Archives

Diabetes, Cardiomyopathy, and Heart Failure

ABSTRACT

 

Heart failure (HF) is an underappreciated complication of diabetes. HF occurs in individuals with diabetes at higher rates, even in the absence of other HF risk factors such as coronary artery disease and hypertension. Comorbid ischemic heart disease and cardiovascular risk factors significantly contribute to the etiology of cardiomyopathy and HF in patients with diabetes. In addition, long-standing diabetes can independently cause subclinical alteration in cardiac structure and function, eventually leading to the development and progression of HF. A complex interplay between numerous mechanisms underlies the pathophysiologic links between diabetes and HF. Patients with concurrent diabetes and HF have impaired quality of life and a poor prognosis with a high risk of hospitalization and mortality. Despite the solid epidemiologic link between poor glycemic control and HF risk, the effects of intensified glycemic control in preventing HF remain controversial. Large-scale cardiovascular outcome trials published since 2015 have confirmed the efficacy and safety of sodium-glucose co-transporter-2 inhibitors (SGLT2) inhibitors in preventing HF among patients with type 2 diabetes mellitus. In addition, several dedicated major clinical trials confirmed the cardiovascular benefits of SGLT2 inhibitors in patients with established HF, regardless of left ventricular ejection fraction or diabetes status. Furthermore, high-quality data from these clinical trials transformed SGLT2 inhibitors from glucose-lowering agents to HF drugs. This chapter outlines the complex relationship between HF and diabetes, focusing on the epidemiology, pathophysiology, and prognostic implications. Additionally, we review the current knowledge on identifying subclinical cardiac remodeling, predicting HF risk, and preventing HF in diabetes. We also summarize the recent evidence and guideline recommendations for the pharmacological treatment of patients with coexisting HF and diabetes. 

 

INTRODUCTION

 

Diabetes is a risk factor for cardiomyopathy and heart failure (HF) independent of traditional cardiovascular (CV) disease (CVD) risk factors such as hypertension and coronary artery disease (CAD) (1–4). The universal definition of HF recognizes patients with diabetes as “at risk for HF” (Stage A).  Asymptomatic individuals with at least one of the following: 1) evidence of structural heart disease, 2) abnormal cardiac function, or 3) elevated cardiac natriuretic peptide or troponins are considered to have “pre-HF” (stage B). According to this classification, HF (stage C) is defined as a clinical syndrome with signs or symptoms of HF caused by an abnormality in cardiac structure and function and corroborated by elevated natriuretic peptide or objective evidence of cardiogenic congestion (pulmonary or systemic) (3,5).

 

The prevalence of diabetes is approximately 10.2% in the U.S. population, and HF affects 9 to 22% of patients with diabetes (6–10). In clinical trials of antidiabetic agents, HF was present in 4 to 30% of participants with diabetes (11). On the other hand, the prevalence of pre-diabetes or diabetes was 30 to 40% among individuals enrolled in HF trials (12,13).

 

Longstanding diabetes alters cardiac structure and function, resulting from the direct effects of abnormal myocardial metabolism and insulin resistance (IR) even without atherosclerotic CAD (14). The pathophysiologic link between diabetes and HF is complex and multifactorial, involving various abnormal biochemical pathways including but not limited to abnormal calcium signaling, deranged glucose/fatty acid metabolism, and inflammatory pathways contributing to myocardial fibrosis, stiffness, and hypertrophy (7,15,16). A complex interaction of these mechanisms can cause asymptomatic diastolic and systolic dysfunction, eventually leading to the clinical syndrome of HF. Conversely, HF is also associated with a higher prevalence of diabetes and is considered a predictor of future risk of type 2 diabetes mellitus (T2DM) (17).

 

Left ventricular (LV) dysfunction in patients with diabetes may present with three different HF phenotypes, such as HF with preserved LV ejection fraction (LVEF ≥ 50%; HFpEF), HF with mildly-reduced LVEF (HFmrEF; LVEF 40-49%), and HF with reduced LVEF (HFrEF; LVEF ≤40%) (3). Diagnosing HFpEF and HFmrEF is often challenging since the symptomatology of HF may overlap with other comorbidities such as obesity, lung disease, and chronic kidney disease (CKD). Therefore, the guidelines usually recommend incorporating additional objective diagnostic criteria such as elevated natriuretic peptides or imaging evidence of either structural heart disease or diastolic dysfunction (18).

 

The coexistence of diabetes and HF is a poor prognostic factor, posing a greater risk of HF hospitalization, all-cause mortality, and CVD mortality. For instance, epidemiologic studies indicated a 50-90% higher risk of CVD mortality in patients with HF and diabetes, regardless of HF phenotype (12,19). HF patients without DM are at increased risk of developing glycemic abnormalities. In addition, newly diagnosed pre-diabetes was associated with a significantly higher risk of all-cause and CV mortality in HF patients. These findings underscore the importance of screening for pre-diabetes or diabetes among patients with HF (17,20,21). Moreover, early assessment with echocardiography can be helpful for the detection of subclinical structural abnormalities and myocardial dysfunction in asymptomatic patients with diabetes.

 

The pathophysiology of diabetes-related HF is complex, and despite significant advances over the past decades, many areas are still poorly understood. Since 2015, several landmark clinical trials on sodium-glucose co-transporter-2 (SGLT2) inhibitors and glucagon-like peptide-1 receptor agonists (GLP1-RA) have revolutionized our understanding of CVD risk reduction in patients with T2DM and have led to a paradigm shift in the clinical practice recommendations for the management of T2DM (22). Incredible evidence from the CVD outcome trials (CVOTs) has confirmed the significant improvement in HF outcomes with SGLT2 inhibitors in patients with or without diabetes. These findings increased medical communities' awareness and interest in the links between diabetes and HF.

 

The American Diabetes Association (ADA) now recommends using SGLT2 inhibitors as first-line agents in T2DM patients with a high risk of or established HF (23). In addition, several dedicated major randomized clinical trials (RCTs) confirmed the CVD benefits of SGLT2 inhibitors in patients with established HF, regardless of LVEF or diabetes status. Moreover, these RCTs transformed SGLT2 inhibitors from glucose-lowering agents to HF drugs. The 2022 American College of Cardiology (ACC) Foundation / American Heart Association (AHA) / Heart Failure Society of America (HFSA) guideline for the management of HF recommended the use of SGLT2 inhibitors for the treatment of HF, regardless of LVEF (3).

 

This chapter outlines the complex relationship between HF and diabetes, focusing on the epidemiology, pathophysiology, and prognostic implications. Additionally, we review the current knowledge on identifying subclinical cardiac remodeling, predicting HF risk, and preventing HF in diabetes. We also summarize the recent evidence and guideline recommendations for the pharmacological treatment of patients with coexisting HF and diabetes. 

 

EPIDEMIOLOGY OF DIABETES AND HF

 

There is a bidirectional link between diabetes and HF (24). Diabetes, either type 1 or type 2, is a strong risk factor for developing HF (25–27). In addition, HF may contribute to the pathogenesis of IR and T2DM (28). The shared underlying risk factors and the overlap of the pathophysiological mechanisms play a critical role in the frequent coexistence of T2DM and HF.  

 

Based on the data from the National Health and Nutrition Examination Survey (NHANES) from 2015 to 2018, the prevalence of HF is 2.3% in the US general adult population (29). The prevalence of HF in individuals with diabetes ranges between 9% and 22%, depending on the characteristics of the population studied (6,8,9). Diabetes is also highly prevalent among patients with HF. In major contemporary drug trials of HF, 32% to 43% of patients with chronic HF had coexisting diabetes (12,30,31). A report from a nationwide US registry (NHANES 2005-2016) demonstrated that, among patients with HF, the prevalence rates of diagnosed and undiagnosed T2DM were 34.7% and 12.8%, respectively (32).

 

HF is a common but often neglected complication of diabetes (33). HF and cardiomyopathy have a heterogeneous etiology in patients with diabetes (Figure 1 and Figure 2). The strong link between diabetes and CAD, hypertension, and renal disease plays a significant role in the development of cardiomyopathy and HF in patients with diabetes (34). Moreover, HF occurs in individuals with diabetes at higher rates, even in the absence of other HF risk factors (16,35).

 

Diabetic Cardiomyopathy

 

Diabetic cardiomyopathy, which lacks a standardized clinical definition, generally refers to diabetes-related myocardial dysfunction without other potential causes (36). A report by Lundbæk in 1954 described the concept of diabetes directly causing myocardial dysfunction (37). In 1972, a landmark study by Rubler et al. described diabetic cardiomyopathy as a new clinical entity by reporting the post-mortem results of 4 patients with diabetes-related HF and dilated cardiomyopathy without other apparent causes of myocardial dysfunction (14). The initial reports of diabetic cardiomyopathy referred to a dilated LV with eccentric hypertrophy and LV systolic dysfunction (HFrEF). Nevertheless, more recent clinical studies described HFpEF with concentric LV hypertrophy and LV diastolic dysfunction as a distinct phenotype of cardiomyopathy rather than being an intermediate form between risk factors and HFrEF (38). The transition from HFpEF to HFrEF does not appear to occur as commonly as it was once presumed.  

 

Epidemiologic Evidence

 

Evidence from large-scale epidemiologic studies has confirmed the strong link between diabetes and HF. For instance, reports from the prospective Framingham Heart Study in the 1970s indicated that individuals with diabetes had 2-fold (in men) to 5-fold (in women) higher risk of developing HF than individuals without diabetes after adjustment for other risk factors (2,39). Similarly, contemporary cohort studies suggested that diabetes is independently associated with a 1.7 to 2.5-fold greater risk of HF (6,40).

 

A recent nationwide cohort study from Sweden including >679.000 patients with T2DM and >2 million matched control subjects demonstrated that a diagnosis of T2DM was associated with HF risk even if CVD risk factors, such as glycated hemoglobin, systolic blood pressure (BP), estimated glomerular filtration rate, and lipids were within a target range (41). The study also demonstrated that CVD complications have significantly declined over the past 20 years in individuals with and without T2DM. However, the decline in the rate of HF in patients with T2DM has plateaued over recent years. One potential explanation for this finding is the obesity epidemic, as adiposity plays a major role in the development of HF in patients with diabetes. For instance, a recent analysis of 2 US cohort studies demonstrated that overall obesity, abdominal obesity, and fat mass were strongly associated with a greater risk of HF in participants with diabetes. Interestingly, a similar independent association was absent in those without T2DM (42).  

 

Ischemic heart disease is more frequently seen in HF patients with coexistent T2DM than those without T2DM (63% vs. 47%). Moreover, ~90% of the patients with T2DM and HF of non-ischemic etiology have at least one comorbidity that can contribute to HF development, such as hypertension, atrial fibrillation, valvular disease, or pulmonary disease (43).

 

Figure 1. Heart failure with reduced ejection fraction due to ischemic cardiomyopathy in a patient with uncontrolled type 2 diabetes. 57-year-old female patient with a history of uncontrolled type 2 diabetes (HbA1c = 12.0) and active tobacco abuse presented with a 2-day history of intermittent midsternal chest pain. (A) Her ECG on presentation demonstrated findings of acute/recent anteroseptal myocardial infarct and old/age indeterminate inferior myocardial infarct, and her serum troponin I was markedly elevated. (B and C) Her echocardiography revealed a dilated left ventricle with severely reduced systolic function, an ejection fraction of 20-25%, and akinetic anterior and inferior wall segments. Her coronary angiography, which was performed emergently, demonstrated subacute occlusion of the proximal segment of the anterior descending artery (arrow in image D) and chronic total occlusion of the middle segment of the right coronary artery (arrow in image E).

Figure 2. Heart failure with reduced ejection fraction due to non-ischemic cardiomyopathy in a patient with uncontrolled type 2 diabetes. 59-year-old male patient with a history of hypertension (medically managed), class I obesity, hyperlipidemia, moderate alcohol consumption, and undiagnosed type 2 diabetes (HbA1c = 13.5) presented with dyspnea on exertion, orthopnea, and leg edema and he was admitted with the diagnoses of acute decompensated heart failure. (A) His ECG on presentation demonstrated left ventricular hypertrophy with repolarization abnormalities. (B and C) His echocardiography revealed a dilated left ventricle with severely reduced systolic function and diffuse hypokinesis, an ejection fraction of 25-30%, and eccentric left ventricular hypertrophy. (D and E) His coronary angiography, performed for ischemic evaluation, demonstrated no evidence of significant epicardial coronary artery disease. The patient’s non-ischemic cardiomyopathy was attributed to a mixed presentation of alcoholic and diabetic cardiomyopathy and hypertensive heart disease.

 

Diabetes is an independent predictor of progression from preclinical HF (stage A and stage B) to clinic HF (stage C) (44). A population-based analysis on the National Scottish Register found that HF hospitalization risk was ~2-fold higher among patients with diabetes than those without diabetes (45). A prospective cohort study, including individuals from the southeastern U.S., demonstrated that hypertension and diabetes were associated with the highest HF risk in white and black participants (46). The population-attributable risk of HF was highest for hypertension (31.8%), followed by diabetes (17%). A population-based case-control study also observed an attributable risk of HF at ~17% for diabetes, without any significant difference between HFpEF and HFrEF (47).

 

Epidemiologic studies have demonstrated a higher incidence of HF in men than in women with diabetes (27,40). This finding is consistent with the strong association between HF risk and male sex in the general population. However, interestingly, diabetes contributes to the future risk of HF more in women than in men, as supported by multiple epidemiologic studies (27,39). In a meta-analysis of 47 cohort studies including >12 million individuals, type 1 diabetes mellitus (T1DM) and T2DM were associated with a 47% and 9% greater excess risk of HF in women than men, respectively (48). The basis for the sex-specific disparity in HF risk attributable to diabetes remains unclear. 

 

Prediabetes and HF Risk

 

Some epidemiologic studies have suggested that prediabetes may pose a risk for cardiomyopathy and HF(49). In a population-based cohort study, prediabetes was independently associated with HF with an odds ratio of 1.7 (9). A modest but significant association exists between fasting plasma glucose levels and the risk of HF independent of an individual’s diabetes status (50).

 

Glycemic Control and HF Risk

 

Glycemic exposure predicts HF risk in individuals with T1DM and T2DM (26,27,51). A population-based prospective case-control study from the Swedish National Diabetes Register evaluated the impact of glycemic control on the future risk of HF hospitalization over a mean follow-up of 7.9 years (26). Compared to a population-based control group without diabetes, patients with T1DM had a four times higher risk of HF hospitalization. Nevertheless, the risk markedly varied depending on glycemic control or comorbidities; hazard-ratio (HR) of 2.2 (1.5–3.0; p<0·0001) in patients with hemoglobin A1c (HbA1c) ≤6.9%, HR of 11.2 (8·4–14·9; p<0·0001) in patients with HbA1c ≥9.7%. Another report from the same dataset revealed that each 1% increase in HbA1c correlated with a 20% higher risk of HF in patients with myocardial infarction and T1DM (52). Among individuals with T2DM, the excess risk of HF attributable to glycemic control varies depending on the patient’s age. For instance, poor glycemic control correlates more strongly with excess risk of HF among middle-aged adults (<55 years old). In contrast, the correlation between HbA1c and the risk of HF markedly attenuates with advanced age (27).

  

Age at Diagnosis of Diabetes and HF Risk

 

Diagnosis of diabetes at a younger age correlates with a higher risk of HF (45,53,54). A report by Rawshani et al. using the data from the Swedish National Diabetes Register demonstrated that compared to a control group without diabetes, individuals with onset of T1DM before age ten years had 12 times and those with onset during young adulthood (20 to 29 years) had five times increased risk of HF. Sattar et al., using the same registry data, evaluated the association between age at diagnosis and future HF risk among participants with T2DM (54). Their analysis revealed that adults diagnosed with T2DM before 41 years of age had a five times higher risk of HF than their counterparts without diabetes. Interestingly, T2DM diagnosis after the age of 80 years did not increase the risk of HF and was associated with a lower risk of all-cause and CV mortality. Consistently, an analysis from a US cohort demonstrated that every 5-year increase in the duration of DM was independently associated with a 17% rise in the risk of incident HF (55). As expected, the association between diabetes duration and HF risk was more prominent in patients with elevated HbA1c.

 

The explanations behind the association of duration and age at diabetes diagnosis and future HF risk are likely multifaceted, with a variation between T1DM and T2DM. The total glycemic load, defined as the cumulative exposure to the effects of hyperglycemia, is a predictor of complications of diabetes. The main components of the total glycemic load are the glycemic variability and the duration of diabetes determined by the age of diabetes onset, more prominently in T1DM (53). Individuals who develop T2DM at a younger age are more likely to have other comorbidities such as obesity, dyslipidemia, hypertension, nephropathy, smoking, and lower socioeconomic status when compared to their counterparts without diabetes. Furthermore, this comorbidity burden likely contributes to the relative excess risk of HF observed in patients diagnosed with T2DM at a relatively young age (54). These findings highlight the significance of delaying diabetes onset as one focus of HF prevention efforts (55).

  

The Relationship Between Diabetes and Comorbidities

 

Traditional modifiable CVD risk factors, such as hypertension, obesity, dyslipidemia, and cigarette smoking, are prevalent among individuals with diabetes. Hypertension affects 66% to 76% of adults with diabetes in the US (56). According to the 2020 National Diabetes Statistics Report, 45.8% of adults with diabetes are obese (body-mass-index [BMI] of 30 to 39.9 kg/m2), and 15.5% are morbidly obese (BMI of ≥40 kg/m2) (10).

 

Coexisting CVD risk factors significantly contribute to the risk of HF in patients with diabetes. A large prospective cohort study, including >270,000 participants with T2DM in the Swedish National Diabetes Register, examined the relationship between five risk factors (elevated HbA1C, dyslipidemia, albuminuria, smoking, and high BP) and CVD outcomes after a median follow-up of 5.7 years (57), The analyses revealed that participants with T2DM who had no risk-factor variables outside the target ranges had a 45% higher risk of hospitalization for HF when compared to that of a control group without diabetes. However, the excess risk of hospitalization for HF was substantially higher (HR vs. control, 11.35; 95% CI, 7.16 to 18.01) when patients with T2DM had all five risk-factor variables outside the target range. These findings indicated the importance of controlling coexisting CV risk factors for preventing HF in diabetes.

 

Recent reports have indicated that comorbid mental disorders may increase HF risk in individuals with diabetes. A retrospective analysis of nationwide health claims data of Korean participants demonstrated an independent association between HF risk and the number of mental disorders in patients with diabetes (58).

 

HF as a Risk Factor for Diabetes

 

Patients with HF are at risk of developing incident DM over time. Data from clinical trials showed that the incidence of new-onset diabetes among patients with HF is 7 to 11% over a 3- to 5-year follow-up period (59,60). Even though the published data is sparse, some evidence has emerged over the past two decades supporting the possible independent role of HF as a risk factor for incident T2DM (61). Analyses of prospective cohort studies and clinical trial participants demonstrated that HF at baseline might predispose the future risk of new-onset diabetes (61–64). Significant predictors of incident diabetes among individuals with HF are elevated glucose and HbA1c, higher BMI and waist circumference, longer duration of HF, and higher functional class of HF (28,61–63).

 

IMPACT OF DIABETES ON CARDIAC STRUCTURE AND FUNCTION

 

The frequent coexistence of diabetes with other comorbidities, such as hypertension and obesity, makes it difficult to understand the relative contribution of each disease entity in cardiac remodeling and dysfunction in clinical practice (65). However, growing evidence has supported an independent association between diabetes and various alterations in cardiac structure and function. These asymptomatic subclinical alterations at earlier stages can be detrimental and increase the risk of developing HF and CVD morbidity and mortality in general (44). Recognizing these subclinical alterations is critical for the early identification of high-risk patients and preventing overt HF and diabetic cardiomyopathy.

 

Left Ventricular Hypertrophy

 

LV hypertrophy (LVH) is characterized by increased LV mass due to myocardial remodeling. LVH is usually caused by a complex interaction between several factors, including hypertension, diabetes, metabolic syndrome, obesity, gender, ethnicity, and genetic and neurohumoral factors (66). There are three distinct LV geometric abnormality patterns: concentric remodeling (normal LV mass with increased relative wall thickness), concentric LVH (increased LV mass and increased relative wall thickness), and eccentric LVH (increased LV mass and normal relative wall thickness) (Figure 3).

 

Figure 3. Based on linear measurements, relative wall thickness and left ventricular mass index determine left ventricular geometric patterns. LVH, left ventricular hypertrophy.

 

LVH has long been recognized as a target organ response and a strong independent risk factor for HF, CAD, stroke, and CVD mortality (66,67). LVH leads to LV diastolic dysfunction by reduced LV compliance, impaired diastolic filling, prolonged isovolumetric relaxation, and increased LV and left atrial filling pressures (16). The universal definition of HF recognizes asymptomatic LVH, LV systolic dysfunction, and LV diastolic dysfunction as “pre-HF” to emphasize the progressive nature of HF and the importance of HF prevention (5).

 

LVH is common among adults with diabetes, with an estimated prevalence as high as 70% (68). A pooled analysis of 3 epidemiological cohort studies, including 2900 individuals with T2DM and no known CVD, demonstrated that 67% of the participants had at least one of the following echocardiographic abnormalities: LVH, left atrial enlargement, or diastolic dysfunction (44). Coexistent hypertension appears to be the main contributor to LVH in patients with diabetes (69). However, several studies have also demonstrated an independent association between diabetes and LVH. In the Framingham Heart Study, serum glucose, insulin levels, and IR were significantly linked to concentric LV remodeling, a finding that was more striking in women than in men (70,71). Results from a prospective cohort study with a 25-year follow-up period indicated that long-standing glycemic abnormalities have a cumulative effect on LV remodeling, and patients with early-onset diabetes tend to have a worse degree of LVH (72).

 

Diabetic cardiomyopathy may present with distinct LVH features (34). Thickened and stiff LV walls with normal LV volume usually characterize diabetic cardiomyopathy with HFpEF phenotype. Furthermore, at the cellular level, cardiomyocytes appear hypertrophied with normal sarcomere structure and increased collagen deposition in the interstitial space. Contrarily, diabetic cardiomyopathy with HFrEF phenotype usually manifests with eccentric LVH with dilated LV volume. At the cellular level, cardiomyocytes appear to have damage with loss of sarcomeres and replacement of some cardiomyocytes with interstitial fibrosis (38).

  

LV Diastolic Dysfunction

 

Diastolic dysfunction is common among otherwise asymptomatic individuals with diabetes, and its prevalence varies between 20% and 60% depending on the diagnostic criteria used and the population studied (73–75). Prospective cohort studies have confirmed that diabetes and poor glycemic control can independently contribute to the development of diastolic dysfunction (72). Even though diastolic dysfunction is often linked to LVH, it can occur in patients with diabetes, even in the absence of LVH (34).

 

Mild diastolic dysfunction (delayed myocardial relaxation) has a weak prognostic significance. However, the progression of diastolic dysfunction and increased LV filling pressure findings on echocardiography predispose to the future risk of HF and mortality in patients with diabetes (73,75). Moreover, among asymptomatic individuals with baseline diastolic dysfunction, diabetes is an independent predictor of progression to HFpEF or HFrEF (76).

 

LV Systolic Dysfunction

 

Traditionally, impaired LVEF is the primary marker of cardiomyopathy and systolic dysfunction. LVEF is a simple measure commonly used in the CV risk evaluation and management of CVD. However, LVEF does not capture the full spectrum of myocardial function (77).  Global longitudinal strain (GLS) evaluated by speckle-tracking echocardiography is a robust technique that measures tissue deformation in a longitudinal direction (Figure 4). Reduced GLS is a marker of reduced contractility (75). GLS is more sensitive than conventional LVEF as a measure of systolic function and has an additional prognostic value (77,78). Therefore, it is now commonly used to detect subclinical LV systolic dysfunction.

 

Figure 4. Global longitudinal strain by speckle tracking echocardiography. Assessment of global longitudinal strain (GLS) in a healthy, asymptomatic individual with a GLS of -25%. The top row displays a regional strain map superimposed on the grayscale two-dimensional echocardiographic images in apical four-chamber (A4C), apical two-chamber (A2C), and apical three-chamber (A3C) views. The bottom left bullseye displays regional longitudinal strain for each segment of a 16-segment left ventricle model. Bright red denotes the most negative normal values of GLS. The bottom right bullseye shows the time (ms) between aortic valve opening and peak longitudinal strain, a measure of desynchrony, for each segment.

 

Impaired GLS is highly prevalent in asymptomatic, normotensive patients with diabetes and normal LVEF (67,79,80). Diabetes is associated with reduced GLS, even in adolescents and young adults with T1DM or T2DM (81,82). Moreover, an inverse correlation exists between HbA1C and GLS regardless of diabetes status, race, and sex (83). Not surprisingly, impaired GLS is a robust independent predictor of new-onset HF and mortality in patients with diabetes (67).

 

Diabetes can lead to clinical HF syndrome in individuals with asymptomatic LV systolic dysfunction. An RCT that included adults with asymptomatic LV systolic dysfunction demonstrated that diabetes increased the risk of HF development by 53% and doubled the risk of HF hospitalization over a median follow-up of 3 years (84).

 

Better glycemic control in patients with diabetes can lead to improvements in LV systolic and diastolic function indices. In a prospective cohort study of subjects with uncontrolled T2DM, lowering of average HbA1c from 10.3% to 8.3% over 12 months was associated with a 21% increase in GLS and a 24% increase in septal e’ velocity, a marker of myocardial relaxation (85).

 

 

A complex interplay between numerous mechanisms underlies the pathophysiologic links between diabetes and HF. These pathophysiologic mechanisms include but are not limited to impaired cardiac insulin signaling, glucotoxicity, lipotoxicity, mitochondrial dysfunction, myocardial fibrosis, oxidative stress, impaired myocardial calcium handling, CV autonomic dysfunction, endocardial dysfunction, overactivation of the renin-angiotensin-aldosterone system (7)(Figure 5). The relative contribution of each pathophysiologic mechanism and their relationship with the phenotype of diabetic cardiomyopathy are still poorly understood. The pathophysiology of cardiomyopathy and HF vary depending on the type of diabetes (T1DM vs. T2DM) and type of HF (HFrEF vs. HFpEF) (86).

 

Figure 5. Pathophysiological mechanisms, subclinical abnormalities, and clinical manifestations of diabetic cardiomyopathy. AGEs, advanced glycation end-products; CMP, cardiomyopathy; EF, ejection fraction; HF, heart failure; LV, left ventricular; RAAS, renin-angiotensin-aldosterone system. *In patients with type-1 diabetes mellitus.

 

Alterations in Myocardial Energy Substrate

 

Under normal circumstances, the heart predominantly consumes free fatty acids (FFA; ~70%) and glucose (~30%) and can adapt its choice of fuels depending on their availability (15). In T2DM, cardiomyocyte substrate utilization shifts towards FFA, and glucose utilization decreases in response to IR. As a result, the heart becomes metabolically less flexible and almost completely reliant on FFA. These alterations lead to inefficient energy metabolism since FFA oxidation requires more oxygen for energy production than glucose or ketone bodies (15,87). Moreover, the increased FFA uptake causes the accumulation of triglycerides in the cardiomyocytes and promotes lipotoxicity, mitochondrial dysfunction, oxidative stress, and apoptosis (34). A prospective study elegantly evaluated the impact of diabetes on lipid accumulation by analyzing endomyocardial biopsy samples from 158 adult heart transplant recipients (88). The investigators demonstrated that cardiomyocyte samples of transplanted healthy hearts begin to show evidence of lipid accumulation (triacylglycerol and ceramide) as early as three months after a transplant in diabetic recipients. In comparison, no lipid accumulation was present in cardiomyocyte samples of transplant recipients without diabetes. Not surprisingly, cardiomyocyte lipid accumulation was an independent predictor of early systolic and diastolic dysfunction in recipients with diabetes after 12 months of a transplant.

 

Hyperinsulinemia and Insulin Resistance

 

In broad terms, IR is defined as the inability of insulin to carry on its metabolic actions at the cellular level (34). IR is the central defect in the pathogenesis of metabolic syndrome and T2DM. Moreover, HF is a well-known insulin-resistant state, and HF risk and prognosis are markedly affected by IR (34,89).

 

IR increases lipolysis, hepatic lipogenesis, and hepatic gluconeogenesis. These changes lead to substrate overload and myocardial dysfunction through lipotoxicity and glucotoxicity (75). IR and related hyperinsulinemia can affect the signaling pathways involved in cardiomyocyte hypertrophy (38).

 

Oxidative Stress

 

Oxidative stress is the imbalance between the increased generation of reactive oxygen species and reduced antioxidant defense (75). Exposure to hyperglycemia induces oxidative stress by activating NADPH oxidase, promoting mitochondrial production of superoxides, and increasing the formation of advanced glycation end products (AGEs) due to nonenzymatic glycation and oxidation of proteins and lipids (34,87).

 

Oxidative stress contributes to increased cardiac remodeling, reduced cardiac contractility and relaxation, and impaired cardiomyocyte calcium handling (75). Moreover, oxidative stress contributes to myocardial dysfunction by causing protein and DNA damage, increasing myocardial inflammation, and impairing intracellular signaling pathways (34). 

 

Endoplasmic Reticulum Stress and Impaired Calcium Handling

 

Myocardial intracellular calcium levels regulate myocardial contractility during a cardiac cycle. Alterations in the complex mechanism of calcium handling can impact myocardial contraction and relaxation (90). The endoplasmic reticulum has a major role in Ca2+ handling, lipid synthesis, and protein folding and modification (91). Moreover, cytosolic Ca2+ levels regulate cellular metabolism and cell signaling. Hyperglycemia and IR trigger endoplasmic reticulum stress, leading to unfolded proteins accumulating and impairing Ca2+ handling. In diabetic cardiomyopathy, impaired Ca2+ reuptake by the endoplasmic reticulum prolongs diastolic relaxation time (91). Studies on animal models have indicated that impaired cardiomyocyte Ca2+ handling plays a crucial role in the pathophysiology of diabetic cardiomyopathy (7,90).

 

Endothelial and Microvascular Dysfunction

 

Endothelial dysfunction, which disturbs endothelial-cardiomyocyte communication and vascular function, is common in patients with diabetes and CVD (15). Diabetes induces the deposition of AGEs in the endothelial and smooth muscle cells of the myocardial microvasculature (92). Furthermore, the deposition of AGEs triggers vascular inflammation, which reduces endothelial nitric oxide production. Low myocardial nitric oxide bioavailability levels predispose to concentric LV remodeling and diastolic dysfunction (38). Diabetes has also been linked to capillary rarefaction and pericyte loss. Microcirculatory rarefaction can impair myocardial perfusion, reduce coronary flow reserve, lead to tissue hypoxia, increase myocardial stiffness, and decrease contractility (38,93).

   

Inflammation

 

Systemic inflammation is a central component of the association between obesity, diabetes, CAD, and HF (34). In individuals with obesity, the expanding adipose tissue recruits immune cells and causes overproduction of proinflammatory cytokines, leading to obesity-mediated chronic inflammation (94).  Chronic low-grade inflammation predisposes to IR and T2DM and contributes to diabetes-related complications (94). Similarly, systemic inflammation is highly prevalent in patients with HF, contributing to the development, progression, and poor prognosis of HF regardless of LVEF (95). Animal model studies have shown a complex interaction between various inflammatory pathways implicated in cardiac inflammation and the development of diabetic cardiomyopathy (96,97). One potential link between HF and diabetes is S100A12, an inflammatory protein that increases with hyperglycemic stress. A prospective cohort study including 1345 patients with T2DM demonstrated an independent association between increased A100A12 and risk of HF hospitalization (98).

 

Epicardial Adipose Tissue Expansion

 

Diabetes and obesity can independently contribute to the expansion and transformation of epicardial adipose tissue (86,99). Epicardial adipose tissue expansion has been associated with LV systolic and diastolic dysfunction in patients with T2DM (100). Epicardial fat volume correlates with myocardial fibrosis (101), vascular stiffness (102), and reduced coronary microcirculation (103). Epicardial adipose tissue expansion and transformation contribute to the alterations in the cardiac structure and function through several pathophysiological mechanisms such as proinflammatory effects of adipokines (i.e., leptin, tumor necrosis factor-a, interleukin-1b and interleukin-6) secreted from epicardial fat and oxidative stress induced by reactive oxygen species released from adipocytes (86,104).

  

Autoimmunity

 

Autoimmunity is implicated in the pathogenesis of CVD among patients with T1DM (38,105). A recent report from a prospective cohort study showed that participants with T1DM and uncontrolled glycemia (HbA1c  9.0%) have a high prevalence of cardiac autoantibodies with an antibody profile similar to that seen in patients with chronic Chagas cardiomyopathy (106). Moreover, cardiac autoantibody positivity predicted elevated high-sensitivity C-reactive protein and the future risk of CVD events. Interestingly, cardiac autoimmunity was lower in participants with controlled T1DM (HbA1c <7.0%) compared to those with uncontrolled (HbA1c 9.0%). The specific role of cardiac autoimmunity in the development and progression of diabetic cardiomyopathy remains to be further explored.

 

Overactivation of the Renin-Angiotensin-Aldosterone System

 

Overactivation of the renin-angiotensin-aldosterone system (RAAS) constitutes a robust pathophysiologic link between diabetes, obesity, hypertension, and HF (34). The typical features of RAAS overactivation are elevated serum angiotensin and aldosterone levels and the upregulation of angiotensin and mineralocorticoid receptors (91,107). There is a bidirectional relationship between RAAS activation and dysglycemia. Hyperglycemia and IR can activate the RAAS, and in return, RAAS induces systemic and cardiac IR and contributes to oxidative stress in cardiomyocytes by increasing the activity of NADPH oxidase (7).

 

Autonomic Dysfunction

 

Autonomic nervous system dysfunction is highly prevalent in patients with diabetes. CV autonomic neuropathy (CAN) differentially impacts the cardiac innervation's sympathetic and parasympathetic components, leading to sympathetic overactivation in the earlier stages (16). This imbalance is believed to contribute to the pathogenesis of CVD. CAN induces LV remodeling, LV systolic and diastolic dysfunction, and myocardial ischemia (108–110). CAN is usually asymptomatic at earlier stages, and at advanced stages, it may present with resting tachycardia, orthostatic hypotension, abnormal BP regulation, blunted heart rate response to exercise, and impaired heart rate variability (34,111).

   

Myocardial Fibrosis

 

Myocardial fibrosis, detected by histopathology or cardiac MRI, is a hallmark of diabetes-induced cardiac remodeling and related myocardial dysfunction. In patients with diabetes, the degree of myocardial fibrosis directly correlates with HbA1c levels (112). Diabetes-induced myocardial fibrosis is characterized by the remodeling of extracellular matrix with the deposition and crosslinking of stiff collagen, progressive elimination of muscular fibrils, perivascular fibrosis, basement membrane thickening, coronary microvascular sclerosis, and formation of microaneurysms (16). Myocardial fibrosis is often considered an end product of various pathophysiologic abnormalities such as hyperglycemia, hyperinsulinemia, oxidative stress, CAN, inflammation, and the overactivation of RAAS.

 

Apoptosis

 

Apoptosis, the process of programmed cell death, has been implicated in the development of diabetic cardiomyopathy and HF (113). Studies from animal models and human subjects have demonstrated a strong association between hyperglycemia and cardiomyocyte apoptosis (114,115). The primary drivers of cardiomyocyte apoptosis in diabetes are oxidative stress, endoplasmic reticulum stress, and dysregulation of autophagy, the lysosomal process that degrades and recycles cellular proteins and organelles (34,116).

  

PROGNOSTIC IMPLICATIONS OF DIABETES IN HF

 

Data from epidemiologic studies and clinical trials have consistently demonstrated that individuals with concurrent diabetes and HF have impaired quality of life, are at high risk of hospitalization and mortality, and have an overall poor prognosis (33,117). In addition, coexistent prediabetes also appears to increase the risk of morbidity and mortality in patients with HF as well (118).  

 

A large meta-analysis including >380,000 subjects with acute and chronic HF from 43 registries and clinical trials revealed that diabetes was associated with a 28% increased risk of all-cause mortality and ~35% increased risk of both CV death and hospitalization (mostly from HF) over a three year follow up (119). Interestingly, the adverse impact of diabetes on the risk of hospitalization and mortality did not differ according to the LVEF group (≤35% vs. >35%) but was higher in patients with chronic HF than those with acute HF. Observational studies have suggested a U-shaped relationship between HbA1c and mortality in patients with coexisting HF and diabetes. Aguliar et al. reported that HbA1c of 7.1% to ≤7.8% was associated with the lowest risk of mortality when compared with the other quantiles of HbA1c in a cohort of ambulatory patients with HF who were receiving medical therapy for diabetes in the early 2000s (120).

 

Several potential mechanisms have been proposed to explain the prognostic impacts of diabetes in HF. Diabetes is linked to multimorbidity, which significantly alters HF outcomes. Moreover, diabetes induces myocardial fibrosis, inflammation, and endothelial dysfunction, leading to higher LV pressures, poor functional status, and impaired exercise capacity in patients with HF (121–125). Diabetes predisposes neurohumoral overactivation and alterations of renal sodium handling, which may lead to congestion, cardiorenal syndrome, and impaired diuretic responsiveness (121). Also, hyperglycemia in patients with diabetes upregulates SGLT2, which leads to increased renal sodium absorption and volume expansion (121). Moreover, the increased burden of ischemic heart disease and other diabetes-related comorbidities, such as CKD, contribute to the detrimental effects of HF (126).

 

Data from the Swedish Heart Failure Registry demonstrated that the diabetes-associated mortality risk is more pronounced in individuals with HF of ischemic etiology than those with nonischemic etiology (43). The 2-year survival rate was less than 50% among those with HF, T2DM, and ischemic heart disease.

 

The presence of preexisting microvascular disease is an independent risk factor for the risk of future HF events in patients with T2DM (44). In addition, microvascular disease portends an increased risk of mortality and morbidity in patients with HF. A post hoc analysis of a large RCT

demonstrated a significant association between a history of microvascular complications and future risk of adverse events among study subjects with diabetes and HFpEF (127).

 

PREVENTION OF HF IN PATIENTS WITH DIABETES

 

The critical importance of HF prevention is underscored by HF staging, where risk factors such as hypertension and diabetes are classified as stage A (at risk for HF) (3). Because of the detrimental prognostic impact of clinical HF, the prevention of asymptomatic cardiac remodeling and dysfunction (pre-HF, stage B) and symptomatic HF (stage C) are among the primary goals of the management of patients with diabetes (128).

 

Prevention of HF by Preventing Diabetes

 

Preventing the onset of diabetes during young adulthood or middle age is an effective strategy for reducing HF risk later in life. An analysis of a large pooled US cohort evaluated the cumulative and relative impact of the absence of five modifiable HF risk factors (diabetes, hypertension, obesity, dyslipidemia, smoking) in middle age (45 to 55 years of age) (129). The data showed that the absence of diabetes in middle age strongly predicted HF-free survival, with a more than 60% lower risk of incident HF than those with diabetes. In addition, subjects without diabetes, hypertension, and obesity at ages 45 to 55 years, compared to those with all 3 of these risk factors, had an average >10 years longer HF-free survival and 13 years longer overall survival (129).

 

Prediction of HF Risk in Patients with DM

 

HF risk stratification is essential for prevention in patients with DM or prediabetes who do not have ASCVD. Even though echocardiography can detect cardiac remodeling in patients with diabetes, its use is not routinely recommended for asymptomatic individuals because of concerns about cost-effectiveness (4). However, measuring natriuretic peptides or high-sensitivity cardiac troponin can help identify patients with pre-HF or those at risk of progression to HF. Therefore, patients with diabetes should have a measurement of natriuretic peptides or high-sensitivity cardiac troponin yearly to identify high-risk individuals and assist with HF prevention (4).

 

Several risk prediction tools and algorithms have been developed to predict incident HF in patients with dysglycemia. Pandey et al. described a simple biomarker-based risk score including high-sensitivity cardiac troponin T ≥6 ng/l, high-sensitivity C-reactive protein ≥3 mg/l, N-terminal pro-B-type natriuretic peptide (NT-proBNP) ≥125 pg/ml, and LVH by ECG, with each abnormal parameter counting as 1 point. This risk score was tested in participants from 3 major US cohort studies. This risk score had good risk stratification for predicting 5-year incident HF risk in patients with both diabetes and prediabetes (130). More complex risk prediction tools (WATCH-DM and TRS-HFDM) incorporating a more extensive list of clinical variables have also been developed. Validation studies on participants from different clinical trials demonstrated that these risk scores could stratify HF risk among patients with T2DM (131–133). However, prospective studies have not evaluated these risk scores, and their clinical utility remains uncertain.  

 

Glycemic control, obesity management, and BP control are well-established therapeutic options to lower the risk of microvascular or macrovascular complications in patients with diabetes. The clinical implications of these therapeutic options in HF prevention will be reviewed here.

 

Impact of Blood Pressure Control on HF Prevention

 

Diabetes and hypertension commonly coexist because of the overlap of underlying risk factors and pathophysiological mechanisms (134,135). The coexistence of diabetes and hypertension synergistically contributes to the risk of microvascular and macrovascular complications and CVD. Therefore, BP control with lifestyle modifications and antihypertensive medications is a primary target for reducing the risk of complications due to coexisting diabetes and hypertension (134). The ACC/AHA guidelines recommend initiating an antihypertensive agent when patients with diabetes have an office BP of ≥140/90 mmHg. The recommended BP target is below 140/90 mmHg for low-risk patients and below 130/80 for individuals with established or high risk for atherosclerotic CVD (136).

 

BP lowering has strong benefits in preventing HF among individuals with diabetes (35). However, the magnitude of this benefit appears to be smaller in patients with diabetes than in those without diabetes. Large meta-analyses of RCTs of BP-lowering therapy demonstrated that every 10 mmHg reduction in systolic BP (SBP) was associated with a 16% to 25% lower risk of HF among individuals with diabetes and 25% to 43% risk reduction among those without diabetes (137,138). The landmark ACCORD BP (Action to Control Cardiovascular Risk in Diabetes Blood Pressure) trial compared the impact of intensive (SBP goal <120 mmHg) versus standard BP control (SBP <140 mmHg) on major adverse CV events (MACE) in hypertensive patients with diabetes (139). In this trial, intensive BP control did not improve the risk of MACE or HF but increased the risk of adverse events. Therefore, the 2018 European Society of Cardiology (ESC)/European Society of Hypertension guidelines for the management of arterial hypertension recommended avoiding a target SBP <120 mmHg in patients with diabetes (140).

 

The antihypertensive drug classes with strong CV risk reduction in individuals with diabetes are thiazide diuretics, angiotensin-converting enzyme (ACE) inhibitors, angiotensin receptor blockers (ARBs), and dihydropyridine calcium channel blockers. Therefore, any of these agents can be considered a first-line therapy for lowering BP in individuals with diabetes. Because of their renal protection benefits, an ACE inhibitor or an ARB should be a part of the initial therapy for those with albuminuria (136,141). It should be noted that beta-blockers are not among the first-line antihypertensive agents for patients with or without diabetes. Because there is insufficient evidence on the mortality benefits of beta-blockers when used for the sole purpose of BP reduction in the absence of HF or CAD (136,141). The landmark Antihypertensive and Lipid-Lowering Treatment to Prevent Heart Attack (ALLHAT) has been the largest trial designed to assess the relative efficacy of different antihypertensive agents in protection against CVD. The results of this trial demonstrated that chlorthalidone (a thiazide diuretic) was superior to lisinopril (an ACE inhibitor) and amlodipine (a calcium-channel blocker) in the prevention of HF among patients with or without diabetes (142,143). A subsequent large meta-analysis of RCTs revealed that diuretics and renin-angiotensin system blockers were independently superior to other antihypertensive agents in the prevention of HF among patients with diabetes (138).

 

Loop diuretics are considered to have a neutral effect on glycemic control in patients with diabetes. Evidence from posthoc analysis and meta-analysis of antihypertensive drug trials has shown a small but significant association between thiazide diuretic use and higher fasting plasma glucose (144). A meta-analysis of 26 RCTs demonstrated that thiazide diuretic use was associated with a 4.6 mg/dL higher fasting plasma glucose than non-thiazide agents or placebo in patients with hypertension (145). A possible link between thiazide diuretic use and new-onset diabetes in patients with hypertension was supported by some studies (146) but not all (147). Overall, the CV benefits of thiazide diuretics outweigh the risk of new-onset diabetes in non-diabetic individuals and the risk of uncontrolled glycemia in patients with diabetes (144). Therefore, neither hypertension nor HF management guidelines recommend avoidance of thiazide diuretics in patients with or at risk for diabetes.

 

The ACC/AHA guidelines recommend the use of steroidal mineralocorticoid receptor antagonists (MRA, i.e., spironolactone and eplerenone) as an add-on therapy to the first-line antihypertensive agents in the treatment of resistant hypertension (136). Besides modest BP reduction, these agents can provide anti-fibrotic, anti-inflammatory, and anti-proteinuric benefits (148,149). The clinical CV benefits of these agents have been proven in patients with HFrEF. However, we lack large-scale RCTs demonstrating their CV benefits in primary prevention settings among patients with hypertension or diabetes. In a small RCT including 140 patients with T2DM and high CVD risk, adding high-dose eplerenone to standard treatment reduced LV mass and decreased NT-ProBNP and a circulating serum marker of myocardial fibrosis (150). Our knowledge of MRAs in patients with diabetes has expanded with two Phase III landmark RCTs evaluating the cardiorenal benefits of Fineranone, a recently discovered non-steroidal MRA with high affinity and specificity for the mineralocorticoid receptor (151). FIDELIO-DKD and FIGARO-DKD trials demonstrated that fineranone therapy on top of maximally tolerated RAAS inhibitor treatment was renally protective and reduced the risk of the primary endpoint of CVD outcomes in patients with T2DM and CKD (152,153). More specifically, fineranone therapy significantly reduced the risk of hospitalization for HF (HR; 0.71 [95% CI, 0.56-0.90]) in the FIGARO-DKD trial (153). Based on these results, the U.S. Food and Drug Administration (FDA) recently approved fineranone to reduce the risk of estimated glomerular filtration rate (eGFR) decline, end-stage renal disease, CVD death, non-fatal MI, and HF hospitalization in patients with CKD and T2DM (151).    

     

Obesity Management and Lifestyle Modifications

 

Studies have consistently demonstrated that purposeful weight reduction, achieved via diet, exercise, or bariatric surgery, has favorable impacts on glycemic control, IR, BP, and lipid profiles and reduces the need for antidiabetics in obese individuals with T2DM (34,154,155). Moreover, weight reduction can delay the progression from prediabetes to T2DM (156). The ADA recommends lifestyle modification to achieve at least a 5% weight loss for all overweight or obese individuals with prediabetes or diabetes (157). Also, the guidelines emphasize the need for an individualized medical nutrition therapy program for individuals with diabetes to achieve treatment goals. The recommended exercise regimen for most individuals with T1DM and T2DM is at least 150 minutes of moderate to vigorous aerobic activity per week, spread over at least three days/week, and an additional 2-3 sessions/week of resistance exercise (157).

 

Despite the well-established favorable effects of weight loss in patients with diabetes, the role of lifestyle changes and weight loss in preventing HF among diabetic patients remains uncertain. A meta-analysis of 36 prospective cohort studies published before 2014 demonstrated that achieving recommended physical activity levels (150 minutes of moderate‐intensity aerobic activity per week) was associated with reduced risk of incident HF (relative risk; 0.81 [0.76, 0.86]) in patients with diabetes (112).

 

The Look AHEAD (Action for Health in Diabetes) has been the largest RCT evaluating the CV effects of an intensive lifestyle intervention that promoted weight loss through decreased caloric intake and increased physical activity in overweight or obese participants with T2DM (154). The study subjects in the intensive lifestyle intervention group lost 8.6% of body weight at one year and, by the end of the 10-year follow-up, maintained a modest (6%) weight loss compared to 3.5% in the control group. Despite the achieved relative weight loss and improved physical fitness and HbA1c, the intensive lifestyle intervention did not reduce the risk of CVD mortality and morbidity, including HF risk, which was a secondary outcome. A post hoc analysis by Pandey et al. evaluated the impact of cardiorespiratory fitness and the degree of weight loss on the HF risk among the Look AHEAD trial participants with an extended follow-up period (158). The investigators observed a 20% reduction in incident HF risk as a response to a 10% reduction in BMI over a 4-year follow-up. Moreover, a higher baseline and improvement of cardiorespiratory fitness over time predicted a lower risk of incident HF. Interestingly, subgroup analysis revealed a more significant correlation between baseline fitness and incident HFpEF than incident HFrEF.  Future dedicated studies are needed to explore the HF risk reduction effects of more intense weight loss and exercise training interventions to promote sustained improvements in body weight and cardiorespiratory fitness in patients with T2DM (158).

 

Metabolic surgeries, when performed as a part of a comprehensive weight management strategy, are effective treatment options for achieving more significant and durable weight loss in individuals with severe obesity (159). Individuals with T2DM and severe obesity who undergo metabolic surgeries experience improvement in glycemic control and insulin sensitivity and may have remission of diabetes (159). Evidence from RCTs and observational studies has demonstrated that metabolic surgery, as compared to conventional lifestyle modifications and medical therapy, can reduce overall CV risk and improve the quality of life in individuals with T2DM and severe obesity (160–162). The impact of metabolic surgeries on incident HF risk has not yet been evaluated in large-scale RCTs. In a large nationwide prospective observational study of obese individuals without a known history of HF, metabolic surgery was associated with a >50% reduction in the risk of incident HF (163). Another retrospective cohort study from the Cleveland Clinic Health System in the U.S. demonstrated that metabolic surgery among patients with T2DM and obesity was associated with approximately 40% relative reduction of major adverse CV event risk and more than 60% relative reduction of incident HF risk over a median follow-up of 3.9 years (164).

 

Impact of Glycemic Control

 

In patients with T1DM and T2DM, intensive glycemic control significantly reduces the risk and severity of microvascular complications (34). However, despite the solid epidemiologic link between poor glycemic control and HF risk, the effects of intensified glycemic control in preventing HF remain controversial. Early clinical trials that established the CV benefits and risks of intensive glycemic control did not include HF as a primary endpoint. However, post hoc or secondary outcome analyses of prospective trials have shed light on the relationship between glycemic control and HF prevention.

 

In the UK Prospective Study of Diabetes (UKPDS) published more than 20 years ago, intensive glycemic control with metformin, sulfonylureas, or insulin was compared to conventional therapy in adults with recently diagnosed T2DM. A post hoc analysis of the main trial demonstrated that each 1% reduction in mean HbA1c was associated with a 16% decrease in incident HF (165). However, similar results were not replicated by subsequent large-scale RCTs such as the ACCORD (166,167), the ADVANCE (Action in Diabetes and Vascular Disease: Preterax and Diamicron Modified Release Controlled Evaluation (168), and the VADT (Veterans Affair Diabetes Trial) (169) which failed to show any reduction in HF risk with intensive glucose control in patients with T2DM. Consistently, a meta-analysis of all these trials showed no overall effect of intensive glucose control on HF risk despite a modest (9%) reduction in the risk of major CV outcomes (170). These observations confirmed that blood-glucose-lowering and improvement of HbA1c are insufficient targets for preventing HF in patients with diabetes.

  

The treatment section below discusses the impact of specific antidiabetic agents on the prevention of HF in high-risk patients.

 

TREATMENT OF HF IN PATIENTS WITH DIABETES

 

The primary objectives in managing HF are to reduce mortality, prevent HF hospitalization, and improve patients’ clinical status, quality of life, and functional capacity (18). The major components of managing HF are lifestyle changes, education and support for HF self-management, monitoring, control of the underlying causes and associated comorbidities, pharmacologic therapy, cardiac rehabilitation, device therapies, mechanical circulatory support, and cardiac transplantation (171). The major society guidelines for the management of patients with HF include the ACC/AHA/HFSA guidelines published in 2022 (3) and the European Society of Cardiology (ESC) guidelines published in 2021 (18).

 

The main components of lifestyle changes recommended for patients with HF are physical exercise, smoking cessation, restriction of or abstinence from alcohol consumption, dietary modifications, and avoidance of obesity (18). ACC/AHA/HFSA and ESC guidelines strongly recommend regular aerobic exercise and exercise training to improve functional capacity and symptoms in patients with HF who can participate. The ACC/AHA/HFSA guideline indicated that avoiding excessive dietary sodium intake is reasonable (class IIa) for patients with symptomatic HF to reduce congestive symptoms (3). However, because of a lack of affirmative evidence from clinical trials, the guidelines did not provide precise recommendations about the limit of daily sodium intake and whether it should vary depending on the type, stage, or severity of HF or comorbidities.

 

Diuretic Therapy in HF

 

Diuretics increase urinary sodium excretion and reduce physical signs and symptoms of congestion in HF patients by inhibiting sodium or chloride reabsorption in the renal tubules. Loop diuretics such as bumetanide, furosemide, and torsemide act in the loop of Henle, whereas thiazide diuretics such as hydrochlorothiazide, metolazone, chlorthalidone, and potassium-sparing diuretics such as spironolactone, eplerenone, and triamterene act in the distal position of the tubule (172). Loop diuretics, which produce a shorter and more intense diuresis than thiazides, are usually the preferred agents for achieving and maintaining euvolemia and reducing the risk of HF hospitalization in patients with HF. Large-scale RCTs have not evaluated the effects of the loop and thiazide diuretics on mortality and morbidity in patients with HF.

  

First-Line Pharmacological Treatment of HFrEF

 

Neurohumoral antagonists (RAAS inhibitors and beta-blockers) and SGLT2 inhibitors with proven morbidity and mortality benefits are the cornerstone of guideline-directed medical therapy for patients with chronic HFrEF (173). The guidelines generally do not recommend specific therapeutic approaches in patients with diabetes compared to those without diabetes.

 

RAAS AND NEPRILYSIN INHIBITORS

 

The guidelines from AHA/ACC/HFSA (3) and ESC (18) recommend (Class I) the use of angiotensin receptor-neprilysin inhibitor (ARNI; sacubitril/valsartan) as a first-line therapy for patients with HFrEF (New York Heart Association [NYHA] Class II or III). ACE inhibitors (Class I recommendation) are recommended to reduce the risk of morbidity and mortality in patients with HFrEF when using ARNI is not feasible. ARBs are considered acceptable vasodilator treatment options (class I recommendation) as a first-line alternative to ACE inhibitors for patients intolerant of ACE inhibitors because of cough or angioedema and when using ARNI is not feasible.

 

Subgroup analysis or meta-analysis of major HF trials demonstrated that the effectiveness of RAAS and neprilysin inhibitors in HF does not vary based on patients’ diabetes status. Therapy with ACE inhibitors, ARBs, or an ARNI reduces the risk of morbidity and mortality among HF patients without significant effect variation based on diabetesstatus (33,61,122,174). In addition, ARNI therapy provides a similar degree of natriuretic peptide improvement and cardiac reverse remodeling in patients with or without diabetes (175).

 

Post hoc analysis of RCTs revealed that ACE inhibitors and ARBs might reduce the risk of incident diabetes in patients with HFrEF (59,61,176). However, the data on the impact of these agents on glycemic control in patients with HF and preexisting diabetes remains limited. Neprilysin inhibition with sacubitril appears to have more favorable effects on glycemic control than ACE inhibitors (61). In the PARADIGM-HF trial, HFrEF patients enrolled in the enalapril and sacubitril-valsartan arms experienced an average of 0.16% and 0.26% HbA1c reduction after one year of treatment (177). In addition, sacubitril-valsartan use was associated with a 29% reduction in new insulin use compared to enalapril.

 

Diabetes confers a higher risk of diabetic nephropathy and CKD. Diabetic nephropathy can lead to increased renal sodium retention and a higher risk of hyperkalemia. Therefore, it is critical to monitor serum electrolytes and creatinine when starting or escalating the dose of RAAS inhibitors in patients with HF and diabetes (178). Of note, ARNI therapy may cause a higher rate of hypotension than ACE inhibitors or ARB (4).

 

BETA-BLOCKERS

 

Beta-blocker therapy is recommended (class I) for all patients with stable, symptomatic HFrEF (3,18). Beta-blocker therapy reduces the risk of death and hospitalization and improves LVEF and clinical status in patients with HFrEF. The ACC/AHA/HFSA guidelines recommend using one of the three beta-blockers with proven mortality benefits (e.g., metoprolol succinate, carvedilol, and bisoprolol).

 

Based on RCTs, the morbidity and mortality benefits of beta-blockers are similar in HFrEF patients with or without diabetes (174,179,180). A prospective cohort study from the UK suggested that increasing beta-blocker dose was associated with a more significant prognostic advantage in HF patients with diabetes than those without diabetes(181).

 

Data from some old observational studies and clinical trials raised concerns for a slight increase in the risk of new-onset diabetes associated with using propranolol, a first-generation non-selective beta-blocker, to treat hypertension (182,183). However, such an adverse effect concern is not present with newer-generation beta-blockers in HF populations (184). Compared to other beta-blockers, carvedilol use may even reduce HbA1c, fasting insulin levels, and risk of new-onset diabetes in patients with HFrEF (60,184).

 

Beta-blockers can potentially mask hypoglycemia symptoms by preventing palpitations and tremors and could prolong recovery from hypoglycemia by reducing glucose production in the liver (178). A post hoc analysis of the ACCORD trial demonstrated a significant association between beta-blocker use and severe hypoglycemia risk in patients with diabetes (185). However, a post hoc analysis of the MERIT-HF trial (Metoprolol CR/XL Randomized Intervention Trial in  Chronic  Heart  Failure) did not show a similar association between beta-blocker use and hypoglycemia events in patients with HF and coexisting T2DM (186).

     

MINERALCORTICOID RECEPTOR INHIBITORS       

 

MRAs (i.e., spironolactone or eplerenone) are recommended for all patients with HFrEF (LVEF ≤35%) and NYHA class II to IV symptoms if eGFR is >30 ml/min/1.73 m2 and serum potassium is <5 mEq/L (3,18). Similar to other RAAS inhibitors, the clinical benefits of MRAs have been consistent in HF patients with or without diabetes (187). Based on limited data, MRAs appear to have a neutral effect on glycemic parameters and diabetes risk in individuals with HFrEF (64,188). MRAs can cause hyperkalemia; therefore, monitoring electrolytes while initiating or maintaining MRA therapy is crucial.

 

SGLT2 INHIBITORS

 

SGLT2 inhibitors are recommended (class I) to reduce CVD mortality and HF hospitalization in patients with symptomatic HFrEF, irrespective of the presence of T2DM (3,18). Empagliflozin, Dapagliflozin, and Sotagliflozin are the SGLT2 inhibitors approved by the U.S. FDA to reduce CVD death and HF hospitalization in patients with HF across the range of LVEF (HFrEF, HFpEF, HFmrEF).

 

Dedicated landmark trials proved the CV benefits of SGLT2 inhibitors in patients with HFrEF, regardless of T2DM status (Table 1). The DAPA-HF (Dapagliflozin and Prevention of Adverse Outcomes in Heart Failure) (189) and EMPEROR-Reduced (EMPagliflozin outcomE tRial in Patients With chrOnic heaRt Failure With Reduced Ejection Fraction) (190) trials enrolled symptomatic chronic HFrEF (LVEF ≤40%, NYHA class II to IV, and elevated natriuretic peptides) and were already on guideline-directed medical therapy. Patients with T1DM and advanced CKD were excluded.  In these trials, compared to placebo, SGLT2 inhibitors reduced the composite of CVD death and HF hospitalization by ~25%. In addition, SGLT2 inhibitors slowed the progression of renal disease. The SGLT2 inhibitors' CV benefits are independent of their glucose-lowering effects (191). In addition, SGLT2 inhibitor therapy appears to improve the clinical stability and functional status of patients with HF. An analysis from the EMPEROR-Reduced trial demonstrated that empagliflozin therapy was associated with improvement in the NYHA class and requirement of diuretic intensification when compared to placebo (192).

 

Sotagliflozin is a dual SGLT1/SGLT2 inhibitor that increases urinary glucose excretion by SGLT2 inhibition and delays intestinal glucose absorption by SGLT1 inhibition. The SOLOIST-WHF (Effect of Sotagliflozin on Cardiovascular Events in Patients With Type 2 Diabetes And Worsening Heart Failure) trial evaluated the efficacy and safety of sotagliflozin in patients with T2DM who were hospitalized with HF (HFpEF and HFrEF) (190). In this trial, Sotagliflozin therapy initiated before or shortly after hospital discharge reduced the combined endpoint of CVD death, HF hospitalization, or urgent HF visits by 33%.

 

Despite the guideline recommendations and strong clinical trial evidence supporting their benefits, SGLT2 inhibitor therapy utilization among patients with HF remains low. A recent nationwide retrospective cohort study analyzed the STLT2 inhibitor prescription patterns among patients hospitalized with HFrEF between July 2021 and July 2022 (193). Only 20% of eligible patients in this cohort were prescribed SGLT2 inhibitors at discharge. Moreover, the utilization was low even among patients with multiple indications, such as comorbid CKD and T2DM. 

Table 1. Trials of SGLT2 Inhibitors in Patients with HF

Medication

Trial

Publication Year

Patient Characteristics

History of T2DM

Follow-up Period

Primary Outcome*

(HR, 95% CI)

Empagliflozin

EMPEROR-REDUCED

(190)

2020

Symptomatic stable HF

(LVEF ≤40%)

50%

16 months

0.75

(0.65 – 0.86)

Empagliflozin

EMPEROR-PRESERVED

(194)

2021

Symptomatic stable HF

(LVEF > 40%)

49%

26 months

0.79

(0.69 – 0.90)

Dapagliflozin

DAPA-HF(189)

2019

Symptomatic stable HF

(LVEF ≤40%)

42%

18 months

0.74

(0.0.65 - 0.85)

Dapagliflozin

DELIVER(118)

2022

Symptomatic stable HF

(LVEF > 40%)

45%

2.3 years

0.82

(0.73 – 0.92)

Sotagliflozin

SOLOIST-WHF (195)

2021

Recently hospitalized HF (All LVEF groups)

100%

9 months

0.67

(0.52 – 0.85)

*Primary outcome was a composite of cardiovascular death or hospitalization for HF.

 

Device Therapies for HFrEF

 

Implantable cardioverter-defibrillator (ICD) is strongly recommended (Class I) for primary prevention of sudden cardiac death in patients with symptomatic HFrEF who have an LVEF ≤35% despite guideline-directed medical therapy for >3 months (3,18). HFrEF patients with diabetes carry a significantly higher risk of sudden cardiac death than those without diabetes (122). This observation highlights the importance of considering ICD in appropriately selected cases with HFrEF and diabetes. Strong evidence from major ICD trials has confirmed the sudden cardiac death risk reduction benefits of ICDs in individuals with coexisting HFrEF and diabetes (196,197).

 

Cardiac resynchronization therapy (CRT) is a well-established therapeutic modality in patients with HFrEF and prolonged QRS duration. In appropriately selected cases, CRT with biventricular pacing can improve LV systolic function and reduce the risk of morbidity and mortality through its ability to reverse the remodeling of LV (198). In major CRT trials, HFrEF patients with and without diabetes experienced similar overall effectiveness of CRT for reducing mortality and HF hospitalization (199–201). However, observational studies suggested that the magnitude of LV reverse remodeling and improvement of systolic and diastolic function may be less pronounced in individuals with diabetes than in those without diabetes (198,202,203).

 

Treatment of HFpEF

 

Until recently, the management of patients with HFpEF and HFmrEF lacked specific therapies shown to improve morbidity and mortality definitively. Clinical trials of pharmacologic agents with proven benefits in HFrEF have predominantly revealed neutral results in populations with HFpEF (30,31,204).

 

The guidelines focus on aggressive management of risk factors and comorbidities, exercise training, and symptom management with diuretics when volume overload findings are present in patients with HFpEF (3). The landscape of medical management of HFpEF has dramatically changed with the data from clinical trials evaluating the safety and efficacy of SGLT2 inhibitor therapy in patients with HFpEF (Table 1). The EMPEROR-Preserved (Empagliflozin Outcome Trial in Patients with Chronic Heart Failure with Preserved Ejection Fraction) enrolled 5988 symptomatic patients with HF with LVEF >40% and elevated natriuretic peptides (194). In this trial, compared to placebo, empagliflozin therapy reduced the primary composite outcome of CVD death or HF hospitalization by 21%, primarily driven by a 29% reduction in HF hospitalization. SGLT2 inhibitor therapy was beneficial regardless of the presence or absence of T2DM. Based on the results of the EMPEROR-Preserved trial, the 2022 ACC/AHA/HFSA guidelines recommended (class IIa) SGLT2 inhibitor therapy to reduce CV death and HF hospitalization in patients with HFpEF and HFmrEF (3). Since the publication of these guidelines, the DELIVER (Dapagliflozin Evaluation to Improve the Lives of Patients with Preserved Ejection Fraction Heart Failure) trial was completed (118). This trial demonstrated similar benefits of dapagliflozin therapy in patients with HF and LVEF >40%. Data from the PRESERVED-HF trial, a relatively small-size multicentric RCT, evaluated the impact of dapagliflozin on the quality of life and symptoms in patients with HFpEF. In this trial, 12 weeks of dapagliflozin treatment significantly improved patient-reported physical limitations and symptoms and objectively measured exercise tolerance (205).

 

The 2022 ACC/AHA/HFSA guidelines indicate that based on a subgroup analysis of RCTs, ARBs, MRAs, and ARNI (in appropriately selected patients) might be considered (Class IIb) in patients with HFpEF or HFmrEF to decrease hospitalizations (3).

 

PHARMACOLOGIC THERAPY OF T2DM IN PATIENTS WITH HF

Lifestyle therapy is essential to managing patients with diabetes and established or high risk for HF. We point the readers to documents from ADA and ACC/AHA for detailed review and recommendations on lifestyle therapy in this patient population (3,4,157).

 

In this chapter, we provide a focused review of the effects of glucose-lowering agents from a HF perspective. Glycemic control is essential in patients with diabetes who have additional CV risk factors or established CVD. The ADA Standards of Medical Care in Diabetes recommend a holistic, multifactorial, and patient-centered approach when choosing antidiabetic medications. As per the guidelines, antidiabetic therapy should be selected according to patient-specific goals such as cardiorenal protection or achieving and maintaining glycemic and weight management goals. Moreover, considering comorbidities, such as HF and CKD, is essential when determining management goals (23,141).

 

SGLT2 Inhibitors

 

In light of the evidence from CVOTs showing the benefits of CVD risk reduction and renal protection, the ADA recommends using GLP1-RAs or SGLT2 inhibitors as first-line agents in T2DM patients with high-risk or established ASCVD (23). In addition, SGLT2 inhibitors are the preferred first-line antidiabetic in patients with known HF or CKD (eGFR <60 ml/min/1.73 m2 or albuminuria). This approach is a change from before, as the guidelines no longer require first-line metformin therapy before initiating SGLT2 inhibitors or GLP1-RAs when the therapy is started with the goal of cardiorenal risk reduction in high-risk patients with T2DM. (Table 2).

 

Table 2.  Pharmacologic Therapy with a Goal of Cardiorenal Risk Reduction in High-Risk Patients with T2DM

Risk Profile

First-line Therapy

Second-line Therapy if A1C is Above the Target

ASCVD*

Indicators of High Risk**

GLP1-RA with proven CVD benefit

Or

SGLT2 inhibitor with proven CVD benefit

GLP1-RA with proven CVD benefit

Or

SGLT2 inhibitor with proven CVD benefit

Heart Failure

SGLT2 inhibitor with proven HF benefit

Follow the algorithm for the achievement of glycemic and weight management goals.

CKD***

SGLT2 Inhibitor with proven CKD benefits

GLP1-RA with proven CVD benefit

*ASCVD, atherosclerotic cardiovascular disease: Individuals with established cardiovascular disease such as myocardial infarction, stroke, revascularization procedure, amputation, or symptomatic/asymptomatic coronary artery disease. **Indicators of high risk: ≥55 years of age with two or more additional risk factors such as obesity, hypertension, dyslipidemia, smoking, and albuminuria. ***CKD, chronic kidney disease: GFR <60 ml/min/1.73 m2 or albuminuria (albumin-creatinine ratio ≥30 mg/g). GLP1-RA, glucagon-like peptide-1 receptor agonists; A1C, hemoglobin A1c, SGLT2, sodium-glucose co-transporter -2.

 

A wealth of high-quality data proves the HF benefits of SGLT2 inhibitors in patients with T2DM.  Since the publication of EMPA-REG OUTCOME (Empagliflozin Cardiovascular Outcome Event Trial in Type 2 Diabetes Mellitus Patients) trial results in 2015, (206) several large-scale CVOTs have revolutionized our understanding of the prevention of CV events and HF in patients with T2DM (Table 1 and 3).

 

In the EMPA-REG OUTCOME, empagliflozin, in the CANVAS (Canagliflozin Cardiovascular Assessment Study), canagliflozin and in the DECLARE-TIMI 58 (Dapagliflozin Effect on Cardiovascular Events–Thrombolysis in Myocardial Infarction 58), dapagliflozin significantly reduced incident HF events as a secondary end-point in patients with established or high risk for CVD (206–208). The CREDENCE (Canagliflozin and Renal Events in Diabetes with Established Nephropathy Clinical Evaluation) trial, designed to assess renal outcomes of canagliflozin, also showed a reduction in hospitalization for HF (HR:0.69, p<0.001) (209). In the VERTIS-CV (Cardiovascular Outcomes Following Ertugliflozin Treatment in Patients with Type 2 Diabetes Mellitus and Atherosclerotic Cardiovascular Disease) trial, ertugliflozin was non-inferior to placebo in regards to major CVD events in 8264 patients with T2DM and established CVD. In this trial, Ertugliflozin reduced the risk of HF hospitalization (an exploratory secondary outcome) by 30% (hazard ratio 0.70 [95% CI, 0.54 to 0.90]) (210).

 

A meta-analysis of 6 RCTs (EMPAREG-OUTCOME, CANVAS, DECLARE-TIMI 58, and CREDENCE, VERTIS-CV) explored the CV benefits of 4 SGLT2 inhibitors in a combined sample size of 46969 patients with T2DM. In this meta-analysis, SGLT2 inhibitor therapy was associated with a reduced risk of MACE (HR, 0.90; 95% CI, 0.85-0.95), kidney outcomes (HR, 0.62; 95% CI, 0.56-0.70), and a combined outcome of CVD death and HF hospitalization (HR, 0.78; 95% CI, 0.73-0.84) (118).

 

The SCORE trial (Effect of Sotagliflozin on Cardiovascular and Renal Events in Patients with Type 2 Diabetes and Moderate Renal Impairment Who Are at Cardiovascular Risk) evaluated the CV benefits of sotagliflozin, a dual SGLT1/SGLT2 inhibitor, against placebo in 10584 patients with T2DM, CKD, and risk of CVD. In this trial, Sotagliflozin reduced the coprimary end-point of MACE (HR; 0.84 [95% CI, 0.72 to 0.99])  and secondary end-point of HF hospitalization (HR; 0.67 [0.55–0.82]) (210).

 

In Europe and Japan, SGLT-2 inhibitors are approved as adjunctive therapy for T1DM in patients with a BMI of at least 27 kg/m2. This recommendation is primarily based on the assumption that CV and renal protective effects of SGLT2 inhibitors can be generalized to T1DM populations. However, we still lack large-scale RCTs assessing the cardiorenal benefits of SGLT2 inhibitors in T1DM (211). Considering the high risk of diabetic ketoacidosis and the lack of proven diabetic ketoacidosis risk mitigation strategies, the U.S. FDA denied the approval of SGLT2 inhibitors for T1DM in the U.S.

 

Table 3. Heart Failure Hospitalization Risk with SGLT-2 Inhibitors in Patients with T2DM

Medication

Trial

Publication Year

Patient Characteristics

History of HF

Follow-up Period

HF hospitalization (HR, 95% CI)

Empagliflozin

EMPA-REG OUTCOME

(206)

2015

Established CVD

10%

3.1 years

0.65

(0.50 - 0.85)

Empagliflozin

EMPA-KIDNEY

(212)

2023

CKD with or without albuminuria*

Not reported

2 years

0.84

(0.67 – 1.07) **

[p=0.15]

Canagliflozin

CANVAS Program

(207)

2017

CV risk factors (34%)

Established CVD (66%)

14%

3.2 years

0.67

(0.52 - 0.87)

Canagliflozin

CREDENCE

(209)

2019

CKD with albuminuria

15%

2.6 years

0.61

(0.47 - 0.80)

Dapagliflozin

DECLARE-TIMI 58(208)

2019

CV risk factors (59%)

Established CVD (41%)

10%

4.2 years

0.73

(0.61 - 0.88)

Dapagliflozin

DAPA-CKD

(213)

2020

CKD with albuminuria

11%

2.4 years

0.71

(0.55 – 0.92) **

Ertugliflozin

VERTIS-CV

(210)

2020

Established CVD

 24%

3.5 years

0.70

(0.54 - 0.90)

Sotagliflozin

SCORED

(214)

2020

CKD with a high risk for CVD

31%

16 months

0.67

(0.55 – 0.82)

*Estimated eGFR of 20 to 45 ml/min/1.73 m2, or eGFR of 45 to 90 ml/min/1.73 m2 with a urinary albumin-to-creatinine ratio of ≥ 200 mg/g. **Composite secondary outcome of cardiovascular death and heart failure hospitalization. CKD, chronic kidney disease; CVD, cardiovascular disease.

 

Glucagon-like Peptide-1 Receptor Agonists

 

As mentioned above, GLP1-RAs are among the first-line agents for glucose-lowering and cardiorenal risk reduction in T2DM patients with indicators of high-risk or established ASCVD (23). In addition, GLP1-RAs are recommended as second-line therapy in patients with CKD if SGLT2 inhibitor therapy is contraindicated or not tolerated or if the HbA1c remains above target despite using an SGLT2 inhibitor.

 

Landmark CVOTs have confirmed the favorable effects of GLP1-RAs on the risk of major CV events such as CV death, nonfatal myocardial infarction, or nonfatal stroke in patients with T2DM. However, individual trials revealed mostly neutral results regarding the effect of GLP1-RAs on HF outcomes in patients with T2DM and at risk for HF (Table 4) (215,216). Efpeglenatide was the only GLP-RA that significantly reduced HF hospitalization as a secondary end-point in a major CVOT, including T2DM patients with a history of CVD or CKD (217). U.S. FDA has not yet approved this agent.

 

Some early small-size trials raised concerns for increased risk of hospitalization or arrhythmia in response to GLP1-RA treatment in patients HFrEF (218). However, A recent meta-analysis of 7 RCTs, including 54,092 ambulatory patients with T2DM, revealed that GLP1-RAs reduced the composite of HF hospitalization and CVD death (HR 0.84, 95% CI: 0.76-0.92.) in patients without a prior history of HF. However, compared to placebo, GLP1-RAs did not reduce the same outcome in patients with a previous history of HF (219). Based on available data, GLP1-RAs may potentially prevent HF in patients with no history of HF. However, GLP1-RAs do not appear to reduce HF-related events in patients with a history of HF and coexisting diabetes (218).

 

GLP1-RAs (i.e., liraglutide and semaglutide) have also been approved as a medical therapy for weight loss in non-diabetic individuals with overweight or obesity. And in a landmark trial, treatment with tirzepatide, a novel combined glucose-dependent insulinotropic polypeptide and GLP1-RA, led to substantial and sustained weight loss and improvement of cardiometabolic parameters in individuals with obesity (220). Several ongoing CVOTs are expected to shed light on the CV-related efficacy and safety of semaglutide or tirzepatide therapy in people with obesity or overweight but without diabetes.

 

STEP-HFpEF (Effect of Semaglutide 2.4 mg Once Weekly on Function and Symptoms in Subjects with Obesity-related Heart Failure with Preserved Ejection Fraction) trial has been the first RCT demonstrating the benefits of weight loss with a GLP1-RA in non-diabetic patients with HFpEF and obesity (BMI  kg/m2) (221). In this trial, semaglutide once-weekly therapy for one year led to 13.3% weight loss and was associated with significant reductions in HF-related symptoms and physical limitations. In addition, patients treated with semaglutide experienced significant improvement in 6-minute wall distance and reduction levels of natriuretic peptide and c-reactive protein. Interestingly, fewer serious adverse events were observed with semaglutide than placebo. This trial was not powered to evaluate mortality and HF hospitalization outcomes properly. Another ongoing trial is investigating the impact of semaglutide in patients with HFpEF, obesity, and T2DM.

 

Metformin

 

As per the recent guidelines, metformin remains the preferred initial pharmacologic agent for treating T2DM if the treatment goal is achieving and maintaining glycemic control and weight management (23). The efficacy and safety of metformin in patients with HF have not been evaluated in dedicated prospective CVOTs. Therefore, metformin is no longer considered the first-line agent for cardiorenal renal risk reduction in T2DM patients with established or increased risk of HF. However, metformin can be used for glucose lowering in patients with T2DM and stable HF if eGFR remains >30 ml/min/1.73 m2. However, it should be avoided in hospitalized patients with HF (141).

 

Data from retrospective studies and post hoc analysis of prospective RCTs predominantly supported the benefits and safety of metformin in HF populations (222). A meta-analysis of 9 observational studies conducted in the 2000s and early 2010s showed reduced mortality and no change in safety outcomes with the use of metformin compared to sulfonylurea therapy (predominantly) in HF populations (223). It should be noted that the U.S. FDA removed HF from the contraindication list of metformin in 2006 since the lactic acidosis risk is rare, and the benefits of metformin use in patients with HF were supported by observational studies (61).

 

Table 4. Effect of Glucose-Lowering Drugs on Heart Failure Hospitalization Risk in Patients with T2DM

Drug Class

Medication

HF Hospitalization

SGLT-2 Inhibitors

Empagliflozin

35% reduced risk

Canagliflozin

33% reduced risk

Dapagliflozin

27% reduced risk

Ertugliflozin

30% reduced risk*

Dual SGLT1/SGLT2 Inhibitor

Sotagliflozin

33% reduced risk

GLP-1 Receptor Agonists

Liraglutide

Neutral effect

Lixisenatide

Neutral effect

Semaglutide

Neutral effect

Albiglutide

Neutral effect

Exenatide

Neutral effect

Efpeglenatide

39% reduced risk

DPP-4 Inhibitors

Saxagliptin

27% increased risk

Alogliptin

Neutral effect**

Sitagliptin

Neutral effect

Vildagliptin

Neutral effect

Linagliptin

Neutral effect

Thiazolidinediones

Rosiglitazone

Increased risk***

Pioglitazone

41% Increased risk

*Exploratory secondary outcome (primary outcome of MACE was similar to placebo). **Possible increase in the risk of HF hospitalization in patients without a history of HF at baseline (HR 1.76, 1·07–2·90). *** Increased risk of HF hospitalization and HF-related death (HR 2.10, 1.35-3.27).

 

DPP4 Inhibitors

 

CV safety and HF outcomes of DPP4 inhibitors have been examined in several large-scale CVOTs (Table 4). In the Savor-TIMI-53 trial, there was no significant difference between Saxagliptin and placebo regarding the primary outcome of CV events. However, saxagliptin use was associated with a 27% relative increase in the risk of HF hospitalizations (224). In the EXAMINE (Cardiovascular Outcomes Study of Alogliptin in Patients With Type 2 Diabetes and Acute Coronary Syndrome) trial, alogliptin use showed a non-significant trend towards (3.9% vs. 3.3%, p = 0.22) increased risk of HF hospitalizations in the entire study population with a history of T2DM and recent acute coronary syndrome (225). However, the subgroup analysis showed an unexpected increase (2.2% vs. 1.3%, p = 0.026) in the risk of HF hospitalizations among subjects without a previous history of HF at baseline. Contrarily, a similar increase in the risk of HF hospitalization was not observed with other DPP4 inhibitors in dedicated CVOTs (226,227). The mechanism of increased HF hospitalization risk with saxagliptin and alogliptin remains unknown.

 

A recent retrospective comparative-effectiveness study using a US health insurance data set compared the CVD outcomes among patients with T2DM who were prescribed a DPP4 inhibitor vs. an SGLT2 inhibitor (193). In this cohort, the DPP4 inhibitor therapy group experienced a higher risk of MACE and hospitalization for HF than those initiated on an SGLT2 inhibitor.

 

DPP4 inhibitors have become less popular due to their cost and limited effectiveness in CV risk reduction compared to SGLT2 inhibitors and GLP1-RAs. Therefore, they are not among the preferred first- or second-line agents for managing T2DM in patients with HF. Based on available data, saxagliptin should be avoided in patients with known or at risk for HF. Moreover, HF risk is listed in the prescribing precautions of alogliptin. 

 

Insulin

 

No solid evidence exists to suggest that, in T2DM, improving glycemic control with insulin lowers HF risk in at-risk patients or improves outcomes in those with established HF. Even though some observational studies suggested an increased risk of CV mortality and HF in T2DM patients treated with insulin (228), limited prospective trial data did not support such an association. The ORIGIN Trial (Outcome Reduction With Initial Glargine Intervention) was a CVOT that examined the early use of basal insulin (glargine) versus standard care in patients with T2DM or prediabetes and a high risk of CVD. The study found that insulin glargine was  neutral with regards to CV outcomes and HF events (229).

 

Sulfonylureas

 

Sulfonylureas (i.e., glipizide, glyburide, and glimepiride) are widely used glucose-lowering agents that promote weight gain and fluid retention (4). No dedicated RCTs evaluated the efficacy and safety of sulfonylureas in patients with HF. Data from post hoc analysis of prospective trials and observational studies have suggested either no change or worsening of HF outcomes with the use of sulfonylureas in patients with or without known HF. A recent population-based cohort study including older diabetic patients hospitalized for HF between 2006 and 2014 demonstrated that sulfonylurea initiation was associated with increased risk of future HF hospitalization (HR: 1.22; 95% CI: 1.00-1.48; P = 0.050)  and mortality  (HR: 1.24; 95% CI: 1.00-1.52; P = 0.045) (193).  Based on current evidence, SGLT2 inhibitors, GLP1-RAs, and metformin are strongly preferable; sulfonylurea use should be avoided in patients with established or high risk for HF (4,141).

 

Thiazolidinediones

 

Thiazolidinediones (rosiglitazone and pioglitazone) have been effective oral antidiabetics in treating T2DM, significantly reducing HbA1c. However, they carry a risk of HF exacerbation and thus should be avoided in patients with established or at risk for HF. (Table 4). Therefore, thiazolidinediones should not be used in patients with pre-HF or clinical HF (4).

 

European Medicines Agencies and the U.S. FDA restricted the use of rosiglitazone, citing concerns about increased HF exacerbation after the publication of the results of the RECORD trial (Rosiglitazone Evaluated for Cardiac Outcomes and Regulation of Glycaemia in Diabetes) (230). Similarly, the PROactive study (PROspective pioglitAzone Clinical Trial In macroVascular Events) showed an improvement in MACE but an increased risk of HF events with pioglitazone therapy in high-risk patients with T2DM (231). The worsening HF outcomes with the treatment of thiazolidinediones have mainly been attributed to their effect on fluid retention. Some studies have raised concerns about the adverse impact of thiazolidinediones on myocardial metabolism and remodeling (61,222). Because of the availability of better options and their potential unfavorable side effect profile, the popularity of thiazolidinediones has significantly declined.

 

CONCLUSIONS

 

HF and cardiomyopathy have a heterogeneous etiology in patients with diabetes. Diabetes-related comorbidities, such as CAD and hypertension, contribute to the pathogenesis of HF in patients with diabetes. The pathophysiologic link between diabetes and HF is multifactorial, involving various abnormal biochemical pathways. A complex interaction of these mechanisms contributes to the development of asymptomatic diastolic and systolic dysfunction, eventually leading to the clinical syndrome of HF.

 

The coexistence of diabetes and HF is a poor prognostic factor, and it poses a higher risk of HF hospitalization, all-cause mortality, and CVD mortality. Therefore, the prevention of asymptomatic cardiac remodeling and progression into symptomatic HF are among the primary goals of the clinical management of patients with diabetes. BP lowering has substantial benefits in preventing HF among individuals with diabetes. Despite the well-established favorable effects of weight loss, the role of lifestyle changes and weight loss in preventing HF among diabetic patients remains uncertain. Observational data demonstrated a significant reduction in HF outcomes in response to metabolic surgeries among patients with diabetes and morbid obesity.

 

Guideline-directed medical therapy for HF has robust morbidity and mortality benefits in individuals with or without diabetes. Subgroup analysis or meta-analysis of major HF RCTs demonstrated that the effectiveness of HF therapies such as beta-blockers, RAAS inhibitors, ARNI, and MRAs do not vary based on patients’ diabetes status, and these therapies lead to similar reductions in morbidity and mortality among HF patients with or without diabetes.

 

Since 2015, several landmark clinical trials of SGLT2 inhibitors and GLP1-RA have revolutionized our understanding of CVD risk reduction in patients with T2DM and have led to a paradigm shift in the clinical practice recommendations for managing T2DM. SGLT-2 inhibitors are the preferred agents in the glucose-lowering regimen independent of baseline HbA1c in T2DM patients with known or at risk for HF.

 

Several dedicated major clinical trials confirmed the CVD benefits of SGLT2 inhibitors in patients with established HF, regardless of LVEF or diabetes status. High-quality data from these clinical trials transformed SGLT2 inhibitors from a glucose-lowering agent to a HF drug. Moreover, SGLT2 inhibitors are the only medication class with U.S. FDA approval and strong guideline indication (class I or IIa) for all HF patients across different LVEF groups.

 

ACKNOWLEDGMENT

 

We thank Halis Kaan Akturk for his contribution to the previous version of this chapter.

 

REFERENCES

 

  1. Chen YT, Vaccarino V, Williams CS, Butler J, Berkman LF, Krumholz HM. Risk factors for heart failure in the elderly: A prospective community- based study. Am. J. Med. 1999;106(6):605–612.
  2. Kannel WBB, Hjortland M, Castelli WPP. Role of diabetes in congestive heart failure: The Framingham study. Am. J. Cardiol. 1974;34(1):29–34.
  3. Heidenreich PA, Bozkurt B, Aguilar D, Allen LA, Byun JJ, Colvin MM, Deswal A, Drazner MH, Dunlay SM, Evers LR, Fang JC, Fedson SE, Fonarow GC, Hayek SS, Hernandez AF, Khazanie P, Kittleson MM, Lee CS, Link MS, Milano CA, Nnacheta LC, Sandhu AT, Stevenson LW, Vardeny O, Vest AR, Yancy CW. 2022 AHA/ACC/HFSA Guideline for the Management of Heart Failure: A Report of the American College of Cardiology/American Heart Association Joint Committee on Clinical Practice Guidelines. Circulation 2022;145(18):E895–E1032.
  4. Pop-Busui R, Januzzi JL, Bruemmer D, Butalia S, Green JB, Horton WB, Knight C, Levi M, Rasouli N, Richardson CR. Heart Failure: An Underappreciated Complication of Diabetes. A Consensus Report of the American Diabetes Association. Diabetes Care 2022;45(7):1670–1690.
  5. Bozkurt B, Coats AJ, Tsutsui H, Abdelhamid M, Adamopoulos S, Albert N, Anker SD, Atherton J, Böhm M, Butler J, Drazner MH, Felker GM, Filippatos G, Fonarow GC, Fiuzat M, Gomez-Mesa JE, Heidenreich P, Imamura T, Januzzi J, Jankowska EA, Khazanie P, Kinugawa K, Lam CSP, Matsue Y, Metra M, Ohtani T, Francesco Piepoli M, Ponikowski P, Rosano GMC, Sakata Y, SeferoviĆ P, Starling RC, Teerlink JR, Vardeny O, Yamamoto K, Yancy C, Zhang J, Zieroth S. Universal Definition and Classification of Heart Failure: A Report of the Heart Failure Society of America, Heart Failure Association of the European Society of Cardiology, Japanese Heart Failure Society and Writing Committee of the Universal Definition o. J. Card. Fail.2021;27(4):387–413.
  6. Nichols GA, Gullion CM, Koro CE, Ephross SA, Brown JB. The incidence of congestive heart failure in type 2 diabetes: An update. Diabetes Care 2004;27(8):1879–1884.
  7. Jia G, Hill MAA, Sowers JRR. Diabetic cardiomyopathy: An update of mechanisms contributing to this clinical entity. Circ. Res. 2018;122(4):624–638.
  8. Bertoni AG, Hundley WG, Massing MW, Bonds DE, Burke GL, Goff DC. Heart Failure Prevalence, Incidence, and Mortality in the Elderly with Diabetes. Diabetes Care 2004;27(3):699–703.
  9. Thrainsdottir IS, Aspelund T, Thorgeirsson G, Gudnason V, Hardarson T, Malmberg K, Sigurdsson G, Rydén L.The association between glucose abnormalities and heart failure in the population-based Reykjavík Study. Diabetes Care 2005;28(3):612–616.
  10. Centers for Disease Control and Prevention. National Diabetes Statistics Report, 2020. Atlanta, GA: Centers for Disease Control and Prevention, US Department of Health and Human Services; 2020.
  11. Marso SP, Bain SC, Consoli A, Eliaschewitz FG, Jódar E, Leiter LA, Lingvay I, Rosenstock J, Seufert J, Warren ML, Woo V, Hansen O, Holst AG, Pettersson J, Vilsbøll T. Semaglutide and Cardiovascular Outcomes in Patients with Type 2 Diabetes. N. Engl. J. Med. 2016;375(19):1834–1844.
  12. McMurray JJV, Packer M, Desai AS, Gong J, Lefkowitz MP, Rizkala AR, Rouleau JL, Shi VC, Solomon SD, Swedberg K, Zile MR. Angiotensin–Neprilysin Inhibition versus Enalapril in Heart Failure. N. Engl. J. Med.2014;371(11):993–1004.
  13. Pfeffer MA, Swedberg K, Granger CB, Held P, McMurray JJV, Michelson EL, Olofsson B, Östergren J, Yusuf S.Effects of candesartan on mortality and morbidity in patients with chronic heart failure: The CHARM-overall programme. Lancet 2003;362(9386):759–766.
  14. Rubler S, Dlugash J, Yuceoglu YZ, Kumral T, Branwood AW, Grishman A. New type of cardiomyopathy associated with diabetic glomerulosclerosis. Am. J. Cardiol. 1972;30(6):595–602.
  15. Maack C, Lehrke M, Backs J, Heinzel FRR, Hulot JSS, Marx N, Paulus WJJ, Rossignol P, Taegtmeyer H, Bauersachs J, Bayes-Genis A, Brutsaert D, Bugger H, Clarke K, Cosentino F, De Keulenaer G, Cas ADD, González A, Huelsmann M, Iaccarino G, Lunde IGG, Lyon ARR, Pollesello P, Rena G, Riksen NPP, Rosano G, Staels B, Van Laake LWW, Wanner C, Farmakis D, Filippatos G, Ruschitzka F, Seferovic P, De Boer RAA, Heymans S. Heart failure and diabetes: Metabolic alterations and therapeutic interventions: A state-of-The-Art review from the Translational Research Committee of the Heart Failure Association-European Society of Cardiology. Eur. Heart J. 2018;39(48):4243–4254.
  16. Murtaza G, Virk HUHUH, Khalid M, Lavie CJJ, Ventura H, Mukherjee D, Ramu V, Bhogal S, Kumar G, Shanmugasundaram M, Paul TKK. Diabetic cardiomyopathy - A comprehensive updated review. Prog. Cardiovasc. Dis. 2019;62(4):315–326.
  17. Amato L, Paolisso G, Cacciatore F, Ferrara N, Ferrara P, Canonico S, Varricchio M, Rengo F. Congestive heart failure predicts the development of non-insulin-dependent diabetes mellitus in the elderly. The Osservatorio Geriatrico Regione Campania Group. Diabetes Metab. 1997;23(3):213–8.
  18. McDonagh TA, Metra M, Adamo M, Baumbach A, Böhm M, Burri H, Čelutkiene J, Chioncel O, Cleland JGF, Coats AJS, Crespo-Leiro MG, Farmakis D, Gardner RS, Gilard M, Heymans S, Hoes AW, Jaarsma T, Jankowska EA, Lainscak M, Lam CSP, Lyon AR, McMurray JJV, Mebazaa A, Mindham R, Muneretto C, Piepoli MF, Price S, Rosano GMC, Ruschitzka F, Skibelund AK, de Boer RA, Schulze PC, Abdelhamid M, Aboyans V, Adamopoulos S, Anker SD, Arbelo E, Asteggiano R, Bauersachs J, Bayes-Genis A, Borger MA, Budts W, Cikes M, Damman K, Delgado V, Dendale P, Dilaveris P, Drexel H, Ezekowitz J, Falk V, Fauchier L, Filippatos G, Fraser A, Frey N, Gale CP, Gustafsson F, Harris J, Iung B, Janssens S, Jessup M, Konradi A, Kotecha D, Lambrinou E, Lancellotti P, Landmesser U, Leclercq C, Lewis BS, Leyva F, Linhart A, Løchen ML, Lund LH, Mancini D, Masip J, Milicic D, Mueller C, Nef H, Nielsen JC, Neubeck L, Noutsias M, Petersen SE, Petronio AS, Ponikowski P, Prescott E, Rakisheva A, Richter D, Schlyakhto E, Seferovic P, Senni M, Sitges M, Sousa-Uva M, Tocchetti CG, Touyz R, Tschoepe C, Waltenberger J, Krim M, Hayrapetyan H, Moertl D, Mustafayev I, Kurlianskaya A, Depauw M, Kušljugić Z, Gatzov P, Agathangelou P, Melenovský V, Løgstrup BB, Mostafa AM, Uuetoa T, Lassus J, Logeart D, Kipiani Z, Chrysohoou C, Sepp R, Ingimarsdóttir IJ, O’Neill J, Gotsman I, Iacoviello M, Bajraktari G, Lunegova O, Kamzola G, Massih TA, Benlamin H, Žaliaduonyte D, Noppe S, Moore A, Vataman E, Boskovic A, Bennis A, Manintveld OC, Kostovska ES, Gulati G, Straburzyńska-Migaj E, Silva-Cardoso J, Rimbaş RC, Lopatin Y, Foscoli M, Stojkovic S, Goncalvesova E, Fras Z, Segovia J, Lindmark K, Maeder MT, Bsata W, Abid L, Altay H, Voronkov L, Davies C, Abdullaev T, Baigent CN, Antoniou S, Collet JP, Halvorsen S, Koskinas KC. 2021 ESC Guidelines for the diagnosis and treatment of acute and chronic heartfailure. Eur. Heart J. 2021;42(36):3599–3726.
  19. Targher G, Dauriz M, Laroche C, Temporelli PL, Hassanein M, Seferovic PM, Drozdz J, Ferrari R, Anker S, Coats A, Filippatos G, Crespo-Leiro MG, Mebazaa A, Piepoli MF, Maggioni A Pietro, Tavazzi L. In-hospital and 1-year mortality associated with diabetes in patients with acute heart failure: results from the ESC-HFA Heart Failure Long-Term Registry. Eur. J. Heart Fail. 2017;19(1):54–65.
  20. Demant MN, Gislason GH, Køber L, Vaag A, Torp-Pedersen C, Andersson C. Association of heart failure severity with risk of diabetes: A Danish nationwide cohort study. Diabetologia 2014;57(8):1595–1600.
  21. Pavlović A, Polovina M, Ristić A, Seferović JP, Veljić I, Simeunović D, Milinković I, Krljanac G, Ašanin M, Oštrić-Pavlović I, Seferović PM. Long-term mortality is increased in patients with undetected prediabetes and type-2 diabetes hospitalized for worsening heart failure and reduced ejection fraction. Eur. J. Prev. Cardiol.2019;26(1):72–82.
  22. Wilcox T, De Block C, Schwartzbard AZ, Newman JD. Diabetic Agents, From Metformin to SGLT2 Inhibitors and GLP1 Receptor Agonists: JACC Focus Seminar. J. Am. Coll. Cardiol. 2020;75(16):1956–1974.
  23. 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 M Lou, Prahalad P, Pratley RE, Seley JJ, Stanton RC, Gabbay RA. 9. Pharmacologic Approaches to Glycemic Treatment: Standards of Care in Diabetes—2023. Diabetes Care 2023;46(supp):S140–S157.
  24. Marsico F, Gargiulo P, Marra AM, Parente A, Paolillo S. Glucose Metabolism Abnormalities in Heart Failure Patients: Insights and Prognostic Relevance. Heart Fail. Clin. 2019;15(3):333–340.
  25. Braunwald E. Diabetes, heart failure, and renal dysfunction: The vicious circles. Prog. Cardiovasc. Dis.2019;62(4):298–302.
  26. Rosengren A, Vestberg D, Svensson AM, Kosiborod M, Clements M, Rawshani A, Pivodic A, Gudbjörnsdottir S, Lind M. Long-term excess risk of heart failure in people with type 1 diabetes: A prospective case-control study. Lancet Diabetes Endocrinol. 2015;3(11):876–885.
  27. Rosengren A, Edqvist J, Rawshani A, Sattar N, Franzén S, Adiels M, Svensson AM, Lind M, Gudbjörnsdottir S.Excess risk of hospitalisation for heart failure among people with type 2 diabetes. Diabetologia2018;61(11):2300–2309.
  28. Tenenbaum A, Motro M, Fisman EZ, Leor J, Freimark D, Boyko V, Mandelzweig L, Adler Y, Sherer Y, Behar S.Functional class in patients with heart failure is associated with the development of diabetes. Am. J. Med.2003;114(4):271–275.
  29. Tsao CW, Aday AW, Almarzooq ZI, Anderson CAM, Arora P, Avery CL, Baker-Smith CM, Beaton AZ, Boehme AK, Buxton AE, Commodore-Mensah Y, Elkind MSV, Evenson KR, Eze-Nliam C, Fugar S, Generoso G, Heard DG, Hiremath S, Ho JE, Kalani R, Kazi DS, Ko D, Levine DA, Liu J, Ma J, Magnani JW, Michos ED, Mussolino ME, Navaneethan SD, Parikh NI, Poudel R, Rezk-Hanna M, Roth GA, Shah NS, St-Onge MP, Thacker EL, Virani SS, Voeks JH, Wang NY, Wong ND, Wong SS, Yaffe K, Martin SS. Heart Disease and Stroke Statistics - 2023 Update: A Report from the American Heart Association. Circulation 2023;147(8):E93–E621.
  30. Solomon SD, McMurray JJV, Anand IS, Ge J, Lam CSP, Maggioni AP, Martinez F, Packer M, Pfeffer MA, Pieske B, Redfield MM, Rouleau JL, van Veldhuisen DJ, Zannad F, Zile MR, Desai AS, Claggett B, Jhund PS, Boytsov SA, Comin-Colet J, Cleland J, Düngen H-D, Goncalvesova E, Katova T, Kerr Saraiva JF, Lelonek M, Merkely B, Senni M, Shah SJ, Zhou J, Rizkala AR, Gong J, Shi VC, Lefkowitz MP. Angiotensin–Neprilysin Inhibition in Heart Failure with Preserved Ejection Fraction. N. Engl. J. Med. 2019;381(17):1609–1620.
  31. Pitt B, Pfeffer MA, Assmann SF, Boineau R, Anand IS, Claggett B, Clausell N, Desai AS, Diaz R, Fleg JL, Gordeev I, Harty B, Heitner JF, Kenwood CT, Lewis EF, O’Meara E, Probstfield JL, Shaburishvili T, Shah SJ, Solomon SD, Sweitzer NK, Yang S, McKinlay SM. Spironolactone for Heart Failure with Preserved Ejection Fraction. N. Engl. J. Med. 2014;370(15):1383–1392.
  32. Ishikawa Y, Lewis RD, Laing EM, Anderson AK, Zhang D, Quyyumi AA, Dunbar SB, Trivedi-Kapoor R, Sattler ELP. Prevalence and trends of type 2 diabetes mellitus and prediabetes among community-dwelling heart failure patients in the United States. Diabetes Res. Clin. Pract. 2022;184. doi:10.1016/J.DIABRES.2022.109191.
  33. Kristensen SL, Preiss D, Jhund PS, Squire I, Cardoso JS, Merkely B, Martinez F, Starling RC, Desai AS, Lefkowitz MP, Rizkala AR, Rouleau JL, Shi VC, Solomon SD, Swedberg K, Zile MR, McMurray JJV, Packer M.Risk Related to Pre-Diabetes Mellitus and Diabetes Mellitus in Heart Failure with Reduced Ejection Fraction: Insights from Prospective Comparison of ARNI with ACEI to Determine Impact on Global Mortality and Morbidity in Heart Failure Trial. Circ. Hear. Fail. 2016;9(1):1–12.
  34. Oktay AA, Akturk HK, Esenboğa K, Javed F, Polin NM, Jahangir E. Pathophysiology and Prevention of Heart Disease in Diabetes Mellitus. Curr. Probl. Cardiol. 2018;43(3):68–110.
  35. Bensimhon HF, Cavender MA. Hypertension Treatment in Diabetes: Focus on Heart Failure Prevention. Heart Fail. Clin. 2019;15(4):551–563.
  36. Albakri A. Diabetic cardiomyopathy: a review of literature on clinical status and meta-analysis of tissue doppler diagnostic method and the clinical value of intensive glycemic control. Med. Clin. Arch. 2018;2(4):1–12.
  37. Lundbæk K. DIABETIC ANGIOPATHY. A SPECIFIC VASCULAR DISEASE. Lancet 1954;263(6808):377–379.
  38. Seferović PM, Paulus WJ. Clinical diabetic cardiomyopathy: A two-faced disease with restrictive and dilated phenotypes. Eur. Heart J. 2015;36(27):1718–1727.
  39. Kannel WB, McGee DL. Diabetes and Cardiovascular Disease: The Framingham Study. JAMA1979;241(19):2035–2038.
  40. Uijl A, Koudstaal S, Direk K, Denaxas S, Groenwold RHH, Banerjee A, Hoes AW, Hemingway H, Asselbergs FW. Risk factors for incident heart failure in age- and sex-specific strata: a population-based cohort using linked electronic health records. Eur. J. Heart Fail. 2019;21(10):1197–1206.
  41. Sattar N, McMurray J, Borén J, Rawshani A, Omerovic E, Berg N, Halminen J, Skoglund K, Eliasson B, Gerstein HC, McGuire DK, Bhatt D, Rawshani A. Twenty Years of Cardiovascular Complications and Risk Factors in Patients With Type 2 Diabetes: A Nationwide Swedish Cohort Study. Circulation 2023;147(25):1872–1886.
  42. Patel K V., Segar MW, Lavie CJ, Kondamudi N, Neeland IJ, Almandoz JP, Martin CK, Carbone S, Butler J, Powell-Wiley TM, Pandey A. Diabetes Status Modifies the Association Between Different Measures of Obesity and Heart Failure Risk Among Older Adults: A Pooled Analysis of Community-Based NHLBI Cohorts. Circulation2022;145(4):268.
  43. Johansson I, Dahlström U, Edner M, Näsman P, Rydén L, Norhammar A. Prognostic Implications of Type 2 Diabetes Mellitus in Ischemic and Nonischemic Heart Failure. J. Am. Coll. Cardiol. 2016;68(13):1404–1416.
  44. Echouffo-Tcheugui JB, Ndumele CE, Zhang S, Florido R, Matsushita K, Coresh J, Skali H, Shah AM, Selvin E.Diabetes and Progression of Heart Failure: The Atherosclerosis Risk In Communities (ARIC) Study. J. Am. Coll. Cardiol. 2022;79(23):2285–2293.
  45. McAllister DA, Read SH, Kerssens J, Livingstone S, McGurnaghan S, Jhund P, Petrie J, Sattar N, Fischbacher C, Kristensen SL, McMurray J, Colhoun HM, Wild SH. Incidence of hospitalization for heart failure and case-fatality among 3.25 million people with and without diabetes mellitus. Circulation 2018;138(24):2774–2786.
  46. Kubicki DM, Xu M, Akwo EA, Dixon D, Muñoz D, Blot WJ, Wang TJ, Lipworth L, Gupta DK. Race and Sex Differences in Modifiable Risk Factors and Incident Heart Failure. JACC Hear. Fail. 2020;8(2):122–130.
  47. Chamberlain AM, Boyd CM, Manemann SM, Dunlay SM, Gerber Y, Killian JM, Weston SA, Roger VL. Risk Factors for Heart Failure in the Community: Differences by Age and Ejection Fraction. Am. J. Med.2020;133(6):e237–e248.
  48. Ohkuma T, Komorita Y, Peters SAE, Woodward M. Diabetes as a risk factor for heart failure in women and men: a systematic review and meta-analysis of 47 cohorts including 12 million individuals. Diabetologia2019;62(9):1550–1560.
  49. Sinha A, Ning H, Cameron N, Bancks M, Carnethon MR, Allen NB, Wilkins JT, Lloyd-jones DM, Khan SS.Atherosclerotic Cardiovascular Disease or Heart Failure: First Cardiovascular Event in Adults With Prediabetes and Diabetes. 2022. doi:10.1016/j.cardfail.2022.10.426.
  50. Held C, Gerstein HC, Yusuf S, Zhao F, Hilbrich L, Anderson C, Sleight P, Teo K, ONTARGET/TRANSCEND Investigators. Glucose levels predict hospitalization for congestive heart failure in patients at high cardiovascular risk. Circulation 2007;115(11):1371–5.
  51. Nelson AJ, Peterson ED, Pagidipati NJ. Atherosclerotic cardiovascular disease and heart failure: Determinants of risk and outcomes in patients with diabetes. Prog. Cardiovasc. Dis. 2019;62(4):306–314.
  52. Nyström T, Sartipy U, Contardi A, Lind M, Bellocco R, Eliasson B, Svensson AM, Holzmann MJ. Glycated Hemoglobin A1c Levels in Type 1 Diabetes Mellitus and Outcomes after Myocardial Infarction. Circulation2019;139(20):2380–2382.
  53. Rawshani A, Sattar N, Franzén S, Rawshani A, Hattersley AT, Svensson AM, Eliasson B, Gudbjörnsdottir S.Excess mortality and cardiovascular disease in young adults with type 1 diabetes in relation to age at onset: a nationwide, register-based cohort study. Lancet 2018;392(10146):477–486.
  54. Sattar N, Rawshani A, Franzén S, Rawshani A, Svensson AM, Rosengren A, Mcguire DK, Eliasson B, Gudbjörnsdottir S. Age at Diagnosis of Type 2 Diabetes Mellitus and Associations With Cardiovascular and Mortality Risks: Findings From the Swedish National Diabetes Registry. Circulation 2019;139(19):2228–2237.
  55. Echouffo-Tcheugui JB, Zhang S, Florido R, Hamo C, Pankow JS, Michos ED, Goldberg RB, Nambi V, Gerstenblith G, Post WS, Blumenthal RS, Ballantyne CM, Coresh J, Selvin E, Ndumele CE. Duration of Diabetes and Incident Heart Failure The ARIC (Atherosclerosis Risk In Communities) Study. 2021. doi:10.1016/j.jchf.2021.06.005.
  56. Shin D, Bohra C, Kongpakpaisarn K. Impact of the Discordance Between the American College of Cardiology/American Heart Association and American Diabetes Association Recommendations on Hypertension in Patients With Diabetes Mellitus in the United States. Hypertension 2018;72(2):256–259.
  57. Rawshani A, Rawshani A, Franzén S, Sattar N, Eliasson B, Svensson AM, Zethelius B, Miftaraj M, McGuire DK, Rosengren A, Gudbjörnsdottir S. Risk factors, mortality, and cardiovascular outcomes in patients with type 2 diabetes. N. Engl. J. Med. 2018;379(7):633–644.
  58. Yoo TK, Han K Do, Rhee EJ, Lee WY. Impact of mental disorders on the risk of heart failure among Korean patients with diabetes: a cohort study. Cardiovasc. Diabetol. 2023;22(1):1–13.
  59. Yusuf S, Ostergren JB, Gerstein HC, Pfeffer MA, Swedberg K, Granger CB, Olofsson B, Probstfield J, McMurray J V, Mortality C in HF-A of R in, Morbidity Program I. Effects of candesartan on the development of a new diagnosis of diabetes mellitus in patients with heart failure.[Erratum appears in Circulation. 2005 Oct 25;112(7):e292]. Circulation 2005;112(1):48–53.
  60. Torp-Pedersen C, Metra M, Charlesworth A, Spark P, Lukas MA, Poole-Wilson PA, Swedberg K, Cleland JGF, Di Lenarda A, Remme WJ, Scherhag A. Effects of metoprolol and carvedilol on pre-existing and new onset diabetes in patients with chronic heart failure: Data from the Carvedilol or Metoprolol European Trial (COMET). Heart 2007;93(8):968–973.
  61. Dunlay SM, Givertz MM, Aguilar D, Allen LA, Chan M, Desai AS, Deswal A, Dickson VV, Kosiborod MN, Lekavich CL, McCoy RG, Mentz RJ, Piña IL. Type 2 Diabetes Mellitus and Heart Failure: A Scientific Statement From the American Heart Association and the Heart Failure Society of America: This statement does not represent an update of the 2017 ACC/AHA/HFSA heart failure guideline update. Circulation 2019;140(7):e294–e324.
  62. Sud M, Wang X, Austin PC, Lipscombe LL, Newton GE, Tu J V., Vasan RS, Lee DS. Presentation blood glucose and death, hospitalization, and future diabetes risk in patients with acute heart failure syndromes. Eur. Heart J. 2015;36(15):924–931.
  63. Preiss D, Zetterstrand S, McMurray JJV, Östergren J, Michelson EL, Granger CB, Yusuf S, Swedberg K, Pfeffer MA, Gerstein HC, Sattar N. Predictors of development of diabetes in patients with chronic heart failure in the Candesartan in Heart Failure Assessment of Reduction in Mortality and Morbidity (CHARM) program. Diabetes Care 2009;32(5):915–920.
  64. Preiss D, Van Veldhuisen DJ, Sattar N, Krum H, Swedberg K, Shi H, Vincent J, Pocock SJ, Pitt B, Zannad F, McMurray JJV. Eplerenone and new-onset diabetes in patients with mild heart failure: Results from the Eplerenone in Mild Patients Hospitalization and Survival Study in Heart Failure (EMPHASIS-HF). Eur. J. Heart Fail. 2012;14(8):909–915.
  65. Ernande L, Audureau E, Jellis CL, Bergerot C, Henegar C, Sawaki D, Czibik G, Volpi C, Canoui-Poitrine F, Thibault H, Ternacle J, Moulin P, Marwick TH, Derumeaux G. Clinical Implications of Echocardiographic Phenotypes of Patients With Diabetes Mellitus. J. Am. Coll. Cardiol. 2017;70(14):1704–1716.
  66. Yildiz M, Oktay AA, Stewart MH, Milani R V., Ventura HO, Lavie CJ. Left ventricular hypertrophy and hypertension. Prog. Cardiovasc. Dis. 2020;63:10–21.
  67. Wang Y, Yang H, Huynh Q, Nolan M, Negishi K, Marwick TH. Diagnosis of Nonischemic Stage B Heart Failure in Type 2 Diabetes Mellitus: Optimal Parameters for Prediction of Heart Failure. JACC Cardiovasc. Imaging2018;11(10):1390–1400.
  68. Dawson A, Morris AD, Struthers AD. The epidemiology of left ventricular hypertrophy in type 2 diabetes mellitus. Diabetologia 2005;48(10):1971–1979.
  69. Hamid A, Yimer WK, Oshunbade AA, Kamimura D, Clark D, Fox ER, Min YI, Muntner P, Shimbo D, Pandey A, Shah AM, Mentz RJ, Jones DW, Bertoni AG, Hall JE, Correa A, Butler J, Hall ME. Impact of Diabetes and Hypertension on Left Ventricular Structure and Function: The Jackson Heart Study. J. Am. Hear. Assoc. Cardiovasc. Cerebrovasc. Dis. 2023;12(6):26463.
  70. Velagaleti RS, Gona P, Chuang ML, Salton CJ, Fox CS, Blease SJ, Yeon SB, Manning WJ, O’Donnell CJ.Relations of insulin resistance and glycemic abnormalities to cardiovascular magnetic resonance measures of cardiac structure and function: the Framingham Heart Study. Circ. Cardiovasc. Imaging 2010;3(3):257–63.
  71. Rutter MK, Parise H, Benjamin EJ, Levy D, Larson MG, Meigs JB, Nesto RW, Wilson PWF, Vasan RS. Impact of glucose intolerance and insulin resistance on cardiac structure and function: Sex-related differences in the Framingham Heart Study. Circulation 2003;107(3):448–454.
  72. Kishi S, Gidding SS, Reis JP, Colangelo LA, Venkatesh BA, Armstrong AC, Isogawa A, Lewis CE, Wu C, Jacobs DR, Liu K, Lima JAC. Association of Insulin Resistance and Glycemic Metabolic Abnormalities With LV Structure and Function in Middle Age: The CARDIA Study. JACC Cardiovasc. Imaging 2017;10(2):105–114.
  73. From AM, Scott CG, Chen HH. The development of heart failure in patients with diabetes mellitus and pre-clinical diastolic dysfunction a population-based study. J. Am. Coll. Cardiol. 2010;55(4):300–5.
  74. Galderisi M. Diastolic Dysfunction and Diabetic Cardiomyopathy. Evaluation by Doppler Echocardiography. J. Am. Coll. Cardiol. 2006;48(8):1548–1551.
  75. Marwick TH, Ritchie R, Shaw JE, Kaye D. Implications of Underlying Mechanisms for the Recognition and Management of Diabetic Cardiomyopathy. J. Am. Coll. Cardiol. 2018;71(3):339–351.
  76. Zhang L, Liebelt JJ, Madan N, Shan J, Taub CC. Comparison of Predictors of Heart Failure With Preserved Versus Reduced Ejection Fraction in a Multiracial Cohort of Preclinical Left Ventricular Diastolic Dysfunction. Am. J. Cardiol. 2017;119(11):1815–1820.
  77. Potter E, Marwick TH. Assessment of Left Ventricular Function by Echocardiography: The Case for Routinely Adding Global Longitudinal Strain to Ejection Fraction. JACC Cardiovasc. Imaging 2018;11(2P1):260–274.
  78. Smiseth OA, Torp H, Opdahl A, Haugaa KH, Urheim S. Myocardial strain imaging: how useful is it in clinical decision making? Eur. Heart J. 2016;37(15):1196–1207.
  79. Holland DJ, Marwick TH, Haluska BA, Leano R, Hordern MD, Hare JL, Fang ZY, Prins JB, Stanton T.Subclinical LV dysfunction and 10-year outcomes in type 2 diabetes mellitus. Heart 2015;101(13):1061–1066.
  80. Enomoto M, Ishizu T, Seo Y, Yamamoto M, Suzuki H, Shimano H, Kawakami Y, Aonuma K. Subendocardial Systolic Dysfunction in Asymptomatic Normotensive Diabetic Patients. Circ. J. 2015;79(8):1749–1755.
  81. Haley JE, Zhiqian G, Philip KR, Nicolas ML, Thomas KR, Lawrence DM, Elaine UM. Reduction in myocardial strain is evident in adolescents and young adults with obesity and type 2 diabetes. Pediatr. Diabetes2020;21(2):243–250.
  82. Rakha S, Aboelenin HM. Left ventricular functions in pediatric patients with ten years or more type 1 diabetes mellitus: Conventional echocardiography, tissue Doppler, and two-dimensional speckle tracking study. Pediatr. Diabetes 2019;20(7):946–954.
  83. Skali H, Shah A, Gupta DK, Cheng S, Claggett B, Liu J, Bello N, Aguilar D, Vardeny O, Matsushita K, Selvin E, Solomon S. Cardiac Structure and Function Across the Glycemic Spectrum in Elderly Men and Women Free of Prevalent Heart Disease: The Atherosclerosis Risk in the Community Study. Circ. Hear. Fail. 2015;8(3):448–454.
  84. Rørth R, Jhund PS, Mogensen UM, Kristensen SL, Petrie MC, Køber L, McMurray JJV. Risk of incident heart failure in patients with diabetes and asymptomatic left ventricular systolic dysfunction. Diabetes Care2018;41(6):1285–1291.
  85. Leung M, Wong VW, Hudson M, Leung DY. Impact of Improved Glycemic Control on Cardiac Function in Type 2 Diabetes Mellitus. Circ. Cardiovasc. Imaging 2016;9(3):e003643.
  86. Packer M. Differential Pathophysiological Mechanisms in Heart Failure With a Reduced or Preserved Ejection Fraction in Diabetes. JACC Hear. Fail. 2021;9(8):535–549.
  87. Bertero E, Maack C. Metabolic remodelling in heart failure. Nat. Rev. Cardiol. 2018;15(8):457–470.
  88. Marfella R, Amarelli C, Cacciatore F, Balestrieri ML, Mansueto G, D’Onofrio N, Esposito S, Mattucci I, Salerno G, De Feo M, D’Amico M, Golino P, Maiello C, Paolisso G, Napoli C. Lipid Accumulation in Hearts Transplanted From Nondiabetic Donors to Diabetic Recipients. J. Am. Coll. Cardiol. 2020;75(11):1249–1262.
  89. Doehner W, Rauchhaus M, Ponikowski P, Godsland IF, von Haehling S, Okonko DO, Leyva F, Proudler AJ, Coats AJS, Anker SD. Impaired insulin sensitivity as an independent risk factor for mortality in patients with stable chronic heart failure. J. Am. Coll. Cardiol. 2005;46(6):1019–26.
  90. Sanganalmath SK, Dubey S, Veeranki S, Narisetty K, Krishnamurthy P. The interplay of inflammation, exosomes and Ca2+ dynamics in diabetic cardiomyopathy. Cardiovasc. Diabetol. 2023;22(1):1–22.
  91. Jia G, Whaley-Connell A, Sowers JR. Diabetic cardiomyopathy: a hyperglycaemia- and insulin-resistance-induced heart disease. Diabetologia 2018;61(1):21–28.
  92. Falcão-Pires I, Hamdani N, Borbély A, Gavina C, Schalkwijk CG, van der Velden J, van Heerebeek L, Stienen GJM, Niessen HWM, Leite-Moreira AF, Paulus WJ. Diabetes mellitus worsens diastolic left ventricular dysfunction in aortic stenosis through altered myocardial structure and cardiomyocyte stiffness. Circulation2011;124(10):1151–9.
  93. Hinkel R, Hoewe A, Renner S, Ng J, Lee S, Klett K, Kaczmarek V, Moretti A, Laugwitz KL, Skroblin P, Mayr M, Milting H, Dendorfer A, Reichart B, Wolf E, Kupatt C. Diabetes Mellitus–Induced Microvascular Destabilization in the Myocardium. J. Am. Coll. Cardiol. 2017;69(2):131–143.
  94. DeFuria J, Belkina AC, Jagannathan-Bogdan M, Snyder-Cappione J, Carr JD, Nersesova YR, Markham D, Strissel KJ, Watkins A a, Zhu M, Allen J, Bouchard J, Toraldo G, Jasuja R, Obin MS, McDonnell ME, Apovian C, Denis G V, Nikolajczyk BS. B cells promote inflammation in obesity and type 2 diabetes through regulation of T-cell function and an inflammatory cytokine profile. Proc. Natl. Acad. Sci. U. S. A. 2013;110(13):5133–8.
  95. Murphy SP, Kakkar R, McCarthy CP, Januzzi JL. Inflammation in Heart Failure: JACC State-of-the-Art Review. J. Am. Coll. Cardiol. 2020;75(11):1324–1340.
  96. Vulesevic B, McNeill B, Giacco F, Maeda K, Blackburn NJR, Brownlee M, Milne RW, Suuronen EJ.Methylglyoxal-induced endothelial cell loss and inflammation contribute to the development of diabetic cardiomyopathy. Diabetes 2016;65(6):1699–1713.
  97. Tan X, Hu L, Shu Z, Chen L, Li X, Du M, Sun D, Mao X, Deng S, Huang K, Zhang F. Role of CCR2 in the development of streptozotocin-treated diabetic cardiomyopathy. Diabetes 2019;68(11):2063–2073.
  98. Gellen B, Thorin-Trescases N, Thorin E, Gand E, Ragot S, Montaigne D, Pucheu Y, Mohammedi K, Gatault P, Potier L, Liuu E, Hadjadj S, Saulnier PJ, Hadjadj S, Marechaud R, Ragot S, Piguel X, Saulnier PJ, Javaugue V, Gand E, Hulin-Delmotte C, Llatty P, Ducrocq G, Roussel R, Rigalleau V, Pucheu Y, Zaoui P, Montaigne D, Halimi JM, Gatault P, Sosner P, Gellen B. Increased serum S100A12 levels are associated with higher risk of acute heart failure in patients with type 2 diabetes. ESC Hear. Fail. 2022;9(6):3909–3919.
  99. Groves EM, Erande AS, Le C, Salcedo J, Hoang KC, Kumar S, Mohar DS, Saremi F, Im J, Agrawal Y, Nadeswaran P, Naderi N, Malik S. Comparison of epicardial adipose tissue volume and coronary artery disease severity in asymptomatic adults with versus without diabetes mellitus. Am. J. Cardiol. 2014;114(5):686–691.
  100. Christensen RH, Hansen CS, von Scholten BJ, Jensen MT, Pedersen BK, Schnohr P, Vilsbøll T, Rossing P, Jørgensen PG. Epicardial and pericardial adipose tissues are associated with reduced diastolic and systolic function in type 2 diabetes. Diabetes, Obes. Metab. 2019;21(8):2006–2011.
  101. Ng ACT, Strudwick M, van der Geest RJ, Ng ACC, Gillinder L, Goo SY, Cowin G, Delgado V, Wang WYS, Bax JJ. Impact of Epicardial Adipose Tissue, Left Ventricular Myocardial Fat Content, and Interstitial Fibrosis on Myocardial Contractile Function. Circ. Cardiovasc. Imaging 2018;11(8):e007372.
  102. Al-Talabany S, Mordi I, Graeme Houston J, Colhoun HM, Weir-McCall JR, Matthew SZ, Looker HC, Levin D, Belch JJF, Dove F, Khan F, Lang CC. Epicardial adipose tissue is related to arterial stiffness and inflammation in patients with cardiovascular disease and type 2 diabetes. BMC Cardiovasc. Disord. 2018;18(1):1–8.
  103. Nakanishi K, Fukuda S, Tanaka A, Otsuka K, Taguchi H, Shimada K. Relationships Between Periventricular Epicardial Adipose Tissue Accumulation, Coronary Microcirculation, and Left Ventricular Diastolic Dysfunction. Can. J. Cardiol. 2017;33(11):1489–1497.
  104. Lavie CJ, Oktay AA, Pandey A. Pericardial Fat and CVD: Is All Fat Created Equally? JACC Cardiovasc. Imaging 2017;10(9):1028–1030.
  105. Hoffman WH, Sharma M, Cihakova D, Talor M V., Rose NR, Mohanakumar T, Passmore GG. Cardiac antibody production to self-antigens in children and adolescents during and following the correction of severe diabetic ketoacidosis. Autoimmunity 2016;49(3):188–196.
  106. Sousa GR, Pober D, Galderisi A, Lv H, Yu L, Pereira AC, Doria A, Kosiborod M, Lipes MA. Glycemic Control, Cardiac Autoimmunity, and Long-Term Risk of Cardiovascular Disease in Type 1 Diabetes Mellitus. Circulation2019;139(6):730–743.
  107. Miller JA, Floras JS, Zinman B, Skorecki KL, Logan AG. Effect of hyperglycaemia on arterial pressure, plasma renin activity and renal function in early diabetes. Clin. Sci. 1996;90(3):189–195.
  108. Spallone V, Ziegler D, Freeman R, Bernardi L, Frontoni S, Pop-Busui R, Stevens M, Kempler P, Hilsted J, Tesfaye S, Low P, Valensi P. Cardiovascular autonomic neuropathy in diabetes: Clinical impact, assessment, diagnosis, and management. Diabetes. Metab. Res. Rev. 2011;27(7):639–653.
  109. Pop-Busui R, Cleary PA, Braffett BH, Martin CL, Herman WH, Low PA, Lima JAC, Bluemke DA. Association between cardiovascular autonomic neuropathy and left ventricular dysfunction: DCCT/EDIC study (diabetes control and complications trial/epidemiology of diabetes interventions and complications). J. Am. Coll. Cardiol.2013;61(4):447–454.
  110. Sacre JW, Franjic B, Jellis CL, Jenkins C, Coombes JS, Marwick TH. Association of cardiac autonomic neuropathy with subclinical myocardial dysfunction in type 2 diabetes. JACC Cardiovasc. Imaging2010;3(12):1207–1215.
  111. Vinik AI, Casellini C, Parson HK, Colberg SR, Nevoret ML. Cardiac autonomic neuropathy in diabetes: A predictor of cardiometabolic events. Front. Neurosci. 2018;12(AUG):591.
  112. Salvador DB, Gamba MR, Gonzalez-Jaramillo N, Gonzalez-Jaramillo V, Raguindin PFN, Minder B, Gräni C, Wilhelm M, Stettler C, Doria A, Franco OH, Muka T, Bano A. Diabetes and Myocardial Fibrosis: A Systematic Review and Meta-Analysis. JACC Cardiovasc. Imaging 2022;15(5):796–808.
  113. Ouyang C, You J, Xie Z. The interplay between autophagy and apoptosis in the diabetic heart. J. Mol. Cell. Cardiol. 2014;71(2014):71–80.
  114. Kuethe F, Sigusch HH, Bornstein SR, Hilbig K, Kamvissi V, Figulla HR. Apoptosis in patients with dilated cardiomyopathy and diabetes: A feature of diabetic cardiomyopathy? Horm. Metab. Res. 2007;39(9):672–676.
  115. He C, Zhu H, Li H, Zou MH, Xie Z. Dissociation of Bcl-2-Beclin1 complex by activated AMPK enhances cardiac autophagy and protects against cardiomyocyte apoptosis in diabetes. Diabetes 2013;62(4):1270–1281.
  116. Ljubkovic M, Gressette M, Bulat C, Cavar M, Bakovic D, Fabijanic D, Grkovic I, Lemaire C, Marinovic J.Disturbed fatty acid oxidation, endoplasmic reticulum stress, and apoptosis in left ventricle of patients with type 2 diabetes. Diabetes 2019;68(10):1924–1933.
  117. Van Den Berge JC, Constantinescu AA, Boiten HJ, Van Domburg RT, Deckers JW, Akkerhuis KM. Short- and long-term prognosis of patients with acute heart failure with and without diabetes: Changes over the last three decades. Diabetes Care 2018;41(1):143–149.
  118. Solomon SD, McMurray JJV, Claggett B, de Boer RA, DeMets D, Hernandez AF, Inzucchi SE, Kosiborod MN, Lam CSP, Martinez F, Shah SJ, Desai AS, Jhund PS, Belohlavek J, Chiang C-E, Borleffs CJW, Comin-Colet J, Dobreanu D, Drozdz J, Fang JC, Alcocer-Gamba MA, Al Habeeb W, Han Y, Cabrera Honorio JW, Janssens SP, Katova T, Kitakaze M, Merkely B, O’Meara E, Saraiva JFK, Tereshchenko SN, Thierer J, Vaduganathan M, Vardeny O, Verma S, Pham VN, Wilderäng U, Zaozerska N, Bachus E, Lindholm D, Petersson M, Langkilde AM. Dapagliflozin in Heart Failure with Mildly Reduced or Preserved Ejection Fraction. N. Engl. J. Med.2022;387(12):1089–1098.
  119. Dauriz M, Mantovani A, Bonapace S, Verlato G, Zoppini G, Bonora E, Targher G. Prognostic impact of diabetes on long-term survival outcomes in patients with heart failure: A meta-analysis. Diabetes Care 2017;40(11):1597–1605.
  120. Aguilar D, Bozkurt B, Ramasubbu K, Deswal A. Relationship of Hemoglobin A1C and Mortality in Heart Failure Patients With Diabetes. J. Am. Coll. Cardiol. 2009;54(5):422–428.
  121. McHugh K, DeVore AD, Wu J, Matsouaka RA, Fonarow GC, Heidenreich PA, Yancy CW, Green JB, Altman N, Hernandez AF. Heart Failure With Preserved Ejection Fraction and Diabetes: JACC State-of-the-Art Review. J. Am. Coll. Cardiol. 2019;73(5):602–611.
  122. MacDonald MR, Petrie MC, Varyani F, Ostergren J, Michelson EL, Young JB, Solomon SD, Granger CB, Swedberg K, Yusuf S, Pfeffer MA, McMurray JJ V, CHARM Investigators. Impact of diabetes on outcomes in patients with low and preserved ejection fraction heart failure: an analysis of the Candesartan in Heart failure: Assessment of Reduction in Mortality and morbidity (CHARM) programme. Eur. Heart J. 2008;29(11):1377–85.
  123. Aguilar D, Deswal A, Ramasubbu K, Mann DL, Bozkurt B. Comparison of Patients With Heart Failure and Preserved Left Ventricular Ejection Fraction Among Those With Versus Without Diabetes Mellitus. Am. J. Cardiol. 2010;105(3):373–377.
  124. Kristensen SL, Mogensen UM, Jhund PS, Petrie MC, Preiss D, Win S, Køber L, McKelvie RS, Zile MR, Anand IS, Komajda M, Gottdiener JS, Carson PE, McMurray JJ V. Clinical and Echocardiographic Characteristics and Cardiovascular Outcomes According to Diabetes Status in Patients With Heart Failure and Preserved Ejection FractionClinical Perspective. Circulation 2017;135(8):724–735.
  125. Lindman BR, Dávila-Román VG, Mann DL, McNulty S, Semigran MJ, Lewis GD, De Las Fuentes L, Joseph SM, Vader J, Hernandez AF, Redfield MM. Cardiovascular phenotype in HFpEF patients with or without diabetes: A RELAX trial ancillary study. J. Am. Coll. Cardiol. 2014;64(6):541–549.
  126. Cavender MA, Steg PG, Smith SC, Eagle K, Ohman EM, Goto S, Kuder J, Im K, Wilson PWF, Bhatt DL. Impact of Diabetes Mellitus on Hospitalization for Heart Failure, Cardiovascular Events, and Death: Outcomes at 4 Years from the Reduction of Atherothrombosis for Continued Health (REACH) Registry. Circulation2015;132(10):923–931.
  127. Sandesara PB, O’Neal WT, Kelli HM, Samman-Tahhan A, Hammadah M, Quyyumi AA, Sperling LS. The prognostic significance of diabetes and microvascular complications in patients with heart failure with preserved ejection fraction. Diabetes Care 2018;41(1):150–155.
  128. Bassi N, Fonarow GC. Prevention of Heart Failure in Patients with Diabetes: Role of Diabetes Medications. Curr. Cardiol. Rep. 2018;20(11):1–7.
  129. Ahmad FS, Ning H, Rich JD, Yancy CW, Lloyd-Jones DM, Wilkins JT. Hypertension, Obesity, Diabetes, and Heart Failure–Free Survival: The Cardiovascular Disease Lifetime Risk Pooling Project. JACC Hear. Fail.2016;4(12):911–919.
  130. Pandey A, Vaduganathan M, Patel K V, Ayers C, Ballantyne CM, Kosiborod MN, Carnethon M, Defilippi C, Mcguire DK, Khan SS, Caughey MC, De Lemos JA, Everett BM. Biomarker-Based Risk Prediction of Incident Heart Failure in Pre-Diabetes and Diabetes. 2021. doi:10.1016/j.jchf.2020.10.013.
  131. Berg DD, Wiviott SD, Scirica BM, Gurmu Y, Mosenzon O, Murphy SA, Bhatt DL, Leiter LA, McGuire DK, Wilding JPH, Johanson P, Johansson PA, Langkilde AM, Raz I, Braunwald E, Sabatine MS. Heart Failure Risk Stratification and Efficacy of Sodium-Glucose Cotransporter-2 Inhibitors in Patients With Type 2 Diabetes Mellitus. Circulation 2019;140(19):1569–1577.
  132. Segar MW, Patel K V, Hellkamp AS, Vaduganathan M, Lokhnygina Y, Green JB, Wan S-H, Kolkailah AA, Holman RR, Peterson ED, Kannan V, Willett DL, McGuire DK, Pandey A. Validation of the WATCH-DM and TRS-HF DM Risk Scores to Predict the Risk of Incident Hospitalization for Heart Failure Among Adults With Type 2 Diabetes: A Multicohort Analysis. J. Am. Hear. Assoc. J Am Hear. Assoc 2022;11:24094.
  133. Segar MW, Vaduganathan M, Patel K V., McGuire DK, Butler J, Fonarow GC, Basit M, Kannan V, Grodin JL, Everett B, Willett D, Berry J, Pandey A. Machine learning to predict the risk of incident heart failure hospitalization among patients with diabetes: The WATCH-DM risk score. Diabetes Care 2019;42(12):2298–2306.
  134. Yildiz M, Esenboğa K, Oktay AA. Hypertension and diabetes mellitus: highlights of a complex relationship. Curr. Opin. Cardiol. 2020;35(4):397–404.
  135. Khangura DS, Waqar Salam M, Brietzke SA, Sowers JR. Hypertension in Diabetes. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, Dungan K, Grossman A, Hershman JM, Kaltsas G, Koch C, Kopp P, Korbonits M, McLachlan R, Morley JE, New M, Perreault L, Purnell J, Rebar R, Singer F, Trence DL, Vinik A, Wilson DP, eds. Endotext. South Dartmouth (MA): MDText.com, Inc.; 2000; 2018.
  136. Whelton PK, Carey RM, Aronow WS, Casey DE, Collins KJ, Dennison Himmelfarb C, DePalma SM, Gidding S, Jamerson KA, Jones DW, MacLaughlin EJ, Muntner P, Ovbiagele B, Smith SC, Spencer CC, Stafford RS, Taler SJ, Thomas RJ, Williams KA, Williamson JD, Wright JT. 2017 ACC/AHA/AAPA/ABC/ACPM/AGS/APhA/ASH/ASPC/NMA/PCNA Guideline for the Prevention, Detection, Evaluation, and Management of High Blood Pressure in Adults: Executive Summary. JACC 2018;71(19):e127–e248.
  137. Ettehad D, Emdin CA, Kiran A, Anderson SG, Callender T, Emberson J, Chalmers J, Rodgers A, Rahimi K.Blood pressure lowering for prevention of cardiovascular disease and death: A systematic review and meta-analysis. Lancet 2016;387(10022):957–967.
  138. Thomopoulos C, Parati G, Zanchetti A. Effects of blood-pressure-lowering treatment on outcome incidence in hypertension:10-Should blood pressure management differ in hypertensive patients with and without diabetes mellitus? Overview and meta-analyses of randomized trials. J. Hypertens. 2017;35(5):922–944.
  139. Accord-BP-Study-Group. Effects of Intensive Blood-Pressure Control in Type 2 Diabetes Mellitus. N. Engl. J. Med. 2010;362(17):1575–1585.
  140. Williams B, Mancia G, Spiering W, Rosei EA, Azizi M, Burnier M, Clement DL, Coca A, De Simone G, Dominiczak A, Kahan T, Mahfoud F, Redon J, Ruilope L, Zanchetti A, Kerins M, Kjeldsen SE, Kreutz R, Laurent S, Lip GYH, McManus R, Narkiewicz K, Ruschitzka F, Schmieder RE, Shlyakhto E, Tsioufis C, Aboyans V, Desormais I, De Backer G, Heagerty AM, Agewall S, Bochud M, Borghi C, Boutouyrie P, Brguljan J, Bueno H, Caiani EG, Carlberg B, Chapman N, Cífková R, Cleland JGF, Collet JP, Coman IM, De Leeuw PW, Delgado V, Dendale P, Diener HC, Dorobantu M, Fagard R, Farsang C, Ferrini M, Graham IM, Grassi G, Haller H, Hobbs FDR, Jelakovic B, Jennings C, Katus HA, Kroon AA, Leclercq C, Lovic D, Lurbe E, Manolis AJ, McDonagh TA, Messerli F, Muiesan ML, Nixdorff U, Olsen MH, Parati G, Perk J, Piepoli MF, Polonia J, Ponikowski P, Richter DJ, Rimoldi SF, Roffi M, Sattar N, Seferovic PM, Simpson IA, Sousa-Uva M, Stanton A V., Van De Borne P, Vardas P, Volpe M, Wassmann S, Windecker S, Zamorano JL. 2018 ESC/ESH Guidelines for themanagement of arterial hypertension. Eur. Heart J. 2018;39(33):3021–3104.
  141. Elsayed NA, Aleppo G, Aroda VR, Bannuru RR, Brown FM, Bruemmer D, Collins BS, Das SR, Hilliard ME, Isaacs D, Johnson EL, Kahan S, Khunti K, Kosiborod M, Leon J, Lyons SK, Perry M Lou, Prahalad P, Pratley RE, Seley JJ, Stanton RC, Gabbay RA. 10. Cardiovascular Disease and Risk Management: Standards of Care in Diabetes—2023. Diabetes Care 2023;46(January):S158–S190.
  142. Whelton PK, Barzilay J, Cushman WC, Davis BR, Ilamathi E, Kostis JB, Leenen FHH, Louis GT, Margolis KL, Mathis DE, Moloo J, Nwachuku C, Panebianco D, Parish DC, Pressel S, Simmons DL, Thadani U. Clinical outcomes in antihypertensive treatment of type 2 diabetes, impaired fasting glucose concentration, and normoglycemia: Antihypertensive and Lipid-Lowering Treatment to Prevent Heart Attack Trial (ALLHAT). Arch. Intern. Med. 2005;165(12):1401–1409.
  143. Wright JT, Probstfield JL, Cushman WC, Pressel SL, Cutler JA, Davis BR, Einhorn PT, Rahman M, Whelton PK, Ford CE, Haywood LJ, Margolis KL, Oparil S, Black HR, Alderman MH. ALLHAT findings revisited in the context of subsequent analyses, other trials, and meta-analyses. Arch. Intern. Med. 2009;169(9):832–842.
  144. Scheen AJ. Type 2 Diabetes and Thiazide Diuretics. Curr. Diab. Rep. 2018;18(2):1–13.
  145. Zhang X, Zhao Q. Association of Thiazide-Type Diuretics With Glycemic Changes in Hypertensive Patients: A Systematic Review and Meta-Analysis of Randomized Controlled Clinical Trials. J. Clin. Hypertens.2016;18(4):342–351.
  146. Elliott WJ, Meyer PM. Incident diabetes in clinical trials of antihypertensive drugs: a network meta-analysis. Lancet 2007;369(9557):201–207.
  147. Yang Y, Xu H. Comparing six antihypertensive medication classes for preventing new-onset diabetes mellitus among hypertensive patients: a network meta-analysis. J. Cell. Mol. Med. 2017;21(9):1742–1750.
  148. Williams B, Macdonald TM, Morant S, Webb DJ, Sever P, McInnes G, Ford I, Cruickshank JK, Caulfield MJ, Salsbury J, Mackenzie I, Padmanabhan S, Brown MJ. Spironolactone versus placebo, bisoprolol, and doxazosin to determine the optimal treatment for drug-resistant hypertension (PATHWAY-2): A randomised, double-blind, crossover trial. Lancet 2015;386(10008):2059–2068.
  149. Agarwal R, Kolkhof P, Bakris G, Bauersachs J, Haller H, Wada T, Zannad F. Steroidal and non-steroidal mineralocorticoid receptor antagonists in cardiorenal medicine. Eur. Heart J. 2021;42(2):152–161.
  150. Brandt-Jacobsen NH, Lav Madsen P, Johansen ML, Rasmussen JJ, Forman JL, Holm MR, Rye Jørgensen N, Faber J, Rossignol P, Schou M, Kistorp C. Mineralocorticoid Receptor Antagonist Improves Cardiac Structure in Type 2 Diabetes: Data From the MIRAD Trial. JACC Hear. Fail. 2021;9(8):550–558.
  151. Pandey AK, Bhatt DL, Cosentino F, Marx N, Rotstein O, Pitt B, Pandey A, Butler J, Verma S. Non-steroidal mineralocorticoid receptor antagonists in cardiorenal disease. Eur. Heart J. 2022;43(31):2931–2945.
  152. Bakris GL, Agarwal R, Anker SD, Pitt B, Ruilope LM, Rossing P, Kolkhof P, Nowack C, Schloemer P, Joseph A, Filippatos G. Effect of Finerenone on Chronic Kidney Disease Outcomes in Type 2 Diabetes. N. Engl. J. Med.2020;383(23):2219–2229.
  153. Pitt B, Filippatos G, Agarwal R, Anker SD, Bakris GL, Rossing P, Joseph A, Kolkhof P, Nowack C, Schloemer P, Ruilope LM. Cardiovascular Events with Finerenone in Kidney Disease and Type 2 Diabetes. N. Engl. J. Med.2021;385(24):2252–2263.
  154. The (Look AHEAD Researh Group). Cardiovascular Effects of Intensive Lifestyle Intervention in Type 2 Diabetes. N. Engl. J. Med. 2013;369(2):145–154.
  155. Wing RR. Long-term effects of a lifestyle intervention on weight and cardiovascular risk factors in individuals with type 2 diabetes mellitus: four-year results of the Look AHEAD trial. Arch. Intern. Med. 2010;170(17):1566–75.
  156. Diabetes Prevention Program Research Group. Reduction in the Incidence of Type 2 Diabetes with Lifestyle Intervention or Metformin. N. Engl. J. Med. 2002;346(6):393–403.
  157. 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 M Lou, Prahalad P, Pratley RE, Seley JJ, Stanton RC, Young-Hyman D, Gabbay RA. 5. Facilitating Positive Health Behaviors and Well-being to Improve Health Outcomes: Standards of Care in Diabetes—2023. Diabetes Care 2023;46(January):S68–S96.
  158. Pandey A, Patel K V, Bahnson JL, Gaussoin SA, Martin CK, Balasubramanyam A, Johnson KC, Mcguire DK, Bertoni AG, Kitzman D, Berry JD. Association of Intensive Lifestyle Intervention, Fitness, and Body Mass Index with Risk of Heart Failure in Overweight or Obese Adults with Type 2 Diabetes Mellitus: An Analysis from the Look AHEAD Trial. Circulation 2020;141(16):1295–1306.
  159. Pareek M, Schauer PR, Kaplan LM, Leiter LA, Rubino F, Bhatt DL. Metabolic Surgery: Weight Loss, Diabetes, and Beyond. J. Am. Coll. Cardiol. 2018;71(6):670–687.
  160. Mingrone G, Panunzi S, De Gaetano A, Guidone C, Iaconelli A, Nanni G, Castagneto M, Bornstein S, Rubino F.Bariatric-metabolic surgery versus conventional medical treatment in obese patients with type 2 diabetes: 5 Year follow-up of an open-label, single-centre, randomised controlled trial. Lancet 2015;386(9997):964–973.
  161. Schauer PR, Bhatt DL, Kirwan JP, Wolski K, Aminian A, Brethauer SA, Navaneethan SD, Singh RP, Pothier CE, Nissen SE, Kashyap SR. Bariatric Surgery versus Intensive Medical Therapy for Diabetes — 5-Year Outcomes. N. Engl. J. Med. 2017;376(7):641–651.
  162. Fisher DP, Johnson E, Haneuse S, Arterburn D, Coleman KJ, O’Connor PJ, O’Brien R, Bogart A, Theis MK, Anau J, Schroeder EB, Sidney S. Association between Bariatric Surgery and Macrovascular Disease Outcomes in Patients with Type 2 Diabetes and Severe Obesity. JAMA - J. Am. Med. Assoc. 2018;320(15):1570–1582.
  163. Sundström J, Bruze G, Ottosson J, Marcus C, Näslund I, Neovius M. Weight Loss and Heart FailureClinical Perspective. Circulation 2017;135(17):1577–1585.
  164. Aminian A, Zajichek A, Arterburn DE, Wolski KE, Brethauer SA, Schauer PR, Kattan MW, Nissen SE.Association of Metabolic Surgery with Major Adverse Cardiovascular Outcomes in Patients with Type 2 Diabetes and Obesity. JAMA - J. Am. Med. Assoc. 2019;322(13):1271–1282.
  165. Stratton IM, Adler AI, Neil HAW, Matthews DR, Manley SE, Cull CA, Hadden D, Turner RC, Holman RR.Association of glycaemia with macrovascular and microvascular complications of type 2 diabetes (UKPDS 35): Prospective observational study. Br. Med. J. 2000;321(7258):405–412.
  166. Gerstein HC, Miller ME, Byington RP, Goff DC, Bigger JT. Effects of intensive glucose lowering in type 2 diabetes. N. Engl. J. Med. 2008;358(24):2545–2559.
  167. Gerstein HC, Miller ME, Genuth S, Ismail-Beigi F, Buse JB, Goff DC, Probstfield JL, Cushman WC, Ginsberg HN, Thomas Bigger J, Grimm RH, Byington RP, Rosenberg YD, Friedewald WT. Long-term effects of intensive glucose lowering on cardiovascular outcomes. N. Engl. J. Med. 2011;364(9):818–828.
  168. Patel A, MacMahon S, Chalmers J, Neal B, Billot L, Woodward M, Marre M, Cooper M, Glasziou P, Grobbee D, Hamet P, Harrap S, Heller S, Liu L, Mancia G, Mogensen CE, Pan C, Poulter N, Rodgers A, Williams B, Bompoint S, De Galan BE, Joshi R, Travert F. Intensive blood glucose control and vascular outcomes in patients with type 2 diabetes. N. Engl. J. Med. 2008;358(24):2560–2572.
  169. Duckworth W, Abraira C, Moritz T, Reda D, Emanuele N, Reaven PD, Zieve FJ, Marks J, Davis SN, Hayward R, Warren SR, Goldman S, McCarren M, Vitek ME, Henderson WG, Huang GD. Glucose Control and Vascular Complications in Veterans with Type 2 Diabetes. N. Engl. J. Med. 2009;360(2):129–139.
  170. Turnbull FM, Abraira C, Anderson RJ, Byington RP, Chalmers JP, Duckworth WC, Evans GW, Gerstein HC, Holman RR, Moritz TE, Neal BC, Ninomiya T, Patel AA, Paul SK, Travert F, Woodward M. Intensive glucose control and macrovascular outcomes in type 2 diabetes. Diabetologia 2009;52(11):2288–2298.
  171. Colucci WS. Overview of the management of heart failure with reduced ejection fraction in adults. In: UpToDate.; 2020.
  172. Ponikowski P, Voors AAA, Anker SDD, Bueno H, Cleland JGFGF, Coats AJSJS, Falk V, González-Juanatey JRR, Harjola VPP, Jankowska EAA, Jessup M, Linde C, Nihoyannopoulos P, Parissis JTT, Pieske B, Riley JPP, Rosano GMCMC, Ruilope LMM, Ruschitzka F, Rutten FHH, Van Der Meer P, Sisakian HSS, Isayev E, Kurlianskaya A, Mullens W, Tokmakova M, Agathangelou P, Melenovsky V, Wiggers H, Hassanein M, Uuetoa T, Lommi J, Kostovska ESS, Juilliere Y, Aladashvili A, Luchner A, Chrysohoou C, Nyolczas N, Thorgeirsson G, Weinstein JMM, Lenarda AD Di, Aidargaliyeva N, Bajraktari G, Beishenkulov M, Kamzola G, Abdel-Massih T, Celutkiene J, Noppe S, Cassar A, Vataman E, AbirKhalil S, van Pol P, Mo R, Straburzynska-Migaj E, Fonseca C, Chioncel O, Shlyakhto E, Zavatta M, Otasevic P, Goncalvesova E, Lainscak M, Molina BDD, Schaufelberger M, Suter T, Yılmaz MBB, Voronkov L, Davies C. 2016 ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure. Eur. Heart J. 2016;37(27):2129-2200m.
  173. van der Meer P, Gaggin HK, Dec GW. ACC/AHA Versus ESC Guidelines on Heart Failure: JACC Guideline Comparison. J. Am. Coll. Cardiol. 2019;73(21):2756–2768.
  174. Shekelle PG, Rich MW, Morton SC, Atkinson SW, Tu W, Maglione M, Rhodes S, Barrett M, Fonarow GC, Greenberg B, Heidenreich PA, Knabel T, Konstam MA, Steimle A, Stevenson LW. Efficacy of angiotensin-converting enzyme inhibitors and beta-blockers in the management of left ventricular systolic dysfunction according to race, gender, and diabetic status: A meta-analysis of major clinical trials. J. Am. Coll. Cardiol.2003;41(9):1529–1538.
  175. Khan MS, Felker GM, Piña IL, Camacho A, Bapat D, Ibrahim NE, Maisel AS, Prescott MF, Ward JH, Solomon SD, Januzzi JL, Butler J. Reverse Cardiac Remodeling Following Initiation of Sacubitril/Valsartan in Patients With Heart Failure With and Without Diabetes. JACC Hear. Fail. 2021;9(2):137–145.
  176. Vermes E, Ducharme A, Bourassa MG, Lessard M, White M, Tardif JC. Enalapril reduces the incidence of diabetes in patients with chronic heart failure: Insight from the studies of left ventricular dysfunction (SOLVD). Circulation 2003;107(9):1291–1296.
  177. Seferovic JP, Claggett B, Seidelmann SB, Seely EW, Packer M, Zile MR, Rouleau JL, Swedberg K, Lefkowitz M, Shi VC, Desai AS, McMurray JJV, Solomon SD. Effect of sacubitril/valsartan versus enalapril on glycaemic control in patients with heart failure and diabetes: a post-hoc analysis from the PARADIGM-HF trial. Lancet Diabetes Endocrinol. 2017;5(5):333–340.
  178. Seferović PM, Petrie MC, Filippatos GS, Anker SD, Rosano G, Bauersachs J, Paulus WJ, Komajda M, Cosentino F, de Boer RA, Farmakis D, Doehner W, Lambrinou E, Lopatin Y, Piepoli MF, Theodorakis MJ, Wiggers H, Lekakis J, Mebazaa A, Mamas MA, Tschöpe C, Hoes AW, Seferović JP, Logue J, McDonagh T, Riley JP, Milinković I, Polovina M, van Veldhuisen DJ, Lainscak M, Maggioni AP, Ruschitzka F, McMurray JJV. Type 2 diabetes mellitus and heart failure: a position statement from the Heart Failure Association of the European Society of Cardiology. Eur. J. Heart Fail. 2018;20(5):853–872.
  179. Bell DSH, Lukas MA, Holdbrook FK, Fowler MB. The effect of carvedilol on mortality risk in heart failure patients with diabetes: Results of a meta-analysis. Curr. Med. Res. Opin. 2006;22(2):287–296.
  180. Haas SJ, Vos T, Gilbert RE, Krum H. Are β-blockers as efficacious in patients with diabetes mellitus as in patients without diabetes mellitus who have chronic heart failure? A meta-analysis of large-scale clinical trials. Am. Heart J. 2003;146(5):848–853.
  181. Witte KK, Drozd M, Walker AMN, Patel PA, Kearney JC, Chapman S, Sapsford RJ, Gierula J, Paton MF, Lowry J, Kearney MT, Cubbon RM. Mortality reduction associated with β-adrenoceptor inhibition in chronic heart failure is greater in patients with diabetes. Diabetes Care 2018;41(1):136–142.
  182. Barzilay JI, Davis BR, Whelton PK. The glycemic effects of antihypertensive medications. Curr. Hypertens. Rep.2014;16(1):1–6.
  183. Helgeland A, Leren P, Foss OP, Hjermann I, Holme I, Lund-Larsen PG. Serum glucose levels during long-term observation of treated and untreated men with mild hypertension. The Oslo study. Am. J. Med. 1984;76(5):802–805.
  184. Wai B, Kearney LG, Hare DL, Ord M, Burrell LM, Srivastava PM. Beta blocker use in subjects with type 2 diabetes mellitus and systolic heart failure does not worsen glycaemic control. Cardiovasc. Diabetol.2012;11(14):1–5.
  185. Tsujimoto T, Sugiyama T, Shapiro MF, Noda M, Kajio H. Risk of Cardiovascular Events in Patients with Diabetes Mellitus on β-Blockers. Hypertension 2017;70(1):103–110.
  186. Deedwania PC, Giles TD, Klibaner M, Ghali JK, Herlitz J, Hildebrandt P, Kjekshus J, Spinar J, Vitovec J, Stanbrook H, Wikstrand J. Efficacy, safety and tolerability of metoprolol CR/XL in patients with diabetes and chronic heart failure: Experiences from MERIT-HF. Am. Heart J. 2005;149(1):159–167.
  187. Eschalier R, McMurray JJV, Swedberg K, Van Veldhuisen DJ, Krum H, Pocock SJ, Shi H, Vincent J, Rossignol P, Zannad F, Pitt B. Safety and efficacy of eplerenone in patients at high risk for hyperkalemia and/or worsening renal function: Analyses of the EMPHASIS-HF study subgroups (eplerenone in mild patients hospitalization and survival study in heart failure). J. Am. Coll. Cardiol. 2013;62(17):1585–1593.
  188. Korol S, White M, O’Meara E, Rouleau JL, White-Guay B, Dorais M, Ahmed A, de Denus S, Perreault S. Is there a potential association between spironolactone and the risk of new-onset diabetes in a cohort of older patients with heart failure? Eur. J. Clin. Pharmacol. 2019;75(6):837–847.
  189. McMurray JJV, Solomon SD, Inzucchi SE, Køber L, Kosiborod MN, Martinez FA, Ponikowski P, Sabatine MS, Anand IS, Bělohlávek J, Böhm M, Chiang C-E, Chopra VK, de Boer RA, Desai AS, Diez M, Drozdz J, Dukát A, Ge J, Howlett JG, Katova T, Kitakaze M, Ljungman CEA, Merkely B, Nicolau JC, O’Meara E, Petrie MC, Vinh PN, Schou M, Tereshchenko S, Verma S, Held C, DeMets DL, Docherty KF, Jhund PS, Bengtsson O, Sjöstrand M, Langkilde A-M. Dapagliflozin in Patients with Heart Failure and Reduced Ejection Fraction. N. Engl. J. Med.2019;381(21):1995–2008.
  190. Packer M, Anker SD, Butler J, Filippatos G, Pocock SJ, Carson P, Januzzi J, Verma S, Tsutsui H, Brueckmann M, Jamal W, Kimura K, Schnee J, Zeller C, Cotton D, Bocchi E, Böhm M, Choi D-J, Chopra V, Chuquiure E, Giannetti N, Janssens S, Zhang J, Gonzalez Juanatey JR, Kaul S, Brunner-La Rocca H-P, Merkely B, Nicholls SJ, Perrone S, Pina I, Ponikowski P, Sattar N, Senni M, Seronde M-F, Spinar J, Squire I, Taddei S, Wanner C, Zannad F. Cardiovascular and Renal Outcomes with Empagliflozin in Heart Failure. N. Engl. J. Med.2020;383(15):1413–1424.
  191. Braunwald E. Gliflozins in the Management of Cardiovascular Disease. N. Engl. J. Med. 2022;386(21):2024–2034.
  192. Packer M, Anker SD, Butler J, Filippatos G, Ferreira JP, Pocock SJ, Carson P, Anand I, Doehner W, Haass M, Komajda M, Miller A, Pehrson S, Teerlink JR, Brueckmann M, Jamal W, Zeller C, Schnaidt S, Zannad F. Effect of Empagliflozin on the Clinical Stability of Patients with Heart Failure and a Reduced Ejection Fraction: The EMPEROR-Reduced Trial. Circulation 2021;143(4):326–336.
  193. Khan MS, Solomon N, DeVore AD, Sharma A, Felker GM, Hernandez AF, Heidenreich PA, Matsouaka RA, Green JB, Butler J, Yancy CW, Peterson PN, Fonarow GC, Greene SJ. Clinical Outcomes With Metformin and Sulfonylurea Therapies Among Patients With Heart Failure and Diabetes. JACC Hear. Fail. 2022;10(3):198–210.
  194. Anker SD, Butler J, Filippatos G, Ferreira JP, Bocchi E, Böhm M, Brunner–La Rocca H-P, Choi D-J, Chopra V, Chuquiure-Valenzuela E, Giannetti N, Gomez-Mesa JE, Janssens S, Januzzi JL, Gonzalez-Juanatey JR, Merkely B, Nicholls SJ, Perrone S V., Piña IL, Ponikowski P, Senni M, Sim D, Spinar J, Squire I, Taddei S, Tsutsui H, Verma S, Vinereanu D, Zhang J, Carson P, Lam CSP, Marx N, Zeller C, Sattar N, Jamal W, Schnaidt S, Schnee JM, Brueckmann M, Pocock SJ, Zannad F, Packer M. Empagliflozin in Heart Failure with a Preserved Ejection Fraction. N. Engl. J. Med. 2021;385(16):1451–1461.
  195. Bhatt DL, Szarek M, Steg PG, Cannon CP, Leiter LA, McGuire DK, Lewis JB, Riddle MC, Voors AA, Metra M, Lund LH, Komajda M, Testani JM, Wilcox CS, Ponikowski P, Lopes RD, Verma S, Lapuerta P, Pitt B.Sotagliflozin in Patients with Diabetes and Recent Worsening Heart Failure. N. Engl. J. Med. 2021;384(2):117–128.
  196. Bardy GH, Lee KL, Mark DB, Poole JE, Packer DL, Boineau R, Domanski M, Troutman C, Anderson J, Johnson G, McNulty SE, Clapp-Channing N, Davidson-Ray LD, Fraulo ES, Fishbein DP, Luceri RM, Ip JH. Amiodarone or an implantable cardioverter-defibrillator for congestive heart failure. N. Engl. J. Med. 2005;352(3):225–237.
  197. Wittenberg SM, Cook JR, Hall WJ, McNitt S, Zareba W, Moss AJ. Comparison of efficacy of implanted cardioverter-defibrillator in patients with versus without diabetes mellitus. Am. J. Cardiol. 2005;96(3):417–419.
  198. Tan ESJ, Lim J, Chan SP, Seow JTK, Singh D, Yeo WT, Lim TW, Kojodjojo P, Seow SC. Effect of Diabetes Mellitus on Cardiac Resynchronization Therapy and to Prognosis in Heart Failure (from the Prospective Evaluation of Asian With Cardiac Resynchronization Therapy for Heart Failure Study). Am. J. Cardiol.2019;124(6):899–906.
  199. Ghali J k., Boehmer J, Feldman AM, Saxon LA, Demarco T, Carson P, Yong P, Galle EG, Leigh J, Ecklund FL, Bristow MR. Influence of Diabetes on Cardiac Resynchronization Therapy With or Without Defibrillator in Patients With Advanced Heart Failure. J. Card. Fail. 2007;13(9):769–773.
  200. Hoppe UC, Freemantle N, Cleland JGF, Marijianowski M, Erdmann E. Effect of cardiac resynchronization on morbidity and mortality of diabetic patients with severe heart failure. Diabetes Care 2007;30(3):722–724.
  201. Sun H, Guan Y, Wang L, Zhao Y, Lv H, Bi X, Wang H, Zhang X, Liu L, Wei M, Song H, Su G. Influence of diabetes on cardiac resynchronization therapy in heart failure patients: A meta-analysis. BMC Cardiovasc. Disord. 2015;15(1). doi:10.1186/s12872-015-0018-0.
  202. Höke U, Thijssen J, Van Bommel RJ, Van Erven L, Van Der Velde ET, Holman ER, Schalij MJ, Bax JJ, Delgado V, Marsan NA. Influence of diabetes on left ventricular systolic and diastolic function and on long-term outcome after cardiac resynchronization therapy. Diabetes Care 2013;36(4):985–991.
  203. Kahr PC, Trenson S, Schindler M, Kuster J, Kaufmann P, Tonko J, Hofer D, Inderbitzin DT, Breitenstein A, Saguner AM, Flammer AJ, Ruschitzka F, Steffel J, Winnik S. Differential effect of cardiac resynchronization therapy in patients with diabetes mellitus: a long-term retrospective cohort study. ESC Hear. Fail.2020;7(5):2773–2783.
  204. Massie BM, Carson PE, McMurray JJ, Komajda M, McKelvie R, Zile MR, Anderson S, Donovan M, Iverson E, Staiger C, Ptaszynska A. Irbesartan in patients with heart failure and preserved ejection fraction. N. Engl. J. Med. 2008;359(23):2456–2467.
  205. Nassif ME, Windsor SL, Borlaug BA, Kitzman DW, Shah SJ, Tang F, Khariton Y, Malik AO, Khumri T, Umpierrez G, Lamba S, Sharma K, Khan SS, Chandra L, Gordon RA, Ryan JJ, Chaudhry SP, Joseph SM, Chow CH, Kanwar MK, Pursley M, Siraj ES, Lewis GD, Clemson BS, Fong M, Kosiborod MN. The SGLT2 inhibitor dapagliflozin in heart failure with preserved ejection fraction: a multicenter randomized trial. Nat. Med.2021;27(11):1954–1960.
  206. Zinman B, Wanner C, Lachin JM, Fitchett D, Bluhmki E, Hantel S. Empagliflozin, cardiovascular outcomes, and mortality in type 2 diabetes. N. Engl. J. Med. 2015;373(22):2117–2128.
  207. Neal B, Perkovic V, Mahaffey KW, De Zeeuw D, Fulcher G, Erondu N, Shaw W, Law G, Desai M, Matthews DR.Canagliflozin and cardiovascular and renal events in type 2 diabetes. N. Engl. J. Med. 2017;377(7):644–657.
  208. Wiviott SD, Raz I, Bonaca MP, Mosenzon O, Kato ET, Cahn A, Silverman MG, Zelniker TA, Kuder JF, Murphy SA, Bhatt DL, Leiter LA, McGuire DK. Dapagliflozin and cardiovascular outcomes in type 2 diabetes. N. Engl. J. Med. 2019;380(4):347–357.
  209. Perkovic V, Jardine MJ, Neal B, Bompoint S, Heerspink HJL, Charytan DM, Edwards R, Agarwal R, Bakris G, Bull S, Cannon CP, Capuano G, Chu PL, De Zeeuw D, Greene T, Levin A, Pollock C, Wheeler DC, Yavin Y, Zhang H, Zinman B, Meininger G, Brenner BM, Mahaffey KW. Canagliflozin and renal outcomes in type 2 diabetes and nephropathy. N. Engl. J. Med. 2019;380(24):2295–2306.
  210. Cannon CP, Pratley R, Dagogo-Jack S, Mancuso J, Huyck S, Masiukiewicz U, Charbonnel B, Frederich R, Gallo S, Cosentino F, Shih WJ, Gantz I, Terra SG, Cherney DZI, McGuire DK. Cardiovascular Outcomes with Ertugliflozin in Type 2 Diabetes. N. Engl. J. Med. 2020;383(15):1425–1435.
  211. Akturk HK, Rewers A, Garg SK. SGLT inhibition: A possible adjunctive treatment for type 1 diabetes. Curr. Opin. Endocrinol. Diabetes Obes. 2018;25(4):246–250.
  212. Empagliflozin in Patients with Chronic Kidney Disease.N. Engl. J. Med. 2023;388(2):117–127.
  213. Heerspink HJL, Stefánsson B V., Correa-Rotter R, Chertow GM, Greene T, Hou F-F, Mann JFE, McMurray JJV, Lindberg M, Rossing P, Sjöström CD, Toto RD, Langkilde A-M, Wheeler DC. Dapagliflozin in Patients with Chronic Kidney Disease. N. Engl. J. Med. 2020;383(15):1436–1446.
  214. Bhatt DL, Szarek M, Pitt B, Cannon CP, Leiter LA, McGuire DK, Lewis JB, Riddle MC, Inzucchi SE, Kosiborod MN, Cherney DZI, Dwyer JP, Scirica BM, Bailey CJ, Díaz R, Ray KK, Udell JA, Lopes RD, Lapuerta P, Steg PG.Sotagliflozin in Patients with Diabetes and Chronic Kidney Disease. N. Engl. J. Med. 2021;384(2):129–139.
  215. Marso SP, Daniels GH, Frandsen KB, Kristensen P, Mann JFE, Nauck MA, Nissen SE, Pocock S, Poulter NR, Ravn LS, Steinberg WM, Stockner M, Zinman B, Bergenstal RM, Buse JB. Liraglutide and cardiovascular outcomes in type 2 diabetes. N. Engl. J. Med. 2016;375(4):311–322.
  216. Gerstein HC, Colhoun HM, Dagenais GR, Diaz R, Lakshmanan M, Pais P. Dulaglutide and cardiovascular outcomes in type 2 diabetes (REWIND): a double-blind, randomised placebo-controlled trial. Lancet2019;394(10193):121–130.
  217. Gerstein HC, Sattar N, Rosenstock J, Ramasundarahettige C, Pratley R, Lopes RD, Lam CSP, Khurmi NS, Heenan L, Del Prato S, Dyal L, Branch K. Cardiovascular and Renal Outcomes with Efpeglenatide in Type 2 Diabetes. N. Engl. J. Med. 2021;385(10):896–907.
  218. Ferreira JP, Sharma A, Butler J, Packer M, Zannad F, Vasques-nóvoa F, Leite-moreira A, Neves JS. Glucagon-Like Peptide-1 Receptor Agonists Across the Spectrum of Heart Failure. 2023;(July):1–6.
  219. Ferreira JP, Saraiva F, Sharma A, Vasques-Nóvoa F, Angélico-Gonçalves A, Leite AR, Borges-Canha M, Carvalho D, Packer M, Zannad F, Leite-Moreira A, Neves JS. Glucagon-like peptide 1 receptor agonists in patients with type 2 diabetes with and without chronic heart failure: A meta-analysis of randomized placebo-controlled outcome trials. Diabetes, Obes. Metab. 2023;25(6):1495–1502.
  220. Jastreboff AM, Aronne LJ, Ahmad NN, Wharton S, Connery L, Alves B, Kiyosue A, Zhang S, Liu B, Bunck MC, Stefanski A. Tirzepatide Once Weekly for the Treatment of Obesity. N. Engl. J. Med. 2022;387(3):205–216.
  221. Kosiborod MN, Abildstrøm SZ, Borlaug BA, Butler J, Rasmussen S, Davies M, Hovingh GK, Kitzman DW, Lindegaard ML, Møller D V, Shah SJ, Treppendahl MB, Verma S, Abhayaratna W, Ahmed FZ, Chopra V, Ezekowitz J, Fu M, Ito H, Lelonek M, Melenovsky V, Merkely B, Núñez J, Perna E, Schou M, Senni M, Sharma K, Van der Meer P, von Lewinski D, Wolf D, Petrie MC, STEP-HFpEF Trial Committees and Investigators.Semaglutide in Patients with Heart Failure with Preserved Ejection Fraction and Obesity. N. Engl. J. Med.2023:1–16.
  222. Kenny HC, Abel ED. Heart Failure in Type 2 Diabetes Mellitus: Impact of Glucose-Lowering Agents, Heart Failure Therapies, and Novel Therapeutic Strategies. Circ. Res. 2019;124(1):121–141.
  223. Eurich DT, Weir DL, Majumdar SR, Tsuyuki RT, Johnson JA, Tjosvold L, Vanderloo SE, McAlister FA.Comparative safety and effectiveness of metformin in patients with diabetes mellitus and heart failure: systematic review of observational studies involving 34,000 patients. Circ. Heart Fail. 2013;6(3):395–402.
  224. Scirica BM, Braunwald E, Raz I, Cavender MA, Morrow DA, Jarolim P, Udell JA, Mosenzon O, Im K, Umez-Eronini AA, Pollack PS, Hirshberg B, Frederich R, Lewis BS, McGuire DK, Davidson J, Steg PG, Bhatt DL.Heart failure, saxagliptin, and diabetes mellitus: Observations from the SAVOR-TIMI 53 randomized trial. Circulation 2014;130(18):1579–1588.
  225. Zannad F, Cannon CP, Cushman WC, Bakris GL, Menon V, Perez AT, Fleck PR, Mehta CR, Kupfer S, Wilson C, Lam H, White WB. Heart failure and mortality outcomes in patients with type 2 diabetes taking alogliptin versus placebo in EXAMINE: A multicentre, randomised, double-blind trial. Lancet 2015;385(9982):2067–2076.
  226. White WB, Cannon CP, Heller SR, Nissen SE, Bergenstal RM, Bakris GL, Perez AT. Alogliptin after acute coronary syndrome in patients with type 2 diabetes. N. Engl. J. Med. 2013;369(14):1327–1335.
  227. McGuire DK, Van De Werf F, Armstrong PW, Standl E, Koglin J, Green JB, Bethel MA, Cornel JH, Lopes RD, Halvorsen S, Ambrosio G, Buse JB, Josse RG, Lachin JM, Pencina MJ, Garg J, Lokhnygina Y, Holman RR, Peterson ED. Association between sitagliptin use and heart failure hospitalization and related outcomes in type 2 diabetes mellitus: Secondary analysis of a randomized clinical trial. JAMA Cardiol. 2016;1(2):126–135.
  228. Hippisley-Cox J, Coupland C. Diabetes treatments and risk of heart failure, cardiovascular disease, and all cause mortality: Cohort study in primary care. BMJ 2016;354. doi:10.1136/bmj.i3477.
  229. Gerstein HC, Bosch J, Dagenais GR, Díaz R, Jung H, Maggioni AP. Basal insulin and cardiovascular and other outcomes in dysglycemia. N. Engl. J. Med. 2012;367(4):319–328.
  230. Home PD, Pocock SJ, Beck-Nielsen H, Curtis PS, Gomis R, Hanefeld M, Jones NP, Komajda M, McMurray JJ.Rosiglitazone evaluated for cardiovascular outcomes in oral agent combination therapy for type 2 diabetes (RECORD): a multicentre, randomised, open-label trial. Lancet 2009;373(9681):2125–2135.
  231. Dormandy JA, Charbonnel B, Eckland DJA, Erdmann E, Massi-Benedetti M, Moules IK, Skene AM, Tan MH, Lefèbvre PJ, Murray GD, Standl E, Wilcox RG, Wilhelmsen L, Betteridge J, Birkeland K, Golay A, Heine RJ, Korányi L, Laakso M, Mokáň M, Norkus A, Pirags V, Podar T, Scheen A, Scherbaum W, Schernthaner G, Schmitz O, Škrha J, Smith U, Tatoň J. Secondary prevention of macrovascular events in patients with type 2 diabetes in the PROactive Study (PROspective pioglitAzone Clinical Trial in macroVascular Events): A randomised controlled trial. Lancet 2005;366(9493):1279–1289.

 

 

 

 

Prevention of Obesity

ABSTRACT

 

Obesity has become a major public problem which is associated with increased risk to health and enhanced mortality along with increased medical costs.  Prevention is obviously the first line of attack and this chapter outlines preventive strategies starting with a model which translates the energy imbalance which produces obesity into the social framework in which this occurs and where intervention must occur. Increased food intake is the major driver with reduced physical activity as a second component, modified by many other factors. Prevention begins with pregnant woman where maintaining a healthy weight gain improves the outcome for both infant and mother with diet and exercise both showing positive results. The early years of life are another important time for prevention. Studies of children have shown that exercise and nutritional quality can be improved by lifestyle intervention, but that impacting body weight change is more difficult. Reducing sugar-sweetened beverage intake and increasing water intake are two potentially useful strategies. Finally, there are many studies examining strategies for prevention of weight gain in adults as a group and in special subsets of adults with variable effect. One can conclude that prevention is the cornerstone for reducing the prevalence of obesity in pregnant women and their offspring, in children and adolescents, and in adults, but that the current strategies may need to be supplemented with additional successful modalities of implementation.

 

INTRODUCTION

 

Obesity is a worldwide problem (1) and affect more than 100 million Americans (2). In the 1980s the prevalence of obesity began to rise more rapidly than before and has now reached epidemic proportions worldwide.  Since 1980 it has more than doubled and In the United States. Data provided by the US National Health and Nutrition Examination Survey (NHANES) in 2009-2010 showed that 35.5 % (95% CI, 31.9%─39.2%) of men were obese (BMI>30 kg/m2) and 35.8% (95% CI, 34.0%─37.7%) of adult women were obese (1, 2). The National Health and Nutrition Examination Surveys (NHANES) in 2014 noted a BMI of ≥25 was present in 71.3% of men 20 years or older and in 65.8% of women 20 years or older. The prevalence of obesity (BMI ≥30) was 33.5% for men and 36.1% for women. Females at any age are disproportionately at greater risk for obesity, and especially extreme obesity (BMI ≥40, 8.3% in females; 4.4% in males). The prevalence of obesity has risen dramatically since 1980, but may have slowed at current higher than desirable levels. The prevalence of obesity among children age 3-5 is alarmingly high at 12.4% in boys and 10% in girls with higher rates in Hispanic and Blacks (3). The increase has continued in extreme obesity in the United States and is rising worldwide. This increase in the prevalence of obesity carries with it increased risks for diabetes, metabolic syndrome, non-alcoholic fatty liver, heart disease and cancer among others (4).   It also has significant costs to the individual and to society (5).  Clearly the “brakes” that prevented a rapid increase in obesity before 1980 are not working well enough, and new preventive strategies are needed. To select papers for this chapter the words prevent, prevention, obesity, and overweight were screened in PubMed and additional references identified from the selected papers.

 

FRAMEWORK FOR DEVELOPMENT OF OBESITY         

 

Figure 1 is a model with the “energy balance” equation at the center and social and environmental factors surrounding it (4).  When an individual becomes obese it is a clear sign that the balance has tipped slightly towards positive energy intake (or reduced expenditure) and that this imbalance has been present for months to years. Primary prevention of obesity would occur if strategies similar to what were in place before 1970-1980 were re-introduced and obesity rates were reversed, or alternatively if equally effective new ones were implemented.  Secondary prevention is the use of techniques to prevent regain of weight in an individual who has gained too much weight as fat and then lost it. These are often also called “maintenance” strategies.

 

Figure 1. Model of Energy Intake and Energy Expenditure on a Balance Influenced by Genetic and Epigenetic Factors and Environmental and Social Influences.

 

The development of obesity, that is moving the central pendulum in Fig 1 to the right in favor of a positive energy balance could occur because there is a small increase in energy expenditure, a small decrease in energy expenditure or a combination of both. 

 

Food available for consumption and food intake, corrected for plate waste, began to increase in the US after 1970 and has continued to the present (6, 7). The extra amount of food is now estimated to be about 400 kcal/d which is enough to account for the calories needed to produce the documented weight gain (8). An alternative explanation would be a decrease in activity in the ordinary duties of the day (9). I tend to favor the rise in food intake as the major factor. 

 

The rise in consumable food items from farms in the United States began after a change in food policy for reimbursing farmers in 1970 (10). There are several outlets for this growing surplus of food. First, it could be stored in warehouses against a future need. It could be destroyed. Or it could be transferred to storage depots on people – obesity (11).   Since the growth of the population is less than 1-2% per year, the growth in food supplies was larger than needed to meet this growing population.  Since we don’t see stores of food in warehouses, we have probably transferred it to the population producing obesity.  As Tillotson has said “…certain of our agricultural and industrial policies have had the unintended and unforeseen consequences of increasing overweight and obesity”. Our current “preventive strategies” thus operate in a setting of agricultural and industrial policies that favor production of more food that is needed to feed the population. Surpluses of food operate in a socio-economic environment. In many groups of women this is reflected in an inverse relationship between the prevalence of obesity and the educational and income levels (10). 

 

Several economic hypotheses related to “food security” have been proposed. These economic factors may need to be part of the preventive strategy if we are to develop a cohesive approach to dealing with obesity around the world.

 

The positive energy balance that we see leading to obesity can be influenced by a variety of factors which are controlled by the individual, by the family, and by society. We know that people can “consciously” control their weight by restrained eating. It is clear from the model that the individual is at the center of this system but their responses in terms of energy intake and energy expenditure are influenced by a myriad of factors over which they have little or no control. Genetics underpins obesity (11, 12, 13). There are some genes that have such potent effects on body weight that when defective obesity is almost certain. Leptin is one of the most potent (11). There are more than 90 other genes that contribute small amounts to differences in body weight accounting for less than 5% of the risk for obesity (11, 13).  

 

Epigenetic factors, environmental influences on the fetus and early life also have important effects. For example, offspring of mothers who smoke during pregnancy or whose mother is a diabetic are at higher risk of obesity later in life and those who gain large amounts of weight (4).  Duration of sleep time also affects body weight. Availability of food and its palatability are environmental factors that have significant effects that can override the controls systems for body weight. It is clear that although many of these environmental factors can be controlled by the individual, the genetic and epigenetic influences make it difficult for many people to maintain a stable body weight. Thus, preventive and therapeutic strategies are needed.

 

STRATEGIES FOR PREVENTING OBESITY

 

At least 4 preventive strategies are available to deal with the epidemic: Education, regulation, modification of the food supply, and changes in the cost of food energy.  Education about good nutrition and healthy weight in the school curriculum would be beneficial in helping all children learn how to select appropriate foods and should be included in school curricula. Foods used in school breakfast and lunch programs should match these educational messages.  

 

However, it is unwise to rely on educational strategies alone, since they have not, so far, prevented the epidemic of obesity. The program of knowing your BMI initially instituted on a state wide basis in Arkansas may have helped that State to reduce the upward trend in obesity.  Regulation is a second strategy. Regulations to provide an improved food label would be one good idea. Better regulations on appropriate serving sizes and caloric value that would be easy for the public to use might be part of the information provided by restaurants on their menus.  This is now required in New York City and if effective may spread around the country.

 

Modification in some components of the food system is a third and very important strategy.  Since the energy we eat comes from the food we eat, we need to modify this system to provide smaller portions and less energy density. One approach is to use differentiated food taxes to promote healthy diets. This is the approach New York State is trying with its tax on soft drinks introduced in 2007, and has been urged by a group of prominent nutritional scientists (15). This strategy has been argued both at the academic level (16, 17, 18), and at the policy level. It may be that economic tools that will shift food choices using cost is the “fluoride” for treating the epidemic of overweight that is described below.

 

Some years ago, I proposed that the best strategy for prevention of obesity may be modeled after the use of fluoride for prevention of dental caries (19).  The addition of fluoride to the water supply had a more profound effect on the incidence of dental caries than brushing and flossing teeth. Brushing and flossing are like diet and exercise. They both require commitment on the part of the individual. Adding something like fluoride to the water supply doesn't require any commitment. Increasing the price of oil may be one such strategy. Between 1940 and 2005, Pollan pointed out that the number of calories from petroleum (oil) used to produce food energy has risen dramatically. In 1940 it was 0.4 cal of oil for each calorie of food energy. In 2005 it had risen over 20-fold with 10 calories of oil needed for each calorie of food. These calories come from the petroleum products used to make fertilizers, to transport food, to process and package food, for pesticides, and so on.  If oil prices rise significantly, this will shift the use of oil for food and shift our consumption patterns (20).

 

Strategies for Preventing Obesity in Pregnant Women

 

Women who gain more weight during pregnancy have increased risk of diabetes, hypertension, pre-eclampsia, and still birth. The offspring is at increased risk for macrosomia and later obesity (21).  A lifestyle intervention program during pregnancy in Denmark (Lifestyle in Pregnancy = LiP), however, failed to alter the metabolic risk factors in the offspring.  A systematic review of evidence relating weight gain during pregnancy and outcomes of pregnancy found that dietary interventions were the most effective type of intervention in pregnancy. They reduced gestational weight gain and the risks of pre-eclampsia, hypertension, and shoulder dystocia in the infant. There was no difference in the incidence of small-for-gestational-age infants as a result of treatments (22, 23). Another systematic review reached similar conclusions and showed that dietary interventions significantly reduced gestational weight gain by 1.92 kg (95% CI -3,65, -0,19), and the incidence of Caesarean section (24). The Cochrane review of this subject in 2015 found that diet, exercise or both reduced the gestational weight gain by an average of 20%. These interventions included low glycemic index diets, supervised or unsupervised exercise program, and diet combined with exercise which were all comparable in their effects. Hypertension was reduced but pre-eclampsia was not. They also found no differences between intervention and control groups in the risk of preterm births, or macrosomia overall. However, the subgroup of women who were overweight or obese did have a 15% reduction in macrosomia. Fetal distress syndrome of was also reduced in women with obesity in the intervention groups (25).

           

Strategies Aimed at Children

 

There is a great deal of concern for the plight of obese children. The pioneering work of the psychoanalyst, Dr. Hilde Bruch, a refugee from Nazi Germany working in the 1940’s did much to alert the public to this important issue (26).

 

Children of overweight parents are a high-risk group for development of overweight (27). In a long-term follow-up study, Berkowitz et al studied 32 high risk children whose maternal pre-pregnancy BMI was 30.4 kg/m2 and compared them to 29 low risk children whose maternal BMI was low at 19.6 kg/m2.  At this age they consumed a test meal in which their eating behavior was assessed, including rate of caloric consumption, mouthfuls/min, and requests for food. Parental prompts for the child to eat also were measured.  Parental feeding prompts were not different between high risk and low risk children, but the rate of eating measured by mouthfuls of food/min, and total caloric intake/min during the test meal predicted an increased risk of being overweight or obese at age 6. Thus, pre-school years are important in setting risks for future obesity

 

Schools have also changed. They were once a place where children could be very active. With security issues and concerns about safety when children walking home from school there is less opportunity for physical exercise. Providing safe pedestrian walk-ways to school could increase physical activity for children more easily than changing the built environment for adults. In a review of school-based programs, 18 studies involving 18,141 children were evaluated. They were primarily elementary school children and had programs that lasted from 6 months to 3 years.  A meta-analysis showed that physical activity interventions did not improve BMI (28).

 

To examine the studies that have looked at prevention of childhood obesity, I have taken data from a Cochrane Collaboration review of preventive strategies for children divided studies into long and short term.  Short-term studies were those with data for 12 weeks or more but less than 52 weeks. Long-term studies were those with data beyond 52 weeks.  I will discuss only the long-term studies.

 

LONG-TERM STUDIES

 

One controlled trial that was deemed of good quality was conducted in the US randomized 26 children and their families to 2 conditions: 1) increasing fruit and vegetable and 2) decreasing fat and sugar (29).  The children were 6-11 years old and at least one parent accompanied them. They received a comprehensive behavioral program.  At the end of 12 months the decrease in percentage of overweight was decreased 1.10% in the fruit and vegetable group and 2.40% in the lower fat and sugar group. These differences were not statistically significant, but are nonetheless tantalizing and suggest the need for applying this to high-risk groups like the children of overweight parents.

 

A second study of good quality was conducted by James et al (30) where 644 children were randomized by school class into 15 intervention and 14 control classes in 6 schools. The baseline prevalence of overweight was comparable. The intervention focused on decreasing consumption of carbonated beverages. The intervention was delivered in 3 one-hour sessions by trained personnel with the assistance of teachers. At 12 months the change in BMI “Z” score was not significantly different between intervention and control classes (mean Z score 0.7 (SD 0.2). However, there was a reduction in the self-reported consumption of soft drinks.

 

Sugar-sweetened beverages have been incriminated in the development of obesity and cardio-metabolic risks in a number of studies (31).  One outcome of this data were 2 randomized clinical trials to reduce the intake of sugar-sweetened beverages among adolescents (32, 33, 34).  At the end of one year the weight gain in one study was significantly smaller in those provided with sugar-free beverages, but most of this benefit, except in the Hispanic children, was lost at 2 years after the treatment program had been discontinued. In the other trial lasting 18 months, the children receiving the artificially sweetened beverages gained less weight than those drinking the sugar-sweetened soft-drinks (33). In a follow-up of this study (34) it was shown that compensation for changing sugar content in beverages was sub-optimal in children in the upper half of the BMI spectrum. Thus, replacing the sugar-sweetened beverages associated with weight gain with lower calorie versions might be beneficial to most children and adolescents.

 

Another trial was conducted in Thailand randomized kindergarten children by class into an exercise group and a control group with 5 classes in each arm (35).  The reduction in the prevalence of obesity tended toward significance (p=0.07). 

 

A US trial including 549 children from 6 schools was stratified by percentage ethnicity (36). This intervention, called SPARK (Sports, Play and Active Recreation for Kids) was a physical education program with a self-management component.  The results for boys showed that the control group had significantly lower BMIs at 6 and 12 months, but not at 18 months. In contrast, the girls in the control group had lower BMIs at each time point that reached statistical significance at 18 months.

 

The Pathways Study (37) is one of the largest studies for prevention of obesity in children.  The participants included 1704 children from 41 American Indian schools.  Children were age 8-11. Pathways was a school-based multi-component, multi-center intervention for reducing percentage body fat. There were 4 components: 1) changing dietary intake; 2) increasing physical activity; 3) A classroom curriculum focused on healthy eating and lifestyle; and 4) a family-involvement program.  At the end of the 3-year study, knowledge improved and fat intake at lunch decreased, but there were no changes in either body composition or activity level measured by motion sensor.

 

The Planet Health study is a high quality randomized controlled trial (38) conducted among 1295 ethnically diverse children in 10 US schools in New England who were randomized by school. The children were 11-12 years old and in the 6-8th grade. The program was a behavioral choice intervention and concentrated on the promotion of physical activity, modification of dietary intake, and reduction of sedentary behavior with an emphasis on reducing time watching television.  At follow-up the percentage of obese girls in the intervention schools was reduced 53% compared with controls, [Odds Ratio 0.47 (95% CI 0.24 to 0.93)].  Each hour of reduction in television time predicted a 15% reduction in obesity [OR 0.85 (95% CI 0.75 to 0.97].  Among the boys there was a decline in BMI in both groups, but no significant difference between them.  Time spent viewing television was reduced among both boys and girls and fruit and vegetable consumption increased significantly. Gortmaker et al (38) concluded that the decline in television watching was a major factor in preventing obesity. 

 

In another clinical trial from Germany, Muller et al (39) randomized a group of 414 children from 6 schools into control or intervention groups in the Kiel Obesity Prevention Study or KOPS. The key messages in the intervention group were to eat more fruits and vegetables each day, to reduce high fat foods, to keep active for at least 1 hour a day, and to decrease television viewing to less than 1 hour a day.  At the end of one year there was no significant difference in change of BMI between the intervention and control groups.

 

In a program called Active Program Promoting Lifestyle in School, or APPLES, 634 children in 10 schools were randomized to intervention or control groups (39). The intervention included teacher training and resources, modification of school meals, support for physical education, and playgroups activities. At one year there was no difference in change in BMI between the children in the two groups, nor was there any difference in dieting behavior.  However, children reported a higher consumption of vegetables. Although APPLES was successful in changing the ethos in the schools and the attitudes of the children, the trial was ineffective in changing weight status.

 

The program “VERB™--It’s What you Do!” was developed by the US Centers for Disease Control and Prevention is another example of a social marketing strategy. It is designed to increase physical activity among ethnically diverse 9- to 13-year-olds (40). The question of whether it is really possible to get long term behavioral change in a society with a vibrant advertising industry remains to be seen.

 

Mind, Exercise, Nutrition – Do it (MEND) is a British program held at a sports center, twice-weekly, for 3 months which consists of behavior modification, physical activity, and nutrition education. In their pilot study, 11 obese children age 7-11 years and their families were recruited and attended a mean of 78% (range 63-88%) of the sessions. Waist circumference, cardiovascular fitness, and self-esteem were all significantly improved at 3 months and continued to improve at 6 months. BMI was significantly improved at 3 months but lost significance by 6 months. This program has now been expanded to many sites through the United Kingdom (41) and Australia (42). The Stockholm Obesity Prevention Program (STOPP) is another randomized trial parents who are overweight or obesity designed to prevent obesity in their children. (43)

 

A meta-analysis examining the impact of lifestyle interventions on body weight and cardio-metabolic outcomes in overweight children found 33 studies with complete data on weight change.  Lifestyle interventions compared to either no treatment control or usual care resulted in significant weight loss of 1.25 to 1.30 kg/m2 (BMI units). In the 15 studies reporting cardio-metabolic outcomes, there were significant reductions in fasting insulin, triglycerides, blood pressure, and LDL-cholesterol, but no effect on HDL-cholesterol (44).  The importance of prevention in childhood and adolescence has prompted 3 Cochrane Reviews of the effect of Lifestyle Interventions. One focused on improvements in school performance (45), another focused on improvements in physical activity and fitness from lifestyle interventions (46), and a third one on the effects on body weight (47).  This latter report showed that lifestyle was effective in in age groups 0 to 5 years, age 6 to 12 and age 13 to 18 with the largest effects seen in the children under 12.  Community wide interventions for Increasing population levels of physical activity are not very effective based on another Cochrane Database Review (48). 

 

Systematic analyses have also been done for special groups of individuals with obesity in the United States. Two of these reviews have examined Latino children and find that lifestyle and physical activity interventions are promising (49, 50).  In a review of studies on Latino and African-American children Robinson et al (51) found that 2 of 17 studies showed benefits for preschool and elementary schools on reducing obesity in African-American children.  Parent-child participation has also been found to be valuable (52). 

 

Strategies Aimed at Adults

 

A large number of trials have been conducted in adults and reviewed in detail by Kumanyika and Daniels (53).  The Pound of Prevention study (54, 55) is probably the largest and most general study for prevention of overweight reported to date.  It demonstrated the feasibility of reaching a large number of people and producing some positive behavioral changes. However, as with many of the studies in children, the interventions were not successful in preventing weight gain relative to the control condition. The decrease of fat intake and increased physical activity were the strongest predictors of weight maintenance (55). Again, behavioral changes were positive and perhaps a higher intensity might have produced different results, but at the present time there is a reasonable argument to be made that money spent on behavioral efforts at changing behavior is wasted. 

 

The results of the 5-year trial from the Healthy Women’s Study is also worth noting (56, 57). Behavioral counseling at 6 months was effective in preventing weight gain during the transition to menopause. The intervention program appears to have been well received, judging from retention rates, but it seems to be labor and cost intensive to deliver.

 

Rural counties in the United States have higher rates of obesity, sedentary lifestyle, and associated chronic diseases than non-rural areas. To tackle this problem, Perri et al worked with the USDA Cooperative Extension Service. They recruited obese women from rural communities who had completed an initial 6-month weight-loss program at Cooperative Extension Service offices in 6 medically underserved rural counties in Florida (n = 234). The women were randomized to extended care program or to an education control group. The extended-care programs entailed problem-solving counseling delivered in 26 biweekly sessions via telephone or face to face. Control group participants received 26 biweekly newsletters containing weight-control advice. The body weight at entry was 96.4 kg, and the women in the intervention group lost 10 kg during the 6-month intervention.  One year after randomization, participants in the telephone and face-to-face extended-care programs regained less weight [1.2± 0.7 and 1.2 ±0.6 kg, (mean± SEM) respectively] than those in the education control group (3.7 ±0.7 kg; P = .03 and .02, respectively). The beneficial effects of extended-care counseling were mediated by greater adherence to behavioral weight-management strategies. Cost analyses indicated that telephone counseling was less expensive than face-to-face intervention, thus offering a strategy of working with the Cooperative Extension service using behavioral weight strategies to modulate body weight (58).

 

Strategies for Preventing Obesity Aimed at the Entire Population 

 

Population-based messages aimed at the public concerning food and exercise are cognitive in nature requiring individual commitment (53). If the “individual” follows the advice in the message this strategy would be sufficient to “overcome” the epidemic of obesity. However, positive preventive messages are delivered in an environment in which there are many alternative messages urging consumption of this or that kind of food or eating at one or another of many different kinds of restaurants. The Low-Fat message of the 1990’s is one example of a message that assumed obesity results from increased fat intake and that reducing fat intake would reverse it. Omitted from this was the realization that eating less fat did not necessarily mean eating fewer calories and thus redressing the energy imbalance.  A test of the low-fat hypothesis as a public health message came from the Women’s Health Initiative. Women were randomly assigned to normal or low-fat diets, but without calorie goals. The women assigned to the low-fat diet lost more weight that the other group, but after the low point, weight was regained (59).   Of particular interest, is that the weight change over 8 years had a strong relation to the level of fat that the women chose to eat. Those with the lower fat intake remained 2 kg or more lighter after 7 years than those in the highest fat intake group indicating that dietary fat is one component of the problem, but that it is not the “whole story”. If one believed that the current epidemic of obesity was due to limited activity, then a campaign like “America on the Move”, which enrolls individuals to use step counters as a way to increase their activity should be an effective strategy (60). The jury is still out on this strategy.

 

An alternative approach might be to re-engineer the built environment to make it both easier to walk and make it more likely that individuals would do so rather than getting into their car (61).  The current housing model of the cluster of houses off a main street where the automobile is essential for mobility will make this strategy a long-term one.  A systematic review by Papas et al (62) identified 20 studies, of which 18 were cross-sectional that examined the relation of obesity to the numbers of outlets for physical activity and food. Seventeen of these found significant relationship between built environment (food outlets or access physical activity) and risk of obesity. The number of recreational facilities and likelihood of overweight in adolescents were significantly related.

 

FOOD            

 

Faith et al (63) reviewed research that used manipulations of the environment to produce weight change. They concluded that easy access to food may influence food purchases, consumption, and possibly weight change while restriction of food availability may accomplish the same goals although this requires further research.  

 

The food industry, for obvious reasons, favors the hypothesis that obesity results from reduced levels of physical activity and strongly supports providing more places for people to exercise and providing them with more healthy alternatives in food stores as a strategy to help overcome the obesity problem (53, 64). However, since “healthy” food items are likely to be more expensive than the ones already on food shelves, and since newer technologies and marketing are needed, it is unclear whether a price-sensitive public can be moved in this direction.

 

Strategies for improving access to healthful foods often focus on fruits and vegetables. The value of a diet high in fruits, vegetables, and low-fat dairy products with a reduction in the level of fat and sugar containing products was found to lower blood pressure across the range of salt intake in individuals who were maintaining their body weight (65, 66).  Regular farmers’ markets, subsidizing the availability of fresh fruits and vegetables to school children, lowering the cost of fruits and vegetables while increasing the price of high-fat or high sugar foods in school or worksite cafeterias, or changing marketing strategies in other ways might increase fruit and vegetable consumption (67, 68, 69, 70, 71, 72). Since we are all price sensitive, these might move choices of food from the lower cost less healthy ones to more healthy choices. However, as Drewnowski has pointed out, the high-fat, high sugar alternatives provide much more food energy for the money than the so-called healthier options (73).

 Another strategy toward this end is to “limit” availability of higher energy foods by making them more expensive.  A review of the use of price of food items by Smed et al (16) has shown that in Europe increasing the tax or reducing the subsidies on “unhealthy” items and reducing the tax on “healthy” items through the value added tax system could shift consumption toward healthier foods (15).   Federally funded programs such as food stamps, school lunches, and meals on wheels could also be used toward this end. Pending the willingness of the public and the politicians to tackle some of the political implications of tax policy to combat the epidemic of obesity, this strategy is likely to remain on the back burner (13). 

 

INCREASING PHYSICAL ACTIVITY AS A FOCUS FOR PREVENTION

 

An overall increase in physical activity would increase energy expenditure and is one strategy for prevention of obesity. One assumption of such a strategy is that activity may have decreased during the time that the epidemic of obesity developed. This is difficult to establish, but a recent publication used total daily energy expenditure in a small number of individuals (some 300) measured over years (74). This strategy would be appropriate, whether there has been a decrease in energy expenditure or not, and the data on this question is unclear. In contrast, Church et al (8) have presented data showing that leisure activity has declined as the obesity epidemic has moved ahead.  Altering the “built environment” such as side-walks and shopping centers is one way to do encourage more physical activity (61). However, there is a 30–40-year lag between initiation of changes in architectural land use and real changes in configuration of sidewalks, making these approaches unlikely to impact this problem in the foreseeable future. 

 

How much has our daily activity level changed?  To examine this question Cutler et al (75) examined levels of activity in various tasks from 1965 to 1995, which covers the period before and to the peak of weight gain. They found that over the 30 years from 1965 to 1995 the major changes in activity have been a decrease in household work and an increase in recreation and communication. These are relatively small compared to the proportion of the daily routine which is spent in paid work and in personal needs and care. We thus think food intake is a more viable strategy for combating the obesity epidemic.

 

Work places are another place where prevention and intervention can occur. In a review of worksite nutrition and physical activity programs Anderson et al (76) noted modest improvements in employee weight status at the 6-12-month follow-up. Based on 9 randomized controlled trials, there was a loss of 2.8 pounds (-1.3 kg) (95% CI=-4.6, -1.0 weight loss) and a decrease of 0.5 (95% CI=-0.8, -0.2) BMI units based on six RCTs. The findings were applicable to both male and female employees, across a range of worksite settings. Most of the studies combined informational and behavioral strategies to influence diet and physical activity; fewer studies modified the work environment (e.g., cafeteria, exercise facilities) to promote healthy choices. This is an area of potential future advance.

 

The nutrition-transition in China provides an interesting example of how the modern way of life makes preservation of physical activity so difficult (77, 78).  As recently as 20 years ago, the bicycle was a major mode of transport for Chinese. This is no longer the case. The automobile and public transport systems are relegating the bicycle to museums. Whether understanding the need for people to move can provide a rescue strategy for weight gain is doubtful.

 

USE OF SOCIAL MARKETING

 

One element in trying to combat the obesity epidemic is to focus on the needs of selected groups – so-called social marketing. The idea is to provide focused messages targeted at specific sub-groups.  Another approach is to focus on specific food groups. The program by the National Cancer Institute to increase Fruits and Vegetable consumption through the “5 A Day for Better Health” program is an example of this idea. Although we would all agree that this is a desirable approach, its effectiveness in changing the consumption of fruits and vegetables has not been overwhelming (79).  

 

SUMMARY AND CONCLUSION

 

Efforts to prevent obesity or to reverse components through changing lifestyle have focused on both adults and children and well as worksites. Although some changes can be documented in these studies, the net effects have been small and the epidemic has continued to move ahead.  The role of economic incentives has received less exploration, but may be more promising as a way to halt this epidemic.

 

REFERENCES

 

  1. Ng M, Fleming T, Robinson M, et al.Global regional, and national prevalence of overweight and obesity in children and adults during 1980-2013: a systematic analysis of the Global burden of Disease Study 2013.  Lancet 2014;384:766-781.
  2. Ogden CL, Carroll MD, Kit BK, Flegal KM. 2014.Prevalence of childhood and adult obesity in the United States, 2011-2012. JAMA. 311(8):806-814.
  3. Lo JC,Maring BChandra MDaniels SRSinaiko ADaley MFSherwood NEKharbanda EOParker EDAdams KFPrineas RJMagid DJO'Connor PJ,Greenspan LC. Prevalence of obesity and extreme obesity in children aged 3-5 years.  Pediatr Obes. 2014 Jun;9(3):167-75.
  4. Finkelstein EA, Graham WC, Malhotra R. Lifetime Direct Medical Costs of Childhood Obesity. Pediatrics. May 2014;133(5):854-862.
  5. Bray GA. A Guide to Obesity and the Metabolic Syndrome. Boca Raton FL: CRC Press A Division of Taylor and Francis, 2011.
  6. Putnam, J Allshouse J, Kantor LS.U.S. per capita food supply trends: More calories, refined carbohydrates and fats.  Food Rev 2002;25 (3):2-15.
  7. Scully T. Public health: Society at large. Nature. 2014;508:S50-1.
  8. Swinburn B, Sacks G, Ravussin E. Increased food energy supply is more than sufficient to explain the US epidemic of obesity. Am J Clin Nutr. 2009;90:1453-6.
  9. Church TS, Thomas DM, Tudor-Locke C, Katzmarzyk PT, Earnest CP, Rodarte RQ, Martin CK, Blair SN, Bouchard C.Trends over 5 decades in U.S. occupation-related physical activity and their associations with obesity.  PLoS One. 2011;6(5):e19657.
  10. Tillotson JE.Role of Agricfulture and the food industry in America’s obesity.  IN: Handbook of Obesity GA Bray and C Bouchard (eds) New York: Informa Health Care Third Edition, 2014;413-426.
  11. Bray GA.From Farm to Fat Cell: Why Aren't We All Fat? Metabolism. 2015 Mar;64(3):349-353.
  12. Loos RJF. Genetic determinants of common obesity and their value in prediction.Best Prac Res Clin Endo Metab 2012:26:211-226.
  13. Ramachandrappa S, Farooqi IS.Genetic approaches to understanding human obesity.  J Clin Invest 2011;121:2080-2086.
  14. Locke AE, Kahali B, Berndt SI, et al Genetic studies of body mass index yield new insights for obesity biology.Nature 2015;Feb 12:518:197-206.
  15. Brownell KD, Farley T, Willett WC, Popkin BM, Chaloupka FJ, Thompson JW, 2009. Ludwig DSThe public health and economic benefits of taxing sugar-sweetened beverages.N Engl J Med. Oct 15;361(16):1599-605.
  16. Smed S, Jensen JD, Denver S.2005b. Differentiated food taxes as a tool in health and nutrition policy.  XIth congress of the European Association of Agricultural Economists Copenhagen DK August 24-27.
  17. Smed S, Denver S.2005a. Taxing as economic tools in health policy.  97th EAAE Seminar University of Reading April 21-22.
  18. WHO/FAO Joint Expert Consultation on: Diet, Nutrition and the Prevention of Chronic Disease.2002. Geneva: WHO Technical Series Report # 916.
  19. Bray, G.A.The epidemic of obesity and changes in food intake: The Fluoride Hypothesis.  Physiol Behav 82:115-121, 2004.
  20. Pollan M 2008. Farmer In Chief.What the next President can and shuld do to remake the way we grow and eat our food. New York Times Magazine;62-68. Oct 12.
  21. Tanvig M, Vinter CA, Jorgensen JS, Wehberg S, Ovesen PG, Beck-Nielsen H, Christesen HT, Jensen DM.Effects of lifestyle intervention in pregnancy and anthropemetrics at birth on offspring metabolic profile at 2.8 years: results from the Lifestyle in Pregnancy and Offspring (LiPO) Study. JCEM 2015:100:175-183.
  22. Thangaratinam S, Rogozinska E, Jolly K, et al Interventions to reduce or prevent obesity in pregnant women: a systematic review.Health Tech:iii-iv, 1-191. 
  23. Thangaratinam S, Rogozinska E, Jolly K, Glinkowski S, Roseboom T, Tomlinson JE, Kunz R, Mol BW, Coomarasamy A, Khan KS.Effects of interventions in pregnancy onmaternal weight and obstetric outcomes: meta-analysis of randomized evidence.   BMJ 2012 May 16;344:e2088.
  24. Tanentsapf, I, Heitman BL, Adegboye AR.Systemataic Review of clinical trials on dietary interventions to prevent excessive weight gain during pregnancy among normal weight, overweight and obese women.  BMC Pregnancy Childbirth 2011 Oc 26;11:81.
  25. Muktabhant B, Lawrite TA, Lumbiganon P, Laopaiboon M.Diet or exercise, or both for preventing excessive weight gain in pregnancy.  Cochrane Database Syst Rev 2015 June 15;6:CD007145.
  26. Bruch, H.The importance of overweight.  New York: W.W. Norton & Co, Inc 1957.
  27. Berkowitz RI, Moore RH, Faith MS, Stallings VA, Kral TV, Stunkard AJ.2009. Identification of an Obese Eating Style in 4-year-old Children Born at High and Low Risk for Obesity.  Obesity (Silver Spring). Sep 24.
  28. Harris KC, Kuramoto LK, Schulzer M, Retallack JE.2009. Effect of a school-based physical activity interventions on body mass index in children: a meta-analysis.  CMAJ;180:719-726.
  29. Epstein LH, Gordy CC, Raynor HA, Beddome M, Kilanowski CK, Paluch R.Increasing fruit and vegetable intake and decreasing fat and sugar intake in families at risk for childhood obesity.  Obes Res. 2001 Mar;9(3):171-8.
  30. James J, Thomas P, Cavan D, Kerr D. 2004. Preventing childhood obesity by reducing consumption of carbonated drinks: cluster randomised controlled trial. BMJ 2004 May 22;328(7450):1237.
  31. Bray GA, Popkin BM.Dietary sugar and body weight: have we reached a crisis in the epidemic of obesity and diabetes?: health be damned! Pour on the sugar.  Diabetes Care. 2014 Apr;37(4):950-6.
  32. Ebbeling CB, Feldman HA, Chomitz VR, Antonelli TA, Gortmaker SL, Osganian SK, Ludwig DS.A randomized trial of sugar-sweetened beverages and adolescent body weight.  N Engl J Med. 2012 Oct 11;367(15):1407-16.
  33. de Ruyter JC, Olthof MR, Seidell JC, Katan MB. A trial of sugar-free or sugar-sweetened beverages and body weight in children.  N Engl J Med. 2012 Oct 11;367(15):1397-406.
  34. Olthof MR, de Ruyter JC, Kuijper LDJ, Chow CC, Hall KD, Katan MB. Impact of masked replacement of sugar-sweetened with sugar-free beverages on body weight increases with initial BMI: an 18 month double-blind trial in children.PLoS One 2016 Jul 22;11(7):e0159771.
  35. Mo-suwan L, Pongprapai S, Junjana C, Puetpaiboon A. Effects of a controlled trial of a school-based exercise program on the obesity indexes of preschool children. Am J Clin Nutr. 1998 Nov;68(5):1006-11.
  36. Dowda M, James F, Sallis JF, McKenzie TL, Rosengard P, Kohl HW 3rd. Evaluating the sustainability of SPARK physical education: a case study of translating research into practice.Res Q Exerc Sport. 2005; Mar;76(1):11-9.
  37. Caballero B, Clay T, Davis SM, Ethelbah B, Rock BH, Lohman T, Norman J, Story M, Stone EJ, Stephenson L, Stevens J; Pathways Study Research Group.Pathways: a school-based, randomized controlled trial for the prevention of obesity in American Indian schoolchildren.  Am J Clin Nutr. 2003 Nov;78(5):1030-8.
  38. Gortmaker SL, Peterson K, Wiecha J, Sobol AM, Dixit S, Fox MK, Laird N.1999. Reducing obesity via a school-based interdisciplinary intervention among youth: Planet Health.  Arch Ped Adol Med;153:409-418.
  39. Muller HL, Bueb K, Bartels U, Roth C, Harz K, Graf N, Korinthenbom R, Bettendorf M, Kuhl J, Gutjahr P, Sorensen N, Calaminus G.2001. Obesity after childhood craniopharyngioms - German multicenter study on pre-operative risk factors and quality of life. Klin Pediatr.213:244-249.
  40. Wong F, Huhman M, Heitzler C, Asbury L, Bretthauer Mueller R, McCarthy S, Londe P.2004. VERB - a social marketing campaign to increase physical activity among youth.  Prev Chronic Dis.;1:A10. 
  41. Sacher PM, Chadwick P, Wells JC, Williams JE, Cole TJ, Lawson MS. Assessing the acceptability and feasibility of the MEND Programme in a small group of obese 7-11-year-old children. J Hum Nutr Diet. 2005 Feb;18(1):3-5.
  42. Skouteris H, Mcabe M, Swinburg B, Hill B.Healthy Eating and obesity prevention for preschoolers: a randomized controlled trial BMC Public Health 2010 Apr 28:10:220.
  43. Sobko T, Svensson V Ek A, Ekstedt M, Karslsson H, Johansson E, Cao Y, Hagstromer M, Marcus C.A randomized controlled trial. BMC Public Health 2011 May 18;11:336.
  44. Ho M, Garnett SP, Baur L et al. Effectiveness of lifestyle intervention in child obesity:Ped 2012647-71.
  45. Martin A, Saunders DH, Shenkin SD, Sproule J. Lifestyle intervention, Cochrane Database Syst Rev 2014; Mar 14;3:CD009728.
  46. Dobbins M, Husson H, DeCorby K, LaRocca RL.School-based physical activity…. Cochrane Databae Syst Rev 2013;2:CD007651.
  47. Waters E, de Silva-Sanigorski A, Hall BJ, et al.  Interventions fo preventing obesity in children.  Cochrane Database Syst Rev 2011;Dec 7;(12):CD001871.
  48. Baker PR, Francis DP, Soares J, Weightman AL, Foster C.Community wide interventions for increasing physical activity.  Cochrane Database Syst Rev 2011 2011 Apr 13;(4):CD008366.
  49. Perez-Morales ME, Bacardi-Gascon M, Jimenez-Cruz A.Childhood overweight and obesity prevention interventions among Hispanic children in the United States: systematic review. Nutr Hosp 2012;27:1415-1421. 
  50. Holub CK, Lobelo F, Mehta SM, Sanchez romero LM, Arredondo EM, Elder JP.  School-wide programs aimed at obesity among Lantino youth in the United States: a review of the evidence.  J Sch Health 2014;84:239-246. 
  51. Robinson LE, Webster EK, Whitt-Glover MD, Ceaser G, Alhassan S.Effectiveness of pre-school and school-based interventions. Obes Rev 2014 Oct;15Suppl 4:5-25.
  52. Barr-Anderson DJ, Adams-Wynn AW, DiSantis KI, Kumanyika S. Family-focused physical activity, diet and obesity interventions in African-American girls: a systematic review. Obes rev 2013Jan; 14:29-51.
  53. Kumanyika SK, Daniels SR.2006. Obesity Endo IN: Overweight and the Metabolic Syndrome: From Bench to Bedside, GA Bray and DH Ryan (eds) Endocrine Updates;233-253.
  54. Jeffery RW, French SA. 1999. Preventing weight gain in adults: the pound of prevention study. Am J Public Health.;89:747-51.
  55. Sherwood NE, Jeffery RW, French SA, Hannan PJ, Murray DM.2000. Predictors of weight gain in the Pound of Prevention study.  Int J Obes Relat Metab Disord. 24:395-403.
  56. Simkin-Silverman LR, Wing RR, Boraz MA, Kuller LH.2003. Lifestyle intervention can prevent weight gain during menopause: results from a 5-year randomized clinical trial. Ann Behav Med.;26:212-20.
  57. Kuller LH, Simkin-Silverman LR, Wing RR, Meilahn EN, Ives DG.2001. Women's Healthy Lifestyle Project: A randomized clinical trial: results at 54 months.  Circulation.;103:32-7.
  58. Perri MG, Limacher MC, Durning PE, Janicke DM, Lutes LD, Bobroff LB, Dale MS, Daniels MJ, Radcliff TA, Martin AD.  2008. Extended-care programs for weight management in rural communities: the treatment of obesity in underserved rural settings (TOURS) randomized trial. Arch Intern Med. Nov 24;168(21):2347-54.
  59. Howard B. V., J. E. Manson, et al. 2006. "Low-fat dietary pattern and weight change over 7 years: the Women's Health Initiative Dietary Modification Trial." Jama 295(1): 39-49.
  60. Hill JO Peters J. Jortberg BT.The Step Diet: Count Steps, Not Calories to Lose Weight and Keep It off Forever2004.
  61. Sallis JF, Glanz K.2009. Physical activity and food environments: solutions to the obesity epidemic. Milbank Q. Mar;87(1):123-54. Review.
  62. Papas MA, Alberg AJ, Ewing R, Helzlsouer KJ, Gary TL, Klassen AC. The built environment and obesity. Epidemiol Rev. 2007;29:129-43.
  63. Faith MS, Fontaine KR, Baskin ML, Allison DB. 2007. Toward the reduction of population obesity: macrolevel environmental approaches to the problems of food, eating, and obesity.Psychol Bull. Mar;133(2):205-26. Review.
  64. James WPT, Gill TP.2008. Prevention of Obesity IN: Handbook of Obesity: Clinical Applications, G.A. Bray and C. Bouchard (eds), New York: Informa Healthcare p 157-175.
  65. Appel LJ, Moore TJ, Obarzanek E, Vollmer WM, Svetkey LP, Sacks FM, Bray GA, Vogt TM, Cutler JA, Windhauser MM, Lin PH, Karanja N.1997. A clinical trial of the effects of dietary patterns on blood pressure. DASH Collaborative Research Group.  N Engl J Med. Apr 17;336(16):1117-24.
  66. Sacks FM, Svetkey LP, Vollmer WM, Appel LJ, Bray GA, Harsha D, Obarzanek E, Conlin PR, Miller ER 3rd, Simons-Morton DG, Karanja N, Lin PH; 2001. DASH-Sodium Collaborative Research Group. Effects on blood pressure of reduced dietary sodium and the Dietary Approaches to Stop Hypertension (DASH) diet. DASH-Sodium Collaborative Research Group.N Engl J Med. Jan 4;344(1):3-10.
  67. Kennedy B.M., S. Paeratakul, D.H. Ryan G.A. Bray.Socioeconomic status and health disparity in the United States.  J Human Behav Soc Envir 15(2/3): 13-23, 2007.
  68. Buzby JC., Guthrie JF, Kantor LS. 2003. Evaluation of the USDA Fruit and Vegetable Pilot Program: Report to Congress. U.S. Department of Agriculture, Economic Research Service. E-FAN-03-006.
  69. Buzby, JC, Guthrie,, J and Kantor, LS. Evaluation of the USDA Fruit and Vegetable Pilot Program: Report to Congress.  www.ers.usda.gov/publications/efan03006.
  1. Glanz K, Hoelscher D. 2004. Increasing fruit and vegetable intake by changing environments, policy and pricing: restaurant-based research, strategies, and recommendations. Prev Med. 39 Suppl 2:S88-93.
  2. Glanz K, Yaroch AL. 2004. Strategies for increasing fruit and vegetable intake in grocery stores and communities: policy, pricing, and environmental change. Prev Med. 39 Suppl 2:S75-80.
  3. French SA, Wechsler H. 2004. School-based research and initiatives: fruit and vegetable environment, policy and pricing workshop. Prev Med. 39 Suppl 2:S101-7).
  4. Drewnowski A, Darmon N.  The economics of obesity: dietary energy density and energy cost.  Am J Clin Nutr.2005 Jul;82(1 Suppl):265S-273S. Review.
  5. Westerterp K, Speakman J.  2008. Physical activity energy expenditure has not declined since the 1980s and matches energy expenditures of wild mammals. Int J Obes (Lond). Aug;32(8):1256-63.
  6. Cutler DM, Glaeser EL, Shapiro JM. Why have Americans become more obese? Journal of Economic Perspectives 17(3):93-118; 2003.
  7. Anderson LM, Quinn TA, Glanz K, Ramirez G, Kahwati LC, Johnson DB, Buchanan LR, Archer WR, Chattopadhyay S, Kalra GP, Katz DL; 2009. Task Force on Community Preventive Services.  The effectiveness of worksite nutrition and physical activity interventions for controlling employee overweight and obesity: a systematic review.  Am J Prev Med. Oct;37(4):340-57. Review.
  8. Zhai F, Wang H, Du S, He Y, Wang Z, Ge K, Popkin BM.  2009. Prospective study on nutrition transition in China.  Nutr Rev. May;67 Suppl 1:S56-61.
  9. Monda KL, Adair LS, Zhai F, Popkin BM.  2008. Longitudinal relationships between occupational and domestic physical activity patterns and body weight in China.  Eur J Clin Nutr. Nov;62(11):1318-25.
  10. Stables GJ, Subar AF, Patterson BH, Dodd K, Heimendinger J, Van Duyn MA, Nebeling L. 2002. Changes in vegetable and fruit consumption and awareness among US adults:results of the 1991 and 1997 5 A Day for Better Health Program surveys.  J Am Diet Assoc.;102:809-17.

 

TEST FOR TABLE DESIGN

WEBSITE TABLES TESTING

Lorem ipsum dolor sit amet, consectetur adipiscing elit, sed do eiusmod tempor incididunt ut labore et dolore magna aliqua. Ut enim ad minim veniam, quis nostrud exercitation ullamco laboris nisi ut aliquip ex ea commodo consequat. Duis aute irure dolor in reprehenderit in voluptate velit esse cillum dolore eu fugiat nulla pariatur. Excepteur sint occaecat cupidatat non proident, sunt in culpa qui officia deserunt mollit anim id est laborum.

Lorem ipsum dolor sit amet, consectetur adipiscing elit, sed do eiusmod tempor incididunt ut labore et dolore magna aliqua. Ut enim ad minim veniam, quis nostrud exercitation ullamco laboris nisi ut aliquip ex ea commodo consequat. Duis aute irure dolor in reprehenderit in voluptate velit esse cillum dolore eu fugiat nulla pariatur. Excepteur sint occaecat cupidatat non proident, sunt in culpa qui officia deserunt mollit anim id est laborum.

Lorem ipsum dolor sit amet, consectetur adipiscing elit, sed do eiusmod tempor incididunt ut labore et dolore magna aliqua. Ut enim ad minim veniam, quis nostrud exercitation ullamco laboris nisi ut aliquip ex ea commodo consequat. Duis aute irure dolor in reprehenderit in voluptate velit esse cillum dolore eu fugiat nulla pariatur. Excepteur sint occaecat cupidatat non proident, sunt in culpa qui officia deserunt mollit anim id est laborum.Lorem ipsum dolor sit amet, consectetur adipiscing elit, sed do eiusmod tempor incididunt ut labore et dolore magna aliqua. Ut enim ad minim veniam, quis nostrud exercitation ullamco laboris nisi ut aliquip ex ea commodo consequat. Duis aute irure dolor in reprehenderit in voluptate velit esse cillum dolore eu fugiat nulla pariatur. Excepteur sint occaecat cupidatat non proident, sunt in culpa qui officia deserunt mollit anim id est laborum.

Table 1.   Widely Held Misconceptions About ZES

1) Gastrinomas, similar to a number of other pNEN (insulinomas, gastrinomas, PPomas), primarily occur in the pancreas.  FACT: In recent studies, 60-100% of gastrinomas in both sporadic ZES and MEN1/ZES occur in the duodenum, with only 0-15% in the pancreas (6,43,50,95,102,127,175-179) (Table 2).

2) MEN1 is uncommon in ZES, similar to other pNEN such as insulinomas (3-5%), glucagonomas (<5%), PPomas/nonfunctional pNEN (<3%). FACT: MEN1 is found in the highest frequency of all pNEN syndromes in ZES patients occurring in 20-25% and is important to diagnose because of its different treatment aspects (30,50,64,72,87,89,95,102).

3) With the increased awareness of ZES and widespread availability of gastrin assays and sensitive imaging modalities, similar to some other pNEN, gastrinomas are being diagnosed earlier. FACT: The time of onset of symptoms to diagnosis of ZES remains 4-7 years (24,26,48,60,62,89,134) and a number of factors are contributing to make the diagnosis even more difficult (See point #4 below).

4) As recommended in all guidelines (9,72,80,152,157,180-182), similar to other functional pNEN syndromes (F-pNENs), ZES is currently diagnosed by demonstrating excess hormone production (fasting hypergastrinemia) in the presence of an unphysiological effect of the hormone hypersecretion (i.e., inappropriate acid hypersecretion (elevated basal acid output>15 mEq/hr., pH<2)) (9,50,51,55,56,59,70,72,73,79,181,183,184). FACT: In contrast to, for example, insulinomas, which are uniformly diagnosed by demonstrating fasting hyperinsulinemia with accompanying hypoglycemia (frequently during a fasting study) (29,50,185-188), in a recent review of the last 20 cases of ZES reported in the literature in 2018 (55), 95% of the diagnoses were reported without performing a gastric analysis or gastric pH assessment (55) and thus not using classical established criteria. This approach has complicated the diagnosis of ZES and the factors leading to this confusion will be discussed below in detail in the ZES diagnosis section.

5) In MEN1 patients, similar to other MEN1 patients with F-pNEN such as insulinomas and glucagonomas, most gastrinomas can be cured by nonaggressive surgical resections in MEN1/ZES patients. FACT:  In contrast to other F-pNEN (29,157,189), the 5-year surgical cure rate of MEN1/ZES is <5% (6,30,43,44,88,190) without aggressive surgical resections such as Whipple resection, which are not recommended (6,9,88,92,93,118,123,157,180,182). However, without these resections, most patients with small tumors and adequate acid secretory control have an excellent prognosis, which has led to controversy in their treatment, and will be discussed in the surgical section later (30,43,47,92,93,95,102,118,157,180,182,191).

Table 1.   Widely Held Misconceptions About ZES

1) Gastrinomas, similar to a number of other pNEN (insulinomas, gastrinomas, PPomas), primarily occur in the pancreas.  FACT: In recent studies, 60-100% of gastrinomas in both sporadic ZES and MEN1/ZES occur in the duodenum, with only 0-15% in the pancreas (6,43,50,95,102,127,175-179) (Table 2).

2) MEN1 is uncommon in ZES, similar to other pNEN such as insulinomas (3-5%), glucagonomas (<5%), PPomas/nonfunctional pNEN (<3%). FACT: MEN1 is found in the highest frequency of all pNEN syndromes in ZES patients occurring in 20-25% and is important to diagnose because of its different treatment aspects (30,50,64,72,87,89,95,102).

3) With the increased awareness of ZES and widespread availability of gastrin assays and sensitive imaging modalities, similar to some other pNEN, gastrinomas are being diagnosed earlier. FACT: The time of onset of symptoms to diagnosis of ZES remains 4-7 years (24,26,48,60,62,89,134) and a number of factors are contributing to make the diagnosis even more difficult (See point #4 below).

4) As recommended in all guidelines (9,72,80,152,157,180-182), similar to other functional pNEN syndromes (F-pNENs), ZES is currently diagnosed by demonstrating excess hormone production (fasting hypergastrinemia) in the presence of an unphysiological effect of the hormone hypersecretion (i.e., inappropriate acid hypersecretion (elevated basal acid output>15 mEq/hr., pH<2)) (9,50,51,55,56,59,70,72,73,79,181,183,184). FACT: In contrast to, for example, insulinomas, which are uniformly diagnosed by demonstrating fasting hyperinsulinemia with accompanying hypoglycemia (frequently during a fasting study) (29,50,185-188), in a recent review of the last 20 cases of ZES reported in the literature in 2018 (55), 95% of the diagnoses were reported without performing a gastric analysis or gastric pH assessment (55) and thus not using classical established criteria. This approach has complicated the diagnosis of ZES and the factors leading to this confusion will be discussed below in detail in the ZES diagnosis section.

5) In MEN1 patients, similar to other MEN1 patients with F-pNEN such as insulinomas and glucagonomas, most gastrinomas can be cured by nonaggressive surgical resections in MEN1/ZES patients. FACT:  In contrast to other F-pNEN (29,157,189), the 5-year surgical cure rate of MEN1/ZES is <5% (6,30,43,44,88,190) without aggressive surgical resections such as Whipple resection, which are not recommended (6,9,88,92,93,118,123,157,180,182). However, without these resections, most patients with small tumors and adequate acid secretory control have an excellent prognosis, which has led to controversy in their treatment, and will be discussed in the surgical section later (30,43,47,92,93,95,102,118,157,180,182,191).

Radiology of the Pituitary

ABSTRACT

 

MRI is the primary imaging modality for the pituitary gland. This chapter reviews and illustrates the normal anatomy and MRI appearances of the pituitary gland and hypothalamic region. The optimal MRI technique relies on thin section T1-weighted sequences in the sagittal and coronal planes before and after gadolinium contrast enhancement. T2-weighted sequences can add useful additional information in some cases but are not a substitute for T1-weighted sequences. Congenital abnormalities with characteristic imaging appearances including the ectopic posterior pituitary and hypothalamic hamartoma are illustrated, along with images of all the common primary pituitary pathologies: adenomas, Rathke’s cysts and hypophysitis (infundibular, lymphocytic, and immunotherapy drug-induced). Non-pituitary origin pathologies which may involve the pituitary or parasellar region are also illustrated, including meningioma, arachnoid cysts, germinoma, craniopharyngioma and chondrosarcoma; they all have distinct imaging appearances. Inflammatory processes such as sarcoid or Langerhans cell histiocytosis can also involve the pituitary stalk or hypothalamus and in these cases diabetes insipidus may be a clinical feature, correlating with stalk involvement on imaging. More recently, IgG4- and immune checkpoint inhibitor-associated hypophysitis have emerged as new diseases entities. Metastases or lymphoma can occasionally involve the pituitary gland and stalk. The granular cell tumor is a rare tumor arising in the pituitary stalk and has a typical appearance, distinct from other inflammatory or infiltrative stalk processes.

 

ANATOMY AND EMBRYOLOGY

 

The pituitary gland sits within the sella turcica which is a cup-shaped depression in the sphenoid bone. The sphenoid air sinus lies below and anterior to the sella turcica (Fig 1). Lying above the pituitary gland is a cerebrospinal fluid (CSF) space, the suprasellar system, which contains the optic chiasm (Figs 1 and 2). The lateral walls of the pituitary fossa are formed by the cavernous sinuses (Fig 1B) which contain the internal carotid arteries as well as a number of cranial nerves: the 3rd, 4th and 6th cranial nerves as well as the first and second divisions of the 5th cranial nerve. The pituitary gland is connected via the pituitary stalk to the hypothalamus, which is a thin plate of tissue making up the floor of the anterior part of the 3rd ventricle (Figs 1 and 3).

Figure 1. Sagittal T1 weighted unenhanced image of the pituitary fossa demonstrate normal anatomy. The anterior pituitary tissue, A, is visible within the sella and the posterior pituitary bright spot, P, is evident behind it. The stalk (arrow) is well seen with a small cleft of CSF visible within it superiorly –the infundibular recess of the third ventricle. The optic chiasm, C, and mamillary bodies, M, are seen in the suprasellar region. B - brainstem, S - sphenoid air sinus, CL - clivus.

Figure 2. Coronal T1 weighted unenhanced image of the pituitary fossa. The anterior pituitary gland, A, is within the fossa. The posterior pituitary bright spot is visible centrally, P. The stalk is seen extending up into the suprasellar region. The optic chiasm, C, is visible. The cavernous segments of the carotid arteries, I, are seen within the cavernous sinuses, which form the lateral boundaries of the pituitary fossa.

Figure 3. Sagittal T1 weighted enhanced image of the pituitary. The pituitary tissue has enhanced, as has the pituitary stalk.

 

The appearance and size of the pituitary gland changes during life. At birth, it is typically globular in shape and shows high signal on T1 weighted images (1). By approximately 6 weeks of age this high signal has diminished, and the anterior pituitary tissue has a similar signal to brain tissue. The posterior pituitary tissue, however, retains a bright signal on T1 weighted sequences. This so-called “posterior pituitary bright spot” is a normal appearance thought to be due to the high neurophysin content (which is not present in the anterior pituitary tissue) (Fig 1).

 

The size of the pituitary gland varies with age and sex. On average it is between 3 and 8mm in height but is generally larger in females than males. The height increases during adolescence due to normal physiological hypertrophy (2). There is also a slight increase in size seen during the sixth decade in females. The most striking physiological changes are seen during pregnancy when the gland progressively enlarges reaching a maximal height immediately after birth when it may reach 10mm in height (3).

 

Embryologically, the anterior and posterior pituitary lobes are distinct. The anterior lobe forms from an invagination of the oral ectoderm known as Rathke’s Pouch. The posterior pituitary forms from a protrusion of the neural ectoderm of the diencephalon. Between the anterior and posterior lobes lies an intermediate lobe which is vestigial and known as the pars intermedia. This is a potential site for small non-functional Rathke’s cysts (Fig 4).

 

Figure 4. Sagittal enhanced T1 weighted image demonstrating a small Rathke’s cyst. This is seen to lie just below the insertion of the pituitary stalk and centrally within the gland. Although it is possible that a small cystic adenoma could have these appearances, this is a very typical location for a Rathke’s cyst arising in the pars intermedia.

 

MR IMAGING

 

MR is the imaging of choice for the pituitary gland. In order to optimize the study, it is necessary to perform thin sections (2mm or 3mm) targeted to the pituitary fossa and performed in both the sagittal and coronal planes. T1 weighted sequences before and after intravenous contrast are the mainstay of pituitary imaging (Fig 1-3) (4). Coronal T2 weighted sequences can also give added information but are less sensitive in the detection of adenomas. Sequences before and after intravenous contrast are the main stay of pituitary imaging (Fig 1-3). Coronal T2 weighted sequences can also give added information but are less sensitive in the detection of adenomas. The higher field strength 3 Tesla MR scanners, which are now in more widespread clinical use, can provide higher resolution pituitary images, the T2 images being reliably improved at 3T. However, artefacts related to vascular flow and patient movement are more pronounced and may outweigh the benefits. CT does not provide such excellent soft tissue resolution as MR but can be a very useful investigation if MR is not possible, and also if it is important to identify the presence of calcification in or around the sella. A dedicated CT study should be performed with a 1mm slice thickness in the axial plane and then reconstructed in the sagittal and coronal planes.

 

There can be some benefit in performing the post-contrast MR sequences in a dynamic fashion (within the first 60 seconds) after contrast injection. This can maximize the conspicuity of adenomas within the pituitary gland, which typically enhance less than the normal pituitary tissue, and this differential enhancement is sometimes best appreciated within the first arterial phase of the contrast injection (Figs 4 and 5) (5). However, in the majority of cases the lesions are adequately demonstrated on a standard acquisition (non-dynamic) scan after the contrast administration (Fig 7) (5).

 

Figure 5. Coronal T1 weighted image of the pituitary gland before contrast. There is a microadenoma in the right side of the gland. On the unenhanced image there is evidence of depression of the floor of the sella on the right side but the microadenoma cannot be visualized within the gland.

Figure 6. Coronal T1 weighted image of the pituitary gland immediately after contrast. There is a microadenoma in the right side of the gland. After contrast a dynamic acquisition shows an area of lesser enhancement indicative of a microadenoma.

Figure 7. Coronal T1 weighted image of the pituitary gland that demonstrates a left sided microadenoma (arrow) which was best seen on this nondynamic post-contrast sequence.

 

The pituitary gland, pituitary stalk and cavernous sinuses are all vascular structures which are seen to enhance after gadolinium injection. The optic chiasm and hypothalamus, however, do not show enhancement if the blood brain barrier is intact (Figs 1-3).

 

PITUITARY ADENOMAS

 

Pituitary adenomas are by far the most common mass lesion seen in the sella and parasellar region. They are slow-growing benign neoplasms arising from the anterior pituitary tissue; radiologically they are simply classified by size: lesions smaller than 10mm transversely are termed microadenomas and those greater than 10mm are macroadenomas. The clinical classification separates adenomas into those that are hormonally activity, e.g., prolactinomas, and those that do not have measurable evidence of hormonal activity referred to as non-functioning adenomas. The histology of the so-called non-functioning adenoma suggests that these arise from gonadotroph cells. The imaging appearances of pituitary adenomas is similar whether they are hormonally functioning or not. Obviously, non-functioning adenomas are more likely to present when they have attained a significant size and are producing effects on local structures around the sella.

 

Pituitary Macroadenomas

 

Pituitary macroadenomas can extend superiorly into the suprasellar cistern (Figs 8-10) and impinge on the optic nerves and/or optic chiasm to produce visual field abnormalities (typically a bitemporal hemianopia). Pituitary macroadenomas with a large suprasellar component characteristically show the appearance of “waisting” (Figs 9 and 10) as they pass through the diaphragma sellae, the sheet of dura which normally lies above the pituitary gland. Lateral growth of a macroadenoma is initially seen to cause deformity of the cavernous sinus; however, adenomas can invade into the cavernous sinus. (Fig 11) This may be associated with symptoms or signs related to involvement of the cranial nerves that run in the cavernous sinus. The third, fourth and sixth cranial nerves run through the cavernous sinus, as do the first and second divisions of the fifth (trigeminal) nerve. If the MR imaging demonstrates adenoma tissue extending beyond the most lateral margin of the cavernous segment of the internal carotid artery, then it is very likely that there is tumor within the cavernous sinus (6). In many cases, however, tumor simply deforms the cavernous sinus flattening the medial wall but not extending more laterally, and in these cases the tumor is typically confined to the sella and has not invaded into the cavernous sinus at surgery. Adenomas can also extend inferiorly into the sphenoid producing remodeling of the bone. Occasionally, macroadenomas will show a very extensive involvement of the skull base (Fig 12) and, exceptionally, can extend out into the infratemporal fossa.

 

Figure 8. Sagittal T1 weighted unenhanced image of a macroadenoma. The sella is enlarged and the macroadenoma is seen to extend upwards into the suprasellar cistern with the optic chiasm stretched and deformed over the surface of the macroadenoma.

Figure 9. Coronal T1 weighted unenhanced image of a macroadenoma. The sella is enlarged and the macroadenoma is seen to extend upwards into the suprasellar cistern with the optic chiasm stretched and deformed over the surface of the macroadenoma.

Figure 10. A coronal T1 weighted enhanced image shows diffuse enhancement of the macroadenoma, no normal pituitary tissue can now be identified. The optic chiasm is easily identified, it does not enhance and is seen to be stretched over the superior aspect of the adenoma.

Figure 11. A coronal enhanced T1 weighted image shows a right-sided pituitary adenoma that has invaded laterally into the right cavernous sinus. A significant component of the tumor lies lateral to the cavernous carotid artery and must therefore be within the cavernous sinus.

Figure 12. A sagittal T1 weighted unenhanced image shows an invasive macroadenoma which has involved much of the central skull base. It has invaded the sphenoid air sinus which can no longer be identified and has also extended down the clivus. There is also a modest suprasellar extension with elevation of the optic chiasm.

 

When pituitary macroadenomas attain a certain size, it is no longer possible to identify any normal pituitary tissue within the sella. The posterior pituitary bright spot may also be difficult to identify but is often seen in the lower aspect of the pituitary stalk which itself may be markedly deformed by the adenoma. Despite these appearances it is very rare for patients with macroadenomas of the pituitary to have diabetes insipidus, which usually points to a different pathology within the pituitary gland or stalk. Macroadenomas may be homogeneous or heterogeneous in their MR signal characteristics. Areas of cystic change and focal areas of hemorrhage are not infrequently identified. It is not uncommon for areas of hemorrhage to be seen within macroadenomas without any correlating clinical event; however, significant hemorrhage with necrosis in an adenoma can produce the syndrome of pituitary apoplexy. The patient reports sudden onset of headache usually associated with visual disturbance. The MRI scan shows an enlarged sella containing a macroadenoma with areas of high T1 signal representing the hemorrhage (Figs 13 and 14). Often there is suprasellar extension and there may also be involvement of the cavernous sinus. Clinically, the patient may present with cranial nerve problems related to involvement of the cavernous sinus. Hemorrhage extending outside the tumor into the subarachnoid space is documented but is very rare.

 

Figure 13. Sagittal T1 non-contrast image showing hemorrhage into an existing pituitary macroadenoma. The area of high signal represents the recent hemorrhage. There is a component of the tumor extending into the left cavernous sinus which does not show hemorrhage. The suprasellar extension is compressing the chiasm particularly on the right side.

Figure 14. Coronal non-contrast image showing hemorrhage into an existing pituitary macroadenoma. The area of high signal represents the recent hemorrhage. There is a component of the tumor extending into the left cavernous sinus which does not show hemorrhage. The suprasellar extension is compressing the chiasm particularly on the right side.

Pituitary Microadenomas

 

Pituitary microadenomas are confined within the sella and are sometimes identified within the normal pituitary gland as an area of lower signal on T1 weighted sequences than the normal pituitary tissue (Fig 15). (7) Local remodeling of the floor of the sella (Fig 5) and remodeling of the dorsum are also useful features to identify the presence of a microadenoma. Although there may be displacement of the pituitary stalk by a lesion, this is not a very reliable indicator (8). Administration of intravenous gadolinium will improve the sensitivity of pituitary MR in identifying the presence of a microadenoma; typically, the microadenomas enhance less avidly than the normal anterior pituitary tissue (Figs 5-7) Acquiring sequences dynamically within the first minute after intravenous injection can slightly further improve the sensitivity of the study (5). This will demonstrate adenomas that appear less vascular on the initial arterial phase of contrast enhancement but then equilibrate to show similar vascularity to the normal gland in the next few minutes. The use of volumetric T1W sequences after contrast (as compared to standard T1W spin echo -SE) improves the detection rate for microadenomas (9), and so is of particular use in Cushing’s disease. A fluid attenuated Inversion recovery (FLAIR) sequence after contrast may further improve the detection of Cushing’s adenomas, if used with the volumetric sequence, which has the highest individual sensitivity (10). The signal characteristics of GH-secreting adenomas on T2 weighted sequences (whether they are of lower or higher signal than adjacent grey matter) can be an indicator of their histological composition: densely granulated GH secreting adenomas are typically hypointense whilst sparsely granulated GH secreting adenomas are hyperintense. The low T2 signal (densely granulated) adenomas seemingly respond better to somatostatin analogues (11) (Fig 16).

 

Figure 15. A coronal unenhanced T1 weighted image shows a small left sided microadenoma (arrow) as an area of lower signal than the rest of the anterior pituitary tissue.

Figure 16. A coronal T2 weighted image shows a low T2 signal adenoma in the left side of the gland (arrow) which represented a densely granulated GH secreting adenoma.

 

After pituitary surgery it takes around 3 to 4 months for the postoperative changes within the sella to regress to allow for assessment of the true volume of residual pituitary tissue (12).

 

RATHKE’S CLEFT CYSTS

 

These benign, non-functioning cysts arise from remnants of squamous epithelium from Rathke’s cleft. They typically arise close to the insertion of the stalk (Fig 17) They are a common incidental finding reported in up to 11% of pituitary glands at autopsy (13). On imaging they may appear proteinaceous rendering their signal higher to the point that they appear hyperintense on the T1 weighted sequence. It is not uncommon for these cysts to lie in the suprasellar region on the surface of the gland where they are usually found anterior to the pituitary stalk (Figs 18 and 19). If they are of significant size, then it may be hard to distinguish a Rathke’s cyst from a cystic craniopharyngioma. However, it is notable that the wall of a Rathke’s cyst shows no more than minimal enhancement. There are no solid enhancing areas and they do not calcify. Rathke’s cysts can be found in all age groups.

 

Figure 17. A coronal unenhanced T1 weighted image demonstrating a small Rathke’s cyst. This is seen to lie just below the insertion of the pituitary stalk and centrally within the gland. Although it is possible that a small cystic adenoma could have these appearances, this is a very typical location for a Rathke’s cyst.

Figure 18. Sagittal enhanced T1 weighted image of a large Rathke’s cyst. The cyst is seen to be sitting on the superior aspect of the pituitary tissue which appears flattened within the sella. The optic chiasm is stretched over the surface of the cyst. After contrast the cyst does not show enhancement. The cyst shows higher signal than the CSF indicating that it has a higher protein content.

Figure 19. Coronal unenhanced T1 weighted image of a large Rathke’s cyst. The cyst is seen to be sitting on the superior aspect of the pituitary tissue which appears flattened within the sella. The optic chiasm is stretched over the surface of the cyst. After contrast the cyst does not show enhancement. The cyst shows higher signal than the CSF indicating that it has a higher protein content.

 

MENINGIOMAS

 

Meningiomas are slow-growing neoplasms that arise from the dura and can arise from any of the dural surfaces around the sella. Suprasellar meningiomas appear as soft tissue mass lesions apparently sitting on the superior surface of the pituitary and often compressing or involving the optic chiasm and nerves (Fig 20) (14) A typical feature is a dural “tail” of enhancement seen extending forward along the planum sphenoidale (Fig 21). Meningiomas have very similar signal to both the brain parenchyma and the pituitary gland, but show prominent homogeneous enhancement after contrast. The dural tail is a helpful distinguishing feature to separate meningiomas from other sella tumors.

 

Figure 20. Coronal enhanced T1 weighted image There is a suprasellar meningioma which appears to be arising from the planum sphenoidale and is seen extending along the floor of the anterior cranial fossa where a dural tail (arrow) is visible. The pituitary gland itself appears normal and a cleft of CSF is visible between the meningioma and the pituitary tissue. The meningioma is displacing and possibly involving the optic nerves anterior to the chiasm.

Figure 21. Sagittal (B) enhanced T1 weighted image. There is a suprasellar meningioma which appears to be arising from the planum sphenoidale and is seen extending along the floor of the anterior cranial fossa where a dural tail (arrow) is visible. The pituitary gland itself appears normal and a cleft of CSF is visible between the meningioma and the pituitary tissue. The meningioma is displacing and possibly involving the optic nerves anterior to the chiasm.

 

Meningiomas can also arise from the cavernous sinus producing a soft tissue mass lying lateral to normal pituitary tissue. The meningioma may encase the cavernous carotid artery and can produce constriction of this vessel (Fig 22). This feature may be helpful in distinguishing between cavernous sinus extension of a pituitary tumor and a cavernous sinus meningioma. Pituitary tumors do not typically cause any vascular constriction. There may be hyperostosis (bony thickening) of any bony surface from which a meningioma has arisen, most typically the anterior clinoid process, though this may be more easily appreciated on CT than MR.

 

Figure 22. A Coronal T1 weighted enhanced image of a left cavernous sinus meningioma. There is expansion of the left cavernous sinus (arrows) and some concentric narrowing of the left cavernous carotid artery (compare to the right carotid).

 

CRANIOPHARYNGIOMAS

 

Craniopharyngiomas are rare epithelial tumors arising in the sella/suprasellar region from the remnants of the craniopharyngeal duct. They are most commonly seen in children between 5 and 10 years of age; however, they can also occur in late adulthood in the sixth decade. They are the commonest lesion to involve the hypothalamic/pituitary region in children. In children it is the adamantinomatous histological subtype that is the most common (15). These shows both cystic and solid components as well as calcification, best appreciated on CT. The fluid within the cysts contains a high content of cholesterol as well as protein and desquamated cells, and this accounts for the cyst fluid often having high signal on T1 weighted unenhanced images (Fig 23) The solid enhancing tumor components are most easily seen after contrast injection (Fig 24). Craniopharyngiomas presenting in adulthood are more likely to be of the papillary subtype. These may be exclusively solid lesions (Fig 25 and 26) or show a mixed solid and cystic morphology (15). They do not typically calcify, and they are less locally infiltrative. The adamantinomatous subtype is particularly adherent to surrounding brain tissue and is therefore difficult to surgically resect; however, imaging cannot distinguish accurately between the histological subtypes. The imaging characteristics are those of a complex suprasellar mass containing both cystic and solid components, the solid components show enhancement after contrast (14). Large lesions may be associated with hydrocephalus (more commonly in children) while the cystic components may show variable signal characteristics from low signal to high signal on the T1 weighted sequences. (Fig 23).

 

Figure 23. Sagittal T1 weighted unenhanced image showing a partially cystic craniopharyngioma. This is a large complex suprasellar mass which extends down into the pituitary fossa and up to deform the third ventricle. There are patchy areas of high signal before contrast which represent the cystic components with a high protein/ lipid content.

Figure 24. Coronal enhanced image showing a partially cystic craniopharyngioma. This is a large complex suprasellar mass which extends down into the pituitary fossa and up to deform the third ventricle. It is of mixed signal intensity and the solid components enhance after contrast.

Figure 25. Sagittal T1 weighted unenhanced image. This shows a solid suprasellar enhancing mass, a solid craniopharyngioma. This is elevating and deforming the chiasm but is not involving the pituitary gland itself which can be seen within the sella.

Figure 26. Sagittal T1 weighted enhanced image. This shows a solid suprasellar enhancing mass, a solid craniopharyngioma. This is elevating and deforming the chiasm but is not involving the pituitary gland itself which can be seen within the sella. The mass enhances after contrast indicating that it is solid, and no cystic areas are evident.

 

HYPOTHALAMIC/OPTIC CHIASM GLIOMAS

 

These tumors present as suprasellar mass lesions and are most commonly seen in children. It is very often hard to determine whether the tumor has arisen in the optic chiasm or the hypothalamus as typically both of these structures are involved. These tumors are often associated with neurofibromatosis type I, in which case there is often involvement of the optic nerves and very often a significant cystic component is evident (14). Otherwise, these tumors appear as well-defined suprasellar mass lesions which may show enhancement (Fig 27 and 28) and may also show reactive signal change in the brain along the optic radiation. Calcification is not seen in these lesions.

 

Figure 27. Sagittal T1 weighted enhanced image show a partially cystic and partially solid hypothalamic glioma in the suprasellar region. The mass is centered on the region of the optic chiasm and is flattening the pituitary stalk and hypothalamus. The pituitary gland is normal.

 

Figure 28. T1 weighted coronal enhanced image show a partially cystic and partially solid hypothalamic glioma in the suprasellar region. The mass is centered on the region of the optic chiasm and is flattening the pituitary stalk and hypothalamus. The pituitary gland is normal.

GERMINOMA

 

These are rare intracranial germ cell tumors which most typically arise in the suprasellar or pineal regions (16,14). They are usually seen in children or young adults. These patients commonly have diabetes insipidus as one of the presenting complaints, and this reflects the involvement of the hypothalamus and pituitary stalk. Characteristic imaging appearances are those of a homogeneous, solidly enhancing mass involving the hypothalamic region with involvement of the upper aspect of the pituitary stalk (Figs 29 and 30) If the lesions are large then the optic chiasm may be also involved. Involvement in both the suprasellar region and the pineal region at the time of presentation is well recognized (16) (Fig 29). Detection of tumor markers, either human chorionic gonadotrophin (HCG) or alpha fetoprotein (AFP) can be detected in the serum/CSF to confirm the diagnosis (16), and may obviate the need for biopsy. The absence of these markers does not exclude the diagnosis and biopsy may be necessary. Dramatic resolution of the imaging findings after treatment can be seen as these tumors are very sensitive to radiation. These tumors can disseminate through the CSF spaces producing enhancement around the ventricular margins.

 

Figure 29. Sagittal T1 weighted enhanced images of a germinoma. Figure demonstrates a large homogeneously enhancing soft tissue mass within the suprasellar region involving the upper aspect of the pituitary stalk and the hypothalamic region with a second separate mass visible in the region of the pineal gland - this is typical of a germinoma.

Figure 30. Sagittal T1 weighted enhanced images of a germinomas. Figure demonstrates a small lesion in the region of the upper aspect of the stalk and hypothalamus (arrow).

 

GRANULAR CELL TUMORS

 

Granular cell tumors are rare, benign WHO grade 1 lesions most typically seen in the suprasellar region. They arise from the neurohypophysis and/or the pituitary stalk. They have also been known as choristomas (17) .Histologically, they appear to arise from the pituicyte which is the main posterior pituitary cell. These are seen as well-defined suprasellar masses related to the pituitary stalk with homogeneous enhancement (Fig 31) (17). Despite their intimate relationship to the pituitary stalk, diabetes insipidus is not usually present although other endocrine disturbance may be present and these lesions may even be asymptomatic. If they have reached significant size, they may be associated with visual disturbance or headache. They are benign and slow growing with a low recurrence rate after surgery. Similar in appearance to the granular cell tumor, and equally rare, are the so called pituicytoma and the spindle cell oncocytoma. It seems that histologically these 3 entities show morphological overlap and may arise from a common lineage: they cannot be distinguished on imaging appearances (18).

 

 

Figure 31. Sagittal enhanced T1 weighted image shows a well-defined enhancing mass within the pituitary stalk. The pituitary gland is normal, as is the hypothalamus. This was a granular cell tumor.

 

LANGERHANS CELL HISTIOCYTOSIS

 

This is a rare disease of childhood due to a proliferative disorder of the Langerhans cell of the dendritic cell line; CNS involvement is common but it is rarely the only site involved, skeletal involvement being the most frequent (19). A common location for intracranial involvement is the hypothalamo-pituitary axis. The MRI appearances characteristically show thickening of the pituitary stalk with enhancement (Fig 32). The posterior pituitary bright spot is usually absent. These appearances correlate with the typical clinical presentation of diabetes insipidus. This is often an isolated intracranial abnormality but extra-axial/osseous intracranial mass lesions and degenerative changes in the cerebellum and basal ganglia are also recognized (19).

 

Figure 32. Sagittal T1 enhanced image shows diffuse thickening of the pituitary stalk in a patient with Langerhans cell histiocytosis.

 

HYPOPHYSITIS

 

Hypophysitis is an inflammatory disorder of the pituitary gland which can be primary or secondary to a known infection or systemic disease. Primary hypophysitis includes autoimmune hypophysitis and other inflammatory forms of unknown cause. Distinct histopathological types are recognized; lymphocytic hypophysitis, granulomatous hypophysitis and xanthomatous hypophysitis (20). Lymphocytic and granulomatous hypophysitis have similar indistinguishable MR imaging features: enlargement of the gland producing the appearance of a sellar mass lesion with suprasellar involvement of the stalk (Figs 33 and 34). The gland may appear heterogeneous after contrast and there may be distortion of the chiasm if the suprasellar component is sizeable, which is not uncommon. Lymphocytic hypophysitis is the most common form occurring more often in females and classically presenting at the end of pregnancy or in the post-partum period with endocrine dysfunction (20) There may be also be diabetes insipidus which may correlate with loss of the posterior pituitary bright spot.

 

A variant of this condition is the so-called lymphocytic infundibulo-neurohypophysitis where the inflammatory process selectively involves the pituitary stalk and the posterior pituitary tissue, and imaging reflects that showing enlargement of the posterior gland and stalk (21).

 

Figure 33. Sagittal T1 weighted enhanced image of lymphocytic hypophysitis. There is slight enlargement and heterogeneity of the gland with thickening of the lower part of the stalk. The fossa is not enlarged.

Figure 34. Coronal T1 weighted enhanced image of lymphocytic hypophysitis. There is slight enlargement and heterogeneity of the gland with thickening of the lower part of the stalk. The fossa is not enlarged.

 

Secondary hypophysitis can be a reaction to a local process (e.g., infection or cyst rupture) or due to systemic disease, neoplastic processes or drugs. IgG4 plasma cell hypophysitis (22) and hypophysitis caused by the immune checkpoint inhibitors (ICPi) used in oncology (23) are now well described.

 

The cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) agents (e.g., ipilimumab) and the programmed death 1 (PD-1) agents (e.g., nivolumab) can both cause hypophysitis, but the CTLA-4 agents more frequently do so, and the combination of CTLA-4 and PD-1 carries the highest risk of hypophysitis (23). Ipilimumab-related hypophysitis has been reported in up to 11% of patients and is commoner in men (24). ICPi-related hypophysitis usually occurs within several weeks to months of starting therapy and manifests with headache, fatigue and anterior pituitary hormonal deficiency. Diabetes Insipidus is very rare (23).

 

MR typically shows a diffuse enlargement of the gland (more rarely the stalk or the gland plus stalk) which regresses after discontinuation of treatment. Enlargement that involves the chiasm is rare (21). Pituitary function may not recover despite the imaging normalization (Fig 35). As many as 23% of cases may have a normal MR despite clinical evidence of drug-induced hypophysitis (25).

 

Figure 35. Coronal T1 weighted enhanced image of ipilimumab-induced hypophysitis show diffuse enlargement of the gland with slightly heterogenous enhancement.

 

IgG4 related hypophysitis is one manifestation of IgG4 related disease. In around 40% of cases it may be an isolated manifestation, but in the majority of cases there is multi-organ involvement, most commonly lung or retroperitoneum (26). Clinical symptoms can relate to the sella mass effects, central diabetic insipidus, or anterior hypopituitarism. In distinction to drug-related hypophysitis, there is a high incidence of diabetes insipidus. A recent case series of 76 cases (26) identified a pituitary mass in 22.4%, a thickened stalk in 26.3%, while 51.3% had both a pituitary mass and a thickened stalk. As it is a fibro-inflammatory process the involved pituitary/ stalk may show low T2W signal (Figs 36 and 37).

 

A paraneoplastic syndrome with Anti-Pit 1 antibodies is a recognized cause of hypophysitis (with no specific radiological feature from other causes of hypophysitis). Systemic disease such as Granulomatosis with polyangiitis can occasionally involve the pituitary gland, the low T2 signal seen in IgG4 disease (Fig 37) would also be a feature of this type of granulomatous involvement

 

Figure 36. Sagittal enhanced T1 weighted image shows a diffusely enlarged gland and enlarged stalk, of low T2 signal -see Fig 37- due to IgG4 hypophysitis.

Figure 37. Coronal T2 weighted image shows a diffusely enlarged gland and enlarged stalk, of low T2 signal due to IgG4 hypophysitis.

 

All forms of hypophysitis can ultimately result in hypopituitarism with a small volume pituitary gland on imaging (Figs 38 and 39).

 

Figure 38. Sagittal enhanced T1 weighted images show a diffusely enlarged gland and enlarged stalk due to hypophysitis.

Figure 39. Sagittal enhanced T1 one year after hypophysitis has resulted in a small volume pituitary gland.

 

Granulomatous hypophysitis is less common and is found equally in males and females. MR appearances are the same as those described for lymphocytic hypophysitis (20).

 

Xanthomatous hypophysitis is the least common form and is characterized by a distinctly cystic appearance on imaging. An intrasellar cyst without suprasellar extension or involvement of the stalk is the most recognized appearance (20).

 

THE EMPTY SELLA

 

An empty sella contains only CSF without any visible pituitary tissue (anterior or posterior). The pituitary stalk will be visible and typically extends down to the floor of the sella (Figs 40 and 41). An empty sella may be the result of previously documented pituitary/hypothalamic pathology or treatment (e.g., surgery, radiotherapy, hypophysitis, Sheehan’s syndrome or pituitary apoplexy). However, it may also be discovered incidentally during MR scanning or in the course of investigating a new endocrine problem, then referred to as a “primary empty sella” (27). A partially empty sella implies that some residual pituitary tissue can still be seen along the floor of the fossa. In the case of a primary empty sella it is thought that a defect in the diaphragma sella (the sheet of dura over the surface of the sella through which the stalk passes) has allowed increased communication with the pulsatile CSF in the subarachnoid space. Primary empty sella is more common in women and has been associated with intracranial hypertension, obesity, visual disturbance, and spontaneous CSF leaks. Endocrine problems may be seen in up to 25% of cases with a primary empty sella (27), the majority within this group being specifically investigated for a suspected endocrine abnormality. However, a small proportion of patients found to have an empty sella unexpectedly may have endocrine abnormalities on detailed testing.

 

Figure 40. Sagittal enhanced T1 weighted image shows an empty sella. No pituitary tissue is visible, and the stalk extends down to the floor of the sella. The optic chiasm has prolapsed inferiorly, a not uncommon appearance after a large mass has been removed.

Figure 41. Coronal enhanced T1 weighted image shows an empty sella. No pituitary tissue is visible, and the stalk extends down to the floor of the sella. The optic chiasm has prolapsed inferiorly, a not uncommon appearance after a large mass has been removed.

 

ARACHNOID CYSTS

 

Arachnoid cysts are benign, congenital CSF-containing cysts that arise within the arachnoid membrane. They can be found in the suprasellar region where they may displace and distort the chiasm and stalk and/or flatten the pituitary tissue (Figs 42 and 43). They will always show the same signal as CSF on all MR imaging sequences, do not show any enhancement after contrast, and do not calcify.

 

Figure 42. Sagittal unenhanced T1 weighted image shows an arachnoid cyst in the suprasellar region. This is markedly elevating the hypothalamus and stretching the pituitary stalk.

Figure 43. Coronal unenhanced T1 weighted images show an arachnoid cyst in the suprasellar region with elevation of the right side of the chiasm by the cyst. No cyst wall is evident and the pituitary tissue itself is normal.

 

CONGENITAL PITUITARY ABNORMALITIES

 

 Congenital hypopituitarism can manifest as isolated growth hormone deficiency (IGHD) or combined pituitary hormone deficiency (CPHD), which can be related to anatomical abnormalities of the hypothalamic/pituitary structures on MR imaging. Imaging features include an ectopic location to the posterior pituitary which is seen to be undescended and is visible as a high signal area in the region of the median eminence (Fig 44). There may be absence of the pituitary stalk and/or hypoplasia of the anterior pituitary tissue. Other congenital abnormalities of midline structures may be associated with these hypothalamic/pituitary features: optic nerve hypoplasia, absence of the septum pellucidum, and corpus callosum abnormalities (28). CPHD is more often associated with callosal and stalk abnormalities than the milder forms of hypopituitarism such as IGHD (28). There are many genetic associations with both IGHD and CPHD, covered in detail in other sections.

 

Figure 44. Sagittal T1 weighted unenhanced image. This demonstrates a small pituitary fossa containing a reduced volume of anterior pituitary tissue. The stalk cannot be seen and the posterior pituitary bright spot (arrow) is lying in an ectopic location within the hypothalamus.

 

An incidental but uncommon imaging finding is that of a lipoma in the suprasellar cistern (Fig 45). This is identified as an entirely high T1 signal mass adjacent to normal hypothalamic and pituitary structures. The fatty nature of the mass can be confirmed with a fat-saturation sequence which will obliterate the signal from the mass.

 

Figure 45. Sagittal T1 weighted unenhanced image. There is a high signal mass lying just below the hypothalamus and behind the pituitary stalk in the suprasellar cistern. This is a lipoma. The pituitary tissue and stalk are normal.

 

HYPOTHALMIC HAMARTOMAS

 

These are benign developmental mass lesions that arise in the tuber cinereum of the hypothalamus and can be associated with central precocious puberty, gelastic (laughing) seizures, and sometimes developmental delay. The lesions are easily identified on MR imaging as they have a distinctive appearance as an almost pedunculated rounded mass hanging from the hypothalamus. They are of similar signal to the grey matter of the brain and do not show enhancement after contrast injection (Fig 46). Histologically, they are composed of well differentiated neurons and glial cells. They are sometimes approached surgically if the epilepsy is proving refractory to treatment.

 

Figure 46. Sagittal T1 weighted enhanced image of a hypothalamic hamartoma. There is a non-enhancing mass visible arising from the under surface of the hypothalamus and lying behind the pituitary stalk.

 

SARCOID

 

CNS involvement is seen in about 25% of cases of sarcoid (29) but many of these are subclinical. Neurosarcoid has a wide spectrum of intracranial imaging appearances including high T2 lesions in the white matter, meningioma-like dural masses, optic nerve lesions, and leptomeningeal enhancement (30). Involvement of the pituitary stalk/hypothalamus is one manifestation and may be part of more widespread leptomeningeal enhancement or may be isolated (Figs 47 and 48). If there is widespread leptomeningeal enhancement then appearances may be indistinguishable from TB meningitis (30)

 

Figure 47. Sagittal T1 weighted enhanced images show thickening of the pituitary stalk and nodular enhancement of the right side of the chiasm in a patient with neurosarcoid.

 

Figure 48. Coronal T1 weighted enhanced images show thickening of the pituitary stalk and nodular enhancement of the right side of the chiasm in a patient with neurosarcoid.

 

SKULL BASE TUMORS

 

Tumors of the central skull base may involve the sella or parasellar region and are in the differential diagnosis of any large mass around the sella which has significant bony involvement. Primary tumors occurring in this region are the chordoma and the chondrosarcoma (14) Chordomas arise from remnants of the primitive notochord and are seen as well-defined, centrally-located, expansile masses arising from the clivus. They show distinctive high signal on T2 weighted images and bony destruction on CT (Fig 49). Chondrosarcomas (malignant mesenchymal tumors) have a similar appearance but tend to arise just lateral to the clivus at the site of the petroclival synchondrosis (Fig 50 and 51); however, it is often not possible to distinguish between these two tumor types radiologically (31). Any destructive bony malignancy such as metastatic disease or plasmacytoma may involve the central skull base.

 

Figure 49. Sagittal CT reconstruction shows a destructive bony lesion in the clivus – a chordoma, the sella itself is preserved.

Figure 50. Axial T2 image of a chondrosarcoma. This shows the typical bubbly high T2 signal and involves the right side of the pituitary fossa, the right cavernous sinus and extends into the right middle cranial fossa.

Figure 51. Axial-enhanced T1 weighted image of a chondrosarcoma. This shows the typical bubbly high T2 signal and involves the right side of the pituitary fossa, the right cavernous sinus and extends into the right middle cranial fossa.

 

ECCHORDOSIS PHYSALIPHORA

 

Ecchordosis physaliphora is a benign, congenital, hamartomatous notochord remnant usually found in the retroclival region. They are typically small, asymptomatic and can be found in up to 2 % of autopsies.

 

They are histologically identical to chordomas, which are typically sizeable and present with brainstem or cranial nerve compression.

 

On MR imaging they lie in the retroclival, prepontine region, are similar signal to CSF, notably high signal on T2 sequences (Fig 52) and show variable (usually none to little) enhancement. CT classically shows a well-defined, non-aggressive clival bony defect. A bony stalk may be seen, and this is considered pathognomonic (Fig 53). Although previously considered a distinct clinical entity they are now considered to lie at the benign end of a pathological spectrum that extends to chordomas (32).

 

Fig 52. Axial T2 volumetric scan shows a small nodule of T2 hyperintense tissue extending through a well-defined midline bony defect in the clivus(arrow).

Fig 53. Axial CT of the skull base shows a well-defined cortical defect (arrow) in the clivus with a small bony spur just to the Rt of the defect (not the same case as image 51).

 

METASTASES

 

Metastatic spread of systemic tumors to the pituitary gland/stalk is relatively uncommon and is usually seen in the context of diffuse malignancy. Breast and lung are the commonest primary tumors to do this although other primary tumors have been reported in the literature (33,14). A clinical presentation with diabetes insipidus reflects the involvement of the stalk in many cases. Diffuse enlargement of the gland with thickening of the stalk (Fig 54) in a patient with known malignancy might suggest this diagnosis.

 

 

Figure 54. Coronal enhanced T1 weighted image shows a diffusely enlarged pituitary gland with bilateral enlargement of both cavernous sinuses and some thickening of the stalk, this was metastatic disease in a patient with breast carcinoma.

 

REFERENCES

 

  1. Dietrich RB, Lis LE, Greensite FS, Pitt D. Normal MR appearance of the pituitary gland in the first 2 years of life. Am J Neuroradiol 1995;16:1413-1419
  2. Tsunoda A, Okuda O, Sato K. MR height of the pituitary gland as a function of age and sex: especially physiological hypertrophy in adolescence and in climacterium. Am J Neuroradiol 1997;18:551-554
  3. Dinc H, Esen F, Demirci A, Sari A, Resit Gumele H. Pituitary dimensions and volume measurements in pregnancy and post partum. MR assessment. Acta Radiol 1998;39:64-69
  4. Steiner E, Wimberger D, Imhof H, Knosp E, Hajek P. [Gd-DTPA in the MR diagnosis of pituitary adenomas]. Rofo 1989;150:323-327
  5. Kucharczyk W, Bishop JE, Plewes DB, Keller MA, George S. Detection of pituitary microadenomas: comparison of dynamic keyhole fast spin-echo, unenhanced, and conventional contrast-enhanced MR imaging. Am J Roentgenol 1994;163:671-679
  6. Knosp E, Steiner E, Kitz K, Matula C. Pituitary adenomas with invasion of the cavernous sinus space: a magnetic resonance imaging classification compared with surgical findings. Neurosurgery 1993;33:610-617; discussion 617-618
  7. Kucharczyk W, Davis DO, Kelly WM, Sze G, Norman D, Newton TH. Pituitary adenomas: high-resolution MR imaging at 1.5 T. Radiology 1986;161:761-765
  8. Ahmadi H, Larsson EM, Jinkins JR. Normal pituitary gland: coronal MR imaging of infundibular tilt. Radiology 1990;177:389-392
  9. Patronas N, Bulakbasi N, Stratakis CA, Lafferty A, Oldfield EH, Doppman J, Nieman LK. Spoiled gradient recalled acquisition in the steady state technique is superior to conventional postcontrast spin echo technique for magnetic resonance imaging detection of adrenocorticotropin-secreting pituitary tumors. J Clin Endocrinol Metab 2003;88:1565-1569
  10. Chatain GP, Patronas N, Smirniotopoulos JG, Piazza M, Benzo S, Ray-Chaudhury A, Sharma S, Lodish M, Nieman L, Stratakis CA, Chittiboina P. Potential utility of FLAIR in MRI-negative Cushing's disease. J Neurosurg 2018;129:620-628
  11. Heck A, Ringstad G, Fougner SL, Casar-Borota O, Nome T, Ramm-Pettersen J, Bollerslev J. Intensity of pituitary adenoma on T2-weighted magnetic resonance imaging predicts the response to octreotide treatment in newly diagnosed acromegaly. Clin Endocrinol (Oxf) 2012;77:72-78
  12. Kremer P, Forsting M, Ranaei G, Wuster C, Hamer J, Sartor K, Kunze S. Magnetic resonance imaging after transsphenoidal surgery of clinically non-functional pituitary macroadenomas and its impact on detecting residual adenoma. Acta Neurochir (Wien) 2002;144:433-443
  13. Teramoto A, Hirakawa K, Sanno N, Osamura Y. Incidental pituitary lesions in 1,000 unselected autopsy specimens. Radiology 1994;193:161-164
  14. Kaltsas GA, Evanson J, Chrisoulidou A, Grossman AB. The diagnosis and management of parasellar tumours of the pituitary. Endocr Relat Cancer 2008;15:885-903
  15. Sartoretti-Schefer S, Wichmann W, Aguzzi A, Valavanis A. MR differentiation of adamantinous and squamous-papillary craniopharyngiomas. Am J Neuroradiol 1997;18:77-87
  16. Packer RJ, Cohen BH, Cooney K. Intracranial germ cell tumors. Oncologist 2000;5:312-320
  17. Cohen-Gadol AA, Pichelmann MA, Link MJ, Scheithauer BW, Krecke KN, Young WF, Jr., Hardy J, Giannini C. Granular cell tumor of the sellar and suprasellar region: clinicopathologic study of 11 cases and literature review. Mayo Clin Proc 2003;78:567-573
  18. Mete O, Lopes MB, Asa SL. Spindle cell oncocytomas and granular cell tumors of the pituitary are variants of pituicytoma. The American journal of surgical pathology 2013;37:1694-1699
  19. D'Ambrosio N, Soohoo S, Warshall C, Johnson A, Karimi S. Craniofacial and intracranial manifestations of langerhans cell histiocytosis: report of findings in 100 patients. Am J Roentgenol 2008;191:589-597
  20. Gutenberg A, Hans V, Puchner MJ, Kreutzer J, Bruck W, Caturegli P, Buchfelder M. Primary hypophysitis: clinical-pathological correlations. Eur J Endocrinol 2006;155:101-107
  21. Abe T. Lymphocytic infundibulo-neurohypophysitis and infundibulo-panhypophysitis regarded as lymphocytic hypophysitis variant. Brain Tumor Pathol 2008;25:59-66
  22. Hsing MT, Hsu HT, Cheng CY, Chen CM. IgG4-related hypophysitis presenting as a pituitary adenoma with systemic disease. Asian journal of surgery / Asian Surgical Association 2013;36:93-97
  23. Chang LS, Barroso-Sousa R, Tolaney SM, Hodi FS, Kaiser UB, Min L. Endocrine Toxicity of Cancer Immunotherapy Targeting Immune Checkpoints. Endocr Rev 2019;40:17-65
  24. Faje AT, Sullivan R, Lawrence D, Tritos NA, Fadden R, Klibanski A, Nachtigall L. Ipilimumab-induced hypophysitis: a detailed longitudinal analysis in a large cohort of patients with metastatic melanoma. J Clin Endocrinol Metab 2014;99:4078-4085
  25. Caturegli P, Di Dalmazi G, Lombardi M, Grosso F, Larman HB, Larman T, Taverna G, Cosottini M, Lupi I. Hypophysitis Secondary to Cytotoxic T-Lymphocyte-Associated Protein 4 Blockade: Insights into Pathogenesis from an Autopsy Series. Am J Pathol 2016;186:3225-3235
  26. Li Y, Gao H, Li Z, Zhang X, Ding Y, Li F. Clinical Characteristics of 76 Patients with IgG4-Related Hypophysitis: A Systematic Literature Review. Int J Endocrinol 2019;2019:5382640
  27. Del Monte P, Foppiani L, Cafferata C, Marugo A, Bernasconi D. Primary "empty sella" in adults: endocrine findings. Endocr J 2006;53:803-809
  28. Mehta A, Hindmarsh PC, Mehta H, Turton JP, Russell-Eggitt I, Taylor D, Chong WK, Dattani MT. Congenital hypopituitarism: clinical, molecular and neuroradiological correlates. Clin Endocrinol (Oxf) 2009;71:376-382
  29. Pawate S, Moses H, Sriram S. Presentations and outcomes of neurosarcoidosis: a study of 54 cases. QJM 2009;102:449-460
  30. Lexa FJ, Grossman RI. MR of sarcoidosis in the head and spine: spectrum of manifestations and radiographic response to steroid therapy. Am J Neuroradiol 1994;15:973-982
  31. Almefty K, Pravdenkova S, Colli BO, Al-Mefty O, Gokden M. Chordoma and chondrosarcoma: similar, but quite different, skull base tumors. Cancer 2007;110:2457-2467
  32. Stevens AR, Branstetter BFt, Gardner P, Pearce TM, Zenonos GA, Arani K. Ecchordosis Physaliphora: Does It Even Exist? Am J Neuroradiol 2023;44:889-893
  33. Komninos J, Vlassopoulou V, Protopapa D, Korfias S, Kontogeorgos G, Sakas DE, Thalassinos NC. Tumors metastatic to the pituitary gland: case report and literature review. J Clin Endocrinol Metab 2004;89:574-580

 

 

Gastrointestinal Neuroendocrine Tumors and The Carcinoid Syndrome

ABSTRACT

 

Neuroendocrine neoplasms originating from the gut are increasingly diagnosed as a result of the rise in radiological and endoscopic procedures, improved pathological classification, and likely an increase in true incidence. The diffuse neuroendocrine gastrointestinal system can trigger cancer formation into a wide variety of neoplasm subtypes, ranging from well-differentiated tumors to poorly differentiated carcinomas. All gastrointestinal neuroendocrine neoplasms have the potential to metastasize and ultimately impair patient survival. In recent years, changes have occurred in the pathophysiological understanding, nomenclature, pathological grading, molecular imaging, and management options for these neuroendocrine neoplasms. This chapter will focus on well-differentiated neuroendocrine tumors of gastrointestinal origin, which find their origin at separate primary locations, all characterized by their specific clinical behavior. A minority of patients suffer from hormonal syndromes due to the secretion of peptides or amines from the neuroendocrine tumor. The carcinoid syndrome is the quintessential hormonal syndrome in gastrointestinal neuroendocrine tumors, particularly those of midgut origin. Patients suffering from the carcinoid syndrome have a reduced survival and quality of life, due to debilitating symptoms of flushing and diarrhea as well as fibrotic complications. We provide an overview of the background of gastrointestinal neuroendocrine tumors as well as the carcinoid syndrome and discuss the diagnostic pathways as well as treatment possibilities for patients presenting with this disease.

 

INTRODUCTION

 

Enteroendocrine cells constitute approximately 1-2% of all cells within the gastrointestinal tract. Quite similarly, neuroendocrine neoplasms (NEN) of the digestive tract form 1-2% of all malignancies in this organ system. When grouped together with pancreatic NEN (panNEN), gastroenteropancreatic (GEP) NEN are the second most common malignancy in the gut, surpassing esophagus, gastric, and pancreatic carcinomas in incidence rates (1). These tumors can arise anywhere along the primitive gut, but are most commonly detected in the small intestine or rectum. Based on histology, NENs are grouped into well-differentiated neuroendocrine tumors (NET) and poorly differentiated neuroendocrine carcinomas (NEC) (2). The former group was previously termed carcinoid tumors, based on the original observation by Siegfried Obendorfer (1876-1944) in 1907 that NETs of the small bowel displayed “carcinoma-like” or “carcinoid” features (3). As this term has led to the common misconception that carcinoid tumors are benign or always indolent, current correct nomenclature of this gastrointestinal malignancy solely uses the term NEN.

 

This chapter focuses on the clinical features, diagnosis, and management of the different well-differentiated NET along the gastrointestinal tract. The reader is referred to chapter “Diffuse hormonal systems” for lung NEN (4), where well-differentiated tumors are still termed typical or atypical carcinoids, and to chapter “Pathophysiology and treatment of pancreatic neuroendocrine tumors” for panNEN (5).

 

EPIDEMIOLOGY

 

NEN are historically considered a rare cancer type with an incidence of all subtypes combined of 5-10 per 100,000 persons per year (6). Two registry studies have shown that the incidence of NEN is rising several fold over the last decades. In the United States of America, NEN incidence increased 6.4-fold from 1.09 to 6.98 per 100,000 population per year between 1973 and 2012 (7), while in the United Kingdom rates rose 3.7-fold from 2.35 to 8.61 per 100,000 population per year between 1995 and 2018 (8). Within the GEP subtypes, small intestinal, pancreatic, and rectal NEN are most prevalent and have seen the clearest rising incidence rates. Part of the increased detection rate is caused by the rise in the absolute number of endoscopy procedures and radiological imaging, shifting the diagnosis more often towards incidentalomas. On the other hand, increased awareness among pathologists and improved classification likely also plays a role. The striking rise of NEN incidence compared to the stable incidence of all other malignant neoplasms in recent decades (7, 8) might suggest that a currently unknown epigenetic or environmental risk factor could stimulate NEN carcinogenesis.

 

The combination of increased detection as well as improved survival leads to an overall increase in NEN prevalence. The recent epidemiological data in the United Kingdom (8) suggest that NEN should not be considered a rare form of cancer anymore, as it comprised the 10th most prevalent cancer.

 

PATHOPHYSIOLOGY

 

Much is unknown about the pathogenesis of gastrointestinal NET (9). Besides the driver function of gastrin in two subtypes of gastric NET (see below), the causative factors for NET formation in the gut remain elusive. Genetic mutations have been identified as driving carcinogenesis across a wide array of malignancies, but – even in late, advanced stages of disease – NET remains among the tumor types with the lowest amount of tumor mutational burden or driver mutations (10). Contrarily, NEC show a high tumor mutational burden with gene mutations in well-known oncogenes or tumor suppressor genes, such as TP53, KRAS, RB1 (11). Dedicated studies of small intestinal NET genotypes with next generation sequencing have failed to detect prevalent mutations. The most commonly mutated gene in small intestinal NET, CDKN1B encoding cyclin-dependent kinase inhibitor p27, was found to be mutated in 10% of cases (12). Germline mutation in CDKN1B also cause the rare endocrine tumor syndrome multiple endocrine neoplasia type 4 (MEN4), which predisposes to the occurrence of gastric, duodenal, and pancreatic NET among other tumor types (13). Whole genome sequencing of synchronous multifocal small intestinal NET also failed to detect common genetic drivers, but instead observed clonal independency of tumors within individuals (14). No clear driver mutations have been identified for the other subtypes of gastrointestinal NET as well. Multiple endocrine neoplasia type 1 (MEN1) is besides primary hyperparathyroidism and pituitary NET primarily associated with the occurrence of pancreatic, bronchial, and thymic NET (15). However, duodenal NET can also arise within the context of MEN1 and these have a predilection to secrete gastrin, leading to gastrinoma or Zollinger-Ellison syndrome. This in turn stimulates secondary gastric NET formation (16). A genome-wide association study of 405 patients compared to more than 600,000 control subjects in two cohorts revealed an association between the occurrence of small intestinal NET and single nucleotide polymorphisms in 6 genes (17). The most interesting locus was of a missense mutation in the intestinal stem cell factor LGR5, suggesting a role for aberrant cellular differentiation in the development of small intestinal NET. Contrary to DNA mutations, chromosomal aberrations are prevalent in gastrointestinal NET. For small intestinal NET copy number variations have been frequently detected. The most prominent observed change is loss of chromosome 18 in up to 70% of cases, followed by losses in chromosomes 9, 11 and 16 and gains in chromosomes 4, 5, 14 and 20 (18). Whether these changes have a causative role in the development of small intestinal NET is currently unknown.

 

Due to the lack of obvious DNA changes contributing to NET pathogenesis, studies have investigated the role of epigenetics, e.g. changes to the chromatin that affect gene transcription without changing the DNA code (19). In the largest study to date in 97 patients with small intestinal NET, integrated genetic, epigenetic, and transcriptomic analysis detected 3 molecular subtypes, that differed in their survival outcome (20). DNA methylation analysis found that small intestinal NET were highly epigenetically dysregulated. The prognostically favorable molecular subgroup was associated with loss of chromosome 18, while another subgroup displayed no copy numbers alterations. NET in the molecular subgroup with inferior survival outcome displayed multiple copy number variations.

 

Because of the link between enteroendocrine cells and the bowel content, there have been speculations on carcinogenic factors in the bowel content. This could include but is not limited to dietary factors, microbial species, and microplastics. Further research is needed before a clear role can be identified for these factors.

 

COMMON FEATURES

 

NET of the gastrointestinal tract share many features owing to their collective origin from enteroendocrine cells. Originally described as APUD (amine precursor uptake and decarboxylation) tumors or APUDomas by Anthony Pearse (1916-2003) these neoplasms retain the potential to produce and secrete several hormonal substances in the form of amines and peptides (3, 21). These secretagogues are stored in intracellular dense-core secretory granules, which are released upon fusion with the plasma membrane. Gastrointestinal NET, like other types of NET, express markers specific for their neuroendocrine phenotype. The two most prevalent markers, synaptophysin and chromogranin A, form the basis for a immunohistochemical diagnosis of a NEN cell (22).

 

Stage

 

Similar to other cancers, NET are staged according to the TNM classification, which signifies key therapeutic and prognostic information (2), Table 1. Whereas stage I and II indicate local disease confined to the presence of the primary tumor (T1-4 N0 M0), stage III signifies the presence of regional spread to lymph node metastases (T1-4 N1 M0). Distant metastases (T1-4 N0-1 M1) are classified as stage IV disease.

 

Table 1. TNM staging of gastrointestinal neuroendocrine neoplasms according to the 8th edition of the AJCC Cancer Staging Manual (2018)

 

Stomach

Duodenum

Small intestine

Appendix

Colon and rectum

Tx

Primary tumor cannot be assessed

T0

No evidence of primary tumor

T1

Invades the lamina propria or submucosa and is ≤ 1 cm in size

Invades the lamina propria or submucosa or confined within the sphincter of Oddi and is ≤ 1 cm in size

Invades the lamina propria or submucosa and is ≤ 1 cm in size

Tumor ≤ 2 cm in size

Invades the lamina propria or submucosa and is ≤ 2 cm in size

  T1a

 

 

 

 

Tumor ≤ 1 cm in size

  T1b

 

 

 

 

Tumor > 1 and ≤ 2 cm in size

T2

Invades the muscularis propria or is > 1 cm in size

Tumor > 2 but ≤ 4 cm in size

Invades the muscularis propria or is > 2 cm in size

T3

Invades into the subserosa

Growth into the pancreas or peripancreatic adipose tissue

Invades into the subserosa

Tumor > 4 cm in size or invades into the subserosa or mesoappendix

Invades into the subserosa

T4

Invades into the (visceral) peritoneum or adjacent organs or structures

Nx

Regional lymph nodes cannot be assessed

N0

No regional lymph node metastasis has occurred

N1

Regional lymph node metastasis

Regional lymph node metastasis in < 12 nodes

Regional lymph node metastasis

 

N2

 

 

Large mesenteric masses (> 2 cm) or extensive nodal deposits (≥ 12)

 

 

 

 

M0

No distant metastasis

M1

Distant metastasis

M1a

Metastasis confined to liver

M1b

Metastasis in at least one extrahepatic site (e.g., lung, ovary, nonregional lymph node, peritoneum, bone)

M1c

Both hepatic and extrahepatic metastasis

 

 

 

 

 

 

 

 

Stage I

T1 N0 M0

 

Stage II

T2-3 N0 M0

 

 

Stage IIA

 

 

 

 

T2 N0 M0

Stage IIB

 

 

 

 

T3 N0 M0

Stage III

Any T N1 M0 or T4 N0 M0

 

 

Stage IIIA

 

 

 

 

 T4 N0 M0

Stage IIIB

 

 

 

 

Any T N1 M0

Stage IV

Any T any N M1

 

Grade

 

The biological behavior of the individual NEN is classified according to the tumor grade. NEN can display a wide array of biological behavior from generally very indolent taking years to significantly grow (e.g., appendix NET) to very aggressive inevitably leading to death (small cell lung NEC) (23). In order to predict prognosis and guide management all gastrointestinal NEN should be examined histologically for differentiation (well versus poorly differentiated), mitotic index (per 10 HPF), and ki67 index. The latter encompasses staining of the nuclear proliferation marker ki67 by the MIB1 antibody. Different grading cut-offs have been used in the past (24), but the WHO 2019 classification of digestive system tumors and 2022 classification of (neuro)endocrine tumors separate well-differentiated NET from poorly differentiated NEC on the basis of the histological phenotype (2, 25). In cases of an ambiguous entity, molecular analysis or staining of Rb1 and p53 can point towards the presence of a NEC (26).

 

NET are divided into grade 1, 2 and 3, whereas NEC by definition are grade 3. NET grading is discerned through the combination of mitotic and ki67 index, with the highest value counted (Table 2) (2, 25). Due to the differences in biological behavior, tumor grading is key to management of GI NET, especially in cases of metastatic and consequently incurable disease.

 

Table 2. Classification of gastrointestinal neuroendocrine neoplasms, according to 2022 WHO classification of endocrine and neuroendocrine tumors and 2019 WHO classification of tumors of the digestive system

Well-differentiated NEN

 

Ki67 proliferation index

Mitotes per 2 mm2

NET Grade 1

<3%

<2

NET Grade 2

3–20%

2–20

NET Grade 3

>20%

>20

Poorly differentiated NEN

Small cell NEC

Large cell NEC

>20%

>20

 

HORMONAL SYNDROMES IN NET

 

Due to their endocrine heritage, gastrointestinal NET can produce and secrete excessive amounts of hormonal substances, that can elicit clinical syndromes in patients (22). All patients presenting with a gastrointestinal NET should be examined by history taking and physical exam for the presence of a hormonal syndrome, as this has important therapeutic and prognostic consequences. In case of a suspected hormonal syndrome, appropriate biochemical analysis should be performed for the elevation of the causative hormonal peptides or amines (27).

 

Carcinoid Syndrome

 

The carcinoid syndrome is the most common hormonal syndrome encountered in gastrointestinal NET and even NEN in general. Estimations fluctuate that around 20% of patients with stage IV midgut NET suffer from carcinoid syndrome (28). It is mainly characterized by symptoms of secretory diarrhea and vasodilatory flushes. Occasionally, bronchospasms can also occur (29). In severe and long-standing cases carcinoid heart disease (CHD) can arise, characterized by plaque-like depositions in mainly right-sided heart valves and endocardium (30). Following acute stressors, some NET associated with carcinoid syndrome are able to secrete massive amounts of vasoactive compounds, leading to hemodynamic instability. This type of vasodilatory shock, also known as carcinoid crisis, can be defined as an acute onset of stressor-induced hemodynamic instability in patients with carcinoid syndrome and can be observed during the induction of anesthesia and after tumor lysis following embolization or peptide receptor radionuclide therapy (31).

 

The principal effector of carcinoid syndrome is thought to be the amine serotonin (5-hydroxytryptamine) (32), which is also secreted physiologically by several subtypes of neuroendocrine cells in the gut and lungs. A variety of preclinical and clinical studies support a central role of serotonin in the pathophysiology of carcinoid syndrome-related diarrhea and CHD, while its role in flushing in carcinoid syndrome patients is still controversial. Other hormonal substances postulated to contribute to the carcinoid syndrome include tachykinins, catecholamines, kallikrein and histamine (33).

 

Carcinoid syndrome predominantly arises in NET of midgut origin, comprised of jejunum, ileum, cecum, and ascending colon. This location specificity is presumably due to carcinogenesis within the enterochromaffin (EC) cell, which uses serotonin as its main secretagogue to communicate with the autonomous nervous system and influence bowel motility (4). This hormonal syndrome can also be encountered in bronchial NET (typical or atypical carcinoid) or NET of other origin (e.g., ovarian, pancreatic, unknown primary). Importantly, tumor seeding beyond the portal circulation is a prerequisite for carcinoid syndrome, as its causative hormones are inactivated by hepatocytes (34). For midgut NET, carcinoid syndrome thus hallmarks spread beyond locoregional disease, with liver metastases being present in more than 90% of cases. Alternatively, the tumor sites may secrete through the retroperitoneal or ovarian/testicular venous drainage, effectively bypassing the portal circulation and drain directly on the inferior caval vein.

 

The presence of carcinoid syndrome is a negative prognostic indicator, which is likely caused by its association with tumor bulk (28, 35). Within this spectrum, CHD is also associated with decreased survival in patients in univariate analyses (36). Because of these features carcinoid syndrome should be diligently investigated in all patients with NET and actively managed alongside antiproliferative therapy (see management section below).

 

Other Functioning Syndromes

 

Besides carcinoid syndrome, other NEN-associated hormonal syndromes are predominantly encountered in patients with a panNEN. Duodenal NET can in rare cases elicit hormonal syndromes that are also seen in pancreatic NET, such as gastrinoma (16), VIPoma (37), and somatostatinoma (38). Ectopic hormonal production has also been described in gastrointestinal NET in limited case reports. However, these functioning syndromes are more frequented encountered in pancreatic (ACTH, PTHrP, GHRH) or lung NET (SIADH, ACTH), see the Endotext chapter on Paraneoplastic syndromes related to Neuroendocrine Tumors (39).

 

PRIMARY NET LOCATIONS

 

Esophagus

 

Well-differentiated NET of the upper alimentary tract are extremely rare. The esophagus is a predilection place for the occurrence of NEC (40). Alternatively, mixed neuroendocrine-non neuroendocrine neoplasms (MiNEN) can be encountered in the esophagus, similar to other gastrointestinal sites. Formerly these tumors were designated as Mixed adeno-neuroendocrine carcinoma (MANEC). This aggressive tumor entity is comprised of both a NEN component (NET or NEC) as well as an adenocarcinoma component, with the latter being responsible for the prognostic outcome (2).

 

Stomach

 

The neuroendocrine cells in the stomach can give rise to several NEN subtypes, depending on the underlying pathophysiology. Central to understanding gastric NEN is the dependency of the histamine-producing enterochromaffin-like (ECL) cells on gastrin stimulation. Chronic hypergastrinemia due to several causes can lead to ECL cell hyperplasia and gastric NET formation, so called ECLoma. When an ECLoma occurs during compensatory gastrin elevations this is termed a type I gastric NET (41), accounting for 75-80% of gastric NEN. This is most commonly caused by atrophic gastritis due to antibodies against intrinsic factor or parietal cells, which is also causative for pernicious anemia. Alternatively, type I gastric NET have been described following Helicobacter pylori infection, chronic use of proton pump inhibitors, or mutations in the proton pump gene (ATP4A) and resulting hypergastrinemia (42-45). When ECLoma arise due to a gastrin-producing NET in the pancreas or duodenum (Zollinger-Ellison syndrome), these are termed type 2 gastric NET, which is responsible for 5% of all gastric NET cases. This pathology is generally restricted to patients with MEN-I and a duodenal gastrinoma (46). A well-differentiated gastric NET arising in the presence of normal fasting gastrin levels is termed a type 3 NET and accounts for approximately 15-20% of gastric NET. Some authors have proposed the rare gastric NEC as the type 4 gastric NEN (9), Table 3.

 

Table 3. Subtypes of Gastric Neuroendocrine Neoplasm

 

Hypergastrinemia, ECL cell hyperplasia

Growth

Features

Gastric NET type 1

Yes

Indolent

Secondary to atrophic gastritis, helicobacter pylori infection, proton pump inhibition or ATP4Amutation

Gastric NET type 2

Yes

Indolent

Secondary to gastrinoma (Zollinger Ellison syndrome)

Gastric NET type 3

No

Intermediate

Sporadic

Gastric NEC type 4

No

Aggressive

Sporadic

 

Biological behavior of the gastric NEN subtypes differs widely with generally indolent course for type 1 and 2 NET, which are predominantly grade 1 and can be characterized by multiplicity (47-49). Only a few metastatic cases have been reported in the literature, without clear evidence of impaired survival (50). Type 3 gastric NET and type 4 gastric NEC were previously considered as a single subtype, which was accompanied by a high rate of metastases and poor survival outcome. However, recent analyses show lower grade, metastatic potential, and better outcome of type 3 gastric NET than previously assumed (51, 52).

 

The vast majority of gastric NET is clinically non-functional, although ghrelin production has been described in NET presumably derived from gastric H cells, see Endotext chapter on Ghrelinoma (53).

 

Duodenum

 

A rare subtype of gastrointestinal NET, duodenal NET are often incidentally discovered during esophagogastroduodenoscopy (Figure 1A). They are characterized by intramural lesions which might sometimes only be visible on endoscopic ultrasound. Bleeding or ulceration is rare, but can be a presenting symptom (54). The majority of duodenal NET are localized and grade 1-2, particularly for tumors smaller than 1.0 cm. Metastatic potential increases with size and can be present at diagnosis or occur during follow-up (55, 56). Due to the nature of the neuroendocrine cells in the duodenum several hormonal syndromes can be encountered, such as gastrinoma or VIPoma. Somatostatin-expressing NET near the ampulla of Vater have been described as part of neurofibromatosis type 1 (57). Some of the larger duodenal NET cannot be effectively localized as originated from either duodenum or pancreas due to the overlapping anatomy.

 

Figure 1. Endoscopy in gastrointestinal NET. (A) Endoscopic image of a 5 mm submucosal lesion in the duodenal bulb. Fine needle aspiration confirmed a grade 1 duodenal NET, which was subsequently removed by endoscopic mucosal resection. (B) Endoscopic view of an 8 mm rectal NET, grade 1, which was successfully resected by endoscopic submucosal dissection.

 

Small Intestinal (Jejunum and Ileum)

 

The classic site for well-differentiated NET in the gastrointestinal tract is the small intestine, particularly the terminal ileum. NET are the most common malignancy in the small intestine, followed in incidence by adenocarcinoma and lymphoma (58). Almost all small intestinal NET are low to intermediate grade and can potentially show indolent growth (59). NEC of the small intestine are extremely rare. As EC cells are the predominant neuroendocrine cell in the small intestine, metastatic small intestinal NET are most often associated with the carcinoid syndrome (60).

 

At presentation, the majority of small intestinal NET are metastasized, with a predilection for lymph node and liver metastases (59). In some cases, the primary tumor cannot be visualized despite modern imaging techniques, such as PET/CT. Lymphogenic spread of small intestinal NET occurs locally within the mesentery. The finding of NET accompanied by a mesenteric mass hints towards a small bowel origin of the NET. Unique to small intestinal NET, mesenteric metastases can develop extensive fibrosis (Figure 2). This is seen on cross-sectional imaging as fibrotic strand radiating from a solid mesenteric mass, in a spoke-wheel pattern (61). This pathognomonic feature of small intestinal NET can lead to chronic bowel ischemia due to compression of venous drainage, leading to intermittent abdominal cramps or colicky pain, particularly after a large meal. Ultimately, ileus or bowel perforation can occur. In one study of 530 patients with small intestinal NET, mesenteric fibrosis was found to be progressive in 13.5% of cases with a median time to growth of 40 months, signifying slow progression (62). Although mesenteric fibrosis can lead to fatal complications and is associated with overall survival in univariate analysis, it was not associated with a worse overall survival in multivariate analysis (63).

 

Figure 2. Mesenteric fibrosis in midgut NET. (A) Transversal and (B) coronal plane contrast-enhanced CT images of a patient with a cecal NET and a mesenteric metastasis (arrow). A desmoplastic reaction consisting of fibrotic strands can be seen radiating from the mesenteric tumor mass, which can compromise venous blood flow from the bowel. The mass is partly calcified.

 

Hepatic metastases of small intestinal NET can be much larger than the primary tumor or lymph nodes. Even in the presence of extensive bilobar metastases, the function of the liver is often preserved, although isolated hyperammonemia due to shunting has been described in selected cases (64).

 

Appendix

 

In the majority of cases, appendix NET are incidentally encountered during appendectomy because of appendicitis. A contributory role of the potentially obstructive tumor has been attributed to the occurrence of appendicitis, but this has not been proven to date. Because of its association with appendicitis, appendix NET have a peak incidence in adolescents and young adults (65). Most appendix NET cases are confined to the appendix and have a favorable proliferation index (grade 1 or low 2). Development of lymph node metastases can be seen in up to 25% of appendix NET patients, whereas distant metastases are rare (66). Contrary to origin NET within the midgut, carcinoid syndrome is rarely encountered in appendix NET patients, potentially due to other cell of origin and limited metastatic spread and tumor bulk.

 

Colon

 

NET arising in the caecum and ascending colon generally show a biological behavior that is similar to that of small intestinal NET. Together these are termed midgut NET due to their common embryological origin and vascularization by the superior mesenteric artery and vein. Consequently, cecal and ascending colonic NET are often low-grade tumors, can be associated with carcinoid syndrome when metastasized beyond the portal circulation, and give rise to fibrotic complications (67).

 

Contrarily, NEN in the transverse and descending colon are more aggressive with a predilection for the occurrence of NEC. These NEC share common features with adenocarcinomas of the colon, like molecular background (11). Hormonal syndromes are seldomly encountered in these colon NEC.

 

Rectum

 

Unlike the distal colon, NEN in the rectum show a preference for well-differentiated NET (68). Most rectal NET are incidentally discovered during colonoscopy (Figure 1B). A rise in rectal NET incidence rates has been detected that coincided with the increased use of diagnostic colonoscopy (69). At the time of detection, tumor size is often small (< 1 cm) signifying indolent behavior and small risk of metastatic spread (70). However, a subset of rectal NET can present in locally advanced stages and be associated with metastatic spread. Although their venous drainage is not connected to the portal vein, rectal NET are rarely associated with hormonal syndromes, presumably due to their neuroendocrine cell type of origin.

 

DIAGNOSIS

 

Histopathology

 

Obtaining histology for evaluation and confirmation of diagnosis remains essential in the work-up of a gastrointestinal NEN, even in the setting of modern imaging techniques and circulating biomarkers. The diagnosis of a NEN can be suggested through histological findings on H&E staining, such as an organoid pattern, absence of necrosis, low nucleus to cytoplasm ratio, and salt and pepper chromatin. Ultimately, the histological diagnosis requires positive immunohistochemical staining of neuroendocrine markers (71). Most commonly used neuroendocrine markers include synaptophysin and chromogranin A, although N-CAM (CD56) has also been advocated as such in the past. Staining with either synaptophysin or chromogranin A should be positive, with the former having a higher positivity rate in gastrointestinal NEN (72). Expert pathological examination is advised in uncertain cases, for instance in neoplasms with overlap with other malignancies, such as carcinomas with neuroendocrine differentiation, amphicrine carcinoma and MiNEN (25, 73).

 

Besides for confirming the diagnosis, histopathological evaluation is required for tumor grading according to the WHO classification. First, the distinction between a poorly differentiated NEC and a well-differentiated NET is crucial as shown above. This distinction is made on the basis of cellular morphology (74). Second, each pathological evaluation of a NET specimen should include grading through evaluation of differentiation, ki67 (MIB1) proliferation index and mitotic index (Table 2). Importantly, tumor grade can be heterogenous within or between tumor lesions as well as change over time (75, 76). The disease course over many years in patients can be accompanied by an increase in proliferation indices and grade over time, providing rationale for repeat biopsies in selected patients with disease progression. Altogether, grading provides key information for clinical decision making across all stages and primary locations of gastrointestinal NET. The subclass of grade 3 well-differentiated gastrointestinal NET was only introduced as recent as 2019 (2), which limits the clinical studies and experience on the management of this rare subtype.

 

Immunohistochemical analysis can also helpful in cases of an unknown primary tumor. Although the prevalence of an unknown primary tumor has decreased due to contemporary PET imaging, up to 5% of NET can present with an unknown primary (77). Positive staining for the following immunohistochemical marker is specific for different primary origins of NET: TTF-1 for foregut tumor, ISL-1 or PAX8 for pancreatic tumor, CDX-2 for midgut tumor, and SATB2 for hindgut tumor (78-81).

 

Biochemistry – General

 

Historically, elevated levels of biochemical markers have been directly linked to the diagnosis of a NET. While this can be true for certain hormones eliciting clinical syndromes when taken under controlled conditions, the vast majority of NET cannot be diagnosed through the use of a circulating biomarker. At most, a biomarker can be used during follow-up when it is elevated in a particular patient as a marker of disease recurrence or activity (27, 82).

 

Chromogranin A (CgA) has been extensively studied since the 1990s as a diagnostic and prognostic biomarker for gastrointestinal and other NET. This acid glycoprotein is stored within the secretory vesicles of neuroendocrine cells and co-secreted with the hormones upon stimulation. In a meta-analysis of 13 studies including 1260 patients with a NET sensitivity of CgA was 73%. In healthy controls, CgA levels were elevated in less than 5%, securing an excellent specificity. However, when compared to subjects with other gastrointestinal, renal, or oncological disease the specificity can drop to ranges of 50-60% (83), making CgA a poor diagnostic marker in patients presenting with abdominal complaints or a tumor. Measurement of CgA for this indication has led to many unnecessary clinical investigations, e.g., endoscopy, cross-sectional and functional imaging, into the cause of an elevated CgA (84) and should be discouraged.

 

Circulating CgA levels are associated with tumor bulk and consequently are correlated to a worse prognostic outcome (85). Because of its link to tumor bulk, CgA can be used during follow-up to track disease activity, although it should never replace imaging due to insufficient sensitivity and specificity of detecting progressive disease.

 

Neuron-specific enolase (NSE) represents another circulating marker on neuroendocrine cells. Mostly studied in small cell lung cancer, NSE is also elevated in a subset of gastrointestinal NET patients. Its sensitivity and specificity for the diagnosis of NET is approximately 40% and 60%, respectively (85, 86), and thereby inferior to that of CgA. Importantly, NSE levels tend to be more increased in aggressive disease. Consequently, a sudden rise in NSE could herald the occurrence of dedifferentiation in a NET.

 

Other circulating neuroendocrine markers, like pancreatic polypeptide and neurokinin A, have been used as diagnostic biomarkers in the past, but due to their overall lack of sensitivity or specificity their use in clinical practice has disappeared (27).

 

Because of the inferior diagnostic characteristics of the peptides described above an mRNA transcript-based marker called the NETest was developed. Through multiplex PCR and a machine learning-based algorithm, the NETest provides a number on a 100-point scale, where an outcome above 20 has been used for optimal diagnostic cut-off (87). In a meta-analysis of 6 studies the sensitivity and specificity of the NETest was 89-94% and 95-98%, respectively (88). An independent study employing serial sampling in 132 patients with gastroenteropancreatic NET showed a high rate of fluctuation in the NETest despite stable disease during follow-up (89). This technique is of interest to the field, but as of yet there are restrictions regarding the availability in clinical practice, costs, and reimbursement. Hopefully, these developments will lead the way towards more superior multianalyte diagnostic biomarkers for gastrointestinal NET in the future.

 

Biochemistry – Specific

 

When patients present with features compatible with a NET-associated functioning syndrome dedicated analysis should be performed. The reader is referred to other Chapters in Endotext for hormonal analysis of Gastrinoma (16), Insulinoma (90), VIPoma (37), Glucagonoma (91), Somatostatinoma (38), Ghrelinoma (53), and Paraneoplastic Syndromes (39). The latter included the hormonal work-up of NET-associated hypercalcemia, hyponatremia, Cushing’s syndrome, acromegaly and hypoglycemia.

 

Although the majority of gastrointestinal NET are not accompanied by a hormonal syndrome, the carcinoid syndrome is the most common hormonal complication. Because patients can be asymptomatic but still at risk for complications such as carcinoid crisis or CHD, all patients with advanced gastrointestinal NET should undergo biochemical evaluation for the carcinoid syndrome at baseline and when clinical suspicion arises during follow-up (29).

 

Serotonin (5-hydroxytryptamine) is the main but not exclusive culprit in the carcinoid syndrome. Upon secretion it is mainly stored in platelets, but a proportion freely circulates in the blood. It is metabolized by hepatocytes to 5-hydroxyindolaceticacid (5-HIAA), which is more stable than serotonin and excreted in the urine. 24-hour urine 5-HIAA levels are the best-established biomarker for the carcinoid syndrome, with 50 µmol/24h used as the optimal diagnostic cut-off (29, 92). Urinary 5-HIAA levels correlate with tumor bulk and multiple studies have described an association in univariate analyses with survival in CS patients, which did not persist in multivariate analyses (93-95). 5-HIAA level associate with the risk of developing CHD, with levels above 300 µmol/24h conferring a 2.7-fold increased risk of the development of CHD (36). Alternatively, 5-HIAA can be measured in plasma or serum, resulting in a slightly lower sensitivity/specificity compared to 24h urine collection (96, 97). Venous sampling saves on the cumbersome collection of 24h urine, but its availability is currently limited. Similarly, platelet serotonin levels are associated with carcinoid syndrome, but few labs can perform the assay (98). Although several other peptides, including neurokinin A, bradykinin, and histamine, have been associated with the occurrence of carcinoid syndrome, these markers have no utility in the diagnostic workup in clinical practice.

 

NT-proBNP is u useful biomarker to screen for the presence of CHD in patients with established carcinoid syndrome (99). An NT-proBNP level below 260 ng/mL (31 pmol/L) has a negative predictive value of 97%, thereby effectively ruling out the presence of CHD (100). Patients with NT-proBNP levels above 260 mg/mL should be referred for echocardiography to confirm or exclude the presence of CHD.

 

Cross-Sectional Imaging

 

Despite the developments in biochemistry and functional imaging, cross-sectional imaging remains the cornerstone of follow-up of NET. Furthermore, as more NET are incidentally discovered on imaging, it is important to be aware of typical or even pathognomonic radiological features of NET. On contrast-enhanced computer tomography (CT) scan gastrointestinal NET typically present as hypervascular lesions in the bowel wall (101). The majority of NET have enhanced intravenous contrast uptake in arterial phase, making it relevant to include an early arterial scan phase next to a venous or portal phase in case of a suspicion of a NET (102). Primary NET lesions in the small intestine tend to be small and can easily be missed, whereas lymph node or distant metastases can be extensive. Fibrosis can occur in mesenteric NET metastases, leading to pathognomonic fibrotic strands radiating from the mesenteric mass (61)(Figure 2). Gastrointestinal NET predominantly metastasize to the liver, where single, multiple or extensive metastases can be found. Again, these are hypervascular and enhancing on arterial phase in the majority of cases (103) (Figure 3).

 

Figure 3. Cross-sectional imaging in gastrointestinal NET. Due to their hypervascular nature, NET primary lesions and metastases can be enhancing in early arterial phase. In case (A) diffuse hypervascular liver metastases of a small intestinal NET are visible. Not all NET (metastases) are hypervascular, as shown in case (B) with a single non-enhancing liver metastasis of small intestinal NET during arterial phase (arrow). The added value of including an early arterial phase after contrast injection (C) op top of venous phase imaging (D) is illustrated within a patient with a small intestinal NET, where visibility of a segment 3 NET metastasis is improved during arterial scan. MRI, particularly diffusion weighted imaging (DWI), can improve the detection rate of small liver NET metastases (E).

 

Magnetic resonance imaging (MRI) is superior to CT with regard to liver and bone metastases, particularly with contrast enhancement and diffusion-weighted imaging (DWI) (104, 105) (Figure 3). For small liver neuroendocrine metastases, MRI even has a higher lesion-based sensitivity than contemporary SSTR-based PET imaging (see below) (106). In rectal NET, MRI is also helpful to stage local growth and lymph node metastases (107). MRI has caveats in the detection of the primary tumor of the bowel, mesenteric, or peritoneal metastases.

 

Endoscopy

 

Endoscopy is often the modality used leading to the incidental detection of a gastrointestinal NET, particularly within primary locations in the stomach or rectum (Figure 1). Primary tumors of gastroduodenal or rectal origin can also be missed on cross-sectional imaging, providing rationale for performing endoscopy or endoscopic ultrasound (EUS) to stage locoregional disease (67, 108). The added value of endoscopy in advanced disease is generally of limited value, unless the aim is to obtain histology. Alternatively, obtaining histology from metastases could be more informative as these can have a higher grade than the primary tumor and ultimately determine the patient prognosis (76).

 

Nuclear Imaging

 

Over 90% of well-differentiated NET express somatostatin receptors, which can be used for functional imaging. Somatostatin is a hormone, whose physiological actions are to inhibit hormonal production and release from neuroendocrine cells, for instance in the pituitary, pancreas, and intestine (109). It binds to one or more of five somatostatin receptor subtypes expressed on the cell membrane. Radiolabeled somatostatin analogues (SSA) were developed in the 1980s to image gastrointestinal and pancreatic NET. First, Octreoscan® with gamma-emitter 111In-pentreotide was shown superior to cross-sectional imaging in NET using planar and SPECT imaging (110). In the recent ten years, 68Gallium-labeled SSA (68Ga-DOTATATE, 68Ga-DOTATOC, 68Ga-DOTANOC) suitable for PET imaging have replaced 111In-pentreotide as the preferred imaging modality. Importantly, 68Ga-DOTA-SSA PET changes clinical management in 40-50% of cases, according to two meta-analyses (111, 112), and as such constitutes a key diagnostic modality in the NET armamentarium (Figure 4). The PET can be combined with diagnostic, contrast-enhanced CT (PET/CT) or MRI (PET/MRI) for hybrid imaging. Pitfalls include PET-positive granulomatous disease, meningioma, renal cell cancer, and lymphoma. Expression of the somatostatin receptors decreases with increasing proliferative capacity in NET, making it very useful in low-to-intermediate grade NET but less sensitive in higher grade NET or NEC. Recently, 64Cu-DOTA-SSA PET/CT and 18F-AIF-NOTA-SSA have been introduced with similar or slighter superior diagnostic capability compared to 68Ga-DOTA-SSA PET (113, 114).

 

Alternatively, 18F-DOPA PET has been advocated by several centers as superior to 68Ga-DOTA-SSA PET, particularly for midgut NET (115). Although this may vary between patients and mostly pertain to tumor count rather than to change in management, 68Ga-DOTA SSA also has therapeutic consequences for theranostics using unlabeled (‘cold’) SSA and peptide receptor radionuclide therapy (PRRT) with radiolabeled (‘hot’) SSA (see below).

 

Figure 4. 68Ga-DOTA-SSA PET imaging. 68Ga-DOTA-SSA PET staging is superior to anatomical imaging and 111In-pentreotide SPECT (Octreoscan). In this case of a patient with stage IV small intestinal NET, PET imaging detected more lesions than Octreoscan, scanned within 3-month timeframe without anatomical progression. In the same patient, multiple liver metastases are detected on hybrid PET/CT imaging (arrow), which were not visible on contrast-enhanced CT (CECT).

 

Similar to other malignancies, a subset of NET metabolize increased amounts of glucose, which makes them amenable to imaging with 18F-fluorodeoxyglucose (FDG) PET. Uptake of 18F-FDG PET in NET increases with aggressiveness, making it the preferred imaging modality in NEC and higher-grade NET (116, 117). Positive FDG uptake of NET is associated with growth potential and consequently several studies have established that FGD uptake constitutes a prognostic marker for a worse survival outcome (118).

 

MANAGEMENT

 

Surgery

 

Radical resection remains the cornerstone in the management of locoregional stages of gastrointestinal NET. Metastatic spread is dependent on the location and size of the primary tumor and adequate staging should be performed accordingly, preferably through hybrid cross-sectional and 68Ga-DOTA-SSA PET imaging (102). If the disease is confined to the local tumor (stage I-II) or locoregional lymph nodes (stage III), the option of a surgical oncological resection should be evaluated. If the NET can be radically resected the outcome is very favorable with 10-years survival outcomes of >90% for all primary sites. A large registry series from Canada did find that recurrence rates can increase up to 60% for small intestinal NET and 40-50% for other NET in a 15-year postoperative period (119). Given the retrospective nature of this series and contemporary preoperative imaging it remains uncertain whether recurrence rates of current therapeutic interventions are still this high.

 

For stage I gastroduodenal NET, metastatic spread is limited and endoscopic resection of the NET can be considered (108). This pertains to gastric type I and type II NET up to 2 cm without muscle invasion and duodenal NET localized at safe distance from the Vater’s ampulla. Similarly, an endoscopic resection can be performed in stage I rectal NET, as the risk of lymph node metastases is limited to less than 3% (67). Resection from both tumor subtypes should be performed by endoscopic mucosal resection (EMR), endoscopic submucosal dissection (ESD), or endoscopic full thickness resection (eFTR) rather than snare polypectomy due to the submucosal growth pattern of NET. Successful removal of type I gastric NET or stage I rectal NET is are high (>85%) with slight superiority of ESD over EMR, while eFTR might approach 100% radical resection rates (120-122). In cases of an inadequate endoscopic resection further imaging should be performed and a step-up endoscopic approach or surgical resection should be considered.

 

Patients with oligometastatic disease might also benefit from an upfront surgical approach. As the liver is the predominant site for metastatic disease, concomitant surgical resection and/or interventional tumor ablation should be considered in patients with limited liver involvement (123). This can potentially cure the patient, but it should be noted that modern imaging techniques detect approximately one-third of liver metastases compared to histological evaluation (124, 125). The presence of micrometastases should be factored into the management process. Despite this, long-term outcomes can be excellent in cases of upfront surgery in oligometastatic disease. A potential advantage of tumor debulking in this setting could be the delay of the need to start systemic therapy. Several series have also described survival benefits of extensive liver metastases resection (126-129), but these concern mostly retrospective series, which might introduce selection bias, and data was often collected before the advent of currently available molecular therapies.

 

Resection of the primary tumor in the context of stage IV or metastatic disease is controversial. Whereas retrospective studies have supported a survival benefit in patients whose primary tumor was resected compared to those that were not operated (130-132), this was later refuted in other series or after propensity score-matched controls (63, 133). Importantly, the disease course locoregionally can be indolent, and in one series only 13% of mesenteric masses showing significant progression after a median follow-up time of 40 months (62). Patients with advanced midgut NET and recurrent complaints from the primary tumor or (fibrotic) mesenteric mass should undergo operation to explore the possibility of a palliative resection or alternatively, an intestinal bypass.

 

Palliative Management

 

Patients with unresectable or advanced gastrointestinal NET are in a palliative setting and the different treatment modalities should be weighed in terms of efficacy and toxicity. Given the wide range of gastrointestinal NET subtypes, the treatment chosen should align with the biological behavior of the tumor as well as the characteristics of the individual patient (Figure 5). Factors to consider in the management of gastrointestinal NET include: tumor grade, growth rate and location(s), symptoms, presence of a hormonal syndrome, performance score, comorbidities, previous therapies, availability of treatments and patient preference.

 

Figure 5. Stage IV gastrointestinal NET. There is a wide heterogeneity in clinical presentation of gastrointestinal NET in advanced or metastatic setting. On these maximal intensity projections of 68Ga-DOTATATE PET, there are 8 different clinical scenarios of stage IV gastrointestinal NET. Despite the similar disease stage, all these patients deserve personalized management of their disease according to several patient- and tumor-specific factors. For optimal management, choice of treatment should be discussed in an experienced multidisciplinary setting.

 

Active Surveillance

 

One potential option to consider is to perform active surveillance in asymptomatic patients with advanced, grade I or low-grade II NET with limited tumor bulk. Evidence for this strategy can be found in placebo-controlled trials. First, the median time to progression in placebo-treated patients was 6 months in the phase III randomized PROMID trial in midgut NET patients (134). Second, in the phase III randomized CLARINET trial in patients with nonfunctioning GEP NET, patients randomized to placebo had a median progression-free survival (PFS) of 18 months (135). Consequently, not all tumors show clear growth potential over time and selected patients can thus safely refrain from costly and potentially toxic medication. This strategy should not be adopted in patients with symptomatic, functioning, high-grade, quickly progressive, or high tumor volume disease. Follow-up cross-sectional imaging every 3-6 months is advised for gastrointestinal NET patients undergoing active surveillance.

 

Somatostatin Analogs

 

Before their role in imaging, SSA were developed for their potential antihormonal effects. The SSA octreotide was found to effectively reduce serotonin production in patients with carcinoid syndrome and other NEN-associated functioning syndromes (136). Following its long-term application in functioning NET, antitumoral efficacy was tested in the PROMID and CLARINET trials. The German multicenter PROMID study randomized 85 midgut NET patients to 4-weekly 30 mg octreotide long-acting release (LAR) injections or placebo injections (134). These patients were in the beginning of their disease course with a median time from diagnosis of 4 months and had on average limited liver tumor load and grade I. In an intention to treated (ITT) analysis median time to progression was 14.3 months in octreotide LAR-treated patients versus 6.0 months in the placebo group (P=0.000072). Overall survival (OS) was not different between the groups. The effect of SSA is predominantly stabilization of disease as only one patient in both treatment groups experienced a partial response. Overall, octreotide LAR treatment was well tolerated, although diarrhea, flatulence, and bile stones were more frequently observed in the SSA-treated group.

 

The international multicenter CLARINET trial randomized 204 patients with advanced nonfunctioning GEP NET to 4-weekly injections of 120 mg lanreotide autogel or placebo injections (135). Tumors were grade I and II with ki-67 index up to 10% and mostly from pancreas or midgut origin. Over 80% of patients had not received previous antitumoral treatment and tumor progression before randomization was only shown in 4-5% of patients. ITT analysis revealed that PFS was significantly longer in lanreotide-treated patients compared to placebo (median not reached versus 18.0 months, P<0.001). The benefit of lanreotide persisted in most predefined subgroups across primary origin, tumor grade, and liver involvement. Safety of lanreotide was good, with known side effects of gastrointestinal complaints, exocrine pancreas insufficiency, and hyperglycemia. Interestingly, the open label extension study of the CLARINET showed a median PFS of 33 months in those continuing lanreotide, while patients in the placebo group – with a median PFS of 14 months - who crossed over to lanreotide after progression had a median second PFS of 18 months (137). This again supports the possibility of considering active surveillance in a subset of patients with indolent disease. Overall survival (OS) in the core CLARINET study was not significantly different between treatment groups, but was also biased by crossover from placebo to lanreotide.

 

Together these landmark trials have positioned SSA as first-line antiproliferative treatment for well-differentiated gastrointestinal NET, particularly in patients without signs of high tumor volume or aggressive disease course. Injections with octreotide LAR or lanreotide are every 4 weeks in the gluteal area intramuscularly or deep subcutaneously, respectively. Overall tolerability is excellent, although patients should be counselled on the potential gastrointestinal adverse effects, e.g., diarrhea, flatulence, nausea, stool discoloration, after the first administration, which tend to dissipate after repeated injections. Long-term concerns include hyperglycemia and bile stones. Although preventive cholecystectomy has been advocated in the past, this practice has been abandoned in most expert centers (138).

 

Several retrospective series and clinical experience supported the use of SSA dose escalation in patients with mild progressive disease (139). These studies suggest that increasing the injected dose or injection frequency might be accompanied by improved antiproliferative control. First prospective evidence of this effect came from the NETTER-1 study designed to investigate the effect of peptide receptor radionuclide therapy (PRRT) with 177Lutetium-DOTA-octreotate (177Lu-DOTATATE) (140). Patients enrolled in this study had advanced, progressive midgut NET on regular dose of SSA and were randomized between PRRT and an escalated dose of 60 mg of octreotide LAR every four weeks. Patients in the high-dose SSA control group had a medium PFS of 8.4 months, supporting some antiproliferative effect of SSA dose escalation after disease progression on a regular dose of SSA. The CLARINET forte single-arm, phase II trial was designed to study the efficacy of lanreotide dose escalation in midgut and pancreatic NET patients with disease progression on standard lanreotide dose in the previous 2 years (141). In the midgut NET cohort, 51 patients were included with grade 1-2 disease and 57% of patients had – generally limited - hepatic metastases. After dose escalation to lanreotide 120 mg every 2 weeks median PFS in this cohort was 8.3 months, while disease control rate (partial response or stable disease as best outcome) was 73%. Importantly, no deterioration of quality of life and no additional treatment-related safety concerns were observed in patients treated with high-dose lanreotide.

 

SSA treatment should be given lifelong in patients with carcinoid syndrome and other SSA-responsive functioning syndromes for which these drugs are registered and approved (29, 142). This includes continuation of treatment after radiological or clinical progression and initiation of a second-line of treatment. Whether SSA should be continued in nonfunctioning gastrointestinal NET disease is a matter of controversy and no prospective data is available to guide this. Intriguingly, 50% of panelists in the NANETS guideline supported continuing SSA treatment, while 50% supported stopping treatment upon progression (143).

 

The pan-somatostatin receptor agonist pasireotide has been investigated in NET based on the hypothesis that targeting more somatostatin receptor subtypes might have an additive antiproliferative effect compared to octreotide and lanreotide, which predominantly target the somatostatin receptor subtype 2 (144). However, early phase clinical trials provided insufficient grounds to pursue further clinical development of this drug in NET (145, 146).

 

Peptide Receptor Radionuclide Therapy

 

Similar to the diagnostics and therapeutics of thyroid disease with radioactive iodine, the discovery of molecular somatostatin receptor imaging also heralded the advent of targeted somatostatin receptor-based radionuclide therapy. Following initial developments with 111In-pentreotide and 90Yttrium-DOTATATE, the short-range beta-emitter 177Lutetium coupled to DOTATATE (177Lu-DOTATATE) was introduced in 2000 (147). This technique of targeting the somatostatin receptor on tumor cells with internal radiation was termed PRRT.

 

Individual phase II trials at several centers showed promising antitumoral effects on somatostatin receptor-positive NET, including gastrointestinal subtypes (148). The multinational phase III randomized NETTER-1 trial established PRRT with 4 cycles of 177Lu-DOTATATE as an effective therapy for advanced, somatostatin receptor-positive midgut NET (140). In this trial, 229 patients were randomized between PRRT, including 30 mg octreotide LAR between cycles and 4-weekly after the fourth cycle, and 60 mg octreotide LAR every four weeks. Patients had a grade 1-2 midgut NET that was progressive on SSA before enrollment. The median PFS in the PRRT group was not reached compared to 8.4 months in the high-dose SSA group. Benefit in PFS prolongation was evident across all pre-specified subgroups. Risk of progression or death was 79% and decreased in the patients treated with PRRT. The study confirmed known side effects of 177Lu-DOTATATE, including nausea, fatigue, abdominal pain, and diarrhea. Two percent of patients experienced grade 3 or higher thrombocytopenia, while 2 patients (1.8%) developed myelodysplastic syndrome following PRRT. In a meta-analysis of 28 studies comprising 7334 patients treated with 90Y-DOTATOC or 177Lu-DOTATATE, the combined incidence of myelodysplastic syndrome and acute myeloid leukemia after PRRT was 2.6% (149). Final analysis of the NETTER-1 study revealed that the median OS in the PRRT group was 48.0 months compared to 36.3 months in the high-dose SSA group, which was not significantly different (150). Crossover of 37% of the patients randomized to high-dose SSA, long-term survival with multiple other treatment lines and insufficient statistical power could have contributed to the failure of reaching this secondary endpoint. Another key secondary endpoint was reached: time to deterioration of quality of life was significantly longer in patients treated with PRRT compared to those treated with high-dose SSA (151).

 

Although the NETTER-1 only included midgut NET patients, the phase II Erasmus MC Rotterdam data were used to obtain regulatory approval of 177Lu-DOTATATE for all gastrointestinal (and pancreatic) NET subtypes (152). Importantly, PRRT also induced tumor response in 18% of midgut NET patients in the NETTER-1 study and 39% of various NET patients in the Rotterdam study, which makes it a potential cytoreductive therapy. Standard protocol of PRRT included four infusions of 7.4 GBq 177Lu-DOTATATE spaced 8 (range 6-12) weeks apart. PRRT should preferably be administered in the absence of long-acting SSA (4-6 weeks) or short-acting SSA (24 hours) due to competition at the receptor level. An amino acid solution of 2.5% lysine and arginine is co-infused with 177Lu-DOTATATE in order to saturate the renal reuptake of radioactive peptide and prevent radiation-induced nephrotoxicity. This limits the incidence of severe renal insufficiency after PRRT to less than 1% (152). Special considerations should be applied to patients with pre-existing cytopenia or clonal hematopoiesis, impaired renal function or hydronephrosis, massive liver tumor bulk, mesenteric fibrosis, or nervous system involvement (153). Patients with a severe functioning syndrome are at risk of an exacerbation of symptoms or hormonal crisis following temporary SSA withdrawal or tumor lysis with PRRT. Although the risk is minor at 1% incidence in retrospective series and limited to patients with severe hormonal hypersecretion (154, 155), adequate management through supportive measures and swift re-introduction of SSA should be employed to prevent a hormonal crisis.

 

There is a possibility for salvage PRRT when progressive disease (re-)occurs after a period of disease control following 4 cycles of PRRT. Several retrospective series have described renewed disease control or even response after additional cycles with 177Lu-DOTATATE after progression. In the largest series to date of 181 patients with gastrointestinal, pancreatic, bronchopulmonary, or unknown origin NET, salvage PRRT with two cycles was administered if disease progression occurred after a period of at least 18 months after the first cycle of the initial PRRT (156). The median PFS after salvage PRRT was 14.6 months and thereby approximately 50% of the initial PRRT, while disease control was observed in 75% of patients. Salvage PRRT was not associated with increased rates of myelotoxicity or nephrotoxicity. In patients that respond favorably to salvage PRRT, future cycles can be considered when progressive disease once again arises, although clinical outcome data of additional treatments are scarce.

 

Targeted Therapy

 

The mammalian target of rapamycin (mTOR) protein is a central proliferative factor in many cancer cells. Inhibition of the mTOR pathway has been investigated for several malignancies, including NEN. The RADIANT-2 multicenter phase III trial investigated whether the oral mTOR inhibitor everolimus had efficacy in patients with advanced NET and carcinoid syndrome (157). In total, 429 patients with progressive and advanced grade 1-2 disease were randomized between everolimus 10 mg q.d. plus octreotide LAR 30 mg every 4 weeks or placebo plus octreotide 30 mg every 4 weeks. Primary sites included among others small intestine (52%), lung (10%), colon (6%), and pancreas (6%). Baseline characteristics between the groups were not well balanced with regard to WHO performance status, primary sites, and prior use of chemotherapy. The median PFS was 16.4 months in the everolimus combination group compared to 11.3 months in the placebo combination group (p=0.026). This analysis encompassing central review of radiological images did not reach the pre-specified cut-off for superiority. Median OS was 35.2 months in the placebo-octreotide LAR group compared to 29.2 months in the everolimus-octreotide LAR group, which was not a statistically significant difference, but more deaths related to respiratory or cardiac disease were observed in the everolimus arm.

 

In the RADIANT-4 phase III trial, patients with advanced, progressive, grade 1-2, nonfunctioning NET of gastrointestinal or lung origin were included (158). Here, 302 patients were randomized 2:1 to everolimus 10 mg q.d. or placebo. Approximately 60% of patients had a gastrointestinal NET, while 80% had liver metastases, generally with limited liver tumor bulk. Median PFS was longer in the everolimus-treated patients at 11.0 months versus 3.9 months in the placebo group. This difference was significant after central radiology review as well as after local review (P<0.00001). Despite a 36% reduction in the risk at death in the everolimus group, overall survival was not significantly improved. Partial response was obtained in 2% of patient treated with everolimus, while stable disease was observed in 81%. Given the outcomes of the RADIANT-2 and RADIANT-4 trials, everolimus appears to be better suited for nonfunctioning NET than functioning NET.

 

Everolimus use is associated with a high rate of side effects, such as stomatitis, rash, diarrhea, fatigue, diabetes, infections, and non-infectious pneumonitis. Dose reductions or interruptions are necessary in up to two-thirds of NET patients taking everolimus (158). No benefit in terms of quality of life has been proven for everolimus (159), with potentially a decrease in quality of life in patients with extrapancreatic NET (160).

 

Multitarget tyrosine kinase inhibitors (MTKI) are another form of targeted therapy that can exert potent anti-cancer effects. Sunitinib is an oral multireceptor MTKI which has been investigated in panNET patients. In a phase II study, suninitib showed encouraging antitumoral activity in 61 pancreatic NET with partial response observed in 17% (161). While the median time to progression of 10.2 months in 41 patients with gastrointestinal and lung NET treated with sunitinib exceeded the 7.7 months observed in panNET patients, further development of sunitinib in gastrointestinal NET was not pursued due to the low response rate of 2.4%. A subsequent phase III trial in panNET patients showed that sunitinib improved PFS and OS in panNET patients (162), which led to registration of this drug for NET of pancreatic origin only.

 

Another MTKI surufatinib was tested in two phase III studies in China in pancreatic and extrapancreatic NET, respectively (163, 164). In the multicenter, randomized SANET-ep trial 198 patients with advanced, grade 1-2, progressive NET of gastrointestinal (47%), thoracic (24%), or other origins were randomized 2:1 to oral surufatinib 300 mg or placebo once daily (164). The median PFS after central review in the surufatinib group was 7.4 months compared to 3.9 months in the placebo group (P=0.037), which appeared to be independent of the subgroups studied. There was a large difference with the local radiology review, which tended to overexaggerate the effect of surufatinib on PFS. OS was not different between the groups at the time of the interim analysis. Partial response and stable disease were observed in 10 (8%) and 88 (70%) out of 126 patients, respectively, in the surufatinib arm. Relevant treatment-related side effects included hypertension, proteinuria, anemia and elevated liver enzymes. Quality of life did not improve in the surufatinib arm, while surufatinib-treated patients experienced more diarrhea than those in the placebo arm (165). Based on the SANET-ep study and its partner SANET-p study in panNET patients, surufatinib is registered in China for the treatment of nonpancreatic and pancreatic NET. Surufatinib is thus far not registered for these indications by the FDA or EMA.

 

Several other MTKI have shown potential for antiproliferative activity in NET patients. These include pazopanib (166), lenvatinib (167), and axitinib (168). Further phase III data are necessary before these MTKI can be considered in gastrointestinal NET.

 

Immunotherapy: Interferon-Alpha and Immune Checkpoint Inhibitors

 

In the 1980s, the advent of interferon as a novel cancer drug was also investigated in NEN. Several uncontrolled series supported antiproliferative and antihormonal effects of interferon alpha in mostly small intestinal NET (169, 170). The proinflammatory effects of interferon alpha however led to side effects of flu-like symptoms, myalgia, asthenia, auto-immune diseases, and diarrhea, limiting its tolerability in patients. Compared to SSA, interferon alpha had comparable antiproliferative effects (171). Long-acting interferon alpha appears to be better tolerated and was shown to produce antitumor effect in a single retrospective series in 17 patients (172). 

 

Immunotherapy with immune checkpoint inhibitors has revolutionized treatment of several malignancies, including melanoma and non-small cell lung cancer. However, infiltration of immune cells, like T-cells, is a rare occurrence in NET samples (173-175). In line with these preclinical findings, immune checkpoint inhibition in clinical (basket) trials have failed to show positive effects in well-differentiated NET (176-178).

 

Chemotherapy

 

In contrast to panNET there are no phase III clinical data to support the use of chemotherapy in gastrointestinal NET. Presumably in part through their well-differentiated nature, response rates to chemotherapy have been disappointing and further clinical development halted (179). Consequently, ENETS 2016 and NANETS 2017 guidelines do not support the use of chemotherapy in gastrointestinal NET (143, 180). The EMSO 2021 guideline does advocate the use of either FOLFOX (5-fluorourical, oxaliplatin) or TEMCAP (temozolomide, capecitabine) in selected cases with high grade 2 gastrointestinal NET in third-line or higher setting, although this is not supported by prospective clinical data (181).

 

Supportive Therapy

 

Due to the primary tumor and metastasis locations as well as the sequalae of hormonal overproduction and therapeutic interventions, patients with gastrointestinal NET can be in a poor clinical condition. Inadequate nutrient intake and uptake in these patients leads to increased incidence rates of weight loss, muscle atrophy, and decreased performance status (182). Consequently, all gastrointestinal NET patients should be screened on dietary intake and referred to dieticians if they are at risk of weight loss. High-protein, high-calorie supplements should be prescribed if regular dietary advice is insufficient to prevent weight loss. In cases of suspected reduced calorie uptake due to exocrine pancreatic insufficiency, often encountered during SSA treatment, or bile acid diarrhea, due to bowel resection, a trial of pancreatic enzyme supplements or bile acid sequestrants can be considered.

 

In some cases, patients can be refractory to these interventions and escalation should be considered. This is particularly true for patients with extensive bowel resections leading to short bowel syndrome and those with severe desmoplastic reaction surrounding mesenteric metastases of small bowel NET. Food intake in the latter group might also be compromised by intermittent venous ischemic pain precipitated by meals. Tube feeding through nasogastric tube should be considered in selected cases. In case enteral feeding fails to improve the clinical situation, total parenteral nutrition can serve as a last resort for these refractory cases. Treatment with total parenteral nutrition up to 5 years has been successfully implemented in severe cases of NET (183).

 

Besides nutritional support, physical therapy should also be offered to patients in order to improve their clinical performance status. Finally, given the impact of an incurable disease and its complaints psychosocial support should be discussed with patients and made accessible, if needed (184).

 

MANAGEMENT OF CARCINOID SYNDROME

 

Patient with gastrointestinal NET and the carcinoid syndrome require dedicated management of their hormonal symptoms. Quality of life in these patients is severely decreased, even when compared to patients with other – generally more aggressive – cancers (185). Prompt recognition of symptoms of flushing and diarrhea is key to specific management, while the complications of mesenteric fibrosis and CHD should also be screened and treated adequately (29).

 

The cornerstone of the management of the carcinoid syndrome is SSA. Since the 1980s octreotide and later lanreotide have been shown to lead to biochemical and clinical responses in patients with the carcinoid syndrome. In a meta-analysis comprising 1945 interventions in 33 studies, SSA significantly decreased 5-HIAA excretion in 45-46% of patients, while flushing and diarrhea were decreased in 69-72% and 65%, respectively (186). Also given its favorable tolerability, all patients should be started on SSA soon after a confirmed diagnosis of carcinoid syndrome.

 

Although patients with carcinoid syndrome in the majority of cases have widespread disease, the option of cytoreductive therapy by surgical resection or ablation or intra-arterial liver embolization can be considered in selected cases. If the vast majority of tumor bulk can be resected or embolized, this can lead to biochemical responses and clinical benefit for the patient (186). These options should be weighed also considering the level of serotonin overproduction, tumor growth rate, and efficacy of SSA. Importantly, SSA should be initiated before interventional therapy is commenced in order to reduce the risk of a carcinoid crisis (187).  

 

Patients with persistent symptoms despite label doses of SSA are designated as having refractory carcinoid syndrome. Several systemic options are available for treatment and these should be weighed on an individual basis guided by tumor bulk, rate of progression, severity of symptoms, and availability. Dose escalation of SSA can be attempted and leads to symptomatic improvement in 72-84% of patients (186). Alternatively, a randomized controlled trial has proven efficacy of the oral drug telotristat ethyl in controlling diarrhea in patients with refractory carcinoid syndrome (188). This serotonin synthesis inhibitor, dosed at 250 mg t.i.d., decreased bowel movements in approximately half of the cases and with a mean reduction of 0.8 bowel movements per day, whilst having no significant effect on flushing. A drug trial of three months is generally advised with stopping of telotristat ethyl if no benefit has been obtained after this time. Clinical symptoms improved in patients treated with PRRT in the NETTER-1 trial (140), although no sub-analysis was performed for carcinoid syndrome patients. In a retrospective series of 24 patients with stable disease or severe, refractory carcinoid syndrome, PRRT with four cycles of 177Lu-DOTATATE effectively reduced flushes and diarrhea in 67% and 47% of patients, respectively (155). Therefore, PRRT constitutes a viable option for refractory carcinoid syndrome patients with aggressive or progressive disease. In the past, interferon-alpha injections have been shown to diminish diarrhea and flushing resulting from carcinoid syndrome. Its antihormonal effect on top of SSA was limited (189), however, and given its poor tolerability interferon-alpha is reserved to selected cases, refractory to the above-mentioned options. Anecdotal reports support the use of serotonin receptor antagonists, like granisetron or ondansetron, and antihistamines (H1 and H2 receptor blockers) in refractory carcinoid syndrome.

 

Importantly, the patient should be counselled on supportive therapy, which could include the use of antidiarrheals, like loperamide or morphine, adaptation of dietary intake, including avoidance of alcohol, tryptophan-containing or spicy foods, and the avoidance of stressors (29). Patients with severe carcinoid syndrome are at a high risk of a catabolic state and vitamin deficiencies. Patients should be referred to a dietician and adequately monitored and supplemented for vitamin deficiencies, particularly for vitamin B3 or niacin and fat-soluble vitamins.

 

Patients suffering from CHD should be evaluated by cardiologists experienced in right-sided cardiac pathology. Dedicated echocardiographic evaluations should be performed, preferably through standardized protocols (190). Fluid and salt restriction comprise first-line treatment of right-sided heart failure due to tricuspid valve regurgitation or pulmonary valve regurgitation or stenosis in the context of CHD. Alternatively, loop diuretics can be prescribed to treat fluid overload and edema. Severe symptomatic patients should be discussed in a multidisciplinary team for evaluation of surgical valve replacement (191).

 

PROGNOSIS AND FOLLOW-UP

 

Resection is the only potential cure for gastrointestinal NET. Recurrence is however frequently observed in NET patients operated on with curative intent (119). Exceptions that are associated with excellent curation rates after local resection include T1-T2 appendiceal, gastric, duodenal, or rectal NET. Long-term imaging follow-up is mandated for the other subtypes of gastrointestinal NET after resection of localized, locoregional, or oligometastatic disease.

 

In a US registry study of almost 100,000 NET patients, median overall survival was 112 months and 62% of patients died of disease-related causes (192). All-cause mortality was 4.3-fold higher in all NET patients, compared to the general population, while patients with stage IV disease had 35-fold elevated risk of mortality. Whereas patients with localized disease still have an elevated standardized mortality ratio, the risk of non-cancer death is higher than cancer-related death in patients with non-metastatic gastrointestinal NET (193). Primary site, stage or grade are tumor-specific prognostic markers, while age, sex, comorbidities and socio-economic status constitute patient-specific factors that are associated with overall survival (7, 8, 192-194). Over the last few decades, NET management has improved considerably with the advent of superior classification, imaging, and biochemical diagnostics and treatment modalities. These developments, combined with expert multidisciplinary team care in dedicated NET centers, have likely contributed to the observed improvement in overall survival in patients with gastrointestinal NET (7, 8). However, survival of gastrointestinal NET patients is still limited, particularly in those with advanced disease, prompting the need for future innovation in the fields of early detection of disease (recurrence), novel druggable targets, and personalized management for NET.

 

REFERENCES

 

  1. Chauhan, A., E. Kohn, and J. Del Rivero, Neuroendocrine Tumors-Less Well Known, Often Misunderstood, and Rapidly Growing in Incidence. JAMA Oncol, 2020. 6(1): p. 21-22.
  2. Nagtegaal, I.D., et al., The 2019 WHO classification of tumours of the digestive system. Histopathology, 2020. 76(2): p. 182-188.
  3. de Herder, W.W., et al., A short history of neuroendocrine tumours and their peptide hormones. Best Pract Res Clin Endocrinol Metab, 2016. 30(1): p. 3-17.
  4. Andersson-Rolf, A., H. Clevers, and T.L. Dayton, Diffuse Hormonal Systems, in Endotext, K.R. Feingold, et al., Editors. 2000: South Dartmouth (MA).
  5. Marien, L., et al., Pathophysiology and Treatment of Pancreatic Neuroendocrine Neoplasms (PNENS): New Developments, in Endotext, K.R. Feingold, et al., Editors. 2000: South Dartmouth (MA).
  6. Fraenkel, M., et al., Incidence of gastroenteropancreatic neuroendocrine tumours: a systematic review of the literature. Endocr Relat Cancer, 2014. 21(3): p. R153-63.
  7. Dasari, A., et al., Trends in the Incidence, Prevalence, and Survival Outcomes in Patients With Neuroendocrine Tumors in the United States. JAMA Oncol, 2017. 3(10): p. 1335-1342.
  8. White, B.E., et al., Incidence and survival of neuroendocrine neoplasia in England 1995-2018: A retrospective, population-based study. Lancet Reg Health Eur, 2022. 23: p. 100510.
  9. Mafficini, A. and A. Scarpa, Genetics and Epigenetics of Gastroenteropancreatic Neuroendocrine Neoplasms. Endocr Rev, 2019. 40(2): p. 506-536.
  10. Priestley, P., et al., Pan-cancer whole-genome analyses of metastatic solid tumours. Nature, 2019. 575(7781): p. 210-216.
  11. van Riet, J., et al., The genomic landscape of 85 advanced neuroendocrine neoplasms reveals subtype-heterogeneity and potential therapeutic targets. Nat Commun, 2021. 12(1): p. 4612.
  12. Francis, J.M., et al., Somatic mutation of CDKN1B in small intestine neuroendocrine tumors. Nat Genet, 2013. 45(12): p. 1483-6.
  13. de Herder, W.W. and J. Hofland, Multiple Endocrine Neoplasia Type 4, in Endotext, K.R. Feingold, et al., Editors. 2000: South Dartmouth (MA).
  14. Makinen, N., et al., Whole genome sequencing reveals the independent clonal origin of multifocal ileal neuroendocrine tumors. Genome Med, 2022. 14(1): p. 82.
  15. Pieterman, C.R.C., et al., Multiple Endocrine Neoplasia Type 1, in Endotext, K.R. Feingold, et al., Editors. 2000: South Dartmouth (MA).
  16. Jensen, R.T. and T. Ito, Gastrinoma, in Endotext, K.R. Feingold, et al., Editors. 2000: South Dartmouth (MA).
  17. Giri, A.K., et al., Genome wide association study identifies 4 novel risk loci for small intestinal neuroendocrine tumors including a missense mutation in LGR5. Gastroenterology, 2023.
  18. Hashemi, J., et al., Copy number alterations in small intestinal neuroendocrine tumors determined by array comparative genomic hybridization. BMC Cancer, 2013. 13: p. 505.
  19. Dawson, M.A. and T. Kouzarides, Cancer epigenetics: from mechanism to therapy. Cell, 2012. 150(1): p. 12-27.
  20. Karpathakis, A., et al., Prognostic Impact of Novel Molecular Subtypes of Small Intestinal Neuroendocrine Tumor. Clin Cancer Res, 2016. 22(1): p. 250-8.
  21. Pearse, A.G., The cytochemistry and ultrastructure of polypeptide hormone-producing cells of the APUD series and the embryologic, physiologic and pathologic implications of the concept. J Histochem Cytochem, 1969. 17(5): p. 303-13.
  22. Hofland, J., G. Kaltsas, and W.W. de Herder, Advances in the Diagnosis and Management of Well-Differentiated Neuroendocrine Neoplasms. Endocr Rev, 2020. 41(2): p. 371-403.
  23. Rindi, G., et al., A common classification framework for neuroendocrine neoplasms: an International Agency for Research on Cancer (IARC) and World Health Organization (WHO) expert consensus proposal. Mod Pathol, 2018. 31(12): p. 1770-1786.
  24. Rindi, G., et al., Competitive Testing of the WHO 2010 versus the WHO 2017 Grading of Pancreatic Neuroendocrine Neoplasms: Data from a Large International Cohort Study. Neuroendocrinology, 2018. 107(4): p. 375-386.
  25. Rindi, G., et al., Overview of the 2022 WHO Classification of Neuroendocrine Neoplasms. Endocr Pathol, 2022. 33(1): p. 115-154.
  26. Kasajima, A., et al., An analysis of 130 neuroendocrine tumors G3 regarding prevalence, origin, metastasis, and diagnostic features. Virchows Arch, 2022. 480(2): p. 359-368.
  27. Hofland, J., W.T. Zandee, and W.W. de Herder, Role of biomarker tests for diagnosis of neuroendocrine tumours. Nat Rev Endocrinol, 2018. 14(11): p. 656-669.
  28. Halperin, D.M., et al., Frequency of carcinoid syndrome at neuroendocrine tumour diagnosis: a population-based study. Lancet Oncol, 2017. 18(4): p. 525-534.
  29. Grozinsky-Glasberg, S., et al., European Neuroendocrine Tumor Society (ENETS) 2022 Guidance Paper for Carcinoid Syndrome and Carcinoid Heart Disease. J Neuroendocrinol, 2022. 34(7): p. e13146.
  30. Grozinsky-Glasberg, S., A.B. Grossman, and D.J. Gross, Carcinoid Heart Disease: From Pathophysiology to Treatment--'Something in the Way It Moves'. Neuroendocrinology, 2015. 101(4): p. 263-73.
  31. Condron, M.E., et al., A prospective study of the pathophysiology of carcinoid crisis. Surgery, 2019. 165(1): p. 158-165.
  32. Oates, J.A., et al., Release of a Kinin Peptide in the Carcinoid Syndrome. Lancet, 1964. 1(7332): p. 514-7.
  33. Clement, D., J. Ramage, and R. Srirajaskanthan, Update on Pathophysiology, Treatment, and Complications of Carcinoid Syndrome. J Oncol, 2020. 2020: p. 8341426.
  34. Ito, T., L. Lee, and R.T. Jensen, Carcinoid-syndrome: recent advances, current status and controversies. Curr Opin Endocrinol Diabetes Obes, 2018. 25(1): p. 22-35.
  35. Zandee, W.T., W.W. de Herder, and H. Jann, Incidence and prognosis of carcinoid syndrome: hormones or tumour burden? Lancet Oncol, 2017. 18(6): p. e299.
  36. Bhattacharyya, S., et al., Risk factors for the development and progression of carcinoid heart disease. Am J Cardiol, 2011. 107(8): p. 1221-6.
  37. de Herder, W.W. and J. Hofland, Vasoactive Intestinal Peptide-Secreting Tumor (VIPoma), in Endotext, K.R. Feingold, et al., Editors. 2000: South Dartmouth (MA).
  38. de Herder, W.W. and J. Hofland, Somatostatinoma, in Endotext, K.R. Feingold, et al., Editors. 2000: South Dartmouth (MA).
  39. Tsoli, M., et al., Paraneoplastic Syndromes Related to Neuroendocrine Tumors, in Endotext, K.R. Feingold, et al., Editors. 2000: South Dartmouth (MA).
  40. Mastracci, L., et al., Neuroendocrine neoplasms of the esophagus and stomach. Pathologica, 2021. 113(1): p. 5-11.
  41. Rindi, G., et al., Three subtypes of gastric argyrophil carcinoid and the gastric neuroendocrine carcinoma: a clinicopathologic study. Gastroenterology, 1993. 104(4): p. 994-1006.
  42. Fossmark, R., et al., ECL-cell carcinoids and carcinoma in patients homozygous for an inactivating mutation in the gastric H(+) K(+) ATPase alpha subunit. APMIS, 2016. 124(7): p. 561-6.
  43. Calvete, O., et al., Exome sequencing identifies ATP4A gene as responsible of an atypical familial type I gastric neuroendocrine tumour. Hum Mol Genet, 2015. 24(10): p. 2914-22.
  44. Sato, Y., et al., Gastric carcinoid tumors without autoimmune gastritis in Japan: a relationship with Helicobacter pylori infection. Dig Dis Sci, 2002. 47(3): p. 579-85.
  45. Trinh, V.Q., C. Shi, and C. Ma, Gastric neuroendocrine tumours from long-term proton pump inhibitor users are indolent tumours with good prognosis. Histopathology, 2020. 77(6): p. 865-876.
  46. Norton, J.A., et al., Gastric carcinoid tumors in multiple endocrine neoplasia-1 patients with Zollinger-Ellison syndrome can be symptomatic, demonstrate aggressive growth, and require surgical treatment. Surgery, 2004. 136(6): p. 1267-74.
  47. Campana, D., et al., Clinical management of patients with gastric neuroendocrine neoplasms associated with chronic atrophic gastritis: a retrospective, multicentre study. Endocrine, 2016. 51(1): p. 131-9.
  48. Rindi, G., et al., Gastric carcinoids and neuroendocrine carcinomas: pathogenesis, pathology, and behavior. World J Surg, 1996. 20(2): p. 168-72.
  49. Thomas, D., et al., Long-term follow-up of a large series of patients with type 1 gastric carcinoid tumors: data from a multicenter study. Eur J Endocrinol, 2013. 168(2): p. 185-93.
  50. Grozinsky-Glasberg, S., et al., Metastatic type 1 gastric carcinoid: a real threat or just a myth? World J Gastroenterol, 2013. 19(46): p. 8687-95.
  51. Exarchou, K., et al., Is local excision sufficient in selected grade 1 or 2 type III gastric neuroendocrine neoplasms? Endocrine, 2021. 74(2): p. 421-429.
  52. Min, B.H., et al., Clinicopathological features and outcome of type 3 gastric neuroendocrine tumours. Br J Surg, 2018. 105(11): p. 1480-1486.
  53. Zandee, W.T., et al., Ghrelinoma, in Endotext, K.R. Feingold, et al., Editors. 2000: South Dartmouth (MA).
  54. Hoffmann, K.M., M. Furukawa, and R.T. Jensen, Duodenal neuroendocrine tumors: Classification, functional syndromes, diagnosis and medical treatment. Best Pract Res Clin Gastroenterol, 2005. 19(5): p. 675-97.
  55. Massironi, S., et al., Heterogeneity of Duodenal Neuroendocrine Tumors: An Italian Multi-center Experience. Ann Surg Oncol, 2018. 25(11): p. 3200-3206.
  56. Margonis, G.A., et al., A Multi-institutional Analysis of Duodenal Neuroendocrine Tumors: Tumor Biology Rather than Extent of Resection Dictates Prognosis. J Gastrointest Surg, 2016. 20(6): p. 1098-105.
  57. Giannakodimos, I., et al., Somatostatinoma of the Ampulla of Vater: A Systematic Review. J Gastrointestin Liver Dis, 2022. 31(4): p. 459-466.
  58. Barsouk, A., et al., Epidemiology of Cancers of the Small Intestine: Trends, Risk Factors, and Prevention. Med Sci (Basel), 2019. 7(3).
  59. Snorradottir, S., et al., Incidence and prognosis of patients with small intestinal neuroendocrine tumors in a population based nationwide study. Cancer Epidemiol, 2022. 79: p. 102197.
  60. Sei, Y., et al., Role of an active reserve stem cell subset of enteroendocrine cells in intestinal stem cell dynamics and the genesis of small intestinal neuroendocrine tumors. Am J Physiol Gastrointest Liver Physiol, 2020. 319(4): p. G494-G501.
  61. Blazevic, A., et al., Small intestinal neuroendocrine tumours and fibrosis: an entangled conundrum. Endocr Relat Cancer, 2018. 25(3): p. R115-R130.
  62. Blazevic, A., et al., Evolution of the Mesenteric Mass in Small Intestinal Neuroendocrine Tumours. Cancers (Basel), 2021. 13(3).
  63. Blazevic, A., et al., Mesenteric fibrosis and palliative surgery in small intestinal neuroendocrine tumours. Endocr Relat Cancer, 2018. 25(3): p. 245-254.
  64. Refardt, J., et al., Prognostic significance of hyperammonemia in neuroendocrine neoplasm patients with liver metastases. Endocr Relat Cancer, 2022. 29(5): p. 241-250.
  65. Pape, U.F., et al., ENETS Consensus Guidelines for Neuroendocrine Neoplasms of the Appendix (Excluding Goblet Cell Carcinomas). Neuroendocrinology, 2016. 103(2): p. 144-52.
  66. Nesti, C., et al., Hemicolectomy versus appendectomy for patients with appendiceal neuroendocrine tumours 1-2 cm in size: a retrospective, Europe-wide, pooled cohort study. Lancet Oncol, 2023. 24(2): p. 187-194.
  67. Rinke, A., et al., European Neuroendocrine Tumor Society (ENETS) 2023 guidance paper for colorectal neuroendocrine tumours. J Neuroendocrinol, 2023. 35(6): p. e13309.
  68. Gallo, C., et al., Rectal neuroendocrine tumors: Current advances in management, treatment, and surveillance. World J Gastroenterol, 2022. 28(11): p. 1123-1138.
  69. Cope, J. and R. Srirajaskanthan, Rectal Neuroendocrine Neoplasms: Why Is There a Global Variation? Curr Oncol Rep, 2022. 24(3): p. 257-263.
  70. Zhao, B., et al., Outcomes for a Large Cohort of Patients with Rectal Neuroendocrine Tumors: an Analysis of the National Cancer Database. J Gastrointest Surg, 2021. 25(2): p. 484-491.
  71. Perren, A., et al., ENETS Consensus Guidelines for the Standards of Care in Neuroendocrine Tumors: Pathology: Diagnosis and Prognostic Stratification. Neuroendocrinology, 2017. 105(3): p. 196-200.
  72. Lloyd, R.V., Practical markers used in the diagnosis of neuroendocrine tumors. Endocr Pathol, 2003. 14(4): p. 293-301.
  73. Merola, E., et al., Histopathological Revision for Gastroenteropancreatic Neuroendocrine Neoplasms in Expert Centers: Does It Make the Difference? Neuroendocrinology, 2021. 111(1-2): p. 170-177.
  74. Elvebakken, H., et al., A Consensus-Developed Morphological Re-Evaluation of 196 High-Grade Gastroenteropancreatic Neuroendocrine Neoplasms and Its Clinical Correlations. Neuroendocrinology, 2021. 111(9): p. 883-894.
  75. Merola, E., et al., High rate of Ki-67 increase in entero-pancreatic NET relapses after surgery with curative intent. J Neuroendocrinol, 2022. 34(10): p. e13193.
  76. Grillo, F., et al., Grade Increases in Gastroenteropancreatic Neuroendocrine Tumor Metastases Compared to the Primary Tumor. Neuroendocrinology, 2016. 103(5): p. 452-9.
  77. Kazmierczak, P.M., et al., The added value of (68)Ga-DOTA-TATE-PET to contrast-enhanced CT for primary site detection in CUP of neuroendocrine origin. Eur Radiol, 2017. 27(4): p. 1676-1684.
  78. Barbareschi, M., et al., CDX-2 homeobox gene product expression in neuroendocrine tumors: its role as a marker of intestinal neuroendocrine tumors. Am J Surg Pathol, 2004. 28(9): p. 1169-76.
  79. Koo, J., et al., Value of Islet 1 and PAX8 in identifying metastatic neuroendocrine tumors of pancreatic origin. Mod Pathol, 2012. 25(6): p. 893-901.
  80. Srivastava, A. and J.L. Hornick, Immunohistochemical staining for CDX-2, PDX-1, NESP-55, and TTF-1 can help distinguish gastrointestinal carcinoid tumors from pancreatic endocrine and pulmonary carcinoid tumors. Am J Surg Pathol, 2009. 33(4): p. 626-32.
  81. Mohanty, S.K., et al., Positivity for SATB2 distinguishes Islet1 positive rectal neuroendocrine tumours from pancreaticoduodenal neuroendocrine tumours. J Clin Pathol, 2021. 74(9): p. 582-588.
  82. Oberg, K., et al., Consensus on biomarkers for neuroendocrine tumour disease. Lancet Oncol, 2015. 16(9): p. e435-e446.
  83. Molina, R., et al., Evaluation of chromogranin A determined by three different procedures in patients with benign diseases, neuroendocrine tumors and other malignancies. Tumour Biol, 2011. 32(1): p. 13-22.
  84. van Balveren, J.A., et al., Awareness of drug laboratory test interactions is important for prevention of unnecessary additional diagnostics: An example. Clin Chim Acta, 2022. 530: p. 99-103.
  85. Nobels, F.R., et al., Chromogranin A as serum marker for neuroendocrine neoplasia: comparison with neuron-specific enolase and the alpha-subunit of glycoprotein hormones. J Clin Endocrinol Metab, 1997. 82(8): p. 2622-8.
  86. Baudin, E., et al., Neuron-specific enolase and chromogranin A as markers of neuroendocrine tumours. Br J Cancer, 1998. 78(8): p. 1102-7.
  87. Modlin, I.M., et al., The clinical utility of a novel blood-based multi-transcriptome assay for the diagnosis of neuroendocrine tumors of the gastrointestinal tract. Am J Gastroenterol, 2015. 110(8): p. 1223-32.
  88. Oberg, K., et al., A meta-analysis of the accuracy of a neuroendocrine tumor mRNA genomic biomarker (NETest) in blood. Ann Oncol, 2020. 31(2): p. 202-212.
  89. van Treijen, M.J.C., et al., NETest: serial liquid biopsies in gastroenteropancreatic NET surveillance. Endocr Connect, 2022. 11(10).
  90. de Herder, W.W. and J. Hofland, Insulinoma, in Endotext, K.R. Feingold, et al., Editors. 2000: South Dartmouth (MA).
  91. de Herder, W.W. and J. Hofland, Glucagon & Glucagonoma Syndrome, in Endotext, K.R. Feingold, et al., Editors. 2000: South Dartmouth (MA).
  92. O'Toole, D., et al., ENETS Consensus Guidelines for the Standards of Care in Neuroendocrine Tumors: biochemical markers. Neuroendocrinology, 2009. 90(2): p. 194-202.
  93. Zandee, W.T., et al., Limited value for urinary 5-HIAA excretion as prognostic marker in gastrointestinal neuroendocrine tumours. Eur J Endocrinol, 2016. 175(5): p. 361-6.
  94. Janson, E.T., et al., Carcinoid tumors: analysis of prognostic factors and survival in 301 patients from a referral center. Ann Oncol, 1997. 8(7): p. 685-90.
  95. Turner, G.B., et al., Circulating markers of prognosis and response to treatment in patients with midgut carcinoid tumours. Gut, 2006. 55(11): p. 1586-91.
  96. de Mestier, L., et al., Plasmatic and Urinary 5-Hydroxyindolacetic Acid Measurements in Patients With Midgut Neuroendocrine Tumors: A GTE Study. J Clin Endocrinol Metab, 2021. 106(4): p. e1673-e1682.
  97. Carling, R.S., et al., Evaluation of whole blood serotonin and plasma and urine 5-hydroxyindole acetic acid in diagnosis of carcinoid disease. Ann Clin Biochem, 2002. 39(Pt 6): p. 577-82.
  98. Kema, I.P., et al., Improved diagnosis of carcinoid tumors by measurement of platelet serotonin. Clin Chem, 1992. 38(4): p. 534-40.
  99. Dobson, R., et al., The association of a panel of biomarkers with the presence and severity of carcinoid heart disease: a cross-sectional study. PLoS One, 2013. 8(9): p. e73679.
  100. Bhattacharyya, S., et al., Usefulness of N-terminal pro-brain natriuretic peptide as a biomarker of the presence of carcinoid heart disease. Am J Cardiol, 2008. 102(7): p. 938-42.
  101. Shinya, T., et al., Small bowel neoplasms: enhancement patterns and differentiation using post-contrast multiphasic multidetector CT. Abdom Radiol (NY), 2017. 42(3): p. 794-801.
  102. Sundin, A., et al., ENETS Consensus Guidelines for the Standards of Care in Neuroendocrine Tumors: Radiological, Nuclear Medicine & Hybrid Imaging. Neuroendocrinology, 2017. 105(3): p. 212-244.
  103. Ronot, M., et al., Neuroendocrine liver metastases: Vascular patterns on triple-phase MDCT are indicative of primary tumour location. Eur J Radiol, 2017. 89: p. 156-162.
  104. Hayoz, R., et al., The combination of hepatobiliary phase with Gd-EOB-DTPA and DWI is highly accurate for the detection and characterization of liver metastases from neuroendocrine tumor. Eur Radiol, 2020. 30(12): p. 6593-6602.
  105. Dromain, C., et al., Detection of liver metastases from endocrine tumors: a prospective comparison of somatostatin receptor scintigraphy, computed tomography, and magnetic resonance imaging. J Clin Oncol, 2005. 23(1): p. 70-8.
  106. Haider, M., et al., Use of MRI and Ga-68 DOTATATE for the detection of neuroendocrine liver metastases. Abdom Radiol (NY), 2022. 47(2): p. 586-595.
  107. Srirajaskanthan, R., et al., Optimising Outcomes and Surveillance Strategies of Rectal Neuroendocrine Neoplasms. Cancers (Basel), 2023. 15(10).
  108. Panzuto, F., et al., European Neuroendocrine Tumor Society (ENETS) 2023 guidance paper for gastroduodenal neuroendocrine tumours (NETs) G1-G3. J Neuroendocrinol, 2023: p. e13306.
  109. Hofland, L.J. and S.W. Lamberts, The pathophysiological consequences of somatostatin receptor internalization and resistance. Endocr Rev, 2003. 24(1): p. 28-47.
  110. Krenning, E.P., et al., Localisation of endocrine-related tumours with radioiodinated analogue of somatostatin. Lancet, 1989. 1(8632): p. 242-4.
  111. Barrio, M., et al., The Impact of Somatostatin Receptor-Directed PET/CT on the Management of Patients with Neuroendocrine Tumor: A Systematic Review and Meta-Analysis. J Nucl Med, 2017. 58(5): p. 756-761.
  112. Graham, M.M., et al., 68Ga-DOTATOC Imaging of Neuroendocrine Tumors: A Systematic Review and Metaanalysis. J Nucl Med, 2017. 58(9): p. 1452-1458.
  113. Hicks, R.J., et al., (64)Cu-SARTATE PET Imaging of Patients with Neuroendocrine Tumors Demonstrates High Tumor Uptake and Retention, Potentially Allowing Prospective Dosimetry for Peptide Receptor Radionuclide Therapy. J Nucl Med, 2019. 60(6): p. 777-785.
  114. Pauwels, E., et al., (18)F-AlF-NOTA-Octreotide Outperforms (68)Ga-DOTATATE/NOC PET in Neuroendocrine Tumor Patients: Results from a Prospective, Multicenter Study. J Nucl Med, 2023. 64(4): p. 632-638.
  115. Haug, A., et al., Intraindividual comparison of 68Ga-DOTA-TATE and 18F-DOPA PET in patients with well-differentiated metastatic neuroendocrine tumours. Eur J Nucl Med Mol Imaging, 2009. 36(5): p. 765-70.
  116. Chan, D.L., et al., Dual Somatostatin Receptor/FDG PET/CT Imaging in Metastatic Neuroendocrine Tumours: Proposal for a Novel Grading Scheme with Prognostic Significance. Theranostics, 2017. 7(5): p. 1149-1158.
  117. Squires, M.H., 3rd, et al., Octreoscan Versus FDG-PET for Neuroendocrine Tumor Staging: A Biological Approach. Ann Surg Oncol, 2015. 22(7): p. 2295-301.
  118. Ezziddin, S., et al., Prognostic stratification of metastatic gastroenteropancreatic neuroendocrine neoplasms by 18F-FDG PET: feasibility of a metabolic grading system. J Nucl Med, 2014. 55(8): p. 1260-6.
  119. Singh, S., et al., Recurrence in Resected Gastroenteropancreatic Neuroendocrine Tumors. JAMA Oncol, 2018. 4(4): p. 583-585.
  120. Zhang, H.P., et al., Endoscopic treatments for rectal neuroendocrine tumors smaller than 16 mm: a meta-analysis. Scand J Gastroenterol, 2016. 51(11): p. 1345-53.
  121. Noh, J.H., et al., Clinical Outcomes of Endoscopic Treatment for Type 1 Gastric Neuroendocrine Tumor. J Gastrointest Surg, 2021. 25(10): p. 2495-2502.
  122. Brand, M., et al., Endoscopic full thickness resection vs. transanal endoscopic microsurgery for local treatment of rectal neuroendocrine tumors - a retrospective analysis. Int J Colorectal Dis, 2021. 36(5): p. 971-976.
  123. Frilling, A., et al., Recommendations for management of patients with neuroendocrine liver metastases. Lancet Oncol, 2014. 15(1): p. e8-21.
  124. Elias, D., et al., Hepatic metastases from neuroendocrine tumors with a "thin slice" pathological examination: they are many more than you think. Ann Surg, 2010. 251(2): p. 307-10.
  125. Gibson, W.E., et al., Hepatic micrometastases are associated with poor prognosis in patients with liver metastases from neuroendocrine tumors of the digestive tract. Hum Pathol, 2018. 79: p. 109-115.
  126. Eriksson, J., et al., Surgery and radiofrequency ablation for treatment of liver metastases from midgut and foregut carcinoids and endocrine pancreatic tumors. World J Surg, 2008. 32(5): p. 930-8.
  127. Gaujoux, S., et al., Synchronous resection of primary and liver metastases for neuroendocrine tumors. Ann Surg Oncol, 2012. 19(13): p. 4270-7.
  128. Norton, J.A., et al., Aggressive surgery for metastatic liver neuroendocrine tumors. Surgery, 2003. 134(6): p. 1057-63; discussion 1063-5.
  129. Zappa, M., et al., Liver-directed therapies in liver metastases from neuroendocrine tumors of the gastrointestinal tract. Target Oncol, 2012. 7(2): p. 107-16.
  130. Levy, S., et al., Primary Tumor Resection is Associated with Improved Disease-Specific Mortality in Patients with Stage IV Small Intestinal Neuroendocrine Tumors (NETs): A Comparison of Upfront Surgical Resection Versus a Watch and Wait Strategy in Two Specialist NET Centers. Ann Surg Oncol, 2022. 29(12): p. 7822-7832.
  131. Norlen, O., et al., Long-term results of surgery for small intestinal neuroendocrine tumors at a tertiary referral center. World J Surg, 2012. 36(6): p. 1419-31.
  132. Soreide, O., et al., Surgical treatment as a principle in patients with advanced abdominal carcinoid tumors. Surgery, 1992. 111(1): p. 48-54.
  133. Daskalakis, K., et al., Association of a Prophylactic Surgical Approach to Stage IV Small Intestinal Neuroendocrine Tumors With Survival. JAMA Oncol, 2018. 4(2): p. 183-189.
  134. Rinke, A., et al., Placebo-controlled, double-blind, prospective, randomized study on the effect of octreotide LAR in the control of tumor growth in patients with metastatic neuroendocrine midgut tumors: a report from the PROMID Study Group. J Clin Oncol, 2009. 27(28): p. 4656-63.
  135. Caplin, M.E., et al., Lanreotide in metastatic enteropancreatic neuroendocrine tumors. N Engl J Med, 2014. 371(3): p. 224-33.
  136. Kvols, L.K., et al., Treatment of the malignant carcinoid syndrome. Evaluation of a long-acting somatostatin analogue. N Engl J Med, 1986. 315(11): p. 663-6.
  137. Caplin, M.E., et al., Anti-tumour effects of lanreotide for pancreatic and intestinal neuroendocrine tumours: the CLARINET open-label extension study. Endocr Relat Cancer, 2016. 23(3): p. 191-9.
  138. Sinnamon, A.J., et al., Prophylactic Cholecystectomy at Time of Surgery for Small Bowel Neuroendocrine Tumor Does Not Increase Postoperative Morbidity. Ann Surg Oncol, 2018. 25(1): p. 239-245.
  139. Strosberg, J.R., et al., Clinical benefits of above-standard dose of octreotide LAR in patients with neuroendocrine tumors for control of carcinoid syndrome symptoms: a multicenter retrospective chart review study. Oncologist, 2014. 19(9): p. 930-6.
  140. Strosberg, J., et al., Phase 3 Trial of 177Lu-Dotatate for Midgut Neuroendocrine Tumors. N Engl J Med, 2017. 376(2): p. 125-135.
  141. Pavel, M., et al., Efficacy and safety of high-dose lanreotide autogel in patients with progressive pancreatic or midgut neuroendocrine tumours: CLARINET FORTE phase 2 study results. Eur J Cancer, 2021. 157: p. 403-414.
  142. Hofland, J., et al., European Neuroendocrine Tumor Society 2023 guidance paper for functioning pancreatic neuroendocrine tumour syndromes. J Neuroendocrinol, 2023: p. e13318.
  143. Strosberg, J.R., et al., The North American Neuroendocrine Tumor Society Consensus Guidelines for Surveillance and Medical Management of Midgut Neuroendocrine Tumors. Pancreas, 2017. 46(6): p. 707-714.
  144. Kvols, L.K., et al., Pasireotide (SOM230) shows efficacy and tolerability in the treatment of patients with advanced neuroendocrine tumors refractory or resistant to octreotide LAR: results from a phase II study. Endocr Relat Cancer, 2012. 19(5): p. 657-66.
  145. Kulke, M.H., et al., A randomized, open-label, phase 2 study of everolimus in combination with pasireotide LAR or everolimus alone in advanced, well-differentiated, progressive pancreatic neuroendocrine tumors: COOPERATE-2 trial. Ann Oncol, 2017. 28(6): p. 1309-1315.
  146. Ferolla, P., et al., Efficacy and safety of long-acting pasireotide or everolimus alone or in combination in patients with advanced carcinoids of the lung and thymus (LUNA): an open-label, multicentre, randomised, phase 2 trial. Lancet Oncol, 2017. 18(12): p. 1652-1664.
  147. Kwekkeboom, D.J., et al., Radiolabeled somatostatin analog (177Lu-DOTA0,Tyr3)octreotate in patients with endocrine gastroenteropancreatic tumors. J Clin Oncol, 2005. 23(12): p. 2754-62.
  148. Hicks, R.J., et al., ENETS Consensus Guidelines for the Standards of Care in Neuroendocrine Neoplasia: Peptide Receptor Radionuclide Therapy with Radiolabeled Somatostatin Analogues. Neuroendocrinology, 2017. 105(3): p. 295-309.
  149. Sonbol, M.B., T.R. Halfdanarson, and T. Hilal, Assessment of Therapy-Related Myeloid Neoplasms in Patients With Neuroendocrine Tumors After Peptide Receptor Radionuclide Therapy: A Systematic Review. JAMA Oncol, 2020. 6(7): p. 1086-1092.
  150. Strosberg, J.R., et al., (177)Lu-Dotatate plus long-acting octreotide versus high‑dose long-acting octreotide in patients with midgut neuroendocrine tumours (NETTER-1): final overall survival and long-term safety results from an open-label, randomised, controlled, phase 3 trial. Lancet Oncol, 2021. 22(12): p. 1752-1763.
  151. Strosberg, J., et al., Health-Related Quality of Life in Patients With Progressive Midgut Neuroendocrine Tumors Treated With (177)Lu-Dotatate in the Phase III NETTER-1 Trial. J Clin Oncol, 2018. 36(25): p. 2578-2584.
  152. Brabander, T., et al., Long-Term Efficacy, Survival, and Safety of (177Lu-DOTA0,Tyr3)octreotate in Patients with Gastroenteropancreatic and Bronchial Neuroendocrine Tumors. Clin Cancer Res, 2017. 23(16): p. 4617-4624.
  153. Becx, M.N., et al., A Clinical Guide to Peptide Receptor Radionuclide Therapy with (177)Lu-DOTATATE in Neuroendocrine Tumor Patients. Cancers (Basel), 2022. 14(23).
  154. Zandee, W.T., et al., Symptomatic and Radiological Response to 177Lu-DOTATATE for the Treatment of Functioning Pancreatic Neuroendocrine Tumors. J Clin Endocrinol Metab, 2019. 104(4): p. 1336-1344.
  155. Zandee, W.T., et al., Peptide Receptor Radionuclide Therapy With 177Lu-DOTATATE for Symptomatic Control of Refractory Carcinoid Syndrome. J Clin Endocrinol Metab, 2021. 106(9): p. e3665-e3672.
  156. van der Zwan, W.A., et al., Salvage peptide receptor radionuclide therapy with ((177)Lu-DOTA,Tyr(3))octreotate in patients with bronchial and gastroenteropancreatic neuroendocrine tumours. Eur J Nucl Med Mol Imaging, 2019. 46(3): p. 704-717.
  157. Pavel, M.E., et al., Everolimus plus octreotide long-acting repeatable for the treatment of advanced neuroendocrine tumours associated with carcinoid syndrome (RADIANT-2): a randomised, placebo-controlled, phase 3 study. Lancet, 2011. 378(9808): p. 2005-2012.
  158. Yao, J.C., et al., Everolimus for the treatment of advanced, non-functional neuroendocrine tumours of the lung or gastrointestinal tract (RADIANT-4): a randomised, placebo-controlled, phase 3 study. Lancet, 2016. 387(10022): p. 968-77.
  159. Pavel, M.E., et al., Health-related quality of life for everolimus versus placebo in patients with advanced, non-functional, well-differentiated gastrointestinal or lung neuroendocrine tumours (RADIANT-4): a multicentre, randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol, 2017. 18(10): p. 1411-1422.
  160. Pavel, M., et al., Safety and QOL in Patients with Advanced NET in a Phase 3b Expanded Access Study of Everolimus. Target Oncol, 2016. 11(5): p. 667-675.
  161. Kulke, M.H., et al., Activity of sunitinib in patients with advanced neuroendocrine tumors. J Clin Oncol, 2008. 26(20): p. 3403-10.
  162. Raymond, E., et al., Sunitinib malate for the treatment of pancreatic neuroendocrine tumors. N Engl J Med, 2011. 364(6): p. 501-13.
  163. Xu, J., et al., Surufatinib in advanced pancreatic neuroendocrine tumours (SANET-p): a randomised, double-blind, placebo-controlled, phase 3 study. Lancet Oncol, 2020. 21(11): p. 1489-1499.
  164. Xu, J., et al., Surufatinib in advanced extrapancreatic neuroendocrine tumours (SANET-ep): a randomised, double-blind, placebo-controlled, phase 3 study. Lancet Oncol, 2020. 21(11): p. 1500-1512.
  165. Li, J., et al., Health-related quality of life in patients with advanced well-differentiated pancreatic and extrapancreatic neuroendocrine tumors treated with surufatinib versus placebo: Results from two randomized, double-blind, phase III trials (SANET-p and SANET-ep). Eur J Cancer, 2022. 169: p. 1-9.
  166. Grande, E., et al., Pazopanib in pretreated advanced neuroendocrine tumors: a phase II, open-label trial of the Spanish Task Force Group for Neuroendocrine Tumors (GETNE). Ann Oncol, 2015. 26(9): p. 1987-1993.
  167. Capdevila, J., et al., Lenvatinib in Patients With Advanced Grade 1/2 Pancreatic and Gastrointestinal Neuroendocrine Tumors: Results of the Phase II TALENT Trial (GETNE1509). J Clin Oncol, 2021. 39(20): p. 2304-2312.
  168. Strosberg, J.R., et al., A phase II study of axitinib in advanced neuroendocrine tumors. Endocr Relat Cancer, 2016. 23(5): p. 411-8.
  169. Moertel, C.G., J. Rubin, and L.K. Kvols, Therapy of metastatic carcinoid tumor and the malignant carcinoid syndrome with recombinant leukocyte A interferon. J Clin Oncol, 1989. 7(7): p. 865-8.
  170. Oberg, K., et al., Treatment of malignant carcinoid tumors with human leukocyte interferon: long-term results. Cancer Treat Rep, 1986. 70(11): p. 1297-304.
  171. Faiss, S., et al., Prospective, randomized, multicenter trial on the antiproliferative effect of lanreotide, interferon alfa, and their combination for therapy of metastatic neuroendocrine gastroenteropancreatic tumors--the International Lanreotide and Interferon Alfa Study Group. J Clin Oncol, 2003. 21(14): p. 2689-96.
  172. Pavel, M.E., et al., Efficacy and tolerability of pegylated IFN-alpha in patients with neuroendocrine gastroenteropancreatic carcinomas. J Interferon Cytokine Res, 2006. 26(1): p. 8-13.
  173. Cives, M., et al., Analysis of the immune landscape of small bowel neuroendocrine tumors. Endocr Relat Cancer, 2019. 26(1): p. 119-130.
  174. Vesely, C., et al., Systematic Evaluation of the Immune Environment of Small Intestinal Neuroendocrine Tumors. Clin Cancer Res, 2022. 28(12): p. 2657-2668.
  175. Bosch, F., et al., Immune checkpoint markers in gastroenteropancreatic neuroendocrine neoplasia. Endocr Relat Cancer, 2019. 26(3): p. 293-301.
  176. Strosberg, J., et al., Efficacy and Safety of Pembrolizumab in Previously Treated Advanced Neuroendocrine Tumors: Results From the Phase II KEYNOTE-158 Study. Clin Cancer Res, 2020. 26(9): p. 2124-2130.
  177. Patel, S.P., et al., A Phase II Basket Trial of Dual Anti-CTLA-4 and Anti-PD-1 Blockade in Rare Tumors (DART SWOG 1609) in Patients with Nonpancreatic Neuroendocrine Tumors. Clin Cancer Res, 2020. 26(10): p. 2290-2296.
  178. Klein, O., et al., Immunotherapy of Ipilimumab and Nivolumab in Patients with Advanced Neuroendocrine Tumors: A Subgroup Analysis of the CA209-538 Clinical Trial for Rare Cancers. Clin Cancer Res, 2020. 26(17): p. 4454-4459.
  179. Lamarca, A., et al., Chemotherapy for advanced non-pancreatic well-differentiated neuroendocrine tumours of the gastrointestinal tract, a systematic review and meta-analysis: A lost cause? Cancer Treat Rev, 2016. 44: p. 26-41.
  180. Pavel, M., et al., ENETS Consensus Guidelines Update for the Management of Distant Metastatic Disease of Intestinal, Pancreatic, Bronchial Neuroendocrine Neoplasms (NEN) and NEN of Unknown Primary Site. Neuroendocrinology, 2016. 103(2): p. 172-85.
  181. Pavel, M., et al., Gastroenteropancreatic neuroendocrine neoplasms: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol, 2020. 31(7): p. 844-860.
  182. Clement, D., et al., Prevalence of Sarcopenia and Impact on Survival in Patients with Metastatic Gastroenteropancreatic Neuroendocrine Tumours. Cancers (Basel), 2023. 15(3).
  183. Clement, D., et al., Outcomes and survival in patients with advanced intestinal neuroendocrine tumours on home parenteral nutrition, an international multicentre retrospective cohort study. Clin Nutr ESPEN, 2023. 54: p. 106-112.
  184. Del Rivero, J., et al., Practical considerations when providing palliative care to patients with neuroendocrine tumors in the context of routine disease management or hospice care. Endocr Relat Cancer, 2023. 30(7).
  185. Beaumont, J.L., et al., Comparison of health-related quality of life in patients with neuroendocrine tumors with quality of life in the general US population. Pancreas, 2012. 41(3): p. 461-6.
  186. Hofland, J., et al., Management of carcinoid syndrome: a systematic review and meta-analysis. Endocr Relat Cancer, 2019: p. doi: 10.1530/ERC-18-0495.
  187. Kaltsas, G., et al., ENETS Consensus Guidelines for the Standards of Care in Neuroendocrine Tumors: Pre- and Perioperative Therapy in Patients with Neuroendocrine Tumors. Neuroendocrinology, 2017. 105(3): p. 245-254.
  188. Kulke, M.H., et al., Telotristat Ethyl, a Tryptophan Hydroxylase Inhibitor for the Treatment of Carcinoid Syndrome. J Clin Oncol, 2017. 35(1): p. 14-23.
  189. Arnold, R., et al., Octreotide versus octreotide plus interferon-alpha in endocrine gastroenteropancreatic tumors: a randomized trial. Clin Gastroenterol Hepatol, 2005. 3(8): p. 761-71.
  190. Hofland, J., et al., Synoptic reporting of echocardiography in carcinoid heart disease (ENETS Carcinoid Heart Disease Task Force). J Neuroendocrinol, 2022. 34(3): p. e13060.
  191. Davar, J., et al., Diagnosing and Managing Carcinoid Heart Disease in Patients With Neuroendocrine Tumors: An Expert Statement. J Am Coll Cardiol, 2017. 69(10): p. 1288-1304.
  192. Sonbol, M.B., et al., Causes of Death After Neuroendocrine Tumors Diagnosis: A US Population-Based Analysis. Pancreas, 2021. 50(1): p. 47-53.
  193. Hallet, J., et al., Risk of Cancer-Specific Death for Patients Diagnosed With Neuroendocrine Tumors: A Population-Based Analysis. J Natl Compr Canc Netw, 2021. 19(8): p. 935-944.
  194. Polee, I.N., et al., Long-term survival in patients with gastroenteropancreatic neuroendocrine neoplasms: A population-based study. Eur J Cancer, 2022. 172: p. 252-263.

 

Hypoglycemia in Neonates, Infants, and Children

ABSTRACT

 

Hypoglycemia in neonates, infants and children should be considered a medical emergency that can cause seizures, permanent neurological injury, and in rare cases, death, if inadequately treated. Under normal conditions, glucose is the primary fuel for brain metabolism. Due to the metabolic demands of the developing brain, infants and children have increased rates of glucose utilization as compared to adults. Normal regulation of glucose during fasting requires integration of glycogenolysis, gluconeogenesis, and fatty acid oxidation coordinated by various hormones. In the first days of life, the time course of the physiologic transitional changes to autonomous glucose regulation may overlap with presentation of inherited and acquired pathologic forms of hypoglycemia, introducing inherent challenges and controversy in addressing neonatal hypoglycemia. Therefore, a careful approach to the neonate with hypoglycemia is critical to determine the precise etiology so that rapid and appropriate interventions can be implemented to avoid permanent neurological injury. After infancy, hypoglycemia is uncommon, but, up to 10% of children older than one year of age presenting to emergency rooms with previously undiagnosed hypoglycemia have a serious underlying condition requiring long-term treatment. An important caveat in the pediatric population is that the classical definition of hypoglycemia, Whipple’s triad, is of limited use as young children are unable to reliably communicate symptoms. At all ages, determining the cause of the hypoglycemia is paramount for establishing specific and effective treatment to prevent further episodes of hypoglycemia and long-term neurological sequelae. The majority of hypoglycemic events in infants and children with hypoglycemic disorders occur during periods of fasting. Evaluation of key metabolic fuels and hormones (the critical sample) during a supervised fast, or at the time of spontaneous hypoglycemia, thus permits classification and relevant treatment of hypoglycemia disorders.

 

INTRODUCTION

 

Hypoglycemia is the biochemical finding of a plasma glucose lower than normal for age.  Hypoglycemia itself is not a diagnosis but rather reflects an underlying perturbation in metabolic adaptation, which may be as simple as prolonged fasting in a child with an intercurrent illness, or a complex genetic disorder. It is critical for the physician at all times to determine the etiology of hypoglycemia while at the same time treating critical low blood glucose and stabilizing the person with hypoglycemia.

 

In this chapter, we outline the basic physiology of glucose regulation, the change in glucose homeostasis in the immediate newborn period and describe the common diseases that cause hypoglycemia in childhood. We discuss the definitions of hypoglycemia, pitfalls in the measurement of blood glucose concentration, the critical importance of glucose as a fuel for energy in the brain, and the implications of hypoglycemia on brain damage. For information on hypoglycemia related to diabetes and its treatment see the chapter entitled “Hypoglycemia During Therapy of Diabetes” in the Diabetes Mellitus and Carbohydrate Metabolism---Diabetes Manager section of Endotext (1).

 

PHYSIOLOGY OF GLUCOSE REGULATION

 

An understanding of the physiology of glucose regulation is critical in order to understand the etiology of the different hypoglycemic disorders. By utilizing a fasting systems approach to diagnosis, one can rapidly pinpoint the general physiological system disrupted and with appropriate examination of blood and urine at the time of hypoglycemia (the critical sample) one can identify the diagnosis relatively quickly. This approach is particularly useful because apart from the newborn period (2), the vast majority of causes of hypoglycemia in children are caused by abnormalities of fasting adaptation. Rare exceptions to this rule include postprandial hypoglycemia typically related to gastrointestinal surgery such as Nissen fundoplication (3), or esophageal atresia repair (4), protein induced hypoglycemia of certain genetic forms of hyperinsulinism (5,6), and the hypoglycemia triggered by the ingestion of fructose in hereditary fructose intolerance (7).

 

The recognition and identification of the etiology of hypoglycemic disorders in the immediate newborn period may be more complicated due to the changes in glucose homeostasis that occur during the transition from intra uterine to extra uterine life, typically defined as the first 12-72 hours of life, and will be discussed separately (2).

 

The Physiology of Fasting

 

The key to diagnosing the etiology of hypoglycemia is a good understanding of the three key metabolic systems that regulate the physiological response to fasting and the hormonal control over these systems. The first, glycogenolysis, involves the breakdown of glycogen and conversion to glucose 6 phosphate (G-6-phos). G-6-phos may either undergo glycolysis and be converted to lactate (8), or converted to glucose by glucose-6-phosphatase (G-6-Pase), and then released from the liver and transported to the brain for fuel. The second, gluconeogenesis, is the pathway by which fuels such as lactate, alanine, fructose and glycerol are converted into glucose. The third, fatty acid oxidation and ketogenesis, are the processes by which ingested fat and fat stores are converted either to acetyl-CoA for entry into the citric acid cycle (Krebs cycle) and generation of energy, or to beta-hydroxybutyrate which is then transported to tissue such as the brain for energy production. An essential role of fatty acid oxidation is the production of an alternate fuel to glucose for energy production. This process conserves glucose for those tissues that can only metabolize glucose (such as red blood cells). Each of these mechanisms of fasting adaptation are finely regulated to maintain the plasma glucose between 70 and 110 mg/dL (3.9 to 6.1 mmol/L) by a combination of insulin to utilize or store glucose, and the counter regulatory hormones to mobilize and release glucose (Table 1). Insulin, the main hormone secreted in the fed state, suppresses glycogenolysis, gluconeogenesis, lipolysis, and ketogenesis. The counter regulatory hormones glucagon, cortisol, growth hormone, and epinephrine are the predominant hormones secreted in the fasting state and have overlapping effects on these processes. Because of the critical importance of these 3 processes (glycogenolysis, gluconeogenesis and fatty acid oxidation) in preventing hypoglycemia, there is overlap in hormonal control of the systems with both glucagon and epinephrine stimulating glycogenolysis, cortisol and glucagon stimulating gluconeogenesis, growth hormone and epinephrine stimulating lipolysis and finally glucagon and epinephrine stimulating ketogenesis. 

 

Table 1. Hormonal Control of Fasting Adaptation

Hormones

Glycogenolysis

Gluconeogenesis

Lipolysis

Ketogenesis

Cortisol

 

 stimulates

 

 

Growth Hormone

 

 

 stimulates

 

Glucagon

 stimulates

 stimulates

 

 stimulate

Epinephrine

 stimulates

 

 stimulates

 stimulates

 

Hormonal Control of Fasting

 

Following a meal, insulin is the predominant secreted hormone as glucose, and other fuels, are absorbed, facilitating glucose entry into cells and its utilization for energy. Under the influence of insulin, excess glucose is stored as glycogen in the liver for use later in fasting. As the plasma glucose starts to fall below 85 mg/dL insulin secretion is reduced and then suppressed (Figure 1). As the blood sugar continues to fall to around 70 mg/dL, the rapidly acting counter regulatory hormones glucagon and epinephrine are secreted. This initially drives glycogenolysis which supplies glucose from the liver for up to 4-8 hours in infants and 8-12 hours in children and young adults. Cortisol and growth hormone secretion stimulate gluconeogenesis and lipolysis providing amino acids, glycerol, and free fatty acids for metabolism to ketones.

Figure 1. Relationship of hormone secretion to plasma glucose levels.

 

Brain Fuel Metabolism/Energy Production

 

As the plasma glucose levels approach 50 mg/dL the ability of the blood brain barrier to transport glucose into the brain for maximal usage, begins to become limited (9). The brain responds by utilizing alternative fuels for energy (lactate, beta-hydroxybutyrate) or in the absence of alternate fuels (as occurs in hyperinsulinism) cuts down nonessential functions in order to conserve energy. When the glucose levels drop below 30 mg/dl cerebral blood flow increases in an attempt to increase glucose delivery (10). In conditions of prolonged fasting, as beta-hydroxybutyrate levels rise, monocarboxylate transporter 1 (MCT1) transports the beta-hydroxybutyrate across the blood brain barrier, which presents the brain with an alternative fuel for energy production. With very prolonged fasting, the brain is able to switch from utilizing almost 100% glucose for energy production to utilizing almost all ketones (11). In disease conditions such as glycogen storage disease (GSD) type 1, glycogenolysis results in elevated lactate levels because of the inability to de-phosphorylate G-6-phos and generate glucose that can be transported out of the liver. As a result, the plasma glucose falls rapidly, and the plasma lactate levels rise to 5-10 mmol/L (4-5 times normal levels). Carriers such as MCT1 transport this lactate across the blood brain barrier where the brain can efficiently metabolize it in the presence of oxygen through the citric acid cycle and produce the energy needed for brain function. Indeed, animal studies have demonstrated that the cerebral metabolic rate of oxygen consumption does not fall significantly during the initial phase of hypoglycemia when either ketones or lactate are available as an alternate substrate. In patients with diabetes, lactate infusions can prevent insulin induced neuroglycopenia (12). In addition, humans undergoing a ketogenic diet were able to transition from utilizing primarily glucose as brain fuel to ketones providing 17% of whole brain energy requirements with no overall change in the cerebral metabolic rate (13). It is for this reason that that hypoglycemia itself does not cause brain damage, it is a lack of fuel (glucose, beta-hydroxybutyrate or lactate combined) that causes brain damage. In addition to a lack of fuels for energy production a lack of oxygen supply to the brain prevents the utilization of lactate which can dramatically worsen energy failure in the brain.

 

In non-physiological conditions when the plasma glucose continues to drop down to 30 mg/dL or lower, cerebral spinal fluid glucose levels become almost zero and in the absence of either ketones or lactate essential functions of the brain stop and cell death begins to occur (14).  Breakdown of the phospholipids in the brain provides free fatty acids and breakdown of protein provides gluconeogenic substrates but also increases brain ammonia levels 10-15 fold (15). Whilst breakdown of tissues provides fuel for energy production, this process also damages vital cells and organelles causing permanent cellular destruction. Once the production of energy is insufficient to maintain adequate high energy phosphate compounds, the infant or child will become comatose and have an isoelectric electroencephalogram. It is not known exactly how long glucose and the alternate fuel levels need to be low to cause harm nor how low the total fuels need to be prior to the onset of permanent brain damage; however, it is likely to differ based on availability of alternative fuels, rates of brain metabolism (increased in seizures), and degree of plasma oxygenation. In conditions such as hyperinsulinism, where there are no alternate fuels available and when patients may have seizures which both increase brain energy needs and decreases the availability of oxygen in the blood stream, all these factors combine and may cause profound brain damage (16).

 

It is clear that not all hypoglycemia is equal, and the responsible physician must assess the etiology of the hypoglycemia, the likelihood of the presence of alternate fuels, and the presence of good blood flow and oxygenation in order to understand the danger of any given episode of hypoglycemia. The importance of correctly identifying the etiology of the hypoglycemia thus affects not just the long-term approach to management but also the immediate approach to the infant or child. This also explains why efforts to define a single blood glucose value that predicts brain damage is impossible and why efforts to define neurological outcome relative to glucose levels alone are not valid.

 

DEFINITION OF HYPOGLYCEMIA

 

For the reasons stated above, a single measurement of glucose does not reflect the total energy availability to the brain. Therefore, when one tries to define hypoglycemia, one should consider why a definition is required. We suggest that the definition of hypoglycemia needs to include three critical aspects. 

 

  • Diagnostic hypoglycemia is the level of glucose required to make a diagnosis of the etiology of the hypoglycemia.
  • Therapeutic hypoglycemia is the level of glucose one should aim to stay above during treatment of hypoglycemic disorders.
  • The concentration of glucose at which hypoglycemia causes brain damage is unknown and practically there is no such value.

 

Diagnostic Hypoglycemia 

 

Evaluation of a blood and urine sample when the plasma glucose is less than 50 mg/dL is essential in determining the etiology of hypoglycemia. This allows a determination of the metabolic profile at the time of hypoglycemia. This sample is often referred to as a “critical sample” and typically includes both blood (plasma) and urine samples, which are analyzed for the analytes in Table 2.

 

Table 2. The Critical Sample: Blood and urine samples to be drawn at a time of hypoglycemia (plasma glucose <50mg/dL) to guide the diagnosis of the etiology of hypoglycemia with additional testing for certain conditions.

Plasma

     Glucose

     Insulin

     Cortisol

     Growth hormone

     Beta-hydroxybutyrate

     Free fatty acids

     Acylcarnitine profile

     Ammonia

     Liver function tests

Urine

     Urine organic acids

In suspected insulin administration

     C-peptide

     Specific insulin assays to measure biological insulin only

     Sulfonylurea screen

In suspected insulinoma

     Proinsulin

     C-peptide

     Sulfonylurea screen

In suspected fatty acid oxidation defects

     Free and total carnitine

 

The critical sample can be drawn either at a time of spontaneous hypoglycemia or at the end of a fasting study in which plasma glucose less than 50 mg/dL is deliberately induced. Once the plasma glucose is <50 mg/dL, the counter regulatory hormones should be elevated, and insulin suppressed. The metabolic systems of glycogenolysis, gluconeogenesis, fatty acid oxidation, and ketone utilization should be underway and with glucagon administration, there should be no glycemic response as the liver should have utilized all the glycogen stores. Interpretation of the critical sample is the key to making the correct diagnosis (Table 3).  Figure 2 is a simple diagnostic algorithm to aid the provider.

 

Table 3. The Differential Diagnosis of Hypoglycemia in Neonates, Infants, and Children Based on the Results of the Critical Sample.

Disorder

Plasma Fuels when glucose <2.8 mmol/L

Plasma Hormones at End of Fast

Clinical Features

Lactate

mmol/L

FFA

mmol/L

BOHB

mmol/L

Insulin (µu/mL)

Cortisol (µg/dL)

GH (ng/mL)

ΔPG following glucagon (mg/dL)

Normal infants

0.7-1.5

>1.7

2-4

<2

>18

>10

<30

Normal

Hyperinsulinism

N

<1.7

<1.8

>1

N

N

>30

LGA

Cortisol deficiency

N

N

N

N

<14*

N

N

Hyperpigmentation if primary

GH deficiency

N

N

N

N

N

<8

N

Short stature

Panhypopituitarism

N

Low-N

Low-N

N

Low

Low

N

Short stature, midline facial malformation, optic hypoplasia, micro/small penis

Epinephrine deficiency (beta-blocker)

N

<1.5

<2

N

N

N

N

 

Debrancher deficiency (GSD III)

N

N

N

N

N

N

N

Hepatomegaly 4+

Phosphorylase deficiency (GSD VI)

N

N

N

N

N

N

N

Hepatomegaly 2+

Phosphorylase kinase deficiency (GSD IX)

N

N

N

N

N

N

N

Hepatomegaly 2+

Glycogen synthase deficiency (GSD 0)

N

N

N

N

N

N

N

 

Glucose 6-phosphatase deficiency (GSD la and lb)

4-8 +

N

<2

N

N

N

Normal glucose elevated lactate

Hepatomegaly 4+

Fructose 1, 6-diphosphatase deficiency

4-8 +

N

N

N

N

N

N

Hepatomegaly 1+

Pyruvate carboxylase deficiency

4-8 +

N

N

N

N

N

N

 

*Depending on sensitivity of the assay used (17). Note that findings of a single low growth hormone or cortisol level at the time of hypoglycemia has poor specificity for deficiency of either hormone (18,19). Stimulation testing may be needed to confirm the diagnosis. FFA, free fatty acids; GH, growth hormone; GSD, glycogen storage disease; LGA, large for gestational age; N, normal; PG, plasma glucose. Adapted from De Leon DD, Thornton P, Stanley CA, Sperling MA. Hypoglycemia in the Newborn and Infant. In: Sperling MA, Majzoub JA, Menon RK, Stratakis CA, eds. Sperling Pediatric Endocrinology. Fifth ed: Elsevier; 2021 (20).

 

Figure 2. Hypoglycemia diagnosis based on plasma metabolic fuel responses. Measurement of lactate as a gluconeogenic substrate, FFA from adipose tissue lipolysis, and BOHB at the time of hypoglycemia segregates major groups of hypoglycemia disorders. FFA, free fatty acids; GH, growth hormone; HCO3, bicarbonate; BOHB, beta-hydroxybutyrate. Adapted from Thornton PS, Stanley CA, De Leon DD, Harris D, Haymond MW, Hussain K, Levitsky LL, Murad MH, Rozance PJ, Simmons RA, Sperling MA, Weinstein DA, White NH, Wolfsdorf JI, Pediatric Endocrine S. Recommendations from the Pediatric Endocrine Society for Evaluation and Management of Persistent Hypoglycemia in Neonates, Infants, and Children. J Pediatr. 2015;167(2):238-245 (21).

 

Therapeutic Hypoglycemia

 

Generally, the target to treat infants and children with known hypoglycemic disorders is to keep the blood glucose above 70 mg/dL. There are certain conditions when the target maybe higher such as patients in acute decompensation with fatty acid oxidation defects where the goal is to stimulate insulin secretion which will prevent lipolysis and then fatty acid oxidation. In these circumstances it may require that the glucose levels are elevated to greater than 85 mg/dL.  Thus, in this case, getting the blood sugar up to 70 mg/dL will prevent the consequences of hypoglycemia but will not rapidly reverse the breakdown of lipids to free fatty acids and glycerol and thus will not stop the accumulation the abnormal metabolites of fatty acid oxidation. In children with GSD 1 the provision of adequate glucose to get the blood sugar greater than 70 mg/dL will provide the brain with adequate glucose for energy, however raising the blood sugar too high will drive glucose to lactate and thus worsen lactate levels. So, for individuals with GSD 1 the goal is to maintain the glucose at a level that will maximally suppress lactate levels (70-85 mg/dL) without triggering insulin release.

 

A second reason to maintain the plasma glucose greater than 70 mg/dL is to prevent hypoglycemic unawareness. Recurrent episodes of glucose levels less than 70 mg/dL over time will blunt the release of epinephrine by the autonomic nervous system. The secretion of epinephrine triggers awareness by the child and young adult of impending neuroglycopenia and allows them to react by food seeking behavior to prevent further hypoglycemia. After every episode of hypoglycemia, this counter regulatory response is blunted for up to 5 days and if the hypoglycemia recurs inside this period, the response is blunted even more. This failure to secrete epinephrine in response to hypoglycemia is hypoglycemia associated autonomic failure (HAAF) (22).

 

NEONATAL TRANSITIONAL GLUCOSE REGULATION

 

The transition from intrauterine to extra uterine life is a critical time for the neonate. In utero, the fetus is exposed to seemingly limitless supplies of glucose, amino acids, and other fuels necessary for growth and development, through the maternal placenta unit. Because of the insulin resistance of pregnancy, maternal plasma glucose levels are higher than in the non-pregnant state. Glucose is transported across the placenta by facilitated diffusion and fetal plasma glucose levels are approximately 8-15 mg/dL lower than maternal levels. Fetal plasma glucose levels are determined by maternal plasma glucose levels, however fetal insulin secretion is regulated by fetal plasma glucose levels. Recent data have shown that the threshold for insulin secretion in the fetus is lower than the adult and that this is controlled at the level of the KATP channel by decreased trafficking of the channel to the beta cell plasma membrane (23). The function of insulin in the fetus is to act as the main anabolic hormone and drive fetal growth. At the time of birth and clamping of the umbilical cord this constant supply of glucose is interrupted. The now newborn has to suddenly adjust initially to no glucose and soon thereafter to limited glucose input via, intermittent feeding and a nutrition source that is primarily fat (colostrum). The recent glucose in well babies study (GLOW) has shown that this transition can last up to 72-96 hours in normal breast-fed babies and consists of two phases (24): initially a hypoketotic hypoglycemic phase, and then a ketotic euglycemic/hypoglycemic stage until the mother’s milk comes in (Figure 3).

 

Figure 3. Glucose and beta-hydroxybutyrate levels in healthy term newborns over the first 96 hours of life demonstrating the period of transitional hyperinsulinism during adaptation to extrauterine life followed by the period of hyperketotic euglycemic/hypoglycemic phase of starvation until breast milk has come in and the neonate has adequate caloric intake. Stanley CA, et al. Figure 1. From Stanley CA, Thornton PS, De Leon DD. New approaches to screening and management of neonatal hypoglycemia based on improved understanding of the molecular mechanism of hypoglycemia. Front Pediatr. 2023;11:1071206 (25) CC BY 4.0. Adapted from Harris DL, Weston PJ, Harding JE. Alternative Cerebral Fuels in the First Five Days in Healthy Term Infants: The Glucose in Well Babies (GLOW) Study. J Pediatr. 2021;231:81-86 e82 (26).

 

In healthy newborns with normal birth weight, the glycogen stores from the liver built up during pregnancy, provide the initial glucose support. In the healthy newborn, liver glycogen concentration is the highest of all times in life and can supply the newborn with glucose for up to 12 hours. In preterm infants and infants born small for gestational age (SGA) or with intra-uterine growth retardation (IUGR), this supply is dramatically diminished thus increasing the risk of hypoglycemia in these babies. In addition, in the immediate newborn period, lactate levels are elevated (24), and gluconeogenesis from lactate rapidly increases over the first few hours of life, as the rate limiting enzyme of gluconeogenesis, phosphoenolpyruvate carboxy-kinase (PEPCK) increases to adult level by 24 hours of life. In the immediate post-natal hours, ketone levels are inappropriately low for the plasma glucose due to the lower threshold for insulin secretion but over the first 48 hours as the threshold for glucose stimulated insulin release rises, plasma ketone concentration begins to increase. In addition, medium chain fatty acids from colostrum are directly absorbed and transported to the liver through the portal circulation and undergo beta oxidation to generate ketones. In starved breastfed infants, ketones can reach 2 mmol/L by 48-72 hours of life (26).

 

The newborn rapidly adjusts from living in a steady anabolic state to an intermittent catabolic state until nutrition becomes adequate to supply sufficient energy to grow. In the relatively hypoxemic environment of in-utero life, insulin has a lower threshold for secretion to ensure the fetus remains in an anabolic state. After birth the newborn must adjust rapidly from the steady secretion of insulin associated with consistent fetal glucose availability to rapidly changing plasma glucose levels with feeding and to be able to deal with intermittent fasting. Over the first 12-24 hours of life the threshold for insulin secretion rises to the more typical 80-85 mg/dL seen in older infants, children and adults. All of this occurs while awaiting maturation of all the enzymes needed for gluconeogenesis and fatty acid oxidation. The consequences of these changes are that in the first 1-2 hours of life the blood glucose concentration falls to a mean of approximately 55 mg/dL (Figure 3). Gradually over the next 12-48 hours the plasma glucose concentration starts to rise to levels (60 mg/dL) approaching normal adult levels and by 72-84 hours of life, transition is complete and plasma glucose levels are in essence similar to adults (70 to 110 mg/dL) (2,24). During the first 24-48 hours of life upwards of 35-45% of normal healthy term babies may have occasional plasma glucose levels less than 50 mg/dL, however by 72-84 hours of life persistent plasma glucose levels less than 60 mg/dL reflect an underlying pathological problem (21).

 

Approach to Neonatal Glucose Screening and Treatment

 

The dilemma for physicians caring for newborns is how to differentiate babies having normal transitional glucose regulation with transient hypoglycemia from those with pathological hypoglycemia due to conditions such as perinatal stress-induced hyperinsulinism (PSHI).  The 2015 Pediatric Endocrine Society (PES) recommendations (21) guide the physician on how to differentiate physiological plasma glucose levels less than 50 mg/dL from pathological causes of hypoglycemia in the first days of life. They recommend that normal healthy term babies with no symptoms of hypoglycemia do not require glucose monitoring. Babies at risk for hypoglycemia such as those with IUGR, those born large for gestational age (LGA) or SGA, those born late pre-term, and those in whom maternal factors increase the risk of hypoglycemia such as maternal hypertension, preeclampsia, and eclampsia should be monitored.  In addition, babies who have symptoms consistent with hypoglycemia (Table 4) should also be screened for hypoglycemia. 

 

Table 4. Symptoms and Signs of Hypoglycemia

Neurogenic symptoms appear when glucose <70 mg/dL (<2.8mmol/L)

     Jitteriness (neonates), shakiness

     Tachycardia

     Pallor

     Hypothermia

     Hunger

     Sweating

     Weakness

Neuroglycopenic symptoms appear when glucose <45-50 mg/dL (2.5-2.8 mmol/L)

     Poor feeding (neonates)

     Apnea (neonates)

     Floppiness (neonates)

     Weak/high-pitched cry (neonates)

     Lip smacking and eye twitching (neonates)

     Headache

     Confusion

     Irritability, outbursts of temper

     Bizarre neurological signs

     Motor and sensory disturbances

     Lethargy

     Decreased muscle tone

     Unconsciousness

     Seizure

           

The PES guidelines recommend that the target to treat these infants be 50 mg/dL but if intravenous glucose is required indicating a more serious problem, the target should be 60-70 mg/dL. After 48 hours of life the target for treatment is 60 mg/dL. The recommendations also state that if a known hypoglycemic disorder is identified then the target for treatment is to maintain the blood glucose greater than 70 mg/dL. These goals should be achieved initially by appropriate resuscitation in the newborn period with the avoidance of cold stress to the baby, early skin to skin contact, early breast feeding or if chosen by mothers, bottle feeding (27).  Supplemental glucose can be given with dextrose gel (28). In neonates with symptomatic hypoglycemia or those in whom the above measures fail to maintain the plasma glucose above 50 mg/dL intravenous glucose therapy should be initiated at a glucose infusion rate (GIR) of 4-6 mg/kg/min with or without a 200 mg/kg bolus of IV dextrose. As shown. in Figure 4, within 10 minutes following an IV dextrose bolus of 200 mg/kg followed by a GIR of 8 mg/kg/min, plasma glucose levels reach close to 90% of the glucose concentration measured at 60 minutes. Thus, in patients with dangerously low glucose levels (<30 mg/dL) plasma glucose levels should be rechecked in 10-15 minutes to demonstrate correction of the hypoglycemia and if the glucose has not achieved the target level, then a repeat bolus and increase in the IV infusion rate of glucose should be implemented. In this manner, severe hypoglycemia can be rapidly corrected without significant overcorrection. At all times during treatment of hypoglycemia, the newborn infant should be encouraged to continue to feed orally, unless the patient is seizing or has significant respiratory distress in which case feeding should be withheld temporarily. 

 

Figure 4. The effect of a 200 mg/kg IV mini-bolus in addition to starting a glucose infusion rate of 8 mg/kg/minute compared to just starting 8 mg/kg/minute infusion rate alone. Reprinted from J. Pediatr, 97(2), Lilien LD, Pildes RS, Srinivasan G, Voora S, Yeh TF., Treatment of neonatal hypoglycemia with minibolus and intravenous glucose infusion, 295-298, 1980, with permission from Elsevier (29).

 

The single most important clinical differentiators of pathological hypoglycemia from transitional hypoglycemia in the newborn period are the presence of neuroglycopenic signs of hypoglycemia (apnea, lethargy, seizures) and the need for intravenous glucose treatment to correct low glucose levels. In both these circumstances the newborn should undergo a fasting study of 6-9 hours to demonstrate that normal glucose regulation has returned (ability to keep glucose >60-70 mg/dL for 6-9 hours) prior to discharge from the hospital. In babies who have hypoglycemia within 6-9 hours of commencing fasting an underlying pathological cause should be sought prior to discharge and appropriate therapy implemented. For babies with persistent glucose <60 mg/dL after 48 hours of life the use of point of care ketone screening may assist in detecting those babies with failed breast feeding from those with the more serious hypoketotic forms of hypoglycemia such as hyperinsulinism, hypopituitarism, or the fatty acid oxidation defects. In the case of persistent hypoglycemia with hypoketosis, further investigations need to be done to determine the etiology.

 

CLINICAL SYMPTOMS AND SIGNS OF HYPOGLYCEMIA

 

Classical symptoms and signs of hypoglycemia are outlined in Table 4. It is important to note that children <5 years of age will generally have hypoglycemic unawareness and not be able to recognize the neurogenic symptoms of hypoglycemia. Neonatal hypoglycemia, by virtue of neonates being unable to communicate in a complex way, may be difficult to detect as many of the signs of hypoglycemia occur in normal neonates (30). Subtle signs such as poor feeding, sleepiness, and jitteriness occur in many non-hypoglycemic babies and clinical judgement as to when to check glucose is required. However, if in doubt it is better to check blood glucose (21). For more serious symptoms, such as lethargy, seizures, apnea and coma, screening point of care glucose should be performed and hypoglycemia simultaneously confirmed by laboratory measured plasma glucose. Older children with acquired hypoglycemia such as that caused by insulinoma, often have hypoglycemic unawareness due to the frequency of hypoglycemia and only demonstrate neuroglycopenic signs. All neonates, infants, and children with neuroglycopenic symptoms or signs must have a plasma glucose concentration checked and if <60-70 mg/dL serious consideration of hypoglycemia as a cause should be entertained.

 

HORMONAL CAUSES OF HYPOGLYCEMIA

 

Hyperinsulinism

 

Hyperinsulinism refers to the group of hypoglycemic disorders caused by dysregulated, excessive insulin secretion or action via signaling. Insulin secretion from pancreatic beta-cells is tightly regulated and predominantly controlled by the plasma glucose concentration (Figure 5). In normal circumstances glucose enters the beta-cell via insulin-independent glucose transporters and is phosphorylated by glucokinase to G-6-phos. Glucokinase acts as the “glucose sensor” setting the threshold for insulin secretion; normally, insulin secretion is triggered when plasma glucose levels are above 85 mg/dL. Metabolism of G-6-phos leads to an increase in the intracellular adenosine triphosphate (ATP) to adenosine diphosphate (ADP) ratio which regulates subsequent closure of the ATP-sensitive plasma membrane KATP channels, membrane depolarization, activation of voltage-gated calcium channels, calcium influx, and release of insulin from stored granules. Amino acid metabolism also influences insulin secretion. Leucine allosterically activates glutamate dehydrogenase, increasing oxidation of glutamate to alpha-ketoglutarate, thereby increasing the ATP-to-ADP ratio, triggering the insulin secretion cascade. Glutamine mediates glucagon-like peptide 1 (GLP-1) receptor signaling, which acts as an “amplification pathway” for insulin secretion.

 

Figure 5. Diagram of the pathways stimulating beta-cell insulin secretion. Glucose enters the beta-cell via glucose transporters and is phosphorylated by GCK to G-6-phos. Oxidation of G-6-phos in mitochondria increases the ATP-to-ADP ratio, leading to closure of plasma membrane ATP-sensitive KATP channels (comprised of SUR1 and Kir6.2 subunits), inhibition of K+ efflux, membrane depolarization, opening of voltage-dependent Ca++ channels, Ca++ influx, and release of insulin from storage granules. Amino acids stimulate insulin secretion through glutamine-mediated amplification of GLP-1 receptor signaling. Leucine stimulates insulin secretion by increasing the oxidation of glutamate via activation of GDH, thereby increasing the ATP-to-ADP ratio and triggering the insulin secretion cascade. HK1 and MCT1 are not normally present in the beta-cell. Diazoxide suppresses insulin secretion by activating the KATP channel to remain open. Somatostatin suppresses insulin secretion downstream of Ca++ signaling. Known sites of defects associated with congenital hyperinsulinism are indicated by bold and underlined font and include GCK (glucokinase), HK1 (hexokinase 1), PGM1 (phosphoglucomutase 1), MCT1 (monocarboxylate transporter 1), UCP2 (uncoupling protein 2), SCHAD (short-chain 3-hydroxyacyl-coenzyme A dehydrogenase), GDH (glutamate dehydrogenase), HNF1A (hepatocyte nuclear factor 1A), HNFA (hepatocyte nuclear factor 4A), SUR1 (sulfonylurea receptor 1), Kir6.2 (inwardly rectifying potassium channel 6.2). Other abbreviations: OAA, oxaloacetate; PEP, phosphoenolpyruvate; α-KG, α-ketoglutarate; GAD, glutamic acid decarboxylase; GABA, γ-aminobutyrate, GHB, γ-hydroxybutyrate; SSA, succinic semialdehyde; Ins, insulin. Reprinted from Stanley CA, Perspective on the Genetics and Diagnosis of Congenital Hyperinsulinism Disorders, J Clin Endocrinol Metab, 2016, 101(3):815-826 by permission of Oxford University Press and Endocrine Society (31).

 

In neonates, hyperinsulinism may occur as a transient issue due to intrauterine factors, in association with recognized clinical syndromes, or may reflect genetic mutations in the pathway of insulin secretion (Figure 5). In older children, acquired forms of hyperinsulinism, including insulinoma, autoimmune causes, and medications are more common. However, late presentation of monogenic forms of hyperinsulinism remain possible (Table 5).

 

Table 5. Classification of Hyperinsulinism Disorders in Infants and Children.

Acquired neonatal HI

Risk Factors

Clinical features

 

Maternal diabetes, including gestational diabetes

Large for gestational age (LGA)

Cardiac Hypertrophy

 

Perinatal stress-induced HI

Small for gestational age (SGA)

Maternal hypertension, pre-eclampsia, eclampsia

 

Maternal drugs

Ritodrine, sulfonylurea, high GIR during labor etc.

Acquired non-neonatal HI

Categories

Clinical features

 

Neoplastic HI

Insulinoma (sporadic or MEN1)

 

Surgically induced HI

Post-gastric bypass, post-fundoplication for gastro-esophageal reflux (NIPHS: non-insulinoma pancreatogenous hypoglycemia syndrome)

 

Drug-induced HI

Antidiabetic medications (Insulin, sulfonylureas)

 

Autoimmune HI (anti-insulin or insulin receptor-activating antibodies)

Spontaneous or associated with drugs or viral infections

Hirata's disease (Insulin Autoimmune Syndrome: anti-insulin antibodies post sulfhydryl medications: methimazole, carbimazole, alpha-lipoic acid and post

measles virus, mumps virus, rubella virus, varicella zoster virus, coxsackie B virus and hepatitis C virus)

Genetic HI:  Isolated HI

Histology

Genes

 

Diffuse form

ABCC8, KCNJ11, GLUD1, GCK, HK1, HNF4A, HNF1A, HADH, SLC16A1, UCP2

 

Focal form

Paternally inherited AR variants of ABCC8 or KCNJ11

 

LINE- HI (Mosaic HI, Atypical HI)

Sporadic mosaic AD variants of ABCC8, GCK, and inappropriate expression of HK1

Genetic HI: Syndromic HI, select forms

Syndrome

Gene

 

Beckwith-Wiedemann syndrome

Genetic or epigenetic changes of imprinted region 11p15.5 (especially paternal UPD11p), mutations of imprinting control genes

Paternal UPD11p combined with paternal recessive ABCC8 or KCNJ11 mutation

 

Kabuki syndrome

KMT2D, KDM6A (usually mosaic)

 

Turner syndrome

Mosaic partial or complete X chromosome monosomy

HI mimickers: Hypoinsulinemic hypoketotic hypoglycemia

 

 

Autoimmune mimicker

Insulin resistance syndrome type B (anti-insulin receptor antibodies post viral infection (HIV, HTLV1, hepatitis C) or lymphoproliferative disease, or autoimmune disease)

 

Paraneoplastic secretion of pro-IGF2

Non-islets cells tumor hypoglycemia (NICTH, Doege-Potter syndrome)

 

Genetic disorders of insulin signaling

Mutations in AKT2, AKT3, PIK3CA, PIK3R2, CCND2, INSR

 

Fatty acid oxidation disorders

Abnormalities in the carnitine cycle, beta-oxidation, electron transfer and ketone synthesis. 

 

Adapted from De Leon DD, Arnoux JB, Banerjee I, Bergada I, Bhatti T, Conwell LS, Fu JF, Flanagan SE, Gillis D, Meissner T, Mohnike K, Pasquini TLS, Shah P, Stanley CA, Vella A, Yorifuji T, Thornton PS. International Guidelines for the Diagnosis and Management of Hyperinsulinism. Horm Res Paediatr. 2023 (32). 

 

Clinically, hyperinsulinism should be suspected when a higher than typical glucose infusion rate (GIR >8 mg/kg/minute) is required to maintain plasma glucose >70 mg/dL. Neonates with hyperinsulinism are often (but not always) born large for gestational age because insulin promotes fetal growth in utero.

 

Diagnosis is established by biochemical findings of inappropriate insulin action at the time of hypoglycemia (Table 6). These include inappropriately suppressed beta-hydroxybutyrate and free fatty acids, and a glycemic response to 1 mg intramuscular or intravenous glucagon administration (rise in plasma glucose >30 mg/dL within 40 minutes following glucagon administration) (33). Importantly, the diagnosis does not rest entirely on the presence or absence of detectable insulin and c-peptide levels at the time of hypoglycemia. Elevated insulin levels may not be observed in cases of HI if the laboratory specimen is hemolyzed (33), or when the plasma insulin concentration is below the detection threshold of the insulin assay utilized. Conversely, with improvements in assay sensitivity, plasma insulin may be reported as detectable in the absence of HI. Thus, accurate diagnosis of HI requires a comprehensive interpretation of biochemical markers of insulin action and does not rely solely upon an insulin level.

 

Table 6. Diagnostic Features of HI at the Time of Hypoglycemia (plasma glucose <50 mg/dL [2.8 mmol/L]).

Evidence of excessive insulin action at the time of hypoglycemia  

Suppressed plasma β-hydroxybutyrate (< 1.8 mmol/L) 

Suppressed plasma free fatty acids (< 1.7 mmol/L) 

Inappropriately large glycemic response to glucagon (≥ 30 mg/dL [≥1.7 mmol/L])

Increased glucose infusion rate required to maintain euglycemia (above normal for age):

>8 mg/kg/min for neonates

>3 mg/kg/min for adults

Evidence of excessive insulin secretion/inadequate suppression of insulin secretion at the time of hypoglycemia (these are less definitive than evidence of excessive insulin action)

Plasma Insulin >1.25 μU/mL (8.7 pmol/L)* 

C-peptide >0.5 ng/mL (> 0.17 nmol/L)*

*Note that these thresholds depend upon the assay utilized. Adapted from De Leon DD, Arnoux JB, Banerjee I, Bergada I, Bhatti T, Conwell LS, Fu JF, Flanagan SE, Gillis D, Meissner T, Mohnike K, Pasquini TLS, Shah P, Stanley CA, Vella A, Yorifuji T, Thornton PS. International Guidelines for the Diagnosis and Management of Hyperinsulinism. Horm Res Paediatr. 2023 (32).

 

Regardless of the etiology, prompt identification and treatment of hyperinsulinism is critical. Since insulin inhibits gluconeogenesis and ketogenesis, ketones and lactate are not sufficiently available as alternative fuels for the brain during hyperinsulinemic hypoglycemia. Consequently, the risk of brain injury is high. Initial management is thus to rapidly correct hypoglycemia via administration of IV dextrose bolus (200 mg/kg or 2 ml/kg D10%). Following this, continuous IV dextrose infusion should be started at a GIR of 4-8 mg/kg/min and quickly titrated (by 4 mg/kg/min if hypoglycemia persists on a GIR of 8 mg/kg/min) as needed to maintain plasma glucose >70 mg/dL. Some infants with hyperinsulinism may require GIRs as high as 20-30 mg/kg/min to maintain euglycemia. Higher concentrations of dextrose (D20%-D50%) may be utilized via a central line to minimize fluid overload in these settings. Glucagon can be administered intramuscularly if IV access is lost, or unable to be obtained, as a temporizing measure to raise plasma glucose. Glucagon can also be administered as a continuous IV infusion (doses of 2-3 mcg/kg/day up to 10 mcg/kg/day, or alternatively, infusion of 1 mg/day) to permit lowering of GIR (and thus fluid load) in cases where fluid overload is a concern.  In general, use of glucagon may be associated with up to 50% reduction in the GIR (34).

 

MONOGENIC FORMS OF HYPERINSULINISM

 

The incidence of congenital hyperinsulinism caused by genetic defects in the insulin secretion pathway is estimated at 1 in 40,000 live births (35). A higher incidence, of up to 1 in 2,500 births, has been described in Saudi Arabia, in a region of Finland, and in some of the Ashkenazi-Jewish population. Congenital hyperinsulinism can be classified by genetic etiology, histology, and response to treatment with diazoxide. At least 11 different monogenic causes have been identified. A high likelihood that yet undiscovered genetic etiologies exist is suggested by the low rate (approximately 50%) of mutation detection in patients with diazoxide-responsive forms of hyperinsulinism (36).

 

First-line treatment for congenital hyperinsulinism is diazoxide, a KATP channel opener that inhibits insulin secretion. Diazoxide therapeutic dose range is 5-15 mg/kg/day. Response to diazoxide should be assessed after 5 days of treatment with a carefully monitored fast (safety fast, Table 7). Responsiveness to diazoxide is both of clinical and diagnostic value (Figure 6). It is defined by maintenance of plasma glucose >70 mg/dL over the fasting period (Table 7) or a rise in beta-hydroxybutyrate >2 mmol/L prior to decline in plasma glucose below 50 mg/dL (37). Side effects of diazoxide include hypertrichosis (prevalence 30%), fluid overload — which may be complicated by pulmonary hypertension (prevalence 2-3%) — neutropenia and thrombocytopenia (prevalence 15%), and hyperuricemia (prevalence 5%) (38,39). To mitigate risks of fluid overload, empiric co-administration of a diuretic is recommended (40). Recommended surveillance on diazoxide includes echocardiogram, complete blood count with differential, electrolytes, and uric acid level at baseline and 5-7 days after diazoxide initiation. Following this, it is recommended to measure complete blood count with differential, electrolytes, and uric acid levels every 6 months (38-40).

 

Table 7. Safety/Cure Fasting Test Procedure

Have blood drawing IV line in place 

Check glucose (POC meter) and beta-hydroxybutyrate every 2-3 hours until glucose <70 mg/dL; then every 2 hours until <60 mg/dL; then hourly until < 50mg/dL

When glucose <60mg/dL by POC meter, send specimen for laboratory confirmation of plasma glucose

Terminate fast when: 

Plasma BOHB >2 mmol/L on two separate samples 1 hour apart, or

Plasma glucose <50 mg/dL, or  

Duration of fasting

     Safety fast: >9-12 hours in <1 month old, or >12 hours in 1 month-1 year old, or >18

hours in >1 year old

     Cure fast: >18 hours in <1 year old, or >36 hours in 1-10 years old, or 72 hours in >10

years old 

Adapted from De Leon DD, Arnoux JB, Banerjee I, Bergada I, Bhatti T, Conwell LS, Fu JF, Flanagan SE, Gillis D, Meissner T, Mohnike K, Pasquini TLS, Shah P, Stanley CA, Vella A, Yorifuji T, Thornton PS. International Guidelines for the Diagnosis and Management of Hyperinsulinism. Horm Res Paediatr. 2023 (32).

 

Notably, most children with hyperinsulinism due to mutations in genes encoding the KATP channel will not respond to diazoxide. Histologically, there are several forms of KATP channel hyperinsulinism including focal, diffuse and atypical. Many of these children will have a focal form of hyperinsulinism that can be cured by surgery (see below). In children with diazoxide-unresponsive hyperinsulinism, genetic testing that includes sequencing of KATP channel genes should be sent on DNA from the affected child and their parents simultaneously. This approach permits timely identification of children likely to have focal hyperinsulinism, and who can be cured by surgery. Sending the parental DNA with the child saves 1-2 weeks of time and many of the laboratories perform the testing free of charge if a mutation is found in the child in which diagnosing the parent of origin will change the management. In addition, rapid genetic testing with a turnaround time of 4-7 days is critical in the management of diazoxide-unresponsive hyperinsulinism because this quickly allows the physician to determine who should be referred to a multidisciplinary hyperinsulinism center for imaging with 18F-DOPA PET scan which can currently be used only under an investigational new drug license (IND) and is only available in several places in the country. Obtaining results of the patient and both parents within 7 days compared to 28-40 days if sent to a conventional genetic laboratory could save upwards of $100,000 to $200,000 and 21 days of hospital stay during which time the patient is at risk of severe hypoglycemia, development of feeding intolerance, line infections, and other iatrogenic complications of prolonged hospitalization.

 

Figure 6. HI Diagnostic and Treatment Algorithm. Once HI is diagnosed, effectiveness of diazoxide treatment needs to be assessed. Responsiveness to diazoxide is shown by demonstrating the ability to fast an age-appropriate interval (minimum 9 hours for neonate) with plasma glucose >70 mg/dL and/or generate beta-hydroxybutyrate >2 mmol/L prior to plasma glucose <50 mg/dL. For patients unresponsive to diazoxide, expedited genetic testing is obtained to differentiate diffuse and focal forms of HI. 18F-DOPA PET/CT is performed when genetic testing is suggestive of possible focal disease. For patients with diffuse, diazoxide-unresponsive disease, intensive medical therapy is initiated, with near-total pancreatectomy reserved for medically unresponsive cases. 18F-DOPA PET/CT 18-fluoro-L-3,4-dihydroxyphenylalanine positron emission tomography.

 

Second-line medical management options include somatostatin analogues – octreotide and the long-acting analog lanreotide - and enteral dextrose. Enteral dextrose is administered as a continuous infusion of 20% dextrose solution via nasogastric or gastrostomy tube (maximum enteral GIR 10 mg/kg/min). Octreotide is a short-acting somatostatin analogue administered multiple times per day subcutaneously (dose range: 2-20 mcg/kg/day, divided every 6-8 hours). Octreotide should not be used in children <2 months of age due to an association with fatal necrotizing enterocolitis. Lanreotide is used in children >1 year of age (administered as 60 mg injection monthly) (41). Several new therapies, including glucagon analogues, oral somatostatin analogues, insulin receptor modulators, and GLP-1 receptor antagonists, are under development (42).

 

In diffuse congenital hyperinsulinism cases that do not respond to medical treatment, near-total pancreatectomy is performed. Near-total pancreatectomy is palliative, not curative; significant hypoglycemia persists in up to 50% of children. Additionally, this procedure is complicated by development of exocrine pancreatic insufficiency and late-onset post-pancreatectomy diabetes.

 

KATP Hyperinsulinism (KATP HI)

 

The most common and severe form of congenital hyperinsulinism, KATP HI, is caused by inactivating mutations in ABCC8 or KCNJ11. Both located on chromosome 11p15.1, these genes each encode a subunit of the pancreatic beta-cell KATP channel. Inactivation of the KATP channel results in beta-cell depolarization (Figure 5), and inappropriate secretion of insulin. In addition to severe fasting hypoglycemia, KATP HI is characterized by protein-induced hypoglycemia mediated by GLP-1 receptor signaling (6).

 

KATP HI is histologically classified as diffuse, in which all beta-cells are affected, focal, in which a localized subset beta-cells are affected, or less commonly, atypical (also termed Localized Islet Nuclear Enlargement [LINE-HI]). Focal KATP HI results from a “two hit” mechanism involving paternal transmission of a recessive KATP HI mutation and a somatic loss of heterozygosity for the maternal 11p15 region yielding imbalanced expression of imprinted tumor suppressor genes (43). Dominant or recessive mutations in KATP HI genes cause diffuse KATP HI. Often, children with atypical histology (LINE-HI) do not have an identifiable mutation in standard sequencing of peripheral blood; however, low-level mosaic mutations are increasingly recognized as causative (44).

 

Infants with diffuse forms of KATP HI are often born large for gestational age and present with symptomatic hypoglycemia in the first few days of life. While infants with the focal form of KATP HI are more likely to have normal birth weight and present at an older age than those with diffuse disease, the two histologic forms are often indistinguishable in clinical practice (45). The recessive, focal, and atypical (LINE-HI) forms of KATP HI are usually not responsive to diazoxide. In contrast, dominant KATP HI may be diazoxide responsive due to retained partial function of KATP channels.

 

For infants with diazoxide-unresponsive hyperinsulinism, the goal is to identify those children with focal KATP HI since these children can be cured by surgery. Findings of a single recessive KATP mutation in the father, but not the mother, will have a 94% positive predictive value for focal hyperinsulinism (36) in the symptomatic infant. When suspected, 18-fluoro-L-3,4-dihydroxyphenylalanine positron emission tomography (18F-DOPA PET) is used to localize the focal lesion and guide surgical excision (46,47). Surgical cure rates for focal hyperinsulinism exceed 95%, when performed by experienced surgeons at a multidisciplinary hyperinsulinism center (48). 

 

For patients with diffuse KATP HI, intensive medical management is attempted, initially with continuous enteral dextrose (maximum enteral GIR 10 mg/kg/min). Octreotide can be added after 2 months of age (as above). Surgical management (near-total pancreatectomy with gastrostomy tube placement) is reserved for patients who fail to achieve adequate glycemic control with intensive medical therapy because this procedure is not curative and carries long-term risks of exocrine pancreatic insufficiency and insulin-dependent diabetes mellitus (45,49). Notably, regardless of initial treatment, the severity of hypoglycemia tends to improve with age in children with diffuse KATP HI. Determination of optimal management strategy thus depends on balancing short/intermediate-term risks of suboptimal hypoglycemia control and labor-intensive home management with potential further increased risk of long-term risk of adverse neurodevelopmental outcomes in the case of medical management versus inevitable post-pancreatectomy complications in the case of surgery. Management decisions are thus informed by the initial severity of hypoglycemia, responsiveness to intensive medical therapy, and the preferences and values of the child’s family. These children thus require specialized care and should be referred to a multidisciplinary hyperinsulinism center.

 

Glycemic status should be assessed in all children following pancreatectomy. Those children who do not require dextrose to maintain euglycemia postoperatively should undergo fasting study to demonstrate whether they are cured or need further medical management (Table 7) (32). Cure of HI can be demonstrated by the development of hyperketonemia (beta-hydroxybutyrate >1.8 mmol/L) prior to development of hypoglycemia (glucose <50 mg/dL). 

 

Later in life, dysregulated insulin secretion in diffuse KATP HI may additionally manifest as gestational, and in some cases insulin-dependent, diabetes mellitus even in the absence of prior pancreatectomy. This finding has been observed both in dominant and recessive forms of diffuse KATP HI (50,51). Mechanisms underlying the switch from hypoglycemia in early life to hyperglycemia later on remain incompletely understood. Impaired glucose-stimulated insulin secretion has been implicated based upon findings of reduced first-phase and maximal glucose-stimulated insulin secretion during oral glucose tolerance testing in adults with dominant KATP (51), and reduced acute insulin response to graded IV glucose infusion in children with recessive KATP HI (50). Impaired insulin secretory response to glucose (beta-cell “glucose blindness”) has also been proposed to underlie progression from hyperinsulinemic hypoglycemia to hyperglycemia in maturity-onset diabetes of the young (MODY) type 1 and type 3 (discussed below) (52-54). In mouse models of KATP HI, increased beta-cell apoptosis has been observed prior to the development of hyperglycemia, suggesting that a progressive decline in beta-cell mass may also play a role (55).

 

GCK Hyperinsulinism (GCK HI)

 

Dominant activating mutations in the GCK gene (chromosome 7p13), encoding glucokinase, cause hyperinsulinism by increasing the affinity of glucokinase for glucose, thereby lowering the threshold for pancreatic beta-cell insulin secretion (Figure 5). Clinical phenotype is highly variable, even within the same family. Presentation ranges from severe hypoglycemia at birth with large for gestational age birth weight, to milder hypoglycemia detected in adulthood. Response to diazoxide is also variable. In severe cases, near-total pancreatectomy may be required.

 

HK1 Hyperinsulinism (HK1 HI)

 

The HK1 gene (chromosome 10q22.1) encodes hexokinase, which has a much higher affinity for glucose than glucokinase. Normally, expression of HK1 is suppressed in pancreatic beta-cells postnatally. Dominantly inherited variations in non-coding regions of the HK1 gene result in aberrant beta-cell expression of hexokinase. As a consequence, appropriate suppression of insulin secretion at low plasma glucose levels is impaired, resulting in hypoglycemia (56). The clinical phenotype is variable. Most affected individuals present in the first weeks of life, but delayed presentation is not infrequently reported. Severity ranges from severe symptoms at birth to mild symptoms detected only following identification of an affected relative. Response to diazoxide is similarly heterogeneous and some affected individuals have required pancreatectomy  (56,57).

 

GDH Hyperinsulinism (GDH HI)

 

Dominant activating mutations in the GLUD1 gene (chromosome 10q23.2), encoding glutamate dehydrogenase (GDH), cause the hyperinsulinism hyperammonemia syndrome. Constitutively hyperactive GDH results in increased oxidation of glutamate to ammonia and alpha-ketoglutarate, the latter of which enters the citric acid cycle, thereby increasing the ATP-to-ADP ratio, triggering insulin secretion (58). Profound protein-induced hypoglycemia occurs because the amino acid leucine is potent allosteric activator of GDH (Figure 5).

 

Individuals with GDH HI typically have normal birth weight and later age of presentation (median age 4-5 months). In addition to fasting and protein-induced hypoglycemia, GDH HI is associated with an increased risk of epilepsy as well as higher rates of intellectual impairment, both of which appear to be independent of hypoglycemic neurological injury (59). Rates of learning and intellectual impairments have ranged from 37-77% in studies of children with GDH HI in which these outcomes were measured (59-61). The characteristic seizure type observed is generalized, atypical absence (62). These seizures occur in the setting of euglycemia and notably are distinct from the focal-onset seizures that may occur following hypoglycemic brain injury (62). Aberrant GDH activity in the central nervous system, and resultant altered glutamate balance, have been hypothesized to underlie these neurological differences. However, the mechanism by which these deficits occur has not yet been definitively established.

 

Persistent hyperammonemia (ammonia elevation 2-5 the normal range) due to GDH overactivity in the kidney is a cardinal feature but appears to be clinically asymptomatic (63). Importantly, affected individuals do not manifest symptoms of acute hyperammonemia encephalopathy (lethargy, headache, vomiting) as do children with urea cycle disorders. Plasma ammonia levels are not influenced by dietary protein load, nor are they lowered by typical therapies for hyperammonemia (sodium benzoate, N-carbamyl-glutamate) (64). The finding of hyperammonemia is thus useful in establishing a clinical diagnosis of GDH HI. However, once identified, plasma ammonia levels do not require serial monitoring or targeted intervention.

 

Hypoglycemia in GDH HI is usually well-managed with diazoxide and dietary modification. While dietary protein should not be restricted, it is imperative that affected individuals consume carbohydrate prior and concomitant to protein intake. Most individuals with GDH HI will respond to a 2:1 gram ratio of carbohydrates to protein to prevent protein induced hypoglycemia but some will need ratios of 3:1. Typically breast milk or formula milk will not trigger hypoglycemia, but once solid foods are introduced, especially meats, care must be taken to prevent protein induced hypoglycemia. The cardinal warning sign of this is patients in whom glucose control was adequate including fasting overnight but who suddenly develop post prandial hypoglycemia.

 

A formalized protein challenge test (Table 8) may be performed to evaluate adequacy of treatment in individuals with GDH HI. This test is also helpful to evaluate protein sensitivity in KATP HI, HADH HI (below), and congenital HI with negative genetic testing. A drop in the plasma glucose of more than 10 mg/dL or below 70 mg/dl is considered an abnormal result (evidence of protein sensitivity). Because this test must be done following a 3-4 hour fast to ensure the glucose levels are stable prior to starting the test it can only be done in patients whose fasting glucose is controlled. Patients who are very protein sensitive may drop the plasma glucose in the first 15-60 minutes.

 

Table 8.  Oral Protein Challenge Test Procedure

Make patient NPO for food and carbohydrate containing fluids for 3-4 hours pre procedure

Insert peripheral intravenous line and have dextrose 10% available for emergency use

Measure baseline glucose and insulin

Administer 1g/kg of food protein PO over 10-15 minutes. Alternatively, protein powder may be used (administered via nasogastric or gastrostomy tube).

Measure glucose and insulin every 30 minutes for 3 hours

If the plasma glucose drops to <60 mg/dL terminate test with carbohydrate drink of 15 g or intravenous push of 200 mg/kg (2ml/kg D10%)

 

HADH Hyperinsulinism (HADH HI)

 

Inactivating mutations in HADH (chromosome 4q25) cause a rare, autosomal recessive form of diazoxide-responsive hyperinsulinism. The HADH gene encodes short chain 3-hydroxyacyl-coenzyme A dehydrogenase (HADH, also referred to as SCHAD) which inhibits GDH and also plays a role in mitochondrial fatty acid beta-oxidation. Loss of normal GDH inhibition results in a similar phenotype as GDH HI with fasting and protein-induced hyperinsulinemic hypoglycemia, but without hyperammonemia. Elevated levels of 3-hydroxybutyryl-carnitine in plasma and 3-hydroxyglutaric acid in urine may serve as clues to the diagnosis in some but are not universally observed (65). Treatment is with diazoxide and dietary modification as in GDH HI. 

 

HNF1A and HNF4A Hyperinsulinism (HNF1A and HNF4A HI)

 

Dominant inactivating mutations in the genes encoding transcription factors hepatocyte nuclear factor 1 alpha and hepatocyte nuclear factor 4 alpha, cause both hyperinsulinism and maturity-onset diabetes of the young (MODY3 and MODY1, respectively). Affected infants are often born large for gestational age. Severity of hyperinsulinism varies from transient neonatal hypoglycemia to persistent hyperinsulinism requiring treatment into school age (66). The p.Arg63Trp HNF4A mutation is associated with an additional extra-pancreatic phenotype of renal Fanconi syndrome and hepatic dysfunction (66,67). Response to diazoxide is often robust. Establishing the diagnosis has important implications both for the affected child, who requires ongoing surveillance after hyperinsulinism resolution due to the risk of developing diabetes later in life, and for family members carrying the mutation who could benefit from early diagnosis of diabetes or targeted therapy (i.e., sulfonylureas).

 

MCT1 Hyperinsulinism (MCT1 HI)

 

Dominant mutations in the non-coding regions of SLC16A1 (chromosome 1p13.2), encoding monocarboxylate transporter 1 (MCT1), result in exercise-induced hyperinsulinism (68). MCT1

catalyzes transport of monocarboxylates, such as lactate, pyruvate and beta-hydroxybutyrate, across the plasma membrane. Normally, MCT1 expression is disallowed in pancreatic beta cells. Promoter-activating mutations induce inappropriate MCT1 expression in beta cells, permitting uptake of pyruvate during exercise when plasma pyruvate is elevated. Pyruvate metabolism leads to increased ATP production, and resultant pyruvate-stimulated (exercise-induced) insulin release despite hypoglycemia (69). Response to diazoxide is variable, and carbohydrate loading prior to exercise is recommended to control hypoglycemia. Nonfunctional variants in the coding regions of SLC16A1 cause ketotic hypoglycemia (see section on Pathologic Ketotic Hypoglycemia and Ketone Utilization and Transport Defects). This is a very uncommon clinical entity.

 

UCP2 Hyperinsulinism

 

Dominant inactivating mutations in the UCP2 gene (chromosome 11q13.4) encoding uncoupling protein 2 (UCP2) have been associated with a diazoxide-responsive form of hyperinsulinism (70). Following initial reports, the prevalence of UCP2 variants in the general population has been found to be high, raising the role of UCP2 variants as a monogenic cause of hyperinsulinism into question (71).

 

SYNDROMIC HYPERINSULINISM

 

Hyperinsulinism is a feature of several recognized syndromes. These include the overgrowth syndromes: Beckwith-Wiedemann, Sotos, Simpson-Golabi-Behmel, and Perlman syndromes, as well as Kabuki syndrome, Turner syndrome, Tyrosinemia type 1, Usher syndrome type 1C (in which there is contiguous gene deletion at 11p15.2 including ABCC8), Rubinstein-Taybi syndrome, and several congenital disorders of glycosylation, among others (72). Beckwith-Wiedemann, Kabuki, and Turner syndromes are most frequently observed (72), and are discussed in the sections below along with the congenital disorders of glycosylation associated with hyperinsulinism.

 

Beckwith-Wiedemann Syndrome

 

Beckwith-Wiedemann syndrome, caused by genetic or epigenetic changes on chromosome 11p15.5, is an overgrowth disorder with classical features of macrosomia, macroglossia, hemihypertrophy, abdominal wall defects, and embryonal tumors. Due to the varying molecular etiologies, and the postzygotic nature of the epigenetic changes in most cases, affected children can present with a variety of clinical features along a spectrum of “classic” to “atypical” to isolated lateralized overgrowth (73). Hyperinsulinism occurs in approximately 50% of all cases and is the presenting symptom of Beckwith-Wiedemann syndrome in 16% of cases (73). While the hyperinsulinism is typically mild in severity and resolves within the first days to years of life, in roughly 5% of cases (particularly cases due to paternal uniparental isodisomy for chromosome 11p), it can be severe and persistent, requiring pancreatectomy (74). Hyperinsulinism persisting beyond the first week of life is considered a cardinal feature of Beckwith-Wiedemann syndrome (75). Thus, all patients presenting with hyperinsulinism should be evaluated for subtle limb asymmetry and other suggestive features (Table 9) (75). Molecular testing for investigation of Beckwith-Wiedemann syndrome should be considered in these patients. Additional suggestive features in the context of a hyperinsulinism evaluation include marked pancreatic enlargement and diffuse 18-F-DOPA uptake, or alternatively, very large areas of focal 18-F-DOPA uptake, on PET/CT imaging (76). In the latter setting, areas of increased involvement on PET imaging may be used to tailor the extent of pancreatectomy. Histologically, resected pancreatic tissue in cases of Beckwith-Wiedemann syndrome are characterized by a dramatic increase in endocrine tissue relative to the amount of exocrine tissue, often with a loss of the normal lobular architecture, and prominent trabecular arrangement of endocrine cells (76,77). Since the genetic or epigenetic changes causing Beckwith-Wiedemann syndrome usually occur during embryonal development, yielding a mosaic pattern, failure to detect these changes in blood (leukocytes) is thus not conclusive, and additional testing (e.g., from skin or pancreas) may be required.

 

Table 9.  Clinical Features of Beckwith-Wiedemann Syndrome (BWS)

Cardinal features (2 points per feature)

Macroglossia

Exomphalos

Lateralized overgrowth

Multifocal and/or bilateral Wilms tumor or nephroblastomatosis

Hyperinsulinism (lasting >1 week and requiring escalated treatment)

Pathology findings: adrenal cortex cytomegaly, placental mesenchymal dysplasia or pancreatic adenomatosis

Suggestive features (1 point per feature)

Birthweight >2SDS above the mean

Facial nevus simplex

Polyhydramnios and/or placentomegaly

Ear creases and/or pits

Transient hypoglycemia (lasting <1 week)

Typical BWS tumors (neuroblastoma, rhabdomyosarcoma, unilateral Wilms tumor, hepatoblastoma, adrenocortical carcinoma or phaeochromocytoma)

Nephromegaly and/or hepatomegaly

Umbilical hernia and/or diastasis recti

Score interpretation

≥4: Clinical diagnosis of classical BWS. Genetic testing for investigation and diagnosis of BWS recommended. Note that clinical diagnosis does not require the molecular confirmation of an 11p15 anomaly.

≥2: Merit genetic testing for investigation and diagnosis of BWS

Patients with a score of ≥2 with negative genetic testing should be considered for an alternative diagnosis and/or referral to a BWS expert for further evaluation

Adapted from Brioude F. et al, Expert consensus document: Clinical and molecular diagnosis, screening and management of Beckwith-Wiedemann syndrome: an international consensus statement. Nat Rev Endocrinol. 2018;14(4):229-249 (75).

 

Kabuki Syndrome

 

Kabuki syndrome is caused by dominant mutations in KMT2D (~75% of cases) or X-linked mutations in KDM6A. While the true incidence of hyperinsulinism in Kabuki syndrome is unknown, it is increasingly recognized as a feature of this disorder, often as the presenting feature (78). Clinically, affected children have distinctive facial features – long palpebral fissures with eversion of the lateral lower eyelid, arched eyebrows, and prominent ears – skeletal anomalies, intellectual disability, and post-natal growth deficiency. Congenital heart defects, genitourinary and gastrointestinal anomalies, and immune dysfunction may also be observed. Haploinsufficiency of KDM6A has been proposed as the pathophysiologic mechanism of hyperinsulinism, and human islets treated with KDM6A inhibitor demonstrate abnormal insulin secretion (79). Most cases are diazoxide-responsive. Somatostatin analogues have been used with success in cases where diazoxide was contraindicated due to cardiac comorbidity.

 

Turner Syndrome

 

The incidence of hyperinsulinism in infants with Turner syndrome is roughly 50 times that expected in the general population (79). As in Kabuki syndrome, haploinsufficiency of KDM6A (located on X chromosome) has been proposed as the underlying mechanism. Many, but not all, children with Turner syndrome and associated hyperinsulinism are diazoxide-responsive.

 

FOXA2 Hyperinsulinism

 

Inactivating mutations in FOXA2, encoding the transcription factor forkhead box A2 (Foxa2), have been associated with a clinical phenotype of congenital hyperinsulinism, hypopituitarism, and endodermal-derived organ anomalies (80,81). Treatment with both pituitary hormone replacement and diazoxide has been reported to be effective.

 

Congenital Disorders of Glycosylation

 

Monogenic defects in the synthesis of oligosaccharides are responsible for congenital disorders of glycosylation (CDG), over 100 of which have been identified to date. Endocrine dysfunction (including growth failure, hypothyroidism, hypogonadotropic hypogonadism) is common to many congenital disorders of glycosylation because both endocrine peptides and their receptor targets are glycosylated. These disorders have a wide phenotypic spectrum, and three have been associated with hyperinsulinism: phosphomannomutase 2 deficiency (PMM2-CDG, formerly CDG-1a), mannose phosphate isomerase deficiency (MPI-CDG, formerly CDG-1b), and phosphoglucomutase 1 deficiency (PGM1-CDG, formerly CDG-1t, also formerly referred to as GSD XIV). PMM2-CDG is the most common congenital disorder of glycosylation. Hyperinsulinism in PMM2-CDG has been proposed to result from impaired function of beta-cell KATP channels and most cases have been diazoxide-responsive (82). MPI-CDG predominantly manifests with gastrointestinal and hepatic involvement (protein losing enteropathy, liver dysfunction and fibrosis), and hyperinsulinemic hypoglycemia. MPI-CDG is treated with mannose, however hyperinsulinism-specific therapies (e.g., diazoxide) may also be required to adequately manage hypoglycemia (83). PGM1 catalyzes the interconversion of G-1-phos and G-6-phos and is thus involved in glycogenesis, glycogenolysis, and gluconeogenesis. Both fasting ketotic hypoglycemia, due to the role of PGM1 in these metabolic pathways, and post-prandial hyperinsulinemic hypoglycemia, due to a lowered threshold for glucose-stimulated insulin secretion, are observed in PGM1-CDG. Thus, this condition mimics GSD 0 and should be considered in that differential diagnosis. Diagnosis can be established by biochemical or molecular testing. Biochemical methods include analysis of serum transferrin glycoforms (also termed carbohydrate-deficient transferrin analysis) by isoelectric focusing or by mass spectroscopy to determine the number and presence of incomplete sialylated N-linked oligosaccharide residues linked to serum transferrin (84). If biochemical testing is not suggestive of a particular CDG, molecular testing approaches include multigene panels or more comprehensive genomic (whole exome, whole genome) testing.

 

TRANSIENT AND PERINATAL STRESS-INDUCED HYPERINSULINISM

 

Transient hyperinsulinism occurs secondary to maternal factors, most commonly gestational diabetes mellitus. In uncontrolled gestational diabetes, hyperglycemia induces fetal hyperinsulinism resulting in macrosomia and hypoglycemia following delivery. Transient hyperinsulinism can also result from maternal use of medications affecting glucose homeostasis, including hypoglycemic agents (e.g., oral sulfonylureas), terbutaline, or propranolol. Hyperinsulinism due to these factors resolves within the first days of life. Resolution can be confirmed by performing a 6-9 hour fast and demonstrating the baby can maintain plasma glucose >60-70 mg/dL throughout. If hyperinsulinism persists beyond 5-7 days of life alternate causes should be sought, particularly perinatal stress-induced hyperinsulinism (see below). 

 

Perinatal factors are also associated with development of perinatal stress-induced hyperinsulinism (PSHI), which has a more prolonged course than transient hyperinsulinism.  Neonates with perinatal complications such as birth asphyxia, maternal preeclampsia, prematurity, intrauterine growth retardation, or other peripartum stress may develop PSHI. As previously noted, fetal hypoxia results in decreased trafficking of KATP channels to the beta-cell membrane, decreasing the threshold for insulin secretion (23,85). Hyperinsulinism spontaneously resolves within weeks to months as beta-cell insulin regulation normalizes. Median age of resolution is six months. By definition, PSHI resolves by one year of age. PSHI typically responds to treatment with diazoxide, typically at doses on the lower end of the therapeutic range (5-7.5 mg/kg/day). Rates of adverse effects of diazoxide may be higher in children with PSHI, and empiric initiation of diuretic and close monitoring are paramount (39). Given these factors, it is recommended to start with a diazoxide dose of 5 mg/kg/day, initially, and to increase the dose after 3-5 days of treatment if adequate response is not achieved. Timing of initiating diazoxide in cases of suspected PSHI should be tailored to the infant’s overall clinical course. For infants with ongoing intensive care nursery needs (e.g., intubation, warming bed, parenteral feeds), plasma glucose support with IV or enteral dextrose-containing fluids offers optimal initial management, especially as some infants will demonstrate resolution of hyperinsulinism before they are otherwise prepared for hospital discharge. In these cases, repeat fasting evaluation should be performed prior to discharge to assess for resolution versus need for initiation of targeted treatment. As above, in these at-risk infants in whom hypoglycemia is considered likely to resolve within a short time, resolution can be confirmed by performing a 6-9 hour fast and demonstrating the baby can maintain plasma glucose >60-70 mg/dL throughout. Diazoxide should be initiated for infants approaching discharge in whom hyperinsulinemic hypoglycemia has not yet resolved, and safety fast (Table 7) should be performed to confirm adequate diazoxide efficacy. While the diagnosis of PSHI may be suspected by the clinical history, it is established only when hyperinsulinism resolution is confirmed by a repeat fasting test after treatment has been discontinued, cure fast (Table 7).

 

ACTIVATING MUTATIONS IN THE INSULIN SIGNALING PATHWAY

 

Activating mutations in insulin signaling pathway genes, including AKT2, AKT3, and PIK3CA, cause hypoglycemia with biochemical findings of inappropriate insulin action, but with low or absent plasma insulin levels (86). Asymmetric somatic overgrowth may serve as a clinical clue to the diagnosis. Frequent feedings or continuous enteral dextrose are effective treatments. 

 

ACQUIRED FORMS OF HYPERINSULINISM

 

Insulinoma

 

Insulinomas are pancreatic neuroendocrine tumors. The incidence of pediatric insulinoma is unknown, however, these lesions are less common in children than in adults (1-3 cases per million per year) and are thus exceedingly rare. While most pediatric cases present in adolescence, presentation as young as 2 years of age has been described.

 

Insulinomas are typically benign, solitary lesions. They can occur sporadically, or in association with multiple endocrine neoplasia, type 1 (MEN1). The frequency of MEN1 mutations in children with insulinoma has been reported to range 26-42% (87,88), which is higher than that in adults (5-10% in adults) (89). Clinically, insulinomas typically manifest with recurrent episodes of fasting hypoglycemia associated with neuroglycopenic symptoms. Weight gain is commonly noted at presentation, and occurs due to increased carbohydrate intake to treat symptoms of hypoglycemia (88). Notably, however, hypoglycemia unawareness is common. This is because repeated and prolonged hypoglycemia episodes can both decrease the counter regulatory hormonal response to hypoglycemia and induce unawareness of the autonomic and neuroglycopenic symptoms of hypoglycemia (90). Consequently, delays in establishing the diagnosis and initial misdiagnosis with neurologic and psychiatric disorders are not uncommon (91).

 

Suppressed beta-hydroxybutyrate, free fatty acids, and IGF-BP1, with inappropriately elevated proinsulin, insulin, and c-peptide levels at the time of hypoglycemia are consistent with the diagnosis. However, these biochemical findings do not differentiate insulinoma from sulfonylurea ingestion or congenital hyperinsulinism. Consequently, surreptitious use of insulin secretagogues (discussed in the section Exogenous hypoglycemia below) must be excluded in all suspected cases of insulinoma. Once the diagnosis is made, localization of the insulinoma is critical to direct the definitive treatment, surgery. Various imaging modalities have been used, including endoscopic ultrasound, computerized tomography (CT), magnetic resonance imaging (MRI), single-photon emission CT (SPECT), positron emission tomography (PET) and intraoperative ultrasound, each with variable sensitivity. The addition of GLP-1 receptor agonists (exendin-4) has increased the sensitivity of nuclear imaging modalities for detecting insulinomas, and exendin-4 PET/CT appears to be more sensitive than exendin-4 SPECT/CT (92). Historically, arterial calcium stimulation with venous insulin sampling was used, however, these invasive procedures have become less common with improvements in the imaging modalities available. Insulinomas are typically small lesions measuring <1 cm, and multiple lesions may be present. Preoperative localization can be challenging, and use of multiple imaging modalities may be required.

 

Diazoxide may be effective treatment in patients awaiting surgery or for whom the lesion cannot be localized. Surgical excision is curative, and prognosis is generally excellent. Genetic testing for MEN1 should be conducted in all patients, and appropriate screening should be initiated if a mutation is found. Insulinomas may be recurrent, with higher risk of recurrence in those with MEN1. For more information on histopathology, risks of malignancy, and suggested follow up protocols see the chapter entitled “Insulinoma” in the Diffuse Hormonal Systems and Endocrine Tumor Syndromes section of Endotext (93).

 

Autoimmune Hypoglycemia

 

Autoimmune hypoglycemia may result from the development of antibodies to insulin, referred to as Hirata disease, or to the insulin receptor. Onset may be triggered by viral infection or medication in a susceptible individual, and association with specific HLA haplotypes has been reported. Biochemically, plasma beta-hydroxybutyrate and free fatty acids are inappropriately low at the time of hypoglycemia, and c-peptide is suppressed. When autoimmune hypoglycemia occurs due to antibodies to insulin, plasma insulin levels may be very high (>1000 pmol/L), due to interference of insulin antibodies with the assay. Detection of insulin antibodies can confirm the diagnosis in patients naïve to exogenous insulin, which must be excluded. Autoimmune hypoglycemia is a spontaneously remitting condition. Various immune modulating treatments, including glucocorticoids, plasmapheresis, intravenous immunoglobulin, and rituximab have been utilized. A comparison of the different therapeutic approaches has not been conducted, owing to the rarity of this condition and its self-resolving course.

 

Post-prandial Hypoglycemia (Late Dumping Syndrome)

 

Post-prandial hypoglycemia (late dumping syndrome) occurs due to disrupted gastric motility, most commonly as a consequence of gastrointestinal surgery. In children, fundoplication surgery is implicated most frequently, whereas in adults, bariatric surgery is the most common cause. Hypoglycemia usually develops 1-3 hours after a meal, and results from imbalance between glucose absorption and insulin secretion. Rapid gastric emptying and intestinal absorption of carbohydrate, result in early hyperglycemia and exaggerated GLP-1 secretion, both of which trigger an exaggerated insulin response (94). The diagnosis can be confirmed by serial monitoring of insulin and glucose following feeding or with formal mixed meal tolerance testing. Fasting studies may be required to fully distinguish fasting hypoglycemia from post-prandial hypoglycemia (which may be comorbid in some patients). When conducting fasting studies in children with risk factors for (gastrostomy tube placement, Nissen fundoplication, esophageal or ileal surgery), or suspected, post-prandial hypoglycemia, it is important to slowly taper off feeds to avoid confounding of the fasting tolerance assessment by “dumping.” Treatment is often dietary manipulation. Decreasing the volume or rate of feeding, tapering the rate of feeding prior to stopping, increasing dietary fat, and decreasing simple carbohydrates may all be helpful. In older children, acarbose, which acts to slow carbohydrate digestion, has been used successfully. The efficacy of these approaches should be confirmed by repeat serial monitoring of glucose following at least two feeds to ensure plasma glucose is maintained within target range. In some cases, continuous enteral feeds, or enteral dextrose may be required. Typical treatments for genetic forms of HI such as diazoxide or octreotide generally have been unsuccessful. However, some of the novel therapies for HI under development (42,95), are also being studied in post bariatric surgery hypoglycemia.

 

Exogenous

 

Exogenous, factitious, and drug-induced hypoglycemia all refer to hypoglycemia that results from the use (intentional or accidental) of insulin or insulin secretagogues. Clinical clues to the diagnosis may include unusual or inconsistent histories, such as severe, recurrent hypoglycemia without typical precipitating factors (e.g., fasting, illness), or access to antidiabetic agents. Biochemical findings of elevated plasma insulin with suppressed c-peptide (or an insulin to c-peptide molar ratio >1) confirm exogenous insulin administration. Various insulin assays differ in their sensitivity to detect insulin analogs, so it is important to understand the detection abilities – and limitations – of the assay used. Failure to do so may result in incorrectly excluding the diagnosis of exogenous insulin administration. Information on the cross-reactivity of commercially available insulin formulations with the ordered insulin assay is available on the test information page of most laboratories and can also be requested. For example, currently, cross-reactivity of most insulin assays with glulisine is very low (96). When clinical suspicion is high, sending specimens for testing using different immunoassays with different insulin formulation cross-reactivity profiles may be helpful. In contrast, insulin secretagogues (e.g., sulfonylureas, meglitinides, GLP-1 receptor agonists) stimulate both insulin and c-peptide secretion. Consequently, the biochemical evaluation of hypoglycemia due to insulin secretagogue use may be indistinguishable from that of insulinoma. Specialized toxicology panels, including measurement of plasma or urine sulfonylureas, may be required to confirm the diagnosis. A high index of suspicion, and knowledge of hypoglycemia agents available in the home can help guide the evaluation. As with exogenous insulin administration, the laboratory evaluation is subject to pitfalls in sensitivity and interpretation, and consultation with the laboratory is recommended (97).

 

Hypoglycemia Due to Growth Hormone and Cortisol Deficiencies

 

Deficiencies in the counter regulatory hormones, growth hormone and cortisol – either in isolation, or more commonly, in combination – cause hypoglycemia. Growth hormone acts to stimulate lipolysis and decrease peripheral glucose uptake. Cortisol stimulates gluconeogenesis and release of gluconeogenic substrates, including alanine, from muscle. Although secretion of both growth hormone and cortisol is triggered by falling plasma glucose, findings of a single low growth hormone or cortisol level at the time of hypoglycemia has poor specificity for deficiency of either hormone (18,19). Stimulation testing may be needed to confirm the diagnosis.

 

In the older child, hypoglycemia due to growth hormone and/or cortisol deficiency is ketotic. However, in neonates, the biochemical picture may mirror that of hyperinsulinism due to both inappropriate conservation of glycogen reserves during hypoglycemia and immature ketogenesis at this age.

 

HYPOPITUITARISM

 

Hypopituitarism may be congenital or acquired. Congenital hypopituitarism may result from malformation of the hypothalamus and pituitary (e.g., holoprosencephaly, septo-optic dysplasia) or from mutations in transcription factors vital for normal hypothalamic-pituitary development (see the chapter entitled “Genetic Etiology of Congenital Hypopituitarism” in the Pediatric Endocrinology section of Endotext (98)). As discussed above, mutations in FOXA2cause both hypopituitarism and hyperinsulinism. Acquired hypopituitarism may develop following trauma, infection, tumor, intracranial surgery, or radiation. In neonates, hypoglycemia is often a presenting feature of panhypopituitarism. Other clinical clues in neonates include unconjugated hyperbilirubinemia, nystagmus, midline developmental defects (e.g., cleft lip and palate), and in males, micropenis or smaller than average penis. An MRI of the hypothalamic-pituitary region is essential and often reveals an ectopic “bright spot” indicating disruption of the normal descent of the neurohypophysis with interruption of hypothalamic releasing factors that regulate hormone secretion. In such cases, prolactin concentration may be elevated, whereas GH, TSH and ACTH are suppressed. In older children, the diagnosis may be suspected based upon clinical history, growth failure, or neuroimaging findings. Hypoglycemia is found more commonly in neonates with multiple pituitary hormone deficiencies compared to isolated growth hormone deficiency and therefore careful consideration of multiple hormonal deficiencies must be given when growth hormone deficiency is found to be the cause of hypoglycemia in neonates (99).  Hypoglycemia is treated by replacement of the deficient hormones. From a practical aspect some neonates and infants will need a higher-than-expected dose of cortisol replacement and may even need growth hormone treatment divided twice daily. A safety fast (Table 7) should be performed prior to discharge to ensure the therapy is effective.

 

ISOLATED GROWTH HORMONE DEFICIENCY

 

Growth hormone deficiency has an estimated prevalence of between 1:4,000-1:10,000. Growth failure is the most common presenting feature, but this typically does not manifest until after the first year of life. Other clinical clues include midface hypoplasia and altered body composition with truncal adiposity. Most cases of isolated growth hormone deficiency are idiopathic. Genetic, anatomic, and acquired causes are detailed in the chapters entitled “Disorders of Growth Hormone in Childhood” and “Genetic Etiology of Congenital Hypopituitarism” in the Pediatric Endocrinology section of Endotext (98,100). Growth factors (IGF-1 and IGFBP-3) are low for age and bone age is delayed. The diagnosis is confirmed via stimulation testing. Treatment is with recombinant growth hormone. Importantly, depending on the underlying etiology, there is the potential for other pituitary hormone deficiencies to develop over time. Periodic screening of pituitary function is thus recommended.

 

ISOLATED CORTISOL DEFICIENCY

 

Cortisol deficiency may result either from defects in the ACTH signaling pathway or from congenital (e.g., congenital adrenal hyperplasia or hypoplasia) or acquired (e.g., bilateral adrenal hemorrhage) defects in adrenal steroidogenesis. Isolated ACTH deficiency is extremely rare. Several genetic etiologies have been described to date, including mutations in TBX19, POMC, PCSK1, and NFKB2 (see the chapter entitled “Genetic Etiology of Congenital Hypopituitarism” the Pediatric Endocrinology section of Endotext (98)). Of these, mutations in TBX19 are most frequently detected. Affected neonates universally present with severe hypoglycemia, often with hypoglycemic seizures. The mortality rate is up to 25%. In contrast to isolated ACTH deficiency, children with adrenal insufficiency due to ACTH resistance or primary adrenal disorders will have elevated plasma ACTH levels and associated skin hyperpigmentation. Primary disorders of the adrenal gland may also manifest with hyponatremia, hyperkalemia, and/or ambiguous genitalia. Congenital adrenal hyperplasia, and other disorders of the adrenal gland are detailed in the chapter entitled “Congenital Adrenal Hyperplasia” in the Adrenal Disease and Function section of Endotext (101). Treatment is with cortisol replacement. Initially, stress dose concentrations of hydrocortisone should be used.

 

GLYCOGEN STORAGE DISORDERS

 

The glycogen storage disorders (GSD) are a group of conditions in which there is either abnormal storage or release of glycogen resulting in hypoglycemia and acidosis. The acidosis may be lactic or ketoacidosis depending on the type of GSD. For the purpose of this discussion, we will include GSD 1 caused by glucose-6-phosphatase (G-6-Pase)deficiency as a glycogen storage disorder although the enzyme also represents the terminal step in gluconeogenesis. The GSDs may affect both liver and muscle glycogen storage and for this chapter we will focus on those with primarily liver expression, which are responsible for the hypoglycemic GSDs.

 

Under fed circumstances, excess glucose is converted into glycogen and stored in liver and muscle. Liver glycogen (but not muscle glycogen as muscle does not have G-6-Pase) later becomes available during fasting to provide glucose for metabolism in the brain and the glucose dependent tissues (red blood cells and proximal convoluted tubule of the kidney). Glucose is phosphorylated to G-6-Phos by glucokinase and then to Glucose-1-phosphate (G-1-Phos) by phosphoglucomutase. G-1-Phos is the starting point for glycogen synthesis by glycogen synthase (GSD 0, gene GYS2, inheritance autosomal recessive [AR]) to form chains with alpha 1-4 linkages. Branch points in these chains are formed by alpha 1-6 linkages approximately every 10 glucose units. During the early stages of fasting, glycogen is broken down by glycogen phosphorylase and then by glycogen debrancher enzymes. First the alpha 1-4 links are cleaved into G-1-Phos by glycogen phosphorylase (GSD VI, gene PGYL, inheritance AR) until 4 glucose units remain and then the debrancher transferase (GSD III, gene AGL, inheritance AR) moves the last 3 alpha 1-4 linked glucose over to another chain and then cleaves the alpha 1-6 branch point releasing a single glucose molecule. At this stage, the G-1-phos molecules are then converted to G-6-phos which then either undergoes glycolysis for energy or dephosphorylation by G-6-Pase (GSD 1, gene GCPC inheritance AR) and released into the blood stream as glucose.

 

Glycogen Storage Disease Type 0

 

GSD 0 is caused by deficiency of glycogen synthetase, which is encoded by the GYS2 gene on chromosome 12p12.2 and inherited in an autosomal recessive manner. The main biochemical manifestations are both fasting ketotic hypoglycemia and postprandial hyperglycemia and hyperlactatemia (102). This is due to the inability of the liver to store excess postprandial glucose resulting in hyperglycemia and glycosuria. The G-6-phos undergoes glycolysis to pyruvate and then lactate. Later as fasting progresses, there are no liver glycogen stores available, and when the glucose drops below 85 mg/dL, insulin secretion is switched off and early ketosis occurs resulting in ketotic hypoglycemia. This typically happens after 6-12 hours of fasting. Dependence on gluconeogenesis also results in over-utilization of protein, and protein deficiency is common.   

 

Clinical features of GSD 0 may range from asymptomatic to recurrent episodes of ketotic hypoglycemia.  Typically, as infants transition off nighttime feeding, episodes of fasting hypoglycemia with hyperketonemia occur. Patients may also present during mild gastrointestinal disorders with ketotic hypoglycemia. They have no hepatomegaly and a normal critical sample with elevated counter regulatory hormones, elevated free fatty acids and ketones. They may have short stature, a history of failure to thrive and hyperlipidemia. In the most severe end of the spectrum, they may present with seizures and developmental delay in addition to hypoglycemia after a very short duration fasting. The condition should be suspected in children with a history of recurrent ketotic hypoglycemia or post prandial hyperglycemia and fasting ketonemia.

 

Diagnosis is made by demonstrating shortened fasting tolerance with ketotic hypoglycemia (103) (typically <12 hours in children under 7 years of age and <18 hours in adolescents). In addition, an oral glucose tolerance test (OGTT) performed after an overnight fast using 1.75 g/kg up to 75 g (one of the few appropriate uses of the OGTT for diagnosis of etiology of hypoglycemia) will demonstrate postprandial hyperglycemia with elevated lactic acid. Finally, a fed (2 hours post meal) glucagon stimulation test will fail to show elevated glucose response to glucagon in the most severe cases but may cause an increase in milder cases. Once the clinical diagnosis is suspected, genetic testing for confirmation of diagnosis is strongly recommended rather than liver biopsy.  

 

Treatment is avoidance of fasting (>3-4 hours), utilizing low glycemic index carbohydrate, and a high protein diet (2-3g/kg/day) during the day with meals and snacks to provide adequate amino acids for gluconeogenesis and overnight dextrose via gastrostomy tube or uncooked cornstarch (UCS) after age 1 year (1.5g/kg every 6 hours overnight). Glycosadeâ a soluble extended-release form of amylopectin cornstarch is available in the US for children over the age of 5 years and may be used in place of uncooked cornstarch. Early diagnosis and treatment may prevent the long-term complications of short stature and osteopenia from the recurrent keto- and lactic acidosis. 

 

Glycogen Storage Disease Type I

 

This is the most severe of the GSDs and causes profound hypoglycemia because of impaired glycogenolysis and gluconeogenesis with lactic acidosis, hyperuricemia, and hyperlipidemia.  Glycogen and triglycerides are stored in the liver resulting in massive hepatomegaly. Long-term consequences of GSD I include hepatic adenoma, renal Fanconi syndrome, renal failure, short stature, and osteoporosis. It occurs in approximately 1:100,000 births (104), and is caused by mutations in the gene for G-6-Pase, G6PC (GSD Ia), or G-6-P translocase 1, G6PT1 (GSD Ib).  Under normal circumstances G-6-P translocase 1 transports G-6-Phos into the endoplasmic reticulum, G-6-Pase then converts G-6-P to glucose which is then transported out of the endoplasmic reticulum by GLUT-2 (hence the similarity of Fanconi Bickel syndrome caused by GLUT2 deficiency to GSD 1).

 

In the postprandial phase, glycogen stored in the liver is broken down to G-6-Phos and from here can enter 3 metabolic pathways: 1) G-6-Phos enters the pentose phosphate shunt resulting in formation of uric acid. 2) G-6-Phos undergoes glycolysis to form pyruvate and then to lactic acid (which is transported to brain and used as fuel for energy production). 3) G-6-Phos undergoes glycolysis and forms acetyl CoA which in turn is converted into malonyl CoA which inhibits carnitine palmitoyl-transferase I leading to decreased oxidation of fatty acids (impaired ketone body production) and increased formation of lipids causing hyperlipidemia. Thus, the cardinal biochemical features of GSD 1 occur (hypoglycemia, hyperuricemia, hyperlactatemia, and hypertriglyceridemia).

 

Clinical features in the newborn may include hypoglycemia with a good response to feeding, however because many newborns breast feed every 2 hours GSD 1 is rarely identified in the new-born period. Despite the occurrence of hypoglycemia, newborns are rarely diagnosed because many physicians erroneously believe that if the glucose responds to feeding then ongoing glucose testing is not needed. However, if the PES recommendations (21) are followed and persistent glucose levels <50 mg/dL are noted in the first 48 hours of life and <60 mg/dL beyond that, then investigations should be done to determine the etiology. A six-hour fasting study will identify these patients and indeed great care must be taken if suspected because glucose levels typically fall very quickly (2.5-3.5 hours after a feed) and fall deeply to the 20-30 mg/dl range. As infants get older and feeding intervals start to stretch out to >6 hours over night, significant hypoglycemia and lactic acidosis occurs and often the children present because of the tachypnea caused by the acid base disturbance, rather than the hypoglycemia.  Neuroglycopenic symptoms are unlikely to occur due to the protective effect of lactate metabolism in the brain, but in circumstances of eating high carbohydrates and a rapid decline in glucose due to insulin release, the lactate levels may be low and the neuroprotective effects of lactate may not occur. In these circumstances seizures, coma, and sudden death may occur.  Clinical features of undiagnosed GSD 1a include massive hepatomegaly, short stature, and failure to thrive. Biochemically, patients have high lactate levels (typically >5mmol/L) with hypoglycemia, low ketones, abnormal transaminases, high uric acid, and hypertriglyceridemia. 

 

GSD Ib is caused by defects in the G6PT1 (also known as SLC37A4) gene and has all the same clinical findings as above but in addition has significant neutrophil dysfunction resulting in recurrent skin infections and later in life severe inflammatory bowel disease similar to Crohn disease. This form of GSD 1 represents about 10% of cases of GSD 1 and is an AR inherited condition with the gene found on chromosome 11q23.  It is estimated to occur in 1:1,000,000 births.

 

It is important to note that untreated GSD 1a/b patients rarely present with neuroglycopenic symptoms of hypoglycemia because lactate may be used a fuel for the brain, and they also rarely present with neurogenic symptoms as they develop hypoglycemic unawareness. Thus, the hepatomegaly, short stature, intermittent tachypnea, and the finding of lipemic serum rather than symptoms of hypoglycemia raise the clinical suspicion.   

 

Diagnosis of GSD Ia or Ib is made by finding the clinical features above and performing genetic testing for mutations in G6PC or G6PT1 to confirm the diagnosis. 

 

Treatment is to prevent hypoglycemia by providing glucose every 2.5-3 hours in neonates using fructose/galactose free formula. In GSD 1, fructose and galactose cannot be converted to glucose due to deficiency of G-6-Pase and so if given will worsen lactic acidosis, hyperlipidemia, and hyperuricemia. The goal of treatment is to maintain the plasma glucose >75mg/dl which is the threshold for the secretion of counter-regulatory hormones that drive glycogenolysis and glycolysis to lactic acid. This can be achieved by frequent oral feeding in newborns, with continuous gastrostomy tube feeds overnight, and in older infants (>1 year), toddlers and children giving uncooked starch (UCS) 1- 1.5 g/kg every 3-6 hours including overnight. Glycosadeâ, an extended-release waxy maize cornstarch, can be used in older children to avoid overnight dosing with a duration of action of 6-8 hours, but is not recommended under 5 years of age. Placement of a gastrostomy tube to provide intra gastric glucose overnight often improves the family’s quality of life and the patient’s metabolic control. One caveat to feeding fast acting carbohydrates is that the insulin production suppresses glycogenolysis and production of lactic acid so treated patients have a higher risk of neuroglycopenic symptoms and brain damage from profound hypoglycemia without lactic acidosis if regular therapy is interrupted or delayed. Typical dietary treatments include 65% carbohydrates, 10-15% protein and low fat of approximately 20-25%. A simple rule of thumb when providing IV glucose or continuous enteral glucose is to start at around 8 mg/kg/min and titrate until lactate is <2 mmol/L and glucose >75mg/dl. Chronic provision of excess glucose is not beneficial so care must be taken to find the ideal amount of glucose (to paraphrase Goldilocks, not too much, not too little, just right!). Because hepatic production of glucose generally is equal to brain utilization of glucose, neonates have a higher glucose requirement than children and children than adults due to the relative sizes of the brain to body. Overall, 30-40% of the daily carbohydrate should be long-acting carbohydrates.

 

Glycogen Storage Disease Type III

 

GSD III is caused by glycogen debrancher enzyme deficiency. This is encoded by the AGL gene on chromosome 1p21 and is inherited in an autosomal recessive manner (105). It occurs in about 1:100,000 similar to GSD I. GSD IIIa occurs in both liver and muscle and represents the majority of cases in the US while GSD IIIb occurs in liver alone. 

 

Clinical features of GSD III include fasting ketotic hypoglycemia and massive and firm hepatomegaly. Unlike GSD I, GSD III is characterized by normal lactic and uric acid levels and more severe elevations in liver function enzymes. In those with GSD IIIa marked elevations of creatine kinase (CK) appear before the clinical signs of proximal muscle wasting. 

 

Clinical diagnosis is made by the finding of short fasting induced hypoglycemia without lactic acidosis, hepatomegaly, marked elevation of transaminases often with high CK. At the time of hypoglycemia, glucagon will not cause a rise in the plasma glucose but 2 hours after a meal in the fed state it will trigger a rise in glucose unlike GSD I.  Genetic testing will identify mutations in the AGL gene.

 

Treatment is the avoidance of prolonged fasting using UCS in doses of 1-1.5g every 4-6 hours but unlike GSD I, patients with GSD III can utilize amino acids and efficiently carry out gluconeogenesis. Thus a high protein diet in addition to UCS can be very effective in preventing hypoglycemia, minimizing hepatomegaly, improving transaminases, improving growth and even proximal muscle wasting (106). Avoidance of too much carbohydrate is also important to avoid filling the liver, heart, and muscles with glycogen, which can cause long term harm.  A hypertrophic cardiomyopathy is a complication of excessive glycogen storage, but the muscle disease appears to be secondary to lack of an energy substrate during activity. A ketotic diet may help improve muscle disease in adults, but it has been associated with worsening of the hepatic transaminases and suboptimal growth in the pediatric population. 

 

Glycogen Storage Disease VI and IX

 

GSD VI is caused by glycogen phosphorylase and GSD IX by glycogen phosphorylase kinase.   Both conditions are remarkably similar and the most common of all the hepatic GSDs. In fact there has been a suggestion that GSD VI and IX together may account for a significant number of children with recurrent ketotic hypoglycemia (107). GSD VI is caused by mutations in PYGL and is inherited in an autosomal recessive form. On the other hand, there are 4 subunits of the phosphorylase kinase enzyme, and each is encoded by different genes including the X-linked gene PHKA2. PHKB mutations lead to an autosomal recessive form of GSD IX as do mutations in PHKG. The most severe form tends to occur in patients with genetic mutations in PHKG.

 

Clinical manifestations of GSD VI and IX are short stature and recurrent episodes of ketotic hypoglycemia. Unlike the hepatomegaly of GSDI and III in which the examiner will need to start palpating the liver from the anterior superior iliac spine in order not to miss a giant liver, the hepatomegaly of VI and IX may be very subtle or absent. Sometimes in younger children morning ketosis is found with euglycemia because after glycogen stores are depleted, gluconeogenesis and ketogenesis are activated and glucose levels remain normal. Thus, if suspecting GSD VI or IX, check morning beta-hydroxybutyrate and if >0.6 mmol/L after a normal overnight fast this would be suggestive of this disease. Also because of the reliance on gluconeogenesis, prealbumin and total protein levels may be low, and patients may complain of intermittent muscle aches without overt weakness.

 

Diagnosis is suspected by finding shortened fasting tolerance with accelerated development of ketosis in a patient with mild short stature and hepatomegaly. Mild elevations of liver transaminases and hyperlipidemia may occur. Absence of hepatomegaly does not rule out GSD VI or IX. Genetic testing is the diagnostic test of choice for those with either recurrent episodes of ketotic hypoglycemia or persistent finding of elevated beta-hydroxybutyrate after a simple overnight fast.

 

Treatment is avoidance of prolonged fasting and administration of UCS 1-2 g/kg at bedtime to prevent early morning ketosis. A high protein diet with complex carbohydrates is effective as gluconeogenesis is unaffected. A program of daytime carbohydrates for intercurrent illness and a plan for IV dextrose with vomiting is encouraged. By the time the child reaches adulthood fasting tolerance is of 12-18 hours overnight without ketones become possible and treatment is rarely required as an adult.

 

FATTY ACID OXIDATION AND KETONE BODY DISORDERS

 

Fatty Acid Oxidation Disorders

 

Fatty acid oxidation (FAO) occurs in the mitochondria and is the major source of energy production in the fasted state i.e., when glucose metabolism is insufficient to provide for energy needs. Typically, FAO starts when glycogen reserves are depleted in conjunction with an increase in gluconeogenesis. As insulin levels fall, hormone-sensitive lipase acts in the adipocytes to release 3 free fatty acids and 1 glycerol from the triacyl-glycerol (triglyceride) stored in the adipose tissue (108). The metabolism of the free fatty acids occurs in 3 major steps: 1) transport into the mitochondria via the carnitine shuttle, 2) long chain fatty acids undergo beta oxidation at the inner mitochondrial membrane, and then 3) medium and short chain fatty acid oxidation (FAO) occurs in the mitochondrial matrix. The result of FAO is the production of acetyl-CoA, which in the heart and skeletal muscle enters into the citric acid cycle and by oxidative phosphorylation generates energy for these tissues. The liver however converts acetyl-CoA into ketone bodies known as beta-hydroxybutyrate (BOHB, the main ketone body in the blood) and acetoacetate via hydroxyl methyl glutaryl-coenzyme A (HMG-CoA) synthetase and HMG-CoA lyase. The ketone bodies are exported to the brain for energy use. The liver also metabolizes the amino acids leucine and isoleucine into acetyl-CoA. Because fatty acids cannot cross the blood brain barrier, fatty acid oxidation is limited in the brain due to a lack of substrate. MCT1, a member of the monocarboxylate transporter family, transports the ketone bodies across the blood brain barrier. BOHB is then converted back to acetoacetate and then to acetyl-CoA which enters the citric acid cycle and generates energy in the form of adenosine triphosphate (ATP).  Studies have shown that as the plasma glucose falls the cerebral metabolic rate of glucose falls and as the plasma acetoacetate levels rise the cerebral metabolic rate acetoacetate rises replacing the lost metabolism of glucose (13). Thus, BOHB can add to glucose as a primary source of energy production by the brain. This protects the brain from hypoglycemic brain damage in circumstances of low glucose and high ketones (with the exception of patients with ketone utilization defects). This also highlights why untreated disorders of fatty acid oxidation plus ketone synthesis and utilization are dangerous as they result in accelerated development of hypoglycemia because of the loss of the glucose sparing ketones, resulting in energy failure in the brain.

 

Abnormalities of FAO tend to present either in periods of prolonged fasting often precipitated by intercurrent illness when the need for energy at a cellular level, increases, or during severe strenuous exercise. Common clinical features of these defects include fasting hypoglycemia with liver failure, hepatic encephalopathy, and muscular hypotonia.  Rhabdomyolysis following exercise is also common. Rare clinical effects might include cardiac arrhythmias and retinitis pigmentosa. The biochemical hallmark of FAO defects is hypoglycemia associated with elevated free fatty acids and inappropriately low ketone bodies (Table 3 and Figure 2). Coagulation defects, elevated liver function tests, and hyperammonemia may occur with massive elevation of serum CK levels.

 

FAO disorders are inherited as autosomal recessive conditions. The overall incidence has been reported to be approximately 1:9000 (109). Since the development of newborn screening for FAO disorders, the clinical presentation has changed with almost no patients presenting with hypoglycemia. Indeed, the incidence of hypoglycemia caused by previously unknown FAO disorder presenting in emergency room situations is dramatically reduced. In a study of approximately 220,000 children presenting at an emergency room between the ages of 0 and 18 years, 160 patients had previously undiagnosed hypoglycemia and none of them had a FAO defect (110). This compares to a study by Weinstein et al prior to universal newborn screen of FAO disorders in which FAO disorders represented 19% of previously undiagnosed hypoglycemia (111). Although most FAO defects are detected on newborn screening, rarely errors in the process may account for a missed case and it is always important to demonstrate in cases of hypoketotic hypoglycemia the absence of a FAO disorder.

 

This chapter will focus primarily on those disorders that primarily cause hypoglycemia and the acute treatment of the hypoglycemia.

 

DISORDERS OF FATTY ACID UPTAKE INTO MITOCHONDRIA AND THE CARNITINE CYCLE

 

Free fatty acids (FFA) are released from adipose tissue and travel to target cells where they are transported across the cell membrane by fatty acid transporter proteins. Once inside the cell they are esterified by acyl-CoA synthases and enter the carnitine cycle to enter the mitochondria. Carnitine palmitoyl transferase I (CPT I) catalyzes transfer of the acyl group from acyl-CoA to form acyl-carnitine and is the rate limiting step in FAO. The acyl-carnitine is transported across the mitochondrial membrane by carnitine acylcarnitine transferase (CACT) and once on the inside the acylcarnitine is converted back to acyl-CoA by CPT II. The acyl-CoA is now ready to undergo beta oxidation. The carnitine for these reactions is transported into the cell by the carnitine transporter. 

 

Disorders of the carnitine transporter, CPT I and CACT all cause hypoketotic hypoglycemia.  Both CPT 1 and CACT cause fasting-induced Reye syndrome with acute liver failure and can be suspected when hyperammonemia is found with hypoketotic hypoglycemia, elevated free fatty acids, and abnormal acylcarnitine profile. Carnitine transporter deficiency cause severe carnitine deficiency and dilated cardiomyopathy that is progressive and generally presents before hypoglycemic episodes. Diagnosis of carnitine transporter deficiency is suspected when total carnitine levels are very low (<10% normal) in conjunction with hypoketotic hypoglycemia and elevated CK levels in a patient with cardiomyopathy. Reye syndrome and acute liver failure also occur in CPT I and CACT and acute myoglobinuria with rhabdomyolysis occur in CPT II.

 

DISORDERS OF BETA OXIDATION

 

Beta-oxidation is the process whereby sequential molecules of acetyl-CoAs are cleaved off the long acyl-CoA inside the mitochondria, until the entire acyl-CoA has been broken down to acetyl-CoA (thus an 18-carbon acyl-CoA will generate 9 acetyl-CoAs). The first step in this reaction is carried out by four individual acyl-CoA dehydrogenase enzymes each acting on a different chain length acyl-CoA. These are the short, medium, long and very long chain acyl CoA dehydrogenase (SCAD, MCAD, LCAD and VLCAD) enzymes. The next three steps of FAO are carried out in conjunction with the tri-functional protein and involve the hydratase enzymes and both long chain and short chain 3-hydroxyacyl-CoA dehydrogenases (LCHAD and SCHAD). Finally electron transport flavoproteins transfer electrons from reducing equivalents produced by multiple pathways to the electron transport chain. Defects in the electron transport chain cause multiple acyl-CoA dehydrogenase deficiency (MADD) also known as Glutaric Aciduria II.

 

Clinical features of defects in beta-oxidation include hypoketotic hypoglycemia, associated with lethargy, vomiting, hypotonia seizures. Acute liver failure or Reye syndrome can occur, and cardiomyopathy is particularly severe in the longer chain disorders. It should be noted that ketones can be produced in variable amounts, but they are not sufficient to prevent hypoglycemia and the ratio of FFA to ketones is increased over normal. Individual FAO disorders have diagnostic acylcarnitine profiles and urine organic acid profiles and diagnosis may be made during an acute decompensation using these tools. However, one should never subject a patient suspected of having a FAO disorder to a fasting study for the purpose of diagnosis as it can precipitate an acute decompensation and death. If a FAO disorder is suspected, perform an acylcarnitine profile, total and free carnitine and test for urine organic acids in the well state, which may diagnose the condition without triggering a decompensation.

 

EMERGENCY TREATMENT OF FATTY ACID OXIDATION DISORDERS

 

Emergency treatment of hypoketotic hypoglycemia caused by FAO disorders involves correcting the hypoglycemia with a 200 mg/kg bolus of dextrose (best provided as 2 ml/kg Dextrose 10%) followed by administration of 5-10 mg/kg/min dextrose infusion with the goal of getting the plasma glucose above 85 mg/dl to turn on insulin secretion and suppress lipolysis. Treatment of elevated ammonia may be required if levels do not promptly fall with glucose administration.  Cardiopulmonary shock if present needs to be aggressively managed and liver failure and rhabdomyolysis need to be suspected and diagnosed early to implement treatment. Cerebral edema may occur particularly associated with elevated ammonia in undiagnosed cases of MCAD deficiency. Diuresis with possible alkalization of the urine can prevent acute renal failure in cases of extreme elevation of CK. Consultation with a metabolic expert is strongly recommended for endocrinologists not familiar with the ongoing management of children with FAO disorders. 

 

Ketogenesis and Ketone Utilization Defects

 

Ketone synthesis involves two primary enzymes involved in converting acetyl-CoA to acetoacetate, hydroxyl methyl glutaryl-coenzyme A (HMG-CoA) synthase and HMG-CoA lyase.  The ketone utilization defects are rare conditions in which ketogenesis is effective but there is an inability to either convert acetoacetate to aceto-acetyl-CoA by succinyl-CoA:acetoacetate transferase (SCOT) or acetoacetyl-CoA to acetyl-CoA by mitochondrial acetoacetic thiolase (MAT). These genetic defects are all autosomal recessive conditions. Finally, ketone transport into the brain can be diminished in patients with loss of function variations in SLC16A1 the gene encoding the monocarboxylate transporter 1 protein (MCT1) (112,113).

 

KETOGENESIS

 

There are significant similarities between the FAO defects and the ketogenesis disorders with hypoketotic hypoglycemia with or without hyperammonemia and hepatic encephalopathy.  Patients present in the newborn period or during intercurrent illness with relatively short episodes of fasting (12-18 hours). The biochemical hallmarks of HMG-CoA synthase deficiency are hypoglycemia with decreased ketones, elevated FFA, normal lactate and ammonia and normal urine organic acids and plasma amino acids whereas HMG-CoA lyase deficiency presents with hypoketotic hypoglycemia with elevated lactate and ammonia and very abnormal urine organic acid profile and elevated 3-methylglutaryl carnitine in the acylcarnitine profile. 

 

Treatment is similar to the FAO defects and includes rapid correction of hypoglycemia with 200 mg/kg intravenous bolus of dextrose (2 ml/kg Dextrose 10%) followed by an infusion of 5-8 mg/kg/min of glucose (3-5 ml/kg/h of Dextrose 10%). Treatment of the lactic acidosis with bicarbonate is not indicated unless life threatening acidosis is occurring as provision of glucose generally corrects the acidosis. Supportive care for hyperammonemia and hepatic encephalopathy should be provided.

 

KETONE UTILIZATION AND TRANSPORT DEFECTS

 

Ketone utilization defects generally present in the newborn period however some patients will survive the neonatal period and present later with intermittent severe metabolic acidosis due to ketoacidosis associated with fasting or intercurrent illness. Outside of an acute episode they may have elevated plasma BOHB >0.4 -0.6 mmol/L even when well fed indicating an inability to suppress ketones. Hypoglycemia is not the most common finding but can occur after prolonged fasting with or without hyperammonemia, hepatomegaly and encephalopathy. During fasting studies plasma BOHB continues to rise until progressive ketoacidosis occurs which is different to the rising and then stable ketosis of physiological fasting that stabilizes out as ketone utilization and ketogenesis balance out. 

 

Treatment is to provide glucose to increase insulin levels and suppress fatty acid oxidation.  This is typically achieved by providing a GIR of 8 mg/kg/min. Treatment of acidosis with bicarbonate therapy is controversial and should be used with caution in the severest cases.

 

 

CLINICAL APPROACH TO THE DIAGNOSIS OF THE ETIOLOGY OF HYPOGLYCEMIA

 

In order to diagnose the etiology of hypoglycemia one must first confirm the presence of hypoglycemia. Normal glucose levels are typically between 70 and 120 mg/dL and glucose levels less than 70 mg/dL in the face neurogenic or neuroglycopenic symptoms are suggestive of hypoglycemic disorders. In children (>5 to 7 years old) and young adults who are reliably able to report the symptoms of hypoglycemia, the finding of Whipple's triad (symptoms consistent with hypoglycemia, a measured plasma glucose confirming hypoglycemia, and improvement of symptoms with administration of glucose and correction of the hypoglycemia) is sufficient to warrant further investigation. For infants and younger children who are unable to reliably communicate symptoms, the Pediatric Endocrine Society (PES) guidelines suggests evaluation and management only of those whose plasma glucose concentrations are documented by laboratory quality assays to be below the normal threshold for neurogenic responses (60 mg/dL) (21).  For any patient with a low glucose value measured either by point of care meter using a finger stick blood sample or using a continuous glucose monitoring system, a plasma glucose needs to be obtained to confirm hypoglycemia prior to commencing a complete evaluation. This recommendation has been made because of the known inaccuracy of point of care glucose testing and of continuous glucose monitor testing for detecting blood glucose less than 70 mg/dL. Rather than providing children and their families with a glucometer for screening for low blood sugars at home, whenever safely possible, a standing order for a plasma glucose to be analyzed immediately, should be offered to the family. When they experience symptoms consistent with hypoglycemia, they can have a plasma glucose drawn in the lab to either confirm the presence of hypoglycemia or to demonstrate that these symptoms are not related to hypoglycemia. Thus, the presence or absence of Whipple's triad may be confirmed and only those with a high likelihood of hypoglycemia undergo further investigations. Studies have shown that the frequency of pathological hypoglycemia in patients in high-risk situations (for example the emergency room or attending a hypoglycemia clinic) who have had a documented low blood sugar associated with intercurrent illness will have a risk of a serious form of hypoglycemia of approximately 10% (110,114). 

 

History

 

The first step in the evaluation is to undertake a careful history specifically questioning for symptoms of hypoglycemia (Table 4), and timing of the symptoms, or measured hypoglycemia, relative to the last meal consumed. In pediatric patients, the vast majority of causes of hypoglycemia are precipitated by fasting. Documenting post prandial hypoglycemia in the absence of fasting hypoglycemic may suggest some very specific conditions, such as late dumping syndrome (Nissen fundoplication induced hypoglycemia, post bariatric surgery induced hypoglycemia), protein induced hypoglycemia found in GDH HI, HADH HI, and KATP HI, hereditary fructose intolerance, or a very rare presentation of insulinoma. Careful review of the history for the presence of neurogenic or neuroglycopenic symptoms should be performed. Neuroglycopenic symptoms are strongly suggestive of a serious underlying form of hypoglycemia and need rapid evaluation. A history of poor growth might suggest growth hormone deficiency, adrenal insufficiency (either primary or secondary), or GSD. Recurrent abdominal pain with nausea and vomiting is a classical symptom of severe adrenal insufficiency. A history of salt craving or darkening skin also are clues to primary adrenal insufficiency.

 

The most common time of presentation of hypoglycemia in childhood is in the neonatal period when the stress of transition from intrauterine to extra uterine life is greatest and genetic or metabolic forms of hypoglycemia most commonly present. It is important to always inquire about the neonatal period because of the possibility of missed diagnosis. Presentation to emergency rooms in association with intercurrent illness is the next most common time of presentation of hypoglycemia in childhood; it is recommended that emergency rooms should have protocols to draw the critical samples (Table 2) in patients who present with hypoglycemia, but with no previous etiology determined. If an emergency room does not have such protocols in place, we recommend their implementation, as up to 10% of all patients presenting to emergency rooms with previously undiagnosed hypoglycemia have a serious underlying condition requiring long-term treatment (110,114).

 

Physical Exam

 

The second step in the evaluation of the etiology of hypoglycemia is the physical exam. There are very few clues that can be obtained; however, it is critical not to miss those that are available. Short stature, with impaired growth velocity, or inappropriate height for the family, should suggest evaluation for growth hormone deficiency. Hyperpigmentation of the skin and or gums could indicate primary adrenal insufficiency with ACTH excess. Failure to thrive and weight loss could indicate chronic adrenal insufficiency. Central malformations such as cleft lip or palate could indicate an underlying pituitary problem. Scars on the abdominal wall or a gastrostomy tube could suggest a postsurgical form of hypoglycemia. Enlargement of the liver at a time when the patient is well would point to glycogen storage disorders, or if the patient is acutely unwell at the time of presentation a fatty acid oxidation defect might be more likely. Abnormal development of the genitalia might indicate adrenal hormonal production problems; a micropenis/small normal penis might indicate a growth hormone problem with or without hypogonadotropic hypogonadism.  Asymmetry of the body, either hemiatrophy or hemihypertrophy, could suggest an underlying syndrome such is Russell Silver Syndrome or Beckwith-Wiedemann Syndrome. Certain syndromes such as Down syndrome are associated with an increased incidence of ketotic hypoglycemia. Overgrowth syndromes, Turner syndrome, and Kabuki syndrome may be associated with hyperinsulinism (72,78,79).

 

Critical Sample at Time of Hypoglycemia

 

As noted above, a critical sample including blood and urine (Table 2) should be collected at the time of glucose less than 50 mg/dL. In this sample, the levels of intermediary metabolites and hormones in blood, ketones and organic acids in urine, and in certain circumstances the presence or absence of drugs such as insulin or sulfonylureas, will aid the physician to diagnose the etiology of the hypoglycemia. Interpretation of the critical sample commences with the determination of the presence or absence of ketosis and lactic acidosis as in Figure 2 and Table 3.

 

When a critical sample has not be obtained during a spontaneous episode of hypoglycemia, a fasting study should be performed to induce hypoglycemia (Table 10). Caution prior to admitting the patient for a fasting study, includes the need to rule out fatty acid oxidation defects by performing an acylcarnitine profile in the well state. Fasting studies should be performed in the inpatient setting in a unit of highly specialized nurses trained in the performance of fasting studies. Safety precautions such as having intravenous lines inserted for both blood drawing and infusing glucose in an emergency situation are very important. It is critical to have the ability to do accurate point of care glucose and ketone testing on venous samples or warmed capillary samples and to have a rapid turnaround on plasma glucose and plasma beta-hydroxybutyrate.  Intravenous glucose should be available for rescue and glucagon should also be available both for stimulation testing and for rescue when hyperinsulinism is suspected.

 

Table 10.  Diagnostic Fasting Test

Perform test only on a unit with trained medical and nursing staff who are experienced in the performance of fasting studies. 

1.     Have IV access and D10% (2-5 ml/kg) for emergency resuscitation.

2.     Measure glucose by POC meter every 2-3 hours until glucose <70 mg/dL (<3.9 mmol/L); then every 2 hours until <60 mg/dL (<3.3 mmol/L); then hourly until <50mg/dL (<2.8 mmol/L) 

a.     When glucose <60 mg/dL (<3.3 mmol/L) send specimen for laboratory confirmation of plasma glucose 

3.     Measure beta-hydroxybutyrate every 2-3 hours and when glucose <50mg/dL (<2.8 mmol/L)

a.     When plasma glucose ≤50 mg/dL (<2.8mmol/L) draw blood for the CRITICAL sample: 

glucose, insulin, beta-hydroxybutyrate, free fatty acids, ammonia, cortisol, growth hormone, lactate, acylcarnitine profile, urine organic acids

i.     Special circumstances: C-peptide, proinsulin, sulfonylurea screen, toxicology screen, serum amino-acids,

4.     Perform Glucagon Stimulation Test once CRITICAL samples are obtained 

1.              Measure glucose using POC meter and then give glucagon 30 mcg/kg or 0.5-1 mg by IM or IV push as long as glucose is <50 mg/dL (<2.8 mmol/L)

2.              Monitor glucose using POC meter every 10 minute for 40 minutes 

3.              Terminate test if glucose is still below 50 mg/dL (<2.8 mmol/L) after 30 minutes 

4.              After 40 minutes, may feed and resume treatment to maintain plasma glucose >70 mg/dL (3.9 mmol/L) 

Adapted from De Leon DD, Arnoux JB, Banerjee I, Bergada I, Bhatti T, Conwell LS, Fu JF, Flanagan SE, Gillis D, Meissner T, Mohnike K, Pasquini TLS, Shah P, Stanley CA, Vella A, Yorifuji T, Thornton PS. International Guidelines for the Diagnosis and Management of Hyperinsulinism. Horm Res Paediatr. 2023 (32).

 

When the results of the critical sample point to the likely area of metabolic perturbation (Figure 2), further testing may be indicated at times not necessarily at the time of hypoglycemia. For conditions such as hyperinsulinism, specific testing can further subclassify the etiology (such as protein sensitivity in GDH HI or HADH HI, or ammonia levels indicating GDH HI). In the case of low counter regulatory hormones found at the time of the critical sample, stimulation testing for growth hormone, cortisol, or ACTH deficiency can confirm if deficiencies in these hormones are the cause of hypoglycemia. One should never diagnose a hormonal deficiency as the cause of hypoglycemia on a single critical sample but rather only when dynamic testing has demonstrated deficiency (18,19). In cases suspected to be due to glycogen storage disease, liver biopsy is no longer indicated due to the morbidity of the procedure and genetic testing is now the preferred diagnostic method. This is also true for the disorders of fatty acid oxidation in which an acylcarnitine profile is not sufficient to make a diagnosis and genetic panels are preferable to skin biopsies the majority of the time. With current genetic testing technology, panels including many hypoglycemia disorder associated genes may be conducted for little more cost than a single gene test.

 

Approach to the Patient with Ketotic Hypoglycemia

 

There is a common misconception that children presenting with ketotic hypoglycemic, who do not possess an already identified underlying cause, have benign physiological idiopathic ketotic hypoglycemia. In recent years it has become clear that this is not correct (110,114). We outline an approach to patients with ketotic hypoglycemia that should allow differentiation between benign physiological ketotic hypoglycemia, pathological ketotic hypoglycemia associated with known underlying conditions and idiopathic pathological ketotic hypoglycemia (IPKH) in which there is clearly an abnormality of glucose regulation, but no genetic cause can be found (Figure 7).

 

Figure 7. Physiological vs. Pathological Ketotic Hypoglycemia.

 

BENIGN PHYSIOLOGICAL KETOTIC HYPOGLYCEMIA

 

Based on the normal physiology of fasting, if a person is fasted for long enough, they will develop a glucose <50 mg/dL and plasma beta-hydroxybutyrate >2 mmol/L. Studies have looked at the normal duration of fasting and found that time to glucose <50 mg/dL increases with age. Children 0-2 years can generally fast about 15-18 hours, children >2 years to 5 years can fast >24 hours, children >5 years to 10 years can fast about 36 hours, and teenagers to adults can fast 48-72 hours (115). Recently Pamar et al.  investigated children admitted for day surgery and found that only a small percentage developed beta-hydroxybutyrate levels >1 mmol/L by 12 hours of fasting (116). Thus, the finding of ketones in blood of >1 mmol/L after 12 hours of fasting in previously healthy and well-nourished children is unusual. However, the finding of ketosis after 24 -30 hours of no, or very poor, oral intake would be considered normal for most children over the age of 2 years. Thus, history is very important in differentiating benign physiological ketotic hypoglycemia due to prolonged starvation or intercurrent illness. It is our recommendation that previously healthy children who present with their first episode of ketotic hypoglycemia, who have a history consistent with prolonged starvation with or without intercurrent illness, who have elevated ketones >2 mmol/L and a normal physical exam, should not require further investigations. Children with recurrent episodes of ketosis despite precautions to avoid prolonged fasting need further investigation which may include admission for a fasting study. In addition to evaluating for an underlying etiology, this permits determination of the time to development of ketosis (by monitoring serial ketone levels, not just at the time of the critical sample) and hypoglycemia, which informs intervention. This is best achieved by a formal fasting study (Table 10) following three days of good feeding.

 

PATHOLOGICAL KETOTIC HYPOGLYCEMIA

 

The term pathological ketotic hypoglycemia is used to indicate that some underlying abnormality is causing disordered fasting tolerance. The key manifestation of this is ketotic hypoglycemia that occurs after an abnormally short fasting period, morning ketosis without hypoglycemia, or symptomatic ketosis causing vomiting and a vicious cycle of worsening ketosis, vomiting and dehydration. This can be secondary to hormonal deficiencies such as isolated growth hormone deficiency or cortisol deficiency (primary, secondary or tertiary). It can be caused by enzyme deficiencies in the glycogen storage pathway, disorders of gluconeogenesis, or disorders of ketone utilization or transport. In a small number of children, no cause can be found despite extensive clinical and genetic investigations. These children have idiopathic pathological ketotic hypoglycemia (IPKH). The approach to management of these children requires knowledge of the duration of fasting required to keep the beta-hydroxybutyrate <0.6 mmol/L and knowledge of how low the glucose will fall despite rising ketone levels. Thus, the therapeutic plan needs to be individually tailored to each child. Typical components will include high protein diet of 3 g/kg/day protein to promote gluconeogenesis and prevent catabolism of muscle and slow-release carbohydrates such as uncooked corn starch (UCS) or Glycosade®.  Many infants and children can be successfully treated with nighttime UCS and frequent high protein snacks during the day, but some will require intensive diets with monitoring of glucose and ketones, similar to individuals with GSD III, VI or IX. Because so little is known of the natural history of the severe forms of IPKH, careful review of metabolic control should be undertaken every few years to prevent under treatment as the patient grows but also to prevent over treatment since the natural ability to fast increases with advancing age.

 

CONCLUSIONS

 

Hypoglycemia in pediatric patients occurs predominately in the newborn period and during times of intercurrent illness, because it is most commonly caused by inherited hormonal or metabolic diseases. Rarer acquired forms of hypoglycemia must be suspected when initial presentation occurs in older children and adolescents. Because of the overlap of the normal transitional changes of glucose regulation in the newborn period with the most common time for the presentation of inherited hypoglycemic conditions, it is critical to screen for hypoglycemia and to determine the precise etiology so that rapid and appropriate interventions can be implemented. Newborn infants cannot be simply labeled as having hypoglycemia and discharged casually on frequent feeds. At the minimum a safety fast should be performed to ensure that the transient forms of hypoglycemia have truly resolved before discharge. Up to 10% of patients presenting with unexplained hypoglycemia in a pediatric emergency room setting will have a serious underlying metabolic or hormonal condition requiring long term care. It is critical that emergency rooms caring for children have protocols in place to identify these 10% of children, because these children are at risk of hypoglycemic brain damage. At all ages rapid intervention can prevent permanent neurological injury caused by the majority of conditions we discuss. 

 

REFERENCES

 

  1. Davis HA, Spanakis EK, Cryer PE, Davis SN. Hypoglycemia During Therapy of Diabetes. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Shah AS, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA)2000.
  2. Stanley CA, Rozance PJ, Thornton PS, De Leon DD, Harris D, Haymond MW, Hussain K, Levitsky LL, Murad MH, Simmons RA, Sperling MA, Weinstein DA, White NH, Wolfsdorf JI. Re-evaluating "transitional neonatal hypoglycemia": mechanism and implications for management. J Pediatr. 2015;166(6):1520-1525 e1521.
  3. Calabria AC, Gallagher PR, Simmons R, Blinman T, De Leon DD. Postoperative surveillance and detection of postprandial hypoglycemia after fundoplasty in children. J Pediatr. 2011;159(4):597-601 e591.
  4. Vukovic R, Milenkovic T, Djordjevic M, Mitrovic K, Todorovic S, Sarajlija A, Hussain K. Postprandial hyperinsulinemic hypoglycemia in a child as a late complication of esophageal reconstruction. J Pediatr Endocrinol Metab. 2017;30(7):791-795.
  5. Kelly A, Ng D, Ferry RJ, Jr., Grimberg A, Koo-McCoy S, Thornton PS, Stanley CA. Acute insulin responses to leucine in children with the hyperinsulinism/hyperammonemia syndrome. J Clin Endocrinol Metab. 2001;86(8):3724-3728.
  6. De Leon DD, Li C, Delson MI, Matschinsky FM, Stanley CA, Stoffers DA. Exendin-(9-39) corrects fasting hypoglycemia in SUR-1-/- mice by lowering cAMP in pancreatic beta-cells and inhibiting insulin secretion. J Biol Chem. 2008;283(38):25786-25793.
  7. Pagliara AS, Karl IE, Keating JP, Brown BI, Kipnis DM. Hepatic fructose-1,6-diphosphatase deficiency. A cause of lactic acidosis and hypoglycemia in infancy. J Clin Invest. 1972;51(8):2115-2123.
  8. Schurr A. Lactate: the ultimate cerebral oxidative energy substrate? J Cereb Blood Flow Metab. 2006;26(1):142-152.
  9. Powers WJ, Rosenbaum JL, Dence CS, Markham J, Videen TO. Cerebral glucose transport and metabolism in preterm human infants. J Cereb Blood Flow Metab. 1998;18(6):632-638.
  10. Pryds O, Christensen NJ, Friis-Hansen B. Increased cerebral blood flow and plasma epinephrine in hypoglycemic, preterm neonates. Pediatrics. 1990;85(2):172-176.
  11. Owen OE, Morgan AP, Kemp HG, Sullivan JM, Herrera MG, Cahill GF, Jr. Brain metabolism during fasting. J Clin Invest. 1967;46(10):1589-1595.
  12. King P, Kong MF, Parkin H, MacDonald IA, Barber C, Tattersall RB. Intravenous lactate prevents cerebral dysfunction during hypoglycaemia in insulin-dependent diabetes mellitus. Clin Sci (Lond). 1998;94(2):157-163.
  13. Courchesne-Loyer A, Croteau E, Castellano CA, St-Pierre V, Hennebelle M, Cunnane SC. Inverse relationship between brain glucose and ketone metabolism in adults during short-term moderate dietary ketosis: A dual tracer quantitative positron emission tomography study. J Cereb Blood Flow Metab. 2017;37(7):2485-2493.
  14. Yager JY. Hypoglycemic injury to the immature brain. Clin Perinatol. 2002;29(4):651-674, vi.
  15. Ghajar JB, Plum F, Duffy TE. Cerebral oxidative metabolism and blood flow during acute hypoglycemia and recovery in unanesthetized rats. J Neurochem. 1982;38(2):397-409.
  16. Menni F, de Lonlay P, Sevin C, Touati G, Peigne C, Barbier V, Nihoul-Fekete C, Saudubray JM, Robert JJ. Neurologic outcomes of 90 neonates and infants with persistent hyperinsulinemic hypoglycemia. Pediatrics. 2001;107(3):476-479.
  17. Javorsky BR, Raff H, Carroll TB, Algeciras-Schimnich A, Singh RJ, Colon-Franco JM, Findling JW. New Cutoffs for the Biochemical Diagnosis of Adrenal Insufficiency after ACTH Stimulation using Specific Cortisol Assays. J Endocr Soc. 2021;5(4):bvab022.
  18. Kelly A, Tang R, Becker S, Stanley CA. Poor specificity of low growth hormone and cortisol levels during fasting hypoglycemia for the diagnoses of growth hormone deficiency and adrenal insufficiency. Pediatrics. 2008;122(3):e522-528.
  19. Hussain K, Hindmarsh P, Aynsley-Green A. Neonates with symptomatic hyperinsulinemic hypoglycemia generate inappropriately low serum cortisol counterregulatory hormonal responses. J Clin Endocrinol Metab. 2003;88(9):4342-4347.
  20. De Leon DD, Thornton P, Stanley CA, Sperling MA. Hypoglycemia in the Newborn and Infant. In: Majzoub JA, Menon RK, Stratakis CA, eds. Sperling Pediatric Endocrinology. Fifth ed: Elsevier; 2021.
  21. Thornton PS, Stanley CA, De Leon DD, Harris D, Haymond MW, Hussain K, Levitsky LL, Murad MH, Rozance PJ, Simmons RA, Sperling MA, Weinstein DA, White NH, Wolfsdorf JI, Pediatric Endocrine S. Recommendations from the Pediatric Endocrine Society for Evaluation and Management of Persistent Hypoglycemia in Neonates, Infants, and Children. J Pediatr. 2015;167(2):238-245.
  22. Cryer PE. Diverse causes of hypoglycemia-associated autonomic failure in diabetes. N Engl J Med. 2004;350(22):2272-2279.
  23. Yang J, Hammoud B, Li C, Ridler A, Yau D, Kim J, Won KJ, Stanley CA, Hoshi T, Stanescu DE. Decreased KATP Channel Activity Contributes to the Low Glucose Threshold for Insulin Secretion of Rat Neonatal Islets. Endocrinology. 2021;162(9).
  24. Harris DL, Weston PJ, Gamble GD, Harding JE. Glucose Profiles in Healthy Term Infants in the First 5 Days: The Glucose in Well Babies (GLOW) Study. J Pediatr. 2020;223:34-41 e34.
  25. Stanley CA, Thornton PS, De Leon DD. New approaches to screening and management of neonatal hypoglycemia based on improved understanding of the molecular mechanism of hypoglycemia. Front Pediatr. 2023;11:1071206.
  26. Harris DL, Weston PJ, Harding JE. Alternative Cerebral Fuels in the First Five Days in Healthy Term Infants: The Glucose in Well Babies (GLOW) Study. J Pediatr. 2021;231:81-86 e82.
  27. LeBlanc S, Haushalter J, Seashore C, Wood KS, Steiner MJ, Sutton AG. A Quality-Improvement Initiative to Reduce NICU Transfers for Neonates at Risk for Hypoglycemia. Pediatrics. 2018;141(3).
  28. Weston PJ, Harris DL, Battin M, Brown J, Hegarty JE, Harding JE. Oral dextrose gel for the treatment of hypoglycaemia in newborn infants. Cochrane Database Syst Rev. 2016(5):CD011027.
  29. Lilien LD, Pildes RS, Srinivasan G, Voora S, Yeh TF. Treatment of neonatal hypoglycemia with minibolus and intraveous glucose infusion. J Pediatr. 1980;97(2):295-298.
  30. Hoermann H, Mokwa A, Roeper M, Salimi Dafsari R, Koestner F, Hagenbeck C, Mayatepek E, Kummer S, Meissner T. Reliability and Observer Dependence of Signs of Neonatal Hypoglycemia. J Pediatr. 2022;245:22-29 e22.
  31. Stanley CA. Perspective on the Genetics and Diagnosis of Congenital Hyperinsulinism Disorders. J Clin Endocrinol Metab. 2016;101(3):815-826.
  32. De Leon DD, Arnoux JB, Banerjee I, Bergada I, Bhatti T, Conwell LS, Fu JF, Flanagan SE, Gillis D, Meissner T, Mohnike K, Pasquini TLS, Shah P, Stanley CA, Vella A, Yorifuji T, Thornton PS. International Guidelines for the Diagnosis and Management of Hyperinsulinism. Horm Res Paediatr. 2023.
  33. Ferrara C, Patel P, Becker S, Stanley CA, Kelly A. Biomarkers of Insulin for the Diagnosis of Hyperinsulinemic Hypoglycemia in Infants and Children. J Pediatr. 2016;168:212-219.
  34. Hawkes CP, Lado JJ, Givler S, De Leon DD. The Effect of Continuous Intravenous Glucagon on Glucose Requirements in Infants with Congenital Hyperinsulinism. JIMD Rep. 2019;45:45-50.
  35. Otonkoski T, Ammala C, Huopio H, Cote GJ, Chapman J, Cosgrove K, Ashfield R, Huang E, Komulainen J, Ashcroft FM, Dunne MJ, Kere J, Thomas PM. A point mutation inactivating the sulfonylurea receptor causes the severe form of persistent hyperinsulinemic hypoglycemia of infancy in Finland. Diabetes. 1999;48(2):408-415.
  36. Snider KE, Becker S, Boyajian L, Shyng SL, MacMullen C, Hughes N, Ganapathy K, Bhatti T, Stanley CA, Ganguly A. Genotype and phenotype correlations in 417 children with congenital hyperinsulinism. J Clin Endocrinol Metab. 2013;98(2):E355-363.
  37. Yau D, Stanley CA. Diazoxide-responsive forms of congenital hyperinsulinism. In: De Leon-Crutchlow DD, Stanley CA, eds. Congenital Hyperinsulinism: A Practical Guide to Diagnosis and Management. Switzerland: Humana Press; 2019.
  38. Herrera A, Vajravelu ME, Givler S, Mitteer L, Avitabile CM, Lord K, De Leon DD. Prevalence of Adverse Events in Children With Congenital Hyperinsulinism Treated With Diazoxide. J Clin Endocrinol Metab. 2018;103(12):4365-4372.
  39. Thornton P, Truong L, Reynolds C, Hamby T, Nedrelow J. Rate of Serious Adverse Events Associated with Diazoxide Treatment of Patients with Hyperinsulinism. Horm Res Paediatr. 2019;91(1):25-32.
  40. Brar PC, Heksch R, Cossen K, De Leon DD, Kamboj MK, Marks SD, Marshall BA, Miller R, Page L, Stanley T, Mitchell D, Thornton P. Management and Appropriate Use of Diazoxide in Infants and Children with Hyperinsulinism. J Clin Endocrinol Metab. 2020;105(12).
  41. Cuff H, Lord K, Ballester L, Scully T, Stewart N, De Leon DD. The Use of Lanreotide in the Treatment of Congenital Hyperinsulinism. J Clin Endocrinol Metab. 2022;107(8):e3115-e3120.
  42. De Cosio AP, Thornton P. Current and Emerging Agents for the Treatment of Hypoglycemia in Patients with Congenital Hyperinsulinism. Paediatr Drugs. 2019;21(3):123-136.
  43. Ryan F, Devaney D, Joyce C, Nestorowicz A, Permutt MA, Glaser B, Barton DE, Thornton PS. Hyperinsulinism: molecular aetiology of focal disease. Arch Dis Child. 1998;79(5):445-447.
  44. Boodhansingh KE, Yang Z, Li C, Chen P, Lord K, Becker SA, States LJ, Adzick NS, Bhatti T, Shyng SL, Ganguly A, Stanley CA, De Leon DD. Localized islet nuclear enlargement hyperinsulinism (LINE-HI) due to ABCC8 and GCK mosaic mutations. Eur J Endocrinol. 2022;187(2):301-313.
  45. Lord K, Dzata E, Snider KE, Gallagher PR, De Leon DD. Clinical presentation and management of children with diffuse and focal hyperinsulinism: a review of 223 cases. J Clin Endocrinol Metab. 2013;98(11):E1786-1789.
  46. States LJ, Saade-Lemus S, De Leon DD. 18-F-L 3,4-Dihydroxyphenylalanine PET/Computed Tomography in the Management of Congenital Hyperinsulinism. PET Clin. 2020;15(3):349-359.
  47. Garg PK, Lokitz SJ, Truong L, Putegnat B, Reynolds C, Rodriguez L, Nazih R, Nedrelow J, Guardia M, Uffman JK, Garg S, Thornton PS. Pancreatic uptake and radiation dosimetry of 6-[18F]fluoro-L-DOPA from PET imaging studies in infants with congenital hyperinsulinism. PLoS One. 2017;12(11):e0186340.
  48. Adzick NS, De Leon DD, States LJ, Lord K, Bhatti TR, Becker SA, Stanley CA. Surgical treatment of congenital hyperinsulinism: Results from 500 pancreatectomies in neonates and children. J Pediatr Surg. 2018.
  49. Beltrand J, Caquard M, Arnoux JB, Laborde K, Velho G, Verkarre V, Rahier J, Brunelle F, Nihoul-Fekete C, Saudubray JM, Robert JJ, de Lonlay P. Glucose metabolism in 105 children and adolescents after pancreatectomy for congenital hyperinsulinism. Diabetes Care. 2012;35(2):198-203.
  50. Grimberg A, Ferry RJ, Jr., Kelly A, Koo-McCoy S, Polonsky K, Glaser B, Permutt MA, Aguilar-Bryan L, Stafford D, Thornton PS, Baker L, Stanley CA. Dysregulation of insulin secretion in children with congenital hyperinsulinism due to sulfonylurea receptor mutations. Diabetes. 2001;50(2):322-328.
  51. Huopio H, Reimann F, Ashfield R, Komulainen J, Lenko HL, Rahier J, Vauhkonen I, Kere J, Laakso M, Ashcroft F, Otonkoski T. Dominantly inherited hyperinsulinism caused by a mutation in the sulfonylurea receptor type 1. J Clin Invest. 2000;106(7):897-906.
  52. Byrne MM, Sturis J, Fajans SS, Ortiz FJ, Stoltz A, Stoffel M, Smith MJ, Bell GI, Halter JB, Polonsky KS. Altered insulin secretory responses to glucose in subjects with a mutation in the MODY1 gene on chromosome 20. Diabetes. 1995;44(6):699-704.
  53. Byrne MM, Sturis J, Menzel S, Yamagata K, Fajans SS, Dronsfield MJ, Bain SC, Hattersley AT, Velho G, Froguel P, Bell GI, Polonsky KS. Altered insulin secretory responses to glucose in diabetic and nondiabetic subjects with mutations in the diabetes susceptibility gene MODY3 on chromosome 12. Diabetes. 1996;45(11):1503-1510.
  54. Cardenas-Diaz FL, Osorio-Quintero C, Diaz-Miranda MA, Kishore S, Leavens K, Jobaliya C, Stanescu D, Ortiz-Gonzalez X, Yoon C, Chen CS, Haliyur R, Brissova M, Powers AC, French DL, Gadue P. Modeling Monogenic Diabetes using Human ESCs Reveals Developmental and Metabolic Deficiencies Caused by Mutations in HNF1A. Cell Stem Cell. 2019;25(2):273-289 e275.
  55. Miki T, Tashiro F, Iwanaga T, Nagashima K, Yoshitomi H, Aihara H, Nitta Y, Gonoi T, Inagaki N, Miyazaki J, Seino S. Abnormalities of pancreatic islets by targeted expression of a dominant-negative KATP channel. Proc Natl Acad Sci U S A. 1997;94(22):11969-11973.
  56. Wakeling MN, Owens NDL, Hopkinson JR, Johnson MB, Houghton JAL, Dastamani A, Flaxman CS, Wyatt RC, Hewat TI, Hopkins JJ, Laver TW, van Heugten R, Weedon MN, De Franco E, Patel KA, Ellard S, Morgan NG, Cheesman E, Banerjee I, Hattersley AT, Dunne MJ, International Congenital Hyperinsulinism C, Richardson SJ, Flanagan SE. Non-coding variants disrupting a tissue-specific regulatory element in HK1 cause congenital hyperinsulinism. Nat Genet. 2022;54(11):1615-1620.
  57. Pinney SE, Ganapathy K, Bradfield J, Stokes D, Sasson A, Mackiewicz K, Boodhansingh K, Hughes N, Becker S, Givler S, Macmullen C, Monos D, Ganguly A, Hakonarson H, Stanley CA. Dominant form of congenital hyperinsulinism maps to HK1 region on 10q. Horm Res Paediatr. 2013;80(1):18-27.
  58. Stanley CA, Lieu YK, Hsu BY, Burlina AB, Greenberg CR, Hopwood NJ, Perlman K, Rich BH, Zammarchi E, Poncz M. Hyperinsulinism and hyperammonemia in infants with regulatory mutations of the glutamate dehydrogenase gene. N Engl J Med. 1998;338(19):1352-1357.
  59. Bahi-Buisson N, Roze E, Dionisi C, Escande F, Valayannopoulos V, Feillet F, Heinrichs C, Chadefaux-Vekemans B, Dan B, de Lonlay P. Neurological aspects of hyperinsulinism-hyperammonaemia syndrome. Dev Med Child Neurol. 2008;50(12):945-949.
  60. Su C, Liang XJ, Li WJ, Wu D, Liu M, Cao BY, Chen JJ, Qin M, Meng X, Gong CX. Clinical and Molecular Spectrum of Glutamate Dehydrogenase Gene Defects in 26 Chinese Congenital Hyperinsulinemia Patients. J Diabetes Res. 2018;2018:2802540.
  61. MacMullen C, Fang J, Hsu BY, Kelly A, de Lonlay-Debeney P, Saudubray JM, Ganguly A, Smith TJ, Stanley CA, Hyperinsulinism/hyperammonemia Contributing I. Hyperinsulinism/hyperammonemia syndrome in children with regulatory mutations in the inhibitory guanosine triphosphate-binding domain of glutamate dehydrogenase. J Clin Endocrinol Metab. 2001;86(4):1782-1787.
  62. Raizen DM, Brooks-Kayal A, Steinkrauss L, Tennekoon GI, Stanley CA, Kelly A. Central nervous system hyperexcitability associated with glutamate dehydrogenase gain of function mutations. J Pediatr. 2005;146(3):388-394.
  63. Treberg JR, Clow KA, Greene KA, Brosnan ME, Brosnan JT. Systemic activation of glutamate dehydrogenase increases renal ammoniagenesis: implications for the hyperinsulinism/hyperammonemia syndrome. Am J Physiol Endocrinol Metab. 2010;298(6):E1219-1225.
  64. De Lonlay P, Benelli C, Fouque F, Ganguly A, Aral B, Dionisi-Vici C, Touati G, Heinrichs C, Rabier D, Kamoun P, Robert JJ, Stanley C, Saudubray JM. Hyperinsulinism and hyperammonemia syndrome: report of twelve unrelated patients. Pediatr Res. 2001;50(3):353-357.
  65. Clayton PT, Eaton S, Aynsley-Green A, Edginton M, Hussain K, Krywawych S, Datta V, Malingre HE, Berger R, van den Berg IE. Hyperinsulinism in short-chain L-3-hydroxyacyl-CoA dehydrogenase deficiency reveals the importance of beta-oxidation in insulin secretion. J Clin Invest. 2001;108(3):457-465.
  66. Stanescu DE, Hughes N, Kaplan B, Stanley CA, De Leon DD. Novel presentations of congenital hyperinsulinism due to mutations in the MODY genes: HNF1A and HNF4A. J Clin Endocrinol Metab. 2012;97(10):E2026-2030.
  67. McGlacken-Byrne SM, Mohammad JK, Conlon N, Gubaeva D, Siersbaek J, Schou AJ, Demirbilek H, Dastamani A, Houghton JAL, Brusgaard K, Melikyan M, Christesen H, Flanagan SE, Murphy NP, Shah P. Clinical and genetic heterogeneity of HNF4A/HNF1A mutations in a multicentre paediatric cohort with hyperinsulinaemic hypoglycaemia. Eur J Endocrinol. 2022;186(4):417-427.
  68. Meissner T, Otonkoski T, Feneberg R, Beinbrech B, Apostolidou S, Sipila I, Schaefer F, Mayatepek E. Exercise induced hypoglycaemic hyperinsulinism. Arch Dis Child. 2001;84(3):254-257.
  69. Otonkoski T, Jiao H, Kaminen-Ahola N, Tapia-Paez I, Ullah MS, Parton LE, Schuit F, Quintens R, Sipila I, Mayatepek E, Meissner T, Halestrap AP, Rutter GA, Kere J. Physical exercise-induced hypoglycemia caused by failed silencing of monocarboxylate transporter 1 in pancreatic beta cells. Am J Hum Genet. 2007;81(3):467-474.
  70. Gonzalez-Barroso MM, Giurgea I, Bouillaud F, Anedda A, Bellanne-Chantelot C, Hubert L, de Keyzer Y, de Lonlay P, Ricquier D. Mutations in UCP2 in congenital hyperinsulinism reveal a role for regulation of insulin secretion. PLoS One. 2008;3(12):e3850.
  71. Flanagan SE, Vairo F, Johnson MB, Caswell R, Laver TW, Lango Allen H, Hussain K, Ellard S. A CACNA1D mutation in a patient with persistent hyperinsulinaemic hypoglycaemia, heart defects, and severe hypotonia. Pediatr Diabetes. 2017;18(4):320-323.
  72. Kostopoulou E, Dastamani A, Guemes M, Clement E, Caiulo S, Shanmugananda P, Dattani M, Gilbert C, Hurst JA, Shah P. Syndromic Forms of Hyperinsulinaemic Hypoglycaemia-A 15-year follow-up Study. Clin Endocrinol (Oxf). 2021;94(3):399-412.
  73. Duffy KA, Cielo CM, Cohen JL, Gonzalez-Gandolfi CX, Griff JR, Hathaway ER, Kupa J, Taylor JA, Wang KH, Ganguly A, Deardorff MA, Kalish JM. Characterization of the Beckwith-Wiedemann spectrum: Diagnosis and management. Am J Med Genet C Semin Med Genet. 2019;181(4):693-708.
  74. Kalish JM, Boodhansingh KE, Bhatti TR, Ganguly A, Conlin LK, Becker SA, Givler S, Mighion L, Palladino AA, Adzick NS, De Leon DD, Stanley CA, Deardorff MA. Congenital hyperinsulinism in children with paternal 11p uniparental isodisomy and Beckwith-Wiedemann syndrome. J Med Genet. 2016;53(1):53-61.
  75. Brioude F, Kalish JM, Mussa A, Foster AC, Bliek J, Ferrero GB, Boonen SE, Cole T, Baker R, Bertoletti M, Cocchi G, Coze C, De Pellegrin M, Hussain K, Ibrahim A, Kilby MD, Krajewska-Walasek M, Kratz CP, Ladusans EJ, Lapunzina P, Le Bouc Y, Maas SM, Macdonald F, Ounap K, Peruzzi L, Rossignol S, Russo S, Shipster C, Skorka A, Tatton-Brown K, Tenorio J, Tortora C, Gronskov K, Netchine I, Hennekam RC, Prawitt D, Tumer Z, Eggermann T, Mackay DJG, Riccio A, Maher ER. Expert consensus document: Clinical and molecular diagnosis, screening and management of Beckwith-Wiedemann syndrome: an international consensus statement. Nat Rev Endocrinol. 2018;14(4):229-249.
  76. Laje P, Palladino AA, Bhatti TR, States LJ, Stanley CA, Adzick NS. Pancreatic surgery in infants with Beckwith-Wiedemann syndrome and hyperinsulinism. J Pediatr Surg. 2013;48(12):2511-2516.
  77. Hussain K, Cosgrove KE, Shepherd RM, Luharia A, Smith VV, Kassem S, Gregory JW, Sivaprasadarao A, Christesen HT, Jacobsen BB, Brusgaard K, Glaser B, Maher EA, Lindley KJ, Hindmarsh P, Dattani M, Dunne MJ. Hyperinsulinemic hypoglycemia in Beckwith-Wiedemann syndrome due to defects in the function of pancreatic beta-cell adenosine triphosphate-sensitive potassium channels. J Clin Endocrinol Metab. 2005;90(7):4376-4382.
  78. Yap KL, Johnson AEK, Fischer D, Kandikatla P, Deml J, Nelakuditi V, Halbach S, Jeha GS, Burrage LC, Bodamer O, Benavides VC, Lewis AM, Ellard S, Shah P, Cody D, Diaz A, Devarajan A, Truong L, Greeley SAW, De Leo-Crutchlow DD, Edmondson AC, Das S, Thornton P, Waggoner D, Del Gaudio D. Congenital hyperinsulinism as the presenting feature of Kabuki syndrome: clinical and molecular characterization of 10 affected individuals. Genet Med. 2018.
  79. Gibson CE, Boodhansingh KE, Li C, Conlin L, Chen P, Becker SA, Bhatti T, Bamba V, Adzick NS, De Leon DD, Ganguly A, Stanley CA. Congenital Hyperinsulinism in Infants with Turner Syndrome: Possible Association with Monosomy X and KDM6A Haploinsufficiency. Horm Res Paediatr. 2018;89(6):413-422.
  80. Vajravelu ME, Chai J, Krock B, Baker S, Langdon D, Alter C, De Leon DD. Congenital Hyperinsulinism and Hypopituitarism Attributable to a Mutation in FOXA2. J Clin Endocrinol Metab. 2018;103(3):1042-1047.
  81. Giri D, Vignola ML, Gualtieri A, Scagliotti V, McNamara P, Peak M, Didi M, Gaston-Massuet C, Senniappan S. Novel FOXA2 mutation causes Hyperinsulinism, Hypopituitarism with Craniofacial and Endoderm-derived organ abnormalities. Hum Mol Genet. 2017;26(22):4315-4326.
  82. Moravej H, Altassan R, Jaeken J, Enns GM, Ellaway C, Balasubramaniam S, De Lonlay P, Coman D, Mercimek-Andrews S, Witters P, Morava E. Hypoglycemia in CDG patients due to PMM2 mutations: Follow up on hyperinsulinemic patients. JIMD Rep. 2020;51(1):76-81.
  83. de Lonlay P, Cuer M, Vuillaumier-Barrot S, Beaune G, Castelnau P, Kretz M, Durand G, Saudubray JM, Seta N. Hyperinsulinemic hypoglycemia as a presenting sign in phosphomannose isomerase deficiency: A new manifestation of carbohydrate-deficient glycoprotein syndrome treatable with mannose. J Pediatr. 1999;135(3):379-383.
  84. Sturiale L, Barone R, Garozzo D. The impact of mass spectrometry in the diagnosis of congenital disorders of glycosylation. J Inherit Metab Dis. 2011;34(4):891-899.
  85. Stanescu DL, Stanley CA. Advances in Understanding the Mechanism of Transitional Neonatal Hypoglycemia and Implications for Management. Clin Perinatol. 2022;49(1):55-72.
  86. Arya VB, Flanagan SE, Schober E, Rami-Merhar B, Ellard S, Hussain K. Activating AKT2 mutation: hypoinsulinemic hypoketotic hypoglycemia. J Clin Endocrinol Metab. 2014;99(2):391-394.
  87. Bhatti TR, Ganapathy K, Huppmann AR, Conlin L, Boodhansingh KE, MacMullen C, Becker S, Ernst LM, Adzick NS, Ruchelli ED, Ganguly A, Stanley CA. Histologic and Molecular Profile of Pediatric Insulinomas: Evidence of a Paternal Parent-of-Origin Effect. J Clin Endocrinol Metab. 2016;101(3):914-922.
  88. Gupta P, Loechner K, Patterson BC, Felner E. Insulinoma in an adolescent female with weight loss: a case report and literature review on pediatric insulinomas. Endocrinol Diabetes Metab Case Rep. 2022;2022.
  89. Thakker RV, Newey PJ, Walls GV, Bilezikian J, Dralle H, Ebeling PR, Melmed S, Sakurai A, Tonelli F, Brandi ML, Endocrine S. Clinical practice guidelines for multiple endocrine neoplasia type 1 (MEN1). J Clin Endocrinol Metab. 2012;97(9):2990-3011.
  90. Mitrakou A, Fanelli C, Veneman T, Perriello G, Calderone S, Platanisiotis D, Rambotti A, Raptis S, Brunetti P, Cryer P, et al. Reversibility of unawareness of hypoglycemia in patients with insulinomas. N Engl J Med. 1993;329(12):834-839.
  91. Service FJ, Dale AJ, Elveback LR, Jiang NS. Insulinoma: clinical and diagnostic features of 60 consecutive cases. Mayo Clin Proc. 1976;51(7):417-429.
  92. Shah R, Garg R, Majmundar M, Purandare N, Malhotra G, Patil V, Ramteke-Jadhav S, Lila A, Shah N, Bandgar T. Exendin-4-based imaging in insulinoma localization: Systematic review and meta-analysis. Clin Endocrinol (Oxf). 2021;95(2):354-364.
  93. de Herder WW, Hofland J. Insulinoma. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Shah AS, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA)2000.
  94. Palladino AA, Sayed S, Levitt Katz LE, Gallagher PR, De Leon DD. Increased glucagon-like peptide-1 secretion and postprandial hypoglycemia in children after Nissen fundoplication. J Clin Endocrinol Metab. 2009;94(1):39-44.
  95. Rosenfeld E, De Leon DD. Bridging the gaps: recent advances in diagnosis, care, and outcomes in congenital hyperinsulinism. Curr Opin Pediatr. 2023.
  96. Egan AM, Galior KD, Maus AD, Fatica E, Simha V, Shah P, Singh RJ, Vella A. Pitfalls in Diagnosing Hypoglycemia Due to Exogenous Insulin: Validation and Utility of an Insulin Analog Assay. Mayo Clin Proc. 2022;97(11):1994-2004.
  97. Folick A, Cheng C, Chu SN, Rick JW, Rushakoff RJ. Discordant Timing of Hypoglycemic Agent Screening Causing Delayed Diagnosis of Sulfonylurea-Induced Hypoglycemia. AACE Clin Case Rep. 2022;8(3):139-141.
  98. Cullingford DJ, Siafarikas A, Choong CS. Genetic Etiology of Congenital Hypopituitarism. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Shah AS, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA)2000.
  99. Mehta A, Hindmarsh PC, Stanhope RG, Turton JP, Cole TJ, Preece MA, Dattani MT. The role of growth hormone in determining birth size and early postnatal growth, using congenital growth hormone deficiency (GHD) as a model. Clin Endocrinol (Oxf). 2005;63(2):223-231.
  100. Murray PG, Clayton PE. Disorders of Growth Hormone in Childhood. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Shah AS, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA)2000.
  101. Yau M, Khattab A, Yuen T, New M. Congenital Adrenal Hyperplasia. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Shah AS, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA)2000.
  102. Aynsley-Green A, Williamson DH, Gitzelmann R. Hepatic glycogen synthetase deficiency. Definition of syndrome from metabolic and enzyme studies on a 9-year-old girl. Arch Dis Child. 1977;52(7):573-579.
  103. Weinstein DA, Correia CE, Saunders AC, Wolfsdorf JI. Hepatic glycogen synthase deficiency: an infrequently recognized cause of ketotic hypoglycemia. Mol Genet Metab. 2006;87(4):284-288.
  104. Burchell A. The molecular basis of the type 1 glycogen storage diseases. Bioessays. 1992;14(6):395-400.
  105. Bao Y, Dawson TL, Jr., Chen YT. Human glycogen debranching enzyme gene (AGL): complete structural organization and characterization of the 5' flanking region. Genomics. 1996;38(2):155-165.
  106. Slonim AE, Coleman RA, Moses WS. Myopathy and growth failure in debrancher enzyme deficiency: improvement with high-protein nocturnal enteral therapy. J Pediatr. 1984;105(6):906-911.
  107. Brown LM, Corrado MM, van der Ende RM, Derks TG, Chen MA, Siegel S, Hoyt K, Correia CE, Lumpkin C, Flanagan TB, Carreras CT, Weinstein DA. Evaluation of glycogen storage disease as a cause of ketotic hypoglycemia in children. J Inherit Metab Dis. 2015;38(3):489-493.
  108. Cohen P, Spiegelman BM. Cell biology of fat storage. Mol Biol Cell. 2016;27(16):2523-2527.
  109. Lindner M, Hoffmann GF, Matern D. Newborn screening for disorders of fatty-acid oxidation: experience and recommendations from an expert meeting. J Inherit Metab Dis. 2010;33(5):521-526.
  110. White K, Truong L, Aaron K, Mushtaq N, Thornton PS. The Incidence and Etiology of Previously Undiagnosed Hypoglycemic Disorders in the Emergency Department. Pediatr Emerg Care. 2018.
  111. Weinstein DA, Butte AJ, Raymond K, Korson MS, Weiner DL, Wolfsdorf JI. High Incidence of Unrecognized Metabolic and Endocrinologic Disorders in Acutely Ill Children with Previously Unrecognized Hypoglycemia. Pediatr Res. 2001;49:103.
  112. Halestrap AP. The SLC16 gene family - structure, role and regulation in health and disease. Mol Aspects Med. 2013;34(2-3):337-349.
  113. Al-Khawaga S, AlRayahi J, Khan F, Saraswathi S, Hasnah R, Haris B, Mohammed I, Abdelalim EM, Hussain K. A SLC16A1 Mutation in an Infant With Ketoacidosis and Neuroimaging Assessment: Expanding the Clinical Spectrum of MCT1 Deficiency. Front Pediatr. 2019;7:299.
  114. Rosenfeld E, Alzahrani O, De Leon DD. Undiagnosed hypoglycaemia disorders in children detected when hypoglycaemia occurs in the setting of illness: a retrospective study. BMJ Paediatr Open. 2023;7(1).
  115. van Veen MR, van Hasselt PM, de Sain-van der Velden MG, Verhoeven N, Hofstede FC, de Koning TJ, Visser G. Metabolic profiles in children during fasting. Pediatrics. 2011;127(4):e1021-1027.
  116. Parmar K, Mosha M, Weinstein DA, Riba-Wolman R. Fasting ketone levels vary by age: implications for differentiating physiologic from pathologic ketotic hypoglycemia. J Pediatr Endocrinol Metab. 2023;36(7):667-673.

Chronic Fatigue Syndrome

ABSTRACT

Myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) is an enigmatic medical condition that has growing prevalence across the globe, often diagnosed after exclusion of other medical or mental illnesses. As there is no clinical test to confirm the presence of this condition, the diagnosis is syndromic based on different clinical definitions. There was mixed evidence to support the use of a specific therapy that provides palliative effect. Pathophysiological hypotheses can be categorized into infection, immune, mitochondrial, neurobehavioral, or stress system (HPA axis and sympathetic nervous system) disorders. The prognosis of ME/CFS is mixed but recovery does occur in many cases, over time.  All-cause mortality rate is not increased.

CLINICAL DEFINITION

Fatigue is a term used to describe unexplained subjective, chronic, pervasive tiredness or weakness physically, mentally, or a combination of both. The term “myalgic encephalomyelitis” was first described in the United Kingdom after an outbreak of serious infection at the Royal Free Hospital in 1955 (1). The US originated term Chronic Fatigue Syndrome (CFS) was introduced by Holmes et al in 1988 (2). Several definitions of CFS have been developed, primarily to standardize research (3,4). The key symptoms expected in this condition was later refined in 1994 and named after Dr Fukada (3). However, it was particularly challenging to reach a consensus on a name for this condition as its etiology and pathology are unexplained. An important milestone was achieved on October 1, 2022 with the update to International Coding Disease (ICD-10-CM) that include a specific diagnostic code for myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS), chronic fatigue syndrome (CFS) and myalgic encephalomyelitis (ME) (5). Prior to this, chronic fatigue syndrome was categorized in the “chronic fatigue, unspecified”, which could limit epidemiologic studies.

 

The 1994 US Centers for Disease Control and Prevention (CDC) Fukuda criteria for chronic fatigue syndrome comprise the following (3):

 

  1. Primary symptoms that are clinically evaluated, unexplained, persistent or relapsing fatigue, lasting at least 6 months. The fatigue is not the result of ongoing physical exertion, and resting, sleeping, or downgrading activity is non-restorative. The fatigue causes significant impairment in personal, social, and/or occupational domains and represents a substantial reduction in premorbid levels of activity and functional capacity.
  2. The concurrent presence of at least 4 of the 8 following symptoms over a 6-month period:
  • Impaired short-term memory or concentration.
  • sore throat.
  • tender lymph nodes/glands.
  •  
  • multiple-joint pain without swelling or redness.
  • headache of new type, pattern, or severity.
  • unrefreshing sleep.
  • post-exertional fatigue/malaise lasting longer than 24 hours.

 

The 2003 Canadian ME/CFS Case Criteria (CCC) specifies (4):

  • Post-exertional malaise must occur with rapid muscle or cognitive fatigability, taking 24 hours or longer to recover.
  • Unrefreshing sleep, myalgia, and arthralgia must be reported.
  • Two or more neurological/cognitive manifestations must be present.
  • At least one of autonomic, neuroendocrine, immune manifestations must be present.

 

This is a stricter criterion, compared to the Fukada Criteria and it is mainly used as case definition in research. Adults are diagnosed after 6 months of symptoms while pediatric cases were diagnosed after 3 months.

 

Nearly two decades after Fukada Criteria was introduced, the US Institute of Medicine (IOM), now known as National Academy of Medicine (NAM) proposed new diagnostic criteria in 2015 for chronic fatigue syndrome (CFS)/myalgic encephalomyelitis (ME) (5). These clinical diagnostic criteria followed a comprehensive analysis of the literature and expert consultation as below.

  1. Substantial reduction/impairment in the ability to engage in pre-illness levels of occupational, educational, social, or personal activities that persists for more than 6 months, is accompanied by fatigue that is often profound, is of new or definite onset, is not the result of ongoing excessive exertion, and is not substantially alleviated by rest.
  2. Post-exertional malaise (PEM).
  3. Unrefreshing sleep.
  4. In addition, patients are required to have at least one of the following two symptoms:
  • Cognitive impairment.
  • Orthostatic intolerance.

 

Symptoms must be present at least half of the time and have moderate, substantial, or severe intensity.

 

As a large group of patients remain stigmatized with the term ‘chronic fatigue syndrome’ (CFS), renaming the condition to 'systemic exertion intolerance disease' (SEID) was recommended to overcome the old stereotypes as CFS is more associated to a mental disorder rather than an organic illness (5). SEID highlights the somewhat unique feature of exertion intolerance, and consequent impaired functional capacity. SEID criteria may help with the treatment by increased diagnosis and awareness, calling attention to the major disabling symptoms, and by validating the major symptoms as real and debilitating. However, the new IOM criteria could increase the prevalence rate of this condition compared to the use of previous Fukada criteria due to the lack of specifying exclusionary illnesses (5).

DIAGNOSTIC APPROACH

The clinical diagnosis of CFS/ME is based on a constellation of symptoms where post-exertional malaise and fatigue are prominent; these are described in some definitions (Table 1) with an algorithm provided in Figure 1 (6). A thorough clinical assessment is necessary to exclude alternative medical and psychiatric diagnoses requiring specific treatment. For example, it is important to differentiate fatigue from weakness, which suggests a neuromuscular disease, and anhedonia from major depression. Hypersomnolence and sleep disorder suggests a need to exclude obstructive sleep apnea, particularly in groups at risk such as the obese.

 

Limited laboratory screening investigations are directed towards the discovery of subtle medical disorders. Unfortunately, there was no test with adequate sensitivity and specificity to verify the diagnosis of CFS/ME. The protean manifestations of CFS/ME suggest diverse causes, hence it is unlikely a single diagnostic test for CFS/ME will be developed. Routine laboratory investigations include a complete blood examination, erythrocyte sedimentation rate (ESR), calcium, phosphate, magnesium, blood glucose, serum electrolytes, thyroid stimulating hormone and free thyroxine levels, protein electrophoresis screen, C-reactive protein (CRP), ferritin, creatinine, rheumatoid factor, antinuclear antibody, creatine kinase and liver function, and routine urinalysis. Any other investigations should be carefully chosen on an individual basis depending on the clinical assessment and risk factors for other conditions. For example, sleep study may be considered in patients who have features of obstructive sleep apnea, while a morning cortisol concentration or a more definitive ACTH stimulation test may be considered for patients who have clinical features suggestive of adrenal insufficiency.

 

Although patients with CFS/ME tend to have more abnormalities on magnetic resonance imaging (MRI) and single-photon emission computed tomography (SPECT), the significance of these findings are unclear, hence routine neuroimaging is not recommended in the diagnostic process (7,8).

 

Some recent studies have suggested reduced circulatory and myocardial function in CFS, although the utility of routine cardiac assessment is not established (9,10).

 

Table 1. Clinical Working Case Definition of ME/CFS, published in 2000 (3,4)

A patient with ME/CFS will meet the criteria for fatigue, post-exertional malaise and/or fatigue, sleep dysfunction, and pain; have two or more neurological/cognitive manifestations and one or more symptoms from two of the categories of autonomic, neuroendocrine and immune manifestations; and adhere to item.

1. Fatigue: The patient must have a significant degree of new onset, unexplained, persistent, or recurrent physical and mental fatigue that substantially reduces activity level.

2. Post-Exertional Malaise and/or Fatigue: There is an inappropriate loss of physical and mental stamina, rapid muscular and cognitive fatigability, post exertional malaise and/or fatigue and/or pain and a tendency for other associated symptoms within the patient’s cluster of symptoms to worsen. There is a pathologically slow recovery period – usually 24 hours or longer.

3. Sleep Dysfunction:* There is unrefreshed sleep or sleep quantity or rhythm disturbances such as reversed or chaotic diurnal sleep rhythms.

4. Pain:* There is a significant degree of myalgia. Pain can be experienced in the muscles and/or joints, and is often widespread and migratory in nature. Often there are significant headaches of new type, pattern or severity.

5. Neurological/Cognitive Manifestations: Two or more of the following difficulties should be present: confusion, impairment of concentration and short-term memory consolidation, disorientation, difficulty with information processing, categorizing and word retrieval, and perceptual and sensory disturbances – e.g., spatial instability and disorientation and inability to focus vision. Ataxia, muscle weakness and fasciculations are common. There may be overload phenomena: cognitive, sensory – e.g., photophobia and hypersensitivity to noise – and/or emotional overload, which may lead to “crash” periods and/or anxiety.

6. At least one symptom from two of the following categories: (i) Autonomic Manifestations: orthostatic intolerance – neurally mediated hypotension (NMH), postural orthostatic tachycardia syndrome (POTS), delayed postural hypotension; lightheadedness; extreme pallor; nausea and irritable bowel syndrome; urinary frequency and bladder dysfunction; palpitations with or without cardiac arrhythmias; exertional dyspnea. (ii) Neuroendocrine Manifestations: loss of thermostatic stability – subnormal body temperature and marked diurnal fluctuation, sweating episodes, recurrent feelings of feverishness and cold extremities; intolerance of extremes of heat and cold; marked weight change – anorexia or abnormal appetite; loss of adaptability and worsening of symptoms with stress. (iii) Immune Manifestations: tender lymph nodes, recurrent sore throat, recurrent flu-like symptoms, general malaise, new sensitivities to food, medications and/or chemicals.

7. The illness persists for at least six months. It usually has a distinct onset, ** although it may be gradual. Preliminary diagnosis may be possible earlier. Three months is appropriate for children.

To be included, the symptoms must have begun or have been significantly altered after the onset of this illness. It is unlikely that a patient will suffer from all symptoms in criteria 5 and 6. The disturbances tend to form symptom clusters that may fluctuate and change over time. Children often have numerous prominent symptoms but their order of severity tends to vary from day to day. *There is a small number of patients who have no pain or sleep dysfunction, but no other diagnosis fits except ME/CFS. A diagnosis of ME/CFS can be entertained when this group has an infectious illness type onset. **Some patients have been unhealthy for other reasons prior to the onset of ME/CFS and lack detectable triggers at onset and/or have more gradual or insidious onset.

Exclusions: Exclude active disease processes that explain most of the major symptoms of fatigue, sleep disturbance, pain, and cognitive dysfunction. It is essential to exclude certain diseases, which would be tragic to miss: Addison’s disease, Cushing’s syndrome, hypothyroidism, hyperthyroidism, iron deficiency, other treatable forms of anemia, iron overload syndrome, diabetes mellitus, and cancer. It is also essential to exclude treatable sleep disorders such as upper airway resistance syndrome and obstructive or central sleep apnea; rheumatological disorders such as rheumatoid arthritis, lupus, polymyositis and polymyalgia rheumatica; immune disorders such as AIDS; neurological disorders such as multiple sclerosis (MS), Parkinsonism, myasthenia gravis and B12 deficiency; infectious diseases such as tuberculosis, chronic hepatitis, Lyme disease, etc.; primary psychiatric disorders and substance abuse. Exclusion of other diagnoses, which cannot be reasonably excluded by the patient’s history and physical examination, is achieved by laboratory testing and imaging. if a potentially confounding medical condition is under control, then the diagnosis of cfs can be entertained if patients meet the criteria otherwise.

Co-Morbid Entities: Fibromyalgia Syndrome (FMS), Myofascial Pain Syndrome (MPS), Temporo- mandibular Joint Syndrome (TMJ), Irritable Bowel Syndrome (IBS), Interstitial Cystitis, Irritable Bladder Syndrome, Raynaud’s Phenomenon, Prolapsed Mitral Valve, Depression, Migraine, Allergies, Multiple Chemical Sensitivities (MCS), Hashimoto’s thyroiditis, Sicca Syndrome, etc. Such comorbid entities may occur in the setting of CFS. Others such as IBS may precede the development of CFS by many years, but then become associated with it. The same holds true for migraines and depression. Their association is thus looser than between the symptoms within the syndrome. CFS and FMS often closely connect and should be considered to be “overlap syndromes.”

Overload phenomena affect sensory modalities where the patient may be hypersensitive to light, sound, vibration, speed, odors, and/or mixed sensory modalities.

Figure 1. Diagnostic algorithm adapted from IOM (6).

EPIDEMIOLOGY

The frequency of CFS has been assessed in two large-scale US community-based studies and a prevalence of 0.23-0.42% has been suggested (11,12). Another study suggested the global prevalence of CFS ranges from 0.4% and 2.5% (13).

 

CFS is at least twice as common in women as in men, occurs more frequently in minority groups, and in those with lower levels of education and occupational status (11, 14). Geographic location has not been shown to influence the prevalence of CFS but more recent study showed the condition is more common in certain countries such as the UK, Australia, and the USA (12, 14). Twin studies suggest that genetic factors play an important role (16). Population studies also associate elevated premorbid stress and childhood trauma, especially if complicated by psychopathology, with an increased risk of CFS (17,18).

 

An Australian sociodemographic cross-sectional study of patients diagnosed with CFS by their primary care physician was conducted over 2 years (2013-2015) (19). Participants were classified according to Fukuda criteria and international consensus ME/ICC criteria. CFS was most prevalent between 45-55 years, with a peak onset between 25-35 years with a high proportion of females affected (78.6%). Patients were predominantly Caucasian and highly educated. Of a total of 535 patients, only 30% met the Fukuda criteria and 32% met both Fukuda and International consensus ME/ICC criteria. 15% did not meet the criteria and 23% had exclusionary conditions. There was higher proportion of participants who were obese or overweight, (41.3% and 43.3% respectively) and were unemployed or on a disability pension. The results of this study may not be representative of all CFS/ME patients in the general population due to sample recruitment bias.

PATHOPHYSIOLOGY OF CHRONIC FATIGUE SYNDROME

Viral/Immune Hypotheses

For many years CFS was suspected to arise from a persistent response to an infection. Abrupt onset of symptoms and the presence of post-infectious fatigue after infections suggest this theory. There were also reports of a high frequency of antibody titers to specific, but varying, infectious agents (20). Epstein-Barr virus, human herpes virus 6, group B Coxsackie virus, human T-cell lymphotrophic virus II, hepatitis C, enteroviruses, and retroviruses, have all been proposed as etiological agents of CFS (21). However, to date, there has been no consistent evidence that CFS results from a specific infection (22). Moreover, there is data to indicate that global increases in humoral immune responses are seen in chronic stress states and that neurohormonal changes may account for these and other immune aberrations (20,23).

 

Recent study has examined the characteristics of cell function and receptors in CFS patients (24). Participants between 20 and 65 years old were recruited, by using the Fukuda criteria. Patient were classified as moderate (mobile) or severely affected (housebound). Blood was collected from all participants between 8am and 11am, and sent for lytic protein analysis, cell activity analysis, respiratory burst analysis and natural killer cell receptors analysis. The study demonstrated that there was significant decrease in natural killer cell cytotoxic activity in CFS patients and there is correlation between low natural killer cells cytotoxic activity and severity of CFS illness. CFS patients have alterations in Natural Killer receptors, adhesion markers and receptors on CD4, and CD8.

 

A prospective population-based cohort of 42,558 atopic patients and 170,232 controls without atopy were recruited between 2005-2007, with follow up until 2011. These 2 groups were similar in sex and age distributions, with a mean age of 47 years. The overall incidence rate for CFS in the atopy cohort (1.37 per 1000 person-year) was higher than in the non-atopy cohort (0.87 per 1000 person-year (25).  This suggests that that atopy might increase the risk of CFS/SEID.

Mitochondrial Hypotheses

Since mitochondria provide cellular energy, hypotheses of impaired mitochondrial function have been suggested to underlie CFS. Early studies have shown some associations between mitochondrial proteins and CFS, but these require confirmation (26).

Neuropsychiatric Hypotheses

Chronic fatigue syndrome has been suspected to be a neuropsychiatric disorder, or a type of depression (28). Although depression is frequent in CFS, most patients do not exhibit the characteristic self-reproach or biological features of endogenous depression. The depression often seen in CFS appears to be reactive and associated with marked frustration. However, the symptoms of depression can overlap with those of CFS. Profound fatigue is more commonly reported amongst CFS patients, than those with depression (28). Cognitive-behavioral models of CFS emphasize the importance of the interactions between cognitive, behavioral and biological variables in attempting to explain the genesis and maintenance of CFS. It may be that while organic factors may precipitate CFS, cognitive-behavioral factors may perpetuate the illness (28). Specifically, when individuals resume normal activity levels following an acute illness, it is common to experience symptoms of physical deconditioning. If individuals attribute these symptoms to signs of ongoing disease rather than deconditioning, they may resort to rest and inactivity in an attempt to "cure" the symptoms. A cycle of avoidance and symptom experience develops, which can lead to loss of control, demoralization and possible depression and anxiety. These psychological states can further perpetuate the illness through generating more symptoms.

 

The cognitive-behavioral model has been expanded to include personality as predisposing factors (29). This model proposes that predisposed people are highly achievement orientated perfectionists and base their self-esteem and the respect from others on their ability to live up to certain high standards (29). When such people are faced with factors that affect their ability to perform, such as a combination of excessive stress and an acute illness, their initial reaction is to persist and to attempt to maintain usual coping strategies. This behavior leads to exhaustion. In making sense of the situation a physical attribution for the exhaustion is made, which protects an individual's self-esteem by avoiding the suggestion that their inability to cope is a sign of personal weakness. The bias may lead to a focus on somatic rather than emotional aspects of the illness, and favors physical rather than psychological explanation. However, this model remains to be fully evaluated and it is poorly integrated with physiological aspects of CFS. There have been few systematic studies undertaken on the relationship between personality and CFS (28). However, a personality trait characterized by "perfectionism, high standards for work performance, responsibility and personal conduct and marked achievement orientation" was reported in interviews with individuals with CFS (30). Interviewees referred to a desire for accomplishment and success, aiming to achieve perfection. These desires were associated with the belief that “failure to meet these standards would indicate failure as a person, or unacceptability to others” (30).

Neurological Hypothesis

CFS as a primary brain disorder has been studied with neuroimaging including Magnetic resonance imaging MRI, Single-photon emission computed tomography (SPECT) Electroencephalogram (EEG), quantitative electroencephalogram (qEEG), and positron emission tomography (PET) (32-36, 40-41). A variety of abnormalities associated with CFS have been reported but the diagnostic or potential pathogenic implications of these findings are unknown.

Neuroendocrine Hypotheses

In recent years, there have been reports indicating neuroendocrine hypofunction, probably of hypothalamic origin, in chronic fatigue states. A tendency to hypocortisolism, has been identified, albeit inconsistently, in CFS patients. Relative hypocortisolism may reflect the primary abnormality in many CFS patients, such as a disorder of the brain regulation, or peripheral elements, of the stress system. Moreover, hypocortisolism may contribute to CFS symptomatology.

 

However, neuroendocrine studies in CFS have often led to contradictory results. Smaller studies may be confounded by differences between subgroups of CFS patients, such as duration of fatigue, concomitant hypotension and/or orthostasis, depression, familial occurrence, and other factors. Although melancholic major depression is associated with mild hypercortisolism, the hypocortisolism of CFS seems to persist in at least some patients with co-morbid depression (28). Moreover, hypocortisolism is a trait shared with other chronic idiopathic disorders, including post-traumatic stress disorder, fibromyalgia, and inflammatory disorders such as rheumatoid arthritis and asthma (18). Wyller et al. studied 120 CFS patients and 68 healthy controls, aged 12-18 years. CFS patients had higher levels of plasma norepinephrine, plasma epinephrine and FT4, with lower urine cortisol/creatinine ratios, (42). This accords with previous studies of attenuation of cortisol secretion and enhancement of the sympathetic nervous system activity in CFS.

THE STRESS SYSTEM AND CFS/SEID

Stress is defined as threat to homeostasis. It is generally accepted that acute stress system responses are adaptive, designed to re-establish homeostasis. However excessive and/or prolonged activation of the stress system can disturb normal physiology. The stress system comprises the hypothalamic-pituitary-adrenal (HPA) axis of which cortisol is the major mediator, and the sympathoadrenal system which produces the catecholamines epinephrine and the sympathoneural system producing norepinephrine. Both glucocorticoids and catecholamines act widely to mediate the stress response.

 

Stress results in stimulation of parvicellular neurons of the paraventricular nucleus (PVN) of the hypothalamus and the release of the neuropeptides corticotropin releasing hormone (CRH) and arginine vasopressin (AVP) into the hypophyseal portal blood system (Figure 2). The combined action of CRH and AVP on the anterior pituitary corticotropes stimulates secretion of adrenocorticotropin hormone (ACTH). Circulating ACTH acts on the zona fasciculata of the adrenal cortex to stimulate cortisol synthesis. Basal (unstressed) cortisol acts to prevent arterial hypotension by augmenting the effects of catecholamines, and maintain normoglycemia through insulin counter-regulation.

 

ACTH secretion is influenced by stress, a light-entrained circadian rhythm, and negative feedback at the hypothalamus. During acute stress, the amplitude and synchronization of the CRH and AVP pulsations in the hypophyseal portal system markedly increases, resulting in increases of ACTH and cortisol secretory episodes (43). Stress-induced cortisol secretion activates the central nervous system, increases blood pressure, elevates blood glucose, and suppresses the inflammatory/immune response to prevent tissue damage (44).

 

Cortisol action is mediated by ubiquitous cytosolic corticosteroid receptors and (45). Free cortisol, unbound to corticosteroid binding globulin (3-10%), diffuses through cell membranes and binds to the carboxy-terminal end of the cytosolic glucocorticoid receptor. On cortisol binding, the ligand-receptor complex translocates into the nucleus, where it interacts with specific glucocorticoid responsive elements (GREs) within DNA to activate gene transcription (45). The activated receptors also inhibit other transcription factors, such as c-Jun/c-Fos and NF-kB, which are positive regulators of the transcription of genes involved in the activation and growth of immune and other cells (46).

Figure 2. The neurohormonal connections of the stress system.

 

Several complementary sets of studies have examined basal and stimulated pituitary-adrenal gland function in CFS.

 

Two different types of heritable disorders of this axis have been described, where fatigue is the principal symptom. These include glucocorticoid resistance due to glucocorticoid receptor abnormalities, and mutations of the corticosteroid-binding globulin gene, the chief cortisol transport protein. These disorders are rare, but reinforce the notion that primary pituitary-adrenal abnormalities may produce chronic fatigue. Studies in the broader CFS patient group have generally detected relative hypocortisolism and altered dynamic responses, providing indirect evidence of a central nervous system under-stimulation of pituitary-adrenal function.

 

Familial glucocorticoid resistance is a rare syndrome characterized by diminished tissue effect of cortisol as a result of a glucocorticoid receptor defect. Glucocorticoid resistance is generally due to a loss of function mutation of the glucocorticoid receptor gene, although the genetic defect has not been identified in all cases. Decreased sensitivity to cortisol results in activation of the HPA axis, with increased ACTH and cortisol levels. In most cases, elevated cortisol levels sufficiently compensate to overcome the hormone resistance, thus these patients do not clinically manifest either cortisol excess or deficiency. Increased ACTH secretion also results in elevated mineralocorticoid and androgen levels resulting in hypertension and hirsutism (47). However, fatigue as an isolated symptom has been described in a 55-year-old woman with glucocorticoid resistance (48). Fatigue in this patient was intermittent, but blood pressure was constantly in the low-normal range, with no postural hypotension. Fatigue was sufficient to prohibit full-time work. Urinary cortisol was elevated (400-800nmol/24h; Range <300nmol/24h), as were plasma cortisol levels. A thermolabile glucocorticoid receptor was noted, specifically a temperature-induced reduction in dexamethasone binding, although a specific glucocorticoid receptor mutation was not reported. It has been proposed that fatigue in such cases is a result of insufficient overproduction of cortisol (49).

 

Further to this, recent studies of glucocorticoid receptor polymorphisms have found an association between certain haplotypes and CFS (50). Although speculative, polymorphisms may result in altered receptor sensitivity to cortisol, and thus, impaired tissue-effect of cortisol, resulting in relative hypocortisolism.

CORTICOSTERIOD BINDING GLOBULIN ABNORMALITIES AND CHRONIC FATIGUE

Corticosteroid-binding globulin (CBG), also known as transcortin, is the high-affinity plasma transport glycoprotein for cortisol (51). It is secreted by hepatocytes as a 383-amino acid polypeptide, after cleavage of a 22-amino acid signal peptide. Each CBG molecule contains a single high-affinity steroid binding site (51). Under circadian conditions, 80% of circulating cortisol is bound to CBG, 10-15% is bound to low-affinity albumin and 5-8% of circulating cortisol is unbound or free (51). Currently, only the free fraction is thought to be biologically active. CBG levels are generally stable. CBG is traditionally thought to function primarily as a carrier molecule for cortisol, but it may also serve as a buffer and as a reservoir, during secretory surges, or during times of reduced cortisol secretion, respectively. CBG may also have a specific-tissue cortisol delivery role, in particular enabling cortisol to act in an immunomodulatory capacity (52). High-affinity cortisol binding is saturated beyond cortisol levels of 500nmol/L, hence free cortisol levels rise exponentially at high cortisol concentrations (53). Under conditions of stress, elevated cortisol levels saturate available CBG and increase the free cortisol to above 20% (53).

 

CBG is involved in the stress response. Immune activation releases interleukin-6 (IL-6) which increases circulating free cortisol levels by two mechanisms. IL-6 stimulates cortisol secretion through activation of hypothalamic CRH neurons and it also inhibits CBG gene transcription thereby increasing the free cortisol fraction and thus, circulating glucocorticoid activity (54,55). In vivo, exogenous IL-6 results in a 50% reduction in CBG levels in humans. Severe illness, such as sepsis and burns, are associated with similar reductions in CBG levels, in conjunction with a similar rise in endogenous IL-6 (56,57). Similar falls in circulating CBG concentrations are seen in septic shock and low CBG concentrations have been shown to be an independent predictor of mortality in ICU patients (58). Stress-induced falls in circulating CBG concentrations may also relate to cortisol elevations, as low CBG levels are seen in Cushing’s syndrome or after anti-inflammatory glucocorticoid doses (57). This effect is probably mediated through the glucocorticoid receptor as glucocorticoid receptor knockout mice exhibit increased hepatic CBG expression and 50% increased plasma CBG levels (59).

 

CBG Lyon refers to a CBG gene mutation that was first described in a 43-year-old Moroccan woman presenting with chronic fatigue, depressed mood and low blood pressure, suggesting adrenal insufficiency (60). She had very low plasma cortisol levels, but normal ACTH levels. She was found to be homozygous for a point mutation in exon 5, leading to an Asp-Asn substitution, and a 4-fold reduction in CBG-cortisol binding affinity. Immunoreactive-CBG levels were 50% of the lower limit of normal, suggesting that the mutation affects CBG secretion or degradation. The proband’s four children were heterozygous for the mutation. A 10-member Brazilian kindred with the same genetic mutation and reduced CBG-binding affinity has also been described, having been discovered after low cortisol levels were detected in the proband, a homozygote, who presented with fatigue (61). One other kindred member was a homozygote, the rest were heterozygotes, all were normotensive and none experienced fatigue.

 

In 2001, a 39-member Italian-Australian family was reported, including 21 heterozygotes and 3 homozygotes with a novel complete loss-of-function (null) CBG gene mutation involving exon 2 (62). The null mutation is a point mutation leading to a premature stop codon corresponding to residue -12 (tryptophan) of the pro-CBG molecule. It resulted in a 50% reduction of or undetectable CBG levels in heterozygotes or homozygotes, respectively. The proband was investigated because of unexplained fatigue and low blood pressure, suggesting glucocorticoid deficiency, and the finding of low plasma but normal urine cortisol levels, suggesting CBG deficiency. Amongst kindred members who were homozygous or heterozygous for the mutation, there was a high prevalence of chronic fatigue and low blood pressure. Surprisingly, five members had the previously reported CBG Lyon mutation.

 

Hence, CBG gene mutations are associated, albeit, inconsistently, with fatigue. Amongst CFS patients, the Lyon and Null mutations have not been detected (63- 65). To date several CBG mutations were identified following investigations of patients presenting with low plasma cortisol in variety of medical conditions such as chronic fatigue (66).

PITUITARY-ADRENAL HORMONE ABNORMALITIES IN CHRONIC FATIGUE SYNDROME

Recent interest in the role of the HPA axis in CFS has arisen from the observation that conditions in which there is low circulating cortisol are characterized by debilitating fatigue. Addison’s disease, glucocorticoid withdrawal, and bilateral adrenalectomy are all associated with fatigue and with other symptoms also seen in CFS, including arthralgia, myalgia, disturbed sleep, and mood (67). Many studies provide inconsistent data on HPA axis function in patients with CFS, in part because of methodological differences, but also reflecting, perhaps, individual variation in HPA axis activity.

 

Urinary free cortisol levels in CFS patients have been found to be significantly lower, or no different to, controls (68-71). Plasma morning and late evening cortisol has been shown to be reduced in CFS/ME, but this finding has not been consistently reproduced, particularly when frequent plasma cortisol sampling has been performed (69,71). Salivary cortisol has emerged as a useful test to detect hypercortisolism because of its non-invasiveness and correlation with free blood cortisol levels. In CFS, salivary cortisol day-curves are blunted compared with controls, evening salivary cortisol levels are lower, and there is a blunted salivary cortisol rise in response to waking (72-75). DHEA and its long half-life sulphated metabolite DHEA-S represent major adrenal gland products in terms of mass. They represent important contributors to circulating androgen activity, particularly in women. DHEA and DHEA-S levels were shown to be lower in 15 CFS patients relative to 11 controls; furthermore, CFS patients did not display the usual decrease in DHEA:cortisol ratio with ACTH stimulation (76). A preliminary study in eight selected CFS patients with a subnormal 1μg ACTH stimulation test showed a 50% reduction in adrenal gland volume on CT scan (77). This finding might indicate that the hypocortisolism of CFS is due to a lack of ACTH stimulation or a primary adrenal abnormality. In a recent study, however, DHEA levels were higher in CFS patients and were correlated with higher disability scores (78).

 

To further examine the endocrine axes, stimulation testing is a classic endocrine paradigm, where subtle hypofunction may become more evident through the administration of stimulatory hormones or neuroactive agents. Nevertheless, as central control of endocrine axes cannot be directly assessed due to the lack of accessibility of the hypothalamic-pituitary circulation, the interpretation of the findings tends to be indirect. Often it is necessary to implicate underlying receptor up or down-regulation or secondary adrenal atrophy. Moreover, neuroactive agents often have incomplete specificity and the central neurotransmitter systems under study may in fact not be exclusively tested.

 

Dynamic endocrine testing with human CRH (pituitary stimulus) in CFS patients revealed a trend towards lower cortisol responses – which became statistically significant if ACTH responses were analyzed as a covariate (79). ACTH responses to CRH may also be blunted in CFS (80). Other studies have found a normal ACTH and cortisol rise to CRH in CFS patients, which contradict the hypothesis, and previous data, suggesting that CFS is associated with a blunting of the HPA axis (81).

 

Insulin hypoglycemia is a profound stimulus of ACTH and cortisol release, as it is likely to induce release of many hypothalamic ACTH secretagogues. Studies in CFS have revealed increased ACTH but normal cortisol responses after insulin hypoglycemia (82). This could be interpreted as indicating low CRH tone, with chronic CRH hyposecretion despite an intact CRH neuron, and secondary adrenal atrophy.

 

Naloxone is thought to stimulate ACTH and cortisol secretion by blocking tonic opioidergic inhibition of the CRH neuron. Naloxone mediated activation may be blunted in CFS suggesting it is the CRH neuron or pathways inhibitory to this neuron that lead to HPA axis hypofunction in CFS, rather than increased opioidergic tone (83). Other studies of CFS patients have a normal ACTH and cortisol response to naloxone (81).

 

The waking cortisol response, where cortisol levels rise 30-50% by 30 mins after waking compared to levels immediately on waking, is attenuated in chronic fatigue syndrome as a result of both higher waking and lower 30 min salivary cortisol levels, as documented in 75 CFS patients versus controls (82).

 

Another explanation for the hypocortisolism of CFS is increased glucocorticoid sensitivity, particularly in relation to the cerebral structures involved in glucocorticoid feedback such as the hypothalamic-paraventricular nucleus, the site of CRH neurons, and the anterior pituitary and hippocampus. Increased glucocorticoid sensitivity has been described in other stress-related hypocortisolemic disorders, such as post-traumatic stress disorder, and has recently been reported in a small study of CFS patients (85).

 

Finally, it is not known if the hypocortisolism of CFS is a response to chronic deconditioning since exercise is a potent stimulator of HPA axis function. Experimental acute exercise deprivation led to some symptoms relating to pain, fatigue and mood as well as lower cortisol in a subset of healthy individuals (86).

 

CFS is associated with prominent features of autonomic dysregulation such as postural hypotension, disturbances in temperature regulation, and altered skin microcirculation. The other arm of the stress system, the sympathetic nervous system with its outflow components, the sympathoneural and sympathoadrenal limbs have been less studied than cortisol in CFS. However, studies of both norepinephrine levels and a variety of tests of autonomic function suggest hyperactivity of the SNS, perhaps as a response to inadequate HPA axis responsivity (87,88).

 

The data suggesting relative hypocortisolism in CFS, along with the co-existence of fatigue, low blood pressure, and mood alterations in both Addison’s disease and CFS, have led to trials of hydrocortisone therapy in CFS. A randomized crossover trial in 32 CFS patients, of low-dose hydrocortisone (5mg or 10mg) treatment compared with placebo showed a reduction in self-reported fatigue scores after 1 month of treatment (89). In 28% of patients taking hydrocortisone, fatigue scores reached a predefined cut-off value similar to the normal population score. Only 9% of patients taking placebo achieved this reduction in fatigue score. However, another trial of hydrocortisone treatment in CFS, have subsequently shown no real benefit of treatment. The trial which included 70 patients, treated with hydrocortisone (16mg/m2 daily in 2 divided doses) for 3 months reported some improvement in symptom scales (90). It is of interest that those with the lowest cortisol levels and adrenal reserve were not the most symptomatic, nor were they more likely to respond to hydrocortisone treatment. Adverse effects including weight gain, increased appetite, and disturbed sleep, occurred in those taking hydrocortisone. Hydrocortisone treatment was also associated with significant adrenal suppression, on the basis of basal and ACTH-stimulated cortisol levels in 12 patients. The authors concluded that the risks of adrenal crisis outweighed any perceived benefit of treatment and therefore that systemic corticosteroids should not be used in the treatment of CFS (90).

 

Blockmans et al., reported six month randomized, placebo-controlled, double-blind, crossover study of hydrocortisone (5mg/day) and fludrocortisone in 100 patients fulfilling the CDC criteria for CFS (91). There was no benefit of treatment on self-reported fatigue or well-being.

 

Fludrocortisone (0.1-0.2mg) was tested in a placebo-controlled, double-blind crossover trial. No improvement in symptoms, treadmill exercise performance, or reaction time was observed in the 20 CFS patients who completed the trial (92).

 

The available scientific data indicates that the symptomatic benefit achieved with hydrocortisone or fludrocortisone replacement is, at best, marginal, and importantly, may be associated with clinically significant adverse effects, including adrenal suppression or features of glucocorticoid excess. These adverse effects outweigh any perceived benefit of treatment. Overall, hydrocortisone and fludrocortisone treatment in CFS patients is not justified. In addition, ACTH stimulation testing has no practical relevance in the routine assessment of CFS patients, and should not be used to formulate management decisions, but may be used to rule out adrenal insufficiency.

 

Although low cortisol may not be the chief source of disability in CFS, it may be a marker of therapeutic significance. For example, the response to cognitive behavioral therapy is reduced in those with lower urine free cortisol or an attenuated diurnal rhythm (93).

 

The COVID-19 pandemic has led to a variety of symptoms after acute illness recovery. The recovery process from COVID-19 varies between individuals, depending on factors such as the illness severity, age, and underlying comorbidities. Despite not having a widely accepted definition, Centers for Disease and Prevention and the World Health Organization (WHO) has agreed the acute symptoms of COVID can last up to four weeks following the onset of the illness (94,95). Various terminologies such as “long COVID’, “post-acute sequalae of SARS-CoV-2 infection”, “post-acute COVID-19” have been used to describe the prolonged symptoms following COVID-19. In this article, we will use Long COVID to describe the condition.

 

While Long COVID and chronic fatigue syndrome/myalgia encephalitis (CFS/ME) are distinct conditions, they do share some similarities in terms of symptoms and impact on individual’s lives. Both conditions are characterized by persistent and debilitating fatigue. It is worth noting that CFS/ME is diagnosed after fatigue present for at least six months, which is not relieved by rest while fatigue experienced in Long COVID can last for weeks, months or longer. The accompanying symptoms of Long COVID syndrome are broad and can affect multiple organ systems including respiratory and cardiac symptoms, which does not typically present in CFS/ME (94- 97). While the triggering event of long COVID is attributed to COVID itself, the triggering event of CFS/ME is not fully understood. Patients with long COVID syndrome may have symptoms consistent with and meet diagnostic criteria of CFS/ME where similar assessment and management strategy can be employed.

MANAGEMENT

Generally, all treatment for CFS/ME must be individualized aiming to address the most debilitating symptom first. No specific treatment is known to be successful for CFS as the current evidence for pharmacological or non-pharmacological interventions was heterogenous and inconclusive (98). However, diagnosis may help patients by providing a basis for prognostic advice and validating their need for assistance in their personal lives and workplace.

Symptomatic treatments, such as non-steroidal anti-inflammatory drugs or non-opiate analgesics for pain and counselling or antidepressants for major depression, are commonly used in ME/CFS although their efficacy has not been the subject of a long-term trial. Developing good sleep hygiene to provide sufficient rest is often part of the management strategy. The latest NICE guideline also suggested dietary strategies including adequate hydration, referral to dietician for patients at risk of weight gain or malnutrition, as well as vitamin D repletion for vitamin D deficiency. It is important to explain to patients with ME/CFS that there is insufficient evidence to support routine vitamin supplementations as treatment for the condition (NICE) (98). Patients with significant cognitive decline should be referred for further neurocognitive evaluation.

 

Cognitive behavioral therapy involves the provision of information and counselling to reduce the psychological impediments to recovery, as well as encouraging the patient to participate at an appropriate level of social and occupational activity. It is important for clinicians to establish a rapport as patients may be mistrustful due to prior negative health care experiences (99). In randomized-controlled trials comparing CBT to control conditions, the intervention has been shown to have a positive overall effect (21). Graded-exercise therapy may also be of benefit (22).

 

No pharmacological agent has been reproducibly shown to be effective in the treatment of chronic fatigue syndrome.

 

Rintatolimod is an antiviral, restricted Toll-like Receptor 3 (TLR3) agonist lacking activation of other primary cellular inducers of innate immunity. It also activates interferon-induced protein. A systemic review suggested some evidence that Rinatotolimod may improve symptoms of ME/CFS (100). Another double blind, randomized, placebo-controlled clinical trial showed statistically significant improvements in primary endpoints in phase II and phase III trials.  About 30-40% of ME/CFS patients can be expected to respond beneficially to Rintatolimod (101). Previous double-blind, randomized clinical trial of Rintatolimod showed an improvement in exercise tolerance and improvement of medication usage for CFS/ME-related symptoms (102). However, the application to the US Food and Drug Administration (FDA) was rejected in 2009 as the previous RCTs that failed to provide credible evidence of efficacy (103).  At present, Rintatolimod is only approved for use in Argentina. Therefore, some authorities suggest Rintatolimod should be considered an experimental drug until confirmatory studies are available (32).

 

Rituximab is an anti-CD20 monoclonal antibody. There may be some benefits shown in a small double-blind, placebo-controlled trials involving 30 patients, particularly in patients with self-reported fatigue, but a subsequent, larger study showed no difference in the treated group and the control group after 24 months of treatment (104,105).

 

A small trial revealed significant improvement in ME/CFS patients who received CoQ10 plus NADH supplementation, but a larger study is warranted to verify its beneficial effect in ME/CFS patients (106).

 

There is a list of therapies that have been trailed in the past, with no proven benefit over placebo. These therapies include acyclovir, antibiotics, cytokine inhibitors, galantamine, glucocorticoids, mofadanil, and methylphenidate (107-113).

PROGNOSIS

Overall, full recovery from untreated ME/CFS is rare but improvement of symptoms in long term is slightly more optimistic (114-116). However, the prognosis of ME/CFS also varies widely among individuals. The reported improvement rates range from 0 to 8% (117-122). Broad range improvement rate is reported at 17-64% (117,120,122,123). A study suggested although most patients with this condition improve, a significant proportion remain functionally impaired over time (118). Another study that was conducted using a questionnaire, reported 73% of patients remain functionally impaired at six weeks to six months but this improved to 33% at two to four years (115).

 

A systematic review showed the median full recovery only happened in 5% of patients (122). Another retrospective study that includes patients with unexplained debilitating fatigue lasting for more than six months but does not fulfil the criteria of ME/CFS showed complete resolution of symptoms only occurred in 2% of these patients (119).

 

As there was lack of operationalized criteria for recovery and improvement, the studies yielded contradictory results in terms of factors that predict the likelihood of recovery. Some studies suggested that old age is associated with poorer outcome while others did not support this hypothesis (118,119,124,125). There has been mixed evidence that shorter duration of illness to be a predictor of better improvement (118,121). Mixed evidence was demonstrated across studies with regards to a worse prognosis in patients with comorbid fibromyalgia (125-127).  There may be an increased risk of suicide (128).

 

ME/CFS has not been associated with increased mortality rate. Treatment is supportive and a defined pathogenesis has not been identified, despite a syndromic definition that is quite frequent and stable across individuals and populations. 

CONCLUSION

Many diagnostic criteria exist for MF/ CFS but the emphasis on exercise intolerance is thought to have significant specificity, although secondary features are also typical. The stress system has been shown to exhibit a reasonably consistent phenotypic pattern comprising relatively low cortisol and elevated sympathetic, particularly sympathoneural function. The etiology of ME/CFS is unknown and the mechanism of altered stress system function is uncertain. Several other pathogenetic mechanisms are proposed. Currently, some treatment trials have been promising and confirmation of their effect is awaited.

REFERENCES

  1. Crownley N, Nelson M, Stovin S. Epidemiological aspects of an outbreak of encephalomyelitis at the Royal Free Hospital, London, in the summer of 1955. J Hyg (Lond). 1957 Mar;55(1):102-22.
  2. Holmes GP, Kaplan JE, Gantz NM, et al. Chronic Fatigue Syndrome: A Working Case Definition. Ann Intern Med. 1988; 108: 387-9.
  3. Fukuda K, Straus SE, Hickie I, et al, and the International Chronic Fatigue Syndrome Study Group. Chronic Fatigue Syndrome: a comprehensive approach to its definition and study. Ann Intern Med 1994;121:953-9.
  4. Carruthers BM, Jain AK, De Meirleir KL, et al. Myalgic Encephalomyelitis/Chronic Fatigue Syndrome: Clinical Working Case Definition, Diagnostic and Treatment Protocols. J Chronic Fatigue Syndr 2003;11(1):7-115.
  5. Jason LA, Sunnquist M, Brown A, Newton JL, Strand EB, Vernon SD. Chronic Fatigue Syndrome versus Systemic Exertion Intolerance Disease. 2015;3(3):127-4
  6. Myalgic Encephalomyelitis/Chronic Fatigue Syndrome. Centre for Disease Control and Prevention (CDC). Updated April, 2021. Accessed June 2023. https://www.cdc.gov/me-cfs/healthcare-providers/diagnosis/iom-2015-diagnostic-criteria.html
  7. Schwartz RB, Garada BM, Komaroff AL, Tice HM, Gleit M, Jolesz FA, Holman BL. Detection of intracranial abnormalities in patients with chronic fatigue syndrome: comparison of MR imaging and SPECT. AJR Am J Roentgenol. 1994 Apr;162(4):935-41. doi: 10.2214/ajr.162.4.8141020.
  8. Buchwald D, Cheney PR, Peterson DL, Henry B, Wormsley SB, Geiger A, Ablashi DV, Salahuddin SZ, Saxinger C, Biddle R, Kikinis R, Jolesz FA, Folks T, Balachandran N, Peter JB, Gallo RC, Komaroff AL. A chronic illness characterized by fatigue, neurologic and immunologic disorders, and active human herpesvirus type 6 infection. Ann Intern Med. 1992 Jan 15;116(2):103-13.
  9. Newton JL, Finkelmeyer A, Petrides G, Frith J, Hodgson T, Maclachlan L, et al. reduced Cardiac volumes in chronic fatigue syndrome associate with plasma volume but not length of disease: a cohort study. Open Heart. 2016;3(1):e000381
  10. Olimulder MA, Galjee MA, Wagenaar LJ, van Es J, van der Palen J, Visser FC, et al. Chronic fatigue syndrome in women assessed with combined cardiac magnetic resonance imaging. Neth Heart J. 2016;24(12):709-16
  11. Reyes M, Nisenbaum R, Hoaglin DC, et al. Prevalence and Incidence of Chronic Fatigue Syndrome in Wichita, Kansas. Arch Intern Med 2003;163:1530-6.
  12. Jason LA, Richman JA, Rademaker AW, et al. A Community-Based Study of Chronic Fatigue Syndrome. Arch Intern Med 1999;159:2129-37.
  13. Twisk FNM. A critical analysis of the proposal of the Institute of Medicine to replace myalgic encephalomyelitis and chronic fatigue syndrome by a new diagnostic entity called systemic exertion intolerance disease. Current Medical Research and Opinion 2015;31:1333–47.
  14. Steele L, Dobbins JG, Fukada K, et al. The epidemiology of chronic fatigue in San Fracisco. Am J Med. 1998; 105 (3A): 83S-90S.
  15. Bakken I,Tveito K,Gunness, et al.Two age peaks in the incidence of chronic fatigue syndrome/Myalgic encephalomyelitis:a population-based registry study from Norway 2008-2012.BMC Med 2014 Oct1:12(1):167
  16. Reeves WC, Jones JF, Maloney E, et al. Prevalence of Chronic Fatigue Syndrome in Metropolitan, Urban and Rural Georgia. Pop Health Metr 2007;5:5.
  17. Kato K, Sullivan PF, Evengård B, et al. Premorbid Predictors of Chronic Fatigue. Arch Gen Psych 2006;63:1267-72.
  18. Heim C, Wagner D, Maloney E, et al. Early Adverse Experience and Risk for Chronic Fatigue Syndrome. Results From a Population-Based Study. Arch Gen Psych 2006;63:1258-66.
  19. Johnston SC, Staines DR, Marshall-Gradisnik SM. Epidemiological characteristics of chronic fatigue syndrome/myalgic encephalomyelitis in Australian patients. Clinical Epidemiology 2016;8:97-107
  20. Clauw DJ, Chrousos GP. Chronic pain and fatigue syndromes: overlapping clinical and neuroendocrine features and potential pathogenic mechanisms. Neuroimmunomodulation 1997;4:134-53
  21. Whiting P, Bagnall A-M, Sowden AJ, et al. Interventions for the treatment and management of Chronic Fatigue Syndrome. JAMA 2001;286:1360-8
  22. White PD, Thomas JM, Amessis J, et al. The existence of a fatigue syndrome after glandular fever. Psychol Med 1995;25:907-16.
  23. Smith WR, Noonan C, Buchwald D. Mortality in a cohort of chronically fatigued patients. Psychol Med 2006;36:1301-6
  24. Hardcastle SL, Brenu EW, Johnston S, Nguyen T, Huth T, Wong N, et al. Characterisation of cell functions and receptors in Chronic Fatigue Syndrome/Myalgic Encephalomyelitis (CFS/ME). BMC Immunol. 2015;16:35
  25. Yang TY, Kuo HT, Chen HJ, Chen CS, Lin WM, Tsai SY, et al. Increased Risk of Chronic Fatigue Syndrome Following Atopy: A Population-Based Study. Medicine (Baltimore). 2015;94(29):e1211
  26. Ciregia F, Kollipara L, Giusti L, Zahedi RP, Giacomelli C, Mazzoni MR et al. Proteomics suggests an association between differential expression of mitochondrial proteins and chronic fatigue syndrome. Transl Psychiatry. 2016;6(9):e904
  27. Komaroff AL, Fagioli LR, Geiger AM, et al. An examination of the working case definition of chronic fatigue syndrome. Am J Med 1996;100:56-64.
  28. Wessely S, Butler S, Chalder T, et al. The cognitive behavioral management of the post-viral fatigue syndrome. In R. Jenkins and J. Mowbrey (Eds.), Postviral fatigue syndrome (pp.305-334). Chichester: John Wiley and Sons.
  29. Surawy C, Hackmann A, Hawton K, et al. Chronic fatigue syndrome: a cognitive approach. Behaviour Research Therapy 1995;33:535-44.
  30. White C, Schwitzer R. The role of personality in the development and perpetuation of chronic fatigue syndrome. J Psychosomatic Med 2000;48:515-24.
  31. Rasouli O, Fors EA, Borchgrevink PC, Ohberg F, Stensdotter AK. Gross and fine motor function in fibromyalgia and chronic fatigue syndrome. J Pain Res. 2017;10:303-9
  32. Gluckman SJ. Treatment of systemic exertion intolerance disease (chronic fatigue syndrome). In: Aronson MD, Mitty J, editors. UpToDate. Waltham, MA: UpToDate; 2017
  33. Wu T, Qi X, Su Y, Teng J, Xu X. Electroencephalogram characteristics in patients with chronic fatigue syndrome. Neuropsychiatr Dis Treat. 2016;12:241-9.
  34. Tuller, David (2014-11-24), "Brains of People With Chronic Fatigue Syndrome Offer Clues About Disorder", NY Times.
  35. Zeineh, Michael M; Kang, James; Atlas, Scott W; et al. (2014-10-29), "Right Arcuate Fasciculus Abnormality in Chronic Fatigue Syndrome", Radiology, 274 (2): 517–526, doi:10.1148/radiol.14141079.
  36. Goldman, Bruce (2014-10-28), "Study finds brain abnormalities in chronic fatigue patients", Stanford Medicine News Center.
  37. Shan, ZY; Kwiatek, R; Burnet, R; Del Fante, P; Staines, DR; Marshall-Gradisnik, SM; Barnden, LR (2016-04-28), "Progressive brain changes in patients with chronic fatigue syndrome: A longitudinal MRI study", Journal of magnetic resonance imaging: JMRI, PMID 27123773, doi:10.1002/jmri.25283.
  38. Jaime S (2016-05-05), "Progressive Brain Changes in Patients with Chronic Fatigue Syndrome: Are our Brains Starved of Oxygen?", #MEAction
  39. Zinn, Marcie L; Zinn, Mark A; Jason, Leonard A (2016), "qEEG / LORETA in Assessment of Neurocognitive Impairment in a Patient with Chronic Fatigue Syndrome: A Case Report", Clinical Research: Open Access ( ISSN 2469-6714 ), 2 (1), ISSN 2469-6714, doi:10.16966/2469-6714.110.
  40. Nakatomi, Yasuhito; Mizuno, Kei; Ishii, Akira; et al. (2014-03-24), "Neuroinflammation in Patients with Chronic Fatigue Syndrome/Myalgic Encephalomyelitis: An ¹¹C-(R)-PK11195 PET Study", Journal of Nuclear Medicine, 2014 Jun;55(6): 945-50, PMID 24665088, doi:10.2967/jnumed.113.131045.
  41. Puri, BK; Jakeman, PM; Agour, M; Gunatilake, KDR; Fernando, KAC; Gurusinghe, AI; Treasaden, IH; Waldman, AD; Gishen, P (2012), "Regional grey and white matter volumetric changes in myalgic encephalomyelitis (chronic fatigue syndrome): a voxel-based morphometry 3 T MRI study", British Journal of Radiology, 85 (1015): e270-3, doi:10.1259/bjr/93889091.
  42. Wyller VB, Vitelli V, Sulheim D, Fagermoen E, Winger A, Godang K, et al. Altered neuroendocrine control and association to clinical symptoms in adolescent chronic fatigue syndrome: a cross-sectional study. J Transl Med. 2016;14(1):121.
  43. Tsigos C, Chrousos GP. Physiology of the hypothalamic-pituitary-adrenal axis in health and dysregulation in psychiatric and autoimmune disorders. Endocrinol Metab Clin Nth Am 1994;23:451-66.
  44. Munck A, Guyre PM, Holbrook NJ. Physiological functions of glucocorticoids in stress and their relation to physiological actions. Endocr Rev 1984;5:25-44.
  45. Pratt WB. Glucocorticoid receptor structure and the initial events in signal transduction. Prog Clin Biol Res 1990;322:119-32.
  46. Scheinman RI, Gualberto A, Jewell CM, et al. Characterisation of mechanisms involved in transrepression of NF-KB by activated glucocorticoid receptors. Mol Cell Biol 1995;15:943-53.
  47. van Rossum EFC and Lamberts SWJ. Glucocorticoid resistance syndrome: a diagnostic and therapeutic approach. Best Pract Res Clin Endocrinol Metab 2006;20:611-26.
  48. Bronnegard M, Werner S, Gustafsson JA. Primary cortisol resistance associated with a thermolabile glucocorticoid receptor in a patient with fatigue as the only symptom. J Clin Invest 1989;78:1270-8.
  49. Huizenga N, De Lange P, Koper JW, et al. Five Patients with Biochemical and/or Clinical Generalised Glucocorticoid Resistance without Alterations in the Glucocorticoid Receptor Gene. J Clin Endocrinol Metab 2000;85:2076-81.
  50. Rajeevan MS, Smith AK, Dimulescu I, et al. Glucocorticoid receptor polymorphisms and haplotypes associated with chronic fatigue syndrome. Genes, Brain and Behav 2007;6:167-76.
  51. Hammond GL. Molecular properties of corticosteroid binding globulin and the sex-steroid binding proteins. Endocr Rev 1990;11:65-79.
  52. Hammond GL, Smith CL, Paterson NA, et al. A role for corticosteroid-binding globulin in delivery of cortisol to activated neutrophils. J Clin Endocrinol Metab 1990;71:34-9.
  53. Ballard PL. Delivery and transport of glucocorticoids to target cells. Monographs on Endocrinology 1979;12:25-48.
  54. Papanicolaou DA, Wilder RL, Manolagas SC, et al. The pathophysiologic roles of interleukin-6 in human disease. Ann Intern Med 1998;128:127-37.
  55. Tsigos C, Kyrou I, Chrousos GP, et al. Prolonged suppression of corticosteroid-binding globulin by recombinant human interleukin-6 in man. J Clin Endocrinol Metab 1998;83:3379.
  56. Ho JT, Al-Musalhi H, Chapman MJ, et al. Septic shock and sepsis: a comparison of total and free plasma cortisol levels. J Clin Endocrinol Metab 2006;91:105-14.
  57. Bernier J, Jobin N, Emptoz-Bonneton A, et al. Decreased corticosteroid-binding globulin in burn patients: relationship with interleukin-6 and fat in nutritional support. Crit Care Med 1998;26:452-60.
  58. Meyer EJ, Nenke MA, Davies ML, et al. Corticosteroid-Binding Globulin Deficiency Indepednently Predicts Mortaility in Septic Shock. J Clin Endocrinol Metab. 2022; 107:1636-1646.
  59. Cole TJ, Harris HJ, Hoong I, et al. The glucocorticoid receptor is essential for maintaining basal and dexamethasone-induced repression of the murine corticosteroid-binding globulin gene. Mol Cell Endocrinol 1999;154:29-36.
  60. Emptoz-Bonneton A, Cousin P, Seguchi K, et al. Novel human corticosteroid-binding globulin variant with low cortisol-binding affinity. J Clin Endocrinol Metab 2000;85:361-7.
  61. Brunner E, Baima J, Vieira TC, et al. Hereditary corticosteroid-binding globulin deficiency due to a missense mutation (Asp367Asn, CBG Lyon) in a Brazilian kindred. Clin Endocrinol 2003;58:756-62.
  62. Torpy DJ, Bachmann AW, Grice JE, et al. Familial Corticosteroid-Binding Globulin Deficiency Due to a Novel Null Mutation: Association with Fatigue and Relative Hypotension. J Clin Endocrinol Metab 2001;86:3692-700.
  63. Torpy DJ, Bachmann AW, Gartside M, et al. Association between chronic fatigue syndrome and the corticosteroid-binding globulin gene ALA SER224 polymorphism. Endocr Res 2004;30:417-29.
  64. Smith CL, Power SG and Hammond GL. A Leu-His substitution at residue 93 in human corticosteroid binding globulin results in reduced affinity for cortisol. J Steroid Biochem Mol Biol 1992;42:671-6.
  65. Torpy DJ and Ho JT. Corticosteroid-binding globulin gene polymorphisms: clinical implications and links to idiopathic chronic fatigue disorders. Clin Endocrinol 2007 Aug;67(2):161-7.
  66. Meyer EJ, Spangenberg L, Rmirez MJ, et al Corticosteroid-Binding Globulin. J Endocr Soc. 2021 Jun 22; 5(9):bvab115.
  67. Cleare AJ. The Neuroendocrinology of Chronic Fatigue Syndrome. Endocr Rev 2003;24:236-52.
  68. Cleare AJ, Blair D, Chambers S, et al. Urinary free cortisol in Chronic Fatigue Syndrome. Am J Psych 2001;158:641-3.
  69. Jerjes WK, Taylor NF, Peters TJ, et al. Urinary cortisol and cortisol metabolite excretion in chronic fatigue syndrome. Psychosom Med 2006;68:578-82.
  70. Crofford, LJ, Young EA, Engleberg NC, et al. Basal circadian and pulsatile ACTH and cortisol secretion in patients with fibromyalgia and/or chronic fatigue syndrome. Brain, Behav Immun 2004;18:314-25.
  71. Jerjes WK, Peters TJ, Taylor NF, et al. Diurnal excretion of urinary cortisol, cortisone and cortisol metabolites in chronic fatigue syndrome. J Psychosom Res 2006;60:145-53.
  72. Jerjes WK, Cleare AJ, Wessely S, et al. Diurnal patterns of salivary cortisol and cortisone output in chronic fatigue syndrome. J Affect Disord 2005;87:299-304.
  73. Strickland P, Morriss R, Wearden A, et al. A comparison of salivary cortisol in chronic fatigue syndrome, community depression and healthy controls. J Affect Disord 1998;47:191-4.
  74. Roberts ADL, Wessely S, Chalder T, et al. Salivary cortisol response to awakening in chronic fatigue syndrome. Br J Psych 2004;184:136-41.
  75. Nater UM, Youngblood LS, Jones JF, Unger ER, Miller AH, Reeves WC, Heim C. Alterations in diurnal salivary cortisol rhythm in a population-based sample of cases with chronic fatigue syndrome. Psychosom Med. 2008 Apr;70(3):298-305.
  76. Scott LV, Salahuddin F, Cooney J, et al. Differences in adrenal steroid profile in chronic fatigue syndrome, in depression and in health. J Affect Disord 1999;52:129-37.
  77. Scott LV, The J, Reznek R, et al. Small adrenal glands in chronic fatigue syndrome: a preliminary computed tomography study. Psychoneuroendocrinology 1999;24:759-68.
  78. Cleare AJ, O’Keane V, Miell JP. Levels of DHEA and DHEAS and responses to CRH stimulation and hydrocortisone treatment in chronic fatigue syndrome. Psychoneuroendocrinology 2004;29:724-32.
  79. Cleare AJ, Miell J, Heap E, et al. Hypothalamo-Pituitary-Adrenal Axis Dysfunction in Chronic Fatigue Syndrome, and the Effects of Low-Dose Hydrocortisone Therapy. J Clin Endocrinol Metab 2001;86:3545-54.
  80. Scott LV, Medbak S, Dinan TG. Blunted adrenocorticotropin and cortisol responses to corticotropin-releasing hormone stimulation in chronic fatigue syndrome. Acta Psychiatr Scand 1998;97:450-7.
  81. Inder WJ, Prickett TCR, Mulder RT. Normal opioid tone and hypothalamic-pituitary-adrenal axis function in chronic fatigue syndrome despite marked functional impairment. Clin Endocrinol (Oxf) 2005;62:343-8.
  82. Bearn J, Allain T, Coskeran P, et al. Neuroendocrine responses to d-fenfluramine and insulin-induced hypoglycemia in chronic fatigue syndrome. Biol Psychiatry 1995;37:245-52.
  83. Scott LV, Burnett F, Medbaks S, et al. Naloxone-mediated activation of the hypothalamic-pituitary-adrenal axis in chronic fatigue syndrome. Psychol Med 1998;28:285-93.
  84. Nater UM, Maloney E, Boneva RS, Gurbaxani BM, Lin JM, Jones JF, Reeves WC, Heim C. Attenuated morning salivary cortisol concentrations in a population-based study of persons with chronic fatigue syndrome and well controls. J Clin Endocrinol Metab. 2008;93:703-9.
  85. DiGiorgio A, Hudson M, Jerjes W, et al. 24-hour pituitary and adrenal hormone profiles in chronic fatigue syndrome. Psychosom Med 2005;67:433-40.
  86. Jerjes WK, Taylor NF, Wood PJ, et al. Enhanced feedback sensitivity to prednisolone in chronic fatigue syndrome. Psychoneuroendocrinology 2007;32:192-8.
  87. Wyller VB, Godang K, Mørkrid L, Saul JP, Thaulow E, Walløe L. Abnormal thermoregulatory responses in adolescents with chronic fatigue syndrome: relation to clinical symptoms. Pediatrics. 2007;120:e129-37.
  88. Wyller VB, Saul JP, Amlie JP, Thaulow E. Sympathetic predominance of cardiovascular regulation during mild orthostatic stress in adolescents with chronic fatigue. Clin Physiol Funct Imaging. 2007;27:231-8.
  89. Cleare AJ, Heap E, Malhi GS, et al. Low-dose hydrocortisone in chronic fatigue syndrome: a randomized crossover trial. Lancet 1999;353:455-8.
  90. McKenzie R, O’Fallon A, Dale J, et al. Low-Dose Hydrocortisone for Treatment of Chronic Fatigue Syndrome. A randomized controlled trial. JAMA 1998;280:1061-6.
  91. Blockmans D, Persoons P, Van Houdenhove B, et al. Combination Therapy with Hydrocortisone and Fludrocortisone Does Not Improve Symptoms in Chronic Fatigue Syndrome: A Randomised Placebo-Controlled, Double-blind Crossover Study. Am J Med 2003;114:736-41.
  92. Peterson PK, Pheley A, Schroeppel J, et al. A preliminary placebo-controlled trial of fludrocortisone for chronic fatigue syndrome. Arch Intern Med 1998;158:908-14.
  93. Roberts AD, Charler ML, Papadopoulos A, Wessely S, Chalder T, Cleare AJ. Does hypocortisolism predict a poor response to cognitive behavioral therapy in chronic fatigue syndrome? Psychol Med. 2010;40:515-22.
  94. Centers for Disease Control and Prevention (CDC). COVID-19: Interim Clinical Guidance for Management of Patients with Confirmed Coronavirus Disease (COVID-19). Available at: http://www.cdc.gov/coronavirus/2019-ncov/hcp/clinical-guidance-managemnet-patients.html. Accessed on 9th July 2023.
  95. Soriano JB, Murthy S, Marshall JC, et al. WHO Clinical Case Definition Working Group on Post-COVID-19 Condition. Lancet Infct Dis. 2022; 22(4)e102. Epub 2021.
  96. Townsend L, et al. Persistent fatigue following SARS-CoV-2 infection is common and independent of severity of initial infection. JAMA Network Open. 2021;4(3):e211641.
  97. Perrin R, et al. Symptoms and signs of COVID-19: A systematic review and meta-analysis. BMJ Open. 2021;11(1):e044327.
  98. National Institute for Health and Care Excellence. Myalgic encephalomyelitis (of encephalopathy)/chronic fatigue syndrome: diagnosis and management. NICE guideline. 2021.
  99. Cluff LE. Medical aspects of delayed convalescence. Rev Infect Dis. 1991 Jan-Feb;13 Suppl 1:S138-40.
  100. Smith ME, Haney E, McDonagh M, et al. Treatment of Myalgic Encephalomyelitis/Chronic Fatigue Syndrome: A systematic Review for a National institutes of Health Pathways to Prevention Workship. Annals of Intern Med. 2015; 162 (12): 841-850.
  101. Mitchell WM. Efficacy of rintatolimod in the treatment of chronic fatigue syndrome/myalgic encephalomyelitis (CFS/ME). Expert Rev Clin Pharmacol 2016;9(6):755-70.
  102. Strayer DR, Carter WA, Stouch BC, et al. A double-blind, placebo-controlled, randomized, clinical trial of the TLR-3 agonist rintatolimod in severe cases of chronic fatigue syndrome.PLoS One. 2012; 7 (3):e31334.
  103. Castro-Marrero J, Saez-Francas N, Santillo D, et al. Treatment and management of chronic fatigue syndrome/myalgic encephalitis: all roads lead to Rome. Brit Jour of Pharm. 2017; 174: 345-69.
  104. Fludge Ø, Bruland O, Risa K, et al. Benefit from B-lymphocyte depletion using the anti-CD20 antibody rituximab in chronic fatigue syndrome. A double-blind and placebo-controlled study. PLoS One. 2011; 6(10).
  105. Fluge Ø, Rekeland IG, Lien K, et al. B-lymphocyte Depletion in Patients with Myalgic Encephalomyelitis/Chronic Fatigue Syndrome: A Randomized, Double-Blind, Placebo-Controlled Trial. Ann Intern Med. 2019 May 7;170(9):585-593.
  106. Castro-Marrero J, Cordero MD, Segundo MJ, Saez-Francas N, Calvo N, Roman-Malo L, et al. Does oral coenzyme Q10 plus NADH supplementation improve fatigue and biochemical parameters in chronic fatigue syndrome? Antioxid Redox Signal 2015;22(8):679-85.
  107. Lightfoot RW Jr, Luft BJ, Rahn DW, et al. Empiric parenteral antibiotic treatment of patients with fibromyalgia and fatigue and a positive serologic result for Lyme disease. A cost-effectiveness analysis. Ann Intern Med 1993; 119:503.
  108. Roerink ME, Bredie SJH, Heijnen M, et al. Cytokine Inhibition in Patients With Chronic Fatigue Syndrome: A Randomized Trial. Ann Intern Med 2017; 166:557.
  109. Blacker CV, Greenwood DT, Wesnes KA, et al. Effect of galantamine hydrobromide in chronic fatigue syndrome: a randomized controlled trial. JAMA 2004; 292:1195.
  110. McKenzie R, O'Fallon A, Dale J, et al. Low-dose hydrocortisone for treatment of chronic fatigue syndrome: a randomized controlled trial. JAMA 1998; 280:1061.
  111. Cleare AJ, Heap E, Malhi GS, et al. Low-dose hydrocortisone in chronic fatigue syndrome: a randomized crossover trial. Lancet 1999; 353:455.
  112. Blockmans D, Persoons P, Van Houdenhove B, Bobbaers H. Does methylphenidate reduce the symptoms of chronic fatigue syndrome? Am J Med 2006; 119:167.e23.
  113. Randall DC, Cafferty FH, Shneerson JM, et al. Chronic treatment with modafinil may not be beneficial in patients with chronic fatigue syndrome. J Psychopharmacol 2005; 19:647.
  114. Ghali A, Lacout C, Fortrat JO, et al. Factors Influencing the Prognosis of Patients with Myalgic Encephalomyelitis/Chronic Fatigue Syndrome. Diagnostics (Basel). 2022 Oct 19;12(10):2540.
  115. Sharpe M, Hawton K, Seagroatt Vet al. Follow up of patients presenting with fatigue to an infectious diseases clinic. BMJ. 1992 Jul 18;305(6846):147-52.
  116. Straus SE, Dale JK, Tobi M, Lawley T, Preble O, Blaese RM, Hallahan C, Henle W. Acyclovir treatment of the chronic fatigue syndrome. Lack of efficacy in a placebo-controlled trial. N Engl J Med. 1988 Dec 29;319(26):1692-8. 
  117. Peterson P.K., Schenck C.H., Sherman R. Chronic Fatigue Syndrome in Minnesota. Minn. Med. 1991;74:21–26.
  118. Wilson A, Hickie I, Lloyd A, et al. Longitudinal Study of Outcome of Chronic Fatigue Syndrome. BMJ. 1994;308:756–759.
  119. Bombardier CH, Buchwald D. Outcome and Prognosis of Patients With Chronic Fatigue vs Chronic Fatigue Syndrome. Arch. Intern. Med. 1995;155:2105–2110.
  120. Vercoulen JH, Swanink CM, Fennis JF, et al. Prognosis in Chronic Fatigue Syndrome: A Prospective Study on the Natural Course. J. Neurol. Neurosurg. Psychiatry. 1996;60:489–494.
  121. van der Werf SP, de Vree B, Alberts M, et al. Natural Course and Predicting Self-Reported Improvement in Patients with Chronic Fatigue Syndrome with a Relatively Short Illness Duration. J. Psychosom. Res. 2002;53:749–753.
  122. Cairns R, Hotopf M. A Systematic Review Describing the Prognosis of Chronic Fatigue Syndrome. Occup. Med. 2005;55:20–31.
  123. Collin SM, Crawley E. Specialist Treatment of Chronic Fatigue Syndrome/ME: A Cohort Study among Adult Patients in England. BMC Health Serv. Res. 2017;17:488.
  124. Clark MR, Katon W, Russo J, et al. Chronic Fatigue: Risk Factors for Symptom Persistence in a 2 1/2-Year Follow-up Study.  J. Med. 1995;98:187–195.
  125. Tiersky LA, DeLuca J, Hill N, et al. Longitudinal Assessment of Neuropsychological Functioning, Psychiatric Status, Functional Disability and Employment Status in Chronic Fatigue Syndrome.  Neuropsychol. 2001;8:41–50.
  126. Ciccone D.S., Chandler H.K., Natelson B.H. Illness Trajectories in the Chronic Fatigue Syndrome: A Longitudinal Study of Improvers versus Non-Improvers.  Nerv. Ment. Dis. 2010;198:486–493.
  127. Joyce J, Hotopf M, Wessely S. The prognosis of chronic fatigue and chronic fatigue syndrome: a systematic review. Q J Med 1997;90:223-33.
  128. Roberts E, Wessely S, Chalder T, et al. Mortality of people with chronic fatigue syndrome: a retrospective cohort study in England and Wales from the South London and Maudsley NHS Foundation Trust Biomedical Research Centre (SLaM BRC) Clinical Record Interactive Search (CRIS) Register. Lancet. 2016 Apr 16;387(10028):1638-43.

 

 

 

 

Growth hormone Stimulation Tests in Assessing Adult Growth Hormone Deficiency

ABSTRACT

 

Adult growth hormone deficiency (GHD) is a clinical syndrome that can manifest either as isolated or associated with additional pituitary hormone deficiencies. Its clinical features are subtle and nonspecific, requiring GH stimulation testing to arrive at a correct diagnosis. However, diagnosing adult GHD can be challenging due to the episodic and pulsatile endogenous GH secretion, concurrently modified by age, gender, and body mass index. Hence, a GH stimulation test is often required to establish the diagnosis, and should only be considered if there is a clinical suspicion of GHD and the intention to treat if the diagnosis is confirmed. Currently, there is no ideal stimulation test and the decision to perform a GH stimulation test must factor in the validity of the chosen test, the appropriate GH cut-points, and the availability of local resources and expertise. For now, the insulin tolerance test remains the gold standard test, while the glucagon stimulation test and macimorelin test are reasonable alternatives to the insulin tolerance test, whereas the arginine test is no longer recommended because arginine is a poor GH secretagogue that requires a very low peak GH cut-point of 0.4 μg/L. In this chapter, we discuss published evidence of the GH stimulation tests used in the United States and the inherent caveats and limitations of each individual test. We propose utilizing the lower GH cut-point to 1mg/L for the glucagon stimulation test to improve its diagnostic accuracy in some overweight and all obese patients based on the clinical suspicion of having adult GHD, and summarize current knowledge and change of status of availability of the oral macimorelin test in the United States.

 

INTRODUCTION

 

Physiological growth hormone (GH) secretion from the anterior pituitary gland is episodic, pulsatile, and accounts for > 85% of total daily GH secretion (1). Due to its pulsatility, serum GH levels vary between peaks and troughs, with very low levels between pulses. Hypothalamic growth hormone–releasing hormone (GHRH) and somatostatin traverse the hypothalamic–pituitary portal system to stimulate and suppress GH production, respectively, by signaling through specific somatotroph cell-surface G protein-coupled receptors (2), while gastric-derived ghrelin also stimulates GH secretion and synergizes the action of GHRH (3). Additionally, other factors such as gender, nutritional status, sleep patterns, physical activity, and metabolic and hormonal signals from other endocrine glands, including glucocorticoids, thyroid hormones, and sex steroids, also play an important role in modulating day-to-day GH secretion (1). Growth hormone regulates its own secretion by a feedback mechanism that involves other peripheral mediators, such as insulin-like growth factor-I (IGF-I), free fatty acids, glucose, and insulin (4). Peripheral GH actions are primarily mediated through IGF-I synthesized mainly by the liver. Because IGF-I has a longer half-life in the circulation than GH, it is considered to provide an integrated measure of GH secretion. Like GH, serum IGF-I levels decline with aging (5), and tend to be low in obesity (6) and in patients with non-alcoholic fatty liver disease (7) that may overlap with the levels observed in younger GH–deficient patients. Hence, for these reasons, the diagnosis of adult GH deficiency (GHD) cannot be established in most patients by a random single measurement of serum GH or IGF-I level.

 

DIAGNOSIS OF ADULT GH DEFICIENCY: CURRENT PERSPECTIVE

 

Adult GHD is a rare heterogeneous disorder that commonly results from a variety of organic causes, including hypothalamic-pituitary tumors and/or their treatment, head trauma, and infiltrative diseases (8). This condition is characterized by decreased lean body mass and increased fat mass, dyslipidemia, cardiac dysfunction, decreased fibrinolysis and premature atherosclerosis, decreased muscle strength and exercise capacity, decreased bone mineral density, increased insulin resistance, and impaired quality of life (9). Treatment with GH replacement improves many, but not all, of these abnormalities (10, 11). However, due to the high cost of GH replacement (GH costs approximately $18,000 to $30,000 per year depending on the dose and brand used) (12) and concerns of potential long-term safety risks, particularly the development of diabetes mellitus, cancer and tumor recurrence, it is imperative that an accurate biochemical diagnosis is made so that appropriate GH replacement is offered to adults who are GH-deficient, and not for non-approved conditions (e.g., aging and sporting enhancement) (13, 14).

 

For the clinician, establishing the diagnosis of adult GHD is challenging because of the lack of a single biological end-point (e.g., growth failure in children with GHD). Other biochemical measurements like IGF-I, IGF-binding protein-3, or GH secretion over a 24-hour period have shown poor diagnostic value as there is an overlap between healthy and adults with GHD, particularly in adults > 40 years of age (5, 15). Hence, a GH stimulation test is often required to establish the diagnosis, and should only be considered if there is a clinical suspicion of GHD and the intention to treat if the diagnosis is confirmed. Currently, there is no ideal stimulation test as each test has its pros and cons, and the decision to consider performing a GH stimulation test to diagnose adult GHD must factor in the validity of the chosen test and its GH cut-points, and the availability of local resources and expertise.

 

Clinical practice guidelines recommend the evaluation of adult GHD to be based on medical history, clinical findings, and utilizing the appropriate GH stimulation test for biochemical confirmation (8, 16-18). The exception of when GH stimulation testing can be exempted include those with organic hypothalamic-pituitary disease with ≥ 3 pituitary hormone deficiencies and low serum IGF-I levels [< -2.0 standard deviation scores (SDS)] (19), patients with genetic defects affecting the hypothalamic-pituitary axes, and those with hypothalamic-pituitary structural brain defects (8, 16, 18). Evaluation for adult GHD should not be performed in patients with no evidence of a suggestive history, e.g., sellar/parasellar mass lesion or a history of a hypothalamic–pituitary insult, such as surgery, radiation therapy, head trauma, or brain tumor. Conversely, GH stimulation testing should not be performed in patients with commonly encountered, generalized, nonspecific symptoms of weakness, frailty, fatigue, or weight gain, without a history of organic hypothalamic/pituitary disease, as such patients are unlikely to benefit from GH therapy (8, 16, 18). These considerations are important for the clinician when deciding which patients to consider testing for possible adult GHD. 

 

All GH stimulation tests are based on the concept that a GH secretagogue agent acutely stimulates pituitary GH secretion, and peak serum GH levels are detected by sequential blood sampling of serum GH levels after administration of the agent. The desired criteria of an ideal GH stimulation test should include the following: the ability to accurately and reliably differentiate adults with GHD from GH-sufficient individuals, high reproducibility, safety with minimal side-effects, affordability, and short test duration. It should also not be unpleasant to the patient and it should be simple to perform.

 

The insulin tolerance test (ITT) has historically been accepted as the gold-standard test for the assessment of adult GHD provided adequate hypoglycemia (blood glucose <40 mg/dL) is achieved (8, 16, 17). However, multiple drawbacks associated with the ITT hamper its wider use (20), and they include the requirement of close medical supervision by a physician throughout the test, the possibility of inducing severe life-threatening hypoglycemia, and the potential of causing seizures and altered consciousness resulting from neuroglycopenia in certain susceptible sub-populations. This test is also contraindicated in the elderly (> 65 years of age) and in patients who are at risk of and/or with a history of cardio-/cerebrovascular disease and seizures.

 

Finding a reliable alternative to the ITT for the diagnosis of adult GHD has been challenging. When the GHRH-arginine test was available in the United States before EMD Serono discontinued manufacturing the GHRH analog (Geref@) in November 2008 (8, 16, 17),  GHRH-arginine test became the most acceptable alternative to the ITT. Since then, the glucagon stimulation test (GST) has grown in popularity replacing the GHRH-arginine test as the test of choice if the ITT cannot be performed or is contraindicated (21). Previous studies have examined the diagnostic utility of the GST for adult GHD, but these studies have either not taken body mass index (BMI) into consideration (22, 23) or included only controls with normal BMIs (24, 25). Several recent retrospective studies have questioned the diagnostic accuracy of the GST when the GH cut-point of 3mg/L is applied to overweight/obese adults (26-29) and in those with glucose intolerance (28, 29), while Hamrahian et al. (30) demonstrated in a prospective study of 28 patients by comparing the GST to the ITT that a lower GH cut-point of 1 mg/L improved its diagnostic accuracy with a 92% sensitivity and 100% specificity.

 

In this document, we will discuss published evidence of the GH stimulation tests used in the United States and the inherent caveats and limitations of each individual test. The lower GH cut-point of 1 mg/L for the GST should be utilized to improve its diagnostic accuracy in some overweight and all obese patients. We will also summarize current knowledge of the oral macimorelin test as the only approved diagnostic test for adult GHD by the United States Food and Drug Administration (FDA) and the European Medicines Agency, and its change in status of availability in the United States.

 

GENERAL LIMITATIONS AND IMPORTANT CAVEATS WHEN INTERPRETING GH STIMULATION TESTS

 

The responses to all GH stimulation tests show intra-individual variability, and the GH cut-points vary depending on the test used. For the ITT and GST, the cut-points advocated by previous consensus guidelines were 3-5 μg/L and 2.5-3 μg/L, respectively (8, 16). Other GH stimulatory agents such as clonidine, L-DOPA, and arginine are weaker GH secretagogues, and would require very low GH cut-points with utilization of sensitive GH assays to achieve adequate specificity (e.g., arginine of 0.4 μg/L) (31). Hence, these tests are not recommended in the United States (8, 16). Other limitations include the relative lack of validated normative data based on age, gender, BMI, glycemic status, and the paucity of data for specific etiologies of adult GHD that have recently been described, such as traumatic brain injury, subarachnoid hemorrhage, ischemic stroke, and central nervous system infections (32, 33).

 

One of the caveats in interpreting the results of GH stimulation tests is that adult GHD itself is complicated by an increased susceptibility to central obesity (34). Obesity per se is a state of relative GHD (35-40), and earlier physiologic studies in obese individuals have shown that spontaneous GH secretion is reduced, GH clearance is enhanced, and stimulated GH secretion is reduced (40-42). Conversely, serum IGF-I levels are unaffected, or even increased, and this discordance is related to the increased hepatic GH responsiveness (43). The decreased serum GH levels in obesity up-regulate GH receptor and sensitivity. Furthermore, non-alcoholic fatty liver disease and non-alcoholic steatohepatitis are now recognized as being highly prevalent in overweight and obese adults with GHD (44), with consequent lower serum IGF-I levels being associated with increased severity of the disease (7). Thus, these data suggest that BMI-specific cut-points should be considered when testing patients for adult GHD. Table 1summarizes the accepted GH cut-points for the GH stimulation tests used in the United States, as recommended by different consensus guidelines.

 

Table 1. Accepted GH Cut-Points (µg/L) for GH Stimulation Tests Used in the United States by Different Consensus Guidelines for Diagnosis of Adult GHD

 

GRS 2007

(17)

 

AACE 2009

(16)

ES 2011

(8)

AACE 2019

(18)

ITT

 

< 3.0

≤ 5.0

< 3.0 to 5.0

≤ 5.0

GHRH-arginine

- BMI < 25 kg/m2

- BMI 25-30 kg/m2

- BMI ≥ 30 kg/m2

 

 

< 11.0

< 8.0

< 4.0

 

≤ 11.0

≤ 8.0

≤ 4.0

 

< 11.0

< 8.0

< 4.0

No recommendation as not commercially available in the United States

Glucagon

- BMI < 25 kg/m2

- BMI 25-30 kg/m2

- BMI ≥ 30 kg/m2

 

 

< 3.0

< 3.0

< 3.0

 

≤ 3.0

≤ 3.0

≤ 3.0

 

< 3.0

< 3.0

< 3.0

 

≤ 3.0

≤ 3.01 or ≤ 1.02

≤ 1.0

Macimorelin

 

Not commercially available in 2007

 

Not commercially available in 2009

 

Not commercially available in 2011

≤ 2.8

Arginine

 

Not recommended to be used

≤ 0.4

 

Not recommended to be used

No longer recommended to be used

 

1GH cut-point of ≤ 3.0 µg/L for patients with a high pre-test probability; 2GH cut-point of ≤ 1.0 µg/L for patients with a low pre-test probability.

AACE, American Association of Clinical Endocrinologists; BMI, body mass index; ES, Endocrine Society; GHRH, growth hormone releasing hormone; GRS, Growth Hormone Research Society; ITT, insulin tolerance test.

 

GROWTH HORMONE STIMULATION TESTS USED IN DIAGNOSING ADULT GH DEFICIENCY

 

Insulin Tolerance Test

 

The ITT remains accepted as the gold standard test for the assessment of adult GHD, with a GH cut-point of 3-5 mg/L when adequate hypoglycemia (blood glucose < 40 mg/dL) is achieved (8, 16, 17). This GH cut-point was originally proposed by Hoffman et al. (45) in 1994 based on GH responses to insulin-induced hypoglycemia, mean 24-hour GH levels derived from 20-min sampling, and serum IGF-I and IGFBP-3 levels in 23 patients considered GH-deficient due to organic pituitary disease, and in 35 sex-matched normal subjects of similar age and BMI. The ranges of stimulated peak GH responses separated GH-deficient (0.2-3.1 mg/L) from GH-sufficient (5.3-42.5 mg/L) patients. However, an overlap in mean 24-hour GH, IGF-I, and IGFBP-3 levels was observed, demonstrating the challenge in utilizing random single serum GH, IGF-I and IGFBP-3 levels to accurately differentiate GH-sufficiency from GHD.

 

Disadvantages of the ITT include the requirement of close medical supervision, may be unpleasant, and cautioned in some patients because of potential adverse effects (e.g., seizures or loss of consciousness resulting from neuroglycopenia), and contraindicated in elderly patients and in patients at risk of and/or with a history of cardio-/cerebrovascular disease and seizures. Furthermore, normoglycemic and/or hyperglycemic obese patients with insulin resistance may fail to achieve adequate hypoglycemia (46), necessitating the use of higher insulin doses (0.15-0.2 IU/kg), thus increasing the risk of delayed hypoglycemia. Although the ITT demonstrates good sensitivity, its reproducibility is another major limitation. Differences in peak GH responses have been demonstrated in healthy subjects undergoing ITT at varying times (47) and in women at different times of their menstrual cycle (48).

 

Table 2. Recommended Protocol for Performing the ITT

CONTRAINDICATIONS:

History of epileptic seizures, coronary artery disease, pregnancy, or age > 55 years.

PRECAUTIONS:

Patients commonly develop neuroglycopenic symptoms during the test and should be encouraged to report these symptoms (administration of IV anti-emetics can be considered).

Late hypoglycemia may occur (patients should be advised to eat small and frequent meals after completion of the test).

PROCEDURE:

Fast from midnight for 8-10 hours.

All morning medications can be taken with water (if the HPA axis is simultaneously assessed, then glucocorticoids should be withheld ≥ 12 hours before testing).

Weigh patient.

1Place IV cannula for IV access in both forearms.

2Administer IV human Regular insulin (standard dose: 0.05-0.1 units/kg for non-diabetic subjects with a BMI < 30 kg/m2 and high dose: 0.15-0.3 units/kg for subjects with a BMI ≥ 30 kg/m2).

SAMPLING AND MEASUREMENTS:

Baseline

Blood is drawn for glucose measurement with a glucometer.

Blood draw for baseline glucose, GH and IGF-I (cortisol and ACTH, if HPA axis is assessed simultaneously) levels will be sent to the laboratory for further analysis.

During the test

Blood samples are drawn from the IV line every 5-10 mins for measurement of glucose levels using a glucometer.

Signs and symptoms of neuroglycopenia are recorded.

When blood glucose levels from the glucometer approaches 45 mg/dL (2.5 mmol/L), blood samples are sent to the laboratory for measurements of blood glucose levels.

When symptomatic hypoglycemia is achieved (laboratory blood glucose < 40 mg/dL or 2.2 mmol/L), additional blood samples are collected to measure glucose and GH (+/- cortisol if the HPA axis is assessed simultaneously) levels at 20, 25, 30, 35, 40, 60 and 90 min.

The patient can begin drinking orange juice and eat to raise his/her blood glucose levels (IV 100 ml of 5% Dextrose can be administered if the patient cannot tolerate oral intake due to nausea or vomiting).

At the end of the test

Blood glucose levels measured from the glucometer should increase to levels > 70 mg/dL (3.9 mmol/L) before the patient is discharged from the testing unit.

INTERPRETATION:

If adequate (symptomatic) hypoglycemia is not achieved (< 40 mg/dL or 2.2 mmol/L), then adult GHD cannot be diagnosed.

Peak serum GH levels ≤ 5 µg/L at any time point during the hypoglycemic phase of the test is diagnostic of adult GHD.

CAUTION:

If adequate (symptomatic) hypoglycemia is not achieved (< 40 mg/dL or 2.2 mmol/L), then adult GHD cannot be diagnosed.

ACTH: adrenocorticotropic hormone, HPA: hypothalamic-pituitary-adrenal, IV: intravenous.

1Two IV lines are placed, one IV line is used for the administration of insulin bolus and possibly for administration of IV 5% Dextrose administration if the patient requires resuscitation from hypoglycemia, while the other IV line is used for repeated blood draws.

2In certain patients with BMIs > 30 kg/m2 who appear muscular with increased insulin sensitivity, clinical discretion is required in deciding the insulin dose for these patients. A dose of 0.05-0.1 units/kg may be more appropriate to prevent severe or delayed hypoglycemia.   

 

Glucagon Stimulation Test

 

Glucagon is reportedly to be more potent than arginine or clonidine in stimulating GH secretion (24, 25). Glucagon is also a more potent GH secretagogue when administered intramuscularly or subcutaneously compared to the intravenous route (49). However, the mechanism/s of glucagon-induced GH stimulation remains unclear, and one hypothesis is that glucagon decreases ghrelin-independent effects of glucose or insulin variations (50).

 

There have been three earlier studies that have assessed the GST in identifying adult GHD in patients with pituitary disorders (22, 23, 51). Gomez et al. (51) and Conceicao et al. (23) compared the diagnostic characteristics of GST to ITT and included a control group matched for age and sex in both studies, and for BMI in one study (51). Using receiver operating characteristic (ROC) analysis, both studies proposed that a GH cut-point of 3 mg/L provided optimal sensitivity and specificity (51, 52). Gomez et al. (51) also demonstrated an inverse correlation between age (R = - 0.389, P = 0.0075) and BMI (R = - 0.329, P = 0.025) with peak GH levels in healthy controls. These data suggest that there is a potential association between relative, but not organic, GHD in aging and obesity. However, this study was conducted in a European cohort, where the frequency and severity of obesity is generally to a lesser degree than in the United States (53). Conversely, Conceicao et al. (23) demonstrated that peak GH levels were unaffected by age in either the control or patient group, and neither were there any gender differences. Additionally, Gomez et al. (51)used intramuscular glucagon doses of 1 mg and 1.5 mg for body weights ≤ 90 kg and > 90 kg respectively, whereas Conceicao et al. (23) used intramuscular glucagon of 1 mg for all subjects. In another study, Berg et al. (22)demonstrated an optimal peak GH cut-point of 2.5 mg/L with 95% sensitivity and 79% specificity using ROC analysis. This study also reported lower peak GH levels with GST compared to ITT (5.1 vs 6.7 mg/L, P < 0.01) and a positive correlation between peak GH levels during ITT and GST (R = 0.88, P < 0.0001), but no correlation between BMI or age to peak GH responses (54, 55). However, these (22, 23, 51) and other earlier studies (24, 25, 49, 56) did not specifically evaluate patients with glucose intolerance; hence, the diagnostic accuracy of the GST in testing for GHD in this population remains unclear.

 

Advantages of the GST is its reproducibility, safety, and lack of influence by gender and hypothalamic GHD (21), whereas disadvantages include the lengthy test duration (3-4 hours), and the need for an intramuscular injection that might not appeal to some patients. Side-effects frequently reported include nausea, vomiting, and headaches ranging from < 10% (22) to 34% (54), mainly occur between 60-210 min and tend to resolve by 240 min into the test, and seem to be more pronounced in elderly subjects, where severe symptomatic hypotension, hypoglycemia, and seizures have been observed (57).    

 

However, since the publication of the 2009 American Association of Clinical Endocrinologists (AACE) (16) and 2011 Endocrine Society (8) Clinical Practice Guidelines, there have been several studies that have suggested that the fixed-dose GST using a GH cut-point of 3 mg/L may potentially over-diagnose adult GHD in a substantial number of overweight/obese subjects and in those with glucose intolerance. In two large retrospective studies, Toogood et al.(58) and Yuen et al. (29) found an inverse correlation between BMI and peak GH during the GST, and that this relationship appeared to be strongest with BMIs between 30 and 40 kg/m2 and seemed to plateau for those with BMIs > 40 kg/m2 (58). Alternatively, a negative correlation between BMI and peak GH following glucagon stimulation has been reported by Gomez et al. (51) in healthy subjects but not in patients with underlying pituitary disease. Dichtel et al. (26) evaluated 3 groups of overweight/obese men, i.e., controls who were younger than the patients, patients with 3-4 pituitary hormone deficits, and patients with 1-2 pituitary hormone deficits. Using ROC analysis, the GH cut-point of 0.94 mg/L provided the optimal sensitivity (90%) and specificity (94%), whereas BMI and amount of visceral adipose tissue inversely correlated with peak GH levels in controls. Almost half of the healthy overweight/obese individuals (45%) failed the GST using the 3 mg/L GH cut-point. Diri et al. (27) evaluated 216 patients with pituitary disease and 26 healthy controls and compared the GST to the ITT. These investigators used a GH cut-point of 3.0 mg/L for the ITT and two GH cut-points of 3.0 mg/L and 1.07 mg/L for the GST, yielding the diagnosis of adult GHD in 86.1%, 74.5%, and 54.2 % patients, respectively. Additionally, patient age, BMI, and number of pituitary hormone deficits correlated with IGF-I and peak GH levels. Twelve out of 26 (46.2 %) healthy subjects failed the GST using a GH cut-point of 3.0 mg/L, but none when the cut-point was lowered to 1.07 mg/L. Wilson et al. (28) studied 42 patients with a high pre-test probability of adult GHD. After excluding 10 patients with severe GHD based on peak GH levels ≤ 0.1 mg/L, these investigators found that body weight negatively correlated with GH area under the curve (AUC) (R = -0.45; P = 0.01) and peak GH response (R = -0.42; P = 0.02) and positively correlated with nadir blood glucose levels (R = 0.48; P < 0.01). Conversely, nadir blood glucose levels during GSTs inversely correlated with GH AUC (r= -0.38; p=0.03) and peak GH (r= -0.37; p=0.04), implying that patients with higher nadir blood glucose levels tended to have a lesser glucagon-induced GH response. Recently, Hamrahian et al. (30) compared the fixed-dose GST (1 mg or 1.5 mg in patients > 90 kg body weight) and weight-based GST (WB-GST: 0.03 mg/kg) with the ITT using a GH cut-point of 3.0 mg/L. Patients with hypothalamic-pituitary disease and 1-2 (n = 14) or ≥ 3 (n = 14) pituitary hormone deficiencies, and control subjects (n = 14) matched for age, sex, estrogen status and BMI undertook the ITT, GST and WB-GST in random order. Using ROC analyses, the optimal GH cut-point was 1.0 (92% sensitivity, 100% specificity) for fixed-dose GST and 2.0 mg/L (96% sensitivity and 100% specificity) for WB-GST. Therefore, lowering the GH cut-point from 3 mg/L to 1 mg/L is important to reduce misclassifying adult GHD in overweight (BMI 25-30 kg/m2) patients with a low pre-test probability and in obese (BMI > 30 kg/m2) patients.

 

It remains unclear whether hyperglycemia influences peak GH responses to glucagon stimulation, independent of central adiposity. No peak GH responses have been studied using the GST in normal controls > 70 years of age, and none of the previous studies included patients with poorly controlled diabetes mellitus. Studies by Yuen et al. (29) and Wilson et al. (28) demonstrated that higher fasting (range 90-316 mg/dL), peak (range 156-336 mg/dL), and nadir (range 52-200 mg/dL) blood glucose levels during the GST were associated with lower peak GH responses. Therefore, stratification of GH responsiveness by the degree of glycemia will be helpful to clinicians in interpreting the GST results in patients with impaired glucose tolerance and diabetes mellitus. Because these data are currently unavailable, caution should be exercised when interpreting abnormal GST results in these patients. Further larger prospective studies are needed to address the effects of varying degrees of hyperglycemia on the ability of glucagon to stimulate GH secretion.

 

Table 3.  Recommended Protocol for Performing the Glucagon Stimulation Test

CONTRAINDICATIONS:

Malnourished patients or patients who have not eaten for > 48 hours.

Severe fasting hyperglycemia > 180 mg/dL.

PRECAUTIONS:

Patients may feel nauseous during the test (administration of IV anti-emetics may be considered).

Late hypoglycemia may occur (patients should be advised to eat small and frequent meals after completion of the test).

PROCEDURE:

Fast from midnight for 8-10 hours.

All morning medications can be taken with water.

Weigh patient.

Place IV cannula for IV access in one forearm.

Administer IM glucagon (1.0 mg if patient body weight ≤ 90 kg and 1.5 mg if patient body weight > 90 kg).

SAMPLING AND MEASUREMENTS:

Blood is drawn for measurements of serum GH1 and blood glucose2 levels at 0, 30, 60, 90, 120, 150, 180, 210 and 240 mins.

INTERPRETATION:

Peak GH levels ≤ 3.0 µg/L in normal-weight (BMI < 25 kg/m2) patients and in

overweight (BMI 25-30 kg/m2) patients with a high pre-test probability, and ≤ 1.0 ug/L in

overweight (BMI 25-30 kg/m2) patients with a low pre-test probability and in obese (BMI >

30 kg/m2) patients at any time point during testing are diagnostic of adult GHD.

CAUTION:

Clinical suspicion of pre-test probability should be taken into consideration when interpreting GST results in patients > 70 years of age and in patients with impaired glucose tolerance and poorly controlled diabetes mellitus, as no peak GH responses have been studied in these patients.

IM: intramuscular, IV: intravenous.

1Serum GH: peak GH levels tend to occur between 120-180 mins; 2blood glucose: usually peaks around 90 mins and then gradually declines (not a requirement to interpret the test).

 

Macimorelin Test

 

Growth hormone secretagogues (GHSs) are peptidyl (GH-releasing peptide [GHRP]) and nonpeptidyl molecules that exert strong dose-dependent and specific stimulatory effects on the animal and human somatotrope secretion (59). These agents act as functional somatostatin antagonists by binding to their specific GH secretagogue receptor-1a in the hypothalamus and pituitary. The natural ligand for this receptor is the gut peptide ghrelin (60). Growth hormone secretagogues are now considered as ghrelin mimetic agents and can be administered parenterally (e.g., GHRP-2, GHRP-6, hexarelin) or orally (e.g., MK-677 and macimorelin).

 

Macimorelin (formerly known as AEZS-130, ARD-07, and EP-01572) is a novel GH secretagogue that binds the GHS-R1a receptor and to pituitary and hypothalamic extracts with a similar affinity to ghrelin (61). In healthy volunteers, it is readily absorbed with good stability and oral bioavailability, and effectively stimulates endogenous GH secretion (61). An open-label, crossover, multicenter trial examined the diagnostic accuracy of a single oral dose of macimorelin (0.5 mg/kg) compared to GHRH plus arginine in adults with GHD and healthy matched controls (62). Peak GH levels were 2.36 ± 5.69 and 17.71 ± 19.11 mg/L in adults with GHD and healthy controls, respectively, with optimal GH cut-points ranging between 2.7 and 5.2 mg/L (62). Macimorelin showed good discrimination comparable to GHRH plus arginine, with peak GH levels that were inversely associated with BMI in controls. In a recent multicenter, open-label, randomized, two-way crossover study, oral macimorelin was compared to the ITT to validate its use for the diagnosis of adult GHD (63). The GH cut-point levels of 2.8 mg/L for macimorelin and 5.1 mg/L for ITT provided 95.4% (95% CI, 87% to 99%) negative agreement, 74.3% (95% CI, 63% to 84%) positive agreement, 87% sensitivity, and 96% specificity. In both studies (62, 63), macimorelin was well-tolerated, reproducible, and safe. In December 2017, the United States FDA approved macimorelin for use as a diagnostic test for adult GHD and mandated the GH cut-point of2.8 mg/L to be used to differentiate patients with normal GH secretion from those with GHD. However, in the study by Garcia et al. (63), when the GH cut-point was increased to 5.1 mg/L for both macimorelin and ITT, negative agreement and specificity was unchanged at 94% (95% CI, 85% to 98%) and 96%, respectively, but interestingly, positive agreement and sensitivity was higher at 82% (95% CI, 72% to 90%) and 92%. Because measured serum GH levels are dependent on the GH assays used, using the GH cut-point of 5.1 mg/L for macrimorelin that is identical to the cut-point accepted for the ITT could be considered in patients with peak serum GH levels between 2.8 mg/L to 5.1 mg/L, especially if the patient has a high pre-test probability, e.g., history of surgery on a sellar/parasellar mass with 1-2 other pituitary hormone deficiencies. It is important to note that this test is not affected by age, BMI, or sex indicating its robustness for diagnosing adult GHD (64).

 

Main advantages of macimorelin are that the drug is orally administered, unlike the ITT, GHRH plus arginine or GST, that requires intravenous or intramuscular administration, and no risk of causing hypoglycemia. In addition, the test only lasts 90 minutes with 3-4 blood sample collections required, in contrast to more blood sample collections over 2 hours for the ITT and 3-4 hours for the GST. The most commonly reported side effect was mild dysgeusia, which did not require any intervention and resolved spontaneously (63). One drug-related serious adverse event was reported; that was in a subject with an asymptomatic QT interval prolongation on the electrocardiogram that resolved spontaneously within 24 h (62). Thus, careful assessment of the patient’s concurrent medications is recommended as well as discontinuation of strong CYP3A4 inducers, provided this is considered safe by the prescribing physician and with sufficient washout time prior to testing.

 

However, in August 2022, a press announcement stated that Novo Nordisk Healthcare AG provided a 270-day notice period to terminate the amended development and commercialization license agreement for macimorelin (MacrilenÒ) in the United States (65). This means that as of May 23, 2023, Aerterna Zentaris regained its full rights in the United States and Canada to macimorelin but because it has yet to find a partner in the United States to market macimorelin, it was further announced that sales of macimorelin will be temporarily discontinued and use of the agent beyond May 2023 will continue until its supplies in the United States runs out (66).

 

Table 4.  Recommended Protocol for Performing the Macimorelin Test

CONTRAINDICATIONS:

Drugs that may increase its plasma levels and prolong QT.

PRECAUTIONS:

Dysgeusia.

PROCEDURE:

Fast from midnight for 8-10 hours.

All morning medications can be taken with water.

Weigh patient.

Place IV cannula for IV access in one forearm.

Dissolve in water 1 (120 ml) or 2 pouches (240 ml) of macimorelin (≤ 120 kg = 1 pouch; > 120 kg = 2 pouches)

Calculate macimorelin dose (0.5 mg/kg as a single oral dose) and volume of water required to reconstitute macimorelin solution (patient body weight X kg = X ml macimorelin solution, e.g., patient with a body weight of 70 kg would require 70 mL of reconstituted macimorelin solution)

After volume of macimorelin is calculated, stir the solution gently and thoroughly for 2-3 min, and use within 30 min of preparation.

Draw the exact macimorelin volume of solution into a needleless syringe, transfer the exact volume of into a drinking glass, and instruct the patient to drink the entire volume of solution within 30 seconds.

SAMPLING AND MEASUREMENTS:

Blood is drawn for measurements of serum GH levels at 30, 45, 60 and 90 min.

INTERPRETATION:

Peak serum GH levels tend to occur between 45-60 mins.

When used according to prescribing package label, peak GH levels ≤ 2.8 µg/L at any time point is diagnostic of adult GHD.

CAUTION:

Peak GH levels ≤ 5.1 µg/L at any time point may be considered in patients with a high-pre-test probability to diagnose adult GHD, as this higher GH cut-point limits the risk of a false-positive diagnosis and maintains a high detection rate for GH-deficient patients because of the more potent GH stimulatory effect of macimorelin compared with the ITT.

Safety and diagnostic performance in patients < 18 and > 65 years of age, and in patients with impaired glucose tolerance and poorly controlled diabetes mellitus, and BMI-adjusted peak GH cut-points for overweight and obese patients is not established.

 

Summary of Tests

 

Table 5 displays a summary of the desirable test characteristics of GH stimulation tests currently available in the United States.

 

Table 5. Summary of Desirable Test Characteristics of each GH Stimulation Test Currently Available in the United States

Test

Accurate?

Safe?

Tolerability?

Simple?

Quick?

Available?

Cost

ITT

Gold standard

No2

No4

No

No

Yes

$

GST

Yes1

Yes3

No3

Yes

No

Yes

$

Macimorelin

Yes

Yes

Yes

Yes

Yes

Yes/No

$$$

1if appropriate BMI-specific GH cut-points are used; 2contraindicated in patients with a history hypoglycemia, history of previous seizures, in the elderly (> 65 years of age), and in patients at risk of and/or with a history of cardio-/cerebrovascular disease; 3caution in patients with propensity for nausea and vomiting, and elderly patients who may be at risk of developing symptomatic hypotension and dizziness (57); 4patients may not tolerate severe symptomatic hypoglycemia. GST, glucagon stimulation test; ITT, insulin tolerance test.

 

STANDARDIZATION OF GH ASSAYS

 

Accurate measurement of GH levels is critical for establishing the diagnosis of adult GHD because the analytical method influences the results of GH stimulation tests, which is dependent on specific GH cut-point levels. However, circulating GH is present in several different isoforms and isomers, including the most common variant of 22 kDa, and other smaller molecules, such as the 20 kDa GH variant. Monoclonal antibodies binding to a specific molecular form of GH are used to limit detection to the 22 kDa GH, but will not detect other GH isoforms. Other molecules similar to GH (e.g., placental GH and prolactin) could potentially cross-react and affect the measurement of GH levels. Growth hormone binding protein, to which approximately 50% of circulating GH is bound, can also cause interference in a GH assay. Furthermore, substantial heterogeneity exists among currently utilized assays due to the use of different standard preparations for calibration of GH immunoassays, and lack of harmonization between various GH assays makes it difficult to directly compare diagnostic cut-points across different published studies. Another source of confusion when interpreting data of GH stimulation tests was that some laboratories reported GH levels in activity (mU/L), whereas others used mass units (mg/L) (67).

 

Due to the heterogeneity of GH assays, it is important that GH assays utilize a universal GH calibration standard 98/574 (National Institute for Biological Standards and Control), a recombinant pituitary GH preparation of high purity (68). All assay manufacturers should also specify the validation of their assay, which should include specification of the GH isoforms detected (20 kDa GH, 22 kDa GH, and other isoforms), the analyte being measured, the specificities of the antibodies used, and the presence or absence of growth hormone binding protein interference.

 

CONCLUSIONS

 

The decision to perform GH stimulation tests should be based on the clinical suspicion of the treating endocrinologist. If the clinical suspicion is high, such as in a patient with history of surgery on a sellar mass, concurrent 1-2 other pituitary hormone deficiencies, and a low (< -2 SDS) or low-normal (< 0 SDS) serum IGF-I level, then performing GH stimulation testing is recommended. If the clinical suspicion is low, such as in cases where there is no suggestive history, such as hypothalamic-pituitary disease, surgery or radiation therapy, head trauma, or childhood-onset GHD, then the diagnosis of adult GHD should not be pursued and GH stimulation testing should not be performed. For now, the ITT remains the gold standard GH stimulation test, and the GST and macimorelin test (where available) are reasonable alternatives to the ITT. As the reliability of the GST GH cut-point of 3 mg/L in overweight/obese subjects and in those with glucose intolerance can misclassify some patients, the utilization of GH cut-points of the GST is now based on the clinician’s level of suspicion of the patient’s pre-test probability and underlying BMI. Macimorelin, a drug administered orally that was approved by the United States FDA in December 2017 is an attractive test because it is easy to conduct with high reproducibility, safe, and has comparable diagnostic accuracy to the ITT and GHRH plus arginine test. The factors that limit its wider is its high cost (one 60 mg macimorelin packet costs approximately $4,500) (69) and the potential of drug-to-drug interactions that may cause QT prolongation. Following the announcement in August 2022 that macimorelin will be temporarily discontinued in the commercial market effective May 2023, after supplies of macimorelin runs out in the United States, the ITT and GST will only be the two GH stimulation tests available to clinicians, limiting the choices of tests that can be used.

 

REFERENCES

 

  1. Gunawardane K, Krarup Hansen T, Sandahl Christiansen J, Lunde Jorgensen JO. Normal physiology of growth hormone in adults. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, Dungan K, Grossman A, et al., editors. Endotext. South Dartmouth (MA) 2000.
  2. Hartman ML, Veldhuis JD, Thorner MO. Normal control of growth hormone secretion. Horm Res. 1993;40(1-3):37-47.
  3. Casanueva FF, Camina JP, Carreira MC, Pazos Y, Varga JL, Schally AV. Growth hormone-releasing hormone as an agonist of the ghrelin receptor GHS-R1a. Proc Natl Acad Sci U S A. 2008;105(51):20452-7.
  4. Muller EE, Locatelli V, Cocchi D. Neuroendocrine control of growth hormone secretion. Physiol Rev. 1999;79(2):511-607.
  5. Hilding A, Hall K, Wivall-Helleryd IL, Saaf M, Melin AL, Thoren M. Serum levels of insulin-like growth factor I in 152 patients with growth hormone deficiency, aged 19-82 years, in relation to those in healthy subjects. J Clin Endocrinol Metab. 1999;84(6):2013-9.
  6. Galli G, Pinchera A, Piaggi P, Fierabracci P, Giannetti M, Querci G, et al. Serum insulin-like growth factor-1 concentrations are reduced in severely obese women and raise after weight loss induced by laparoscopic adjustable gastric banding. Obes Surg. 2012;22(8):1276-80.
  7. Sumida Y, Yonei Y, Tanaka S, Mori K, Kanemasa K, Imai S, et al. Lower levels of insulin-like growth factor-1 standard deviation score are associated with histological severity of non-alcoholic fatty liver disease. Hepatol Res. 2015;45(7):771-81.
  8. Molitch ME, Clemmons DR, Malozowski S, Merriam GR, Vance ML. Evaluation and treatment of adult growth hormone deficiency: an Endocrine Society clinical practice guideline. J Clin Endocrinol Metab. 2011;96(6):1587-609.
  9. Simpson H, Savine R, Sonksen P, Bengtsson BA, Carlsson L, Christiansen JS, et al. Growth hormone replacement therapy for adults: into the new millennium. Growth Horm IGF Res. 2002;12(1):1-33.
  10. Jorgensen AP, Fougner KJ, Ueland T, Gudmundsen O, Burman P, Schreiner T, et al. Favorable long-term effects of growth hormone replacement therapy on quality of life, bone metabolism, body composition and lipid levels in patients with adult-onset growth hormone deficiency. Growth Horm IGF Res. 2011;21(2):69-75.
  11. Widdowson WM, Gibney J. The effect of growth hormone replacement on exercise capacity in patients with GH deficiency: a meta-analysis. J Clin Endocrinol Metab. 2008;93(11):4413-7.
  12. Cook D, Owens G, Jacobs M. Human growth hormone treatment in adults: balancing economics and ethics. Am J Manag Care. 2004;10(13 Suppl):S417-9.
  13. Bartke A. Growth hormone and aging: updated review. World J Mens Health. 2019;37(1):19-30.
  14. Siebert DM, Rao AL. The use and abuse of human growth hormone in sports. Sports Health. 2018:1941738118782688.
  15. Yuen KC, Cook DM, Sahasranam P, Patel P, Ghods DE, Shahinian HK, et al. Prevalence of GH and other anterior pituitary hormone deficiencies in adults with nonsecreting pituitary microadenomas and normal serum IGF-1 levels. Clin Endocrinol (Oxf). 2008;69(2):292-8.
  16. Cook DM, Yuen KC, Biller BM, Kemp SF, Vance ML, American Association of Clinical E. American Association of Clinical Endocrinologists medical guidelines for clinical practice for growth hormone use in growth hormone-deficient adults and transition patients - 2009 update. Endocr Pract. 2009;15 Suppl 2:1-29.
  17. Ho KKY on behalf of the 2007 GH Deficiency Consensus Workshop Participants. Consensus guidelines for the diagnosis and treatment of adults with GH deficiency II: a statement of the GH Research Society in association with the European Society for Pediatric Endocrinology, Lawson Wilkins Society, European Society of Endocrinology, Japan Endocrine Society, and Endocrine Society of Australia. Eur J Endocrinol. 2007;157(6):695-700.
  18. Yuen KCJ (chair of the 2019 AACE Growth Hormone Task Force), Biller BMK, Radovick S, Carmichael JD, Jasim S, Pantalone KM, et al. American Association of Clinical Endocrinologists and American College of Endocrinology Guidelines for management of growth hormone deficiency in adults and patients transitioning from pediatric to adult care. Endocr Pract. 2019 Nov;25(11):1191-1232.
  19. Hartman ML, Crowe BJ, Biller BM, Ho KK, Clemmons DR, Chipman JJ. Which patients do not require a GH stimulation test for the diagnosis of adult GH deficiency? J Clin Endocrinol Metab. 2002;87(2):477-85.
  20. Gordon MB, Levy RA, Gut R, Germak J. Trends in growth hormone stimulation testing and growth hormone dosing in adult growth hormone deficiency patients: results from the ANSWER Program. Endocr Pract. 2016;22(4):396-405.
  21. Yuen KC, Biller BM, Molitch ME, Cook DM. Clinical review: Is lack of recombinant growth hormone (GH)-releasing hormone in the United States a setback or time to consider glucagon testing for adult GH deficiency? J Clin Endocrinol Metab. 2009;94(8):2702-7.
  22. Berg C, Meinel T, Lahner H, Yuece A, Mann K, Petersenn S. Diagnostic utility of the glucagon stimulation test in comparison to the insulin tolerance test in patients following pituitary surgery. Eur J Endocrinol. 2010;162(3):477-82.
  23. Conceicao FL, da Costa e Silva A, Leal Costa AJ, Vaisman M. Glucagon stimulation test for the diagnosis of GH deficiency in adults. J Endocrinol Invest. 2003;26(11):1065-70.
  24. Aimaretti G, Baffoni C, DiVito L, Bellone S, Grottoli S, Maccario M, et al. Comparisons among old and new provocative tests of GH secretion in 178 normal adults. Eur J Endocrinol. 2000;142(4):347-52.
  25. Rahim A, Toogood AA, Shalet SM. The assessment of growth hormone status in normal young adult males using a variety of provocative agents. Clin Endocrinol (Oxf). 1996;45(5):557-62.
  26. Dichtel LE, Yuen KC, Bredella MA, Gerweck AV, Russell BM, Riccio AD, et al. Overweight/obese adults with pituitary disorders require lower peak growth hormone cutoff values on glucagon stimulation testing to avoid overdiagnosis of growth hormone deficiency. J Clin Endocrinol Metab. 2014;99(12):4712-9.
  27. Diri H, Karaca Z, Simsek Y, Tanriverdi F, Unluhizarci K, Selcuklu A, et al. Can a glucagon stimulation test characterized by lower GH cut-off value be used for the diagnosis of growth hormone deficiency in adults? Pituitary. 2015;18(6):884-92.
  28. Wilson JR, Utz AL, Devin JK. Effects of gender, body weight, and blood glucose dynamics on the growth hormone response to the glucagon stimulation test in patients with pituitary disease. Growth Horm IGF Res. 2016;26:24-31.
  29. Yuen KC, Biller BM, Katznelson L, Rhoads SA, Gurel MH, Chu O, et al. Clinical characteristics, timing of peak responses and safety aspects of two dosing regimens of the glucagon stimulation test in evaluating growth hormone and cortisol secretion in adults. Pituitary. 2013;16(2):220-30.
  30. Hamrahian AH, Yuen KC, Gordon MB, Pulaski-Liebert KJ, Bena J, Biller BM. Revised GH and cortisol cut-points for the glucagon stimulation test in the evaluation of GH and hypothalamic-pituitary-adrenal axes in adults: results from a prospective randomized multicenter study. Pituitary. 2016;19(3):332-41.
  31. Biller BM, Samuels MH, Zagar A, Cook DM, Arafah BM, Bonert V, et al. Sensitivity and specificity of six tests for the diagnosis of adult GH deficiency. J Clin Endocrinol Metab. 2002;87(5):2067-79.
  32. Pekic S, Popovic V. Diagnosis of endocrine disease: Expanding the cause of hypopituitarism. Eur J Endocrinol. 2017;176(6):R269-R82.
  33. Tanriverdi F, Kelestimur F. Classical and non-classical causes of GH deficiency in adults. Best Pract Res Clin Endocrinol Metab. 2017;31(1):3-11.
  34. Verhelst J, Abs R. Cardiovascular risk factors in hypopituitary GH-deficient adults. Eur J Endocrinol. 2009;161 Suppl 1:S41-9.
  35. Utz AL, Yamamoto A, Sluss P, Breu J, Miller KK. Androgens may mediate a relative preservation of IGF-I levels in overweight and obese women despite reduced growth hormone secretion. J Clin Endocrinol Metab. 2008;93(10):4033-40.
  36. Pijl H, Langendonk JG, Burggraaf J, Frolich M, Cohen AF, Veldhuis JD, et al. Altered neuroregulation of GH secretion in viscerally obese premenopausal women. J Clin Endocrinol Metab. 2001;86(11):5509-15.
  37. Makimura H, Stanley T, Mun D, You SM, Grinspoon S. The effects of central adiposity on growth hormone (GH) response to GH-releasing hormone-arginine stimulation testing in men. J Clin Endocrinol Metab. 2008;93(11):4254-60.
  38. Iranmanesh A, Lizarralde G, Veldhuis JD. Age and relative adiposity are specific negative determinants of the frequency and amplitude of growth hormone (GH) secretory bursts and the half-life of endogenous GH in healthy men. J Clin Endocrinol Metab. 1991;73(5):1081-8.
  39. Beck P, Koumans JH, Winterling CA, Stein MF, Daughaday WH, Kipnis DM. Studies of insulin and growth hormone secretion in human obesity J Lab Clin Med. 1964;64:654-67.
  40. Langendonk JG, Meinders AE, Burggraaf J, Frolich M, Roelen CA, Schoemaker RC, et al. Influence of obesity and body fat distribution on growth hormone kinetics in humans. Am J Physiol. 1999;277(5):E824-9.
  41. Magiakou MA, Mastorakos G, Gomez MT, Rose SR, Chrousos GP. Suppressed spontaneous and stimulated growth hormone secretion in patients with Cushing's disease before and after surgical cure. J Clin Endocrinol Metab. 1994;78(1):131-7.
  42. Veldhuis JD, Iranmanesh A, Ho KK, Waters MJ, Johnson ML, Lizarralde G. Dual defects in pulsatile growth hormone secretion and clearance subserve the hyposomatotropism of obesity in man. J Clin Endocrinol Metab. 1991;72(1):51-9.
  43. Yuen KC, Cook DM, Rumbaugh EE, Cook MB, Dunger DB. Individual igf-I responsiveness to a fixed regimen of low-dose growth hormone replacement is increased with less variability in obese compared to non-obese adults with severe growth hormone deficiency. Horm Res. 2006;65(1):6-13.
  44. Nishizawa H, Iguchi G, Murawaki A, Fukuoka H, Hayashi Y, Kaji H, et al. Nonalcoholic fatty liver disease in adult hypopituitary patients with GH deficiency and the impact of GH replacement therapy. Eur J Endocrinol. 2012;167(1):67-74.
  45. Hoffman DM, O'Sullivan AJ, Baxter RC, Ho KK. Diagnosis of growth-hormone deficiency in adults. Lancet. 1994;343(8905):1064-8.
  46. Lee P, Greenfield JR, Ho KK. Factors determining inadequate hypoglycaemia during insulin tolerance testing (ITT) after pituitary surgery. Clin Endocrinol (Oxf). 2009;71(1):82-5.
  47. Pfeifer M, Kanc K, Verhovec R, Kocijancic A. Reproducibility of the insulin tolerance test (ITT) for assessment of growth hormone and cortisol secretion in normal and hypopituitary adult men. Clin Endocrinol (Oxf). 2001;54(1):17-22.
  48. Hoeck HC, Vestergaard P, Jakobsen PE, Laurberg P. Test of growth hormone secretion in adults: poor reproducibility of the insulin tolerance test. Eur J Endocrinol. 1995;133(3):305-12.
  49. Ghigo E, Bartolotta E, Imperiale E, Bellone J, Cardinale G, Aimaretti G, et al. Glucagon stimulates GH secretion after intramuscular but not intravenous administration. Evidence against the assumption that glucagon per se has a GH-releasing activity. J Endocrinol Invest. 1994;17(11):849-54.
  50. Arafat MA, Otto B, Rochlitz H, Tschop M, Bahr V, Mohlig M, et al. Glucagon inhibits ghrelin secretion in humans. Eur J Endocrinol. 2005;153(3):397-402.
  51. Gomez JM, Espadero RM, Escobar-Jimenez F, Hawkins F, Pico A, Herrera-Pombo JL, et al. Growth hormone release after glucagon as a reliable test of growth hormone assessment in adults. Clin Endocrinol (Oxf). 2002;56(3):329-34.
  52. Carmichael JD, Danoff A, Milani D, Roubenoff R, Lesser ML, Livote E, et al. GH peak response to GHRH-arginine: relationship to insulin resistance and other cardiovascular risk factors in a population of adults aged 50-90. Clin Endocrinol (Oxf). 2006;65(2):169-77.
  53. Mathus-Vliegen EM, Obesity Management Task Force of the European Association for the study of obesity. Prevalence, pathophysiology, health consequences and treatment options of obesity in the elderly: a guideline. Obes Facts. 2012;5(3):460-83.
  54. Leong KS, Walker AB, Martin I, Wile D, Wilding J, MacFarlane IA. An audit of 500 subcutaneous glucagon stimulation tests to assess growth hormone and ACTH secretion in patients with hypothalamic-pituitary disease. Clin Endocrinol (Oxf). 2001;54(4):463-8.
  55. Littley MD, Gibson S, White A, Shalet SM. Comparison of the ACTH and cortisol responses to provocative testing with glucagon and insulin hypoglycaemia in normal subjects. Clin Endocrinol (Oxf). 1989;31(5):527-33.
  56. Orme SM, Price A, Weetman AP, Ross RJ. Comparison of the diagnostic utility of the simplified and standard i.m. glucagon stimulation test (IMGST). Clin Endocrinol (Oxf). 1998;49(6):773-8.
  57. Tavares AB, Seixas-da-Silva IA, Silvestre DH, Paixao CM, Jr., Vaisman M, Conceicao FL. Potential risks of glucagon stimulation test in elderly people. Growth Horm IGF Res. 2015;25(1):53-6.
  58. Toogood A, Brabant G, Maiter D, Jonsson B, Feldt-Rasmussen U, Koltowska-Haggstrom M, et al. Similar clinical features among patients with severe adult growth hormone deficiency diagnosed with insulin tolerance test or arginine or glucagon stimulation tests. Endocr Pract. 2012;18(3):325-34.
  59. Bowers CY. GH releasing peptides - structure and kinetics. J Pediatr Endocrinol. 1993;6(1):21-31.
  60. Wang G, Lee HM, Englander E, Greeley GH, Jr. Ghrelin - not just another stomach hormone. Regul Pept. 2002;105(2):75-81.
  61. Piccoli F, Degen L, MacLean C, Peter S, Baselgia L, Larsen F, et al. Pharmacokinetics and pharmacodynamic effects of an oral ghrelin agonist in healthy subjects. J Clin Endocrinol Metab. 2007;92(5):1814-20.
  62. Garcia JM, Swerdloff R, Wang C, Kyle M, Kipnes M, Biller BM, et al. Macimorelin (AEZS-130)-stimulated growth hormone (GH) test: validation of a novel oral stimulation test for the diagnosis of adult GH deficiency. J Clin Endocrinol Metab. 2013;98(6):2422-9.
  63. Garcia JM, Biller BMK, Korbonits M, Popovic V, Luger A, Strasburger CJ, et al. Macimorelin as a diagnostic test for adult growth hormone deficiency. J Clin Endocrinol Metab. 2018;103(8):3083-93.
  64. Garcia JM, Biller BMK, Korbonits M, Popovic V, Luger A, Strasburger CJ, Chanson P, Swerdloff R, Wang C, Fleming RR, Cohen F, Ammer N, Mueller G, Kelepouris N, Strobl F, Ostrow V, Yuen KCJ. Sensitivity and specificity of the macimorelin test for diagnosis of AGHD. Endocr Connect. 2021 Jan;10(1):76-83.
  65. Global News Wire. https://www.globenewswire.com/en/news-release/2022/08/29/2505902/0/en/Aeterna-Zentaris-Set-to-Regain-Full-Rights-to-Macrilen-Macimorelin-in-U-S-and-Canada-from-Novo-Nordisk.html. Accessed August 6, 2023.
  66. Global News Wire.https://www.globenewswire.com/en/news-release/2023/04/05/2641603/0/en/Aeterna-Zentaris-Provides-Update-on-Macrilen-Macimorelin-Reiterates-Advancement-of-Pediatric-DETECT-Trial.html. Accessed August 6, 2023.
  67. Junnila RK, Strasburger CJ, Bidlingmaier M. Pitfalls of insulin-like growth factor-I and growth hormone assays. Endocrinol Metab Clin North Am. 2015;44(1):27-34.
  68. Clemmons DR. Consensus statement on the standardization and evaluation of growth hormone and insulin-like growth factor assays. Clin Chem. 2011;57(4):555-9.
  69. Monthly Prescribing Reference. Macrilen Rx. Available at: https://www.empr.com/drug/macrilen/. Accessed October 28, 2019.

Radiotherapy for Pituitary Tumors

ABSTRACT

 

Pituitary adenomas have been historically managed on a multidisciplinary level with surgery, medical therapy, and radiotherapy to control symptoms secondary to mass-effects and hypersecretion of hormones. While transsphenoidal surgery represents the standard initial approach in the majority of cases, radiotherapy is a valuable and effective treatment option for recurrent adenomas, or lesions not amenable to surgery or medical therapy. Following radiotherapy, tumor growth control (over 90% in most series), plus the normalization of hormones, occurs in a large proportion of treated patients, independent of tumor subtype. Over the last decades, radiotherapy technological advances have allowed the reduction of dose to uninvolved brain while maintaining an effective therapeutic dose to the tumor. This has generated debate on the superiority of some radiotherapy techniques over others. The clinical efficacy of conventionally-fractionated treatment (25 to 30 fractions delivered over 5 to 6 weeks), in the form of 3D-conformal radiotherapy (CRT) or intensity-modulated radiotherapy (IMRT) and the more refined “stereotactic” – highly conformal - fractionated radiotherapy (SFRT), can be compared to that provided by “radio-surgical” (SRS) techniques of irradiation (where the tumor is treated with single high dose of radiation). Due to the lack of randomized control trials addressing this issue, the evidence provided in retrospective studies of different radiotherapy technologies is critically reviewed in this chapter. 

 

INTRODUCTION

 

Pituitary adenomas are mostly benign tumors and comprise about 10% of all intracranial tumors [1, 2]. Radiotherapy has an important and long-established role as part of the multi-disciplinary management of both non-functioning and functioning adenomas. There has been a steady evolution in radiotherapy technologies since radiotherapy was first used to treat pituitary adenomas more than 100 years ago [3]. Despite decades of clinical experience, there remains a paucity of randomized clinical trials to enable a robust evidence-based approach to the optimal use of radiotherapy. This is to some extent compensated for by the large number of non-randomized largely retrospective case series which provide evidence on relevant clinical outcomes and toxicities associated with pituitary radiotherapy. Nevertheless, given the nature of the available data, there continue to be areas of controversy regarding the use of particular radiotherapy modalities. We review the available published data on modern radiotherapy techniques for the treatment of pituitary adenomas to provide a rational basis for the selection of radiotherapy technologies.

 

RATIONALE FOR PITUITARY RADIOTHERAPY

 

Traditional practice had been to use post-operative radiotherapy for all patients with a residual non-functioning pituitary adenoma after surgical resection, as it was considered that otherwise most would subsequently progress [4, 5]. With improvements in surgical techniques, and the development of magnetic resonance imaging (MRI), post-operative radiotherapy is no longer routinely used, even in the presence of residual tumor. The use of post-operative pituitary radiotherapy is now based on a risk assessment. In patients with non-functioning adenomas, radiotherapy is generally withheld until the time of progression, unless there are concerns of significant threat to function (vision) with tumor progression, or the histology raises concerns of earlier recurrence risk (e.g., atypical features, silent corticotroph adenoma). When radiotherapy is used for patients with progressive non-functioning adenomas, tumor control is achieved in over 90% of patients at 10 years, and in 85-92% at 20 years [5-13].

 

In patients with functioning adenomas, radiotherapy is used when surgery fails to achieve hormone normalization and/or when medical treatment is insufficient to control hormone secretion or is not considered appropriate, often due to toxicities. Hormone levels decline slowly following radiotherapy, consequently normalization may take from months to years to achieve. The time required to achieve hormone normalization is primarily related to the pre-treatment hormone levels. Nevertheless, despite this temporal delay, the majority of patients will eventually achieve normalization of excess pituitary hormone secretion following radiotherapy [14].

 

CURRENT TECHNIQUES OF PITUITARY RADIOTHERAPY

 

The principal aim of pituitary radiotherapy techniques has always been to deliver an effective treatment dose to the target tumor volume while at the same time minimizing the radiation dose delivered to surrounding normal tissues, thereby minimizing the risk of normal tissue damage. Improved radiotherapy treatment precision, with the use of the modern radiotherapy techniques described in this chapter, relies on the increased accuracy in tumor volume delineation achieved by using modern MRI imaging technology. Over the last twenty years there have been a number of developments in techniques for pituitary radiotherapy which have largely amounted to refinements of existing technologies. However, the overall success of modern high precision pituitary radiotherapy techniques is largely a function of the quality of a treatment center’s infrastructure and its expertise and accuracy in identifying the target tumor volume, rather than of the particular radiotherapy technique that is used to deliver treatment.

 

3D-Conformal RT

 

Until the last decade, the standard of care for pituitary radiotherapy was three-dimensional (3D) conformal radiotherapy (CRT). CRT uses pre-treatment computed tomography (CT) and MRI imaging for computerized 3D radiotherapy treatment planning. CRT treatment is planned and delivered using a non-invasive method of patient immobilization. The tumor is visualized on unenhanced magnetic resonance imaging co-registered with planning computed tomography (CT). The treatment target is delineated on the MRI scan (in the three orthogonal planes), while radiotherapy dosimetry is calculated using the CT scan data.

 

The treatment target comprises the visible residual tumor and also accounts for any pre-operative extension of disease whilst sparing the optic chiasm where possible after decompression. An isotropic margin of 5-10 mm is added to account for areas of uncertainty in volume delineation, the transsphenoidal surgical route and any set-up variation. The whole pre-operative extent of the tumor is not included within the treatment volume as debulking of large, and particularly cranially extending tumors, often leads to the return of normal anatomical structures to their pre-morbid positions with no residual tumor present. On the other hand, tumors are frequently not removed from the walls of the cavernous sinus, particularly if the sinus is involved, and so the lateral extent of the radiotherapy target does not tend to alter with surgery. The resulting volume outlined on the treatment planning system therefore encompasses both the visible tumor and also any regions of presumed residual tumor. Normal tissue structures adjacent to the pituitary, such as the optic chiasm and optic nerves, the brain stem and the hypothalamus, may also be outlined to aid in treatment planning, and also to enable the calculation and recording of normal tissue

dosimetry, although with conventional fractionated radiotherapy all the structures are treated to below the limits of radiation tolerance in terms of structural damage.

Figure 1. CT-MRI co-registration for planning purposes.

 

Reproducible patient immobilization is vital for the delivery of safe and accurate CRT. The immobilization system used should be well tolerated and must reliably minimize patient movement during both pre-treatment imaging and treatment delivery itself. The most commonly used system for immobilization for CRT is a custom-made closely fitting lightweight thermoplastic mask which is applied and molded directly to the patient’s face in the treatment planning process. The repositioning accuracy of this system is very good at around 3-5mm [15], and can be improved to 2-3mm, by using a tighter fitting but less comfortable mask [16].

 

CT imaging for CRT planning is performed with the patient lying in the radiotherapy treatment position within the immobilization system and co-registered with the MRI (Figure 1) 3D computerized radiotherapy planning is followed by robust quality assurance (QA) procedures to ensure the accuracy of the whole process both before and during treatment. The planning system defines the number, shape, and orientation of radiation beams to achieve uniform dose coverage of the target volume with the lowest possible dose to the surrounding normal tissues. As the dose to the tumor is below the radiation tolerance dose of the surrounding normal tissue structures, no specific measures are generally needed, or taken, during treatment planning to avoid the optic apparatus, hypothalamus, and brain stem. In any case, for many patients requiring pituitary irradiation, some of these entire normal structures lie within, or in close proximity to, the target volume and cannot be avoided without compromising the efficacy of treatment.

 

Localized irradiation is achieved using treatment in multiple beams each shaped to conform to the shape of the tumor using a multileaf collimator (MLC). Traditionally, beam arrangements used for CRT consisted of three fixed beams (an antero-superior beam and two lateral beams) (Figure 2).

 

Figure 2. Example of beam arrangement and dose distribution in a traditional CRT plan (one antero-superior beam and two lateral beams).

 

Intensity-Modulated RT

 

Techniques for varying the radiation dose intensity across a beam, by moving MLC leaves into the beam path, are now standard and are collectively referred to as intensity modulated radiotherapy (IMRT). IMRT is a form of 3D CRT which can spare critical structures, especially within a concave PTV. Although IMRT offers no significant advantage in comparison with CRT for target volume dose coverage [

], its improved conformality can allow for reduced radiation dose delivery to adjacent normal tissues. This can be of particular use in tumor with suprasellar extension, where the dose delivered to the medial temporal lobes can reduced. The technique of arcing IMRT (described as VMAT or RapidArc) offers a fast way of delivering complex IMRT and is increasingly used as an alternative to fixed-field techniques (Figure 3).

 

Figure 3. Example of beam arrangement and dose distribution in a static field IMRT plan (left) and in a VMAT plan (right) for the same patient. Note the better conformality of the high radiation dose region to the target volume in comparison with the CRT plan in Figure 2.

 

Patient immobilization and the imaging required for target volume definition are no different for IMRT treatment than for CRT as described above. Similarly, there are robust QA procedures to ensure the accuracy of IMRT treatment planning and delivery.

 

Stereotactic Radiotherapy Techniques

 

The term “stereotactic” is derived from long-established neurosurgical techniques, and denotes a method of determining the position of a lesion within the brain using an external 3D co-ordinate system based on a method of immobilization, usually an invasive neurosurgical stereotactic head frame [18-20]. Stereotactic radiotherapy originally referred to radiotherapy treatment delivered to an intracranial target lesion that was located by stereotactic means in a patient immobilized in a neurosurgical stereotactic head frame.

 

Stereotactic radiotherapy was first delivered with a multiheaded cobalt unit described as the gamma-knife (GK) which uses multiple cobalt-60 sources arranged in a hemispherical distribution with collimators to achieve a circumscribed spherical dose distribution of 4-18mm diameter [20]. Subsequent development of the GK has allowed larger non-spherical tumors to be treated by combining several radiation spheres using a multiple isocenter technique.

 

Due to the invasive nature of the GK stereotactic head frame (surgically fixed to the skull), GK radiation treatment is delivered as a single large dose during one combined treatment planning and delivery session. This single fraction stereotactic radiation technique was termed ‘radiosurgery’ [18]. The GK radiosurgical procedure aimed to create a non-invasive radiation-based analogue of an open neurosurgical ablation of an intra-cranial target lesion. It should be emphasized, however, that aside from the use of a surgically-fitted stereotactic frame, GK radiosurgery and open neurosurgery are quite distinct procedures, and GK radiosurgery is a radiotherapeutic rather than a surgicalintervention, particularly as the commonly used doses are not “ablative”.

 

Subsequently, linear accelerators (linacs) were adapted to deliver radiosurgery (single fraction radiation) using multiple arcs of rotation, achieving the same dose distribution as that delivered by the GK. With the introduction of non-invasive relocatable stereotactic head frames, which enabled stereotactic radiation to be given in a number of treatment sessions, stereotactic radiotherapy was delivered as fractionated treatment to conventional doses [21, 22]. Initially, specifically adapted linacs were required, but the precision of modern linacs is now such that they do not generally require modification for stereotactic radiotherapy. The improved patient immobilization, more accurate tumor target localization using cross-sectional image for treatment planning, and high precision radiation treatment delivery to the tumor target, enabled a reduction in the margins around the radiotherapy target volume (the gross tumor volume (GTV) to planning target volume (PTV) margin), therefore achieving greater sparing of surrounding normal tissues than can be obtained with standard CRT techniques.

 

The miniaturization of a 6MV linear accelerator has allowed for its mounting on a high precision industrial robotic arm, and this has been combined with real time kV imaging for target tracking during treatment to create a robotic frameless stereotactic radiotherapy machine that is commercially known as the Cyberknife (CK) [23]. The CK uses multiple narrow, low dose rate photon beams, which have to be summated, to create a dose distribution equivalent to that achieved with other techniques. The need to summate contributions from multiple narrow beams results in longer treatment times per fraction than with other techniques and requires that CK treatment be given as a single large fraction (SRS), or as a few large fractions delivered over the course of a week or so (hypofractionated stereotactic radiotherapy).

 

While the term stereotactic radiotherapy continues to be used, “stereotaxy” as initially used for neurosurgery and subsequently for target localization in radiotherapy is no longer necessary and not in routine use, as modern MR and CT imaging with on treatment image guidance allow for equivalent high-precision treatment delivery. The appropriate modern terminology for the best and most accurate techniques of treatment delivery should be high precision conformal radiotherapy. Nevertheless, the term stereotactic used in conjunction with fractionated treatment (see below), while largely outmoded, remains in use with no clear meaning other than presumably denoting accuracy. Stereotactic localization, however, largely remains the standard of practice with single fraction treatment (GK radiosurgery).

 

Radiotherapy Fractionation

 

TERMINOLOGY

 

The term ‘radiosurgery’ is used for radiation treatment that is given as a single large dose (a single fraction), and the term radiotherapy is used for treatment that is given as multiple, usually daily, small doses over a period of weeks (fractionated treatment). The fractionation of radiation treatment is a mechanism for protecting normal tissues, through recovery between fractions, and permits the delivery of higher total doses of radiation than can be given as single fractions [24].

 

Similarly, stereotactic radiotherapy to the pituitary can be given in multiple doses as fractionated stereotactic conformal radiotherapy (SCRT or fSRT), or as a single large dose when it is described as stereotactic radiosurgery (SRS). SCRT/fSRT is generally delivered using a linac. SRS has most frequently been delivered using a GK, but can also be delivered using a linac or a robotic arm mounted linac (CK). Treatment given in fewer large fractions is described as hypofractionated RT.

 

BIOLOGICAL RATIONALE

 

The use of single fraction SRS is based on a belief, prevalent in the literature, that there is greater clinical benefit from single fraction rather than fractionated irradiation for pituitary adenomas. This belief was based on radiobiological modelling which defines equivalent radiation doses and fractionation schemes through biologically derived parameters [24, 25], mainly from the radiobiology of malignant tumors and some normal tissues. Such models are not validated for single fraction treatments [26], and the corresponding biological parameters necessary to calculate equivalent radiation doses do not exist for benign tumors. Publications claiming theoretical benefit of single fraction radiosurgery over fractionated irradiation [25] are based on constants that are not derived from experimental data and may therefore be misleading.

 

The therapeutic effect of radiation on malignant tumors is thought to be due to tumor cell attrition, either as apoptosis, or reproductive cell death, secondary to radiation-induced DNA damage. As a consequence, the time taken for an irradiated tissue to manifest radiotherapy related effects is proportional to the rate of cell proliferation in the tissue. In tissues with rapidly proliferating cells (malignant tumors), radiation effects are expressed either during or immediately after a course of radiotherapy, while in a tissue with a slowly proliferating cell population, such as benign tumors, radiotherapy effects may take many months or years to manifest. It is assumed that the beneficial effects of radiation in pituitary adenomas conform to these same mechanistic principles with the radiation-induced depletion of pituitary adenoma tumor cells, and with the adenoma being considered a slowly proliferating tissue. As benign tumors are rarely grown in culture, the precise mechanism of the observed clinical benefit of irradiation is not elucidated and remains largely theoretical. The surrounding normal brain tissue is also considered to consist largely of slowly proliferating cell populations, although critical cell populations with faster turnover, such as blood vessels, are also present and are affected by radiation.

 

DOSE FRACTIONATION SCHEMES FOR PITUITARY ADENOMAS

 

Conventional CRT and fractionated SCRT are given to total dose of 45 to 50 Gy at 1.8 Gy per fractionation, once a day, five days per week. These treatment doses are below the tolerance of central nervous system neural tissue, and the risk of structural damage due to such treatment is <1% [27, 28]. While, theoretically, single large doses of radiation as used in SRS may result in a higher tumor cell kill than the equivalent total dose given over a small number of fractions, this is also true for the normal tissue cell population and leads to normal tissue toxicity which may not be acceptable if it affects critical regions such as optic chiasm [28].

 

As most pituitary adenomas requiring radiation treatment lie in close proximity to the optic apparatus, and to the cranial nerves in the cavernous sinus, SRS is suitable only for small lesions located away from critical structures, and the optic apparatus should not exceed single doses above 8Gy [28]. Fractionated SRT, using up to 5-fractions over a week course, is another feasible alternative fractionation scheme delivered by LINACs, Cyberknife or frameless radiosurgery.

 

For larger NFPA with chiasmatic involvement, hypofractionation can allow for safe delivery of enhanced biologically effective doses compared to conventional fractionation. The safety of this scheme has been recently reported in a cohort of NFPA, the majority with abutment or compression of optic chiasm, who had satisfactory local control compared to SRS with acceptable toxicity for visual preservation [29].

 

Linac Based SCRT/FSRT    

 

For fractionated stereotactic radiotherapy, patients are immobilized in a non-invasive relocatable frame with a relocation accuracy of 1-2mm [21, 22], or a precisely fitting thermoplastic mask system with an accuracy of 2-3mm [16]. Sub-millimeter repositioning accuracy can now be achieved with thermoplastic mask immobilization by means of image guidance techniques which can determine and apply daily online setup corrections [30]. As for conventional CRT, the GTV is outlined on an MRI scan co-registered with a CT scan. The PTV margin used for SCRT is smaller than for conventional CRT, typically in the region of 3-5mm based on the overall accuracy of the treatment system, the principal determinant of which is the repositioning accuracy of the patient in the immobilization device [31] and the ability to correct it with on treatment imaging (image guidance). For such precision treatment, accurate localization of the tumor volume is of paramount importance in order to avoid treatment failure due to exclusion of a part of the tumor from the treatment volume.

 

SCRT employs a larger number of radiotherapy beams than conventional CRT (usually 4-6). Each beam is conformed to the shape of the PTV using a narrow leaf MLC (5mm width known as mini MLC, or 3mm width known as micro MLC). MLC leaves can be used to modulate the intensity of the radiation beam during its delivery as in intensity-modulated radiotherapy (IMRT). More recently, arc-based or rotational techniques (volumetric modulated arc therapy or VMAT) have been introduced in the clinical practice to overcome some of the limitation of IMRT (complex planning and QA process). The continuous rotation of the radiation source allows the patient to be treated from a full 360° beam angle in a shorter time interval. Fractionated SCRT (fSRT) combines the precision of stereotactic patient positioning and treatment delivery with standard radiotherapy fractionation, which preferentially spares normal tissue. Complete avoidance of surrounding normal tissue structures, such as the optic apparatus, is not generally practiced, as the dose fractionation schemes used are below the radiation tolerance doses of the CNS. Nonetheless arc techniques are used to minimize the dose bilaterally to the temporal lobes with the aim of reducing the impact of treatment on patients’ cognitive function. The fractionated SCRT technique is suitable for pituitary adenomas of all sizes, regardless of their relationship to adjacent critical normal tissue structures.

 

Linac Based SRS

 

Linac based SRS can be delivered using either a relocatable or an invasive neurosurgical stereotactic frame. Use of an invasive neurosurgical frame necessitates that the treatment planning and delivery procedures are carried out and completed within a single day. Computerized treatment planning defines the arrangement of the radiation beams, as in SCRT. SRS can be planned either as multiple arcs of rotation, simulating GK SRS treatment, and producing small spherical dose distributions, or as multiple fixed conformal fields. Multiple arc SRS using a linear accelerator, employing multiple isocenters, is a cumbersome and rarely used technique. The use of multiple fixed fields is generally confined to fractionated treatment, although it can also be used for single fraction SRS. Because of the potentially damaging effect of large single fraction radiation doses on normal tissue structures, SRS is only suitable for small pituitary adenomas that are at least 3-5mm away from the optic chiasm.

 

Several dosimetry studies have shown that linear accelerators could deliver the same SRS doses to pituitary tumors as GK, with comparable conformity indices and OAR doses. Linac SRS has the advantage of being available, efficient with a less beam-on time, so could be considered for radiosurgery of pituitary adenomas [32]..

 

Gamma Knife SRS

 

For GK SRS, patients are immobilized in an invasive neurosurgical stereotactic frame. A relocatable non-invasive stereotactic frame has become available, enabling the delivery of hypofractionated stereotactic radiotherapy treatment in addition to SRS, and experience with this system is increasing [33, 34]. GK SRS delivers a single high dose, in a spherical distribution, of 4-18mm diameter. Larger, non-spherical tumors, which represent the majority of pituitary adenomas, are treated by combining several such spherical dose volumes using a multiple isocenter technique. The appropriate number and distribution of isocenters is defined using a 3D computer planning system which also allows for selective plugging of some of the cobalt source positions to enable shaping of the high dose volume envelope. The use of multiple isocenters results in dose inhomogeneity within the target volume, with small areas of high radiation dose (hot spots) in the regions of overlap of the radiation dose spheres. This may lead to radiation damage if critical normal structures, such as cranial nerves, lie within these hot spots. GK SRS is given to doses of 12 - 35Gy to the tumor margin with doses to the optic chiasm and the other cranial nerves in the cavernous sinus limited to 8-10Gy and 16-18 Gy respectively. royal sinus invasion has been reported as a significant predictor of poor outcomes after surgical resection. Different series have shown good local control using GK for positive residuals within the cavernous sinus after surgical resection [35-39].  

 

Although the total dose delivered with fractionated meanings of irradiation is largely consistent within different publications (45-50.4 Gy), the range of dose prescriptions between secretory and non-functioning adenomas treated with single fraction SRS tends to be different. The rationale behind this practice is based on the observation that a more rapid hormone normalization was reported in single studies using higher doses to treat secreting tumors [40, 41]. In absence of a strong radiobiological model and of prospective randomized studies in support, the relationship between dose and endocrine remission warrants further investigation.

 

Robotic Mounted Linac SRS

 

Cyberknife has been used to treat pituitary adenomas using a variety of dose/fractionation regimens, with a tendency to deliver treatment as hypofractionated radiotherapy in 3 to 5 fractions, rather than as single fraction SRS doses.

 

Proton Therapy

 

Proton beams, heavy charged particles with similar radiobiological effectiveness as photons, have been in use at a small number of centers with the relevant facilities since the late 1960s [42, 43]. Proton therapy was initially used in two US centers (Boston, MA, and Loma Linda, CA) and then subsequently in Europe (d’Orsay, France) and Japan (Tsukuba, Japan); these centers have reported the majority of the initial clinical results. The introduction of proton therapy had been underpinned by planning studies demonstrating, in selected cases, improved dose distribution of protons compared with photons.

 

The principal theoretical advantage of proton therapy over photon therapy is the deposition of energy at a defined depth in tissue (the Bragg peak) with little energy deposition beyond that point [44]. These properties make the use of protons appealing for tumors lying in close proximity to critical dose-limiting normal tissues, which is a bar to safe dose escalation using conventional photon radiotherapy, or when a reduction of low dose (the low dose radiation “bath” responsible for the late sequelae of radiotherapy) to the normal brain tissues is of particular clinical evidence, as in children.

 

Current indications for the use of protons within the UK Specialized Commissioning Team include the treatment of craniopharyngiomas and pituitary adenomas up to the age of 24 years old based on theoretical reduction in the possible late side effects of brain radiation, such as second malignancy, neuro-cognitive deficits and cerebrovascular disease [45].

 

Peptide Receptor Radionuclide Therapy (PRRT)

 

PRRT is a form of internal radiation therapy directed to the tumor tissues expressing peptide receptors using gamma emitting radiopharmaceuticals. It is typically used for neuroendocrine tumors; however, it was investigated as a treatment option for aggressive pituitary tumors refractory to other treatment modalities. Different pituitary tumors express somatostatin receptors and show uptake of radiolabeled somatostatin analogues like 68Ga-DOTATATE. The 2018 guidelines of the European Society of Endocrinology listed PRRT as an alternative treatment option for aggressive pituitary tumors refractory to other lines of treatment including temozolomide [46].The treatment doses and the type of nucleotide used varied in the available studies, with only small patient numbers being reported [47, 48].

 

CLINICAL OUTCOMES FOLLOWING PITUITARY RADIOTHERAPY

 

The clinical efficacy of radiotherapy for pituitary adenomas should be assessed by overall survival, actuarial tumor control (progression-free survival, PFS), and quality of life. Few publications focused on quality of life assessment after radiotherapy in pituitary tumors [49-51], while commonly reported endpoints for retrospective studies of radiation treatment for non-functioning pituitary adenomas are local tumor control, and long term morbidity.

 

In patients with functioning pituitary adenomas, the principal endpoint, in addition to PFS and morbidity, is the rate of normalization of elevated pituitary hormone levels. The rate of pituitary hormone decline after irradiation varies with the type of functioning tumor, and the time to reach normal hormone levels is dependent on the initial pre-treatment hormone levels [52]. The appropriate comparative measure for each pituitary hormone is the time to reach 50% of the pre-treatment hormone level, and this should be corrected for the confounding effect of medical treatment.

 

Surrogate endpoints such as ‘tumor control rate’ and the ‘proportion of patients achieving normal hormone levels’ do not, of themselves, provide adequate information on the efficacy of different pituitary irradiation techniques and are potentially misleading [53]. Tumor control rate must be quoted with an indication of the time or duration of follow-up required to achieve the stated level of control. Similarly, the proportion of patients achieving normal hormone levels following treatment is meaningful only when described in terms of the relationship to pre-treatment hormone levels. Due to the use of such surrogate endpoints in published retrospective series, inappropriate and incorrect claims have been made in the literature for superiority of one technique of irradiation over another.

 

Given that the published data on the efficacy of the various available techniques for pituitary irradiation consist entirely of retrospective case-series, the available data inevitably suffer from selection bias. While SCRT is suitable for the treatment of all pituitary tumors, irrespective of size, shape or proximity to critical normal tissue structures, SRS is only suitable for treatment of small tumors away from the optic chiasm. As a result, studies reporting the efficacy of SRS mostly deal with smaller tumors, which are typically associated with lower hormone levels if the adenomas are functioning. Therefore, the reported results of studies of SRS do not necessarily apply to the generality of pituitary adenomas and may give a false impression of greater efficacy if only more favorable cases are treated.

 

THE EFFICACY AND TOXICITIES OF TREATMENT

 

Conventional RT and CRT

 

The efficacy of modern stereotactic pituitary radiotherapy and pituitary radiosurgical techniques must be assessed in the light of the results achieved with standard treatment, which is conventional conformal radiotherapy. Large and mature case series provide data on the long-term effectiveness of CRT in controlling pituitary tumor growth and hormone secretion.

 

TUMOR CONTROL

 

The long-term results following pituitary CRT from case series published in the literature are shown in Table 1 [5-14, 17, 54-66]. The actuarial PFS is in the region of 80%-90% at 10 years and 75%-90% at 20 years [14, 55]. The single largest series of patients with pituitary adenomas treated with conventional fractionated radiotherapy is that from The Royal Marsden Hospital which reported a 10-year PFS of 92% and a 20-year PFS of 88% [8].

Post operative radiotherapy has been reported to provide excellent local control of non-functioning tumors when offered for progressive residual disease with almost no radiological evidence of tumor progression up to 15 years of follow-up [67].

 

ENDOCRINE CONTROL  

 

Fractionated irradiation leads to normalization of excess pituitary hormone secretion in the majority of patients, albeit with some time delay following treatment. For acromegaly, RT achieves normalization of GH/IGF-I levels in 30-50% of patients at 5-10 years, and in 75% of patients at 15 years, after treatment (Table 2) [14, 55]. As the time to normalization of GH levels is related to the pre-treatment GH level, the time to achieve a 50% reduction in GH levels, which takes into account the starting GH level, is in the region of 2 years, with IGF-1 reaching half of pre-treatment levels somewhat after the GH [58, 60].

 

A 10-year follow-up for more than 600 acromegaly patients was published by the Swedish Pituitary Register 2022. It has reported 78% of IGF-1 normalization rate with an annual rate of increased hormonal control of 1.23%. One third of the patients required bi-modality therapy to achieve hormonal control and 5% required triplet therapy i.e. surgical resection, medical treatment and radiotherapy with a trend towards reduced use of conventional radiotherapy doses [68]. 

 

Table 1. Summary of Results of Published Series on Conventional RT for Pituitary Adenomas

Authors

Type of adenoma

Number of patients

Follow-up

(median years)

Actuarial progression free survival (PFS) (%)

Late toxicity (%)

Visual Hypopituitarism

Grigby at al.,1989 [6]

NFA, SA

121

11.7

89.9 at 10 years

1.7

NA

McCollough et al., 1991 [7]

NFA, SA

105

7.8

95 at 10 years

NA

NA

Brada et al., 1993 [8]

NFA, SA

411

10.8

94 at 10 years

88 at 20 years

1.5

30 at 10 years

Tsang et al., 1994 [9]

NFA, SA

160

8.7

87 at 10 years

0

23**

Zierhut et al., 1995 [10]

NFA, SA

138

6.5

95 at 5 years

1.5

27**

Estrada et al., 1997 [56]

SA (ACTH)

30

3.5

73 at 2 years*

0

48**

Rush et al., 1997 [11]

NFA, SA

70

8

NA

NA

42**

Breen et al., 1998 [12]

NFA

120

9

87.5 at 10 years

1

NA

Gittoes et al., 1998 [5]

NFA

126

7.5

93 at 10 and 15 years

NA

NA

Barrande et al., 2000 [57]

SA (GH)

128

11

53 at 10 years*

0

50 at 10 years

Biermasz et al., 2000 [58]

SA (GH)

36

10

60 at 10 years*

0

54 at 10 years

Sasaki et al., 2000 [13]

NFA, SA

91

8.2

93 at 10 years

1

NA

Epaminonda et al., 2001 [59]

SA (GH)

67

10

65 at 15 years*

0

NA

Minniti et al., 2005 [60]

SA (GH)

45

12

52 at 10 years*

0

45 at 10 years

Langsenlehner et al., 2007 [61]

NFA, SA

87

15

93 at 15 years

 

0

88 at 10 years

Minniti et al., 2007 [62]

SA(ACTH)

40

9

78 and 84 at 5 and 10 years*

0

62 at 10 years

Rim et al., 2011 [63]

NFA, SA

60

5.6

96 at 10 years (NFA),

66 at 10 years (SA)

0

76 at 10 years

Kim et al., 2016 [65]

NFA, SA

73

8

98 at 10 years

0

NA

Patt et al., 2016 [66]

SA (GH)

36

4.9 (mean)

89 at 5 years

0

33

NFA, non-functioning adenoma; SA, secreting adenoma; NA, not assessed, ACTH-Cushing, GH- acromegaly, *hormone concentration normalization, **no time specified

 

After RT for Cushing’s disease, urinary free cortisol (UFC) is reduced to 50% of the pre-treatment levels after an interval of 6-12 months, and plasma cortisol after around 12 months [62]. The median time to cortisol level normalization is around 24 months after treatment [62]. The overall tumor and hormone control rates in the reported studies, after a median follow-up of 8 years, are 97% and 74% respectively [64]. Pituitary radiotherapy is rarely used to treat patients with prolactinoma. Occasional patients who fail surgery and medical therapy have been treated with RT, and the reported 10-year tumor and hormone control rates are 90% and 50% respectively [69-71].

 

TOXICITY

 

The toxicity of RT with total treatment doses of 45-50Gy with daily fraction sizes of < 2Gy is low. The principal toxicities reported in studies of CRT are described in Table 1.

 

Hypopituitarism  

 

Hypopituitarism is the most common long-term complication following RT, reported to occur in 30-60 % of patients by 10 years after treatment [8, 9, 14]. Pituitary hormone loss is observed to occur in a characteristic sequence, with GH secretion being affected most frequently, followed by the gonadotrophins, ACTH, and then TSH. Long term follow-up after pituitary irradiation, with intermittent testing for deficiency of all pituitary axes, is therefore an essential part of the post-treatment management of these patients.

 

Visual Pathways Deficit and Other Structural CNS Damage

 

The reported incidence of optic neuropathy resulting in visual deficit following CRT is 1-3% [8, 9]. The occurrence of necrosis of normal brain tissue is almost unknown following pituitary RT, although this complication has been reported to occur in 0.2% of patients [72].

 

Cerebrovascular Disease

 

Pituitary disease is, in itself, associated with increased mortality, principally due to vascular disease [73]. An increased incidence of stroke (relative to the general population) in patients treated with RT for both non-functioning and functioning pituitary adenomas has been reported in a number of retrospective cohort studies [74-77]. Whilst it is has long been known that radiotherapy can lead to vascular injury [78], it is not at present clear how much of the excess stroke risk following RT is attributable to radiotherapy, and how much may be due to other potential causes including the metabolic and cardiovascular consequences of hypopituitarism, the effects of associated endocrine syndromes, and the consequences of surgery.

 

In a retrospective cohort study of 342 patients treated with pituitary surgery and post-operative RT, 31 patients died from stroke after a median follow-up interval of 21 years (range, 2-33) [77] and in all cases the probable location of the stroke lesion was within the irradiated volume. Comparison of stroke patients with matched control patients without stroke drawn from the same cohort showed no significant differences in radiotherapy-dependent variables with the exception of the pre-treatment duration of symptoms of hypopituitarism. This suggests that untreated hormone deficiency may be a significant factor in the pathogenesis of stroke in patients treated for pituitary adenoma, rather than or in addition to treatment with radiotherapy. It is likely that the cause of stroke in patients treated with RT for pituitary adenoma is multi-factorial, and the relative contributions of the various possible contributory factors remains to be determined.

 

Second Brain Tumor

 

Intracranial radiotherapy is associated with the development of second, radiation-induced, brain tumors. The cumulative incidence of gliomas and meningiomas following radiotherapy for pituitary adenomas in retrospective case series is reported to be in the region of 2% at 20 years [77, 79-81]. A large retrospective study of patients who received radiotherapy for pituitary and sellar lesions has shown a relative risk of 3.34 (95% confidence interval 1.06-10.6) for development of malignant brain tumors and 4.06 (95% confidence interval 1.51-10.9) for development of meningiomas in comparison with patients who did not receive radiotherapy. Rates were higher in those treated with radiotherapy at a younger age, and there was no difference in incidence rates between conventional or stereotactic radiotherapy (70).

 

In another large retrospective cohort of more than 3600 patients from six adult endocrinology registries, incidence of secondary brain tumors was compared between irradiated and non-irradiated patients with pituitary adenomas and craniopharyngiomas. The relative risk of secondary brain tumors for irradiated patients was 2·18 (95% CI 1·31-3·62, p<0·0001). Cumulative probability of second brain tumor was 4% for the irradiated and 2·1% for the controls at 20 years. Radiotherapy exposure and older age at pituitary tumor detection were associated with increased risk of second brain tumor [82].

 

Cognitive Deficit

 

Radiotherapy treatment to significant volumes of normal brain in children is associated with subsequent neuro-cognitive impairment [27]. However, the evidence for the effect of radiotherapy treatment to small volumes of brain on neuro-cognitive function in adults is weak [27]. The effect of pituitary radiotherapy on neuro-cognitive function is particularly difficult to discern as this cannot be differentiated from the effect of other treatment interventions, and from the effects of the tumor itself [83-85].

 

A retrospective study of 84 patients following transsphenoidal surgery, of whom 39 received post-operative radiotherapy, compared neuro-cognitive function with a large reference sample, considered to be representative of normal population without pituitary disease. While the pituitary cohort had lower scores on the tests of both memory and executive function in comparison with the reference sample, patients who had received radiotherapy showed no significant difference compared to patients treated with surgery alone [86]. A dosimetric study did not find a correlation between radiotherapy dose to the hippocampus and pre-frontal cortex (brain regions known to be important in memory and executive function) and conformal technique of irradiation with cognitive performance [87].

 

Stereotactic Conformal Radiotherapy (SCRT/FSRT)

 

SCRT achieves tumor control and normalization of pituitary hormone hypersecretion at rates similar to the best reports following conventional RT. Longer duration follow-up is required to demonstrate the presumed lower incidence of long-term morbidity following SCRT compared to conventional RT. The results from reported studies of SCRT are summarized below.

 

TUMOR CONTROL

 

SCRT data for 1166 patients with either non-functioning or functioning pituitary adenomas have been reported in 21 studies to date (Table 2) [14, 17, 55, 64, 88-105]. Analysis of published data up to 2020 shows that, at a corrected median follow-up of 56 months (range 9-152 months), tumor control was achieved in 96% of patients. The 5-year actuarial PFS of 92 patients (67 non-functioning, 25 functioning) treated at The Royal Marsden Hospital was 97% [93]. These results are similar to the results seen in patient cohorts treated with conventional RT (Table 1).

 

ENDOCRINE CONTROL  

 

Detailed data on the rate of pituitary hormone decline are not available, although this is expected to be similar to that seen following conventional RT as the same dose-fractionation is used. In The Royal Marsden case series, 6 of 18 acromegalic patients (35%) had normalization of GH/IGF-I levels after a median follow-up of 39 months [93]. Similarly, in another single center study of 20 patients treated with SCRT, normalization of GH levels was reported in 70%, and local tumor control in 100% after a median follow-up of 26 months [90]. The data available on SCRT for patients with Cushing’s disease are limited. In a small series of 12 patients, control of elevated cortisol was reported in 9 out of 12 patients (75%) after a median follow-up of 29 months [92].

 

TOXICITIES

 

Following SCRT, hypopituitarism has been reported in 22% of patients after an overall corrected median follow-up of 57 months (Table 2). The length of follow-up after SCRT is shorter than reported for the mature cohorts treated with RT. It is likely that the rate of hypopituitarism following SCRT will continue to increase as the duration of follow-up increases particularly as the technique of SCRT generally does not avoid either the hypothalamus or the remaining pituitary gland. Other late complications have been rarely reported after SCRT. While the incidence of treatment-related morbidity with SCRT appears to be low, longer duration follow-up is necessary to detect normal tissue toxicity that may only become manifest at a low frequency many years after treatment.

 

Table 2. Summary of Results on Published Studies on SCRT for Pituitary Adenomas

Authors

Number of patients

Follow-up median (months)

Tumor growth control rate (%)

Late toxicity (%)

Visual  Hypopituitarism

Coke et al., 1997 [88]

19*

9

100

0

0

Mitsumori et al., 1998 [89]

30*

33

86 at 3 years

0

20

Milker-Zabel et al., 2001 [90]

68*

38

93 at 5 years

7

5

Paek et al., 2005 [91]

68

30

98 at 5 years

3

6

Colin et al., 2005 [92]

110*

48

99 at 5 years

2

29 at 4 years

Minniti et al., 2006 [93]

92*

32

98 at 5 years

1

22

Selch et al., 2006 [94]

39*

60

100

0

15

Kong et al., 2007 [95]

64*

37

97 at 4 years

0

11

Snead et al., 2008 [96]

100*

6.7 years

98 and 73 at 10 years for NFA and SA

1

35

Roug et al., 2010 [97]

34*

34

91 (50% hormonal normalization)

-

-

Schalin-Jantti et al., 2010 [98]

30

5.3 years

100

0

23

Weber et al., 2011 [99]

27*

72.4

96

4

8

Wilson et al., 2012 [100]

67

5.12 years

88

2

6

Kim et al., 2013 [101]

76*

6.8 years

97.1 at 7 years

0

48 (one or more hormone)

Kopp et al., 2013 [102]

37

57

91.9

5

43

Liao et al., 2014 [106]

34~

36.8 (mean)

100

0

NA

Minniti et al., 2015 [103]

68

75

97 and 91 at 5 and 10 years

0

26

Puataweepong et al., 2015 [107]

94*

72

95

3

9.6

Diallo et al., 2015 [104]

34*

152 (mean)

97

0

39

Barber et al., 2016 [105]

75*

47.5 (mean)

100

1.5

28

Lian et al., 2020 [108]

113*

36

99

0

28.3

* Case series includes secreting adenomas

 

Radiosurgery (SRS)

 

TUMOR CONTROL  

 

The published results of GK SRS for patients with non-functioning and functioning pituitary adenomas have been summarized in systematic reviews [14, 17, 55, 64] and an update with more recently published studies is given in Table 3 [14, 17, 35, 55, 64, 100, 109-130]. The majority of published reports provide information on tumor ‘control rate’, without specifying a time-frame, and therefore provide little useful information on the efficacy of GK SRS. The summary figure for the actuarial 5-year control rate (PFS) following GK SRS for non-functioning adenomas is 95% at 5 years (few 10-year results are available). This is a lower rate of tumor control than expected following RT & SCRT, particularly when it is considered that only small tumors suitable for GK SRS are treated, compared to that adenoma of all sizes treated with RT, CRT & SCRT.

 

Table 3. Summary of Results of Published Series on SRS for Non-Functioning Pituitary Adenomas

 

Authors

Number of patients

Follow-up median (months)

Tumor control growth rate (%)

Late toxicity (%)

Visual Hypopituitarism

 

Martinez et al., 1998 [109]

14

26-45

100

0

0

 

Pan et al., 1998 [110]

17

29

95

0

0

 

Ikeda et al., 1998 [35]

13

45

100

0

0

 

Mokry et al., 1999 [111]

31

20

98

NA

NA

 

Sheehan et al., 2002 [112]

42

31*

97

2.3

0

 

Wowra et al., 2002 [113]

45

55

93 at 3 years

0

14

 

Petrovich et al., 2003 [114]

56

36

94 at 3 years

4

NA

 

Pollock et al., 2003 [115]

33

43

97 at 5 years

0

28 and 41 at 2 and 5 years

 

Losa et al., 2004 [116]

56

41*

88 at 5 years

0

24

 

Iwai et al., 2005 [117]

34

60

93 at 5 years

0

6

 

Mingione et al., 2006 [118]

100

45*

92

0

25

 

Liscak et al., 2007 [119]

140

60

100

0

2

 

Pollock et al., 2008 [120]

62

63

95 at 3 and 7 years

0

32 at 5 years

 

Kobayashi et al., 2009 [121]

60

>3 years

97

4.3

8.2 worsening

 

Gopalan et al., 2011 [122]

48

80.5

83

9.4

39

 

Park et al., 2011 [124]

125

62

94 at 5 years and

76 at 10 years

1

24 at 2 years

 

Wilson et al., 2012 [100]

51

4.17 years

100

0

0

 

Runge et al., 2012 [123]

61

83

98

0

9.8

 

Starke et al., 2012 [125]

140

4.2 years

97 at 5 and 87 at 10 years

12.8

30.3

 

El-Shehaby et al., 2012 [126]

38

44*

97

0

0

 

Sheehan et al., 2013+ [127]

512

36

95 at 5 years

6.6

21

 

Lee et al., 2014 [128]

41

48

94 at 5 and 85 at 10 years

2.4

24.4

 

Xu et al., 2014 [129]

34

56

73 at 3 years

24

29

 

Hasegawa et al., 2015 [130]

16

98

100

0

6

 

Graffeo et al., 2018[131]

57

48

99

NA

31 at 5years

 

Oh et al., 2018 [132]

76

53.5

96

NA

24.5

 

Cordeiro et al., 2018 [133]

410

51

94.4

NA

34.7

 

Narayan et al., 2018 [134]

87

48.2

90

8.1

20.7

 

Slavinsky P et al., 2022 [135]

 

28

63

94.2

NA

26%

Maldar AN, et al.,2022[136]

63

47

87.3

NA

26% at 5 years

29.7% at 10 years

*Mean follow-up; NA: not available, + multicenter study, 34 patients had prior CFSR

 

ENDOCRINE CONTROL WITH GK SRS

 

The reported endocrine outcomes following GK SRS for acromegaly are shown in Table 4 [14, 36, 40, 55, 64, 109-111, 114, 121, 137-163]. A summary analysis of the published literature up to 2020 shows that -41% of patients achieved normalization of serum GH, after a median follow-up of 46 months. The time to reach 50% of baseline serum GH, reported in only three studies, is in the region of 1.5-2 years with a slower reduction in IGF-I levels [147, 150, 164], which is similar to the rate reported following conventional RT/CRT.

 

Table 4. Summary of Results of Published Series on SRS for GH-Secreting Pituitary Adenomas

 

Authors

Number of patients

Follow-up median (months)

Hormone normalization* (%)

Late toxicity (%)

Visual Hypopituitarism

 

Thoren et al., 1991 [137]

21

64

10

0

15

 

Martinez et al., 1998 [109]

7

26-45

NA

0

0

 

Pan et al., 1998 [110]

15

29

NA

0

0

 

Morange-Ramos et al., 1998 [138]

15

20

20

6

16

 

Lim et al., 1998 [139]

20

26

30

5

5

 

Kim et al., 1999 [165]

11

27

35

NA

NA

 

Landolt et al., 1998 [141]

16

17

50

0

16

 

Mokry et al., 1999 [111]

16

46

31

0

NA

 

Hayashi et al., 1999 [142]

22

>6

41

0

0

 

Inoue et al., 1999 [143]

12

>24

58

0

0

 

Zhang et al., 2000 [144]

68

>12

40

NA

NA

 

Izawa et al., 2000 [145]

29

>6

41

0

0

 

Pollock et al., 2002 [146]

26

36

47

4

16

 

Attanasio et al., 2003 [147]

30

46

23

0

6

 

Choi et al., 2003 [148]

12

43

30

0

0

 

Jane et al., 2003 [149]

64

>18

36

0

28

 

Petrovich et al., 2003 [114]

6

36

100

0

NA

 

Castinetti et al., 2005 [150]

82

49.5*

17

0

18

 

Gutt et al., 2005 [151]

44

22

48

NA

NA

 

Kobayashi et al., 2005 [152]

67

63

17

0

NA

 

Jezkova et al., 2006 [153]

96

54

50

0

26

 

Pollock et al., 2007 [154]

46

63

11 and 60 at 2 and 5 years

0

33 at 5 years

 

Jagannathan et al., 2009 [155]

95

57 *

53

5#

34 (new)

 

Kobayashi, 2009 [121]

49

63

17 (normal or nearly normal)

11

15

 

Wan et al., 2009 [156]

103

60 (minimum)

37

0

1.7**

 

Castinetti et al., 2009 [157]

27

60 (minimum)

42 at 50 months

1.3**

23**

 

Iwai et al., 2010 [158]

26

84

38

0

8

 

Hayashi et al., 2010 [36]

25

36*

40

0

0

 

Erdur et al., 2011 [159]

22

60

55

0

29

 

Sheehan et al., 2011 [40]

130

30

53 at 30 months

0

34

 

Franzin et al., 2012 [160]

103

71

56.9 at 5 years

0

7.8 (new)

 

Liu et al., 2012 [161]

40

72

57.5

0

40 (new)

 

Zeiler et al., 2013 [162]

21

33

30

3.9

13.2

 

Lee et al., 2014 [163]

136

61.5

64.5 and 82.6 at 4 and 8 years

3

33.1

 

Cordeiro et al., 2018 [133]

351

51

NA

NA

38.7

 

Gupta et al.,2018 [166]

25

69.5

28

NA

19.6

Ding et al., 2019 [167]

371

79

59 at 10 years

4

26

 

*mean follow-up; NA not assessed, #3 had previous RT, **whole series

 

A summary analysis of the published literature up to 2020, for patients with Cushing’s disease, shows that 52% achieved biochemical remission (as defined by plasma cortisol and 24-hour UFC level) at a corrected median follow-up of 50 months after SRS (Table 5) [14, 36, 40, 55, 64, 109-111, 114, 121, 138-140, 142, 143, 145, 146, 148, 149, 155-157, 162, 165, 168-178]. The reported time to hormonal normalization ranged from 3 months to 3 years, with no clear difference in the rate of decline of hormone level compared to RT/CRT. The largest single series of GK SRS for Cushing’s disease reported a remission rate of 54%, with 20% of patients who achieved remission subsequently relapsing, suggesting a higher failure rate following GK SRS than following RT/CRT [179].

 

Table 5. Summary of Results of Published Series on SRS for ACTH-Secreting Pituitary Adenomas

 

Authors

Number of patients

Follow-up median (months)

Tumor growth control rate (%)

Hormone normalization*%

Late toxicity

(%)

Visual    Hypopituitarism

 

Degerblad et al., 1986 [168]

29

3-9 years

76

48

NA

55

 

Ganz et al., 1993 [169]

4

18

NA

NA

0

NA

 

Seo et al., 1995 [170]

2

24

100

NA

0

NA

 

Martinez et al., 1998 [109]

3

26-45

100

100

0

0

 

Pan et al., 1998 [110]

4

29

95

NA

0

0

 

Morange-Ramos et al., 1998 [138]

6

20

100

66

0

16

 

Lim et al., 1998 [139]

4

26

NA

25

2

2

 

Mokry et al., 1999 [111]

5

26

93

20

0

2

 

Kim et al., 1999 [165]

8

26

100

60

NA

NA

 

Hayashi et al., 1999 [142]

10

>6

100

10

0

5

 

Inoue et al., 1999 [143]

3

>24

100

100

0

0

 

Izawa et al., 2000 [145]

12

>6

100

17

NA

0

 

Sheehan et al., 2000 [171]

43

44

100

63

2

16

 

Hoybye et al., 2001 [172]

18

17 years

100

83

0

66

 

Kobayashi et al., 2002 [173]

20

60

100

35

NA

NA

 

Pollock et al., 2002 [146]

11

36

85

35

35

8

 

Choi et al., 2003 [148]

9

43

100

55

0

0

 

Jane et al., 2003 [149]

45

>18

100

63

1

31

 

Petrovich et al., 2003 [114]

4

36

NA

50

0

NA

 

Devin et al.,  2004 [174]

35

35

91

49

0

40

 

Castinetti et al., 2007 [175]

40

54

100

42

0

NA

 

Jagannathan et al., 2009 [155]

90

45

96

54

6

22

 

Kobayashi, 2009 [121]

25

64(mean)

100

35

NA

NA

 

Wan et al., 2009 [156]

68

60(minimum)

90

28

0

1.7

 

Castinetti et al., 2009 [157]

18

60(minimum)

NA

50 at 28 months

1.3**

23**

 

Hayashi et al., 2010 [36]

13

36(mean)

97

38

0

0

 

Sicignano et al., 2012 [178]

15

60

97.7

64

NA

12.3

 

Zeiler et al., 2013 [162]

8

35

100

50

3.9

32

 

Sheehan et al., 2013 [177]

96

48

98

70

4

36

 

Marek et al., 2015 [176]

26

78

90.9 at 5 and 10 years

80.7

0

23

 

Cordeiro et al., 2018 [133]

262

51

95.8

NA

NA

26.6

 

Knappe et al., 2020 [180]

119

107

NA

78

NA

NA

 

Gupta et al., 2018  [166]

21

69.5

100

81%

NA

19.6%

*time not specified; NA not assessed

 

In patients with prolactinomas treated with GK SRS the reported time to hormonal response ranged from 5 months to 40 months (Table 6) [14, 40, 55, 64, 109-111, 114, 121, 138-140, 142, 143, 145, 146, 148, 149, 156, 157, 161, 169, 181-186]. At a corrected median follow-up of 43 months (median range 6-60 months), 33% of patients had normalization of serum prolactin concentrations following GK SRS [14]. One study of 35 patients reported a hormonal normalization of 80% after a median of 96 months and a tumor control rate of 97% [184]. There is insufficient information to assess the rate of decline of prolactin following GK SRS in comparison to that following CRT.

 

Table 6. Summary of Results of Published Series on SRS for Prolactin Secreting Pituitary Adenomas

 Authors

Number of patients

Follow-up median (months)

Hormone normalization*%

Late toxicity (%)

Visual        Hypopituitarism

Ganz et al., 1993 [169]

3

18

0

0

NA

Martinez et al., 1998 [109]

5

26-45

0

0

0

Pan et al., 1998 [110]

27

29

30

0

0

Morange-Ramos et al., 1998 [138]

4

20

0

0

16

Lim et al., 1998 [139]

19

26

50

NA

NA

Mokry et al., 1999 [111]

21

31

57

0

19

Kim et al., 1999 [165]

18

27

16

NA

NA

Hayashi et al., 1999 [142]

13

>6

15

NA

5

Inoue et al., 1999 [143]

2

>24

50

0

0

Landolt et al., 2000 [181]

20

29

25

0

NA

Pan et al., 2000 [182]

128

33

41

0

NA

Izawa et al., 2000 [145]

15

>6

16

0

NA

Pollock et al., 2002 [146]

7

26

29

14

16

Choi et al., 2003 [148]

21

43

23

0

0

Jane et al., 2003 [149]

19

>18

11

0

21

Petrovich et al., 2003 [114]

12

36

83

0

NA

Pouratian et al., 2006 [183]

23

55

26

7

28

Jezkova et al., 2009 [184]

35

96

80

NA

NA

Kobayashi, 2009 [121]

27

37(mean)

17

0

0

Wan et al., 2009 [156]

176

60 (minimum)

23

0

1.7

Castinetti et al., 2009 [157]

15

60 (minimum)

46 at 24 months

1.3**

23**

Liu et al., 2013 [185]

22

36

27

-

4.5

Cohen-Inbar et al., 2015 [186]

38

42.3

50

NA

30.3

Ježková et al., 2019 [187]

28

140

82.1

3.6

8.3

 

Early studies of linac based SRS reported results on small numbers of patients, but the available results are broadly equivalent to those reported for GK SRS [17]. In the largest linac based SRS study to date, which included 175 patients with both non-functioning and functioning pituitary adenomas treated using a single dose of 20 Gy, the local tumor control rate was 97% after a minimum of 12 months follow-up [188]. Actuarial 5-year PFS was not reported. Hormonal normalization rates were 47% for GH-secreting adenomas, 65% with Cushing’s disease, and 39% with prolactinomas. The mean time for hormone normalization was 36±24 months. Within the limited follow-up period, 12% developed additional pituitary dysfunction, 3% radiation-induced CNS tissue damage, and 1% radiation-induced optic neuropathy. These results from linac SRS are difficult to evaluate but are broadly similar to those achieved with GK SRS and appear inferior to those obtained with fractionated treatment.

 

TOXICITY

 

In common with other modalities of pituitary irradiation, the most commonly reported complication following GK SRS is hypopituitarism, with a crude incidence ranging from 0% to 66% [14, 55]; the actuarial incidence has not been defined. The expected frequency of visual complications would be low if GK SRS is only offered to patients with a pituitary adenoma at a safe distance from the optic chiasm and nerves (~ 5mm). However, one study in patients with Cushing’s disease reported a 10% incidence of new cranial nerve deficit, with a 6% incidence of optic neuropathy [155]. Similarly, a study reporting results of SRS for prolactinoma noted a 7% incidence of cranial nerve deficit [183]. Although the absolute numbers of patients treated in these studies of GK SRS were small, there is a suggestion that for some patients, possibly with larger tumors, the incidence of optic pathway toxicity with GK SRS is well above what is seen in patients following CRT. Long-term risks of cerebrovascular events and the incidence of second tumors following GK SRS are not yet defined. GK toxicity is expected to be higher when offered after surgical excision rather than as a primary treatment option. In a recently published systematic review and meta-analysis on 1381 patients with pituitary adenomas treated with GK SRS, rates of radiation-induced hypopituitarism were (11.4%) in primary GK compared to (18-32%) in post-operative GK SRS. This highlights the importance of long-term endocrinology follow-up [189].

 

Robotic SRS

 

A small number of retrospective case series on outcomes following CK SRS for pituitary adenomas have been published to date (Table 7) [190-197]. While the published results are comparable to the outcomes achieved with GK SRS, the same criticisms levelled at the GK SRS studies also apply to these early CK SRS series. The duration of follow-up in all the existing CK SRS series is too short to allow meaningful conclusions to be drawn with regard to both efficacy and toxicity outcomes.

 

Table 7. Summary of Results of Published Series on Cyberknife SRS for Functioning & Non-Functioning Pituitary Adenomas

 

Author

Tumor type

Number of patients

Follow-up mean (months)

Tumor Control or Hormone normalization*

(%)

Late toxicity (%)

Visual Hypopituitarism

 

Kajiwara et al., 2005 [190]

14 NFA, 3 PRL, 2 GH, 2 ACTH

21

35.3

95.2TC, 50 HN

4.7

9.5

 

Adler et al., 2006 [191]

12 NFA, 4 GH, 2 ACTH, 1 PRL

19

46

18/19 TC

5.2

NA

 

Roberts et al., 2007 [192]

GH

9

25.4

44.4 HN

0

33

 

Killory et al., 2009 [193]

14 NFA, 4GH, 1 PRL, 1 TSH

20

26.6

100 TC

0

NA

 

Cho et al., 2009 [194]

17 NFA, 3 PRL, 6 GH

26

30

92.3 TC, 44 HN

7.6

0

 

Iwata et al., 2011 [195]

NFA

100

33 median

98 TC

1

4

 

Puataweepong et al., 2015[196]

27 NFA, 7 GH, 5 PRL, 1 ACTH

40

38.5 median

97.5 TC, 54 HN

0

0

 

Iwata et al., 2016 [197]

GH

52

60 median

100 TC, 20.4 HN

0

2.2

 

Plitt et al., 2019 [198]

NFA

53

32.5

98.1 TC

0

1.9

 

Romero-Gameros et al., 2023 [199]

GH

57

48

45.6% HN

0

24.5

                 

TC: Tumor Control; HN: hormone normalization

 

Proton Beam Therapy

 

An early study, published in 1989, of proton beam therapy for pituitary adenomas attempted to compare the effectiveness of this treatment modality to RT/CRT [200]. Follow-up after CRT in 17 patients and after proton therapy in 13 patients found a similar reduction of GH levels in both groups and the small number of patients does not allow for any statistically meaningful comparison. Nevertheless, treatment related side effects, including new hypopituitarism and oculomotor palsies, were more frequent in proton therapy group. Since the efficacy of both pituitary irradiation methods were similar, but proton therapy was associated with a higher incidence of serious side effects, the authors concluded that RT/CRT is the preferred treatment modality [200].

 

In a study from 2006, of 47 patients treated with fractionated proton therapy for both non-functioning and functioning pituitary adenomas reported tumor stabilization in 41 (87%) patients after a minimum 6-month follow-up; 1 patient developed temporal lobe necrosis, 3 developed new significant visual deficits, and 11 developed new hypopituitarism [201]. These are disappointing results suggesting considerably worse outcome both in terms of efficacy and toxicity than seen with photon irradiation.

 

A study of proton beam stereotactic radiosurgery in 22 patients with acromegaly reported normalization of GH in 59% after a median of 42 months. New pituitary deficiency was reported in 38% of patients, but no visual complications were reported [43]. The same group reported on the biochemical response in a larger population of secreting adenomas (74 ACTH-secreting, 50 GH-secreting, 9 PRL-secreting, 8 Nelson’s syndromes, 3 TSH-secreting) treated with the same technique. The study included 27 patients previously irradiated (14 pts) or treated with fractionated proton beam radiotherapy. At a median follow-up of 52 months, 42% of patients did not achieve endocrine control with patients with acromegaly having the longer time to biochemical response (49% at 5 years). The risk of developing hypopituitarism was 62% at 5 years and four patients (3%) experienced post treatment temporal lobe seizures, with associated temporal lobe changes on imaging (1 month to 9 years from proton treatment). [202]).

 

An evidence-based review of proton therapy from ASTRO’s emerging technology committee examined the evidence for proton therapy across multiple tumor sites and concluded that currently available evidence provides only limited indications for proton therapy [203]). The report recommended that robust prospective clinical trials be conducted to determine the appropriate clinical indications for proton therapy. In the present context, the available published reports of proton therapy for pituitary adenoma demonstrate disappointing efficacy and increased toxicity relative to much more readily available photon-based treatment. Also, in dosimetric comparisons, proton beam did not provide superior dose coverage advantage over photon radiation with comparable doses to OARs with both modalities [204]. Therefore, it seems difficult to justify proton therapy to the pituitary outside of the context of a clinical trial.

 

RE-IRRADIATION FOR RECURRENT DISEASE

 

Re-irradiation for progression of pituitary adenoma after previous pituitary radiotherapy is considered to be associated with a high risk of radiation-induced damage due to the presumed cumulative effect of radiation to the optic apparatus, the cranial nerves, and the normal brain tissues. However, re-irradiation using fractionated conventional or stereotactic techniques is feasible, with acceptable toxicity [53], provided that there has been at least a 3-4 year gap following primary radiotherapy treatment to doses below radiation tolerance of the CNS (which is the case for the conventional dose of 45Gy delivered at <1.8Gy per fraction). GK SRS has also been used to re-irradiate small recurrent lesions, particularly if they are not in close proximity to the optic apparatus [205].

 

While the current impression is that late toxicity following pituitary re-irradiation is uncommon, a high incidence of adverse side effects (13% radiation induced optic neuropathy and 13% of temporal lobe necrosis) was reported in a series of 15 patients re-irradiated with both single fraction and fractionated approaches (median time from previous RT 5.8 years) [206]. Nonetheless, there are at present insufficient long-term data to demonstrate the safety of pituitary re-irradiation for recurrent disease, although the use of high precision techniques and fractionation should theoretically reduce late toxicity.

 

With the lack of consensus, variations in the management of pituitary recurrences are discussed in MDT meetings and decisions vary based on expertise and scope of practicing physicians. For example, in a survey study for Canadian neurosurgeons and radiation oncologists, it was observed that physicians newer to practice had a greater tendency to advocate for stereotactic radiosurgery (SRS) or re-resection (54.5% and 36.4%, respectively), as compared to older surgeons who showed a higher propensity (22.2%) to advocate for observation. The presence of cavernous sinus extension encouraged radiation oncologists to offer earlier radiotherapy sooner (61.4%), compared to 40% of neurosurgeons [207].

 

OUTLOOK

 

The techniques of pituitary radiotherapy have gradually evolved over a number of decades with apparent choice between different technologies. All technologies share the aim of concentrating the radiation dose to the tumor with minimal dose to surrounding tissue and the irradiation is given in one, few or many fractions. There has been a lack of randomized comparative studies comparing the techniques to date. Systematic review of case series reported in the literature assessing the efficacy and toxicity provides a reasonably objective assessment of the techniques. While prospective randomized trials would provide the best objective comparative information, the beliefs of practitioners in particular treatment modalities, vested interests in technologies, and general difficulty of conducting studies in diseases with such long natural history make such comparative trials an unlikely prospect. This is compounded by the fact that new radiotherapy technologies continue to be introduced into clinical practice without the need for establishing efficacy as demanded for new drugs. Therefore, controversy will persist with regard to the appropriate and optimal methods for treating pituitary adenomas using radiation, and that all of the treatment modalities described here will continue in clinical use for the foreseeable future despite systematic reviews suggesting that some of the techniques may be less effective and potentially more toxic.

 

Conformal techniques of fractionated pituitary radiotherapy are standard practice, with many centers able to offer the additional accuracy of higher precision radiotherapy previously termed stereotactic but currently part of mainstream high-precision RT. Successful application of high-precision treatment is highly dependent on expertise in accurate target definition using modern MR imaging, on the precision of the immobilization system based on an exhaustive quality assurance program, and on infrastructure particularly in the form of expertise of staff in complex techniques of treatment planning and delivery. It seems most likely that it is the available expertise at all levels of staff in a treatment center that is the principal determinant of the success of pituitary radiotherapy rather than the choice of equipment and the precise treatment technique that is used.

 

SUMMARY

 

Fractionated radiotherapy is an effective treatment for pituitary adenomas, able to achieve excellent disease control and normalization of hormone levels. While the overall safety profile of this treatment modality is favorable, it is not devoid of side effects and it should only be employed when the risks from the disease itself are considered to outweigh the risks from the treatment. The balance of risks should take into account not only the early consequences of the disease and treatment, measured in terms of disease control and immediate morbidity, but also the long-term effects, particularly in terms of the influence of treatment on survival and quality of life, both of which are less well defined.

 

Residual pituitary adenomas, most of which have an indolent natural history, pose little threat to function, unless they lie close to the optic apparatus, or unless they destructively invade adjacent structures, which is an uncommon event. The risks of residual adenoma are therefore often minimal, and in the absence of progression or hormone hypersecretion, there is currently little justification for adjuvant radiation, whether in the form of fractionated or single fraction treatment. However, a policy of postoperative surveillance does require a program of close monitoring, usually in the form of annual MR imaging, and proceeding to timely irradiation if necessary, and certainly well before the need for further surgery. The aim of radiation treatment is to arrest tumor growth without the risks of re-operation.

 

For functioning tumors radiation treatment is generally offered to patients with persistent hormone elevation that is not decreasing at the expected rate following previous intervention of surgery and medical therapy. This usually means persistent hormone elevation in patients with acromegaly, Cushing’s disease, and other functioning adenomas, regardless of how far the actual hormone level is from normal, as the aim in most cases is to achieve normalization. In patients with acromegaly treated with somatostatin analogues, the expense and inconvenience of protracted systemic treatment also warrants early radiation treatment to allow for the withdrawal of medical treatment. The alternative is to continue medical management indefinitely without radiotherapy. It is not clear at present which policy is associated with better long-term survival and quality of life, and this should ideally be the subject of a prospective randomized trial.

 

Current clinical practice is therefore to offer treatment to patients with progressive non-functioning (or functioning) pituitary adenomas considered to be a threat to function, and to patients with functioning pituitary adenomas with persistent hypersecretion. Fractionated radiotherapy, as high-precision IMRT (previously considered as SCRT/fSRT), is the current standard of care for patients requiring radiation treatment for pituitary adenoma. Single fraction radiosurgery can be considered to treat small adenomas away from critical structures in view of the significant risk of radiation-induced damage carried by a high single dose of radiation. Long-term follow-up data are needed to fully evaluate the clinical efficacy of single fraction radiosurgery in comparison with fractionated radiotherapy.

 

ACKNOWLEDGEMENTS

 

MK and NF would like to thank Dr Francesca Solda, Dr Liam Welsh, Dr Thankamma Ajithkumar and Professor Michael Brada who authored previous versions of this review. MK is funded by the NIHR Biomedical Research Centre at University College London Hospitals NHS Foundation Trust and University College London.

 

REFERENCES

 

  1. Surawicz, T.S., et al., Descriptive epidemiology of primary brain and CNS tumors: results from the Central Brain Tumor Registry of the United States, 1990-1994. Neuro Oncol, 1999. 1(1): p. 14-25.
  2. Ezzat, S., et al., The prevalence of pituitary adenomas: a systematic review. Cancer, 2004. 101(3): p. 613-9.
  3. Beclere, J., The radiotherapeutic treatment of tumours of the hypophysis, gigantism and acromegaly. Archives of the Roentgen Ray, 1909. III(1): p. 8.
  4. Turner, H.E., et al., Audit of selected patients with nonfunctioning pituitary adenomas treated without irradiation - a follow-up study. Clin Endocrinol (Oxf), 1999. 51(3): p. 281-4.
  5. Gittoes, N.J., et al., Radiotherapy for non-function pituitary tumours. Clin Endocrinol (Oxf), 1998. 48(3): p. 331-7.
  6. Grigsby, P.W., et al., Prognostic factors and results of surgery and postoperative irradiation in the management of pituitary adenomas. Int J Radiat Oncol Biol Phys, 1989. 16(6): p. 1411-7.
  7. McCollough, W.M., et al., Long-term follow-up of radiotherapy for pituitary adenoma: the absence of late recurrence after greater than or equal to 4500 cGy. Int J Radiat Oncol Biol Phys, 1991. 21(3): p. 607-14.
  8. Brada, M., et al., The long-term efficacy of conservative surgery and radiotherapy in the control of pituitary adenomas. Clin Endocrinol (Oxf), 1993. 38(6): p. 571-8.
  9. Tsang, R.W., et al., Radiation therapy for pituitary adenoma: treatment outcome and prognostic factors. Int J Radiat Oncol Biol Phys, 1994. 30(3): p. 557-65.
  10. Zierhut, D., et al., External radiotherapy of pituitary adenomas. Int J Radiat Oncol Biol Phys, 1995. 33(2): p. 307-14.
  11. Rush, S. and P.R. Cooper, Symptom resolution, tumor control, and side effects following postoperative radiotherapy for pituitary macroadenomas. Int J Radiat Oncol Biol Phys, 1997. 37(5): p. 1031-4.
  12. Breen, P., et al., Radiotherapy for nonfunctional pituitary adenoma: analysis of long-term tumor control. J Neurosurg, 1998. 89(6): p. 933-8.
  13. Sasaki, R., et al., The efficacy of conventional radiation therapy in the management of pituitary adenoma. Int J Radiat Oncol Biol Phys, 2000. 47(5): p. 1337-45.
  14. Minniti, G., D.C. Gilbert, and M. Brada, Modern techniques for pituitary radiotherapy. Rev Endocr Metab Disord, 2009. 10(2): p. 135-44.
  15. Khoo, V.S., et al., Comparison of intensity-modulated tomotherapy with stereotactically guided conformal radiotherapy for brain tumors. Int J Radiat Oncol Biol Phys, 1999. 45(2): p. 415-25.
  16. Karger, C.P., et al., Three-dimensional accuracy and interfractional reproducibility of patient fixation and positioning using a stereotactic head mask system. Int J Radiat Oncol Biol Phys, 2001. 49(5): p. 1493-504.
  17. Ajithkumar, T. and M. Brada, Stereotactic linear accelerator radiotherapy for pituitary tumors. Treat Endocrinol, 2004. 3(4): p. 211-6.
  18. Leksell, L., The stereotaxic method and radiosurgery of the brain. Acta Chir Scand, 1951. 102(4): p. 316-9.
  19. Levy, R., A Short History of Stereotactic Neurosurgery. Cyber Museum of neurosurgery. 1999. Available from: http://www.neurosurgery.org/cybermuseum/stereotactichall/stereoarticle.html.
  20. Lasak, J.M. and J.P. Gorecki, The history of stereotactic radiosurgery and radiotherapy. Otolaryngol Clin North Am, 2009. 42(4): p. 593-9.
  21. Gill, S.S., et al., Relocatable frame for stereotactic external beam radiotherapy. Int J Radiat Oncol Biol Phys, 1991. 20(3): p. 599-603.
  22. Graham, J.D., et al., A non-invasive, relocatable stereotactic frame for fractionated radiotherapy and multiple imaging. Radiother Oncol, 1991. 21(1): p. 60-2.
  23. Adler, J.R., Jr., et al., The Cyberknife: a frameless robotic system for radiosurgery. Stereotact Funct Neurosurg, 1997. 69(1-4 Pt 2): p. 124-8.
  24. Steel, G.G., Basic Clinical Radiobiology. 3 ed. Vol. Arnold. 2002, London.
  25. Larson, D.A., J.C. Flickinger, and J.S. Loeffler, The radiobiology of radiosurgery. Int J Radiat Oncol Biol Phys, 1993. 25(3): p. 557-61.
  26. Park, C., et al., Universal survival curve and single fraction equivalent dose: useful tools in understanding potency of ablative radiotherapy. Int J Radiat Oncol Biol Phys, 2008. 70(3): p. 847-52.
  27. Lawrence, Y.R., et al., Radiation dose-volume effects in the brain. Int J Radiat Oncol Biol Phys, 2010. 76(3 Suppl): p. S20-7.
  28. Mayo, C., et al., Radiation dose-volume effects of optic nerves and chiasm. Int J Radiat Oncol Biol Phys, 2010. 76(3 Suppl): p. S28-35.
  29. Khattab, M.H., et al., Stereotactic Radiosurgery and Hypofractionated Stereotactic Radiotherapy for Nonfunctioning Pituitary Adenoma. J Neurol Surg B Skull Base, 2021. 82(Suppl 3): p. e51-e58.
  30. Rosenfelder, N.A., et al., Achieving the relocation accuracy of stereotactic frame-based cranial radiotherapy in a three-point thermoplastic shell. Clin Oncol (R Coll Radiol), 2013. 25(1): p. 66-73.
  31. Kumar, S., et al., Treatment accuracy of fractionated stereotactic radiotherapy. Radiother Oncol, 2005. 74(1): p. 53-9.
  32. Veselsky, T., et al., Pituitary adenoma treatment plan quality comparison between linear accelerator volumetric modulated arc therapy and Leksell Gamma Knife(R) radiosurgery. Med Dosim, 2021. 46(4): p. 440-448.
  33. Sayer, F.T., et al., Initial experience with the eXtend System: a relocatable frame system for multiple-session gamma knife radiosurgery. World Neurosurg, 2011. 75(5-6): p. 665-72.
  34. Zhao, K., et al., Fractionated Gamma Knife surgery for giant pituitary adenomas. Clin Neurol Neurosurg, 2016. 150: p. 139-142.
  35. Ikeda, H., H. Jokura, and T. Yoshimoto, Gamma knife radiosurgery for pituitary adenomas: usefulness of combined transsphenoidal and gamma knife radiosurgery for adenomas invading the cavernous sinus. Radiat Oncol Investig, 1998. 6(1): p. 26-34.
  36. Hayashi, M., et al., Gamma Knife robotic microradiosurgery of pituitary adenomas invading the cavernous sinus: treatment concept and results in 89 cases. J Neurooncol, 2010. 98(2): p. 185-94.
  37. Kim, E.H., et al., Postoperative Gamma Knife Radiosurgery for Cavernous Sinus-Invading Growth Hormone-Secreting Pituitary Adenomas. World Neurosurg, 2018. 110: p. e534-e545.
  38. Hayashi, M., et al., Gamma Knife Radiosurgery for Pituitary Adenomas Invading the Cavernous Sinus: Tokyo Women's Medical University Experience. Acta Neurochir Suppl, 2021. 128: p. 29-41.
  39. Lee, W.J., et al., Gamma Knife Radiosurgery as a Primary Treatment for Nonfunctioning Pituitary Adenoma Invading the Cavernous Sinus. Stereotact Funct Neurosurg, 2020. 98(6): p. 371-377.
  40. Sheehan, J.P., et al., Gamma Knife surgery for pituitary adenomas: factors related to radiological and endocrine outcomes. J Neurosurg, 2011. 114(2): p. 303-9.
  41. Grant, R.A., et al., Efficacy and safety of higher dose stereotactic radiosurgery for functional pituitary adenomas: a preliminary report. World Neurosurg, 2014. 82(1-2): p. 195-201.
  42. Kjellberg, R.N., et al., Proton-beam therapy in acromegaly. N Engl J Med, 1968. 278(13): p. 689-95.
  43. Petit, J.H., et al., Proton stereotactic radiosurgery in management of persistent acromegaly. Endocr Pract, 2007. 13(7): p. 726-34.
  44. Durante, M. and J.S. Loeffler, Charged particles in radiation oncology. Nat Rev Clin Oncol, 2010. 7(1): p. 37-43.
  45. Specialised Commissioning Team, N.E. B01/P/c Proton Beam Therapy for Cancer in Teenagers and Young Adults 2015; Available from: https://www.england.nhs.uk/commissioning/wp-content/uploads/sites/12/2015/10/b01-pc-prtn-bm-thrpy-teens-yng-oct15.pdf.
  46. Raverot, G., et al., European Society of Endocrinology Clinical Practice Guidelines for the management of aggressive pituitary tumours and carcinomas. Eur J Endocrinol, 2018. 178(1): p. G1-G24.
  47. Giuffrida, G., et al., Peptide receptor radionuclide therapy for aggressive pituitary tumors: a monocentric experience. Endocr Connect, 2019. 8(5): p. 528-535.
  48. Maclean, J., et al., Peptide receptor radionuclide therapy for aggressive atypical pituitary adenoma/carcinoma: variable clinical response in preliminary evaluation. Pituitary, 2014. 17(6): p. 530-8.
  49. Page, R.C., et al., An account of the quality of life of patients after treatment for non-functioning pituitary tumours. Clin Endocrinol (Oxf), 1997. 46(4): p. 401-6.
  50. van Beek, A.P., et al., Radiotherapy is not associated with reduced quality of life and cognitive function in patients treated for nonfunctioning pituitary adenoma. Int J Radiat Oncol Biol Phys, 2007. 68(4): p. 986-91.
  51. Yang, C.J., et al., Symptom distress and quality of life after stereotactic radiosurgery in patients with pituitary tumors: a questionnaire survey. PLoS One, 2014. 9(2): p. e88460.
  52. Jenkins, P.J., et al., Conventional pituitary irradiation is effective in lowering serum growth hormone and insulin-like growth factor-I in patients with acromegaly. J Clin Endocrinol Metab, 2006. 91(4): p. 1239-45.
  53. Brada, M. and P. Jankowska, Radiotherapy for pituitary adenomas. Endocrinol Metab Clin North Am, 2008. 37(1): p. 263-75, xi.
  54. Rowland, N.C. and M.K. Aghi, Radiation treatment strategies for acromegaly. Neurosurg Focus, 2010. 29(4): p. E12.
  55. Brada, M., T.V. Ajithkumar, and G. Minniti, Radiosurgery for pituitary adenomas. Clin Endocrinol (Oxf), 2004. 61(5): p. 531-43.
  56. Estrada, J., et al., The long-term outcome of pituitary irradiation after unsuccessful transsphenoidal surgery in Cushing's disease. N Engl J Med, 1997. 336(3): p. 172-7.
  57. Barrande, G., et al., Hormonal and metabolic effects of radiotherapy in acromegaly: long-term results in 128 patients followed in a single center. J Clin Endocrinol Metab, 2000. 85(10): p. 3779-85.
  58. Biermasz, N.R., H.V. Dulken, and F. Roelfsema, Postoperative radiotherapy in acromegaly is effective in reducing GH concentration to safe levels. Clin Endocrinol (Oxf), 2000. 53(3): p. 321-7.
  59. Epaminonda, P., et al., Efficacy of radiotherapy in normalizing serum IGF-I, acid-labile subunit (ALS) and IGFBP-3 levels in acromegaly. Clin Endocrinol (Oxf), 2001. 55(2): p. 183-9.
  60. Minniti, G., et al., The long-term efficacy of conventional radiotherapy in patients with GH-secreting pituitary adenomas. Clin Endocrinol (Oxf), 2005. 62(2): p. 210-6.
  61. Langsenlehner, T., et al., Long-term follow-up of patients with pituitary macroadenomas after postoperative radiation therapy: analysis of tumor control and functional outcome. Strahlenther Onkol, 2007. 183(5): p. 241-7.
  62. Minniti, G., et al., Long-term follow-up results of postoperative radiation therapy for Cushing's disease. J Neurooncol, 2007. 84(1): p. 79-84.
  63. Rim, C.H., et al., Radiotherapy for pituitary adenomas: long-term outcome and complications. Radiat Oncol J, 2011. 29(3): p. 156-63.
  64. Ajithkumar, T.B., M., Pituitary radiotherapy. Oxford Textbook of Endocrinology and Diabetes. 2011: Oxford Press University.
  65. Kim, M.Y., et al., Long-term outcomes of surgery and radiotherapy for secreting and non-secreting pituitary adenoma. Radiation Oncology Journal, 2016. 34(2): p. 121-127.
  66. Patt, H., et al., High-Precision Conformal Fractionated Radiotherapy Is Effective in Achieving Remission in Patients with Acromegaly after Failed Transsphenoidal Surgery. Endocr Pract, 2016. 22(2): p. 162-72.
  67. Hussein, Z., et al., Non-functioning pituitary macroadenoma following surgery: long-term outcomes and development of an optimal follow-up strategy. Front Surg, 2023. 10: p. 1129387.
  68. Arnardottir, S., et al., Long-term outcomes of patients with acromegaly: a report from the Swedish Pituitary Register. Eur J Endocrinol, 2022. 186(3): p. 329-339.
  69. Tsagarakis, S., et al., Megavoltage pituitary irradiation in the management of prolactinomas: long-term follow-up. Clin Endocrinol (Oxf), 1991. 34(5): p. 399-406.
  70. Johnston, D.G., et al., The long-term effects of megavoltage radiotherapy as sole or combined therapy for large prolactinomas: studies with high definition computerized tomography. Clin Endocrinol (Oxf), 1986. 24(6): p. 675-85.
  71. Mehta, A.E., F.I. Reyes, and C. Faiman, Primary radiotherapy of prolactinomas. Eight- to 15-year follow-up. Am J Med, 1987. 83(1): p. 49-58.
  72. Becker, G., et al., Radiation therapy in the multimodal treatment approach of pituitary adenoma. Strahlenther Onkol, 2002. 178(4): p. 173-86.
  73. Sherlock, M., et al., Mortality in patients with pituitary disease. Endocr Rev, 2010. 31(3): p. 301-42.
  74. Brada, M., et al., Cerebrovascular mortality in patients with pituitary adenoma. Clin Endocrinol (Oxf), 2002. 57(6): p. 713-7.
  75. Brada, M., et al., The incidence of cerebrovascular accidents in patients with pituitary adenoma. Int J Radiat Oncol Biol Phys, 1999. 45(3): p. 693-8.
  76. Tomlinson, J.W., et al., Association between premature mortality and hypopituitarism. West Midlands Prospective Hypopituitary Study Group. Lancet, 2001. 357(9254): p. 425-31.
  77. Erfurth, E.M., et al., Risk factors for cerebrovascular deaths in patients operated and irradiated for pituitary tumors. J Clin Endocrinol Metab, 2002. 87(11): p. 4892-9.
  78. Fajardo, L.F., The pathology of ionizing radiation as defined by morphologic patterns. Acta Oncol, 2005. 44(1): p. 13-22.
  79. Bliss, P., G.R. Kerr, and A. Gregor, Incidence of second brain tumours after pituitary irradiation in Edinburgh 1962-1990. Clin Oncol (R Coll Radiol), 1994. 6(6): p. 361-3.
  80. Tsang, R.W., et al., Glioma arising after radiation therapy for pituitary adenoma. A report of four patients and estimation of risk. Cancer, 1993. 72(7): p. 2227-33.
  81. Brada, M., et al., Risk of second brain tumour after conservative surgery and radiotherapy for pituitary adenoma. BMJ, 1992. 304(6838): p. 1343-6.
  82. Hamblin, R., et al., Risk of second brain tumour after radiotherapy for pituitary adenoma or craniopharyngioma: a retrospective, multicentre, cohort study of 3679 patients with long-term imaging surveillance. Lancet Diabetes Endocrinol, 2022. 10(8): p. 581-588.
  83. Peace, K.A., et al., Cognitive dysfunction in patients with pituitary tumour who have been treated with transfrontal or transsphenoidal surgery or medication. Clin Endocrinol (Oxf), 1998. 49(3): p. 391-6.
  84. Guinan, E.M., et al., Cognitive effects of pituitary tumours and their treatments: two case studies and an investigation of 90 patients. J Neurol Neurosurg Psychiatry, 1998. 65(6): p. 870-6.
  85. McCord, M.W., et al., Radiotherapy for pituitary adenoma: long-term outcome and sequelae. Int J Radiat Oncol Biol Phys, 1997. 39(2): p. 437-44.
  86. Brummelman, P., et al., Cognitive functioning in patients treated for nonfunctioning pituitary macroadenoma and the effects of pituitary radiotherapy. Clin Endocrinol (Oxf), 2011. 74(4): p. 481-7.
  87. Brummelman, P., et al., Cognitive performance after postoperative pituitary radiotherapy: a dosimetric study of the hippocampus and the prefrontal cortex. Eur J Endocrinol, 2012. 166(2): p. 171-9.
  88. Coke, C., et al., Multiple fractionated stereotactic radiotherapy of residual pituitary macroadenomas: initial experience. Stereotact Funct Neurosurg, 1997. 69(1-4 Pt 2): p. 183-90.
  89. Mitsumori, M., et al., Initial clinical results of LINAC-based stereotactic radiosurgery and stereotactic radiotherapy for pituitary adenomas. Int J Radiat Oncol Biol Phys, 1998. 42(3): p. 573-80.
  90. Milker-Zabel, S., et al., Fractionated stereotactically guided radiotherapy and radiosurgery in the treatment of functional and nonfunctional adenomas of the pituitary gland. Int J Radiat Oncol Biol Phys, 2001. 50(5): p. 1279-86.
  91. Paek, S.H., et al., Integration of surgery with fractionated stereotactic radiotherapy for treatment of nonfunctioning pituitary macroadenomas. Int J Radiat Oncol Biol Phys, 2005. 61(3): p. 795-808.
  92. Colin, P., et al., Treatment of pituitary adenomas by fractionated stereotactic radiotherapy: a prospective study of 110 patients. Int J Radiat Oncol Biol Phys, 2005. 62(2): p. 333-41.
  93. Minniti, G., et al., Fractionated stereotactic conformal radiotherapy for secreting and nonsecreting pituitary adenomas. Clin Endocrinol (Oxf), 2006. 64(5): p. 542-8.
  94. Selch, M.T., et al., Stereotactic radiotherapy for the treatment of pituitary adenomas. Minim Invasive Neurosurg, 2006. 49(3): p. 150-5.
  95. Kong, D.S., et al., The efficacy of fractionated radiotherapy and stereotactic radiosurgery for pituitary adenomas: long-term results of 125 consecutive patients treated in a single institution. Cancer, 2007. 110(4): p. 854-60.
  96. Snead, F.E., et al., Long-term outcomes of radiotherapy for pituitary adenomas. Int J Radiat Oncol Biol Phys, 2008. 71(4): p. 994-8.
  97. Roug, S., et al., Fractionated stereotactic radiotherapy in patients with acromegaly: an interim single-centre audit. Eur J Endocrinol, 2010. 162(4): p. 685-94.
  98. Schalin-Jantti, C., et al., Outcome of fractionated stereotactic radiotherapy in patients with pituitary adenomas resistant to conventional treatments: a 5.25-year follow-up study. Clin Endocrinol (Oxf), 2010. 73(1): p. 72-7.
  99. Weber, D.C., et al., Adjuvant or radical fractionated stereotactic radiotherapy for patients with pituitary functional and nonfunctional macroadenoma. Radiat Oncol, 2011. 6: p. 169.
  100. Wilson, P.J., et al., A single centre's experience of stereotactic radiosurgery and radiotherapy for non-functioning pituitary adenomas with the Linear Accelerator (Linac). J Clin Neurosci, 2012. 19(3): p. 370-4.
  101. Kim, J.O., et al., Long-term outcomes of fractionated stereotactic radiation therapy for pituitary adenomas at the BC Cancer Agency. Int J Radiat Oncol Biol Phys, 2013. 87(3): p. 528-33.
  102. Kopp, C., et al., Fractionated stereotactic radiotherapy in the treatment of pituitary adenomas. Strahlenther Onkol, 2013. 189(11): p. 932-7.
  103. Minniti, G., et al., Fractionated stereotactic radiotherapy for large and invasive non-functioning pituitary adenomas: long-term clinical outcomes and volumetric MRI assessment of tumor response. Eur J Endocrinol, 2015. 172(4): p. 433-41.
  104. Diallo, A.M., et al., Long-term results of fractionated stereotactic radiotherapy as third-line treatment in acromegaly. Endocrine, 2015. 50(3): p. 741-8.
  105. Barber, S.M., B.S. Teh, and D.S. Baskin, Fractionated Stereotactic Radiotherapy for Pituitary Adenomas: Single-Center Experience in 75 Consecutive Patients. Neurosurgery, 2016. 79(3): p. 406-17.
  106. Liao, H.I., et al., Fractionated stereotactic radiosurgery using the Novalis system for the management of pituitary adenomas close to the optic apparatus. J Clin Neurosci, 2014. 21(1): p. 111-5.
  107. Puataweepong, P., et al., Outcomes for Pituitary Adenoma Patients Treated with Linac- Based Stereotactic Radiosurgery and Radiotherapy: a Long Term Experience in Thailand. Asian Pac J Cancer Prev, 2015. 16(13): p. 5279-84.
  108. Lian, X., et al., Intensity-modulated Radiotherapy for Pituitary Somatotroph Adenomas. J Clin Endocrinol Metab, 2020. 105(12).
  109. Martinez, R., et al., Pituitary tumors and gamma knife surgery. Clinical experience with more than two years of follow-up. Stereotact Funct Neurosurg, 1998. 70 Suppl 1: p. 110-8.
  110. Pan, L., et al., Pituitary adenomas: the effect of gamma knife radiosurgery on tumor growth and endocrinopathies. Stereotact Funct Neurosurg, 1998. 70 Suppl 1: p. 119-26.
  111. Mokry, M., et al., A six year experience with the postoperative radiosurgical management of pituitary adenomas. Stereotact Funct Neurosurg, 1999. 72 Suppl 1: p. 88-100.
  112. Sheehan, J.P., et al., Radiosurgery for residual or recurrent nonfunctioning pituitary adenoma. J Neurosurg, 2002. 97(5 Suppl): p. 408-14.
  113. Wowra, B. and W. Stummer, Efficacy of gamma knife radiosurgery for nonfunctioning pituitary adenomas: a quantitative follow up with magnetic resonance imaging-based volumetric analysis. J Neurosurg, 2002. 97(5 Suppl): p. 429-32.
  114. Petrovich, Z., et al., Gamma knife radiosurgery for pituitary adenoma: early results. Neurosurgery, 2003. 53(1): p. 51-9; discussion 59-61.
  115. Pollock, B.E. and P.C. Carpenter, Stereotactic radiosurgery as an alternative to fractionated radiotherapy for patients with recurrent or residual nonfunctioning pituitary adenomas. Neurosurgery, 2003. 53(5): p. 1086-91; discussion 1091-4.
  116. Losa, M., et al., Gamma knife surgery for treatment of residual nonfunctioning pituitary adenomas after surgical debulking. J Neurosurg, 2004. 100(3): p. 438-44.
  117. Iwai, Y., K. Yamanaka, and K. Yoshioka, Radiosurgery for nonfunctioning pituitary adenomas. Neurosurgery, 2005. 56(4): p. 699-705; discussion 699-705.
  118. Mingione, V., et al., Gamma surgery in the treatment of nonsecretory pituitary macroadenoma. J Neurosurg, 2006. 104(6): p. 876-83.
  119. Liscak, R., et al., Gamma knife radiosurgery for endocrine-inactive pituitary adenomas. Acta Neurochir (Wien), 2007. 149(10): p. 999-1006; discussion 1006.
  120. Pollock, B.E., et al., Pituitary tumor type affects the chance of biochemical remission after radiosurgery of hormone-secreting pituitary adenomas. Neurosurgery, 2008. 62(6): p. 1271-6; discussion 1276-8.
  121. Kobayashi, T., Long-term results of stereotactic gamma knife radiosurgery for pituitary adenomas. Specific strategies for different types of adenoma. Prog Neurol Surg, 2009. 22: p. 77-95.
  122. Gopalan, R., et al., Long-term outcomes after Gamma Knife radiosurgery for patients with a nonfunctioning pituitary adenoma. Neurosurgery, 2011. 69(2): p. 284-93.
  123. Runge, M.J., et al., LINAC-radiosurgery for nonsecreting pituitary adenomas. Long-term results. Strahlenther Onkol, 2012. 188(4): p. 319-25.
  124. Park, K.J., et al., Long-term outcomes after gamma knife stereotactic radiosurgery for nonfunctional pituitary adenomas. Neurosurgery, 2011. 69(6): p. 1188-99.
  125. Starke, R.M., et al., Gamma Knife surgery for patients with nonfunctioning pituitary macroadenomas: predictors of tumor control, neurological deficits, and hypopituitarism. J Neurosurg, 2012. 117(1): p. 129-35.
  126. El-Shehaby, A.M., et al., Low-dose Gamma Knife surgery for nonfunctioning pituitary adenomas. J Neurosurg, 2012. 117 Suppl: p. 84-8.
  127. Sheehan, J.P., et al., Gamma Knife radiosurgery for the management of nonfunctioning pituitary adenomas: a multicenter study. J Neurosurg, 2013. 119(2): p. 446-56.
  128. Lee, C.C., et al., Initial Gamma Knife radiosurgery for nonfunctioning pituitary adenomas. J Neurosurg, 2014. 120(3): p. 647-54.
  129. Xu, Z., et al., Silent corticotroph adenomas after stereotactic radiosurgery: a case-control study. Int J Radiat Oncol Biol Phys, 2014. 90(4): p. 903-10.
  130. Hasegawa, T., et al., Stereotactic Radiosurgery as the Initial Treatment for Patients with Nonfunctioning Pituitary Adenomas. World Neurosurg, 2015. 83(6): p. 1173-9.
  131. Graffeo, C.S., et al., Hypopituitarism After Single-Fraction Pituitary Adenoma Radiosurgery: Dosimetric Analysis Based on Patients Treated Using Contemporary Techniques. Int J Radiat Oncol Biol Phys, 2018. 101(3): p. 618-623.
  132. Oh, J.W., et al., Hypopituitarism after Gamma Knife surgery for postoperative nonfunctioning pituitary adenoma. J Neurosurg, 2018. 129(Suppl1): p. 47-54.
  133. Cordeiro, D., et al., Hypopituitarism after Gamma Knife radiosurgery for pituitary adenomas: a multicenter, international study. J Neurosurg, 2018.
  134. Narayan, V., et al., Long-Term Outcome of Nonfunctioning and Hormonal Active Pituitary Adenoma After Gamma Knife Radiosurgery. World Neurosurg, 2018. 114: p. e824-e832.
  135. Slavinsky, P., et al., Gamma knife radiosurgery in pituitary adenomas. A single-center experience. Medicina (B Aires), 2022. 82(1): p. 111-116.
  136. Maldar, A.N., et al., Long-term hormonal and imaging outcomes of adjunctive gamma knife radiosurgery in non-functioning pituitary adenomas: a single center experience. J Neurooncol, 2022. 158(3): p. 423-433.
  137. Thoren, M., et al., Stereotactic radiosurgery with the cobalt-60 gamma unit in the treatment of growth hormone-producing pituitary tumors. Neurosurgery, 1991. 29(5): p. 663-8.
  138. Morange-Ramos, I., et al., Short-term endocrinological results after gamma knife surgery of pituitary adenomas. Stereotact Funct Neurosurg, 1998. 70 Suppl 1: p. 127-38.
  139. Lim, Y.L., et al., Four years' experiences in the treatment of pituitary adenomas with gamma knife radiosurgery. Stereotact Funct Neurosurg, 1998. 70 Suppl 1: p. 95-109.
  140. Kim, M.S., S.I. Lee, and J.H. Sim, Gamma Knife radiosurgery for functioning pituitary microadenoma. Stereotact Funct Neurosurg, 1999. 72 Suppl 1: p. 119-24.
  141. Landolt, A.M., et al., Stereotactic radiosurgery for recurrent surgically treated acromegaly: comparison with fractionated radiotherapy. J Neurosurg, 1998. 88(6): p. 1002-8.
  142. Hayashi, M., et al., Gamma Knife radiosurgery for pituitary adenomas. Stereotact Funct Neurosurg, 1999. 72 Suppl 1: p. 111-8.
  143. Inoue, H.K., et al., Pituitary adenomas treated by microsurgery with or without Gamma Knife surgery: experience in 122 cases. Stereotact Funct Neurosurg, 1999. 72 Suppl 1: p. 125-31.
  144. Zhang, N., et al., Radiosurgery for growth hormone-producing pituitary adenomas. J Neurosurg, 2000. 93 Suppl 3: p. 6-9.
  145. Izawa, M., et al., Gamma knife radiosurgery for pituitary adenomas. J Neurosurg, 2000. 93 Suppl 3: p. 19-22.
  146. Pollock, B.E., et al., Results of stereotactic radiosurgery in patients with hormone-producing pituitary adenomas: factors associated with endocrine normalization. J Neurosurg, 2002. 97(3): p. 525-30.
  147. Attanasio, R., et al., Gamma-knife radiosurgery in acromegaly: a 4-year follow-up study. J Clin Endocrinol Metab, 2003. 88(7): p. 3105-12.
  148. Choi, J.Y., et al., Radiological and hormonal responses of functioning pituitary adenomas after gamma knife radiosurgery. Yonsei Med J, 2003. 44(4): p. 602-7.
  149. Jane, J.A., Jr. and E.R. Laws, Jr., The management of non-functioning pituitary adenomas. Neurol India, 2003. 51(4): p. 461-5.
  150. Castinetti, F., et al., Outcome of gamma knife radiosurgery in 82 patients with acromegaly: correlation with initial hypersecretion. J Clin Endocrinol Metab, 2005. 90(8): p. 4483-8.
  151. Gutt, B., et al., Gamma-knife surgery is effective in normalising plasma insulin-like growth factor I in patients with acromegaly. Exp Clin Endocrinol Diabetes, 2005. 113(4): p. 219-24.
  152. Kobayashi, T., et al., Long-term results of gamma knife surgery for growth hormone-producing pituitary adenoma: is the disease difficult to cure? J Neurosurg, 2005. 102 Suppl: p. 119-23.
  153. Jezkova, J., et al., Gamma knife radiosurgery for acromegaly--long-term experience. Clin Endocrinol (Oxf), 2006. 64(5): p. 588-95.
  154. Pollock, B.E., et al., Radiosurgery of growth hormone-producing pituitary adenomas: factors associated with biochemical remission. J Neurosurg, 2007. 106(5): p. 833-8.
  155. Jagannathan, J., et al., Stereotactic radiosurgery for pituitary adenomas: a comprehensive review of indications, techniques and long-term results using the Gamma Knife. J Neurooncol, 2009. 92(3): p. 345-56.
  156. Wan, H., O. Chihiro, and S. Yuan, MASEP gamma knife radiosurgery for secretory pituitary adenomas: experience in 347 consecutive cases. J Exp Clin Cancer Res, 2009. 28: p. 36.
  157. Castinetti, F., et al., Long-term results of stereotactic radiosurgery in secretory pituitary adenomas. J Clin Endocrinol Metab, 2009. 94(9): p. 3400-7.
  158. Iwai, Y., et al., Gamma knife radiosurgery for growth hormone-producing adenomas. J Clin Neurosci, 2010. 17(3): p. 299-304.
  159. Erdur, F.M., et al., Gammaknife radiosurgery in patients with acromegaly. J Clin Neurosci, 2011. 18(12): p. 1616-20.
  160. Franzin, A., et al., Results of gamma knife radiosurgery in acromegaly. Int J Endocrinol, 2012. 2012: p. 342034.
  161. Liu, X., et al., Gamma knife radiosurgery for clinically persistent acromegaly. J Neurooncol, 2012. 109(1): p. 71-9.
  162. Zeiler, F.A., et al., Gamma knife in the treatment of pituitary adenomas: results of a single center. Can J Neurol Sci, 2013. 40(4): p. 546-52.
  163. Lee, C.C., et al., Stereotactic radiosurgery for acromegaly. J Clin Endocrinol Metab, 2014. 99(4): p. 1273-81.
  164. Jagannathan, J., et al., Gamma knife radiosurgery for acromegaly: outcomes after failed transsphenoidal surgery. Neurosurgery, 2008. 62(6): p. 1262-9; discussion 1269-70.
  165. Kim, S.H., et al., Gamma Knife radiosurgery for functioning pituitary adenomas. Stereotact Funct Neurosurg, 1999. 72 Suppl 1: p. 101-10.
  166. Gupta, A., et al., Upfront Gamma Knife radiosurgery for Cushing's disease and acromegaly: a multicenter, international study. J Neurosurg, 2018. 131(2): p. 532-538.
  167. Ding, D., et al., Stereotactic Radiosurgery for Acromegaly: An International Multicenter Retrospective Cohort Study. Neurosurgery, 2019. 84(3): p. 717-725.
  168. Degerblad, M., et al., Long-term results of stereotactic radiosurgery to the pituitary gland in Cushing's disease. Acta Endocrinol (Copenh), 1986. 112(3): p. 310-4.
  169. Ganz, J.C., E.O. Backlund, and F.A. Thorsen, The effects of Gamma Knife surgery of pituitary adenomas on tumor growth and endocrinopathies. Stereotact Funct Neurosurg, 1993. 61 Suppl 1: p. 30-7.
  170. Seo, Y., et al., Gamma knife surgery for Cushing's disease. Surg Neurol, 1995. 43(2): p. 170-5; discussion 175-6.
  171. Sheehan, J.M., et al., Radiosurgery for Cushing's disease after failed transsphenoidal surgery. J Neurosurg, 2000. 93(5): p. 738-42.
  172. Hoybye, C., et al., Adrenocorticotropic hormone-producing pituitary tumors: 12- to 22-year follow-up after treatment with stereotactic radiosurgery. Neurosurgery, 2001. 49(2): p. 284-91; discussion 291-2.
  173. Kobayashi, T., Y. Kida, and Y. Mori, Gamma knife radiosurgery in the treatment of Cushing disease: long-term results. J Neurosurg, 2002. 97(5 Suppl): p. 422-8.
  174. Devin, J.K., et al., The efficacy of linear accelerator radiosurgery in the management of patients with Cushing's disease. Stereotact Funct Neurosurg, 2004. 82(5-6): p. 254-62.
  175. Castinetti, F., et al., Gamma knife radiosurgery is a successful adjunctive treatment in Cushing's disease. Eur J Endocrinol, 2007. 156(1): p. 91-8.
  176. Marek, J., et al., Gamma knife radiosurgery for Cushing's disease and Nelson's syndrome. Pituitary, 2015. 18(3): p. 376-84.
  177. Sheehan, J.P., et al., Results of gamma knife surgery for Cushing's disease. J Neurosurg, 2013. 119(6): p. 1486-92.
  178. Sicignano, G., et al., Dosimetric factors associated with pituitary function after Gamma Knife Surgery (GKS) of pituitary adenomas. Radiother Oncol, 2012. 104(1): p. 119-24.
  179. Jagannathan, J., et al., Gamma Knife surgery for Cushing's disease. J Neurosurg, 2007. 106(6): p. 980-7.
  180. Knappe, U.J., et al., Fractionated radiotherapy and radiosurgery in acromegaly: analysis of 352 patients from the German Acromegaly Registry. Eur J Endocrinol, 2020. 182(3): p. 275-284.
  181. Landolt, A.M. and N. Lomax, Gamma knife radiosurgery for prolactinomas. J Neurosurg, 2000. 93 Suppl 3: p. 14-8.
  182. Pan, L., et al., Gamma knife radiosurgery as a primary treatment for prolactinomas. J Neurosurg, 2000. 93 Suppl 3: p. 10-3.
  183. Pouratian, N., et al., Gamma knife radiosurgery for medically and surgically refractory prolactinomas. Neurosurgery, 2006. 59(2): p. 255-66; discussion 255-66.
  184. Jezkova, J., et al., Use of the Leksell gamma knife in the treatment of prolactinoma patients. Clin Endocrinol (Oxf), 2009. 70(5): p. 732-41.
  185. Liu, X., et al., Gamma knife stereotactic radiosurgery for drug resistant or intolerant invasive prolactinomas. Pituitary, 2013. 16(1): p. 68-75.
  186. Cohen-Inbar, O., et al., Gamma Knife radiosurgery for medically and surgically refractory prolactinomas: long-term results. Pituitary, 2015. 18(6): p. 820-30.
  187. Jezkova, J., et al., Role of gamma knife radiosurgery in the treatment of prolactinomas. Pituitary, 2019. 22(4): p. 411-421.
  188. Voges, J., et al., Linear accelerator radiosurgery for pituitary macroadenomas: a 7-year follow-up study. Cancer, 2006. 107(6): p. 1355-64.
  189. Palmisciano, P., et al., Endocrine disorders after primary gamma knife radiosurgery for pituitary adenomas: A systematic review and meta-analysis. Pituitary, 2022. 25(3): p. 404-419.
  190. Kajiwara, K., et al., Image-guided stereotactic radiosurgery with the CyberKnife for pituitary adenomas. Minim Invasive Neurosurg, 2005. 48(2): p. 91-6.
  191. Adler, J.R., Jr., et al., Visual field preservation after multisession cyberknife radiosurgery for perioptic lesions. Neurosurgery, 2006. 59(2): p. 244-54; discussion 244-54.
  192. Roberts, B.K., et al., Efficacy and safety of CyberKnife radiosurgery for acromegaly. Pituitary, 2007. 10(1): p. 17.
  193. Killory, B.D., et al., Hypofractionated CyberKnife radiosurgery for perichiasmatic pituitary adenomas: early results. Neurosurgery, 2009. 64(2 Suppl): p. A19-25.
  194. Cho, C.B., et al., Stereotactic Radiosurgery with the CyberKnife for Pituitary Adenomas. J Korean Neurosurg Soc, 2009. 45(3): p. 157-63.
  195. Iwata, H., et al., Hypofractionated stereotactic radiotherapy with CyberKnife for nonfunctioning pituitary adenoma: high local control with low toxicity. Neuro Oncol, 2011. 13(8): p. 916-22.
  196. Puataweepong, P., et al., The Clinical Outcome of Hypofractionated Stereotactic Radiotherapy With CyberKnife Robotic Radiosurgery for Perioptic Pituitary Adenoma. Technol Cancer Res Treat, 2016. 15(6): p. NP10-NP15.
  197. Iwata, H., et al., Long-term results of hypofractionated stereotactic radiotherapy with CyberKnife for growth hormone-secreting pituitary adenoma: evaluation by the Cortina consensus. J Neurooncol, 2016. 128(2): p. 267-75.
  198. Plitt, A.R., et al., Fractionated CyberKnife Stereotactic Radiotherapy for Perioptic Pituitary Adenomas. World Neurosurg, 2019. 126: p. e1359-e1364.
  199. Romero-Gameros, C.A., et al., Efficiency and Safety of CyberKnife Robotic Radiosurgery in the Multimodal Management of Patients with Acromegaly. Cancers (Basel), 2023. 15(5).
  200. Ludecke, D.K., B.S. Lutz, and G. Niedworok, The choice of treatment after incomplete adenomectomy in acromegaly: proton--versus high voltage radiation. Acta Neurochir (Wien), 1989. 96(1-2): p. 32-8.
  201. Ronson, B.B., et al., Fractionated proton beam irradiation of pituitary adenomas. Int J Radiat Oncol Biol Phys, 2006. 64(2): p. 425-34.
  202. Wattson, D.A., et al., Outcomes of proton therapy for patients with functional pituitary adenomas. Int J Radiat Oncol Biol Phys, 2014. 90(3): p. 532-9.
  203. Allen, A.M., et al., An evidence based review of proton beam therapy: the report of ASTRO's emerging technology committee. Radiother Oncol, 2012. 103(1): p. 8-11.
  204. Sud, S., et al., Dosimetric Comparison of Proton Versus Photon Radiosurgery for Treatment of Pituitary Adenoma. Adv Radiat Oncol, 2021. 6(6): p. 100806.
  205. Edwards, A.A., F.M. Swords, and P.N. Plowman, Focal radiation therapy for patients with persistent/recurrent pituitary adenoma, despite previous radiotherapy. Pituitary, 2009. 12(1): p. 30-4.
  206. Verma, J., et al., Feasibility and outcome of re-irradiation in the treatment of multiply recurrent pituitary adenomas. Pituitary, 2014. 17(6): p. 539-45.
  207. Kasper, G., et al., Practice patterns in the management of recurrent and residual non-functioning pituitary adenomas: Results from a Canada-wide survey. eNeurologicalSci, 2021. 22: p. 100317.

Endocrine Testing Protocols: Hypothalamic Pituitary Adrenal Axis

ABSTRACT

 

Abnormalities in the hypothalamic pituitary adrenal (HPA) axis are identified by a careful analysis of both direct and non-stimulated measurements of the hormones as well as provocative tests.  Dynamic testing is useful to determine if elevated levels are suppressible and whether there is sufficient hormone reserve when low levels are measured under stimulation. A combination of all these analyses can distinguish between normal physiology and the consequences of clinical disease in the HPA axis.  While clinical suspicion drives the testing performed, arrival at the correct diagnosis by laboratory testing is crucial for cure of the patient.  Knowledge of the methodologies used in measuring cortisol and ACTH and associated hormones and binding proteins is essential for correct interpretation of the tests.  In this review we compare methodologies available, sensitivity and specificity of the various assays and volumes of sample needed. There are at least 7 different types of dexamethasone suppression testing and they are compared and described in detail. Confirmation of the anatomic source of the hormone is necessary. Petrosal sinus sampling and adrenal vein sampling are reviewed and the clinical indications for each discussed. Finally, once the endocrine diagnosis is reached based on endocrine testing, imaging studies are then reviewed which can confirm the endocrine diagnosis. An abnormality in the HPA axis is a laboratory diagnosis and radiologic imaging is reserved for the last step in the diagnosis of endocrine disease.

 

NONSTIMULATED HORMONE MEASUREMENTS

 

Overview

 

In evaluation of the hypothalamic pituitary adrenal (HPA) axis, static measurement of hormones is seldom useful due to the variable nature of cortisol and ACTH secretion in normal physiological states. In general, if one is suspicious of hypofunction of the HPA axis, then measurement of morning cortisol at 8 am when it is expected to be at its peaks is a good screening strategy. Depending on the result, this might need to be followed by dynamic testing to stimulate either adrenocorticotrophic hormone (ACTH) or cortisol for confirmatory purposes. On the other hand, if one is concerned about Cushing’s syndrome (CS), an overproduction of cortisol or ACTH, then measurement of cortisol should be performed late at night, when it is expected to be at its nadir. Alternatively, one could test cortisol’s response to suppression with dexamethasone.

 

The American Endocrine Society Clinical Guidelines recommend one of the following tests for the initial CS testing: at least two measurements of urinary-free cortisol (UFC), two measurements of late night salivary cortisol (LNSC), 1 mg overnight dexamethasone suppression test (DST) or a longer low-dose DST (1). Cortisol measurement (serum, UFC or salivary) is the end point for each recommended test.

 

Despite recent literature reports describing utility of direct salivary and urine cortisol measurements in CS diagnosis (2-4), most clinicians prefer provocative testing due to the variable nature of cortisol and ACTH secretion in normal physiological states. Cortisol is secreted under the direction of ACTH and follows a diurnal variation, with peak values at 08:00 h and a nadir at 22:00 h. In CS, diurnal variation is lost and PM cortisol level is inappropriately elevated. Superimposed on this diurnal pattern are 8-10 pulsatile peaks released during the course of a 24-hour period. Therefore, depending on the instance when blood is sampled, there can be significant variation in the absolute values of ACTH and serum cortisol. Due to this variability of cortisol and ACTH levels, it may be challenging to distinguish pituitary-dependent Cushing’s disease from pseudo-Cushing’s states. Cunningham et al conducted a study where blood was sampled and cortisol measured every 20 to 30 minutes for 24 hours. The group demonstrated that both circadian and pulse amplitudes of cortisol secretion were decreased in Cushing’s disease (5).

 

This section provides and overview of methodologies commonly used in clinical laboratories for direct determinations of cortisol and ACTH, regardless of whether they are a part of a provocative testing series or direct, non-stimulated hormone assessment.

 

Cortisol

 

Methods currently available for measuring serum cortisol levels include automated immunoassays and liquid chromatography-tandem mass spectrometry (LC-MS/MS).

 

CORTISOL IMMUNOASSAYS (TOTAL CORTISOL)

 

Cortisol immunoassays are widely available, have been in use for a long time, and automated methods provide high throughput with minimal manual sample manipulations. Virtually all immunoassay methods are based on the competitive binding principle, where cortisol from the patient sample and exogenous, labeled cortisol compete for the binding sites available on the anti-cortisol antibody. The major difference between the assays is in the label design and chemistry enabling antibody-antigen binding. All currently available cortisol methods have limit of detection below 1 µg/dL, providing sufficient sensitivity to support interpretation of CS dynamic testing results.

 

A widely recognized disadvantage of immunoassays is a potential of interferences from auto-, anti-animal or heterophilic antibodies. In addition, the older generations of cortisol assays had significant cross-reactivity with other steroids, such as 6-b-hydroxycortisol or prednisolone, due to the use of less specific polyclonal antibody in the assay formulation. However, the majority of current immunoassay methods have transitioned to a more specific monoclonal antibody format, minimizing or eliminating cross-reactivity with other steroids. It should also be noted that some immunoassay vendors use biotinylated antibodies in their assay design. In these instances, biotin may interfere with the assay, causing spuriously elevated cortisol measurement. The presence and magnitude of interference is vendor-specific and the potential of biotin interference should be checked with the laboratory that performs the testing. In general, none of the assays manufactured by Abbott use biotin in reagent formulation, while all assays manufactured by Roche do. The Roche cortisol assay should not be used to measure serum cortisol in a patient taking daily doses of biotin exceeding 5 mg, unless blood is obtained at least 8 hours following the last biotin ingestion.

 

LC-MS/MS CORTISOL ASSAYS

 

The LC-MS/MS assays utilize liquid chromatography to separate cortisol from other serum/plasma components and tandem mass spectrometry to detect and quantify compounds of interest. LC-MS/MS based methods offer superior analytical sensitivity and specificity over immunoassays.

 

Serum Free Cortisol

 

In conditions where CBG concentrations are affected, such as pregnancy or critical illness for example, total serum cortisol may not always reflect the true pituitary-adrenal status. In these cases, assessment of serum free cortisol is preferred. Free serum cortisol concentration are directly measured by separating free serum cortisol fraction using equilibrium dialysis (6) or ultrafiltration (7, 8) followed by cortisol determination, usually performed using LC-MS/MS method. Alternatively, free serum cortisol can be estimated by calculating the ratio of serum cortisol and CBG to obtain serum cortisol index (6).  Although not affected by CBG levels, free cortisol is also secreted in episodic fashion and thus not much more useful than random total serum cortisol levels in assessment of HPA axis functionality.

 

Urinary Free Cortisol

 

Cortisol is excreted in urine in an unbound (free) form and, like free serum cortisol is unaffected by fluctuations in CBG levels. Properly collected 24-hour urine specimens can be used to eliminate fluctuations that would affect serum cortisol levels, due to the pulsatile nature of its release. Therefore, measurement of UFC from 24 hour urine collections has become a valuable diagnostic tool for evaluation of adrenal cortical function and it is one of the first line tests recommended for CS diagnostic testing (1). In the unstressed patient, with normal renal function, elevation of UFC in 24-hour urine specimen is usually sufficient to diagnose CS. A normal result is strong evidence against that diagnosis. Although this test has long been used, its utility in CS diagnosis still remains somewhat controversial. Studies show wide variability in clinical utility of UFC for diagnosis of CS with clinical sensitivity ranging from 53% to over 90% and specificity ranging from 79% to 90% (2, 3, 9). These differences are due to differences in study design, cut-off, and methodology used. Furthermore, in a careful study of normal subjects de Boss Kuil et al found that urinary excretion of free cortisol can differ by as much as 50% between the two consecutive urine collections, while the creatinine values can differ by as much as five fold (10). Since the ratio of free cortisol/creatinine also varies considerably (range 1.0-3.7; median 1.3), intra-variation in urinary cortisol excretion could not be attributed to variation in creatinine excretion. In addition to biological variation, other factors include difficulty in over or under collection of urine. Given such wide discrepancies in reported clinical sensitivity and specificity of UFC measurements and significant intra-individual UFC variability, this test may not be an ideal choice for initial screening of CS.

 

Methodology used for UFC quantitation is the same as for serum cortisol. In terms of specimen collection, an 8:00 AM to the following day’s 8:00 AM collection is desirable. Samples should be refrigerated during collection and, while preservatives are not required, boric acid is usually acceptable. Quantitation of urine cortisol with a more sensitive and specific LC-MS/MS method is generally preferred over immunoassays. Typically, all the LC-MS/MS UFC assays involve a sample pretreatment with an organic solvent which removes the interfering substances. However, some UFC assays immunoassays either do not include this pretreatment step or offer it as an optional step to the user. As a result, UFC reference ranges vary widely between the assay manufacturers, methodologies, and different laboratories. To increase sensitivity, it is recommended that the upper limit of normal for any UFC assay be used as a positive test (5). It would be thus incorrect to make a diagnosis of adrenal insufficiency relying solely on 24-hour urine collections.

 

Salivary Cortisol

 

Late-night (23:00-24:00 h) salivary cortisol (LNSC) is one of the first line tests used to screen for CS. Most studies report high diagnostic sensitivity of this test (80-90%), but there are discrepancies in reported specificities (70-90%), resulting mostly from difference in methodologies and populations studied (2, 3, 11-13) Interestingly, mass spectrometry assays demonstrate high sensitivity, but low specificity (75%) for the diagnosis of CS (11). One potential explanation, as postulated by Raff, is that higher analytical specificity of mass spectrometry actually leads to lower diagnostic specificity, suggesting that cortisol metabolites and precursors picked up by immunoassays may be diagnostically relevant (14). Kannankeril et al recently reported that LNSC has excellent negative predictive value (99.8%) but poor positive predictive value (16.8%) for diagnosis of ACTH-dependent CS (12). Thus, a negative LNSC can be used to rule out ACTH-dependent CS, but complementary tests of adrenal function are needed to establish the diagnosis.

 

Salivary cortisol concentration is not dependent on CBG and could therefore be useful during an ACTH stimulation testing in patients with increased CBG concentrations due to increased estrogen or decreased plasma binding globulins due to critical illness.

 

Similar to UFC, the assay methodology remains the same as serum cortisol with the differences in specimen collection.

 

ACTH

 

ACTH measurements, while subject to the same circadian variability as cortisol (actually it is the variability of the ACTH that is directly responsible for the variability of the cortisol), are not subject to the effects of CBG. Values of ACTH > 100 pg/ml in the setting of possible adrenal insufficiency are usually suggestive of primary adrenal insufficiency, while values >500 pg/ml are diagnostic. Low concentrations of plasma ACTH are not diagnostic, except for the undetectable levels observed in patients with cortisol producing adrenal adenomas. Plasma ACTH concentration is also low in patients taking exogenous steroids.

 

Unlike widely available cortisol assays, the availability of clinical ACTH assays is limited. All currently available methods are immunoassays based on the “sandwich” principle, where two antibodies that recognize different ACTH epitopes are utilized. The first antibody, designated as capture antibody, detects one specific site on ACTH molecule and is used to pull the antigen from the patient’s plasma. The second antibody that detects a different ACTH epitope is then used to “sandwich” the antigen and generate a signal.

 

As is the case with any immunoassay, ACTH assays are susceptible to heterophilic antibody interferences. Several cases have been described in literature where aberrant, falsely elevated ACTH results were inconsistent with clinical picture and lead to unnecessary testing, misdiagnosis, and in some cases surgical interventions. These cases emphasize the importance of interaction between clinicians and the laboratory to identify any interference present and ensure that each patient is appropriately managed (15, 16). In addition, just as is the case with cortisol immunoassays, some vendors use biotinylated antibodies in the capture antibody design. Unlike cortisol, however, biotin interference may result in falsely decreased ACTH levels. The two most commonly used ACTH assays are manufactured by Siemens and Roche. Siemens ACTH assay is not affected by biotin, while the recommendation for Roche ACTH assay is not to use the test in patients ingesting >5 mg biotin daily, unless at least 8 hours had elapsed following the last biotin dose (cf. Roche Elecsys ACTH Package Insert V 12.0, 2020-11).

 

The preferred specimen for ACTH is EDTA plasma. ACTH is heat labile, and if not collected and preserved on ice, will lead to proteolysis, which can reduce the plasma concentration leading to falsely lower values.

 

Miscellaneous Non-Stimulated Measurements

 

CORTISOL BINDING GLOBULIN (CBG)

 

As mentioned earlier, the majority of cortisol (~92%) is bound to CBG, a serum protein. CBG levels increase in pregnancy and patients on oral contraceptives or supplemental estrogen. CBG is decreased in hyperinsulinemic states, nephrotic syndrome, starvation, severe illness, and chronic liver disease. This test is useful for the assessment of unexpected serum cortisol values. It is offered by large reference laboratories and uses a radioimmunoassay method.

 

11-DEOXYCORTISOL (COMPOUND S)

 

This is the immediate precursor of cortisol and is typically increased when ACTH is elevated or in 11 beta-hydroxylase deficiency. The method for 11-deoxycortisol measurement is now available by LC-MS/MS technology and is offered by most reference laboratories.

 

ANTI-ADRENAL ANTIBODIES

 

The measurement of anti-adrenal antibodies has been suggested to be useful in detecting early evidence of adrenal insufficiency, before cortisol values are decreased even in response to stimuli. The only test currently clinically available is a test that detects 21-hydroxylase autoantibodies, which are present in the common autoimmune form of Addison’s disease (17). This test is offered by major reference laboratories and is based on the radioimmunoassay format.

 

CORTICOTROPHIN RELEASING HORMONE (CRH)

 

Serum concentration of CRH is markedly elevated in pregnancy, presumably due to the production of CRH by the placenta. High levels are associated with high levels of CRH binding protein. Although mentioned as useful in the diagnosis of ectopic CRH syndromes, little data is available in this regard. CRH testing is not commonly done and we have not been able to find a commercial laboratory that is currently performing this test.

 

DYNAMIC TESTING

 

Glucocorticoid Deficiency

 

Adrenal insufficiency is a life-threatening disorder and prompt diagnosis is important because adequate hormonal replacement therapy can be lifesaving.

 

Despite that more than 35 years have elapsed since the initial description of the use of the insulin tolerance test (ITT) to diagnose adrenocortical deficiency (18), and more than 200 scientific publications in this area, clinicians today still argue as to which is the most sensitive and specific test to diagnose adrenocortical deficiency. The ITT is still regarded as the gold standard upon which to compare all other tests of HPA axis function. Unfortunately, this test has a considerable spectrum of intra-individual and inter-individual variation (19, 20). Therefore, when comparing other tests to the "gold standard", if the standard is not reliable, how can one determine the effectiveness of the other forms of testing? The problem lies in the ability of a single laboratory to know what the values are for their tests. Therefore, ranges from an ITT test response in normal subjects performed in one laboratory may not be normal for another laboratory. Taking this into account there are some general guidelines that are available for evaluating patients with suspected adrenal insufficiency.

 

PRIMARY ADRENAL INSUFFICIENCY

 

High Dose ACTH Stimulation Test

 

WHEN TO USE THIS TEST: Patients acutely ill in the hospital or clinic who present with signs and symptoms suggestive of primary adrenal insufficiency. Patients who are thermodynamically unstable should be resuscitated with crystalloids and given dexamethasone prior to testing if the diagnosis of primary adrenal insufficiency is being considered.

 

PROCEDURE: An intravenous (IV) line is placed 30 minutes before the test for rapid phlebotomy and to eliminate a temporary rise in cortisol associated with a needle stick. The IV line is to be kept open with 0.9% sodium chloride (NaCl) at a rate of 50 ml/hr. Blood is drawn at 0 min for ACTH (2 ml in a lavender top tube on ice) and cortisol (2 ml in a red top tube). Cosyntropin, 0.25 mg is administered as an IV bolus over 2 minutes. The cosyntropin comes as a lyophilized powder which should be reconstituted with 1 ml of 0.9% NaCl. Thirty min after the injection, blood is obtained from the IV line (2 ml) for cortisol. The same is repeated at 60 min (2 ml) for cortisol.

 

SPECIAL CONSIDERATIONS: The test can be performed at any time of the day. If the patient is receiving hydrocortisone or cortisone acetate, the medication should be held for at least 12 hours prior to testing (if possible). Although the test can be performed while the patient is receiving dexamethasone, there is some cross-reactivity in some assays and cortisol levels may not be accurate. Each laboratory should determine for itself, the effect of dexamethasone on their assay.

 

Patients with known sensitivity to cosyntropin or its preservatives should not have it administered. Oral estrogen use may result in elevation of the total serum cortisol level due to increased corticosteroid binding globulin (21). Patients with albumin <2.5 g/dL may also have a low cortisol level (21, 22).

 

CONTRAINDICATIONS: Hypersensitivity to cosyntropin or any component of the formulation.

 

WARNINGS / PRECAUTIONS: Use with caution in patients with pre-existing allergic disease or a history of allergic reactions to corticotropin. Class C in pregnancy.

 

ADVERSE REACTIONS 1% to 10%: Cardiovascular: Flushing. Central nervous system: Mild fever. Dermatologic: Pruritus. Gastrointestinal: Chronic pancreatitis. <1%: Hypersensitivity reactions

 

DRUG INTERACTIONS: Decreased effect: May decrease the effect of anticholinesterases in patients with myasthenia gravis; nondepolarizing neuromuscular blockers, phenytoin and barbiturates may decrease effect of cosyntropin

 

INTERPRETATION OF RESULTS: Baseline cortisol values <5 µg/dl and ACTH concentrations >100 pg/ml are usually diagnostic of primary adrenal insufficiency. The normal peak cortisol value post stimulation should be an increment no less than 7µg/dl. A peak stimulated cortisol value of >18 µg/dl at 30 min is considered normal. Since 37% of subjects had a peak response to cosyntropin at 30 min and 63% had a peak response at 60 min, both time points are analyzed in all patients and if either the 30 min or 60 min sample reaches the criteria as noted above, the test is considered normal (23).  However, there is some suggestion that new generation cortisol assays may have different cutoff values, but these have not been verified (24). 

 

Free cortisol, instead of total cortisol can be measured using a value of >1.2 µg/dl at 30 or 60 min as a normal result. This can be indicated in patient with albumin levels <2.5 g/dL or those with low cortisol binding globulin.

 

Serum aldosterone can be measured in 0 min, 30 min and 60 min blood samples as ACTH stimulation of the adrenal cortex will also stimulate aldosterone. It has been suggested that a normal aldosterone response to ACTH in the presence of a suboptimal cortisol response is diagnostic of secondary adrenal insufficiency (25).

 

Low dose ACTH stimulation Test

 

WHEN TO USE THIS TEST: Patients with subtle signs of adrenal insufficiency or patients who have been treated with glucocorticoids in whom determination of adrenal reserve is necessary. Patients who have autoimmune disease and may have early adrenocortical insufficiency may be best assessed with this test.

 

PROCEDURE: An intravenous line is placed 30 minutes before the test for rapid phlebotomy and to eliminate a temporary rise in cortisol associated with a needle stick. The IV line is to be kept open with 0.9% NaCl at a rate of 50 ml/hr. Blood is drawn at 0 min for ACTH (2 ml in a lavender top tube on ice) and cortisol (2 ml in a red top tube).

 

Cosyntropin, 1 µg is administered as an IV bolus over 2 minutes. The injection material was prepared according to the method of Dickstein as follows: The cosyntropin was diluted with 50 ml of sterile saline to a stock concentration of 5 µg/ml. Aliquots of 0.2 ml were added into sterile plastic tubes and kept at 4oC for a maximum of 4 months (26). Immediately prior to testing 0.8 ml of saline is added to the tube (final dilution 1 µg/ml) and 1 ml is injected into the patient. Thirty min after the injection blood is obtained from the IV line (2 ml) for cortisol. The same is repeated at 60 min (2 ml) for cortisol.

 

SPECIAL CONSIDERATIONS: Same as for high dose ACTH stimulation test, see above.

 

INTERPRETATION OF RESULTS: This test was originally developed to be more sensitive for diagnosing secondary adrenal insufficiency because it was more of a "physiologic" dose. It is much better at diagnosing secondary adrenal insufficiency than the high dose, although it is not at all recommended in acute or recent hypopituitarism when the intact adrenal glands can still respond normally to any dose of ACTH. Although probably not useful for the initial purpose of secondary adrenal insufficiency, it may be more sensitive at distinguishing milder forms of primary adrenal insufficiency (27). Furthermore, this low dose test was helpful in identifying mild adrenal suppression in asthmatic children being treated with inhaled steroids (28). As noted above, each laboratory should establish their normal values, however in general, a stimulated value at 30 or 60 min greater than 20 µg/dl would be considered normal.

 

A meta-analysis of 30 studies enrolling 1209 adults and 228 children with secondary adrenal insufficiency, evaluating the diagnostic accuracy of high and low dose ACTH stimulation concluded that they have similar diagnostic accuracy. They are both adequate to rule in, but not rule out, secondary adrenal insufficiency

 

SECONDARY ADRENAL INSUFFICIENCY (PITUITARY OR HYPOTHALAMIC)

 

Insulin Tolerance Testing (ITT)

 

WHEN TO USE THIS TEST: Patients in whom pituitary or hypothalamic disease may result in impaired corticotroph (or somatotroph) activity. Patients following pituitary surgery or pituitary radiation can be tested at any time, unlike the ACTH stimulation tests described above which are not useful in the acute setting. A random serum cortisol should be drawn prior to scheduling the test if the value is > 20 µg/dl, the test may not be necessary This test, can be performed in the outpatient clinic, however while relatively safe it requires a trained endocrine registered nurse to be present with the patient during the course of the test.

 

PROCEDURE: A 50 ml vial of 50% Dextrose should be at the patient's bedside in a syringe ready for injection before beginning the procedure.

 

An intravenous line is placed 30 minutes before the test for rapid phlebotomy, to eliminate a temporary rise in cortisol associated with a needle stick, and in order to have IV access for 50% Dextrose in the event of severe hypoglycemia. The IV line is to be kept open with 0.9% NaCl at a rate of 50 ml/hr. Blood is drawn at 0' for cortisol (2 ml in a red top tube) and glucose (1 ml in a gray top tube). Blood glucose is also checked at the bedside with a glucose monitor.

 

Regular (short acting) insulin is administered as an IV bolus at a dose of 0.1 units/kg. Blood is sampled for cortisol and glucose as noted above at 10min, 15min, 30min, 45min, 60min, 90min and 120min. A bedside nurse should monitor the blood glucose more frequently if glucose drops below 60 mg/dl on the glucometer or if the patient complains of neuroglycopenic symptoms, such as fatigue, diaphoresis, hunger, lightheadedness, or nausea. The test should continue until the blood glucose concentration drops below 40 mg/dl.

 

In patients with diabetes on insulin, consideration should be given that they may be insulin resistant. In which case, larger doses of insulin may be given. We usually begin with a single bolus of 0.1 U/kg and then re-bolus with insulin depending on the response to the initial dose (either give the same dose again if there was some response but insufficient, or double the dose if there was only minimal response to blood glucose, or give half the dose if the hypoglycemic response was close to the desired goal). This can be repeated several times until adequate hypoglycemia is reached.

 

Once the response goal of a glucose < 40 mg/dl is reached, patients can be fed a meal such as crackers and orange juice. Blood glucose should be checked at 5min, 10min and 15min post feeding. If there is no increase in glucose or a clinical response within 5min, intravenous glucose should be administered. If no response, then a repeat bolus of glucose is suggested. If no response or IV access is lost, glucagon 1 mg intramuscular can be given.

 

SPECIAL CONSIDERATIONS: The test can be performed at any time of the day, although due to the need for patients to be fasting it is most conveniently done in the morning. If the patient is receiving hydrocortisone or cortisone acetate, the medication should be held for at least 12 hours prior to testing (if possible). Unlike the ACTH stimulation tests, the ITT cannot be performed while the patient is receiving dexamethasone, due to suppression of the hypothalamic pathways necessary to respond to hypoglycemia.

 

In general ITT is not recommended in patients with uncontrolled seizure disorder or significant coronary artery disease.

 

In order to determine if the level of dysfunction is at the hypothalamus or at the pituitary this test is sometimes used in addition to the CRH stimulation test. When the ITT fails to stimulate cortisol, but the CRH test does stimulate it is likely that the patient is having hypothalamic dysfunction.

 

INTERPRETATION OF RESULTS: Serum cortisol should increase within 30 min of the hypoglycemic response to > 20 µg/dl. If the serum cortisol at baseline is 18 ug/dl the test may not be diagnostic. If the baseline serum cortisol is higher than 19 µg, adrenal insufficiency is unlikely. Although the response of cortisol is more reproducible than that of growth hormone in the ITT, intra-subject differences have been reported (20, 30).

 

Metyrapone Testing

 

WHEN TO USE THIS TEST: This test is perhaps the most sensitive to determine whether the HPA axis is intact. Although metyrapone is not generally available from your neighborhood pharmacy, it can be obtained by calling Novartis Pharmaceutical Corp. at 1-800-988-7768 on weekdays. Metyrapone blocks 11-b hydroxylase and results in the inhibition of conversion of 11-deoxycortisol to cortisol. Serum levels of cortisol decrease and concentration of 11-deoxycortisol increases, however 11-deoxycortisol does not down regulate ACTH. Therefore, in a normally functioning HPA axis there is an increase in 11-deoxycortisol. This metabolite can be directly measured in the serum or measured in the urine as 17-OH corticosteroids. This test can help differentiate primary adrenal deficiency from ACTH deficiency. It has a similar diagnostic performance to the ITT and it’s a potential alternative when there is a contraindication to ITT.

 

PROCEDURE: For assessment of adrenal or pituitary insufficiency the test can be performed as an overnight test. Metyrapone is given orally (30 mg/kg body weight, or 2 grams for <70 kg, 2.5 grams for 70 to 90 kg, and 3 grams for >90 kg body weight) at midnight with a glass of milk or a small snack (24). Serum 11-deoxycortisol and cortisol are measured at 8 AM the next morning; it is also recommended to measure plasma ACTH levels (31).

 

SPECIAL CONSIDERATIONS: The concurrent use of glucocorticoids will interfere with the test. Any medications that the patient is taking which increase the P450 enzymes will increase the metabolism and clearance of metyrapone (such as rifampin, phenobarbital, and phenytoin) (32). Similarly, hypothyroidism or hyperthyroidism will affect clearance of metyrapone and the adrenal responsiveness. Therefore, thyroid function tests should be measured prior to performing this test. Measurement of 11-deoxycortisol, like cortisol itself is dependent on CBG and drugs such as estrogens and oral contraceptives will falsely increase the concentrations of 11-deoxycortisol (33).

 

PREGNANCY IMPLICATIONS - Use during pregnancy only if clearly needed. Subnormal response may occur in pregnant women and the fetal pituitary may be affected.

 

LACTATION - Excretion in breast milk unknown/use caution

 

ADVERSE REACTIONS - Frequency not defined. Central nervous system: Headache, dizziness, sedation. Dermatologic: Allergic rash. Gastrointestinal: Nausea, vomiting, abdominal discomfort or pain. Hematologic: Rarely, decreased white blood cell count or bone marrow suppression.

 

INTERPRETATION OF RESULTS: 8 AM serum 11-deoxycortisol concentrations should be >7 µg/dL with serum cortisol less than 5 µg/dL (138 nmol/L), confirming adequate metyrapone blockade. The plasma ACTH concentration at 8 AM should exceed 75 pg/mL (17 pmol/L), confirming that any increases in serum 11-deoxycortisol concentrations are ACTH-dependent, thereby separating primary from secondary adrenal insufficiency (34, 35).

 

Glucocorticoid Excess

 

DEXAMETHASONE SUPPRESSION TEST

 

Measurement of endogenous cortisol production in response to exogenous dexamethasone suppression was the first provocative test and still remains among the most useful tests used for the evaluation of excess cortisol. Dexamethasone, due to its high affinity to the glucocorticoid receptor is a potent inhibitor of ACTH synthesis and release. In addition, most of modern immunoassays for cortisol (both urine and serum) utilize an antibody that does not cross-react with dexamethasone. Therefore, the combination of being able to use relatively low doses and at the same time not interfere with the measurement of cortisol make dexamethasone suppression useful for establishing the presence of a perturbation in the pituitary - adrenal axis and for diagnosing the etiology of hypercortisolism.

 

At least five different tests have been described using dexamethasone, which differ in the dose and timing of dexamethasone treatment and differ in whether there is measurement of urine or serum cortisol or 17-OH-corticostseroids (Table 1). Although the endocrine basis for the tests are in general the same, none are perfect. Confirming the diagnosis of patients with suspected hypercortisolism requires several tests for accurate diagnosis.

 

TABLE 1. Various Dexamethasone Suppression Tests

Dex Supp Test

Dex Dose

Time of Admin

Normal Response

Sens/Spec

Low dose Oral/Night

1 mg

@23:00 x1

<1.8 mcg/dl or <5 mcg/dl

87% / 100%

High dose Oral/Night

8 mg

@23:00 x 1

<50% basal

92% / 100%

Low Dose 2day

0.5 mg

q 6h x 2 days

<10 µg/24h in urine

74-98%/69-100%

High Dose 2 day

2.0 mg

q 6h x 2 days

<50% basal

79% / 100%

Very High dose

8 mg

q 6h x 1 day

<50% basal

74% / 100%

Note: To assure patient compliance and determine whether there is abnormal metabolism of the dexamethasone, serum levels of dexamethasone can be measured. However, this is not a common diagnostic test. Testing can be done by specialized laboratories, such as Esoterix inc. CA. The principle of the assay is RIA after chromatographic sample separation and requires 1 ml of serum sample.

 

All these tests require significant patient participation as the patients are required to self-administer the dexamethasone at inconvenient hours of the day (11PM) or up to 4 times a day. Sampling requires either collection of urine for 24 hours or coming to the physician's office at 8 AM for multiple blood sampling. Drugs that induce hepatic cytochrome P-450 enzymes, such as barbiturates, phenytoin, rifampin, and aminoglutethimide, increase the metabolism of dexamethasone and other steroids. Measurement of serum dexamethasone a few hours after the last dose will help determine if there is abnormal metabolism. All these caveats are in addition to the other problems associated with measurement of cortisol as noted above, including the variable diurnal variation as well as interference with concurrent administration of glucocorticoids, estrogen, or other medications that increase cortisol binding globulin.

 

A popular screening test for confirming hypercortisolism is the overnight 1 mg dexamethasone. A single dose of 1 mg is administered (or 0.3 mg/Kg for children (34) at 11PM and blood is obtained by 8 AM the following morning. The dexamethasone dose is given prior to the diurnal rise in endogenous ACTH release and therefore suppresses the early AM cortisol. A normal response would be a serum cortisol concentration of <1.8 mcg/dl, alternatively a cut point of < 5 µg/dl can be used which will yield more specificity with less sensitivity. If cortisol is >10 µg/dl the likelihood of hypercortisolism is high. The other dexamethasone suppression tests are reviewed in Table VIII. Patients with corticotroph macroadenomas or very active tumors, may have urine free cortisol in excess of 1000 µg/dl which will require higher doses of dexamethasone to confirm suppressibility and/or rule out ectopic ACTH production (36).

 

The two- day low dose dexamethasone suppression test can be used to differentiate Cushing’s syndrome from pseudo-Cushing’s which can present with many of the signs and symptoms associated with hypercortisolism in the setting of other clinical conditions such as depression, alcoholism, PCOS, obesity, and uncontrolled diabetes (37, 38). Dexamethasone 0.5 mg is delivered orally Q6 hours for 48 hours. Serum cortisol is measured 2 hours after the last dose and a cutoff level of <1.4 µg/dl is consistent with pseudo-Cushing’s. Measurement of 24 hour urine excretion of 17-hydroxycorticosteroid and creatinine during the administration of dexamethasone starting at 1200h, has also been suggested with a cut point of 11 umol/day or higher considered positive for Cushing’s syndrome (39). This test however, can misclassify as many as 15% of patients with Cushing’s syndrome and up to 15% of patients with pseudo Cushing’s.

 

The overnight high dose dexamethasone suppression test can help differentiate Cushing’s disease from ectopic ACTH syndrome in patients with ACTH-dependent Cushing’s syndrome. The basis for this differentiation is the fact that ACTH secretion in Cushing’s disease is only relatively resistant to glucocorticoid negative feedback inhibition. Cortisol levels will not suppress normally with overnight 1 mg but will suppress with a higher dose of 8 mg of dexamethasone. Serum cortisol concentration at 8 AM is <5 µg/dL in most patients with Cushing’s disease and is usually undetectable in normal individuals. A more than 50% decrease in cortisol on the day after taking 8 mg dexamethasone supports a diagnosis of Cushing’s disease over ectopic ACTH production. In patients with non-ACTH dependent hypercortisolism, a lack of suppression of cortisol by more than 50% with a low normal ACTH level (5-20 pg/ml) suggests an adrenal etiology.

 

CRH STIMULATION TEST

 

WHEN TO USE THIS TEST: This test is one of the most sensitive to determine if there is an abnormality in the HPA axis and for diagnosing the etiology of hypercortisolism in ACTH dependent Cushing’s.  Although CRH is expensive ($300), when one considers the cost of multiple urine collections and analyses of cortisol as well as the cost of a single MRI of the pituitary (which generally exceeds $1500), CRH is at least cost effective when one considers the overall expense in the evaluation of these patients.

 

PROCEDURE: An intravenous line is placed 30 min before the test for rapid phlebotomy and to eliminate a temporary rise in cortisol associated with a needle stick. Blood is drawn at -15' and 0' for cortisol and ACTH (2 ml in a lavender top tube on ice). CRH is then injected IV at a dose of 1 µg/Kg up to a maximum of 200 µg. Blood is obtained at 15, 30, 60, 90, 120, 180 and 210 min for cortisol and ACTH (2 ml in a lavender top tube on ice).

 

SPECIAL CONSIDERATIONS: The test can be performed at any time of the day, although the initial studies describing the test have been done in the morning.

 

Side effects: The patient may experience slight nausea, metallic taste, urgency to urinate, a change in blood pressure (either increase or decrease), a change in heart rate, headaches, abdominal discomfort, facial flushing, and lightheadedness. These side effects are mild and last for only few minutes. Category C in pregnancy.

 

INTERPRETATION OF RESULTS: The mean ACTH concentrations at 15 and 30 min after CRH should increase by at least 35% above the mean basal value at -15 and 0 min in patients with Cushing's disease, but not in patients with ectopic ACTH secretion. This measure gave the best sensitivity (93%) and specificity (100%) (40, 41).  The best cortisol criterion was a mean increase at 30 and 45 min of 20% or more above mean basal values, which gave a sensitivity of 91% and a specificity of 88%. It should be noted that the criterion for Cushing's disease is based on the presence of hypercortisolism. The CRH test will not adequately differentiate subjects with pseudo-Cushing’s and those with true pituitary dependent Cushing's disease.

 

CRH TEST WITH DEXAMETHSONE

 

WHEN TO USE THIS TEST: Several investigators have found that modifications of the CRH stimulation test can increase further the sensitivity and specificity in the diagnosis of the etiology of Cushing's disease. While the simultaneous use of vasopressin can augment the response to CRH, dexamethasone can be used to suppress all but pathologic responses to CRH stimulation [33]. Without dexamethasone the sensitivity and specificity of the CRH test is 65 and 100%, respectively, while with dexamethasone the CRH test is 100% sensitive and specific. This test is also particularly useful to differentiate true Cushing’s from pseudo-Cushing’s state.

 

PROCEDURE: Dexamethasone, 0.5 mg is self-administered orally by the patient every 6 hours for 2 days, at 6 AM, 12 Noon, 6 PM and midnight. On the morning of the 3rd day an additional dose of dexamethasone is given at 6 AM. The patient arrives at the testing center by 8 AM and an intravenous line is placed 30 minutes before the test for rapid phlebotomy and to eliminate a temporary rise in cortisol associated with a needle stick. Blood is drawn at -15' and 0' for cortisol and ACTH (2 ml in a lavender top tube on ice). CRH is then injected IV at a dose of 1 µg/Kg up to a maximum of 200 µg. Blood is obtained at 15, 30 60, 90 120, 180 and 210 min for cortisol and ACTH (2 ml in a lavender top tube on ice).

 

SPECIAL CONSIDERATIONS: The test can be performed at any time of the day, although it is usually done in the morning.

 

Side effects that the patient may experience are: slight nausea, metallic taste, urgency to urinate, a change in blood pressure (either increase or decrease), a change in heart rate, headaches, abdominal discomfort, facial flushing, and lightheadedness. These side effects are mild and last for only few minutes.

 

Similar to the dexamethasone suppression test, the results should be interpreted with caution in patients taking estrogen therapy as they can present with falsely elevated cortisol levels due to an increase of cortisol-binding globulin. Drugs such as phenytoin, phenobarbitone, carbamazepine, rifampicin and alcohol induce hepatic enzymatic clearance of dexamethasone, mediated through CYP 3A4, thereby reducing the plasma concentration and may be associated with false positive results (42).

 

INTERPRETATION OF RESULTS: A normal response would be a plasma cortisol concentration less than 1.3 µg/dl measured 15 minutes after the administration of CRH.  Values of cortisol greater than 1.3 µg/dl correctly identified all cases of Cushing's syndrome and all cases of pseudo-Cushing's states (100% specificity, sensitivity, and diagnostic accuracy). While this is a general recommendation, each laboratory should confirm based on the sensitivity of the respective cortisol assay. Furthermore, it is important to confirm the serum level of dexamethasone at the time of the blood draw to assure patient compliance with the dexamethasone regimen.  Patients with ectopic ACTH production will have nonsuppressed cortisol and ACTH levels that are not stimulated by CRH.

 

DDAVP STIMULATION TEST

 

WHEN TO USE THIS TEST: This test can be used as part of the workup of ACTH dependent hypercortisolism. It can be used in addition to the CRH stimulation test as studies have shown that the combination of these two tests performs better than either of the tests separately. It can also be performed in lieu to the CRH test in situations in which CRH is not available. The aberrant expression of vasopressin V2 receptor in pituitary ACTH-secreting adenomas is the rationale for the use of the desmopressin test to differentiate corticotroph adenomas (which should respond to desmopressin injection) from ectopic ACTH secreting tumors or pseudo Cushing’s (which should not respond)(43-45).

 

PROCEDURE: An intravenous line is placed 30 minutes before the test for rapid phlebotomy and to eliminate a temporary rise in cortisol associated with a needle stick. Blood is drawn at -15' and 0' for cortisol and ACTH (2 ml in a lavender top tube on ice). DDAVP is then injected IV at a dose of 5 to 10 ug. Blood is obtained at 15, 30, 60, 90, 120, 180 and 210 minutes for cortisol and ACTH (2 ml in a lavender top tube on ice) (45).

 

INTERPRETATION OF RESULTS: No definitive cutoff values have been standardized for the interpretation of this test. The published established criteria for this test have generally been based on studies with small series of subjects. Malerbi et al. proposed a cortisol increase over baseline of 12% to be consistent with diagnosis of Cushing’s disease (46). An absolute ACTH increase over baseline equal or greater than 6 pmol/L yielded higher sensitivity and specificity to differentiate Cushing’s disease from pseudo Cushing’s in a different study. Alternatively the criteria used for the CRH stimulation test can be used in  the interpretation of the results (47).

 

INFERIOR PETROSAL SINUS SAMPLING (IPSS) WITH CRH STIMULATION

 

WHEN TO USE THIS TEST: Once the diagnosis of ACTH dependent Cushing's syndrome has been made based on endocrinologic testing, the next step in the evaluation of such patients should be an MRI of the pituitary to confirm the presence of a pituitary mass. Unfortunately, MRI imaging of the pituitary as a primary diagnostic tool is distinctly unhelpful due to the fact that 10% of all normal individuals may have slight abnormalities of their pituitary and that in many subjects with Cushing's disease, the tumor may be too small to be imaged with MRI scans. However, subjecting a patient to surgical pituitary exploration in the absence of a demonstrable mass is likely to result in an unsuccessful surgery. Furthermore, if previous dexamethasone and/or CRH testing is equivocal, then IPSS should be performed to further confirm the pituitary as the source of the ACTH (34). Although this test is less reliable in lateralizing the ACTH source (i.e., left versus right), than it is in confirming that the ACTH is central in origin, it can rule out ectopic ACTH production by a tumor (although ectopic CRH secreting tumors would be difficult to distinguish from true Cushings' disease based on IPSS). Simultaneous measurement of prolactin in the central samples can normalize the data if there is any difference in the location of the catheters (48).

 

It is recommended that active hypercortisolism is confirmed by measuring a 24-hour UFC or overnight UFC the day preceding IPSS. Misleading results have been reported when this test is performed “out of cycle” in patients with cyclical Cushing’s.

 

PROCEDURE: This test is done in conjunction with a skilled interventional neuroradiologist. It is important that the endocrinologist is personally present in the room during the procedure so that assurance can be made that the proper blood tests were drawn at the specified times. The patient is brought to the angiogram suite without sedation. A large bore IV line is placed in an antecubital fossa (to be certain there is access to peripheral blood sampling and CRH injection). Catheters (5 French) are placed in the femoral veins and threaded under fluoroscopic guidance to the inferior petrosal sinus. Injection of IV contrast confirms proper placement of the catheters.

 

Patients are on constant, pulse, blood pressure and oxygenation monitors during the course of the procedure. Test tubes are prechilled in ice and labeled so that during the rapid sampling period, blood can be placed in the tubes without delay.

 

It is recommended to routinely obtain 4 baseline measurements at -15, -10, -5 and at 0 minutes. This allows for practice allowing proper coordination between the radiologists drawing blood from the IPSS and the individual drawing blood from the brachial vein. Appropriate amounts of blood should be removed to discard the dead space of the catheter (this varies depending on the size of the catheter used). 2 ml of blood is obtained in lavender top vacutainer tubes on ice for measurement of cortisol (on peripheral samples); ACTH and prolactin (on central samples).

 

At 0' CRH is then injected as described above for the peripheral CRH test. Alternatively, a combination of CRH and 10ug of desmopressin can be used, especially if the patient has had a negative response to a prior CRH test. If CRH is not available, IPSS can also be performed with desmopressin alone per the protocol described above. Blood is then sampled from both central and peripheral lines at 2', 5' 10' and 15'. After the 15' time point and right before the IPSS catheters are removed, repeat fluoroscopic localization of the catheters should be performed to confirm that there was no displacement during the sampling. However, sampling on peripheral blood may continue as described in the CRH test discussed above.

 

SPECIAL CONSIDERATIONS: The test can be performed at any time of the day, although it is usually done in the morning.

 

Side effects that the patient may experience are: slight nausea, metallic taste, urgency to urinate, a change in blood pressure (either increase or decrease), a change in heart rate, headaches, abdominal discomfort, facial flushing, and lightheadedness. These side effects are mild and last for only few minutes.

 

Patients greater than 300 pounds in weight may not be able to be supported by the standard fluoroscopic table. Furthermore, such large patients may have an abdominal pannus that precludes reasonable access to the femoral veins. In such instances the IPSS can be performed via catheters placed in the antecubital vein with the patient immobilized in the sitting position.

 

Strokes have been reported in the literature as a potential complication (36). To minimize this possibility, it is recommended that the catheters remain in the petrosal sinus for no more than 30 min.

 

Freeze/thawing can decrease the ACTH concentration (see above); therefore, we recommend that the samples be brought to the endocrine lab and analyzed within 24 hours with the plasma separated and kept on ice during this time. If the analysis is not possible within 24 hours, the samples should be aliquoted and frozen to minimize the amount of freeze/thawing.

 

INTERPRETATION OF RESULTS: Plasma ACTH values are normalized to the prolactin value in order to correct for possible different localization of the catheters, or movement of the catheters during the study. The post CRH ACTH/Prolactin value of the central catheters should be >2.1 fold the ACTH/Prolactin value of the peripheral sample. In most cases of pituitary dependent Cushing’s, the increase is > 5.0-fold. A central to peripheral ACTH gradient higher or equal to 2 before CRH administration or higher or equal to 3, 10 min after CRH infusion is considered diagnostic of a pituitary source of ACTH (Cushing's disease). Lateralization would mean that the ratio of the left to right side is >2.0. Frequently the ratio criteria can be met without the need for CRH stimulation, however, the diagnostic accuracy increases from 86% to 90% with CRH (37).

 

The workup of ACTH dependent Cushing’s to differentiate Cushing’s disease from ectopic ACTH source can be quite challenging and often times requires combination of different dynamic testing in addition to imaging that often ultimately led to costly and invasive diagnostic procedures such as IPSS to be able to establish an accurate diagnosis. A retrospective study involving 167 patients with Cushing’s disease and 27 patients with ectopic Cushing’s found that using thresholds of a cortisol increase >17% with an ACTH increase >37% during CRH test and a cortisol increase >18% with an ACTH increase >33% during desmopressin test, the combination of both tests gave 73% sensitivity and 98% PPV of Cushing’s disease. The PPV was 100% in patients with positive response to both tests, with a negative pituitary MRI and whole-body CT scan. The NPV was 100% in patients with negative response to both tests, with negative pituitary MRI and positive whole body CT scan. This combination of dynamic tests with imaging studies is proposed as an accurate, cost-effective diagnostic strategy for the workup of ACTH depended Cushing’s that might minimize the need for IPSS which can be invasive, costly and unavailable in all institutions (43)

 

ADRENAL VEIN SAMPLING

 

WHEN TO USE THIS TEST: Patients diagnosed with ACTH independent Cushing’s and found to have bilateral adrenal tumors on imaging pose a particular challenge to the clinician. The differential diagnosis in these cases includes unilateral cortisol secreting adenoma (or carcinoma) with contralateral non-functioning cortical adenoma, bilateral cortisol secreting adenomas, macronodular adrenal hyperplasia, and primary pigmented nodular adrenocortical disease. Adrenal vein sampling measuring cortisol can be very helpful in this scenario and give valuable information to elucidate the proper diagnosis and guide therapy.

 

PROCEDURE: This test is done in conjunction with a skilled interventional radiologist under sedation. The procedure is usually performed early morning after an overnight fast on the second day of either a low dose (0.5 mg orally every 6 hours) or high dose (2 mg orally every 6 hours) of dexamethasone administration. This eliminates the probability of endogenous ACTH secretion causing interference with the interpretation of autonomous adrenal gland cortisol secretion. The adrenal veins can be catheterized by the percutaneous femoral vein approach, the position of the catheter tip should be verified by venogram. Concentrations of cortisol and aldosterone should be measured in blood obtained from both adrenal veins and the external iliac vein (for the detection of peripheral venous concentrations) (49).

 

SPECIAL CONSIDERATIONS: Potential complications include thrombosis with subsequent infarction or hemorrhage adrenal insufficiency and hypertensive crisis, however these are rare (48).

 

The aldosterone concentrations are usually much higher on the right adrenal vein compared to the left, this is presumably due to the anatomy differences and the catheter proximity to the right adrenal medulla. For this reason, although plasma epinephrine is measured to confirm success of adrenal vein catheterization, it cannot be used to correct for blood sample dilution between the 2 adrenal veins. There have been few case reports in which aldosterone has been used for side-to-side dilution differences, however whether it can be used for this purpose remains unclear (49, 50).

 

INTERPRETATION: Catheterization of each adrenal vein can be considered successful if plasma aldosterone concentration in the adrenal vein exceeds peripheral venous concentration by more than 100 pg/ml. An adrenal-to-peripheral venous cortisol gradient greater than 6.5 can be considered consistent with a cortisol secreting adenoma. Lateralization can be determined by measuring the side-to-side cortisol gradient (high-side to low-side). A ration of 2.3 or greater is consistent with autonomous cortical secretion from predominately 1 adrenal gland (49).

 

IMAGING STUDIES IN THE HPA AXIS EVALUATION

 

The evaluation of the HPA axis function should always be approached through biochemical measurements. With few exceptions, imaging studies provide no information about hormonal function but can be very useful for the localization of tumors or lesions. Once a biochemical diagnosis of either deficiency or excess of glucocorticoid production has been established, imaging studies can complement and assists the hormonal evaluation, providing valuable information about etiology, prognosis, and management.

 

Pituitary Imaging

 

In the vast majority of cases of ACTH dependent Cushing’s syndrome (CS), the source of ACTH is in the pituitary (Cushing’s Disease), so performing imaging studies of the pituitary gland in this scenario to try to localize a tumor is appropriate. However given the high incidence of non-functioning pituitary adenomas in the general population (up to 10%) (51) and the increasing sensitivity of the high resolution imaging modalities available that can lead to false positive results, it is important to perform a thorough dynamic testing evaluation of each case and consider inferior petrosal sinus sampling if appropriate (see section above), before committing a patient to pituitary surgery. Adrenocorticotropic pituitary tumors represent about 10% of all pituitary tumors (52) and  ACTH-secreting adenomas are most commonly microadenomas (<1cm). In cases of macroadenoma, assessment of extrasellar extension including chiasmatic compression and cavernous sinus involvement is imperative (53).

 

The other scenario in which pituitary imaging is indicated and can be useful in the evaluation of the HPA axis function, is in patients diagnosed with secondary adrenal insufficiency who have no history of recent exogenous glucocorticoid exposure or any other clear explanation for the clinical presentation. In these cases, a mass lesion disrupting the HPA function should be suspected, especially if the patient presents with deficiencies of other pituitary hormones and/or elevated prolactin, as isolated adrenal insufficiency from a non-functioning tumor affecting the pituitary is very rare.

 

PITUITARY MRI

 

Magnetic resonance imaging (MRI) is the mainstay of pituitary assessment. MRI is more sensitive than computed tomography (CT) in detecting corticotroph adenomas, but still detects only about 50% of these tumors (54) and has a false positive rate of 12-19% (55, 56)]. Standard pituitary imaging protocols typically include thin-section (2 or 3 mm) of T1-weighted (w) spin echo sequences (SE) performed both in coronal and sagittal planes through the pituitary fossa, which are repeated after administration of intravenous gadolinium contrast medium, associated with a T2-weighted sequence in the coronal plane (57, 58). High spatial detail can be achieved by using thin slices, a fine matrix size and a small field of view focused on the pituitary (58). The classic MR features of a corticotroph adenoma include a less than 1 cm focal area of lesser enhancement on T1-w images following contrast administration, hyperintense or hypointense on T2-w images as compared with the normal pituitary gland, remodeling of the pituitary sella floor and deformity of the gland contour (59). Acquiring dynamic sequences in the first 1-2 minutes after contrast injection can increase the sensitivity (60), but this technique has not been unequivocally demonstrated to improve the usefulness of MR in Cushing’s (61). The use of three-dimensional (3D) spoiled gradient recalled acquisition in the steady state (SPGR) sequence allows for superior soft tissue contrast compared to conventional spin echo sequences, this technique can be further optimized with thin-slice imaging (<1mm) (58).  Compared to T1-w SE sequence, SPGR has been reported to increase sensitivity but also has a higher false positive rate (62, 63).

 

PITUITARY CT SCAN

 

Pituitary computed tomography (CT) scanning is less sensitive than MRI for the detection of pituitary adenomas (64)and it is usually reserved for those patients who cannot safely undergo brain MRI. Acquisition of 1 mm (or less) axial sections through the pituitary fossa with coronal reconstructions can be helpful in the assessment of macroadenomas (57). It is also very helpful preoperatively in patients planned for transsphenoidal pituitary surgery to delineate the bony anatomy (65)

 

Adrenal Gland Imaging

 

There are a couple of scenarios in which adrenal gland imaging plays a role in the evaluation of the HPA axis. It is indicated and particularly important in the evaluation of patients diagnosed with ACTH independent CS, which is most commonly caused by adrenocortical adenomas or carcinomas and less frequently bilateral micronodular and macronodular hyperplasia. It can also be considered in cases of primary adrenal insufficiency. Tumors in the adrenal are fairly common in humans, they have been found to be present in 3% of autopsies performed in persons older than 50 years of age (66) and have been reported to be incidentally discovered in up to 5% of cross-sectional abdominal imaging carried out for unrelated problems (67). Most of these incidentally found adrenal tumors are nonfunctioning, 10 to 15% secrete excess amounts of hormones (68) of these, adrenocortical tumors are the most common. On the basis of imaging characteristics alone, no distinction can be made between a benign hyperfunctioning and a non-functioning adenoma, and this can only be differentiated based on clinical and biochemical diagnosis. Adrenal carcinoma represents <10% of adrenal tumors, 30 to 40% of these are hyperfunctioning in adults (69).  There are multiple important imaging characteristics that can help differentiate benign adrenal adenomas from pheochromocytomas, adrenocortical carcinomas and metastasis, like percentage of lipid content, tumor size, homogeneity, border regularity, presence of calcifications, invasion of surrounding tissue, and lymph node enlargements (Table 2).

 

ADRENAL CT SCAN

 

Unenhanced thin- section CT scan followed by contrast-enhanced examination is the cornerstone of imaging of adrenal tumors. Unenhanced CT is important to provide density measurements of lesions (70). The rich intracytoplasmic fat in adenomas results in a low attenuation on nonenhanced CT. The Hounsfield (HU) scale is a semiquantitative method to measure radiograph attenuation. If an adrenal mass measures <10 HU on unenhanced CT, the likelihood that it is a benign adenoma is nearly 100% (71).  However up to 30% of benign adenomas might not contain large amounts of lipid and present with higher HU on nonenhanced CT scan. This is when measuring the contrast washout on delayed images is very useful. Ten minutes after the administration of contrast, an absolute medium washout of more than 50% has been reported to be close to a 100% sensitive and specific for benign adenoma (72). Non-adenomas include metastases, pheochromocytomas and carcinomas.

 

Adrenal carcinomas usually appear as a unilateral mass, >4 cm in size with an inhomogeneous appearance due to necrosis, hemorrhage, fibrosis, and calcification. Careful assessment of the draining venous structures is essential on imaging, together with identification of direct infiltration of adjacent viscera (57).

 

ADRENAL MRI

 

When lesions cannot be characterized adequately with CT, MRI evaluation (with T1 and T2-weighted sequences, chemical shift and fat-suppression refinements) can be sought. Adrenal adenomas usually show low homogeneous signal on T1-weighted images and a signal intensity equivalent or higher than the liver on T2-weighted images. Chemical shift imaging will readily identify the lipid rich adenomas with signal loss on the out-of-phase sequences (73). This loss of signal can be measured using the adreno-splenic-ratio (ASR) and the signal intensity index (SII). An ASR ratio of <70% has been shown to be highly specific for adenomas and has a 78% sensitivity. Using the SII, a minimum of 5% signal loss characterizes an adrenal adenoma with accuracy of 100% [61]. MRI can also be particularly useful to evaluate for local and distant invasion of adrenocortical carcinomas.

 

Primary pigmented nodular adrenocortical disease is a rare cause of Cushing’s syndrome that has a female predilection and may be familial or associated with Carney complex. On imaging the adrenal glands may appear normal or minimally hyperplastic with multiple, usually <5 mm, unilateral or bilateral benign cortical nodules. The adrenal nodules are macroscopically pigmented; they demonstrate a lower T1 and T2 signal intensity on MRI compared to surrounding atrophic cortical tissue. When nodules are 1-2 cm in size, there might be atrophy of the intervening cortex, which helps distinguish this condition from ACTH- dependent hyperplasia (57).

 

Another rare cause of Cushing’s syndrome is ACTH-independent macronodular adrenal hyperplasia, which has a male predilection. The imaging appearance of the adrenal glands is striking with massive bilateral adrenal enlargement, nodularity. and distortion of adrenal contour. Nodules can measure 1 to 5.5 cm. On MRI they are hypointense relative to liver on T1-w images and hyperintense or isointense in T2-w images. On chemical shift imaging, nodules lose signal intensity on out-of-phase due to their high lipid content (57).

 

OTHER ADRENAL IMAGING MODALITIES

 

Patients that harbor adrenal masses, which are not adequately characterized by CT or MRI, can be further evaluated with functional nuclear medicine modalities that include single photon emission computed tomography (SPECT) scintigraphy with various radionuclide tracers, and positron emission tomography (PET) scintigraphy with various radionuclides.  PET images provide a higher spatial resolution compared to SPECT (70).

 

PET scan with either Fluorodeoxyglucose (FDG) or 11C-metomidate (MTO) can be useful in selected cases to differentiate benign adrenal adenomas from adrenocortical carcinomas. An elevated uptake on the FDG scan correlates with high metabolic activity and raises the suspicion for malignancy (74) with high sensitivity and specificity (75). Limitations of this technique include physiological excretion of FDG into renal inflammatory system and high metabolic uptake in inflammatory and infectious processes as well as in benign pheochromocytomas, leading to false positive results (64). Metomidate is an inhibitor of 11 beta-hydroxylase (CYP11B1) and aldosterone synthetase (CYP11B2), and based on this property its use can help differentiate tumors of adrenocortical origin from non-cortical lesions. Originally developed as a PET imaging agent radiolabeled with 11C, more recently it has been labeled with 18F and 123I, allowing SPECT and SPECT/CT imaging (76).

 

Integrated or “fused” PET-CT imaging allows to combine CT attenuation measurements with the intensity of FDG uptake, as described by the standardized uptake value (SUV), improving the performance of either imaging technique alone (77).

 

Scintigraphy with Iodine-131-Iodomethyl-19-norcholesterol (NP 59) is a functional nuclear medicine imaging modality that can be used to differentiate adrenal cortical adenomas from carcinomas. This is a labeled cholesterol analogue that specifically binds to low-density lipoproteins and after receptor-mediated uptake it is stored in the adrenocortical cells (70).  NP 59 uptake is regulated by ACTH and suppressed by dexamethasone, concentrating in hyperfunctioning cortisol and aldosterone secreting adenomas and showing low uptake in adrenocortical carcinomas because of the inefficient concentration of radiotracer by malignant tissue (78).

 

Other Imaging Modalities for Ectopic Cushing’s

 

Patients diagnosed with ACTH dependent Cushing’s whose biochemical dynamic tests suggests an ectopic source, pose a special challenge to the clinician. In 12 to 20% of these patients, the source remains undiscovered despite repeated biochemical and radiological investigations (55).

 

In the setting of ectopic ACTH production, imaging studies play a crucial role in trying to identify the source of the tumor causing the disease and guide management and prognosis. The optimal imaging study to detect these tumors has not been defined. CT, MRI, PET scan, 111In-pentetreotide (OCT) scintigraphy at conventional or higher radionuclide doses, as well as newer molecular imaging techniques like 131I/123-metaiodobenzylguanidine (MIBG), 18F-fluoro-2-2-deoxyglucose-positron emission tomography (FDG-PET), 18F-fluorodopa-PET (F-DOPA-PET), 68Ga-DOTATATE-PET/CT or 68Ga-DOTATOC-PET/CT scan (68Gallium-SSTR-PET/CT) are complementary and have been shown to be useful in different scenarios with variable sensitivity and specificity (79-83). For the most part at least two different imaging modalities are needed to establish a diagnosis and sometimes, repeated imaging over several months is required to identify the source. The choice of imaging modalities is guided by the sensitivity of the procedure balanced with the risk of false-positive findings (72).

 

A good approach is to start by obtaining images of the chest, since most ACTH-secreting tumors are located in this area. The most common causes are bronchial carcinoid tumors and small cell lung cancer. Other sources of excess ACTH production include neuroendocrine tumors of the thymus, bowel and pancreas, medullary carcinoma of the thyroid, pheochromocytomas, and mesotheliomas.

 

CT of the chest, abdomen and pelvis with intravenous contrast medium injection is the most commonly used initial imaging test performed and is very useful in many cases. In patients with equivocal CT imaging findings, MRI can be useful, particularly for tumors within the abdomen. It is recommended to follow CT and MRI imaging with a functional imaging modality, being OCT scintigraphy the most widely used. Functional imaging reduces false-positive results because it relies on the specific properties of tumor cells, not just their anatomic characteristics. However, tumors lacking the relevant receptor can have false negative results (83). Site-specific differences occur and different imaging modalities might have higher sensitivity and specificity depending on this. A recent systematic review showed that FDG-PET can be very sensitive in the detection of neuroendocrine tumors with high proliferation index, particularly in the pancreas. This review also showed that 68Gallium-SSTR-PET/CT had a 100% sensitivity but this is an imaging technique that has limited availability and was only performed in a minority of the patients in their series (80).

 

Table 2. Imaging Characteristics of Adrenal Tumors

Characteristic

Adenoma

Carcinoma

Pheochromocytoma

Metastasis

Size

<4 cm

>4 cm

Variable

>4 cm

Shape

Round

Irregular

Round

Irregular

Border

Smooth

Irregular

Well delineated

Irregular

Laterality

Unilateral

Unilateral

May be bilateral or unilateral

May be bilateral

Appearance

Round, homogeneous

Inhomogeneous with central necrosis. May have calcifications

Cystic and hemorrhagic changes.

Inhomogeneous

Vascularity

Normal

Increased

Increased

Increased

Growth rate

Slow (1 cm/year)

Fast (>2 cm/year)

Slow (0.5-1 cm/year)

Variable/Fast

Lipid content

Lipid rich or poor

Lipid poor

Lipid poor

Lipid poor

CT attenuation

 

<10 HU unenhanced.

>50% absolute washout.

>20 HU unenhanced.

<50% absolute washout.

>20 HU unenhanced.

<50% absolute washout.

>20 HU unenhanced.

<50% absolute washout.

MRI

Isointense with liver in T1 and T2-w.

Chemical shift

Hypointense compared to liver on T1-w

High to intermediate signal on T2-w

High signal intensity on T2-w

Hypointense compared to liver on T1-w

High to intermediate signal on T2-w

FDG-PET-CT

Low SUV

High SUV

Variable SUV

High SUV

Other

 

Evidence of invasion or metastasis

 

History of prior cancer

 

 REFERENCES

 

  1. Nieman LK, Biller BM, Findling JW, Newell-Price J, Savage MO, Stewart PM, Montori VM 2008 The diagnosis of Cushing's syndrome: an Endocrine Society Clinical Practice Guideline. J Clin Endocrinol Metab 93:1526-1540.
  2. Ceccato F, Barbot M, Zilio M, Frigo AC, Albiger N, Camozzi V, Antonelli G, Plebani M, Mantero F, Boscaro M, Scaroni C 2015 Screening Tests for Cushing's Syndrome: Urinary Free Cortisol Role Measured by LC-MS/MS. J Clin Endocrinol Metab 100:3856-3861.
  3. Elias PC, Martinez EZ, Barone BF, Mermejo LM, Castro M, Moreira AC 2014 Late-night salivary cortisol has a better performance than urinary free cortisol in the diagnosis of Cushing's syndrome. J Clin Endocrinol Metab 99:2045-2051.
  4. Pappachan JM, Hariman C, Edavalath M, Waldron J, Hanna FW 2017 Cushing's syndrome: a practical approach to diagnosis and differential diagnoses. J Clin Pathol 70:350-359.
  5. Cunningham JM, Buxton OM, Weiss RE 2002 Circadian variation in Cushing's disease and pseudo-Cushing states by analysis of F and ACTH pulsatility. J Endocrinol Invest 25:791-799.
  6. Bonte HA, van den Hoven RJ, van der Sluijs Veer G, Vermes I 1999 The use of free cortisol index for laboratory assessment of pituitary-adrenal function. Clin Chem Lab Med 37:127-132.
  7. Durber SM, Daly JR 1976 A simple method for the determination of free cortisol in plasma, with normal levels in men and women, including women taking low oestrogen contraceptives. Clin Chim Acta 68:43-48.
  8. McWhinney BC, Briscoe SE, Ungerer JP, Pretorius CJ 2010 Measurement of cortisol, cortisone, prednisolone, dexamethasone and 11-deoxycortisol with ultra high performance liquid chromatography-tandem mass spectrometry: Application for plasma, plasma ultrafiltrate, urine and saliva in a routine laboratory. J Chromatogr B Analyt Technol Biomed Life Sci 878:2863-2869.
  9. Aranda G, Careaga M, Hanzu FA, Patrascioiu I, Rios P, Mora M, Morales-Romero B, Jimenez W, Halperin I, Casals G 2016 Accuracy of immunoassay and mass spectrometry urinary free cortisol in the diagnosis of Cushing's syndrome. Pituitary 19:496-502.
  10. de Bos Kuil MJJ, Endert E, Fliers E, Prummel MF, Romijn JA, Wiersinga WM 1998 Establishment of reference values for endocrine tests.I:  Cushing's syndrome. Neth J Med 53:153-163.
  11. Erickson D, Singh RJ, Sathananthan A, Vella A, Bryant SC 2012 Late-night salivary cortisol for diagnosis of Cushing's syndrome by liquid chromatography/tandem mass spectrometry assay. Clin Endocrinol (Oxf) 76:467-472.
  12. Kannankeril J, Carroll T, Findling JW, Javorsky B, Gunsolus IL, Phillips J, Raff H 2020 Prospective Evaluation of Late-Night Salivary Cortisol and Cortisone by EIA and LC-MS/MS in Suspected Cushing Syndrome. J Endocr Soc 4:bvaa107.
  13. Raff H 2009 Utility of salivary cortisol measurements in Cushing's syndrome and adrenal insufficiency. J Clin Endocrinol Metab 94:3647-3655.
  14. Raff H 2013 Update on late-night salivary cortisol for the diagnosis of Cushing's syndrome: methodological considerations. Endocrine 44:346-349.
  15. Greene LW, Geer EB, Page-Wilson G, Findling JW, Raff H 2019 Assay-Specific Spurious ACTH Results Lead to Misdiagnosis, Unnecessary Testing, and Surgical Misadventure-A Case Series. J Endocr Soc 3:763-772.
  16. Morita K, Ogawa M, Kimura M, Okawa Y, Oki Y, Edo N, Tsukamoto K, Ishikawa T 2019 Falsely elevated plasma ACTH levels measured by the Elecsys assay related to heterophilic antibody in a case of secondary adrenocortical insufficiency. Endocr J 66:563-569.
  17. Husebye ES, Allolio B, Arlt W, Badenhoop K, Bensing S, Betterle C, Falorni A, Gan EH, Hulting AL, Kasperlik-Zaluska A, Kampe O, Lovas K, Meyer G, Pearce SH 2014 Consensus statement on the diagnosis, treatment and follow-up of patients with primary adrenal insufficiency. J Intern Med 275:104-115.
  18. Landon J, Greenwood FC, Stamp TC, Wynn V 1966 The plasma sugar, free fatty acid, cortisol, and growth hormone response to insulin, and the comparison of this procedure with other tests of pituitary and adrenal function. II. In patients with hypothalamic or pituitary dysfunction or anorexia nervosa. J Clin Invest 45:437-449.
  19. Dickstein G 2001 Hypothalamo-pituitary-adrenal axis testing:nothis is sacred and caution in interpretation is needed. Clin Endocrinol 54:15-16.
  20. Pfeifer M, Kanc K, Verhovec R, Kocijancic A 2001 Reproducibility of the insulin tolerance test (ITT) for assessment of growth hormone and cortisol secretion in normal and hypopituitary adult men. Clin Endocrinol (Oxf) 54:17-22.
  21. Hamrahian AH, Oseni TS, Arafah BM 2004 Measurements of serum free cortisol in critically ill patients. N Engl J Med 350:1629-1638.
  22. Klose M, Lange M, Rasmussen AK, Skakkebaek NE, Hilsted L, Haug E, Andersen M, Feldt-Rasmussen U 2007 Factors influencing the adrenocorticotropin test: role of contemporary cortisol assays, body composition, and oral contraceptive agents. J Clin Endocrinol Metab 92:1326-1333.
  23. Dokmetas HS, Colak R, Kelestimur F, Selcuklu A, Unluhizarci K, Bayram F 2000 A comparison between the 1-microg adrenocorticotropin (ACTH) test, the short ACTH (250 microg) test, and the insulin tolerance test in the assessment of hypothalamo-pituitary-adrenal axis immediately after pituitary surgery. J Clin Endocrinol Metab 85:3713-3719.
  24. Javorsky BR, Raff H, Carroll TB, Algeciras-Schimnich A, Singh RJ, Colon-Franco JM, Findling JW 2021 New Cutoffs for the Biochemical Diagnosis of Adrenal Insufficiency after ACTH Stimulation using Specific Cortisol Assays. J Endocr Soc 5:bvab022.
  25. Oilkers W, Diederich S, Bahr V 1992 Diagnosis and therapy surveillance in addison's disease:Rapid adrenocorticotropin (ACTH) test and measurement of plasma ACTH, renin activity, and aldosterone. J Clin Endocrinol Metab 75:259-264.
  26. Dickstein G 1997 Low-dose ACTH test- a word of caution to the word of caution:when ahd how to use it (Letter). J Clin Endocrinol Metab 82:322.
  27. Laureti S, Arvat E, Candeloro P, Di Vito L, Ghigo E, Santeusanio F, Falorni A 2000 Low dose (1 microg) ACTH test in the evaluation of adrenal dysfunction in pre-clinical Addison's disease. Clin Endocrinol (Oxf) 53:107-115.
  28. Kannisto S, Korppi M, Remes K, Voutilainen R 2000 Adrenal suppression, evaluated by a low dose adrenocorticotropin test, and growth in asthmatic children treated with inhaled steroids. J Clin Endocrinol Metab 85:652-657.
  29. Ospina NS, Al Nofal A, Bancos I, Javed A, Benkhadra K, Kapoor E, Lteif AN, Natt N, Murad MH 2016 ACTH Stimulation Tests for the Diagnosis of Adrenal Insufficiency: Systematic Review and Meta-Analysis. J Clin Endocrinol Metab 101:427-434.
  30. Dickstein G 2001 Peak cortisol response to insulin tolerance test: how reproducible is it really? Clin Endocrinol (Oxf) 55:819-820; author reply 820-811.
  31. Fiad TM, Kirby JM, Cunningham SK, McKenna TJ 1994 The overnight single-dose metyrapone test is a simple and reliable index of the hypothalamic-pituitary-axis. Clin Endocrinol 40:603-609.
  32. Jubiz W, Levinson RA, Meikle AW, West CD, Tyler FH 1970 Absorption and conjugation of metyrapone during diphenylhydantoin therapy:mechanism of the abnormal response to oral metyrapone. Endocrinology 86:328-331.
  33. Meikle AW, Jubiz W, Matsukura S, Harada G, West CD, Tyler FH 1970 Effect of estrogen on the metabolism of metyrapone and release of ACTH. J Clin Endocrinol Metab 30:259-263.
  34. Hindmarsh PC, Brook CGD 1985 Single dose dexamethasone suppression test in children:dose relationship to body size. Clin Endocrinol 23:67-.
  35. Mahajan DK, Wahlen JD, Tyler FH, West CD 1972 Plasma 11-deoxycortisol radioimmunoassay for metyrapone tests. Steroids 20:609.
  36. Al-Saadi N, Diederich S, Oelkers W 1998 A very high dose dexamethasone suppression test for the differential diagnosis of Cushing's syndrome. Clin Endocrinol 48:45-51.
  37. Alwani RA, Schmit Jongbloed LW, de Jong FH, van der Lely AJ, de Herder WW, Feelders RA 2014 Differentiating between Cushing's disease and pseudo-Cushing's syndrome: comparison of four tests. Eur J Endocrinol 170:477-486.
  38. Chabre O 2018 The difficulties of pseudo-Cushing's syndrome (or "non-neoplastic hypercortisolism"). Ann Endocrinol (Paris) 79:138-145.
  39. Yanovski JA, Cutler GB, Jr., Chrousos GP, Nieman LK 1998 The dexamethasone-suppressed corticotropin-releasing hormone stimulation test differentiates mild Cushing's disease from normal physiology. J Clin Endocrinol Metab 83:348-352.
  40. Chrousos GP, Schulte HM, Oldfield EH, Gold PW, Cutler GB, Jr., Loriaux DL 1984 The corticotropin-releasing factor stimulation test.An aid in the evaluation of patient's with Cushing's syndrome. N Engl J Med 310:622-626.
  41. Nieman LK, Oldfield EH, Wesley R, Chrousos GP, Loriaux DL, Cutler GB, Jr. 1993 A simplified morning ovine corticotropin-releasing hormone stimulation test for the differential diagnosis of adrenocorticotropin-dependent Cushing's syndrome. J Clin Endocrinol Metab 77:1308-1312.
  42. Kyriazopoulou V, Vagenakis AG 1992 Abnormal overnight dexamethasone suppression test in subjects receiving rifampicin therapy. J Clin Endocrinol Metab 75:315-317.
  43. Frete C, Corcuff JB, Kuhn E, Salenave S, Gaye D, Young J, Chanson P, Tabarin A 2020 Non-invasive Diagnostic Strategy in ACTH-dependent Cushing's Syndrome. J Clin Endocrinol Metab 105.
  44. Newell-Price J, Perry L, Medbak S, Monson J, Savage M, Besser M, Grossman A 1997 A combined test using desmopressin and corticotropin-releasing hormone in the differential diagnosis of Cushing's syndrome. J Clin Endocrinol Metab 82:176-181.
  45. Vassiliadi DA, Tsagarakis S 2018 DIAGNOSIS OF ENDOCRINE DISEASE: The role of the desmopressin test in the diagnosis and follow-up of Cushing's syndrome. Eur J Endocrinol 178:R201-R214.
  46. Malerbi DA, Mendonca BB, Liberman B, Toledo SP, Corradini MC, Cunha-Neto MB, Fragoso MC, Wajchenberg BL 1993 The desmopressin stimulation test in the differential diagnosis of Cushing's syndrome. Clin Endocrinol (Oxf) 38:463-472.
  47. Moro M, Putignano P, Losa M, Invitti C, Maraschini C, Cavagnini F 2000 The desmopressin test in the differential diagnosis between Cushing's disease and pseudo-Cushing states. J Clin Endocrinol Metab 85:3569-3574.
  48. Manni A, Latshaw RF, Page R, Santen RJ 1983 Simultanoues bilateral venous sampling for adrenocorticotropin in pituitary-dependent Cushing's disease:Evidence for lateralization of pituitary venous drainage. J Clin Endocrinol Metab 57:1070-1073.
  49. Wiggam MI, Heaney AP, McIlrath EM, McCance DR, Sheridan B, Hadden DR, Atkinson AB 2000 Bilateral inferior petrosal sinus sampling in the differential diganosis of adrenocorticotropin-dependent Cushing's syndrome:A comparison with other diagnostic tests. J Clin Endocrinol Metab 85:1525-1532.
  50. Obuobie K, Davies JS, DOgunko A, Scanlon MF 2000 Venous thrombo-emoblism following inferior petrosal sinus sampling in Cushing's disease. J Enocrinol Invest 23:542-544.
  51. Buurman H, Saeger W 2006 Subclinical adenomas in postmortem pituitaries: classification and correlations to clinical data. Eur J Endocrinol 154:753-758.
  52. Osamura RY, Kajiya H, Takei M, Egashira N, Tobita M, Takekoshi S, Teramoto A 2008 Pathology of the human pituitary adenomas. Histochem Cell Biol 130:495-507.
  53. Woo YS, Isidori AM, Wat WZ, Kaltsas GA, Afshar F, Sabin I, Jenkins PJ, Monson JP, Besser GM, Grossman AB 2005 Clinical and biochemical characteristics of adrenocorticotropin-secreting macroadenomas. J Clin Endocrinol Metab 90:4963-4969.
  54. Hall WA, Luciano MG, Doppman JL, Patronas NJ, Oldfield EH 1994 Pituitary magnetic resonance imaging in normal human volunteers: occult adenomas in the general population. Ann Intern Med 120:817-820.
  55. Ilias I, Torpy DJ, Pacak K, Mullen N, Wesley RA, Nieman LK 2005 Cushing's syndrome due to ectopic corticotropin secretion: twenty years' experience at the National Institutes of Health. J Clin Endocrinol Metab 90:4955-4962.
  56. Testa RM, Albiger N, Occhi G, Sanguin F, Scanarini M, Berlucchi S, Gardiman MP, Carollo C, Mantero F, Scaroni C 2007 The usefulness of combined biochemical tests in the diagnosis of Cushing's disease with negative pituitary magnetic resonance imaging. Eur J Endocrinol 156:241-248.
  57. Sahdev A, Reznek RH, Evanson J, Grossman AB 2007 Imaging in Cushing's syndrome. Arq Bras Endocrinol Metabol 51:1319-1328.
  58. Vitale G, Tortora F, Baldelli R, Cocchiara F, Paragliola RM, Sbardella E, Simeoli C, Caranci F, Pivonello R, Colao A, Group ABC 2017 Pituitary magnetic resonance imaging in Cushing's disease. Endocrine 55:691-696.
  59. Colombo N, Loli P, Vignati F, Scialfa G 1994 MR of corticotropin-secreting pituitary microadenomas. AJNR Am J Neuroradiol 15:1591-1595.
  60. Friedman TC, Zuckerbraun E, Lee ML, Kabil MS, Shahinian H 2007 Dynamic pituitary MRI has high sensitivity and specificity for the diagnosis of mild Cushing's syndrome and should be part of the initial workup. Horm Metab Res 39:451-456.
  61. Tabarin A, Laurent F, Catargi B, Olivier-Puel F, Lescene R, Berge J, Galli FS, Drouillard J, Roger P, Guerin J 1998 Comparative evaluation of conventional and dynamic magnetic resonance imaging of the pituitary gland for the diagnosis of Cushing's disease. Clin Endocrinol (Oxf) 49:293-300.
  62. Kasaliwal R, Sankhe SS, Lila AR, Budyal SR, Jagtap VS, Sarathi V, Kakade H, Bandgar T, Menon PS, Shah NS 2013 Volume interpolated 3D-spoiled gradient echo sequence is better than dynamic contrast spin echo sequence for MRI detection of corticotropin secreting pituitary microadenomas. Clin Endocrinol (Oxf) 78:825-830.
  63. Patronas N, Bulakbasi N, Stratakis CA, Lafferty A, Oldfield EH, Doppman J, Nieman LK 2003 Spoiled gradient recalled acquisition in the steady state technique is superior to conventional postcontrast spin echo technique for magnetic resonance imaging detection of adrenocorticotropin-secreting pituitary tumors. J Clin Endocrinol Metab 88:1565-1569.
  64. Escourolle H, Abecassis JP, Bertagna X, Guilhaume B, Pariente D, Derome P, Bonnin A, Luton JP 1993 Comparison of computerized tomography and magnetic resonance imaging for the examination of the pituitary gland in patients with Cushing's disease. Clin Endocrinol (Oxf) 39:307-313.
  65. Faje A, Tritos NA, Swearingen B, Klibanski A 2016 Neuroendocrine disorders: pituitary imaging. Handb Clin Neurol 136:873-885.
  66. Grumbach MM, Biller BM, Braunstein GD, Campbell KK, Carney JA, Godley PA, Harris EL, Lee JK, Oertel YC, Posner MC, Schlechte JA, Wieand HS 2003 Management of the clinically inapparent adrenal mass ("incidentaloma"). Ann Intern Med 138:424-429.
  67. Bovio S, Cataldi A, Reimondo G, Sperone P, Novello S, Berruti A, Borasio P, Fava C, Dogliotti L, Scagliotti GV, Angeli A, Terzolo M 2006 Prevalence of adrenal incidentaloma in a contemporary computerized tomography series. J Endocrinol Invest 29:298-302.
  68. Cawood TJ, Hunt PJ, O'Shea D, Cole D, Soule S 2009 Recommended evaluation of adrenal incidentalomas is costly, has high false-positive rates and confers a risk of fatal cancer that is similar to the risk of the adrenal lesion becoming malignant; time for a rethink? Eur J Endocrinol 161:513-527.
  69. Loh HH, Yee A, Loh HS, Sukor N, Kamaruddin NA 2017 The natural progression and outcomes of adrenal incidentaloma: a systematic review and meta-analysis. Minerva Endocrinol 42:77-87.
  70. Ilias I, Sahdev A, Reznek RH, Grossman AB, Pacak K 2007 The optimal imaging of adrenal tumours: a comparison of different methods. Endocr Relat Cancer 14:587-599.
  71. Dinnes J, Bancos I, Ferrante di Ruffano L, Chortis V, Davenport C, Bayliss S, Sahdev A, Guest P, Fassnacht M, Deeks JJ, Arlt W 2016 MANAGEMENT OF ENDOCRINE DISEASE: Imaging for the diagnosis of malignancy in incidentally discovered adrenal masses: a systematic review and meta-analysis. Eur J Endocrinol 175:R51-64.
  72. Hamrahian AH, Ioachimescu AG, Remer EM, Motta-Ramirez G, Bogabathina H, Levin HS, Reddy S, Gill IS, Siperstein A, Bravo EL 2005 Clinical utility of noncontrast computed tomography attenuation value (hounsfield units) to differentiate adrenal adenomas/hyperplasias from nonadenomas: Cleveland Clinic experience. J Clin Endocrinol Metab 90:871-877.
  73. Rockall AG, Babar SA, Sohaib SA, Isidori AM, Diaz-Cano S, Monson JP, Grossman AB, Reznek RH 2004 CT and MR imaging of the adrenal glands in ACTH-independent cushing syndrome. Radiographics 24:435-452.
  74. Groussin L, Bonardel G, Silvera S, Tissier F, Coste J, Abiven G, Libe R, Bienvenu M, Alberini JL, Salenave S, Bouchard P, Bertherat J, Dousset B, Legmann P, Richard B, Foehrenbach H, Bertagna X, Tenenbaum F 2009 18F-Fluorodeoxyglucose positron emission tomography for the diagnosis of adrenocortical tumors: a prospective study in 77 operated patients. J Clin Endocrinol Metab 94:1713-1722.
  75. Boland GW, Dwamena BA, Jagtiani Sangwaiya M, Goehler AG, Blake MA, Hahn PF, Scott JA, Kalra MK 2011 Characterization of adrenal masses by using FDG PET: a systematic review and meta-analysis of diagnostic test performance. Radiology 259:117-126.
  76. Hahner S, Stuermer A, Kreissl M, Reiners C, Fassnacht M, Haenscheid H, Beuschlein F, Zink M, Lang K, Allolio B, Schirbel A 2008 [123 I]Iodometomidate for molecular imaging of adrenocortical cytochrome P450 family 11B enzymes. J Clin Endocrinol Metab 93:2358-2365.
  77. Metser U, Miller E, Lerman H, Lievshitz G, Avital S, Even-Sapir E 2006 18F-FDG PET/CT in the evaluation of adrenal masses. J Nucl Med 47:32-37.
  78. Wong KK, Miller BS, Viglianti BL, Dwamena BA, Gauger PG, Cook GJ, Colletti PM, Rubello D, Gross MD 2016 Molecular Imaging in the Management of Adrenocortical Cancer: A Systematic Review. Clin Nucl Med 41:e368-382.
  79. de Herder WW, Krenning EP, Malchoff CD, Hofland LJ, Reubi JC, Kwekkeboom DJ, Oei HY, Pols HA, Bruining HA, Nobels FR, et al. 1994 Somatostatin receptor scintigraphy: its value in tumor localization in patients with Cushing's syndrome caused by ectopic corticotropin or corticotropin-releasing hormone secretion. Am J Med 96:305-312.
  80. Isidori AM, Sbardella E, Zatelli MC, Boschetti M, Vitale G, Colao A, Pivonello R, Group ABCS 2015 Conventional and Nuclear Medicine Imaging in Ectopic Cushing's Syndrome: A Systematic Review. J Clin Endocrinol Metab 100:3231-3244.
  81. Pacak K, Ilias I, Chen CC, Carrasquillo JA, Whatley M, Nieman LK 2004 The role of [(18)F]fluorodeoxyglucose positron emission tomography and [(111)In]-diethylenetriaminepentaacetate-D-Phe-pentetreotide scintigraphy in the localization of ectopic adrenocorticotropin-secreting tumors causing Cushing's syndrome. J Clin Endocrinol Metab 89:2214-2221.
  82. Santhanam P, Taieb D, Giovanella L, Treglia G 2015 PET imaging in ectopic Cushing syndrome: a systematic review. Endocrine 50:297-305.
  83. Zemskova MS, Gundabolu B, Sinaii N, Chen CC, Carrasquillo JA, Whatley M, Chowdhury I, Gharib AM, Nieman LK 2010 Utility of various functional and anatomic imaging modalities for detection of ectopic adrenocorticotropin-secreting tumors. J Clin Endocrinol Metab 95:1207-1219.