 

Diabetic Nephropathy

 

The Diabetes Atherosclerosis Intervention Study (DAIS) evaluated the effect of fenofibrate therapy (n= 155) vs. placebo (n=159) on changes in urinary albumin excretion in patients with T2DM (251). Fenofibrate significantly reduced the worsening of albumin excretion (fenofibrate 8% vs. placebo 18%; P < 0.05). This effect was primarily due to reduced progression from normal albumin excretion to microalbuminuria (fenofibrate 3% vs. 18% placebo; P < 0.001).

 

In the FIELD trial, fenofibrate reduced urine albumin/creatinine ratio by 24% vs 11% in placebo group (p < 0.001), with 14% less progression and 18% more albuminuria regression (p < 0.001) in the fenofibrate group than in participants on placebo (252). As expected, fenofibrate therapy acutely increased plasma creatinine levels and decreased eGFR but over the long term, the increase in plasma creatinine was decreased in the fenofibrate group compared to the placebo group (14% decrease; p=0.01). Similarly, there was a slower annual decrease in eGFR in the fenofibrate group (1.19 vs 2.03 mL/min/1.73m2   annually, p < 0.001). The effect of fenofibrate on kidney function was greater in those with higher TG and lower HDL levels. End-stage renal disease, dialysis, renal transplant, and renal death were similar in the fenofibrate and placebo groups, but the incidence was low.

 

In the ACCORD-LIPID trial the post-randomization incidence of microalbuminuria was 38.2% in the fenofibrate group and 41.6% in the placebo group (p=0.01) and post-randomization incidence of macroalbumuria was 10.5% in the fibrate group and 12.3% in the placebo group (p=0.04) indicating a modest reduction in the development of proteinuria in patients treated with fenofibrate (189). There was no significant difference in the incidence of end-stage renal disease or need for dialysis between the fenofibrate group and the placebo group.

 

These studies suggest that fibrates may have a beneficial effect on diabetic kidney disease. One should recognize that reducing proteinuria is a surrogate marker and may not indicate a reduction in the development of end stage renal disease. The mechanisms accounting for decreased in proteinuria are unknown.

 

Amputations

 

In the FIELD study the risks of first amputation were decreased by 36% (p=0.02) and minor amputation events without known large-vessel disease by 47% (p=0.027) in the fenofibrate treated group (253). The reduction in amputations was independent of glucose control or dyslipidemia. No difference between the risks of major amputations was seen in the placebo and fenofibrate groups. The basis for this reduction in amputations is unknown.

 

Do Fibrates have an Independent Effect on Microvascular Disease?

 

Multiple studies cited above have now shown that fenofibrate decreases retinopathy, nephropathy, and amputation in the absence of large vessel disease. The effects are independent of blood glucose control. Given that there also was no effect of fenofibrate on cardiovascular (macrovascular) disease, these results may suggest that fenofibrate has an independent effect on microvascular disease.  Further studies are warranted, but these results should be taken into account when considering treatment of marked hypertriglyceridemia in patients with diabetes. 

 

BILE ACID SEQUESTRANTS

 

Bile acid sequestrants are relatively difficult to take due to GI toxicity (mainly constipation) (141). Diabetic subjects have an increased prevalence of constipation, which may be exacerbated by the use of bile acid sequestrants. On the other hand, in diabetic patients with diarrhea, the use of bile acid sequestrants may be advantageous. Bile acid sequestrants may also increase serum TG levels, which can be a problem in patients with diabetes who are already hypertriglyceridemic (141). An additional difficulty in using bile acid sequestrants is their potential for binding other drugs (141). Many drugs should be taken either two hours before or four hours after taking bile acid sequestrants to avoid the potential of decreased drug absorption. Patients with diabetes are frequently on multiple drugs for glycemic control, hypertension, etc., and it can sometimes be difficult to time the ingestion of bile resin sequestrants to avoid these other drugs. Colesevelam (Welchol) is a bile acid sequestrant that comes in pill, powder, or chewable bars and causes fewer side effects and has fewer interactions with other drugs than other preparations (254). The usual dose is3.75 grams per day and can be given as tablets (​take 6 tablets once daily or 3 tablets twice daily), oral suspension (​take one packet once daily), or chewable bars (take one bar once daily). Of particular note is that a number of studies have shown that colesevelam improves glycemic control in patients with diabetes resulting in an approximately 0.5% decrease in A1c levels (255).

 

NIACIN

 

Niacin is well known to cause skin flushing and itching and GI upset (256). Additionally, niacin reduces insulin sensitivity (i.e., causes insulin resistance), which can worsen glycemic control (256). Studies have shown that niacin is usually well tolerated in diabetic subjects who are in good glycemic control (257,258). In patients with poor glycemic control, niacin is more likely to adversely impact glucose levels. In the HPS2-Thrive trial, niacin therapy significantly worsened glycemic control in patients with diabetes and induced new onset diabetes in 1.3% of subjects that were non-diabetic (195). High doses of niacin are more likely to adversely affect glycemic control. Niacin can also increase serum uric acid levels and induce gout, both of which are already common in obese patients with T2DM (256). Additionally recent trials have reported an increased incidence of infection and bleeding with niacin therapy (256). However, niacin is the most effective drug in increasing HDL-C levels, which are frequently low in patients with diabetes. 

 

OMEGA-3-FATTY ACIDS

 

A Cochrane review of fish oil in patients with diabetes have demonstrated that this is a safe approach and does not result in worsening of glycemic control in patients with diabetes (242). Fish oil effectively lowers TG levels but, in some patients, particularly those with significant hypertriglyceridemia, high dose fish oil increases LDL-C levels (242). It should be noted that fish oil products that contain just EPA (Vascepa) do not adversely affect LDL-C levels (259). When using fish oil to lower serum TG levels it is important to recognize that one is aiming to provide 3-4 grams of DHA/EPA per day. The quantity of these active omega-3-fatty acids can vary greatly from product to product. Prescription fish oil products contain large amounts of these active ingredients whereas the amount of DHA/EPA in food supplements can vary greatly and in some products levels are very low. Additionally, while prescription omega-3-fatty acid preparations have high levels of quality control, omega-3-fish oil food supplements may have contaminants and the dosage may not be precisely controlled.

 

PCSK9 INHIBITORS

 

The beneficial effects of PCSK9 inhibitors in patients with diabetes is similar to what is observed in non-diabetic patients. Additionally, except for local reactions at the injection sites PCSK9 inhibitors do not seem to cause major side effects. PCSK9 inhibitors do not appear to increase the risk of developing new-onset diabetes (260,261).

 

BEMPEDOIC ACID

 

The effect of bempedoic acid on LDL-C levels in patients with diabetes are similar to the decreases seen on non-diabetics. Patients with T2DM often have elevated uric acid levels and an increased risk of gouty attacks and a major side effect of bempedoic acid is elevating uric acid levels (141). In clinical trials, 26% of bempedoic acid-treated patients with normal baseline uric acid values experienced hyperuricemia one or more times versus 9.5% in the placebo group. Elevations in blood uric acid levels may lead to the development of gout and gout was reported in 1.5% of patients treated with bempedoic acid vs. 0.4% of patients treated with placebo. The risk for gout attacks were higher in patients with a prior history of gout (11.2% for bempedoic acid treatment vs. 1.7% in the placebo group). In patients with no prior history of gout only 1% of patients treated with bempedoic acid and 0.3% of the placebo group had a gouty attack.

 

In a meta-analysis, bempedoic acid therapy was associated with a decrease in the onset of diabetes and worsening of diabetes  (RR 0.65, p = 0.03) (7/100 vs 6.4/100 patient years) (262). In a study focusing solely on the development of new onset diabetes it was reported that new-onset diabetes/hyperglycemia occurred less frequently with bempedoic acid vs placebo (263). In the bempedoic acid cardiovascular outcome trial (CLEAR Outcomes) the development of diabetes and worsening of pre-existing diabetes was similar in the bempedoic acid and placebo groups (187).  

 

Table 12. Effect of Lipid Lowering Drugs

 

LDL-C

HDL-C

TG

 

Statins

↓ 20-60%

↑ 5-15%

↓ 0-35%*

Bile acid sequestrants

↓ 10-30%

↑ 0-10%

↑ 0-10%**

Fibrates

↓ 0-15%***

↑ 5-15%

↓ 20-50%

Niacin

↓ 10-25%

↑ 10-30%

↓ 20-50%

Ezetimibe

↓ 15-25%

↑ 1-3%

↓ 10-20%

PCSK9 Inhibitors

↓ 50-60%

↑ 5-15%

↓ 5-20%

Bempedoic Acid

↓ 15-25%

↓ 5-6%

No change

High Dose Fish Oil

↑ 0- 50%***

↑ 4- 9%

↓ 20- 50%*

 *Patients with elevated TG have largest decrease

** In patients with high TG may cause marked increase

*** In patients with high TG may increase LDL

 

Therapeutic Approach

 

FIRST PRIORITY- LDL-C

 

The first priority in treating lipid disorders in patients with diabetes is to lower the LDL-C levels to goal, unless TG are markedly elevated (> 500- 1000mg/dL), which increases the risk of pancreatitis. LDL-C is the first priority because the database linking lowering LDL-C with reducing ASCVD is extremely strong and we now have the ability to markedly decrease LDL-C levels. Dietary therapy is the initial step but, in almost all patients, will not be sufficient to achieve the LDL-C goals. If patients are willing and able to make major changes in their diet it is possible to achieve significant reductions in LDL-C levels but this seldom occurs in clinical practice (264).

 

Statins are the first-choice drugs to lower LDL-C levels and the vast majority of diabetic patients will require statin therapy. There are several statins currently available in the US and they are available as generic drugs and therefore relatively inexpensive. The particular statin used may be driven by price, ability to lower LDL-C levels, and potential drug interactions. Patients with ASCVD (secondary prevention patients) should be started on intensive statin therapy (atorvastatin 40-80mg per day or rosuvastatin 20-40mg per day). Given the extensive data showing that the lower the LDL-C the greater the reduction in ASCVD events most secondary prevention patients would benefit from the addition of ezetimibe to maximize LDL-C lowering. Ezetimibe is now a generic drug and therefore this strategy will not markedly increase costs. Similarly, primary prevention patients who are at high risk for cardiovascular events will also benefit from the use of high intensity statin therapy in combination with ezetimibe. Primary prevention patients at moderate risk can be started on moderate intensity statin therapy.

 

If a patient is unable to tolerate statins or statins as monotherapy are not sufficient to lower LDL-C to goal the second-choice drug is either ezetimibe or a PCSK9 inhibitor. Ezetimibe can be added to any statin. PCSK9 inhibitors can also be added to any statin and are the drug of choice if a large decrease in LDL-C is required to reach goal (PCSK9 inhibitors will lower LDL-C levels by 50-60% when added to a statin, whereas ezetimibe will only lower LDL-C by approximately 20%).  Bile acid sequestrants and bempedoic acid are alternatives with the use of a bile acid sequestrant particularly useful if a reduction in A1c level is also needed. It should be noted that in statin intolerant patients with ASCVD or at high risk (approximately 45% with diabetes), bempedoic acid has been shown to reduce cardiovascular events by 13% (HR 0.87; 95% CI 0.79 to 0.96; P = 0.004) (187). Ezetimibe, PCSK9 inhibitors, bempedoic acid, and bile acid sequestrants additively lower LDL-C levels when used in combination with a statin, because these drugs increase hepatic LDL receptor levels by different mechanisms, thereby resulting in a reduction in serum LDL-C levels (141). Niacin and the fibrates also lower LDL-C levels but are not usually employed to lower LDL-C levels. 

 

SECOND PRIORITY- NON-HDL-C

 

The second priority should be non-HDL-C (non-HDL-C = total cholesterol – HDL-C), which is particularly important in patients with elevated TG levels (>150mg/dL). Non-HDL-C is a measure of all the pro-atherogenic apolipoprotein B containing particles. Numerous studies have shown that non-HDL-C is a strong risk factor for the development of ASCVD (265). The non-HDL-C goals are approximately 30mg/dL greater than the LDL-C goals. For example, if the LDL goal is <100mg/dL then the non-HDL-C goal would be <130mg/dL. Drugs that reduce either LDL-C or TG levels will reduce non-HDL-C levels. To lower TG levels initial therapy should focus on glycemic control and lifestyle changes including weight loss if indicated and a decrease in simple sugars and ethanol intake. Additionally, if possible, discontinue medications that increase triglyceride levels. As discussed above, studies with the omega-3-fatty acid icosapent ethyl (EPA; Vascepa) added to statin therapy have reduced the risk of cardiovascular events. The National Lipid Association has recommended “that for patients aged ≥45 years with clinical ASCVD, or aged ≥50 years with diabetes mellitus requiring medication plus ≥1 additional risk factor, with fasting TGs 135 to 499 mg/dL on high-intensity or maximally tolerated statin therapy (±ezetimibe), treatment with icosapent ethyl is recommended for ASCVD risk reduction” (266). As noted earlier in this chapter there is controversy regarding the benefits of icosapent ethyl on cardiovascular outcomes with some experts interpreting the beneficial results of the REDUCE-IT trial as being due to the adverse effects of the mineral oil placebo (217).

 

VERY HIGH TG

 

Patients with very high TG levels (> 500-1000 mg/dL) are at risk of pancreatitis and therefore lifestyle interventions including diet, exercise, and weight loss if indicated should be initiated early. Treatment is a low-fat diet and glycemic control. Additionally, decreasing simple sugars and avoiding alcohol is beneficial. When the TG levels are very elevated (> 1000mg/dL) a very low-fat diet (5-20% of calories as fat) should be the primary treatment until the TG levels decrease to < 1000mg/dL. Treating secondary disorders that raise TG levels and when possible, stopping drugs that increase TG levels is essential. If the TG levels remain above 500mg/dL the addition of fenofibrate or omega-3-fatty acids is indicated.

 

LOW HDL-C

 

While there is strong epidemiologic data linking low HDL-C levels with ASCVD there is no clinical trials demonstrating that increasing HDL-C levels reduce ASCVD. Thus, the use of drugs such as niacin to raise HDL-C levels is not recommended.

 

CONCLUSION

 

Patients with diabetes, particularly T2DM, often have dyslipidemia. Modern therapy of patients with diabetes demands that we aggressively treat lipids to reduce the high risk of ASCVD in this susceptible population and in those with very high TG to reduce the risk of pancreatitis.

 

REFERENCES

 

  1. Milicevic Z, Raz I, Beattie SD, Campaigne BN, Sarwat S, Gromniak E, Kowalska I, Galic E, Tan M, Hanefeld M. Natural history of cardiovascular disease in patients with diabetes: role of hyperglycemia. Diabetes Care 2008; 31 Suppl 2:S155-160
  2. Feingold KR, Siperstein MD. Diabetic vascular disease. Adv Intern Med 1986; 31:309-340
  3. Regensteiner JG, Golden S, Huebschmann AG, Barrett-Connor E, Chang AY, Chyun D, Fox CS, Kim C, Mehta N, Reckelhoff JF, Reusch JE, Rexrode KM, Sumner AE, Welty FK, Wenger NK, Anton B. Sex Differences in the Cardiovascular Consequences of Diabetes Mellitus: A Scientific Statement From the American Heart Association. Circulation 2015; 132:2424-2447
  4. Fox CS, Golden SH, Anderson C, Bray GA, Burke LE, de Boer IH, Deedwania P, Eckel RH, Ershow AG, Fradkin J, Inzucchi SE, Kosiborod M, Nelson RG, Patel MJ, Pignone M, Quinn L, Schauer PR, Selvin E, Vafiadis DK. Update on Prevention of Cardiovascular Disease in Adults With Type 2 Diabetes Mellitus in Light of Recent Evidence: A Scientific Statement From the American Heart Association and the American Diabetes Association. Diabetes Care 2015; 38:1777-1803
  5. Low Wang CC, Hess CN, Hiatt WR, Goldfine AB. Clinical Update: Cardiovascular Disease in Diabetes Mellitus: Atherosclerotic Cardiovascular Disease and Heart Failure in Type 2 Diabetes Mellitus - Mechanisms, Management, and Clinical Considerations. Circulation 2016; 133:2459-2502
  6. Kannel WB, McGee DL. Diabetes and cardiovascular disease. The Framingham study. JAMA 1979; 241:2035-2038
  7. Members Writing Group, Mozaffarian D, Benjamin EJ, Go AS, Arnett DK, Blaha MJ, Cushman M, Das SR, de Ferranti S, Despres JP, Fullerton HJ, Howard VJ, Huffman MD, Isasi CR, Jimenez MC, Judd SE, Kissela BM, Lichtman JH, Lisabeth LD, Liu S, Mackey RH, Magid DJ, McGuire DK, Mohler ER, 3rd, Moy CS, Muntner P, Mussolino ME, Nasir K, Neumar RW, Nichol G, Palaniappan L, Pandey DK, Reeves MJ, Rodriguez CJ, Rosamond W, Sorlie PD, Stein J, Towfighi A, Turan TN, Virani SS, Woo D, Yeh RW, Turner MB. Executive Summary: Heart Disease and Stroke Statistics--2016 Update: A Report From the American Heart Association. Circulation 2016; 133:447-454
  8. Haffner SM, Lehto S, Ronnemaa T, Pyorala K, Laakso M. Mortality from coronary heart disease in subjects with type 2 diabetes and in nondiabetic subjects with and without prior myocardial infarction. N Engl J Med 1998; 339:229-234
  9. Evans JM, Wang J, Morris AD. Comparison of cardiovascular risk between patients with type 2 diabetes and those who had had a myocardial infarction: cross sectional and cohort studies. BMJ 2002; 324:939-942
  10. Wannamethee SG, Shaper AG, Whincup PH, Lennon L, Sattar N. Impact of diabetes on cardiovascular disease risk and all-cause mortality in older men: influence of age at onset, diabetes duration, and established and novel risk factors. Arch Intern Med 2011; 171:404-410
  11. Howard BV, Best LG, Galloway JM, Howard WJ, Jones K, Lee ET, Ratner RE, Resnick HE, Devereux RB. Coronary heart disease risk equivalence in diabetes depends on concomitant risk factors. Diabetes Care 2006; 29:391-397
  12. Rawshani A, Rawshani A, Franzen S, Sattar N, Eliasson B, Svensson AM, Zethelius B, Miftaraj M, McGuire DK, Rosengren A, Gudbjornsdottir S. Risk Factors, Mortality, and Cardiovascular Outcomes in Patients with Type 2 Diabetes. N Engl J Med 2018; 379:633-644
  13. Lind M, Svensson AM, Kosiborod M, Gudbjornsdottir S, Pivodic A, Wedel H, Dahlqvist S, Clements M, Rosengren A. Glycemic control and excess mortality in type 1 diabetes. N Engl J Med 2014; 371:1972-1982
  14. de Ferranti SD, de Boer IH, Fonseca V, Fox CS, Golden SH, Lavie CJ, Magge SN, Marx N, McGuire DK, Orchard TJ, Zinman B, Eckel RH. Type 1 diabetes mellitus and cardiovascular disease: a scientific statement from the American Heart Association and American Diabetes Association. Diabetes Care 2014; 37:2843-2863
  15. Maahs DM, Daniels SR, de Ferranti SD, Dichek HL, Flynn J, Goldstein BI, Kelly AS, Nadeau KJ, Martyn-Nemeth P, Osganian SK, Quinn L, Shah AS, Urbina E. Cardiovascular disease risk factors in youth with diabetes mellitus: a scientific statement from the American Heart Association. Circulation 2014; 130:1532-1558
  16. Huxley RR, Peters SA, Mishra GD, Woodward M. Risk of all-cause mortality and vascular events in women versus men with type 1 diabetes: a systematic review and meta-analysis. Lancet Diabetes Endocrinol 2015; 3:198-206
  17. Rawshani A, Sattar N, Franzen S, Rawshani A, Hattersley AT, Svensson AM, Eliasson B, Gudbjornsdottir S. Excess mortality and cardiovascular disease in young adults with type 1 diabetes in relation to age at onset: a nationwide, register-based cohort study. Lancet 2018; 392:477-486
  18. Chillaron JJ, Flores Le-Roux JA, Benaiges D, Pedro-Botet J. Type 1 diabetes, metabolic syndrome and cardiovascular risk. Metabolism 2014; 63:181-187
  19. Constantino MI, Molyneaux L, Limacher-Gisler F, Al-Saeed A, Luo C, Wu T, Twigg SM, Yue DK, Wong J. Long-term complications and mortality in young-onset diabetes: type 2 diabetes is more hazardous and lethal than type 1 diabetes. Diabetes Care 2013; 36:3863-3869
  20. Preis SR, Hwang SJ, Coady S, Pencina MJ, D'Agostino RB, Sr., Savage PJ, Levy D, Fox CS. Trends in all-cause and cardiovascular disease mortality among women and men with and without diabetes mellitus in the Framingham Heart Study, 1950 to 2005. Circulation 2009; 119:1728-1735
  21. Matuleviciene-Anangen V, Rosengren A, Svensson AM, Pivodic A, Gudbjornsdottir S, Wedel H, Kosiborod M, Haraldsson B, Lind M. Glycaemic control and excess risk of major coronary events in persons with type 1 diabetes. Heart 2017; 103:1687-1695
  22. Miller RG, Orchard TJ, Costacou T. 30-Year Cardiovascular Disease in Type 1 Diabetes: Risk and Risk Factors Differ by Long-term Patterns of Glycemic Control. Diabetes Care 2022; 45:142-150
  23. Abraira C, Colwell JA, Nuttall FQ, Sawin CT, Nagel NJ, Comstock JP, Emanuele NV, Levin SR, Henderson W, Lee HS. Veterans Affairs Cooperative Study on glycemic control and complications in type II diabetes (VA CSDM). Results of the feasibility trial. Veterans Affairs Cooperative Study in Type II Diabetes. Diabetes Care1995; 18:1113-1123
  24. Goldner MG, Knatterud GL, Prout TE. Effects of hypoglycemic agents on vascular complications in patients with adult-onset diabetes. 3. Clinical implications of UGDP results. JAMA 1971; 218:1400-1410
  25. Meinert CL, Knatterud GL, Prout TE, Klimt CR. A study of the effects of hypoglycemic agents on vascular complications in patients with adult-onset diabetes. II. Mortality results. Diabetes 1970; 19:Suppl:789-830
  26. The effect of intensive treatment of diabetes on the development and progression of long-term complications in insulin-dependent diabetes mellitus. The Diabetes Control and Complications Trial Research Group. N Engl J Med 1993; 329:977-986
  27. Ohkubo Y, Kishikawa H, Araki E, Miyata T, Isami S, Motoyoshi S, Kojima Y, Furuyoshi N, Shichiri M. Intensive insulin therapy prevents the progression of diabetic microvascular complications in Japanese patients with non-insulin-dependent diabetes mellitus: a randomized prospective 6-year study. Diabetes Res Clin Pract 1995; 28:103-117
  28. Shichiri M, Kishikawa H, Ohkubo Y, Wake N. Long-term results of the Kumamoto Study on optimal diabetes control in type 2 diabetic patients. Diabetes Care 2000; 23 Suppl 2:B21-29
  29. Lachin JM, Orchard TJ, Nathan DM, Group DER. Update on cardiovascular outcomes at 30 years of the diabetes control and complications trial/epidemiology of diabetes interventions and complications study. Diabetes Care 2014; 37:39-43
  30. Nathan DM, Cleary PA, Backlund JY, Genuth SM, Lachin JM, Orchard TJ, Raskin P, Zinman B, Diabetes C, Complications Trial/Epidemiology of Diabetes I, Complications Study Research Group. Intensive diabetes treatment and cardiovascular disease in patients with type 1 diabetes. N Engl J Med 2005; 353:2643-2653
  31. Intensive blood-glucose control with sulphonylureas or insulin compared with conventional treatment and risk of complications in patients with type 2 diabetes (UKPDS 33). UK Prospective Diabetes Study (UKPDS) Group. Lancet 1998; 352:837-853
  32. Holman RR, Paul SK, Bethel MA, Matthews DR, Neil HA. 10-year follow-up of intensive glucose control in type 2 diabetes. N Engl J Med 2008; 359:1577-1589
  33. Malmberg K. Prospective randomised study of intensive insulin treatment on long term survival after acute myocardial infarction in patients with diabetes mellitus. DIGAMI (Diabetes Mellitus, Insulin Glucose Infusion in Acute Myocardial Infarction) Study Group. BMJ 1997; 314:1512-1515
  34. Mellbin LG, Malmberg K, Norhammar A, Wedel H, Ryden L, Investigators D. The impact of glucose lowering treatment on long-term prognosis in patients with type 2 diabetes and myocardial infarction: a report from the DIGAMI 2 trial. Eur Heart J 2008; 29:166-176
  35. Action to Control Cardiovascular Risk in Diabetes Study Group, Gerstein HC, Miller ME, Byington RP, Goff DC, Jr., Bigger JT, Buse JB, Cushman WC, Genuth S, Ismail-Beigi F, Grimm RH, Jr., Probstfield JL, Simons-Morton DG, Friedewald WT. Effects of intensive glucose lowering in type 2 diabetes. N Engl J Med 2008; 358:2545-2559
  36. Gerstein HC, Miller ME, Ismail-Beigi F, Largay J, McDonald C, Lochnan HA, Booth GL, ACCORD STUDY Group. Effects of intensive glycaemic control on ischaemic heart disease: analysis of data from the randomised, controlled ACCORD trial. Lancet 2014; 384:1936-1941
  37. ACCORD STUDY Group. Nine-Year Effects of 3.7 Years of Intensive Glycemic Control on Cardiovascular Outcomes. Diabetes Care 2016; 39:701-708
  38. Advance Collaborative Group, Patel A, MacMahon S, Chalmers J, Neal B, Billot L, Woodward M, Marre M, Cooper M, Glasziou P, Grobbee D, Hamet P, Harrap S, Heller S, Liu L, Mancia G, Mogensen CE, Pan C, Poulter N, Rodgers A, Williams B, Bompoint S, de Galan BE, Joshi R, Travert F. Intensive blood glucose control and vascular outcomes in patients with type 2 diabetes. N Engl J Med 2008; 358:2560-2572
  39. Zoungas S, Chalmers J, Neal B, Billot L, Li Q, Hirakawa Y, Arima H, Monaghan H, Joshi R, Colagiuri S, Cooper ME, Glasziou P, Grobbee D, Hamet P, Harrap S, Heller S, Lisheng L, Mancia G, Marre M, Matthews DR, Mogensen CE, Perkovic V, Poulter N, Rodgers A, Williams B, MacMahon S, Patel A, Woodward M, Group A-OC. Follow-up of blood-pressure lowering and glucose control in type 2 diabetes. N Engl J Med 2014; 371:1392-1406
  40. Duckworth W, Abraira C, Moritz T, Reda D, Emanuele N, Reaven PD, Zieve FJ, Marks J, Davis SN, Hayward R, Warren SR, Goldman S, McCarren M, Vitek ME, Henderson WG, Huang GD, Investigators V. Glucose control and vascular complications in veterans with type 2 diabetes. N Engl J Med 2009; 360:129-139
  41. Hayward RA, Reaven PD, Wiitala WL, Bahn GD, Reda DJ, Ge L, McCarren M, Duckworth WC, Emanuele NV, Investigators V. Follow-up of glycemic control and cardiovascular outcomes in type 2 diabetes. N Engl J Med2015; 372:2197-2206
  42. Reaven PD, Emanuele NV, Wiitala WL, Bahn GD, Reda DJ, McCarren M, Duckworth WC, Hayward RA, Investigators V. Intensive Glucose Control in Patients with Type 2 Diabetes - 15-Year Follow-up. N Engl J Med2019; 380:2215-2224
  43. Buehler AM, Cavalcanti AB, Berwanger O, Figueiro M, Laranjeira LN, Zazula AD, Kioshi B, Bugano DG, Santucci E, Sbruzzi G, Guimaraes HP, Carvalho VO, Bordin SA. Effect of tight blood glucose control versus conventional control in patients with type 2 diabetes mellitus: a systematic review with meta-analysis of randomized controlled trials. Cardiovasc Ther 2013; 31:147-160
  44. Control Group, Turnbull FM, Abraira C, Anderson RJ, Byington RP, Chalmers JP, Duckworth WC, Evans GW, Gerstein HC, Holman RR, Moritz TE, Neal BC, Ninomiya T, Patel AA, Paul SK, Travert F, Woodward M. Intensive glucose control and macrovascular outcomes in type 2 diabetes. Diabetologia 2009; 52:2288-2298
  45. Effect of intensive blood-glucose control with metformin on complications in overweight patients with type 2 diabetes (UKPDS 34). UK Prospective Diabetes Study (UKPDS) Group. Lancet 1998; 352:854-865
  46. Kooy A, de Jager J, Lehert P, Bets D, Wulffele MG, Donker AJ, Stehouwer CD. Long-term effects of metformin on metabolism and microvascular and macrovascular disease in patients with type 2 diabetes mellitus. Arch Intern Med 2009; 169:616-625
  47. Hong J, Zhang Y, Lai S, Lv A, Su Q, Dong Y, Zhou Z, Tang W, Zhao J, Cui L, Zou D, Wang D, Li H, Liu C, Wu G, Shen J, Zhu D, Wang W, Shen W, Ning G. Effects of metformin versus glipizide on cardiovascular outcomes in patients with type 2 diabetes and coronary artery disease. Diabetes Care 2013; 36:1304-1311
  48. Goldberg RB, Orchard TJ, Crandall JP, Boyko EJ, Budoff M, Dabelea D, Gadde KM, Knowler WC, Lee CG, Nathan DM, Watson K, Temprosa M. Effects of Long-term Metformin and Lifestyle Interventions on Cardiovascular Events in the Diabetes Prevention Program and Its Outcome Study. Circulation 2022; 145:1632-1641
  49. Goldberg RB, Aroda VR, Bluemke DA, Barrett-Connor E, Budoff M, Crandall JP, Dabelea D, Horton ES, Mather KJ, Orchard TJ, Schade D, Watson K, Temprosa M. Effect of Long-Term Metformin and Lifestyle in the Diabetes Prevention Program and Its Outcome Study on Coronary Artery Calcium. Circulation 2017; 136:52-64
  50. Fitch K, Abbara S, Lee H, Stavrou E, Sacks R, Michel T, Hemphill L, Torriani M, Grinspoon S. Effects of lifestyle modification and metformin on atherosclerotic indices among HIV-infected patients with the metabolic syndrome. AIDS 2012; 26:587-597
  51. Rosenstock J, Kahn SE, Johansen OE, Zinman B, Espeland MA, Woerle HJ, Pfarr E, Keller A, Mattheus M, Baanstra D, Meinicke T, George JT, von Eynatten M, McGuire DK, Marx N. Effect of Linagliptin vs Glimepiride on Major Adverse Cardiovascular Outcomes in Patients With Type 2 Diabetes: The CAROLINA Randomized Clinical Trial. JAMA 2019;
  52. Navigator Study Group, Holman RR, Haffner SM, McMurray JJ, Bethel MA, Holzhauer B, Hua TA, Belenkov Y, Boolell M, Buse JB, Buckley BM, Chacra AR, Chiang FT, Charbonnel B, Chow CC, Davies MJ, Deedwania P, Diem P, Einhorn D, Fonseca V, Fulcher GR, Gaciong Z, Gaztambide S, Giles T, Horton E, Ilkova H, Jenssen T, Kahn SE, Krum H, Laakso M, Leiter LA, Levitt NS, Mareev V, Martinez F, Masson C, Mazzone T, Meaney E, Nesto R, Pan C, Prager R, Raptis SA, Rutten GE, Sandstroem H, Schaper F, Scheen A, Schmitz O, Sinay I, Soska V, Stender S, Tamas G, Tognoni G, Tuomilehto J, Villamil AS, Vozar J, Califf RM. Effect of nateglinide on the incidence of diabetes and cardiovascular events. N Engl J Med 2010; 362:1463-1476
  53. Dormandy JA, Charbonnel B, Eckland DJ, Erdmann E, Massi-Benedetti M, Moules IK, Skene AM, Tan MH, Lefebvre PJ, Murray GD, Standl E, Wilcox RG, Wilhelmsen L, Betteridge J, Birkeland K, Golay A, Heine RJ, Koranyi L, Laakso M, Mokan M, Norkus A, Pirags V, Podar T, Scheen A, Scherbaum W, Schernthaner G, Schmitz O, Skrha J, Smith U, Taton J. Secondary prevention of macrovascular events in patients with type 2 diabetes in the PROactive Study (PROspective pioglitAzone Clinical Trial In macroVascular Events): a randomised controlled trial. Lancet 2005; 366:1279-1289
  54. Kernan WN, Viscoli CM, Furie KL, Young LH, Inzucchi SE, Gorman M, Guarino PD, Lovejoy AM, Peduzzi PN, Conwit R, Brass LM, Schwartz GG, Adams HP, Jr., Berger L, Carolei A, Clark W, Coull B, Ford GA, Kleindorfer D, O'Leary JR, Parsons MW, Ringleb P, Sen S, Spence JD, Tanne D, Wang D, Winder TR. Pioglitazone after Ischemic Stroke or Transient Ischemic Attack. N Engl J Med 2016; 374:1321-1331
  55. Vaccaro O, Masulli M, Nicolucci A, Bonora E, Del Prato S, Maggioni AP, Rivellese AA, Squatrito S, Giorda CB, Sesti G, Mocarelli P, Lucisano G, Sacco M, Signorini S, Cappellini F, Perriello G, Babini AC, Lapolla A, Gregori G, Giordano C, Corsi L, Buzzetti R, Clemente G, Di Cianni G, Iannarelli R, Cordera R, La Macchia O, Zamboni C, Scaranna C, Boemi M, Iovine C, Lauro D, Leotta S, Dall'Aglio E, Cannarsa E, Tonutti L, Pugliese G, Bossi AC, Anichini R, Dotta F, Di Benedetto A, Citro G, Antenucci D, Ricci L, Giorgino F, Santini C, Gnasso A, De Cosmo S, Zavaroni D, Vedovato M, Consoli A, Calabrese M, di Bartolo P, Fornengo P, Riccardi G. Effects on the incidence of cardiovascular events of the addition of pioglitazone versus sulfonylureas in patients with type 2 diabetes inadequately controlled with metformin (TOSCA.IT): a randomised, multicentre trial. Lancet Diabetes Endocrinol 2017; 5:887-897
  56. Vaccaro O, Lucisano G, Masulli M, Bonora E, Del Prato S, Rivellese AA, Giorda CB, Mocarelli P, Squatrito S, Maggioni AP, Riccardi G, Nicolucci A. Cardiovascular Effects of Pioglitazone or Sulfonylureas According to Pretreatment Risk: Moving Toward Personalized Care. J Clin Endocrinol Metab 2019; 104:3296-3302
  57. Mazzone T, Meyer PM, Feinstein SB, Davidson MH, Kondos GT, D'Agostino RB, Sr., Perez A, Provost JC, Haffner SM. Effect of pioglitazone compared with glimepiride on carotid intima-media thickness in type 2 diabetes: a randomized trial. JAMA 2006; 296:2572-2581
  58. Pfutzner A, Marx N, Lubben G, Langenfeld M, Walcher D, Konrad T, Forst T. Improvement of cardiovascular risk markers by pioglitazone is independent from glycemic control: results from the pioneer study. J Am Coll Cardiol2005; 45:1925-1931
  59. Nissen SE, Nicholls SJ, Wolski K, Nesto R, Kupfer S, Perez A, Jure H, De Larochelliere R, Staniloae CS, Mavromatis K, Saw J, Hu B, Lincoff AM, Tuzcu EM. Comparison of pioglitazone vs glimepiride on progression of coronary atherosclerosis in patients with type 2 diabetes: the PERISCOPE randomized controlled trial. JAMA2008; 299:1561-1573
  60. Nissen SE, Wolski K. Effect of rosiglitazone on the risk of myocardial infarction and death from cardiovascular causes. N Engl J Med 2007; 356:2457-2471
  61. Singh S, Loke YK, Furberg CD. Long-term risk of cardiovascular events with rosiglitazone: a meta-analysis. JAMA 2007; 298:1189-1195
  62. Home PD, Pocock SJ, Beck-Nielsen H, Curtis PS, Gomis R, Hanefeld M, Jones NP, Komajda M, McMurray JJ. Rosiglitazone evaluated for cardiovascular outcomes in oral agent combination therapy for type 2 diabetes (RECORD): a multicentre, randomised, open-label trial. Lancet 2009; 373:2125-2135
  63. Home PD, Pocock SJ, Beck-Nielsen H, Gomis R, Hanefeld M, Jones NP, Komajda M, McMurray JJ. Rosiglitazone evaluated for cardiovascular outcomes--an interim analysis. N Engl J Med 2007; 357:28-38
  64. Mahaffey KW, Hafley G, Dickerson S, Burns S, Tourt-Uhlig S, White J, Newby LK, Komajda M, McMurray J, Bigelow R, Home PD, Lopes RD. Results of a reevaluation of cardiovascular outcomes in the RECORD trial. Am Heart J 2013; 166:240-249 e241
  65. Bach RG, Brooks MM, Lombardero M, Genuth S, Donner TW, Garber A, Kennedy L, Monrad ES, Pop-Busui R, Kelsey SF, Frye RL. Rosiglitazone and outcomes for patients with diabetes mellitus and coronary artery disease in the Bypass Angioplasty Revascularization Investigation 2 Diabetes (BARI 2D) trial. Circulation 2013; 128:785-794
  66. Feingold KR. Oral and Injectable (Non-Insulin) Pharmacological Agents for Type 2 Diabetes. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, Dungan K, Grossman A, Hershman JM, Kaltsas G, Koch C, Kopp P, Korbonits M, McLachlan R, Morley JE, New M, Perreault L, Purnell J, Rebar R, Singer F, Trence DL, Vinik A, Wilson DP, eds. Endotext. South Dartmouth (MA)2022.
  67. Scirica BM, Bhatt DL, Braunwald E, Steg PG, Davidson J, Hirshberg B, Ohman P, Frederich R, Wiviott SD, Hoffman EB, Cavender MA, Udell JA, Desai NR, Mosenzon O, McGuire DK, Ray KK, Leiter LA, Raz I. Saxagliptin and cardiovascular outcomes in patients with type 2 diabetes mellitus. N Engl J Med 2013; 369:1317-1326
  68. Scirica BM, Braunwald E, Raz I, Cavender MA, Morrow DA, Jarolim P, Udell JA, Mosenzon O, Im K, Umez-Eronini AA, Pollack PS, Hirshberg B, Frederich R, Lewis BS, McGuire DK, Davidson J, Steg PG, Bhatt DL. Heart failure, saxagliptin, and diabetes mellitus: observations from the SAVOR-TIMI 53 randomized trial. Circulation 2014; 130:1579-1588
  69. White WB, Cannon CP, Heller SR, Nissen SE, Bergenstal RM, Bakris GL, Perez AT, Fleck PR, Mehta CR, Kupfer S, Wilson C, Cushman WC, Zannad F. Alogliptin after acute coronary syndrome in patients with type 2 diabetes. N Engl J Med 2013; 369:1327-1335
  70. Zannad F, Cannon CP, Cushman WC, Bakris GL, Menon V, Perez AT, Fleck PR, Mehta CR, Kupfer S, Wilson C, Lam H, White WB. Heart failure and mortality outcomes in patients with type 2 diabetes taking alogliptin versus placebo in EXAMINE: a multicentre, randomised, double-blind trial. Lancet 2015; 385:2067-2076
  71. Green JB, Bethel MA, Armstrong PW, Buse JB, Engel SS, Garg J, Josse R, Kaufman KD, Koglin J, Korn S, Lachin JM, McGuire DK, Pencina MJ, Standl E, Stein PP, Suryawanshi S, Van de Werf F, Peterson ED, Holman RR. Effect of Sitagliptin on Cardiovascular Outcomes in Type 2 Diabetes. N Engl J Med 2015; 373:232-242
  72. Rosenstock J, Perkovic V, Johansen OE, Cooper ME, Kahn SE, Marx N, Alexander JH, Pencina M, Toto RD, Wanner C, Zinman B, Woerle HJ, Baanstra D, Pfarr E, Schnaidt S, Meinicke T, George JT, von Eynatten M, McGuire DK. Effect of Linagliptin vs Placebo on Major Cardiovascular Events in Adults With Type 2 Diabetes and High Cardiovascular and Renal Risk: The CARMELINA Randomized Clinical Trial. JAMA 2019 Jan 1;321(1):69-79.
  73. Zinman B, Wanner C, Lachin JM, Fitchett D, Bluhmki E, Hantel S, Mattheus M, Devins T, Johansen OE, Woerle HJ, Broedl UC, Inzucchi SE. Empagliflozin, Cardiovascular Outcomes, and Mortality in Type 2 Diabetes. N Engl J Med 2015; 373:2117-2128
  74. Fitchett D, Inzucchi SE, Cannon CP, McGuire DK, Scirica BM, Johansen OE, Sambevski S, Kaspers S, Pfarr E, George JT, Zinman B. Empagliflozin Reduced Mortality and Hospitalization for Heart Failure Across the Spectrum of Cardiovascular Risk in the EMPA-REG OUTCOME Trial. Circulation 2019; 139:1384-1395
  75. Fitchett D, Butler J, van de Borne P, Zinman B, Lachin JM, Wanner C, Woerle HJ, Hantel S, George JT, Johansen OE, Inzucchi SE. Effects of empagliflozin on risk for cardiovascular death and heart failure hospitalization across the spectrum of heart failure risk in the EMPA-REG OUTCOME(R) trial. Eur Heart J 2018; 39:363-370
  76. Neal B, Perkovic V, Mahaffey KW, de Zeeuw D, Fulcher G, Erondu N, Shaw W, Law G, Desai M, Matthews DR. Canagliflozin and Cardiovascular and Renal Events in Type 2 Diabetes. N Engl J Med 2017; 377:644-657
  77. Perkovic V, Jardine MJ, Neal B, Bompoint S, Heerspink HJL, Charytan DM, Edwards R, Agarwal R, Bakris G, Bull S, Cannon CP, Capuano G, Chu PL, de Zeeuw D, Greene T, Levin A, Pollock C, Wheeler DC, Yavin Y, Zhang H, Zinman B, Meininger G, Brenner BM, Mahaffey KW. Canagliflozin and Renal Outcomes in Type 2 Diabetes and Nephropathy. N Engl J Med 2019; 380:2295-2306
  78. Jardine MJ, Zhou Z, Mahaffey KW, Oshima M, Agarwal R, Bakris G, Bajaj HS, Bull S, Cannon CP, Charytan DM, de Zeeuw D, Di Tanna GL, Greene T, Heerspink HJL, Levin A, Neal B, Pollock C, Qiu R, Sun T, Wheeler DC, Zhang H, Zinman B, Rosenthal N, Perkovic V. Renal, Cardiovascular, and Safety Outcomes of Canagliflozin by Baseline Kidney Function: A Secondary Analysis of the CREDENCE Randomized Trial. J Am Soc Nephrol2020; 31:1128-1139
  79. Wiviott SD, Raz I, Bonaca MP, Mosenzon O, Kato ET, Cahn A, Silverman MG, Zelniker TA, Kuder JF, Murphy SA, Bhatt DL, Leiter LA, McGuire DK, Wilding JPH, Ruff CT, Gause-Nilsson IAM, Fredriksson M, Johansson PA, Langkilde AM, Sabatine MS. Dapagliflozin and Cardiovascular Outcomes in Type 2 Diabetes. N Engl J Med2018;
  80. Furtado RHM, Bonaca MP, Raz I, Zelniker TA, Mosenzon O, Cahn A, Kuder J, Murphy SA, Bhatt DL, Leiter LA, McGuire DK, Wilding JPH, Ruff CT, Nicolau JC, Gause-Nilsson IAM, Fredriksson M, Langkilde AM, Sabatine MS, Wiviott SD. Dapagliflozin and Cardiovascular Outcomes in Patients With Type 2 Diabetes Mellitus and Previous Myocardial Infarction. Circulation 2019; 139:2516-2527
  81. Cannon CP, Pratley R, Dagogo-Jack S, Mancuso J, Huyck S, Masiukiewicz U, Charbonnel B, Frederich R, Gallo S, Cosentino F, Shih WJ, Gantz I, Terra SG, Cherney DZI, McGuire DK. Cardiovascular Outcomes with Ertugliflozin in Type 2 Diabetes. N Engl J Med 2020; 383:1425-1435
  82. Cosentino F, Cannon CP, Cherney DZI, Masiukiewicz U, Pratley R, Dagogo-Jack S, Frederich R, Charbonnel B, Mancuso J, Shih WJ, Terra SG, Cater NB, Gantz I, McGuire DK. Efficacy of Ertugliflozin on Heart Failure-Related Events in Patients With Type 2 Diabetes Mellitus and Established Atherosclerotic Cardiovascular Disease: Results of the VERTIS CV Trial. Circulation 2020; 142:2205-2215
  83. Feingold KR. Oral and Injectable (Non-Insulin) Pharmacological Agents for the Treatment of Type 2 Diabetes. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Shah AS, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA)2022.
  84. Pfeffer MA, Claggett B, Diaz R, Dickstein K, Gerstein HC, Kober LV, Lawson FC, Ping L, Wei X, Lewis EF, Maggioni AP, McMurray JJ, Probstfield JL, Riddle MC, Solomon SD, Tardif JC. Lixisenatide in Patients with Type 2 Diabetes and Acute Coronary Syndrome. N Engl J Med 2015; 373:2247-2257
  85. Marso SP, Daniels GH, Brown-Frandsen K, Kristensen P, Mann JF, Nauck MA, Nissen SE, Pocock S, Poulter NR, Ravn LS, Steinberg WM, Stockner M, Zinman B, Bergenstal RM, Buse JB. Liraglutide and Cardiovascular Outcomes in Type 2 Diabetes. N Engl J Med 2016; 375:311-322
  86. Mann JFE, Fonseca V, Mosenzon O, Raz I, Goldman B, Idorn T, von Scholten BJ, Poulter NR. Effects of Liraglutide Versus Placebo on Cardiovascular Events in Patients With Type 2 Diabetes Mellitus and Chronic Kidney Disease. Circulation 2018; 138:2908-2918
  87. Verma S, Poulter NR, Bhatt DL, Bain SC, Buse JB, Leiter LA, Nauck MA, Pratley RE, Zinman B, Orsted DD, Monk Fries T, Rasmussen S, Marso SP. Effects of Liraglutide on Cardiovascular Outcomes in Patients With Type 2 Diabetes Mellitus With or Without History of Myocardial Infarction or Stroke. Circulation 2018; 138:2884-2894
  88. Marso SP, Bain SC, Consoli A, Eliaschewitz FG, Jodar E, Leiter LA, Lingvay I, Rosenstock J, Seufert J, Warren ML, Woo V, Hansen O, Holst AG, Pettersson J, Vilsboll T. Semaglutide and Cardiovascular Outcomes in Patients with Type 2 Diabetes. N Engl J Med 2016;
  89. Husain M, Birkenfeld AL, Donsmark M, Dungan K, Eliaschewitz FG, Franco DR, Jeppesen OK, Lingvay I, Mosenzon O, Pedersen SD, Tack CJ, Thomsen M, Vilsboll T, Warren ML, Bain SC. Oral Semaglutide and Cardiovascular Outcomes in Patients with Type 2 Diabetes. N Engl J Med 2019; 381:841-851
  90. Holman RR, Bethel MA, Mentz RJ, Thompson VP, Lokhnygina Y, Buse JB, Chan JC, Choi J, Gustavson SM, Iqbal N, Maggioni AP, Marso SP, Ohman P, Pagidipati NJ, Poulter N, Ramachandran A, Zinman B, Hernandez AF. Effects of Once-Weekly Exenatide on Cardiovascular Outcomes in Type 2 Diabetes. N Engl J Med 2017; 377:1228-1239
  91. Hernandez AF, Green JB, Janmohamed S, D'Agostino RB, Sr., Granger CB, Jones NP, Leiter LA, Rosenberg AE, Sigmon KN, Somerville MC, Thorpe KM, McMurray JJV, Del Prato S. Albiglutide and cardiovascular outcomes in patients with type 2 diabetes and cardiovascular disease (Harmony Outcomes): a double-blind, randomised placebo-controlled trial. Lancet 2018; 392:1519-1529
  92. Gerstein HC, Colhoun HM, Dagenais GR, Diaz R, Lakshmanan M, Pais P, Probstfield J, Riesmeyer JS, Riddle MC, Ryden L, Xavier D, Atisso CM, Dyal L, Hall S, Rao-Melacini P, Wong G, Avezum A, Basile J, Chung N, Conget I, Cushman WC, Franek E, Hancu N, Hanefeld M, Holt S, Jansky P, Keltai M, Lanas F, Leiter LA, Lopez-Jaramillo P, Cardona Munoz EG, Pirags V, Pogosova N, Raubenheimer PJ, Shaw JE, Sheu WH, Temelkova-Kurktschiev T. Dulaglutide and cardiovascular outcomes in type 2 diabetes (REWIND): a double-blind, randomised placebo-controlled trial. Lancet 2019; 394:121-130
  93. Gerstein HC, Hart R, Colhoun HM, Diaz R, Lakshmanan M, Botros FT, Probstfield J, Riddle MC, Ryden L, Atisso CM, Dyal L, Hall S, Avezum A, Basile J, Conget I, Cushman WC, Hancu N, Hanefeld M, Jansky P, Keltai M, Lanas F, Leiter LA, Lopez-Jaramillo P, Munoz EGC, Pogosova N, Raubenheimer PJ, Shaw JE, Sheu WH, Temelkova-Kurktschiev T. The effect of dulaglutide on stroke: an exploratory analysis of the REWIND trial. Lancet Diabetes Endocrinol 2020; 8:106-114
  94. Kristensen SL, Rorth R, Jhund PS, Docherty KF, Sattar N, Preiss D, Kober L, Petrie MC, McMurray JJV. Cardiovascular, mortality, and kidney outcomes with GLP-1 receptor agonists in patients with type 2 diabetes: a systematic review and meta-analysis of cardiovascular outcome trials. Lancet Diabetes Endocrinol 2019; 7:776-785
  95. Sposito AC, Berwanger O, de Carvalho LSF, Saraiva JFK. GLP-1RAs in type 2 diabetes: mechanisms that underlie cardiovascular effects and overview of cardiovascular outcome data. Cardiovasc Diabetol 2018; 17:157
  96. Sattar N, McGuire DK, Pavo I, Weerakkody GJ, Nishiyama H, Wiese RJ, Zoungas S. Tirzepatide cardiovascular event risk assessment: a pre-specified meta-analysis. Nat Med 2022; 28:591-598
  97. Chiasson JL, Josse RG, Gomis R, Hanefeld M, Karasik A, Laakso M. Acarbose treatment and the risk of cardiovascular disease and hypertension in patients with impaired glucose tolerance: the STOP-NIDDM trial. JAMA 2003; 290:486-494
  98. Yun P, Du AM, Chen XJ, Liu JC, Xiao H. Effect of Acarbose on Long-Term Prognosis in Acute Coronary Syndromes Patients with Newly Diagnosed Impaired Glucose Tolerance. J Diabetes Res 2016; 2016:1602083
  99. Holman RR, Coleman RL, Chan JCN, Chiasson JL, Feng H, Ge J, Gerstein HC, Gray R, Huo Y, Lang Z, McMurray JJ, Ryden L, Schroder S, Sun Y, Theodorakis MJ, Tendera M, Tucker L, Tuomilehto J, Wei Y, Yang W, Wang D, Hu D, Pan C. Effects of acarbose on cardiovascular and diabetes outcomes in patients with coronary heart disease and impaired glucose tolerance (ACE): a randomised, double-blind, placebo-controlled trial. Lancet Diabetes Endocrinol 2017; 5:877-886
  100. Gaziano JM, Cincotta AH, O'Connor CM, Ezrokhi M, Rutty D, Ma ZJ, Scranton RE. Randomized clinical trial of quick-release bromocriptine among patients with type 2 diabetes on overall safety and cardiovascular outcomes. Diabetes Care 2010; 33:1503-1508
  101. Younk LM, Davis SN. Evaluation of colesevelam hydrochloride for the treatment of type 2 diabetes. Expert Opin Drug Metab Toxicol 2012; 8:515-525
  102. The Lipid Research Clinics Coronary Primary Prevention Trial results. I. Reduction in incidence of coronary heart disease. JAMA 1984; 251:351-364
  103. The Lipid Research Clinics Coronary Primary Prevention Trial results. II. The relationship of reduction in incidence of coronary heart disease to cholesterol lowering. JAMA 1984; 251:365-374
  104. Investigators OT, Gerstein HC, Bosch J, Dagenais GR, Diaz R, Jung H, Maggioni AP, Pogue J, Probstfield J, Ramachandran A, Riddle MC, Ryden LE, Yusuf S. Basal insulin and cardiovascular and other outcomes in dysglycemia. N Engl J Med 2012; 367:319-328
  105. Origin Trial Investigators. Cardiovascular and Other Outcomes Postintervention With Insulin Glargine and Omega-3 Fatty Acids (ORIGINALE). Diabetes Care 2016; 39:709-716
  106. Marso SP, McGuire DK, Zinman B, Poulter NR, Emerson SS, Pieber TR, Pratley RE, Haahr PM, Lange M, Brown-Frandsen K, Moses A, Skibsted S, Kvist K, Buse JB. Efficacy and Safety of Degludec versus Glargine in Type 2 Diabetes. N Engl J Med 2017; 377:723-732
  107. Chaitman BR, Hardison RM, Adler D, Gebhart S, Grogan M, Ocampo S, Sopko G, Ramires JA, Schneider D, Frye RL. The Bypass Angioplasty Revascularization Investigation 2 Diabetes randomized trial of different treatment strategies in type 2 diabetes mellitus with stable ischemic heart disease: impact of treatment strategy on cardiac mortality and myocardial infarction. Circulation 2009; 120:2529-2540
  108. Bari D. Study Group, Frye RL, August P, Brooks MM, Hardison RM, Kelsey SF, MacGregor JM, Orchard TJ, Chaitman BR, Genuth SM, Goldberg SH, Hlatky MA, Jones TL, Molitch ME, Nesto RW, Sako EY, Sobel BE. A randomized trial of therapies for type 2 diabetes and coronary artery disease. N Engl J Med 2009; 360:2503-2515
  109. ElSayed NA, Aleppo G, Aroda VR, Bannuru RR, Brown FM, Bruemmer D, Collins BS, Hilliard ME, Isaacs D, Johnson EL, Kahan S, Khunti K, Leon J, Lyons SK, Perry ML, Prahalad P, Pratley RE, Seley JJ, Stanton RC, Gabbay RA, on behalf of the American Diabetes A. 9. Pharmacologic Approaches to Glycemic Treatment: Standards of Care in Diabetes-2023. Diabetes Care 2023; 46:S140-S157
  110. de Ferranti SD, de Boer IH, Fonseca V, Fox CS, Golden SH, Lavie CJ, Magge SN, Marx N, McGuire DK, Orchard TJ, Zinman B, Eckel RH. Type 1 diabetes mellitus and cardiovascular disease: a scientific statement from the American Heart Association and American Diabetes Association. Circulation 2014; 130:1110-1130
  111. Martin-Timon I, Sevillano-Collantes C, Segura-Galindo A, Del Canizo-Gomez FJ. Type 2 diabetes and cardiovascular disease: Have all risk factors the same strength? World J Diabetes 2014; 5:444-470
  112. Turner RC, Millns H, Neil HA, Stratton IM, Manley SE, Matthews DR, Holman RR. Risk factors for coronary artery disease in non-insulin dependent diabetes mellitus: United Kingdom Prospective Diabetes Study (UKPDS: 23). BMJ 1998; 316:823-828
  113. Hovingh GK, Rader DJ, Hegele RA. HDL re-examined. Curr Opin Lipidol 2015; 26:127-132
  114. Brunzell JD, Davidson M, Furberg CD, Goldberg RB, Howard BV, Stein JH, Witztum JL. Lipoprotein management in patients with cardiometabolic risk: consensus statement from the American Diabetes Association and the American College of Cardiology Foundation. Diabetes Care 2008; 31:811-822
  115. Ference BA, Kastelein JJP, Ray KK, Ginsberg HN, Chapman MJ, Packard CJ, Laufs U, Oliver-Williams C, Wood AM, Butterworth AS, Di Angelantonio E, Danesh J, Nicholls SJ, Bhatt DL, Sabatine MS, Catapano AL. Association of Triglyceride-Lowering LPL Variants and LDL-C-Lowering LDLR Variants With Risk of Coronary Heart Disease. JAMA 2019; 321:364-373
  116. Nordestgaard BG. Triglyceride-Rich Lipoproteins and Atherosclerotic Cardiovascular Disease: New Insights From Epidemiology, Genetics, and Biology. Circ Res 2016; 118:547-563
  117. Ganjali S, Dallinga-Thie GM, Simental-Mendia LE, Banach M, Pirro M, Sahebkar A. HDL functionality in type 1 diabetes. Atherosclerosis 2017; 267:99-109
  118. Ginsberg HN, MacCallum PR. The obesity, metabolic syndrome, and type 2 diabetes mellitus pandemic: Part I. Increased cardiovascular disease risk and the importance of atherogenic dyslipidemia in persons with the metabolic syndrome and type 2 diabetes mellitus. J Cardiometab Syndr 2009; 4:113-119
  119. Goldberg IJ. Clinical review 124: Diabetic dyslipidemia: causes and consequences. J Clin Endocrinol Metab2001; 86:965-971
  120. Krauss RM. Lipids and lipoproteins in patients with type 2 diabetes. Diabetes Care 2004; 27:1496-1504
  121. Wu L, Parhofer KG. Diabetic dyslipidemia. Metabolism 2014; 63:1469-1479
  122. Taskinen MR, Boren J. New insights into the pathophysiology of dyslipidemia in type 2 diabetes. Atherosclerosis2015; 239:483-495
  123. Feingold KR, Grunfeld C, Pang M, Doerrler W, Krauss RM. LDL subclass phenotypes and triglyceride metabolism in non-insulin-dependent diabetes. Arterioscler Thromb 1992; 12:1496-1502
  124. Feingold KR. Obesity and Dyslipidemia. In: De Groot LJ, Beck-Peccoz P, Chrousos G, Dungan K, Grossman A, Hershman JM, Koch C, McLachlan R, New M, Rebar R, Singer F, Vinik A, Weickert MO, eds. Endotext. South Dartmouth (MA)2023.
  125. Morgantini C, Natali A, Boldrini B, Imaizumi S, Navab M, Fogelman AM, Ferrannini E, Reddy ST. Anti-inflammatory and antioxidant properties of HDLs are impaired in type 2 diabetes. Diabetes 2011; 60:2617-2623
  126. Apro J, Tietge UJ, Dikkers A, Parini P, Angelin B, Rudling M. Impaired Cholesterol Efflux Capacity of High-Density Lipoprotein Isolated From Interstitial Fluid in Type 2 Diabetes Mellitus-Brief Report. Arterioscler Thromb Vasc Biol 2016; 36:787-791
  127. Manjunatha S, Distelmaier K, Dasari S, Carter RE, Kudva YC, Nair KS. Functional and proteomic alterations of plasma high density lipoproteins in type 1 diabetes mellitus. Metabolism 2016; 65:1421-1431
  128. Enkhmaa B, Anuurad E, Berglund L. Lipoprotein (a): impact by ethnicity and environmental and medical conditions. J Lipid Res 2016; 57:1111-1125
  129. Durrington PN, Schofield JD, Siahmansur T, Soran H. Lipoprotein (a): gene genie. Curr Opin Lipidol 2014; 25:289-296
  130. Gudbjartsson DF, Thorgeirsson G, Sulem P, Helgadottir A, Gylfason A, Saemundsdottir J, Bjornsson E, Norddahl GL, Jonasdottir A, Jonasdottir A, Eggertsson HP, Gretarsdottir S, Thorleifsson G, Indridason OS, Palsson R, Jonasson F, Jonsdottir I, Eyjolfsson GI, Sigurdardottir O, Olafsson I, Danielsen R, Matthiasson SE, Kristmundsdottir S, Halldorsson BV, Hreidarsson AB, Valdimarsson EM, Gudnason T, Benediktsson R, Steinthorsdottir V, Thorsteinsdottir U, Holm H, Stefansson K. Lipoprotein(a) Concentration and Risks of Cardiovascular Disease and Diabetes. J Am Coll Cardiol 2019; 74:2982-2994
  131. Ferrannini E, DeFronzo RA. Impact of glucose-lowering drugs on cardiovascular disease in type 2 diabetes. Eur Heart J 2015; 36:2288-2296
  132. Wulffele MG, Kooy A, de Zeeuw D, Stehouwer CD, Gansevoort RT. The effect of metformin on blood pressure, plasma cholesterol and triglycerides in type 2 diabetes mellitus: a systematic review. J Intern Med 2004; 256:1-14
  133. Ratner R, Goldberg R, Haffner S, Marcovina S, Orchard T, Fowler S, Temprosa M, Diabetes Prevention Program Research Group. Impact of intensive lifestyle and metformin therapy on cardiovascular disease risk factors in the diabetes prevention program. Diabetes Care 2005; 28:888-894
  134. Spanheimer R, Betteridge DJ, Tan MH, Ferrannini E, Charbonnel B. Long-term lipid effects of pioglitazone by baseline anti-hyperglycemia medication therapy and statin use from the PROactive experience (PROactive 14). Am J Cardiol 2009; 104:234-239
  135. Deeg MA, Buse JB, Goldberg RB, Kendall DM, Zagar AJ, Jacober SJ, Khan MA, Perez AT, Tan MH. Pioglitazone and rosiglitazone have different effects on serum lipoprotein particle concentrations and sizes in patients with type 2 diabetes and dyslipidemia. Diabetes Care 2007; 30:2458-2464
  136. Goldberg RB, Kendall DM, Deeg MA, Buse JB, Zagar AJ, Pinaire JA, Tan MH, Khan MA, Perez AT, Jacober SJ. A comparison of lipid and glycemic effects of pioglitazone and rosiglitazone in patients with type 2 diabetes and dyslipidemia. Diabetes Care 2005; 28:1547-1554
  137. Sanchez-Garcia A, Simental-Mendia M, Millan-Alanis JM, Simental-Mendia LE. Effect of sodium-glucose co-transporter 2 inhibitors on lipid profile: A systematic review and meta-analysis of 48 randomized controlled trials. Pharmacol Res 2020; 160:105068
  138. Lamos EM, Levitt DL, Munir KM. A review of dopamine agonist therapy in type 2 diabetes and effects on cardio-metabolic parameters. Prim Care Diabetes 2016; 10:60-65
  139. Holt RI, Barnett AH, Bailey CJ. Bromocriptine: old drug, new formulation and new indication. Diabetes Obes Metab 2010; 12:1048-1057
  140. Raskin P, Cincotta AH. Bromocriptine-QR therapy for the management of type 2 diabetes mellitus: developmental basis and therapeutic profile summary. Expert Rev Endocrinol Metab 2016; 11:113-148
  141. Feingold KR. Cholesterol Lowering Drugs. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, Dungan K, Grossman A, Hershman JM, Kaltsas G, Koch C, Kopp P, Korbonits M, McLachlan R, Morley JE, New M, Perreault L, Purnell J, Rebar R, Singer F, Trence DL, Vinik A, Wilson DP, eds. Endotext. South Dartmouth (MA)2021.
  142. Nauck MA, Meier JJ, Cavender MA, Abd El Aziz M, Drucker DJ. Cardiovascular Actions and Clinical Outcomes With Glucagon-Like Peptide-1 Receptor Agonists and Dipeptidyl Peptidase-4 Inhibitors. Circulation 2017; 136:849-870
  143. Kanbay M, Copur S, Siriopol D, Yildiz AB, Gaipov A, van Raalte DH, Tuttle KR. Effect of tirzepatide on blood pressure and lipids: A meta-analysis of randomized controlled trials. Diabetes Obes Metab 2023;
  144. Ginsberg HN. Diabetic dyslipidemia: basic mechanisms underlying the common hypertriglyceridemia and low HDL cholesterol levels. Diabetes 1996; 45 Suppl 3:S27-30
  145. Ginsberg HN, Zhang YL, Hernandez-Ono A. Metabolic syndrome: focus on dyslipidemia. Obesity (Silver Spring)2006; 14 Suppl 1:41S-49S
  146. Klop B, Elte JW, Cabezas MC. Dyslipidemia in obesity: mechanisms and potential targets. Nutrients 2013; 5:1218-1240
  147. Jorgensen AB, Frikke-Schmidt R, Nordestgaard BG, Tybjaerg-Hansen A. Loss-of-function mutations in APOC3 and risk of ischemic vascular disease. N Engl J Med 2014; 371:32-41
  148. Tg,Hdl Working Group of the Exome Sequencing Project, National Heart LungBlood, Institute, Crosby J, Peloso GM, Auer PL, Crosslin DR, Stitziel NO, Lange LA, Lu Y, Tang ZZ, Zhang H, Hindy G, Masca N, Stirrups K, Kanoni S, Do R, Jun G, Hu Y, Kang HM, Xue C, Goel A, Farrall M, Duga S, Merlini PA, Asselta R, Girelli D, Olivieri O, Martinelli N, Yin W, Reilly D, Speliotes E, Fox CS, Hveem K, Holmen OL, Nikpay M, Farlow DN, Assimes TL, Franceschini N, Robinson J, North KE, Martin LW, DePristo M, Gupta N, Escher SA, Jansson JH, Van Zuydam N, Palmer CN, Wareham N, Koch W, Meitinger T, Peters A, Lieb W, Erbel R, Konig IR, Kruppa J, Degenhardt F, Gottesman O, Bottinger EP, O'Donnell CJ, Psaty BM, Ballantyne CM, Abecasis G, Ordovas JM, Melander O, Watkins H, Orho-Melander M, Ardissino D, Loos RJ, McPherson R, Willer CJ, Erdmann J, Hall AS, Samani NJ, Deloukas P, Schunkert H, Wilson JG, Kooperberg C, Rich SS, Tracy RP, Lin DY, Altshuler D, Gabriel S, Nickerson DA, Jarvik GP, Cupples LA, Reiner AP, Boerwinkle E, Kathiresan S. Loss-of-function mutations in APOC3, triglycerides, and coronary disease. N Engl J Med 2014; 371:22-31
  149. Gaudet D, Brisson D, Tremblay K, Alexander VJ, Singleton W, Hughes SG, Geary RS, Baker BF, Graham MJ, Crooke RM, Witztum JL. Targeting APOC3 in the familial chylomicronemia syndrome. N Engl J Med 2014; 371:2200-2206
  150. Arca M, D'Erasmo L, Minicocci I. Familial combined hypolipidemia: angiopoietin-like protein-3 deficiency. Curr Opin Lipidol 2020; 31:41-48
  151. Khovidhunkit W, Kim MS, Memon RA, Shigenaga JK, Moser AH, Feingold KR, Grunfeld C. Effects of infection and inflammation on lipid and lipoprotein metabolism: mechanisms and consequences to the host. J Lipid Res2004; 45:1169-1196
  152. Lara-Castro C, Fu Y, Chung BH, Garvey WT. Adiponectin and the metabolic syndrome: mechanisms mediating risk for metabolic and cardiovascular disease. Curr Opin Lipidol 2007; 18:263-270
  153. Feingold KR, Grunfeld C. The acute phase response inhibits reverse cholesterol transport. J Lipid Res 2010; 51:682-684
  154. Feingold KR, Grunfeld C. Effect of inflammation on HDL structure and function. Curr Opin Lipidol 2022; 27:521-530
  155. Lu B, Moser A, Shigenaga JK, Grunfeld C, Feingold KR. The acute phase response stimulates the expression of angiopoietin like protein 4. Biochem Biophys Res Commun 2010; 391:1737-1741
  156. Feingold KR, Grunfeld C. The Effect of Inflammation and Infection on Lipids and Lipoproteins. In: De Groot LJ, Beck-Peccoz P, Chrousos G, Dungan K, Grossman A, Hershman JM, Koch C, McLachlan R, New M, Rebar R, Singer F, Vinik A, Weickert MO, eds. Endotext. South Dartmouth (MA)2019.
  157. Rosenblat M, Karry R, Aviram M. Paraoxonase 1 (PON1) is a more potent antioxidant and stimulant of macrophage cholesterol efflux, when present in HDL than in lipoprotein-deficient serum: relevance to diabetes. Atherosclerosis 2006; 187:74-81
  158. Ahima RS. Adipose tissue as an endocrine organ. Obesity (Silver Spring) 2006; 14 Suppl 5:242S-249S
  159. Christou GA, Kiortsis DN. Adiponectin and lipoprotein metabolism. Obes Rev 2013; 14:939-949
  160. Rashid S, Kastelein JJ. PCSK9 and resistin at the crossroads of the atherogenic dyslipidemia. Expert Rev Cardiovasc Ther 2013; 11:1567-1577
  161. Yu YH, Ginsberg HN. Adipocyte signaling and lipid homeostasis: sequelae of insulin-resistant adipose tissue. Circ Res 2005; 96:1042-1052
  162. Cholesterol Treatment Trialists, Collaborato, Kearney PM, Blackwell L, Collins R, Keech A, Simes J, Peto R, Armitage J, Baigent C. Efficacy of cholesterol-lowering therapy in 18,686 people with diabetes in 14 randomised trials of statins: a meta-analysis. Lancet 2008; 371:117-125
  163. Collins R, Armitage J, Parish S, Sleigh P, Peto R, Heart Protection Study Collaborative Group. MRC/BHF Heart Protection Study of cholesterol-lowering with simvastatin in 5963 people with diabetes: a randomised placebo-controlled trial. Lancet 2003; 361:2005-2016
  164. Heart Protection Study Collaborative Group. MRC/BHF Heart Protection Study of cholesterol lowering with simvastatin in 20,536 high-risk individuals: a randomised placebo-controlled trial. Lancet 2002; 360:7-22
  165. Wanner C, Krane V, Marz W, Olschewski M, Mann JF, Ruf G, Ritz E, German, Diabetes

            Dialysis Study, Investigators. Atorvastatin in patients with type 2 diabetes mellitus undergoing hemodialysis. N Engl J Med 2005; 353:238-248

  1. Colhoun HM, Betteridge DJ, Durrington PN, Hitman GA, Neil HA, Livingstone SJ, Thomason MJ, Mackness MI, Charlton-Menys V, Fuller JH. Primary prevention of cardiovascular disease with atorvastatin in type 2 diabetes in the Collaborative Atorvastatin Diabetes Study (CARDS): multicentre randomised placebo-controlled trial. Lancet2004; 364:685-696
  2. Nissen SE, Tuzcu EM, Schoenhagen P, Brown BG, Ganz P, Vogel RA, Crowe T, Howard G, Cooper CJ, Brodie B, Grines CL, DeMaria AN. Effect of intensive compared with moderate lipid-lowering therapy on progression of coronary atherosclerosis: a randomized controlled trial. JAMA 2004; 291:1071-1080
  3. Nissen SE, Nicholls SJ, Sipahi I, Libby P, Raichlen JS, Ballantyne CM, Davignon J, Erbel R, Fruchart JC, Tardif JC, Schoenhagen P, Crowe T, Cain V, Wolski K, Goormastic M, Tuzcu EM. Effect of very high-intensity statin therapy on regression of coronary atherosclerosis: the ASTEROID trial. JAMA 2006; 295:1556-1565
  4. Stegman B, Puri R, Cho L, Shao M, Ballantyne CM, Barter PJ, Chapman MJ, Erbel R, Libby P, Raichlen JS, Uno K, Kataoka Y, Nissen SE, Nicholls SJ. High-intensity statin therapy alters the natural history of diabetic coronary atherosclerosis: insights from SATURN. Diabetes Care 2014; 37:3114-3120
  5. Ahmed S, Cannon CP, Murphy SA, Braunwald E. Acute coronary syndromes and diabetes: Is intensive lipid lowering beneficial? Results of the PROVE IT-TIMI 22 trial. Eur Heart J 2006; 27:2323-2329
  6. Cannon CP, Braunwald E, McCabe CH, Rader DJ, Rouleau JL, Belder R, Joyal SV, Hill KA, Pfeffer MA, Skene AM, Pravastatin or Atorvastatin E, Infection Therapy-Thrombolysis in Myocardial Infarction I. Intensive versus moderate lipid lowering with statins after acute coronary syndromes. N Engl J Med 2004; 350:1495-1504
  7. LaRosa JC, Grundy SM, Waters DD, Shear C, Barter P, Fruchart JC, Gotto AM, Greten H, Kastelein JJ, Shepherd J, Wenger NK, Treating to New Targets I. Intensive lipid lowering with atorvastatin in patients with stable coronary disease. N Engl J Med 2005; 352:1425-1435
  8. Shepherd J, Barter P, Carmena R, Deedwania P, Fruchart JC, Haffner S, Hsia J, Breazna A, LaRosa J, Grundy S, Waters D. Effect of lowering LDL cholesterol substantially below currently recommended levels in patients with coronary heart disease and diabetes: the Treating to New Targets (TNT) study. Diabetes Care 2006; 29:1220-1226
  9. Pedersen TR, Faergeman O, Kastelein JJ, Olsson AG, Tikkanen MJ, Holme I, Larsen ML, Bendiksen FS, Lindahl C, Szarek M, Tsai J, Incremental Decrease in End Points Through Aggressive Lipid Lowering Study Group. High-dose atorvastatin vs usual-dose simvastatin for secondary prevention after myocardial infarction: the IDEAL study: a randomized controlled trial. JAMA 2005; 294:2437-2445
  10. Cholesterol Treatment Trialists Colloboration, Baigent C, Blackwell L, Emberson J, Holland LE, Reith C, Bhala N, Peto R, Barnes EH, Keech A, Simes J, Collins R. Efficacy and safety of more intensive lowering of LDL cholesterol: a meta-analysis of data from 170,000 participants in 26 randomised trials. Lancet 2010; 376:1670-1681
  11. Keech A, Simes RJ, Barter P, Best J, Scott R, Taskinen MR, Forder P, Pillai A, Davis T, Glasziou P, Drury P, Kesaniemi YA, Sullivan D, Hunt D, Colman P, d'Emden M, Whiting M, Ehnholm C, Laakso M. Effects of long-term fenofibrate therapy on cardiovascular events in 9795 people with type 2 diabetes mellitus (the FIELD study): randomised controlled trial. Lancet 2005; 366:1849-1861
  12. Jun M, Foote C, Lv J, Neal B, Patel A, Nicholls SJ, Grobbee DE, Cass A, Chalmers J, Perkovic V. Effects of fibrates on cardiovascular outcomes: a systematic review and meta-analysis. Lancet 2010; 375:1875-1884
  13. Sacks FM, Carey VJ, Fruchart JC. Combination lipid therapy in type 2 diabetes. N Engl J Med 2010; 363:692-694; author reply 694-695
  14. Robins SJ, Collins D, Wittes JT, Papademetriou V, Deedwania PC, Schaefer EJ, McNamara JR, Kashyap ML, Hershman JM, Wexler LF, Rubins HB. Relation of gemfibrozil treatment and lipid levels with major coronary events: VA-HIT: a randomized controlled trial. JAMA 2001; 285:1585-1591
  15. Rubins HB, Robins SJ, Collins D, Nelson DB, Elam MB, Schaefer EJ, Faas FH, Anderson JW. Diabetes, plasma insulin, and cardiovascular disease: subgroup analysis from the Department of Veterans Affairs high-density lipoprotein intervention trial (VA-HIT). Arch Intern Med 2002; 162:2597-2604
  16. Koskinen P, Manttari M, Manninen V, Huttunen JK, Heinonen OP, Frick MH. Coronary heart disease incidence in NIDDM patients in the Helsinki Heart Study. Diabetes Care 1992; 15:820-825
  17. Effect of fenofibrate on progression of coronary-artery disease in type 2 diabetes: the Diabetes Atherosclerosis Intervention Study, a randomised study. Lancet 2001; 357:905-910
  18. Elkeles RS, Diamond JR, Poulter C, Dhanjil S, Nicolaides AN, Mahmood S, Richmond W, Mather H, Sharp P, Feher MD. Cardiovascular outcomes in type 2 diabetes. A double-blind placebo-controlled study of bezafibrate: the St. Mary's, Ealing, Northwick Park Diabetes Cardiovascular Disease Prevention (SENDCAP) Study. Diabetes Care 1998; 21:641-648
  19. Clofibrate and niacin in coronary heart disease. JAMA 1975; 231:360-381
  20. Canner PL, Furberg CD, Terrin ML, McGovern ME. Benefits of niacin by glycemic status in patients with healed myocardial infarction (from the Coronary Drug Project). Am J Cardiol 2005; 95:254-257
  21. Ouchi Y, Sasaki J, Arai H, Yokote K, Harada K, Katayama Y, Urabe T, Uchida Y, Hayashi M, Yokota N, Nishida H, Otonari T, Arai T, Sakuma I, Sakabe K, Yamamoto M, Kobayashi T, Oikawa S, Yamashita S, Rakugi H, Imai T, Tanaka S, Ohashi Y, Kuwabara M, Ito H. Ezetimibe Lipid-Lowering Trial on Prevention of Atherosclerotic Cardiovascular Disease in 75 or Older (EWTOPIA 75): A Randomized, Controlled Trial. Circulation 2019; 140:992-1003
  22. Nissen SE, Lincoff AM, Brennan D, Ray KK, Mason D, Kastelein JJP, Thompson PD, Libby P, Cho L, Plutzky J, Bays HE, Moriarty PM, Menon V, Grobbee DE, Louie MJ, Chen CF, Li N, Bloedon L, Robinson P, Horner M, Sasiela WJ, McCluskey J, Davey D, Fajardo-Campos P, Petrovic P, Fedacko J, Zmuda W, Lukyanov Y, Nicholls SJ. Bempedoic Acid and Cardiovascular Outcomes in Statin-Intolerant Patients. N Engl J Med 2023; 388:1353-1364
  23. Nissen SE, Menon V, Nicholls SJ, Brennan D, Laffin L, Ridker P, Ray KK, Mason D, Kastelein JJP, Cho L, Libby P, Li N, Foody J, Louie MJ, Lincoff AM. Bempedoic Acid for Primary Prevention of Cardiovascular Events in Statin-Intolerant Patients. JAMA 2023; 330:131-140
  24. Group AS, Ginsberg HN, Elam MB, Lovato LC, Crouse JR, 3rd, Leiter LA, Linz P, Friedewald WT, Buse JB, Gerstein HC, Probstfield J, Grimm RH, Ismail-Beigi F, Bigger JT, Goff DC, Jr., Cushman WC, Simons-Morton DG, Byington RP. Effects of combination lipid therapy in type 2 diabetes mellitus. N Engl J Med 2010; 362:1563-1574
  25. ACCORD Study Group, Group AES, Chew EY, Ambrosius WT, Davis MD, Danis RP, Gangaputra S, Greven CM, Hubbard L, Esser BA, Lovato JF, Perdue LH, Goff DC, Jr., Cushman WC, Ginsberg HN, Elam MB, Genuth S, Gerstein HC, Schubart U, Fine LJ. Effects of medical therapies on retinopathy progression in type 2 diabetes. N Engl J Med 2010; 363:233-244
  26. Keech AC, Mitchell P, Summanen PA, O'Day J, Davis TM, Moffitt MS, Taskinen MR, Simes RJ, Tse D, Williamson E, Merrifield A, Laatikainen LT, d'Emden MC, Crimet DC, O'Connell RL, Colman PG. Effect of fenofibrate on the need for laser treatment for diabetic retinopathy (FIELD study): a randomised controlled trial. Lancet 2007; 370:1687-1697
  27. Das Pradhan A, Glynn RJ, Fruchart JC, MacFadyen JG, Zaharris ES, Everett BM, Campbell SE, Oshima R, Amarenco P, Blom DJ, Brinton EA, Eckel RH, Elam MB, Felicio JS, Ginsberg HN, Goudev A, Ishibashi S, Joseph J, Kodama T, Koenig W, Leiter LA, Lorenzatti AJ, Mankovsky B, Marx N, Nordestgaard BG, Pall D, Ray KK, Santos RD, Soran H, Susekov A, Tendera M, Yokote K, Paynter NP, Buring JE, Libby P, Ridker PM. Triglyceride Lowering with Pemafibrate to Reduce Cardiovascular Risk. N Engl J Med 2022; 387:1923-1934
  28. Investigators AIM-HIGH, Boden WE, Probstfield JL, Anderson T, Chaitman BR, Desvignes-Nickens P, Koprowicz K, McBride R, Teo K, Weintraub W. Niacin in patients with low HDL cholesterol levels receiving intensive statin therapy. N Engl J Med 2011; 365:2255-2267
  29. Guyton JR, Slee AE, Anderson T, Fleg JL, Goldberg RB, Kashyap ML, Marcovina SM, Nash SD, O'Brien KD, Weintraub WS, Xu P, Zhao XQ, Boden WE. Relationship of lipoproteins to cardiovascular events: the AIM-HIGH Trial (Atherothrombosis Intervention in Metabolic Syndrome With Low HDL/High Triglycerides and Impact on Global Health Outcomes). J Am Coll Cardiol 2013; 62:1580-1584
  30. Hps Thrive Collaborative Group, Landray MJ, Haynes R, Hopewell JC, Parish S, Aung T, Tomson J, Wallendszus K, Craig M, Jiang L, Collins R, Armitage J. Effects of extended-release niacin with laropiprant in high-risk patients. The New England journal of medicine 2014; 371:203-212
  31. Cannon CP, Blazing MA, Giugliano RP, McCagg A, White JA, Theroux P, Darius H, Lewis BS, Ophuis TO, Jukema JW, De Ferrari GM, Ruzyllo W, De Lucca P, Im K, Bohula EA, Reist C, Wiviott SD, Tershakovec AM, Musliner TA, Braunwald E, Califf RMI. Ezetimibe Added to Statin Therapy after Acute Coronary Syndromes. N Engl J Med 2015; 372:2387-2397
  32. Bohula EA, Morrow DA, Giugliano RP, Blazing MA, He P, Park JG, Murphy SA, White JA, Kesaniemi YA, Pedersen TR, Brady AJ, Mitchel Y, Cannon CP, Braunwald E. Atherothrombotic Risk Stratification and Ezetimibe for Secondary Prevention. J Am Coll Cardiol 2017; 69:911-921
  33. Giugliano RP, Cannon CP, Blazing MA, Nicolau JC, Corbalan R, Spinar J, Park JG, White JA, Bohula EA, Braunwald E. Benefit of Adding Ezetimibe to Statin Therapy on Cardiovascular Outcomes and Safety in Patients With Versus Without Diabetes Mellitus: Results From IMPROVE-IT (Improved Reduction of Outcomes: Vytorin Efficacy International Trial). Circulation 2018; 137:1571-1582
  34. Lee YJ, Cho JY, You SC, Lee YH, Yun KH, Cho YH, Shin WY, Im SW, Kang WC, Park Y, Lee SY, Lee SJ, Hong SJ, Ahn CM, Kim BK, Ko YG, Choi D, Hong MK, Jang Y, Kim JS. Moderate-intensity statin with ezetimibe vs. high-intensity statin in patients with diabetes and atherosclerotic cardiovascular disease in the RACING trial. Eur Heart J 2023; 44:972-983
  35. Sabatine MS, Giugliano RP, Keech AC, Honarpour N, Wiviott SD, Murphy SA, Kuder JF, Wang H, Liu T, Wasserman SM, Sever PS, Pedersen TR. Evolocumab and Clinical Outcomes in Patients with Cardiovascular Disease. N Engl J Med 2017; 376:1713-1722
  36. Sabatine MS, Leiter LA, Wiviott SD, Giugliano RP, Deedwania P, De Ferrari GM, Murphy SA, Kuder JF, Gouni-Berthold I, Lewis BS, Handelsman Y, Pineda AL, Honarpour N, Keech AC, Sever PS, Pedersen TR. Cardiovascular safety and efficacy of the PCSK9 inhibitor evolocumab in patients with and without diabetes and the effect of evolocumab on glycaemia and risk of new-onset diabetes: a prespecified analysis of the FOURIER randomised controlled trial. Lancet Diabetes Endocrinol 2017;
  37. Sabatine MS, Leiter LA, Wiviott SD, Giugliano RP, Deedwania P, De Ferrari GM, Murphy SA, Kuder JF, Gouni-Berthold I, Lewis BS, Handelsman Y, Pineda AL, Honarpour N, Keech AC, Sever PS, Pedersen TR. Cardiovascular safety and efficacy of the PCSK9 inhibitor evolocumab in patients with and without diabetes and the effect of evolocumab on glycaemia and risk of new-onset diabetes: a prespecified analysis of the FOURIER randomised controlled trial. Lancet Diabetes Endocrinol 2017; 5:941-950
  38. Giugliano RP, Keech A, Murphy SA, Huber K, Tokgozoglu SL, Lewis BS, Ferreira J, Pineda AL, Somaratne R, Sever PS, Pedersen TR, Sabatine MS. Clinical Efficacy and Safety of Evolocumab in High-Risk Patients Receiving a Statin: Secondary Analysis of Patients With Low LDL Cholesterol Levels and in Those Already Receiving a Maximal-Potency Statin in a Randomized Clinical Trial. JAMA Cardiol 2017; 2:1385-1391
  39. Giugliano RP, Pedersen TR, Park JG, De Ferrari GM, Gaciong ZA, Ceska R, Toth K, Gouni-Berthold I, Lopez-Miranda J, Schiele F, Mach F, Ott BR, Kanevsky E, Pineda AL, Somaratne R, Wasserman SM, Keech AC, Sever PS, Sabatine MS. Clinical efficacy and safety of achieving very low LDL-cholesterol concentrations with the PCSK9 inhibitor evolocumab: a prespecified secondary analysis of the FOURIER trial. Lancet 2017; 390:1962-1971
  40. Schwartz GG, Steg PG, Szarek M, Bhatt DL, Bittner VA, Diaz R, Edelberg JM, Goodman SG, Hanotin C, Harrington RA, Jukema JW, Lecorps G, Mahaffey KW, Moryusef A, Pordy R, Quintero K, Roe MT, Sasiela WJ, Tamby JF, Tricoci P, White HD, Zeiher AM. Alirocumab and Cardiovascular Outcomes after Acute Coronary Syndrome. N Engl J Med 2018; 379:2097-2107
  41. Nicholls SJ, Puri R, Anderson T, Ballantyne CM, Cho L, Kastelein JJ, Koenig W, Somaratne R, Kassahun H, Yang J, Wasserman SM, Scott R, Ungi I, Podolec J, Ophuis AO, Cornel JH, Borgman M, Brennan DM, Nissen SE. Effect of Evolocumab on Progression of Coronary Disease in Statin-Treated Patients: The GLAGOV Randomized Clinical Trial. JAMA 2016; 316:2373-2384
  42. ORIGIN Trial Investigators OT, Bosch J, Gerstein HC, Dagenais GR, Diaz R, Dyal L, Jung H, Maggiono AP, Probstfield J, Ramachandran A, Riddle MC, Ryden LE, Yusuf S. n-3 fatty acids and cardiovascular outcomes in patients with dysglycemia. N Engl J Med 2012; 367:309-318
  43. Ascend Study Collaborative Group, Bowman L, Mafham M, Wallendszus K, Stevens W, Buck G, Barton J, Murphy K, Aung T, Haynes R, Cox J, Murawska A, Young A, Lay M, Chen F, Sammons E, Waters E, Adler A, Bodansky J, Farmer A, McPherson R, Neil A, Simpson D, Peto R, Baigent C, Collins R, Parish S, Armitage J. Effects of n-3 Fatty Acid Supplements in Diabetes Mellitus. N Engl J Med 2018; 379:1540-1550
  44. Feingold KR. Triglyceride Lowering Drugs. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, Dungan K, Grossman A, Hershman JM, Kaltsas G, Koch C, Kopp P, Korbonits M, McLachlan R, Morley JE, New M, Perreault L, Purnell J, Rebar R, Singer F, Trence DL, Vinik A, Wilson DP, eds. Endotext. South Dartmouth (MA)2021.
  45. Yokoyama M, Origasa H, Matsuzaki M, Matsuzawa Y, Saito Y, Ishikawa Y, Oikawa S, Sasaki J, Hishida H, Itakura H, Kita T, Kitabatake A, Nakaya N, Sakata T, Shimada K, Shirato K. Effects of eicosapentaenoic acid on major coronary events in hypercholesterolaemic patients (JELIS): a randomised open-label, blinded endpoint analysis. Lancet 2007; 369:1090-1098
  46. Saito Y, Yokoyama M, Origasa H, Matsuzaki M, Matsuzawa Y, Ishikawa Y, Oikawa S, Sasaki J, Hishida H, Itakura H, Kita T, Kitabatake A, Nakaya N, Sakata T, Shimada K, Shirato K. Effects of EPA on coronary artery disease in hypercholesterolemic patients with multiple risk factors: sub-analysis of primary prevention cases from the Japan EPA Lipid Intervention Study (JELIS). Atherosclerosis 2008; 200:135-140
  47. Bhatt DL, Steg PG, Miller M, Brinton EA, Jacobson TA, Ketchum SB, Doyle RT, Jr., Juliano RA, Jiao L, Granowitz C, Tardif JC, Ballantyne CM. Cardiovascular Risk Reduction with Icosapent Ethyl for Hypertriglyceridemia. N Engl J Med 2018;
  48. Mason RP, Libby P, Bhatt DL. Emerging Mechanisms of Cardiovascular Protection for the Omega-3 Fatty Acid Eicosapentaenoic Acid. Arterioscler Thromb Vasc Biol 2020; 40:1135-1147
  49. Nicholls SJ, Lincoff AM, Bash D, Ballantyne CM, Barter PJ, Davidson MH, Kastelein JJP, Koenig W, McGuire DK, Mozaffarian D, Pedersen TR, Ridker PM, Ray K, Karlson BW, Lundstrom T, Wolski K, Nissen SE. Assessment of omega-3 carboxylic acids in statin-treated patients with high levels of triglycerides and low levels of high-density lipoprotein cholesterol: Rationale and design of the STRENGTH trial. Clin Cardiol 2018; 41:1281-1288
  50. Nicholls SJ, Lincoff AM, Garcia M, Bash D, Ballantyne CM, Barter PJ, Davidson MH, Kastelein JJP, Koenig W, McGuire DK, Mozaffarian D, Ridker PM, Ray KK, Katona BG, Himmelmann A, Loss LE, Rensfeldt M, Lundstrom T, Agrawal R, Menon V, Wolski K, Nissen SE. Effect of High-Dose Omega-3 Fatty Acids vs Corn Oil on Major Adverse Cardiovascular Events in Patients at High Cardiovascular Risk: The STRENGTH Randomized Clinical Trial. JAMA 2020;
  51. Ridker PM, Rifai N, MacFadyen J, Glynn RJ, Jiao L, Steg PG, Miller M, Brinton EA, Jacobson TA, Tardif JC, Ballantyne CM, Mason RP, Bhatt DL. Effects of Randomized Treatment With Icosapent Ethyl and a Mineral Oil Comparator on Interleukin-1beta, Interleukin-6, C-Reactive Protein, Oxidized Low-Density Lipoprotein Cholesterol, Homocysteine, Lipoprotein(a), and Lipoprotein-Associated Phospholipase A2: A REDUCE-IT Biomarker Substudy. Circulation 2022; 146:372-379
  52. Goff ZD, Nissen SE. N-3 polyunsaturated fatty acids for cardiovascular risk. Curr Opin Cardiol 2022; 37:356-363
  53. Mason RP, Sherratt SCR, Eckel RH. Omega-3-fatty acids: Do they prevent cardiovascular disease? Best Pract Res Clin Endocrinol Metab 2023; 37:101681
  54. ElSayed NA, Aleppo G, Aroda VR, Bannuru RR, Brown FM, Bruemmer D, Collins BS, Das SR, Hilliard ME, Isaacs D, Johnson EL, Kahan S, Khunti K, Kosiborod M, Leon J, Lyons SK, Perry ML, Prahalad P, Pratley RE, Seley JJ, Stanton RC, Gabbay RA, on behalf of the American Diabetes A. 10. Cardiovascular Disease and Risk Management: Standards of Care in Diabetes-2023. Diabetes Care 2023; 46:S158-S190
  55. Grundy SM, Stone NJ, Bailey AL, Beam C, Birtcher KK, Blumenthal RS, Braun LT, de Ferranti S, Faiella-Tommasino J, Forman DE, Goldberg R, Heidenreich PA, Hlatky MA, Jones DW, Lloyd-Jones D, Lopez-Pajares N, Ndumele CE, Orringer CE, Peralta CA, Saseen JJ, Smith SC, Jr., Sperling L, Virani SS, Yeboah J. 2018 AHA/ACC/AACVPR/AAPA/ABC/ACPM/ADA/AGS/APhA/ASPC/NLA/PCNA Guideline on the Management of Blood Cholesterol: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. Circulation 2019; 139:e1082-e1143
  56. Jellinger PS, Handelsman Y, Rosenblit PD, Bloomgarden ZT, Fonseca VA, Garber AJ, Grunberger G, Guerin CK, Bell DSH, Mechanick JI, Pessah-Pollack R, Wyne K, Smith D, Brinton EA, Fazio S, Davidson M. American Association of Clinical Endocrinologists and American College of Endocrinology Guidelines for Management of Dyslipidemia and Prevention of Cardiovascular Disease. Endocr Pract 2017; 23:1-87
  57. Handelsman Y, Jellinger PS, Guerin CK, Bloomgarden ZT, Brinton EA, Budoff MJ, Davidson MH, Einhorn D, Fazio S, Fonseca VA, Garber AJ, Grunberger G, Krauss RM, Mechanick JI, Rosenblit PD, Smith DA, Wyne KL. Consensus Statement by the American Association of Clinical Endocrinologists and American College of Endocrinology on the Management of Dyslipidemia and Prevention of Cardiovascular Disease Algorithm - 2020 Executive Summary. Endocr Pract 2020; 26:1196-1224
  58. Mach F, Baigent C, Catapano AL, Koskinas KC, Casula M, Badimon L, Chapman MJ, De Backer GG, Delgado V, Ference BA, Graham IM, Halliday A, Landmesser U, Mihaylova B, Pedersen TR, Riccardi G, Richter DJ, Sabatine MS, Taskinen MR, Tokgozoglu L, Wiklund O. 2019 ESC/EAS Guidelines for the management of dyslipidaemias: lipid modification to reduce cardiovascular risk. Eur Heart J 2020; 41:111-188
  59. Marx N, Federici M, Schutt K, Muller-Wieland D, Ajjan RA, Antunes MJ, Christodorescu RM, Crawford C, Di Angelantonio E, Eliasson B, Espinola-Klein C, Fauchier L, Halle M, Herrington WG, Kautzky-Willer A, Lambrinou E, Lesiak M, Lettino M, McGuire DK, Mullens W, Rocca B, Sattar N. 2023 ESC Guidelines for the management of cardiovascular disease in patients with diabetes. Eur Heart J 2023;
  60. SCORE2-Diabetes Working Group. SCORE2-Diabetes: 10-year cardiovascular risk estimation in type 2 diabetes in Europe. Eur Heart J 2023; 44:2544-2556
  61. Feingold KR. Maximizing the benefits of cholesterol-lowering drugs. Curr Opin Lipidol 2019; 30:388-394
  62. Feingold KR, Chait A. Approach to patients with elevated low-density lipoprotein cholesterol levels. Best Pract Res Clin Endocrinol Metab 2023; 37:101658
  63. Evert AB, Dennison M, Gardner CD, Garvey WT, Lau KHK, MacLeod J, Mitri J, Pereira RF, Rawlings K, Robinson S, Saslow L, Uelmen S, Urbanski PB, Yancy WS, Jr. Nutrition Therapy for Adults With Diabetes or Prediabetes: A Consensus Report. Diabetes Care 2019; 42:731-754
  64. Feingold KR. The Effect of Diet on Cardiovascular Disease and Lipid and Lipoprotein Levels. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Shah AS, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA)2021.
  65. Look ARG, Wing RR, Bolin P, Brancati FL, Bray GA, Clark JM, Coday M, Crow RS, Curtis JM, Egan CM, Espeland MA, Evans M, Foreyt JP, Ghazarian S, Gregg EW, Harrison B, Hazuda HP, Hill JO, Horton ES, Hubbard VS, Jakicic JM, Jeffery RW, Johnson KC, Kahn SE, Kitabchi AE, Knowler WC, Lewis CE, Maschak-Carey BJ, Montez MG, Murillo A, Nathan DM, Patricio J, Peters A, Pi-Sunyer X, Pownall H, Reboussin D, Regensteiner JG, Rickman AD, Ryan DH, Safford M, Wadden TA, Wagenknecht LE, West DS, Williamson DF, Yanovski SZ. Cardiovascular effects of intensive lifestyle intervention in type 2 diabetes. N Engl J Med 2013; 369:145-154
  66. Estruch R, Ros E, Salas-Salvado J, Covas MI, Corella D, Aros F, Gomez-Gracia E, Ruiz-Gutierrez V, Fiol M, Lapetra J, Lamuela-Raventos RM, Serra-Majem L, Pinto X, Basora J, Munoz MA, Sorli JV, Martinez JA, Martinez-Gonzalez MA. Primary prevention of cardiovascular disease with a Mediterranean diet. N Engl J Med2013; 368:1279-1290
  67. Estruch R, Ros E, Salas-Salvado J, Covas MI, Corella D, Aros F, Gomez-Gracia E, Ruiz-Gutierrez V, Fiol M, Lapetra J, Lamuela-Raventos RM, Serra-Majem L, Pinto X, Basora J, Munoz MA, Sorli JV, Martinez JA, Fito M, Gea A, Hernan MA, Martinez-Gonzalez MA. Primary Prevention of Cardiovascular Disease with a Mediterranean Diet Supplemented with Extra-Virgin Olive Oil or Nuts. N Engl J Med 2018; 378:e34
  68. Estruch R, Martinez-Gonzalez MA, Corella D, Salas-Salvado J, Ruiz-Gutierrez V, Covas MI, Fiol M, Gomez-Gracia E, Lopez-Sabater MC, Vinyoles E, Aros F, Conde M, Lahoz C, Lapetra J, Saez G, Ros E. Effects of a Mediterranean-style diet on cardiovascular risk factors: a randomized trial. Ann Intern Med 2006; 145:1-11
  69. Delgado-Lista J, Alcala-Diaz JF, Torres-Pena JD, Quintana-Navarro GM, Fuentes F, Garcia-Rios A, Ortiz-Morales AM, Gonzalez-Requero AI, Perez-Caballero AI, Yubero-Serrano EM, Rangel-Zuniga OA, Camargo A, Rodriguez-Cantalejo F, Lopez-Segura F, Badimon L, Ordovas JM, Perez-Jimenez F, Perez-Martinez P, Lopez-Miranda J. Long-term secondary prevention of cardiovascular disease with a Mediterranean diet and a low-fat diet (CORDIOPREV): a randomised controlled trial. Lancet 2022; 399:1876-1885
  70. de Lorgeril M, Renaud S, Mamelle N, Salen P, Martin JL, Monjaud I, Guidollet J, Touboul P, Delaye J. Mediterranean alpha-linolenic acid-rich diet in secondary prevention of coronary heart disease. Lancet 1994; 343:1454-1459
  71. de Lorgeril M, Salen P, Martin JL, Monjaud I, Delaye J, Mamelle N. Mediterranean diet, traditional risk factors, and the rate of cardiovascular complications after myocardial infarction: final report of the Lyon Diet Heart Study. Circulation 1999; 99:779-785
  72. Adams TD, Gress RE, Smith SC, Halverson RC, Simper SC, Rosamond WD, Lamonte MJ, Stroup AM, Hunt SC. Long-term mortality after gastric bypass surgery. N Engl J Med 2007; 357:753-761
  73. Romeo S, Maglio C, Burza MA, Pirazzi C, Sjoholm K, Jacobson P, Svensson PA, Peltonen M, Sjostrom L, Carlsson LM. Cardiovascular events after bariatric surgery in obese subjects with type 2 diabetes. Diabetes Care 2012; 35:2613-2617
  74. Sjostrom L, Peltonen M, Jacobson P, Sjostrom CD, Karason K, Wedel H, Ahlin S, Anveden A, Bengtsson C, Bergmark G, Bouchard C, Carlsson B, Dahlgren S, Karlsson J, Lindroos AK, Lonroth H, Narbro K, Naslund I, Olbers T, Svensson PA, Carlsson LM. Bariatric surgery and long-term cardiovascular events. JAMA 2012; 307:56-65
  75. Sheng B, Truong K, Spitler H, Zhang L, Tong X, Chen L. The Long-Term Effects of Bariatric Surgery on Type 2 Diabetes Remission, Microvascular and Macrovascular Complications, and Mortality: a Systematic Review and Meta-Analysis. Obes Surg 2017; 27:2724-2732
  76. Fisher DP, Johnson E, Haneuse S, Arterburn D, Coleman KJ, O'Connor PJ, O'Brien R, Bogart A, Theis MK, Anau J, Schroeder EB, Sidney S. Association Between Bariatric Surgery and Macrovascular Disease Outcomes in Patients With Type 2 Diabetes and Severe Obesity. JAMA 2018; 320:1570-1582
  77. Farmer A, Montori V, Dinneen S, Clar C. Fish oil in people with type 2 diabetes mellitus. Cochrane Database Syst Rev 2001:CD003205
  78. Preiss D, Sattar N. Statins and the risk of new-onset diabetes: a review of recent evidence. Curr Opin Lipidol2011; 22:460-466
  79. Sattar N, Preiss D, Murray HM, Welsh P, Buckley BM, de Craen AJ, Seshasai SR, McMurray JJ, Freeman DJ, Jukema JW, Macfarlane PW, Packard CJ, Stott DJ, Westendorp RG, Shepherd J, Davis BR, Pressel SL, Marchioli R, Marfisi RM, Maggioni AP, Tavazzi L, Tognoni G, Kjekshus J, Pedersen TR, Cook TJ, Gotto AM, Clearfield MB, Downs JR, Nakamura H, Ohashi Y, Mizuno K, Ray KK, Ford I. Statins and risk of incident diabetes: a collaborative meta-analysis of randomised statin trials. Lancet 2010; 375:735-742
  80. Erqou S, Lee CC, Adler AI. Statins and glycaemic control in individuals with diabetes: a systematic review and meta-analysis. Diabetologia 2014; 57:2444-2452
  81. Livingstone SJ, Looker HC, Akbar T, Betteridge DJ, Durrington PN, Hitman GA, Neil HA, Fuller JH, Colhoun HM. Effect of atorvastatin on glycaemia progression in patients with diabetes: an analysis from the Collaborative Atorvastatin in Diabetes Trial (CARDS). Diabetologia 2016; 59:299-306
  82. Chiu SW, Pratt CM, Feinn R, Chatterjee S. Proprotein Convertase Subtilisin/Kexin Type 9 Inhibitors and Ezetimibe on Risk of New-Onset Diabetes: A Systematic Review and Meta-Analysis of Large, Double-Blinded Randomized Controlled Trials. J Cardiovasc Pharmacol Ther 2020; 25:409-417
  83. Shah NP, McGuire DK, Cannon CP, Giugliano RP, Lokhnygina Y, Page CB, Tershakovec AM, Braunwald E, Blazing MA. Impact of Ezetimibe on New-Onset Diabetes: A Substudy of IMPROVE-IT. J Am Heart Assoc 2023; 12:e029593
  84. Kellick KA, Bottorff M, Toth PP, The National Lipid Association's Safety Task F. A clinician's guide to statin drug-drug interactions. J Clin Lipidol 2014; 8:S30-46
  85. Linz PE, Lovato LC, Byington RP, O'Connor PJ, Leiter LA, Weiss D, Force RW, Crouse JR, Ismail-Beigi F, Simmons DL, Papademetriou V, Ginsberg HN, Elam MB. Paradoxical reduction in HDL-C with fenofibrate and thiazolidinedione therapy in type 2 diabetes: the ACCORD Lipid Trial. Diabetes Care 2014; 37:686-693
  86. Ansquer JC, Foucher C, Rattier S, Taskinen MR, Steiner G. Fenofibrate reduces progression to microalbuminuria over 3 years in a placebo-controlled study in type 2 diabetes: results from the Diabetes Atherosclerosis Intervention Study (DAIS). Am J Kidney Dis 2005; 45:485-493
  87. Davis TM, Ting R, Best JD, Donoghoe MW, Drury PL, Sullivan DR, Jenkins AJ, O'Connell RL, Whiting MJ, Glasziou PP, Simes RJ, Kesaniemi YA, Gebski VJ, Scott RS, Keech AC, Event Lowering in Diabetes Study i. Effects of fenofibrate on renal function in patients with type 2 diabetes mellitus: the Fenofibrate Intervention and Event Lowering in Diabetes (FIELD) Study. Diabetologia 2011; 54:280-290
  88. Rajamani K, Colman PG, Li LP, Best JD, Voysey M, D'Emden MC, Laakso M, Baker JR, Keech AC. Effect of fenofibrate on amputation events in people with type 2 diabetes mellitus (FIELD study): a prespecified analysis of a randomised controlled trial. Lancet 2009; 373:1780-1788
  89. Aldridge MA, Ito MK. Colesevelam hydrochloride: a novel bile acid-binding resin. Ann Pharmacother 2001; 35:898-907
  90. Bays HE. Colesevelam hydrochloride added to background metformin therapy in patients with type 2 diabetes mellitus: a pooled analysis from 3 clinical studies. Endocr Pract 2011; 17:933-938
  91. Song WL, FitzGerald GA. Niacin, an old drug with a new twist. J Lipid Res 2013; 54:2586-2594
  92. Elam MB, Hunninghake DB, Davis KB, Garg R, Johnson C, Egan D, Kostis JB, Sheps DS, Brinton EA. Effect of niacin on lipid and lipoprotein levels and glycemic control in patients with diabetes and peripheral arterial disease: the ADMIT study: A randomized trial. Arterial Disease Multiple Intervention Trial. JAMA 2000; 284:1263-1270
  93. Grundy SM, Vega GL, McGovern ME, Tulloch BR, Kendall DM, Fitz-Patrick D, Ganda OP, Rosenson RS, Buse JB, Robertson DD, Sheehan JP, Diabetes Multicenter Research Group. Efficacy, safety, and tolerability of once-daily niacin for the treatment of dyslipidemia associated with type 2 diabetes: results of the assessment of diabetes control and evaluation of the efficacy of niaspan trial. Archives of internal medicine 2002; 162:1568-1576
  94. Weintraub H. Update on marine omega-3 fatty acids: management of dyslipidemia and current omega-3 treatment options. Atherosclerosis 2013; 230:381-389
  95. Khan SU, Rahman H, Okunrintemi V, Riaz H, Khan MS, Sattur S, Kaluski E, Lincoff AM, Martin SS, Blaha MJ. Association of Lowering Low-Density Lipoprotein Cholesterol With Contemporary Lipid-Lowering Therapies and Risk of Diabetes Mellitus: A Systematic Review and Meta-Analysis. J Am Heart Assoc 2019; 8:e011581
  96. Carugo S, Sirtori CR, Corsini A, Tokgozoglu L, Ruscica M. PCSK9 Inhibition and Risk of Diabetes: Should We Worry? Curr Atheroscler Rep 2022; 24:995-1004
  97. Wang X, Zhang Y, Tan H, Wang P, Zha X, Chong W, Zhou L, Fang F. Efficacy and safety of bempedoic acid for prevention of cardiovascular events and diabetes: a systematic review and meta-analysis. Cardiovasc Diabetol2020; 19:128
  98. Bays HE, Banach M, Catapano AL, Duell PB, Gotto AM, Jr., Laufs U, Leiter LA, Mancini GBJ, Ray KK, Bloedon LT, Sasiela WJ, Ye Z, Ballantyne CM. Bempedoic acid safety analysis: Pooled data from four phase 3 clinical trials. J Clin Lipidol 2020; 14:649-659 e646
  99. Jenkins DJ, Jones PJ, Lamarche B, Kendall CW, Faulkner D, Cermakova L, Gigleux I, Ramprasath V, de Souza R, Ireland C, Patel D, Srichaikul K, Abdulnour S, Bashyam B, Collier C, Hoshizaki S, Josse RG, Leiter LA, Connelly PW, Frohlich J. Effect of a dietary portfolio of cholesterol-lowering foods given at 2 levels of intensity of dietary advice on serum lipids in hyperlipidemia: a randomized controlled trial. JAMA 2011; 306:831-839
  100. Feingold KR. Utility of Advanced Lipoprotein Testing in Clinical Practice. In: De Groot LJ, Beck-Peccoz P, Chrousos G, Dungan K, Grossman A, Hershman JM, Koch C, McLachlan R, New M, Rebar R, Singer F, Vinik A, Weickert MO, eds. Endotext. South Dartmouth (MA)2023.
  101. Orringer CE, Jacobson TA, Maki KC. National Lipid Association Scientific Statement on the use of icosapent ethyl in statin-treated patients with elevated triglycerides and high or very-high ASCVD risk. J Clin Lipidol 2019; 13:860-872

 

 

 

Role Of Glucose And Lipids In The Atherosclerotic Cardiovascular Disease In Patients With Diabetes

ABSTRACT

Atherosclerotic cardiovascular disease (ASCVD) is a major cause of morbidity and mortality in both men and women with T1DM and T2DM. In patients with T1DM, intensive glycemic control results in a reduction in ASCVD. However, intensive glycemic control does not have a major impact in reducing ASCVD in patients with T2DM. Metformin, pioglitazone, SGLT2 inhibitors, and certain GLP-1 receptor agonists have been shown to decrease major cardiovascular events in patients with T2DM to a greater extent than other treatment modalities. In patients with T2DM other risk factors including, hypertension and dyslipidemia, play a major role in inducing ASCVD and control of these risk factors is paramount. In patients with T1DM in good glycemic control, the lipid profile is very similar to the general population. In contrast, in patients with T2DM, even with good glycemic control, there are frequently lipid abnormalities (elevated TG and non-HDL-C, decreased HDL-C, and an increase in small dense LDL). In both T1DM and T2DM, poor glycemic control increases TG levels and decreases HDL-C levels with modest effects on LDL-C levels.  Extensive studies have demonstrated that statins decrease ASCVD in patients with diabetes. Treatment with high doses of potent statins reduces ASCVD events to a greater extent than low dose statin therapy. Adding fibrates or niacin to statin therapy has not been shown to further decrease ASCVD events. In contrast, studies have shown that the combination of a statin and ezetimibe or a statin and a PCSK9 inhibitor result in a greater decrease in ASCVD events than statins alone. Studies have suggested that EPA, an omega-3-fatty acid, when added to statins also reduces ASCVD events but this result is controversial. In statin intolerant patients with T2DM bempedoic acid decreases ASCVD events. Current recommendations state that most patients with diabetes should be on statin therapy. In certain patients with diabetes ezetimibe, PCSK9 inhibitors, and bempedoic acid can play a role in reducing ASCVD.  

 

INTRODUCTION

 

Atherosclerotic cardiovascular disease (ASCVD) is a major cause of morbidity and mortality in both men and women with diabetes (1-5). In addition to coronary disease, ASCVD includes stroke and peripheral arterial disease (PAD).  PAD is common in diabetes, may be the first presentation of ASCVD and should be recognized as needing aggressive treatment of risk factors. The risk of ASCVD is increased approximately 2-fold in men and 3-4-fold in women (2-4,6,7). In the Framingham study, the annual rate of ASCVD was similar in men and women with diabetes, emphasizing that woman with diabetes need as aggressive preventive treatment as men with diabetes (2,6). In addition, several but not all studies, have shown that patients with diabetes with no history of ASCVD have a similar risk of having a myocardial infarction as non-diabetic patients who have a history of ASCVD, i.e., diabetes is an equivalent risk factor as a history of a previous cardiovascular event (8,9). The duration of diabetes and the presence of other risk factors or complications of diabetes likely determine whether a patient with diabetes has a risk equivalent to patients with a history of previous ASCVD events (10,11). In one study patients with T2DM who had the following risk factors within the target range, HbA1c, LDL-C, albuminuria, smoking, and blood pressure, the risk of an acute myocardial infarction or stroke was similar to individuals without diabetics (12). Moreover, numerous studies have shown that patients with diabetes who have ASCVD are at a very high risk of having another event, indicating that this population of patient’s needs especially aggressive preventive measures (1,8). This increased risk for the development of ASCVD in patients with diabetes is seen both in populations where the prevalence of ASCVD is high (Western societies) and low (for example, Japan) (2). However, in societies where the prevalence of ASCVD is low, the contribution of ASCVD as a cause of morbidity and mortality in patients with diabetes is relatively low compared to Western societies (2).

 

While the database is not as robust, the evidence indicates that patients with T1DM are also at high risk for the development of ASCVD (1,13-15). Interestingly, women with T1DM have twice the excess risk of fatal and nonfatal vascular events compared to men with T1DM (16,17). Additionally, developing T1DM at a young age increases the risk of ASCVD to a greater degree than late onset T1DM (17). Approximately 50% of patients with T1DM are obese or overweight and between 8% and 40% meet the criteria for the metabolic syndrome, which increases their risk of developing ASCVD (18).

 

While the development of diabetes at a young age increases the risk of ASCVD in patients with both T1DM and T2DM the deleterious impact is greater in patients with T2DM (19). Lastly, in patients with both T1DM and T2DM the presence of renal disease increases the risk of ASCVD (4,14). Of note is that the risk of developing ASCVD events in patients with diabetes has decreased recently, most likely due to better lipid and blood pressure control, which again reinforces the need to aggressively treat these risk factors in patients with diabetes (5,7,20). 

 

ROLE OF GLYCEMIC CONTROL

 

Epidemiological studies have shown an association between the level of glycemic control and the development of ASCVD in both T1DM and T2DM (1,4,5,21,22). However, the association of glycemic control with ASCVD is considerably weaker than the association of glycemic control with the microvascular complications of diabetes, such as retinopathy and nephropathy (4). It must be recognized that epidemiological studies can only demonstrate associations and that confounding variables could account for the association between poor glycemic control and ASCVD. For example, patients with poor glycemic control may not undertake other preventive measures that could reduce ASCVD such as exercise, healthy diet, etc. Furthermore, the patients with poor glycemic control may have less compliance with therapies that reduce lipids and blood pressure. Therefore, randomized studies are essential in determining the role of glycemic control on ASCVD. 

 

Early randomized studies, such as the University Group Diabetes Project (UGDP) and VA cooperative study, did not demonstrate a reduction in cardiovascular events in patients who were aggressively treated for glucose control (23-25). In fact, the data from these early studies suggested that improvements in glycemic control (VA cooperative study) or the use of certain drugs to treat diabetes (oral sulfonylureas in UGDP) may actually increase the risk of ASCVD.

 

Diabetes Control and Complications Trial (DCCT) and Kumamoto Studies

 

Latter studies, the DCCT in patients with T1DM and the Kumamoto study in patients with T2DM, while finding a decrease in ASCVD events (DCCT 41% decrease) in the subjects randomized to improved glycemic control did not have enough ASCVD events to demonstrate a statistically significant reduction (DCCT studied a population at low risk for ASCVD and the Kumamoto study had a very small number of subjects) (26-28). In contrast, both the DCCT and the Kumamoto study clearly demonstrated that improvements in glycemic control resulted in a reduction in microvascular disease (26-28). However, the long- term follow-up of the DCCT has demonstrated that those in the intensive glycemic control group had a decrease in ASCVD in subsequent years (29,30). The initial DCCT compared intensive vs. conventional therapy for a mean of 6.5 years. At the end of the study, a very large proportion of subjects agreed to participate in a follow-up observational study (Epidemiology of Diabetes Interventions and Complications- EDIC). During this follow-up period, glycemic control was relatively similar between the intensive therapy and conventional therapy group (glycosylated hemoglobin 7.9% vs. 7.8%) but during the actual trial there was a large difference in glycosylated hemoglobin levels (7.4% vs. 9.1%). After a mean 17 years of observation, the risk of any cardiovascular event was reduced by 42% and the risk of nonfatal myocardial infarction (MI), stroke, or death from ASCVD was reduced by 57% in the intensive control group. This study demonstrates that being in the intensive glycemic control group (for 6.5 of the 17 years of observation) is sufficient to have long-term beneficial effects on the risk of developing ASCVD in patients with T1DM. This beneficial effect was not entirely due to the prevention of microvascular complications as the differences between the intensive and conventional treatment groups for ASCVD persisted after adjusting for microalbuminuria and albuminuria. When an outcome of improved glycemic control is seen, or persists for years after the trial is over the phenomenon is called a “metabolic memory” effect.

 

UK Prospective Diabetes Study (UKPDS)

 

A similar finding has been reported with regard to T2DM. The UKPDS studied a large number of newly diagnosed patients with T2DM at risk for ASCVD. In this study improved glycemic control, with either insulin or sulfonylureas, reduced ASCVD by 16%, which just missed being statistically significant (p=0.052) (31). In the UKPDS, the improvement in glycemic control was modest (HbA1c reduced by approximately 0.9%) and the 16% reduction in ASCVD was in the range predicted based on epidemiological studies. The results of a 10-year follow-up of the UKPDS study have been reported (total duration of observation 25 years) (32). After termination of the study, glycosylated hemoglobin levels became very similar between the control and treatment groups. Nevertheless, risk reductions for MI became statistically significant for the insulin and the sulfonylurea group compared to controls (15% decrease, p=0.01).

 

DiGami Studies

 

Similarly, the DiGami study, which used insulin infusion during the peri-MI period to improve glycemic control followed by long-term glycemic control, demonstrated that survival post MI was significantly improved by good glycemic control (33). While this study focused on a highly-selected population and time period (patients undergoing a MI), the results are consistent with the hypothesis that improvements in glycemic control will reduce ASCVD. However, the DiGami 2 study did not confirm the benefits of tight glucose control beginning in the peri-MI period on outcomes (34). It must be noted though that the differences in glucose control achieved in DiGami 2 were much smaller than planned and the number of patients recruited was less than anticipated. Together these deficiencies could account for the failure to demonstrate significant differences in ASCVD events in this study.

 

ACCORD Study

 

Because of the need for more definitive data on the effect of glycemic control on ASCVD in T2DM, three large randomized trials, the ACCORD, ADVANCE, and VA Diabetes Trial, have been carried out. Much to everyone’s surprise and disappointment, improvement in glycemic control did not clearly result in a significant reduction in ASCVD in these trials.

 

The ACCORD study randomized 10,251 subjects with T2DM in the US and Canada with either a history of ASCVD or at increased risk for the development of ASCVD (35). Multiple different treatment protocols were used with the goal of achieving an A1c level < 6% in the intensive group and between 7-7.9% in the standard glycemic control group. During the trial the A1c levels were 6.4% in the intensive group and 7.5% in the standard group. As expected, the use of insulin therapy was much greater in the intensive group, as was the occurrence of hypoglycemia and weight gain. After a mean duration of 3.5 years this study was stopped early by the data safety monitoring board due to an increased all-cause mortality in the intensive treatment group (1.41% vs. 1.14% per year; hazard ratio 1.22 CI 1.01- 1.46). The primary outcome (MI, stroke, ASCVD death) was reduced by 10% in the intensive control group but this was not statistically significant (p=0.16). Of note, intensive glycemic control reduced the incidence of any MI (i.e. fatal or non-fatal) by 16%, nonfatal MI by 19%, coronary revascularization by 16%, and unstable angina by 19% (36).The explanation for the increased death rate in the intensive treatment arm remains unknown, but it has been speculated that the increased deaths might have been due to hypoglycemia, weight gain, too rapidly lowering A1c levels, or unrecognized drug toxicity. Long term follow-up of the ACCORD study did not reveal any beneficial effects on the primary outcome (nonfatal MI, nonfatal stroke, or cardiovascular death), death from any cause, and an expanded composite outcome that included all-cause death in the intensive glycemic control group (37).

 

ADVANCE Study

 

The ADVANCE study randomized 11,140 subjects with T2DM in Europe, Asia, Australia/New Zealand, and Canada who either had ASCVD or at least one other risk factor for ASCVD (38). In the intensive group the goal A1c was <6.5%. The achieved A1c levels during the trial were 6.3% in the intensive group and 7.3% in the standard treatment group. Of note is that compared to the ACCORD study, less insulin use was required to achieve these A1c levels. With regard to macrovascular disease (MI, stroke, and cardiovascular death), no significant differences were observed between the intensive and standard treatment groups (HR 0.94, CI 0.84-1.06, p=0.32). In contrast to the ACCORD trial, no increase in overall or cardiovascular mortality in the intensive treatment group was observed in the ADVANCE study. Long term follow-up did not demonstrate a decrease in the risk of death from any cause or major macrovascular events between the intensive-glucose-control group and the standard-glucose-control group (39).

 

VA Diabetes Trial

 

The VA Diabetes Trial randomized 1,791 subjects with poor glycemic control on maximal oral agent therapy or insulin (entry A1c 9.4%) (40). In the intensive group, the goal A1c was <6.0%. The achieved A1c levels during the trial were 6.9% in the intensive group and 8.5% in the standard treatment group. Similar to the other trials, a significant reduction in ASCVD was not observed in the intensive glycemic control group (HR 0.88, CI 0.74-1.05, p=0.12). Notably there were more ASCVD deaths and sudden deaths in the intensive treatment group, but this increase was not statistically significant. With long-term follow-up (approximately 10 years), the intensive-therapy group had a significantly lower risk of MI, stroke, congestive heart failure, amputation for ischemic gangrene, or cardiovascular-related death than did the standard-therapy group (hazard ratio, 0.83; P=0.04 (41). However, there was no reduction in cardiovascular or total mortality. Furthermore, with a longer period of follow-up (15 years) the risks of major cardiovascular events or death were not lower in the intensive-therapy group than in the standard-therapy group (42). In a careful analysis it was noted that that the risk of cardiovascular events was 17% lower in the intensive treatment group than in the standard control group during the approximate 10-year period when there was a separation of the glycated hemoglobin curves between the two groups, suggesting that glycemic control was reducing the risk of cardiovascular events (42).

 

Meta-analyses

 

In a meta-analysis of 6 randomized studies (UKPDS, Kumamoto, VA Feasibility study, ACCORD, ADVANCE, and VA Diabetes Trial) of intensive vs. conventional glycemic control in patients with T2DM (27,654 patients) there was no significant effect of tight blood glucose control on all-cause mortality (RR 1.03; 95% CI 0.90-1.17), cardiovascular mortality (RR 1.04; 95% CI 0.83-1.29), or nonfatal stroke (RR 1.02; 95% CI 0.88-1.17) but tight glucose control reduced the risk for nonfatal MI (RR 0.85; 95% CI 0.76-0.95) (43). In a meta-analysis of 4 studies (UKPDS, ACCORD, ADVANCE, and VA Diabetes Trial) the primary outcome was the composite of death from cardiovascular causes (including sudden death), non-fatal MI and non-fatal stroke, which was decreased by 9% (HR 0.91, 95% CI 0.84–0.99) in the intensive control group (44). Of note the risk of non-fatal/fatal MI was reduced by 15% (HR 0.85, 95% CI 0.76–0.94) in the intensive group without significant reductions in the risk of non-fatal/fatal stroke, fatal heart failure, all-cause mortality, or cardiovascular death.  

 

Limitations of Cardiovascular Outcome Studies

 

Thus, while the epidemiological data strongly suggests that glycemic control would favorably impact ASCVD the recent randomized trials that were designed specifically to prove this hypothesis have failed to definitively demonstrate a clear link. There are several explanations for why these trials may not have worked as planned.

 

First, in the ACCORD, ADVANCE, and VA Diabetes Trial, other ASCVD risk factors were aggressively treated (lipid and BP lowering, ASA therapy). As a result of these treatments, the actual number of ASCVD events was considerably less than expected in these trials. The lower event rate may have reduced the ability to see a beneficial effect of glucose control. Additionally, the beneficial effects of glucose control maybe more robust if other risk factors are not aggressively controlled. In this regard, it is worth noting that in the earlier UKPDS, which showed that improved glycemic control reduced ASCVD events, both BP and lipids were not aggressively treated by current standards (systolic BP 135-140mm Hg, LDL-C 135-142mg/dL), which could be why this older trial demonstrated a benefit of improving glycemic control on ASCVD.

 

Second, these three recent trials were comparing relatively low A1c levels in both the intensive and usual control groups (A1c in intensive from ~6.4-6.9% and usual control group from ~7.0-8.4%). It is likely that both levels are on the “flatter” portion of the glycemic control-cardiovascular risk curve and that if one compared patients with intensive glycemic control with a control group with higher A1c values one would see more impressive results. If the difference in A1c levels were greater in the intensive and control groups the likelihood of seeing a reduction in cardiovascular events in the intensive group would be enhanced.

 

Third, all three trials were carried out by initiating tight control in patients with long standing diabetes who either had pre-existing ASCVD or were at high risk for ASCVD. It is possible that patients with a different clinical profile would be more likely to benefit from intensive glucose control. Subgroup analysis from these trials have suggested that patients with a shorter duration of diabetes, less severe diabetes, or the absence of pre-existing ASCVD actually benefited from intensive control. It may be that glycemic control is most important prior to the development of significant atherosclerosis. Clearly additional studies on different types of patients (i.e., newly diagnosed without evidence of ASCVD) will be necessary to definitively determine the role of glycemic control in different diabetic populations.

 

Fourth, the duration of these studies was relatively short and it is possible that a much longer duration of glycemic control is required to show benefits on ASCVD. In the UKPDS study the beneficial effects of intensive glucose control was not statistically significant at the end of the study but with an extended duration of follow-up (15-25 years) became statistically significant.

 

Fifth, it may be that glycemic control will be more important in patients with T1DM where abnormalities in glucose metabolism are a major reason for the increased risk of ASCVD. In contrast, patients with T2DM have multiple risk factors for ASCVD (dyslipidemia, hypertension, inflammation, insulin resistance, coagulation disorders, etc.) and glucose may play only a minor role in the increased risk. The differences in other cardiovascular risk factors could account for why intensive glycemic control produced a marked reduction in ASCVD in the DCCT (T1DM trial) and had only minimal effects in the trials carried out in patients with T2DM.

 

Finally, it is possible that certain treatments have side effects that mask the beneficial effects of glucose control. For example, hypoglycemia and weight gain could counterbalance the beneficial effects of improvements in glycemic control. It is possible that different treatment strategies could lead to more profound benefits (see below). 

 

Summary

 

Thus, the currently available data do not definitively indicate that glycemic control will have major effects on reducing ASCVD in patients with T2DM. Furthermore, there are concerns that too tight control in patients with advanced disease could be harmful. In contrast, in patients with T1DM intensive glucose control appears to have a major beneficial effect on ASCVD based on the results of the DCCT.

 

THE EFFECT OF GLUCOSE LOWERING DRUGS ON ASCVD

 

Metformin

 

In the UKPDS, metformin, while producing a similar improvement in glycemic control as insulin or sulfonylureas, markedly reduced ASCVD by approximately 40% (45). In the ten-year follow-up the patients randomized to metformin in the UKPDS continued to show a reduction in MI and all-cause mortality (32). Two other randomized controlled trials have also demonstrated cardiovascular benefits with metformin therapy.

 

A study by Kooy et al compared the effect of adding metformin or placebo in overweight or obese patients already on insulin therapy (46). After a mean follow-up of 4.3 years this study observed a reduction in macrovascular events (HR 0.61 CI- 0.40-0.94, p=0.02), which was partially accounted for by metformin’s beneficial effects on weight. In this study the difference in A1c between the metformin and placebo group was only 0.3%.

 

Hong et al randomized non-obese patients with coronary artery disease to glipizide vs. metformin therapy for three years (47). A1c levels were similar, but there was a marked reduction in cardiovascular events in the metformin treated group (HR 0.54 CI 0.30- 0.90, p=0.026).

 

In contrast, long term follow-up (21 years) of individuals in the Diabetes Prevention Program with “pre-diabetes” did not demonstrate a reduction in cardiovascular event in individuals treated with metformin (48). A reduction in the use of metformin when the formal study ended after 3 years coupled with out-of-study metformin use over time may have diluted the potential effects of metformin therapy.

 

Support for the beneficial effects of metformin on atherosclerosis comes from long term follow-up of the Diabetes Prevention Program, which compared the effect of lifestyle changes or metformin in patients at high risk of developing diabetes (49). Coronary artery calcium scores were measured on average 13-14 years after randomization (49). There were no differences in coronary artery calcium scores between the lifestyle and placebo groups. However, in males, coronary artery calcium scores were significantly lower in the metformin group vs. the placebo group. In females treated with metformin coronary artery calcium scores were similar to the placebo group. The absence of a beneficial effect of metformin in women could be due to the lower baseline coronary artery calcium scores making it more difficult to demonstrate a beneficial effect. In HIV-infected patients with the metabolic syndrome metformin similarly reduced the progression of coronary artery calcium scores (50).

 

Thus, while there are no large cardiovascular outcome trials with metformin, together, the above results suggest that metformin may reduce ASCVD and that this effect is not due to improving glucose control. Metformin decreases weight or prevents weight gain and lowers lipid levels and these or other non-glucose effects may account for the beneficial effects on ASCVD.

 

Sulfonylureas

 

Based on the University Group Diabetes Project (UGDP) sulfonylureas carry a warning regarding an increased risk of ASCVD (24,25). However, the UKPDS studied a large number of newly diagnosed patients with T2DM at risk for ASCVD and in this study improved glycemic control with sulfonylureas reduced ASCVD by approximately 16%, which just missed being statistically significant (p=0.052) (31). In the UKPDS, A1c was reduced by approximately 0.9% and the 16% reduction in ASCVD was in the range predicted based on epidemiological studies. Thus, the reduction in cardiovascular events was likely due to improvements in glycemic control and not a direct benefit of sulfonylurea treatment. In support of this conjecture is that in the UKPDS, insulin treatment resulted in a similar decrease in A1c levels and reduction in cardiovascular events (31). Additionally, a large randomized cardiovascular outcome study (Carolina Study) reported that linagliptin, a DPP-4 inhibitor, and glimepiride, a sulfonylurea, had similar effects on cardiovascular events (hazard ratio 0.98) (51). Similarly, in the ADVANCE trial patients in the intensive therapy group were randomized to gliclazide and as noted above the occurrence of cardiovascular events was similar to the control group (38).Taken together these results suggest that sulfonylureas have a neutral effect on ASCVD.

 

Meglitinides

 

The Navigator study was a double-blind, randomized clinical trial in 9,306 individuals with impaired glucose tolerance and either ASCVD or cardiovascular risk factors who received nateglinide (up to 60 mg three times daily) or placebo (52). After 5 years, nateglinide administration did not alter the incidence of cardiovascular outcomes suggesting that meglitinides do not have an adverse or beneficial effect on cardiovascular events.

 

Thiazolidinediones

 

Studies with pioglitazone have suggested a beneficial effect on ASCVD. The PROactive study was a randomized controlled trial that examined the effect of pioglitazone vs. placebo over a 3-year period in T2DM with pre-existing macrovascular disease (53). With regard to the primary endpoint (a composite of all-cause mortality, non-fatal MI including silent MI, stroke, acute coronary syndrome, endovascular or surgical intervention in the coronary or leg arteries, and amputation above the ankle), there was a 10% reduction in events in the pioglitazone group but this difference was not statistically significant (p=0.095). It should be noted that both leg revascularization and leg amputations are not typical primary end points in ASCVD trials and these could be affected by pioglitazone induced edema. When one focuses on standard ASCVD endpoints, the pioglitazone treated group did demonstrate a 16% reduction in the main secondary endpoint (composite of all-cause mortality, non-fatal MI, and stroke) that was statistically significant (p=0.027). In the pioglitazone treated group, blood pressure, A1c, triglyceride, and HDL-C levels were all improved compared to the placebo group making it very likely that the mechanism by which pioglitazone decreased vascular events was multifactorial.

 

A multicenter, double-blind trial (IRIS Trial), randomly assigned 3,876 patients with insulin resistance (defined as score of more than 3.0 on the homeostasis model assessment  of insulin resistance [HOMA-IR] index) but without diabetes and a recent ischemic stroke or TIA to treatment with either pioglitazone (target dose, 45 mg daily) or placebo (54). After 4.8 years, the primary outcome of fatal or nonfatal stroke or MI occurred in 9.0% of the pioglitazone group and 11.8% of the placebo group (hazard ratio 0.76; P=0.007). All components of the primary outcome were reduced in the pioglitazone treated group. Fasting glucose, fasting TG, and systolic and diastolic blood pressure were lower while HDL-C and LDL-C levels were higher in the pioglitazone group than in the placebo group. Although this study excluded patients with diabetes the results are consistent with and support the results of a protective effect of pioglitazone observed in the PROactive study.

 

In contrast, the TOSCA.IT study compared the effect of pioglitazone vs. sulfonylurea on ASCVD and did not observe a reduction in events with pioglitazone treatment (55). Patients with T2DM (n= 3028), inadequately controlled with metformin monotherapy (2-3 g per day), were randomized to pioglitazone or sulfonylurea and followed for a median of 57 months. Only 11% of the participants had a previous cardiovascular event. The primary outcome, was a composite of first occurrence of all-cause death, non-fatal MI, non-fatal stroke, or urgent coronary revascularization and occurred in 6.8% of the patients treated with pioglitazone and 7.2% of the patients treated with a sulfonylurea (HR 0.96; NS) (55). Limitations of this study are the small number of events due to the low-risk population studied and the relatively small number of participants. Additionally, 28% of the subjects randomized to pioglitazone prematurely discontinued the medication. Furthermore, it should be noted that when patients in this study were analyzed based on the risk of developing ASCVD those at high risk had a marked reduction in events when treated with pioglitazone compared to the sulfonylurea (56).Thus, the results of this study should be interpreted with caution.

 

Further support for the beneficial effects of pioglitazone on atherosclerosis is provided by studies that have examined the effect of pioglitazone on carotid intima-medial thickness. Both the Chicago and Pioneer studies demonstrated favorable effects on carotid intima-medial thickness in patients treated with pioglitazone compared to patients treated with sulfonylureas (57,58). Similarly, Periscope, a study that measured atheroma volume in the coronary arteries by intravascular ultrasonography, also demonstrated less atherosclerosis in the pioglitazone treated group compared to patients treated with sulfonylureas (59).

 

While the data from a variety of different types of studies strongly suggests that pioglitazone is anti-atherogenic, the results with rosiglitazone are different. Several meta-analyses of small and short-duration rosiglitazone trials suggested that rosiglitazone was associated with an increased risk of adverse cardiovascular outcomes (60,61). However, the final results of the RECORD study, a randomized trial that was specifically designed to compare the effect of rosiglitazone vs. either metformin or sulfonylurea therapy as a second oral drug in those receiving either metformin or a sulfonylurea on ASCVD events, have been published and did not reveal a difference in ASCVD death, MI, or stroke (62-64). Similarly, an analysis of patients on rosiglitazone in the BARI 2D trial also did not suggest an increase or decrease in ASCVD events in the patients treated with rosiglitazone (65). Thus, while the available data suggests that pioglitazone is anti-atherogenic, the data for rosiglitazone suggests a neutral effect. Whether these differences between pioglitazone and rosiglitazone are accounted for by their differential effects on lipid levels are unknown (see below for information on the effects of these drugs on lipid levels).

 

Numerous studies have shown that both pioglitazone and rosiglitazone increase the risk of heart failure (66).

 

DPP4 Inhibitors

 

Because of the importance of ASCVD in patients with diabetes the FDA is requiring manufacturers of new drugs to treat diabetes to carry out studies addressing ASCVD endpoints. The effect of the DPP4 inhibitors saxagliptin, alogliptin, sitagliptin, and linagliptin on ASCVD endpoints has been reported. In the saxagliptin study (SAVOR‐TIMI 53 trial), 16,492 patients with T2DM who had a history of cardiovascular events or who were at high risk were randomized to saxagliptin or placebo for 2.1 years (67). Saxagliptin did not increase or decrease cardiovascular death, MI, or ischemic stroke. Interestingly more patients treated with saxagliptin were admitted to the hospital for heart failure. The risk of heart failure with saxagliptin was greatest in patients at a high overall risk of heart failure (i.e., history of heart failure, impaired renal function, or elevated baseline levels of NT-proBNP) (68). Additionally, in the patients treated with saxagliptin the increase in heart failure was an early event with a 6-month rate of 1.1% vs. 0.6% in the placebo group (HR 1.80, p=0·001) and a 12-month rate of 1·9% vs. 1·3% (1.46; p=0.002) (68). In contrast, after 12 months no difference in the rate of heart failure was observed in the saxagliptin and placebo groups indicating that the development of heart failure is an early event (68).

 

In the alogliptin trial (EXAMINE), 5,380 patients with either an acute MI or unstable angina within the previous 15-90 days were randomized to alogliptin or placebo and followed for a median of 18 months (69). As seen in the saxagliptin study the rates of ASCVD events were similar in the alogliptin and placebo groups. The risk of hospitalization for heart failure was not statistically increased in the entire subset of patients treated with alogliptin (70). However, the hazard ratio for the subgroup of patients without heart failure at baseline was 1.76, p=0.026) (70). 

 

In the sitagliptin trial (TECOS), 14,671 patients with established ASCVD were randomized to sitagliptin or placebo for 3 years (71). Sitagliptin did not decrease the risk of major adverse cardiovascular events or increase hospitalization for heart failure. Finally, in the linagliptin trial (CARMELINA), 6,979 patients at high risk for ASCVD were randomized to linagliptin or placebo for a median follow-up of 2.2 years (72). As in the other DPP4 inhibitor studies, linagliptin did not have a beneficial effect on ASCVD events. Additionally, linagliptin did not increase the risk of hospitalization for heart failure. Thus, these results indicate that DPP4 inhibitors do not increase or decrease ASCVD. The extent to which specific DPP4 inhibitors affect heart failure needs further investigation.

 

SGLT2 Inhibitors

 

EMPA-REG OUTCOME TRIAL   

 

The effects of empagliflozin on cardiovascular morbidity and mortality in patients with T2DM has been reported (73). In this study, 7,020 patients at high risk for ASCVD were randomly assigned to receive 10 mg or 25 mg of empagliflozin or placebo once daily and were followed for 3.1 years. In the combined empagliflozin treated groups there was a statistically significant 14% reduction in the primary outcome (death from cardiovascular causes, nonfatal MI, or nonfatal stroke). As compared with placebo, empagliflozin treatment did not result in a significant difference in the occurrence of non-fatal MI or strokes. However, empagliflozin resulted in a significantly lower risk of death from cardiovascular causes (hazard ratio, 0.62), death from any cause (hazard ratio, 0.68), and hospitalization for heart failure (hazard ratio, 0.65). The beneficial effects of empagliflozin were noted to occur very rapidly and the beneficial effects on heart failure appeared to be the dominant effect compared to effects on ASCVD events. Decreases in cardiovascular outcomes and mortality with empagliflozin occurred across the range of cardiovascular risk (74). Additionally, the reduction in hospitalizations for heart failure and cardiovascular death were observed both in patients with and without heart failure at baseline (75).

 

CANVAS TRIAL

 

The effects of placebo vs. canagliflozin were determined in two combined trials involving a total of 10,142 participants with T2DM and high cardiovascular risk (76). The primary outcome was a composite of death from cardiovascular causes, nonfatal MI, or nonfatal stroke and the mean follow-up was 188 weeks. The primary outcome was reduced in the canagliflozin group (hazard ratio, 0.86; P=0.02). Death from any cause (hazard ratio 0.87; 95% CI 0.74-1.01) and death from ASCVD (hazard ratio 0.87; 95% CI 0.72-1.06) were reduced but were not statistically significant. Similarly, canagliflozin treatment did not result in a significant difference in non-fatal strokes or non-fatal MI (hazard ratio 0.90 for stroke and 0.85 for myocardial infarction). As seen with empagliflozin, hospitalization for heart failure was markedly reduced (hazard ratio 0.67; 95% CI 0.52-0.87) and this beneficial effect occurred rapidly. Notably, there was an increased risk of amputation (hazard ratio, 1.97; 95% CI, 1.41 to 2.75), which were primarily at the level of the toe or metatarsal. The basis for the increase in amputations is unknown. In other SGLT2 inhibitor studies an increase in amputations was not noted.

 

CREDENCE TRIAL

 

In a second canagliflozin trial that focused on kidney disease, a decrease in cardiovascular events was also observed (77). In this double-blind trial 4,401 patients with chronic kidney disease and T2DM were randomized to canagliflozin 100mg per day or placebo and followed for a median of 2.62 years. All the patients had an eGFR of 30 to <90 ml per minute per 1.73 m2 and albuminuria (ratio of albumin [mg] to creatinine [g], >300 to 5000). In this trial hospitalization for heart failure was reduced by 39%. The relative benefits of canagliflozin for cardiovascular outcomes was similar in individuals across the spectrum of eGFR levels (78). In contrast to the CANVAS trial, an increased risk of amputations was not observed.

 

DECLARE–TIMI 58 TRIAL

 

The effect of dapagliflozin on cardiovascular events has also been reported (79). 17,160 patients, including 10,186 without ASCVD were randomized to dapagliflozin or placebo and followed for a median of 4.2 years. The primary outcome was a composite of major adverse cardiovascular events (MACE), defined as cardiovascular death, MI, or ischemic stroke. The primary efficacy outcomes were MACE and a composite of cardiovascular death or hospitalization for heart failure. Dapagliflozin did not result in a lower rate of major adverse cardiovascular events (8.8% in the dapagliflozin group and 9.4% in the placebo group; hazard ratio, 0.93; P=0.17) but did result in a lower rate of cardiovascular death or hospitalization for heart failure (4.9% vs. 5.8%; hazard ratio, 0.83; P=0.005), which reflected a lower rate of hospitalization for heart failure (hazard ratio, 0.73; 95% CI, 0.61 to 0.88). Interestingly, in the patients with a history of a previous MI, dapagliflozin reduced the risk of a MACE (HR 0.84; P=0.039), whereas there was no effect in patients without a previous MI (80). Additionally, there was no increase in lower extremities amputations in the dapagliflozin treated group.

 

VERTIS CV

 

Patients with ASCVD and T2DM were randomized to ertugliflozin 5mg (n=2752), 15mg (2747), or placebo (n=2747) and the primary composite outcome of cardiovascular death and non-fatal MI or stroke was determined after a mean duration of follow-up of 3.5 years (81). This trial did not demonstrate a significant difference in the primary endpoint (MACE) nor any components of the primary endpoint. However, heart failure hospitalizations were significantly reduced by 30% in the patients treated with ertugliflozin (HR 0.70; CI 0.54–0.90). The benefits on heart failure were observed in both patients with a history of heart failure (decreased 37%) and patients without a history of heart failure (decreased 21%) (82).

 

SUMMARY

 

Thus, all four SGLT2 inhibitor studies demonstrated a decrease in heart failure with SGLT2 inhibitor therapy without consistent effects on ASCVD events. For additional information on the beneficial effects of SGLT2 inhibitors and SGLT1/SGLT2 inhibitors on ASCVD and heart failure see the Endotext chapter entitled “Oral and Injectable (Non-Insulin) Pharmacological Agents for the Treatment of Type 2 Diabetes” (83).

 

GLP-1 Receptor Agonists

 

The effect of six GLP-1 receptor agonists on ASCVD has been reported.

 

ELIXA

 

In the ELIXA trial 6,068 patients with T2DM who recently had a MI or been hospitalized for unstable angina were randomized to placebo or lixisenatide and followed for a median of 25 months (84). The primary end point of cardiovascular death, MI, stroke, or hospitalization for unstable angina was similar in the placebo or lixisenatide groups.

 

LEADER TRIAL

 

In contrast, the LEADER trial has shown that liraglutide decreased cardiovascular events (85). In this trial 9,340 patients at high cardiovascular risk were randomly assigned to receive liraglutide or placebo. After a median time of 3.5 years, the primary outcome of death from cardiovascular causes, nonfatal MI, or nonfatal stroke occurred in significantly fewer patients in the liraglutide group (13.0%) than in the placebo group (14.9%) (hazard ratio, 0.87, P=0.01). Additionally, deaths from cardiovascular causes (hazard ratio 0.78, P=0.007) or any cause was lower in the liraglutide group than in the placebo group (hazard ratio, 0.85; P=0.02). Interestingly patients with established ASCVD or decreased renal function (eGFR < 60) appeared to derive the greatest benefit of liraglutide treatment (86,87). As expected, weight and blood pressure were decreased in the liraglutide treated group and A1c levels were also decreased by 0.4%.

 

SUSTAIN 6 TRIAL

 

In support of the beneficial effects of GLP1 receptor agonists to reduce cardiovascular events, semaglutide, a long acting GLP-1 receptor agonist, has been shown to also reduce cardiovascular events (88). In this trial, 3,297 patients with T2DM with established ASCVD, chronic heart failure, chronic kidney disease, or age >60 with at least one cardiovascular risk factor were randomized to receive once-weekly semaglutide (0.5 mg or 1.0 mg) or placebo for 104 weeks. The primary outcome of cardiovascular death, nonfatal MI, or nonfatal stroke occurred in 6.6% of the semaglutide group and 8.9% of the placebo group (hazard ratio, 0.74; P = 0.02). In this study, both body weight and A1c levels were decreased in the patients treated with semaglutide.

 

PIONEER 6

 

In the PIONEER 6 study 3,183 patients with T2DM at high cardiovascular risk (age ≥50 years with established cardiovascular or chronic kidney disease, or age ≥60 years with cardiovascular risk factors) were randomly assigned to receive oral semaglutide or placebo (89). After a median time of 15.9 months, major adverse cardiovascular events, the primary outcome, occurred in 3.8% of the subjects treated with oral semaglutide and 4.8% of the placebo group (HR 0.79; 95% CI 0.57 to 1.11). Deaths from cardiovascular causes were 0.9% in the oral semaglutide group and 1.9% in the placebo group (HR 0.49; 95% CI, 0.27 to 0.92) while death from any cause occurred in 1.4% in the oral semaglutide group and 2.8% in the placebo group (HR 0.51; 95% CI, 0.31 to 0.84). It should be noted that the primary outcome was not statistically decreased in this study, which may be due to the relatively small number of subjects studied and the short duration of the study that together resulted in a small number of events. Additionally, more patients in the placebo group received treatment with an SGLT2 inhibitor than in the oral semaglutide group and SGLT2 inhibitors are well recognized to reduce cardiovascular events, which could also have diminished the ability to observe a decrease in events in the oral semaglutide group. Because the direction of change in cardiovascular events in PIONEER 6 and glucose lowering, weight loss, and many other effects of oral semaglutide are very similar to injected semaglutide many experts consider the effects on cardiovascular to also be similar. 

 

EXSCEL TRIAL

 

The effect of once weekly exenatide vs. placebo on cardiovascular outcomes was tested in 14,752 patients, 73% who had ASCVD (90). The primary outcome was the occurrence of death from cardiovascular causes, nonfatal MI, or nonfatal stroke. After a median follow-up of 3.2 years (duration of drug exposure 2.4 years) the primary outcome was reduced in the exenatide treated group but this difference just missed achieving statistical significance (hazard ratio 0.91; 95% CI 0.83-1.00; p=0.06). While not statistically significant these results are consistent with the results observed with liraglutide and semaglutide treatment. It should be recognized that a high percentage of patients discontinued exenatide therapy in this trial (>40%) and this could have adversely affected the ability of exenatide treatment to favorably effect ASCD outcomes.

 

HARMONY OUTCOMES TRIAL

 

The effect of once weekly albiglutide vs. placebo was tested in 9,463 patients with ASCVD (91). The primary outcome was first occurrence of cardiovascular death, MI, or stroke. After a median follow-up of 1.6 years a 22% decrease in the primary endpoint was observed in the albiglutide group (hazard ratio 0·78, p<0·0001). It should be noted that albiglutide is no longer available as it was removed from the market due to commercial considerations by Glaxo.

 

REWIND TRIAL  

 

REWIND was a randomized study of weekly subcutaneous injection of dulaglutide (1.5 mg) or placebo in 9,901 patients with T2DM who had either a previous cardiovascular event or cardiovascular risk factors (approximately 70% of patients did not have prior ASCVD) (92).  During a median follow-up of 5.4 years the primary outcome of non-fatal MI, non-fatal stroke, or death from cardiovascular causes was decreased by 12% in the dulaglutide treated group (HR 0.88, p=0.026). The decrease in events was similar in participants with and without previous ASCVD. In an analysis that focused on stroke it was noted that dulaglutide reduced ischemic stroke by 25% compared to placebo but had no effect on hemorrhagic stroke (93).

 

SUMMARY

 

Thus, four studies have clearly demonstrated that treatment with GLP-1 receptor agonists reduces cardiovascular events, two studies has provided data consistent with these results, and one study failed to demonstrate benefit (Table 1). In a meta-analysis of these seven trials it was observed that cardiovascular death, stroke, or MI was decreased by 12% (HR 0.88, p<0.0001), death from cardiovascular causes by 12% (HR 0.88, p=0.003), fatal or non-fatal stroke by 16% (HR 0.84, p<0.0001) and fatal and non-fatal MI by 9% (HR 0.91, p=0.043) (Table 1) (94). Why there are differences in results between these studies is unknown but could be due to differential effects of the GLP-1 receptor agonists, differences in the patient populations studied, or other unrecognized variables. For additional information on the beneficial effects of GLP-1 receptor agonists on ASCVD see the Endotext chapter entitled “Oral and Injectable (Non-Insulin) Pharmacological Agents for the Treatment of Type 2 Diabetes” (83).

 

Table 1. Summary of GLP-1 Receptor Agonist Cardiovascular Outcome Trials

 

Number

Prior CVD

HbA1c

Mean Follow-up (years)

Hazard Ratio* (95% CI)

P value

ELIXA
Lixisenatide

6068

100%

7.7%

2.1

1.02
(0.89-1.17)

0.78

LEADER
Liraglutide

9340

81%

8.7%

3.8

0.87
(0.78-0.97)

0.015

SUSTAIN 6
Semaglutide

3297

83%

8.7%

2.1

0.74
(0.58-0.95)

0.016

EXSCEL
Exenatide

14,752

73%

8.0%

3.2

0.91
(0.83-1.00)

0.061

HARMONY
Albiglutide

9463

100%

8.7%

1.6

0.78
(0.68-0.90)

<0.001

REWIND
Dulaglutide

9901

31%

7.3%

5.4

0>88
(0.79-0.99)

0.026

PIONEER 6**
Semaglutide oral

3183

85%

8.2%

1.3

0.79
(0.57-1.11)

0.17

Overall (94)

       

0.88
(0.82-0.94)

<0.001

*CVD death, MI, Stroke.

 

The mechanism accounting for this decrease in ASCVD is uncertain but could be related to reductions in HbA1c, body weight, systolic blood pressure, postprandial triglyceride levels, or the direct effect of activation of GLP-1 receptors on the atherosclerotic process such as improving endothelial function (95).

 

Tirzepatide

 

Tirzepatide activates both GLP-1 and GIP receptors. Long term cardiovascular trials with tirzepatide are underway. In a meta-analysis of seven randomized controlled trials with a duration of at least 26 weeks with 4,887 participants treated with tirzepatide and 2,328 control participants a 20% decrease in cardiovascular events was observed in the tirzepatide group (HR 0.80; 95% CI 0.57-1.11) suggesting that the effect of tirzepatide will be similar to the GLP-1 receptor agonists (96).   

 

Acarbose

 

In the STOP-NIDDM trial 1,429 subjects with impaired glucose tolerance were randomized to placebo vs. acarbose and followed for 3.3 years (97). In the acarbose group a 49% relative risk reduction in the development of ASCVD events (hazard ratio 0.51; P =0.03) was observed. Among cardiovascular events, the major reduction was in the risk of MI (HR, 0.09; P =.02). In a smaller trial, 135 patients hospitalized for the acute coronary syndrome who were newly diagnosed with IGT were randomly assigned to acarbose or placebo (98). During a mean follow-up of 2.3 years the risk of recurrent major adverse cardiovascular event was decreased significantly in the acarbose group compared with that in control group (26.7% versus 46.9%, P < 0.05).

 

Despite these favorable observations a large trial failed to demonstrate a beneficial effect of acarbose in Chinese patients with impaired glucose tolerance (99). In a randomized trial acarbose vs. placebo was compared in 6,522 patients with coronary heart disease and impaired glucose tolerance. The primary outcome was cardiovascular death, non-fatal MI, non-fatal stroke, hospital admission for unstable angina, and hospital admission for heart failure and patients were followed for a median of 5 years. The primary outcome was similar in the acarbose and placebo groups (hazard ratio 0.98; 95% CI 0.86-1.11, p=0·73). No significant differences were seen for death from any cause, cardiovascular death, fatal or non-fatal MI, fatal or non-fatal stroke, hospital admission for unstable angina, hospital admission for heart failure, or impaired renal function.

 

Thus, whether acarbose favorably affects ASCVD in patients at high risk for developing diabetes is uncertain. Moreover, the effect of acarbose on ASCVD in patients with diabetes is unknown. 

 

Cycloset

 

Cycloset is a quick-release bromocriptine formulation (bromocriptine-QR) that activates the D2 dopamine receptor and is approved for the treatment of diabetes. A 52 week, randomized, double-blind, multicenter trial evaluated cardiovascular safety in 3,095 patients with T2DM treated with bromocriptine-QR or placebo (100). The composite end point of first MI, stroke, coronary revascularization, or hospitalization for angina or congestive heart failure occurred in 1.8% of the bromocriptine-QR treated vs. 3.2% of the placebo-treated patients resulting in a 40% decrease in cardiovascular events (HR 0.60; CI 0.37– 0.96). Clearly further studies to confirm this finding and to elucidate the mechanism of this beneficial effect are required.

 

Bile Acid Sequestrants

 

Colesevelam is a non-absorbed, polymeric, LDL-C lowering and glucose lowering agent that is a high-capacity bile acid-binding molecule. This drug was developed primarily to lower LDL-C levels and was later noted to have favorable effects on blood glucose levels and was approved for improving glycemic control in patients with T2DM (101).

 

There have been no randomized studies that have examined the effect of bile acid sequestrants on cardiovascular end points in subjects with diabetes. In non-diabetic-subjects bile acid sequestrants have reduced cardiovascular events(102,103). Since bile acid sequestrants have a similar beneficial impact on LDL-C levels in diabetic and non-diabetic subjects one would anticipate that these drugs would also result in a reduction in events in the diabetic population.

 

Insulin

 

As described above in patients with T1DM the DCCT trial and in T2DM in the UKPDS trial demonstrated that insulin therapy reduced cardiovascular events by improving glycemic control (29-32). In the Origin Trial 12,537 people with cardiovascular risk factors plus impaired fasting glucose, impaired glucose tolerance, or T2DM were randomized to receive insulin glargine or standard care (104). The cardiovascular outcomes, which included nonfatal MI, nonfatal stroke, death from cardiovascular causes, revascularization, or hospitalization for heart failure, were similar in the glargine and placebo groups. Extended follow-up also did not demonstrate favorable effects on cardiovascular events in the glargine treated patients (105). Additionally, in patients with T2DM at high risk for cardiovascular events the occurrence of major cardiovascular events was similar in patients treated with degludec insulin or glargine insulin (106). These studies demonstrate that insulin does not accelerate atherosclerosis and by lowering glucose levels may decrease atherosclerosis, although the protective effects are mainly observed in patients with T1DM over a protracted period of time.

 

Other Studies

 

Finally, the Bari 2D study compared the effect of insulin sensitizers (metformin/TZD- mostly rosiglitazone) vs. insulin provision therapy (sulfonylureas/insulin) on cardiovascular outcomes in patients with T2DM and coronary artery disease (> 50% stenosis and positive stress test or > 70% stenosis and classic angina) (107,108). In this study, no differences in survival or cardiovascular endpoints were observed between metformin/TZD therapy vs. sulfonylurea/insulin therapy for the entire study. However, in the group with more severe coronary artery disease who were selected for coronary artery bypass surgery, the combination of coronary artery bypass and treatment with insulin sensitizers was associated with a lower rate of cardiovascular events. Why the metformin/TZD group only derived an enhanced benefit in the coronary artery bypass patients in this study is unknown. It should be noted that the vast majority of patients on TZD therapy were treated with rosiglitazone and, as discussed above, the effects of rosiglitazone on ASCVD do not appear to be as beneficial as pioglitazone. 

 

Summary

 

These studies clearly demonstrate that the method by which one improves glycemic control may be very important with different drugs having effects in addition to glucose lowering that reduce cardiovascular events (table 2). While previous treatment algorithms have primarily focused on the effect of drugs on glycemic control, current treatment recommendations for patients with diabetes are using the results of these ASCVD trials to decide which drugs should be employed. For example, the ADA is recommending that in patients with high risk or established ASCVD an SGLT inhibitor or GLP1 receptor agonist with proven cardiovascular benefit should be part of the initial treatment regimen independent of A1c levels (109).

 

Table 2. Effect of Glucose Lowering Drugs on Atherosclerotic ASCVD

Metformin

Studies suggest benefit

Sulfonylureas

No effect

Meglitinides

No effect

Thiazolidinediones

Rosiglitazone no effect; Pioglitazone- studies suggest benefit

DPP4 Inhibitors

No effect on atherosclerosis.

SGLT2 Inhibitors

Marginal effect on ASCVD, Large effect on heart failure

GLP-1 Receptor Agonists

Decrease events

Tirzepatide

Study ongoing

Acarbose

No effect

Cycloset

Further studies required

Bile Acid Sequestrants

Decrease events, further studies required

Insulin

No effect

Thiazolidinediones clearly increase the risk of heart failure while saxagliptin and alogliptin may increase risk of heart failure. SGLT2 inhibitors decrease the risk of heart failure.

 

ROLE OF OTHER RISK FACTORS IN ASCVD

 

Numerous studies have demonstrated that the traditional risk factors for ASCVD play an important role in patients with diabetes (2,4,5,110). Patients with diabetes without other risk factors have a relatively low risk of ASCVD (in most studies higher than similar non-diabetic patients), whereas the increasing prevalence of other risk factors markedly increases the risk of developing ASCVD (2). The major reversible traditional risk factors are hypertension, cigarette smoking, and lipid abnormalities (2,4,5,14,111). Other risk factors include obesity (particularly visceral obesity), insulin resistance, small dense LDL, elevated TG, low HDL-C, procoagulant state (increased PAI-1, fibrinogen), family history of early ASCVD, homocysteine, Lp (a), renal disease, albuminuria, and inflammation (C-reactive protein, SAA, cytokines) (2,4,5,110,111). In the last decade, it has become clear that to reduce the risk of ASCVD in patients with diabetes, one will not only need to improve glycemic control but also address these other cardiovascular risk factors. In the remainder of this chapter, I will focus on the dyslipidemia that occurs in patients with diabetes.

  

ROLE OF LIPIDS IN ASCVD

 

As in non-diabetic populations, epidemiological studies have shown that increased LDL-C and non-HDL-C levels and decreased HDL-C levels are associated with an increased risk of ASCVD in patients with diabetes (2,4,110,111). In the UKPDS cohort LDL-C levels were the strongest predictor of coronary artery disease (112). While it is universally accepted that elevated levels of LDL-C and non-HDL-C cause atherosclerosis and ASCVD the role of HDL-C is uncertain. Genetic studies and studies of drugs that raise HDL-C have not supported low HDL-C levels as a causative factor for atherosclerosis (113). Rather it is currently thought that HDL function is associated with atherosclerosis risk and that this does not precisely correlate with HDL-C levels (113). In patients with diabetes, elevations in serum triglyceride (TG) levels also are associated with an increased risk of ASCVD (4,111,114). With regard to TG, it is not clear whether they are a causative factor for ASCVD or whether the elevation in TG is a marker for other abnormalities (4,111,114,115). Recent Mendelian randomization studies have provided support for the hypothesis that elevated TG levels play a causal role in atherosclerosis (115,116). Unfortunately, as will be discussed later in this chapter lowering TG levels in patients on statin therapy has not decreased cardiovascular events.

 

LIPID ABNORMALITIES IN PATIENTS WITH DIABETES

 

In patients with T1DM in good glycemic control, the lipid profile is very similar to lipid profiles in the general population (110). In some studies HDL-C levels are modestly increased in patients with T1DM (117). In contrast, in patients with T2DM, even when in good glycemic control, there are abnormalities in lipid levels (118-121). It is estimated that 30-60% of patients with T2DM have dyslipidemia (5,122). Specifically, patients with T2DM often have an increase in serum TG levels, increased VLDL and IDL, and decreased HDL-C levels. Non-HDL-C levels are increased due to the increase in VLDL and IDL. LDL-C levels are typically not markedly different than in normal subjects but there is an increase in small dense LDL, a lipoprotein particle that may be particularly pro-atherogenic (123). As a consequence, there are more LDL particles, which coupled with the increases in VLDL and IDL, leads to an increase in apolipoprotein B levels (118-121). Additionally, the postprandial increase in serum TG is accentuated and elevations in postprandial lipids may increase the risk of ASCVD (118-121).

 

It should be recognized that the lipid changes in patients with T2D are characteristic of the alterations in lipid profile seen in obesity and the metabolic syndrome (insulin resistance syndrome) (124). Since a high percentage of patients with T2DM are obese, insulin resistant, and have the metabolic syndrome, it is not surprising that the prevalence of increased TG and small dense LDL and decreased HDL-C is common in patients with T2DM even when these patients are in good glycemic control. Obesity is also accompanied by increased systemic inflammation. The increasing prevalence of obesity/overweight in patients with T1D will likely result in an increased prevalence of dyslipidemia in this population.

 

Studies have shown that the anti-oxidant and anti-inflammatory functions of HDL isolated from patients with T1DM and T2DM are reduced (117,125). Additionally, the ability of HDL to facilitate cholesterol efflux is reduced in patients with T1DM and T2DM (126,127). Together these findings indicate that HDL-C levels per se may not fully reflect risk of ASCVD in patients with diabetes and that HDL function is perturbed in patients with diabetes.

 

In both T1DM and T2DM, poor glycemic control increases serum TG levels, VLDL, and IDL, and decreases HDL-C levels (119). Poor glycemic control can also result in a modest increase in LDL-C, which because of the elevation in TG is often in the small dense LDL subfraction. It is therefore important to optimize glycemic control in patients with diabetes because this will have secondary beneficial effects on lipid levels.

 

Lp(a) levels are usually within the normal range in patients with T1DM and T2DM (128). Some studies have observed no impact of diabetes mellitus on Lp(a) concentrations while other studies reported an elevation or a decrease in Lp(a) concentrations (128). The development of microalbuminuria and the onset of renal disease are associated with an increase in Lp (a) levels (129). Of note low Lp(a) levels are associated with an increased risk of developing T2DM (128). A recent very large case control study found that an Lp(a) concentration in the bottom 10% increases T2DM risk (130).

 

Table 3. Lipid Abnormalities in Patients with Diabetes

T1DM

Lipid profile is similar to controls if glycemic control is good

T2DM

Increased TG, VLDL, IDL, and non-HDL-C. Decreased HDL-C. Normal LDL-C but increase in small dense LDL, LDL particle number, and apolipoprotein B.

Poor glycemic control

Increased TG, VLDL, IDL, and non-HDL-C.  Decreased HDL-C. Modest increase in LDL-C with increase in small dense LDL, LDL particle number, and apolipoprotein B.

 

EFFECT OF GLUCOSE LOWERING DRUGS ON LIPIDS

 

Some therapies used to improve glycemic control may have an impact on lipid levels above and beyond their effects on glucose metabolism. In reviewing the literature, it is often very difficult to separate improvements in glycemic control vs. direct effects of drugs. Additionally, many of the changes induced by drug therapy result in only small changes in LDL-C, HDL-C, and TG levels, are variable from study to study, and are of questionable clinical significance. Insulin, sulfonylureas, meglinitides, DPP4 inhibitors, and alpha-glucosidase inhibitors do not appear to markedly alter fasting lipid profiles other than by improving glucose control (there are data indicating that DPP4 inhibitors and acarbose decrease postprandial triglyceride excursions, but they do not markedly alter fasting lipid levels) (131). In contrast, metformin, thiazolidinediones, GLP1 receptor agonists, bromocriptine-QR, and SGLT2 inhibitors have effects independent of glycemic control on serum lipid levels.

 

Metformin may decrease serum TG levels and LDL-C levels without altering HDL-C levels (131). In a meta-analysis of 37 trials with 2,891 patients, metformin decreased TG by 11.4mg/dL when compared with control treatment (p=0.003) (132). In an analysis of 24 trials with 1,867 patients, metformin decreased LDL-C by 8.4mg/dL compared to control treatment (p<0.001) (132). In contrast, metformin did not significantly alter HDL-C levels (132). It should be noted that in the Diabetes Prevention Program 3,234 individuals with impaired glucose metabolism were randomized to placebo, intensive lifestyle, or metformin therapy. In the metformin therapy group no significant changes were noted in TG, LDL-C, or HDL-C levels compared to the placebo group (133). Thus, metformin may have small effects on lipid levels.    

 

The effect of thiazolidinediones depends on which agent is used. Rosiglitazone increases serum LDL-C levels, increases HDL-C levels, and only decreases serum TG if the baseline TG levels are high (131). In contrast, pioglitazone has less impact on LDL-C levels, but increases HDL-C levels, and decreases TG (131). In the PROactive study, a large randomized cardiovascular outcome study, pioglitazone decreased TG levels by approximately 10%, increased HDL-C levels by approximately 10%, and increased LDL-C by 1-4% (134). It should be noted that reductions in the small dense LDL subfraction and an increase in the large buoyant LDL subfraction are seen with both thiazolidinediones (131). In a randomized head-to-head trial, it was shown that pioglitazone decreased TG levels and increased serum HDL-C levels to a greater degree than rosiglitazone treatment (135,136). Additionally, pioglitazone increased LDL-C levels less than rosiglitazone. In contrast to the differences in lipid parameters, both rosiglitazone and pioglitazone decreased A1c and C-reactive protein to a similar extent. The mechanism by which pioglitazone induces more favorable changes in lipid levels than rosiglitazone despite similar changes in glucose levels is unclear, but differential actions of ligands for nuclear hormone receptors are well described.

 

Treatment with SGLT2 inhibitors results in a small increase in LDL-C and HDL-C levels (131). In a meta-analysis of 48 randomized controlled trials SGLT2 inhibitors significantly increased LDL-C (3.8mg/dL, p < 0.00001), HDL-C (2.3mg/dL, p < 0.00001), and decreased TG levels (8.8mg/dL, p < 0.00001) (137). The mechanism for these increases in LDL and HDL cholesterol is unknown but could be due to a decrease in plasma volume. The decrease in TG levels could be secondary to weight loss.

 

Bromocriptine-QR (Cycloset) treatment decreases TG levels but has no significant effect on LDL-C or HDL-C levels (138,139). The decrease in TG levels is thought to be due to a decrease in hepatic TG synthesis, likely due to a decrease in adipose tissue lipolysis resulting in decreased blood free fatty acid levels and reduced delivery of fatty acids to the liver for TG synthesis (140).

 

Colesevelam, a bile acid sequestrant that is approved for glucose lowering, lowers LDL-C levels by 15-20% and has only a modest effect on HDL-C levels (101,141). The effect of bile acid sequestrants on TG levels varies (141). In patients with normal TG levels, bile acid sequestrants increase TG levels by a small amount. However, as baseline TG levels increase, the effect of bile acid sequestrants on TG levels becomes greater, and can result in substantial increases in TG levels (141). In patients with TG > 500mg/dL the use of bile acid sequestrants is contraindicated (141).

 

Finally, GLP-1 receptor agonists can favorably affect the lipid profile by inducing weight loss (decreasing TG and very modestly decreasing LDL-C levels) (131). In a review by Nauck and colleagues it was noted that GLP-1 receptor agonists lowered TG levels by 18 to 62mg/dL depending upon the specific GLP-1 receptor agonist while decreasing LDL-C by 3-8mg/dL and increasing HDL-C by less than 1mg/dL (142). Additionally, GLP-1 receptor agonists reduce postprandial TG by reducing circulating chylomicrons by decreasing intestinal lipoprotein production (131,142). DPP4 inhibitors have a similar effect on postprandial TG levels as GLP-1 receptor agonists while having minimal effects on fasting lipid levels (142).

 

In the SURPASS trials, tirzepatide studies TG levels were consistently decreased by 13-25% (83,143). In most studies with the exception of SURPASS 5, HDL cholesterol levels increased by 3-11% (83,143). Total cholesterol and LDL cholesterol levels were modestly decreased in most studies (83,143). Not unexpectedly given the decrease in TG levels small LDL particles were decreased. For details see the Endotext chapter Oral and Injectable (Non-Insulin) Pharmacological Agents for the Treatment of Type 2 Diabetes (83).

 

Table 4. Effect of Glucose Lowering Drugs on Lipid Levels

Metformin

Modestly decrease TG and LDL-C

Sulfonylureas

No effect

DPP4 inhibitors

Decrease postprandial TG

GLP1 analogues

Decrease fasting and postprandial TG, modestly decrease LDL-C

Tirzepatide

Decrease TG, modestly decrease LDL-C, increase HDL-C

Acarbose

Decrease postprandial TG

Pioglitazone

Rosiglitazone

Decrease TG and increase HDL-C. Small increase LDL-C but a decrease in small dense LDL

SGLT2 inhibitors

Small increase in LDL-C and HDL-C

Colesevelam

Decrease LDL-C. May increase TG

Bromocriptine-QR

Decrease TG

Insulin

No effect

 

PATHOPHYSIOLOGY OF THE DYSLIPIDEMIA OF DIABETES

Figure 1. Pathophysiology of the Dyslipidemia of Diabetes

 

Multiple mechanisms account for the dyslipidemia seen in patients with T2DM, which are affected both by the level of glucose control and by factors such as obesity and inflammation that also contribute to dyslipidemia.

 

Increase in TG

 

There are a number of different abnormalities that contribute to the dyslipidemia seen in patients with T2DM and obesity (figure 1) (119-122,144-146).

 

OVERPRODUCTION OF VLDL BY THE LIVER

 

A key abnormality is the overproduction of VLDL by the liver, which is a major contributor to the elevations in serum TG levels. The rate of secretion of VLDL is highly dependent on TG availability, which is determined by the levels of fatty acids available for the synthesis of TG in the liver. An abundance of TG prevents the intra-hepatic degradation of Apo B-100 allowing for increased VLDL formation and secretion. There are three major sources of fatty acids in the liver all of which may be altered in patients with T2DM. First, the flux of fatty acids from adipose tissue to the liver is increased. An increased mass of adipose tissue, particularly visceral stores, results in increased fatty acid delivery to the liver. Additionally, insulin suppresses the lipolysis of TG to free fatty acids in adipose tissue; thus, in patients with either poorly controlled diabetes due to a decrease in insulin or a decrease in insulin activity due to insulin resistance, the inhibition of TG lipolysis is blunted and there is increased TG breakdown leading to increased fatty acid deliver to the liver. A second source of fatty acids in the liver is de novo fatty acid synthesis. Numerous studies have shown that fatty acid synthesis is increased in the liver in patients with T2DM. This increase may be mediated by the hyperinsulinemia seen in patients with insulin resistance. While the liver is resistant to the effects of insulin on carbohydrate metabolism, the liver remains sensitive to the effects of insulin stimulating lipid synthesis. Specifically, insulin stimulates the activity of SREBP-1c, a transcription factor that increases the expression of the enzymes required for the synthesis of fatty acids. Thus, while the liver is resistant to the effects of insulin on carbohydrate metabolism the liver remains sensitive to the effects of insulin stimulating lipid synthesis. Additionally, in the presence of hyperglycemia, glucose can induce another transcription factor, carbohydrate responsive element binding protein (ChREBP), which also stimulates the transcription of the enzymes required for fatty acid synthesis. The third source of fatty acids is the uptake of TG rich lipoproteins by the liver. Studies have shown an increase in intestinal fatty acid synthesis and the enhanced secretion of chylomicrons in animal models of T2DM. This increase in chylomicrons leads to the increased delivery of fatty acids to the liver. The increase in hepatic fatty acids produced by these three pathways results in an increase in the synthesis of TG in the liver and the protection of Apo B-100 from degradation resulting in the increased formation and secretion of VLDL. Finally, insulin stimulates the post translational degradation of Apo B-100 in the liver and a decrease in insulin activity in patients with T2DM also allows for the enhanced survival of Apo B-100 promoting increased VLDL formation.

 

DECREASED DEGRADATION OF TRIGLYCERIDE RICH LIPOPROTEINS

 

While the overproduction of triglyceride rich lipoproteins by the liver and intestine are important contributors to the elevations in serum TG levels in patients with T2DM, there are also abnormalities in the metabolism of these TG rich lipoproteins. First, there is a modest decrease in lipoprotein lipase activity, the key enzyme that metabolizes TG rich lipoproteins. The expression of lipoprotein lipase is stimulated by insulin and decreased insulin activity in patients with T2DM results in a decrease in lipoprotein lipase, which plays a key role in the hydrolysis of the TG carried in chylomicrons and VLDL. Additionally, patients with T2DM have an increase in Apo C-III levels, a key regulator of TG rich lipoprotein clearance. Glucose stimulates and insulin suppresses Apo C-III expression; thus, diabetes with hyperglycemia and either insulin deficiency or insulin resistance contribute to an increase in Apo C-III. Apo C-III is an inhibitor of lipoprotein lipase activity and thereby reduces the clearance of TG rich lipoproteins. In addition, Apo C-III also inhibits the cellular uptake of lipoproteins. Studies have shown that loss of function mutations in Apo C-III lead to lower serum TG levels and a reduced risk of ASCVD (147,148). Interestingly, inhibition of Apo C-III expression results in a decrease in serum TG levels even in patients deficient in lipoprotein lipase, indicating that the ability of Apo C-III to modulate serum TG levels is not dependent solely on regulating lipoprotein lipase activity (149). Lastly, insulin resistance is associated with an increase in Angptl3, an inhibitor of LPL (150). Thus, in patients with diabetes, a decrease in clearance of TG rich lipoproteins also contributes to the elevation in serum triglyceride levels.  

 

Mechanism for the Increase in Small Dense LDL and Decrease in HDL

 

The elevation in TG rich lipoproteins in turn has effects on other lipoproteins. Specifically, cholesterol ester transfer protein (CETP) mediates the exchange of TG from TG rich VLDL and chylomicrons to LDL and HDL. The increase in TG rich lipoproteins per se leads to an increase in CETP mediated exchange, increasing the TG content of both LDL and HDL. The TG on LDL and HDL is then hydrolyzed by hepatic lipase and lipoprotein lipase leading to the production of small dense LDL and small HDL. Notably hepatic lipase activity is increased in patients with T2DM, which will also facilitate the removal of TG from LDL and HDL resulting in small lipoprotein particles. The affinity of Apo A-I for small HDL particles is reduced, leading to the disassociation of Apo A-I, which in turn leads to the accelerated clearance and breakdown of Apo A-I by the kidneys. Additionally, the production of Apo A-I may be reduced in patients with diabetes. High glucose levels can activate ChREBP and this transcription factor inhibits Apo A-I expression. Furthermore, insulin stimulates Apo A-I expression and a reduction in insulin activity due to insulin resistance or decreased insulin levels may also lead to a decrease in Apo A-I expression. The net result is lower levels of Apo A-I and HDL-C levels in patients with T2DM.

 

Role of Poor Glycemic Control

 

The above-described changes lead to the typical dyslipidemia observed in patients with T2DM (increased TG, decreased HDL-C, and an abundance of small dense LDL and small HDL). In patients with both Type 1 and T2DM, poor glycemic control can further adversely affect lipid and lipoprotein metabolism. As noted above the expression of lipoprotein lipase is stimulated by insulin. If insulin activity is very low the expression of lipoprotein lipase is severely suppressed and the metabolism of TG rich lipoproteins is markedly impaired. This leads to the delayed clearance of both chylomicrons and VLDL and elevations of TG rich lipoproteins. Additionally, insulinopenia results in a marked increase in lipolysis in adipose tissue, leading to the release of free fatty acids into the circulation. This increase in serum fatty acids results in the increased delivery of fatty acids to the liver, enhanced TG synthesis in the liver, and the increased production and secretion of VLDL. Whereas patients with T1DM who are well controlled and not obese or overweight typically have normal serum lipid profiles, if their control deteriorates, they will develop hypertriglyceridemia. In patients with T2DM deterioration of glycemic control will further exacerbate their underlying dyslipidemia resulting in greater increases in TG levels. If the synthesis of new VLDL is increased sufficiently this can result in an increase in LDL-C levels. HDL-C levels may decrease due to the formation of small HDL that are more susceptible to accelerated clearance. Improvements in glycemic control can markedly lower TG levels and may increase serum HDL-C levels. In patients with poorly controlled diabetes improvements in glycemic control may also lower LDL-C levels.

 

Role of Obesity and Inflammation

  

Most patients with T2DM and many patients with T1D are obese or overweight. Obesity is a pro-inflammatory state due to the macrophages that infiltrate adipose tissue. The cytokines produced by these macrophages and the adipokines that are produced by fat cells also alter lipid metabolism (151,152). The pro-inflammatory cytokines, TNF and IL-1, decrease the expression of lipoprotein lipase and increase the expression of angiopoietin like protein 4, an inhibitor of lipoprotein lipase. Together these changes decrease lipoprotein lipase activity, thereby delaying the clearance of TG rich lipoproteins. In addition, pro-inflammatory cytokines stimulate lipolysis in adipocytes increasing circulating free fatty acid levels, which will provide substrate for hepatic TG synthesis. In the liver, pro-inflammatory cytokines stimulate de novo fatty acid and TG synthesis. These alterations will lead to the increased production and secretion of VLDL. Thus, increases in the levels of pro-inflammatory cytokines will stimulate the production of TG rich lipoproteins and delay the clearance of TG rich lipoproteins, which together will contribute to the increase in serum TG that occurs in obese patients.

 

Obesity and the increase in pro-inflammatory cytokines may also affect HDL-C levels (153-155). First, pro-inflammatory cytokines inhibit the production of Apo A-I, the main protein constituent of HDL. Second, in many tissues pro-inflammatory cytokines decrease the expression of ABCA1 and ABCG1, which will lead to a decrease in the efflux of phospholipids and cholesterol from the cell to HDL decreasing the formation of mature HDL. Third, pro-inflammatory cytokines inhibit the production and activity of LCAT, which will limit the conversion of cholesterol to cholesterol esters in HDL. This conversion step is required for the formation of a normal spherical HDL particle and is crucial for the ability of HDL to increase the efflux of cholesterol from cells (including macrophages). Together these effects may lead to a decrease in HDL-C levels and a decrease in reverse cholesterol transport. Reverse cholesterol transport plays an important role in preventing cholesterol accumulation in macrophages and thereby reduces atherosclerosis.

 

Inflammation also decreases other important functions of HDL, such as its ability to prevent LDL oxidation (156). This reduction in the ability of HDL to protect from oxidation may be mediated in part by inflammation inducing lower levels of the enzyme paraoxonase, which is commonly seen in patients with diabetes (151,157). In parallel inflammation increases the oxidation of LDL and the amount of small dense LDL that is more susceptible to oxidation.

 

Role of Adipokines

 

Adipokines, such as leptin, adiponectin, and resistin, regulate lipid metabolism and the levels are altered in obese patients. Obesity increases serum leptin levels and leptin stimulates lipolysis in adipocytes which will increase serum free fatty acid levels (158). The circulating levels of adiponectin are decreased in subjects who are obese (159). Decreased adiponectin levels are associated with elevations in serum TG levels and decreases in HDL-C levels (159). This association is thought to be causal as studies in mice have shown that overexpressing adiponectin (transgenic mice) decreases TG and increases HDL-C levels while conversely, adiponectin knock-out mice have increased TG and decreased HDL-C levels (159). The adiponectin induced decrease in TG levels is mediated by an increased catabolism of TG rich lipoproteins due to an increase in lipoprotein lipase activity and a decrease Apo C-III, an inhibitor of lipoprotein lipase (159). The increase in HDL-C levels induced by adiponectin is mediated by an increase in hepatic Apo A-I and ABCA1, which results in the increased production of HDL particles (159).

 

Resistin is increased in subjects who are obese and the levels of resistin directly correlate with plasma TG levels (160). Moreover, resistin has been shown to stimulate hepatic VLDL production and secretion due to an increase in the synthesis of Apo B, TG, and cholesterol (160,161). Finally, resistin is associated with a decrease in HDL-C and Apo A-I levels (160).

 

EFFECT OF LIPID LOWERING ON ASCVD EVENTS IN PATIENTS WITH DIABETES

 

Monotherapy Studies

 

STATINS

 

The Cholesterol Treatment Trialists analyzed data from 18,686 subjects with diabetes (mostly T2DM) from 14 randomized trials (162). In the statin treated group there was a 9% decrease in all-cause mortality, a 13% decrease in vascular mortality, and a 21% decrease in major vascular events per 39mg/dL (1mmol/L) reduction in LDL-C. The beneficial effect of statin therapy was seen in both primary and secondary prevention patients. The effect of statin treatment on cardiovascular events in patients with diabetes was similar to that seen in non-diabetic subjects. Thus, these studies indicate that statins are beneficial in reducing ASCVD in patients with diabetes. Because of the large number of patients with diabetes included in the Heart Protection Study (HPS) and CARDS these two studies will be discussed in greater depth.

 

The HPS was a double-blind randomized trial that focused on patients at high risk for the development of cardiovascular events, including patients with a history of MIs, other atherosclerotic lesions, diabetes, and/or hypertension (163,164). Patients were between 40 and 80 years of age and had to have total serum cholesterol levels greater than 135mg/dL (thus very few patients were excluded because they did not have a high enough cholesterol level). The major strength of this trial was the large number of patients studied (>20,000). The diabetes subgroup included 5,963 subjects and thus was as large as many other prevention trials. The study was a 2x2 study design comparing simvastatin 40mg a day vs. placebo and anti-oxidant vitamins (vitamin E 600mg, vitamin C 250mg, and beta-carotene 20mg) vs. placebo and lasted approximately 5 years. Analysis of the group randomized to the anti-oxidant vitamins revealed no beneficial or harmful effects. In contrast, simvastatin therapy (40mg per day) reduced cardiovascular events, including MIs and strokes, by approximately 25% in all participants and to a similar degree in the diabetic subjects (total ASCVD reduced 27%, coronary mortality 20%, MI 37%, stroke 24%). Further analysis of the subjects with diabetes revealed that the reduction in cardiovascular events with statin therapy was similar in individuals with diabetes diagnosed for a short duration (<6 years) and for a long duration (>13 years). Similarly, subjects with diabetes in good control (HbA1c <7%) and those not in ideal control (HbA1c >7%) also benefited to a similar degree with statin therapy. Moreover, both T1DM and T2DM patients had a comparable reduction in ASCVD with simvastatin therapy. The decrease in cardiovascular events in patients with T1DM was not statistically significant because of the small number of subjects. Nevertheless, this is the only trial that included patients with T1DM and suggests that patients with T1DM will benefit from statin therapy similar to T2DM. In general, statin therapy reduced ASCVD in all subgroups of subjects with diabetes (females, males, older age, renal disease, hypertension, high TG, low HDL, ASA therapy, etc.) i.e., statin therapy benefits all patients with diabetes (note this study did not include patients with end stage renal disease but other studies have failed to show benefits of statin therapy in patients with diabetes and end stage renal disease (165)).

 

The CARDS trial specifically focused on subjects with diabetes (166). The subjects in this trial were males and females with T2DM between the ages of 40 to 75 years of age who were at high risk of developing ASCVD based on the presence of hypertension, retinopathy, renal disease, or current smoking. Of particular note, the subjects did not have any evidence of clinical atherosclerosis (myocardial disease, stroke, peripheral vascular disease) at entry and hence this study is a primary prevention trial. Inclusion criteria included LDL-C levels less than 160mg/dL and TG levels less than 600mg/dL. It is important to recognize that the average LDL-C in this trial was approximately 118mg/dL, indicating relatively low LDL-C levels. A total of 2,838 T2DM subjects were randomized to either placebo or atorvastatin 10mg a day. Atorvastatin therapy resulted in a 40% decrease in LDL-C levels with over 80% of patients achieving LDL-C levels less than 100mg/dL. Most importantly, atorvastatin therapy resulted in a 37% reduction in cardiovascular events. In addition, strokes were reduced by 48% and coronary revascularization by 31%. As seen in the HPS, subjects with relatively low LDL-C levels (LDL <120mg/dL) benefited to a similar extent as subjects with higher LDL-C levels (>120mg/dL).

 

HPS and CARDS, in combination with the other statin trials, provide conclusive evidence that statin therapy will reduce cardiovascular events in patients with diabetes. Importantly, the benefits of statin therapy are seen in patients with diabetes in both primary and secondary prevention trials. 

 

Effect of Aggressive LDL-C Lowering with Statins

 

Studies have compared reductions of LDL-C to approximately 100mg/dL to more aggressive reductions in LDL-C on atheroma volume. The Reversal Trial studied 502 symptomatic coronary artery disease patients with an average LDL-C of 150mg/dL (167). Approximately 19% of the patients in this trial had diabetes. Patients were randomized to moderate LDL lowering therapy with pravastatin 40mg per day or to aggressive lipid lowering with atorvastatin 80mg per day. As expected, LDL-C levels were considerably lower in the atorvastatin treated group (pravastatin LDL= 110mg/dL vs. atorvastatin LDL= 79mg/dL). Most importantly, when one analyzed the change in atheroma volume determined after 18 months of therapy using intravascular ultrasound, the group treated aggressively with atorvastatin had a much lower progression rate than the group treated with pravastatin. Compared with baseline values, patients treated with atorvastatin had no change in atheroma burden (there was a very slight regression of lesions), whereas patients treated with pravastatin showed progression of lesions. When one compares the extent of the reduction in LDL-C to the change in atheroma volume, a 50% reduction in LDL (LDL-C levels of approximately 75mg/dL) resulted in the absence of lesion progression. This study suggests that lowering the LDL-C to levels well below 100mg/dL is required to prevent disease progression as measured by intravascular ultrasound. Other studies, such as Asteroid, have shown that marked reductions in LDL-C (in Asteroid the mean LDL-C levels were 61mg/dL) can also result in the regression of coronary artery atherosclerosis determined by intravascular ultrasound measurements (168). Additionally, the Saturn trial demonstrated that aggressive lipid lowering with either atorvastatin 80mg or rosuvastatin 40mg would induce regression of coronary artery atherosclerosis to a similar degree in patients with and without diabetes if the LDL-C levels were reduced to less than 70mg/dL (169). Together these trials indicate that aggressive lowering of LDL-C levels to below 70mg/dL can induce regression of atherosclerotic lesions.

 

The Prove-It trial determined in patients recently hospitalized for an acute coronary syndrome whether aggressively lowering of LDL-C with atorvastatin 80mg per day vs. moderate LDL-C lowering with pravastatin 40mg per day would have a similar effect on cardiovascular end points such as death, MI, documented unstable angina requiring hospitalization, revascularization, or stroke (170,171). In this trial, approximately 18% of the patients were diabetic. As expected, the on-treatment LDL-C levels were significantly lower in patients aggressively treated with atorvastatin compared to the moderate treated pravastatin group (atorvastatin LDL-C = approximately 62 vs. pravastatin LDL-C = approximately 95mg/dL). Of great significance, death or major cardiovascular events was reduced by 16% over the two years of the study in the group aggressively treated with atorvastatin. Moreover, the risk reduction in the patients with diabetes in the aggressive treatment group was similar to that observed in non-diabetics.

 

In the treating to new targets trial (TNT) patients with stable coronary heart disease and LDL-C levels less than 130mg/dL were randomized to either 10mg or 80mg atorvastatin and followed for an average of 4.9 years (172,173). Approximately 15% of the patients had diabetes. As expected, LDL-C levels were lowered to a greater extent in the patients treated with 80mg atorvastatin than with 10mg atorvastatin (77mg/dL vs. 101mg/dL). Impressively, the occurrence of major cardiovascular events was reduced by 22% in the group treated with atorvastatin 80mg (p<0.001). In the patients with diabetes events were reduced by 25% in the high dose statin group.

 

Finally, the IDEAL trial was a randomized study that compared atorvastatin 80mg vs. simvastatin 20-40mg in 8,888 patients with a history of ASCVD (174). Approximately 12% of the patients had diabetes. As expected, LDL-C levels were reduced to a greater extent in the atorvastatin treated group than the simvastatin treated group (approximately 81mg/dL vs. 104mg/dL). Once again, the greater reduction in LDL-C levels was associated with a greater reduction in cardiovascular events. Specifically, major coronary events defined as coronary death, nonfatal MI, or cardiac arrest was reduced by 11% (p=0.07), while nonfatal acute MI were reduced by 17% (p=0.02).

 

Combining the results of the Heart Protection Study, CARDS, Reversal, Saturn, Asteroid, Prove-It, TNT, and IDEAL leads one to the conclusion that aggressive lowering of LDL-C with statin therapy will be beneficial and suggests that in high-risk patients lowering the LDL to levels well below 100mg/dL is desirable. Moreover, the Cholesterol Treatment Trialists reviewed five trials with 39,612 subjects that were designed to determine the effect of usual vs. aggressive reductions in LDL-C (175). They reported that intensive control (approximately a 19mg/dL difference in LDL-C) resulted in a 15% decrease in major vascular events, a 13% reduction in coronary death or non-fatal MI, a 19% decrease in coronary revascularization, and a 16% decrease in strokes. As will be discussed below treatment guidelines reflect the results of these studies. Additionally, as described in detail below, studies of the addition of either ezetimibe or PCSK9 inhibitors to statins further demonstrates that aggressive lowering of LDL-C levels further reduces cardiovascular events

 

FIBRATES

 

The beneficial effect of monotherapy with fibrates (e.g., gemfibrozil, fenofibrate) on ASCVD in patients with diabetes is shown in Table 5. The results of these randomized trials suggest that monotherapy with this class of drug might reduce cardiovascular events in patients with diabetes, but the data is not very robust. The largest trial was the Field Trial (176). In this trial, 9,795 patients with T2DM between the ages of 50 and 75 not taking statin therapy were randomized to fenofibrate or placebo and followed for approximately 5 years. Fenofibrate therapy resulted in a 12% decrease in LDL-C, a 29% decrease in TG, and a 5% increase in HDL-C levels. The primary outcome was coronary events (coronary heart disease death and non-fatal MI), which were reduced by 11% in the fenofibrate group but did not reach statistical significance (p= 0.16). However, there was a 24% decrease in non-fatal MI in the fenofibrate treated group (p=0.01) and a non-significant increase in coronary heart disease mortality. Total ASCVD events (coronary events plus stroke and coronary or carotid revascularization) were reduced 11% (p=0.035). These beneficial effects of fenofibrate therapy on ASCVD were observed in patients without a previous history of ASCVD. In patients with a previous history of ASCVD no benefits were observed. Additionally, the beneficial effect of fenofibrate therapy was seen only in those subjects less than 65 years of age. The beneficial effects of fenofibrate in this study may have been muted by the increased use of statins in the placebo group, which reduced the differences in lipid levels between the placebo and fenofibrate groups. If one adjusted for the addition of lipid-lowering therapy, fenofibrate reduced the risk of coronary heart disease events by 19% (p=0.01) and of total ASCVD events by 15% (p=0.004). Thus, while the results of this large trial are intriguing they do not clearly show a benefit of fibrate therapy reducing ASCVD events. The number of patients with diabetes in the other fibrate trials are relatively small (table 4).

 

While the results of the monotherapy fibrate trials have been very heterogeneous it should be noted that fibrate trials in patients with elevated TG levels have reported a greater reduction of cardiovascular events (177). Additionally, subgroup analysis of several fibrate trials has also suggested that the benefit of fibrates was greatest in patients with elevated TG levels (177,178).

 

The mechanism by which fibrates may reduce cardiovascular events is unclear. These drugs lower serum TG levels and increase HDL-C, but it should be recognized that the beneficial effects of fibrates could be due to other actions of these drugs. Specifically, these drugs activate the nuclear hormone receptor PPAR alpha, which is present in the cells that comprise the atherosclerotic lesions, and it is possible that these compounds directly affect lesion formation and development. In addition, fibrates are anti-inflammatory. In fact, analysis of the VA-HIT study suggested that much of the benefit of fibrate therapy was not due to changes in serum lipoprotein levels (179,180).

 

To summarize, while in general the studies suggest that monotherapy with fibrates may reduce ASCVD in patients with diabetes, the results are not very robust or consistent as seen in the statin trials. Of note fibrate therapy appeared to be most effective in patients with increased TG levels and decreased HDL levels, a lipid profile typically seen in patients with T2DM. However, as will be presented in detail below (combination therapy section) the addition of fibrates to statins does not reduce ASCVD.

 

Table 5. Effect of Fibrate Monotherapy on Cardiovascular Outcomes

Study

Drug

#Diabetic subjects

%Decrease controls

% Decrease diabetics

Helsinki Heart Study (181)

Gemfibrozil

135

34

60*

VA-HIT (180)

Gemfibrozil

620

24

24

DIAS (182)

Fenofibrate

418

-

23*

Sendcap (183)

Bezafibrate

164

-

70

Field (176)

Fenofibrate

9795

-

11*

* Not statistically significant

 

NIACIN

 

A single randomized trial, the Coronary Drug Project, has examined the effect of niacin monotherapy on cardiovascular outcomes (184). This trial was carried out from 1966 to 1974 (before the introduction of statin therapy) in men with a history of a prior MI and demonstrated that niacin therapy reduced cardiovascular events. The results of this study were re-analyzed to determine the effect of niacin therapy in subjects with varying baseline fasting and 1-hour post meal glucose levels (185). It was noted that 6 years of niacin therapy reduced the risk of coronary heart disease death or nonfatal MI by approximately 15-25% regardless of baseline fasting or 1-hour post glucose challenge glucose levels. Particularly notable is that reductions in events were seen in the subjects who had a fasting glucose level >126mg/dL or 1-hour glucose levels >220mg/dL (i.e., patients with diabetes). Thus, based on this single study, niacin monotherapy reduces cardiovascular events both in normal subjects and patients with diabetes. However, as will be presented in detail below (combination therapy section) the addition of niacin to statins does not reduce ASCVD.

 

EZETIMIBE

 

A multicenter, randomized trial in Japan examined the efficacy of ezetimibe in patients aged ≥75 years with elevated LDL-C (≥140 mg/dL) without a history of coronary artery disease who were not taking lipid lowering drugs (186). Patients were randomized to ezetimibe (n=1716) or usual care (n=1695) and followed for 4.1 years. The primary outcome was a composite of sudden cardiac death, MI, coronary revascularization, or stroke. In the ezetimibe group LDL-C was decreased by 25.9% and non-HDL-C by 23.1% while in the usual care group LDL-C was decreased by 18.5% and non-HDL-C by 16.5% (p<0.001 for both lipid parameters). By the end of the trial 9.6% of the patients in the usual care group and 2.1% of the ezetimibe group were taking statins. Ezetimibe reduced the incidence of the primary outcome by 34% (HR 0.66; P=0.002). Additionally, composite cardiac events were reduced by 60% (HR 0.60; P=0.039) and coronary revascularization by 62% (HR 0.38; P=0.007) in the ezetimibe group vs. the control group. There was no difference in the incidence of stroke or all-cause mortality between the groups. Approximately 25% of the patients in this trial had diabetes and the beneficial effects were similar in the diabetic and non-diabetic subjects. It should be noted that the reduction in cardiovascular events was much greater than one would expect based on the absolute difference in LDL-C levels (121mg/dL in ezetimibe group vs. 132mg/dL). As stated by the authors “Given the open-label nature of the trial, its premature termination, and issues with follow-up, the magnitude of benefit observed should be interpreted with caution.” Nevertheless, this study provides suggestive evidence that ezetimibe monotherapy may reduce cardiovascular events in patients with diabetes.

 

BEMPEDOIC ACID

 

A multicenter study of bempedoic acid in statin intolerant patients with ASCVD or at high risk for ASCVD was recently reported (187). Patients were randomized to bempedoic acid, 180 mg daily (n=6992), or placebo (n=6978) and the primary end point was death from cardiovascular causes, nonfatal MI, nonfatal stroke, or coronary revascularization. Bempedoic acid therapy reduced LDL-C and hsCRP levels by approximately 22% compared to the placebo group. The primary composite endpoint was reduced by 13% in the bempedoic acid group (HR 0.87; 95% CI, 0.79 to 0.96; P = 0.004). The four individual components of the primary endpoint were also significantly reduced in the bempedoic acid treatment group. In this trial approximately 45% of the patients had diabetes. In an analysis of the patients without clinical ASCVD, (i.e., primary prevention) (bempedoic acid n = 2100 or placebo n = 2106), there was a 30% decrease in cardiovascular events (HR 0.70: 95% CI, 0.55-0.89; P = .002) (188). In this subgroup analysis 66% of the patients had diabetes. This study clearly indicates that monotherapy with bempedoic acid will reduce cardiovascular events.

 

OTHER DRUGS

 

With regard to PCSK9 inhibitors and bile acid sequestrants there have been no randomized monotherapy studies that have examined the effect of these drugs on cardiovascular end points in subjects with diabetes. In non-diabetic subjects, monotherapy with bile acid sequestrants have reduced cardiovascular events (102,103). Since bile acid sequestrants have a similar beneficial impact on serum lipid levels in diabetic and non-diabetic subjects one would anticipate that these drugs would also result in a reduction in events in the diabetic population. Additionally, bile acid sequestrants improve glycemic control (101). However, bile acid sequestrants can raise TG levels and therefore must be used with caution in hypertriglyceridemic patients. There are no outcome studies with PCSK9 inhibitor monotherapy in patients with diabetes but given that these drugs reduce LDL-C levels and in combination with statins reduce cardiovascular events one would anticipate that PCSK9 inhibitor monotherapy will also reduce cardiovascular events.

 

Combination Therapy

 

The studies with statins have been so impressive that most patients with diabetes over the age of 40 are routinely treated with statin therapy and younger patients with diabetes at high risk for ASCVD are also typically on statin therapy (see Current Guidelines Section). Therefore, a key issue is whether the addition of other lipid lowering drugs to statins will result in a further reduction in cardiovascular events. A difficulty with such studies is that the reduction in cardiovascular events induced by statin therapy is so robust that very large trials may be required to see additional benefit.

 

STATINS + FIBRATES

 

The ACCORD-LIPID trial was designed to determine if the addition of fenofibrate to aggressive statin therapy would result in a further reduction in ASCVD in patients with T2DM (189). In this trial, 5,518 patients on statin therapy were randomized to placebo or fenofibrate therapy. The patients had diabetes for approximately 10 years and either had pre-existing ASCVD or were at high risk for developing ASCVD. During the trial, LDL-C levels were approximately 80mg/dL in both groups. There was only a small difference in HDL-C with the fenofibrate groups having a mean HDL-C of 41.2mg/dL while the control group had an HDL-C of 40.5mg/dL. Differences in TG levels were somewhat more impressive with the fenofibrate group having a mean TG level of 122mg/dL while the control group had a TG level of 144mg/dL. First occurrence of nonfatal MI, nonfatal stroke, or death from cardiovascular causes was the primary outcome and there was no statistical difference between the fenofibrate treated group and the placebo group. Additionally, there were also no statistically significant differences between the groups with regards to any of the secondary outcome measures of ASCVD. Of note, the addition of fenofibrate to statin therapy did not result in an increase in either muscle or liver side effects. On further analysis, there was a possible benefit of fenofibrate therapy in the patients in whom the baseline TG levels were elevated (>204mg/dL) and HDL-C levels decreased (<34mg/dL). Finally, similar to what has been reported in other trials, fenofibrate had beneficial effects on the progression of microvascular disease (190,191).

 

The PROMINENT trial studied the effect of pemafibrate, a new selective PPAR-alpha activator, in reducing cardiovascular outcomes in 10,497 patients (66.9% with previous ASCVD) with diabetes (192). This was a double-blind, randomized, controlled trial, in patients with T2DM, with mild-to-moderate hypertriglyceridemia (TG level, 200 to 499 mg/dL), LDL-C < 100mg/dL, and HDL-C levels < 40 mg/dL) who received either pemafibrate (0.2-mg tablets twice daily) or placebo in addition to statin therapy (96% on statins). The primary end point was a composite of nonfatal MI, ischemic stroke, coronary revascularization, or death from cardiovascular causes. Baseline fasting TG was 271 mg/dL, HDL-C 33 mg/dL, and LDL-C 78 mg/dL. Compared with placebo, pemafibrate decreased TG by 26.2%, while HDL-C increased 5.1% and LDL-C increased 12.3%. Notably non-HDL-C levels were unchanged and Apo B levels increased 4.8%. The primary endpoint was similar in the pemafibrate and placebo group (HR 1.03; 95% CI 0.91 to 1.15). The increase in LDL-C and Apo B levels likely account for the failure to reduce cardiovascular events.

 

Taken together the ACCORD study and the PROINENT trial indicate that the addition of fibrate therapy to statin therapy will not result in a reduction in cardiovascular events in patients with diabetes.

 

STATIN + NIACIN

 

The AIM-HIGH trial was designed to determine if the addition of Niaspan to aggressive statin therapy would result in a further reduction in cardiovascular events in patients with pre-existing ASCVD (193). In this trial 3,314 patients were randomized to Niaspan vs. placebo. Approximately 33% of the patients had diabetes. On trial, LDL-C levels were in the 60-70mg/dL range in both groups. As expected, HDL-C levels were increased in the Niaspan treated group (approximately 44mg/dL vs. 38mg/dL), while TG were decreased (approximately 121mg/dL vs. 155mg/dL). However, there were no differences in the primary endpoint between the control and Niaspan treated groups (Primary endpoint consisted of the first event of death from coronary heart disease, nonfatal MI, ischemic stroke, hospitalization for an acute coronary syndrome, or symptom-driven coronary or cerebral revascularization). There were also no differences in secondary endpoints except for a possible increase in strokes in the Niaspan treated group. The addition of Niaspan to statin therapy did not result in a significant increase in either muscle or liver toxicity. Thus, this study does not provide support for the addition of niacin to statins. However, it should be recognized that this was a relatively small study and a considerable number of patients stopped taking the Niaspan during the course of the study (25.4% of patients discontinued Niaspan therapy). In addition, most of the patients included in this study did not have a lipid profile that one would typically consider treating with niacin therapy. In the subset of patients with TG > 198mg/dL and HDL-C < 33mg/dL niacin showed a trend towards benefit (hazard ratio 0.74; p=0.073), suggesting that if the appropriate patient population was studied the results may have been positive (194).

 

HPS 2 Thrive also studied the effect of niacin added to statin therapy (195). This trial utilized extended-release niacin combined with laropiprant, a prostaglandin D2 receptor antagonist that reduces the flushing side effect of niacin treatment. HPS 2 Thrive was a very large trial with over 25,000 patients randomized to either niacin therapy or placebo. Approximately 32% of the patients in this trial had diabetes. The LDL-C level was 63mg/dL, the HDL-C 44mg/dL, and the TG 125mg/dL at baseline. As expected, niacin therapy resulted in a modest reduction in LDL-C (10mg/dL), a modest increase in HDL-C (6mg/dL), and a larger reduction in TG (33mg/dL). However, despite these lipid changes there were no significant differences in major cardiovascular events between the niacin and control group (risk ratio 0.96 CI 0.90- 1.03). It is unknown whether laropiprant, the prostaglandin D2 receptor antagonist, might have effects that worsen atherosclerosis and increase event rates. Similar to the AIM-HIGH study, the group of patients included in the HPS 2 Thrive trial were not the ideal patient population to test for the beneficial effects of niacin treatment added to statin therapy. Ideally, patients with high TG and high non-HDL-C levels coupled with low HDL-C levels should be studied. Nevertheless, the results of the AIM-HIGH and HPS 2 Thrive trials do not provide support for the addition of niacin to statin therapy in patients with diabetes.

 

STATIN + EZETIMIBE

 

The IMPROVE-IT trial tested whether the addition of ezetimibe to statin therapy would provide an additional beneficial effect in patients with the acute coronary syndrome (196). This was a large trial with over 18,000 patients randomized to statin therapy vs. statin therapy + ezetimibe. Approximately 27% of the patients in this trial had diabetes. On treatment LDL-C levels were 70mg/dL in the statin alone group vs. 53mg/dL in the statin + ezetimibe group. There was a small but significant 6.4% decrease in major cardiovascular events (Cardiovascular death, MI, documented unstable angina requiring re-hospitalization, coronary revascularization, or stroke) in the statin + ezetimibe group (HR 0.936 CI (0.887, 0.988) p=0.016). Cardiovascular death, non-fatal MI, or non-fatal stroke were reduced by 10% (HR 0.90 CI (0.84, 0.97) p=0.003). These beneficial effects were particularly pronounced in the patients with diabetes (Primary endpoint hazard ratio, 0.85; 95% confidence interval, 0.78-0.94) (197,198). This trial provides evidence that the combination of a statin + ezetimibe that results in a greater reduction in LDL-C levels will lead to a larger decrease in cardiovascular events than statin alone. It should be noted that the observed reduction in events was in the range expected based on the decrease in LDL-C levels.

 

The RACING trial compared rosuvastatin 10 mg plus ezetimibe 10 mg (combination therapy) vs. rosuvastatin 20mg in 3,780 patients (1,398 patients (37.0%) with diabetes) at 26 centers in South Korea (199). In the patients with diabetes the baseline LDL-C levels was 74mg/dL and during the study the median LDL-C was 53mg/dL in the combination therapy group and 61mg/dL in the high-intensity statin group (P < 0.001). After a median follow-up of 3 years the rate of cardiovascular events in patients with diabetes was 10.0% in the combination therapy group and 11.3% in the high-intensity statin group (HR: 0.89; 95% CI: 0.64–1.22; P = 0.460). Interestingly the rate of discontinuation or dose reduction of the study drug due to intolerance was lower in the combination therapy group than in the high-intensity statin group (5.2 vs. 8.7%; P = 0.014). This study demonstrates that cardiovascular outcomes were comparable between those receiving combination therapy vs. high-intensity statin monotherapy and that combination therapy significantly reduced the rate of drug discontinuation or dose reduction due to intolerance.

 

STATIN + PCSK9 INHIBITORS

 

The FOURIER trial was a randomized, double-blind, placebo-controlled trial of evolocumab vs. placebo in 27,564 patients with atherosclerotic ASCVD and an LDL-C level of 70 mg/dL or higher who were on statin therapy (200). Approximately 40% of the patients had diabetes (201). The primary end point was cardiovascular death, MI, stroke, hospitalization for unstable angina, or coronary revascularization and the key secondary end point was cardiovascular death, MI, or stroke. The median duration of follow-up was 2.2 years. Baseline LDL-C levels were 92mg/dL and evolocumab resulted in a 59% decrease in LDL-C levels (LDL-C level on treatment approximately 30mg/dL). Evolocumab treatment significantly reduced the risk of the primary end point (HR 0.85; 95% CI 0.79 to 0.92; P<0.001) and the key secondary end point (HR 0.80; 95% CI 0.73 to 0.88; P<0.001). The results were consistent across key subgroups, including the subgroup of patients in the lowest quartile for baseline LDL-C levels (median, 74 mg/dL). Of note, a similar decrease in cardiovascular events occurred in patients with diabetes treated with evolocumab and glycemic control was not altered (202). Further analysis has shown that in the small number of patients with a baseline LDL-C level less than 70mg/dL, evolocumab reduced cardiovascular events to a similar degree as in the patients with an LDL-C greater than 70mg/dL (203). Finally, the lower the on-treatment LDL-C levels (down to levels below 20mg/dL), the lower the cardiovascular event rate, suggesting that greater reductions in LDL-C levels will result in greater reductions in ASCVD (204).

 

The ODYSSEY trial was a multicenter, randomized, double-blind, placebo-controlled trial involving 18,924 patients who had an acute coronary syndrome 1 to 12 months earlier, an LDL-C level of at least 70 mg/dL, a non-HDL-C level of at least 100 mg/dL, or an Apo B level of at least 80 mg/dL while on high intensity statin therapy or the maximum tolerated statin dose (205). Approximately 29% of the patients had diabetes. Patients were randomly assigned to receive alirocumab 75 mg every 2 weeks or matching placebo. The dose of alirocumab was adjusted to target an LDL-C level of 25 to 50 mg/dL. The primary end point was a composite of death from coronary heart disease, nonfatal MI, fatal or nonfatal ischemic stroke, or unstable angina requiring hospitalization. During the trial LDL-C levels in the placebo group was 93-103mg/dL while in the alirocumab group LDL-C levels were 40mg/dL at 4 months, 48mg/dL at 12 months, and 66mg/dL at 48 months (the increase with time was due to discontinuation of alirocumab or a decrease in dose). The primary endpoint was reduced by 15% in the alirocumab group (HR 0.85; 95% CI 0.78 to 0.93; P<0.001). In addition, total mortality was reduced by 15% in the alirocumab group (HR 0.85; 95% CI 0.73 to 0.98). The absolute benefit of alirocumab was greatest in patients with a baseline LDL-C level > than 100mg/dL. In patients with an LDL-C level > than 100mg/dL the number needed to treat with alirocumab to prevent an event was only 16. It should be noted that similar to the FOURIER trial the duration of this trial was very short (median follow-up 2.8 years) which may have minimized the beneficial effects. Additionally, because alirocumab 75mg every 2 weeks was stopped if the LDL-C level was < 15mg/dL on two consecutive measurements the beneficial effects may have been blunted (7.7% of patients randomized to alirocumab were switched to placebo).

 

It should be noted that that the duration of the PCSK9 outcome trials were relatively short and it is well recognized from previous statin trials that the beneficial effects of lowering LDL-C levels takes time with only modest effects observed during the first year of treatment. In the FOURIER trial the reduction of cardiovascular death, MI, or stroke was 16% during the first year but was 25% beyond 12 months. In the ODYSSEY trial the occurrence of cardiovascular events was similar in the alirocumab and placebo group during the first year of the study with benefits of alirocumab appearing after year one. Thus, the long-term benefits of treatment with a PCSK9 inhibitor may be greater than that observed during these relatively short-term studies.

 

Additional support for the benefits of further lowering of LDL-C levels with a PCSK9 inhibitor added to statin therapy is seen in the GLAGOV trial (206). This trial was a double-blind, placebo-controlled, randomized trial of evolocumab vs. placebo in 968 patients presenting for coronary angiography. Approximately 20-21% of the patients had diabetes. The primary efficacy measure was the change in percent atheroma volume (PAV) from baseline to week 78, measured by serial intravascular ultrasonography (IVUS) imaging. Secondary efficacy measures included change in normalized total atheroma volume (TAV) and percentage of patients demonstrating plaque regression. As expected, there was a marked decrease in LDL-C levels in the evolocumab group (Placebo 93mg/dL vs. evolocumab 37mg/dL; p<0.001). PAV increased 0.05% with placebo and decreased 0.95% with evolocumab (P < .001) while TAV decreased 0.9 mm3 with placebo and 5.8 mm3 with evolocumab (P < .001). There was a linear relationship between achieved LDL-C and change in PAV (i.e., the lower the LDL-C the greater the regression in atheroma volume down to an LDL-C of 20mg/dL). Additionally, evolocumab induced plaque regression in a greater percentage of patients than placebo (64.3% vs 47.3%; P < .001 for PAV and 61.5% vs 48.9%; P < .001 for TAV). The results in the patients with diabetes were similar to the non-diabetic patients.

 

Taken together these trials demonstrate that further lowering LDL-C levels with PCSK9 monoclonal antibodies in patients taking statins will have beneficial effects on ASCVD outcomes. A study of the effect of inclisiran on ASCVD endpoints is currently in progress.

 

The results of the ezetimibe and PCSK9 trials have several important implications. First, it demonstrates that combination therapy may have benefits above and beyond statin therapy alone. Second, it provides further support for the hypothesis that lowering LDL per se will reduce cardiovascular events. Third, it suggests that lowering LDL levels to much lower levels than usual will have significant benefits. These results have implications for determining goals of therapy.

 

STATINS + LOW DOSE OMEGA-3-FATTY ACIDS

 

ORIGIN was a double-blind study in 12,536 patients at high risk for ASCVD who had impaired fasting glucose, impaired glucose tolerance, or diabetes (207).  Patients were randomized to receive a 1-gram capsule containing at least 900mg of ethyl esters of omega-3 fatty acids (EPA 465mg and DHA 375mg) or placebo for approximately 6 years. Greater than 50% of the patients were on statin therapy. The primary outcome was death from cardiovascular causes. TG levels were reduced by 14.5mg/dL in the group receiving omega-3-fatty acids compared to the placebo group (P<0.001), without a significant effect on other lipids. The incidence of the primary outcome was not significantly decreased among patients receiving omega-3-fatty acids as compared with those receiving placebo. The use of omega-3-fatty acids also had no significant effect on the rates of major vascular events, death from any cause, or death from arrhythmia.

 

A Study of Cardiovascular Events in Diabetes (ASCEND) was a randomized, placebo controlled, double blind trial of 1-gram omega-3-fattys acids (400mg EPA and 300mg DHA ethyl esters) vs. olive oil placebo in 15,480 patients with diabetes without a history of ASCVD (primary prevention trial) (208). Approximately 75% of patients were on statin therapy. The primary end point was serious vascular events (non-fatal MI, non-fatal stroke, transient ischemic attack, or vascular death). Total cholesterol, HDL-C, and non-HDL-C levels were not significantly altered by omega-3-fatty acid treatment (changes in TG levels were not reported). After a mean follow-up of 7.4 years the composite outcome of a serious vascular event or revascularization occurred in 882 patients (11.4%) on omega-3-fatty acids and 887 patients (11.5%) on placebo (rate ratio, 1.00; 95% CI, 0.91 to 1.09). Serious adverse events were similar in placebo and omega-3-fatty acid treated groups.

 

Taken together these studies indicate that low dose omega-3-fatty acids do not reduce cardiovascular events in patients with diabetes. Studies in non-diabetics have also found little effect of low dose omega-3-fatty acids on ASCVD (209).

 

STATINS + HIGH DOSE OMEGA-3-FATTY ACIDS

 

The Japan EPA Lipid Intervention Study (JELIS) was an open label non-placebo controlled study in patients on statin therapy with total cholesterol levels > 254mg/dL with (n= 3664) or without ASCVD (n=14,981) who were randomly assigned to be treated with 1800 mg of EPA (Vascepa) + statin (n=9326) or statin alone (n= 9319) with a 5 year follow-up (210). Approximately 16% of the patients had diabetes. The mean baseline TG level was 153mg/dL. The primary endpoint was any major coronary event, including sudden cardiac death, fatal and non-fatal MI, and other non-fatal events including unstable angina pectoris, angioplasty, stenting, or coronary artery bypass grafting. On treatment total cholesterol, LDL-C, and HDL-C levels were similar in the two groups but plasma TG were modestly decreased in the EPA treated group (5% decrease in EPA group compared to controls; p = 0.0001). In the EPA + statin group the primary endpoint occurred in 2.8% of the patients vs. 3.5% of the patients in the statin alone group (19% decrease; p = 0.011). Unstable angina and non-fatal coronary events were also significantly reduced in the EPA group but in this study sudden cardiac death and coronary death did not differ between groups. Unstable angina was the main component contributing to the primary endpoint and this is a more subjective endpoint than other endpoints such as a MI, stroke, or cardiovascular death. A subjective endpoint has the potential to be an unreliable endpoint in an open label study and is a major limitation of the JELIS Study. The reduction in events was similar in the subgroup of patients with diabetes. In patients with TG levels >150mg/dL and HDL-C levels < 40mg/dL there was a 53% decrease in events (211). In the EPA group, small increases in the occurrence of bleeding (1.1% vs. 0.6%, p=0.0006), gastrointestinal disturbance (3.8%% vs. 1.7%, p<0.0001) and skin abnormalities (1.7 vs. 0.7%, p<0.0001) were seen. 

 

The Reduction of Cardiovascular Events with EPA – Intervention Trial (REDUCE-IT) was a randomized, double blind trial of 2 grams twice per day of EPA ethyl ester (icosapent ethyl) (Vascepa) vs. placebo (mineral oil) in 8,179 patients with hypertriglyceridemia (135mg/dL to 499mg/dL) and established ASCVD or high ASCVD risk (diabetes plus one risk factor) who were on stable statin therapy (212). Approximately 60% of the patients in this trial had diabetes. The primary end point was a composite of cardiovascular death, nonfatal MI, nonfatal stroke, coronary revascularization, or unstable angina. At baseline, the median LDL-C level was 75.0 mg/dL, HDL-C level was 40.0 mg/dL, and TG level was 216.0 mg/dL. The median change in TG level from baseline to 1 year was a decrease of 18.3% (−39.0 mg/dL) in the EPA group and an increase of 2.2% (4.5 mg/dL) in the placebo group. After a median of 4.9 years the primary end-point occurred in 17.2% of the patients in the EPA group vs. 22.0% of the patients in the placebo group (HR 0.75; 95% CI 0.68 to 0.83; P<0.001), indicating a 25% decrease in events. The beneficial effects were similar in patients with and without diabetes. The number needed to treat to avoid one primary end-point event was 21. The reduction in cardiovascular events was noted after approximately 2 years of EPA treatment. Additionally, the risk of cardiovascular death was decreased by 20% in the EPA group (HR 0.80; 95% CI, 0.66 to 0.98; P=0.03). The cardiovascular benefits of EPA were similar across baseline levels of TG (<150, ≥150 to <200, and ≥200 mg/dL). Moreover, the cardiovascular benefits of EPA appeared to occur irrespective of the attained TG level at 1 year (≥150 or <150 mg/dL), suggesting that the cardiovascular risk reduction was not associated with attainment of a normal TG levels. An increase in hospitalization for atrial fibrillation or flutter (3.1% vs. 2.1%, P=0.004) occurred in the EPA group. In addition, serious bleeding events occurred in 2.7% of the patients in the EPA group and in 2.1% in the placebo group (P=0.06). There were no fatal bleeding events in either group and the rates of hemorrhagic stroke, serious central nervous system bleeding, and serious gastrointestinal bleeding were not significantly higher in the EPA group.

 

These results demonstrate that EPA treatment reduces ASCVD events. Of note the reduction in TG levels is relatively modest and would not be expected to result in the magnitude of the decrease in ASCVD observed in the JELIS and REDUCE-IT trials. Other actions of EPA, such as decreasing platelet function, anti-inflammation, decreasing lipid oxidation, stabilizing membranes, etc. could account for or contribute to the reduction in cardiovascular events (213). It is likely that the beneficial effects of EPA seen in the JELIS and REDUCE-IT trials are multifactorial.

 

The Statin Residual Risk Reduction with Epanova in High Risk Patients with Hypertriglyceridemia (STRENGTH) trial was a randomized, placebo controlled, double blind trial of 4 grams per day of omega-3-fatty acids (Epanova) (carboxylic acid formulation of EPA and DHA) vs. placebo (corn oil) in 13,000 patients on statins with hypertriglyceridemia (180-500mg/dL), optimal LDL-C levels (< 100mg/dL or on maximal statin therapy), low HDL-C (<42mg/dL in men and < 47mg/dL in women), and either ASCVD or high risk for ASCVD (214). The primary outcome was major atherosclerotic cardiovascular events (cardiovascular death, MI, stroke, coronary revascularization or hospitalization for unstable angina). The primary end point occurred in 785 patients (12.0%) treated with omega-3 CA vs 795 (12.2%) treated with corn oil (HR, 0.99: [95% CI, 0.90-1.09]; P = .84) (215). Thus, in contrast to EPA alone this omega-3-fatty acid formulation failed to show benefits despite reducing TG levels (18% decrease) to a similar degree as in the REDUCE-IT trial.

 

Whether EPA has special properties that resulted in the reduction in cardiovascular events in the REDUCE-IT trial or there were flaws in the trial design (the use of mineral oil as the placebo) is uncertain and debated. It should be noted that in the REDUCE-IT trial LDL-C and non-HDL-C levels were increased by approximately 10% (LDL-C by approximately 9mg/dL and non-HDL-C by approximately 10mg/dL) in the mineral oil placebo group (212). Additionally, Apo B levels were increased by 7% (6mg/dL) by mineral oil (212). Finally, an increase in hsCRP (20-30%) and other biomarkers of atherosclerosis (oxidized LDL-C, IL-6, IL-1 beta, and lipoprotein-associated phospholipase A2) were noted in the mineral oil group (212,216). In the STRENGTH trial there were no differences in LDL-C, Non-HDL-C, HDL-C, Apo B, or hsCRP levels between the treated vs. placebo groups (215). Whether EPA has special properties compared to DHA leading to a reduction in cardiovascular events or the mineral oil placebo resulted in adverse changes increasing ASCVD in the placebo resulting in an artifactual decrease in the EPA group is debated (217,218). Ideally, another large randomized cardiovascular trial with EPA ethyl ester (icosapent ethyl) (Vascepa) using a placebo other than mineral oil would resolve this controversy.

 

CURRENT GUIDELINES FOR SERUM LIPIDS

 

There are several different guidelines for treating lipids in patients with diabetes. While they all focus on lowering LDL-C there are differences between the various guidelines.

 

American Diabetes Association Guidelines

 

The 2023 American Diabetes Association (ADA) recommends that adult patients with diabetes have their lipid profile determined at the time of diabetes diagnosis and at least every 5 years thereafter or more frequently if indicated (219). This profile includes total cholesterol, HDL-C, TG, and calculated LDL-C. A lipid panel should be obtained immediately prior to initiating statin therapy. Once a patient is on statin therapy testing should be carried out 4-12 weeks after initiating therapy and annually thereafter to monitor adherence and efficacy. Lifestyle modifications including a reduction in saturated fat, trans fat, and cholesterol intake, weight loss if indicated, an increase in omega-3-fatty acids, viscous fiber, and plant stanols /sterol intake, and increased physical activity is indicated in all patients with diabetes. A focus on a Mediterranean style diet or Dietary Approaches to Stop Hypertension (DASH) diet should be encouraged. In patients with elevated TG levels glycemic control is beneficial and dietary changes and lifestyle changes including weight loss and abstinence from alcohol should be undertaken. Secondary disorders and medications that raise TG levels should be evaluated. Optimize glycemic control to improve TG and HDL-C levels. The recommendations for lipid lowering therapy are shown in table 6. If one follows these recommendations almost all patients with diabetes over the age of 40 will be on statin therapy and many under the age of 40 will also be treated with statins. The addition of ezetimibe should be considered to further lower LDL-C levels in high-risk primary prevention patients. In very high-risk patients with ASCVD if the LDL-C level on statin therapy is greater than 70mg/dL the use of ezetimibe or a PCSK9 inhibitor should be considered. The use of fibrates or niacin with statins were generally not recommended as there is no evidence of benefit. However, in patients with ASCVD or other cardiovascular risk factors on a statin with controlled LDL-C but elevated TG levels (135-499mg/dL) the addition of icosapent ethyl can be considered. Finally, in patients with fasting TG levels greater than 500mg/dL an evaluation for secondary causes of hypertriglyceridemia should be initiated and consideration of drug therapy to reduce the risk of pancreatitis.

 

Table 6. ADA Recommendations for Lipid Lowering Therapy

Primary Prevention

Age 20-39: With additional risk factors may be reasonable to initiate statin therapy

Age 40-75: Use moderate-intensity statin therapy* in addition to lifestyle therapy

Age 40-75: If at higher cardiovascular risk, including those with one or more ASCVD risk factors, it is recommended to use high intensity statin therapy to reduce LDL cholesterol by >50% and to target an LDL-C <70 mg/dL

Age 40-75: If at higher cardiovascular risk, especially those with multiple ASCVD risk factors and an LDL-C >70 mg/dL, it may be reasonable to add ezetimibe or a PCSK9 inhibitor to maximum tolerated statin therapy

Age > 75: Initiating moderate intensity statin therapy is reasonable after discussion and in patient already on statin therapy it is reasonable to continue statin therapy

Secondary Prevention

All ages: High intensity statin therapy**/maximally tolerated stain

For people with diabetes and ASCVD, treatment with high intensity statin therapy is recommended to target an LDL-C reduction of >50% and an LDL-C l goal of <55 mg/dL. Addition of ezetimibe or a PCSK9 inhibitor is recommended if this goal is not achieved

*Moderate intensity statin- atorvastatin 10-20mg, rosuvastatin 5-10mg, simvastatin 20-40mg, pravastatin 40-80mg, lovastatin 40mg, Fluvastatin XL 80mg, pitavastatin 3-4mg.

**High Intensity statin- atorvastatin 40-80mg, rosuvastatin 20-40mg.

 

American College of Cardiology and American Heart Association Guidelines

 

The 2018 American College of Cardiology and American Heart Association (ACC/AHA) guidelines recommend the following (220). “In patients 40 to 75 years of age with diabetes mellitus and LDL-C ≥70 mg/dL (≥1.8 mmol/L), start moderate-intensity statin therapy without calculating 10-year ASCVD risk. In patients with diabetes mellitus at higher risk, especially those with multiple risk factors or those 50 to 75 years of age, it is reasonable to use a high-intensity statin to reduce the LDL-C level by ≥50%.” In patients with diabetes and ASCVD they recommend “In patients with clinical ASCVD, reduce LDL-C with high-intensity statin therapy or maximally tolerated statin therapy. The more LDL-C is reduced on statin therapy, the greater will be subsequent risk reduction. Use a maximally tolerated statin to lower LDLC levels by ≥50%. In very high-risk ASCVD, use an LDL-C threshold of 70 mg/dL (1.8 mmol/L) to consider addition of non-statins to statin therapy. Very high-risk includes a history of multiple major ASCVD events or 1 major ASCVD event and multiple high-risk conditions. In very high-risk ASCVD patients, it is reasonable to add ezetimibe to maximally tolerated statin therapy when the LDL-C level remains ≥70 mg/dL (≥1.8 mmol/L). In patients at very high risk whose LDL-C level remains ≥70 mg/dL (≥1.8 mmol/L) on maximally tolerated statin and ezetimibe therapy, adding a PCSK9 inhibitor is reasonable, although the long-term safety (>3 years) is uncertain and cost effectiveness is low at mid-2018 list prices.” With regards to testing they recommend “Assess adherence and percentage response to LDL-C–lowering medications and lifestyle changes with repeat lipid measurement 4 to 12 weeks after statin initiation or dose adjustment, repeated every 3 to 12 months as needed”. Finally, there are several diabetes specific risk enhancers that are independent of other risk factors that should be considered in deciding the risk of cardiovascular events in a patient with diabetes (Table 7).

 

Table 7. Diabetes Specific Risk Enhancers That are Independent of Other Risk Factors in Diabetes

Long duration (≥10 years for type 2 diabetes mellitus or ≥20 years for type 1 diabetes mellitus

Albuminuria ≥30 mcg of albumin/mg creatinine

eGFR <60 mL/min/1.73 m2

Retinopathy

Neuropathy

ABI <0.9

ABI indicates ankle-brachial index

 

American Association of Clinical Endocrinologists/American College of Endocrinology Guidelines

 

The American Association of Clinical Endocrinologists and American College of Endocrinology guidelines consider individuals with T2DM to be at high, very high, or extreme risk for ASCVD (221,222). Patients with T1DM and a duration of diabetes of more than 15 years or two or more risk factors, poorly controlled A1c, or insulin resistance with metabolic syndrome should be considered to have an equivalent risk to patients with T2DM (221). The recommended treatment goals are shown in Table 8.

 

Table 8. ASCVD Risk Categories and Treatment Goals

Risk Category

Risk Factors/10-year risk

LDL-C mg/dL

Non-HDL-C mg/dL

Apo B mg/dL

TG

mg/dL

Extreme Risk

Diabetes and clinical ASCVD

<55

<80

<70

<150

Very High Risk

Diabetes with one or more risk factors

<70

<100

<80

<150

High Risk

Diabetes and no other risk factors

<100

<130

<90

<150

 

European Society of Cardiology and European Atherosclerosis Society Guidelines

 

The European Society of Cardiology (ESC) and European Atherosclerosis Society (EAS) has 2019 guidelines for the treatment of lipids in patients with diabetes (223). These guidelines classify patients with diabetes as very high risk, high risk, or moderate risk (table 9). The recommended goals of therapy based on risk classification are shown in table 10. As with other guidelines intensification of statin therapy should be considered before the introduction of combination therapy. If the goal is not reached, statin combination with ezetimibe should be considered next.

 

Table 9. ESC/EAS Classification of Risk in Patients with Diabetes

Very High Risk- target organ damage, or at least three major risk factors, or early onset of T1DM of long duration (>20 years)

High Risk- without target organ damage, with DM duration >10 years or another additional risk factor

Moderate Risk- Young patients (T1DM <35 years; T2DM <50 years) with DM duration <10 years, without other risk factors. Calculated SCORE >1 % and <5% for 10-year risk of fatal CVD

 

Table 10. ESC/EAS Goals of Therapy in Patients with Diabetes

 

LDL-C

Non-HDL-C

Apo B

Very High Risk

>50% reduction and <55mg/dL (<1.4mmol/L)

<85mg/d;

<65mg/dL

High Risk

>50% reduction and <70mg/dL (<1.8mmol/L)

<100mg/dL

<80mg/dL

Moderate Risk

<100mg/dL

<130mg/dL

<100mg/dL

 

European Society of Cardiology

 

The ESC has updated their guidelines in 2023 (224). An important new recommendation is that in patients with T2DM without symptomatic ASCVD or severe target organ damage, it is recommended to estimate 10-year CVD risk using SCORE2-Diabetes (225). This has resulted in a new classification of risk in patients with T2DM (table 9). The LDL-C goals for each category are shown in table 11.

 

Table 11. Cardiovascular Risk Categories and Goals in Patients with Type 2 Diabetes

Very high risk

Clinically established ASCVD or

Severe target organ damage or

10-year risk of CVD> 20% using SCORE2-Diabetes

LDL-C < 55mg/dl

Non-HDL-C <85mg/dL

High risk*

10-year risk of CVD 10% to < 20% using SCORE2-Diabetes

LDL-C < 70mg/dL

Non-HDL-C <100mg/dL

Moderate risk*

10-year risk of CVD 5% to <10% using SCORE2-Diabetes

LDL-C < 100mg/dL

Low Risk*

10-year risk of CVD <5% using SCORE2-Diabetes

no recommendations

*Patients not meeting the very high-risk category.

Severe target organ damage is defined as eGFR <45 mL/min/1.73 m2 irrespective of albuminuria; or eGFR 45–59 mL/min/1.73 m2 and microalbuminuria (UACR 30–300 mg/g; stage A2); or proteinuria (UACR>300 mg/g; stage A3), or presence of microvascular disease in at least three different sites [e.g., microalbuminuria (stage A2) plus retinopathy plus neuropathy].

 

My Guideline Recommendations

 

Thus, different organizations have proposed somewhat different recommendations for the treatment of lipids in patients with diabetes. Despite these differences it is clear that the vast majority of patients with diabetes will need to be treated with statins regardless of which guidelines one elects to follow.

 

The approach I use is to combine these recommendations (Tables 12 and 13). In patients with diabetes who have pre-existing ASCVD I initiate intensive statin therapy. I prefer LDL or non-HDL-C goals over percent reduction goals. Given the extensive data showing that the lower the LDL-C the greater the reduction in cardiovascular events most secondary prevention patients would benefit from the addition of ezetimibe to maximize LDL-C lowering without markedly increasing costs (226). In patients with diabetes 40-75 years of age without pre-existing ASCVD I calculate the 10-year risk of developing ASCVD (http://www.cvriskcalculator.com/) and identify risk enhancing factors (Table 7) and other factors that increase risk that are not included in the calculator (for example family history, inflammatory disorders, etc.). I initiate intensive statin therapy if the 10-year risk is > 7.5% or if there are multiple risk factors/risk enhancers. I initiate moderate statin therapy if the risk is < 7.5% without multiple risk factors/enhancers. Four to twelve weeks after initiating statin therapy I obtain a lipid panel to determine if the LDL and non-HDL-C levels are at goal. In patients with pre-existing ASCVD or multiple risk factors/risk enhancers (i.e., very high-risk patients) my goal is an LDL-C < 55mg/dL and a non-HDL-C < 80mg/dL. In patients that are at high-risk the goal my goal is an LDL-C < 70mg/dL and a non-HDL-C < 100mg/dL. In patients with moderate risk an LDL-C goal of < 100mg/dL and a non-HDL c < 130mg/dL is appropriate. If the levels are not at goal, I first adjust the statin dose until the patient is taking the maximally tolerated statin dose and then consider adding additional medications.  In patients with diabetes who are less than 40 years of age I initiate statin therapy if the patient has overt ASCVD, long standing diabetes, or risk factors/risk enhancers for ASCVD and the LDL and non-HDL-C levels are not at goal. In these younger patients I also calculate the life time risk of ASCVD events to start a discussion of beginning early therapy given the abundance of data indicating that initiating LDL-C lowering therapy early has great potential in markedly lowering ASCVD risk (226,227). In patients over 75 years of age with a reasonable life expectancy I begin moderate statin therapy and adjust based on response. When there is difficulty classifying a patient’s risk, I will obtain a coronary calcium score and use the score to help stratify the patient’s risk. In all cases the benefits and risks of lipid lowering therapy needs to be discussed with patients and the patient’s personnel preferences taken into account.

 

Table 12. ASCVD Risk Categories and Treatment Goals

Risk Category

Risk Factors/10-year risk

LDL-C mg/dL

Non-HDL-C mg/dL

Very High Risk

Diabetes and clinical ASCVD, multiple risk factors, 10-year risk > 20%

<55

<85

High Risk

Diabetes with one or more risk factors, 10-year risk >7.5% to <20%

<70

<100

Moderate Risk

Diabetes and no other risk factors. 10-year risk <7.5%

<100

<130

 

Table 13. Drug Therapy According to Risk Category that is Typically Required

Very High Risk

Intensive statin therapy + ezetimibe. Add PCSK9 if not close to goal

High Risk

Intensive statin therapy. Add ezetimibe if not at goal

Moderate Risk

Moderate statin therapy. Increase to intensive statin therapy if not at goal

 

TREATMENT OF LIPID ABNORMALITIES IN PATIENT WITH DIABETES

 

Life Style Changes and Weight Loss

 

Initial treatment of lipid disorders should focus on lifestyle changes. There is little debate that exercise is beneficial and that all patients with diabetes should, if possible, exercise for at least 150 minutes per week (for example 30 minutes 5 times per week). Exercise will decrease serum TG levels and increase HDL-C levels (an increase in HDL-C requires vigorous exercise) (124). Exercise increases fitness and improves insulin resistance even with limited weight loss; reductions in obesity are even more beneficial. It should be noted that many patients with diabetes may have substantial barriers to participating in exercise programs, such as comorbidities that limit exercise tolerance, risk of hypoglycemia, and presence of microvascular complications (visual impairment, neuropathy) that make exercise difficult.

 

Diet is debated to a greater extent and for detailed information on nutrition therapy for adults with diabetes see the consensus report by the American Diabetes Association (228). Everyone agrees that weight loss in obese patients is essential (124). But how this can be achieved is hotly debated with many different "experts" advocating different dietary approaches. The wide diversity of approach is likely due to the failure of any approach to be effective in the long term for the majority of obese patients with diabetes. If successful, weight loss will decrease serum TG levels, increase HDL-C levels, and modestly reduce LDL-C (124,229). To reduce LDL-C levels, it is important that the diet decrease saturated fat, trans fatty acids, and cholesterol intake. Increasing soluble fiber is also helpful.

 

It is debated whether a low fat, high complex carbohydrate diets vs. a high monounsaturated fat  diet is ideal for obese patients with diabetes (124). One can find "experts" in favor of either of these approaches and there are pros and cons to each approach. It is essential to recognize that both approaches reduce simple sugars, saturated fat, trans fatty acids, and cholesterol intake. The high complex carbohydrate diet will increase serum TG levels in some patients and if the amount of fat in the diet is markedly reduced serum HDL-C levels may decrease. In obese patients, it has been postulated that a diet high in monounsaturated fats, because of the increase in caloric density, will lead to an increase in weight gain. Both diets reduce saturated fat and cholesterol intake that will result in reductions in LDL-C levels. Additionally, both diets also reduce trans-fatty acid intake, which will have a beneficial effect on LDL and HDL-C levels and simple sugars, which will have a beneficial effect on TG levels. Very high levels of TG (>1000mg/dL), require diets that are very low in fat.

 

The available data do not indicate that any particular diet is best for inducing weight loss and it is essential to adapt the diet to fit the food preferences of the patient. Ultimately no weight loss diet will be successful if the patient cannot follow the diet for the long term and therefore the diet needs to be tailored to the specific preferences of the patient. For more detailed information on the effect of diet on lipid and lipoprotein levels and cardiovascular disease see the Endotext chapter “The Effect of Diet on Cardiovascular Disease and Lipid and Lipoprotein Levels” (229).

 

While it is widely accepted that lifestyle changes will decrease ASCVD events it should be recognized that the Look Ahead trial failed to demonstrate a reduction in ASCVD events (230). In this trial, over 5,000 overweight or obese patients with T2DM were randomized to either an intensive lifestyle intervention group that promoted weight loss through decreased caloric intake and increased physical activity or to a group that received diabetes support and education (control group). After a median follow-up of 9.6 years there was no difference in cardiovascular events (hazard ratio in the intervention group, 0.95; 95% CI 0.83 to 1.09; P=0.51). A major limitation of this study was that while the weight difference between groups was impressive during the first year of the trial, over time the differences greatly narrowed such that at the end of the trial the intensive group had a 6.0% weight loss while the control group had a 3.5% weight loss. This very modest weight difference demonstrates the difficulty in sustaining long term lifestyle changes. Thus, while weight loss and diet therapy are likely to be beneficial in reducing cardiovascular events, in clinical practice they are seldom sufficient because long-term life style changes are very difficult for most patients to maintain.

 

In contrast to the failure of lifestyle therapy in the Look Ahead trial to reduce cardiovascular events, the PREDIMED trial employing a Mediterranean diet (increased monounsaturated fats) did reduce the incidence of major ASCVD events (231,232). In this multicenter trial center trial, carried out in Spain, over 7,000 patients at high risk for developing ASCVD were randomized to three diets (primary prevention trial). A Mediterranean diet supplemented with extra-virgin olive oil, a Mediterranean diet supplemented with mixed nuts, or a control diet. Approximately 50% of the patients in this trial had T2DM. In the patients assigned to the Mediterranean diets there was 29% decrease in the primary end point (MI, stroke, and death from ASCVD). Subgroup analysis demonstrated that the Mediterranean diet was equally beneficial in patients with and without diabetes. The Mediterranean diet resulted in only a small but significant increase in HDL-C levels and a small decrease in both LDL-C and TG levels, suggesting that the beneficial effects were not mediated by changes in lipids (233).

 

The CORDIOPREV study was a single center randomized trial that compared a Mediterranean diet to a low-fat diet in 1,002 patients with ASCVD (234). Approximately 50% of the patients had diabetes. The Mediterranean diet contained a minimum of 35% of the calories as fat (22% monounsaturated fatty acids, 6% polyunsaturated fatty acids, and <10% saturated fat), 15% proteins, and a maximum of 50% carbohydrates while the low-fat diet contained less than 30% of total fat (<10% saturated fat, 12–14% monounsaturated fatty acids, and 6–8% polyunsaturated fatty acids), 15% protein, and a minimum of 55% carbohydrates. The risk of an ASCVD event was reduced by approximately 25-30% in the Mediterranean diet group. Whether these diets differed in their effects on fasting lipid levels is unknown.

 

Finally, another secondary prevention trial of a Mediterranean diet has also demonstrated a reduction in cardiovascular events. The Lyon Diet Heart Study randomized 584 patients who had a MI within 6 months to a Mediterranean type diet vs usual diet (235,236). There was a marked reduction in events in the group of patients randomized to the Mediterranean diet (cardiac death and nonfatal MI rate was 4.07 per 100 patient years in the control diet vs. 1.24 in the Mediterranean diet; p<0.0001). Unfortunately, there is no indication of the number of patients with diabetes in the Lyon Diet Heart Study or whether patients with diabetes responded similar to the entire group. Lipid levels were similar in both groups in this trial (235).

 

The results of these three randomized trials indicate that we should be encouraging our patients to follow a Mediterranean type diet. It is likely that the beneficial effects of the Mediterranean diet on ASCVD is mediated by multiple mechanisms with alterations in lipid levels making only a minor contribution.

 

For additional information on the effect of diets on lipid levels and ASCVD see the chapter entitled “The Effect of Diet on Cardiovascular Disease and Lipid and Lipoprotein Levels” in the Lipids and Lipoproteins section of Endotext (229).

 

Bariatric surgery can have profound effects on weight and can result in marked improvements in lipid profiles with a decrease in TG and LDL-C and an increase in HDL-C (124,229) Additionally, observational studies have shown a decrease in cardiovascular events following bariatric surgery in patients with and without diabetes (237-241). For additional information see the chapter entitled “Obesity and Dyslipidemia” (124).

 

Ethanol and simple sugars, in particular fructose, increase serum TG levels in susceptible patients. In patients with hypertriglyceridemia efforts should be made to reduce the intake of ethanol, simple sugars, and fructose (229).

 

Lastly, in the past some "experts" advocated the addition of fish oil supplements to reduce cardiovascular events. However, both the Origin Trial and the ASCEND Trial did not demonstrate that fish oil supplements were beneficial in patients with T2DM or patients at high risk for the development of T2DM (207,208). It should be recognized that higher doses of fish oil are required to lower serum triglyceride levels (~ 3-4 grams of DHA/EPA per day) and are useful in treating patients with high TG levels (242). Most studies of fish oil supplements in patients with diabetes have demonstrated that this is a safe approach and that worsening of glycemic control does not occur in patients with diabetes treated with fish oil supplements (242). Additionally, in some patient's high dose fish oil increases LDL-C levels, particularly when serum TG levels are very high (242). For additional information on fish oil see the chapter on Triglyceride Lowering Drugs (209).

 

Drug Therapy

 

The effect of statins, fibrates, niacin, ezetimibe, omega-3-fatty acids, bile acid sequestrants, bempedoic acid, and PCSK9 inhibitors on lipid levels in patients with diabetes is virtually identical to that seen in non-diabetic patients (Table 14). Below we will highlight issues particularly relevant to the use of these drugs in patients with diabetes. For detailed information on lipid lowering drugs see the chapters on Triglyceride Lowering Drugs and Cholesterol Lowering Drugs (141,209).

 

STATINS

 

Statins are easy to use and generally well tolerated by patients with diabetes. However, statins can adversely affect glucose homeostasis. In patients without diabetes the risk of developing diabetes is increased by approximately 10% with higher doses of statin causing a greater risk than more moderate doses (243,244). The mechanism for this adverse effect is unknown but older, obese patients with higher baseline glucose levels are at greatest risk. In patients with diabetes, an analysis of 9 studies with over 9,000 patients with diabetes reported that the patients randomized to statin therapy had a 0.12% higher HbA1c than the placebo group indicating that statin therapy is associated with only a very small increase in HbA1c levels in patients with diabetes, which is unlikely to be clinically significant (245). Individual studies such as CARDS and the Heart Protection Study have also shown only a very modest effect of statins on HbA1c levels in patients with diabetes (163,166,246). Muscle symptoms occur in patients with diabetes similar to what is observed in patients without diabetes.

 

EZETIMIBE

 

Ezetimibe is easy to use and generally well tolerated by patients with diabetes. Ezetimibe does not appear to increase the risk of new onset diabetes (199,247,248).

 

FIBRATES

 

Fibrates are easy to use and generally well tolerated by patients with diabetes. When combining fibrates with statin therapy it is best to use fenofibrate as the risk of inducing myositis is much less than when statins are used in combination with gemfibrozil, which can inhibit statin metabolism (249). In the ACCORD-LIPID Trial the incidence of muscle disorders was not increased in the statin + fenofibrate group compared to statin alone (189). The dose of fenofibrate needs to be adjusted in patients with renal disease and fenofibrate itself can induce a reversible increase in serum creatinine levels. It should be noted that marked reductions in HDL-C levels can occur in some patients treated with both fenofibrate and a TZD (250).

 

Diabetic Retinopathy

 

Fenofibrate has been shown to have beneficial effects on diabetic eye disease. The FIELD study, described earlier, was a randomized trial of fenofibrate vs. placebo in patients with T2DM. Laser treatment for retinopathy was significantly lower in the fenofibrate group than in the placebo group (3.4% patients on fenofibrate vs 4.9% on placebo; p=0.0002) (191). Fenofibrate therapy reduced the need for laser therapy to a similar extent for maculopathy (31% decrease) and for proliferative retinopathy (30% decrease). In the ophthalmology sub-study (n=1012), the primary endpoint of 2-step progression of retinopathy grade did not differ significantly between the fenofibrate and control groups (9.6% patients on fenofibrate vs 12.3% on placebo; p=0.19). In patients without pre-existing retinopathy there was no difference in progression (11.4% vs 11.7%; p=0.87). However, in patients with pre-existing retinopathy, significantly fewer patients on fenofibrate had a 2-step progression than did those on placebo (3.1% patients vs 14.6%; p=0.004). A composite endpoint of 2-step progression of retinopathy grade, macular edema, or laser treatments was significantly reduced in the fenofibrate group (HR 0.66, 95% CI 0.47-0.94; p=0.022).

 

In the ACCORD Study a subgroup of participants was evaluated for the progression of diabetic retinopathy by 3 or more steps on the Early Treatment Diabetic Retinopathy Study Severity Scale or the development of diabetic retinopathy necessitating laser photocoagulation or vitrectomy over a four-year period (190). At 4 years, the rates of progression of diabetic retinopathy were 6.5% with fenofibrate therapy (n=806) vs. 10.2% with placebo (n=787) (adjusted odds ratio, 0.60; 95% CI, 0.42 to 0.87; P = 0.006). Of note, this reduction in the progression of diabetic retinopathy was of a similar magnitude as intensive glycemic treatment vs. standard therapy.

 

Taken together these results indicate that fibrates have beneficial effects on the progression of diabetic retinopathy. The mechanisms by which fibrates decrease diabetic retinopathy are unknown.

 

Diabetic Nephropathy

 

The Diabetes Atherosclerosis Intervention Study (DAIS) evaluated the effect of fenofibrate therapy (n= 155) vs. placebo (n=159) on changes in urinary albumin excretion in patients with T2DM (251). Fenofibrate significantly reduced the worsening of albumin excretion (fenofibrate 8% vs. placebo 18%; P < 0.05). This effect was primarily due to reduced progression from normal albumin excretion to microalbuminuria (fenofibrate 3% vs. 18% placebo; P < 0.001).

 

In the FIELD trial, fenofibrate reduced urine albumin/creatinine ratio by 24% vs 11% in placebo group (p < 0.001), with 14% less progression and 18% more albuminuria regression (p < 0.001) in the fenofibrate group than in participants on placebo (252). As expected, fenofibrate therapy acutely increased plasma creatinine levels and decreased eGFR but over the long term, the increase in plasma creatinine was decreased in the fenofibrate group compared to the placebo group (14% decrease; p=0.01). Similarly, there was a slower annual decrease in eGFR in the fenofibrate group (1.19 vs 2.03 mL/min/1.73m2   annually, p < 0.001). The effect of fenofibrate on kidney function was greater in those with higher TG and lower HDL levels. End-stage renal disease, dialysis, renal transplant, and renal death were similar in the fenofibrate and placebo groups, but the incidence was low.

 

In the ACCORD-LIPID trial the post-randomization incidence of microalbuminuria was 38.2% in the fenofibrate group and 41.6% in the placebo group (p=0.01) and post-randomization incidence of macroalbumuria was 10.5% in the fibrate group and 12.3% in the placebo group (p=0.04) indicating a modest reduction in the development of proteinuria in patients treated with fenofibrate (189). There was no significant difference in the incidence of end-stage renal disease or need for dialysis between the fenofibrate group and the placebo group.

 

These studies suggest that fibrates may have a beneficial effect on diabetic kidney disease. One should recognize that reducing proteinuria is a surrogate marker and may not indicate a reduction in the development of end stage renal disease. The mechanisms accounting for decreased in proteinuria are unknown.

 

Amputations

 

In the FIELD study the risks of first amputation were decreased by 36% (p=0.02) and minor amputation events without known large-vessel disease by 47% (p=0.027) in the fenofibrate treated group (253). The reduction in amputations was independent of glucose control or dyslipidemia. No difference between the risks of major amputations was seen in the placebo and fenofibrate groups. The basis for this reduction in amputations is unknown.

 

Do Fibrates have an Independent Effect on Microvascular Disease?

 

Multiple studies cited above have now shown that fenofibrate decreases retinopathy, nephropathy, and amputation in the absence of large vessel disease. The effects are independent of blood glucose control. Given that there also was no effect of fenofibrate on cardiovascular (macrovascular) disease, these results may suggest that fenofibrate has an independent effect on microvascular disease.  Further studies are warranted, but these results should be taken into account when considering treatment of marked hypertriglyceridemia in patients with diabetes. 

 

BILE ACID SEQUESTRANTS

 

Bile acid sequestrants are relatively difficult to take due to GI toxicity (mainly constipation) (141). Diabetic subjects have an increased prevalence of constipation, which may be exacerbated by the use of bile acid sequestrants. On the other hand, in diabetic patients with diarrhea, the use of bile acid sequestrants may be advantageous. Bile acid sequestrants may also increase serum TG levels, which can be a problem in patients with diabetes who are already hypertriglyceridemic (141). An additional difficulty in using bile acid sequestrants is their potential for binding other drugs (141). Many drugs should be taken either two hours before or four hours after taking bile acid sequestrants to avoid the potential of decreased drug absorption. Patients with diabetes are frequently on multiple drugs for glycemic control, hypertension, etc., and it can sometimes be difficult to time the ingestion of bile resin sequestrants to avoid these other drugs. Colesevelam (Welchol) is a bile acid sequestrant that comes in pill, powder, or chewable bars and causes fewer side effects and has fewer interactions with other drugs than other preparations (254). The usual dose is 3.75 grams per day and can be given as tablets (​take 6 tablets once daily or 3 tablets twice daily), oral suspension (​take one packet once daily), or chewable bars (take one bar once daily). Of particular note is that a number of studies have shown that colesevelam improves glycemic control in patients with diabetes resulting in an approximately 0.5% decrease in A1c levels (255).

 

NIACIN

 

Niacin is well known to cause skin flushing and itching and GI upset (256). Additionally, niacin reduces insulin sensitivity (i.e., causes insulin resistance), which can worsen glycemic control (256). Studies have shown that niacin is usually well tolerated in diabetic subjects who are in good glycemic control (257,258). In patients with poor glycemic control, niacin is more likely to adversely impact glucose levels. In the HPS2-Thrive trial, niacin therapy significantly worsened glycemic control in patients with diabetes and induced new onset diabetes in 1.3% of subjects that were non-diabetic (195). High doses of niacin are more likely to adversely affect glycemic control. Niacin can also increase serum uric acid levels and induce gout, both of which are already common in obese patients with T2DM (256). Additionally recent trials have reported an increased incidence of infection and bleeding with niacin therapy (256). However, niacin is the most effective drug in increasing HDL-C levels, which are frequently low in patients with diabetes. 

 

OMEGA-3-FATTY ACIDS

 

A Cochrane review of fish oil in patients with diabetes have demonstrated that this is a safe approach and does not result in worsening of glycemic control in patients with diabetes (242). Fish oil effectively lowers TG levels but, in some patients, particularly those with significant hypertriglyceridemia, high dose fish oil increases LDL-C levels (242). It should be noted that fish oil products that contain just EPA (Vascepa) do not adversely affect LDL-C levels (259). When using fish oil to lower serum TG levels it is important to recognize that one is aiming to provide 3-4 grams of DHA/EPA per day. The quantity of these active omega-3-fatty acids can vary greatly from product to product. Prescription fish oil products contain large amounts of these active ingredients whereas the amount of DHA/EPA in food supplements can vary greatly and in some products levels are very low. Additionally, while prescription omega-3-fatty acid preparations have high levels of quality control, omega-3-fish oil food supplements may have contaminants and the dosage may not be precisely controlled.

 

PCSK9 INHIBITORS

 

The beneficial effects of PCSK9 inhibitors in patients with diabetes is similar to what is observed in non-diabetic patients. Additionally, except for local reactions at the injection sites PCSK9 inhibitors do not seem to cause major side effects. PCSK9 inhibitors do not appear to increase the risk of developing new-onset diabetes (260,261).

 

BEMPEDOIC ACID

 

The effect of bempedoic acid on LDL-C levels in patients with diabetes are similar to the decreases seen on non-diabetics. Patients with T2DM often have elevated uric acid levels and an increased risk of gouty attacks and a major side effect of bempedoic acid is elevating uric acid levels (141). In clinical trials, 26% of bempedoic acid-treated patients with normal baseline uric acid values experienced hyperuricemia one or more times versus 9.5% in the placebo group. Elevations in blood uric acid levels may lead to the development of gout and gout was reported in 1.5% of patients treated with bempedoic acid vs. 0.4% of patients treated with placebo. The risk for gout attacks were higher in patients with a prior history of gout (11.2% for bempedoic acid treatment vs. 1.7% in the placebo group). In patients with no prior history of gout only 1% of patients treated with bempedoic acid and 0.3% of the placebo group had a gouty attack.

 

In a meta-analysis, bempedoic acid therapy was associated with a decrease in the onset of diabetes and worsening of diabetes  (RR 0.65, p = 0.03) (7/100 vs 6.4/100 patient years) (262). In a study focusing solely on the development of new onset diabetes it was reported that new-onset diabetes/hyperglycemia occurred less frequently with bempedoic acid vs placebo (263). In the bempedoic acid cardiovascular outcome trial (CLEAR Outcomes) the development of diabetes and worsening of pre-existing diabetes was similar in the bempedoic acid and placebo groups (187).  

 

Table 14. Effect of Lipid Lowering Drugs

 

LDL-C

HDL-C

TG

 

Statins

↓ 20-60%

↑ 5-15%

↓ 0-35%*

Bile acid sequestrants

↓ 10-30%

↑ 0-10%

↑ 0-10%**

Fibrates

↓ 0-15%***

↑ 5-15%

↓ 20-50%

Niacin

↓ 10-25%

↑ 10-30%

↓ 20-50%

Ezetimibe

↓ 15-25%

↑ 1-3%

↓ 10-20%

PCSK9 Inhibitors

↓ 50-60%

↑ 5-15%

↓ 5-20%

Bempedoic Acid

↓ 15-25%

↓ 5-6%

No change

High Dose Fish Oil

↑ 0- 50%***

↑ 4- 9%

↓ 20- 50%*

 *Patients with elevated TG have largest decrease

** In patients with high TG may cause marked increase

*** In patients with high TG may increase LDL

 

Therapeutic Approach

 

FIRST PRIORITY- LDL-C

 

The first priority in treating lipid disorders in patients with diabetes is to lower the LDL-C levels to goal, unless TG are markedly elevated (> 500- 1000mg/dL), which increases the risk of pancreatitis. LDL-C is the first priority because the database linking lowering LDL-C with reducing ASCVD is extremely strong and we now have the ability to markedly decrease LDL-C levels. Dietary therapy is the initial step but, in almost all patients, will not be sufficient to achieve the LDL-C goals. If patients are willing and able to make major changes in their diet it is possible to achieve significant reductions in LDL-C levels but this seldom occurs in clinical practice (264).

 

Statins are the first-choice drugs to lower LDL-C levels and the vast majority of diabetic patients will require statin therapy. There are several statins currently available in the US and they are available as generic drugs and therefore relatively inexpensive. The particular statin used may be driven by price, ability to lower LDL-C levels, and potential drug interactions. Patients with ASCVD (secondary prevention patients) should be started on intensive statin therapy (atorvastatin 40-80mg per day or rosuvastatin 20-40mg per day). Given the extensive data showing that the lower the LDL-C the greater the reduction in ASCVD events most secondary prevention patients would benefit from the addition of ezetimibe to maximize LDL-C lowering. Ezetimibe is now a generic drug and therefore this strategy will not markedly increase costs. Similarly, primary prevention patients who are at high risk for cardiovascular events will also benefit from the use of high intensity statin therapy in combination with ezetimibe. Primary prevention patients at moderate risk can be started on moderate intensity statin therapy.

 

If a patient is unable to tolerate statins or statins as monotherapy are not sufficient to lower LDL-C to goal the second-choice drug is either ezetimibe or a PCSK9 inhibitor. Ezetimibe can be added to any statin. PCSK9 inhibitors can also be added to any statin and are the drug of choice if a large decrease in LDL-C is required to reach goal (PCSK9 inhibitors will lower LDL-C levels by 50-60% when added to a statin, whereas ezetimibe will only lower LDL-C by approximately 20%).  Bile acid sequestrants and bempedoic acid are alternatives with the use of a bile acid sequestrant particularly useful if a reduction in A1c level is also needed. It should be noted that in statin intolerant patients with ASCVD or at high risk (approximately 45% with diabetes), bempedoic acid has been shown to reduce cardiovascular events by 13% (HR 0.87; 95% CI 0.79 to 0.96; P = 0.004) (187). Ezetimibe, PCSK9 inhibitors, bempedoic acid, and bile acid sequestrants additively lower LDL-C levels when used in combination with a statin, because these drugs increase hepatic LDL receptor levels by different mechanisms, thereby resulting in a reduction in serum LDL-C levels (141). Niacin and the fibrates also lower LDL-C levels but are not usually employed to lower LDL-C levels. 

 

SECOND PRIORITY- NON-HDL-C

 

The second priority should be non-HDL-C (non-HDL-C = total cholesterol – HDL-C), which is particularly important in patients with elevated TG levels (>150mg/dL). Non-HDL-C is a measure of all the pro-atherogenic apolipoprotein B containing particles. Numerous studies have shown that non-HDL-C is a strong risk factor for the development of ASCVD (265). The non-HDL-C goals are approximately 30mg/dL greater than the LDL-C goals. For example, if the LDL goal is <100mg/dL then the non-HDL-C goal would be <130mg/dL. Drugs that reduce either LDL-C or TG levels will reduce non-HDL-C levels. To lower TG levels initial therapy should focus on glycemic control and lifestyle changes including weight loss if indicated and a decrease in simple sugars and ethanol intake. Additionally, if possible, discontinue medications that increase triglyceride levels. As discussed above, studies with the omega-3-fatty acid icosapent ethyl (EPA; Vascepa) added to statin therapy have reduced the risk of cardiovascular events. The National Lipid Association has recommended “that for patients aged ≥45 years with clinical ASCVD, or aged ≥50 years with diabetes mellitus requiring medication plus ≥1 additional risk factor, with fasting TGs 135 to 499 mg/dL on high-intensity or maximally tolerated statin therapy (±ezetimibe), treatment with icosapent ethyl is recommended for ASCVD risk reduction” (266). As noted earlier in this chapter there is controversy regarding the benefits of icosapent ethyl on cardiovascular outcomes with some experts interpreting the beneficial results of the REDUCE-IT trial as being due to the adverse effects of the mineral oil placebo (217).

 

VERY HIGH TG

 

Patients with very high TG levels (> 500-1000 mg/dL) are at risk of pancreatitis and therefore lifestyle interventions including diet, exercise, and weight loss if indicated should be initiated early. Treatment is a low-fat diet and glycemic control. Additionally, decreasing simple sugars and avoiding alcohol is beneficial. When the TG levels are very elevated (> 1000mg/dL) a very low-fat diet (5-20% of calories as fat) should be the primary treatment until the TG levels decrease to < 1000mg/dL. Treating secondary disorders that raise TG levels and when possible, stopping drugs that increase TG levels is essential. If the TG levels remain above 500mg/dL the addition of fenofibrate or omega-3-fatty acids is indicated.

 

LOW HDL-C

 

While there is strong epidemiologic data linking low HDL-C levels with ASCVD there is no clinical trials demonstrating that increasing HDL-C levels reduce ASCVD. Thus, the use of drugs such as niacin to raise HDL-C levels is not recommended.

 

CONCLUSION

 

Patients with diabetes, particularly T2DM, often have dyslipidemia. Modern therapy of patients with diabetes demands that we aggressively treat lipids to reduce the high risk of ASCVD in this susceptible population and in those with very high TG to reduce the risk of pancreatitis.

 

REFERENCES

 

  1. Milicevic Z, Raz I, Beattie SD, Campaigne BN, Sarwat S, Gromniak E, Kowalska I, Galic E, Tan M, Hanefeld M. Natural history of cardiovascular disease in patients with diabetes: role of hyperglycemia. Diabetes Care 2008; 31 Suppl 2:S155-160
  2. Feingold KR, Siperstein MD. Diabetic vascular disease. Adv Intern Med 1986; 31:309-340
  3. Regensteiner JG, Golden S, Huebschmann AG, Barrett-Connor E, Chang AY, Chyun D, Fox CS, Kim C, Mehta N, Reckelhoff JF, Reusch JE, Rexrode KM, Sumner AE, Welty FK, Wenger NK, Anton B. Sex Differences in the Cardiovascular Consequences of Diabetes Mellitus: A Scientific Statement From the American Heart Association. Circulation 2015; 132:2424-2447
  4. Fox CS, Golden SH, Anderson C, Bray GA, Burke LE, de Boer IH, Deedwania P, Eckel RH, Ershow AG, Fradkin J, Inzucchi SE, Kosiborod M, Nelson RG, Patel MJ, Pignone M, Quinn L, Schauer PR, Selvin E, Vafiadis DK. Update on Prevention of Cardiovascular Disease in Adults With Type 2 Diabetes Mellitus in Light of Recent Evidence: A Scientific Statement From the American Heart Association and the American Diabetes Association. Diabetes Care 2015; 38:1777-1803
  5. Low Wang CC, Hess CN, Hiatt WR, Goldfine AB. Clinical Update: Cardiovascular Disease in Diabetes Mellitus: Atherosclerotic Cardiovascular Disease and Heart Failure in Type 2 Diabetes Mellitus - Mechanisms, Management, and Clinical Considerations. Circulation 2016; 133:2459-2502
  6. Kannel WB, McGee DL. Diabetes and cardiovascular disease. The Framingham study. JAMA 1979; 241:2035-2038
  7. Members Writing Group, Mozaffarian D, Benjamin EJ, Go AS, Arnett DK, Blaha MJ, Cushman M, Das SR, de Ferranti S, Despres JP, Fullerton HJ, Howard VJ, Huffman MD, Isasi CR, Jimenez MC, Judd SE, Kissela BM, Lichtman JH, Lisabeth LD, Liu S, Mackey RH, Magid DJ, McGuire DK, Mohler ER, 3rd, Moy CS, Muntner P, Mussolino ME, Nasir K, Neumar RW, Nichol G, Palaniappan L, Pandey DK, Reeves MJ, Rodriguez CJ, Rosamond W, Sorlie PD, Stein J, Towfighi A, Turan TN, Virani SS, Woo D, Yeh RW, Turner MB. Executive Summary: Heart Disease and Stroke Statistics--2016 Update: A Report From the American Heart Association. Circulation 2016; 133:447-454
  8. Haffner SM, Lehto S, Ronnemaa T, Pyorala K, Laakso M. Mortality from coronary heart disease in subjects with type 2 diabetes and in nondiabetic subjects with and without prior myocardial infarction. N Engl J Med 1998; 339:229-234
  9. Evans JM, Wang J, Morris AD. Comparison of cardiovascular risk between patients with type 2 diabetes and those who had had a myocardial infarction: cross sectional and cohort studies. BMJ 2002; 324:939-942
  10. Wannamethee SG, Shaper AG, Whincup PH, Lennon L, Sattar N. Impact of diabetes on cardiovascular disease risk and all-cause mortality in older men: influence of age at onset, diabetes duration, and established and novel risk factors. Arch Intern Med 2011; 171:404-410
  11. Howard BV, Best LG, Galloway JM, Howard WJ, Jones K, Lee ET, Ratner RE, Resnick HE, Devereux RB. Coronary heart disease risk equivalence in diabetes depends on concomitant risk factors. Diabetes Care 2006; 29:391-397
  12. Rawshani A, Rawshani A, Franzen S, Sattar N, Eliasson B, Svensson AM, Zethelius B, Miftaraj M, McGuire DK, Rosengren A, Gudbjornsdottir S. Risk Factors, Mortality, and Cardiovascular Outcomes in Patients with Type 2 Diabetes. N Engl J Med 2018; 379:633-644
  13. Lind M, Svensson AM, Kosiborod M, Gudbjornsdottir S, Pivodic A, Wedel H, Dahlqvist S, Clements M, Rosengren A. Glycemic control and excess mortality in type 1 diabetes. N Engl J Med 2014; 371:1972-1982
  14. de Ferranti SD, de Boer IH, Fonseca V, Fox CS, Golden SH, Lavie CJ, Magge SN, Marx N, McGuire DK, Orchard TJ, Zinman B, Eckel RH. Type 1 diabetes mellitus and cardiovascular disease: a scientific statement from the American Heart Association and American Diabetes Association. Diabetes Care 2014; 37:2843-2863
  15. Maahs DM, Daniels SR, de Ferranti SD, Dichek HL, Flynn J, Goldstein BI, Kelly AS, Nadeau KJ, Martyn-Nemeth P, Osganian SK, Quinn L, Shah AS, Urbina E. Cardiovascular disease risk factors in youth with diabetes mellitus: a scientific statement from the American Heart Association. Circulation 2014; 130:1532-1558
  16. Huxley RR, Peters SA, Mishra GD, Woodward M. Risk of all-cause mortality and vascular events in women versus men with type 1 diabetes: a systematic review and meta-analysis. Lancet Diabetes Endocrinol 2015; 3:198-206
  17. Rawshani A, Sattar N, Franzen S, Rawshani A, Hattersley AT, Svensson AM, Eliasson B, Gudbjornsdottir S. Excess mortality and cardiovascular disease in young adults with type 1 diabetes in relation to age at onset: a nationwide, register-based cohort study. Lancet 2018; 392:477-486
  18. Chillaron JJ, Flores Le-Roux JA, Benaiges D, Pedro-Botet J. Type 1 diabetes, metabolic syndrome and cardiovascular risk. Metabolism 2014; 63:181-187
  19. Constantino MI, Molyneaux L, Limacher-Gisler F, Al-Saeed A, Luo C, Wu T, Twigg SM, Yue DK, Wong J. Long-term complications and mortality in young-onset diabetes: type 2 diabetes is more hazardous and lethal than type 1 diabetes. Diabetes Care 2013; 36:3863-3869
  20. Preis SR, Hwang SJ, Coady S, Pencina MJ, D'Agostino RB, Sr., Savage PJ, Levy D, Fox CS. Trends in all-cause and cardiovascular disease mortality among women and men with and without diabetes mellitus in the Framingham Heart Study, 1950 to 2005. Circulation 2009; 119:1728-1735
  21. Matuleviciene-Anangen V, Rosengren A, Svensson AM, Pivodic A, Gudbjornsdottir S, Wedel H, Kosiborod M, Haraldsson B, Lind M. Glycaemic control and excess risk of major coronary events in persons with type 1 diabetes. Heart 2017; 103:1687-1695
  22. Miller RG, Orchard TJ, Costacou T. 30-Year Cardiovascular Disease in Type 1 Diabetes: Risk and Risk Factors Differ by Long-term Patterns of Glycemic Control. Diabetes Care 2022; 45:142-150
  23. Abraira C, Colwell JA, Nuttall FQ, Sawin CT, Nagel NJ, Comstock JP, Emanuele NV, Levin SR, Henderson W, Lee HS. Veterans Affairs Cooperative Study on glycemic control and complications in type II diabetes (VA CSDM). Results of the feasibility trial. Veterans Affairs Cooperative Study in Type II Diabetes. Diabetes Care1995; 18:1113-1123
  24. Goldner MG, Knatterud GL, Prout TE. Effects of hypoglycemic agents on vascular complications in patients with adult-onset diabetes. 3. Clinical implications of UGDP results. JAMA 1971; 218:1400-1410
  25. Meinert CL, Knatterud GL, Prout TE, Klimt CR. A study of the effects of hypoglycemic agents on vascular complications in patients with adult-onset diabetes. II. Mortality results. Diabetes 1970; 19:Suppl:789-830
  26. The effect of intensive treatment of diabetes on the development and progression of long-term complications in insulin-dependent diabetes mellitus. The Diabetes Control and Complications Trial Research Group. N Engl J Med 1993; 329:977-986
  27. Ohkubo Y, Kishikawa H, Araki E, Miyata T, Isami S, Motoyoshi S, Kojima Y, Furuyoshi N, Shichiri M. Intensive insulin therapy prevents the progression of diabetic microvascular complications in Japanese patients with non-insulin-dependent diabetes mellitus: a randomized prospective 6-year study. Diabetes Res Clin Pract 1995; 28:103-117
  28. Shichiri M, Kishikawa H, Ohkubo Y, Wake N. Long-term results of the Kumamoto Study on optimal diabetes control in type 2 diabetic patients. Diabetes Care 2000; 23 Suppl 2:B21-29
  29. Lachin JM, Orchard TJ, Nathan DM, Group DER. Update on cardiovascular outcomes at 30 years of the diabetes control and complications trial/epidemiology of diabetes interventions and complications study. Diabetes Care2014; 37:39-43
  30. Nathan DM, Cleary PA, Backlund JY, Genuth SM, Lachin JM, Orchard TJ, Raskin P, Zinman B, Diabetes C, Complications Trial/Epidemiology of Diabetes I, Complications Study Research G. Intensive diabetes treatment and cardiovascular disease in patients with type 1 diabetes. N Engl J Med 2005; 353:2643-2653
  31. Intensive blood-glucose control with sulphonylureas or insulin compared with conventional treatment and risk of complications in patients with type 2 diabetes (UKPDS 33). UK Prospective Diabetes Study (UKPDS) Group. Lancet 1998; 352:837-853
  32. Holman RR, Paul SK, Bethel MA, Matthews DR, Neil HA. 10-year follow-up of intensive glucose control in type 2 diabetes. N Engl J Med 2008; 359:1577-1589
  33. Malmberg K. Prospective randomised study of intensive insulin treatment on long term survival after acute myocardial infarction in patients with diabetes mellitus. DIGAMI (Diabetes Mellitus, Insulin Glucose Infusion in Acute Myocardial Infarction) Study Group. BMJ 1997; 314:1512-1515
  34. Mellbin LG, Malmberg K, Norhammar A, Wedel H, Ryden L, Investigators D. The impact of glucose lowering treatment on long-term prognosis in patients with type 2 diabetes and myocardial infarction: a report from the DIGAMI 2 trial. Eur Heart J 2008; 29:166-176
  35. Action to Control Cardiovascular Risk in Diabetes Study Group, Gerstein HC, Miller ME, Byington RP, Goff DC, Jr., Bigger JT, Buse JB, Cushman WC, Genuth S, Ismail-Beigi F, Grimm RH, Jr., Probstfield JL, Simons-Morton DG, Friedewald WT. Effects of intensive glucose lowering in type 2 diabetes. N Engl J Med 2008; 358:2545-2559
  36. Gerstein HC, Miller ME, Ismail-Beigi F, Largay J, McDonald C, Lochnan HA, Booth GL, ACCORD STUDY Group. Effects of intensive glycaemic control on ischaemic heart disease: analysis of data from the randomised, controlled ACCORD trial. Lancet 2014; 384:1936-1941
  37. ACCORD STUDY Group. Nine-Year Effects of 3.7 Years of Intensive Glycemic Control on Cardiovascular Outcomes. Diabetes Care 2016; 39:701-708
  38. Advance Collaborative Group, Patel A, MacMahon S, Chalmers J, Neal B, Billot L, Woodward M, Marre M, Cooper M, Glasziou P, Grobbee D, Hamet P, Harrap S, Heller S, Liu L, Mancia G, Mogensen CE, Pan C, Poulter N, Rodgers A, Williams B, Bompoint S, de Galan BE, Joshi R, Travert F. Intensive blood glucose control and vascular outcomes in patients with type 2 diabetes. N Engl J Med 2008; 358:2560-2572
  39. Zoungas S, Chalmers J, Neal B, Billot L, Li Q, Hirakawa Y, Arima H, Monaghan H, Joshi R, Colagiuri S, Cooper ME, Glasziou P, Grobbee D, Hamet P, Harrap S, Heller S, Lisheng L, Mancia G, Marre M, Matthews DR, Mogensen CE, Perkovic V, Poulter N, Rodgers A, Williams B, MacMahon S, Patel A, Woodward M, Group A-OC. Follow-up of blood-pressure lowering and glucose control in type 2 diabetes. N Engl J Med 2014; 371:1392-1406
  40. Duckworth W, Abraira C, Moritz T, Reda D, Emanuele N, Reaven PD, Zieve FJ, Marks J, Davis SN, Hayward R, Warren SR, Goldman S, McCarren M, Vitek ME, Henderson WG, Huang GD, Investigators V. Glucose control and vascular complications in veterans with type 2 diabetes. N Engl J Med 2009; 360:129-139
  41. Hayward RA, Reaven PD, Wiitala WL, Bahn GD, Reda DJ, Ge L, McCarren M, Duckworth WC, Emanuele NV, Investigators V. Follow-up of glycemic control and cardiovascular outcomes in type 2 diabetes. N Engl J Med2015; 372:2197-2206
  42. Reaven PD, Emanuele NV, Wiitala WL, Bahn GD, Reda DJ, McCarren M, Duckworth WC, Hayward RA, Investigators V. Intensive Glucose Control in Patients with Type 2 Diabetes - 15-Year Follow-up. N Engl J Med2019; 380:2215-2224
  43. Buehler AM, Cavalcanti AB, Berwanger O, Figueiro M, Laranjeira LN, Zazula AD, Kioshi B, Bugano DG, Santucci E, Sbruzzi G, Guimaraes HP, Carvalho VO, Bordin SA. Effect of tight blood glucose control versus conventional control in patients with type 2 diabetes mellitus: a systematic review with meta-analysis of randomized controlled trials. Cardiovasc Ther 2013; 31:147-160
  44. Control Group, Turnbull FM, Abraira C, Anderson RJ, Byington RP, Chalmers JP, Duckworth WC, Evans GW, Gerstein HC, Holman RR, Moritz TE, Neal BC, Ninomiya T, Patel AA, Paul SK, Travert F, Woodward M. Intensive glucose control and macrovascular outcomes in type 2 diabetes. Diabetologia 2009; 52:2288-2298
  45. Effect of intensive blood-glucose control with metformin on complications in overweight patients with type 2 diabetes (UKPDS 34). UK Prospective Diabetes Study (UKPDS) Group. Lancet 1998; 352:854-865
  46. Kooy A, de Jager J, Lehert P, Bets D, Wulffele MG, Donker AJ, Stehouwer CD. Long-term effects of metformin on metabolism and microvascular and macrovascular disease in patients with type 2 diabetes mellitus. Arch Intern Med 2009; 169:616-625
  47. Hong J, Zhang Y, Lai S, Lv A, Su Q, Dong Y, Zhou Z, Tang W, Zhao J, Cui L, Zou D, Wang D, Li H, Liu C, Wu G, Shen J, Zhu D, Wang W, Shen W, Ning G. Effects of metformin versus glipizide on cardiovascular outcomes in patients with type 2 diabetes and coronary artery disease. Diabetes Care 2013; 36:1304-1311
  48. Goldberg RB, Orchard TJ, Crandall JP, Boyko EJ, Budoff M, Dabelea D, Gadde KM, Knowler WC, Lee CG, Nathan DM, Watson K, Temprosa M. Effects of Long-term Metformin and Lifestyle Interventions on Cardiovascular Events in the Diabetes Prevention Program and Its Outcome Study. Circulation 2022; 145:1632-1641
  49. Goldberg RB, Aroda VR, Bluemke DA, Barrett-Connor E, Budoff M, Crandall JP, Dabelea D, Horton ES, Mather KJ, Orchard TJ, Schade D, Watson K, Temprosa M. Effect of Long-Term Metformin and Lifestyle in the Diabetes Prevention Program and Its Outcome Study on Coronary Artery Calcium. Circulation 2017; 136:52-64
  50. Fitch K, Abbara S, Lee H, Stavrou E, Sacks R, Michel T, Hemphill L, Torriani M, Grinspoon S. Effects of lifestyle modification and metformin on atherosclerotic indices among HIV-infected patients with the metabolic syndrome. AIDS 2012; 26:587-597
  51. Rosenstock J, Kahn SE, Johansen OE, Zinman B, Espeland MA, Woerle HJ, Pfarr E, Keller A, Mattheus M, Baanstra D, Meinicke T, George JT, von Eynatten M, McGuire DK, Marx N. Effect of Linagliptin vs Glimepiride on Major Adverse Cardiovascular Outcomes in Patients With Type 2 Diabetes: The CAROLINA Randomized Clinical Trial. JAMA 2019;
  52. Navigator Study Group, Holman RR, Haffner SM, McMurray JJ, Bethel MA, Holzhauer B, Hua TA, Belenkov Y, Boolell M, Buse JB, Buckley BM, Chacra AR, Chiang FT, Charbonnel B, Chow CC, Davies MJ, Deedwania P, Diem P, Einhorn D, Fonseca V, Fulcher GR, Gaciong Z, Gaztambide S, Giles T, Horton E, Ilkova H, Jenssen T, Kahn SE, Krum H, Laakso M, Leiter LA, Levitt NS, Mareev V, Martinez F, Masson C, Mazzone T, Meaney E, Nesto R, Pan C, Prager R, Raptis SA, Rutten GE, Sandstroem H, Schaper F, Scheen A, Schmitz O, Sinay I, Soska V, Stender S, Tamas G, Tognoni G, Tuomilehto J, Villamil AS, Vozar J, Califf RM. Effect of nateglinide on the incidence of diabetes and cardiovascular events. N Engl J Med 2010; 362:1463-1476
  53. Dormandy JA, Charbonnel B, Eckland DJ, Erdmann E, Massi-Benedetti M, Moules IK, Skene AM, Tan MH, Lefebvre PJ, Murray GD, Standl E, Wilcox RG, Wilhelmsen L, Betteridge J, Birkeland K, Golay A, Heine RJ, Koranyi L, Laakso M, Mokan M, Norkus A, Pirags V, Podar T, Scheen A, Scherbaum W, Schernthaner G, Schmitz O, Skrha J, Smith U, Taton J. Secondary prevention of macrovascular events in patients with type 2 diabetes in the PROactive Study (PROspective pioglitAzone Clinical Trial In macroVascular Events): a randomised controlled trial. Lancet 2005; 366:1279-1289
  54. Kernan WN, Viscoli CM, Furie KL, Young LH, Inzucchi SE, Gorman M, Guarino PD, Lovejoy AM, Peduzzi PN, Conwit R, Brass LM, Schwartz GG, Adams HP, Jr., Berger L, Carolei A, Clark W, Coull B, Ford GA, Kleindorfer D, O'Leary JR, Parsons MW, Ringleb P, Sen S, Spence JD, Tanne D, Wang D, Winder TR. Pioglitazone after Ischemic Stroke or Transient Ischemic Attack. N Engl J Med 2016; 374:1321-1331
  55. Vaccaro O, Masulli M, Nicolucci A, Bonora E, Del Prato S, Maggioni AP, Rivellese AA, Squatrito S, Giorda CB, Sesti G, Mocarelli P, Lucisano G, Sacco M, Signorini S, Cappellini F, Perriello G, Babini AC, Lapolla A, Gregori G, Giordano C, Corsi L, Buzzetti R, Clemente G, Di Cianni G, Iannarelli R, Cordera R, La Macchia O, Zamboni C, Scaranna C, Boemi M, Iovine C, Lauro D, Leotta S, Dall'Aglio E, Cannarsa E, Tonutti L, Pugliese G, Bossi AC, Anichini R, Dotta F, Di Benedetto A, Citro G, Antenucci D, Ricci L, Giorgino F, Santini C, Gnasso A, De Cosmo S, Zavaroni D, Vedovato M, Consoli A, Calabrese M, di Bartolo P, Fornengo P, Riccardi G. Effects on the incidence of cardiovascular events of the addition of pioglitazone versus sulfonylureas in patients with type 2 diabetes inadequately controlled with metformin (TOSCA.IT): a randomised, multicentre trial. Lancet Diabetes Endocrinol2017; 5:887-897
  56. Vaccaro O, Lucisano G, Masulli M, Bonora E, Del Prato S, Rivellese AA, Giorda CB, Mocarelli P, Squatrito S, Maggioni AP, Riccardi G, Nicolucci A. Cardiovascular Effects of Pioglitazone or Sulfonylureas According to Pretreatment Risk: Moving Toward Personalized Care. J Clin Endocrinol Metab 2019; 104:3296-3302
  57. Mazzone T, Meyer PM, Feinstein SB, Davidson MH, Kondos GT, D'Agostino RB, Sr., Perez A, Provost JC, Haffner SM. Effect of pioglitazone compared with glimepiride on carotid intima-media thickness in type 2 diabetes: a randomized trial. JAMA 2006; 296:2572-2581
  58. Pfutzner A, Marx N, Lubben G, Langenfeld M, Walcher D, Konrad T, Forst T. Improvement of cardiovascular risk markers by pioglitazone is independent from glycemic control: results from the pioneer study. J Am Coll Cardiol2005; 45:1925-1931
  59. Nissen SE, Nicholls SJ, Wolski K, Nesto R, Kupfer S, Perez A, Jure H, De Larochelliere R, Staniloae CS, Mavromatis K, Saw J, Hu B, Lincoff AM, Tuzcu EM. Comparison of pioglitazone vs glimepiride on progression of coronary atherosclerosis in patients with type 2 diabetes: the PERISCOPE randomized controlled trial. JAMA2008; 299:1561-1573
  60. Nissen SE, Wolski K. Effect of rosiglitazone on the risk of myocardial infarction and death from cardiovascular causes. N Engl J Med 2007; 356:2457-2471
  61. Singh S, Loke YK, Furberg CD. Long-term risk of cardiovascular events with rosiglitazone: a meta-analysis. JAMA 2007; 298:1189-1195
  62. Home PD, Pocock SJ, Beck-Nielsen H, Curtis PS, Gomis R, Hanefeld M, Jones NP, Komajda M, McMurray JJ. Rosiglitazone evaluated for cardiovascular outcomes in oral agent combination therapy for type 2 diabetes (RECORD): a multicentre, randomised, open-label trial. Lancet 2009; 373:2125-2135
  63. Home PD, Pocock SJ, Beck-Nielsen H, Gomis R, Hanefeld M, Jones NP, Komajda M, McMurray JJ. Rosiglitazone evaluated for cardiovascular outcomes--an interim analysis. N Engl J Med 2007; 357:28-38
  64. Mahaffey KW, Hafley G, Dickerson S, Burns S, Tourt-Uhlig S, White J, Newby LK, Komajda M, McMurray J, Bigelow R, Home PD, Lopes RD. Results of a reevaluation of cardiovascular outcomes in the RECORD trial. Am Heart J 2013; 166:240-249 e241
  65. Bach RG, Brooks MM, Lombardero M, Genuth S, Donner TW, Garber A, Kennedy L, Monrad ES, Pop-Busui R, Kelsey SF, Frye RL. Rosiglitazone and outcomes for patients with diabetes mellitus and coronary artery disease in the Bypass Angioplasty Revascularization Investigation 2 Diabetes (BARI 2D) trial. Circulation 2013; 128:785-794
  66. Feingold KR. Oral and Injectable (Non-Insulin) Pharmacological Agents for Type 2 Diabetes. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, Dungan K, Grossman A, Hershman JM, Kaltsas G, Koch C, Kopp P, Korbonits M, McLachlan R, Morley JE, New M, Perreault L, Purnell J, Rebar R, Singer F, Trence DL, Vinik A, Wilson DP, eds. Endotext. South Dartmouth (MA)2022.
  67. Scirica BM, Bhatt DL, Braunwald E, Steg PG, Davidson J, Hirshberg B, Ohman P, Frederich R, Wiviott SD, Hoffman EB, Cavender MA, Udell JA, Desai NR, Mosenzon O, McGuire DK, Ray KK, Leiter LA, Raz I. Saxagliptin and cardiovascular outcomes in patients with type 2 diabetes mellitus. N Engl J Med 2013; 369:1317-1326
  68. Scirica BM, Braunwald E, Raz I, Cavender MA, Morrow DA, Jarolim P, Udell JA, Mosenzon O, Im K, Umez-Eronini AA, Pollack PS, Hirshberg B, Frederich R, Lewis BS, McGuire DK, Davidson J, Steg PG, Bhatt DL. Heart failure, saxagliptin, and diabetes mellitus: observations from the SAVOR-TIMI 53 randomized trial. Circulation2014; 130:1579-1588
  69. White WB, Cannon CP, Heller SR, Nissen SE, Bergenstal RM, Bakris GL, Perez AT, Fleck PR, Mehta CR, Kupfer S, Wilson C, Cushman WC, Zannad F. Alogliptin after acute coronary syndrome in patients with type 2 diabetes. N Engl J Med 2013; 369:1327-1335
  70. Zannad F, Cannon CP, Cushman WC, Bakris GL, Menon V, Perez AT, Fleck PR, Mehta CR, Kupfer S, Wilson C, Lam H, White WB. Heart failure and mortality outcomes in patients with type 2 diabetes taking alogliptin versus placebo in EXAMINE: a multicentre, randomised, double-blind trial. Lancet 2015; 385:2067-2076
  71. Green JB, Bethel MA, Armstrong PW, Buse JB, Engel SS, Garg J, Josse R, Kaufman KD, Koglin J, Korn S, Lachin JM, McGuire DK, Pencina MJ, Standl E, Stein PP, Suryawanshi S, Van de Werf F, Peterson ED, Holman RR. Effect of Sitagliptin on Cardiovascular Outcomes in Type 2 Diabetes. N Engl J Med 2015; 373:232-242
  72. Rosenstock J, Perkovic V, Johansen OE, Cooper ME, Kahn SE, Marx N, Alexander JH, Pencina M, Toto RD, Wanner C, Zinman B, Woerle HJ, Baanstra D, Pfarr E, Schnaidt S, Meinicke T, George JT, von Eynatten M, McGuire DK. Effect of Linagliptin vs Placebo on Major Cardiovascular Events in Adults With Type 2 Diabetes and High Cardiovascular and Renal Risk: The CARMELINA Randomized Clinical Trial. JAMA 2018;
  73. Zinman B, Wanner C, Lachin JM, Fitchett D, Bluhmki E, Hantel S, Mattheus M, Devins T, Johansen OE, Woerle HJ, Broedl UC, Inzucchi SE. Empagliflozin, Cardiovascular Outcomes, and Mortality in Type 2 Diabetes. N Engl J Med 2015; 373:2117-2128
  74. Fitchett D, Inzucchi SE, Cannon CP, McGuire DK, Scirica BM, Johansen OE, Sambevski S, Kaspers S, Pfarr E, George JT, Zinman B. Empagliflozin Reduced Mortality and Hospitalization for Heart Failure Across the Spectrum of Cardiovascular Risk in the EMPA-REG OUTCOME Trial. Circulation 2019; 139:1384-1395
  75. Fitchett D, Butler J, van de Borne P, Zinman B, Lachin JM, Wanner C, Woerle HJ, Hantel S, George JT, Johansen OE, Inzucchi SE. Effects of empagliflozin on risk for cardiovascular death and heart failure hospitalization across the spectrum of heart failure risk in the EMPA-REG OUTCOME(R) trial. Eur Heart J 2018; 39:363-370
  76. Neal B, Perkovic V, Mahaffey KW, de Zeeuw D, Fulcher G, Erondu N, Shaw W, Law G, Desai M, Matthews DR. Canagliflozin and Cardiovascular and Renal Events in Type 2 Diabetes. N Engl J Med 2017; 377:644-657
  77. Perkovic V, Jardine MJ, Neal B, Bompoint S, Heerspink HJL, Charytan DM, Edwards R, Agarwal R, Bakris G, Bull S, Cannon CP, Capuano G, Chu PL, de Zeeuw D, Greene T, Levin A, Pollock C, Wheeler DC, Yavin Y, Zhang H, Zinman B, Meininger G, Brenner BM, Mahaffey KW. Canagliflozin and Renal Outcomes in Type 2 Diabetes and Nephropathy. N Engl J Med 2019; 380:2295-2306
  78. Jardine MJ, Zhou Z, Mahaffey KW, Oshima M, Agarwal R, Bakris G, Bajaj HS, Bull S, Cannon CP, Charytan DM, de Zeeuw D, Di Tanna GL, Greene T, Heerspink HJL, Levin A, Neal B, Pollock C, Qiu R, Sun T, Wheeler DC, Zhang H, Zinman B, Rosenthal N, Perkovic V. Renal, Cardiovascular, and Safety Outcomes of Canagliflozin by Baseline Kidney Function: A Secondary Analysis of the CREDENCE Randomized Trial. J Am Soc Nephrol 2020; 31:1128-1139
  79. Wiviott SD, Raz I, Bonaca MP, Mosenzon O, Kato ET, Cahn A, Silverman MG, Zelniker TA, Kuder JF, Murphy SA, Bhatt DL, Leiter LA, McGuire DK, Wilding JPH, Ruff CT, Gause-Nilsson IAM, Fredriksson M, Johansson PA, Langkilde AM, Sabatine MS. Dapagliflozin and Cardiovascular Outcomes in Type 2 Diabetes. N Engl J Med2018;
  80. Furtado RHM, Bonaca MP, Raz I, Zelniker TA, Mosenzon O, Cahn A, Kuder J, Murphy SA, Bhatt DL, Leiter LA, McGuire DK, Wilding JPH, Ruff CT, Nicolau JC, Gause-Nilsson IAM, Fredriksson M, Langkilde AM, Sabatine MS, Wiviott SD. Dapagliflozin and Cardiovascular Outcomes in Patients With Type 2 Diabetes Mellitus and Previous Myocardial Infarction. Circulation 2019; 139:2516-2527
  81. Cannon CP, Pratley R, Dagogo-Jack S, Mancuso J, Huyck S, Masiukiewicz U, Charbonnel B, Frederich R, Gallo S, Cosentino F, Shih WJ, Gantz I, Terra SG, Cherney DZI, McGuire DK. Cardiovascular Outcomes with Ertugliflozin in Type 2 Diabetes. N Engl J Med 2020; 383:1425-1435
  82. Cosentino F, Cannon CP, Cherney DZI, Masiukiewicz U, Pratley R, Dagogo-Jack S, Frederich R, Charbonnel B, Mancuso J, Shih WJ, Terra SG, Cater NB, Gantz I, McGuire DK. Efficacy of Ertugliflozin on Heart Failure-Related Events in Patients With Type 2 Diabetes Mellitus and Established Atherosclerotic Cardiovascular Disease: Results of the VERTIS CV Trial. Circulation 2020; 142:2205-2215
  83. Feingold KR. Oral and Injectable (Non-Insulin) Pharmacological Agents for the Treatment of Type 2 Diabetes. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Shah AS, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA)2022.
  84. Pfeffer MA, Claggett B, Diaz R, Dickstein K, Gerstein HC, Kober LV, Lawson FC, Ping L, Wei X, Lewis EF, Maggioni AP, McMurray JJ, Probstfield JL, Riddle MC, Solomon SD, Tardif JC. Lixisenatide in Patients with Type 2 Diabetes and Acute Coronary Syndrome. N Engl J Med 2015; 373:2247-2257
  85. Marso SP, Daniels GH, Brown-Frandsen K, Kristensen P, Mann JF, Nauck MA, Nissen SE, Pocock S, Poulter NR, Ravn LS, Steinberg WM, Stockner M, Zinman B, Bergenstal RM, Buse JB. Liraglutide and Cardiovascular Outcomes in Type 2 Diabetes. N Engl J Med 2016; 375:311-322
  86. Mann JFE, Fonseca V, Mosenzon O, Raz I, Goldman B, Idorn T, von Scholten BJ, Poulter NR. Effects of Liraglutide Versus Placebo on Cardiovascular Events in Patients With Type 2 Diabetes Mellitus and Chronic Kidney Disease. Circulation 2018; 138:2908-2918
  87. Verma S, Poulter NR, Bhatt DL, Bain SC, Buse JB, Leiter LA, Nauck MA, Pratley RE, Zinman B, Orsted DD, Monk Fries T, Rasmussen S, Marso SP. Effects of Liraglutide on Cardiovascular Outcomes in Patients With Type 2 Diabetes Mellitus With or Without History of Myocardial Infarction or Stroke. Circulation 2018; 138:2884-2894
  88. Marso SP, Bain SC, Consoli A, Eliaschewitz FG, Jodar E, Leiter LA, Lingvay I, Rosenstock J, Seufert J, Warren ML, Woo V, Hansen O, Holst AG, Pettersson J, Vilsboll T. Semaglutide and Cardiovascular Outcomes in Patients with Type 2 Diabetes. N Engl J Med 2016;
  89. Husain M, Birkenfeld AL, Donsmark M, Dungan K, Eliaschewitz FG, Franco DR, Jeppesen OK, Lingvay I, Mosenzon O, Pedersen SD, Tack CJ, Thomsen M, Vilsboll T, Warren ML, Bain SC. Oral Semaglutide and Cardiovascular Outcomes in Patients with Type 2 Diabetes. N Engl J Med 2019; 381:841-851
  90. Holman RR, Bethel MA, Mentz RJ, Thompson VP, Lokhnygina Y, Buse JB, Chan JC, Choi J, Gustavson SM, Iqbal N, Maggioni AP, Marso SP, Ohman P, Pagidipati NJ, Poulter N, Ramachandran A, Zinman B, Hernandez AF. Effects of Once-Weekly Exenatide on Cardiovascular Outcomes in Type 2 Diabetes. N Engl J Med 2017; 377:1228-1239
  91. Hernandez AF, Green JB, Janmohamed S, D'Agostino RB, Sr., Granger CB, Jones NP, Leiter LA, Rosenberg AE, Sigmon KN, Somerville MC, Thorpe KM, McMurray JJV, Del Prato S. Albiglutide and cardiovascular outcomes in patients with type 2 diabetes and cardiovascular disease (Harmony Outcomes): a double-blind, randomised placebo-controlled trial. Lancet 2018; 392:1519-1529
  92. Gerstein HC, Colhoun HM, Dagenais GR, Diaz R, Lakshmanan M, Pais P, Probstfield J, Riesmeyer JS, Riddle MC, Ryden L, Xavier D, Atisso CM, Dyal L, Hall S, Rao-Melacini P, Wong G, Avezum A, Basile J, Chung N, Conget I, Cushman WC, Franek E, Hancu N, Hanefeld M, Holt S, Jansky P, Keltai M, Lanas F, Leiter LA, Lopez-Jaramillo P, Cardona Munoz EG, Pirags V, Pogosova N, Raubenheimer PJ, Shaw JE, Sheu WH, Temelkova-Kurktschiev T. Dulaglutide and cardiovascular outcomes in type 2 diabetes (REWIND): a double-blind, randomised placebo-controlled trial. Lancet 2019; 394:121-130
  93. Gerstein HC, Hart R, Colhoun HM, Diaz R, Lakshmanan M, Botros FT, Probstfield J, Riddle MC, Ryden L, Atisso CM, Dyal L, Hall S, Avezum A, Basile J, Conget I, Cushman WC, Hancu N, Hanefeld M, Jansky P, Keltai M, Lanas F, Leiter LA, Lopez-Jaramillo P, Munoz EGC, Pogosova N, Raubenheimer PJ, Shaw JE, Sheu WH, Temelkova-Kurktschiev T. The effect of dulaglutide on stroke: an exploratory analysis of the REWIND trial. Lancet Diabetes Endocrinol 2020; 8:106-114
  94. Kristensen SL, Rorth R, Jhund PS, Docherty KF, Sattar N, Preiss D, Kober L, Petrie MC, McMurray JJV. Cardiovascular, mortality, and kidney outcomes with GLP-1 receptor agonists in patients with type 2 diabetes: a systematic review and meta-analysis of cardiovascular outcome trials. Lancet Diabetes Endocrinol 2019; 7:776-785
  95. Sposito AC, Berwanger O, de Carvalho LSF, Saraiva JFK. GLP-1RAs in type 2 diabetes: mechanisms that underlie cardiovascular effects and overview of cardiovascular outcome data. Cardiovasc Diabetol 2018; 17:157
  96. Sattar N, McGuire DK, Pavo I, Weerakkody GJ, Nishiyama H, Wiese RJ, Zoungas S. Tirzepatide cardiovascular event risk assessment: a pre-specified meta-analysis. Nat Med 2022; 28:591-598
  97. Chiasson JL, Josse RG, Gomis R, Hanefeld M, Karasik A, Laakso M. Acarbose treatment and the risk of cardiovascular disease and hypertension in patients with impaired glucose tolerance: the STOP-NIDDM trial. JAMA 2003; 290:486-494
  98. Yun P, Du AM, Chen XJ, Liu JC, Xiao H. Effect of Acarbose on Long-Term Prognosis in Acute Coronary Syndromes Patients with Newly Diagnosed Impaired Glucose Tolerance. J Diabetes Res 2016; 2016:1602083
  99. Holman RR, Coleman RL, Chan JCN, Chiasson JL, Feng H, Ge J, Gerstein HC, Gray R, Huo Y, Lang Z, McMurray JJ, Ryden L, Schroder S, Sun Y, Theodorakis MJ, Tendera M, Tucker L, Tuomilehto J, Wei Y, Yang W, Wang D, Hu D, Pan C. Effects of acarbose on cardiovascular and diabetes outcomes in patients with coronary heart disease and impaired glucose tolerance (ACE): a randomised, double-blind, placebo-controlled trial. Lancet Diabetes Endocrinol 2017; 5:877-886
  100. Gaziano JM, Cincotta AH, O'Connor CM, Ezrokhi M, Rutty D, Ma ZJ, Scranton RE. Randomized clinical trial of quick-release bromocriptine among patients with type 2 diabetes on overall safety and cardiovascular outcomes. Diabetes Care 2010; 33:1503-1508
  101. Younk LM, Davis SN. Evaluation of colesevelam hydrochloride for the treatment of type 2 diabetes. Expert Opin Drug Metab Toxicol 2012; 8:515-525
  102. The Lipid Research Clinics Coronary Primary Prevention Trial results. I. Reduction in incidence of coronary heart disease. JAMA 1984; 251:351-364
  103. The Lipid Research Clinics Coronary Primary Prevention Trial results. II. The relationship of reduction in incidence of coronary heart disease to cholesterol lowering. JAMA 1984; 251:365-374
  104. Investigators OT, Gerstein HC, Bosch J, Dagenais GR, Diaz R, Jung H, Maggioni AP, Pogue J, Probstfield J, Ramachandran A, Riddle MC, Ryden LE, Yusuf S. Basal insulin and cardiovascular and other outcomes in dysglycemia. N Engl J Med 2012; 367:319-328
  105. Origin Trial Investigators. Cardiovascular and Other Outcomes Postintervention With Insulin Glargine and Omega-3 Fatty Acids (ORIGINALE). Diabetes Care 2016; 39:709-716
  106. Marso SP, McGuire DK, Zinman B, Poulter NR, Emerson SS, Pieber TR, Pratley RE, Haahr PM, Lange M, Brown-Frandsen K, Moses A, Skibsted S, Kvist K, Buse JB. Efficacy and Safety of Degludec versus Glargine in Type 2 Diabetes. N Engl J Med 2017; 377:723-732
  107. Chaitman BR, Hardison RM, Adler D, Gebhart S, Grogan M, Ocampo S, Sopko G, Ramires JA, Schneider D, Frye RL. The Bypass Angioplasty Revascularization Investigation 2 Diabetes randomized trial of different treatment strategies in type 2 diabetes mellitus with stable ischemic heart disease: impact of treatment strategy on cardiac mortality and myocardial infarction. Circulation 2009; 120:2529-2540
  108. Bari D. Study Group, Frye RL, August P, Brooks MM, Hardison RM, Kelsey SF, MacGregor JM, Orchard TJ, Chaitman BR, Genuth SM, Goldberg SH, Hlatky MA, Jones TL, Molitch ME, Nesto RW, Sako EY, Sobel BE. A randomized trial of therapies for type 2 diabetes and coronary artery disease. N Engl J Med 2009; 360:2503-2515
  109. ElSayed NA, Aleppo G, Aroda VR, Bannuru RR, Brown FM, Bruemmer D, Collins BS, Hilliard ME, Isaacs D, Johnson EL, Kahan S, Khunti K, Leon J, Lyons SK, Perry ML, Prahalad P, Pratley RE, Seley JJ, Stanton RC, Gabbay RA, on behalf of the American Diabetes A. 9. Pharmacologic Approaches to Glycemic Treatment: Standards of Care in Diabetes-2023. Diabetes Care 2023; 46:S140-S157
  110. de Ferranti SD, de Boer IH, Fonseca V, Fox CS, Golden SH, Lavie CJ, Magge SN, Marx N, McGuire DK, Orchard TJ, Zinman B, Eckel RH. Type 1 diabetes mellitus and cardiovascular disease: a scientific statement from the American Heart Association and American Diabetes Association. Circulation 2014; 130:1110-1130
  111. Martin-Timon I, Sevillano-Collantes C, Segura-Galindo A, Del Canizo-Gomez FJ. Type 2 diabetes and cardiovascular disease: Have all risk factors the same strength? World J Diabetes 2014; 5:444-470
  112. Turner RC, Millns H, Neil HA, Stratton IM, Manley SE, Matthews DR, Holman RR. Risk factors for coronary artery disease in non-insulin dependent diabetes mellitus: United Kingdom Prospective Diabetes Study (UKPDS: 23). BMJ 1998; 316:823-828
  113. Hovingh GK, Rader DJ, Hegele RA. HDL re-examined. Curr Opin Lipidol 2015; 26:127-132
  114. Brunzell JD, Davidson M, Furberg CD, Goldberg RB, Howard BV, Stein JH, Witztum JL. Lipoprotein management in patients with cardiometabolic risk: consensus statement from the American Diabetes Association and the American College of Cardiology Foundation. Diabetes Care 2008; 31:811-822
  115. Ference BA, Kastelein JJP, Ray KK, Ginsberg HN, Chapman MJ, Packard CJ, Laufs U, Oliver-Williams C, Wood AM, Butterworth AS, Di Angelantonio E, Danesh J, Nicholls SJ, Bhatt DL, Sabatine MS, Catapano AL. Association of Triglyceride-Lowering LPL Variants and LDL-C-Lowering LDLR Variants With Risk of Coronary Heart Disease. JAMA 2019; 321:364-373
  116. Nordestgaard BG. Triglyceride-Rich Lipoproteins and Atherosclerotic Cardiovascular Disease: New Insights From Epidemiology, Genetics, and Biology. Circ Res 2016; 118:547-563
  117. Ganjali S, Dallinga-Thie GM, Simental-Mendia LE, Banach M, Pirro M, Sahebkar A. HDL functionality in type 1 diabetes. Atherosclerosis 2017; 267:99-109
  118. Ginsberg HN, MacCallum PR. The obesity, metabolic syndrome, and type 2 diabetes mellitus pandemic: Part I. Increased cardiovascular disease risk and the importance of atherogenic dyslipidemia in persons with the metabolic syndrome and type 2 diabetes mellitus. J Cardiometab Syndr 2009; 4:113-119
  119. Goldberg IJ. Clinical review 124: Diabetic dyslipidemia: causes and consequences. J Clin Endocrinol Metab2001; 86:965-971
  120. Krauss RM. Lipids and lipoproteins in patients with type 2 diabetes. Diabetes Care 2004; 27:1496-1504
  121. Wu L, Parhofer KG. Diabetic dyslipidemia. Metabolism 2014; 63:1469-1479
  122. Taskinen MR, Boren J. New insights into the pathophysiology of dyslipidemia in type 2 diabetes. Atherosclerosis2015; 239:483-495
  123. Feingold KR, Grunfeld C, Pang M, Doerrler W, Krauss RM. LDL subclass phenotypes and triglyceride metabolism in non-insulin-dependent diabetes. Arterioscler Thromb 1992; 12:1496-1502
  124. Feingold KR. Obesity and Dyslipidemia. In: De Groot LJ, Beck-Peccoz P, Chrousos G, Dungan K, Grossman A, Hershman JM, Koch C, McLachlan R, New M, Rebar R, Singer F, Vinik A, Weickert MO, eds. Endotext. South Dartmouth (MA)2023.
  125. Morgantini C, Natali A, Boldrini B, Imaizumi S, Navab M, Fogelman AM, Ferrannini E, Reddy ST. Anti-inflammatory and antioxidant properties of HDLs are impaired in type 2 diabetes. Diabetes 2011; 60:2617-2623
  126. Apro J, Tietge UJ, Dikkers A, Parini P, Angelin B, Rudling M. Impaired Cholesterol Efflux Capacity of High-Density Lipoprotein Isolated From Interstitial Fluid in Type 2 Diabetes Mellitus-Brief Report. Arterioscler Thromb Vasc Biol 2016; 36:787-791
  127. Manjunatha S, Distelmaier K, Dasari S, Carter RE, Kudva YC, Nair KS. Functional and proteomic alterations of plasma high density lipoproteins in type 1 diabetes mellitus. Metabolism 2016; 65:1421-1431
  128. Enkhmaa B, Anuurad E, Berglund L. Lipoprotein (a): impact by ethnicity and environmental and medical conditions. J Lipid Res 2016; 57:1111-1125
  129. Durrington PN, Schofield JD, Siahmansur T, Soran H. Lipoprotein (a): gene genie. Curr Opin Lipidol 2014; 25:289-296
  130. Gudbjartsson DF, Thorgeirsson G, Sulem P, Helgadottir A, Gylfason A, Saemundsdottir J, Bjornsson E, Norddahl GL, Jonasdottir A, Jonasdottir A, Eggertsson HP, Gretarsdottir S, Thorleifsson G, Indridason OS, Palsson R, Jonasson F, Jonsdottir I, Eyjolfsson GI, Sigurdardottir O, Olafsson I, Danielsen R, Matthiasson SE, Kristmundsdottir S, Halldorsson BV, Hreidarsson AB, Valdimarsson EM, Gudnason T, Benediktsson R, Steinthorsdottir V, Thorsteinsdottir U, Holm H, Stefansson K. Lipoprotein(a) Concentration and Risks of Cardiovascular Disease and Diabetes. J Am Coll Cardiol 2019; 74:2982-2994
  131. Ferrannini E, DeFronzo RA. Impact of glucose-lowering drugs on cardiovascular disease in type 2 diabetes. Eur Heart J 2015; 36:2288-2296
  132. Wulffele MG, Kooy A, de Zeeuw D, Stehouwer CD, Gansevoort RT. The effect of metformin on blood pressure, plasma cholesterol and triglycerides in type 2 diabetes mellitus: a systematic review. J Intern Med 2004; 256:1-14
  133. Ratner R, Goldberg R, Haffner S, Marcovina S, Orchard T, Fowler S, Temprosa M, Diabetes Prevention Program Research Group. Impact of intensive lifestyle and metformin therapy on cardiovascular disease risk factors in the diabetes prevention program. Diabetes Care 2005; 28:888-894
  134. Spanheimer R, Betteridge DJ, Tan MH, Ferrannini E, Charbonnel B. Long-term lipid effects of pioglitazone by baseline anti-hyperglycemia medication therapy and statin use from the PROactive experience (PROactive 14). Am J Cardiol 2009; 104:234-239
  135. Deeg MA, Buse JB, Goldberg RB, Kendall DM, Zagar AJ, Jacober SJ, Khan MA, Perez AT, Tan MH. Pioglitazone and rosiglitazone have different effects on serum lipoprotein particle concentrations and sizes in patients with type 2 diabetes and dyslipidemia. Diabetes Care 2007; 30:2458-2464
  136. Goldberg RB, Kendall DM, Deeg MA, Buse JB, Zagar AJ, Pinaire JA, Tan MH, Khan MA, Perez AT, Jacober SJ. A comparison of lipid and glycemic effects of pioglitazone and rosiglitazone in patients with type 2 diabetes and dyslipidemia. Diabetes Care 2005; 28:1547-1554
  137. Sanchez-Garcia A, Simental-Mendia M, Millan-Alanis JM, Simental-Mendia LE. Effect of sodium-glucose co-transporter 2 inhibitors on lipid profile: A systematic review and meta-analysis of 48 randomized controlled trials. Pharmacol Res 2020; 160:105068
  138. Lamos EM, Levitt DL, Munir KM. A review of dopamine agonist therapy in type 2 diabetes and effects on cardio-metabolic parameters. Prim Care Diabetes 2016; 10:60-65
  139. Holt RI, Barnett AH, Bailey CJ. Bromocriptine: old drug, new formulation and new indication. Diabetes Obes Metab 2010; 12:1048-1057
  140. Raskin P, Cincotta AH. Bromocriptine-QR therapy for the management of type 2 diabetes mellitus: developmental basis and therapeutic profile summary. Expert Rev Endocrinol Metab 2016; 11:113-148
  141. Feingold KR. Cholesterol Lowering Drugs. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, Dungan K, Grossman A, Hershman JM, Kaltsas G, Koch C, Kopp P, Korbonits M, McLachlan R, Morley JE, New M, Perreault L, Purnell J, Rebar R, Singer F, Trence DL, Vinik A, Wilson DP, eds. Endotext. South Dartmouth (MA)2021.
  142. Nauck MA, Meier JJ, Cavender MA, Abd El Aziz M, Drucker DJ. Cardiovascular Actions and Clinical Outcomes With Glucagon-Like Peptide-1 Receptor Agonists and Dipeptidyl Peptidase-4 Inhibitors. Circulation 2017; 136:849-870
  143. Kanbay M, Copur S, Siriopol D, Yildiz AB, Gaipov A, van Raalte DH, Tuttle KR. Effect of tirzepatide on blood pressure and lipids: A meta-analysis of randomized controlled trials. Diabetes Obes Metab 2023;
  144. Ginsberg HN. Diabetic dyslipidemia: basic mechanisms underlying the common hypertriglyceridemia and low HDL cholesterol levels. Diabetes 1996; 45 Suppl 3:S27-30
  145. Ginsberg HN, Zhang YL, Hernandez-Ono A. Metabolic syndrome: focus on dyslipidemia. Obesity (Silver Spring)2006; 14 Suppl 1:41S-49S
  146. Klop B, Elte JW, Cabezas MC. Dyslipidemia in obesity: mechanisms and potential targets. Nutrients 2013; 5:1218-1240
  147. Jorgensen AB, Frikke-Schmidt R, Nordestgaard BG, Tybjaerg-Hansen A. Loss-of-function mutations in APOC3 and risk of ischemic vascular disease. N Engl J Med 2014; 371:32-41
  148. Tg,Hdl Working Group of the Exome Sequencing Project, National Heart LungBlood, Institute, Crosby J, Peloso GM, Auer PL, Crosslin DR, Stitziel NO, Lange LA, Lu Y, Tang ZZ, Zhang H, Hindy G, Masca N, Stirrups K, Kanoni S, Do R, Jun G, Hu Y, Kang HM, Xue C, Goel A, Farrall M, Duga S, Merlini PA, Asselta R, Girelli D, Olivieri O, Martinelli N, Yin W, Reilly D, Speliotes E, Fox CS, Hveem K, Holmen OL, Nikpay M, Farlow DN, Assimes TL, Franceschini N, Robinson J, North KE, Martin LW, DePristo M, Gupta N, Escher SA, Jansson JH, Van Zuydam N, Palmer CN, Wareham N, Koch W, Meitinger T, Peters A, Lieb W, Erbel R, Konig IR, Kruppa J, Degenhardt F, Gottesman O, Bottinger EP, O'Donnell CJ, Psaty BM, Ballantyne CM, Abecasis G, Ordovas JM, Melander O, Watkins H, Orho-Melander M, Ardissino D, Loos RJ, McPherson R, Willer CJ, Erdmann J, Hall AS, Samani NJ, Deloukas P, Schunkert H, Wilson JG, Kooperberg C, Rich SS, Tracy RP, Lin DY, Altshuler D, Gabriel S, Nickerson DA, Jarvik GP, Cupples LA, Reiner AP, Boerwinkle E, Kathiresan S. Loss-of-function mutations in APOC3, triglycerides, and coronary disease. N Engl J Med 2014; 371:22-31
  149. Gaudet D, Brisson D, Tremblay K, Alexander VJ, Singleton W, Hughes SG, Geary RS, Baker BF, Graham MJ, Crooke RM, Witztum JL. Targeting APOC3 in the familial chylomicronemia syndrome. N Engl J Med 2014; 371:2200-2206
  150. Arca M, D'Erasmo L, Minicocci I. Familial combined hypolipidemia: angiopoietin-like protein-3 deficiency. Curr Opin Lipidol 2020; 31:41-48
  151. Khovidhunkit W, Kim MS, Memon RA, Shigenaga JK, Moser AH, Feingold KR, Grunfeld C. Effects of infection and inflammation on lipid and lipoprotein metabolism: mechanisms and consequences to the host. J Lipid Res2004; 45:1169-1196
  152. Lara-Castro C, Fu Y, Chung BH, Garvey WT. Adiponectin and the metabolic syndrome: mechanisms mediating risk for metabolic and cardiovascular disease. Curr Opin Lipidol 2007; 18:263-270
  153. Feingold KR, Grunfeld C. The acute phase response inhibits reverse cholesterol transport. J Lipid Res 2010; 51:682-684
  154. Feingold KR, Grunfeld C. Effect of inflammation on HDL structure and function. Curr Opin Lipidol 2022; 27:521-530
  155. Lu B, Moser A, Shigenaga JK, Grunfeld C, Feingold KR. The acute phase response stimulates the expression of angiopoietin like protein 4. Biochem Biophys Res Commun 2010; 391:1737-1741
  156. Feingold KR, Grunfeld C. The Effect of Inflammation and Infection on Lipids and Lipoproteins. In: De Groot LJ, Beck-Peccoz P, Chrousos G, Dungan K, Grossman A, Hershman JM, Koch C, McLachlan R, New M, Rebar R, Singer F, Vinik A, Weickert MO, eds. Endotext. South Dartmouth (MA)2019.
  157. Rosenblat M, Karry R, Aviram M. Paraoxonase 1 (PON1) is a more potent antioxidant and stimulant of macrophage cholesterol efflux, when present in HDL than in lipoprotein-deficient serum: relevance to diabetes. Atherosclerosis 2006; 187:74-81
  158. Ahima RS. Adipose tissue as an endocrine organ. Obesity (Silver Spring) 2006; 14 Suppl 5:242S-249S
  159. Christou GA, Kiortsis DN. Adiponectin and lipoprotein metabolism. Obes Rev 2013; 14:939-949
  160. Rashid S, Kastelein JJ. PCSK9 and resistin at the crossroads of the atherogenic dyslipidemia. Expert Rev Cardiovasc Ther 2013; 11:1567-1577
  161. Yu YH, Ginsberg HN. Adipocyte signaling and lipid homeostasis: sequelae of insulin-resistant adipose tissue. Circ Res 2005; 96:1042-1052
  162. Cholesterol Treatment Trialists, Collaborato, Kearney PM, Blackwell L, Collins R, Keech A, Simes J, Peto R, Armitage J, Baigent C. Efficacy of cholesterol-lowering therapy in 18,686 people with diabetes in 14 randomised trials of statins: a meta-analysis. Lancet 2008; 371:117-125
  163. Collins R, Armitage J, Parish S, Sleigh P, Peto R, Heart Protection Study Collaborative Group. MRC/BHF Heart Protection Study of cholesterol-lowering with simvastatin in 5963 people with diabetes: a randomised placebo-controlled trial. Lancet 2003; 361:2005-2016
  164. Heart Protection Study Collaborative Group. MRC/BHF Heart Protection Study of cholesterol lowering with simvastatin in 20,536 high-risk individuals: a randomised placebo-controlled trial. Lancet 2002; 360:7-22
  165. Wanner C, Krane V, Marz W, Olschewski M, Mann JF, Ruf G, Ritz E,German, Diabetes

            Dialysis Study, Investigators. Atorvastatin in patients with type 2 diabetes mellitus undergoing hemodialysis. N Engl J Med 2005; 353:238-248

  1. Colhoun HM, Betteridge DJ, Durrington PN, Hitman GA, Neil HA, Livingstone SJ, Thomason MJ, Mackness MI, Charlton-Menys V, Fuller JH. Primary prevention of cardiovascular disease with atorvastatin in type 2 diabetes in the Collaborative Atorvastatin Diabetes Study (CARDS): multicentre randomised placebo-controlled trial. Lancet2004; 364:685-696
  2. Nissen SE, Tuzcu EM, Schoenhagen P, Brown BG, Ganz P, Vogel RA, Crowe T, Howard G, Cooper CJ, Brodie B, Grines CL, DeMaria AN. Effect of intensive compared with moderate lipid-lowering therapy on progression of coronary atherosclerosis: a randomized controlled trial. JAMA 2004; 291:1071-1080
  3. Nissen SE, Nicholls SJ, Sipahi I, Libby P, Raichlen JS, Ballantyne CM, Davignon J, Erbel R, Fruchart JC, Tardif JC, Schoenhagen P, Crowe T, Cain V, Wolski K, Goormastic M, Tuzcu EM. Effect of very high-intensity statin therapy on regression of coronary atherosclerosis: the ASTEROID trial. JAMA 2006; 295:1556-1565
  4. Stegman B, Puri R, Cho L, Shao M, Ballantyne CM, Barter PJ, Chapman MJ, Erbel R, Libby P, Raichlen JS, Uno K, Kataoka Y, Nissen SE, Nicholls SJ. High-intensity statin therapy alters the natural history of diabetic coronary atherosclerosis: insights from SATURN. Diabetes Care 2014; 37:3114-3120
  5. Ahmed S, Cannon CP, Murphy SA, Braunwald E. Acute coronary syndromes and diabetes: Is intensive lipid lowering beneficial? Results of the PROVE IT-TIMI 22 trial. Eur Heart J 2006; 27:2323-2329
  6. Cannon CP, Braunwald E, McCabe CH, Rader DJ, Rouleau JL, Belder R, Joyal SV, Hill KA, Pfeffer MA, Skene AM, Pravastatin or Atorvastatin E, Infection Therapy-Thrombolysis in Myocardial Infarction I. Intensive versus moderate lipid lowering with statins after acute coronary syndromes. N Engl J Med 2004; 350:1495-1504
  7. LaRosa JC, Grundy SM, Waters DD, Shear C, Barter P, Fruchart JC, Gotto AM, Greten H, Kastelein JJ, Shepherd J, Wenger NK, Treating to New Targets I. Intensive lipid lowering with atorvastatin in patients with stable coronary disease. N Engl J Med 2005; 352:1425-1435
  8. Shepherd J, Barter P, Carmena R, Deedwania P, Fruchart JC, Haffner S, Hsia J, Breazna A, LaRosa J, Grundy S, Waters D. Effect of lowering LDL cholesterol substantially below currently recommended levels in patients with coronary heart disease and diabetes: the Treating to New Targets (TNT) study. Diabetes Care 2006; 29:1220-1226
  9. Pedersen TR, Faergeman O, Kastelein JJ, Olsson AG, Tikkanen MJ, Holme I, Larsen ML, Bendiksen FS, Lindahl C, Szarek M, Tsai J, Incremental Decrease in End Points Through Aggressive Lipid Lowering Study Group. High-dose atorvastatin vs usual-dose simvastatin for secondary prevention after myocardial infarction: the IDEAL study: a randomized controlled trial. JAMA 2005; 294:2437-2445
  10. Cholesterol Treatment Trialists Colloboration, Baigent C, Blackwell L, Emberson J, Holland LE, Reith C, Bhala N, Peto R, Barnes EH, Keech A, Simes J, Collins R. Efficacy and safety of more intensive lowering of LDL cholesterol: a meta-analysis of data from 170,000 participants in 26 randomised trials. Lancet 2010; 376:1670-1681
  11. Keech A, Simes RJ, Barter P, Best J, Scott R, Taskinen MR, Forder P, Pillai A, Davis T, Glasziou P, Drury P, Kesaniemi YA, Sullivan D, Hunt D, Colman P, d'Emden M, Whiting M, Ehnholm C, Laakso M. Effects of long-term fenofibrate therapy on cardiovascular events in 9795 people with type 2 diabetes mellitus (the FIELD study): randomised controlled trial. Lancet 2005; 366:1849-1861
  12. Jun M, Foote C, Lv J, Neal B, Patel A, Nicholls SJ, Grobbee DE, Cass A, Chalmers J, Perkovic V. Effects of fibrates on cardiovascular outcomes: a systematic review and meta-analysis. Lancet 2010; 375:1875-1884
  13. Sacks FM, Carey VJ, Fruchart JC. Combination lipid therapy in type 2 diabetes. N Engl J Med 2010; 363:692-694; author reply 694-695
  14. Robins SJ, Collins D, Wittes JT, Papademetriou V, Deedwania PC, Schaefer EJ, McNamara JR, Kashyap ML, Hershman JM, Wexler LF, Rubins HB. Relation of gemfibrozil treatment and lipid levels with major coronary events: VA-HIT: a randomized controlled trial. JAMA 2001; 285:1585-1591
  15. Rubins HB, Robins SJ, Collins D, Nelson DB, Elam MB, Schaefer EJ, Faas FH, Anderson JW. Diabetes, plasma insulin, and cardiovascular disease: subgroup analysis from the Department of Veterans Affairs high-density lipoprotein intervention trial (VA-HIT). Arch Intern Med 2002; 162:2597-2604
  16. Koskinen P, Manttari M, Manninen V, Huttunen JK, Heinonen OP, Frick MH. Coronary heart disease incidence in NIDDM patients in the Helsinki Heart Study. Diabetes Care 1992; 15:820-825
  17. Effect of fenofibrate on progression of coronary-artery disease in type 2 diabetes: the Diabetes Atherosclerosis Intervention Study, a randomised study. Lancet 2001; 357:905-910
  18. Elkeles RS, Diamond JR, Poulter C, Dhanjil S, Nicolaides AN, Mahmood S, Richmond W, Mather H, Sharp P, Feher MD. Cardiovascular outcomes in type 2 diabetes. A double-blind placebo-controlled study of bezafibrate: the St. Mary's, Ealing, Northwick Park Diabetes Cardiovascular Disease Prevention (SENDCAP) Study. Diabetes Care 1998; 21:641-648
  19. Clofibrate and niacin in coronary heart disease. JAMA 1975; 231:360-381
  20. Canner PL, Furberg CD, Terrin ML, McGovern ME. Benefits of niacin by glycemic status in patients with healed myocardial infarction (from the Coronary Drug Project). Am J Cardiol 2005; 95:254-257
  21. Ouchi Y, Sasaki J, Arai H, Yokote K, Harada K, Katayama Y, Urabe T, Uchida Y, Hayashi M, Yokota N, Nishida H, Otonari T, Arai T, Sakuma I, Sakabe K, Yamamoto M, Kobayashi T, Oikawa S, Yamashita S, Rakugi H, Imai T, Tanaka S, Ohashi Y, Kuwabara M, Ito H. Ezetimibe Lipid-Lowering Trial on Prevention of Atherosclerotic Cardiovascular Disease in 75 or Older (EWTOPIA 75): A Randomized, Controlled Trial. Circulation 2019; 140:992-1003
  22. Nissen SE, Lincoff AM, Brennan D, Ray KK, Mason D, Kastelein JJP, Thompson PD, Libby P, Cho L, Plutzky J, Bays HE, Moriarty PM, Menon V, Grobbee DE, Louie MJ, Chen CF, Li N, Bloedon L, Robinson P, Horner M, Sasiela WJ, McCluskey J, Davey D, Fajardo-Campos P, Petrovic P, Fedacko J, Zmuda W, Lukyanov Y, Nicholls SJ. Bempedoic Acid and Cardiovascular Outcomes in Statin-Intolerant Patients. N Engl J Med 2023; 388:1353-1364
  23. Nissen SE, Menon V, Nicholls SJ, Brennan D, Laffin L, Ridker P, Ray KK, Mason D, Kastelein JJP, Cho L, Libby P, Li N, Foody J, Louie MJ, Lincoff AM. Bempedoic Acid for Primary Prevention of Cardiovascular Events in Statin-Intolerant Patients. JAMA 2023; 330:131-140
  24. Group AS, Ginsberg HN, Elam MB, Lovato LC, Crouse JR, 3rd, Leiter LA, Linz P, Friedewald WT, Buse JB, Gerstein HC, Probstfield J, Grimm RH, Ismail-Beigi F, Bigger JT, Goff DC, Jr., Cushman WC, Simons-Morton DG, Byington RP. Effects of combination lipid therapy in type 2 diabetes mellitus. N Engl J Med 2010; 362:1563-1574
  25. ACCORD Study Group, Group AES, Chew EY, Ambrosius WT, Davis MD, Danis RP, Gangaputra S, Greven CM, Hubbard L, Esser BA, Lovato JF, Perdue LH, Goff DC, Jr., Cushman WC, Ginsberg HN, Elam MB, Genuth S, Gerstein HC, Schubart U, Fine LJ. Effects of medical therapies on retinopathy progression in type 2 diabetes. N Engl J Med 2010; 363:233-244
  26. Keech AC, Mitchell P, Summanen PA, O'Day J, Davis TM, Moffitt MS, Taskinen MR, Simes RJ, Tse D, Williamson E, Merrifield A, Laatikainen LT, d'Emden MC, Crimet DC, O'Connell RL, Colman PG. Effect of fenofibrate on the need for laser treatment for diabetic retinopathy (FIELD study): a randomised controlled trial. Lancet 2007; 370:1687-1697
  27. Das Pradhan A, Glynn RJ, Fruchart JC, MacFadyen JG, Zaharris ES, Everett BM, Campbell SE, Oshima R, Amarenco P, Blom DJ, Brinton EA, Eckel RH, Elam MB, Felicio JS, Ginsberg HN, Goudev A, Ishibashi S, Joseph J, Kodama T, Koenig W, Leiter LA, Lorenzatti AJ, Mankovsky B, Marx N, Nordestgaard BG, Pall D, Ray KK, Santos RD, Soran H, Susekov A, Tendera M, Yokote K, Paynter NP, Buring JE, Libby P, Ridker PM. Triglyceride Lowering with Pemafibrate to Reduce Cardiovascular Risk. N Engl J Med 2022; 387:1923-1934
  28. Investigators AIM-HIGH, Boden WE, Probstfield JL, Anderson T, Chaitman BR, Desvignes-Nickens P, Koprowicz K, McBride R, Teo K, Weintraub W. Niacin in patients with low HDL cholesterol levels receiving intensive statin therapy. N Engl J Med 2011; 365:2255-2267
  29. Guyton JR, Slee AE, Anderson T, Fleg JL, Goldberg RB, Kashyap ML, Marcovina SM, Nash SD, O'Brien KD, Weintraub WS, Xu P, Zhao XQ, Boden WE. Relationship of lipoproteins to cardiovascular events: the AIM-HIGH Trial (Atherothrombosis Intervention in Metabolic Syndrome With Low HDL/High Triglycerides and Impact on Global Health Outcomes). J Am Coll Cardiol 2013; 62:1580-1584
  30. Hps Thrive Collaborative Group, Landray MJ, Haynes R, Hopewell JC, Parish S, Aung T, Tomson J, Wallendszus K, Craig M, Jiang L, Collins R, Armitage J. Effects of extended-release niacin with laropiprant in high-risk patients. The New England journal of medicine 2014; 371:203-212
  31. Cannon CP, Blazing MA, Giugliano RP, McCagg A, White JA, Theroux P, Darius H, Lewis BS, Ophuis TO, Jukema JW, De Ferrari GM, Ruzyllo W, De Lucca P, Im K, Bohula EA, Reist C, Wiviott SD, Tershakovec AM, Musliner TA, Braunwald E, Califf RMI. Ezetimibe Added to Statin Therapy after Acute Coronary Syndromes. N Engl J Med 2015; 372:2387-2397
  32. Bohula EA, Morrow DA, Giugliano RP, Blazing MA, He P, Park JG, Murphy SA, White JA, Kesaniemi YA, Pedersen TR, Brady AJ, Mitchel Y, Cannon CP, Braunwald E. Atherothrombotic Risk Stratification and Ezetimibe for Secondary Prevention. J Am Coll Cardiol 2017; 69:911-921
  33. Giugliano RP, Cannon CP, Blazing MA, Nicolau JC, Corbalan R, Spinar J, Park JG, White JA, Bohula EA, Braunwald E. Benefit of Adding Ezetimibe to Statin Therapy on Cardiovascular Outcomes and Safety in Patients With Versus Without Diabetes Mellitus: Results From IMPROVE-IT (Improved Reduction of Outcomes: Vytorin Efficacy International Trial). Circulation 2018; 137:1571-1582
  34. Lee YJ, Cho JY, You SC, Lee YH, Yun KH, Cho YH, Shin WY, Im SW, Kang WC, Park Y, Lee SY, Lee SJ, Hong SJ, Ahn CM, Kim BK, Ko YG, Choi D, Hong MK, Jang Y, Kim JS. Moderate-intensity statin with ezetimibe vs. high-intensity statin in patients with diabetes and atherosclerotic cardiovascular disease in the RACING trial. Eur Heart J 2023; 44:972-983
  35. Sabatine MS, Giugliano RP, Keech AC, Honarpour N, Wiviott SD, Murphy SA, Kuder JF, Wang H, Liu T, Wasserman SM, Sever PS, Pedersen TR. Evolocumab and Clinical Outcomes in Patients with Cardiovascular Disease. N Engl J Med 2017; 376:1713-1722
  36. Sabatine MS, Leiter LA, Wiviott SD, Giugliano RP, Deedwania P, De Ferrari GM, Murphy SA, Kuder JF, Gouni-Berthold I, Lewis BS, Handelsman Y, Pineda AL, Honarpour N, Keech AC, Sever PS, Pedersen TR. Cardiovascular safety and efficacy of the PCSK9 inhibitor evolocumab in patients with and without diabetes and the effect of evolocumab on glycaemia and risk of new-onset diabetes: a prespecified analysis of the FOURIER randomised controlled trial. Lancet Diabetes Endocrinol 2017;
  37. Sabatine MS, Leiter LA, Wiviott SD, Giugliano RP, Deedwania P, De Ferrari GM, Murphy SA, Kuder JF, Gouni-Berthold I, Lewis BS, Handelsman Y, Pineda AL, Honarpour N, Keech AC, Sever PS, Pedersen TR. Cardiovascular safety and efficacy of the PCSK9 inhibitor evolocumab in patients with and without diabetes and the effect of evolocumab on glycaemia and risk of new-onset diabetes: a prespecified analysis of the FOURIER randomised controlled trial. Lancet Diabetes Endocrinol 2017; 5:941-950
  38. Giugliano RP, Keech A, Murphy SA, Huber K, Tokgozoglu SL, Lewis BS, Ferreira J, Pineda AL, Somaratne R, Sever PS, Pedersen TR, Sabatine MS. Clinical Efficacy and Safety of Evolocumab in High-Risk Patients Receiving a Statin: Secondary Analysis of Patients With Low LDL Cholesterol Levels and in Those Already Receiving a Maximal-Potency Statin in a Randomized Clinical Trial. JAMA Cardiol 2017; 2:1385-1391
  39. Giugliano RP, Pedersen TR, Park JG, De Ferrari GM, Gaciong ZA, Ceska R, Toth K, Gouni-Berthold I, Lopez-Miranda J, Schiele F, Mach F, Ott BR, Kanevsky E, Pineda AL, Somaratne R, Wasserman SM, Keech AC, Sever PS, Sabatine MS. Clinical efficacy and safety of achieving very low LDL-cholesterol concentrations with the PCSK9 inhibitor evolocumab: a prespecified secondary analysis of the FOURIER trial. Lancet 2017; 390:1962-1971
  40. Schwartz GG, Steg PG, Szarek M, Bhatt DL, Bittner VA, Diaz R, Edelberg JM, Goodman SG, Hanotin C, Harrington RA, Jukema JW, Lecorps G, Mahaffey KW, Moryusef A, Pordy R, Quintero K, Roe MT, Sasiela WJ, Tamby JF, Tricoci P, White HD, Zeiher AM. Alirocumab and Cardiovascular Outcomes after Acute Coronary Syndrome. N Engl J Med 2018; 379:2097-2107
  41. Nicholls SJ, Puri R, Anderson T, Ballantyne CM, Cho L, Kastelein JJ, Koenig W, Somaratne R, Kassahun H, Yang J, Wasserman SM, Scott R, Ungi I, Podolec J, Ophuis AO, Cornel JH, Borgman M, Brennan DM, Nissen SE. Effect of Evolocumab on Progression of Coronary Disease in Statin-Treated Patients: The GLAGOV Randomized Clinical Trial. JAMA 2016; 316:2373-2384
  42. ORIGIN Trial Investigators OT, Bosch J, Gerstein HC, Dagenais GR, Diaz R, Dyal L, Jung H, Maggiono AP, Probstfield J, Ramachandran A, Riddle MC, Ryden LE, Yusuf S. n-3 fatty acids and cardiovascular outcomes in patients with dysglycemia. N Engl J Med 2012; 367:309-318
  43. Ascend Study Collaborative Group, Bowman L, Mafham M, Wallendszus K, Stevens W, Buck G, Barton J, Murphy K, Aung T, Haynes R, Cox J, Murawska A, Young A, Lay M, Chen F, Sammons E, Waters E, Adler A, Bodansky J, Farmer A, McPherson R, Neil A, Simpson D, Peto R, Baigent C, Collins R, Parish S, Armitage J. Effects of n-3 Fatty Acid Supplements in Diabetes Mellitus. N Engl J Med 2018; 379:1540-1550
  44. Feingold KR. Triglyceride Lowering Drugs. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, Dungan K, Grossman A, Hershman JM, Kaltsas G, Koch C, Kopp P, Korbonits M, McLachlan R, Morley JE, New M, Perreault L, Purnell J, Rebar R, Singer F, Trence DL, Vinik A, Wilson DP, eds. Endotext. South Dartmouth (MA)2021.
  45. Yokoyama M, Origasa H, Matsuzaki M, Matsuzawa Y, Saito Y, Ishikawa Y, Oikawa S, Sasaki J, Hishida H, Itakura H, Kita T, Kitabatake A, Nakaya N, Sakata T, Shimada K, Shirato K. Effects of eicosapentaenoic acid on major coronary events in hypercholesterolaemic patients (JELIS): a randomised open-label, blinded endpoint analysis. Lancet 2007; 369:1090-1098
  46. Saito Y, Yokoyama M, Origasa H, Matsuzaki M, Matsuzawa Y, Ishikawa Y, Oikawa S, Sasaki J, Hishida H, Itakura H, Kita T, Kitabatake A, Nakaya N, Sakata T, Shimada K, Shirato K. Effects of EPA on coronary artery disease in hypercholesterolemic patients with multiple risk factors: sub-analysis of primary prevention cases from the Japan EPA Lipid Intervention Study (JELIS). Atherosclerosis 2008; 200:135-140
  47. Bhatt DL, Steg PG, Miller M, Brinton EA, Jacobson TA, Ketchum SB, Doyle RT, Jr., Juliano RA, Jiao L, Granowitz C, Tardif JC, Ballantyne CM. Cardiovascular Risk Reduction with Icosapent Ethyl for Hypertriglyceridemia. N Engl J Med 2018;
  48. Mason RP, Libby P, Bhatt DL. Emerging Mechanisms of Cardiovascular Protection for the Omega-3 Fatty Acid Eicosapentaenoic Acid. Arterioscler Thromb Vasc Biol 2020; 40:1135-1147
  49. Nicholls SJ, Lincoff AM, Bash D, Ballantyne CM, Barter PJ, Davidson MH, Kastelein JJP, Koenig W, McGuire DK, Mozaffarian D, Pedersen TR, Ridker PM, Ray K, Karlson BW, Lundstrom T, Wolski K, Nissen SE. Assessment of omega-3 carboxylic acids in statin-treated patients with high levels of triglycerides and low levels of high-density lipoprotein cholesterol: Rationale and design of the STRENGTH trial. Clin Cardiol 2018; 41:1281-1288
  50. Nicholls SJ, Lincoff AM, Garcia M, Bash D, Ballantyne CM, Barter PJ, Davidson MH, Kastelein JJP, Koenig W, McGuire DK, Mozaffarian D, Ridker PM, Ray KK, Katona BG, Himmelmann A, Loss LE, Rensfeldt M, Lundstrom T, Agrawal R, Menon V, Wolski K, Nissen SE. Effect of High-Dose Omega-3 Fatty Acids vs Corn Oil on Major Adverse Cardiovascular Events in Patients at High Cardiovascular Risk: The STRENGTH Randomized Clinical Trial. JAMA 2020;
  51. Ridker PM, Rifai N, MacFadyen J, Glynn RJ, Jiao L, Steg PG, Miller M, Brinton EA, Jacobson TA, Tardif JC, Ballantyne CM, Mason RP, Bhatt DL. Effects of Randomized Treatment With Icosapent Ethyl and a Mineral Oil Comparator on Interleukin-1beta, Interleukin-6, C-Reactive Protein, Oxidized Low-Density Lipoprotein Cholesterol, Homocysteine, Lipoprotein(a), and Lipoprotein-Associated Phospholipase A2: A REDUCE-IT Biomarker Substudy. Circulation 2022; 146:372-379
  52. Goff ZD, Nissen SE. N-3 polyunsaturated fatty acids for cardiovascular risk. Curr Opin Cardiol 2022; 37:356-363
  53. Mason RP, Sherratt SCR, Eckel RH. Omega-3-fatty acids: Do they prevent cardiovascular disease? Best Pract Res Clin Endocrinol Metab 2023; 37:101681
  54. ElSayed NA, Aleppo G, Aroda VR, Bannuru RR, Brown FM, Bruemmer D, Collins BS, Das SR, Hilliard ME, Isaacs D, Johnson EL, Kahan S, Khunti K, Kosiborod M, Leon J, Lyons SK, Perry ML, Prahalad P, Pratley RE, Seley JJ, Stanton RC, Gabbay RA, on behalf of the American Diabetes A. 10. Cardiovascular Disease and Risk Management: Standards of Care in Diabetes-2023. Diabetes Care 2023; 46:S158-S190
  55. Grundy SM, Stone NJ, Bailey AL, Beam C, Birtcher KK, Blumenthal RS, Braun LT, de Ferranti S, Faiella-Tommasino J, Forman DE, Goldberg R, Heidenreich PA, Hlatky MA, Jones DW, Lloyd-Jones D, Lopez-Pajares N, Ndumele CE, Orringer CE, Peralta CA, Saseen JJ, Smith SC, Jr., Sperling L, Virani SS, Yeboah J. 2018 AHA/ACC/AACVPR/AAPA/ABC/ACPM/ADA/AGS/APhA/ASPC/NLA/PCNA Guideline on the Management of Blood Cholesterol: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. Circulation 2019; 139:e1082-e1143
  56. Jellinger PS, Handelsman Y, Rosenblit PD, Bloomgarden ZT, Fonseca VA, Garber AJ, Grunberger G, Guerin CK, Bell DSH, Mechanick JI, Pessah-Pollack R, Wyne K, Smith D, Brinton EA, Fazio S, Davidson M. American Association of Clinical Endocrinologists and American College of Endocrinology Guidelines for Management of Dyslipidemia and Prevention of Cardiovascular Disease. Endocr Pract 2017; 23:1-87
  57. Handelsman Y, Jellinger PS, Guerin CK, Bloomgarden ZT, Brinton EA, Budoff MJ, Davidson MH, Einhorn D, Fazio S, Fonseca VA, Garber AJ, Grunberger G, Krauss RM, Mechanick JI, Rosenblit PD, Smith DA, Wyne KL. Consensus Statement by the American Association of Clinical Endocrinologists and American College of Endocrinology on the Management of Dyslipidemia and Prevention of Cardiovascular Disease Algorithm - 2020 Executive Summary. Endocr Pract 2020; 26:1196-1224
  58. Mach F, Baigent C, Catapano AL, Koskinas KC, Casula M, Badimon L, Chapman MJ, De Backer GG, Delgado V, Ference BA, Graham IM, Halliday A, Landmesser U, Mihaylova B, Pedersen TR, Riccardi G, Richter DJ, Sabatine MS, Taskinen MR, Tokgozoglu L, Wiklund O. 2019 ESC/EAS Guidelines for the management of dyslipidaemias: lipid modification to reduce cardiovascular risk. Eur Heart J 2020; 41:111-188
  59. Marx N, Federici M, Schutt K, Muller-Wieland D, Ajjan RA, Antunes MJ, Christodorescu RM, Crawford C, Di Angelantonio E, Eliasson B, Espinola-Klein C, Fauchier L, Halle M, Herrington WG, Kautzky-Willer A, Lambrinou E, Lesiak M, Lettino M, McGuire DK, Mullens W, Rocca B, Sattar N. 2023 ESC Guidelines for the management of cardiovascular disease in patients with diabetes. Eur Heart J 2023;
  60. SCORE2-Diabetes Working Group. SCORE2-Diabetes: 10-year cardiovascular risk estimation in type 2 diabetes in Europe. Eur Heart J 2023; 44:2544-2556
  61. Feingold KR. Maximizing the benefits of cholesterol-lowering drugs. Curr Opin Lipidol 2019; 30:388-394
  62. Feingold KR, Chait A. Approach to patients with elevated low-density lipoprotein cholesterol levels. Best Pract Res Clin Endocrinol Metab 2023; 37:101658
  63. Evert AB, Dennison M, Gardner CD, Garvey WT, Lau KHK, MacLeod J, Mitri J, Pereira RF, Rawlings K, Robinson S, Saslow L, Uelmen S, Urbanski PB, Yancy WS, Jr. Nutrition Therapy for Adults With Diabetes or Prediabetes: A Consensus Report. Diabetes Care 2019; 42:731-754
  64. Feingold KR. The Effect of Diet on Cardiovascular Disease and Lipid and Lipoprotein Levels. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Shah AS, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA)2021.
  65. Look ARG, Wing RR, Bolin P, Brancati FL, Bray GA, Clark JM, Coday M, Crow RS, Curtis JM, Egan CM, Espeland MA, Evans M, Foreyt JP, Ghazarian S, Gregg EW, Harrison B, Hazuda HP, Hill JO, Horton ES, Hubbard VS, Jakicic JM, Jeffery RW, Johnson KC, Kahn SE, Kitabchi AE, Knowler WC, Lewis CE, Maschak-Carey BJ, Montez MG, Murillo A, Nathan DM, Patricio J, Peters A, Pi-Sunyer X, Pownall H, Reboussin D, Regensteiner JG, Rickman AD, Ryan DH, Safford M, Wadden TA, Wagenknecht LE, West DS, Williamson DF, Yanovski SZ. Cardiovascular effects of intensive lifestyle intervention in type 2 diabetes. N Engl J Med 2013; 369:145-154
  66. Estruch R, Ros E, Salas-Salvado J, Covas MI, Corella D, Aros F, Gomez-Gracia E, Ruiz-Gutierrez V, Fiol M, Lapetra J, Lamuela-Raventos RM, Serra-Majem L, Pinto X, Basora J, Munoz MA, Sorli JV, Martinez JA, Martinez-Gonzalez MA. Primary prevention of cardiovascular disease with a Mediterranean diet. N Engl J Med2013; 368:1279-1290
  67. Estruch R, Ros E, Salas-Salvado J, Covas MI, Corella D, Aros F, Gomez-Gracia E, Ruiz-Gutierrez V, Fiol M, Lapetra J, Lamuela-Raventos RM, Serra-Majem L, Pinto X, Basora J, Munoz MA, Sorli JV, Martinez JA, Fito M, Gea A, Hernan MA, Martinez-Gonzalez MA. Primary Prevention of Cardiovascular Disease with a Mediterranean Diet Supplemented with Extra-Virgin Olive Oil or Nuts. N Engl J Med 2018; 378:e34
  68. Estruch R, Martinez-Gonzalez MA, Corella D, Salas-Salvado J, Ruiz-Gutierrez V, Covas MI, Fiol M, Gomez-Gracia E, Lopez-Sabater MC, Vinyoles E, Aros F, Conde M, Lahoz C, Lapetra J, Saez G, Ros E. Effects of a Mediterranean-style diet on cardiovascular risk factors: a randomized trial. Ann Intern Med 2006; 145:1-11
  69. Delgado-Lista J, Alcala-Diaz JF, Torres-Pena JD, Quintana-Navarro GM, Fuentes F, Garcia-Rios A, Ortiz-Morales AM, Gonzalez-Requero AI, Perez-Caballero AI, Yubero-Serrano EM, Rangel-Zuniga OA, Camargo A, Rodriguez-Cantalejo F, Lopez-Segura F, Badimon L, Ordovas JM, Perez-Jimenez F, Perez-Martinez P, Lopez-Miranda J. Long-term secondary prevention of cardiovascular disease with a Mediterranean diet and a low-fat diet (CORDIOPREV): a randomised controlled trial. Lancet 2022; 399:1876-1885
  70. de Lorgeril M, Renaud S, Mamelle N, Salen P, Martin JL, Monjaud I, Guidollet J, Touboul P, Delaye J. Mediterranean alpha-linolenic acid-rich diet in secondary prevention of coronary heart disease. Lancet 1994; 343:1454-1459
  71. de Lorgeril M, Salen P, Martin JL, Monjaud I, Delaye J, Mamelle N. Mediterranean diet, traditional risk factors, and the rate of cardiovascular complications after myocardial infarction: final report of the Lyon Diet Heart Study. Circulation 1999; 99:779-785
  72. Adams TD, Gress RE, Smith SC, Halverson RC, Simper SC, Rosamond WD, Lamonte MJ, Stroup AM, Hunt SC. Long-term mortality after gastric bypass surgery. N Engl J Med 2007; 357:753-761
  73. Romeo S, Maglio C, Burza MA, Pirazzi C, Sjoholm K, Jacobson P, Svensson PA, Peltonen M, Sjostrom L, Carlsson LM. Cardiovascular events after bariatric surgery in obese subjects with type 2 diabetes. Diabetes Care2012; 35:2613-2617
  74. Sjostrom L, Peltonen M, Jacobson P, Sjostrom CD, Karason K, Wedel H, Ahlin S, Anveden A, Bengtsson C, Bergmark G, Bouchard C, Carlsson B, Dahlgren S, Karlsson J, Lindroos AK, Lonroth H, Narbro K, Naslund I, Olbers T, Svensson PA, Carlsson LM. Bariatric surgery and long-term cardiovascular events. JAMA 2012; 307:56-65
  75. Sheng B, Truong K, Spitler H, Zhang L, Tong X, Chen L. The Long-Term Effects of Bariatric Surgery on Type 2 Diabetes Remission, Microvascular and Macrovascular Complications, and Mortality: a Systematic Review and Meta-Analysis. Obes Surg 2017; 27:2724-2732
  76. Fisher DP, Johnson E, Haneuse S, Arterburn D, Coleman KJ, O'Connor PJ, O'Brien R, Bogart A, Theis MK, Anau J, Schroeder EB, Sidney S. Association Between Bariatric Surgery and Macrovascular Disease Outcomes in Patients With Type 2 Diabetes and Severe Obesity. JAMA 2018; 320:1570-1582
  77. Farmer A, Montori V, Dinneen S, Clar C. Fish oil in people with type 2 diabetes mellitus. Cochrane Database Syst Rev 2001:CD003205
  78. Preiss D, Sattar N. Statins and the risk of new-onset diabetes: a review of recent evidence. Curr Opin Lipidol2011; 22:460-466
  79. Sattar N, Preiss D, Murray HM, Welsh P, Buckley BM, de Craen AJ, Seshasai SR, McMurray JJ, Freeman DJ, Jukema JW, Macfarlane PW, Packard CJ, Stott DJ, Westendorp RG, Shepherd J, Davis BR, Pressel SL, Marchioli R, Marfisi RM, Maggioni AP, Tavazzi L, Tognoni G, Kjekshus J, Pedersen TR, Cook TJ, Gotto AM, Clearfield MB, Downs JR, Nakamura H, Ohashi Y, Mizuno K, Ray KK, Ford I. Statins and risk of incident diabetes: a collaborative meta-analysis of randomised statin trials. Lancet 2010; 375:735-742
  80. Erqou S, Lee CC, Adler AI. Statins and glycaemic control in individuals with diabetes: a systematic review and meta-analysis. Diabetologia 2014; 57:2444-2452
  81. Livingstone SJ, Looker HC, Akbar T, Betteridge DJ, Durrington PN, Hitman GA, Neil HA, Fuller JH, Colhoun HM. Effect of atorvastatin on glycaemia progression in patients with diabetes: an analysis from the Collaborative Atorvastatin in Diabetes Trial (CARDS). Diabetologia 2016; 59:299-306
  82. Chiu SW, Pratt CM, Feinn R, Chatterjee S. Proprotein Convertase Subtilisin/Kexin Type 9 Inhibitors and Ezetimibe on Risk of New-Onset Diabetes: A Systematic Review and Meta-Analysis of Large, Double-Blinded Randomized Controlled Trials. J Cardiovasc Pharmacol Ther 2020; 25:409-417
  83. Shah NP, McGuire DK, Cannon CP, Giugliano RP, Lokhnygina Y, Page CB, Tershakovec AM, Braunwald E, Blazing MA. Impact of Ezetimibe on New-Onset Diabetes: A Substudy of IMPROVE-IT. J Am Heart Assoc 2023; 12:e029593
  84. Kellick KA, Bottorff M, Toth PP, The National Lipid Association's Safety Task F. A clinician's guide to statin drug-drug interactions. J Clin Lipidol 2014; 8:S30-46
  85. Linz PE, Lovato LC, Byington RP, O'Connor PJ, Leiter LA, Weiss D, Force RW, Crouse JR, Ismail-Beigi F, Simmons DL, Papademetriou V, Ginsberg HN, Elam MB. Paradoxical reduction in HDL-C with fenofibrate and thiazolidinedione therapy in type 2 diabetes: the ACCORD Lipid Trial. Diabetes Care 2014; 37:686-693
  86. Ansquer JC, Foucher C, Rattier S, Taskinen MR, Steiner G. Fenofibrate reduces progression to microalbuminuria over 3 years in a placebo-controlled study in type 2 diabetes: results from the Diabetes Atherosclerosis Intervention Study (DAIS). Am J Kidney Dis 2005; 45:485-493
  87. Davis TM, Ting R, Best JD, Donoghoe MW, Drury PL, Sullivan DR, Jenkins AJ, O'Connell RL, Whiting MJ, Glasziou PP, Simes RJ, Kesaniemi YA, Gebski VJ, Scott RS, Keech AC, Event Lowering in Diabetes Study i. Effects of fenofibrate on renal function in patients with type 2 diabetes mellitus: the Fenofibrate Intervention and Event Lowering in Diabetes (FIELD) Study. Diabetologia 2011; 54:280-290
  88. Rajamani K, Colman PG, Li LP, Best JD, Voysey M, D'Emden MC, Laakso M, Baker JR, Keech AC. Effect of fenofibrate on amputation events in people with type 2 diabetes mellitus (FIELD study): a prespecified analysis of a randomised controlled trial. Lancet 2009; 373:1780-1788
  89. Aldridge MA, Ito MK. Colesevelam hydrochloride: a novel bile acid-binding resin. Ann Pharmacother 2001; 35:898-907
  90. Bays HE. Colesevelam hydrochloride added to background metformin therapy in patients with type 2 diabetes mellitus: a pooled analysis from 3 clinical studies. Endocr Pract 2011; 17:933-938
  91. Song WL, FitzGerald GA. Niacin, an old drug with a new twist. J Lipid Res 2013; 54:2586-2594
  92. Elam MB, Hunninghake DB, Davis KB, Garg R, Johnson C, Egan D, Kostis JB, Sheps DS, Brinton EA. Effect of niacin on lipid and lipoprotein levels and glycemic control in patients with diabetes and peripheral arterial disease: the ADMIT study: A randomized trial. Arterial Disease Multiple Intervention Trial. JAMA 2000; 284:1263-1270
  93. Grundy SM, Vega GL, McGovern ME, Tulloch BR, Kendall DM, Fitz-Patrick D, Ganda OP, Rosenson RS, Buse JB, Robertson DD, Sheehan JP, Diabetes Multicenter Research Group. Efficacy, safety, and tolerability of once-daily niacin for the treatment of dyslipidemia associated with type 2 diabetes: results of the assessment of diabetes control and evaluation of the efficacy of niaspan trial. Archives of internal medicine 2002; 162:1568-1576
  94. Weintraub H. Update on marine omega-3 fatty acids: management of dyslipidemia and current omega-3 treatment options. Atherosclerosis 2013; 230:381-389
  95. Khan SU, Rahman H, Okunrintemi V, Riaz H, Khan MS, Sattur S, Kaluski E, Lincoff AM, Martin SS, Blaha MJ. Association of Lowering Low-Density Lipoprotein Cholesterol With Contemporary Lipid-Lowering Therapies and Risk of Diabetes Mellitus: A Systematic Review and Meta-Analysis. J Am Heart Assoc 2019; 8:e011581
  96. Carugo S, Sirtori CR, Corsini A, Tokgozoglu L, Ruscica M. PCSK9 Inhibition and Risk of Diabetes: Should We Worry? Curr Atheroscler Rep 2022; 24:995-1004
  97. Wang X, Zhang Y, Tan H, Wang P, Zha X, Chong W, Zhou L, Fang F. Efficacy and safety of bempedoic acid for prevention of cardiovascular events and diabetes: a systematic review and meta-analysis. Cardiovasc Diabetol2020; 19:128
  98. Bays HE, Banach M, Catapano AL, Duell PB, Gotto AM, Jr., Laufs U, Leiter LA, Mancini GBJ, Ray KK, Bloedon LT, Sasiela WJ, Ye Z, Ballantyne CM. Bempedoic acid safety analysis: Pooled data from four phase 3 clinical trials. J Clin Lipidol 2020; 14:649-659 e646
  99. Jenkins DJ, Jones PJ, Lamarche B, Kendall CW, Faulkner D, Cermakova L, Gigleux I, Ramprasath V, de Souza R, Ireland C, Patel D, Srichaikul K, Abdulnour S, Bashyam B, Collier C, Hoshizaki S, Josse RG, Leiter LA, Connelly PW, Frohlich J. Effect of a dietary portfolio of cholesterol-lowering foods given at 2 levels of intensity of dietary advice on serum lipids in hyperlipidemia: a randomized controlled trial. JAMA 2011; 306:831-839
  100. Feingold KR. Utility of Advanced Lipoprotein Testing in Clinical Practice. In: De Groot LJ, Beck-Peccoz P, Chrousos G, Dungan K, Grossman A, Hershman JM, Koch C, McLachlan R, New M, Rebar R, Singer F, Vinik A, Weickert MO, eds. Endotext. South Dartmouth (MA)2023.
  101. Orringer CE, Jacobson TA, Maki KC. National Lipid Association Scientific Statement on the use of icosapent ethyl in statin-treated patients with elevated triglycerides and high or very-high ASCVD risk. J Clin Lipidol 2019; 13:860-872

 

 

 

Idiopathic Short Stature and Growth Failure of Unknown Etiology

ABSTRACT

 

Idiopathic short stature (ISS) is defined by a height standard deviation score (SDS) ≤ -2.25 (≤1.2nd percentile) in pediatric patients for whom diagnostic evaluation has excluded other causes of short stature, and growth hormone levels are above 10 nanograms per milliliter in response to physiologic/pharmacologic stimulation. This definition, while comprehensive, is static and does not consider the importance of longitudinal follow up of growth, and consideration of evolving causes of growth hormone deficiency in these children. Using three illustrative cases, we highlight the importance of longitudinal evaluation of children with short stature to establish a growth pattern, identifying evolving causes of GH deficiency which may not be apparent on the initial work up, as well as exclusion of non-growth hormone related factors that affect growth including malabsorptive diseases, familial short stature, constitutional delay in puberty, Turner's syndrome, and SHOX Gene haploinsufficiency to accurately make a diagnosis of growth failure of unknown etiology (GFUE). 

 

INTRODUCTION 

 

In the 1980s, data on growth outcomes on patients who received growth hormone treatment were reported in an observational study (1); this included children who had a normal peak growth hormone response to stimulation, a group that was termed “constitutional short stature”.  The term, idiopathic short stature (ISS), was later adopted to describe such patients (2).   Guidelines for the use of growth hormone in children published by the Pediatric Endocrine Society defined ISS by a height standard deviation score (SDS) ≤-2.25 (≤1.2nd percentile) in pediatric patients for whom diagnostic evaluation excludes other causes of short stature, and with growth hormone levels above 10 nanograms per milliliter in response to physiologic/pharmacologic stimulation (3). The evaluation of children with short stature includes observation over a prolonged period to establish a growth pattern as well as the exclusion of non-growth hormone related factors that affect growth (4, 5).  To monitor children's growth patterns in the United States, the Center for Disease Control (CDC) chart utilizes percentiles, with the 3rd and 95th percentiles serving as the defined limits. Across the globe, the World Health Organization (WHO) charts are used to track a child's growth progression, and the limits  are set at 2.3rd and 97.7th percentiles (5). There are wide variations in normal growth patterns and an even greater variety of conditions that manifest with growth abnormalities.  Various factors contribute to the etiology of short stature, and this includes normal variants of growth such as familial short stature, constitutional delay in growth and puberty, as well as systemic conditions such as malabsorptive diseases, Turner's syndrome, and SHOX Gene haploinsufficiency, among others (Table 1).  Diagnostic tests at initial presentation by pediatric endocrinologists focus on elucidating the underlying cause of poor growth (Table 2).  Globally, malnutrition remains the main cause of poor growth (5).

 

In 2003, the Food and Drug Administration (FDA) approved the use of growth hormone (GH) for children with ISS. Two decades later, growth hormone therapy for idiopathic short stature remains controversial. ISS is not universally accepted as an indication for treatment with GH. This medication was approved for ISS in US, Canada, and Latin America, but not in the European Union or Japan (6). Along with other investigators, we have expressed concerns about the definition of ISS (7). The Pediatric Endocrine Society consensus statement did not specifically exclude patients with normal variants of growth such as constitutional growth delay and familial short stature (3). The definition focuses on stature, rather than growth and growth failure, therefore making GH treatment of such patients open to criticism for its use as only a height enhancer. In addition, as we have previously argued, a child whose height decelerates from the 75 to the 25th percentile without any known cause would also be of concern even though the height would not fit the ISS definition (7).  If no cause is found after clinical, appropriate biochemical and radiologic evaluation, such a patient may have what we would prefer to term idiopathic growth failure (IGF). In as much as those initials are already in use for Insulin Like Growth Factor, we prefer the term GFUE, or growth failure of unknown etiology, thereby emphasizing the concern about growth failure, not just stature.

 

To illustrate the potential dilemmas in diagnosis, we present three patients with similar patterns of growth and a normal response to initial growth hormone stimulation testing with 2 provocative agents; each of these children may have been called ISS but demonstrate radically different outcomes. 

Table 1. Common Causes of Short Stature and Growth Failure

Normal variants of growth

 
 

Familial short stature

 

Constitutional delay in growth and puberty (CDGP)

Systemic disorders

 

Endocrine

Growth hormone deficiency

 

Hypothyroidism

Cortisol excess (endogenous or exogenous)

Non-endocrine

Malnutrition

 

Malabsorptive diseases

 

Genetic syndromes: Turner's syndrome, Noonan’s syndrome, Achondroplasia, SHOX gene haploinsufficiency

 

Chronic inflammatory conditions (e.g.: Inflammatory bowel diseases)

 

Chronic medical conditions (e.g.: asthma on inhaled steroids)

 

Table 2. Diagnostic Tests at Initial Investigation of Idiopathic Short Stature and Growth Failure of Unknown Etiology

CBC, ESR

Urinalysis

Basic metabolic panel- BUN, creatinine, electrolytes

Celiac screen

Karyotype

TSH, Free T4

IGF1, IGFBP3

Bone age x ray

 

PATIENT 1

 

A boy aged 12 years and 5 months was evaluated for poor growth; both the father and paternal uncle were “late bloomers”. The height was at the 6th percentile, weight at 9th percentile, and Body Mass Index or BMI (weight in Kg divided by the square of height in meters) was at 32nd percentile, and his growth velocity was 3.8 cm per year. Clinical examination indicated Tanner stage 1 pubertal development with testes volume of 2-3mL bilaterally. Laboratory tests performed by the referring pediatrician before endocrine consultation showed normal thyroid function tests, sedimentation rate, C-Reactive protein, and no thyroid autoantibodies. A radiological bone age study confirmed delayed skeletal maturation, with a bone age of 10 years.  No additional tests were recommended at the Pediatric endocrinology clinic, and he was observed clinically.

 

At 14 years of age, the patient showed no signs of puberty. Figure 1 shows his longitudinal growth during follow up.  His height was the 2nd percentile, weight at the 6th percentile, BMI at the 31st percentile, and growth velocity remained at 3.8 cm per year. Delayed puberty in males is defined as the absence of testicular growth at an age that is 2 to 2.5 SD later than the reference population, which usually is around 14 years of age. (7) Laboratory results revealed low IGF-1 levels (-1.8 SDS for age and gender), while LH levels were close to pubertal levels, and testosterone levels remained prepubertal. A growth hormone (GH) stimulation test with the provocative agents, arginine and clonidine, was conducted due to the declining growth velocity and the low IGF-1 levels. Upon stimulation, peak GH level was 15 ng/ml.  At 14 years and 6 months old, the patient's height was at the1st percentile and growth velocity had decreased to 1.9 cm per year. The laboratory tests at that visit showed that IGF-1 levels had increased to 226 ng/mL and testosterone was now 45.7 ng/dL, consistent with Tanner stage 2 pubic hair, and 4 ml testicular volume noted on examination.  A bone age study confirmed delayed skeletal maturation, with a bone age of 10 years and 6 months. Since the patient had not progressed in puberty at the age of 14 years and 6 months, testosterone priming at a dose of 100 mg intramuscularly, once in 4 weeks for 3 months was initiated. At 15 years of age, his growth velocity had accelerated to 16.4 cm per year and his height was at the 5th percentile. Laboratory tests performed at 15.5 years showed that testosterone level was 301 ng/dL with a LH of 0.57 mIU/ml and IGF-1 levels had increased to 340 ng/ml. A bone age study showed the bone age to be consistent with 14 years and 6 months old. At 16 years and 11 months of age, the patient's height had reached the 36th percentile. Pubertal development had advanced with Tanner stage 3-4 for pubic hair development and testicular volume of 15mL bilaterally.

 

Figure 1. Patient 1 growth chart: Longitudinal growth chart in patient 1 before and after sex hormone priming.

 

DISCUSSION PATIENT 1

 

This patient’s height at presentation met the criteria for the diagnosis of Idiopathic Short Stature (ISS) since the patient's height was below -2.25 SD (8). However, with longitudinal follow-up, patient 1 ultimately proved to have constitutional growth delay (Figure 1). The diagnostic approach employed for this patient centered on monitoring. Once testosterone priming was employed, puberty progressed, and he reached his expected adult height. Through a targeted endocrine evaluation and longitudinal monitoring, the patient's condition was successfully managed without growth hormone treatment. This case underscores the importance of judicious use of testing.

 

The onset of puberty differs between boys and girls. In girls, 95% manifest at least one sign of puberty by 13 years of age whereas boys should begin signs of puberty by 14 years of age. In patients with delayed puberty and slow growth, a careful family history may be consistent with constitutional delay of growth and puberty. There often may be a history of a family member who also did not begin puberty until late into the teenage years, as in the case with the proband’s father and uncle. These patients can be difficult to diagnose because they may appear to be slowing in growth as they cross growth percentiles around the time of the anticipated pubertal growth spurt. Patients with constitutional growth delay fall behind their peers initially but have increased growth velocity later when puberty progresses and most reach adequate adult heights. Patients with constitutional growth delay have delayed bone ages and do not require treatment with growth hormone.

 

PATIENT 2

 

A boy presents at 12 years 9 months of age with growth failure and short stature. The patient's height had declined from 25th percentile to 10th percentile over 1.5 years, BMI was 40th percentile, growth velocity was 3.8 cm per year, and he was prepubertal. The parental target height was 176.7 cm (50th percentile). Laboratory tests showed that the testosterone level was 12 ng/dL, IGF-1 level was 125ng/mL (-2.06 SD), and bone age study was consistent with 12 years of age. Because of the low growth velocity and IGF1 level, the patient had a growth hormone (GH) stimulation test with provocative agents, arginine, and clonidine. The results showed a peak GH level of 16.9 ng/ml. At 13.5 years old, the patient’s height further declined to be at -2.25 SD and growth velocity was 2.84 cm per year.  Figure 2 shows patient 2’s growth trajectory during follow-up.  His testicular volume was 6mL bilaterally while tests showed IGF-1 to be114 ng/mL (-2.21 SD), thyroid function tests were normal, early morning cortisol level was 15.2 ug/dL, prolactin was 10 ng/ml, testosterone level 79 ng/dL, the luteinizing hormone was 3.2 mIU/mL, and the follicle-stimulating hormone was 2.1 mIU/mL, all consistent with pubertal progression. As puberty progressed, the growth velocity did not increase leading to a decline in height standard deviation.  In the setting of this growth deceleration despite pubertal progression, a second growth hormone stimulation test with arginine and L-dopa was performed. The GH stimulation test revealed a peak GH level of 5.4 ng/ml. This low peak GH level is consistent with growth hormone deficiency (GHD).  On pituitary magnetic resonance imaging, the pituitary gland appeared normal but small. The patient was treated with daily recombinant human growth hormone for 4.8 years. He reached an adult height at the 90th percentile (1.3 SDS), slightly above mid parental height (Figure 2).

 

Figure 2. Patient 2 growth chart: Longitudinal growth chart in patient 2 before and after growth hormone therapy. GH denotes growth hormone. MPTH denotes mid-parental target height.

 

DISCUSSION PATIENT 2

 

This patient's referral to a pediatric endocrinologist stemmed from decline in height, subpar growth velocity considering stage of puberty, and low levels of insulin-like growth factor 1 (IGF-1). Based on his normal response to the first growth hormone (GH) stimulation test, the patient’s diagnosis was consistent with Idiopathic Short Stature (ISS). With continued monitoring, it was apparent that further growth deceleration occurred despite advent and progression of puberty. With an additional growth hormone (GH) stimulation test, the patient was ultimately diagnosed to have evolving growth hormone deficiency. GHD can be congenital or acquired, developing over time. The mechanism of idiopathic GHD might be due to a functional, transient decrease in somatomedin secretion insufficient to maintain growth as puberty sets in. Isolated GHD can be  diagnosed using a combination of measuring growth factors, bone age X-rays, and growth hormone stimulation testing (9). Long-term monitoring as children progress into puberty is essential to uncover these instances of evolving GHD (9). It is imperative to emphasize the significance of prolonged follow-up and repeated testing to ensure the accurate diagnosis of GHD, rather than classifying the patient under the category of ISS or constitutional delay in growth and puberty (CDGP). In this case, physicians employed a meticulous approach of closely observing the decline in height and height velocity as puberty advanced, alongside screening tests such as IGF-1 measurement and multiple growth hormone stimulation tests. This comprehensive methodology allowed for the accurate diagnosis of GHD.

 

Despite the recognition of their many flaws, including poor reproducibility, non-physiologic assessment, and practical considerations, provocative tests remain part of the comprehensive evaluation of growth and are essential for the diagnosis of GHD (10).  For GH stimulation testing, two agents that provoke GH secretion from the pituitary (L-dopa, clonidine, arginine, glucagon) are administered following an overnight fast. These provocative agents are not physiological and do not replicate normal secretory dynamics. In this patient with short stature, the clinical picture of low growth velocity as puberty progressed plus low IGF-1 levels are indicative of late, or evolving GHD. 

 

PATIENT 3

 

A girl aged 13 years and 6 months was referred to a pediatric endocrinology clinic to evaluate her short stature. The patient had a medical history of ADHD diagnosed at the age of 7 years, and multiple surgeries for strabismus in both eyes. Her mid-parental height was at the 5th percentile. Axillary hair developed at 11 years of age and breast development started around 10-11 years. At age 13.5 years, the height was at the 1st percentile (-2.27 SD), weight was at <1 percentile, and BMI was at the 6th percentile. Tanner stage of breast development was 3 and pubic hair had been shaved. The patient had small hands and feet, bilateral fifth finger clinodactyly, brachydactyly, and a slightly increased carrying angle. The bone age was12.5 years old with shortening and broadening of the metacarpals which raised concerns for hypochondroplasia. Chromosomal analysis showed a normal female karyotype, 46XX. SHOX gene analysis did not reveal mutations or deletions. This patient had a normal response to growth hormone stimulation test with provocative agents, arginine, and clonidine; peak growth hormone level was 18 ng/ml. The patient was referred to a medical geneticist to undergo further genetic testing to explain her short stature. Repeated chromosome analysis counting 50 cells, had normal female karyotype of 46XX excluding mosaic Turner Syndrome. The microarray results found gain in a 1.5 Mb copy at 10p15.3. The Fragile X test was negative, it did not reveal FMR1 CGG repeat expansions. A genetic testing panel for skeletal dysplasia investigated twenty-nine genes associated with skeletal dysplasia and the results showed no mutations or deletions. Whole exome sequencing did not reveal any variant in disease genes possibly associated with a short stature phenotype. In this patient with height -3 SDS and low mid parental height, treatment with growth hormone resulted in modest gains in height SDS. Figure 3 shows this patient’s growth before and after growth hormone therapy. 

 

Figure 3. Patient 3 growth chart: Longitudinal growth chart in patient 2 before and after growth hormone therapy. GH denotes growth hormone.

 

DISCUSSION PATIENT 3

 

The cause of this patient’s short stature was not apparent despite extensive testing; thus, the diagnosis remains Growth Failure of Unknown Etiology, (GFUE) or Idiopathic short stature (ISS). After repeated genetic testing and thorough whole exome sequencing the etiology of the patient’s growth failure remained elusive. Case three presents the roadmap for a proper diagnosis of GFUE/ ISS using diagnoses of exclusion. The patient's height declined below >2.25 SD score below the mean height for a given age and sex, and through extensive genetic testing, there were no abnormalities found. Through a rigorous process of extensive evaluations and the systematic elimination of alternative causes for short stature, including malabsorptive diseases, familial short stature, constitutional delay in puberty, Turner syndrome, and SHOX Gene haploinsufficiency, the patient ultimately received a diagnosis of GFUE or idiopathic short stature.

 

ISS or GFUE may exist as a primary diagnosis alone or may be subcategorized to include ISS with familial short stature or constitutional delay of growth and puberty. Patients with ISS are often predicted to have an adult height below that expected based on mid parental height. It is thought that GH may increase the adult height of patients with ISS by 3.5 to 7.5 cm when compared with controls (11).  GH-treated children with ISS have also been reported to achieve height gain similar to patients with isolated GHD (6). Additional considerations for treatment in conjunction with GH include gonadotropin- releasing hormone (GnRH) analogs and aromatase inhibitors. Both classes of medications seek to decrease bone age maturation while allowing the child to continue to grow. These agents are still considered experimental when used for improving growth and more studies are needed (12, 13).  IGF-1 has received some attention recently as a potential treatment for ISS. Currently, IGF-1 is only approved for those patients with proven IGF-1 deficiency (IGF-1 levels –3 SD from the mean) with heights below –3 SD from the mean for age and normal stimulated GH levels. There is a lack of prospective studies that show clear benefit of IGF-1 for ISS. IGF-1 has also been associated with potential side effects, including hypoglycemia, headaches, lymphoid tissue hypertrophy, and coarsening of facial features (4).  Further studies must be performed before IGF-1 can be considered a treatment option for GFUE/ISS.

 

The concept that short stature alone is a problem that must be treated often leads to patients seeking treatment for cosmetic, rather than medical reasons to attain an adult height the patient or family feel is appropriate. It is important to stress that short stature itself is not necessarily a medical problem. However, a pattern of growth consistent with growth failure (decreasing growth velocity, growth velocity consistently <50 percentile) regardless of height percentile should be evaluated further. If an underlying cause of growth failure cannot be found, we propose that it is appropriate to use a term such as “idiopathic growth failure” or “growth failure of unknown etiology” (GFUE) rather than “idiopathic short stature.” These terms take the focus off stature as the overriding problem, placing it into an element that all can agree needs investigation and, if persistent, needs intervention for its reversal. Height predictions always, but especially in the face of subnormal growth rates, are likely to be inaccurate and should be provided with a great deal of caution (5).  We believe that familial short stature and constitutional delay of growth and puberty should be removed from the current overall category of “ISS” and placed under the category “normal variant.”

 

A child should be labeled as GFUE/ISS only after an in depth evaluation for other etiologies has been performed (7). As Next Generation Sequencing (NGS) and other diagnostic techniques become established, the number of patients with unidentified causes for growth failure is likely to diminish.

 

CONCLUSION

 

The three illustrative cases above help to show the significance of comprehensive testing, thorough evaluations, and rigorous longitudinal monitoring, to exclude currently known causes of short stature. Every effort should be made to avoid any bias in referral and evaluation of short stature based on gender, ethnic or racial differences (14).  With these guidelines, two of the three patients initially presented with features aligned with the diagnostic criteria for growth failure of unknown etiology (GFUE)/ISS. Through subsequent testing and prolonged monitoring, alternative underlying diagnoses were identified: constitutional growth delay, and acquired, idiopathic, isolated growth hormone deficiency. In the third case, we present the roadmap for a proper diagnosis of GFUE/ ISS that is based on specific phenotypic characteristics. The diagnosis of ISS/GFUE should only be assigned after an exhaustive exploration of all diagnostic possibilities, ensuring that patients are provided with precise diagnoses and subsequently, if indicated, appropriate medication-based treatments to achieve height gain.

 

REFERENCES

 

  1. Gertner, J.M., et al., Prospective clinical trial of human growth hormone in short children without growth hormone deficiency. J Pediatr, 1984. 104(2): p. 172-6.
  2. Hintz, R.L., Growth hormone treatment of idiopathic short stature. Horm Res, 1996. 46(4-5): p. 208-14.
  3. Wilson, T.A., et al., Update of guidelines for the use of growth hormone in children: the Lawson Wilkins Pediatric Endocrinology Society Drug and Therapeutics Committee. J Pediatr, 2003. 143(4): p. 415-21.
  4. Grimberg, A., et al., Guidelines for Growth Hormone and Insulin-Like Growth Factor-I Treatment in Children and Adolescents: Growth Hormone Deficiency, Idiopathic Short Stature, and Primary Insulin-Like Growth Factor-I Deficiency. Horm Res Paediatr, 2016. 86(6): p. 361-397.
  5. Graber, E. and R. Rapaport, Growth and growth disorders in children and adolescents. Pediatr Ann, 2012. 41(4): p. e1-9.
  6. Child, C.J., et al., Height Gain and Safety Outcomes in Growth Hormone-Treated Children with Idiopathic Short Stature: Experience from a Prospective Observational Study. Horm Res Paediatr, 2019. 91(4): p. 241-251.
  7. Rapaport, R., J.M. Wit, and M.O. Savage, Growth failure: 'idiopathic' only after a detailed diagnostic evaluation. Endocr Connect, 2021. 10(3): p. R125-R138.
  8. Cohen, P., et al., Consensus statement on the diagnosis and treatment of children with idiopathic short stature: a summary of the Growth Hormone Research Society, the Lawson Wilkins Pediatric Endocrine Society, and the European Society for Paediatric Endocrinology Workshop. J Clin Endocrinol Metab, 2008. 93(11): p. 4210-7.
  9. Samuels, J.G., Chimatapu, S. N., Savage, M. O., Rapaport, R, Late-Onset Isolated Growth Hormone Deficiency. JCEM Case Reports, 2023. 1(2).
  10. Yau, M. and R. Rapaport, Growth Hormone Stimulation Testing: To Test or Not to Test? That Is One of the Questions. Front Endocrinol (Lausanne), 2022. 13: p. 902364.
  11. Allen DB, Cuttler L. Clinical practice. Short stature in childhood--challenges and choices. N Engl J Med. 2013 Mar 28;368(13):1220-8. doi: 10.1056/NEJMcp1213178. PMID: 23534561; PMCID: PMC5754004.
  12. Mauras, N., et al., Randomized Trial of Aromatase Inhibitors, Growth Hormone, or Combination in Pubertal Boys with Idiopathic, Short Stature. J Clin Endocrinol Metab, 2016. 101(12): p. 4984-4993.
  13. Carel, J.C., Management of short stature with GnRH agonist and co-treatment with growth hormone: a controversial issue. Mol Cell Endocrinol, 2006. 254-255: p. 226-33.
  14. Beliard, K., et al., Identifying and addressing disparities in the evaluation and treatment of children with growth hormone deficiency. Front Endocrinol (Lausanne), 2022. 13: p. 989404.

Lipoprotein (a)

ABSTRACT

 

Lipoprotein(a) is an apo B-containing lipoprotein which has a unique plasminogen-like apolipoprotein(a) attached to apo B-100. The levels of lipoprotein(a) are under strong genetic regulation and vary by several hundred-fold in the general population. Although the exact function of lipoprotein(a) is still a mystery, recent studies have shown that lipoprotein(a) has a causal role in atherosclerosis and its level has been shown to be a risk factor for atherosclerotic cardiovascular disease (ASCVD) and calcific aortic valvular disease. Measurement of lipoprotein(a) level and comparison between different assays are challenging due to differences in reporting units and the absence of a reference method. Various guidelines recommend measurement of lipoprotein(a) levels in order to define cardiovascular risk. Lifestyle modifications have a minimal effect in reducing lipoprotein(a) levels. Currently available lipid-lowering therapies also result in only modest reductions in lipoprotein(a) levels. At present, there is no approved pharmacological treatment option for lowering lipoprotein(a) levels. More potent lipoprotein(a)-lowering medications are under active investigation to prove that lowering lipoprotein(a) level reduces ASCVD events. Lipoprotein(a), therefore, remains a viable and attractive therapeutic target in ASCVD risk reduction.   

 

HISTORY

 

Lipoprotein(a) was first described by Kare Berg in 1963 (1) and later purified and characterized by Ehnholm and colleagues as part of human lipoproteins (2). Subsequent studies linked lipoprotein(a) to coronary artery disease (CAD) (3,4) and the threshold of 30 mg/dL was introduced (5). In 1987, amino acid sequences of apolipoprotein(a) [apo(a)], a major protein of lipoprotein(a), were identified and the LPA gene, encoding apo(a), was cloned (6,7). The sequence showed homology to plasminogen, a fibrinolytic proenzyme, but the protease domain was inactive (6). Apo(a) size polymorphism and its underlying molecular genetics were discovered and the inverse correlation between apo(a) isoform and lipoprotein(a) levels were demonstrated (8). Early observational and prospective studies showed higher levels of lipoprotein(a) in patients with CAD and myocardial infarction (MI) (4,5,9-12), but others did not (13,14). Lipoprotein(a) was later identified as a main carrier of oxidized phospholipids in the plasma (15). Results from large population-based studies along with Mendelian randomization studies in the past 2 decades have shown that lipoprotein(a) is a causal risk factor for atherosclerotic cardiovascular disease (ASCVD), especially MI and atherosclerotic stenosis (16-19). A surprise finding in 2013 also identified lipoprotein(a) as a key player in calcific aortic valvular disease (CAVD) (20). Currently, lipoprotein(a) is an active therapeutic target and results from ongoing phase 3 clinical studies will soon clarify the lipoprotein(a) hypothesis in ASCVD and CAVD.           

 

STRUCTURE AND ASSEMBLY

 

Lipoprotein(a) is an apoB-containing lipoprotein resembling low-density lipoprotein (LDL), but apolipoprotein B-100 is covalently linked to a unique glycoprotein, called apolipoprotein(a) or apo(a). Apo(a), encoded by the LPA gene, is structurally similar to plasminogen, an important protein involved in fibrinolysis. In vitro, apo(a) has been shown to inhibit fibrinolysis (21)

 

Apo(a) is present only in a subset of primates, including humans, Old World monkeys, and orangutans, but not in New World monkeys or other primates. The exception is the occurrence of an apo(a)-like protein in the hedgehog (22). A characteristic feature of apo(a) is the presence of loop-like structure stabilized by 3 internal disulfide bonds, called “kringles”. These kringle domains are triple-loop structures found in other coagulation factors, including plasminogen. While plasminogen has 5 kringle domains (KI, KII, KIII, KIV and KV) and one protease domain at the end, apo(a) has only KIV, KV and an inactive protease domain (Figure 1).

 

Figure 1. Structure of lipoprotein(a) (from www.familyheart.org).

 

In apo(a), the number of the fourth kringle domain is highly variable due to expansion and differentiation of KIV into 10 different types of KIV domains, called KIV1-KIV10. While KIV1 and KIV3-KIV10 are present as single copies, kringle IV type 2 (KIV2) are further expanded, resulting in multiallelic copy number variation (1 to >40 copies). This expansion of KIV2 leads to a size polymorphism of apo(a), ranging from 300-800 kDa (23). Circulating levels of lipoprotein(a) are determined by the number of KIV2 copies. A low number of KIV2 copies results in small apo(a) isoforms, which lead to efficient secretion and contribute to higher levels of lipoprotein(a). On the other hand, the high number of KIV2 copies leads to large apo(a) isoforms and low levels of circulating lipoprotein(a).     

 

Apo(a) is mainly synthesized in the liver. Newly formed apo(a) is then bound to apo B-100 of LDL to become lipoprotein(a). The assembly of lipoprotein(a) particles is a two-step process (24). The first step is the non-covalent docking of the lysine-binding site at the KIV7-KIV8 domains of apo(a) to the lysine residues at the N-terminus of apo B-100. The second step is the covalent binding through the disulfide bond between the 2 free cysteines in the KIV-9 of apo(a) and apo B-100 (25). The site of lipoprotein(a) assembly has been controversial whether it occurs intracellularly or extracellularly (23,25,26), but a recent study using a hepatocyte cell model suggests that the first non-covalent step occurs intracellularly, whereas the second covalent bond formation occurs extracellularly (27,28).

 

FUNCTION

 

The physiological function of lipoprotein(a) remains obscure (23). Earlier, it has been suggested that lipoprotein(a) may assist in wound healing (16,29). Lipoprotein(a) can interact with fibrin and other components of the extracellular matrix (30) via the lysine-binding sites in its kringle domains and it delivers cholesterol to the site of injury. Besides carrying cholesterol esters, free cholesterol, triglycerides, and phospholipids, lipoprotein(a) is the main carrier of oxidized phospholipids in the circulation (15). Lipoprotein(a) might also act as a carrier/scavenger for oxidized lipids since oxidized phospholipids and platelet-activating factor acetylhydrolase, an enzyme involved in hydrolyzing PAF, are found on lipoprotein(a) particles. In vitro, lipoprotein(a) can interfere with many steps of blood clotting and fibrinolysis (21), but the evidence for its thrombogenic role in vivo is less convincing (16).

 

In the Finnish population, loss-of-function variants in the LPA gene have been found (31). Although these subjects have very low levels of lipoprotein(a), no identifiable clinical abnormalities have been demonstrated (31). An increased risk of type 2 diabetes has been reported in those who have very low levels of lipoprotein(a) (32-36). Whether low levels of lipoprotein(a) are causally associated with type 2 diabetes is still unclear.

 

METABOLISM

 

Circulating levels of lipoprotein(a) are primarily determined by isoform size-dependent production rate and not catabolism (37). Fractional catabolic rates for lipoprotein(a) with short and long isoforms have been shown to be relatively indifferent (37). Liver is the main site of lipoprotein(a) catabolism, with a minor contribution from the kidney (26). Apo(a), apo B-100 and oxidized phospholipids carried on lipoprotein(a) may play a role in catabolism of lipoprotein(a) particles by acting as ligands for various receptors. A number of receptors have been shown to be associated with clearance of lipoprotein(a), including lipoprotein receptors, scavenger receptors, plasminogen receptors, Toll-like receptors (TLRs), and carbohydrate receptors or lectins (38). However, the relative contribution of these different receptors in lipoprotein(a) clearance remains to be explored. The lack of suitable animal models and the conflicting data on the role of certain receptors on the catabolism of lipoprotein(a) also make it difficult to conclude the definite role of these receptors. Following uptake into the cells, it has been shown that lipoprotein(a) dissociates into 2 components, with LDL being degraded in lysosomes and apo(a) being recycled to be re-secreted (39). It has been estimated that approximately 30% of apo(a) is recycled after lipoprotein(a) internalization into the cells (39), which could potentially contribute to the circulating level of lipoprotein(a).

 

GENETICS

 

Lipoprotein(a) levels are strongly determined by the genetic locus at the LPA gene on chromosome 6, which encodes apo(a). The LPA gene is closely related to the PLG gene, which encodes plasminogen. These 2 genes diverged about 40 million years ago with the loss of KI-KIII and the expansion and differentiation of KIV domain (23). The major genetic determinant is a copy number variation of the KIV2 repeat, which explains approximately 30-70% of the variability in lipoprotein(a) levels, depending on ethnicity (40). The apo(a) size is highly variable among individuals and depends on the number of KIV2 repeats in the LPA gene. Expression of a low number (10-22) of KIV2 repeats is characterized by small apo(a) isoforms and higher lipoprotein(a) levels, whereas a higher number (>23) of KIV2 repeats results in large apo(a) isoforms and lower lipoprotein(a) levels. Smaller apo(a) isoforms are associated with higher apo(a) production rates from the liver (41), whereas larger isoforms are more susceptible to proteosomal degradation in the endoplasmic reticulum (42). Subjects who carry a low number of KIV repeats have 4-5 times higher median lipoprotein(a) levels than those with a high number of KIV repeats (43).

 

Because every individual has 2 copies of this gene, 2 different isoforms may be present in plasma. In general, plasma levels of lipoprotein(a) are determined by the net production of apo(a) in each isoform. In Caucasians, with an increasing number of KIV repeats, and hence the larger apo(a) isoform, the frequency of the non-expressed allele is also increasing (44). As a result, the major isoform in circulation is mainly driven by the smaller ones. However, in about one-quarter of Caucasians, the larger allele is reported to be the dominant one (44) so the smaller allele is not always dominant (45). The frequency of the non-expressed alleles in Caucasians was highest in the mid-range whereas in African Americans, the non-expressed alleles were fairly distributed across apo(a) sizes (44).        

 

It is of note that among those who carry the same number of KIV-2 repeats, there is a wide range of variability in the lipoprotein(a) levels. This could be explained by the presence of other genetic variants beyond the KIV repeat polymorphism that are also known to influence lipoprotein(a) levels (23,43,45) with a variable impact among various populations. A large genome-wide association meta-analysis has identified a number of independent single nucleotide polymorphisms (SNPs) around the LPA gene which are significantly associated with lipoprotein(a) levels (46). Certain SNPs are functionally lipoprotein(a)-increasing, such as rs1800769 and rs1853021, whereas other SNPS, such as rs10455872 and rs3798220, have no functional effect, but they are in linkage disequilibrium with other small apo(a) isoforms or other variants associated with elevated lipoprotein(a) levels (47).  

 

On the contrary, other genetic variants of the LPA gene are associated with low levels of lipoprotein(a), such as the common splice variants, 4733G>A (48) and 4925G>A (49) in the KIV-2 repeat, and the missense variant rs41267813 (50). Other SNPs are null alleles which decrease lipoprotein(a) levels, such as rs41272114, rs41259144 and rs139145675. Individuals who carry these variants are found to be protected against the development of ASCVD.

 

Other genes, besides the LPA gene, have also been shown to influence lipoprotein(a) levels (45) although a meta-analysis has not confirmed the findings (51), suggesting that further studies with large sample sizes are required to replicate these findings. A recent genome-wide association study from the UK Biobank in almost 300,000 individuals has identified APOE, APOH, and CETP as additional loci that affect lipoprotein(a) levels (52).

 

Although lipoprotein(a) is under a strong genetic regulation, currently, there is no recommendation or advice to perform genetic testing for the LPA gene. Measuring lipoprotein(a) level in the circulation is considered sufficient since it reflects the overall genetic interaction of all variants.

 

LIPOPROTEIN(A) LEVEL

 

The distribution of lipoprotein(a) level in plasma is highly skewed with a tail toward higher levels as shown in Figure 2 (16,17,53). The range varies widely from <0.1 mg/dL to >300 mg/dL (<0.2 – 750 nmol/L). In people of European descent, 80% of the population have a serum level of lipoprotein(a) <40 mg/dL or 90 nmol/L (18). It is well known that plasma levels of lipoprotein(a) are different among various ethnicities, which are predominantly determined by lipoprotein(a) isoform size and other genetic variants in the LPA locus (45). Data from the UK Biobank showed that the mean lipoprotein(a) levels were lowest in Chinese individuals (16 nmol/L), followed by White (19 nmol/L), South Asian (31 nmol/L), and highest in Black individuals (75 nmol/L) (17). Data from subjects with various ethnicities have demonstrated that the level of lipoprotein(a) is slightly higher in women than in men (17,53-56), although other studies have shown a lack of difference between men and women (57,58).

 

Figure 2. Distribution of lipoprotein(a) in the Danish general population (from (16)).

 

Lipoprotein(a) level is fully expressed by the age of 2 years and the adult levels are usually achieved by 5 years (59). The level of lipoprotein(a) seems to be stable throughout life because it is under a strong genetic influence (60). Except for certain medical conditions, plasma lipoprotein(a) levels are relatively stable across the lifespan independent of lifestyle. A long-term study over a period of 15 years has shown that the overall absolute change in lipoprotein(a) levels is relatively modest but may be more pronounced in subjects with very high lipoprotein(a) levels (60). The level is also not affected by fasting (61).

 

Because lipoprotein(a) levels are predominantly determined by genetics, lifestyle modifications appear to have minimal effects (45). A low carbohydrate/high saturated fat diet was shown to result in a 15% decrease in lipoprotein(a) levels (62), whereas a decrease in saturated fat was associated with a 10-15% increase in lipoprotein(a) levels (63). Dietary changes may affect lipoprotein(a) levels in the opposite direction to LDL-C. Physical activity and exercise may affect lipoprotein(a) levels but the results are inconsistent and it may depend on the host, type, intensity, and duration (64).

 

As lipoprotein(a) is produced by the liver, a reduction in lipoprotein(a) levels is observed in liver disease (65,66). Liver transplantation has been shown to change apo(a) isoform to that of the donor with corresponding changes in lipoprotein(a) levels (67). In contrast, elevations of lipoprotein(a) levels are shown in nephrotic syndrome and chronic kidney diseases, either from impaired catabolism or increased hepatic production in response to protein loss in urine or in dialysis (68,69). A large increase is found among subjects who carry large apo(a) isoform sizes (70). Initiation of dialysis has no effect on lipoprotein(a) levels (71). Kidney transplantation can rapidly normalize lipoprotein(a) levels within several weeks (72-74).

 

Infection and inflammation also affect lipoprotein(a) levels. In acute and chronic inflammatory conditions, such as autoimmune diseases, lipoprotein(a) levels were increased (10,75), but in severe life-threatening conditions, such as sepsis and burns, lipoprotein(a) levels were decreased (76). Lipoprotein(a) levels increased during COVID infections, which might be responsible for increased thromboembolic events (77,78). The LPA gene contains interleukin-6 response elements (79) and elevated lipoprotein(a) levels could be decreased using interleukin-6 receptor blockade by tocilizumab (80). Following acute myocardial infarction, earlier results on the changes in lipoprotein(a) were conflicting, which could be due to differences in assay system (81,82).

 

Changes in endogenous sex hormones, including menopause and ovariectomy, have been found to have minimal effects on lipoprotein(a) levels (83,84), although a 27% increase after menopause has also been reported (85). Castration and orchidectomy have been shown to slightly increase lipoprotein(a) levels (86,87). Hormone replacement therapy in postmenopausal women reduces lipoprotein(a) levels by approximately 12-25% (85,88).

 

Longitudinal studies during pregnancy have shown that lipoprotein(a) levels are increased during the first trimester, peak at the middle of the second trimester and return to baseline after childbirth (89-91). Other studies, however, report no changes in lipoprotein(a) during pregnancy (92,93).

 

Hyperthyroidism is associated with a reduction in lipoprotein(a) level and treatment is found to increase it (94). Hypothyroidism is also associated with an elevation in lipoprotein(a) level and treatment with thyroxine can reduce it (94). Growth hormone (GH) therapy for GH-deficient adults can markedly increase lipoprotein(a) levels (95). 

 

A decrease in lipoprotein(a) level has been reported in several genetic disorders of lipoprotein metabolism including abetalipoproteinemia, lecithin-cholesterol acyltransferase (LCAT) deficiency, and lipoprotein lipase deficiency (96). In contrast, an increase in lipoprotein(a) level is observed in familial hypercholesterolemia (FH) and familial defective apo B (96).

 

Conditions that have been reported to increase or decrease lipoprotein(a) levels are shown in Table 1.

 

Table 1. Conditions Associated with Changes in Lipoprotein(a) Levels

Increase

Decrease

No change

High carbohydrate/low fat diet (10-15%)

Low carbohydrate/high fat diet (10-15%)

Lifestyle intervention

Hypothyroidism (5-20%)

Hyperthyroidism (20-25%)

Alcohol consumption

Pregnancy (200%)

Hormone replacement therapy (25%)

Fasting

Castration/ovariectomy (small)

Severe acute-phase reactions (burn/sepsis)

Endogenous sex hormone

Growth hormone therapy (25-100%)

Tocilizumab (30-40%)

Menopause

Chronic kidney disease (200-400%)

Hepatitis/cirrhosis

 

Nephrotic syndrome (300-500%)

 

 

Severe inflammatory condition

 

 

Protease inhibitors/antiretroviral therapy

 

 

 

ISSUES IN LIPOPROTEIN(A) MEASUREMENT AND REPORTING

 

The wide variation in apo(a) isoform size among individuals poses challenges in measuring and reporting lipoprotein(a) levels. Most commercially available immunoassays use polyclonal antibodies which may cross-react with multiple KIV2 repeats (97). As a result, it may overestimate or underestimate lipoprotein(a) levels in subjects with large or small apo(a) isoforms, respectively. Currently, the latex-enhanced immunoturbidimetric method by Denka Seiken, Japan, has been shown to be less affected to apo(a) isoform size variability with high concordance of values when compared with the reference enzyme-linked immunosorbent assay (ELISA) method (98,99). Although the antibodies used in the Denka assay are still isoform dependent, the impact of apo(a) size variation is minimized by the use of five different calibrators (99). Nevertheless, results comparing six commercially available immunoassays, all of which use five-point calibrators, show a wide range of discrepancy among various assays (100). The reference ELISA method using monoclonal antibodies developed by a group at the University of Washington is considered the least apo(a) isoform size-sensitive immunoassay available (98,99). Recently, a newly developed LC-MS/MS method has been shown to be unaffected by the apo(a) isoform size polymorphism and proposed as a candidate reference method for standardization of lipoprotein(a) assay (101).

 

Besides the issues in measuring lipoprotein(a) levels, the reporting of lipoprotein(a) levels is also challenging. Currently, there are 2 types of commercially available assays to measure and report lipoprotein(a) levels. The first one reports the level in total lipoprotein(a) mass concentrations, which include the mass of apo(a), apo B-100, lipid, and carbohydrate components. The values are based on assay calibrators and are expressed in mg/dL. In the first method, there is no traceability of the various calibrators to the reference material (102). In the second method, the level is reported in lipoprotein(a) particle number and expressed in nmol/L of apo(a). The assay calibrators used in the second approach are traceable to the World Health Organization/International Federation of Clinical Chemistry and Laboratory Medicine (WHO/IFCCLM) secondary reference material (98) and the values are compared to the “gold standard” monoclonal antibody-based ELISA developed by Marcovina et al. (103). It is recommended that lipoprotein(a) levels should be measured with an assay which is least subjected to apo(a) isoform size and has been calibrated with the WHO/IFCCLM reference material. Current recommendations also encourage the reporting of lipoprotein(a) levels in molar units, but if it is not available, the units in which the assay is calibrated should be used for reporting (59,104,105).

 

It is known that different apo(a) isoform sizes give different molecular weights (106), therefore, direct conversion between molar and mass concentrations (i.e. nmol/L and mg/dL) using a single conversion factor could be misleading and is discouraged (59,99,106). Nevertheless, a factor of 2.0-2.5 is traditionally used to convert from a mass unit to a molar unit (97), although it may vary from 1.85 for a large apo(a) size to 2.85 for a small apo(a) size (99).

 

Since lipoprotein(a) and LDL have relatively similar size and density, cholesterol contained in lipoprotein(a) particles cannot be separated from that in LDL particles and is therefore collectively reported as LDL-C concentration. Previous experiments of isolated lipoprotein(a) particles showed that cholesterol accounted for approximately 30% of lipoprotein(a) mass concentration, therefore, lipoprotein(a)-cholesterol could be estimated by multiplying lipoprotein(a) mass (mg/dL) by 0.3 and used to correct LDL-C (lipoprotein(a)-cholesterol-corrected LDL-C) (59). However, a recent study directly measured lipoprotein(a)-cholesterol relative to lipoprotein(a) mass demonstrated that the percentage varied widely, ranging from 5.8% to 57.3% with a median of 17.3% (107), therefore, routine correction of LDL-C for lipoprotein(a)-cholesterol is currently not recommended in clinical practice (59). There are certain exceptions to this. First, in patients with clinically suspected familiar hypercholesterolemia (FH) and elevated lipoprotein(a) levels, measurement of lipoprotein(a)-cholesterol may be warranted since the corrected value may help exclude the diagnosis of FH and avoid unnecessary genetic testing (108). Second, correcting LDL-C for lipoprotein(a)-cholesterol may help explain the suboptimal response or resistance to statin therapy. High levels of lipoprotein(a) result in falsely elevated LDL-C. Since statins do not lower lipoprotein(a) levels, cholesterol in lipoprotein(a) is considered a statin-resistant fraction of LDL-C.

 

EFFECTS OF LIPOPROTEIN(A) ON VASCULAR DISEASE

 

Lipoprotein(a) exerts multiple effects that could be proatherogenic, proinflammatory, and prothrombotic.

 

Lipoprotein(a) could be atherogenic since lipoprotein(a) is small enough (<70 nm in diameter) to enter and become trapped in the vascular wall. Cholesterol carried on lipoprotein(a) particle could then be deposited in the arterial intima and aortic valvular leaflets. However, the number of circulating lipoprotein(a) particles is substantially lower than that of LDL particles. As a result, the amount of cholesterol deposited from lipoprotein(a) is expected to be much lower than that from LDL (109). Apo(a), the main protein of lipoprotein(a), also contains lysine-binding sites that could tightly bind to exposed surface of denuded endothelium of the vascular wall or the aortic valve leaflets. In an animal experiment of endothelial injury, it was found that lipoprotein(a) preferentially accumulated at injured sites, compared with LDL (110). These findings suggest that accumulation of lipoprotein(a) at the site of vascular injury could be the primary mechanism by which elevated lipoprotein(a) causes cardiovascular disease.   

 

There is increasing evidence that oxidized phospholipids carried on lipoprotein(a) play a major role in atherogenesis and valvular calcification. In plasma, lipoprotein(a) is the main carrier of oxidized phospholipids (15). Interaction between oxidized phospholipids and apo(a) is mediated by the histidine residues in the KIV-10 domain of apo(a) (111,112). These oxidized phospholipids are both proatherogenic and proinflammatory (113). Oxidized phospholipids can induce inflammation by binding TLR2, TLR4, CD14 and CD36 on monocytes, macrophages, and endothelial cells (79,114). The result is an activation of multiple cytokines, chemokines, and adhesion molecules that mediate monocyte activation and migration into the vascular wall, endothelial dysfunction and proliferation, proliferation and migration of vascular smooth muscle cells into the atheromatous plaques, generation of reactive oxygen species, progression of vascular wall inflammation, and cell apoptosis that could lead to plaque rupture (115,116).

 

Structurally, apo(a) is similar to plasminogen, but the protease domain of apo(a) is catalytically inactive. Therefore, apo(a) may inhibit binding of plasminogen to endothelial cell surface receptor, compete with plasminogen for fibrin affinity sites, and interfere with fibrinolytic activity of plasminogen through a molecular mimicry mechanism (21). However, lowering lipoprotein(a) levels does not affect ex vivo fibrinolysis (117), suggesting that the role of lipoprotein(a) in thrombosis may involve other mechanisms beyond fibrinolysis (118). Lipoprotein(a) has also been shown to induce tissue factor expression, inhibit tissue factor pathway inhibitor, promote platelet activation and aggregation, and alter the structure of fibrin, which ultimately leads to thrombosis (118).

 

It is of note that these proatherogenic, proinflammatory, and prothrombotic effects of lipoprotein(a) have been demonstrated mainly from in vitro studies, and the clinical significance of these findings needs to be established from human studies.  

 

LIPOPROTEIN(A) AND CARDIOVASCULAR DISEASE

 

Over the past two decades, data from epidemiological studies, meta-analyses, genome-wide association studies, and Mendelian randomization studies, have provided conclusive evidence that elevated lipoprotein(a) levels are associated with a higher risk of ASCVD (16,18,47,119-121). This is true in all ethnicities studied to date (17,55,122). The result of a genome-wide association study revealed that the most potent genetic association with coronary artery disease (CAD) was the LPA locus (121). A number of Mendelian randomization studies have shown that genetic variants associated with high lipoprotein(a) levels are more prevalent in subjects with ASCVD (18,47), whereas genetic variants associated with low lipoprotein(a) levels are protective against the development of ASCVD (31,49).

 

Several epidemiological studies, both in primary prevention and secondary prevention settings, have shown an association between lipoprotein(a) levels and ASCVD (17,19,55,120,123). The association is continuous and linear in different ethnicities without evidence of a threshold effect (17,120). Data from a large prospective cohort study from the UK Biobank showed a broadly linear relationship between lipoprotein(a) levels and ASCVD with an 11% increased risk of ASCVD with every 50 nmol/L increase in lipoprotein(a) levels (17). Another study also reported a relatively similar result, showing a 2.9% increased risk of CVD with every 18 nmol/L (10 mg/dL) increment in lipoprotein(a) levels (124).

 

Results from the clinical trials suggest that lipoprotein(a) might be a determinant of residual risk of ASCVD in patients who achieved low levels of LDL-C. For example, in the JUPITER trial using high-intensity rosuvastatin, on-statin lipoprotein(a) levels were associated with a residual risk of ASCVD even at low LDL-C levels (125). Similarly, findings from the FOURIER and ODYSSEY-OUTCOMES trials, using statins and PCSK9 inhibitors evolocumab and alirocumab, respectively, also demonstrated that patients with the higher baseline lipoprotein(a) levels were at an elevated risk of ASCVD irrespective of LDL-C levels (126,127). Lipoprotein(a) level is not associated with coronary artery calcium score, but both are independently associated with ASCVD risk (128). Elevated levels of lipoprotein(a) are associated with accelerated progression of atherosclerotic plaque (129).

 

The association between lipoprotein(a) levels and ASCVD is strongest for MI, atherosclerotic stenosis, and calcific aortic valvular disease (CAVD) (16). Additionally, very high levels of lipoprotein(a) levels are also associated with an increased risk of ischemic stroke (120,130) and heart failure (131), although the associations are weaker.  

 

Not only that lipoprotein(a) is a determinant of ASCVD in patients with lower risk of ASCVD, but in subjects who are at higher risk of ASCVD, such as those with FH, elevated lipoprotein(a) levels have also been shown to be associated with a higher risk of ASCVD (132,133). Lipoprotein(a) is also an independent predictor of MI and mortality in patients with CKD (134).

 

Regarding prothrombotic effects of lipoprotein(a), although in vitro data demonstrated that lipoprotein(a) might impair fibrinolysis, a potential role for lipoprotein(a) in prothrombotic and antifibrinolytic activity in vivo remains unproven (21). Epidemiological studies and Mendelian randomization studies have failed to establish the major role of lipoprotein(a) in venous thrombosis in adults (135,136). In children, however, there is some evidence that elevated lipoprotein(a) level is a risk factor for arterial ischemic stroke and venous thromboembolism (137).

 

LIPOPROTEIN(A) AND CALCIFIC AORTIC VALVULAR DISEASE

 

Calcific aortic valvular disease (CAVD) is the disease of the aortic valve that includes early valvular sclerosis and advanced aortic valvular stenosis. CAVD is the leading cause of aortic valve replacement in developed countries, but there is currently no available medical treatment for CAVD. CAVD is characterized by thickening of the aortic valve leaflets with progressive stenosis of the aortic valve. Although the early stages of CAVD are asymptomatic, severe aortic stenosis may lead to significant left ventricular outflow obstruction and subsequent development of syncope, angina and heart failure.

 

Early cross-sectional studies have shown a strong association between elevated lipoprotein(a) and CAVD (138,139). Such association, however, cannot be used to indicate that lipoprotein(a) is causal for CAVD. In 2013, a genome-wide association study in subjects of European ancestry reported an association between genetic variants in the LPA locus and CAVD (20), which was also confirmed across multiple ethnic groups (20,140). Subsequent cohort and case-control studies have verified this finding and further linked lipoprotein(a) levels and oxidized phospholipids to CAVD (141-144). Lipoprotein(a) is now considered as a new causal risk factor for CAVD (16). Two recent meta-analyses show that plasma lipoprotein(a) levels ³50 mg/dL are associated with a 1.76-1.79-fold increased risk of CAVD (145,146). Elevated lipoprotein(a) levels have also been associated with the faster progression of CAVD and the increased risk for aortic valve replacement and cardiovascular death (143,146-149). In contrast, genetically lowered levels of lipoprotein(a) are associated with a 37% lower risk of aortic stenosis (150).

 

The normal aortic valve consists of endothelial cells and valvular interstitial cells. In the early stage of CAVD, increased permeability of valvular endothelial cells occurs and results in endothelial barrier dysfunction. Mechanical shear stress may cause further injury to the valvular endothelial cells, leading to endothelium denudation. As a result, lipid infiltration by LDL and lipoprotein(a) follows, similar to the development of atherosclerosis. Inflammatory responses ensue and induce the valvular interstitial cells to gain a myofibroblast-like phenotype and release collagen matrix along with other bone-related proteins. The later stage of CAVD is characterized by the osteogenic differentiation of the valvular interstitial cells, resulting in progressive calcification of the aortic valve. Apoptosis of the valvular interstitial cells can lead to formation of hydroxyapatite crystals and further calcification, creating the propagation of both inflammation and calcification. Progressive fibrosis and widespread calcification ultimately result in thickening and dysfunction of the aortic valvular leaflets.  

 

The role of lipoprotein(a) in the molecular pathogenesis of CAVD is now accumulating. Apo(a) may increase the endothelial cell permeability through the Rho/RhoK signaling pathway, which is dependent on the lysine-binding site in the KIV10 domain. Since lipoprotein(a) is a major carrier of oxidized phospholipids in plasma, infiltration of lipoprotein(a) in the valvular tissue can deliver oxidized phospholipids to the valve. Oxidized phospholipids can directly attract monocytes and promote the transformation into macrophages. Oxidized phospholipids can then interact with various receptors on the macrophage, resulting in the release of several pro-inflammatory cytokines and subsequent formation of foam cells. Lipoprotein-associated phospholipase A2 (Lp-PLA2) from the macrophage carried on lipoprotein(a) can also act on oxidized phospholipids and generate lysophosphatidylcholine. Autotaxin, a lysophospholipase D enzyme secreted by the valvular interstitial cells and carried on lipoprotein(a) particles (151), can convert lysophosphatidylcholine into lysophosphatidic acid. Lysophosphatidic acid is both pro-inflammatory and pro-calcifying. By binding the lysophospholipid receptor on the valvular interstitial cells, it can activate the nuclear factor kappa B (NF-kB) and the Wnt-b-catenin pathways, leading to increased transcripts of IL-6 and various osteogenic genes, including runt-related transcription factor 2 (RUNX2) and bone morphogenetic protein 2 (BMP2). The result is the production of alkaline phosphatase, calcium deposition, inflammation, and calcification of the aortic valve (152).

 

These molecular pathways involved in the pathogenesis of CAVD provide several targets of interest, in addition to lipoprotein(a), that could be targeted and may lead to novel therapies for CAVD in the future.

 

GUIDELINE RECOMMENDATIONS FOR MEASUREMENT OF LIPOPROTEIN(A) LEVELS

 

Measurement of lipoprotein(a) levels could help refine the cardiovascular risk in certain conditions and various professional societies have recommended specific clinical conditions that warrant the measurement of lipoprotein(a) levels (59,105,153-156) as shown in Table 2.

 

Table 2. Guideline Recommendations for Lipoprotein(a) Measurement

Indications

AHA

2018

HEART UK 2019

NLA

2019

CCS 2021

BHS

2022

EAS 2022

1. Familial hypercholesterolemia

 

All
Individuals

All Adults

2. Family history (1stdegree relatives) of premature ASCVD

3. Family history (1stdegree relatives) of elevated lipoprotein(a)

 

4. Personal history of premature ASCVD

 

 

5. Recurrent CVD despite optimal statin treatment

 

 

 

6. Inadequate LDL-C reduction in response to statin

 

 

 

7. Borderline or intermediate risk of ASCVD

8. Calcific valvular aortic stenosis

 

It has been pointed out that different recommendations on measurement of lipoprotein(a) among various guidelines may be related to when the guidelines have been written (157). Most of the earlier guidelines list a number of conditions in which lipoprotein(a) should be measured, but the 2019 European Society of Cardiology/European Atherosclerosis Society (ESC/EAS) dyslipidemia guidelines have simplified this and recommended that lipoprotein(a) measurement should be considered at least once in each adult person’s lifetime (158). The rationale behind this recommendation is to identify individuals with very high levels of lipoprotein(a), i.e., >180 mg/dL or >430 nmol/L, who have a lifetime risk of ASCVD equivalent to that of heterozygous FH (158). Thus, measuring lipoprotein(a) levels in those with very high levels could make a significant contribution in the overall management to reduce cardiovascular risk. For the latest EAS lipoprotein(a) consensus statement published in 2022, it is recommended that lipoprotein(a) levels should be measured at least once in every adult (59). Incorporating lipoprotein(a) levels in the cardiovascular risk assessment could help improve risk stratification and provide comprehensive ASCVD risk evaluation. In addition, failure to consider an individual’s lipoprotein(a) level could lead to an underestimate of the absolute cardiovascular risk (59). For example, in a person with baseline risk of ASCVD events of 25%, a lipoprotein(a) level of 150 mg/dL will substantially increase the risk to 68% (59). A new risk calculator incorporating lipoprotein(a) levels and other traditional cardiovascular risk factors is now publicly available at http://www.lpaclinicalguidance.com.

 

Elevated lipoprotein(a) level is transmitted as a co-dominant trait. When an index subject is found to have high lipoprotein(a) level, screening for high lipoprotein(a) level in other family members is recommended and could help identify other affected family members (59). This approach is relatively similar to cascade screening in FH, except that genetic testing is not required. For children, there is a recommendation that children who have history of hemorrhagic or ischemic stroke, children of a parent with premature ASCVD and no other identifiable risk factors, and children of a parent with high levels of lipoprotein(a) should have lipoprotein(a) testing (59,154).

 

In general, the higher the lipoprotein(a) level, the greater risk for ASCVD. Therefore, the use of a specific threshold is biologically implausible and not appropriate. However, clinical practice guidelines prefer the use of a certain threshold for therapeutic decisions. An early study in patients who suffered acute MI proposed the lipoprotein(a) level of >30 mg/dL to reflect a higher risk of MI (5), and this cutoff values have been referred to in several guideline recommendations from various international societies (156,159,160). Another threshold level of lipoprotein(a) that has been used by other professional societies to confer an increased risk of ASCVD is 50 mg/dL or 100-125 nmol/L (153,154), which corresponds to the 80th percentile in the European descent (45). A recent 2022 consensus statement from the EAS proposed a threshold of ³50 mg/dL (125 nmol/L) to rule in and <30 mg/dL (75 nmol/L) to rule out cardiovascular risk, whereas levels between 30-50 mg/dL (75-125 nmol/L) are grey zones (59). Different thresholds according to ethnicities have been proposed (161), although a large study from the UK Biobank has shown that the ASCVD risk among various ethnicities is similar when using a uniform threshold of ³150 nmol/L (17). At present, current management should remain the same regardless of ethnicity.

 

Interestingly, an inverse relationship between lipoprotein(a) levels and the risk of type 2 diabetes has been demonstrated from various studies and subjects with very low levels of lipoprotein(a) are at increased risk of diabetes (32-36,162). Some, but not all, genetic studies also suggest that low lipoprotein(a) level is causally associated with the risk of diabetes (33-35). The exact mechanism underlying this association is currently unclear. Whether potent specific lipoprotein(a)-lowering therapy might increase the risk of diabetes remains to be determined.

 

LIPOPROTEIN(A)-LOWERING TREATMENT

 

Currently, there is no approved medication for lowering lipoprotein(a) levels. Currently available lipid-lowering medications result in modest (10-30%) reduction in lipoprotein(a) levels (table 3) (163). Data from the Mendelian randomization studies have suggested that lowering lipoprotein(a) levels by approximately 100 mg/dL (215 nmol/L) would produce a similar benefit of reducing ASCVD risk by 22% as shown by lowering LDL-C levels by 38.6 mg/dL (1 mmol/L) using statin therapy (164). A separate analysis using data from primary prevention studies suggested that this number might be overestimated and lowering lipoprotein(a) levels by 65 mg/dL would be enough to give 22% ASCVD risk reduction (165,166). Similarly, data from the Copenhagen General Population Study demonstrated that lowering lipoprotein(a) by 50 mg/dL (105 nmol/L) within a 5-year period would produce a 20% reduction in major adverse cardiovascular events for secondary prevention (167). It is of note that lowering LDL-C levels of 38.6 mg/dL to give a reduction of an adverse cardiovascular event of 22% was used only as a benchmark and it is possible that lowering lipoprotein(a) levels by smaller amount may also be beneficial as previously shown for LDL-C lowering.

 

Although early studies report an increase in lipoprotein(a) levels after statin therapy, recent meta-analyses of statin therapy show that there is heterogeneity across studies. In addition, the changes in lipoprotein(a) levels are relatively small and may not be clinically meaningful (168,169). Currently, it is widely accepted that the benefits of statin in lowering LDL-C levels and decreasing ASCVD risk outweigh the potential risk associated with a small increase in lipoprotein(a) levels. 

 

PCSK9 inhibitors, including evolocumab, alirocumab and inclisiran, can lower lipoprotein(a) levels by 10-30% and the absolute reduction is greatest in those with high lipoprotein(a) levels at baseline (126,127,170). A post-hoc analysis from the FOURIER trial using evolocumab reported a 23% decrease in adverse cardiovascular events in those with a baseline lipoprotein(a) level >37 nmol/L and a 7% reduction in those with <37 nmol/L (126). A significant relationship between a 15% lower risk per 25 nmol/L reduction of lipoprotein(a) levels was also observed. These data suggest that a small lowering in lipoprotein(a) level might have a clinical benefit. Results from the ODYSSEY Outcomes trial using alirocumab also showed a significant reduction in major cardiovascular events among those in the two highest quartiles of baseline lipoprotein(a) levels (127).    

 

Interestingly, an exploratory analysis of the FOURIER trial further showed that lipoprotein(a) level was associated with future aortic stenosis (AS) events (new or worsening CAVD or aortic valve replacement) and that evolocumab therapy beyond one year might reduce these AS events (171). However, it should be noted that this post hoc analysis was performed in only a small number of subjects and the original trial was not designed to evaluate the impact of PCSK9 inhibitors on CAVD.

 

Other lipid-lowering medications, such as fibrates, ezetimibe, bempedoic acid and bile acid sequestrants, also have modest effects on lipoprotein(a) levels (163). Niacin can lower lipoprotein(a) levels by 20% due to decreased LPA mRNA and apo(a) production rate (172). Mipomersen and CETP inhibitors also decrease lipoprotein(a) levels by 20-30% (173). Currently, the use of these medications to lower lipoprotein(a) level is not advised.

 

Lipoprotein apheresis is an option to lower lipoprotein(a) levels in clinical practice, although levels of all apoB-containing lipoproteins are reduced after treatment (174). A median reduction in lipoprotein(a) level by 70% was observed (175) but the time-averaged reduction was around 30-35% (176). Notably, a reduction in CAD events has been reported from several retrospective studies (177-180). Lipoprotein apheresis could therefore be considered in those with very high levels of lipoprotein(a) and progressive cardiovascular disease despite optimal management of other risk factors (174). Worldwide, lipoprotein apheresis is not commonly performed except in Germany where it is approved for ASCVD patients who have elevated lipoprotein(a) levels (>120 nmol/L or >60 mg/dL) and recurrent ASCVD events, irrespective of LDL-C levels (175). Recently, it is also approved in the United States for patients with lipoprotein(a) levels >60 mg/dL and LDL-C levels >100 mg/dL with documented CAD or peripheral arterial disease (163).

 

Without specific lipoprotein(a)-lowering therapy yet available, in subjects with elevated lipoprotein(a) levels, intensive risk factor management, such as healthy diet and lifestyle behavior modifications, is recommended along with the intensification of statin therapy to lower the risk of ASCVD (59). Data from the EPIC-Norfolk population-based study showed that in subjects with elevated lipoprotein(a) above 50 mg/dL, those who modified their lifestyles to maintain ideal cardiovascular health had about one third of cardiovascular risk compared to those with poor cardiovascular health (181). These modifiable cardiovascular health scores included body mass index, healthy diet, physical activity, smoking status, high blood pressure, diabetes and cholesterol concentration (181). In those whose LDL-C target levels are not achieved, ezetimibe or PCSK9 inhibitors could be considered (154).

 

Aspirin has been shown to be associated with a cardiovascular risk reduction in subjects who carried the SNP rs3798220-C with elevated lipoprotein(a) levels from the Women’s Health Study and the Aspirin in Reducing Events in the Elderly (APREE) trial (182). However, this SNP was present in only a small percentage of Caucasian subjects (approximately 3-4%) in these 2 trials. Therefore, further studies with a larger number of subjects in broader populations are needed to confirm the benefits of aspirin.

 

NEW SPECIFIC LIPOPROTEIN(A)-LOWERING TREATMENT

 

Since apo(a) is exclusively produced by the liver, novel therapies targeting hepatic apo(a) production using antisense oligonucleotide (ASO) and small interfering RNA (siRNA) technologies are under active investigation. 

 

Pelacarsen, formerly known as TQJ230, IONIS-APO(a)-LRx and AKCEA-APO(a)-LRx, is a second generation of apo(a) ASO, conjugated with N-acetylgalactosamine (GalNAc). Since GalNac is preferentially bound to asialoglycoprotein receptor on the cell surface of hepatocytes, the design of this GalNac-conjugated molecule will ensure the selective uptake by hepatocytes. Administered by subcutaneous injection every 4 weeks, it can bind apo(a) RNA in the hepatocytes, leading to an approximately 80% reduction in lipoprotein(a) levels in subjects with lipoprotein(a) level >150 nmol/L or approximately 60 mg/dL (183). Pelacarsen is generally well tolerated with the most frequently reported adverse event being injection site reactions. Currently, a phase 3 Lp(a)-HORIZON (NCT04023552) cardiovascular outcome study is underway to investigate the efficacy and safety of pelacarsen at the dose of 80 mg monthly for 4 years in subjects with ASCVD and lipoprotein(a) ³70 mg/dL.

 

Olpasiran, formerly known as AMG890 and ARO-LPA, is a GalNAc-conjugated siRNA that could be given every 3-6 months and result in a greater (>90%) reduction in lipoprotein(a) levels in subjects with lipoprotein(a) level >150 nmol/L or approximately 70 mg/dL (184). The most common adverse events were injection site reactions and hypersensitivity reactions. A cardiovascular outcome study (NCT05581303) is currently investigating the effects of olpasiran given every 12 weeks for 4 years in subjects with ASCVD and lipoprotein(a) levels ³200 nmol/L.

 

Zerlasiran, also known as SLN360, is another GalNAc-conjugated siRNA (185). A Phase 1 study investigated the safety and tolerability of zerlasiran after single ascending doses and multiple doses in healthy subjects with elevated lipoprotein(a) level ³150 nmol/L. A dose-dependent and sustained reduction in lipoprotein(a) level has been observed with a 98% reduction in the group receiving 600 mg of zerlasiran (185). A phase 2 ALPACAR-360 study (NCT05537571) to evaluate the Lipoprotein (a) lowering efficacy, safety and tolerability of zerlasiran in adult participants with elevated lipoprotein(a) (³125 nmol/L) at high risk of ASCVD is now underway.

 

Besides nucleic acid therapeutics to inhibit hepatic production of apo(a), small molecule inhibitors of lipoprotein(a) formation have been developed. Muvalaplin, an oral small molecule that blocks the initial noncovalent binding between apo(a) and apo B-100, thus disrupting lipoprotein(a) formation, has been reported to lower lipoprotein(a) level by 65% after 14 days of daily dosing in a phase 1 study (186). No serious adverse effects have been noted.

 

The effect of lipid-lowering medications on lipoprotein(a) levels is shown in Table 3.

 

Table 3. Effect of Lipid-Lowering Medications on Lipoprotein(a) Levels

Statins

No effect or slight increase

Ezetimibe

No effect or slight increase

Fibrates

No effect

Bempedoic acid

Minimal effect

Bile acid sequestrants

Minimal effect

Omega-3 fatty acids

No effect

Niacin

Decrease 15-25%

Lomitapide

Decrease 15-20%

Mipomersen*

Decrease 20-30%

CETP inhibitors*

Decrease 20-30%

Estrogen

Decrease 20-35%

PCSK9 inhibitors

Decrease 10-30%

Apo(a) ASO*

Decrease 80%

Apo(a) siRNA*

Decrease 90-98%

Apo(a) small molecule inhibitor*

Decrease 65%

*not currently available

 

The results of these ongoing and future clinical trials of lipoprotein(a) reduction are eagerly awaited and are expected to be the last piece of evidence supporting and confirming the causal relationship between lipoprotein(a) and ASCVD. Until a specific therapy for elevated lipoprotein(a) is available, intensive management of other modifiable cardiovascular risk factors is strongly recommended. Whether these lipoprotein(a)-lowering treatments would also be beneficial in CAVD remains to be further explored.

 

CONCLUSION

 

After 60 years of its discovery, several issues of lipoprotein(a) remain unresolved, including its function and metabolism. While the causality of lipoprotein(a) in ASCVD and CAVD has now been firmly established by epidemiological, genetic association, and Mendelian randomization studies, the next challenge is to prove that lowering lipoprotein(a) levels also leads to cardiovascular benefit in patients with elevated lipoprotein(a) levels.

 

REFERENCES

 

  1. Berg K. A new serum type system in man--the Lp system. Acta Pathol Microbiol Scand. 1963;59:369-382.
  2. Ehnholm C, Garoff H, Simons K, Aro H. Purification and quantitation of the human plasma lipoprotein carrying the Lp(a) antigen. Biochim Biophys Acta. 1971;236(2):431-439.
  3. Frick MH, Dahlen G, Furbery C, Ericson C, Wiljasalo M. Serum pre-beta-1 lipoprotein fraction in coronary atherosclerosis. Acta Med Scand. 1974;195(5):337-340.
  4. Berg K, Dahlen G, Frick MH. Lp(a) lipoprotein and pre-beta1-lipoprotein in patients with coronary heart disease. Clin Genet. 1974;6(3):230-235.
  5. Kostner GM, Avogaro P, Cazzolato G, Marth E, Bittolo-Bon G, Qunici GB. Lipoprotein Lp(a) and the risk for myocardial infarction. Atherosclerosis. 1981;38(1-2):51-61.
  6. Eaton DL, Fless GM, Kohr WJ, McLean JW, Xu QT, Miller CG, Lawn RM, Scanu AM. Partial amino acid sequence of apolipoprotein(a) shows that it is homologous to plasminogen. Proc Natl Acad Sci U S A. 1987;84(10):3224-3228.
  7. McLean JW, Tomlinson JE, Kuang WJ, Eaton DL, Chen EY, Fless GM, Scanu AM, Lawn RM. cDNA sequence of human apolipoprotein(a) is homologous to plasminogen. Nature. 1987;330(6144):132-137.
  8. Utermann G, Menzel HJ, Kraft HG, Duba HC, Kemmler HG, Seitz C. Lp(a) glycoprotein phenotypes. Inheritance and relation to Lp(a)-lipoprotein concentrations in plasma. J Clin Invest. 1987;80(2):458-465.
  9. Rosengren A, Wilhelmsen L, Eriksson E, Risberg B, Wedel H. Lipoprotein (a) and coronary heart disease: a prospective case-control study in a general population sample of middle aged men. BMJ. 1990;301(6763):1248-1251.
  10. Dahlen GH, Guyton JR, Attar M, Farmer JA, Kautz JA, Gotto AM, Jr. Association of levels of lipoprotein Lp(a), plasma lipids, and other lipoproteins with coronary artery disease documented by angiography. Circulation.1986;74(4):758-765.
  11. Bostom AG, Cupples LA, Jenner JL, Ordovas JM, Seman LJ, Wilson PW, Schaefer EJ, Castelli WP. Elevated plasma lipoprotein(a) and coronary heart disease in men aged 55 years and younger. A prospective study. JAMA.1996;276(7):544-548.
  12. Seman LJ, DeLuca C, Jenner JL, Cupples LA, McNamara JR, Wilson PW, Castelli WP, Ordovas JM, Schaefer EJ. Lipoprotein(a)-cholesterol and coronary heart disease in the Framingham Heart Study. Clin Chem.1999;45(7):1039-1046.
  13. Ridker PM, Hennekens CH, Stampfer MJ. A prospective study of lipoprotein(a) and the risk of myocardial infarction. JAMA. 1993;270(18):2195-2199.
  14. Ridker PM, Stampfer MJ, Hennekens CH. Plasma concentration of lipoprotein(a) and the risk of future stroke. JAMA. 1995;273(16):1269-1273.
  15. Bergmark C, Dewan A, Orsoni A, Merki E, Miller ER, Shin MJ, Binder CJ, Horkko S, Krauss RM, Chapman MJ, Witztum JL, Tsimikas S. A novel function of lipoprotein [a] as a preferential carrier of oxidized phospholipids in human plasma. J Lipid Res. 2008;49(10):2230-2239.
  16. Nordestgaard BG, Langsted A. Lipoprotein (a) as a cause of cardiovascular disease: insights from epidemiology, genetics, and biology. J Lipid Res. 2016;57(11):1953-1975.
  17. Patel AP, Wang M, Pirruccello JP, Ellinor PT, Ng K, Kathiresan S, Khera AV. Lp(a) (Lipoprotein[a]) Concentrations and Incident Atherosclerotic Cardiovascular Disease: New Insights From a Large National Biobank. Arterioscler Thromb Vasc Biol. 2021;41(1):465-474.
  18. Kamstrup PR, Tybjaerg-Hansen A, Steffensen R, Nordestgaard BG. Genetically elevated lipoprotein(a) and increased risk of myocardial infarction. JAMA. 2009;301(22):2331-2339.
  19. Kamstrup PR, Benn M, Tybjaerg-Hansen A, Nordestgaard BG. Extreme lipoprotein(a) levels and risk of myocardial infarction in the general population: the Copenhagen City Heart Study. Circulation. 2008;117(2):176-184.
  20. Thanassoulis G, Campbell CY, Owens DS, Smith JG, Smith AV, Peloso GM, Kerr KF, Pechlivanis S, Budoff MJ, Harris TB, Malhotra R, O'Brien KD, Kamstrup PR, Nordestgaard BG, Tybjaerg-Hansen A, Allison MA, Aspelund T, Criqui MH, Heckbert SR, Hwang SJ, Liu Y, Sjogren M, van der Pals J, Kalsch H, Muhleisen TW, Nothen MM, Cupples LA, Caslake M, Di Angelantonio E, Danesh J, Rotter JI, Sigurdsson S, Wong Q, Erbel R, Kathiresan S, Melander O, Gudnason V, O'Donnell CJ, Post WS, Group CECW. Genetic associations with valvular calcification and aortic stenosis. N Engl J Med. 2013;368(6):503-512.
  21. Boffa MB, Koschinsky ML. Lipoprotein (a): truly a direct prothrombotic factor in cardiovascular disease? J Lipid Res. 2016;57(5):745-757.
  22. Lawn RM, Schwartz K, Patthy L. Convergent evolution of apolipoprotein(a) in primates and hedgehog. Proc Natl Acad Sci U S A. 1997;94(22):11992-11997.
  23. Schmidt K, Noureen A, Kronenberg F, Utermann G. Structure, function, and genetics of lipoprotein (a). J Lipid Res.2016;57(8):1339-1359.
  24. Trieu VN, McConathy WJ. A two-step model for lipoprotein(a) formation. J Biol Chem. 1995;270(26):15471-15474.
  25. Dieplinger H, Utermann G. The seventh myth of lipoprotein(a): where and how is it assembled? Curr Opin Lipidol.1999;10(3):275-283.
  26. Kostner KM, Kostner GM. Lipoprotein (a): a historical appraisal. J Lipid Res. 2017;58(1):1-14.
  27. Youssef A, Clark JR, Marcovina SM, Boffa MB, Koschinsky ML. Apo(a) and ApoB Interact Noncovalently Within Hepatocytes: Implications for Regulation of Lp(a) Levels by Modulation of ApoB Secretion. Arterioscler Thromb Vasc Biol. 2022;42(3):289-304.
  28. Boffa MB, Koschinsky ML. Understanding the ins and outs of lipoprotein (a) metabolism. Curr Opin Lipidol.2022;33(3):185-192.
  29. Brown MS, Goldstein JL. Plasma lipoproteins: teaching old dogmas new tricks. Nature. 1987;330(6144):113-114.
  30. van der Hoek YY, Sangrar W, Cote GP, Kastelein JJ, Koschinsky ML. Binding of recombinant apolipoprotein(a) to extracellular matrix proteins. Arterioscler Thromb. 1994;14(11):1792-1798.
  31. Lim ET, Wurtz P, Havulinna AS, Palta P, Tukiainen T, Rehnstrom K, Esko T, Magi R, Inouye M, Lappalainen T, Chan Y, Salem RM, Lek M, Flannick J, Sim X, Manning A, Ladenvall C, Bumpstead S, Hamalainen E, Aalto K, Maksimow M, Salmi M, Blankenberg S, Ardissino D, Shah S, Horne B, McPherson R, Hovingh GK, Reilly MP, Watkins H, Goel A, Farrall M, Girelli D, Reiner AP, Stitziel NO, Kathiresan S, Gabriel S, Barrett JC, Lehtimaki T, Laakso M, Groop L, Kaprio J, Perola M, McCarthy MI, Boehnke M, Altshuler DM, Lindgren CM, Hirschhorn JN, Metspalu A, Freimer NB, Zeller T, Jalkanen S, Koskinen S, Raitakari O, Durbin R, MacArthur DG, Salomaa V, Ripatti S, Daly MJ, Palotie A, Sequencing Initiative Suomi P. Distribution and medical impact of loss-of-function variants in the Finnish founder population. PLoS Genet. 2014;10(7):e1004494.
  32. Mora S, Kamstrup PR, Rifai N, Nordestgaard BG, Buring JE, Ridker PM. Lipoprotein(a) and risk of type 2 diabetes. Clin Chem. 2010;56(8):1252-1260.
  33. Kamstrup PR, Nordestgaard BG. Lipoprotein(a) concentrations, isoform size, and risk of type 2 diabetes: a Mendelian randomisation study. Lancet Diabetes Endocrinol. 2013;1(3):220-227.
  34. Ye Z, Haycock PC, Gurdasani D, Pomilla C, Boekholdt SM, Tsimikas S, Khaw KT, Wareham NJ, Sandhu MS, Forouhi NG. The association between circulating lipoprotein(a) and type 2 diabetes: is it causal? Diabetes.2014;63(1):332-342.
  35. Gudbjartsson DF, Thorgeirsson G, Sulem P, Helgadottir A, Gylfason A, Saemundsdottir J, Bjornsson E, Norddahl GL, Jonasdottir A, Jonasdottir A, Eggertsson HP, Gretarsdottir S, Thorleifsson G, Indridason OS, Palsson R, Jonasson F, Jonsdottir I, Eyjolfsson GI, Sigurdardottir O, Olafsson I, Danielsen R, Matthiasson SE, Kristmundsdottir S, Halldorsson BV, Hreidarsson AB, Valdimarsson EM, Gudnason T, Benediktsson R, Steinthorsdottir V, Thorsteinsdottir U, Holm H, Stefansson K. Lipoprotein(a) Concentration and Risks of Cardiovascular Disease and Diabetes. J Am Coll Cardiol. 2019;74(24):2982-2994.
  36. Paige E, Masconi KL, Tsimikas S, Kronenberg F, Santer P, Weger S, Willeit J, Kiechl S, Willeit P. Lipoprotein(a) and incident type-2 diabetes: results from the prospective Bruneck study and a meta-analysis of published literature. Cardiovasc Diabetol. 2017;16(1):38.
  37. Rader DJ, Cain W, Ikewaki K, Talley G, Zech LA, Usher D, Brewer HB, Jr. The inverse association of plasma lipoprotein(a) concentrations with apolipoprotein(a) isoform size is not due to differences in Lp(a) catabolism but to differences in production rate. J Clin Invest. 1994;93(6):2758-2763.
  38. McCormick SPA, Schneider WJ. Lipoprotein(a) catabolism: a case of multiple receptors. Pathology.2019;51(2):155-164.
  39. Sharma M, Redpath GM, Williams MJ, McCormick SP. Recycling of Apolipoprotein(a) After PlgRKT-Mediated Endocytosis of Lipoprotein(a). Circ Res. 2017;120(7):1091-1102.
  40. Scholz M, Kraft HG, Lingenhel A, Delport R, Vorster EH, Bickeboller H, Utermann G. Genetic control of lipoprotein(a) concentrations is different in Africans and Caucasians. Eur J Hum Genet. 1999;7(2):169-178.
  41. Chan DC, Watts GF, Coll B, Wasserman SM, Marcovina SM, Barrett PHR. Lipoprotein(a) Particle Production as a Determinant of Plasma Lipoprotein(a) Concentration Across Varying Apolipoprotein(a) Isoform Sizes and Background Cholesterol-Lowering Therapy. J Am Heart Assoc. 2019;8(7):e011781.
  42. White AL, Guerra B, Wang J, Lanford RE. Presecretory degradation of apolipoprotein [a] is mediated by the proteasome pathway. J Lipid Res. 1999;40(2):275-286.
  43. Coassin S, Kronenberg F. Lipoprotein(a) beyond the kringle IV repeat polymorphism: The complexity of genetic variation in the LPA gene. Atherosclerosis. 2022;349:17-35.
  44. Rubin J, Paultre F, Tuck CH, Holleran S, Reed RG, Pearson TA, Thomas CM, Ramakrishnan R, Berglund L. Apolipoprotein [a] genotype influences isoform dominance pattern differently in African Americans and Caucasians. J Lipid Res. 2002;43(2):234-244.
  45. Enkhmaa B, Anuurad E, Berglund L. Lipoprotein (a): impact by ethnicity and environmental and medical conditions. J Lipid Res. 2016;57(7):1111-1125.
  46. Mack S, Coassin S, Rueedi R, Yousri NA, Seppala I, Gieger C, Schonherr S, Forer L, Erhart G, Marques-Vidal P, Ried JS, Waeber G, Bergmann S, Dahnhardt D, Stockl A, Raitakari OT, Kahonen M, Peters A, Meitinger T, Strauch K, Kedenko L, Paulweber B, Lehtimaki T, Hunt SC, Vollenweider P, Lamina C, Kronenberg F. A genome-wide association meta-analysis on lipoprotein (a) concentrations adjusted for apolipoprotein (a) isoforms. J Lipid Res.2017;58(9):1834-1844.
  47. Clarke R, Peden JF, Hopewell JC, Kyriakou T, Goel A, Heath SC, Parish S, Barlera S, Franzosi MG, Rust S, Bennett D, Silveira A, Malarstig A, Green FR, Lathrop M, Gigante B, Leander K, de Faire U, Seedorf U, Hamsten A, Collins R, Watkins H, Farrall M, Consortium P. Genetic variants associated with Lp(a) lipoprotein level and coronary disease. N Engl J Med. 2009;361(26):2518-2528.
  48. Schachtl-Riess JF, Kheirkhah A, Gruneis R, Di Maio S, Schoenherr S, Streiter G, Losso JL, Paulweber B, Eckardt KU, Kottgen A, Lamina C, Kronenberg F, Coassin S, Investigators G. Frequent LPA KIV-2 Variants Lower Lipoprotein(a) Concentrations and Protect Against Coronary Artery Disease. J Am Coll Cardiol. 2021;78(5):437-449.
  49. Coassin S, Erhart G, Weissensteiner H, Eca Guimaraes de Araujo M, Lamina C, Schonherr S, Forer L, Haun M, Losso JL, Kottgen A, Schmidt K, Utermann G, Peters A, Gieger C, Strauch K, Finkenstedt A, Bale R, Zoller H, Paulweber B, Eckardt KU, Huttenhofer A, Huber LA, Kronenberg F. A novel but frequent variant in LPA KIV-2 is associated with a pronounced Lp(a) and cardiovascular risk reduction. Eur Heart J. 2017;38(23):1823-1831.
  50. Said MA, Yeung MW, van de Vegte YJ, Benjamins JW, Dullaart RPF, Ruotsalainen S, Ripatti S, Natarajan P, Juarez-Orozco LE, Verweij N, van der Harst P. Genome-Wide Association Study and Identification of a Protective Missense Variant on Lipoprotein(a) Concentration: Protective Missense Variant on Lipoprotein(a) Concentration-Brief Report. Arterioscler Thromb Vasc Biol. 2021;41(5):1792-1800.
  51. Zabaneh D, Kumari M, Sandhu M, Wareham N, Wainwright N, Papamarkou T, Hopewell J, Clarke R, Li K, Palmen J, Talmud PJ, Kronenberg F, Lamina C, Summerer M, Paulweber B, Price J, Fowkes G, Stewart M, Drenos F, Shah S, Shah T, Casas JP, Kivimaki M, Whittaker J, Hingorani AD, Humphries SE. Meta analysis of candidate gene variants outside the LPA locus with Lp(a) plasma levels in 14,500 participants of six White European cohorts. Atherosclerosis. 2011;217(2):447-451.
  52. Hoekstra M, Chen HY, Rong J, Dufresne L, Yao J, Guo X, Tsai MY, Tsimikas S, Post WS, Vasan RS, Rotter JI, Larson MG, Thanassoulis G, Engert JC. Genome-Wide Association Study Highlights APOH as a Novel Locus for Lipoprotein(a) Levels-Brief Report. Arterioscler Thromb Vasc Biol. 2021;41(1):458-464.
  53. Varvel S, McConnell JP, Tsimikas S. Prevalence of Elevated Lp(a) Mass Levels and Patient Thresholds in 532 359 Patients in the United States. Arterioscler Thromb Vasc Biol. 2016;36(11):2239-2245.
  54. Nago N, Kayaba K, Hiraoka J, Matsuo H, Goto T, Kario K, Tsutsumi A, Nakamura Y, Igarashi M. Lipoprotein(a) levels in the Japanese population: influence of age and sex, and relation to atherosclerotic risk factors. The Jichi Medical School Cohort Study. Am J Epidemiol. 1995;141(9):815-821.
  55. Virani SS, Brautbar A, Davis BC, Nambi V, Hoogeveen RC, Sharrett AR, Coresh J, Mosley TH, Morrisett JD, Catellier DJ, Folsom AR, Boerwinkle E, Ballantyne CM. Associations between lipoprotein(a) levels and cardiovascular outcomes in black and white subjects: the Atherosclerosis Risk in Communities (ARIC) Study. Circulation. 2012;125(2):241-249.
  56. Gurdasani D, Sjouke B, Tsimikas S, Hovingh GK, Luben RN, Wainwright NW, Pomilla C, Wareham NJ, Khaw KT, Boekholdt SM, Sandhu MS. Lipoprotein(a) and risk of coronary, cerebrovascular, and peripheral artery disease: the EPIC-Norfolk prospective population study. Arterioscler Thromb Vasc Biol. 2012;32(12):3058-3065.
  57. Jenner JL, Ordovas JM, Lamon-Fava S, Schaefer MM, Wilson PW, Castelli WP, Schaefer EJ. Effects of age, sex, and menopausal status on plasma lipoprotein(a) levels. The Framingham Offspring Study. Circulation.1993;87(4):1135-1141.
  58. Cobbaert C, Kesteloot H. Serum lipoprotein(a) levels in racially different populations. Am J Epidemiol.1992;136(4):441-449.
  59. Kronenberg F, Mora S, Stroes ESG, Ference BA, Arsenault BJ, Berglund L, Dweck MR, Koschinsky M, Lambert G, Mach F, McNeal CJ, Moriarty PM, Natarajan P, Nordestgaard BG, Parhofer KG, Virani SS, von Eckardstein A, Watts GF, Stock JK, Ray KK, Tokgozoglu LS, Catapano AL. Lipoprotein(a) in atherosclerotic cardiovascular disease and aortic stenosis: a European Atherosclerosis Society consensus statement. Eur Heart J.2022;43(39):3925-3946.
  60. Deshotels MR, Sun C, Nambi V, Virani SS, Matsushita K, Yu B, Ballantyne CM, Hoogeveen RC. Temporal Trends in Lipoprotein(a) Concentrations: The Atherosclerosis Risk in Communities Study. J Am Heart Assoc.2022;11(21):e026762.
  61. Langsted A, Kamstrup PR, Nordestgaard BG. Lipoprotein(a): fasting and nonfasting levels, inflammation, and cardiovascular risk. Atherosclerosis. 2014;234(1):95-101.
  62. Ebbeling CB, Knapp A, Johnson A, Wong JMW, Greco KF, Ma C, Mora S, Ludwig DS. Effects of a low-carbohydrate diet on insulin-resistant dyslipoproteinemia-a randomized controlled feeding trial. Am J Clin Nutr.2022;115(1):154-162.
  63. Enkhmaa B, Petersen KS, Kris-Etherton PM, Berglund L. Diet and Lp(a): Does Dietary Change Modify Residual Cardiovascular Risk Conferred by Lp(a)? Nutrients. 2020;12(7).
  64. Enkhmaa B, Berglund L. Non-genetic influences on lipoprotein(a) concentrations. Atherosclerosis. 2022;349:53-62.
  65. Feely J, Barry M, Keeling PW, Weir DG, Cooke T. Lipoprotein(a) in cirrhosis. BMJ. 1992;304(6826):545-546.
  66. Alessandri C, Basili S, Maurelli M, Andreozzi P, Violi F, Cordova C. Relationship between lipoprotein(a) levels in serum and some indices of protein synthesis in liver cirrhosis. Clin Chim Acta. 1994;224(2):125-129.
  67. Kraft HG, Menzel HJ, Hoppichler F, Vogel W, Utermann G. Changes of genetic apolipoprotein phenotypes caused by liver transplantation. Implications for apolipoprotein synthesis. J Clin Invest. 1989;83(1):137-142.
  68. Kronenberg F. Causes and consequences of lipoprotein(a) abnormalities in kidney disease. Clin Exp Nephrol.2014;18(2):234-237.
  69. Hopewell JC, Haynes R, Baigent C. The role of lipoprotein (a) in chronic kidney disease. J Lipid Res.2018;59(4):577-585.
  70. Kronenberg F, Lingenhel A, Lhotta K, Rantner B, Kronenberg MF, Konig P, Thiery J, Koch M, von Eckardstein A, Dieplinger H. The apolipoprotein(a) size polymorphism is associated with nephrotic syndrome. Kidney Int.2004;65(2):606-612.
  71. Barbagallo CM, Averna MR, Sparacino V, Galione A, Caputo F, Scafidi V, Amato S, Mancino C, Cefalu AB, Notarbartolo A. Lipoprotein (a) levels in end-stage renal failure and renal transplantation. Nephron.1993;64(4):560-564.
  72. Kronenberg F, Konig P, Lhotta K, Ofner D, Sandholzer C, Margreiter R, Dosch E, Utermann G, Dieplinger H. Apolipoprotein(a) phenotype-associated decrease in lipoprotein(a) plasma concentrations after renal transplantation. Arterioscler Thromb. 1994;14(9):1399-1404.
  73. Black IW, Wilcken DE. Decreases in apolipoprotein(a) after renal transplantation: implications for lipoprotein(a) metabolism. Clin Chem. 1992;38(3):353-357.
  74. Segarra A, Chacon P, Vilardell M, Piera LL. Cyclosporin and serum lipids in renal transplant recipients. Lancet.1993;341(8847):766; author reply 767.
  75. Missala I, Kassner U, Steinhagen-Thiessen E. A Systematic Literature Review of the Association of Lipoprotein(a) and Autoimmune Diseases and Atherosclerosis. Int J Rheumatol. 2012;2012:480784.
  76. Mooser V, Berger MM, Tappy L, Cayeux C, Marcovina SM, Darioli R, Nicod P, Chiolero R. Major reduction in plasma Lp(a) levels during sepsis and burns. Arterioscler Thromb Vasc Biol. 2000;20(4):1137-1142.
  77. Feingold KR. Lipid and Lipoprotein Levels in Patients with COVID-19 Infections. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Shah AS, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA)2000.
  78. Moriarty PM, Gorby LK, Stroes ES, Kastelein JP, Davidson M, Tsimikas S. Lipoprotein(a) and Its Potential Association with Thrombosis and Inflammation in COVID-19: a Testable Hypothesis. Curr Atheroscler Rep.2020;22(9):48.
  79. Dzobo KE, Kraaijenhof JM, Stroes ESG, Nurmohamed NS, Kroon J. Lipoprotein(a): An underestimated inflammatory mastermind. Atherosclerosis. 2022;349:101-109.
  80. Schultz O, Oberhauser F, Saech J, Rubbert-Roth A, Hahn M, Krone W, Laudes M. Effects of inhibition of interleukin-6 signalling on insulin sensitivity and lipoprotein (a) levels in human subjects with rheumatoid diseases. PLoS One. 2010;5(12):e14328.
  81. Maeda S, Abe A, Seishima M, Makino K, Noma A, Kawade M. Transient changes of serum lipoprotein(a) as an acute phase protein. Atherosclerosis. 1989;78(2-3):145-150.
  82. Andreassen AK, Berg K, Torsvik H. Changes in Lp(a) lipoprotein and other plasma proteins during acute myocardial infarction. Clin Genet. 1994;46(6):410-416.
  83. Derby CA, Crawford SL, Pasternak RC, Sowers M, Sternfeld B, Matthews KA. Lipid changes during the menopause transition in relation to age and weight: the Study of Women's Health Across the Nation. Am J Epidemiol. 2009;169(11):1352-1361.
  84. Anagnostis P, Antza C, Trakatelli C, Lambrinoudaki I, Goulis DG, Kotsis V. The effect of menopause on lipoprotein (a) concentrations: A systematic review and meta-analysis. Maturitas. 2023;167:39-45.
  85. Simony SB, Mortensen MB, Langsted A, Afzal S, Kamstrup PR, Nordestgaard BG. Sex differences of lipoprotein(a) levels and associated risk of morbidity and mortality by age: The Copenhagen General Population Study. Atherosclerosis. 2022;355:76-82.
  86. Denti L, Pasolini G, Cortellini P, Ferretti S, Sanfelici L, Ablondi F, Valenti G. Effects of androgen suppression by gonadotropin-releasing hormone agonist and flutamide on lipid metabolism in men with prostate cancer: focus on lipoprotein(a). Clin Chem. 1996;42(8 Pt 1):1176-1181.
  87. Henriksson P, Angelin B, Berglund L. Hormonal regulation of serum Lp (a) levels. Opposite effects after estrogen treatment and orchidectomy in males with prostatic carcinoma. J Clin Invest. 1992;89(4):1166-1171.
  88. Salpeter SR, Walsh JM, Ormiston TM, Greyber E, Buckley NS, Salpeter EE. Meta-analysis: effect of hormone-replacement therapy on components of the metabolic syndrome in postmenopausal women. Diabetes Obes Metab. 2006;8(5):538-554.
  89. Zechner R, Desoye G, Schweditsch MO, Pfeiffer KP, Kostner GM. Fluctuations of plasma lipoprotein-A concentrations during pregnancy and post partum. Metabolism. 1986;35(4):333-336.
  90. Sattar N, Clark P, Greer IA, Shepherd J, Packard CJ. Lipoprotein (a) levels in normal pregnancy and in pregnancy complicated with pre-eclampsia. Atherosclerosis. 2000;148(2):407-411.
  91. Manten GT, Franx A, van der Hoek YY, Hameeteman TM, Voorbij HA, Smolders HC, Westers P, Visser GH. Changes of plasma lipoprotein(a) during and after normal pregnancy in Caucasians. J Matern Fetal Neonatal Med.2003;14(2):91-95.
  92. Rymer J, Constable S, Lumb P, Crook M. Serum lipoprotein (A) and apolipoproteins during pregnancy and postpartum in normal women. J Obstet Gynaecol. 2002;22(3):256-259.
  93. Chiang AN, Yang ML, Hung JH, Chou P, Shyn SK, Ng HT. Alterations of serum lipid levels and their biological relevances during and after pregnancy. Life Sci. 1995;56(26):2367-2375.
  94. Kotwal A, Cortes T, Genere N, Hamidi O, Jasim S, Newman CB, Prokop LJ, Murad MH, Alahdab F. Treatment of Thyroid Dysfunction and Serum Lipids: A Systematic Review and Meta-analysis. J Clin Endocrinol Metab.2020;105(12).
  95. Eden S, Wiklund O, Oscarsson J, Rosen T, Bengtsson BA. Growth hormone treatment of growth hormone-deficient adults results in a marked increase in Lp(a) and HDL cholesterol concentrations. Arterioscler Thromb.1993;13(2):296-301.
  96. Kronenberg F, Utermann G. Lipoprotein(a): resurrected by genetics. J Intern Med. 2013;273(1):6-30.
  97. Kronenberg F. Lipoprotein(a) measurement issues: Are we making a mountain out of a molehill? Atherosclerosis.2022;349:123-135.
  98. Marcovina SM, Albers JJ, Scanu AM, Kennedy H, Giaculli F, Berg K, Couderc R, Dati F, Rifai N, Sakurabayashi I, Tate JR, Steinmetz A. Use of a reference material proposed by the International Federation of Clinical Chemistry and Laboratory Medicine to evaluate analytical methods for the determination of plasma lipoprotein(a). Clin Chem.2000;46(12):1956-1967.
  99. Marcovina SM, Albers JJ. Lipoprotein (a) measurements for clinical application. J Lipid Res. 2016;57(4):526-537.
  100. Scharnagl H, Stojakovic T, Dieplinger B, Dieplinger H, Erhart G, Kostner GM, Herrmann M, Marz W, Grammer TB. Comparison of lipoprotein (a) serum concentrations measured by six commercially available immunoassays. Atherosclerosis. 2019;289:206-213.
  101. Marcovina SM, Clouet-Foraison N, Koschinsky ML, Lowenthal MS, Orquillas A, Boffa MB, Hoofnagle AN, Vaisar T. Development of an LC-MS/MS Proposed Candidate Reference Method for the Standardization of Analytical Methods to Measure Lipoprotein(a). Clin Chem. 2021;67(3):490-499.
  102. Tsimikas S, Fazio S, Ferdinand KC, Ginsberg HN, Koschinsky ML, Marcovina SM, Moriarty PM, Rader DJ, Remaley AT, Reyes-Soffer G, Santos RD, Thanassoulis G, Witztum JL, Danthi S, Olive M, Liu L. NHLBI Working Group Recommendations to Reduce Lipoprotein(a)-Mediated Risk of Cardiovascular Disease and Aortic Stenosis. J Am Coll Cardiol. 2018;71(2):177-192.
  103. Marcovina SM, Albers JJ, Gabel B, Koschinsky ML, Gaur VP. Effect of the number of apolipoprotein(a) kringle 4 domains on immunochemical measurements of lipoprotein(a). Clin Chem. 1995;41(2):246-255.
  104. Reyes-Soffer G, Ginsberg HN, Berglund L, Duell PB, Heffron SP, Kamstrup PR, Lloyd-Jones DM, Marcovina SM, Yeang C, Koschinsky ML, American Heart Association Council on Arteriosclerosis T, Vascular B, Council on Cardiovascular R, Intervention, Council on Peripheral Vascular D. Lipoprotein(a): A Genetically Determined, Causal, and Prevalent Risk Factor for Atherosclerotic Cardiovascular Disease: A Scientific Statement From the American Heart Association. Arterioscler Thromb Vasc Biol. 2022;42(1):e48-e60.
  105. Cegla J, Neely RDG, France M, Ferns G, Byrne CD, Halcox J, Datta D, Capps N, Shoulders C, Qureshi N, Rees A, Main L, Cramb R, Viljoen A, Payne J, Soran H, Heart Uk Medical S, Research C. HEART UK consensus statement on Lipoprotein(a): A call to action. Atherosclerosis. 2019;291:62-70.
  106. Tsimikas S, Fazio S, Viney NJ, Xia S, Witztum JL, Marcovina SM. Relationship of lipoprotein(a) molar concentrations and mass according to lipoprotein(a) thresholds and apolipoprotein(a) isoform size. J Clin Lipidol.2018;12(5):1313-1323.
  107. Yeang C, Witztum JL, Tsimikas S. Novel method for quantification of lipoprotein(a)-cholesterol: implications for improving accuracy of LDL-C measurements. J Lipid Res. 2021;62:100053.
  108. Langsted A, Kamstrup PR, Benn M, Tybjaerg-Hansen A, Nordestgaard BG. High lipoprotein(a) as a possible cause of clinical familial hypercholesterolaemia: a prospective cohort study. Lancet Diabetes Endocrinol.2016;4(7):577-587.
  109. Nielsen LB, Gronholdt ML, Schroeder TV, Stender S, Nordestgaard BG. In vivo transfer of lipoprotein(a) into human atherosclerotic carotid arterial intima. Arterioscler Thromb Vasc Biol. 1997;17(5):905-911.
  110. Nielsen LB, Stender S, Kjeldsen K, Nordestgaard BG. Specific accumulation of lipoprotein(a) in balloon-injured rabbit aorta in vivo. Circ Res. 1996;78(4):615-626.
  111. Leibundgut G, Scipione C, Yin H, Schneider M, Boffa MB, Green S, Yang X, Dennis E, Witztum JL, Koschinsky ML, Tsimikas S. Determinants of binding of oxidized phospholipids on apolipoprotein (a) and lipoprotein (a). J Lipid Res. 2013;54(10):2815-2830.
  112. Koschinsky ML, Boffa MB. Oxidized phospholipid modification of lipoprotein(a): Epidemiology, biochemistry and pathophysiology. Atherosclerosis. 2022;349:92-100.
  113. van der Valk FM, Bekkering S, Kroon J, Yeang C, Van den Bossche J, van Buul JD, Ravandi A, Nederveen AJ, Verberne HJ, Scipione C, Nieuwdorp M, Joosten LA, Netea MG, Koschinsky ML, Witztum JL, Tsimikas S, Riksen NP, Stroes ES. Oxidized Phospholipids on Lipoprotein(a) Elicit Arterial Wall Inflammation and an Inflammatory Monocyte Response in Humans. Circulation. 2016;134(8):611-624.
  114. Seimon TA, Nadolski MJ, Liao X, Magallon J, Nguyen M, Feric NT, Koschinsky ML, Harkewicz R, Witztum JL, Tsimikas S, Golenbock D, Moore KJ, Tabas I. Atherogenic lipids and lipoproteins trigger CD36-TLR2-dependent apoptosis in macrophages undergoing endoplasmic reticulum stress. Cell Metab. 2010;12(5):467-482.
  115. Simantiris S, Antonopoulos AS, Papastamos C, Benetos G, Koumallos N, Tsioufis K, Tousoulis D. Lipoprotein(a) and inflammation- pathophysiological links and clinical implications for cardiovascular disease. J Clin Lipidol.2023;17(1):55-63.
  116. Coassin S, Kronenberg F. Mechanistic insights into lipoprotein(a): from infamous to 'inflammous'. Eur Heart J.2020;41(24):2272-2274.
  117. Boffa MB, Marar TT, Yeang C, Viney NJ, Xia S, Witztum JL, Koschinsky ML, Tsimikas S. Potent reduction of plasma lipoprotein (a) with an antisense oligonucleotide in human subjects does not affect ex vivo fibrinolysis. J Lipid Res. 2019;60(12):2082-2089.
  118. Boffa MB. Beyond fibrinolysis: The confounding role of Lp(a) in thrombosis. Atherosclerosis. 2022;349:72-81.
  119. Tsimikas S. A Test in Context: Lipoprotein(a): Diagnosis, Prognosis, Controversies, and Emerging Therapies. J Am Coll Cardiol. 2017;69(6):692-711.
  120. Emerging Risk Factors C, Erqou S, Kaptoge S, Perry PL, Di Angelantonio E, Thompson A, White IR, Marcovina SM, Collins R, Thompson SG, Danesh J. Lipoprotein(a) concentration and the risk of coronary heart disease, stroke, and nonvascular mortality. JAMA. 2009;302(4):412-423.
  121. Consortium CAD, Deloukas P, Kanoni S, Willenborg C, Farrall M, Assimes TL, Thompson JR, Ingelsson E, Saleheen D, Erdmann J, Goldstein BA, Stirrups K, Konig IR, Cazier JB, Johansson A, Hall AS, Lee JY, Willer CJ, Chambers JC, Esko T, Folkersen L, Goel A, Grundberg E, Havulinna AS, Ho WK, Hopewell JC, Eriksson N, Kleber ME, Kristiansson K, Lundmark P, Lyytikainen LP, Rafelt S, Shungin D, Strawbridge RJ, Thorleifsson G, Tikkanen E, Van Zuydam N, Voight BF, Waite LL, Zhang W, Ziegler A, Absher D, Altshuler D, Balmforth AJ, Barroso I, Braund PS, Burgdorf C, Claudi-Boehm S, Cox D, Dimitriou M, Do R, Consortium D, Consortium C, Doney AS, El Mokhtari N, Eriksson P, Fischer K, Fontanillas P, Franco-Cereceda A, Gigante B, Groop L, Gustafsson S, Hager J, Hallmans G, Han BG, Hunt SE, Kang HM, Illig T, Kessler T, Knowles JW, Kolovou G, Kuusisto J, Langenberg C, Langford C, Leander K, Lokki ML, Lundmark A, McCarthy MI, Meisinger C, Melander O, Mihailov E, Maouche S, Morris AD, Muller-Nurasyid M, Mu TC, Nikus K, Peden JF, Rayner NW, Rasheed A, Rosinger S, Rubin D, Rumpf MP, Schafer A, Sivananthan M, Song C, Stewart AF, Tan ST, Thorgeirsson G, van der Schoot CE, Wagner PJ, Wellcome Trust Case Control C, Wells GA, Wild PS, Yang TP, Amouyel P, Arveiler D, Basart H, Boehnke M, Boerwinkle E, Brambilla P, Cambien F, Cupples AL, de Faire U, Dehghan A, Diemert P, Epstein SE, Evans A, Ferrario MM, Ferrieres J, Gauguier D, Go AS, Goodall AH, Gudnason V, Hazen SL, Holm H, Iribarren C, Jang Y, Kahonen M, Kee F, Kim HS, Klopp N, Koenig W, Kratzer W, Kuulasmaa K, Laakso M, Laaksonen R, Lee JY, Lind L, Ouwehand WH, Parish S, Park JE, Pedersen NL, Peters A, Quertermous T, Rader DJ, Salomaa V, Schadt E, Shah SH, Sinisalo J, Stark K, Stefansson K, Tregouet DA, Virtamo J, Wallentin L, Wareham N, Zimmermann ME, Nieminen MS, Hengstenberg C, Sandhu MS, Pastinen T, Syvanen AC, Hovingh GK, Dedoussis G, Franks PW, Lehtimaki T, Metspalu A, Zalloua PA, Siegbahn A, Schreiber S, Ripatti S, Blankenberg SS, Perola M, Clarke R, Boehm BO, O'Donnell C, Reilly MP, Marz W, Collins R, Kathiresan S, Hamsten A, Kooner JS, Thorsteinsdottir U, Danesh J, Palmer CN, Roberts R, Watkins H, Schunkert H, Samani NJ. Large-scale association analysis identifies new risk loci for coronary artery disease. Nat Genet. 2013;45(1):25-33.
  122. Cai DP, He YM, Yang XJ, Zhao X, Xu HF. Lipoprotein (a) is a risk factor for coronary artery disease in Chinese Han ethnic population modified by some traditional risk factors: A cross-sectional study of 3462 cases and 6125 controls. Clin Chim Acta. 2015;451(Pt B):278-286.
  123. Pare G, Caku A, McQueen M, Anand SS, Enas E, Clarke R, Boffa MB, Koschinsky M, Wang X, Yusuf S, Investigators I. Lipoprotein(a) Levels and the Risk of Myocardial Infarction Among 7 Ethnic Groups. Circulation.2019;139(12):1472-1482.
  124. Welsh P, Welsh C, Celis-Morales CA, Brown R, Ho FK, Ferguson LD, Mark PB, Lewsey J, Gray SR, Lyall DM, Gill JMR, Pell JP, de Lemos JA, Willeit P, Sattar N. Lipoprotein(a) and cardiovascular disease: prediction, attributable risk fraction, and estimating benefits from novel interventions. Eur J Prev Cardiol. 2021;28(18):1991-2000.
  125. Khera AV, Everett BM, Caulfield MP, Hantash FM, Wohlgemuth J, Ridker PM, Mora S. Lipoprotein(a) concentrations, rosuvastatin therapy, and residual vascular risk: an analysis from the JUPITER Trial (Justification for the Use of Statins in Prevention: an Intervention Trial Evaluating Rosuvastatin). Circulation. 2014;129(6):635-642.
  126. O'Donoghue ML, Fazio S, Giugliano RP, Stroes ESG, Kanevsky E, Gouni-Berthold I, Im K, Lira Pineda A, Wasserman SM, Ceska R, Ezhov MV, Jukema JW, Jensen HK, Tokgozoglu SL, Mach F, Huber K, Sever PS, Keech AC, Pedersen TR, Sabatine MS. Lipoprotein(a), PCSK9 Inhibition, and Cardiovascular Risk. Circulation.2019;139(12):1483-1492.
  127. Bittner VA, Szarek M, Aylward PE, Bhatt DL, Diaz R, Edelberg JM, Fras Z, Goodman SG, Halvorsen S, Hanotin C, Harrington RA, Jukema JW, Loizeau V, Moriarty PM, Moryusef A, Pordy R, Roe MT, Sinnaeve P, Tsimikas S, Vogel R, White HD, Zahger D, Zeiher AM, Steg PG, Schwartz GG, Committees OO, Investigators. Effect of Alirocumab on Lipoprotein(a) and Cardiovascular Risk After Acute Coronary Syndrome. J Am Coll Cardiol. 2020;75(2):133-144.
  128. Mehta A, Vasquez N, Ayers CR, Patel J, Hooda A, Khera A, Blumenthal RS, Shapiro MD, Rodriguez CJ, Tsai MY, Sperling LS, Virani SS, Blaha MJ, Joshi PH. Independent Association of Lipoprotein(a) and Coronary Artery Calcification With Atherosclerotic Cardiovascular Risk. J Am Coll Cardiol. 2022;79(8):757-768.
  129. Kaiser Y, Daghem M, Tzolos E, Meah MN, Doris MK, Moss AJ, Kwiecinski J, Kroon J, Nurmohamed NS, van der Harst P, Adamson PD, Williams MC, Dey D, Newby DE, Stroes ESG, Zheng KH, Dweck MR. Association of Lipoprotein(a) With Atherosclerotic Plaque Progression. J Am Coll Cardiol. 2022;79(3):223-233.
  130. Langsted A, Nordestgaard BG, Kamstrup PR. Elevated Lipoprotein(a) and Risk of Ischemic Stroke. J Am Coll Cardiol. 2019;74(1):54-66.
  131. Kamstrup PR, Nordestgaard BG. Elevated Lipoprotein(a) Levels, LPA Risk Genotypes, and Increased Risk of Heart Failure in the General Population. JACC Heart Fail. 2016;4(1):78-87.
  132. Seed M, Hoppichler F, Reaveley D, McCarthy S, Thompson GR, Boerwinkle E, Utermann G. Relation of serum lipoprotein(a) concentration and apolipoprotein(a) phenotype to coronary heart disease in patients with familial hypercholesterolemia. N Engl J Med. 1990;322(21):1494-1499.
  133. Holmes DT, Schick BA, Humphries KH, Frohlich J. Lipoprotein(a) is an independent risk factor for cardiovascular disease in heterozygous familial hypercholesterolemia. Clin Chem. 2005;51(11):2067-2073.
  134. Bajaj A, Damrauer SM, Anderson AH, Xie D, Budoff MJ, Go AS, He J, Lash JP, Ojo A, Post WS, Rahman M, Reilly MP, Saleheen D, Townsend RR, Chen J, Rader DJ, Chronic Renal Insufficiency Cohort Study I. Lipoprotein(a) and risk of myocardial infarction and death in chronic kidney disease: findings from the CRIC study (Chronic Renal Insufficiency Cohort). Arterioscler Thromb Vasc Biol. 2017;37(10):1971-1978.
  135. Kamstrup PR, Tybjaerg-Hansen A, Nordestgaard BG. Genetic evidence that lipoprotein(a) associates with atherosclerotic stenosis rather than venous thrombosis. Arterioscler Thromb Vasc Biol. 2012;32(7):1732-1741.
  136. Helgadottir A, Gretarsdottir S, Thorleifsson G, Holm H, Patel RS, Gudnason T, Jones GT, van Rij AM, Eapen DJ, Baas AF, Tregouet DA, Morange PE, Emmerich J, Lindblad B, Gottsater A, Kiemeny LA, Lindholt JS, Sakalihasan N, Ferrell RE, Carey DJ, Elmore JR, Tsao PS, Grarup N, Jorgensen T, Witte DR, Hansen T, Pedersen O, Pola R, Gaetani E, Magnadottir HB, Wijmenga C, Tromp G, Ronkainen A, Ruigrok YM, Blankensteijn JD, Mueller T, Wells PS, Corral J, Soria JM, Souto JC, Peden JF, Jalilzadeh S, Mayosi BM, Keavney B, Strawbridge RJ, Sabater-Lleal M, Gertow K, Baldassarre D, Nyyssonen K, Rauramaa R, Smit AJ, Mannarino E, Giral P, Tremoli E, de Faire U, Humphries SE, Hamsten A, Haraldsdottir V, Olafsson I, Magnusson MK, Samani NJ, Levey AI, Markus HS, Kostulas K, Dichgans M, Berger K, Kuhlenbaumer G, Ringelstein EB, Stoll M, Seedorf U, Rothwell PM, Powell JT, Kuivaniemi H, Onundarson PT, Valdimarsson E, Matthiasson SE, Gudbjartsson DF, Thorgeirsson G, Quyyumi AA, Watkins H, Farrall M, Thorsteinsdottir U, Stefansson K. Apolipoprotein(a) genetic sequence variants associated with systemic atherosclerosis and coronary atherosclerotic burden but not with venous thromboembolism. J Am Coll Cardiol. 2012;60(8):722-729.
  137. Kenet G, Lutkhoff LK, Albisetti M, Bernard T, Bonduel M, Brandao L, Chabrier S, Chan A, deVeber G, Fiedler B, Fullerton HJ, Goldenberg NA, Grabowski E, Gunther G, Heller C, Holzhauer S, Iorio A, Journeycake J, Junker R, Kirkham FJ, Kurnik K, Lynch JK, Male C, Manco-Johnson M, Mesters R, Monagle P, van Ommen CH, Raffini L, Rostasy K, Simioni P, Strater RD, Young G, Nowak-Gottl U. Impact of thrombophilia on risk of arterial ischemic stroke or cerebral sinovenous thrombosis in neonates and children: a systematic review and meta-analysis of observational studies. Circulation. 2010;121(16):1838-1847.
  138. Stewart BF, Siscovick D, Lind BK, Gardin JM, Gottdiener JS, Smith VE, Kitzman DW, Otto CM. Clinical factors associated with calcific aortic valve disease. Cardiovascular Health Study. J Am Coll Cardiol. 1997;29(3):630-634.
  139. Gotoh T, Kuroda T, Yamasawa M, Nishinaga M, Mitsuhashi T, Seino Y, Nagoh N, Kayaba K, Yamada S, Matsuo H, et al. Correlation between lipoprotein(a) and aortic valve sclerosis assessed by echocardiography (the JMS Cardiac Echo and Cohort Study). Am J Cardiol. 1995;76(12):928-932.
  140. Cairns BJ, Coffey S, Travis RC, Prendergast B, Green J, Engert JC, Lathrop M, Thanassoulis G, Clarke R. A Replicated, Genome-Wide Significant Association of Aortic Stenosis With a Genetic Variant for Lipoprotein(a): Meta-Analysis of Published and Novel Data. Circulation. 2017;135(12):1181-1183.
  141. Arsenault BJ, Boekholdt SM, Dube MP, Rheaume E, Wareham NJ, Khaw KT, Sandhu MS, Tardif JC. Lipoprotein(a) levels, genotype, and incident aortic valve stenosis: a prospective Mendelian randomization study and replication in a case-control cohort. Circ Cardiovasc Genet. 2014;7(3):304-310.
  142. Kamstrup PR, Tybjaerg-Hansen A, Nordestgaard BG. Elevated lipoprotein(a) and risk of aortic valve stenosis in the general population. J Am Coll Cardiol. 2014;63(5):470-477.
  143. Capoulade R, Chan KL, Yeang C, Mathieu P, Bosse Y, Dumesnil JG, Tam JW, Teo KK, Mahmut A, Yang X, Witztum JL, Arsenault BJ, Despres JP, Pibarot P, Tsimikas S. Oxidized Phospholipids, Lipoprotein(a), and Progression of Calcific Aortic Valve Stenosis. J Am Coll Cardiol. 2015;66(11):1236-1246.
  144. Kamstrup PR, Hung MY, Witztum JL, Tsimikas S, Nordestgaard BG. Oxidized Phospholipids and Risk of Calcific Aortic Valve Disease: The Copenhagen General Population Study. Arterioscler Thromb Vasc Biol.2017;37(8):1570-1578.
  145. Liu Q, Yu Y, Xi R, Li J, Lai R, Wang T, Fan Y, Zhang Z, Xu H, Ju J. Association Between Lipoprotein(a) and Calcific Aortic Valve Disease: A Systematic Review and Meta-Analysis. Front Cardiovasc Med. 2022;9:877140.
  146. Pantelidis P, Oikonomou E, Lampsas S, Zakynthinos GE, Lysandrou A, Kalogeras K, Katsianos E, Theofilis P, Siasos G, Vavuranakis MA, Antonopoulos AS, Tousoulis D, Vavouranakis M. Lipoprotein(a) and calcific aortic valve disease initiation and progression: a systematic review and meta-analysis. Cardiovasc Res. 2023;119(8):1641-1655.
  147. Zheng KH, Tsimikas S, Pawade T, Kroon J, Jenkins WSA, Doris MK, White AC, Timmers N, Hjortnaes J, Rogers MA, Aikawa E, Arsenault BJ, Witztum JL, Newby DE, Koschinsky ML, Fayad ZA, Stroes ESG, Boekholdt SM, Dweck MR. Lipoprotein(a) and Oxidized Phospholipids Promote Valve Calcification in Patients With Aortic Stenosis. J Am Coll Cardiol. 2019;73(17):2150-2162.
  148. Perez de Isla L, Watts GF, Alonso R, Diaz-Diaz JL, Muniz-Grijalvo O, Zambon D, Fuentes F, de Andres R, Padro T, Lopez-Miranda J, Mata P. Lipoprotein(a), LDL-cholesterol, and hypertension: predictors of the need for aortic valve replacement in familial hypercholesterolaemia. Eur Heart J. 2021;42(22):2201-2211.
  149. Capoulade R, Yeang C, Chan KL, Pibarot P, Tsimikas S. Association of Mild to Moderate Aortic Valve Stenosis Progression With Higher Lipoprotein(a) and Oxidized Phospholipid Levels: Secondary Analysis of a Randomized Clinical Trial. JAMA Cardiol. 2018;3(12):1212-1217.
  150. Emdin CA, Khera AV, Natarajan P, Klarin D, Won HH, Peloso GM, Stitziel NO, Nomura A, Zekavat SM, Bick AG, Gupta N, Asselta R, Duga S, Merlini PA, Correa A, Kessler T, Wilson JG, Bown MJ, Hall AS, Braund PS, Samani NJ, Schunkert H, Marrugat J, Elosua R, McPherson R, Farrall M, Watkins H, Willer C, Abecasis GR, Felix JF, Vasan RS, Lander E, Rader DJ, Danesh J, Ardissino D, Gabriel S, Saleheen D, Kathiresan S, Consortium CH-HF, Consortium CAE. Phenotypic Characterization of Genetically Lowered Human Lipoprotein(a) Levels. J Am Coll Cardiol. 2016;68(25):2761-2772.
  151. Bourgeois R, Devillers R, Perrot N, Despres AA, Boulanger MC, Mitchell PL, Guertin J, Couture P, Boffa MB, Scipione CA, Pibarot P, Koschinsky ML, Mathieu P, Arsenault BJ. Interaction of Autotaxin With Lipoprotein(a) in Patients With Calcific Aortic Valve Stenosis. JACC Basic Transl Sci. 2020;5(9):888-897.
  152. Mathieu P, Boulanger MC. Autotaxin and Lipoprotein Metabolism in Calcific Aortic Valve Disease. Front Cardiovasc Med. 2019;6:18.
  153. Grundy SM, Stone NJ, Bailey AL, Beam C, Birtcher KK, Blumenthal RS, Braun LT, de Ferranti S, Faiella-Tommasino J, Forman DE, Goldberg R, Heidenreich PA, Hlatky MA, Jones DW, Lloyd-Jones D, Lopez-Pajares N, Ndumele CE, Orringer CE, Peralta CA, Saseen JJ, Smith SC, Jr., Sperling L, Virani SS, Yeboah J. 2018 AHA/ACC/AACVPR/AAPA/ABC/ACPM/ADA/AGS/APhA/ASPC/NLA/PCNA Guideline on the Management of Blood Cholesterol: Executive Summary: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. J Am Coll Cardiol. 2019;73(24):3168-3209.
  154. Wilson DP, Jacobson TA, Jones PH, Koschinsky ML, McNeal CJ, Nordestgaard BG, Orringer CE. Use of Lipoprotein(a) in clinical practice: A biomarker whose time has come. A scientific statement from the National Lipid Association. J Clin Lipidol. 2019;13(3):374-392.
  155. Pearson GJ, Thanassoulis G, Anderson TJ, Barry AR, Couture P, Dayan N, Francis GA, Genest J, Gregoire J, Grover SA, Gupta M, Hegele RA, Lau D, Leiter LA, Leung AA, Lonn E, Mancini GBJ, Manjoo P, McPherson R, Ngui D, Piche ME, Poirier P, Sievenpiper J, Stone J, Ward R, Wray W. 2021 Canadian Cardiovascular Society Guidelines for the Management of Dyslipidemia for the Prevention of Cardiovascular Disease in Adults. Can J Cardiol. 2021;37(8):1129-1150.
  156. Li JJ, Ma CS, Zhao D, Yan XW, Beijing Heart S, Expert C. Lipoprotein(a) and Cardiovascular Disease in Chinese Population: A Beijing Heart Society Expert Scientific Statement. JACC Asia. 2022;2(6):653-665.
  157. Kronenberg F, Mora S, Stroes ESG. Consensus and guidelines on lipoprotein(a) - seeing the forest through the trees. Curr Opin Lipidol. 2022;33(6):342-352.
  158. Mach F, Baigent C, Catapano AL, Koskinas KC, Casula M, Badimon L, Chapman MJ, De Backer GG, Delgado V, Ference BA, Graham IM, Halliday A, Landmesser U, Mihaylova B, Pedersen TR, Riccardi G, Richter DJ, Sabatine MS, Taskinen MR, Tokgozoglu L, Wiklund O, Group ESCSD. 2019 ESC/EAS Guidelines for the management of dyslipidaemias: lipid modification to reduce cardiovascular risk. Eur Heart J. 2020;41(1):111-188.
  159. Anderson TJ, Gregoire J, Pearson GJ, Barry AR, Couture P, Dawes M, Francis GA, Genest J, Jr., Grover S, Gupta M, Hegele RA, Lau DC, Leiter LA, Lonn E, Mancini GB, McPherson R, Ngui D, Poirier P, Sievenpiper JL, Stone JA, Thanassoulis G, Ward R. 2016 Canadian Cardiovascular Society Guidelines for the Management of Dyslipidemia for the Prevention of Cardiovascular Disease in the Adult. Can J Cardiol. 2016;32(11):1263-1282.
  160. Joint committee for guideline r. 2016 Chinese guidelines for the management of dyslipidemia in adults. J Geriatr Cardiol. 2018;15(1):1-29.
  161. Guan W, Cao J, Steffen BT, Post WS, Stein JH, Tattersall MC, Kaufman JD, McConnell JP, Hoefner DM, Warnick R, Tsai MY. Race is a key variable in assigning lipoprotein(a) cutoff values for coronary heart disease risk assessment: the Multi-Ethnic Study of Atherosclerosis. Arterioscler Thromb Vasc Biol. 2015;35(4):996-1001.
  162. Ding L, Song A, Dai M, Xu M, Sun W, Xu B, Sun J, Wang T, Xu Y, Lu J, Wang W, Bi Y, Ning G. Serum lipoprotein (a) concentrations are inversely associated with T2D, prediabetes, and insulin resistance in a middle-aged and elderly Chinese population. J Lipid Res. 2015;56(4):920-926.
  163. Schwartz GG, Ballantyne CM. Existing and emerging strategies to lower Lipoprotein(a). Atherosclerosis.2022;349:110-122.
  164. Burgess S, Ference BA, Staley JR, Freitag DF, Mason AM, Nielsen SF, Willeit P, Young R, Surendran P, Karthikeyan S, Bolton TR, Peters JE, Kamstrup PR, Tybjaerg-Hansen A, Benn M, Langsted A, Schnohr P, Vedel-Krogh S, Kobylecki CJ, Ford I, Packard C, Trompet S, Jukema JW, Sattar N, Di Angelantonio E, Saleheen D, Howson JMM, Nordestgaard BG, Butterworth AS, Danesh J, European Prospective Investigation Into C, Nutrition-Cardiovascular Disease C. Association of LPA Variants With Risk of Coronary Disease and the Implications for Lipoprotein(a)-Lowering Therapies: A Mendelian Randomization Analysis. JAMA Cardiol. 2018;3(7):619-627.
  165. Lamina C, Kronenberg F, Lp GC. Estimation of the Required Lipoprotein(a)-Lowering Therapeutic Effect Size for Reduction in Coronary Heart Disease Outcomes: A Mendelian Randomization Analysis. JAMA Cardiol.2019;4(6):575-579.
  166. Kronenberg F. Therapeutic lowering of lipoprotein(a): How much is enough? Atherosclerosis. 2019;288:163-165.
  167. Madsen CM, Kamstrup PR, Langsted A, Varbo A, Nordestgaard BG. Lipoprotein(a)-Lowering by 50 mg/dL (105 nmol/L) May Be Needed to Reduce Cardiovascular Disease 20% in Secondary Prevention: A Population-Based Study. Arterioscler Thromb Vasc Biol. 2020;40(1):255-266.
  168. de Boer LM, Oorthuys AOJ, Wiegman A, Langendam MW, Kroon J, Spijker R, Zwinderman AH, Hutten BA. Statin therapy and lipoprotein(a) levels: a systematic review and meta-analysis. Eur J Prev Cardiol. 2022;29(5):779-792.
  169. Willeit P, Ridker PM, Nestel PJ, Simes J, Tonkin AM, Pedersen TR, Schwartz GG, Olsson AG, Colhoun HM, Kronenberg F, Drechsler C, Wanner C, Mora S, Lesogor A, Tsimikas S. Baseline and on-statin treatment lipoprotein(a) levels for prediction of cardiovascular events: individual patient-data meta-analysis of statin outcome trials. Lancet. 2018;392(10155):1311-1320.
  170. Leiter LA, Teoh H, Kallend D, Wright RS, Landmesser U, Wijngaard PLJ, Kastelein JJP, Ray KK. Inclisiran Lowers LDL-C and PCSK9 Irrespective of Diabetes Status: The ORION-1 Randomized Clinical Trial. Diabetes Care.2019;42(1):173-176.
  171. Bergmark BA, O'Donoghue ML, Murphy SA, Kuder JF, Ezhov MV, Ceska R, Gouni-Berthold I, Jensen HK, Tokgozoglu SL, Mach F, Huber K, Gaciong Z, Lewis BS, Schiele F, Jukema JW, Pedersen TR, Giugliano RP, Sabatine MS. An exploratory analysis of proprotein convertase subtilisin/kexin type 9 inhibition and aortic stenosis in the FOURIER trial. JAMA Cardiol. 2020;5(6):709-713.
  172. Chemello K, Chan DC, Lambert G, Watts GF. Recent advances in demystifying the metabolism of lipoprotein(a). Atherosclerosis. 2022;349:82-91.
  173. Santos RD, Raal FJ, Catapano AL, Witztum JL, Steinhagen-Thiessen E, Tsimikas S. Mipomersen, an antisense oligonucleotide to apolipoprotein B-100, reduces lipoprotein(a) in various populations with hypercholesterolemia: results of 4 phase III trials. Arterioscler Thromb Vasc Biol. 2015;35(3):689-699.
  174. Waldmann E, Parhofer KG. Lipoprotein apheresis to treat elevated lipoprotein (a). J Lipid Res. 2016;57(10):1751-1757.
  175. Schettler VJJ, Neumann CL, Peter C, Zimmermann T, Julius U, Roeseler E, Heigl F, Grutzmacher P, Blume H, Vogt A, Scientific Board of GftGAWG. The German Lipoprotein Apheresis Registry (GLAR) - almost 5 years on. Clin Res Cardiol Suppl. 2017;12(Suppl 1):44-49.
  176. Julius U, Tselmin S, Schatz U, Fischer S, Birkenfeld AL, Bornstein SR. Actual situation of lipoprotein apheresis in patients with elevated lipoprotein(a) levels. Atheroscler Suppl. 2019;40:1-7.
  177. Jaeger BR, Richter Y, Nagel D, Heigl F, Vogt A, Roeseler E, Parhofer K, Ramlow W, Koch M, Utermann G, Labarrere CA, Seidel D, Group of Clinical I. Longitudinal cohort study on the effectiveness of lipid apheresis treatment to reduce high lipoprotein(a) levels and prevent major adverse coronary events. Nat Clin Pract Cardiovasc Med. 2009;6(3):229-239.
  178. Leebmann J, Roeseler E, Julius U, Heigl F, Spitthoever R, Heutling D, Breitenberger P, Maerz W, Lehmacher W, Heibges A, Klingel R, ProLiFe Study G. Lipoprotein apheresis in patients with maximally tolerated lipid-lowering therapy, lipoprotein(a)-hyperlipoproteinemia, and progressive cardiovascular disease: prospective observational multicenter study. Circulation. 2013;128(24):2567-2576.
  179. Roeseler E, Julius U, Heigl F, Spitthoever R, Heutling D, Breitenberger P, Leebmann J, Lehmacher W, Kamstrup PR, Nordestgaard BG, Maerz W, Noureen A, Schmidt K, Kronenberg F, Heibges A, Klingel R, ProLiFe-Study G. Lipoprotein Apheresis for Lipoprotein(a)-Associated Cardiovascular Disease: Prospective 5 Years of Follow-Up and Apolipoprotein(a) Characterization. Arterioscler Thromb Vasc Biol. 2016;36(9):2019-2027.
  180. Moriarty PM, Gray JV, Gorby LK. Lipoprotein apheresis for lipoprotein(a) and cardiovascular disease. J Clin Lipidol. 2019;13(6):894-900.
  181. Perrot N, Verbeek R, Sandhu M, Boekholdt SM, Hovingh GK, Wareham NJ, Khaw KT, Arsenault BJ. Ideal cardiovascular health influences cardiovascular disease risk associated with high lipoprotein(a) levels and genotype: The EPIC-Norfolk prospective population study. Atherosclerosis. 2017;256:47-52.
  182. Bhatia HS. Aspirin and lipoprotein(a) in primary prevention. Curr Opin Lipidol. 2023;34(5):214-220.
  183. Tsimikas S, Karwatowska-Prokopczuk E, Gouni-Berthold I, Tardif JC, Baum SJ, Steinhagen-Thiessen E, Shapiro MD, Stroes ES, Moriarty PM, Nordestgaard BG, Xia S, Guerriero J, Viney NJ, O'Dea L, Witztum JL, Investigators AK-A-LS. Lipoprotein(a) Reduction in Persons with Cardiovascular Disease. N Engl J Med. 2020;382(3):244-255.
  184. O'Donoghue ML, Rosenson RS, Gencer B, Lopez JAG, Lepor NE, Baum SJ, Stout E, Gaudet D, Knusel B, Kuder JF, Ran X, Murphy SA, Wang H, Wu Y, Kassahun H, Sabatine MS, Investigators O-DT. Small Interfering RNA to Reduce Lipoprotein(a) in Cardiovascular Disease. N Engl J Med. 2022;387(20):1855-1864.
  185. Nissen SE, Wolski K, Balog C, Swerdlow DI, Scrimgeour AC, Rambaran C, Wilson RJ, Boyce M, Ray KK, Cho L, Watts GF, Koren M, Turner T, Stroes ES, Melgaard C, Campion GV. Single Ascending Dose Study of a Short Interfering RNA Targeting Lipoprotein(a) Production in Individuals With Elevated Plasma Lipoprotein(a) Levels. JAMA. 2022;327(17):1679-1687.
  186. Nicholls SJ, Nissen SE, Fleming C, Urva S, Suico J, Berg PH, Linnebjerg H, Ruotolo G, Turner PK, Michael LF. Muvalaplin, an Oral Small Molecule Inhibitor of Lipoprotein(a) Formation: A Randomized Clinical Trial. JAMA.2023;330(11):1042-1053.

Paget’s Disease of Bone

ABSTRACT

 

Sir James Paget described a skeletal disorder affecting one or more areas of the skeleton in 1876.  It is most common in England and in countries to which the English migrated. In recent years the prevalence in most countries has decreased. A common feature is skeletal deformity which evolves over many years and is most visible in the skull and lower extremities. Pathological fractures are most likely to occur in the femurs. Pain is a common feature in patients with Paget’s disease and may be of skeletal, joint, neurologic, or muscle origin. The radiologic features begin with a localized area of osteolysis which advances very slowly in the absence of therapy. Over time the lesion becomes osteosclerotic and once an entire bone is affected the entire lesion is sclerotic with areas of osteolysis remaining. Bone scans utilizing technetium99m-labeled bisphosphonates exhibit markedly increased uptake in the untreated state. Histologic evaluation of early lesions reveals an increased number of osteoclasts advancing at the interface of normal bone. They are often larger than normal and contain many more nuclei than normal osteoclasts. Subsequently numerous osteoblasts are found to be producing a large amount of disorganized bone. Associated with the increase in osteoclasts and osteoblasts there is a highly vascular fibrocellular marrow replacing the hematopoietic marrow. The osteoclasts have an abnormal ultrastructure featuring nuclear inclusions, and sometimes, cytoplasmic inclusions resembling nucleocapsid-like structures of the Paramyxoviridae family. Measurement of serum or urine N- or C-telopeptides documents the degree of bone resorption and serum total alkaline phosphatase activity, serum bone specific alkaline phosphatase and serum procollagen type 1 amino-propeptide document bone formation. Serum total alkaline phosphatase activity is the least expensive and most widely used test. Patients may develop sarcomas or giant cell tumors in affected bone but this is rare. Metabolic complications include hypercalcemia associated with immobilization and hyperuricemia and gout in patients with more extensive disease. Increased cardiac output may occur in patients with extensive disease due to the vascularity of the lesions. The earliest effective treatment was calcitonin but with the increased efficacy of the more potent bisphosphonates calcitonin is seldom prescribed. The treatment of choice is presently an intravenous infusion of 5 mg zoledronate. This normalizes bone resorption and formation markers for up to six and a half years in most patients. Indications for treatment include bone pain, hypercalcemia, neurologic deficits with vertebral disease, congestive heart failure, preparation for orthopedic surgery, and prevention of complications such as hearing loss and deformity. Surgery most commonly is needed for lower extremity joint replacement and correction of deformities of the lower extremity. The etiology remains somewhat controversial with some studies indicating a role for measles virus. The observation that the prevalence of the disease has decreased could be explained by the introduction of measles vaccine in 1963. Clearly genetic factors also play a role. Mutations in the sequestosome 1 gene produce susceptibility to develop Paget’s disease but not all family members with the mutation develop Paget’s disease. Many other gene abnormalities may also increase disease susceptibility.

 

HISTORICAL ASPECTS

 

In 1876, Sir James Paget (Figure 1), a prominent English surgeon, described five men who had at least two deformed areas of the skeleton (1). His description of the disorder he called osteitis deformans included clinical features, and gross and histologic pathology. He believed he was describing a rare inflammatory disorder, but by the start of the new century, numerous publications describing similar patients appeared in England, France, and the United States. A small number of reports also came from Australia, Germany, Holland, Italy, and Sweden. By this time, the condition commonly became known as Paget's disease of bone.

Figure 1. The bust of Sir James Paget in the Museum of St. Bartholomew's Hospital.

 

Further realization that Paget's disease was not a rare disorder came about after the discovery of X-rays in 1895 by Roentgen. It was then possible to detect affected bones, which exhibited no external manifestations of the disease. The first X-ray report appeared in 1896 (2) and osteolytic disease was recognized by1901 (3).

 

The first biochemical marker of Paget's disease was recognized in 1929 by Kay (4). He reported elevated alkaline phosphatase activity in the patients' sera. Over time, it came to be appreciated that serum alkaline phosphatase activity could reach higher levels in Paget's disease than in any other disorder.

 

EPIDEMIOLOGY

 

The distribution of Paget's disease throughout the world is one of its most striking features. While commonly found in the population of England, the United States, Australia, New Zealand, Canada, South Africa, and France, it appears to be rare throughout Asia and Scandinavia. Estimates of the prevalence in individual British cities even suggest a striking variability within one country (5). Analysis of hospital radiographs indicated prevalence ranging from 2.3% in Aberdeen, Scotland to 8% in Lancaster, England. Recently analysis of 1000 CT scans of the abdomen revealed a striking decrease in prevalence to 0.8% in the Lancashire region (6). The most recent prevalence estimate in the United States is 1-2% (7), and in France is 1.1-1.8% (8).  In many countries the prevalence of Paget’s disease appears to have decreased (9-13) although this has not been observed in Italy (14) or in the Salamanca province of Spain (15).   It is particularly difficult to obtain a true estimate of prevalence in a population as serum alkaline phosphatase activity may be elevated in as few as 14% of individuals with x-ray evidence of Paget’s disease (16).

 

Paget's disease probably occurs equally often in men and women and clearly increases in prevalence with age (17). The diagnosis is nearly always determined in individuals over the age of 50 years. The prevalence in the past approached 10% by 90 years and affected individuals are rarely discovered before 20 years.

 

The occurrence of Paget's disease in more than one member of a family was first reported in 1883 (18). Analysis of numerous kindreds indicates an autosomal dominant mode of inheritance (19). A positive family history of Paget's disease was reported in nearly 15% of patients in two large studies (20,21). In a clinic in Spain, 40% of the patients had at least one first-degree relative with Paget's disease after screening with bone scans (22). Gene analysis of Paget's disease kindreds will be discussed subsequently.

 

CLINICAL FEATURES

 

Paget's disease is a localized disorder of the skeleton with a wide range of skeletal involvement. One bone was affected in 5% of patients whereas the average number of lesions was about 6.5 per patient in a series of 197 patients (23). In a more recent study younger patients had a 47% prevalence of monostotic disease while 28% of older patients had monostotic disease (24).  In patients with familial disease there may be somewhat more bones affected than in patients with sporadic disease (25).

 

Deformity

 

A common feature of Paget's disease is skeletal deformity. This clearly evolves over a period of many years (probably decades) in most patients. The deformity is most visible in the skull and lower extremities.

 

Asymmetric enlargement of the cranium may first come to attention in those individuals who notice an increase in hat size. An increase in the size of superficial scalp veins, best appreciated over the frontal and temporal bones, is not uncommon. In patients with cranial enlargement, hearing loss is a common complication. Hearing loss correlates with loss of bone mineral density in the cochlear capsule (26). Inexplicably, despite the common skull involvement, Paget's disease is quite unusual in the facial bones. Facial disfigurement may be a consequence of enlargement of the maxilla and/or mandible and can be accompanied by spreading of the teeth, malocclusion, and loss of teeth (27).

 

One or, less often, two clavicles may become enlarged. An enlarged scapula is uncommonly appreciated perhaps because of its location.

 

The spine is a common source of morbidity from Paget's disease. The lumbar vertebrae and sacrum are most frequently affected. A single vertebra or multiple vertebrae may be involved. Over time, the vertebrae generally enlarge, but in some instances, vertebral compression may produce significant kyphosis.

 

Although Paget's disease is commonly found to affect the pelvis, only in its most severe form is it apparent on physical examination that the bone is thicker than normal. It is much easier to detect in the extremities, particularly when bowing of the femur and/or tibia is present (Figure 2). An increase in skin temperature is more readily detected over the tibia, a reflection of increased blood flow to the bone and surrounding soft tissue. Bowing of the upper extremity long bones is much less common than in the lower extremity, presumably because these are not weight-bearing bones.

Figure 2. Typical bowing of the leg due to Paget's disease involving the right tibia.

Pathological fractures in the lower extremity are most likely to occur in the femur and typically are transverse in nature (Figure 3). They are much more likely to result in nonunion than are tibial fractures (28).

Figure 3. Transverse fracture of the left femur.

 

Pain

 

Pain is a quite common symptom in patients with Paget's disease. It may be of skeletal, joint, neurologic, or muscle origin. Surprisingly, bone pain is usually absent even in patients with extensive disease or, when present, is mild to moderate in severity. The pain is usually dull in quality and often persists during the night. Weight bearing seldom produces a significant increase in bone pain.

 

Severe pain in a patient with Paget's disease is most likely to be due to osteoarthritis. This commonly occurs in the hip joint. Deterioration of the cartilage can occur when Paget's disease affects the acetabulum alone (Figure 4), but is likely to be more severe when both the acetabulum and head of the femur are affected by Paget's disease. If the femoral head is the only site of the disease, osteoarthritis is a less likely complication. A major feature of the pain in these patients is a significant increase in severity with weight bearing. In some patients, the combination of pain and impaired motion of the joint severely limits mobility. Knee pain and joint effusion may be prominent features in patients with bowing of the tibia. Back pain due to osteoarthritis also occurs in association with Paget's disease (Figure 5). Pain from osteoarthritis of the shoulder joint is relatively uncommon.

Figure 4. Paget's disease involving the left hemipelvis and right femur. There is severe osteoarthritis of the left hip but a relatively normal joint space in the right hip.

The most severe chronic pain in patients with Paget's disease is probably of neurologic origin. Pain from compression of the spinal cord or nerve roots may follow from enlargement of the vertebral bodies, pedicles, or laminae as well as from compression fractures. Pain from this source is more likely to arise from Paget's disease affecting the thoracic spine.

Figure 5. Patient with back pain who has multiple vertebrae affected by Paget's disease, large osteophytes, and narrowed disc spaces.

 

In a number of individuals, the weight of the skull may be so great that they have difficulty in keeping the head erect. This can produce neck pain and tension headaches due to muscle spasm. Deformity of the spine may also be associated with intermittent pain due to spasm of the paravertebral muscles.

 

RADIOLOGY

 

The radiologic features of Paget's disease include osteolytic, osteosclerotic, and mixed lesions.

The earliest lesions are osteolytic in character and are most readily appreciated in the skull (Figure 6). Circumscribed osteolytic skull lesions were called "osteoporosis circumscripta" by Schuller (29). These most often are seen in the frontal and occipital regions and with time may slowly coalesce. The other region where osteolytic lesions are commonly observed is the long bones of the lower extremity. The lesions usually arise at either end of the bone, seldom in the diaphysis. At the junction of the lesion with normal bone, the osteolytic lesion has the shape of a flame or inverted V (Figure 7). Such lesions have been noted to extend into normal bone at an average rate of about 1 cm per year (30).

Figure 6. Large osteolytic lesion in the skull of a woman with Paget's disease.

Figure 7. Osteolytic lesion of the distal left femur which is progressing proximally.

 

A heterogeneous region of osteosclerotic bone slowly develops in areas of the skeleton previously exhibiting a purely osteolytic character. This can be readily seen in long bones where the advancing front of osteolysis is trailed by patchy sclerosis superimposed on the earlier osteolytic process. With more time, the character of the bone may evolve into a dominant osteosclerotic appearance. This is often accompanied by periosteal new bone formation, which results in an increase in circumference of the bone. In the first observations reported by Paget, the thickness of the calvarium was fourfold greater than normal in one patient (1). The most severe skull involvement may be associated with basilar impression (Figure 8) which can produce compression of the structures in the posterior fossa resulting in ataxia, muscle weakness, and respiratory distress. With the evolution of the sclerotic phase of the disease, the lower extremity long bones often exhibit lateral and anterior bowing. Another radiologic feature in the long bones of the lower extremity is the presence of linear transverse radiolucencies in the cortex of the convex aspect of the bowed bone. These have been termed fissure fractures. Multiple fissure fractures may be seen. Although they usually remain stable, a small percent progress to complete transverse fractures.

Figure 8. Far advanced Paget's disease of the skull. Note the thickened inner and outer tables, the chaotic new bone deposition termed cotton-wool patch, and basilar impression.

 

It has been observed that after a dominant sclerotic lesion has developed, there may be secondary osteolytic lesions superimposed upon the sclerotic bone. These are most readily seen as clefts in the cortex of the long bones.

 

Computerized tomography (CT) and magnetic resonance imaging (MRI) are generally not needed in the evaluation of most patients (31).  CT may be needed to detect subtle fractures, spinal stenosis and secondary neoplasms.  MRI may be particularly useful in evaluating spinal complications.

 

The commercial availability of a technetium99m-labeled bisphosphonate in 1974 ushered in the era of routine use of bone scans in clinical medicine (32). In patients with Paget's disease, the affected bone has increased nuclide activity five minutes after intravenous administration of the bone-seeking tracer when compared with normal bone. The nuclide activity is 3-5 times higher than in normal bone. A bone scan is a very effective means of determining the extent of the disease and is clearly more sensitive than X-rays in determining the presence of small osteolytic areas of the disease (Figure 9). Since occult fractures and bone metastases may mimic some lesions of Paget's disease, it is necessary to do X-rays or CT scans of areas of increased nuclide uptake to distinguish the nature of the lesions. Very seldom is it necessary to do a bone biopsy to ascertain the diagnosis.

Figure 9. A technetium 99m-bisphosphonate bone scan of a patient with polyostotic Paget's disease.

 

In addition to the classical bone scan using a technetium-labeled bisphosphonate, gallium scans (33), fluorine-18-FDG PET scans (34), and Tl-201 scans (35) have been observed to delineate lesions of Paget's disease. In one study, the response to calcitonin treatment was more rapid with gallium scan than with a bone scan (33).

 

PATHOLOGY

 

Based on histological examinations of the interface of normal bone with an advancing osteolytic focus of Paget's disease, it has been concluded that the primary abnormality is a localized excess of osteoclastic bone resorption. An increased number of osteoclasts are present in Howship's lacunae in cortical and trabecular bone (Figure 10). They are frequently larger than normal and may have up to 100 nuclei in a single cross-section rather than the 3-10 found in normal osteoclasts (36). With progression of osteoclastic activity in the cortex, bone volume is reduced and individual osteons become confluent. The bone volume in trabecular bone of the medullary cavities is similarly reduced by osteoclastic activity. In association with the intense osteoclastic activity, the normal fatty or hematopoietic marrow is replaced by a fibrocellular stroma, which is highly vascular.

Figure 10. A bone biopsy of the iliac crest revealing an intense area of osteoclastic bone resorption. The osteoclasts are increased in size and have a greater number of nuclei than average.

 

In the mature lesion, there is a mixture of lamellar and woven bone, which transforms the matrix into a chaotic "mosaic" pattern of irregularly juxtaposed pieces of lamellar bone, interspersed with woven bone (Figure 11). The normal outer and inner circumferential lamellae and interstitial lamellae of the cortex are completely disrupted. Plump osteoblasts are found in large numbers on surfaces of new bone formation. There is an abundance of osteoid on bone surfaces but there is no increase in thickness of the osteoid seams (37). It has been noted that the size of the periosteocytic lacunae in the woven bone is greater than in the lamellar bone of Paget’s disease (37).  Since this is also the finding in woven bone from non-pagetic individuals the relevance of this observation is unclear.

Figure 11. A bone biopsy demonstrating the "mosaic" pattern of bone matrix in Paget's disease. Note the chaotic lamellar pattern intermixed with woven bone as demonstrated with polarized light.

 

There is some evidence of a "burned out" phase of Paget's disease in which the abnormal matrix persists but cellular activity is nearly absent and the marrow space is mainly filled with fat. It is more likely that such a finding does not occur throughout an entire lesion, but is found with all stages of the disease in a single bone.

 

Studies of the ultrastructure of osteoclasts in Paget's disease have demonstrated that many of these cells harbor microfilaments in the nucleus and occasionally, in the cytoplasm (38,39) (Figure 12). The microfilaments have the same structural features as nucleocapsids of viruses of the Paramyxoviridae family, a family of RNA viruses known to cause childhood infections such as measles and pneumonia due to respiratory syncytial virus. The nucleocapsid-like structures have not been found in osteoblasts, osteocytes, or bone marrow cells in the same specimens containing the osteoclast microfilaments. Identical microfilaments have been found in a small percentage of the osteoclasts in giant cell tumors of bone and in the osteoclasts of some patients with osteopetrosis and pyknodysostosis (40).

Figure 12. Electron microscopic examination of an iliac crest biopsy revealing microfilaments within remnants of a degenerating osteoclast nucleus. The arrow indicates microfilaments in adjacent cytoplasm. Magn. x 25,800

 

In addition to the structural evidence for the presence of viral nucleocapsids in the osteoclasts of Paget's disease, evidence of paramyxoviridae nucleocapsid proteins (41,42), and mRNA (43) has been reported, although not by all investigators (44). In one study addressing the identity of the osteoclast microfilaments, the full-length sequence for the measles virus nucleocapsid gene was delineated from the bone marrow of one patient as were more than 700 base pairs of the nucleocapsid gene in three additional patients (45).

 

BIOCHEMICAL ASSESSMENT

 

The radiologic and histologic evidence of increased bone resorption and formation in patients with Paget's disease is readily assessed by measuring biochemical markers of bone turnover. In general, these tests reflect the extent and activity of the disease.

 

Bone Resorption

 

Since the underlying cellular abnormality in Paget's disease seems to be increased bone resorption, one might expect that serum calcium and/or urinary calcium levels would be increased in some individuals with active Paget's disease. In the absence of fractures, immobilization, primary hyperparathyroidism, or bone metastases, this is not the case (46). Presumably, this is explained by a concomitant increase in bone formation, which has been defined by histopathology and by kinetic analysis of plasma disappearance rates and skeletal uptake of radiocalcium (46). Evidence of increased bone matrix resorption was first provided by the demonstration of increased urinary hydroxyproline excretion, a component of all types of collagen. Subsequently, more specific indices of bone resorption have been developed including pyridinoline, deoxypyridinoline, type I collagen N-telopeptide, and C-telopeptide. The latter two collagen components are the most specific markers of bone collagen resorption (47); serum and urine assays of the telopeptides are widely available for clinical use.

 

Bone Formation

 

Measurement of total serum alkaline phosphatase activity has been a means of evaluating Paget's disease for 90 years (4). The enzyme activity, which is localized in the plasma membrane of osteoblasts before extracellular release, correlates with the extent of the disease on skeletal surveys (48) and with parameters of bone resorption (48). The circulating enzyme activity usually increases gradually or does not change during long-term follow up of patients who are untreated (49). In patients with liver disease or who might be pregnant, it would be preferable to measure bone-specific alkaline phosphatase levels by immunoassay (50). Several of these assays have been developed which have little cross-reactivity with non-skeletal alkaline phosphatase. Measurement of serum procollagen type1-N-terminal peptide has proven to be valuable in assessing the response to teriparatide in osteoporosis patients. While these assays may have an advantage over the nonspecific total alkaline phosphatase activity with respect to specificity, no study has been done which indicates that they should replace this inexpensive assay for routine clinical use (51).

 

Other markers of bone formation such as serum osteocalcin or type I procollagen carboxyl-terminal peptide are not as sensitive as total or bone-specific alkaline phosphatase levels in assessing the response to therapy (47).

 

Sclerostin is an important protein produced by osteocytes which inhibits bone formation.  Serum sclerostin has been noted to be elevated in patients with Paget’s disease (52) but is not correlated with serum C-telopeptide or serum procollagen type1-N-terminal peptide levels.  The relevance of this finding remains to be established.

 

Calciotropic Hormones

 

Serum parathyroid hormone levels are generally normal in patients with Paget's disease (23). Elevated levels are found in the presence of concomitant primary hyperparathyroidism (53) and would be expected to also be increased in the presence of renal failure or vitamin D deficiency. Serum calcitonin levels are normal in Paget's disease (54) although there was prior speculation that low levels might contribute to the pathogenesis of the disease. In the absence of vitamin D deficiency, serum 25-hydroxy-vitamin D and 1, 25 dihydroxyvitamin D levels are normal. Inexplicably, 24,25-dihydroxyvitamin D levels have been reported to be low (55).

 

NEOPLASTIC COMPLICATIONS

 

Sarcoma

Sarcomas develop in the lesions of Paget's disease more often than in an age-matched normal population, although the incidence is less than 1% overall (56). However, in patients with extensive disease, the incidence has been estimated at 10% (57), although a subsequent study suggests this is not so (58). Rarely, sarcomas have been known to develop in multiple members of a family.

 

A sarcoma should be suspected when new pain and swelling develop in a bone previously affected by Paget's disease. The most common sites are in the pelvis, femur, humerus, skull, and facial bones.

 

There is a variable histology of the sarcomas of Paget's disease including osteosarcoma, fibrosarcoma, chondrosarcoma, and anaplastic sarcoma (57). Several types of histology may be present in a single tumor. Multinucleated giant cells (probably osteoclasts) may be scattered throughout a tumor. They may contain the nuclear microfilaments seen in the osteoclasts and are not thought to be neoplastic in nature (59).

 

Because of the underlying distortion of the pagetic bone, it is difficult to detect an early stage of a sarcoma. Typically, a radiolucent focus with speckled regions of calcification will be observed to disrupt the cortex of the bone (Figure 13). The best means of delineating the extent of the tumor mass is by CT or MRI.

Figure 13. Multiple sites of osteogenic sarcoma in a patient with Paget's disease of the right hemipelvis. Note the extension of the tumor through the cortex of right ischium.

 

Perhaps because of a failure of early diagnosis in most patients with sarcoma arising in Paget's disease, survival is brief. Only 7.5%-10% of patients survive five years and despite the multiple modalities of therapy presently available, the prognosis remains poor (56,60).

 

Giant Cell Tumor

 

Giant cell tumors of bone may arise in lesions of Paget's disease, often in the skull and facial bones (61). They are nearly always benign and appear to be less common than sarcoma in Paget's disease. As is the case with sarcoma, they rarely may appear in multiple family members who have Paget's disease (62).

 

A prominent feature of the tumors is the presence of large numbers of multinucleated giant cells, a small percentage of which contain the nuclear microfilaments typical of the osteoclasts of Paget's disease (61). The neoplastic component of the giant cell tumor is a spindle-shaped cell with fusiform nuclei and clumped chromatin. These cells rarely have mitoses. There may be some difficulty in distinguishing these tumors from giant cell reparative granulomas, which commonly arise in the jaws (63).

 

Giant cell tumors in Paget's disease are usually successfully treated with surgery and radiation therapy. In a few patients, high doses of dexamethasone have been shown to shrink the tumors (64).  Denosumab has been effective in treating patients with giant cell tumors (65) but has not been studied in patients with giant cell tumors arising in the lesions of Paget’s disease.

 

Other Neoplasia

 

Other neoplastic processes such as lymphoma (56), multiple myeloma (57), various carcinomas, and parathyroid tumors (53) have been reported in association with Paget's disease, but are probably chance occurrences. Metastatic cancers have been reported to metastasize to the highly vascular lesions of Paget's disease.

 

SYSTEMIC COMPLICATIONS AND ASSOCIATED DISEASES

 

Hypercalcemia

 

Hypercalcemia may occur as a consequence of immobilization in patients with Paget's disease (66), although this is an unusual clinical event. This is believed to occur because immobility results in increased bone resorption and decreased bone formation.

 

Hypercalcemia can also occur in association with a malignancy (67). More commonly hypercalcemia in Paget's disease occurs as a consequence of primary hyperparathyroidism (53). Correction of the hyperparathyroidism by surgery produced a decrease of 68% in plasma alkaline phosphatase in a series of 18 patients (53). The clinical features of these patients were quite similar to hyperparathyroid patients without Paget's disease, prompting the investigators to speculate that the two diseases were associated by chance.

 

Hyperuricemia and Gout

 

Hyperuricemia has been observed to be common in males with relatively severe Paget's disease (48). Clinical episodes of gouty arthritis occurred in almost half of these individuals. In a larger population of Paget's disease patients, hyperuricemia (20%) and gout (4%) were not felt to be increased in incidence (68). The differences in hyperuricemia and gout might be explained by the severity of the disease in the two study populations. With extensive skeletal involvement, a high turnover of nucleic acids in the lesions of Paget's disease could increase the urate pool enough to produce a clinical disturbance of urate metabolism (69).

 

Cardiovascular Dysfunction

 

A hallmark of the pathology of Paget's disease is the increased vascularity of affected bones. Further evidence for this has been documented by demonstration of an increase in blood flow to the extremities (70), although it has been suggested that this is mainly caused by cutaneous vasodilation (71). An echocardiographic study of cardiac function in Paget's disease found that patients with more severe disease had lower peripheral vascular resistance and higher stroke volume (72). These observations help account for the finding that patients with 15% or more of their skeleton affected by Paget's disease have increased cardiac output (73). High output congestive heart failure can occur.

 

It is possible that increased cardiac output in patients with Paget's disease accounts for an increased incidence in calcific aortic stenosis through causing turbulence across the valve. Patients with Paget's disease have a 4-6 times higher incidence of this lesion than control subjects (74,75). Calcification of the interventricular septum has also been reported in patients with Paget's disease and may be associated with complete heart block (75,76).  It also has been reported that arterial calcification is more common in Paget’s disease than in control subjects in the aorta as well as in iliac, femoral, gluteal and pelvic arteries (77). The explanation for this is unknown.

 

A less certain consequence of an increase in vascularity of bone and surrounding soft tissues is a variety of vascular steal syndromes. Patients with marked enlargement of the skull have been noted to be withdrawn, somnolent, and weak. These findings might be explained by shunting of blood from brain vessels to the external carotid artery system (78). It has also been proposed that spinal cord dysfunction might be a consequence of shunting of blood flow from the spinal arteries to the bone (79).

 

TREATMENT

 

Prior to 1975, a number of nonspecific treatments were used to attempt to alleviate some of the manifestations of Paget's disease. With the exception of pain medications none were of value. With the development of salmon calcitonin, a new era of effective treatment began. Presently, there are a number of highly effective agents which make possible excellent control of the disease.

 

Pretreatment Evaluation

 

The initial goal of patient evaluation is to establish which bones are affected by Paget's disease and what symptoms the lesions produce. A search for skeletal deformity may indicate one or more bones are involved, but this should be confirmed by X-rays. The full extent of the disease would best be ascertained by full body bone scan followed by radiologic confirmation of the disease in areas of increased tracer uptake. The decision as to which patient requires a bone scan is an individual one. For example, a 90-year old asymptomatic patient who is found to have Paget's disease in the pelvis during an intravenous pyelogram probably does not need a scan.

 

There is now a considerable choice of bone resorption and bone formation parameters which could be used to determine the overall metabolic activity of the disease. For routine clinical purposes, in most patients, measurement of total serum alkaline phosphatase activity is an effective and inexpensive test.

 

Drug Therapy

 

Calcitonin

 

Calcitonin is a peptide hormone whose main pharmacologic effect is rapid inhibition of bone resorption. This is mediated by binding of the hormone to its receptor on the surface of osteoclasts.

 

Salmon calcitonin was the first calcitonin species approved by regulatory agencies for treatment of Paget's disease. A dose of 50 to 100 U given daily or three times a week produces relief of bone pain in most patients within 2-6 weeks. Following suppression of the metabolic activity of the disease cardiac output is reduced (80) as is the skin temperature over affected tibiae. In addition, some patients have had dramatic improvement of neurologic deficits (81). Stabilization of hearing loss has also been noted (82). Because the drug has been shown to reduce the vascularity of bone affected by Paget's disease, it has been given preoperatively to reduce the degree of hemorrhage in patients scheduled for orthopedic procedures (83).

 

A single injection results in an immediate decrease in urinary hydroxyproline reflecting an acute inhibition of bone resorption. A maximal effect occurs in several months. Serum alkaline phosphatase activity falls more slowly; a significant decrease is generally not seen for one month. Within 3-6 months, both hydroxyproline excretion and alkaline phosphatase activity decrease on average by 50%. If treatment is stopped, urinary hydroxyproline gradually increases over several months followed by an increase in alkaline phosphatase activity back to pretreatment levels. With chronic treatment osteolytic lesions generally are reversed (84). However, if treatment is not continuous, the osteolytic lesion will recur. Reduced uptake of radiolabeled bisphosphonate (85) and gallium (33) occurs during long term treatment. Bone biopsies exhibit a reduced number of bone cells, a decrease in marrow fibrosis, and a reduction of woven bone volume (86).

 

Since salmon calcitonin is a foreign protein, it is not surprising that more than half of patients on long-term treatment develop specific antibodies against the hormone in the circulation (87). High titers of these antibodies almost always impair the response to continuing treatment so that up to 26% of patients have become resistant to the drug. Although no longer available for clinical use, human calcitonin was effective in inducing remissions in salmon calcitonin-resistant patients. Presently, any of the bisphosphonates can be used to treat these patients.

Salmon calcitonin injections may cause nausea and facial flushing in 10-20% of patients. Vomiting, abdominal pain, diarrhea, and polyuria are much less common side effects. Rarely tetany and allergic reactions have been reported. Nasal spray salmon calcitonin is much less likely to cause side effects but has lower potency (88). At this time, salmon calcitonin is used much less frequently than in the past because of the development of potent bisphosphonates.

 

Bisphosphonates

 

The development of the bisphosphonates for the treatment of skeletal disorders associated with increased bone resorption has been a major advance in the management of Paget's disease (89). These drugs, initially known as diphosphonates, are analogues of inorganic pyrophosphonate, a factor believed to be a necessary component for the mineralization of bone. All bisphosphonates have a central P-C-P core, which was substituted for the naturally occurring P-O-P core of pyrophosphate, because unlike P-O-P, the P-C-P structure is impervious to metabolic degradation. The bisphosphonates have a profound influence on bone metabolism, in part, because they bind to hydroxyapatite. The primary effect of bisphosphonates is to inhibit osteoclastic bone resorption, which in vivo is followed by a secondary decrease in bone formation. The earliest bisphosphonates which were developed, etidronate and clodronate, appear to achieve their effects by generating nonhydrolyzable analogues of adenosine triphosphate, while the later generation of more potent aminobisphosphonates, such as pamidronate and risedronate, inhibit protein prenylation through inhibition of farnesyl pyrophosphate synthase, a key enzyme in the mevalonate pathway. Although it is generally believed that bisphosphonates act directly on the differentiation and function of osteoclasts, evidence has accumulated which indicates that some bisphosphonates regulate cell proliferation, differentiation, and gene expression in human osteoblasts in vitro (90). How such observations translate into in vivo actions of bisphosphonates is unclear.

 

In table I, the bisphosphonates presently approved for treatment of Paget's disease in the United States are listed with their recommended regimes. There are four oral bisphosphonates available whose recommended daily treatment courses range from two months to six months and two intravenous bisphosphonates.

 

Table 1. Bisphosphonates Approved for Treatment of Paget's Disease

Bisphosphonates available in U.S.A.

 Administration and Dosage

 

Etidronate

 

Trade Name: Didronel®

 

FDA approval: 1977 

 

1. Tablet

2. 200 to 400 mg once daily for 6 months

200-400 mg dose is approved; 400 mg dose is preferred

3. Must be taken with 6-8 ounces of water on an empty stomach (no food, beverages, or medications for 2 hours before and after dose).

4. Course of Didronel® should not exceed 6 months.

5. Repeat courses can be given after rest periods of 3-6 months duration.

 

Pamidronate

 

Trade Name: Aredia®

 

FDA approval: 1994 

Generic available

1. Intravenous

2. Approved regimen is 30 mg intravenous infusion over 4 hours on 3 consecutive days

3. A more commonly used regimen is a 60 mg or 90 mg intravenous infusion over 2-4 hours and repeated as clinically indicated.

4. A single infusion is sometimes effective in mild disease; 2-3 or more infusions may be required in more severe disease.

5. A course of Aredia® may be readministered at intervals as needed.

 

Alendronate

 

Trade Name: Fosamax®

 

FDA approval: 1995 

Generic available

1. Tablet

2. 40 mg once daily for 6 months· Must be taken on an empty stomach, with 6-8 ounces of water, in the morning.

3. Wait at least 30 minutes after taking Fosamax® before eating any food, drinking anything other than tap water, or taking any medication.

4. Do not lie down for at least 30 minutes after taking Fosamax®. (Patient may sit.)

5. Available by mail order to the general public.

 

Tiludronate

 

Trade Name: Skelid®

 

FDA approval: 1997

 

1. Tablet

2. 400 mg (two 200 mg tablets) once daily for 3 months

3. Must be taken on an empty stomach with 6-8 ounces of water.

4. Skelid® may be taken any time of day, as long as there is a period of 2 hours before and after resuming food, beverages, and medications.

 

Risedronate

 

Trade Name: Actonel®

 

FDA approval: 1998 

 

1. Tablet

2. 30 mg once daily for 2 months

3. Must be taken on an empty stomach, with 6-8 ounces of water in the morning.

4. Wait at least 30 minutes after taking Actonel® before eating any food, drinking anything other than tap water, or taking any medication.

5. Do not lie down for at least 30 minutes after taking Actonel®. (Patient may sit.)

 

Zoledronic Acid

 

Trade Name: Reclast®

 

FDA approval: 2007

 

1. Intravenous

2. A 15 minute infusion of 5mg

3. Creatinine clearance must be >35 ml/min

4. Correct vitamin D deficiency and/or hypocalcemia before infusion

5. To reduce the risk of hypocalcemia after infusion, patients should receive 1500mg calcium and 1000 units vitamin D3 daily for two weeks

*Adapted from Information for Patients about Bisphosphonates, A Publication of the Paget Foundation for Paget's Disease of Bone and Related Disorders (2007).

 

The least potent bisphosphonate, etidronate, is similar to salmon calcitonin with respect to suppression of the metabolic activity of Paget's disease. The more potent aminobisphosphonates, pamidronate, alendronate, risedronate, and zoledronic acid can induce biochemical remissions in the majority of patients. In the past patients with extensive disease and markedly elevated biochemical parameters may have impressive reductions in serum alkaline phosphatase activity yet not reach normal levels (91). However, patients treated with zoledronic acid, no matter how high the baseline serum alkaline phosphatase activity, nearly always reach the normal range of enzyme activity (92). Most of the clinical benefits attributed to salmon calcitonin are produced by the aminobisphosphonates, yet it remains to be demonstrated whether long term biochemical remissions with any agent can reduce the incidence of future complications such as hearing loss and deformity.

 

The oral bisphosphonates are poorly absorbed and must be taken with water only. In clinical trials, side effects involving the gastrointestinal tract were not greater in patients receiving the drug than in the placebo group. However, some individuals experience abdominal distress or diarrhea. Patients receiving an oral aminobisphosphonate are advised to remain upright for at least 30 minutes after taking the drug to reduce the chance of esophageal irritation. A small percentage of patients may experience a transient increase in bone pain. The first infusion of pamidronate (93) or zoledronic acid (92) may produce an acute phase reaction in 30-50% of patients manifested by fever, myalgia, and elevation of circulating interleukin 6 levels (93).  Subsequent infusions produce little or no side effects. The mechanism responsible for the acute phase reaction appears to be release of cytokines from gamma delta T cells (94), which is worsened by vitamin D deficiency (95). Vitamin D supplementation in patients with low levels is very effective in preventing acute phase reactions and all patients who will be receiving pamidronate or zoledronic acid should have normal levels of serum 25OHD prior to the infusion (96). Allergic reactions to bisphosphonates are rare and most commonly manifest as inflammatory eye reactions due to pamidronate (97). If etidronate is used at a dose greater than 5 mg/kg body weight, osteomalacia may be a consequence (98). Another disadvantage of etidronate use is that osteolytic lesions may progress despite evidence of biochemical improvement (99).

 

Treatment with a potent bisphosphonate may produce long remissions. This is the most likely to be seen after treatment with zoledronic acid. A single infusion restores biochemical markers of bone turnover into the normal range and this is maintained for up to six and a half years in most patients (100). This response is largely independent of pretreatment disease activity. However, with the older bisphosphonates induction of a remission correlates well with the extent and activity (alkaline phosphatase) of the disease (101). Patients with less extensive disease and lower alkaline phosphatase activity are more likely to achieve remission. With respect to the duration of a remission, this appears to be dose-dependent as well as correlated with the nadir value of serum alkaline phosphatase activity, the number of affected bones, and the number of previous therapies (101).  Intravenous ibandronate may produce a prolonged response but is not an FDA-approved therapy for Paget’s disease (102).

 

Resistance to etidronate therapy is commonly seen after two six-month courses of the drug (103). There is also evidence that resistance to intravenous pamidronate (101) or clodronate (104) can occur. In pamidronate-resistant patients, treatment with alendronate was effective (104). In the clodronate-resistant patients, either risedronate or pamidronate was effective (105). There is no information which explains the mechanism responsible for apparent decreased efficacy of these agents with time. It is possible that an increase in the disease activity is responsible for these observations rather than a change in efficacy of the drugs.

Considerable publicity has been given to the development of osteonecrosis of the jaw in patients treated with bisphosphonates (106). This mainly is seen in cancer patients given monthly infusions and is rare in patients with Paget’s disease.

 

Miscellaneous Agents

 

Other inhibitors of bone resorption such as plicamycin and gallium nitrate, approved for treatment of hypercalcemia of malignancy, are effective in treating Paget's disease (107,108). In view of the safety and efficacy of the aminobisphosphonates, there is very little present use of these agents. The most potent antiresorptive agent, denosumab, has been reported to decrease disease activity in two patients with Paget’s disease (109, 110). In 1971 glucagon infusions were reported to markedly reduce of bone turnover parameters in four patients with Paget’s disease (111) but large trials have not been reported.

 

Treatment and Posttreatment Evaluation

 

Assessment of total serum alkaline phosphatase activity is generally sufficient to determine the success of treatment. The frequency of evaluation does not need to be more frequently than every 3 months after the onset of the treatment and can be extended to every 6-12 months after a nadir has been reached. Serial nuclear scans of the skeleton are more sensitive in defining no residual disease activity than biochemical results as reported in one study (112).  Minimal to significant disease activity was found in two-thirds of patients who had normal biochemical parameters after zoledronic acid infusions.  A second infusion produced complete remission.  It is uncertain how clinically important it is to produce complete suppression of radioisotope uptake in patients with normal biochemistry after treatment. If a patient has a well- defined osteolytic lesion on X-ray, it can be assessed annually to assure the disease is well-controlled. 

 

Indications for Treatment

 

Effective drug treatment for Paget's disease has evolved over 45 years, but there have been no large, randomized, long-term clinical trials, which can provide definitive guidelines for treatment. Nevertheless, in table 2, indications for drug treatment of Paget's disease are listed. These are based on a review of the literature and a large personal experience.

 

Table 2.  Indications for Drug Treatment of Paget's Disease

1. Bone pain

2. Hypercalcemia due to immobilization

3. Neurologic deficit associated with vertebral disease

4. High-output congestive heart failure

5. Preparation for orthopedic surgery

6. Prevention of complications including hearing loss, deformity

 

Although bone pain is not a problem in the majority of patients, it is a clear indication for treatment. In patients in whom bone pain is difficult to distinguish from joint pain treatment of the Paget's disease will usually clarify the source of pain. Treatment should also correct hypercalcemia in an immobilized patient, a rare situation. Neurologic deficits may improve with treatment, also a very unusual complication. High output heart failure should respond favorably to a treatment which lowers the cardiac workload. Reducing the vascularity of the bone and surrounding soft tissue before elective orthopedic surgery should reduce perioperative bleeding.

A major indication for treatment could be the prevention of future complications. There is some evidence that progression of hearing loss is reduced by treating patients with cranial disease. Prevention of deformity of lower extremity long bones and secondary osteoarthritis is a reasonable possibility. Presumably, early treatment would reduce the incidence of future fractures. It would be more speculative as to whether the incidence of sarcoma or giant cell tumor formation would be influenced.

 

To achieve the long-term goals of therapy such as prevention of future complications, it may be necessary to maintain the serum alkaline phosphatase activity within the normal range. Future very long-term studies would be needed to determine if complications can be abolished.

 

Surgery

 

In table 3, the various surgical procedures which have been utilized in the management of Paget's disease are listed.

 

Table 3. Surgical Procedures for Management of Paget's Disease

1. Total hip replacement

2. Total knee replacement

3. Femoral osteotomy

4. Tibial osteotomy

5. Suboccipital craniectomy and upper cervical vertebral laminectomy for basilar impression

6. Ventricular shunting for hydrocephalus

7. Stapes mobilization or stapedectomy

8. Surgery for correction of spinal stenosis or nerve root compression

 

Total hip replacement is probably the most common elective orthopedic procedure in patients with Paget's disease (113,114). Pain relief and improved mobility occur in a high percentage of patients. Postoperatively, heterotopic ossification may be somewhat more common, but is seldom a significant problem. For patients with severe osteoarthritis of the knees, total knee replacement is an effective treatment (115). Knee pain and joint effusions associated with osteoarthritis and tibial bowing may be effectively treated by tibular and fibular osteotomy (83).

 

There is much less experience with neurosurgical procedures in treating Paget's disease. However, successful relief of symptoms is expected after surgery for spinal stenosis or nerve root compression (116).  Percutaneous vertebroplasty might be considered in patients thought to have vertebral bone pain who do not respond to conservative therapy (117). Stapes mobilization or stapedectomy has not proven to be effective in improving hearing loss. One patient treated with cochlear implantation was reported to have improved speech perception (118).

 

ETIOLOGY

 

Slow Virus Infection

 

The possibility that Paget's disease fell into the category of a slow virus infection was suggested by the observation that the osteoclasts in this disorder harbored nuclear and cytoplasmic microfilaments which were essentially identical in structure to nucleocapsids of the Paramyxoviridae virus family (38,39). Immunochemical studies (40-42), and sequence analysis of nucleocapsid transcripts (43) have supported the initial hypothesis although not all studies have been positive with respect to a viral presence in the osteoclasts (44). Indirect support for the role of measles virus comes from a consideration of the availability of measles vaccine throughout the world (119). The vaccine was introduced in the United States in 1963, in Australia in 1967, in the United Kingdom and France in 1968, in New Zealand in 1969, in the Netherlands and Italy in 1976 and in Spain in 1978.  Availability of the vaccine for more than 50 years in several countries might explain a decrease in prevalence whereas delayed availability might explain why other regions may not have had a decrease as yet.

 

Non-Viral Environmental Influences

 

A number of reports have suggested that toxins such as arsenic and lead and animal exposure to dogs and cattle may be factors in the pathogenesis of Paget’s disease (119).  The most recent study indicates that exposure to woodburning during childhood, living near a mine, and hunting may be related to developing the disorder (120).  Exactly how these factors might produce pagetic lesions is unknown.

 

Genetics

 

Since there is clearly a familial aggregation of Paget's disease in up to 40% of patients with Paget's disease (17,19), a search for a predisposition gene or genes has been undertaken by a number of investigators. The initial attempts to define genetic susceptibility in Paget's disease centered on chromosome 6 because of known associations between disease susceptibility and histocompatibility loci on this chromosome (121). Although there is some evidence for human leucocyte antigen linkage in families with Paget's disease no gene locus has yet been defined on chromosome 6.

 

The initial localization of a predisposition gene for Paget's disease came from linkage studies with chromosome 18 markers (122,123). Attention was given to chromosome 18 because of the discovery that mutations of receptor activator of nuclear factor kB (RANK), a critical osteoclastogenic factor, were responsible for the skeletal disorder, familial expansile osteolysis (124), a condition which bears some resemblance to Paget's disease (125). Although chromosome markers have indicated linkage to Paget's disease in the region of the RANK gene on chromosome 18, no RANK mutations have been found in families of typical Paget's disease.

 

A second susceptibility locus, not associated with RANK, has been identified on chromosome 18 in a large Australian kindred (126). A further finding of interest on chromosome 18 is that sarcomas arising in Paget's disease may harbor a tumor suppressor gene in the same region as the first locus to be described (127).

 

In 2002 Laurin and colleagues (128) reported that mutations in the sequestosome 1 gene on chromosome 5 were associated with Paget’s disease in 11/24 French Canadian families and in 18/112 apparently sporadic patients. Mutations of this gene were subsequently found in families in the United Kingdom, Australia, New Zealand, the United States, and The Netherlands. Mutations have also been found in a smaller percentage of patients with sporadic disease. More than 20 different mutations have been described, nearly all of which are clustered around the ubiquitin binding domain of the sequestosome 1 protein (128-131). This protein modulates activity of the NF-κβ pathway, an important mediator of osteoclast function, and has also been implicated in the process of autophagy in osteoclasts (132).

 

A second gene abnormality has been described in the rare syndrome of inclusion body myopathy, frontotemporal dementia, and Paget’s disease (133).  More than 50 mutations have been identified in the VCP gene (134).  Only about 50% of the individuals with a mutation have demonstrable Paget’s disease (135).  VCP has a ubiquitin-binding domain as does sequestosome 1 and like sequestosome 1 is thought to play a role in autophagy (136). A search for VCP mutations in familial and sporadic Paget’s disease patients was negative in one study (137) and revealed a polymorphism associated with sporadic Paget’s disease in another study (138).

 

Further evidence of the heterogeneity of the genetics of Paget’s disease has come from studies reporting linkage of the disease with candidate loci at chromosome 5q31 and 5q35-qter (139), chromosome 2q36 (140), and chromosome 10p13 (140,141).

 

In a relatively small group of Paget’s disease patients, no mutations of the osteoprotegerin gene were found and a statistically significant increased frequency for the C allele in exon 2 was noted compared to control subjects (142). In a larger study a common polymorphism of the osteoprotegerin gene, G1181C, was found to predispose to the development of sporadic and familial Paget’s disease (143). Estrogen receptor-a and calcium-sensing receptor genotyping were significantly different in Paget’s disease versus control subjects in another study (144).

In the past 10 years 14 new susceptibility loci for Paget’s disease have been reported (145-158).  Most of these are likely to influence bone metabolism (CSF1, TNFRSF11A, PML, TM7SF4, UCMA/GRP, DKK1, CTHRC1, OPTN, RIN3, hnRNPA2B7, FKBP5, ZNF687, BER, C9ORF72 hexanucleotide repeat expansion frequency).  In one study single nucleotide polymorphisms were believed to amplify the effect of sequestosome 1 mutations and thereby magnify the severity of Paget’s disease (159).

 

In giant cell tumors arising in pagetic lesions H3F3A mutations have been detected and are associated with a higher number of osteoclast-like giant cells and an increased number of nuclei per cell (154) as compared with giant cell tumors occurring without Paget’s disease.

 

Clearly great progress has been made in studies of the genetics of Paget’s disease but the data suggest that an individual with a mutation has an increased susceptibility to develop particularly severe Paget’s disease but may not ever manifest evidence of the disorder. In a study of 84 offspring from 10 families whose Paget’s disease was associated with sequestosome 1 mutations, only 17% of the 23 offspring (mean age 45 years) who had mutations had evidence of the disease as indicated by bone scans (160). The offspring with normal scans had a mean age of 44 years and the mean age of the parents at the time of diagnosis was 48 years. There is incomplete penetrance of the Paget’s disease trait in these families although it is possible more offspring with aging will develop Paget’s disease in the future. In another study only 52% of patients with a VCP/p97 gene mutation were noted to have a lesion of Paget’s disease after extensive radiologic surveys (134). No assessment of viruses in the bone of these patients has been reported. In the most recent report from The Netherlands after 15.9 years of follow-up of sequestosome 1 mutation family members of pagetic patients only one individual (7.1%) was found to develop Paget’s disease (161). Fourteen individuals were followed and their ages ranged from 52 to 74 years.

 

Animal Models and In Vitro Models of Paget’s Disease 

 

Transgenic mice have been utilized to investigate the potential roles of the measles virus nucleocapsid gene and the sequestosome1/p62 and VCP/p97 gene mutations in the pathogenesis of Paget’s disease. Targeting of the measles virus nucleocapsid gene into osteoclasts of transgenic mice produced lesions in some vertebrae which strongly resembled the lesions of Paget’s disease, increased osteoclastic activity with exuberant new bone formation often of woven character was observed (162).  The same investigators targeted the most common sequestosome1/p62 mutation in familial Paget’s disease, P392L, into the osteoclasts of transgenic mice (163). They observed increased numbers of osteoclasts associated with bone loss but no increase in osteoblastic activity characteristic of Paget’s disease. Transduced osteoclast precursors isolated from the mice were hyperresponsive to receptor activator of NF-kappa B ligand (RANKL) and TNF-alpha but did not exhibit increased 1,25 (OH)2D3 responsivity, TAF(11)-17 expression, or increased number of nuclei per osteoclast, features found in osteoclast precursors isolated from individuals with Paget’s disease. In contrast to this study, Daroszewska and colleagues did find that targeting the P392L mutation into transgenic mice produced a Paget-like bone pathology predominantly in the lower limbs (164). They also found that osteoclast precursors had increased sensitivity to RANKL in vitro but did not examine 1,25(OH)2D3 sensitivity or TAF(11)-17 expression.  They found nuclear inclusions also but it was not certain that they were identical to those found in patients.  The explanation for the different results in these two studies is not apparent.

 

To examine the potential interaction of the measles virus nucleocapsid gene and the P392L mutation Kurihara and colleagues undertook studies of bone marrow specimens from patients with familial Paget’s disease who had the P392L mutation, utilizing specimens from pagetic and normal bone as well as from normal volunteers (165). The effects of antisense-measles virus nucleocapsid protein (MVNP) on osteoclast characteristics were different in marrow specimens from pagetic bone versus nonpagetic bone. The patient specimens which had MVNP expression responded to the antisense –MVNP with a reduction in osteoclast number, TATA box-binding protein associated factor 12 expression, 1,25 (OH)2D3 –stimulated IL-6 production and bone resorption, observations indicating a reversal of the usual features of osteoclasts generated from the marrow of sporadic or familial Paget’s disease patients. The results indicate an important role of measles virus in pagetic lesions.  A contribution of the P392L mutation to the pathologic process was suggested by the fact that there was hyperresponsiveness to RANKL in both the pagetic and nonpagetic bone marrows obtained from the familial patients.  These investigators then went on to carry out experiments in transgenic mice by cross breeding mice with MVNP and P392L mutations (165). The mice who harbored both MVNP and P392L had more severe Paget-like lesions than mice with MVNP alone.  As expected zoledronic acid treatment of p62P394L mice with a high rate of bone resorption produced marked suppression of bone resorption (166).

 

Two groups have generated transgenic mice expressing mutant forms of the VCP/p97 gene (167,168). In both studies increased osteoclastic activity appeared to be the dominant histologic feature with a modest amount of sclerotic bone being present.  No studies evaluating the characteristics of the osteoclast precursors were reported.

 

In a mouse study it was discovered that osteoclasts obtained from optineurin mutant mice have an increase in NF-kB activation and a reduction in response to RANKL as compared to wild-type mice (169). Bone histology revealed increased osteoclastic activity and increased bone formation but the overall histology was not typical of Paget’s disease.

 

A model to explain the coupling of bone resorption to bone formation in Paget’s disease has been developed utilizing osteoclasts from pagetic patients and transgenic mice harboring a knock-in of p62P394L (170).  It was concluded that in Paget’s disease, measles virus nucleocapsid protein upregulates IL-6 and IGF1 in osteoclasts to increase ephrinB2-EphB4 coupling and thereby promotes bone formation.  This is the first hypothesis to explain the masked level of bone formation in Paget’s disease.

 

REFERENCES

 

  1. Paget J. On a form of chronic inflammation of bones. (Osteitis deformans). Med Chir Trans. 1877;60:37.
  2. Leopold-Levi M, Londe A. Application des rayons de Roentgen a l'etude de la texture. N iconog. de la Salpetriere, Paris. 1897;X:198-201.
  3. Beclere M. Radiographie d'un cas de maladie de Paget. Bull Mem Soc Med Hop Paris. 1901;18:929-931.
  4. Kay H. Plasma phosphatase in osteitis deformans and in other diseases of bone. Brit J Exp Path. 1929;10:253.
  5. Barker DJ, Chamberlain AT, Guyer PB, Gardner MJ. Paget's disease of bone: the Lancashire focus. Br Med J. 1980;280:1105-1107.
  6. Abdulla O, Naqvi MJ, Shanshuddin S, Bukhari M, Proctor R. Prevalence of Paget’s disease of bone in Lancaster:  Time for an update. Rheumatology. 2019 57:931-932
  7. Altman RD, Bloch DA, Hochberg MC, Murphy WA. Prevalence of pelvic Paget's disease of bone in the United States. J Bone Miner Res. 2000;15:461-465.
  8. Lecuyer N, Grados F, Dargent-Molina P, Deramond H, Meunier PJ, Fardellone P. Prevalence of Paget's disease of bone and spinal hemangioma in French women older than 75 years: data from the EPIDOS study. Joint Bone Spine. 2000;67:315-318.
  9. Cundy HR, Gamble G, Wattie D, Rutland M, Cundy T. Paget’s disease of bone in New Zealand: Continued decline in disease severity. Calcif Tissue Int. 2004:75;358-364.
  10. Poor G, Donath J, Fornet B, Copper C. Epidemiology of Paget’s disease in Europe: the prevalence is decreasing. J Bone Miner Res. 2006;21(10):1545-9.
  11. Tiegs RD, Lohse CM, Wollan PC, Melton LJ. Long-term trends in the incidence of Paget's disease of bone. Bone. 2000;27:423-427.
  12. Corral-Gudino, L, Borao-Cengotita-Bengos, M, Del Pino-Montes, J, Ralston, S. Epidemiology of Paget’s disease of bone: a systemic review and meta-analysis of secular changes. Bone. 2013;55:347-352.
  13. Michou L, Orcel P. Has Paget’s bone disease become rare? Joint Bone Spine. 2019;86:538-541.
  14. Gennari, L, Merlotti, D, Martini, G, Nuti, R. Paget’s disease of bone in Italy.  J Bone Miner Res. 2006;21:14-21.
  15. Corral-Gudino, L, Secular changes in Paget’s disease:contrasting changes in the number of new referrals and in disease severity in two neighboring regions of Spain. Osteoporosis Int. 2013;443-450.
  16. Eekhoff ME, van der Klift M, Kroon HM, Cooper C, Hofman A, Pols HA, Papapoulos SE. Paget’s disease of bone in The Netherlands: a population-based radiological and biochemical survey—the Rotterdam Study. J Bone Miner Res. 2004:19;566-570.
  17. Schmorl G. Uber Osteitis deformans Paget. Virchows Arch. 1932;283:694-751.
  18. Pick. Osteitis deformans. Lancet. 1883;2:1125-1126.
  19. McKusick V. Heritable Disorders of Connective Tissue. St. Louis: Mosby; 1972.
  20. Sofaer JA, Holloway SM, Emery AE. A family study of Paget's disease of bone. J Epidemiol Community Health. 1983;37:226-231.
  21. Siris ES, Ottman R, Flaster E, Kelsey JL. Familial aggregation of Paget's disease of bone. J Bone Miner Res. 1991;6:495-500.
  22. Morales-Piga AA, Rey-Rey JS, Corres-Gonzalez J, Garcia-Sagredo JM, Lopez-Abente G. Frequency and characteristics of familial aggregation of Paget's disease of bone. J Bone Miner Res. 1995;10:663-670.
  23. Kanis J. Pathophysiology and treatment of Paget's disease of bone. 1991; Fogelman I, ed. London: Martin Dunitz Ltd.
  24. Haddaway MJ, Davie MW, McCall IW, Howdle S. Effect of age and gender on the number and distribution of sites in Paget’s disease of bone. Br J Radiol. 2007;80(955):532-6.
  25. Visconti MR, Langston AL, Goodman K, Selby PL, Fraser WD, Ralston SH. Mutations of SQSTM1 are associated with severity and clinical outcome in Paget disease of bone. J Bone Miner Res 2010: 25: 2368-2373.
  26. Monsell EM. The mechanism of hearing loss in Paget’s disease of bone. Laryngoscope. 2004;114;598-606.
  27. Smith BJ, Eveson JW. Paget's disease of bone with particular reference to dentistry. J Oral Pathol. 1981;10:233-247.
  28. Dove J. Complete fractures of the femur in Paget's disease of bone. J Bone Joint Surg Br. 1980;62-B:12-17.
  29. Schuller A. Dysostosis hypophysaria. Brit. J Radiol. 1926;31:156-158.
  30. Maldague B, Malghem J. Dynamic radiologic patterns of Paget's disease of bone. Clin Orthop. 1987; 217:126-151.
  31. Theodorou DJ, Theodorou SJ, Kakitsubata Y. Imaging of Paget disease of bone and its musculoskeletal complications: Review.  AJR Am J Roentgenol. 2011; (6 Suppl): 196: S64-75.
  32. Miller SW, Castronovo FP, Jr., Pendergrass HP, Potsaid MS. Technetium 99m labeled diphosphonate bone scanning in Paget's disease. Am J Roentgenol Radium Ther Nucl Med. 1974;121:177-183.
  33. Waxman AD, McKee D, Siemsen JK, Singer FR. Gallium scanning in Paget's disease of bone: effect of calcitonin. Am J Roentgenol. 1980;134:303-306.
  34. Cook GJ, Maisey MN, Fogelman I. Fluorine-18-FDG PET in Paget's disease of bone. J Nucl Med. 1997;38:1495-1497.
  35. Abamor E, Kitapci MT, Cila E, Gokcora N, Uluoglu O. Increased accumulation of Tl-201 in monostotic Paget's disease of the patella: evaluation with quantitative analysis. Clin Nucl Med. 2001;26:615-618.
  36. Rubinstein M, Smelin A, Freedman A. Osteoblasts and osteoclasts in bone marrow aspiration. Arch Intern Med. 1953;92:684-696.
  37. Meunier PJ, Coindre JM, Edouard CM, Arlot ME. Bone histomorphometry in Paget's disease. Quantitative and dynamic analysis of pagetic and nonpagetic bone tissue. Arthritis Rheum. 1980;23:1095-1103.
  38. Rebel A, Malkani K, Basle M. Nuclear anomalies in osteoclasts in Paget's bone disease. Nouv Presse Med. 1974;3:1299-1301.
  39. Mills BG, Singer FR. Nuclear inclusions in Paget's disease of bone. Science. 1976;194:201-202.
  40. Singer FR, Mills BG, Gruber HE, Windle JJ, Roodman GD. Ultrastructure of bone cells in Paget’s disease of bone. J Bone Miner Res. 2006;21(Supp 2):51-54.
  41. Rebel A, Basle M, Pouplard A, Kouyoumdjian S, Filmon R, Lepatezour A. Viral antigens in osteoclasts from Paget's disease of bone. Lancet. 1980;2:344-346.
  42. Mills BG, Singer FR, Weiner LP, Suffin SC, Stabile E, Holst P. Evidence for both respiratory syncytial virus and measles virus antigens in the osteoclasts of patients with Paget's disease of bone. Clin Orthop.1984;183:303-311.
  43. Reddy SV, Singer FR, Roodman GD. Bone marrow mononuclear cells from patients with Paget's disease contain measles virus nucleocapsid messenger ribonucleic acid that has mutations in a specific region of the sequence. J Clin Endocrinol Metab. 1995;80:2108-2111.
  44. Helfrich MH, Hobson RP, Grabowski PS, Zurbriggen A, Cosby SL, Dickson GR, Fraser WD, Ooi CG, Selby PL, Crisp AJ, Wallace RG, Kahn S, Ralston SH. A negative search for a paramyxoviral etiology of Paget's disease of bone: molecular, immunological, and ultrastructural studies in UK patients. J Bone Miner Res. 2000;15:2315-2329.
  45. Freidrichs WE, Reddy S, V., Bruder JM, Cundy T, Cornish J, Singer FR, Roodman GD. Sequence analysis of measles virus nucleocapsid transcripts in patients with Paget's disease. J Bone and Miner Res. 2002;17:145-151.
  46. Nagant de Deuxchaisnes C, Krane S. Paget's disease of bone: Clinical and metabolic observations. Medicine. 1964;43:233-266.
  47. Randall AG, Kent GN, Garcia-Webb P, Bhagat CI, Pearce DJ, Gutteridge DH, Prince RL, Stewart G, Stuckey B, Will RK, Retallack RW, Price RI, Ward L. Comparison of biochemical markers of bone turnover in Paget disease treated with pamidronate and a proposed model for the relationships between measurements of the different forms of pyridinoline cross-links. J Bone Miner Res. 1996;11:1176-1184.
  48. Franck WA, Bress NM, Singer FR, Krane SM. Rheumatic manifestations of Paget's disease of bone. Am J Med. 1974;56:592-603.
  49. Woodard HQ, Marcove RC. A comparison of the chemistry of blood from bone and peripheral veins. Clin Orthop. 1969;66:254-264.
  50. Panigrahi K, Delmas PD, Singer F, Ryan W, Reiss O, Fisher R, Miller PD, Mizrahi I, Darte C, Kress BC, Christenson RH. Characteristics of a two-site immunoradiometric assay for human skeletal alkaline phosphatase in serum. Clin Chem. 1994;40:822-828.
  51. Reid IR, Davidson JS, Wattie D, Wu F, Lucas J, Gamble GD, Rutland MD, Cundy T. Comparative responses of bone turnover markers to bisphosphonate therapy in Paget’s disease of bone. Bone. 2004;35:224-230.
  52. Yavropoulou MP, van Lierop, AH, Hamdy NA, Rizzoli R, Papapoulos, SE. Serum sclerostin levels in Paget’s disease and prostate cancer with bone metastases with a wide range of bone turnover.  Bone. 2012; 51:153-157.
  53. Gutteridge DH, Gruber HE, Kermode DG, Worth GK. Thirty cases of concurrent Paget's disease and primary hyperparathyroidism: sex distribution, histomorphometry, and prediction of the skeletal response to parathyroidectomy. Calcif Tissue Int. 1999;65:427-435.
  54. Kanis JA, Heynen G, Walton RJ. Plasma calcitonin in Paget's disease of bone. Clin Sci Mol Med. 1977;52:329-332.
  55. Guillard-Cumming DF, Beard DJ, Douglas DL, Johnson SK, Lawson-Matthew PJ, Russell RG, Kanis JA. Abnormal vitamin D metabolism in Paget's disease of bone. Clin Endocrinol (Oxf). 1985;22:559-566.
  56. Hadjipavlou A, Lander P, Srolovitz H, Enker IP. Malignant transformation in Paget disease of bone. Cancer. 1992;70:2802-2808.
  57. Jaffe H. Metabolic, Degenerative, and Inflammatory Diseases of Bones and Joints. Philadelphia: Lea and Febiger; 1972.
  58. Deyrup AT, Montag AG, Inwards CY, Xu Z, Swee RG, Krishnan Unni K. Sarcomas arising in Paget disease of bone: a clinicopathologic analysis of 70 cases. Arch Pathol Lab Med. 2007:131:942-6.
  59. Seret P, Basle MF, Rebel A, Renier JC, Saint-Andre JP, Bertrans G, Audran M. Sarcomatous degeneration in Paget's bone disease. J Cancer Res Clin Oncol. 1987;113:392-399.
  60. Huvos AG. Osteogenic sarcoma of bones and soft tissues in older persons. A clinicopathologic analysis of 117 patients older than 60 years. Cancer. 1986;57:1442-1449.
  61. Singer FR, Mills BG. Giant cell tumor arising in Paget's disease of bone. Recurrences after 36 years. Clin Orthop. 1993:293-301.
  62. Rendina D, Gennari L, DeFilippo G, Merlotti D, De Campora E, Fazioli F, Scarano G, Nuti R, Strazzullo P, Mossetti G. Evidence for increased clinical severity of familial and sporadic Paget’s disease of bone in Campania, southern Italy. J Bone Miner Res. 2006: 21: 1828-1838.
  63. Upchurch KS, Simon LS, Schiller AL, Rosenthal DI, Campion EW, Krane SM. Giant cell reparative granuloma of Paget's disease of bone: a unique clinical entity. Ann Intern Med. 1983;98:35-40.
  64. Ziambaras K, Totty WA, Teitelbaum SL, Dierkes M, Whyte MP. Extraskeletal osteoclastomas responsive to dexamethasone treatment in Paget bone disease. J Clin Endocrinol Metab. 1997;82:3826-3834.
  65. Chawla S, Blay JY, Rutkowski P, Le Chesne A, Reichardt P, Gelderblom H, Grimer RJ, Choy E, Skubitz K, Seeger L, Schuetze SM,  Henshaw R, Dai T, Jandial D, Palmerini E. Denosumab in patients with giant cell tumour of bone: a multicenter, open-label, phase 2 study. Lancet Oncol. 2019;20:1719-1729.
  66. Reifenstein, EC Jr, Albright F. Paget's disease: Its pathologic physiology and the importance of this in the complications arising from fracture and immobilization. N Engl J Med. 1944;231:343-355.
  67. Rosenkrantz J, Gluckman E. Coexistence of Paget's disease of bone and multiple myeloma. AJR. 1957;78:30-38.
  68. Altman RD. Musculoskeletal manifestations of Paget's disease of bone. Arthritis Rheum. 1980;23:1121-1127.
  69. Fennelly JJ, Hogan A. Pseudouridine excretion--a reflection of high RNA turnover in Paget's disease. Ir J Med Sci. 1972;141:103-107.
  70. Edholm O, Howarth S. Studies on the peripheral circulation in osteitis deformans. Clin Sci. 1953;12:277-285.
  71. Heistad DD, Abboud FM, Schmid PG, Mark AL, Wilson WR. Regulation of blood flow in Paget's disease of bone. J Clin Invest. 1975;55:69-74.
  72. Morales-Piga AA, Moya JL, Bachiller FJ, Munoz-Malo MT, Benavides J, Abraira V. Assessment of cardiac function by echocardiography in Paget's disease of bone. Clin Exp Rheumatol. 2000;18:31-37.
  73. Arnalich F, Plaza I, Sobrino JA, Oliver J, Barbado J, Pena JM, Vasquez JJ. Cardiac size and function in Paget's disease of bone. Int J Cardiol. 1984;5:491-505.
  74. Strickberger SA, Schulman SP, Hutchins GM. Association of Paget's disease of bone with calcific aortic valve disease. Am J Med. 1987;82:953-956.
  75. Hultgren HN. Osteitis deformans (Paget's disease) and calcific disease of the heart valves. Am J Cardiol. 1998;81:1461-1464.
  76. Harrison C, Lennox B. Heart block in osteitis deformans. Br Heart J. 1948;10:167-176.
  77. Laroche M, Delmotte A. Increased arterial calcification in Paget’s disease of bone. Calcif Tissue Int. 2005;77:129-33.
  78. Blotman F, Blard JM, Labauge R, Simon L. [Ultrasonic study of the brain circulation in patients with Paget's disease. Preliminary results]. Rev Rhum Mal Osteoartic. 1975;42:647-651.
  79. Douglas DL, Duckworth T, Kanis JA, Jefferson AA, Martin TJ, Russell RG. Spinal cord dysfunction in Paget's disease of bone. Has medical treatment a vascular basis? J Bone Joint Surg Br. 1981;4:495-503.
  80. Woodhouse NJ, Crosbie WA, Mohamedally SM. Cardiac output in Paget's disease: response to long-term salmon calcitonin therapy. Br Med J. 1975;4:686.
  81. Chen JR, Rhee RS, Wallach S, Avramides A, Flores A. Neurologic disturbances in Paget disease of bone: response to calcitonin. Neurology. 1979;29:448-457.
  82. el Sammaa M, Linthicum FH, Jr., House HP, House JW. Calcitonin as treatment for hearing loss in Paget's disease. Am J Otol. 1986;7:241-243.
  83. Meyers MH, Singer FR. Osteotomy for tibia vara in Paget's disease under cover of calcitonin. J Bone Joint Surg Am. 1978;60:810-814.
  84. Nagant de Deuxchaisnes C, Maldague B, Malghem J, Devogelaer JP, Huax JP, Rombouts-Lindemans C. The action of the main therapeutic regimes on Paget's disease of bone with a note on the effect of vitamin D deficiency. Arthritis Rhem. 1980;23:1215-1234.
  85. Waxman AD, Ducker S, McKee D, Siemsen JK, Singer FR. Evaluation of 99mTc bisphosphonate kinetics and bone scans in patients with Paget’s disease before and after calcitonin treatment. Radiology. 1977;125:761-764.
  86. Fornasier VL, Stapleton K, Williams CC. Histologic changes in Paget's disease treated with calcitonin. Hum Pathol. 1978;9:455-461.
  87. Singer FR, Fredericks RS, Minkin C. Salmon calcitonin therapy for Paget's disease of bone. The problem of acquired clinical resistance. Arthritis Rheum. 1980;23:1148-1154.
  88. Nagant de Deuxchaisnes C, Devogelaer JP, Huaux JP, Dufour JP, Esselinckx W, Engelbeen JP, Stasse P, Hermans P, de Buisseret JP. New modes of administration of salmon calcitonin in Paget's disease. Nasal spray and suppository. Clin Orthop.1987;217:56-71.
  89. Ebetino FH, Hogan AM, Sun S, Tsoumpra MK, Duan X, Triffitt JT, Kwaasi AA, Dunford JE, Barnett BL, Oppermann U, Lundy MW, Boyde A, Kashemirov BA, McKenna CE, Russell RG. The relationship between the chemistry and biological activity of the bisphosphonates. Bone. 2011;49:20-33.
  90. Reinholz GG, Getz B, Pederson L, Sanders ES, Subramaniam M, Ingle JN, Spelsberg TC. Bisphosphonates directly regulate cell proliferation, differentiation, and gene expression in human osteoblasts. Cancer Res. 2000; 60:6001-6007.
  91. Cundy T, Wattie D, King AR. High-dose pamidronate in the management of resistant Paget's disease. Calcif Tissue Int. 1996;58:6-8.
  92. Reid IR, Miller P, Lyles K, Fraser W, Brown JP, Saidi Y, Mesenbrink P, Su G, Pak J, Zelenakas K, Luchi M, Richardson P, Hosking D. Comparison of a single infusion of zoledronic acid with risedronate for Paget’s disease. N Engl J Med. 2005;353:898-908.
  93. Schweitzer DH, Oostendorp-van de Ruit M, Van der Pluijm G, Lowik CW, Papapoulos SE. Interleukin-6 and the acute phase response during treatment of patients with Paget's disease with the nitrogen-containing bisphosphonate dimethylaminohydroxypropylidene bisphosphonate. J Bone Miner Res. 1995;10:956-962.
  94. Rossini M, Adami S, Viapiana O, Vella A, Fracassi E, Gatti D. Circulatin APDg gamma delta T cells and the risk of acute-phase response after zoledronic acid administration. J Bone Miner Res. 2011:
  95. Bertoldo F, Pancheri S, Zenari S, Boldini S, Giovanazzi B, Zanatta M, Valenti MT, Dalle Carbonare L, Lo Cascio V. Serum 25-hydroxyvitamin D levels modulate the acute-phase response associated with the first nitrogen-containing bisphosphonate infusion. J Bone Miner Res. 2010;25:447-454.
  96. Merlotti D, Rendina D, Muscariello, R, Picchoni T, Alessandri M, De Filippo M, Bianciardi S, Franci MB, Lucani B, Cenci S, Strazzullo P, Nuti R, Gennari L. Preventive role of vitamin D supplementation for acute phase reaction after bisphosphonate infusion in Paget’s disease. J Clin Endocrinol Metab . 2019 Oct 22 (Epub ahead of print).
  97. Macarol V, Fraunfelder FT. Pamidronate disodium and possible ocular adverse drug reactions. Am J Ophthalmol. 1994;118:220-224.
  98. Krane SM. Etidronate disodium in the treatment of Paget's disease of bone. Ann Intern Med. 1982;96:619-625.
  99. Dodd GW, Ibbertson HK, Fraser TR, Holdaway IM, Wattle D. Radiological assessment of Paget’s disease of bone after treatment with the bisphosphonates EHDP and APD. Br J Radiol 1987; 60: 849-860.
  100. Reid IR, Lyles K, Su G, Brown jp, Walsh jp, del Pino-Montes J, Miller PD, Fraser WD, Cafoncelli S, Bucci-Rechtweg C, Hosking DJ. A single infusion of zoledronic acid produces sustained remissions in Paget disease: data to 6.5 years.  J Bone Miner Res. 2011:26: 2261-2270.
  101. Eekhoff ME, Zwinderman AH, Haverkort DM, Cremers SC, Hamdy NA, Papapoulos SE. Determinants of induction and duration of remission of Paget’s disease of bone after bisphosphonate (olpadronate) therapy. Bone. 2003;33:831-838.
  102. Reid IR, Wattie D, Gamble GD, Kalluru R, Cundy T. Long-term effects of intravenous ibandronate in Paget’s disease of bone. Calcif Tissue Int. 2017;100:250-254.
  103. Meunier P, Ravault A. Treatment of Paget's disease with etidronate disodium. In Singer FR, Wallach S (eds): Paget's Disease of Bone, Clinical Assessment, Present and Future Therapy. New York: Elsevier; 1991:86-99.
  104. Gutteridge DH, Ward LC, Stewart GO, et al. Paget's disease: acquired resistance to one aminobisphosphonate with retained response to another. J Bone Miner Res. 1999;14 Suppl 2:79-84.
  105. Rendina D, Mossetti G, Viceconti R, Sorrentino M, Nunziata V. Risedronate and pamidronate treatment in the clinical management of patients with severe Paget’s disease of bone and acquired resistance to bisphosphonates. Calcif Tissue Int. 2004;75:189-196.
  106. Murad OM, Arora S, Farag AF, Guber HA. Bisphosphates and osteonecrosis of the jaw: a retrospective study. Endocr Pract. 2007;13:232-8.
  107. Ryan WG. Two decades of experience in the treatment of Paget's disease of bone with plicamycin (mithramycin). In Singer, FR, Wallach S (eds): Paget's Disease of Bone, Clinical Assessment, Present and Future Therapy. New York: Elsevier; 1991:176-190.
  108. Warrell RP, Jr., Bosco B, Weinerman S, Levine B, Lane J, Bockman RS. Gallium nitrate for advanced Paget disease of bone: effectiveness and dose-response analysis. Ann Intern Med. 1990;113:847-851.
  109. Schwarz P, Rasmussen AQ, Kvist TM, Andersen UB, Jorgenson NR. Paget’s disease of bone after treatment with denosumab: A case report. Bone. 2012;50:1023-1025.
  110. Reid IR, Sharma S, Kalluru R, Eagleton C. Treatment of Paget’s disease of bone with denosumab: Case report and literature review.  Calcif Tissue Int. 2016;99:322-325.
  111. Condon JR. Glucagon in the treatment of Paget’s disease of bone. Brit Med J. 1971:4:719-721.
  112. Reid IR, Maslowski K. Long-term bone scintigraphy results after intravenous zoledronate in Paget’s disease of bone. Calcif Tissue Int. 2017;101:43-49.
  113. Hanna SA, Dawson-Bowling S, Millington S, Bhumbra R, Achan P. Total hip arthroplasty in patients with Paget’s disease of bone: A systematic review. World J Orthop. 2018; 8:357-363.
  114. Wegrzyn J, Pibarot V, Chapurlat R, Carret J-P, Bejui-Hugues J, Guyen O. Cementless total hip arthroplasty in Paget’s disease of bone: a retrospective review. Int Orthop .2010; 34:1103-1109.
  115. Popat R, Tsitskaris K, Millington S, Dawson-Bowling S, Hanna SA. Total knee arthroplasty in patients with Paget’s disease of bone: A systematic review. World J Orthop. 2018; 9:229-234.
  116. Hartman JT, Dohn DF. Paget's disease of the spine with cord or nerve-root compression. J Bone Joint Surg Am. 1966;48:1079-1084.
  117. Pedicelli A, Papacci F, Leone A, De Simone C, Meglio M, Bonomo L, Colosimo C. Vertebroplasty for symptomatic monostotic Paget disease. J Vasc Interv Radiol. 2011;22:400-403.
  118. Wolfovitz A, Shihada R, Shpak T, Braun J, Luntz M. Cochlear implantation in a patient with Paget’s disease. Laryngoscope. 2011;121:358-360.
  119. Singer FR. Paget’s disease of bone-genetic and environmental factors. Nat Rev Endocrinol. 2015;11:662-671.
  120. Audet M-C, Jean S, Beaudain C, Guay-Belanger S, Dumont J, Brown JP, Michou L. Environmental factors associated with familial or nonfamial forms of Paget’s disease of bone. 2017;84:719-723.
  121. Leach RJ, Singer FR, Roodman GD. The genetics of Paget's disease of the bone. J Clin Endocrinol Metab. 2001;86:24-28.
  122. Cody JD, Singer FR, Roodman GD, Otterund B, Lewis TB, Leppert M, Leach RJ. Genetic linkage of Paget disease of the bone to chromosome 18q. Am J Hum Genet. 1997;61:1117-1122.
  123. Haslam SI, Van Hul W, Morales-Piga A, Balemans W, San-Millan JL, Nakatsuka K, Willems P, Haites NE, Ralston SH. Paget's disease of bone: evidence for a susceptibility locus on chromosome 18q and for genetic heterogeneity. J Bone Miner Res. 1998;13:911-917.
  124. Hughes AE, Ralston SH, Marken J, Bell C, MacPherson H, Wallace RG, van Hul W, Whyte MP, Nakatsuka K, Hovy L, Anderson DM. Mutations in TNFRSF11A, affecting the signal peptide of RANK, cause familial expansile osteolysis. Nat Genet. 2000;24:45-48.
  125. Wallace RG, Barr RJ, Osterberg PH, Mollan RA. Familial expansile osteolysis. Clin Orthop. 1989;248:265-277.
  126. Good DA, Busfield F, Fletcher BH, Duffy DL, Kesting JB, Andersen J, Shaw JT. Linkage of Paget Disease of Bone to a Novel Region on Human Chromosome 18q23. Am J Hum Genet. 2001;70:517-525.
  127. Nellissery MJ, Padalecki SS, Brkanac Z, Singer FR, Roodman GD, Unni KK, Leach RJ, Hansen MF. Evidence for a novel osteosarcoma tumor-suppressor gene in the chromosome 18 region genetically linked with Paget disease of bone. Am J Hum Genet. 1998;63:817-824.
  128. Laurin N, Brown JP, Morissette J, Raymond V. Recurrent mutation of the gene encoding sequestosome 1 (SQSTM1/p62) in Paget disease of bone. Am J Hum Genet. 2002; 70: 1582-1588.
  129. Rea SL, Walsh JP, Ward L, Yip K, Kent GN, Steer JH, Xu J, Ratajczak T. A novel mutation (K378X) in the sequestosome 1 gene associated with increased NF-kappaB signaling and Paget’s disease of bone with a severe phenotype. J Bone Miner Res. 2006;21(7):1136-45.
  130. Collet C, Michou L, Audran M, Chasseigneaux S, Hilliquin P, Bardin T, Lemaire I, Cornelis F, Launay JM, Orcel P, Laplanche JL. Paget’s disease of bone in the French population: novel SQSTM1 mutations, functional analysis, and genotype-phenotype correlations. J Bone Miner Res. 2007;22(2):310-317.
  131. Michou L, Morrissette J, Gagnon ER, Marquis A, Dellabadia M, Brown JP, Siris ES. Novel SQSTM1 mutations in patients with Paget’s disease of bone in an unrelated multiethnic American population.  Bone 2011:48:456-460.
  132. Hocking LJ, Mellis DJ, McCabe PS, Helfrich MH, Rogers MJ. Functional interaction between sequestosome-1/p62 and autophagy-linked FYVE-containing protein WDFY3 in human osteoclasts.  Biochem Biophys Res Commun. 2010;402;543-548.
  133. Watts GD, Wymer J, Kovach MJ, Mehta SG, Mumm S, Darvish D, Pestronk A, Whyte MP, Kimonis VE. Inclusion body myopathy associated with Paget disease of bone and frontotemporal dementia is caused by mutant valosin-containing protein. Nature Genetics. 2004;36:377-381. 
  134. Plewa J, Surampalli A, Wencel M, Milad M, Donkerervoort S, Caiozzo VJ, Goyal N, Mozaffar T, Kimonis V. A cross-sectional analysis of clinical evaluation in 35 individuals with mutations of the valosin-conatining protein gene. Neuromuscul Disord. 2018;28:778-786.
  135. Farpour F, Tehranzadeh J, Donkervoort S, Smith C, Martin B,, Vanjara P, Osann K, Kimonis VE. Radiological features of Paget disease of bone associated with VCP myopathy.  Skeletal Radiol. 2012; 41:329-337.
  136. Ju J-S, Weihl CC. Inclusion body myopathy, Paget’s disease of bone and fronto-temporal dementia: a disorder of autophagy. Hum Mol Genet. 2010;19: R38-R45.
  137. Lucas GJ, Mehta SG, Hocking LJ, Stewart TL, Cundy T, Nicholson GC, Walsh JP, Fraser WD, Watts GD, Ralston SH, Kimonis VE. Evaluation of the role of valosin-containing protein in the pathogenesis of familial and sporadic Paget’s disease of bone. Bone. 2006;38:280-5.
  138. Chung PY, Beyens G, de Freitas F, Boonen S, Geusens P, Vanhoenakker F, Verbruggen L, Van Offel J, Goemaere S, Zmierczak HG, Devogelaer JP, Van Hul W. Indications for a genetic association of a VCP polymorphism with the pathogenesis of sporadic Paget’s disease of bone, but not for TNFSF11 (RANKL) and IL-6 polymorphisms.  Mol Genet Metab. 2011;103:287-292.
  139. Laurin N, Brown JP, Lemainque A, Duchesne A, Huot D, Lacourciere Y, Drapeau G, Verrault J, Raymond V, Morissette J. Paget disease of bone: mapping of two loci at 5Q35-qter and 5q31. Am J Hum Genet. 2001;69:528-543.
  140. Hocking LJ, Herbert CA, Nicholls RK, Williams F, Bennett ST, Cundy T, Nicholson GC, Wuyts W, Van Hul W, Ralston SH. Genomewide search in familial Paget disease of bone shows evidence of genetic heterogeneity with candidate loci on chromosomes 2q36, 10p13, and 5q35. Am J Hum Genet. 2001;69:1055-1061.
  141. Lucas GJ, Riches PL, Hocking LJ, Cundy T, Nicholson GC, Walsh JP, Ralston SH. Identification of a major locus for Paget’s disease on chromosome 10p13 in families of british descent. J Bone Miner Res. 2008;23:58-63.
  142. Wuyts W, Van Wesenbeeck L, Morales-Piga A, Ralston S, Hocking L, Vanhoenacker R, Westhovens R, Verbruggen L, Anderson D, Hughes A, Van Hul W. Evaluation of the role of RANK and OPG genes in Paget's disease of bone. Bone. 2001;28:104-107.
  143. Daroszewska A, Hocking LJ, McGuigan FE, Langdahl B, Stone MD, Cundy T, Nicholson GC, Fraser WD, Ralston SH. Susceptibility to Paget’s disease of bone is influenced by a common polymorphic variant of osteoprotegerin. J Bone Miner Res. 2004;19:1506-1511.
  144. Donath J, Speer G, Poor G, Gergely P Jr, Tabak A, Lakatos P. Vitamin D receptor, oestrogen receptor-alpha and calcium-sensing receptor genotypes, bone mineral density and biochemical markers in Paget’s disease of bone. Rheumatology (Oxford). 2004;43:692-695.
  145. Chung PY, Beyens G, Boonen, S, Papapoulos S, Geusens P, Karperoen M, Vanhoenacker F, Verbruggen L, Fransen L, Van Offel J, Goemare S, Zmierczak HG, Westhovens R, Devogelaer JP, Van Hul W. The majority of the genetic risk for Paget’s disease of bone is explained by genetic variants close to the CSF1, OPTN, TMSF4, andTNFRSF11A genes. Hum Genet. 2010;128:615-626.
  146. Albagha OM, Visconti MR, Alonso N, Langston AL, Cundy T, Dargie R, Dunlop MG, Fraser WD, Hooper MJ, Isaia G, Nicholson GC, del Pino Montes J, Gonzalez-Sarmiento R, diStefano M, Teresa A, Walsh JP, Ralston SH. Genome-wide association study identifies variants at CSF1, OPTN and TNFRSF11A as genetic risk factors for Paget’s disease of bone. Nat Genet. 2010;42:520-524.
  147. Albagha OME, Wani SE, Visconti MR, Alonso N, Goodman K, Brandi ML, Cundy T, Chung PYJ, Dargie R, Devogelaer J-P, Falchetti A, Fraser WD, Gennari L, Gianfrancesco F, Hooper MJ, Van Hul W, Isaia G, Nicholson GC, Nuti R, Papapoulos S, del Pinto Morales J, Ratajczak T, Rea SL, Rendina D, Gonzalez-Sarmiento R, Di Stefano M, tifies Ward LC, Walsh JP. Genome-wide association identifies three new susceptibility loci for Paget’s disease of bone. Nat Genet. 2011;43:685-689.
  148. Michou L, Conceicao N, Morissette J, Gagnon E, Miltenberger-Miltenyi G, Siris ES, Brown JP, Cancela ML. Genetic association study of UCMA/GRP and OPTN genes (PDB6 locus) with Paget’s disease of bone. Bone. 2012;51:720-728.
  149. Beauregard M, Gagnon E, Guay-Belanger S, Morissette J, Brown JP, Michou l. Identification of rare genetic variants in novel loci associated with Paget’s disease of bone. Hum Genet. 2014;133:755-768.
  150. Beauregard M, Gagnon E, Guay-Belanger S, Siris ES, Morissette, J, Brown JP, Michou l. Genetic association study of Dickkopf 1 and sclerostin genes with Paget disease of bone. Calcif Tissue Int. 2013;93:405-412.
  151. Vallet M, Soares DC, Wani S, Sophocleus A, Warner J, Salter DM, Ralston SH, Albagha OME. Targeting sequencing of the Paget’s disease associated 14q32 locus identifies several missense coding variants in RIN3 that predispose to Paget’s disease of bone. Hum Mol Gen. 2015;24:3286-3295.
  152. De Ridder R, Boudin E, Vandeweyer G, Devogelaer J-P, Fransen E, Mortier G, Van Hul W. Calcif Tissue Int. 2019;104:613-621.
  153. Divisato G, Formicola D, Esposito T, Merlotti D, Pazzaglia L, Del Fattore A, Siris E, Orcel P, Brown JP, Nutti R, Strazzullo P, Benassi MS, Cancela ML, Michou L, Rendina D, Gennari L, Gianfrancesco F. ZNF687 mutations in severe Paget disease of bone associated with giant cell tumor, Am J Hum Gen. 2016;98:275-286.
  154. Divisato G, Scotto di Carlo F, Pazzzaglia L, Rizzo, R, Coviello DA, Benassi MS, Picci P, Esposito T, Gianfresco F.The distinct clinical features of giant cell tumor of bone in pagetic and non-pagetic patients are associated with genetic, biochemical and histological differences. Oncotarget. 2017;8:63121-63131.
  155. Qi X, Wang J, Zhao Z, Wang O, Xu L, Mao J, Jiang Y, Li M, Xing x, Yu W, Asan, Xia W. Familial early-onset Paget’s disease of bone associated with a novel hnRNPA2B1 mutation. Calcif Tissue Int. 2017;101:159-169.
  156. Lu B, Jiao Y, Wang Y, Dong J, Wei M, Cui B, Sun Y, Wang L, Zhang B, Chen Z, Zhao Y. A FKBP5 mutation is associated with Paget’s disease of bone and enhances osteoclastogenesis. Exp Mol Med. 2017;49:
  157. Usategui-Martin R, Gutierrez-Cerrajero C, Jimenez-Vazquez S, Calero-Paniagua I, Garcia-Aparicio J, Corral-Gudino L, DelPino-Montes J, Gonzalez-Sarmiento R. Polymorphisms in genes implicated in base excision repair (BER) pathway are associated with susceptibility to Paget’s disease of bone. Bone. 2018;112:19-23.
  158. Rubino E, DiStefano M, Galimberti D, Serpente M, Scarpente M, Scarpini E, Fenoglio C, Bo M,Rainero I. C90RF72 hexanucleotide repeat expansion frequency in patients with Paget’s disease of bone. Neurobiol Aging. 2019;
  159. Gianfrancesco F, Rendina D, DiStefano M, Mingione A, Esposito T, Merlotti D, Gallone S, Magliocca S, Goode A, Formicola D, Morello G, Layfield R, Frattini A, DeFilippo G, Nuti R, Searle M, Strazzullo P, Isaia G, Mossetti G, Gennari L. A nonsynonymous TNFRSF11A variation increases NFkB activity and the severity of Paget’s disease. J Bone Miner Res. 2012;27:443-452.
  160. Bolland MJ, Tong PC, Naot D, Callon KE, Wattie DJ, Gamble GD, Cundy T. Delayed development of Paget’s disease in offspring inheriting SQSTM1 mutations.  J Bone Miner Res 2007; 22:411-415.
  161. Peeters JMM, De Ridder R, Mamoen EC, Marelise E, Eckhoff W, Smit F, Boudin E, Van Hul W, Papapoulos SE, Appelman-Dijkstra, NM. Familial Paget’s disease of bone: Long-term fpllow-up of unaffected relatives with and without sequestome 1 mutations. Bone. 2019;Nov 1.
  162. Kurihara N, Zhou H, Reddy SV, Garcia Palacios V, Subler MA, Dempster DW, Windle JJ, Roodman GD. Expression of measles virus nucleocapsid protein in osteoclasts induces Paget’s disease-like bone lesions in mice. J Bone Miner Res. 2006; 21:446-455.
  163. Kurihara N, Hiruma Y, Zhou H, Subler MA, Dempster DW, Singer FR, Reddy SV, Gruber HE, Windle JJ, Roodman GD. Mutation of the sequestosome 1 (p62) gene increases osteoclastogenesis but does not induce Paget disease. J Clin Invest. 2007; 117: 133-142.
  164. Daroszewska A, van’t Hof RJ, Rojas JA, Layfield R, Landao-Basonga E, Rose K, Ralston SH. A point mutation in the ubiquitin-associated domain of SQSMT1 is sufficient to cause a Paget’s disease-like disorder in mice. Hum Mol Genet. 2011; 20: 2734-2744.
  165. Kurihara N, Hiruma Y, Yamana K, Michou L, Rousseau C, Morissette J, Galson DL, Teramachi J, Zhou H, Dempster DW, Windle JJ, Brown JP, Roodman GD. Contributions of the measles virus nucleocapsid gene and the SQSTM1/p62 (P392L) mutation to Paget’s disease. Cell Metab. 2011; 13: 23-34.
  166. Daroszewska A, Rose L, Sarsam N, Charlesworth G, Prior A, Rose K, Ralston SH, van ‘t Hof RJ. Zoledronic acid prevents pagetic-like lesions and accelerated bone loss in the p62P394L model of Paget’s disease. Dis Model Mech. 2018;Aug 23.
  167. Custer SK, Neumann M, Lu H, Wright AC, Taylor JP. Transgenic mice expressing mutant forms VCP/p97 recapitulate the full spectrum of IBMPFD including degeneration in muscle, brain and bone. Hum Mol Genet. 2010; 19: 1741-1755.
  168. Badadani M, Nalbandian A, Watts GD, Vesa J, Kitazawa M, Su H, Tanaja J, Dec E, Wallace DC, Mukherjee J, Caiozzo V, Warman M, Kimonis VE. VCP associated inclusion body myopathy and Paget disease of bone knock-in mouse model exhibits tissue pathology typical of human disease. PLoS ONE. 2010; 5:1-15.
  169. Obaid R, Wani SE, Azfer A, Hurd T, Jones R, Cohen P, Ralston SH, Albagha OME. Optineurin negatively regulates osteoclast differentiation by modulating NF-kB and interferon signaling: Implications for Paget’s disease. Cell Reports. 2015;13:1096-1102.
  170. Teramachi J, Nagata Y, Mohammad K, Inagaki Y, Ohata Y, Guise T, Michou L, Brown JP, Windle JJ, Kurihara N, Roodman GD. Measles virus nucleocapsid protein increases osteoblast differentiation in Paget’s disease. J Clin Invest. 2016;126:1012-1022.

 

The Epidemiology and Pathogenesis of Osteoporosis

ABSTRACT

 

Osteoporosis is a multifactorial disorder associated with low bone mass and enhanced skeletal fragility. Although most prevalent in older females, some men are also at high risk. Risk factors in men and women include smoking, family history of fracture, age greater than 65 years, and low but also high BMI particularly in men.  Secondary causes of osteoporosis include chronic treatment with glucocorticoids, gastrointestinal disorders, diabetes mellitus (T1D, T2D), rheumatoid arthritis, liver disease, gluten enteropathy, multiple myeloma and other hematologic disorders.  However, primary osteoporosis is most often related to either postmenopausal estrogen loss or age-related deterioration of skeletal microarchitecture; both are due to uncoupling in the bone remodeling unit. Reduced bone formation with age is almost certainly a function of impaired stem cell differentiation into the osteoblast lineage with a resultant increase in marrow adipogenesis. Increased bone resorption also characterizes most forms of osteoporosis but the etiology is multifactorial. Changes in local and systemic growth factors are often responsible for uncoupling between resorption and formation. However, alterations in peak bone acquisition contribute years later to low bone mass and enhanced skeletal fragility. Fracture risk assessment tools (e.g. FRAX) in handheld apps and computers which combine bone density score and risk factors, have provided rapid assessments of future osteoporotic fractures and can be performed at the bedside. Newer methods of measuring bone quality have led to insights into micro-architectural deterioration that contributes to skeletal fragility. Notwithstanding, low areal bone mineral density by DEXA remains the strongest predictor of subsequent fracture beyond age, and this is potentially measurable in everyone after age 65.

 

INTRODUCTION

 

Osteoporosis is a disorder characterized by reduced bone mass, impaired bone quality, and a propensity to fracture. For decades, this disease was considered a syndrome characterized by back pain, vertebral fractures, and osteopenia on plain films. Identifying secondary causes of low bone mass was the principle objective of most clinicians. However, osteoporosis is now classified as a primary disorder of the skeleton related to profound metabolic changes not only in bone but also related to changes in whole body homeostasis. Significant progress has been made in both defining this disorder and in understanding its complex pathogenesis. In addition, a consensus has emerged concerning the strength of the association between low bone mineral density and fracture risk, and the importance of qualitative aspects of the skeleton, as additional risk determinants.

 

Dual energy x-ray absorptiometry (subsequently referred to as DEXA) revolutionized our ability to predict fractures in large numbers of subjects by measuring areal bone mineral density (subsequently referred to as BMD).  Virtually all population studies have confirmed that for every one standard deviation below young normal mean bone mineral density (at virtually any skeletal site) there is a nearly two-fold greater risk of a subsequent hip fracture (1). Some clinicians, as well as the WHO and the National Osteoporosis Foundation define osteoporosis purely on a bone mineral density (BMD) T-score more than 2.5 standard deviations below young normal reference ranges for the spine, hip or radius (2). Although this "bar" has been used to establish prevalence estimates and to define high risk individuals who should be considered for treatment, it is also evident that even this definition demands a better understanding of the pathophysiologic processes that result in low bone mass, and a more thorough review of 'bone quality'. Indeed, despite the strength of the association between BMD and fracture risk, qualitative measurements of the skeleton, such as bone turnover, mineralization, and trabecular connectivity also contribute to risk. In this chapter, the mechanisms responsible for altering bone microarchitecture and strength in such a way as to enhance the likelihood of fragility fractures will be reviewed. Irrespective of the epidemiology and pathogenesis of osteoporosis, the stark fact remains that this disease has significant morbidity, mortality, and economic costs. Just as importantly, understanding how this disorder develops and progresses, has important socio- economic as well as medical consequences.

 

EPIDEMIOLOGY OF OSTEOPOROSIS

 

Estimating exactly how many women have osteoporosis depends on the working definition of this disease and the appropriate diagnostic criteria (3). Prior to the widespread application of DEXA, osteoporosis was rarely diagnosed and then only in women with symptomatic vertebral fractures or osteopenia noted by x-ray for other reasons. Indeed, for too long, hip fractures, the end point of this metabolic syndrome, were either written off as a consequence of aging, or ignored in respect to treatment. BMD measurements by DEXA and CT changed all that, especially when it became clear that a single BMD measurement at any site was a very strong predictor of future spine and hip fractures (1,2,4). As such, the definition of osteoporosis began to evolve, and estimates of how many people were affected also changed. When the World Health Organization (WHO) set a cut off point of 2.5 standard deviations below a young normal mean value for BMD in the spine or hip of postmenopausal women, as an indicator of osteoporotic risk, estimates of disease prevalence increased (5). These were confirmed by publication of larger epidemiologic studies in women such as The Study of Osteoporotic Fractures (SOF) and MrOs, a large international cohort of men which provided better estimates of disease prevalence, onset and clinical course (4,6).

 

Currently most estimates suggest that there are approximately 0.3 million hip fractures per annum in the U.S. and 1.7 million hip fractures in Europe (7,8).  With the introduction of readily available treatments, and clear prevention messaging annual hip fracture rates in the early 2000s started to decline. However, by 2015 those rates had flattened out and were trending upwards, following widespread reporting of atypical femoral fractures in patients treated with bisphosphonates and denosumab. Virtually all hip fractures can be attributed to osteoporosis, whether primary or secondary. Moreover, in most if not all cases, falls are a primary event leading to the fracture. The female to male ratio of hip fractures is approximately 2:1.0 (6,9). Not surprisingly, the occurrence of these fractures increases exponentially with age. In contrast, the incidence of wrist fractures in the UK and the US ranges from about 400- 800 per 100,000 women but is relatively stable over several decades of older life (9). Women are far more likely to suffer a Colle's fracture than a man (i.e. a ratio upwards of 10:1 by age 75) (9). Compression fractures of the vertebrae are much more difficult to estimate because often these can be asymptomatic. Best estimates are that more than a million American postmenopausal women will suffer a spine fracture in the course of a single year (9-11). The female to male ratio of occurrence is approximately 2:1. Moreover, both symptomatic, and radiographic (morphometric) fractures are associated with significant morbidity and disability (10). Finally, estimates about disease prevalence in women and men without fractures, but with low BMD (-2.5 or lower) vary greatly, but place the overall number at close to 25 million Americans and many more world-wide (9,12).

 

Bone loss as a result of aging/and or estrogen deficiency is the predominant pathophysiologic disorder of primary osteoporosis. However, the frequent use of glucocorticoids in both men and women, contribute dramatically to the total number of individuals with very low BMD and/or osteoporotic fractures (11). It is estimated that more than 5 million American men are afflicted with osteoporosis, based on either the presence of osteoporotic fractures (i.e. vertebral compressions, wrist fractures, hip fractures, or humerus/tibial fractures) or low BMD (11). However, the number of cohort studies in men with this disease is somewhat limited, and a more complete epidemiologic picture of male osteoporosis is becoming clearer with the MrOS study. Importantly, and somewhat surprisingly, in this cohort of more than 5000 men over the age of 65, obesity was associated with a significantly increased risk of fracture (11-13).

 

More frightening than estimates of the extent of low bone mass is the concern about the lower frequency of diagnosing osteoporosis and the poor adherence to therapy. It is now estimated that more than 70% of individuals who are at risk for osteoporosis, and who are receiving therapy, will not continue beyond the first year (14). Moreover, prescribing rates for bisphosphonates have fallen significantly due to the perceived risk of atypical femoral fractures (15,16). Whereas, osteoporosis was once considered a disorder of old Northern European women, it is now clear that this disease can occur in postmenopausal African Americans, that it is much more common in men than previously appreciated, and that the use of glucocorticoids and/or immunosuppressive therapies for transplant patients, markedly enhances that risk. Interestingly, and somewhat alarmingly although the prevalence of hip fractures has increased slightly, (possibly due to poor compliance from the perceived adverse events with anti-osteoporosis therapy) the diagnosis of osteoporosis by primary physicians has dropped.

 

FACTORS THAT AFFECT BONE QUANTITY AND QUALITY

 

Several risk factors predispose individuals to osteoporotic fractures. For a hip fracture, these include age greater than 65 years, a previous spine or hip fracture, maternal history of a hip fracture, poor neuromuscular function, weight loss after the age of 50, and low body mass index (1,18) (See Table 1). Falls are a major cause of fractures, and in all clinical situations, some degree of trauma can be linked to the injury (19). But many osteoporotic patients suffer fractures with minimal trauma, and this feature is pathognomonic of the skeletal fragility which accompanies low bone mass. It is for this reason that the most significant risk factor for fractures of the spine, hip or wrist remains low BMD (20).  This association has been confirmed by use of the FRAX score (www.shef.ac.uk/frax) that includes areal BMD plus clinical risk factors including family history, BMI, smoking and use of glucocorticoids (21). The continuous but inverse relationship between BMD and fracture is consistent at all points below the mean suggesting there is no threshold effect (18,19). And, it is applicable at virtually every skeletal site by multiple types of measurements from the spine to finger to the calcaneus. Moreover, the advent of newer technology to measure bone mass has allowed widespread screening for risk as well as defining risk reduction with therapy. Notwithstanding the strength of the inverse relationship of BMD to fracture risk, it is important to note that the presence of a previous fracture is also an extremely important, BMD-independent risk for subsequent fracture. This is relevant clinically since the recognition of fracture at any BMD defines a skeleton that has poor bone quality, and hence is likely to fracture again.

 

Table 1. Phenotypic Characteristics that Adversely Affect Bone Strength

Characteristic

 

Recognized Risk Factor for Fracture

 

Clinically Measurable

 

Bone Mineral Density (areal)

 

Yes

 

Yes (DXA)

 

Bone Mineral Density (volumetric)

 

Yes

 

Yes (QCT)

 

Microcracks

 

No

 

No (Histology)

 

Trabecular Connectivity

 

Yes

 

TBS (DXA)

 

Periosteal Circumference

 

No

 

Yes (pQCT/DXA)

 

Cortical Thickness

 

+/-

 

Yes (pQCT/DXA)

 

Bone Turnover

 

Yes

 

Yes (markers)

 

Previous Osteoporotic Fracture

 

Yes

 

Yes (radiograms or IVA)

 

Mineralization and cortical porosity

 

?

 

No/yes (back scatter EM) and sometimes with high resolution microCT

 

See references 22-26  for more discussion about bone quality and its relationship to fracture risk. uCT- micro CT, pQCT- peripheral quantitative CT, DXA- dual energy X-ray absorptiometry, TBS-trabecular bone score.

IVA- instant vertebral assessment- DXA scan for morphometric changes in the thoracic and lumbar spine.

 

Bone mass measurement defines mineral content per area of bone. In the laboratory, bone density by DEXA is a very strong predictor of bone strength and accounts for about 80% of the variability in the breaking strength of a single femur. Thus, a very low BMD can be linked to increased skeletal fragility with a great degree of confidence.  Indeed, the FRAX tool has been independently validated as the most accurate tool to measure fracture risk and includes bone mineral density as a major component (21). But there are other determinants of bone strength, often referred to as qualitative measures, including the rate of bone turnover, the extent of trabecular connectivity, cortical and periosteal bone size, and skeletal morphometry (See Figure 1 and Table 1). Indeed, much progress has been made in quantifying several aspects of bone 'quality' utilizing tools such as single energy QCT of the spine/hip, ‘extreme’ CT (i.e. high resolution hr pQCT) of the radius or tibia (See Figure 1), TBS (i.e. trabecular bone score), histomorphometry and magnetic resonance imaging of the radius (22-26). However, more work still needs to be done to ascertain their role in clinical medicine. Still, BMD represents the most accurate, cost effective, and easiest parameter for risk assessment (21). In part, two-dimensional DEXA measurements integrate actual bone mineral content in both trabecular and cortical compartments with bone size. Due to the strong association between bone mineral density and future fractures, this phenotype remains an excellent surrogate for defining both the genetic and acquired components of the disease process. TBS, (trabecular bone score) in the vertebrae, which can be measured from DXA, provides a relatively rapid way of assessing bone quality, although its role in providing additional risk assessment remains to be determined (24).

 

Figure 1. This is a high resolution QCT image of the distal radius of an individual patient. One can image the trabecular bone of the peripheral skeleton and define measures of bone “quality” by specific measurements. It is still not clear whether these measurements provide a better insight into fracture risk than DXA.

 

BONE REMODELING AND ITS RELATIONSHIP TO BONE QUANTITY

 

Adult bone mineral density represents the end result of two processes; acquisition of peak bone mass during adolescence and maintenance of bone mass during the middle and later years. Changes in bone mass result from physiologic and pathophysiologic processes in the bone remodeling cycle (25). This can occur during the stage of accelerated linear growth in adolescence, or much later in life, usually after menopause in women. The bone remodeling cycle is a tightly coupled process whereby bone is resorbed at approximately the same rate as new bone is formed. Basic multicellular units (BMUs) compose the remodeling unit of bone and include: osteoclasts which stimulate bone resorption, osteoblasts, which are responsible for new bone formation, and osteocytes, older osteoblasts surrounded by bone and present in a reduced state of activity (26) (See Figure 2). Activation of the remodeling cycle serves two functions in the adult skeleton: 1) to produce a supply rapidly, as well as chronically, of calcium to the extracellular space; 2) to provide elasticity and strength to the skeleton. When the remodeling process is uncoupled so that resorption exceeds formation, bone is lost. On the other hand, during peak bone acquisition, formation exceeds resorption resulting in a net gain of bone. Remodeling is more pronounced in the trabecular skeleton (e.g. spine, calcaneus and proximal femur) due to much greater surface area, and is the most metabolically active component of bone, in part because of its proximity to the marrow space. However, trabecular bone is also extremely vulnerable to perturbations by local or systemic factors that can cause significant imbalances in bone turnover.

 

Figure 2. The bone remodeling cycle. The osteoblast (OB) orchestrates the orderly process of bone remodeling through activation signals from systemic factors including growth hormone (GH), interleukins (IL-1,IL-6), parathyroid hormone (PTH), and withdrawal of estrogen (-E2). M-CSF and RANKL are the two major OB mediated factors which regulate the recruitment and differentiation of the osteoclast (OC). Osteoprotogerin (OPG) is also synthesized by OBs and serves as a soluble decoy receptor blocking activation of RANK. Inhibition or knockout of these signals from OB-OC results in reduction in bone resorption. Other cells including activated T lymphocytes may contribute to the marrow milieu. Not pictured are IGFs which are released during bone resorption and serve as coupling factors to recruit new OBs to the surface. These peptides may also be important for osteoclast activity. Also sclerostin, a peptide produced by osteocytes reduces new bone formation by blocking Wnt signaling.

The initiation of the bone remodeling cycle remains unclear. The long-standing dogma was that activation of resting osteoblasts on the surface of bone and marrow stromal cells began the process (27-33). This would mean that there were initiating signals, either paracrine or endocrine that would stimulate osteogenesis. One possibility is that osteocytes, which can sense fluid shifts and microcracks, and are imbedded deep within the matrix of the skeleton, induce the remodeling sequence, by paracrine signaling to the osteoblast (27-29). But those same cells secrete factors that can initiate osteoclast differentiation as well as alter osteoblast mediated bone formation. Osteocytes, for those reasons, are now considered the ‘command and control’ system for remodeling. More recently, investigators believe that RANKL, released from osteocytes as well as stromal cells, drives osteoclast differentiation, beginning the process of active resorption prior to osteoblast differentiation. This can occur as a result of hormonal signals or mechanical loading. Resting lining cells or osteoblast progenitors as well as mesenchymal stromal cells can become activated at the same time when osteoclast differentiation is ongoing. Those progenitors also signal to osteoclast precursors to induce further differentiation and induction of hematopoietic cells (29).  Osteoclasts, once differentiated, may also elaborate growth factors, such as the Wnts and sphingosine to signal back to osteoblast progenitors.  After osteoclast- induced bone resorption, matrix components such as TGF-beta and IGF-I, as well as collagen, osteocalcin, and other protein and mineral components, are released into the micro environment of the niche. Growth factors released by resorption contribute to the recruitment of new osteoblasts to the bone surface, which begin the process of collagen synthesis and mineralization. But, in addition stromal factors such as the Wnts can induce further osteoblast differentiation via the LRP5/6 signaling pathway. In healthy adults as many as two million remodeling sites may be active at any given time, and it is estimated that nearly one fourth of all trabecular bone is remodeled each year. In general resorption takes only 10-13 days, while formation is much more deliberate and can take upwards of three months (Figure 2). Under ideal circumstances, by the end of the cycle, the amount of bone resorbed equals the amount reformed. Cessation of bone formation almost certainly occurs via osteocyte mediated sclerostin (see below) which blocks further Wnt signaling. In sum, remodeling begins at the surface of the trabecular and cortical bone via signals from the osteocytes, probably starting with osteoclast differentiation and then signaling backwards to osteoblasts, and vice versa.

 

In contrast to normal remodeling, osteoporosis has been classically defined in a pathogenic manner as an uncoupling in which resorption exceeds formation resulting in a net loss of bone. However, it is also apparent that some individuals have impaired peak bone acquisition. This scenario may be more common than previously appreciated and almost certainly represents inherited or acquired alterations in the rate of either bone formation or bone resorption during a critical period when several hormones in synchrony orchestrate a marked increase in bone mass (see later).

 

There are several key components of the remodeling cycle which are susceptible to systemic and local alterations and when perturbed, can lead to deleterious changes in bone mass. In particular, activation of remodeling via the osteoblast, and recruitment of osteoclasts, represent the two most vulnerable sites in the cycle. A third cell, altered by disease states is the osteocyte, an entombed fully differentiated osteoblast that connects to the surface osteoblasts, and likely senses mechanical stimulation. Osteocyte apoptosis may contribute to age-related osteoporosis either directly or through the elaboration of systemic peptides. Interestingly, remodeling may end with the osteocyte as well, since it produces a protein, sclerostin, which inhibits osteoblast activity by antagonizing the Wnt and BMP pathways (see below) (34).  Monoclonal antibodies that bind to sclerostin have been developed and one (e.g. romosozumab) has been approved for the treatment of postmenopausal osteoporosis (35-36). That monoclonal antibody enhanced bone formation, increased bone mineral density by 13-15% at one year, suppressed bone resorption (via Wnt mediated RANKL), and reduced overall fractures of the spine (32). Finally, one could consider macrophages to have an important role in remodeling, since these cells are present in the bone marrow niche and respond to injury with inflammatory cytokines and immune modulators (37).  On the other hand, the bone marrow niche may be protected against macrophage induced cytokine release due to a protective ‘canopy’ of lining cells, although recent evidence suggests that there is a pro-inflammatory response in the marrow with certain perturbations such as diet induced obesity or aging (29).

 

Uncoupled remodeling occurs during menopause, with estrogen deprivation or antagonists, or in response to endogenous parathyroid hormone fluxes, cytokine stimulation, growth hormone surges, glucocorticoid excess, or changes in serum calcium. For the most part, estrogen deprivation remains one of the most common and critical elements in shifting resorption rates to a higher set point (38-42). Although bone formation initially can "catch up", the length of time for each component of the remodeling cycle clearly favor resorption over formation as the process of laying down new bone requires the interaction of several processes (see Figure 2). But, it is still unclear why falling estrogen levels, which is a universal event during the menopausal years, causes such rapid bone loss in a relatively small percentage of women (43). Clearly, factors such as peripheral conversion of testosterone to estradiol, adrenal androgen production, FSH levels, and genetic determinants, as well as other local signals may also be important. In regards to FSH, SWAN (Studies of Women Across the Ages) reported that higher FSH precedes estrogen loss and this is associated with longitudinal bone loss more closely than circulating estrogen.  Although not identified in humans, mice have strong heritable determinants that affect the rate of age-related bone loss and in one mouse model high FSH levels drove significant bone loss independent of estrogen.

 

The nature of the osteocyte-osteoblast-osteoclast interaction has been one of the most active areas of recent investigation (see Figure 2). External signals (such as PTH, growth hormone, interleukin-1, estrogen deprivation) to resting osteoblasts and stromal cells cause these cells to release a potpourri of cytokines (i.e. interleukins such as IL-1, -6, -11 as well as m-CSF, tumor necrosis factor (TNF), and TGF-beta) that enhance the recruitment and differentiative function of multinucleated giant cells destined to become bone resorbing cells (38). However, one of the most critical pathways in the osteoblast-osteoclast interaction scheme is the RANKL- Osteoprotogerin (OPG) relationship. OPG is a soluble peptide originally described as a factor which markedly inhibited bone resorption and osteoclast differentiation in vitro (42). This protein is a member of the TNF receptor super-family and its role in bone remodeling is to act as a decoy receptor for the peptide known as osteoprotegerin ligand i.e. OPGL (or RANKL) (42). In fact, RANKL is a surface peptide which when expressed on the osteoblast, binds to the true OPGL receptor (also called RANK-receptor activator of NFkB Ligand) on osteoclasts, and initiates cell-cell contact necessary for osteoclast activation and subsequent bone resorption (42).  More recently RANKL has been shown to be produced by osteocytes and can result in osteocytic osteolysis during states of high calcium demand such as lactation, estrogen deficiency, and even acute exercise (43).

 

The OPG, OPGL and RANK system that affects osteoclast differentiation, in addition to the effects of m-CSF on osteoclast proliferation, provides the critical link among osteocytes, osteoclasts and osteoblasts. It has also led to the synthesis of RANKL antibodies. Denosumab, (brand name Prolia) was the first approved monoclonal antibody against RANKL for the treatment of postmenopausal osteoporosis due to its strong efficacy in reducing spine and hip fractures (44-46). It is administered once every six months and it suppresses bone resorption by 80-90%.  Unlike the bisphosphonates, denosumab’s anti-resorptive effect wanes in 4-6 months, thereby providing a margin of safety in terms of total suppression in remodeling (46). On the other hand, because the anti-resorptive effect wanes rapidly, there is concern about post treatment rebound and fractures, particularly of the vertebral spine (47).

 

The osteoblast functions not only to signal osteoclasts during remodeling as well as receive signals from them, but also to lay down collagen and orchestrate mineralization of previously resorbed lacunae in the skeletal matrix. These complex functions are tied to differentiation of mesenchymal stromal cells which become terminally differentiated osteoblasts and rest on the surface of the remodeling space (29). Recruitment of stromal cells or lining cells into osteoblasts, rather than adipocytes is a critical step in bone formation and requires a series of transcription factors that enhance differentiation. This is particularly important since one of the features of estrogen deficiency and age-related osteoporosis is the development of increased bone marrow adiposity (48,49). Indeed, the presence of excess bone marrow fat may be a major risk factor for osteoporotic fractures (50).

 

One of the most important components driving osteogenesis is Runx2, a unique transcription factor which is essential in the early differentiation pathway of osteoprogenitors (51). Regulation of Runx2 has become a major focus of work in as investigators have begun to consider novel ways to enhance bone formation and reduce marrow adipogenesis (50). Also, it should be noted that there are metabolic programs activated by transcription factors that are essential for fueling the work of the osteoblast. Conditions of substrate insufficiency; e.g. anorexia nervosa, diabetes mellitus etc. also impair bone formation by altering their metabolic programs.

 

With activation of resting osteoblasts and lining cells, osteoblasts synthesize several types of collagen as well as elaborating a series of growth factors such as IGF-I, IGF-II, TGF-b. These, in turn, are necessary for further recruitment of bone forming cells (52). In addition, osteoblasts deposit growth factors in the skeletal matrix where they are stored in latent forms, and released during subsequent remodeling cycles. After deposition of new bone, some osteoblasts are encased by matrix. These osteocytes, are still viable, although less metabolic and, can, through newly developed caniculi, provide signals to other bone cells. Indeed, most evidence suggests that osteocytic signals are important in the so-called "mechanostat", the gravitary sensing device which modulates bone formation, as well as initiating normal remodeling sequences.  As noted earlier, osteocytes may participate in bone resorption of the cortex by secreting RANKL. This is termed ‘osteocytic osteolysis’ and may occur during lactation and other states of high calcium demand.

 

In the last half-decade, the Wnt/b catenin signaling pathway has emerged as a major regulator of bone formation and a potential mediator of the mechanostat.  Wnts belong to a large family of peptides that bind to two membrane bound receptors, Lrp5 or 6 and Frizzled (see Figure 3) (53). Once these ligands bind, activation occurs through a complex intracellular signaling pathway mediated by /b catenin which translocates to the nucleus and stimulates the transcription of several genes through the TCF/Lef network.  Sclerostin (SOST), the osteocyte specific protein that inhibits bone formation does so by binding to Lrp5 and blocking Wnt signaling (54). The anti-sclerostin antibody, romosozumab, was shown to markedly increase bone mass and reduce fractures in postmenopausal women after only 1 year of treatment (55,56). It was approved by the FDA and European Regulatory Agencies for the treatment of postmenopausal osteoporosis.

 

Figure 3. The Wnt/-catenin signaling pathway that is critical for osteoblast differentiation (53).

 

In summary, the bone remodeling cycle is complex and redundant. The three major cells, osteocytes, osteoblasts and osteoclasts, arise from different stem cells (mesenchymal for osteoblasts and osteocytes, and hematopoietic for osteoclasts) and are under the control of various factors which in harmony orchestrate an orderly remodeling sequence. Their birth and death (i.e. the cycle of recruitment, proliferation and programmed cell death) and the regulatory factors which control those events, are also complex, yet vitally important for understanding the pathogenesis of osteoporosis. Alterations at any stage along the process of recruitment, activation, differentiation, or cell death can lead to imbalances in remodeling which eventually would result in bone loss, reduced bone mass and ultimately fractures. Some of those perturbations are noted below.

 

SYSTEMIC AND LOCAL ALTERATIONS IN THE BONE REMODELING SEQUENCE LEAD TO BONE LOSS

 

The importance of estrogen in maintaining calcium homeostasis for the postmenopausal woman was first established by Fuller Albright, more than 70 years ago (57). Since that time more evidence has accumulated from randomized intervention trials demonstrating that hormone replacement (estrogen with or without progesterone) reduces bone turnover and increases bone mass (58). However, these data provide only indirect evidence that estrogen levels are important as pathogenic components of the osteoporosis syndrome. More recent studies provide stronger evidence of the association between low estradiol concentrations and low bone mass. Several investigators have demonstrated that the lowest estradiol levels in postmenopausal women (i.e. <5 pg/ml) are associated with the lowest bone mineral density and the greatest likelihood of fracture (59). In addition, at least one study has shown that males with osteoporosis have lower serum levels of estradiol then do age-matched men who do not have low bone mass (60). Moreover, there are now two case reports describing mutations in either aromatase activity or the estrogen receptor, which produced a phenotype of severe osteoporosis in men (61,62). In the former case, estrogen replacement therapy for this young man, resulted in a marked increase in spine and hip bone mineral density. In both situations the lack of functional estrogen, despite normal to high levels of testosterone, resulted in very low BMD (63).

 

Although declining estradiol levels contribute to the osteoporosis syndrome, the precise molecular events or sequences that result from changes in ambient hormonal concentrations are not clear. In some animal models, estrogen deprivation is associated with a marked increase in IL-6 synthesis from stromal and osteoblastic cells. This is consistent with experimental findings which demonstrate that estrogen regulates the transcriptional activity of the IL-6 promoter (39,41). However, results in other studies are conflicting. In other experimental paradigms, changes in TNF, IL-11 and IL-1 can all be associated with increased bone resorption (40). Recently RANKL has been identified as a major regulator of osteoclast differentiation. Thus, it seems likely that several cytokines, working in concert, are active during estrogen deprivation, and each can accelerate the process of bone resorption. RANKL, however may be the most critical and necessary for full activation of remodeling. Enhanced bone resorption eventually leads to bone loss from estrogen deprivation since bone formation rates cannot keep up with rates of bone resorption (31,42, 43). Moreover, the faster the resorption, the greater the loss resulting in more damage to the structure integrity of the skeleton.

 

In contrast to the plethora of studies on bone loss and estrogen, there are few good studies relating androgen deprivation to bone loss in women. Androgen receptors are present on osteoblasts. However, both in vitro and in vivo studies in men have yielded conflicting results. Like estrogen, androgens can regulate the IL-6 promoter and in experimental animals, orchiectomy has been associated with increased IL-6 production and bone loss (64). In men, chronically low androgen levels have been associated with low bone mass, and testosterone replacement can enhance bone mineral density (65). However, estradiol levels in men may be a more important risk factor for fracture than androgen levels and correlate more closely with trabecular bone volume as measured by QCT. At the present time it is not clear precisely what role androgens play in the maintenance of bone mass in both men and women except in deficiency syndromes, particularly hypogonadism.

 

Other more common causes of male osteoporosis include alcoholism, glucocorticoid excess, and hypercalciuria (see Table 2). In the first two cases, hypogonadism remains a pathogenic feature of that osteoporosis syndrome. Less frequently, men with gluten enteropathy or an endocrinopathy such as thyrotoxicosis or primary hyperparathyroidism can present with multiple fractures and low bone mass (see Table 2).

 

Table 2. Etiology of Osteoporosis in Men

Etiology

 

Age-years

 

Clinical Features

 

Hypogonadism

 

30-80

 

low Test, low E2, increased resorption

 

Alcoholism

 

40-80

 

low test, E2+/-, +/- turnover

 

Glucocorticoids

 

20-80

 

 +/- test, E2 +/-, increased resorption, Decreased formation

 

 Multiple myeloma

 

 40-80

 

 Hypercalcemia, ESR

Hypercalcuria

30-80

Test, E2 nl; increased resorption Hypercalcuria, inc PTH, kidney stones

Idiopathic Osteoporosis-

 

40-80

 

fractures, low formation, low IGF-I

 

Gluten enteropathy

 

20-80

 

low 25OHD, turnover increased

 

Endocrine Disorders

 

15-90

 

Cushing’s ±

PHPT

 

15-90

PTH increased in all cases; increased resorption

 

Thyrotoxicosis

15-90

Decreased PTH

E2- estradiol; Test-testosterone; PTH-parathyroid hormone; PHPT-primary hyperparathyroidism; Inc- increased

 

In women, bone loss is accelerated immediately after menopause. However, recent studies demonstrate that markers of bone resorption are also very high later in life. In particular, women in their 80s and 90s have been noted to lose bone at a rate of greater than 1% per year from the spine and hip (65), Contrary to earlier studies, it is now evident that the older woman who is not as physically active, and is not on estrogen, is at extremely high risk of bone loss and subsequent fractures. The pathogenesis of this process is multifactorial although dietary calcium deficiency, leading to secondary hyperparathyroidism, certainly plays a central role. The average calcium intake of women in their 8th and 9th decades of life is now estimated to be between 800-1000 mg/day (66). If vitamin D intake is also sub-optimal, and serum levels of 25 OH vitamin D <20 ng/ml, or 50nmol/l, secondary hyperparathyroidism may occur, although there are other causes for increases in PTH, including chronic renal insufficiency and low calcium diet (67). PTH stimulates osteoblasts and provokes the remodeling sequence including the elaboration of several cytokines that accelerate bone resorption. Unfortunately, in most elders, bone formation is not enhanced although the reasons for this are not entirely clear beyond the simplistic notion that stem cell recruitment is impaired in the elderly. Nonetheless, this leads to further uncoupling in the bone remodeling cycle, and significant bone loss. Among elders with poor calcium intake who live in northern latitudes, seasonal changes in vitamin D lowering levels below 20 ng/ml might aggravate bone loss (68,69). Whether increased bone loss from vitamin D deficiency is an independent risk factor for future fractures in the elderly remains somewhat controversial necessitating further studies to define such a risk. Overall, there is evidence that vitamin D deficiency is associated with a greater risk of fracture in frail elderly institutionalized men and women (<20 ng/ml; 50 nmol/l), whereas it is unlikely that those individuals with 25 OHD levels between 20-30 ng/ml are at a greater risk (70).  The recently completed VITAL trial demonstrated that 2000 IU of vitamin D to healthy men and women did not prevent bone loss or alter bone turnover (LeBoff, personal communication and abstract ASBMR,2019).

 

There is growing evidence that low serum levels of vitamin D, through impaired calcium absorption, can stimulate PTH release and increase bone turnover in the elderly. Increased PTH enhances 1,25 dihydroxyvitamin D, and this in turn could suppress further bone formation and mineralization (71). Thus, bone loss is associated with uncoupled remodeling. Many older individuals already have established osteoporosis. Coincidental vitamin D deficiency due to poor intake, absent sunlight exposure, or impaired conversion of vitamin D to its active metabolite, can result in osteomalacia as well as aggravating pre-existent osteoporosis (70).  LeBoff et al reported that more than 50% of elders who presented with a hip fracture were frankly vitamin D deficient (71). Therefore, the combination of vitamin D deficiency with inadequate calcium intake enhances the likelihood of rapid bone loss in the very susceptible elderly population. Still, it is unclear how secondary hyperparathyroidism causes bone loss. Chronic elevations in PTH secretion due to primary or tertiary hyperparathyroidism, have been associated with low bone mass at several skeletal sites including the radius. Elevated PTH levels in older women have been associated with bone loss in some studies but not in others. In elderly individuals, it has been reported that PTH levels are closely correlated with increased synthesis of an insulin-like growth factor binding protein (IGFBP-4) which suppresses IGF action on bone cells and may increase sclerostin secretion (72, 34). Since IGF-I is an important growth factor for osteoblasts, it is conceivable that PTH down regulates IGF activity during states of relative calcium/and or vitamin D deficiency. This would shift the remodeling balance towards preserving intravascular calcium concentrations, while inhibiting new calcium incorporation into the skeletal matrix. In sum, there is little doubt that calcium and vitamin D insufficiency are prominent causes of accelerated bone loss in the elderly (73). However, in the healthy postmenopausal population, there is little evidence that vitamin D supplementation prevents bone loss or fractures.

 

As noted previously, high circulating levels of glucocorticoids have a significant impact on bone acquisition and maintenance. In 1932 Harvey Cushing recognized the syndrome of endogenous steroid excess which included marked osteopenia and fractures (74). Long term exposure to pharmacologic doses of glucocorticoids results in significant bone loss and enhanced marrow adipogenesis as marrow stromal cells differentiate down the fat lineage. In addition to having direct effects on the osteoclast and osteoblast, glucocorticoids also induce secondary hypogonadism and hyperparathyroidism, impaired vitamin D metabolism, muscle atrophy, and hypercalciuria (See Table 3). All these factors contribute to a rapid and sustained loss of bone during the first few months of steroid therapy (75). The addition of other immunosuppressants such as cyclosporine has been shown to aggravate bone loss by further increasing bone resorption. As the number of organ transplants have increased exponentially over the last decade, the prevalence of post-transplantation osteoporosis has risen substantially. Steroid induced osteoporosis is now considered the second most common cause of low bone mass in the general population and one of the most common causes of osteoporotic fractures (75). It is listed in the FRAX data set as one critical risk factor to assess in determining 10-year fracture likelihood.

 

Table 3. Effects of Glucocorticoids on Bone Mass

 Response to Glucocorticoids

 

Effects on Bone Remodeling

 

Effects on Bone Mass

 

Increased PTH secretion

 

Increased bone resorption

? decreased bone formation

 

rapid loss of bone

 

Decreased LH/FSH secretion

 

 

Increased bone resorption due loss of estrogen

 

 

loss of bone

 

Impaired calcium absorption due to decreased 1,25 D

 

Increased PTH, increased bone resorption

 

loss of bone

 

Increased calcium loss in urine

 

Secondary increase in PTH, Increased bone resorption

 

 

loss of bone

 

Acute suppression of osteoblasts and apoptosis

 

Reduced bone formation

 

gradual bone loss

 

Stimulation of osteoclastogenesis

 

Increased bone resorption rapid

 

loss of bone

 

 

PATHOGENIC FACTORS WHICH IMPAIR PEAK BONE MASS

 

Peak bone mass is acquired between the ages of 10-16 years. It is the zenith of bone acquisition and represents the sum of several processes including a marked increase in bone formation (76,77). Boys tend to reach peak 2 years later than girls and their bone mineral density is higher than women at all skeletal sites. In part this relates to a greater cross-sectional bone area in males than females (78). Peak bone mass results from linear growth and consolidation of cortical and trabecular components. Acquisition is most rapid during the latter stages of puberty and coincides with maximum growth hormone secretion, high serum IGF-I levels, and rising levels of estradiol and testosterone. In addition, calcium absorption is maximal and skeletal accretion is optimal. All these processes combine over a relatively short period of time to produce a bone mass that subsequently plateaus and then falls during later life. It is estimated that more than 60% of adult bone mass can be related to peak acquisition. Hence understanding the mechanisms responsible for low bone mass must include perturbations in peak bone acquisition.

 

There are several hormonal, environmental, nutritional and heritable determinants of peak bone mass. These include estrogen/testosterone, growth hormone/IGF-I, adequate nutrition, calcium/vitamin D, and unknown genetic factors. If each is perturbed, dramatic alterations in peak bone mass may occur, setting the stage for low bone density throughout life. Gonadal steroids are important not only to bone maintenance but also to its acquisition. During puberty, estrogen and testosterone levels rise and contribute to consolidation of bone mass. Estrogen is also necessary for epiphysial closure. Studies of a male with an estrogen receptor mutation and men with an aromatase deficiency established that estradiol is critical for bone acquisition (61,62). These young men share several phenotypic characteristics including tall stature unfused epiphysis, and very low bone mass. Hence, there must be a threshold effect for estradiol in men, and this effect must be time dependent. Similar conclusions can be drawn from studies in women. Acquired deficiencies in estrogen, such as anorexia nervosa, or chemotherapy induced ovarian dysfunction, result in low peak bone mass and lead to subsequent risk for osteoporosis (79,80,81). Nearly identical findings have been noted in patients with untreated Turner's syndrome and in men with Klinefelter's syndrome, although the high FSH levels could also contribute to bone loss.

 

The timing of gonadal steroid surges is critical for bone acquisition since there is a relatively short window of time in which bone formation is favored and matrix synthesis is markedly enhanced. That window is likely to be less than three years and earlier in girls than boys. Probably the best study which addressed this issue comes from a retrospective analysis of men in their thirties who underwent late onset of puberty (i.e. at the age of 17 or 18) but were otherwise normal by full endocrine testing. These men had significantly lower bone mineral density in their thirties than age matched men who went through puberty at a normal time. These data suggest that timing as well as quantity of gonadal steroids is critical for bone acquisition.

 

Pubertal surges of estrogen and androgens are also important for priming the growth hormone/ IGF-I axis. Rising levels of both contribute to growth hormone surges that lead to increases in circulating and tissue expression of IGF-I, an essential growth factor for chondrocyte hypertrophy and expansion. IGF-I may also be critical in defining the cross-sectional size of bone, a potentially important determinant of bone strength. Once again, studies in growth hormone deficient, or growth hormone resistant individuals have established that low levels of circulating IGF-I, especially during puberty, are associated with reduced bone mass (82). In addition, rhGH replacement has been shown to restore linear growth and improve peak bone mass acquisition. Several studies in experimental animals, including inbred strains of mice, have established that IGF-I is important for bone acquisition and the timing of IGF-I peaks coincide with maximal rates of bone formation. Impairment in production of IGF-I due to acquired disorders such as anorexia nervosa, malnutrition, and delayed puberty can also impede peak bone acquisition (80). Recently, it has become apparent that Type I IDDM can impact the bone marrow niche and suppress bone formation and increase resorption. This uncoupling can lead to an impairment in peak bone mass, although there are other determinants of glucose intolerance that can impact the skeleton; e.g. increased advanced glycation end products, glucose toxicity, material property changes in the matrix.

 

Hormonal abnormalities not only enhance bone resorption in older individuals, but may blunt the capacity of bone cells to maximize bone formation during adolescence. Clearly, hypogonadal boys and girls have impaired peak bone mass, resulting in low adult bone mineral density. Even one form of contraception, Depo-provera, may reduce estrogen concentrations enough in the teen girl, to reduce her capacity to acquire peak bone mass. Similarly, it seems likely although not proven that smoking during the teen years could impair osteoblast activity and flatten projected trajectories for peak bone acquisition.

 

More recently diabetes mellitus has been established as a secondary cause of osteoporosis. TIDM, particularly at early onset (e.g. childhood or adolescence) is associated with low bone mass and fractures (83).  Both hip and vertebral fractures are more common in adult T1DM, whereas in T2D, in which areal BMD is normal, fracture risk is increased by 20%, but the type of fractures are usually peripheral in nature (84). Importantly use of rosiglitazone, a TZD is also associated with an increased risk of fracture whereas DPP4 inhibitors and GLP1 agonists are bone neutral or favor a slight increase in bone mass.  The newer SGLT-2 inhibitors are associated with a greater risk of fracture as a class. Despite the greater risk with glucose intolerance, the pathophysiology of these disorders is not well defined, although changes in advanced glycation end products, enhanced reactive oxygen species, and glucose toxicity are all considered likely contributors.

 

In order to mineralize newly synthesized bone, calcium must become bioavailable to the skeletal matrix. In experimental studies in rodents and humans, it is clear that the several pools of available calcium are markedly enhanced during puberty. These include calcium efflux from the gastrointestinal tract, and the calcium pool available for incorporation in the matrix. It is no coincidence that growth hormone surges not only increases IGF-I, (thereby enhancing skeletal growth and matrix biosynthesis) but also result in increases in 1,25 dihydroxyvitamin D (possibly via IGF-I induction of 1, alpha hydroxylase activity), the active metabolite of vitamin D which markedly enhances calcium absorption from the gut. Although there are no longitudinal studies in pubertal individuals with prolonged calcium deficiency, several randomized placebo-controlled trials in pubertal and pre-pubertal girls and boys have established that supplemental calcium can enhance bone mineral density. In a twin study, in which one twin receives 1200 mg of calcium supplementation, and one receives placebo, radial BMD increases by as much as 5% after three years compared to placebo (81). This study suggests that there is significant gene-environmental interaction, and that even in those individuals with heritable determinants of low peak bone mass, calcium supplementation may provide an important and relatively simple means of protecting individuals from future osteoporotic fractures.

 

GENETIC DETERMINANTS OF PEAK BONE MASS

 

Probably the most important determinant of peak bone mass, but one that has lacked clear definition is the genetic contribution. In part this is due to the complex nature of bone mineral density as a trait. Nevertheless, low peak bone mass may be the most important pathogenic factor in the osteoporosis syndrome of later life. And, it appears that at least 50% of peak bone mass is determined by genetic factors (85). What are these determinants and how are they modified by environmental factors?

 

Efforts to define heritable determinants of peak bone mass have been complicated by a number of issues which are also common to analyses of other complex traits. These include the following:

  1. A quantifiable phenotype;
  2. Heterogeneity within a given population under study;
  3. The polygenic nature of the disorder;
  4. Epistasis (gene-gene interaction)
  5. Pleiotropy- phenotypic differences with identical genotypes, and
  6. Gene by environmental interactions.

 

Notwithstanding these complexities it is clear that BMD is an acceptable phenotype for defining heritable determinants of future risk. Bone mineral density in the population is distributed in a gaussian manner, thereby allowing analyses at the extremes (<-2.0 SD or > 2.0 SD) of the density distribution. Large homogeneous and heterogeneous populations are now being studied as part of the GEFOS consortium, to ascertain genetic determinants of BMD and fractures in humans (86-91). Candidate genes are those associated previously by biologic determinants or previous studies as being important for skeletal maintenance.  Indeed, many of these have been identified by whole genome studies (GWAS) include RANKL, OPG, the vitamin D receptor, collagen IA1, the estrogen receptor, interleukin-1, and IGF-I. Additionally several other candidate genes that have not been previously associated with osteoporosis have been identified. These studies have been reviewed in depth elsewhere (86-89).  Depending on the cohort, the phenotype, and the number of individuals studied, it is predicted there will be hundreds of genes that contribute to individual variation in bone mass (90,91). Twin studies examining discordant or concordant phenotypes are also helpful, as are sib-pair studies, although the results have been less generalizable.

 

One example of a candidate gene that has also been shown to have strong heritability and predictive value for osteoporosis from GWAS studies is Lrp5 (92). Originally Recker and colleagues identified an extended family with very high bone density and fine mapped the locus to a region in chromosome 11. After several years of intense high through put analysis, that group identified a 'high bone density' gene, LDL receptor related protein 5 (LRP-5) that was mutated in this family (92). The low affinity lipoprotein receptor is important in binding Wnts, ligands critical for cell differentiation in several organisms. At the same time, Warman and colleagues identified several children with osteoporosis pseudoganglioma syndrome and subsequently mapped the gene which resulted in 'loss of function' in these individuals (93). This turned out to also be Lrp5. The potential pathways that direct osteoblast function and mineralization through Lrp5, and its co-receptor frizzled, have opened up new areas of investigation (See Figure 3 and discussion above). Moreover, natural antagonists to the Wnt/Lrp5 signaling system including sclerostin and DKK-1 have been studied using genetic engineering in mice. This pathway continues to show great promise for therapeutic interventions such as romosozumab, recently approved by the FDA for the treatment of postmenopausal osteoporosis (54, 99).

 

In the past five years, Lrp5 has been studied extensively both in its function and its allelic effects through genome wide association studies as well as in translational bench work. Hence this pathway is important in regulating peak bone mass. But it is also clear that since BMD is a polygenic trait, other genes are now being discovered. Moreover, gene environmental interactions must play a major role in defining heritable risk for low BMD and fracture. In addition to the search for osteoporosis genes, intervention studies in adolescents have provided insight into at least one of the environmental impacts on genetic determinants (94-97). A twin study in Indiana revealed that as long as calcium supplementation continued during puberty, young boys could enhance their peak bone mass. In a Swiss study, younger pre-pubertal girls supplemented with a protein product had a significant increase in spine bone density, as did a cohort of pubertal girls receiving a milk powder in England (96,97). Remarkably, in the latter cohort, serum IGF-I levels also rose dramatically, providing further indirect evidence of a link between pubertal status, bone mass, and the growth hormone/IGF-I axis. Thus, there is strong evidence that nutritional, hormonal and environmental factors play a major role in regulating peak bone mass.

 

SUMMARY

 

The epidemiology of osteoporosis is well established and risk factors have been defined. On the other hand, the pathogenesis of osteoporosis is complex and multifactorial (41,98). Alterations in bone mineral density almost certainly represent the final common pathway by which pathologic factors affect risk of future osteoporotic fracture. The interplay of various physiologic processes which result in peak bone mass, and maintenance of adult bone mass are key to understanding the pathogenesis of this disease. Changes in hormonal status, and in particular estradiol, clearly are important factors in regulating both bone formation and bone resorption in men and women. Perturbations in growth hormone activity, musculoskeletal function, dietary intake of calcium and vitamin D, and genetic determinants are also important pathogenic factors. Defining the role of genetic factors and their interaction with many of the environmental and hormonal determinants that have been established as potential etiologic agents responsible for low bone mineral density and fracture will certainly be the most difficult challenge facing basic and clinical researchers. On, the other hand, the strength of data from basic and clinical studies over the last decade, now allows practitioners to confidently diagnose osteoporosis. New treatment strategies offer greater hope for patients suffering from this disease.

 

REFERENCES

 

  1. Cummings SR, Nevitt MC, Browner WS, Stone K, Fox KM, Ensrud, KE, Cauley J, Black D, Vogt TH. Risk factors for hip fracture in white women. N Engl J Med 1995;332:767-773.
  2. Kanis JA, Melton LJ, Christiansen C, Johnston CC, Khaltaev N. The diagnosis of osteoporosis. J Bone Min Res 1994;9:1137-1141.
  3. Melton LJ, Chrischilles EA, Cooper C. Perspective: How many women have osteoporosis? J Bone Min Res 1992; 7: 1005-1010.
  4. Seeley DG, Browner WS, Nevitt MC. Which fractures are associated with low appendicular bone mass in elderly women? The Study of Osteoporotic Fractures. Ann Intern Med 1991; 115:837-842.
  5. Kanis JA. Assessment of fracture risk and its application to screening for postmenopausal osteoporosis: synopsis of a WHO report. Osteoporosis Int 1994; 4: 368-371.
  6. Eliffors I, Allander E, Kanis JAS. The variable incidence of hip fracture in southern Europe: The Medos Study. Osteoporosis Int; 1994: 4: 253-261.
  7. Cooper C, Campion G, Melton LJ. Hip fractures in the elderly: a world wide projection. Osteoporosis Int 1992; 2: 285-289.
  8. European Commission: Report on Osteoporosis in the European Community-Action for Prevention. Brussels; European Commission 1998.
  9. DeLaet CEDH, Pols HAP. Fractures in the elderly: epidemiology and demography. Bailliere's Clin Endocrinol Metab 2000; 14: 171-179.
  10. Ross PD. Clinical Consequences of vertebral fractures. Am J Med 1997; 103: 425-435.
  11. Cosman F, Krege JH, Looker AC, Schousboe JT, Fan B, Sarafrazi Isfahani N, Shepherd JA, Krohn KD, Steiger P, Wilson KE, Genant HK. Spine fracture prevalence in a nationally representative sample of US women and men aged ≥40 years: results from the National Health and Nutrition Examination Survey (NHANES) 2013-2014 Osteoporosis Int. 2017; 28(6):1857-1866.
  12. O Neill TW, Felsenberg D, Varlow J. The prevalence of vertebral deformity in European men and women: the European Vertebral Osteoporosis Study. J Bone Min Res 1996; 11: 1010-1018.
  13. Cauley JA, Cawthon PM, Peters KE, Cummings SR, Ensrud KE, Bauer DC, Taylor BC, Shikany JM, Hoffman AR, Lane NE, Kado DM, Stefanick ML, Orwoll ES; Risk Factors for Hip Fracturein Older Men: The Osteoporotic Fractures in Men Study (MrOS). Osteoporotic Fractures in Men (MrOS) Study Research Group. J Bone Miner Res. 2016 Oct;31(10):1810-1819. doi: 10.1002/jbmr.2836.
  14. Lindsay BR, Olufade T, Bauer J, Babrowicz J, Hahn R. Patient-reported barriers to osteoporosistherapy. Arch Osteoporosis. 2016;11:19. doi: 10.1007/s11657-016-0272-5.
  15. Khan M, Cheung AM, Khan AA. Drug-Related Adverse Events of Osteoporosis Therapy. Endocrinol Metab Clin North Am. 2017 Mar;46(1):181-192
  16. Adler RA. Bisphosphonates and atypical femoral fractures. Curr Opin Endocrinol Diabetes Obes. 2016 Dec;23(6):430-434.
  17. Whittier X, Saag KG. Glucocorticoid-inducedOsteoporosis. Rheum Dis Clin North Am. 2016 Feb;42(1):177-89
  18. Marshall D, Johnell O, and Wedel H. Meta-analysis of how well measures on bone mineral density predict occurrence of osteoporotic fractures. Br Med J 1996; 312: 1254-1259.
  19. Stewart A, Walker LG, Potter RW, Primrose WR, Reid DM. Prediction of a second hip fracture: the potential role of DXA, ultrasound, and other risk factors for targeting of preventative therapy. J Clin Densitometry 1999;2(4):363-370.
  20. Tinetti ME, Speechely M, Gunter SF. Risk factors for falls among elderly persons living in the community. N Engl J Med 1988;319:1701-1707

21     Miller PD. Guidelines for the clinical utilization of bone mass measurements in the adult population. Calcif Tiss Int 1995;57:252-252.

  1. Marques A, Ferreira RJ, Santos E, Loza E, Carmona L, da Silva JA. . The accuracy of osteoporotic fracturerisk prediction tools: a systematic review and meta-analysis. Ann Rheum Dis. 2015 Nov;74(11):1958-67.
  2. Riggs BL, Wahner HW, Seeman E. Changes in bone mineral density of the proximal femur and spine with ang: differences between the postmenopausal and senile osteoporosis syndromes. J Clin Invest 1982;70:716-723.
  3. Boutroy S, Bouxsein ML, Munoz F, Delmas PD. In vivo assessment of trabecular bone architecture by high resolution peripheral quantitative CT. J Clin Endocrinol Metab 2005 90:6508-6515.
  4. Martineau P, Leslie WD. Trabecular bone score (TBS): Method and applications. Bone. 2017 Nov;104:66-72.
  5. 26. Manhard MK, Nyman JS, Does MD. Advances in imaging approaches to fracture risk evaluation. Transl Res. 2017 Mar;181:1-14.
  6. Lian JB and Stein GS. The Cells of Bone in Principles of Bone and Cartilage Metabolism. Ed by Seibel MJ, Robbins S and Bilezikian JP. Academic Press, San Diego 1999 pp 165-185.
  7. Bonewald LF. The Role of the Osteocyte in Bone and Nonbone Disease. Endocrinol Metab Clin North Am. 2017 Mar;46(1):1-18.
  8. Reagan MR, Rosen CJ. Navigating the bone marrow niche: translational insights and cancer-driven dysfunction.Nat Rev Rheumatol. 2016 Mar;12(3):154-68.
  9. Lorenzo JA and Raisz LG. Cytokines and Prostaglandins in Principles of Bone and Cartilage Metabolism Ed by Seibel MJ, Robbins S and Bilezikian JP. Academic Press San Diego CA 2005 pp 97-109.
  10. Udagawa N, Takahashi N, Jimi E, Matsuzaki K, Tsurukai T, Itoh K, Nakagawa N et al Osteoblasts/Stromal Cells Stimulate Osteoclast Differentiation factor/RANKL but not Macrophage Colony Stimulating Factor. Bone 1999; 25:517-523.
  11. Emery JG, McDonnell P, Burke MB, Deen KC, Lyn S, Silverman C, Dul E, Appelbaum ER, et al Osteoprotegrin is a receptor for the cytotoxic ligand TRAIL J Biol Chem 1998;273:14363-14367.
  12. Suda T, Takahashi N and Martin TJ. Modulation of osteoclast differentiation. Endocrine Rev 1992;12:66-80.
  13. Ellies DL, Viviano B, McCarthy J, Rey JP, Itasaki N, Saunders S. Bone density ligand, sclerostin, directly interacts with LRP5 but not LRP5G171V to modulate Wnt activity. J Bone Miner Res 2006; 21:1738-1749.
  14. Xiadong L, Ominsky MS, Warmington KS, Marony S, Gong J, Cao J, Yongming G, Shalhoub V, Tipton B, Haldankar R, Chen Q, Winters A, Boone T, Zhaopo G, Qing-Tian N, Hua Zhu K, Kosteniuk PT, Scott Simont W, Lacey DL, Paszty C. Sclerostin antibody treatment increases bone formation, bone mass, and bone strength in a rat model of postmenopausal osteoporosis. J Bone Miner Res 2009;24:578-588.

36     Cosman F, Crittenden DB, Adachi JD, Binkley N, Czerwinski E, Ferrari S, Hofbauer LC, Lau E, Lewiecki EM, Miyauchi A, Zerbini CA, Milmont CE, Chen L, Maddox J, Meisner PD, Libanati C, Grauer A. Romosozumab treatment in postmenopausal women with osteoporosis. N Engl J Med. 2016 Oct 20;375(16):1532-1543.

  1. Michalski MN, McCauley LK. Macrophages and skeletal Health. Pharmacol Ther. 2017 Jun;174:43-54.
  2. Eghbali-Fatourechi G, Khosla S, Sanyal A, Boyle WJ, Lacey DL, Riggs BL. Role of RANK ligand in mediating increased bone resorption in early postmenopausal women. J Clin Invest 2003;111(8):1221-1230.
  3. Jilka RG, Girsole GH, Passeri G, Williams D, Abrams J, Boyce B, Broxmeyer H and Manoloagas S. Increased osteoclast development after estrogen loss: mediation by IL-6. Science 1992;257:88-91.
  4. Pacifici R, Brown C, Puscheck E. The effect of surgical menopause and estrogen replacement on cytokine release from human blood monocytes. Proc Natl Acad Sci USA 1991;88:5134-5138.
  5. Manolagas SC, Jilka RL Emerging insights into the pathophysiology of osteoporosis. N Engl J Med 1995;332:305-311
  6. Fujiwara Y, Piemontese M, Liu Y, Thostenson JD, Xiong J, O'Brien CA. RANKL(Receptor activator of NFκB ligand) produced by osteocytes Is required for the Increase in B cells and bone loss caused by estrogen deficiency in mice. J Biol Chem. 2016 Nov 25;291(48):24838-24850.
  7. Xiong J, Piemontese M, Onal M, Campbell J, Goellner JJ, Dusevich V, Bonewald L, Manolagas SC, O'Brien CA Osteocytes, not osteoblasts or lining cells, are the main source of the RANKLrequired for osteoclast formation in remodeling bone. PLoS One. 2015 Sep 22;10(9):e0138189.
  8. Zebaze R, Libanati C, McClung MR, Zanchetta JR, Kendler DL, Høiseth A, Wang A, Ghasem-Zadeh A, Seeman E. Denosumabreduces cortical porosity of the proximal femoral shaft in postmenopausal women with osteoporosis. J Bone Miner Res. 2016 Oct;31(10):1827-1834.
  9. Cummings, SR, San Martin J, McClung MR, Siris ES, Eastell R, Reid IR, Delmas P, Zoog HB, Austin M, Wong A, Kutilek S, Adami S, Zanchetti J, Libanati C, Siddhanti S, Christiansen C: FREEDOM Trial. Denosumab for prevention of fractures in postmenopausal women with osteoporosis. N Engl J Med 209 Aug 20:361(8),756-765.
  10. Ottawa (ON): Denosumab (Xgeva) [Internet]. Canadian Agency for Drugs and Technologies in Health; 2016 Nov.
  11. Lamy O, Gonzalez-Rodriguez E, Stoll D, Hans D, Aubry-Rozier B. Severe rebound-associated vertebral fracturesafter denosumab discontinuation: nine clinical cases report. J Clin Endocrinol Metab. 2017 Feb 1:102(2):354-358.
  12. Sheu Y, Amati F, Schwartz AV, Danielson ME, Li X, Boudreau R, Cauley JA; Osteoporotic Fracturesin Men (MrOS) Research Group. Vertebral bone marrow fat, bone mineral density and diabetes: The Osteoporotic Fractures in Men (MrOS) study. Bone. 2017 Feb 4;97:299-305
  13. Greco EA, Lenzi A, Migliaccio S The obesity of bone. Ther Adv Endocrinol Metab. 2015 Dec;6(6):273-86.
  14. Veldhuis-Vlug AG, Rosen CJ. Mechanisms of marrow adiposity and its implications for skeletal health. Metabolism. 2017;67:106-114.                                               
  15. Ducy P, Zhang RR, Geoffroy V, Ridall AL, and Karsenty G. Osf2/Cbfa1: a transcriptional activator of osteoblast differentiation. Cell 1997; 89:747-754.                            
  16. Rosen CJ. Growth hormone, IGF-I and the elderly: Clues to potential therapeutic intervention. Endocrine.1997 Aug;7(1):39-40.                                                   
  17. Baron R, Rawadi G. Targeting the Wnt/beta-catenin pathway to regulate bone formation in the adult skeleton. Endocrinology 2007Jun;148(6):2635-2643.                           
  18. Poole, KE, van Bezoolijen RL, Loveridge N, Hamersma H, Papapoulos SE, Lowik CW, Reeve J. Sclerostin is a delayed secreted product of osteocytes that inhibits bone formation. FASEB J 2005 Nov;19(13):1842-1844.
  19. Appelman-Dijkstra NM, Papapoulos SE. Sclerostin inhibition in the management of osteoporosis. Calcif Tissue Int. 2016Apr;98(4):370-380.
  20. Bandeira L, Lewiecki EM, Bilezikian JP. Romosozumabfor the treatment of osteoporosis. Expert Opin Biol Ther. 2017 Feb;17(2):255-263.
  21. Albright F. Postmenopausal osteoporosis. JAMA 1941; 116 2465-2474.
  22. Chestnut C,Notelovitz M, Clark G, Drinkwater B, Rosen C, Bell N, English S, Johnston CC, Cain D, Flessland K, Mallinak N. Use of the N-telopeptide of type I collagen to monitor the effect of therapy and predict changes in bone mineral density in postmenopausal women treated with hormone replacement therapy. Am J Med;1997:102:29-37.
  23. Ettinger B, Pressman A, Sklarin P, Bauer D Cualey JA and Cummings SR. Associations between low levels of serum estradiol, bone density and fractures among elderly women: SOF. J Clin Endocrinol Metab 1998;83:2239-2243.
  24. Greendale GA, Edelstein S Barrett-Connor E. Endogenous sex steroids and bone mineral density in older women and men. J Bone Miner Res 1997;12:1833-1837.
  25. Smith EP, Boyd J, Frank GR, Takahasi H, Cohen RM, Specker B, Williams TC, Lubahn DB, Korach KS. Estrogen resistance caused by a mutation in the estrogen receptor gene in a man. N Engl J Med 1994;331:1056-1061.
  26. Morishima A, Grumbach MM, Simpson ER, Fisher C, Qin K. Aromatase deficiency in male and female siblings caused by a novel mutation in the physiological role of estrogens. J Clin Endocrinol Metab 1995;3689-3698.
  27. Carani C, Qin K, simoni M, Faustini-Faustini S, Boyd J, Korach KS, Simpson ER. Effect of testosterone and estradiol in a man with aromatase deficiency. N Engl J Med 1997; 337:91-95.
  28. Snyder PJ, Peachey H, Hannoush P, Berlin JA, Loh L, Lenrow DA, Holmes JH, Diewati A, Santana J, Rosen CJ and Strom BL. Effect of testosterone treatment on body composition and muscle strength in men over 65.J Clin Endo Metab 1999;84:2647-2653.
  29. Dresner-Pollak R, Parker RA, Poku M, Thompson J Seibel MJ, Greenspan SL. Biochemical markers of bone turnover reflect femoral bone loss in elderly women. Calcif Tissue Int 1996:59:328-333.
  30. Food and Nutrition Board Institute of Medicine 2011; Dietary Reference Intakes for Calcium, Phosphorus, Magnesium Vitamin D and Fluoride. Washington DC, National Academy Press. www.iom/vitaminD.edu
  31. Storm D, Smith-Porter E, Musgrave KO, Vereault D, Patton C, Kessenich CR, Eslin R, Mohan S, Chen T, Holick MF and Rosen CJ. Calcium supplementation prevents seasonal bone loss and changes in biochemical markers of bone turnover in elderly New England women: A Randomized Placebo-Controlled Trial. J Clin Endocrinol Metab 1998;83;3817-3826
  32. Rosen CJ Morrison A, Zhou H, Storm D, Hunter S, Musgrave KO, Chen T, et al. Elderly women in northern new England exhibit seasonal changes in bone mineral density and calciotropic hormones. Bone Miner 1994;25:83-92.
  33. Ross AC, Manson JE, Abrams SA, Aloia JF, Brannon PM, Clinton SK, Durazo-Arvizu RA, Gallagher JC, Gallo RL, Jones G, Kovacs CS, Mayne ST, Rosen CJ, Shapses SA. The 2011 report on dietary reference intakes for calcium and vitamin D from the Institute of Medicine: what clinicians need to know. J Clin Endocrinol Metab. 2011 Jan;96(1):53-58.
  34. Kahwati LC, Weber RP, Pan H, Gourlay M, LeBlanc E, Coker-Schwimmer M, Viswanathan M Vitamin D, Calcium, or Combined Supplementation for the Primary Prevention of Fractures in Community-Dwelling Adults: Evidence Report and Systematic Review for the US Preventive Services Task Force. JAMA. 2018 Apr 17;319(15):1600-1612.
  35. LeBoff MS, Kohlmeier L, Hurwitz S, Franklin J, Wright J Glowacki J. Occult vitamin D deficiency in postmenopausal US women with acute hip fracture. JAMA 1999; 282:1505-1511.
  36. Rosen C, Donahue LR, Hunter S, Holick M, Kavookjian H, Kirshenbaum A, Mohan S and Baylink DJ. The 24/25kD serum insulin-like growth factor binding protein is increased in elderly women with fractures. J Clin Endocrinol Metab. 1992;74:24-28.
  37. Heaney RP, McCarron DA, Dawson Hughes B, Oparil S, Berga SL, Stern JS, Barr SI, Rosen CJ. Dietary Changes favorably affect bone remodeling in older adults. JADA:99:1228-1233.
  38. Cushing H. Basophile adenomas of the pituitary body and their clinical manifestations. Bull Johns Hopkins Hop 1932;50:137-145.
  39. Thacker H. Glucocorticoid-induced Osteoporosis. Cleveland Clinic J Med 2010 77 843-844.
  40. Teegarden D, Proulx WR, martin BR. Peak bone mass in young women. J Bone Miner Res 1995;10:711-715.
  41. Prior JC, Vigna Y, Schechter MT, Burgess AE. Spinal bone loss and ovulatory disturbances. N Engl J Med 1990;323:1221-1227.
  42. Weaver CM, Gordon CM, Janz KF, Kalkwarf HJ, Lappe JM, Lewis R, O'Karma M, Wallace TC, Zemel BS. The National Osteoporosis Foundation's position statement on peak bone mass development and lifestyle factors: a systematic review and implementation recommendations. Osteoporosis Int. 2016 Apr;27(4):1281-386.
  43. Gilsanz V, Loro ML, Roe TF, Syrre J, Gilsanz R and Schulz EE. Gender differences in vertebral size in adults: biomechanical implications. J Clin Invest 1995;95:2332-2337.
  44. Bachrach LK, Guido D, Katzman D, Litt IF, Marcus R. Decreased bone density in adolescent girls with anorexia nervosa. Pediatrics 1990;86:440-447.
  45. Johnston CC, Miller JZ, Slemenda CW, Reister TK, Hui S, Christian JC, Peacock M. Calcium supplementation and increases in bone mineral density in children. N Engl J Med 1992; 327 82-87.
  46. Menaa C, Vrtovsnik F, Freidlander G, Corvol M and Garbedian M. IGF-I a unique calcium dependent stimulator of 1,25 vitamin D production. J Biol Chem 1995;270:25461-25467.
  47. Schacter GI, Leslie WD Diabetes and BoneDisease. Endocrinol Metab Clin North Am. 2017 Mar;46(1):63-85
  48. Shanbhogue VV, Mitchell DM, Rosen CJ, Bouxsein ML. Type 2 diabetesand the skeleton: new insights into sweet bones. Lancet Diabetes Endocrinol. 2016 Feb;4(2):159-73. 
  49. McKay HA, Bailey DA, Wilkinson AA, Houston CS. Familial comparison of bone mineral density at the proximal femur and lumbar spine. Bone Miner 1994;24:95-107.
  50. Smith DM, Nance WE, Kang KW, Christian JC, Johnston CC. Genetic factors in determining bone mass. J Clin Invest 1973; 52:2800-2808.
  51. Seeman E. Genetic Determinants of the Population Variance in Bone Mineral Density. In The Aging Skeleton eds by CJ Rosen, J Glowacki and JP Bilezikian 1999; Academic Press San Diego, CA pp77-84.
  52. Rogers J, Mahaney MC, Beamer WG, Donahue LR and Rosen CJ. Beyond one gene-one disease: Alternative strategies for deciphering genetic determinants of osteoporosis Calcif Tiss Int 1997;60:225-228.
  53. Utterlinden AG, Burger H, Huang Q, Fang Y, MCGuigan FEA, Grant SFA, Hofman A et al. Relation of alleles of the collagen type I alpha 1 gene to bone density and the risk of osteoporotic fractures in postmenopausal women. N Engl J Med 1998;338:1016-1021.
  54. Hsu YH, Kiel DP. Clinical review: Genome-wide association studies of skeletal phenotypes: what we have learned and where we are headed. J Clin Endocrinol Metab. 2012 Oct;97(10):
  55. Richards JB, Zheng HF, Spector TD. Genetics of osteoporosis from genome-wide association studies: advances and challenges. Nat Rev Genet. 2012 Jul 18;13(8):576-88.
  56. Boyden LM, Mao J, Belsky J, et al. High bone density due to mutation in LDL-receptor related protein-5. New Engl J Med 2002 May:346(20):1513-1521.
  57. Gong Y, Vikkula M, Boon L, Liu J, Beighton P, Ramesar R, Peltonen L, Somer H, Hirose T, Dallapiccola B, De Pappe A, Swoboda W, Zabel B, Superti-Furga A, Steinmann B, Brunner HG, Jans A, Boles RG, Adkins W, van den Boogaard MJ, Olsen BR, Warman ML Osteoporosis-pseudogliomasyndrome, a disorder affecting skeletal strength and vision, is assigned to chromosome region 11q12-13. Am J Hum Genet. 1996 Jul;59(1):146-51
  58. Bonjour JP, Carrie AL, Ferrari S. Calcium enriched foods and bone mass growth in prepubertal girls: a randomized double blind placebo controlled trial. J Clin Invest 1997 99:1287-1294.
  59. CadoganJ, Blumensen A, Barker M, Eastell R. A longitudinal study of bone gain in pubertal girls: anthropometric and biochemical correlates. J Bone Min Res 1998;13:1602-1612.
  60. Weaver CM, Peacock M and Johnston CC. Adolescent nutrition in the prevention of postmenopausal osteoporosis. J Clin Endocrinol Metab;1999: 84:1839-1843.
  61. Bonjour JP, Chevalley T. Pubertal timing, boneacquisition, and risk of fracture throughout life. Endocr Rev. 2014 Oct;35(5):820-847.
  62. Black DM and Rosen CJ. Postmenopausal Osteoporosis. 2016 NEJM 374: 257-262.

99     Shoback D, Rosen CJ, Black DM, Cheung AM, Murad MH, Eastell R. Pharmacological Management of Osteoporosis in Postmenopausal Women: An Endocrine Society Guideline Update. J Clin Endocrinol Metab. 2020 Mar;105(3):587-594.

Gastrinoma

ABSTRACT

 

Gastrinomas are neuroendocrine neoplasms (NENs), that occur primarily in the duodenum and pancreas, which ectopically secrete gastrin, resulting in the Zollinger-Ellison syndrome (ZES), which is due to marked hypersecretion of gastric acid causing severe gastro-esophageal peptic disease. ZES patients have two management problems that must be dealt with: control of the acid hypersecretion and control of the gastrinoma, which is malignant in 60-90% of cases. Most gastrinomas are sporadic, but 20-25% of patients have it as part of the Multiple Endocrine Neoplasia-type 1 syndrome (MEN1), an autosomal dominant disorder characterized by endocrine tumors/hyperplasia of multiple endocrine organs (parathyroid> pancreatic islets>pituitary>adrenal). It is important to identify those with ZES/MEN1 as their management differs from those with sporadic disease. Acid hypersecretion is now controlled medically both acutely and long term, with proton pump inhibitors (PPI) the drugs of choice. In patients with sporadic ZES, after detailed imaging with cross-sectional imaging and somatostatin receptor imaging (SRI), resection of the gastrinomas should be considered whenever possible, with cures reported in 20-45% of patients. The role of surgical resection of the gastrinomas in MEN1/ZES is controversial and it is generally recommended it be reserved for patients with tumors>1.5/2 cm because of the multiplicity of small gastrinomas resulting in very low cure rates. The diagnosis of ZES requires demonstrating fasting hypergastrinemia in the presence of inappropriate acid secretion (pH<2), however, because of the widespread use of PPIs and the lack of gastric acid testing, the diagnosis of ZES is becoming more difficult and referral to a specialty group is frequently required. Patients with advanced metastatic disease are treated as other patients with advanced NENs including with somatostatin analogues, chemotherapy, everolimus, sunitinib, liver directed therapies, and peptide radio-receptor therapy (PRRT) with radiolabeled somatostatin analogues.

 

GENERAL/DEFINITIONS

 

ZES was first described in 1955 by two surgeons, RM Zollinger and EH Ellison, in two patients with intractable peptic ulcer disease (1). Although previous cases had been described (2,3), including one well described case by Roar Strom in 1952 (3-5), in Zollinger/Ellison’s two patients the authors were the first to propose the important association between the gastric hypersecretion and the presence of a pancreatic neuroendocrine neoplasm (PNEN) (1-3,6). Presently, the term gastrinoma and ZES are often used synonymous, however, in the past the term gastrinoma was also used to refer to a neoplasm synthesizing gastrin and ZES to the clinical manifestations (7).  Numerous NENs and non-NENs can synthesize gastrin precursors which are not processed to the biologically active gastrin-17 or gastrin-34 as in ZES, and thus are generally not called gastrinomas by clinicians or in most current classification systems of pNEN (7-9). In addition to being well-described in humans, Zollinger-Ellison syndrome due to a gastrinoma have also been reported in dogs (10-17), cats (10,18-22), and a Mexican gray wolf (23).

 

Like most other functional pNEN syndromes (F-pNEN) (insulinomas, glucagonomas, VIPomas, etc.), in ZES the functional syndrome due to the ectopic hormone secretion requires immediate treatment because it was the most frequent cause of morbidity/death prior to effective treatments (24-38). In addition, treatment must be directed at the gastrinomas itself, because similar to all other pNEN, except insulinomas, the majority (60-90%) are malignant (9,26,27,39-42). Whereas effective surgical resection would cure both problems, in <50% of ZES patients is curative resection possible because of advanced disease or the patients have MEN1/ZES, which can only be cured with Whipple resections, which are not generally recommended (discussed below) (6,9,43-49). Therefore, treatment of patients with ZES requires management of two different treatment problems: the acid hypersecretion and the malignant nature of the gastrinoma.

 

This chapter will review important aspects of the management of patients with ZES and important treatment issues at present, including the most recent studies up to 2023. It will concentrate on the most current important aspects and not cover comprehensively all areas of ZES or numerous areas in depth. For more in depth considerations the reader is referred to recent papers/reviews which cover ZES generally (6,33,38,40,46,49-54); its diagnosis (29,51,55-61), clinical features (24,25,41,62-69); acid hypersecretion (24,50,70-72); gastrin provocative testing and the diagnosis of hypergastrinemia (36,50,51,55,57,73-86); MEN1/ZES (30,44,47,57,64,85-96,96-102); medical treatment of acid hypersecretion (50,51,69,72,78,80,103-108);clinical course and prognosis (41,65,87,93,109-117);  surgical treatment of the gastrinoma (6,44,50-52,80,92,95,96,99,100,102,103,118-128); imaging and tumor localization (37,50,90,112,124,125,129-141);  treatment of advanced disease in ZES and other NENs (42,48,50,51,58,135,142-156); diagnosis and treatment of all/functional pNEN (24-26,34-36,36,48,48-50,54,58,148,156-165) and pathology, pathogenesis  and classification of gastrinomas/NENs (9,50,58,86,96,117,158,166-174).

 

Before considering the diagnosis and management of ZES in more detail it is important to realize that there are a number of misconceptions about ZES, often because of comparison with other pNEN and these need to be kept in mind. They are listed in the Table 1 below and briefly discussed in the following sections.

 

Table 1.   Widely Held Misconceptions About ZES

1) Gastrinomas, similar to a number of other pNEN (insulinomas, gastrinomas, PPomas), primarily occur in the pancreas.  FACT: In recent studies, 60-100% of gastrinomas in both sporadic ZES and MEN1/ZES occur in the duodenum, with only 0-15% in the pancreas (6,43,50,95,102,127,175-179) (Table 2).

2) MEN1 is uncommon in ZES, similar to other pNEN such as insulinomas (3-5%), glucagonomas (<5%), PPomas/nonfunctional pNEN (<3%). FACT: MEN1 is found in the highest frequency of all pNEN syndromes in ZES patients occurring in 20-25% and is important to diagnose because of its different treatment aspects (30,50,64,72,87,89,95,102).

3) With the increased awareness of ZES and widespread availability of gastrin assays and sensitive imaging modalities, similar to some other pNEN, gastrinomas are being diagnosed earlier. FACT: The time of onset of symptoms to diagnosis of ZES remains 4-7 years (24,26,48,60,62,89,134) and a number of factors are contributing to make the diagnosis even more difficult (See point #4 below).

4) As recommended in all guidelines (9,72,80,152,157,180-182), similar to other functional pNEN syndromes (F-pNENs), ZES is currently diagnosed by demonstrating excess hormone production (fasting hypergastrinemia) in the presence of an unphysiological effect of the hormone hypersecretion (i.e., inappropriate acid hypersecretion (elevated basal acid output>15 mEq/hr., pH<2)) (9,50,51,55,56,59,70,72,73,79,181,183,184). FACT: In contrast to, for example, insulinomas, which are uniformly diagnosed by demonstrating fasting hyperinsulinemia with accompanying hypoglycemia (frequently during a fasting study) (29,50,185-188), in a recent review of the last 20 cases of ZES reported in the literature in 2018 (55), 95% of the diagnoses were reported without performing a gastric analysis or gastric pH assessment (55) and thus not using classical established criteria. This approach has complicated the diagnosis of ZES and the factors leading to this confusion will be discussed below in detail in the ZES diagnosis section.

5) In MEN1 patients, similar to other MEN1 patients with F-pNEN such as insulinomas and glucagonomas, most gastrinomas can be cured by nonaggressive surgical resections in MEN1/ZES patients. FACT:  In contrast to other F-pNEN (29,157,189), the 5-year surgical cure rate of MEN1/ZES is <5% (6,30,43,44,88,190) without aggressive surgical resections such as Whipple resection, which are not recommended (6,9,88,92,93,118,123,157,180,182). However, without these resections, most patients with small tumors and adequate acid secretory control have an excellent prognosis, which has led to controversy in their treatment, and will be discussed in the surgical section later (30,43,47,92,93,95,102,118,157,180,182,191).

 

The misconceptions listed in Table 1 above as well as the factors specific to ZES that led to these misconceptions have led to controversies that are complicating numerous aspects of the management of ZES patients. These extend particularly to the current diagnosis of ZES, the management of both gastrinomas and nonfunctional pNENs in MEN1/ZES patients, and various aspects of the surgical management of these patients. Each of these will be discussed in more detail in the specific later sections in this chapter.

 

EPIDEMIOLOGY: ZES

 

PNEN account in different series for 1-10% of all pancreatic tumors with a prevalence of 1/100,000 and annual incidence of 1-4/million, which is increasing in frequency (192-194).  In older series, insulinomas, gastrinomas, and NF-pNEN were reported with similar frequencies, however, in recent series of pNEN patients NF-pNEN make up 60-80% of all cases (24,48). Currently, for F-pNENs, insulinomas and gastrinomas are the most frequent, with incidences of 0.5-3/million in different series (26,50). Generally, insulinomas/gastrinomas are 8-10-fold more frequent than VIPomas, 17-fold more than glucagonomas, and >20 fold more the other F-pNENs (GRFomas, pancreatic ACTHomas, etc.) (26,50). Gastrinomas are the most frequent malignant F-pNEN, because 60-90% are malignant, like the other less common F-PNEN, in contrast to insulinomas, which are malignant in only 5-10% in most series (26,50,158,188).

 

Gastrinoma, as well as other pNENs, can occur both sporadically or as part of an inherited syndrome (30,158,195-197). Gastrinomas occur more frequently with an associated inherited pNEN syndrome than other F-pNEN, particularly in the case of MEN1, where 20-25% of all ZES patients have MEN1/ZES, compared to <3-5% of other F-pNEN syndromes (30,50,65,86,87,89). ZES is also rarely reported in other inherited syndromes associated with pNEN including the autosomal dominant syndromes, von Hippel –Lindau Disease (30,196,198,199), tuberous sclerosis (30,200), and neurofibromatosis type 1 and type 2 (30,199,201-204).

 

PATHOPHYSIOLOGY: CLINICAL FEATURES

 

In the majority of patients with ZES (>90%), the presenting symptoms are due to the marked gastric acid hypersecretion (24,28,62,64,70,205,206). Generally, only in patients with advanced disease late in the disease course are the prominent symptoms due to the tumor per se (abdominal pain, weight loss, anorexia, etc.) (24,28,40,62,205,206).  The acid dependency of the above symptoms is shown by numerous studies reporting in a typical ZES patient, all of the presenting symptoms (including the PUD, pain, diarrhea, GERD symptoms, weight loss) disappear if the gastric acid hypersecretion is adequately controlled by any means (surgical, medical, acid aspiration) (7,27,28,40,103,106,207).

 

The ectopic release of gastrin by the gastrinoma is the direct cause of the gastric hypersecretion (49,170,208). In a typical ZES patient the fasting hypergastrinemia results in a markedly increased basal acid output (BAO) of approximately 4-fold (42-mEq/hr.) (70) and in some patients the BAO is increased more than >10-fold (27,28,70,206,209-212). Chronic hypergastrinemia also has trophic effects on the gastric mucosa, stimulating an increase in number of parietal cells and gastric enterochromaffin-like cells (ECL cells) (7,76,213-217) with the result the parietal cell mass is increased up to 4-6-times normal (27,76,218,219). This contributes to both the elevated BAO and increased maximal capacity to secrete acid, as shown by ZES patients having increased maximal acid outputs (MAOs) (27,70,76,212,219-221). Diarrhea which is seen in >70% of ZES patients (Table 3) in recent prospective studies is due to the effects of the gastric acid hypersecretion by causing structural damage to the small intestine, it interferes with fat transport; inactivates pancreatic lipase; can precipitate bile acids; and if prolonged, leads to steatorrhea (27,158,222).

 

Long-standing hypergastrinemia stimulates proliferation of the gastric enterochromaffin-like cells (ECL cells), which show such a response in ZES-patients (223). Gastric ECL cells are increased a mean of twofold in ZES (76,212,223-225). ZES patients can develop advanced ECL-proliferative responses, similar to the findings in animal studies of chronic hypergastrinemia induced by various methods, and which, in some cases, results in neoplastic changes (7,76,213,217,226,227).  It has been proposed that with chronic hypergastrinemia, the ECL cells undergo a progressive hyperplasia-neoplasia sequence of events beginning with simple hyperplasia, followed by linear hyperplasia, micronodular hyperplasia, adenomatoid hyperplasia, dysplasia (pre-carcinoid) and finally the development of carcinoids (7,76,217,226,228).  In the prospective NIH studies greater than 98% of ZES patients demonstrated ECL hyperplasia (217,227), with 50% having advanced changes with sporadic ZES (7% dysplasia) (217) and 53% with MEN1/ZES (2%-dysplasia) (227). In ZES, there is a close correlation between the degree of ECL hyperplasia and the fasting serum gastrin level (76,217,227). Even though advanced ECL proliferative changes are seen in both sporadic and MEN1/ZES-patients, they have a marked difference in the rate of occurrence of gastric carcinoids. Gastric carcinoids occur in 0-33% of MEN1/ZES-patients (76,227), and in the one perspective NIH study were found in 23% (87,224,227,229-231). However, gastric carcinoids rarely occur (<1%) in sporadic-ZES patients (212,217,232-234), and it has been estimated they occur at least with 70-fold greater frequency in MEN1/ZES-patients (227). An important finding of the prospective NIH studies of ZES patients is there was no threshold effect of fasting gastrin on ECL growth, as had been previously proposed, with any increase in FSG being associated with increased ECL proliferation (76,217,227).

 

PATHOLOGY AND TUMOR CLASSFICATION

 

In the past, gastrinomas were frequently reported as nonbeta islet cell tumors (1), because they were originally thought to originate in the pancreas from the islets and to generally be pancreatic in location, similar to insulinomas (1,51,185,205,235,236). They were reported to occur in the pancreas with a distribution of pancreatic head: body: tail of 4:1:4 (27,40,63,205,237,238). Later studies described a small percentage of duodenal gastrinomas (239,240). Currently, duodenal gastrinomas are found 2-10 times more frequently than pancreatic (Table 2) (43,64,95,102,175-177,241-245). Therefore, prior to the mid-1980s, 80-95% of gastrinomas were reported in the pancreas, whereas now 45-100% are duodenal, and 0-45% pancreatic (40,175-177,236,241-244).  Even as late as 1998, in Soga’s review of 359 cases of ZES, only 11% of the patients had a duodenal gastrinoma (7,27,43,63,235).This likely occurred because of the analogies to insulinomas which are almost always in the pancreas, as well as the fact that duodenal tumors were being missed on preoperative localization studies or with a standard laparotomy because of their small size (Table 2) (27,43,175-177,241) and in many series no gastrinoma was found in a significant percentage of patients (7,27,28,40,63,235). Furthermore, a number of the early cases were patients with MEN1/ZES, and intra-pancreatic tumors were found (which were generally NF-pNEN) and these were attributed to be the source of the gastrin, with the true source being in a duodenal gastrinoma, which was not explored for or detected. Recent studies show that when careful attention is paid to the duodenum at surgery (duodenotomy, intraduodenal palpation, transillumination on occasion), more duodenal tumors were found (6,45,95,102,175,176,241-244,246-248). Primary gastrinomas are rarely found in other intra-abdominal sites: (particularly the ovary and liver/bile duct,  as well as very uncommonly in the pylorus, spleen, mesentery, stomach,  kidney)  and in a few cases(<5 total) (<0.5%) in extra-abdominal locations, including the cardiac intraventricular spectrum and due to nonsmall cell lung cancer (Table 2) (40,45,109,110,121,126,176,236,249-265).  A number of studies provide strong evidence that gastrinomas can arise in lymph nodes as the primary site, however, this is not universally accepted and some have proposed that they represent metastases from occult primaries (27,40,43,258,259,266-274). The possibility that a lymph node primary tumor may occur is supported by studies demonstrating long-term cure after resection of only a lymph node gastrinoma (40,258,259,267). Furthermore, in 3-25% of patients without pNEN, chromogranin-positive rests occur in abdominal lymph-nodes (266,275). In the NIH prospective series, 11% of patients are classified as having primary lymph node gastrinomas (Table 2).

 

At surgery, it has been recently emphasized that 60-90% of gastrinomas occur within the “gastrinoma-triangle”, which is an area formed by the junction of the cystic/common bile ducts posteriorly, the junction of the second/third parts of the duodenum inferiorly, and the junction of the pancreatic neck/body medially (40,176,177,244,276). This occurs primarily because of the high frequency of duodenal gastrinomas which are now found that fall into this area. Duodenal gastrinomas do not occur in equal proportion in all parts of the duodenum, but instead demonstrate a decreasing occurrence distally, with almost 90% of duodenal gastrinomas occurring in the 1st/2nd part of the duodenum (Table 2) (175,277,278).

 

In early studies, 60-90% of gastrinomas were associated with metastases (primarily lymph-node/liver) and therefore they should all be considered potentially malignant (9,39,205,236,257,279). The presence of metastases or gross invasion of normal tissue remains the only generally accepted criterion for the diagnosis of malignancy (27,40,280). Gastrinomas metastasize initially primarily to regional lymph nodes and the liver (27,109,236). Duodenal gastrinomas are characteristically small in size (Table 2), frequently <1 cm in diameter; however, they are associated with lymph node metastases in 47% of the cases in the NIH prospective studies (20-80%-literature), which is a similar percentage seen with the larger pancreatic gastrinomas (mean size 3.8 cm) (Table 2). From this data it has been proposed that gastrinomas in these two sites are equally malignant (109,110,175,281). However, from the NIH prospective studies it is also proposed that duodenal and pancreatic gastrinomas are not equally aggressive, because liver metastases occur in 52% of the NIH patients with a pancreatic gastrinoma (15-45%-literature) (Table 2), whereas liver metastases occur in only 5% of duodenal gastrinomas (10%-literature) (Table 2) (109,110,281). This is a similar rate to a recent collective series of 24 ZES cases with a duodenal gastrinoma in which  4 of the patients (16%) had liver metastases but 75% had lymph node metastases (282).Similarly in a recent review of 52 ZES patients(33-sporadic/19-MEN1/ZES) the rate of liver metastases was significantly lower in those with MEN1/ZES (21% vs 51%, p=0.031) (64).   At presence the basis for this difference in aggressive behavior of pancreatic and duodenal gastrinomas is unclear. A genomic analysis (172) identified a number of molecular similarities and differences between duodenal gastrinomas and pNENs. In a comparison of RNA-seq data, duodenal gastrinomas and pancreatic pNENs shared 1233 common co-expressed transcripts, however duodenal gastrinomas expressed 909 distinct transcripts not seen in either normal duodenum or pancreatic pNENs and pancreatic pNENs had 588 unique transcripts not shared in normal pancreas or duodenal gastrinomas (172).The duodenal gastrinomas  strongly expressed two inflammatory mediators (IL-17 and TGF-alpha), enrichment of mesenchymal, cytoskeletal, neuroactive-ligand receptor interaction, and calcium signaling pathway genes (64). In both duodenal and pancreatic neuroendocrine tumors alterations in expression of genes were found that were involved in cellular signaling cascades as well as in associated immune cells, and presence of proinflammatory cytokines, however it is unclear how these are related to the differences in biologic behavior of these two groups of NENs.

 

Duodenal gastrinomas in sporadic cases (75-80%) differ from those in MEN1/ZES patients in that they are usually solitary tumors, whereas in MEN1/ZES they are multicentric, smaller and multiple (64,178,283,284).

 

Duodenal gastrinomas account for 44-66% of all duodenal NENs (285,286), however only 58 % are associated with the development of ZES (286). In a recent study (286) the characteristics of sporadic duodenal gastrinomas associated with ZES (n=24) or not associated with ZES (n=17) were compared. The duodenal gastrinomas associated with ZES had a higher mean Ki-67(1.74 vs 0.85, p=0.012), more frequently had associated lymph node metastases (75 vs 6%, p=0.012), more frequently were associated with liver metastases and presented more frequently with TNM stage ≥III (75 vs 6%, p<0.0010). In a recent collective study of 108 sporadic ZES patients (127) in which 68 had duodenal gastrinomas and 19 pancreatic tumors, the overall 5-yr survival was 94% and not affected by gastrinoma location. However, pancreatic location was associated with higher recurrence rate (p=0.0001) (127).

 

In the past literature, approximately one-third of ZES patients presented with metastatic liver disease, approximately one-third with no tumor found and one-third with localized disease (Table 2) (27,40). Some recent studies suggest an increasing proportion are being seen with earlier disease stages, without advanced disease (40) (Table 2).  For example, in the last 221 patients seen at the NIH, the majority (65%) at presentation had localized disease, and in the remaining 35% of the patients, they were divided between those with hepatic metastases and those with no primary tumor found (40,109,110) (Table 2). This distribution of gastrinoma extent differs from that reported in various surgical series, because not all ZES patients are included in these series with exclusion of all non-operated patients including those with patients with diffuse liver metastases, most with MEN1/ZES and those with contra-indications to surgery (9,43,181,287,288). In the last 155 patients undergoing surgical exploration at NIH, 85% had limited disease and the remaining 15% either had limited hepatic metastases (8%) or no tumor was found (7%) (40). In older studies, up to 50% of patients had no tumor found (Table 2), whereas at present, gastrinomas are more frequently found, as evidenced by the recent NIH data in which in the last 81 patients explored for possible cure at NIH, a gastrinoma was found in all (43). As pointed out above this difference is almost entirely due to the careful exploration of the duodenal area with a Kocher maneuver, duodenotomy, intraluminal palpation, and transillumination, (43,63,175,176,244,247,289).  It is likely the detection rate of primary gastrinomas will increase even further with the recent development and widespread use of somatostatin receptor imaging (SRI), which has superior sensitivity to conventional cross-sectional imaging (129,133,134,136,137,139,153). SRI was initially performed  with 111Indium (diethylenediamine penta-acetic-D-phenylalanine-1) octreotide with single photon emission CT (SPECT) detection, but has now been replaced by 68Gallium DOTA (9,4,7,10-tetraazacyclododecane-1,4,7,10-tetracetic acid) labeled somatostatin analogues (generally 68Ga-DOTATOC PET/CT) with positron-emission tomography detection because of its even greater sensitivity (129,134-137,139,153,290-293).

 

Distant, extrahepatic, metastases can occur with advanced gastrinomas (112,294-299). Metastases to bone are reported in 31% of ZES patients with advanced disease which occur primarily in the axial skeleton initially, however, they are uncommonly seen in ZES patients that do not have liver metastases (112,294,297,300). Their identification is important, because their detection frequently alters management (109,112,294,296,298,299).

 

Histologically, gastrinomas show the typical features of NENs, with cubical cells generally with few mitoses and having a granular, eosinophilic cytoplasm (236,280). They can demonstrate trabecular, gyriform or glandular morphology; however, no specific pattern is predictive of biologic behavior (27,235,280). Duodenal gastrinomas occur in the submucosa, frequently infiltrate the mucosa and in the case of tumors >1 cm, the muscular layer (236).  Duodenal gastrinomas usually have proliferative rates <10%, whereas pancreatic gastrinomas frequently have higher proliferative rates (236,286). Both duodenal and pancreatic gastrinomas may demonstrate blood vessel invasion (236,280). Gastrinomas are usually identified as a NEN by their histological appearance and positivity with immunohistochemistry for the NEN markers (chromogranin A, synaptophysin) (27,236,280,301). Gastrin immunoreactivity (Gastrin-IR) can be detected in most gastrinomas (27,236,302,303) and approximately one-half produce multiple hormones (27,236,302,303).

 

Recently, it has been proposed that gastrinomas, as well as all pNEN/GI-NENs (carcinoid tumors), should have a common classification as NENs (166,168,304-306). Several classification systems (International Union for Cancer Control/American Joint Cancer Committee (UICC/AJCC), World Health Organization (WHO), European Neuroendocrine tumor Society (ENENs)) for both staging and grading NENs have been proposed recently, validated for pNEN, GI-NENs (carcinoids) and NENs (carcinoids) in other locations and recently updated (158,166,168,306-308). The use of these classification systems is essential to the management of NEN patients because they not only have overall prognostic significance, they also have predictive value for different treatment approaches and thus can dictate the treatment approach in some cases (115,115,116,116,158,166,167,306,308). These classification systems use primarily tumor size, extent, differentiation of the tumor and invasion for determination of stage (306). The grade of the tumor is determined by evaluating proliferative indices (Ki-67 and mitotic index (MI)) and the degree of differentiation of the tumor (well vs poor). NENs are divided into three grades based on the proliferative indices with Grade 1(G1) or low grade, having a  Ki67<3%(MI <2 mitoses/10-HPF; Grade 2(G2) or intermediate grade having a Ki67>3-20%(MI-2-20/10 HPF), and high grade or Grade 3(G3) having a Ki67>20%(MI>20 mitosis/10 HPF) (115,116,158,166,167,306-309). Recently (WHO2017, 2019) Grade 3 was divided into two different groups depending on tumor differentiation with G3NEN having well differentiated tumor cells, and G3NEC (neuroendocrine cancer) having poorly differentiated tumor cells (115,116,158,166,167,306,308,310).  Recent studies show G3NENs and G3NECs not only vary markedly in survival, but they also vary in their molecular pathogenesis and their treatment approaches (115,116,158,166,167,306,310,311). Proper classification of gastrinomas is essential, because recent studies demonstrate it has prognostic value and may affect the type of treatment recommended (115,116,304-306,312). Most gastrinomas are well-differentiated, pNEN Grade 1 or grade 2 (38,64,236,286). In one recent retrospective cohort study (64) (n=52), the grades of gastrinomas in patients with MEN1/ZES differed from those with sporadic ZES in having lower grade (G1: 83 % vs 39%; G2 (11% vs 54%) G3: (5.6% vs 6.1%), as well as being smaller in size (1.7 cm vs 3.1 cm).  A review of 171 gastrinomas in various papers published up to 10/2020 in which tumor grade was reported, shows that 74% of the gastrinomas were grade 1, 22% were Grade 2 and only 4% were Grade 3(313). There is limited data on the correlation of tumor grade in ZES patients with survival. In one study (65)on univariate analysis in MEN1/ZES and sporadic ZES patients(n=37) the presence of grade 3 gastrinomas correlated with decreased survival (p=0.008), however not on multivariate analysis. In a recent review (127) of 108 patients with sporadic ZES, no predictive factors for survival, including tumor grade, were identified, however, for recurrence post- surgical resection, only tumor size (p=0.005) and tumor grade (p=0.01) were independent predictors of tumor recurrence. Two recent analyses (115,116) of prognostic factors in patients with any pNEN demonstrate that grade of the tumor was the most frequent significant prognostic factor cited in the studies analyzed both for overall survival and for disease free survival post-surgery (116) and in treatment of advanced resistant disease (115). These data would strongly suggest that the tumor grade of the gastrinoma in ZES patients will likely be a very important prognostic factor for assessing various aspects of long-term tumor behavior (survival/recurrence/aggressive growth).

 

Table 2. Characteristics of Gastrinomas (NIH Prospective Studies and Literature)

Characteristic

NIH Data (n=221)

Mean (range) Percent

Literature

Mean (range) Percent

Primary Location

Pancreas

Duodenum

Lymph node

Other (1)

Unknown

 

24

49

11

9

16

 

42 (0 – 70%)

15 (0 – 100%)

<1%

2 (0 – 18%)

30 (7 – 48%)

Duodenal Location

D-1

D-2

D-3

D-4

 

57

32

6

3

 

ND

ND

ND

ND

Percent Extent of Disease

No tumor found

Localized disease

Metastatic disease to liver

 

13

70

17

 

30 (7 – 50%)

36 (23 – 52%)

34 (13 – 54%)

Extent Metastases

Primary only

Primary + lymph nodes

Primary + liver metastases

Liver metastases only

Lymph node metastases only

 

36

29

23

3

16

 

32 (23 – 50%)

23 (8 – 61%)

32 (15 – 40%)

10 (4 – 15%)

11 (4 – 24)

Gastrinoma Size (cm)

Mean (largest)

Duodenal

Pancreatic

 

2 ± 0.2(0.1-4.8)

0.9 ±0.1(0.1-5)

4 ± 0.3 (0.5-7)

 

(1-6)

(0.2-5.5)

(0.5-10)

Metastases: Duo vs Pancreatic

 

Lymph node Metastases (%)

Duodenal

Pancreas

 

Liver Metastases (%)

Duodenal

Pancreas

 

 

 

47

48

 

 

5

52

 

 

 

 

(20-80%)

(up to 48% of patients had no primary 0-60%)

 

10

(15-45%)

Abbreviations: Duo-duodenal; D1-4-duodenal regions, 1,2,3,4;

Data are from (2,109,110,175,177,178,243,244,246,281,314-317).

(1) Other tumor locations include additional intra-abdominal sites (liver, bile duct, spleen pylorus, mesentery, ovary, lymph nodes) and very rarely extra-abdominal sites (heart, nonsmall cell lung cancer.

 

Tumors in a given patient in multiple locations can be monoclonal or polyclonal. In MEN1, multiple gastrinomas were reported to arise by independent clonal events in one study (318). A more recent study (114) which include 137 microscopic and macroscopic duodeno-pancreatic NENs and 36 matched metastases in 10 patients with MEN1 assessed tumoral ARX, PDX1, Ki67, gastrin expression and alternative lengthening of telomere. Most metastases (91%) originated from a single NET of origin, however, a few patients had likely multiple, metastatic primary NETS.  In 6 patients with hypergastrinemia with MEN1, periduodeno-pancreatic lymph node metastases expressed gastrin and clustered with minute duodenal gastrinomas, not with larger pNEN. The pNEN frequently clustered with high grade or alternative lengthening of telomere positive primary tumors. It was concluded that in MEN-1 patients with ZES and pNEN a duodenal origin of the periduodeno-pancreatic lymph node metastases is likely even if preoperative localization studies do not reveal a duodenal tumor (114). Clonality (319) was analyzed in 20 sporadic gastrinomas from eight patients in whom the tumor was present in at least two separate sites. A combination of methods was used to assess clonality, including MEN1 gene mutation analysis, loss of heterozygosity analysis of the MEN1 locus, and analysis of X-chromosome inactivation at the human androgen receptor locus (human androgen receptor analysis). In three patients, a somatic MEN1 gene mutation was detected in the tumor. Identical mutations were found in other tumors at different sites within the same patients. Human androgen receptor analysis in three informative patients and loss of heterozygosity analysis in five patients revealed identical clonal patterns in the tumors from multiple sites in each patient. This study (319) concluded that sporadic gastrinomas at multiple sites are monoclonal and that MEN1 gene alterations in gastrinomas occur before the development of tumor metastases.

 

TUMOR BIOLOGY

 

Similar to other NENs, gastrinomas frequently synthesize (pancreatic polypeptide, insulin, glucagon, somatostatin) and also secrete multiple, gastrointestinal peptides as well as chromogranins, alpha-subunits of the glycoprotein hormones, and neuron-specific enolase (27,280,303,320-322).  In one study (303) plasma levels of hormones other than gastrin are elevated in 62% of ZES-patients, with one additional hormone elevated in 44% and two in 18%. Motilin is the most common plasma hormone also elevated (30%), followed by human pancreatic polypeptide (27%), neurotensin (20%) and gastrin-releasing peptide (10%) (303). The occurrence of a second F-pNEN syndrome does occur in ZES patients (27,303,323,324) with cases of concomitant ZES and insulinoma (87,303,324-331),GRFomas (326,332,333), ectopic Cushing’s syndrome (66-68,113,325,334-345), glucagonomas (87,324,328,342,343,346-348), VIPoma (324,325), somatostatinomas (339,349), carcinoid syndrome (87,325,327,343,350) and PTHrPomas (351)  all described.  Even though secondary F-pNEN syndromes have been described in ZES, in general they are relatively infrequent, except for the development of Cushing’s syndrome in patients with advanced metastatic gastrinomas (66,68,109,113,325,336). In a prospective study from NIH of 45 ZES patients with a mean follow-up of 146 mos. from ZES, only one patient (2%) developed a second F-pNEN syndrome onset for a rate of 0.16%/yr (1% of patients every 6 yrs. of follow-up). This rate was considerably less than that reported in another study (352) of 353 patients with all pNEN(169=gastrinomas) in which 6.8% of all patients developed a secondary pNEN syndrome over a 19-mo. mean follow-up(rate=4.3%/yr.). Ectopic Cushing's syndrome has been more frequently reported in patients with ZES (27,113,334,336,345,353) as well as other pancreatic endocrine tumors (353-356). In a prospective study from the NIH (109) ectopic Cushing's syndrome developed in 4% of all patients with ZES studied (9/212), 17% (9/54) with liver metastases, 21% (7/33) dying of ZES-related causes and 25% (5/20) with bone metastases.  It was an independent predictor of poor survival (p <0.005) with patients having a 10-year survival of 0%.  Ectopic Cushing's syndrome only developed in patients with metastatic liver disease.  Similar to bone metastases, development of ectopic Cushing's syndrome was a strong predictor of poor prognosis with patients only surviving a mean of 1.7+0.4 years after its onset (109). 

 

The gastrin-gene covers a 4 kilobase area and consists of 3 exons and 2 introns, with the coding region translating into a 101-amino acid peptide, pre-progastrin (7,7,8,170,357,358). In normal antral G-cells, pre-progastrin undergoes a number of post-translational processing steps including dibasic cleavages, removal of the glycine extended COOH-terminal amino acids and sulfation, leading to the formation of progastrin, then COOH-terminal glycine-extended forms and finally the biologically active forms consisting of 2 COOH-amidated gastrins, gastrin-17 (G-17) and gastrin-34 (G-34), existing in sulfated and non-sulfated forms (7,8,170,357,358). Normally, >90% of antral gastrin is G-17, while in the duodenum only 40-50% is G-17(7,8,357,358).  In the circulation, normal G34 is the predominant form (>60%) and sulfated/non-sulfated forms occur equally (7,8,357,358). In contrast, in patients with gastrinomas the relative concentrations of G-17 are higher (74-80%), and increased concentrations of partially processed forms are found (progastrin, NH2- and COOH- terminal fragments, COOH-glycine extended fragments, incompletely amidated fragments) (7,8,27,357-360).  Alterations in post-translational processing have been correlated with the presence of metastatic disease (7,8,27,359,360); however, no prospective studies have established their usefulness in an individual case (7) and they are currently rarely measured.

 

Chromogranin A (CgA) is a 48-kilodalton protein stored in secretory granules of neuroendocrine cells and is widely used as an immunocytochemical marker to identify tumors as NENs (27,236,280,301,322,361-364).   CgA is released simultaneously with the release of polypeptides and thus can be used as a general plasma tumor marker for NENs (322,361-363,365-368). Plasma CgA levels are elevated in 80-100% of ZES patients, as is the case in patients with other pNEN/GI-NENs (carcinoids) (322,365-370).  Changes in plasma CgA levels are reported to be useful for assessing changes in tumor mass in some studies; however, in other studies, including in patients with gastrinomas, it has been found to be a relatively insensitive marker for tumor progression and/or NEN identification (115,116,364-369,371-375). One major problem with using plasma CgA as a tumor marker in ZES patients is that the chronic hypergastrinemia causes gastric ECL cell proliferation which increases plasma CgA (24,362-364,368,376). Thus, in ZES, elevated plasma CgA can come from the gastrinoma or from hyperplastic ECL cells (24,377-379). Unfortunately, plasma CgA is also increased by inflammatory disorders, other endocrine diseases, the use of proton pump inhibitors, gastrointestinal disorders, cardiovascular disorders and altered renal function, and therefore minimally or moderately elevated plasma CgA levels in the range frequently seen with small gastrinomas/pNEN overlap with values found in these other disorders (361-364,368).

 

In patients with gastrinoma, a number of agents stimulate the release of gastrin including secretin (61,74,75,84,380-384), glucagon (385-387), bombesin/GRP (380,388), muscarinic cholinergic agonists (380), beta-adrenergic agonists(389), calcium (74,75,380,383,384,390) and a standard meal (74,384,391,392); in addition, native and synthetic somatostatin analogues (octreotide, lanreotide) can decrease serum gastrin (7,103,393-396).  Studies demonstrate that gastrinomas possess secretin receptors, somatostatin receptors, bombesin/GRP receptors, and calcium-sensing receptors (380,388,397-401). These findings have been used clinically for ZES diagnosis with the development of secretin, calcium, glucagon and standard meal provocative tests and the use of somatostatin analogues to control acid hypersecretion (7,56,74,75,103,391,394).  The clinical aspects of gastrin provocative testing will be discussed in a later section on ZES diagnosis. Currently, somatostatin analogues are uncommonly used to control acid hypersecretion in ZES patient, because they must be given parenterally, whereas effective long-acting, oral antisecretory agents such as PPIs are available and are the drugs of choice (29,103,142,151,181,182,396). Somatostatin analogues are used for their anti-growth effects or to control ectopic secretion of other hormones in gastrinoma patients, as in other F-pNEN (25,58,142,148,152,155,402,403), and this will be discussed in later sections. Furthermore, the presence of somatostatin receptors on gastrinomas, as well as on other pNEN/NENs, is used for tumor localization, as well as to deliver cytotoxic radiotherapy to patients with advanced tumors (51,129,133,134,136,137,139,142,185,404), both of which will be discussed later in the treatment sections.

 

The exact mechanisms by which secretin, calcium, glucagon, or a meal stimulate an increase, and somatostatin analogues a decrease, in serum gastrin in ZES patients is not completely clear (27,74,397). The most likely explanation is a direct effect on gastrin release from the gastrinoma through activation of specific receptors which are known to be present on these cells, although others have proposed (in the case of the secretin-test) that it is an exaggerated physiological response (397,405,406). The evidence for a direct effect is that presence of receptors for these agents which have been shown on gastrinomas. Furthermore, in dispersed/cultured gastrinoma cells, calcium and secretin stimulate gastrin release, and secretin activates adenylate-cyclase in these cells which stimulates gastrin release (380,393,397,399,407-409). whereas somatostatin causes inhibition (393,409). Also, a direct relationship has been shown between the magnitude of expression of secretin receptors on gastrinomas and the magnitude of the secretin-stimulated response in ZES patients (397).

 

The exact pathogenesis or cell-of-origin of pancreatic or duodenal gastrinomas remains unclear. As mentioned above, gastrinomas and other pNEN were frequently called islet cell tumors, however it is still controversial that those arising in the pancreas actually originate from pancreatic islets (410,411). Numerous older studies have reported that gastrin is found only in the fetal/developing pancreas in islet cells so if pancreatic gastrinomas arose from islets, the possible cell of origin was unclear (8,27,412-414). Passaro and colleagues proposed two different subpopulations of gastrinomas existed (414-416). One group occurred in the gastrinoma triangle (duodenum, pancreatic head, peri-duodenal lymph nodes), which were to the right of the superior mesenteric artery, which originated form the ventral pancreatic bud and were relatively more benign with frequent positive lymph nodes, low rate of liver metastases and high cure rate (414-417). In contrast, the second group occurred outside the gastrinoma triangle, were entirely within the pancreas, were to the left of the superior mesenteric artery, arose from the dorsal pancreatic bud, and were more aggressive with lower cure rates and liver frequency of liver metastases (414-417).   Numerous studies support the conclusion that duodenal and pancreatic gastrinomas differ in biologic behavior (109,110,127,170,172,281,415,418-420).  Furthermore, in numerous studies gastrin-producing G cells were found in the adult duodenum, but not in the adult pancreas; therefore, supporting the proposal that different cells-of-origin were likely for duodenal and pancreatic gastrinomas (27,40,412,413,418,419). This proposal is further supported by a study (420) which demonstrates that all 15 duodenal gastrinomas show sonic hedgehog expression with none showing expression of pancreatic-duodenal homeobox 1, whereas the reverse pattern was seen in 11 pancreatic gastrinomas. It has been suggested that gastrinomas in the gastrinoma triangle area originate from stem cells in the ventral pancreatic bud,  and that these cells become dispersed in lymphoid and duodenal tissue and give rise to the gastrinomas in this area (414)  Others have proposed that gastrinomas originate from multi-potential, endocrine- programmed stem cells that undergo inappropriate and incomplete differentiation toward the G-cell in the islets/pancreas (27,413,418). Although some recent studies propose that cancer stem cells, which have been described in a number of solid tumors, could also be important in the pathogenesis of pNEN or GI-NENs, at present they have not been convincingly identified and isolated in GEP-NEN pathologic samples (421). A recent detailed lineage tracing study of gastrin expressing cells in pancreas provides some of the strongest evidence that pancreatic gastrinomas in sporadic ZES cases may originate from the islets (412). In this study (412) during fetal stages up to postnatal day 7 gastrin expressing cells were abundant, whereas a small population of gastrin expressing cells existed in adult islets which co-expressed glucagon or insulin and the pancreatic gastrin positive cells were found to originate from PTF1a+ and neurogenin 3 expressing progenitors that were a subpopulation of alpha and beta cells. Furthermore, disruption of the MEN1 gene in the progenitor cells, resulted in the development of pancreatic gastrin-expressing tumors, but no animals developed ZES (412).  Recent studies provide evidence that gastrinomas in MEN1/ZES may have different pathogenesis than sporadic gastrinomas and also the development of the duodenal gastrinomas and pancreatic tumors differ in these patients. In MEN1/ZES patients, it has been proposed that the duodenal gastrinomas arise from the G cells by a process of hyperplasia similar to proposed for the response of ECL cells to gastrin in the stomach (422,423). In MEN1/ZES patients, it is proposed that the pivotal event in the development of the multifocal gastrin neoplasms is the allelic deletion of the second MEN1 allele (422,424).  However, this sequence was not seen in sporadic duodenal gastrinomas (422,424). Previous studies (424) have reported that in MEN1 gastrinomas only 46% of the tumors exhibited LOH at the MEN1 locus with the remaining 55% not exhibiting allelic loss of the MEN1 gene locus, despite having precursor lesions such as hyperplastic G cells in the crypt base or in Brunner’s glands, suggesting that mechanisms besides loss of the wild type MEN1 allele may be involved in the transition from G-cell hyperplasia to duodenal gastrinoma (425). Recent studies (170,173,174,426,427) using mice with targeted MEN1 deletion bred onto a somatostatin null background and treated with omeprazole to induce hypergastrinemia developed gastric carcinoids as well as hyperplastic gastrin-expressing cells in the lamina propria of the proximal duodenum expressing markers for enteric glial cells such as glial fibrillary acidic protein.  Because in these experiments, the MEN1gene had been deleted from the epithelial cells, this suggested a possible non-cell autonomous mechanism was involved. This conclusion was supported by a study (427) reporting duodenal gastrinomas as well as their metastatic lymph nodes showed immunohistochemical staining for enteric glial cell markers, whereas it was not seen in pancreatic gastrinomas (170,174,427). From these findings the authors (170,174) proposed that duodenal gastrinomas in these patients may arise from a neural crest-derived cell and /or an endodermally derived epithelial cell.

 

 For pancreatic pNEN in MEN1 patients, two studies have come to different conclusions, with one concluding that PETs arise from duct cells (411) and the other concluding that they arise from islet cells (422,428).

 

Important insights into the natural history and prognosis of the gastrinoma per se have been provide by a number of long-term studies of patients with or without MEN1 (64,87,89,93,109-111,258,314,315,429-434). In ZES patients without MEN1(sporadic ZES), 25% of their gastrinomas show aggressive growth behavior (109,110). Aggressive growth is associated with a decreased ten-year survival (30%) compared to the excellent survival in those with nonaggressive disease (10 yr.-survival=96%) (110).  A similar aggressive growth pattern has been described in patients with MEN1/ZES; however, the percentages are different, with only 14% demonstrating aggressive growth (111). In the sporadic ZES patients, those with aggressive growth are characterized by more frequently having liver metastases, a pancreatic primary, a large primary (>3 cm), a short disease history, higher gastrin levels, female gender, and sporadic ZES (109,110). In general, patients with MEN1/ZES have a better prognosis than patients with sporadic ZES (110). Finally, long-term studies demonstrate that even in patients with liver metastases, their rate of tumor growth may markedly vary with 42% demonstrating rapid growth, 26% having no tumor growth and 32% demonstrating a slow growth over a three-year period (429). Deaths only occurred in the subgroup with rapid tumor growth (62% died during follow-up) (429). This result has important implications for treatment in gastrinomas as well as other NENs with a number of studies demonstrating the rate of tumor growth prior to treatment is an important prognostic predictor of patient’s survival, outcome and even response to different therapies (402,429,435-440).

 

MOLECULAR PATHOGENESIS

 

The molecular pathogenesis of gastrinomas, similar to other pNEN /NENs, differs from more common adenocarcinomas, but has remained largely unknown until recently (29,170,172,174,427,441-443,443-448). In contrast to many adenocarcinomas, mutations of common tumor suppressor genes (p53, retinoblastoma, etc.) and oncogenes (Ras, myc, jun, Src, etc.), are infrequent in gastrinomas and other pNEN (29,158,308,310,311,441,443,443,446,448-453). This is not the case with G3NECs, which are uncommon in gastrinomas (<5%), which have a higher mutation rate for p53, Rb and p16(158,310).  Whereas mutations of common oncogenes or tumor suppressor genes are uncommon in pNEN, recent studies provide evidence that both the p53 pathway and the retinoblastoma (RB) pathway are frequently altered in pNEN (454-457). The Rb pathway is inactivated in most pNEN (including gastrinomas) (455) by amplification of genes encoding the cyclin-dependent kinases Cdk4/Cdk6. A second study (454) found a low rate of p53 mutations in pNEN (<3%); however, the p53 pathway was altered in 70% of pNEN through aberrant activation of its negative regulators- MDM2 (22%), MDDM4 (320%), and WIPI (15%). A third study found the p53 target gene PHLDA3 is frequently inactivated in pNEN and this correlates with tumor progression and poor prognosis (456,457)

 

As discussed above, gastrinomas, as well as other pNEN not only occur sporadically (75%-gastrinomas), but can also occur as part of various inherited syndromes (30,114,158,195,197,458,458-462), including MEN1, tuberous sclerosis, neurofibromatosis, von Recklinghausen’s disease and von Hippel-Lindau disease (VHL), and investigations of the altered genes in these diseases have provided insights into the molecular pathogenesis of pNEN (30,195,196,442,449). Approximately 20-25% of patients (Table 3) (27,30,87,89,463) with ZES have Multiple Endocrine Neoplasia type 1 syndrome (Wermer’s syndrome) (MEN1/ZES). MEN1 is an autosomal dominant disorder due to mutations in the MEN1 gene on the long arm of chromosome 11 (11q13). The MEN1 gene has 10-exons encoding for a 610 amino acid protein, MENIN (30,87,97,317,448,464). A recent sequencing study (446) showed in sporadic pNEN, MENIN is also important with 44% having an inactivating mutations of the Multiple Endocrine Neoplasia-type 1(MEN1) gene. Mutations in the MEN1 gene occur in one-third of sporadic gastrinomas (30,441,449,450,465). Furthermore, 5-95% of patients with sporadic pNEN have loss of heterozygosity (LOH) at the MEN1 locus(11q13) including in 44% of sporadic gastrinomas (30,318,463). These results strongly suggest alterations in MENIN are important in the pathogenesis of sporadic gastrinomas and in the inherited syndrome, MEN1.  The exact molecular alteration that occurs with MENIN mutations that results in pNEN, including gastrinomas, is not clear. However, it is known that MENIN is a nuclear protein that interacts with a large number of proteins (30,98,463,464,466,467). MENIN interacts with SMAD3; RPA2(a DNA-processing-factor); the AP1-transcription factor, JunD; nuclear factor-B(NF-B), Pem, FANCD2 (a DNA-repair-factor), nucleoside diphosphate kinase, NM23 cytoskeletal-associated proteins and various histone-modifying enzymes (30,463,464,466,467). A recent large WGS study (443) of pNENs found an MEN1 mutation in 41% of the pNENs and altered copy number in 70% and concluded that MEN1 played a central core pathway role in pNENs molecular pathogenesis interacting with each of the key cascades found to be altered in these tumors. This included MEN1(443) interacting with altered key genes involved in DNA damage repair (MLH1-4, MSH5, etc.), chromatin modification (SETD2, MLL3, etc.), altered telomere length (DAXX, ATRX, etc.),  mTOR signaling (PTEN, TSC1-2,etc), homologous recombination and double break repair(CHEK2, BRAC1,TP53, etc.) and cell cycle regulation(CDK2C, JNK, etc.).

 

In recent sequence studies(446) of pNENs was carried out, and it was found that in addition to alterations in the MEN1 gene in 21-100%% (443,446,448), mutations were found in frequently in genes encoding for two subunits of a transcription/chromatin remodeling complex consisting of DAXX (death-domain associated-protein) (25-40%) and ATRX (alpha-thalassemia/mental retardation syndrome X-linked) (18-35%), followed by mutations in mTor pathway genes (15-54%) (443,446,448).  MEN1/DAXX/ATRX are important in the epigenetic landscape including DNA methylation, histone modifications, posttranscriptional regulation, and are thought to play important roles in the pathogenesis of pNEN (308,443,446,448,452). Recent studies provide evidence that pNEN are heterogeneous (308,447,452,468,469). The presence of the MEN1/DAXX/ATRX mutant phenotype, which is present in 60% of pNEN, has been reported to correlate with a worse prognosis (448,452,470-474). The MEN1/DAXX/ATRX mutant profile of pNEN is associated with an islet alpha-cell lineage pattern (high ARX, low PDX1, high HNF1A expression) and has a much worse recurrence free survival (470). Numerous recent studies in pNENs (114,444,445,475,476) including gastrinomas support the importance of the cell lineage (alpha cell, beta cell, intermediate pattern), as well as alterations in DAXX, ATRX, alternative lengthening of telomeres and MEN1 mutations as determinants and prognostic factors for identify patients with pNENs showing  aggressive growth and cohorts associated with decreased survival.

 

The VHL locus occurs at 3p25, and chromosome 3 alterations are reported in 21-50% of sporadic pNEN (449,477). However, these chromosome 3 alterations are rarely associated with a mutation at the VHL locus, suggesting that it is not involved in pNEN development; however, a locus telomeric to the VHL locus may be involved. Recent studies provide evidence for the importance in pNEN/gastrinomas of alterations in the DPC4/SMAD gene (20% in pNEN), the p16/MTS1 tumor suppressor gene (50-90%), mTor/Akt/PI3K pathway, amplification of the HER-2/neu proto-oncogene, as well as increased expression of a number of growth factors and/or their receptors (platelet-derived growth factor, hepatocyte growth-factor, epidermal growth factor, insulin-like growth-factor 1) (441,442,449,450,478,479). Numerous recent studies provide evidence that the mTor/Akt/PI3K pathway is particularly important for mediating the growth of pNEN (478,479). This evidence includes the success of the mTOR inhibitor, everolimus, in extending disease-free survival in patients with advanced pNEN (480), but also studies showing the mTor/Akt/PI3K/ signaling cascade plays a central role in pNEN cell growth and proliferation (442,478,479,481). Additional evidence for the importance of the mTor/Akt/PI3K pathway comes from a study showing mutations in mTor pathway genes (15%) in sporadic pNEN (443,446) as well as from a study (482) reporting the effects of a single nucleotide polymorphism. Replacing arginine by glycine in codon 388 (R388)) of the fibroblast growth factor receptor 4 (FGF4) (482) diminishes the responsiveness to mTor inhibitors in pNEN, and its presence in pNEN is associated with advanced tumor stage and liver metastases.

 

Numerous chromosomal alterations have been identified in sporadic pNEN and accumulate with advancing stage and tumor progression (158,308,452). Comparative genomic hybridization (CGH) and genomic-wide allelotyping studies report that chromosomal gains/losses occur frequently in pNEN, including in gastrinomas, and that the distribution of these changes differs between GI-NENs (carcinoids) and pNEN, supporting the conclusion that they have a different pathogenesis (29,48,449-451). In pNEN, allelic losses occur most frequently at chromosomal locus 1p (25-75%), 1q (20-90%), 3p (40-95%), 11p (30-50%), 11q (30-70%) and 22q (40-95%) (441,449,450,478). With pNEN, chromosomal gains occur most frequently at 17q (10-55%), 7q (15-70%), and 4 q (33%) (441,449,450,478). A number of these alterations are associated with malignant behavior including deletions at chromosome 1, 3p, 6, 11q, 17p and 22p, and gains on chromosome 4, 7, 14q, Xp (441,449,450,478).  Deletions are more frequently seen in the primary tumor and gains in the metastases (452). The commonly mutated genes in pancreatic cancer such as KRAS, TP53, p16/cdk2A and SMAD4 and not commonly mutated in pNEN (446,469).

 

Results have been reported from a number of studies in which pNEN were studied using microarrays to perform gene expression profiling (449,450,478,483-485). Results from 8 studies in pNEN have been summarized (478) and they demonstrate a wide variation in the number of genes up-regulated (45-668) or down-regulated (25-323). These studies and others (483,484,486) describe a number of gene alterations that correlate with prognosis, survival, and relapse, but it is not clear presently which gene changes are of most important in the molecular pathogenesis of the pNEN.

 

CLINICAL FEATURES AND PRESENTATION: ZES

 

ZES most frequently occurs between the ages of 35-65 with a mean age of 41 yrs. (range-41-53) (7,27,62,235) but is reported in both children (487,488,488-491) and the elderly (27,62,63,235). There is a slight male predominance and in most series 20-35% of cases occur as part of the MEN1 syndrome (Table 3) (30,62,87,88). The main presenting symptoms are summarized in Table 3. Abdominal pain remains the most prominent symptom (>70%), and it is most frequently due to the presence of a duodenal ulcer, with a lesser subset presenting with pain due to gastro-esophageal reflux disease (GERD 20-44%%) (62). Whereas, in the older literature the ulcer was frequently described as occurring in abnormal locations outside the duodenum or as multiple ulcers, at present, most ZES patients present with a typical duodenal ulcer that is indistinguishable form that seen in idiopathic peptic ulcer disease (27,28,62). Similarly, the pain at presentation is similar to that seen in patients with idiopathic gastro-esophageal peptic disease (28,62). Diarrhea was uncommonly reported in older series, however in more recent series it is present in more than one-half the patients, and in 9-20% of patients it is the principal or a prominent presenting feature (Table 3) (51,55,60,62,87,490,492-495). The diarrhea differs from that seen with VIPomas in that it is characteristically not large volume (<1 L/day) and is more characterized by increased frequency and mild steatorrhea, if it is present (28,62,222). The presence of the diarrhea is an important clinical clue that when associated with peptic ulcer disease, should suggest the diagnosis of ZES (9,24,28,51,55,59,62,181), and this will be discussed in more detail in a later section on diagnosis of ZES.

 

Table 3. Clinical Features of Patients with ZES

Feature

NIH data (n= 261)

Literature data (range)

INITIAL SYMPTOM (percentage)

Abdominal pain

75

26–98

Diarrhea

73

17–89

Heartburn

44

0–56

Nausea

30

8–37

Vomiting

25

26–51

Bleeding

24

8–75

Pain and bleeding

19

19–44

Pain and diarrhea

55

28–56

FINDINGS AT PRESENTATION

Prominent gastric folds

94%

(10-30%)

OTHER CLINICAL FEATURES

Gender (percentage male)

56

44–70

Mean age onset (years)

41

41–53

MEN1 present (percentage)

22

10–48

PAST CLINICAL FEATURES

History-confirmed peptic ulcer (percentage)

71

71–93

History of Esophageal stricture (percentage)

4

4–6

History of Abdominal perforation (percentage)

5

5–18

Note. NIH data are from 261 patients with ZES prospectively studied (62). Literature data are from 11 series (50,64). Abbreviations: ZES-Zollinger-Ellison syndrome, MEN1-Multiple Endocrine Neoplasia type 1, ND-no data

 

In the past before effective nonsurgical methods to control acid hypersecretion was available, many ZES patients with ZES developed severe complications of the gastric acid hypersecretion (1,28,205,235). These included severe peptic ulcer disease (with perforation or penetration, with or without fistula formation), bleeding (22-45%), strictures leading to gastric outlet obstruction) (up to 20%) or GERD complications (esophageal ulcers, strictures, ulcers, bleeding, Barrett’s, rarely perforation) (up to 20) (1,28,62,205,235,496,497). At present, because of the widespread off label antisecretory drug use, it is uncommon to have patients present with symptoms due to complications from advanced peptic ulcer disease /GERD (62,498-501).  In the NIH prospective study (62), only 4% of the 261 ZES patients had a perforation due to a peptic ulcer disease and 5% had esophageal strictures, although 10% had duodenal scarring due to chronic peptic ulcer disease (Table 3).  At present, while a duodenal ulcer is usually present at diagnosis, it is not advanced, with 18-65% having no ulcer present (27,62,205), although up to 91% have a history of peptic ulcer disease (Table 3).

 

The diarrhea is a consequence of the acid hypersecretion and not due directly to the hypergastrinemia per se, as shown in numerous studies which report any method that controls the acid hypersecretion (nasogastric section, medications, surgery), without changing the level of hypergastrinemia, all lead to a decrease or cessation of the diarrhea (9,28,55,62,222,502).

 

In early studies of ZES patients, gastroesophageal reflux disease (GERD) symptoms (i.e., heartburn, pain) were either uncommon or not reported, so that 7 early series of ZES patients reported before 1986, the GERD symptoms were reported to occur in only at 0-2% of all patients (62). More recent GERD symptoms are increasingly reported in series of ZES patients, with 44% of 261 ZES patients having GERD symptoms at presentation in the prospective NIH series(62), and 49-61% in other series in the recent literature (Table 3) (62,498,503). Other gastrointestinal symptoms such as nausea (30%) and vomiting (25%) as well as weight loss (17%) are not infrequent in ZES patients at presentation (Table 3). The cause of the weight loss can be multifactorial, including from effect of the gastric acid hypersecretion on intestinal absorption causing malabsorption, decreased appetite, or from advanced metastatic disease resulting in anorexia, pain or other symptoms (62). In most patients early in their disease course or without widespread metastatic disease, the weight loss is due to maldigestion and malabsorption (28,62,222).

 

Approximately 20-25% of patients (Table 3) (27,30,87,89,190,463) with ZES have Multiple Endocrine Neoplasia type 1 syndrome (Wermer’s syndrome) (MEN1/ZES) and these patients have a number of important differences including clinical presentation and disease course from patients with ZES without MEN1(sporadic ZES) (27,30,64,87,89,190,463). These aspects will be discussed in the next section.

 

MEN1/ZES-GENERAL AND CLINCIAL FEATURES

 

MEN1/ZES-General

 

As discussed above, MEN1 is an autosomal dominant disorder resulting from mutations in the MEN1 gene located on the long arm of chromosome 11 (11q13) (30,87,317,464). The MEN1 gene has 10-exons with 9-exons encoding for a 610 amino acid protein, MENIN (30,87,317,464). The exact molecular alteration that occurs with MENIN mutations that results in pNEN, including gastrinomas, is not clear.

 

MEN1 causes NENs and hyperplasia in multiple endocrine organs (Table 4) that classically includes: hyperparathyroidism due to multi-gland parathyroid hyperplasia; pancreatic NENs (nonfunctional pNEN>gastrinoma> insulinoma>>other) (Table 4); and pituitary adenomas (prolactinomas>ACTH-secreting>growth hormone-secreting) (Table 4) (30,87,463,504,505). Each may be associated with a functional syndrome. The most frequent pNEN is a nonfunctional pNEN (NF-pNEN) with 80-100% developing microscopic NF-pNEN; however, NF-pNEN cause symptoms in only 0-13% (30,87).  Gastrinomas are the most frequent functional pNEN (mean 54%, range 20-61%) (62,87,463,505,506) (Table 3). In addition, classically, adrenal tumors (rarely functional) and thyroid disease can occur in <50%, and these patients have an increased incidence of carcinoid (stomach, lung, thymus) (Table 4). Recently it has become recognized that these patients can develop a number of other tumors including smooth muscle tumors (leiomyomas, leiomyosarcomas), CNS tumors (meningiomas, schwannomas, ependymomas); and skin tumors (angiofibromas> collagenomas >lipomas >melanoma) (Table 4).

 

As will be discussed in the separate sections below the presence of MEN1 in ZES patients is important to recognize because it affects all aspects of the disease including: the pathogenesis, the pathologic findings; the clinical presentation; the treatment approaches; the prognosis and the role of surgery; and the need for genetic counseling (9,30,64,87-89,93,181,190,463,505,507-509).

 

Table 4. Clinical Features of Multiple Endocrine Neoplasia - Type I (MEN1)

 

Average Frequency (range)

% of all patients

Hyperparathyroidism

97 (78-100)

Pancreatic Endocrine Tumors

Pancreatic endocrine tumors (panNENs)

81-100

Nonfunctional or PPomas

80-100 (microscopic)

0-13 (symptomatic)

Gastrinomas

54 (20-60)

Insulinomas

18 (7-30)

Glucagonomas

3 (1-8)

VIPomas

1 (1-15)

Somatostatinomas

0-1

GRFoma

<1

Pituitary Tumors

54-65 (15-100)

Prolactin-secreting

15-45

Growth-hormone secreting

6-20

Cushing's syndrome

16

Adrenal Tumors

    Cortical adenomas

    Hyperplasia, carcinoma (uncommon)

27-36 (symptoms<2%)

 

Thyroid Tumors- adenomas

0-10 (0-30) (<1% symptomatic)

Carcinoid Tumors

Gastric (ECLoma)

7-35 (symptomatic<5%)

Lung

0-8

Thymic

0-8

Skin Tumors

40-100

Angiofibromas> collagenomas> café-au-lai> macules> lipomas

88%>72>38>34(symptomatic<1%)

Smooth muscle tumors- Leiomyomas, leiomyosarcomas

1-8% (symptomatic<1%)

CNS tumors- Meningiomas>ependymomas, schwanomas

0-8%>0-1% (symptomatic<1%)

Data from references (27,30,87,89,257,463,505).

 

MEN1/ZES-Clinical Features

 

For the 20-25% of patients (Tables 4 and 5) (27,30,87,89,463) with ZES with the Multiple Endocrine Neoplasia type 1 syndrome (MEN1/ZES), the presentation of mild hyperparathyroidism is best detected by an assessment of plasma ionized calcium levels, combined with assessment of plasma parathormone levels using a more sensitive assay such as intact PTH-IRMA assays (87,510). In general, the clinical manifestations of ZES are largely similar to those of patients with sporadic and MEN1/ZES, although patients with MEN1/ZES tend to have diarrhea less frequently as one of the presenting symptoms (26% vs 53%) (511). A carefully taken clinical, personal, and family history of endocrinopathies can be particularly important in suspecting MEN1/ZES, because up to 75% have a family history of MEN1 (Table 4) and 24-42% have a personal history compatible with renal colic (30,87,89). The presence of the MEN1 can affect the manifestations of ZES and aspects of its presentation, which will be discussed in a later section dealing with the diagnosis of ZES. In one study (64) the delay in diagnosis of ZES was greater in MEN1/ZES patients than in sporadic cases (7.4 ± 4.9 yrs. vs 3.9 ± 0.2 yrs, p=0.022).

 

Table 5. Features of Patients with MEN1/ ZES

Feature

NIH Data (n=106)

Mean (range)

Literature (range)

I.   MEN1 Tumor/hyperplasia

Hyperparathyroidism

100 (94%)

88% (78-100%)

Pituitary disease

60%

31% (28-60%)

Adrenal abnormality

45%

13% (13-35%)

Other functional pNEN)

6%

15.7% 

Smooth muscle tumor

7%

0.2%

Thyroid disease

6%

5% (3-25%)

CNS tumor (meningioma, etc.)

8%

<1%

Carcinoid

Gastric

Bronchial

Thymic

30%

20%

8%

6%

6%

4%

2%

2%

Skin tumor

Lipoma

Melanoma

Collagenoma

Angiofibroma

 

5%

2%

72%

88%

 

3%

<1%

<1%

<1%

II. Age/duration

Age (yrs.

Age at study

Age at onset ZES

Age onset MEN1

 

51.2 ± 1.2 (23.8 – 80)

29.8 ± 1.1 (10.2 – 61)

34.7 ± 1.0 (12.1 – 61)

 

43.5 ± 0.5 (43-51)

36.6 ± 0.6

34.1 ± 0.5

Duration (yrs.)

Of ZES

Of MEN1

 

16.6 ± 0.9 (1.4 – 43)

21.5 ± 1.1 (1.4 – 58)

 

ND

ND

III. Other MEN1 feature

Family History of MEN1

70%

76%

First MEN1 symptom

Asymptomatic (screening)

HPT

ZES

Pituitary

other

 

5%

38%

45%

8%

2%

 

1.3%

38%

41%

12%

8%

Abbreviations:  MEN1 = multiple endocrine neoplasia type-1; ZES = Zollinger-Ellison syndrome; HPT = hyperparathyroidism; NIH = National Institutes of Health; ND=no data

NIH data are from (27,62,87,227,512-514)

 

DIAGNOSIS AND DIFFERENTIAL DIAGNOSIS

 

Differential Diagnosis: When Should You Suspect ZES

 

Despite many articles on the diagnosis of ZES, the diagnosis is continuing to be delayed by 4-7 years from disease onset with no shortening occurring over the last few years (27,29,56,59,62,184,511), and in fact, numerous studies support the conclusion the diagnosis is becoming more difficult and may be delayed even further in the future (24,51,55,56,59,77,78,515-517).  The diagnosis of ZES has historically been frequently missed and delayed, because ZES is an uncommon cause of PUD (1-3 new cases/million population/year), whereas idiopathic PUD is 1000-fold more frequent (2300 cases/ year /million) and their initial clinical manifestations can closely resemble each other (27,40,55,56,511,515). In the past when there was ineffective gastric antisecretory medications, ZES would often present with advanced, refractory peptic disease suggesting the diagnosis, however, at present, most patients present with a typical appearing duodenal ulcer, without complicated disease, as seen in patients with idiopathic PUD (28,40,511). This is occurring primarily because of the widespread available of potent gastric acid suppressant drugs (i.e., PPIs), which in conventional doses used to treat idiopathic GERD/PUD, also generally control the acid hypersecretion occurring in most ZES patients (70,105,518,519). The result of this change and others, are making the  diagnosis more difficult primarily for two reasons: first, the widespread use of PPIs can both lead to a false negative diagnosis of ZES because the symptoms and acid secretion are well controlled on the PPI, as well as lead to a false-positive diagnosis of ZES because it can induce fasting hypergastrinemia (24,51,55,56,76-78,515,517). Secondly, there is an increasing unreliability of serum gastrin assays which are essential for the diagnosis of ZES (55,56,516,520-522).  Each of these points will be discussed in detail later in this section.

 

A number of clinical/laboratory findings should suggest the diagnosis of ZES, and these are summarized in Table 6.

 

The presence of diarrhea with PUD is a particularly important clue to the possible presence of ZES, because in recent series when a history for diarrhea is careful sought it is present in >60% of ZES patients (Table 3,4,6). Conversely, in patients with idiopathic PUD/GERD, the occurrence of diarrhea is now uncommon, because the use of high doses of Mg containing antacids is now rare, which were a frequent cause of diarrhea in the past in patients with PUD/GERD (523,524). 

 

Table 6. Findings That Should Suggest Possible Diagnosis of ZES

I.  SYMPTOMS

A.  Peptic ulcer disease or gastro-esophageal reflux disease (GERD) with:

      diarrhea (>60%)

         without H. pylori or use of NSAIDs (PUD) (10-50%)

         with a long history of persistent or severe symptoms (i.e., >3 yrs.) (>50%)

         with refractoriness to treatment

         with a PUD complication (bleeding, perforation, penetration) (10-15%) 

         with a GERD complication (esophageal stricture, perforation, ulcer) (<5%)

         with weight loss (15-20%)        

         with family history of PUD or GERD

            with family history of endocrinopathy (esp. renal lithiasis, hyperparathyroidism)

B.  Persistent diarrhea (50-80%) which is:

         responsive to gastric acid antisecretory drug treatment (H2-R, PPIs)

         secretory

         associated with abdominal pain (50-70%)

         associated with malabsorption that is unexplained

         unexplained

          with esophageal disease/symptoms (40-70%)

          not responding to specific treatments of diarrheal diseases

         with weight loss (15%)

         with history of endocrinopathies or peptic ulcer disease (25%)

         with family history of endocrinopathies (esp. renal lithiasis, hyperparathyroidism)

II.  SIGNS

        Multiple peptic ulcers in unusual locations (<10%)

        Gastric outlet obstruction due to peptic ulcer disease (PUD) (3-10%)

        Esophageal stricture due to peptic ulcer disease (3-5%)

        PUD/GERD with findings of endocrinopathy or with MEN1-related tumors

        Prominent gastric folds on UGI endoscopy/Imaging (94%)

III.  LABORATORY/RADIOLOGY FINDINGS

PUD/GERD/unexplained diarrhea with:

         Hypergastrinemia

         Hypercalcemia

         Positive somatostatin receptor imaging

         Positive pancreatic mass

Numbers in parenthesis refer to percentage of ZES patients with these features.  Table prepared from data in ref. (56,62,73,87,89,133,512,525).

 

Furthermore, in any patient with chronic diarrhea without an evident cause, especially if it is fasting in nature, ZES should be suspected (Tables 3,4,6) (60,62,63,87,183,222,490,492-494,498).

 

In idiopathic PUD, H. pylori infection (>80%) or the widespread use of NSAID/aspirin are a frequent contributing factor, whereas they are frequently not present in ZES patients with a duodenal ulcer (approximately 50%), thus the lack of their presence should raise the possibility of ZES (60,80,526-531). Although less common than in the past, any patient with severe PUD/GERD or with a PUD/GERD complication (stricture, obstruction, perforation, bleeding, penetration), ZES should be suspected (Tables 3,5,6).  Because of the frequent occurrence of MEN1 in ZES patients (20-25%) (Table 5), any patient with PUD/GERD/unexplained diarrhea with a personal or family history of an endocrinopathy or a laboratory finding suggesting an endocrinopathy (especially hyperparathyroidism, renal stones, pituitary disease) should lead to suspicion of ZES (Tables 4,5,6).  An unappreciated finding that was not emphasized in the past, but which recent studies show is present in up to 94% of ZES patients is the presence of prominent gastric folds on upper gastrointestinal endoscopy or imaging studies (Table 3) (62).

 

Establishing ZES Diagnosis

 

If ZES is suspected, a fasting serum gastrin level (FSG) is generally the initial study performed (9,29,55,56,59,182,184). FSG levels are elevated in almost all patients with ZES (>99%), except in some unusual circumstances, such as post-parathyroidectomy in MEN1/ZES or post-noncurative gastrinoma resection (73,74,190,498,508,509,511,532-534). Because of its high sensitivity, the assessment of FSG is an excellent screening test (40,56,498). However, an elevation of FSG alone has a low specificity for establishing the diagnosis of ZES, and no matter how high the FSG level, is not sufficient for a ZES diagnosis (29,40,51,55,56,59,184,498). Many physicians assume that a very high level of FSG (>10-100-fold elevated) is indicative of ZES; however, similar magnitudes of elevation in FSG levels can occur in patients with chronic atrophic gastritis/pernicious anemia, renal failure or those taking PPIs (55,56). For example, FSG levels 10-20-fold elevated are not uncommon in patients with chronic atrophic gastritis (367,535-537). Furthermore, in patients without ZES taking PPIs, although hypergastrinemia is frequent seen (see next paragraph) (80-100%), the FSG is usually increased<3-fold, although in some patients it is increased >10-fold (55,56,59,76,78,376,515,538-543).

 

 Hypergastrinemia can either be physiological which develops as a physiological response to anything causing chronic hypo-/achlorhydria or it may be pathological or inappropriate which occurs in the presence of normal or even elevated gastric acid secretion, which would physiologically suppress gastrin release (Table 7). In humans the disorders causing physiological hypergastrinemia are due to CAG/pernicious anemia, use of PPIs, or H. pylori infections, which are much more frequent than ZES, and thus need to be excluded as a cause of the hypergastrinemia to establish a firm diagnosis of ZES.

 

Table 7. Causes of Chronic Hypergastrinemia

A. Associated with gastric acid hyposecretion/achlorhydria

Chronic atrophic gastritis (CAG)

Pernicious anemia

Treatment with potent gastric acid antisecretory agents (especially PPIs/uncommonly-H2-R)

H. pylori infections

Chronic renal failure

Post acid-reducing surgery/vagotomy

Inherited inactivating mutations in H+K+ATPase (1)

B. Associated with gastric acid hypersecretion

H. pylori infections

Antral G cell hyperfunction/hyperplasia

Gastric outlet obstruction

Chronic renal failure

Short bowel syndrome (rare)

Retained gastric antrum syndrome (rare)

ZES

1)-includes ATP4R mutations encoding for the alpha subunit of H+K+ATPase (544-547)

 

Therefore, historically, the next study generally recommended in a patient in whom fasting hypergastrinemia was detected and the possibility of ZES was being considered, was an assessment of gastric pH or fasting basal gastric secretory output (9,27,29,55,56,59,72,76,181,182,185,548). Gastric secretory rates are now rarely measured (55,70,72,105) and are available in only a few specialty centers and thus will be discussed briefly below for completeness. If the patient has fasting hypergastrinemia with a gastric pH≤2, ZES should be strongly suspected (55,59,70,73), as summarized in Table 8, because an NIH ZES study found that all ZES patients off of any antisecretory drug had a fasting gastric pH≤2 (70). As shown in Table 8 the diagnosis is established in the group with FSG increased>10-fold combined with gastric pH≤2. However, in the 60% of patients with FSG<10 fold elevated, the diagnosis is strongly suspected but not proven, because there are a number of other diseases (majority=rare) which can also cause these findings that are not ZES which are listed in Table 7 (51,55,56,59,73,74,76).

 

Table 8. Established and Recently Proposed (untested) Criteria for the Diagnosis of ZES

I.               Established Criteria for diagnosis of ZES

Required all: Fasting serum hypergastrinemia (FSG) and gastric fluid pH≤2.

1. If FSG> 10 times elevated (over ULN) and gastric pH≤2, the diagnosis of ZES is established (exclude retained antrum almost always by history) (40% of ZES patients)

2. If FSG is <10 fold elevated and gastric pH≤2, need to perform additional testing to exclude other causes of FSG/ hyperchlorhydria) (60%)

a. Secretin test positive (≥120 pg/ml increase)

b. Elevated basal acid output (>15 mEq/Hr)

II.        Possible new criteria for diagnosing ZES in patients with Fasting Hypergastrinemia in the absence of PPI therapy (gastric pH data not available) (Proposed; not evaluated or/and should not be routinely used)

 

A. Strongly supportive of ZES diagnosis

1. Active peptic ulcer disease (PUD) or a history compatible with recent PUD or improvement in diarrhea with PPIs combined with:

a. a positive somatostatin receptor scintigraphy imaging (SRI) with either 68Ga-DOTATATE PET/CT or 111In-DTPA-octreotide with SPECT/CT imaging.

b. a positive biopsy or cytology for a neuroendocrine tumor (NEN) (stronger support if a gastrinoma is found)

c. a positive secretin test.

d. known or strongly suspected MEN1 syndrome (i.e., a positive family history, hyperparathyroidism, or pituitary disease)

2.  A patient with known MEN1 or strongly suspected MEN1 (i.e., a positive family history, hyperparathyroidism, or pituitary disease) with a positive gastrinoma by cytology/biopsy

 

B. Moderately supportive of ZES diagnosis (consider this a tentative diagnosis)

1. Positive somatostatin receptor scintigraphy imaging (SRI) with either 68Ga-DOTATATE PET/CT or 111In-DTPA-octreotide with SPECT/CT imaging (sporadic disease only) or positive cytology or biopsy for a NEN, ideally a gastrinoma, (sporadic disease or MEN1 syndrome present) with a biopsy-proven absence of atrophic gastritis and negative autoimmune markers. (1,2)

 

C. Weakly supportive of ZES diagnosis (insufficient alone for even a tentative diagnosis)

1.  A patient with known MEN1 or strongly suspected MEN1 (i.e., a positive family history, hyperparathyroidism, or pituitary disease) with positive imaging or an SRI (1)

2. MEN1 syndrome absent but positive SRI or imaging for possible tumor (3). 

III.            Possible new criteria supporting the diagnosis of ZES in patients with Fasting Hypergastrinemia taking PPIs (4) (gastric pH data not available) (Proposed; not evaluated or and should not be routinely used)

 

A. Moderately supportive of ZES diagnosis (ZES is likely)

1. In a patient with or without MEN1 with active peptic ulcer disease (PUD) or a history compatible with recent PUD or improvement in diarrhea with PPIs combined with a positive biopsy or cytology for a neuroendocrine tumor (NEN) (stronger support if a gastrinoma is found.

2. In a patient without MEN1 with active peptic ulcer disease (PUD) or a history compatible with recent PUD or improvement in diarrhea with PPIs combined with

a positive somatostatin receptor scintigraphy imaging (SRI) with either 68Ga-DOTATATE PET/CT or 111In-DTPA-octreotide with SPECT/CT imaging. (5)

 

B. Weakly supportive of ZES diagnosis (consider this a tentative diagnosis)

1. In a patient without active PUD or history of diarrhea responding to PPIs without MEN1 with a biopsy-proven absence of atrophic gastritis and negative autoimmune markers with a positive SRI (6,7)

2. In a patient without active PUD or history of diarrhea responding to PPIs with known MEN1 or strongly suspected MEN1 (i.e., a positive family history, hyperparathyroidism, or pituitary disease) with a biopsy-proven absence of atrophic gastritis. (6) and negative autoimmune markers. (7)

 

C. Minimally supportive of ZES diagnosis (consider this a possible diagnosis only)

1. In a patient without active PUD or history of diarrhea responding to PPIs without MEN1 with a positive SRI

2. A patient with known MEN1 or strongly suspected MEN1 (i.e., a positive family history, hyperparathyroidism, or pituitary disease) without active PUD or history of diarrhea responding to PPIs with prominent gastric folds (8).

Part II and part III are from (55), Part I data from (55,73,74)

(1)   Under such conditions a NEN is confirmed but since MEN1 patients develop multiple NENs in various locations NEN(s) identified on SRI may not be a gastrinoma(s) (30,89,90,549).

(2)   Five biopsies (2-antrum, 2-corpus,1- incisura angularis) of the stomach are recommended to diagnose atrophic gastritis) (550,551).

(3)   SRI can be positive in nonngastrinoma NENs, numerous other tumors and both physiological and pharmacologic processes, so alone is not specific for gastrinoma (134,401,552).

(4)   The potential for a false-positive secretin test in patients with hypo-/achlorhydria limits the usefulness of the secretin test in patients taking PPIs unless the gastric pH≤2.

(5)   Under these conditions a NEN is likely but since MEN1 patients develop multiple NENs in various locations NEN(s) a positive SRI or biopsy may not be a gastrinoma(s) (30,89,90,549)

(6)   Five biopsies (2-antrum, 2-corpus,1- incisura angularis) of the stomach are recommended to diagnose atrophic gastritis) (550,551).

(7)   Biopsy and autoimmune markers can both be negative in confirmed autoimmune gastropathy (550,551).

(8)   Prominent gastric folds are present in 94% of ZES patients when initially seen, however they are not specific for ZES (62)

Abbreviations: ULN-upper limit of normal; FSG-fasting serum gastrin level; CAG-chronic atrophic gastritis, PPI-proton pump inhibitor; PUD-peptic ulcer disease; SRI-somatostatin imaging

 

The increased widespread use of PPIs has made the diagnosis of ZES more difficult (51,55,56,59,78,185). PPIs are potent gastric acid suppressants and because of their long durations of action (up to one week) (40,55,553-556) they induce hypergastrinemia in 80-100% of normal (55,56,59,78,184,376,515,538-543). The hypergastrinemia with PPIs develops rapidly (within 5 days); is a common finding among patients even without gastroesophageal disease since these agents are widely prescribed; are now available as over-the-counter medications; and are one of the most over-prescribed medications (185). The degree of hypergastrinemia is variable among PPI users, however in >20% of those taking PPIs in some studies the FSG increased >4-fold, and FSG levels>5-fold are not infrequent, with FSG levels even exceeding >10-fold increased have been reported (55,56,59,78,376,515,538-543). Furthermore, in contrast to H2R antagonists (cimetidine, ranitidine, nizatidine, famotidine), PPIs control symptoms in most ZES patients at conventional doses used in the treatment of idiopathic PUD/GERD (103,518,519,557-559), whereas with H2R antagonists, higher doses and/or more frequent dosing are usually needed than used to treat the typical patient with idiopathic GERD/PUD (40,72,103-106,559-562). In the past, ZES patients treated with conventional doses of H2R antagonists continued to have symptoms suggesting the diagnosis, whereas this is not the case with PPIs (56,56,59,515). Therefore, PPIs both mask and delay the diagnosis of ZES because of their effective symptom control at conventional doses and they also complicate the diagnosis of ZES by their ability to cause a false suspicion for ZES by inducing hypergastrinemia in normal subjects (56,515). However, the characteristic of PPIs which has most complicated the ability to diagnose ZES is their long duration of action which makes it difficult to take patients off the PPI, especially if ZES is present and can lead to complications (25,51,55,57,59,77,78,563) as will be discussed below.

 

If the gastric fluid is sampled in a patient with an elevated FSG while the patient is being treated with a PPI, and the gastric pH is >2 it is not possible with this information alone, to determine whether the hypergastrinemia is physiological or pathological. To resolve this problem, both historically and in the more recent American NANETs and European ENETs guidelines, as well as recommendation by experts for the diagnosis of ZES, it was recommended to stop the PPI for up to one week and then determining gastric pH and FSG (7,9,27,55,78,79,103,181,182,235,517,564). This approach should be performed with caution (25,55-57,59,77,520). In each of the above guidelines, it is pointed that this must be performed only after taking a careful history of the prior effects of stopping the PPIs, that high-dose H2R antagonists be substituted for the PPI (equivalent to ranitidine-300-600-every 4-6 hours), and this only be performed after it is established that acute PUD/GERD lesions are healed and the patient can be carefully followed during this time (55,56,59,77,520). After 5-7 days, the H2R can be stopped, antacids used and on the following day the repeat testing performed. A recent study (77) reported two patients with ZES who developed severe PUD/GERD complications when PPIs were suddenly stopped and recommended the diagnosis of ZES should be established by not stopping the PPI. A number of subsequently papers (55,56,59) have pointed out that it may be possible in some patients to decrease the dose/frequency of PPI to obtain gastric pH≤2 or use other findings (presence of gastrinoma) to establish the diagnosis; however, in most cases this will not be possible. The only established criteria, which usually require discontinuation of PPIs, are listed in Table 8 (Part I). Because of the potential risk in a patient who does have ZES, it has been recommended that in a patient suspected of having ZES on PPIs, that the trial off PPIs in such a patient is best performed at experienced centers (9,55,56,181,565).

 

In the past, gastric acid secretory studies were performed in most centers and the results used for ZES diagnosis. A study of gastric acid secretory results in 234 NIH ZES patients and 984 ZES patients from the literature reported study found that most ZES patients without previous gastric acid-reducing surgery have elevated basal and maximal acid outputs (BAO, MAO) with a mean BAO=42mEq/hr (normal<10 mEq/hr) and mean MAO=62.7 mEq/hr. (normal 48 mEq/hr. (men)/ 30 mEq/hr. (women) (70). In this study various levels of BAO, MAO, BAO/MAO ratios as well as basal gastric fluid volume and basal/maximal acid concentration or pH were proposed to identify ZES patients (70). A number of these secretory criteria had high sensitivity for identifying ZES patients with the commonly used BAO criteria of ≥15 mEq/hr. (no previous gastric surgery) or ≥5 mEq/hr (with previous gastric surgery) having a sensitivity of 87-90% and 81-100%, respectively (70). However, gastric acid secretion studies are now performed by very few centers, and thus not generally available, so these secretory criteria are no longer used. However, the above NIH study (70) demonstrated that >99% of ZES had a fasting gastric pH ≤2 off antisecretory drugs; therefore, this is a useful criterion that can be applied widely today. A recent study (566) described the validity of measuring gastric pH at the time of gastrointestinal endoscopy in ZES patients, so this criterion can be generally applied (70).

 

In a patient suspected of having ZES, who is found to have a FSG level >10-fold elevated and a gastric pH≤2 (which in 40% of ZES patients), the diagnosis is established without further testing (Table 8 (part I)), if the possibility of a retained antrum syndrome, which can mimic ZES (Table 7), has been ruled out by previous history/records (27,552,567). Unfortunately, most ZES patients (60%) present with a FSG<10-fold elevated (27,73,75,212) and are found to have a gastric-pH ≤2, which overlaps with a number of other disorders that can cause hyperchlorhydria with hypergastrinemia (Table 7, 8 (part b)) (7,28,81,105,134,183,184,527). The most frequent of this group are patients with H. pylori infection, which is most frequently thought to be associated with acid hyposecretion, but which can also result in hyperchlorhydria with hypergastrinemia (485,527,568,569), and may thus be particularly confusing. To exclude these other disorders (Table 7,8 (part b)) it is now recommended that a BAO and a secretin provocative test be performed (Table 8 (part b)). In the past, a number of gastrin provocative tests were reported to help identity the patients with ZES, which included tests using secretin (27,28,74,75,381,384,570), calcium (28,40,74,75,384,390,390,570) or a standard meal (27,28,74,384,391). The secretin/calcium tests were based on the finding that these agents stimulated an increase in serum gastrin in ZES patients compared to normal subjects (381,390), while with the standard meal test, ZES patients generally show <100%-increase in serum gastrin (74,384,391), whereas patients with antral G-cell hyperfunction/hyperplasia have an augmented and much larger response (74,384,391,571). At present, only the secretin test is widely used because of its convenience, sensitivity, specificity, and lack of side effects (33,61,74,83). A NIH study of 293 ZES patients (NIH) and 537 ZES cases(literature) (74) demonstrated that a value of 120 pg/mL increase with secretin had a sensitivity of 94% and specificity of 100% for ZES (74), and was more sensitive than previously proposed criteria of increases of  200 pg/ml, 50% over basal or 110 pg./ml (75,382,384,570), and therefore is the criterion recommended today (29,84,181,182). In some countries, secretin is not available, and a glucagon stimulation test has been proposed as an alternative (572); however, there is much less experience with the glucagon stimulation test. Unfortunately, the secretin tests results can be affected by PPI-inducted hypo/achlorhydria or by the presence of hypo/achlorhydria for other reasons; therefore, it cannot be reliably performed while the patient is taking PPIs or is hypo- or achlorhydric (573,574).

 

The availability of a reliable serum/plasma gastrin-assay is essential in all phases of the diagnostic evaluation of a patient with possible ZES. Unfortunately, a recent study (516) examined the accuracy of 12 widely used commercial assays for FSG assessment used by laboratories in both the US and Europe demonstrated and reported that only 5 assays reliably measured gastrin concentrations, with the others either overestimating or under-estimating the true value. Hence, 7 assays produced FSG values that could lead to false diagnoses or missed diagnoses (56,516,521). The inaccuracy occurred because inadequately characterized antibodies were used that either recognized precursor/inactive fragments or did not interact with all biologically active forms. The lack of a reliable FSG assay invalidates both the assessment of the FSG levels and the results of the secretin test. This is a potential major problem, and the best approach is to check to see if the laboratory performing the FSG assay for your patients uses one of the 5 reliable gastrin-assays listed in this paper or to obtain advice from a center that routinely performs FSG studies in your area for the assay they recommend.  A recent study (82) reports a rapid method to measure serum G17 an G34 using liquid chromatography-tandem mass spectroscopy which might prove useful to circumvent the above problems using RIA’s.

 

A recent study (55) pointed out in regular practice the criteria that most physicians are using to make the diagnosis of ZES are not those outlined above. This report (55) for the first time proposing new criteria which would support the diagnosis of ZES that did not involve the assessment of gastric pH, because it was found that in the last 20 cases of ZES reported in the literature, in only 5% (1/20) was an assessment of gastric acidity used in establishing the proposed ZES diagnosis of the cases reported in these studies. This has occurred primarily because of the difficulty physicians are having in assessing the gastric fluid acidity in these patients. The failure to measure gastric acidity in newly selected patients is due to a number of different contributing factors.  First, ln almost all community as well as many university hospitals, the assessment of gastric acid acidity is complicated by the general lack of its availability and the widespread use of PPIs. Secondly the vast majority of newly diagnosed ZES patients when the diagnosis is first suspected, the patients are almost all being treated with PPIs. As discussed above, these drugs have a long duration of action (up to 1 week), making it is difficult to assess the unsuppressed gastric acidity which can only be done by stopping the PPI for up to one week (40,55,70,74,75,254,553,555,556) which makes it difficult to take patients off the PPI. Third, because in a patient who has ZES, this approach is not without potential risk (51,55-57,59,77) and must be performed under control conditions, often using high doses of histamine H2 receptor antagonists, hence it is uncommonly performed. Although many current reports use the presence of an elevated FSG in combination with a positive SRI study to make the diagnosis of ZES (55), unfortunately, this is not specific for ZES, as patients can be achlorhydric/hypochlorhydria and have a non-gastrinoma neuroendocrine tumor that will be positive on SRI, and thus not have ZES. Furthermore, some propose the use of provocative test on PPIs to circumvent the need to stop the PPI to assess gastric pH (55), however a number of studies (573,574), but not all (61)conclude this is not a reliable alternative as the secretin test results are not reliable in a patient taking PPIs which frequently cause achlorhydria/marked hypochlorhydria which causes unreliable results (573,574). In a recent study (61) this conclusion has been challenged, because in 28 patients taking PPIs, no false positive or false negative secretin tests occurred and the sensitivity, specificity and positive predictive values for the secretin test were the same in patients taking or not taking PPIs.

 

It is important to remember that the new criteria (55) have been proposed to support the diagnosis of ZES, have not been widely evaluated and are not as strong as the classical criteria requiring increased FSG and gastric pH<2. These criteria were only proposed because 95% of physicians are not using the established criteria for the diagnosis of ZES (Table 8 (Part I)), and it is not apparent this practice will be reversed in the future. At present, it is best to refer these patients to a center that has expertise in the diagnosis of ZES to firmly establish the diagnosis by the established criteria. This is importance because it will dictate the course of management acutely and long term in the patient if ZES is present or not present (9,50,55).

 

TUMOR LOCALIZATION: ASSESSMENT OF PRIMARY LOCATION AND DISEASE

 

An assessment of both the primary tumor location and the tumor extent by various tumor localization modalities is needed at all steps in the management of ZES patients, similar to patients with other malignant NENS (9,27,29,33,53,135,140,140,142,181,575-582). It is initial needed in ZES patients to determine whether surgery should be considered and if so, to determine  the extent of surgery; to determine the location, extent and in some cases the rate of growth of metastatic disease prior to any anti-tumor treatment; to assess in MEN1 patients the possible presence of extra-duodenal-pancreatic NENs, such as carcinoid tumors (especially of the lung/thymus); to assess post-resection status; and to assess changes in tumor load with antitumor therapies or extent of recurrence, with time (9,27,29,33,53,129,135,140,140,142,142,181,291,575-578,578,579,579-582). Generally, more than one imaging modalities is used in different patients with the most frequent cross-sectional imaging study being a being a triphasic CT scan with intravenous contrast. In the case of SRI, in the past primarily somatostatin receptor scintigraphy (SRS) using 111Indium-labeled somatostatin analogues with SPECT imaging was used (141,582). But now in most centers it is replaced by the use of SRI with 68Gallium-labeled somatostatin analogues with positron emission tomographic imaging (PET-scanning) (45,129,134-137,139,141,157,291,293,578,582).

 

A wide range of different imaging modalities have been used in the evaluation of ZES patients (Table 9) (9,130,134,135,141,291,293,576,582,583). These include cross-sectional imaging (CT scanning, magnetic-resonance imaging (MRI), transabdominal ultrasound); selective angiography; somatostatin receptor scintigraphy (SRS) using 111Indium-labeled somatostatin analogues with SPECT imaging or 68Gallium-labeled somatostatin analogues with positron emission tomographic imaging (PET-scanning); endoscopic ultrasound (EUS); and the assessment of serum gastrin gradients either determined in the portal venous drainage through transhepatic venous sampling or in hepatic veins after selective, intra-arterial secretin injections (9,27,29,37,130-132,135,138,142,153,181,291,293,323,577,583-592) vary in sensitivities for detection of the primary tumor, as well as metastatic tumor (Table 9).

 

Table 9. Tumor Localization Results in Patients with ZES

 

NIH studies

Mean(range)

Literature

Mean(range)

Literature

Mean(range)

Extra-hepatic lesions

Ultrasound

13 (9-16)

24 (0-28)

92 (92-93)

MRI

40 (30-57)

22 (20-25)

100 (99-100)

CT scan

38 (31-51)

38 (0-59)

90 (83-100)

Angiography

43 (28-57)

68 (35-68)

89 (84-94)

SRS

69 (58-78)

72 (57-77)

86 (86-100)

PVS

71

68 (60-94)

ND

Intra-arterial Secretin test

86

89 (40-100)

ND

EUS

ND

70 (28-86)

85 (80-93)

IOUS

83

83 (75-100)

 

Liver Metastases

Ultrasound

46

40 (15-77)

100 (99-100)

MRI

71

63 (60-75)

92 (88-100)

CT scan

42

48 (37-56)

99 (99-100)

Angiography

65

62 (33-86)

98 96-100

SRS

92

97 (92-100)

95 (90-100)

Intra-art. Secretin test

40

ND

ND

Data are from (9,27,40,119,129,133,142,295,525,591,593).

ND-no data.

 

At present, most patients when initially evaluated have performed a cross-sectional imaging study (CT, MRI. Ultrasound) and an SRI study to determine whether surgical resection should be considered (6,9,44,88,181,182,584). Gastrinomas, similar to other pNEN, are hypervascular and are thus their detection on imaging studies can be enhanced by the of administration of contrast; hence, in most patients, either a triphasic CT with intravenous contrast or an MRI with intravenous contrast (gadolinium (129,291,576,577,583). With the cross-sectional imaging modalities, the detection of lesions is influenced by their (40,141,525,577,591). In patients with gastrinoma lesions <1 cm in diameter, only <10-20% are detected, with 1-3 cm in diameter it increases to 15-40%, and with tumor lesions >3 cm, >80-90% are detected (40,525,577,591). Therefore, cross-sectional imaging studies will miss most primary duodenal gastrinomas, which are characteristically <1 cm in diameter; however, they detect most pancreatic primaries which are frequently > 3 cm in diameter (43,45,109,110,175,176,577). As summarized in Table 9, the sensitivity of cross-sectional imaging for detection of primary gastrinomas varies markedly among different series, with generally excellent specificity. In general, they detect <50% of the primaries, with lower yields in series with a high percentage of duodenal gastrinomas.  For detection of a patient with liver metastases, cross-sectional CT/ultrasound identify approximately one-half the patients, whereas MRI detects nearly three-quarters (Table 9). 

 

Selective angiography was widely used in the past, but is infrequently used now, however it is a sensitive method to image gastrinomas, (27,43,241,525,591,593). In most studies angiography was more sensitive than cross-sectional imaging studies for localizing primary gastrinomas, but it still did not localized approximately half of all primary gastrinomas, particularly missing small duodenal gastrinomas (40,43,175,241,525,591) (Table 9). However, angiography is increasingly not used, because it is an invasive procedure, but more importantly because of the increasing sensitivity of both cross-sectional imaging, and the increased availability of SRI which has a significantly higher sensitivity of SRS (Table 9). In the past frequently at the time of angiography, selective hormonal sampling was also used, and still used today in some centers for patients with ZES  who have negative cross-sectional imaging and negative SRI studies (138,323,585,588,589,593-597)  Two different methods for gastrin hormonal sampling have been  used, with the first being the transhepatic catheterization of portal venous tributaries draining the pancreas (portal venous sampling (PVS)) (323,593,595)  and the second,  which is more frequently used, is the assessment of hepatic venous gastrin concentrations performed after secretin injection into selective arteries to various pancreatic/duodenal regions (585,588,589,593,594,596,597). This method is not dependent on tumor size and involves functional localization which can be very sensitive (Table 9), however, it is now rarely used being replaced by cross sectional imaging and SRI (588,589).

 

Greater than 90% of NENs/NTs including gastrinomas are well differentiated tumors which overexpress or ectopically express one of the subtypes of somatostatin receptors (sst1-5) in >90% of cases (primarily sst2) with the result that somatostatin receptor imaging (SRI) with various radiolabeled various somatostatin analogues is now widely used (90,134-137,139,141,153,291,292,582,598,599). This method is particularly sensitive method to identify both the primary and metastatic gastrinoma location (181,401,525,576,600-602). Of the five classes of somatostatin receptors (sst1-5), all can be detected in various gastrinomas; however, sst2(80-100%) and sst5 (30-60%) are the most often overexpressed (603). Whereas native somatostatin (som14) interacts with all 5 receptor subtypes with high affinity; it is rapidly degraded in the circulation, hence is not useful therapeutically or for radio-imaging studies (601,603). Two synthetic analogues of somatostatin, octreotide and lanreotide, which have high affinity only for sst2 and sst5, are metabolically stable, and are now widely used for both SRI, for their anti-tumor effects both alone or coupled to radiolabels that are cytotoxic to the tumor and will be discussed in the treatment section later (see PRRT) (90,129,292,601,603-609). Specifically in gastrinomas, in the NIH ZES prospective studies (Table 9), SRS using 111In-labeled somatostatin analogues (Octreoscan) and single photon emission computed tomographic scanning (SPECT) imaging detected primaries in 69% of patients and in one prospective study of 80 consecutive ZES patients (133), SRS was more sensitive than any single cross-sectional imaging study or angiography, and was equal in sensitivity to the combination of all three cross-sectional imaging studies (US, CT, MRI) and angiography together (58% vs 48%)(133).  The sensitivity of SRS, similar to cross-sectional imaging, is influence by the size of the gastrinoma, with SRS using 111In-labeled somatostatin analogues (Octreoscan) and SPECT imaging visualizing only 20% of gastrinomas <0.5 cm in diameter, 30-40% <1 cm in diameter (610).  Because the mean size of duodenal gastrinomas is <1 cm, SRS detects only 32% of duodenal gastrinomas (175,610). The use of 68Ga-labelled somatostatin analogues with positron emission tomography (PET-scanning) has greater resolution with increased sensitivity (129,134,141,290-292,582) and thus is an important recent advance. In the US and in many countries, the most commonly used ligand for SRI is now 68Gallium DOTA (9,4,7,10-tetraazacyclododecane-1,4,7,10-tetracetic acid) labeled somatostatin analogue (generally 68Ga-DOTATOC PET/CT) with positron-emission tomography detection (129,134,141,290,582). This SRI method has generally replaced the use of 111Indium (diethylenediamine penta-acetic-D-phenylalanine-1) octreotide with single photon emission CT (SPECT) detection, because of it greater sensitivity (129,134,290-292).  SRI at present is the most sensitive method for assessing whole body localization of advanced NENs (129,134,290,292).

 

SRI is of particularly valuable for detecting distant metastases both to the liver and more distant, especially to bone, with a detection rate of 97% for identifying a patient with metastatic disease in the liver (Table 9). Studies demonstrate that bone metastases are relatively common in patients with advanced NENs including gastrinomas, in which they occur in up to 31% of ZES patients with liver metastases (58,112,294,611,612). The detection of bone metastases in ZES patient has been shown to have a high clinical importance, because they may not only require specific treatment, they also have important prognostic significance) (109,294,299,612,613). In one prospective study from NIH (112), SRS had greater sensitivity than bone scans for detecting bone metastases, and for imaging metastases in the spine was equal in sensitivity to MRI (112). Because 15-25% of the initial metastases occur outside the axial skeleton, SRI is recommended as the initial study over MRI to detect bone metastases (112).

 

 Whereas endoscopic ultrasound (EUS) has proven to be one of the most sensitive modalities for detecting insulinomas/NF-pNEN and is reported to be sensitive for localizing gastrinomas in some studies, its use is controversial in gastrinomas (40,119,245,614-616). EUS detected a mean of 70% of gastrinomas in different studies (Table 9) and has the advantage of allowing histological verification of the presence of a NEN as well as obtaining samples for determining the grade of the NEN which is particularly important for prognosis (617,618). However, EUS’s result is operator-dependent and false positives can occur (119,133,245,615). An important issue in patients with ZES is EUS’s sensitivity for detecting gastrinomas in different locations such as the duodenum, which is the source of controversy in is use in ZES as opposed to its general use in patients with entirely intra-pancreatic NENs (insulinoma, NF-pNEN, etc.). In one review of EUS in ZES patients, EUS detected a pancreatic gastrinoma in 83%, whereas it detected a duodenal gastrinoma in only 43 % (119). This is a major problem for EUS in patients with ZES because in recent studies 3-10 times more gastrinomas are found duodenal than pancreatic (9,181,182). Because of this difference, many experts do not recommend EUS as a routine preoperative imaging study in patients with ZES, especially in the 75-85% of patients with sporadic ZES (119). As will be discussed further in a later section, serial EUS studies may be used in patients with MEN1/ZES to evaluate the possible growth of the pNETs in patients who do not undergo routine exploration (30,44,88,90,619,620). At present it is recommended that a cross-sectional imaging study and SRS with SPECT imaging be performed in all ZES patients to evaluate tumor location/extent (9,42,50,181,182). If negative, but where the diagnosis of ZES has otherwise been confirmed, MEN1/ZES is not present and surgery is being considered, there is not complete agreement on which if any localization procedure should be performed prior to surgery (45,119,254). This issue will be discussed further under the section on surgical management.

 

Recently, there has been increased interest in pNENs, including gastrinomas, as well as NENs in other locations of the use of 18F-Fluordeoxyglucose (18F-FDG) PET imaging particularly as a prognostic marker (42,90,115,116,129,134,291,621-623). 18F-FDG PET/CT assesses tumor metabolic activity by determining the glucose uptake and therefore measures a different tumor parameter than SRI which is assessing somatostatin receptor expression. Although 18F-FDG PET/CT is widely used in oncology, until recently, it was generally not thought helpful in patients with pNENs/NETs (134,624). However, numerous recent studies report high uptake by a proportion of NETs (291,625-627). In a number of studies, the high uptake/SUV of 18F-FDG PET/CT   was reported to be associated with higher Ki67 values and was a predictor of overall survival as well as PFS (42,115,116,134,291,621-623). Lately there have been an increasing number of papers advocating either the use of FDG either alone or combined in dual imaging with 68Ga-DOTA-SSA PET/CT (42,622,623,625-631). Similar to its increasing use in other pNENs/NENs the use of 18F-FDG PET/CT in patients with gastrinomas may help in identifying those with aggressive disease, particular as a postoperative tool to stratify patients that may benefit by more aggressive postoperative treatments.

 

TREATMENT (ACID SECRETION/LOCALIZED DISEASE)

 

General Aspects (Not Advanced Metastatic Disease)

 

Like patients with other F-NEN syndrome, patients with ZES have two different aspects that require treatment and often can’t be controlled by a single treatment strategy (25,48,54,158,632-634): control of the hormone excess state and treatment directed at the NEN per se, because, except for insulinomas, but similar for gastrinomas and the other F-NEN, these NENs are malignant in 50-100%  of cases  and require treatment. Specifically, in the case of ZES treatment needs to be directed at two different problems: the control of the marked acid hypersecretion and the gastrinoma itself. Whereas a curative resection would solve both problems; unfortunately, it is possible in <30% of patients. Furthermore, in patients with MEN1/ZES which comprise 20-25%, treatment must be also directed at the other endocrinopathies these patients frequently develop, as well as genetic family counseling (30,65,96,99,100,203,463,505). The first section will discuss management of the acid hypersecretion, followed by the surgical management of the gastrinoma in patients without advanced metastatic disease. In the last section of treatment, the management of patients with advanced/metastatic disease will be discussed. 

 

Management of Gastric Hypersecretion  

 

GENERAL MANAGEMENT OF GASTRIC HYPERSECRETION

 

Numerous studies, especially older studies prior to adequate drug therapy to control the gastric acid hypersecretion in ZES patients, demonstrate that both the acute and long-term control of the acid hypersecretion is essential for long-term survival (9,46,72,89,103,181,184,205,313,635-637). Prior to the availability of effective acid antisecretory drugs, most ZES patients who did not have a total gastrectomy, eventually developed complications of the gastric acid hypersecretion, and the majority died from these complications rather than from tumor progression (1,27,28,46,72,89,103,184,205,235,313,635,638). This occurred largely because of the direct effect of the marked acid hypersecretion, with the mean basal-acid output (BAO) in ZES patients typically 4-times normal but reaching as high as 12-times the upper limit of normal in some patients (40,70). In a given patient it is not possible to predict when these elevated acid levels will overcome the defense mechanism (increased bicarbonate secretion, increased duodenal secretion, etc.), thus in all patients it is essential to acutely control the acid hypersecretion as soon as ZES is suspected and as the initial step in management (1,9,28,51,56,72,181,182,313,558,639).

 

SURGICAL TREATMENT OF GASTRIC HYPERSECRETION

 

While surgical management of the gastric acid hypersecretion in ZES patients is now rarely used, in the past (prior to the 1970’ s), the only effective means of adequately controlling gastric acid hypersecretion in these patients was by total gastrectomy (1,28,205,207,237,620,638,640-642). Lesser operations were almost invariably inadequate to prevent recurrence long-term (1,28,237,403,620,638,641). Because in ZES patients prior to any other means of controlling the acid hypersecretion, the total gastrectomy was often performed as an emergency procedure and was associated with considerable morbidity/mortality (1,40,205,638). However, later with the availability of histamine H2receptor antagonists, starting in the 1970’s, allowing preoperatively control of the acid hypersecretion medically in most patients, the total gastrectomy could then be performed electively and was relatively safe, with an overall mortality of 5.8% in 248 cases since 1980, and 2.4% for elective cases (207).  However, the long-term morbidity remained unclear, and in some studies up to 50% of patients have moderate or severe side-effects, including weight loss, pain, stenosis of the anastomoses, vomiting and early satiety (27,40,643). At present, because of the effectiveness of medical therapy especially the PPIs, total gastrectomy is rarely performed and reserved for patients (<0.2%) (9,27,51,71,103,108,644,645) who cannot or will not regularly take oral antisecretory drugs.

 

Both vagotomy, as well as medical treatment with anticholinergic agents, can reduce the levels of gastric acid hypersecretion in ZES patients and also, they can potentiate the effectiveness of histamine H2R antagonists when added (27,40,646-648). After the availability of histamine H2R antagonists (1970+), but prior to the availability of PPIs(mid-1980s), most ZES patients were not cured at surgery, and because many continued to require frequent histamine H2R antagonists, it was proposed that parietal cell vagotomy, be performed at the time of surgery in ZES patients (211).  In ZES patients that underwent selective vagotomy, (211,648), the BAO decreased by a mean of 50%, the histamine H2R antagonist dosage could be reduced by 40%, and in 36% of patients all antisecretory drugs could be stopped postoperatively. Today, with the development and availability of PPIs, which are highly effective in ZES patients, a form of vagotomy is rarely necessary or used.

 

In patients with MEN1/ZES with hyperparathyroidism, an effective parathyroidectomy can markedly reduce fasting gastrin levels (FSG), the BAO and can increase the sensitivity to gastric antisecretory drugs (190,508,509,533), with a mean decrease in BAO of 56% and the FSG of 55% (40,190,508,509).  Moreover, in some patients, the FSG levels can decrease to the normal range, as well as a positive secretin-test can become negative (40,190,508,509). MEN1 patients, with or without ZES, have parathyroid hyperplasia which involves all four parathyroid glands, if recurrent hyperparathyroidism is to be avoided post-parathyroidectomy, it is recommend that either a 3.5 parathyroid gland resection or a 4-gland resection, with a parathyroid implant, should be performed in these patients (463,508,509,649-653).

 

Long-term, curative gastrinoma resection is possible in < 40% of patients with sporadic ZES undergoing surgery with the recommended surgical approaches in most guidelines with no-aggressive resections (non-Whipple resection) (6,43,175,254); and even when curative, it does not completely correct the gastric acid hypersecretion in some patients (654-656). In the NIH prospective studies of acid hypersecretion post-curative resection, the MAO decreased 50%, BAO decreased 75% within 6-12 mos. and then remained unchanged for up to 4 years, and the histamine H2R antagonists’ dose could be reduced by >60% (654-656).  However, even though the BAO decreased by 75% after curative resection for up to 4 years, 60% of the patients remained acid hypersecretors (654,655). This group included 34% who were mild hypersecretors (BAO-15-24.9 mEq/hr.) and 28% who had marked to extreme hypersecretion (≥25 mEq/hr. (range-25-69 mEq/hr.)) (655). The mechanism of this continued hypersecretion post-curative resection is unclear (655). Practically, it means that all ZES patients should continue to be followed carefully post-curative resection and many will continue to need low doses of antisecretory drugs (655).

 

MEDICAL TREATMENT OF GASTRIC HYPERSECRETION

 

In all recent guidelines, medical treatment with oral gastric acid antisecretory drugs is the recommended method to control the gastric acid hypersecretion seen in ZES patients, both acutely and long-term (9,29,103,157,164,180,181). PPIs (omeprazole, esomeprazole, lansoprazole, pantoprazole, rabeprazole) are the recommended drug of choice because of their long durations of action and potency (7,9,25,29,40,72,103,157,164,180-182,657,658). Most ZES patients without complicated disease (MEN1/ZES, moderate-severe GERD, post-Billroth II surgery) require only once a day dosing and many are controlled on PPI doses equivalent to those used in idiopathic PUD disease (i.e., equivalent to 20 mg/day omeprazole) (103,184,518,519,557). In patients with complicated disease (MEN1/ZES (especially with active hyperparathyroidism), moderate-severe GERD, post-Billroth II surgery) higher doses/frequency are usually needed (72,184,518,519,557,659). For patients requiring higher doses, in general, increasing the dose frequency is more effective than increasing the dosage once-per-day (72,518,519).  Most long-term studies were performed with omeprazole or lansoprazole as the PPI, however; other PPIs (pantoprazole, rabeprazole, esomeprazole) are effective in ZES, and it is not apparent anyone has an advantage over the others (103,184,660-662). There is no complete agreement on the starting dose of PPI to be recommended. This becomes an important point in patients with ZES because many of the PPI formulations are acid-labile and thus starting a patient on a low PPI-dose could delay its action, and in acutely ill ZES-patients with PUD this could result in complications (663). One study attempted to address this question (663) by starting patients with ZES on a low dose of omeprazole (20 mg/day) and found that in 32% acid secretion was not controlled and higher omeprazole doses were needed. This study proposed that ZES patients with uncomplicated ZES (no MEN1/ZES, moderate-severe-GERD, post-Billroth II surgery) be started on higher PPI doses (equivalent to omeprazole 60 mg/day) and then doses reduced during follow-up. Both the US NANETs guidelines (182) and the European ENETs guidelines (181) recommend that ZES patients with uncomplicated disease (no-MEN1/ZES, moderate-severe GERD, post-Billroth II surgery) be started on the equivalent of 60 mg/day of omeprazole and that patients with complicated disease be started on PPI doses equivalent to omeprazole 40-60 mg BID and then, with time, dose reduction be attempted. It is ideal to titrate the PPI dose to control the acid output (<10 mEq/hr for no gastric surgery, <5 mEq/hr for previous gastric surgery) (27,40,72,103,106,184,503,660), but few physicians now have access to units measuring gastric acid output. Symptom control (particularly diarrhea, pain, heartburn) can be used to guide management, and if mucosal disease is present, repeat UGI endoscopy should be performed after 6-8 weeks. Because of their potency, dose titration is less important with PPIs; however, it is essential with histamine H2R antagonists (see comments below in this section).

 

Only a few studies have reported the long-term results of continuous treatment with PPIs in ZES patients for 9-15 years (72,76,518,557,661). Tachyphylaxis does not develop with long-term PPI treatment in ZES patients, and on average <20% of patients require a PPI-dose increase/year (rate-0.13/patient), whereas with long-term histamine H2R antagonist treatment, an average of at least one dose increase/year was required (27,72,103,104,518,558-561). Long-term PPI use has proven safe; with fewer than 0.1% of patients stopping treatment because of a side-effect (103). A potential concern of long-term PPI-treatment is the drug-induced hypo-/achlorhydria, which may lead to effects on nutrient absorption (vitamin B12, iron, calcium) as well as enhanced hypergastrinemia resulting in an increased risk of gastric carcinoid tumors (76,502,664-669). Low vitamin B12 levels are frequent in ZES patients (666,667,670,671), are more frequent in ZES patients treated with PPIs, and correlate with the PPI-induced hypo/achlorhydria (670). While the PPI induced decrease in serum VB12 levels in ZES patients in the above study (670) was established in a prospective study of these patients, the question of whether PPIs systematically decrease VB12 levels in the nonZES, general population and thus should be monitored for, remains controversial (76,667,672).

 

In another study of ZES patients (673), deficiencies in body iron stores were not found with long-term PPI treatment. Recently, epidemiological, and various correlative studies report in the general population that long-term PPI use may result in an increased incidence of bone fractures, particularly in the spine/ hip, but there are no specific studies in ZES patients (76,666,667,674). In addition, in similar correlative studies in the general population a number of other possible side effects of long-term PPI treatment have been proposed: these are controversial and except for malabsorption of vitamin B12 have not been reported with increased occurrence in ZES patients (76,667). The proposed PPI-side-effects include an increased occurrence of such diverse problems as:  dementia, chronic renal disease, hypomagnesemia, malabsorption of various nutrients (vitamin B12, iron, etc.), well as increased growth of various other cancers including gastric, pancreatic, and colorectal tumors (76,502,667,668,675,676). Hypomagnesemia has been rarely reported (3 case reports) in ZES patients (76,645,672,677) and in the prospective NIH studies involving 250 ZES patients, only a single patient developed hypomagnesemia despite chronic, continuous PPI with many patients taking higher PPI doses and with a mean treatment time >10 years, for rate of 0.4% over the treatment period (76). On the basis of these studies, it has been proposed (76,181,673) that only the serum vitamin B12-levels should be periodically assessed once a year in ZES patients with long-term PPI treatment, especially the group of patients who might have low vitamin B12 level initially or a poorer nutritional status (elderly patients with a long history of malabsorption).  

 

Recently, there have been an increasing number of reports of medical failure in ZES patients of the long-term use of H2R (27,558,562,586,678) for maintenance acid control, and also even problems controlling acid with PPI therapy long-term (71,71,95,102,108,136,285,403,494,644,645,672,679-686). This is occurring in large part due to the lack of data from any extended long-term/ lifetime treatment studies (i.e., >10 yrs.-lifetime) of antisecretory acid control in ZES patients. This is in contrast to a number of studies of acute acid control and short-term (<5-6 yrs.)  control with small number of ZES patients (225,518,519,554,561,661,662,677,687-693). This lack of information about the long-term efficacy of acid antisecretory drug’s in ZES is a particular problem because of the unique acid secretory condition in ZES. In ZES there is a constant hypersecretory drive due to constant ectopic secretion of gastrinoma from the gastrinoma, resulting in a constant acid hypersecretory state, which results in a constant requirement to inhibit the acid secretion, which because it is unique to ZES, its treatment can only be addressed by long-term/lifetime study data in these patients.  In contrast to ZES, there are numerous long-term PPI studies in nonZES patients, particularly in patients with advanced idiopathic GERD, and these can provide evidence for safety issues that might occur with lifetime PPI treatment (76,667,694-696), which is applicable to chronic treatment of ZES patients, however, this is not the case with long-term/lifetime efficacy data in ZES. Another important variable contributing to the need to have data on long-term/lifelong antisecretory efficacy in ZES patients, occurs because of the marked variation of the dose requirement between individual ZES patients as well as in each patient, which has been well-examined in short-term ZES acid secretory studies (225,518,519,554,561,661,662,677,687-689,697). This issue was recently addressed (72)in an analysis of the results of acid antisecretory treatment in ZES patients, which examined in detailed the efficacy/pharmacology of long-term/lifetime medical treatment of acid hypersecretion in a large cohort of ZES patients. This study included results from all 303 patients with established ZES who were prospectively followed and had acid antisecretory treatment with either H2Rs or PPIs who had antisecretory doses individually titrated by the results of regular gastric acid testing. It includes both patients treated for short-term periods (<5 years), as well as patients treated long-term (>5 yrs.), and with lifetime treatment (30%), followed for up to 48 yrs. (mean-14 yrs.). Long-term/lifelong acid antisecretory treatment with H2Rs/PPIs could be successfully carried out in all patients with both uncomplicated and complicated ZES (i.e., with MEN1/ZES, previous Billroth 2, severe GERD). Successful treatment in this study was only possible because the drug doses were individually set by assessing acid secretory control by measuring the acid secretory rate and adjusting the various drug doses to establish proven criteria, with regular reassessments and readjustments. Frequent dose changes both up and down were needed; as well as regulation of dose-frequency and a primary reliance on the use of PPs. In this study (72) prognostic factors predicting patients who required PPI dose-changes were identified which need to be studied prospectively to develop a useful predictive algorithm which could be clinically useful for tailored long-term/lifetime therapy in these patients. These results clearly establish that long-term/lifelong medical control of the acid hypersecretion is possible in all ZES patients who can take acid antisecretory drugs, but requires it be performed in centers with the capability of titrating the drug dose over time by assessing the acid secretory rate, thus it is best if these patients are referred to centers with this capability.

 

Chronic hypergastrinemia in animals and man stimulates gastric enterochromaffin-like (ECL) cell proliferation and in animal models, gastric carcinoid tumors (ECLomas) can develop, some of which are malignant (76,217,224,227,502,665,666,698-701). In patients with ZES, ECL cell proliferative changes develop in >90% (76,217,227). However, patients with sporadic ZES (no MEN1) (75-80%), rarely develop gastric carcinoids (76,103,217,502), whereas MEN1/ZES patients have >70 greater risk of developing a gastric carcinoid (227). In one prospective study (227), 23% of MEN1/ZES patients had gastric carcinoids and other studies have indicated that these can be malignant in 10-30% of patients (76,227,502,666,702,703). In a similar prospective study of 106 patients (217) with sporadic ZES, none of the patients had a gastric carcinoid tumor, although 99% had ECL cell hyperplasia, and 50% had advanced ECL cell proliferative changes, including 7% with dysplasia. Even though there are a few case reports of gastric carcinoids found in sporadic ZES patients (76,212,228,229,232,233,704-709), the prospective NIH study discussed above(217), demonstrates that this is very uncommon, and differs markedly from the chronic atrophic gastritis patients in which 0.4-7% have gastric carcinoids on a routine endoscopy, and 5-35% in some series with long-term follow-up (76,710,711). There is no evidence the long-term use of PPIs accelerates gastric carcinoids development either in patients with sporadic ZES or with MEN1/ZES (76,103,502). However, because of the association of hypergastrinemia with gastric carcinoids, all patients with ZES should undergo an initial upper gastrointestinal endoscopy; those with MEN1/ZES should have a repeat UGI endoscopy yearly, while in those with sporadic ZES, if there are no upper GI symptoms, follow-up UGI endoscopy can be less frequent.

 

During the subsequent clinical course of many ZES patients after diagnosis, for their frequently occurs brief periods where they cannot take the oral antisecretory drugs (e.g., after surgery, chemotherapy, etc.)  and during this period a parenterally administered gastric antisecretory drug may be necessary. Parental histamine H2R antagonists can be used, however, continuous infusions of high doses are required (27,103,105,586,712,713). In contrast, with parenteral PPIs (omeprazole, esomeprazole, lansoprazole, pantoprazole, etc.), because of their long durations of action, intermittent parenteral administration (every 6-12 hours) can be used (103,555,714-716).

 

At present histamine H2R antagonists are much less frequently used than in the past (76). Although histamine H2R antagonists can be effective if properly administered, they usually have to be taken every 4-6 hours, and the oral dose needs to be titrated so that acid hypersecretion one hour prior to the next dose is decreased to <10 mEq/hr (no previous-gastric-surgery, <5 mEq/hr.-previous gastric-acid surgery) (28,103-105,558,560,562,717). In most patients at this level of control, symptoms will be controlled, and mucosal lesions heal (27,103,105,106,586). For patients with complicated ZES (MEN1/ZES, moderate-severe GERD, previous Billroth II surgery), acid hypersecretion may have to be reduced to <1 mEq/hr in order to achieve complete healing (27,105,503,519,659). Using dose-titration, the average daily doses needed of oral histamine H2R antagonists in the prospective NIH studies were 4.9, 2.2 and 0.33 g/day for cimetidine, ranitidine, and famotidine, respectively (40,103). Despite these high doses, the drugs were generally free of dose related side effects, except for anti-androgen effects with cimetidine (gynecomastia, impotence) and were effective long-term, although approximately one dose-increase/ year was needed (27,40,103,104,558,561,718). Because of this need to titrate the histamine H2R antagonist dose for each patient, the need for frequent, high dosing and the need to adjust of dosage with time,  PPIs (omeprazole, esomeprazole, lansoprazole, pantoprazole, rabeprazole) have now largely replaced the use of histamine H2R antagonists,  and are currently the recommended drugs of choice, because of their long durations of action and potency (7,9,29,40,76,103,157,180-182,657,719). Most ZES patients without complicated disease (MEN1/ZES, moderate-severe GERD, post-Billroth II surgery) require only once a day dosing and many are controlled on PPI doses equivalent to those used in idiopathic PUD disease (i.e., equivalent to 20 mg/day omeprazole) (103,184,518,519,557). In patients with complicated disease (MEN1/ZES (especially with active hyperparathyroidism), moderate-severe GERD, post-Billroth II surgery) higher doses/frequency are usually needed (184,518,519,557).

 

Currently, somatostatin analogues are uncommonly used to control acid hypersecretion in ZES patient, because they must be given parenterally, whereas effective inexpensive long-acting, oral antisecretory agents such as PPIs are available and are the drugs of choice (29,103,142,151,181,182,396).

 

SURGICAL TREATMENT (NOT FOR ADVANCED METASTATIC DISEASE)

 

At presents most authorities, as well as all guidelines, agree that surgical resection for attempted cure should be performed in ZES patients whenever possible without undue risk, similar to the treatment of other potentially resectable pNENs (9,43,46,95,99,102,119,120,122,157,180,182,267,461,720-728). This approach is, in contrast to that in the recent past, wherein the role of routine surgery for cure was controversial, with some recommending that surgery not routinely be performed, because gastrinomas were frequently not found at surgery and cure was uncommon (729-731). In addition, many patients had negative preoperative imaging, and most patients with non-imaged or small gastrinomas had a good prognosis without surgery (729,731). The situation has changed because of results from a number of more systematic studies. In a NIH prospective surgical study (43) of sporadic ZES patients (n=123), the immediate postoperative cure rate was 51% and after 10-years was 34%. A number of other NIH surgical studies (45,175,176,241,254,258,732,733), as well as studies from other institutions (46,95,99,102,122,177,724) have provided additional support for routine surgery. This approach is further supported by two NIH studies on survival/disease course post-surgical resection of the primary gastrinoma, with the one study (732) demonstrating that patients who underwent routine exploration had a lower incidence of developing liver metastases post-resection (3% vs 23%, p<0.003). A subsequent NIH study (733) with more patients (n=160) and a longer follow-up (mean 12 yrs. postresection) demonstrated that patients undergoing surgery had a better overall survival (15 yrs., 98% vs 74%, p=0.0002); the survival advantage was disease-related (p=0.0012), due to less tumor progression, and fewer patients developed liver metastases (5% vs 29%, p=0.0002). This is a particularly important finding, because two NIH studies (109,110) in patients with gastrinomas, as well as a number of other studies both in patients with gastrinomas and other pNEN (115,116,235,276,433,639,734), have demonstrated that the development/presence of liver metastases is one of the most important prognostic markers of long-term survival in these patients. Neither of above NIH studies were randomized, but in each case the comparative groups were well matched (109,110,242,733).

 

 In the past, imaging studies were not infrequently all negative on preoperative studies, and because these ZES patients had an excellent prognosis without surgery, and because surgery was often negative in these patients, this led a number of investigators to recommend against surgical exploration in this group (45,515,729-731,735-737). A subsequent NIH study (45) provided important information to challenge this approach by reporting the value of surgery in patients with preoperative negative imaging in an expert treatment center. In this study (45), in 58 ZES patients with negative preoperative imaging (40%=negative SRS), at surgical exploration, a gastrinoma was found in almost every patient (98%), and nearly 50% were cured.  The postoperative cure rate was not different from ZES patients with positive preoperative imaging studies treated in a similar manner (45). This study demonstrated that if the diagnosis of ZES is appropriately established, that an experienced surgeon can find gastrinoma in almost every patient, even if imaging studies and negative, and almost one-half will be cured (45). This improvement in the surgical success of finding and curing gastrinomas in sporadic ZES patients has occurred because of a number of factors: particularly important is the appreciation that the majority of gastrinomas are not in the pancreas, as previously thought, and are, in fact, small duodenal tumors (often <1 cm) (6,109,110,175,177,236,286,738), which are frequently missed on even the most sensitive pre-operative imaging studies, including SRI (293); which will be missed at standard surgical operations if special duodenal gastrinoma localization procedures are not used, such as duodenotomy with or without duodenal-transillumination (175,176,244,247); the use of improved imaging including SRI (549);  at surgical exploration the  routine resection of pancreatic head area lymph nodes  because of the possibility of lymph node primaries (258,259,268,269,269-272,739); and an understanding that patients with sporadic ZES have a different surgical outcome than those with MEN1/ZES (9,30,43,179,723).

 

The standard operation includes besides a careful inspection of the duodenum, pancreas and general abdominal inspection, a Kocher maneuver to explore the pancreatic/head; a duodenotomy  with or without duodenal transillumination; routine resection of pancreatic/duodenal lymph nodes; careful inspection of biliary tract and liver, and  an  intra-operative ultrasound(IOUS) examination of the pancreas (43,95,120,175,176,247,249,258,288,738,740-743). This detailed examination is based on the fact that the relative order of occurrence of gastrinomas is duodenum>>pancreas>lymph node primary>primary liver/biliary tract> other (ovary, mesentery, gastric, etc.) (27,40,50,744). The most important procedure is a careful inspection of the duodenum. This requires the performance of a duodenotomy which is characteristically a 3-cm longitudinal duodenotomy centered on the anterolateral surface of the descending part (second portion) of the duodenum accompanied in the NIH protocol with transillumination of the duodenum (175,176,244,247). A duodenotomy is required to carefully inspect the duodenum because intraoperative ultrasound (IOUS) has been found to be relatively insensitive for duodenal wall tumors in patients with ZES (740). The use of a duodenotomy was proposed by Norman Thompson, University of Michigan in 1989 (246) at a time when most physicians though gastrinomas were primarily intrapancreatic, similar to insulinomas and its benefits and risks for detecting occult duodenal gastrinomas was debated (175,175,246,247,721). Its routine use was firmly established by a prospective study at NIH involving 35 patients with ZES in which all patients first had the standard exploration for a duodenal tumor involving careful palpation without a duodenotomy/or intraoperative transillumination of duodenum, followed IOUS, then duodenal transillumination and finally a duodenotomy (244).  Standard palpation identified only 61% of all duodenal tumors found by any method, IOUS found only 26% and no new lesions; transillumination identified 64% of all duodenal tumors and 6 of these were new tumors, whereas duodenotomy identified all 31 duodenal tumors of which 5 were not identified by any other method.

 

This result was corroborated by another NIH study (176) which compared the surgical results from 36 patients (Group 1) who underwent the standard laparotomy (1980-1986) without duodenotomy (prior to its routine use) to a group receiving the same operation but with transillumination and a duodenotomy (19987-1990-37 patients) (Group 2). Gastrinomas were found in significantly more patients in Group 2(92% vs 64%, p<0.01); this increase was due to more duodenal gastrinomas detected in Group 2 (43% vs 11%, p<0.01) which resulted in an increased disease-free rate in group 2 (176). Most importantly, a NIH 2004 study (175) examined the long-term effects of adding a duodenotomy on the long-term cure rate. This study (176) compared results in 143 ZES patients, of which all had the standard exploration protocol, but 79 had a duodenotomy and the others had not had one. Gastrinomas were found in a higher percentage of patients who had underwent a duodenotomy (98 vs 76%, p<0.000011); as were duodenal gastrinomas (62 vs 18%, p<0.00001), whereas the detection rate of pancreatic tumors was similar; the duodenotomy group had a postoperative cure rate that was higher (62 vs 44%, p=0.010) as well as the long-term cure rate (52 vs 26%, p=0.0012). These results have established the need for all patients to have duodenotomy at exploration (9,157,181,182,745).

 

The routine resection of peri-duodena/peripancreatic lymph nodes is recommend for two reasons. First, as discussed earlier, a number of different groups have reported lymph node primary gastrinomas (27,40,43,258,259,266-274)which in the NIH series (258) was the third largest primary tumor group after pancreaticoduodenal tumors, comprising 10 % and their resection resulted in a disease-free state. Secondly, lymphadenopathy is reported to increase the disease-free rate postresection in ZES patients, as well as to increase overall survival in patients with sporadic ZES (746).

 

In contrast to the situation with sporadic ZES (no MEN1), the surgical management of MEN1/ZES patients remains controversial (9,30,87,92,93,95,95,96,99,120,122,123,128,157,181,182,745,747). This has occurred because almost all studies demonstrate that these patients are rarely cured by the standard ZES operation involving local tumor resection/enucleation even with a duodenotomy, and that cure only occurs if a Whipple resection is performed, which is not routinely recommended (9,43,44,88,89,92,122,181,182,747,748). Even though pancreaticoduodenectomy (Whipple resection) will cure the ZES in MEN1 patients (30,92,119,747,748), it is not routinely or generally recommended by most groups or in most guidelines in patients with MEN1/ZES, primarily because of the long-term potential complications (119,748,749). Also, in patients with NF-pNEN, because of the multiplicity of small adenomas, a total pancreatectomy would be required, which because of its morbidity, is not recommended (30,750). This low cure rate with nonaggressive resections occurs because MEN1/ZES patients almost invariably have multiple, duodenal gastrinomas which are microscopic to small in size (many <0.5 cm) and thus difficult to find at surgery, as well as >50% have metastatic lymph nodes at surgery (43,95,119,178,179,191,284). On preoperative imaging studies in MEN1/ZES patients, duodenal-pancreatic NENs are frequently visualized, however, the peripancreatic tumors are often not the primary but an adjacent positive metastatic lymph node, whereas the pancreatic NENs frequently visualized are usually not gastrinomas (0-<15%) (mostly-nonfunctional-pNEN) (88,191,284). Numerous studies report that if the preoperative imaging studies identify a tumor <1.5-2 cm in diameter, that these patients have an excellent long-term prognosis; in fact, survival is not different from MEN1 patients without a pNEN seen in some studies (9,27,191,751).

 

A number of other points complicate the decision for surgery and the management of the pancreatic-duodenal lesions in MEN1 patients and have particularly importance in recommending a more conserve approach than aggressive surgical resection in MEN1/ZES patients. First, pNEN present approximately 10-years earlier in MEN1 than sporadic cases (30,87), even occasionally occurring in patients < 20 years old (101,752).  This has led to added controversy on whether such young patients should undergo surgery or have continued surveillance. Second, MEN1 patients have an increased incidence of glucose intolerance and diabetes (753,754). This could become an important consideration, particularly in younger patients if they underwent extensive pancreatic resections such as Whipple resection, because the occurrence of glucose intolerance/diabetes after such procedures is reported in different series as, 10% (749), 34 % (755),40% (102) and 86%(756) if MEN1 patients underwent a major pancreatic resection and in 8-27 % of any patients undergoing pancreaticoduodenectomy (102,757,758). Furthermore, pancreatic insufficiency develops in 41-50% after major resections or un 40% after Whipple resections (102) which can complicate the post-surgical clinical management.  Third, is the potential importance of continued radiation exposure in MEN1 patients who require life-long monitoring (90). This could become an important issue if these patients are followed, and continued imaging surveillance is required. Some recommend endoscopic ultrasound (EUS) for this purpose (90), which is the most sensitive modality, however, it is an invasive procedure which is done under general anesthesia in many centers, and therefore other imaging modalities that allow serial assessment of changes in pNEN size, would be of value, such as repeated cross-sectional imaging studies (MRI, CT scanning), however they are less sensitive. These cross-sectional imaging modalities (CT, MRI) very frequently miss small pNEN<1.5-2 cm in diameter, a group that numerous studies shows do not have an increased mortality from pNEN (181,505,751,759,760). For the above reasons, there has been increased interest in MEN1 patients, especially younger patients, in imaging studies not involving radiation such as MRI, but because MRI does not detect a significant number of small pNEN in MEN1 patients, there also is increased interest in more sensitive imaging studies such as 68Ga-DOTATOC positron emission tomographic/CT imaging (68Ga-DOTATOC-PET/CT) which involve radiation. This interest has especially increased with recent studies reporting for the first time prospective (549,761,762) and non-prospective studies (763,764) demonstrating enhanced sensitivity/specificity for localizing NETs, including pNEN, in MEN1 patients, using 68Ga-DOTATOC-PET/CT.  Lifetime exposure to radiation may be a particular issue in MEN1 patients because basic science studies demonstrate that menin, the protein altered in patients with MEN1, is involved in DNA repair, cell cycle control and transcriptional regulation, and when there is a loss of menin activity, as occurs in MEN1 patients, cells become more sensitive to the effects of ionizing radiation as well as other cell damaging injuries (765-767). As a result, a number of studies have raised concerns about the use of imaging studies involving radiation in younger patients (without MEN1) (768-770), and whether younger MEN1 patients are at increased risk is unclear. These points raise controversies about when and how frequent these serial imaging studies should be used.

 

For these reasons, most current guidelines, and expert opinions (9,44,88,157,180-182) for the treatment of pNEN in MEN1 patients recommend that MEN1/ZES patients with preoperative imaging studies demonstrating pNETs <1.5-2 cm in diameter not undergo routine surgical exploration.  These guidelines also recommend that when surgical exploration is performed that Whipple resections not be routinely performed.

 

There are however, increasing concerns raised by the number of recent studies with this general conservative approach. Important points being raised is that these patients have a markedly shortened life- expectancy (i.e., 55 yrs. in the large prospective NIH review (89) with the major cause of death being malignant NENs, although presumed pNEN in origin it is not proven at this point (89). A second major point reviewed above is the enhanced ability to localize the primary NENs and their extent preoperative with the availability of SLI, allowing an enhanced ability to plan the operation and extend of surgery needed and likely enhancing the probability of cure. A third major point is that there have been a number of series (95,99,102) from different institutions reporting high cure rates and excellent long term survival in these patients after Whipple resections (95,99,102,119,178,191,683,747,748,771-801) . Furthermore, in a number of these studies the rate of post operative diabetes is less than previous reported in some studies and even not higher than seen with the recommended more conservative resections (273).

 

Increasingly, both patients with sporadic ZES, as well as those with MEN1/ZES, who had undergone an initial surgical resection, are being reoperated with time for either a recurrence after being initial rendered disease-free or due to increasing tumor growth with persistent disease (92,127,254,747,802-805). In a recent prospective NIH study of 52 ZES patients (254) with recurrence who underwent reoperation, the reoperation occurred a mean of 6 years after the initial surgery. After the reoperation, 35% were disease-free immediately postoperative and on the last follow-up after the repeat surgery (mean-8 years), 25% remained disease free, which are lower percentages than seen with the initial operation in NIH studies (43,45,175,254,805).  In this study (254), the 20-year survival was 84% and the presence or absence of MEN1/ZES did not affect survival, but the length of the disease-free interval postresection and presence of liver metastases did.  A recent study (127) reported recurrence in 108 sporadic ZES patients who had underwent an initial elective surgery between 2000-2020 in 15 different European hospitals. In these patients (127) 68 had duodenal gastrinomas, 19 (18%) had pancreatic gastrinomas, and 21 (19%) had a primary lymph node gastrinoma in the original surgery. During the initial surgery 74% of the patients with duodenal gastrinomas had a pancreaticoduodenectomy (Whipple Resection). For all gastrinoma patients (127) their mean OS was 173 mos., 5-yr survival 94%, and no predictive factors were found. The median DF-survival was 93 mos., and the 5 yr DF survival rate was 63%. For recurrence, significant prognostic factors were tumor size> 2 cm, (P=0.00001), tumor grade (p=0.00001) and pancreatic gastrinoma location (p=0.0001), however on multivariate analysis only tumor size >2 cm (p=0.005) and grade (p-0.013) were significant.  Specifically, not a significant prognostic factor was age, sex, preoperative gastrin level, lymphadenopathy <10 nodes or metastatic lymph nodes in resected nodes.  Also, for duodenal gastrinoma the recurrence rate was similar in patients with a Whipple operation to that in patients with excisions of duodenal tumors and lymphadenectomy (127).

 

A recent study also reported the results of duodenopancreatic reoperations in patients with MEN1(12 patients), of whom 5 patients (42%) had MEN1/ZES (92,747). In this study (92,747) the mean time to reoperation was 5.5 yrs., and with a long-term mean follow-up of 18 years, 83% (10/12) remained alive.  The authors (92,747) concluded reoperations in this group of patients are not uncommon, there is no increased perioperative morbidity with reoperation in a specialty center, the patients can have prolonged survival after reoperation and that organ-sparing resections are preferred in these patients. 

 

TREATMENT OF ADVANCED METASTATIC DISEASE

 

General Points

 

With the increased ability to medically control the gastric acid hypersecretory state in ZES patients, the natural history/growth of the gastrinoma is becoming the major determinant of long-term survival in ZES patients (46,89,109,110,314,433). Natural history studies show that gastrinomas are malignant in 60-90% of patients, and at present, approximately one-third of ZES patients present with metastatic disease to the liver, and because most patients are not cured surgically, an increasing proportion develop advanced metastatic disease over time (40,46,89,109,110,254,314,433,639). Overall, in NIH prospective studies, 25% of patients with sporadic ZES (109,110) and 15% of MEN1/ZES patients (111) have tumors showing an aggressive growth pattern, and in 40% of patients with hepatic metastases, aggressive growth occurs (429). As a result, currently one-half of ZES patients have tumor-related deaths (109).

 

A number of clinical, laboratory, pathological and other tumoral features in ZES patients are associated with a poor prognosis and are summarized in Table 10. A number of studies report one of the most important prognostic factors is the presence of any liver metastases (initially or their development) (Table 10). For example, in the NIH studies, the 10-year survival of ZES patients with no liver metastases initially is 96%, with liver metastases limited to one hepatic lobe is 78%, and with diffuse liver metastases is 16% (109,110). If liver metastases develop for the first time during the follow-up period after an initial evaluation wherein no liver metastases were present, the ten-year survival is decreased to 85% (110). However, in different studies at different times in the NIH cohort of ZES patients, the presence of lymph node metastases alone was, at best, only a weak predictor of poor prognosis, and in fact, was not predictive in a number of early studies (Table 10) (109,110,314,806). In a detailed analysis (806) of 216 pNEN patients at NIH in which >90% were ZES, with a prolonged follow-up (mean 11 years), overall survival decreased not only in patients with any lymph node positive, but also the extent of decrease in survival correlated with the number of positive lymph nodes. This result is consistent with some general studies in patients with various pNEN (807-809), but differs from others, which found no effect of lymph node metastases on survival in patients with various pNEN (130,314,806,810-813).

 

Numerous characteristics of the gastrinoma itself correlate with decreased survival including (Table 10): pancreatic location over duodenal location; increasing primary size; rate of growth overtime; in addition to the presence of liver or lymph node metastases the development of bone metastases has a poor prognosis. The development of ectopic, Cushing’s syndrome or bone-metastases has a particularly poor prognosis with survival averaging only one year (109,113,294,611).  The fact that duodenal and pancreatic gastrinomas are equally malignant (40-70%=lymph node metastases), but not equally aggressive, with liver metastases present in 25-40% of pancreatic gastrinomas, but in only 2% of duodenal gastrinomas; results in pancreatic gastrinomas having a worse prognosis (Table 10) (27,109,110,314,433). Other features of gastrinomas associated with a poor prognosis including advanced ENET/WHO classification, higher ENET/WHO grade, poor differentiation, other histological features, and rapid growth (Table 10) (65,142,301,814).

 

As mentioned earlier in the pathology section of this paper, both the recently developed TNM tumor classification systems (ENETs, UICC/AJCC/WHO) and the tumor grading systems have been shown to be the most important single factors in numerous multivariate analyses for predicting overall survival or disease-free survival in all NENs ( pNEN, GI-NENs (Carcinoids) (Table 10) (115,116,142,815-817). Most (>90%) of gastrinomas are well differentiated NENs (Grade G1 or G2), and at present there is only one study just including only gastrinomas showing the importance prognostic effect of grade on survival of ZES patients (65). However, because of the almost universal importance of these classification/grading systems in studies involving all pNEN, it is almost certain this will be true of gastrinomas also.

 

Table 10. Prognostic Factors in Patients with Gastrinomas (overall survival or associated with increased development liver metastases)

Prognostic factor for decreased survival

 Reference(s)

I. GASTRINOMAS ONLY

I.A. Acid Control

Uncontrolled acid hypersecretion

(1,27,205,433,638)

I.B. Demographic Features

Female gender (p=0.024)

(109,110)

Diagnosis before 1980 (p=0.010)

(315)

Older age at diagnosis (p=0.001)

(65,315)

I.C. Disease Clinical/Lab Features

MEN1 absent (sporadic ZES) (p<0.03) (Fig.3.D)

(64,65,110,207,734,818)

Short disease history prior diagnosis (<3 yrs.) (p<0.001)

(109,110,819)

High gastrin (p=0.022)

(109,110,191,507,819)

I.D. Disease Course

Recurrence postop with short disease-free interval

(254)

Develop ectopic Cushing’s syndrome (p=0.0049)

(109,113)

Primary gastrinoma location

 

Pancreatic >duodenal (p<0.004)

(109,110,281,315,734,819)

I.E. Tumor size, Location, Extent, Growth Rate

Large primary tumor size (>2-3 cm)

(109,110,315,433,507,819)

Gastrinoma located to the Left of the SMA> right of SMA (gastrinoma triangle)

(415)

Presence of Lymph node metastases (p<0.004)

(109,734,806)

Extent/presence of liver metastases (p<0.0001)

(27,64,109,110,254,314,314,433,507,734)

Diffuse>localized (p<0.0001)

 

Diffuse>both lobes>single lobe>none

 

Rate of growth of liver metastases or tumor

 

Rapid> slow, none

(111,402,429)

Time liver metastases diagnosed

 

Present initially>develop on follow-up (p=0.02)

(109)

Develop bone or extrahepatic metastases (p<0.0001)

(65,109,112,433,611)

I.F. Specific Tumor Features

Flow cytometric results

 

High S phase, low % nontetraploid aneuploid, multiple stem line aneuploid frequent

(820)

Molecular changes

(821)

(Chromosome 1qLOH) (p=0.019))

(822)

(Chromosome XLOH) (p=0.042))

 

Tumor grade

(65)

II. ADDITIONAL FEATURES SHARED WITH OTHER pNEN

II.A. Classification

Advanced TNM classification (ENETs, UICC/AJCC/WHO)

(41,115,116,142,816,817,823)

IIB. Histological Features

Poorly differentiated

(41,115,116,142,824,825)

High Ki67>low Ki67 proliferative index

(115,116,142,815-817)

Cytokeratin 19-IR positivity

(115,116,826-828)

Vascular, neural invasion

(115,116,639,829)

Decreased expression of autophagic genes

(117)

Alternative lengthening of telomeres, ATRX/DAXX loss

(444)

Alpha cell origin over Beta cell origin

(444)

IIC. Other Features

Age

(41,825)

Gender

(825)

Poor symptom control post-resection

(32)

No surgical resection

(41,825)

NF-pNEN rather than F-pNEN

(825)

Tumor size

(825)

Abbreviations: SMA, superior mesenteric artery; LOH, loss of heterozygosity; IR, immunoreactivity; ENETs, European Neuroendocrine Tumor network; postop-postoperative

 

A wide range of different anti-tumor treatments is used in patients with advanced pNEN, which are similar to that used in all advanced NENs. These include: surgical resection including cytoreductive (debunking) surgery; liver-directed therapies including radio-frequency ablation (RFA)/other local ablative therapies; trans-arterial embolization (TAE) or chemo-embolization (TACE), radio-embolization or selective internal radiation therapy (SIRT); chemotherapy; biotherapy with somatostatin analogues or interferon-Alfa; molecular targeted therapy with mTOR (everolimus) or tyrosine kinase inhibitors; peptide radio-receptor therapy (PRRT),  liver transplantation and immunotherapy (38,50,142,158,604,830-839). There are only a few small, specific studies including only patients with metastatic gastrinomas as they are usually included in series with other metastatic pNEN, and in some cases even with GI-NENs (Carcinoids). Thus, below the results will primarily be from series containing pNEN with some gastrinomas.

 

One of the main problems with all forms of anti-tumor treatment in patients with advanced pNENs and other NENs, is the development of resistant to the therapies with time. This occurs to varying degrees at different times with all of the current therapies except complete surgical removal without recurrence (840,841). Numerous experimental approaches have been tried after failure of the different primary therapies, with the most frequent approach used is to switch to another primary established approach (840,841). The development of resistant with treatment and approaches recommended to deal with it will be briefly covered in each of the following sections dealing with the various established specific primary anti-tumor therapies.

 

Cytoreductive Surgery 

 

In patients with gastrinomas with advanced metastatic disease, similar to all malignant NENs, it is recommended that the possibility of surgical  removal of all resectable tumor (cytoreductive surgery, debunking surgery) should be considered by many authorities although there are no controlled studies to support its value (9,41,128,142,143,157,171,181,191,258,288,831,842-862,862-864). Surgery is generally recommended if ≥90% of all imageable disease can be removed, although others, have recommended lower numbers. There are only a few reports containing primarily gastrinomas treated with this approach (122,258,288,403,842-844,865), with most studies reporting results from different malignant pNEN and in some cases combined with GI-NENs (carcinoids). In various studies using this approach five-year survivals of 75-80% are reported and increased survival over patients not undergoing such surgery (128,142,143,157,831,849-859,861,862,862,864,866). This approach is primarily used in patients with well differentiated NENs, which is the case in >90% of gastrinomas (G1, G2,G3NET). Unfortunately, this approach is possible in the minority of even patients with advanced well differentiated gastrinomas or other NENs (<15-20%), and because of lack of control studies establishing its efficacy, it is not uniformly used. In only a small minority of G3NEC patients, is such an approach considered and even then, it is controversial (857,867).

 

At the time of any abdominal surgery, it is generally recommended that prophylactic cholecystectomy be performed because of the widespread use of somatostatin analogues for their anti-tumor activity and the ability of long-term treatment with them to cause biliary stasis and gallstones (143,180,849,868).  Lastly, recent non-controlled studies, report that removal of the primary tumor increases the survival rate with PRRT and suggest it routinely be performed, although this approach is not widely used at present (869).

 

Liver Directed Therapies  

 

GENERAL

 

These approaches include the use of local ablative techniques (radiofrequency ablation (RFA), ethanol injections, cryotherapy), which are frequently used in combination with other anti-tumor treatments, as well as various more general hepatic cytotoxic approaches using trans-arterial embolization (TAE)/chemo-embolization (TACE) or radioembolization (42,142,143,831,834,851,859,870-872,872-877). The embolization approaches in gastrinoma patients are generally reserved for patients who have metastatic unresectable hepatic metastases either limited to the liver or with liver-predominant disease, particularly if locally symptomatic, whereas in patients with other F-NENs,  they are also frequently used for patients in whom the symptoms due to F-NEN  excess-state not controlled by other modalities (25,142,143,831,834,851,870-872,876,878,879).

 

RADIOFREQUENCY AND OTHER ABLATIVE THERAPIES

 

 Of the all of the liver-directed therapies, RFA is the most widely used, which converts RF waves to heat resulting in cellular destruction (142,831,880-883). RFA and other ablative techniques (cryotherapy, ethanol injections) are administered either at the time of surgery (+/- laparoscopic) to ablate isolated metastases or by radiological techniques for guidance (25,142,844,847,881,882,884-887).In different studies, relative contra-indications to its use are the presence of large lesions (>3.5-5 cm), a large number of lesions (>5-15), and he presence of metastases near vital structures (831,848,880,880,881,883,885,887-889). RFA has response rates of 80-95% which last up to 3 years, has the lowest complication rate of all liver-directed therapies (<15%), and can be used alone for a palliative procedure or to supplement a surgical resection by removing additional isolated liver metastases (25,831,880,885,888).

 

Embolization and Chemoembolization

 

Embolization of advanced metastases in the liver in patients with metastatic gastrinomas or other NENs, can be used because the blood supply to the tumor is primarily arterial, whereas in normal liver only 20-25% is arterial, with the majority coming from the portal vein (42,142,831,870,872-876,880,882). Therefore, interrupting the tumoral area arterial supply preferentially affects metastases (142,870,880,882). At present, this procedure is usually performed radiological, rather than at surgery and can be done alone (trans-arterial embolization or TAE) or accompanied by administration of chemotherapeutic agents (trans-arterial chemoembolization or TACE) such as doxorubicin, cisplatin, 5-fluorouracil, mitomycin C or streptozotocin (42,142,837,870,872,872,873,875,876,880,882,888,890).   TAE or TACE is performed using gel foam powder or polyvinyl alcohol particles. In various studies a response is seen in 55-100% of symptomatic patients, 25-85% have an objective tumor response, and responses last from 6-45 mos. (142,831,834,870,872,880,891). Five-year survival rates for TAE/TACE are 20-35% and the progression free survival is 1.5 years (142,870,892). Contra-indications are the presence of portal venous occlusion, liver failure, extensive liver involvement (>50-75%), poor performance score, and previous biliary surgical reconstruction (142,870,880,888,890-893). Both TAE and TACE are associated with side-effects including a mortality rate of <6%, complications in 10-80%, particularly post embolization syndrome (pain, fever, nausea/vomiting), and occasionally gallbladder necrosis, hepatic failure, abscess formation and liver/renal failure (831,834,870,880,882,888,890,891,894).  TAE/TACE are generally considered for palliative therapy in patients with non-resectable liver metastases with hepatic predominant disease (142,143,180,834,872,891). There are no prospective studies that have established the value of TAE/TACE; however, both the NANETS and ENETs guidelines recommend TAE/TACE be considered for palliative treatment in an experienced center if the patient has hepatic-only or hepatic-predominant disease that is not surgically resectable (142,143,180,182,834,848,891).

 

Radioembolization or Selective Internal Radiation Therapy (SIRT)

 

Radio-embolization or selective internal radiation therapy (SIRT) utilizes 90Yttrium-labeled microspheres (Sir-spheres-20-60 um diameter, load-50Bq/sphere or Theraspheres-glass sphere, 20-30um diameter, 2500 Bq/sphere), which are administered by selective intra-arterial injection after a pretreatment angiogram to allow correct catheter localization (9,142,181,182,831,848,871,872,876,876-878,895-905). Prior to their administration, the position of the catheter tip needs to be properly established so that microsphere administration does not enter the cystic or duodenal arteries, which can result in cholecystitis or ulceration, and the amount of lung shunting must be determined to avoid radiation pneumonitis (142,831,871,871,872). Contraindications include: the presence of excessive shunting to the lung/GI tract; inadequate liver reserve; and the inability to isolate the liver arterial tree from the gastric/small intestinal branches (142,831,871,872,905,906). The mean objective response rate from 12 studies in patients with advanced NENs was 55% (range-12-90%) with stable disease seen in 32% (range-10-60% (142,831,871,905,907) and the disease control rate was 91% in a recent multicenter international study (908). The mean survival is 30-months and 50% of patients have symptomatic improvement in quality-of-life indices (142,871,897-899). Side-effects include post-embolization (fever, nausea, vomiting, abdominal pain) (25-45%), \and rarely ulceration or cholecystitis (<1%) if the catheter is not properly positioned (142,831,871,905,907,909).

 

At present the exact embolization procedure that is preferable in which clinical situation is not clear because of lack of prospective comparative studies. There have been no randomized control trials comparing radioembolization to the other liver-directed therapies, so at present at is unclear which should be preferred.

 

Medical Treatment of Advanced Metastatic Disease

 

CHEMOTHERAPY

 

Chemotherapy has a poor response rate (<15%) in well-differentiated NENs outside the pancreas (lung/GI-NETs, carcinoids) and thus is uncommonly used for these tumors, it has higher response rates in different series of malignant, well-differentiated pNEN, varying from  25% to 70 % (9,147,181,182,832,848,893,904,910-920). Until recently the generally used chemotherapeutic regimen was streptozotocin (STZ) based for advanced pNEN (most frequently combined with doxorubicin, 5-FU or cyclophosphamide) (9,142,147,181,182,832,878,893,910,916-919,921). STZ is a glycosamine-nitrourea derivative which was found to have cytotoxic effect on pancreatic islets (28), and since 1968 has been used for the treatment of patients with metastatic pNEN/gastrinomas (9,142,181,182,878,893,916-918,921). However, recent studies report temozolomide and capecitabine may have at least similar if not better activity than STX based regimens (827,830,911-913,918-920,922-927). Recently, in a randomized, prospective trial (ECOG-ACRIN E2211) (928) the Eastern Cooperative Oncology group compared the combination of temozolomide (TMZ) and capecitabine (CAP)(CAPTEM) to TMZ alone in 144 patients with advanced G1/G2 pNENs.  At the interim analysis the mean PFS was 14.4 mos. in the TMZ alone group and 22.7 MS FR CAPTEM (P=0.022).  At the final analysis the mean S was 53,6 mos. for TMZ and 58.7. mos. with CAPTEM. MGMT deficiency was associated with the response in this study. The authors concluded that the CAPTEM combination was superior to TMZ alone and that MGMT deficiency correlated with a response (928). In a recent systematic review of CAPTEM treatment of patients with advanced NENs, involving 42 articles with 1818 patients the overall disease control rate was 77% (range 44-100%), the median PFS ranged from 4 to 38 mos., and the media OS ranged from 8 to 103 mos. In this review (920) the safety analysis showed an occurrence of G3-G4 toxicities in 16% of the patients treated.  The most common toxicities were hematological (27%), gastrointestinal (8%) and cutaneous (3%). This systematic analysis (920) concluded CAPTEM was an effective and relatively safe treatment for patients with well-moderately differentiated NENs of pancreatic, GI, lung and unknown origin.

 

 STZ-based regimens have considerable morbidity with 70-100% developing some side-effect including nausea/vomiting (70-100%), abnormalities in hepatic function, leukopenia, and thrombocytopenia in 6%, and 15-40% developing some degree of renal toxicity including proteinuria (40-60%) and decreased creatinine clearance (893,913,916,917). The combination of STZ/doxorubicin (±5-FU) has an objective response rate of 20-45%, but complete responses are rare, and the median-response duration is 5-20 months (9,142,181,182,830,878,893,914,916-919,921). In patients with advanced gastrinomas only, the response rate varied from 5 to 40% (147,929).

 

Poorly differentiated pNETs comprise only 2-6.5% of all pNETs; however, it is important that they be identified because they have an aggressive course and poor prognoses and are generally treated differently than well-differentiated pNEN (54,142,848,912,913,918,930-935). The recommended chemotherapeutic drug combinations for treatment of well differentiated pNEN differs from that for treatment of poorly differentiated NENs (Grade 3) in any location (142,143,180,912,913,932,935-937). In contrast to the combinations listed above to treat advanced well differentiated pNEN, in patients with poorly differentiated NENs, a cisplatin-based drug combination with etoposide is generally the initial treatment (142,143,180,848,912,913,918,919,931,932,936,938,939). This combination results in an objective response in 30-80% of patients with mean duration of <12 months (142,143,180,912,932,936). The median survival is 4-16 mos., and the 5-year survival is 11% (range 0-31%) (142,143,180,912,913,918,931,932,936,938). This chemotherapeutic regiment can be associated with significant toxicity including GI toxicity (nausea/vomiting), myeloid-suppression and renal toxicity (142,143,180,912,913,931,932,936,938,939). In a recent multicenter study promising results were reported with the use of temozolomide/capecitabine (92%/8%=TMZ alone) (CAPTEM) in patients with Gr3 GP-NENs (933). In this study (933) the results of treatment with CAPTEM were reported from 130 patients (67% pNENs) and a radiological response was seen in 36%, the median TTF was 3.6 mos., OS was 9.2 mos., with the TTF being longer in pNENs than patients with GI-NENs Gr 3 tumors (5.8 vs 1.8 m0s, p=0.04). The role of surgery in patients with high grade NENs is controversial (940), although it is reported to be associated with higher survival in those with Gr3 WDs in some studies (940).

 

Recently, some studies (926,928,941-948) report that the effectiveness of alkylating agents in NENs correlated with the expression of, but not all (489,945,949-951) the DNA repair enzyme, O6-methylguanine DNA methyl transferase (MGMT) in these tumors. MGMT, in its role as a DNA repair enzyme, specifically removes the methyl/alkyl group form the O6 position of guanine, whereas alkylating agents induce methylation at this site which leads to DNA mismatch occurring and results in cell death/apoptosis. Some studies show that pNEN have a higher response rate to alkylating agents due to their low level of MGMT (926) compared to GI-NENs (carcinoids) having higher MGMT levels and lower response rate. Perspective studies are needed before recommending the routine determination of NEN tumoral MGMTs to help predict, for a given patient the subsequent response to an alkylating agent, and therefore its routine use is not generally recommended at this time (928,945-948). 

 

BIOTHERAPY

 

Somatostatin Analogues

 

Similar to NENs in general, most well-differentiated G1, G2 pNEN, as well as a proportion of G3 pNEN, overexpress one of the 5 subtypes of somatostatin receptors (sst1-5) (most frequently sst2) (142,851,952-955).  Numerous studies, including both non- controlled and randomized controlled studies (PROMID, CLARINET studies)  on NENs and pNEN(including gastrinomas) (142,954,956,957), demonstrate that somatostatin agonist analogs (octreotide, lanreotide) are  not only are effective for controlling the hormone-excess state in F-NENs(discussed in a previous section), but also have anti-tumor growth effects in NENs (25,142,155,402,603,609,848,851,952-955,958-965). The exact molecular basis for this antiproliferative-effect is not entirely clear, but somatostatin analogues inhibit the release of growth factors from NETs, have antiproliferative effects on neighboring cells (stromal, immune, vascular, etc.) and activate intracellular cascades that have antiproliferative effects (phosphatases, inhibition of adenylate -cyclase, etc.) (603,954,959,962-964). In these studies the anti-tumor effect of the somatostatin analogues is almost entirely a tumoristatic effect (only 10-15% show decreased tumor-size), resulting in disease stabilization with prolongation of progressive free survival, and because of study design and the multiple treatments the patients received, an effect on overall survival has not been established (155,402,603,609,848,956,958,961-964,966). However, these agents are very well tolerated, and are generally the first line treatment of patients with advanced well-differentiated gastrinomas and other pNEN as well as with lung/GI-NENs(carcinoids) (25,142,143,150,164,180,833,851,952-956). In a recent study of the use somatostatin analogs (SSAs) on tumor progression in 12 ZES patients (WD, G1, G2) (155), 67% had a sustained response to SSAs and 33% showed early progression. There was a significant difference in PFS between the early and late progression groups (84 vs 2 mos., p=0.004) (155). However, there was no difference OS or PFS between these 12 SSA treated ZES patients and 21 other ZES patients not treated with SSA analogues (155).

 

With time the tumor may become refractory to the antigrowth effect of the somatostatin analogue, and its efficacy may be restored by either increasing the dosage or shortening the time interval between doses (965). Side effects that result in somatostatin analogue therapy discontinuation are rare with any side-effect occurring in 50% of patients (including pain at injection site, GI symptoms), which may improve with continued treatment (25,29,142,848,954,955,958,961,964,967). Long-term more serious side-effects include the development of biliary sludge/gallstones which cause symptomatic disease in <1%, developing glucose intolerance/diabetes or developing steatorrhea which is usually mild (25,29,142,142,848,868,954,955,958,961,964,967).

 

Interferon

 

Interferon-alpha, similar to somatostatin analogues, is able to control symptoms of the hormone hypersecretory state and has anti-proliferative effects in pNEN/NENs which result in primarily disease stabilization, rather than a decrease in tumor size (<15%) (142,848,961,968-973). In the only study of interferon limited to gastrinoma patients (13 patients, advanced metastatic progressive disease) (972), 46% of patients showed disease stabilization, and in 23% it lasted almost 2 years. The antiproliferative effect on pNENs/NETs of interferon is partially mediated by blocking cell-cycle progression in G1, inhibiting DNA synthesis, stimulating an increase in Bcl-2, inhibiting protein synthesis, inhibiting angiogenesis, and induction of apoptosis (961,968,970,971). Side effects develop in the majority of patients (>70%) with the most frequent being flu-like symptoms (40-80%), weight loss/anorexia (60%), and fatigue (51%), which frequently decrease in severity with continued treatment or with decreased dose (848,961,970-972). More serious side effects include hepatotoxicity (31%), hyperlipemia (31%), and bone marrow toxicity; autoimmune disorders particularly thyroid disease and rarely CNS side effects such as depression or mental disorders (142,961,968,970-972). While interferon-alpha was frequently used in the past either alone or with somatostatin, at present it is uncommonly used because of the availability of other agents with fewer side effects.

 

MOLECULAR TARGETED THERAPIES  

 

mTor-Inhibitors (Everolimus)

 

Both extensive in vitro and in vivo studies demonstrate that activation of the mTOR cascade, plays and important role in the proliferation, growth, and apoptosis of pNEN, as well as NENs in other locations (142,150,479,882,915,957,964,974-979). mTor is a serine-threonine kinase critically involved in a variety of cellular functions including apoptosis, cell-growth, and proliferation (150,957,974,977,978,980). A number of different mTOR antagonists have been developed and shown to have anti-proliferative effects in NENs in various in vitro and in vivo studies, however, only one, everolimus, has been approved by the FDA for patients with advanced NENs (pNEN (including gastrinomas) and GI-NENs (carcinoids) (142,150,480,974-976,978-981). The approval of everolimus for use in both advanced pNEN and GI-NENs(carcinoids) was based on the positive results of two randomized, double-blind, prospective, placebo-controlled studies, RADIANT-3 (pNEN) (480) and RADIANT-4 (lung/GI-NENs) (982), which each demonstrated almost a 3-fold increase in PFS (p<0.001). There are no specific studies on the effects of everolimus on gastrinomas only, and the only data comes from general trials of all pNEN.

 

Everolimus treatment was associated with a 2-fold increase in adverse events, the majority being grade 1 or 2, with grade 3 or 4 side effects occurring in 3-7% (primarily hematological, stomatitis, or hyperglycemia) which could be managed by dose-reduction or drug interruption (480). At present, it is not established whether everolimus’ ability to increase progression-free survival will result in an increase in overall survival (142,981).

           

Long-term treatment with everolimus is associated with primary and acquired resistance, which has frequently limited its long-term benefit for patients with advanced NEN (150,983-985). Numerous mechanisms for this have been described but none has been sufficiently successful to lead to its widespread use or its ability to function as a biomarker for the occurrence of resistance (983-985).

 

Numerous studies have provided information on the importance of angiogenesis in the pathogenesis of pNENs as well as other NENs (986). A recent randomized Phase II study compared the effectiveness of everolimus alone versus everolimus plus the anti-VEGF agent, bevacizumab in 150 patients with advanced pNENs (987). The combination resulted in improved PFS compared to everolimus alone (16.7 mos. vs 14 mos., complete tumor response in 31% with the combination compared to 12% with everolimus alone (p=0.0053), with similar median overall responses.  Toxicities were more frequently seen in the combination group (987). The authors concluded that these results support the need for continued evaluation of VEGF pathway inhibitors for the treatment of advanced pNENs (987).

 

Tyrosine Kinase Receptor Inhibitors (Sunitinib and Surufatinib)

 

PNEN including gastrinomas, similar to other NENs, as well as other neoplasms and normal tissues, frequently possess multiple tyrosine kinase (TK) receptors, which are important in mediating growth, angiogenesis, differentiation, and apoptosis (26,142,848,895,957,964,988-995). TK receptors comprise >20 families of transmembrane receptors that mediate the actions of a number of different growth factors that include the receptors for insulin-like growth factor (IGF1R), epidermal growth factor family (EGFRs); hepatocyte growth factor (c-Met); platelet-derived growth factor family (PDGFRs); vascular endothelial growth factor family (VEGFRs); stem cell factor (c-Kit) and a number of others (142,957,992,994). A number to tyrosine kinase receptor antagonists (sunitinib, axitinib, cabozantinib, famitinib, nintedanib, pazopanib, sorafinib, sulfatinib, surufatinib) (979,994,996-998,998-1001) have been shown to have anti-growth/anti-angiogenic effects on pNEN/NENs in both in vitro and in vivo animal studies, however, at present the only two are approved for use in various countries which are sunitinib (US, Europe, other countries) which is an inhibitor of a number of tyrosine kinase receptors (PDGFR, VEGFR1/2,c-KIT, FLT-3) (142,914,989,991,994,996,1002-1004) and surufatinib (approved in China) which is an inhibitor of VEGFR, FGFR1, and colony stimulating factor 1 (CSF1R) (998,998-1001). In a Phase 3 double blind, randomized trial (1002) in 171 patients with progressive well-differentiated nonresectable pancreatic NENs (including 19 patients with ZES), sunitinib resulted in a doubling of PFS (11.4 vs. 4.5 mos., p<0.001), which lead to its approval for advanced pNEN. There are no specific studies on the effects of sunitinib on gastrinomas only, and the only data comes from general trials of all pNEN.

 

Sunitinib treatment is associated with frequent grade 1/2 side effects and some grade 3/4 side effects particularly neutropenia (12%) and hypertension (10%) (994,1002,1005). A quality-of -life analysis (1002) showed sunitinib did not have a significant effect, with most side-effects able to be managed by dose-reduction and/or temporary cessation of treatment.

 

Surufatinib was evaluated in two multicenter studies, one a randomized, double-blind, placebo controlled Phase III involving 172 patients with advanced pNENs in 21 different Chinese hospitals (999) and a second study (SANET-ep) involving 198 patients with  advanced extra-pancreatic NENs 1in 24 hospitals in China (998),  In the first study in patients with pNENs  (999) the PFS was 19.3 mos. with surufatinib vs 3.7 with placebo (p=0.00110) with the most common Gr ¾ side-effects being hypertension (38% vs 7%), proteinuria (10% vs 1%) and hypertriglyceridemia (7% vs 0%). In the second study on patients with advanced extra-pancreatic NENs (998) the PFS was 9.2 mos. with surufatinib vs 3.8 with placebo (p<0.0001) with the most common Gr ¾ side-effects being hypertension (47% vs 13%) and proteinuria (19% vs 0%). Currently, surufatinib is not approved by either the FDA or European regulatory agencies.

 

Long-term treatment with sunitinib is associated with primary and acquired resistance, which has frequently limited their long-term benefit for patients with advanced NEN (840,841,983,984,994,995). Numerous mechanisms for this have been described but none has been sufficiently successful to lead to its widespread use or its ability to function as a biomarker for the occurrence of resistance (840,841,983,984,995). 

 

Peptide Radioreceptor Therapy (PRRT) Using Radiolabeled Somatostatin Analogues

 

PRRT utilizes the fact that almost all well-differentiated NENs, as well as a proportion of G3NECs, overexpress somatostatin receptors (sst1-5) particularly sst2, which can bind radiolabeled somatostatin analogues resulting in the targeted delivery of cytotoxic radiation to the tumor cells (142,154,603-605,607,608,836,952,953,1006-1018). Two different isotopes have been used in most studies: 90Yttrium(90Y)- or 177Lutetium(177Lu)- labeled somatostatin analogues (142,154,836,1008-1016,1018). 177Lu emits beta-particles and gamma rays, has a maximum tissue penetration of 2 mm, and a half-life of 6.7 days, whereas 90Y strongly emits beta-particles, has a maximal tissue penetration of 12 mm, and a half-life of 2.7 days (1011-1015). A number of different synthetic somatostatin analogues have been used, with the most frequent being octreotide or octreotate coupled to the radiolabel by different chelators, including diethylene triamine penta-acetic acid (DTPA) and 1,4,7,10-tetraazaacyclododecane-1,4,7,10-tetraacetic acid (DOTA) (142,1010,1011,1013,1014).

 

At present the only approved formulation for pNEN is 177Lu-DOTATATE (604,605,607,608,1006-1010). The approval of this therapy is based on results of a double-blinded, control phase 3 trial (NETTER-1) (1019) in patients with advanced unresectable, midgut carcinoids which showed a marked prolongation of PFS (from 8.4 mos. to >40 mos., p<0.0001), with an increased overall survival from 3 to 18%, combined with the results of treatment of 510 patients with advanced pNEN and other NENs, in Rotterdam which showed complete response in 2%, partial response in 28%, and tumor stabilization in 35% (404,1015). Of 440 patients (10 studies) with various malignant pNEN/NETs, including gastrinomas, treated with 90Y-labeled somatostatin analogues, complete tumor remission was rare (0-6%), partial remission occurred in 7-37%, and tumor stabilization in 40-86% (29,142,1011,1013,1014). In a recent meta-analysis of 22 studies (1758 patients) with advanced NENs treated with PRRT the pooled disease response rate (complete/partial tumor response) was 33% with RECIST criteria, and the pooled disease control rate (compete/partial response or stable disease) was 79% (1020). In a second recent meta-analysis of PRRT results (1018) involving 15 studies selected from 715 references in patients with advanced NENs, the pooled response rate was 27.6% by RECIST criteria, and 20.6 % by SWOG criteria, with respective DCR rates of 79.1% and 78.3% by the two criteria demonstrating excellent agreement with these two different criteria of response.

 

In two different studies there was no significant difference in the PFS overall survival rates between NEN patients treated with PRRT with advanced pNEN compared to NENs in other sites (869,1010).

 

Gastrinomas are one of the malignant pNEN/NETs that were most responsive to PRRT (42%-3mos); however, they also had one of the highest recurrence rates leading to a poorer prognosis (142,404,1011,1013-1015,1021). In one detailed study of 11 patients with metastatic ZES (1022) treated with either 90Y-and/or 177Lu-labeled somatostatin analogues, the mean serum gastrin decreased by 81%, compete response occurred in 9%, partial tumor response in 45%, tumor stabilization in 45%, and in 64% the antitumor effect persisted for a median period of 14 months. In a second study (1023) involving 30 gastrinoma patients treated with 90Y- labeled somatostatin analogues the tumoral partial response rate was 33% with a mean overall survival time of 40 mos.

 

The treatment was well tolerated when given with renal protective amino acid infusions, with no Grade 3/4 nephrotoxicity. In various studies the most serious side effects are hematological (15%-transient, 0.8% developing a myeloproliferative disorder), liver toxicity (0.6%) and renal toxicity, with the latter occurring primarily in patients receiving 90Y-labeled somatostatin analogues (154,404,604,605,607,608,1006,1007,1010,1015,1019).

 

Recent studies show that in patients with refractory hormonal symptoms due to a F-NEN, PRRT may be of great help in control the hormone excess state’s symptoms independent of its effect on tumor proliferation (25,148,350,860,1010,1024). Although this is almost never an issue in patients with gastrinoma because of the effectiveness of PPIs in controlling the symptoms of the gastric acid hypersecretion, numerous recent studies suggest that PRRT may be particularly helpful in patients with refractory advanced F-NEN syndromes in controlling the hormone excess state, particularly in patients with carcinoid syndrome, VIPomas and insulinomas (25,148,350,860,1010,1024).

 

With 177Lu-(DOTA0, Tyr3) octreotate, a number of prognostic factors were identified which predicted a poor outcome after PRRT, which included; presence of progressive metastatic disease prior to treatment; Karnofsky performance score of ≤70; no tumor-response to PRRT; weight loss at the time of treatment; presence of bone metastases; extensive liver involvement; poor uptake of the radiolabeled analogue by the tumor; the presence of malignant gastrinoma, VIPoma, or insulinoma and the presence of positive lesions on 18F-FDG Pet scans(142,404,612,1011,1013-1015,1021,1025,1026).

 

Recent studies support the conclusion that retreatment of NEN patients with PRRT who develop progressive disease after an initial PRRT treatment, is a feasible option with good efficacy and acceptable toxicity (1027,1028). This conclusion was supported by two recent systematic reviews and met analyses (1027,1029). In the report by (1027) 9 articles with 426 patients were analyzed and the ORR after retreatment was 17.1%, DCR was 76.9%, PFS was 14.1 mos. and OS was 26.9 mos. Pooled proportions of hematologic and renal toxicities were 11% and 0.7%. In a subgroup allowing direct comparison to initial PRRT treatment data, the salvage PRRT had a lower therapeutic efficacy (ORR, DCR, p<0.001) and shorter PFS (P=0.03), despite similar hematologic and renal toxicity. In a second analysis (1029) 13 studies were identified containing retreatment data (177Lu-PRRT, or 90Y-PRRT) from 567 original studies. With 177Lu-PRRT retreatment in 7 studies PFS was 12.5 mos., mean OS was 26.8 mos., and DCT was 71%. Grade ¾ toxicity occurred in 5% of 177Lu-PRRT retreated patients, with no Gr ¾ renal toxicity and no myelodysplastic and acute myeloid leukemia incidence.

 

Liver Transplantation

 

In contrast to many metastatic tumors, liver transplantation is both recommended for pNEN/NENs (including patients with advanced gastrinomas) and is selectively used in a small number of patients with metastatic advance metastatic pNEN/NENs, although it use remains controversial (142,143,1030-1038). In one recent systematic analysis of reported NEN series the 1-,3- and 5-yr survival rates were 89%, 69% and 63% with a recurrence rate after transplantation of 31-57% (1032). In another recent (2020) review of 206 patients with metastatic pNEN/NENs who underwent liver transplantation the overall survival rates at 1,3,5 and 10 years were 89%,75%, 65% and 46% respectively (1039). In this study (1039) the recurrence rate was 34% with a mean time to recurrence of 28 mos. Important prognostic factors in the systematic analysis (1032) were >50% liver involvement by tumor, high Ki67, or the presence of a pancreatic primary over a GI-NEN. In other studies important other factors predict a poorer prognosis include: older age of patient(>50), older age of donor, cold ischemic time MELD score, tumor recurrence, presence of a symptomatic tumor, a primary tumor in the pancreas as opposed to an extra-pancreatic NEN (carcinoid), poor tumor differentiation, transplantation associated with a simultaneous and extensive digestive tract  resection, presence of hepatomegaly,  presence of extra-hepatic metastases and a patient with a primary tumor not resected (1034-1036,1039). Liver transplantation is generally reserved for patients with life-threatening hormonal disturbances refractory to other treatments (which is very rare in ZES) or to selected patients with pNEN/NENs with diffuse liver involvement refractory to all other treatments (26,142,1033-1040).

 

If liver transplantation is considered, important selection criteria include the presence of a well-differentiated NEN/pNEN; age <45-50; a Ki-67 index <10%; <50% liver involvement; the absence of extra-hepatic metastases as determined using the most sensitive methodology (68Ga-labeled somatostatin-analogues and PET imaging) ;  the absence of extra-hepatic disease(resected primary tumor); the absence of other resections at the time of liver-transplantation; and some groups consider various histological features such as E-cadherin-tumor staining characteristics (142,848,930,1032,1034-1039,1041,1042).

 

Immunotherapy

 

Since the recent widespread effectiveness of immune check point inhibitors in a number of tumors (melanomas, etc.), including in patients with poorly differentiated lung NENs with small cell neuroendocrine tumors, there have been several studies of the efficacy and safety in other tumors such as patients with advanced NENs including pNEN such as advanced gastrinomas (995,1043-1049).

 

A number of these studies suggest single agent (Anti-PDL1) immunotherapy may be a useful approach in only small subset of patients with advanced disease (12% in Keynote 02 study (1047) , 0-5% in other studies not selecting for PD-L1 activity (1049-1051)  and this subset is primarily patients with high-grade poorly differentiated NENs (995,1043-1046).  In various GEP-NENs the presence of high tumor-infiltrating lymphocyte (TILs) numbers and high PD-1 expression show a significant correlation with decreased survival and higher grading of the tumor supporting the finding that immunotherapy might be most promising in GEP-NENs with high TILs (1044).  In other tumors particularly melanomas, renal cell carcinoma and non-small cell lung tumors, a combination of immunotherapies using anti-PD-L1 and CTLA-4 blockade resulted in increased efficacy, and a recent study in patients with various advanced NENs reported an objective response rate of 24 % with increased activity in patients with both atypical lung NENs and with high-grade pNEN (1046). Other studies report ORR of 14.7-25% almost entirely in patients with high grade NENs (995). Ongoing and future studies are in progress to exactly define the best combinations as well as define which specific NEN types will best respond (995,1043-1045).

 

Neoadjuvant Therapy  

 

Because the large majority (>80%) of patients with advanced gastrinomas or other advanced NENs present with surgically unresectable disease, primarily with widespread liver metastases or unresectable locally invasive tumors, the possible benefit of surgical resection is not an option. Therefore, with the increasingly effectiveness of various anti-tumor modalities, there is increasing interest in a possible role of neoadjuvant treatments which could allow tumor downsizing to the point surgical resection can become an option (1052,1053). In one recent systematic review and meta-analysis of 9 studies from the literature (1052), involving 468 patients with advanced pNENs who had adjuvant therapies, there were no complete responders, 43.6% had a partial response; 51.3% had stable disease; and 4.3% had progressive disease. The estimated resection rate was 68.2%, and the RO resection rate was 60.2% (1052). There was no difference in the resection rate between different chemotherapy regimens (41% vs 34%), as well as the RO resection rate (62% to 68%) and the ORR was similar with CAPTEM and FAS (42% and 34%). PRRT showed a higher numerical ORR than chemotherapy although the difference did not reach statistical significance (49% vs 37%, p=0.154). The authors concluded these results were promising for some patients, however the best neoadjuvant regime to use remains unclear (1052).

 

REFERENCES

 

  1.             Zollinger RM, Ellison EH. Primary peptic ulcerations of the jejunum associated with islet cell tumors of the pancreas. Ann Surg 1955;142:709-728
  2. Stabile BE. Gastrinoma before Zollinger and Ellison. Am J Surg 1997;174:232-236
  3. Wilson SD, Ellison EC. Invited Commentary. Ann Surg 2019;270:e22
  4. Soreide JA, Lea D. The Gastrinoma Saga Before Zollinger and Ellison: The Strom Case Revisited. Ann Surg 2019;270:e19-e21
  5. Soreide JA, Hem E. Roar Strom - the Norwegian surgeon who was three years ahead of Zollinger and Ellison. Tidsskr Nor Laegeforen 2019;139:
  6. Norton JA, Foster DS, Ito T , et al. Gastrinomas: Medical and SurgicalTreatment. Endocrinol Metab Clin North Am 2018;47:577-601
  7. Jensen RT. Gastrointestinal endocrine tumors.Gastrinoma. Bailliere's Clin Gastroenterol 1996;10:555-766
  8. Rehfeld JF, van Solinge WW. The tumor biology of gastrin and cholecystokinin. Adv Cancer Res 1994;63:295-347
  9. Jensen RT, Niederle B, Mitry E , et al. Gastrinoma (duodenal and pancreatic). Neuroendocrinology 2006;84:173-182
  10. Shaw DH. Gastrinoma (Zollinger-Ellison Syndrome) in the Dog and cat. Can Vet J 1988;29:448-452
  11. Hoenerhoff M, Kiupel M. Concurrent gastrinoma and somatostatinoma in a 10-year-old Portuguese water dog. J Comp Pathol 2004;130:313-318
  12. Brooks D, Watson GL. Omeprazole in a dog with gastrinoma. J Vet Intern Med 1997;11:379-381
  13. Green RA, Gartrell CL. Gastrinoma: a retrospective study of four cases (1985-1995). J Am Anim Hosp Assoc 1997;33:524-527
  14. Hayden DW, Henson MS. Gastrin-secreting pancreatic endocrine tumor in a dog (putative Zollinger-Ellison syndrome). J Vet Diagn Invest 1997;9:100-103
  15. Straus E, Johnson GF, Yalow RS. Canine Zollinger-Ellison syndrome. Gastroenterology 1977;72:380-381
  16. Fukushima U, Sato M, Okano S , et al. A case of gastrinoma in a Shih-Tzu dog. J Vet Med Sci 2004;66:311-313
  17. Hughes SM. Canine gastrinoma: a case study and literature review of therapeutic options. N Z Vet J 2006;54:242-247
  18. Patnaik AK, Lieberman PH, Erlandson RA , et al. Hepatobiliary neuroendocrine carcinoma in cats: a clinicopathologic, immunohistochemical, and ultrastructural study of 17 cases. Vet Pathol 2005;42:331-337
  19. van den Ingh ThS, Lamers CB, Lindeman J. Zollinger-Ellison syndrome in a cat. Vet Q 1988;10:151-155
  20. Middleton DJ, Watson AD, Vasak E , et al. Duodenal ulceration associated with gastrin-secreting pancreatic tumor in a cat. J Am Vet Med Assoc 1983;183:461-462
  21. Lane M, Larson J, Hecht S , et al. Medical management of gastrinoma in a cat. JFMS Open Rep 2016;2:2055116916646389
  22. Myers-Nodes J, Mazepa ASW. Combined surgical and medical management of a cat with gastrinoma. J Small Anim Pract 2022;63:632-634
  23. Struthers JD, Robl N, Wong VM , et al. Gastrinoma and Zollinger-Ellison syndrome in canids: a literature review and a case in a Mexican gray wolf. J Vet Diagn Invest 2018;30:584-588
  24. Ito T, Igarashi H, Jensen RT. Pancreatic neuroendocrine tumors: clinical features, diagnosis and medical treatment: Advances. Best Pract Res Clin Gastroenterol 2012;26:737-753
  25. Ito T, Lee L, Jensen RT. Treatment of symptomatic neuroendocrine tumor syndromes: recent advances and controversies. Expert Opin Pharmacother 2016;17:2191-2205
  26. Jensen RT, Norton JA, Oberg K. Neuroendocrine Tumors. In: Feldman M, Friedman LS, Brandt LJ eds. Sleisenger and Fordtran's Gastrointestinal and Liver Diseases. Philadelphia: Elsevier Saunders; 2016:501-541.
  27. Jensen RT, Gardner JD. Gastrinoma. In: Go VLW, DiMagno EP, Gardner JD et al.  eds. The Pancreas: Biology, Pathobiology and Disease. New York: Raven Press Publishing Co.; 1993:931-978.
  28. Jensen RT, Gardner JD, Raufman JP , et al. Zollinger-Ellison syndrome: current concepts and management. Ann Intern Med 1983;98:59-75
  29. Metz DC, Jensen RT. Gastrointestinal neuroendocrine tumors:; Pancreatic endocrine tumors. Gastroenterology 2008;135:1469-1492
  30. Jensen RT, Berna MJ, Bingham MD , et al. Inherited pancreatic endocrine tumor syndromes: advances in molecular pathogenesis, diagnosis, management and controversies. Cancer 2008;113(7 suppl):1807-1843
  31. Norton JA, Krampitz G, Zemek A , et al. Better Survival But Changing Causes of Death in Patients With Multiple Endocrine Neoplasia Type 1. Ann Surg 2015;261:e147-e148
  32. Zaidi MY, Lopez-Aguiar AG, Poultsides GA , et al. The impact of failure to achieve symptom control after resection of functional neuroendocrine tumors: An 8-institution study from the US Neuroendocrine Tumor Study Group. J Surg Oncol 2019;119:5-11
  33. Rossi RE, Elvevi A, Citterio D , et al. Gastrinoma and Zollinger Ellison syndrome: A roadmap for the management between new and old therapies. World J Gastroenterol 2021;27:5890-5907
  34. Bevere M, Gkountakos A, Martelli FM , et al. An Insight on Functioning Pancreatic Neuroendocrine Neoplasms. Biomedicines 2023;11:303
  35. Magi L, Marasco M, Rinzivillo M , et al. Management of Functional Pancreatic Neuroendocrine Neoplasms. Curr Treat Options Oncol 2023;24:725-741
  36. Hofland J, de Herder WW. Effective strategies for adequate control of hormonal secretion in functioning neuroendocrine neoplasms. Best Pract.Res Clin Endocrinol Metab , 101787. 2023.
  37. Knapp TG, Duan S, Merchant JL , et al. Quantitative characterization of duodenal gastrinoma autofluorescence using multiphoton microscopy. Lasers Surg Med 2023;55:208-225
  38. Rydzewska G, Strzelczyk J, Bednarczuk T , et al. Gastroduodenal neuroendocrine neoplasms including gastrinoma - update of the diagnostic and therapeutic guidelines (recommended by the Polish Network of Neuroendocrine Tumours). Endokrynol Pol 2022;73:455-490
  39. Creutzfeldt W, Arnold R, Creutzfeldt C , et al. Pathomorphologic, biochemical and diagnostic aspects of gastrinomas (Zollinger-Ellison syndrome). Hum Pathol 1975;6:47-76
  40. Jensen RT. Zollinger-Ellison syndrome. In: Doherty GM, Skogseid B eds. Surgical Endocrinology: Clinical Syndromes. Philadelphia: Lippincott Williams & Wilkins; 2001:291-344.
  41. Kaur A, Wang S, Herbert J , et al. Analysis of Clinical Characteristics and Survival in Patients With Functional Neuroendocrine Tumors of Gastrointestinal Origin. Pancreas 2022;51:1171-1178
  42. Clift AK, Thomas R, Frilling A. Developments in interventional management of hepatic metastases from neuroendocrine tumours. Best Pract Res Clin Endocrinol Metab 2023;101798
  43. Norton JA, Fraker DL, Alexander HR , et al. Surgery to cure the Zollinger-Ellison syndrome. N Engl J Med 1999;341:635-644
  44. Jensen RT, Norton JA. Treatment of Pancreatic Neuroendocrine Tumors in Multiple Endocrine Neoplasia Type 1: Some Clarity But Continued Controversy. Pancreas 2017;46:589-594
  45. Norton JA, Fraker DL, Alexander HR , et al. Value of surgery in patients with negative imaging and sporadic zollinger-ellison syndrome. Ann Surg 2012;256:509-517
  46. Ellison EC, Johnson JA. The Zollinger-Ellison syndrome: a comprehensive review of historical, scientific, and clinical considerations. Curr Probl Surg 2009;46:13-106
  47. Nieman LK. Cushing's syndrome: update on signs, symptoms and biochemical screening. Eur J Endocrinol 2015;173:M33-M38
  48. Wolin EM, Jensen RT. Neuroendocrine Tumors. In: Goldman L, Schaffer AI, Crow MK et al.  eds. GOLDMAN-CECIL MEDICINE. Philadelphia: Elsevier; 2020:1520-1528.
  49. Metz DC, Jensen RT. Zollinger-Ellison syndrome. In: Podoloff DA, Camilleri M, Fitz JGKAN et al.  eds. Yamada"s Textbook of Gastroenterogy. Chichester,West Sussex,UK: Wiley-Blackwell; 2020:In press.
  50. Jensen RT. Zollinger-Ellison syndrome. In: Podolsky DK, Camilleri M, Fitz JGKAN et al.  eds. Yamada's Textbook of Gastroenterology. West Sussex. UK: John Wiley and Sons,Ltd.; 2016:1078-1102.
  51. Ito T, Igarashi H, Jensen RT. Zollinger-Ellison syndrome: Recent advances and controversies. Current Opinion in Gastroenterology 2013;29:650-661
  52. Krampitz GW, Norton JA. Current management of the Zollinger-Ellison syndrome. Adv Surg 2013;47:59-79
  53. Chatzipanagiotou O, Schizas D, Vailas M , et al. All you need to know about gastrinoma today | Gastrinoma and Zollinger-Ellison syndrome: A thorough update. J Neuroendocrinol 2023;35:e13267
  54. Chang A, Sherman SK, Howe JR , et al. Progress in the Management of Pancreatic Neuroendocrine Tumors. Annu Rev Med 2022;73:213-229
  55. Metz DC, Cadiot G, Poitras P , et al. Diagnosis of Zollinger-Ellison syndrome in the era of PPIs, faulty gastrin assays, sensitive imaging and limited access to acid secretory testing. Int J Endocr Oncol 2017;4:167-185
  56. Ito T, Cadiot G, Jensen RT. Diagnosis of Zollinger-Ellison syndrome: Increasingly difficult. World J Gastroenterol 2012;18:5495-5503
  57. Singh Ospina N, Donegan D, Rodriguez-Gutierrez R , et al. Assessing for Multiple Endocrine Neoplasia Type 1 in Patients Evaluated for Zollinger-Ellison Syndrome-Clues to a Safer Diagnostic Process. Am J Med 2017;130:603-605
  58. Hofland J, Kaltsas G, de Herder WW. Advances in the diagnosis and management of well-differentiated neuroendocrine neoplasms. Endocr Rev 2019;41:371-403
  59. Metz DC. Diagnosis of the Zollinger-Ellison syndrome. Clin Gastroenterol Hepatol 2012;10:126-130
  60. Klimko A, Plotogea O, Constantinescu A , et al. Delayed Management of Zollinger-Ellison Syndrome in a Noncompliant Patient. Cureus 2020;12:e8471
  61. Bhattacharya S, Blau JE, Cochran C , et al. Validity of Secretin Stimulation Testing on Proton Pump Inhibitor Therapy for Diagnosis of Zollinger-Ellison Syndrome. Am J Gastroenterol 2021;116:2216-2221
  62. Roy PK, Venzon DJ, Shojamanesh H , et al. Zollinger-Ellison syndrome: clinical presentation in 261 patients. Medicine (Baltimore) 2000;79:379-411
  63. Soga J, Yakuwa Y. The gastrinoma/Zollinger-Ellison syndrome: statistical evaluation of a Japanese series of 359 cases. J Hep Bil Pancr Surg 1998;5:77-85
  64. Guo Y, Chen LH, Liu M , et al. (Comparison of clinical characteristics between sporadic gastrinoma and multiple endocrine neoplasia type 1-related gastrinoma). Zhonghua Wei Chang Wai Ke Za Zhi 2021;24:875-882
  65. Massironi S, Rossi RE, Laffusa A , et al. Sporadic and MEN1-related gastrinoma and Zollinger-Ellison syndrome: differences in clinical characteristics and survival outcomes. J Endocrinol Invest 2023;46:957-965
  66. Zheng M, Chen L, Nie X , et al. Pancreatic neuroendocrine tumor with ectopic adrenocorticotropic hormone syndrome: a case report and 5-year follow-up. Endocr J 2022;69:243-251
  67. Nakaoka K, Hashimoto S, Kawabe N , et al. A rare case of pancreatic neuroendocrine neoplasm causing Cushing's syndrome. Clin J Gastroenterol 2022;15:256-262
  68. Wydra A, Cylke-Falkowska K, Czajka-Oraniec I , et al. Severe ectopic Cushing syndrome in a transgender man with a metastatic gastrinoma and an adrenal tumor-A case report and review of the literature. Front Endocrinol (Lausanne) 2023;14:1135016
  69. Bahrin MH, Hagag R, Ali A , et al. Benign Oesophageal Stricture and Chronic Diarrhoea As Atypical Presenting Symptoms of an Advanced Metastatic Pancreatic Gastrinoma: A Case Report and Review of Literature. Cureus 2021;13:e16593
  70. Roy PK, Venzon DJ, Feigenbaum KM , et al. Gastric secretion in Zollinger-Ellison syndrome: correlation with clinical expression, tumor extent and role in diagnosis - A prospective NIH study of 235 patients and review of the literature in 984 cases. Medicine(Baltimore) 2001;80:189-222
  71. Perrier M, Delemer B, Deguelte S , et al. Total gastrectomy for severe proton pump inhibitor-induced hypomagnesemia in a MEN1/Zollinger Ellison syndrome patient. Pancreatology 2021;21:236-239
  72. Ito T, Ramos-Alvarez I, Jensen RT. Successful lifetime/lifelong medical treatment of acid in Zollinger-Ellison syndrome (ZES): Myth or fact? Insights from an analysis of results of NIH-long-term prospective studies of ZES. Cancers (Basel) 2023;15:1377
  73. Berna MJ, Hoffmann KM, Serrano J , et al. Serum gastrin in Zollinger-Ellison syndrome: I. Prospective study of fasting serum gastrin in 309 patients from the National Institutes of Health and comparison with 2229 cases from the literature. Medicine (Baltimore) 2006;85:295-330
  74. Berna MJ, Hoffmann KM, Long SH , et al. Serum gastrin in Zollinger-Ellison syndrome: II. Prospective study of gastrin provocative testing in 293 patients from the National Institutes of Health and comparison with 537 cases from the literature. evaluation of diagnostic criteria, proposal of new criteria, and correlations with clinical and tumoral features. Medicine (Baltimore) 2006;85:331-364
  75. Frucht H, Howard JM, Slaff JI , et al. Secretin and calcium provocative tests in the Zollinger-Ellison syndrome: A prospective study. Ann Intern Med 1989;111:713-722
  76. Lee L, Ramos-Alvarez I, Ito T , et al. Insights into Effects/Risks of Chronic Hypergastrinemia and Lifelong PPI Treatment in Man Based on Studies of Patients with Zollinger-Ellison Syndrome. Int J Mol Sci 2019;20:E5128
  77. Poitras P, Gingras MH, Rehfeld JF. The Zollinger-Ellison syndrome: dangers and consequences of interrupting antisecretory treatment. Clin Gastroenterol Hepatol 2012;10:199-202
  78. Banasch M, Schmitz F. Diagnosis and treatment of gastrinoma in the era of proton pump inhibitors. Wien Klin Wochenschr 2007;119:573-578
  79. Mendelson AH, Donowitz M. Catching the Zebra: Clinical Pearls and Pitfalls for the Successful Diagnosis of Zollinger-Ellison Syndrome. Dig Dis Sci 2017;62:2258-2265
  80. De Angelis C, Cortegoso VP, Venezia L , et al. Diagnosis and management of Zollinger-Ellison syndrome in 2018. Minerva Endocrinol 2018;43:212-220
  81. Murakami T, Usui T, Nakamoto Y , et al. Challenging Differential Diagnosis of Hypergastremia and Hyperglucagonemia with Chronic Renal Failure: Report of a Case with Multiple Endocrine Neoplasia Type 1. Intern Med 2017;56:1375-1381
  82. Yu S, Wang D, Ma X , et al. Analytical and Clinical Performance of a Liquid Chromatography-Tandem Mass Spectrometry Method for Measuring Gastrin Subtypes G34 and G17 in Serum. Clin Chem 2021;67:1220-1229
  83. Veysey-Smith R, Moore AR, Murugesan SV , et al. Effects of Proton Pump Inhibitor Therapy, H. pylori Infection and Gastric Preneoplastic Pathology on Fasting Serum Gastrin Concentrations. Front Endocrinol (Lausanne) 2021;12:741887
  84. Giusti F, Cioppi F, Fossi C , et al. Secretin Stimulation Test and Early Diagnosis of Gastrinoma in MEN1 Syndrome: Survey on the MEN1 Florentine Database. J Clin Endocrinol Metab 2022;107:e2110-e2123
  85. Endall R, Thompson M, Parameswaran V , et al. The Relationship of Gastrinoma in MEN 1 to Helicobacter pylori infection. J Clin Endocrinol Metab 2020;105:e 676-e682
  86. Hackeng WM, Dreijerink KMA, Offerhaus GJA , et al. A Parathyroid-Gut Axis: Hypercalcemia and the Pathogenesis of Gastrinoma in Multiple Endocrine Neoplasia 1. Mol Cancer Res 2021;19:946-949
  87. Gibril F, Schumann M, Pace A , et al. Multiple endocrine neoplasia type 1 and Zollinger-Ellison syndrome. A prospective study of 107 cases and comparison with 1009 patients from the literature. Medicine (Baltimore) 2004;83:43-83
  88. Norton JA, Krampitz G, Jensen RT. Multiple Endocrine Neoplasia: Genetics and Clinical Management. Surg Oncol Clin N Am 2015;24:795-832
  89. Ito T, Igarashi H, Uehara H , et al. Causes of Death and Prognostic Factors in Multiple Endocrine Neoplasia Type 1: A Prospective Study: Comparison of 106 MEN1/Zollinger-Ellison Syndrome Patients With 1613 Literature MEN1 Patients With or Without Pancreatic Endocrine Tumors. Medicine (Baltimore) 2013;92:135-181
  90. Ito T, Jensen RT. Imaging in multiple endocrine neoplasia type 1: recent studies show enhanced sensitivities but increased controversies. Int J Endocr Oncol 2016;3:53-66
  91. Mandl A, Welch JM, Kapoor G , et al. Two distinct classes of thymic tumors in patients with MEN1 show LOH at the MEN1 locus. Endocr Relat Cancer 2021;28:L15-L19
  92. Albers MB, Manoharan J, Bollmann C , et al. Results of Duodenopancreatic Reoperations in Multiple Endocrine Neoplasia Type 1. World J Surg 2019;43:552-558
  93. Vinault S, Mariet AS, Le BM , et al. Metastatic Potential and Survival of Duodenal and Pancreatic Tumors in Multiple Endocrine Neoplasia Type 1: A GTE and AFCE Cohort Study (Groupe d'etude des Tumeurs Endocrines and Association Francophone de Chirurgie Endocrinienne). Ann Surg 2020;272:1094-1101
  94. Febrero B, Segura P, Ruiz-Manzanera JJ , et al. Uncommon tumors in multiple endocrine neoplasia (MEN) type 1: Do they have a relationship with the prognosis of these patients? J Endocrinol Invest 2021;44:1327-1330
  95. Imamura M, Komoto I, Taki Y. How to treat gastrinomas in patients with multiple endocrine neoplasia type1: surgery or long-term proton pump inhibitors? Surgery Today . 2023. (In Press)
  96. Geslot A, Vialon M, Caron P , et al. New therapies for patients with multiple endocrine neoplasia type 1. Ann Endocrinol (Paris) 2021;82:112-120
  97. Mele C, Mencarelli M, Caputo M , et al. Phenotypes Associated With MEN1 Syndrome: A Focus on Genotype-Phenotype Correlations. Front Endocrinol (Lausanne) 2020;11:591501
  98. Naruoka A, Ohnami S, Nagashima T , et al. Genomic profiling of multiple tissues in two patients with multiple endocrine neoplasia type 1. Biomed Res 2021;42:89-94
  99. Santucci N, Gaujoux S, Binquet C , et al. Pancreatoduodenectomy for Neuroendocrine Tumors in Patients with Multiple Endocrine Neoplasia Type 1: An AFCE (Association Francophone de Chirurgie Endocrinienne) and GTE (Groupe d'étude des Tumeurs Endocrines) Study. World J Surg 2021;45:1794-1802
  100. Gong S, Li Z, Liu XB , et al. Gastrinoma in multiple endocrine neoplasia type 1 after total pancreatectomy: A case report. Medicine (Baltimore) 2019;98:e18275
  101. Shariq OA, Lines KE, English KA , et al. Multiple endocrine neoplasia type 1 in children and adolescents: Clinical features and treatment outcomes. Surgery 2022;171:77-87
  102. Kong W, Albers MB, Manoharan J , et al. Pancreaticoduodenectomy Is the Best Surgical Procedure for Zollinger-Ellison Syndrome Associated with Multiple Endocrine Neoplasia Type 1. Cancers (Basel) 2022;14:1928
  103. Ito T, Igarashi H, Uehara H , et al. Pharmacotherapy  of Zollinger-Ellison syndrome. Expert Opin Pharmacotherapy 2013;14:307-321
  104. Collen MJ, Howard JM, McArthur KE , et al. Comparison of ranitidine and cimetidine in the treatment of gastric hypersecretion. Ann Intern Med 1984;100:52-58
  105. Metz DC, Pisegna JR, Fishbeyn VA , et al. Control of gastric acid hypersecretion in the management of patients with Zollinger-Ellison syndrome. World J Surg 1993;17:468-480
  106. Raufman JP, Collins SM, Pandol SJ , et al. Reliability of symptoms in assessing control of gastric acid secretion in patients with Zollinger-Ellison syndrome. Gastroenterology 1983;84:108-113
  107. Kusano K, Uno K, Koike T , et al. A case of refractory esophageal stricture due to occult gastrinoma of the duodenum. Clin J Gastroenterol 2022;15:859-863
  108. Martino BR, Manibusan P. Zollinger Ellison Syndrome Refractory to Medical Therapy in the Setting of Multiple Endocrine Neoplasia Type I. Cureus 2022;14:e26468
  109. Yu F, Venzon DJ, Serrano J , et al. Prospective study of the clinical course, prognostic factors and survival in patients with longstanding Zollinger-Ellison syndrome. J Clin Oncol 1999;17:615-630
  110. Weber HC, Venzon DJ, Lin JT , et al. Determinants of metastatic rate and survival in patients with Zollinger-Ellison syndrome: a prospective long-term study. Gastroenterology 1995;108:1637-1649
  111. Gibril F, Venzon DJ, Ojeaburu JV , et al. Prospective study of the natural history of gastrinoma in patients with MEN1: Definition of an aggressive and a nonaggressive form. J Clin Endocrinol Metab 2001;86:5282-5293
  112. Gibril F, Doppman JL, Reynolds JC , et al. Bone metastases in patients with gastrinomas: a prospective study of bone scanning, somatostatin receptor scanning, and MRI in their detection, their frequency, location and effect of their detection on management. J Clin Oncol 1998;16:1040-1053
  113. Maton PN, Gardner JD, Jensen RT. Cushing's syndrome in patients with Zollinger-Ellison syndrome. N Engl J Med 1986;315:1-5
  114. Hackeng WM, van Beek DJ, Kok ASM , et al. Metastatic Patterns of Duodenopancreatic Neuroendocrine Tumors in Patients With Multiple Endocrine Neoplasia Type 1. Am J Surg Pathol 2022;46:159-168
  115. Lee L, Ramos-Alvarez I, Jensen RT. Predictive Factors for Resistant Disease with Medical/Radiologic/Liver-Directed Anti-Tumor Treatments in Patients with Advanced Pancreatic Neuroendocrine Neoplasms: Recent Advances and Controversies. Cancers (Basel) 2022;14:1250
  116. Lee L, Ito T, Jensen RT. Prognostic and predictive factors on overall survival and surgical outcomes in pancreatic neuroendocrine tumors: recent advances and controversies. Expert Rev Anticancer Ther 2019;19:1029-1050
  117. Matrood S, Melms LE, Bartsch DK , et al. The Expression of Autophagy-Associated Genes Represents a Valid Footprint for Aggressive Pancreatic Neuroendocrine Neoplasms. Int J Mol Sci 2023;24:3636
  118. Bartsch DK, Albers MB. Controversies in surgery for multiple endocrine neoplasia type-1- associated Zollinger-Ellison syndrome. Int J Endo Oncol 2015;2:263-271
  119. Norton JA, Jensen RT. Resolved and unresolved controversies in the surgical management of patients with Zollinger-Ellison syndrome. Ann Surg 2004;240:757-773
  120. Souche R, Hobeika C, Hain E , et al. Surgical Management of Neuroendocrine Tumours of the Pancreas. J Clin Med 2020;9:2993
  121. Sampathirao N, Basu S. Rare Occurrence Of Hypergastrinemia due to Thoracic Neuroendocrine Tumor: Detection and Characterization by 68Ga-DOTATATE PET-CT. J Nucl.Med.Technol.  2016.
  122. Shao QQ, Zhao BB, Dong LB , et al. Surgical management of Zollinger-Ellison syndrome: Classical considerations and current controversies. World J Gastroenterol 2019;25:4673-4681
  123. Sauvanet A. Gastroenteropancreatic neuroendocrine tumors: Role of surgery. Ann Endocrinol (Paris) 2019;80:175-181
  124. Cockburn KC, Toumi Z, Mackie A , et al. Radioguided Surgery for Gastroenteropancreatic Neuroendocrine Tumours: a Systematic Literature Review. J Gastrointest Surg 2021;25:3244-3257
  125. Okasha HH, Aboubakr A, Elenin SA , et al. Unexpected adverse events of endoscopic polypectomy of a juxtapapillay duodenal gastrinoma. Gastrointest Endosc 2022;95:589-590
  126. Okamoto T, Yoshimoto T, Ohike N , et al. Spontaneous regression of gastric gastrinoma after resection of metastases to the lesser omentum: A case report and review of literature. World J Gastroenterol 2021;27:129-142
  127. Robin L, Sauvanet A, Walter T , et al. Recurrence after surgical resection of nonmetastatic sporadic gastrinoma: Which prognostic factors and surgical procedure? Surgery 2023;173:1144-1152
  128. Titan AL, Norton JA, Fisher AT , et al. Evaluation of Outcomes Following Surgery for Locally Advanced Pancreatic Neuroendocrine Tumors. JAMA Netw Open 2020;3:e2024318
  129. Lee L, Ito T, Jensen RT. Imaging of pancreatic neuroendocrine tumors: recent advances, current status and controversies. Expert Rev Anticancer Ther 2018;18:837-860
  130. Krudy AG, Doppman JL, Jensen RT , et al. Localization of islet cell tumors by dynamic CT: Comparison with plain CT, arteriography, sonography and venous sampling. Am J Roentgenol 1984;143:585-589
  131. Maton PN, Miller DL, Doppman JL , et al. Role of selective angiography in the management of Zollinger- Ellison syndrome. Gastroenterology 1987;92:913-918
  132. Frucht H, Doppman JL, Norton JA , et al. Gastrinomas: Comparison of MR Imaging with CT, angiography and US. Radiology 1989;171:713-717
  133. Gibril F, Reynolds JC, Doppman JL , et al. Somatostatin receptor scintigraphy: its sensitivity compared with that of other imaging methods in detecting primary and metastatic gastrinomas: a prospective study. Ann Intern Med 1996;125:26-34
  134. Ito T, Jensen RT. Molecular imaging in neuroendocrine tumors: recent advances, controversies, unresolved issues, and roles in management. Curr Opin Endocrinol Diabetes Obes 2017;24:15-24
  135. Desai H, Borges-Neto S, Wong TZ. Molecular Imaging and Therapy for Neuroendocrine Tumors. Curr Treat Options Oncol 2019;20:78
  136. Lui J, Terra SBSP, de la FJ. Metastatic Gastrinoma Localized on Gallium-68 DOTATATE Imaging. Mayo Clin Proc 2021;96:285-286
  137. Stolniceanu CR, Grierosu IC, Matovic M , et al. Somatostatin receptor molecular imaging in a misdiagnosed gastrinoma case. World J Nucl Med 2020;19:417-420
  138. Higashino N, Sonomura T, Ito D , et al. Usefulness of computed tomography during arteriography in a selective arterial calcium agent injection test for pancreatic gastrinoma: a case description. Quant Imaging Med Surg 2021;11:3898-3903
  139. Jayasekera M, Sartin S, Bhargava P. Ga-68 DOTATATE PET/CT in a patient with Zollinger-Ellison syndrome. Radiol Case Rep 2023;18:1046-1048
  140. Pellegrino F, Granata V, Fusco R , et al. Diagnostic Management of Gastroenteropancreatic Neuroendocrine Neoplasms: Technique Optimization and Tips and Tricks for Radiologists. Tomography 2023;9:217-246
  141. Duan H, Iagaru A. Neuroendocrine Tumor Diagnosis: PET/MR Imaging. PET Clin 2023;18:259-266
  142. Ito T, Igarashi H, Jensen RT. Therapy of metastatic pancreatic neuroendocrine tumors (pNETs): recent insights and advances. J Gastroenterol 2012;47:941-960
  143. Pavel M, O'Toole D, Costa F , et al. ENETS Consensus Guidelines Update for the Management of Distant Metastatic Disease of Intestinal, Pancreatic, Bronchial Neuroendocrine Neoplasms (NEN) and NEN of Unknown Primary Site. Neuroendocrinology 2016;103:172-185
  144. Akirov A, Larouche V, Alshehri S , et al. Treatment Options for Pancreatic Neuroendocrine Tumors. Cancers (Basel) 2019;11:828
  145. Dimitriadis GK, Weickert MO, Randeva HS , et al. Medical management of secretory syndromes related to gastroenteropancreatic neuroendocrine tumours. Endocr Relat Cancer 2016;23:R423-R436
  146. Gut P, Waligorska-Stachura J, Czarnywojtek A , et al. Management of the hormonal syndrome of neuroendocrine tumors. Arch Med Sci 2017;13:515-524
  147. von Schrenck T, Howard JM, Doppman JL , et al. Prospective study of chemotherapy in patients with metastatic gastrinoma. Gastroenterology 1988;94:1326-1334
  148. Zandee WT, Brabander T, Blazevic A , et al. Symptomatic and Radiological Response to 177Lu-DOTATATE for the Treatment of Functioning Pancreatic Neuroendocrine Tumors. J Clin Endocrinol Metab 2019;104:1336-1344
  149. Halfdanarson TR, Strosberg JR, Tang L , et al. The North American Neuroendocrine Tumor Society Consensus Guidelines for Surveillance and Medical Management of Pancreatic Neuroendocrine Tumors. Pancreas 2020;49:863-881
  150. Lee L, Ito T, Jensen RT. Everolimus in the treatment of neuroendocrine tumors: efficacy, side-effects, resistance, and factors affecting its place in the treatment sequence. Expert Opin Pharmacother 2018;19:909-928
  151. Guarnotta V, Martini C, Davi MV , et al. The Zollinger-Ellison syndrome: is there a role for somatostatin analogues in the treatment of the gastrinoma? Endocrine 2018;60:15-27
  152. Shah MH, Goldner WS, Halfdanarson TR , et al. NCCN Guidelines Insights: Neuroendocrine and Adrenal Tumors, Version 2.2018. J Natl Compr Canc Netw 2018;16:693-702
  153. Patil VA, Goroshi MR, Shah H , et al. Comparison of (68)Ga-DOTA-NaI(3)-Octreotide/tyr(3)-octreotate positron emission tomography/computed tomography and contrast-enhanced computed tomography in localization of tumors in multiple endocrine neoplasia 1 syndrome. World J Nucl Med 2020;19:99-105
  154. Hofland J, Brabander T, Verburg FA , et al. Peptide Receptor Radionuclide Therapy. J Clin Endocrinol Metab 2022;107:3199-3208
  155. Massironi S, Cavalcoli F, Elvevi A , et al. Somatostatin analogs in patients with Zollinger Ellison syndrome (ZES): an observational study. Endocrine 2022;75:942-948
  156. Ito T, Masui T, Komoto I , et al. JNETS clinical practice guidelines for gastroenteropancreatic neuroendocrine neoplasms: diagnosis, treatment, and follow-up: a synopsis. J Gastroenterol 2021;56:1033-1044
  157. Falconi M, Eriksson B, Kaltsas G , et al. ENETS Consensus Guidelines Update for the Management of Patients with Functional Pancreatic Neuroendocrine Tumors and Non-Functional Pancreatic Neuroendocrine Tumors. Neuroendocrinology 2016;103:153-171
  158. Jensen RT. Neuroendocrine Tumors of the Gastrointestintal Tract (GI) and Pancreas. In: Jamieson LL, Fauci AS, Kasper DL et al.  eds. Harrison's Principles of Internal Medicine-ED.20. New York, New York: McGraw Hill Education Medical Publishing Division; 2018:596-615.
  159. Del Olmo-Garcia MI, Muros MA, Lopez-de-la-Torre M , et al. Prevention and Management of Hormonal Crisis during Theragnosis with LU-DOTA-TATE in Neuroendocrine Tumors. A Systematic Review and Approach Proposal. J Clin Med 2020;9:2203
  160. Wolin EM, Benson III AB. Systemic Treatment Options for Carcinoid Syndrome: A Systematic Review. Oncology 2019;96:273-289
  161. Pavel M, Oberg K, Falconi M , et al. Gastroenteropancreatic neuroendocrine neoplasms: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol 2020;31:844-860
  162. Tsoli M, Alexandraki K, Xanthopoulos C , et al. Medical Treatment of Gastrointestinal Neuroendocrine Neoplasms. Horm Metab Res 2020;52:614-620
  163. Murakami M, Fujimori N, Matsumoto K , et al. A clinical analysis on functioning pancreatic neuroendocrine tumors (focusing on VIPomas): a single-center experience. Endocr J 2022;69:1201-1209
  164. Ito T, Jensen RT. Perspectives on the Current Pharmacotherapeutic Strategies for Management of Functional Neuroendocrine Tumor Syndromes. Expert Opin Pharmacother 2021;22:685-693
  165. Marchese U, Gaillard M, Pellat A , et al. Multimodal Management of Grade 1 and 2 Pancreatic Neuroendocrine Tumors. Cancers (Basel) 2022;14:433
  166. Klimstra DS. Pathologic Classification of Neuroendocrine Neoplasms. Hematol Oncol Clin North Am 2016;30:1-19
  167. Inzani F, Petrone G, Rindi G. The New World Health Organization Classification for Pancreatic Neuroendocrine Neoplasia. Endocrinol Metab Clin North Am 2018;47:463-470
  168. Klimstra DS, La Rosa S, Rindi G. Classification of Neuroendocrine Neopasms of the digestive system. In: WHO Classsification of Tumors Editorial Board ed. WHO classification of tumours:digestive system tumours. Lyon, France: IARC Press; 2019:16-19.
  169. Kasajima A, Kloppel G. Neuroendocrine neoplasms of lung, pancreas and gut: a morphology-based comparison. Endocr Relat Cancer 2020;27:R417-R432
  170. Duan S, Rico K, Merchant JL. Gastrin: From Physiology to Gastrointestinal Malignancies. Function (Oxf) 2022;3:zqab062
  171. Maekawa A, Kudo A, Kishino M , et al. Hormonal tumor mapping for liver metastases of gastroenteropancreatic neuroendocrine neoplasms: a novel therapeutic strategy. J Cancer Res Clin Oncol 2022;148:697-706
  172. Rico K, Duan S, Pandey RL , et al. Genome analysis identifies differences in the transcriptional targets of duodenal versus pancreatic neuroendocrine tumours. BMJ Open Gastroenterol 2021;8:e000765
  173. Duan S, Sawyer TW, Sontz RA , et al. GFAP-directed Inactivation of Men1 Exploits Glial Cell Plasticity in Favor of Neuroendocrine Reprogramming. Cell Mol Gastroenterol Hepatol 2022;14:1025-1051
  174. Elvis-Offiah UB, Duan S, Merchant JL. MENIN-mediated regulation of gastrin gene expression and its role in gastrinoma development. FASEB J 2023;37:e22913
  175. Norton JA, Alexander HR, Fraker DL , et al. Does the use of routine duodenotomy (DUODX) affect rate of cure, development of liver metastases or survival in patients with Zollinger-Ellison syndrome (ZES)? Ann Surg 2004;239:617-626
  176. Norton JA, Doppman JL, Jensen RT. Curative resection in Zollinger-Ellison syndrome: Results of a 10-year prospective study. Ann Surg 1992;215:8-18
  177. Howard TJ, Zinner MJ, Stabile BE , et al. Gastrinoma excision for cure. A prospective analysis. Ann Surg 1990;211:9-14
  178. Pipeleers-Marichal M, Somers G, Willems G , et al. Gastrinomas in the duodenums of patients with multiple endocrine neoplasia type 1 and the Zollinger-Ellison syndrome. N Engl J Med 1990;322:723-727
  179. Anlauf M, Garbrecht N, Henopp T , et al. Sporadic versus hereditary gastrinomas of the duodenum and pancreas: distinct clinico-pathological and epidemiological features. World J Gastroenterol 2006;12:5440-5446
  180. Kunz PL, Reidy-Lagunes D, Anthony LB , et al. Consensus Guidelines for the Management and Treatment of Neuroendocrine Tumors. Pancreas 2013;42:557-577
  181. Jensen RT, Cadiot G, Brandi ML , et al. ENETS Consensus Guidelines for the Management of Patients with Digestive Neuroendocrine Neoplasms: Functional Pancreatic Endocrine Tumor Syndromes. Neuroendocrinology 2012;95:98-119
  182. Kulke MH, Anthony LB, Bushnell DL , et al. NANETS Treatment Guidelines: Well-Differentiated Neuroendocrine Tumors of the Stomach and Pancreas. Pancreas 2010;39:735-752
  183. Phan J, Benhammou JN, Pisegna JR. Gastric Hypersecretory States: Investigation and Management. Curr Treat Options Gastroenterol 2015;13:386-397
  184. Osefo N, Ito T, Jensen RT. Gastric Acid hypersecretory States: recent insights and advances. Curr Gastroenterol Rep 2009;11:433-441
  185. Ito T, Jensen RT. Neuroendocrine Neoplasms and Functional Syndromes. In: Weber HC ed. Gastrointestinal Endocrinology. New York: Springer; 2023(In Press).
  186. Grant CS. Insulinoma. Best Pract Res Clin Gastroenterol 2005;19:783-798
  187. Bansal N, Weinstock RS. Non-Diabetic Hypoglycemia. Endotext(Internet) 2000;
  188. Maggio I, Mollica V, Brighi N , et al. The functioning side of the pancreas: a review on insulinomas. J Endocrinol Invest 2020;43:139-148
  189. O'Toole D, Kianmanesh R, Caplin M. ENETS 2016 Consensus Guidelines for the Management of Patients with Digestive Neuroendocrine Tumors: An Update. Neuroendocrinology 2016;103:113-195
  190. Jensen RT. Management of the Zollinger-Ellison syndrome in patients with multiple endocrine neoplasia type 1. J Intern Med 1998;243:477-488
  191. Norton JA, Alexander HR, Fraker DL , et al. Comparison of surgical results in patients with advanced and limited disease with multiple endocrine neoplasia type 1 and Zollinger-Ellison syndrome. Ann Surg 2001;234:495-506
  192. Dasari A, Shen C, Halperin D , et al. Trends in the Incidence, Prevalence, and Survival Outcomes in Patients With Neuroendocrine Tumors in the United States. JAMA Oncol 2017;3:1335-1342
  193. Yao JC, Eisner MP, Leary C , et al. Population-based study of islet cell carcinoma. Ann Surg Oncol 2007;14:3492-3500
  194. Yao JC, Hassan M, Phan A , et al. One hundred years after "carcinoid": epidemiology of and prognostic factors for neuroendocrine tumors in 35,825 cases in the United States. J Clin Oncol 2008;26:3063-3072
  195. Geurts JL. Inherited syndromes involving pancreatic neuroendocrine tumors. J Gastrointest Oncol 2020;11:559-566
  196. Glasker S, Vergauwen E, Koch CA , et al. Von Hippel-Lindau Disease: Current Challenges and Future Prospects. Onco Targets Ther 2020;13:5669-5690
  197. Carrera S, Sancho A, Azkona E , et al. Hereditary pancreatic cancer: related syndromes and clinical perspective. Hered Cancer Clin Pract 2017;15:9
  198. Sala Hernandez A, Montalva Oron EM, Pareja Ibars E , et al. Management of pancreatic gastrinoma associated with Von Hippel-Lindau disease: a case report. Rev Esp Enferm Dig 2016;109:
  199. Alshikho MJ, Noureldine SI, Talas JM , et al. Zollinger-Ellison Syndrome Associated with von Recklinghausen Disease: Case Report and Literature Review. Am J Case Rep 2016;17:398-405
  200. Schwarzkopf G, Pfisterer J. Metastasizing gastrinoma and tuberous sclerosis complex. Zentralbl Pathol 1994;139:477-481
  201. Chagnon JP, Barge J, Henin D , et al. (Recklinghausen's disease with digestive localizations associated with gastric acid hypersecretion suggesting Zollinger-Ellison syndrome). Gastroenterol Clin Biol 1985;9(1):65-69
  202. Massironi S, Zilli A, Rossi RE , et al. Gastrinoma and neurofibromatosis type 2: the first case report and review of the literature. BMC Gastroenterol 2014;14:110
  203. Arif AA, Kim PTW, Melck A , et al. Pancreatic Gastrinoma, Gastrointestinal Stromal Tumor (GIST), Pheochromocytoma, and Hürthle Cell Neoplasm in a Patient with Neurofibromatosis Type 1: A Case Report and Literature Review. Am J Case Rep 2021;22:e927761
  204. Gauci J, Azzopardi N, Babic D , et al. Neurofibromatosis Type 1: A Rare Predisposition for Gastrinomas and Other Neuroendocrine Tumors. Pancreas 2022;51:559-562
  205. Ellison EH, Wilson SD. The Zollinger-Ellison syndrome: Re-appraisal and evaluation of 260 registered cases. Ann Surg 1964;160:512-530
  206. Stage JG, Stadil F. The clinical diagnosis of the Zollinger-Ellison syndrome. Scand J Gastroenterol Suppl 1979;53:79-91
  207. Thompson JC, Lewis BG, Wiener I , et al. The role of surgery in the Zollinger-Ellison syndrome. Ann Surg 1983;197(5):594-607
  208. Schubert ML. Hormonal regulation of gastric acid secretion. Curr Gastroenterol Rep 2008;10:523-527
  209. Aoyagi T, Summerskill WH. Gastric secretion with ulcerogenic islet cell tumor. Importance of basal acid output. Arch Intern Med 1966;117:667-672
  210. Way L, Goldman L, Dunphy JE. Zollinger-Ellison syndrome. An analysis of twenty-five cases. Am J Surg 1968;116(2):293-304
  211. Richardson CT, Peters MN, Feldman M , et al. Treatment of Zollinger-Ellison syndrome with exploratory laparotomy, proximal gastric vagotomy, and H2-receptor antagonists. A prospective study. Gastroenterology 1985;89:357-367
  212. Jensen RT. Gastrinoma as a model for prolonged hypergastrinemia in man. In: Walsh JH ed. Gastrin. New York, NY: Raven Press Publishing Co.; 1993:373-393.
  213. Willems G. Trophic action of gastrin on specific target cells in the gut. In: Mignon M, Jensen RT eds. Endocrine Tumors of the Pancreas: Recent Advances in Research and Management. Series: Frontiers of Gastrointestinal Research. Basel, Switzerland: S. Karger; 1995:30-44.
  214. Hakanson R, Oscarson J, Sundler F. Gastrin and the trophic control of gastric mucosa. Scand J Gastroenterol 1986;Suppl 118:18-30
  215. Hakanson R, Sundler FH. Regulation of gastric endocrine cell proliferation. In: Walsh JH ed. Gastrin. New York: Raven Press, Ltd.; 1993:307-318.
  216. Hakanson R, Ekelund M, Sundler F. Activation and proliferation of gastric endocrine cells. In: Falkmer S, Hakanson R, Sundler F eds. Evolution and tumor pathology of the neuroendocrine system. Amsterdam: Elsevier; 1984:371-398.
  217. Peghini PL, Annibale B, Azzoni C , et al. Effect of chronic hypergastrinemia on human enterochromaffin-like cells: insights from patients with sporadic gastrinomas. Gastroenterology 2002;123:68-85
  218. Neuburger P, Lewin M, Recherche Cd , et al. Parietal and chief cell population in four cases of the Zollinger-Ellison syndrome. Gastroenterology 1972;63:937-942
  219. Polacek MA, Ellison EH. Parietal cell mass and gastric acid secretion in the Zollinger-Ellison syndrome. Surgery 1966;60:606-614
  220. Isenberg JI, Walsh JH, Grossman MI. Zollinger-Ellison syndrome. Gastroenterology 1973;65:140-165
  221. Plockinger U, Rindi G, Arnold R , et al. Guidelines for the diagnosis and treatment of neuroendocrine gastrointestinal tumours. A consensus statement on behalf of the European Neuroendocrine Tumour Society (ENETS). Neuroendocrinology 2004;80:394-424
  222. Jensen RT. Overview of chronic diarrhea caused by functional neuroendocrine neoplasms. Semin Gastrointest Dis 1999;10:156-172
  223. D'Adda T, Corleto V, Pilato FP , et al. Quantitative ultrastructure of endocrine cells of oxyntic mucosa in Zollinger-Ellison syndrome. Correspondence with light microscopic findings. Gastroenterology 1990;99:17-26
  224. Maton PN, Lack EE, Collen MJ , et al. The effect of Zollinger-Ellison syndrome and omeprazole therapy on gastric oxyntic endocrine cells. Gastroenterology 1990;99:943-950
  225. Lehy T, Mignon M, Cadiot G , et al. Gastric endocrine cell behavior in Zollinger-Ellison patients upon long-term potent antisecretory treatment. Gastroenterology 1989;96:1029-1040
  226. Creutzfeldt W. The achlorhydria-carcinoid sequence: role of gastrin. Digestion 1988;39:61-79
  227. Berna MJ, Annibale B, Marignani M , et al. A prospective study of gastric carcinoids and enterochromaffin-like cells changes in Multple Endocrine Neoplaisa Type 1 and Zollinger-Ellison syndrome: Identification of risk factors. J Clin Endocrinol Metab 2008;93:1582-1591
  228. Solcia E, Bordi C, Creutzfeldt W , et al. Histopathological classification of nonantral gastric endocrine growths in man. Digestion 1988;41:185-200
  229. Rindi G, Luinetti O, Cornaggia M , et al. Three subtypes of gastric argyrophil carcinoid and the gastric neuroendocrine carcinoma: a clinicopathologic study. Gastroenterology 1993;104:994-1006
  230. Cadiot G, Lehy T, Ruszniewski P , et al. Gastric endocrine cell evolution in patients with Zollinger-Ellison syndrome. Influence of gastrinoma growth and long-term omeprazole treatment. Dig Dis Sci 1993;38:1307-1317
  231. Lehy T, Cadiot G, Mignon M , et al. Influence of multiple endocrine neoplasia type 1 on gastric endocrine cells in patients with the Zollinger-Ellison syndrome. Gut 1992;33:1275-1279
  232. Feurle GE. Argyrophil cell hyperplasia and a carcinoid tumour in the stomach of a patient with sporadic Zollinger-Ellison syndrome. Gut 1994;35:275-277
  233. Cadiot G, Vissuzaine C, Potet F , et al. Fundic argyrophil carcinoid tumor in a patient with sporadic-type Zollinger-Ellison syndrome. Dig Dis Sci 1995;40:1275-1278
  234. Rais R, Trikalinos NA, Liu J , et al. Enterochromaffin-like Cell Hyperplasia-Associated Gastric Neuroendocrine Tumors May Arise in the Setting of Proton Pump Inhibitor Use. Arch Pathol Lab Med 2022;146:366-371
  235. Jensen RT, Doppman JL, Gardner JD. Gastrinoma. In: Go VLW, Brooks FA, DiMagno EP et al.  eds. The Exocrine Pancreas: Biology, Pathobiology and Disease. New York: Raven Press; 1986:727-744.
  236. Kloppel G, Anlauf M. Gastrinoma - morphological aspects. Wien Klin Wochenschr 2007;119:579-584
  237. Zollinger RM, Ellison EC, Fabri PJ , et al. Primary peptic ulcerations of the jejunum associated with islet cell tumors. Twenty-five-year appraisal. Ann Surg 1980;192:422-430
  238. Deveney CW, Deveney KS, Stark D , et al. Resection of gastrinomas. Ann Surg 1983;198:546-553
  239. Oberhelman HA, Jr., Nelsen TS, Johnson AN , et al. Ulcerogenic tumors of the duodenum. Ann Surg 1961;153:214-227
  240. Hofmann JW, Fox PS, Wilson SD. Duodenal wall tumors and the Zollinger-Ellison syndrome.  Surgical management. Arch Surg 1973;107:334-339
  241. Norton JA, Doppman JL, Collen MJ , et al. Prospective study of gastrinoma localization and resection in patients with Zollinger-Ellison syndrome. Ann Surg 1986;204:468-479
  242. Raufman JP, Kasbekar DK, Jensen RT , et al. Potentiation of pepsinogen secretion from dispersed glands from rat stomach. Am J Physiol 1983;245:G525-G530
  243. Thompson NW. Current concepts in the surgical management of multiple endocrine neoplasia type 1 pancreatic-duodenal disease. Results in the treatment of 40 patients with Zollinger-Ellison syndrome, hypoglycaemia or both. J Intern Med 1998;243:495-500
  244. Sugg SL, Norton JA, Fraker DL , et al. A prospective study of intraoperative methods to diagnose and resect duodenal gastrinomas. Ann Surg 1993;218:138-144
  245. Cadiot G, Lebtahi R, Sarda L , et al. Preoperative detection of duodenal gastrinomas and peripancreatic lymph nodes by somatostatin receptor scintigraphy. Gastroenterology 1996;111:845-854
  246. Thompson NW, Vinik AI, Eckhauser FE. Microgastrinomas of the duodenum. A cause of failed operations for the Zollinger-Ellison syndrome. Ann Surg 1989;209:396-404
  247. Frucht H, Norton JA, London JF , et al. Detection of duodenal gastrinomas by operative endoscopic transillumination: a prospective study. Gastroenterology 1990;99:1622-1627
  248. Thompson NW, Pasieka J, Fukuuchi A. Duodenal gastrinomas, duodenotomy, and duodenal exploration in the surgical management of Zollinger-Ellison syndrome. World J Surg 1993;17:455-462
  249. Norton JA, Foster DS, Blumgart LH , et al. Incidence and Prognosis of Primary Gastrinomas in the Hepatobiliary Tract. JAMA Surg 2018;153:e175083
  250. Norton JA, Harris EJ, Chen Y , et al. Pancreatic endocrine tumors with major vascular abutment, involvement, or encasement and indication for resection. Arch Surg 2011;146:724-732
  251. Moraes AB, Treistman N, Studart MC , et al. Gastrinoma of Cystic Duct: A Rare Association With Multiple Endocrine Neoplasia Type 1. J Clin Med Res 2018;10:843-847
  252. Pipek LZ, Jardim YJ, de Mesquita GHA , et al. Large primary hepatic gastrinoma in young patient treated with trisegmentectomy: A case report and review of the literature. World J Hepatol 2018;10:517-522
  253. Wu PC, Alexander HR, Bartlett DL , et al. A prospective analysis of the frequency, location, and curability of ectopic (non-pancreaticoduodenal, non-nodal) gastrinoma. Surgery 1997;122:1176-1182
  254. Norton JA, Krampitz GW, Poultsides GA , et al. Prospective Evaluation of Results of Reoperation in Zollinger-Ellison Syndrome. Ann Surg 2018;267:782-788
  255. Gibril F, Curtis LT, Termanini B , et al. Primary cardiac gastrinoma causing Zollinger-Ellison syndrome. Gastroenterology 1997;112:567-574
  256. Abou-Saif A, Lei J, McDonald TJ , et al. A new cause of Zollinger-Ellison syndrome: non-small cell lung cancer. Gastroenterology 2001;120:1271-1278
  257. Gibril F, Jensen RT. Advances in evaluation and management of gastrinoma in patients with Zollinger-Ellison syndrome. Curr Gastroenterol Rep 2005;7:114-121
  258. Norton JA, Alexander HA, Fraker DL , et al. Possible primary lymph node gastrinomas: occurrence, natural history and predictive factors: A prospective study. Ann Surg 2003;237:650-659
  259. Arnold WS, Fraker DL, Alexander HR , et al. Apparent lymph node primary gastrinoma. Surgery 1994;116:1123-1130
  260. Ito T, Jensen RT. Primary hepatic gastrinoma: an unusual case of zollinger-ellison syndrome. Gastroenterol Hepatol (N Y ) 2010;6:57-59
  261. Tonelli F, Giudici F, Nesi G , et al. Biliary tree gastrinomas in multiple endocrine neoplasia type 1 syndrome. World J Gastroenterol 2013;19:8312-8320
  262. Luiso D, Zuccarino F, Tizon-Marcos H , et al. Primary intracardiac gastrinoma causing Zollinger-Ellison syndrome. Eur Heart J 2019;41:3376
  263. Rafieian S, Vahedi M, Jahanbin B , et al. Primary thoracic gastrinoma causing Zollinger-Ellison syndrome. Indian J Thorac Cardiovasc Surg 2021;37:706-709
  264. Gonzalez-Lopez R, Ramlrez-Castaneda J, Ortega-Jimenez JA , et al. Primary hepatic gastrinoma, extremely rare case presentation and its surgical resolution, in a third level hospital in Mexico. Cir Cir 2022;90:109-113
  265. Rosiek V. Sequential therapies in gastric gastrinoma. Endokrynol Pol 2022;73:171-172
  266. Perrier ND, Batts KP, Thompson GB , et al. An immunohistochemical survey for neuroendocrine cells in regional pancreatic lymph nodes: a plausible explanation for primary nodal gastrinomas? Surgery 1995;118:957-965
  267. Atema JJ, Amri R, Busch OR , et al. Surgical treatment of gastrinomas: a single-centre experience. HPB (Oxford) 2012;14:833-838
  268. Anlauf M, Enosawa T, Henopp T , et al. Primary lymph node gastrinoma or occult duodenal microgastrinoma with lymph node metastases in a MEN1 patient: the need for a systematic search for the primary tumor. Am J Surg Pathol 2008;32:1101-1105
  269. Chen Y, Deshpande V, Ferrone C , et al. Primary lymph node gastrinoma: A single institution experience. Surgery 2017;162:1088-1094
  270. Singh D, Lal SB, Sood A , et al. Management of Primary Lymph Nodal Gastrinoma With Liver Metastases Resulting in Zollinger-Ellison Syndrome. Clin Nucl Med 2019;44:e36-e39
  271. Abu Ghanimeh M, Abuamr K, Sadeddin E , et al. Severe chronic diarrhoea secondary to primary lymph node gastrinoma. BMJ Case Rep 2017;bcr2016216855
  272. Cavalcanti E, Stasi E, Coletta S , et al. Primary lymph node gastrinoma: a case report and review of the literature. World J Surg Oncol 2020;18:80
  273. Marcos Enriquez JC, Coayla CF, Caceres R.M. , et al. (Primary lymph node gastrinoma). Rev Gastroenterol Peru 2022;42:122-125
  274. Zhang G, Jiang Q, Zhang S , et al. The sun beside the pancreatic neck: The lymph node gastrinoma (with video). Endosc Ultrasound 2023;12:152-154
  275. Herrmann ME, Ciesla MC, Chejfec G , et al. Primary Nodal gastrinomas - an immunohistochemical study in support of a theory. Arch Pathol Lab Med 2000;124:832-835
  276. Stabile BE, Morrow DJ, Passaro E, Jr. The gastrinoma triangle: operative implications. Am J Surg 1984;147:25-31
  277. Thom AK, Norton JA, Axiotis CA , et al. Location, incidence and malignant potential of duodenal gastrinomas. Surgery 1991;110:1086-1093
  278. Delcore R, Jr., Cheung LY, Friesen SR. Characteristics of duodenal wall gastrinomas. Am J Surg 1990;160:621-623
  279. Metz DC, Jensen RT, Bale AE et al.  Multiple endocrine neoplasia type 1: clinical features and management. In: Bilezekian JP, Levine MA, Marcus R eds. The Parathyroids. New York: Raven Press Publishing Co.; 1994:591-646.
  280. Kloppel G, Schroder S, Heitz PU. Histopathology and immunopathology of pancreatic endocrine tumors. In: Mignon M, Jensen RT eds. Endocrine Tumors of the Pancreas: Recent advances in research and management. Series: Frontiers of Gastrointestinal Research. Basel, Switzerland: S. Karger; 1995:99-120.
  281. Donow C, Pipeleers-Marichal M, Schroder S , et al. Surgical pathology of gastrinoma: site, size, multicentricity, association with multiple endocrine neoplasia type 1, and malignancy. Cancer 1991;68:1329-1334
  282. Creutzfeldt W, Arnold R. Somatostatin and the stomach: exocrine and endocrine aspects. Metabolism 1978;27:1309-1315
  283. Pipeleers-Marichal M, Donow C, Heitz PU , et al. Pathologic aspects of gastrinomas in patients with Zollinger-Ellison syndrome with and without multiple endocrine neoplasia type I. World J Surg 1993;17:481-488
  284. MacFarlane MP, Fraker DL, Alexander HR , et al. A prospective study of surgical resection of duodenal and pancreatic gastrinomas in multiple endocrine neoplasia-Type 1. Surgery 1995;118:973-980
  285. Hoffmann KM, Gibril F, Entsuah LK , et al. Patients with multiple endocrine neoplasia type 1 with gastrinomas have an increased risk of severe esophageal disease including stricture and the premalignant condition, Barrett's esophagus. J Clin Endocrinol Metab 2006;91:204-212
  286. Rosentraeger MJ, Garbrecht N, Anlauf M , et al. Syndromic versus non-syndromic sporadic gastrin-producing neuroendocrine tumors of the duodenum: comparison of pathological features and biological behavior. Virchows Arch 2016;468:277-287
  287. Norton JA, Jensen RT. Current surgical management of Zollinger-Ellison syndrome (ZES) in patients without multiple endocrine neoplasia-type 1 (MEN1). Surg Oncol 2003;12:145-151
  288. Norton JA, Doherty GD, Fraker DL , et al. Surgical treatment of localized gastrinoma within the liver: A prospective study. Surgery 1998;124:1145-1152
  289. Zogakis TG, Gibril F, Libutti SK , et al. Management and outcome of patients with sporadic gastrinomas arising in the duodenum. Ann Surg 2003;238:42-48
  290. Oberg K, Sundin A. Imaging of Neuroendocrine Tumors. Front Horm Res 2016;45:142-151
  291. Sundin A, Arnold R, Baudin E , et al. ENETS Consensus Guidelines for the Standards of Care in Neuroendocrine Tumors: Radiological, Nuclear Medicine & Hybrid Imaging. Neuroendocrinology 2017;105:212-244
  292. Sundin A. Novel Functional Imaging of Neuroendocrine Tumors. Endocrinol Metab Clin North Am 2018;47:505-523
  293. Gabriel S, Garrigue P, Dahan L , et al. Prospective evaluation of (68) Ga-DOTATATE PET/CT in limited disease neuroendocrine tumours and/or elevated serum neuroendocrine biomarkers. Clin Endocrinol (Oxf) 2018;89:155-163
  294. Lebtahi R, Cadiot G, Delahaye N , et al. Detection of bone metastases in patients with endocrine gastroenteropancreatic tumors: bone scintigraphy compared with somatostatin receptor scintigraphy. J Nucl Med 1999;40:1602-1608
  295. Jensen RT. Endocrine Neoplasms of the Pancreas. In: Yamada T, Alpers DH, Kalloo AN et al.  eds. Textbook of Gastroenterology. Oxford, England: Wiley-Blackwell; 2009:1875-1920.
  296. Kos-Kudla B, O'Toole D, Falconi M , et al. ENETS consensus guidelines for the management of bone and lung metastases from neuroendocrine tumors. Neuroendocrinology 2010;91:341-350
  297. Leboulleux S, Dromain C, Vataire AL , et al. Prediction and diagnosis of bone metastases in well-differentiated gastro-entero-pancreatic endocrine cancer: a prospective comparison of whole body magnetic resonance imaging and somatostatin receptor scintigraphy. J Clin Endocrinol Metab 2008;93:3021-3028
  298. Van Loon K, Zhang L, Keiser J , et al. Bone metastases and skeletal-related events from neuroendocrine tumors. Endocr Connect 2015;4:9-17
  299. Altieri B, Di Dato C, Martini C , et al. Bone Metastases in Neuroendocrine Neoplasms: From Pathogenesis to Clinical Management. Cancers (Basel) 2019;11:1332
  300. Cortes M, Mora J, Benitez A , et al. (Bone metastasis from gastrinoma without hepatic involvement. Utility of the scintigraphy with 111 In-pentetreotide). Rev Esp Med Nucl 2001;20:23-26
  301. Klimstra DS, Modlin IR, Coppola D , et al. The pathologic classification of neuroendocrine tumors: a review of nomenclature, grading, and staging systems. Pancreas 2010;39:707-712
  302. Heitz PU, Kasper M, Polak JM , et al. Pancreatic endocrine tumors. Immunohistochemial analysis of 125 tumors. Hum Pathol 1982;13:263-271
  303. Chiang HC, O'Dorisio TM, Huang SC , et al. Multiple hormone elevations in patients with Zollinger-Ellison syndrome: Prospective study of clinical significance and of the development of a second symptomatic pancreatic endocrine tumor syndrome. Gastroenterology 1990;99:1565-1575
  304. Klimstra DS. Pathology reporting of neuroendocrine tumors: essential elements for accurate diagnosis, classification, and staging. Semin Oncol 2013;40:23-36
  305. Kloppel G. Classification and pathology of gastroenteropancreatic neuroendocrine neoplasms. Endocr Relat Cancer 2011;18 Suppl 1:S1-S16
  306. Rindi G, Mete O, Uccella S , et al. Overview of the 2022 WHO Classification of Neuroendocrine Neoplasms. Endocr Pathol 2022;33:115-154
  307. Kloppel G. Neuroendocrine Neoplasms: Dichotomy, Origin and Classifications. Visc Med 2017;33:324-330
  308. Tang LH. Pancreatic Neuroendocrine Neoplasms: Landscape and Horizon. Arch Pathol Lab Med 2020;144:816-828
  309. Anonymous. WHO classification of tumours of endocrine organs. Vol. 10, 4th Edition. Lyon, France: Agency for Research on Cancer; 2017.
  310. Sorbye H, Baudin E, Perren A. The Problem of High-Grade Gastroenteropancreatic Neuroendocrine Neoplasms: Well-Differentiated Neuroendocrine Tumors, Neuroendocrine Carcinomas, and Beyond. Endocrinol Metab Clin North Am 2018;47:683-698
  311. Milione M, Maisonneuve P, Spada F , et al. The Clinicopathologic Heterogeneity of Grade 3 Gastroenteropancreatic Neuroendocrine Neoplasms: Morphological Differentiation and Proliferation Identify Different Prognostic Categories. Neuroendocrinology 2017;104:85-93
  312. Rindi G, Falconi M, Klersy C , et al. TNM Staging of Neoplasms of the Endocrine Pancreas: Results From a Large International Cohort Study. J Natl Cancer Inst 2012;104:764-777
  313. Jensen RT, Ito T. Gastrinoma. Endotext(Internet){www endotext org) 2020;
  314. Stabile BE, Passaro E, Jr. Benign and malignant gastrinoma. Am J Surg 1985;49:144-150
  315. Cadiot G, Vuagnat A, Doukhan I , et al. Prognostic factors in patients with Zollinger-Ellison syndrome and multiple endocrine neoplasia type 1. Gastroenterology 1999;116:286-293
  316. Ruszniewski P, Podevin P, Cadiot G , et al. Clinical, anatomical, and evolutive features of patients with the Zollinger-Ellison syndrome combined with type I multiple endocrine neoplasia. Pancreas 1993;8:295-304
  317. Chandrasekharappa SC, Guru SC, Manickam P , et al. Positional cloning of the gene for multiple endocrine neoplasia-Type 1. Science 1997;276:404-407
  318. Debelenko LV, Zhuang ZP, Emmert-Buck MR , et al. Allelic deletions on chromosome 11q13 in Multiple Endocrine Neoplasia Type-I-associated sporadic gastrinomas and pancreatic endocrine tumors. Cancer Res 1997;57:2238-2243
  319. Goebel SU, Vortmeyer O, Zhang ZP , et al. Identical clonality of sporadic gastrinomas at multiple sites. Cancer Res 2000;60:60-63
  320. Gurevich L, Kazantseva I, Isakov VA , et al. The analysis of immunophenotype of gastrin-producing tumors of the pancreas and gastrointestinal tract. Cancer 2003;98:1967-1976
  321. Kimura H, Ohtsuka T, Fujimoto T , et al. Different Hormonal Expression Patterns Between Primary Pancreatic Neuroendocrine Tumors and Metastatic Sites. Pancreas 2016;45:947-952
  322. Hong L, Wang Y, Zhang T , et al. Chromogranin A: A Valuable Serum Diagnostic Marker for Non-Insulinoma Neuroendocrine Tumors of the Pancreas in a Chinese Population. Med Sci Monit 2020;26:e926635
  323. Cherner JA, Doppman JL, Norton JA , et al. Selective venous sampling for gastrin to localize gastrinomas. A prospective study. Ann Intern Med 1986;105:841-847
  324. Wynick D, Williams SJ, Bloom SR. Symptomatic secondary hormone syndromes in patients with established malignant pancreatic endocrine tumors. N Engl J Med 1988;319:605-607
  325. Lase I, Strele I, Gronberg M , et al. Multiple hormone secretion may indicate worse prognosis in patients with ectopic Cushing's syndrome. Hormones (Athens ) 2020;19:351-360
  326. Price DE, Absalom SR, Davidson K , et al. A case of multiple endocrine neoplasia: hyperparathyroidism, insulinoma, GRF-oma, hypercalcitoninaemia and intractable peptic ulceration. Clin Endocrinol (Oxf) 1992;37:187-188
  327. Liaw CC, Lin JT, Chen TJ. Multiple-hormone-producing islet cell carcinoma: report of a case. Taiwan Yi Xue Hui Za Zhi 1989;88:722-725
  328. Murray-Lyon IM, Eddleston AL, Williams R , et al. Treatment of multiple-hormone-producing malignant islet-cell tumor with streptozotocin. Lancet 1968;2:895-898
  329. Lodish MB, Powell AC, Abu-Asab M , et al. Insulinoma and gastrinoma syndromes from a single intrapancreatic neuroendocrine tumor. J Clin Endocrinol Metab 2008;93:1123-1128
  330. Rustagi T, Siegel RD. Zollinger-ellison syndrome with subsequent association of insulinoma. JOP 2010;11:486-488
  331. Mizuno N, Naruse S, Kitagawa M , et al. Insulinoma with subsequent association of Zollinger-Ellison syndrome. Intern Med 2001;40:386-390
  332. Wilson DM, Ceda GP, Bostwick DG , et al. Acromegaly and Zollinger-Ellison syndrome secondary to an islet cell tumor: characterization and quantification of plasma and tumor human growth hormone-releasing factor. J Clin Endocrinol Metab 1984;59:1002-1005
  333. Ching JL, Christofides ND, Kraenzlin ME , et al. Growth hormone secretion dynamics in a patient with ectopic growth hormone-releasing factor production. Am J Med 1985;79:135-138
  334. Kubicka E, Zawadzka K, Syrycka J , et al. A case of gastrinoma associated with ectopic Cushing syndrome. Pol Arch Intern Med 2020;130:328-329
  335. Amikura K, Alexander HR, Norton JA , et al. The role of surgery in the management of ACTH-producing islet cell tumors of the pancreas. Surgery 1995;118:1125-1130
  336. Doppman JL, Nieman LK, Cutler GB, Jr. , et al. Adrenocorticotropic hormone-secreting islet cell tumors: Are they always malignant? Radiology 1994;190:59-64
  337. Ilias I, Torpy DJ, Pacak K , et al. Cushing's syndrome due to ectopic corticotropin secretion: twenty years' experience at the National Institutes of Health. J Clin Endocrinol Metab 2005;90:4955-4962
  338. Raddatz D, Horstmann O, Basenau D , et al. Cushing's syndrome due to ectopic adrenocorticotropic hormone production by a non-metastatic gastrinoma after lonterm conservative treatment of Zollinger-Ellison syndrome. Ital J Gastroenterol Hepatol 1998;30:636-640
  339. Asa SL, Kovacs K, Killinger DW , et al. Pancreatic islet cell carcinoma producing gastrin, ACTH, alpha- endorphin, somatostatin and calcitonin. Am J Gastroenterol 1980;74(1):30-35
  340. Law DH, Liddle GW, Scott HW, Jr. , et al. Ectopic production of multiple hormones (ACTH, MSH and gastrin) by a single malignant tumor. N Engl J Med 1965;273:292-296
  341. Lyons DF, Eisen BR, Clark MR , et al. Concurrent Cushing's and Zollinger-Ellison syndromes in a patient with islet cell carcinoma: case report and review of the literature. Am J Med 1984;76:729-733
  342. Belchetz PE, Brown CL, Makin HL , et al. ACTH, glucagon and gastrin production by a pancreatic islet cell carcinoma and its treatment. Clin Endocrinol 1973;2:307-316
  343. O'Neal LW, Kipnis DM, Luse SA , et al. Secretion of various endocrine substances by ACTH-secreting tumors--gastrin, melanotropin, norepinephrine, serotonin, parathormone, vasopressin, glucagon. Cancer 1968;21:1219-1232
  344. Park SY, Rhee Y, Youn JC , et al. Ectopic Cushing's Syndrome Due to Concurrent Corticotropin-Releasing Hormone (CRH) and Adrenocorticotropic Hormone (ACTH) Secreted by Malignant Gastrinoma. Exp Clin Endocrinol Diabetes 2007;115:13-16
  345. Ahmad NA, Furth EE, Schwartz SS , et al. Sporadic Zollinger-Ellison syndrome with ectopic production of corticotropin: surgical management. Endocr Pract 1999;5:261-265
  346. Kido-Nakahara M, Nakahara T, Miki M , et al. Necrolytic migratory erythema associated with alteration from predominantly gastrin-secreting to predominantly glucagon-secreting pancreatic neuroendocrine tumor. Eur J Dermatol 2014;24:702-703
  347. Balian A, Fromont C, Naveau S , et al. A particularly aggressive combined glucagonoma and gastrinoma syndrome. Eur J Gastroenterol Hepatol 1999;11:1417-1419
  348. Lokich J, Bothe A, O'Hara C , et al. Metastatic islet cell tumor with ACTH, gastrin, and glucagon secretion. Clinical and pathologic studies with multiple therapies. Cancer 1987;59:2053-2058
  349. Soga J, Yakuwa Y. Somatostatinoma/inhibitory syndrome: a statistical evaluation of 173 reported cases as compared to other pancreatic endocrinomas. J Exp Clin Cancer Res 1999;18:13-22
  350. Ito T, Lee L, Jensen RT. Carcinoid-syndrome: recent advances, current status and controversies. Curr Opin Endocrinol Diabetes Obes 2018;25:22-35
  351. Morita Y, Suzuki S, Sakaguchi T , et al. Pancreatic neuroendocrine cell tumor secreting parathyroid hormone-related protein and gastrin: Report of a case. Surg Today 2010;40:1192-1196
  352. Cho KJ, Vinik AI, Thompson NW , et al. Localization of the source of hyperinsulinism: percutaneous transhepatic portal and pancreatic vein catheterization with hormone assay. AJR Am J Roentgenol 1982;139:237-245
  353. Fraker DL, Jensen RT. Pancreatic endocrine tumors. In: DeVita VT, Hellman S, Rosenberg SA eds. Cancer: Principles and Practice of Oncology. Philadelphia, PA: Lippincott-Raven Publishers; 1997:1678-1704.
  354. Jensen RT, Norton JA. Endocrine neoplasms of the pancreas. In: Yamada T, Alpers DH, Owyang C et al.  eds. Textbook of Gastroenterology. Philadelphia: J.B. Lippincott Company; 1998:2193-2228.
  355. Jensen RT, Norton JA. Endocrine tumors of the pancreas. In: Feldman M, Scharschmidt BF, Sleisenger MH eds. Gastrointestinal and Liver Disease. Philadelphia: W.B. Saunders; 1998:871-894.
  356. Jensen RT. Pancreatic endocrine tumors: recent advances. Ann Oncol 1999;10:170-176
  357. Rehfeld JF, Friis-Hansen L, Goetze JP , et al. The biology of cholecystokinin and gastrin peptides. Curr Top Med Chem 2007;7:1154-1165
  358. Walsh JH. Gastrin. In: Walsh JH, Dockray GJ eds. Gut Peptides. New York: Raven Press; 1994:75-121.
  359. Bardram L. Progastrin in pancreas and the Zollinger-Ellison syndrome. Scand J Gastroenterol 1990;25:1185-1195
  360. Bardram L. Progastrin in serum from Zollinger-Ellison patients. An indicator of malignancy? Gastroenterology 1990;98:1420-1426
  361. d'Herbomez M, Do Cao C, Vezzosi D , et al. Chromogranin A assay in clinical practice. Ann Endocrinol (Paris) 2010;71:274-280
  362. Lawrence B, Gustafsson BI, Kidd M , et al. The clinical relevance of chromogranin A as a biomarker for gastroenteropancreatic neuroendocrine tumors. Endocrinol Metab Clin North Am 2011;40:111-34, viii
  363. Baekdal J, Krogh J, Klose M , et al. Limited Diagnostic Utility of Chromogranin A Measurements in Workup of Neuroendocrine Tumors. Diagnostics (Basel) 2020;10:881
  364. Jacoba IM, Weber HC. Biomarkers in gastroenteropancreatic neuroendocrine neoplasms. Curr Opin Endocrinol Diabetes Obes 2023;30:175-180
  365. Nobels FR, Kwekkeboom DJ, Bouillon R , et al. Chromogranin A: its clinical value as marker of neuroendocrine tumours. Eur J Clin Invest 1998;28:431-440
  366. Goebel SU, Serrano J, Yu F , et al. Prospective study of the value of serum chromogranin A or serum gastrin levels in assessment of the presence, extent, or growth of gastrinomas. Cancer 1999;85:1470-1483
  367. Nobels FR, Kwekkeboom DJ, Coopmans W , et al. Chromogranin A as serum marker for neuroendocrine neoplasia: comparison with neuron-specific enolase and the alpha-subunit of glycoprotein hormones. J Clin Endocrinol Metab 1997;82:2622-2628
  368. Ito T, Igarashi H, Jensen RT. Serum pancreastatin: the long sought universal, sensitive, specific tumor marker for neuroendocrine tumors? Pancreas 2012;41:505-507
  369. Abou-Saif A, Gibril F, Ojeaburu JV , et al. Prospective study of the ability of serial measurements of serum chromogranin A and gastrin to detect changes in tumor burden in patients with gastrinomas. Cancer 2003;98:249-261
  370. Rehfeld JF, Bardram L, Hilsted L , et al. An evaluation of chromogranin A versus gastrin and progastrin in gastrinoma diagnosis and control. Biomark Med 2014;8:571-580
  371. Massironi S, Rossi RE, Casazza G , et al. Chromogranin A in diagnosing and monitoring patients with gastroenteropancreatic neuroendocrine neoplasms: a large series from a single institution. Neuroendocrinology 2014;100:240-249
  372. Rehfeld JF. Chromogranin A in gastrinomas: Promises and pitfalls. Clin Chim Acta 2015;446:15-20
  373. Malczewska A, Oberg K, Kos-Kudla B. NETest is superior to chromogranin A in neuroendocrine neoplasia: a prospective ENETS CoE analysis. Endocr Connect 2021;10:110-123
  374. Malczewska A, Witkowska M, Wójcik-Giertuga M , et al. Prospective Evaluation of the NETest as a Liquid Biopsy for Gastroenteropancreatic and Bronchopulmonary Neuroendocrine Tumors: An ENETS Center of Excellence Experience. Neuroendocrinology 2021;111:304-319
  375. Modlin IM, Kidd M, Falconi M , et al. A multigenomic liquid biopsy biomarker for neuroendocrine tumor disease outperforms CgA and has surgical and clinical utility. Ann Oncol 2021;32:1425-1433
  376. Raines D, Chester M, Diebold AE , et al. A prospective evaluation of the effect of chronic proton pump inhibitor use on plasma biomarker levels in humans. Pancreas 2012;41:508-511
  377. Syversen U, Mignon M, Bonfils S , et al. Chromogranin A and pancreastatin-like immunoreactivity in serum of gastrinoma patients. Acta Oncol 1993;32:161-165
  378. Stabile BE, Howard TJ, Passaro E, Jr. , et al. Source of plasma chromogranin A elevation in gastrinoma patients. Arch Surg 1990;125:451-453
  379. Hirschowitz BI, Worthington J, Mohnen J , et al. Chromogranin A in patients with acid hypersecretion and/or hypergastrinaemia. Aliment Pharmacol Ther 2007;26:869-878
  380. Elouaer-Blanc L, Sobhani I, Ruszniewski P , et al. Gastrin secretion by gastrinoma cells in long-term culture. Am J Physiol 1988;255:G596-G602
  381. Isenberg JI, Walsh JH, Passaro E, Jr. , et al. Unusual effect of secretin on serum gastrin, serum calcium and gastric acid secretion in a patient with suspected Zollinger- Ellison syndrome. Gastroenterology 1972;62:626-631
  382. McGuigan JE, Wolfe MM. Secretin injection test in the diagnosis of gastrinoma. Gastroenterology 1980;79:1324-1331
  383. Deveney CW, Deveney KS, Jaffe BM , et al. Use of calcium and secretin in the diagnosis of gastrinoma (Zollinger-Ellison syndrome). Ann Intern Med 1977;87:680-686
  384. Lamers CB, Van Tongeren JHM. Comparative study of the value of calcium, secretin, and meal stimulated increase in serum gastrin in the diagnosis of the Zollinger-Ellison syndrome. Gut 1977;18:128-134
  385. Korman MG, Soveny C, Hansky J. Effect of glucagon on serum gastrin. II. Studies in pernicious anaemia and the Zollinger-Ellison syndrome. Gut 1973;14:459-461
  386. Shibata C, Funayama Y, Fukushima K , et al. Role of selective arterial secretin injection test in treatment of gastrinoma. Hepatogastroenterology 2006;53:960-963
  387. Shibata C, Funayama Y, Fukushima K , et al. The Glucagon Provocative Test for the Diagnosis and Treatment of Zollinger-Ellison Syndrome. J Gastrointest Surg 2008;12:344-349
  388. Kawai K, Mukai H, Yuzawa K , et al. Effects of neuromedin B and GRP-10 on gastrin and insulin release from cultured tumor cells of a malignant gastrinoma. Endocrinol Jpn 1990;37:857-865
  389. Sakamoto T, Miyata M, Hamaji M , et al. Beta-adrenergic regulation of gastrin release from gastrinoma cells. Surgery 1990;107:282-288
  390. Passaro E, Jr., Basso N, Walsh JH. Calcium challenge in the Zollinger-Ellison syndrome. Surgery 1972;72:60-67
  391. Frucht H, Howard JM, Stark HA , et al. Prospective study of the standard meal provocative test in Zollinger-Ellison syndrome. Am J Med 1989;87:528-536
  392. Malagelada JR. Pathophysiological responses to meals in the Zollinger-Ellison syndrome: 1. Paradoxical postprandial inhibition of gastric secretion. Gut 1978;19:284-289
  393. Ellison EC, Gower WR, Elkhammas E , et al. Characterization of the in vivo and in vitro inhibition of gastrin secretion from gastrinoma by a somatostatin analogue (SMS 201-995). Am J Med 1986;81:56-64
  394. Vinik AI, Tsai S, Moattari AR , et al. Somatostatin analogue (SMS 201-995) in patients with gastrinomas. Surgery 1988;104:834-842
  395. Ruszniewski P, Ramdani A, Cadiot G , et al. Long-term treatment with octreotide in patients with the Zollinger-Ellison syndrome. Eur J Clin Invest 1993;23:296-301
  396. Auernhammer CJ, Goke B. Medical treatment of gastrinomas. Wien Klin Wochenschr 2007;119:609-615
  397. Long SH, Berna MJ, Thill M , et al. Secretin-receptor and secretin-receptor-variant expression in gastrinomas: correlation with clinical and tumoral features and secretin and calcium provocative test results. J Clin Endocrinol Metab 2007;92:4394-4402
  398. Goebel SU, Peghini PL, Goldsmith PK , et al. Expression of the calcium-sensing receptor in gastrinomas. J Clin Endocrinol Metab 2000;85:4131-4137
  399. Itami A, Kato M, Komoto I , et al. Human gastrinoma cells express calcium-sensing receptor. Life Sci 2001;70:119-129
  400. Reubi JC, Kvols LK, Waser B , et al. Detection of somatostatin receptors in surgical and percutaneous needle biopsy samples of carcinoids and islet cell carcinomas. Cancer Res 1990;50:5969-5977
  401. Krenning EP, Kwekkeboom DJ, Bakker WH , et al. Somatostatin receptor scintigraphy with (111In-DTPA-D-Phe1)- and (123I-Tyr3)-octreotide: the Rotterdam experience with more than 1000 patients. Eur J Nucl Med 1993;20:716-731
  402. Shojamanesh H, Gibril F, Louie A , et al. Prospective study of the anti-tumor efficacy of long-term octreotide treatment in patients with progressive metastatic gastrinomas. Cancer 2002;94:331-343
  403. Daniels LM, Khalili M, Morano WF , et al. Case report: optimal tumor cytoreduction and octreotide with durable disease control in a patient with MEN-1 and Zollinger-Ellison syndrome-over a decade of follow-up. World J Surg Oncol 2019;17:213
  404. Kwekkeboom DJ, de Herder WW, Kam BL , et al. Treatment with the radiolabeled somatostatin analog (177 Lu-DOTA 0,Tyr3)octreotate: toxicity, efficacy, and survival. J Clin Oncol 2008;26:2124-2130
  405. Brady CE, III. Secretin provocation test in the diagnosis of Zollinger-Ellison syndrome. Am J Gastroenterol 1991;86:129-134
  406. Brady CE, Utts SJ, Dev J. Secretin provocation in normal and duodenal ulcer subjects. Is the gastrin rise in Zollinger-Ellison syndrome paradoxic or exaggeration? Dig Dis Sci 1987;32:232-238
  407. Gower WR, Jr., Ellison EC, Knierim TH , et al. Gastrinoma in vitro: morphological and physiological studies of primary cell cultures. Gastroenterology 1990;98:936-954
  408. Imamura M, Adachi H, Takahashi K , et al. Gastrin release from gastrinoma cells stimulated with secretin. Dig Dis Sci 1982;27:1130-1136
  409. Chiba T, Yamatani T, Yamaguchi A , et al. Mechanism for increase of gastrin release by secretin in Zollinger-Ellison syndrome. Gastroenterology 1989;96:1439-1444
  410. Sawicki MP, Wan YJ, Johnson CL , et al. Loss of heterozygosity on chromosome 11 in sporadic gastrinomas. Hum Genet 1992;89:445-449
  411. Vortmeyer AO, Huang S, Lubensky I , et al. Non-islet origin of pancreatic islet cell tumors. J Clin Endocrinol Metab 2004;89:1934-1938
  412. Bonnavion R, Teinturier R, Jaafar R , et al. Islet Cells Serve as Cells of Origin of Pancreatic Gastrin-Positive Endocrine Tumors. Mol Cell Biol 2015;35:3274-3283
  413. Larsson LI, Rehfeld JF, Goltermann N. Gastrin in the human fetus: Distribution and molecular forms of gastrin in the antro-pyloric gland area, duodenum and pancreas. Scand J Gastroenterol 1977;12:869-872
  414. Passaro E, Jr., Howard TJ, Sawicki MP , et al. The origin of sporadic gastrinomas within the gastrinoma triangle: a theory. Arch Surg 1998;133:13-16
  415. Howard TJ, Sawicki MP, Stabile BE , et al. Biologic behavior of sporadic gastrinoma located to the right and left of the superior mesenteric artery. Am J Surg 1993;165:101-105
  416. Howard TJ, Stabile BE, Zinner MJ , et al. Anatomic distribution of pancreatic endocrine tumors. Am J Surg 1990;159:258-264
  417. Sawicki MP, Howard TJ, Dalton M , et al. The dichotomous distribution of gastrinomas. Arch Surg 1990;125:1584-1587
  418. Solcia E, Capella C, Buffa R , et al. Endocrine cells of the gastrointestinal tract and related tumors. Pathobiol Annu 1979;9:163-204
  419. Solcia E, Capella C, Buffa R , et al. Pathology of the Zollinger-Ellison syndrome. Prog Surg Pathol 1980;I:119-133
  420. Fendrich V, Ramerth R, Waldmann J , et al. Sonic hedgehog and pancreatic-duodenal homeobox 1 expression distinguish between duodenal and pancreatic gastrinomas. Endocr Relat Cancer 2009;16:613-622
  421. Grande E, Capdevila J, Barriuso J , et al. Gastroenteropancreatic neuroendocrine tumor cancer stem cells: do they exist? Cancer Metastasis Rev 2012;31:47-53
  422. Kloppel G, Anlauf M, Perren A. Endocrine precursor lesions of gastroenteropancreatic neuroendocrine tumors. Endocr Pathol 2007;18:150-155
  423. Anlauf M, Perren A, Meyer CL , et al. Precursor lesions in patients with multiple endocrine neoplasia type 1-associated duodenal gastrinomas. Gastroenterology 2005;128:1187-1198
  424. Anlauf M, Perren A, Henopp T , et al. Allelic deletion of the MEN1 gene in duodenal gastrin and somatostatin cell neoplasms and their precursor lesions. Gut 2007;56:637-644
  425. Anlauf M, Perren A, Kloppel G. Endocrine precursor lesions and microadenomas of the duodenum and pancreas with and without MEN1: criteria, molecular concepts and clinical significance. Pathobiology 2007;74:279-284
  426. Sundaresan S, Kang AJ, Hayes MM , et al. Deletion of Men1 and somatostatin induces hypergastrinemia and gastric carcinoids. Gut 2017;66:1012-1021
  427. Sundaresan S, Meininger CA, Kang AJ , et al. Gastrin Induces Nuclear Export and Proteasome Degradation of Menin in Enteric Glial Cells. Gastroenterology 2017;153:1555-1567
  428. Perren A, Anlauf M, Henopp T , et al. Multiple endocrine neoplasia type 1 (MEN1): loss of one MEN1 allele in tumors and monohormonal endocrine cell clusters but not in islet hyperplasia of the pancreas. J Clin Endocrinol Metab 2007;92:1118-1128
  429. Sutliff VE, Doppman JL, Gibril F , et al. Growth of newly diagnosed, untreated metastatic gastrinomas and predictors of growth patterns. J Clin Oncol 1997;15:2420-2431
  430. Goudet P, Peschaud F, Mignon M , et al. Les gastrinomes dans les neoplasies endocriniennes multiples de type 1. Une etude de cohorte de 127 cas du groupe des tumeurs endocrines (GTE). Ann Chir 2004;129:149-155
  431. Goudet P, Murat A, Binquet C , et al. Risk factors and causes of death in MEN1 disease. A GTE (Groupe d'Etude des Tumeurs Endocrines) cohort study among 758 patients. World J Surg 2010;34:249-255
  432. Giudici F, Cavalli T, Giusti F , et al. Natural History of MEN1 GEP-NET: Single-Center Experience After a Long Follow-Up. World J Surg 2017;41:2312-2323
  433. Mignon M, Cadiot G. Natural history of gastrinoma: lessons from the past. Ital J Gastroenterol Hepatol 1999;31:S98-S103
  434. Thompson JC, Reeder DD, Villar HV , et al. Natural history and experience with diagnosis and treatment of the Zollinger-Ellison syndrome. Surg Gynecol Obstet 1975;140:721-739
  435. Madeira I, Terris B, Voss M , et al. Prognostic factors in patients with endocrine tumours of the duodenopancreatic area. Gut 1998;43:422-427
  436. Dromain C, Pavel ME, Ruszniewski P , et al. Tumor growth rate as a metric of progression, response, and prognosis in pancreatic and intestinal neuroendocrine tumors. BMC Cancer 2019;19:66
  437. Dromain C, Sundin A, Najran P , et al. Tumour Growth Rate to predict the outcome of patients with Neuroendocrine Tumours: Performance and sources of variability. Neuroendocrinology 2021;111:831-839
  438. Palazzo M, Lombard-Bohas C, Cadiot G , et al. Ki67 proliferation index, hepatic tumor load, and pretreatment tumor growth predict the antitumoral efficacy of lanreotide in patients with malignant digestive neuroendocrine tumors. Eur J Gastroenterol Hepatol 2013;25:232-238
  439. Lamarca A, Ronot M, Moalla S , et al. Tumor Growth Rate as a Validated Early Radiological Biomarker Able to Reflect Treatment-Induced Changes in Neuroendocrine Tumors: The GREPONET-2 Study. Clin Cancer Res 2019;25:6692-6699
  440. Lamarca A, Crona J, Ronot M , et al. Value of Tumor Growth Rate (TGR) as an Early Biomarker Predictor of Patients' Outcome in Neuroendocrine Tumors (NET)-The GREPONET Study. Oncologist 2019;24:e1082-e1090
  441. Oberg K. The genetics of neuroendocrine tumors. Semin Oncol 2013;40:37-44
  442. Zhang J, Francois R, Iyer R , et al. Current understanding of the molecular biology of pancreatic neuroendocrine tumors. J Natl Cancer Inst 2013;105:1005-1017
  443. Scarpa A. The landscape of molecular alterations in pancreatic and small intestinal neuroendocrine tumours. Ann Endocrinol (Paris) 2019;80:153-158
  444. Hackeng WM, Assi HA, Westerbeke FHM , et al. Prognostic and Predictive Biomarkers for Pancreatic Neuroendocrine Tumors. Surg Pathol Clin 2022;15:541-554
  445. Di Domenico A, Pipinikas CP, Maire RS , et al. Epigenetic landscape of pancreatic neuroendocrine tumours reveals distinct cells of origin and means of tumour progression. Commun Biol 2020;3:740
  446. Jiao Y, Shi C, Edil BH , et al. DAXX/ATRX, MEN1, and mTOR pathway genes are frequently altered in pancreatic neuroendocrine tumors. Science 2011;331:1199-1203
  447. Cejas P, Drier Y, Dreijerink KMA , et al. Enhancer signatures stratify and predict outcomes of non-functional pancreatic neuroendocrine tumors. Nat Med 2019;25:1260-1265
  448. Mohindroo C, McAllister F, De Jesus-Acosta A. Genetics of Pancreatic Neuroendocrine Tumors. Hematol Oncol Clin North Am 2022;36:1033-1051
  449. Duerr EM, Chung DC. Molecular genetics of neuroendocrine tumors. Best Pract Res Clin Endocrinol Metab 2007;21:1-14
  450. Corleto VD, Delle Fave G, Jensen RT. Molecular insights into gastrointestinal neuroendocrine tumors: importance and recent advances. Dig Liver Dis 2002;34:668-680
  451. Di Domenico A, Wiedmer T, Marinoni I , et al. Genetic and epigenetic drivers of neuroendocrine tumours (NET). Endocr Relat Cancer 2017;24:R315-R334
  452. Pipinikas CP, Berner AM, Sposito T , et al. The evolving (epi)genetic landscape of pancreatic neuroendocrine tumours. Endocr Relat Cancer 2019;26:R519-R544
  453. Wang H, Sun L, Bao H , et al. Genomic dissection of gastrointestinal and lung neuroendocrine neoplasm. Chin J Cancer Res 2019;31:918-929
  454. Hu W, Feng Z, Modica I , et al. Gene Amplifications in Well-Differentiated Pancreatic Neuroendocrine Tumors Inactivate the p53 Pathway. Genes Cancer 2010;1:360-368
  455. Tang LH, Contractor T, Clausen R , et al. Attenuation of the retinoblastoma pathway in pancreatic neuroendocrine tumors due to increased cdk4/cdk6. Clin Cancer Res 2012;18:4612-4620
  456. Chen Y, Ohki R. p53-PHLDA3-Akt Network: The Key Regulators of Neuroendocrine Tumorigenesis. Int J Mol Sci 2020;21:
  457. Ohki R, Saito K, Chen Y , et al. PHLDA3 is a novel tumor suppressor of pancreatic neuroendocrine tumors. Proc Natl Acad Sci U S A 2014;111:E2404-E2413
  458. Mowrey K, Northrup H, Rougeau P , et al. Frequency, Progression, and Current Management: Report of 16 New Cases of Nonfunctional Pancreatic Neuroendocrine Tumors in Tuberous Sclerosis Complex and Comparison With Previous Reports. Front Neurol 2021;12:627672
  459. Ahmad S, Naber MR, Giles RH , et al. Diagnostic and management strategies for pNETs in Von Hippel-Lindau: a systematic review. Endocr Relat Cancer 2021;28:151-160
  460. Evans LM, Geenen KR, O'Shea A , et al. Tuberous sclerosis complex-associated nonfunctional pancreatic neuroendocrine tumors: Management and surgical outcomes. Am J Med Genet A 2022;188:2666-2671
  461. Halperin R, Tirosh A. Non-Interventional Management of Advanced Pancreatic Neuroendocrine Neoplasms in Patients with von Hippel-Lindau Disease. Cancers (Basel) 2023;15:1739
  462. Mestre-Alagarda C, Srirajaskanthan R, Zen Y , et al. Genetic and epigenetic prognosticators of neuroendocrine tumours of the GI tract, liver, biliary tract and pancreas: A systematic review and meta-analysis. Histopathology 2023 (In Press)
  463. Thakker RV. Multiple endocrine neoplasia type 1. Endocrinol Metab Clin North Am 2000;29:541-567
  464. Thakker RV. Multiple endocrine neoplasia type 1 (MEN1). Best Pract Res Clin Endocrinol Metab 2010;24:355-370
  465. Goebel SU, Heppner C, Burns AD , et al. Geneotype/phenotype correlations of MEN1 gene mutations in sporadic gastrinoma. J Clin Endocrinol Metab 2000;85:116-123
  466. Falchetti A, Marini F, Luzi E , et al. Multiple endocrine neoplasms. Best Pract Res Clin Rheumatol 2008;22:149-163
  467. Duan S, Sheriff S, Elvis-Offiah UB , et al. Clinically Defined Mutations in MEN1 Alter Its Tumor-suppressive Function Through Increased Menin Turnover. Cancer Res Commun 2023;3:1318-1334
  468. Scarpa A, Chang DK, Nones K , et al. Whole-genome landscape of pancreatic neuroendocrine tumours. Nature 2017;543:65-71
  469. Yachida S, Vakiani E, White CM , et al. Small cell and large cell neuroendocrine carcinomas of the pancreas are genetically similar and distinct from well-differentiated pancreatic neuroendocrine tumors. Am J Surg Pathol 2012;36:173-184
  470. Chan CS, Laddha SV, Lewis PW , et al. ATRX, DAXX or MEN1 mutant pancreatic neuroendocrine tumors are a distinct alpha-cell signature subgroup. Nat Commun 2018;9:4158
  471. Marinoni I, Kurrer AS, Vassella E , et al. Loss of DAXX and ATRX are associated with chromosome instability and reduced survival of patients with pancreatic neuroendocrine tumors. Gastroenterology 2014;146:453-460
  472. Wang F, Xu X, Ye Z , et al. Prognostic Significance of Altered ATRX/DAXX Gene in Pancreatic Neuroendocrine Tumors: A Meta-Analysis. Front Endocrinol (Lausanne) 2021;12:691557
  473. Heaphy CM, Singhi AD. The diagnostic and prognostic utility of incorporating DAXX, ATRX, and alternative lengthening of telomeres to the evaluation of pancreatic neuroendocrine tumors. Hum Pathol 2022;129:11-20
  474. Hong X, Qiao S, Li F , et al. Whole-genome sequencing reveals distinct genetic bases for insulinomas and non-functional pancreatic neuroendocrine tumours: leading to a new classification system. Gut 2020;69:877-887
  475. Lund KA, Lazar CS, Chen WS , et al. Phosphorylation of the epidermal growth factor receptor at threonine 654 inhibits ligand-induced internalization and down- regulation. J Biol Chem 1990;265:20517-20523
  476. Simon T, Riemer P, Jarosch A , et al. DNA methylation reveals distinct cells of origin for pancreatic neuroendocrine carcinomas and pancreatic neuroendocrine tumors. Genome Med 2022;14:24
  477. Amato E, Barbi S, Malpeli G , et al. Chromosome 3p alterations in pancreatic endocrine neoplasia. Virchows Arch 2011;458:39-45
  478. Capurso G, Festa S, Valente R , et al. Molecular pathology and genetics of pancreatic endocrine tumours. J Mol Endocrinol 2012;49:R37-R50
  479. Wolin EM. PI3K/Akt/mTOR Pathway Inhibitors in the Therapy of Pancreatic Neuroendocrine Tumors. Cancer Lett 2013;335:1-8
  480. Yao JC, Shah MH, Ito T , et al. Everolimus for Advanced Pancreatic Neuroendocrine Tumors. N Engl J Med 2011;364:514-523
  481. Missiaglia E, Dalai I, Barbi S , et al. Pancreatic Endocrine Tumors: Expression Profiling Evidences a Role for AKT-mTOR Pathway. J Clin Oncol 2010;28:245-255
  482. Serra S, Zheng L, Hassan M , et al. The FGFR4-G388R single-nucleotide polymorphism alters pancreatic neuroendocrine tumor progression and response to mTOR inhibition therapy. Cancer Res 2012;72:5683-5691
  483. Zhou H, Chen Q, Tan W , et al. Integrated clinicopathological features and gene microarray analysis of pancreatic neuroendocrine tumors. Gene 2017;625:72-77
  484. Wang DD, Liu ZW, Han MM , et al. Microarray based analysis of gene expression patterns in pancreatic neuroendocrine tumors. Eur Rev Med Pharmacol Sci 2015;19:3367-3374
  485. Waldum HL, Hauso O, Fossmark R. The regulation of gastric acid secretion - clinical perspectives. Acta Physiol (Oxf) 2014;210:239-256
  486. Hua J, Shi S, Xu J , et al. Expression Patterns and Prognostic Value of DNA Damage Repair Proteins in Resected Pancreatic Neuroendocrine Neoplasms. Ann Surg 2022;275:443-452
  487. Wilson SD. The role of surgery in children with the Zollinger-Ellison syndrome. Surgery 1982;92(4):682-692
  488. Nazir Z. Long-term follow-up of a child with primary lymph node gastrinoma and Zollinger-Ellison syndrome. J Pediatr Surg 2011;46:969-972
  489. Cives M, Ghayouri M, Morse B , et al. Analysis of potential response predictors to capecitabine/temozolomide in metastatic pancreatic neuroendocrine tumors. Endocr Relat Cancer 2016;23:759-767
  490. Chang FY, Liao KY, Wu L , et al. An uncommon cause of abdominal pain and diarrhea-gastrinoma in an adolescent. Eur J Pediatr 2010;169:355-357
  491. Citak EC, Taskinlar H, Arpaci RB , et al. Primary lymph node gastrinoma: a rare cause of abdominal pain in childhood. J Pediatr Hematol Oncol 2013;35:394-398
  492. Goyal R, Debi U, Dey P , et al. Zollinger-Ellison syndrome: an unusual case of chronic diarrhoea in a child. Malays J Pathol 2016;38:321-325
  493. Anderson B, Sweetser S. Gastrointestinal: Zollinger-Ellison Syndrome: A rare cause of chronic diarrhea and abdominal pain. J Gastroenterol Hepatol 2017;32:1281
  494. Aamar A, Madhani K, Virk H , et al. Zollinger-Ellison Syndrome: A Rare Case of Chronic Diarrhea. Gastroenterology Res 2016;9:103-104
  495. Eads JR, Reidy-Lagunes D, Soares HP , et al. Differential Diagnosis of Diarrhea in Patients With Neuroendocrine Tumors. Pancreas 2020;49:1123-1130
  496. Bondeson AG, Bondeson L, Thompson NW. Stricture and perforation of the esophagus: overlooked threats in the Zollinger-Ellison syndrome. World J Surg 1990;14:361-363
  497. Siewert R, Jennewein HM, Arnold R , et al. The lower oesophageal sphincter in the Zollinger-Ellison syndrome. Ger Med 1973;3:101-102
  498. Mignon M, Jais P, Cadiot G et al.  Clinical features and advances in biological diagnostic criteria for Zollinger-Ellison syndrome. In: Mignon M, Jensen RT eds. Endocrine Tumors of the Pancreas: Recent Advances in Research and Management. Series: Frontiers of Gastrointestinal Research. Basel, Switzerland: S. Karger; 1995:223-239.
  499. Freund MR, Reissman P, Schwarz AD. Jejunal Perforations in Zollinger-Ellison Syndrome: Expect the Unexpected. Am Surg 2020;86:171-172
  500. Waxman I, Gardner JD, Jensen RT , et al. Peptic ulcer perforation as the presentation of Zollinger- Ellison syndrome. Dig Dis Sci 1991;16:19-24
  501. Pakzad N, Salonia J, Mathew J. A Young Man With Abdominal Pain, Shock, and Respiratory Distress. Chest 2018;154:e119-e121
  502. Jensen RT. Consequences of long-term proton pump blockade: Highlighting insights from studies of patients with gastrinomas. Basic Clin Pharmacol Toxicol 2006;98:4-19
  503. Miller LS, Vinayek R, Frucht H , et al. Reflux esophagitis in patients with Zollinger-Ellison syndrome. Gastroenterology 1990;98:341-346
  504. Kimura W, Kuroda A, Morioka Y. Clinical pathology of endocrine tumors of the pancreas. Analysis of autopsy cases. Dig Dis Sci 1991;36:933-942
  505. Thakker RV, Newey PJ, Walls GV , et al. Clinical Practice Guidelines for Multiple Endocrine Neoplasia Type 1 (MEN1). J Clin Endocrinol Metab 2012;97:2990-3011
  506. Langer P, Wild A, Celik I , et al. Prospective controlled trial of a standardized meal stimulation test in the detection of pancreaticoduodenal endocrine tumours in patients with multiple endocrine neoplasia type 1. Br J Surg 2001;88:1403-1407
  507. van Beek DJ, Nell S, Pieterman CRC , et al. Prognostic factors and survival in MEN1 patients with gastrinomas: Results from the DutchMEN study group (DMSG). J Surg Oncol 2019;120:966-975
  508. Norton JA, Cornelius MJ, Doppman JL , et al. Effect of parathyroidectomy in patients with hyperparathyroidism, Zollinger-Ellison syndrome and multiple endocrine neoplasia Type I: A prospective study. Surgery 1987;102:958-966
  509. Norton JA, Venzon DJ, Berna MJ , et al. Prospective study of surgery for primary hyperaparathyroidism (HPT) in Multiple Endocrine Neoplasia type 1 (MEN1), and Zollinger-Ellison syndrome (ZES): longterm outcome of a more virulent form of HPT. Ann Surgery 2008;247:501-510
  510. Benya RV, Metz DC, Venzon DJ , et al. Zollinger-Ellison syndrome can be the initial endocrine manifestation in patients with multiple endocrine neoplasia-type 1. Am J Med 1994;97:436-444
  511. Mignon M, Cadiot G. Diagnostic and therapeutic criteria in patients with Zollinger-Ellison syndrome and multiple endocrine neoplasia type 1. J Intern Med 1998;243:489-494
  512. Asgharian B, Turner ML, Gibril F , et al. Cutaneous tumors in patients with MEN1 and gastrinomas: Prospective study of frequency and development of criteria with high sensitivity and specificity. J Clin Endocrinol Metab 2004;89:5328-5336
  513. Asgharian B, Chen YJ, Patronas NJ , et al. Meningiomas may be a component tumor of MEN1. Clin Cancer Res 2004;10:869-880
  514. Gibril F, Chen Y-J, Schrump DS , et al. Prospective study of thymic carcinoids in patients with Multiple Endocrine Neoplasia Type 1. J Clin Endocrinol Metab 2003;88:1066-1081
  515. Corleto VD, Annibale B, Gibril F , et al. Does the widespread use of proton pump inhibitors mask, complicate and/or delay the diagnosis of Zollinger-Ellison syndrome? Aliment Pharmacol Ther 2001;15:1555-1561
  516. Rehfeld JF, Gingras MH, Bardram L , et al. The Zollinger-Ellison Syndrome and Mismeasurement of Gastrin. Gastroenterology 2011;140:1444-1453
  517. Murugesan SV, Varro A, Pritchard DM. Review article: Strategies to determine whether hypergastrinaemia is due to Zollinger Ellison syndrome rather than a more common benign cause. Aliment Pharmacol Ther 2009;29:1055-1068
  518. Metz DC, Strader DB, Orbuch M , et al. Use of omeprazole in Zollinger-Ellison: A prospective nine-year study of efficacy and safety. Aliment Pharmacol Ther 1993;7:597-610
  519. Metz DC, Pisegna JR, Fishbeyn VA , et al. Currently used doses of omeprazole in Zollinger-Ellison syndrome are too high. Gastroenterology 1992;103:1498-1508
  520. Poitras P, Gingras MH, Rehfeld JF. Secretin stimulation test for gastrin release in zollinger-ellison syndrome: to do or not to do? Pancreas 2013;42:903-904
  521. Rehfeld JF, Bardram L, Hilsted L , et al. Pitfalls in diagnostic gastrin measurements. Clin Chem 2012;58:831-836
  522. Rehfeld JF. The art of measuring gastrin in plasma: a dwindling diagnostic discipline? Scand J Clin Lab Invest 2008;68:353-361
  523. Chassany O, Michaux A, Bergmann JF. Drug-induced diarrhoea. Drug Saf 2000;22:53-72
  524. Green FW, Norton RA, Kaplan MM. Pharmacology and clinical use of antacids. Am J Hosp Pharm 1975;32:425-429
  525. Gibril F, Jensen RT. Diagnostic uses of radiolabelled somatostatin-receptor analogues in gastroenteropancreatic endocrine tumors. Dig Liver Dis 2004;36:S106-S120
  526. Saeed ZA, Evans DJ, Jr., Evans DG , et al. Helicobacter pylori and the Zollinger-Ellison syndrome. Dig Dis Sci 1991;36:15-18
  527. Metz DC, Weber HC, Orbuch M , et al. Helicobacter pylori infection: a reversible cause of hypergastrinemia and hyperchlorhydria which can mimic Zollinger-Ellison syndrome. Dig Dis Sci 1995;40:153-159
  528. Fich A, Talley NJ, Shorter RG , et al. Zollinger-Ellison syndrome. Relation to Helicobacter pylori-associated chronic gastritis and gastric acid secretion. Dig Dis Sci 1991;36:10-14
  529. Weber HC, Venzon DJ, Jensen RT , et al. Studies on the interrelation between Zollinger-Ellison syndrome, Helicobacter pylori and proton pump inhibitor therapy. Gastroenterology 1997;112:84-91
  530. Watanabe T, Matsushima Y, Nakase H , et al. Effects of Helicobacter pylori infection on Zollinger-Ellison syndrome. J Gastroenterol 2000;35:735-741
  531. McColl KE. Helicobacter pylori-negative nonsteroidal anti-inflammatory drug-negative ulcer. Gastroenterol Clin North Am 2009;38:353-361
  532. Fishbeyn VA, Norton JA, Benya RV , et al. Assessment and prediction of long-term cure in patients with Zollinger-Ellison syndrome: the best approach. Ann Intern Med 1993;119:199-206
  533. McCarthy DM, Peikin SR, Lopatin RN , et al. Hyperparathyroidism a reversible cause of cimetidine-resistant gastric hypersecretion. Br Med J 1979;1:1765-1766
  534. Winczura A, Saggi B, Savage-Lobeck D. Gastrinoma With Relatively Low Gastrin Levels: A Case Report. Cureus 2023;15:e41686
  535. Borch K, Renvall H, Liedberg G , et al. Relations between circulating gastrin and endocrine cell proliferation in the atrophic gastric fundic mucosa. Scand J Gastroenterol 1986;21:357-363
  536. Vannella L, Sbrozzi-Vanni A, Lahner E , et al. Development of type I gastric carcinoid in patients with chronic atrophic gastritis. Aliment Pharmacol Ther 2011;33:1361-1369
  537. Delle Fave G, Marignani M, Moretti A , et al. Hypergastrinemia and enterochromaffin-like cell hyperplasia. Yale J Biol Med 1998;71:291-301
  538. Dhillo WS, Jayasena CN, Lewis CJ , et al. Plasma gastrin measurement cannot be used to diagnose a gastrinoma in patients on either proton pump inhibitors or histamine type-2 receptor antagonists. Ann Clin Biochem 2006;43:153-155
  539. Lamberts R, Creutzfeldt W, Stockmann F , et al. Long term omeprazole treatment in man: effects on gastric endocrine cell populations. Digestion 1988;39:126-135
  540. Pregun I, Herszenyi L, Juhasz M , et al. Effect of proton-pump inhibitor therapy on serum chromogranin a level. Digestion 2011;84:22-28
  541. Jansen JB, Klinkenberg-Knol EC, Meuwissen SG , et al. Effect of long-term treatment with omeprazole on serum gastrin and serum group A and C pepsinogens in patients with reflux esophagitis. Gastroenterology 1990;99:621-628
  542. Arnold R. Diagnosis and differential diagnosis of hypergastrinemia. Wien Klin Wochenschr 2007;119:564-569
  543. Lundell L, Vieth M, Gibson F , et al. Systematic review: the effects of long-term proton pump inhibitor use on serum gastrin levels and gastric histology. Aliment Pharmacol Ther 2015;42:649-663
  544. Calvete O, Reyes J, Zuniga S , et al. Exome sequencing identifies ATP4A gene as responsible of an atypical familial type I gastric neuroendocrine tumour. Hum Mol Genet 2015;24:2914-2922
  545. Fossmark R, Calvete O, Mjones P , et al. ECL-cell carcinoids and carcinoma in patients homozygous for an inactivating mutation in the gastric H(+) K(+) ATPase alpha subunit. APMIS 2016;124:561-566
  546. Ihara Y, Umeno J, Hori Y. Type IV Gastric Carcinoids in the Stomach Caused by ATP4A Gene Mutations. Clin Gastroenterol Hepatol 2020;18:A22
  547. Calvete O, Varro A, Pritchard DM , et al. A knockin mouse model for human ATP4aR703C mutation identified in familial gastric neuroendocrine tumors recapitulates the premalignant condition of the human disease and suggests new therapeutic strategies. Dis Model Mech 2016;9:975-984
  548. Maltz C, Lightdale CJ. The Zollinger-Ellison syndrome: a new approach. Clin Bull 1979;9:165-167
  549. Sadowski SM, Millo C, Cottle-Delisle C , et al. Results of (68)Gallium-DOTATATE PET/CT Scanning in Patients with Multiple Endocrine Neoplasia Type 1. J Am Coll Surg 2015;221:509-517
  550. Minalyan A, Benhammou JN, Artashesyan A , et al. Autoimmune atrophic gastritis: current perspectives. Clin Exp Gastroenterol 2017;10:19-27
  551. Dixon MF, Genta RM, Yardley JHCP. Classification and grading of gastritis. The upgraded Sydney System. International workshop on the histopathology of gastritis, Houston 1994. Am J Surg Pathol 1996;20:1161-1181
  552. Gibril F, Reynolds JC, Chen CC , et al. Specificity of somatostatin receptor scintigraphy: a prospective study and the effects of false positive localizations on management in patients with gastrinomas. J Nucl Med 1999;40:539-553
  553. Festen HP, Thijs JC, Lamers CB , et al. Effect of oral omeprazole on serum gastrin and serum pepsinogen I levels. Gastroenterology 1984;87:1030-1034
  554. McArthur KE, Collen MJ, Maton PN , et al. Omeprazole: effective, convenient therapy for Zollinger-Ellison syndrome. Gastroenterology 1985;88:939-944
  555. Vinayek R, Amantea MA, Maton PN , et al. Pharmacokinetics of oral and intravenous omeprazole in patients with the Zollinger-Ellison syndrome. Gastroenterology 1991;101:138-147
  556. Metz DC, Pisegna JR, Ringham GL , et al. Prospective study of efficacy and safety of lansoprazole in Zollinger-Ellison syndrome. Dig Dis Sci 1993;38:245-256
  557. Hirschowitz BI, Mohnen J, Shaw S. Long-term treatment with lansoprazole for patients with Zollinger-Ellison syndrome. Aliment Pharmacol Ther 1996;10:507-522
  558. Jensen RT. Use of omeprazole and other proton pump inhibitors in the Zollinger-Ellison syndrome. In: Olbe L ed. Milestones in Drug Therapy. Basel, Switzerland: Birkhauser Verlag AG Publish. Co.; 1999:205-221.
  559. Vinayek R, Howard JM, Maton PN , et al. Famotidine in the therapy of gastric hypersecretory states. Am J Med 1986;81:49-59
  560. Jensen RT, Collen MJ, McArthur KE , et al. Comparison of the effectiveness of ranitidine and cimetidine in inhibiting acid secretion in patients with gastric acid hypersecretory states. Am J Med 1984;77(5B):90-105
  561. Howard JM, Chremos AN, Collen MJ , et al. Famotidine, a new, potent, long-acting histamine H2-receptor antagonist: comparison with cimetidine and ranitidine in the treatment of Zollinger-Ellison syndrome. Gastroenterology 1985;88:1026-1033
  562. Jensen RT. Basis for failure of cimetidine in patients with Zollinger- Ellison syndrome. Dig Dis Sci 1984;29:363-366
  563. Jensen RT. Endocrine tumors of the Gastrointestinal Tract and Pancreas. In: Longo DL, Fauci AS, Kasper DL et al.  eds. Harrison's Principles of Internal Medicine. New York: McGraw-Hill companies; 2012:3056-3072.
  564. Arnold R. Endocrine tumours of the gastrointestinal tract. Introduction: definition, historical aspects, classification, staging, prognosis and therapeutic options. Best Pract Res Clin Gastroenterol 2005;19:491-505
  565. Kulke MH, Shah MH, Benson AB , et al. Neuroendocrine Tumors: version 3.2017: NCCN Clinical Practice Guidelines in Oncology. NCCN Clinical Practice Guidelines in Oncology 2017;1-116
  566. Oh DS, Wang HS, Ohning GV , et al. Validation of a new endoscopic technique to assess acid output in Zollinger-Ellison syndrome. Clin Gastroenterol Hepatol 2006;4:1467-1473
  567. Gibril F, Lindeman RJ, Abou-Saif A , et al. Retained gastric antrum syndrome. A forgotten, treatable cause of refractory peptic ulcer disease. Dig Dis Sci 2001;46:610-617
  568. Chu S, Schubert ML. Gastric secretion. Curr Opin Gastroenterol 2012;28:587-593
  569. Malfertheiner P. The intriguing relationship of Helicobacter pylori infection and acid secretion in peptic ulcer disease and gastric cancer. Dig Dis 2011;29:459-464
  570. Deveney CW, Deveney KE, Jones S, Jaffe BM. Calcium and secretin tests in diagnosis of gastrinoma. Gastroenterology 70, 968. 1976.
  571. Annibale B, Rindi G, D'Ambra G , et al. Antral gastrin cell hyperfunction and helicobacter pylori infection. Aliment Pharmacol Ther 1996;10:607-615
  572. Shibata C, Kakyo M, Kinouchi M , et al. Criteria for the glucagon provocative test in the diagnosis of gastrinoma. Surg Today 2013;43:1281-1285
  573. Feldman M, Schiller LR, Walsh JH , et al. Positive intravenous secretin test in patients with achlorhydria-related hypergastrinemia. Gastroenterology 1987;93:59-62
  574. Shah P, Singh MH, Yang YX , et al. Hypochlorhydria and achlorhydria are associated with false-positive secretin stimulation testing for zollinger-ellison syndrome. Pancreas 2013;42:932-936
  575. Leung D, Schwartz L. Imaging of neuroendocrine tumors. Semin Oncol 2013;40:109-119
  576. Sundin A. Radiological and nuclear medicine imaging of gastroenteropancreatic neuroendocrine tumours. Best Pract Res Clin Gastroenterol 2012;26:803-818
  577. Klose KJ, Heverhagen JT. Localisation and staging of gastrin producing tumours using cross-sectional imaging modalities. Wien Klin Wochenschr 2007;119:588-592
  578. Singh A, Hines JJ, Friedman B. Multimodality Imaging of the Pancreatic Neuroendocrine Tumors. Semin Ultrasound CT MR 2019;40:469-482
  579. Fottner C, Ferrata M, Weber MM. Hormone secreting gastro-entero-pancreatic neuroendocrine neoplasias (GEP-NEN): When to consider, how to diagnose? Rev Endocr Metab Disord 2017;18:393-410
  580. Gibril F, Doppman JD, Jensen RT. Comparative analysis of tumor localization techniques for neuroendocrine tumors. Yale J Biol Med 1997;70:481-500
  581. Jensen RT. Carcinoid and pancreatic endocrine tumors: recent advances in molecular pathogenesis, localization, and treatment. Curr Opin Oncol 2000;12:368-377
  582. Szidonya L, Park EA, Kwak JJ , et al. Molecular and Anatomic Imaging of Neuroendocrine Tumors. Surg Oncol Clin N Am 2022;31:649-671
  583. Rockall AG, Reznek RH. Imaging of neuroendocrine tumours (CT/MR/US). Best Pract Res Clin Endocrinol Metab 2007;21:43-68
  584. Wank SA, Doppman JL, Miller DL , et al. Prospective study of the ability of computerized axial tomography to localize gastrinomas in patients with Zollinger-Ellison syndrome. Gastroenterology 1987;92:905-912
  585. Doppman JL, Miller DL, Chang R , et al. Gastrinomas: localization by means of selective intraarterial injection of secretin. Radiology 1990;174:25-29
  586. Metz DC, Jensen RT. Advances in gastric antisecretory therapy in Zollinger-Ellison syndrome. In: Mignon M, Jensen RT eds. Endocrine Tumors of the Pancreas: Recent advances in research and management. Series: Frontiers of Gastrointestinal Research. Basel, Switzerland: S. Karger; 1995:240-257.
  587. Sundin A. Imaging of neuroendocrine tumors. Expert Opin Med Diagn 2012;6:473-483
  588. Hussain JS, Srinivasa RN, Hage A , et al. Bringing SASI back: Single session selective arterial secretin injection and transarterial embolization of intrahepatic pancreatic neuroendocrine metastasis in a MEN-1 patient. Radiol Case Rep 2018;13:333-335
  589. Hayashi R, Minami I, Sasahara Y , et al. Diagnostic accuracy of selective arterial calcium injection test for localization of gastrinoma. Endocr J 2020;67:305-315
  590. Manoharan J, Bollmann C, Kann PH , et al. Gender Differences in Multiple Endocrine Neoplasia Type 1: Implications for Screening? Visc Med 2020;36:3-9
  591. Orbuch M, Doppman JL, Strader DB et al.  Imaging for pancreatic endocrine tumor localization: recent advances. In: Mignon M, Jensen RT eds. Endocrine Tumors of the Pancreas: Recent advances in research and management.  Frontiers of Gastrointestinal Research. Basel, Switzerland: S. Karger; 1995:268-281.
  592. Yang J, Kan Y, Ge BH , et al. Diagnostic role of Gallium-68 DOTATOC and Gallium-68 DOTATATE PET in patients with neuroendocrine tumors: a meta-analysis. Acta Radiol 2014;55:389-398
  593. Strader DB, Doppman JL, Orbuch M et al.  Functional localization of pancreatic endocrine tumors. In: Mignon M, Jensen RT eds. Endocrine Tumors of the Pancreas: Recent Advances in Research and Management. Series: Frontiers of Gastrointestinal Research. Basel, Switzerland: Karger Publishing Co.; 1995:282-297.
  594. Thom AK, Norton JA, Doppman JL , et al. Prospective study of the use of intraarterial secretin injection and portal venous sampling to localize duodenal gastrinomas. Surgery 1992;112(#6):1002-1008
  595. Miller DL, Doppman JL, Metz DC , et al. Zollinger-Ellison syndrome: technique, results and complications of portal venous sampling. Radiology 1992;182:235-241
  596. Gibril F, Doppman JL, Chang R , et al. Metastatic gastrinomas: localization with selective arterial injection of secretin. Radiology 1996;198:77-84
  597. Imamura M, Takahashi K. Use of selective arterial secretin injection test to guide surgery in patients with Zollinger-Ellison syndrome. World J Surg 1993;17:433-438
  598. Vannella L, Lahner E, Annibale B. Risk for gastric neoplasias in patients with chronic atrophic gastritis: a critical reappraisal. World J Gastroenterol 2012;18:1279-1285
  599. Otto AI, Marschalko M, Zalatnai A , et al. Glucagon cell adenomatosis: a new entity associated with necrolytic migratory erythema and glucagonoma syndrome. J Am Acad Dermatol 2011;65:458-459
  600. el-Omar EM, Penman ID, Ardill JE , et al. Helicobacter pylori infection and abnormalities of acid secretion in patients with duodenal ulcer disease. Gastroenterology 1995;109:681-691
  601. Kwekkeboom DJ, Kam BL, Van Essen M , et al. Somatostatin-receptor-based imaging and therapy of gastroenteropancreatic neuroendocrine tumors. Endocr Relat Cancer 2010;17:R53-R73
  602. Behe M, Gotthardt M, Behr TM. Imaging of gastrinomas by nuclear medicine methods. Wien Klin Wochenschr 2007;119:593-596
  603. Oberg KE, Reubi JC, Kwekkeboom DJ , et al. Role of somatostatins in gastroenteropancreatic neuroendocrine tumor development and therapy. Gastroenterology 2010;139:742-53, 753
  604. Kong G, Hicks RJ. Peptide Receptor Radiotherapy: Current Approaches and Future Directions. Curr Treat Options Oncol 2019;20:77
  605. Kwekkeboom DJ, Krenning EP. Peptide Receptor Radionuclide Therapy in the Treatment of Neuroendocrine Tumors. Hematol Oncol Clin North Am 2016;30:179-191
  606. Murtha TD, Lupsa BC, Majumdar S , et al. A Systematic Review of Proinsulin-Secreting Pancreatic Neuroendocrine Tumors. J Gastrointest Surg 2017;21:1335-1341
  607. Bushnell DL, Bodeker KL. Overview and Current Status of Peptide Receptor Radionuclide Therapy. Surg Oncol Clin N Am 2020;29:317-326
  608. Feijtel D, de Jong M, Nonnekens J. Peptide receptor radionuclide therapy: Looking back, looking forward. Curr Top Med Chem 2020;20:2959-2969
  609. La Salvia A, Modica R, Rossi RE , et al. Targeting neuroendocrine tumors with octreotide and lanreotide: Key points for clinical practice from NET specialists. Cancer Treat Rev 2023;117:102560
  610. Alexander HR, Fraker DL, Norton JA , et al. Prospective study of somatostatin receptor scintigraphy and its effect on operative outcome in patients with Zollinger-Ellison syndrome. Ann Surg 1998;228:228-238
  611. Barton JC, Hirschowitz BI, Maton PN , et al. Bone metastases in malignant gastrinoma. Gastroenterology 1986;91:1179-1185
  612. Mathew A, Fendler WP, Theysohn J , et al. Bone Metastases in Patients with Pancreatic NETs: Prevalence and Prognosis. Horm Metab Res 2023 (In Press)
  613. Cives M, Pelle E, Rinzivillo M , et al. BONE METASTASES IN NEUROENDOCRINE TUMORS: MOLECULAR PATHOGENESIS AND IMPLICATIONS IN CLINICAL PRACTICE. Neuroendocrinology 2020;111:207-216
  614. Ruszniewski P, Amouyal P, Amouyal G , et al. Localization of gastrinomas by endoscopic ultrasonography in patients with Zollinger-Ellison syndrome. Surgery 1995;117:629-635
  615. Thompson NW, Czako PF, Fritts LL , et al. Role of endoscopic ultrasonography in the localization of insulinomas and gastrinomas. Surgery 1994;116:1131-1138
  616. Rosch T, Lightdale CJ, Botet JF , et al. Localization of pancreatic endocrine tumors by endoscopic ultrasonography. N Engl J Med 1992;326:1721-1726
  617. Kamata K, Ashida R, Yasukawa S , et al. Histological diagnosis and grading of pancreatic neuroendocrine tumor by endoscopic ultrasound-guided fine needle biopsy using a 25-gauge needle with a core trap: A multicenter prospective trial. Pancreatology 2020;20:1428-1433
  618. Kann PH. Is endoscopic ultrasonography more sensitive than magnetic resonance imaging in detecting and localizing pancreatic neuroendocrine tumors? Rev Endocr Metab Disord 2018;19:133-137
  619. Thomas-Marques L, Murat A, Delemer B , et al. Prospective endoscopic ultrasonographic evaluation of the frequency of nonfunctioning pancreaticoduodenal endocrine tumors in patients with multiple endocrine neoplasia type 1. Am J Gastroenterol 2006;101:266-273
  620. Kann PH, Balakina E, Ivan D , et al. Natural course of small, asymptomatic neuroendocrine pancreatic tumours in multiple endocrine neoplasia type 1: an endoscopic ultrasound imaging study. Endocr Relat Cancer 2006;13:1195-1202
  621. Binderup T, Knigge U, Loft A , et al. 18F-fluorodeoxyglucose positron emission tomography predicts survival of patients with neuroendocrine tumors. Clin Cancer Res 2010;16:978-985
  622. Carideo L, Prosperi D, Panzuto F , et al. Role of Combined ((68)Ga)Ga-DOTA-SST Analogues and ((18)F)FDG PET/CT in the Management of GEP-NENs: A Systematic Review. J Clin Med 2019;8:1032
  623. Mohamed A, Asa SL, Lee Z , et al. The predictive impact of dual somatostatin receptor/fluorodeoxyglucose (FDG) positron emission tomography (PET) in metastatic gastroenteropancreatic neuroendocrine tumors (GEP-NETs): review of literature and a single institution experience. J Gastrointest Oncol 2023;14:1087-1094
  624. Dromain C, Deandreis D, Scoazec JY , et al. Imaging of neuroendocrine tumors of the pancreas. Diagn Interv Imaging 2016;97:1241-1257
  625. Naswa N, Sharma P, Gupta SK , et al. Dual tracer functional imaging of gastroenteropancreatic neuroendocrine tumors using 68Ga-DOTA-NOC PET-CT and 18F-FDG PET-CT: competitive or complimentary? Clin Nucl Med 2014;39:e27-e34
  626. Rinzivillo M, Partelli S, Prosperi D , et al. Clinical Usefulness of (18)F-Fluorodeoxyglucose Positron Emission Tomography in the Diagnostic Algorithm of Advanced Entero-Pancreatic Neuroendocrine Neoplasms. Oncologist 2018;23:186-192
  627. Sharma P, Naswa N, Kc SS , et al. Comparison of the prognostic values of Ga-DOTANOC PET/CT and F-FDG PET/CT in patients with well-differentiated neuroendocrine tumor. Eur J Nucl Med Mol Imaging 2014;41:2194-2202
  628. Has Simsek D, Kuyumcu S, Turkmen C , et al. Can complementary 68Ga-DOTATATE and 18F-FDG PET/CT establish the missing link between histopathology and therapeutic approach in gastroenteropancreatic neuroendocrine tumors? J Nucl Med 2014;55:1811-1817
  629. Hindie E. The NETPET Score: Combining FDG and Somatostatin Receptor Imaging for Optimal Management of Patients with Metastatic Well-Differentiated Neuroendocrine Tumors. Theranostics 2017;7:1159-1163
  630. Chan DL, Pavlakis N, Schembri GP , et al. Dual Somatostatin Receptor/FDG PET/CT Imaging in Metastatic Neuroendocrine Tumours: Proposal for a Novel Grading Scheme with Prognostic Significance. Theranostics 2017;7:1149-1158
  631. Nilica B, Waitz D, Stevanovic V , et al. Direct comparison of (68)Ga-DOTA-TOC and (18)F-FDG PET/CT in the follow-up of patients with neuroendocrine tumour treated with the first full peptide receptor radionuclide therapy cycle. Eur J Nucl Med Mol Imaging 2016;43:1585-1592
  632. Ito T, Hijioka S, Masui T , et al. Advances in the diagnosis and treatment of pancreatic neuroendocrine neoplasms in Japan. J Gastroenterol 2017;52:9-18
  633. Mignon M, Bonfils S. Diagnosis and treatment of Zollinger-Ellison syndrome. Bailliere's Clin Gastroenterol 1988;2:677-698
  634. Maton PN, Gardner JD, Jensen RT. Diagnosis and management of Zollinger-Ellison syndrome. Endocrinol Metab Clin North Am 1989;18:519-543
  635. Atalar K, Warren OJ, Jacyna M , et al. Laparoscopic resection for primary lymph node gastrinoma. Pancreas 2013;42:723-725
  636. Jensen RT. Gastrin-producing tumors. Cancer Treat Res 1997;89:293-334
  637. Jensen RT, Maton PN, Gardner JD. Current management of Zollinger-Ellison syndrome. Drugs 1986;32:188-196
  638. Fox PS, Hofmann JW, Wilson SD , et al. Surgical management of the Zollinger-Ellison syndrome. Surg Clin North Am 1974;54:395-407
  639. Jensen RT. Natural history of digestive endocrine tumors. In: Mignon M, Colombel JF eds. Recent advances in pathophysiology and management of inflammatory bowel diseases and digestive endocrine tumors. Paris, France: John Libbey Eurotext Publishing Co.; 1999:192-219.
  640. Quatrini M, Castoldi L, Rossi G , et al. A follow-up study of patients with Zollinger-Ellison syndrome in the period 1966-2002: effects of surgical and medical treatments on long-term survival. J Clin Gastroenterol 2005;39:376-380
  641. Zollinger RM, Ellison EC, O'Dorisio TM , et al. Thirty years' experience with gastrinoma. World J Surg 1984;8:427-435
  642. Jensen RT. Gastrinomas: advances in diagnosis and management. Neuroendocrinology 2004;80:23-27
  643. Malagelada JR, Edis AJ, Adson MA , et al. Medical and surgical options in the management of patients with gastrinoma. Gastroenterology 1983;84:1524-1532
  644. Franz RC, Penzhorn HO. Is total gastrectomy still a viable option in the management of patients with the Zollinger-Ellison syndrome? S Afr J Surg 2007;45:58-60
  645. Stroker E, Leone L, Vandeput Y , et al. Severe symptomatic hypomagnesaemia induced by the chronic use of proton pump inhibitors: a case report of a patient with Zollinger-Ellison syndrome. Acta Clin Belg 2014;69:62-65
  646. Richardson CT, Walsh JH. The value of a histamine H2-receptor antagonist in the management of patients with the Zollinger-Ellison syndrome. N Engl J Med 1976;294:133-135
  647. Richardson CT, Feldman M, McClelland RN , et al. Effect of vagotomy in Zollinger-Ellison syndrome. Gastroenterology 1979;77:682-686
  648. McArthur KE, Richardson CT, Barnett CC , et al. Laparotomy and proximal gastric vagotomy in Zollinger-Ellison syndrome: results of a 16-year prospective study. Am J Gastroenterol 1996;91:1104-1111
  649. Lamas C, Navarro E, Casteras A , et al. MEN1-associated primary hyperparathyroidism in the Spanish Registry: clinical characterictics and surgical outcomes. Endocr Connect 2019;8:1416-1424
  650. Goudet P, Cougard P, Vergès B , et al. Hyperparathyroidism in multiple endocrine neoplasia type I: surgical trends and results of a 256-patient series from Groupe d'Etude des Néoplasies Endocriniennes Multiples study group. World J Surg 2001;25:886-890
  651. Nilubol N, Weinstein LS, Simonds WF , et al. Limited Parathyroidectomy in Multiple Endocrine Neoplasia Type 1-Associated Primary Hyperparathyroidism: A Setup for Failure. Ann Surg Oncol 2016;23:416-423
  652. Kivlen MH, Bartlett DL, Libutti SK , et al. Reoperation for hyperparathyroidism in multiple endocrine neoplasia type 1. Surgery 2001;130:991-998
  653. Elaraj DM, Skarulis MC, Libutti SK , et al. Results of initial operation for hyperparathryoidism (HPT) in patients with Multiple Endocrine Neoplasia Type 1 (MEN1). Surgery 2003;134:858-864
  654. Pisegna JR, Norton JA, Slimak GG , et al. Effects of curative resection on gastric secretory function and antisecretory drug requirement in the Zollinger-Ellison syndrome. Gastroenterology 1992;102:767-778
  655. Ojeaburu JV, Ito T, Crafa P , et al. Mechanism of Acid hypersecretion post curative gastrinoma resection. Dig Dis Sci 2011;56:139-154
  656. Metz DC, Benya RV, Fishbeyn VA , et al. Prospective study of the need for long-term antisecretory therapy in patients with Zollinger-Ellison syndrome following successful curative gastrinoma resection. Aliment Pharmacol Ther 1993;7(#3):247-257
  657. Weinstein MC, Feinberg HV. Clinical decision analysis. Philadelphia, PA: W.B. Saunders; 1980.
  658. Maton PN, Vinayek R, Frucht H , et al. Long-term efficacy and safety of omeprazole in patients with Zollinger-Ellison syndrome: a prospective study. Gastroenterology 1989;97:827-836
  659. Maton PN, Frucht H, Vinayek R , et al. Medical management of patients with Zollinger-Ellison syndrome who have had previous gastric surgery: A prospective study. Gastroenterology 1988;94:294-299
  660. Gibril F, Jensen RT. Zollinger-Ellison syndrome revisited: diagnosis, biologic markers, associated inherited disorders, and acid hypersecretion. Curr Gastroenterol Rep 2004;6:454-463
  661. Hirschowitz BI, Simmons J, Mohnen J. Clinical outcome using lansoprazole in acid hypersecretors with and without Zollinger-Ellison syndrome: a 13-year prospective study. Clin Gastroenterol Hepatol 2005;3:39-48
  662. Hirschowitz BI, Simmons J, Mohnen J. Long-term lansoprazole control of gastric acid and pepsin secretion in ZE and non-ZE hypersecretors: a prospective 10-year study. Aliment Pharmacol Ther 2001;15:1795-1806
  663. Termanini B, Gibril F, Stewart CA , et al. A prospective study of the effectiveness of low dose omeprazole as initial therapy in Zollinger-Ellison syndrome. Aliment Pharmacol Ther 1996;10:61-71
  664. Heidelbaugh JJ, Metz DC, Yang YX. Proton pump inhibitors: are they overutilised in clinical practice and do they pose significant risk? Int J Clin Pract 2012;66:582-591
  665. Frucht H, Maton PN, Jensen RT. Use of omeprazole in patients with the Zollinger-Ellison syndrome. Dig Dis Sci 1991;36:394-404
  666. Yang YX, Metz DC. Safety of proton pump inhibitor exposure. Gastroenterology 2010;139:1115-1127
  667. Ito T, Jensen RT. Association of long-term proton pump inhibitor therapy with bone fractures and effects on absorption of calcium, vitamin b(12), iron, and magnesium. Curr Gastroenterol Rep 2010;12:448-457
  668. Linder L, Tamboue C, Clements JN. Drug-Induced Vitamin B12 Deficiency: A Focus on Proton Pump Inhibitors and Histamine-2 Antagonists. J Pharm Pract 2017;30:639-642
  669. Jensen RT. Involvement of cholecystokinin/gastrin-related peptides and their receptors in clinical gastrointestinal disorders. Pharmacol Toxicol 2002;91:333-350
  670. Termanini B, Gibril F, Sutliff VE, III , et al. Effect of long-term gastric acid suppressive therapy on serum vitamin B12 levels in patients with Zollinger-Ellison syndrome. Am J Med 1998;104:422-430
  671. Hirschowitz BI, Worthington J, Mohnen J. Vitamin B12 deficiency in hypersecretors during long-term acid suppression with proton pump inhibitors. Aliment Pharmacol Ther 2008;27:1110-1121
  672. Eyal A, Sueissa A, Braun E , et al. From hypomagnesaemia to Zollinger-Ellison syndrome: an adverse effect of a proton pump inhibitor. BMJ Case Rep 2014;2014:bcr2014205165
  673. Stewart CA, Termanini B, Sutliff VE , et al. Assessment of the risk of iron malabsorption in patients with Zollinger-Ellison syndrome treated with long-term gastric acid antisecretory therapy. Aliment Pharmacol Ther 1998;12:83-98
  674. Yang YX, Lewis JD, Epstein S , et al. Long-term proton pump inhibitor therapy and risk of hip fracture. JAMA 2006;296:2947-2953
  675. Nehra AK, Alexander JA, Loftus CG , et al. Proton Pump Inhibitors: Review of Emerging Concerns. Mayo Clin Proc 2018;93:240-246
  676. Jaynes M, Kumar AB. The risks of long-term use of proton pump inhibitors: a critical review. Ther Adv Drug Saf 2018;10:2042098618809927
  677. Metz DC, Sostek MB, Ruszniewski P , et al. Effects of esomeprazole on Acid output in patients with zollinger-ellison syndrome or idiopathic gastric Acid hypersecretion. Am J Gastroenterol 2007;102:2648-2654
  678. Deveney CW, Stein S, Way LW. Cimetidine in the treatment of Zollinger-Ellison syndrome. Am J Surg 1983;146:116-123
  679. Pratap T, Jalal MJA, Jacob D , et al. Radiological Features of Zollinger-Ellison Syndrome: A Report of Two Cases. Indian J Radiol Imaging 2022;32:395-402
  680. Venkat PG, Longstreth GF. Gastroesophageal Reflux Disease and the Zollinger-Ellison Syndrome. Am J Gastroenterol 2022;117:1012-1013
  681. Simmons LH, Guimaraes AR, Zukerberg LR. Case records of the Massachusetts General Hospital. Case 6-2013. A 54-year-old man with recurrent diarrhea. N Engl J Med 2013;368:757-765
  682. Bhattacharya S, Blau JE, Koh C. Acute oesophageal necrosis in multiple endocrine neoplasia type 1: an undescribed complication. BMJ Case Rep 2021;14:e238214
  683. Matsubayashi H, Kawata N, Kakushima N , et al. A case of type 1 multiple endocrine neoplasia with esophageal stricture successfully treated with endoscopic balloon dilation and local steroid injection combined with surgical resection of gastrinomas. BMC Gastroenterol 2017;17:37
  684. Gaztambide S, Vazquez JA. Short- and long-term effect of a long-acting somatostatin analogue, lanreotide (SR-L) on metastatic gastrinoma. J Endocrinol Invest 1999;22:144-146
  685. Wilson SD, Doffek KM, Krzywda EA , et al. Zollinger-Ellison syndrome associated with a history of alcohol abuse: Coincidence or consequence? Surgery 2011;150:1129-1135
  686. Thodiyil PA, El-Masry NS, Williamson RC. Supergastrinoma: simultaneous peptic ulceration of esophagus, stomach, and small intestine. Int Surg 2003;88:155-158
  687. Lloyd-Davies KA, Rutgersson K, Solvell L. Omeprazole in the treatment of Zollinger-Ellison syndrome: a 4-year international study. Aliment Pharmacol Ther 1988;2:13-32
  688. Morocutti A, Merrouche M, Bjaaland T , et al. An open-label study of rabeprazole in patients with Zollinger-Ellison syndrome or idiopathic gastric acid hypersecretion. Aliment Pharmacol Ther 2006;24:1439-1444
  689. Metz DC, Comer GM, Soffer E , et al. Three-year oral pantoprazole administration is effective for patients with Zollinger-Ellison syndrome and other hypersecretory conditions. Aliment Pharmacol Ther 2006;23:437-444
  690. Riff BP, Leiman DA, Bennett B , et al. Weight Gain in Zollinger-Ellison Syndrome After Acid Suppression. Pancreas 2016;45:193-197
  691. Paul G, Ramdani A, Mignon M , et al. (Comparative efficacy of lansoprazole and omeprazole on the intragastric pH measured over a period of 24 hours and on the basal). Gastroenterol Clin Biol 1994;18:695-701
  692. Mignon M, Hochlaf S, Forestier S , et al. (Dose-response effect of lansoprazole in patients with Zollinger-Ellison syndrome). Gastroenterol Clin Biol 1994;18:13-16
  693. Ramdani A, Mignon M, Samoyeau R. Effect of pantoprazole versus other proton pump inhibitors on 24-hour intragastric pH and basal acid output in Zollinger-Ellison syndrome. Gastroenterol Clin Biol 2002;26:355-359
  694. Strand DS, Kim D, Peura DA. 25 Years of Proton Pump Inhibitors: A Comprehensive Review. Gut Liver 2017;11:27-37
  695. Katz PO, Dunbar KB, Schnoll-Sussman FH , et al. ACG Clinical Guideline for the Diagnosis and Management of Gastroesophageal Reflux Disease. Am J Gastroenterol 2022;117:27-56
  696. Chinzon D, Domingues G, Tosetto N , et al. SAFETY OF LONG-TERM PROTON PUMP INHIBITORS: FACTS AND MYTHS. Arq Gastroenterol 2022;59:219-225
  697. Richter JE, Pandol SJ, Castell DO , et al. Gastroesophageal reflux disease in the Zollinger-Ellison syndrome. Ann Intern Med 1981;95:37-43
  698. Waldum HL, Fossmark R, Bakke I , et al. Hypergastrinemia in animals and man: causes and consequences. Scand J Gastroenterol 2004;39:505-509
  699. Waldum HL, Sordal O, Fossmark R. Proton pump inhibitors (PPIs) may cause gastric cancer - clinical consequences. Scand J Gastroenterol 2018;53:639-642
  700. Wormsley KG. Is chronic long-term inhibition of gastric secretion really dangerous. Scand J Gastroenterol Suppl 1988;146:166-174
  701. Mignon M, Lehy T, Bonnefond A , et al. Development of gastric argyrophil carcinoid tumors in a case of Zollinger-Ellison syndrome with primary hyperparathyroidism during long term antisecretory treatment. Cancer 1987;59:1959-1962
  702. Norton JA, Melcher ML, Gibril F , et al. Gastric carcinoid tumors in multiple endocrine neoplasia-1 patients with Zollinger-Ellison syndrome can be symptomatic, demonstrate aggressive growth, and require surgery. Surgery 2004;136:1267-1274
  703. Scherubl H, Cadiot G, Jensen RT , et al. Neuroendocrine tumors of the stomach (gastric carcinoids) are on the rise: small tumors, small problems? Endoscopy 2010;42:664-671
  704. Bordi C, D'Adda T, Azzoni C , et al. Hypergastrinemia and gastric enterochromaffin-like cells. Am J Surg Pathol 1995;19:S8-S19
  705. Borch K, Renvall H, Liedberg G. Gastric endocrine cell hyperplasia and carcinoid tumors in pernicious anemia. Gastroenterology 1985;88:638-648
  706. Hage E, Hendel L, Gustafsen J , et al. Histopathology of the gastric oxyntic mucosa in two different patient groups during long-term treatment with omeprazole. Eur J Gastroenterol Hepatol 2003;15:781-789
  707. Konturek SJ, Konturek PC, Bielanski W , et al. Case presentation of gastrinoma combined with gastric carcinoid with the longest survival record--Zollinger-Ellison syndrome: pathophysiology, diagnosis and therapy. Med Sci Monit 2002;8:CS43-CS59
  708. Lee HW, Chung JW, Kim YJ , et al. Synchronous Peripancreatic Lymph Node Gastrinoma and Gastric Neuroendocrine Tumor Type 2. Clin Endosc 2016;49:483-487
  709. Tariq H, Kamal MU, Vootla V , et al. A Rare Cause of Abdominal Pain and Mass in an 18-Year-Old Patient: A Diagnostic Dilemma. Gastroenterology Res 2018;11:75-78
  710. Campana D, Ravizza D, Ferolla P , et al. Risk factors of type 1 gastric neuroendocrine neoplasia in patients with chronic atrophic gastritis. A retrospective, multicentre study. Endocrine 2017;56:633-638
  711. Massironi S, Zilli A, Elvevi A , et al. The changing face of chronic autoimmune atrophic gastritis: an updated comprehensive perspective. Autoimmun Rev 2019;18:215-222
  712. Vinayek R, Hahne WF, Euler AR , et al. Parenteral control of gastric hypersecretion in patients with Zollinger-Ellison syndrome. Dig Dis Sci 1993;38:1857-1865
  713. Saeed ZA, Norton JA, Frank WO , et al. Parenteral antisecretory drug therapy in patients with Zollinger- Ellison syndrome. Gastroenterology 1989;96:1393-1402
  714. Metz DC, Forsmark C, Lew EA , et al. Replacement of oral proton pump inhibitors with intravenous pantoprazole to effectively control gastric acid hypersecretion in patients with Zollinger-Ellison syndrome. Am J Gastroenterol 2001;96:3274-3280
  715. Vinayek R, Frucht H, London JF , et al. Intravenous omeprazole in patients with Zollinger-Ellison syndrome undergoing surgery. Gastroenterology 1990;99:10-16
  716. Pang SH, Graham DY. A clinical guide to using intravenous proton-pump inhibitors in reflux and peptic ulcers. Therap Adv Gastroenterol 2010;3:11-22
  717. Maton PN, Gardner JD, Jensen RT. Recent advances in the management of gastric acid hypersecretion in patients with Zollinger-Ellison syndrome. Gastroenterol Clin North Am 1989;18:847-863
  718. Jensen RT, Collen MJ, Allende HD , et al. Cimetidine-induced impotence and breast changes in patients with gastric hypersecretory states. N Engl J Med 1983;308:883-887
  719. Jensen RT, Metz DC, Koviack PD , et al. Prospective study of the long-term efficacy and safety of lansoprazole in patients with Zollinger-Ellison syndrome. Aliment Pharmacol Ther 1993;7 Suppl 1:41-50
  720. Hajri A, Aprahamian M, Damge C. Effect of a new CCK-receptor antagonist, CR 1409, on pancreatic growth induced by caerulein, CCK-8, bombesin and gastrin-releasing peptide in the rat. Digestion 1989;43:66-72
  721. Norton JA, Jensen RT. Unresolved surgical issues in the management of patients with the Zollinger-Ellison syndrome. World J Surg 1991;15:151-159
  722. Ellison EC, Sparks J, Verducci JS , et al. 50-year appraisal of gastrinoma: recommendations for staging and treatment. J Am Coll Surg 2006;202:897-905
  723. Lorenz K, Dralle H. Surgical treatment of sporadic gastrinoma. Wien Klin Wochenschr 2007;119:597-601
  724. Fendrich V, Langer P, Waldmann J , et al. Management of sporadic and multiple endocrine neoplasia type 1 gastrinomas. Br J Surg 2007;94:1331-1341
  725. Fendrich V, Bartsch DK. Surgical Therapy of Sporadic Pancreatic Neuroendocrine Neoplasias G1/G2. Visc Med 2017;33:344-350
  726. Norton JA. Neuroendocrine tumors of the pancreas and duodenum. Curr Probl Surg 1994;31:1-156
  727. Norton JA, Jensen RT. Role of surgery in Zollinger-Ellison syndrome. J Am Coll Surg 2007;205:S34-S37
  728. Mignon M, Ruszniewski P, Haffar S , et al. Current approach to the management of tumoral process in patients with gastrinoma. World J Surg 1986;10:703-710
  729. McCarthy DM. The place of surgery in the Zollinger-Ellison syndrome. N Engl J Med 1980;302:1344-1347
  730. Hirschowitz BI. Surgery to cure the Zollinger-Ellison syndrome. N Engl J Med 1999;341:2096-2097
  731. Hirschowitz BI. Clinical course of nonsurgically treated Zollinger-Ellison syndrome. In: Mignon M, Jensen RT eds. Endocrine Tumors of the Pancreas: Recent advances in research and management. Frontiers of Gastrointestinal Research. Basel, Switzerland: S. Karger; 1995:360-371.
  732. Fraker DL, Norton JA, Alexander HR , et al. Surgery in Zollinger-Ellison syndrome alters the natural history of gastrinoma. Ann Surg 1994;220:320-330
  733. Norton JA, Fraker DL, Alexander HR , et al. Surgery increases survival in patients with gastrinoma. Ann Surg 2006;244:410-419
  734. Zollinger RM. Gastrinoma: factors influencing prognosis. Surgery 1985;97:49-54
  735. McNamara D, Lewis T, O'Moran C. Zollinger-Ellison syndrome with fasting hypoglycaemia. J Royal Soc Med 1998;91:92-93
  736. Wilcox CM, Seay T, Arcury JT , et al. Zollinger-Ellison syndrome: presentation, response to therapy, and outcome. Dig Liver Dis 2011;43:439-443
  737. Collins JSA, Buchanan KD, Kennedy TL , et al. Changing patterns in presentation and management of the Zollinger-Ellison syndrome in Northern Ireland, 1970-1988. Q J Med 1991;78:215-225
  738. Hoffmann KM, Furukawa M, Jensen RT. Duodenal neuroendocrine tumors: Classification, functional syndromes, diagnosis and medical treatment. Best Pract Res Clin Gastroenterol 2005;19:675-697
  739. Harper S, Carroll RW, Frilling A , et al. Primary lymph node gastrinoma: 2 cases and a review of the literature. J Gastrointest Surg 2015;19:651-655
  740. Norton JA. Intraoperative methods to stage and localize pancreatic and duodenal tumors. Ann Oncol 1999;10:182-184
  741. Norton JA, Cromack DT, Shawker TH , et al. Intraoperative ultrasonographic localization of islet cell tumors. A prospective comparison to palpation. Ann Surg 1988;207:160-168
  742. Norton JA. Surgery and prognosis of duodenal gastrinoma as a duodenal neuroendocrine tumor. Best Pract Res Clin Gastroenterol 2005;19:699-704
  743. Norton JA. Surgical treatment of islet cell tumors with special emphasis on operative ultrasound. In: Mignon M, Jensen RT eds. Endocrine Tumors of the Pancreas: Recent advances in research and management. Frontiers in Gastrointestinal Research. Basel, Switzerland: S. Karger; 1995:309-332.
  744. Maton PN, Mackem SM, Norton JA , et al. Ovarian carcinoma as a cause of Zollinger-Ellison syndrome. Natural history, secretory products and response to provocative tests. Gastroenterology 1989;97:468-471
  745. Carty SE, Jensen RT, Norton JA. Prospective study of aggressive resection of metastatic pancreatic endocrine tumors. Surgery 1992;112:1024-1031
  746. Bartsch DK, Waldmann J, Fendrich V , et al. Impact of lymphadenectomy on survival after surgery for sporadic gastrinoma. Br J Surg 2012;99:1234-1240
  747. Albers MB, Manoharan J, Bartsch DK. Contemporary surgical management of the Zollinger-Ellison syndrome in multiple endocrine neoplasia type 1. Best Pract Res Clin Endocrinol Metab 2019;33:101318
  748. Lopez CL, Falconi M, Waldmann J , et al. Partial pancreaticoduodenectomy can provide cure for duodenal gastrinoma associated with multiple endocrine neoplasia type 1. Ann Surg 2013;257:308-314
  749. Lopez CL, Waldmann J, Fendrich V , et al. Long-term results of surgery for pancreatic neuroendocrine neoplasms in patients with MEN1. Langenbecks Arch Surg 2011;396:1187-1197
  750. Falconi M, Bartsch DK, Eriksson B , et al. ENETS Consensus Guidelines for the Management of Patients with Digestive Neuroendocrine Neoplasms of the Digestive System: Well-Differentiated Pancreatic Non-Functioning Tumors. Neuroendocrinology 2012;95:120-134
  751. Triponez F, Goudet P, Dosseh D , et al. Is surgery beneficial for MEN1 patients with small (< or = 2 cm), nonfunctioning pancreaticoduodenal endocrine tumor? An analysis of 65 patients from the GTE. World J Surg 2006;30:654-662
  752. Goudet P, Dalac A, Le BA , et al. MEN1 disease occurring before 21 years old. A 160-patient cohort study from the GTE (Groupe d'etude des Tumeurs Endocrines). J Clin Endocrinol Metab 2015;100:1568-1577
  753. van Wijk JP, Dreijerink KM, Pieterman CR , et al. Increased prevalence of impaired fasting glucose in MEN1 gene mutation carriers. Clin Endocrinol (Oxf) 2012;76:67-71
  754. McCallum RW, Parameswaran V, Burgess JR. Multiple endocrine neoplasia type 1 (MEN 1) is associated with an increased prevalence of diabetes mellitus and impaired fasting glucose. Clin Endocrinol (Oxf) 2006;65:163-168
  755. Kouvaraki MA, Shapiro SE, Cote GJ , et al. Management of pancreatic endocrine tumors in multiple endocrine neoplasia type 1. World J Surg 2006;30:643-653
  756. Sakurai A, Katai M, Yamashita K , et al. Long-term follow-up of patients with multiple endocrine neoplasia type 1. Endocr J 2007;54:295-302
  757. Timofte D, Livadariu R, Bintintan V , et al. Metabolic disorders in patients operated for pancreatic cancer. Rev Med Chir Soc Med Nat Iasi 2014;118:392-398
  758. van Beek DJ, Nell S, Vorselaars WMCM , et al. Complications After Major Surgery for Duodenopancreatic Neuroendocrine Tumors in Patients with MEN1: Results from a Nationwide Cohort. Ann Surg Oncol 2021;28:4387-4399
  759. Barbe C, Murat A, Dupas B , et al. Magnetic resonance imaging versus endoscopic ultrasonography for the detection of pancreatic tumours in multiple endocrine neoplasia type 1. Dig Liver Dis 2012;44:228-234
  760. Yates CJ, Newey PJ, Thakker RV. Challenges and controversies in management of pancreatic neuroendocrine tumours in patients with MEN1. Lancet Diabetes Endocrinol 2015;3:895-905
  761. Froeling V, Elgeti F, Maurer MH , et al. Impact of Ga-68 DOTATOC PET/CT on the diagnosis and treatment of patients with multiple endocrine neoplasia. Ann Nucl Med 2012;26:738-743
  762. Lastoria S, Marciello F, Faggiano A , et al. Role of Ga-DOTATATE PET/CT in patients with multiple endocrine neoplasia type 1 (MEN1). Endocrine 2016;52:488-494
  763. Sharma P, Mukherjee A, Karunanithi S , et al. Accuracy of 68Ga DOTANOC PET/CT Imaging in Patients With Multiple Endocrine Neoplasia Syndromes. Clin Nucl Med 2015;40:e351-e356
  764. Kumar Gupta S, Singla S, Damle NA , et al. Diagnosis of Men-I Syndrome on (68)Ga-DOTANOC PET-CT and Role of Peptide Receptor Radionuclide Therapy With (177)Lu-DOTATATE. Int J Endocrinol Metab 2012;10:629-633
  765. Kottemann MC, Bale AE. Characterization of DNA damage-dependent cell cycle checkpoints in a menin-deficient model. DNA Repair (Amst) 2009;8:944-952
  766. Busygina V, Kottemann MC, Scott KL , et al. Multiple endocrine neoplasia type 1 interacts with forkhead transcription factor CHES1 in DNA damage response. Cancer Res 2006;66:8397-8403
  767. Busygina V, Suphapeetiporn K, Marek LR , et al. Hypermutability in a Drosophila model for multiple endocrine neoplasia type 1. Hum Mol Genet 2004;13:2399-2408
  768. Miglioretti DL, Johnson E, Williams A , et al. The use of computed tomography in pediatrics and the associated radiation exposure and estimated cancer risk. JAMA Pediatr 2013;167:700-707
  769. Hall EJ, Brenner DJ. Cancer risks from diagnostic radiology. Br J Radiol 2008;81:362-378
  770. Pearce MS, Salotti JA, Little MP , et al. Radiation exposure from CT scans in childhood and subsequent risk of leukaemia and brain tumours: a retrospective cohort study. Lancet 2012;380:499-505
  771. Stadil F. Treatment of gastrinomas with pancreaticoduodenectomy. In: Mignon M, Jensen RT eds. Endocrine Tumors of the Pancreas: Recent advances in research and management. Series: Frontiers in Gastrointestinal Research. Basel, Switzerland: S. Karger; 1995:333-341.
  772. Imamura M, Komoto I, Ota S , et al. Biochemically curative surgery for gastrinoma in multiple endocrine neoplasia type 1 patients. World J Gastroenterol 2011;17:1343-1353
  773. Imamura M, Takahashi K, Isobe Y , et al. Curative resection of multiple gastrinomas aided by selective arterial secretin injection and intraoperative secretin test. Ann Surg 1989;210:710-715
  774. Bartsch DK, Fendrich V, Langer P , et al. Outcome of duodenopancreatic resections in patients with multiple endocrine neoplasia type 1. Ann Surg 2005;242:757-64, discussion
  775. Waddell WR, Coppinger WR, Loughry RW. Pancreaticoduodenectomy for Zollinger-Ellison syndrome. Ann Surg 1968;168(4):641-654
  776. Oberhelman HA, Jr. Excisional therapy for ulcerogenic tumors of the duodenum: long- term results. Arch Surg 1972;104:447-453
  777. Roche A, Raisonnier A, Gillon-Savouret MC. Pancreatic venous sampling and arteriography in localizing insulinomas and gastrinomas: procedure and results in 55 cases. Radiology 1982;145:621-627
  778. Cavina E, Pagliai EF, Goletti O , et al. Pylorus preserving pancreatoduodenectomy for gastrinoma. A case report. Ital J Surg Sci 1986;16:291-295
  779. Stamm B, Hedinger CE, Saremaslani P. Duodenal and ampullary carcinoid tumors. A report of 12 cases with pathological characteristics, polypeptide content and relation to MEN 1 syndrome and von Recklinghausen's disease (neurofibromatosis). Virchows Arch A Pathol Anat Histopathol 1986;408:475-489
  780. Vogel SB, Wolfe MM, McGuigan JE , et al. Localization and resection of gastrinomas in Zollinger-Ellison syndrome. Ann Surg 1987;205:550-556
  781. Lind T, Olbe L. Long term follow up of patients with Zollinger-Ellison syndrome (ZES). Acta Chir Scand 1989;155:383-388
  782. Rosato FE, Bonn F, Shapiro M , et al. Selective arterial stimulation of secretin in localization of gastrinomas. Surg Gynecol Obstet 1990;171:196-200
  783. Delcore R, Friesen SR. Role of pancreatoduodenectomy in the management of primary duodenal wall gastrinomas in patients with Zollinger-Ellison syndrome. Surgery 1992;112:1016-1022
  784. Farley DR, Van Heerden JA, Grant CS , et al. The Zollinger-Ellison syndrome. A collective surgical experience. Ann Surg 1992;215:561-569
  785. Udelsman R, Yeo CJ, Hruban RH , et al. Pancreaticoduodenectomy for selected pancreatic endocrine tumors. Surg Gynecol Obstet 1993;177:269-278
  786. Stadil F, Bradram L, Gustafsen J , et al. Surgical treatment of the Zollinger-Ellison syndrome. World J Surg 1993;17:463-467
  787. Schroder W, Holscher AH, Beckurts KTE , et al. Chirurgische therapie des gastrinoms mit assoziiertem Zollinger-Ellison-Syndrom. Z Gastroenterol 1996;34:465-472
  788. Phan GQ, Yeo CJ, Cameron JL , et al. Pancreaticoduodenectomy for selected periampullary neuroendocrine tumors: Fifty patients. Surgery 1997;122:989-997
  789. Jordan PH, Jr. A personal experience with pancreatic and duodenal neuroendocrine tumors. J Am Coll Surg 1999;189:470-482
  790. Partensky C, Berger F, Owono P , et al. Cephalic duodenopancreatectomy for endocrine tumor of the ampulla of Vater and of the minor papilla. Gastroenterol Clin Biol 1999;23:832-836
  791. Tonelli F, Fratini G, Falchetti A , et al. Surgery for gastroenteropancreatic tumours in multiple endocrine neoplasia type 1: review and personal experience. J Intern Med 2005;257:38-49
  792. Bartsch DK, Langer P, Wild A , et al. Pancreaticoduodenal endocrine tumors in multiple endocrine neoplasia type 1: Surgery or surveillance? Surgery 2000;128:958-966
  793. Kato M, Imamura M, Hosotani R , et al. Curative resection of microgastrinomas based on the intraoperative secretin test. World J Surg 2000;24:1425-1430
  794. Lairmore TC, Chen VY, DeBenedetti MK , et al. Duodenopancreatic resections in patients with multiple endocrine neoplasia type 1. Ann Surg 2000;231:909-918
  795. Siech M, Mattfeldt T, Schlosser W , et al. Duodenum-preserving pancreatic head resection in patients with benign and borderline tumors of the pancreatic head. Langenbeck's Arch Surg 2000;385:229-233
  796. Thodiyil PA, El-Masry NS, Williamson RC. Achieving eugastrinaemia in Zollinger-Ellison syndrome: resection or enucleation? Dig Surg 2001;18:118-123
  797. Huang JJ, Yeo CJ, Sohn TA , et al. Quality of life and outcomes after pancreaticoduodenectomy. Ann Surg 2000;231:890-898
  798. Tartaglia A, Portela-Gomes GM, Oberg K , et al. Chromogranin A in gastric neuroendocrine tumours: an immunohistochemical and biochemical study with region-specific antibodies. Virchows Arch 2006;448:399-406
  799. Price TN, Thompson GB, Lewis JT , et al. Zollinger-Ellison syndrome due to primary gastrinoma of the extrahepatic biliary tree: three case reports and review of literature. Endocr Pract 2009;15:737-749
  800. Giovinazzo F, Butturini G, Monsellato D , et al. Lymph nodes metastasis and recurrences justify an aggressive treatment of gastrinoma. Updates Surg 2013;65:19-24
  801. Fernandez-Cruz L, Pelegrina A. Surgery for gastrinoma: Short and long-term results. Cir Esp 2015;93:390-395
  802. Maire F, Sauvanet A, Couvelard A , et al. Recurrence after surgical resection of gastrinoma: who, when, where and why? Eur J Gastroenterol Hepatol 2012;24:368-374
  803. Grobmyer SR, Vogel SB, McGuigan JE , et al. Reoperative surgery in sporadic Zollinger-Ellison Syndrome: longterm results. J Am Coll Surg 2010;208:718-722
  804. Nishio K, Nishio A, Nishikawa T , et al. Recurrent gastrinoma in the mesentery 19 years after primary resection. Dig Dis Sci 2007;52:3104-3108
  805. Jaskowiak NT, Fraker DL, Alexander HR , et al. Is reoperation for gastrinoma excision in Zollinger-Ellison syndrome (ZES) indicated? Surgery 1996;120:1057-1063
  806. Krampitz GW, Norton JA, Poultsides GA , et al. Lymph nodes and survival in duodenal and pancreatic neuroendocrine tumors. Arch Surg 2012;147:820-827
  807. Tomassetti P, Campana D, Piscitelli L , et al. Endocrine pancreatic tumors: factors correlated with survival. Ann Oncol 2005;16:1806-1810
  808. Bettini R, Boninsegna L, Mantovani W , et al. Prognostic factors at diagnosis and value of WHO classification in a mono-institutional series of 180 non-functioning pancreatic endocrine tumours. Ann Oncol 2008;19:903-908
  809. Boninsegna L, Panzuto F, Partelli S , et al. Malignant pancreatic neuroendocrine tumour: Lymph node ratio and Ki67 are predictors of recurrence after curative resections. Eur J Cancer 2012;48:1608-1615
  810. Roland CL, Bian A, Mansour JC , et al. Survival impact of malignant pancreatic neuroendocrine and islet cell neoplasm phenotypes. J Surg Oncol 2011;
  811. Chu QD, Hill HC, Douglass HO, Jr. , et al. Predictive factors associated with long-term survival in patients with neuroendocrine tumors of the pancreas. Ann Surg Oncol 2002;9:855-862
  812. Bilimoria KY, Talamonti MS, Tomlinson JS , et al. Prognostic score predicting survival after resection of pancreatic neuroendocrine tumors: analysis of 3851 patients. Ann Surg 2008;247:490-500
  813. DiNorcia J, Lee MK, Reavey PL , et al. One hundred thirty resections for pancreatic neuroendocrine tumor: evaluating the impact of minimally invasive and parenchyma-sparing techniques. J Gastrointest Surg 2010;14:1536-1546
  814. Capelli P, Fassan M, Scarpa A. Pathology - Grading and staging of GEP-NETs. Best Pract Res Clin Gastroenterol 2012;26:705-717
  815. Arnold R, Rinke A, Klose KJ , et al. Octreotide versus octreotide plus interferon-alpha in endocrine gastroenteropancreatic tumors: a randomized trial. Clin Gastroenterol Hepatol 2005;3:761-771
  816. Strosberg J, Gardner N, Kvols L. Survival and prognostic factor analysis in patients with metastatic pancreatic endocrine carcinomas. Pancreas 2009;38:255-258
  817. Strosberg JR, Cheema A, Weber J , et al. Prognostic validity of a novel American Joint Committee on Cancer Staging Classification for pancreatic neuroendocrine tumors. J Clin Oncol 2011;29:3044-3049
  818. Zollinger RM. The Zollinger-Ellison syndrome. World J Surg 1981;5:773-775
  819. Berger AC, Gibril F, Venzon DJ , et al. Prognostic value of initial fasting serum gastrin level in patients with Zollinger-Ellison syndrome. J Clin Oncol 2001;19:3051-3057
  820. Metz DC, Kuchnio M, Fraker DL , et al. Flow cytometry and Zollinger-Ellison syndrome: relationship to clinical course. Gastroenterology 1993;105:799-813
  821. Chen Y-J, Zortmeyer A, Zhuang ZP , et al. Chromosome X loss of heterozygosity frequently occurs in gastrinomas and is correlated with aggressive tumor growth. Cancer 2004;100:1379-1387
  822. Chen YJ, Vortmeyer A, Zhuang Z , et al. Loss of heterozygosity of chromosome 1q in gastrinomas: occurrence and prognostic significance. Cancer Res 2003;63:817-823
  823. Pape UF, Jann H, Muller-Nordhorn J , et al. Prognostic relevance of a novel TNM classification system for upper gastroenteropancreatic neuroendocrine tumors. Cancer 2008;113:256-265
  824. Lepage C, Rachet B, Coleman MP. Survival from malignant digestive endocrine tumors in England and Wales: a population-based study. Gastroenterology 2007;132:899-904
  825. Yang Z, Shi G. Comparative outcomes of pancreatic neuroendocrine neoplasms: A population-based analysis of the SEER database. Eur J Surg Oncol 2022;48:2181-2187
  826. Schmitt AM, Anlauf M, Rousson V , et al. WHO 2004 criteria and CK19 are reliable prognostic markers in pancreatic endocrine tumors. Am J Surg Pathol 2007;31:1677-1682
  827. Maire F, Hammel P, Faivre S , et al. Temozolomide: a safe and effective treatment for malignant digestive endocrine tumors. Neuroendocrinology 2009;90:67-72
  828. La Rosa S, Rigoli E, Uccella S , et al. Prognostic and biological significance of cytokeratin 19 in pancreatic endocrine tumours. Histopathology 2007;50:597-606
  829. La Rosa S, Sessa F, Capella C , et al. Prognostic criteria in nonfunctioning pancreatic endocrine tumours. Virchows Arch 1996;429:323-333
  830. Hauser H, Gerson DS, Reidy-Lagunes D , et al. Systemic Therapies for Metastatic Pancreatic Neuroendocrine Tumors. Curr Treat Options Oncol 2019;20:87
  831. Dermine S, Palmieri LJ, Lavole J , et al. Non-Pharmacological Therapeutic Options for Liver Metastases in Advanced Neuroendocrine Tumors. J Clin Med 2019;8:1907
  832. Scoville SD, Cloyd JM, Pawlik TM. New and emerging systemic therapy options for well-differentiated gastroenteropancreatic neuroendocrine tumors. Expert Opin Pharmacother 2020;21:183-191
  833. Mohamed A, Strosberg JR. Medical Management of Gastroenteropancreatic Neuroendocrine Tumors: Current Strategies and Future Advances. J Nucl Med 2019;60:721-727
  834. Kanabar R, Barriuso J, McNamara MG , et al. Liver embolisation for patients with neuroendocrine neoplasms: systematic review. Neuroendocrinology 2021;111:354-369
  835. Li YL, Cheng ZX, Yu FH , et al. Advances in medical treatment for pancreatic neuroendocrine neoplasms. World J Gastroenterol 2022;28:2163-2175
  836. Ngongoni R, Visser B. Surgery, Liver Directed Therapy and Peptide Receptor Radionuclide Therapy for Pancreatic Neuroendocrine Tumor Liver Metastases. Cancers (Basel) 2022;14:
  837. Chauhan A, De Rivero J, Ramirez RA , et al. Treatment Sequencing Strategies in Advanced Neuroendocrine Tumors: A Review. Cancers (Basel) 2022;14:5049
  838. Modica R, Liccardi A, Minotta R , et al. Therapeutic strategies for patients with neuroendocrine neoplasms: current perspectives. Expert Rev Endocrinol Metab 2022;17:389-403
  839. Cuthbertson DJ, Shankland R, Srirajaskanthan R. Diagnosis and management of neuroendocrine tumours. Clin Med (Lond) 2023;23:119-124
  840. Shi C, Morse MA. Mechanisms of Resistance in Gastroenteropancreatic Neuroendocrine Tumors. Cancers (Basel) 2022;14:6114
  841. McClellan K, Chen EY, Kardosh A , et al. Therapy Resistant Gastroenteropancreatic Neuroendocrine Tumors. Cancers (Basel) 2022;14:
  842. Norton JA, Sugarbaker PH, Doppman JL , et al. Aggressive resection of metastatic disease in selected patients with malignant gastrinoma. Ann Surg 1986;203:352-359
  843. Norton JA, Warren RS, Kelly MG , et al. Aggressive surgery for metastatic liver neuroendocrine tumors. Surgery 2003;134:1057-1065
  844. Norton JA, Kirlen MA, Li M , et al. Morbidity and mortality of aggressive resections in patients with advanced neuroendocrine tumors. Arch Surg 2003;138:859-866
  845. Que FG, Sarmiento JM, Nagorney DM. Hepatic surgery for metastatic gastrointestinal neuroendocrine tumors. Adv Exp Med Biol 2006;574:43-56
  846. Sarmiento JM, Que FG. Hepatic surgery for metastases from neuroendocrine tumors. Surg Oncol Clin North Am 2003;12:231-242
  847. Wang SC, Fidelman N, Nakakura EK. Management of well-differentiated gastrointestinal neuroendocrine tumors metastatic to the liver. Semin Oncol 2013;40:69-74
  848. Pavel M, Baudin E, Couvelard A , et al. ENETS Consensus Guidelines for the Management of Patients with Liver and Other Distant Metastases from Neuroendocrine Neoplasms of Foregut, Midgut, Hindgut, and Unknown Primary. Neuroendocrinology 2012;95:157-176
  849. Howe JR, Cardona K, Fraker DL , et al. The Surgical Management of Small Bowel Neuroendocrine Tumors: Consensus Guidelines of the North American Neuroendocrine Tumor Society. Pancreas 2017;46:715-731
  850. Tamburrino D, Spoletini G, Partelli S , et al. Surgical management of neuroendocrine tumors. Best Pract Res Clin Endocrinol Metab 2016;30:93-102
  851. Uri I, Grozinsky-Glasberg S. Current treatment strategies for patients with advanced gastroenteropancreatic neuroendocrine tumors (GEP-NETs). Clin Diabetes Endocrinol 2018;4:16
  852. Chan MY, Ma KW, Chan A. Surgical management of neuroendocrine tumor-associated liver metastases: a review. Gland Surg 2018;7:28-35
  853. Ejaz A, Reames BN, Maithel S , et al. Cytoreductive debulking surgery among patients with neuroendocrine liver metastasis: a multi-institutional analysis. HPB (Oxford) 2018;20:277-284
  854. Lesurtel M, Nagorney DM, Mazzaferro V , et al. When should a liver resection be performed in patients with liver metastases from neuroendocrine tumours? A systematic review with practice recommendations. HPB (Oxford) 2015;17:17-22
  855. Addeo P, Bertin JB, Imperiale A , et al. Outcomes of Simultaneous Resection of Small Bowel Neuroendocrine Tumors with Synchronous Liver Metastases. World J Surg 2020;44:2377-2384
  856. Jeune F, Taibi A, Gaujoux S. Update on the Surgical Treatment of Pancreatic Neuroendocrine Tumors. Scand J Surg 2020;109:42-52
  857. Merola E, Rinke A, Partelli S , et al. Surgery with Radical Intent: Is There an Indication for G3 Neuroendocrine Neoplasms? Ann Surg Oncol 2020;27:1348-1355
  858. Pozzari M, Maisonneuve P, Spada F , et al. Systemic therapies in patients with advanced well-differentiated pancreatic neuroendocrine tumors (PanNETs): When cytoreduction is the aim. A critical review with meta-analysis. Cancer Treat Rev 2018;71:39-46
  859. Kacmaz E, Heidsma CM, Besselink MGH , et al. Treatment of Liver Metastases from Midgut Neuroendocrine Tumours: A Systematic Review and Meta-Analysis. J Clin Med 2019;8:403
  860. Veltroni A, Cosaro E, Spada F , et al. Clinico-pathological features, treatments and survival of malignant insulinomas: a multicenter study. Eur J Endocrinol 2020;182:439-446
  861. Sham JG, Ejaz A, Gage MM , et al. The Impact of Extent of Liver Resection Among Patients with Neuroendocrine Liver Metastasis: an International Multi-institutional Study. J Gastrointest Surg 2019;23:484-491
  862. Cloyd JM, Wiseman JT, Pawlik TM. Surgical management of pancreatic neuroendocrine liver metastases. J Gastrointest Oncol 2020;11:590-600
  863. Jensen RT, Bodei L, Capdevila J , et al. Unmet Needs in Functional and Nonfunctional Pancreatic Neuroendocrine Neoplasms. Neuroendocrinology 2019;108:26-36
  864. Tran CG, Sherman SK, Chandrasekharan C , et al. Surgical Management of Neuroendocrine Tumor Liver Metastases. Surg Oncol Clin N Am 2021;30:39-55
  865. Cherner JA, Sawyers JL. Benefit of resection of metastatic gastrinoma in multiple endocrine neoplasia type I. Gastroenterology 1992;102:1049-1053
  866. Masui T, Nagai K, Anazawa T , et al. Risk factors for short recurrence-free survival after resection of pancreatic neuroendocrine tumor (PanNET) liver metastases: which patients should undergo resection? Scand J Gastroenterol 2020;55:479-484
  867. Merola E, Falconi M, Rinke A , et al. Radical intended surgery for highly selected stage IV neuroendocrine neoplasms G3. Am J Surg 2020;220:284-289
  868. Brighi N, Lamberti G, Maggio I , et al. Biliary stone disease in patients receiving somatostatin analogs for neuroendocrine neoplasms. A retrospective observational study. Dig Liver Dis 2019;51:689-694
  869. Bertani E, Fazio N, Radice D , et al. Resection of the Primary Tumor Followed by Peptide Receptor Radionuclide Therapy as Upfront Strategy for the Treatment of G1-G2 Pancreatic Neuroendocrine Tumors with Unresectable Liver Metastases. Ann Surg Oncol 2016;23:981-989
  870. Nazario J, Gupta S. Transarterial liver-directed therapies of neuroendocrine hepatic metastases. Semin Oncol 2010;37:118-126
  871. Vyleta M, Coldwell D. Radioembolization in the treatment of neuroendocrine tumor metastases to the liver. Int J Hepatol 2011;2011(Article ID 785315):1-5
  872. Barat M, Cottereau AS, Kedra A , et al. The Role of Interventional Radiology for the Treatment of Hepatic Metastases from Neuroendocrine Tumor: An Updated Review. J Clin Med 2020;9:2302
  873. Kessler J, Singh G, Ituarte PHG , et al. A Comparison of Liver-Directed Therapy and Systemic Therapy for the Treatment of Liver Metastases in Patients with Gastrointestinal Neuroendocrine Tumors: Analysis of the California Cancer Registry. J Vasc Interv Radiol 2021;32:393-402
  874. Nanno Y, Toyama H, Ueshima E , et al. Transarterial chemoembolization for liver metastases of a pancreatic neuroendocrine neoplasm: a single-center experience. Surg Today 2023 (In Press)
  875. Cazzato RL, Hubele F, De Marini P , et al. Liver-Directed Therapy for Neuroendocrine Metastases: From Interventional Radiology to Nuclear Medicine Procedures. Cancers (Basel) 2021;13:6368
  876. Egger ME, Armstrong E, Martin RC , et al. Transarterial Chemoembolization vs Radioembolization for Neuroendocrine Liver Metastases: A Multi-Institutional Analysis. J Am Coll Surg 2020;230:363-370
  877. Ramdhani K, Braat AJAT. The Evolving Role of Radioembolization in the Treatment of Neuroendocrine Liver Metastases. Cancers (Basel) 2022;14:3415
  878. The NCCN Clinical Practice Guidelines in Oncology for Neuroendocrine tumors version 1.2012. ( online. go to www.nccn.org.: 2012.
  879. Klein A, Clemens J, Cameron J. Periampullary neoplasms in von Recklinghausen's disease. Surgery 1989;106:815-819
  880. O'Toole D, Ruszniewski P. Chemoembolization and other ablative therapies for liver metastases of gastrointestinal endocrine tumours. Best Pract Res Clin Gastroenterol 2005;19:585-594
  881. Elias D, Goere D, Leroux G , et al. Combined liver surgery and RFA for patients with gastroenteropancreatic endocrine tumors presenting with more than 15 metastases to the liver. Eur J Surg Oncol 2009;35:1092-1097
  882. Harring TR, Nguyen NT, Goss JA , et al. Treatment of liver metastases in patients with neuroendocrine tumors: a comprehensive review. Int J Hepatol 2011;2011:1-11
  883. Figueiredo Ferreira M, Garces-Duran R, Eisendrath P , et al. EUS-guided radiofrequency ablation of pancreatic/peripancreatic tumors and oligometastatic disease: an observational prospective multicenter study. Endosc Int Open 2022;10:E1380-E1385
  884. Guillermet-Guibert J, Lahlou H, Pyronnet S , et al. Somatostatin receptors as tools for diagnosis and therapy: Molecular aspects. Best Pract Res Clin Gastroenterol 2005;19:535-551
  885. Karabulut K, Akyildiz HY, Lance C , et al. Multimodality treatment of neuroendocrine liver metastases. Surgery 2011;150:316-325
  886. Akyildiz HY, Mitchell J, Milas M , et al. Laparoscopic radiofrequency thermal ablation of neuroendocrine hepatic metastases: Long-term follow-up. Surgery 2010;148:1288-1293
  887. Eriksson J, Stalberg P, Nilsson A , et al. Surgery and radiofrequency ablation for treatment of liver metastases from midgut and foregut carcinoids and endocrine pancreatic tumors. World J Surg 2008;32:930-938
  888. Vogl TJ, Naguib NN, Zangos S , et al. Liver metastases of neuroendocrine carcinomas: interventional treatment via transarterial embolization, chemoembolization and thermal ablation. Eur J Radiol 2009;72:517-528
  889. Mazzaglia PJ, Berber E, Milas M , et al. Laparoscopic radiofrequency ablation of neuroendocrine liver metastases: a 10-year experience evaluating predictors of survival. Surgery 2007;142:10-19
  890. Kvols LK, Turaga KK, Strosberg J , et al. Role of interventional radiology in the treatment of patients with neuroendocrine metastases in the liver. J Natl Compr Canc Netw 2009;7:765-772
  891. de Mestier L, Zappa M, Hentic O , et al. Liver transarterial embolizations in metastatic neuroendocrine tumors. Rev Endocr Metab Disord 2017;18:459-471
  892. Maire F, Lombard-Bohas C, O'Toole D , et al. Hepatic arterial embolization versus chemoembolization in the treatment of liver metastases from well-differentiated midgut endocrine tumors: a prospective randomized study. Neuroendocrinology 2012;96:294-300
  893. Toumpanakis C, Meyer T, Caplin ME. Cytotoxic treatment including embolization/chemoembolization for neuroendocrine tumours. Best Pract Res Clin Endocrinol Metab 2007;21:131-144
  894. Lewis MA, Jaramillo S, Roberts L , et al. Hepatic Artery Embolization for Neuroendocrine Tumors: Postprocedural Management and Complications. Oncologist 2012;17:725-731
  895. Memon K, Lewandowski RJ, Mulcahy MF , et al. Radioembolization for Neuroendocrine Liver Metastases: Safety, Imaging, and Long-term Outcomes. Int J Radiat Oncol Biol Phys 2012;83:887-894
  896. Kennedy A, Coldwell D, Sangro B , et al. Integrating Radioembolization into the Treatment Paradigm for Metastatic Neuroendocrine Tumors in the Liver. Am J Clin Oncol 2012;35:293-301
  897. Paprottka PM, Hoffmann RT, Haug A , et al. Radioembolization of symptomatic, unresectable neuroendocrine hepatic metastases using yttrium-90 microspheres. Cardiovasc Intervent Radiol 2012;35:334-342
  898. Lacin S, Oz I, Ozkan E , et al. Intra-arterial treatment with 90yttrium microspheres in treatment-refractory and unresectable liver metastases of neuroendocrine tumors and the use of 111in-octreotide scintigraphy in the evaluation of treatment response. Cancer Biother Radiopharm 2011;26:631-637
  899. Shaheen M, Hassanain M, Aljiffry M , et al. Predictors of response to radio-embolization (TheraSphere(R)) treatment of neuroendocrine liver metastasis. HPB (Oxford) 2012;14:60-66
  900. King J, Quinn R, Glenn DM , et al. Radioembolization with selective internal radiation microspheres for neuroendocrine liver metastases. Cancer 2008;113:921-929
  901. Whitney R, Valek V, Fages JF , et al. Transarterial chemoembolization and selective internal radiation for the treatment of patients with metastatic neuroendocrine tumors: a comparison of efficacy and cost. Oncologist 2011;16:594-601
  902. Kennedy AS, Dezarn WA, McNeillie P , et al. Radioembolization for unresectable neuroendocrine hepatic metastases using resin 90Y-microspheres: early results in 148 patients. Am J Clin Oncol 2008;31:271-279
  903. Saxena A, Chua TC, Bester L , et al. Factors predicting response and survival after yttrium-90 radioembolization of unresectable neuroendocrine tumor liver metastases: a critical appraisal of 48 cases. Ann Surg 2010;251:910-916
  904. Ramage JK, Ahmed A, Ardill J , et al. Guidelines for the management of gastroenteropancreatic neuroendocrine (including carcinoid) tumours (NETs). Gut 2012;61:6-32
  905. Deleporte A, Flamen P, Hendlisz A. State of the art: radiolabeled microspheres treatment for liver malignancies. Expert Opin Pharmacother 2010;11:579-586
  906. Lee E, Leon Pachter H, Sarpel U. Hepatic arterial embolization for the treatment of metastatic neuroendocrine tumors. Int J Hepatol 2012;2012(Article ID 471203):1-8
  907. Barbier CE, Garske-Roman U, Sandstrom M , et al. Selective internal radiation therapy in patients with progressive neuroendocrine liver metastases. Eur J Nucl Med Mol Imaging 2016;43:1425-1431
  908. Braat AJAT, Kappadath SC, Ahmadzadehfar H , et al. Radioembolization with (90)Y Resin Microspheres of Neuroendocrine Liver Metastases: International Multicenter Study on Efficacy and Toxicity. Cardiovasc Intervent Radiol 2019;42:413-425
  909. Currie BM, Hoteit MA, Ben-Josef E , et al. Radioembolization-Induced Chronic Hepatotoxicity: A Single-Center Cohort Analysis. J Vasc Interv Radiol 2019;30:1915-1923
  910. Krug S, Gress TM, Michl P , et al. The Role of Cytotoxic Chemotherapy in Advanced Pancreatic Neuroendocrine Tumors. Digestion 2017;96:67-75
  911. de Mestier L, Walter T, Brixi H , et al. Comparison of Temozolomide-Capecitabine to 5-Fluorouracile-Dacarbazine in 247 Patients with Advanced Digestive Neuroendocrine Tumors Using Propensity Score Analyses. Neuroendocrinology 2019;108:343-353
  912. Cives M, Pelle' E, Quaresmini D , et al. The Role of Cytotoxic Chemotherapy in Well-Differentiated Gastroenteropancreatic and Lung Neuroendocrine Tumors. Curr Treat Options Oncol 2019;20:72
  913. Chan DL, Singh S. Current Chemotherapy Use in Neuroendocrine Tumors. Endocrinol Metab Clin North Am 2018;47:603-614
  914. Palmieri LJ, Dermine S, Barre A , et al. Medical Treatment of Advanced Pancreatic Neuroendocrine Neoplasms. J Clin Med 2020;9:1860
  915. Riccardi F, Rizzo M, Festino L , et al. Therapy innovation for the treatment of pancreatic neuroendocrine tumors. Expert Opin Ther Targets 2012;16 Suppl 2:S91-102
  916. Kouvaraki MA, Ajani JA, Hoff P , et al. Fluorouracil, doxorubicin, and streptozocin in the treatment of patients with locally advanced and metastatic pancreatic endocrine carcinomas. J Clin Oncol 2004;22:4762-4771
  917. Delaunoit T, Ducreux M, Boige V , et al. The doxorubicin-streptozotocin combination for the treatment of advanced well-differentiated pancreatic endocrine carcinoma; a judicious option? Eur J Cancer 2004;40:515-520
  918. Kulke MH. Systemic therapy for advanced pancreatic neuroendocrine tumors. Semin Oncol 2013;40:75-83
  919. Costa FP, Gumz B, Pasche B. Selecting patients for cytotoxic therapies in gastroenteropancreatic neuroendocrine tumours. Best Pract Res Clin Gastroenterol 2012;26:843-854
  920. Arrivi G, Verrico M, Roberto M , et al. Capecitabine and Temozolomide (CAPTEM) in Advanced Neuroendocrine Neoplasms (NENs): A Systematic Review and Pooled Analysis. Cancer Manag Res 2022;14:3507-3523
  921. O'Toole D, Hentic O, Corcos O , et al. Chemotherapy for gastro-enteropancreatic endocrine tumours. Neuroendocrinology 2004;80 Suppl 1:79-84
  922. Lu Y, Zhao Z, Wang J , et al. Safety and efficacy of combining capecitabine and temozolomide (CAPTEM) to treat advanced neuroendocrine neoplasms: A meta-analysis. Medicine (Baltimore) 2018;97:e12784
  923. Thomas K, Voros BA, Meadows-Taylor M , et al. Outcomes of Capecitabine and Temozolomide (CAPTEM) in Advanced Neuroendocrine Neoplasms (NENs). Cancers (Basel) 2020;12:206
  924. Strosberg JR, Fine RL, Choi J , et al. First-line chemotherapy with capecitabine and temozolomide in patients with metastatic pancreatic endocrine carcinomas. Cancer 2011;117:268-275
  925. Ekeblad S, Sundin A, Janson ET , et al. Temozolomide as monotherapy is effective in treatment of advanced malignant neuroendocrine tumors. Clin Cancer Res 2007;13:2986-2991
  926. Kulke MH, Hornick JL, Frauenhoffer C , et al. O6-methylguanine DNA methyltransferase deficiency and response to temozolomide-based therapy in patients with neuroendocrine tumors. Clin Cancer Res 2009;15:338-345
  927. Spada F, Maisonneuve P, Fumagalli C , et al. Temozolomide alone or in combination with capecitabine in patients with advanced neuroendocrine neoplasms: an Italian multicenter real-world analysis. Endocrine 2021;72:268-278
  928. Kunz PL, Graham NT, Catalano PJ , et al. Randomized Study of Temozolomide or Temozolomide and Capecitabine in Patients With Advanced Pancreatic Neuroendocrine Tumors (ECOG-ACRIN E2211). J Clin Oncol 2023;41:1359-1369
  929. Ruszniewski P, Hochlaf S, Rougier P , et al. (Intravenous chemotherapy with streptozotocin and 5 fluorouracil for hepatic metastases of Zollinger-Ellison syndrome. A prospective multicenter study in 21 patients). Gastroenterol Clin Biol 1991;15:393-398
  930. Steinmuller T, Kianmanesh R, Falconi M , et al. Consensus guidelines for the management of patients with liver metastases from digestive (neuro)endocrine tumors: foregut, midgut, hindgut, and unknown primary. Neuroendocrinology 2008;87:47-62
  931. Nilsson O, Van Cutsem E, Delle Fave G , et al. Poorly differentiated carcinomas of the foregut (gastric, duodenal and pancreatic). Neuroendocrinology 2006;84:212-215
  932. Smith J, Reidy-Lagunes D. The management of extrapulmonary poorly differentiated (high-grade) neuroendocrine carcinomas. Semin Oncol 2013;40:100-108
  933. Chan DL, Bergsland EK, Chan JA , et al. Temozolomide in grade III gastroenteropancreatic neuroendocrine neoplasms (G3 GEPNENs): A multicentre retrospective review. Oncologist 2021;26:950-955
  934. Garcia-Alvarez A, Cubero JH, Capdevila J. What Is the Status of Immunotherapy in Neuroendocrine Neoplasms? Curr Oncol Rep 2022;24:451-461
  935. Alheraki SZ, Almquist DR, Starr JS , et al. Treatment landscape of advanced high-grade neuroendocrine neoplasms. Clin Adv Hematol Oncol 2023;21:16-26
  936. Iwasa S, Morizane C, Okusaka T , et al. Cisplatin and etoposide as first-line chemotherapy for poorly differentiated neuroendocrine carcinoma of the hepatobiliary tract and pancreas. Jpn J Clin Oncol 2010;40:313-318
  937. Garcia-Carbonero R, Anton-Pascual B, Modrego A , et al. Advances in the Treatment of Gastroenteropancreatic Neuroendocrine Carcinomas: Are we Moving Forward? Endocr Rev 2023;44:724-736
  938. Strosberg JR, Coppola D, Klimstra DS , et al. The NANETS Consensus Guidelines for the Diagnosis and Management of Poorly Differentiated (High-Grade) Extrapulmonary Neuroendocrine Carcinomas. Pancreas 2010;39:799-800
  939. Olsen IH, Langer SW, Jepsen I , et al. First-line treatment of patients with disseminated poorly differentiated neuroendocrine carcinomas with carboplatin, etoposide, and vincristine: a single institution experience. Acta Oncol 2012;51:97-100
  940. Que QY, Zhang LC, Bao JQ , et al. Role of surgical treatments in high-grade or advanced gastroenteropancreatic neuroendocrine neoplasms. World J Gastrointest Surg 2022;14:397-408
  941. Walter T, van Brakel B, Vercherat C , et al. O6-Methylguanine-DNA methyltransferase status in neuroendocrine tumours: prognostic relevance and association with response to alkylating agents. Br J Cancer 2015;112:523-531
  942. Campana D, Walter T, Pusceddu S , et al. Correlation between MGMT promoter methylation and response to temozolomide-based therapy in neuroendocrine neoplasms: an observational retrospective multicenter study. Endocrine 2018;60:490-498
  943. Schmitt AM, Pavel M, Rudolph T , et al. Prognostic and predictive roles of MGMT protein expression and promoter methylation in sporadic pancreatic neuroendocrine neoplasms. Neuroendocrinology 2014;100:35-44
  944. Cros J, Hentic O, Rebours V , et al. MGMT expression predicts response to temozolomide in pancreatic neuroendocrine tumors. Endocr Relat Cancer 2016;23:625-633
  945. Lemelin A, Barritault M, Hervieu V , et al. O6-methylguanine-DNA methyltransferase (MGMT) status in neuroendocrine tumors: a randomized phase II study (MGMT-NET). Dig Liver Dis 2019;51:595-599
  946. Yagi K, Ono H, Kudo A , et al. MGMT is frequently inactivated in pancreatic NET-G2 and is associated with the therapeutic activity of STZ-based regimens. Sci Rep 2023;13:7535
  947. Brighi N, Lamberti G, Andrini E , et al. Prospective Evaluation of MGMT-Promoter Methylation Status and Correlations with Outcomes to Temozolomide-Based Chemotherapy in Well-Differentiated Neuroendocrine Tumors. Curr Oncol 2023;30:1381-1394
  948. Trillo Aliaga P, Spada F, Peveri G , et al. Should temozolomide be used on the basis of O(6)-methylguanine DNA methyltransferase status in patients with advanced neuroendocrine tumors? A systematic review and meta-analysis. Cancer Treat Rev 2021;99:102261
  949. Wang W, Zhang Y, Peng Y , et al. A Ki-67 Index to Predict Treatment Response to the Capecitabine Temozolomide (CAPTEM) Regimen in Neuroendocrine Neoplasms: A Retrospective Multicenter Study. Neuroendocrinology 2021;111:752-763
  950. Girot P, Dumars C, Mosnier JF , et al. Short article: Evaluation of O6-methylguanine-DNA methyltransferase as a predicting factor of response to temozolomide-based chemotherapy in well-differentiated metastatic pancreatic neuroendocrine tumors. Eur J Gastroenterol Hepatol 2017;29:826-830
  951. Raj N, Klimstra DS, Horvat N , et al. O6-Methylguanine DNA Methyltransferase Status Does Not Predict Response or Resistance to Alkylating Agents in Well-Differentiated Pancreatic Neuroendocrine Tumors. Pancreas 2017;46:758-763
  952. Zhang J, Kulkarni HR, Singh A , et al. Peptide Receptor Radionuclide Therapy in Grade 3 Neuroendocrine Neoplasms: Safety and Survival Analysis in 69 Patients. J Nucl Med 2019;60:377-385
  953. Sorbye H, Kong G, Grozinsky-Glasberg S. PRRT in high-grade gastroenteropancreatic neuroendocrine neoplasms (WHO G3). Endocr Relat Cancer 2020;27:R67-R77
  954. Stueven AK, Kayser A, Wetz C , et al. Somatostatin Analogues in the Treatment of Neuroendocrine Tumors: Past, Present and Future. Int J Mol Sci 2019;20:3049
  955. Narayanan S, Kunz PL. Role of Somatostatin Analogues in the Treatment of Neuroendocrine Tumors. Hematol Oncol Clin North Am 2016;30:163-177
  956. Caplin ME, Pavel M, Cwikla Jb , et al. Lanreotide in metastatic enteropancreatic neuroendocrine tumors. N Engl J Med 2014;371:224-233
  957. Capurso G, Fazio N, Festa S , et al. Molecular target therapy for gastroenteropancreatic endocrine tumours: biological rationale and clinical perspectives. Crit Rev Oncol Hematol 2009;72:110-124
  958. Appetecchia M, Baldelli R. Somatostatin analogues in the treatment of gastroenteropancreatic neuroendocrine tumours, current aspects and new perspectives. J Exp Clin Cancer Res 2010;29:19-31
  959. Strosberg J, Kvols L. Antiproliferative effect of somatostatin analogs in gastroenteropancreatic neuroendocrine tumors. World J Gastroenterol 2010;16:2963-2970
  960. Sideris L, Dube P, Rinke A. Antitumor Effects of Somatostatin Analogs in Neuroendocrine Tumors. Oncologist 2012;17:747-755
  961. Plockinger U, Wiedenmann B. Biotherapy. Best Pract Res Clin Endocrinol Metab 2007;21:145-162
  962. Toumpanakis C, Caplin ME. Update on the role of somatostatin analogs for the treatment of patients with gastroenteropancreatic neuroendocrine tumors. Semin Oncol 2013;40:56-68
  963. Modlin IM, Pavel M, Kidd M , et al. Review article: somatostatin analogues in the treatment of gastroenteropancreatic neuroendocrine (carcinoid) tumours. Aliment Pharmacol Ther 2010;31:169-188
  964. Panzuto F, Di Francesco V, Iannicelli E , et al. Long-term clinical outcome of somatostatin analogues for treatment of progressive, metastatic, well-differentiated entero-pancreatic endocrine carcinoma. Ann Oncol 2006;17:461-466
  965. Sharp AJ, Hayes AR, Grossman A. High-dose Somatostatin Analogues for Progressive Neuroendocrine Tumours. Eur Endocrinol 2020;16:93-95
  966. Rinke A, Muller HH, Schade-Brittinger C , et al. Placebo-controlled, double-blind, prospective, randomized study on the effect of octreotide LAR in the control of tumor growth in patients with metastatic neuroendocrine midgut tumors: a report from the PROMID Study Group. J Clin Oncol 2009;27:4656-4663
  967. Oberg K, Ferone D, Kaltsas G , et al. ENETS Consensus Guidelines for the Standards of Care in Neuroendocrine Tumors: biotherapy. Neuroendocrinology 2009;90:209-213
  968. Shah T, Caplin M. Biotherapy for metastatic endocrine tumours. Best Pract Res Clin Gastroenterol 2005;19:617-636
  969. Amoroso V, Pavel M, Claps M , et al. IFN-a  in advanced well-differentiated neuroendocrine tumors: the neglected drug? Future Oncol 2018;14:897-899
  970. Öberg K. Interferon in the management of neuroendocrine GEP-tumors. Digestion 2000;62:92-97
  971. Oberg K. Biotherapies for GEP-NETs. Best Pract Res Clin Gastroenterol 2012;26:833-841
  972. Pisegna JR, Slimak GG, Doppman JL , et al. An evaluation of human recombinant alpha interferon in patients with metastatic gastrinoma. Gastroenterology 1993;105:1179-1183
  973. Ozdirik B, Tacke F, Benz F , et al. A case report of an excellent response to interferon-a in a patient with functional metastasized neuroendocrine tumor refractory to other treatments. Medicine (Baltimore) 2020;99:e20820
  974. Cingarlini S, Bonomi M, Corbo V , et al. Profiling mTOR pathway in neuroendocrine tumors. Target Oncol 2012;7:183-188
  975. Lamberti G, Brighi N, Maggio I , et al. The Role of mTOR in Neuroendocrine Tumors: Future Cornerstone of a Winning Strategy? Int J Mol Sci 2018;19:747
  976. Gajate P, Alonso-Gordoa T, Martinez-Saez O , et al. Prognostic and predictive role of the PI3K-AKT-mTOR pathway in neuroendocrine neoplasms. Clin Transl Oncol 2018;20:561-569
  977. Yao JC. Molecular targeted therapy for carcinoid and islet-cell carcinoma. Best Pract Res Clin Endocrinol Metab 2007;21:163-172
  978. Grozinsky-Glasberg S, Pavel M. Inhibition of mTOR in carcinoid tumors. Target Oncol 2012;7:189-195
  979. Vijayvergia N, Dasari A. Targeted Therapies in the Management of Well-Differentiated Digestive and Lung Neuroendocrine Neoplasms. Curr Treat Options Oncol 2020;21:96
  980. Fazio N, Cinieri S, Lorizzo K , et al. Biological targeted therapies in patients with advanced enteropancreatic neuroendocrine carcinomas. Cancer Treat Rev 2010;36 Suppl 3:S87-S94
  981. Jensen RT, Delle Fave G. Promising advances in the treatment of malignant pancreatic endocrine tumors. N Engl J Med 2011;364:564-565
  982. Yao JC, Fazio N, Singh S , et al. Everolimus for the treatment of advanced, non-functional neuroendocrine tumours of the lung or gastrointestinal tract (RADIANT-4): a randomised, placebo-controlled, phase 3 study. Lancet 2016;387:968-977
  983. Beyens M, Vandamme T, Peeters M , et al. Resistance to targeted treatment of gastroenteropancreatic neuroendocrine tumors. Endocr Relat Cancer 2019;26:R109-R130
  984. Pozas J, San RM, Alonso-Gordoa T , et al. Targeting Angiogenesis in Pancreatic Neuroendocrine Tumors: Resistance Mechanisms. Int J Mol Sci 2019;20:4949
  985. Fazio N. Neuroendocrine tumors resistant to mammalian target of rapamycin inhibitors: A difficult conversion from biology to the clinic. World J Clin Oncol 2015;6:194-197
  986. Lauricella E, Mandriani B, Cavallo F , et al. Angiogenesis in NENs, with a focus on gastroenteropancreatic NENs: from biology to current and future therapeutic implications. Front Oncol 2022;12:957068
  987. Kulke MH, Ou FS, Niedzwiecki D , et al. Everolimus with or without bevacizumab in advanced pNET: CALGB 80701 (Alliance). Endocr Relat Cancer 2022;29:335-344
  988. Fjallskog ML, Lejonklou MH, Oberg KE , et al. Expression of molecular targets for tyrosine kinase receptor antagonists in malignant endocrine pancreatic tumors. Clin Cancer Res 2003;9:1469-1473
  989. Capdevila J, Salazar R. Molecular targeted therapies in the treatment of gastroenteropancreatic neuroendocrine tumors. Target Oncol 2009;4:287-296
  990. Gilbert JA, Adhikari LJ, Lloyd RV , et al. Molecular markers for novel therapies in neuroendocrine (carcinoid) tumors. Endocr Relat Cancer 2010;17:623-636
  991. Faivre S, Sablin MP, Dreyer C , et al. Novel anticancer agents in clinical trials for well-differentiated neuroendocrine tumors. Endocrinol Metab Clin North Am 2010;39:811-826
  992. Raymond E, Hobday T, Castellano D , et al. Therapy innovations: tyrosine kinase inhibitors for the treatment of pancreatic neuroendocrine tumors. Cancer Metastasis Rev 2011;30 Suppl 1:19-26
  993. Pavel ME, Wiedenmann B. Novel therapeutic agents for the treatment of gastroenteropancreatic neuroendocrine tumors. Horm Metab Res 2011;43:844-853
  994. Fazio N, Cella CA, Del Re M , et al. Pharmacodynamics, clinical findings and approval status of current and emerging tyrosine-kinase inhibitors for pancreatic neuroendocrine tumors. Expert Opin Drug Metab Toxicol 2019;15:993-1004
  995. Mosalem O, Sonbol MB, Halfdanarson TR , et al. Tyrosine Kinase Inhibitors and Immunotherapy Updates in Neuroendocrine Neoplasms. Best Pract Res Clin Endocrinol Metab 2023;101796
  996. Grillo F, Florio T, Ferrau F , et al. Emerging multitarget tyrosine kinase inhibitors in the treatment of neuroendocrine neoplasms. Endocr Relat Cancer 2018;25:R453-R466
  997. Toumpanakis C, Fazio N, Tiensuu JE , et al. Unmet Needs in Appendiceal Neuroendocrine Neoplasms. Neuroendocrinology 2019;108:37-44
  998. Xu J, Shen L, Zhou Z , et al. Surufatinib in advanced extrapancreatic neuroendocrine tumours (SANET-ep): a randomised, double-blind, placebo-controlled, phase 3 study. Lancet Oncol 2020;21:1500-1512
  999. Xu J, Shen L, Bai C , et al. Surufatinib in advanced pancreatic neuroendocrine tumours (SANET-p): a randomised, double-blind, placebo-controlled, phase 3 study. Lancet Oncol 2020;21:1489-1499
  1000. Ali MA, Shah SS, Tahir N , et al. Efficacy and toxicity of surufatinib in neuroendocrine tumors: A systematic review and meta-analysis. J Neuroendocrinol 2022;34:e13149
  1001. Lu X, Yan S, Koral KA , et al. Surufatinib for the treatment of advanced extrapancreatic neuroendocrine tumors. Expert Rev Anticancer Ther 2021;21:917-926
  1002. Raymond E, Dahan L, Raoul JL , et al. Sunitinib Malate for the Treatment of Pancreatic Neuroendocrine Tumors. N Engl J Med 2011;364:501-513
  1003. Blumenthal GM, Cortazar P, Zhang JJ , et al. FDA approval summary: sunitinib for the treatment of progressive well-differentiated locally advanced or metastatic pancreatic neuroendocrine tumors. Oncologist 2012;17:1108-1113
  1004. Raymond E, Kulke MH, Qin S , et al. Efficacy and Safety of Sunitinib in Patients with Well-Differentiated Pancreatic Neuroendocrine Tumours. Neuroendocrinology 2018;107:237-245
  1005. Valle JW, Borbath I, Rosbrook B , et al. Sunitinib in patients with pancreatic neuroendocrine tumors: update of safety data. Future Oncol 2019;15:1219-1230
  1006. Cives M, Strosberg J. Radionuclide Therapy for Neuroendocrine Tumors. Curr Oncol Rep 2017;19:9
  1007. Hicks RJ, Kwekkeboom DJ, Krenning E , et al. ENETS Consensus Guidelines for the Standards of Care in Neuroendocrine Neoplasia: Peptide Receptor Radionuclide Therapy with Radiolabeled Somatostatin Analogues. Neuroendocrinology 2017;105:295-309
  1008. Ramage J, Naraev BG, Halfdanarson TR. Peptide receptor radionuclide therapy for patients with advanced pancreatic neuroendocrine tumors. Semin Oncol 2018;45:236-248
  1009. Alsadik S, Yusuf S, Al-Nahhas A. Peptide Receptor Radionuclide Therapy for Pancreatic Neuroendocrine Tumours. Curr Radiopharm 2019;12:126-134
  1010. Starr JS, Sonbol MB, Hobday TJ , et al. Peptide Receptor Radionuclide Therapy for the Treatment of Pancreatic Neuroendocrine Tumors: Recent Insights. Onco Targets Ther 2020;13:3545-3555
  1011. van Vliet EI, Teunissen JJ, Kam BL , et al. Treatment of Gastroenteropancreatic Neuroendocrine Tumors with Peptide Receptor Radionuclide Therapy. Neuroendocrinology 2013;97:74-85
  1012. Kwekkeboom DJ, de Herder WW, Krenning EP. Somatostatin receptor-targeted radionuclide therapy in patients with gastroenteropancreatic neuroendocrine tumors. Endocrinol Metab Clin North Am 2011;40:173-185
  1013. Bergsma H, van Vliet EI, Teunissen JJ , et al. Peptide receptor radionuclide therapy (PRRT) for GEP-NETs. Best Pract Res Clin Gastroenterol 2012;26:867-881
  1014. Van Essen M, Sundin A, Krenning EP , et al. Neuroendocrine tumours: the role of imaging for diagnosis and therapy. Nat Rev Endocrinol 2014;10:102-114
  1015. Kwekkeboom DJ, Teunissen JJ, Bakker WH , et al. Radiolabeled somatostatin analog (177Lu-DOTA0,Tyr3)octreotate in patients with endocrine gastroenteropancreatic tumors. J Clin Oncol 2005;23:2754-2762
  1016. Merola E, Grana CM. Peptide Receptor Radionuclide Therapy (PRRT): Innovations and Improvements. Cancers (Basel) 2023;15:
  1017. Kipnis ST, Hung M, Kumar S , et al. Laboratory, Clinical, and Survival Outcomes Associated With Peptide Receptor Radionuclide Therapy in Patients With Gastroenteropancreatic Neuroendocrine Tumors. JAMA Netw Open 2021;4:e212274
  1018. Zhang J, Song Q, Cai L , et al. The efficacy of (177)Lu-DOTATATE peptide receptor radionuclide therapy (PRRT) in patients with metastatic neuroendocrine tumours: a systematic review and meta-analysis. J Cancer Res Clin Oncol 2020;146:1533-1543
  1019. Strosberg J, El-Haddad G, Wolin E , et al. Phase 3 Trial of (177)Lu-Dotatate for Midgut Neuroendocrine Tumors. N Engl J Med 2017;376:125-135
  1020. Wang LF, Lin L, Wang MJ , et al. The therapeutic efficacy of 177Lu-DOTATATE/DOTATOC in advanced neuroendocrine tumors: A meta-analysis. Medicine (Baltimore) 2020;99:e19304
  1021. Forrer F, Valkema R, Kwekkeboom DJ , et al. Neuroendocrine tumors.Peptide receptor radionuclide therapy. Best Pract Res Clin Endocrinol Metab 2007;21:111-129
  1022. Grozinsky-Glasberg S, Barak D, Fraenkel M , et al. Peptide receptor radioligand therapy is an effective treatment for the long-term stabilization of malignant gastrinomas. Cancer 2011;117:1377-1385
  1023. Dumont RA, Seiler D, Marincek N , et al. Survival after somatostatin based radiopeptide therapy with (90)Y-DOTATOC vs. (90)Y-DOTATOC plus (177)Lu-DOTATOC in metastasized gastrinoma. Am J Nucl Med Mol Imaging 2015;5:46-55
  1024. Hofland J, Herrera-Martinez AD, Zandee WT , et al. Management of carcinoid syndrome: a systematic review and meta-analysis. Endocr Relat Cancer 2019;26:R145-R156
  1025. Zhang J, Liu Q, Singh A , et al. Prognostic Value of (18)F-FDG PET/CT in a Large Cohort of 495 Patients with Advanced Metastatic Neuroendocrine Neoplasms (NEN) Treated with Peptide Receptor Radionuclide Therapy (PRRT). J Nucl Med 2020;61:1560-1569
  1026. Binderup T, Knigge U, Johnbeck CB , et al. (18)F-FDG-PET is superior to WHO grading as prognostic tool in neuroendocrine neoplasms and useful in guiding peptide receptor radionuclide therapy: a prospective 10-year follow-up study of 166 patients. J Nucl Med 2020;
  1027. Kim YI. Salvage peptide receptor radionuclide therapy in patients with progressive neuroendocrine tumors: a systematic review and meta-analysis. Nucl Med Commun 2021;42:451-458
  1028. Strosberg J, Leeuwenkamp O, Siddiqui MK. Peptide receptor radiotherapy re-treatment in patients with progressive neuroendocrine tumors: A systematic review and meta-analysis. Cancer Treat Rev 2021;93:102141
  1029. Sachs G, Shin JM, Briving C , et al. The pharmacology of the gastric acid pump: the H+,K+ ATPase. Annu Rev Pharmacol Toxicol 1995;35:277-305
  1030. Gedaly R, Daily MF, Davenport D , et al. Liver Transplantation for the Treatment of Liver Metastases From Neuroendocrine Tumors: An Analysis of the UNOS Database. Arch Surg 2011;146:953-958
  1031. Le Treut YP, Gregoire E, Klempnauer J , et al. Liver Transplantation for Neuroendocrine Tumors in Europe-Results and Trends in Patient Selection: A 213-Case European Liver Transplant Registry Study. Ann Surg 2013;257:807-815
  1032. Moris D, Tsilimigras DI, Ntanasis-Stathopoulos I , et al. Liver transplantation in patients with liver metastases from neuroendocrine tumors: A systematic review. Surgery 2017;162:525-536
  1033. Kim J, Zimmerman MA, Hong JC. Liver transplantation in the treatment of unresectable hepatic metastasis from neuroendocrine tumors. J Gastrointest Oncol 2020;11:601-608
  1034. Fan ST, Le Treut YP, Mazzaferro V , et al. Liver transplantation for neuroendocrine tumour liver metastases. HPB (Oxford) 2015;17:23-28
  1035. Lim C, Lahat E, Osseis M , et al. Liver Transplantation for Neuroendocrine Tumors: What Have We Learned? Semin Liver Dis 2018;38:351-356
  1036. Spolverato G, Bagante F, Tsilimigras DI , et al. Liver transplantation in patients with liver metastases from neuroendocrine tumors. Minerva Chir 2019;74:399-406
  1037. Muller PC, Pfister M, Eshmuminov D , et al. Liver transplantation versus liver resection for treatment of neuroendocrine liver metastasis: Appraisal of the current evidence. Hepatobiliary Pancreat Dis Int 2023;
  1038. Shah T, Manas DM, Ford SJ , et al. Where Are We Now with Liver Transplantation in Neuroendocrine Neoplasms? The Place of Liver Transplantation for Grades 1 and 2 Well-Differentiated Unresectable Liver Metastatic Neuroendocrine Tumours. Curr Oncol Rep 2023;25:135-144
  1039. Valvi D, Mei X, Gupta M , et al. Younger Age Is Associated with Improved Survival in Patients Undergoing Liver Transplantation Alone for Metastatic Neuroendocrine Tumors. J Gastrointest Surg 2020;
  1040. Vilchez V, Gedaly R. Liver transplantation for the treatment of neuroendocrine liver metastases. Best Pract Res Clin Endocrinol Metab 2016;30:141-147
  1041. Gregoire E, Le Treut YP. Liver transplantation for primary or secondary endocrine tumors. Transpl Int 2010;23:704-711
  1042. Le Treut YP, Gregoire E, Belghiti J , et al. Predictors of long-term survival after liver transplantation for metastatic endocrine tumors: an 85-case French multicentric report. Am J Transplant 2008;8:1205-1213
  1043. Maggio I, Manuzzi L, Lamberti G , et al. Landscape and Future Perspectives of Immunotherapy in Neuroendocrine Neoplasia. Cancers (Basel) 2020;12:832
  1044. Bosch F, Bruwer K, Altendorf-Hofmann A , et al. Immune checkpoint markers in gastroenteropancreatic neuroendocrine neoplasia. Endocr Relat Cancer 2019;26:293-301
  1045. Weber MM, Fottner C. Immune Checkpoint Inhibitors in the Treatment of Patients with Neuroendocrine Neoplasia. Oncol Res Treat 2018;41:306-312
  1046. Klein O, Kee D, Markman B , et al. Immunotherapy of Ipilimumab and Nivolumab in Patients with Advanced Neuroendocrine Tumors: A Subgroup Analysis of the CA209-538 Clinical Trial for Rare Cancers. Clin Cancer Res 2020;26:4454-4459
  1047. Mehnert JM, Bergsland E, O'Neil BH , et al. Pembrolizumab for the treatment of programmed death-ligand 1-positive advanced carcinoid or pancreatic neuroendocrine tumors: Results from the KEYNOTE-028 study. Cancer 2020;126:3021-3030
  1048. Xu G, Wang Y, Zhang H , et al. Immunotherapy and potential predictive biomarkers in the treatment of neuroendocrine neoplasia. Future Oncol 2020;
  1049. Al-Toubah T, Cives M, Strosberg J. Novel immunotherapy strategies for treatment of neuroendocrine neoplasms. Transl Gastroenterol Hepatol 2020;5:54
  1050. Rindi G, Wiedenmann B. Neuroendocrine neoplasia of the gastrointestinal tract revisited: towards precision medicine. Nat Rev Endocrinol 2020;16:590-607
  1051. Young K, Lawlor RT, Ragulan C , et al. Immune landscape, evolution, hypoxia-mediated viral mimicry pathways and therapeutic potential in molecular subtypes of pancreatic neuroendocrine tumours. Gut 2020;
  1052. Li Y, Fan Z, Zhang F , et al. Neoadjuvant therapy in pancreatic neuroendocrine neoplasms: A systematic review and meta-analysis. Front Oncol 2022;12:981575
  1053. Lania A, Ferraù F, Rubino M , et al. Neoadjuvant Therapy for Neuroendocrine Neoplasms: Recent Progresses and Future Approaches. Front Endocrinol (Lausanne) 2021;12:651438