Archives

Adolescent Bariatric Surgery

ABSTRACT

 

The prevalence of adolescent obesity has rapidly increased over the past several decades. With this increase, there has also been a rise in the prevalence of complications of obesity leading to premature mortality. While lifestyle and medical management remain a part of the initial treatment of obesity, these therapies have been shown to be inferior when compared to metabolic and bariatric surgery (MBS) for adolescents with severe obesity. A multidisciplinary approach is recommended to evaluate medically eligible candidates for MBS, prepare patients for surgery, and guide postoperative management. Laparoscopic sleeve gastrectomy (SG) and Roux-en-Y gastric bypass (RYGB) are the most common MBS procedures performed in both adolescent and adult patients. Postoperative hospital stays are generally short and long-term routine follow-up with the MBS team is recommended to monitor weight loss, resolution of complications of obesity, and to monitor for postoperative complications. Most adolescent MBS studies demonstrate an average percent body mass index loss between 25-29% after surgery. This is also associated with resolution or improvement of most complications of obesity at rates that are similar or superior to adult studies. Resolution and prevention of type 2 diabetes mellitus (T2DM) after MBS is a particularly compelling reason to pursue surgical treatment due to the complications from T2DM that occur over a patient’s lifetime as well as the overall burden of health-related costs. These adverse consequences of T2DM can be mitigated by early use of MBS. MBS is generally well tolerated. Complication rates are similar to adult patients therefore it is recommended to refer patients for MBS whenever they are medically qualified. Most common short-term (<30 days) complications include leak, bleeding, and surgical site infections. Most common long-term (>30 days) complications are nutritional deficiencies.

 

INTRODUCTION

 

The prevalence of worldwide overweight and obesity in adolescents has more than quadrupled since 1975. Currently, it is estimated that over 14 million children and adolescents age 2-19 years suffer from obesity in the United States alone (1, 2). Adolescents with obesity are at risk for developing significant comorbidities including insulin resistance, type 2 diabetes mellitus (T2DM), hypertension, dyslipidemia, obstructive sleep apnea, nonalcoholic fatty liver disease, depression, polycystic ovarian syndrome, impaired quality of life, cardiovascular disease, and longer term, certain malignancies (3-9). Similar to obesity, the prevalence of T2DM has been increasing dramatically (3). Obesity is a major risk factor the development of T2DM with overweight adolescents having close to a three times greater risk of developing T2DM when compared to adolescents with normal weight (10-12). Additionally, obesity in adolescence is associated with persistent obesity into adulthood, increased risk for obesity related comorbidities, and premature mortality in adulthood (13-15). Lifestyle and medical management remain the first-line treatment for adolescent obesity. However, current evidence suggests that pharmacotherapy, dietary, and behavioral modifications rarely lead to long-term weight loss in adolescents with severe obesity (16-18). The use of metabolic and bariatric surgery (MBS) in adolescents with severe obesity and complications of obesity has been shown to have superior results in both efficacy and durability (19). Despite growing evidence of the efficacy and durability of MBS for the treatment of severe obesity in adolescent patients, utilization of MBS in adolescent patients is low and there have been documented racial and socioeconomic disparities (20-22).

 

PREOPERATIVE EVALUATION

 

Multidisciplinary Program

 

A multidisciplinary approach is recommended when considering MBS for an adolescent (23, 24). At a minimum, this includes a bariatric surgeon with adolescent experience, pediatrician, dietitian, nurse, and pediatric psychologist. It is also important that the core providers have access to additional pediatric specialists including anesthesiologists, radiologists, and appropriate specialists to aid the management of complications of obesity (e.g., pulmonology, endocrinology, gastroenterology/hepatology). Adolescents undergoing preoperative work-up should be evaluated for the presence and severity of complications of obesity. Additionally, it is important for the multidisciplinary team to determine a potential patient and caregivers’ ability to assess the risks and benefits of surgery as well as to adhere to postoperative requirements including daily vitamin regimens and attending postoperative visits.

 

Patient Selection

 

BODY MASS INDEX (BMI)

 

The following criteria have been recommended by multiple panels of experts for consideration of weight loss surgery in adolescents under 18 years old: (4, 19, 25)

  • BMI ≥ 120 percent of the 95th percentile for BMI for age or BMI ≥ 35kg/m2, whichever is lower, with complications of obesity that have a significant effect on health (Table 1).
  • OR -
  • BMI ≥ 140 percent of the 95th percentile of BMI for age or BMI ≥ 40 kg/m2, whichever is lower

Of note, the BMI threshold for adult patients for the recommendation has been reduced in the 2022 guidelines, therefore changes to adolescent recommendations may follow suit in future updates (26).

 

Table 1. Qualifying Comorbidities for Consideration of MBS in Adolescents (4).

Obstructive sleep apnea (apnea-hypoxia index > 5)

Type 2 diabetes mellitus

Idiopathic intracranial hypertension

Nonalcoholic steatohepatitis

Blount’s disease

Slipped capital femoral epiphysis

Gastroesophageal Reflux Disease

Hypertension

 

CONTRAINDICATIONS

 

Contraindications to adolescent MBS are listed in Table 2.

 

Table 2. Contraindications to Adolescent MBS

Medically correctable cause of obesity

Ongoing substance abuse problem (within the preceding year)

Medical, psychiatric, psychosocial, or cognitive condition that prevents adherence to postoperative dietary and medication regimens or impairs decisional capacity

Current or planned pregnancy within 18 months of the procedure

Inability for patient or caregivers to comprehend risks and benefits of surgical weight loss procedure

 

AGE

 

A recent cohort analysis of more than 600,000 adolescents aged 13-17 found that 1 in 23 adolescents met criteria for MBS (27). A retrospective review of the Metabolic and Bariatric Surgery Accreditation and Quality Improvement Program (MBSAQIP) data registry from 2015 to 2018 demonstrated that adolescents and young adults only represented 3.7% of total MBS cases performed, suggesting significant underutilization within this population (28). Multiple studies have evaluated the safety and efficacy of MBS in younger adolescents. Current evidence suggests there are no significant clinical differences in outcomes between MBS in younger (e.g., <13 years) versus older adolescents (e.g., ≥13 years) (29-35). It is therefore not recommended to limit access to MBS based on patient’s age, physical maturity (e.g., bone age), or pubertal status. These findings have prompted increased advocacy for the use of MBS in the adolescent population by the American Academy of Pediatrics (19, 36).

 

TYPES OF SURGERY

 

Sleeve Gastrectomy

 

A laparoscopic sleeve gastrectomy (SG) results in the removal of the greater curvature of the stomach resulting in a smaller, tubular stomach that has a reduced capacity (Figure 1). Given the procedure is less complex than the Roux-en-Y gastric bypass (RYGB) and has less risk for micronutrient deficiencies, it is an appealing option for adolescents. Sleeve gastrectomies currently account for approximately 80% of bariatric procedures in adolescents (28, 37-39). A SG may also be converted to RYGB in the event additional MBS is indicated or in the setting of postoperative medically refractory gastroesophageal reflux disease (GERD).

 

Figure 1. Sleeve Gastrectomy.

 

Roux-en-Y Gastric Bypass

 

Laparoscopic Roux-en-Y gastric bypass (RYGB) involves creating a small, proximal gastric pouch which is separated from the remnant stomach and anastomosed to a Roux-limb of small bowel 70-150 cm distally (Figure 2). The RYGB results in similar weight loss when compared to SG and dramatically improves glycemic control (38, 40). The incidence of postoperative GERD is significantly less following RYGB compared to SG, making the procedure an attractive option for adolescents with GERD at baseline (41).

Figure 2. Roux-en-Y Gastric Bypass.

 

Others

 

Additional procedures including intragastric balloons are not currently approved by the United States Food and Drug Administration (FDA) for use in adolescents. Adjustable gastric bands have been previously used in the adolescent population. However, they have fallen out of favor due inferior efficacy compared to SG and RYGB (42).

 

POSTOPERATIVE MANAGEMENT

 

Inpatient  

 

Average inpatient stay is typically <2 days following both a SG and RYGB (43). Patients are monitored for immediate postoperative complications including a leak, bleeding, and venous thromboembolism (VTE). Following discharge, patients are seen at regular postoperative visits to monitor body weight, nutritional status, and to manage complications of obesity.

 

Diet

 

Following a SG or RYGB, patients gradually progress from a high protein liquid diet to incorporating small volumes of regular food. Patients are encouraged to eat three to four protein-rich meals a day while avoiding carbohydrate rich foods. Supplemental sugar-free fluids between meals are also essential following surgery to avoid dehydration. Patients are typically encouraged to avoid excessive fluids with meals to minimize nausea and maximize nutritional intake with meals due to the restrictive component of both procedures.

 

Postoperative nausea is not uncommon following surgery but typically self resolves. Meals high in carbohydrates or sugar and fats can result in dumping syndrome or weight regain following surgery. Some providers recommend limiting carbonated or caffeinated beverages following MBS based on theoretical concerns, however there is minimal evidence to support this apprehension.

 

Similar to non-operative weight loss recommendations, general recommendations including exercising for 30 to 60 minutes daily, drinking sugar-free fluids, and portion-controlled protein rich meals are the same. Overall, it is recommended that patient and caregiver meet with a dietitian prior to discharge to develop a plan tailored to patient’s specific nutritional needs. Regular follow-up visits with a dietitian are also recommended to assist with postoperative weight management and to monitor for nutritional deficiencies.

 

Nutritional Supplements and Monitoring

 

Although SG may be associated with a decreased risk of nutritional deficiencies when compared to RYGB, lifelong supplementation with vitamins and minerals is recommended following both operations (Table 3) (44). Patients are particularly at risk for deficiencies in iron, vitamin B12, and vitamin D. Additionally, lifelong annual monitoring of nutritional and micronutrient status is recommended with annual laboratory testing (Table 3). Adjustments in supplements may need to be made over time as specific deficiencies emerge. 

 

Table 3. Nutritional Supplementation and Monitoring Recommendations (45)

Nutritional Supplements

Standard multivitamin with folate or iron, or prenatal vitamin if female (once or twice daily)

Vitamin B12, 500mcg sublingually daily, or 1000mcg intramuscularly monthly

Calcium, 1200 to 1500mg daily (measured as elemental calcium) with 800 to 1000 international units of vitamin D.

Annual Nutritional Monitoring

Complete blood cell count with differential

Serum iron and ferritin

Red blood cell folate, serum vitamin B12, and serum homocysteine

Serum thiamin (vitamin B1)

Hepatic panel (including albumin, total protein, serum aminotransferase levels, gamma-glutamyl transpeptidase, and alkaline phosphatase

Calcium, 25-hydroxyvitamin D, and parathyroid hormone

Dual-energy x-ray absorptiometry (DXA) scan to monitor bone density (optimal frequency not yet established)

 

Pregnancy Prevention

 

Pregnancy should be avoided for 12 to 18 months following MBS to allow patients to achieve weight maintenance and to avoid potential micronutrient deficiencies which may affect both patient and fetus (46). Obesity can result in decreased fertility secondary to irregular menstruation and ovulatory dysfunction (47, 48). Weight loss after MBS has been shown to result in more regular ovulation and improved fertility (49, 50). In a retrospective review of 47 adolescents who underwent MBS surgery, seven pregnancies occurred, six of them within 10 to 22 months following surgery (51). While all six deliveries were healthy and at term, the twofold higher than anticipated pregnancy rate highlights the need for contraception counseling following MBS.

 

Multiple studies have evaluated the efficacy of hormonal contraceptive methods in patients with elevated BMIs and no definitive association was found between higher BMI and effectiveness of hormonal contraceptives (52). Due to concern for malabsorption after intestinal bypass procedures and the subsequent potential for decreased oral contraceptive efficacy, the American College of Obstetrics and Gynecology recommend using non-oral forms of hormonal contraception in patients who have undergone malabsorptive MBS (53). Additionally, oral contraceptives are associated with increased risk of venous thromboembolism (VTE) which may be worrisome for adolescents with elevated BMIs who already have a higher predisposition for VTE (54, 55).

 

Intrauterine devices (IUDs) are an appealing option following MBS in adolescent patients as they are one of the most effective contraception methods, do not increase risk of VTE, and can be placed at the time of surgery (56). Levonorgestrel-releasing IUDs have the added benefit of promoting amenorrhea which could help reduce the risk of iron deficiency anemia following surgery (57). Regardless of the form of contraception selection, adolescents should be counseled on safe sex practices including the use of barrier protection against sexually transmitted infections.

 

Adolescent patients who become pregnant following MBS should be counseled on adequate nutritional intake with close monitoring of iron, folate, and vitamin B12 levels. Additionally, one must be cautious when screening for gestational diabetes in pregnant patients who have undergone MBS. In a study of a 119 post-bariatric surgery pregnant patients, oral glucose tolerance test resulted in hypoglycemia in 83% of patients with history of RYGB and 55% of patients with history of SG (58). Alternative methods for screening such as capillary blood glucose measurements are therefore recommended. These methods recommend obtaining capillary blood glucose values before and after each meal for 3-7 days and using pre-and post-prandial capillary glucose values according to  recommended cut-off values for defining diabetes mellitus (59, 60).

 

Comorbidity Reassessment

 

Regular reassessment of complications of obesity should occur at routine intervals in the postoperative phase to monitor for resolution or need for continued management. Patients with T2DM should be evaluated by their endocrinologist every three months. Repeat polysomnography is generally obtained between three to six months after surgery for patients previously on continuous positive airway pressure therapy (61, 62). Twenty-four-hour blood pressure monitoring can also be repeated three months after surgery to demonstrate resolution or persistence of hypertension. Medication may be restarted if blood pressure is consistently ≥120 mmHg systolic or ≥80 mmHg diastolic. Patients with biopsy proven nonalcoholic fatty liver disease may be re-biopsied 12 months after surgery to document regression. Finally, patients’ mental health needs should be re-evaluated by a pediatric psychologist at 6 and 12 months after surgery.

 

In the setting of weight regain, patients should be monitored for complications of obesity. There is emerging evidence however, that some complications of obesity may be weight dependent and others non-weight dependent (63). Some surgeons will routinely obtain an upper gastrointestinal contrast study at 12 months after surgery or as needed to assess anatomy which may lead to weight regain. Anatomical abnormalities that may contribute to weight regain include a dilated gastric sleeve or gastrogastric fistula.

 

Follow Up

 

Close follow up with the multidisciplinary team including the bariatric surgeon, pediatrician, dietitian, and pediatric psychologist is strongly recommended. Patients are typically followed by a pediatrician to ensure ongoing continuity of care. It is important for the core providers to have access to pediatric specialists including endocrinology, gastroenterology/hepatology, and pulmonology as needed in those with complications of obesity that require ongoing monitoring or management. Additionally, a gynecologist for contraception counseling may be required for female patients. The transition from pediatric to adult medicine can be challenging in patients with chronic medical conditions and frequently requires assistance from multiple members of the team for transition care coordination and preparation as well as to ensure adequate communication, support, and education (64-66). 

 

OUTCOMES

 

Percent BMI Loss

 

Both SG and RYGB have resulted in clinically significant weight loss in adolescents. The efficacy of both procedures appears to be similar in the adolescent population(67). In a large, multicenter analysis of 177 adolescents who underwent RYGB and 306 adolescents who underwent SG, there was a three-year postoperative average percent BMI loss of 29% (95% CI, 26 to 33) and 25% (95% CI, 22 to 28) for RYGB and SG, respectively (38). Similar results were seen in the Teen Longitudinal Assessment of Bariatric Surgery (Teen-LABS) the largest prospective, observational study to date of 228 adolescents undergoing either RYGB or SG.  The three-year analysis showed an average 28% reduction in BMI following RYGB compared to 26% reduction following SG (62). In the 10 year analysis of Teen-LABS data for Roux-en-Y gastric bypass (RYGB, n = 161) and sleeve gastrectomy (SG, n = 99), 83% of those eligible for 10 year follow up completed the full decade of data collection (68). The findings revealed long-term BMI reductions for both procedures, with RYGB showing a 20.6% decrease and SG a 19.2% decrease. Furthermore, initial BMI loss (within the first six months) proved to be a strong predictor of 10-year outcomes.  It is noteworthy that some smaller, single center studies have demonstrated long term (7-14 year) BMI reductions after RYGB up to 30% in patients who underwent surgery in their adolescence (69-71). 

 

Complications of Obesity

 

TYPE 2 DIABETES MELLITUS  

 

Multiple studies have demonstrated improved glycemic control, even remission as well as prevention of T2DM following MBS, making a compelling case of MBS as a treatment for T2DM (40, 70, 72-75). Of the 242 adolescents enrolled in Teen-LABS, 29 had T2DM at baseline. By 3 years after the procedure, remission of T2DM occurred in 95% (95% CI, 85-100) of participants with no new cases of T2DM in those without the condition at baseline (62). Additionally, 19 participants had prediabetes at baseline with a 76% (95% CI, 56-97) rate of remission at 3 years (62). These remission rates in Teen-LABS were compared to adults who underwent MBS. The Teen-LABS study's 10-year metabolic findings demonstrated sustained improvements in comorbidities for most participants, with a remission rate of 55% for type 2 diabetes (68). In contrast to adult studies, diabetes outcomes when surgery was used in adolescents were comparable between RYGB and SG.  It is also worth mentioning that the long-term remission rate for type 2 diabetes (55% at 10 years) significantly exceeds that observed in adults undergoing bariatric surgery for diabetes, where long term remission rates have been estimated to be around 15% (76). These results highlight the durability of both weight loss and diabetes remission in adolescents undergoing RYGB. Finally, among those who underwent RYGB, adolescents were more likely to have remission of T2DM at 5 years with a remission rate of 86% compared to 53% in adults (77). These data underscore that the health benefits of bariatric surgery may be more pronounced in adolescents than in adults.

 

Similar findings were demonstrated in another study of 226 adolescents undergoing SG, of which 23% of patients were found to have T2DM. Eighty-five percent of patients with T2DM were on medication for diabetes prior to surgery and 89% achieved normal fasting plasma glucose and hemoglobin A1c levels without the use of medication postoperatively (61).

 

To compare surgical versus medical therapy for T2DM in adolescents with severe obesity, data from participants with T2DM enrolled in the Teen-LABS study were compared to participants of similar age and racial distribution from the Treatment Options of Type 2 Diabetes in Adolescents and Youth (TODAY) studies. Teen-LABS participants underwent MBS. TODAY participants were randomized to metformin alone or in combination with rosiglitazone or intensive lifestyle intervention, with insulin therapy given for glycemic progression. At two years, mean hemoglobin A1c concentration decreased from 6.8% to 5.5% in patients who underwent MBS compared to an increase from 6.4% to 7.8% in those enrolled in the TODAY study. Compared to baseline, average BMI decreased by 29% in Teen-LABS participants while the average BMI increased by 3.7% in TODAY participants (78). Cardiovascular disease (CVD) risk reduction was also explored in a secondary analysis of this study and despite higher pretreatment risk for CVD, treatment with MBS resulted in reduction of estimated CVD that were sustained at 5-year follow-up where medical therapy was associated with an increase in risk of CVD in adolescents with T2DM and severe obesity (79).

 

While these initial results are promising of the beneficial effects of MBS for the treatment of T2DM, no studies have prospectively compared the efficacy of MBS with that of medical therapy for the treatment of T2DM in adolescents with obesity. Additionally, the majority of initial MBS data in adolescents were from those who underwent RYGB which is no longer the primary MBS procedure performed in adolescents due to its inferior safety profile. In 2019, the National Institute of Health funded the Surgical or Medical Treatment for Pediatric T2DM (ST2OMP) trial which will compare SG to advanced medical therapy (80, 81).

 

OTHER COMORBIDITIES

 

In the Teen-LABS study described above, a mean 74% (95% CI, 64 to 84) remission of hypertension (HTN), 66% (95% CI 57 to 74) remission of dyslipidemia, and 86% (95% CI 72 to 100) resolution of abnormal kidney function was found at 3 years (62). In a secondary analysis of Teen-LABS and TODAY data, medical management of adolescents with obesity was associated with higher odds of diabetic kidney disease when compared to MBS (82). Greater weight loss after MBS in adolescents has also been associated with greater remission of T2DM, HTN, and dyslipidemia (63, 83). In a comparison of adolescents and adults who underwent RYGB, adolescents were more likely to have remission of HTN at 5 years compared to adults (68% vs 41%) (77).

 

Additional studies have demonstrated a 66% to 84% remission of obstructive sleep apnea as well as improvements in liver disease and polycystic ovarian syndrome (8, 61, 84). Improvements in functional mobility as well as reduction in musculoskeletal pain have also been well described (85, 86).

 

Mental Health

 

Multiple studies have reported higher rates of depression, emotional and behavioral disorders, and suicidal ideation in adolescents with obesity (87-90). Additionally, binge and loss of control eating is prevalent among more than one quarter of adolescents with overweight and obesity (91, 92). A recent prospective study demonstrated that undergoing MBS in adolescence did not heighten or lower the risk of suicidal thoughts or behaviors following the initial 4 years after surgery (93). While still unclear whether obesity leads to psychopathology, or vice versa, the association highlights the need for appropriate psychological services in the pre- and postoperative period (87).

 

MBS can lead to improvements in psychosocial outcomes, although the improvements in some studies are transient. In the TEEN-Labs study, quality of life measured by the Impact of Weight on Quality of Life and Short Form 36 Health Survey improved after MBS (62, 86). When compared to a nonsurgical control, Teen-LABS participants also demonstrated significantly higher levels of self-worth and romantic self-perceptions 6 years after surgery (94). Several studies have demonstrated improved depressive and anxiety symptoms in the months following MBS, although the results were not maintained after the first postoperative year (95, 96). In a multisite study assessing two year follow up of psychopathology prevalence in adolescents undergoing MBS, most patients retained their symptomatic or non-symptomatic psychopathology status at two years, although remission of symptoms was more prevalent than the development of new symptoms (97). These results emphasize the need for long-term psychosocial monitoring following MBS as well as early treatment in those with psychopathology.

 

Substance and alcohol abuse have been observed in post-MBS adolescents and adults. Pre and postoperative screening and education regarding substance and alcohol addiction should be integrated in long-term follow up care (98).  

 

Short-Term Complications

 

Short-term complications (<30 days after surgery) in adolescents undergoing MBS are similar to those seen in adults. Early postoperative complications, though rare, include surgical site infections, bleeding, leak, strictures, and pulmonary embolism. In a retrospective review of 21,592 adolescents and young adults who underwent SG or RYGB between 2015 and 2018, 3.7% of patients required readmission, 1.1% of patients required reoperation, and 3.3% required percutaneous, endoscopic, or other intervention (28). Major complications were rare; the most common complication was bleeding (0.4%), followed by leak (0.4%), and deep surgical site infections (0.2%). RYGB was associated with higher rates of reoperation (2.1% vs. 0.8%), readmission (6.3% vs. 3.0%), and serious complications (5.5% vs. 1.8%) compared to SG. Mortality occurred in 0.05% of patients and there were no differences in mortality noted between groups (28). Similar complication rates were found in a more recent analysis of the Metabolic and Bariatric Surgery Accreditation and Quality Improvement Program database in patients aged 10-19 years old (99).  In an additional retrospective review of 483 adolescents (SG n=306, RYGB n=177) no perioperative deaths occurred and the rate of major adverse events were too rare for statistical comparison. VTEs occurred in only 0.4% of patients and failure to discharge in 30 days was observed in 0.7% of patients (38).

 

Multiple studies have also suggested that MBS may be safer in adolescents when compared with adults. In a large study evaluating perioperative outcomes of MBS between 309 adolescents and 55,192 adults, the overall 30-day complication rate was significantly lower in adolescents (5.5%) as compared with adults (9.8%). No in-hospital mortalities occurred in the adolescent group compared to 0.2% in the adult group. The 30-day morbidity for adolescents following SG was zero compared to 4.3% following RYGB (100). In an additional study evaluating 1047 adolescents,10,429 college-aged individuals, and 24,841 young adults who underwent SG or RYGB, there were no differences in 30-day complication rates between age groups (101).

 

Long-Term Complications

 

NUTRITIONAL DEFICIENCIES  

 

Long-term complications after MBS in adolescents are primarily nutritional. Patients are particularly at risk for deficiencies in iron, vitamin B12, and vitamin D. Iron deficiency is common in premenopausal females due to menstruation. Some patients may require iron infusion if oral supplementation is not adequate. Symptomatic thiamine deficiency following MBS is rare, however can have serious consequences (102-104).These risks are higher for patients who undergo RYGB compared to SG due to potential malabsorption. In a Teen-LABS study evaluating nutritional deficiencies at 5 years postoperatively, low serum ferritin levels were seen in 71% of patients who underwent RYGB compared to 45% following a SG indicating iron deficiency (102). Iron deficiency anemia can occasionally be severe in adolescent women following MBS which can be compounded by menstruation and challenges in recognizing symptoms therefore daily supplementation and routine nutritional monitoring is essential following MBS.

 

Vitamin B12 deficiency was seen in approximately 12% of patients after either procedure. Approximately 40% of patients had low vitamin D levels at baseline with no significant change at follow up. Parathyroid hormone concentrations increased in patients undergoing RYGB from an average baseline concentration of 44 pg/ml to 59 pg/ml at 5 years with the risk of abnormal parathyroid hormone levels nearly sixfold higher after RYGB compared with SG (102). Elevated parathyroid hormone is utilized as a surrogate for calcium deficiency and raises concerns about long-term bone health. In adolescents, reduced bone mass has been noted 5-11 years after MBS, whether this increases long term fracture risk remains unclear.(105, 106). Concerns of growth retardation after MBS have been refuted and the most recent adolescent ASMBS guidelines have removed the recommendation of patients reaching physical maturity prior to MBS (4, 30).

 

The risk of nutritional deficiencies decreases with adherence to prescribed micronutrient supplements and increases with pregnancy (102). Given the high prevalence of nutritional deficiencies, lifelong micronutrient supplementation is required following surgery. One concern emphasized in the adolescent population is adherence to regular multivitamin use. In a prospective study of 41 adolescents who underwent MBS, multivitamin adherence was only 29.8%  23.9 (107).

 

WEIGHT REGAIN

 

Current data demonstrates satisfactory maintenance of weight loss at long-term follow up with both SG and RYGB (38, 71, 108), but in other detailed analysis of weight regain trajectories, there are distinct groups that emerge in a large enough dataset. The Teen-LABS data at 10 years showed that 38% of the cohort experienced moderate weight regain. One trajectory group experienced a nadir of 25% weight loss at five years but only maintained 13% loss at 10 years.  Another group (11% of the cohort) showed poor results with a weight loss peak at 20% at five years, but then proceeding to regain all weight from 5-10 years, resulting in a 7% BMI increase (above baseline) by 10 years (68). Some adult studies have demonstrated utility of anti-obesity medications after MBS to mitigate weight regain after surgery, however this has not been thoroughly explored in the adolescent population (109, 110). More research is needed to fully understand the mechanisms behind long-term weight maintenance after MBS.

 

OTHER COMPLICATIONS  

 

Cholelithiasis is a common complication due to rapid weight loss following MBS in both adolescents and adults. In the Teen-LABS study, cholecystectomy was required within three years in 9.9% of adolescents who underwent RYGB and 5.1% who underwent SG (62). Five percent of Teen-LABS participants required other abdominal operations including lysis of adhesions, gastrostomy, ventral hernia repair, or internal hernia repair (62). Symptoms of GERD, nausea, bloating, and diarrhea can also increase following MBS. During five years of follow up, the incidence of GERD increased from 2% to 8% in adolescents who underwent RYGB and from 11% to 24% in those who underwent SG. At five years postoperatively, the SG group had more than fourfold greater odds of having gastrointestinal distress symptoms when compared to RYGB (41). Dumping syndrome can been seen after both procedures, however it is much more common after RYGB compared to SG (111, 112). The incidence of dumping syndrome (~12%) in adolescents after RYGB was similar to adult patients two years after surgery(113).

 

There are no current established guidelines for surveillance of Barrett’s esophagus after SG for adolescent patients, however routine screening is recommended for adult patients after SG, therefore it would be prudent for adolescent patients to undergo intermittent surveillance also as the length of possible GERD exposure is theoretically longer (114). Similarly, there are no established guidelines for monitoring of bone density following use of MBS in adolescence, but due to inadequate vitamin D levels and rising PTH at least in those who underwent RYGB, periodic monitoring with DEXA may be prudent.

 

Emerging Evidence

 

Current evidence evaluating the outcomes and efficacy of adolescent MBS is generally limited to ≤10 years of follow up. Smaller, long-term studies with data available for up to 18 years post-operatively in patients who primary underwent RYGB demonstrate the durability of weight loss and similar rates of complications, although inference is limited due to small sample sizes with reduced attrition rates (70-72, 115, 116). Characteristics including study size, length of follow up, and attrition rate of available studies on MBS published from 2012 to present are available in Table 4. As SG is now the most predominate MBS procedure performed in the Unites States long-term data with this procedure is required. While some longitudinal studies are ongoing (Table 4), there remains a paucity of long-term data in the adolescent population.

 

Table 4. Characteristics of Studies on MBS, 2012 – Present

Author; Year

Study Design

Sample Size (N)

Type of MBS

 

 

RYBG     SG

Longest follow up

N

(1 yr)

N

(3 yr)

N

(5 yr)

Comments

Inge;

2018 (38)

RO 

483

177

306

5 yr

466 (96%)

153 (32%)

41 (8%)

The PCORnet bariatric study (2005 – 2015)

Olbers; 2017 (69)

CC

81

81

0

5 yr

81

(100%)

n/a

81 (100%)

Adolescent Morbid Obesity Surgery (AMOS) study

Inge; 2017(70)

PO

74

74

0

12.5 yr

n/a

n/a

58 (81%)

Adolescent Bariatric Surgery at 5 Plus Years (FABS-5+) study (2001-2007); mean follow up 8.0 yr

Inge; 2016 (62)

PO 

228

161

67

3 yr

205 (90%)

194 (85%)

n/a

Teen-Longitudinal Assessment of Bariatric Surgery (Teen-LABS) study; (2007-2012)

Vilallonga; 2016 (72)

RO

19

19

0

10.2 yr

n/a

n/a

n/a

Mean follow up 7.2 years; (2003-2008)

Al-Sabah; 2015 (73)

RO

125

0

135

4 yr

54 (40%)

n/a

n/a

2 yr follow up: 46 (34%); (2008-2012)

Cozacov; 2014 (115)

RO

18

8

10

7 yr

15 (83%)

10 (56%)

n/a

7 yr follow up: 3 (17%); (2002 – 2011)

Messiah; 2013 (84)

PO

454

454

0

1 yr

108 (24%)

n/a

n/a

Bariatric Outcomes Longitudinal Database (BOLD) (2004-2010)

Alqahtani; 2012 (31)

RO

108

0

108

2 yr

41 (38%)

n/a

n/a

2 yr follow-up: 8 (7%); (2008 – 2011)

Nijhawan; 2012 (116)

RO

25

25

0

9 yr

n/a

n/a

20 (80%)

Study dates not provided

 

de la Cruz-Muñoz; 2022 (71)

RO

96

87

1

18 yr

n/a

n/a

n/a

Mean follow up 14.2 years (2002-2010).

RO- Retrospective observational; CC- Case-control; PO- Prospective observational

 

CONCLUSION

 

Surgical weight loss is an appropriate consideration for adolescents with severe obesity and/or complications of obesity who have failed to lose weight through other obesity management options. It is essential that adolescents undergoing evaluation for MBS do so in the context of a multidisciplinary program with specific expertise in adolescent medicine and MBS. SG and RYGB are safe and effective treatment options in adolescents. Weight loss outcomes are comparable between SG and RYGB. Both procedures also result in substantial improvement in complications of obesity, including T2DM. SG appears to have an improved safety profile when compared to RYGB and is now the most common adolescent bariatric procedure performed in the United States. Emerging evidence demonstrates advantages of earlier surgical intervention in those with obesity including improved weight loss, increased resolution of comorbidities, and decreased adverse events when compared to adults (77, 117). Perioperative complications in adolescents undergoing MBS are similar to those in adults but occur less frequently (100, 101). Long-term complications are primarily nutritional and life-long vitamin and mineral supplementation is recommended. Regular follow up is required following MBS and it is important for patients to have access to appropriate medical, dietary, and psychological care.

 

REFERENCES

 

  1. Obesity and overweight World Health Organization2020 [Available from: https://www.who.int/news-room/fact-sheets/detail/obesity-and-overweight.
  2. Prevention CfDCa. Childhood Obesity Facts[updated APRIL 2, 2024; cited 2024 April 2, 2024]. Available from: https://www.cdc.gov/obesity/php/data-research/childhood-obesity-facts.html.
  3. Rosenbaum M. Special Considerations Relevant to Pediatric Obesity. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dhatariya K, et al., editors. Endotext. South Dartmouth (MA)2000.
  4. Pratt JSA, Browne A, Browne NT, Bruzoni M, Cohen M, Desai A, et al. ASMBS pediatric metabolic and bariatric surgery guidelines, 2018. Surg Obes Relat Dis. 2018;14(7):882-901.
  5. Xanthakos SA, Jenkins TM, Kleiner DE, Boyce TW, Mourya R, Karns R, et al. High Prevalence of Nonalcoholic Fatty Liver Disease in Adolescents Undergoing Bariatric Surgery. Gastroenterology. 2015;149(3):623-34.e8.
  6. Zeller MH, Roehrig HR, Modi AC, Daniels SR, Inge TH. Health-related quality of life and depressive symptoms in adolescents with extreme obesity presenting for bariatric surgery. Pediatrics. 2006;117(4):1155-61.
  7. Legro RS, Arslanian SA, Ehrmann DA, Hoeger KM, Murad MH, Pasquali R, et al. Diagnosis and treatment of polycystic ovary syndrome: an Endocrine Society clinical practice guideline. J Clin Endocrinol Metab. 2013;98(12):4565-92.
  8. Kaar JL, Morelli N, Russell SP, Talker I, Moore JM, Inge TH, et al. Obstructive sleep apnea and early weight loss among adolescents undergoing bariatric surgery. Surg Obes Relat Dis. 2021;17(4):711-7.
  9. Avgerinos KI, Spyrou N, Mantzoros CS, Dalamaga M. Obesity and cancer risk: Emerging biological mechanisms and perspectives. Metabolism. 2019;92:121-35.
  10. Tirosh A, Shai I, Afek A, Dubnov-Raz G, Ayalon N, Gordon B, et al. Adolescent BMI trajectory and risk of diabetes versus coronary disease. N Engl J Med. 2011;364(14):1315-25.
  11. Abdullah A, Stoelwinder J, Shortreed S, Wolfe R, Stevenson C, Walls H, et al. The duration of obesity and the risk of type 2 diabetes. Public Health Nutr. 2011;14(1):119-26.
  12. Lee JM, Gebremariam A, Vijan S, Gurney JG. Excess body mass index-years, a measure of degree and duration of excess weight, and risk for incident diabetes. Arch Pediatr Adolesc Med. 2012;166(1):42-8.
  13. Fontaine KR, Redden DT, Wang C, Westfall AO, Allison DB. Years of life lost due to obesity. Jama. 2003;289(2):187-93.
  14. Greenberg JA. Obesity and early mortality in the United States. Obesity (Silver Spring, Md). 2013;21(2):405-12.
  15. Inge TH, King WC, Jenkins TM, Courcoulas AP, Mitsnefes M, Flum DR, et al. The effect of obesity in adolescence on adult health status. Pediatrics. 2013;132(6):1098-104.
  16. Berkowitz RI, Wadden TA, Gehrman CA, Bishop-Gilyard CT, Moore RH, Womble LG, et al. Meal replacements in the treatment of adolescent obesity: a randomized controlled trial. Obesity (Silver Spring, Md). 2011;19(6):1193-9.
  17. Andela S, Burrows TL, Baur LA, Coyle DH, Collins CE, Gow ML. Efficacy of very low-energy diet programs for weight loss: A systematic review with meta-analysis of intervention studies in children and adolescents with obesity. Obes Rev. 2019;20(6):871-82.
  18. Srivastava G, Fox CK, Kelly AS, Jastreboff AM, Browne AF, Browne NT, et al. Clinical Considerations Regarding the Use of Obesity Pharmacotherapy in Adolescents with Obesity. Obesity (Silver Spring, Md). 2019;27(2):190-204.
  19. Armstrong SC, Bolling CF, Michalsky MP, Reichard KW, Section On Obesity SOS. Pediatric Metabolic and Bariatric Surgery: Evidence, Barriers, and Best Practices. Pediatrics. 2019;144(6).
  20. Bouchard ME, Tian Y, Linton S, De Boer C, O'Connor A, Ghomrawi H, et al. Utilization Trends and Disparities in Adolescent Bariatric Surgery in the United States 2009-2017. Child Obes. 2022;18(3):188-96.
  21. Salimi-Jazi F, Chkhikvadze T, Shi J, Pourmehdi-Lahiji A, Moshksar A, Rafeeqi TA, et al. Trends in Adolescent Bariatric Procedures: a 15-Year Analysis of the National Inpatient Survey. Obes Surg. 2022;32(11):3658-65.
  22. Messiah SE, Xie L, Atem F, Mathew MS, Qureshi FG, Schneider BE, et al. Disparity Between United States Adolescent Class II and III Obesity Trends and Bariatric Surgery Utilization, 2015-2018. Ann Surg. 2022;276(2):324-33.
  23. Inge TH, Krebs NF, Garcia VF, Skelton JA, Guice KS, Strauss RS, et al. Bariatric surgery for severely overweight adolescents: concerns and recommendations. Pediatrics. 2004;114(1):217-23.
  24. Styne DM, Arslanian SA, Connor EL, Farooqi IS, Murad MH, Silverstein JH, et al. Pediatric Obesity-Assessment, Treatment, and Prevention: An Endocrine Society Clinical Practice Guideline. J Clin Endocrinol Metab. 2017;102(3):709-57.
  25. Cuda S, O'Hara V, Censani M, Conroy R, Sweeney B, Paisley J, et al. Special considerations for the adolescent with obesity: An obesity medicine association (OMA) clinical practice statement (CPS) 2024. Obes Pillars. 2024;9:100096.
  26. Eisenberg D, Shikora SA, Aarts E, Aminian A, Angrisani L, Cohen RV, et al. 2022 American Society of Metabolic and Bariatric Surgery (ASMBS) and International Federation for the Surgery of Obesity and Metabolic Disorders (IFSO) Indications for Metabolic and Bariatric Surgery. Obes Surg. 2023;33(1):3-14.
  27. Shapiro WL, Kunani P, Sidell MA, Li X, Anderson SR, Slezak JM, et al. Prevalence of Adolescents Meeting Criteria for Metabolic and Bariatric Surgery. Pediatrics. 2024;153(3).
  28. Mocanu V, Lai K, Dang JT, Switzer NJ, Birch DW, Ball GDC, et al. Evaluation of the Trends, Characteristics, and Outcomes in North American Youth Undergoing Elective Bariatric Surgery. Obes Surg. 2021;31(5):2180-7.
  29. Ogle SB, Dewberry LC, Jenkins TM, Inge TH, Kelsey M, Bruzoni M, et al. Outcomes of Bariatric Surgery in Older Versus Younger Adolescents. Pediatrics. 2021;147(3).
  30. Alqahtani A, Elahmedi M, Qahtani AR. Laparoscopic Sleeve Gastrectomy in Children Younger Than 14 Years: Refuting the Concerns. Ann Surg. 2016;263(2):312-9.
  31. Alqahtani AR, Antonisamy B, Alamri H, Elahmedi M, Zimmerman VA. Laparoscopic sleeve gastrectomy in 108 obese children and adolescents aged 5 to 21 years. Ann Surg. 2012;256(2):266-73.
  32. El Chaar M, King K, Al-Mardini A, Galvez A, Claros L, Stoltzfus J. Thirty-Day Outcomes of Bariatric Surgery in Adolescents: a First Look at the MBSAQIP Database. Obes Surg. 2021;31(1):194-9.
  33. Poliakin L, Roberts A, Thompson KJ, Raheem E, McKillop IH, Nimeri A. Outcomes of adolescents compared with young adults after bariatric surgery: an analysis of 227,671 patients using the MBSAQIP data registry. Surg Obes Relat Dis. 2020;16(10):1463-73.
  34. Shah A, Liang NE, Bruzoni M, Pratt JSA, Zitsman J, Nadler EP. Outcomes after metabolic and bariatric surgery in preteens versus teens using the Metabolic and Bariatric Surgery Accreditation and Quality Improvement Program database and center-specific data. Obesity (Silver Spring, Md). 2024;32(1):150-5.
  35. Liang NE, Herdes RE, Balili R, Pratt JSA, Bruzoni M. Sleeve gastrectomy for the treatment of adolescent obesity in children aged 13 and under: a retrospective study. Surg Obes Relat Dis. 2024;20(4):354-61.
  36. Steinberger AE, Nickel KB, Keller M, Wallendorf M, Sprague J, Nicol GE, et al. National Trends in Pediatric Metabolic and Bariatric Surgery: 2010-2017. Pediatrics. 2022;150(6).
  37. Alqahtani A, Alamri H, Elahmedi M, Mohammed R. Laparoscopic sleeve gastrectomy in adult and pediatric obese patients: a comparative study. Surg Endosc. 2012;26(11):3094-100.
  38. Inge TH, Coley RY, Bazzano LA, Xanthakos SA, McTigue K, Arterburn D, et al. Comparative effectiveness of bariatric procedures among adolescents: the PCORnet bariatric study. Surg Obes Relat Dis. 2018;14(9):1374-86.
  39. Jackson WL, Lewis SR, Bagby JP, Hilton LR, Milad M, Bledsoe SE. Laparoscopic sleeve gastrectomy versus laparoscopic Roux-en-Y gastric bypass in the pediatric population: a MBSAQIP analysis. Surg Obes Relat Dis. 2020;16(2):254-60.
  40. Inge TH, Miyano G, Bean J, Helmrath M, Courcoulas A, Harmon CM, et al. Reversal of type 2 diabetes mellitus and improvements in cardiovascular risk factors after surgical weight loss in adolescents. Pediatrics. 2009;123(1):214-22.
  41. Dewberry LC, Khoury JC, Ehrlich S, Jenkins TM, Beamish AJ, Kalkwarf HJ, et al. Change in gastrointestinal symptoms over the first 5 years after bariatric surgery in a multicenter cohort of adolescents. J Pediatr Surg. 2019;54(6):1220-5.
  42. Dewberry LC, Jalivand A, Gupta R, Jenkins TM, Beamish A, Inge TH, et al. Weight Loss and Health Status 5 Years After Adjustable Gastric Banding in Adolescents. Obes Surg. 2020;30(6):2388-94.
  43. Svetanoff WJ, Diefenbach K, Hall B, Craver A, Rutledge S, McManaway C, et al. Utilization of an Enhanced Recovery After Surgery (ERAS) protocol for pediatric metabolic and bariatric surgery. J Pediatr Surg. 2023;58(4):695-701.
  44. Parrott J, Frank L, Rabena R, Craggs-Dino L, Isom KA, Greiman L. American Society for Metabolic and Bariatric Surgery Integrated Health Nutritional Guidelines for the Surgical Weight Loss Patient 2016 Update: Micronutrients. Surg Obes Relat Dis. 2017;13(5):727-41.
  45. Mechanick JI, Apovian C, Brethauer S, Timothy Garvey W, Joffe AM, Kim J, et al. Clinical Practice Guidelines for the Perioperative Nutrition, Metabolic, and Nonsurgical Support of Patients Undergoing Bariatric Procedures - 2019 Update: Cosponsored by American Association of Clinical Endocrinologists/American College of Endocrinology, The Obesity Society, American Society for Metabolic and Bariatric Surgery, Obesity Medicine Association, and American Society of Anesthesiologists. Obesity (Silver Spring, Md). 2020;28(4):O1-o58.
  46. Mechanick JI, Youdim A, Jones DB, Garvey WT, Hurley DL, McMahon MM, et al. Clinical practice guidelines for the perioperative nutritional, metabolic, and nonsurgical support of the bariatric surgery patient--2013 update: cosponsored by American Association of Clinical Endocrinologists, The Obesity Society, and American Society for Metabolic & Bariatric Surgery. Obesity (Silver Spring, Md). 2013;21 Suppl 1(0 1):S1-27.
  47. Broughton DE, Moley KH. Obesity and female infertility: potential mediators of obesity's impact. Fertil Steril. 2017;107(4):840-7.
  48. Silvestris E, de Pergola G, Rosania R, Loverro G. Obesity as disruptor of the female fertility. Reprod Biol Endocrinol. 2018;16(1):22.
  49. Teitelman M, Grotegut CA, Williams NN, Lewis JD. The impact of bariatric surgery on menstrual patterns. Obes Surg. 2006;16(11):1457-63.
  50. Escobar-Morreale HF, Santacruz E, Luque-Ramírez M, Botella Carretero JI. Prevalence of 'obesity-associated gonadal dysfunction' in severely obese men and women and its resolution after bariatric surgery: a systematic review and meta-analysis. Hum Reprod Update. 2017;23(4):390-408.
  51. Roehrig HR, Xanthakos SA, Sweeney J, Zeller MH, Inge TH. Pregnancy after gastric bypass surgery in adolescents. Obes Surg. 2007;17(7):873-7.
  52. Lopez LM, Bernholc A, Chen M, Grey TW, Otterness C, Westhoff C, et al. Hormonal contraceptives for contraception in overweight or obese women. Cochrane Database Syst Rev. 2016(8):Cd008452.
  53. ACOG practice bulletin no. 105: bariatric surgery and pregnancy. Obstet Gynecol. 2009;113(6):1405-13.
  54. de Bastos M, Stegeman BH, Rosendaal FR, Van Hylckama Vlieg A, Helmerhorst FM, Stijnen T, et al. Combined oral contraceptives: venous thrombosis. Cochrane Database Syst Rev. 2014(3):Cd010813.
  55. Lassandro G, Palmieri VV, Palladino V, Amoruso A, Faienza MF, Giordano P. Venous Thromboembolism in Children: From Diagnosis to Management. Int J Environ Res Public Health. 2020;17(14).
  56. ACOG Committee Opinion No. 735: Adolescents and Long-Acting Reversible Contraception: Implants and Intrauterine Devices. Obstet Gynecol. 2018;131(5):e130-e9.
  57. Apter D. International Perspectives: IUDs and Adolescents. J Pediatr Adolesc Gynecol. 2019;32(5s):S36-s42.
  58. Rottenstreich A, Elazary R, Ezra Y, Kleinstern G, Beglaibter N, Elchalal U. Hypoglycemia during oral glucose tolerance test among post-bariatric surgery pregnant patients: incidence and perinatal significance. Surg Obes Relat Dis. 2018;14(3):347-53.
  59. Benhalima K, Minschart C, Ceulemans D, Bogaerts A, Van Der Schueren B, Mathieu C, et al. Screening and Management of Gestational Diabetes Mellitus after Bariatric Surgery. Nutrients. 2018;10(10).
  60. Minschart C, Beunen K, Benhalima K. An Update on Screening Strategies for Gestational Diabetes Mellitus: A Narrative Review. Diabetes Metab Syndr Obes. 2021;14:3047-76.
  61. Alqahtani AR, Elahmedi MO, Al Qahtani A. Co-morbidity resolution in morbidly obese children and adolescents undergoing sleeve gastrectomy. Surg Obes Relat Dis. 2014;10(5):842-50.
  62. Inge TH, Courcoulas AP, Jenkins TM, Michalsky MP, Helmrath MA, Brandt ML, et al. Weight Loss and Health Status 3 Years after Bariatric Surgery in Adolescents. N Engl J Med. 2016;374(2):113-23.
  63. Derderian SC, Patten L, Kaizer AM, Moore JM, Ogle S, Jenkins TM, et al. Influence of Weight Loss on Obesity-Associated Complications After Metabolic and Bariatric Surgery in Adolescents. Obesity (Silver Spring, Md). 2020;28(12):2397-404.
  64. Wafa S, Nakhla M. Improving the Transition from Pediatric to Adult Diabetes Healthcare: A Literature Review. Can J Diabetes. 2015;39(6):520-8.
  65. Coyne I, Sheehan A, Heery E, While AE. Healthcare transition for adolescents and young adults with long-term conditions: Qualitative study of patients, parents and healthcare professionals' experiences. J Clin Nurs. 2019;28(21-22):4062-76.
  66. Butalia S, McGuire KA, Dyjur D, Mercer J, Pacaud D. Youth with diabetes and their parents' perspectives on transition care from pediatric to adult diabetes care services: A qualitative study. Health Sci Rep. 2020;3(3):e181.
  67. Herouvi D, Soldatou A, Paschou SA, Kalpia C, Karanasios S, Karavanaki K. Bariatric surgery in the management of childhood and adolescence obesity. Endocrine. 2023;79(3):411-9.
  68. Ryder JR, Jenkins TM, Xie C, Courcoulas AP, Harmon CM, Helmrath MA, et al. Ten-Year Outcomes after Bariatric Surgery in Adolescents. N Engl J Med. 2024;391(17):1656-8.
  69. Olbers T, Beamish AJ, Gronowitz E, Flodmark CE, Dahlgren J, Bruze G, et al. Laparoscopic Roux-en-Y gastric bypass in adolescents with severe obesity (AMOS): a prospective, 5-year, Swedish nationwide study. Lancet Diabetes Endocrinol. 2017;5(3):174-83.
  70. Inge TH, Jenkins TM, Xanthakos SA, Dixon JB, Daniels SR, Zeller MH, et al. Long-term outcomes of bariatric surgery in adolescents with severe obesity (FABS-5+): a prospective follow-up analysis. Lancet Diabetes Endocrinol. 2017;5(3):165-73.
  71. de la Cruz-Munoz N, Xie L, Quiroz HJ, Kutlu OC, Atem F, Lipshultz SE, et al. Long-Term Outcomes after Adolescent Bariatric Surgery. J Am Coll Surg. 2022;235(4):592-602.
  72. Vilallonga R, Himpens J, van de Vrande S. Long-Term (7 Years) Follow-Up of Roux-en-Y Gastric Bypass on Obese Adolescent Patients (<18 Years). Obes Facts. 2016;9(2):91-100.
  73. Al-Sabah SK, Almazeedi SM, Dashti SA, Al-Mulla AY, Ali DA, Jumaa TH. The efficacy of laparoscopic sleeve gastrectomy in treating adolescent obesity. Obes Surg. 2015;25(1):50-4.
  74. Syn NL, Cummings DE, Wang LZ, Lin DJ, Zhao JJ, Loh M, et al. Association of metabolic-bariatric surgery with long-term survival in adults with and without diabetes: a one-stage meta-analysis of matched cohort and prospective controlled studies with 174 772 participants. Lancet. 2021;397(10287):1830-41.
  75. Stefater MA, Inge TH. Bariatric Surgery for Adolescents with Type 2 Diabetes: an Emerging Therapeutic Strategy. Curr Diab Rep. 2017;17(8):62.
  76. Courcoulas AP, Patti ME, Hu B, Arterburn DE, Simonson DC, Gourash WF, et al. Long-Term Outcomes of Medical Management vs Bariatric Surgery in Type 2 Diabetes. Jama. 2024;331(8):654-64.
  77. Inge TH, Courcoulas AP, Jenkins TM, Michalsky MP, Brandt ML, Xanthakos SA, et al. Five-Year Outcomes of Gastric Bypass in Adolescents as Compared with Adults. N Engl J Med. 2019;380(22):2136-45.
  78. Inge TH, Laffel LM, Jenkins TM, Marcus MD, Leibel NI, Brandt ML, et al. Comparison of Surgical and Medical Therapy for Type 2 Diabetes in Severely Obese Adolescents. JAMA Pediatr. 2018;172(5):452-60.
  79. Ryder JR, Xu P, Nadeau KJ, Kelsey MM, Xie C, Jenkins T, et al. Effect of surgical versus medical therapy on estimated cardiovascular event risk among adolescents with type 2 diabetes and severe obesity. Surg Obes Relat Dis. 2021;17(1):23-33.
  80. Shah AS, Nadeau KJ, Helmrath MA, Inge TH, Xanthakos SA, Kelsey MM. Metabolic outcomes of surgery in youth with type 2 diabetes. Semin Pediatr Surg. 2020;29(1):150893.
  81. Shah AS, Helmrath MA, Inge TH, Xanthakos SA, Kelsey MM, Jenkins T, et al. Study protocol: a prospective controlled clinical trial to assess surgical or medical treatment for paediatric type 2 diabetes (ST(2)OMP). BMJ Open. 2021;11(8):e047766.
  82. Bjornstad P, Hughan K, Kelsey MM, Shah AS, Lynch J, Nehus E, et al. 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(1):187-95.
  83. Michalsky MP, Inge TH, Jenkins TM, Xie C, Courcoulas A, Helmrath M, et al. Cardiovascular Risk Factors After Adolescent Bariatric Surgery. Pediatrics. 2018;141(2).
  84. Messiah SE, Lopez-Mitnik G, Winegar D, Sherif B, Arheart KL, Reichard KW, et al. Changes in weight and co-morbidities among adolescents undergoing bariatric surgery: 1-year results from the Bariatric Outcomes Longitudinal Database. Surg Obes Relat Dis. 2013;9(4):503-13.
  85. Ryder JR, Edwards NM, Gupta R, Khoury J, Jenkins TM, Bout-Tabaku S, et al. Changes in Functional Mobility and Musculoskeletal Pain After Bariatric Surgery in Teens With Severe Obesity: Teen-Longitudinal Assessment of Bariatric Surgery (LABS) Study. JAMA Pediatr. 2016;170(9):871-7.
  86. Bout-Tabaku S, Gupta R, Jenkins TM, Ryder JR, Baughcum AE, Jackson RD, et al. Musculoskeletal Pain, Physical Function, and Quality of Life After Bariatric Surgery. Pediatrics. 2019;144(6).
  87. Burton ET, Mackey ER, Reynolds K, Cadieux A, Gaffka BJ, Shaffer LA. Psychopathology and Adolescent Bariatric Surgery: A Topical Review to Support Psychologists in Assessment and Treatment Considerations. J Clin Psychol Med Settings. 2020;27(2):235-46.
  88. Kalarchian MA, Marcus MD. Psychiatric comorbidity of childhood obesity. Int Rev Psychiatry. 2012;24(3):241-6.
  89. Rankin J, Matthews L, Cobley S, Han A, Sanders R, Wiltshire HD, et al. Psychological consequences of childhood obesity: psychiatric comorbidity and prevention. Adolesc Health Med Ther. 2016;7:125-46.
  90. Zeller MH, Reiter-Purtill J, Jenkins TM, Ratcliff MB. Adolescent suicidal behavior across the excess weight status spectrum. Obesity (Silver Spring, Md). 2013;21(5):1039-45.
  91. He J, Cai Z, Fan X. Prevalence of binge and loss of control eating among children and adolescents with overweight and obesity: An exploratory meta-analysis. Int J Eat Disord. 2017;50(2):91-103.
  92. Goldschmidt AB, Khoury JC, Mitchell JE, Jenkins TM, Bond DS, Zeller MH, et al. Loss of Control Eating and Health Indicators Over 6 Years in Adolescents Undergoing Metabolic and Bariatric Surgery. Obesity (Silver Spring, Md). 2021;29(4):740-7.
  93. Zeller MH, Reiter-Purtill J, Jenkins TM, Kidwell KM, Bensman HE, Mitchell JE, et al. Suicidal thoughts and behaviors in adolescents who underwent bariatric surgery. Surg Obes Relat Dis. 2020;16(4):568-80.
  94. Reiter-Purtill J, Decker KM, Jenkins TM, Zeller MH. Self-worth and developmental outcomes in young adults after pediatric bariatric surgery. Health Psychol. 2023;42(2):92-102.
  95. Järvholm K, Olbers T, Marcus C, Mårild S, Gronowitz E, Friberg P, et al. Short-term psychological outcomes in severely obese adolescents after bariatric surgery. Obesity (Silver Spring, Md). 2012;20(2):318-23.
  96. Zeller MH, Reiter-Purtill J, Ratcliff MB, Inge TH, Noll JG. Two-year trends in psychosocial functioning after adolescent Roux-en-Y gastric bypass. Surg Obes Relat Dis. 2011;7(6):727-32.
  97. Hunsaker SL, Garland BH, Rofey D, Reiter-Purtill J, Mitchell J, Courcoulas A, et al. A Multisite 2-Year Follow Up of Psychopathology Prevalence, Predictors, and Correlates Among Adolescents Who Did or Did Not Undergo Weight Loss Surgery. J Adolesc Health. 2018;63(2):142-50.
  98. White GE, Boles RE, Courcoulas AP, Yanovski SZ, Zeller MH, Jenkins TM, et al. A Prospective Cohort of Alcohol Use and Alcohol-related Problems Before and After Metabolic and Bariatric Surgery in Adolescents. Ann Surg. 2023;278(3):e519-e25.
  99. Hoeltzel GD, Swendiman RA, Tewksbury CM, Parks EP, Williams NN, Dumon KR. How safe is adolescent bariatric surgery? An analysis of short-term outcomes. J Pediatr Surg. 2022;57(8):1654-9.
  100. Varela JE, Hinojosa MW, Nguyen NT. Perioperative outcomes of bariatric surgery in adolescents compared with adults at academic medical centers. Surg Obes Relat Dis. 2007;3(5):537-40; discussion 41-2.
  101. Grant HM, Perez-Caraballo A, Romanelli JR, Tirabassi MV. Metabolic and bariatric surgery is likely safe, but underutilized in adolescents aged 13-17 years. Surg Obes Relat Dis. 2021;17(6):1146-51.
  102. Xanthakos SA, Khoury JC, Inge TH, Jenkins TM, Modi AC, Michalsky MP, et al. Nutritional Risks in Adolescents After Bariatric Surgery. Clin Gastroenterol Hepatol. 2020;18(5):1070-81.e5.
  103. Armstrong-Javors A, Pratt J, Kharasch S. Wernicke Encephalopathy in Adolescents After Bariatric Surgery: Case Report and Review. Pediatrics. 2016;138(6).
  104. Towbin A, Inge TH, Garcia VF, Roehrig HR, Clements RH, Harmon CM, et al. Beriberi after gastric bypass surgery in adolescence. J Pediatr. 2004;145(2):263-7.
  105. Mitchell DM, Singhal V, Animashaun A, Bose A, Carmine B, Stanford FC, et al. Skeletal Effects of Sleeve Gastrectomy in Adolescents and Young Adults: A 2-Year Longitudinal Study. J Clin Endocrinol Metab. 2023;108(4):847-57.
  106. Wasserman H, Jenkins T, Inge T, Ryder J, Michalsky M, Sisley S, et al. Bone mineral density 5 to 11 years after metabolic and bariatric surgery in adolescents with severe obesity compared to peers. Res Sq. 2023.
  107. Modi AC, Zeller MH, Xanthakos SA, Jenkins TM, Inge TH. Adherence to vitamin supplementation following adolescent bariatric surgery. Obesity (Silver Spring, Md). 2013;21(3):E190-5.
  108. Elhag W, El Ansari W. Durability of Cardiometabolic Outcomes Among Adolescents After Sleeve Gastrectomy: First Study with 9-Year Follow-up. Obes Surg. 2021;31(7):2869-77.
  109. Doyle WN, Reinhart N, Reddy NC, Diab AF, Sujka JA, DuCoin CG, et al. Anti-obesity Medication Use for Adolescent Metabolic and Bariatric Surgery Patients: A Systematic Literature Review. Cureus. 2023;15(12):e50905.
  110. Murvelashvili N, Xie L, Schellinger JN, Mathew MS, Marroquin EM, Lingvay I, et al. Effectiveness of semaglutide versus liraglutide for treating post-metabolic and bariatric surgery weight recurrence. Obesity (Silver Spring, Md). 2023;31(5):1280-9.
  111. Ramadan M, Loureiro M, Laughlan K, Caiazzo R, Iannelli A, Brunaud L, et al. Risk of Dumping Syndrome after Sleeve Gastrectomy and Roux-en-Y Gastric Bypass: Early Results of a Multicentre Prospective Study. Gastroenterol Res Pract. 2016;2016:2570237.
  112. Ahmad A, Kornrich DB, Krasner H, Eckardt S, Ahmad Z, Braslow A, et al. Prevalence of Dumping Syndrome After Laparoscopic Sleeve Gastrectomy and Comparison with Laparoscopic Roux-en-Y Gastric Bypass. Obes Surg. 2019;29(5):1506-13.
  113. Laurenius A, Olbers T, Naslund I, Karlsson J. Dumping syndrome following gastric bypass: validation of the dumping symptom rating scale. Obes Surg. 2013;23(6):740-55.
  114. Campos GM, Mazzini GS, Altieri MS, Docimo S, Jr., DeMaria EJ, Rogers AM. ASMBS position statement on the rationale for performance of upper gastrointestinal endoscopy before and after metabolic and bariatric surgery. Surg Obes Relat Dis. 2021;17(5):837-47.
  115. Cozacov Y, Roy M, Moon S, Marin P, Lo Menzo E, Szomstein S, et al. Mid-term results of laparoscopic sleeve gastrectomy and Roux-en-Y gastric bypass in adolescent patients. Obes Surg. 2014;24(5):747-52.
  116. Nijhawan S, Martinez T, Wittgrove AC. Laparoscopic gastric bypass for the adolescent patient: long-term results. Obes Surg. 2012;22(9):1445-9.
  117. Benedix F, Krause T, Adolf D, Wolff S, Lippert H, Manger T, et al. Perioperative Course, Weight Loss and Resolution of Comorbidities After Primary Sleeve Gastrectomy for Morbid Obesity: Are There Differences Between Adolescents and Adults? Obes Surg. 2017;27(9):2388-97.

Platelets, Coagulation, and Antithrombotic Therapy in Diabetes

ABSTRACT

 

Diabetes mellitus is a strong, independent risk factor for the development of atherosclerotic cardiovascular disease (ASCVD) and therefore for atherothrombotic events. Compared to those without diabetes, individuals with diabetes are also at increased risk of cardioembolic stroke in the presence of atrial fibrillation (AF) and of venous thromboembolism. Activation of platelets and the coagulation cascade are the central mechanisms of thrombosis. A range of antiplatelet and anticoagulant drugs are now available. Antithrombotic therapy should be considered in all those with diabetes and established ASCVD or AF. Intensification of antithrombotic therapy is typically indicated during the acute phase of an atherothrombotic event or in those with chronic coronary syndromes who are at high ischemic risk, provided this outweighs bleeding risk. Clinical decisions regarding antithrombotic therapy should be made by assessing an individual’s ischemic and bleeding risks, in consultation with the recipient and reviewed upon any change in circumstances.

 

LIST OF ABBREVIATIONS

 

5HT

5-hydroxytryptamine

ACS

acute coronary syndrome

ADP

adenosine diphosphate

AF

atrial fibrillation

ALI

acute limb ischemia

APT

antiplatelet therapy

ASCVD

atherosclerotic cardiovascular disease

ATP

adenosine triphosphate

ATT

antithrombotic therapy

CAD

coronary artery disease

CCS

chronic coronary syndromes

CI

confidence interval

COX

cyclo-oxygenase

DAPT

dual antiplatelet therapy

DATT

dual antithrombotic therapy

DM

diabetes mellitus

DVT

deep vein thrombosis

GP

glycoprotein

HR

hazard ratio

LEAD

lower extremity artery disease

MACE

major adverse cardiovascular event

MI

myocardial infarction

miR

microribonucleic acid

NOAC

non-vitamin K antagonist oral anticoagulant

OAC

oral anticoagulant

PAD

peripheral artery disease

PAR

protease-activated receptor

PCI

percutaneous coronary intervention

PGI2

prostacyclin

RCTs

randomized controlled trials

RRR

relative risk reduction

SAPT

single antiplatelet therapy

TIMI

thrombolysis in myocardial infarction

TP

thromboprostanoid

TXA2

thromboxane A2

UA

unstable angina

VKA

vitamin K antagonist

vWF

von Willebrand factor

 

INTRODUCTION

 

Despite a century of advances in understanding and management of diabetes mellitus (DM), it continues to increase in prevalence and, furthermore, remains an independent risk factor for atherosclerotic cardiovascular disease (ASCVD), leading to a significant burden of premature mortality and morbidity (1).

 

ASCVD includes a spectrum of clinical syndromes. This can include acute presentations such as acute coronary syndromes (ACS, including myocardial infarction [MI] or unstable angina [UA]), thrombotic stroke, or acute limb ischemia (ALI) (Figure 1). Similarly, ASCVD can lead to chronic conditions such as chronic coronary syndromes (CCS, for example those with stable angina or a history of MI >1 year previously) or chronic lower extremity arterial disease (LEAD) (2).

 

Most acute events in ASCVD are caused by thrombosis. The hemostatic response has an important physiological role in the response to trauma but, if it becomes activated inappropriately, thrombosis can be triggered (3). The clinical effects of thrombosis arise primarily from its location, such as in the coronary arteries leading to acute coronary syndrome (ACS, including myocardial infarction [MI] and unstable angina [UA]), cerebral arteries leading to thrombotic stroke, peripheral arteries leading to acute limb ischemia or deep limb veins leading to deep vein thrombosis (DVT). Alternatively, a thrombus formed at a site can embolize, leading to presentations such as acute pulmonary embolism (typically embolism of a DVT to the pulmonary arteries) or embolic stroke (typically left atrial thrombus to the cerebral arteries) (2,4). In addition to atherosclerotic diseases, individuals with DM who have atrial fibrillation are at higher risk of stroke, secondary to atrial thrombosis and subsequent cardioembolic events (5).

 

There are clear links between pathological processes associated with DM and those responsible for atherogenesis and thrombosis, including inflammation, platelet activation, and coagulation (6,7). Alongside control of glucose levels and optimization of other risk factors, such as dyslipidemia, hypertension, and smoking cessation, antithrombotic therapy (ATT), including antiplatelet therapy (APT) and oral anticoagulation (OAC), has become a key component of the treatment and prevention of atherothrombotic and cardioembolic events. ATT has evolved greatly in the last decades, both in terms of the range of drugs available but also our understanding of how best to deploy them (8).

 

Whilst ATT reduces thrombotic risk, in particular reducing the composite of major adverse cardiovascular events (MACE, typically defined as cardiovascular death, stroke or MI), it also leads to an increased risk of bleeding. Balancing these risks is central to interpretation of clinical trial data and development of treatment recommendations, including in those with DM (9).

 

In this chapter, we will review the underlying pathophysiological mechanisms of thrombosis and the pharmacology of commonly prescribed drugs during ATT. With specific reference to individuals with DM, we will appraise evidence for ATT in a broad range of clinical settings, highlighting current treatment recommendations and particular areas in which more data are needed.

Figure 1. The spectrum of acute cardiovascular events relating to thrombosis and hemostasis in DM.

THE THROMBOTIC RESPONSE AND ITS PHARMACOLOGY

 

As described in Virchow’s triad, prothrombotic changes in the blood flow, constituents and/or vessel wall can trigger thrombosis (10). Broadly, thrombosis involves the activation of platelets and the coagulation cascade (Figure 2). Understanding these processes provides insights into how pharmacological modulation may improve ischemic risk and increase bleeding risk as well as how the individual components of combination ATT interact, including in those with DM.

 

Platelet Activation

 

Platelet activation typically occurs upon endothelial injury and atherosclerotic plaque rupture or erosion, resulting in exposure of blood constituents to prothrombotic substances such as collagen. Collagen exposure leads to platelets adhering to the vessel wall via the glycoprotein (GP) Ia receptor and activation via GPVI (11,12). GPIb forms a complex with clotting factors IX, V and von Willebrand Factor (vWF), strengthening adhesion (13).

 

Platelet activation involves several key processes. Alterations in the cytoskeleton lead to shape change with the formation of filopodia, which increase surface area to volume ratio and may facilitate mechanical adhesion to the vessel wall, other platelets and fibrin strands (14). Platelet activation also involves the release of arachidonic acid from the cell membrane, which is then locally converted to thromboxane A2 (TXA2) by cyclo-oxygenase (COX) 1 and TXA2synthase. TXA2, via the platelet TP-α receptor, contributes further to platelet activation (15). Aspirin (acetylsalicylic acid) irreversibly inhibits COX1, thereby blocking the downstream release of TXA2 for the platelet’s lifespan (around 8-10 days in healthy individuals) as, unlike nucleated cells, platelets cannot regenerate the enzyme (8). Endothelial COX1 and 2 generate the antiplatelet and vasodilatory substance prostacyclin (PGI2). The facts that aspirin is short-lived in the systemic circulation, that platelets are exposed to higher levels of aspirin than endothelium, due to travel through the portal circulation, and that aspirin has relative selectivity for COX1 over COX2 leads to aspirin’s net antiplatelet effect at low doses (16).

 

Platelets also undergo degranulation on activation; a granules contain procoagulant and proinflammatory factors, including platelet P-selectin (also known as CD62P), the surface expression of which is therefore increased. P-selectin mediates platelet-leukocyte aggregation and therefore contributes to an associated inflammatory response (17). Dense granules contain adenosine triphosphate (ATP), adenosine diphosphate (ADP) and 5-hydroxytryptamine (5HT, also known as serotonin). In particular, ADP stimulates platelet activation via P2Y1 and, most significantly, P2Y12 receptors (18,19).

 

Stimulation of the P2Y12 receptor leads to central amplification of the response to a range of agonists and contributes significantly to activation of platelet surface GPIIb/IIIa receptors, the final pathway of platelet aggregation (20). Via vWF and fibrinogen bridges, GPIIb/IIIa mediates platelet-platelet interaction (21).

Figure 2. Pathophysiology of the thrombotic response showing targets for antithrombotic drugs discussed in this chapter. 5HT, 5-hydroxytryptamine (serotonin); AA, arachidonic acid; ADP, adenosine diphosphate; ATP, adenosine triphosphate; Ca2+, calcium; COX1, cyclo-oxygenase 1; GP, glycoprotein; IXa, activated factor IX; P2X1, platelet ATP receptor; P2Y1/P2Y12, platelet ADP receptors; PAR, protease activated receptor; PLA2, phospholipase A2; PSGL1, P-selectin glycoprotein ligand 1; TF, tissue factor; TPα, thromboxane receptor α; TXA2, thromboxane A2; TXA2s, thromboxane A2 synthase; Va, activated factor V; VIIa, activated factor VII; VIIIa, activated factor VIII; VASP, vasodilator-stimulated phosphoprotein; vWF, von Willebrand factor; Xa, activated factor X; XIa, activated factor XI; XIIa, activated factor XII; XIIIa, activated factor XII. Modified from (22).

Several oral platelet P2Y12 receptor antagonists (‘P2Y12 inhibitors’) are currently available (23). Clopidogrel and prasugrel are irreversibly-binding thienopyridines (8). As pro-drugs, they require hepatic metabolism to be activated. In the case of prasugrel this pathway is reliable, whereas there is interindividual variation in the metabolism of clopidogrel meaning around one-third of recipients have poor response when assessed using aggregometry (22). Ticagrelor is a reversibly-binding cyclopentyl-triazolopyrimidine that does not require metabolism to be active. Prasugrel or ticagrelor provide more potent and reliable platelet inhibition compared with clopidogrel (24).

 

Parenterally administered P2Y12 inhibitors have also been developed. Cangrelor is a reversibly-binding ATP analogue that is potent and has rapid onset and offset (25). Selatogrel is a novel, parenterally-active, reversibly-binding P2Y12 inhibitor formulated for subcutaneous administration, but has not yet completed phase III trials and is yet to be marketed (26).

 

Activation of the Coagulation Cascade

 

Although likely an oversimplification of the in vivo state, the coagulation cascade can be summarized as two key pathways made up of factors that converge on a final pathway (27).

 

Loss of endothelium leads to exposure of subendothelial extracellular matrix and contact activation of factor XII, triggering the chain of clotting factor activation known as the intrinsic pathway (28). Tissue factor, expressed on subendothelial cells and released in microparticles from atheromatous plaques, can activate factor IX when in a complex with factor VII: this is the extrinsic pathway (29).

 

Initiation of either pathway can lead to activation of factor X, which associates with activated factor V, calcium (released from damaged tissue) and phospholipids to form the prothrombinase complex (30). Prothrombin (II) is thus broken down to thrombin (IIa), which completes the process through cleavage of fibrinogen to fibrin, the latter being insoluble and forming strands. Tissue factor pathway inhibitor and antithrombin limit this response, but, as recruitment of activated platelets contributes to higher levels of thrombin generation, this endogenous inhibition is quickly overwhelmed (31). Once fibrin is formed, factor XIIIa, activated by thrombin, stabilizes the structure of clot by forming crosslinks between strands and by crosslinking anti-fibrinolytic proteins into the clot (32).

 

Fibrin is lysed by plasmin, a proteolytic enzyme that degrades into variously termed fragments (33). Plasmin is cleaved from its precursor, plasminogen, by tissue plasminogen activator, and is endogenously inhibited by antiplasmin.

 

A number of drugs target the coagulation cascade. During chronic administration, vitamin K antagonists (VKA) such as warfarin reduce the biological activity of prothrombotic vitamin-K-dependent factors (II, VII, IX, X) more than antithrombotic factors (e.g., proteins C and S) (34). Non-vitamin K antagonist oral anticoagulants (NOACs) include the Xa inhibitors apixaban, edoxaban and rivaroxaban and the thrombin inhibitor dabigatran (35).

 

Crosstalk Between Platelets and the Coagulation Cascade

 

Despite the fact that platelets and coagulation are often considered separately when discussing physiology and pharmacology, there is significant crosstalk between the two. Thrombin is generated upon activation of coagulation, and is able to stimulate platelet activation via action on protease-activated receptor (PAR) 1 and, at higher concentrations, PAR4 (36). Conversely platelets can contribute to thrombin generation, increasing coagulability, via scramblase activity that leads to greater surface expression of phosphatidylserine, supporting the assembly of prothrombinase complex on the activated platelet surface, which potentiates thrombin generation (37).

 

SPECIAL PATHOPHYSIOLOGICAL CONSIDERATIONS IN DIABETES

 

DM is an independent risk factor for atherothrombosis and also thrombosis after vascular interventions (38).  Individuals with DM have greater average atherosclerotic plaque burden than those without (39), and onset is at an earlier age (40). There is also some evidence that atherosclerosis in people with DM is more likely to involve distal vessels than those without DM (41). The reasons for this are not completely understood and are likely multifactorial, but a number of relevant pathological processes such as hyperglycemia, chronic inflammation, and oxidative stress are prominent in DM. These contribute to both endothelial injury/dysfunction and increased platelet reactivity, resulting in a prothrombotic milieu (42-44).

 

Platelet activation markers are enhanced in people with DM (45). Effects of hyperglycemia on platelets include increased expression of GPIba, GPIIb/IIIa, and P2Y12, and reduced platelet membrane fluidity (46,47).Hyperglycemia-induced changes in intracellular magnesium and calcium signaling increase sensitivity of platelets to agonists such as ADP, epinephrine and thrombin (48). TXA2 and F2-isoprostane synthesis is increased, the latter via oxidative stress, leading to increased TPa receptor stimulation (49). Reduced sensitivity to PGI2, nitric oxide and insulin, which inhibit platelet activation, also contributes to hyper-reactivity (50,51).

 

Platelet turnover is accelerated in those with DM compared to those without (52). This increased activity in the creation and destruction of circulating platelets means a higher proportion of immature platelets, which are hyper-reactive, are present at any time (53). As well as increasing baseline platelet reactivity, the more frequent appearance of aspirin-naïve platelets in the circulation means more have uninhibited COX1 between doses (54).

 

There is also evidence that DM affects expression of platelet-associated microRNAs (miR-223, miR-26b, miR-126, miR-140), which play a role in the expression in a wide range of genes including those encoding the P2Y12 receptor and P-selectin, though the significance of this remains to be fully established (55,56).

 

As well as platelet activation, DM may affect coagulation and fibrinolysis (57). Changes include increased levels of tissue factor, prothrombin, factor VII and fibrinogen leading to impaired anticoagulant and fibrinolytic activity (58). Increased levels of fibrinogen and its levels of glycation and oxidation lead to more compact, densely-packed fibrin networks and reduced fibrinolysis (59). Hyperglycemia inhibits the fibrinolytic activity of plasminogen through inducing qualitative changes (60). Fibrinolysis is further impaired by elevated levels of plasminogen activator inhibitor 1 and thrombin-activatable fibrinolysis inhibitor as well as incorporation into clot of complement C3 and plasmin inhibitor (59,61).

 

DM also appears to enhance the crosstalk between platelets and clotting factors, leading to tendency to more externalization of phosphatidylserine in the outer platelet membrane, promoting clotting factor assembly and tissue factor activation (62).

 

Finally, individuals with DM frequently have other metabolic conditions such as obesity, dyslipidemia, and increased systemic inflammation. These may interact with diabetes to further enhance platelet reactivity and impair fibrinolysis (59).

 

CURRENT EVIDENCE AND TREATMENT RECOMMENDATIONS FOR ANTITHROMBOTIC THERAPY IN DIABETES

 

The Need for Therapeutic Oral Anticoagulation

 

Broadly, when considering the need for antithrombotic therapy (ATT), including in people with DM, it is helpful to make first a distinction between those with an indication for therapeutic anticoagulation and those without. The most common indication is for prevention of cardioembolic stroke in those with current or previous atrial fibrillation (AF). Individuals with atrial flutter are typically regarded as having similar thrombotic risk to those with AF so similar recommendations are followed (63).

 

DM increases the risk of developing AF by around 40% (64,65). Whilst difficult to completely exclude the effects of confounders such as obesity and hypertension, epidemiological data suggest a causal association between DM and AF, including that poor glycemic control and longer diabetes duration increase AF risk (66). A raised level of HbA1c is also associated with a higher chance of AF recurrence after catheter ablation (67). Hyperglycemia and glycemic fluctuations may contribute to the development of AF though exact mechanisms remain to be determined. Disappointingly, however, there is no clear evidence that intensive glycemic control reduces AF risk, though prospective trials are lacking (66). Treatment with metformin, thiazolidinediones, or dapagliflozin is associated with lower AF risk, suggesting that hypoglycemia avoidance may play a role but adequately designed studies to investigate this possibility are lacking (68-71). AF is often clinically silent and screening with simple pulse checking or using wearable devices should be considered in those over 65 years old (72).

 

Presence of DM is incorporated into the CHA2DS2VASc score used to assess stroke risk when determining whether to recommend oral anticoagulation in people with AF (Table 1 and 2) (73). Long-term oral anticoagulation is strongly recommended in those with AF/atrial flutter and a CHA2DS2VASc score of ³2 (if male) or ³3 (if female), and should be considered when the score is 1 (male) or 2 (female). Individuals with DM, technically defined for the purposes of calculating the score as treatment with oral hypoglycemic drugs and/or insulin or fasting blood glucose >7.0 mmol/L (126 mg/dL), will have a score of at least 1 (males) or 2 (females), therefore OAC should be considered in all people with DM and concurrent AF (63). Bleeding risk should also be considered when weighing the benefits and risks of OAC, but there is no concrete evidence that DM itself increases this, including in those with complications such as retinopathy (74). For people with non-valvular AF (i.e., those without at least moderate mitral valve stenosis or a mechanical valve prothesis), there is now good evidence that, unless contraindicated, a NOAC should be preferred over a VKA, offering better stroke prevention whilst leading to less bleeding, including in individuals with DM (75).

 

Components of the CHA2DS2VASc score are shown in Table 1 and the relation of the score with stroke risk is shown in table 2 (76-78).

 

Table 1. Components of the CHA2DS2VASc Score

Abbreviation

Criterion

Contribution to score

Details

C

Congestive heart failure

1

LVEF £40%

H

Hypertension

1

Includes patients receiving antihypertensive medication

A

Age ³75 years

2

 

D

Diabetes

1

Treatment with oral hypoglycemic drugs and/or insulin or fasting blood glucose >7.0 mmol/L (126 mg/dL)

S

Stroke/TIA/thromboembolism

2

 

V

Vascular disease

1

Atherosclerotic disease e.g., prior MI, PAD or aortic plaque

A

Age 65-74

1

 

Sc

Sex category female

1

 

LVEF, left ventricular ejection fraction; MI, myocardial infarction; PAD, peripheral artery disease; TIA, transient ischemic attack.

 

Table 2. Relation of CHA2DS2VASc Score with Stroke Risk

Total CHA2DS2VASc score

Adjusted stroke risk (% per year)

0

<1

1

1.3

2

2.2

3

3.2

4

4.0

5

6.7

6

9.8

7

9.6

8

6.7

9

15.2

 

When choosing between individual non-vitamin K antagonist oral anticoagulants (NOACs), beyond considering specific drug interactions, there is little evidence to support the use of one agent over another as these have never undergone head-to-head clinical outcome-driven randomized controlled trials (RCTs), although observational data have emerged to provide some insights. In a large retrospective observational study of 434,046 participants with non-valvular AF comparing treatment with apixaban, dabigatran, rivaroxaban and warfarin, apixaban led to a lower risk of stroke against both dabigatran (HR 0.72 [ 95% CI 0.60-0.85]) and rivaroxaban (0.80 [0.73-0.89]), whilst also leading to less bleeding (major bleeding: vs. dabigatran 0.78 [0.70-0.87]; vs. rivaroxaban 0.80 [0.55-0.59]) (79). These findings remain hypothesis-generating, however, and prospective trials would clarify this issue more definitively.

 

Although not discussed in detail in this chapter, OAC may also be indicated for the treatment and prevention of venous thromboembolism. Whilst DM is regarded as a weak risk factor for VTE, beyond this there are no particular considerations relating to DM and usual clinical guidelines as for non-DM individuals should generally be followed (4). Of specific note, however, is that people with DM who are experiencing hyperosmolar states such as ketoacidosis or hyperosmolar hyperglycemic syndrome are at particular risk of VTE. There is ongoing debate around the intensity of anticoagulation that is appropriate for thromboprophylaxis in this group. Consensus is that at least prophylactic doses of low molecular weight heparin, for example, are warranted, with others advocate therapeutic doses (80,81). A robustly-powered clinical outcomes-driven RCT would be welcome to definitively address this issue.

 

Where indications for both anti-platelet therapy (APT) and therapeutic levels of oral anticoagulant therapy (OAC) exist, the general principle is to prioritize continuation of OAC. Co-prescription of APT and OAC should in general be reserved for those with acute coronary syndrome (ACS), recent percutaneous coronary intervention (PCI) or indication for long-term therapy in selected individuals with chronic coronary syndromes (CCS) where ischemic risk is felt to significantly outweigh bleeding risk (22).

 

Treating Acute Atherothrombotic Events

 

ACUTE CORONARY SYNDROMES (ACS)

 

Current guidelines recommend 12 months of dual antiplatelet therapy (DAPT) with aspirin and a P2Y12 inhibitor, including in those with DM, as the default antithrombotic strategy for ACS (72,82-84).

 

There is robust evidence for aspirin therapy in ACS. For example, ISIS-2 demonstrated that aspirin led to an odds reduction in 30-day vascular mortality of 23% in those with acute MI (85). Current recommendations advise a loading dose of around 300 mg followed by maintenance therapy with 75 mg once daily, including in those with DM. However, because of higher platelet turnover in people with DM, 24-hour platelet inhibition is greater with twice-daily compared with once-daily aspirin administration (86-88). Any effects of clinical outcomes are yet to be determined, but are being studied in the ANDAMAN trial that aims to recruit 2573 participants (NCT02520921) and is estimated to finish in December 2023.

 

In ACS, the newer P2Y12 inhibitors prasugrel and ticagrelor are recommended in preference to clopidogrel due to their greater pharmacodynamic and clinical efficacy (83,84). Post-hoc analysis of the TRITON-TIMI trial suggested an impressive benefit of prasugrel over clopidogrel in people with DM (89). Similar findings were noted with regards to ticagrelor over clopidogrel in the PLATO trial, for which post-hoc analysis showing that the absolute benefit of was greatest in individuals with both DM and chronic kidney disease (90).

 

Table 3. Key Double-Blinded Randomized Controlled Trials of Dual Antiplatelet Therapy in Acute Coronary Syndrome, Including in People with Diabetes.

 

Trial

 

n

ACS group included

Group 1

Group 2

Primary efficacy endpoint – whole trial population

Number with DM

Primary efficacy endpoint – DM subgroup

CURE

(91)

12,562

 

NSTE-ACUTE CORONARY SYNDROME

Aspirin + Clopidogrel

Aspirin + Placebo

CV death/MI/stroke:11.4% vs. 9.3%, HR 0.80 [95% CI 0.72-0.90], p<0.001), ARR 2.1%.

2840 (23%)

CV death/MI/stroke:14.2% vs. 16.7%. RR 0.85. ARR 2.5%.

 

CLARITY

(92)

3491

STEMI

Aspirin + Clopidogrel

Aspirin + Placebo

Occluded infarct-related artery/death/recurrent MI: 15.0% vs. 21.7%, odds reduction 36% [95% CI 24-47], p<0.001, ARR 6.7%.

575 (16%)

NR

COMMIT

(93)

45,852

 

STEMI

Aspirin + Clopidogrel

Aspirin + Placebo

Death/reinfarction/stroke: 9.2% vs. 10.1%, OR 0.91 [95% CI 0.86-0.97], p=0.002, ARR 0.9%.

NR

NR

TRITON-THROMBOLYSIS IN MYOCARDIAL INFARCTION 38

(94)

13,608

ACUTE CORONARY SYNDROME with scheduled PCI

Aspirin + Prasugrel

Aspirin + Clopidogrel

CV death/MI/stroke: 9.9% vs. 12.1%, HR 0.81 [95% CI 0.73-0.90], p<0.001, ARR 2.2%.

3146 (23%)

CV death/MI/stroke: 12.2% vs. 17.0%, HR 0.70, ARR 4.8%.

TRILOGY ACUTE CORONARY SYNDROME

(95)

7243

NSTE-ACUTE CORONARY SYNDROME

with medical management

Aspirin + Prasugrel

Aspirin + Clopidogrel

CV death/MI/stroke: 13.9% vs. 16.0%, HR 0.91 [95% CI 0.79-1.05], p=0.21, ARR 2.1%.

2811 (39%)

CV death/MI/stroke: 17.8% vs. 20.4%, HR 0.90 [95% CI 0.73 to 1.09]), ARR=2.6%, interaction-p for DM status 0.71, ARR 2.6%.

PLATO

(96)

18,624

All ACUTE CORONARY SYNDROME (STEMI

patients included only if for PPCI)

Aspirin + Ticagrelor

Aspirin + Clopidogrel

CV death/MI/stroke: 9.8% vs. 11.7%, HR 0.84 [95% CI 0.77-0.92], p<0.001, ARR 1.9%.

 

 

4662 (25%)

CV death/MI/stroke: 14.1% vs. 16.2, HR 0.88 [95% CI 0.76-1.03], interaction-p for DM status 0.49, ARR 2.1%.

ACS, acute coronary syndrome; ARR, absolute risk reduction; CV, cardiovascular; DM, diabetes mellitus; HR, hazard ratio; MI, myocardial infarction; NR, not reported; NSTE-ACS, non-ST elevation ACS; OR, odds ratio; PCI, percutaneous coronary intervention; PPCI, primary PCI; PPM, permanent pacemaker; RR, relative risk; STEMI, ST elevation MI; NR, not recorded

 

The recent ISAR-REACT-5 study demonstrated superiority of a prasugrel-based strategy over a ticagrelor-based strategy in reducing cardiovascular events in ACS patients but was an open-label trial with limited power (97,98). Furthermore, data from the pre-specified subgroup with DM suggested there was no difference between the drugs (99).

 

Early de-escalation from dual antiplatelet therapy (DAPT) to ticagrelor monotherapy after PCI, including for ACS, has recently been trialed as an alternative strategy. In the TWILIGHT study, de-escalation from aspirin and ticagrelor to ticagrelor monotherapy at 3 months after PCI for ACS or stable coronary artery disease (CAD) was compared with continued DAPT in 7,119 participants (100). De-escalating to ticagrelor monotherapy led to a lower incidence at 12 months of the primary end point of Bleeding Academic Research Consortium type 2, 3, or 5 bleeding compared with DAPT (4.0% vs 7.1%, HR 0.56 [95% 0.45-0.68], p<0.001). This finding appeared similar regardless of DM status. There was no evidence of an increase in the secondary combined endpoint of death, MI or stroke. Conversely, 1 month of DAPT followed by ticagrelor alone for 23 months was not superior to 12 months of standard DAPT followed by 12 months of aspirin alone in reducing the primary endpoint of all-cause mortality or new Q-wave MI following PCI in the GLOBAL LEADERS trial, in which 47% of participants had ACS (101). Antiplatelet strategy had no significant effect on BARC type 3 or 5 bleeding in those with and without DM (102). Currently, de-escalation of DAPT may be an option for individuals with high bleeding risk and relatively low risk of vascular re-occlusion but guidelines are yet to recommend more widespread adoption.

 

In summary, following ACS in individuals with diabetes, DAPT for 12 months with aspirin and prasugrel or aspirin and ticagrelor is recommended by the majority of guidelines/experts and early de-escalation should be reserved to those at high bleeding risk. Longer term DAPT should be considered in those at high thrombosis/low bleeding risk, which is further detailed below. 

 

ACUTE ISCHEMIC STROKE

 

If no contraindications exist, the first-line treatment for significant acute ischemic stroke is thrombolysis with an intravenous tissue plasminogen activator, or percutaneous mechanical thrombectomy (103). Antiplatelet therapy (APT), typically aspirin monotherapy, is then administered from 24 hours later (104,105).

 

In those with minor stroke (National Institutes of Health Stroke Score <3), high-risk transient ischemic attack (TIA) (Age, blood pressure, clinical feature, duration and presence of diabetes score>4) or TIA not requiring thrombolysis or thrombectomy, APT can be initiated as soon as hemorrhagic stroke is excluded. The current regimen of choice may be dual antiplatelet therapy (DAPT) with aspirin 75-100 mg once daily and clopidogrel 75 mg once daily, based on findings from the CHANCE and POINT trials (106,107). After 21 days, DAPT should be de-escalated to clopidogrel monotherapy (105).

 

Both ticagrelor monotherapy and aspirin plus ticagrelor have also been compared to aspirin alone after acute non-severe ischemic stroke or high-risk TIA. The SOCRATES trial narrowly failed to demonstrate statistically-significant difference in the primary endpoint of stroke, MI or death (6.7% vs. 7.5%, HR 0.89 [95% CI 0.78-1.01], p=0.07) between participants receiving ticagrelor vs. aspirin (108). However, exploratory analysis suggested those who received both aspirin and ticagrelor in the peri-event period appeared to gain more benefit compared to individuals not having aspirin pre-randomization (HR 0.76 [95% CI 0.61-0.95], p=0.02; vs. 0.96 [0.82-1.12]). This was explored further in the THALES trial, which demonstrated a significant reduction in the primary composite endpoint of stroke or death at 30 days (5.5% vs. 6.6%, HR 0.83 [95% CI 0.71-0.96, p=0.02) when receiving aspirin plus ticagrelor compared to aspirin alone, but at the expense of more frequent severe bleeding (0.5% vs. 0.1%, HR 3.99 [95% CI 1.74-9.14], p=0.001), defined using the Global Utilization of Streptokinase and Tissue Plasminogen Activator for Occluded Coronary Arteries trial criteria (109). Findings from SOCRATES appeared similar in the subgroups with and without DM, whereas in THALES there was less signal of benefit of DAPT in those with DM vs. those without (HR 0.93 [95% CI 0.72-1.20] vs. 0.78 [0.64-0.94]).

 

In summary, following major stroke requiring thrombolysis or thrombectomy, aspirin monotherapy should be administered 24 hours later. In minor stroke or high-risk TIA, DAPT should be initiated as soon as intracerebral bleeding is ruled out and continued for 21 days with aspirin then withdrawn and individuals treated with long-term clopidogrel monotherapy.

 

Preventing Atherothrombotic Events in Individuals with Diabetes and Established Cardiovascular Disease

 

CORONARY ARTERY DISEASE

 

In those with established CAD, even without an ACS event in the last 12 months, the benefits of antiplatelet therapy (APT) are well-established. Robust evidence for vs. against use of APT in patients with ASCVD, including CAD comes, for example, from the Antithrombotic Trialists Collaboration, who performed a meta-analysis including 135,000 individuals (110). This demonstrated clear benefit, mainly with aspirin as single-antiplatelet therapy (SAPT), in reducing MACE by around a quarter (110).  The incidence of diabetes in these studies, many of which are now several decades old, was relatively low, however.

 

There is evidence from trials with both pharmacodynamic and clinical outcomes that increasing daily aspirin dose beyond 75-100 mg in patients with DM leads to neither greater platelet inhibition nor improved outcomes (111,112).

 

Daily doses of aspirin in the range 75-100 mg and no higher are recommended for use as APT. Recent data on clinical outcomes relating to aspirin dosing comes from the ADAPTABLE trial, in which the regimens 81 mg OD and 325 mg OD were compared in 15,076 patients with ASCVD (113). After a median of 26 months, there was no significant difference in the rates of a composite primary endpoint of all-cause death, hospitalization for myocardial infarction or hospitalization for stroke (7.28% [81 mg] vs. 7.51% [325 mg], HR 1.02, 95% CI 0.91-1.14; p=0.75). Furthermore, this finding appeared replicated in the subgroup (n=5676) with diabetes (HR 0.99 [0.84-1.17]). This is supported by pharmacodynamic data showing that, whilst individuals with DM have reduced response to aspirin 75 mg once daily compared with healthy controls, increasing the dose to 300 mg does not alter the response (111).

 

In the CAPRIE study, clopidogrel 75 mg once daily was compared with aspirin 325 mg once daily (114). There was a slightly lower rate of MI, ischemic stroke or CV death with clopidogrel (5.32% vs. 5.83%, RRR 8.7% [95% CI 0.3-16.5], p=0.043) as well as less gastrointestinal bleeding. A fifth of participants in CAPRIE had diabetes and a retrospective subgroup analysis suggested an amplified benefit of clopidogrel over aspirin compared to those without diabetes. Clopidogrel monotherapy is currently recommended in those people with chronic coronary syndromes (CCS) who are unable to take aspirin, or, based on pre-specified subgroup analyses of CAPRIE suggesting particular benefit, as a first-line agent in those with either concurrent CAD and cerebrovascular disease or PAD.

 

Beyond single antiplatelet therapy (SAPT), there is good evidence for intensification of antithrombotic therapy in select people with CAD who are at high risk of ischemic events but without high risk of bleeding. The Clopidogrel for High Atherothrombotic Risk and Ischemic Stabilization, Management, and Avoidance (CHARISMA) study randomized 19,185 stable aspirin-treated individuals with established atherothrombotic disease or multiple risk factors to receive clopidogrel 75 mg once daily or placebo (115). Though the point estimate of the hazard ratio was below 1, there was no significant reduction in the primary efficacy endpoint of MACE when receiving dual antithrombotic therapy (DAPT) vs. aspirin alone (HR 0.93, [95% CI 0.83-1.05], p=0.22). However, in the subgroup with prior MI, prior stroke or PAD, there was some evidence of benefit (0.77 [0.61-0.98], p=0.031) (116). Around 30% of the participants in CHARISMA had DM and there was in fact a trend towards less benefit of DAPT over SAPT in this group compared to those without DM.

 

The DAPT study similarly showed that 30 vs. 12 months of clopidogrel (65%) or prasugrel (35%) given to aspirin-treated individuals undergoing PCI significantly reduced death, MI or stroke in those with prior MI (HR 0.56 [95% CI 0.42-0.76], p<0.001), but not those without (0.83 [0.68-1.02], p=0.08) (117). Like CAPRIE, there was some evidence that those in the trial with DM gained less benefit in reduction of MACE from continued thienopyridine vs. placebo, when compared to those without DM (6.6% vs. 7.0% in those with DM, p=0.55; 3.3% vs. 5.2% in those without, p<0.001; interaction-p=0.03). Conversely, DM did not appear to be an interacting factor with regards to stent thrombosis or bleeding.

 

There is perhaps more convincing evidence, particularly in those with DM, for use of long-term ticagrelor-based DAPT. In the PEGASUS-TIMI 54 study, DAPT with aspirin plus ticagrelor, either 60 mg or 90 mg twice-daily, reduced MACE vs. aspirin alone (e.g. 60 mg twice-daily vs. placebo: HR 0.84 [95% CI 0.74-0.95], p=0.008) in participants with prior MI (>1 year ago) and an additional risk factor (age ≥65 years, DM, recurrent MI, multivessel CAD or non-end stage CKD) (118). Thrombolysis In Myocardial Infarction (TIMI)-major bleeding was significantly more frequent in ticagrelor-treated individuals, but serious events such as intracranial hemorrhage, hemorrhagic stroke or fatal bleeding showed no increase. In contrast to the thienopyridine trials, the 6806 participants with diabetes demonstrated a significant benefit of DAPT over SAPT in reducing MACE (HR 0.84 [95% CI 0.72-0.99], p=0.035) with a greater absolute risk reduction than in the cohort without diabetes (1.5% vs. 1.1%) (119). Patients without a history of anemia or hospitalization for bleeding, important risk factors for bleeding, appeared to derive greater benefit from long-term DAPT (120).

 

As well as in those with prior MI, ticagrelor-based DAPT has also been tested against aspirin alone in people with type 2 DM and chronic coronary syndromes (CCS) but without prior MI. THEMIS included 19,220 participants randomized to receive ticagrelor (90 mg twice daily, reduced to 60 mg during the trial) or placebo, on a background of aspirin treatment (121). After an average follow-up of 40 months, there was a lower incidence of MACE in those receiving ticagrelor when compared to placebo (HR 0.90 [95% CI 0.81-0.99], p=0.04).  Notably, however, there was a relatively greater increase in TIMI-major bleeding (2.32 [1.82-2.94], p<0.001). Whilst meeting its primary endpoint, the net clinical benefit has not supported adoption in European practice, although subgroup analysis has suggested this may have been more favorable in those patients with prior PCI (122). Furthermore, based on the THEMIS data, the US Food and Drug Administration has recently extended the licensed indication for ticagrelor to include the prevention of a first MI or stroke in people with CCS at high risk of MI or stroke, including in those with DM (123).

 

An alternative to long-term DAPT is low-dose dual antithrombotic therapy (DATT) with aspirin 75-100 mg once daily and rivaroxaban 2.5 mg twice daily.  The COMPASS trial included randomization of 27,395 participants with prior MI or multivessel CAD (38% with DM) or PAD to receive either low-dose DATT, rivaroxaban 5 mg twice daily alone or aspirin alone (124). Compared to aspirin alone, low-dose DATT led to a significantly reduced incidence of MACE [4.1% vs 5.4%, HR 0.76 [95% CI 0.66-0.86], p<0.001], people with DM gaining an even greater absolute net benefit.

 

Current guidelines recommend long-term DAPT or low-dose dual antithrombotic therapy (DATT) in those individuals with CCS without an indication for therapeutic oral anticoagulant (OAC) who are at high ischemic risk but not high bleeding risk (22).

 

In those undergoing PCI for stable CAD, including in those individuals with DM, the standard DATT regimen is DAPT with aspirin and clopidogrel for 6 months (125).

 

In summary, individuals with DM who have CCS should be treated with at least one antiplatelet agent, usually aspirin, although clopidogrel can be used if aspirin is contraindicated. However, more recent evidence indicates that those with a previous MI benefit from long-term DAPT (aspirin and ticagrelor) or a combination of antiplatelet and anticoagulant (DATT with aspirin and rivaroxaban) provided they have a low bleeding risk. Individuals with significant CAD but without a previous MI may also benefit from DAPT or DATT, which is best reserved for people with high vascular risk but low bleeding risk. 

 

CEREBROVASCULAR DISEASE  

 

There is good evidence for use of APT with aspirin, clopidogrel, ticlopidine or aspirin and dipyridamole in combination for secondary prevention in people with cerebrovascular disease, including those who also have DM (126). Aspirin plus dipyridamole offers better long-term protection than aspirin alone, but has a frequent adverse effect of headache that can limit its use (127). Clopidogrel monotherapy, without this side effect, offers similar levels of secondary prevention to aspirin plus dipyridamole and is the current preferred agent. In the first 3 months after an ischemic stroke, if reperfusion therapy has been given, aspirin alone is typically prescribed. In cases where reperfusion therapy has not been given, there is good evidence for using either aspirin and clopidogrel or aspirin and ticagrelor over aspirin alone (128,129).  After 3 months, typically clopidogrel monotherapy is then given long-term, though aspirin and dipyridamole or aspirin alone are used instead at some centers (127,130,131).

 

PERIPHERAL ARTERY DISEASE

 

The effectiveness of APT for secondary prevention of ASCVD, including in those with symptomatic PAD, was established by the Antithrombotic Trialists’ Collaboration as discussed above. Similarly, in the CAPRIE trial, P2Y12inhibitor monotherapy with clopidogrel was compared with aspirin, including in people with PAD (114). Whilst in the overall trial population there was only a modest reduction in MACE, there was evidence of greater efficacy in the subgroup with PAD, meaning clopidogrel may be preferred to aspirin. Current ESC guidelines recommend either aspirin or clopidogrel for patients with symptomatic PAD and/or those who have required revascularization, including in individuals with DM (132).

 

In those with symptomatic PAD, ticagrelor monotherapy has also been compared with clopidogrel in the EUCLID trial (133). There was no significant difference in the primary composite endpoint of MACE during a median follow-up period of 30 months and therefore ticagrelor monotherapy is not licensed for use in PAD. Prasugrel monotherapy has not been well tested in clinical-outcome studies but may offer pharmacodynamic advantages over clopidogrel, including in individuals with DM (134).

 

Comparison of DAPT (aspirin plus clopidogrel) with aspirin alone in people with PAD was included in CHARISMA (n=3,096 with PAD, 36.2% with DM). There was no significant difference in MACE (7.6% vs 8.9%, HR 0.85 [0.66–1.08], p=0.18) (135).

 

Conversely, there is good evidence for intensification of aspirin monotherapy to low-dose DATT with aspirin 75-100 mg once daily and rivaroxaban 2.5 mg twice daily in people with PAD, supported by the analysis of 7,470 participants with PAD in the COMPASS trial (136). The combination of rivaroxaban and aspirin reduced incidence of MACE over a median follow up of 21 months versus aspirin alone [5.1% vs 6.9%, HR 0.72 (0.57-0.90); p=0.0047]. Particularly important benefits observed included a lower incidence of major adverse limb events [1% vs 2%, HR 0·63 [95% CI 0.41–0.96], p=0·032], and lower incidence of major amputation [0.30 [0.11–0.80], p=0.011].

 

Subsequently, the evidence base for low-dose DATT in people PAD has been enhanced by the results of the VOYAGER-PAD trial, which randomized 6564 individuals with PAD treated by revascularization to receive either low-dose DATT or aspirin alone (137). After a median follow-up of 28 months (interquartile range 22-34), the primary composite endpoint of acute limb ischemia, amputation, MI, ischemic stroke or CV death occurred in 17.3% vs. 19.9% (HR 0.85 [0.76-0.96], p=0.009) without a significant increase in the incidence of TIMI major bleeding (2.65% vs. 1.87%, HR 1.43 [0.97-2.10, p=0.07). Forty percent of the trial population had DM with a similar response observed in this group.

 

It should be noted that DM individuals with symptomatic PAD are likely to have extensive vascular pathology and therefore DATT is likely to offer benefit in more than one vascular bed. Discussion of antithrombotic therapy for those people with DM and asymptomatic PAD is included in the next section.

 

Preventing First Atherothrombotic Event in Patients with Diabetes and No Symptomatic Atherosclerotic Cardiovascular Disease

 

It is rational to hypothesize that antithrombotic therapy (ATT) therapy may reduce the chance of a first atherothrombotic event or limit its severity by preventing thrombosis or reducing its impact.  ATT in several distinct groups with DM but without symptomatic ASCVD have been investigated in a number of trials. The largest individual-level meta-analysis was performed in 2009 and included 95,000 participants from 6 trials (138). In individuals with DM, though aspirin led to a 12% proportional reduction in the rate of serious vascular events, this did not reach statistical significance. However, the point estimate was consistent with the statistically significant benefit of aspirin in the non-DM population and the DM population showed an identical trend. Three further trials have been added to the literature since this meta-analysis was performed. Two, JPAD (n=2539) and POPADAD (n=1276) were not adequately powered to draw firm conclusions (139,140). However, most recently ASCEND provided data from 15,480 individuals with DM but without symptomatic ASCVD who were randomized to receive aspirin 100 mg once daily or placebo (141). After a mean follow up of 7.4 years, those randomized to aspirin had a significantly reduced rate of serious vascular events (MI, stroke or TIA, or vascular death excluding intracranial hemorrhage) (RR 0.88 [95 % CI 0.79-0.97], p=0.01). However, major bleeding was significantly more frequent when receiving aspirin (1.24 [1.09-1.52], p=0.003), the majority being gastrointestinal. The investigators concluded that the absolute benefits were largely counterbalanced by the risks, despite a favorable, albeit modest, risk-benefit ratio.

 

Antiplatelet drugs other than aspirin have not been widely studied for primary prevention in individuals with DM and this remains an area for future research.

 

CONCLUSIONS

 

DM leads to a prothrombotic milieu that increases the risk of atherothrombotic and thromboembolic events compared to the non-DM population. Changes in platelets, coagulation, and inflammation appear central to this increased risk. Antithrombotic therapy (ATT) can help treat or prevent thrombotic events but increases bleeding risk. In those with a history of symptomatic ASCVD, long-term antiplatelet therapy (APT) with aspirin or clopidogrel is indicated. Intensification to long-term dual antiplatelet therapy (DAPT) or low-dose dual antithrombotic therapy (DATT) should be considered in those with chronic coronary syndromes (CCS) who have high ischemic risk but not high bleeding risk. Low-dose DATT can also be beneficial to people with symptomatic PAD. Therapeutic levels of oral anticoagulant (OAC) should be considered in all individuals with DM who develop AF. Accurately assessing and balancing a patient’s risk of ischemic and bleeding events is key to making rational treatment recommendations for ATT in DM (Figure 3).

 

Looking to the future, further work to determine more precisely an individual’s thrombotic and bleeding risk would greatly enhance our ability to make the best treatment recommendations for patients with DM. Whether this is achieved by more complex statistical modelling, novel imaging techniques, and/or better appreciation of circulating biomarkers remains to be determined. This would allow a greater move towards personalized strategies in order to more appropriately balance the benefits and risks of ATT. People with DM often have complex co-morbidities meaning choosing the best regimen is difficult, but is at the same time crucial to ensure an optimal outcome.

 

Emerging strategies such as early de-escalation of DAPT are encouraging new tools giving more options for subtle adjustment of ATT intensity, but require definitive proof they lead to no significant ischemic penalty and ratification by guideline committees before wider adoption can be recommended. No doubt further clarity will follow in the coming years.

 

The lack of an ability of ATT to meaningfully improve net clinical outcomes in those with DM without established ASCVD is a source of disappointment and demands future attention. Trials have focused on aspirin but it is clear that people with DM may have a poor response (111). As well as trials exploring novel regimens of aspirin, trials testing P2Y12 inhibitor monotherapy, which may offer pharmacodynamic advantages over aspirin in this group, are warranted (134).

 

Finally, targeting the pathological abnormalities that cause hypofibrinolysis in diabetes, such as inhibition of PAI-1 activity, may offer an alternative management strategy to further reduce vascular occlusive disease in diabetes, while keeping the risk of bleeding to a minimum.

Figure 3. Principles to consider when deciding on the optimal regimen of antithrombotic therapy in a person with diabetes. ACS, acute coronary syndrome; AF, atrial fibrillation; ASCVD, atherosclerotic cardiovascular disease; CAD, coronary artery disease; CI, contraindication; DAPT, dual antiplatelet therapy; DATT, dual antithrombotic therapy; DM, diabetes mellitus; eGFR, estimated glomerular filtration rate; GI, gastrointestinal; OAC, oral anticoagulation; PAD, peripheral artery disease; PCI, percutaneous coronary intervention.

REFERENCES

 

  1. Sarwar N, Gao P, Seshasai SR, Gobin R, Kaptoge S, Di Angelantonio E, Ingelsson E, Lawlor DA, Selvin E, Stampfer M, Stehouwer CD, Lewington S, Pennells L, Thompson A, Sattar N, White IR, Ray KK, Danesh J. Diabetes mellitus, fasting blood glucose concentration, and risk of vascular disease: a collaborative meta-analysis of 102 prospective studies. Lancet. 2010;375(9733):2215-2222.
  2. Parker WAE, Gorog DA, Geisler T, Vilahur G, Sibbing D, Rocca B, Storey RF. Prevention of stroke in patients with chronic coronary syndromes or peripheral arterial disease. Eur Heart J Suppl. 2020;22(Supplement_M):M26-M34.
  3. Jackson SP. Arterial thrombosis--insidious, unpredictable and deadly. Nat Med. 2011;17(11):1423-1436.
  4. Konstantinides SV, Meyer G, Becattini C, Bueno H, Geersing G-J, Harjola V-P, Huisman MV, Humbert M, Jennings CS, Jiménez D, Kucher N, Lang IM, Lankeit M, Lorusso R, Mazzolai L, Meneveau N, Ní Áinle F, Prandoni P, Pruszczyk P, Righini M, Torbicki A, Van Belle E, Zamorano JL, ESC Scientific Document Group. 2019 ESC Guidelines for the diagnosis and management of acute pulmonary embolism developed in collaboration with the European Respiratory Society (ERS): The Task Force for the diagnosis and management of acute pulmonary embolism of the European Society of Cardiology (ESC). Eur Heart J. 2020;41(4):543-603.
  5. Gage BF, Waterman AD, Shannon W, Boechler M, Rich MW, Radford MJ. Validation of clinical classification schemes for predicting stroke: results from the National Registry of Atrial Fibrillation. JAMA. 2001;285(22):2864-2870.
  6. Libby P, Buring JE, Badimon L, Hansson GK, Deanfield J, Bittencourt MS, Tokgozoglu L, Lewis EF. Atherosclerosis. Nat Rev Dis Primers. 2019;5(1):56.
  7. Biondi-Zoccai GGL, Abbate A, Liuzzo G, Biasucci LM. Atherothrombosis, inflammation, and diabetes. J Am Coll Cardiol. 2003;41(7):1071-1077.
  8. Patrono C, Morais J, Baigent C, Collet JP, Fitzgerald D, Halvorsen S, Rocca B, Siegbahn A, Storey RF, Vilahur G. Antiplatelet agents for the treatment and prevention of coronary atherothrombosis. J Am Coll Cardiol. 2017;70(14):1760-1776.
  9. Storey RF. The long journey of individualizing antiplatelet therapy after acute coronary syndromes. Eur Heart J. 2020;41:3546-3548.
  10. Bagot CN, Arya R. Virchow and his triad: a question of attribution. Br J Haematol. 2008;143(2):180-190.
  11. Moroi M, Jung SM, Okuma M, Shinmyozu K. A patient with platelets deficient in glycoprotein VI that lack both collagen-induced aggregation and adhesion. J Clin Invest. 1989;84(5):1440-1445.
  12. Santoro SA, Rajpara SM, Staatz WD, Woods VL, Jr. Isolation and characterization of a platelet surface collagen binding complex related to VLA-2. Biochem Biophys Res Commun. 1988;153(1):217-223.
  13. Handa M, Titani K, Holland LZ, Roberts JR, Ruggeri ZM. The von Willebrand factor-binding domain of platelet membrane glycoprotein Ib. Characterization by monoclonal antibodies and partial amino acid sequence analysis of proteolytic fragments. J Biol Chem. 1986;261(27):12579-12585.
  14. Aslan JE, Itakura A, Gertz JM, McCarty OJ. Platelet shape change and spreading. Methods Mol Biol. 2012;788:91-100.
  15. Patrono C. Aspirin as an antiplatelet drug. N Engl J Med. 1994;330(18):1287-1294.
  16. Parker WAE, Orme RC, Hanson J, Stokes HM, Bridge CM, Shaw PA, Sumaya W, Thorneycroft K, Petrucci G, Porro B, Judge HM, Ajjan RA, Rocca B, Storey RF. Very-low-dose twice-daily aspirin maintains platelet inhibition and improves haemostasis during dual-antiplatelet therapy for acute coronary syndrome. Platelets. 2019;30(2):148-157.
  17. Harrison P, Cramer EM. Platelet alpha-granules. Blood Rev. 1993;7:52-62.
  18. Fagura MS, Dainty IA, McKay GD, Kirk IP, Humphries RG, Robertson MJ, Dougall IG, Leff P. P2Y1-receptors in human platelets which are pharmacologically distinct from P2Y(ADP)-receptors. Br J Pharmacol. 1998;124(1):157-164.
  19. Mahaut-Smith MP, Jones S, Evans RJ. The P2X1 receptor and platelet function. Purinergic Signal. 2011;7(3):341-356.
  20. Storey RF, Sanderson HM, White AE, May JA, Cameron KE, Heptinstall S. The central role of the P(2T) receptor in amplification of human platelet activation, aggregation, secretion and procoagulant activity. Br J Haematol. 2000;110(4):925-934.
  21. Pytela R, Pierschbacher MD, Ginsberg MH, Plow EF, Ruoslahti E. Platelet membrane glycoprotein IIb/IIIa: member of a family of Arg-Gly-Asp--specific adhesion receptors. Science. 1986;231(4745):1559-1562.
  22. Parker WAE, Storey RF. Antithrombotic therapy for patients with chronic coronary syndromes. Heart 2021;107:925-933.
  23. Parker WAE, Storey RF. Novel approaches to P2Y12 inhibition and aspirin dosing. Platelets. 2020;32(1):7-14.
  24. Joshi RR, Hossain R, Morton AC, Ecob R, Judge HM, Wales C, Walker JV, Karunakaran A, Storey RF. Evolving pattern of platelet P2Y12 inhibition in patients with acute coronary syndromes. Platelets. 2014;25(6):416-422.
  25. Parker WA, Bhatt DL, Prats J, Day JRS, Steg PG, Stone GW, Hamm CW, Mahaffey KW, Price MJ, Gibson CM, White HD, Storey RF. Characteristics of dyspnoea and associated clinical outcomes in the CHAMPION PHOENIX study. Thromb Haemost. 2017;117(6):1093-1100.
  26. Parker W, Storey R. Pharmacology and Potential Role of Selatogrel, a Subcutaneous Platelet P2Y 12 Receptor Antagonist. Expert Opin Emerg Drugs. 2020;25(1):1-6.
  27. Ajjan R, Grant PJ. Coagulation and atherothrombotic disease. Atherosclerosis. 2006;186(2):240-259.
  28. Renné T, Schmaier AH, Nickel KF, Blombäck M, Maas C. In vivo roles of factor XII. Blood. 2012;120(22):4296-4303.
  29. Mackman N, Tilley RE, Key NS. Role of the extrinsic pathway of blood coagulation in hemostasis and thrombosis. Arterioscler Thromb Vasc Biol. 2007;27(8):1687-1693.
  30. Krishnaswamy S, Jones KC, Mann KG. Prothrombinase complex assembly. Kinetic mechanism of enzyme assembly on phospholipid vesicles. J Biol Chem. 1988;263(8):3823-3834.
  31. Crawley JT, Lane DA. The haemostatic role of tissue factor pathway inhibitor. Arterioscler Thromb Vasc Biol. 2008;28(2):233-242.
  32. Ariens RA, Lai TS, Weisel JW, Greenberg CS, Grant PJ. Role of factor XIII in fibrin clot formation and effects of genetic polymorphisms. Blood. 2002;100(3):743-754.
  33. Verstraete M. The fibrinolytic system: from petri dishes to genetic engineering. Thromb Haemost. 1995;74:25-35.
  34. Hurlen M, Abdelnoor M, Smith P, Erikssen J, Arnesen H. Warfarin, aspirin, or both after myocardial infarction. N Engl J Med. 2002;347(13):969-974.
  35. Yeh CH, Hogg K, Weitz JI. Overview of the new oral anticoagulants: opportunities and challenges. Arterioscler Thromb Vasc Biol. 2015;35(5):1056-1065.
  36. Kahn ML, Nakanishi-Matsui M, Shapiro MJ, Ishihara H, Coughlin SR. Protease-activated receptors 1 and 4 mediate activation of human platelets by thrombin. J Clin Invest. 1999;103(6):879-887.
  37. Monroe DM, Hoffman M, Roberts HR. Platelets and thrombin generation. Arterioscler Thromb Vasc Biol. 2002;22(9):1381-1389.
  38. Mi Y, Yan S, Lu Y, Liang Y, Li C. Venous thromboembolism has the same risk factors as atherosclerosis: A PRISMA-compliant systemic review and meta-analysis. Medicine (Baltimore). 2016;95(32):e4495.
  39. Ibebuogu UN, Nasir K, Gopal A, Ahmadi N, Mao SS, Young E, Honoris L, Nuguri VK, Lee RS, Usman N, Rostami B, Pal R, Flores F, Budoff MJ. Comparison of atherosclerotic plaque burden and composition between diabetic and non diabetic patients by non invasive CT angiography. Int J Cardiovasc Imaging. 2009;25(7):717-723.
  40. Larsen JR, Tsunoda T, Tuzcu EM, Schoenhagen P, Brekke M, Arnesen H, Hanssen KF, Nissen SE, Dahl-Jorgensen K. Intracoronary ultrasound examinations reveal significantly more advanced coronary atherosclerosis in people with type 1 diabetes than in age- and sex-matched non-diabetic controls. Diab Vasc Dis Res. 2007;4(1):62-65.
  41. Lowry D, Saeed M, Narendran P, Tiwari A. A review of distribution of atherosclerosis in the lower limb arteries of patients with diabetes mellitus and peripheral vascular disease. Vasc Endovascular Surg. 2018;52(7):535-542.
  42. Carrizzo A, Izzo C, Oliveti M, Alfano A, Virtuoso N, Capunzo M, Di Pietro P, Calabrese M, De Simone E, Sciarretta S, Frati G, Migliarino S, Damato A, Ambrosio M, De Caro F, Vecchione C. The main determinants of diabetes mellitus vascular complications: endothelial dysfunction and platelet hyperaggregation. Int J Mol Sci. 2018;19(10).
  43. Stentz FB, Umpierrez GE, Cuervo R, Kitabchi AE. Proinflammatory cytokines, markers of cardiovascular risks, oxidative stress, and lipid peroxidation in patients with hyperglycemic crises. Diabetes. 2004;53(8):2079-2086.
  44. Chaudhuri A, Umpierrez GE. Oxidative stress and inflammation in hyperglycemic crises and resolution with insulin: implications for the acute and chronic complications of hyperglycemia. J Diabetes Complications. 2012;26(4):257-258.
  45. Patrono C, Rocca B. Measurement of thromboxane biosynthesis in health and disease. Front Pharmacol. 2019;10:1244.
  46. Soma P, Swanepoel AC, du Plooy JN, Mqoco T, Pretorius E. Flow cytometric analysis of platelets type 2 diabetes mellitus reveals 'angry' platelets. Cardiovasc Diabetol. 2016;15:52.
  47. Watala C, Boncler M, Golański J, Koziołkiewcz W, Trojanowski Z, Walkowiak B. Platelet membrane lipid fluidity and intraplatelet calcium mobilization in type 2 diabetes mellitus. Eur J Haematol. 1998;61(5):319-326.
  48. Gawaz M, Ott I, Reininger AJ, Neumann FJ. Effects of magnesium on platelet aggregation and adhesion. Magnesium modulates surface expression of glycoproteins on platelets in vitro and ex vivo. Thromb Haemost. 1994;72(6):912-918.
  49. Santilli F, Simeone P, Liani R, Davì G. Platelets and diabetes mellitus. Prostaglandins Other Lipid Mediat. 2015;120:28-39.
  50. Hunter RW, Hers I. Insulin/IGF-1 hybrid receptor expression on human platelets: consequences for the effect of insulin on platelet function. J Thromb Haemost. 2009;7(12):2123-2130.
  51. Westein E, Hoefer T, Calkin AC. Thrombosis in diabetes: a shear flow effect? Clin Sci (Lond). 2017;131(12):1245-1260.
  52. Vernstrom L, Funck KL, Grove EL, Laugesen E, Baier JM, Hvas AM, Poulsen PL. Antiplatelet effect of aspirin during 24h in patients with type 2 diabetes without cardiovascular disease. Thromb Res. 2018;161:1-6.
  53. Neergaard-Petersen S, Hvas AM, Ajjan R, Larsen SB, Würtz M, Kristensen SD, Grove EL. Platelet count, platelet turnover and fibrin clot structure in patients with coronary artery disease. Thromb Res. 2014;133(6):1161-1163.
  54. Parker WAE, Storey RF. Aspirin dosing in atherosclerotic cardiovascular disease: should we be more ADAPTABLE? Cardiovasc Res. 2021;117(10):e123-e125.
  55. Fejes Z, Poliska S, Czimmerer Z, Kaplar M, Penyige A, Gal Szabo G, Beke Debreceni I, Kunapuli SP, Kappelmayer J, Nagy B, Jr. Hyperglycaemia suppresses microRNA expression in platelets to increase P2RY12 and SELP levels in type 2 diabetes mellitus. Thromb Haemost. 2017;117(3):529-542.
  56. Pordzik J, Jakubik D, Jarosz-Popek J, Wicik Z, Eyileten C, De Rosa S, Indolfi C, Siller-Matula JM, Czajka P, Postula M. Significance of circulating microRNAs in diabetes mellitus type 2 and platelet reactivity: bioinformatic analysis and review. Cardiovasc Diabetol. 2019;18(1):113.
  57. Alzahrani SH, Ajjan RA. Coagulation and fibrinolysis in diabetes. Diab Vasc Dis Res. 2010;7:260-273.
  58. Kim HK, Kim JE, Park SH, Kim YI, Nam-Goong IS, Kim ES. High coagulation factor levels and low protein C levels contribute to enhanced thrombin generation in patients with diabetes who do not have macrovascular complications. J Diabetes Complications. 2014;28(3):365-369.
  59. Kearney K, Tomlinson D, Smith K, Ajjan R. Hypofibrinolysis in diabetes: a therapeutic target for the reduction of cardiovascular risk. Cardiovasc Diabetol. 2017;16(1):34.
  60. Ajjan RA, Gamlen T, Standeven KF, Mughal S, Hess K, Smith KA, Dunn EJ, Anwar MM, Rabbani N, Thornalley PJ, Philippou H, Grant PJ. Diabetes is associated with posttranslational modifications in plasminogen resulting in reduced plasmin generation and enzyme-specific activity. Blood. 2013;122(1):134-142.
  61. Du XL, Edelstein D, Rossetti L, Fantus IG, Goldberg H, Ziyadeh F, Wu J, Brownlee M. Hyperglycemia-induced mitochondrial superoxide overproduction activates the hexosamine pathway and induces plasminogen activator inhibitor-1 expression by increasing Sp1 glycosylation. Proc Natl Acad Sci U S A. 2000;97(22):12222-12226.
  62. Wang Y, Beck W, Deppisch R, Marshall SM, Hoenich NA, Thompson MG. Advanced glycation end products elicit externalization of phosphatidylserine in a subpopulation of platelets via 5-HT2A/2C receptors. Am J Physiol Cell Physiol. 2007;293(1):C328-336.
  63. Hindricks G, Potpara T, Dagres N, Arbelo E, Bax JJ, Blomström-Lundqvist C, Boriani G, Castella M, Dan G-A, Dilaveris PE, Fauchier L, Filippatos G, Kalman JM, La Meir M, Lane DA, Lebeau J-P, Lettino M, Lip GYH, Pinto FJ, Thomas GN, Valgimigli M, Van Gelder IC, Van Putte BP, Watkins CL, ESC Scientific Document Group. 2020 ESC Guidelines for the diagnosis and management of atrial fibrillation developed in collaboration with the European Association for Cardio-Thoracic Surgery (EACTS): The Task Force for the diagnosis and management of atrial fibrillation of the European Society of Cardiology (ESC) Developed with the special contribution of the European Heart Rhythm Association (EHRA) of the ESC. Eur Heart J. 2021;42(5):373-498.
  64. Pallisgaard JL, Schjerning AM, Lindhardt TB, Procida K, Hansen ML, Torp-Pedersen C, Gislason GH. Risk of atrial fibrillation in diabetes mellitus: A nationwide cohort study. Eur J Prev Cardiol. 2016;23(6):621-627.
  65. Overvad TF, Skjøth F, Lip GY, Lane DA, Albertsen IE, Rasmussen LH, Larsen TB. Duration of diabetes mellitus and risk of thromboembolism and bleeding in atrial fibrillation: nationwide cohort study. Stroke. 2015;46(8):2168-2174.
  66. Dublin S, Glazer NL, Smith NL, Psaty BM, Lumley T, Wiggins KL, Page RL, Heckbert SR. Diabetes mellitus, glycemic control, and risk of atrial fibrillation. J Gen Intern Med. 2010;25(8):853-858.
  67. Anselmino M, Matta M, D'Ascenzo F, Pappone C, Santinelli V, Bunch TJ, Neumann T, Schilling RJ, Hunter RJ, Noelker G, Fiala M, Frontera A, Thomas G, Katritsis D, Jais P, Weerasooriya R, Kalman JM, Gaita F. Catheter ablation of atrial fibrillation in patients with diabetes mellitus: a systematic review and meta-analysis. Europace. 2015;17(10):1518-1525.
  68. Fatemi O, Yuriditsky E, Tsioufis C, Tsachris D, Morgan T, Basile J, Bigger T, Cushman W, Goff D, Soliman EZ, Thomas A, Papademetriou V. Impact of intensive glycemic control on the incidence of atrial fibrillation and associated cardiovascular outcomes in patients with type 2 diabetes mellitus (from the Action to Control Cardiovascular Risk in Diabetes Study). Am J Cardiol. 2014;114(8):1217-1222.
  69. Chang SH, Wu LS, Chiou MJ, Liu JR, Yu KH, Kuo CF, Wen MS, Chen WJ, Yeh YH, See LC. Association of metformin with lower atrial fibrillation risk among patients with type 2 diabetes mellitus: a population-based dynamic cohort and in vitro studies. Cardiovasc Diabetol. 2014;13:123.
  70. Zhang Z, Zhang X, Korantzopoulos P, Letsas KP, Tse G, Gong M, Meng L, Li G, Liu T. Thiazolidinedione use and atrial fibrillation in diabetic patients: a meta-analysis. BMC Cardiovasc Disord. 2017;17(1):96.
  71. Zelniker TA, Bonaca MP, Furtado RHM, Mosenzon O, Kuder JF, Murphy SA, Bhatt DL, Leiter LA, McGuire DK, Wilding JPH, Budaj A, Kiss RG, Padilla F, Gause-Nilsson I, Langkilde AM, Raz I, Sabatine MS, Wiviott SD. Effect of dapagliflozin on atrial fibrillation in patients with type 2 diabetes mellitus. Circulation. 2020;141(15):1227-1234.
  72. Cosentino F, Grant PJ, Aboyans V, Bailey CJ, Ceriello A, Delgado V, Federici M, Filippatos G, Grobbee DE, Hansen TB, Huikuri HV, Johansson I, Juni P, Lettino M, Marx N, Mellbin LG, Ostgren CJ, Rocca B, Roffi M, Sattar N, Seferovic PM, Sousa-Uva M, Valensi P, Wheeler DC. 2019 ESC Guidelines on diabetes, pre-diabetes, and cardiovascular diseases developed in collaboration with the EASD. Eur Heart J. 2020;41(2):255-323.
  73. Lip GY, Nieuwlaat R, Pisters R, Lane DA, Crijns HJ. Refining clinical risk stratification for predicting stroke and thromboembolism in atrial fibrillation using a novel risk factor-based approach: the euro heart survey on atrial fibrillation. Chest. 2010;137(2):263-272.
  74. Lip GYH, Clementy N, Pierre B, Boyer M, Fauchier L. The impact of associated diabetic retinopathy on stroke and severe bleeding risk in diabetic patients with atrial fibrillation: the loire valley atrial fibrillation project. Chest. 2015;147(4):1103-1110.
  75. Patti G, Di Gioia G, Cavallari I, Nenna A. Safety and efficacy of nonvitamin K antagonist oral anticoagulants versus warfarin in diabetic patients with atrial fibrillation: A study-level meta-analysis of phase III randomized trials. Diabetes Metab Res Rev. 2017;33(3).
  76. Olesen JB, Lip GYH, Hansen ML, Hansen PR, Tolstrup JS, Lindhardsen J, Selmer C, Ahlehoff O, Olsen A-MS, Gislason GH, Torp-Pedersen C. Validation of risk stratification schemes for predicting stroke and thromboembolism in patients with atrial fibrillation: nationwide cohort study. BMJ. 2011;342:d124.
  77. January CT, Wann LS, Alpert JS, Calkins H, Cigarroa JE, Cleveland JC, Conti JB, Ellinor PT, Ezekowitz MD, Field ME, Murray KT, Sacco RL, Stevenson WG, Tchou PJ, Tracy CM, Yancy CW. 2014 AHA/ACC/HRS guideline for the management of patients with atrial fibrillation: a report of the American College of Cardiology/American Heart Association Task Force on practice guidelines and the Heart Rhythm Society. J Am Coll Cardiol. 2014;64(21):e1-e76.
  78. Olesen JB, Torp-Pedersen C, Hansen ML, Lip GY. The value of the CHA2DS2-VASc score for refining stroke risk stratification in patients with atrial fibrillation with a CHADS2 score 0-1: a nationwide cohort study. Thromb Haemost. 2012;107(6):1172-1179.
  79. Lip GYH, Keshishian A, Li X, Hamilton M, Masseria C, Gupta K, Luo X, Mardekian J, Friend K, Nadkarni A, Pan X, Baser O, Deitelzweig S. Effectiveness and Safety of Oral Anticoagulants Among Nonvalvular Atrial Fibrillation Patients. Stroke. 2018;49(12):2933-2944.
  80. Sim SY, Morrison A, Gregory R, Kong M. Anticoagulation in hyperosmolar hyperglycaemic state: a case report and review of the literature. Brit J Diabetes. 2021;21(2):250-254.
  81. Dhatariya K, Weston P. The argument against everyone with hyperosmolar hyperglycaemic syndrome being given prophylactic treatment dose anticoagulation. Brit J Diabetes. 2021;21(2):282-283.
  82. Cosentino F, Ceriello A, Baeres FMM, Fioretto P, Garber A, Stough WG, George JT, Grant PJ, Khunti K, Langkilde AM, Plutzky J, Rydén L, Scheen A, Standl E, Tuomilehto J, Zannad F. Addressing cardiovascular risk in type 2 diabetes mellitus: a report from the European Society of Cardiology Cardiovascular Roundtable. Eur Heart J. 2019;40(34):2907-2919.
  83. Collet J-P, Thiele H, Barbato E, Barthélémy O, Bauersachs J, Bhatt DL, Dendale P, Dorobantu M, Edvardsen T, Folliguet T, Gale CP, Gilard M, Jobs A, Jüni P, Lambrinou E, Lewis BS, Mehilli J, Meliga E, Merkely B, Mueller C, Roffi M, Rutten FH, Sibbing D, Siontis GCM, Group ESD. 2020 ESC Guidelines for the management of acute coronary syndromes in patients presenting without persistent ST-segment elevation: The Task Force for the management of acute coronary syndromes in patients presenting without persistent ST-segment elevation of the European Society of Cardiology (ESC). Eur Heart J. 2021;42(14):1289-1367.
  84. Ibanez B, James S, Agewall S, Antunes MJ, Bucciarelli-Ducci C, Bueno H, Caforio ALP, Crea F, Goudevenos JA, Halvorsen S, Hindricks G, Kastrati A, Lenzen MJ, Prescott E, Roffi M, Valgimigli M, Varenhorst C, Vranckx P, Widimský P, Group ESD. 2017 ESC Guidelines for the management of acute myocardial infarction in patients presenting with ST-segment elevation: The Task Force for the management of acute myocardial infarction in patients presenting with ST-segment elevation of the European Society of Cardiology (ESC). Eur Heart J. 2017;39(2):119-177.
  85. ISIS-2 Collaborative Group. Randomised trial of intravenous streptokinase, oral aspirin, both, or neither among 17,187 cases of suspected acute myocardial infarction: ISIS-2. Lancet. 1988;2(8607):349-360.
  86. Rocca B, Santilli F, Pitocco D, Mucci L, Petrucci G, Vitacolonna E, Lattanzio S, Mattoscio D, Zaccardi F, Liani R, Vazzana N, Del Ponte A, Ferrante E, Martini F, Cardillo C, Morosetti R, Mirabella M, Ghirlanda G, Davi G, Patrono C. The recovery of platelet cyclooxygenase activity explains interindividual variability in responsiveness to low-dose aspirin in patients with and without diabetes. J Thromb Haemost. 2012;10(7):1220-1230.
  87. Capodanno D, Patel A, Dharmashankar K, Ferreiro JL, Ueno M, Kodali M, Tomasello SD, Capranzano P, Seecheran N, Darlington A, Tello-Montoliu A, Desai B, Bass TA, Angiolillo DJ. Pharmacodynamic effects of different aspirin dosing regimens in type 2 diabetes mellitus patients with coronary artery disease. Circ Cardiovasc Interv. 2011;4:180-187.
  88. Spectre G, Arnetz L, Ostenson CG, Brismar K, Li N, Hjemdahl P. Twice daily dosing of aspirin improves platelet inhibition in whole blood in patients with type 2 diabetes mellitus and micro- or macrovascular complications. Thromb Haemost. 2011;106(3):491-499.
  89. Wiviott SD, Braunwald E, Angiolillo DJ, Meisel S, Dalby AJ, Verheugt FW, Goodman SG, Corbalan R, Purdy DA, Murphy SA, McCabe CH, Antman EM, for the TRITON-TIMI Investigators. Greater clinical benefit of more intensive oral antiplatelet therapy with prasugrel in patients with diabetes mellitus in the trial to assess improvement in therapeutic outcomes by optimizing platelet inhibition with prasugrel-thrombolysis in myocardial infarction 38. Circulation. 2008;118:1626-1636.
  90. Franchi F, James SK, Ghukasyan Lakic T, Budaj AJ, Cornel JH, Katus HA, Keltai M, Kontny F, Lewis BS, Storey RF, Himmelmann A, Wallentin L, Angiolillo DJ. impact of diabetes mellitus and chronic kidney disease on cardiovascular outcomes and platelet P2Y(12) receptor antagonist effects in patients with acute coronary syndromes: insights from the PLATO trial. J Am Heart Assoc. 2019;8(6):e011139.
  91. Yusuf S, Zhao F, Mehta S, Chrolavicius S, Tognoni G, Fox K. Effects of clopidogrel in addition to aspirin in patients with acute coronary syndromes without ST-segment elevation. New Eng J Med. 2001;345:494-502.
  92. Sabatine MS, Cannon CP, Gibson CM, Lopez-Sendon JL, Montalescot G, Theroux P, Claeys MJ, Cools F, Hill KA, Skene AM, McCabe CH, Braunwald E. Addition of clopidogrel to aspirin and fibrinolytic therapy for myocardial infarction with ST-segment elevation. N Engl J Med. 2005;352(12):1179-1189.
  93. Chen ZM, Jiang LX, Chen YP, Xie JX, Pan HC, Peto R, Collins R, Liu LS, COMMIT (ClOpidogrel and Metoprolol in Myocardial Infarction Trial) collaborative group. Addition of clopidogrel to aspirin in 45 852 patients with acute myocardial infarction: randomised placebo-controlled trial. Lancet. 2005;366(9497):1607-1621.
  94. Wiviott S, Braunwald E, McCabe C, Montalescot G, Ruzyllo W, Gottlieb S, Neumann F-J, Ardissino D, De Servi S, Murphy S, Riesmeyer J, Weerakkody G, Gibson C, Antman E. Prasugrel versus clopidogrel in patients with acute coronary syndromes. New Eng J Med. 2007;357:2001-2015.
  95. Roe MT, Armstrong PW, Fox KA, White HD, Prabhakaran D, Goodman SG, Cornel JH, Bhatt DL, Clemmensen P, Martinez F, Ardissino D, Nicolau JC, Boden WE, Gurbel PA, Ruzyllo W, Dalby AJ, McGuire DK, Leiva-Pons JL, Parkhomenko A, Gottlieb S, Topacio GO, Hamm C, Pavlides G, Goudev AR, Oto A, Tseng CD, Merkely B, Gasparovic V, Corbalan R, Cinteza M, McLendon RC, Winters KJ, Brown EB, Lokhnygina Y, Aylward PE, Huber K, Hochman JS, Ohman EM. Prasugrel versus clopidogrel for acute coronary syndromes without revascularization. N Engl J Med. 2012;367(14):1297-1309.
  96. Wallentin L, Becker RC, Budaj A, Cannon CP, Emanuelsson H, Held C, Horrow J, Husted S, James S, Katus H, Mahaffey KW, Scirica BM, Skene A, Steg PG, Storey RF, Harrington RA. Ticagrelor versus clopidogrel in patients with acute coronary syndromes. N Engl J Med. 2009;361(11):1045-1057.
  97. Schupke S, Neumann FJ, Menichelli M, Mayer K, Bernlochner I, Wohrle J, Richardt G, Liebetrau C, Witzenbichler B, Antoniucci D, Akin I, Bott-Flugel L, Fischer M, Landmesser U, Katus HA, Sibbing D, Seyfarth M, Janisch M, Boncompagni D, Hilz R, Rottbauer W, Okrojek R, Mollmann H, Hochholzer W, Migliorini A, Cassese S, Mollo P, Xhepa E, Kufner S, Strehle A, Leggewie S, Allali A, Ndrepepa G, Schuhlen H, Angiolillo DJ, Hamm CW, Hapfelmeier A, Tolg R, Trenk D, Schunkert H, Laugwitz KL, Kastrati A. Ticagrelor or prasugrel in patients with acute coronary syndromes. N Engl J Med. 2019;381(16):1524-1534.
  98. Storey RF. Ticagrelor versus prasugrel for PCI-managed myocardial infarction: the battle of the giants continues. Heart. 2021;107(14):1111.
  99. Ndrepepa G, Kastrati A, Menichelli M, Neumann F-J, Wöhrle J, Bernlochner I, Richardt G, Witzenbichler B, Sibbing D, Gewalt S, Angiolillo Dominick J, Hamm Christian W, Hapfelmeier A, Trenk D, Laugwitz K-L, Schunkert H, Schüpke S, Mayer K. Ticagrelor or prasugrel in patients with acute coronary syndromes and diabetes mellitus. JACC: Cardiovasc Intervent. 2020;13(19):2238-2247.
  100. Mehran R, Baber U, Sharma SK, Cohen DJ, Angiolillo DJ, Briguori C, Cha JY, Collier T, Dangas G, Dudek D, Dzavik V, Escaned J, Gil R, Gurbel P, Hamm CW, Henry T, Huber K, Kastrati A, Kaul U, Kornowski R, Krucoff M, Kunadian V, Marx SO, Mehta SR, Moliterno D, Ohman EM, Oldroyd K, Sardella G, Sartori S, Shlofmitz R, Steg PG, Weisz G, Witzenbichler B, Han YL, Pocock S, Gibson CM. Ticagrelor with or without aspirin in high-risk patients after PCI. N Engl J Med. 2019;381(21):2032-2042.
  101. Vranckx P, Valgimigli M, Juni P, Hamm C, Steg PG, Heg D, van Es GA, McFadden EP, Onuma Y, van Meijeren C, Chichareon P, Benit E, Mollmann H, Janssens L, Ferrario M, Moschovitis A, Zurakowski A, Dominici M, Van Geuns RJ, Huber K, Slagboom T, Serruys PW, Windecker S. Ticagrelor plus aspirin for 1 month, followed by ticagrelor monotherapy for 23 months vs aspirin plus clopidogrel or ticagrelor for 12 months, followed by aspirin monotherapy for 12 months after implantation of a drug-eluting stent: a multicentre, open-label, randomised superiority trial. Lancet. 2018;392(10151):940-949.
  102. Chichareon P, Modolo R, Kogame N, Takahashi K, Chang CC, Tomaniak M, Botelho R, Eeckhout E, Hofma S, Trendafilova-Lazarova D, Kőszegi Z, Iñiguez A, Wykrzykowska JJ, Piek JJ, Garg S, Hamm C, Steg PG, Jüni P, Vranckx P, Valgimigli M, Windecker S, Onuma Y, Serruys PW. Association of diabetes with outcomes in patients undergoing contemporary percutaneous coronary intervention: Pre-specified subgroup analysis from the randomized GLOBAL LEADERS study. Atherosclerosis. 2020;295:45-53.
  103. Bhalla A, Patel M, Birns J. An update on hyper-acute management of ischaemic stroke. Clin Med (Lond). 2021;21(3):215-221.
  104. Capodanno D, Alberts M, Angiolillo DJ. Antithrombotic therapy for secondary prevention of atherothrombotic events in cerebrovascular disease. Nat Rev Cardiol. 2016;13(10):609-622.
  105. Powers WJ, Rabinstein AA, Ackerson T, Adeoye OM, Bambakidis NC, Becker K, Biller J, Brown M, Demaerschalk BM, Hoh B, Jauch EC, Kidwell CS, Leslie-Mazwi TM, Ovbiagele B, Scott PA, Sheth KN, Southerland AM, Summers DV, Tirschwell DL, null n. Guidelines for the early management of patients with acute ischemic Stroke: 2019 update to the 2018 guidelines for the early management of acute ischemic stroke: a guideline for healthcare professionals from the American Heart Association/American Stroke Association. Stroke. 2019;50(12):e344-e418.
  106. Wang Y, Wang Y, Zhao X, Liu L, Wang D, Wang C, Wang C, Li H, Meng X, Cui L, Jia J, Dong Q, Xu A, Zeng J, Li Y, Wang Z, Xia H, Johnston SC. Clopidogrel with aspirin in acute minor stroke or transient ischemic attack. New Eng J Med. 2013;369(1):11-19.
  107. Johnston SC, Easton JD, Farrant M, Barsan W, Conwit RA, Elm JJ, Kim AS, Lindblad AS, Palesch YY. Clopidogrel and aspirin in acute ischemic stroke and high-risk TIA. N Engl J Med. 2018;379(3):215-225.
  108. Johnston SC, Amarenco P, Albers GW, Denison H, Easton JD, Evans SR, Held P, Jonasson J, Minematsu K, Molina CA, Wang Y, Wong KS. Ticagrelor versus aspirin in acute stroke or transient ischemic attack. N Engl J Med. 2016;375(1):35-43.
  109. Johnston SC, Amarenco P, Denison H, Evans SR, Himmelmann A, James S, Knutsson M, Ladenvall P, Molina CA, Wang Y. Ticagrelor and aspirin or aspirin alone in acute ischemic stroke or tia. New Eng J Med. 2020;383(3):207-217.
  110. Antiplatelet Trialists' Collaboration. Collaborative meta-analysis of randomised trials of antiplatelet therapy for prevention of death, myocardial infarction, and stroke in high risk patients. BMJ. 2002;324(7329):71-86.
  111. Parker WAE, Sagar R, Kurdee Z, Hawkins F, Naseem KM, Grant PJ, Storey RF, Ajjan RA. A randomised controlled trial to assess the antithrombotic effects of aspirin in type 1 diabetes: role of dosing and glycaemic control. Cardiovasc Diabetol. 2021;20(1):238.
  112. CURRENT-OASIS 7 Investigators, Mehta S, Bassand J, Chrolavicius S, Diaz R, Eikelboom J, Fox K, Granger C, Jolly S, Joyner C, Rupprecht H, Widimsky P, Afzal R, Pogue J, Yusuf S. Dose comparisons of clopidogrel and aspirin in acute coronary syndromes. New Eng J Med. 2010;363(10):930-942.
  113. Jones WS, Mulder H, Wruck LM, Pencina MJ, Kripalani S, Muñoz D, Crenshaw DL, Effron MB, Re RN, Gupta K, Anderson RD, Pepine CJ, Handberg EM, Manning BR, Jain SK, Girotra S, Riley D, DeWalt DA, Whittle J, Goldberg YH, Roger VL, Hess R, Benziger CP, Farrehi P, Zhou L, Ford DE, Haynes K, VanWormer JJ, Knowlton KU, Kraschnewski JL, Polonsky TS, Fintel DJ, Ahmad FS, McClay JC, Campbell JR, Bell DS, Fonarow GC, Bradley SM, Paranjape A, Roe MT, Robertson HR, Curtis LH, Sharlow AG, Berdan LG, Hammill BG, Harris DF, Qualls LG, Marquis-Gravel G, Modrow MF, Marcus GM, Carton TW, Nauman E, Waitman LR, Kho AM, Shenkman EA, McTigue KM, Kaushal R, Masoudi FA, Antman EM, Davidson DR, Edgley K, Merritt JG, Brown LS, Zemon DN, McCormick TE, 3rd, Alikhaani JD, Gregoire KC, Rothman RL, Harrington RA, Hernandez AF. Comparative effectiveness of aspirin dosing in cardiovascular disease. N Engl J Med. 2021;384(21):1981-1990.
  114. CAPRIE Steering Committee. A randomised, blinded, trial of clopidogrel versus aspirin in patients at risk of ischaemic events (CAPRIE). Lancet. 1996;348(9038):1329-1339.
  115. Bhatt DL, Fox KAA, Hacke W, Berger PB, Black HR, Boden WE, Cacoub P, Cohen EA, Creager MA, Easton JD, Flather MD, Haffner SM, Hamm CW, Hankey GJ, Johnston SC, Mak K-H, Mas J-L, Montalescot G, Pearson TA, Steg PG, Steinhubl SR, Weber MA, Brennan DM, Fabry-Ribaudo L, Booth J, Topol EJ, the CI. Clopidogrel and aspirin versus aspirin alone for the prevention of atherothrombotic events. New Eng J Med. 2006;354(16):1706-1717.
  116. Bhatt DL, Flather MD, Hacke W, Berger PB, Black HR, Boden WE, Cacoub P, Cohen EA, Creager MA, Easton JD, Hamm CW, Hankey GJ, Johnston SC, Mak KH, Mas JL, Montalescot G, Pearson TA, Steg PG, Steinhubl SR, Weber MA, Fabry-Ribaudo L, Hu T, Topol EJ, Fox KA. Patients with prior myocardial infarction, stroke, or symptomatic peripheral arterial disease in the CHARISMA trial. J Am Coll Cardiol. 2007;49(19):1982-1988.
  117. Mauri L, Elmariah S, Yeh RW, Cutlip DE, Steg PG, Windecker S, Wiviott SD, Cohen DJ, Massaro JM, D'Agostino RB, Sr., Braunwald E, Kereiakes DJ. Causes of late mortality with dual antiplatelet therapy after coronary stents. Eur Heart J. 2016;37(4):378-385.
  118. Bonaca MP, Bhatt DL, Cohen M, Steg PG, Storey RF, Jensen EC, Magnani G, Bansilal S, Fish MP, Im K, Bengtsson O, Ophuis TO, Budaj A, Theroux P, Ruda M, Hamm C, Goto S, Spinar J, Nicolau JC, Kiss RG, Murphy SA, Wiviott SD, Held P, Braunwald E, Sabatine MS. Long-term use of ticagrelor in patients with prior myocardial infarction. New Engl J Med. 2015;372:1791-1800.
  119. Bhatt DL, Bonaca MP, Bansilal S, Angiolillo DJ, Cohen M, Storey RF, Im K, Murphy SA, Held P, Braunwald E, Sabatine MS, Steg PG. Reduction in ischemic events with ticagrelor in diabetic patients: from the PEGASUS-TIMI 54 trial. J Am Coll Cardiol. 2016;67:2732-2740.
  120. Magnani G, Ardissino D, Im K, Budaj A, Storey RF, Steg PG, Bhatt DL, Cohen M, Oude Ophius T, Goudev A, Parkhomenko A, Kamensky G, Angiolillo DJ, López-Sendón J, Johanson P, Braunwald E, Sabatine MS, Bonaca MP. Predictors, type, and impact of bleeding on the net clinical benefit of long-term ticagrelor in stable patients with prior myocardial infarction. J Am Heart Assoc. 2021;10(4):e017008.
  121. Steg PG, Bhatt DL, Simon T, Fox K, Mehta SR, Harrington RA, Held C, Andersson M, Himmelmann A, Ridderstrale W, Leonsson-Zachrisson M, Liu Y, Opolski G, Zateyshchikov D, Ge J, Nicolau JC, Corbalan R, Cornel JH, Widimsky P, Leiter LA. Ticagrelor in Patients with Stable Coronary Disease and Diabetes. N Engl J Med. 2019;381(14):1309-1320.
  122. Bhatt DL, Steg PG, Mehta SR, Leiter LA, Simon T, Fox K, Held C, Andersson M, Himmelmann A, Ridderstrale W, Chen J, Song Y, Diaz R, Goto S, James SK, Ray KK, Parkhomenko AN, Kosiborod MN, McGuire DK, Harrington RA. Ticagrelor in patients with diabetes and stable coronary artery disease with a history of previous percutaneous coronary intervention (THEMIS-PCI): a phase 3, placebo-controlled, randomised trial. Lancet. 2019;394(10204):1169-1180.
  123. Lackey LG, Garnett CE, Senatore F. Applying decision analysis to inform the US Food and Drug Administration’s benefit–risk assessment of ticagrelor for primary prevention of myocardial infarction or stroke based on THEMIS. Circulation. 2021;144(8):655-658.
  124. Eikelboom JW, Connolly SJ, Bosch J, Dagenais GR, Hart RG, Shestakovska O, Diaz R, Alings M, Lonn EM, Anand SS, Widimsky P, Hori M, Avezum A, Piegas LS, Branch KRH, Probstfield J, Bhatt DL, Zhu J, Liang Y, Maggioni AP, Lopez-Jaramillo P, O'Donnell M, Kakkar AK, Fox KAA, Parkhomenko AN, Ertl G, Stork S, Keltai M, Ryden L, Pogosova N, Dans AL, Lanas F, Commerford PJ, Torp-Pedersen C, Guzik TJ, Verhamme PB, Vinereanu D, Kim JH, Tonkin AM, Lewis BS, Felix C, Yusoff K, Steg PG, Metsarinne KP, Cook Bruns N, Misselwitz F, Chen E, Leong D, Yusuf S. Rivaroxaban with or without aspirin in stable cardiovascular disease. N Engl J Med. 2017;377(14):1319-1330.
  125. Knuuti J, Wijns W, Saraste A, Capodanno D, Barbato E, Funck-Brentano C, Prescott E, Storey RF, Deaton C, Cuisset T, Agewall S, Dickstein K, Edvardsen T, Escaned J, Gersh BJ, Svitil P, Gilard M, Hasdai D, Hatala R, Mahfoud F, Masip J, Muneretto C, Valgimigli M, Achenbach S, Bax JJ. 2019 ESC Guidelines for the diagnosis and management of chronic coronary syndromes. Eur Heart J. 2020;41(3):407-477.
  126. Antithrombotic Trialists' Collaboration. Collaborative meta-analysis of randomised trials of antiplatelet therapy for prevention of death, myocardial infarction, and stroke in high risk patients. BMJ. 2002;324(7329):71-86.
  127. Halkes PH, van Gijn J, Kappelle LJ, Koudstaal PJ, Algra A. Aspirin plus dipyridamole versus aspirin alone after cerebral ischaemia of arterial origin (ESPRIT): randomised controlled trial. Lancet. 2006;367(9523):1665-1673.
  128. Pan Y, Elm JJ, Li H, Easton JD, Wang Y, Farrant M, Meng X, Kim AS, Zhao X, Meurer WJ, Liu L, Dietrich D, Johnston SC. Outcomes associated with clopidogrel-aspirin use in minor stroke or transient ischemic attack: a pooled analysis of Clopidogrel in High-risk patients with Acute Non-disabling Cerebrovascular Events (CHANCE) and Platelet-Oriented Inhibition in New TIA and minor ischemic stroke (POINT) trials. JAMA Neurol. 2019;76(12):1466-1473.
  129. Johnston SC, Amarenco P, Denison H, Evans SR, Himmelmann A, James S, Knutsson M, Ladenvall P, Molina CA, Wang Y. The acute stroke or transient ischemic attack treated with ticagrelor and aspirin for prevention of stroke and death (THALES) trial: rationale and design. Int J Stroke. 2019:1747493019830307.
  130. Benavente OR, Hart RG, McClure LA, Szychowski JM, Coffey CS, Pearce LA. Effects of clopidogrel added to aspirin in patients with recent lacunar stroke. N Engl J Med. 2012;367(9):817-825.
  131. Greving JP, Diener HC, Reitsma JB, Bath PM, Csiba L, Hacke W, Kappelle LJ, Koudstaal PJ, Leys D, Mas JL, Sacco RL, Algra A. Antiplatelet Therapy After Noncardioembolic Stroke. Stroke. 2019;50(7):1812-1818.
  132. Aboyans V, Ricco JB, Bartelink MEL, Bjorck M, Brodmann M, Cohnert T, Collet JP, Czerny M, De Carlo M, Debus S, Espinola-Klein C, Kahan T, Kownator S, Mazzolai L, Naylor AR, Roffi M, Rother J, Sprynger M, Tendera M, Tepe G, Venermo M, Vlachopoulos C, Desormais I. 2017 ESC Guidelines on the Diagnosis and Treatment of Peripheral Arterial Diseases, in collaboration with the European Society for Vascular Surgery (ESVS): Document covering atherosclerotic disease of extracranial carotid and vertebral, mesenteric, renal, upper and lower extremity arteriesEndorsed by: the European Stroke Organization (ESO)The Task Force for the Diagnosis and Treatment of Peripheral Arterial Diseases of the European Society of Cardiology (ESC) and of the European Society for Vascular Surgery (ESVS). Eur Heart J. 2018;39(9):763-816.
  133. Hiatt WR, Fowkes FG, Heizer G, Berger JS, Baumgartner I, Held P, Katona BG, Mahaffey KW, Norgren L, Jones WS, Blomster J, Millegård M, Reist C, Patel MR. Ticagrelor versus clopidogrel in symptomatic peripheral artery disease. N Engl J Med. 2017;376:32-40.
  134. Parker WAE, Schulte C, Barwari T, Phoenix F, Pearson SM, Mayr M, Grant PJ, Storey RF, Ajjan RA. Aspirin, clopidogrel and prasugrel monotherapy in patients with type 2 diabetes mellitus: a double-blind randomised controlled trial of the effects on thrombotic markers and microRNA levels. Cardiovasc Diabetol. 2020;19(1):3.
  135. Cacoub PP, Bhatt DL, Steg PG, Topol EJ, Creager MA. Patients with peripheral arterial disease in the CHARISMA trial. Eur Heart J. 2009;30(2):192-201.
  136. Anand SS, Bosch J, Eikelboom JW, Connolly SJ, Diaz R, Widimsky P, Aboyans V, Alings M, Kakkar AK, Keltai K, Maggioni AP, Lewis BS, Stork S, Zhu J, Lopez-Jaramillo P, O'Donnell M, Commerford PJ, Vinereanu D, Pogosova N, Ryden L, Fox KAA, Bhatt DL, Misselwitz F, Varigos JD, Vanassche T, Avezum AA, Chen E, Branch K, Leong DP, Bangdiwala SI, Hart RG, Yusuf S. Rivaroxaban with or without aspirin in patients with stable peripheral or carotid artery disease: an international, randomised, double-blind, placebo-controlled trial. Lancet. 2018;391(10117):219-229.
  137. Bonaca MP, Bauersachs RM, Anand SS, Debus ES, Nehler MR, Patel MR, Fanelli F, Capell WH, Diao L, Jaeger N, Hess CN, Pap AF, Kittelson JM, Gudz I, Mátyás L, Krievins DK, Diaz R, Brodmann M, Muehlhofer E, Haskell LP, Berkowitz SD, Hiatt WR. Rivaroxaban in peripheral artery disease after revascularization. New Eng J Med. 2020;382(21):1994-2004.
  138. Baigent C, Blackwell L, Collins R, Emberson J, Godwin J, Peto R, Buring J, Hennekens C, Kearney P, Meade T, Patrono C, Roncaglioni MC, Zanchetti A. Aspirin in the primary and secondary prevention of vascular disease: collaborative meta-analysis of individual participant data from randomised trials. Lancet. 2009;373(9678):1849-1860.
  139. Belch J, MacCuish A, Campbell I, Cobbe S, Taylor R, Prescott R, Lee R, Bancroft J, MacEwan S, Shepherd J, Macfarlane P, Morris A, Jung R, Kelly C, Connacher A, Peden N, Jamieson A, Matthews D, Leese G, McKnight J, O’Brien I, Semple C, Petrie J, Gordon D, Pringle S, MacWalter R. The prevention of progression of arterial disease and diabetes (POPADAD) trial: factorial randomised placebo controlled trial of aspirin and antioxidants in patients with diabetes and asymptomatic peripheral arterial disease. BMJ. 2008;337.
  140. Ogawa H, Nakayama M, Morimoto T, Uemura S, Kanauchi M, Doi N, Jinnouchi H, Sugiyama S, Saito Y. Low-dose aspirin for primary prevention of atherosclerotic events in patients with type 2 diabetes: a randomized controlled trial. JAMA. 2008;300(18):2134-2141.
  141. 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 aspirin for primary prevention in persons with diabetes mellitus. N Engl J Med. 2018;379(16):1529-1539.

Dysbetalipoproteinemia (Type III Hyperlipoproteinemia)

ABSTRACT

 

Dysbetalipoproteinemia is an underrecognized and underdiagnosed genetic lipid disorder characterized by pathogenic variants in the APOE gene, which encodes apolipoprotein (apo) E. It leads to the abnormal accumulation of triglyceride-rich remnant lipoproteins, elevated levels of both cholesterol and triglycerides, and an increased risk of cardiovascular disease. Typically, patients with autosomal recessive form of dysbetalipoproteinemia are homozygous for the e2 allele, which is associated with decreased binding of apo E to the LDL receptor and/or heparan sulfate proteoglycans, resulting in impaired remnant clearance. However, only a minority of apo e2 homozygotes become hyperlipidemic, often due to metabolic conditions that either increase lipoprotein production or decrease remnant clearance. Rarer variants in the APOE gene are linked to autosomal dominant dysbetalipoproteinemia. Palmar xanthoma is considered a characteristic feature of dysbetalipoproteinemia, although it is observed in fewer than half of affected individuals. Both total cholesterol and triglyceride levels are typically elevated and may be of similar magnitude. A low apo B level relative to a high total cholesterol level or a discrepancy between calculated LDL-cholesterol (LDL-C) and direct LDL-C levels can raise suspicion of this condition. There is no simple diagnostic test for dysbetalipoproteinemia, and diagnosis traditionally requires the detection of b-VLDL (remnant lipoproteins) and pathogenic variants in the APOE gene, both of which are not routinely available in clinical laboratories. Several algorithms using various lipid and apo B parameters have been proposed for screening and selecting candidates for genetic testing. Recent data suggest that the phenotype of dysbetalipoproteinemia is heterogeneous. The term multifactorial remnant cholesterol disease has been proposed to describe a milder form of dysbetalipoproteinemia in individuals without the apo e2/e2 genotype, differentiating them from the more severe form associated with apo e2/e2 genotype. Patients with dysbetalipoproteinemia are at an increased risk of cardiovascular diseases, particularly coronary artery disease and peripheral arterial disease. However, they generally respond well to lifestyle modifications and conventional lipid-lowering therapies, including statins and fibrates.

 

INTRODUCTION

 

Dysbetalipoproteinemia has been recognized since the 1950s and several names have been used in the literature, including xanthoma tuberosum, familial dysbetalipoproteinemia, broad beta disease, type III hyperlipoproteinemia, and remnant removal disease. Originally described by Gofman and colleagues, affected patients developed tuberous xanthoma of the extensor tendons and palmar xanthoma of the skin creases (1). An abnormal lipoprotein profile determined by analytical ultracentrifugation showed an increase in lipoproteins corresponding to small very low-density lipoprotein (VLDL) and intermediate-density lipoprotein (IDL). Using a combination of ultracentrifugation and paper electrophoresis, these cholesterol-enriched lipoprotein fractions displayed abnormal flotation and b electrophoretic mobility, instead of the normal pre-b mobility (2), and were referred to as floating beta lipoproteins or b-VLDL (3,4). The term “dysbetalipoproteinemia” was used to describe the presence of these b-VLDL in the circulation although the amount may not be high enough to cause elevated lipid levels. Overt hyperlipidemia observed in certain patients with dysbetalipoproteinemia was identified as being identical to type III hyperlipidemia, as classified by Fredrickson et al. in 1967 (4). The term “broad beta disease” represented the peculiar migration pattern of these abnormal b-VLDL on paper electrophoresis (4). These b-VLDL were shown to be remnants of apolipoprotein (apo) B-containing lipoproteins of both hepatic and intestinal origin (5), which accumulated in the plasma due to defective clearance (6). Havel and Kane later demonstrated that subjects with type III hyperlipidemia exhibited elevated levels of apo E, originally referred to as arginine-rich protein  (7). Using isoelectric focusing, Utermann et al. found that apo E3 was absent in these subjects (8)and homozygosity for the pathogenic variant of apo E, referred to as apo E2 as opposed to the normal apo E3, was later found to be the underlying genetic defect (9,10). The complete amino acid sequences of different isoforms of apo E were determined by Mahley et al., which helped define the molecular abnormality of apo E in the pathogenesis of dysbetalipoproteinemia (11). Apo E was subsequently shown to be a major ligand for the LDL receptor and heparan sulfate proteoglycans (HSPGs), establishing apo E as the main apolipoprotein responsible for the uptake of remnant particles into hepatocytes (12). Defective binding of apo E to the receptors and impaired hepatic uptake could therefore explain the accumulation of remnant lipoproteins in these patients (13), although other environmental factors could modulate the expression of the abnormal lipid profile.

 

It is important to note that different terminology is often used in the literature to describe dysbetalipoproteinemia. In general, the term dysbetalipoproteinemia is used to indicate the presence of b-VLDL remnant particles in the circulation, whereas type III hyperlipidemia or type III hyperlipoproteinemia refers to the hyperlipidemic phenotype resulting from the accumulation of these remnant lipoproteins. In this article, we use the term dysbetalipoproteinemia to refer to the lipoprotein disorder characterized by the presence of b-VLDL in the circulation, which is often associated with hyperlipidemic phenotype.

 

EPIDEMIOLOGY

 

The prevalence of dysbetalipoproteinemia varies depending upon the definition used for diagnosis and the study population. Using the original gold standard diagnostic criteria by Fredrickson et al. (14) (a VLDL-cholesterol/plasma triglyceride (VLDL-C/TG) ratio >0.30 and the presence of b-VLDL on gel electrophoresis without requiring apo E genotype), the population-based prevalence of dysbetalipoproteinemia in the Northern American population was reported around 0.4% (1 in 250) in men aged 20 years or older and 0.2% (1 in 500) in similarly aged women (15,16). The prevalence is higher in men than in women and it tends to occur earlier in men (17,18). A similar prevalence of 0.2-0.4% was reported from a free-living population in California and in Vermont using lipoprotein electrophoresis as a diagnostic tool (19,20). In the 2011-2014 National Health and Nutrition Examination Survey (NHANES) participants in the U.S., the prevalence of 0.2-0.8% was reported using the lipoprotein levels from ultracentrifugation, but it increased to 1.97% when using only lipid and apo B levels (21). In studies using both lipid levels and the apo E genotype, a prevalence of 0.1% (1 in 889) was reported among 8,888 Dutch population (22), and a prevalence of 0.2% (1 in 469) was reported from 452,469 UK Biobank participants (23). The prevalence among different genetic ancestries was relatively similar and did not exceed 0.2% in any ancestry (23). Another study in Russia showed a prevalence of 0.67% (1 in 150) using the apo E genotype and triglyceride level ³130 mg/dL or 1.5 mmol/L (24). Collectively, the overall prevalence of dysbetalipoproteinemia, based on gold standard criteria and genetic testing, is estimated to be around 0.1–0.8%. Interestingly, this estimate is comparable to that of familial hypercholesterolemia (25).

 

GENETICS

 

Dysbetalipoproteinemia is caused by a genetic defect in the APOE gene, which encodes apo E. Apo E is a polymorphic glycoprotein found in various lipoprotein particles, including chylomicrons, chylomicron remnants, VLDL, VLDL remnants, and HDL. The main function of apo E is to mediate the interaction between apo E-containing lipoproteins and lipoprotein receptors. The N-terminal domain of apo E is involved in the interaction with the LDL receptor, the LDL receptor-related protein (LRP), and HSPGs, whereas the C-terminal domain is responsible for lipid binding. Amino acid residues 154-168 in the N-terminus contained several critical basic amino acids (i.e., arginine and lysine) that interact with acidic amino acid residues of the lipoprotein receptors and HSPGs.

 

The APOE gene is in the apolipoprotein gene cluster on the long arm of chromosome 19. It has 4 exons and 3 introns. Apo E is primarily synthesized in the liver, but other tissues can also produce apo E, including the brain, spleen, lung, kidneys, adrenals, ovaries, macrophages, and smooth muscle cells (26). After cleavage of the 18-amino acid signal peptide, the mature apo E protein has 299 amino acids. Three major isoforms of apo E (E3, E2, and E4) exist, which are caused by a single amino acid substitution at two different sites of the protein (27) as shown in Table 1. The differences among these isoforms result from different apo E alleles. The alleles are given designations using the Greek letter epsilon, i.e., e2, e3, and e4. Although the e3 is suggested to be a normal or wild-type allele, evidence exists that e4 allele may be the ancestral allele (28). The 3 apo E alleles yield 6 possible phenotypes, i.e., E2/E2, E2/E3, E2/E4, E3/E3, E3/E4, and E4/E4. The classical molecular abnormality causing dysbetalipoproteinemia is the homozygous variant known as the E2/E2 phenotype, which leads to a substitution of arginine for cysteine at position 176 (p.Arg176Cys). This variant is associated with an autosomal recessive inheritance of dysbetalipoproteinemia.

 

Table 1. Major Isoforms of Apo E Due to Different Amino Acids and Charges

Isoform

E2

E3

E4

Apo E allele

e2

e3

e4

rs number

rs7412

-

rs429358

HGVSc

c.526C>T

-

c.388T>C

HGVSp

p.Arg176Cys

-

p.Cys130Arg

Residue 130 (112*)

Cys

Cys

Arg

Residue 176 (158*)

Cys

Arg

Arg

Charge

0

+1

+2

Lipoprotein preference

HDL

HDL

VLDL

* Used in the old literature, which does not include the 18-amino acid signal peptide.

 

In almost all populations, the e3 allele makes up a majority of the apo E gene pool (70-80%), followed by e4 (10-15%) and e2 (5-10%). Therefore, the most common phenotype is the apo E3/E3 phenotype, which is found in 50-70% of the population, whereas the apo E2/E2 phenotype is relatively rare (24). Data from the UK Biobank indicate that apo E2 homozygosity is present in 0.2–1.3% of individuals, depending on genetic ancestry (23), and less than 20% of those with the apo E2/E2 phenotype develop overt hyperlipidemia (22), despite having demonstrable b-VLDL in the plasma, characteristic of dysbetalipoproteinemia.

 

Rarer variants in the APOE gene cause an autosomal dominant form of dysbetalipoproteinemia. Except for APOELeiden, which has a tandem duplication of 21 nucleotides coding for 7 amino acids, most of these rare variants involve substitutions of neutral or acidic amino acids for basic ones in the critical amino acid residues 154-168 that interact with lipoprotein receptors. The p.Arg163Cys variant is particularly common in subjects of African descents with the prevalence of 5-12% (29). Another rare cause of autosomal dominant dysbetalipoproteinemia is due to apo E deficiency (30,31).

 

PATHOPHYSIOLOGY

 

Chylomicrons produced by the small intestine and VLDL produced by the liver are both processed by lipoprotein lipase in the lipolytic cascade, resulting in triglyceride hydrolysis and the formation of chylomicron remnants and VLDL remnants, respectively. Normally, these remnant lipoproteins are cleared by receptors in the liver, including the LDL receptor and LDL receptor-related protein (LRP). Apo E plays a critical role in the binding, uptake, and hepatic clearance of remnant lipoprotein particles. Synthesized primarily by hepatocytes, apo E is secreted into the space of Disse, where it associates with remnant lipoproteins. Two major pathways mediate the clearance of remnant lipoprotein particles (32,33). First, apo E-containing remnant lipoproteins directly interact with the LDL receptor and are internalized into hepatocytes via the classical LDL receptor-mediated pathway. Second, apo E-enriched remnant lipoproteins interact with the cell-surface HSPGs before being transferred to LRP and internalized into hepatocytes through LRP. In addition, HSPGs alone can directly mediate lipoprotein uptake. HSPGs are transmembrane core proteins with attached heparan sulfate chains. These heparan sulfate chains are highly negatively charged sugar polymers capable of capturing lipoproteins and other ligands (33). Thus, the ligand-binding domains of HSPGs are carbohydrates, not proteins. Syndecan-1, an abundant HSPG on the hepatic surface in the space of Disse, is particularly important for remnant clearance (34,35). Furthermore, there is evidence suggesting that scavenger receptor class B type I (SR-BI) may also serve as an additional hepatic remnant receptor (36). Figure 1 illustrates the pathways involved in the clearance of remnant lipoproteins.

 

Figure 1. Clearance pathways of remnant lipoproteins.

 

The presence of abnormal apo E, which is defective in binding to HSPGs or hepatic receptors, or the absence of apo E could lead to impaired clearance of these remnant particles and the accumulation in the circulation, resulting in elevated levels of cholesterol and triglyceride. Different apo E variants likely affect different pathways of remnant lipoprotein particles, resulting in different patterns of hyperlipidemia and inheritance (37). Patients with homozygous familial hypercholesterolemia (FH) who lack LDL receptors do not have remnant accumulation (38). However, mice lacking syndecan-1, a core protein of HSPG, develop remnant accumulation despite intact LDL receptors (35). These studies suggest that HSPGs play a more important role in remnant clearance than the LDL receptor.

 

Accumulation of cholesterol-rich remnant particles leads to their uptake by macrophages, resulting in foam cells found in atherosclerotic lesions and xanthoma. Elevated levels of remnant cholesterol have been associated with an increased risk of cardiovascular disease, with a hazard ratio similar to that of elevated LDL-C (39,40).

 

Besides its role in the uptake and clearance of remnant particles, apo E also modulates lipolytic activity. Elevated levels of apo E can impair triglyceride hydrolysis by displacing or masking apo C-II, a cofactor for lipoprotein lipase, resulting in hypertriglyceridemia (41). In addition, apo E has been shown to stimulate hepatic VLDL production in animals, further increasing circulating triglyceride levels (42). However, evidence in humans is rather limited. In individuals with complete apo E deficiency, hypertriglyceridemia is usually not observed since there is no excess apo E and triglyceride lipolysis is not impaired.

 

Pathogenic variants in the APOE gene play a key role in the pathophysiology of dysbetalipoproteinemia. Most cases of dysbetalipoproteinemia are autosomal recessive, with the majority of affected individuals harboring two e2 alleles. Apo E2 has a binding capacity for the LDL receptor that is only 1–2% of that of apo E3 (43). Notably, the amino acid residue 176 lies outside the critical binding region involved in ionic interaction with lipoprotein receptors. This amino acid change appears to reorganize the salt bridges and alter the conformation of the amino acid residues 154-168, thereby indirectly impairing the receptor binding (44,45). In contrast, apo E2 retains significant binding affinity for HSPGs and the HSPG/LRP (37,46). Therefore, relatively normal binding of apo E2 to HSPG may compensate for defective binding to the LDL receptor, thereby protecting against the development of hyperlipidemia. In fact, most subjects with the E2/E2 phenotype are either normolipidemic or even hypolipidemic (9) and have a reduced risk for coronary artery disease (CAD) (47). Overt hyperlipidemia, also known as type III hyperlipidemia or type III hyperlipoproteinemia, develops only in the presence of additional environmental or genetic factors. These secondary factors may involve conditions associated with overproduction of VLDL or impaired clearance via the LDL receptor or the HSPG/LRP pathways as shown in Table 2. Insulin resistance, for example, is associated with the activation of the SULF2 gene, which encodes sulfatase 2 and causes degradation of HSPGs in mice (48). Thus, the presence of apoE2/E2 is necessary but not sufficient to cause an abnormal lipid profile. In the recessive form of dysbetalipoproteinemia, elevated lipid levels rarely appear before adulthood. Estrogen is known to enhance both LDL receptor expression and the lipolytic process. Therefore, women who are e2/e2 homozygotes are protected against the development of overt hyperlipidemia until after menopause. Additionally, common gene polymorphisms involved in triglyceride metabolism influence susceptibility to overt hyperlipidemia (49).

 

Table 2. Metabolic Conditions Known to Precipitate Hyperlipidemia in Dysbetalipoproteinemia

Lipoprotein overproduction

Impaired clearance

- Insulin resistance

- Type 2 diabetes

- Nephrotic syndrome

- Excess alcohol intake

- Estrogen treatment

- Pregnancy

- High fat diets

- Medications: corticosteroids, retinoids, atypical antipsychotics, antiretrovirals, immunosuppressive drugs

- Increased age

- Menopause

- Hypothyroidism

- Insulin resistance

 

 

In approximately 10% of patients with dysbetalipoproteinemia, the disease is caused by autosomal dominant pathogenic variants in the APOE gene. These rare variants typically involve single amino acid substitutions within the critical binding region of apo E (residues 154–168) that interacts with the LDL receptor, thereby directly impairing receptor binding (50). Other variants disrupt the receptor binding of apo E or result in apo E deficiency. Furthermore, these dominant variants exhibit severely impaired binding to HSPGs. This defective HSPG binding in the dominant form of dysbetalipoproteinemia suggests that normal LDL receptor binding alone is not sufficient to ensure proper clearance of remnant lipoproteins. As a result, the HSPG binding affinity of apo E variants is considered a key determinant of the inheritance pattern of dysbetalipoproteinemia. In the autosomal dominant form, a single allele carrying these variants is sufficient to cause overt hyperlipidemia without the need for secondary factors and lipid abnormalities in these cases presumably begin at birth. To date, approximately 30 APOE variants associated with autosomal dominant dysbetalipoproteinemia have been reported (50-52). Autosomal dominant dysbetalipoproteinemia can occasionally be misdiagnosed as FH (50). The key differences between the autosomal recessive and autosomal dominant forms of dysbetalipoproteinemia are shown in Table 3.

 

Table 3. Characteristics of Autosomal Recessive and Autosomal Dominant Dysbetalipoproteinemia

 

Recessive

Dominant

Presence of b-VLDL

Yes

Yes

Lipoprotein preference of apo E

HDL

Triglyceride-rich lipoproteins

LDL receptor binding

Defective

Defective

HSPG binding

Normal

Defective

Defect in receptor binding

Indirect

Direct

Secondary factors

Required

Not required

Occurrence of hyperlipidemia

Adulthood

From birth

 

Hypercholesterolemia in dysbetalipoproteinemia arises from impaired receptor-mediated clearance of cholesterol-rich remnant lipoproteins, while hypertriglyceridemia results from both impaired lipolytic processing of remnant particles and increased hepatic VLDL production driven by elevated levels of apo E (41,42). Low LDL-cholesterol levels in individuals with dysbetalipoproteinemia are primarily due to impaired conversion of VLDL to IDL, caused by elevated levels of apo E, and reduced hepatic lipase-mediated conversion of IDL to LDL by apo E2. Apo E plays a crucial role in hepatic lipase activity, with apo E3 and apo E4 being more effective than apo E2 in activating hepatic lipase-mediated lipolysis (53,54). Animal studies also suggest that low LDL-cholesterol levels are not due to upregulation of LDL receptors or enhanced hepatic clearance of LDL (41).

 

CLINICAL FEATURES

 

Patients with dysbetalipoproteinemia exhibit variable clinical features. Cutaneous xanthomas, especially palmar xanthoma and tuberous or tuberoeruptive xanthoma, are commonly observed. Palmar xanthoma (or xanthoma striata palmaris) is characterized by yellowish lipid deposits in the palmar creases and is found in 18–72% of patients (figure 2) (17,18,55-57). Although once considered specific to dysbetalipoproteinemia, it is now recognized in other conditions (57). Tuberous xanthoma, frequently found on the knuckles, elbows, knees, and buttocks, may be more common than palmar xanthoma (17,18). Tendon xanthoma is also present in some cases. Neither tuberous xanthoma nor tendon xanthoma is unique to dysbetalipoproteinemia; they can occur in other types of dyslipidemia. These xanthomas typically disappear once lipid levels are brought under control. Several metabolic conditions, including type 2 diabetes, hyperinsulinemia, obesity, hyperuricemia, and hypothyroidism, are associated with dysbetalipoproteinemia, as outlined in Table 2.

 

Figure 2. Palmer xanthoma

In a large cohort collected over a 35-year period in Canada, 524 patients met the gold standard Fredrickson criteria for dysbetalipoproteinemia (plasma triglyceride between 150-1,000 mg/dL and VLDL-cholesterol/triglyceride mass ratio >0.30) (58). However, only 197 subjects (38%) had the apo e2/e2 genotype. This finding contrasts with earlier reports based on a smaller number of subjects, which indicated that 90% of patients with dysbetalipoproteinemia carried the apo e2/e2 genotype. Clinically, patients who met the Fredrickson criteria and had the apo e2/e2 genotype exhibited more severe phenotypes than those without it. These individuals had significantly higher levels of remnant cholesterol, a greater frequency of xanthomas, and a higher prevalence of atherosclerotic cardiovascular disease (ASCVD) (58). Additionally, those with the apo e2/e2 genotype demonstrated an 11-fold increased risk of peripheral artery disease (PAD) compared to those without it. This study suggests that dysbetalipoproteinemia may manifest as a less severe multifactorial remnant cholesterol disease in individuals without the apo e2/e2 genotype and as a more severe form associated with the apo e2/e2 genotype (58).

 

Premature ASCVD, particularly CAD and PAD of the lower extremities, is more common in patients with dysbetalipoproteinemia and elevated lipid levels (17,55,59). The risk of CAD is reported to increase by approximately 5- to 10-fold (59). For PAD, the risk is elevated 13-fold compared to normolipidemic controls and 3-fold compared to patients with FH (60). Factors such as age, hypertension, smoking, waist circumference, triglyceride levels, and a polygenic risk score are significant predictors of cardiovascular disease in these individuals (61,62).

 

A contemporary cross-sectional study of 305 patients with dysbetalipoproteinemia in Europe found CAD in 19% of participants, while PAD was present in 11% (63). Similarly, among 964 patients in the UK Biobank, CAD was identified in 12% and PAD in 3% (23). Notably, as with other genetic lipid disorders, standard risk calculators for estimating the 10-year risk of ASCVD are not applicable, as they tend to underestimate the actual risk.

 

Rare mutations in the APOE gene are associated with lipoprotein glomerulopathy, a condition most commonly reported in Japan. The most frequent mutation identified is APOEc.488G>C (p.Arg163Pro), also known as apoE Sendai (64,65). This disorder is characterized by progressive proteinuria. Histologically, lipoprotein thrombi are observed in markedly dilated glomeruli. Approximately half of the reported cases progress to renal failure.

 

BIOCHEMICAL FEATURES

 

The lipid profile of subjects with dysbetalipoproteinemia is highly variable and sensitive to day-to-day changes in diet (66). Typically, there is an increase in both total cholesterol and triglyceride levels. Plasma triglyceride levels may be within the same range with the total cholesterol levels (cholesterol to triglyceride molar ratio around 2:1) or sometimes higher than total cholesterol levels. Severe hypertriglyceridemia resulting in acute pancreatitis has been reported in some cases of dysbetalipoproteinemia. Although total cholesterol levels are usually elevated, LDL-C levels are almost always reduced (17). The cause of low LDL-C levels in dysbetalipoproteinemia is due to an impairment in the apo E-mediated conversion of remnant lipoproteins to LDL (67). Normally, once apo E on remnant lipoproteins binds to HSPGs on hepatocytes, HSPG-bound hepatic lipase removes triglyceride from these remnants and converts them into LDL. The presence of abnormal apo E2 in dysbetalipoproteinemia appears to impair this process, leading to low levels of LDL-C.

 

Since remnant lipoproteins are enriched in cholesterol with a higher VLDL-cholesterol/triglyceride (VLDL-C/TG) ratio, a fixed ratio of VLDL-C/TG used in the Friedewald formula is invalid. In fact, dysbetalipoproteinemia is listed as one of the exceptions in the original report that the Friedewald formula should not be used (68). VLDL-C levels, calculated by triglyceride/5, are therefore underestimated, leading to overestimation of calculated LDL-C. Calculated LDL-C levels derived from the Friedewald formula or the NIH equation, as well as measured LDL-C levels from a homogeneous direct LDL-C assay, have been shown to overestimate plasma LDL-C levels in patients with dysbetalipoproteinemia (69,70). HDL cholesterol levels are also modestly reduced in subjects with dysbetalipoproteinemia. Apo B levels are typically not markedly elevated. Although Apo E levels are higher in individuals with dysbetalipoproteinemia, there is an overlap with those without the condition (71).

 

Based on lipid phenotypes, dysbetalipoproteinemia should be suspected in the following situations (72).

(1) dyslipidemia patients whose total cholesterol and triglyceride levels are both elevated and approximately equal

(2) mixed hyperlipidemia in which apo B level is relatively low in relation to total cholesterol level

(3) Significant disparity between calculated LDL-C and directly measured LDL-C levels

 

DIAGNOSIS

 

Dysbetalipoproteinemia cannot be diagnosed with a single straightforward test, nor can it be identified solely through conventional lipid values. Historically, diagnosis usually requires a biochemical approach to demonstrate the presence of remnant accumulation in the circulation and a genetic approach to characterize the apo E genotype. The presence of b-VLDL indicates dysbetalipoproteinemia regardless of whether hyperlipidemia is present or not.

 

Lipoprotein electrophoresis is a classical method originally used to characterize different lipoproteins and to classify various types of dyslipidemia. Different lipoprotein families display distinct bands on serum electrophoresis. Using paper, agarose, or cellulose acetate as the media, chylomicron stays at the origin whereas HDL migrates to the most advanced position, which is called an a band. Between the origin and the a band, a b band indicates LDL, whereas a pre-b band represents VLDL. On polyacrylamide gel, however, the migration pattern is slightly different in that VLDL (pre-blipoproteins) migrate behind instead of in front of the LDL (b-lipoproteins) (73).

 

Serum agarose gel electrophoresis has been traditionally used to detect the remnant lipoproteins and to diagnose dysbetalipoproteinemia. On paper, agarose, or cellulose acetate electrophoresis, the demonstration of a broad b band, extending from the b position into the pre-b position, indicates the presence of remnant lipoproteins (74) as shown in Figure 3. However, a broad b band is found in less than half of patients (75) and can be found in other conditions (76), suggesting that the presence of a broad b-band in lipoprotein electrophoresis is neither sensitive nor specific for the diagnosis of dysbetalipoproteinemia. On polyacrylamide gel electrophoresis, the presence of small VLDL and IDL along with the absence of a b-migrating lipoprotein band have also been used to indicate dysbetalipoproteinemia (73,75).

 

Figure 3. Plasma lipoprotein electrophoresis in 0.5% agarose gel demonstrated a broad  band in a patient with dysbetalipoproteinemia (left) and a normal pattern in a normal subject (right) (from (31)).

 

Another method used to characterize different lipoproteins is ultracentrifugation. Using preparative ultracentrifugation to isolate various lipoprotein families, lipid composition of different lipoprotein fractions can then be determined. Compared to the normal pre b-VLDL, b-VLDL remnant particles are more cholesterol-enriched and triglyceride-depleted. Normally, the cholesterol/triglyceride mass ratio in VLDL is 0.2 or less and the cholesterol/triglyceride ratio in b-VLDL is higher. A VLDL-C/VLDL TG mass ratio (both in mg/dL) >0.42 or VLDL-C/VLDL TG molar ratio (both in mmol/L) >0.97 is considered diagnostic of dysbetalipoproteinemia (77). Several studies have also tried to identify cut points for detection of b-VLDL using a VLDL-C/plasma or total TG ratio. The most frequently used cutoff for diagnosis of dysbetalipoproteinemia is the Fredrickson criteria, which is VLDL-C/TG >0.30 (mass ratio) or >0.69 (molar ratio) and plasma triglyceride level between 150-1,000 mg/dL (14). A mass ratio between 0.25-0.30 or a molar ratio between 0.57-0.69 is considered suggestive of dysbetalipoproteinemia (14,78).

 

Both lipoprotein electrophoresis and preparative ultracentrifugation described above are, however, not readily available in routine clinical laboratories. Therefore, other diagnostic methods using common lipid and apolipoprotein levels have been explored.

 

Two methods for estimating the VLDL-C level without a need for ultracentrifugation have recently been described (79). The first method used results obtained from the standard lipid panel and the Sampson-NIH equation. At a cut point of 0.194, a sensitivity of 74% and a specificity of 83% were reported (79). The second method modified the Sampson-NIH equation by including apo B level for predicting VLDL-C. At a cut-point of 0.209, a better sensitivity of 97% and a better specificity of 95% were demonstrated (79).

 

Remnant lipoprotein cholesterol (RLP-C) can be measured in serum and serum RLP-C/triglyceride ratio has been shown to be an effective alternative to VLDL-C/triglyceride ratio (80,81). Serum RLP-C/triglyceride ratio >0.23 is highly correlated with the presence of b-VLDL and has been demonstrated to be useful for screening for dysbetalipoproteinemia (81,82)

 

Since plasma apo B levels are not typically elevated in subjects with dysbetalipoproteinemia, Blom et al. showed that an apo B (in g/L)/total cholesterol (in mmol/L) ratio of <0.15 could identify patients with dysbetalipoproteinemia with a sensitivity of 89% (95% confidence interval [CI] 78-96%) and a specificity of 97% (95% CI 94-98%) among 333 patients with mixed hyperlipidemia with 57 having confirmed dysbetalipoproteinemia (83). In another study of 1,771 patients with various types of dyslipidemia along with 38 confirmed cases of dysbetalipoproteinemia, Sniderman et al. reported that a total cholesterol (in mmol/L)/apo B (in g/L) ratio of >6.2 and a triglyceride/apo B ratio of <10.0 have been shown to discriminate among other types of dyslipidemia based on the Fredrickson classification (84). However, when this method was compared to the ultracentrifugation-based definition of dysbetalipoproteinemia among 3,695 individuals (16 with dysbetalipoproteinemia), a higher prevalence was found (1.43% vs. 0.43%), suggesting that the method of Sniderman et al. using lipids and apo B levels might not be specific (16). When the triglyceride cutoff was raised from 160 mg/dL to 200 mg/dL, the specificity is significantly improved, indicating that triglyceride level is also important in this screening algorithm (16). With increasing levels of triglyceride, more severe cases of dysbetalipoproteinemia may be identified using the apo B algorithm but the sensitivity to detect milder cases drops significantly (85). Similarly, a recent study from Germany using the apo B/total cholesterol ratio as diagnostic criteria proposed by Sniderman et al. (84) or Blom et al. (83), showed that although subjects with the apo e2/e2 genotype were more likely to develop dysbetalipoproteinemia, most subjects with dysbetalipoproteinemia did not have the apo e2/e2 genotype (86). Resequencing of APOE gene further identified only a few cases of rare APOE variants (86). These studies suggest that using only lipid phenotypes and apo B alone without the apo e2 genotype tends to either include more false positive cases or capture milder cases of true dysbetalipoproteinemia (16,21).

 

In addition to using the total cholesterol/apo B ratio as a screening criterion, the non-HDL-cholesterol (non-HDL-C)/apo B ratio has also been examined. A small study in 9 Japanese patients with dysbetalipoproteinemia proposed a non HDL-C/apo B ratio (both in mg/dL) of >2.6 to differentiate from those with combined hyperlipidemia (87), whereas a subsequent larger study in England (n = 1,637) with 63 cases of dysbetalipoproteinemia showed that a non HDL-C (in mmol/L)/apo B (in g/L) ratio of >4.91 had better diagnostic performance than a total cholesterol/apo B ratio (88).

 

A study from Canada has also evaluated different lipid values among 4,891 patients with mixed hyperlipidemia (total cholesterol ³5.2 mmol/L [200 mg/dL] and triglyceride ³2.0 mmol/L [175 mg/dL]), 188 of whom had dysbetalipoproteinemia defined from elevated VLDL-C/plasma TG ratio and the presence of apo e2/e2 genotype (56). In this cohort, Paquette et al. showed that the non-HDL-C/apo B ratio was the best predictor of dysbetalipoproteinemia, which was better than the total cholesterol/apo B ratio (56). The non HDL-C/apo B ratio cut point of 3.69 mmol/g or 1.43 in conventional units (both in mg/dL) followed by the identification of apo e2/e2 genotype provided a good sensitivity (94.8%) and specificity (99%) with 99.4% accuracy (56). A review of previous diagnostic strategies proposed for dysbetalipoproteinemia further demonstrated that all other criteria (16,82-84,87-89) exhibited either low sensitivity or low specificity when validated using this cohort.

 

A combination of non HDL-C/apo B ratio of ³1.7 and TG/apo B ratio of ³1.35, all in mg/dL (non HDL-C in mmol/L/apo B in mg/dL ³4.4 and TG in mmol/L/apo B in mg/dL ³3.5) has recently been proposed as a screening tool for further APOEgenotyping in subjects with TG >150 mg/dL, LDL-C >160 mg/dL or non HDL-C >190 mg/dL (90). This algorithm has been shown to give excellent sensitivity and high specificity compared with other algorithms. Although apo B levels are affected by lipid-lowering therapy, this algorithm has been proposed to be used in those with and without lipid-lowering medications. In the population with lower levels of apo B, however, the algorithm that used non HDL-C/apo B ratio has been shown to give excellent sensitivity but very low specificity for detecting apo e2/e2 genotype (24).

 

More recently, a large study of dysbetalipoproteinemia patients (n=964) from the UK Biobank has been reported (23). Dysbetalipoproteinemia was diagnosed using the apo e2/e2 genotype and mixed hyperlipidemia (total cholesterol ³200 mg/dL [5.2 mmol/l] and triglyceride ³175 mg/dL [2.0 mmol/l]). The performances of 6 different criteria (56,79,83,84,88,90)were evaluated and 3 criteria by Boot et al.(88), Blom et al. (83), and Sniderman et al. (84) showed sensitivity, specificity, and accuracy >90% with the area under the curve (AUC) of ³0.95 and the negative predictive value of 100% (23). The number of those who met the criteria and should be assessed for APOE genotype in these 3 criteria ranged from 1-6%. When the non HDL-C/apo B cutoff ratio originally proposed by Paquette et al. (56) was raised from ³1.43 (in mg/dL) [3.69 (in mmol/g)] to ³1.78 (in mg/dL) [4.61 (in mmol/g)], the sensitivity, specificity, accuracy and the AUC were all improved similar to the 3 criteria, and the number of individuals that should undergo APOE genetic testing was lower from 23% to 3% (23). It is important to note that all of these criteria should be used for screening for further genetic testing and should not be used solely for diagnosis of dysbetalipoproteinemia. All of these screening criteria have very low positive predictive value, meaning that only a few of those who meet the criteria actually have dysbetalipoproteinemia when tested for APOE genotype (23).

 

The description of various criteria proposed for further evaluation for dysbetalipoproteinemia is shown in Table 4.

 

Table 4. Criteria Proposed for Further Evaluation for Dysbetalipoproteinemia

References

Proposed criteria

Apo B assay

Blom et al., 2005 (83)

- apo B (in g/L)/total cholesterol (in mmol/L) <0.15

Sniderman et al., 2007 (84)

- total cholesterol (in mmol/L)/apo B (in g/L) >6.2

- triglyceride (in mmol/L)/apo B (in g/L) <10.0

- triglyceride >75th percentile for age and gender

Murase et al., 2010 (87)

- non-HDL-C/apo B ratio (both in mg/dL) >2.6

Hopkins et al., 2014 (16)

- total cholesterol (in mmol/L)/apo B (in g/L) >6.2

- triglyceride (in mmol/L)/apo B (in g/L) <10.0

- triglyceride >200 mg/dL (>2.3 mmol/L)

Boot et al., 2019 (88)

- total cholesterol >5.0 mmol/L (>193 mg/dL) and triglyceride >1.5 mmol/L (>133 mg/dL)

- non-HDL-C (in mmol/L)/apo B (in g/L) >4.91

Paquette et al., 2020 (56)

- total cholesterol ³5.2 mmol/L (³200 mg/dL) and triglyceride ³2.0 mmol/L (³175 mg/dL)

- non-HDL-C/apo B >3.69 mmol/g or 1.43 (both in mg/dL)

Bea et al, 2023 (90)

- triglyceride >150 mg/dL and LDL-C >160 mg/dL or non-HDL-C >190 mg/dL

- non-HDL-C/apo B ³1.7 (both in mg/dL) or non-HDL-C (in mmol/L) /apo B (in mg/dL) ³4.4

- triglyceride/apo B ³1.35 (both in mg/dL) or triglyceride (in mmol/L) /apo B (in mg/dL) ³3.5

Remnant lipoprotein cholesterol assay

Nakajima et al., 2007 (82)

- RLP-C/triglyceride >0.23

 

Identification of apo E phenotype and/or genotype can help establish the diagnosis of dysbetalipoproteinemia. Nowadays, conventional apo E phenotyping by isoelectric focusing is replaced by a number of simple and more accurate APOE genotyping methods. When apo e2/e2 homozygosity is discovered in subjects with dysbetalipoproteinemia, immediate family members should be screened for the presence of hyperlipidemia. The presence of apo e2/e2 by itself without overt hyperlipidemia is not a critical risk factor for premature ASCVD. Therefore, counseling should be focused on eliminating secondary factors known to cause hyperlipidemia, such as obesity, diabetes, or alcohol consumption.

 

In patients with suspected dysbetalipoproteinemia, if the apo e2/e2 homozygosity is excluded, next generation sequencing can be performed to identify rare APOE variants associated with the autosomal dominant form. Since not all identified variants in the APOE gene are causally related to dysbetalipoproteinemia, a comprehensive approach is advised to determine the pathogenicity of the variants detected using both in vitro and in vivo functional studies (52). In this condition, 50% of first-degree relatives are affected. Therefore, cascade screening should be performed in a manner similar to that for FH. Once the diagnosis is confirmed, appropriate treatment should be initiated.

 

TREATMENT

 

Dysbetalipoproteinemia responds well to therapy (17). However, data from the UK Biobank and the US NHANES cohorts show that the majority of subjects with dysbetalipoproteinemia remain untreated despite their high atherogenic risk (21,23). Dietary modifications and addressing secondary metabolic factors form the cornerstone of therapy. Restriction of caloric intake in those who are overweight and reduction of saturated fat and cholesterol in the diet help normalize lipid levels (18). There are no specific dietary guidelines for patients with dysbetalipoproteinemia (91); however, reducing dietary cholesterol and saturated fat while increasing polyunsaturated fat intake is recommended (18). Weight reduction, glycemic control of diabetes, cessation of alcohol intake, and treatment of hypothyroidism can also lower lipid levels.

 

LDL-C levels cannot be accurately measured or calculated in patients with dysbetalipoproteinemia (70). In addition, LDL-C levels are typically not elevated and do not reflect high cardiovascular risk in these patients. Therefore, LDL-C levels should not be used as a treatment target in dysbetalipoproteinemia. It is recommended that the primary target of treatment is non-HDL-C level (50,73), which can be reliably measured using standard assays of total cholesterol and HDL-C (70). The secondary target of treatment is triglyceride level. In some cases, medications are required to lower cholesterol and triglyceride levels, and statins and fibrates are the medications of choice, respectively. Evolocumab, a PCSK9 inhibitor, has also been shown to reduce total cholesterol, remnant cholesterol, and triglyceride levels in patients with dysbetalipoproteinemia (92,93). Resolution in xanthomas and regression of atherosclerotic lesions have been observed after normalization of lipid levels (94).

 

CONCLUSION

 

Dysbetalipoproteinemia remains an underrecognized genetic lipid disorder. Pathogenic variants in the APOE gene lead to defective apo E-mediated remnant clearance and accumulation of remnant lipoproteins characterized by elevation of both total cholesterol and triglyceride levels, palmar and tuberous xanthomas, and an increased risk of CAD and PAD. The HSPG-binding affinity of the apo E variants appears to be an important determinant of the different modes of inheritance. Historically, diagnosis requires sophisticated methods to demonstrate the presence of remnant lipoprotein particles (b-VLDL) and the pathogenic variants in the APOE gene. Currently, a simple diagnostic test for dysbetalipoproteinemia does not exist and several algorithms using various lipid and apo B parameters have been proposed for screening for this condition and further genetic testing. Recent data suggest that the phenotype of dysbetalipoproteinemia may be more heterogeneous and a milder form of dysbetalipoproteinemia without the apo e2/e2 genotype is called multifactorial remnant cholesterol disease to differentiate it from the more severe form in those with apo e2/e2 genotype. Nevertheless, subjects with dysbetalipoproteinemia are usually responsive to lifestyle modifications and conventional lipid-modifying therapy, including statins and fibrates. Despite renewed interest and recent advances in understanding this condition, several knowledge gaps remain. These include the precise mechanisms involved in the clearance of remnant lipoproteins, the true prevalence within the general population, the roles of genetic and environmental factors in modifying disease expression, the underlying mechanisms of PAD involvement, the development of a simplified diagnostic test for clinical use, the establishment of standard guidelines for screening, and the creation of evidence-based guidelines for optimal treatment and cardiovascular risk reduction.

 

REFERENCES

 

  1. McGinley J, Jones H, Gofman J. Lipoproteins and xanthomatous diseases. J Invest Dermatol. 1952;19(1):71-82.
  2. Fredrickson DS, Levy RI, Lindgren FT. A comparison of heritable abnormal lipoprotein patterns as defined by two different techniques. J Clin Invest. 1968;47(11):2446-2457.
  3. Quarfordt S, Levy RI, Fredrickson DS. On thelipoprotein abnormality in type 3 hyperlipoproteinemia. J Clin Invest.1971;50(4):754-761.
  4. Fredrickson DS, Levy RI, Lees RS. Fat transport in lipoproteins--an integrated approach to mechanisms and disorders. N Engl J Med. 1967;276(4):215-225.
  5. Kane JP, Chen GC, Hamilton RL, Hardman DA, Malloy MJ, Havel RJ. Remnants of lipoproteins of intestinal and hepatic origin in familial dysbetalipoproteinemia. Arteriosclerosis. 1983;3(1):47-56.
  6. Chait A, Brunzell JD, Albers JJ, Hazzard WR. Type-III Hyperlipoproteinaemia ("remnant removal disease"). Insight into the pathogenetic mechanism. Lancet. 1977;1(8023):1176-1178.
  7. Havel RJ, Kane JP. Primary dysbetalipoproteinemia: predominance of a specific apoprotein species in triglyceride-rich lipoproteins. Proc Natl Acad Sci U S A. 1973;70(7):2015-2019.
  8. Utermann G, Jaeschke M, Menzel J. Familial hyperlipoproteinemia type III: deficiency of a specific apolipoprotein (apo E-III) in the very-low-density lipoproteins. FEBS Lett. 1975;56(2):352-355.
  9. Utermann G, Hees M, Steinmetz A. Polymorphism of apolipoprotein E and occurrence of dysbetalipoproteinaemia in man. Nature. 1977;269(5629):604-607.
  10. Zannis VI, Breslow JL. Characterization of a unique human apolipoprotein E variant associated with type III hyperlipoproteinemia. J Biol Chem. 1980;255(5):1759-1762.
  11. Rall SC, Jr., Weisgraber KH, Mahley RW. Human apolipoprotein E. The complete amino acid sequence. J Biol Chem. 1982;257(8):4171-4178.
  12. Windler E, Chao Y, Havel RJ. Determinants of hepatic uptake of triglyceride-rich lipoproteins and their remnants in the rat. J Biol Chem. 1980;255(11):5475-5480.
  13. Havel RJ, Chao Y, Windler EE, Kotite L, Guo LS. Isoprotein specificity in the hepatic uptake of apolipoprotein E and the pathogenesis of familial dysbetalipoproteinemia. Proc Natl Acad Sci U S A. 1980;77(7):4349-4353.
  14. Fredrickson DS, Morganroth J, Levy RI. Type III hyperlipoproteinemia: an analysis of two contemporary definitions. Ann Intern Med. 1975;82(2):150-157.
  15. LaRosa JC, Chambless LE, Criqui MH, Frantz ID, Glueck CJ, Heiss G, Morrison JA. Patterns of dyslipoproteinemia in selected North American populations. The Lipid Research Clinics Program Prevalence Study. Circulation. 1986;73(1 Pt 2):I12-29.
  16. Hopkins PN, Brinton EA, Nanjee MN. Hyperlipoproteinemia type 3: the forgotten phenotype. Curr Atheroscler Rep.2014;16(9):440.
  17. Morganroth J, Levy RI, Fredrickson DS. The biochemical, clinical, and genetic features of type III hyperlipoproteinemia. Ann Intern Med. 1975;82(2):158-174.
  18. Brewer HB, Jr., Zech LA, Gregg RE, Schwartz D, Schaefer EJ. NIH conference. Type III hyperlipoproteinemia: diagnosis, molecular defects, pathology, and treatment. Ann Intern Med. 1983;98(5 Pt 1):623-640.
  19. Wood PD, Stern MP, Silvers A, Reaven GM, von der Groeben J. Prevalence of plasma lipoprotein abnormalities in a free-living population of the Central Valley, California. Circulation. 1972;45(1):114-126.
  20. Gibson TC, Whorton EB. The prevalence of hyperlipidemia in a natural community. J Chronic Dis. 1973;26(4):227-236.
  21. Pallazola VA, Sathiyakumar V, Park J, Vakil RM, Toth PP, Lazo-Elizondo M, Brown E, Quispe R, Guallar E, Banach M, Blumenthal RS, Jones SR, Marais D, Soffer D, Sniderman AD, Martin SS. Modern prevalence of dysbetalipoproteinemia (Fredrickson-Levy-Lees type III hyperlipoproteinemia). Arch Med Sci. 2020;16(5):993-1003.
  22. de Beer F, Stalenhoef AF, Hoogerbrugge N, Kastelein JJ, Gevers Leuven JA, van Duijn CM, Havekes LM, Smelt AH. Expression of type III hyperlipoproteinemia in apolipoprotein E2 (Arg158 --> Cys) homozygotes is associated with hyperinsulinemia. Arterioscler Thromb Vasc Biol. 2002;22(2):294-299.
  23. Paquette M, Trinder M, Guay SP, Brunham LR, Baass A. Prevalence of Dysbetalipoproteinemia in the UK Biobank According to Different Diagnostic Criteria. J Clin Endocrinol Metab. 2024.
  24. Blokhina AV, Ershova AI, Kiseleva AV, Sotnikova EA, Zharikova AA, Zaicenoka M, Vyatkin YV, Ramensky VE, Kutsenko VA, Shalnova SA, Meshkov AN, Drapkina OM. Applicability of Diagnostic Criteria and High Prevalence of Familial Dysbetalipoproteinemia in Russia: A Pilot Study. Int J Mol Sci. 2023;24(17).
  25. Beheshti SO, Madsen CM, Varbo A, Nordestgaard BG. Worldwide Prevalence of Familial Hypercholesterolemia: Meta-Analyses of 11 Million Subjects. J Am Coll Cardiol. 2020;75(20):2553-2566.
  26. Mahley RW. Apolipoprotein E: cholesterol transport protein with expanding role in cell biology. Science.1988;240(4852):622-630.
  27. Weisgraber KH, Rall SC, Jr., Mahley RW. Human E apoprotein heterogeneity. Cysteine-arginine interchanges in the amino acid sequence of the apo-E isoforms. J Biol Chem. 1981;256(17):9077-9083.
  28. Hanlon CS, Rubinsztein DC. Arginine residues at codons 112 and 158 in the apolipoprotein E gene correspond to the ancestral state in humans. Atherosclerosis. 1995;112(1):85-90.
  29. Abou Ziki MD, Strulovici-Barel Y, Hackett NR, Rodriguez-Flores JL, Mezey JG, Salit J, Radisch S, Hollmann C, Chouchane L, Malek J, Zirie MA, Jayyuosi A, Gotto AM, Jr., Crystal RG. Prevalence of the apolipoprotein E Arg145Cys dyslipidemia at-risk polymorphism in African-derived populations. Am J Cardiol. 2014;113(2):302-308.
  30. Ghiselli G, Schaefer EJ, Gascon P, Breser HB, Jr. Type III hyperlipoproteinemia associated with apolipoprotein E deficiency. Science. 1981;214(4526):1239-1241.
  31. Schaefer EJ, Gregg RE, Ghiselli G, Forte TM, Ordovas JM, Zech LA, Brewer HB, Jr. Familial apolipoprotein E deficiency. J Clin Invest. 1986;78(5):1206-1219.
  32. Mahley RW, Ji ZS. Remnant lipoprotein metabolism: key pathways involving cell-surface heparan sulfate proteoglycans and apolipoprotein E. J Lipid Res. 1999;40(1):1-16.
  33. Williams KJ, Chen K. Recent insights into factors affecting remnant lipoprotein uptake. Curr Opin Lipidol.2010;21(3):218-228.
  34. Fuki IV, Kuhn KM, Lomazov IR, Rothman VL, Tuszynski GP, Iozzo RV, Swenson TL, Fisher EA, Williams KJ. The syndecan family of proteoglycans. Novel receptors mediating internalization of atherogenic lipoproteins in vitro. J Clin Invest. 1997;100(6):1611-1622.
  35. Stanford KI, Bishop JR, Foley EM, Gonzales JC, Niesman IR, Witztum JL, Esko JD. Syndecan-1 is the primary heparan sulfate proteoglycan mediating hepatic clearance of triglyceride-rich lipoproteins in mice. J Clin Invest.2009;119(11):3236-3245.
  36. Out R, Kruijt JK, Rensen PC, Hildebrand RB, de Vos P, Van Eck M, Van Berkel TJ. Scavenger receptor BI plays a role in facilitating chylomicron metabolism. J Biol Chem. 2004;279(18):18401-18406.
  37. Ji ZS, Fazio S, Mahley RW. Variable heparan sulfate proteoglycan binding of apolipoprotein E variants may modulate the expression of type III hyperlipoproteinemia. J Biol Chem. 1994;269(18):13421-13428.
  38. Rubinsztein DC, Cohen JC, Berger GM, van der Westhuyzen DR, Coetzee GA, Gevers W. Chylomicron remnant clearance from the plasma is normal in familial hypercholesterolemic homozygotes with defined receptor defects. J Clin Invest. 1990;86(4):1306-1312.
  39. Varbo A, Benn M, Nordestgaard BG. Remnant cholesterol as a cause of ischemic heart disease: evidence, definition, measurement, atherogenicity, high risk patients, and present and future treatment. Pharmacol Ther.2014;141(3):358-367.
  40. Varbo A, Freiberg JJ, Nordestgaard BG. Extreme nonfasting remnant cholesterol vs extreme LDL cholesterol as contributors to cardiovascular disease and all-cause mortality in 90000 individuals from the general population. Clin Chem. 2015;61(3):533-543.
  41. Huang Y, Liu XQ, Rall SC, Jr., Mahley RW. Apolipoprotein E2 reduces the low density lipoprotein level in transgenic mice by impairing lipoprotein lipase-mediated lipolysis of triglyceride-rich lipoproteins. J Biol Chem.1998;273(28):17483-17490.
  42. Huang Y, Liu XQ, Rall SC, Jr., Taylor JM, von Eckardstein A, Assmann G, Mahley RW. Overexpression and accumulation of apolipoprotein E as a cause of hypertriglyceridemia. J Biol Chem. 1998;273(41):26388-26393.
  43. Weisgraber KH, Innerarity TL, Mahley RW. Abnormal lipoprotein receptor-binding activity of the human E apoprotein due to cysteine-arginine interchange at a single site. J Biol Chem. 1982;257(5):2518-2521.
  44. Innerarity TL, Weisgraber KH, Arnold KS, Rall SC, Jr., Mahley RW. Normalization of receptor binding of apolipoprotein E2. Evidence for modulation of the binding site conformation. J Biol Chem. 1984;259(11):7261-7267.
  45. Wilson C, Mau T, Weisgraber KH, Wardell MR, Mahley RW, Agard DA. Salt bridge relay triggers defective LDL receptor binding by a mutant apolipoprotein. Structure. 1994;2(8):713-718.
  46. Kowal RC, Herz J, Weisgraber KH, Mahley RW, Brown MS, Goldstein JL. Opposing effects of apolipoproteins E and C on lipoprotein binding to low density lipoprotein receptor-related protein. J Biol Chem. 1990;265(18):10771-10779.
  47. Bennet AM, Di Angelantonio E, Ye Z, Wensley F, Dahlin A, Ahlbom A, Keavney B, Collins R, Wiman B, de Faire U, Danesh J. Association of apolipoprotein E genotypes with lipid levels and coronary risk. JAMA. 2007;298(11):1300-1311.
  48. Chen K, Liu ML, Schaffer L, Li M, Boden G, Wu X, Williams KJ. Type 2 diabetes in mice induces hepatic overexpression of sulfatase 2, a novel factor that suppresses uptake of remnant lipoproteins. Hepatology.2010;52(6):1957-1967.
  49. Satny M, Todorovova V, Altschmiedova T, Hubacek JA, Dlouha L, Lanska V, Soska V, Kyselak O, Freiberger T, Bobak M, Vrablik M. Genetic risk score in patients with the APOE2/E2 genotype as a predictor of familial dysbetalipoproteinemia. J Clin Lipidol. 2024;18(2):e230-e237.
  50. Koopal C, Marais AD, Westerink J, Visseren FL. Autosomal dominant familial dysbetalipoproteinemia: A pathophysiological framework and practical approach to diagnosis and therapy. J Clin Lipidol. 2017;11(1):12-23 e11.
  51. Khalil YA, Rabes JP, Boileau C, Varret M. APOE gene variants in primary dyslipidemia. Atherosclerosis.2021;328:11-22.
  52. Heidemann BE, Koopal C, Baass A, Defesche JC, Zuurbier L, Mulder MT, Roeters van Lennep JE, Riksen NP, Boot C, Marais AD, Visseren FLJ. Establishing the relationship between familial dysbetalipoproteinemia and genetic variants in the APOE gene. Clin Genet. 2022;102(4):253-261.
  53. Thuren T, Weisgraber KH, Sisson P, Waite M. Role of apolipoprotein E in hepatic lipase catalyzed hydrolysis of phospholipid in high-density lipoproteins. Biochemistry. 1992;31(8):2332-2338.
  54. Ehnholm C, Mahley RW, Chappell DA, Weisgraber KH, Ludwig E, Witztum JL. Role of apolipoprotein E in the lipolytic conversion of beta-very low density lipoproteins to low density lipoproteins in type III hyperlipoproteinemia. Proc Natl Acad Sci U S A. 1984;81(17):5566-5570.
  55. Blom DJ, Byrnes P, Jones S, Marais AD. Dysbetalipoproteinaemia--clinical and pathophysiological features. S Afr Med J. 2002;92(11):892-897.
  56. Paquette M, Bernard S, Blank D, Pare G, Baass A. A simplified diagnosis algorithm for dysbetalipoproteinemia. J Clin Lipidol. 2020;14(4):431-437.
  57. Roy N, Gaudet D, Brisson D. Palmar striated xanthomas in clinical practice. J Endocr Soc. 2022;6(8):bvac103.
  58. Paquette M, Bernard S, Pare G, Baass A. Dysbetalipoproteinemia: Differentiating Multifactorial Remnant Cholesterol Disease From Genetic ApoE Deficiency. J Clin Endocrinol Metab. 2022;107(2):538-548.
  59. Hopkins PN, Wu LL, Hunt SC, Brinton EA. Plasma triglycerides and type III hyperlipidemia are independently associated with premature familial coronary artery disease. J Am Coll Cardiol. 2005;45(7):1003-1012.
  60. Paquette M, Bernard S, Baass A. Dysbetalipoproteinemia Is Associated With Increased Risk of Coronary and Peripheral Vascular Disease. J Clin Endocrinol Metab. 2022;108(1):184-190.
  61. Paquette M, Bernard S, Pare G, Baass A. Triglycerides, hypertension, and smoking predict cardiovascular disease in dysbetalipoproteinemia. J Clin Lipidol. 2020;14(1):46-52.
  62. Paquette M, Trinder M, Guay SP, Brunham LR, Baass A. Predictors of cardiovascular disease in individuals with dysbetalipoproteinemia: a prospective study in the UK Biobank. J Clin Endocrinol Metab. 2024.
  63. Koopal C, Retterstol K, Sjouke B, Hovingh GK, Ros E, de Graaf J, Dullaart RP, Bertolini S, Visseren FL. Vascular risk factors, vascular disease, lipids and lipid targets in patients with familial dysbetalipoproteinemia: a European cross-sectional study. Atherosclerosis. 2015;240(1):90-97.
  64. Saito T, Oikawa S, Sato H, Sato T, Ito S, Sasaki J. Lipoprotein glomerulopathy: significance of lipoprotein and ultrastructural features. Kidney Int Suppl. 1999;71:S37-41.
  65. Oikawa S, Matsunaga A, Saito T, Sato H, Seki T, Hoshi K, Hayasaka K, Kotake H, Midorikawa H, Sekikawa A, Hara S, Abe K, Toyota T, Jingami H, Nakamura H, Sasaki J. Apolipoprotein E Sendai (arginine 145-->proline): a new variant associated with lipoprotein glomerulopathy. J Am Soc Nephrol. 1997;8(5):820-823.
  66. Sniderman AD, de Graaf J, Thanassoulis G, Tremblay AJ, Martin SS, Couture P. The spectrum of type III hyperlipoproteinemia. J Clin Lipidol. 2018;12(6):1383-1389.
  67. Mahley RW, Huang Y, Rall SC, Jr. Pathogenesis of type III hyperlipoproteinemia (dysbetalipoproteinemia). Questions, quandaries, and paradoxes. J Lipid Res. 1999;40(11):1933-1949.
  68. Friedewald WT, Levy RI, Fredrickson DS. Estimation of the concentration of low-density lipoprotein cholesterol in plasma, without use of the preparative ultracentrifuge. Clin Chem. 1972;18(6):499-502.
  69. Higgins V, Leiter LA, Delaney SR, Beriault DR. Validating the NIH LDL-C equation in a specialized lipid cohort: Does it add up? Clin Biochem. 2022;99:60-68.
  70. Heidemann BE, Koopal C, Roeters van Lennep JE, Stroes ES, Riksen NP, Mulder MT, van Vark-van der Zee LC, Blackhurst DM, Visseren FLJ, Marais AD. Low-density lipoprotein cholesterol and non-high-density lipoprotein cholesterol measurement in Familial Dysbetalipoproteinemia. Clin Chim Acta. 2023;539:114-121.
  71. Todo Y, Kobayashi J, Higashikata T, Kawashiri M, Nohara A, Inazu A, Koizumi J, Mabuchi H. Detailed analysis of serum lipids and lipoproteins from Japanese type III hyperlipoproteinemia with apolipoprotein E2/2 phenotype. Clin Chim Acta. 2004;348(1-2):35-40.
  72. Marais AD, Solomon GA, Blom DJ. Dysbetalipoproteinaemia: a mixed hyperlipidaemia of remnant lipoproteins due to mutations in apolipoprotein E. Crit Rev Clin Lab Sci. 2014;51(1):46-62.
  73. Koopal C, Marais AD, Visseren FL. Familial dysbetalipoproteinemia: an underdiagnosed lipid disorder. Curr Opin Endocrinol Diabetes Obes. 2017;24(2):133-139.
  74. Beaumont JL, Carlson LA, Cooper GR, Fejfar Z, Fredrickson DS, Strasser T. Classification of hyperlipidaemias and hyperlipoproteinaemias. Bull World Health Organ. 1970;43(6):891-915.
  75. Blom DJ, Byrnes P, Jones S, Marais AD. Non-denaturing polyacrylamide gradient gel electrophoresis for the diagnosis of dysbetalipoproteinemia. J Lipid Res. 2003;44(1):212-217.
  76. Boot CS, Luvai A, Neely RDG. The clinical and laboratory investigation of dysbetalipoproteinemia. Crit Rev Clin Lab Sci. 2020;57(7):458-469.
  77. Hazzard WR, Porte D, Jr., Bierman EL. Abnormal lipid composition of very low density lipoproteins in diagnosis of broad-beta disease (type 3 hyperlipoproteinemia). Metabolism. 1972;21(11):1009-1019.
  78. Mishkel MA, Nazir DJ, Crowther S. A longitudinal assessment of lipid ratios in the diagnosis of type III hyperlipoproteinaemia. Clin Chim Acta. 1975;58(2):121-136.
  79. Sampson M, Wolska A, Meeusen JW, Donato LJ, Jaffe AS, Remaley AT. Identification of dysbetalipoproteinemia by an enhanced Sampson-NIH equation for very low-density lipoprotein-cholesterol. Front Genet. 2022;13:935257.
  80. Nakajima K, Saito T, Tamura A, Suzuki M, Nakano T, Adachi M, Tanaka A, Tada N, Nakamura H, Murase T. A new approach for the detection of type III hyperlipoproteinemia by RLP-cholesterol assay. J Atheroscler Thromb.1994;1(1):30-36.
  81. Wang T, Nakajima K, Leary ET, Warnick GR, Cohn JS, Hopkins PN, Wu LL, Cilla DD, Zhong J, Havel RJ. Ratio of remnant-like particle-cholesterol to serum total triglycerides is an effective alternative to ultracentrifugal and electrophoretic methods in the diagnosis of familial type III hyperlipoproteinemia. Clin Chem. 1999;45(11):1981-1987.
  82. Nakajima K, Daimon M, Kamiyama K, Takanashi K, Suzuki Y, Watanabe M, Kubono K, Saniabadi AR, Takashima S, Sakurabayashi I. Serum remnant lipoprotein cholesterol/triglyceride ratio as an index for screening familial type III hyperlipidaemia. Ann Clin Biochem. 2007;44(Pt 4):353-359.
  83. Blom DJ, O'Neill FH, Marais AD. Screening for dysbetalipoproteinemia by plasma cholesterol and apolipoprotein B concentrations. Clin Chem. 2005;51(5):904-907.
  84. Sniderman A, Tremblay A, Bergeron J, Gagne C, Couture P. Diagnosis of type III hyperlipoproteinemia from plasma total cholesterol, triglyceride, and apolipoprotein B. J Clin Lipidol. 2007;1(4):256-263.
  85. Varghese B, Park J, Chew E, Sajja A, Brownstein A, Pallazola VA, Sathiyakumar V, Jones SR, Sniderman AD, Martin SS. Importance of the triglyceride level in identifying patients with a Type III Hyperlipoproteinemia phenotype using the ApoB algorithm. J Clin Lipidol. 2021;15(1):104-115 e109.
  86. Evans D, Beil FU, Aberle J. Resequencing the APOE gene reveals that rare mutations are not significant contributory factors in the development of type III hyperlipidemia. J Clin Lipidol. 2013;7(6):671-674.
  87. Murase T, Okubo M, Takeuchi I. Non-HDL-cholesterol/apolipoprotein B ratio: a useful distinguishing feature in the screening for type III hyperlipoproteinemia. J Clin Lipidol. 2010;4(2):99-104.
  88. Boot CS, Middling E, Allen J, Neely RDG. Evaluation of the non-HDL cholesterol to apolipoprotein B ratio as a screening test for dysbetalipoproteinemia. Clin Chem. 2019;65(2):313-320.
  89. de Graaf J, Couture P, Sniderman A. A diagnostic algorithm for the atherogenic apolipoprotein B dyslipoproteinemias. Nat Clin Pract Endocrinol Metab. 2008;4(11):608-618.
  90. Bea AM, Cenarro A, Marco-Bened V, Laclaustra M, Martn C, Ibarretxe D, Pint X, Arrobas T, Vials C, Civeira F, Olmos S. Diagnosis of Familial Dysbetalipoproteinemia Based on the Lipid Abnormalities Driven by APOE2/E2 Genotype. Clin Chem. 2023;69(2):140-148.
  91. Paquette M, Blais C, Fortin A, Bernard S, Baass A. Dietary recommendations for dysbetalipoproteinemia: A need for better evidence. J Clin Lipidol. 2023;17(4):549-556.
  92. Waldmann E, Wu L, Busygina K, Altenhofer J, Henze K, Folwaczny A, Parhofer KG. Effect of PCSK9 inhibition with evolocumab on lipoprotein subfractions in familial dysbetalipoproteinemia (type III hyperlipidemia). PLoS One.2022;17(3):e0265838.
  93. Heidemann BE, Koopal C, Roeters van Lennep JE, Stroes ESG, Riksen NP, Mulder MT, van der Zee L, Blackhurst DM, Marais AD, Visseren FLJ. Effect of evolocumab on fasting and post fat load lipids and lipoproteins in familial dysbetalipoproteinemia. J Clin Lipidol. 2023;17(1):112-123.
  94. Cho EJ, Min YJ, Oh MS, Kwon JE, Kim JE, Kim CJ. Disappearance of angina pectoris by lipid-lowering in type III hyperlipoproteinemia. Am J Cardiol. 2011;107(5):793-796.

 

Diabetes and Depression

ABSTRACT  

 

Depression is characterized by a disturbance of mood, affecting between 3–5% of the general population at any one time. The prevalence of depression is approximately doubled in people with diabetes compared to the general population, with similar rates between type 1 diabetes and type 2 diabetes. Although many cases of depression are coincidental to the presence of diabetes, certain diabetes factors including diabetes-related complications, diabetes treatments and obesity are associated with an increased risk of depression. There is a bi-directional relationship between diabetes and depression with specific disease and treatment factors explaining why diabetes pre-disposes to depression and vice versa. Genetics, early intra-uterine development, and social determinants of health may create a “common soil” for both conditions. The presence of depression in people with diabetes worsens both diabetes and depression outcomes. Mortality is increased, quality of life diminished, and healthcare costs are increased. Diabetes self-management is also impaired. It may be possible to reduce the incidence of depression in people with diabetes by considering the way in which the diagnosis of diabetes is conveyed and the psychosocial support that is given through an individual’s journey with diabetes. Several short screening questionnaires have been validated in people with diabetes. A diagnosis should be confirmed by a diagnostic interview. The main aims of treatment are to improve both diabetes and mental health outcomes with complete remission of depressive symptoms.  Various psychological treatments, including cognitive behavioral therapy, problem-solving, and psychodynamic techniques have been used to treat depression in people with diabetes. Antidepressants reduce depressive symptoms in people with diabetes as well as the general population. All antidepressants appear to have similar effects on depressive symptoms as long as adequate doses are used. Treatment should be maintained for at least 4–6 months after remission of symptoms to reduce the risk of relapse and recurrence. The choice of antidepressant depends largely on the side-effect profile, individual preference, and response. Selective serotonin reuptake inhibitors are widely used as first-choice agents. A common model of care for depression is the Stepped Care Model which is designed to provide a rational approach to the treatment of depression, while reducing costs and side effects of antidepressants through more appropriate prescribing. A case management model known as collaborative care is a clinical- and cost-effective treatment of depression that also improves diabetes outcomes by involving a multidisciplinary team that works together to identify and treat depression within primary care settings. Although diabetes and depression remain a considerable clinical challenge, there are grounds for considerable optimism as the scientific knowledge that underpins clinical practices has expanded markedly in the last two decades. However, further research is needed to understand what can be done to prevent depression in people with diabetes and to identify the optimal treatment for an individual that improves both depressive symptoms and diabetes outcomes.

 

INTRODUCTION

 

The importance of considering the interaction between mind and body in the management and outcome of chronic diseases has been recognized for over 2,500 years but is particularly poignant for people with diabetes. Diabetes places high behavioral demands on those living with the condition whilst mental disorders, such as depression, may compromise an individual’s ability to perform the self-care needed to optimize health and prevent the long-term consequences of diabetes. Much of diabetes care is focused around the identification and treatment of long-term diabetes-related complications and diabetes healthcare professionals are adept in managing microvascular and macrovascular conditions. An appreciation, however, of the psychological consequences of diabetes has lagged behind, despite these being common and the morbidity, mortality, and health costs associated with the co-morbidity being disproportionately increased compared with the effects of either condition alone (1, 2).

 

Several factors contribute to the poorer health outcomes seen in people with diabetes and mental disorders. Despite the increased burden of disease, people with co-morbid mental disorders have often been disadvantaged by health services and have received sub-optimal medical care (3). They have been less likely to receive the necessary diabetes processes of care, self-management education, and cardiovascular preventative medication despite increased visits to their primary healthcare teams and despite similar interest in caring for their physical illness as the general population (4). Clinicians may fail to recognize that those with mental illness are more likely to develop physical illnesses and so physical complaints are either ignored or not taken seriously. Mental health symptoms often “over-shadow” other complaints leading healthcare professionals to focus on the mental illness to the detriment of any physical illness (5). Health services are often poorly configured with clinics focusing on either the treatment of physical illnesses or mental disorders rather than treating both conditions at the same time (3). The stigma associated with mental illness may prevent people and their families from seeking help for mental illness, thereby depriving them of effective treatments, which not only harms mental well-being but is detrimental to diabetes management (6).

 

Diabetes and mental disorders are both common, and so a degree of co-occurrence would be expected purely by chance. The evidence suggests, however, that diabetes is more frequently associated with a range of mental and psychosocial disorders than expected (7, 8). Furthermore, several mental disorders, including depression, are associated with an increased risk of developing diabetes. An understanding of this complex interaction is crucial to the management and outcome of people with diabetes.

 

This chapter focuses on the co-morbidity of diabetes and depression; both are common, are relatively easy to diagnose, and have established effective treatments (8). They may also serve as an exemplar for other mental disorders and provide a model for the successful management of other co-morbid mental and physical health conditions. The chapter will describe the epidemiology of diabetes and depression and will discuss the mechanisms underlying the association between diabetes and depression. It will also highlight the clinical implications and consequences of co-occurring diabetes and depression. Diabetes healthcare professionals need to be aware of how to screen for depression and to provide “first response” management, while recognizing when to refer for specialist psychiatric care (9).

 

DEPRESSION

 

Depression belongs to a group of mental disorders where the primary abnormality is a disturbance of mood. The cardinal features of depression are low mood, and loss of interest or pleasure, lasting longer than two weeks. During a depressive episode, other symptoms may include poor concentration, feelings of excessive guilt or low self-worth, hopelessness about the future, thoughts about dying or suicide, disrupted sleep, changes in appetite or weight, and feeling especially tired or low in energy. People with depression are at an increased risk of suicide. Depressive symptoms exist on a continuum of severity, and it is important to recognize that depression is different from usual fluctuations in mood that occur with everyday life. 

 

Major depressive disorder (also known as clinical depression, unipolar depression, or major depression) is defined by the diagnostic criteria of the American Psychiatric Association Diagnostic and Statistical Manual of Mental Disorders(DSM-5), based on the number and duration of symptoms (Table 1) (10). The DSM-5 definition approximates a level of severity of symptoms that is associated with both disability and dysfunction. DSM-5 also recognizes several sub-types of depressive disorder where the symptoms are less severe but nevertheless may still compromise diabetes self-care and outcomes.

 

Table 1. DSM-5 “Major” Depressive Episode

A.        Five (or more) of the following symptoms have been present during the same 2-week period and represent a change from previous functioning; at least one of the symptoms is either (1) depressed mood, or (2) loss of interest or pleasure.

•           Depressed mood most of the day, nearly every day, as indicated by either subjective report (e.g. feels sad or empty) or observation made by others (e.g. appears tearful).

•           Markedly diminished interest or pleasure in all, or almost all, activities most of the day, nearly every day (as indicated by either subjective account or observation made by others).

•           Significant weight loss when not dieting or weight gain (e.g. a change of >5% of body weight in a month) or decrease or increase in appetite nearly every day.

•           Insomnia or hypersomnia nearly every day.

•           Psychomotor agitation or retardation nearly every day (observable by others, not merely subjective feelings of restlessness or being slowed down).

•           Fatigue or loss of energy nearly every day.

•           Feelings of worthlessness or excessive or inappropriate guilt (which may be delusional) nearly every day (not merely self-reproach or guilt about being sick).

•           Diminished ability to think or concentrate, or indecisiveness, nearly every day (either by subjective account or as observed by others).

•           Recurrent thoughts of death (not just fear of dying), recurrent suicidal ideation without a specific plan, or a suicide attempt or a specific plan for committing suicide.

B.    B.        The symptoms cause clinically significant distress or impairment in social, occupational, or other important areas of functioning.

C.    C.        The symptoms are not due to the direct physiological effects of a substance (e.g. a drug of abuse, a medication) or a general medical condition (e.g. hypothyroidism).

 

Epidemiology of Depression

 

Depression is one of the commonest mental disorders, affecting between 3–5% of the general population at any one time; the lifetime prevalence is 8-12% but there is considerable geographical variation ranging from 3% in Japan to 17% in India. The highest rates of depression are found in the United States of America (USA), the Middle East and South Asia (11). In 2019, 280 million people were living with depression, including 23 million children and adolescents (12). The incidence of depression worldwide increased by 50% between 1990 and 2017 to an age standardized rate of 3.25 per 1000 (13). Depressive disorders are now ranked as the single largest contributor to non-fatal health loss (7.5% of all years lost to disability) (14).

 

Depression can affect any age group, with depression being seen in infants as young as 6 months old after separation from their mothers (15). The commonest age of onset is between the ages of 30 and 40 years, but there is a second, smaller peak in incidence between ages 50 and 60 years (16). The risk of depression is doubled in women compared with men (17). Although the cause of depression remains uncertain, other risk factors for a depressive episode are recognized and include a family history of depression, certain personality types, childhood adversity, the postpartum period, social isolation, and a lack of close interpersonal relationships (18). An episode of depression is often triggered by a stressful life event, particularly for the first few depressive episodes (Table 2). Depression may accompany other mental illnesses and long-term physical conditions (19). All these risk factors apply to people living with diabetes and so consequently much of the depression seen in people living with diabetes is coincidental to the diabetes.

 

Table 2. Risk Factors for Depression

·       Female gender

·       Age

·       Family History

o   Genetics

o   Shared Environment

·       Personality type

o   Introversion

o   Neuroticism

§  Insecure, worried

§  Obsessive

o   Unassertive, dependent

o   Low conscientiousness

o   Disorganization

o   Stress-sensitive

·       Childhood adversity

o   Separation

o   Neglect

o   Abuse

§  Mental

§  Physical

§  Sexual

o   Early bereavement

o   Unequal parental treatment of siblings

·       Life Events

o   Childbirth

o   Menopause

o   Rape or assault

o   Natural disasters

o   Job loss or unemployment

o   Stress

o   Financial difficulties

o   Bullying

o   Relationship or marriage breakdown

o   Bereavement

o   Catastrophic injury

·       Mental illness

o   Schizophrenia

o   Drug or alcohol misuse

·       Physical illness

o   Infection

§  HIV

o   Nutritional deficiencies

o   Cardiovascular disease

§  Myocardial infarction

§  Stroke

o   Pernicious anemia

o   Neurological

§  Parkinsonism

§  Multiple sclerosis

o   Endocrine

§  Diabetes

§  Obesity

§  Hypoandrogenism

§  Cushing syndrome

§  Hypothyroidism

§  Hyperparathyroidism

o   Cancer

o   Arthritis

o   Chronic pain

o   Inflammation

·       Side effects of medication

o   β-blockers

o   α-interferon

o   Finasteride

o   Isotretinoin

o   Dopamine receptor agonists

o   Some anticonvulsants

o   Some antimigraine agents

o   Some antipsychotics

 

 

DIABETES AND DEPRESSION

 

The association between diabetes and depression has been recognized for many years. In the 17th century, Thomas Willis, an English physician, described how “diabetes is a consequence of prolonged sorrow” (20). Until the last two decades, the focus of any discussion of mood and diabetes was on the increased likelihood of depression in people with diabetes, where the comorbidity was viewed as an understandable reaction to the difficulties and challenges of living with a demanding and life-limiting long-term physical illness. In this regard, it was treated no differently from other long-term physical conditions that are also associated with increased rates of depression. We now understand that the relationship between the two conditions is more complex and, at least for type 2 diabetes, is bidirectional (8).

 

Understanding the scale of the co-morbidity is challenging because the epidemiological studies examining the relationship have been hampered by considerable variation in measurement, study design, and use of terminology that have contributed to significant heterogeneity and inconsistency between studies (8). An example of this variability is illustrated by one meta-analysis that reported a range of prevalence rates of depression in people with diabetes from 1.8% to 88% (21).

 

The gold standard for the diagnosis of depression is a diagnostic interview, but many studies have relied on self-rating scales (22). These scales identify depressive symptoms rather than depression and do not differentiate between symptoms that could be caused by either diabetes or depression, for example, fatigue or weight change. This can lead to significant overestimates of the prevalence of depression in those with diabetes, as shown in an early meta-analysis where the prevalence of depression in people with diabetes identified by diagnostic interview was 11.4% compared to 31.0% in studies using self-rating questionnaire (23). However, a more recent meta-analysis argued that the symptom overlap does not affect prevalence (21). Nevertheless, the authors argue that depression measures that focus on the cardinal symptoms of depression rather than overlapping symptoms may most accurately diagnose depression.

 

Studies have often used mixed populations of people with type 1 diabetes and type 2 diabetes. This distinction is important because people with type 2 diabetes are generally older, and depression prevalence varies with age (16).There are differences in the prevalence of diabetes-related complications and other comorbid conditions such as obesity and heart disease, which independently affect the likelihood of developing depression (24). The pathogenesis and etiology of the two main types of diabetes differ, which may affect the risk of depression in different ways (25). Finally, the burden of management differs between type 1 diabetes and type 2 diabetes.

 

Many studies have not taken broad population approaches but have concentrated on convenience samples, usually drawn from specialist diabetes clinics, where the participants may not reflect the wider population of people living with diabetes. This bias is illustrated in the meta-analysis by Farooqi et al which reported that the prevalence of depression in people with diabetes was 36% in studies carried out in specialist care compared to 12% in community or primary care settings (26). Biases may also occur where there are low or unknown response rates, because the presence of depressive symptoms may affect the willingness of an individual to participate.

 

A further confounding factor is the concept of diabetes distress which describes the emotional response to living with diabetes, including the demands of self-management, the threat of complications, the social impact of stigma and discrimination, and the financial costs of treatment (27). Diabetes distress can fluctuate over time and may peak during challenging periods such as soon after diagnosis, during major treatment changes, or during the development or worsening of long-term complications (Table 3). Diabetes distress is distinct from depression in its association with self-management and glycemic levels, but the two conditions may co-exist in about 5-15% of people with diabetes. By contrast depression occurs without distress in 5-10% of people while distress alone affects 20-30% (28). The importance of diabetes distress was first proposed in 1995, and it is likely that early studies of diabetes and depression were capturing distress as well as depression, thereby contributing to an overestimate of the prevalence of depression.

 

Table 3. Common Features of Diabetes Distress

•           Feeling overwhelmed by the demands of living with diabetes

•           Feeling concerned about “failures” with diabetes management

•           Feeling powerless or hopeless

•           Worrying over the risk of low blood glucose or long-term complications

•           Feeling frustrated that diabetes cannot be predicted or controlled from one day to the next

•           Feeling frustrated with care givers

•           Feeling guilty when the diabetes management go ‘off track’.

 

Despite these caveats, several meta-analyses have indicated that the prevalence of depression is approximately doubled in people with diabetes compared to the background population (23, 26), with the prevalence of depression similar between type 1 diabetes (22%) and type 2 diabetes (19%) (26). Although most studies come from Western Europe or North America, the increased rates of depression or depressive symptoms have been found across the world (29, 30), with the prevalence being higher in low- and middle-income countries (26). A meta-analysis of 248 observational studies including over 8 million people with type 2 diabetes found a global prevalence of depression of 28%, with the highest rates observed in Asia and Australia whilst the lowest rates were found in studies from Europe (31). Cohort studies have shown an increase in incident depression in people with diabetes; one meta-analysis of 11 studies involving approximately 50,000 people with type 2 diabetes but without depression at baseline reported that the incidence of depression was 24% higher in people with diabetes (32), while another meta-analysis of 13 studies found incident depression was increased by 15% in people with diabetes (33).

 

An increased prevalence of depression has also been found in children with diabetes. A meta-analysis of 109 studies involving over 50,000 children with diabetes estimated that the prevalence of depression was 22.2% among children with type 1 diabetes and 22.7% in children with type 2 diabetes (34). Consistent with studies in adults, the prevalence of depression was higher among girls than boys (29.7% vs. 19.7%) and in low- to- middle-income countries.

 

Type 2 diabetes is a common comorbidity in people with mental disorders, with a reported prevalence ranging from 5% to 22% depending on the specific psychiatric disorder. Overall, the prevalence of diabetes in people with depression is 9% but there is considerable heterogeneity between studies (35). Consistent with this finding, the incidence of diabetes in people with depression is increased by 18-60% (33, 36, 37). People with depression, however, may receive more screening for diabetes than those without because of their increased contact with healthcare professionals and an awareness of the risk of diabetes by mental health practitioners, which may lead to an overestimate of the difference in diabetes risk between those with and without depression, particularly in studies that rely on routinely collected healthcare data.

 

Diabetes Factors That Increase the Risk of Depression

 

Although many cases of depression are coincidental to the presence of diabetes, certain diabetes factors including diabetes-related complications and obesity are associated with an increased risk of depression (38).

 

DIABETES COMPLICATIONS

 

The development of macrovascular and microvascular complications increases the risk of depression in people with type 1 diabetes and type 2 diabetes  (24, 39). Overall, the presence of diabetes complications increases the risk of incident depressive disorder by 14% but the increased risk of developing depression is 24% higher for microvascular complications compared with 9% higher for those with macrovascular complications (39). The risk of depression increases as more complications develop such that the presence of two or more complications more than doubled the risk of depression in people with type 2 diabetes in one specialized outpatient clinic, with neuropathy and nephropathy showing the strongest association (40).

 

DIABETES TREATMENTS

 

The use of insulin in type 2 diabetes is associated with higher rates of depression compared to non-insulin medications or dietary and lifestyle interventions alone. One meta-analysis of 28 studies reported an overall 59% higher risk of developing depression in people taking insulin and 42% higher risk when compared with oral anti-diabetes agents (41). It seems unlikely that insulin per se increases depressive symptoms, but insulin is associated with higher treatment demands that not only include self-injection but more intensive self-monitoring, which may adversely affect depressive symptoms (42). Insulin is also used in those with longer duration of type 2 diabetes, which may be associated with a higher prevalence of diabetes-related complications and elevated HbA1c, a further risk factor of depression (43). Insulin has been used erroneously as a threat by healthcare professionals to encourage people to follow certain health behaviors or take medications. As type 2 diabetes is associated with progressive β-cell decline, many people ultimately need insulin. Where insulin has been used as a threat, commencing insulin can evoke feelings of guilt, blame or failure, which may increase the likelihood of depression. Furthermore, people may have strongly held beliefs about insulin usage, seeing it as an “end of the road” treatment. They may also associate insulin with the development of diabetes complications or death if they have seen a family member with diabetes developing complications while using insulin. Insulin therapy is associated with significant weight gain, again a risk factor for depression, and an increased risk of hypoglycemia. In a 10-year study of 3,742 people with type 1 diabetes requiring emergency room visit or hospitalization, those admitted with a hypoglycemic event were 74% more likely to develop depression (43).

 

Other anti-diabetes treatments, by contrast, may be associated with a reduced risk of depression and there has been discussion about whether these could be re-purposed as treatments for depression (44). A meta-analysis of current therapies indicated that pioglitazone was associated with improved depressive symptoms, more so in women, but metformin had no consistent benefit (44). A more recent systematic review, however, suggested that metformin might help treat comorbid depression, although the evidence was too weak to recommend its use for this indication (45). A review of observational studies also indicated that metformin was associated with reduced depressive symptoms (46). GLP-1 receptors are widely expressed in the brain and have putative neuroprotective properties. A meta-analysis of six studies including 2,071 participants showed a small but significant reduction in depressive symptoms in those treated with GLP-1 receptor agonists (47). Whether the improvement is a direct effect or mediated through weight loss is unknown. This observation was also seen in a large population-based cohort and nested case-control study that found that low doses of metformin, dipeptidyl peptidase-4 (DPP4) inhibitors, GLP-1 receptor agonists, and sodium-glucose transporter 2 (SGLT2) inhibitors were associated with lower risk of depression in people with diabetes compared to those using other treatments, with the lowest risk seen with SGLT2 inhibitors (48). Further studies of the impact of SGLT2 inhibitors are warranted as abnormal brain bioenergetic metabolism occurs in depression and endogenous ketones may exert a neuroprotective effect that might improve mood. As SGLT2 inhibitors induce ketogenesis, there is a rationale to test whether SGLT2 inhibitors improve depressive symptoms (49).

 

 

No one mechanism explains the association between diabetes and depression, but specific disease and treatment factors may account for why diabetes pre-disposes to depression and vice versa (Figure 1). These are, however, superimposed on other factors, such as genetics, early intra-uterine development, and social determinants of health, that create a “common soil” for both conditions.

 

Figure 1. The possible mechanisms that lead to the co-morbidity of diabetes and depression.

 

Underlying Factors That Predispose to Both Diabetes and Depression

 

GENETICS

 

Modern genetic technologies, such as genome-wide association (GWAS) studies and Mendelian randomization analyses have revolutionized our understanding of how genetics may underlie many polygenic mental and physical disorders and their association as well as health behavior traits, such as smoking and alcohol consumption that influence health. These studies have shown overlap of genetic polymorphisms that increase the risk of several mental disorders, including depression, and physical disorders, including diabetes, metabolic syndrome, and obesity (50). There are nearly 500 single nuclear polymorphisms that are associated with an increased risk of both diabetes and depression across a broad range of pathways that include immune function, lipid metabolism, cancer-related pathways, and cell signaling (51).

 

FETAL DEVELOPMENT

 

The ‘developmental origins of health and disease’ hypothesis emerged from epidemiological studies that found that infants with low birth weight had an increased risk of cardiovascular disease, diabetes, and other chronic conditions in adulthood (52, 53). Although the early studies focused on physical illness, the fetal environment is linked to psychiatric conditions, including depression, although the effect size is weak and inconsistent across studies (54).

 

SOCIAL DETERMINANTS OF HEALTH

 

The broad conditions in which people are born, live, learn, work, play, worship, and age affect a wide range of health, functioning, and quality-of-life outcomes and risks throughout the life course, which together are known as the ‘social determinants of health’ (55). These can be divided into macro-level factors, such as government policy, meso-level factors, such as neighborhood and workplace, and individual factors, such as health behaviors and are broadly grouped into five domains: economic stability, educational access and quality, health care access and quality, neighborhood and built environment, and social and community context, which include gender and race. These social factors are more robust predictors of population health than either the provision of healthcare services or individual health behaviors and explain up to 80% of a person’s health (56).

 

Many of the risk factors for depression described earlier in the chapter include or are affected by social factors, such as childhood adversity, low socio-economic status, or lived environment. Many of these same factors also affect the risk of diabetes. For example, access to recreational spaces, safe housing and surroundings, clean air, and shops that sell nutritious and wholesome foods provide an environment where an individual can more easily choose behaviors that would reduce the risk of diabetes.

 

Diabetes-Specific Factors

 

Both psychological and biological mechanisms contribute to the increased risk of depression in people with diabetes.

 

PSYCHOLOGIOCAL FACTORS

 

The psychological model proposes that depression is an understandable response to the difficulties of living with a demanding and life-shortening long-term physical illness that is associated with potentially debilitating complications. This model is supported by a systematic review of 11 studies that found no difference in the prevalence of depression between those with undiagnosed diabetes, those with impaired glucose metabolism, and people with normal glucose metabolism (57). By contrast, an increased prevalence of depression was only found in those with diagnosed diabetes suggesting that the knowledge of the diagnosis and the burden of managing the condition and its complications are associated with the development of depression. A more recent meta-analysis showed an 11% and 27% increased risk of depression in those with pre-diabetes and undiagnosed diabetes, respectively, compared with people with normal glucose metabolism but this was lower than the 80% increase in those with known diabetes (58). The increase was only seen in people aged less than 60 years old and was partially explained by the presence of comorbid cardiovascular disease. Since this publication, studies from Mexico (59), Germany (60) and rural China (61) did not find increased rates of depression in people with undiagnosed diabetes. However, a large study from the Netherlands found a similarly increased rate of depression in those with diagnosed and undiagnosed diabetes (62).

 

Although these findings generally support the psychological model, it is important to recognize that the people with undiagnosed diabetes differ from those with diagnosed diabetes by more than just the knowledge of their condition. For example, those with diagnosed diabetes are likely to have had diabetes for longer and have developed complications and other co-morbidities that may affect their risk of depression.

 

EFFECT OF DIABETES ON BRAIN STRUCTURE AND FUNCTION

 

It is well recognized that acute hyperglycemia and hypoglycemia can affect mood (63, 64). This is unsurprising because the brain is dependent on a continuous supply of glucose as its principal source of energy, and changes in blood glucose levels rapidly affect cerebral function. However, longer term effects of diabetes on brain structure and function have also been seen in animal models of diabetes and in humans. In animals, diabetes negatively affects hippocampal integrity and neurogenesis, both of which are areas that are important in cognition and mood (65). In adults with type 1 diabetes, magnetic resonance imaging (MRI) studies have shown hippocampal atrophy together with increased prefrontal glutamate-glutamine-gamma-aminobutyric acid (GABA) levels in a way that correlates with mild depressive symptoms (65, 66). In the brain, insulin stimulates glucose uptake, in part by increasing the synthesis of the glucose transporters in neurons and neuroglia (67). Consequently, abnormal insulin signaling in the brain could affect glucose transport across the blood-brain barrier leading to reduced neuronal glucose uptake and neuronal loss. As the amygdala and hippocampus are the regions that contain a high density of insulin receptors, they may be disproportionately affected by insulin resistance, which is independently associated with depression (68).

 

At a cellular level within the hippocampus, diabetes is associated with an increase in astrocytes and microglia reactivity and apoptosis of pyramidal neurons, and reduced neurogenesis and synaptic plasticity with dendritic retraction (69). In addition to the changes in GABA, there are other molecular changes, including increased glucocorticoid signaling, reduced brain-derived neurotrophic factor (BDNF) production, increased mGluR2/3 activity and caspase 3 activation, and an increase in the TLR4/NFκB signaling pathway, together with increased production of reactive oxygen species and pro-inflammatory cytokines, such as TNF-β. These changes lead to increased apoptosis and decreased progenitor proliferation, which in turn lead to a decrease in the hippocampal size.

 

Depression-Specific Factors

 

Depression may increase the risk of diabetes through health behaviors as well as the biological effects of depression and its treatment with antidepressants.

 

ADULT HEALTH BEHAVIORS

 

Health behaviors play an important role in determining an individual’s risk of developing diabetes.

 

Diet

 

A healthy diet protects against many long-term conditions including diabetes, obesity, and depression. Both epidemiological studies and intervention studies have shown how maintaining normal weight, reducing fat, particularly saturated fat, and increasing the fiber content of the diet reduces the risk of type 2 diabetes (70, 71). Certain dietary patterns, such as the Mediterranean diet, are associated with a lower risk of diabetes (72).

 

High levels of refined sugars and saturated fats may also increase the risk of depression, while a Mediterranean diet and diets that include more vegetables, fruits, fish, and whole grains seem to be protective (73). Once present, depression may entrench less prudent eating habits, creating a vicious cycle where poor diet and depression reinforce each other while simultaneously increasing the risk of diabetes. Depression increases the risk of obesity, with those with depression being 58% more likely to develop obesity than those without (74).

 

Excessive alcohol intake is associated with an increased risk of diabetes (75). Alcohol misuse is one of the most prevalent mental disorders, especially in more affluent countries (76). About a quarter of those with alcohol dependency have co-morbid mental disorders including depression, where alcohol is often used as a coping mechanism to manage stress, anxiety, or depressive symptoms.

 

Physical Activity

 

The health benefits of physical activity are overwhelming and include a lowered risk of type 2 diabetes and depression. There is a graded response with some physical activity being better than none, but further benefits accrue with more physical activity. Avoidance of sedentary behavior is also important for health. People with depression are less physically active than the general population; a meta-analysis of 24 studies including 2901 people with major depression disorder found that compared to the general population, those with depression spent less time engaged in overall and moderate to vigorous physical activity and were more likely to be sedentary. People with depression were 50% less likely to meet the recommended physical activity guidelines of taking at least 150 minutes of moderate-to-high intensity physical activity in a week through a variety of activities (77). Over two-thirds of people with depression do not reach this target (78).

 

Smoking

 

Tobacco use is the single most preventable cause of death and disease throughout the life-course and increases the risk of diabetes by 30-40% (79, 80). Smoking is one of the most important modifiable risk factors of physical morbidity and mortality in people with mental illness (81). Adults with depression are twice as likely to smoke as adults without depression. There also appears to be a bi-directional relationship where smoking increases the risk of depression while people with depression are more likely to start smoking (82).

 

Sleep

 

The health benefits of sleep include a lower risk of diabetes and maintenance of a healthy weight (83). Sleep problems are a cardinal feature of depression with difficulty falling asleep and waking during the night, being common symptoms of depression (10).

 

BIOLOGICAL EFFECTS OF DEPRESSION

 

Several biological changes occur during an episode of depression that might increase the risk of diabetes. First, acute episodes of depression are associated with hyperinsulinemia and insulin resistance and are unaffected by antidepressant treatment (68). Depression is also associated with a state of chronic inflammation that is characterized by increased C-reactive protein, TNF-α, and proinflammatory cytokines that might partially explain the change in insulin sensitivity. These proteins are linked to sickness behavior in animal models of depression and in humans are associated with an increase in type 2 diabetes and the metabolic syndrome (84, 85). Depression is further associated with abnormalities of hypothalamic-pituitary adrenal (HPA) axis function, which manifests as subclinical hypercortisolism, blunted diurnal cortisol rhythm, or hypocortisolism with impaired glucocorticoid sensitivity (86). Brain-derived neurotrophic factor (BDNF) is a neurotrophic factor expressed in several tissues, including the brain, gut, and pancreas. As described earlier, BDNF plays an important role in maintaining neuronal plasticity, including neurogenesis, synaptogenesis, and neuronal maturation. Depression decreases BDNF expression in the hippocampus and prefrontal cortex (87). Outside the brain, BDNF activation leads to reduced hepatic gluconeogenesis, increased hepatic insulin signal transduction, and protects against pancreatic β-cell loss. Serum BDNF concentrations are lower in people with diabetes (88).

 

ANTIDEPRESSANTS

 

Although essential components of the management of depression, it is possible that the use of antidepressants contribute to the risk of diabetes. Case reports, and observational studies have generally shown that people receiving antidepressant medications have a higher risk of diabetes but whether this relationship is causative remains unproven (89, 90). Randomized controlled trials have emphasized that antidepressants vary considerably in their association with weight gain and both hyperglycemia and hypoglycemic effects have been observed (89). Some antidepressants, including paroxetine, mirtazapine, and various tricyclic antidepressants are associated with significant weight gain which could increase the risk of diabetes in the long term. By contrast, buproprion is associated with weight loss (91).

 

CONSEQUENCES OF DIABETES AND DEPRESSION CO-MORBIDITY

 

The presence of depression in people with diabetes worsens both diabetes and depression outcomes (Figure 2).

Figure 2. The consequences of living with diabetes and depression.

 

Mortality

 

Mortality rates from physical illnesses, including cardiovascular disease, cancer, and diabetes, are higher in people with depression. Among people with diabetes, depression increases the risk of mortality by approximately 50% (92, 93). Where depression co-exists with anxiety, which is also more common in people with diabetes, mortality rates are further increased (94).

 

Depression Outcomes

 

Once depressive symptoms occur or a diagnosis of depression is made, the symptoms appear to be persistent and likely to recur in people with diabetes. A longitudinal study of 2,460 people with type 2 diabetes in a primary care setting found that 26% met the criterion for depression on at least one occasion, with incident depression occurring in 14% over a 3-year period (95). Recurrent or persistent depression occurred in two-thirds of those with baseline depression. In two other studies from the USA, self-reported depressive symptoms persisted in 73% of people 12 months after a diabetes education program (96) while major depressive disorder relapsed in 79% of people with diabetes over a 5-year period (97).

 

Depression is a major cause of excess hospitalization in people with diabetes and is the leading cause of psychiatric admissions in people with diabetes accounting for 6.1 admissions per 1,000 person years in people with type 1 diabetes and 7.05 admissions in people with type 2 diabetes in Australia, with higher admission rates for women (98). These rates are 2-3-fold higher than the general Australian population (99).

 

People with diabetes have an increased risk of completed suicide compared with the general population and are more likely to report suicidal ideation, one of the strongest predictors of completed suicide, and intentional self-harm (100). The risk of suicidal behavior is highest in young people with type 1 diabetes, in whom suicide may account for up to 7% of deaths. Suicidal ideation is also increased among adolescents and young adults with type 1 diabetes compared with the general population (15.0% vs. 9.4%) and is seen in all ethnic groups (101). It is likely, however, that the true incidence of suicidal attempts and completed suicides is considerably higher amongst people with diabetes because of ineffective identification and coding (100). Many hospital admissions for diabetes ketoacidosis or hypoglycemia of ‘unknown’ etiology result from insulin omission or overdose, but whether these are deliberate acts is frequently unrecognized or unrecorded. There is also anecdotal evidence that healthcare professionals are unwilling to record suicide as a cause of death because of the associated stigma. Suicide rates are higher in people with long-term health conditions than in people in the general population, but what makes diabetes stand out is access to insulin, providing a means for suicide either by omission or overdose, the latter of which is the commonest method of suicide in people with insulin-treated diabetes. 

 

Diabetes Outcomes

 

ACUTE METABOLIC COMPLICATIONS

 

People with type 1 diabetes and depression have an increased risk of admission to hospital with diabetic ketoacidosis and severe hypoglycemia. In one study involving 3,742 people with type 1 diabetes who attended the emergency room or who were admitted to hospital between 1996 and 2015, those with depression had a 2.5-fold increased risk of severe hyperglycemia events and an 89% increased risk of severe hypoglycemia (43). The risk was greatest within the first 6 months following a diagnosis of depression, when the risk was 7.14-fold higher for hyperglycemia events and 5.58-fold higher for hypoglycemia.

 

MICROVASCULAR COMPLICATIONS

 

An early meta-analysis showed that the risk of microvascular complications was increased with small to moderate weighted effect sizes of 0.17 to 0.32 (24). The increased risk was similar in people with type 1 diabetes and people with type 2 diabetes while sexual dysfunction and painful peripheral neuropathy seemed to be associated with the highest risk. Most of these studies were cross-sectional but a more recent meta-analysis of 16 studies that examined the relationship between baseline depression and incident diabetes complications found that depression was associated with a 33% increased risk of incident microvascular disease (39). Most studies reported a composite of neuropathy, retinopathy, and nephropathy but one study reported nephropathy alone and found a 18% increase in incident chronic kidney disease (102). Depression is associated with a 68% increased risk of a first diabetes-related foot ulcer but not ulcer recurrence (103). This contrasted an earlier study that found a third of all individuals with diabetes-related foot ulcer had depression and that depression was associated with a threefold increased risk of dying (104).

 

MACROVASCULAR COMPLICATIONS

 

A recent meta-analysis has reported that incident macrovascular complications were increased by 38% in people with baseline depression (39). Most studies used a composite macrovascular outcome which included atherosclerotic vascular disease, myocardial infarction, and stroke as well as congestive heart failure and stroke. Some studies also included cardiovascular procedures, such as coronary artery bypass grafting or other revascularization techniques. Only one study reported separate outcomes for stroke (HR 1.22) and coronary heart disease (HR 1.32) (102).

 

QUALITY OF LIFE

 

Quality of life has been assessed by several different measures in people with diabetes and depression, including the Diabetes Specific Quality of Life scale and SF-36. These studies consistently show that quality of life is impaired in people with the co-morbidity (105). The effect of diabetes and depression appears to be additive across several domains with the exception of mental health where most of the effect stems from depression (106).

 

COST OF TREATMENT

 

The presence of depression among people with diabetes can substantially increase health care costs. An analysis of 147,095 adults living in the US found that depression and diabetes alone increased healthcare expenditure by $2,654 and $2,692, respectively, compared with neither condition but when both conditions occurred together, the cost was increased by $6,037 (107). Based on these figures, the estimated total cost of treating co-morbid diabetes and depression in the US was $77.6 billion per year. A more recent study found that total health costs increased from $11,550 for people with diabetes alone to $16,511 for people with diabetes and depression (108). This was in part driven by higher rates of hospitalization (26.1% vs 17.4%) and emergency room visits (55.3% vs 43.0%). Ironically, the increased cost occurred despite decreased healthcare utilization. In a US study of 22,642 people with diabetes enrolled in the 2019 Behavioral Risk Factor Surveillance System, those with a diagnosis of a depressive disorder were 82% more likely to report not seeing a doctor because of healthcare costs in the previous year (109).

 

DIABETES MANAGEMENT

 

Glycemic Levels

 

It has been hypothesized that changes in glycemic levels may mediate the association between depression and diabetes micro- and macrovascular complications and mortality. However, studies that have investigated this have produced inconsistent findings. An early meta-analysis reported a small to moderate association between depression and elevated HbA1c for type 1 diabetes and type 2 diabetes, but included mainly cross-sectional studies that precluded an inference on temporality (110). A recent meta-analysis investigated the longitudinal association between self-reported depressive symptoms and HbA1c. The six longitudinal studies had a combined sample size of 3,683 participants who were followed for a mean period of 37 months (range six months to five years). There was a small significant association between baseline depressive symptoms and subsequent HbA1c levels (111). A further meta-analysis of 14 studies in children and adolescents with type 1 diabetes reported a correlation between depressive symptoms and HbA1c (112). The timing of the diagnosis of diabetes and depression may also be important. In a study of 11,837 people with type 2 diabetes registered with the UK Biobank followed for a median of 6.9 years, those diagnosed with major depression decades prior to type 2 diabetes had lower HbA1c over time compared to individuals without depression and those diagnosed closer to their diabetes diagnosis date (113). For individuals whose depression was diagnosed after diabetes, the time since the onset of depression also shaped the trajectory of HbA1c, with the adverse effect of a diagnosis of depression on HbA1cbeing greatest in those whose diagnosis occurred shortly after the onset of diabetes. Furthermore, the variability of HbA1c within any individual was 16% higher in those with post-diabetes depression (113).

 

Diabetes Self-Management

 

Optimizing glucose levels is highly dependent on self-care activities that include regular glucose monitoring, taking medication as prescribed, and engaging in health behavior change to improve diet and physical activity. Depression compromises an individual’s ability to self-manage their diabetes. A meta-analysis of 47 studies reported that depression significantly reduced the likelihood of engaging in self-management behaviors, including missed medical appointments, less medication taking and glucose monitoring, and less foot care (114, 115). Similar to anti-diabetes medications, people with co-morbid depression are also less likely to take anti-hypertensive medications and lipid-lowering therapy (115). Depression is associated with a less nutritious diet that is characterized by lower consumption of fruit and vegetables and increased refined carbohydrates (116). Physical activity is reduced while smoking and alcohol consumption is increased (115). The effect of depression appears to be mediated through its adverse effects on self-efficacy and illness perception (115).

 

MANAGEMENT OF DIABETES AND DEPRESSION

 

Preventing Depression in People with Diabetes

 

The implication of the psychological model of depression in diabetes is that healthcare professionals could play an important role in moderating the psychological burden associated with diabetes by considering the way in which the diagnosis of diabetes is conveyed and the psychosocial support that is given through an individual’s journey with diabetes. Many people with diabetes experience stigma, some of which stem from their healthcare team. They report feelings of shame and blame because they are held responsible for developing overweight, obesity, or diabetes (117). However well-intentioned the healthcare professional is, it is clear that evoking these feelings is associated with worse clinical outcomes. By contrast, the use of person-centered, non-stigmatizing language can create a trusted and safe space for meaningful clinical discussion (118).

 

Several individual and group-based interventions with the aim of preventing the development of depressive symptoms have been trialed in people with diabetes. Of the twelve studies reported in a recent narrative review, half had a positive effect (119). Features associated with a reduction in the likelihood of developing depressive symptoms included diabetes self-management education and support, problem-solving and resilience-focused approaches, and emotion-targeted techniques.

 

Screening and Diagnosis of Depression

 

Given the importance of depression in people with diabetes and the availability of effective treatment, there is a strong rationale to screen for depression in people with diabetes (120), not least because depression is under-recognized in clinical practice. Primary care doctors do not diagnose and treat depression in 50–77% of cases (121, 122), while diabetologists only initiate antidepressant treatment in approximately one-third of their patients with clinical depression (123). Similarly diabetes nurses do not recognize depression and anxiety, missing 75-80% of those with the conditions (124).

 

A formal diagnosis of depression requires a validated interview method, such as the Mini International Neuropsychiatric Interview (MINI) or Composite International Diagnostic Interview (CIDI). No laboratory investigations are needed to diagnose depression, but it is prudent to rule out general medical conditions that may mimic the symptoms of a depressive episode. The diagnostic interviews are too labor-intensive to make them suitable for population screening or for screening in primary care or other clinical settings. However, numerous brief screening instruments or questionnaires that are simple to administer and have reasonable clinical specificity and sensitivity have been developed (Table 4). Not all screening questionnaires are appropriate because of the overlap of symptoms of depression and diabetes, including tiredness, lethargy, lack of energy, appetite changes, and sleeping difficulties. However, the Beck Depression Inventory (BDI), the Centre for Epidemiologic Studies Depression Scale (CED), the Patient Health Questionnaire (PHQ-9), and the Hospital Anxiety and Depression Scale (HADS) are all suitable for use in people with diabetes (22). Of these, the PHQ-9 is the best validated and most widely used in people with diabetes (125). It is also short, containing nine questions making it easy to administer in primary and secondary care settings. It has been suggested that the cut-off for major depression, which is ≥10 in primary care populations, should be increased by two points to ≥12 points in people with diabetes to help differentiate between diabetes-related symptoms and depressive symptoms (126).

 

Table 4. Reported sensitivity, specificity, positive and negative predictive value and reliability and validity in tools screening for depression in people with diabetes. Adapted from (22).

Tool

Sensitivity (%)

Specificity (%)

PPV (%)

NPV (%)

Reliability/

Validity (α)

CES.

60.0 – 100

86.7

28.6

 97.0

0.80

PHQ-9

66.0 - 100

52.0 - 80.0

43.8 - 64.4

93.4 - 100

0.80 - 0.84

BDI

82.0 - 90.0

84.0 - 89.0

59.0 - 89.0

82.0 - 97.0

 

HADS

74.0 - 85.0

37.5 – 86.0

28.0 – 49.0

95.0 - 96.0

0.55 – 0.78

Sensitivity= Number of true positives (cases of depression)/Number of true positives + number of false negatives; Specificity = Number of true negatives/ Number of true negatives + false positives; Positive Predictive Value (PPV) = the proportion of cases with positive test results who are correctly diagnosed. Negative Predictive Value (NPV) = the proportion of cases with negative test results who are correctly diagnosed.

 

Another simple approach with good sensitivity and reasonable specificity is to ask two questions (127):

  • During the past month, have you been bothered by having little interest or pleasure in doing things?
  • During the past month, have you been bothered by feeling down, depressed, or hopeless?

 

If the answer to either is yes, and the person with diabetes wants helps with this problem, the healthcare professional should undertake a diagnostic interview and offer appropriate referral and treatment.

 

Although screening for depression is acceptable to people living with diabetes (128), its value has not been proven and remains controversial despite being recommended by several professional bodies, including the International Diabetes Federation (129), American Diabetes Association (130), and the UK National Institute for Health and Clinical Excellence (131). A 2008 Cochrane review reported that depression screening alone in the general population had little or no impact on the detection and management of depression (132). However, a more recent meta-analysis of depression screening interventions, many of which included additional components beyond screening, showed these were associated with less depression or depressive symptoms in the general population 6-12 months after screening (133). This issue is important because of the potential harms of screening, which include the stigma associated with depression, the risk of transient distress being labelled as having depression, and discrimination from insurance companies, and so studies demonstrating the effectiveness of screening in people with diabetes are needed.

 

Primary care depression screening in people with diabetes was introduced in the UK in 2006 as part of the Quality and Outcomes Framework (a performance management and payment scheme for NHS general practitioners) with mixed results. In one semi-rural general practice, 365 of 435 eligible people with diabetes or ischemia heart disease were screened, but only three people without a current diagnosis of depression screened positive and none were subsequently diagnosed with depression (134). By contrast, in a study of 112 general practices in Leeds, UK, the rates of diagnosis of depression increased from 21 to 94 per 100,000 population per month after introduction of the screening compared with 27 to 77 per 100,000 population per month in people without screening (135). Despite the increased diagnosis, after an initial increase in antidepressant treatment, screening had little impact on prescribing habits.

 

Two clinical trials of depression screening in people with diabetes have not demonstrated a benefit. In the first study from the Netherlands, written feedback was provided to both the person with diabetes and the doctor following depression screening, but this did not change use of mental health services or improve depression scores compared with routine care (136). The second study from the USA examined the benefits of training healthcare technicians about the importance of discussing mental health with their patients (137). Although there was an improvement in depressive symptoms, this was no different from the control group and all the participants continued to have moderate to severe depressive symptoms.

 

Several reasons may explain the lack of effectiveness of depression screening in people with diabetes including a low acceptance of screening and subsequent referral to further care by people with diabetes, failure to screen those at highest risk of depression, reluctance by healthcare professionals, and generally poor quality of depression care in primary care systems (138). While identification of those with depression is an essential first step in treatment, screening alone will not improve clinical outcomes unless linked to appropriate care pathways and treatment (120, 139).  By contrast, studies of care pathways that have clearly linked screening to diagnosis and treatment improved depression outcomes (140, 141).

 

Treatment of Depression

 

The main aims of treatment are to improve both mental health and diabetes outcomes (Table 5). Ideally, any depression treatment for people with diabetes would simultaneously improve both sets of outcomes, but from a clinical perspective, the rapid improvement or remission of depression should be the first priority (138). This recommendation partly reflects the time course of treatment responses, which can be seen within 2–4 weeks for depression but also because treating the depression may aid optimal diabetes self-management.

 

Table 5. Aims of Depression Treatment in People with Diabetes

Mental health outcomes

Diabetes outcomes

Decreased depressive symptoms

Optimal diabetes self-management

Remission of depression

Decreased HbA1c

Suicide prevention

Increased time in glucose range

Improved health-related quality of life

Less hypoglycemia

Restoration of psychosocial functioning

Reduced long-term complications

 

Reduced mortality

 

Until relatively recently, people with diabetes have been under-represented in trials of depression treatment and so, there were few studies examining antidepressant and psychological treatment of depression. However, over the last two decades, the evidence base for treatment has grown substantially and has clearly indicated that treatment with either psychological therapies or antidepressant medication is effective (142) .

 

PSYCHOLOGICAL TREATMENT

 

Various psychological treatments, including cognitive behavioral therapy, problem-solving, and psychodynamic techniques have been used to treat depression in people with diabetes. Different members of the healthcare team have been utilized to deliver these interventions in primary and secondary care either in person or virtually through the internet or telephone (142, 143). The majority of trials have included people with type 2 diabetes with no trials conducted solely in people with type 1 diabetes.

 

A meta-analysis of psychological treatments, including group-based and online therapies, reported they were effective for the treatment of depression with large effect sizes (142). The follow-up ranged between 4 weeks and 1 year and thus, the longer term effects are unknown. Cognitive behavioral therapy is the most studied intervention, with its core components being cognitive restructuring, behavioral activation, and problem solving. This intervention was judged to be moderately effective in two recent meta-analyses (144, 145). Mindfulness also has an moderate benefit in treating depression (146), but there is mixed evidence on the benefit of motivational interviewing in reducing depressive symptoms (147, 148). Although psychological treatments are better than no treatment, the rates of recovery are low post-psychological intervention (17% vs 9% in controls) (149), indicating that many people will need additional support if they are to recover fully from their depression.

 

There is more debate about the effect of psychological interventions on diabetes outcomes (138) with one systematic review reporting a reduction in HbA1c of ~0.6 % (6 mmol/mol) (150) while another only reporting a non-significant improvement in glycemic levels (151). A meta-analysis of cognitive behavioral therapy showed a statistically significant but clinically insignificant reduction in HbA1c of 0.14% (1 mmol/mol). There was a greater effect on HbA1c if the intervention was delivered in a group-based and face-to-face fashion and included psycho-education, behavioral, cognitive, goal-setting, and homework assignment strategies as central components (145).

 

One of the challenges in delivering psychological interventions is the lack of trained personnel, a situation which appears to be worsening, at least in the United Kingdom (152). To address this deficiency, interventions have been designed to be delivered online or using mobile technologies, a trend which has increased dramatically since the Covid-19 pandemic. This has the potential to increase accessibility to treatment while limiting costs (142). One meta-analysis reported large effect sizes on depressive symptoms for online therapy and a moderate effect for telephone interventions, although no change in diabetes outcomes was seen (142). The beneficial effect on depressive symptoms up to 12 months after the interventions was supported by another systematic review, albeit again without improvement in diabetes outcomes (153). However, this finding was contradicted by a recent meta-analysis of 24 randomized controlled trials, 14 non-randomized controlled trials and three observational studies which reported no significant effect on depression outcomes (154). The discrepancy may partly explained by drop-out rates which vary from 13% to 42%; for those who remain in treatment, the interventions appear effective (155) and so the challenge will be to deliver services that engage people with diabetes and depression.

 

In the United Kingdom in 2008, the National Health Service introduced the Improving Access to Psychological Therapies (now NHS Talking Therapies for anxiety and depression) program to improve the delivery of, and access to, psychological therapies for depression. By 2021/22, nearly 1.2 million people had accessed these services. Although the clinical workforce is appropriately trained and supervised, many practitioners do not have experience of the challenges of living with diabetes. To address this issue, the Southampton diabetes team has formed a partnership with the local NHS Talking Therapies service. A practitioner joins the multidisciplinary team in the young adult clinic once a fortnight, helping to engage the person with diabetes and facilitate referral and access to the service. There is also a wider benefit to the city as the NHS Talking Therapies team have become much more aware of the challenges of living with diabetes. Introducing this service led to reductions in depressive symptoms and diabetes distress and was well received by people with diabetes and staff alike (156).

 

ANTIDEPRESSANTS

 

Antidepressants have been used to treat depressive symptoms since the late 1950s. There are many different antidepressants, but these fall into five main categories:

  • Selective Serotonin Reuptake Inhibitors (SSRI)
  • Serotonin and Noradrenaline Reuptake Inhibitors (SNRI)
  • Noradrenaline and Specific Serotoninergic Antidepressants (NASSA)
  • Tricyclics (TCA)
  • Monoamine Oxidase Inhibitors (MAOI)

 

The discovery of the first clinically useful antidepressants paved the way for an understanding of the underlying biological or neuroanatomical basis for depression. The first antidepressant was the tricyclic antidepressant (TCA), imipramine. It was first synthesized in 1951 as a potential antipsychotic and derived from work on chlorpromazine, which had pronounced sedative and antihistamine effects (157). Although imipramine has no antipsychotic effect, it was found to possess antidepressant effects. Other TCAs, such as amitriptyline, were subsequently synthesized by modifying the structure of imipramine. Iproniazid, a monoamine oxidase inhibitor (MAOI), was the next antidepressant to be discovered; again, this drug was initially developed for a different purpose, the treatment of tuberculosis, before its antidepressant effect was recognized. MAOIs prevent the breakdown of monoamine neurotransmitters (e.g. noradrenaline, dopamine and serotonin) while TCAs block the uptake of serotonin and noradrenaline resulting in an elevation of the synaptic concentrations of these transmitters. An understanding of the pharmacology led to the hypothesis that depression was caused by low catecholamine levels in the central nervous system (158). Both TCA and MAOI affect the serotonin system and in 1967, Coppen proposed that this was a more important neurotransmitter in depression than noradrenaline (159). Fluoxetine was the first in the class of selective serotonin re-uptake inhibitors (SSRI) and was developed by design following a search for molecules that could selectively block the re-uptake of serotonin. A major advantage of this approach was that it minimized adverse effects such as cardiovascular toxicity and anticholinergic effects. First synthesized in 1972 and launched in 1987, fluoxetine became the most widely prescribed drug in North America by 1990 (157). Although better tolerated than earlier antidepressants, SSRI still caused side effects, including sexual dysfunction, appetite change, nausea and vomiting, irritability, anxiety, insomnia, and headaches. In an attempt to reduce these adverse effects, other antidepressant classes were developed, including serotonin and noradrenaline reuptake inhibitors (SNRI, e.g. venlafaxine) and noradrenaline and specific serotoninergic antidepressants (NASSA, e.g. mirtazapine).

 

Antidepressants reduce depressive symptoms in people with diabetes as well as the general population; however, there have been relatively few formal efficacy trials in people with diabetes and even these have been are limited to a small group of antidepressants, including fluoxetine, sertraline, paroxetine, citalopram, escitalopram, agomelatine, nortriptyline, and vortioxetine (138, 160). Furthermore, most studies are short-term and so the medium- and long-term sustainability of pharmacological interventions after treatment cessation is uncertain. A systematic review and meta-analysis suggested that all antidepressants have similarly large effect size on depression outcomes as long as adequate doses are used (151). However, a more recent network meta-analysis of 12 randomized controlled trials involving 792 participants reported that there may be a greater reduction in depressive symptoms with escitalopram and agomelatine (160). Vortioxetine was associated with the greatest reduction in HbA1c with escitalopram, agomelatine, sertraline and fluoxetine also associated with a fall in HbA1c. No antidepressant was found to disrupt glucose levels (160). These differences should be viewed with caution as the number of trials and participants for each drug is small. Further head-to-head randomized controlled trials would help us understand more about the relative benefits and safety of different antidepressants on depression and glucose metabolism.

 

Given the similar efficacy between antidepressants, the treatment of choice depends largely on the side-effect profile, individual preference, and response. SSRI are widely used as first-choice agents because they are less cardiotoxic than TCA and are safer in overdose. Some antidepressants, notably mirtazapine, paroxetine and some TCA, may cause undesirable weight gain (91) and should be used with caution in people with type 2 diabetes. By contrast, buproprion, which is available in the USA, is associated with weight loss and, unlike SSRIs, does not appear to worsen sexual function (161).

 

The aim of treatment is complete remission of depressive symptoms. Treatment should be maintained at an adequate dose for at least 4–6 months after remission of symptoms to reduce the risk of relapse and recurrence. Recovery from depression may lead to a change in the individual’s behavior and routine which may have an effect on diabetes self-management. For example, if appetite improves, insulin requirements may increase, while on the other hand, if the person becomes more active, they may decrease. An individual approach is therefore needed to support the person’s glycemic management. There are important drug–drug interactions between antidepressants and oral anti-diabetes agents through inhibition of the cytochrome P450 3A4 and 2C9 isoenzyme. For example, the use of fluoxetine may potentiate the effect of sulfonylureas precipitating hypoglycemia (84).

 

EXERCISE

 

Many depression guidelines recommend exercise and other aspects of a healthy lifestyle as an integral component of management. Coupled with the importance of exercise in glycemic management, interventions to increase physical activity have been trialed and shown to be effective in reducing depressive symptoms and improving glycemic measures (162).

 

PREVENTING SUICIDE

 

A detailed description of the many effective interventions to prevent suicide is beyond the scope of this article (163),however, it is important for diabetes healthcare professionals to understand how to identify those at risk, particularly given the high prevalence of suicidal ideation and acts and immediate availability of a means of suicide. It is crucial that suicide can be discussed with people with diabetes in a safe and non-judgmental way. Talking about suicide remains highly stigmatized and choosing to reveal suicidal ideation can be difficult. The isolation that suicidal people feel can be reinforced by a critical response from the healthcare team. A recent survey of diabetes healthcare professionals found that the vast majority of respondents believed it is their professional responsibility to ask about suicide and self-harm, with nearly half reporting that this should be addressed every visit (164). Around three-quarters reported feeling comfortable discussing these issues, but those who were more reluctant to do so were concerned about their lack of training and uncertainty about what to do if someone reported self-harming behavior. Once an individual has discussed suicidal intention, it is important that the person is supported and has access to mental healthcare and suicide prevention interventions.

 

ORGANIZATION OF CARE

 

A common model of care of depression is the Stepped Care Model which provides a framework in which service delivery is organized to help those with depression, their caregivers, and healthcare professionals identify and access the most effective interventions (Figure 3) (131). The model utilizes a sequenced treatment process depending on the severity of depressive symptoms and response to previous treatments. The model allows a rational approach to the treatment of depression, while reducing costs and side effects of antidepressant through more appropriate prescribing.

 

Figure 3. Stepped Care Model for management of depression (131).

 

The first step involves the recognition of depression through screening and diagnostic interview and is reserved for those with suspected depression. For those with mild depression, step 2 involves the use of guided self-help, computerized CBT, and brief psychological interventions which can be delivered by the primary healthcare team or psychologist. The third step, which is also delivered in primary care settings, is indicated for those who do not respond to step 2 interventions, or for those with moderate and severe depression. Treatments include medication, high-intensity psychological interventions, or combined treatment. Step 4 corresponds to severe or treatment-resistant depression; the interventions are similar to those used in step 3 but now involve the mental health team. The final step is for those with life-threatening depression and/or severe self-neglect. In addition to medication and high-intensity psychological interventions, electroconvulsive therapy may be required under the supervision of a mental health crisis services and involve hospitalization.

 

A meta-analysis of 18 randomized controlled trial of stepped care showed improvements in depressive symptoms and better remission rates (165). A significant benefit on quality of life was also observed. More people in the stepped care model were prescribed antidepressants.

 

The increasing fragmentation of medical services and super-specialization in modern medicine has resulted in clinicians focusing on the conditions with which they are most familiar and being unable or unwilling to recognize and treat comorbid illnesses when they occur (166). The need for integrated holistic healthcare has never been greater but many diabetes healthcare professionals feel ill-equipped to manage depression. To address this, a case management model known as collaborative care was developed in the U.S., that involves a multidisciplinary team working together to identify and treat depression within primary care settings. The prototype intervention led to improvements in depression symptoms but without change in glycemia (167). Subsequently, greater attention was paid to intervention strategies for diabetes, resulting in simultaneous improvements in glycemic and blood pressure management and improved depressive symptoms (140). In a meta-analysis of five studies from the U.S., collaborative care was shown to be a clinical- and cost-effective treatment of depression, with a moderate effect size for depression outcomes and a small effect size for glycemic levels (142, 168).

 

CONCLUSION

 

Diabetes and depression remain a considerable clinical challenge. While an awareness of this co-morbidity has increased in recent years, this has not necessarily translated into better care or outcomes. Effective treatments are available and these need to be made available to those with diabetes and depression in clear treatment pathways. There are grounds for considerable optimism as the scientific knowledge that underpins clinical practices has expanded markedly in the last two decades. However, further research is needed to understand what can be done to prevent depression in people with diabetes and to identify the optimal treatment for an individual that improves both depressive symptoms and diabetes outcomes.

 

REFERENCES

 

  1. Katon WJ. Epidemiology and treatment of depression in patients with chronic medical illness. Dialogues Clin Neurosci. 2011;13(1):7-23.
  2. Ciechanowski PS, Katon WJ, Russo JE. Depression and diabetes: impact of depressive symptoms on adherence, function, and costs. Arch Intern Med. 2000;160(21):3278-85.
  3. Mitchell AJ, Malone D, Doebbeling CC. Quality of medical care for people with and without comorbid mental illness and substance misuse: systematic review of comparative studies. Br J Psychiatry. 2009;194(6):491-9.
  4. Buhagiar K, Parsonage L, Osborn DP. Physical health behaviours and health locus of control in people with schizophrenia-spectrum disorder and bipolar disorder: a cross-sectional comparative study with people with non-psychotic mental illness. BMC Psychiatry. 2011;11:104.
  5. Disability Rights Commission. Equal Treatment: Closing the gap. A formal investigation into physical Health inequalities experienced by people with learning difficulties and mental health problems. London: Disability Rights Commission; 2006.
  6. Schomerus G, Angermeyer MC. Stigma and its impact on help-seeking for mental disorders: what do we know? Epidemiol Psichiatr Soc. 2008;17(1):31-7.
  7. Holt RI, Mitchell AJ. Diabetes mellitus and severe mental illness: mechanisms and clinical implications. Nat Rev Endocrinol. 2015;11(2):79-89.
  8. Holt RI, de Groot M, Golden SH. Diabetes and depression. Curr Diab Rep. 2014;14(6):491.
  9. Royal College of Psychiatrists. Positive Cardiometabolic Health Resource: Don't just screen, intervene! 2014 [Available from: https://www.rcpsych.ac.uk/docs/default-source/improving-care/ccqi/national-clinical-audits/ncap-library/rcp17056-lester-uk-adaptation-4pp-a4-leaflet_4_updated.pdf?sfvrsn=7bc177e9_2.
  10. American Psychiatric Association. Diagnostic and statistical manual of mental disorders. Arlington: American Psychiatric Association; 2013. 1-947 p.
  11. Ferrari AJ, Charlson FJ, Norman RE, Patten SB, Freedman G, Murray CJ, et al. Burden of depressive disorders by country, sex, age, and year: findings from the global burden of disease study 2010. PLoS Med. 2013;10(11):e1001547.
  12. GBD Mental Disorders Collaborators. Global, regional, and national burden of 12 mental disorders in 204 countries and territories, 1990-2019: a systematic analysis for the Global Burden of Disease Study 2019. Lancet Psychiatry. 2022;9(2):137-50.
  13. Liu Q, He H, Yang J, Feng X, Zhao F, Lyu J. Changes in the global burden of depression from 1990 to 2017: Findings from the Global Burden of Disease study. J Psychiatr Res. 2020;126:134-40.
  14. World Health Organisation. Depression and Other Common Mental Disorders: Global Health Estimates. Geneva: World Health Organisation; 2017. 1-24 p.
  15. Ayuso-Mateos JL, Vazquez-Barquero JL, Dowrick C, Lehtinen V, Dalgard OS, Casey P, et al. Depressive disorders in Europe: prevalence figures from the ODIN study. Br J Psychiatry. 2001;179:308-16.
  16. Eaton WW, Anthony JC, Gallo J, Cai G, Tien A, Romanoski A, et al. Natural history of Diagnostic Interview Schedule/DSM-IV major depression. The Baltimore Epidemiologic Catchment Area follow-up. Arch Gen Psychiatry. 1997;54(11):993-9.
  17. Ford DE, Erlinger TP. Depression and C-reactive protein in US adults: data from the Third National Health and Nutrition Examination Survey. Arch Intern Med. 2004;164(9):1010-4.
  18. Ferenchick EK, Ramanuj P, Pincus HA. Depression in primary care: part 1-screening and diagnosis. BMJ. 2019;365:l794.
  19. Holt RI, Phillips DI, Jameson KA, Cooper C, Dennison EM, Peveler RC. The relationship between depression and diabetes mellitus: findings from the Hertfordshire Cohort Study. Diabet Med. 2009;26(6):641-8.
  20. Willis T. Pharmaceutice rationalis sive diabtriba de medicamentorum operantionibus in humano corpore. Oxford; 1675.
  21. Harding KA, Pushpanathan ME, Whitworth SR, Nanthakumar S, Bucks RS, Skinner TC. Depression prevalence in Type 2 diabetes is not related to diabetes-depression symptom overlap but is related to symptom dimensions within patient self-report measures: a meta-analysis. Diabet Med. 2019;36(12):1600-11.
  22. Roy T, Lloyd CE, Pouwer F, Holt RI, Sartorius N. Screening tools used for measuring depression among people with Type 1 and Type 2 diabetes: a systematic review. Diabet Med. 2012;29(2):164-75.
  23. Anderson BJ, Edelstein S, Abramson NW, Katz LE, Yasuda PM, Lavietes SJ, et al. Depressive symptoms and quality of life in adolescents with type 2 diabetes: baseline data from the TODAY study. Diabetes Care. 2011;34(10):2205-7.
  24. de Groot M, Anderson R, Freedland KE, Clouse RE, Lustman PJ. Association of depression and diabetes complications: a meta-analysis. Psychosom Med. 2001;63(4):619-30.
  25. Mehdi S, Wani SUD, Krishna KL, Kinattingal N, Roohi TF. A review on linking stress, depression, and insulin resistance via low-grade chronic inflammation. Biochem Biophys Rep. 2023;36:101571.
  26. Farooqi A, Gillies C, Sathanapally H, Abner S, Seidu S, Davies MJ, et al. A systematic review and meta-analysis to compare the prevalence of depression between people with and without Type 1 and Type 2 diabetes. Prim Care Diabetes. 2022;16(1):1-10.
  27. Fisher L, Polonsky WH, Hessler D. Addressing diabetes distress in clinical care: a practical guide. Diabet Med. 2019;36(7):803-12.
  28. Skinner TC, Joensen L, Parkin T. Twenty-five years of diabetes distress research. Diabet Med. 2020;37(3):393-400.
  29. Mommersteeg PM, Herr R, Pouwer F, Holt RI, Loerbroks A. The association between diabetes and an episode of depressive symptoms in the 2002 World Health Survey: an analysis of 231,797 individuals from 47 countries. Diabet Med. 2013;30(6):e208-e14.
  30. Hussain S, Habib A, Singh A, Akhtar M, Najmi AK. Prevalence of depression among type 2 diabetes mellitus patients in India: A meta-analysis. Psychiatry Res. 2018;270:264-73.
  31. Khaledi M, Haghighatdoost F, Feizi A, Aminorroaya A. The prevalence of comorbid depression in patients with type 2 diabetes: an updated systematic review and meta-analysis on huge number of observational studies. Acta Diabetol. 2019;56(6):631-50.
  32. Nouwen A, Winkley K, Twisk J, Lloyd CE, Peyrot M, Ismail K, et al. Type 2 diabetes mellitus as a risk factor for the onset of depression: a systematic review and meta-analysis. Diabetologia. 2010;53(12):2480-6.
  33. Mezuk B, Eaton WW, Albrecht S, Golden SH. Depression and type 2 diabetes over the lifespan: a meta-analysis. Diabetes Care. 2008;31(12):2383-90.
  34. Akbarizadeh M, Naderi Far M, Ghaljaei F. Prevalence of depression and anxiety among children with type 1 and type 2 diabetes: a systematic review and meta-analysis. World J Pediatr. 2022;18(1):16-26.
  35. Lindekilde N, Scheuer SH, Rutters F, Knudsen L, Lasgaard M, Rubin KH, et al. Prevalence of type 2 diabetes in psychiatric disorders: an umbrella review with meta-analysis of 245 observational studies from 32 systematic reviews. Diabetologia. 2022;65(3):440-56.
  36. Yu M, Zhang X, Lu F, Fang L. Depression and Risk for Diabetes: A Meta-Analysis. Can J Diabetes. 2015;39(4):266-72.
  37. Graham EA, Deschenes SS, Khalil MN, Danna S, Filion KB, Schmitz N. Measures of depression and risk of type 2 diabetes: A systematic review and meta-analysis. J Affect Disord. 2020;265:224-32.
  38. Busili A, Kumar K, Kudrna L, Busaily I. The risk factors for mental health disorders in patients with type 2 diabetes: An umbrella review of systematic reviews with and without meta-analysis. Heliyon. 2024;10(7):e28782.
  39. Nouwen A, Adriaanse MC, van Dam K, Iversen MM, Viechtbauer W, Peyrot M, et al. Longitudinal associations between depression and diabetes complications: a systematic review and meta-analysis. Diabet Med. 2019;36(12):1562-72.
  40. van Steenbergen-Weijenburg KM, van Puffelen AL, Horn EK, Nuyen J, van Dam PS, van Benthem TB, et al. More co-morbid depression in patients with Type 2 diabetes with multiple complications. An observational study at a specialized outpatient clinic. Diabet Med. 2011;28(1):86-9.
  41. Bai X, Liu Z, Li Z, Yan D. The association between insulin therapy and depression in patients with type 2 diabetes mellitus: a meta-analysis. BMJ Open. 2018;8(11):e020062.
  42. Li C, Ford ES, Strine TW, Mokdad AH. Prevalence of depression among U.S. adults with diabetes: findings from the 2006 behavioral risk factor surveillance system. Diabetes Care. 2008;31(1):105-7.
  43. Gilsanz P, Karter AJ, Beeri MS, Quesenberry CP, Jr., Whitmer RA. The Bidirectional Association Between Depression and Severe Hypoglycemic and Hyperglycemic Events in Type 1 Diabetes. Diabetes Care. 2018;41(3):446-52.
  44. Moulton CD, Hopkins CWP, Ismail K, Stahl D. Repositioning of diabetes treatments for depressive symptoms: A systematic review and meta-analysis of clinical trials. Psychoneuroendocrinology. 2018;94:91-103.
  45. Hamal C, Velugoti L, Tabowei G, Gaddipati GN, Mukhtar M, Alzubaidee MJ, et al. Metformin for the Improvement of Comorbid Depression Symptoms in Diabetic Patients: A Systematic Review. Cureus. 2022;14(8):e28609.
  46. Zhang Y, Chan VK, Chan SSM, Chan EWY, Lee CH, Wong IC, et al. Effect of metformin on the risk of depression: A systematic review and meta-regression of observational studies. Asian J Psychiatr. 2024;92:103894.
  47. Chen X, Zhao P, Wang W, Guo L, Pan Q. The Antidepressant Effects of GLP-1 Receptor Agonists: A Systematic Review and Meta-Analysis. Am J Geriatr Psychiatry. 2024;32(1):117-27.
  48. Wium-Andersen IK, Osler M, Jorgensen MB, Rungby J, Wium-Andersen MK. Diabetes, antidiabetic medications and risk of depression - A population-based cohort and nested case-control study. Psychoneuroendocrinology. 2022;140:105715.
  49. Liebers DT, Ebina W, Iosifescu DV. Sodium-Glucose Cotransporter-2 Inhibitors in Depression. Harv Rev Psychiatry. 2023;31(4):214-21.
  50. Postolache TT, Del Bosque-Plata L, Jabbour S, Vergare M, Wu R, Gragnoli C. Co-shared genetics and possible risk gene pathway partially explain the comorbidity of schizophrenia, major depressive disorder, type 2 diabetes, and metabolic syndrome. Am J Med Genet B Neuropsychiatr Genet. 2019;180(3):186-203.
  51. Ji HF, Zhuang QS, Shen L. Genetic overlap between type 2 diabetes and major depressive disorder identified by bioinformatics analysis. Oncotarget. 2016;7(14):17410-4.
  52. Barker DJ. The Wellcome Foundation Lecture, 1994. The fetal origins of adult disease. Proc Biol Sci. 1995;262(1363):37-43.
  53. Barker DJ. Fetal origins of coronary heart disease. BMJ. 1995;311(6998):171-4.
  54. Schlotz W, Phillips DI. Fetal origins of mental health: evidence and mechanisms. Brain Behav Immun. 2009;23(7):905-16.
  55. Marmot M, Friel S, Bell R, Houweling TA, Taylor S, Commission on Social Determinants of H. Closing the gap in a generation: health equity through action on the social determinants of health. Lancet. 2008;372(9650):1661-9.
  56. Hood CM, Gennuso KP, Swain GR, Catlin BB. County Health Rankings: Relationships Between Determinant Factors and Health Outcomes. Am J Prev Med. 2016;50(2):129-35.
  57. Nouwen A, Nefs G, Caramlau I, Connock M, Winkley K, Lloyd CE, et al. Prevalence of depression in individuals with impaired glucose metabolism or undiagnosed diabetes: a systematic review and meta-analysis of the European Depression in Diabetes (EDID) Research Consortium. Diabetes Care. 2011;34(3):752-62.
  58. Chen S, Zhang Q, Dai G, Hu J, Zhu C, Su L, et al. Association of depression with pre-diabetes, undiagnosed diabetes, and previously diagnosed diabetes: a meta-analysis. Endocrine. 2016;53(1):35-46.
  59. Olvera RL, Fisher-Hoch SP, Williamson DE, Vatcheva KP, McCormick JB. Depression in Mexican Americans with diagnosed and undiagnosed diabetes. Psychol Med. 2016;46(3):637-46.
  60. Weikert B, Buttery AK, Heidemann C, Rieckmann N, Paprott R, Maske UE, et al. Glycaemic status and depressive symptoms among adults in Germany: results from the German Health Interview and Examination Survey for Adults (DEGS1). Diabet Med. 2018;35(11):1552-61.
  61. Li Z, Guo X, Jiang H, Sun G, Sun Y, Abraham MR. Diagnosed but Not Undiagnosed Diabetes Is Associated with Depression in Rural Areas. Int J Environ Res Public Health. 2016;13(11).
  62. Meurs M, Roest AM, Wolffenbuttel BH, Stolk RP, de Jonge P, Rosmalen JG. Association of Depressive and Anxiety Disorders With Diagnosed Versus Undiagnosed Diabetes: An Epidemiological Study of 90,686 Participants. Psychosom Med. 2016;78(2):233-41.
  63. Sommerfield AJ, Deary IJ, Frier BM. Acute hyperglycemia alters mood state and impairs cognitive performance in people with type 2 diabetes. Diabetes Care. 2004;27(10):2335-40.
  64. Hermanns N, Scheff C, Kulzer B, Weyers P, Pauli P, Kubiak T, et al. Association of glucose levels and glucose variability with mood in type 1 diabetic patients. Diabetologia. 2007;50(5):930-3.
  65. Ho N, Sommers MS, Lucki I. Effects of diabetes on hippocampal neurogenesis: links to cognition and depression. Neurosci Biobehav Rev. 2013;37(8):1346-62.
  66. Lyoo IK, Yoon S, Jacobson AM, Hwang J, Musen G, Kim JE, et al. Prefrontal cortical deficits in type 1 diabetes mellitus: brain correlates of comorbid depression. ArchGenPsychiatry. 2012;69(12):1267-76.
  67. Leonard BE, Wegener G. Inflammation, insulin resistance and neuroprogression in depression. Acta Neuropsychiatr. 2020;32(1):1-9.
  68. Fernandes BS, Salagre E, Enduru N, Grande I, Vieta E, Zhao Z. Insulin resistance in depression: A large meta-analysis of metabolic parameters and variation. Neurosci Biobehav Rev. 2022;139:104758.
  69. Mazala-de-Oliveira T, Silva BT, Campello-Costa P, Carvalho VF. The Role of the Adrenal-Gut-Brain Axis on Comorbid Depressive Disorder Development in Diabetes. Biomolecules. 2023;13(10).
  70. Crandall JP, Knowler WC, Kahn SE, Marrero D, Florez JC, Bray GA, et al. The prevention of type 2 diabetes. Nat Clin Pract Endocrinol Metab. 2008;4(7):382-93.
  71. Knowler WC, Barrett-Connor E, Fowler SE, Hamman RF, Lachin JM, Walker EA, et al. Reduction in the incidence of type 2 diabetes with lifestyle intervention or metformin. N Engl J Med. 2002;346(6):393-403.
  72. Dyson PA, Twenefour D, Breen C, Duncan A, Elvin E, Goff L, et al. Diabetes UK evidence-based nutrition guidelines for the prevention and management of diabetes. Diabet Med. 2018;35(5):541-7.
  73. Shafiei F, Salari-Moghaddam A, Larijani B, Esmaillzadeh A. Adherence to the Mediterranean diet and risk of depression: a systematic review and updated meta-analysis of observational studies. Nutr Rev. 2019;77(4):230-9.
  74. Luppino FS, de Wit LM, Bouvy PF, Stijnen T, Cuijpers P, Penninx BW, et al. Overweight, obesity, and depression: a systematic review and meta-analysis of longitudinal studies. Arch Gen Psychiatry. 2010;67(3):220-9.
  75. Knott C, Bell S, Britton A. Alcohol Consumption and the Risk of Type 2 Diabetes: A Systematic Review and Dose-Response Meta-analysis of More Than 1.9 Million Individuals From 38 Observational Studies. Diabetes Care. 2015;38(9):1804-12.
  76. Carvalho AF, Heilig M, Perez A, Probst C, Rehm J. Alcohol use disorders. Lancet. 2019;394(10200):781-92.
  77. King AC, Whitt-Glover MC, Marquez DX, Buman MP, Napolitano MA, Jakicic J, et al. Physical Activity Promotion: Highlights from the 2018 Physical Activity Guidelines Advisory Committee Systematic Review. Med Sci Sports Exerc. 2019;51(6):1340-53.
  78. Schuch F, Vancampfort D, Firth J, Rosenbaum S, Ward P, Reichert T, et al. Physical activity and sedentary behavior in people with major depressive disorder: A systematic review and meta-analysis. J Affect Disord. 2017;210:139-50.
  79. World Health Organisation. Effects of tobacco on health 2016 [Available from: https://www.who.int/europe/news-room/fact-sheets/item/effects-of-tobacco-on-health.
  80. Pan A, Wang Y, Talaei M, Hu FB, Wu T. Relation of active, passive, and quitting smoking with incident type 2 diabetes: a systematic review and meta-analysis. Lancet Diabetes Endocrinol. 2015;3(12):958-67.
  81. Tam J, Warner KE, Meza R. Smoking and the Reduced Life Expectancy of Individuals With Serious Mental Illness. Am J Prev Med. 2016;51(6):958-66.
  82. Fluharty M, Taylor AE, Grabski M, Munafo MR. The Association of Cigarette Smoking With Depression and Anxiety: A Systematic Review. Nicotine Tob Res. 2017;19(1):3-13.
  83. Cappuccio FP, D'Elia L, Strazzullo P, Miller MA. Sleep duration and all-cause mortality: a systematic review and meta-analysis of prospective studies. Sleep. 2010;33(5):585-92.
  84. Musselman DL, Betan E, Larsen H, Phillips LS. Relationship of depression to diabetes types 1 and 2: epidemiology, biology, and treatment. Biol Psychiatry. 2003;54(3):317-29.
  85. Dantzer R, O'Connor JC, Freund GG, Johnson RW, Kelley KW. From inflammation to sickness and depression: when the immune system subjugates the brain. Nat Rev Neurosci. 2008;9(1):46-56.
  86. Champaneri S, Wand GS, Malhotra SS, Casagrande SS, Golden SH. Biological basis of depression in adults with diabetes. Curr Diab Rep. 2010;10(6):396-405.
  87. Duman RS, Deyama S, Fogaca MV. Role of BDNF in the pathophysiology and treatment of depression: Activity-dependent effects distinguish rapid-acting antidepressants. Eur J Neurosci. 2021;53(1):126-39.
  88. Moosaie F, Mohammadi S, Saghazadeh A, Dehghani Firouzabadi F, Rezaei N. Brain-derived neurotrophic factor in diabetes mellitus: A systematic review and meta-analysis. PLoS One. 2023;18(2):e0268816.
  89. Barnard K, Peveler RC, Holt RI. Antidepressant medication as a risk factor for type 2 diabetes and impaired glucose regulation: systematic review. Diabetes Care. 2013;36(10):3337-45.
  90. Greene CRL, Ward-Penny H, Ioannou MF, Wild SH, Wu H, Smith DJ, et al. Antidepressant and antipsychotic drug prescribing and diabetes outcomes: A systematic review of observational studies. Diabetes Res Clin Pract. 2023;199:110649.
  91. Serretti A, Mandelli L. Antidepressants and body weight: a comprehensive review and meta-analysis. J Clin Psychiatry. 2010;71(10):1259-72.
  92. Park M, Katon WJ, Wolf FM. Depression and risk of mortality in individuals with diabetes: a meta-analysis and systematic review. Gen Hosp Psychiatry. 2013;35(3):217-25.
  93. van Dooren FE, Nefs G, Schram MT, Verhey FR, Denollet J, Pouwer F. Depression and risk of mortality in people with diabetes mellitus: a systematic review and meta-analysis. PLoS One. 2013;8(3):e57058.
  94. Meier SM, Mattheisen M, Mors O, Mortensen PB, Laursen TM, Penninx BW. Increased mortality among people with anxiety disorders: total population study. Br J Psychiatry. 2016;209(3):216-21.
  95. Nefs G, Pouwer F, Denollet J, Pop V. The course of depressive symptoms in primary care patients with type 2 diabetes: results from the Diabetes, Depression, Type D Personality Zuidoost-Brabant (DiaDDZoB) Study. Diabetologia. 2012;55(3):608-16.
  96. Peyrot M, Rubin RR. Persistence of depressive symptoms in diabetic adults. Diabetes Care. 1999;22(3):448-52.
  97. Lustman PJ, Griffith LS, Clouse RE. Depression in adults with diabetes. Results of 5-yr follow-up study. Diabetes Care. 1988;11(8):605-12.
  98. Tomic D, Morton JI, Salim A, Lambert T, Magliano DJ, Shaw JE. Hospitalisation for mental health disorders in Australians with type 1 or type 2 diabetes. Diabetes Res Clin Pract. 2023;196:110244.
  99. Draper B, Low LF. Patterns of hospitalisation for depressive and anxiety disorders across the lifespan in Australia. J Affect Disord. 2009;113(1-2):195-200.
  100. Barnard-Kelly KD, Naranjo D, Majidi S, Akturk HK, Breton M, Courtet P, et al. Suicide and Self-inflicted Injury in Diabetes: A Balancing Act. J Diabetes Sci Technol. 2020;14(6):1010-6.
  101. Majidi S, O'Donnell HK, Stanek K, Youngkin E, Gomer T, Driscoll KA. Suicide Risk Assessment in Youth and Young Adults With Type 1 Diabetes. Diabetes Care. 2020;43(2):343-8.
  102. Novak M, Mucsi I, Rhee CM, Streja E, Lu JL, Kalantar-Zadeh K, et al. Increased Risk of Incident Chronic Kidney Disease, Cardiovascular Disease, and Mortality in Patients With Diabetes With Comorbid Depression. Diabetes Care. 2016;39(11):1940-7.
  103. Westby M, Norman G, Vedhara K, Game F, Cullum N. Psychosocial and behavioural prognostic factors for diabetic foot ulcer development and healing: a systematic review. Diabet Med. 2020;37(8):1244-55.
  104. Ismail K, Winkley K, Stahl D, Chalder T, Edmonds M. A cohort study of people with diabetes and their first foot ulcer: the role of depression on mortality. Diabetes Care. 2007;30(6):1473-9.
  105. Jing X, Chen J, Dong Y, Han D, Zhao H, Wang X, et al. Related factors of quality of life of type 2 diabetes patients: a systematic review and meta-analysis. Health Qual Life Outcomes. 2018;16(1):189.
  106. Goldney RD, Phillips PJ, Fisher LJ, Wilson DH. Diabetes, depression, and quality of life: a population study. Diabetes Care. 2004;27(5):1066-70.
  107. Egede LE, Bishu KG, Walker RJ, Dismuke CE. Impact of diagnosed depression on healthcare costs in adults with and without diabetes: United States, 2004-2011. J Affect Disord. 2016;195:119-26.
  108. Kangethe A, Lawrence DF, Touya M, Chrones L, Polson M, Evangelatos T. Incremental burden of comorbid major depressive disorder in patients with type 2 diabetes or cardiovascular disease: a retrospective claims analysis. BMC Health Serv Res. 2021;21(1):778.
  109. McClintock HF, Edmonds SE, Bogner HR. Depression and Cost-Related Health Care Utilization Among Persons with Diabetes. Popul Health Manag. 2023;26(4):232-8.
  110. Lustman PJ, Anderson RJ, Freedland KE, de GM, Carney RM, Clouse RE. Depression and poor glycemic control: a meta-analytic review of the literature. Diabetes Care. 2000;23(7):934-42.
  111. Beran M, Muzambi R, Geraets A, Albertorio-Diaz JR, Adriaanse MC, Iversen MM, et al. The bidirectional longitudinal association between depressive symptoms and HbA(1c) : A systematic review and meta-analysis. Diabet Med. 2022;39(2):e14671.
  112. Buchberger B, Huppertz H, Krabbe L, Lux B, Mattivi JT, Siafarikas A. Symptoms of depression and anxiety in youth with type 1 diabetes: A systematic review and meta-analysis. Psychoneuroendocrinology. 2016;70:70-84.
  113. Gillett AC, Hagenaars SP, Handley D, Casanova F, Young KG, Green H, et al. The impact of major depressive disorder on glycaemic control in type 2 diabetes: a longitudinal cohort study using UK Biobank primary care records. BMC Med. 2024;22(1):211.
  114. Gonzalez JS, Peyrot M, McCarl LA, Collins EM, Serpa L, Mimiaga MJ, et al. Depression and diabetes treatment nonadherence: a meta-analysis. Diabetes Care. 2008;31(12):2398-403.
  115. Derese A, Gebreegzhiabhere Y, Medhin G, Sirgu S, Hanlon C. Impact of depression on self-efficacy, illness perceptions and self-management among people with type 2 diabetes: A systematic review of longitudinal studies. PLoS One. 2024;19(5):e0302635.
  116. Sumlin LL, Garcia TJ, Brown SA, Winter MA, Garcia AA, Brown A, et al. Depression and adherence to lifestyle changes in type 2 diabetes: a systematic review. Diabetes Educ. 2014;40(6):731-44.
  117. Speight J, Holmes-Truscott E, Garza M, Scibilia R, Wagner S, Kato A, et al. Bringing an end to diabetes stigma and discrimination: an international consensus statement on evidence and recommendations. Lancet Diabetes Endocrinol. 2024;12(1):61-82.
  118. Holt RIG, Speight J. The language of diabetes: the good, the bad and the ugly. Diabet Med. 2017;34(11):1495-7.
  119. Guerin E, Jaafar H, Amrani L, Prud'homme D, Aguer C. Intervention Strategies for Prevention of Comorbid Depression Among Individuals With Type 2 Diabetes: A Scoping Review. Front Public Health. 2019;7:35.
  120. Holt RI, van der Feltz-Cornelis CM. Key concepts in screening for depression in people with diabetes. J Affect Disord. 2012;142 Suppl:S72-S9.
  121. Mitchell AJ, Vaze A, Rao S. Clinical diagnosis of depression in primary care: a meta-analysis. Lancet. 2009;374(9690):609-19.
  122. Mitchell AJ, Rao S, Vaze A. Can general practitioners identify people with distress and mild depression? A meta-analysis of clinical accuracy. J Affect Disord. 2011;130(1-2):26-36.
  123. Lustman PJ, Harper GW. Nonpsychiatric physicians' identification and treatment of depression in patients with diabetes. Compr Psychiatry. 1987;28(1):22-7.
  124. Pouwer F, Beekman AT, Lubach C, Snoek FJ. Nurses' recognition and registration of depression, anxiety and diabetes-specific emotional problems in outpatients with diabetes mellitus. Patient Educ Couns. 2006;60(2):235-40.
  125. Kroenke K, Spitzer RL, Williams JB, Lowe B. The Patient Health Questionnaire Somatic, Anxiety, and Depressive Symptom Scales: a systematic review. Gen Hosp Psychiatry. 2010;32(4):345-59.
  126. van Steenbergen-Weijenburg KM, de Vroege L, Ploeger RR, Brals JW, Vloedbeld MG, Veneman TF, et al. Validation of the PHQ-9 as a screening instrument for depression in diabetes patients in specialized outpatient clinics. BMC Health Serv Res. 2010;10:235.
  127. Whooley MA, Avins AL, Miranda J, Browner WS. Case-finding instruments for depression. Two questions are as good as many. J Gen Intern Med. 1997;12(7):439-45.
  128. Lloyd CE, Dyer PH, Barnett AH. Prevalence of symptoms of depression and anxiety in a diabetes clinic population. Diabet Med. 2000;17(3):198-202.
  129. International Diabetes Federation Guideline Development Group. Global guideline for type 2 diabetes. Diabetes Res Clin Pract. 2014;104(1):1-52.
  130. American Diabetes Association Professional Practice C. 4. Comprehensive Medical Evaluation and Assessment of Comorbidities: Standards of Care in Diabetes-2024. Diabetes Care. 2024;47(Suppl 1):S52-S76.
  131. National Collaborating Centre for Mental Health. Depression in adults with a chronic physical health problem CG91. London: The British Psychological Society and The Royal College of Psychiatrists; 2010.
  132. Gilbody S, Sheldon T, House A. Screening and case-finding instruments for depression: a meta-analysis. CMAJ. 2008;178(8):997-1003.
  133. O'Connor EA, Perdue LA, Coppola EL, Henninger ML, Thomas RG, Gaynes BN. Depression and Suicide Risk Screening: Updated Evidence Report and Systematic Review for the US Preventive Services Task Force. JAMA. 2023;329(23):2068-85.
  134. Subramanian DN, Hopayian K. An audit of the first year of screening for depression in patients with diabetes and ischaemic heart disease under the Quality and Outcomes Framework. Qual Prim Care. 2008;16(5):341-4.
  135. McLintock K, Russell AM, Alderson SL, West R, House A, Westerman K, et al. The effects of financial incentives for case finding for depression in patients with diabetes and coronary heart disease: interrupted time series analysis. BMJ Open. 2014;4(8):e005178.
  136. Pouwer F, Tack CJ, Geelhoed-Duijvestijn PH, Bazelmans E, Beekman AT, Heine RJ, et al. Limited effect of screening for depression with written feedback in outpatients with diabetes mellitus: a randomised controlled trial. Diabetologia. 2011;54(4):741-8.
  137. Scollan-Koliopoulos M, Herrera I, Romano K, Gregory C, Rapp K, Bleich D. Healthcare technician delivered screening of adults with diabetes to improve primary care provider recognition of depression. J Family Med Prim Care. 2012;1(2):97-102.
  138. Petrak F, Baumeister H, Skinner TC, Brown A, Holt RI. Depression and diabetes: treatment and health-care delivery. Lancet Diabetes Endocrinol. 2015;3(6):472-85.
  139. Hermanns N, Caputo S, Dzida G, Khunti K, Meneghini LF, Snoek F. Screening, evaluation and management of depression in people with diabetes in primary care. Prim Care Diabetes. 2013;7(1):1-10.
  140. Katon WJ, Lin EH, Von KM, Ciechanowski P, Ludman EJ, Young B, et al. Collaborative care for patients with depression and chronic illnesses. N Engl J Med. 2010;363(27):2611-20.
  141. Ell K, Katon W, Xie B, Lee PJ, Kapetanovic S, Guterman J, et al. Collaborative care management of major depression among low-income, predominantly Hispanic subjects with diabetes: a randomized controlled trial. Diabetes Care. 2010;33(4):706-13.
  142. van der Feltz-Cornelis C, Allen SF, Holt RIG, Roberts R, Nouwen A, Sartorius N. Treatment for comorbid depressive disorder or subthreshold depression in diabetes mellitus: Systematic review and meta-analysis. Brain Behav. 2021;11(2):e01981.
  143. van Bastelaar KM, Pouwer F, Cuijpers P, Riper H, Snoek FJ. Web-based depression treatment for type 1 and type 2 diabetic patients: a randomized, controlled trial. Diabetes Care. 2011;34(2):320-5.
  144. An Q, Yu Z, Sun F, Chen J, Zhang A. The Effectiveness of Cognitive Behavioral Therapy for Depression Among Individuals with Diabetes: a Systematic Review and Meta-Analysis. Curr Diab Rep. 2023;23(9):245-52.
  145. Li Y, Storch EA, Ferguson S, Li L, Buys N, Sun J. The efficacy of cognitive behavioral therapy-based intervention on patients with diabetes: A meta-analysis. Diabetes Res Clin Pract. 2022;189:109965.
  146. Ni YX, Ma L, Li JP. Effects of mindfulness-based intervention on glycemic control and psychological outcomes in people with diabetes: A systematic review and meta-analysis. J Diabetes Investig. 2021;12(6):1092-103.
  147. Berhe KK, Gebru HB, Kahsay HB. Effect of motivational interviewing intervention on HgbA1C and depression in people with type 2 diabetes mellitus (systematic review and meta-analysis). PLoS One. 2020;15(10):e0240839.
  148. Bilgin A, Muz G, Yuce GE. The effect of motivational interviewing on metabolic control and psychosocial variables in individuals diagnosed with diabetes: Systematic review and meta-analysis. Patient Educ Couns. 2022;105(9):2806-23.
  149. Mather S, Fisher P, Nevitt S, Cherry MG, Maturana C, Warren JG, et al. The limited efficacy of psychological interventions for depression in people with Type 1 or Type 2 diabetes: An Individual Participant Data Meta-Analysis (IPD-MA). J Affect Disord. 2022;310:25-31.
  150. Ismail K, Winkley K, Rabe-Hesketh S. Systematic review and meta-analysis of randomised controlled trials of psychological interventions to improve glycaemic control in patients with type 2 diabetes. Lancet. 2004;363(9421):1589-97.
  151. Baumeister H, Hutter N, Bengel J. Psychological and pharmacological interventions for depression in patients with diabetes mellitus and depression. Cochrane Database Syst Rev. 2012;12:CD008381.
  152. Winocour PH, Gosden C, Walton C, Nagi D, Turner B, Williams R, et al. Association of British Clinical Diabetologists (ABCD) and Diabetes-UK survey of specialist diabetes services in the UK, 2006. 1. The consultant physician perspective. Diabet Med. 2008;25(6):643-50.
  153. Varela-Moreno E, Carreira Soler M, Guzman-Parra J, Jodar-Sanchez F, Mayoral-Cleries F, Anarte-Ortiz MT. Effectiveness of eHealth-Based Psychological Interventions for Depression Treatment in Patients With Type 1 or Type 2 Diabetes Mellitus: A Systematic Review. Front Psychol. 2021;12:746217.
  154. Fernandez-Rodriguez R, Zhao L, Bizzozero-Peroni B, Martinez-Vizcaino V, Mesas AE, Wittert G, et al. Are e-Health Interventions Effective in Reducing Diabetes-Related Distress and Depression in Patients with Type 2 Diabetes? A Systematic Review with Meta-Analysis. Telemed J E Health. 2024;30(4):919-39.
  155. Franco P, Gallardo AM, Urtubey X. Web-Based Interventions for Depression in Individuals with Diabetes: Review and Discussion. JMIR Diabetes. 2018;3(3):e13.
  156. Holt RIG. Transforming the diabetes outcomes for people with mental illness. Practical Diabetes. 2024;41(4):3-8.
  157. Pereira VS, Hiroaki-Sato VA. A brief history of antidepressant drug development: from tricyclics to beyond ketamine. Acta Neuropsychiatr. 2018;30(6):307-22.
  158. Schildkraut JJ, Gordon EK, Durell J. Catecholamine metabolism in affective disorders. I. Normetanephrine and VMA excretion in depressed patients treated with imipramine. J Psychiatr Res. 1965;3(4):213-28.
  159. Coppen A. The biochemistry of affective disorders. Br J Psychiatry. 1967;113(504):1237-64.
  160. Srisurapanont M, Suttajit S, Kosachunhanun N, Likhitsathian S, Suradom C, Maneeton B. Antidepressants for depressed patients with type 2 diabetes mellitus: A systematic review and network meta-analysis of short-term randomized controlled trials. Neurosci Biobehav Rev. 2022;139:104731.
  161. Sayuk GS, Gott BM, Nix BD, Lustman PJ. Improvement in sexual functioning in patients with type 2 diabetes and depression treated with bupropion. Diabetes Care. 2011;34(2):332-4.
  162. Mohammad Rahimi GR, Aminzadeh R, Azimkhani A, Saatchian V. The Effect of Exercise Interventions to Improve Psychosocial Aspects and Glycemic Control in Type 2 Diabetic Patients: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Biol Res Nurs. 2022;24(1):10-23.
  163. Hofstra E, van Nieuwenhuizen C, Bakker M, Ozgul D, Elfeddali I, de Jong SJ, et al. Effectiveness of suicide prevention interventions: A systematic review and meta-analysis. Gen Hosp Psychiatry. 2020;63:127-40.
  164. Majidi S, Cohen L, Holt RIG, Clements M, O'Neill S, Renard E, et al. Healthcare Professional Experiences and Opinions on Depression and Suicide in People With Diabetes. J Diabetes Sci Technol. 2024;18(3):667-75.
  165. Rivero-Santana A, Perestelo-Perez L, Alvarez-Perez Y, Ramos-Garcia V, Duarte-Diaz A, Linertova R, et al. Stepped care for the treatment of depression: a systematic review and meta-analysis. J Affect Disord. 2021;294:391-409.
  166. Holt RI, Katon WJ. Dialogue on Diabetes and Depression: Dealing with the double burden of co-morbidity. JAffectDisord. 2012;142 Suppl:S1-S3.
  167. Katon WJ, Von Korff M, Lin EH, Simon G, Ludman E, Russo J, et al. The Pathways Study: a randomized trial of collaborative care in patients with diabetes and depression. Arch Gen Psychiatry. 2004;61(10):1042-9.
  168. Simon GE, Katon WJ, Lin EH, Rutter C, Manning WG, Von Korff M, et al. Cost-effectiveness of systematic depression treatment among people with diabetes mellitus. Arch Gen Psychiatry. 2007;64(1):65-72.

Diabetes In The Tropics

ABSTRACT

 

Diabetes mellitus (DM), an important non-communicable disease, is a major global health problem. Of the major three types of DM, namely type 1 diabetes mellitus (T1DM), type 2 diabetes mellitus (T2DM), and gestational diabetes mellitus, T2DM constitutes more than 90% of cases. The diabetes prevalence is on the rise in tropical regions. T1DM, caused by an autoimmune process, is influenced not only by genetic susceptibility but also to a greater extent by environmental factors. Among the multitude of environmental factors viral triggers, environmental toxins, exposure to exogenous antigens, and geographical factors play a significant role in T1DM pathogenesis. The hygiene hypothesis as explained by prevalent helminthic infection in the tropics, intense ultraviolet exposure translating to improved vitamin D synthesis serving as immune modulator, delayed exposure to cow’s milk and gluten thereby avoiding the allergen provoking beta cell autoimmunity, are a few of the postulated protective mechanisms for T1DM in tropical regions. Tropical regions comprise almost 40 percent of the world’s diabetic load with six countries in the top ten countries with DM. Reports from the IDF also predict a great increase in the coming decades with the maximum increment expected in Africa, Middle East, South-East Asia and South America. The incidence rate of diabetes among those with prediabetes in the Indian subcontinent is also one of the highest reported when compared to the Caucasian population yet comparable to Native Americans and Micronesian populations. World estimates indicate that 16.7% of pregnancies are complicated by some form of hyperglycemia. More than 80% of this is due to gestational DM. While the majority of hyperglycemia in pregnancy is seen in low- and middle-income countries, the prevalence between countries in tropical regions varies with South-East Asian countries topping the prevalence list while Middle East countries and northern Africa show the lowest prevalence (IDF). Diet patterns including greater consumption of tropical fruits with moderate or high glycemic index have been postulated to increase the likelihood of gestational DM. Fibro calculus pancreatic diabetes (FCPD) is a rare but unique and unexplored type of DM found specifically in tropical countries including India, Indonesia, Bangladesh, Sri-Lanka, Brazil and a few African countries. Most of the chronic pancreatitis originates from chronic alcoholism in developed countries contrasting with FCPD, which develops in the absence of alcohol use. Despite rising awareness about DM, the problem of ignorance about DM still exists. Data indicates the alarming fact that one in two adults with DM were unaware of their condition.  The increasing incidence and prevalence of DM in the tropics add to the infectious disease load and severity in the tropics. Infections are an important cause of morbidity and mortality in DM. Although disease duration and glycemic control are important risk factors, ethnicity may also play a role as a risk factor for complications.

 

INTRODUCTION

 

Diabetes mellitus (DM), an important non-communicable disease, is a major global health problem. Of the major three types of DM, namely type 1 diabetes mellitus (T1DM), type 2 diabetes mellitus (T2DM), and gestational DM, T2DM constitutes more than 90% of cases. According to the International Diabetes Federation (IDF), the prevalence of DM was 537 million with an almost equal number of the population having impaired glucose tolerance, which if not acted upon will contribute to the future burden of DM. Keeping in mind the current trends, the estimated worldwide prevalence of DM in two decades will be 784 million. Despite originally conceived as a disease of the affluent in developed, temperate countries, DM has skyrocketed to an alarming rate in developing tropical countries.

 

The IDF report also highlights the same fact that the burden of DM is increasing at a more rapid pace in low- as well as middle-income countries in comparison to high-income countries. Apart from geographical differences between tropical and temperate countries, they also diverge in terms of DM in substantial ways. Tropical region includes all the South-East Asian countries, part of Middle Eastern countries, a major part of Africa, South America, and a portion of Australia. Among the top ten countries contributing to the world’s DM’ burden, tropical countries occupy six slots including India which solely harbors one sixth of the world’s burden. Not only the disease burden, but also the pattern, types, demography, and epidemiology of DM is considerably different in tropical regions in comparison to the rest of the globe. It is also highlighted that most countries in tropical regions show lower rates of DM disease detection and limited access to management measures.

 

History dates to almost a century ago when the distinct aspects of people with DM in the tropics were beginning to be acknowledged as evidenced by an opinion by Sir Havelock Charles in the British Medical Journal (1). In 1950s Hugh Jones reported an unclassifiable form of DM in a small proportion of people with DM in Jamaica which he named as J-type (Jamaican type) (2). J-type was different from the then recognized two major forms of DM namely insulin dependent and noninsulin dependent DM in having mixed characteristics like young onset of disease, ketosis resistance, lean body habitus but with insulin resistance. Similar forms of DM were reported from the Indian subcontinent and Africa (3,4). With observation and follow-up, the clinical spectrum became more inconstant and perplexing with some patients becoming insulin independent, while a few becoming ketosis prone (5).

 

Amplifying the perplexity, abundance of nomenclature like tropical diabetes, Z-type, M-type, type 3 DM, pancreatic DM, malnutrition- related DM, fibro-calculous pancreatic DM, protein-deficient pancreatic DM, etc. exist. The WHO study group in 1985 recognized this group of DM and conglomerated it as a distinct entity naming it as malnutrition–related DM and also put forward the existence of two subtypes: protein-deficient pancreatic DM and fibro-calculous pancreatic diabetes (FCPD) (8). Likewise, a consensus statement from a workshop conducted in India also drew attention to those special types of DM peculiar to the tropics (6). Yet the most recent WHO classification of DM categorizes FCPD in the other specific types of DM (WHO 2019). Strangely, T2DM, which is the most common form of DM even in tropical regions has been overlooked when DM in the tropics is discussed. This review discusses the various aspects of all forms of DM in the tropics.

 

EPIDEMIOLOGY           

 

The prevalence of DM is on the rise globally, including in tropical regions. The epidemiology of all types of DM in the tropical region is as follows.

 

Type 1 Diabetes Mellitus

 

The global prevalence of T1DM in children and adolescents below 20 years is estimated to be more than one million (IDF). T1DM, caused by an autoimmune process, is influenced not only by genetic susceptibility but also to a greater extent by environmental factors. Among the multitude of environmental factors viral triggers, environmental toxins, exposure to exogenous antigens, and geographical factors play a very significant role in T1DM pathogenesis (7,8). The prevalence of disease follows a latitudinal gradient showing a direct relationship with the distance from the equator. Tropical regions show a lower prevalence while European countries exhibit a higher rate (9).

 

The incidence rates are highest in the northern European population and not surprisingly among the top ten countries based on incidence rates of T1DM, only one tropical country is included (IDF). The hygiene hypothesis as explained by prevalent helminthic infection in the tropics, intense ultraviolet exposure translating to improved vitamin D synthesis serving as an immune modulator, delayed exposure to cow’s milk and gluten thereby avoiding the allergen provoking beta cell autoimmunity are a few of the postulated protective mechanisms for T1DM in tropical regions (10–12). Data from temperate regions suggest the onset of T1DM occurs more often in winter while such differences were not shown in tropical countries (13,14). Likewise, the prevalence of anti-islet cell antibodies seems heterogenous in studies with some showing concordance to Western data while others showing higher antibody negative T1DM in tropical countries than in Western countries, although the number of antibodies tested vary between studies (15–17).

 

Type 2 Diabetes Mellitus

 

DM is estimated to affect around one-tenth of the adults aged 20-79 years of which the major type is T2DM. Tropical regions comprise almost 40 percent of the world’s diabetic load with six countries in the top ten countries with DM. Reports from the IDF also predict a rampant increase in the coming decades with maximum increment expected in the tropical regions (Africa, Middle East, South-East Asia, and South America). This increase in DM could be due to increasing life expectancy, improved access to health care, urbanization, and Westernization of lifestyle (18). Poorer lifestyle in the form of unhealthy food options, inaccessible recreational physical activity, and lack of awareness of healthy living accelerate the DM risk. Much evidence for T2DM in the tropics come from India which throws light on the distinguishing features from that of the Western population. The distribution of DM in developed nations is predominantly among the underprivileged strata whereas in developing countries including India, DM has been a disease of the affluent populations. However, it is bothersome to note that the prevalence is now increasing even among the lower socioeconomic strata. The nationwide study which investigated the epidemiology of DM in India showed that the prevalence was higher in urban in comparison to rural areas, although the rate of prediabetes is comparable in urban and rural areas thereby projecting that rural areas will reach the DM numbers akin to urban regions in the near future (ICMR) (19). Higher socioeconomic status remained a risk factor for DM in rural areas while the same was not true in urban areas reflecting the epidemiological transition in urban areas, probably owing to improved health awareness among higher socioeconomic strata. It was also shown that male sex is considered an independent risk factor for developing DM, which is at variance to what is shown in temperate zone showing female preponderance. The plausible explanation for this disparity between regions could be due to neglected healthcare among women in certain communities. Likewise, evidence from the same study did not favor smoking and alcohol as independent risk factors for DM, which contrast with the data from wealthier nations.

 

The incidence rate of DM among those with prediabetes in the Indian subcontinent is also one of the highest reported when compared to the Caucasian population yet comparable to the Pima Indians, Native Americans, and Micronesian population (20). Besides it is also interesting to note that Asian Indians develop DM at younger ages and at lesser obesity levels as compared to Western counterparts. This could be explained by virtue of South Asians having higher abdominal fat, more insulin resistance, and higher C-reactive protein levels despite lower body mass index (21,22). Among the individuals with prediabetes, prevalence of impaired fasting glucose is higher than that of impaired glucose tolerance in Asian Indians. This finding is in accordance with the evidence that an insulin secretory defect plays a significant role in T2DM pathogenesis in Indians compared to other ethnicities (1). This is also in line with the evidence from IDF which shows that age adjusted prevalence of impaired glucose tolerance is lowest in the southeast Asian region while that of impaired fasting glucose is considerably higher in southeast Asian region than other regions.

 

Hyperglycemia in Pregnancy

 

World estimates indicate that 16.7% of pregnancies are complicated by some form of hyperglycemia. More than 80% of this is due to gestational DM. While the majority of hyperglycemia in pregnancy is seen in low- and middle-income countries, the prevalence between tropical countries is vast with South-East Asian countries topping the prevalence list while Middle East countries and northern Africa show the least prevalence (IDF). Studies have also elucidated the possible effect of season and temperature on the prevalence of gestational DM with increased prevalence in summer and higher temperature (23,24). Factors like diet patterns including greater consumption of tropical fruits with moderate or high glycemic index have been postulated to increase the likelihood of gestational DM (25).

 

Maturity Onset Diabetes of the Young

 

The pioneering reports from Fajans and Tattersal described an intermediate form of DM different from the two classical forms of insulin dependent and non-insulin dependent DM (26). They coined the term Maturity Onset Diabetes of the young (MODY) showcasing its characteristics of age of onset below 25 years, absence of ketosis, and glycemic control without insulin for a minimum of at least 2 years. They also demonstrated an autosomal dominant inheritance (27). Since its recognition, reports confirm the existence of such youth onset DM which if different from the insulin dependent DM. The prevalence of this distinct form of DM also shows divergence between regions of the world.

 

Fibro Calculus Pancreatic Diabetes (FCPD)

 

Fibro calculus pancreatic diabetes (FCPD) is a rare but unique and unexplored type of DM found specifically in tropical countries including India, Indonesia, Bangladesh, Sri-Lanka, Brazil and a few African countries (28). FCPD was earlier called “tropical calcific pancreatitis (TCP)”. According to the American Diabetes Association (ADA) classification, DM originating secondary to any pancreatic origin is classified as type 3c DM. Chronic pancreatitis remains the most common etiology for the development of type 3c DM and chronic alcoholism in developed countries is a major cause of chronic pancreatitis contrasting with FCPD, which develops in the absence of alcohol intake.

 

FCPD is seen in the spectrum of DM associated with chronic pancreatitis and characterized by the presence of large calculi in the dilated pancreatic ducts along with significant fibrosis and atrophy of the gland. This leads to both exocrine and endocrine dysfunction. Population based studies of FCPD are scarce and the majority are reported from India. The reported population prevalence of FCPD was 0.019% among all DM patients (29). FCPD prevalence has declined from 1.6% in 1991-1995 to 0.2% during 2006-2010 while the BMI in FCPD increased (30). Similarly, Balakrishnan et al (31)found that only 3.8% among type 3c DM patients have FCPD, making chronic idiopathic chronic pancreatitis as the major contributor. The prevalence is approximately15% in young patients referred to a tertiary center (32). The declining prevalence of FCPD is principally attributed to an improvement in nutritional status  however the real cause remains to be determined.

 

GAPS AND CHALLENGES

 

Despite rising awareness of DM, the problem of ignorance about DM still exists. Data reveal the alarming fact that one in two adults with DM were ignorant about their condition. The majority of the undiagnosed cases occur in low- and middle-income countries. Likewise, the proportion of DM that is undiagnosed differs between regions. More than half of the patients living with DM in tropical regions like Africa and South-East Asia are undiagnosed while in European and North American countries the proportion undiagnosed is remarkably lower (IDF). Such high rates of undiagnosed cases reflect insufficient access to healthcare, poorer capacity of healthcare models, lack of pertinent diagnostic modalities, trained personnel, and inadequate patient health education. It should also be borne in mind that such remarkable levels of undiagnosed cases unquestionably impact morbidity and mortality because the later the diagnosis the higher the chances of disease complications. Additionally, people with a delayed diagnosis of DM, place an extra pressure on the healthcare structure due to higher complications (31).

 

Existing international guidelines for DM management are based on research conducted in developed countries. Extrapolating and applying such evidence to low- and middle-income countries may not be appropriate and requires a shift from being only developed country centric to more inclusive and international. Widespread effective patient awareness, modified affordable screening methods, appropriate diagnostic strategies, the need to diagnose people with DM earlier, and an increase in the coverage of preventive counselling is needed. In addition to diagnosing DM, efforts to diagnose complications earlier with non-invasive affordable screening tools could also improve outcomes (33,34). Lifestyle modification the most promising tool to prevent DM becomes the foremost inexpensive and best option in resource poor settings (35). DM takes a huge toll and is a major economic burden both to the individual as well as at the national level. The total DM related health expenditure has shown a steady rise over time, and this has been found to be lower in tropical regions as compared to temperate zones.

 

The North American and European regions display higher total DM related health expenditure while the tropical regions, despite having more than a third of the DM population are responsible for only one-tenth of the global DM related health expenditure. Along the same lines, DM becomes a major contributor to total health expenditure. In tropical regions like South America, Middle East, and North Africa expenditure due to DM contributes to almost one fifth of the total health expenditure while in Europe, DM expenditure as a proportion of total health expenditure is less than one-tenth. This can plausibly be explained by the fact that delayed diagnosis and greater chances of pre-existing complications result in higher expenditures. It is estimated that one third of all deaths from DM occur in the working age group which contributes to the economic burden. The Middle East and North Africa regions have the highest proportion of total deaths related to DM in the working age group. Similar challenges exist for T1DM as well with most developing countries reporting a very sub-optimal glycemic target achievement and control. Guidelines coordinating with existing government programs and primary care facilities aid in benefiting patients (36).

 

COMPLICATIONS

 

DM related mortality contributes to 12% of all-cause mortality in the 20-79 years age group. DM related morbidity also augments the economic burden of the disease globally. The microvascular and macrovascular complications of DM account for the majority of the morbidity & mortality. Although disease duration and glycemic control are important risk factors, ethnicity may also be a risk factor for complications.

 

Microvascular Complications

 

The prevalence of retinopathy varies between tropical countries. Indian studies report a prevalence ranging from 12-18% (37–39) which is lower than in Western cohorts. In contrast, data from Tanzania and other regions in Africa show a prevalence of 27-31% (70). In India, the prevalence of retinopathy at diagnosis was also strikingly lower among Indians with diabetes than seen in Western counterparts (40–43). Although duration of diabetes and glycemic control are consistent risk factors for retinopathy, diet patterns with increased antioxidants may serve as a probable protective factor (39).

 

Racial differences in the prevalence of diabetic nephropathy exist with Asian and African groups showing a higher nephropathy prevalence. Risk of end stage renal disease is higher in these populations than in Western populations (44). The prevalence of nephropathy ranges from 30-36% in various tropical regions (45,46) while one study from India reported a lower prevalence (47).

 

The prevalence of DM neuropathy is very heterogenous among different regions of the tropics. The prevalence estimates from Indian studies show lower figures in comparison to other tropical countries like Cuba, Mexico, Peru, and Caribbean countries (48–50). The different definitions used for DM neuropathy and characteristics of study populations may account for the differences in prevalence data. Asians when compared to Caucasians have a lower prevalence of neuropathy and possible explanations include lower smoking rates resulting in better peripheral vascularity and preserved skin micro-vascularization, and shorter height of Asians (increased height is a known risk factor for neuropathy).

 

Macrovascular Complications

 

Cardiovascular disease is the most common reason for mortality in people with DM. The pattern and prevalence vary between regions. The prevalence is less in tropical countries compared to Western countries, probably due to the relatively young population, lack of diagnostic facilities, and death due to other causes which prevail in most tropical countries (51). With Western countries now showing a progressive decrease in cardiovascular deaths, several tropical regions have reported a significant increase in cardiovascular mortality in recent decades (50,52). This could be attributable to the rampant Westernization with harmful transition in lifestyle increasing cardiovascular risk factors, growing population, and aging (53). Data from India show the susceptibility of Asian Indians to coronary artery disease. Asian Indians show early onset, more severe disease, and higher risk of mortality that Caucasians (54). DM by virtue of its insulin resistance and atherogenic dyslipidemia further aggravate this risk. On the contrary, peripheral vascular disease is comparatively rare among Indians. Younger age of onset of DM and lesser prevalence of smoking contributes to the decreased prevalence (55).

 

Diabetic Foot Ulcer & Tropical Diabetic Hand Syndrome

 

DM foot, a serious chronic complication of DM, shows an increasing prevalence worldwide owing to the rising DM prevalence and increased life expectancy. The prevalence of DM foot ulcer is heterogenous even between tropical regions with the Africa region showing higher caseloads than Asia and Australia. Still the overall prevalence is greater in North America and the reason for such differences could be increased prevalence of smoking among Americans than South Asians or poor screening processes in tropical countries (56). One of the important risk factors for developing a foot ulcer is barefoot walking, which prevails in most communities in Africa and Asia. Other risk factors, which are peculiar to these populations, include utilizing inappropriate footwear, more susceptible to rodent bites during farming activities, etc. (57). It is also shown that the duration between DM onset and onset of foot ulceration is shorter probably due to late diagnosis of DM (58). The bacteriology of foot infections also depends on climatic conditions with gram negative organism showing higher prevalence in the tropical and sub-tropical regions. DM foot ulcers increase healthcare costs, risk of amputation, and mortality (59). In the tropical regions, native practices to treat DM foot ulcers with plant parts remain widespread even in the modern era.

 

Tropical DM hand syndrome (TDHS) is a comparatively less recognized complication than DM foot. Since its earliest description from Africa, many cases have been reported in Africa and India (60). It is distinct from the DM hand syndrome where the latter predominantly involves joints and skin, leading to limited mobility. TDHS usually follows a trivial trauma and involves cellulitis ultimately progressing to fulminant sepsis or gangrene. Early aggressive antibiotic therapy with or without surgical intervention is needed for adequate management (61).

 

Acute Complications

 

Hyperglycemic emergencies constitute an important cause of emergency presentation of T1DM as well as T2DM. Up to a maximum of 80% of T1DM patients present with diabetic ketoacidosis (DKA) at diagnosis and this varies between countries. It has been shown that countries with higher background prevalence of T1DM have lower frequency of DKA at presentation with Sweden showing the lowest frequency of DKA at presentation while the United Arab Emirates and Saudi Arabia show the highest frequency. The same study showed that the frequency of DKA at presentation progressively decreases with increasing latitude thereby demonstrating a higher risk in tropical countries (62). Heat exposure has been shown to be associated with hyperglycemic emergencies through various mechanisms such as reduced insulin activity in insulin preparations exposed to high ambient temperatures, higher environmental temperature leading to increased counter regulatory hormones, higher risk of dehydration, and decreased thermoregulatory activity in the elderly (99). Interestingly higher environmental temperature is also an important risk factor for hypoglycemia. Asian countries also report a higher frequency of DKA among T2DM than Western populations (63). The mortality rates are also comparatively higher in tropical countries (64). It is also important to note that FCPD despite beta cell destruction, does not commonly lead to DKA episodes (65).

 

Diabetes & Infections

 

Uncontrolled DM is a well-known risk factor for infections and poor outcomes, due to altered immune responses (97). For some infections, there is evidence that poor glycemic control correlates with infection risk as well as severity. The following mechanisms confer increased susceptibility to infections in people with DM:

 

(i) Altered skin flora and increased risk of breach in integrity due to neuropathy and angiopathy (66)

(ii) Altered gut microbiome (67)

(iii) Impaired neutrophil function (68)

(iv) Impaired function of macrophages, T-cells, and NK cells (69)

(iv) Endothelial dysfunction, oxidative stress (70)

 

Some infections like rhino cerebral mucormycosis, Klebsiella pneumoniae related liver abscess, emphysematous pyelonephritis or cholecystitis, and Fournier’s gangrene are DM specific (71,72). Certain infections like candidiasis, bacterial pneumonia, urinary tract infections, skin and soft tissue infections, and bloodstream infections, although not exclusive for patients with DM are more common and severe among people with DM (73,74).

 

The increasing incidence and prevalence of DM in the tropics add to the infectious disease load and severity in the tropics. Infections are an important cause of morbidity and mortality in people with DM. Tropical regions are home to endemic infections like tuberculosis, dengue, melioidosis, leishmaniasis, helminthic, and parasitic infections. Patients with DM are affected out of proportion by tuberculosis, malaria, and Human immunodeficiency virus (HIV). The plausible reason for a higher risk of infections in people with DM in the tropics include:

 

(i) higher possibility of DM being undiagnosed or the diagnosis delayed

(ii) poorly controlled DM due to suboptimal management

(iii) co-existing malnutrition and poor hygiene

(iv) reduced access to healthcare facilities & infection care

 

Therefore, the tropical countries face a double disease burden, persisting communicable diseases and DM worsening the communicable disease burden. While DM, a proven risk factor for infections, confers higher rates of infection with common bacterial organisms in high income countries in tropical countries, in addition to higher rates of common organisms, the risk of tropical infections poses additional concerns.

 

BACTERIAL INFECTIONS

 

Among bacterial infections, tuberculosis has a bidirectional relationship with DM. People with uncontrolled DM have a three times higher risk of developing active tuberculosis, more atypical presentation, higher rate of treatment failure, and recurrence (75). Conversely active tuberculosis leads to stress hyperglycemia (76). Recent reports also suggest the number of patients of tuberculosis with coexisting DM exceeds the number of TB-HIV coinfection (77). Among the top 10 countries harboring global tuberculosis cases, most of the countries also show a high prevalence of DM (78). Understanding the impact of DM on tuberculosis, some countries have implemented collaborative interventions to improve detection of DM among patients with tuberculosis by screening all TB cases for DM (78). In contrast, screening all patients with DM for TB, despite being very important, still has practical difficulty owing to the limitations of available screening tests.

 

Melioidosis, another important tropical disease, is caused by Burkholderia pseudomallei, a gram-negative bacterium. DM increases the risk of melioidosis, which is usually prevalent in rice farming countries such as South-East Asia (79). With the rising prevalence of DM in tropical countries along with the increasing life expectancy, the burden of melioidosis may prove disastrous. Contradictory evidence also exists with regards to the impact of sulfonylurea treatment on the immune effects against melioidosis (80). DM serves as an independent risk factor for severity of scrub typhus, a rickettsial disease in tropical regions (81).

 

VIRAL INFECTIONS

 

Viral infections of significance in the tropics include dengue, arbovirus, severe acute respiratory syndrome, Middle East respiratory syndrome virus, and Ebola virus. Dengue, a mosquito borne infection, has shown a relation with DM. DM is associated with more severe dengue-induced thrombocytopenia, dengue shock syndrome, and higher risk of acute kidney injury (82–84). Similar evidence of DM predisposing to more severe chikungunya, West Nile fever disease does exist although such evidence on zika virus is inadequate (85,86). Likewise, DM is an important risk factor for Middle East Respiratory Syndrome (MERS) and is associated with higher mortality among severe acute respiratory syndrome (SARS) virus (87,88). Hepatitis B virus (HBV) also shows a close association with DM. Its prevalence is higher among people with DM, and DM is described to be associated with HBV disease progression. On the other hand, people with chronic HBV have an increased risk of developing DM. DM is more common among people living with HIV with data from tropical regions showing a more consistent and stronger association then those from high income countries (89,90). Advancements in retroviral therapy have transformed HIV infection from being associated with acquired immunodeficiency syndrome to a chronic disease associated with DM. The fact that DM in patients living with HIV develops at a much younger age than the general population is of great public health importance (61).

 

PARASITIC INFECTIONS

 

Malaria, caused by Plasmodium sp., is transmitted via mosquito bites. Africa accounts for the majority of cases. With co-existent increasing DM prevalence in Africa, a study from Ghana found that people with DM had a 46% increased risk of Plasmodium falciparum infection (91). Malaria infection during pregnancy is associated with intrauterine growth retardation, which in later life heightens the risk of insulin resistance and DM risk. DM is linked to an increased risk of leishmaniasis, whereas hyperglycemia was more common in patients with Chagas disease and cardiomyopathy than patients without cardiomyopathy (92,93). Interestingly a few helminthic infections like Schistosomiasis, round worm, and hook worm have a possible protective effect against DM (94).

 

FUNGAL INFECTIONS

 

Certain fungi are more frequent in the tropics than temperate regions due to the hotter and wetter conditions prevailing in the tropics. Fungal infections, typically the invasive ones, are also more common among immunocompromised individuals constituting opportunistic infections. Uncontrolled DM, a relatively immunocompromised state, and the climatic conditions of tropical regions favoring the prevalence of fungi, lead to fungal infections contributing importantly to the infection disease burden in the tropics. Mucormycosis caused by Zygomycetes, presents specifically as rhino-orbital-cerebral disease in people with DM. Although different forms of mucormycosis like pulmonary, gastrointestinal, and cutaneous types exist, rhino-orbital-cerebral mucormycosis is specifically associated with poorly controlled DM (95). Records from tropics show that among all patients with Mucormycosis, DM was seen in more than three-fourths of the patients. In the low-income countries, mortality due to mucormycosis is also higher than that in the developed countries owing to the shortcomings in medical and surgical management as well as poor glycemic control (96). Additionally, invasive aspergillus infections are also on the rise in the tropical regions.

 

FIBRO CALCULUS PANCREATIC DIABETES (FCPD)

 

Pathogenesis

 

The exact pathogenesis remains elusive. Factors like environmental toxins, nutrient deficiency, and genetic factors in combination may have a role.

 

(i) Environmental toxins: The most popular concept in the pathogenesis was the cassava hypothesis by McMillan and Geevarghese (4). Cassava contains cyanogenic glycosides. Cyanide detoxification in the body requires sulfur containing amino acids. In coexisting malnutrition, cyanide detoxification is impaired leading to pancreatic damage. Although cyanide ingestion in experiment rat models lead to transient hyperglycemia, permanent diabetes was not reported even with long term cassava consumption in rat models, thereby questioning this hypothesis (4). Geographic distribution of FCPD coincides with areas that consume cassava yet other areas where cassava consumption is not documented also have FCPD. The possible role of other cyanide containing foods such as jowar and sorghum may play a role.

 

(ii) Nutrient deficiency: The role of nutrient deficiency in the pathogenesis of FCPD has been a matter of debate. Nutrient deficiency could be the cause of as well as an effect of FCPD. Micronutrient deficiency and low vitamin C, vitamin E and beta carotene intake leading to oxidative stress may play a role in the etiology. Oxidative stress may also play a crucial role as evidenced by higher malondialdehyde levels and reduced antioxidant markers. Special interest with regards to selenium deficiency has been proposed. Western data showing that serum selenium levels were lower in those with chronic pancreatitis than in controls has kindled the hypothesis that lower selenium levels are associated with an accelerated course leading to DM in tropical pancreatitis. Yet a study comparing the selenium levels in healthy volunteers and patients with chronic pancreatitis in tropical and temperate regions did not confirm that selenium levels are involved in the DM that occurs in association with chronic pancreatitis (97).

 

(iii) Genetic factors: Studies have shown a familial aggregation, observed in up to one-tenth of cases (91) (98). Alterations in genes such as serum protease inhibitor Kazal type (SPINK1), cationic trypsinogen (PRSS1), anionic trypsinogen (PRSS2), and chymotrypsinogen C have been highlighted in FCPD cases.

 

Overall, no one factor is responsible for the pathogenesis while probable involvement of multiple factors may explain the occurrence. The pathogenesis of DM includes defective insulin secretion as well as insulin resistance in FCPD. Deficiency of pancreatic polypeptide and storage of triglyceride in liver due to reduced fat store, contributes to insulin resistance (65).

 

Unfortunately, there is no specific etiology identified in FCPD despite several proposed theories. The initial explanation of cassava (tapioca) induced injury to the pancreatic acini through cyanide generation is flawed by the fact that not all with cassava intake develop the disease (6,7). A diet pattern of high carbohydrate and low protein is also implicated but the underlying mechanism is not known. Perhaps, genetic predisposition partly explains the FCPD etiology. A recent study has shown that 62.5 percent of FCPD patients harbor variation in the serine protease inhibitor Kazal type 1 (SPINK1) gene , particularly the N34S polymorphism (8). The role of other genes like PRSS1, PRSS2, CFTR, CTSB are not fully elucidated.

 

Apart from pancreatitis development, how diabetes develops is also not understood. Insulin deficiency and beta cell dysfunction are the primary pathology found. However, newer evidence suggests that altered glucagon dynamics, incretin abnormalities, and the presence of abnormal body composition leading to selective insulin resistance may contribute to the development of diabetes in FCPD (1,9) (99).

 

Clinical Features and Diagnosis of FCPD

 

The clinical features of FCPD are unique. The natural history usually progresses through three distinct stages. The initial period, where recurrent abdomen pain is the main clinical feature, often occurs in adolescents or early second decades. The second stage is characterized by classical exocrine pancreatic insufficiency with resultant steatorrhea symptoms. Steatorrhea is characterized by recurrent diarrhea with bulky, foul smelling, greasy stool, and predominant symptoms of fat malabsorption. Subsequently, fat related vitamin deficiency (A,D,E,K) features ensue. Other macro and micronutrients deficiencies are also present. In the third stage, that usually occurs in the late second to third decade, frank hyperglycemia occurs. FCPD patients are classically lean and malnourished. They have very brittle DM with high glycemic variability that is difficult to control.

 

Abdominal pain is conspicuously absent or less in intensity and frequency in the third stage, but the full-blown picture of both endocrine and exocrine deficiency persists. 50% of patients with FCPD without DM at baseline develop DM after 5 years of follow-up (100), mostly at the third decade, however the percentage is even higher as age advances. The diagnostic criteria proposed by the Mohan et al encompasses all the clinical features described (See Table 1, Adapted from reference) (101). Despite a very high glucose, FCPD patients do not develop diabetic ketoacidosis (DKA). The reasons proposed are : 1) simultaneous destruction of pancreatic alpha cells leading to absence of glucagon which is a crucial hormone for ketogenesis in the liver. This is coupled with absence of absolute insulin deficiency in FCPD, as compared to T1DM, preventing lipolysis; 2) these patients are chronically malnourished and have very low free fatty acid reserve thus adequate substrate for ketone generation is usually absent and 3) carnitine deficiency as a part of generalized malnutrition as carnitine is required for the mitochondrial beta oxidation (28,101).

 

TABLE 1. DIAGNOSTIC CRITERIA FOR FCPD

1.     Occurrence in a tropical country

2.     Diabetes as per standard diagnostic criteria

3.     Evidence of chronic pancreatitis:

a.     Pancreatic calculi on X-ray or

b.     At least 3 of the following:

i.     Abnormal pancreatic morphology by imaging

ii.     Chronic abdominal pain since childhood

iii.     Steatorrhea

c.     Abnormal pancreatic function test

4.     Absence of other causes of pancreatitis like alcoholism, hyperparathyroidism, marked hypertriglyceridemia, hepatobiliary disease etc.

Table from the Endotext chapter entitled Fibrocalculus Pancreatic Diabetes

 

The diagnosis of FCPD is mostly clinical and supported by imaging. Since FCPD has an asymptomatic course when the pancreatic injury has happened and frank glycemia is not present, the diagnosis is often delayed and there is an unmet need for screening in such patients. The classical patient is a lean malnourished patient with severe hyperglycemia requiring multiple insulin doses. Abdominal imaging, particularly computed tomography (CT) scans are helpful to diagnose pancreatic pathology. The CT hallmark is 1) pancreatic duct dilation, 2) large pancreatic calculi involving the major ducts and 3) pancreatic atrophy and fibrosis.  These features differ from alcoholic chronic pancreatitis where the pancreatic stones are smaller and have speckled pattern and involves smaller pancreatic ducts (101). However, other common causes of pancreatitis like alcohol, hyperparathyroidism, gallstones, and hypertriglyceridemia should be ruled out in such patients.

 

Despite having high glycemic variability and an elevated HbA1C, FCPD patients are at lower risk of micro and macrovascular complications compared to classical (T2DM). In a study from the Southern part of India it was shown that the prevalence of coronary artery disease, cerebrovascular stroke, and retinopathy was significantly higher in the T2DM patients compared to FCPD patients confirming this notion (102). This difference is possibly related to the absence of insulin resistance and other risk factors like obesity and dyslipidemia in FCPD patients, but further work is required to better understand this dichotomy. Nevertheless, all patients should undergo careful investigation for the routine micro and macrovascular complications. Periodontal disease is common in FCPD similar to that seen in T2DM and the severity correlates with poor glycemia (103). Hypoglycemia unawareness  is found in 73% of FCPD patients and classically is related to the lower fasting and post prandial glucagon levels in a subset of patients and contributes to the higher glycemic variability (104).

 

It is important to evaluate pancreatic exocrine insufficiency by fecal elastase estimation. However, fat malabsorptive features may be absent in tropical countries since the diet may be low in fat. Other abnormalities such as high triglycerides during hyperglycemia can be seen. Pancreatic amylase and lipase are not elevated in the chronic phase but may rise if an acute attack is present. Pancreatic carcinoma is a dreaded long-term complication of FCPD. Usually, pancreatic carcinoma develops much earlier, around the 5th decade in these patients and is diagnosed at an advanced stage (105). Unexplained weight loss and sudden deterioration in glycemic control along with abdominal pain distinct from the usual pancreatitis pain, warrants urgent investigation for pancreatic carcinoma (101).

 

Management and Challenges in FCPD

 

The mainstay of management of hyperglycemia is insulin. However, the doses may be quite variable and require frequent adjustment. Sometimes metformin and sulphonylureas are sufficient to control glycemia in milder cases. Lack of evidence of management of FCPD is a concern and should be addressed urgently. Incretin mimetics like DPP-4 inhibitors and GLP-1 analogues should be used cautiously if at all in FCPD. Ideally a closed loop system for continuous subcutaneous insulin delivery coupled with continuous glucose monitoring should be used whenever possible in FCPD patients but need further studies for this recommendation. Recent studies have shown the impact of SGLT-2 inhibitors in pancreatectomized patients, and hence they can be an important choice in FCPD patients, but one must be cautious about potential weight loss (106).

 

With documented exocrine pancreatic insufficiency in FCPD, pancreatic enzyme replacement therapy (PERT) may help improve glycemia and hence should be considered in patients with chronic pancreatitis (107). The usual dose is 10,000-25,000 lipase units and requires escalation to higher dose up to 50,000 per meal or less for snacks. The capsules should be spread throughout the meal to maximize the benefits. Usually, dietary fat restrictions are not advised but a high fiber diet may aggravate abdominal symptoms in FCPD, thus better avoided. Fat soluble vitamin replacement is necessary. Eventually some patients having refractory pain require either endoscopic or open surgical procedures for drainage. Unfortunately, there is no therapy to halt the progression from the pancreatitis phase to the FCPD phase, however antifibrotic treatment like pirfenidone has shown some effect in experimental preclinical studies (108).

 

REFERENCES

 

  1. Gujral UP, Narayan KMV, Kahn SE, Kanaya AM. The relative associations of β-cell function and insulin sensitivity with glycemic status and incident glycemic progression in migrant Asian Indians in the United States: the MASALA study. J Diabetes Complications. J Diabetes Complications. 2014;28(1):45-50. Available from: https://pubmed.ncbi.nlm.nih.gov/24211090/
  2. Hugh-Jones P. Diabetes in Jamaica. Lancet. 1955;269(6896):891–7. Available from: https://pubmed.ncbi.nlm.nih.gov/13264638/
  3. Tripathy BB, Kar BC. Observations on Clinical Patterns of Diabetes Mellitus in India. Diabetes. 1965;14:404-412. Available from: http://diabetesjournals.org/ diabetes/article-pdf/14/7/404/338504/14-7-404.pdf
  4. McMillan DE, Geevarghese PJ. Dietary cyanide and tropical malnutrition diabetes. Diabetes Care. 1979;2(2):202-208. Available from: https://pubmed.ncbi.nlm.nih.gov/574813/
  5. Tulloch JA, Macintosh D. “J”-type diabetes. Lancet. 1961;2(7194):119-121.Available from: https://pubmed.ncbi.nlm.nih.gov/13778587/
  6. Hoet JJ, Tripathy BB. Report of the International Workshop on types of Diabetes Peculiar to the Tropics. Diabetes Care. 1996;19(9):1014. Available from: https://pubmed.ncbi.nlm.nih.gov/8875102/
  7. Zorena K, Michalska M, Kurpas M, Jaskulak M, Murawska A, Rostami S. Environmental Factors and the Risk of Developing Type 1 Diabetes-Old Disease and New Data. Biology (Basel). 2022;11(4):608. Available from: https://pubmed.ncbi.nlm.nih.gov/35453807/
  8. Alruhaili M, Alruhaili MH. Type 1 diabetes in the tropics: the protective effects of environmental factors. African Journal of Diabetes Medicine. 2010;18(2):1-4.
  9. Mohr SB, Garland CF, Gorham ED, Garland FC. The association between ultraviolet B irradiance, vitamin D status and incidence rates of type 1 diabetes in 51 regions worldwide. Diabetologia. 2008;51(8):1391-1398. Available from: https://pubmed.ncbi.nlm.nih.gov/18548227/
  10. Zaccone P, Fehervari Z, Phillips JM, Dunne DW, Cooke A. Parasitic worms and inflammatory diseases. Parasite Immunol. 2006;28(10):515-523. Available from: https://pubmed.ncbi.nlm.nih.gov/16965287/
  11. Virtanen SM, Räsänen L, Aro A, Lindström J, Sippola H, Lounamaa R, et al. Infant feeding in Finnish children less than 7 yr of age with newly diagnosed IDDM. Childhood Diabetes in Finland Study Group. Diabetes Care. 1991;14(5):415-417. Available from: https://pubmed.ncbi.nlm.nih.gov/2060453/
  12. Norris JM, Barriga K, Klingensmith G, Hoffman M, Eisenbarth GS, Erlich HA, et al. Timing of Initial Cereal Exposure in Infancy and Risk of Islet Autoimmunity. JAMA. 2003;290(13):1713-1720. Available from: https://jamanetwork.com/ journals/jama/fullarticle/197392
  13. Variation and trends in incidence of childhood diabetes in Europe. EURODIAB ACE Study Group. [published correction appears in Lancet 2000 Nov 11;356(9242):1690]. Lancet. 2000;355(9207):873-876. Available from: https://pubmed.ncbi.nlm.nih.gov/10752702/
  14. Basu M, Banerjee M, Mukhopadhyay P, Ghosh S. Trends of Seasonality and Age of onset in T1DM: A snapshot from Eastern India. Indian J Endocrinol Metab. 2020;24(2):219-220. Available from: /pmc/articles/PMC7333758/
  15. Basu M, Pandit K, Banerjee M, Mondal SA, Mukhopadhyay P, Ghosh S. Profile of Auto-antibodies (Disease Related and Other) in Children with Type 1 Diabetes. Indian J Endocrinol Metab. 2020;24(3):256-259. Available from: /pmc/articles/PMC7539037/
  16. Balasubramanian K, Dabadghao P, Bhatia V, Colman PG, Gellert SA, Bharadwaj U, et al. High frequency of type 1B (idiopathic) diabetes in North Indian children with recent-onset diabetes. Diabetes Care. 2003;26(9):2697. Available from: https://pubmed.ncbi.nlm.nih.gov/12941746/
  17. Shivaprasad C, Mittal R, Dharmalingam M, Kumar P. Zinc transporter-8 autoantibodies can replace IA-2 autoantibodies as a serological marker for juvenile onset type 1 diabetes in India. Indian J Endocrinol Metab. 2014;18(3):345-9. Available from: /pmc/articles/PMC4056133/
  18. van Crevel R, van de Vijver S, Moore DAJ. The global diabetes epidemic: what does it mean for infectious diseases in tropical countries? Lancet Diabetes Endocrinol. 2017 Jun;5(6):457-468.
  19. Mohan V, Anjana RM, Tandon N. Lessons Learnt from the ICMR-INDIAB Study. Natl Med J India. 2023;36(3):137-139. Available from: https://pubmed.ncbi.nlm.nih.gov/38692603/
  20. Anjana RM, Rani CSS, Deepa M, Pradeepa R, Sudha V, Nair HD, et al. Incidence of diabetes and prediabetes and predictors of progression among Asian Indians: 10-year follow-up of the Chennai urban rural epidemiology study (CURES). Diabetes Care. 2015;38(8):1441-1448.
  21. Unnikrishnan R, Anjana RM, Mohan V. Diabetes in South Asians: Is the Phenotype Different? Diabetes. 2014;63(1):53-55. Available from: https://dx.doi.org/10.2337/db13-1592
  22. Ramachandran A, Snehalatha C, Naik RAS, Mohan V, Shobana R, Viswanathan M. Significance of impaired glucose tolerance in an Asian Indian population: a follow-up study. Diabetes Res Clin Pract. 1986;2(3):173-178. Available from: https://pubmed.ncbi.nlm.nih.gov/3527626/
  23. Retnakaran R, Ye C, Kramer CK, Hanley AJ, Connelly PW, Sermer M, et al. Impact of daily incremental change in environmental temperature on beta cell function and the risk of gestational diabetes in pregnant women. Diabetologia. 2018;61(12):2633-2642. Available from: https://pubmed.ncbi.nlm.nih.gov /30112689/
  24. Wang P, Wu CS, Li CY, Yang CP, Lu MC. Seasonality of gestational diabetes mellitus and maternal blood glucose levels: Evidence from Taiwan. Medicine (Baltimore). 2020;99(41):e22684. Available from: https://pubmed. ncbi.nlm.nih.gov/33031338/
  25. Huang WQ, Lu Y, Xu M, Huang J, Su YX, Zhang CX. Excessive fruit consumption during the second trimester is associated with increased likelihood of gestational diabetes mellitus: a prospective study. Sci Rep. 2017;7:43620. Available from: https://www.nature.com/articles/srep43620
  26. Tattersall RB. Mild familial diabetes with dominant inheritance. Q J Med. 1974;43(170):339-357. Available from: https://pubmed.ncbi. nlm.nih.gov/4212169/
  27. Tattersall RB, Fajans SS. A difference between the inheritance of classical juvenile-onset and maturity-onset type diabetes of young people. Diabetes. 1975;24(1):44-53. Available from: https://pubmed.ncbi.nlm.nih.gov/1122063/
  28. Dasgupta R, Naik D, Thomas N. Emerging concepts in the pathogenesis of diabetes in fibrocalculous pancreatic diabetes. J Diabetes. 2015;7(6):754-761. Available from: https://pubmed.ncbi.nlm.nih.gov/25707547/
  29. Mohan V, Farooq S, Deepa M. Prevalence of fibrocalculous pancreatic diabetes in Chennai in South India. JOP. 2008;9(4):489-492. Available from: https://pubmed.ncbi.nlm.nih.gov/18648140/
  30. Papita R, Nazir A, Anbalagan VP, et al. Secular trends of fibrocalculous pancreatic diabetes and diabetes secondary to alcoholic chronic pancreatitis at a tertiary care diabetes centre in South India. JOP. 2012;13(2):205-209. Available from: https://pubmed.ncbi.nlm.nih.gov/22406602/
  31. Balakrishnan V, Unnikrishnan AG, Thomas V, Choudhuri G, Veeraraju P, Singh SP, et al. Chronic pancreatitis. A prospective nationwide study of 1,086 subjects from India. JOP. 2008;9(5):593-600. Available from: https://europepmc.org/ article/med/18762690
  32. Sahoo S, Zaidi G, Vipin V, Chapla A, Thomas N, Yu L, et al. Heterogeneity in the aetiology of diabetes mellitus in young adults: A prospective study from north India. Indian J Med Res. 2019;149(4):479-488. Available from: https://pubmed.ncbi.nlm.nih.gov/31411171/
  33. Banerjee M, Mukhopadhyay P, Basu M, Ghosh S. Corneal Confocal Microscopy Identifies Structural Small Fibre Abnormalities in an Adolescent with Type 1 Diabetes and Impaired Awareness of Hypoglycaemia. J ASEAN Fed Endocr Soc. 2023;38(2):128-130. Available from: https://asean-endocrinejournal.org/ index.php/JAFES/article/view/2689
  34. Banerjee M, Mukhopadhyay P, Ghosh S, Basu M, Pandit A, Malik R, et al. Corneal Confocal Microscopy Abnormalities in Children and Adolescents With Type 1 Diabetes. Endocr Pract. 2023;29(9):692-698. Available from: https://pubmed.ncbi.nlm.nih.gov/37343765/
  35. Echouffo-Tcheugui JB, Dagogo-Jack S. Preventing diabetes mellitus in developing countries. [published correction appears in Nat Rev Endocrinol. 2012 Nov;8(11):692]. Nat Rev Endocrinol. 2012;8(9):557-562. Available from: https://pubmed.ncbi.nlm.nih.gov/22488646/
  36. Yasmin M, Mukhopadhyay P, Ghosh S. Model of care for Type 1 diabetes in India: Integrated approach for its incorporation in future national health care policy. Lancet Reg Health Southeast Asia. 2022;3:100014.Available from: http://www.thelancet.com/article/S2772368222000142/fulltext
  37. Raman R, Rani PK, Reddi Rachepalle S, Gnanamoorthy P, Uthra S, Kumaramanickavel G, et al. Prevalence of diabetic retinopathy in India: Sankara Nethralaya Diabetic Retinopathy Epidemiology and Molecular Genetics Study report 2. Ophthalmology. 2009;116(2):311-318. Available from: https://pubmed.ncbi.nlm.nih.gov/19084275/
  38. Pradeepa R, Rema M, Vignesh J, Deepa M, Deepa R, Mohan V. Prevalence and risk factors for diabetic neuropathy in an urban south Indian population: the Chennai Urban Rural Epidemiology Study (CURES-55). Diabet Med. 2008;25(4):407-412. Available from: https://pubmed.ncbi.nlm.nih.gov/ 18294224/
  39. Rema M, Pradeepa R. Diabetic retinopathy: an Indian perspective. Indian J Med Res. 2007;125(3):297-310. Available from: https://pubmed.ncbi.nlm.nih.gov/ 17496357/
  40. Kohner EM, Aldington SJ, Stratton IM, Manley SE, Holman RR, Matthews DR, et al. United Kingdom Prospective Diabetes Study, 30: diabetic retinopathy at diagnosis of non-insulin-dependent diabetes mellitus and associated risk factors. Arch Ophthalmol. 1998;116(3):297-303. Available from: https://pubmed.ncbi.nlm.nih.gov/9514482/
  41. Harris MI, Klein R, Welborn TA, Knuiman MW. Onset of NIDDM occurs at least 4-7 yr before clinical diagnosis. Diabetes Care. 1992;15(7):815-819. Available from: https://pubmed.ncbi.nlm.nih.gov/1516497/
  42. Ramachandran A, Snehalatha C, Vijay V, Viswanathan M. Diabetic retinopathy at the time of diagnosis of NIDDM in South Indian subjects. Diabetes Res Clin Pract. 1996;32(1-2):111-114.
  43. Scanlon PH, Nevill CR, Stratton IM, Maruti SS, Massó-González EL, Sivaprasad S, et al. Prevalence and incidence of diabetic retinopathy (DR) in the UK population of Gloucestershire. Acta Ophthalmol. 2022;100(2):e560-e570. Available from: https://pubmed.ncbi.nlm.nih.gov/34180581/
  44. Young BA, Maynard C, Boyko EJ. Racial differences in diabetic nephropathy, cardiovascular disease, and mortality in a national population of veterans. Diabetes Care. 2003;26(8):2392-2399. Available from: https://pubmed.ncbi.nlm.nih.gov/12882868/
  45. Wagnew F, Eshetie S, Kibret GD, Zegeye A, Dessie G, Mulugeta H, et al. Diabetic nephropathy and hypertension in diabetes patients of sub-Saharan countries: a systematic review and meta-analysis. BMC Res Notes. 2018;11(1):565. Available from: https://pubmed.ncbi.nlm.nih.gov/30081966/
  46. Chandie Shaw PK, Baboe F, Van Es LA, Van Der Vijver JC, Van De Ree MA, De Jonge N, et al. South-Asian type 2 diabetic patients have higher incidence and faster progression of renal disease compared with Dutch-European diabetic patients. Diabetes Care. 2006;29(6):1383-1385. Available from: https://pubmed.ncbi.nlm.nih.gov/16732026/
  47. Unnikrishnan I R, Rema M, Pradeepa R, Deepa M, Shanthirani CS, Deepa R, et al. Prevalence and Risk Factors of Diabetic Nephropathy in an Urban South Indian PopulationThe Chennai Urban Rural Epidemiology Study (CURES 45). Diabetes Care. 2007;30(8):2019-2024. Available from: https://dx.doi.org/ 10.2337/dc06-2554
  48. Yovera-Aldana M, Velasquez-Rimachi V, Huerta-Rosario A, More-Yupanqui MD, Osores-Flores M, Espinoza R, et al. Prevalence and incidence of diabetic peripheral neuropathy in Latin America and the Caribbean: A systematic review and meta-analysis. PLoS One. 2021;16(5):e0251642. Available from: /pmc/articles/PMC8118539/
  49. Dutta A, Naorem S, Singh TP, Wangjam K. Prevalence of Peripheral Neuropathy In Newly Diagnosed Type 2 Diabetics Mellitus. Int J Diab Dev Countries. 2005;25:30-33.
  50. Sacco RL, Roth GA, Reddy KS, Arnett DK, Bonita R, Gaziano TA, et al. The Heart of 25 by 25: Achieving the Goal of Reducing Global and Regional Premature Deaths From Cardiovascular Diseases and Stroke: A Modeling Study From the American Heart Association and World Heart Federation. Circulation. 2016;133(23):e674-e690. Available from: https:// pubmed.ncbi.nlm.nih.gov/27162236/
  51. Kengne AP, Amoah AGB, Mbanya JC. Cardiovascular complications of diabetes mellitus in sub-Saharan Africa. Circulation. 2005;112(23):3592-3601. Available from: https://pubmed.ncbi.nlm.nih.gov/16330701/
  52. Gouda HN, Charlson F, Sorsdahl K, Ahmadzada S, Ferrari AJ, Erskine H, et al. Burden of non-communicable diseases in sub-Saharan Africa, 1990-2017: results from the Global Burden of Disease Study 2017. Lancet Glob Health. 2019;7(10):e1375-e1387. Available from: https://pubmed.ncbi.nlm. nih.gov/31537368/
  53. Minja NM, Nakagaayi D, Aliku T, Zhang W, Ssinabulya I, Nabaale J, et al. Cardiovascular diseases in Africa in the twenty-first century: Gaps and priorities going forward. Front Cardiovasc Med. 2022;9:1008335. Available from: https://pubmed.ncbi.nlm.nih.gov/36440012/
  54. Mohan V, Venkatraman JV, Pradeepa R. Epidemiology of cardiovascular disease in type 2 diabetes: the Indian scenario. Diabetes Sci Technol. 2010;4(1):158-170. Available from: https://pubmed.ncbi.nlm.nih.gov/20167181/
  55. Pradeepa R, Chella S, Surendar J, Indulekha K, Anjana RM, Mohan V. Prevalence of peripheral vascular disease and its association with carotid intima-media thickness and arterial stiffness in type 2 diabetes: the Chennai urban rural epidemiology study (CURES 111). Diab Vasc Dis Res. 2014;11(3):190-200. Available from: https://pubmed.ncbi.nlm.nih.gov/ 24627461/
  56. Zhang P, Lu J, Jing Y, Tang S, Zhu D, Bi Y. Global epidemiology of diabetic foot ulceration: a systematic review and meta-analysis. Ann Med. 2017;49(2):106-116. Available from: https://pubmed.ncbi.nlm.nih.gov/27585063/
  57. Abbas ZG, Boulton AJM. Diabetic foot ulcer disease in African continent: ‘From clinical care to implementation’ - Review of diabetic foot in last 60 years - 1960 to 2020. Diabetes Res Clin Pract. 2022;183:109155. Available from: https://pubmed.ncbi.nlm.nih.gov/34838640/
  58. Okunola O, Akinwusi P, Kolawole B, Oluwadiya K. Diabetic foot ulcer in a Tropical setting: Presentation and outcome. Nigerian Endo Practice. 2012;6(1):27–31. Available from: https://www.ajol.info/index.php/nep/ article/view/84844
  59. Rabin B, Lockwood SM, Martinson E, Urquhart-Foster K, Bhanushali P, Raymond J, et al. 2901. Associations between climate and diabetic foot infection microbiology: a systematic review and meta-analysis. Open Forum Infect Dis. 2023;10(2):99-101.
  60. Abbas ZG, Archibald LK. Tropical diabetic hand syndrome. Epidemiology, pathogenesis, and management. Am J Clin Dermatol. 2005;6(1):21-28. Available from: https://pubmed.ncbi.nlm.nih.gov/15675887/
  61. Tiwari S, Chauhan A, Sethi NT. Tropical diabetic hand syndrome. Int J Diabetes Dev Ctries. 2008 Oct;28(4):130-1. Available from: /pmc/articles/PMC2822157/
  62. Usher-Smith JA, Thompson M, Ercole A, Walter FM. Variation between countries in the frequency of diabetic ketoacidosis at first presentation of type 1 diabetes in children: a systematic review. Diabetologia. 2012;55(11):2878-2894. Available from: https://pubmed.ncbi.nlm.nih.gov/22933123/
  63. Tan H, Zhou Y, Yu Y. Characteristics of diabetic ketoacidosis in Chinese adults and adolescents -- a teaching hospital-based analysis. Diabetes Res Clin Pract. 2012;97(2):306-312. Available from: https://pubmed.ncbi.nlm.nih.gov/ 22704172/
  64. Musoma SN, Omar A, Mutai BC, Laigong P. Outcomes of Children and Adolescents Admitted with Diabetic Ketoacidosis at Kenyatta National Hospital (KNH), Kenya. J Diabetes Res. 2020;2020:8987403.
  65. Unnikrishnan A, Kumaran S, Kalra S. Fibrocalculous Pancreatic Diabetes. Endotext [Updated 2024 Sep 13]. In: Feingold KR, Anawalt B, Blackman MR, et al., editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000- Available from: https://www.ncbi.nlm.nih.gov/books/NBK578126/
  66. Ahluwalia A, Sood A, Sood A, Lakshmy R, Kapil A, Pandey RM. Nasal colonization with Staphylococcus aureus in patients with diabetes mellitus. Diabet Med. 2000;17(6):487-488. Available from: https://pubmed.ncbi. nlm.nih.gov/10975221/
  67. Forslund K, Hildebrand F, Nielsen T, Falony G, Le Chatelier E, Sunagawa S, et al. Disentangling type 2 diabetes and metformin treatment signatures in the human gut microbiota. Nature. 2017 May 3;545(7652):116. Available from: https://pubmed.ncbi.nlm.nih.gov/26633628/
  68. Hulme KD, Gallo LA, Short KR. Influenza Virus and Glycemic Variability in Diabetes: A Killer Combination? Front Microbiol. 2017;8:861. Available from: https://pubmed.ncbi.nlm.nih.gov/28588558/
  69. Hodgson K, Morris J, Bridson T, Govan B, Rush C, Ketheesan N. Immunological mechanisms contributing to the double burden of diabetes and intracellular bacterial infections. Immunology. 2015;144(2):171-185. Available from: https://pubmed.ncbi.nlm.nih.gov/25262977/
  70. Roberts AC, Porter KE. Cellular and molecular mechanisms of endothelial dysfunction in diabetes. Diab Vasc Dis Res. 2013;10(6):472-482. Available from: https://pubmed.ncbi.nlm.nih.gov/24002671/
  71. Singh V, Sharma B, Sen R, Agrawal S, Bhagol A, Bali R. Rhinocerebral mucormycosis: a diagnostic challenge and therapeutic dilemma in immunocompetent host. J Oral Maxillofac Surg. 2012;70(6):1369-1375. Available from: https://pubmed.ncbi.nlm.nih.gov/21864966/
  72. Siu LK, Yeh KM, Lin JC, Fung CP, Chang FY. Lancet Infect Dis. 2012;12(11):881-887. Available from: https://pubmed.ncbi.nlm.nih. gov/23099082/
  73. Hirji I, Guo Z, Andersson SW, Hammar N, Gomez-Caminero A. Incidence of urinary tract infection among patients with type 2 diabetes in the UK General Practice Research Database (GPRD). J Diabetes Complications. 2012;26(6):513-516. Available from: https://pubmed.ncbi.nlm. nih.gov/22889712/
  74. Kornum JB, Thomsen RW, Riis A, Lervang HH, Schønheyder HC, Sørensen HT. Type 2 diabetes and pneumonia outcomes: a population-based cohort study. Diabetes Care. 2007;30(9):2251-2257. Available from: https://pubmed.ncbi.nlm.nih.gov/17595354/
  75. Jeon CY, Murray MB. Diabetes mellitus increases the risk of active tuberculosis: a systematic review of 13 observational studies. PLoS Med. 2008;5(7):e152. Available from: https://pubmed.ncbi.nlm.nih.gov/18630984/
  76. Magee MJ, Salindri AD, Kyaw NTT, Auld SC, Haw JS, Umpierrez GE. Stress Hyperglycemia in Patients with Tuberculosis Disease: Epidemiology and Clinical Implications. Curr Diab Rep. 2018;18(9):71. Available from: https://pubmed.ncbi.nlm.nih.gov/30090969/
  77. Restrepo BI. Diabetes and Tuberculosis. Schlossberg D, editor. Microbiol Spectr. 2016;4(6).10.1128. Available from: https://pubmed.ncbi. nlm.nih.gov/28084206/
  78. World Health Organization (WHO) - Global tuberculosis report 2023. Available from: https://iris.who.int/.
  79. Jenjaroen K, Chumseng S, Sumonwiriya M, Ariyaprasert P, Chantratita N, Sunyakumthorn P, et al. T-Cell Responses Are Associated with Survival in Acute Melioidosis Patients. PLoS Negl Trop Dis. 2015;9(10):e0004152. Available from: https://pubmed.ncbi.nlm.nih.gov/26495852/
  80. Liu X, Foo G, Lim WP, Ravikumar S, Sim SH, Win MS, et al. Sulphonylurea usage in melioidosis is associated with severe disease and suppressed immune response. PLoS Negl Trop Dis. 2014;8(4):e2795. Available from: https://pubmed.ncbi.nlm.nih.gov/24762472/
  81. Park SW, Lee CS, Lee CK, Kwak YG, Moon C, Kim BN, et al. Severity predictors in eschar-positive scrub typhus and role of serum osteopontin. Am J Trop Med Hyg. 2011;85(5):924-30. Available from: https://pubmed.ncbi.nlm. nih.gov/22049051/
  82. Mehta P, Hotez PJ. NTD and NCD Co-morbidities: The Example of Dengue Fever. PLoS Negl Trop Dis. 2016;10(8):e0004619. Available from: https://pubmed.ncbi.nlm.nih.gov/27561091/
  83. Htun NSN, Odermatt P, Eze IC, Boillat-Blanco N, D’Acremont V, Probst-Hensch N. Is diabetes a risk factor for a severe clinical presentation of dengue?--review and meta-analysis. PLoS Negl Trop Dis. 2015;9(4):e0003741. Available from: https://pubmed.ncbi.nlm.nih.gov/25909658/
  84. Toledo J, George L, Martinez E, Lazaro A, Han WW, Coelho GE, et al. Relevance of Non-communicable Comorbidities for the Development of the Severe Forms of Dengue: A Systematic Literature Review. PLoS Negl Trop Dis. 2016;10(1):e0004284. Available from: https://pubmed.ncbi.nlm. nih.gov/26727113/
  85. Lindsey NP, Staples JE, Lehman JA, Fischer M. Medical risk factors for severe West Nile Virus disease, United States, 2008-2010. Am J Trop Med Hyg. 2012;87(1):179-84. Available from: https://pubmed.ncbi.nlm.nih.gov/22764311/
  86. Jean-Baptiste E, Von Oettingen J, Larco P, Raphaël F, Larco NC, Cauvin MM, et al. Chikungunya Virus Infection and Diabetes Mellitus: A Double Negative Impact. Am J Trop Med Hyg. 2016;95(6):1345-1350. Available from: https://pubmed.ncbi.nlm.nih.gov/27729569/
  87. Yang JK, Feng Y, Yuan MY, Yuan SY, Fu HJ, Wu BY, et al. Plasma glucose levels and diabetes are independent predictors for mortality and morbidity in patients with SARS. Diabet Med. 2006;23(6):623-8. Available from: https://pubmed.ncbi.nlm.nih.gov/16759303/
  88. Assiri A, Al-Tawfiq JA, Al-Rabeeah AA, Al-Rabiah FA, Al-Hajjar S, Al-Barrak A, et al. Epidemiological, demographic, and clinical characteristics of 47 cases of Middle East respiratory syndrome coronavirus disease from Saudi Arabia: a descriptive study. Lancet Infect Dis. 2013;13(9):752-61. Available from: https://pubmed.ncbi.nlm.nih.gov/23891402/
  89. 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(S1):27-39. Available from: https://pubmed.ncbi.nlm.nih.gov/25117959/
  90. Lo YC, Chen MY, Sheng WH, Hsieh SM, Sun HY, Liu WC, et al. Risk factors for incident diabetes mellitus among HIV-infected patients receiving combination antiretroviral therapy in Taiwan: a case-control study. HIV Med. 2009;10(5):302-309. Available from: https://pubmed.ncbi.nlm.nih.gov/19220492/
  91. Danquah I, Bedu-Addo G, Mockenhaupt FP. Type 2 diabetes mellitus and increased risk for malaria infection. Emerg Infect Dis. 2010;16(10):1601-1604. Available from: https://pubmed.ncbi.nlm.nih.gov/20875289/
  92. dos Santos VM, da Cunha SF, Teixeira Vde P, et al. Freqüência de diabetes mellitus e hiperglicemia em mulheres chagásicas e não-chagásicas [Frequency of diabetes mellitus and hyperglycemia in chagasic and non-chagasic women]. Rev Soc Bras Med Trop. 1999;32(5):489-496. Available from: https://pubmed.ncbi.nlm.nih.gov/10881081/
  93. Sharquie KE, Najim RA, Hussein AK. Reinfestation in cutaneous leishmaniasis: a new look at predisposing conditions. Saudi Med J. 2000;21(5):464-467. Available from: https://pubmed.ncbi.nlm.nih.gov/11500682/
  94. Cooke A, Tonks P, Jones FM, O’Shea H, Hutchings P, Fulford AJC, et al. Infection with Schistosoma mansoni prevents insulin dependent diabetes mellitus in non-obese diabetic mice. Parasite Immunol. 1999;21(4):169-176. Available from: https://pubmed.ncbi.nlm.nih.gov/10320614/
  95. Taborda CP, Muñoz JE, Gonzalez A. Editorial: Tropical fungal diseases. Front Cell Infect Microbiol. 2022;12:1104519.
  96. Singhal I, Arora M, Dave A, Saluja P. Diabetes and Fungal Infection - A Didactic Relationship. J clin and diag res. 2023;17(4):1-4.
  97. Yadav S, Day JP, Mohan V, Snehalatha C, Braganza JM. Selenium and Diabetes in the Tropics. Pancreas. 1991;6(5):528-533.
  98. Mohan V, Chari ST, Hitman GA, Suresh S, Madanagopalan N, Ramachandran A, et al. Familial aggregation in tropical fibrocalculous pancreatic diabetes. Pancreas. 1989;4(6):690-693. Available from: https://pubmed.ncbi.nlm. nih.gov/2813331/
  99. Ghosh I, Mukhopadhyay P, Das K, Anne M B, Ali Mondal S, Basu M, et al. Incretins in fibrocalculous pancreatic diabetes: A unique subtype of pancreatogenic diabetes. J Diabetes. 2021;13(6):506-511.
  100. Mohan V, Barman KK, Rajan VS, Chari ST, Deepa R. Natural history of endocrine failure in tropical chronic pancreatitis: a longitudinal follow-up study. J Gastroenterol Hepatol. 2005;20(12):1927-1934. Available from: https://pubmed.ncbi.nlm.nih.gov/16336455/
  101. Unnikrishnan R, Mohan V. Fibrocalculous Pancreatic Diabetes. Current Diabetes Reports. 2020;20:19.
  102. Shivaprasad C, Anish K, Aiswarya Y, Atluri S, Rakesh B, Anupam B, et al. A comparative study of the clinical profile of fibrocalculous pancreatic diabetes and type 2 diabetes mellitus. Diabetes Metab Syndr. 2019;13(2):1511-1516. Available from: https://pubmed.ncbi.nlm.nih.gov/31336514/
  103. Beatrice Anne M, Chakraborty P, Mukhopadhyay P, Ghosh S. Periodontal disease in fibrocalculous pancreatic diabetes (FCPD): common complication of an uncommon disease. Int J Diabetes Dev Ctries. 2023;43(5):709–14. Available from: https://link.springer.com/article/10.1007/s13410-022-01148-2
  104. Dasgupta R, Jebasingh FK, Anoop S, Seenivasan S, Kurian ME, Christina F, et al. Comprehensive evaluation of patterns of hypoglycemia unawareness (HUA) and glycemic variability (GV) in patients with fibrocalculous pancreatic diabetes (FCPD): A cross-sectional study from South India. PLoS One. 2022;17(7):e0270788. Available from: /pmc/articles/PMC9275701/
  105. Chari ST, Mohan V, Pitchumoni CS, Viswanathan M, N. Madanagopalan, Lowenfels AB. Risk of pancreatic carcinoma in tropical calcifying pancreatitis: an epidemiologic study. Pancreas. 1994;9(1):62-66. Available from: https://pubmed.ncbi.nlm.nih.gov/8108373/
  106. Baekdal M, Nielsen SW, Hansen CP, Storkholm JH, Van Hall G, Hartmann B, et al. Empagliflozin Normalizes Fasting Hyperglycemia and Improves Postprandial Glucose Tolerance in Totally Pancreatectomized Patients: A Randomized, Double-Blind, Placebo-Controlled Crossover Study. Diabetes Care. 2024;47(1):71-80. Available from: https://pubmed.ncbi.nlm.n nih.gov/37703527/
  107. Ghosh I, Basu M, Anne B, Mukhopadhyay P, Ghosh S. Exocrine Pancreatic Dysfunction in Diabetes: An Observational Study. Indian J Endocrinol Metab. 2021;25(1):67-68. Available from: /pmc/articles/PMC8323623/
  108. Palathingal Bava E, George J, Iyer S, Sahay P, Tarique M, Jain T, et al. Pirfenidone ameliorates chronic pancreatitis in mouse models through immune and cytokine modulation. Pancreatology. 2022;22(5):553-563. Available from: https://pubmed.ncbi.nlm.nih.gov/35570091/

Genetic Obesity Syndromes

ABSTRACT

 

Genetic factors play a major role in the regulation of body weight and in the susceptibility to obesity in the population. A subset of people with severe obesity carry rare, highly penetrant genetic variants that can result in severe childhood-onset obesity. Current estimates suggest that up to 20% of children with severe obesity may carry pathogenic chromosomal abnormalities or mutations. The diagnosis of a genetic obesity syndrome can provide information that has value for the patient and their family and may help them deal with the social stigma that comes with severe obesity in childhood. In an increasing number of cases, the finding of a genetic cause for a patient’s obesity can inform clinical care and the use of targeted therapies.

 

INTRODUCTION

 

At a population level, obesity is driven by an increase in the intake of easily available, energy-dense, highly palatable foods and a decrease in physical activity at school/work and in leisure time. However, variation in body mass index (BMI: weight in kg/height in meters squared) within the population is influenced by genetic factors (1,2). Studies in twins and families have shown that food intake, satiety responsiveness (fullness after a fixed meal), basal metabolic rate, the amount of energy utilized during a fixed amount of exercise, and body fat distribution are all heritable traits (3).

 

Genome-wide association studies (GWAS’s) in large population-based cohorts have identified thousands of common variants (minor allele frequency > 5%) that are associated with BMI and/or obesity (4). While individually each variant often has a small effect on BMI, cumulatively, the effect of millions of common and rare variants can now be combined to compute a polygenic risk score which can predict the development of severe obesity (5). Studies are ongoing to test how such scores might be useful in the clinical setting. Most common obesity associated variants lie in noncoding areas of the genome so identifying the mechanism by which they affect body weight can be challenging. It is interesting to note that most obesity- or BMI-associated variants lie in or near genes which are expressed in the brain and some of these variants have been associated with increased food intake (4,6). In contrast, GWAS associations for body fat distribution/waist-to-hip ratio, mostly seem to be linked to genes expressed in adipose tissue (7).

 

CLINICAL APPROACH TO DIAGNOSIS OF GENETIC OBESITY SYNDROMES

 

Rare (less than 1% minor allele frequency), highly penetrant genetic variants in multiple genes have been associated with severe obesity that often presents in childhood. Whilst these disorders are rare, cumulatively up to 20% of children with severe obesity have chromosomal abnormalities or other penetrant rare variants that drive their obesity (8). The assessment of children and adults with severe obesity should be directed at screening for endocrine, neurological, and genetic disorders (9). Important information can be obtained from a detailed family history to identify potential consanguineous relationships, the presence of other family members with severe obesity and those who have had bariatric surgery, and the ethnic origin of family members (Figure 1). The clinical history and examination can then guide the appropriate use of diagnostic tests. For the purposes of clinical assessment, it remains useful to categorize the genetic obesity syndromes as those with and without associated developmental delay.

 

Figure 1. Diagnosis of genetic obesity syndromes.

 

OBESITY SYNDROMES WITHOUT DEVELOPMENTAL DELAY

 

The adipocyte-derived hormone leptin acts mainly to defend against starvation (10), with a fall in leptin levels (as seen in weight loss, acute caloric restriction or congenital leptin deficiency) causing an increase in food intake and physiological responses that act to restore energy balance (11). In most people, circulating leptin levels correlate closely with fat mass (12), although there is considerable variation in leptin levels at any given BMI, which is as yet unexplained. Leptin signals through the long isoform of the leptin receptor, which is widely expressed in the hypothalamus and other brain regions involved in energy homeostasis (13). In the arcuate nucleus of the hypothalamus (which has a permeable blood-brain barrier), there are several neuronal populations known to be important in weight regulation expressing the leptin receptor. In the fed state, leptin stimulates the expression of pro-opiomelanocortin (POMC), which is processed to generate the melanocortin peptides that, in turn, activate the melanocortin 4 receptor (MC4R) on second-order neurons in the paraventricular nucleus. Leptin also inhibits adjacent neurons containing Agouti-related protein (AgRP), a MC4R antagonist. The integration of these two actions leads to reduced food intake (Figure 2). In the fasted state and with weight loss, a drop in leptin levels reduces the activation state of POMC neurons and increases AgRP signaling to cause an increase in food intake. These hypothalamic pathways interact with other brain centers to affect not just eating behaviors but also energy expenditure.

 

Severe obesity can result from mutations that disrupt key components of the leptin-melanocortin pathway (Figure 2). People with these genetic disorders experience an intense drive to eat (hunger), find food to be highly rewarding, and have impaired fullness (satiety) leading to hyperphagia (increased food intake), resulting in excessive weight gain from early childhood.

 

Figure 2. Genes involved in the leptin-melanocortin pathway whose disruption causes obesity.

 

Leptin and Leptin Receptor Deficiency

 

Congenital leptin (LEP protein; LEP gene) and leptin receptor (LEPR protein; LEPR gene) deficiency are rare, autosomal recessive disorders associated with severe obesity from a very young age (before 1 year) (14,15). Homozygous frameshift, nonsense, and missense mutations involving LEP and LEPR have been identified in 1% and 2-3% of patients with severe obesity from consanguineous families, respectively (16-18). Leptin receptor mutations have been found in some non-consanguineous families, where both parents were unrelated but carried rare heterozygous variants.

 

Serum leptin is a useful test in patients with severe early onset obesity as an undetectable serum leptin suggests a diagnosis of congenital leptin deficiency. Very rare mutations that result in a detectable but bio-inactive form of leptin or a form of leptin that antagonizes the leptin receptor, have also been described (19,20). Serum leptin concentrations are appropriate for the degree of obesity in leptin receptor deficiency and as such an elevated serum leptin concentration is not necessarily a predictor of leptin receptor deficiency (17). In some patients, particular LEPR mutations that result in abnormal cleavage of the extracellular domain of LEPR (which then acts as a leptin binding protein), are associated with markedly elevated leptin levels (15).

 

The clinical phenotypes associated with leptin and leptin receptor deficiencies are similar. Patients are born of normal birth weight, experience intense hyperphagia with food seeking behavior and rapid weight gain in the first few months of life resulting in severe obesity (16). While measurable changes in resting metabolic rate or total energy expenditure have not been demonstrated in affected individuals, reduced sympathetic nerve function is associated with impaired fat oxidation and may contribute to obesity (21). Children with leptin deficiency have abnormalities of T cell number and function (16), consistent with reported high childhood infection rates and childhood mortality from infection, particularly in environments where infectious diseases are prevalent (21).

 

In keeping with severe obesity, patients with leptin and leptin receptor deficiency are hyperinsulinemic and some adults develop type 2 diabetes in the 3rd to 4th decade. Affected individuals can exhibit hypothalamic (secondary) hypothyroidism characterized by low free thyroxine levels and inappropriately normal (or high-normal) levels of serum thyroid stimulating hormone (TSH) (14,15). Typically, adults with leptin or leptin receptor deficiency have biochemical evidence of hypogonadotropic hypogonadism and do not undergo normal pubertal development (see below). However, there are reports of delayed spontaneous onset of menses in some leptin and leptin receptor deficient adults (17). Linear growth is appropriate in childhood, but in the absence of a pubertal growth spurt, final height is reduced.

 

Although leptin deficiency is very rare, it is entirely treatable with daily subcutaneous injections of recombinant human leptin (16,22). The major effect of leptin replacement in these patients is on food intake, with normalization of hyperphagia and enhanced satiety. Leptin administration does not enhance energy expenditure. However, weight loss by caloric restriction is associated with decreased total energy expenditure; the absence of this decrease in patients with congenital leptin deficiency, suggests that leptin does affect energy expenditure (23). Leptin replacement permits progression of appropriately-timed pubertal development, along with expression of secondary sexual characteristics (21). These reproductive system effects are likely mediated through leptin action on hypothalamic neurons containing kisspeptin, which signals via GPR54 to modify the release of gonadotrophin-releasing hormone (24).

 

Although leptin treatment is not be effective for patients with LEPR deficiency, these patients can now treated with a melanocortin receptor agonist (setmelanotide, Figure 3), which is now licensed in the UK, Europe and USA (25).  

 

Leptin treatment is not clinically effective in people with common obesity (26,27), which may be a manifestation of leptin resistance or defects in downstream neuronal pathways. Studies in heterozygous carriers of LEP mutations who have partial leptin deficiency and an increase in fat mass (28), suggest that people with relatively low leptin levels may benefit from leptin therapy.

 

Pro-opiomelanocortin Deficiency

 

Due to impaired production of melanocortin stimulating hormone peptides (a/b MSH) and diminished or absent MC4R signaling (Figure 2), homozygous or compound heterozygous mutations in POMC cause hyperphagia and early-onset obesity (29). People deficient in POMC also have pale skin and red or light colored hair due to the lack of signaling of pigment-inducing melanocortin 1 receptors in the skin (29). In the pituitary gland, POMC is the precursor for adrenocorticotrophin (ACTH). As such, complete POMC deficiency presents in neonatal life with features of ACTH and cortisol deficiency: hypoglycemia and cholestatic jaundice requiring long-term corticosteroid replacement therapy (30). Typically, patients with primary or secondary cortisol deficiency present with hypophagia and weight loss, so adrenal insufficiency with hyperphagia in the absence of a structural hypothalamic abnormality should raise suspicion for a POMC defect. POMC deficiency may also impair the timing of puberty, an effect that appears to be mediated by the melanocortin 3 receptor (MC3R) (31).

 

Complete POMC deficiency can be treated with a melanocortin receptor agonist (setmelanotide) (32) (Figure 3). Heterozygous missense mutations directly affecting the function of POMC peptides have been described (Figure 2) (33). These variants significantly increase obesity risk but are not invariably associated with obesity. The potential efficacy of MC4R agonists in patients with these heterozygous mutations is currently being tested in clinical trials.

 

Figure 3. Medical treatment of patients with genetic obesity syndromes. POMC: pro-opiomelanocortin. PCSK1: prohormone convertase-1. MC4R: melanocortin 4 receptor. GLP-1: glucagon receptor-1.

 

Prohormone Convertase-1-Deficiency

 

Prohormone convertase-1 (PCSK1, also known as PC1/3) is an enzyme that acts upon a range of substrates including proinsulin, proglucagon, and POMC. Compound heterozygous or homozygous mutations in PCSK1 cause neonatal small bowel enteropathy, glucocorticoid deficiency (secondary to ACTH deficiency), hypogonadotropic hypogonadism, and postprandial hypoglycemia due to impaired processing of proinsulin to insulin, as well as severe, early onset obesity (34,35). Elevated plasma levels of proinsulin and 32/33 split proinsulin in the context of low levels of mature insulin are diagnostic for this disorder. Setmelanotide is now licensed for the treatment of this condition (Figure 3).

 

Melanocortin 4 Receptor Deficiency

 

Heterozygous melanocortin 4 receptor (MC4R) mutations have been reported in people with obesity from various ethnic groups (www.mc4r.org.uk) and occur at a frequency of 1 in 300 people in the population (36), 1% of adults with a BMI > 30 kg/m2, and 3-5% of children with severe obesity (37,38). MC4R mutations are inherited in a co-dominant manner, with variable penetrance and expression; homozygous mutations have also been reported. In several studies, MC4R deficiency is the most common genetic form of obesity (37-39).

 

Given the importance of MC4R for leptin signaling (Figure 2), the clinical features of MC4R deficiency include hyperphagia and rapid weight gain, which often emerges in the first few years of life. Alongside the increase in fat mass, MC4R-deficient subjects also have an increase in lean mass and a marked increase in bone mineral density that exceeds what would be expected for their increased body size and, thus, they often appear “big-boned” (37). They exhibit accelerated linear growth in early childhood, which may be a consequence of disproportionate early hyperinsulinemia and effects on pulsatile growth hormone (GH) secretion, which is retained in MC4R-deficient adults in contrast to common forms of obesity (40). Despite this early hyperinsulinemia, adult subjects with obesity who are heterozygous for mutations in the MC4R gene have a comparable risk of developing impaired glucose intolerance and type 2 diabetes to controls of similar age and adiposity. Reduced sympathetic nervous system activity in MC4R-deficient patients is likely to explain the lower prevalence of hypertension and lower systolic and diastolic blood pressures compared to control populations (41). Thus, central melanocortin signaling appears to play an important role in the regulation of blood pressure and its coupling to changes in weight.

 

At present, there is no specific therapy for MC4R deficiency, but patients with heterozygous MC4R mutations do respond to Glucagon-like peptide- (GLP-1) receptor agonists (42) and to Roux-en-Y-bypass surgery (43) (Figure 3), with a variation in weight loss response that is comparable to people with a normal MC4R gene sequence.

 

Albright’s Hereditary Osteodystrophy/Pseudohypoparathyroidism

 

Albright hereditary osteodystrophy (AHO) is an autosomal dominant disorder due to germline mutations in GNAS, an imprinted gene that encodes the G alpha s (Gs) protein, which mediates signaling by multiple G-protein coupled receptors (GPCRs). Classically, heterozygous loss-of-function mutations in GNAS affecting the maternal allele lead to short stature, obesity, skeletal defects, and resistance to several hormones that activate Gs in their target tissues (pseudohypoparathyroidism type IA), while paternal transmission leads only to the AHO phenotype (pseudopseudohypoparathyroidism) (44). GNAS mutations affect coupling to, or signaling by, MC4R, which explains hyperphagia and obesity in affected patients (45). Some patients will carry mutations that affect signaling by other GPCRs including the beta-2 and beta-3 adrenoreceptors which contribute to low basal metabolic rate and other clinical phenotypes (45). These patients may not have classical features such as short stature. As such, this diagnosis should be considered in all patients with severe early-onset obesity (45).

 

SRC Homology 2B (SH2B1) 1 Deficiency

 

Deletion of a 220-kb segment of chromosome 16p11.2 is associated with highly penetrant, severe, early-onset obesity and insulin resistance (46). This deletion includes a small number of genes, one of which is SH2B1 (Src homology 2B1)known to be involved in leptin, insulin, and Brain-Derived Neurotrophic Factor (BDNF) signaling. These patients gain weight in the first years of life, with hyperphagia and fasting plasma insulin levels that are disproportionately elevated, with increased risk for type 2 diabetes in early adulthood (47). In some patients loss of function mutations in the SH2B1 gene have also been reported in association with early-onset obesity, severe insulin resistance, and behavioral abnormalities (48).

 

OBESITY SYNDROMES WITH DEVELOPMENTAL DELAY

 

Prader-Willi Syndrome

 

Prader-Willi syndrome is an autosomal dominant disorder caused by deletion or disruption of a paternally imprinted region on chromosome 15q11.2-q12 (49) The clinical features of Prader-Willi syndrome (PWS) include diminished fetal activity, hypotonia, and feeding difficulties in infancy followed by hyperphagia, obesity, developmental delay, short stature, hypogonadotropic hypogonadism, and small hands and feet (Figure 4). Children with PWS display diminished growth, reduced lean body mass and increased fat mass. These body composition abnormalities can be explained, in part, by growth hormone (GH) deficiency and improved with growth hormone treatment, which should be started in early childhood.

 

Figure 4. Infancy and childhood clinical features of Prader-Willi Syndrome (PWS).

 

Contained within the 4.5Mb PWS region in 15q11-q13 are silenced paternally imprinted genes and a family of small nucleolar RNAs (snoRNAs) known as the HBII-85 snoRNAs. Small deletions exclusively encompassing these snoRNAs result in the key features of PWS including obesity (Figure 4) (50,51) suggesting that these snoRNAs play a critical role in the development of this syndrome. Histopathological studies on post-mortem brain samples from PWS patients have demonstrated reduced levels of oxytocin expression in the hypothalamus (52) and trials of intranasal administration in PWS are ongoing (53). Brain-derived neurotrophic factor (BDNF) expression is also reduced in PWS, potentially contributing to both the obesity and neurobehavioral features including stereotyped behaviors (54).

 

Bardet Biedl Syndrome

 

Bardet-Biedl syndrome (BBS) is a rare, autosomal recessive disease caused by mutations in over 25 genes and characterized by obesity, developmental delay, syndactyly, brachydactyly or polydactyly, retinal dystrophy or pigmentary retinopathy, hypogonadism, and structural abnormalities of the kidney or renal impairment (55). The differential diagnosis includes Biemond syndrome II (iris coloboma, hypogenitalism, obesity, polydactyly) and Alstrom syndrome (retinitis pigmentosa, obesity, diabetes mellitus, and deafness). To date, BBS proteins are all involved in basal body and centrosomal function and impact on ciliary development and transport (56). There is some evidence that BBS genes affect leptin signaling and trafficking of MC4Rs in cilia. Clinical trials of setmelanotide have shown some benefit in treating hyperphagia in these patients (57) and this drug is licensed for BBS in some countries (Figure 3).

 

 

Brain-Derived neurotrophic factor (BDNF) activates signaling by the tropomycin-related kinase B (TrkB) to play a key role in the development and maintenance of neurons. Rare chromosomal rearrangements and heterozygous point mutations in BDNF and TrkB are associated with speech and language delay, hyperphagia, and impaired pain sensation (58-60). Disordered behaviors including hyperactivity, fearlessness, anxiety, and aggression are also features of these conditions, which can often present as de-novo genetic abnormalities (61).

 

Single Minded 1 Deficiency

 

Single minded 1 (SIM1) is a transcription factor involved in the development of the paraventricular and supraoptic nuclei of the hypothalamus. Chromosomal rearrangements and heterozygous missense mutations in SIM1 and in a closely related transcription factor OTP (Orthopedia) cause severe obesity (62-64). Clinical features of these patients resemble those seen in MC4R deficiency with, in addition, a variable phenotype of developmental delay with autistic like features noted in some, but not all, patients (63).

 

Other Rare Genetic Mutations

 

Rare penetrant variants in multiple genes can be associated with, but do not invariably cause, obesity that is inherited in a classical Mendelian manner (Figure 2). Examples include heterozygous loss of function variants affecting the Semaphorin 3 ligands, receptors and co-receptors that direct the development of POMC projections (65); mutations in Steroid receptor coactivator-1 (SRC-1) (66) and Pleckstrin-homology-domain interacting protein, PHIP, which modulate POMC transcription (67); disruption of Serotonin 2c receptor, HTR2C which regulates the electrical activity of POMC neurons causing obesity, social anxiety and impaired memory (68,69) and deletions affecting TRPC5 on the X chromosome, which cause obesity, anxiety, autism (in males), and postnatal depression (in females) (70). Variants in genes that regulate MC4R trafficking (MRAP2) (71) and genes whose precise function in the hypothalamus is not as yet clear, such as Kinase Suppressor of Ras-2 (KSR2) (72) have also been associated with obesity.

 

FUTURE PERSPECTIVES

 

The diagnosis of a genetic obesity syndrome can provide information that has diagnostic value for the family to whom genetic counselling can be provided. A genetic diagnosis can help children and their families deal with the social stigma that comes with severe obesity and, in some instances, has prevented children from being taken into care by social services when obesity is blamed on parental neglect. A genetic diagnosis can inform management (many such patients are relatively refractory to weight loss through changes in diet and exercise) and can inform clinical decision-making. For example, bariatric surgery (particularly Roux-en-Y bypass surgery) is contraindicated in many genetic obesity syndromes as it does not reverse the strong hypothalamic drive to eat and continued overeating can be harmful. Importantly, an increasing number of genetic obesity syndromes are now treatable with mechanism-based pharmacologic therapies.

 

REFERENCES    

 

  1. Maes HH, Neale MC, Eaves LJ. Genetic and environmental factors in relative body weight and human adiposity. Behav Genet. 1997;27(4):325-351.
  2. Stunkard AJ HJ, Pedersen NL, McClearn GE. The body-mass index of twins who have been reared apart. N Engl J Med. 1990;322:1483-1487.
  3. Bouchard C. Genetics of Obesity: What We Have Learned Over Decades of Research. Obesity (Silver Spring).2021;29(5):802-820.
  4. Loos RJF, Yeo GSH. The genetics of obesity: from discovery to biology. Nature reviews Genetics. 2022;23(2):120-133.
  5. Khera AV, Chaffin M, Wade KH, Zahid S, Brancale J, Xia R, Distefano M, Senol-Cosar O, Haas ME, Bick A, Aragam KG, Lander ES, Smith GD, Mason-Suares H, Fornage M, Lebo M, Timpson NJ, Kaplan LM, Kathiresan S. Polygenic Prediction of Weight and Obesity Trajectories from Birth to Adulthood. Cell. 2019;177(3):587-596 e589.
  6. Locke AE, Kahali B, Berndt SI, Justice AE, Pers TH, Day FR, Powell C, Vedantam S, Buchkovich ML, Yang J, Croteau-Chonka DC, Esko T, Fall T, Ferreira T, Gustafsson S, Kutalik Z, Luan J, Magi R, Randall JC, Winkler TW, Wood AR, Workalemahu T, Faul JD, Smith JA, Zhao JH, Zhao W, Chen J, Fehrmann R, Hedman AK, Karjalainen J, Schmidt EM, Absher D, Amin N, Anderson D, Beekman M, Bolton JL, Bragg-Gresham JL, Buyske S, Demirkan A, Deng G, Ehret GB, Feenstra B, Feitosa MF, Fischer K, Goel A, Gong J, Jackson AU, Kanoni S, Kleber ME, Kristiansson K, Lim U, Lotay V, Mangino M, Leach IM, Medina-Gomez C, Medland SE, Nalls MA, Palmer CD, Pasko D, Pechlivanis S, Peters MJ, Prokopenko I, Shungin D, Stancakova A, Strawbridge RJ, Sung YJ, Tanaka T, Teumer A, Trompet S, van der Laan SW, van Setten J, Van Vliet-Ostaptchouk JV, Wang Z, Yengo L, Zhang W, Isaacs A, Albrecht E, Arnlov J, Arscott GM, Attwood AP, Bandinelli S, Barrett A, Bas IN, Bellis C, Bennett AJ, Berne C, Blagieva R, Bluher M, Bohringer S, Bonnycastle LL, Bottcher Y, Boyd HA, Bruinenberg M, Caspersen IH, Chen YI, Clarke R, Daw EW, de Craen AJM, Delgado G, Dimitriou M, Doney ASF, Eklund N, Estrada K, Eury E, Folkersen L, Fraser RM, Garcia ME, Geller F, Giedraitis V, Gigante B, Go AS, Golay A, Goodall AH, Gordon SD, Gorski M, Grabe HJ, Grallert H, Grammer TB, Grassler J, Gronberg H, Groves CJ, Gusto G, Haessler J, Hall P, Haller T, Hallmans G, Hartman CA, Hassinen M, Hayward C, Heard-Costa NL, Helmer Q, Hengstenberg C, Holmen O, Hottenga JJ, James AL, Jeff JM, Johansson A, Jolley J, Juliusdottir T, Kinnunen L, Koenig W, Koskenvuo M, Kratzer W, Laitinen J, Lamina C, Leander K, Lee NR, Lichtner P, Lind L, Lindstrom J, Lo KS, Lobbens S, Lorbeer R, Lu Y, Mach F, Magnusson PKE, Mahajan A, McArdle WL, McLachlan S, Menni C, Merger S, Mihailov E, Milani L, Moayyeri A, Monda KL, Morken MA, Mulas A, Muller G, Muller-Nurasyid M, Musk AW, Nagaraja R, Nothen MM, Nolte IM, Pilz S, Rayner NW, Renstrom F, Rettig R, Ried JS, Ripke S, Robertson NR, Rose LM, Sanna S, Scharnagl H, Scholtens S, Schumacher FR, Scott WR, Seufferlein T, Shi J, Smith AV, Smolonska J, Stanton AV, Steinthorsdottir V, Stirrups K, Stringham HM, Sundstrom J, Swertz MA, Swift AJ, Syvanen AC, Tan ST, Tayo BO, Thorand B, Thorleifsson G, Tyrer JP, Uh HW, Vandenput L, Verhulst FC, Vermeulen SH, Verweij N, Vonk JM, Waite LL, Warren HR, Waterworth D, Weedon MN, Wilkens LR, Willenborg C, Wilsgaard T, Wojczynski MK, Wong A, Wright AF, Zhang Q, LifeLines Cohort S, Brennan EP, Choi M, Dastani Z, Drong AW, Eriksson P, Franco-Cereceda A, Gadin JR, Gharavi AG, Goddard ME, Handsaker RE, Huang J, Karpe F, Kathiresan S, Keildson S, Kiryluk K, Kubo M, Lee JY, Liang L, Lifton RP, Ma B, McCarroll SA, McKnight AJ, Min JL, Moffatt MF, Montgomery GW, Murabito JM, Nicholson G, Nyholt DR, Okada Y, Perry JRB, Dorajoo R, Reinmaa E, Salem RM, Sandholm N, Scott RA, Stolk L, Takahashi A, Tanaka T, van 't Hooft FM, Vinkhuyzen AAE, Westra HJ, Zheng W, Zondervan KT, Consortium AD, Group A-BW, Consortium CAD, Consortium CK, Glgc, Icbp, Investigators M, Mu TC, Consortium MI, Consortium P, ReproGen C, Consortium G, International Endogene C, Heath AC, Arveiler D, Bakker SJL, Beilby J, Bergman RN, Blangero J, Bovet P, Campbell H, Caulfield MJ, Cesana G, Chakravarti A, Chasman DI, Chines PS, Collins FS, Crawford DC, Cupples LA, Cusi D, Danesh J, de Faire U, den Ruijter HM, Dominiczak AF, Erbel R, Erdmann J, Eriksson JG, Farrall M, Felix SB, Ferrannini E, Ferrieres J, Ford I, Forouhi NG, Forrester T, Franco OH, Gansevoort RT, Gejman PV, Gieger C, Gottesman O, Gudnason V, Gyllensten U, Hall AS, Harris TB, Hattersley AT, Hicks AA, Hindorff LA, Hingorani AD, Hofman A, Homuth G, Hovingh GK, Humphries SE, Hunt SC, Hypponen E, Illig T, Jacobs KB, Jarvelin MR, Jockel KH, Johansen B, Jousilahti P, Jukema JW, Jula AM, Kaprio J, Kastelein JJP, Keinanen-Kiukaanniemi SM, Kiemeney LA, Knekt P, Kooner JS, Kooperberg C, Kovacs P, Kraja AT, Kumari M, Kuusisto J, Lakka TA, Langenberg C, Marchand LL, Lehtimaki T, Lyssenko V, Mannisto S, Marette A, Matise TC, McKenzie CA, McKnight B, Moll FL, Morris AD, Morris AP, Murray JC, Nelis M, Ohlsson C, Oldehinkel AJ, Ong KK, Madden PAF, Pasterkamp G, Peden JF, Peters A, Postma DS, Pramstaller PP, Price JF, Qi L, Raitakari OT, Rankinen T, Rao DC, Rice TK, Ridker PM, Rioux JD, Ritchie MD, Rudan I, Salomaa V, Samani NJ, Saramies J, Sarzynski MA, Schunkert H, Schwarz PEH, Sever P, Shuldiner AR, Sinisalo J, Stolk RP, Strauch K, Tonjes A, Tregouet DA, Tremblay A, Tremoli E, Virtamo J, Vohl MC, Volker U, Waeber G, Willemsen G, Witteman JC, Zillikens MC, Adair LS, Amouyel P, Asselbergs FW, Assimes TL, Bochud M, Boehm BO, Boerwinkle E, Bornstein SR, Bottinger EP, Bouchard C, Cauchi S, Chambers JC, Chanock SJ, Cooper RS, de Bakker PIW, Dedoussis G, Ferrucci L, Franks PW, Froguel P, Groop LC, Haiman CA, Hamsten A, Hui J, Hunter DJ, Hveem K, Kaplan RC, Kivimaki M, Kuh D, Laakso M, Liu Y, Martin NG, Marz W, Melbye M, Metspalu A, Moebus S, Munroe PB, Njolstad I, Oostra BA, Palmer CNA, Pedersen NL, Perola M, Perusse L, Peters U, Power C, Quertermous T, Rauramaa R, Rivadeneira F, Saaristo TE, Saleheen D, Sattar N, Schadt EE, Schlessinger D, Slagboom PE, Snieder H, Spector TD, Thorsteinsdottir U, Stumvoll M, Tuomilehto J, Uitterlinden AG, Uusitupa M, van der Harst P, Walker M, Wallaschofski H, Wareham NJ, Watkins H, Weir DR, Wichmann HE, Wilson JF, Zanen P, Borecki IB, Deloukas P, Fox CS, Heid IM, O'Connell JR, Strachan DP, Stefansson K, van Duijn CM, Abecasis GR, Franke L, Frayling TM, McCarthy MI, Visscher PM, Scherag A, Willer CJ, Boehnke M, Mohlke KL, Lindgren CM, Beckmann JS, Barroso I, North KE, Ingelsson E, Hirschhorn JN, Loos RJF, Speliotes EK. Genetic studies of body mass index yield new insights for obesity biology. Nature. 2015;518(7538):197-206.
  7. Rask-Andersen M, Karlsson T, Ek WE, Johansson A. Genome-wide association study of body fat distribution identifies adiposity loci and sex-specific genetic effects. Nat Commun. 2019;10(1):339.
  8. Hendricks AE, Bochukova EG, Marenne G, Keogh JM, Atanassova N, Bounds R, Wheeler E, Mistry V, Henning E, Korner A, Muddyman D, McCarthy S, Hinney A, Hebebrand J, Scott RA, Langenberg C, Wareham NJ, Surendran P, Howson JM, Butterworth AS, Danesh J, Nordestgaard BG, Nielsen SF, Afzal S, Papadia S, Ashford S, Garg S, Millhauser GL, Palomino RI, Kwasniewska A, Tachmazidou I, O'Rahilly S, Zeggini E, Barroso I, Farooqi IS, Understanding Society Scientific G, Consortium E-C, Consortium UK. Rare Variant Analysis of Human and Rodent Obesity Genes in Individuals with Severe Childhood Obesity. Sci Rep. 2017;7(1):4394.
  9. Styne DM, Arslanian SA, Connor EL, Farooqi IS, Murad MH, Silverstein JH, Yanovski JA. Pediatric Obesity-Assessment, Treatment, and Prevention: An Endocrine Society Clinical Practice Guideline. J Clin Endocrinol Metab. 2017;102(3):709-757.
  10. Ahima RS, Prabakaran D, Mantzoros C, Qu D, Lowell B, Maratos-Flier E, Flier JS. Role of leptin in the neuroendocrine response to fasting. Nature. 1996;382(6588):250-252.
  11. Flier JS, Maratos-Flier E. Leptin's Physiologic Role: Does the Emperor of Energy Balance Have No Clothes? Cell metabolism. 2017;26(1):24-26.
  12. Considine RV, Sinha MK, Heiman ML, Kriauciunas A, Stephens TW, Nyce MR, Ohannesian JP, Marco CC, McKee LJ, Bauer TL, et al. Serum immunoreactive-leptin concentrations in normal-weight and obese humans. N Engl J Med. 1996;334(5):292-295.
  13. Gautron L, Elmquist JK, Williams KW. Neural control of energy balance: translating circuits to therapies. Cell.2015;161(1):133-145.
  14. Montague CT, Farooqi IS, Whitehead JP, Soos MA, Rau H, Wareham NJ, Sewter CP, Digby JE, Mohammed SN, Hurst JA, Cheetham CH, Earley AR, Barnett AH, Prins JB, O'Rahilly S. Congenital leptin deficiency is associated with severe early-onset obesity in humans. Nature. 1997;387(6636):903-908.
  15. Clement K, Vaisse C, Lahlou N, Cabrol S, Pelloux V, Cassuto D, Gourmelen M, Dina C, Chambaz J, Lacorte JM, Basdevant A, Bougneres P, Lebouc Y, Froguel P, Guy-Grand B. A mutation in the human leptin receptor gene causes obesity and pituitary dysfunction. Nature. 1998;392(6674):398-401.
  16. Farooqi IS, Matarese G, Lord GM, Keogh JM, Lawrence E, Agwu C, Sanna V, Jebb SA, Perna F, Fontana S, Lechler RI, DePaoli AM, O'Rahilly S. Beneficial effects of leptin on obesity, T cell hyporesponsiveness, and neuroendocrine/metabolic dysfunction of human congenital leptin deficiency. J Clin Invest. 2002;110(8):1093-1103.
  17. Farooqi IS, Wangensteen T, Collins S, Kimber W, Matarese G, Keogh JM, Lank E, Bottomley B, Lopez-Fernandez J, Ferraz-Amaro I, Dattani MT, Ercan O, Myhre AG, Retterstol L, Stanhope R, Edge JA, McKenzie S, Lessan N, Ghodsi M, De Rosa V, Perna F, Fontana S, Barroso I, Undlien DE, O'Rahilly S. Clinical and molecular genetic spectrum of congenital deficiency of the leptin receptor. N Engl J Med. 2007;356(3):237-247.
  18. Saeed S, Bonnefond A, Manzoor J, Shabbir F, Ayesha H, Philippe J, Durand E, Crouch H, Sand O, Ali M, Butt T, Rathore AW, Falchi M, Arslan M, Froguel P. Genetic variants in LEP, LEPR, and MC4R explain 30% of severe obesity in children from a consanguineous population. Obesity (Silver Spring). 2015;23(8):1687-1695.
  19. Wabitsch M, Funcke JB, Lennerz B, Kuhnle-Krahl U, Lahr G, Debatin KM, Vatter P, Gierschik P, Moepps B, Fischer-Posovszky P. Biologically inactive leptin and early-onset extreme obesity. N Engl J Med. 2015;372(1):48-54.
  20. Funcke JB, Moepps B, Roos J, von Schnurbein J, Verstraete K, Frohlich-Reiterer E, Kohlsdorf K, Nunziata A, Brandt S, Tsirigotaki A, Dansercoer A, Suppan E, Haris B, Debatin KM, Savvides SN, Farooqi IS, Hussain K, Gierschik P, Fischer-Posovszky P, Wabitsch M. Rare Antagonistic Leptin Variants and Severe, Early-Onset Obesity. N Engl J Med. 2023;388(24):2253-2261.
  21. Ozata M, Ozdemir IC, Licinio J. Human leptin deficiency caused by a missense mutation: multiple endocrine defects, decreased sympathetic tone, and immune system dysfunction indicate new targets for leptin action, greater central than peripheral resistance to the effects of leptin, and spontaneous correction of leptin-mediated defects. J Clin Endocrinol Metab. 1999;84(10):3686-3695.
  22. Farooqi IS, Jebb SA, Langmack G, Lawrence E, Cheetham CH, Prentice AM, Hughes IA, McCamish MA, O'Rahilly S. Effects of recombinant leptin therapy in a child with congenital leptin deficiency. N Engl J Med.1999;341(12):879-884.
  23. Galgani JE, Greenway FL, Caglayan S, Wong ML, Licinio J, Ravussin E. Leptin replacement prevents weight loss-induced metabolic adaptation in congenital leptin-deficient patients. J Clin Endocrinol Metab. 2010;95(2):851-855.
  24. Navarro VM. Metabolic regulation of kisspeptin — the link between energy balance and reproduction. Nature Reviews Endocrinology. 2020;16(8):407-420.
  25. Clement K, Biebermann H, Farooqi IS, Van der Ploeg L, Wolters B, Poitou C, Puder L, Fiedorek F, Gottesdiener K, Kleinau G, Heyder N, Scheerer P, Blume-Peytavi U, Jahnke I, Sharma S, Mokrosinski J, Wiegand S, Muller A, Weiss K, Mai K, Spranger J, Gruters A, Blankenstein O, Krude H, Kuhnen P. MC4R agonism promotes durable weight loss in patients with leptin receptor deficiency. Nat Med. 2018;24(5):551-555.
  26. Heymsfield SB, Greenberg AS, Fujioka K, Dixon RM, Kushner R, Hunt T, Lubina JA, Patane J, Self B, Hunt P, McCamish M. Recombinant leptin for weight loss in obese and lean adults: a randomized, controlled, dose-escalation trial. JAMA. 1999;282(16):1568-1575.
  27. Zelissen PM, Stenlof K, Lean ME, Fogteloo J, Keulen ET, Wilding J, Finer N, Rössner S, Lawrence E, Fletcher C, McCamish M. Effect of three treatment schedules of recombinant methionyl human leptin on body weight in obese adults: a randomized, placebo-controlled trial. Diabetes, obesity & metabolism. 2005;7(6):755-761.
  28. Farooqi IS, Keogh JM, Kamath S, Jones S, Gibson WT, Trussell R, Jebb SA, Lip GY, O'Rahilly S. Partial leptin deficiency and human adiposity. Nature. 2001;414(6859):34-35.
  29. Krude H, Biebermann H, Luck W, Horn R, Brabant G, Gruters A. Severe early-onset obesity, adrenal insufficiency and red hair pigmentation caused by POMC mutations in humans. Nat Genet. 1998;19(2):155-157.
  30. Krude H, Biebermann H, Schnabel D, Tansek MZ, Theunissen P, Mullis PE, Gruters A. Obesity due to proopiomelanocortin deficiency: three new cases and treatment trials with thyroid hormone and ACTH4-10. J Clin Endocrinol Metab. 2003;88(10):4633-4640.
  31. Lam BYH, Williamson A, Finer S, Day FR, Tadross JA, Gonçalves Soares A, Wade K, Sweeney P, Bedenbaugh MN, Porter DT, Melvin A, Ellacott KLJ, Lippert RN, Buller S, Rosmaninho-Salgado J, Dowsett GKC, Ridley KE, Xu Z, Cimino I, Rimmington D, Rainbow K, Duckett K, Holmqvist S, Khan A, Dai X, Bochukova EG, Trembath RC, Martin HC, Coll AP, Rowitch DH, Wareham NJ, van Heel DA, Timpson N, Simerly RB, Ong KK, Cone RD, Langenberg C, Perry JRB, Yeo GS, O'Rahilly S. MC3R links nutritional state to childhood growth and the timing of puberty. Nature. 2021;599(7885):436-441.
  32. Kuhnen P, Clement K, Wiegand S, Blankenstein O, Gottesdiener K, Martini LL, Mai K, Blume-Peytavi U, Gruters A, Krude H. Proopiomelanocortin Deficiency Treated with a Melanocortin-4 Receptor Agonist. N Engl J Med.2016;375(3):240-246.
  33. Lee YS, Challis BG, Thompson DA, Yeo GS, Keogh JM, Madonna ME, Wraight V, Sims M, Vatin V, Meyre D, Shield J, Burren C, Ibrahim Z, Cheetham T, Swift P, Blackwood A, Hung CC, Wareham NJ, Froguel P, Millhauser GL, O'Rahilly S, Farooqi IS. A POMC variant implicates beta-melanocyte-stimulating hormone in the control of human energy balance. Cell metabolism. 2006;3(2):135-140.
  34. Jackson RS, Creemers JW, Ohagi S, Raffin-Sanson ML, Sanders L, Montague CT, Hutton JC, O'Rahilly S. Obesity and impaired prohormone processing associated with mutations in the human prohormone convertase 1 gene. Nat Genet. 1997;16(3):303-306.
  35. Jackson RS, Creemers JW, Farooqi IS, Raffin-Sanson ML, Varro A, Dockray GJ, Holst JJ, Brubaker PL, Corvol P, Polonsky KS, Ostrega D, Becker KL, Bertagna X, Hutton JC, White A, Dattani MT, Hussain K, Middleton SJ, Nicole TM, Milla PJ, Lindley KJ, O'Rahilly S. Small-intestinal dysfunction accompanies the complex endocrinopathy of human proprotein convertase 1 deficiency. J Clin Invest. 2003;112(10):1550-1560.
  36. Wade KH, Lam BYH, Melvin A, Pan W, Corbin LJ, Hughes DA, Rainbow K, Chen JH, Duckett K, Liu X, Mokrosinski J, Morseburg A, Neaves S, Williamson A, Zhang C, Farooqi IS, Yeo GSH, Timpson NJ, O'Rahilly S. Loss-of-function mutations in the melanocortin 4 receptor in a UK birth cohort. Nat Med. 2021;27(6):1088-1096.
  37. Farooqi IS, Keogh JM, Yeo GS, Lank EJ, Cheetham T, O'Rahilly S. Clinical spectrum of obesity and mutations in the melanocortin 4 receptor gene. N Engl J Med. 2003;348(12):1085-1095.
  38. Stutzmann F, Tan K, Vatin V, Dina C, Jouret B, Tichet J, Balkau B, Potoczna N, Horber F, O'Rahilly S, Farooqi IS, Froguel P, Meyre D. Prevalence of melanocortin-4 receptor deficiency in Europeans and their age-dependent penetrance in multigenerational pedigrees. Diabetes. 2008;57(9):2511-2518.
  39. Collet TH, Dubern B, Mokrosinski J, Connors H, Keogh JM, Mendes de Oliveira E, Henning E, Poitou-Bernert C, Oppert JM, Tounian P, Marchelli F, Alili R, Le Beyec J, Pepin D, Lacorte JM, Gottesdiener A, Bounds R, Sharma S, Folster C, Henderson B, O'Rahilly S, Stoner E, Gottesdiener K, Panaro BL, Cone RD, Clement K, Farooqi IS, Van der Ploeg LHT. Evaluation of a melanocortin-4 receptor (MC4R) agonist (Setmelanotide) in MC4R deficiency. Mol Metab. 2017;6(10):1321-1329.
  40. Martinelli CE, Keogh JM, Greenfield JR, Henning E, van der Klaauw AA, Blackwood A, O'Rahilly S, Roelfsema F, Camacho-Hubner C, Pijl H, Farooqi IS. Obesity due to melanocortin 4 receptor (MC4R) deficiency is associated with increased linear growth and final height, fasting hyperinsulinemia, and incompletely suppressed growth hormone secretion. J Clin Endocrinol Metab. 2011;96(1):E181-188.
  41. Greenfield JR, Miller JW, Keogh JM, Henning E, Satterwhite JH, Cameron GS, Astruc B, Mayer JP, Brage S, See TC, Lomas DJ, O'Rahilly S, Farooqi IS. Modulation of blood pressure by central melanocortinergic pathways. N Engl J Med. 2009;360(1):44-52.
  42. Iepsen EW, Zhang J, Thomsen HS, Hansen EL, Hollensted M, Madsbad S, Hansen T, Holst JJ, Holm JC, Torekov SS. Patients with Obesity Caused by Melanocortin-4 Receptor Mutations Can Be Treated with a Glucagon-like Peptide-1 Receptor Agonist. Cell metabolism. 2018;28(1):23-32.e23.
  43. Hatoum IJ, Stylopoulos N, Vanhoose AM, Boyd KL, Yin DP, Ellacott KL, Ma LL, Blaszczyk K, Keogh JM, Cone RD, Farooqi IS, Kaplan LM. Melanocortin-4 receptor signaling is required for weight loss after gastric bypass surgery. J Clin Endocrinol Metab. 2012;97(6):E1023-1031.
  44. Weinstein LS, Chen M, Liu J. Gs(alpha) mutations and imprinting defects in human disease. Ann N Y Acad Sci.2002;968:173-197.
  45. Mendes de Oliveira E, Keogh JM, Talbot F, Henning E, Ahmed R, Perdikari A, Bounds R, Wasiluk N, Ayinampudi V, Barroso I, Mokrosinski J, Jyothish D, Lim S, Gupta S, Kershaw M, Matei C, Partha P, Randell T, McAulay A, Wilson LC, Cheetham T, Crowne EC, Clayton P, Farooqi IS. Obesity-Associated GNAS Mutations and the Melanocortin Pathway. N Engl J Med. 2021;385(17):1581-1592.
  46. Bochukova EG, Huang N, Keogh J, Henning E, Purmann C, Blaszczyk K, Saeed S, Hamilton-Shield J, Clayton-Smith J, O'Rahilly S, Hurles ME, Farooqi IS. Large, rare chromosomal deletions associated with severe early-onset obesity. Nature. 2010;463(7281):666-670.
  47. Hanssen R, Auwerx C, Joeloo M, Sadler MC, Estonian Biobank Research T, Henning E, Keogh J, Bounds R, Smith M, Firth HV, Kutalik Z, Farooqi IS, Reymond A, Lawler K. Chromosomal deletions on 16p11.2 encompassing SH2B1 are associated with accelerated metabolic disease. Cell Rep Med. 2023;4(8):101155.
  48. Doche ME, Bochukova EG, Su HW, Pearce LR, Keogh JM, Henning E, Cline JM, Saeed S, Dale A, Cheetham T, Barroso I, Argetsinger LS, O'Rahilly S, Rui L, Carter-Su C, Farooqi IS. Human SH2B1 mutations are associated with maladaptive behaviors and obesity. J Clin Invest. 2012;122(12):4732-4736.
  49. Cassidy SB, Schwartz S, Miller JL, Driscoll DJ. Prader-Willi syndrome. Genet Med. 2012;14(1):10-26.
  50. Sahoo T, del Gaudio D, German JR, Shinawi M, Peters SU, Person RE, Garnica A, Cheung SW, Beaudet AL. Prader-Willi phenotype caused by paternal deficiency for the HBII-85 C/D box small nucleolar RNA cluster. Nat Genet. 2008;40(6):719-721.
  51. de Smith AJ, Purmann C, Walters RG, Ellis RJ, Holder SE, Van Haelst MM, Brady AF, Fairbrother UL, Dattani M, Keogh JM, Henning E, Yeo GS, O'Rahilly S, Froguel P, Farooqi IS, Blakemore AI. A deletion of the HBII-85 class of small nucleolar RNAs (snoRNAs) is associated with hyperphagia, obesity and hypogonadism. Hum Mol Genet.2009;18(17):3257-3265.
  52. Swaab DF, Purba JS, Hofman MA. Alterations in the hypothalamic paraventricular nucleus and its oxytocin neurons (putative satiety cells) in Prader-Willi syndrome: a study of five cases. J Clin Endocrinol Metab.1995;80(2):573-579.
  53. Tauber M, Boulanouar K, Diene G, Cabal-Berthoumieu S, Ehlinger V, Fichaux-Bourin P, Molinas C, Faye S, Valette M, Pourrinet J, Cessans C, Viaux-Sauvelon S, Bascoul C, Guedeney A, Delhanty P, Geenen V, Martens H, Muscatelli F, Cohen D, Consoli A, Payoux P, Arnaud C, Salles JP. The Use of Oxytocin to Improve Feeding and Social Skills in Infants With Prader-Willi Syndrome. Pediatrics. 2017;139(2).
  54. Bochukova EG, Lawler K, Croizier S, Keogh JM, Patel N, Strohbehn G, Lo KK, Humphrey J, Hokken-Koelega A, Damen L, Donze S, Bouret SG, Plagnol V, Farooqi IS. A Transcriptomic Signature of the Hypothalamic Response to Fasting and BDNF Deficiency in Prader-Willi Syndrome. Cell Rep. 2018;22(13):3401-3408.
  55. Forsythe E, Beales PL. Bardet-Biedl syndrome. Eur J Hum Genet. 2013;21(1):8-13.
  56. Forsythe E, Sparks K, Best S, Borrows S, Hoskins B, Sabir A, Barrett T, Williams D, Mohammed S, Goldsmith D, Milford DV, Bockenhauer D, Foggensteiner L, Beales PL. Risk Factors for Severe Renal Disease in Bardet-Biedl Syndrome. J Am Soc Nephrol. 2017;28(3):963-970.
  57. Haqq AM, Chung WK, Dollfus H, Haws RM, Martos-Moreno GA, Poitou C, Yanovski JA, Mittleman RS, Yuan G, Forsythe E, Clement K, Argente J. Efficacy and safety of setmelanotide, a melanocortin-4 receptor agonist, in patients with Bardet-Biedl syndrome and Alstrom syndrome: a multicentre, randomised, double-blind, placebo-controlled, phase 3 trial with an open-label period. The lancet Diabetes & endocrinology. 2022;10(12):859-868.
  58. Gray J, Yeo GS, Cox JJ, Morton J, Adlam AL, Keogh JM, Yanovski JA, El Gharbawy A, Han JC, Tung YC, Hodges JR, Raymond FL, O'Rahilly S, Farooqi IS. Hyperphagia, severe obesity, impaired cognitive function, and hyperactivity associated with functional loss of one copy of the brain-derived neurotrophic factor (BDNF) gene. Diabetes. 2006;55(12):3366-3371.
  59. Han JC, Liu QR, Jones M, Levinn RL, Menzie CM, Jefferson-George KS, Adler-Wailes DC, Sanford EL, Lacbawan FL, Uhl GR, Rennert OM, Yanovski JA. Brain-derived neurotrophic factor and obesity in the WAGR syndrome. N Engl J Med. 2008;359(9):918-927.
  60. Yeo GS, Connie Hung CC, Rochford J, Keogh J, Gray J, Sivaramakrishnan S, O'Rahilly S, Farooqi IS. A de novo mutation affecting human TrkB associated with severe obesity and developmental delay. Nat Neurosci.2004;7(11):1187-1189.
  61. Sonoyama T, Stadler LKJ, Zhu M, Keogh JM, Henning E, Hisama F, Kirwan P, Jura M, Blaszczyk BK, DeWitt DC, Brouwers B, Hyvonen M, Barroso I, Merkle FT, Appleyard SM, Wayman GA, Farooqi IS. Human BDNF/TrkB variants impair hippocampal synaptogenesis and associate with neurobehavioural abnormalities. Sci Rep.2020;10(1):9028.
  62. Holder LJ, Nancy F. Butte & Andrew R. Zinn. Profound obesity associated with a balanced translocatioin that disrupts the SIM1 gene. Hum Mol Genet. 2000;9(1):101-108.
  63. Ramachandrappa S, Raimondo A, Cali AM, Keogh JM, Henning E, Saeed S, Thompson A, Garg S, Bochukova EG, Brage S, Trowse V, Wheeler E, Sullivan AE, Dattani M, Clayton PE, Datta V, Bruning JB, Wareham NJ, O'Rahilly S, Peet DJ, Barroso I, Whitelaw ML, Farooqi IS. Rare variants in single-minded 1 (SIM1) are associated with severe obesity. J Clin Invest. 2013;123(7):3042-3050.
  64. Moir L, Bochukova EG, Dumbell R, Banks G, Bains RS, Nolan PM, Scudamore C, Simon M, Watson KA, Keogh J, Henning E, Hendricks A, O'Rahilly S, Barroso I, Sullivan AE, Bersten DC, Whitelaw ML, Kirsch S, Bentley E, Farooqi IS, Cox RD. Disruption of the homeodomain transcription factor orthopedia homeobox (Otp) is associated with obesity and anxiety. Mol Metab. 2017;6(11):1419-1428.
  65. van der Klaauw AA, Croizier S, Mendes de Oliveira E, Stadler LKJ, Park S, Kong Y, Banton MC, Tandon P, Hendricks AE, Keogh JM, Riley SE, Papadia S, Henning E, Bounds R, Bochukova EG, Mistry V, O'Rahilly S, Simerly RB, Interval, Consortium UK, Minchin JEN, Barroso I, Jones EY, Bouret SG, Farooqi IS. Human Semaphorin 3 Variants Link Melanocortin Circuit Development and Energy Balance. Cell. 2019;176(4):729-742 e718.
  66. Cacciottolo TM, Henning E, Keogh JM, Bel Lassen P, Lawler K, Bounds R, Ahmed R, Perdikari A, Mendes de Oliveira E, Smith M, Godfrey EM, Johnson E, Hodson L, Clement K, van der Klaauw AA, Farooqi IS. Obesity Due to Steroid Receptor Coactivator-1 Deficiency Is Associated With Endocrine and Metabolic Abnormalities. J Clin Endocrinol Metab. 2022;107(6):e2532-e2544.
  67. Marenne G, Hendricks AE, Perdikari A, Bounds R, Payne F, Keogh JM, Lelliott CJ, Henning E, Pathan S, Ashford S, Bochukova EG, Mistry V, Daly A, Hayward C, Interval UKKC, Wareham NJ, O'Rahilly S, Langenberg C, Wheeler E, Zeggini E, Farooqi IS, Barroso I. Exome Sequencing Identifies Genes and Gene Sets Contributing to Severe Childhood Obesity, Linking PHIP Variants to Repressed POMC Transcription. Cell metabolism.2020;31(6):1107-1119 e1112.
  68. He Y, Brouwers B, Liu H, Liu H, Lawler K, Mendes de Oliveira E, Lee DK, Yang Y, Cox AR, Keogh JM, Henning E, Bounds R, Perdikari A, Ayinampudi V, Wang C, Yu M, Tu L, Zhang N, Yin N, Han J, Scarcelli NA, Yan Z, Conde KM, Potts C, Bean JC, Wang M, Hartig SM, Liao L, Xu J, Barroso I, Mokrosinski J, Xu Y, Sadaf Farooqi I. Human loss-of-function variants in the serotonin 2C receptor associated with obesity and maladaptive behavior. Nat Med.2022;28(12):2537-2546.
  69. Liu H, He Y, Liu H, Brouwers B, Yin N, Lawler K, Keogh JM, Henning E, Lee DK, Yu M, Tu L, Zhang N, Conde KM, Han J, Yan Z, Scarcelli NA, Liao L, Xu J, Tong Q, Zheng H, Sun Z, Yang Y, Wang C, He Y, Farooqi IS, Xu Y. Neural circuits expressing the serotonin 2C receptor regulate memory in mice and humans. Sci Adv.2024;10(26):eadl2675.
  70. Li Y, Cacciottolo TM, Yin N, He Y, Liu H, Liu H, Yang Y, Henning E, Keogh JM, Lawler K, Mendes de Oliveira E, Gardner EJ, Kentistou KA, Laouris P, Bounds R, Ong KK, Perry JRB, Barroso I, Tu L, Bean JC, Yu M, Conde KM, Wang M, Ginnard O, Fang X, Tong L, Han J, Darwich T, Williams KW, Yang Y, Wang C, Joss S, Firth HV, Xu Y, Farooqi IS. Loss of transient receptor potential channel 5 causes obesity and postpartum depression. Cell.2024;187(16):4176-4192 e4117.
  71. Asai M, Ramachandrappa S, Joachim M, Shen Y, Zhang R, Nuthalapati N, Ramanathan V, Strochlic DE, Ferket P, Linhart K, Ho C, Novoselova TV, Garg S, Ridderstrale M, Marcus C, Hirschhorn JN, Keogh JM, O'Rahilly S, Chan LF, Clark AJ, Farooqi IS, Majzoub JA. Loss of function of the melanocortin 2 receptor accessory protein 2 is associated with mammalian obesity. Science. 2013;341(6143):275-278.
  72. Pearce LR, Atanassova N, Banton MC, Bottomley B, van der Klaauw AA, Revelli JP, Hendricks A, Keogh JM, Henning E, Doree D, Jeter-Jones S, Garg S, Bochukova EG, Bounds R, Ashford S, Gayton E, Hindmarsh PC, Shield JP, Crowne E, Barford D, Wareham NJ, consortium UK, O'Rahilly S, Murphy MP, Powell DR, Barroso I, Farooqi IS. KSR2 mutations are associated with obesity, insulin resistance, and impaired cellular fuel oxidation. Cell. 2013;155(4):765-777.

 

Osteoporosis: Clinical Evaluation

ABSTRACT

 

The identification of a patient at high-risk of fracture should be followed by evaluation for factors contributing to low bone mass, skeletal fragility, falls, and fractures. Components of the evaluation include a bone density test, osteoporosis-directed medical history and physical exam, laboratory studies, and possibly skeletal imaging. A bone density test with dual-energy X-ray absorptiometry (DXA) is useful for diagnostic classification, assessment of fracture risk, and establishing a baseline for monitoring the skeletal effects of treatment. FRAX is a fracture risk algorithm that includes input of femoral neck bone mineral density measured by DXA. The DXA T-score, prior fracture history, and FRAX estimation of fracture risk are used with clinical practice guidelines to determine whether treatment is indicated. The medical history may reveal underlying causes of osteoporosis (e.g., nutritional deficiencies, gastric surgery, medications with adverse skeletal effects) and important risk factors for fracture (e.g., past history of fracture, family history of osteoporosis, or recent falls). Physical exam may show skeletal deformities due to unrecognized fractures (e.g., loss of height, kyphosis, or diminished rib-pelvis space), identify possible secondary causes of skeletal fragility (e.g., blue sclera with osteogenesis imperfecta, urticarial pigmentosa with systemic mastocytosis, dermatitis herpetiformis with celiac disease, or bone tenderness with osteomalacia), and help to recognize patients with poor balance and frailty that might lead to falls. Laboratory studies may show potentially reversible abnormalities (e.g., vitamin D deficiency, hypocalcemia, or impaired kidney function) that must be assessed and corrected, if possible, before starting pharmacological therapy. Disorders other than osteoporosis, requiring other types of treatment, may be found; for example, low serum alkaline phosphatase suggests hypophosphatasia, M-component may be due to myeloma, or hypocalciuria due to malabsorption with celiac disease. There are important safety considerations that can be derived from a pre-treatment assessment, as well. A patient with a blood clotting disorder should not be treated with raloxifene, a history of esophageal stricture is a contraindication for oral bisphosphonates, and previous skeletal radiation therapy precludes treatment with teriparatide or abaloparatide. Skeletal imaging may be helpful when a fracture, malignancy, or Paget’s disease of bone is suspected. Bone biopsy is rarely performed in clinical practice, but may be helpful in some situations, such as when it is necessary to determine the underlying bone disease in a patient with severe chronic kidney disease.

 

INTRODUCTION

 

Osteoporosis is a common systemic skeletal disease characterized by low bone strength that results in an increased risk of fracture (1). Fractures are associated with serious clinical consequences, including pain, disability, loss of independence, and death, as well as high healthcare costs. Early identification and intervention with patients at high risk for fracture is needed to reduce the burden of osteoporotic fractures (2). Management of a patient with a confirmed diagnosis of osteoporosis or low bone mass (osteopenia) includes assessment of fracture risk, evaluation for secondary causes of skeletal fragility, decisions on initiation of treatment, and identification of all relevant clinical factors that may influence patient management. This is a review of the key components in the care of patients prior to treatment.

 

DIAGNOSIS OF OSTEOPOROSIS

 

The World Health Organization (WHO) diagnostic classification (Table 1) (3) is made by bone mineral density (BMD) testing with dual-energy X-ray absorptiometry (DXA) using the T-score, calculated by subtracting the mean BMD (in g/cm2) of a young-adult reference population from the patient’s BMD and dividing by the standard deviation (SD) of the young-adult reference population. The International Society for Clinical Densitometry (ISCD) recommends that BMD be measured at the lumbar spine (ideally L1-L4), total hip, and femoral neck, with the 33% radius (1/3 radius) being measured when the lumbar spine and/or hip cannot be measured (e.g., obese patient who exceeds weight limit of table), is invalid (e.g., patient with lumbar laminectomy or bilateral total hip replacements), or when the patient has hyperparathyroidism (4). Osteoporosis cannot be diagnosed by BMD measurement at skeletal sites other than lumbar spine, total hip, femoral neck, and 33% radius or with technologies other than DXA, except for total hip and femoral neck T-scores calculated from 2D projections of quantitative computed tomography (QCT) data. The quality of DXA instrument maintenance, acquisition, analysis, interpretation, and reporting is important in obtaining valid results that can be used for making appropriate clinical decisions (4-6). In a patient with a fragility fracture, a clinical diagnosis of osteoporosis may be considered independently of BMD results, assuming other causes of skeletal fragility (e.g., osteomalacia, multiple myeloma) are not responsible for the fracture. Establishing a diagnosis of osteoporosis is clinically useful because it facilitates communication among healthcare providers and patients concerning a disease with potentially serious consequences; in some countries, such as the United States (US), a diagnosis is necessary in order to select a numerical code for submission of insurance claims for reimbursement for medical services. The US Bone Health & Osteoporosis Foundation (BHOF) (7) recommends that osteoporosis be diagnosed in postmenopausal women and men over the age ≥ 50 years in any of the following circumstances: T-score ≤ −2.5 at the lumbar spine, femoral neck, total hip, or 33% radius; low-trauma fracture of the hip, spine, and/or forearm; or T-score between -1.0 and -2.5 with FRAX 10-year probability of major osteoporotic fracture ≥ 20% or 10-year probability of hip fracture ≥ 3%.

 

Table 1. World Health Organization Criteria for Classification of Patients with Bone Mineral Density Measured by Dual-Energy X-ray Absorptiometry (3)

Classification

T-score

Normal

-1.0 or greater

Low bone mass (osteopenia)

Between 1-.0 and -2.5

Osteoporosis

-2.5 and below

Severe osteoporosis

-2.5 and below + fragility fracture

 

The BHOF indications for BMD testing in the US (7), which are similar to the ISCD Official Positions (4) are listed in Table 2. BMD testing should be done when it is likely to have an influence on patient management decisions. Other organizations and other countries with different economic resources and health care priorities have used a variety of methodologies to develop alternative recommendations (8-10).

 

Table 2. The BHOF Recommends that Bone Mineral Density Testing be Considered at DXA Facilities using Accepted Quality Assurance Procedures for the Following individuals (7).

Women age ≥ 65 years and men age ≥ 70 years

Postmenopausal women and men age 50-69 years, based on risk profile

Postmenopausal women and men age ≥ 50 years with history of adult-age fracture

Adults with a condition (e.g., rheumatoid arthritis, organ transplant) or taking a medication (e.g., glucocorticoids, aromatase inhibitors, androgen deprivation therapy) associated with low bone mass or bone loss

 

FRACTURE RISK ASSESSMENT

 

There is a robust correlation between BMD and fracture risk, with approximately a 2-fold increase in fracture risk for every 1 SD decrease in BMD (11). However, many or most patients with a hip fracture have a T-score better than -2.5 (12); although fracture risk is higher in patients with very low BMD, there are numerically many more patients with a T-score better than -2.5 than with a T-score ≤ -2.5, therefore there are numerically more fractures in those with higher T-scores. The presence of clinical risk factors (CRFs) that are independent of BMD, particularly advancing age, prior fracture, and recency/number/severity of fracture(s), can identify patients at high-risk for fracture by providing information on fracture risk that is complementary to BMD. The BHOF has provided an extensive list of CRFs (summarized in Table 3) for osteoporosis and fractures. Since most fractures occur with a fall, it is helpful to recognize risk factors for falling (summarized in Table 4) so that appropriate interventions can be made, when possible, to reduce the chances of falling.

 

Table 3. Conditions, Diseases, and Medications that Cause or Contribute to Osteoporosis and Fractures (adapted from guidelines of the BHOF (7)).

Lifestyle Factors

Low Calcium Intake

Vitamin D Insufficiency

Excess Vitamin A

Excessive Thinness

High Salt Intake

Immobilization

Inadequate Physical Activity

 

Smoking

Frequent Falling

 

 

Genetic Factors

Cystic Fibrosis

Homocystinuria

Osteogenesis Imperfecta

Ehlers-Danlos Syndrome

Hypophosphatasia

Gaucher’s Disease

Idiopathic Hypercalciuria

Porphyria

Glycogen storage diseases

Marfan Syndrome

Riley-Day Syndrome

Hemochromatosis

Menkes Steely Hair Syndrome

Parental History of Hip Fracture

Androgen Insensitivity

Turner’s & Klinefelter’s Syndromes

Endocrine Disorders

Obesity

Diabetes Mellitus

Hyperthyroidism

Cushing’s Syndrome

Hyperparathyroidism

Hypogonadism

Panhypopituitarism

Female athlete triad

Anorexia Nervosa

Hyperprolactinemia

Premature Menopause

Androgen Insensitivity

Gastrointestinal Disorders

Celiac Disease

Inflammatory Bowel Disease

Primary Biliary Cirrhosis

Gastric Bypass

Malabsorption

GI Surgery

Pancreatic Disease

 

Hematologic Disorders

Hemophilia

Monoclonal Gammopathies

Systemic Mastocytosis

Leukemia

Lymphoma

Sickle Cell Disease

Thalassemia

Multiple Myeloma

Rheumatic and Autoimmune Diseases

Ankylosing Spondylitis

Systemic Lupus

Rheumatoid Arthritis

Multiple Sclerosis

Muscular Dystrophy

Parkinson’s Disease

Spinal Cord Injury

Stroke

Epilepsy

 

 

 

Miscellaneous Conditions and Diseases

Chronic Obstructive Pulmonary Disease

Weight Loss

Amyloidosis

End Stage Renal Disease

Parenteral Nutrition

Chronic Metabolic Acidosis

Hyponatremia

Post-Transplant Bone Disease

Congestive Heart Failure

Idiopathic Scoliosis

Prior Fracture as an Adult

Depression

HIV/AIDS

Sarcoidosis

 

 

Medications

Anticoagulants (heparin)

Cancer Chemotherapy

Gonadotropin Releasing Hormone Agonists

Anticonvulsants

Lithium

Aromatase Inhibitors

Depo-medroxyprogesterone

Barbiturates

Glucocorticoids (> 5mg of prednisone or equivalent for > 3 months)

Cyclosporine A

Tacrolimus

Aluminum-containing Antacids

Proton Pump Inhibitors

Tamoxifen (premenopausal)

Selective Serotonin Reuptake Inhibitors

Thiazolidinediones

 

Table 4. Risk Factors for Falls Adapted from Guidelines of the BHOF (7).

Environmental Risk Factors: lack of assistive devices in bathrooms, loose throw rugs, low level lighting, obstacles in the walking path, stairs, slippery outdoor conditions

Medical Risk Factors: advanced age, anxiety and agitation, arrhythmias, dehydration, depression, female gender, impaired transfer and mobility, malnutrition, orthostatic hypotension, poor vison and use of bifocals, previous fall, reduced mental acuity and diminished cognitive skills, urgent urinary incontinence, Vitamin D insufficiency (serum 25-OH-D < 30 ng/mL [75 nmol/L]), medications causing over-sedation (narcotic analgesics, anticonvulsants, psychotropics), diabetes

Neurological and Musculoskeletal Risk Factors: kyphosis, poor balance, reduced proprioception, weak muscles

Psychological Risk Factors: fear of falling

The presence of any of these risk factors should trigger consideration of further evaluation and treatment to reduce the risk of falls and fall-related injuries.

 

VERTEBRAL FRACTURE ASSESSMENT (VFA)

 

VFA is a method for imaging the thoracic and lumbar spine by DXA for the purpose of detecting vertebral fracture deformities. Identification of a previously unrecognized vertebral fracture may alter diagnostic classification, change estimation of fracture risk, and influence treatment decisions (13). VFA compares favorably with standard radiographs of the spine, with good correlation for detecting moderate (grade 2) and severe (grade 3) vertebral fractures, a smaller dose of ionizing irradiation, greater patient convenience (i.e., it may be done at the same visit and with the same instrument as BMD testing by DXA), and lower cost. In a study of women age 65 years and older, using the Genant semi-quantitative (SC) method of classifying vertebral deformities (14), the sensitivity of VFA for diagnosing moderate and severe vertebral fractures was 87-93%, with a specificity of 93-95% (15). Indications for vertebral imaging are listed in Table 5. Optimal use of DXA and VFA requires training and adherence to well established quality standards (4).

 

Table 5. ISCD Indications for Lateral Spine Imaging by Standard Radiography or Vertebral Fracture Assessment (VFA) (4)

Vertebral imaging is indicated when the T-score is < -1.0 and one or more of the following is present:

Women ≥ 70 years of age or men ≥ 80 years of age

Historical height loss > 4 cm (1.5 inches)

Self-reported but undocumented prior vertebral fracture

Glucocorticoid therapy equivalent to ≥ 5 mg of prednisone or equivalent per day for ≥ 3 months

 

QUALITY OF DXA AND VFA

 

DXA and VFA should be performed by well-trained and experienced staff operating an instrument that has been maintained and calibrated according to the manufacturer’s standards. Precision assessment and least significant change (LSC) calculation by each DXA technologist are required in order to make quantitative comparisons of serial BMD measurements. The LSC is the smallest change in BMD that is statistically significant, usually with a 95% level of confidence. The use of the correct scan modes, proper patient positioning, consistent vertebral body labeling, and bone edge detection are among the essential elements for serial comparisons of BMD. VFA should be done by a technologist properly trained in acquisition techniques and interpreted by a clinician familiar with methods of diagnosing vertebral fractures using this technology. Bone densitometry facilities should be supervised by a clinician who knows current methods for BMD measurement and fully understands the standards for quality control, interpretation, and reporting of the findings. Poor quality studies may result in inappropriate clinical decisions, generate unnecessary healthcare expenses, and be harmful to patients (5). Assurances of high quality DXA can be attained through education, training, and certification of DXA technologists and interpreters by organizations such as the ISCD. DXA facilities should understand and adhere to ISCD Official Positions (4) and DXA Best Practices (6, 14); facility accreditation (15) provides assurance of adherence to DXA quality standards.

 

TECHNOLOGIES FOR ASSESSMENT OF SKELETAL HEALTH

 

Dual-energy X-ray Absorptiometry (DXA)

 

Devices that measure or estimate BMD differ according to their clinical utility, cost, portability, and use of ionizing radiation. DXA is the “gold standard” method for measuring bone density in clinical practice. There is a strong correlation between mechanical strength and BMD measured by DXA biomechanical studies (16). In observational studies of untreated patients, there is a robust relationship between fracture risk and BMD measured by DXA (11). The WHO diagnostic classification of osteoporosis is based primarily on reference data obtained by DXA (3), and femoral neck BMD provides input into the FRAX algorithm. Most randomized clinical trials showing reduction in fracture risk with pharmacological therapy have selected study participants according to BMD measured by DXA (17). There is a relationship between BMD increases with drug therapy and fracture risk reduction (18, 19). Accuracy and precision of DXA are excellent (20). Radiation exposure with DXA is very low (21). BMD of the 33% (one-third) radius, measured either by a dedicated peripheral DXA (pDXA) device or a central DXA instrument with appropriate software, may be used for diagnostic classification with the WHO criteria and to assess fracture risk, but is generally not clinically useful in monitoring the effects of treatment. DXA measures bone mineral content (BMC in grams [g]) and bone area (cm2), then calculates areal BMD in g/cm2 and derives parameters, such as the T-score and Z-score. DXA is used for diagnostic classification, assessment of fracture risk, and for monitoring changes in BMD over time.

 

Quantitative Ultrasound (QUS)

 

QUS devices emit inaudible high frequency sound waves in the ultrasonic range, typically between 0.1 and 1.0 megahertz (MHz). The sound waves are produced and detected by means of high-efficiency piezoelectric transducers, which must have good acoustical contact with the skin over the bone being tested. Technical differences among QUS systems are great, with different instruments using variable frequencies, different transducer sizes, and sometimes measuring different regions of interest, even at the same skeletal site. The calcaneus is the skeletal site most often tested, although other bones, including the radius, tibia, and finger phalanges, can be used. Commercial QUS systems usually measure two parameters- the speed of sound (SOS) and broadband ultrasound attenuation (BUA). A proprietary value, such as the “quantitative ultrasound index” (QUI) with the Hologic Sahara or “stiffness index” with the GE Healthcare Achilles Express, may be calculated from a combination of these measurements. SOS varies according to the type of bone, with a typical range of 3000-3600 meters per second (m/sec) with cortical bone and 1650-2300 m/sec for trabecular bone (22). A higher bone density is associated with a higher SOS. BUA, reported as decibels per megahertz (dB/MHz), is a measurement of the loss of energy, or attenuation, of the sound wave as it passes through bone. As with SOS, a higher bone density is associated with a higher BUA. Values obtained from calculations using ultrasound parameters may be used to generate an estimated BMD and a T-score. The T-score derived from a QUS measurement is not the same as a T-score from a DXA. QUS cannot be used for diagnostic classification and is not clinically useful to monitor the effects of therapy (23).

 

Radiofrequency Echographic Multi Spectrometry (REMS) assesses bone health and fracture risk with an ultrasound scan of the lumbar spine and proximal femur, thereby overcoming the limitation of QUS of only measuring peripheral skeletal sites. REMS technology uses a portable device with a transducer that transmits ultrasound waves to the target axial skeletal site and a receiver that captures the resultant back-scattered waveforms with B-mode image reconstruction of the region of interest. There are studies that support a strong correlation between REMS and DXA measurements of BMD (24). Potential clinical applications include its use in frail patients with limited mobility, bedside measurements in hospitalized patients, and special populations such as pregnant women and children.

 

Quantitative Computed Tomography (QCT) and Peripheral QCT (pQCT)

 

QCT and pQCT measure trabecular and cortical volumetric BMD at the axial skeleton and peripheral skeletal sites, respectively. QCT is a useful research tool to enhance understanding of the pathophysiology of osteoporosis and the mechanism of action of pharmacological agents used to treat osteoporosis. QCT predicts fracture risk, with the correlation varying according to skeletal site and bone compartment measured, type of fracture predicted, and population assessed (4). The ISCD Official Positions state that “spinal trabecular BMD as measured by QCT has at least the same ability to predict vertebral fractures as AP spinal BMD measured by central DXA in postmenopausal women with lack of sufficient evidence to support this position in men; pQCT of the forearm at the ultra-distal radius predicts hip, but not spine, fragility fractures in postmenopausal women with lack of sufficient evidence to support this position in men (4).” QCT is more expensive than DXA and QUS and uses higher levels of ionizing radiation than DXA. T-scores by QCT are typically lower than with DXA (27), thereby overestimating the prevalence of osteoporosis, with the exception of total hip and femoral neck T-scores calculated from 2D projections of QCT data, which are similar to DXA-derived T-scores at the same regions of interest and may be used for diagnosis of osteoporosis in accordance with the WHO criteria. T-scores and femoral neck BMD derived from 2D projections of QCT data may also be used as input for the FRAX algorithm to estimate 10-year fracture probabilities.

 

Other Technologies of Interest

 

Pulse-echo ultrasonography (PEUS) uses a portable handheld ultrasound device to estimate the thickness of cortical bone at peripheral skeletal sites. When connected to a computer with proprietary software, a value can be generated that that is correlated with hip BMD measured by DXA, with the potential benefit of identifying patients who are likely or unlikely to have osteoporosis (25).  

 

Biomechanical CT (BCT) is an opportunistic analysis of data from pre-existing CT scans of the hip and/or spine that provides DXA-equivalent T-scores for the hip, QCT-equivalent vBMD at the spine, and an estimate of bone strength with finite element analysis (FEA) (26).  

 

3D-Shaper is software that can be added to a DXA system using statistical modelling to reconstruct the 3D shape and density distribution of the proximal femur from 2D DXA data scans. A recent study found that this technology provided an estimation of femur strength that was similar to that derived from QCT FEA (27).

 

FRACTURE RISK ASSESSMENT TOOL (FRAX® and FRAXplus)

 

The combination of BMD and clinical risk factors (CRFs) predicts fracture risk better than BMD or CRFs alone (28,29) (2). A fracture risk assessment tool (FRAX) combines CRFs and femoral neck BMD in a computer-based algorithm that estimates the 10-year probability of hip fracture and major osteoporotic fracture (i.e., clinical spine, hip, proximal humerus, and distal forearm fracture). FRAX can be accessed online at http://www.shef.ac.uk/FRAX (Figure 1), on most software versions of DXA systems, and on smartphones. FRAX is based on analysis of data from 12 large prospective observational studies in about 60,000 untreated men and women in different world regions, having over 250,000 person-years of observation and more than 5,000 reported fractures reported.

Figure 1. FRAX online for US Caucasian patients. This example shows a 65-year-old woman who has no clinical risk factors for fracture and a femoral neck BMD of 0.582 g/cm2 with a Hologic instrument. The 10-year probability of major osteoporotic fracture is 11% and the 10-year probability of hip fracture is 2.2%. These levels do not meet the Bone Health & Osteoporosis Foundation guidelines for initiation of pharmacological therapy in the US (7). Image reproduced with permission from Eugene McCloskey, University of Sheffield, Sheffield, UK.

 

The input for FRAX is the patient’s age, sex, height, weight, a “yes” or “no” response indicating the presence or absence for each of 7 CRFs: 1. previous ‘spontaneous’ or fragility fracture as an adult; 2. parent with hip fracture; 3. current tobacco smoking; 4. ever use of chronic glucocorticoids at least 5 mg prednisolone for at least 3 months; 5. confirmed rheumatoid arthritis; 6. secondary osteoporosis, such as type 1 diabetes, osteogenesis imperfecta in adults, untreated longstanding hyperthyroidism and hypogonadism, or premature menopause (note: this is a “dummy” risk factor that has no effect on the fracture risk calculation unless no femoral neck BMD value is entered); 7. alcohol intake greater than 3 units per day, with a unit of alcohol defined as equivalent to a glass of beer, an ounce of spirits or a medium-sized glass of wine), and if available, femoral neck BMD and trabecular bone score (TBS). Since the introduction of FRAX, upgrades have been introduced to correct errors, enhance its usability, and incorporate new data that have become available.

 

Benefits of FRAX

 

The use of FRAX provides a quantitative estimation of fracture risk that is based on robust data in large populations of men and women with ethnic and geographic diversity. Expression of fracture risk as a probability provides greater clinical utility than relative risk. When combined with cost-utility analysis, a fracture risk level at which it is cost-effective to treat may be derived. FRAX can be used to estimate fracture probability without femoral neck BMD, allowing it to be used when DXA in unavailable or inaccessible. FRAX is incorporated into many clinical practice guidelines.

 

Limitations of FRAX

 

To generate a valid FRAX output, the responses to CRF questions must be correct; for example, an incorrect entry of self-reported rheumatoid arthritis or use of glucocorticoids could skew the results toward overestimation of fracture risk. FRAX may underestimate or overestimate fracture risk due to dichotomized (yes or no) input for CRFs that in reality are associated with a range of risk that varies according to dose, duration of exposure, or severity; for example, fracture risk may be underestimated when a patient is on high-dose glucocorticoid therapy or has had multiple recent fragility fractures, even when a “yes” response is entered for these CRFs. FRAX is validated only in untreated patients and may overestimate fracture risk when the patient is being treated; the NOF(BHOF)/ISCD guidance on FRAX suggests that “untreated” may be interpreted as never treated or if previously treated, no bisphosphonate for the past 2 years (unless it is an oral agent taken for less than 2 months); and no estrogen, raloxifene, calcitonin, or denosumab for the past 1 year (7). In this context, calcium and vitamin D do not constitute treatment. FRAX in the US allows input for 4 ethnicities (Caucasian, Black, Hispanic, Asian); it is not clear how to use FRAX for patients of other ethnicities or a mix of these ethnicities. Answering “yes” for the category of secondary osteoporosis has no effect on the fracture risk calculation as long as a value for femoral neck BMD is entered. The range of error for a fracture probability generated by FRAX is unknown but may be substantial in some cases.

 

Some important risk factors, such as falls and frailty, are not directly entered into FRAX, although they are indirectly included insofar as they are a component of aging. FRAX may underestimate fracture risk when the lumbar spine BMD is substantially lower than femoral neck BMD, as may occur in about 15% of patients (30). Despite the limitations of FRAX, it is a helpful clinical tool when used with a good understanding of factors that may result in underestimation or overestimation of fracture risk. FRAX may enhance discussion of risk with the patient and help to identify those who are at sufficiently high for fracture to benefit from therapy.

 

FRAXplus

 

FRAXplus (https://www.fraxplus.org/) is an updated version of FRAX that addresses some of the limitations of traditional FRAX, allowing input for these additional rick factors: recency of osteoporotic fracture, high exposure to oral glucocorticoids, type 2 diabetes, concurrent data on lumbar spine BMD, trabecular bone score, falls history, and hip axis length. For patients with fracture risk that is close to the intervention threshold for the applicable clinical practice guideline, the use of an additional risk factor with FRAXplus might influence the decision to treat or not treat with a pharmacological agent.

 

MEDICAL HISTORY

 

A thorough medical history may identify risk factors for osteoporosis and fractures, suggesting that a bone density test and/or further evaluation is indicated. The medical history may also reveal symptoms of potentially correctable causes of skeletal fragility (e.g., gluten intolerance with celiac disease) or co-morbidities that could influence treatment decisions (e.g., esophageal stricture suggests that oral bisphosphonates should not be given). A history of falls is a predictor of future falls, with that risk potentially modifiable though appropriate interventions. Finally, some symptoms may trigger further evaluation for the presence of fractures (e.g., historical height loss or development of kyphotic posture suggests the possibility of vertebral fractures that may warrant spine imaging). Table 6 provides examples of helpful information that might be obtained from a thoughtful interactive discussion with the patient.

 

Medical History for Patients with Osteoporosis

 

A thorough review of systems and history of relevant familial disorders, previous surgical procedures, medications, dietary supplements, food intolerances, and lifestyle provides helpful information in the management of patients with osteoporosis. Such historical information may play a role in determining who should have a bone density test, assessing fracture risk, providing input for FRAX, evaluating for secondary causes of osteoporosis, selecting the most appropriate treatment to reduce fracture risk, and finding factors contributing to suboptimal response to therapy. Listed here are key components of the skeletal health history and examples of the potential impact on patient care.

 

Table 6. Clinical Utility of the Medical History

Clinical Utility

Medical History

Assist in determining who need a bone density test

See Table 3

Assessing fracture risk

See Table 3 and 4

Input for FRAX

Age, sex, weight, height, previous fracture, parent with hip fracture, current tobacco smoking, ever use of glucocorticoids, rheumatoid arthritis, secondary osteoporosis, alcohol intake 3 or more units per day, and if available, femoral neck bone mineral density and trabecular bone score

Evaluating for secondary causes of osteoporosis

See Table 3

Selecting most appropriate treatment

Identify co-morbidities of clinical significance. For example, high-risk of breast cancer favors raloxifene use, while history of thrombophlebitis suggests that raloxifene should not be used; esophageal stricture is a contraindication for oral bisphosphonate use; a patient with a skeletal malignancy should not be treated with teriparatide.

Factors contributing to suboptimal response to therapy

Compliance and persistence to therapy; adequacy of calcium and vitamin D; comorbidities listed in Table 3.

 

PHYSICAL EXAM

 

Findings of importance on the physical exam of a patient with osteoporosis may be the sequelae of old fractures (e.g., kyphosis due to old vertebral fractures), a consequence of a recent fracture (e.g., localized vertebral spinous process tenderness with a new vertebral fracture), or abnormalities suggestive of a secondary cause of osteoporosis (e.g., thyromegaly with thyrotoxicosis). An accurate measurement of height with a wall-mounted stadiometer is a helpful office tool for evaluating patients at risk for fracture. A height loss of 1.5 inches (4.0 cm) or more compared to the historical maximum (28, 29) or a loss of 0.75 inches (2.0 cm) or more compared to a previous measured height (30) suggests a high likelihood of vertebral fracture. Body weight measurement is part of the osteoporosis evaluation because low body weight (less than 127 lbs) (31), low BMI (20 kg/m2 or less) (32), and weight loss of 5% or more ((33)36) are associated with increased risk of fracture. Localized tenderness of the spine, kyphosis, or diminished distance between the lower ribs and the pelvic brim may be the result of one or more vertebral fractures. Abnormalities of gait, posture, balance, muscle strength, or the presence of postural hypotension or impaired level of consciousness may be associated with increased risk of falling. Bone tenderness may be caused by osteomalacia. Atrophic testicles suggest hypogonadism. Patients should be observed for stigmata of hyperthyroidism or Cushing’s syndrome. Blue sclera, hearing loss, and yellow-brown teeth are suggestive of osteogenesis imperfecta. Joint hypermobility and skin fragility could be due to Ehlers-Danlos syndrome. Urticaria pigmentosa may occur with systemic mastocytosis. Table 7 shows examples of abnormal physical exam findings with osteoporosis.

 

Table 7. Focused Physical Examination in a Patient with Osteoporosis

Component of physical exam

Example of finding of potential skeletal importance

Potential clinical implications for skeletal health

Vital signs

Low body weight or body mass index

Anorexia nervosa

Loss of height

Vertebral fracture

Loss of weight

Malignancy, malabsorption

Skin

Urticaria pigmentosa

Dermatitis herpetiformis

Systemic mastocytosis

Celiac disease

Striae, acne

Cushing’s syndrome, exogenous glucocorticoids

Head

Cranial dysostosis

Hypophosphatasia

Eyes

Blue sclera

Osteogenesis imperfect

Ears

Hearing loss

Osteogenesis imperfecta, sclerosteosis

Nose

Anosmia

Kallmann syndrome

Throat

Poor dentition

Increased risk of osteonecrosis of the jaw

Neck

Thyromegaly

Thyrotoxicosis

Lungs

Decreased breath sounds

Chronic obstructive pulmonary disease

Heart

Aortic insufficiency

Marfan’s syndrome

Musculoskeletal

Kyphosis

Vertebral fractures

Spinous process tenderness

Acute vertebral fracture

Decreased space between lower ribs and pelvis

Vertebral fractures

Tender bones

Osteomalacia

Inflammatory joint disease

Rheumatoid arthritis

Hypermobility of joints

Ehlers-Danlos syndrome

Muscle weakness

Vitamin D deficiency, osteomalacia

Abdomen

Hepatomegaly

Chronic liver disease

Surgical scars

Bariatric surgery, gastrectomy

Genitalia

Testicular atrophy

Hypogonadism

Neurological

Poor balance

High fall risk, vitamin D deficiency

Dementia

Poor adherence to therapy, high fall risk

This table provides examples of findings on physical exam that may be helpful in the evaluation of skeletal health. It is not intended to show all findings of importance.

 

EVALUATION FOR SECONDARY CAUSES OF OSTEOPOROSIS

 

The possibility of previously unrecognized causes of skeletal fragility should be considered in every patient with osteoporosis (34), understanding that some patients with a T-score ≤ -2.5 may have a skeletal disease other than osteoporosis and some patients with osteoporosis have contributing disorders and conditions other than estrogen deficiency and advancing age that can be corrected. Collectively, these contributing factors are sometimes called secondary causes of osteoporosis. After an initial medical history is taken and physical exam is performed, appropriate laboratory testing and imaging may provide information that is critical for ongoing patient care.

 

The reported prevalence of secondary osteoporosis varies depending on the study population, the extent of the medical evaluation, and definitions for laboratory abnormalities. It is likely that many or most patients with osteoporosis have clinically significant contributing factors that may influence patient management. In a study of North American women receiving osteoporosis therapy, it was found that 52% had vitamin D inadequacy, defined as serum 25-hydroxyvitamin D (25-OH-D) levels less than 30 ng/ml (35). In another study of patients referred to an osteoporosis clinic, over 60% were found to have elements of secondary osteoporosis when vitamin D deficiency was very conservatively defined as serum 25-OH-D level less than 12.5 ng/ml (36, 37). In the same study, the number of patients with secondary osteoporosis was much higher when vitamin D inadequacy was more appropriately defined as serum 25-OH-D less than 33 ng/ml (38, 39).

 

It has been proposed by some that a bone density that is less than expected compared to an age- and sex-matched population, as represented by a low Z-score (e.g., less than -2.0), suggests a high likelihood of secondary osteoporosis and should be one of the triggers for further investigation (40, 41). While there may be some merit to this concept, there are few if any studies validating the use of a Z-score cutoff for this purpose. Since secondary causes of osteoporosis are common, a more effective strategy is to screen all patients with osteoporosis for contributing factors (42). The results of a metabolic evaluation may identify previously unrecognized diseases and conditions that require treatment in addition to, or instead of, standard osteoporosis pharmacological therapy.

 

Depending on the patient population being studied, different causes of secondary osteoporosis may predominate. Calcium deficiency, vitamin D deficiency, and sedentary lifestyle are common contributing factors for all patients. In women referred to an osteoporosis clinic with previously recognized medications or diseases contributing to osteoporosis, the most common were history of glucocorticoid use (36%), premature ovarian failure (21%), history of unintentional weight loss (10%), history of alcoholism (10%), and history of liver disease (10%) (36). When patients without previously recognized contributing factors were evaluated at the same specialty clinic, most (55%) were found to have vitamin D deficiency or insufficiency (serum 25-OH-D less than 33 ng/ml) (39), while 10% had hypercalciuria, 8% had malabsorption, and 7% had primary or secondary hyperparathyroidism (36). In men, the most common secondary causes of osteoporosis are long-term glucocorticoid use, hypogonadism, and alcoholism (43, 44). The increasing use of aromatase inhibitor therapy for breast cancer in women and androgen deprivation therapy for prostate cancer in men (45) is now recognized as an important factor in the development of osteoporosis in these patients. Other common causes for low BMD and fractures include multiple myeloma (46), gastric bypass surgery (47) and gastric resection (48). Treatable but easily missed secondary causes of osteoporosis include asymptomatic primary hyperparathyroidism (49), subclinical hyperthyroidism (50), mild Cushing’s syndrome (51), and malabsorption due to unrecognized celiac disease (52). Table 8 lists some of the causes of low BMD by category.

 

Table 8. Causes of Low Bone Mineral Density

Inherited

Nutritional

Endocrine

Drugs

Other

Osteogenesis imperfecta

Malabsorption

Hypogonadism

Glucocorticoids

Multiple myeloma

Homocystinuria

Chronic liver disease

Hyperthyroidism

Anticonvulsants

Rheumatoid arthritis

Marfan’s syndrome

Alcoholism

Hyperparathyroidism

Long-term heparin

Systemic mastocytosis

Hypophosphatasia

Calcium deficient diet

Cushing’s syndrome

Excess thyroid

Immobilization

 

Vitamin D deficiency

Eating disorder

GnRH agonists

 
     

Aromatase inhibitors

 

 

Although a variety of testing strategies have been proposed as screening for all patients with osteoporosis, a minimal cost-effective work-up for all patients consists of a complete blood count (CBC), serum calcium, phosphorus, creatinine with calculated or measured creatinine clearance, alkaline phosphatase, 24-hour urinary calcium, and serum 25-OH-D. Other laboratory tests may be indicated according to the patient’s clinical profile and the practice setting. A summary of useful common and uncommon laboratory studies with comments on their possible skeletal significance is provided below.

 

CLINICAL CASE

 

A 52-year-old postmenopausal woman with a history of irritable bowel syndrome (IBS) and a family history of osteoporosis (mother with hip fracture) is found to have osteoporosis on a DXA study. Evaluation for secondary causes of osteoporosis is unremarkable except for mild iron deficiency anemia (a long-standing problem, previously attributed to heavy menses) and a low 24-hour urinary calcium of 30 mg, with adequate calcium intake and normal renal function. Serum 25-OH-D is 29 ng/ml. Additional work-up shows a high titer of IgA endomysial antibodies consistent with celiac disease. This diagnosis is confirmed by a small bowel biopsy showing villous atrophy. She is started on a gluten-free diet, resulting in resolution of her “IBS” symptoms and correction of her anemia. One year later, with no pharmacological therapy for osteoporosis, there is a statistically significant BMD increase of 9% at the lumbar spine.

 

Celiac disease may result in osteoporosis due to calcium malabsorption, even in the absence of gastrointestinal symptoms. Treatment is strict lifelong adherence to a gluten-free diet, which may sometimes be followed by a substantial increase in BMD, as seen in this patient. A 24-hour urinary calcium is an inexpensive screening test for calcium malabsorption that should be considered a routine part of the initial evaluation of osteoporosis.

 

BASIC BLOOD TESTS

 

CBC- Anemia may be seen in patients with myeloma or malnutrition

 

Sedimentation rate- May be elevated with myeloma and rheumatic diseases.

 

Calcium- Among the many causes of hypercalcemia are primary and secondary hyperparathyroidism, hyperthyroidism, renal failure, vitamin D intoxication, and Paget’s disease of bone. Hypocalcemia may be seen with vitamin D deficiency and hyperphosphatemia.

 

Phosphorus- Hyperphosphatemia may occur with hypoparathyroidism, renal failure, and possibly with bisphosphonate therapy. Hypophosphatemia may be seen with primary or secondary hyperparathyroidism, vitamin D deficiency, tumor induced osteomalacia, and X-linked hypophosphatemia.

 

Alkaline phosphatase- High values can be seen with healing fractures, osteomalacia, and Paget’s disease, as well as occurring normally in growing children. Low values occur with hypophosphatasia, a rare genetic disorder that causes impaired mineralization of bone and dental tissue.

 

Vitamin D- The test that best reflects vitamin D stores is the serum 25-OH-D. While there is no consensus on the optimal range of serum 25-OH-D, a reasonable target for good skeletal health is approximately 30-50 ng/ml. This is likely to maximize intestinal absorption of calcium and minimize serum PTH levels. Interpretation of serum 25-OH-D levels is confounded by assay variability (59). Serum 1,25-(OH)2-D3 is usually not helpful in the evaluation of osteoporosis patients, unless there are concerns regarding renal conversion of 25-OH-D to 1,25-(OH)2-D3. Deficiency or insufficiency of vitamin D is very common and play a role in the pathogenesis of osteoporosis and osteomalacia.

 

Creatinine- Chronic kidney disease may cause an elevated creatinine level and renal osteodystrophy. Elderly patients with small muscle mass may have impaired renal function with a “normal” serum creatinine. An estimated glomerular filtration rate can be calculated using one of many formulae, such as that of Cockcroft and Gault (53) or modification of diet in renal disease study equation (54). Impaired renal function not only has adverse skeletal effects but also raises considerations regarding the type and dose of pharmacologic agents used.

 

TSH- Hyperthyroidism from any cause, including excess thyroid replacement, can usually be recognized by a low TSH. High bone turnover associated hyperthyroidism is associated with loss of bone mass.

 

Liver enzymes- Abnormalities may be caused by chronic liver disease, which is a risk factor for osteoporosis.

 

BASIC URINE TESTS

 

Urinalysis- Proteinuria may occur with multiple myeloma or chronic kidney disease. Abnormal cells may suggest kidney disease.

 

24-hour urine for calcium- A well-collected 24-hour urine for calcium is a helpful screening test for identifying patients with common disorders of calcium metabolism. The “normal” range of urinary calcium is not well established and varies according to many dietary factors and estrogen status in women (55). As a “rule of thumb,” urinary calcium may be considered elevated when it is greater than 250 mg per 24 hours in women; greater than 300 mg per 24 hours in men; or greater than 4 mg/kg body weight per 24 hours in either sex. It has been proposed that hypercalciuria can be easily classified as “renal” (renal calcium leak), “resorptive” (excess skeletal loss of calcium) or “absorptive” (increased intestinal absorption of calcium) (56). However, in clinical practice, these distinctions are not so easily established. Idiopathic hypercalciuria, perhaps the most common type of hypercalciuria (57), may be diagnosed if there are no underlying medical disorders (e.g., hyperparathyroidism, vitamin D toxicity, Paget’s disease of bone, multiple myeloma, sarcoidosis) and no obvious dietary excesses (e.g., calcium, sodium, protein, carbohydrates, alcohol) or deficiencies (e.g., phosphate, potassium) that are associated with hypercalciuria. In the absence of dietary calcium deficiency, vitamin D deficiency, malabsorption, liver disease, or chronic renal failure, low urinary calcium (less than 50 mg per 24 hours in women or men) is suggestive of calcium malabsorption and warrants further investigation. Celiac disease is a common (58) cause of asymptomatic malabsorption in osteoporosis that is treatable with a gluten-free diet.

 

ADDITIONAL STUDIES IN SELECTED PATIENTS

 

Celiac antibodies- Anti-endomysial antibody and tissue transglutaminase antibody are currently the serological markers of choice, with a higher sensitivity and specificity than anti-gliadin antibody and anti-reticulin antibody. If a serological marker is abnormal, or if there is a high clinical suspicion for celiac disease, the patient should be referred for endoscopy and small bowel biopsy.

 

Intact PTH- This may be elevated in patients with primary hyperparathyroidism or with secondary hyperparathyroidism due to disorders such as chronic kidney disease, vitamin D deficiency, or calcium malabsorption.

 

Serum protein electrophoresis and serum kappa/lambda light chain ratio- These are helpful tests to screen for possible multiple myeloma. Abnormal results may require further evaluation by an oncologist.

 

Overnight 1 mg dexamethasone suppression test or 24-hour urinary free cortisol- This is helpful to evaluate patients with suspected Cushing’s syndrome.

 

Serum total or free testosterone level- May be helpful in the assessment of men with osteoporosis.

 

Serum homocysteine- Elevated circulating homocysteine levels are associated with an increased risk of fractures (59). It is unknown whether reduction of homocysteine levels by increasing dietary intake of folic acid and vitamins B6 and B12 reduces the risk of fracture.

 

Serum tryptase and 24-hour urine for N-methylhistamine- Systemic mastocytosis is a rare cause of osteoporosis that can be diagnosed by a biopsy of typical skin lesions of urticaria pigmentosa, when present. Patients with systemic mastocytosis may sometimes present with osteoporosis and no other manifestations of the disease (60). When this disorder is suspected but skin lesions are not present, the finding of an elevated serum tryptase and/or urinary N-methyl histamine can be helpful, especially during or soon after a symptomatic episode of histamine release. However, normal values do not exclude the diagnosis. Bone marrow aspiration or biopsy, or non-decalcified double tetracycline labeled transiliac bone biopsy, may be necessary to confirm the diagnosis.

 

Serum bicarbonate- Renal tubular acidosis (RTA) has been associated with osteoporosis (61). With distal (type I) RTA, the serum bicarbonate is usually less than 15 mmol/l with a urine pH greater than 5.5 despite having systemic acidosis. This is due to the impaired ability of the distal nephron to secrete hydrogen ions effectively, which is a hallmark of the condition.

 

BONE TURNOVER MARKERS

 

Bone turnover markers (BTMs) are noninvasive laboratory tests of serum and urine that are readily available in clinical practice. While BTMs cannot be used to diagnose osteoporosis or determine the cause to osteoporosis, they have been very helpful in research to understand the pathophysiology of osteoporosis and other skeletal diseases and the mechanism of action of interventions used in the treatment of osteoporosis. In clinical practice, BTMs offer the potential of predicting fracture risk independently of BMD and may be useful in monitoring the metabolic effects of therapy (62). Drugs that are approved for the management of osteoporosis modulate bone remodeling in ways that are reflected by changes in BTMs. A decrease in BTMs with antiresorptive therapy is predictive of a subsequent increase in BMD (63)and reduction in fracture risk (64). The magnitude of BTM decrease with antiresorptive therapy is significantly associated with the level of fracture risk reduction, although the proportion of treatment effect due to the reduction in BTMs appears to vary according to the type of drug used (65). Teriparatide and abaloparatide, analogs of PTH and PTHrP, respectfully, are bone forming drugs associated with an increase in bone remodeling, with bone formation markers rising sooner and greater than bone resorption makers. Romosozumab is bone forming drug that uncouples bone resorption and formation, with an initial increase in bone formation markers and decrease in bone resorption markers.

 

Markers of bone resorption are mostly fragments of type I collagen, the main component of the organic bone matrix, which are released during osteoclastic bone resorption. These are measured in the serum or urine, with those available for clinical use including N-telopeptide of type I collagen (NTX), C-telopeptide of type I collagen (CTX), deoxypyridinoline (DPD), and pyridinoline (PYD). Bone formation markers are proteins secreted by osteoblasts or byproducts of type I collagen production by osteoblasts. They are measured in the serum and include bone specific alkaline phosphatase (BSAP), N-terminal propeptide of type I collagen (P1NP), and osteocalcin. CTX and P1NP have been proposed as the reference BTMs for clinical trials (66) and for clinical practice (67).

 

Clinical use of BTMs requires knowledge of their limitations as well as benefits. BTMs are subject to pre-analytical (biological) and analytical variability (62). Uncontrollable sources of pre-analytical variability include age, sex, menopausal status, pregnancy, lactation, fractures, co-existing diseases (e.g., diabetes mellitus, impaired renal function, and liver disease), drugs (e.g., glucocorticoids, anticonvulsants, and gonadotropin hormone releasing agonists), and immobility. Controllable pre-analytical sources of variability include time of day (circadian variability), fasting status, and exercise. Analytical sources of variability include specimen processing (e.g., collection, handling, and storage). Between-laboratory variability may be large (reported to be as much as a 7.3-fold difference), casting doubt on the validity of comparing specimens sent to different labs (68). Reference ranges for BTMs are not well established and may vary according to the population tested, the type of BTM, and the circumstances under which it is collected and processed.

 

In order to compare BTMs measurements longitudinally, it would be ideal to know the least significant change (LSC) and use this in a manner similar to what should be (but is probably not) common practice with DXA. However, the standards for calculating an LSC for a BTM are not as clear as with DXA, and the opportunity to do precision assessment for a BTM may not present itself. The Belgian Bone Club suggests using an estimated LSC by assuming an LSC of about 30% for serum BTMs and about 50-60% for urine BTMs (69). While the LSC for BTMs is almost always greater than for DXA, the magnitude of likely change is greater than DXA, with the “signal to noise ratio” that may be as good or even better than DXA. One strategy for the use of BTMs to monitor patients on antiresorptive therapy is to use absolute values rather that percent changes, as follows: treatment effect can be considered optimal when serum CTX has decreased by 100 ng/L or is below 280 ng/L, or when P1NP has decreased by 10 mcg/L or is less than 35 mcg/L (70).

 

A significant change of a BTM level in the appropriate direction following therapy is evidence that the patient is taking the drug regularly, taking it correctly, and that it is being absorbed and having the expected effect in modulating bone remodeling. Failure to achieve such a change in the BTM level is cause for concern and suggests that evaluation and possibly a reconsideration of treatment strategies. The use of BTMs allows assessment of drug effect sooner than with DXA, so that evaluation and corrective action, if needed, can be taken early in the course of therapy rather than later. Monitoring BTMs, especially in association with regular contact by a healthcare provider, may improve persistence with therapy (70). Despite the well-described limitations of BTMs, there is emerging support for their use in clinical practice, particularly in the assessment of response to therapy (62). Clinicians who are familiar with the benefits and limitations of BTMs may find them a helpful tool, in association with BMD testing, for managing patients with osteoporosis.

 

IMAGING STUDIES

 

Standard X-rays are used to diagnose fractures of all types and may sometimes suggest secondary causes of osteoporosis. Pseudofractures (Looser’s zones) are radiolucent lines running perpendicular to the bone cortex that may be seen in patients with osteomalacia. These probably represent stress fractures that have healed with poorly mineralized osteoid. Punctate radiolucencies may be seen in bone X-rays of patients with systemic mastocytosis. Primary hyperparathyroidism may cause bone cysts, subperiosteal bone resorption, brown tumors, and demineralization (‘salt and pepper’ pattern) of the skull. MRI, CT scanning, or nuclear imaging may be used to detect stress fractures that are not visible on X-ray. MRI of the spine is commonly used prior to vertebroplasty or kyphoplasty to determine the age of the fracture, the likelihood of the fracture being from causes other than osteoporosis, and whether there is retropulsion of bony fragments than could impair neurological function.

 

BONE BIOPSY

 

Non-decalcified double tetracycline labeled iliac crest bone biopsy is rarely used in clinical practice but may be helpful with difficult diagnostic problems. In the evaluation of renal osteodystrophy, a bone biopsy can distinguish between high turnover and low turnover bone disease, and possibly be an aid in the selection of therapy. With infiltrative disorders of bone, such as systemic mastocytosis, a bone biopsy or bone marrow aspiration may sometimes be the only way to make the diagnosis. In patients who are not responding to therapy as expected, or in patients with unusual presentations of osteoporosis, a bone biopsy may be indicated. Bone biopsies are required by the FDA for safety monitoring in clinical trials of osteoporosis drugs.

 

SUMMARY

 

Osteoporosis is a common skeletal disease with serious clinical consequences. Effective management of skeletal health includes appropriate selection of patients for bone density testing and assessment of risk factors for fracture. Prior to treatment, and when response to treatment is suboptimal, patients should be evaluated for secondary causes of osteoporosis. All reversible factors should be corrected and treatment should be individualized based on the clinical circumstances.

 

REFERENCES

 

  1. NIH Consensus Development Panel. Osteoporosis prevention, diagnosis, and therapy. JAMA. 2001;285(6):785-95.
  2. US Department of Health and Human Services. Bone Health and Osteoporosis: A Report of the Surgeon General. Rockville, MD: US Department of Health and Human Services, Office of the Surgeon General; 2004.
  3. WHO Study Group on Assessment of Fracture Risk and its Application to Screening for Postmenopausal Osteoporosis. Technical Report Series 843: Assessment of fracture risk and its application to screening for postmenopausal osteoporosis. Geneva: World Health Organization; 1994.
  4. Shuhart C, Cheung A, Gill R, Gani L, Goel H, Szalat A. Executive Summary of the 2023 Adult Position Development Conference of the International Society for Clinical Densitometry: DXA Reporting, Follow-up BMD Testing and Trabecular Bone Score Application and Reporting. J Clin Densitom. 2024;27(1):101435.
  5. Lewiecki EM, Binkley N, Petak SM. DXA Quality Matters. J Clin Densitom. 2006;9(4):388-92.
  6. Lewiecki EM, Binkley N, Morgan SL, Shuhart CR, Camargos BM, Carey JJ, et al. Best Practices for Dual-Energy X-ray Absorptiometry Measurement and Reporting: International Society for Clinical Densitometry Guidance. J Clin Densitom. 2016;19(2):127-40.
  7. LeBoff MS, Greenspan SL, Insogna KL, Lewiecki EM, Saag KG, Singer AJ, et al. The clinician's guide to prevention and treatment of osteoporosis. Osteoporos Int. 2022;33(10):2049-102.
  8. Qaseem A, Hicks LA, Etxeandia-Ikobaltzeta I, Shamliyan T, Cooney TG, Clinical Guidelines Committee of the American College of P, et al. Pharmacologic Treatment of Primary Osteoporosis or Low Bone Mass to Prevent Fractures in Adults: A Living Clinical Guideline From the American College of Physicians. Ann Intern Med. 2023;176(2):224-38.
  9. Gregson CL, Armstrong DJ, Bowden J, Cooper C, Edwards J, Gittoes NJL, et al. UK clinical guideline for the prevention and treatment of osteoporosis. Archives of osteoporosis. 2022;17(1):58.
  10. Kanis JA, Harvey NC, McCloskey E, Bruyere O, Veronese N, Lorentzon M, et al. Algorithm for the management of patients at low, high and very high risk of osteoporotic fractures. Osteoporos Int. 2020;31(1):1-12.
  11. Marshall D, Johnell O, Wedel H. Meta-analysis of how well measures of bone mineral density predict occurrence of osteoporotic fractures. BMJ. 1996;312(7041):1254-9.
  12. Wainwright SA, Marshall LM, Ensrud KE, Cauley JA, Black DM, Hillier TA, et al. Hip fracture in women without osteoporosis. J Clin Endocrinol Metab. 2005;90(5):2787-93.
  13. Lewiecki EM, Laster AJ. Clinical applications of vertebral fracture assessment by dual-energy X-ray absorptiometry. J Clin Endocrinol Metab. 2006;91(11):4215-22.
  14. Khan AA, Slart R, Ali DS, Bock O, Carey JJ, Camacho P, et al. Osteoporotic Fractures: Diagnosis, Evaluation, and Significance From the International Working Group on DXA Best Practices. Mayo Clin Proc. 2024;99(7):1127-41.
  15. International Society for Clinical Densitometry. Facility AccreditationMarch 11, 2018. Available from: http://www.iscd.org/accreditation/.
  16. Lotz JC, Cheal EJ, Hayes WC. Fracture prediction for the proximal femur using finite element models: Part I--Linear analysis. J Biomech Eng. 1991;113(4):353-60.
  17. Cranney A, Guyatt G, Griffith L, Wells G, Tugwell P, Rosen C, et al. Summary of meta-analyses of therapies for postmenopausal osteoporosis. Endocr Rev. 2002;23(4):570-8.
  18. Black DM, Bauer DC, Vittinghoff E, Lui LY, Grauer A, Marin F, et al. Treatment-related changes in bone mineral density as a surrogate biomarker for fracture risk reduction: meta-regression analyses of individual patient data from multiple randomised controlled trials. Lancet Diabetes Endocrinol. 2020;8(8):672-82.
  19. Bouxsein ML, Eastell R, Lui LY, Wu LA, de Papp AE, Grauer A, et al. Change in Bone Density and Reduction in Fracture Risk: A Meta-Regression of Published Trials. J Bone Miner Res. 2019;34(4):632-42.
  20. Hangartner TN. A study of the long-term precision of dual-energy X-ray absorptiometry bone densitometers and implications for the validity of the least-significant-change calculation. Osteoporos Int. 2007;18(4):513-23.
  21. Njeh CF, Fuerst T, Hans D, Blake GM, Genant HK. Radiation exposure in bone mineral density assessment. Applied Radiation & Isotopes. 1999;50(1):215-36.
  22. Bonnick SL. Bone densitometry in clinical practice- application and interpretation. 2nd ed ed. Totowa, N.J: Humana Press; 2004 2004.
  23. Lewiecki EM, Richmond B, Miller PD. Uses and misuses of quantitative ultrasonography in managing osteoporosis. CleveClin J Med. 2006;73(8):742-52.
  24. Fuggle NR, Reginster JY, Al-Daghri N, Bruyere O, Burlet N, Campusano C, et al. Radiofrequency echographic multi spectrometry (REMS) in the diagnosis and management of osteoporosis: state of the art. Aging Clin Exp Res. 2024;36(1):135.
  25. Lewiecki EM. Assessment of Skeletal Strength: Bone Density Testing and Beyond. Endocrinol Metab Clin North Am. 2021;50(2):299-317.
  26. Keaveny TM, Clarke BL, Cosman F, Orwoll ES, Siris ES, Khosla S, et al. Biomechanical Computed Tomography analysis (BCT) for clinical assessment of osteoporosis. Osteoporos Int. 2020;31(6):1025-48.
  27. Qasim M, Lopez Picazo M, Ruiz Wills C, Noailly J, Di Gregorio S, Del Rio Barquero LM, et al. 3D-DXA Based Finite Element Modelling for Femur Strength Prediction: Evaluation Against QCT. J Clin Densitom. 2024;27(2):101471.
  28. Vogt TM, Ross PD, Palermo L, Musliner T, Genant HK, Black D, et al. Vertebral fracture prevalence among women screened for the Fracture Intervention Trial and a simple clinical tool to screen for undiagnosed vertebral fractures. Fracture Intervention Trial Research Group. Mayo Clin Proc. 2000;75(9):888-96.
  29. Gunnes M, Lehmann EH, Mellstrom D, Johnell O. The relationship between anthropometric measurements and fractures in women. Bone. 1996;19(4):407-13.
  30. Siminoski K, Jiang G, Adachi JD, Hanley DA, Cline G, Ioannidis G, et al. Accuracy of height loss during prospective monitoring for detection of incident vertebral fractures. Osteoporos Int. 2005;16(4):403-10.
  31. Margolis KL, Ensrud KE, Schreiner PJ, Tabor HK. Body size and risk for clinical fractures in older women. Study of Osteoporotic Fractures Research Group. AnnIntern Med. 2000;133(2):123-7.
  32. de Laet C, Kanis JA, Oden A, Johanson H, Johnell O, Delmas P, et al. Body mass index as a predictor of fracture risk: a meta-analysis. OsteoporosInt. 2005;16(11):1330-8.
  33. Ensrud KE, Ewing SK, Stone KL, Cauley JA, Bowman PJ, Cummings SR. Intentional and unintentional weight loss increase bone loss and hip fracture risk in older women. J AmGeriatrSoc. 2003;51(12):1740-7.
  34. Lewiecki EM. Evaluation of the osteoporosis patient. In: Dempster DW, Cauley JA, Bouxsein ML, Cosman F, editors. Marcus and Feldman’s Osteoporosis. 2. Fifth ed. San Diego, CA, USA: Elsevier; 2021. p. 1475-500.
  35. Holick MF, Siris ES, Binkley N, Beard MK, Khan A, Katzer JT, et al. Prevalence of Vitamin D inadequacy among postmenopausal North American women receiving osteoporosis therapy. J Clin Endocrinol Metab. 2005;90(6):3215-24.
  36. Tannenbaum C, Clark J, Schwartzman K, Wallenstein S, Lapinski R, Meier D, et al. Yield of laboratory testing to identify secondary contributors to osteoporosis in otherwise healthy women. J Clin Endocrinol Metab. 2003;87:4431-7.
  37. Wagman RB, Marcus R. Beyond bone mineral density-navigating the laboratory assessment of patients with osteoporosis. J Clin Endocrinol Metab. 2002;87(10):4429-30.
  38. Barzel US. Recommended testing in patients with low bone density. J Clin Endocrinol Metab. 2003;88(3):1404-5.
  39. Luckey MM, Tannenbaum C. Authors' response: Recommended testing in patients with low bone density. J Clin Endocrinol Metab. 2003;88(3):1405.
  40. Harper KD, Weber TJ. Secondary osteoporosis - Diagnostic considerations. Endocrinol Metab Clin North Am. 1998;27(2):325-48.
  41. Eastell R. Treatment of postmenopausal osteoporosis. NEnglJMed. 1998;338(11):736-46.
  42. Favus MJ. Postmenopausal osteoporosis and the detection of so-called secondary causes of low bone density. J Clin Endocrinol Metab. 2005;90(6):3800-1.
  43. Looker AC, Orwoll ES, Johnston CCJ, Lindsay RL, Wahner HW, Dunn WL, et al. Prevalence of low femoral bone density in older U.S. adults from NHANES III. JBone MinerRes. 1997;12:1761-8.
  44. Orwoll ES, Klein RF. Osteoporosis in men. EndocrRev. 1995;16:87-116.
  45. Bedatsova L, Drake MT. The skeletal impact of cancer therapies. Br J Clin Pharmacol. 2019;85(6):1161-8.
  46. Rasch S, Lund T, Asmussen JT, Lerberg Nielsen A, Faebo Larsen R, Osterheden Andersen M, et al. Multiple Myeloma Associated Bone Disease. Cancers (Basel). 2020;12(8).
  47. Yu EW, Lee MP, Landon JE, Lindeman KG, Kim SC. Fracture Risk After Bariatric Surgery: Roux-en-Y Gastric Bypass Versus Adjustable Gastric Banding. J Bone Miner Res. 2017;32(6):1229-36.
  48. Adachi Y, Shiota E, Matsumata T, Iso Y, Yoh R, Kitano S. Osteoporosis after gastrectomy: bone mineral density of lumbar spine assessed by dual-energy X-ray absorptiometry. CalcifTissue Int. 2000;66(2):119-22.
  49. Bilezikian JP, Silverberg SJ, Bandeira F, Cetani F, Chandran M, Cusano NE, et al. Task Force #8: Management of Primary Hyperparathyroidism. J Bone Miner Res. 2022;Epub.
  50. Földes J, Tarján G, Szathmari M, Varga F, Krasznai I, Horvath C. Bone mineral density in patients with endogenous subclinical hyperthyroidism:Is this thyroid status a risk factor for osteoporosis. ClinEndocrinol(Oxf). 1993;39:521-7.
  51. Mancini T, Doga M, Mazziotti G, Giustina A. Cushing's Syndrome and Bone. Pituitary. 2004;7:249-52.
  52. Katz S, Weinerman S. Osteoporosis and gastrointestinal disease. Gastroenterol Hepatol (N Y). 2010;6(8):506-17.
  53. Cockcroft DW, Gault MH. Prediction of creatinine clearance from serum creatinine. Nephron. 1976;16(1):31-41.
  54. Levey AS, Coresh J, Greene T, Stevens LA, Zhang YL, Hendriksen S, et al. Using standardized serum creatinine values in the modification of diet in renal disease study equation for estimating glomerular filtration rate. AnnInternMed. 2006;145(4):247-54.
  55. Heaney RP, Recker RR, Ryan RA. Urinary calcium in perimenopausal women: normative values. Osteoporos Int. 1999;9(1):13-8.
  56. Power ML, Heaney RP, Kalkwarf HJ, Pitkin RM, Repke JT, Tsang RC, et al. The role of calcium in health and disease. Am J Obstet Gynecol. 1999;181(6):1560-9.
  57. Pacifici R. Idiopathic hypercalciuria and osteoporosis--distinct clinical manifestations of increased cytokine-induced bone resorption? J Clin Endocrinol Metab. 1997;82(1):29-31.
  58. Fasano A, Berti I, Gerarduzzi T, Not T, Colletti RB, Drago S, et al. Prevalence of celiac disease in at-risk and not-at-risk groups in the United States: a large multicenter study. ArchInternMed. 2003;163(3):286-92.
  59. McLean RR, Jacques PF, Selhub J, Tucker KL, Samelson EJ, Broe KE, et al. Homocysteine as a predictive factor for hip fracture in older persons. NEnglJ Med. 2004;350(20):2042-9.
  60. Lidor C, Frisch B, Gazit D, Gepstein R, Hallel T, Mekori YA. Osteoporosis as the sole presentation of bone marrow mastocytosis. J Bone Miner Res. 1990;5(8):871-6.
  61. Weger M, Deutschmann H, Weger W, Kotanko P, Skrabal F. Incomplete renal tubular acidosis in 'primary' osteoporosis. Osteoporos Int. 1999;10(4):325-9.
  62. Brown JP, Don-Wauchope A, Douville P, Albert C, Vasikaran SD. Current use of bone turnover markers in the management of osteoporosis. Clin Biochem. 2022;109-110:1-10.
  63. Greenspan SL, Parker RA, Ferguson L, Rosen HN, Maitland-Ramsey L, Karpf DB. Early changes in biochemical markers of bone turnover predict the long-term response to alendronate therapy in representative elderly women: a randomized clinical trial. J Bone Miner Res. 1998;13:1431-8.
  64. Eastell R, Barton I, Hannon RA, Chines A, Garnero P, Delmas PD. Relationship of early changes in bone resorption to the reduction in fracture risk with risedronate. J Bone Miner Res. 2003;18:1051-6.
  65. Bouxsein ML, Delmas PD. Considerations for development of surrogate endpoints for antifracture efficacy of new treatments in osteoporosis: a perspective. J Bone Miner Res. 2008;23(8):1155-67.
  66. Vasikaran S, Cooper C, Eastell R, Griesmacher A, Morris HA, Trenti T, et al. International Osteoporosis Foundation and International Federation of Clinical Chemistry and Laboratory Medicine Position on bone marker standards in osteoporosis. Clin Chem Lab Med. 2011;49(8):1271-4.
  67. Lee J, Vasikaran S. Current recommendations for laboratory testing and use of bone turnover markers in management of osteoporosis. Ann Lab Med. 2012;32(2):105-12.
  68. Seibel MJ, Lang M, Geilenkeuser WJ. Interlaboratory variation of biochemical markers of bone turnover. Clin Chem. 2001;47(8):1443-50.
  69. Bergmann P, Body JJ, Boonen S, Boutsen Y, Devogelaer JP, Goemaere S, et al. Evidence-based guidelines for the use of biochemical markers of bone turnover in the selection and monitoring of bisphosphonate treatment in osteoporosis: a consensus document of the Belgian Bone Club. Int J Clin Pract. 2009;63(1):19-26.
  70. Fontalis A, Eastell R. The challenge of long-term adherence: The role of bone turnover markers in monitoring bisphosphonate treatment of osteoporosis. Bone. 2020;136:115336.

 

Management of Diabetes and Hyperglycemia in Hospitalized Patients

ABSTRACT

 

Diabetes is the most prevalent metabolic disorder, and in 2021, the International Diabetes Federation estimated that it affected 537 million adults globally. In 2024, the United States Centers for Disease Control reported that 38.1 million adult Americans, or 14.7% of the adult population, have diabetes. Patients with diabetes have a 3-4-fold greater chance ofhospitalization compared to those without diabetes. In 2020, in the U.S., there were over 7.86 million hospital discharges for adults listed as having diabetes. Hyperglycemia, defined as a blood glucose greater than 140 mg/dl (7.8 mmol/l), isreported in 22-46% of non-critically ill hospitalized patients. Extensive data indicates that inpatient hyperglycemia, in patients with or without a prior diagnosis of diabetes, is associated with an increased risk of complications and mortality. In 2025, the American Diabetes Association (ADA) recommends that once therapy is initiated, a glycemic goal of 140–180 mg/dL (7.8–10.0 mmol/L) is recommended for most critically ill (ICU) individuals with hyperglycemia. More stringent individualized glycemic goals may be appropriate for selected critically ill individuals if they are achieved without significant hypoglycemia. However, for non-critically ill (non-ICU) individuals, a glycemic goal of 100-180 mg/dL (5.6-10.0 mmol/L) is recommended, if achieved without significant hypoglycemia. Insulin remains the best way to control hyperglycemia in the inpatient setting, especially in critically ill patients. Intravenously administered insulin is the preferredmethod to achieve the recommended glycemic target in the ICU. In 2025, the ADA changed its recommendations on using SGLT2 inhibitors in inpatients. They now suggest that in people with type 2 diabetes and heart failure, SGLT2 inhibitors may be started or continued if there are no contraindications (which include prolonged fasting or post-operative recovery). The use of GLP-1 receptor agonists was not recommended in previous guidelines because of the need for more safety and efficacy studies in the inpatient setting. However, increasing evidence indicates that treatment with oral agents such as DPP4 inhibitors, alone or combined with basal insulin, is safe and effective in general medicine andsurgery patients with mild to moderate hyperglycemia.

 

INTRODUCTION

 

Diabetes is the most prevalent metabolic disorder, affecting more than 537 million adults globally and is projected to rise to almost 800 million (10.9% of the adult population) by 2045 (1). In the United States, data from the National Diabetes Statistics Report in 2023 estimated that 38.4 million people of all ages or 14.7% of all U.S. adults had diabetes (2). The percentage of the population with diagnosed diabetes is expected to rise, with one study projecting that as many as onein three U.S. adults will have diabetes during their lifetime (3). People with diabetes have a 35% greater chance of referral for elective operations and a 3-4-fold greater chance of hospitalization compared to those without diabetes (4-7). Data from the US and Scotland estimate that of those individuals with a discharge diagnosis of diabetes, 30% will require two or more hospitalizations in any given year (5; 6; 8). In 2020, in the U.S., there were over 7.86 million hospital discharges for adults listed as having diabetes, (i.e., diabetes as either a principal diagnosis for hospitalization or as a secondary diagnosis, coexisting condition) (9). Data from the USA suggest that the prevalence of diabetes in the adult inpatient population has increased by 2.5% annually from 17.1% to 27.3% between 2000 and 2018 (10). In the UK, the annual National Diabetes Inpatient Audit suggested that the prevalence of diabetes amongst inpatients had risen from15% in 2010 to almost 20% in 2019 (11). In addition, those hospitalized with a diagnosis of diabetes stay in the hospitalfor longer than those without a diagnosis of diabetes admitted for the same condition (12; 13).

 

Diabetes was the 8th leading cause of death in the United States in 2021, accounting for 31.1 deaths per 100,000 of thepopulation (2). A further 120.3 per 100,000 people had diabetes listed as a contributing factor towards the cause of death (2). Not only does diabetes have a significant economic impact on those living with the condition, but it also imposes a substantial burden on the economy, with a total estimated cost of treating people diagnosed with diabetes in the UnitedStates in 2022 of $413 billion – or 25% of all health care spending in the US (14). This included $306.6 billion in directmedical costs. It is estimated that a further cost of $96.5 billion is incurred due to reduced productivity (14). Data from Ireland estimated that the overall cost of treating diabetes represented between 12 and 14% of the annual health budget. The cost per admission for someone with type 1 or type 2 diabetes was €4,027 and €5,026, respectively (15). Globally,diabetes care costs have been estimated at $1.3 trillion, rising to an estimated $2.1-2.5 trillion by 2030 (16; 17). This represents a rise in spending on diabetes as a proportion of global gross domestic product from 1.8% in 2015 to 2.2% in 2030 (17). Other than the costs of diabetes medications, the most significant component of this medical expenditure is hospital inpatient care (13; 18).

 

Hyperglycemia is defined as a blood glucose concentration greater than 140 mg/dl (7.8 mmol/l) (19-21). It is not just found in those with a pre-existing diagnosis of diabetes but in those with stress hyperglycemia or previously undiagnosed diabetes. The prevalence has been reported to be 22% to 46% in non-critically ill hospitalized patients (8; 19). Extensiveobservational and trial data indicate that inpatient hyperglycemia, in patients with or without a prior diagnosis of diabetes,is associated with an increased risk of complications and mortality, a longer hospital stay, a higher admission rate to the intensive care unit (ICU), and a higher need for transitional or nursing home care after hospital discharge (8; 22; 23).

 

Several studies and meta-analyses have shown that attempting ‘tight’ glycemic control using intensive insulin therapy isassociated with an increased risk of hypoglycemia (24-28), which has been associated with increased morbidity andmortality in hospitalized patients (19; 29-34). Thus, while insulin therapy is recommended for managing hyperglycemia inhospitalized patients, the concern about hypoglycemia has led leading professional organizations worldwide to recommend targets that avoid the risk of hypoglycemia (20; 27; 35-38).

 

This chapter reviews the pathophysiology of hyperglycemia during illness, the mechanisms for increased complicationsand mortality due to hyperglycemia and hypoglycemia, and the evidence supporting different therapies and approaches for the management of inpatient diabetes and hyperglycemia in critical care, general medicine, and surgical settings.

 

PREVALENCE OF DIABETES AND HYPERGLYCEMIA IN THE HOSPITALIZED PATIENT

 

Observational studies have reported a prevalence of hyperglycemia and diabetes ranging from 38% to 40% in hospitalized patients (8) and in 70-80% of those with diabetes who have a critical illness or cardiac surgery (39-41). A2017 report using point-of-care bedside glucose tests data in almost 3.5 million people (653,359 ICU and 2,831,436 non-ICU) from 575 hospitals in the United States reported a prevalence of hyperglycemia (defined as a glucose level >180mg/dl [10.0 mmol/l]) of 32.2% in ICU patients and in 32.0% of non-ICU patients (39). A study of 893 people across 69 ICUs in France reported a prevalence of hyperglycemia (>180 mg/dl [10 mmol/l]) of 45% (42). Other USA data suggest that between 2000 and 2018, the prevalence of diabetes amongst adult inpatients increased by 2.5% per year from 17.1% to 27.3% (10), and that over 33% of all hospital discharges in 2020 had diabetes listed as a diagnosis (9). However, this does not include those individuals who develop stress hyperglycemia. The American Diabetes Association (ADA) and American Association of Clinical Endocrinologists (AACE) consensus on inpatient hyperglycemia defined stress hyperglycemia or hospital-related hyperglycemia as any blood glucose concentration >140 mg/dl (>7.8 mmol/l) in patients without a prior history of diabetes (19; 20). The data from the US included those with newly identified diabetes or stress hyperglycemia as well as those with a prior diagnosis of diabetes (39). Although stress hyperglycemia typicallyresolves as the acute illness or surgical stress abates, a significant proportion (up to 60% in some reports) develop confirmed diabetes at 6-12 months after discharge (43; 44). A guide from the UK on the management of ‘diabetes at the front door’, also recommends that any individual without diabetes who presents acutely unwell should have a capillary glucose measurement and blood/urine ketone measurement taken, but that if it is high on admission (i.e. >140mg/dl [7.8 mmol/l]) and subsequently goes down to normal, then a diagnosis of stress hyperglycemia should be made and documented to the primary care team (21).

 

Measurement of HbA1c is indicated in people with hyperglycemia without a history of diabetes to differentiate betweenstress-induced hyperglycemia and previously undiagnosed diabetes (21; 45-48). The ADA also recommends that an HbA1c be done in those with diabetes who have not had it measured in the preceding 3 months (48). The Endocrine Society and the UK Joint British Diabetes Societies for Inpatient Care (JBDS) recommendations indicate that people hospitalized with elevated blood glucose >140 mg/dl (7.8 mmol/l) and an HbA1c of 6.5% (48 mmol/mol) or higher can be identified as having diabetes (19; 21). Given the increasing prevalence of diabetes, the UK has also produced a calculator to help teams work out their optimal staffing levels (49).

 

PATHOPHYSIOLOGY OF HYPERGLYCEMIA DURING ILLNESS

 

In subjects without diabetes during the fasted state, plasma glucose is maintained between 70 – 100 mg/dl (3.9 – 5.6 mmol/l) by a finely regulated balance between glucose production from the liver and kidneys and glucose utilization inperipheral tissues. Maintenance of near-normal glucose concentration is essential for cardiovascular and central nervous system function because the brain can neither synthesize nor store glucose (50; 51).

 

Systemic glucose balance is maintained by dynamic, minute-to-minute regulation of endogenous glucose production and glucose utilization by peripheral tissues (52). Glucose production is accomplished by gluconeogenesis or glycogenolysisprimarily in the liver and, to a lesser degree, by the kidneys (53; 54). Gluconeogenesis results from converting non-carbohydrate precursors such as lactate, alanine, and glycerol to glucose in the liver (55). Excess glucose is polymerized into glycogen, mainly stored in the liver and muscle. Hyperglycemia develops because of three processes: 1) increased gluconeogenesis, 2) accelerated glycogenolysis, and 3) impaired glucose utilization by peripheral tissues (Figure 1).

 

Figure 1. Pathogenesis of hyperglycemia. Hyperglycemia results from increased hepatic glucose production and impaired glucose utilization in peripheral tissues. Reduced insulin and excess counter-regulatory hormones (glucagon, cortisol, catecholamines, and growth hormone) increase lipolysis and protein breakdown (proteolysis) and impair glucose uptake by peripheral tissues. Hyperglycemia causes osmotic diuresis, leading to volume depletion, decreasing glomerular filtration rate, and worsening hyperglycemia. At the cellular level, increased blood glucose concentrations result in mitochondrial injury by generating reactive oxygen species and endothelial dysfunction by inhibiting nitric oxide production. Hyperglycemia increases levels of pro-inflammatory cytokines such as tumor necrosis factor-α (TNF-α) and interleukin [IL]-6, leading to immune system dysfunction. These changes can eventually lead to an increased risk of infection, impaired wound healing, multiple organ failure, prolonged hospital stay, and death. Adapted from ref (25).

 

From the quantitative standpoint, inappropriately increased hepatic glucose production represents the major pathogenic disturbance. Increased hepatic glucose production results from the high availability of gluconeogenic precursors. These include the amino acids alanine and glutamine, which result from accelerated proteolysis and decreased proteinsynthesis; lactate, which results from increased muscle glycogenolysis; glycerol, which results from increased lipolysis;and the increased activity of gluconeogenic enzymes (phosphoenol pyruvate carboxykinase, fructose-1,6-bisphosphatase, and pyruvate carboxylase) (53; 55).

 

Glucose metabolism is maintained by an interaction of glucoregulatory hormones – insulin and counter-regulatory hormones (glucagon, cortisol, epinephrine, norepinephrine, and growth hormone). Insulin controls hepatic glucose production by suppressing hepatic gluconeogenesis and glycogenolysis. Depending on the concentration in the circulation, insulin inhibits glycogenolysis and protein breakdown and, at higher concentrations, promotes protein anabolism ininsulin-sensitive tissues such as muscle, glucose uptake, and glycogen synthesis (52; 56; 57). In addition, insulin is a potent inhibitor of lipolysis, free fatty acid oxidation, and ketogenesis (56-58).

 

Counter-regulatory hormones also play an essential role in regulating glucose production and utilization. Glucagon is themost important glycogenolytic hormone, and therefore regulates hepatic glucose production in healthy individuals and in every state of hyperglycemia (53). During stress, excess concentration of counter-regulatory hormones results in altered carbohydrate metabolism by inducing insulin resistance, increasing hepatic glucose production, and reducing peripheralglucose utilization. In addition, high epinephrine levels stimulate glucagon secretion and inhibit insulin release bypancreatic β-cells (59; 60).

 

The development of hyperglycemia results in an inflammatory state characterized by an elevation of pro-inflammatory cytokines and increased oxidative stress markers (61-63). Circulating levels of TNF-α, IL-6, IL1-ß, IL-8, and C-reactiveprotein are significantly increased two- to fourfold on admission in people with severe hyperglycemia compared withcontrol subjects, and levels returned to normal levels after insulin treatment and resolution of hyperglycemic crises (61).Raised concentrations of TNF-α lead to insulin resistance at the level of the insulin receptor and through altered regulationof the insulin-signaling pathway (62; 64). In addition, preventing insulin-mediated activation of phosphatidylinositol 3-kinase TNF-α reduces insulin-stimulated glucose uptake in peripheral tissues (62; 64; 65).

 

CONSEQUENCES OF HYPERGLYCEMIA IN THE HOSPITALIZED PATIENTS

 

A large body of literature, including observational and prospective randomized clinical trials, in people with and withoutdiabetes, as well as those who are critically or non-critically ill has shown a strong association between hyperglycemia (in particular, a blood glucose >200mg/dl [11.0mmol/l]) and poor clinical outcomes, such as mortality, infections, and hospital complications compared to those with a glucose concentration of <100mg/dl (5.6mmol/l) (5; 66-76). This association correlates with the severity of hyperglycemia prior to or on admission and during the hospital stay (72; 77-79). Of interest, increasing evidence indicates an increased risk of complications and mortality in patients without a history of diabetes(stress-induced) compared to patients with a known diagnosis of diabetes (8; 69; 75; 77; 80; 81). It is not clear if stresshyperglycemia is the direct cause of poor outcomes or if it is a general marker of the severity of illness. However, there are data to show that those without a prior history of diabetes have fewer point-of-care glucose concentrations measured compared to those with diabetes, even when glucose concentrations are just as high (75; 82). In those who had diabetes, having more point-of-care tests increases contact with the ward staff, suggesting that impending complications may be picked up sooner, resulting in lower mortality. These data correlate with other work that also shows that those with lower preoperative HbA1c lower the number of post-operative glucose checks in a general surgical population (83).

 

The mechanisms implicated in the detrimental effects of hyperglycemia during acute illnesses are not entirely understood.Current evidence indicates that severe hyperglycemia results in impaired neutrophil granulocyte function, high circulating free fatty acids, and overproduction of pro-inflammatory cytokines and reactive oxygen species (ROS) that can result in direct cellular damage and endothelial and immune dysfunction (84; 85).

 

The majority of evidence linking hyperglycemia and poor outcomes comes from studies in the ICU. Falciglia et al., in a retrospective study of over 250,000 veterans admitted to various ICUs, reported that hyperglycemia is an independentrisk factor for mortality and complications (77). In a nonrandomized, prospective study, Furnary et al. followed 3,554 people with diabetes who underwent coronary artery bypass graft. These were treated with either intermittent subcutaneous insulin (SCI) or with a continuous intravenous insulin infusion (CIII). The group treated with SCI achieved an average blood glucose of 214 mg/dl (11.9 mmol/l), compared to 177 mg/dl (9.8 mmol/l) in the CIII group. The CIII group had significantly fewer deep sternal wound infections and a 50% lower risk-adjusted mortality (73; 86). In other ICU studies, patients with blood glucose levels >200 mg/dl (>11.1 mmol/l) were shown to have higher mortality comparedto those with blood glucose levels <200 mg/dl (<11.1 mmol/) (72; 75). Importantly however, once again it has been shown that it was those people who were not previously known to have diabetes yet who developed hyperglycemia on the ICU who fared worse (75; 87). This was confirmed by another ICU study looking at almost 350,000 people, looking at the outcomes of those with sepsis (88). These authors showed that having hyperglycemia without a prior diagnosis of diabetes was associated with an increased stay in hospital and ICU and greater 90-day mortality (88). However, there was no difference in outcomes for those with diabetes unless they had experienced severe hypoglycemia (<40 mg/dl [2.2 mmol/l]), in which case mortality rose (OR 2.95 95%CI 1.19-7.32) (88). Another ICU study randomized 9230 people who were not given early parenteral nutrition to liberal glucose control (insulin only started if glucose rose to >215 mg/dl [>11.9 mmol/l]), or tight glucose control with glucose concentrations maintained between 80 and 110 mg/dl (4.4 – 6.1 mmol/l). These authors showed no differences in outcome, including length of time in ICU, infection rates, time on respiratory or hemodynamic support, or mortality. The only differences were lower severe acute kidney injury incidence and cholestatic liver dysfunction in the tight glycemic control arm (89). 

 

The association of hyperglycemia and poor outcomes also applies to those not in ICU but admitted to general medicine, surgery, or mental health services. In such individuals, hyperglycemia is associated with poor hospital outcomes, including prolonged hospital stay, infections, disability after hospital discharge, and death (5; 8; 66; 67; 81; 90). In a study of 1,886 patients admitted to a community hospital, mortality in the general floors was significantly higher in patients withnewly diagnosed hyperglycemia and with known diabetes compared to subjects with normal glucose values (10% vs. 1.7% vs. 0.8%, respectively, p < 0.01) (8). In a prospective cohort multicenter study of 2,471 patients with community-acquired pneumonia, those with an admission glucose level of >198 mg/dl (>11.0 mmol/l) had a greater risk of mortality and complications than those with glucose <198 mg/dl (<11.0 mmol/l) (91). The risk of complications increased by 3% foreach 18 mg/dl (1.0 mmol/l) increase in admission glucose (91). In a retrospective study of 348 patients with chronic obstructive pulmonary disease and respiratory tract infection, the relative risk of death was 2.1 in those with a bloodglucose of 126-160 mg/dl (7.0-8.9 mmol/l), and 3.4 for those with a blood glucose of >162 mg/dl (9.0 mmol/l) compared topatients with a blood glucose of 108 mg/dl (6.0 mmol/l) (92). Similar data from a systematic review and meta-analysis from 38 studies of people who needed hospitalization for community-acquired pneumonia showed that in those without a prior diagnosis of diabetes, hyperglycemia was associated with an almost doubling of the need for ICU admission (crude OR 1.82, 95% CI 1.17 to 2.84) and in-hospital mortality (adjusted OR 1.28, 95% CI 1.09 to 1.50) (81). Those people already known to have diabetes had no increased risk of either. 

 

General surgery patients with hyperglycemia during the perioperative period are also at increased risk for adverse outcomes. Reviews of diabetes and the risk of surgical site infection across a variety of surgical specialties have shown that high peri-operative glucose is associated with an increased risk of infection (93; 94). In a case-control study, elevated preoperative glucose levels increased the risk of postoperative mortality in patients undergoing elective non-cardiac non-vascular surgery (95). Patients with glucose levels of 110-200 mg/dl (5.6-11.1 mmol/l) and those with glucose levels of >200 mg/dl (>11.1 mmol/l) had, respectively, 1.7-fold and  2.1-fold increased mortality compared to those with glucose levels <5.6 mmol/l (<110 mg/dl) (95). In another study, patients with glucose levels >220 mg/dl (>12.2 mmol/l) on the first postoperative day had a rate of infection 2.7 times higher than those who had serum glucose levels <220 mg/dl (<12.2 mmol/l) (96). Other authors showed an increase of postoperative infection rate by 30% for every 40mg/dl (2.2 mmol/l) rise in postoperative glucose level above 110 mg/dl (6.1 mmol/l) (96). Further, a study looking at perioperativeglycemic control and the effect on surgical site infections in people with diabetes undergoing foot and ankle surgery showed that 11.9% of those with a serum glucose ≥200 mg/dl (11.1 mmol/l) during the admission developed a surgicalsite infection versus only 5.2% of those with a serum glucose <200 mg/dl (11.1 mmol/l) (odds ratio = 2.45; 95% CI 1.09-5.52, P = 0.03) (97). Lastly, a prospective randomized study looking at the impact of glycemic control at 1-year post livertransplant showed that in those randomized to glycemic control of blood glucose below 140 mg/dl (7.8 mmol/l), any infection within one year occurred in 35 of the 82 patients (42.7%) versus 54 of 82 (65.9%) in those randomized to glycemic control of 180 mg/dl (10.0 mmol/l) (P = 0.0046) (98).

 

Emerging evidence suggests that early intervention and the use of technology allowing proactive identification of people at risk help to reduce hospital-acquired infection rates, episodes of hyper- and hypoglycemia, and, in some cases, length of stay (99-102). A meta-analysis also shows that improving peri-operative glycemic control reduced postoperative infection rates (103).

 

In summary, despite a large amount of work having been done, and the numerous data showing the association – but not causation – between hyperglycemia and poor outcomes, and because there remain a very few robust intervention studies showing a benefit of glycemic control, the optimal blood glucose concentration for people on ICU has yet to be determined (104; 105).

 

GLYCEMIC TARGETS IN THE ICU AND NON-ICU SETTINGS

 

The American Diabetes Association (ADA) and American Association of Clinical Endocrinology (AACE) task force on inpatient glycemic control and other groups recommended differing glycemic targets in the ICU setting (20) (Table 1). These guidelines suggest targeting a BG level between 140 and 180 mg/dl (7.8 and 10.0 mmol/l) for the majority of ICU patients and a lower glucose target between 110 and 140 mg/dl (6.1 and 7.8 mmol/l) in selected ICU patients (i.e., centers with extensive experience and appropriate nursing support, cardiac surgical patients, patients with stableglycemic control without hypoglycemia). Glucose targets >180 mg/dl (>10.0 mmol/l) or <110 mg/dl (<6.1 mmol/l) are not recommended in ICU patients. There is an argument that lowering glucose thresholds for hospital patients will likely be associated with harm (32). Still, an equally persuasive argument suggests that implementing the thresholds advocated by national and organizational guidelines has led to safer care (106).

 

The Society of Critical Care Medicine (SCCM) guidelines for the management of hyperglycemia in critically ill (ICU)patients recently “recommended against” titrating an insulin infusion to a lower glucose target of 80–139 mg/dL (4.4–7.7 mmol/L) as compared with a higher BG target range of 140–200 mg/dL (7.8–11.1 mmol/L) to reduce the risk of hypoglycemia (107). They also recommended that clinicians should initiate glycemic management protocols and procedures to treat persistent hyperglycemia greater than or equal to 180 mg/dL (10 mmol/L) to maintain target glucose below <180 mg/dl (<10.0 mmol/l) in critically ill adults (107). They also suggest that the insulin regimen and monitoringsystem be designed to avoid and detect hypoglycemia (blood glucose <70 mg/dl [<3.9 mmol/l]) and to minimize glycemicvariability.

 

Table 1. Major Guidelines for Treatment of Hyperglycemia in a Hospital Setting

 

ICU

Non-ICU

ADA/AACE (20; 108)

Initiate insulin therapy for persistent hyperglycemia (glucose >180 mg/dl [>10 mmol/l]).Treatment goal: For most people, target a glucose level between 140 – 180 mg/dl (7.8 – 10.0 mmol/l].More stringent goals (110 – 140 mg/dl [6.1 – 7.8 mmol/l]) or 100 – 180 mg/dL (5.6 –10.0 mmol/L), may be appropriate for selected patients and are acceptable if they can be achieved without significant hypoglycemia No specific guidelines.Insulin therapy should be initiated for the treatment of persistent hyperglycemia ≥180 mg/dL (10.0 mmol/L) and targeted to a glucose range of 140 –180 mg/dL (7.8 – 10.0 mmol/L) for most critically ill patients.
More stringent goals, such as 110–140 mg/dL (6.1–7.8 mmol/L), may be appropriate for selected patients (e.g., critically ill postsurgical patients or patients with cardiac surgery) as long as they can be achieved without significant hypoglycemia.Less stringent targets (e.g., >250 mg/dL (13.9 mmol/L) maybe appropriate in people withsevere comorbidities or end of life care.

ACP (27)

Recommends against intensiveinsulin therapy in those with orwithout diabetes in surgical / medical ICUs

Treatment goal: target glucosebetween 140 – 200 mg/dl (7.8 – 11.0 mmol/l), in people with or without diabetes, in surgical / medical ICUs

 

Critical Care Society (107)

BG >180 mg/dl (>10.0 mmol/l) should trigger insulin therapy.

Treatment goal: maintainglucose <180 mg/dl (<10.0 mmol/l) for most adults in ICU.

Maintain glucose levels <180mg/dl (10.0 mmol/l) while avoiding hypoglycemia.

 

Endocrine Society (19; 109)

 

Pre-meal glucose target <140mg/dl (<7.8mmol/l) and random blood glucose <180 mg/dl (<10.0 mmol/l). Those with insulin treated diabetes aim for a target glucose of 100 – 180 mg/dL (5.6 – 10 mmol/L). A lower target range may beappropriate in people able toachieve and maintain glycemiccontrol without hypoglycemia. Aglucose of <180 – 200 mg/dl(<10.0 – 11.0 mmol/l) isappropriate in those withterminal illness and/or withlimited life expectancy or at high risk for hypoglycemia.

Adjust antidiabetic therapywhen glucose falls <100 mg/dl (<5.6 mmol/l) to avoidhypoglycemia.

Society of ThoracicSurgeons (110)

Continuous insulin infusionpreferred over SC or intermittent intravenousboluses.

Treatment goal: Recommendglucose <180 mg/dl (<10.0 mmol/l) during surgery (≤110 mg/dl [≤6.1 mmol/l] in fasting and pre-meal states)

 

Joint British Diabetes Society for Inpatient Care(111)

 

Target blood glucose levels inmost people of between 108 – 180 mg/dl (6.0 – 10 mmol/l) with an acceptable range of between 72 – 216 mg/dl (4.0 – 12.0 mmol/l).

AACE/ADA, American Association of Endocrinologists and American Diabetes Association joint guidelines; ACP, American College of Physicians; ADA, American Diabetes Association; ICU, intensive care unit; SC, subcutaneous.

 

In the non-ICU setting, the Endocrine Society and the ADA/AACE Practice Guidelines recommended a pre-meal glucose of <140 mg/dl (<7.8 mmol/l) and a random BG of <180 mg/dl (<10.0 mmol/l) for the majority of non-critically ill patients treated with insulin (19; 20; 35; 109). More recently, the American Diabetes Association has recommended that targetglucose for most general medicine and surgery patients in non-ICU settings should be between 140 – 180 mg/dl (7.8 – 10.0 mmol/l) (108). In contrast, higher glucose ranges (>200 mg/dl [>11.1 mmol/l]) may be acceptable in people who are terminally ill or in those with severe comorbidities as a way of avoiding symptomatic hyperglycemia (19; 112).

 

Guidelines from the JBDS in the UK published over the last few years aim for target blood glucose concentrations in most people between 108 – 180 mg/dl (6.0 – 10.0 mmol/l) with an acceptable range of between 72 – 216 mg/dl (4.0 –12.0 mmol/l) (111). Table 1 summarizes the currently available guidelines for managing hyperglycemia in the hospitalsetting.

 

EVIDENCE FOR CONTROLLING HYPERGLYCEMIA IN ICU AND NON – ICU SETTINGS

 

In 2001, the Leuven surgical ICU study promoted intensive glycemic control in the critical care setting (113). This study randomized 1,548 people admitted to the surgical ICU (63% cardiac cases, 13% with diabetes, with most receiving earlyparenteral nutrition). Individuals were randomized to either conventional therapy, with target glucose between 180 – 200 mg/dl (10.0 – 11.1 mmol/l), or intensive treatment to target glucose between 80 – 110 mg/dl (4.4 – 6.1 mmol/l). Those in the conventional arm had a mean daily glucose average of 153 mg/dl (8.5 mmol/l), and those in the intensive arm had an average glucose of 103 mg/dl (5.7 mmol/l). Those in the intensive group had significantly less bacteremia, fewer antibioticrequirements, lower length of ventilator dependency, lower number of ICU days, and an overall 34% reduction in mortality(113). Following a similar study design, the same group of investigators randomized people to a medical ICU (18% withdiabetes) and reported that intensive insulin therapy (mean daily glucose of 111 mg/dl [6.2 mmol/l]) resulted in less ICU and total hospital complications in those with three days of insulin treatment (114). These two studies together, based on the positive outcomes on morbidity and mortality, suggested a glycemic target in the ICU of 140 – 180 mg/dl (7.8 – 10.0mmol/l) (20). There was also a realization that while lower targets may be appropriate for selected individuals, a target of <110 mg/dl (<6.1 mmol/l) was not recommended (20).

 

Many well-designed randomized controlled trials and meta-analyses have shown that such low glucose targets aredifficult to achieve, even in environments with high staff-to-patient ratios, without increasing the risk for severe hypoglycemia (24; 115-117). In addition, these and other studies failed to show improvement in clinical outcomes andhave even shown increased mortality risk with intensive glycemic control, targeting glucose concentrations of (80 – 110 mg/dl [4.4 – 6.1 mmol/l]) versus conventional glycemic control (140 – 200 mg/dl [7.8 – 11.0 mmol/l]) (Table 2) (29; 115-118). Most of these studies showed no differences in clinical outcomes between groups but had an increased risk of severe hypoglycemia in the intensively treated arms.  One study in ICU patients was the Normoglycemia in Intensive Care Evaluation and Surviving Using Glucose Algorithm Regulation (NICE-SUGAR) trial, which randomized over 6104subjects to receive either conventional glycemic control to target glucose <180 mg/dl [<10.0 mmol/l]) or intensiveglycemic control (target 81 – 108 mg/dl [4.5 – 6.0 mmol/l]). This study also reported no difference in hospital mortality butfound increased mortality at 90 days of follow-up (24.9% vs. 27.5%, p=0.02) (24). In a subsequent analysis of the trial, the NICE-SUGAR investigators reported a higher frequency of hypoglycemia in the intensive arm (6.8% vs. 0.5%), and those with hypoglycemia had a ~2-fold increase in mortality compared to patients without hypoglycemia (29). More recently, Gunst et al. recently published the results of a multicenter, randomized trial involving 9230 patients in medical and surgical ICUs (89). 4622 patients were assigned to liberal glucose control, where insulin was initiated only when the blood glucose level was above 215 mg per deciliter (11.9 mmol per liter), and 4608 patients were assigned to tight glucose control, targeting a glucose level between 80 and 110 mg per deciliter. The primary outcome, the duration of time in ICU care, did not differ significantly between the two trial groups. The hazard ratio for earlier discharge alive with tight glucose control was 1.00 with a 95% confidence interval of 0.96 to 1.04. Effective glycemic separation between the groups was observed, with a median absolute difference of -28 mg/dl (-1.6 mmol/l) in daily blood glucose levels. Additionally, the safety outcome, mortality within 90 days after randomization, was 10.1% in the liberal-control group and 10.5% in the tight-control group. The incidence of other secondary outcomes, including severe hypoglycemia, time to discharge alive from the hospital, use of respiratory support, or in-hospital mortality, were no different between intensive and relaxed glycemic targets, except for a trend in lower rates of liver and kidney injury in the tight control group.  Umpierrez recently summarized the data on mortality and outcomes of ICU RCTs (119).

 

Increasing evidence indicates that high pre-admission glycemic control – as measured by HbA1c >8.0% (64mmol/mol) is associated with lower mortality than those with an HbA1c <6.5% (48mmol/mol) (120). Whether this is due to an increased risk of hypoglycemia in the low HbA1c group or an increased frequency of monitoring or bedside vigilance in those with higher glucose or preadmission HbA1c remains unknown (75; 83).

 

Table 2. Clinical Trials of Intensive Glycemic Control in ICU Populations

Study

Setting

Population

Percentage with diabetes

Clinical Outcome

Malmberg, 1994 (121)

CCU

People with diabetes with suspected or confirmed acute MI

100

28% decreasemortality after 1 year

Furnary, 1999 (73)

CCU

People with diabetes undergoing CABG

100

65% decrease indeep sternal woundinfection rate

Van den Berghe,2001 (113)

Surgical ICU

Mixed, with CABG

13

34% decrease in mortality

Furnary, 2003 (86)

CCU

People with diabetes undergoing CABG

100

50% decrease inadjusted mortalityrate

Krinsley, 2003 (72)

Medical and surgical ICU

Mixed

22.4

27% decrease inmortality

Lazar, 2004 (122)

Operating room and ICU

People with diabetes undergoing CABG

100

 

60% decrease of post - operative atrialfibrillation

Van den Berghe,2006 (114)

Medical ICU

Mixed

17

18% decreasemortality

Gandhi, 2007 (123)

OperatingRoom

Mixed, undergoing cardiac surgery

19.6

No difference inmortality; increase instroke rate in the intensive treatmentarm

VISEP, 2008 (115)

Medical ICU

Mixed, admitted withsepsis

30

No differences in 28-day or 90-daymortality, end-organ failure, length of stay

De La Rosa, 2008(116)

Medical and surgical ICU

Mixed

12

No differences in 28-day mortality orinfection rate

Glucontrol, 2009 (124)

Medical and surgical ICU

Mixed

18

No difference in 28-day mortality

NICE-SUGAR,2009/2012 (24; 29)

Medical and surgical ICU

Mixed

20

No difference in 90-day mortality

Boston Children’s(SPECS), 2012 (125; 126)

Cardiac ICU

Cardiac surgery,people without diabetes

0

No differences in 30-day mortality, length of stay, in the cardiac ICU, length ofhospital, duration ofmechanicalventilation andvasoactive support,or measures of organfailure

ChiP, 2014 (127)

Pediatric ICU

Criticalillness/injury/majorsurgery, those without diabetes.

0

No difference in 30-day mortality.Increasedhypoglycemia in theintensive treatedgroup

CGAO–REA, 2014 (128; 129)

Medical ICU

Mixed

23

No difference in 90-day mortality. Increasedhypoglycemia in theintensive treatedgroup

Okabayashi, 2014 (130)

 

Surgical ICU

Mixed

25.3

Decreased surgicalsite infection in theintensive treatedgroup

Umpierrez (GLUCOCABG) 2015

Surgical ICU

CABG

50%

No difference in mortality

 

Gunst et al

ICU

ICU

 

XX

No difference in mortality

 

MI, myocardial infarction, ICU – Intensive Care Unit, CABG – Coronary artery bypass graft. Mixed-study enrolled those with and without diabetes.

 

The GLUCO-CABG trial was a randomized open-label clinical study that included those with and without diabetesundergoing CABG who experienced perioperative hyperglycemia, defined as a BG >140 mg/dl (>7.8 mmol/l) 6069 (70). A total of 302 people between 18 and 80 years of age were randomized to the intensive glycemic control group (target BG 100 – 140 mg/dl [5.6 – 7.8 mmol/l]) or the control group (BG 141 – 180 mg/dl [7.9 – 10.0 mmol/l]) in the ICU. Aftertransitioning from the ICU to the telemetry floor, patients were managed with a single treatment protocol to maintain a glucose target of <140 mg/dl (<7.8 mmol/l) before meals during the hospital stay. The primary outcome included differences between intensive and conservative glucose control on a composite of perioperative complications, includingsternal wound infection, bacteremia, respiratory failure, pneumonia, acute kidney injury, major adverse cardiovascular events including acute coronary syndrome, stroke, heart failure, and cardiac arrhythmias (70). The mean BG during the ICU stay was 132±14 mg/dl (7.3±0.8 mmol/l) in the intensive and 152±17 mg/dl (8.4±1.0 mmol/l) in the conservative group. Intensive glucose treatment resulted in a 20% reduction in perioperative complications compared to theconservative group (42% vs. 52%). Of interest, there were no differences in the rate of complications among patients with diabetes treated with intensive or conservative regimens (42% vs. 52%, p=0.08); however, intensive treatment wasassociated with a significantly lower rate of complications compared to the conservative group in those without diabetes (34% vs. 55%, p=0.008) (70). Hospitalization costs were lower in the intensive group (median [IQR] $36,681 [28,488 – 46,074] vs. $40,913 [31,464 – 56,629], p=0.04), with an average total cost savings of $3,654 per case compared to conservative glucose control (131).

 

To date, few large studies have been conducted to determine if improved control in those not in ICU may result inreduced morbidity and mortality in general medical and surgical patients – indeed, until recently, for most people in hospital with diabetes while there are observational data to show that dysglycemia is harmful, there were little data to show that improving glycemic control helps (132). A randomized controlled trial from 2011 reported that improved glucose control using a basal-bolus regimen may reduce hospital complications in general surgery patients (71). Improving glucose control with a basal-bolus regimen significantly reduced the frequency of composite complications, including postoperative wound infection, pneumonia, bacteremia, and acute renal and respiratory failure (71). In that study, treatment with basal-bolus insulin reduced average total inpatient costs per day by 14% or $751 compared totreatment with a correction bolus dose insulin alone (133). Another study from Australia has shown that in a randomized study of 1371 surgical inpatients, 680 with even a single glucose value >200 mg/dl (11.1 mmol/l) received early intervention from the diabetes inpatient team (134). This led to reductions in glucose of a modest -5.4 mg/dl (-0.3 mmol/l), which still equated to a 4.6%, statistically significant reduction in hospital-acquired infections compared to those receiving standard care (134).

 

HYPOGLYCEMIA

 

Hypoglycemia is the most common side effect of treatment of all types of diabetes and stress hyperglycemia in thehospital setting. It presents a significant barrier to satisfactory long-term glycemic control. Hypoglycemia results from animbalance between glucose supply, glucose utilization, and current insulin levels. Hypoglycemia is defined as a lower-than-normal level of blood glucose. Hypoglycemia is defined as any glucose level <70 mg/dl (<3.9 mmol/l) (108; 135) for hospital inpatients. Level 1 hypoglycemia is a glucose concentration of 54 – 70 mg/dL (3.0 – 3.9 mmol/L). Level 2 hypoglycemia is a blood glucose concentration of <54 mg/dL (3.0 mmol/L) (108). Severe hypoglycemia has been defined by many as <40 mg/dl (<2.2 mmol/l) (136), but a newer definition, Level 3 hypoglycemia, is a glucose concentration low enough where the individual requires third-party assistance to aid recovery (108). The UK JBDS guideline suggests that the lower limit of glucose in the inpatient population should be 108 mg/dl (6.0 mmol/l), and that the range between 72 – 108 mg/dl (4.0 – 6.0 mmol/l) be designated ‘looming’ hypoglycemia, to alert ward staff to take action because of the possibility that lower glucose levels may be associated with harm (137). The exception to this is those people on a diet only or those people on an insulin pump / closed loop system who can self-manage their diabetes while in the hospital.

 

The incidence of severe hypoglycemia in the different trials ranged between 5% and 28%, depending on the intensity ofglycemic control in the ICU (138). Rates from subcutaneous insulin trials in non-critically ill patients range from less than 1% to 33% (71; 139; 140). In 2017, the UK National Diabetes Inpatient Audit (NaDIA) data showed that 18% of peoplewith diabetes in hospital experienced one or more hypoglycemic episodes with a blood glucose <72mg/dl (<4.0 mmol/l) –down from 26% in 2011, with 7% (1 in 14) of all inpatients experiencing episodes requiring third party assistance to administer rescue therapy (141). The NaDIA data also showed that those with type 1 diabetes had the highestprevalence, with 25% experiencing a severe hypoglycemic episode (141). Furthermore, 1.3% (1 in 80) of those in hospitals with diabetes required some form of injectable rescue treatment (i.e., IV glucose or IM glucagon), down from2.1% in 2011 (141). The same data showed that the highest proportion of episodes occurred overnight (28%) between 05:00 and 09.00 AM when snack availability was likely the lowest (140; 141).

 

Table 3 lists some key factors that predict the likelihood of someone experiencing a hypoglycemic event whilehospitalized. These also include older age, greater illness severity, diabetes, and the use of oral glucose-loweringmedications and/or insulin (137; 142-145). In-hospital processes of care that contribute to the risk for hypoglycemia include unexpected changes in nutritional intake that are not accompanied by associated changes in the glycemic management regimen. Examples include (but are not limited to) cessation of nutrition for procedures, an adjustment in the amount of nutritional support, interruption of the established routine for glucose monitoring, deviations from the established glucose control protocols, and failure to adjust therapy when glucose is trending down, or steroid therapy isbeing tapered (137; 146; 147). A common cause of inpatient hypoglycemia is when handwritten insulin prescriptions lead to errors, including misreading, e.g., when ‘U’ is used for units (i.e., 4U becoming 40 units) or confusing the insulin name with the dose (e.g., Humalog Mix25 becoming Humalog 25 units) (148). Electronic prescribing has been associated with a lower rate of prescription errors (141).

 

However, other factors may also be involved, such as concurrent use of drugs with hypoglycemic agents, e.g., warfarin, quinine, salicylates, fibrates, sulphonamides (including co-trimoxazole), monoamine oxidase inhibitors, NSAIDs, probenecid, somatostatin analogs, or selective serotonin reuptake inhibitors. Secondary causes of inpatienthypoglycemia include loss of counter-regulatory hormone function, e.g., Addison’s disease, growth hormone deficiency,hypothyroidism, or hypopituitarism.

 

Table 3. Common Risk Factors for Developing Hypoglycemia in the Hospital

Prior episode of hypoglycemia

Older age

Chronic kidney disease

Congestive heart failure

Liver Failure

Sepsis

Malnutrition

Erratic eating patterns / Nutritional interruptions / Lack of access to carbohydrates

Malignancies

Insulin regimen

Type 1 diabetes

Mental status changes

Certain concomitants use of medications

Duration of diabetes

 

The development of hypoglycemia is associated with adverse hospital outcomes (29; 30; 117; 118; 124; 144; 149-155). Turchin et al. examined data from 4,368 admission episodes for people with diabetes, of which one-third were on regular insulin therapy (30). Patients experiencing inpatient hypoglycemia experienced a 66% increased risk of death within one year and spent 2.8 days longer in the hospital compared to those not experiencing hypoglycemia. A 2019 systematic review and meta-analysis of hospital-acquired hypoglycemia in non-ICU patients suggested that adults exposed to glucose levels <72 mg/dl (<4.0 mmol/l) experienced a mean increased length of hospital stay of 4.1 days (95% CI 2.36 – 5.79) compared to those who did not experience hypoglycemia (144). The same dataset suggested an increased relative risk of in-hospital mortality for non-ICU patients of 2.09 (95% CI 1.64 – 2.67) (144). There was a non-significant reduction in mortality for those in ICU of 0.75 (95% CI 0.49 – 1.16) (144). The odds ratio (95% confidence interval) for mortality associated with one or more episodes was 2.28 (1.41-3.70, p=0.0008) among a cohort of 5,365 patients admitted to amixed medical-surgical ICU (142). In a larger cohort of over 6,000 patients, hypoglycemia was associated with longer ICU stays and greater hospital mortality, especially for patients with more than one episode of hypoglycemia (29). These data strengthen the argument to have potentially less strict glycemic targets for those not on ICU (32; 137). For example, if an individual has a glucose of 75 mg/dl (4.2 mmol/l), and is on an intravenous insulin infusion, by the time their bedside capillary glucose is next measured, they may have a glucose well below 72 mg/dl (4.0 mmol/l), thus they have come to potential harm. Indeed, data published from previous NaDIA surveys and NaDIA Harms using data from over 100 hospitals across the UK showed several serious adverse events, including seizures, permanent cerebral damage, cardiac arrests, and deaths. Insulin therapy was implicated in several of these events (33; 34; 156; 157). The counterargument is that there are initiatives to reduce the risk of developing inpatient hypoglycemia and having national guidance has led to improved patient care overall (106; 158). As with the outpatient population, the increased use of technology may help avoid hypoglycemia (159).

 

Hypoglycemia has been associated with adverse cardiovascular outcomes, such as increased myocardial contractility, prolonged QT interval (possibly due to the rapid drop in potassium concentrations due to the increased circulating epinephrine and norepinephrine), ischemic electrocardiogram changes and repolarization abnormalities, angina, arrhythmias, increased inflammation, and sudden death, (51; 160-162). The mechanisms for the poor outcome have yet to be entirely understood. Still, hypoglycemia has been associated with increases in pro-inflammatory cytokines (TNFα,IL-1β, IL-6, and IL-8), markers of lipid peroxidation, acute changes in endothelial dysfunction with associated vasoconstriction, increased blood coagulability, cellular adhesion, and oxidative stress (163; 164).

 

Despite these observations, the direct causal effect of iatrogenic hypoglycemia on outcome is still debatable. Kosiborod et al. reported that spontaneous hypoglycemia, but not insulin–induced hypoglycemia, was associated with higherhospital mortality (152). Similarly, another study among 31,970 patients also reported that hypoglycemia is associatedwith increased in-hospital mortality. Still, the risk was limited to patients with spontaneous hypoglycemia and not topatients with drug-associated hypoglycemia (165). These studies raise the possibility that hypoglycemia, like hyperglycemia, despite the biochemical and other changes described, is a marker of disease burden rather than a directcause of death.

 

RECOMMENDATIONS FOR MANAGING HYPERGLYCEMIA IN THE  HOSPITAL ENVIRONMENT

 

Knowledge of Diabetes Management Amongst Medical Staff

 

The burden on inpatient diabetes falls most frequently on junior medical staff, who often have little or no specialist diabetes training. As such, it is perhaps unsurprising that errors occur. In the UK, a survey of junior doctors showed that unlike other commonly encountered medical conditions, such as acute asthma or angina, their knowledge about and confidence in managing diabetes was significantly lower (166). In 2019, this was also shown in a multicenter study from the US – with the major difference being that whilst most staff felt confident and comfortable managing diabetes, when challenged on how to manage certain situations, and in particular identifying glucose targets for those who were critically ill or the threshold for defining hypoglycemia, their confidence was far higher than their knowledge – a potentially devastating combination (167). Given the high prevalence of diabetes amongst hospital inpatients, essential diabetes management should be part of mandatory training. However, studies have found that despite the implementation of training programs, structured staff education has not shown to be of significant benefit in terms of improved patient outcomes (168; 169)

 

Management of Inpatient Hyperglycemia in the ICU

 

Insulin is the best way to control hyperglycemia in the inpatient setting, especially in critically ill patients. A variable-rate intravenous insulin infusion is the preferred method to achieve the recommended glycemic target (ADA Standards of Care 2025). The short half-life of intravenous insulin makes it ideal in this setting because it allows flexibility in the eventof unpredicted changes in an individual’s health, medications, and nutrition.

 

When someone is identified as having hyperglycemia (blood glucose ≥180 mg/dl [≥10.0 mmol/l]), a variable rate intravenous insulin infusion should be started to maintain blood glucose levels <180 mg/dl (<10.0 mmol/l). A variety ofintravenous infusion protocols are effective in achieving glycemic control with a low rate of hypoglycemic events and in improving hospital outcomes (73; 86; 113; 121; 170-174). A proper protocol should allow flexible blood glucose targets to be modified based on the individual’s clinical situation. Further, it should have clear instructions about the blood glucosethreshold for initiating an insulin infusion and the initial rate. The appropriate fluids should also be prescribed. It should bevalidated to avoid hyperglycemia if adjusted too slowly and hypoglycemia if adjusted too fast. Accurate insulin administration requires a reliable infusion pump that can deliver the insulin dose in increments of 0.1 units per hour (138; 172).

 

There is no ideal insulin protocol for managing hyperglycemia in the critically ill patient. In addition, no clear evidence demonstrates the benefit of one protocol/algorithm versus any other (138). Implementing any of these algorithms requires close follow-up by the nursing staff and is prone to human errors. Some institutions have developedcomputerized protocols that can be implemented to avoid errors in dosing (138; 175-179). Essential elements thatincrease protocol success of continuous insulin infusion are: 1) rate adjustment considers the current and previous glucose value and the current rate of insulin infusion, 2) rate adjustment considers the rate of change (or lack of change) from the previous reading, and 3) frequent glucose monitoring (hourly until stable glycemia is established, and then at least every 2 – 3 hours) (138; 171; 180-182).

 

Several computer-based algorithms aiming to direct the nursing staff in adjusting the insulin infusion rate have become commercially available (175-177; 179; 183). Retrospective cohorts and controlled trials have reported a more rapid and tighter glycemic control with computer-guided algorithms than standard paper form protocols in ICU patients (176; 184), as well as lower glycemic variability than patients treated with the standard insulin infusion regimens. Despite differences in glycemic control between insulin algorithms, another study showed no difference between computerized protocolsversus conventional glucose control (128). Thus, most insulin algorithms appear to be appropriate alternatives for managing hyperglycemia in critically ill patients, and the choice depends upon the physician’s preferences, staffing availability, and cost considerations. As mentioned, the increasing implementation of available technology, in particular the use of closed loops should improve the management of dysglycemia over the coming years (185-187).

 

Managing Hyperglycemia in the Non-ICU Setting

 

Subcutaneous insulin is the preferred therapeutic agent for glucose control in those admitted to non-ICU settings under general medicine and surgery. A recent study suggested that the use of bolus correction doses of subcutaneous insulin (“subcutaneous sliding scale insulin” (SSI)) is an acceptable way of controlling dysglycemia, particularly in those whose admission glucose levels were <180 mg/dl (10 mmol/l) (188; 189). However, many studies do not agree and advocate against using this method as the only way to control glucose levels because it results in undesirable hypoglycemia andhyperglycemia or inadequately controls dysglycemia (109; 190-193). It has become evident in recent years that the useof scheduled subcutaneous insulin therapy with basal (e.g. glargine, detemir or degludec) once daily or with intermediate-acting insulin (NPH) given twice daily alone or in combination with short (regular) or rapid-acting insulin (lispro, aspart, glulisine) prior to meals is effective and safe for the management of most patients with hyperglycemia and diabetes (20; 108; 194).

 

The basal-bolus (prandial) insulin regimen is considered the physiologic approach as it addresses the three componentsof insulin requirement: basal (what is required in the fasting state), nutritional (what is needed for peripheral glucosedisposal following a meal), and supplemental (what is necessary for unexpected glucose elevations, or to dispose ofglucose in hyperglycemia (195).

 

A prospective, randomized multi-center trial compared the efficacy and safety of a basal/bolus insulin regimen with basal-bolus regimen and SSI in people with type 2 diabetes admitted to a general medicine service (139). The use of a basal-bolus insulin regimen improved blood glucose control more than the subcutaneous sliding scale alone. A blood glucose target <140 mg/dl (<7.8 mmol/l) was achieved in 66% of those in the glargine plus glulisine group and 38% in the slidingscale group (139). The incidence of hypoglycemia, defined as a BG <60 mg/dl (<3.3 mmol/l), was less than 5% in thosetreated with basal-bolus or SSI. A different study on general surgery inpatients also compared the efficacy and safety of a basal-bolus regimen to SSI in those with type 2 diabetes (71). The basal-bolus regimen resulted in a significant improvement in glucose control and a reduction in the frequency of the composite of postoperative complications, including wound infection, pneumonia, respiratory failure, acute renal failure, and  bacteremia.

 

Multi-dose human NPH and regular insulin have been compared to basal-bolus treatment with insulin analogs in an open-label, controlled, multicenter trial in 130 medical admissions with type 2 diabetes (196). This study found that both treatment regimens significantly improved inpatient glycemic control with a glucose target of <140 mg/dl (<7.8 mmol/l) before meals and no difference in the rate of hypoglycemic events. Thus, a similar improvement in glycemic control can be achieved with either basal-bolus therapy with insulin analogs or with NPH/regular human insulin in people with type 2diabetes.

 

Most people in the hospital have reduced caloric intake due to a lack of appetite, medical procedures, or surgical intervention. In the Basal Plus trial, people with type 2 diabetes who were treated with diet, oral antidiabetic agents, orlow-dose insulin (≤ 0.4 unit/kg/day) prior to admission were randomized to receive a standard basal-bolus regimen withglargine once daily and glulisine before meals or a single daily dose of glargine. In addition, supplemental doses ofglulisine were administered for correction of hyperglycemia (>140 mg/dl [>7.8 mmol/l]) per sliding scale (197). This study reported that the basal approach resulted in similar improvement in glycemic control and the frequency of hypoglycemia compared to a standard basal-bolus regimen (197). Thus, in insulin-naive individuals or those receiving low-dose insulin on admission, as well as those with reduced oral intake, the use of a basal plus regimen is an effective alternative to basal-bolus (108).

 

The recommended total daily insulin dose should start between 0.3 to 0.5 units per Kg (139; 147; 198; 199) for mostpeople with diabetes. Starting doses greater than 0.6 – 0.8 units/kg/day have been associated with 3-fold higher odds of hypoglycemia than doses lower than 0.2 U/kg/day. In elderly individuals or those with impaired renal function, lower initial daily doses (≤ 0.3 units/kg) may lower the risk of hypoglycemia (200).

 

Hospital Use of Non-Insulin Therapy in Non-Critical Care Settings

 

Several other classes of non-insulin glucose-lowering agents have been tried in the hospital setting. However, most are not suitable for use. Metformin, while the first line for type 2 diabetes in the outpatient setting, may not be appropriate where there is any evidence of dehydration, renal impairment, or if intravenous contrast is due to be administered due to the risk of lactic acidosis or worsening of renal function (195). Despite the lack of robust evidence of benefit, it remains in everyday use in many countries (201). Thiazolidinediones are excellent at lowering glucose but are used rarely, and possibly inappropriately, in hospitalized patients because they take several weeks to reach their maximum effect, may precipitate heart failure, and may cause peripheral edema due to fluid retention (202-204).

 

Sulfonylureas work rapidly and are often the drugs of choice for worsening diabetes in an outpatient setting (205). They remain in everyday use in many countries, with up to 20% of inpatients with diabetes in the USA and UK remaining on them (140; 203). However, they increase the risk of hypoglycemia. There is data to show that they remain one of the most frequent causes of inpatient hypoglycemia, thus extending the length of hospital stay and increasing the risk of inpatient mortality (141; 206-208).

 

Oral glucose-lowering medication use is limited by the delay and unpredictability of onset of action, and there is also concern regarding the cardiovascular effects of sulfonylureas and the contraindication of metformin use in patients with renal or liver dysfunction (19; 209). Recent work using the sodium-glucose co-transporter 2 inhibitors for corticosteroid-induced hyperglycemia in acute exacerbation of chronic obstructive pulmonary disease (COPD) or used in COVID infections failed to demonstrate an improvement in outcomes (210; 211). Indeed, despite their clear benefits in the outpatient population with and without diabetes, robust evidence for the benefit of SGLT2i use in the inpatient population (in people with diabetes) is lacking (212; 213).

 

The use of oral antidiabetic agents was not recommended in previous guidelines because of the need for more safety and efficacy studies in the inpatient setting (20). However, increasing evidence indicates that treatment with dipeptidyl peptidase-4 (DPP4) inhibitors, alone or in combination with basal insulin, is safe and effective in general medicine and surgery with mild to moderate hyperglycemia (48). In a pilot study, general medicine and surgical inpatients with bloodglucose between 140 and 400 mg/dl (7.8 – 22.2 mmol/l) treated with diet, oral antidiabetic drugs, or low-dose insulin (≤0.4 U/kg/day) were randomized to sitagliptin once daily, sitagliptin and basal insulin, or basal-bolus insulin (214). All groups received correction doses of lispro before meals and bedtime for blood glucose >140 mg/dl (>7.8 mmol/l). In those with mild-moderate hyperglycemia (<180 mg/dl [<10 mmol/l]), the use of sitagliptin plus supplemental (correctiondoses) or in combination with basal insulin resulted in no significant differences in mean daily blood glucose, frequency of hypoglycemia or the number of treatment failures compared to the basal-bolus regimen (214). The SITA-HOSPITAL trial, a multicenter, randomized controlled study in 279 general medicine and surgery individuals with type 2 diabetes previously treated with oral anti-diabetic agents or low-dose insulin (<0.6 U/kg/d), also reported similar glycemic control,hypoglycemia rate, hospital length-of-stay, treatment failures or hospital complications (including acute kidney injury orpancreatitis) between the combination of oral sitagliptin plus basal insulin to the more labor-intensive basal-bolus insulin regimen (215).

 

Analysis from prospective studies using DPP4-i in various inpatient situations with type 2 diabetes (T2D) reported that treatment with DPP4-i alone or with basal insulin suggested they were safe and lowered glucose concentrations without increasing the risk of hypoglycemia (216; 217).

 

For people with type 2 diabetes hospitalized with heart failure, the ADA has recommended that the use of a sodium-glucose cotransporter 2 (SGLT2) inhibitor be initiated or continued during hospitalization and upon discharge if there are no contraindications and after recovery from the acute illness (218-221). In patients with acute heart failure, empagliflozin was well tolerated, resulting in significant clinical benefits, including heart failure readmissions and quality of life (221). However, SGLT2 inhibitors should be avoided in cases of severe illness, in people with type 1 diabetes, ketonemia, or ketonuria, and during prolonged fasting and surgical procedures. Proactive adjustment of diuretic dosing is recommended during hospitalization and/or discharge, especially in collaboration with a cardiology/heart failure consult team. The FDA has warned that SGLT2 inhibitors should be stopped three days before scheduled surgeries (4 days in the case of ertugliflozin) (222). This differs from the UK guideline, which states that these drugs should be omitted from the day before a procedure (223).

 

Staffing Levels in the Hospital

 

Inadequate levels of appropriately knowledgeable staff are a concern for patients with diabetes (224). An insufficient level of specialist diabetes staff is a factor that inhibits safe and optimal care (34). Recently, the UK JBDS group developed a simple calculator into which individual teams could enter data to calculate their staffing needs (49). That data showed the discrepancy between the number of people delivering care and the number of people that specialist teams felt was necessary to provide safe and effective care for five days or seven days per week. This was for senior medical staff, specialist nursing staff, dieticians, podiatrists, pharmacists and psychologists (49).

 

Glucose Monitoring in the Hospital

 

All patients admitted to the hospital with a diagnosis of diabetes and those with newly discovered hyperglycemia should be monitored closely (21). The frequency and method of monitoring and the schedule of the blood glucose checks will depend on the nutritional intake, patient treatment, and insulin schedule, as well as the ability of the individual to self-manage their diabetes (225). There is some controversy regarding the best method to monitor blood glucose. However,considering the convenience and wide availability of capillary point of care (POC) testing, we suggest this as the bestapproach if done with a monitoring device that has demonstrated accuracy (226-228). When using POC blood glucose meters, it is important to keep several things in mind. In particular, overall clinical conditions that might affect the POC value, such as hemoglobin level, perfusion, and medications, as well as the policy of the health care organization in guiding the patient and the staff on the use of POC devices or newer technologies.

 

Bedside point-of-care (POC) capillary glucose testing is usually ordered before meals and bedtime to assess glycemic control and adjust insulin therapy in the hospital (19; 228). However, this approach has been shown to fail to detect hypoglycemia, particularly nocturnal and asymptomatic hypoglycemia, which is a common scenario in the hospital setting (229; 230).

 

Continuous glucose monitoring (CGM) has increased over the last few years, helping to improve glycemic management in the ICU. The use of this technology was accelerated during the COVID pandemic, where the use of CGM meant that close contact with sick individuals was avoided using remote sensing (231-234). The use of CGM is questioned, with the accuracy of readings when dealing with hypoglycemia or in the operating room (235; 236). However, in general, most studies have been associated with overall benefit (234; 237; 238).

 

CGM is reliable compared to point-of-care testing and laboratory values in the inpatient setting. It is currently being evaluated for managing ICU and general ward patients (159; 185; 235; 239-242). Studies have shown that CGM offers advantages over intermittent capillary monitoring in the ICU. CGM can help identify and prevent severe hyperglycemia and hypoglycemia by allowing for more rapid and accurate adjustments to insulin infusions compared to capillary blood glucose testing. Research has also demonstrated that CGM is better at detecting hypoglycemia, predominantly asymptomatic and nocturnal hypoglycemia, than capillary glucose testing (243; 244). Additionally, CGM is as safe and effective as standard care in hospitalized patients and can lead to a significant decrease in recurrent hypoglycemia events compared with standard point-of-care testing (243; 245). Regulatory approval for CGM use in hospitals is still pending, but consensus guidelines suggest that the use of CGM in the hospital setting has the potential to provide a better glycemic assessment than capillary glucose testing (Walia et al.; other, Endo Soc Guidelines).  Furthermore, advanced technology in guiding insulin therapy using machine learning and artificial intelligence is being integrated more frequently into diabetes care (246). A proof-of-concept trial in patients with type 2 diabetes evaluated the efficacy and safety of a model-based reinforcement learning framework in titrating insulin dosing. After applying the intervention, the mean daily BG was lower by approximately 56 mg/dl (3 mmol/l) with no severe hypo- or hyperglycemia (247).

 

The American Diabetes Association (ADA) and UK JBDS recently recommended that people with diabetes who use a personal continuous glucose monitoring (CGM) device should be allowed to continue during hospitalization (48; 159; 248). Both organizations also recommend that confirmatory point-of-care (POC) glucose measurements be used for insulin dosing decisions, hypoglycemia assessment, and treatment.

 

A recent survey of inpatient teams across the UK showed significant variations in accessing and using technologies (249). These included networked glucose and ketone meters, and wearable diabetes technologies such as CGM, pumps, or closed loop systems. While almost two-thirds of respondents agreed that technology would help prevent hypoglycemia, there was a wide variety of specialist diabetes nursing or medical staff support available to help non-specialists, particularly on weekends or outside of regular working hours (249).

 

Medical Nutrition Therapy (MNT) in Hospitalized Patients with Diabetes

 

Medical nutrition therapy (MNT) is a key component of the comprehensive management of diabetes and hyperglycemia in the inpatient setting. Maintaining adequate nutrition is essential for glycemic control and to meet adequate caloric demands. Caloric demand in acute illness will differ from that in the outpatient setting. Achieving the proper nutritionalbalance in the inpatient setting is challenging. Anyone admitted to the hospital with diabetes or hyperglycemia should beassessed to determine the need for a modified diet to meet caloric demands.

 

The general approach to addressing MNT in the inpatient setting is usually based on expert opinions and patients' needs. Limited data exist regarding the best approach or method to achieve the ideal caloric supply. To determine the best approach, method, and caloric needs of their patients, providers should work closely with the nutrition professional.

 

All patients with diabetes or hyperglycemia should receive an individualized assessment. Most patients will generallyreceive adequate caloric needs with 3 discrete meals daily. Further, the metabolic need for patients with diabetes isusually provided by 25 to 35 calories/kg, whereas some critically ill patients might require less than 15 to 25 calories/kg per day (250; 251). A consistent carbohydrate meal-planning system might help to facilitate glycemic control and insulin dosing in the inpatient setting. Most patients require 1,500-2000 calories daily with 12-15 grams of carbohydrates per meal (19). Ideally, the carbohydrates should come from low glycemic index foods such as whole grains and vegetables.

 

Those individuals unable to achieve these goals should be evaluated to determine the need for enteral or parenteral nutrition. Enteral nutrition is the second-best option after oral nutrition and should be preferred over parenteral nutrition in hospitalized individuals (252-254). There are several advantages of enteral feeding versus parenteral feeding, including low cost, low risk of complications, a physiologic route, less risk for gastric mucosa atrophy, and lower risk of infectiousand thrombotic complications compared with the latter form of therapy (252-254). The benefit of parental nutrition has been documented in critically ill patients. However, some research has shown a detrimental effect on patients withdiabetes and hyperglycemia. Parental nutrition should be considered only in patients who cannot receive enteral nutritionand should be coordinated with the institution’s parenteral nutrition team. There has been guidance published in the surgical population on peri-operative nutrition, but the recommendations for people with diabetes is lacking because the literature remains scanty (251). A recent UK survey of diabetes teams showed no consensus on enteral feeding regimens (253). For those tube-fed, there were 3 main regimens:  continuous 24-hour feeding, a single feed with one break in 24 hours, or multiple feeds in 24 hours. In addition, there were multiple insulin regimens used: premixed insulin, isophane insulin, analog basal insulin, variable rate intravenous insulin, or basal-bolus insulin. None of these provided adequate glycemic control (253).

 

Enteral and parenteral nutrition  can prevent the effects of starvation and malnutrition (252). Enteral nutrition over parenteral nutrition is preferred whenever possible due to a lower risk of infectious and thrombotic complications (254-256). Standard enteral formulas reflect the reference values for macro- and micronutrients for a healthy population and contain 1-2 cal/ml. Most standard formulas contain whole protein, lipids in the form of long-chain triglycerides, andcarbohydrates. Standard diabetes-specific formulas provide low amounts of lipids (30% of total calories) combined with a high carbohydrate (257) content (55–60% of total calories); however, newer “diabetic” formulas have replaced part of carbohydrates with monounsaturated fatty acids (up to 35% of total calories) and also include 10-15 g/l dietary fiber andup to 30% fructose (257; 258).

 

“Diabetic” enteral formulas containing low-carbohydrate high–monounsaturated fatty acid (LCHM) are preferable to standard high-carbohydrate formulas in hospitalized patients with type 1 and type 2 diabetes (257; 258). In a meta-analysis of studies comparing relatively newer enteral LCHM formulas with older formulations, the postprandial rise inblood glucose was reduced by 18- 29 mg/dl [1.0-1.6 mmol/l] with the newer formulations (258). Table 4 depicts the composition of standard and diabetic-specific enteral formulas commonly used in hospitalized patients.

 

Table 4. Composition of Standard and Diabetic Specific Enteral Formulas Commonly Usedin Hospitalized Patients in the USA

 

Calories(kcal/mL)

Carbohydrate(g/l)

Fat (g/l)

Protein (g/l)

Manufacture

Standard formula

 

Jevity® 1.0 Cal

1.0

140

35

40

Abbott Nutrition

Nutren® 1.0

1.0

109

27

70

Nestle Nutrition

Osmolite® 1.2 Cal

1.2

158

39

56

Abbott Nutrition

Jevity® 1.2

1.2

169

39

56

Nestle Nutrition

Fibersource® HN

1.2

164

40

54

Nestle Nutrition

Isosource® 1.5 Cal

1.5

176

60

68

Nestle Nutrition

Jevity® 1.5

1.5

216

50

64

Nestle Nutrition

Diabetes specific formula

Glucerna® 1.0 Cal

1.0

75

54

50

Abbott Nutrition

Nutren® Glytrol®

1.0

100

48

45

Nestle Nutrition

Glucerna® 1.2 Cal

1.2

114

60

60

Abbott Nutrition

Diabetisource® AC

1.2

100

59

60

Nestle Nutrition

Glucerna® 1.5 Cal

1.5

133

75

83

Abbott Nutrition

             

The UK Joint British Diabetes Societies has updated its guidelines for the management of diabetes in enterally fed people (259).

 

Corticosteroid Therapy – Impact on Blood Glucose

 

Steroid use in hospitalized patients is common. A single-center cross-sectional study showed that 12.8% of all the people in the hospital were on glucocorticoids (260). Steroids may be administered by various regimes and at variable doses. A single daily dose of steroid (e.g., prednisolone/prednisone) in the morning may be the most standard mode ofadministration (205; 260-262). Limbachia et al. showed that, in susceptible individuals, steroid use will often result in arise in blood glucose by late morning that continues through to the evening (263). Overnight, the blood glucose generallyfalls back to baseline levels by the following day. They also showed the differential effects between different steroid types, with oral or IV dexamethasone or methylprednisolone leading to higher glucose excursions than prednisolone or hydrocortisone (262). Thus, treatment should be tailored to treating the hyperglycemia while avoiding nocturnal and earlymorning hypoglycemia. Multiple daily doses of steroid, be it intravenous hydrocortisone or oral dexamethasone, can cause a hyperglycemic effect throughout the 24-hour period. It may be, however, that a twice-daily premixed or basal-bolus regimen may need to be started if oral medication or once-daily insulin proves insufficient to control hyperglycemia (205). Close attention will therefore need to be paid to blood glucose monitoring, and early intervention may benecessary.

 

Glucose levels in most individuals can be predicted to rise approximately 4 to 8 hours following the administration of once-daily oral steroids and sooner following the administration of intravenous steroids. Again, capillary blood glucose monitoring is paramount to guide appropriate therapeutic interventions. Conversely, glucose levels may improve to pre-steroid levels 24 hours after intravenous steroids are discontinued. When oral steroids are weaned down, the glucose levels may decline in a dose-dependent fashion, but this may not occur, particularly in those with pre-existing undiagnosed diabetes.

 

At the commencement of steroid therapy, or for those already on a supraphysiological dose of corticosteroid, capillary blood glucose testing should occur before meals and at bedtime, in particular before lunch or evening meal, when thehyperglycemic effects of a morning dose of steroid are likely to be greatest (205; 262).

 

Subcutaneous insulin using a basal or multiple daily injection regimen will likely be the most appropriate choice for most patients to achieve glycemic control in the event of hyperglycemia. While the UK has advocated for short-acting sulfonylureas (205), the morning administration of basal human insulin may closely fit the glucose excursion induced by a single morning dose of oral steroid. Basal analog insulin may be appropriate if hyperglycemia is present for more prolonged periods. However, if long-acting insulin analogs are used in this context, care should be taken to identify and protect against hypoglycemia overnight and in the early morning. Subsequent titration of the insulin dose may be required to maintain glucose control in the face of increasing or decreasing the steroid dose.

 

When a patient is discharged from the hospital on steroid therapy, a clear strategy for managing hyperglycemia or potential hyperglycemia and the titration of treatment to address the hyperglycemia should be communicated to the community diabetes team and primary care team. Patients who commenced on steroids as inpatients and were discharged after a short stay with the intention of continuing high-dose steroids should receive standard diabetes education, encompassing the risks associated with hyperglycemia and hypoglycemia.

 

Closed Loop Technology

 

Several organizations have recommended that people who are well enough to do so should continue to use their insulin pumps in hospitals (108; 109; 240; 264). However, only a few recent studies have reported using closed-loop systems, also referred to as the artificial pancreas or automated insulin delivery systems, in hospitalized inpatients. Small randomized trials have reported good efficacy with improved time in target and lower mean daily blood glucose without an increased rate of hypoglycemia in the ICU (265-267) and non-ICU settings (268-271). However, some of these studies were done in those with type 2 diabetes (268; 270). In one non-ICU study, the time in the target range between 100-180 mg/dl (5.6-10.0 mmol/l) was reported as 59.8% in patients using the closed-loop technology compared to 38.1% with standard subcutaneous insulin regimen (269).

 

Similarly, a closed-loop study in patients receiving nutritional support also reported higher time in target glucose (68% vs 36.4%) and lower mean glucose values (153 vs 205 mg/dl [8.5-11.4 mmol/l]) compared to a standard insulin regimen (270). As with the use of CGM in the hospital, treatment with artificial pancreas is still experimental, and larger studies are needed to prove its safety and efficacy in ICU and non-ICU settings. Further challenges lie ahead because of the unfamiliarity of these systems, with non-specialist staff the primary carers for people with diabetes.

 

The ADA has recommended that insulin pumps or automated insulin delivery (closed-loop) systems be continued for hospitalized individuals with diabetes when clinically appropriate. Confirmatory POC blood glucose measurements should be used for insulin-dosing decisions and for assessing and treating hypoglycemia. However, this depends on the availability of required supplies and resources, proper training, ongoing competency assessments, and the implementation of institutional diabetes technology protocols (48).

 

As with the CGM, those who are well and can self-manage can look after their devices and diabetes. However, in those who are unwell or incapacitated, the systems must be disengaged from automatic and set to ‘manual’ mode to allow the diabetes teams to help manage the diabetes. The systems may not also be able to cope with the acute changes that occur in the hospital, including (but not limited to) change in oral carbohydrate intake, the use of glucocorticoids or other medications inducing insulin resistance; peri-operative use, nausea and vomiting; enteral or parenteral nutrition. Once again, the use of ‘manual’ mode is recommended in these situations, and the diabetes is managed in conjunction with the specialist diabetes team.

 

REFERENCES

 

  1. IDF Diabetes Atlas 2021 [article online], 2021. Available from https://diabetesatlas.org/.
  2. National Diabetes Statistics Report - 2024 [article online], 2024. Available from https://www.cdc.gov/diabetes/php/data-research/index.html.
  3. Koyama AK, Cheng YJ, Brinks R, Xie, Gregg EW, Hoyer A, Pavkov ME, Imperatore G. Trends in lifetime risk and years of potential life lost from diabetes in the United States, 1997–2018. PLOS ONE 2022;17:e0268805
  4. Pournaras DJ, Photi ES, Barnett N, Challand CP, Chatzizacharias NA, Dlamini NP, Doulias T, Foley A, Hernon J, Kumar B, Martin J, Nunney I, Panagiotopoulou I, Sengupta N, Shivakumar O, Sinclair P, Stather P, Than MM, Wells AC, Xanthis A, Dhatariya K. Assessing the quality of primary care referrals to surgery of patients with diabetes in the East of England: A multi-centre cross-sectional cohort study. International Journal of Clinical Practice 2017;71:e12971
  5. Jiang HJ, Stryer D, Friedman B, Andrews R. Multiple hospitalizations for patients with diabetes. Diabetes Care 2003;26:1421-1426
  6. Donnan PT, Leese GP, Morris AD. Hospitalizations for people with type 1 and type 2 diabetes compared with the nondiabetic population of Tayside, Scotland: a retrospective cohort study of resource use. Diabetes Care 2000;23:1774-1779
  7. Harding JL, Benoit SR, Gregg EW, Pavkov ME, Perreault L. Trends in rates of infections requiring hospitalization among adults with versus without diabetes in the U.S., 2000 - 2015. Diabetes Care 2020;43:106-116
  8. Umpierrez GE, Isaacs SD, Bazargan N, You X, Thaler LM, Kitabchi AE. Hyperglycemia: An independent marker of in-hospital mortality in patients with undiagnosed diabetes. Journal of Clinical Endocrinology Metabolism 2002;87:978-982
  9. National Diabetes Statistics Report 2022 [article online], 2022. Available from https://www.cdc.gov/diabetes/data/statistics-report/index.html.
  10. Zhang Y, McKeever Bullard K, Imperatore G, Holliday CS, Benoit SR. Proportions and trends of adult hospitalizations with diabetes, United States, 2000–2018. Diabetes Research and Clinical Practice 2022;187:109862
  11. National Diabetes Inpatient Audit (NaDIA) - 2019 [article online], 2020. Available from https://digital.nhs.uk/data-and-information/publications/statistical/national-diabetes-inpatient-audit/2019.
  12. Sampson MJ, Dozio N, Ferguson B, Dhatariya K. Total and excess bed occupancy by age, speciality and insulin use for nearly one million diabetes patients discharged from all English Acute Hospitals. Diabetes Research & Clinical Practice 2007;77:92-98
  13. American Diabetes Association. Economic costs of diabetes in the U.S. in 2017. Diabetes Care 2018;41:917-928
  14. Parker ED, Lin J, Mahoney T, Ume N, Yang G, Gabbay RA, ElSayed NA, Bannuru RR. Economic costs of diabetes in the U.S. in 2022. Diabetes Care 2023;47:26-43
  15. Friel KM, Gillespie P, Coates V, McCauley C, McCann M, O’Kane M, McGuigan K, Khamis A, Manktelow M. Estimating and examining the costs of inpatient diabetes care in an Irish Public Hospital. Diabetic Medicine 2022;39:e14753
  16. Bommer C, Heesemann E, Sagalova V, Manne-Goehler J, Atun R, Barnighausen T, Vollmer S. The global economic burden of diabetes in adults aged 20-79 years: a cost-of-illness study. Lancet Diabetes and Endocrinology 2017;5:423-430
  17. Bommer C, Sagalova V, Heesemann E, Manne-Goehler J, Atun R, Barnighausen T, Davies J, Vollmer S. Global economic burden of diabetes in adults: Projections from 2015 to 2030. Diabetes Care 2018;41:963-970
  18. Nolan JJ, O'Halloran D, McKenna TJ, Firth R, Redmond S. The cost of treating type 2 diabetes (CODEIRE). Irish Medical Journal 2006;99:307-310
  19. Umpierrez GE, Hellman R, Korytkowski MT, Kosiborod M, Maynard GA, Montori VM, Seley JJ, Van den Berghe G. Management of hyperglycemia in hospitalized patients in non-critical care setting: An Endocrine Society clinical practice guideline. Journal of Clinical Endocrinology Metabolism 2012;97:16-38
  20. Moghissi ES, Korytkowski MT, Dinardo MM, Hellman R, Hirsch IB, Inzucchi S, Ismail-Beigi F, Kirkman MS, Umpierrez GE. American Association of Clinical Endocrinologists and American Diabetes Association consensus statement on inpatient glycemic control. Diabetes Care 2009;32:1119-1131
  21. Dhatariya K, James J, Kong M-F, Berrington R. Diabetes at the front door. A guideline for dealing with glucose related emergencies at the time of acute hospital admission from the Joint British Diabetes Society (JBDS) for Inpatient Care Group. Diabetic Medicine 2020;37:1578-1589
  22. Dhatariya K, Mustafa OG, Rayman G. Safe care for people with diabetes in hospital. Clinical Medicine 2020;20:21-27
  23. A good inpatient diabetes service [article online], 2019. Available from https://abcd.care/resource/jbds-14-good-inpatient-diabetes-service.
  24. NICE SUGAR investigators. Intensive versus conventional glucose control in critically ill patients. New England Journal of Medicine 2009;360:1283-1297
  25. Hulkower RD, Pollack RM, Zonszein J. Understanding hypoglycemia in hospitalized patients. Diabetes Management (Lond) 2014;4:165-176
  26. McDonnell ME, Umpierrez GE. Insulin therapy for the management of hyperglycemia in hospitalized patients. Endocrinology & Metabolism Clinics of North America 2012;41:175-201
  27. Qaseem A, Humphrey LL, Chou R, Snow V, Shekelle P, Physicians for the Clinical Guidelines Committee of the American College of Physicians. Use of intensive insulin therapy for the management of glycemic control in hospitalized patients: A clinical practice guideline from the American College of Physicians. Annals of Internal Medicine 2011;154:260-267
  28. Cook CB, Kongable GL, Potter DJ, Abad VJ, Leija DE, Anderson M. Inpatient glucose control: a glycemic survey of 126 U.S. hospitals. Journal of Hospital Medicine 2009;4:E7-E14
  29. NICE SUGAR investigators. Hypoglycemia and risk of death in critically ill patients. New England Journal of Medicine 2012;367:1108-1118
  30. Turchin A, Matheny ME, Shubina M, Scanlon JV, Greenwood B, Pendergrass ML. Hypoglycemia and clinical outcomes in patients with diabetes hospitalized in the general ward. Diabetes Care 2009;32:1153-1157
  31. Yamada T, Shojima N, Noma H, Yamauchi T, Kadowaki T. Glycemic control, mortality, and hypoglycemia in critically ill patients: a systematic review and network meta-analysis of randomized controlled trials. Intensive Care Medicine 2017;43:1-15
  32. Levy N, Hall GM. National guidance contributes to the high incidence of inpatient hypoglycaemia. Diabetic Medicine 2019;36:120-121
  33. National Diabetes Inpatient Audit - Harms 2020 England [article online], 2020. Available from https://digital.nhs.uk/data-and-information/publications/statistical/national-diabetes-inpatient-audit---harms/national-diabetes-inpatient-audit---harms-2020.
  34. National Diabetes Inpatient Safety Audit (NDISA) 2018-2021 [article online], 2022. Available from https://digital.nhs.uk/data-and-information/publications/statistical/national-diabetes-inpatient-safety-audit-ndisa/ndisa-2018-2021.
  35. Handelsman Y, Bloomgarden ZT, Grunberger G, Umpierrez G, Zimmerman RS, Bailey TS, Blonde L, Bray GA, Cohen AJ, Dagogo-Jack S, Davidson JA, Einhorn D, Ganda AP, Garber AJ, Garvey WT, Henry RR, Hirsch IB, Horton ES, Hurley DL, Jellinger PS, Jovanovic L, Lebovitz HE, LeRoith D, Levy P, McGill JB, Mechanick JI, Mestman JH, Moghissi ES, Orzeck EA, Pessah-Pollack R, Rosenblit PD, Vinik AI, Wyne K, Zangeneh F. American Association of Clinical Endocrinologists and American College of Endocrinology - clinical practice guidelines for developing a diabetes mellitus comprehensive care plan - 2015. Endocrine Practice 2015;21:1-87
  36. Jacobi J, Bircher N, Krinsley J, Agus M, Braithwaite SS, Duutschman C, Freire AX, Geehan D, Kohl B, Nasraway SA, Rigby M, Sands K, Schallom L, Taylor B, Umpierrez G, Mazuski J, Schunemann H. Guidelines for the use of an insulin infusion for the management of hyperglycemia in critically ill patients. Critical Care Medicine 2012;40:3251-3276
  37. Kitabchi AE, Umpierrez GE, Miles JM, Fisher JN. Hyperglycemic crises in adult patients with diabetes. Diabetes Care 2009;32:1335-1343
  38. 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. Diabetes care in the hospital: Standards of care in diabetes—2023. Diabetes Care 2023;46:S267-S278
  39. Swanson C, Potter D, Kongable G, Cook C. Update on inpatient glycemic control in hospitals in the United States. Endocrine Practice 2017;17:853-861
  40. Schmeltz LR, DeSantis AJ, Thiyagarajan V, Schmidt K, Shea-Mahler E, Johnson D, Henske J, McCarthy PM, Gleason TG, McGee EC, Molitch ME. Reduction of surgical mortality and morbidity in diabetic patients undergoing cardiac surgery with a combined intravenous and subcutaneous insulin glucose management strategy. Diabetes Care 2007;30:823-828
  41. Carpenter DL, Gregg SR, Xu K, Buchman TG, Coopersmith CM. Prevalence and impact of unknown diabetes in the ICU. Critical Care Medicine 2015;43:e541-e550
  42. Desgrouas M, Demiselle J, Stiel L, Brunot V, Marnai R, Sarfati S, Fiancette M, Lambiotte F, Thille AW, Leloup M, Clerc S, Beuret P, Bourion A-A, Daum J, Malhomme R, Ravan R, Sauneuf B, Rigaud J-P, Dequin P-F, Boulain T. Insulin therapy and blood glucose management in critically ill patients: a 1-day cross-sectional observational study in 69 French intensive care units. Annals of Intensive Care 2023;13:53
  43. Van Ackerbroeck S, Schepens T, Janssens K, Jorens PG, Verbrugge W, Collet S, Van Hoof V, Van Gaal L, De Block C. Incidence and predisposing factors for the development of disturbed glucose metabolism and DIabetes mellitus AFter Intensive Care admission: the DIAFIC study. Critical Care 2015;19:355
  44. Kar P, Plummer MP, Ali Abdelhamid Y, Giersch EJ, Summers MJ, Weinel LM, Finnis ME, Phillips LK, Jones KL, Horowitz M, Deane AM. Incident diabetes in survivors of critical illness and mechanisms underlying persistent glucose intolerance: A prospective cohort study. Critical Care Medicine 2019;47:e103-e111
  45. Gomez AM, Imitola Madero A, Henao Carrillo DC, Rondon M, Munoz OM, Robledo MA, Rebolledo M, Garcia Jaramillo M, Leon Vargas F, Umpierrez G. Hypoglycemia incidence and factors associated in a cohort of patients with type 2 diabetes hospitalized in general ward treated with basal bolus insulin regimen assessed by continuous glucose monitoring. Journal of Diabetes Science and Technology 2020;14:233-239
  46. Greci LS, Kailasam M, Malkani S, Katz DL, Hulinsky I, Ahmadi R, Nawaz H. Utility of HbA1c levels for diabetes case finding in hospitalized patients with hyperglycemia. Diabetes Care 2003;26:1064-1068
  47. Mazurek JA, Hailpern SM, Goring T, Nordin C. Prevalence of hemoglobin A1c greater than 6.5% and 7.0% among hospitalized patients without known diagnosis of diabetes at an urban inner city hospital. Journal of Clinical Endocrinology & Metabolism 2010;95:1344-1348
  48. American Diabetes Association. 16. Diabetes care in the hospital: Standards of Care in Diabetes–2025. Diabetes Care 2025;48 (Suppl 1).
  49. Dashora U, Flanagan D, Rayman G, Mustafa OG, Walden E, Dhatariya K, The Joint British Diabetes Societies for Inpatient Care. Optimal staffing for a good quality inpatient diabetes service. Diabetic Medicine 2023;40:e15151
  50. Cryer PE. Hypoglycemia, functional brain failure, and brain death. Journal of Clinical Investigation 2014;117:868-870
  51. Amiel SA, Aschner P, Childs B, Cryer PE, de Galan BE, Frier BM, Gonder-Frederick L, Heller SR, Jones T, Khunti K, Leiter LA, Luo Y, McCrimmon RJ, Pedersen-bjergaard U, Seaquist ER, Zoungas S. Hypoglycaemia, cardiovascular disease, and mortality in diabetes: epidemiology, pathogenesis, and management. Lancet Diabetes & Endocrinology 2019;7:385-396
  52. Corssmit EP, Romijn JA, Sauerwein HP. Regulation of glucose production with special attention to nonclassical regulatory mechanisms: A review. Metabolism 2014;50:742-755
  53. Boden G. Gluconeogenesis and glycogenolysis in health and diabetes. Journal of Investigative Medicine 2004;52:375-378
  54. Gerich JE, Meyer C, Woerle HJ, Stumvoll M. Renal gluconeogenesis. Diabetes Care 2001;24:382-391
  55. van de Werve G, Jeanrenaud B. Liver glycogen metabolism: An overview. Diabetes/Metabolism Reviews 1987;3:47-78
  56. Gerich JE, Lorenzi M, Bier DM, Tsalikian E, Schneider V, Karam JH, Forsham PH. Effects of physiologic levels of glucagon and growth hormone on human carbohydrate and lipid metabolism. Studies involving administration of exogenous hormone during suppression of endogenous hormone secretion with somatostatin. Journal of Clinical Investigation 1976;57:875-884
  57. Rizza RA, Mandarino LJ, Gerich JE. Dose-response characteristics for effects of insulin on production and utilization of glucose in man. American Journal of Physiology - Endocrinology & Metabolism 1981;240:E630-E639
  58. Dhatariya KK, Glaser NS, Codner E, Umpierrez GE. Diabetic ketoacidosis. Nature Reviews Disease Primers 2020;6:40
  59. Dungan KM, Braithwaite SS, Preiser J-C. Stress hyperglycaemia. Lancet 2009;373:1798-1807
  60. McCowen KC, Malhotra A, Bistrian BR. Stress-induced hyperglycemia. Critical Care Clinics 2001;17:107-124
  61. Stentz FB, Umpierrez GE, Cuervo R, Kitabchi AE. Proinflammatory cytokines, markers of cardiovascular risks, oxidative stress, and lipid peroxidation in patients with hyperglycemic crises. Diabetes 2004;53:2079-2086
  62. Chaudhuri A, Umpierrez GE. Oxidative stress and inflammation in hyperglycemic crises and resolution with insulin: implications for the acute and chronic complications of hyperglycemia. Journal of Diabetes and its Complications 2012;26:257-258
  63. Reyes-Umpierrez D, Davis G, Cardona S, Pasquel FJ, Peng L, Jacobs S, Vellanki P, Fayfman M, Haw S, Halkos M, Guyton RA, Thourani VH, Umpierrez GE. Inflammation and oxidative stress in cardiac surgery patients treated to intensive versus conservative glucose targets. Journal of Clinical Endocrinology & Metabolism 2016;102:309-315
  64. Esposito K, Nappo F, Marfella R, Giugliano G, Giugliano F, Ciotola M, Quagliaro L, Ceriello A, Giugliano D. Inflammatory cytokine concentrations are acutely increased by hyperglycemia in humans: Role of oxidative stress. Circulation 2002;106:2067-2072
  65. Fan J, Li YH, Wojnar MM, Lang CH. Endotoxin-induced alterations in insulin-stimulated phosphorylation of insulin receptor, IRS-1, and MAP kinase in skeletal muscle. Shock 1996;6:164-170
  66. Evans NR, Dhatariya KK. Assessing the relationship between admission glucose levels, subsequent length of hospital stay, readmission and mortality. Clinical Medicine 2012;12:137-139
  67. Haddadin F, Clark A, Evans N, Dhatariya K. Admission blood glucose helps predict 1 year, but not 2 years, mortality in an unselected cohort of acute general medical admissions. International Journal of Clinical Practice 2014;69:643-648
  68. Frisch A, Chandra P, Smiley D, Peng L, Rizzo M, Gatcliffe C, Hudson M, Mendoza J, Johnson R, Lin E, Umpierrez GE. Prevalence and clinical outcome of hyperglycemia in the perioperative period in noncardiac surgery. Diabetes Care 2010;33:1783-1788
  69. Kotagal M, Symons RG, Hirsch IB, Umpierrez GE, Dellinger EP, Farrokhi ET, Flum DR. Perioperative hyperglycemia and risk of adverse events among patients with and without diabetes. Annals of Surgery 2016;261:97-103
  70. Umpierrez G, Cardona S, Pasquel F, Jacobs S, Peng L, Unigwe M, Newton CA, Smiley-Byrd D, Vellanki P, Halkos M, Puskas JD, Guyton RA, Thourani VH. Randomized controlled trial of intensive versus conservative glucose control in patients undergoing coronary artery bypass graft surgery: GLUCO-CABG trial. Diabetes Care 2015;38:1665-1672
  71. Umpierrez GE, Smiley D, Jacobs S, Peng L, Temponi A, Mulligan P, Umpierrez D, Newton C, Olson D, Rizzo M. Randomized study of basal-bolus insulin therapy in the inpatient management of patients with type 2 diabetes undergoing general surgery (RABBIT 2 Surgery). Diabetes Care 2011;34:256-261
  72. Krinsley JS. Association between hyperglycemia and increased hospital mortality in a heterogeneous population of critically ill patients. Mayo Clinic Proceedings 2003;78:1471-1478
  73. Furnary AP, Zerr KJ, Grunkemeier GL, Starr A. Continuous intravenous insulin infusion reduces the incidence of deep sternal wound infection in diabetic patients after cardiac surgical procedures. Annals of Thoracic Surgery 1999;67:352-362
  74. Montori VM, Bistrian BR, McMahon MM. Hyperglycemia in acutely ill patients. JAMA 2002;288:2167-2169
  75. Adie SK, Ketcham SW, Marshall VD, Farina N, Sukul D. The association of glucose control on in-hospital mortality in the cardiac intensive care unit. Journal of Diabetes and its Complications 2023;37:108453
  76. Levy N, Dhatariya K. Pre-operative optimisation of the surgical patient with diagnosed and undiagnosed diabetes: a practical review. Anaesthesia 2019;74:58-66
  77. Falciglia M, Freyberg RW, Almenoff PL, D'Alessio DA, Render ML. Hyperglycemia-related mortality in critically ill patients varies with admission diagnosis. Critical Care Medicine 2009;37:3001-3009
  78. Bruno A, Gregori D, Caropreso A, Lazzarato F, Petrinco M, Pagano E. Normal glucose values are associated with a lower risk of mortality in hospitalized patients. Diabetes Care 2008;31:2209-2210
  79. Critchley JA, Carey IM, Harris T, DeWilde S, Hosking FJ, Cook DG. Glycemic control and risk of infections among people with type 1 or type 2 diabetes in a large primary care cohort study. Diabetes Care 2018;41:2127-2135
  80. Blaha J, Mraz M, Kopecky P, Stritesky M, Lips M, Matias M, Kunstyr J, Porizka M, Kotulak T, Kolnikova I, Simanovska B, Zakharchenko M, Rulisek J, Sachl R, Anyz J, Novak D, Linder J, Hovorka R, Svacina S, Haluzik M. Perioperative tight glucose control reduces postoperative adverse events in nondiabetic cardiac surgery patients. Journal of Clinical Endocrinology & Metabolism 2015;100:3081-3089
  81. Barmanray RD, Cheuk N, Fourlanos S, Greenberg PB, Colman PG, Worth LJ. In-hospital hyperglycemia but not diabetes mellitus alone is associated with increased in-hospital mortality in community-acquired pneumonia (CAP): a systematic review and meta-analysis of observational studies prior to COVID-19. BMJ Open Diabetes Research & Care 2022;10:e002880
  82. Shiffermiller J, Anderson M, Thompson R. Postoperative length of stay in patients with stress hyperglycemia compared to patients with diabetic hyperglycemia: A retrospective cohort study. Journal of Diabetes Science and Technology 2024;18:556-561
  83. Jones CE, Graham LA, Morris MS, Richman JS, Hollis RH, Wahl TS, Copeland LA, Burns EA, Itani KM, Hawn MT. Association between preoperative hemoglobin A1c levels, postoperative hyperglycemia, and readmissions following gastrointestinal surgery. JAMA Surgery 2017;152:1031-1038
  84. Rains JL, Jain SK. Oxidative stress, insulin signaling, and diabetes. Free Radical Biology and Medicine 2011;50:567-575
  85. Meza CA, La Favor JD, Kim DH, Hickner RC. Endothelial dysfunction: Is there a hyperglycemia-induced imbalance of NOX and NOS? International  Journal of Molecular Science 2019;20
  86. Furnary AP, Gao G, Grunkemeier GL, Wu Y, Zerr KJ, Bookin SO, Floten HS, Starr A. Continuous insulin infusion reduces mortality in patients with diabetes undergoing coronary artery bypass grafting. Journal of Thoracic & Cardiovascular Surgery 2003;125:1007-1021
  87. Krinsley JS, Maurer P, Holewinski S, Hayes R, McComsey D, Umpierrez GE, Nasraway SA. Glucose control, diabetes status, and mortality in critically ill patients. Mayo Clinic Proceedings 2017;92:1019-1029
  88. van Vught LA, Holman R, de Jong E, de Keizer NF, van der Poll T. Diabetes is not associated with increased 90-day mortality risk in critically ill patients with sepsis. Critical Care Medicine 2017;45:e1026-e1035
  89. Gunst J, Debaveye Y, Güiza F, Dubois J, De Bruyn A, Dauwe D, De Troy E, Casaer MP, De Vlieger G, Haghedooren R, Jacobs B, Meyfroidt G, Ingels C, Muller J, Vlasselaers D, Desmet L, Mebis L, Wouters PJ, Stessel B, Geebelen L, Vandenbrande J, Brands M, Gruyters I, Geerts E, De Pauw I, Vermassen J, Peperstraete H, Hoste E, De Waele JJ, Herck I, Depuydt P, Wilmer A, Hermans G, Benoit DD, Van den Berghe G. Tight blood-glucose control without early parenteral nutrition in the ICU. New England Journal of Medicine 2023;389:1180-1190
  90. Mehta G, Lo B, Memarpour M, Chan A. Impact of diabetes on inpatient length of stay in adult mental health services in a community hospital setting: A retrospective cohort study. Canadian Journal of Diabetes 2022;46:678-682
  91. McAlister FA, Majumdar SR, Blitz S, Rowe BH, Romney J, Marrie TJ. The relation between hyperglycemia and outcomes in 2,471 patients admitted to the hospital with community-acquired pneumonia. Diabetes Care 2005;28:810-815
  92. Baker EH, Janaway CH, Philips BJ, Brennan AL, Baines DL, Wood DM, Jones PW. Hyperglycaemia is associated with poor outcomes in patients admitted to hospital with acute exacerbations of chronic obstructive pulmonary disease. Thorax 2006;61:284-289
  93. Noordzij PG, Boersma E, Schreiner F, Kertai MD, Feringa HH, Dunkelgrun M, Bax JJ, Klein J, Poldermans D. Increased preoperative glucose levels are associated with perioperative mortality in patients undergoing noncardiac, nonvascular surgery. European Journal of Endocrinology 2007;156:137-142
  94. Martin ET, Kaye KS, Knott C, Nguyen H, Santarossa M, Evans R, Bertran E, Jaber L. Diabetes and risk of surgical site infection: A systematic review and meta-analysis. Infection Control & Hospital Epidemiology 2016;37:88-99
  95. Pomposelli JJ, Baxter JK, Babineau TJ, Pomfret EA, Driscoll DF, Forse RA, Bistrian BR. Early postoperative glucose control predicts nosocomial infection rate in diabetic patients. Journal of Parenteral and Enteral Nutrition 1998;22:77-81
  96. Ramos M, Khalpey Z, Lipsitz S, Steinberg J, Panizales MT, Zinner M, Rogers SO. Relationship of perioperative hyperglycemia and postoperative infections in patients who undergo general and vascular surgery. Annals of Surgery 2015;248:585-591
  97. Sadoskas D, Suder NC, Wukich DK. Perioperative glycemic control and the effect on surgical site infections in diabetic patients undergoing foot and ankle surgery. Foot and Ankle Specialist 2016;9:24-30
  98. Wallia A, Schmidt K, Oakes DJ, Pollack T, Welsh N, Kling-Colson S, Gupta S, Fulkerson C, Aleppo G, Parikh N, Levitsky J, Norvell JP, Rademaker A, Molitch ME. Glycemic control reduces infections in post-liver transplant patients: Results of a prospective, randomized study. Journal of Clinical Endocrinology & Metabolism 2017;102:451-459
  99. Sheen YJ, Huang CC, Huang SC, Hung MD, Lin CH, Lee IT, Lin SY, Sheu WH. Implimentation of an electronic dashboard with a remote management system to improve glycemic management among hospitalized adults. Endocrine Practice 2020;26:179-191
  100. Garg R, Schuman B, Bader A, Hurwitz S, Turchin A, Underwood P, Metzger C, Rein R, Lortie M. Effect of preoperative diabetes management on glycemic control and clinical outcomes after elective surgery. Annals of Surgery 2018;267:858-862
  101. Gregory NS, Seley JJ, Ukena J, Shah S, Fred MR, Dargar SK, Mauer E, Kim RJ. Decreased rates of inpatient hypoglycemia following implementation of an automated tool in the electronic medical record for identifying root causes. Journal of Diabetes Science and Technology 2018;12:63-68
  102. Pichardo-Lowden A, Umpierrez G, Lehman EB, Bolton MD, DeFlitch CJ, Chinchilli VM, Haidet PM. Clinical decision support to improve management of diabetes and dysglycemia in the hospital: a path to optimizing practice and outcomes. BMJ Open Diabetes Research & Care 2021;9:e001557
  103. Wang YY, Hu SF, Ying HM, Chen L, Li HL, Tian F, Zhou ZF. Postoperative tight glycemic control significantly reduces postoperative infection rates in patients undergoing surgery: A meta-analysis. BMC Endocrine Disorders 2018;18:42
  104. Vanhorebeek I, Gunst J, Van den Berghe G. Critical care management of stress-induced hyperglycemia. Current Diabetes Reports 2018;18:17
  105. Gunst J, De Bruyn A, Van den Berghe G. Glucose control in the ICU. Current Opinion in Anesthesiology 2019;32:156-162
  106. Dashora U, George S, Sampson M, Walden E. National guidelines have contributed to safer care for inpatients with diabetes. Diabetic Medicine 2019;36:124-126
  107. Honarmand K, Sirimaturos M, Hirshberg EL, Bircher NG, Agus MS, Carpenter DL, Downs CR, Farrington EA, Freire AX, Grow A, Irving SY, Krinsley JS, Lanspa MJ, Long MT, Nagpal D, Preiser JC, Srinivasan V, Umpierrez GE, Jacobi J. Society of Critical Care Medicine guidelines on glycemic control for critically ill children and adults 2024. Critical Care Medicine 2024;52:e161-e181
  108. Association AD. Diabetes care in the hospital: Standards of care in diabetes—2024. Diabetes Care 2023;47:S295-S306
  109. Korytkowski MT, Muniyappa R, Antinori-Lent K, Donihi AC, Drincic AT, Hirsch IB, Luger A, McDonnell ME, Murad MH, Nielsen C, Pegg C, Rushakoff RJ, Santesso N, Umpierrez GE. Management of hyperglycemia in hospitalized adult patients in non-critical care settings: An Endocrine Society clinical practice guideline. Journal of Clinical Endocrinology & Metabolism 2022;107:2101–2128
  110. Lazar HL, McDonnell M, Chipkin SR, Furnary AP, Engelman RM, Sadhu AR, Bridges CR, Haan CK, Svedjeholm R, Taegtmeyer H, Shemin RJ. The Society of Thoracic Surgeons Practice Guideline Series: Blood glucose management during adult cardiac surgery. Annals of Thoracic Surgery 2009;87:663-669
  111. Guidelines for the management of inpatient diabetes [article online], 2022. Available from https://abcd.care/jbds-ip.
  112. End of life diabetes care [article online], 2021. Available from https://www.diabetes.org.uk/professionals/position-statements-reports/diagnosis-ongoing-management-monitoring/end-of-life-care.
  113. Van den Berghe G, Wouters P, Weekers F, Verwaest C, Bruyninckx F, Schetz M, Vlasselaers D, Ferdinande P, Bouillon R, Lauwers P. Intensive insulin therapy in critically ill patients. New England Journal of Medicine 2001;345:1359-1367
  114. Van den Berghe G, Wilmer A, Hermans G, Meersseman W, Wouters PJ, Milants I, Van Wijngaerden E, Bobbaers H, Bouillon R. Intensive insulin therapy in the medical ICU. New England Journal of Medicine 2006;354:449-461
  115. Brunkhorst FM, Engel C, Bloos F, Meier-Hellmann A, Ragaller M, Weiler N, Moerer O, Gruendling M, Oppert M, Grond S, Olthoff D, Jaschinski U, John S, Rossaint R, Welte T, Schaefer M, Kern P, Kuhnt E, Kiehntopf M, Hartog C, Natanson C, Loeffler M, Reinhart K. Intensive insulin therapy and pentastarch resuscitation in severe sepsis. New England Journal of Medicine 2008;358:125-139
  116. De La Rosa G, Donado JH, Restrepo AH, Quintero AM, Gonzalez LG, Saldarriaga NE, Bedoya M, Toro JM, Velasquez JB, Valencia JC, Arango CM, Aleman PH, VAsquez EM, Chavarriaga JC, Yepes A, Pulido W, Cadavid CA. Strict glycaemic control in patients hospitalised in a mixed medical and surgical intensive care unit: a randomised clinical trial. Critical Care 2008;12:R120
  117. Preiser J-C, Brunkhorst F. Tight glucose control and hypoglycemia. Critical Care Medicine 2008;36:1391-1392
  118. Marik PE. Tight glycemic control in acutely ill patients: low evidence of benefit, high evidence of harm! Intensive Care Medicine 2016;42:1475-1477
  119. Umpierrez GE. Glucose control in the ICU. New England Journal of Medicine 2023;389:1234-1237
  120. Krinsley JS, Rule P, Pappy L, Ahmed A, Huley-Rodrigues C, Prevedello D, Preiser J-C. The interaction of acute and chronic glycemia on the relationship of hyperglycemia, hypoglycemia, and glucose variability to mortality in the critically ill. Critical Care Medicine 2020;48:1744-1751
  121. Malmberg KA, Efendic S, Ryden LE. Feasibility of insulin-glucose infusion in diabetic patients with acute myocardial infarction: A report from the multicenter trial: DIGAMI. Diabetes Care 1994;17:1007-1014
  122. Lazar HL, Chipkin SR, Fitzgerald CA, Bao Y, Cabral H, Apstein CS. Tight glycemic control in diabetic coronary artery bypass graft patients improves perioperative outcomes and decreases recurrent ischemic events. Circulation 2004;109:1497-1502
  123. Gandhi GY, Nuttall GA, Abel MD, Mullany CJ, Schaff HV, O'Brien PC, Johnson MG, Williams AR, Cutshall SM, Mundy LM, Rizza RA, McMahon MM. Intensive intraoperative insulin therapy versus conventional glucose management during cardiac surgery: A randomized trial. Annals of Internal Medicine 2007;146:233-243
  124. Preiser J-C, Devos P, Ruiz-Santana S, Melot C, Annane D, Groeneveld J, Iapichino G, Leverve XM, Nitenberg G, Singer P, Wernerman J, Joannidis M, Stecher A, Chiolero R. A prospective randomised multi-centre controlled trial on tight glucose control by intensive insulin therapy in adult intensive care units: the Glucontrol study. Intensive Care Medicine 2009;35:1738-1748
  125. Agus MS, Steil GM, Wypij D, Costello JM, Laussen PC, Langer M, Alexander JL, Scoppettuolo LA, Pigula FA, Charpie JR, Ohye RG, Gaies MG. Tight glycemic control versus standard care after pediatric cardiac surgery. New England Journal of Medicine 2012;367:1208-1219
  126. Agus MS, Wypij D, Hirshberg EL, Srinivasan V, Faustino EV, Luckett PM, Alexander JL, Asaro LA, Curley MA, Steil GM, Nadkarni VM. Tight glycemic control in critically ill children. New England Journal of Medicine 2017;376:729-741
  127. Macrae D, Grieve R, Allen E, Sadique Z, Morris K, Pappachan J, Parslow R, Tasker RC, Elbourne D. A randomized trial of hyperglycemic control in pediatric intensive care. New England Journal of Medicine 2014;370:107-118
  128. Kalfon P, Giraudeau B, Ichai C, Guerrini A, Brechot N, Cinotti R, Dequin P-F, Riu-Poulenc B, Montravers P, Annane D, Dupont H, Sorine M, Riou B. Tight computerized versus conventional glucose control in the ICU: a randomized controlled trial. Intensive Care Medicine 2014;40:171-181
  129. Cinotti R, Ichai C, Orban JC, Kalfon P, Feuillet F, Roquilly A, Riou B, Blanloeil Y, Asehnoune K, Rozec B. Effects of tight computerized glucose control on neurological outcome in severely brain injured patients: A multicenter sub-group analysis of the randomized-controlled open-label CGAO-REA study. Critical Care 2014;18:498
  130. Okabayashi T, Shima Y, Sumiyoshi T, Kozuki A, Tokumaru T, Iiyama T, Sugimoto T, Kobayashi M, Yokoyama M, Hanazaki K. Intensive versus intermediate glucose control in surgical intensive care unit patients. Diabetes Care 2014;37:1516
  131. Cardona S, Pasquel FJ, Fayfman M, Peng L, Jacobs S, Vellanki P, Weaver J, Halkos M, Guyton RA, Thourani VH, Umpierrez GE. Hospitalization costs and clinical outcomes in CABG patients treated with intensive insulin therapy. Journal of Diabetes and its Complications 2017;31:742-747
  132. Dhatariya K. Should inpatient hyperglycaemia be treated? British Medical Journal 2013;346:f134
  133. Phillips VL, Byrd AL, Adeel S, Peng L, Smiley DD, Umpierrez GE. A comparison of inpatient cost per day in general surgery patients with type 2 diabetes treated with basal-bolus versus sliding scale insulin regimens. PharmacoEconomics - Open 2017;1:109-115
  134. Barmanray RD, Kyi M, Colman PG, Rowan L, Raviskanthan M, Collins L, Donaldson L, Montalto S, Tsan J, Sun E, Le M, Worth LJ, Thomson B, Fourlanos S. The specialist treatment of inpatients: Caring for diabetes in surgery (STOIC-D Surgery) trial: A randomized controlled trial of early intervention with an electronic specialist-led model of diabetes care. Diabetes Care 2024;47:948-955
  135. Cryer PE, Axelrod L, Grossman AB, Heller SR, Montori VM, Seaquist ER, Service FJ. Evaluation and management of adult hypoglycemic disorders: An Endocrine Society clinical practice guideline. Journal of Clinical Endocrinology & Metabolism 2009;94:709-728
  136. Seaquist ER, Anderson J, Childs B, Cryer P, Dagogo-Jack S, Fish L, Heller SR, Rodriguez H, Rosenzweig J, Vigersky R. Hypoglycemia and diabetes: A report of a workgroup of the American Diabetes Association and The Endocrine Society. Diabetes Care 2013;36:1384-1395
  137. The hospital management of hypoglycaemia in adults with diabetes mellitus. Revised April 2021 [article online], 2021. Available from https://abcd.care/resource/current/jbds-01-hospital-management-hypoglycaemia-adults-diabetes-mellitus.
  138. Krikorian A, Ismail-Beigi F, Moghissi ES. Comparisons of different insulin infusion protocols: a review of recent literature. Current Opinion in Clinical Nutrition & Metabolic Care 2010;13:198-204
  139. Umpierrez GE, Smiley D, Zisman A, Prieto LM, Palacio A, Ceron M, Puig A, Mejia R. Randomized study of basal-bolus insulin therapy in the inpatient management of patients with type 2 diabetes (RABBIT 2 trial). Diabetes Care 2007;30:2181-2186
  140. Rajendran R, Kerry C, Rayman G, on behalf of the MAGIC group. Temporal patterns of hypoglycaemia and burden of sulfonylurea-related hypoglycaemia in UK hospitals: a retrospective multicentre audit of hospitalised patients with diabetes. BMJ Open 2014;4
  141. National Diabetes Inpatient Audit (NaDIA) - 2017 [article online], 2018. Available from https://digital.nhs.uk/data-and-information/publications/statistical/national-diabetes-inpatient-audit/national-diabetes-inpatient-audit-nadia-2017.
  142. Krinsley JS, Grover A. Severe hypoglycemia in critically ill patients: Risk factors and outcomes. Critical Care Medicine 2007;35:2262-2267
  143. Kagansky N, Levy S, Rimon E. Hypoglycemia as a predictor of mortality in hospitalized elderly patients. Archives of Internal Medicine 2003;163:1825-1829
  144. Lake A, Arthur A, Byrne C, Davenport K, Yamamoto JM, Murphy HR. The effect of hypoglycaemia during hospital admission on health-related outcomes for people with diabetes: a systematic review and meta-analysis. Diabetic Medicine 2019;36:1349-1359
  145. Dendy J, Chockalingam V, Tirumalasetty N, Dornelles A, Blonde L, Bolton P, Meadows R, Andrews S. Identifying risk factors for severe hypoglycemia in hospitalized patients with diabetes. Endocrine Practice 2014;20:1051-1056
  146. Smith WD, Winterstein AG, Johns T, Rosenberg E, Sauer BC. Causes of hyperglycemia and hypoglycemia in adult inpatients. American Journal of Health-System Pharmacy 2005;62:714-719
  147. Maynard G, Lee J, Phillips G, Fink E, Renvall M. Improved inpatient use of basal insulin, reduced hypoglycemia, and improved glycemic control: Effect of structured subcutaneous insulin orders and an insulin management algorithm. Journal of Hospital Medicine 2009;4:3-15
  148. Insulin safety. Reducing harm associated with the unsafe use of insulin products [article online], 2010. Available from https://www.centreformedicinesoptimisation.co.uk/new-insulin-safety-guidance-issued-to-reduce-wrong-dosages/#:~:text=Learning%20from%20previous%20incidents%20like,insulin%20administration%20times%3B%20and%20putting.
  149. Griesdale DE, de Souza RJ, van Dam RM, Heyland DK, Cook DJ, Malhotra A, Dhaliwal R, Henderson WR, Chittock DR, Finfer S, Talmor D. Intensive insulin therapy and mortality among critically ill patients: a meta-analysis including NICE-SUGAR study data. Canadian Medical Association Journal 2009;180:821-827
  150. Zoungas S, Patel A, Chalmers J, de Galan BE, Li Q, Billot L, Woodward M, Ninomiya T, Neal B, MacMahon S, Grobbee DE, Kengne AP, Marre M, Heller S. Severe hypoglycemia and risks of vascular events and death. New England Journal of Medicine 2010;363:1410-1418
  151. Wiener RS, Wiener DC, Larson RJ. Benefits and risks of tight glucose control in critically ill adults: A meta-analysis. JAMA 2008;300:933-944
  152. Kosiborod M, Inzucchi S, Goyal A, Krumholz HM, Masoudi FA, Xiao L, Spertus JA. Relationship between spontaneous and iatrogenic hypoglycemia and mortality in patients hospitalized with acute myocardial infarction. JAMA 2009;301:1556-1564
  153. Arabi YM, Tamin HM, Rishu AH. Hypoglycemia with intensive insulin therapy in critically ill patients: Predisposing factors and association with mortality. Critical Care Medicine 2009;37:2536-2544
  154. Vriesendorp TM, DeVries JH, van Santen S, Moeniralam HS, de Jong E, Roos YB, Schultz MJ, Rosendaal FR, Hoekstra JB. Evaluation of short-term consequences of hypoglycemia in an intensive care unit. Critical Care Medicine 2006;34:2714-2718
  155. Krinsley JS, Schultz MJ, Spronk PE, Harmsen RE, van Braam Houckgeest F, van der Sluijs JP, Melot C, Preiser JC. Mild hypoglycemia is independently associated with increased mortality in the critically ill. Critical Care 2011;15:R173
  156. National Diabetes Inpatient Audit (NaDIA) - 2016 [article online], 2017. Available from http://www.content.digital.nhs.uk/catalogue/PUB23539.
  157. National Diabetes Inpatient Audit - Harms, 2019 [article online], 2020. Available from https://digital.nhs.uk/data-and-information/publications/statistical/national-diabetes-inpatient-audit---harms/national-diabetes-inpatient-audit---harms-2019/2019.
  158. Dashora U, Sampson M, Castro E, Stanisstreet D, Hillson R. The best hypoglycaemia avoidance initiative in the UK. British Journal of Diabetes 2017;17:74-77
  159. Flanagan D, Avari P, Choudhary P, Lumb A, Misra S, Rayman G, Dhatariya K. Using technology to improve diabetes care in hospital: The challenge and the opportunity. Journal of Diabetes Science and Technology 2023;17:503-508
  160. Gill GV, Woodward A, Casson IF, Weston PJ. Cardiac arrhythmia and nocturnal hypoglycaemia in type 1 diabetes—the 'dead in bed' syndrome revisited. Diabetologia 2009;52:42-45
  161. Desouza C, Salazar H, Cheong B, Murgo J, Fonseca V. Association of hypoglycemia and cardiac ischemia. Diabetes Care 2003;26:1485-1489
  162. Petersen KG, Schluter KJ, Kerp L. Regulation of serum potassium during insulin-induced hypoglycemia. Diabetes 1982;31:615-617
  163. Razavi Nematollahi L, Kitabchi AE, Stentz FB, Wan JY, Larijani BA, Tehrani MM, Gozashti MH, Omidfar K, Taheri E. Proinflammatory cytokines in response to insulin-induced hypoglycemic stress in healthy subjects. Metabolism - Clinical and Experimental 2009;58:443-448
  164. Desouza CV, Bolli GB, Fonseca V. Hypoglycemia, diabetes, and cardiovascular events. Diabetes Care 2010;33:1389-1394
  165. Boucai L, Southern WN, Zonszein J. Hypoglycemia-associated mortality is not drug-associated but linked to comorbidities. American Journal of Medicine 2011;124:1028-1035
  166. George JT, Warriner D, McGrane DJ, Rozario KS, Price HC, Wilmot EG, Kar P, Stratton IM, Jude EB, McKay GA. Lack of confidence among trainee doctors in the management of diabetes: the Trainees Own Perception of Delivery of Care (TOPDOC) Diabetes Study. QJM 2011;104:761-766
  167. Horton WB, Law S, Darji M, Conaway MR, Akbashev MY, Kubiak NT, Kirby JL, Thigpen SC. A multicenter study evaluating perceptions and knowledge of inpatient glycemic control among resident physicians: analyzing themes to inform and improve care. Endocrine Practice 2019;25:1295-1303
  168. Dei Cas A, Aldigeri R, Ridolfi V, Vazzana A, Ciardullo AV, Manicardi V, Sforza A, Tomasi F, Zavaroni D, Zavaroni I, Bonadonna RC. Efficacy of a training programme for the management of diabetes mellitus in the hospital: A randomized study (stage 2 of GOVEPAZ healthcare). Diabetes/Metabolism Research and Reviews 2023;39:e3708
  169. Hommel I, Wollersheim H, Tack CJ, Mulder J, van Gurp PJ, Hulscher ME. Impact of a multifaceted strategy to improve perioperative diabetes care. Diabetic Medicine 2017;34:278-285
  170. Brown G, Dodek P. Intravenous insulin nomogram improves blood glucose control in the critically ill. Critical Care Medicine 2001;29:1714-1719
  171. Rea RS, Donihi AC, Bobeck M, Herout P, McKaveney TP, Kane-Gill SL, Korytkowski MT. Implementing an intravenous insulin infusion protocol in the intensive care unit. American Journal of Health-System Pharmacy 2007;64:385-395
  172. George S, Dale J, Stanisstreet D. A guideline for the use of variable rate intravenous insulin infusion in medical inpatients. Diabetic Medicine 2015;32:706-713
  173. Miller EE, Lalla M, Zaidi A, Elgash M, Zhao H, Rubin DJ. Eating and glycemic control among critically ill patients receiving continuous intravenous insulin. Endocrine Practice 2020;26:43-50
  174. Salinas PD, Mendez CE. Glucose management technologies for the critically ill. Journal of Diabetes Science and Technology 2019;13:682-690
  175. Ullal J, Aloi JA. Subcutaneous insulin dosing calculators for inpatient glucose control. Current Diabetes Reports 2019;19:120
  176. Bouw JW, Campbell N, Hull MA, Juneja R, Guzman O, Overholser BR. A retrospective cohort study of a nurse-driven computerized insulin infusion program versus a paper-based protocol in critically ill patients. Diabetes Technology & Therapeutics 2012;14:125-130
  177. Juneja R, Roudebush C, Kumar N, Macy A, Golas A, Wall D, Wolverton C, Nelson D, Carroll J, Flanders SJ. Utilization of a computerized intravenous insulin infusion program to control blood glucose in the intensive care unit. Diabetes Technology & Therapeutics 2007;9:232-240
  178. Marvin MR, Inzucchi SE, Besterman BJ. Computerization of the Yale insulin infusion protocol and potential insights into causes of hypoglycemia with intravenous insulin. Diabetes Technology & Therapeutics 2013;15:246-252
  179. Bouldin MG, Hong B, Setji T, Greenlee J, Cooper A, Thompson J, Capes K. Evaluation of the efficacy and safety of an eglycemic management system in a community hospital setting. Journal of Diabetes Science and Technology 2021;15:236-241
  180. Goldberg PA, Siegel MD, Sherwin RS, Halickman JI, Lee M, Bailey VA, Lee SL, Dziura JD, Inzucchi SE. Implementation of a safe and effective insulin infusion protocol in a medical intensive care unit. Diabetes Care 2004;27:461-467
  181. Boutin JM, Gauthier L. Insulin infusion therapy in critically ill patients. Canadian Journal of Diabetes 2014;38:144-150
  182. DeSantis AJ, Schmeltz LR, Schmidt K, O'Shea-Mahler E, Rhee C, Wells A, Brandt S, Peterson S, Molitch ME. Inpatient management of hyperglycemia: The Northwestern experience. Endocrine Practice 2006;12:491-505
  183. Davidson PC, Steed RD, Bode BW. Glucommander. A computer-directed intravenous insulin system shown to be safe, simple, and effective in 120,618 h of operation. Diabetes Care 2005;28:2418-2423
  184. Newton CA, Smiley D, Bode BW, Kitabchi AE, Davidson PC, Jacobs S, Steed RD, Stentz F, Peng L, Mulligan P, Freire AX, Temponi A, Umpierrez GE. A comparison study of continuous insulin infusion protocols in the medical intensive care unit: Computer-guided vs. standard column-based algorithms. Journal of Hospital Medicine 2010;5:432-437
  185. Leelarathna L, English SW, Thabit H, Caldwell K, Allen JM, Kumareswaran K, Wilinska ME, Nodale M, Mangat J, Evans ML, Burnstein R, Hovorka R. Feasibility of fully automated closed-loop glucose control using continuous subcutaneous glucose measurements in critical illness: a randomized controlled trial. Critical Care 2013;17:R159
  186. Gómez Medina AM, Henao-Carrillo DC, Yepes C, Silva J, Gómez González JA, Cortes D, Robledo S, Mejía G, Rondon M. Glycemic control metrics in a cohort of hospitalized patients with type 1 diabetes using hybrid closed-loop and advanced hybrid closed-loop systems. Diabetes Research and Clinical Practice 2023;204:110897
  187. Avari P, Lumb A, Flanagan D, Rayman G, Misra S, Choudhary P, Dhatariya K. Insulin pumps and hybrid close loop systems within hospital: A scoping review and practical guidance From the Joint British Diabetes Societies for Inpatient Care. Journal of Diabetes Science and Technology 2023;17:625-634
  188. Migdal AL, Fortin-Leung C, Pasquel F, Wang H, Peng L, Umpierrez GE. Inpatient glycemic control with sliding scale insulin in noncritical patients with type 2 diabetes: Who can slide? Journal of Hospital Medicine 2021;16:462-468
  189. Migdal AL, Idrees T, Umpierrez GE. Selecting insulin regimens for the management of non-ICU patients with type 2 diabetes. Journal of the Endocrine Society 2021;5
  190. Hirsch IB. Sliding scale insulin--time to stop sliding. JAMA 2009;301:213-214
  191. Umpierrez GE, Maynard G. Glycemic chaos (not glycemic control) still the rule for inpatient care. How do we stop the insanity? Journal of Hospital Medicine 2006;1:141-144
  192. Swan E, Dhatariya K. Differential effects of intravenous and subcutaneous sliding scale insulin regimes used to improve blood glucose levels in a tertiary care setting. Practical Diabetes International 2009;26:2i
  193. Vellanki P, Cardona S, Galindo RJ, Urrutia MA, Pasquel FJ, Davis GM, Fayfman M, Migdal A, Peng L, Umpierrez GE. Efficacy and safety of intensive versus nonintensive supplemental insulin with a basal-bolus insulin regimen in hospitalized patients with type 2 diabetes: A randomized clinical study. Diabetes Care 2022;45:2217-2223
  194. King AB, Armstrong DU. Basal bolus dosing: A clinical experience. Current Diabetes Reviews 2005;1:215-220
  195. Galindo RJ, Dhatariya K, Gomez-Peralta F, Umpierrez GE. Safety and efficacy of inpatient diabetes management with non‑insulin agents: an overview of international practices. Current Diabetes Reports 2022;22:237-246
  196. Umpierrez GE, Hor T, Smiley D, Temponi A, Umpierrez D, Ceron M, Munoz C, Newton C, Peng L, Baldwin D. Comparison of inpatient insulin regimens with detemir plus aspart versus neutral protamine hagedorn plus regular in medical patients with type 2 diabetes. Journal of Clinical Endocrinology Metabolism 2009;94:564-569
  197. Umpierrez GE, Smiley D, Hermayer K, Khan A, Olson DE, Newton C, Jacobs S, Rizzo M, Peng L, Reyes D, Pinzon I, Fereira ME, Hunt V, Gore A, Toyoshima MT, Fonseca VA. Randomized study comparing a basal-bolus with a basal plus correction insulin regimen for the hospital management of medical and surgical patients with type 2 diabetes: Basal plus trial. Diabetes Care 2013;36:2169-2174
  198. Pietras S, Hanrahan P, Arnold L, Sternthal E, McDonnell M. State-of-the-art inpatient diabetes care: The evolution of an academic hospital. Endocrine Practice 2010;16:512-521
  199. Clement S, Bowen-Wright H. Twenty-four hour action of insulin glargine (Lantus) may be too short for once-daily dosing: A case report. Diabetes Care 2002;25:1479-1147a
  200. Rubin DJ, Rybin D, Doros G, McDonnell ME. Weight-based, insulin dose-related hypoglycemia in hospitalized patients with diabetes. Diabetes Care 2011;34:1723-1728
  201. Koufakis T, Mustafa OG, Zebekakis P, Kotsa K. Oral antidiabetes agents for the management of inpatient hyperglycaemia: so far, yet so close. Diabetic Medicine 2020;37:1418-1426
  202. Nesto RW, Bell D, Bonow RO, Fonseca V, Grundy SM, Horton ES, Le Winter M, Porte D, Semenkovich CF, Smith S, Young LH, Kahn R. Thiazolidinedione use, fluid retention, and congestive heart failure. Diabetes Care 2004;27:256-263
  203. Khalam A, Dilip C, Shinu C. Drug use evaluation of diabetes mellitus in hospitalized patients of a tertiary care referral hospital. Journal of Basic and Clinical Physiology and Pharmacology 2012;23:173-177
  204. Willi SM, Kennedy A, Brant BP, Wallace P, Rogers NL, Garvey WT. Effective use of thiazolidinediones for the treatment of glucocorticoid-induced diabetes. Diabetes Research & Clinical Practice 2002;58:87-96
  205. Roberts A, James J, Dhatariya K, on behalf of the Joint british Diabetes Societies for Inpatient Care Group. Management of hyperglycaemia and steroid (glucocorticoid) therapy: a guideline from the Joint British Diabetes Societies (JBDS) for Inpatient Care group. Diabetic Medicine 2018;35:1011-1017
  206. Nirantharakumar K, Marshall T, Kennedy A, Hemming K, Coleman JJ. Hypoglycaemia is associated with increased length of stay and mortality in people with diabetes who are hospitalized. Diabetic Medicine 2012;29:e445-e448
  207. Deusenberry CM, Coley KC, Korytkowski MT, Donihi AC. Hypoglycemia in hospitalized patients treated with sulfonylureas. Pharmacotherapy 2012;32:613-617
  208. Stuart K, Adderley NJ, Marshall T, Rayman G, Sitch A, Manley S, Ghosh S, Toulis KA, Nirantharakumar K. Predicting inpatient hypoglycaemia in hospitalized patients with diabetes: a retrospective analysis of 9584 admissions with diabetes. Diabetic Medicine 2017;34:1385-1391
  209. Roumie CL, Greevy RA, Grijalva CG, Hung AM, Liu X, Murff HJ, Elasy TA, Griffin MR. Association between intensification of metformin treatment with insulin vs sulfonylureas and cardiovascular events and all-cause mortality among patients with diabetes. JAMA 2014;311:2288-2296
  210. Gerards MC, Venema GE, Patberg KW, Kross M, Potter van Loon BJ, Hageman IM, Snijders D, Brandjes DP, Hoekstra JB, Vriesendorp TM, Gerdes VE. Dapagliflozin for prednisone-induced hyperglycemia in acute exacerbation of chronic obstructive pulmonary disease. Diabetes, Obesity and Metabolism 2018;20:1306-1310
  211. Empagliflozin to be investigated as a possible treatment for COVID-19 in the RECOVERY trial [article online], 2022. Available from https://www.recoverytrial.net/news/empagliflozin-to-be-investigated-as-a-possible-treatment-for-covid-19-in-the-recovery-trial.
  212. Cohen B, Harris YT, Schulman-Rosenbaum R. SGLT2 inhibitors should be avoided for inpatient management of hyperglycemia. Endocrine Practice 2024;
  213. Dhatariya K. Initiation and continuation of sodium-glucose cotransporter 2 inhibitors in hospital inpatients – ready for prime time? Diabetes Care 2022;45:2806-2807
  214. Umpierrez GE, Gianchandani R, Smiley D, Wesorick DH, Newton C, Farrokhi F, Peng L, Lathkar-Pradhan S, Pasquel F. Safety and efficacy of sitagliptin therapy for the inpatient management of general medicine and surgery patients with type 2 diabetes: A pilot, randomized, controlled study. Diabetes Care 2013;36:3430-3435
  215. Pasquel FJ, Gianchandani R, Rubin DJ, Dungan KM, Anzola I, Gomez PC, Peng L, Hodish I, Bodnar T, Wesorick D, Balakrishnan V, Osei K, Umpierrez GE. Efficacy of sitagliptin for the hospital management of general medicine and surgery patients with type 2 diabetes (Sita-Hospital): a multicentre, prospective, open-label, non-inferiority randomised trial. Lancet Diabetes and Endocrinology 2017;5:125-133
  216. Lorenzo-Gonzalez C, Atienza-Sanchez E, Reyes-Umpierrez D, Vellanki P, Davis GM, Pasquel FJ, Cardona S, Fayman M, Peng L, Umpierrez GE. Safety and efficacy of DDP-4 inhibitors for management of hospitalized general medicine and surgery patients with type 2 diabetes. Endocrine Practice 2020;26:722-728
  217. Hulst AH, Plummer MP, Hollmann MW, DeVries JH, Preckel B, Deane AM, Hermanides J. Systematic review of incretin therapy during peri-operative and intensive care. Critical Care 2018;22:299
  218. Kosiborod MN, Angermann CE, Collins SP, Teerlink JR, Ponikowski P, Biegus J, Comin-Colet J, Ferreira JP, Mentz RJ, Nassif ME, Psotka MA, Tromp J, Brueckmann M, Blatchford JP, Salsali A, Voors AA. Effects of empagliflozin on symptoms, physical limitations and quality of life in patients hospitalized for acute heart failure - results from the EMPULSE trial. Circulation 2022;146
  219. Tamaki S, Yamada T, Watanabe T, Morita T, Furukawa Y, Kawasaki M, Kikuchi A, Kawai T, Seo M, Abe M, Nakamura J, Yamamoto K, Kayama K, Kawahira M, Tanabe K, Fujikawa K, Hata M, Fujita Y, Umayahara Y, Taniuchi S, Sanada S, Shintani A, Fukunami M. Effect of empagliflozin as an add-on therapy on decongestion and renal function in patients with diabetes hospitalized for acute decompensated heart failure. Circulation: Heart Failure 2021;14:e007048
  220. Cunningham JW, Vaduganathan M, Claggett BL, Kulac IJ, Desai AS, Jhund PS, de Boer RA, DeMets D, Hernandez AF, Inzucchi SE, Kosiborod MN, Lam CS, Martinez F, Shah SJ, McGrath MM, O’Meara E, Wilderäng U, Lindholm D, Petersson M, Langkilde A, McMurray JJ, Solomon SD. Dapagliflozin in patients recently hospitalized with heart failure and mildly reduced or preserved ejection fraction. Journal of the American College of Cardiology 2022;80:1302-1310
  221. Voors AA, Angermann CE, Teerlink JR, Collins SP, Kosiborod M, Biegus J, Ferreira JP, Nassif ME, Psotka MA, Tromp J, Borleffs CJ, Ma C, Comin-Colet J, Fu M, Janssens SP, Kiss RG, Mentz RJ, Sakata Y, Schirmer H, Schou M, Schulze PC, Spinarova L, Volterrani M, Wranicz JK, Zeymer U, Zieroth S, Brueckmann M, Blatchford JP, Salsali A, Ponikowski P. The SGLT2 inhibitor empagliflozin in patients hospitalized for acute heart failure: a multinational randomized trial. Nature Medicine 2022;28:568-574
  222. FDA revises labels of SGLT2 inhibitors for diabetes to include warnings about too much acid in the blood and serious urinary tract infections [article online], 2022. Available from https://www.fda.gov/Drugs/DrugSafety/ucm475463.htm.
  223. Guideline for perioperative care for people with diabetes mellitus undergoing elective and emergency surgery [article online], 2022. Available from https://cpoc.org.uk/sites/cpoc/files/documents/2022-12/CPOC-Diabetes-Guideline-Updated2022.pdf.
  224. Rutter CL, Jones C, Dhatariya KK, James J, Irvine L, Wilson EC, Singh H, Walden E, Holland R, Harvey I, Bradley C, Sampson MJ. Determining in-patient diabetes treatment satisfaction in the UK—the DIPSat study. Diabetic Medicine 2013;30:731-738
  225. Avari P, Lumb A, Flanagan D, Rayman G, Misra S, Dhatariya K, Choudhary P. Continuous glucose monitoring within hospital: A scoping review and summary of guidelines from the Joint British Diabetes Societies for Inpatient Care. Journal of Diabetes Science and Technology 2023;17:611-624
  226. Rice MJ, Smith JL, Coursin DB. Glucose measurement in the ICU: Regulatory intersects reality. Critical Care Medicine 2017;45:741-743
  227. Pilackas K, El-Oshar S, Carter C. Clinical reliability of point-of-care glucose testing in critically ill patients. Journal of Diabetes Science and Technology 2020;14:65-69
  228. Misra S, Avari P, Lumb A, Flanagan D, Choudhary P, Rayman G, Dhatariya K. How can point-of-care technologies support in-hospital diabetes care? Journal of Diabetes Science and Technology 2023;17:509-516
  229. Gomez AM, Umpierrez GE. Continuous glucose monitoring in insulin-treated patients in non-ICU settings. Journal of Diabetes Science and Technology 2014;8:930-936
  230. Cardona S, Gomez PC, Vellanki P, Anzola I, Ramos C, Urrutia MA, Haw JS, Fayfman M, Wang H, Galindo RJ, Pasquel FJ, Umpierrez GE. Clinical characteristics and outcomes of symptomatic and asymptomatic hypoglycemia in hospitalized patients with diabetes. BMJ Open Diabetes Research & Care 2018;6:e000607
  231. Galindo RJ, Aleppo G, Klonoff DC, Spanakis EK, Agarwal S, Vellanki P, Olson DE, Umpierrez GE, Davis GM, Pasquel FJ. Implementation of continuous glucose monitoring in the hospital: Emergent considerations for remote glucose monitoring during the COVID-19 pandemic. Journal of Diabetes Science and Technology 2020;14:822-832
  232. Chow KW, Kelly DJ, Rieff MC, Skala PA, Kravets I, Charitou MM, Morley EJ, Gupta R, Miller JD. Outcomes and healthcare provider perceptions of real-time continuous glucose monitoring (rtCGM) in patients with diabetes and COVID-19 admitted to the ICU. Journal of Diabetes Science and Technology 2021;15:607-614
  233. Klarskov CK, Kristensen PL. Experience from implementing telemetric in-hospital continuous glucose monitoring during the COVID-19 pandemic. Journal of Diabetes Science and Technology 2021;15:715-716
  234. Faulds ER, Boutsicaris A, Sumner L, Jones L, McNett M, Smetana KS, May CC, Buschur E, Exline MC, Ringel MD, Dungan K. Use of continuous glucose monitor in critically ill COVID-19 patients requiring insulin infusion: An observational study. Journal of Clinical Endocrinology & Metabolism 2021;106:e4007-e4016
  235. Perez-Guzman MC, Duggan E, Gibanica S, Cardona S, Corujo-Rodriguez A, Faloye A, Halkos M, Umpierrez GE, Peng L, Davis GM, Pasquel FJ. Continuous glucose monitoring in the operating room and cardiac intensive care unit. Diabetes Care 2021;44:e50-e52
  236. Longo RR, Elias H, Khan M, Seley JJ. Use and accuracy of inpatient CGM during the COVID-19 pandemic: An observational study of general medicine and ICU patients. Journal of Diabetes Science and Technology 2022;16:1136-1143
  237. Sweeney AT, Pena S, Sandeep J, Hernandez B, Chen Y, Breeze JL, Bulut A, Feghali K, Abdelrehim M, Abdelazeem M, Srivoleti P, Salvucci L, Cann SB, Norman C. Use of a continuous glucose monitoring system in high-risk hospitalized noncritically ill patients with diabetes after cardiac surgery and during their transition of care from the intensive care unit during COVID-19: A pilot study. Endocrine Practice 2022;28:615-621
  238. Tingsarat W, Buranasupkajorn P, Khovidhunkit W, Boonchaya-anant P, Laichuthai N. The accuracy of continuous glucose monitoring in the medical intensive care unit. Journal of Diabetes Science and Technology 2022;16:1550-1554
  239. Wernerman J, Desaive T, Finfer S, Foubert L, Furnary A, Holzinger U, Hovorka R, Joseph J, Kosiborod M, Krinsley J, Mesotten D, Nasraway S, Rooyackers O, Schultz M, Van Herpe T, Vigersky R, Preiser JC. Continuous glucose control in the ICU: report of a 2013 round table meeting. Critical Care 2014;18:226
  240. Clinical Guideline: Guidelines for managing continuous subcutaneous insulin infusion (CSII, or 'insulin pump') therapy in hospitalised patients [article online], 2019. Available from https://abcd.care/sites/abcd.care/files/CSII_DTN_FINAL%20210218.pdf.
  241. Wang M, Singh LG, Spanakis EK. Advancing the use of CGM devices in a non-ICU setting. Journal of Diabetes Science and Technology 2019;13:674-681
  242. Tsur A, Cahn A, Israel M, Feldhamer I, Hammerman A, Pollack R. Impact of flash glucose monitoring on glucose control and hospitalization in type 1 diabetes: A nationwide cohort study. Diabetes/Metabolism Research and Reviews 2021;37:e3355
  243. Spanakis EK, Urrutia A, Galindo RJ, Vellanki P, Migdal AL, Davis G, Fayfman M, Idrees T, Pasquel FJ, Coronado WZ, Albury B, Moreno E, Singh LG, Marcano I, Lizama S, Gothong C, Munir K, Chesney C, Maguire R, Scott WH, Perez-Guzman MC, Cardona S, Peng L, Umpierrez GE. Continuous glucose monitoring–guided insulin administration in hospitalized patients with diabetes: A randomized clinical trial. Diabetes Care 2022;45:2369-2375
  244. Pasquel FJ, Lansang MC, Dhatariya K, Umpierrez GE. Management of diabetes and hyperglycaemia in the hospital. Lancet Diabetes & Endocrinology 2021;9:174-188
  245. Singh LG, Satyarengga M, Marcano I, Scott WH, Pinault LF, Feng Z, Sorkin JD, Umpierrez GE, Spanakis EK. Reducing inpatient hypoglycemia in the general wards using real-time continuous glucose monitoring: The glucose telemetry system, a randomized clinical trial. Diabetes Care 2020;43:2736-2743
  246. Davis GM, Galindo RJ, Migdal AL, Umpierrez GE. Diabetes technology in the inpatient setting for management of hyperglycemia. Endocrinology and Metabolism Clinics 2020;49:79-93
  247. Wang G, Liu X, Ying Z, Yang G, Chen Z, Liu Z, Zhang M, Yan H, Lu Y, Gao Y, Xue K, Li X, Chen Y. Optimized glycemic control of type 2 diabetes with reinforcement learning: a proof-of-concept trial. Nature Medicine 2023;29:2633-2642
  248. Using technology to support diabetes care in hospital: A guideline from the Joint British Diabetes Societies for Inpatient Care (JBDS-IP) Group and Diabetes Technology Network (DTN) [article online], 2024. Available from https://abcd.care/resource/current/jbds-20-using-technology-support-diabetes-care-hospital.
  249. Lumb A, Misra S, Rayman G, Avari P, Flanagan D, Choudhary P, Dhatariya K. Variation in the current use of technology to support diabetes management in UK hospitals: Results of a survey of health care professionals. Journal of Diabetes Science and Technology 2023;17:733-741
  250. McMahon MM, Nystrom E, Braunschweig C, Miles J, Compher C, Nutrition ASfPaE. A.S.P.E.N. clinical guidelines: Nutrition support of adult patients with hyperglycemia. Journal of Parenteral and Enteral Nutrition 2013;37:23-36
  251. Lobo DN, Gianotti L, Adiamah A, Barazzoni R, Deutz NE, Dhatariya K, Greenhaff PL, Hiesmayr M, Hjort Jakobsen D, Klek S, Krznaric Z, Ljungqvist O, McMillan DC, Rollins KE, Panisic Sekeljic M, Skipworth RJ, Stanga Z, Stockley A, Stockley R, Weimann A. Perioperative nutrition: Recommendations from the ESPEN expert group. Clinical Nutrition 2020;39:3211-3227
  252. JBDS guideline on the glycaemic management during the inpatient enteral feeding of stroke patients with diabetes [article online], 2012. Available from http://www.diabetologists-abcd.org.uk/JBDS/JBDS_IP_Enteral_Feeding_Stroke.pdf.
  253. Flanagan D, Lake AK, Green M, Roberts A, Dhatariya K, on behalf of the Joint British Diabetes Societies for Inpatient Care Group. A UK national survey of enteral feed use in people with diabetes 2022. Diabetic Medicine 2023;n/a:e15216
  254. McMahon MM, Rizza RA. Nutrition support in hospitalized patients with diabetes mellitus. Mayo Clinic Proceedings 1996;71:587-594
  255. Singer P, Blaser AR, Berger MM, Alhazzani W, Calder PC, Casaer MP, Hiesmayr M, Mayer K, Montejo JC, Pichard C, Preiser J-C, van Zanten AR, Oczkowski S, Szczeklik W, Bischoff SC. ESPEN guideline on clinical nutrition in the intensive care unit. Clinical Nutrition 2019;38:48-79
  256. Preiser J-C, Arabi YM, Berger MM, Casaer M, McClave S, Montejo-González JC, Peake S, Reintam Blaser A, Van den Berghe G, van Zanten A, Wernerman J, Wischmeyer P. A guide to enteral nutrition in intensive care units: 10 expert tips for the daily practice. Critical Care 2021;25:424
  257. Via MA, Mechanick JI. Inpatient enteral and parental nutrition for patients with diabetes. Current Diabetes Reports 2011;11:99-105
  258. Elia M, Ceriello A, Laube H, Sinclair AJ, Engfer M, Stratton RJ. Enteral nutritional support and use of diabetes-specific formulas for patients with diabetes: A systematic review and meta-analysis. Diabetes Care 2005;28:2267-2279
  259. Glycaemic management during the inpatient enteral feeding of stroke patients with diabetes [article online], 2024. Available from https://abcd.care/resource/jbds-05-glycaemic-management-during-inpatient-enteral-feeding-stroke-patients-diabetes.
  260. Narwani V, Swafe L, Stavraka C, Dhatariya K. How frequently are bedside glucose levels measured in hospital inpatients on glucocorticoids? Clinical Medicine 2014;14:327-328
  261. Sudlow A, O'Connor HM, Narwani V, Swafe L, Dhatariya K. Assessing the prevalence of dexamethasone use in patients undergoing surgery, and subsequent glucose measurements: a retrospective cohort study. Practical Diabetes 2017;34:117-121
  262. Limbachia V, Nunney I, Page DJ, Barton HA, Patel LK, Thomason GN, Green SL, Lewis KF, Dhatariya K. The effect of different types of oral or intravenous corticosteroids on capillary blood glucose levels in hospitalized inpatients with and without diabetes. Clinical Therapeutics 2024;46
  263. Limbachia V, Nunney I, Page DJ, Barton HA, Patel LK, Thomason GN, Green SL, Lewis KF, Dhatariya K. The effect of different types of oral or intravenous corticosteroids on capillary blood glucose levels in hospitalized inpatients with and without diabetes. Clinical Therapeutics 2024;46:e59-363
  264. Flanagan D, Dhatariya K, Kilvert A, on behalf of the Joint British Diabetes Societies for Inpatient Care (JBDS-IP) Group. Self-management of diabetes in hospital: a guideline from the Joint British Diabetes Societies (JBDS) for Inpatient Care group. Diabetic Medicine 2018;35:992-996
  265. Yatabe T, Yamazaki R, Kitagawa H, Okabayashi T, Yamashita K, Hanazaki K, Yokoama M. The evaluation of the ability of closed-loop glycemic control device to maintain the blood glucose concentration in intensive care unit patients. Critical Care Medicine 2011;39:575-578
  266. Levitt DL, Spanakis EK, Ryan KA, Silver KD. Insulin pump and continuous glucose monitor initiation in hospitalized patients with type 2 diabetes mellitus. Diabetes Technology & Therapeutics 2018;20:32-38
  267. Pelkey MN, Boyle ME, Long A, Castro JC, Cook CB, Thompson B. Hybrid closed loop insulin pump technology can be safely used In the inpatient setting. Endocrine Practice 2023;29:24-28
  268. Bally L, Thabit H, Hartnell S, Andereggen E, Ruan Y, Wilinska ME, Evans ML, Wertli MM, Coll AP, Stettler C, Hovorka R. Closed-loop insulin delivery for glycemic control in noncritical care. New England Journal of Medicine 2018;379:547-556
  269. Thabit H, Hartnell S, Allen JM, Lake A, Wilinska ME, Ruan Y, Evans ML, Coll AP, Hovorka R. Closed-loop insulin delivery in inpatients with type 2 diabetes: a randomised, parallel-group trial. Lancet Diabetes & Endocrinology 2020;5:117-124
  270. Boughton CK, Bally L, Martignoni F, Hartnell S, Herzig D, Vogt A, Wertli MM, Wilinska ME, Evans ML, Coll AP, Stettler C, Hovorka R. Fully closed-loop insulin delivery in inpatients receiving nutritional support: a two-centre, open-label, randomised controlled trial. Lancet Diabetes & Endocrinology 2019;7:368-377
  271. Stewart ZA, Wilinska ME, Hartnell S, Temple RC, Rayman G, Stanley KP, Simmons D, Law GR, Scott EM, Hovorka R, Murphy HR. Closed-loop insulin delivery during pregnancy in women with type 1 diabetes. New England Journal of Medicine 2016;375:644-654

 

Assessing Insulin Sensitivity and Resistance in Humans

ABSTRACT

 

In this chapter we discuss a representative variety of methods currently available for estimating insulin sensitivity/resistance. These range from complex, time consuming, labor-intensive, invasive procedures to simple testsinvolving a single fasting blood sample. It is important to understand the physiological concepts informing each method so that relative merits and limitations of particular approaches are appropriately matched with proposed applications and data is interpreted correctly. The glucose clamp method is the reference standard for direct measurement of insulin sensitivity. Regarding simple surrogates, QUICKI and Log (HOMA) are among the best and most extensively validated. Dynamic tests are useful if information about both insulin secretion and insulin action are needed.

 

INTRODUCTION

 

Insulin resistance plays a major pathophysiological role in type 2 diabetes and is tightly associated with major public health problems including obesity, hypertension, coronary artery disease, dyslipidemias, and a cluster of metabolic and cardiovascular abnormalities that define the metabolic syndrome (1, 2, 3).

 

A global epidemic of obesity is driving the increased incidence and prevalence of type 2 diabetes and its cardiovascular complications (4). Insulin resistance is commonly associated with visceral adiposity, glucose intolerance, hypertension, dyslipidemia, hypercoagulable state, endothelial dysfunction, and/or elevated markers of inflammation. Therefore, the presence of these clinical abnormalities is usually characteristic of an insulin resistant state. In addition to clinical manifestations of the “Insulin Resistance Syndrome,” insulin resistance predisposes to accelerated cardiovascular disease (CVD). Therefore, it is of great importance to develop tools for quantifying insulin sensitivity/resistance in humans that may be used to appropriately investigate the epidemiology, pathophysiological mechanisms, outcomes of therapeutic interventions, and clinical course of patients with insulin resistance (5). In this chapter, we will discuss some currently used methods for assessing insulin sensitivity, their applications, merits, and limitations.

 

INSULIN SENSITIVITY AND RESISTANCE

 

Metabolic actions of insulin help to maintain glucose homeostasis and promote glucose utilization (6). Insulin increases glucose utilization in peripheral organs (e.g., skeletal muscle and adipose tissue) and suppresses hepatic glucose production (HGP) and adipose tissue lipolysis. In addition to these classical metabolic target tissues, insulin has many other important physiological targets. These include the brain, pancreatic β-cells, heart, and vascular endothelium that help to coordinate and couple metabolic and cardiovascular homeostasis under healthy conditions (6-9). Insulin has concentration- dependent saturable actions to increase whole-body glucose disposal. The maximal effect of insulin defines “insulin responsiveness” while the insulin concentration required for a half-maximal response defines “insulin sensitivity” (Fig. 1). Although, other actions of insulin on fat and amino-acid metabolism, cardiovascular, kidney, and brain function also exhibit a concentration-dependent response, the term “insulin sensitivity” typically refers to insulin’s metabolic actions to promote glucose disposal.

 

The concept of insulin resistance was proposed as early as 1936 to describe diabetic patients requiring high doses of insulin (10). Insulin resistance is typically defined as decreased sensitivity and/or responsiveness to insulin- mediatedglucose disposal and/or inhibition of HGP and adipose tissue lipolysis. Rigorous evaluation of altered sensitivity and responsiveness therefore requires a comparison of insulin dose-response curves.

 

Figure 1. Schematic representation of concentration-response relationships between plasma insulin concentrations and insulin-mediated whole-body glucose disposal. Curve a: normal insulin sensitivity and responsiveness. Curve b: rightward shift in insulin concentration-response curve. This represents decreased insulin sensitivity (increased EC50) with normal insulin responsiveness. Curve c: Decreased insulin sensitivity (increased EC50) and reduced insulin responsiveness. Curve d: Leftward shift in the insulin concentration- response curve. This represents increased insulin sensitivity (decreased EC50) with normal insulin responsiveness.

 

DIRECT MEASURES OF INSULIN SENSITIVITY

 

Hyperinsulinemic Euglycemic Glucose Clamp

 

PROCEDURE

 

The glucose clamp technique, originally developed by Andres and DeFronzo is widely accepted as the reference standard for directly determining metabolic insulin sensitivity in humans (11). After an overnight fast, insulin is infused intravenously at a constant rate that may range from 5 - 120 mU/m2/min (dose per body surface area per minute). This constant insulin infusion results in a new steady-state insulin level that is above the fasting level (hyperinsulinemic). As a consequence, glucose disposal in skeletal muscle and adipose tissue is increased while HGP is suppressed. Under these conditions, a bedside glucose analyzer is used to frequently monitor blood glucose levels at 5 – 10 min intervals while 20% dextrose is given intravenously at a variable rate in order to “clamp” blood glucose concentrations in the normal range (euglycemic). An infusion of potassium phosphate is also given to prevent hypokalemia resulting from hyperinsulinemia and increased glucose disposal. After several hours of constant insulin infusion, steady-state conditions are typically achieved for plasma insulin, blood glucose, and the glucose infusion rate (GIR). Assuming that the hyperinsulinemic state is sufficient to completely suppress hepatic glucose production, and since there is no net change in blood glucose concentrations under steady- state clamp conditions, the GIR must be equal to the glucose disposal rate (M) (Fig. 2). Thus, whole body glucose disposal at a given level of hyperinsulinemia can be directly determined. M is typically normalized to body weight or fat-free mass to generate an estimate of insulin sensitivity.Alternatively, an insulin sensitivity index derived from clamp data can be defined as SIClamp = M/(G x ΔI), where M is normalized for G (steady-state blood glucose concentration) and ΔI (difference between fasting and steady-state plasma insulin concentrations) (12).

 

Figure 2 Schematic representation of the “steady state” dynamics of glucose and insulin during an euglycemic hyperinsulinemic glucose clamp.

 

The validity of glucose clamp measurements of insulin sensitivity depends on achieving steady-state conditions. “Steady-state” is often defined as a period greater than 30- min (at least 1 h after initiation of insulin infusion) during which the coefficient of variation for blood glucose, plasma insulin, and GIR are less than 5% (12, 13). It is possible to use stable isotope or radio-labeled glucose tracer under clamp conditions to estimate HGP so that appropriate corrections can be made to M in the event HGP is not completely suppressed (14, 15, 16,17). An alternative approach is to use an insulin infusion rate sufficiently high to completely suppress HGP according to the insulin sensitivity/resistance of the population to be studied. M is routinely obtained at only a single insulin infusion rate and therefore comparisons between M or SIClamp among different subjects is valid only if the same insulin infusion rate is used for all subjects. When glucose tracers are used during a clamp study, the tracer is infused at constant rate throughout the study. HGP estimated during the last 20 or 30 min of the clamp is a measure of insulin- mediated suppression of HGP, an estimate of hepatic insulin sensitivity. Similarly, lipolytic rates can be assessed at baseline and hyperinsulinemia during clamp by using isotopic tracers (e.g., palmitate). A single or multistep hyperinsulinemic euglycemic clamp can be used to measure adipose tissue insulin sensitivity. The linear relationship between log transformed rates of palmitate flux and plasma insulin concentrations provides an IC50 (pmol/L) for suppression of lipolysis (18).

 

ADVANTAGES AND LIMITATIONS

 

The principal advantage of the glucose clamp in humans is that it directly measures whole body glucose disposal at a given level of insulinemia under steady-state conditions. Conceptually, the approach is straightforward and there are a limited number of assumptions which are clearly defined. In research settings where assessing insulin sensitivity/resistance is of primary interest and feasibility is not an issue (e.g., study population < 100) it is appropriate to use the reference standard glucose clamp technique. The main limitations of the clamp approach are that it is time-consuming, labor intensive, expensive, and requires an experienced operator to manage technical difficulties. Thus, for epidemiological studies, large clinical investigations, or routine clinical applications (e.g., following changes in insulinresistance after therapeutic intervention in individual patients) application of the glucose clamp is not feasible. Nevertheless, when measured in relatively large cohorts, the M values showed a bimodal pattern, with an optimal cutoff of 5 mg/min/kg-FFM using a 40 mU/min·m2. However, IR was defined as a glucose disposal rate below 4.9 mg/min/kg, using a 120 mU/min·m2 dose (80, 81).

 

Insulin-Suppression Test (IST)

 

PROCEDURE

 

The insulin-suppression test, another method that directly measures metabolic insulin sensitivity/resistance, was introduced by Shen et. al. in 1970 and subsequently modified by Harano et. al. (19, 20). After an overnight fast, somatostatin (250 μg/h) or the somatostatin analogue octreotide (25 µg bolus, followed by 0.5 µg/min) (21) is intravenously infused to suppress endogenous secretion of insulin and glucagon. Simultaneously, insulin (25 mU/m2/min) and glucose (240 mg/m2/min) are infused into the same antecubital vein over 3 h. From the contralateral arm, blood samples for glucose and insulin determinations are taken every 30 min for 2.5 h and then at 10 min intervals from 150 - 180 min of the IST. The constant infusions of insulin and glucose determine steady-state plasma insulin (SSPI) andglucose (SSPG) concentrations. The steady-state period is assumed to be from 150 - 180 min after initiation of the IST. SSPI concentrations are generally (but not always) similar among subjects. Therefore, the SSPG concentration will be higher in insulin resistant subjects and lower in insulin sensitive subjects. That is, SSPG values are inversely related to insulin sensitivity. The IST provides a direct measure (SSPG) of the ability of exogenous insulin to mediate disposal of an intravenous glucose load under steady-state conditions where endogenous insulin secretion is suppressed.

 

ADVANTAGES AND LIMITATIONS

 

The SSPG is a highly reproducible direct measure of metabolic actions of insulin that is less labor-intensive and less technically demanding than the glucose clamp. Indeed, since there are no variable infusions with the IST, steady-state conditions are more easily achieved with the IST than with the glucose clamp. Estimates of insulin sensitivity determined by SSPG correlate well with reference standard glucose clamp estimates in normal subjects (r = 0.93) and in patients with type 2 diabetes mellitus (r = 0.91). (22, 23). Indeed, SSPG has positive predictive power for cardiovascular disease events and onset of type 2 diabetes (24, 25). In research settings where assessing insulin sensitivity/resistance is of primary interest and feasibility is not an issue, it is appropriate to use the IST. Moreover, the IST can be used for larger populations that may pose difficulties for application of the glucose clamp (26). Many of the limitations of the IST are similar to those described above for the glucose clamp (with the exception that the IST is less technically demanding).Thus, it is impractical to apply the IST in large epidemiological studies or in the clinical care setting. SSPG under ideal conditions determines primarily skeletal muscle insulin sensitivity and is not designed to reflect hepatic insulin sensitivity.

 

INDIRECT MEASURES OF INSULIN SENSITIVITY

 

Minimal Model Analysis of Frequently Sampled Intravenous Glucose Tolerance Test (FSIVGTT)

 

PROCEDURE

 

The minimal model, developed by Bergman, Cobelli, and colleagues in 1979, provides an indirect measure of metabolic insulin sensitivity/resistance based on glucose and insulin data obtained during an FSIVGTT (27). After an overnight fast, an intravenous bolus of glucose (0.3 g/kg body weight) is infused over 2 min starting at time 0. Currently, a modified FSIVGTT is used where exogenous insulin (4 mU/kg/min) is also infused over 5 min beginning 20 min after the intravenous glucose bolus (28, 29,30). Some studies use tolbutamide instead of insulin in the modified FSIVGTT to stimulate endogenous insulin secretion (15, 29, 31, 32, 27). Blood samples are taken for plasma glucose and insulinmeasurements at -10, -1, 1, 2, 3, 4, 5, 6, 7, 8, 10, 12, 14, 16, 20, 22, 23, 24, 25, 27, 30, 40, 50, 60, 70, 80, 90, 100,120, 160, and 180 min. These data are then subjected to minimal model analysis using the computer program MINMOD to generate an index of insulin sensitivity (SI).

 

The minimal model is defined by two coupled differential equations with four model parameters (Fig. 3). The first equation describes plasma glucose dynamics in a single compartment. The second equation describes insulin dynamics in a “remote compartment”. The structure of the minimal model allows MINMOD to uniquely identify model parameters that determine a best fit to glucose disappearance during the modified FSIVGTT. SI is calculated from two of these model parameters and is defined as fractional glucose disappearance per insulin concentration unit. In addition to SI, other minimal model parameters may be used to estimate a “glucose effectiveness” index (SG). SG is defined as the ability of glucose per se to promote its own disposal and inhibit HGP in the absence of an incremental insulin effect (i.e., when insulin is at basal or constant concentrations).

 

Recently the minimal model has been used to assess free fatty acid (FFA) insulin sensitivity. Using a one compartment nonlinear model of FFA kinetics during FSIVGTT, showed that the FFA insulin sensitivity parameter correlated well with minimal model indices (33). Furthermore, this model also showed that glucose modulates disposal of FFAs.

 

Figure 3. Schematic equations, and parameters for the minimal model of glucose metabolism. Differential equations describing glucose dynamics (G(t)) in a monocompartmental “glucose space” and insulin dynamics in a “remote compartment” (X(t)) are shown at the top. Glucose leaves or enters its space at a rate proportional to the difference between plasma glucose level, G(t) and the basal fasting level, Gb. In addition, glucose also disappears from its compartment at a rate proportional to insulin levels in the “remote” compartment (X(t)). In this model, t = time; G(t) = plasma glucose at time t; I(t) = plasma insulin concentration at time t; X(t) = insulin concentration in “remote” compartment at time t; Gb = basal plasma concentration; Ib = basal plasma insulin concentration; G(0) = G0 (assuming instantaneous mixing of the IV glucose load); p1, p2, p3, and G0 = unknown parameters in the model that are uniquely identifiable from FSIVGTT; glucose effectiveness, SG = p1; and insulin sensitivity, SI = p3/p2.

 

ADVANTAGES AND LIMITATIONS

 

Minimal model analysis of the modified FSIVGTT is easier than the glucose clamp method because it is slightly less labor intensive, steady-state conditions are not required, and there are no intravenous infusions that require constant adjustment. Unlike the glucose clamp or IST, information about insulin sensitivity, glucose effectiveness, and β-cellfunction can be derived from a single dynamic test. The minimal model generates excellent predictions of glucose disappearance during the FSIVGTT. SI is a strong predictor of the development of diabetes in a prospective study of children of diabetic parents (34). Moreover, the insulin-modified FSIVGT may be used in relatively large- scale population studies (35). Therefore, in research settings where assessing insulin sensitivity along with glucose effectiveness and β-cell function is of interest, minimal model analysis of the insulin-modified FSIVGTT may be appropriate. The minimal model approach is simpler than direct methods for determining insulin sensitivity. Nevertheless, it still involves intravenous infusions with multiple blood sampling over a 3 h period that is nearly as labor intensive as the glucose clamp or IST. In addition, many limitations of minimal model analysis stem from the fact that the model oversimplifies the physiology of glucose homeostasis and is discussed in detail elsewhere (5).

 

Oral Glucose Tolerance Test (OGTT)

 

The oral glucose tolerance test (OGTT) is a simple test widely used in clinical practice to diagnose glucose intolerance and type 2 diabetes (36). After overnight fast, blood samples for determinations of glucose and insulin concentrations are taken at 0, 30, 60, and 120 min following a standard 75g oral glucose load. Oral glucose tolerance reflects the efficiency of the body to dispose of glucose after an oral glucose load or meal. The OGTT mimics the glucose and insulin dynamics of physiological conditions more closely than conditions of the glucose clamp, IST, or FSIVGTT. However, it is important to recognize that glucose tolerance and insulin sensitivity are not equivalent concepts. In addition to metabolic actions of insulin, insulin secretion, incretin effects, and other factors contribute importantly to glucose tolerance. Thus, the OGTT and meal tolerance tests provide useful information about glucose tolerance but not insulin sensitivity/resistance per se.

 

Intravenous and Oral Tracer Studies

 

The use of tracers for estimation of insulin sensitivity was first introduced in 1986 to overcome the shortcomings of FSIVGTT (37) The minimal model method does not allow segregation of glucose production from liver from exogenously administered glucose during calculations of insulin sensitivity and thus induces error in the insulin sensitivity calculations. Labeled intravenous glucose can be differentiated from endogenously produced glucose and thus use of labeled glucose during IVGTT provides more precise and accurate measurements (38,39) Similarly, labeled glucose has been used in oral glucose tolerance test and insulin sensitivity has been calculated by minimal model technique similar toFSIVGTT(40). There is a strong correlation of insulin sensitivity calculated from labeled oral minimal model with insulin sensitivity calculated from gold standard euglycemic hyperinsulinemic clamp, r=0.7, p<0.001 (41). There are dual tracer and triple tracer methods as well to estimate the hepatic/endogenous glucose production and discussion of these methods is beyond the scope of this review (42). Basal hepatic insulin resistance index can then be estimated as the product of HGP rate and the fasting plasma insulin concentration. Use of tracer definitely allows for improvement over the FSIVGTT. Use of labeled oral glucose allows for more precise measurements of insulin sensitivity and glucose disposal from a simple OGTT and this can be a useful tool in large studies. The triple tracer method is cumbersome and cannot be employed in large studies.

 

SIMPLE SURROGATE INDEXES FOR INSULIN SENSITIVITY/RESISTANCE

 

Surrogates Derived from Fasting Steady-state Conditions

 

PROCEDURE

 

After an overnight fast, a single blood sample is taken for determination of blood glucose and plasma insulin. In healthy humans, the fasting condition represents a basal steady-state where glucose is homeostatically maintained in the normal range such that insulin levels are not significantly changing and HGP is constant. That is, basal insulin secretion by pancreatic β cells determines a relatively constant level of insulinemia that will be lower or higher in accordance with insulin sensitivity/resistance such that HGP matches whole body glucose disposal under fasting conditions. Surrogate indexes based on fasting glucose and insulin concentrations reflect primarily hepatic insulin sensitivity/resistance. However, under most conditions, hepatic and skeletal muscle insulin sensitivity/resistance are proportional to each other. In the diabetic state with fasting hyperglycemia, fasting insulin levels are inappropriately low and insufficient to maintain euglycemia. Therefore, definitions of the more useful surrogate indexes take these considerations into account. Due to lack of a standardized insulin assay, it is not possible to use surrogate indexes to define universal cutoff points for insulin resistance.

 

ADVANTAGES AND LIMITATIONS

 

Simple surrogate indexes of insulin sensitivity/resistance are inexpensive quantitative tools that can be easily applied in almost every setting including epidemiological studies, large clinical trials, clinical research investigations, and clinical practice. If a direct measure of insulin sensitivity is not required, not feasible to obtain, or if insulin sensitivity is ofsecondary interest, it may be appropriate to use a surrogate index. The relative merits and limitations of individual surrogate indexes are discussed below.

 

The Homeostasis Model Assessment (HOMA)

 

HOMA, developed in 1985, is a model of interactions between glucose and insulin dynamics that is then used to predict fasting steady-state glucose and insulin concentrations for a wide range of possible combinations of insulin resistance and β-cell function (43). The model assumes a feedback loop between the liver and β-cell (43, 44, 45); glucose concentrations are regulated by insulin- dependent HGP while insulin levels depend on the pancreatic β-cell response to glucose concentrations. Thus, deficient β-cell function reflects a diminished response to glucose-stimulated insulinsecretion. Likewise, insulin resistance is reflected by diminished suppressive effect of insulin on HGP. HOMA model describes this glucose-insulin homeostasis by a set of empirically derived non-linear equations. The model predicts fasting steady- state levels of plasma glucose and insulin for any given combination of pancreatic β-cell function and insulin sensitivity. Computer simulations have been used to generate a grid from which mathematical transformations of fasting glucose and insulin β-cell function (HOMA %B) from steady-state conditions. An important caveat for HOMA is that it imputes dynamic β-cell function (i.e., glucose-stimulated insulin secretion) from fasting steady- state data. In the absence of dynamic data, it is difficult, if not impossible, to determine the true dynamic function of β-cell insulin secretion.

 

In practical terms, most studies using HOMA employ an approximation described by a simple equation to determine a surrogate index of insulin resistance. This is defined by the product of the fasting glucose and fasting insulin divided by a constant. Thus, HOMA-IR = fasting insulin (μU/ml) × fasting glucose (mmol/l) / 22.5. The constant is a normalizing factor, the product of fasting plasma insulin of 5 µU/mL and plasma glucose of 4.5 mmol/L obtained from an “ideal” and “normal” individual. Therefore, for an individual with normal insulin sensitivity, HOMA-IR = 1. It is important to note that over wide ranges of insulin sensitivity/resistance Log (HOMA-IR), (which normalizes the skewed distribution of fasting insulinvalues) determines a much stronger linear correlation with glucose clamp estimates of insulin sensitivity (12). HOMA orLog  (HOMA) is extensively used in large epidemiological studies, prospective clinical trials, and clinical research studies (45, 46, 47). In research settings where assessing insulin sensitivity/resistance is of secondary interest or feasibility issues preclude the use of direct measures by glucose clamp, it may be appropriate to use Log (HOMA-IR). However, as discussed below, other surrogate indexes have certain advantages over HOMA or Log (HOMA) in some circumstances.

 

Quantitative Insulin Sensitivity Check Index (QUICKI)

 

QUICKI is an empirically-derived mathematical transformation of fasting blood glucose and plasma insulin concentrations that provides a reliable, reproducible, and accurate index of insulin sensitivity with excellent positive predictive power (12, 48,13, 49, 50). Since fasting insulin levels have a non-normal skewed distribution, log transformation improves its linear correlation with SIclamp. However, as with 1/(fasting insulin) and the G/I ratio, this correlation is not maintained in diabetic subjects with fasting hyperglycemia and impaired β-cell function that is insufficient to maintain euglycemia. To accommodate these clinically important circumstances where fasting glucose is inappropriately high and insulin is inappropriately low, addition of log (fasting glucose) to log (fasting insulin) provides a reasonable correction such that the linear correlation with SIClamp is maintained in both diabetic and non-diabetic subjects. The reciprocal of this sum results in further transformation of the data generating an insulin sensitivity index that has a positive correlation with SIclamp. Thus, QUICKI = 1/Log (Fasting Insulin, µU/ml) + Log (Fasting Glucose, mg/dl). Over a wide range of insulin sensitivity/resistance, QUICKI has a substantially better linear correlation with SIclamp (r ≈ 0.8 – 0.9) than SI derived from the minimal model or HOMA-IR (12, 48, 49). Log (HOMA) is roughly comparable to QUICKI in this regard. Multiple independent studies find excellent linear correlations between QUICKI and glucose clampestimates (either GIR or SIClamp) in healthy subjects, obesity, diabetes, hypertension, and many other insulin- resistant states (49, 51, 52, 53, 54, 55, 56). QUICKI is among the most thoroughly evaluated and validated surrogate index for insulin sensitivity. As a simple, useful, inexpensive, and minimally invasive surrogate for glucose clamp-derived measures of insulin sensitivity, QUICKI is appropriate and effective for use in large epidemiological or clinical researchstudies, to follow changes after therapeutic interventions, and for use in studies where evaluation of insulin sensitivity is not of primary interest.

 

Adipose Tissue Insulin Resistance Index (Adipo-IR)

 

Adipo-IR is a measure similar to HOMA-IR in that it is obtained from a fasting level of FFA and insulin (product of FFA and insulin levels). Recent studies have shown that Adipo-IR correlates well with the gold standard measure of adipose tissue insulin sensitivity derived from one-step hyperinsulinemic-euglycemic clamp technique using a palmitate tracer (57). Age and physical fitness were however shown to affect the predictive values. Thus, Adipo-IR may be suitable for larger population studies, however the multistep pancreatic clamp technique is probably needed for mechanistic studies of adipose tissue insulin action.

 

Surrogates Derived from Dynamic Tests

 

PROCEDURE

 

Surrogate indexes of insulin sensitivity that use information derived from dynamic tests include OGTT, meal tolerance tests, and IVGTT. Procedures for these tests have been described in a previous section. Specific indexes including Matsuda index (58), Stumvoll index (59), Avignon index (60), oral glucose insulin sensitivity index (OGSI) (61), Gutt index (62), and Belfiore index (63) use particular sampling protocols during the OGTT or the meal. In addition, minimal model approaches have been used to model plasma glucose and insulin dynamics during an OGTT or a meal to determine insulin sensitivity/resistance (64). Glucose disposal of an oral glucose load or a meal is mediated by a complex dynamic process that includes gut absorption, glucose effectiveness, neurohormonal actions, incretin actions, insulin secretion, and metabolic actions of insulin that primarily determine the balance between peripheral glucose utilization and HGP. Surrogate indexes that depend on dynamic testing take into account both fasting steady-state and dynamic post-glucose load plasma glucose and insulin levels. After an oral glucose challenge, the HGP is maximally suppressed for approximately 60 min and remains suppressed at a constant level for the subsequent 60–120 min time period. Therefore, glucose uptake by peripheral tissues (e.g., muscle and adipose tissue) primarily determines the rate ofdecrease in plasma glucose concentration from its peak value to its nadir during an OGTT. Based on this observation, surrogate indices of hepatic and muscle insulin sensitivity/ resistance from an OGTT has been widely used (65). Recent studies comparing the OGTT- derived, tissue-specific surrogate indices hepatic insulin resistance index (HIRI) andmuscle insulin sensitivity index (MISI) with clamp measurements showed that surrogate indices derived from an OGTT are accurate in predicting insulin sensitivity but are not tissue-specific (66). Studies using oral tracers in OGTT, with measurement of insulin sensitivity from OGTT and then comparing these to clamp measurements, would be crucial to ascertain the validity these measures. Indeed, recent studies have shown that it is possible to measure hepatic insulin sensitivity in healthy volunteers and in prediabetes with the use of single tracer (67).

 

ADVANTAGES AND LIMITATIONS

 

Many surrogate measures derived from dynamic data correlate reasonably well with glucose clamp estimates of insulin sensitivity (58, 61,62). Estimates of insulin sensitivity derived from OGTT predict the development of type 2 diabetes in epidemiologic studies ( 50, 68, 65). The advantage of surrogates based on dynamic testing is that information about insulin secretion can be obtained at the same time as information about insulin action. However, if one is only interested in estimating insulin sensitivity/resistance, fasting surrogates may be preferable to dynamic surrogates because they are simpler to obtain. The oral route of glucose delivery is more physiological than intravenous glucose infusion. However, poor reproducibility of the OGTT and meal tolerance test due to variable glucose absorption, splanchnic glucose uptake, and additional incretin effects need to be considered. Thus, distinguishing direct metabolic actions of insulin following oral ingestion of glucose or a mixed meal is more problematic than after FSIVGTT. In addition, as with many other measures of insulin sensitivity, surrogates derived from dynamic testing generally incorporate both peripheral and hepatic insulin sensitivity. Although OGTT involves considerably less work than FSIVGTT, dynamic testing in general requires more effort and cost than fasting blood sampling.

 

ETHNIC DIFFERENCES

 

Hispanics, African Americans, and South Asians are highly prone to develop diabetes. A meta-analysis showed that non-diabetic Africans have lower insulin sensitivity and higher insulin response after an intravenous glucose load comparedto Caucasians and East Asians ( 69). In a study that compared euglycemic hyperinsulinemic clamp derived glucosedisposal rates (GDR) with HOMA-IR, QUICKI, and OGTT-derived indices, fasting insulin levels and HOMA-IR did not correlate with GDR whereas Matsuda index derived from OGTT significantly correlated with GDR in African American men (70). Similarly, in another study in Afro-Caribbean adults, HOMA-IR did not correlate with insulin sensitivity calculated from FSIVGTT and M-value calculated from hyperinsulinemic euglycemic clamp (71). Likewise, IR predictive ability of QUICKI and HOMA-IR was limited in Asian-Indian men (72). Recent studies highlight that minimal model may underestimate insulin sensitivity between groups when acute insulin response (AIR) is higher in one group (73). African Americans have reduced insulin clearance and higher AIR than Whites, suggesting that the minimal model may underestimate insulin sensitivity in African Americans (73). These studies suggest that at least in some ethnic groups, QUICKI and HOMA-IR may only be useful as secondary outcome measurements in assessing insulin sensitivity/resistance and studies inferring lower insulin sensitivity in non- diabetic African Americans based on FSIVGTT and minimal modeling should be interpreted cautiously.

 

METABOLOMICS

 

Metabolomics is an interrogation and quantification of small-molecule metabolites in body fluids and tissues. It aims at identifying and quantifying small molecules in the sample by either using mass spectrometry (MS) or nuclear magnetic resonance (NMR) spectroscopy. The details of the methodology and its application in diabetes research are beyond the scope of this chapter. In this chapter, we will focus on new markers of insulin resistance that have been discovered using this approach. Using a non- targeted approach, Gall et al. metabolically profiled fasting plasma samples from 399 non-diabetic, clinically healthy subjects (74). Insulin sensitivity was measured using euglycemic hyperinsulinemic clamps. Individuals in the bottom tertile of the cohort were designated as insulin- resistant. Among the 485 candidate biomarkers identified, plasma α-hydroxybutyrate levels were inversely related to insulin sensitivity and this association was independent of age, sex and BMI. Other metabolites such as linoleoyl- glycerophosphocholine (L-GPC), glycine, and creatine were also highly correlated with insulin sensitivity. Using 26 metabolites from this study, the group went on to develop a model called Quantose algorithm to predict insulin resistance. Fasting insulin, α-hydroxybutyrate, L- GPC and oleate levels were included in this model. Quantose IR as a fasting surrogate of insulin sensitivity was superior to other simple surrogate measures and was able to predict the progression from normal glucose tolerance to impaired glucose tolerance (75). Branched chain amino acids (BCAAs) were found to significantly increase in obese compared to lean subjects and a BCAA based index correlated with HOMA (76). The elevation of BCAA in subjects with impaired fasting glucose and diabetes has been confirmed in subsequent studies (77).

 

Lipoprotein insulin resistance score (LPIR) is a novel metabolomic biomarker based on nuclear magnetic resonance (NMR) quantification of lipoprotein levels and sizes. This index has been shown to predict future type 2 diabetes mellitus is some cohorts (78). LPIR is derived from the weighted score of six lipoproteins (VLDL, LDL, and HDL sizes and concentrations) that are more strongly related to IR than each of its individual subclasses (79). A risk score of between0-100 is estimated, with a score of 100 denoting being most insulin resistant. These metabolomic studies are promising since they can measure hundreds of metabolites in a very small sample. However, the pricing, technology, and access, precludes its use clinically. Further studies using this approach are necessary in larger more heterogeneous cohorts to replicate and validate surrogate insulin resistance markers derived through metabolomics

 

Table 1. Methods for Assessing Insulin Sensitivity and Resistance in Humans

Method

Measure of Insulin sensitivity

Direct Measures

Hyperinsulinemic Euglycemic Glucose Clamp

Average glucose infusion rate (GIR) = glucose disposal rate (M). SIClamp = M/(G x ΔI), where M is normalized for G (steady-state blood glucose concentration) and ΔI (difference between fastingand steady-state plasma insulin concentrations)

Insulin-suppression Test (IST)

Steady-state plasma glucose (SSPG) concentrations duringconstant infusions of insulin and glucose with suppressed endogenous insulin secretion

Indirect Measures

Minimal Model Analysis of Frequently Sampled IntravenousGlucose Tolerance Test (FSIVGTT)

Minimal model uniquely identifies model parameters thatdetermine a best fit to glucose disappearance during the modified FSIVGTT. SI : fractional glucose disappearance per insulin concentration unit; SG (glucose effectiveness): ability of glucose per se to promote its own disposal and inhibit HGP in the absenceof an incremental insulin effect (i.e., when insulin is at basal levels).

Simple Surrogate Indexes

Surrogates Derived from Fasting Steady-state Conditions

The Homeostasis Model Assessment (HOMA)

HOMA-IR = [(Fasting Insulin (µU/mL)) X (Fasting Glucose(mmol/L))]/22.5

Quantitative Insulin Sensitivity Check Index (QUICKI)

QUICKI = 1/[Log (Fasting Insulin, µU/ml) + Log (Fasting Glucose,mg/dl)]

Surrogates Derived from Dynamic Tests (OGTT)

Matsuda Index

ISI(Matsuda) = 10000/√[(Gfasting (mg/dl) x Ifasting (µU/ml) x(Gmean x Imean)]

Gutt Index - ISI (0, 120) (mg.l2.mmol-1.mIU-1.min-1)

ISI (0, 120) = 75000 + (G0-G120)(mg/l) x 0.19 x BW / 120 xGmean (0,

120min) (mmol/l) x Log [Imean (0, 120min)](mU/l)

Gmean, mean plasma glucose concentration during OGTT; Go, plasma glucose concentration during fasting; G120, plasma glucose concentration at 120 min; Gmean, mean plasma glucose concentration during OGTT; Imean, mean insulin concentration during OGTT; Io, plasma insulin concentration during fasting; I120, plasma insulin concentration at 120 min.

 

SUMMARY

 

In this chapter we have discussed a representative variety of methods currently available for estimating insulin sensitivity/resistance (but this is by no means an exhaustive review) (Table 1). These range from complex, timeconsuming, labor-intensive, invasive procedures to simple tests involving a single fasting blood sample. It is important to understand the physiological concepts informing each method so that relative merits and limitations of particular approaches are appropriately matched with proposed applications and data is interpreted correctly. The glucose clamp method is the reference standard for direct measurement of insulin sensitivity. Regarding simple surrogates, QUICKI and Log (HOMA) are among the best and most extensively validated. Dynamic tests are useful if information about both insulin secretion and insulin action are needed.

 

ACKNOWLEDGEMENTS

 

This work was supported by the Intramural Research Program, NIDDK, NIH

 

REFERENCES

 

  1. Reaven, G.M., The insulin resistance syndrome: definition and dietary approaches to treatment. Annu Rev Nutr, 2005. 25: p. 391-406.
  2. Petersen, K.F., et al., The role of skeletal muscle insulin resistance in the pathogenesis of the metabolic syndrome. Proc Natl Acad Sci U S A, 2007. 104(31): p. 12587-94.
  3. DeFronzo, R.A. and E. Ferrannini, Insulin resistance. A multifaceted syndrome responsible for NIDDM, obesity, hypertension, dyslipidemia, and atherosclerotic cardiovascular disease. Diabetes Care, 1991. 14(3): p. 173-94.
  4. Poirier, P., et al., Obesity and cardiovascular disease: pathophysiology, evaluation, and effect of weight loss: an update of the 1997 American Heart Association Scientific Statement on Obesity and Heart Disease from the Obesity Committee of the Council on Nutrition, Physical Activity, and Metabolism. Circulation, 2006. 113(6): p. 898-918.
  5. Muniyappa, R., et al., Current approaches for assessing insulin sensitivity and resistance in vivo: advantages, limitations, and appropriate usage. Am J Physiol Endocrinol Metab, 2008. 294(1): p. E15-26.
  6. Accili, D., Lilly lecture 2003: the struggle for mastery in insulin action: from triumvirate to republic. Diabetes, 2004. 53(7): p. 1633-42.
  7. Prodi, E. and S. Obici, Minireview: the brain as a molecular target for diabetic therapy. Endocrinology, 2006. 147(6): p. 2664-9.
  8. Muniyappa, R., et al., Cardiovascular actions of insulin. Endocr Rev, 2007. 28(5): p. 463-91.
  9. Kahn, S.E., R.L. Hull, and K.M. Utzschneider, Mechanisms linking obesity to insulin resistance and type 2 diabetes. Nature, 2006. 444(7121): p. 840-6.
  10. Himsworth, H.P., Diabetes mellitus: its differentiation into insulin-sensitive and insulin-insensitive types. 1936. Int J Epidemiol, 2013. 42(6): p. 1594-8.
  11. DeFronzo, R.A., J.D. Tobin, and R. Andres, Glucose clamp technique: a method for quantifying insulin secretion and resistance. Am J Physiol, 1979. 237(3): p. E214-23.
  12. Katz, A., et al., Quantitative insulin sensitivity check index: a simple, accurate method for assessing insulin sensitivity in humans. J Clin Endocrinol Metab, 2000. 85(7): p. 2402-10.
  13. Chen, H., G. Sullivan, and M.J. Quon, Assessing the predictive accuracy of QUICKI as a surrogate index for insulin sensitivity using a calibration model. Diabetes, 2005. 54(7): p. 1914-25.
  14. Rizza, R.A., L.J. Mandarino, and J.E. Gerich, Dose- response characteristics for effects of insulin on production and utilization of glucose in man. Am J Physiol, 1981. 240(6): p. E630-9.
  15. Finegood, D.T., R.N. Bergman, and M. Vranic, Estimation of endogenous glucose production during hyperinsulinemic-euglycemic glucose clamps. Comparison of unlabeled and labeled exogenous glucose infusates. Diabetes, 1987. 36(8): p. 914-24.
  16. McMahon, M.M., et al., Underestimation of glucose turnover measured with 6-3H- and 6,6-2H- but not 6-14Cglucose during hyperinsulinemia in humans. Diabetes, 1989. 38(1): p. 97-107.
  17. Radziuk, J. and S. Pye, Quantitation of basal endogenous glucose production in Type II diabetes: importance of the volume of distribution. Diabetologia, 2002. 45(8): p. 1053- 84.
  18. Sondergaard, E. and M.D. Jensen, Quantification of adipose tissue insulin sensitivity. J Investig Med, 2016. 64(5): p. 989-91.
  19. Shen, S.W., G.M. Reaven, and J.W. Farquhar, Comparison of impedance to insulin-mediated glucose uptake in normal subjects and in subjects with latent diabetes. J Clin Invest, 1970. 49(12): p. 2151-60.
  20. Harano, Y., et al., Glucose, insulin, and somatostatin infusion for the determination of insulin sensitivity in vivo. Metabolism, 1978. 27(9 Suppl 1): p. 1449-52.
  21. Pei, D., et al., Evaluation of octreotide to assess insulin- mediated glucose disposal by the insulin suppression test. Diabetologia, 1994. 37(8): p. 843-5.
  22. Greenfield, M.S., et al., Assessment of insulin resistance with the insulin suppression test and the euglycemic clamp. Diabetes, 1981. 30(5): p. 387-92.
  23. Mimura, A., et al., Insulin sensitivity test using a somatostatin analogue, octreotide (Sandostatin). Horm Metab Res, 1994. 26(4): p. 184-7.
  24. Yip, J., F.S. Facchini, and G.M. Reaven, Resistance to insulin-mediated glucose disposal as a predictor of cardiovascular disease. J Clin Endocrinol Metab, 1998. 83(8): p. 2773-6.
  25. Facchini, F.S., et al., Insulin resistance as a predictor of age-related diseases. J Clin Endocrinol Metab, 2001. 86(8): p. 3574-8.
  26. Yeni-Komshian, H., et al., Relationship between several surrogate estimates of insulin resistance and quantification of insulin-mediated glucose disposal in 490 healthy nondiabetic volunteers. Diabetes Care, 2000. 23(2): p. 171-5.
  27. Bergman, R.N., et al., Equivalence of the insulin sensitivity index in man derived by the minimal model method and the euglycemic glucose clamp. J Clin Invest, 1987. 79(3): p. 790-800.
  28. Finegood, D.T., I.M. Hramiak, and J. Dupre, A modified protocol for estimation of insulin sensitivity with the minimal model of glucose kinetics in patients with insulin-dependent diabetes. J Clin Endocrinol Metab, 1990. 70(6): p. 1538-49.
  29. Saad, M.F., et al., Differences between the tolbutamide- boosted and the insulin-modified minimal model protocols. Diabetes, 1997. 46(7): p. 1167-71.
  30. Quon, M.J., et al., Direct comparison of standard and insulin modified protocols for minimal model estimation of insulin sensitivity in normal subjects. Diabetes Res, 1994. 25(4): p. 139- 49.
  31. Yang, Y.J., J.H. Youn, and R.N. Bergman, Modified protocols improve insulin sensitivity estimation using the minimal model. Am J Physiol, 1987. 253(6 Pt 1): p. E595-602.
  32. Beard, J.C., et al., The insulin sensitivity index in nondiabetic man. Correlation between clamp-derived and IVGTT- derived values. Diabetes, 1986. 35(3): p. 362-9.
  33. Stefanovski, D., et al., Insulin Action, Glucose Homeostasis and Free Fatty Acid Metabolism: Insights From a Novel Model. Front Endocrinol (Lausanne), 2021. 12: p. 625701.
  34. Martin, B.C., et al., Role of glucose and insulin resistance in development of type 2 diabetes mellitus: results of a 25-year follow-up study. Lancet, 1992. 340(8825): p. 925-9.
  35. Howard, G., et al., Insulin sensitivity and atherosclerosis. The Insulin Resistance Atherosclerosis Study (IRAS) Investigators. Circulation, 1996. 93(10): p. 1809-17.
  36. American Diabetes, A., Diagnosis and classification of diabetes mellitus. Diabetes Care, 2007. 30 Suppl 1: p. S42-7.
  37. Cobelli, C., et al., Estimation of insulin sensitivity and glucose clearance from minimal model: new insights from labeled IVGTT. Am J Physiol, 1986. 250(5 Pt 1): p. E591-8.
  38. Toffolo, G. and C. Cobelli, The hot IVGTT two- compartment minimal model: an improved version. Am J Physiol Endocrinol Metab, 2003. 284(2): p. E317-21.
  39. Krudys, K.M., et al., Integrated model of hepatic and peripheral glucose regulation for estimation of endogenous glucose production during the hot IVGTT. Am J Physiol Endocrinol Metab, 2005. 288(5): p. E1038-46.
  40. Cobelli, C., et al., The oral minimal model method. Diabetes, 2014. 63(4): p. 1203-13.
  41. Dalla Man, C., et al., Insulin sensitivity by oral glucose minimal models: validation against clamp. Am J Physiol Endocrinol Metab, 2005. 289(6): p. E954-9.
  42. Dube, S., et al., Assessment of insulin action on carbohydrate metabolism: physiological and non-physiological methods. Diabet Med, 2013. 30(6): p. 664-70.
  43. Matthews, D.R., et al., Homeostasis model assessment: insulin resistance and beta-cell function from fasting plasma glucose and insulin concentrations in man. Diabetologia, 1985. 28(7): p. 412-9.
  44. Levy, J.C., D.R. Matthews, and M.P. Hermans, Correct homeostasis model assessment (HOMA) evaluation uses the computer program. Diabetes Care, 1998. 21(12): p. 2191-2.
  45. Wallace, T.M., J.C. Levy, and D.R. Matthews, Use and abuse of HOMA modeling. Diabetes Care, 2004. 27(6): p. 1487- 95.
  46. Haffner, S.M., H. Miettinen, and M.P. Stern, The homeostasis model in the San Antonio Heart Study. Diabetes Care, 1997. 20(7): p. 1087-92.
  47. Dabelea, D., et al., Birth weight, type 2 diabetes, and insulin resistance in Pima Indian children and young adults. Diabetes Care, 1999. 22(6): p. 944-50.
  48. Chen, H., et al., QUICKI is a useful index of insulin sensitivity in subjects with hypertension. Am J Physiol Endocrinol Metab, 2003. 284(4): p. E804-12.
  49. Mather, K.J., et al., Repeatability characteristics of simple indices of insulin resistance: implications for research applications. J Clin Endocrinol Metab, 2001. 86(11): p. 5457-64.
  50. Hanley, A.J., et al., Prediction of type 2 diabetes using simple measures of insulin resistance: combined results from the San Antonio Heart Study, the Mexico City Diabetes Study, and the Insulin Resistance Atherosclerosis Study. Diabetes, 2003. 52(2): p. 463-9.
  51. Bastard, J.P., et al., Is quantitative insulin sensitivity check index, a fair insulin sensitivity index in humans? Diabetes Metab, 2001. 27(1): p. 69-70.
  52. Yokoyama, H., et al., Quantitative insulin sensitivity check index and the reciprocal index of homeostasis model assessment are useful indexes of insulin resistance in type 2 diabetic patients with wide range of fasting plasma glucose. J Clin Endocrinol Metab, 2004. 89(3): p. 1481-4.
  53. Skrha, J., et al., Comparison of the insulin action parameters from hyperinsulinemic clamps with homeostasis model assessment and QUICKI indexes in subjects with different endocrine disorders. J Clin Endocrinol Metab, 2004. 89(1): p. 135-41.
  54. Rabasa-Lhoret, R., et al., Modified quantitative insulin sensitivity check index is better correlated to hyperinsulinemic glucose clamp than other fasting-based index of insulin sensitivity in different insulin-resistant states. J Clin Endocrinol Metab, 2003. 88(10): p. 4917-23.
  55. Katsuki, A., et al., QUICKI is useful for following improvements in insulin sensitivity after therapy in patients with type 2 diabetes mellitus. J Clin Endocrinol Metab, 2002. 87(6): p. 2906-8.
  56. Uwaifo, G.I., et al., Indices of insulin action, disposal, and secretion derived from fasting samples and clamps in normal glucose-tolerant black and white children. Diabetes Care, 2002. 25(11): p. 2081-7.
  57. Sondergaard, E., et al., How to Measure Adipose Tissue Insulin Sensitivity. J Clin Endocrinol Metab, 2017. 102(4): p. 1193-1199.
  58. Matsuda, M. and R.A. DeFronzo, Insulin sensitivity indices obtained from oral glucose tolerance testing: comparison with the euglycemic insulin clamp. Diabetes Care, 1999. 22(9): p. 1462-70.
  59. Stumvoll, M., et al., Use of the oral glucose tolerance test to assess insulin release and insulin sensitivity. Diabetes Care, 2000. 23(3): p. 295-301.
  60. Avignon, A., et al., Assessment of insulin sensitivity from plasma insulin and glucose in the fasting or post oral glucose- load state. Int J Obes Relat Metab Disord, 1999. 23(5): p. 512-7.
  61. Mari, A., et al., A model-based method for assessing insulin sensitivity from the oral glucose tolerance test. Diabetes Care, 2001. 24(3): p. 539-48.
  62. Gutt, M., et al., Validation of the insulin sensitivity index (ISI(0,120)): comparison with other measures. Diabetes Res Clin Pract, 2000. 47(3): p. 177-84.
  63. Belfiore, F., S. Iannello, and G. Volpicelli, Insulin sensitivity indices calculated from basal and OGTT-induced insulin, glucose, and FFA levels. Mol Genet Metab, 1998. 63(2):
  64. 134-41.
  65. Cobelli, C., et al., Assessment of beta-cell function in humans, simultaneously with insulin sensitivity and hepatic extraction, from intravenous and oral glucose tests. Am J Physiol Endocrinol Metab, 2007. 293(1): p. E1-E15.
  66. Abdul-Ghani, M.A., et al., What is the best predictor of future type 2 diabetes? Diabetes Care, 2007. 30(6): p. 1544-8.
  67. Muniyappa, R., et al., Predictive Accuracy of Surrogate Indices for Hepatic and Skeletal Muscle Insulin Sensitivity. J Endocr Soc, 2019. 3(1): p. 108-118.
  68. Visentin, R., et al., Hepatic insulin sensitivity in healthy and prediabetic subjects: from a dual- to a single-tracer oral minimal model. Am J Physiol Endocrinol Metab, 2015. 309(2): p. E161-7.
  69. Hanson, R.L., et al., Evaluation of simple indices of insulin sensitivity and insulin secretion for use in epidemiologic studies. Am J Epidemiol, 2000. 151(2): p. 190-8.
  70. Kodama, K., et al., Ethnic differences in the relationship between insulin sensitivity and insulin response: a systematic review and meta-analysis. Diabetes Care, 2013. 36(6): p. 1789-96.
  71. Pisprasert, V., et al., Limitations in the use of indices using glucose and insulin levels to predict insulin sensitivity: impact of race and gender and superiority of the indices derived from oral glucose tolerance test in African Americans. Diabetes Care, 2013. 36(4): p. 845-53.
  72. Thompson, D.S., et al., Limitations of fasting indices in the measurement of insulin sensitivity in Afro-Caribbean adults. BMC Res Notes, 2014. 7: p. 98.
  73. Muniyappa, R., et al., Limited predictive ability of surrogate indices of insulin sensitivity/resistance in Asian-Indian men. Am J Physiol Endocrinol Metab, 2010. 299(6): p. E1106-12.
  74. Ha, J., et al., When MINMOD Artifactually Interprets Strong Insulin Secretion as Weak Insulin Action. Front Physiol, 2021. 12: p. 601894.
  75. Gall, W.E., et al., alpha-hydroxybutyrate is an early biomarker of insulin resistance and glucose intolerance in a nondiabetic population. PLoS One, 2010. 5(5): p. e10883.
  76. Cobb, J., et al., A novel fasting blood test for insulin resistance and prediabetes. J Diabetes Sci Technol, 2013. 7(1):p. 100-10.
  77. Newgard, C.B., et al., A branched-chain amino acid- related metabolic signature that differentiates obese and lean humans and contributes to insulin resistance. Cell Metab, 2009. 9(4): p. 311-26.
  78. Menni, C., et al., Biomarkers for type 2 diabetes and impaired fasting glucose using a nontargeted metabolomics approach. Diabetes, 2013. 62(12): p. 4270-6.
  79. Mackey, R.H., et al., Lipoprotein particles and incident type 2 diabetes in the multi-ethnic study of atherosclerosis. Diabetes Care, 2015. 38(4): p. 628-36.
  80. Shalaurova, I., et al., Lipoprotein insulin resistance index: a lipoprotein particle-derived measure of insulin resistance. Metab Syndr Relat Disord, 2014. 12(8): p. 422-9.
  81. Stern, S.E., et al., Identification of individuals with insulin resistance using routine clinical measurements. Diabetes, 2005. 54(2): p. 333-339.
  82. Tam, C.S., et al., Defining insulin resistance from hyperinsulinemic-euglycemic clamps. Diabetes Care, 2012. 35(7): p.1605-1610.

 

Physiology of GnRH and Gonadotropin Secretion

ABSTRACT

 

Gonadotropin hormone-releasing hormone (GnRH) is the key regulator of the reproductive axis.  Its pulsatile secretion determines the pattern of secretion of the gonadotropins, follicle stimulating hormone and luteinizing hormone, which then regulate both the endocrine function and gamete maturation in the gonads. Recent years have seen rapid developments in how GnRH secretion is regulated, with the discovery of the kisspeptin-neurokinin-dynorphin neuronal network in the hypothalamus. This mediates both positive and negative sex steroid feedback control of GnRH secretion, in conjunction with other neuropeptides and neurotransmitters. This chapter describes the main features of this regulatory system, including how its anatomical arrangements interact with functional control, and describes key differences between rodent and larger mammalian systems.

 

INTRODUCTION

 

Since the discovery of Gonadotropin Releasing Hormone (GnRH), an extensive body of literature has established it as the pivotal central regulator of human reproduction. However, the GnRH neuronal network, per se lacks the cellular machinery to fully integrate developmental, environmental, endocrine, and metabolic factors that influence its secretion. For example, GnRH neurons do not express the principal estrogen receptor alpha (ER-alpha), which is required for sex-steroid mediated control of gonadotropin secretion (1). Intermediate signaling pathways must therefore exist to mediate gonadal steroid feedback. Current evidence, accumulated since the discovery of Kisspeptin-Neurokinin B-Dynorphin (KNDy) neuronal network in the last decade, suggests a pivotal role for this network in the regulation of pulsatile GnRH secretion by integrating nutrient, endocrine, and environmental signals, and thus the control of downstream hypothalamic-pituitary-gonadal (HPG) axis.

 

The HPG axis anatomically comprises:

 

  1. The hypothalamus (especially the infundibular nucleus, the human homologue of the arcuate nucleus) where the KNDy and GnRH-producing neurons are located.
  2. The anterior pituitary, where Luteinizing Hormone (LH) and Follicle-Stimulating Hormone (FSH) are secreted by gonadotropes.
  3. The gonads, responsible for the production of both sex steroids and gametes, under the influences of LH and FSH.

 

As with other endocrine systems, positive and negative feedback regulate HPG axis (2,3). In this chapter, we have focused on human data. Where human data is limited, data from other species are leveraged.

 

GONADOTROPIN RELEASING HORMONE (GnRH) – THE PRINCIPAL REGULATOR OF REPRODUCTION

 

The Discovery of GnRH

 

GnRH was isolated from porcine hypothalami and structurally identified as a decapeptide (pGlu-His-Trp-Ser-Tyr-Gly-Leu-Arg-Pro-Gly·NH2) five decades ago (4-6). This decapeptide was shown to potently stimulate LH and FSH release from the pituitary in a number of mammalian species (6,7). Early literature referred to this peptide as the ‘Luteinizing Hormone-Releasing Hormone (LH-RH)’, but more recently, it is widely referenced as Gonadotropin Releasing Hormone (GnRH) -reflecting the stimulatory role on the secretion of both gonadotropins – i.e., LH and FSH (8).

 

Diverse forms of GnRH and its receptor exist among vertebrates, with over twenty primary structures across species, suggesting that GnRH system developed early in the evolutionary sequence (9,10). The GnRH structure was first identified in mammals and is therefore referred to as GnRH I (9,11).  Subsequently, another structurally different vertebrate GnRH sequence was first identified from chicken brain -this is now referred to as GnRH II (pGlu-His-Trp-Ser-His-Gly-Trp-Tyr-Pro-Gly-NH2) (10,12). A third form has also been described in fish - GnRH III (9,12). In mammals, hypophysiotropic functions are limited to GnRH I, therefore in the human context GnRH I is referred to as GnRH (13) and we will use this terminology for this review.

 

Neuroanatomy of GnRH Neurons

 

GnRH neurons originate in the medial olfactory placode during embryological development and migrate along the olfactory bulb to their final positions within the hypothalamus (14,15). A number of factors contributing to this GnRH neuron migratory process have been identified: anosmin-1 (the product of KAL gene) (16), neuropilins (17), leukemia inhibitory factor (18), fibroblast growth factor receptor 1 (19), fibroblast growth factor receptor 8 (20), polysialic form of neural adhesion molecule (PSA-NCAM) (21), among others (22). Defective GnRH migration leads to Kallmann syndrome, characterized by hypogonadotropic hypogonadism due to GnRH deficiency and anosmia (15). Mutations in prokineticin genes (PROK1 and PROK2) lead to hypogonadotropic hypogonadism without anosmia, suggesting that factors other than suboptimal migration can also lead to functional deficiencies in GnRH (15,23,24).

 

GnRH cell bodies are located in the medial preoptic area (POA) and in the arcuate/infundibular nucleus of the hypothalamus, forming a neuronal network with projections to the median eminence (25). GnRH is secreted from the median eminence into the fenestrated capillaries of portal circulation, carried to the anterior pituitary (25). In humans, the number of GnRH neurons has been estimated to range between 1000 to 1500 (9,14). The co-location of GnRH neurons with other central regulators allows the GnRH network to be influenced by a range of neuroendocrine and metabolic inputs (26).

 

GnRH Secretion and Pulsatility

 

Two distinct modes of GnRH secretion have been described: pulsatile and surge modes (26). Pulsatile mode refers to episodic release of GnRH, with distinct pulses of GnRH secretion into the portal circulation with undetectable GnRH concentrations between pulses. The surge mode of GnRH secretion occurs in females, during the pre-ovulatory phase, in which the presence of GnRH in the portal circulation appears to be persistent (26,27).

 

Direct pulsatile GnRH release was initially demonstrated in ovariectomized rhesus monkeys using serial samples of portal blood (28). Pulsatile pattern of GnRH secretion was demonstrated subsequently in humans through serial blood sampling during pituitary surgery (29). Abolishment of LH pulses by GnRH antisera (30,31) and its reestablishment with GnRH analogues (30) suggest that LH pulses are determined by the underlying GnRH pulsatility.  The LH pulsatility was first detected during an attempt to validate a radioimmunoassay to measure serum LH in rhesus monkeys, where marked variations in LH levels was noticed (32). Further studies confirmed the pulsatile nature of LH secretion (33-35). In women, the frequency and amplitude of LH pulses were noted to be dependent on the menstrual cycle phase, with pulses every 1 to 2 hours during the early follicular phase eventually merging into a continuous mid-cycle surge, and decreased pulse frequency to every 4 hours during the luteal phase (36). In humans, LH pulse frequency is used as a surrogate of GnRH pulsatility, as ethical considerations and technical challenges preclude sampling of hypophyseal blood or cerebrospinal fluid to measure GnRH concentrations directly (37,38).

 

The importance of GnRH pulsatility on LH and FSH secretion was first demonstrated in rhesus monkeys, where endogenous GnRH secretion was abolished by hypothalamic radiofrequency. Pulsatile GnRH reinstated gonadotropin secretion in these animals, whereas continuous GnRH only elicited a transient response. Moreover, the switch from continuous to pulsatile GnRH administration allowed recovery of gonadotropin secretion (39).

 

GnRH neurons coordinate their activity, but the precise mechanism of this remains unclear (27,40), and is the subject of continuing investigations. Episodic multi-unit electrical activity at medial basal hypothalami (MBH) is correlated with LH release, suggesting that ‘GnRH pulse generator’ is anatomically located at MBH – or closely linked to it neurohormonally(41,42). GnRH neurons show intrinsic electrical pulsatility. GnRH cell lines derived from mouse hypothalamic and fetal olfactory placode GnRH neurons both demonstrate intrinsic pulsatility in vitro (26,43,44). Functionally, the ‘GnRH pulse generator’ relies on complex relations between glutamatergic cells, GnRH and other neurons, and likely other elements are involved, of which the kisspeptin-neurokinin B-opioid pathway may have a pivotal intermediary role in the regulation of GnRH pulsatility (45).

 

Differential Regulation of LH and FSH

 

The stimulatory effects of GnRH on LH and FSH secretion are not identical (46).  FSH secretion is more irregular than LH in both humans and sheep, which is essentially related to the pulsatility and different stimulatory effects of GnRH, but other factors also might be relevant, such as differences in LH and FSH storage (more scarce for the FSH), existence of different gonadotropes subpopulations, or diverse response times to GnRH (47). In ovariectomized sheep administered GnRH antisera, pulsatile secretion of LH was completely inhibited (undetectable LH levels within 24 hours), while the FSH concentration fell more slowly and remained detectable (30). It has been estimated that 93% of the GnRH pulses were associated with FSH pulses and, unlike LH, a constitutive secretion of FSH appears to exist (48). The frequency of GnRH input has been demonstrated to selectively regulate gonadotropin subunit gene transcription: rapid GnRH pulse rates increase α and LH-β and slow GnRH pulse frequency increases FSH-β gene transcription (49-51). Moreover, with progressive increases in GnRH frequency (from one pulse every 120 to 60 min, from 60 to 30 min, and from 30 to 15 min) in GnRH deficient men, mean LH rose concurrently with a decrease in LH pulse amplitude, while FSH remained unchanged (52).

 

Biological and Clinical Relevance of GnRH Pulsatility

 

Appropriate modulation of LH pulse frequency is essential for pubertal maturation and reproductive function. In infancy, LH pulsatile secretion is increased (often termed mini-puberty), likely reflecting pulsatile GnRH secretion, but soon becomes quiescent (53). This pre-pubertal suppression of HPG axis has been shown to occur in agonadal humans (54)and primates (55), suggesting that hypothalamo-hypophyseal factors play a role in post-natal quiescence of the reproductive axis, until puberty sets in.

 

The onset of pubertal maturation is heralded by the development of a pattern of steady acceleration in LH pulsatility (56). In children, higher basal and GnRH stimulated LH concentrations are observed in early childhood (<5 years). This is subdued mid-childhood (5-11 years) and increase thereafter with pubertal development (54,57).  Conceptually, an abnormal reactivation of GnRH pulse frequency is the central mechanism associated with precocious or delayed puberty (14).

 

In women, the pattern of GnRH secretion is essential for the regulation of the menstrual cycle (Figure 1) (58,59). LH pulse frequency is slow in the luteal phase, and increasingly speeds up during the follicular and the pre-ovulatory phases, presumably reflecting changes in GnRH pulse frequency (60). Abnormalities in GnRH - and hence LH pulse frequency - are associated with a number of reproductive endocrine disorders. In hypothalamic amenorrhea, a condition associated with anovulatory amenorrhea and hypoestrogenism, LH pulse frequency (and by inference GnRH) is lower than expected for the prevailing steroid profile and is comparable to luteal phase pulsatility (37). LH pulse frequency in hyperprolactinemic women is also lower than in healthy women, requiring dopaminergic agonist preparations, such as bromocriptine to regulate prolactin secretion and restore LH pulse frequency (38). In polycystic ovary syndrome LH pulse frequency and amplitude are higher throughout the menstrual cycle in comparison to that observed in healthy women, contributing to chronic anovulation (61-64).

 

Figure 1. Hormonal oscillations through the menstrual cycle. In the early follicular phase of the menstrual cycle, the initial increase in FSH stimulates follicular recruitment and maturation. The consequent secretion of estradiol (E2) selectively inhibits FSH release (needed for selection of the dominant follicle) and maintains rapid GnRH pulsatility during the late follicular phase. The persistent rapid GnRH pulses increase LH, which further stimulates E2 secretion, culminating in positive E2 feedback to produce the mid-cycle LH surge. During the LH surge, GnRH levels appear to be consistently elevated and remain elevated as LH declines, suggesting that the frequency of GnRH pulse has become very rapid or continuous, which results in desensitization of LH secretion (possibly the mechanism to terminate the LH surge). After ovulation, luteinization of the ruptured follicle results in progesterone secretion which reduces the frequency of GnRH pulses. With the demise of corpus luteum, E2, progesterone and inhibin levels fall, and the GnRH pulse frequency increases, leading to follicular maturation in the next cycle. (Adapted from: Marshall JC, Dalkin AC, Haisenleder DJ, Paul SJ, Ortolano GA, Kelch RP. Gonadotropin-releasing hormone pulses: regulators of gonadotropin synthesis and ovulatory cycles. Recent Prog Horm Res. 1991;47:155-187).

 

NEURONAL REGULATION OF GnRH SECRETION: THE KISSPEPTIN-NEUROKININ B-DYNORPHIN (KNDy) NEURONAL NETWORK

 

Whilst the central role attributed to GnRH remains undisputed, its effective function requires input from other neuronal networks. For instance, the absence of estrogen receptor alpha (ER-alpha) expression on GnRH neurons suggests the need for an intermediate signaling pathway to mediate gonadal steroid feedback (1). The discovery of kisspeptin signaling in neuroendocrine regulation of human reproduction revolutionized the current understanding of the HPG axis. Kisspeptin signaling pathway is increasingly recognized as essential for normal puberty, gonadotropin secretion, and regulation of reproduction (65-67). Other relevant kisspeptin roles have been identified such as regulation of sexual and social behavior, emotional brain processing, mood, audition, olfaction, metabolism, body composition, cardiac function, among others (68-74).

 

Discovery of KNDy Neuronal Network

 

KiSS1, the gene encoding kisspeptins, was first described in 1996 as a suppressor of metastasis in human malignant melanoma (75,76). This gene was discovered in Hershey and named in accordance with the famous chocolates ‘Hershey’s Kisses’; the inclusion of ‘SS’ is indicative of ‘suppressor sequence’. The KiSS1 gene maps to chromosome 1q32 and includes four exons of which the first two are not translated (77). The gene encodes the precursor 145 amino acid peptide, which is cleaved down to a 54 amino acid peptide. This peptide can be truncated to 14, 13 and 10 amino-acid peptides, all sharing the C-terminal sequence (78,79). These peptides are collectively referred to as kisspeptins - and Kp-10, Kp-13, Kp-14 and Kp-54 are suggested abbreviations for human kisspeptins (80). In 2001, kisspeptins were identified as ligands for the orphan G–protein receptor 54 (GPR54) (81-83), currently named KISS1R (80). KISS1R is localized to human chromosome 19p13.3 and it has five exons, encoding a 398-amino acid protein with seven trans-membrane domains (79,82). Upon binding by kisspeptin, KISS1R activates phospholipase C and recruits intracellular messengers, inositol triphosphate and diacylglycerol, which in turn lead to the release of calcium and activation of protein kinase C (82-84).

 

A reproductive role for kisspeptin in humans became apparent from patients with pubertal disorders which were associated with KISS1R mutations (85-87). A number of inactivating mutations of Kiss1 and Kiss1r have since been reported in animal models with phenotypes characterized by pubertal delay (88). An activating mutation in KISS1R has been described in a girl with precocious puberty: when compared to cells with wild-type transfected GPR54, cells with this mutation showed prolonged inositol phosphate accumulation and phosphorylation of extracellular signal–regulated kinase, suggesting extended activation of intracellular signaling by the mutant GPR54 (89). Missense mutations have also been reported in KISS1 gene in three unrelated children with central precocious puberty (90). Functional studies of these mutant peptides demonstrated higher resistance to in vitro degradation but normal affinity to KISS1R, thus suggestive of increased bioavailability as the mechanism by which these abnormal kisspeptins induce precocious puberty (90).

 

Recently in an Asian cohort, potentially regulatory polymorphisms, as rs5780218 and rs12998, in KiSS1 gene were significantly associated to genetic susceptibility to central precocious puberty in Chinese girls by single-locus analysis (91). Nevertheless, these findings are inconsistently reported in literature and require additional validation in functional studies.

 

A role for neurokinin B in the hypothalamic regulation was also demonstrated when genetic studies in patients from consanguineous families with hypogonadotropic hypogonadism were found to have missense mutations in TAC3 (encodes neurokinin B) and TACR3 (encodes neurokinin B receptor) (92). Other cases have been reported since (93-96).

 

There is also long-standing evidence for the role of opioid systems in reproduction. In 1980, Wilkes reported the localization of β endorphin in the human hypothalamus (97). Studies involving the administration of naloxone and naltrexone (opioid antagonists) to humans showed stimulatory effects on LH secretion (98,99), and other studies supported the notion that endogenous opioids play a role in the control of HPG axis (100-104). In 2007, it was demonstrated that dynorphin and kisspeptin are co-localized along with neurokinin B in the same hypothalamic neuronal population in sheep, therefore termed KNDy (Kisspeptin-Neurokinin B-Dynorphin) neurons, highlighting the possible interconnection between these neuropeptides in the control of GnRH and gonadotropin secretion (105-107). The co-localization of kisspeptin, neurokinin B and dynorphin has also been demonstrated in humans (108).

 

Kisspeptin neurons have also other important neuroanatomical relationships, such as with neuronal nitric oxide synthase neurons as demonstrated in prepubertal female sheep (109), or with somatostatin neurons in the rat hypothalamus (110).

 

Neuroanatomy of KNDy Neuronal Network

 

The location of kisspeptin neurons is different between rodents and human species. In humans, kisspeptin neurons are distributed in the rostral Pre-optic Area (POA) and in the infundibular nucleus in the hypothalamus (Figure 2) (108,111). In both male and female autopsy samples, the majority of kisspeptin cell bodies are identified in the infundibular nucleus, and a second dense population of kisspeptin neurons in the rostral POA (108). The infundibular nucleus (arcuate nucleus in non-human species) is similar across species, but the rostral region is more species specific (108,112,113). In rodents, the rostral population is located in the anteroventral periventricular nucleus (AVPV) and the periventricular nucleus (PeN), the continuum of this region named as the rostral periventricular region of the third ventricle (RP3V) (112,114). Humans and ruminants lack this well-defined RP3V population of kisspeptin neurons, which are more scattered within the preoptic region (113,115).

 

Kisspeptin axons form dense plexuses in the human infundibular stalk, where the secretion of GnRH occurs (108). Axo-somatic, axo-dendritic, and axo-axonal contacts between kisspeptin and GnRH axons were demonstrated at this level, showing that kisspeptin and GnRH networks are in close proximity (108,116). Moreover, GnRH neurons express Kiss1rmRNA, reinforcing the notion of kisspeptin involvement in GnRH secretion (117-119).

 

Figure 2. Neuroanatomy of kisspeptin-GnRH pathway and the control of HPG axis in humans and rodents. Kisspeptin signals directly to GnRH neurons, which express KISS1R. The location of kisspeptin neurons within the hypothalamus is species specific, residing within the anteroventral periventricular nucleus (AVPV) and the arcuate nucleus in rodents, and within the preoptic area (POA) and the infundibular nucleus in humans. Kisspeptin neurons in the infundibular nucleus (humans)/arcuate nucleus (rodents) co-express neurokinin B and dynorphin (KNDy neurons), which autosynaptically regulate kisspeptin secretion (via neurokinin B receptor and kappa opioid peptide receptor). In humans, infundibular KNDy neurons relay negative (red) and positive (green) feedback, whereas in rodents the negative and positive steroid feedback are mediated via arcuate nucleus and AVPV respectively. The role of human POA kisspeptin neurons in sex steroid feedback is not yet clear. (Adapted from: Skorupskaite K, George JT, Anderson RA. The kisspeptin-GnRH pathway in human reproductive health and disease. Human Reproduction Update. 2014;20:485-500).

 

Three-quarters of kisspeptin-immunoreactive cells in the human infundibular nucleus of the hypothalamus co-express neurokinin B and dynorphin (KNDy neurons) (108,120). KNDy neurons in rodents and ruminants are localized in the arcuate nucleus of the hypothalamus. However, neurokinin B and dynorphin are absent from kisspeptin neurons in the hypothalamic POA (Figure 2) (67,115). This differential expression of neuropeptides may reflect distinct functions of these two kisspeptin populations with kisspeptin neurons in the AVPV acting as LH surge generators, while those in the ARC (including KNDy neurons) acting as LH pulse generators.

 

Significant kisspeptin expression was also demonstrated in central extra-hypothalamic sites, including in limbic and paralimbic brain regions, such as medial amygdala, cingulate, globus pallidus, hippocampus, putamen and thalamus, key areas of neurobiological control of sexual and emotional behaviors (reviewed in detail in (121)), as well as peripherally in organs like ovary, testis, uterus and placenta where the kisspeptin system may also play a part in reproduction function (122,123).

 

Apart from reproductive and central kisspeptin expression, the kisspeptin signaling system has been demonstrated in several peripheral tissues, namely, in pancreas (involved in glucose-stimulated insulin secretion); in endothelial cells of different vascular beds as coronary artery, aorta and umbilical vein (triggering vasoconstriction); in the kidney, namely, in tubular cells, collecting duct cells and vascular smooth muscle cells (involved in function and renal morphogenesis); as well as in bone, fat and liver tissue (78,124,125).

 

Interactions Between Kisspeptin, Neurokinin B and Dynorphin

 

KDNy neurons act synergistically to induce coordinated and pulsatile GnRH secretion by regulating the neuroactivity of other KDNy cells. This is supported by the existence of neurokinin B and kappa opioid peptide receptors (receptor for dynorphin) within the KNDy cells, but not kisspeptin receptors, which are predominantly expressed on GnRH neurons (107,120,126). Neuron-neuron and neuron-glia communications via gap junctions contribute for the synchronized activities among KNDy neurons (127).  

 

Neurokinins (A and B) are members of the tachykinin family of peptides, which stimulate three related GPCRs (encoded by TACR1, TACR2 and TACR3) (128) This family also contains substance P, neuropeptide K, neuropeptide γ, hemokinin-1, and more recently endokinins. Neurokinin B acts predominantly on TACR3. Neurokinin B increases the membrane potential of KNDy neurons, leading to an increase in KNDy neuron pulsatile activity which, in turn, will promote the secretion of kisspeptin leading, ultimately, to GnRH secretion (67,129,130). Neurokinin B signaling regulates GnRH/LH secretion in healthy women, and it is crucial for the mediation of the estrogenic positive and negative feedback on LH secretion (131-133). There is rapidly increasing interest in the therapeutic value of neurokinin antagonists in several indications in reproductive health, recently reviewed in (134).

 

In women with polycystic ovary syndrome, the relationship between kisspeptin and gonadotropin levels has been widely explored in those with anovulatory cycles (135-138). Most studies have shown higher serum levels of kisspeptin and LH when the oligomenorrhea phenotype is present, despite the high heterogeneity observed. In this context, potential treatments targeting neuroendocrine dysfunction emerged. The administration of neurokinin 3 receptor antagonists markedly reduced serum LH concentration and pulse frequency, as well as serum testosterone (139-141). A recent study confirmed a complex crosstalk between neurokinin B and kisspeptin pathways in the regulation of GnRH secretion in polycystic ovary syndrome. In this study, kisspeptin-10 infusion given to women with polycystic ovary syndrome increased LH secretion with a direct relationship to estradiol exposure. Neurokinin 3 receptor antagonism reduced LH secretion and pulsatility, and whilst LH response to kisspeptin-10 was preserved, its relationship with circulating estradiol was not. More interestingly, although kisspeptin-10 increased LH pulse frequency, changes in other parameters of LH secretory pattern were prevented when co-administered with neurokinin 3 receptor antagonists (141).

 

In postmenopausal women,  seven day treatment with neurokinin 3 receptor antagonist decreased LH secretion, but not FSH secretion, as well as lead to a remarkable reduction in hot flushes (142). Neurokinin 3 receptor antagonism efficiency in treating menopausal hot flushes has been also demonstrated in other clinical trials (143,144). Fezolinetant administrated in a single daily dose regimen (30 or 45mg/daily) for treatment of moderate to severe vasomotor symptoms reduced these symptoms by over 50% from baseline within the first week and persistently during the 52-week treatment period, and is now approved for the treatment of menopausal vasomotor symptoms (145-148).

 

A second comparable drug, elinzanetant, which differs in its pharmacology in that it is an antagonist at the NK1 as well as NK3 receptor, has also been recently demonstrated to reduce the severity and frequency of moderate-to-severe vasomotor symptoms and also to improve sleep quality and menopause-related quality of life (149). The importance of NK1 receptor antagonism in these effects is unclear.

 

In healthy men, neurokinin B signaling display a central role for the reproductive function, and this is functionally upstream of kisspeptin-mediated GnRH secretion: LH, FSH and testosterone secretion decreased during the administration of a neurokinin 3 receptor antagonist, while kisspeptin-10 administration restored LH secretion to the same degree before and during neurokinin 3 receptor antagonist treatment (150).

 

An increase in the expression of Kiss1 in the hypothalamic neurons was observed following senktide (agonist of neurokinin B) administration (151), and its stimulatory effects were abolished in Gpr54 knock-out male (152). In ovariectomized goats, neurokinin B stimulated LH secretion through electrical multi-unit activity corresponded to LH secretion, suggesting a hypothalamic site for this GnRH pulse generation (153). GnRH antagonists abolished the stimulatory effect of neurokinin B, demonstrating its site of action to be functionally higher than the GnRH receptor (154,155).

 

Dynorphins act as the decelerator that inhibits KNDy neurons pulsatility. Studies involving the administration of opioid antagonists to humans have shown stimulatory effects on LH secretion in late follicular and mid-luteal phase (98,99), and together with other studies (100-104), highlight the inhibitory input by dynorphins on kisspeptin signalling, and consequently on GnRH/gonadotropin secretion. Through the stimulatory effects of neurokinin B and kisspeptin, and the inhibitory action of dynorphin, these neuropeptides coordinate pulsatile GnRH and LH secretion (Figure 2) (156,157).

 

Kisspeptin-mediated GnRH secretion is sex steroid dependent. Estrogen and progesterone directly modulate kisspeptin activity though the sex-steroid receptors expressed on kisspeptin neurons at both AVPV and the arcuate nucleus (158-160). Furthermore, two distinct populations of kisspeptin neurons, the infundibular/arcuate region of which interacts with neurokinin B and dynorphin, appear to mediate distinct sex-steroid pathways (discussed in more detail in sections 4.1-4.4).

 

Briefly, in humans, KNDy neurons in the infundibular nucleus alone are involved in negative and positive sex-steroid feedback, whereas in rodents positive sex-steroid feedback seems to be mediated via kisspeptin neurons in the AVPV region and negative sex-steroid feedback via the arcuate KNDy neurons (Figure 2) (67,111,160,161).

 

Stimulatory Effect of Kisspeptin on GnRH and Gonadotropin Secretion

 

Kisspeptin is a potent stimulator of the HPG axis – and in fact, it is the most potent GnRH secretagogue currently known. Kisspeptin signals directly to the hypothalamic GnRH neurons via kisspeptin receptor to release GnRH into the portal circulation, which in turn stimulates the anterior pituitary gonadotropes to produce LH and FSH (129,162).

 

The stimulatory effects of kisspeptin on LH secretion have been documented in animal models (163-166). This is consistent with human studies, where kisspeptin increases both LH and FSH secretion with the preferential stimulatory effect on the former (67,167-177). Kissppetin-54 was first administered in healthy men as an intravenous infusion with dose-dependent rise in LH secretion (169). Since then kisspeptin was administered in different isoforms (kisspeptin-54 and kisspeptin-10), different routes (subcutaneous and intravenous), different types of exposure (continuous and bolus), to healthy men and women and in endocrine disease models with low gonadotropin output, all showing stimulatory effect of kisspeptin on LH secretion (fully reviewed in (67)).

 

Pulsatile GnRH secretion correlates with LH pulsatility, prompting investigation of the effect of kisspeptin on regulating LH pulse frequency. LH pulse frequency and amplitude were increased following intravenous infusion of kisspeptin-10 in healthy men (172), and subcutaneous bolus of kisspeptin-54 in healthy women (174). The hypothalamic response to kisspeptin-54 and the pituitary response to GnRH are preserved in healthy older men (178). Kisspeptin also stimulates LH pulse frequency in reproductive endocrine disorders of low LH pulsatility, including hypothalamic amenorrhea, defects in the neurokinin B pathway and hypogonadal men with diabetes (96,179,180). Indeed, kisspeptin-54 and kisspeptin-10, as well as kisspeptin agonists like MVT-602 (previously known as TAK-448) are able to stimulate physiological reproductive hormone secretion in individuals with functional hypogonadism related to deficient GnRH secretion, such as in hypothalamic amenorrhea or polycystic ovary syndrome (181,182).

 

In addition, recent findings have explored further the effects of MVT-602. LH concentration increased in a dose-dependent manner, resembling the amplitude and duration found in the physiological mid-cycle LH surge, proving to be safe and well tolerated throughout the dose range (0.3 – 3.0 mg) (183). This approach to mimicking the physiological response during oocyte maturation and ovulation may have clinical utility for women during medically assisted reproduction.

 

Kisspeptin regulates GnRH and subsequently gonadotropin secretion through Kiss1r, as suggested by Messager who demonstrated no detectable LH levels in response to kisspeptin in Kiss1r knockout mice (119). The prevention of the stimulatory effect of kisspeptin on LH secretion by GnRH antagonists indicate that kisspeptin action is GnRH-mediated (118,164,184-186). This is further supported by the observation that kisspeptin cause depolarization of GnRH neurons (117) and stimulate GnRH release from hypothalamic explants (187,188). The expression of GnRH mRNA is upregulated in GnRH neurons following kisspeptin administration (189). Moreover, in patients with impaired functional capacity of GnRH neurons (idiopathic hypogonadotropic hypogonadism), the same dose of kisspeptin failed to induce LH response seen in healthy men and women (190). In female rats, ablation of KNDy neurons resulted in hypogonadotropic hypogonadism, confirming its role in the maintenance of normal LH levels and to estrous cyclicity (191).

 

Some investigators have demonstrated a direct stimulatory effect of kisspeptin on gonadotropes, but this direct stimulatory action of kisspeptin on gonadotropes remains debatable (192-196). Kiss1 and Kiss1r gene expression has been shown in gonadotropes, and gonadotropin secretion from the pituitary explants was observed following exposure to kisspeptin (78,192-195). Moreover, LHβ and FSHβ gene expression was upregulated in the primary pituitary cells treated with kisspeptin. Whilst kisspeptin can directly regulate gonadotropins at the transcriptional level, it appears to be less relevant than the GnRH-mediated action (67,195,196).

 

Desensitization Effect of Chronic or Continuous Exposure to Kisspeptin

 

Continuous administration of GnRH desensitizes the HPG axis by downregulation of GnRH receptors and desensitization of gonadotropes, following an initial stimulatory effect (39). It is therefore important to ascertain the effects of continuous exposure to kisspeptin on the HPG axis. Efforts have been made to assess the impact of continuous infusions of kisspeptin in a number of animal experiments (119,197-200).

 

In adult rats, continuous administration of kisspeptin-54 increased serum LH and free testosterone on day one, but this stimulatory effect was lost after 2 days, indicative of kisspeptin receptor desensitization (200). In rhesus monkeys, the continuous administration of kisspeptin-10 resulted in suppression of LH secretion, indicating desensitization of kisspeptin receptor (198). The kisspeptin receptor has been shown to desensitize in vitro (197). In sheep, infusion of kisspeptin-10 resulted in acute increase in serum LH levels, which declined by the end of 4-hour infusion, while GnRH remained elevated following the discontinuation of kisspeptin-10 administration. This suggests that desensitization to GnRH could be occurring at the level of pituitary gonadotropes (119).  

 

Consistent with animal studies, Jayasena et al. demonstrated that in women with hypothalamic amenorrhea an initial increase in LH and FSH secretion was not sustained following twice daily subcutaneous kisspeptin-54 administration for two weeks (176). Other studies in humans employing continuous or repeated kisspeptin administration provide conflicting evidence for kisspeptin-mediated desensitization and appear to be dose-related (172,180). High doses of kisspeptin may induce desensitization, but this is not apparent at lower doses (67). Sustained LH secretion and increased LH pulsatility was demonstrated with lower dose of kisspeptin-54 (0.01-1nmol/kg/h) infusion for 8 hours in women with hypothalamic amenorrhea (180) and kisspeptin-10 (3.1 nmol/kg/h) infusion for 22.5 hours in healthy men (172). In contrast, LH secretion was not maintained in three healthy men during the 24 hour infusion of kisspeptin-10 at 9.2 nmol/kg/h (the highest dose used in humans so far), although serum LH did not fall to the castrate levels and remained well above baseline at end of infusion (201).

 

Kisspeptin receptor agonist analogues, TAK-488 and TAK-683, induce desensitization when administered to healthy men (202,203). However, the ability of natural kisspeptin fragments to downregulate the HPG axis in humans remains to be established, and is to date complicated by differences in study protocols, in terms of isoform of kisspeptin used, duration (8 hours-2 weeks), mode and route of kisspeptin administration, lower doses of kisspeptin in human studies compared to animal, and the endocrine profile of the study participants (men versus women versus hypothalamic amenorrhea). 

 

Sexual Dimorphism in Kisspeptin Signaling

 

The response to kisspeptin is different in men and women. In men, kisspeptin potently stimulates the release of LH, but in women the effect of kisspeptin is variable and dependent on the phase of menstrual cycle (67). Whilst men respond to the modest doses of kisspeptin, LH response to kisspeptin in healthy women is minimal and inconsistent in the early follicular phase but greatest in the pre-ovulatory phase of the menstrual cycle (169-171,177). This indicates that in addition to the fluctuations in sex-steroid milieu, other mechanisms, such as changes in pituitary sensitivity to GnRH or GnRH network responsiveness to kisspeptin regulate the sensitivity to kisspeptin throughout the menstrual cycle (67,126,204).

 

Not only there is sexual dimorphism in gonadotropin response to kisspeptin, but there are also anatomical differences. Female hypothalami have significantly more kisspeptin fibers and kisspeptin cell bodies than men (173). Only a few kisspeptin cell bodies are present in the male infundibular nucleus and none in the rostral periventricular nucleus, which is on contrary to the female hypothalami with abundant kisspeptin network in both of these hypothalamic nuclei (108). These sex differences in kisspeptin neurons appear to be established early during perinatal development through the action of sex steroids (126,205).

 

These marked functional and anatomical differences may reflect sexually dimorphic roles of kisspeptin between both sexes, influencing their reproductive functions, namely the sex steroid feedback in GnRH and gonadotropin secretion (67).

 

Kisspeptin, GnRH and Puberty

 

Kisspeptin is crucial for normal pubertal development, the discovery of which formed the basis for the obligate role of kisspeptin signaling in the control of reproductive function (206). More than a decade ago two independent groups identified ‘inactivating’ mutations in KISS1R in patients with hypogonadotropic hypogonadism presenting with pubertal delay (85,86). Recently, a male patient with a biallelic loss-of-function KISS1R mutation was described who had undergone a normal and timely puberty, although as a child he had presented with microphallus and bilateral cryptorchidism. This suggests different levels of dependence of the hypothalamic-pituitary-gonadal axis on kisspeptin signaling during the reproductive life span, with the mini-puberty of infancy appearing more dependent on the kisspeptin system than is adolescent puberty (207). On the other hand, activating mutations in KISS1R and KISS1 were then described in children with central precocious puberty (89,90).

 

Hypothalamic expression of Kiss1 and Kiss1R mRNA is upregulated at puberty (117,165,208), and the percentage of GnRH neurons depolarizing in response to kisspeptin increases from juvenile (25%) to pubertal (50%) and to adult mice (>90%) (117), suggesting that GnRH neurons may acquire sensitivity to kisspeptin across puberty. In monkeys, kisspeptin-54 secretion and pulsatility increased at the onset of puberty (209). Moreover, the exogenous administration of kisspeptin resulted in earlier puberty in rats and monkeys (208,210), whereas kisspeptin antagonists delayed puberty in rats (186) and inhibited GnRH release in pubertal monkeys (211). In other study, daily injections of a synthetic kisspeptin analogue have been shown to significantly advance puberty in prepubertal female mice (212). GnRH neuron-specific Kiss1r knockout mouse showed a delay in pubertal onset, abnormal estrous cyclicity in female and abnormal external genitalia in male (microphallus, decreased anogenital distance associated with failure of preputial gland separation) (213).

 

Exogenous kisspeptin stimulated GnRH-induced LH secretion in patients with hypogonadotropism resulted in a spontaneous and permanent activation of their hypothalamic-pituitary-gonadal axis, whereas patients with idiopathic hypogonadotropic hypogonadism and no spontaneous LH pulsatility did not respond to kisspeptin, suggesting that the reversal of hypogonadism, sexual maturation and puberty may well be associated with the acquisition of kisspeptin responsiveness which in turn signals the emergence of reproductive endocrine activity (214). A recent study, 15 children with delayed puberty were administered intravenous kisspeptin and displayed divergent responses, with seven subjects having no response to kisspeptin, whereas others having either robust response (comparable to those of adults) or intermediate responses as perceived in one case (215).

 

GnRH release during puberty appears to require a cooperative mechanism between the kisspeptin/NKB networks in close interaction with different neuropeptides, as substance P, NKA, RFRP-3 and alpha-MSH, working as partners to regulate puberty timing influenced, naturally, by a combination of genetic, environmental, and gene-environment interactions (216).

 

Agonists and antagonists of kisspeptin and NKB were administered into the stalk-median eminence (region with high concentration of GnRH, kisspeptin and NKB neuroterminal fibers), and it was found that both kisspeptin-10 and the NK3R agonist senktide stimulated GnRH release in a dose-responsive manner in prepubertal and pubertal monkeys. However, senktide-induced GnRH release was blocked in the presence of a KISS1R antagonist and the kisspeptin-induced GnRH release was blocked in the presence of NK3R antagonist in pubertal monkeys, leading to the notion that a reciprocal signaling mechanism between kisspeptin and NKB exists and is possibly necessary for a normal puberty (217). These data together emphasizes that disrupted kisspeptin-GPR54-NKB signaling leads to hypogonadotropic hypogonadism, reinforcing the critical role of kisspeptin in puberty.

 

REGULATION OF GnRH AND GONADOTROPIN SECRETION

 

Development and maintenance of normal reproductive function requires a coordinated interplay between neuroendocrine, metabolic, and environmental factors. The GnRH-gonadotropin system plays a central role in the regulation of reproduction by integrating different signals and factors (Figure 3) (126,204). 

 

Figure 3. Neuroendocrine regulation of GnRH/gonadotropin secretion.
The GnRH-gonadotropin system plays a central role in the regulation of reproduction by integrating different neuroendocrine, metabolic and environmental signals/factors. The KNDy signaling has a key role in this process by integrating some of these signals and by regulating GnRH neurons.

 

Overview of Sex Steroid Feedback

 

A crucial role for sex steroids in the regulation of GnRH neurons and/or gonadotropes in humans was initially proposed as serial blood sampling and gonadotropin assays in women through phases of menstrual cycle showed an uneven distribution, with a clear mid-cycle surge in LH and FSH. Two mechanisms were proposed to mediate this effect: first, GnRH secretion is altered in response to the steroid milieu; second, sensitivity of the gonadotropes to a GnRH input is sex-steroid dependent, although the exact mechanism remains controversial due to inter-species variation (218).

 

Hypothalamic secretion of GnRH increases during proesterus in rats (219), sheep (220), and non-human primates (221). Pulsatile once hourly administration of exogenous GnRH restored ovulation in Rhesus monkeys with hypothalamic lesions which abolished GnRH secretion, suggesting that it was the ‘ebb and flow’ of ovarian estrogen feedback acting directly on the pituitary which triggered an LH surge (222). In humans, endogenous GnRH secretion is potentially diminished during the pre-ovulatory LH surge and the suppression of gonadotropin secretion is greater with lower doses of a GnRH receptor antagonist during the mid-cycle surge in comparison to the other phases of the menstrual cycle (223). This suggests that pituitary gonadotrope sensitivity to GnRH is enhanced during the mid-cycle surge. Administration of exogenous estradiol or testosterone in men with hypogonadotropic hypogonadism receiving pulsatile GnRH therapy, decreased gonadotropin concentrations, demonstrating inhibitory effects of sex-steroids at the level of pituitary (224). A direct effect of estrogen on gonadotropes is further demonstrated by the inhibition of LH secretion from rat pituitary gonadotropes in vitro (225). Literature to date suggests that there is dual-site sex-steroid feedback in the regulation of gonadotropin secretion, occurring at the level of both pituitary and hypothalamus (226-231).

 

Estrogen Feedback

 

Patterns of GnRH and LH secretion across the menstrual cycle are modulated by estradiol feedback. A biphasic effect of estradiol on gonadotropin secretion has long been established and it is essential for normal menstrual cycle, with an initial negative feedback (greater suppression of FSH) and a subsequent positive feedback (more prominent for LH) (32). However, the basis for estrogen feedback has been unclear for a long time. GnRH neurons do not express estrogen receptor alpha (ER-alpha) (232,233), and therefore a mediator between gonads and hypothalamus was missed. Recent evidence suggests that kisspeptin and neurokinin B (132) appears to be providing this “missing link” as a key regulator of both negative and positive estrogen feedback (67,126).

 

KNDy neurons in the infundibular nucleus in humans and the arcuate nucleus in other mammals mediate negative estrogen feedback. Estrogen suppresses kisspeptin and neurokinin B release from KNDy neurons, which reduce their stimulatory input to GnRH neurons. Simultaneously, there is a relative deficiency in dynorphin signaling as part of this negative feedback, releasing the inhibitory action on kisspeptin signaling (Figure 2) (67). Immunohistochemical staining of the postmenopausal women hypothalami showed up-regulated expression of KISS1 mRNA and hypertrophy of kisspeptin neurons in the infundibular nucleus when compared to the premenopausal women (111). These hypertrophied kisspeptin neurons co-localized with ER-alpha, had increased expression of neurokinin B and decreased levels of prodynorphin mRNA (234-236). The above evidence for the involvement of the infundibular KNDy system in mediating negative estrogen feedback in humans is consistent with animal studies. Kisspeptin neurons in the arcuate nucleus show frequent co-localization with ER-alpha (160,237). In ovariectomized animals, the expression of Kiss1 and neurokinin B mRNA was up-regulated but prodynorphin mRNA reduced in the arcuate nucleus (equivalent to the infundibular nucleus in humans), and this was reversed by estrogen replacement (102,115,120,238-242). Postmenopausal women are resistant to the stimulatory effect of kisspeptin on LH secretion (142,243), but postmenopausal women receiving estradiol replacement therapy are only resistant to kisspeptin initially and then they do demonstrate a remarkable increase in LH pulse amplitude with direct correlation to the circulating levels of estradiol and duration of kisspeptin administration (243). However, neurokinin B regulates gonadotropin secretion in postmenopausal women, and antagonizing the neurokinin 3 receptor modestly decreases LH secretion in this context (142).

 

Interestingly, the use of neurokinin 3 receptor antagonists has been shown to effectively reduce the frequency and severity of menopause-related vasomotor symptoms owing to their inhibitory effect in the hypothalamic thermoregulatory center, and thus presenting a potential non-hormonal treatment option for menopausal women (144,148,149).

 

Negative estrogen feedback switches to positive feedback in the late follicular phase of menstrual cycle, in order to induce the pre-ovulatory LH surge. Ovarian estradiol seems to be the predominant signal to trigger this switch, via ER-alpha, stimulating RP3V kisspeptin neurons while it inhibits arcuate kisspeptin neurons. Recent evidence supports the role of kisspeptin in generating the LH surge: during an assisted conception cycle, kisspeptin-54, used instead of a routinely administered human chorionic gonadotropin, induced an LH surge, and oocyte maturation, with a subsequent live term birth (241). Repeated twice-daily administration of kisspeptin-54 shortened the menstrual cycle, suggesting that the onset of LH surge was advanced (173). This is further supported by antagonistic studies in animal models, where the administration of kisspeptin antiserum or antagonists blunt/prevent LH peak, whilst kisspeptin advances LH surge (211,244,245).

 

However, kisspeptin-mediated positive estrogen feedback has marked anatomical variations between humans and other species. In rodents, positive estrogen feedback is mediated via the AVPV nucleus, which is absent in humans, other primates and sheep (Figure 2). AVPV neurons are sexually dimorphic, with higher density of ER-alpha described in females and AVPV kisspeptin neurons, as a subset of AVPV neurons, share this pattern (246). There seems to be functional specialization, since only a subset of AVPV kisspeptin neurons (~1/3) are synaptically connected to GnRH cell bodies, but of these, nearly all express estrogen sensitivity and most co-express tyrosine hydroxylase to facilitate positive feedback (247). The expression of Kiss1 mRNA in the AVPV nucleus is low following an ovariectomy but is dramatically increased with estrogen treatment and at the time of LH surge (160,161). In sheep, positive estrogen feedback is mediated though the arcuate nucleus, where the expression of Kiss1 mRNA is the greatest at the pre-ovulatory LH surge (195).

There are no studies looking at the anatomical region of estrogen mediating positive feedback in humans. Although there does not appear to be two distinct anatomical populations of kisspeptin neurons to relay negative and positive sex-steroid feedback in humans, it is possible that separate signaling pathways exists to mediate gonadal steroid feedback.

 

Whilst it is clear that kisspeptin is involved in estrogen-induced mid-cycle gonadotropin surge, the role of KNDy neurons in positive estrogen feedback is less obvious. In sheep, the expression of neurokinin B mRNA was increased during the LH surge, and neurokinin B receptor agonist senktide induced LH secretion mimicking its mid-cycle surge (248,249). However, this has not been reproduced in other species, including humans (180). In summary, KNDy neurons mediate negative estrogen feedback in the infundibular nucleus in humans and the arcuate nucleus in other species. Positive estrogen feedback is mediated via kisspeptin neurons, which show marked inter-species anatomical variation.

 

In addition to the gonads, the brain is one of the major organs producing estradiol, and recently a number of studies demonstrated that estradiol is synthesized and released in the hypothalamus (i.e. neuroestradiol) contributing to the regulation of GnRH release, particularly regarding its positive feedback effect during the preovulatory GnRH/LH surge (250).

 

Progesterone Feedback

 

Progesterone reduces LH pulse frequency in healthy women. LH secretory pattern in women exposed to exogenous progesterone was comparable to LH profile observed in the mid-luteal phase, demonstrating that progesterone plays a central role in the luteal phase of menstrual cycle (251). These inhibitory effects of progesterone on gonadotropin secretion are mediated by the progesterone receptor (PR) (252). The suppressive effect of progesterone on LH secretion was diminished in the context of estrogen deficiency, while co-administration of estradiol restored it (252), suggesting an interplay between these sex steroids. However, the presence of PR on only a small subset of GnRH neurons (253-255)led to the notion that intermediaries are involved in mediating inhibitory progesterone signal to GnRH neurons.

 

There is evidence that KNDy neurons play a role in mediating progesterone feedback on GnRH through dynorphin signaling (Figure 2) (102,120). PR have been demonstrated to be co-localized with dynorphin in the KNDy neurons (159)and progesterone increased dynorphin concentrations (256). Moreover, the number of preprodynorphin mRNA expressing cells decreased in postmenopausal women (236) and in ovariectomized ewes, but normalized with exogenous progesterone to luteal levels (256).

 

Testosterone Feedback

 

Testosterone exerts negative feedback on gonadotropin secretion. Early studies verified that LH and FSH pulse frequency are enhanced in hypogonadal men and exogenous testosterone decreases gonadotropin secretion, suggesting that testosterone have an inhibitory effect on GnRH secretion (230,257).

 

Few GnRH neurons express androgen receptors (AR) (258). GnRH neurons were thus considered to be reliant on an intermediary neuronal population to mediate testosterone feedback. A key role for KNDy neurons in this mediation has been suggested, as these neurons express AR which directly mediate the androgen feedback. The androgen feedback may also rely on the aromatization of testosterone, as testosterone-induced suppression of Kiss1 mRNA in the rodent arcuate nucleus is identical to that observed with estradiol, but more than that observed with dihydrotestosterone administration (259). The cross-talk between AR and ER was suggested from animal studies: AR expression was downregulated in the prostate following neonatal estrogen exposure (260), and AR transcription was modulated following a co-transfection of AR and ER (261).

 

Navarro has described a role for KNDy neurons in mediating the negative testosterone feedback on GnRH secretion, and provided evidence that neurokinin B released from KNDy neurons is part of an auto-feedback loop that generates the pulsatile secretion of Kiss1 and GnRH in male mice: Kiss1 and dynorphin mRNA are regulated by testosterone through estrogen and androgen receptor-dependent pathways; KNDy neurons express neurokinin B receptor whereas GnRH neurons do not, and senktide (an agonist for the neurokinin B receptor) activates KNDy neurons leading to gonadotropin secretion but has no discernible effect on GnRH neurons (262). Other studies demonstrated that the suppression of gonadotropin secretion using testosterone is associated with a reduction of Kiss1 mRNA in the hypothalamus (118,208,263). Moreover, post-orchidectomy rise in LH in rodents can be blocked by kisspeptin antagonists, further suggesting that kisspeptin system mediates the hypothalamic androgen feedback (186).

 

Stress and Glucocorticoids

 

Physical and psychological stress is associated with hypothalamic amenorrhea, possibly though the activation of hypothalamic-pituitary-adrenal (HPA) axis (264,265). Experimental evidence points towards a cortisol-mediated suppression of gonadotropin secretion as the main key pathway to explain stress-induced gonadotropin suppression(55,266-273). The negative effect of cortisol on HPG axis is recognized to occur at both pituitary and hypothalamic levels. There are also data suggesting that upstream factors in the HPA axis, such as Corticotropin Releasing Hormone (CRH) and vasopressin may play a mediatory role (274,275).

 

Cortisol secretion in women with hypothalamic amenorrhea is elevated (267), and evening adrenocorticotropic hormone (ACTH) and cortisol concentrations are higher in excessive exercise (266,270). Administration of exogenous glucocorticoids to eugonadal women was associated with a decrease in LH pulse frequency, suggesting that glucocorticoids have a negative action on GnRH secretion (273). In ovine portal blood, cortisol administration led to a decrease in GnRH pulse frequency (272). Inferences of cortisol effects on gonadotropin secretion were also derived from observations in women and men with Cushing’s syndrome (condition associated with excessive cortisol secretion), where exogenous GnRH preferentially stimulates FSH whilst LH remains unchanged (268,271). The resolution of male hypogonadotropic hypogonadism was also observed in men with remission of Cushing’s disease (271). This negative input of cortisol on the HPG axis may be modulated by sex-steroid hormones, and kisspeptin signaling has also been implicated in the process.

 

Cortisol alone had no impact on GnRH pulsatility in ovariectomized ewes, but the co-administration of estradiol and progesterone led to a 70% decrease in GnRH secretion (272). Decreased hypothalamic Kiss1 mRNA expression has been observed during exposure to stress or exogenous glucocorticoids. The role of kisspeptin in mediating stress inputs is further supported by the expression of glucocorticoid receptor on murine kisspeptin neurons (276). Colocalization by immunohistochemistry of CRH receptor (CRH-R) in most hypothalamic kisspeptin neurons in the AVPV/PeN and ARC nuclei as well as glucocorticoid receptor (GR) in AVPV/PeN kisspeptin neurons support a relevant direct role of kisspeptin neurons in the inhibitory effects of CRH/ glucocorticoids (277).

 

Hypothalamic CRH neurons, important regulators of the stress response, also directly modulate GnRH excitability in a dose-dependent and receptor-specific manner, and the GnRH response to CRH is influenced by estrogens (278). Intracerebroventricular administration of CRH in female rats suppressed LH pulsatility and the LH surge, and this suppression was enhanced by estrogens (279).

 

Animal models have also linked increased exposure to RFamide-related peptide-3 (RFRP-3) during acute and chronic stress and hypothalamic expression of GnIH mRNA. Along these lines, the surface of GnIH neurons has glucocorticoid receptors and hydrocortisone administration was associated to an increased GnIH mRNA expression, ultimately leading to lower GnRH activity and dysregulation of the HPG axis (280). Together, these findings emphasize that kisspeptin as GnIH provide relevant inputs that contribute to an inhibitory effect of corticosteroids on gonadal axis during stress.

 

Prolactin

 

Prolactin is a well-known inhibitor of GnRH release and a suppressor of the HPG axis. The association between hyperprolactinemia and reproductive dysfunction has long been established, accounting for 14% of secondary amenorrhea and hypogonadism cases (281) and for a third of women presenting with infertility (282,283). Hyperprolactinemia is evident in 16% of men with erectile dysfunction and in 11% of men with oligospermia (284). The decreased pulsatility of LH in hyperprolactinemia responds to bromocriptine (285). GnRH therapy has restored ovulation and normal luteal function in bromocriptine resistant hyperprolactinemia women (286,287), suggesting that prolactin exerts inhibition through direct reduction of GnRH secretion.

 

The neuroendocrine pathway by which prolactin inhibits GnRH pulse frequency remains to be fully elucidated. A direct action of prolactin on the GnRH neuronal network is possible (288,289). Prolactin has also been demonstrated to influence other systems, including GABA (290), β endorphins (291), neuropeptide Y (292) and dopaminergic systems (via tuberoinfundibular dopamine (TIDA) neurons) (293).

 

Nevertheless, data suggest that prolactin receptors are expressed in most kisspeptin neurons but only in a small proportion of GnRH neurons, indicating that kisspeptin signaling may have a role in this context (288,294). In rodent models, kisspeptin neurons in the arcuate nucleus modulate dopamine release from dopaminergic neurons, thereby regulating prolactin secretion (295). Kiss1 expression is decreased in lactation, a physiological state associated with hyperprolactinemia (296). Prolactin-sensitive GABA and kisspeptin neurons were identified in regions of the rat hypothalamus (294). Moreover, in a mouse model of anovulatory hyperprolactinemia (induced by a continuous infusion of prolactin), Kiss1 mRNA levels were diminished and peripheral administration of kisspeptin restored gonadotropin secretion and ovarian cyclicity (297). There are also other animal studies reporting an inhibitory effect of prolactin on Kiss1 expression (298,299). This data suggests that kisspeptin is a possible link between hyperprolactinemia and GnRH deficiency. The administration of kisspeptin-10 reactivated the gonadotropin secretion in women with hyperprolactinemia-induced hypogonadotropic amenorrhea, suggesting that GnRH deficiency in the context of hyperprolactinemia is, at least in part, mediated by an impaired hypothalamic kisspeptin secretion (300).

 

On the other hand, kisspeptins appears to have a stimulatory effect on prolactin release, as demonstrated in a recent study in ovariectomized rats which had intracerebroventricular injections of kisspeptin-10 with subsequent increase in prolactin release, and this required the estrogen receptor-alpha and was potentiated by progesterone via progesterone receptor activation (301).

 

Nutrition and Metabolism

 

A link between energy balance and reproductive function enables organisms to survive to reproductive maturity and to withstand the energy needs of parturition, lactation, and other parental behaviors. This link optimizes reproductive success under fluctuating metabolic conditions (302). Kisspeptin signaling may link nutrition/metabolic status and reproduction by sensing energy stores and translate this information into GnRH secretion (303). These relations elucidate further associations between reproductive dysfunction and metabolic disturbances, such as diabetes, obesity or anorexia nervosa (67,304,305).

 

Food deprivation impairs GnRH and gonadotropin secretion, and leptin (a satiety hormone secreted by adipose tissue, the levels of which drop in response to fasting) plays a role in this inter-regulation by stimulating LH release (67,306-308). Periods of fasting and calorie restriction decrease LH pulse frequency and increase pulse amplitude (302,309-311). Administration of recombinant leptin increased LH pulse frequency in women with hypothalamic amenorrhea (312) and prevented fasting-induced drop in testosterone and LH pulsatility in healthy men (313). Moreover, humans with mutations in leptin or in leptin receptor show hypogonadism (314). Thus, the crosstalk between kisspeptin and leptin is relevant for reproduction and fertility (71), including in the setting of assisted reproduction techniques (315).

 

Kisspeptin neurons may have a role in mediating the metabolic signals of leptin on the control of HPG axis, as 40% of the arcuate kisspeptin neurons express leptin receptors in contrast to the GnRH neurons, where leptin receptors are absent (316-319). Food deprivation is associated with a decrease in kisspeptin, and subsequent reduction in gonadotropin secretion (320-323). Levels of low Kiss1 mRNA expression in the leptin-deficient ob/ob mice are partially upregulated by exogenous leptin (161). Moreover, exogenous kisspeptin restored vaginal opening (marker of sexual maturation) in malnourished rodents (320). Animal models of type 1 diabetes, characterized by insulin deficiency and impaired cellular nutrition, had hypogonadotropic hypogonadism and decreased Kiss1 mRNA expression. Repeated administration of kisspeptin to these rodents increased prostate and testis weight (324). It is plausible that a relative deficiency of kisspeptin secretion is a mechanism for hypogonadotropic hypogonadism in patients with obesity and diabetes (179). In hypogonadal men with type 2 diabetes, kisspeptin-10 increased LH secretion and pulse frequency (179). Although early studies appeared to suggest a direct link between kisspeptin and leptin, it seems that the neuronal pathway whereby leptin modulates GnRH is far more complex (325,326). Only partial restoration in Kiss1 mRNA in leptin-deficiency and normal pubertal development and fertility observed in selective leptin receptor deletion from kisspeptin neurons suggest that kisspeptin may link reproduction and metabolism through other ways than leptin (161,327). Proopiomelanocortin (POMC), agouti-related peptide, neuropeptide Y, ghrelin, and cocaine- and amphetamine-regulated transcript (CART) expressing neurons have been linked to this process (303,319). Kisspeptin neurons communicate with POMC and neuropeptide Y neurons and are able to modulate the expression of relevant genes in these cells (316). This link between kisspeptin and other peptides classically associated to food intake (as POMC and neuropeptide Y) was explored due to the anorexigenic effect of intracerebroventricular administration of KP-10 in male rats mediated via anorectic neuropeptides, nesfatin-1 and oxytocin, expressed in various hypothalamic nuclei. Diminished food intake and anorexia was significantly abolished by pretreatment with oxytocin receptor antagonist (328,329).

 

Several studies have also suggested that ghrelin can interact directly with hypothalamic neurons leading to suppression of gonadotropins release, and thus impairing fertility, an effect that is dependent of the estradiol milieu (303,330-332).

 

GABA (Gamma-Amino Butyric Acid)

 

GABA has also been implicated as a regulator of GnRH secretion. Although GABA is classically an inhibitory neurotransmitter in the central nervous system, most mature GnRH neurons are stimulated by GABA, which has attributed to GABA an excitatory action in HPG axis. The precise physiology of this mechanism is still unclear (333-337), but it may be related to the bidirectional interactions between GABA and kisspeptin pathways, as well as between these and GnRH neurons, in a variety of ways throughout development (338). In early development, GABA seems to increase KISS1 expression in embryonic phase and early postnatally, while in the absence of GABAergic input the expression of KISS1 declines (338,339). In the prepubertal period, the central restraint on GnRH secretion seems to be mediated by GABA possibly acting directly via kisspeptin neurons (338). In the peri-pubertal phase, the antagonism of GABA and the intrinsic disinhibition of kisspeptin neurons seem to be critical in puberty initiation and development (340,341). In adulthood, the interactions between GnRH-GABA-kisspeptin become more complex with HPG axis function critically dependent on such interactions. For instance, the preovulatory surge does not occur in the absence of GABA signaling, thus neurons co-expressing GABA and kisspeptin seem crucial in providing double excitatory input to GnRH neurons at the time of ovulation (338,342).

 

Additionally, in healthy men, total endogenous GABA levels in the anterior cingulate cortex, a key limbic structure, significantly decreased after intravenous infusion of kisspeptin (1 nmol/kg/h) demonstrating a potent inhibitory effect of kisspeptin on GABA levels which could be a fundamental concept in understanding the central limbic effect of kisspeptin in the human brain (343).

 

Other Neuropeptides

 

In addition to KNDy system and GABA, other peptides and neurotransmitters have been shown to influence GnRH-gonadotrope system: vasoactive intestinal polypeptide (VIP), vasopressin, catecholamines, nitric oxide, neurotensin, gonadotropin-inhibitory hormone (GnIH) /RFamide related peptide-3 (RFRP-3) (337), nucleobindin-2/nesfatin-1 (344). Excitatory inputs to the HPG axis may be mediated by VIP, catecholamines, glutamate and possibly vasopressin, whereas GnIH in birds, or its mammalian homolog RFRP-3, provide inhibitory inputs (345-349). RFRP neuronal populations have been detected mainly in the hypothalamic dorsomedial nucleus or adjacent regions, and they have projections to several hypothalamic areas including the arcuate nucleus, paraventricular nucleus, ventromedial nucleus and the lateral hypothalamus, all areas with major roles in the regulation of reproduction and energy balance (350,351). RFRP-3, encoded by the gene Rfrp, inhibits the electric firing of GnRH and kisspeptin neurons (346,352), which results in a suppression of GnRH-induced gonadotropin release with consequent inhibition of the reproductive axis (353). This RFRP-3 inhibitory input on the gonadotropin release is influenced by estrogens and may well be involved in their negative feedback. Estrogens reduce RFRP-3 expression and RFRP-3 neuronal activation (354,355).

 

Particular attention has been paid to the role of glutamate as a stimulatory modulator of the activity of ARC kisspeptin neurons, reaffirming the role of kisspeptin as a major neural integrator of inputs to GnRH neurons. Data from Kiss1 KO rats showed failure to increase GnRH/LH secretion following monosodium glutamate/NMDA administration (356).

 

SUMMARY

 

Complex neuroendocrine networks coordinate the regulation of reproduction, integrating a wide range of internal and external environmental inputs and signals. GnRH, the principal regulator of reproduction integrates cues from sex steroids, stress, glucocorticoids, nutritional and metabolic status, prolactin and other peptides, to controls gonadotropin secretion and subsequently gonadal function. Recently, the KNDy neuronal network has emerged as essential gatekeeper of GnRH release and thus reproduction, fertility and puberty. Translational clinical studies, exploring kisspeptin and neurokinin B activity in various physiological and pathological states are pivotal to explore potential clinical applications for these novel neuropeptides and their agonists as well as antagonists, may underpin future management of some disorders with dysfunctional GnRH pulsatility, such polycystic ovary syndrome, hypothalamic amenorrhea, infertility, obesity, pubertal disorders and menopause-related symptoms.

 

REFERENCES

 

  1. Herbison AE, Theodosis DT. Localization of oestrogen receptors in preoptic neurons containing neurotensin but not tyrosine hydroxylase, cholecystokinin or luteinizing hormone-releasing hormone in the male and female rat. Neuroscience. 1992;50(2):283-298.
  2. Fink G. Neuroendocrine regulation of pituitary function: general principles. Neuroendocrinology in Physiology and Medicine. 2000:p.107-134.
  3. Tena-Sempere M. Hypothalamic KiSS-1: the missing link in gonadotropin feedback control? Endocrinology. 2005;146(9):3683-3685.
  4. Baba Y, Matsuo H, Schally AV. Structure of the porcine LH- and FSH-releasing hormone. II. Confirmation of the proposed structure by conventional sequential analyses. Biochem Biophys Res Commun. 1971;44(2):459-463.
  5. Matsuo H, Arimura A, Nair RM, Schally AV. Synthesis of the porcine LH- and FSH-releasing hormone by the solid-phase method. Biochem Biophys Res Commun. 1971;45(3):822-827.
  6. Schally AV, Arimura A, Kastin AJ, Matsuo H, Baba Y, Redding TW, Nair RM, Debeljuk L, White WF. Gonadotropin-releasing hormone: one polypeptide regulates secretion of luteinizing and follicle-stimulating hormones. Science. 1971;173(4001):1036-1038.
  7. Schally AV, Arimura A, Baba Y, Nair RM, Matsuo H, Redding TW, Debeljuk L. Isolation and properties of the FSH and LH-releasing hormone. Biochem Biophys Res Commun. 1971;43(2):393-399.
  8. Schally AV. Use of GnRH in preference to LH-RH terminology in scientific papers. Hum Reprod. 2000;15(9):2059-2061.
  9. Millar RP. GnRHs and GnRH receptors. Anim Reprod Sci. 2005;88(1-2):5-28.
  10. Okubo K, Nagahama Y. Structural and functional evolution of gonadotropin-releasing hormone in vertebrates. Acta Physiol (Oxf). 2008;193(1):3-15.
  11. Miyamoto K, Hasegawa Y, Nomura M, Igarashi M, Kangawa K, Matsuo H. Identification of the second gonadotropin-releasing hormone in chicken hypothalamus: evidence that gonadotropin secretion is probably controlled by two distinct gonadotropin-releasing hormones in avian species. Proc Natl Acad Sci U S A. 1984;81(12):3874-3878.
  12. White SA, Bond CT, Francis RC, Kasten TL, Fernald RD, Adelman JP. A second gene for gonadotropin-releasing hormone: cDNA and expression pattern in the brain. Proc Natl Acad Sci U S A. 1994;91(4):1423-1427.
  13. Gault PM, Maudsley S, Lincoln GA. Evidence that gonadotropin-releasing hormone II is not a physiological regulator of gonadotropin secretion in mammals. J Neuroendocrinol. 2003;15(9):831-839.
  14. Balasubramanian R, Dwyer A, Seminara SB, Pitteloud N, Kaiser UB, Crowley WF, Jr. Human GnRH deficiency: a unique disease model to unravel the ontogeny of GnRH neurons. Neuroendocrinology. 2010;92(2):81-99.
  15. Mitchell AL, Dwyer A, Pitteloud N, Quinton R. Genetic basis and variable phenotypic expression of Kallmann syndrome: towards a unifying theory. Trends Endocrinol Metab. 2011;22(7):249-258.
  16. Cariboni A, Pimpinelli F, Colamarino S, Zaninetti R, Piccolella M, Rumio C, Piva F, Rugarli EI, Maggi R. The product of X-linked Kallmann's syndrome gene (KAL1) affects the migratory activity of gonadotropin-releasing hormone (GnRH)-producing neurons. Hum Mol Genet. 2004;13(22):2781-2791.
  17. Cariboni A, Hickok J, Rakic S, Andrews W, Maggi R, Tischkau S, Parnavelas JG. Neuropilins and their ligands are important in the migration of gonadotropin-releasing hormone neurons. J Neurosci. 2007;27(9):2387-2395.
  18. Magni P, Dozio E, Ruscica M, Watanobe H, Cariboni A, Zaninetti R, Motta M, Maggi R. Leukemia inhibitory factor induces the chemomigration of immortalized gonadotropin-releasing hormone neurons through the independent activation of the Janus kinase/signal transducer and activator of transcription 3, mitogen-activated protein kinase/extracellularly regulated kinase 1/2, and phosphatidylinositol 3-kinase/Akt signaling pathways. Mol Endocrinol. 2007;21(5):1163-1174.
  19. Dode C, Levilliers J, Dupont JM, De Paepe A, Le Du N, Soussi-Yanicostas N, Coimbra RS, Delmaghani S, Compain-Nouaille S, Baverel F, Pecheux C, Le Tessier D, Cruaud C, Delpech M, Speleman F, Vermeulen S, Amalfitano A, Bachelot Y, Bouchard P, Cabrol S, Carel JC, Delemarre-van de Waal H, Goulet-Salmon B, Kottler ML, Richard O, Sanchez-Franco F, Saura R, Young J, Petit C, Hardelin JP. Loss-of-function mutations in FGFR1 cause autosomal dominant Kallmann syndrome. Nat Genet. 2003;33(4):463-465.
  20. Kim SH, Hu Y, Cadman S, Bouloux P. Diversity in fibroblast growth factor receptor 1 regulation: learning from the investigation of Kallmann syndrome. J Neuroendocrinol. 2008;20(2):141-163.
  21. Yoshida K, Rutishauser U, Crandall JE, Schwarting GA. Polysialic acid facilitates migration of luteinizing hormone-releasing hormone neurons on vomeronasal axons. J Neurosci. 1999;19(2):794-801.
  22. Kaprara A, Huhtaniemi IT. The hypothalamus-pituitary-gonad axis: Tales of mice and men. Metabolism. 2017.
  23. Martin C, Balasubramanian R, Dwyer AA, Au MG, Sidis Y, Kaiser UB, Seminara SB, Pitteloud N, Zhou QY, Crowley WF, Jr. The role of the prokineticin 2 pathway in human reproduction: evidence from the study of human and murine gene mutations. Endocr Rev. 2011;32(2):225-246.
  24. Pitteloud N, Zhang C, Pignatelli D, Li JD, Raivio T, Cole LW, Plummer L, Jacobson-Dickman EE, Mellon PL, Zhou QY, Crowley WF, Jr. Loss-of-function mutation in the prokineticin 2 gene causes Kallmann syndrome and normosmic idiopathic hypogonadotropic hypogonadism. Proc Natl Acad Sci U S A. 2007;104(44):17447-17452.
  25. Clifton DKS, R.A. . Neuroendocrinology of reproduction. Yen & Jaffe's Reproductive Endocrinology JF Strauss and RL Barberi, Elsevier. 2009.
  26. Maeda K, Ohkura S, Uenoyama Y, Wakabayashi Y, Oka Y, Tsukamura H, Okamura H. Neurobiological mechanisms underlying GnRH pulse generation by the hypothalamus. Brain Res. 2010;1364:103-115.
  27. Moenter SM, DeFazio AR, Pitts GR, Nunemaker CS. Mechanisms underlying episodic gonadotropin-releasing hormone secretion. Front Neuroendocrinol. 2003;24(2):79-93.
  28. Carmel PW, Araki S, Ferin M. Pituitary stalk portal blood collection in rhesus monkeys: evidence for pulsatile release of gonadotropin-releasing hormone (GnRH). Endocrinology. 1976;99(1):243-248.
  29. Antunes JL, Carmel PW, Housepian EM, Ferin M. Luteinizing hormone-releasing hormone in human pituitary blood. J Neurosurg. 1978;49(3):382-386.
  30. Caraty A, Martin GB, Montgomery G. A new method for studying pituitary responsiveness in vivo using pulses of LH-RH analogue in ewes passively immunized against native LH-RH. Reprod Nutr Dev. 1984;24(4):439-448.
  31. Lincoln GA, Fraser HM. Blockade of episodic secretion of luteinizing hormone in the ram by the administration of antibodies to luteinizing hormone releasing hormone. Biol Reprod. 1979;21(5):1239-1245.
  32. Knobil E, Plant TM, Wildt L, Belchetz PE, Marshall G. Control of the rhesus monkey menstrual cycle: permissive role of hypothalamic gonadotropin-releasing hormone. Science. 1980;207(4437):1371-1373.
  33. Bolt DJ. Changes in the concentration of luteinizing hormone in plasma of rams following administration of oestradiol, progesterone or testosterone. J Reprod Fertil. 1971;24(3):435-438.
  34. Dierschke DJ, Bhattacharya AN, Atkinson LE, Knobil E. Circhoral oscillations of plasma LH levels in the ovariectomized rhesus monkey. Endocrinology. 1970;87(5):850-853.
  35. Naftolin F, Yen SS, Tsai CC. Rapid cycling of plasma gonadotrophins in normal men as demonstrated by frequent sampling. Nat New Biol. 1972;236(64):92-93.
  36. Yen SS, Tsai CC. The biphasic pattern in the feedback action of ethinyl estradiol on the release of pituitary FSH and LH. J Clin Endocrinol Metab. 1971;33(6):882-887.
  37. Reame NE, Sauder SE, Case GD, Kelch RP, Marshall JC. Pulsatile gonadotropin secretion in women with hypothalamic amenorrhea: evidence that reduced frequency of gonadotropin-releasing hormone secretion is the mechanism of persistent anovulation. J Clin Endocrinol Metab. 1985;61(5):851-858.
  38. Sauder SE, Frager M, Case GD, Kelch RP, Marshall JC. Abnormal patterns of pulsatile luteinizing hormone secretion in women with hyperprolactinemia and amenorrhea: responses to bromocriptine. J Clin Endocrinol Metab. 1984;59(5):941-948.
  39. Belchetz PE, Plant TM, Nakai Y, Keogh EJ, Knobil E. Hypophysial responses to continuous and intermittent delivery of hypopthalamic gonadotropin-releasing hormone. Science. 1978;202(4368):631-633.
  40. Rasmussen DD. Episodic gonadotropin-releasing hormone release from the rat isolated median eminence in vitro. Neuroendocrinology. 1993;58(5):511-518.
  41. Thiery JC, Pelletier J. Multiunit activity in the anterior median eminence and adjacent areas of the hypothalamus of the ewe in relation to LH secretion. Neuroendocrinology. 1981;32(4):217-224.
  42. Wilson RC, Kesner JS, Kaufman JM, Uemura T, Akema T, Knobil E. Central electrophysiologic correlates of pulsatile luteinizing hormone secretion in the rhesus monkey. Neuroendocrinology. 1984;39(3):256-260.
  43. Martinez de la Escalera G, Choi AL, Weiner RI. Generation and synchronization of gonadotropin-releasing hormone (GnRH) pulses: intrinsic properties of the GT1-1 GnRH neuronal cell line. Proc Natl Acad Sci U S A. 1992;89(5):1852-1855.
  44. Terasawa E, Keen KL, Mogi K, Claude P. Pulsatile release of luteinizing hormone-releasing hormone (LHRH) in cultured LHRH neurons derived from the embryonic olfactory placode of the rhesus monkey. Endocrinology. 1999;140(3):1432-1441.
  45. Ezzat A, Pereira A, Clarke IJ. Kisspeptin is a component of the pulse generator for GnRH secretion in female sheep but not the pulse generator. Endocrinology. 2015;156(5):1828-1837.
  46. Millar RP, Lu ZL, Pawson AJ, Flanagan CA, Morgan K, Maudsley SR. Gonadotropin-releasing hormone receptors. Endocr Rev. 2004;25(2):235-275.
  47. Pincus SM, Padmanabhan V, Lemon W, Randolph J, Rees Midgley A. Follicle-stimulating hormone is secreted more irregularly than luteinizing hormone in both humans and sheep. J Clin Invest. 1998;101(6):1318-1324.
  48. Padmanabhan V, McFadden K, Mauger DT, Karsch FJ, Midgley AR, Jr. Neuroendocrine control of follicle-stimulating hormone (FSH) secretion. I. Direct evidence for separate episodic and basal components of FSH secretion. Endocrinology. 1997;138(1):424-432.
  49. Dalkin AC, Haisenleder DJ, Ortolano GA, Ellis TR, Marshall JC. The frequency of gonadotropin-releasing-hormone stimulation differentially regulates gonadotropin subunit messenger ribonucleic acid expression. Endocrinology. 1989;125(2):917-924.
  50. Haisenleder DJ, Dalkin AC, Ortolano GA, Marshall JC, Shupnik MA. A pulsatile gonadotropin-releasing hormone stimulus is required to increase transcription of the gonadotropin subunit genes: evidence for differential regulation of transcription by pulse frequency in vivo. Endocrinology. 1991;128(1):509-517.
  51. Kaiser UB, Jakubowiak A, Steinberger A, Chin WW. Differential effects of gonadotropin-releasing hormone (GnRH) pulse frequency on gonadotropin subunit and GnRH receptor messenger ribonucleic acid levels in vitro. Endocrinology. 1997;138(3):1224-1231.
  52. Spratt DI, Finkelstein JS, Butler JP, Badger TM, Crowley WF, Jr. Effects of increasing the frequency of low doses of gonadotropin-releasing hormone (GnRH) on gonadotropin secretion in GnRH-deficient men. J Clin Endocrinol Metab. 1987;64(6):1179-1186.
  53. Waldhauser F, Weissenbacher G, Frisch H, Pollak A. Pulsatile secretion of gonadotropins in early infancy. Eur J Pediatr. 1981;137(1):71-74.
  54. Conte FA, Grumbach MM, Kaplan SL, Reiter EO. Correlation of luteinizing hormone-releasing factor-induced luteinizing hormone and follicle-stimulating hormone release from infancy to 19 years with the changing pattern of gonadotropin secretion in agonadal patients: relation to the restraint of puberty. J Clin Endocrinol Metab. 1980;50(1):163-168.
  55. Pohl CR, deRidder CM, Plant TM. Gonadal and nongonadal mechanisms contribute to the prepubertal hiatus in gonadotropin secretion in the female rhesus monkey (Macaca mulatta). J Clin Endocrinol Metab. 1995;80(7):2094-2101.
  56. Boyar R, Finkelstein J, Roffwarg H, Kapen S, Weitzman E, Hellman L. Synchronization of augmented luteinizing hormone secretion with sleep during puberty. N Engl J Med. 1972;287(12):582-586.
  57. Roth JC, Kelch RP, Kaplan SL, Grumbach MM. FSH and LH response to luteinizing hormone-releasing factor in prepubertal and pubertal children, adult males and patients with hypogonadotropic and hypertropic hypogonadism. J Clin Endocrinol Metab. 1972;35(6):926-930.
  58. Marshall JC, Dalkin AC, Haisenleder DJ, Griffin ML, Kelch RP. GnRH pulses--the regulators of human reproduction. Trans Am Clin Climatol Assoc. 1993;104:31-46.
  59. Marshall JC, Dalkin AC, Haisenleder DJ, Paul SJ, Ortolano GA, Kelch RP. Gonadotropin-releasing hormone pulses: regulators of gonadotropin synthesis and ovulatory cycles. Recent Prog Horm Res. 1991;47:155-187; discussion 188-159.
  60. Yen SS, Tsai CC, Naftolin F, Vandenberg G, Ajabor L. Pulsatile patterns of gonadotropin release in subjects with and without ovarian function. J Clin Endocrinol Metab. 1972;34(4):671-675.
  61. Arroyo A, Laughlin GA, Morales AJ, Yen SS. Inappropriate gonadotropin secretion in polycystic ovary syndrome: influence of adiposity. J Clin Endocrinol Metab. 1997;82(11):3728-3733.
  62. Morales AJ, Laughlin GA, Butzow T, Maheshwari H, Baumann G, Yen SS. Insulin, somatotropic, and luteinizing hormone axes in lean and obese women with polycystic ovary syndrome: common and distinct features. J Clin Endocrinol Metab. 1996;81(8):2854-2864.
  63. Waldstreicher J, Santoro NF, Hall JE, Filicori M, Crowley WF, Jr. Hyperfunction of the hypothalamic-pituitary axis in women with polycystic ovarian disease: indirect evidence for partial gonadotroph desensitization. J Clin Endocrinol Metab. 1988;66(1):165-172.
  64. Yen SS, Vela P, Rankin J. Inappropriate secretion of follicle-stimulating hormone and luteinizing hormone in polycystic ovarian disease. J Clin Endocrinol Metab. 1970;30(4):435-442.
  65. Beltramo M, Dardente H, Cayla X, Caraty A. Cellular mechanisms and integrative timing of neuroendocrine control of GnRH secretion by kisspeptin. Mol Cell Endocrinol. 2014;382(1):387-399.
  66. Jayasena CN, Abbara A, Izzi-Engbeaya C, Comninos AN, Harvey RA, Gonzalez Maffe J, Sarang Z, Ganiyu-Dada Z, Padilha AI, Dhanjal M, Williamson C, Regan L, Ghatei MA, Bloom SR, Dhillo WS. Reduced levels of plasma kisspeptin during the antenatal booking visit are associated with increased risk of miscarriage. J Clin Endocrinol Metab. 2014;99(12):E2652-2660.
  67. Skorupskaite K, George JT, Anderson RA. The kisspeptin-GnRH pathway in human reproductive health and disease. Hum Reprod Update. 2014;20(4):485-500.
  68. Clarke SA, Dhillo WS. Kisspeptin across the human lifespan:evidence from animal studies and beyond. J Endocrinol. 2016;229(3):R83-98.
  69. Comninos AN, Dhillo WS. Emerging Roles of Kisspeptin in Sexual and Emotional Brain Processing. Neuroendocrinology. 2018;106(2):195-202.
  70. Dudek M, Ziarniak K, Sliwowska JH. Kisspeptin and Metabolism: The Brain and Beyond. Front Endocrinol (Lausanne). 2018;9:145.
  71. Wahab F, Atika B, Ullah F, Shahab M, Behr R. Metabolic Impact on the Hypothalamic Kisspeptin-Kiss1r Signaling Pathway. Front Endocrinol (Lausanne). 2018;9:123.
  72. Wolfe A, Hussain MA. The Emerging Role(s) for Kisspeptin in Metabolism in Mammals. Front Endocrinol (Lausanne). 2018;9:184.
  73. Yang L, Comninos AN, Dhillo WS. Intrinsic links among sex, emotion, and reproduction. Cell Mol Life Sci. 2018;75(12):2197-2210.
  74. Mills EG, Yang L, Abbara A, Dhillo WS, Comninos AN. Current Perspectives on Kisspeptins Role in Behaviour. Front Endocrinol (Lausanne). 2022;13:928143.
  75. Lee JH, Miele ME, Hicks DJ, Phillips KK, Trent JM, Weissman BE, Welch DR. KiSS-1, a novel human malignant melanoma metastasis-suppressor gene. J Natl Cancer Inst. 1996;88(23):1731-1737.
  76. Ciaramella V, Della Corte CM, Ciardiello F, Morgillo F. Kisspeptin and Cancer: Molecular Interaction, Biological Functions, and Future Perspectives. Front Endocrinol (Lausanne). 2018;9:115.
  77. West A, Vojta PJ, Welch DR, Weissman BE. Chromosome localization and genomic structure of the KiSS-1 metastasis suppressor gene (KISS1). Genomics. 1998;54(1):145-148.
  78. Kotani M, Detheux M, Vandenbogaerde A, Communi D, Vanderwinden JM, Le Poul E, Brezillon S, Tyldesley R, Suarez-Huerta N, Vandeput F, Blanpain C, Schiffmann SN, Vassart G, Parmentier M. The metastasis suppressor gene KiSS-1 encodes kisspeptins, the natural ligands of the orphan G protein-coupled receptor GPR54. J Biol Chem. 2001;276(37):34631-34636.
  79. Pasquier J, Kamech N, Lafont AG, Vaudry H, Rousseau K, Dufour S. Molecular evolution of GPCRs: Kisspeptin/kisspeptin receptors. J Mol Endocrinol. 2014;52(3):T101-117.
  80. Gottsch ML, Navarro VM, Zhao Z, Glidewell-Kenney C, Weiss J, Jameson JL, Clifton DK, Levine JE, Steiner RA. Regulation of Kiss1 and dynorphin gene expression in the murine brain by classical and nonclassical estrogen receptor pathways. J Neurosci. 2009;29(29):9390-9395.
  81. Lee DK, Nguyen T, O'Neill GP, Cheng R, Liu Y, Howard AD, Coulombe N, Tan CP, Tang-Nguyen AT, George SR, O'Dowd BF. Discovery of a receptor related to the galanin receptors. FEBS Lett. 1999;446(1):103-107.
  82. Muir AI, Chamberlain L, Elshourbagy NA, Michalovich D, Moore DJ, Calamari A, Szekeres PG, Sarau HM, Chambers JK, Murdock P, Steplewski K, Shabon U, Miller JE, Middleton SE, Darker JG, Larminie CG, Wilson S, Bergsma DJ, Emson P, Faull R, Philpott KL, Harrison DC. AXOR12, a novel human G protein-coupled receptor, activated by the peptide KiSS-1. J Biol Chem. 2001;276(31):28969-28975.
  83. Ohtaki T, Shintani Y, Honda S, Matsumoto H, Hori A, Kanehashi K, Terao Y, Kumano S, Takatsu Y, Masuda Y, Ishibashi Y, Watanabe T, Asada M, Yamada T, Suenaga M, Kitada C, Usuki S, Kurokawa T, Onda H, Nishimura O, Fujino M. Metastasis suppressor gene KiSS-1 encodes peptide ligand of a G-protein-coupled receptor. Nature. 2001;411(6837):613-617.
  84. Liu X, Lee K, Herbison AE. Kisspeptin excites gonadotropin-releasing hormone neurons through a phospholipase C/calcium-dependent pathway regulating multiple ion channels. Endocrinology. 2008;149(9):4605-4614.
  85. de Roux N, Genin E, Carel JC, Matsuda F, Chaussain JL, Milgrom E. Hypogonadotropic hypogonadism due to loss of function of the KiSS1-derived peptide receptor GPR54. Proc Natl Acad Sci U S A. 2003;100(19):10972-10976.
  86. Seminara SB, Messager S, Chatzidaki EE, Thresher RR, Acierno JS, Jr., Shagoury JK, Bo-Abbas Y, Kuohung W, Schwinof KM, Hendrick AG, Zahn D, Dixon J, Kaiser UB, Slaugenhaupt SA, Gusella JF, O'Rahilly S, Carlton MB, Crowley WF, Jr., Aparicio SA, Colledge WH. The GPR54 gene as a regulator of puberty. N Engl J Med. 2003;349(17):1614-1627.
  87. Tenenbaum-Rakover Y, Commenges-Ducos M, Iovane A, Aumas C, Admoni O, de Roux N. Neuroendocrine phenotype analysis in five patients with isolated hypogonadotropic hypogonadism due to a L102P inactivating mutation of GPR54. J Clin Endocrinol Metab. 2007;92(3):1137-1144.
  88. Chan YM, Broder-Fingert S, Wong KM, Seminara SB. Kisspeptin/Gpr54-independent gonadotrophin-releasing hormone activity in Kiss1 and Gpr54 mutant mice. J Neuroendocrinol. 2009;21(12):1015-1023.
  89. Teles MG, Bianco SD, Brito VN, Trarbach EB, Kuohung W, Xu S, Seminara SB, Mendonca BB, Kaiser UB, Latronico AC. A GPR54-activating mutation in a patient with central precocious puberty. N Engl J Med. 2008;358(7):709-715.
  90. Silveira LG, Noel SD, Silveira-Neto AP, Abreu AP, Brito VN, Santos MG, Bianco SD, Kuohung W, Xu S, Gryngarten M, Escobar ME, Arnhold IJ, Mendonca BB, Kaiser UB, Latronico AC. Mutations of the KISS1 gene in disorders of puberty. J Clin Endocrinol Metab. 2010;95(5):2276-2280.
  91. Dong L, Zhou W, Lin Z, Tang L, Deng X, Chen B, Huang W, Xiong Q. Polymorphism rs5780218, rs12998 and rs10158616 in KISS1 gene among the Hubei province Chinese girls with the central precocious puberty. Endocrine. 2024;84(3):1229-1237.
  92. Topaloglu AK, Reimann F, Guclu M, Yalin AS, Kotan LD, Porter KM, Serin A, Mungan NO, Cook JR, Imamoglu S, Akalin NS, Yuksel B, O'Rahilly S, Semple RK. TAC3 and TACR3 mutations in familial hypogonadotropic hypogonadism reveal a key role for Neurokinin B in the central control of reproduction. Nat Genet. 2009;41(3):354-358.
  93. Gianetti E, Tusset C, Noel SD, Au MG, Dwyer AA, Hughes VA, Abreu AP, Carroll J, Trarbach E, Silveira LF, Costa EM, de Mendonca BB, de Castro M, Lofrano A, Hall JE, Bolu E, Ozata M, Quinton R, Amory JK, Stewart SE, Arlt W, Cole TR, Crowley WF, Kaiser UB, Latronico AC, Seminara SB. TAC3/TACR3 mutations reveal preferential activation of gonadotropin-releasing hormone release by neurokinin B in neonatal life followed by reversal in adulthood. J Clin Endocrinol Metab. 2010;95(6):2857-2867.
  94. Guran T, Tolhurst G, Bereket A, Rocha N, Porter K, Turan S, Gribble FM, Kotan LD, Akcay T, Atay Z, Canan H, Serin A, O'Rahilly S, Reimann F, Semple RK, Topaloglu AK. Hypogonadotropic hypogonadism due to a novel missense mutation in the first extracellular loop of the neurokinin B receptor. J Clin Endocrinol Metab. 2009;94(10):3633-3639.
  95. Young J, Bouligand J, Francou B, Raffin-Sanson ML, Gaillez S, Jeanpierre M, Grynberg M, Kamenicky P, Chanson P, Brailly-Tabard S, Guiochon-Mantel A. TAC3 and TACR3 defects cause hypothalamic congenital hypogonadotropic hypogonadism in humans. J Clin Endocrinol Metab. 2010;95(5):2287-2295.
  96. Young J, George JT, Tello JA, Francou B, Bouligand J, Guiochon-Mantel A, Brailly-Tabard S, Anderson RA, Millar RP. Kisspeptin restores pulsatile LH secretion in patients with neurokinin B signaling deficiencies: physiological, pathophysiological and therapeutic implications. Neuroendocrinology. 2013;97(2):193-202.
  97. Wilkes MM, Watkins WB, Stewart RD, Yen SS. Localization and quantitation of beta-endorphin in human brain and pituitary. Neuroendocrinology. 1980;30(2):113-121.
  98. Quigley ME, Yen SS. The role of endogenous opiates in LH secretion during the menstrual cycle. J Clin Endocrinol Metab. 1980;51(1):179-181.
  99. Veldhuis JD, Rogol AD, Samojlik E, Ertel NH. Role of endogenous opiates in the expression of negative feedback actions of androgen and estrogen on pulsatile properties of luteinizing hormone secretion in man. J Clin Invest. 1984;74(1):47-55.
  100. Boukhliq R, Goodman RL, Berriman SJ, Adrian B, Lehman MN. A subset of gonadotropin-releasing hormone neurons in the ovine medial basal hypothalamus is activated during increased pulsatile luteinizing hormone secretion. Endocrinology. 1999;140(12):5929-5936.
  101. Genazzani AD, Gastaldi M, Petraglia F, Battaglia C, Surico N, Volpe A, Genazzani AR. Naltrexone administration modulates the neuroendocrine control of luteinizing hormone secretion in hypothalamic amenorrhoea. Hum Reprod. 1995;10(11):2868-2871.
  102. Goodman RL, Coolen LM, Anderson GM, Hardy SL, Valent M, Connors JM, Fitzgerald ME, Lehman MN. Evidence that dynorphin plays a major role in mediating progesterone negative feedback on gonadotropin-releasing hormone neurons in sheep. Endocrinology. 2004;145(6):2959-2967.
  103. Shoupe D, Montz FJ, Lobo RA. The effects of estrogen and progestin on endogenous opioid activity in oophorectomized women. J Clin Endocrinol Metab. 1985;60(1):178-183.
  104. Walsh JP, Clarke IJ. Effects of central administration of highly selective opioid mu-, delta- and kappa-receptor agonists on plasma luteinizing hormone (LH), prolactin, and the estrogen-induced LH surge in ovariectomized ewes. Endocrinology. 1996;137(9):3640-3648.
  105. Cheng G, Coolen LM, Padmanabhan V, Goodman RL, Lehman MN. The kisspeptin/neurokinin B/dynorphin (KNDy) cell population of the arcuate nucleus: sex differences and effects of prenatal testosterone in sheep. Endocrinology. 2010;151(1):301-311.
  106. Goodman RL, Lehman MN, Smith JT, Coolen LM, de Oliveira CV, Jafarzadehshirazi MR, Pereira A, Iqbal J, Caraty A, Ciofi P, Clarke IJ. Kisspeptin neurons in the arcuate nucleus of the ewe express both dynorphin A and neurokinin B. Endocrinology. 2007;148(12):5752-5760.
  107. Burke MC, Letts PA, Krajewski SJ, Rance NE. Coexpression of dynorphin and neurokinin B immunoreactivity in the rat hypothalamus: Morphologic evidence of interrelated function within the arcuate nucleus. J Comp Neurol. 2006;498(5):712-726.
  108. Hrabovszky E, Ciofi P, Vida B, Horvath MC, Keller E, Caraty A, Bloom SR, Ghatei MA, Dhillo WS, Liposits Z, Kallo I. The kisspeptin system of the human hypothalamus: sexual dimorphism and relationship with gonadotropin-releasing hormone and neurokinin B neurons. Eur J Neurosci. 2010;31(11):1984-1998.
  109. Bedenbaugh MN, O'Connell RC, Lopez JA, McCosh RB, Goodman RL, Hileman SM. Kisspeptin, gonadotrophin-releasing hormone and oestrogen receptor alpha colocalise with neuronal nitric oxide synthase neurones in prepubertal female sheep. J Neuroendocrinol. 2018;30(1).
  110. Dufourny L, Delmas O, Teixeira-Gomes AP, Decourt C, Sliwowska JH. Neuroanatomical connections between kisspeptin neurones and somatostatin neurones in female and male rat hypothalamus: a possible involvement of SSTR1 in kisspeptin release. J Neuroendocrinol. 2018:e12593.
  111. Rometo AM, Krajewski SJ, Voytko ML, Rance NE. Hypertrophy and increased kisspeptin gene expression in the hypothalamic infundibular nucleus of postmenopausal women and ovariectomized monkeys. J Clin Endocrinol Metab. 2007;92(7):2744-2750.
  112. Clarkson J, Herbison AE. Postnatal development of kisspeptin neurons in mouse hypothalamus; sexual dimorphism and projections to gonadotropin-releasing hormone neurons. Endocrinology. 2006;147(12):5817-5825.
  113. Pompolo S, Pereira A, Estrada KM, Clarke IJ. Colocalization of kisspeptin and gonadotropin-releasing hormone in the ovine brain. Endocrinology. 2006;147(2):804-810.
  114. Clarkson J, d'Anglemont de Tassigny X, Colledge WH, Caraty A, Herbison AE. Distribution of kisspeptin neurones in the adult female mouse brain. J Neuroendocrinol. 2009;21(8):673-682.
  115. Oakley AE, Clifton DK, Steiner RA. Kisspeptin signaling in the brain. Endocr Rev. 2009;30(6):713-743.
  116. Uenoyama Y, Inoue N, Pheng V, Homma T, Takase K, Yamada S, Ajiki K, Ichikawa M, Okamura H, Maeda KI, Tsukamura H. Ultrastructural evidence of kisspeptin-gonadotrophin-releasing hormone (GnRH) interaction in the median eminence of female rats: implication of axo-axonal regulation of GnRH release. J Neuroendocrinol. 2011;23(10):863-870.
  117. Han SK, Gottsch ML, Lee KJ, Popa SM, Smith JT, Jakawich SK, Clifton DK, Steiner RA, Herbison AE. Activation of gonadotropin-releasing hormone neurons by kisspeptin as a neuroendocrine switch for the onset of puberty. J Neurosci. 2005;25(49):11349-11356.
  118. Irwig MS, Fraley GS, Smith JT, Acohido BV, Popa SM, Cunningham MJ, Gottsch ML, Clifton DK, Steiner RA. Kisspeptin activation of gonadotropin releasing hormone neurons and regulation of KiSS-1 mRNA in the male rat. Neuroendocrinology. 2004;80(4):264-272.
  119. Messager S, Chatzidaki EE, Ma D, Hendrick AG, Zahn D, Dixon J, Thresher RR, Malinge I, Lomet D, Carlton MB, Colledge WH, Caraty A, Aparicio SA. Kisspeptin directly stimulates gonadotropin-releasing hormone release via G protein-coupled receptor 54. Proc Natl Acad Sci U S A. 2005;102(5):1761-1766.
  120. Lehman MN, Coolen LM, Goodman RL. Minireview: kisspeptin/neurokinin B/dynorphin (KNDy) cells of the arcuate nucleus: a central node in the control of gonadotropin-releasing hormone secretion. Endocrinology. 2010;151(8):3479-3489.
  121. Mills EGA, Dhillo WS, Comninos AN. Kisspeptin and the control of emotions, mood and reproductive behaviour. J Endocrinol. 2018;239(1):R1-R12.
  122. Cao Y, Li Z, Jiang W, Ling Y, Kuang H. Reproductive functions of Kisspeptin/KISS1R Systems in the Periphery. Reprod Biol Endocrinol. 2019;17(1):65.
  123. Hu KL, Chang HM, Zhao HC, Yu Y, Li R, Qiao J. Potential roles for the kisspeptin/kisspeptin receptor system in implantation and placentation. Hum Reprod Update. 2019;25(3):326-343.
  124. Bhattacharya M, Babwah AV. Kisspeptin: beyond the brain. Endocrinology. 2015;156(4):1218-1227.
  125. Guzman S, Dragan M, Kwon H, de Oliveira V, Rao S, Bhatt V, Kalemba KM, Shah A, Rustgi VK, Wang H, Bech PR, Abbara A, Izzi-Engbeaya C, Manousou P, Guo JY, Guo GL, Radovick S, Dhillo WS, Wondisford FE, Babwah AV, Bhattacharya M. Targeting hepatic kisspeptin receptor ameliorates nonalcoholic fatty liver disease in a mouse model. J Clin Invest. 2022;132(10).
  126. Pinilla L, Aguilar E, Dieguez C, Millar RP, Tena-Sempere M. Kisspeptins and reproduction: physiological roles and regulatory mechanisms. Physiol Rev. 2012;92(3):1235-1316.
  127. Ikegami K, Minabe S, Ieda N, Goto T, Sugimoto A, Nakamura S, Inoue N, Oishi S, Maturana AD, Sanbo M, Hirabayashi M, Maeda KI, Tsukamura H, Uenoyama Y. Evidence of involvement of neurone-glia/neurone-neurone communications via gap junctions in synchronised activity of KNDy neurones. J Neuroendocrinol. 2017;29(6).
  128. Topaloglu AK, Semple RK. Neurokinin B signalling in the human reproductive axis. Mol Cell Endocrinol. 2011;346(1-2):57-64.
  129. Navarro VM, Bosch MA, Leon S, Simavli S, True C, Pinilla L, Carroll RS, Seminara SB, Tena-Sempere M, Ronnekleiv OK, Kaiser UB. The integrated hypothalamic tachykinin-kisspeptin system as a central coordinator for reproduction. Endocrinology. 2015;156(2):627-637.
  130. Ikegami K, Watanabe Y, Nakamura S, Goto T, Inoue N, Uenoyama Y, Tsukamura H. Cellular and molecular mechanisms regulating the KNDy neuronal activities to generate and modulate GnRH pulse in mammals. Front Neuroendocrinol. 2022;64:100968.
  131. Skorupskaite K, George JT, Veldhuis JD, Anderson RA. Neurokinin B Regulates Gonadotropin Secretion, Ovarian Follicle Growth, and the Timing of Ovulation in Healthy Women. J Clin Endocrinol Metab. 2018;103(1):95-104.
  132. Skorupskaite K, George JT, Veldhuis JD, Millar RP, Anderson RA. Interactions Between Neurokinin B and Kisspeptin in Mediating Estrogen Feedback in Healthy Women. J Clin Endocrinol Metab. 2016;101(12):4628-4636.
  133. Skorupskaite K, George J, Anderson RA. Role of a neurokinin B receptor antagonist in the regulation of ovarian function in healthy women. Lancet. 2015;385 Suppl 1:S92.
  134. Skorupskaite K, Anderson RA. Hypothalamic neurokinin signalling and its application in reproductive medicine. Pharmacol Ther. 2021:107960.
  135. Akad M, Socolov R, Furnica C, Covali R, Stan CD, Crauciuc E, Pavaleanu I. Kisspeptin Variations in Patients with Polycystic Ovary Syndrome-A Prospective Case Control Study. Medicina (Kaunas). 2022;58(6).
  136. de Assis Rodrigues NP, Lagana AS, Zaia V, Vitagliano A, Barbosa CP, de Oliveira R, Trevisan CM, Montagna E. The role of Kisspeptin levels in polycystic ovary syndrome: a systematic review and meta-analysis. Arch Gynecol Obstet. 2019;300(5):1423-1434.
  137. Zhao T, Zhang Q, Xiao X, Tao X, Gao M, He W, Wu X, Yuan T. Associations of the KiSS-1 and GPR54 genetic polymorphism with polycystic ovary syndrome in Yunnan, China. Gynecol Endocrinol. 2022;38(9):790-794.
  138. Zhao T, Xiao X, Li L, Tao X, He W, Zhang Q, Wu X, Yuan T. Role of kisspeptin in polycystic ovarian syndrome: A metabolomics study. Clin Endocrinol (Oxf). 2023;99(3):315-325.
  139. George JT, Kakkar R, Marshall J, Scott ML, Finkelman RD, Ho TW, Veldhuis J, Skorupskaite K, Anderson RA, McIntosh S, Webber L. Neurokinin B Receptor Antagonism in Women With Polycystic Ovary Syndrome: A Randomized, Placebo-Controlled Trial. J Clin Endocrinol Metab. 2016;101(11):4313-4321.
  140. Fraser GL, Obermayer-Pietsch B, Laven J, Griesinger G, Pintiaux A, Timmerman D, Fauser B, Lademacher C, Combalbert J, Hoveyda HR, Ramael S. Randomized Controlled Trial of Neurokinin 3 Receptor Antagonist Fezolinetant for Treatment of Polycystic Ovary Syndrome. J Clin Endocrinol Metab. 2021.
  141. Skorupskaite K, George JT, Veldhuis JD, Millar RP, Anderson RA. Kisspeptin and neurokinin B interactions in modulating gonadotropin secretion in women with polycystic ovary syndrome. Hum Reprod. 2020;35(6):1421-1431.
  142. Skorupskaite K, George JT, Veldhuis JD, Millar RP, Anderson RA. Neurokinin 3 Receptor Antagonism Reveals Roles for Neurokinin B in the Regulation of Gonadotropin Secretion and Hot Flashes in Postmenopausal Women. Neuroendocrinology. 2018;106(2):148-157.
  143. Prague JK, Roberts RE, Comninos AN, Clarke S, Jayasena CN, Nash Z, Doyle C, Papadopoulou DA, Bloom SR, Mohideen P, Panay N, Hunter MS, Veldhuis JD, Webber LC, Huson L, Dhillo WS. Neurokinin 3 receptor antagonism as a novel treatment for menopausal hot flushes: a phase 2, randomised, double-blind, placebo-controlled trial. Lancet. 2017;389(10081):1809-1820.
  144. Depypere H, Timmerman D, Donders G, Sieprath P, Ramael S, Combalbert J, Hoveyda HR, Fraser GL. Treatment of Menopausal Vasomotor Symptoms With Fezolinetant, a Neurokinin 3 Receptor Antagonist: A Phase 2a Trial. J Clin Endocrinol Metab. 2019;104(12):5893-5905.
  145. Prague JK, Dhillo WS. Treating hot flushes with a neurokinin 3 receptor antagonist. Oncotarget. 2017;8(63):106153-106154.
  146. Prague JK, Dhillo WS. Neurokinin 3 receptor antagonism - the magic bullet for hot flushes? Climacteric. 2017;20(6):505-509.
  147. Anderson RA, Skorupskaite K, Sassarini J. The neurokinin B pathway in the treatment of menopausal hot flushes. Climacteric. 2019;22(1):51-54.
  148. Johnson KA, Martin N, Nappi RE, Neal-Perry G, Shapiro M, Stute P, Thurston RC, Wolfman W, English M, Franklin C, Lee M, Santoro N. Efficacy and Safety of Fezolinetant in Moderate to Severe Vasomotor Symptoms Associated With Menopause: A Phase 3 RCT. J Clin Endocrinol Metab. 2023;108(8):1981-1997.
  149. Pinkerton JV, Simon JA, Joffe H, Maki PM, Nappi RE, Panay N, Soares CN, Thurston RC, Caetano C, Haberland C, Haseli Mashhadi N, Krahn U, Mellinger U, Parke S, Seitz C, Zuurman L. Elinzanetant for the Treatment of Vasomotor Symptoms Associated With Menopause: OASIS 1 and 2 Randomized Clinical Trials. JAMA. 2024.
  150. Skorupskaite K, George JT, Veldhuis JD, Millar RP, Anderson RA. Neurokinin 3 receptor antagonism decreases gonadotropin and testosterone secretion in healthy men. Clin Endocrinol (Oxf). 2017;87(6):748-756.
  151. Navarro VM, Castellano JM, McConkey SM, Pineda R, Ruiz-Pino F, Pinilla L, Clifton DK, Tena-Sempere M, Steiner RA. Interactions between kisspeptin and neurokinin B in the control of GnRH secretion in the female rat. Am J Physiol Endocrinol Metab. 2011;300(1):E202-210.
  152. Garcia-Galiano D, van Ingen Schenau D, Leon S, Krajnc-Franken MA, Manfredi-Lozano M, Romero-Ruiz A, Navarro VM, Gaytan F, van Noort PI, Pinilla L, Blomenrohr M, Tena-Sempere M. Kisspeptin signaling is indispensable for neurokinin B, but not glutamate, stimulation of gonadotropin secretion in mice. Endocrinology. 2012;153(1):316-328.
  153. Wakabayashi Y, Nakada T, Murata K, Ohkura S, Mogi K, Navarro VM, Clifton DK, Mori Y, Tsukamura H, Maeda K, Steiner RA, Okamura H. Neurokinin B and dynorphin A in kisspeptin neurons of the arcuate nucleus participate in generation of periodic oscillation of neural activity driving pulsatile gonadotropin-releasing hormone secretion in the goat. J Neurosci. 2010;30(8):3124-3132.
  154. Ramaswamy S, Seminara SB, Ali B, Ciofi P, Amin NA, Plant TM. Neurokinin B stimulates GnRH release in the male monkey (Macaca mulatta) and is colocalized with kisspeptin in the arcuate nucleus. Endocrinology. 2010;151(9):4494-4503.
  155. Ramaswamy S, Seminara SB, Plant TM. Evidence from the agonadal juvenile male rhesus monkey (Macaca mulatta) for the view that the action of neurokinin B to trigger gonadotropin-releasing hormone release is upstream from the kisspeptin receptor. Neuroendocrinology. 2011;94(3):237-245.
  156. Navarro VM. Interactions between kisspeptins and neurokinin B. Adv Exp Med Biol. 2013;784:325-347.
  157. Navarro VM, Gottsch ML, Chavkin C, Okamura H, Clifton DK, Steiner RA. Regulation of gonadotropin-releasing hormone secretion by kisspeptin/dynorphin/neurokinin B neurons in the arcuate nucleus of the mouse. J Neurosci. 2009;29(38):11859-11866.
  158. Ciofi P, Krause JE, Prins GS, Mazzuca M. Presence of nuclear androgen receptor-like immunoreactivity in neurokinin B-containing neurons of the hypothalamic arcuate nucleus of the adult male rat. Neurosci Lett. 1994;182(2):193-196.
  159. Foradori CD, Coolen LM, Fitzgerald ME, Skinner DC, Goodman RL, Lehman MN. Colocalization of progesterone receptors in parvicellular dynorphin neurons of the ovine preoptic area and hypothalamus. Endocrinology. 2002;143(11):4366-4374.
  160. Smith JT, Cunningham MJ, Rissman EF, Clifton DK, Steiner RA. Regulation of Kiss1 gene expression in the brain of the female mouse. Endocrinology. 2005;146(9):3686-3692.
  161. Smith JT, Acohido BV, Clifton DK, Steiner RA. KiSS-1 neurones are direct targets for leptin in the ob/ob mouse. J Neuroendocrinol. 2006;18(4):298-303.
  162. George JT, Seminara SB. Kisspeptin and the hypothalamic control of reproduction: lessons from the human. Endocrinology. 2012;153(11):5130-5136.
  163. Arreguin-Arevalo JA, Lents CA, Farmerie TA, Nett TM, Clay CM. KiSS-1 peptide induces release of LH by a direct effect on the hypothalamus of ovariectomized ewes. Anim Reprod Sci. 2007;101(3-4):265-275.
  164. Gottsch ML, Cunningham MJ, Smith JT, Popa SM, Acohido BV, Crowley WF, Seminara S, Clifton DK, Steiner RA. A role for kisspeptins in the regulation of gonadotropin secretion in the mouse. Endocrinology. 2004;145(9):4073-4077.
  165. Shahab M, Mastronardi C, Seminara SB, Crowley WF, Ojeda SR, Plant TM. Increased hypothalamic GPR54 signaling: a potential mechanism for initiation of puberty in primates. Proc Natl Acad Sci U S A. 2005;102(6):2129-2134.
  166. Smith JT, Li Q, Pereira A, Clarke IJ. Kisspeptin neurons in the ovine arcuate nucleus and preoptic area are involved in the preovulatory luteinizing hormone surge. Endocrinology. 2009;150(12):5530-5538.
  167. Chan YM, Butler JP, Pinnell NE, Pralong FP, Crowley WF, Jr., Ren C, Chan KK, Seminara SB. Kisspeptin resets the hypothalamic GnRH clock in men. J Clin Endocrinol Metab. 2011;96(6):E908-915.
  168. Chan YM, Butler JP, Sidhoum VF, Pinnell NE, Seminara SB. Kisspeptin administration to women: a window into endogenous kisspeptin secretion and GnRH responsiveness across the menstrual cycle. J Clin Endocrinol Metab. 2012;97(8):E1458-1467.
  169. Dhillo WS, Chaudhri OB, Patterson M, Thompson EL, Murphy KG, Badman MK, McGowan BM, Amber V, Patel S, Ghatei MA, Bloom SR. Kisspeptin-54 stimulates the hypothalamic-pituitary gonadal axis in human males. J Clin Endocrinol Metab. 2005;90(12):6609-6615.
  170. Dhillo WS, Chaudhri OB, Thompson EL, Murphy KG, Patterson M, Ramachandran R, Nijher GK, Amber V, Kokkinos A, Donaldson M, Ghatei MA, Bloom SR. Kisspeptin-54 stimulates gonadotropin release most potently during the preovulatory phase of the menstrual cycle in women. J Clin Endocrinol Metab. 2007;92(10):3958-3966.
  171. George JT, Anderson RA, Millar RP. Kisspeptin-10 stimulation of gonadotrophin secretion in women is modulated by sex steroid feedback. Hum Reprod. 2012;27(12):3552-3559.
  172. George JT, Veldhuis JD, Roseweir AK, Newton CL, Faccenda E, Millar RP, Anderson RA. Kisspeptin-10 is a potent stimulator of LH and increases pulse frequency in men. J Clin Endocrinol Metab. 2011;96(8):E1228-1236.
  173. Jayasena CN, Comninos AN, Nijher GM, Abbara A, De Silva A, Veldhuis JD, Ratnasabapathy R, Izzi-Engbeaya C, Lim A, Patel DA, Ghatei MA, Bloom SR, Dhillo WS. Twice-daily subcutaneous injection of kisspeptin-54 does not abolish menstrual cyclicity in healthy female volunteers. J Clin Endocrinol Metab. 2013;98(11):4464-4474.
  174. Jayasena CN, Comninos AN, Veldhuis JD, Misra S, Abbara A, Izzi-Engbeaya C, Donaldson M, Ghatei MA, Bloom SR, Dhillo WS. A single injection of kisspeptin-54 temporarily increases luteinizing hormone pulsatility in healthy women. Clin Endocrinol (Oxf). 2013;79(4):558-563.
  175. Jayasena CN, Nijher GM, Abbara A, Murphy KG, Lim A, Patel D, Mehta A, Todd C, Donaldson M, Trew GH, Ghatei MA, Bloom SR, Dhillo WS. Twice-weekly administration of kisspeptin-54 for 8 weeks stimulates release of reproductive hormones in women with hypothalamic amenorrhea. Clin Pharmacol Ther. 2010;88(6):840-847.
  176. Jayasena CN, Nijher GM, Chaudhri OB, Murphy KG, Ranger A, Lim A, Patel D, Mehta A, Todd C, Ramachandran R, Salem V, Stamp GW, Donaldson M, Ghatei MA, Bloom SR, Dhillo WS. Subcutaneous injection of kisspeptin-54 acutely stimulates gonadotropin secretion in women with hypothalamic amenorrhea, but chronic administration causes tachyphylaxis. J Clin Endocrinol Metab. 2009;94(11):4315-4323.
  177. Jayasena CN, Nijher GM, Comninos AN, Abbara A, Januszewki A, Vaal ML, Sriskandarajah L, Murphy KG, Farzad Z, Ghatei MA, Bloom SR, Dhillo WS. The effects of kisspeptin-10 on reproductive hormone release show sexual dimorphism in humans. J Clin Endocrinol Metab. 2011;96(12):E1963-1972.
  178. Abbara A, Narayanaswamy S, Izzi-Engbeaya C, Comninos AN, Clarke SA, Malik Z, Papadopoulou D, Clobentz A, Sarang Z, Bassett P, Jayasena CN, Dhillo WS. Hypothalamic Response to Kisspeptin-54 and Pituitary Response to Gonadotropin-Releasing Hormone Are Preserved in Healthy Older Men. Neuroendocrinology. 2018;106(4):401-410.
  179. George JT, Veldhuis JD, Tena-Sempere M, Millar RP, Anderson RA. Exploring the pathophysiology of hypogonadism in men with type 2 diabetes: kisspeptin-10 stimulates serum testosterone and LH secretion in men with type 2 diabetes and mild biochemical hypogonadism. Clin Endocrinol (Oxf). 2013;79(1):100-104.
  180. Jayasena CN, Abbara A, Veldhuis JD, Comninos AN, Ratnasabapathy R, De Silva A, Nijher GM, Ganiyu-Dada Z, Mehta A, Todd C, Ghatei MA, Bloom SR, Dhillo WS. Increasing LH pulsatility in women with hypothalamic amenorrhoea using intravenous infusion of Kisspeptin-54. J Clin Endocrinol Metab. 2014;99(6):E953-961.
  181. Abbara A, Eng PC, Phylactou M, Clarke SA, Richardson R, Sykes CM, Phumsatitpong C, Mills E, Modi M, Izzi-Engbeaya C, Papadopoulou D, Purugganan K, Jayasena CN, Webber L, Salim R, Owen B, Bech P, Comninos AN, McArdle CA, Voliotis M, Tsaneva-Atanasova K, Moenter S, Hanyaloglu A, Dhillo WS. Kisspeptin receptor agonist has therapeutic potential for female reproductive disorders. J Clin Invest. 2020;130(12):6739-6753.
  182. Szeliga A, Podfigurna A, Bala G, Meczekalski B. Kisspeptin and neurokinin B analogs use in gynecological endocrinology: where do we stand? J Endocrinol Invest. 2020;43(5):555-561.
  183. Abbara A, Ufer M, Voors-Pette C, Berman L, Ezzati M, Wu R, Lee TY, Ferreira JCA, Migoya E, Dhillo WS. Endocrine profile of the kisspeptin receptor agonist MVT-602 in healthy premenopausal women with and without ovarian stimulation: results from 2 randomized, placebo-controlled clinical tricals. Fertil Steril. 2024;121(1):95-106.
  184. Li XF, Kinsey-Jones JS, Cheng Y, Knox AM, Lin Y, Petrou NA, Roseweir A, Lightman SL, Milligan SR, Millar RP, O'Byrne KT. Kisspeptin signalling in the hypothalamic arcuate nucleus regulates GnRH pulse generator frequency in the rat. PLoS One. 2009;4(12):e8334.
  185. Millar RP, Roseweir AK, Tello JA, Anderson RA, George JT, Morgan K, Pawson AJ. Kisspeptin antagonists: unraveling the role of kisspeptin in reproductive physiology. Brain Res. 2010;1364:81-89.
  186. Roseweir AK, Kauffman AS, Smith JT, Guerriero KA, Morgan K, Pielecka-Fortuna J, Pineda R, Gottsch ML, Tena-Sempere M, Moenter SM, Terasawa E, Clarke IJ, Steiner RA, Millar RP. Discovery of potent kisspeptin antagonists delineate physiological mechanisms of gonadotropin regulation. J Neurosci. 2009;29(12):3920-3929.
  187. Thompson EL, Patterson M, Murphy KG, Smith KL, Dhillo WS, Todd JF, Ghatei MA, Bloom SR. Central and peripheral administration of kisspeptin-10 stimulates the hypothalamic-pituitary-gonadal axis. J Neuroendocrinol. 2004;16(10):850-858.
  188. Tovar S, Vazquez MJ, Navarro VM, Fernandez-Fernandez R, Castellano JM, Vigo E, Roa J, Casanueva FF, Aguilar E, Pinilla L, Dieguez C, Tena-Sempere M. Effects of single or repeated intravenous administration of kisspeptin upon dynamic LH secretion in conscious male rats. Endocrinology. 2006;147(6):2696-2704.
  189. Novaira HJ, Ng Y, Wolfe A, Radovick S. Kisspeptin increases GnRH mRNA expression and secretion in GnRH secreting neuronal cell lines. Mol Cell Endocrinol. 2009;311(1-2):126-134.
  190. Chan YM, Lippincott MF, Butler JP, Sidhoum VF, Li CX, Plummer L, Seminara SB. Exogenous kisspeptin administration as a probe of GnRH neuronal function in patients with idiopathic hypogonadotropic hypogonadism. J Clin Endocrinol Metab. 2014;99(12):E2762-2771.
  191. Mittelman-Smith MA, Krajewski-Hall SJ, McMullen NT, Rance NE. Ablation of KNDy Neurons Results in Hypogonadotropic Hypogonadism and Amplifies the Steroid-Induced LH Surge in Female Rats. Endocrinology. 2016;157(5):2015-2027.
  192. Gutierrez-Pascual E, Martinez-Fuentes AJ, Pinilla L, Tena-Sempere M, Malagon MM, Castano JP. Direct pituitary effects of kisspeptin: activation of gonadotrophs and somatotrophs and stimulation of luteinising hormone and growth hormone secretion. J Neuroendocrinol. 2007;19(7):521-530.
  193. Richard N, Galmiche G, Corvaisier S, Caraty A, Kottler ML. KiSS-1 and GPR54 genes are co-expressed in rat gonadotrophs and differentially regulated in vivo by oestradiol and gonadotrophin-releasing hormone. J Neuroendocrinol. 2008;20(3):381-393.
  194. Smith JT, Coolen LM, Kriegsfeld LJ, Sari IP, Jaafarzadehshirazi MR, Maltby M, Bateman K, Goodman RL, Tilbrook AJ, Ubuka T, Bentley GE, Clarke IJ, Lehman MN. Variation in kisspeptin and RFamide-related peptide (RFRP) expression and terminal connections to gonadotropin-releasing hormone neurons in the brain: a novel medium for seasonal breeding in the sheep. Endocrinology. 2008;149(11):5770-5782.
  195. Smith JT, Rao A, Pereira A, Caraty A, Millar RP, Clarke IJ. Kisspeptin is present in ovine hypophysial portal blood but does not increase during the preovulatory luteinizing hormone surge: evidence that gonadotropes are not direct targets of kisspeptin in vivo. Endocrinology. 2008;149(4):1951-1959.
  196. Witham EA, Meadows JD, Hoffmann HM, Shojaei S, Coss D, Kauffman AS, Mellon PL. Kisspeptin regulates gonadotropin genes via immediate early gene induction in pituitary gonadotropes. Mol Endocrinol. 2013;27(8):1283-1294.
  197. Pampillo M, Camuso N, Taylor JE, Szereszewski JM, Ahow MR, Zajac M, Millar RP, Bhattacharya M, Babwah AV. Regulation of GPR54 signaling by GRK2 and {beta}-arrestin. Mol Endocrinol. 2009;23(12):2060-2074.
  198. Ramaswamy S, Seminara SB, Pohl CR, DiPietro MJ, Crowley WF, Jr., Plant TM. Effect of continuous intravenous administration of human metastin 45-54 on the neuroendocrine activity of the hypothalamic-pituitary-testicular axis in the adult male rhesus monkey (Macaca mulatta). Endocrinology. 2007;148(7):3364-3370.
  199. Seminara SB, Dipietro MJ, Ramaswamy S, Crowley WF, Jr., Plant TM. Continuous human metastin 45-54 infusion desensitizes G protein-coupled receptor 54-induced gonadotropin-releasing hormone release monitored indirectly in the juvenile male Rhesus monkey (Macaca mulatta): a finding with therapeutic implications. Endocrinology. 2006;147(5):2122-2126.
  200. Thompson EL, Murphy KG, Patterson M, Bewick GA, Stamp GW, Curtis AE, Cooke JH, Jethwa PH, Todd JF, Ghatei MA, Bloom SR. Chronic subcutaneous administration of kisspeptin-54 causes testicular degeneration in adult male rats. Am J Physiol Endocrinol Metab. 2006;291(5):E1074-1082.
  201. Lippincott MFS, V.F.; Seminara, S. Continuously administered kisspeptin as a pseudo-antagonist of the kisspeptin receptor. The Endocrine Society´s 95th Annual Meeting, ENDO 2013, San Francisco, Abstract SAT 2134-2163. 2013.
  202. MacLean DB, Matsui H, Suri A, Neuwirth R, Colombel M. Sustained exposure to the investigational Kisspeptin analog, TAK-448, down-regulates testosterone into the castration range in healthy males and in patients with prostate cancer: results from two phase 1 studies. J Clin Endocrinol Metab. 2014;99(8):E1445-1453.
  203. Scott G, Ahmad I, Howard K, MacLean D, Oliva C, Warrington S, Wilbraham D, Worthington P. Double-blind, randomized, placebo-controlled study of safety, tolerability, pharmacokinetics and pharmacodynamics of TAK-683, an investigational metastin analogue in healthy men. Br J Clin Pharmacol. 2013;75(2):381-391.
  204. Christian CA, Moenter SM. The neurobiology of preovulatory and estradiol-induced gonadotropin-releasing hormone surges. Endocr Rev. 2010;31(4):544-577.
  205. Kauffman AS. Coming of age in the kisspeptin era: sex differences, development, and puberty. Mol Cell Endocrinol. 2010;324(1-2):51-63.
  206. Terasawa E, Garcia JP, Seminara SB, Keen KL. Role of Kisspeptin and Neurokinin B in Puberty in Female Non-Human Primates. Front Endocrinol (Lausanne). 2018;9:148.
  207. Shahab M, Lippincott M, Chan YM, Davies A, Merino PM, Plummer L, Mericq V, Seminara S. Discordance in the Dependence on Kisspeptin Signaling in Mini Puberty vs Adolescent Puberty: Human Genetic Evidence. J Clin Endocrinol Metab. 2018;103(4):1273-1276.
  208. Navarro VM, Castellano JM, Fernandez-Fernandez R, Barreiro ML, Roa J, Sanchez-Criado JE, Aguilar E, Dieguez C, Pinilla L, Tena-Sempere M. Developmental and hormonally regulated messenger ribonucleic acid expression of KiSS-1 and its putative receptor, GPR54, in rat hypothalamus and potent luteinizing hormone-releasing activity of KiSS-1 peptide. Endocrinology. 2004;145(10):4565-4574.
  209. Keen KL, Wegner FH, Bloom SR, Ghatei MA, Terasawa E. An increase in kisspeptin-54 release occurs with the pubertal increase in luteinizing hormone-releasing hormone-1 release in the stalk-median eminence of female rhesus monkeys in vivo. Endocrinology. 2008;149(8):4151-4157.
  210. Plant TM, Ramaswamy S, Dipietro MJ. Repetitive activation of hypothalamic G protein-coupled receptor 54 with intravenous pulses of kisspeptin in the juvenile monkey (Macaca mulatta) elicits a sustained train of gonadotropin-releasing hormone discharges. Endocrinology. 2006;147(2):1007-1013.
  211. Pineda R, Garcia-Galiano D, Roseweir A, Romero M, Sanchez-Garrido MA, Ruiz-Pino F, Morgan K, Pinilla L, Millar RP, Tena-Sempere M. Critical roles of kisspeptins in female puberty and preovulatory gonadotropin surges as revealed by a novel antagonist. Endocrinology. 2010;151(2):722-730.
  212. Decourt C, Robert V, Anger K, Galibert M, Madinier JB, Liu X, Dardente H, Lomet D, Delmas AF, Caraty A, Herbison AE, Anderson GM, Aucagne V, Beltramo M. A synthetic kisspeptin analog that triggers ovulation and advances puberty. Sci Rep. 2016;6:26908.
  213. Novaira HJ, Sonko ML, Hoffman G, Koo Y, Ko C, Wolfe A, Radovick S. Disrupted kisspeptin signaling in GnRH neurons leads to hypogonadotrophic hypogonadism. Mol Endocrinol. 2014;28(2):225-238.
  214. Lippincott MF, Chan YM, Delaney A, Rivera-Morales D, Butler JP, Seminara SB. Kisspeptin Responsiveness Signals Emergence of Reproductive Endocrine Activity: Implications for Human Puberty. J Clin Endocrinol Metab. 2016;101(8):3061-3069.
  215. Chan YM, Lippincott MF, Kusa TO, Seminara SB. Divergent responses to kisspeptin in children with delayed puberty. JCI Insight. 2018;3(8).
  216. Sobrino V, Avendano MS, Perdices-Lopez C, Jimenez-Puyer M, Tena-Sempere M. Kisspeptins and the neuroendocrine control of reproduction: Recent progress and new frontiers in kisspeptin research. Front Neuroendocrinol. 2022;65:100977.
  217. Garcia JP, Guerriero KA, Keen KL, Kenealy BP, Seminara SB, Terasawa E. Kisspeptin and Neurokinin B Signaling Network Underlies the Pubertal Increase in GnRH Release in Female Rhesus Monkeys. Endocrinology. 2017;158(10):3269-3280.
  218. Midgley AR, Jr., Jaffe RB. Regulation of human gonadotropins. X. Episodic fluctuation of LH during the menstrual cycle. J Clin Endocrinol Metab. 1971;33(6):962-969.
  219. Levine JE, Bauer-Dantoin AC, Besecke LM, Conaghan LA, Legan SJ, Meredith JM, Strobl FJ, Urban JH, Vogelsong KM, Wolfe AM. Neuroendocrine regulation of the luteinizing hormone-releasing hormone pulse generator in the rat. Recent Prog Horm Res. 1991;47:97-151; discussion 151-153.
  220. Clarke IJ. Gonadotrophin-releasing hormone secretion (GnRH) in anoestrous ewes and the induction of GnRH surges by oestrogen. J Endocrinol. 1988;117(3):355-360.
  221. Pau KY, Berria M, Hess DL, Spies HG. Preovulatory gonadotropin-releasing hormone surge in ovarian-intact rhesus macaques. Endocrinology. 1993;133(4):1650-1656.
  222. Knobil E, Plant TM. The hypothalamic regulation of LH and FSH secretion in the rhesus monkey. Res Publ Assoc Res Nerv Ment Dis. 1978;56:359-372.
  223. Hall JE, Taylor AE, Martin KA, Rivier J, Schoenfeld DA, Crowley WF, Jr. Decreased release of gonadotropin-releasing hormone during the preovulatory midcycle luteinizing hormone surge in normal women. Proc Natl Acad Sci U S A. 1994;91(15):6894-6898.
  224. Bagatell CJ, Dahl KD, Bremner WJ. The direct pituitary effect of testosterone to inhibit gonadotropin secretion in men is partially mediated by aromatization to estradiol. J Androl. 1994;15(1):15-21.
  225. Emons G, Ortmann O, Thiessen S, Knuppen R. Effects of estradiol and some antiestrogens (clomiphene, tamoxifen, and hydroxytamoxifen) on luteinizing hormone secretion by rat pituitary cells in culture. Arch Gynecol. 1986;237(4):199-211.
  226. Finkelstein JS, O'Dea LS, Whitcomb RW, Crowley WF, Jr. Sex steroid control of gonadotropin secretion in the human male. II. Effects of estradiol administration in normal and gonadotropin-releasing hormone-deficient men. J Clin Endocrinol Metab. 1991;73(3):621-628.
  227. Hayes FJ, Seminara SB, Decruz S, Boepple PA, Crowley WF, Jr. Aromatase inhibition in the human male reveals a hypothalamic site of estrogen feedback. J Clin Endocrinol Metab. 2000;85(9):3027-3035.
  228. Kerin JF, Liu JH, Phillipou G, Yen SS. Evidence for a hypothalamic site of action of clomiphene citrate in women. J Clin Endocrinol Metab. 1985;61(2):265-268.
  229. Kesner JS, Wilson RC, Kaufman JM, Hotchkiss J, Chen Y, Yamamoto H, Pardo RR, Knobil E. Unexpected responses of the hypothalamic gonadotropin-releasing hormone "pulse generator" to physiological estradiol inputs in the absence of the ovary. Proc Natl Acad Sci U S A. 1987;84(23):8745-8749.
  230. Pitteloud N, Dwyer AA, DeCruz S, Lee H, Boepple PA, Crowley WF, Jr., Hayes FJ. Inhibition of luteinizing hormone secretion by testosterone in men requires aromatization for its pituitary but not its hypothalamic effects: evidence from the tandem study of normal and gonadotropin-releasing hormone-deficient men. J Clin Endocrinol Metab. 2008;93(3):784-791.
  231. Veldhuis JD, Evans WS, Rogol AD, Thorner MO, Stumpf P. Actions of estradiol on discrete attributes of the luteinizing hormone pulse signal in man. Studies in postmenopausal women treated with pure estradiol. J Clin Invest. 1987;79(3):769-776.
  232. Herbison AE. Multimodal influence of estrogen upon gonadotropin-releasing hormone neurons. Endocr Rev. 1998;19(3):302-330.
  233. McDevitt MA, Glidewell-Kenney C, Jimenez MA, Ahearn PC, Weiss J, Jameson JL, Levine JE. New insights into the classical and non-classical actions of estrogen: evidence from estrogen receptor knock-out and knock-in mice. Mol Cell Endocrinol. 2008;290(1-2):24-30.
  234. Rance NE, McMullen NT, Smialek JE, Price DL, Young WS, 3rd. Postmenopausal hypertrophy of neurons expressing the estrogen receptor gene in the human hypothalamus. J Clin Endocrinol Metab. 1990;71(1):79-85.
  235. Rance NE, Young WS, 3rd. Hypertrophy and increased gene expression of neurons containing neurokinin-B and substance-P messenger ribonucleic acids in the hypothalami of postmenopausal women. Endocrinology. 1991;128(5):2239-2247.
  236. Rometo AM, Rance NE. Changes in prodynorphin gene expression and neuronal morphology in the hypothalamus of postmenopausal women. J Neuroendocrinol. 2008;20(12):1376-1381.
  237. Franceschini I, Lomet D, Cateau M, Delsol G, Tillet Y, Caraty A. Kisspeptin immunoreactive cells of the ovine preoptic area and arcuate nucleus co-express estrogen receptor alpha. Neurosci Lett. 2006;401(3):225-230.
  238. Abel TW, Voytko ML, Rance NE. The effects of hormone replacement therapy on hypothalamic neuropeptide gene expression in a primate model of menopause. J Clin Endocrinol Metab. 1999;84(6):2111-2118.
  239. Eghlidi DH, Haley GE, Noriega NC, Kohama SG, Urbanski HF. Influence of age and 17beta-estradiol on kisspeptin, neurokinin B, and prodynorphin gene expression in the arcuate-median eminence of female rhesus macaques. Endocrinology. 2010;151(8):3783-3794.
  240. Goodman RL, Holaskova I, Nestor CC, Connors JM, Billings HJ, Valent M, Lehman MN, Hileman SM. Evidence that the arcuate nucleus is an important site of progesterone negative feedback in the ewe. Endocrinology. 2011;152(9):3451-3460.
  241. Jayasena CN, Abbara A, Comninos AN, Nijher GM, Christopoulos G, Narayanaswamy S, Izzi-Engbeaya C, Sridharan M, Mason AJ, Warwick J, Ashby D, Ghatei MA, Bloom SR, Carby A, Trew GH, Dhillo WS. Kisspeptin-54 triggers egg maturation in women undergoing in vitro fertilization. J Clin Invest. 2014;124(8):3667-3677.
  242. Sandoval-Guzman T, Rance NE. Central injection of senktide, an NK3 receptor agonist, or neuropeptide Y inhibits LH secretion and induces different patterns of Fos expression in the rat hypothalamus. Brain Res. 2004;1026(2):307-312.
  243. Lippincott MF, Chan YM, Rivera Morales D, Seminara SB. Continuous Kisspeptin Administration in Postmenopausal Women: Impact of Estradiol on Luteinizing Hormone Secretion. J Clin Endocrinol Metab. 2017;102(6):2091-2099.
  244. Clarkson J, d'Anglemont de Tassigny X, Moreno AS, Colledge WH, Herbison AE. Kisspeptin-GPR54 signaling is essential for preovulatory gonadotropin-releasing hormone neuron activation and the luteinizing hormone surge. J Neurosci. 2008;28(35):8691-8697.
  245. Kinoshita M, Tsukamura H, Adachi S, Matsui H, Uenoyama Y, Iwata K, Yamada S, Inoue K, Ohtaki T, Matsumoto H, Maeda K. Involvement of central metastin in the regulation of preovulatory luteinizing hormone surge and estrous cyclicity in female rats. Endocrinology. 2005;146(10):4431-4436.
  246. Watson RE, Jr., Langub MC, Jr., Engle MG, Maley BE. Estrogen-receptive neurons in the anteroventral periventricular nucleus are synaptic targets of the suprachiasmatic nucleus and peri-suprachiasmatic region. Brain Res. 1995;689(2):254-264.
  247. Kumar D, Candlish M, Periasamy V, Avcu N, Mayer C, Boehm U. Specialized subpopulations of kisspeptin neurons communicate with GnRH neurons in female mice. Endocrinology. 2015;156(1):32-38.
  248. Billings HJ, Connors JM, Altman SN, Hileman SM, Holaskova I, Lehman MN, McManus CJ, Nestor CC, Jacobs BH, Goodman RL. Neurokinin B acts via the neurokinin-3 receptor in the retrochiasmatic area to stimulate luteinizing hormone secretion in sheep. Endocrinology. 2010;151(8):3836-3846.
  249. Li Q, Millar RP, Clarke IJ, Smith JT. Evidence that Neurokinin B Controls Basal Gonadotropin-Releasing Hormone Secretion but Is Not Critical for Estrogen-Positive Feedback in Sheep. Neuroendocrinology. 2015;101(2):161-174.
  250. Terasawa E. Neuroestradiol in regulation of GnRH release. Horm Behav. 2018.
  251. Soules MR, Steiner RA, Clifton DK, Cohen NL, Aksel S, Bremner WJ. Progesterone modulation of pulsatile luteinizing hormone secretion in normal women. J Clin Endocrinol Metab. 1984;58(2):378-383.
  252. Skinner DC, Evans NP, Delaleu B, Goodman RL, Bouchard P, Caraty A. The negative feedback actions of progesterone on gonadotropin-releasing hormone secretion are transduced by the classical progesterone receptor. Proc Natl Acad Sci U S A. 1998;95(18):10978-10983.
  253. Fox SR, Harlan RE, Shivers BD, Pfaff DW. Chemical characterization of neuroendocrine targets for progesterone in the female rat brain and pituitary. Neuroendocrinology. 1990;51(3):276-283.
  254. King JC, Tai DW, Hanna IK, Pfeiffer A, Haas P, Ronsheim PM, Mitchell SC, Turcotte JC, Blaustein JD. A subgroup of LHRH neurons in guinea pigs with progestin receptors is centrally positioned within the total population of LHRH neurons. Neuroendocrinology. 1995;61(3):265-275.
  255. Skinner DC, Caraty A, Allingham R. Unmasking the progesterone receptor in the preoptic area and hypothalamus of the ewe: no colocalization with gonadotropin-releasing neurons. Endocrinology. 2001;142(2):573-579.
  256. Foradori CD, Goodman RL, Adams VL, Valent M, Lehman MN. Progesterone increases dynorphin a concentrations in cerebrospinal fluid and preprodynorphin messenger ribonucleic Acid levels in a subset of dynorphin neurons in the sheep. Endocrinology. 2005;146(4):1835-1842.
  257. Matsumoto AM, Bremner WJ. Modulation of pulsatile gonadotropin secretion by testosterone in man. J Clin Endocrinol Metab. 1984;58(4):609-614.
  258. Herbison AE, Skinner DC, Robinson JE, King IS. Androgen receptor-immunoreactive cells in ram hypothalamus: distribution and co-localization patterns with gonadotropin-releasing hormone, somatostatin and tyrosine hydroxylase. Neuroendocrinology. 1996;63(2):120-131.
  259. Smith JT, Dungan HM, Stoll EA, Gottsch ML, Braun RE, Eacker SM, Clifton DK, Steiner RA. Differential regulation of KiSS-1 mRNA expression by sex steroids in the brain of the male mouse. Endocrinology. 2005;146(7):2976-2984.
  260. Woodham C, Birch L, Prins GS. Neonatal estrogen down-regulates prostatic androgen receptor through a proteosome-mediated protein degradation pathway. Endocrinology. 2003;144(11):4841-4850.
  261. Kumar MV, Leo ME, Tindall DJ. Modulation of androgen receptor transcriptional activity by the estrogen receptor. J Androl. 1994;15(6):534-542.
  262. Navarro VM, Gottsch ML, Wu M, Garcia-Galiano D, Hobbs SJ, Bosch MA, Pinilla L, Clifton DK, Dearth A, Ronnekleiv OK, Braun RE, Palmiter RD, Tena-Sempere M, Alreja M, Steiner RA. Regulation of NKB pathways and their roles in the control of Kiss1 neurons in the arcuate nucleus of the male mouse. Endocrinology. 2011;152(11):4265-4275.
  263. Shibata M, Friedman RL, Ramaswamy S, Plant TM. Evidence that down regulation of hypothalamic KiSS-1 expression is involved in the negative feedback action of testosterone to regulate luteinising hormone secretion in the adult male rhesus monkey (Macaca mulatta). J Neuroendocrinol. 2007;19(6):432-438.
  264. Chrousos GP, Torpy DJ, Gold PW. Interactions between the hypothalamic-pituitary-adrenal axis and the female reproductive system: clinical implications. Ann Intern Med. 1998;129(3):229-240.
  265. Kalantaridou SN, Makrigiannakis A, Zoumakis E, Chrousos GP. Stress and the female reproductive system. J Reprod Immunol. 2004;62(1-2):61-68.
  266. Barron JL, Noakes TD, Levy W, Smith C, Millar RP. Hypothalamic dysfunction in overtrained athletes. J Clin Endocrinol Metab. 1985;60(4):803-806.
  267. Berga SL, Mortola JF, Girton L, Suh B, Laughlin G, Pham P, Yen SS. Neuroendocrine aberrations in women with functional hypothalamic amenorrhea. J Clin Endocrinol Metab. 1989;68(2):301-308.
  268. Boccuzzi G, Angeli A, Bisbocci D, Fonzo D, Giadano GP, Ceresa F. Effect of synthetic luteinizing hormone releasing hormone (LH-RH) on the release of gonadotropins in Cushing's disease. J Clin Endocrinol Metab. 1975;40(5):892-895.
  269. Breen KM, Oakley AE, Pytiak AV, Tilbrook AJ, Wagenmaker ER, Karsch FJ. Does cortisol acting via the type II glucocorticoid receptor mediate suppression of pulsatile luteinizing hormone secretion in response to psychosocial stress? Endocrinology. 2007;148(4):1882-1890.
  270. Luger A, Deuster PA, Kyle SB, Gallucci WT, Montgomery LC, Gold PW, Loriaux DL, Chrousos GP. Acute hypothalamic-pituitary-adrenal responses to the stress of treadmill exercise. Physiologic adaptations to physical training. N Engl J Med. 1987;316(21):1309-1315.
  271. Luton JP, Thieblot P, Valcke JC, Mahoudeau JA, Bricaire H. Reversible gonadotropin deficiency in male Cushing's disease. J Clin Endocrinol Metab. 1977;45(3):488-495.
  272. Oakley AE, Breen KM, Clarke IJ, Karsch FJ, Wagenmaker ER, Tilbrook AJ. Cortisol reduces gonadotropin-releasing hormone pulse frequency in follicular phase ewes: influence of ovarian steroids. Endocrinology. 2009;150(1):341-349.
  273. Saketos M, Sharma N, Santoro NF. Suppression of the hypothalamic-pituitary-ovarian axis in normal women by glucocorticoids. Biol Reprod. 1993;49(6):1270-1276.
  274. Breen KM, Karsch FJ. New insights regarding glucocorticoids, stress and gonadotropin suppression. Front Neuroendocrinol. 2006;27(2):233-245.
  275. Chen MD, Ordog T, O'Byrne KT, Goldsmith JR, Connaughton MA, Knobil E. The insulin hypoglycemia-induced inhibition of gonadotropin-releasing hormone pulse generator activity in the rhesus monkey: roles of vasopressin and corticotropin-releasing factor. Endocrinology. 1996;137(5):2012-2021.
  276. Kinsey-Jones JS, Li XF, Knox AM, Wilkinson ES, Zhu XL, Chaudhary AA, Milligan SR, Lightman SL, O'Byrne KT. Down-regulation of hypothalamic kisspeptin and its receptor, Kiss1r, mRNA expression is associated with stress-induced suppression of luteinising hormone secretion in the female rat. J Neuroendocrinol. 2009;21(1):20-29.
  277. Takumi K, Iijima N, Higo S, Ozawa H. Immunohistochemical analysis of the colocalization of corticotropin-releasing hormone receptor and glucocorticoid receptor in kisspeptin neurons in the hypothalamus of female rats. Neurosci Lett. 2012;531(1):40-45.
  278. Phumsatitpong C, Moenter SM. Estradiol-Dependent Stimulation and Suppression of Gonadotropin-Releasing Hormone Neuron Firing Activity by Corticotropin-Releasing Hormone in Female Mice. Endocrinology. 2018;159(1):414-425.
  279. Mitchell JC, Li XF, Breen L, Thalabard JC, O'Byrne KT. The role of the locus coeruleus in corticotropin-releasing hormone and stress-induced suppression of pulsatile luteinizing hormone secretion in the female rat. Endocrinology. 2005;146(1):323-331.
  280. Choi YJ, Habibi HR, Kil GS, Jung MM, Choi CY. Effect of cortisol on gonadotropin inhibitory hormone (GnIH) in the cinnamon clownfish, Amphiprion melanopus. Biochem Biophys Res Commun. 2017;485(2):342-348.
  281. Bohnet HG, Dahlen HG, Wuttke W, Schneider HP. Hyperprolactinemic anovulatory syndrome. J Clin Endocrinol Metab. 1976;42(1):132-143.
  282. Diaz S, Seron-Ferre M, Cardenas H, Schiappacasse V, Brandeis A, Croxatto HB. Circadian variation of basal plasma prolactin, prolactin response to suckling, and length of amenorrhea in nursing women. J Clin Endocrinol Metab. 1989;68(5):946-955.
  283. Molitch ME. Pregnancy and the hyperprolactinemic woman. N Engl J Med. 1985;312(21):1364-1370.
  284. De Rosa M, Zarrilli S, Di Sarno A, Milano N, Gaccione M, Boggia B, Lombardi G, Colao A. Hyperprolactinemia in men: clinical and biochemical features and response to treatment. Endocrine. 2003;20(1-2):75-82.
  285. Moult PJ, Rees LH, Besser GM. Pulsatile gonadotrophin secretion in hyperprolactinaemic amenorrhoea an the response to bromocriptine therapy. Clin Endocrinol (Oxf). 1982;16(2):153-162.
  286. Lecomte P, Lecomte C, Lansac J, Gallier J, Sonier CB, Simonetta C. Pregnancy after intravenous pulsatile gonadotropin-releasing hormone in a hyperprolactinaemic woman resistant to treatment with dopamine agonists. Eur J Obstet Gynecol Reprod Biol. 1997;74(2):219-221.
  287. Polson DW, Sagle M, Mason HD, Adams J, Jacobs HS, Franks S. Ovulation and normal luteal function during LHRH treatment of women with hyperprolactinaemic amenorrhoea. Clin Endocrinol (Oxf). 1986;24(5):531-537.
  288. Grattan DR, Jasoni CL, Liu X, Anderson GM, Herbison AE. Prolactin regulation of gonadotropin-releasing hormone neurons to suppress luteinizing hormone secretion in mice. Endocrinology. 2007;148(9):4344-4351.
  289. Milenkovic L, D'Angelo G, Kelly PA, Weiner RI. Inhibition of gonadotropin hormone-releasing hormone release by prolactin from GT1 neuronal cell lines through prolactin receptors. Proc Natl Acad Sci U S A. 1994;91(4):1244-1247.
  290. Kolbinger W, Beyer C, Fohr K, Reisert I, Pilgrim C. Diencephalic GABAergic neurons in vitro respond to prolactin with a rapid increase in intracellular free calcium. Neuroendocrinology. 1992;56(2):148-152.
  291. Sarkar DK, Yen SS. Hyperprolactinemia decreases the luteinizing hormone-releasing hormone concentration in pituitary portal plasma: a possible role for beta-endorphin as a mediator. Endocrinology. 1985;116(5):2080-2084.
  292. Li C, Chen P, Smith MS. Neuropeptide Y and tuberoinfundibular dopamine activities are altered during lactation: role of prolactin. Endocrinology. 1999;140(1):118-123.
  293. Moore KE. Interactions between prolactin and dopaminergic neurons. Biol Reprod. 1987;36(1):47-58.
  294. Kokay IC, Petersen SL, Grattan DR. Identification of prolactin-sensitive GABA and kisspeptin neurons in regions of the rat hypothalamus involved in the control of fertility. Endocrinology. 2011;152(2):526-535.
  295. Szawka RE, Ribeiro AB, Leite CM, Helena CV, Franci CR, Anderson GM, Hoffman GE, Anselmo-Franci JA. Kisspeptin regulates prolactin release through hypothalamic dopaminergic neurons. Endocrinology. 2010;151(7):3247-3257.
  296. Yamada S, Uenoyama Y, Kinoshita M, Iwata K, Takase K, Matsui H, Adachi S, Inoue K, Maeda KI, Tsukamura H. Inhibition of metastin (kisspeptin-54)-GPR54 signaling in the arcuate nucleus-median eminence region during lactation in rats. Endocrinology. 2007;148(5):2226-2232.
  297. Sonigo C, Bouilly J, Carre N, Tolle V, Caraty A, Tello J, Simony-Conesa FJ, Millar R, Young J, Binart N. Hyperprolactinemia-induced ovarian acyclicity is reversed by kisspeptin administration. J Clin Invest. 2012;122(10):3791-3795.
  298. Araujo-Lopes R, Crampton JR, Aquino NS, Miranda RM, Kokay IC, Reis AM, Franci CR, Grattan DR, Szawka RE. Prolactin regulates kisspeptin neurons in the arcuate nucleus to suppress LH secretion in female rats. Endocrinology. 2014;155(3):1010-1020.
  299. Brown RS, Herbison AE, Grattan DR. Prolactin regulation of kisspeptin neurones in the mouse brain and its role in the lactation-induced suppression of kisspeptin expression. J Neuroendocrinol. 2014;26(12):898-908.
  300. Millar RP, Sonigo C, Anderson RA, George J, Maione L, Brailly-Tabard S, Chanson P, Binart N, Young J. Hypothalamic-Pituitary-Ovarian Axis Reactivation by Kisspeptin-10 in Hyperprolactinemic Women With Chronic Amenorrhea. J Endocr Soc. 2017;1(11):1362-1371.
  301. Aquino NSS, Araujo-Lopes R, Henriques PC, Lopes FEF, Gusmao DO, Coimbra CC, Franci CR, Reis AM, Szawka RE. alpha-Estrogen and Progesterone Receptors Modulate Kisspeptin Effects on Prolactin: Role in Estradiol-Induced Prolactin Surge in Female Rats. Endocrinology. 2017;158(6):1812-1826.
  302. Schneider JE. Energy balance and reproduction. Physiol Behav. 2004;81(2):289-317.
  303. Yeo SH, Colledge WH. The Role of Kiss1 Neurons As Integrators of Endocrine, Metabolic, and Environmental Factors in the Hypothalamic-Pituitary-Gonadal Axis. Front Endocrinol (Lausanne). 2018;9:188.
  304. Dandona P, Dhindsa S. Update: Hypogonadotropic hypogonadism in type 2 diabetes and obesity. J Clin Endocrinol Metab. 2011;96(9):2643-2651.
  305. George JT, Millar RP, Anderson RA. Hypothesis: kisspeptin mediates male hypogonadism in obesity and type 2 diabetes. Neuroendocrinology. 2010;91(4):302-307.
  306. Bergendahl M, Aloi JA, Iranmanesh A, Mulligan TM, Veldhuis JD. Fasting suppresses pulsatile luteinizing hormone (LH) secretion and enhances orderliness of LH release in young but not older men. J Clin Endocrinol Metab. 1998;83(6):1967-1975.
  307. Bergendahl M, Veldhuis JD. Altered pulsatile gonadotropin signaling in nutritional deficiency in the male. Trends Endocrinol Metab. 1995;6(5):145-159.
  308. Weigle DS, Duell PB, Connor WE, Steiner RA, Soules MR, Kuijper JL. Effect of fasting, refeeding, and dietary fat restriction on plasma leptin levels. J Clin Endocrinol Metab. 1997;82(2):561-565.
  309. Loucks AB, Thuma JR. Luteinizing hormone pulsatility is disrupted at a threshold of energy availability in regularly menstruating women. J Clin Endocrinol Metab. 2003;88(1):297-311.
  310. Loucks AB, Verdun M, Heath EM. Low energy availability, not stress of exercise, alters LH pulsatility in exercising women. J Appl Physiol (1985). 1998;84(1):37-46.
  311. Schreihofer DA, Amico JA, Cameron JL. Reversal of fasting-induced suppression of luteinizing hormone (LH) secretion in male rhesus monkeys by intragastric nutrient infusion: evidence for rapid stimulation of LH by nutritional signals. Endocrinology. 1993;132(5):1890-1897.
  312. Welt CK, Chan JL, Bullen J, Murphy R, Smith P, DePaoli AM, Karalis A, Mantzoros CS. Recombinant human leptin in women with hypothalamic amenorrhea. N Engl J Med. 2004;351(10):987-997.
  313. Chan JL, Heist K, DePaoli AM, Veldhuis JD, Mantzoros CS. The role of falling leptin levels in the neuroendocrine and metabolic adaptation to short-term starvation in healthy men. J Clin Invest. 2003;111(9):1409-1421.
  314. Farooqi IS, O'Rahilly S. Leptin: a pivotal regulator of human energy homeostasis. Am J Clin Nutr. 2009;89(3):980S-984S.
  315. Rehman R, Jamil Z, Khalid A, Fatima SS. Cross talk between serum Kisspeptin-Leptin during assisted reproduction techniques. Pak J Med Sci. 2018;34(2):342-346.
  316. Backholer K, Smith JT, Rao A, Pereira A, Iqbal J, Ogawa S, Li Q, Clarke IJ. Kisspeptin cells in the ewe brain respond to leptin and communicate with neuropeptide Y and proopiomelanocortin cells. Endocrinology. 2010;151(5):2233-2243.
  317. Cunningham MJ, Clifton DK, Steiner RA. Leptin's actions on the reproductive axis: perspectives and mechanisms. Biol Reprod. 1999;60(2):216-222.
  318. Fernandez-Fernandez R, Martini AC, Navarro VM, Castellano JM, Dieguez C, Aguilar E, Pinilla L, Tena-Sempere M. Novel signals for the integration of energy balance and reproduction. Mol Cell Endocrinol. 2006;254-255:127-132.
  319. Quennell JH, Mulligan AC, Tups A, Liu X, Phipps SJ, Kemp CJ, Herbison AE, Grattan DR, Anderson GM. Leptin indirectly regulates gonadotropin-releasing hormone neuronal function. Endocrinology. 2009;150(6):2805-2812.
  320. Castellano JM, Navarro VM, Fernandez-Fernandez R, Nogueiras R, Tovar S, Roa J, Vazquez MJ, Vigo E, Casanueva FF, Aguilar E, Pinilla L, Dieguez C, Tena-Sempere M. Changes in hypothalamic KiSS-1 system and restoration of pubertal activation of the reproductive axis by kisspeptin in undernutrition. Endocrinology. 2005;146(9):3917-3925.
  321. Luque RM, Kineman RD, Tena-Sempere M. Regulation of hypothalamic expression of KiSS-1 and GPR54 genes by metabolic factors: analyses using mouse models and a cell line. Endocrinology. 2007;148(10):4601-4611.
  322. Roa J, Garcia-Galiano D, Varela L, Sanchez-Garrido MA, Pineda R, Castellano JM, Ruiz-Pino F, Romero M, Aguilar E, Lopez M, Gaytan F, Dieguez C, Pinilla L, Tena-Sempere M. The mammalian target of rapamycin as novel central regulator of puberty onset via modulation of hypothalamic Kiss1 system. Endocrinology. 2009;150(11):5016-5026.
  323. Wahab F, Ullah F, Chan YM, Seminara SB, Shahab M. Decrease in hypothalamic Kiss1 and Kiss1r expression: a potential mechanism for fasting-induced suppression of the HPG axis in the adult male rhesus monkey (Macaca mulatta). Horm Metab Res. 2011;43(2):81-85.
  324. Castellano JM, Navarro VM, Fernandez-Fernandez R, Roa J, Vigo E, Pineda R, Dieguez C, Aguilar E, Pinilla L, Tena-Sempere M. Expression of hypothalamic KiSS-1 system and rescue of defective gonadotropic responses by kisspeptin in streptozotocin-induced diabetic male rats. Diabetes. 2006;55(9):2602-2610.
  325. Elias CF. Leptin action in pubertal development: recent advances and unanswered questions. Trends Endocrinol Metab. 2012;23(1):9-15.
  326. Hill JW, Elmquist JK, Elias CF. Hypothalamic pathways linking energy balance and reproduction. Am J Physiol Endocrinol Metab. 2008;294(5):E827-832.
  327. Donato J, Jr., Cravo RM, Frazao R, Gautron L, Scott MM, Lachey J, Castro IA, Margatho LO, Lee S, Lee C, Richardson JA, Friedman J, Chua S, Jr., Coppari R, Zigman JM, Elmquist JK, Elias CF. Leptin's effect on puberty in mice is relayed by the ventral premammillary nucleus and does not require signaling in Kiss1 neurons. J Clin Invest. 2011;121(1):355-368.
  328. Schalla MA, Stengel A. Central mechanisms of kisspeptin-induced inhibition of food intake. Peptides. 2021;135:170475.
  329. Saito R, Tanaka K, Nishimura H, Nishimura K, Sonoda S, Ueno H, Motojima Y, Yoshimura M, Maruyama T, Yamamoto Y, Kusuhara K, Ueta Y. Centrally administered kisspeptin suppresses feeding via nesfatin-1 and oxytocin in male rats. Peptides. 2019;112:114-124.
  330. Frazao R, Dungan Lemko HM, da Silva RP, Ratra DV, Lee CE, Williams KW, Zigman JM, Elias CF. Estradiol modulates Kiss1 neuronal response to ghrelin. Am J Physiol Endocrinol Metab. 2014;306(6):E606-614.
  331. Kluge M, Schussler P, Schmidt D, Uhr M, Steiger A. Ghrelin suppresses secretion of luteinizing hormone (LH) and follicle-stimulating hormone (FSH) in women. J Clin Endocrinol Metab. 2012;97(3):E448-451.
  332. Martini AC, Fernandez-Fernandez R, Tovar S, Navarro VM, Vigo E, Vazquez MJ, Davies JS, Thompson NM, Aguilar E, Pinilla L, Wells T, Dieguez C, Tena-Sempere M. Comparative analysis of the effects of ghrelin and unacylated ghrelin on luteinizing hormone secretion in male rats. Endocrinology. 2006;147(5):2374-2382.
  333. Donoso AO, Lopez FJ, Negro-Vilar A. Cross-talk between excitatory and inhibitory amino acids in the regulation of luteinizing hormone-releasing hormone secretion. Endocrinology. 1992;131(3):1559-1561.
  334. Leranth C, MacLusky NJ, Sakamoto H, Shanabrough M, Naftolin F. Glutamic acid decarboxylase-containing axons synapse on LHRH neurons in the rat medial preoptic area. Neuroendocrinology. 1985;40(6):536-539.
  335. Mijiddorj T, Kanasaki H, Sukhbaatar U, Oride A, Kyo S. DS1, a delta subunit-containing GABA(A) receptor agonist, increases gonadotropin subunit gene expression in mouse pituitary gonadotrophs. Biol Reprod. 2015;92(2):45.
  336. Moore AM, Prescott M, Marshall CJ, Yip SH, Campbell RE. Enhancement of a robust arcuate GABAergic input to gonadotropin-releasing hormone neurons in a model of polycystic ovarian syndrome. Proc Natl Acad Sci U S A. 2015;112(2):596-601.
  337. Watanabe M, Fukuda A, Nabekura J. The role of GABA in the regulation of GnRH neurons. Front Neurosci. 2014;8:387.
  338. Di Giorgio NP, Bizzozzero-Hiriart M, Libertun C, Lux-Lantos V. Unraveling the connection between GABA and kisspeptin in the control of reproduction. Reproduction. 2019;157(6):R225-R233.
  339. Kanasaki H, Tumurbaatar T, Oride A, Hara T, Okada H, Kyo S. Gamma-aminobutyric acidA receptor agonist, muscimol, increases KiSS-1 gene expression in hypothalamic cell models. Reprod Med Biol. 2017;16(4):386-391.
  340. Kurian JR, Keen KL, Guerriero KA, Terasawa E. Tonic control of kisspeptin release in prepubertal monkeys: implications to the mechanism of puberty onset. Endocrinology. 2012;153(7):3331-3336.
  341. Terasawa E, Kurian JR, Guerriero KA, Kenealy BP, Hutz ED, Keen KL. Recent discoveries on the control of gonadotrophin-releasing hormone neurones in nonhuman primates. J Neuroendocrinol. 2010;22(7):630-638.
  342. Piet R, Kalil B, McLennan T, Porteous R, Czieselsky K, Herbison AE. Dominant Neuropeptide Cotransmission in Kisspeptin-GABA Regulation of GnRH Neuron Firing Driving Ovulation. J Neurosci. 2018;38(28):6310-6322.
  343. Comninos AN, Yang L, O'Callaghan J, Mills EG, Wall MB, Demetriou L, Wing VC, Thurston L, Owen BM, Abbara A, Rabiner EA, Dhillo WS. Kisspeptin modulates gamma-aminobutyric acid levels in the human brain. Psychoneuroendocrinology. 2021;129:105244.
  344. Hatef A, Unniappan S. Gonadotropin-releasing hormone, kisspeptin, and gonadal steroids directly modulate nucleobindin-2/nesfatin-1 in murine hypothalamic gonadotropin-releasing hormone neurons and gonadotropes. Biol Reprod. 2017;96(3):635-651.
  345. Christian CA, Moenter SM. Vasoactive intestinal polypeptide can excite gonadotropin-releasing hormone neurons in a manner dependent on estradiol and gated by time of day. Endocrinology. 2008;149(6):3130-3136.
  346. Ducret E, Anderson GM, Herbison AE. RFamide-related peptide-3, a mammalian gonadotropin-inhibitory hormone ortholog, regulates gonadotropin-releasing hormone neuron firing in the mouse. Endocrinology. 2009;150(6):2799-2804.
  347. Lee EJ, Moore CT, Hosny S, Centers A, Jennes L. Expression of estrogen receptor-alpha and c-Fos in adrenergic neurons of the female rat during the steroid-induced LH surge. Brain Res. 2000;875(1-2):56-65.
  348. Miller BH, Olson SL, Levine JE, Turek FW, Horton TH, Takahashi JS. Vasopressin regulation of the proestrous luteinizing hormone surge in wild-type and Clock mutant mice. Biol Reprod. 2006;75(5):778-784.
  349. Palm IF, Van Der Beek EM, Wiegant VM, Buijs RM, Kalsbeek A. Vasopressin induces a luteinizing hormone surge in ovariectomized, estradiol-treated rats with lesions of the suprachiasmatic nucleus. Neuroscience. 1999;93(2):659-666.
  350. Leon S, Tena-Sempere M. Dissecting the Roles of Gonadotropin-Inhibitory Hormone in Mammals: Studies Using Pharmacological Tools and Genetically Modified Mouse Models. Front Endocrinol (Lausanne). 2015;6:189.
  351. Qi Y, Oldfield BJ, Clarke IJ. Projections of RFamide-related peptide-3 neurones in the ovine hypothalamus, with special reference to regions regulating energy balance and reproduction. J Neuroendocrinol. 2009;21(8):690-697.
  352. Liu X, Herbison AE. Kisspeptin Regulation of Neuronal Activity throughout the Central Nervous System. Endocrinol Metab (Seoul). 2016;31(2):193-205.
  353. Acevedo-Rodriguez A, Kauffman AS, Cherrington BD, Borges CS, Roepke TA, Laconi M. Emerging insights into Hypothalamic-pituitary-gonadal (HPG) axis regulation and interaction with stress signaling. J Neuroendocrinol. 2018.
  354. Kriegsfeld LJ, Mei DF, Bentley GE, Ubuka T, Mason AO, Inoue K, Ukena K, Tsutsui K, Silver R. Identification and characterization of a gonadotropin-inhibitory system in the brains of mammals. Proc Natl Acad Sci U S A. 2006;103(7):2410-2415.
  355. Molnar CS, Kallo I, Liposits Z, Hrabovszky E. Estradiol down-regulates RF-amide-related peptide (RFRP) expression in the mouse hypothalamus. Endocrinology. 2011;152(4):1684-1690.
  356. Uenoyama Y, Nakamura S, Hayakawa Y, Ikegami K, Watanabe Y, Deura C, Minabe S, Tomikawa J, Goto T, Ieda N, Inoue N, Sanbo M, Tamura C, Hirabayashi M, Maeda KI, Tsukamura H. Lack of pulse and surge modes and glutamatergic stimulation of luteinising hormone release in Kiss1 knockout rats. J Neuroendocrinol. 2015;27(3):187-197.