Archives

Dyslipidemia in Chronic Kidney Disease

ABSTRACT

Chronic kidney disease (CKD) is associated with a dyslipidemia comprising high triglycerides, low HDL-cholesterol and altered lipoprotein composition. Cardiovascular diseases are the leading cause of mortality in CKD, especially in end stage renal disease patients. Thus, therapies to reduce cardiovascular risk are urgently needed in CKD. Robust clinical trial evidence has found that use of statins in pre-end stage CKD patients, as well as in renal transplant recipients, can decrease cardiovascular events; however, providers need to be aware of dose restrictions for statin therapy in CKD subjects. Furthermore, statin therapy does not reduce cardiovascular events in dialysis patients, nor does statin therapy confer any protection against progression of renal disease. Niacin and fibrates are effective in lipid lowering in CKD and appear to have some cardiovascular benefit but further study is needed to clearly define their role. Novel therapies with PCSK 9 inhibitors, bempedoic acid, and inclisiran have all been shown to decrease LDL cholesterol levels but there is currently limited data for reduction of cardiovascular events or mortality in patients with CKD/ESRD. This article reviews the epidemiology of CKD, association of CKD with cardiovascular events, and the effects of CKD on lipid levels and metabolism. The article discusses clinical trial evidence for and against statin and non-statin lipid lowering therapy in CKD patients.

CHRONIC KIDNEY DISEASE (CKD) EPIDEMIOLOGY

Chronic kidney disease (CKD) is defined as renal impairment greater than 3 months duration that results in an estimated glomerular filtration rate (eGFR) < 60ml/min/1.73m2. CKD is classified into 5 stages based on the eGFR (Table 1). CKD is a world-wide health problem with rising incidence and prevalence. CKD, especially in the early stages is often asymptomatic; thus, the actual prevalence may be even higher than estimated. End stage renal disease (ESRD) is defined as needing dialysis or transplant, and the prevalence and incidence of ESRD have doubled over the past 10 years (1). The annual mortality rate of dialysis patients is greater than 20%. The burden of co-morbidities and the cost of caring for CKD patients is high, and thus a major focus is increased screening and early detection of CKD when interventions to delay or prevent progression to ESRD may be effective. There are multiple causes of CKD with the most common causes in Westernized nations being hypertension and diabetes; however, a wide range of etiologies including infectious, auto-immune, genetic, obstructive, and ischemic injury are all prevalent. There are ethnic differences in susceptibility with increased prevalence in Mexican-Americans and non-Hispanic blacks compared to Caucasians (2).

Table 1. Stages of CKD

CKD stage

GFR (ml/min/1.73 m2)

CKD 1

≥ 90 (with renal damage or injury)

CKD 2 (mild)

60-89

CKD 3 (moderate)

30-59

CKD 4 (severe)

15-29

CKD 5 (end stage)

<15, dialysis, or transplant

While the burden of CKD itself is significant, the leading causes of morbidity and mortality in CKD are cardiovascular diseases (CVD), primarily atherosclerotic coronary artery disease. Risk factors for CVD in CKD include the traditional risk factors – hypertension, sex, age, smoking, and family history and CKD patients appear to benefit similar to non-CKD patients from therapies targeting these risk factors. Regardless of the cause of CKD, patients with CKD are at increased risk for CVD, which has led to the National Kidney Foundation classifying all patients with CKD as “highest risk” for CVD regardless of their levels of traditional CVD risk factors. The focus of this chapter is on the dyslipidemia of CKD and the risk of CVD in CKD.

Nephrotic Syndrome

Nephrotic syndrome differs from other types of CKD in its presentation and risks. Nephrotic syndrome is comprised of significant proteinuria (typically > 3g/24h), hypoalbuminemia, peripheral (+/- central) edema, and significant hyperlipidemia and lipiduria may also be seen. It is frequently seen in children, and the etiology includes minimal change disease (up to 85%), focal segmental glomerulosclerosis (up to 15%), and secondary causes (rare) including systemic lupus erythematosus, Henoch Schonlein Purpura, or membrano-proliferative glomerulopathy. In adults, the etiology is more likely to involve a systemic disease such as diabetes, amyloidosis, or lupus. Nephrotic syndrome may be transient or persistent. Most (approximately 80% of children) cases of nephrotic syndrome are successfully treated with glucocorticoids with resolution of all features including hyperlipidemia; however, steroid-resistant nephrotic syndrome patients often have persistent dyslipidemia, which may place them at increased risk for CVD. For example, a small study found increased CVD markers including pulse wave velocity, carotid artery intima-media thickness, and left ventricular mass in patients with steroid-resistant nephrotic syndrome compared to controls (3), implying increased risk for CVD events. Treatment of nephrotic syndrome dyslipidemia includes therapies specifically targeting the renal disease (primarily glucocorticoids, but also renin-angiotensin system antagonists which can help decrease proteinuria) and lipid lowering agents.

 CVD IN CKD

CVD accounts for 40-50% of all deaths in ESRD patients, with CVD mortality rates approximately 15 times that seen in the general population (4). However, CVD is highly prevalent in patients who progress to ESRD implying that earlier stages of CKD increase the development of CVD. A number of factors have been proposed as risk factors for CVD in CKD including proteinuria, inflammation, anemia, malnutrition, oxidative stress, and uremic toxins (5). Ongoing research is investigating whether these (and other) markers may be therapeutic targets. Interestingly, proteinuria correlates with blood pressure, total cholesterol, triglycerides, and inversely correlates with HDL-cholesterol (6). Thus, it remains unclear if proteinuria itself is a risk factor (e.g., a cause of CVD) or a biomarker. Meta-analyses of general population and high-risk population cohorts found that both lower eGFR (<60 ml/min/1.73 m2) and higher albuminuria (>10 mg/g creatinine) are predictors of total mortality and CVD mortality; furthermore, eGFR and albuminuria are independent of each other and of traditional CVD risk factors (7, 8). Estimated GFR > 60 ml/min/1.73 m2 is not a risk factor for CVD or total mortality.

Dyslipidemia in CKD

EFFECT OF CKD ON LIPID LEVELS

CKD is associated with a dyslipidemia comprised of elevated triglycerides and low HDL-cholesterol. Levels of LDL-cholesterol (and thus, total cholesterol) are generally not elevated; however, proteinuria correlates with cholesterol and triglycerides. CKD leads to a down regulation of lipoprotein lipase and the LDL-receptor, and increased triglycerides in CKD are due to delayed catabolism of triglyceride rich lipoproteins, with no differences in production rate (9). CKD is associated with lower levels of apoA-I (due to decreased hepatic expression (10)) and higher apoB/apoA-I. Decreased lecithin-cholesterol acyltransferase (LCAT) activity and increased cholesteryl ester transfer protein (CETP) activity contribute to decreased HDL-cholesterol levels. Beyond decreased HDL cholesterol levels, the HDL in CKD is less effective in its anti-oxidative and anti-inflammatory functions [for review see (11)].

As CKD progresses the dyslipidemia often worsens. In an evaluation of 2001-2010 National Health and Nutrition Examination Survey (NHANES), the prevalence of dyslipidemia increased from 45.5% in CKD stage 1 to 67.8% in CKD stage 4; similarly, the use of lipid lowering agents increased from 18.1% in CKD stage 1 to 44.7% in CKD stage 4 (12).  Of more than 1000 hemodialysis patients studied only 20% had “normal” lipid levels (defined as LDL<130 mg/dl, HDL > 40 and triglycerides < 150); of 317 peritoneal dialysis patients only 15% had “normal” lipid levels (13). A larger study evaluating dyslipidemia in > 21,000 incident dialysis patients found 82% prevalence of dyslipidemia and suggested a threshold of non-HDL cholesterol > 100 mg/dl (2.6mmol/L) to identify dyslipidemia in CKD stage 5 subjects (14). Peritoneal dialysis is associated with higher cholesterol levels than hemodialysis, although the reasons aren’t fully understood. In subjects who switched from peritoneal dialysis to hemodialysis there was a drop in cholesterol levels of almost 20% following transition (15). The National Kidney Foundation recommends routine screening of all adults and adolescents with CKD using a standard fasting lipid profile (total cholesterol, LDL-cholesterol, HDL-cholesterol and triglycerides), and follows the classification of the National Cholesterol Education Panel for levels (desirable, borderline or high). Although some studies have found associations between Lp(a) and dialysis patients, this is not well defined and there is no current indication for routine screening of Lp(a).

EFFECT OF CKD ON LIPOPROTEIN COMPOSITION

Beyond simply measuring lipid levels, emerging evidence implies that lipoprotein particle size and composition is altered in CKD, with increased small dense LDL and decreased larger LDL particles in CKD subjects compared to controls (16). Small dense LDL is thought to be more atherogenic than larger LDL particles. An emerging theory is that beyond lipid levels or lipoprotein size, lipoprotein particle “cargo” can affect atherosclerosis development and progression. Lipoprotein particles transport numerous bioactive lipids, microRNAs, other small RNAs, proteins, hormones, etc. For example, a recent study compared LDL particle composition between subjects with stage 4/5 CKD and non-CKD controls, and found similar total lipid and cholesterol content, but altered content of various lipid subclasses; for example decreased phosphatidylcholines, sulfatides, and ceramides and increased N-acyltaurines (17). Many of these lipid species are known to have either pro- or anti-atherogenic properties and thus could directly affect atherogenesis.

EFFECT OF RENAL TRANSPLANTATION ON LIPID LEVELS

Dyslipidemia is frequently seen in renal transplant recipients, including increased total cholesterol, LDL-cholesterol, and triglycerides, and decreased HDL-cholesterol. The dyslipidemia may have existed pre-transplant or be related to transplantation associated factors. Cyclosporine increases LDL-cholesterol via both increased production and decreased clearance. Corticosteroids increase both cholesterol and triglyceride levels in a dose-dependent manner. The adverse effects of cyclosporine and corticosteroids on lipid levels appear to be additive (18). Tacrolimus and azathioprine appear to have less induction of dyslipidemia than cyclosporine (19). Sirolimus increases both cholesterol and triglycerides, in part due to decreased LDL-clearance (20).

EFFECT OF NEPHROTIC SYNDROME ON LIPID LEVELS

The dyslipidemia in nephrotic syndrome can be striking with significant elevations of cholesterol, LDL-cholesterol, triglycerides, and lipoprotein(a); HDL cholesterol is often low, especially HDL2. The cause of elevated lipid levels is multi-factorial, including reduction in oncotic pressure which stimulates apoB synthesis (although the exact mechanism by which this occurs is not known), decreased metabolism of lipoproteins, and decreased clearance. Patients with nephrotic syndrome have decreased LDL-receptor activity and increased acyl-CoA cholesterol acytransferase (ACAT) and HMG-CoA reductase activity leading to increased LDL-cholesterol levels (21, 22). Low HDL-cholesterol is thought to be due at least in part to LCAT deficiency secondary to accelerated renal loss of LCAT (23). Triglycerides are elevated due to impaired clearance of chylomicrons and triglyceride-rich lipoproteins, as well as increased triglyceride production (24).

EVIDENCE FOR/AGAINST LIPID LOWERING THERAPY IN CKD FOR CVD OUTCOMES

Given the high prevalence of CVD in CKD, and the robust clinical evidence in non-CKD subjects that lipid lowering reduces CVD outcomes, there is great interest in using lipid lowering therapy in CKD subjects. Statins are the most commonly used lipid-lowering medications and thus far have been shown to reduce CVD events and/or mortality in virtually every population studied. However, CKD patients seem to be a unique population in that at present there is no evidence of benefit for CVD outcomes in dialysis patients with statin therapy. The Canadian Journal of Cardiology lists CKD as a statin indicated condition in its newest guidelines published in 2021 (25) while AHA/ACC lists CKD as a risk enhancer but not a high-risk condition based on 2018 guidelines (26).  Despite growing evidence to support CKD as a CVD risk equivalent, the use of statin therapy in CKD does not appear to be rising more than in the non-CKD population based on data from Mefford et al looking at trends in statin use amongst US adults with CKD from 1999-2014 (27). As discussed below it appears that statins can reduce CVD events in pre-end stage CKD subjects, and in post-renal transplant subjects, but not in dialysis patients (Table 2).

Use of Statins in Pre-ESRD CKD Patients

Although many of the initial statin CVD studies did not include many CKD patients, evidence from sub-group analyses of large statin studies suggested that CKD subjects had similar benefits to non-CKD individuals. For example, the Heart Protection Study (HPS) which assessed >20,000 subjects at high risk of CVD included a subgroup of 1329 subjects with impaired kidney function. In this subgroup those that received simvastatin had a 28% proportional risk reduction and an 11% absolute risk reduction of a major cardiovascular event compared to those randomized to placebo; similar to the effect on the overall cohort (28). Further, in the Pravastatin Pooling Project, 4,991 subjects with CKD3 were examined and a 23% reduction in cardiovascular events was seen in the pravastatin group (29).  In a retrospective study with 47,200 subjects followed through the Department of Veterans Affairs, starting statin therapy 12 months prior to transitioning to ESRD conferred a reduction in 12-month all-cause mortality (HR 0.79), cardiovascular events (HR 0.83) and hospitalization rate (HR 0.89) (30). Several other studies or meta-analyses similarly predicted that CKD subjects would have reduction in CVD with statin therapy. For example, a meta-analysis of 38 studies with >37,000 participants with CKD but not yet on dialysis found a consistent reduction in major cardiovascular events, all-cause mortality, cardiovascular death, and myocardial infarction in statin users compared to placebo groups. There was no clear effect of statin on stroke, nor was there any effect of statin use on progression of the renal disease (31). Thus, for CKD patients with pre-end stage renal disease statins effectively lower total cholesterol and LDL-cholesterol levels and decrease CVD risk. The different statins have different degrees of renal involvement in their metabolism, and providers should be aware of dose restrictions in CKD (Table 3).

Unclear Whether to Use Statins in Subjects with Nephrotic Syndrome

Several small clinical studies have investigated the use of lipid lowering therapies in nephrotic syndrome, but data is only available for statins and fibrates, and no CVD outcomes data is available. Several small studies using statins have found efficacy in lowering LDL-cholesterol, and that statins were safe and well tolerated (32, 33). Thus, the use of statins in nephrotic syndrome appears to be safe and efficacious in terms of lipid lowering; however, it is not clear if there is any corresponding benefit on either CVD or renal outcomes.

No Benefit to Statins in Subjects with only Microalbuminuria

The Prevention of Renal and Vascular Endstage Disease Intervention Trial (PREVEND IT) randomized 864 subjects with persistent microalbuminuria (urinary albumin of 15-300mg/24h x 2 samples) to fosinopril (an angiotensin converting enzyme inhibitor) or placebo and to pravastatin 20 mg or placebo. Inclusion criteria for the study included blood pressure <160/100 mm Hg and no use of antihypertensive medications and total cholesterol < 300 mg/dl (8 mmol/L) or < 192 mg/dl (5 mmol/L) if patient had known CVD and no use of lipid lowering medications. Although diabetes was not an exclusion criteria, <3% of the subjects had diabetes (34). The use of statin did not affect either urinary albumin excretion or cardiovascular events; however, the use of fosinopril significantly decreased albuminuria and had a trend to reduction in cardiovascular events. Thus, in the absence of other indications for statin therapy, there appears to be no benefit in subjects that solely have microalbuminuria. However, a subsequent analysis found that the subjects with isolated microalbuminuria had an increased risk for CVD events and mortality compared to those without risk factors (35); thus, isolated microalbuminuria appears to indicate high risk and further study is needed to determine effective therapies to reduce risk.

No Benefit to Statins in Dialysis Patients

Studies specifically examining the role of statins in ESRD subjects have not found a benefit. The Deutsche Diabetes Dialyse Studie (4D) randomized 1255 type 2 diabetic subjects on maintenance hemodialysis to either 20 mg atorvastatin or placebo daily. The cholesterol and LDL-cholesterol reduction was similar to that seen in non-dialysis patients; however, unlike non-CKD subjects there was no significant reduction in cardiovascular death, nonfatal myocardial infarction or stroke with atorvastatin compared to placebo (36). A long-term follow-up of the 4D study population found similar effects after 11.5 years as were found at the end of the original study: no CVD benefit, but also no evidence of harm (37). Similarly, A Study to Evaluate the Use of Rosuvastatin in Subjects on Regular Hemodialysis (AURORA) randomized 2776 subjects on maintenance hemodialysis to rosuvastatin 10 mg or placebo. Again, the LDL-cholesterol lowering in dialysis patients was similar to that seen in other studies in non-dialysis patients, but there was no significant effect on the primary endpoint of cardiovascular death, nonfatal myocardial infarction or stroke (38). The Study of Heart and Renal Protection (SHARP) randomized 9270 CKD patients (3023 on dialysis) to simvastatin plus ezetimibe versus placebo. Unlike 4D and AURORA, the SHARP study did report a significant reduction in major atherosclerotic events in the simvastatin plus ezetimibe group, including the dialysis subgroup (39). However, a meta-analysis of 25 studies involving 8289 dialysis patients found no benefit of statin therapy on major cardiovascular events, cardiovascular mortality, all-cause mortality or myocardial infarction, despite efficacious lipid lowering (40). Nevertheless, a post-hoc analysis of the 4D study did demonstrate a benefit of statin therapy in the subgroup that had LDL cholesterol > 145 mg/dl (3.76mmol/l) (41). Although the use of statins in dialysis patients does not clearly cause harm, at present there is no indication for use in dialysis patients, with the exception of a possible benefit in those with significant elevation in LDL-cholesterol.

WHY IS STATIN THERAPY INEFFECTIVE IN DIALYSIS SUBJECTS?

Given the robust data demonstrating statin efficacy in CVD risk reduction in virtually all other populations studied, the lack of efficacy in ESRD subjects in perplexing. However, it may be due to different mechanisms of disease progression in ESRD populations compared to other populations. In ESRD subjects there is increased inflammation and oxidative stress as well as increased non-lipid-associated pro-atherogenic factors, which may be the major cause of atherosclerosis development or progression in CKD subjects [for review see (42)]. Therefore, the relative impact of dyslipidemia on CVD development and progression in ESRD subjects may be less than in other CKD and non-CKD subjects, and thus the potential benefit of lipid lowering therapy is reduced. In ESRD subjects with significant hyperlipidemia (such as genetic hyperlipidemias) there may still be a role for statins, or other lipid lowering therapies. Furthermore, while no benefit has been found for statins in dialysis subjects, there is no evidence of increased harm, and thus consideration of lipid lowering medications in particular individuals with ESRD is warranted.

Use of Statins in Renal Transplant Recipients

The Assessment of Lescol in Renal Transplant (ALERT) study randomized 2102 renal transplant recipients to fluvastatin or placebo. There was a non-significant 17% reduction in the combined primary endpoint (cardiac mortality, nonfatal myocardial infarction or coronary intervention procedures) but a significant reduction in cardiac death or myocardial infarction (43, 44). Furthermore, a post hoc analysis suggested that earlier initiation of statins post-transplant was associated with greater benefit (45). However, a recent small study found no benefit of statin therapy on coronary calcification in renal transplant patients (46). Furthermore, as with pre-end stage CKD patients there did not appear to be any benefit from statin therapy on progression of renal disease or graft loss in statin treated transplant recipients (47). Thus, following renal transplant patients should be considered for statin therapy for CVD risk reduction, but not for graft preservation. Several of the statins have drug interactions, particularly with cyclosporine, thus providers must be aware of dose and drug restrictions (Table 3).

Table 2. Use of Statins in Various CKD Subgroups

Patient population

Statin indicated? Yes/no

Microalbuminuria*

No

CKD 1-4

Yes

Nephrotic syndrome

Unclear

Dialysis patients

No

Renal transplant recipients

Yes

* In the absence of any other indication

EVIDENCE FOR/AGAINST LIPID LOWERING THERAPY IN CKD FOR RENAL OUTCOMES

Given the evidence that renal lipid deposition is associated with progression of renal disease itself, there has been an ongoing interest in whether targeting dyslipidemia in CKD can help delay the progression of the renal disease. The dyslipidemia in CKD is associated not only with increased CVD but also with adverse renal prognosis (48, 49). Biopsy studies have found that the amount of renal apoB/apoE is correlated with increased progression of the renal disease itself (50). Animal studies have supported this concept. A meta-analysis of several small, older studies suggested that the rate of decline in GFR was decreased in subjects receiving a lipid-lowering agent (the included studies mainly used statins but the meta-analysis also included a study using gemfibrozil and another using probucol) (51). However, the relationship between lipid levels and renal disease is unclear, as prospective cohort studies have not found any relationship of lipid levels to progression of kidney disease (52). Furthermore, the SHARP study, which included subjects with earlier stages of CKD (stages 3-5 were included) found no benefit of lipid lowering therapy on the progression of renal disease. A meta-analysis of statins in pre-end stage CKD patients found no overall effect of statins on renal disease progression (31) and the ALERT study found no benefit of statin use on renal graft or renal disease parameters (47). Thus, there does not appear to be any use for statins to improve renal function or CKD itself.

SAFETY OF STATINS IN CKD

Statin Safety– Renal Outcomes

An observational study using administrative databases containing information on > 2 million patients suggested that the use of high potency statins was associated with acute kidney injury, especially within the first 120 days of statin use (53). However, a subsequent analysis of 24 placebo-controlled statin studies and 2 high versus low-dose statin studies found no evidence of renal injury from statin use (54). These discrepant results can be explained by the quality of the data: in randomized controlled trials, albeit not designed or powered to look at renal injury, data quality tends to be higher than that in administrative data sets, which often contain bias for selection, ascertainment, and classification. Furthermore, statins appear to have a nephron-protective role in the prevention of contrast induced acute kidney injury. A meta-analysis of 15 trials examining the effect of statin pre-treatment before coronary angiography found a significant reduction in acute kidney injury in those treated with high dose statin compared to controls treated with either placebo or low dose statin (55). One study specifically examined the use of statins in subjects with diabetes and existing CKD undergoing angiography, and found a benefit to statin pre-treatment in reducing the risk of contrast induced acute renal injury (56). As discussed above, use of statins in pre-end stage CKD or post-renal transplant patients demonstrates neither benefit nor harm on renal outcomes. Thus, based on available evidence there does not seem to be any renal harm from statin use, and the presence of CKD should not be a contra-indication to statin use, although some statins require dose restrictions in CKD (Table 3).

Statin Safety – Diabetes Outcomes

As a class, emerging evidence demonstrates that statins increase new diagnoses of diabetes (57). As diabetes can lead to or exacerbate renal injury, this is another potential harm of statins. However, there is no evidence that statin therapy acutely raises normal fasting glucose into the diabetic range and rather the evidence from clinical trials suggests that statin therapy instead leads individuals at high risk of diabetes to progress to diabetes diagnosis sooner than may have happened without statin therapy. A subsequent meta-analysis of 5 statin trials with >32,000 patients without diabetes at baseline found that high dose statin was associated with increased risk for new diabetes diagnosis compared to low or moderate dose statin therapy (58). However, the number needed to harm (induce diabetes) is 498 whereas the number needed to treat (prevent cardiovascular events) is 155 for intensive statin therapy; implying that despite the increased risk of new onset diabetes, statin therapy’s benefits outweigh the risks.

Which Statins to Use in CKD?

The various statins have different degrees of renal clearance; thus, with CKD patients it is important to be aware of the metabolism of the agent of interest and understand if/when dose adjustments are needed. Most statins are primarily metabolized through hepatic pathways, and dose adjustment in early CKD is typically not needed (eGFR> 30 ml/min). However, with more advanced CKD, eGFR< 30 ml/min (or ESRD, although statins are not indicated in this population) most agents have maximum dose restrictions (Table 3). 

Table 3. Statin Dosing in CKD

Statin

Usual dose range (mg/d)

Clearance route

Dose range for CKD stages1-3

Dose range for CKD stages4-5

Use with cyclosporine

Atorvastatin

10-80

Liver

10-80

10-80

Avoid use with cyclosporine

Fluvastatin

20-80

Liver

20-80

20-40

Max dose 20 mg/d with cyclosporine

Lovastatin

10-80

Liver

10-80

10-20

Avoid use with cyclosporine

Pitavastatin

1-4

Liver/Kidney

1-2

1-2

Avoid use with cyclosporine

Pravastatin

10-80

Liver/Kidney

10-80

10-20

Max dose 20 mg/d when used with cyclosporine

Rosuvastatin

10-40

Liver/Kidney

5-40

5-10

Max dose 5 mg/d with cyclosporine

Simvastatin

5-40

Liver

5-40

5-40

Avoid use with cyclosporine

BEYOND STATINS

There has been relatively little research into the use of non-statin lipid lowering agents in CKD. There is an emerging interest in niacin in CKD patients for its phosphorus-lowering effects, and niacin has similar lipid-altering efficacy in CKD as opposed to non-CKD subjects. Fibrates are metabolized via the kidney and thus generally contraindicated in CKD. Ezetimibe has been shown to be safe and effective in reducing LDL for patients with CKD; however, studies have typically compared treatment with ezetimibe added to statin therapy vs control and few studies compare ezetimibe monotherapy vs control.  PCSK9-inhibitors have been shown to be safe in CKD and efficacious in lowering LDL but there remains limited data regarding morbidity and mortality outcomes with this therapy.  Newer therapies include bempedoic acid and inclisiran which both remain relatively unstudied in CKD/ESRD. The following sections summarize the available data on the use of other lipid lowering agents in CKD (Table 4).

Niacin

As niacin is not cleared via the kidney it is theoretically safe in CKD; however, its use is limited due to side effects (predominantly flushing) and a lack of data. Several short-term studies have evaluated niacin in CKD patients and it is efficacious in lipid lowering. There is an emerging interest in use of niacin or its analog niacinimide in CKD and ESRD patients for their effects to decrease phosphate levels. A meta-analysis of randomized controlled trials of niacin and niacinamide in dialysis patients found that niacin reduced serum phosphorus but did not change serum calcium levels; furthermore niacin increased HDL levels but had no significant effect on LDL-cholesterol, triglycerides, or total cholesterol levels; no CVD outcomes data were provided (59). Animal studies have suggested a beneficial effect of niacin on renal outcomes (60), and clinical literature is suggestive that this may occur in humans (61). Kang et al treated patients with CKD stages 2-4 with niacin 500mg/d x 6 months; niacin led to increased HDL-cholesterol and decreased triglyceride levels, and improved GFR compared to baseline levels (62). Laropiprant has been developed as an inhibitor of prostaglandin-medicated niacin-induced flushing. In a sub-study examining the use of niacin with laropiprant in dyslipidemic subjects with impaired renal function, the use of niacin resulted in a mean decrease in serum phosphorus of 11% with similar effects between those with eGFR above or below 60 ml/min/1.73 m2 (63); the parent study reported significant reduction in lipid parameters including a decrease in LDL-cholesterol of 18%, decrease in triglycerides of 25%, and an increase in HDL of 20% (64). Thus, there may be an indication for use of niacin in CKD subjects beyond lipid lowering considerations. However, cardiovascular outcome studies evaluating the combination of statin plus niacin have not found any additional benefit compared to statin alone (65, 66); thus, at this time further research is needed in CKD subjects to determine if niacin may be more beneficial than statins, or if the addition of niacin to statin may confer non-CVD benefit, for example, from phosphorus lowering.

Fibrates

Fibric acid derivatives are used primarily to raise HDL-cholesterol and lower triglycerides; thus, they target two major components of CKD associated dyslipidemia. However, fibrates are known to decrease renal blood flow and glomerular filtration and they are cleared renally (67); therefore, there is significant concern regarding their use in CKD. Furthermore, the fibric acid derivatives raise serum creatinine levels and may thus trigger medical investigations into renal disease progression. Thus, there is concern regarding their use in CKD. However, there is a potential for fibric acid derivatives to improve both CVD and CKD outcomes. The acute changes in serum creatinine do not necessarily indicate adverse renal effects. A meta-analysis (68)  examined the use of fibrates in CKD subjects and reported beneficial effects to reduce total cholesterol and triglyceride levels and raise HDL-cholesterol levels with no effect on LDL-cholesterol levels. In addition, 3 trials reporting on > 14,000 patients reported that fibrates reduced risk of albuminuria progression in diabetic subjects, with 2 trials (>2,000 patients) reporting albuminuria regression (69-71). This was associated with a reduction in major cardiovascular events, CVD death, stroke, and all-cause mortality in subjects with moderate renal dysfunction, but not in those with eGFR > 60 ml/min/1.73m2.  Thus, despite the elevations in serum creatinine seen with fibrates, there is the potential for both cardiac and renal benefit, and further studies specifically designed to evaluate these outcomes in CKD subjects are urgently needed. At this point, providers are encouraged to consider fibrate therapy for appropriate subjects, especially if statins are not tolerated or are contra-indicated.

Ezetimibe

Ezetimibe is presently the only member of the class of cholesterol absorption inhibitors. As monotherapy it can lower LDL approximately 15%; however, the majority of research has examined ezetimibe in combination with a statin (primarily simvastatin) where the addition of ezetimibe can induce a further 25% lowering of LDL cholesterol. Ezetimibe is metabolized through intestinal and hepatic metabolism, and does not require any dose adjustment in CKD or ESRD, making it potentially attractive therapy in CKD. The Improved Reduction of Outcomes: Vytorin Efficacy International Trial (IMPROVE IT) study demonstrated that the combination of statin + ezetimibe led to further LDL lowering and improved CVD outcomes compared to statin alone in high-risk patients (72).  A secondary analysis of this study evaluating outcomes based on eGFR shows that compared to statin alone, the combination of statin + ezetimibe was more effective in reducing risk of CVD outcomes in those with eGFR < 60/ml/min/1.73m2(73). The Study of Heart and Renal Protection (SHARP) compared CVD and renal effects in CKD patients treated with statin + ezetimibe versus placebo. There was a reduction in CVD events (39); however, there was no effect on renal disease progression (74). Note, neither of these studies included an ezetimibe only arm; thus, the effects of ezetimibe monotherapy on outcomes are unknown, although it can be expected to reduce CVD events in proportion to its degree of LDL-cholesterol lowering. A small study evaluating ezetimibe monotherapy in CKD patients found it safe and effective (75). Thus, the use of ezetimibe with or without statin is likely to benefit pre-end stage CKD patients in terms of CVD outcomes (given that the impact of ezetimibe is on lowering LDL-cholesterol we can anticipate lack of CVD benefit in ESRD subjects based on the statin studies and SHARP).

Fish Oil

Omega-3 polyunsaturated fatty acids can lower triglyceride levels, making them a potential therapy in CKD. The role of fish oil/ omega-3 supplements in the general population for prevention of CVD events remains unclear, with some studies suggesting benefit but others finding no CVD protection. A recent meta-analysis found no evidence for CVD protection (76) while a meta-analysis of thirteen randomized control trials involving 127,477 patients demonstrated marine omega-3 supplementation was associated with small but significantly lower risk of MI, CHD death, total CHD, CVD death and total CVD with linear relationship to dose (77).  In CKD patients there is little data to support the use of fish oil and much of the data it is conflicting. A small randomized study evaluated omega-3 fish oil supplements, coenzyme Q10, or both in subjects with CKD stage 3 for 8 weeks. The group that received the omega-3 supplements had decreased heart rate and blood pressure and triglycerides, but there was no effect on renal function (eGFR, or albuminuria) (78). Conversely, a study evaluating dietary omega-3 intake found that higher consumption was associated with reduced likelihood of CKD (79). A randomized controlled trial in patients with CKD and microalbuminuria showed that omega-3 fatty acid supplementation had no effect on urine albumin excretion; however, there was a beneficial effect on serum triglyceride levels and pulse wave velocity (80). Fish oil supplementation has not been found to have any clear benefit on hemodialysis arteriovenous graft function (81, 82)or on cardiovascular events or mortality in hemodialysis patients (83). Thus, there is no clear benefit to the use of fish oil supplements in CKD, but further research is needed.

Bile Acid Resins

The bile acid resins tend to be used less commonly than other classes of lipid lowering agents overall, and their use in CKD is limited by a lack of data. Bile acid resins as a class can lower LDL-cholesterol by 10-20% so they are less effective than statins; furthermore, they can raise triglyceride levels and their use is contra-indicated with elevated triglyceride levels, for example > 400-500 mg/dl (>4.5 – 5.6 mmol/L). Thus, overall bile acid resins are rarely used in CKD patients. However, their metabolism is intestinal and thus there are no required modifications for their use in mild-moderate CKD. Although there are no theoretical concerns regarding their use in ESRD there is no data to address safety or efficacy.

PCSK9 Inhibitors

Monoclonal antibodies against proprotein convertase subtilisin/kexin type 9 (PCSK9) have been developed and approved for patients for patients with clinical atherosclerotic CVD not meeting lipid goals despite maximally tolerated statin therapy. This class of drugs lowers LDL-C in addition to statin-mediated lowering and has been shown to decrease CVD events in outcome studies in secondary prevention populations (84). Two PCSK9 monoclonal antibody inhibitors are presently available in the US – evolucumab and alirocumab. PCSK9 plasma levels are not influenced by eGFR in CKD patients (85) but are increased in nephrotic syndrome (86). As monoclonal antibodies the inhibitors are not cleared by the kidney and thus are approved to use in CKD and ESRD with no dose adjustment. The ODYSSEY OUTCOME trial randomized post-acute coronary syndrome patients with LDL > 70mg/dL to maximally tolerated statin with placebo vs alirocumab; the intervention arm with alirocumab had nearly twice the absolute reduction in cardiovascular events (87). Of note patients with eGFR < 30 ml/min/m2 were excluded from the ODYSSEY OUTCOME trial. However, a later subanalysis looked at the effect of alirocumab on major adverse cardiovascular events based on renal function. The subanalysis showed that irrespective of eGFR alirocumab was efficacious in reducing LDL. Further, annualized incidence rates of major adverse cardiovascular events and death increased with decreasing eGFR but rates were lower in the alirocumab group compared to placebo and there was no significant difference in incidence of major adverse cardiovascular events between treatment groups with eGFR < 60 ml/min/m2 (88).  Further, data from a pooled analysis of nine trials comparing alirocumab vs control showed that among patients with ASCVD and LDL > 100 mg/dL those with additional risk factors including CKD had the greatest absolute cardiovascular benefit from alirocumab therapy in addition to maximally tolerated statin compared to placebo (89).  Studies remain ongoing to further look at mortality and morbidity outcome in PCSK-9 inhibitors specifically in at risk patients such as those with CKD. There remains very limited data to use in patients with ESRD and PCSK-9 inhibitor use as monotherapy for dyslipidemia.

Bempedoic Acid

Currently approved for use in combination with maximally tolerated statin therapy, bempedoic acid facilitates further LDL reduction by inhibiting cholesterol synthesis in the liver through blocking adenosine triphosphate-citrate lyase (ACL).  Currently, use in CKD is approved without dosage adjustment for eGFR > 30ml/minute/1.73m2; however, below this eGFR threshold there is insufficient data to guide its use. As bempedoic acid has hepatic metabolism it is presumably safe in CKD.  A 52-week study in very high-risk CVD patients demonstrated that bempedoic acid added to maximally tolerated statin therapy was safe and led to a significant reduction in LDL levels (90).  Further, combination with ezetimibe is safe and can increase the cholesterol-lowering effect more than either agent alone when added to standard therapy (91). A cardiovascular outcome study is presently underway (92) but at this time there are limited data regarding mortality and morbidity benefit and use in ESRD.

Inclisiran

Newest to the market, inclisiran is a small interfering RNA (siRNA) that acts in hepatocytes to break down mRNA for PCSK-9 which increases LDL cholesterol receptor recycling thus increasing LDL cholesterol uptake. It is FDA approved for use in heterozygous familial hypercholesterolemia and in secondary prevention of cardiovascular events as an adjunct to lifestyle and maximally tolerated statin. It is administered by subcutaneous injections at 3 and then 6-month intervals. There are no cardiovascular outcomes studies yet available. There is no recommended dosage adjustment in CKD, but there have been no studies done in patients with ESRD. An analysis of the ORION-1 and ORION-7 studies compared inclisiran in patients with renal impairment and those with normal renal function, and found similar safety and efficacy, suggesting no dose adjustment is needed in CKD (93). However, no patients on dialysis were studied in these trials.

Table 4. Non-Statin Treatments

Agent

Usual dose range (mg/d)

Clearance route

Dose range for CKD stages1-3

Dose range for CKD stages4-5

Use with cyclosporine

Niaspan

500-2000

Hepatic/renal

No data

No data

No data

Gemfibrozil

1200

Renal

Avoid if creatinine > 2.0 mg/dl

Avoid if creatinine > 2.0 mg/dl

Cautious use

Fenofibrate

40-200

renal

40-60

avoid

Cautious use

Ezetimibe

10

Intestinal/hepatic

10

10

Cautious use

Colsevelam

3750 (6 x 625 mg tablets daily)

Intestinal

No change

unknown

May reduce levels of cyclosporine

Fish oil

4000

 

No change

Caution

No data

PCSK9 inhibitors

Alirocumab 75-150mg SC q 2 weeks

Evolocumab 140mg weekly SC - 420mg monthly SC

Unknown

No change

Not defined

No data

Bempedoic acid

180 mg daily

Hepatic

No change

Not defined

No data

Inclisiran

284 mg SC at 0 and 3 months then every 6 months

Nucleases

No change

Not defined

No data

SUMMARY

CVD is the leading cause of mortality in CKD, and as with the non-CKD population dyslipidemia is a significant contributor. The dyslipidemia of CKD comprises primarily high triglyceride levels and low HDL-cholesterol levels; however, emerging data suggests that the composition of the lipoprotein particles is altered by CKD, and that altered composition and/or lipoprotein cargo may be a cause of the increased CVD in CKD. The use of statins has been shown to be safe and efficacious in lipid lowering in CKD, and of benefit in reducing CVD events in individuals with pre-end stage CKD, or post renal transplant, but not in dialysis patients. The various available agents have different clearance routes, and some statins need dose adjustment in CKD. In patients that cannot tolerate or who have contra-indications to statin therapy, there may be some benefit from use of PCSK9 inhibitors, fibrates niacin or newer therapies such as bempedoic acid and inclisiran, but further studies are needed to better investigate their use.

REFERENCES

  1. National Kidney Foundation. 2012. KDOQI Clinical Practice Guideline for Diabetes and CKD: 2012 Update. Am J Kidney Dis 60: 850-886.
  2. Collins, A. J., R. N. Foley, B. Chavers, D. Gilbertson, C. Herzog, K. Johansen, B. Kasiske, N. Kutner, J. Liu, W. St Peter, H. Guo, S. Gustafson, B. Heubner, K. Lamb, S. Li, Y. Peng, Y. Qiu, T. Roberts, M. Skeans, J. Snyder, C. Solid, B. Thompson, C. Wang, E. Weinhandl, D. Zaun, C. Arko, S. C. Chen, F. Daniels, J. Ebben, E. Frazier, C. Hanzlik, R. Johnson, D. Sheets, X. Wang, B. Forrest, E. Constantini, S. Everson, P. Eggers, and L. Agodoa. 2012. 'United States Renal Data System 2011 Annual Data Report: Atlas of chronic kidney disease & end-stage renal disease in the United States. Am J Kidney Dis 59: A7, e1-420.
  3. Candan, C., N. Canpolat, S. Gokalp, N. Yildiz, P. Turhan, M. Tasdemir, L. Sever, and S. Caliskan. 2014. Subclinical cardiovascular disease and its association with risk factors in children with steroid-resistant nephrotic syndrome. Pediatric nephrology 29: 95-102.
  4. Foley, R. N., P. S. Parfrey, and M. J. Sarnak. 1998. Clinical epidemiology of cardiovascular disease in chronic renal disease. Am J Kidney Dis 32: S112-119.
  5. Parfrey, P. S., and R. N. Foley. 1999. The clinical epidemiology of cardiac disease in chronic renal failure. J Am Soc Nephrol 10: 1606-1615.
  6. Sarnak, M. J., B. E. Coronado, T. Greene, S. R. Wang, J. W. Kusek, G. J. Beck, and A. S. Levey. 2002. Cardiovascular disease risk factors in chronic renal insufficiency. Clin Nephrol 57: 327-335.
  7. Chronic Kidney Disease Prognosis, C., K. Matsushita, M. van der Velde, B. C. Astor, M. Woodward, A. S. Levey, P. E. de Jong, J. Coresh, and R. T. Gansevoort. 2010. Association of estimated glomerular filtration rate and albuminuria with all-cause and cardiovascular mortality in general population cohorts: a collaborative meta-analysis. Lancet 375: 2073-2081.
  8. van der Velde, M., K. Matsushita, J. Coresh, B. C. Astor, M. Woodward, A. Levey, P. de Jong, R. T. Gansevoort, C. Chronic Kidney Disease Prognosis, M. van der Velde, K. Matsushita, J. Coresh, B. C. Astor, M. Woodward, A. S. Levey, P. E. de Jong, R. T. Gansevoort, A. Levey, M. El-Nahas, K. U. Eckardt, B. L. Kasiske, T. Ninomiya, J. Chalmers, S. Macmahon, M. Tonelli, B. Hemmelgarn, F. Sacks, G. Curhan, A. J. Collins, S. Li, S. C. Chen, K. P. Hawaii Cohort, B. J. Lee, A. Ishani, J. Neaton, K. Svendsen, J. F. Mann, S. Yusuf, K. K. Teo, P. Gao, R. G. Nelson, W. C. Knowler, H. J. Bilo, H. Joosten, N. Kleefstra, K. H. Groenier, P. Auguste, K. Veldhuis, Y. Wang, L. Camarata, B. Thomas, and T. Manley. 2011. Lower estimated glomerular filtration rate and higher albuminuria are associated with all-cause and cardiovascular mortality. A collaborative meta-analysis of high-risk population cohorts. Kidney Int 79: 1341-1352.
  9. Chan, D. T., G. K. Dogra, A. B. Irish, E. M. Ooi, P. H. Barrett, D. C. Chan, and G. F. Watts. 2009. Chronic kidney disease delays VLDL-apoB-100 particle catabolism: potential role of apolipoprotein C-III. J Lipid Res 50: 2524-2531.
  10. Vaziri, N. D., G. Deng, and K. Liang. 1999. Hepatic HDL receptor, SR-B1 and Apo A-I expression in chronic renal failure. Nephrol Dial Transplant 14: 1462-1466.
  11. Schuchardt, M., M. Tolle, and M. van der Giet. 2015. High-density lipoprotein: structural and functional changes under uremic conditions and the therapeutic consequences. Handbook of experimental pharmacology 224: 423-453.
  12. Kuznik, A., J. Mardekian, and L. Tarasenko. 2013. Evaluation of cardiovascular disease burden and therapeutic goal attainment in US adults with chronic kidney disease: an analysis of national health and nutritional examination survey data, 2001-2010. BMC nephrology 14: 132.
  13. Longenecker, J. C., J. Coresh, N. R. Powe, A. S. Levey, N. E. Fink, A. Martin, and M. J. Klag. 2002. Traditional cardiovascular disease risk factors in dialysis patients compared with the general population: the CHOICE Study. J Am Soc Nephrol 13: 1918-1927.
  14. Pennell, P., B. Leclercq, M. I. Delahunty, and B. A. Walters. 2006. The utility of non-HDL in managing dyslipidemia of stage 5 chronic kidney disease. Clin Nephrol 66: 336-347.
  15. Rao, R., D. Ansell, J. A. Gilg, S. J. Davies, E. J. Lamb, and C. R. Tomson. 2009. Effect of change in renal replacement therapy modality on laboratory variables: a cohort study from the UK Renal Registry. Nephrol Dial Transplant 24: 2877-2882.
  16. Chu, M., A. Y. Wang, I. H. Chan, S. H. Chui, and C. W. Lam. 2012. Serum small-dense LDL abnormalities in chronic renal disease patients. British journal of biomedical science 69: 99-102.
  17. Reis, A., A. Rudnitskaya, P. Chariyavilaskul, N. Dhaun, V. Melville, J. Goddard, D. J. Webb, A. R. Pitt, and C. M. Spickett. 2014. Top-down lipidomics of low density lipoprotein reveal altered lipid profiles in advanced chronic kidney disease. J Lipid Res.
  18. Hricik, D. E., J. T. Mayes, and J. A. Schulak. 1991. Independent effects of cyclosporine and prednisone on posttransplant hypercholesterolemia. Am J Kidney Dis 18: 353-358.
  19. Marcen, R., J. Chahin, A. Alarcon, and J. Bravo. 2006. Conversion from cyclosporine microemulsion to tacrolimus in stable kidney transplant patients with hypercholesterolemia is related to an improvement in cardiovascular risk profile: a prospective study. Transplantation proceedings 38: 2427-2430.
  20. Ma, K. L., X. Z. Ruan, S. H. Powis, Y. Chen, J. F. Moorhead, and Z. Varghese. 2007. Sirolimus modifies cholesterol homeostasis in hepatic cells: a potential molecular mechanism for sirolimus-associated dyslipidemia. Transplantation 84: 1029-1036.
  21. Vaziri, N. D. 2003. Molecular mechanisms of lipid disorders in nephrotic syndrome. Kidney Int 63: 1964-1976.
  22. Vaziri, N. D., T. Sato, and K. Liang. 2003. Molecular mechanisms of altered cholesterol metabolism in rats with spontaneous focal glomerulosclerosis. Kidney Int 63: 1756-1763.
  23. Vaziri, N. D., K. Liang, and J. S. Parks. 2001. Acquired lecithin-cholesterol acyltransferase deficiency in nephrotic syndrome. Am J Physiol Renal Physiol 280: F823-828.
  24. Vaziri, N. D., C. H. Kim, D. Phan, S. Kim, and K. Liang. 2004. Up-regulation of hepatic Acyl CoA: Diacylglycerol acyltransferase-1 (DGAT-1) expression in nephrotic syndrome. Kidney Int 66: 262-267.
  25. Pearson, G. J., G. Thanassoulis, T. J. Anderson, A. R. Barry, P. Couture, N. Dayan, G. A. Francis, J. Genest, J. Gregoire, S. A. Grover, M. Gupta, R. A. Hegele, D. Lau, L. A. Leiter, A. A. Leung, E. Lonn, G. B. J. Mancini, P. Manjoo, R. McPherson, D. Ngui, M. E. Piche, P. Poirier, J. Sievenpiper, J. Stone, R. Ward, and W. Wray. 2021. 2021 Canadian Cardiovascular Society Guidelines for the Management of Dyslipidemia for the Prevention of Cardiovascular Disease in Adults. The Canadian journal of cardiology 37: 1129-1150.
  26. Grundy, S. M., N. J. Stone, A. L. Bailey, C. Beam, K. K. Birtcher, R. S. Blumenthal, L. T. Braun, S. de Ferranti, J. Faiella-Tommasino, D. E. Forman, R. Goldberg, P. A. Heidenreich, M. A. Hlatky, D. W. Jones, D. Lloyd-Jones, N. Lopez-Pajares, C. E. Ndumele, C. E. Orringer, C. A. Peralta, J. J. Saseen, S. C. Smith, Jr., L. Sperling, S. S. Virani, and J. Yeboah. 2019. 2018 AHA/ACC/AACVPR/AAPA/ABC/ACPM/ADA/AGS/APhA/ASPC/NLA/PCNA Guideline on the Management of Blood Cholesterol: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. J Am Coll Cardiol 73: e285-e350.
  27. Mefford, M. T., R. S. Rosenson, L. Deng, R. M. Tanner, V. Bittner, M. M. Safford, B. Coll, K. E. Mues, K. L. Monda, and P. Muntner. 2019. Trends in Statin Use Among US Adults With Chronic Kidney Disease, 1999-2014. Journal of the American Heart Association 8: e010640.
  28. 2002. MRC/BHF Heart Protection Study of cholesterol lowering with simvastatin in 20,536 high-risk individuals: a randomised placebo-controlled trial. Lancet 360: 7-22.
  29. Tonelli, M., C. Isles, G. C. Curhan, A. Tonkin, M. A. Pfeffer, J. Shepherd, F. M. Sacks, C. Furberg, S. M. Cobbe, J. Simes, T. Craven, and M. West. 2004. Effect of pravastatin on cardiovascular events in people with chronic kidney disease. Circulation 110: 1557-1563.
  30. Soohoo, M., H. Moradi, Y. Obi, C. M. Rhee, E. O. Gosmanova, M. Z. Molnar, M. L. Kashyap, D. L. Gillen, C. P. Kovesdy, K. Kalantar-Zadeh, and E. Streja. 2019. Statin Therapy Before Transition to End-Stage Renal Disease With Posttransition Outcomes. Journal of the American Heart Association 8: e011869.
  31. Palmer, S. C., S. D. Navaneethan, J. C. Craig, D. W. Johnson, V. Perkovic, J. Hegbrant, and G. F. Strippoli. 2014. HMG CoA reductase inhibitors (statins) for people with chronic kidney disease not requiring dialysis. Cochrane Database Syst Rev 5: CD007784.
  32. Toto, R. D., S. M. Grundy, and G. L. Vega. 2000. Pravastatin treatment of very low density, intermediate density and low density lipoproteins in hypercholesterolemia and combined hyperlipidemia secondary to the nephrotic syndrome. Am J Nephrol 20: 12-17.
  33. Matzkies, F. K., U. Bahner, M. Teschner, H. Hohage, A. Heidland, and R. M. Schaefer. 1999. Efficiency of 1-year treatment with fluvastatin in hyperlipidemic patients with nephrotic syndrome. Am J Nephrol 19: 492-494.
  34. Asselbergs, F. W., G. F. Diercks, H. L. Hillege, A. J. van Boven, W. M. Janssen, A. A. Voors, D. de Zeeuw, P. E. de Jong, D. J. van Veldhuisen, W. H. van Gilst, R. Prevention of, and I. Vascular Endstage Disease Intervention Trial. 2004. Effects of fosinopril and pravastatin on cardiovascular events in subjects with microalbuminuria. Circulation 110: 2809-2816.
  35. Scheven, L., M. Van der Velde, H. J. Lambers Heerspink, P. E. De Jong, and R. T. Gansevoort. 2013. Isolated microalbuminuria indicates a poor medical prognosis. Nephrol Dial Transplant 28: 1794-1801.
  36. Wanner, C., V. Krane, W. Marz, M. Olschewski, J. F. Mann, G. Ruf, and E. Ritz. 2005. Atorvastatin in patients with type 2 diabetes mellitus undergoing hemodialysis. N Engl J Med 353: 238-248.
  37. Krane, V., K. R. Schmidt, L. J. Gutjahr-Lengsfeld, J. F. Mann, W. Marz, F. Swoboda, C. Wanner, and D. S. Investigators. 2016. Long-term effects following 4 years of randomized treatment with atorvastatin in patients with type 2 diabetes mellitus on hemodialysis. Kidney Int 89: 1380-1387.
  38. Fellstrom, B. C., A. G. Jardine, R. E. Schmieder, H. Holdaas, K. Bannister, J. Beutler, D. W. Chae, A. Chevaile, S. M. Cobbe, C. Gronhagen-Riska, J. J. De Lima, R. Lins, G. Mayer, A. W. McMahon, H. H. Parving, G. Remuzzi, O. Samuelsson, S. Sonkodi, D. Sci, G. Suleymanlar, D. Tsakiris, V. Tesar, V. Todorov, A. Wiecek, R. P. Wuthrich, M. Gottlow, E. Johnsson, and F. Zannad. 2009. Rosuvastatin and cardiovascular events in patients undergoing hemodialysis. N Engl J Med 360: 1395-1407.
  39. Baigent, C., M. J. Landray, C. Reith, J. Emberson, D. C. Wheeler, C. Tomson, C. Wanner, V. Krane, A. Cass, J. Craig, B. Neal, L. Jiang, L. S. Hooi, A. Levin, L. Agodoa, M. Gaziano, B. Kasiske, R. Walker, Z. A. Massy, B. Feldt-Rasmussen, U. Krairittichai, V. Ophascharoensuk, B. Fellstrom, H. Holdaas, V. Tesar, A. Wiecek, D. Grobbee, D. de Zeeuw, C. Gronhagen-Riska, T. Dasgupta, D. Lewis, W. Herrington, M. Mafham, W. Majoni, K. Wallendszus, R. Grimm, T. Pedersen, J. Tobert, J. Armitage, A. Baxter, C. Bray, Y. Chen, Z. Chen, M. Hill, C. Knott, S. Parish, D. Simpson, P. Sleight, A. Young, and R. Collins. 2011. The effects of lowering LDL cholesterol with simvastatin plus ezetimibe in patients with chronic kidney disease (Study of Heart and Renal Protection): a randomised placebo-controlled trial. Lancet 377: 2181-2192.
  40. Palmer, S. C., S. D. Navaneethan, J. C. Craig, D. W. Johnson, V. Perkovic, S. U. Nigwekar, J. Hegbrant, and G. F. Strippoli. 2013. HMG CoA reductase inhibitors (statins) for dialysis patients. Cochrane Database Syst Rev 9: CD004289.
  41. Marz, W., B. Genser, C. Drechsler, V. Krane, T. B. Grammer, E. Ritz, T. Stojakovic, H. Scharnagl, K. Winkler, I. Holme, H. Holdaas, C. Wanner, D. German, and I. Dialysis Study. 2011. Atorvastatin and low-density lipoprotein cholesterol in type 2 diabetes mellitus patients on hemodialysis. Clin J Am Soc Nephrol 6: 1316-1325.
  42. Vaziri, N. D., and K. C. Norris. 2013. Reasons for the lack of salutary effects of cholesterol-lowering interventions in end-stage renal disease populations. Blood purification 35: 31-36.
  43. Holdaas, H., B. Fellstrom, A. G. Jardine, I. Holme, G. Nyberg, P. Fauchald, C. Gronhagen-Riska, S. Madsen, H. H. Neumayer, E. Cole, B. Maes, P. Ambuhl, A. G. Olsson, A. Hartmann, D. O. Solbu, and T. R. Pedersen. 2003. Effect of fluvastatin on cardiac outcomes in renal transplant recipients: a multicentre, randomised, placebo-controlled trial. Lancet 361: 2024-2031.
  44. Jardine, A. G., H. Holdaas, B. Fellstrom, E. Cole, G. Nyberg, C. Gronhagen-Riska, S. Madsen, H. H. Neumayer, B. Maes, P. Ambuhl, A. G. Olsson, I. Holme, P. Fauchald, C. Gimpelwicz, T. R. Pedersen, and A. S. Investigators. 2004. fluvastatin prevents cardiac death and myocardial infarction in renal transplant recipients: post-hoc subgroup analyses of the ALERT Study. Am J Transplant 4: 988-995.
  45. Holdaas, H., B. Fellstrom, A. G. Jardine, G. Nyberg, C. Gronhagen-Riska, S. Madsen, H. H. Neumayer, E. Cole, B. Maes, P. Ambuhl, J. O. Logan, B. Staffler, C. Gimpelewicz, and A. S. Group. 2005. Beneficial effect of early initiation of lipid-lowering therapy following renal transplantation. Nephrol Dial Transplant 20: 974-980.
  46. Yazbek, D. C., A. B. de Carvalho, C. S. Barros, J. O. Medina Pestana, and M. E. Canziani. 2016. Effect of Statins on the Progression of Coronary Calcification in Kidney Transplant Recipients. PLoS One 11: e0151797.
  47. Fellstrom, B., H. Holdaas, A. G. Jardine, I. Holme, G. Nyberg, P. Fauchald, C. Gronhagen-Riska, S. Madsen, H. H. Neumayer, E. Cole, B. Maes, P. Ambuhl, A. G. Olsson, A. Hartmann, J. O. Logan, T. R. Pedersen, and I. Assessment of Lescol in Renal Transplantation Study. 2004. Effect of fluvastatin on renal end points in the Assessment of Lescol in Renal Transplant (ALERT) trial. Kidney Int 66: 1549-1555.
  48. Samuelsson, O., H. Mulec, C. Knight-Gibson, P. O. Attman, B. Kron, R. Larsson, L. Weiss, H. Wedel, and P. Alaupovic. 1997. Lipoprotein abnormalities are associated with increased rate of progression of human chronic renal insufficiency. Nephrol Dial Transplant 12: 1908-1915.
  49. Samuelsson, O., P. O. Attman, C. Knight-Gibson, R. Larsson, H. Mulec, L. Weiss, and P. Alaupovic. 1998. Complex apolipoprotein B-containing lipoprotein particles are associated with a higher rate of progression of human chronic renal insufficiency. J Am Soc Nephrol 9: 1482-1488.
  50. Sato, H., S. Suzuki, H. Kobayashi, S. Ogino, A. Inomata, and M. Arakawa. 1991. Immunohistological localization of apolipoproteins in the glomeruli in renal disease: specifically apoB and apoE. Clin Nephrol 36: 127-133.
  51. Fried, L. F., T. J. Orchard, and B. L. Kasiske. 2001. Effect of lipid reduction on the progression of renal disease: a meta-analysis. Kidney Int 59: 260-269.
  52. Rahman, M., W. Yang, S. Akkina, A. Alper, A. H. Anderson, L. J. Appel, J. He, D. S. Raj, J. Schelling, L. Strauss, V. Teal, D. J. Rader, and C. S. Investigators. 2014. Relation of serum lipids and lipoproteins with progression of CKD: The CRIC study. Clin J Am Soc Nephrol 9: 1190-1198.
  53. Dormuth, C. R., B. R. Hemmelgarn, J. M. Paterson, M. T. James, G. F. Teare, C. B. Raymond, J. P. Lafrance, A. Levy, A. X. Garg, P. Ernst, and S. Canadian Network for Observational Drug Effect. 2013. Use of high potency statins and rates of admission for acute kidney injury: multicenter, retrospective observational analysis of administrative databases. BMJ 346: f880.
  54. Bangalore, S., R. Fayyad, G. K. Hovingh, R. Laskey, L. Vogt, D. A. DeMicco, D. D. Waters, C. Treating to New Targets Steering, and Investigators. 2014. Statin and the risk of renal-related serious adverse events: Analysis from the IDEAL, TNT, CARDS, ASPEN, SPARCL, and other placebo-controlled trials. Am J Cardiol 113: 2018-2020.
  55. Lee, J. M., J. Park, K. H. Jeon, J. H. Jung, S. E. Lee, J. K. Han, H. L. Kim, H. M. Yang, K. W. Park, H. J. Kang, B. K. Koo, S. H. Jo, and H. S. Kim. 2014. Efficacy of short-term high-dose statin pretreatment in prevention of contrast-induced acute kidney injury: updated study-level meta-analysis of 13 randomized controlled trials. PLoS One 9: e111397.
  56. Han, Y., G. Zhu, L. Han, F. Hou, W. Huang, H. Liu, J. Gan, T. Jiang, X. Li, W. Wang, S. Ding, S. Jia, W. Shen, D. Wang, L. Sun, J. Qiu, X. Wang, Y. Li, J. Deng, J. Li, K. Xu, B. Xu, R. Mehran, and Y. Huo. 2014. Short-term rosuvastatin therapy for prevention of contrast-induced acute kidney injury in patients with diabetes and chronic kidney disease. J Am Coll Cardiol 63: 62-70.
  57. Sattar, N., D. Preiss, H. M. Murray, P. Welsh, B. M. Buckley, A. J. de Craen, S. R. Seshasai, J. J. McMurray, D. J. Freeman, J. W. Jukema, P. W. Macfarlane, C. J. Packard, D. J. Stott, R. G. Westendorp, J. Shepherd, B. R. Davis, S. L. Pressel, R. Marchioli, R. M. Marfisi, A. P. Maggioni, L. Tavazzi, G. Tognoni, J. Kjekshus, T. R. Pedersen, T. J. Cook, A. M. Gotto, M. B. Clearfield, J. R. Downs, H. Nakamura, Y. Ohashi, K. Mizuno, K. K. Ray, and I. Ford. 2010. Statins and risk of incident diabetes: a collaborative meta-analysis of randomised statin trials. Lancet 375: 735-742.
  58. Preiss, D., S. R. Seshasai, P. Welsh, S. A. Murphy, J. E. Ho, D. D. Waters, D. A. DeMicco, P. Barter, C. P. Cannon, M. S. Sabatine, E. Braunwald, J. J. Kastelein, J. A. de Lemos, M. A. Blazing, T. R. Pedersen, M. J. Tikkanen, N. Sattar, and K. K. Ray. 2011. Risk of incident diabetes with intensive-dose compared with moderate-dose statin therapy: a meta-analysis. JAMA 305: 2556-2564.
  59. He, Y. M., L. Feng, D. M. Huo, Z. H. Yang, and Y. H. Liao. 2014. Benefits and harm of niacin and its analog for renal dialysis patients: a systematic review and meta-analysis. Int Urol Nephrol 46: 433-442.
  60. Cho, K. H., H. J. Kim, V. S. Kamanna, and N. D. Vaziri. 2010. Niacin improves renal lipid metabolism and slows progression in chronic kidney disease. Biochim Biophys Acta 1800: 6-15.
  61. Streja, E., C. P. Kovesdy, D. A. Streja, H. Moradi, K. Kalantar-Zadeh, and M. L. Kashyap. 2015. Niacin and progression of CKD. Am J Kidney Dis 65: 785-798.
  62. Kang, H. L., D. Y. Kim, S. M. Lee, K. H. Kim, S. H. Han, H. K. Nam, K. H. Kim, S. E. Kim, Y. K. Son, and W. S. An. 2013. Effect of low-dose niacin on dyslipidemia, serum phophorus levels and adverse effects in patients with chornic kidney disease. Kidney Res Clin Pract 32: 21-26.
  63. Maccubbin, D., D. Tipping, O. Kuznetsova, W. A. Hanlon, and A. G. Bostom. 2010. Hypophosphatemic effect of niacin in patients without renal failure: a randomized trial. Clin J Am Soc Nephrol 5: 582-589.
  64. Maccubbin, D., H. E. Bays, A. G. Olsson, V. Elinoff, A. Elis, Y. Mitchel, W. Sirah, A. Betteridge, R. Reyes, Q. Yu, O. Kuznetsova, C. M. Sisk, R. C. Pasternak, and J. F. Paolini. 2008. Lipid-modifying efficacy and tolerability of extended-release niacin/laropiprant in patients with primary hypercholesterolaemia or mixed dyslipidaemia. International journal of clinical practice 62: 1959-1970.
  65. Boden, W. E., J. L. Probstfield, T. Anderson, B. R. Chaitman, P. Desvignes-Nickens, K. Koprowicz, R. McBride, K. Teo, and W. Weintraub. 2011. Niacin in patients with low HDL cholesterol levels receiving intensive statin therapy. N Engl J Med 365: 2255-2267.
  66. HPS2-THRIVE Collaborative Group, M. J. Landray, R. Haynes, J. C. Hopewell, S. Parish, T. Aung, J. Tomson, K. Wallendszus, M. Craig, L. Jiang, R. Collins, and J. Armitage. 2014. Effects of extended-release niacin with laropiprant in high-risk patients. N Engl J Med 371: 203-212.
  67. Sica, D. A. 2009. Fibrate therapy and renal function. Curr Atheroscler Rep 11: 338-342.
  68. Jun, M., B. Zhu, M. Tonelli, M. J. Jardine, A. Patel, B. Neal, T. Liyanage, A. Keech, A. Cass, and V. Perkovic. 2012. Effects of fibrates in kidney disease: a systematic review and meta-analysis. J Am Coll Cardiol 60: 2061-2071.
  69. Davis, T. M., R. Ting, J. D. Best, M. W. Donoghoe, P. L. Drury, D. R. Sullivan, A. J. Jenkins, R. L. O'Connell, M. J. Whiting, P. P. Glasziou, R. J. Simes, Y. A. Kesaniemi, V. J. Gebski, R. S. Scott, A. C. Keech, I. Fenofibrate, and i. Event Lowering in Diabetes Study. 2011. Effects of fenofibrate on renal function in patients with type 2 diabetes mellitus: the Fenofibrate Intervention and Event Lowering in Diabetes (FIELD) Study. Diabetologia 54: 280-290.
  70. Tonelli, M., D. Collins, S. Robins, H. Bloomfield, and G. C. Curhan. 2004. Gemfibrozil for secondary prevention of cardiovascular events in mild to moderate chronic renal insufficiency. Kidney Int 66: 1123-1130.
  71. Ting, R. D., A. C. Keech, P. L. Drury, M. W. Donoghoe, J. Hedley, A. J. Jenkins, T. M. Davis, S. Lehto, D. Celermajer, R. J. Simes, K. Rajamani, and K. Stanton. 2012. Benefits and safety of long-term fenofibrate therapy in people with type 2 diabetes and renal impairment: the FIELD Study. Diabetes Care 35: 218-225.
  72. Cannon, C. P., M. A. Blazing, R. P. Giugliano, A. McCagg, J. A. White, P. Theroux, H. Darius, B. S. Lewis, T. O. Ophuis, J. W. Jukema, G. M. De Ferrari, W. Ruzyllo, P. De Lucca, K. Im, E. A. Bohula, C. Reist, S. D. Wiviott, A. M. Tershakovec, T. A. Musliner, E. Braunwald, R. M. Califf, and IMPROVE-IT Investigators. 2015. Ezetimibe Added to Statin Therapy after Acute Coronary Syndromes. N Engl J Med 372: 2387-2397.
  73. Stanifer, J. W., D. M. Charytan, J. White, Y. Lokhnygina, C. P. Cannon, M. T. Roe, and M. A. Blazing. 2017. Benefit of Ezetimibe Added to Simvastatin in Reduced Kidney Function. J Am Soc Nephrol 28: 3034-3043.
  74. Haynes, R., D. Lewis, J. Emberson, C. Reith, L. Agodoa, A. Cass, J. C. Craig, D. de Zeeuw, B. Feldt-Rasmussen, B. Fellstrom, A. Levin, D. C. Wheeler, R. Walker, W. G. Herrington, C. Baigent, M. J. Landray, S. C. Group, and S. C. Group. 2014. Effects of lowering LDL cholesterol on progression of kidney disease. J Am Soc Nephrol 25: 1825-1833.
  75. Morita, T., S. Morimoto, C. Nakano, R. Kubo, Y. Okuno, M. Seo, K. Someya, M. Nakahigashi, H. Ueda, N. Toyoda, M. Kusabe, F. Jo, N. Takahashi, T. Iwasaka, and I. Shiojima. 2014. Renal and vascular protective effects of ezetimibe in chronic kidney disease. Internal medicine 53: 307-314.
  76. Rizos, E. C., E. E. Ntzani, E. Bika, M. S. Kostapanos, and M. S. Elisaf. 2012. Association between omega-3 fatty acid supplementation and risk of major cardiovascular disease events: a systematic review and meta-analysis. JAMA 308: 1024-1033.
  77. Hu, Y., F. B. Hu, and J. E. Manson. 2019. Marine Omega-3 Supplementation and Cardiovascular Disease: An Updated Meta-Analysis of 13 Randomized Controlled Trials Involving 127 477 Participants. Journal of the American Heart Association 8: e013543.
  78. Mori, T. A., V. Burke, I. Puddey, A. Irish, C. A. Cowpland, L. Beilin, G. Dogra, and G. F. Watts. 2009. The effects of [omega]3 fatty acids and coenzyme Q10 on blood pressure and heart rate in chronic kidney disease: a randomized controlled trial. J Hypertens 27: 1863-1872.
  79. Gopinath, B., D. C. Harris, V. M. Flood, G. Burlutsky, and P. Mitchell. 2011. Consumption of long-chain n-3 PUFA, alpha-linolenic acid and fish is associated with the prevalence of chronic kidney disease. The British journal of nutrition 105: 1361-1368.
  80. Bunout, D., G. Barrera, S. Hirsch, and E. Lorca. 2021. A Randomized, Double-Blind, Placebo-Controlled Clinical Trial of an Omega-3 Fatty Acid Supplement in Patients With Predialysis Chronic Kidney Disease. J Ren Nutr 31: 64-72.
  81. Lok, C. E., L. Moist, B. R. Hemmelgarn, M. Tonelli, M. A. Vazquez, M. Dorval, M. Oliver, S. Donnelly, M. Allon, and K. Stanley. 2012. Effect of fish oil supplementation on graft patency and cardiovascular events among patients with new synthetic arteriovenous hemodialysis grafts: a randomized controlled trial. Jama 307: 1809-1816.
  82. Irish, A. B., A. K. Viecelli, C. M. Hawley, L. S. Hooi, E. M. Pascoe, P. A. Paul-Brent, S. V. Badve, T. A. Mori, A. Cass, P. G. Kerr, D. Voss, L. M. Ong, K. R. Polkinghorne, A. Omega-3 Fatty, and G. Aspirin in Vascular Access Outcomes in Renal Disease Study Collaborative. 2017. Effect of Fish Oil Supplementation and Aspirin Use on Arteriovenous Fistula Failure in Patients Requiring Hemodialysis: A Randomized Clinical Trial. JAMA Intern Med 177: 184-193.
  83. Svensson, M., E. B. Schmidt, K. A. Jorgensen, and J. H. Christensen. 2006. N-3 fatty acids as secondary prevention against cardiovascular events in patients who undergo chronic hemodialysis: a randomized, placebo-controlled intervention trial. Clin J Am Soc Nephrol 1: 780-786.
  84. Sabatine, M. S., R. P. Giugliano, A. C. Keech, N. Honarpour, S. D. Wiviott, S. A. Murphy, J. F. Kuder, H. Wang, T. Liu, S. M. Wasserman, P. S. Sever, T. R. Pedersen, F. S. Committee, and Investigators. 2017. Evolocumab and Clinical Outcomes in Patients with Cardiovascular Disease. N Engl J Med 376: 1713-1722.
  85. Morena, M., C. Le May, L. Chenine, L. Arnaud, A. M. Dupuy, M. Pichelin, H. Leray-Moragues, L. Chalabi, B. Canaud, J. P. Cristol, and B. Cariou. 2017. Plasma PCSK9 concentrations during the course of nondiabetic chronic kidney disease: Relationship with glomerular filtration rate and lipid metabolism. J Clin Lipidol 11: 87-93.
  86. Pavlakou, P., E. Liberopoulos, E. Dounousi, and M. Elisaf. 2017. PCSK9 in chronic kidney disease. Int Urol Nephrol 49: 1015-1024.
  87. Schwartz, G. G., P. G. Steg, M. Szarek, D. L. Bhatt, V. A. Bittner, R. Diaz, J. M. Edelberg, S. G. Goodman, C. Hanotin, R. A. Harrington, J. W. Jukema, G. Lecorps, K. W. Mahaffey, A. Moryusef, R. Pordy, K. Quintero, M. T. Roe, W. J. Sasiela, J. F. Tamby, P. Tricoci, H. D. White, A. M. Zeiher, O. O. Committees, and Investigators. 2018. Alirocumab and Cardiovascular Outcomes after Acute Coronary Syndrome. N Engl J Med 379: 2097-2107.
  88. Tunon, J., P. G. Steg, D. L. Bhatt, V. A. Bittner, R. Diaz, S. G. Goodman, J. W. Jukema, Y. U. Kim, Q. H. Li, C. Mueller, A. Parkhomenko, R. Pordy, P. Sritara, M. Szarek, H. D. White, A. M. Zeiher, G. G. Schwartz, and O. O. Investigators. 2020. Effect of alirocumab on major adverse cardiovascular events according to renal function in patients with a recent acute coronary syndrome: prespecified analysis from the ODYSSEY OUTCOMES randomized clinical trial. Eur Heart J 41: 4114-4123.
  89. Vallejo-Vaz, A. J., K. K. Ray, H. N. Ginsberg, M. H. Davidson, R. H. Eckel, L. V. Lee, L. Bessac, R. Pordy, A. Letierce, and C. P. Cannon. 2019. Associations between lower levels of low-density lipoprotein cholesterol and cardiovascular events in very high-risk patients: Pooled analysis of nine ODYSSEY trials of alirocumab versus control. Atherosclerosis 288: 85-93.
  90. Ray, K. K., H. E. Bays, A. L. Catapano, N. D. Lalwani, L. T. Bloedon, L. R. Sterling, P. L. Robinson, C. M. Ballantyne, and C. H. Trial. 2019. Safety and Efficacy of Bempedoic Acid to Reduce LDL Cholesterol. N Engl J Med 380: 1022-1032.
  91. Powell, J., and C. Piszczatoski. 2021. Bempedoic Acid: A New Tool in the Battle Against Hyperlipidemia. Clin Ther 43: 410-420.
  92. Nicholls, S., A. M. Lincoff, H. E. Bays, L. Cho, D. E. Grobbee, J. J. Kastelein, P. Libby, P. M. Moriarty, J. Plutzky, K. K. Ray, P. D. Thompson, W. Sasiela, D. Mason, J. McCluskey, D. Davey, K. Wolski, and S. E. Nissen. 2021. Rationale and design of the CLEAR-outcomes trial: Evaluating the effect of bempedoic acid on cardiovascular events in patients with statin intolerance. Am Heart J 235: 104-112.
  93. Wright, R. S., M. G. Collins, R. M. Stoekenbroek, R. Robson, P. L. J. Wijngaard, U. Landmesser, L. A. Leiter, J. J. P. Kastelein, K. K. Ray, and D. Kallend. 2020. Effects of Renal Impairment on the Pharmacokinetics, Efficacy, and Safety of Inclisiran: An Analysis of the ORION-7 and ORION-1 Studies. Mayo Clin Proc 95: 77-89.

 

Fibrocalculous Pancreatic Diabetes

ABSTRACT

 

In tropical countries like India, there are several reports of a unique form of diabetes called fibrocalculous pancreatic diabetes (FCPD). In general, FCPD occurs in young, lean individuals with diabetes, abdominal pain, and steatorrhea. People with FCPD are at a higher risk of developing pancreatic cancer. Diabetes is typically ketosis resistant. Recent studies have shown that a proportion of the cases may have genetic factors and gene mutations that confer the risk of developing the disease. Recent studies have suggested a changing profile of the disease which could also be present in older individuals having a normal body mass index and better survival. The changing profile of the illness could be due to better exocrine, and endocrine (diabetes-related) care being offered to people with FCPD. The treatment profile of exocrine illnesses in FCPD is like conventional therapy of chronic pancreatitis. Hyperglycemia in FCPD is managed by a combination of lifestyle intervention, monitoring, and medications. Researchers and clinicians working with people having FCPD must, in addition to good medical care, focus on prevention and early diagnosis of future illness like pancreatic cancer as well as diabetes-related vascular complications.

 

INTRODUCTION

 

It is well known that diabetes in tropical and developing countries is different from that seen in the Western World. For example, people with diabetes in India are leaner and develop diabetes earlier (1). In earlier times, this leanness was attributable to factors like malnutrition and pancreatic damage (2). However, with increasing improvement in nutritional status in India, malnutrition has become less frequent. Thus, leanness in people with type 2 diabetes in India is now increasingly attributed to the “thin-fat” Indian concept, i.e., the combination of visceral adiposity in thinly built persons (3). In addition, some people with lean type diabetes in India could have a latent autoimmune diabetes of the adult (4).

 

However, there was another subtype of diabetes seen with leanness that required better characterization. This form of diabetes, typically occurring in young people, with a low body mass index (BMI), and with the presence of clinical features of malnutrition, was unique because of structural changes in the pancreas, associated with pancreatic damage and presence of calculi in the pancreas. This form of diabetes is referred to as fibrocalcific pancreatic diabetes or fibrocalculous pancreatic diabetes (FCPD) (5-8). Before the onset of diabetes, there is a pre-diabetic phase of pancreatic damage in these subjects, which is referred to as TCP or tropical chronic pancreatitis, a name suggesting its occurrence in tropical countries such as India. A large clinical study of chronic pancreatitis in India documented that the occurrence of TCP and FCPD is seen in about 3.5% of subjects with chronic pancreatitis (9).

 

This chapter discusses the unique nature of FCPD as well as glycemic aspects of managing FCPD, recognizing its overlap with other forms of pancreatic diabetes in India, focusing on the changing clinical spectrum of these disorders.

 

EPIDEMIOLOGY AND HISTORICAL ASPECTS

 

It was in 1959, that Zudeima reported a series of cases of pancreatic calculi with features of undernutrition, particularly in lower socioeconomic groups (10). This was followed by reports from India, Brazil, Thailand and other countries (11,12). Importantly it was the disease-characterization work of Dr Geevarghese that the disease became known worldwide (13). Over the next decades, several states of India have reported and characterized FCPD. However, it was after a national prospective multicenter study, that the currently accepted version of FCPD and the changing profile of the disease (see later) became well established, i.e., that FCPD could overlap with idiopathic pancreatitis, and that with better treatment, leanness and malnutrition are no longer characteristic of chronic pancreatitis (9,14). Indeed, there is evidence to suggest that people with chronic pancreatitis now live longer, and people who develop diabetes go on to develop micro- and macrovascular complications of the disease.  Following this nationwide study, other reports have documented the changing profile of FCPD over the years (15).

 

Earlier, it was reported that FCPD is an important cause of diabetes in the young (16). However, more recent reports indicate that this scenario is changing and that FCPD accounts for only a small proportion of people with young-onset diabetes (17). It is also well known that hyperglycemia leads to microvascular complications, and because of this, people with FCPD are prone to microvascular complications like neuropathy, nephropathy, and retinopathy. While macrovascular complications are thought to be rare in FCPD, they have been reported.

 

PATHOGENESIS

 

The exact pathogenesis of FCPD is not known. Studies have focused on environmental toxins, oxidant stress, micronutrient deficiency, dietary toxins, autoimmune factors, and a combination of the above. Genetic analysis has shown variants in the SPINK gene in 33% of people with FCPD, according to a study by Hassan et al. (18,19). The authors concluded that the N34S variant of the SPINK gene poses a susceptibility to FCPD in the Indian subcontinent.

 

One of the favored theories of FCPD was the consumption of cassava. However, in one study, the long-term feeding of cassava, up to one year, in a rat model failed to detect features of FCPD (20). This questioned the cassava theory. Malnutrition has also been favored as a cause of FCPD, but recent reports clearly indicate that malnutrition is the consequence rather than the cause of FCPD.

 

A disbalance between the oxidative stress and antioxidant responses in the body have also been postulated in FCPD. Higher levels of malondialdehyde and reduced levels of antioxidant markers have been reported. A heightened oxidative detoxification reaction has been reported in FCPD (21).

 

Earlier, genetic studies had focused on familial aggregations as well as on HLA associations, while recent studies highlight altered expression of several genes in FCPD, notably: serum protease inhibitor Kazal type 1 (SPINK1), cationic trypsinogen (PRSS1), anionic trypsinogen (PRSS2), and chymotrypsinogen C. Taken together, most studies suggest that FCPD is of heterogenous origin, and may be due to a gene-environment interaction (22). Clearly, no single factor can explain FCPD, simply because no single factor is sufficient for the disease phenotype. For instance, a genetic change may render one susceptible to pancreatic damage, and this may be heightened by oxidative stress due to micronutrient injury.

 

Table 1. Pathophysiological Factors Postulated in the Etio-Pathogenesis of FCPD

Genetic:

   Familial aggregations

   Altered gene expressions involving several genes:

     Serum protease inhibitor Kazal type 1 (SPINK1)- especially N34S mutation of SPINK-1

     Cationic trypsinogen (PRSS1)

     Anionic trypsinogen (PRSS2)

     Chymotrypsinogen C

Environmental:

   Toxins

   Micronutrient deficiency

   Cassava consumption

   Oxidative stress

   Malnutrition

Additional:

   A two-hit model (Mahurkar et al,):

      First hit: mutation of one or more genes resulting in the formation of supertrypsin in

                    the acinar cells of pancreas.                    

      Second hit: probably involves unidentified genes resulting in one or more of

                         phenotypes like stone formation, fibrosis and/ or diabetes.                                                                                                                      

 

 

PATHOGENESIS OF DIABETES IN FCPD

 

The pathogenesis of diabetes in FCPD deserves a special section. It has been reported that insulin secretion, as well as stimulated insulin levels after a glucose tolerance test are reduced in people with FCPD levels as compared to controls, but insulin levels were numerically higher than in people with type 1 diabetes, suggesting some preservation of beta cell function. Clinically, the absence of occurrence of ketoacidosis following withdrawal of insulin suggests that there is some pancreatic beta cell reserve. However, rarely ketoacidosis has been reported (23). Studies have also suggested that an exocrine function marker could correlate with endocrine dysfunction. While it is simple and logical to presume that any pancreatic pathology would reduce insulin production, the situation is more complex than meets the eye. There is evidence of insulin resistance (IR) in people with FCPD. Mean glucose disposal rates and markers of IR such as homeostasis model assessment (HOMA) IR have suggested that IR could be a factor in FCPD (6). A deficiency of pancreatic polypeptide (PP) has been postulated to underlie IR in FCPD, particularly at the level of the liver (6). The liver seems to be a particular site of insulin resistance in FCPD, because defects in the internalization of liver glucose transporter-2 (GLUT-2) as well as altered bioavailability/ function of the hepatic insulin receptor has been invoked to play a role in FCPD (6). Recent reports have suggested that a reduced fat store in people with FCPD may lead to the storage of triglyceride in the liver, and that this could predispose to insulin resistance (6).

 

CLINICAL PRESENTATION

 

FCPD has been classically described in young, lean BMI, malnourished people from tropical countries with a low socioeconomic background. There is evidence to suggest that this classical presentation is changing, and there is better longevity too (24). The disease is increasingly affecting people with a normal BMI as well as subjects of an older age group. Classical descriptions such as a cyanotic hue, a distended abdomen, and parotid gland swellings are no longer seen (5).

 

The usual age of occurrence is 10-30 years, but FCPD is often seen in younger as well as older individuals. The first symptom is abdominal pain. This pain is usually seen in the upper abdomen and is relieved by stooping forward or lying in a prone position. The pain radiates to the back- and these descriptions of pain are evocative of pancreatitis-like pain. The pain lessens in frequency and severity with passage of time and this decrease coincides with a rise in blood glucose and eventual diabetes. A sign of exocrine pancreatic insufficiency is that the stools become oily and frothy, and this signals fat malabsorption. Rarely, frank steatorrhea may be present (5).

 

Diabetes presents one to two decades after the onset of abdominal pain (26). Despite hyperglycemia, episodes of diabetic ketoacidosis do not commonly occur- this is attributed to partial beta cell destruction with some insulin secretory activity present, reduced glucagon reserve because of alpha cell destruction, and finally, reduced availability of non-esterified fatty acids which are a substrate for ketogenesis. The reduced availability of non-esterified fatty acids is due to the lack of subcutaneous fat. While diabetes can be managed with lifestyle changes and oral drugs in a few cases, insulin is required in many cases as the subjects are often lean and malnourished. Ketoacidosis is rare despite withdrawal of insulin, but may occur. Studies have shown that stimulated insulin secretion is low in people with FCPD and certain recent reports have also suggested that insulin resistance does occur in FCPD. This is an area for more research.

 

The clinical profile of FCPD seems to be changing, and previously accepted criteria may no longer be applicable. In a nationwide prospective study from India, by our group, it was shown that FCPD accounted for only 3.2% of chronic pancreatitis (9). However, in this study, very strict criteria, including a low BMI were used. If the BMI criteria were removed, then virtually all chronic pancreatitis with diabetes in tropical countries like India, which is of “idiopathic” i.e., non-alcoholic, could be labelled as FCPD. Clearly, this represents a large number, and geography alone cannot determine a disease. Hence, we call for development of newer criteria for FCPD.  This nationwide prospective study was also the first to show the changing profile and suggested that people with FCPD may overlap with idiopathic chronic pancreatitis (9).

 

With appropriate diabetes care and good pancreatic enzyme supplementation, malnutrition is rare, as is death due to diabetes in adolescence. Probably, like in chronic pancreatitis, people with FCPD could also now have a BMI that is relatively higher, i.e., not lean. Tropical chronic pancreatitis is the pre-diabetes phase of the disease, and once diabetes begins, the disease is referred to as FCPD (9). Not all people with chronic pancreatitis have diabetes. About 40% of people with chronic pancreatitis have diabetes, and a female gender and alcohol intake seem to predispose to diabetes in the background of chronic pancreatitis (9). Studies suggest that people with FCPD, probably owing to longer survival, are at risk of micro-and macrovascular diabetes-related complications. More research is needed to characterize FCPD beyond simple clinical criteria, and a biological approach may yield alternatives. Thus, in addition to the typical, young and malnourished patient with FCPD, it may be important to consider an “extended spectrum” of FCPD, with people being older, higher BMI, and at risk of diabetes vascular complications also being considered in the spectrum.

 

NATURAL HISTORY OF FCPD

 

The natural history of FCPD has undergone a shift since earlier days. In the classic descriptions of those early days, FCPD was considered to present with abdominal pain in childhood, diabetes in adolescence and death in early adulthood. However, unlike the reports of early mortality in the past, now, it is well known that people with FCPD now live longer, with 80% living beyond 35 years since the onset of abdominal pain. Microvascular complications of diabetes are common in FCPD, as was shown in a publication by Jyotsna et al. Neuropathy especially is very common, reportedly seen in 43% of people with FCPD (7). Macrovascular complications of diabetes were thought to be rare in FCPD because of relative youth, lower cholesterol levels, and leanness. However, this no longer seems to be the case, as macrovascular diseases like stroke and peripheral vascular diseases have been reported, probably due to longer survival (5,6). Pancreatic adenocarcinoma and diabetes-related nephropathy are important causes of death in people with FCPD. The main causes of death include diabetic complications especially nephropathy, infections and pancreatic cancer.

 

DIAGNOSIS AND WORK UP OF FCPD

 

 FCPD is an important, though less common cause of diabetes in the young (25). FCPD should be suspected in young, lean people from tropical countries, who have a history of abdominal pain and steatorrhea, if they have features suggesting malnutrition along with diabetes – and if the diabetes is ketosis resistant (26). Also, given the changing profile of the disease, the extended spectrum mentioned above, the disease may now also be seen in older people with a lower BMI. The diagnosis of FCPD is made by the presence of diabetes mellitus plus evidence of abnormal pancreatic morphology on x ray or ultrasound. The presence of large and discrete pancreatic stones is a classical feature of the diagnosis of FCPD (11). Sometimes, however, these typical calculi are absent and therefore computerized tomography (CT), magnetic resonance imaging (MRI), or endoscopic retrograde cholangiopancreatography (ERCP) may be required to clinch the diagnosis. (25,26) The presence of large intraductal calculi, and the presence of fibrosis / ductal changes such as dilatation are all characteristic features.

Figure 1. X-ray of the abdomen showing multiple tiny opacities in epigastric region, suggestive of pancreatic calcification.

Figure 2. Ultrasound of abdomen showing atrophic pancreas, with dilated main pancreatic duct and multiple calculi in main pancreatic duct.

Figure 3. CT scan of abdomen showing atrophic pancreas with few parenchymal foci of calcification. Main pancreatic duct is dilated along with hyper dense calculi.

Figure 4. MRI scan of abdomen (axial -T2 weight HASTE sequence) showing atrophic pancreas with dilated MPD and few filling defects within, suggestive of calculi.

Figure 5. MRI scan of abdomen (coronal - T2 weight HASTE sequence): Atrophic pancreas with dilated MPD and few filling defects within, suggestive of calculi.

Figure 6. Maximum Intensity Projection (MIP) Image showing irregular dilatation of MPD with calculi within.

Conventionally, the disease is thought to occur only in tropical countries; however currently, there is no evidence to link any geographical factor with the etiology of the disease. Typical clinical features of exocrine pancreatic dysfunction such as abdominal discomfort and steatorrhea may also be present (see above). The absence of a detectable secondary cause of chronic pancreatitis is also important. The presence of exocrine pancreatic dysfunction can be determined by several tests. Older tests like the secretin pancreozymin test, in which pancreatic juice is extracted after stimulation with secretin and pancreozymin, shows a reduction in volume, bicarbonate, lipase, and trypsin content. Fecal chymotrypsin assessments are also useful tests. Fecal elastase assay is of practical significance. Endocrine tests like the measurement of C-peptide rarely have a role in diagnosis of FCPD, though they might be of clinical significance in management, when a low C-peptide level signal the need for insulin therapy.  Particularly, CA 19.9 testing is important in FCPD for the early diagnosis of pancreatic cancer. Workup should include a gastroenterological and an endocrinological consultation- and the latter should focus on tests such as glycosylated hemoglobin, renal function, lipid profile testing, urine albumin creatinine ratio to detect microalbuminuria, foot assessment, retinal screening for diabetic retinopathy and when indicated screening for cardiovascular disease by appropriate tests.

 

Table 2. Diagnostic Criteria for FCPD

1.         Occurrence in a tropical country

2.         Diabetes as per standard diagnostic criteria

3.         Evidence of chronic pancreatitis: pancreatic calculi on X-ray or at least 3 of the following: a) Abnormal pancreatic morphology by sonography

                b) Chronic abdominal pain since childhood

                c) Steatorrhea

                d) Abnormal pancreatic function test

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

 

MANAGEMENT

 

This section focuses on the management of diabetes, but before that it is necessary to mention other aspects of management.

 

Nutritional Management

 

As in other form of diabetes, medical nutrition therapy (MNT) is the cornerstone of FCPD management. As most FCPD patients are lean, calorie restriction should not be prescribed. A balanced diet with adequate carbohydrates, fat, and proteins must be ensured. A dietary consultant, to assess correlation of steatorrhea, and other gastrointestinal symptoms, with various food should help in planning appropriate meals. Foods with potential toxic effect on the pancreas, such as cassava (tapioca) should be avoided. Fat soluble vitamins must be replaced, as their absorption may be limited in FCPD. Smoking and alcohol must be stopped. The deleterious effects of substance abuse, including its effect on pancreatic adenocarcinoma, must be made very clear to the patient and the family. If available, sublingual or parenteral formulations of vitamin D should be preferred. Nano formulations of vitamin D have a hypothetical advantage in persons with FCPD.

 

Pain Management

 

Pain is a common symptom of FCPD. Pain can occur due to acute inflammation of the pancreas, increased pressure within the parenchyma and the ductal system, ischemia of the gland, microvascular complications such as neuritis, and the space-occupying effect of pseudocysts. Extra-pancreatic disorders such as bile duct or duodenal stenosis may also contribute to pain. Abdominal pain is ideally managed by non-opioid analgesic, though in later stages opioid analgesics could be required (27). Drugs such as tramadol and tapentadol can be used in low doses, but one should be mindful of the risk of addiction. Octreotide, a synthetic somatostatin analogue, has been used subcutaneously to reduce the pain of chronic pancreatitis. It inhibits pancreatic secretion and increases the contractility of the sphincter of Oddi. Micronutrition supplementation has also been thought to help in pain management by reducing oxidative stress in the pancreas. Endoscopic interventions to remove stones and lithotripsy may be helpful (28). Antioxidants are not of proven benefit in pain reduction, and pancreatic enzyme supplementation, while important from the exocrine therapy perspective, have not been shown to reduce the abdominal pain. Pancreatic enzyme replacement therapy (PERT) degrades cholecystokinin-releasing peptide in the duodenum, and facilitates inhibition of endogenous pancreatic activity and thus may help with pain reduction. Severe pain may require surgery- ductal decompression, drainage procedures like pancreatojejunostomy, and ablative procedures like subtotal pancreatectomy are advised. Coeliac blockage has also been tried as a pain-relieving intervention.

 

Exocrine Management

 

Steatorrhea responds to pancreatic enzyme supplements, which also helps improve glucose control. Pancreatic enzyme supplementation is often prescribed, at initial doses of 500-1000 lipase units/kg of body weight with each meal, up to maximum of 2,500 lipase units/kg per meal. Vitamin supplementation, particularly fat-soluble vitamin supplementation is important. A low-fat diet helps in the management of steatorrhea (8).

 

Pancreatic enzyme replacement therapy (PERT) is necessary to ensure adequate nutrient absorption and prevent down- stream effects such as protein-energy and vitamin malnutrition. It is important to prescribe an optimal dose of PERT, at the right time. Enteric coated PERT should be preferred, as they prevent the contents from being denatured by the acidic medium of the stomach, and allow them to reach the duodenum. An average dose of at least 25,000 IU (75,000 USO units) of lipase should reach the intestine for optimal fat absorption. Smaller snacks may require 10,000 IU (30,000 USP units). In practice, lesser doses (10,000-20,000IU/meal) often suffice in persons with diabetes, as they have been counseled to take low-fat meals. Ideally, patients should take half the dose with first bite of a meal, and the remaining half in the middle, or at the end. Most patients, however, prefer a reduced pill burden. To ensure adherence and persistence, therefore, the entire dose can be taken with the first bite of food. Patient should avoid ingesting the tablet with copious quantities of water, in order to prevent rapid transit through the upper gut. The adequacy of PERT replacement is determined clinically. Patient should be asked about stool consistency, presence or absence of oily droplets in the stool, and weight gain. Fecal fat and breath testing can be used, in research settings, to evaluate response to PERT.  Fecal elastase-1 is not impacted by PERT and cannot be used as a monitoring tool. PERT can be combined with proton pump inhibitors and/or histamine 2 blockers, as these increase the responsiveness to PERT by reducing acidic degradation of the formulations.

 

Diabetes Management

 

This chapter will focus on the management of diabetes. A balanced and healthy diet with whole grains and adequate micro-and macronutrient intake is important; in the presence of diabetes some degree of carbohydrate restriction is important. Restriction of simple carbohydrates can be advised. Adequate protein intake is important. In the presence of steatorrhea, a low-fat diet is important. Adequate physical activity, like brisk walking, and stress reduction are part of other lifestyle measures (8).

 

Oral anti-diabetes agents are useful only in a minority of patients with FCPD, as insulin is the anti-diabetes medication of choice. Among secretagogues, short acting drugs like repaglinide, which stimulate insulin secretion may be considered. Metformin has the benefit of glucose control without hypoglycemia. In addition, there is evidence that metformin could protect against pancreatic cancer. However, metformin may worsen gastrointestinal symptoms and lead to diarrhea and undesirable weight loss, and low doses may be used cautiously. Alfa glucosidase inhibitors are better avoided due to gastrointestinal symptoms. Incretin based drugs, including glucagon like peptide 1 agonists or dipeptidyl peptidase 4 inhibitors should not be prescribed in FCPD, given the possible link between incretin-based therapies and pancreatitis. Indeed, there is data from India that glucagon like peptide-1, an important incretin, increases two-fold in people with FCPD compared to people with type 2 diabetes and controls (29). Finally, future studies need to look at the role of thiazolidinediones and oral SGLT2 inhibitors, especially the latter, given the association between FCPD mortality and nephropathy (8).

 

Studies have shown that FCPD is associated with some degree of insulin resistance. However, it has also been noted that insulin levels are lower in FCPD compared with type 2 diabetes and in general- insulin is the drug of choice. Long-acting basal insulin and multiple doses of rapid acting insulins at mealtime are the ideal therapy for diabetes in FCPD. Glycemic targets to be attained with anti-diabetes therapy are similar to those recommended for commoner subtypes of diabetes. Control of other cardio-metabolic risk factors such as hypertension and dyslipidemia are also important. Very high triglyceride levels > 500 mg/dl put people with FCPD at risk of pancreatitis, and therefore may need additional lifestyle intervention and medications such as fibrates (8).

 

SUMMARY

 

FCPD is a unique form of diabetes often seen in the tropics. Currently, it is considered an example of a secondary, pancreatic form of diabetes- also referred to as “type 3c diabetes”. There is a need for further research into its etiology. The clinical spectrum is changing, and the classical young, malnourished subject with FCPD is now not the only clinical phenotype. Indeed, an “extended spectrum” of the clinical phenotype now includes older people with higher BMI, no malnutrition, and predilection to both exocrine and endocrine (diabetes-related) complications. Early detection of pancreatic cancer is important in these subjects, as is the diagnosis of diabetes related complications. Management includes lifestyle modifications and medications, among which insulin injections are an ideal choice for managing glycemic control in people with FCPD. One of the fascinating areas of research is the presence of insulin resistance in subjects with FCPD- which appears to be independent of intra-abdominal adipose tissue (30). Future research on the genetic and environmental aspects of FCPD causation will help uncover new therapeutic approaches, that ultimately promise to improve the lives of people with FCPD.

 

ACKOWLEDGEMENTS

 

We thank Dr. Abhishek B. Yashod, MD (Radio-Diagnosis), Chellaram Hospital: Diabetes Care & Multispecialty, Pune; Dr Aseem Katyal, Faith Ultrasound, Karnal; Dr Sangeeta Dhawan, Dhawan Ultrasound, Karnal; for providing the CT scan and MRI images.

 

REFERENCES

 

1        Unnikrishnan, R., Anjana, R. & Mohan, V. Diabetes mellitus and its complications in India. Nat Rev Endocrinol 12, 357–370 (2016). https://doi.org/10.1038/nrendo.2016.53

2        Samal KC, Kanungo A, Sanjeevi CB. Clinicoepidemiological and Biochemical Profile of Malnutrition-Modulated Diabetes Mellitus. Annals of the New York Academy of Sciences 2006; 958: 131–137.

3        Wells JCK, Pomeroy E, Walimbe SR, Popkin BM, Yajnik CS. The Elevated Susceptibility to Diabetes in India: An Evolutionary Perspective. Frontiers in Public Health 2016; https://www.frontiersin.org/article/10.3389/fpubh.2016.00145 (accessed 15 Jan 2022).

4        Unnikrishnan AG, Singh SK, Sanjeevi CB. Prevalence of GAD65 Antibodies in Lean Subjects with Type 2 Diabetes. Annals of the New York Academy of Sciences 2004; 1037: 118–121.

5        Unnikrishnan R, Mohan V. Fibrocalculous pancreatic diabetes (FCPD). Acta Diabetol 2015; 52: 1–9.

6        Dasgupta R, Naik D, Thomas N. Emerging concepts in the pathogenesis of diabetes in fibrocalculous pancreatic diabetes: J Diabetes 2015; 7: 754–761.

7        Jyotsna VP, Singh SK, Gopal D, Unnikrishnan AG, Agrawal NK, Singh SK et al. Clinical and biochemical profiles of young diabetics in North-Eastern India. J Assoc Physicians India 2002; 50: 1130–1134.

8        Kumaran S, Unnikrishnan AG. Fibrocalculous pancreatic diabetes. Journal of Diabetes and its Complications 2021; 35: 107627.

9        Balakrishnan V, Unnikrishnan AG, Thomas V, Choudhuri G, Veeraraju P, Singh SP et al. Chronic pancreatitis. A prospective nationwide study of 1,086 subjects from India. JOP 2008; 9: 593–600.

10      Zuidema PJ. Cirrhosis and disseminated calcification of the pancreas in patients with malnutrition. Trop Geogr Med 1959; 11: 70–74.

11      Barman KK, Premalatha G, Mohan V. Tropical chronic pancreatitis. Postgraduate Medical Journal 2003; 79: 606–615.

12      Dani R, Penna FJ, Nogueira CED. Etiology of chronic calcifying pancreatitis in Brazil: a report of 329 consecutive cases. International Journal of Pancreatology 1986; 1: 399–406.

13      Geevarghese PJ, Pitchumoni CS, Nair SR. Is protein malnutrition an initiating cause of pancreatic calcification? J Assoc Physicians India 1969; 17: 417–419.

14      Garg PK, Narayana D. Changing phenotype and disease behaviour of chronic pancreatitis in India: evidence for gene–environment interactions. Global Health, Epidemiology and Genomics 2016; 1. doi:10.1017/gheg.2016.13.

15      Papita R, Nazir A, Anbalagan VP, Anjana RM, Pitchumoni C, Chari S et al. Secular trends of fibrocalculous pancreatic diabetes and diabetes secondary to alcoholic chronic pancreatitis at a tertiary care diabetes centre in South India. JOP 2012; 13: 205–209.

16      Unnikrishnan R, Mohan V. Fibrocalculous Pancreatic Diabetes. Curr Diab Rep 2020; 20: 19.

17      Hassan Z, Mohan V, Ali L, Allotey R, Barakat K, Faruque MO et al. SPINK1 is a susceptibility gene for fibrocalculous pancreatic diabetes in subjects from the Indian subcontinent. Am J Hum Genet 2002; 71: 964–968.

18      Mahurkar S, Reddy DN, Rao GV, Chandak GR. Genetic mechanisms underlying the pathogenesis of tropical calcific pancreatitis. World J Gastroenterol. 2009;15(3):264-269. doi:10.3748/wjg.15.264

19      Mathangi DC, Deepa R, Mohan V, Govindarajan M, Namasivayam A. Long-Term Ingestion of Cassava (Tapioca) Does Not Produce Diabetes or Pancreatitis in the Rat Model. IJGC 2000; 27: 203–208.

20      Chaloner C, Sandle LN, Mohan V, Snehalatha C, Viswanathan M, Braganza JM. Evidence for induction of cytochrome P-450I in patients with tropical chronic pancreatitis. Int J Clin Pharmacol Ther Toxicol 1990; 28: 235–240.

21      Rossi L, Pfützer RH, Parvin S, Ali L, Sattar S, Kahn AKA et al. SPINK1/PSTI Mutations Are Associated with Tropical Pancreatitis in Bangladesh. Pancreatology 2001; 1: 242–245.

22      Melki G, Laham L, Karim G, Komal F, Kumar V, Barham S et al. Chronic Pancreatitis Leading to Pancreatogenic Diabetes Presenting in Diabetic Ketoacidosis: A Rare Entity. Gastroenterology Res 2019; 12: 208–210.

23      Mohan V, Premalatha G, Padma A, Chari ST, Pitchumoni CS. Fibrocalculous Pancreatic Diabetes: Long-term survival analysis. Diabetes Care 1996; 19: 1274–1278.

24     Unnikrishnan AG, Bhatia E, Bhatia V, Bhadada SK, Sahay RK, Kannan A, Kumaravel V, Sarma D, Ganapathy B, Thomas N, John M, Jayakumar RV, Kumar H, Nair V, Sanjeevi CB. Type 1 diabetes versus type 2 diabetes with onset in persons younger than 20 years of age. Ann N Y Acad Sci. 2008 Dec;1150:239-44. doi: 10.1196/annals.1447.056. PMID: 19120303.

25      Tandon RK, Garg PK. Tropical pancreatitis. Dig Dis. 2004;22(3):258-66. doi: 10.1159/000082797. PMID: 15753608.

26       Praveen, G., Mohan, V. Fibrocalculous pancreatic diabetes—current scenario in  

           developing countries. Int J Diabetes Dev Ctries 38, 131–132 (2018).   

           https://doi.org/10.1007/s13410-018-0638-x

27      Drewes AM, Bouwense SAW, Campbell CM, Ceyhan GO, Delhaye M, Demir IE, Garg PK, van Goor H, Halloran C, Isaji S, Neoptolemos JP, Olesen SS, Palermo T, Pasricha PJ, Sheel A, Shimosegawa T, Szigethy E, Whitcomb DC, Yadav D; Working group for the International (IAP – APA – JPS – EPC) Consensus Guidelines for Chronic Pancreatitis. Guidelines for the understanding and management of pain in chronic pancreatitis. Pancreatology. 2017 Sep-Oct;17(5):720-731. doi: 10.1016/j.pan.2017.07.006. Epub 2017 Jul 13. PMID: 28734722.

28     Kitano M, Gress TM, Garg PK, Itoi T, Irisawa A, Isayama H, Kanno A, Takase K, Levy M, Yasuda I, Lévy P, Isaji S, Fernandez-Del Castillo C, Drewes AM, Sheel ARG, Neoptolemos JP, Shimosegawa T, Boermeester M, Wilcox CM, Whitcomb DC. International consensus guidelines on interventional endoscopy in chronic pancreatitis. Recommendations from the working group for the international consensus guidelines for chronic pancreatitis in collaboration with the International Association of Pancreatology, the American Pancreatic Association, the Japan Pancreas Society, and European Pancreatic Club. Pancreatology. 2020 Sep;20(6):1045-1055. doi: 10.1016/j.pan.2020.05.022. Epub 2020 Jul 10. PMID: 32792253.

29     Ghosh I, Mukhopadhyay P, Das K, Anne M B, Ali Mondal S, Basu M, Nargis T, Pandit K, Chakrabarti P, Ghosh S. Incretins in fibrocalculous pancreatic diabetes: A unique subtype of pancreatogenic diabetes. J Diabetes. 2021 Jun;13(6):506-511. doi: 10.1111/1753-0407.13139. Epub 2020 Dec 16. PMID: 33247879.

30     Shivaprasad C, Gautham K, Palani P, Gupta S, Shah K. Intra-abdominal fat estimation by bio-electrical impedance analysis in patients with fibrocalculous pancreatic diabetes compared with BMI matched type 2 diabetic subjects and healthy controls. Diabetes Metab Syndr. 2020 Sep-Oct;14(5):789-795. doi: 10.1016/j.dsx.2020.05.038. Epub 2020 May 27. PMID: 32531743.

Immune Checkpoint Inhibitors Related Endocrine Adverse Events

ABSTRACT

 

Immune checkpoint inhibitors (ICIs) are currently used for the treatment of various types of cancers. Despite the important clinical benefits, these medications can lead to a spectrum of side effects called immune-related adverse events (irAEs). Endocrine irAEs are among the most common irAEs that have been reported in clinical trials and post-marketing settings with an overall incidence of around 10% of patients treated with ICIs. These include hypothyroidism, hyperthyroidism, hypophysitis, primary adrenal insufficiency, insulin‐deficient diabetes mellitus, hypogonadism, hypoparathyroidism, hypocalcemia, and other less commonly reported side effects. The symptoms can sometimes be nonspecific but life-threatening. Hence, physicians should be aware of the endocrine irAEs which can occur anytime during treatment or even after discontinuation of the medications. In this chapter, we will be discussing in detail the ICI-related endocrine irAEs and their management. In addition, we will be suggesting an algorithm to be used in the clinical setting for screening and monitoring of the endocrine iRAEs.

 

INTRODUCTION

 

Immune checkpoint inhibitors (ICIs) are currently approved by the US Food and Drug Administration (FDA) for the treatment of various types of cancers and have significantly improved clinical outcomes and survival. Antigen-presenting cells (APCs) process and express antigens (including tumor antigens) on major histocompatibility complexes recognized by receptors on T cells, which then stimulates a cascade either to kill the cell expressing the antigen (via CD8+ effector/cytotoxic T cells) or recruit other components of the immune system (via CD4+ helper cells) (1). Many of the ligands presented by the APCs can bind to multiple receptors and deliver stimulatory or inhibitory signals, the latter being referred to as immune checkpoints. Various ligand-receptor interactions between antigen-presenting cells and T cells regulate the T cell response to the antigen (Figure 1). Agonists of stimulatory receptors or antagonists of inhibitory signals can result in amplification of antigen-specific T-cell responses (2). Cancer cells can develop tolerance to the immune system by upregulating the expression of immune checkpoint molecules like programmed cell death ligand (PD-L1) leading to peripheral T cell exhaustion or lose surface antigen expression leading to immunologic escape. ICIs help overcoming this tolerance by inhibiting the checkpoints and these inhibitory compounds currently used in pharmacologic intervention target three ligands/receptors- CTLA-4, PD-1, and PD-L1 (3).

 

Cytotoxic T-Lymphocyte-Associated Protein 4 (CTLA-4) Inhibitors

 

CTLA-4 was first described by Leach et. al. in 1996 as a receptor on T cells (3), where it acts as a physiologic brake on the T-cell activation. It competes with the CD28 stimulatory receptor present on T cells (1). Both bind CD80 and CD86 ligands (also known as B7.1 and B7.2 respectively, collectively as B7) seen on APCs, but CTLA-4 has a 500-2500 times higher affinity for these ligands than CD28 does. Blocking CTLA-4: B7 interactions favors CD28:B7 interactions, which results in proliferation of T cells, increased T cell survival, activation of T effector cells, and increased diversity of T cell responses on tumors. This is the basis of CTLA-4 inhibitor therapy with ipilimumab (trade name Yervoy) and tremelimumab (4, 5).

 

Programmed Death-1 (PD-1) and Programmed Death-Ligand 1 (PD-L1) Inhibitors

 

PD-1 receptors on the T cell interact with PD-L1 (another member of the B7 family) and inhibit T-cell expression and decrease expression of proinflammatory cytokines such as interferon-gamma (IFN-gamma), tumor necrosis factor-alpha (TNF-alpha), and interleukin -2 (IL-2) similar to CTLA-4. PD-L1 is found on leukocytes, nonlymphoid tissue, and tumor cells and modulates CD8+ T cell function (1). PD-L1 is aberrantly expressed on many cancers, including lung, ovary, colon, head and neck, and breast (6) and results in tumor cells evading the immune system (7).  Inhibition of PD-1: PD-L1 interaction increases the number of T cells and inflammatory markers at tumor sites, creating an environment more conducive to tumor suppression. Drugs that target PD-1 include pembrolizumab (Keytruda), nivolumab (Opdivo), and dostarlimab (Jemperli) while PD-L1 inhibitors include atezolizumab (Tecentriq), avelumab (Bevancio), and Durvalumab (Imfinzi). PDL-2 is expressed on dendritic cells, monocytes, and mast cells and modulates CD4+ function.

Figure 1. Interactions between antigen-presenting cells (APCs) and T cells that regulate T-cell responses. From DM Pardoll (2)

 

Immune checkpoints normally inhibit the function of T cells, which helps prevent autoimmunity but can also benefit cancer cells. ICIs prevent the apoptosis and downregulation of T cells, which allows the immune system to naturally fight malignant cells. Despite the important clinical benefits, this unique mechanism of action itself can lead to a spectrum of side effects called immune-related adverse events (irAEs). Endocrine irAEs are among the most common irAEs that have been reported in clinical trials and post-marketing settings with a meta-analysis of 38 randomized trials showing an overall incidence of endocrinopathies among 10% of patients treated with ICIs (8). These include hypothyroidism, hyperthyroidism, hypophysitis, primary adrenal insufficiency (PAI), and insulin‐deficient diabetes mellitus. Median time to onset of moderate to severe endocrinopathy is 1.75-5 months with ipilimumab and 1.4-4.9 months for any endocrinopathy with PD-1 inhibitors (9, 10). Patients with pre-existing autoimmune disorders are at higher risk of exacerbation of the autoimmune condition as well as development of an unrelated irAEs (11). Multiple large prospective studies and meta-analyses showed that irAEs are associated with improved treatment outcomes suggesting the activated immune system is also concurrently targeting the cancer (12-14).  Hence, the general principle of management of irAEs is to control symptoms with minimum amount of immunosuppression. In this article, we will be discussing in detail the ICI-related endocrine irAEs and its management. We will be suggesting algorithm for screening, monitoring and treatment of the patients and we will be listing a summary of the side effects grading system and incidence in different ICI. (Figure 2-4, Table 3-4).

 

Immune Checkpoint Inhibitor Induced Thyroid Diseases

 

ICI-mediated thyroid disease is one of the common endocrine irAEs. It can manifest as primary hypothyroidism secondary to destructive thyroiditis or as hyperthyroidism due to Graves' disease.

 

HYPOTHYROIDISM

 

ICI-mediated hypothyroidism can present as primary or secondary hypothyroidism (secondary to hypophysitis, which is discussed below). Primary hypothyroidism usually ensues after an occurrence of ICI-induced thyrotoxicosis. In a study by Abdel-Rahman et. al., authors found a higher risk of all-grade hypothyroidism compared to hyperthyroidism associated with ICIs therapy (15).

 

Incidence 

 

The incidence of hypothyroidism with the use of immune checkpoint inhibitors varies based on the type of immune checkpoint inhibitors used and monotherapy vs combination therapy. In the largest meta-analysis of 38 randomized control trials comprising 7551 patients, the overall incidence of hypothyroidism was found to be 6.6%. The incidence of hypothyroidism ranged from 3.8% with ipilimumab to 13.2% (95% CI, 6.9%-23.8%) with combination therapy (8). Various other studies have also found similar findings of higher incidence of hypothyroidism with the use of PD-1 inhibitors (7-21%) compared to CTLA-4 inhibitor (0-6%) ipilimumab (16).

 

Pathophysiology 

 

Anti-thyroid antibodies are often absent in ICI-associated hypothyroidism, suggesting a role of cell-mediated rather than humoral autoimmunity (17). In addition, some studies have suggested an increased risk of ICI-induced thyroid dysfunction among patients with pre-existing anti-thyroid antibodies compared to those without these antibodies suggesting unmasking of autoimmune destruction with the use of ICIs (18, 19). The complete pathophysiology behind the development of thyroid dysfunction is not completely understood, but increased cytokine levels following anti-PD1 therapy have been found to correlate with thyroid dysfunction (20). Fine-needle aspiration biopsy obtained during active ICI-induced thyroiditis showed lymphocytic infiltrate along with CD163+ histiocytes (21).

 

Clinical Characteristics

 

The median time to thyroid dysfunction following initiation of ICIs is 6 weeks and most of the patients develop biochemical hypothyroidism (22). Nonetheless, thyroid dysfunction can happen at any time during therapy. Most of the patient are asymptomatic or have very few symptoms. Common presenting symptoms include fatigue, depressed mood, mild weight gain, and constipation however with severe hypothyroidism, the patient can present with altered mental status (23).

 

Screening and Monitoring

 

Thyroid function tests should be performed in all the patients receiving ICIs, by measuring TSH (thyroid stimulating hormone) and free T4 (free thyroxine). In the setting of abnormal thyroid function tests, routine monitoring is recommended at 4-6 weeks or more frequently if clinically indicated. However, in presence of normal thyroid function tests, the frequency could be increased to every 12-18 weeks. ICI-induced hypothyroidism is diagnosed by the presence of elevated TSH and decreased free T4. However, TSH is the more sensitive and preferred test. Currently, anti-thyroid antibodies have not been proven to be helpful in the screening and treatment of these patients. For patients who have subclinical hypothyroidism (elevated TSH and normal Free T4), routine monitoring is recommended while continuing treatment with immunotherapy.

 

Treatment

 

The diagnosis of primary hypothyroidism is based on elevated TSH (>10 mIU/L) and low free T4 along with clinical symptoms. Once the diagnosis is established, treatment is recommended with levothyroxine supplementation. For young patients with TSH >10 and low free T4, a full replacement dose at 1.6 mcg/kg should be considered. However, in elderly patients or among patients with cardiovascular comorbidities, a lower starting dose of 50 mcg is recommended. The dose should be changed by ~10% every 4-6 weeks to achieve reference range or age-appropriate range TSH and free T4. ICIs are usually continued while treating hypothyroidism with mild to moderate symptoms (24, 25). Although the guidelines to diagnose and treat ICI–associated primary hypothyroidism is well established, the recommendations for the management of patients with subclinical hypothyroidism (mildly elevated TSH with normal free T4) is not well established and should be based on the patient’s symptoms, age, and co-morbid conditions (26, 27).

 

THYROTOXICOSIS

 

ICI-mediated thyrotoxicosis can present as transient thyrotoxicosis or persistent hyperthyroidism. Transient thyrotoxicosis is far more common among patients treated with ICIs and is often followed by primary hypothyroidism; persistent hyperthyroidism is less frequent. Hyperthyroidism is more commonly reported with combination therapy and is rare with PD-L1 inhibitors. Patients with hyperthyroidism can be symptomatic and need supportive care with beta-blockers and anti-thyroid medications in some cases.

 

Incidence 

 

The prevalence of ICI-associated transient thyrotoxicosis has varied significantly among the studies and can range from 3.0-9.0% (23, 28) and is followed by primary hypothyroidism (8). The incidence of transient thyrotoxicosis is higher among patients treated with combination therapy compared to monotherapy with anti-PD1 or anti-PD-L1 therapy (23). In the largest to date meta-analysis, the overall incidence of hyperthyroidism was estimated to be 2.9%. The incidence of hyperthyroidism ranged from 0.6% with the PD-L1 inhibitor to 8.0% with combination therapy. Combination therapy was found to have an increased risk of higher-grade hyperthyroidism compared to monotherapy. Moreover, the risk of hyperthyroidism was greater with PD-1 inhibitors compared to PD-L1 inhibitors (8). ICI-induced Graves’ disease is extremely rare, with only a few reported cases in the literature (29).

 

Pathophysiology 

 

The pathophysiology of ICI-thyrotoxicosis remains poorly understood. Autoimmunity is believed to play a critical role in leading to thyroiditis among patients treated with ICIs. In one study, combination ICI therapy (ipilimumab and nivolumab) resulted in a more robust antibody response compared to monotherapy with nivolumab, leading to faster destruction of the thyroid (30). Moreover, patients with elevated anti-TPO or antithyroglobulin antibodies required a higher dose of levothyroxine compared to those who did not have elevated antibodies. In addition to antibody-mediated thyroid destruction, circulating CD56, CD16, and natural killer cells have been implicated in the development of pembrolizumab-induced thyroiditis in one study (17). Another study found an association between PD-L1 and PD-L2 expression on the thyroid gland and destructive thyroiditis (31). Worsening of pre-existing autoimmune thyroid disease and subclinical hypothyroidism in patients treated with ICIs have also been reported, suggesting synergistic roles of autoimmunity and inflammatory mechanisms (30).

 

Clinical Characteristics

 

Most patients with thyrotoxicosis are asymptomatic or present with symptoms such as palpitation, agitation, anxiety, and insomnia (23). Although uncommon, ICI-induced Graves' disease following use of CTLA-4-inhibitor and PD1-inhibitors have also been reported and can be associated with graves orbitopathy (23). Graves’ orbitopathy can occur with and without TRAb antibodies among patients treated with ICIs (29). ICI-induced thyroid storm is extremely rare and has only been reported a few times in the literature (32, 33). Toxic autonomous nodules or toxic multinodular goiter is not associated with ICIs and if seen among patients treated with ICIs, should be considered a co-incidental finding (23).

 

Screening and Monitoring

 

Screening of ICI-induced thyrotoxicosis is performed by TSH and free T4. Thyrotoxicosis is defined as suppressed TSH and it can either be (i) clinical when free T4 is elevated or (ii) subclinical when free T4 is normal. The most common cause of ICI-induced thyrotoxicosis is thyroiditis, which is due to the destruction of thyroid follicular cells with the release of preformed thyroid hormone. This is often associated with transient thyrotoxicosis and eventually progresses to hypothyroidism in the majority (50 to 90%) of the cases (22, 28). Hence, monitoring of TFTs with TSH and free T4 every 4-6 weeks is recommended. The usual duration of thyrotoxicosis with ICIs is about 4-6 weeks (30, 34) and if thyrotoxicosis persists beyond this period, evaluation for Graves’ disease should be considered by checking thyroid-stimulating hormone receptor antibody (TRAb) or thyroid-stimulating immunoglobulin (TSI) or a thyroid uptake scan (28)( Figure 2).

 

Treatment

 

For patients with minimal symptoms of thyroiditis-associated thyrotoxicosis, and presence of suppressed TSH and elevated free T4, supportive treatment with non-selective beta-blockers such as propranolol should be considered (24). When propranolol is used, the recommended dose is 10-20 mg every 4 to 6 hours for symptomatic management and until thyrotoxicosis resolves. As most of the time, patients with ICI-induced thyrotoxicosis progress to develop primary hypothyroidism (defined by elevated TSH levels), further treatment with thyroid hormone replacement should be considered. However, in the minority of cases (such as prominent initial symptoms, significantly elevated free T4 levels, signs of Graves’ orbitopathy, or persistent thyrotoxicosis), further evaluation and treatment for Graves’ disease should be considered (30, 34). Graves’ disease should be treated with anti-thyroid medications, radioactive iodine, or surgery depending on the clinical setting and patient preference (35). Rarely, patients can develop thyroid storm and high-dose steroids should be used in conjunction with standard management among these patients (34). If asymptomatic or only mildly symptomatic, continuation of ICIs is recommended (24, 25).

Figure 2. Algorithm suggested to diagnose and treat ICI thyroid disease.

Hypophysitis

 

Hypophysitis is one of the more common endocrine side effects reported with the use of ICIs particularly with CTLA-4 antibodies and combination therapy including both CTLA-4 and PD1 or PD-L1 inhibitors. It is less likely with PD-1 or PD-L1 inhibitor monotherapy. Hypophysitis is characterized by infiltration and inflammation of the pituitary gland. It can occur in the first few weeks of treatment with frequent hormonal deficiencies at the time of diagnosis. Pituitary enlargement is considered both a highly sensitive and specific indicator of hypophysitis after ruling out metastatic disease. Moreover, the symptoms of hypophysitis can sometimes be non-specific, hence the importance of close monitoring of these patients for early diagnosis and prompt treatment (36, 37).

 

INCIDENCE

 

Hypophysitis estimated incidence was one in nine million people per year (38). ICI-induced hypophysitis has been reported in 0-17% of ICI-treated patients. Some studies showed the incidence increased up to 25% while using higher doses of ipilimumab of 10 mg/kg (36, 37, 39). There have been some variations in the observed incidence rate of ICI-induced hypophysitis which has been attributed to not only the dose of the medication but also to the difference in the use, the intensity, and the frequency of hormonal monitoring, in addition to clinical awareness of and suspicion for the condition (37, 40).

 

Adrenocorticotropic hormone (ACTH) is one of the most common hormone deficiencies in hypophysitis. In a study of ipilimumab-induced hypophysitis, 80% had central adrenal insufficiency. Lu et. al. found hypophysitis occurred in 3.25% of patients using ICIs. Of these, it was more common with combination therapy at 7.68% followed by anti-CTLA-4 at 4.53% then anti-PD-1 and anti-PD-L1 at less than 1% of cases (41). Chang et. al. found the combination of ipilimumab (anti-CTLA-4) and nivolumab (anti-PD-1) caused hypophysitis in 6.4% of patients. Incidence of anti-CTLA-4 alone was 3.2%, anti-PD-1 alone is 0.4%, and anti-PD-L1 alone was 0.1% (42). Overall, the evidence suggests that combination therapy and anti-CTLA-4 have the highest incidence of hypophysitis, while anti-PD-1 and anti-PD-L1 are less common causes of hypophysitis.

 

PATHOPHYSIOLOGY

 

The actual pathogenesis is not well defined. Since many patients had no previous immune-related disease before the development of ICI-associated hypophysitis, it was suggested that this condition is not triggered by a pre-existing immune condition. Hypophysitis was initially considered a specific irAE of ipilimumab considering the presence of pituitary expression of CTLA-4 antigens in the TSH, follicle stimulating hormone (FSH), ACTH, and prolactin-secreting cells. Now more recent data suggested that it can occur with any ICI target (CTLA-4, PD-1, or PD-L1) (43, 44). Garon Czmin et. al. reported that the time to develop hypophysitis following initiation of ICIs was significantly shorter with ipilimumab alone or combined with nivolumab (83 days) compared to nivolumab or pembrolizumab alone (165 days). Moreover, ICI-associated hypophysitis is more common in men while autoimmune lymphocytic hypophysitis has a higher prevalence in the female population (43, 45). In one study, hypophysitis with anti-CTLA was four times more common in males compared to females. This may be related to more men having melanoma, but studies controlling for this factor have found similar results (42). Corticotrophs and thyrotrophs are the most common cell types affected while gonadotroph deficiency was more common in male patients. The somatotroph axis and prolactin levels were rarely involved (36).

 

CLINICAL CHARACTERISTICS

 

Hypophysitis can occur weeks to months after the initiation of ICIs. In the study by Albarel et. al., mean hypophysitis occurred at 9-9.5 weeks ± 6 weeks after the treatment initiation with a mean age at diagnosis of 55.2 years (36). The initial symptoms are usually related to tumor mass or hormone deficiencies, and rarely visual disturbance or diabetes insipidus. Symptoms may be acute or subacute, but they are usually nonspecific, including headaches, anorexia, dizziness, nausea, weight loss, and/ or fatigue. More serious signs are hypotension, lethargy, confusion, and electrolyte abnormalities including hyponatremia. Hyponatremia occurs due to increased antidiuretic hormone (ADH) stimulated by hypothalamic secretion of CRH. The most common hormone deficiencies include TSH, ACTH, FSH, luteinizing hormone (LH). Panhypopituitarism is less likely to happen. Although hypogonadotropic hypogonadism and central hypothyroidism may resolve, central adrenal insufficiency is permanent requiring lifelong treatment (36, 42). Although, hypo enhancing lesions in the anterior pituitary are characteristic of ICI-associated hypophysitis, a few cases of PD-1/PD-L1 induced-hypophysitis have been reported in the absence of any radiographic abnormalities and just on clinical grounds (46). This suggests that PD-1/PD-L1 may not always show classic pituitary enlargement or enhancement on MRI (47).

 

SCREENING AND MONITORING

 

The National Comprehensive Cancer Network (NCCN) guidelines recommend initial serum pituitary hormonal evaluation including morning cortisol, ACTH, TSH, FT3, LH, FSH, testosterone in men, estrogen in premenopausal women, prolactin, growth hormone, and IGF1. The sodium and potassium levels should also be checked. Cosyntropin stimulation test can be normal in acute secondary adrenal insufficiency. Diagnostics radiology reports of brain MRIs in patients receiving ICIs should routinely include comparisons of pituitary size with prior studies. In case of suspected hypophysitis, a dedicated pituitary MRI is recommended.  MRI usually showed a pituitary enlargement with or without mass effect however some cases showed pituitary adenoma, empty sella syndrome, or a normal pituitary gland on the imaging studies.

 

TREATMENT

 

Once high suspicion for ICI-induced hypophysitis, an endocrinology consult is recommended. High-dose glucocorticoids should be initiated for patients with ipilimumab-induced hypophysitis who have serious mass-effect-related symptoms, such as severe headache, visual-field disturbance, or simultaneously the presence of other irAEs. Patients should be started on methylprednisolone/prednisone at 1-2 mg/kg /day until symptoms resolve, typically 1-2 weeks then taper the steroids rapidly to a physiological dose. In patients without mass effect, studies have suggested that high dose glucocorticoid therapy was not associated with improved outcomes in patients nor change in the natural history of hypophysitis, thus physiological replacement doses can be considered in these patients (28, 36, 48).ICIs should be held until acute symptoms or symptoms related to mass effect have resolved and hormone replacement is initiated (42). One study compared discontinuing ipilimumab to restarting ipilimumab and found no effect on the resolution of hypophysitis (42). In the case of central hypothyroidism, replacement should be started after steroids are initiated. Testosterone and estrogen replacement should be considered in patients with central hypogonadism after discussing the risks and benefits of the medications (28).

 

Adrenalitis

 

Primary adrenal insufficiency (PAI), although being a rare endocrine irAEs is a potentially serious condition with significant morbidity and mortality if not identified early. Metastasis to the adrenal gland should be excluded. Other causes of adrenal insufficiency include sudden withdrawal of glucocorticoids and central adrenal insufficiency related to hypophysitis (49).

 

INCIDENCE

 

PAI is a rare side effect of ICIs, but early identification is essential given the risk of severe outcomes including death. Early evidence of adrenal insufficiency from ICIs came from case reports, but increasingly more evidence is available from larger studies and meta-analyses (50).

A review and meta-analysis by Barroso-Sousa et. al. that included 62 studies with 5831 patients, found the incidence of PAI was 0.7% for single ICI and 4.2% for combinations of ICIs (8). Another review and meta-analysis by Lu et. al. that included 160 studies and 40,432 patients, examined the rate of pituitary-adrenal dysfunction but did not distinguish the cause of adrenal insufficiency. One complicating factor in the study of PAI is that similar symptoms could occur from hypophysitis or discontinuation of steroids (41). Lu et. al. found adrenal insufficiency occurred in patients on ICIs in 2.43% of cases (ranging from 0-6.4% in studies) with serious grade adrenal effects in 0.15% of cases (ranging from 0-3.3%). Anti-CTLA-4 was associated with higher rates of adrenal insufficiency at 5.32% and serious grade events at 0.42%. Combination therapy also resulted in higher rates of adrenal insufficiency at 4.05%. Anti-PD-1 and anti-PD-L1 accounted for a smaller proportion of events at 0.49% and 0.43% respectively (41).

 

Grouthier et. al. used the World Health Organization global database, VigiBase, to examine individual safety reports for PAI and ICIs (4). The study found 451 cases of PAI, of which 45 were definite PAI and 406 possible PAI. In the study, 90% of cases involved significant morbidity including prolonged hospitalization, life-threatening illness, and disability. The mortality rate was 7.3%. Importantly the mortality rate appeared to be similar across immunotherapy treatments and combination treatments (4). This suggests that despite a relatively low incidence rate of PAI from ICIs, providers need to be able to identify these cases to prevent the significant risk of morbidity and mortality. 

 

PATHOPHYSIOLOGY

 

PAI is most frequently caused by autoimmune adrenal insufficiency (AI). Autoimmune AI is seen predominantly in women who make up between 54% to 83% of cases. In contrast, males accounted for the majority of ICI-related PAI cases at 58%, while females accounted for 36% of cases. In the remaining 6%, sex was unspecified. Autoimmune AI generally occurs between 30 to 50 years of age. In contrast, the age of onset with PAI caused by ICIs was 66 years on average with a range of 30-95 years old (1). In autoimmune AI, antibodies to the adrenal cortex including anti-21-hydroxylase are found in 83% to 88% of cases (4). The same antibodies have been found in case reports of ICI-related PAI (42). Adrenal metastasis should be excluded during the workup of adrenal insufficiency.

 

CLINICAL CHARACTERISTICS

 

Symptoms of PAI related to ICIs are similar to PAI from other causes. Symptoms include fatigue, postural dizziness, orthostatic hypotension, anorexia, weight loss, and abdominal pain. Adrenal crisis is suggested by altered mental status, weakness, syncope, nausea, and vomiting (42). In 52% of cases, other irAEs were also present. Other endocrinopathies made up 14.9% of these irAEs. The median time to onset was 120 days (ranging from 6-576 days) from starting the ICIs (4). Lab findings include hyponatremia, hyperkalemia, hypoglycemia, hypercalcemia, low aldosterone, elevated renin, elevated ACTH, and low to low normal cortisol. Imaging may reveal adrenalitis with enlarged adrenal glands. Interestingly, one case report found imaging evidence of adrenalitis present on a positron emission tomography (PET) scan after starting ipilimumab, but no symptoms or biochemical evidence of adrenal insufficiency. Repeat imaging revealed normal adrenal glands months later. This case suggests adrenalitis may occur without adrenal insufficiency (42).

 

SCREENING AND MONITORING

 

The NCCN guidelines recommend checking morning cortisol before each treatment or every four weeks during treatment. Additionally, follow-up testing is recommended for an additional six to twelve weeks. If cortisol is low or subnormal, ACTH monitoring is recommended. To monitor for pituitary and thyroid dysfunction, TSH and T4 monitoring at similar intervals are also recommended (28). In a review by Chang et. al., monitoring was recommended only in symptomatic patients, but a low index for suspicion was recommended as symptoms are nonspecific. When a patient has suspicious symptoms for adrenal dysfunction, ACTH and cortisol should be obtained before corticosteroid treatment only if this can be done safely. Additionally, measuring renin and aldosterone is helpful to determine if mineralocorticoid deficiency is present. This can be particularly helpful as case reports of central and PAI coexisting have been reported. The utility of adrenal autoantibodies, including 21-hydroxylase, is not well-established (42).

 

TREATMENT

 

PAI caused by ICIs is treated the same as other causes of PAI. If adrenal crisis or other critical illness is present, stress dose steroids with 100mg IV then 50mg IV every six hours is initiated. In stable patients, 15-25mg hydrocortisone is started in divided doses. Fludrocortisone is used to treat mineralocorticoid deficiency in PAI starting at 0.5-1mg daily. Additionally, patients will need to be educated on sick day rules and be provided with medical alert bracelets, and have high-dose corticosteroids for emergency purposes (28). The other important aspect of treatment is the decision to continue the ICIs. Holding the ICIs is recommended upon identification of adrenal insufficiency. Restarting immunotherapy can be considered after stabilization on hydrocortisone and fludrocortisone replacement.

 

Type 1 Diabetes

 

Rapid onset of autoimmune diabetes has been reported with ICIs use.  It is a rare but life-threatening side effect as it can present with diabetic ketoacidosis (DKA). The diabetes is permanent and requires lifelong treatment with insulin therapy (51). Notably, ICI-induced type 1 diabetes (T1D) has been reported with all clinically available PD-1 (nivolumab, pembrolizumab) and PD-L1 inhibitors (avelumab, durvalumab, atezolizumab) but rarely with the CTLA-4 inhibitor (ipilimumab).

 

INCIDENCE

 

The incidence of ICI-induced T1D comes from large case series at academic medical centers reporting 27 cases out of 2960 patients receiving ICI therapy (0.9%) (52) and 1/1163 (1.8%) (53).  Additionally, the prescription label for nivolumab reports that 17/1994 (0.9%) cases developed T1D (54). However, when examining the clinical trials evaluating the efficacy of PD-1 and PD-L1 inhibitors, there is a wide range of reported hyperglycemia and diabetes (55-64) (Table 1). From this analysis, hyperglycemia or diabetes was reported in approximately 2.5% of treated individuals.

 

Table 1. Clinical Trials Reporting Hyperglycemia/Diabetes with ICIs Use

Authors, Journal and Publication Year

Cases (n)

Study

Participants

(n)

Sideeffect (%)

Side effect

Drug

Cancer Type

Hamid et. al. NEJM, 2013 (55)

4

135

2.96

Hyperglycemia

Lambrolizumab

Melanoma

Borghaei et. al. NEJM, 2015 (56)

13

287

4.52

Hyperglycemia

Nivolumab

Lung

Motzer et. al. NEJM, 2015 (57)

9

406

2.21

Hyperglycemia

Nivolumab

Renal cell

Robert et. al. NEJM, 2016 (58)

1

206

0.48

Diabetes

Nivolumab

Melanoma

Nghiem et. al. NEJM, 2016 (59)

1

26

3.84

Hyperglycemia

Pembrolizumab

Merkel-cell

Kaufman et. al. Lancet, 2016 (60)

1

88

1.13

Type 1 Diabetes

Avelumab

Merkel-cell

Reck et. al. NEJM, 2016 (61)

1

154

0.64

Type 1 Diabetes

Pembrolizumab

Lung

Heery et. al. Lancet, 2017 (62)

3

53

5.66

Hyperglycemia

Avelumab

Solid tumors

Weber et. al. NEJM, 2017 (63)

2

452

0.44

Diabetes

Nivolumab

Melanoma

Choueiri et. al. Lancet, 2018 (64)

7

55

12.7

Hyperglycemia

Avelumab

Renal cell

 

Of note, most of the clinical trials in Table 1 excluded patients with a preexisting autoimmune condition, and some even excluded patients with a family history of autoimmunity. As these therapies are now being more widely used in clinical practice, there is an increased reporting of ICIs-induced diabetes (65). This is likely due to the increasing use of ICIs therapy and differences in patient populations between phase 2/3 clinical trials and clinical practice. Although T1D is a relatively rare occurrence with ICIs therapy, the events are clinically significant.

 

PATHOPHYSIOLOGY

 

The first case series reporting ICIs-induced autoimmune diabetes was described in 2015 (66). In this series of five patients, both humoral and cellular diabetes-associated autoimmunity were described. Some patients had positive T1D associated autoantibodies and diabetes-specific CD8+ T cells in the peripheral blood, consistent with findings from childhood-onset T1D (66).

The role of the PD-1/PD-L1 pathway in preclinical animal models of T1D has been appreciated for over a decade. Non-obese-diabetic (NOD) mice develop spontaneous autoimmune diabetes so it has been used extensively as an animal model to understand the mechanisms of T1D development (67). NOD mice with a knockout of either PD-1 or PD-L1 (but not PD-L2) have accelerated onset of diabetes with lymphocytic infiltration of the pancreatic islets (e.g., insulitis) compared to mice with these immune regulatory molecules (68, 69). Furthermore, administration of anti-PD-1or PD-L1 monoclonal antibodies to NOD mice also accelerated the onset of T1D (70). When examining the islets in NOD mice, insulin-producing beta-cells express PD-L1 during the progression of autoimmune diabetes (71). Similar to NOD mice, human islets from T1D organ donors exhibit upregulation of PD-L1, which was strongly associated with insulitis (72). This likely represents a protective mechanism for beta-cells to lessen their autoimmune destruction. These studies may explain why anti-PD-1/PD-L1 therapies induce T1D, while there is an absence of diabetes with anti-CTLA-4 therapy, whose ligands are CD80 and CD86 on antigen-presenting cells such as B cells, dendritic cells, and macrophages.

 

CLINICAL CHARACTERISTICS

 

Over the last 4 years, cases have described rapid-onset insulin-dependent diabetes with undetectable C-peptide levels (a measure of residual beta-cell function) and both positive and negative T1D associated autoantibodies at presentation (73, 74). Cases of ICIs-induced T1D have remained insulin-dependent even upon stopping therapy. Steroid treatment has not been able to reverse T1D, and as expected, blood glucose worsens with steroid administration (75, 76).

 

ICIs-induced T1D is mostly reported in older patients (50-70 years old) due to the nature of end-stage cancers developing later in life. More cases have been reported with anti-PD-1 therapies (nivolumab and pembrolizumab) as these agents were approved before monoclonal antibodies targeting anti-PD-L1 (51, 66, 73, 74, 77). Melanoma is the most common cancer in patients that present with ICIs-induced T1D, likely due to this being the first approved indication for ICIs therapy, and more patients with melanoma have been exposed to ICIs therapy compared to other cancer types. However, with the expanding indications and recent approval of ICIs therapy for use in pediatric cancers, ICIs-induced T1D may increase and present in younger individuals (78).

 

METABOLIC FEATURES

 

ICIs-induced T1D presents within days to a year after the initiation of PD-1 or PD-L1 therapy. HbA1c, which is a measure of the average blood glucose over the preceding three months, is generally lower than 10% at presentation with most patients presenting between 7 to 8%. As these values are mildly elevated, this suggests significant hyperglycemia over a short period rather than a gradual increase in hyperglycemia over a longer period. Most of the patients present with severe DKA that can be life-threatening. In most cases, C-peptide levels were inappropriately low for the presenting blood glucose or undetectable; ‘honeymoon’ periods tend to be absent after diagnosis. These observations suggest a destruction of beta-cell mass. In some patients, increased amylase and/or lipase has beenreported, suggesting more generalized pancreatic inflammation (52, 79).

 

IMMUNOLOGIC FEATURES

 

At least one T1D associated autoantibody, directed against insulin, glutamic acid decarboxylase (GAD), islet antigen-2 (IA-2), and zinc transporter 8 (ZnT8), was reported in 40-50% of the cases (52, 79). Almost all antibody-positive cases had GADA antibodies; however, not all four major autoantibodies were reported or measured in these case series. It is speculated that there is an association between antibody presence and earlier onset of ICIs-induced T1D in a subset of patients. In one case, positive conversion of antibodies after ICIs therapy was reported (52). Polyclonal and predominantly IgG1 subclass for GADA was shown at the presentation of another case that developed T1D five days after the initiation of PD-1 inhibitor therapy. Since IgG antibodies are involved in memory immune response and the short time interval from the initiation of anti-PD-1 treatment to the onset of T1D, these antibodies were likely present before the start of therapy (51). Based on these findings, a subset of patients developing ICIs-induced T1D likely have preexisting T1D associated antibodies which may be an early form of latent autoimmune diabetes of adulthood (LADA); however, prospective studies measuring T1D associated antibodies before the start of ICIs therapy are needed to evaluate this hypothesis.

 

GENETIC RISKS

 

Human leukocyte antigen (HLA) genes on chromosome 6 confer genetic risk for many autoimmune disorders including childhood-onset T1D (80). The polymorphic class II HLA genes (DQ, DR, and DP) confer this risk, especially the DR4-DQ8 and DR3-DQ2 haplotypes (81, 82). Only a small number of cases with ICIs-induced T1D have reported HLA genes with some having T1D risk alleles. In one case series, the frequency of HLA-DR4 was found to be enriched in those with ICIs-induced T1D compared to rates among Caucasians in the US population (52, 79). Further research is necessary to identify HLA and other genetic variants that may confer risk for ICIs-induced T1D.

 

COMPARISON TO CHILDHOOD-ONSET TYPE 1 DIABETES

 

We believe it is useful to compare the current knowledge of ICIs-induced T1D to prototypical childhood-onset T1D (Table 2). The age of onset is distinctly different between the two types of diabetes. Presentation with DKA is more common and the onset of diabetes more rapid than traditional T1D. T1D associated autoantibodies are present in ~90% of children and adolescents with T1D compared to half of the reported cases in ICIs-induced T1D. There is a predominance of GAD autoantibodies at the presentation of ICIs-induced T1D; however, more research is needed to measure all four major T1D associated autoantibodies in these patients and those directed against post-translationally modified antigens may also reveal insights into the pathogenesis of the disorder. C-peptide levels are low or undetected in those treated with ICIs therapy that develops T1D compared to C-peptide levels that vary and gradually go down after the diagnosis of childhood T1D. As a corollary, the honeymoon phase is generally absent in ICIs-induced T1D (80-83).

 

Table 2. Comparison Between Prototypical and Immune Checkpoint Inhibitor-Induced Type 1 Diabetes

Characteristics

Prototypical Type 1 Diabetes

Immune Checkpoint Inhibitor-Induced Type 1 Diabetes

Age of Onset

Peak in early childhood & adolescence

Later adulthood, 60’s

Diabetic ketoacidosis at Onset

Common

Very common

Pathophysiology

Autoimmune (years)

Autoimmune (days to months)

Autoantibodies

Present in 90-95%

Present in ~50%*

HLA Risk Genes

~90%

75-80%+

C-peptide at presentation

Varies

Low/absent

Honeymoon phase

Present

Absent

*Predominantly GADA antibodies;

+Small sample size, as not all cases report HLA alleles; there is an association with HLA-DR4

 

SCREENING AND MONITORING

 

The most updated recommendation on screening for diabetes in patient receiving ICIss comes from 2018 American Society of Clinical Oncology (ASCO) clinical practice guidelines, which recommends monitoring blood glucose at baseline, with each treatment cycle for 12 weeks and then every 3-6 weeks thereafter (24). In cases with suspected T1DM such as new onset hyperglycemia >200 mg/dl, random blood sugar >250 or history of T2DM with glucose levels >250 mg/dl, further testing for ketosis and anion gap is recommended (24). Discussing the risk of developing T1D with patients and educating them about the signs and symptoms of diabetes and DKA are recommended. Based on the current evidence, patients who have positive T1D associated antibodies and certain HLA alleles may have an increased risk to develop diabetes, so screening antibodies and reporting HLA alleles before the initiation of treatment may identify these patients with greater risk.

 

A retrospective study evaluated fasting blood glucose levels of patients receiving ICIs treatment during patient visits and showed no detectable upward drift of glycemia before DKA presentation (83). This is likely due to the rapid onset and progression of ICI-induced T1D. However, we believe monitoring blood glucose and HbA1c levels during patient visits are still necessary. Considering the rapid onset of diabetes, this approach alone may miss a significant amount of hyperglycemia and DKA. We recommend routine self-monitoring of blood glucose by patients and/or using continuous glucose monitoring to recognize hyperglycemia before DKA presentation. Close monitoring of patients with preexisting autoimmunity may also be useful (51). Our suggested screening and monitoring algorithm is depicted in Figure 3.

 

Figure 3. Proposed algorithm to screen and manage patients for ICI-induced T1D. (DKA = diabetic ketoacidosis; HbA1c = Hemoglobin A1c, T1D= Type 1 diabetes, HLA = human leukocyte antigen, CGM = continuous glucose monitor)

Hypogonadism

 

The effects of ICIs on sexual function are not very well known. ICI-induced primary hypogonadism is rare but a life-changing side effect, as it can potentially lead to infertility. Notably, gonadal dysfunction has been reported for ipilimumab monotherapy or in combination with PD-1/PD-L1 inhibitors (84). The long-term effects are still largely unknown. ICI-induced male hypogonadism is characterized by a deficiency in testosterone, which can be due to testicular, hypothalamic, or pituitary abnormalities. ICI-associated hypophysitis is discussed separately, and this section will primarily focus on ICI-induced primary hypogonadism.

 

INCIDENCE

 

Although, ICI-associated hypogonadism can be seen in patients who develop panhypopituitarism secondary to ICI-associated hypophysitis, the true occurrence of primary hypogonadism is uncommon and is based on a few case reports and ongoing studies (84-86). A recent analysis of VigiBase, the WHO global database of individual case safety reports between 2011 and 2019, found only 1 case of primary hypogonadism (87). This surprisingly low incidence may in fact be due to lack of proper evaluation looking for primary hypogonadism. For example, many studies reporting occurrence of secondary hypogonadism lacked data on the levels of pituitary gonadotropins, FSH and LH, which is necessary to differentiate between primary and secondary hypogonadism (43). Moreover, the majority of the pivotal trials leading to FDA approval of ICIs lacked information regarding fertility, menopause status, sex hormone levels, or sexual health-related quality of life. Additionally, not much is known about ICI-associated infertility. In a study of patients with malignant melanoma treated with ICIs, 6 of 7 men (86%) with testicular autopsy tissue samples had impaired spermatogenesis (88). This may suggest higher prevalence of infertility among men receiving ICIs. No data on potential effects on female fertility are currently available.

 

PATHOPHYSIOLOGY

 

ICIs may cause irAEs affecting any organ in the body by blocking regulators of self-tolerance. The understanding of pathophysiologic mechanism of ICI-induced primary hypogonadism comes from limited number of cases reports (85, 86). In the first case, the patient developed bilateral orchitis two weeks following administration of nivolumab and laboratory workup confirmed diagnosis of primary hypogonadism (decreased testosterone with elevated LH) (85). However, it self-resolved within one week without use of steroids or any other therapy, and there was no recurrence.  The intensity and timing of the orchitis suggests an intense immune stimulation leading to orchitis and primary hypogonadism (85). In another case, the patient developed bilateral epididymo-orchitis following administration of the third dose of pembrolizumab and needed high-dose steroids resulting in complete resolution (86). The testis is considered an immune-privileged organ due to its ability to tolerate autoantigens. The use of experimental autoimmune orchitis (EAO) in rats has allowed analysis of the autoimmune inflammatory response to spermatic antigens, providing a steppingstone towards understanding the ICI-induced primary hypogonadism. The main mechanisms responsible for preventing autoimmune disease of testes are: (a) secretion of immunosuppressive factors by macrophages, Sertoli cells, and Leydig cells, (b) presence of blood-testis barrier (BTB), and (c) presence of regulatory T cells. There is a fine equilibrium between dendritic cells, macrophages, T cells, and cytokines in maintaining immunosuppression in testes. While there have been no studies to date specifically evaluating the mechanism of ICI-induced orchitis, the examination of the normal and altered autoimmune immunobiology elucidates the possible mechanisms involved (89). This is briefly described below:

 

Secretion of Immunosuppressive Factors

 

In the normal testis macrophages, Sertoli and Leydig cells create an immunosuppressor microenvironment by secreting factors and cytokines that inhibit immune reactions. These include transforming growth factor-beta, granulocyte-macrophage colony stimulating factor, alpha-endorphin, and insulin-growth factor-1 (89, 90). In the setting of EAO, there is increased recruitment and activation of immune cells to the interstitium which bring along with them secretion of pro-inflammatory cytokines (IL-6, IFN-gamma, TNF-alpha, IL-17, IL-23). This brings about a cascade of events leading to germ cell apoptosis, primarily via the section of TNF-alpha (89, 91)

 

Blood-Testis Barrier (BTB)

 

In the normal testis, the BTB limits the interaction between germ cell antigens and interstitial immune cells. Secretion of pro-inflammatory cytokines mentioned above act on adherens and tight junctions, altering the BTB permeability (92). After crossing the BTB, these cytokines enter the seminiferous tubules inducing apoptosis of germ cells and facilitating the release of spermatic antigens, which then go on to interact with interstitial immune cells (92).

 

Presence of Regulatory T Cells (Tregs)

 

In the normal testis, there are several subsets of T cells present, regulating immune responses. Tregs specifically, mediate tolerance to self-antigens and their suppression sets the stage for autoimmunity. While there are increased Tregs seen in chronic inflamed testis, these are overwhelmed by the inhibitory effects of effector T cells, affecting the ability of Tregs to control autoimmunity (93). CTLA-4 inhibits effector T cells and PD-1/PDL-1 binding promotes the conversion of Teff to Treg. Therefore, it is plausible that the use of the combination of ipilimumab with an anti-PD-1/PDL-1 antibody, tips the balance between Tregs and effector cells toward the effector T cells. Consequently, creating a pro-inflammatory state resulting in orchitis.         

 

LONG-TERM OUTCOMES AND TREATMENT

 

It is well established that inflammation and infection of the male reproductive tract may lead to infertility in males (94). Therefore, it is reasonable to postulate that ICI-induced orchitis may also lead to male infertility, a consequence that should be addressed by providers. The long-term outcomes of ICIs are just beginning to be explored. One retrospective review assessed patients who became infertile after ICI therapy and subsequently died. Retrospective cohort cadaver study analyzing tissue specimens of the testes showed 86% of men who received ICI therapy had impaired spermatogenesis (88).  Notably, there was no increased peritubular hyalinization or fibrosis in the treated group, and no changes in Leydig cells (88). These findings support the previously mentioned pathophysiology of ICI-induced orchitis and address the possibility of infertility as a long-term consequence. Given the limited information on the effects of ICIs in spermatogenesis, providers should provide patients with their options, such as sperm banking and cryopreservation (95).

 

Other Uncommon Endocrine Side Effects  

 

ACQUIRED GENERALIZED LIPODYSTROPHY

 

Lipodystrophy is characterized by absent of visceral or subcutaneous adipose tissue in the settings of normal non-starvation nutritional state. It is a known common side effect from certain medications such as older HIV protease inhibitor, which is a reversible side effect. While the mechanism of lipodystrophy from ICIs is currently unclear, it is believed that the medication may induced an autoimmune process that leads to fat destruction by forming anti-adipocyte antibodies. In ICI-induced lipodystrophy, the more common form appears to be acquired generalized lipodystrophy (AGL) in which all fat tissues are affected but may spare the neck and face region. Onset of AGL, can be as early as 2-4 months which is roughly after 4-5 doses of ICIs. Currently, most of the cases of ICI-induced AGL are associated with nivolumab therapy (96, 97).

 

HYPOPARATHYROIDISM AND HYPOCALCEMIA  

 

Another rare but crucial endocrine irAEs is hypocalcemia secondary to hypoparathyroidism. While the exact mechanism is unclear, the proposed etiology is due to calcium-sensing receptor (CaSR) activating autoantibodies. This antibody is also present in patients with autoimmune polyendocrine syndrome type 1 (APS1) or idiopathic hypoparathyroidism. The clinical presentation can be as abrupt as an acute symptomatic hypocalcemia episode which includes paresthesia, tetany, and potential arrhythmias requiring hospitalization but may also present as very mild to asymptomatic hypocalcemia. For both circumstances, calcium and vitamin D replacement are adequate therapy but patients should be closely monitored for severe symptoms (98).

 

CENTRAL DIABETES INSIPIDUS  

 

Posterior pituitary hormone secretion can also be affected with ICIs, mainly antidiuretic hormones (ADH), which can subsequently lead to sodium and water dysregulation. To our knowledge only 3 cases of central diabetes insipidus (CDI) has been reported with the use of nivolumab (PD-1 inhibitor) and Azelumab (PD-L1 inhibitor) (99-101).The patients presented with classic polyuria/polydipsia symptoms along with hypernatremia which responded well to desmopressin (99-101). In the case report described by Fosci et. al., the authors described coexistence of metastatic localization and infundibulo-neurohypophysitis on MRI (100) while in the case report by Deligiori et. al., there was no signs of hypophysitis on imaging (99). Thus, further investigation is needed to fully understand the possible mechanisms for CDI.

 

SYNDROME OF INAPPROPRIATE ANTIDIURETIC HORMONE SECRETION (SIADH)

 

SIADH is the opposite scenario in which patients present with euvolemic hyponatremia. It is somewhat difficult to distinguish for certain that SIADH is truly from ICIs since SIADH is quite common in patients with underlying malignancies. Additionally, pain in patients with cancer itself can be the underlying cause of SIADH. Additionally, there are some reports of hyponatremia as a manifestation of adrenal insufficiency in patients on ICIs and hence it is crucial to rule out adrenal insufficiency for any patient with hyponatremia, as immediate recognition and treatment can be lifesaving (102, 103).

 

VITILIGO

 

Depigmentation of skin or vitiligo is thought to be from inducing an immune response to normal melanocyte antigens leading to the destructive process. While vitiligo itself may not be directly endocrine-related, its presence has been strongly associated with common endocrinopathies such as thyroid and adrenal disease as well as autoimmune diabetes. Interestingly, when vitiligo is present as one of the side effects from ICIs, this may represent a better prognosis in melanoma cases (104).

Figure 4. Proposed algorithm to screen and manage patients with endocrine irAEs

 

Table 3. Summary of the Common Terminology Criteria for Adverse Events (28)

Grade

Severity of Adverse events

Management

1

Mild (asymptomatic or mild symptoms)

Clinical or diagnostic observation

2

Moderate

Minimal, local or noninvasive intervention indicated

3

Severe or medically significant but not immediate life threatening

Intervention is required

4

Life threatening

Urgent intervention indication

5

Death

 

 

Table 4. Summary of the Incidence of Endocrine iRAEs (8,16,23, 41, 55-64, 105)

irAEs

PD-1/L1 inhibitors

CTLA-4 inhibitors

Combination

Hypophysitis

Less than 1 %

0-17%

 

 

 

More common than single drug use. 

Hypothyroidism

7-21%

0-6%

hyperthyroidism

Higher in PD1 inhibitors compared to PDL1 inhibitors

Less common than PD-1/PDL1 inhibitors

Primary adrenal insufficiency

Less common than CTLA-4 inhibitors

More common than PD-1/PDL1 inhibitors

Diabetes

Around 2.5%

None reported

 

CONCLUSION

 

Considering the increasing use of immune checkpoint inhibitors in clinical practice, health care providers and patients should be aware of endocrine irAEs. Educating patients receiving and providers using these state-of-the-art therapies about the signs and symptoms of different endocrinopathies is critical for an early diagnosis to prevent life-threatening complications. Developing screening and monitoring guidelines are essential to identify at-risk patients for close monitoring of these unwanted side effect.

 

REFERENCES

 

  1. Thangamathesvaran L, Shah R, Verma R, Mahmoud O. Immune checkpoint inhibitors and radiotherapy-concept and review of current literature. Ann Transl Med. 2018;6(8):155.
  2. Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer. 2012;12(4):252-64.
  3. Leach DR, Krummel MF, Allison JP. Enhancement of antitumor immunity by CTLA-4 blockade. Science. 1996;271(5256):1734-6.
  4. Grouthier V, Lebrun-Vignes B, Moey M, Johnson DB, Moslehi JJ, Salem JE, et al. Immune Checkpoint Inhibitor-Associated Primary Adrenal Insufficiency: WHO VigiBase Report Analysis. Oncologist. 2020;25(8):696-701.
  5. Peggs KS, Quezada SA, Korman AJ, Allison JP. Principles and use of anti-CTLA4 antibody in human cancer immunotherapy. Curr Opin Immunol. 2006;18(2):206-13.
  6. Thompson RH, Dong H, Kwon ED. Implications of B7-H1 expression in clear cell carcinoma of the kidney for prognostication and therapy. Clin Cancer Res. 2007;13(2 Pt 2):709s-15s.
  7. Zang X, Allison JP. The B7 family and cancer therapy: costimulation and coinhibition. Clin Cancer Res. 2007;13(18 Pt 1):5271-9.
  8. Barroso-Sousa R, Barry WT, Garrido-Castro AC, Hodi FS, Min L, Krop IE, et al. Incidence of Endocrine Dysfunction Following the Use of Different Immune Checkpoint Inhibitor Regimens: A Systematic Review and Meta-analysis. JAMA Oncol. 2018;4(2):173-82.
  9. Byun DJ, Wolchok JD, Rosenberg LM, Girotra M. Cancer immunotherapy - immune checkpoint blockade and associated endocrinopathies. Nat Rev Endocrinol. 2017;13(4):195-207.
  10. Weber JS, Dummer R, de Pril V, Lebbé C, Hodi FS. Patterns of onset and resolution of immune-related adverse events of special interest with ipilimumab: detailed safety analysis from a phase 3 trial in patients with advanced melanoma. Cancer. 2013;119(9):1675-82.
  11. Johnson DB, Sullivan RJ, Ott PA, Carlino MS, Khushalani NI, Ye F, et al. Ipilimumab Therapy in Patients With Advanced Melanoma and Preexisting Autoimmune Disorders. JAMA Oncol. 2016;2(2):234-40.
  12. Eggermont AMM, Kicinski M, Blank CU, Mandala M, Long GV, Atkinson V, et al. Association Between Immune-Related Adverse Events and Recurrence-Free Survival Among Patients With Stage III Melanoma Randomized to Receive Pembrolizumab or Placebo: A Secondary Analysis of a Randomized Clinical Trial. JAMA Oncol. 2020;6(4):519-27.
  13. Maher VE, Fernandes LL, Weinstock C, Tang S, Agarwal S, Brave M, et al. Analysis of the Association Between Adverse Events and Outcome in Patients Receiving a Programmed Death Protein 1 or Programmed Death Ligand 1 Antibody. J Clin Oncol. 2019;37(30):2730-7.
  14. Suo A, Chan Y, Beaulieu C, Kong S, Cheung WY, Monzon JG, et al. Anti-PD1-Induced Immune-Related Adverse Events and Survival Outcomes in Advanced Melanoma. Oncologist. 2020;25(5):438-46.
  15. Abdel-Rahman O, ElHalawani H, Fouad M. Risk of endocrine complications in cancer patients treated with immune check point inhibitors: a meta-analysis. Future Oncol. 2016;12(3):413-25.
  16. Kotwal A, Kottschade L, Ryder M. PD-L1 Inhibitor-Induced Thyroiditis Is Associated with Better Overall Survival in Cancer Patients. Thyroid. 2020;30(2):177-84.
  17. Delivanis DA, Gustafson MP, Bornschlegl S, Merten MM, Kottschade L, Withers S, et al. Pembrolizumab-Induced Thyroiditis: Comprehensive Clinical Review and Insights Into Underlying Involved Mechanisms. The Journal of clinical endocrinology and metabolism. 2017;102(8):2770-80.
  18. Kobayashi T, Iwama S, Yasuda Y, Okada N, Tsunekawa T, Onoue T, et al. Patients With Antithyroid Antibodies Are Prone To Develop Destructive Thyroiditis by Nivolumab: A Prospective Study. J Endocr Soc. 2018;2(3):241-51.
  19. Toi Y, Sugawara S, Sugisaka J, Ono H, Kawashima Y, Aiba T, et al. Profiling Preexisting Antibodies in Patients Treated With Anti-PD-1 Therapy for Advanced Non-Small Cell Lung Cancer. JAMA Oncol. 2019;5(3):376-83.
  20. Kurimoto C, Inaba H, Ariyasu H, Iwakura H, Ueda Y, Uraki S, et al. Predictive and sensitive biomarkers for thyroid dysfunctions during treatment with immune-checkpoint inhibitors. Cancer Sci. 2020;111(5):1468-77.
  21. Angell TE, Min L, Wieczorek TJ, Hodi FS. Unique Cytologic Features of Thyroiditis Caused by Immune Checkpoint Inhibitor Therapy for Malignant Melanoma. Genes Dis. 2018;5(1):46-8.
  22. Tan MH, Iyengar R, Mizokami-Stout K, Yentz S, MacEachern MP, Shen LY, et al. Spectrum of immune checkpoint inhibitors-induced endocrinopathies in cancer patients: a scoping review of case reports. Clin Diabetes Endocrinol. 2019;5:1.
  23. Wright JJ, Powers AC, Johnson DB. Endocrine toxicities of immune checkpoint inhibitors. Nat Rev Endocrinol. 2021;17(7):389-99.
  24. Brahmer JR, Lacchetti C, Schneider BJ, Atkins MB, Brassil KJ, Caterino JM, et al. Management of Immune-Related Adverse Events in Patients Treated With Immune Checkpoint Inhibitor Therapy: American Society of Clinical Oncology Clinical Practice Guideline. J Clin Oncol. 2018;36(17):1714-68.
  25. Network NCC. Management of Immunotherapy-Related Toxicities Version 4.2021 2021 [Available from: https://www.nccn.org/professionals/physician_gls/pdf/immunotherapy.pdf.
  26. Bekkering GE, Agoritsas T, Lytvyn L, Heen AF, Feller M, Moutzouri E, et al. Thyroid hormones treatment for subclinical hypothyroidism: a clinical practice guideline. Bmj. 2019;365:l2006.
  27. Pearce SH, Brabant G, Duntas LH, Monzani F, Peeters RP, Razvi S, et al. 2013 ETA Guideline: Management of Subclinical Hypothyroidism. Eur Thyroid J. 2013;2(4):215-28.
  28. Thompson JA, Schneider BJ, Brahmer J, Andrews S, Armand P, Bhatia S, et al. Management of Immunotherapy-Related Toxicities, Version 1.2019. J Natl Compr Canc Netw. 2019;17(3):255-89.
  29. Brancatella A, Viola N, Brogioni S, Montanelli L, Sardella C, Vitti P, et al. Graves' Disease Induced by Immune Checkpoint Inhibitors: A Case Report and Review of the Literature. Eur Thyroid J. 2019;8(4):192-5.
  30. Iyer PC, Cabanillas ME, Waguespack SG, Hu MI, Thosani S, Lavis VR, et al. Immune-Related Thyroiditis with Immune Checkpoint Inhibitors. Thyroid. 2018;28(10):1243-51.
  31. Yamauchi I, Sakane Y, Fukuda Y, Fujii T, Taura D, Hirata M, et al. Clinical Features of Nivolumab-Induced Thyroiditis: A Case Series Study. Thyroid. 2017;27(7):894-901.
  32. Yonezaki K, Kobayashi T, Imachi H, Yoshimoto T, Kikuchi F, Fukunaga K, et al. Combination therapy of ipilimumab and nivolumab induced thyroid storm in a patient with Hashimoto's disease and diabetes mellitus: a case report. Journal of medical case reports. 2018;12(1):171.
  33. Yu C, Chopra IJ, Ha E. A novel melanoma therapy stirs up a storm: ipilimumab-induced thyrotoxicosis. Endocrinol Diabetes Metab Case Rep. 2015;2015:140092.
  34. Ross DS, Burch HB, Cooper DS, Greenlee MC, Laurberg P, Maia AL, et al. 2016 American Thyroid Association Guidelines for Diagnosis and Management of Hyperthyroidism and Other Causes of Thyrotoxicosis. Thyroid. 2016;26(10):1343-421.
  35. Sjolin G, Holmberg M, Torring O, Bystrom K, Khamisi S, de Laval D, et al. The Long-Term Outcome of Treatment for Graves' Hyperthyroidism. Thyroid. 2019;29(11):1545-57.
  36. Albarel F, Gaudy C, Castinetti F, Carre T, Morange I, Conte-Devolx B, et al. Long-term follow-up of ipilimumab-induced hypophysitis, a common adverse event of the anti-CTLA-4 antibody in melanoma. Eur J Endocrinol. 2015;172(2):195-204.
  37. Faje AT, Sullivan R, Lawrence D, Tritos NA, Fadden R, Klibanski A, et al. Ipilimumab-induced hypophysitis: a detailed longitudinal analysis in a large cohort of patients with metastatic melanoma. The Journal of clinical endocrinology and metabolism. 2014;99(11):4078-85.
  38. Caturegli P, Newschaffer C, Olivi A, Pomper MG, Burger PC, Rose NR. Autoimmune hypophysitis. Endocr Rev. 2005;26(5):599-614.
  39. González-Rodríguez E, Rodríguez-Abreu D. Immune Checkpoint Inhibitors: Review and Management of Endocrine Adverse Events. Oncologist. 2016;21(7):804-16.
  40. Torino F, Barnabei A, Paragliola RM, Marchetti P, Salvatori R, Corsello SM. Endocrine side-effects of anti-cancer drugs: mAbs and pituitary dysfunction: clinical evidence and pathogenic hypotheses. Eur J Endocrinol. 2013;169(6):R153-64.
  41. Lu J, Li L, Lan Y, Liang Y, Meng H. Immune checkpoint inhibitor-associated pituitary-adrenal dysfunction: A systematic review and meta-analysis. Cancer Med. 2019;8(18):7503-15.
  42. 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(1):17-65.
  43. Garon-Czmil J, Petitpain N, Rouby F, Sassier M, Babai S, Yéléhé-Okouma M, et al. Immune check point inhibitors-induced hypophysitis: a retrospective analysis of the French Pharmacovigilance database. Scientific reports. 2019;9(1):19419.
  44. Iwama S, De Remigis A, Callahan MK, Slovin SF, Wolchok JD, Caturegli P. Pituitary expression of CTLA-4 mediates hypophysitis secondary to administration of CTLA-4 blocking antibody. Sci Transl Med. 2014;6(230):230ra45.
  45. Falorni A, Minarelli V, Bartoloni E, Alunno A, Gerli R. Diagnosis and classification of autoimmune hypophysitis. Autoimmun Rev. 2014;13(4-5):412-6.
  46. Kurokawa R, Ota Y, Gonoi W, Hagiwara A, Kurokawa M, Mori H, et al. MRI Findings of Immune Checkpoint Inhibitor-Induced Hypophysitis: Possible Association with Fibrosis. AJNR Am J Neuroradiol. 2020;41(9):1683-9.
  47. Lupi I, Brancatella A, Cosottini M, Viola N, Lanzolla G, Sgrò D, et al. Clinical heterogeneity of hypophysitis secondary to PD-1/PD-L1 blockade: insights from four cases. Endocrinol Diabetes Metab Case Rep. 2019;2019.
  48. Blansfield JA, Beck KE, Tran K, Yang JC, Hughes MS, Kammula US, et al. Cytotoxic T-lymphocyte-associated antigen-4 blockage can induce autoimmune hypophysitis in patients with metastatic melanoma and renal cancer. J Immunother. 2005;28(6):593-8.
  49. Haissaguerre M, Hescot S, Bertherat J, Chabre O. Expert opinions on adrenal complications in immunotherapy. Ann Endocrinol (Paris). 2018;79(5):539-44.
  50. Elshimy G, Gandhi A, Guo R, Correa R. Tyrosine Kinase Inhibitors' Newly Reported Endocrine Side Effect: Pazopanib-Induced Primary Adrenal Insufficiency in a Patient With Metastatic Renal Cell Cancer. J Investig Med High Impact Case Rep. 2020;8:2324709620936808.
  51. Akturk HK, Alkanani A, Zhao Z, Yu L, Michels AW. PD-1 Inhibitor Immune-Related Adverse Events in Patients With Preexisting Endocrine Autoimmunity. The Journal of clinical endocrinology and metabolism. 2018;103(10):3589-92.
  52. Stamatouli AM, Quandt Z, Perdigoto AL, Clark PL, Kluger H, Weiss SA, et al. Collateral Damage: Insulin-Dependent Diabetes Induced With Checkpoint Inhibitors. Diabetes. 2018;67(8):1471-80.
  53. Kotwal A, Haddox C, Block M, Kudva YC. Immune checkpoint inhibitors: an emerging cause of insulin-dependent diabetes. BMJ Open Diabetes Res Care. 2019;7(1):e000591.
  54. Nivolumab label information 2018 [Available from: https://packageinserts.bms.com/pi/pi_opdivo.pdf.
  55. Hamid O, Robert C, Daud A, Hodi FS, Hwu WJ, Kefford R, et al. Safety and tumor responses with lambrolizumab (anti-PD-1) in melanoma. The New England journal of medicine. 2013;369(2):134-44.
  56. Borghaei H, Paz-Ares L, Horn L, Spigel DR, Steins M, Ready NE, et al. Nivolumab versus Docetaxel in Advanced Nonsquamous Non-Small-Cell Lung Cancer. The New England journal of medicine. 2015;373(17):1627-39.
  57. Motzer RJ, Escudier B, McDermott DF, George S, Hammers HJ, Srinivas S, et al. Nivolumab versus Everolimus in Advanced Renal-Cell Carcinoma. The New England journal of medicine. 2015;373(19):1803-13.
  58. Robert C, Long GV, Brady B, Dutriaux C, Maio M, Mortier L, et al. Nivolumab in previously untreated melanoma without BRAF mutation. The New England journal of medicine. 2015;372(4):320-30.
  59. Nghiem PT, Bhatia S, Lipson EJ, Kudchadkar RR, Miller NJ, Annamalai L, et al. PD-1 Blockade with Pembrolizumab in Advanced Merkel-Cell Carcinoma. The New England journal of medicine. 2016;374(26):2542-52.
  60. Kaufman HL, Russell J, Hamid O, Bhatia S, Terheyden P, D'Angelo SP, et al. Avelumab in patients with chemotherapy-refractory metastatic Merkel cell carcinoma: a multicentre, single-group, open-label, phase 2 trial. Lancet Oncol. 2016;17(10):1374-85.
  61. Reck M, Rodríguez-Abreu D, Robinson AG, Hui R, Csőszi T, Fülöp A, et al. Pembrolizumab versus Chemotherapy for PD-L1-Positive Non-Small-Cell Lung Cancer. The New England journal of medicine. 2016;375(19):1823-33.
  62. Heery CR, O'Sullivan-Coyne G, Madan RA, Cordes L, Rajan A, Rauckhorst M, et al. Avelumab for metastatic or locally advanced previously treated solid tumours (JAVELIN Solid Tumor): a phase 1a, multicohort, dose-escalation trial. Lancet Oncol. 2017;18(5):587-98.
  63. Weber J, Mandala M, Del Vecchio M, Gogas HJ, Arance AM, Cowey CL, et al. Adjuvant Nivolumab versus Ipilimumab in Resected Stage III or IV Melanoma. The New England journal of medicine. 2017;377(19):1824-35.
  64. Choueiri TK, Larkin J, Oya M, Thistlethwaite F, Martignoni M, Nathan P, et al. Preliminary results for avelumab plus axitinib as first-line therapy in patients with advanced clear-cell renal-cell carcinoma (JAVELIN Renal 100): an open-label, dose-finding and dose-expansion, phase 1b trial. Lancet Oncol. 2018;19(4):451-60.
  65. Wright JJ, Salem JE, Johnson DB, Lebrun-Vignes B, Stamatouli A, Thomas JW, et al. Increased Reporting of Immune Checkpoint Inhibitor-Associated Diabetes. Diabetes care. 2018;41(12):e150-e1.
  66. Hughes J, Vudattu N, Sznol M, Gettinger S, Kluger H, Lupsa B, et al. Precipitation of autoimmune diabetes with anti-PD-1 immunotherapy. Diabetes Care. 2015;38(4):e55-7.
  67. Mullen Y. Development of the Nonobese Diabetic Mouse and Contribution of Animal Models for Understanding Type 1 Diabetes. Pancreas. 2017;46(4):455-66.
  68. Wang J, Yoshida T, Nakaki F, Hiai H, Okazaki T, Honjo T. Establishment of NOD-Pdcd1-/- mice as an efficient animal model of type I diabetes. Proc Natl Acad Sci U S A. 2005;102(33):11823-8.
  69. Keir ME, Liang SC, Guleria I, Latchman YE, Qipo A, Albacker LA, et al. Tissue expression of PD-L1 mediates peripheral T cell tolerance. J Exp Med. 2006;203(4):883-95.
  70. Ansari MJ, Salama AD, Chitnis T, Smith RN, Yagita H, Akiba H, et al. The programmed death-1 (PD-1) pathway regulates autoimmune diabetes in nonobese diabetic (NOD) mice. J Exp Med. 2003;198(1):63-9.
  71. Rui J, Deng S, Arazi A, Perdigoto AL, Liu Z, Herold KC. beta Cells that Resist Immunological Attack Develop during Progression of Autoimmune Diabetes in NOD Mice. Cell metabolism. 2017;25(3):727-38.
  72. Osum KC, Burrack AL, Martinov T, Sahli NL, Mitchell JS, Tucker CG, et al. Interferon-gamma drives programmed death-ligand 1 expression on islet beta cells to limit T cell function during autoimmune diabetes. Scientific reports. 2018;8(1):8295.
  73. Gaudy C, Clévy C, Monestier S, Dubois N, Préau Y, Mallet S, et al. Anti-PD1 Pembrolizumab Can Induce Exceptional Fulminant Type 1 Diabetes. Diabetes Care. 2015;38(11):e182-3.
  74. Mellati M, Eaton KD, Brooks-Worrell BM, Hagopian WA, Martins R, Palmer JP, et al. Anti-PD-1 and Anti-PDL-1 Monoclonal Antibodies Causing Type 1 Diabetes. Diabetes Care. 2015;38(9):e137-8.
  75. Fukui A, Sugiyama K, Yamada T, Tajitsu M, Cao X, Nagai J, et al. A Case of Nivolumab-Induced Fulminant Type 1 Diabetes with Steroidsand Glucagon-Like Peptide 1 Administration during the Early Onset. journal of Clinical Case Reports. 2016;2016.
  76. Kapke J, Shaheen Z, Kilari D, Knudson P, Wong S. Immune Checkpoint Inhibitor-Associated Type 1 Diabetes Mellitus: Case Series, Review of the Literature, and Optimal Management. Case Rep Oncol. 2017;10(3):897-909.
  77. Zaied AA, Akturk HK, Joseph RW, Lee AS. New-onset insulin-dependent diabetes due to nivolumab. Endocrinol Diabetes Metab Case Rep. 2018;2018.
  78. Wedekind MF, Denton NL, Chen CY, Cripe TP. Pediatric Cancer Immunotherapy: Opportunities and Challenges. Paediatr Drugs. 2018;20(5):395-408.
  79. Clotman K, Janssens K, Specenier P, Weets I, De Block CEM. Programmed Cell Death-1 Inhibitor-Induced Type 1 Diabetes Mellitus. The Journal of clinical endocrinology and metabolism. 2018;103(9):3144-54.
  80. Noble JA, Valdes AM. Genetics of the HLA region in the prediction of type 1 diabetes. Curr Diab Rep. 2011;11(6):533-42.
  81. Atkinson MA, Eisenbarth GS, Michels AW. Type 1 diabetes. Lancet. 2014;383(9911):69-82.
  82. Noble JA, Valdes AM, Varney MD, Carlson JA, Moonsamy P, Fear AL, et al. HLA class I and genetic susceptibility to type 1 diabetes: results from the Type 1 Diabetes Genetics Consortium. Diabetes. 2010;59(11):2972-9.
  83. Magis Q, Gaudy-Marqueste C, Basire A, Loundou A, Malissen N, Troin L, et al. Diabetes and Blood Glucose Disorders Under Anti-PD1. J Immunother. 2018;41(5):232-40.
  84. Sood A, Cole D, Abdollah F, Eilender B, Roumayah Z, Deebajah M, et al. Endocrine, Sexual Function, and Infertility Side Effects of Immune Checkpoint Inhibitor Therapy for Genitourinary Cancers. Curr Urol Rep. 2018;19(9):68.
  85. Brunet-Possenti F, Opsomer MA, Gomez L, Ouzaid I, Descamps V. Immune checkpoint inhibitors-related orchitis. Ann Oncol. 2017;28(4):906-7.
  86. Quach HT, Robbins CJ, Balko JM, Chiu CY, Miller S, Wilson MR, et al. Severe Epididymo-Orchitis and Encephalitis Complicating Anti-PD-1 Therapy. Oncologist. 2019;24(7):872-6.
  87. Bai X, Lin X, Zheng K, Chen X, Wu X, Huang Y, et al. Mapping endocrine toxicity spectrum of immune checkpoint inhibitors: a disproportionality analysis using the WHO adverse drug reaction database, VigiBase. Endocrine. 2020;69(3):670-81.
  88. Scovell JM, Benz K, Samarska I, Kohn TP, Hooper JE, Matoso A, et al. Association of Impaired Spermatogenesis With the Use of Immune Checkpoint Inhibitors in Patients With Metastatic Melanoma. JAMA Oncol. 2020;6(8):1297-9.
  89. Jacobo P, Guazzone VA, Theas MS, Lustig L. Testicular autoimmunity. Autoimmun Rev. 2011;10(4):201-4.
  90. Hedger MP, Meinhardt A. Cytokines and the immune-testicular axis. J Reprod Immunol. 2003;58(1):1-26.
  91. Schuppe HC, Meinhardt A. Immune privilege and inflammation of the testis. Chem Immunol Allergy. 2005;88:1-14.
  92. Pérez CV, Sobarzo CM, Jacobo PV, Pellizzari EH, Cigorraga SB, Denduchis B, et al. Loss of occludin expression and impairment of blood-testis barrier permeability in rats with autoimmune orchitis: effect of interleukin 6 on Sertoli cell tight junctions. Biol Reprod. 2012;87(5):122.
  93. Jacobo P. The role of regulatory T Cells in autoimmune orchitis. Andrologia. 2018;50(11):e13092.
  94. P. J. Rowe FC, T. B. Hargreave and A. Mahmoud. WHO Manual for the Standardized Investigation, Diagnosis and Management of the Infertile Male. Cambidge University Press. 2000:102.
  95. Oktay K, Harvey BE, Partridge AH, Quinn GP, Reinecke J, Taylor HS, et al. Fertility Preservation in Patients With Cancer: ASCO Clinical Practice Guideline Update. J Clin Oncol. 2018;36(19):1994-2001.
  96. Haddad N, Vidal-Trecan T, Baroudjian B, Zagdanski AM, Arangalage D, Battistella M, et al. Acquired generalized lipodystrophy under immune checkpoint inhibition. Br J Dermatol. 2020;182(2):477-80.
  97. Jehl A, Cugnet-Anceau C, Vigouroux C, Legeay AL, Dalle S, Harou O, et al. Acquired Generalized Lipodystrophy: A New Cause of Anti-PD-1 Immune-Related Diabetes. Diabetes Care. 2019;42(10):2008-10.
  98. Nalluru SS, Piranavan P, Ning Y, Ackula H, Siddiqui AD, Trivedi N. Hypocalcemia with Immune Checkpoint Inhibitors: The Disparity among Various Reports. International journal of endocrinology. 2020;2020:7459268.
  99. Deligiorgi MV, Siasos G, Vergadis C, Trafalis DT. Central diabetes insipidus related to anti-programmed cell-death 1 protein active immunotherapy. Int Immunopharmacol. 2020;83:106427.
  100. Fosci M, Pigliaru F, Salcuni AS, Ghiani M, Cherchi MV, Calia MA, et al. Diabetes insipidus secondary to nivolumab-induced neurohypophysitis and pituitary metastasis. Endocrinol Diabetes Metab Case Rep. 2021;2021.
  101. Zhao C, Tella SH, Del Rivero J, Kommalapati A, Ebenuwa I, Gulley J, et al. Anti-PD-L1 Treatment Induced Central Diabetes Insipidus. The Journal of clinical endocrinology and metabolism. 2018;103(2):365-9.
  102. Abdulla H. Primary hyperparathyroidism: molecular genetic insights and clinical implications.Society for Endocrinology BES 2017; Harrogate, UK: Bioscientifica; 2017.
  103. Desikan SP, Varghese R, Kamoga R, Desikan R. Acute hyponatremia from immune checkpoint inhibitor therapy for non-small cell lung cancer. Postgrad Med J. 2020;96(1139):570-1.
  104. Kosche C, Mohindra N, Choi JN. Vitiligo in a patient undergoing nivolumab treatment for non-small cell lung cancer. JAAD Case Rep. 2018;4(10):1042-4.
  105. de Filette J, Andreescu CE, Cools F, Bravenboer B, Velkeniers B. A Systematic Review and Meta-Analysis of Endocrine-Related Adverse Events Associated with Immune Checkpoint Inhibitors. Horm Metab Res. 2019 Mar;51(3):145-156. doi: 10.1055/a-0843-3366. Epub 2019 Mar 12. PMID: 30861560.

Medical Interventions for Transgender Youth

ABSTRACT

 

Up to 1.8% of youth and 0.6% of adults in the United States identify as transgender, meaning their gender identity differs from or is opposite their sex designated at birth. This chapter provides an overview including epidemiology and gender development. Then, it aims to summarize medical interventions for transgender youth as outlined in the Endocrine Society Clinical Practice Guidelines and World Professional Association for Transgender Health standards of care. The chapter concludes with research on mental health in this population and future directions.

 

INTRODUCTION

 

Throughout history and across cultures there have been people who live with, what we would now term, gender incongruence (definitions in Table 1). Prior to identification of sex steroids in the 1930s (1-5), and the development of exogenous sex steroids and surgical techniques, there were no options to change one’s secondary sex characteristics. The first modern orchiectomy for gender reassignment was performed in 1930 (6), and the first feminizing genital surgeries in the 1940s and 50s in Germany and Denmark, respectively (7,8). Harry Benjamin, known for his 1966 book, The Transsexual Phenomenon (9), treated Christine Jorgensen, the first widely published case of a transgender female in the United States (U.S.), treated with feminizing hormones and surgery. In 1979, the Harry Benjamin International Gender Dysphoria Association was formed, now the World Professional Association for Transgender Health (WPATH). The first standards of care were published in 1979, with the 7th edition released in 2012, and the 8th edition coming soon. The Endocrine Society first published a clinical practice guideline regarding the care of transgender persons, including support for pubertal suppression and gender affirming hormone therapy (GAHT) in 2009, with an updated guideline released in 2017 (10,11). In the over 40 years since the first edition of the WPATH Standards of Care, transgender rights, access to care, bathroom use, and sports participation, among other topics, are often featured and debated in mainstream media, politics, and healthcare (12). Furthermore, as care becomes increasingly politicized, numerous bills to both expand or limit the rights of transgender people and their access to medical care are being introduced in the U.S.(13). Medical care that respects the gender identity of the patient is recommended by numerous medical organizations, including the American Academy of Pediatrics (14),Endocrine Society (11), and the American Psychological Association (15).

 

Table 1. Definitions

Agender

A person with very little or no connection to the traditional systems of gender; existing without gender

Cisgender

Gender identity aligns with biologic sex

Gender affirming hormone therapy

Hormones, including testosterone and /or estradiol, that are prescribed to eligible individuals to induce development of secondary sex characteristics that align with gender identity

Gender affirming surgery (sometimes referred to as gender-confirming or gender-reassignment surgery)

Surgery or surgeries to align one’s body with one’s gender identity

Gender diverse

Individuals with a variety of gender identities across the gender spectrum, including those who identify as transgender

Gender dysphoria

Distress experienced when gender identity and body are not congruent. Defined in the DSM-5, which replaced “gender identity disorder” in the DSM-IV

Gender expression

External manifestations of gender, expressed through name, pronouns, clothing, haircut, behavior, voice, or other characteristics

Gender identity/experienced gender

One’s internal, deeply held sense of gender; not visible to others

Gender incongruence

Umbrella term used when gender identity and/or expression differ from what is typically/societally associated with their sex designated at birth

Gender role

Behaviors, attitudes, and personality trait that a society (in a given culture and historical period) designates as masculine or feminine and/or that society associates with the typical social role of men or women

Non-binary

A person whose gender identity is neither male nor female, both male and female or some combination of genders

Sex designated at birth

Sex at birth, typically based on external appearance of genitalia

Sex

Attributes that characterize biologic maleness or femaleness; factors that influence sex include sex chromosomes, gonads, sex steroids, internal reproductive structures, external genitalia, secondary sex characteristics

Sexual orientation

Physical and emotional attraction to others. Gender identity and sexual orientation are not the same

Transgender

Gender identity differs from sex designated at birth

Transgender male (also transgender man, female-to-male)

Individuals designed female at birth who identify and live as men

Transgender female (also transgender woman, male-to-female)

Individuals designated male at birth who identify and live as women

Transition

Process during which persons change their physical, social, and/or legal characteristics consistent with their affirmed gender identity

Adapted from Table 1 in the 2017 Endocrine Society Guidelines (11)

 

Centers around the world are seeing a rise in the number of transgender and gender diverse (TGD, Table 1) people seeking care to align their bodies with their identities (16). Despite the rise in the number of TGD people seeking care, there remains a lack of education and knowledge among providers as to how best serve this group (17). This article will define terminology, briefly review gender development, review current guidelines regarding medical treatment of pediatric TGD individuals, and mental health considerations.

 

EPIDEMIOLOGY  

 

Recent population-based studies in the U.S. report that 1.8% of youth and 0.6% of adults identify as transgender (18,19). Gender diverse describes individuals with a variety of gender identities across the gender spectrum, including those who identify as transgender (Table 1). Gender dysphoria, which describes the distress associated with a conflict between gender identity and anatomy or sex, is a listed diagnosis in the Diagnostic and Statistical Manual of Mental Disorders 5th edition (DSM-5, Table 2) (20).

 

Table 2. DSM-5 Criteria for Gender Dysphoria (20)

A.    A marked incongruence between one’s experienced/expressed gender and natal gender of at least 6 months in duration, as manifested by at least two of the following:

1.     A) A marked incongruence between one’s experienced/expressed gender and primary and/or secondary sex characteristics (or in young adolescents, the anticipated secondary sex characteristics)

2.     B) A strong desire to be rid of one’s primary and/or secondary sex characteristics because of a marked incongruence with one’s experienced/expressed gender (or in young adolescents, a desire to prevent the development of the anticipated secondary sex characteristics)

3.     C) A strong desire for the primary and/or secondary sex characteristics of the other gender

4.     D) A strong desire to be of the other gender (or some alternative gender different from one’s designated gender)

5.     E) A strong desire to be treated as the other gender (or some alternative gender different from one’s designated gender)

6.     F) A strong conviction that one has the typical feelings and reactions of the other gender (or some alternative gender different from one’s designated gender)

B.    The condition is associated with clinically significant distress or impairment in social, occupational, or other important areas of functioning. Specify if:

1.     A) The condition exists with a disorder of sex development.

2.     B) The condition is post-transitional, in that the individual has transitioned to full-time living in the desired gender (with or without legalization of gender change) and has undergone (or is preparing to have) at least one sex-related medical procedure or treatment regimen—namely, regular sex hormone treatment or gender reassignment surgery confirming the desired gender (e.g., penectomy, vaginoplasty in natal males; mastectomy or phalloplasty in natal females).

 

One’s sex refers to the physical attributes that characterize biologic maleness or femaleness and is typically assigned or designated at birth based on the appearance of the external genitalia (or prior to birth based on sex chromosome complement and/or the appearance of the genitalia on the prenatal anatomy ultrasound).

 

Note that terminology in this field is constantly evolving, and for clinicians, it is important to ask individuals what terms they use to describe their gender identity, and what that term means to them. Table 3 includes suggestions on how to ask these questions.

 

Table 3. Suggested Ways of Asking About Name and Gender Identity

Name

“Is there a name you go by other than your legal name?”

“What name do you go by?”

“What would you like me to call you?”

Pronouns

“What pronouns do you use?”

“I’d like to use the pronouns that feel best to you. What pronouns would you like me to use?”

Model pronoun use: “Hello, my name is Dr. ____ I use ___ pronouns.”

Gender identity

“How do you identify your gender?”

“What does [gender identity term] mean to you?”

Suggestion for children: “Some kids tell me think of themselves as girls, some as boys, some as part girl and boy, or something entirely different. How do you think about yourself?”

Suggestion for adolescents: “There are lots of ways people think about their gender identity, how do you think of yours?”

 

GENDER IDENTITY DEVELOPMENT AND NATURAL HISTORY OF GENDER INCONGRUENCE  

 

Gender identity is multidimensional with biological, cultural, and environmental contributions (21,22). Studies of gender identity among individuals with differences/disorders of sex development underscore the influence of the hormonal milieu, and prenatal androgen exposure in particular, in gender development (23-26). There are also genetic influences, as some studies show concordance rates of gender dysphoria up to 39% among identical twins (27). Although studies have sought to identify genes associated with transgender identity, results have largely been inconsistent or inconclusive, with a possible role for genes related to sex steroids and their receptors (28-32).

 

In childhood, learning about gender starts early, and progress through many stages (33). In their review of gender development in childhood, Perry, Pauletti, and Cooper describe eight dimensions of gender identity: (1) gender self-categorization, (2) felt same-gender typicality, (3) felt other-gender typicality, (4) gender contentedness, (5) felt pressure for gender differentiation, (6) intergroup bias, (7) gender centrality, and (8) gender frustration. By age 18-24 months most children can categorize their own and others’ gender (34), and by age 6, have a developed gender identity (35). More individuals with a female sex designated at birth express dissatisfaction with their gender (36,37). This reflects the current sex ratio of individuals being referred to gender clinics, with more individuals with a female sex designated at birth currently referred, but the opposite being true prior to the 2000s (38-40).

 

Despite adolescence being a period of identity formation (41,42), there is a surprising lack of research on adolescent gender identity development. Development of identity is an individual and social process and shaped by external surroundings (42). However, there are also numerous psychological and biological factors that influence gender identity, as outlined in a review by Steensma and colleagues (43).

 

Overall, there is still much to learn about gender development among gender variant or nonconforming individuals. Prospective studies of children referred to gender clinics, primarily in Europe, show that less than a quarter of children will remain or meet criteria for gender identity disorder (the DSM-IV diagnosis prior to the DSM-5 gender dysphoria) after adolescence (44-47). In follow-up studies, the period of early adolescence/puberty, age 10-13 is critically important. There are three possible factors that contribute to an increase or decrease in gender discomfort and cross-gender identification: (1) physical puberty, (2) changing environment and being treated as their sex assigned at birth, and (3) the discovery of sexuality (47). More recent studies that reflect the rise in referral rates, and in various places around the world, will be critically important.

 

MEDICAL MANAGEMENT  

 

The WPATH Standards of Care (48) outline three categories of physical interventions for adolescents, indulging (1) fully reversible interventions, such as the use of gonadotropin releasing hormone (GnRH) agonists, medications to suppress menses (such as progestins), and medications to decrease the effects of androgens (such as spironolactone); (2) partially reversible interventions, including testosterone or estradiol; and (3) irreversible interventions, such as surgical procedures. Many individuals also seek care, including behavioral health consultation, for reversible interventions such as name, pronoun and gender marker change, discussing gender identity with friends, family and school, voice therapy, or wearables (including binders and packers to flatten the chest or give the appearance of male genitalia, respectively) (48).

 

It is important to note, as outlined in the WPATH Standards of Care (48), individuals who have gender variance or incongruence, but whom do not experience distress may not require clinical attention or intervention.  Furthermore, there is an increasing recognition that interventions should align with one’s individuals gender goals and gender embodiment (49).

 

Pubertal Blockade

 

The onset of puberty (gonadarche) is characterized by breast budding in people designated female at birth and by testicular enlargement to 4mL or greater in people designated male at birth, characterized as Tanner or Sexual Maturity Rating stage 2 (50,51). The average age of pubertal onset is age 10-11 years in someone designated female at birth (range 8-13 years, can be younger in African Americans), and 11-12 years in individuals designated male at birth (range 9-14 years). For individuals designated male at birth, external virilization typically starts around a testicular volume of 10 mL (11), voice drop at >8-10mL (52), and spermarche at 11-12 mL (53). In individuals designated female at birth, breast developmental progresses from stage 2 to 5 (fully developed) within 4-5 years and menarche typically occurs about 2-2.5 years after breast budding (54). Pubic hair and/or axillary hair and/or body odor reflect the onset of adrenarche or adrenal androgen production, which, by themselves are not indicative of central puberty (50,51). Height velocity increases during puberty and peaks about 2.5 years after the start of pubertal growth acceleration (55). An understanding of typical pubertal development and timing of external secondary sex characteristics is useful in counseling families about the timeliness and risk/benefit of GnRH agonist therapy to halt further pubertal progression. For example, towards the end of puberty or in post-pubertal individuals, GnRH agonist therapy may be used in certain circumstances for sex steroid suppression but would not block any pubertal changes, as these are complete.

 

GnRH agonists were first used in youth for the treatment of central precocious puberty in the 1980s (56). In 1998, Drs. Cohen-Kettenis and van Goozen in the Netherlands published the first report of a transgender patient treated with triptorelin, a GnRH agonist (57). The “Dutch model” of using pubertal suppression followed by gender affirming hormones (testosterone or estradiol) subsequently became incorporated into the WPATH and Endocrine Society standards of care (10,48). Their use became more widespread in the U.S. after publication of the 2009 Endocrine Society guidelines (10). The 2017 Endocrine Society guidelines suggest that “adolescents who meet diagnostic criteria for gender dysphoria/gender incongruence, fulfill criteria for treatment, and are requesting treatment should initially undergo treatment to suppress pubertal development”(11). The guidelines suggest beginning pubertal hormone suppression after the onset of the physical changes of puberty (Tanner Stage or Sexual Maturity Rating 2) for individuals who meet criteria, including being diagnosed with gender dysphoria, experienced worsening dysphoria with the onset of puberty, existing psychological, medical and/or social problems are addressed and the adolescent has sufficient mental capacity to consent to treatment (11). Treatment with a GnRH agonists suppresses gonadotropins (after an initial increase of gonadotropins) (58). There are also gonadotropin releasing hormone antagonists that immediately suppress gonadotropins, but are not available in children. GnRH agonists are typically administered as either an injection (IM or SQ) or as an implant (preparations listed in Table 4). Insurance coverage for this off-label, and costly therapy, varies (59). GnRH agonist treatment will pause or halt pubertal changes and may cause slight regression of breast tissue or testicular volume (11). On their own, these are reversible interventions, and if the individual decided that they wanted to progress through their endogenous puberty, these medications can be discontinued. During GnRH agonist treatment, the Endocrine Society recommends measurement of height, weight, sitting height, blood pressure, and Tanner stages every 3-6 months, measurement of LH, FSH, estradiol or testosterone, and 25OH vitamin D every 6-12 months, and bone density using dual-energy X-ray absorptiometry (DXA) and bone age x-ray of the left hand every 1-2 years (11).

 

Table 4. Hormonal Interventions for Transgender Adolescents

Pubertal blockade/inhibition of sex steroid secretion

GnRH agonist: inhibition of the hypothalamic-pituitary-gonadal access

Leuprolide acetate IM (1-, 3-, 4- or 6-mo preparations) or SQ (1-, 3-, 4- or 6-mo preparation)

Triptorelin IM (4-, 12- or 24-week preparation)

Histrelin acetate SQ implant (one-yearly dosing, although reports of longer effectiveness)

Medroxyprogesterone acetate: inhibition of the hypothalamic-pituitary-gonadal access and direct inhibition of gonadal steroidogenesis

Orally (up to 40 mg/day) or IM (150 mg every 3 mo, may be given more frequently for suppression of sex steroids)

Inhibition of testosterone secretion or action

Spironolactone: inhibition of testosterone synthesis and action

Titrate up to 10-300 mg/day orally (typically in divided doses)

Cyproterone acetate: inhibition of testosterone synthesis and action (not available in US)

25-50 mg/day orally

Finasteride: inhibition of type II 5 α-reductase, blocks conversion of testosterone to 5 α-dihydrotestosterone

2.5-5 mg/day orally

Bicalutamide: androgen receptor blockade

50 mg/day orally

Sex steroids

Estrogen/17β-estradiol

Oral/sublingual: start with lower doses for pubertal induction, titrate up to adult doses 2-6 mg/day

Transdermal: start with lower doses for pubertal induction, titrate up to adult doses 0.025-0.2 mg/day (patches are typically once or twice weekly)

Parenteral: estradiol valerate (5-30 mg every 2 weeks) or cypionate (2-10 mg IM every week)

Testosterone

Parenteral IM or SQ testosterone cypionate or enanthate (start at 12.5 mg/week or 25 mg q2 week with gradual increases to 50-100 mg/week or 100-200 mg every 2 weeks)

Transdermal (typically after full adult dose has been achieved parenterally): patch (2.5-7.5 mg/day or 1% or 1.6% gel

Note that all medications are currently off-label for gender non-conforming/transgender youth. Note that certain GnRH preparations are approved in children for central precocious puberty and other formulations are approved for adults only, with off-label use in children. Different formulations are available in different countries. This table was adapted from the following references (11,178). Note that some centers/providers also use GnRH agonists for testosterone blockade in older adolescents and/or adults. GnRH: gonadotropin releasing hormone, IM: intramuscular, SQ: subcutaneous

 

Recent reviews of puberty blockade have been published (60-62). Small studies have demonstrated effectiveness of GnRH agonist treatment for suppression of the hypothalamic-pituitary-gonadal axis in transgender youth (63). Studies, primarily in Europe, have demonstrated improvements in psychological functioning, behavioral/emotional problems, and depressive symptoms during GnRH agonist treatment in transgender youth (64,65). A recent systemic review found that GnRH agonist therapy is associated with decreased suicidality in adulthood, improved affect and psychological functioning, and improved social life (61).

 

Potential risks of GnRH agonist therapy include impacts on growth, bone health, body composition, fertility, and neurodevelopment, as well as difficulties accessing treatment due to cost and insurance coverage (61,62,66). GnRH agonist use in TGD youth is associated with increased body fat and decrease in lean mass after initiation (67,68), and compared to age- and BMI-matched control youth (69), and may also have an adverse effect on insulin sensitivity (69). If GnRH agonists are started prior to skeletal maturity, they will decrease skeletal advancement during monotherapy due to suppression of sex steroids, which are necessary for growth plate closure (70,71). There is a dearth of research on growth trajectories during treatment with a GnRH agonist in this population. One multicenter study in the U.S. showed that transgender youth treated with GnRH therapy have growth velocity similar to prepubertal children, but those who start GnRH agonist treatment later in puberty have growth velocity below the prepubertal range (72). The growth spurt and skeletal advancement will progress either when exogenous testosterone or estradiol are started, or if the GnRH agonists are discontinued and the individual progresses through their endogenous puberty. There is a growing body of research of bone health in transgender individuals, as well as the impact of GnRH agonists and later gender affirming hormones on bone health. Studies in the Netherlands have demonstrated decreased bone turnover, and a decrease in bone mineral apparent density Z-scores of the lumbar spine in transwomen after initiation of GnRH agonist therapy (73). However, studies in the U.S. (74), United Kingdom (75) and Netherlands (73,76) have also shown decreased bone mineral density Z-scores determined by DXA are low prior to treatment with GnRH agonist, and some studies showing Z-scores did not completely normalize with sex hormone treatment (73,76). In the U.S., the individuals with lower baseline bone mineral density Z-scores also reported less physical activity, an area warrants further research (74).

 

Overall, there is a paucity of research on neurodevelopment and a recent consensus parameter was published with recommended research methodologies to evaluate the neurodevelopmental effects of puberty suppression in this population (77). There is also very little research on sexual function and future surgical options among individuals who received early puberty blockade. Recently, there has been a call for more information to better inform the impacts on future sexual function(78,79) (as GnRH agonists limit penile and testicular growth/size (80)) and on implications for future surgical intervention for those individuals pursuing vaginoplasty (as the scrotal tissue is used to construct the vagina) (80). Finally, treatment with GnRH agonists will impair spermatogenesis and oocyte maturation temporarily, and the Endocrine Society recommends fertility counseling (11). Treatment may be delayed to preserve fertility, but many individuals do not choose this, as delay will also cause further progression of unwanted secondary sex characteristics (11). There are limited options available for early tissue cryopreservation, an area that is gaining more attention (81).

 

If GnRH analogues are not available or are cost prohibitive, medroxyprogesterone may be used as an alternative agent for pubertal suppression (Table 4) (11,48). At high doses, medroxyprogesterone inhibits the pituitary-gonadal axis and suppresses testosterone (82-84).  Medroxyprogesterone was used for treatment of precocious puberty in the 1960s and 70s (85-87). It is typically safe, although may have some side effects, including due to the estrogenic effects (bloating, nausea/vomiting, breast fullness, breakthrough bleeding for those menstruating, irritability, headache, hypertension), progestational effects (headache, breast pain/tenderness, hypertension), and androgenic effects (acne, oily skin, weight gain, hirsutism, fatigue, depression) (88). At extremely high doses (100 mg four times a day), it may cause Cushing’s syndrome, adrenal insufficiency, and diabetes (89). There is one small study of medroxyprogesterone in transgender youth demonstrating effective sex steroid suppression with doses of oral medroxyprogesterone 10-30 mg BID or 150 mg IM every 2-3 months (90).

 

Gender Affirming Hormone Therapy

 

Gender affirming hormone therapy (GAHT) refer to hormones that induce secondary sex characteristics to align the body with one’s gender identity. The Endocrine Society recommends treatment with sex steroids (testosterone or estradiol) “using a gradually increasing dose schedule after a multidisciplinary team of medical and mental health professionals has confirmed the persistence of gender dysphoria/gender incongruence and sufficient mental capacity to give informed consent, which most adolescents have by age 16 years” (full criteria in guidelines) (11). However, they also state that “there may be compelling reasons to initiate sex hormone treatment prior to age 16 years in some adolescents” (11). The WPATH Standards of Care, 8th version are forthcoming, but in the current 7th version, the criteria for hormone therapy are: “(1) persistent, well-documented gender dysphoria; (2) capacity to make a fully informed decision and to consent for treatment; (3) age of majority in a given country; (4) if significant medical or mental concerns are present, they must be reasonably well-controlled” (48). It is recommended, that for adolescents who have not reached the age of majority in their country, that consent from all parents or medical decision-makers is obtained prior to starting this partially irreversible therapy.

 

Feminizing Hormone Therapy  

 

ESTRADIOL THERAPY  

 

For eligible adolescents, the Endocrine Society recommends a gradually increasing dose schedule of oral or transdermal 17β-estradiol (11). This will cause feminization of the body, with expected effects including body fat redistribution, decreased muscle mass/strength, softening of the skin/decreased oiliness, decreased libido/erections, breast growth, decreased testicular volume, decreased sperm production, and thinning and slowed growth of body and facial hair occurring one to several months after treatment with maximum effects generally about 2-3 years or more into treatment (11,48). For younger individuals, the Endocrine Society recommends starting oral estradiol at a dose of 5 µg/kg/day and increasing doses every 6 months up to a dose of 2-6 mg/day for an adult (11). In post-pubertal individuals, the starting dose may be higher and titrated more quickly (start at 1 mg/day for 6 months and increase to 2 mg/day orally) (11). For transdermal estradiol, it is recommended to start at a dose of 6.25-12.5 µg/24 hours and increase the dose every 6 months to an adult dose of 50-200 µg/24 hours. During induction of puberty, it is recommended to measure height, weight, sitting height, blood pressure, and Tanner stages every 3-6 months, and measure prolactin, estradiol and 25OH vitamin D every 6-12 months (11). Additionally, DXA and bone age (if clinically indicated or a growing patient) is recommended every 1-2 years (11).

 

Potential Adverse Effects of Estradiol Therapy

 

Risks with estradiol therapy as outlined in the Endocrine Society guidelines include thromboembolic disease, macroprolactinoma, breast cancer, coronary artery disease, cerebrovascular disease, cholelithiasis, and hypertriglyceridemia (11).

 

In adults, studies using three large cohorts have shown an increased risk of myocardial infarction and venous thromboembolism. In Europe, transgender women on estradiol therapy have a higher risk of stroke and venous thromboembolism than both cisgender reference women and men, and a higher risk of myocardial infarction than cisgender women (but not men) (91). In the U.S., two large cohorts have been used to examine outcomes, the Kaiser STRONG cohort and self-report data from the Behavioral Risk Factor Surveillance System (BRFSS). Data on hormone treatment is not collected in BRFSS. Transgender women in BRFSS were more likely (>2-fold increase risk) to have a history of myocardial infarction than cisgender women (but not men) (92,93). In the Kaiser STRONG cohort (94), both prevalent and incident type 2 diabetes was more common in the transfeminine cohort compared to cisgender females (95). In a meta-analysis commissioned by the Endocrine Society to accompany the 2017 updated guidelines, transgender women on estradiol therapy had increased triglycerides, but no changes in other lipid parameters (96). There were few reports of myocardial infarction, stroke, venous thromboembolism or death (96). It is well-known that transgender women on estradiol therapy have increases in body weight and fat and decreases in lean body mass (97). Estradiol therapy is associated with increases in lumbar spine bone mineral density compared to baseline (98).

 

In youth, there is a growing body of literature on the effects of GAHT, particularly on cardiometabolic health. TGD youth on estradiol have changes in HDL, aspartate aminotransferase, potassium, prolactin, and hemoglobin after about two years (99). One study found that transgender females on estradiol therapy were more insulin resistant than matched cisgender males (100). The presence of obesity attenuates the beneficial effect of estradiol on HDL (101). There are also studies investigating baseline differences between TGD youth and cisgender controls prior to hormone therapy, with recent studies showing TGD youth have lower HDL and low bone mineral density (72,102).

 

Testosterone Blockade/Suppression

 

There are many options for blockade and/or suppression of testosterone (all off-label use, Table 4). When available and affordable, some centers utilize GnRH agonists for suppression of testosterone. For example, in the United Kingdom, GnRH analogues are heavily subsidized (103). There are also many antiandrogens available, and a systemic review of options has recently been published (104). Spironolactone is widely available, inexpensive, and commonly used in the U.S. and Australia. Spironolactone is a weak androgen receptor antagonist (105,106), weak progesterone receptor agonist, and weak estrogen receptor agonist (104). It also partially inhibits 17α-hydroxylase/17,20 lyase, which are involved in testosterone synthesis (107). Even at high doses, spironolactone does not cause a significant reduction in serum total testosterone concentration (108). Although the combination of spironolactone with estradiol does appear to suppress testosterone in transgender women (109). Side effects include irregular menses (only for people who are menstruating, not a consideration for transgender women), hypotension, polyuria, and hyperkalemia (110,111).

 

Cyproterone acetate is available in Europe and Australia, but not in the U.S. and is a moderate androgen receptor antagonist, strong progesterone receptor agonist, and does not have any estrogen receptor activity but does suppress the hypothalamic pituitary gonadal axis (104). Cyproterone acetate has been associated with increased risk of meningiomas (112) and prolactinomas (113). Other side effects include weight gain, headache, gastrointestinal disorders, mood effects, and edema (114).

 

Nonsteroidal antiandrogens, such as bicalutamide have also been used at some centers. Bicalutamide has strong androgen receptor antagonist activity and does not have any estrogen or progesterone agonist activity (104). It does not cause a reduction in testosterone concentrations. There is some feminization, thought to be due to increased aromatization of testosterone to estradiol (115). There is currently one published study of the use of bicalutamide in transgender adolescents as an alternative to GnRH agonists (115). In that study, hepatic enzymes remained normal and there were no adverse effects, however, effectiveness and the potential risk of liver toxicity needs to be examined in larger studies.

 

Finally, 5-alpha reductase inhibitors, such as finasteride, block conversion of testosterone to dihydrotestosterone. These are not recommended by the Endocrine Society due to adverse effects (11), but the WPATH guidelines state, “these medications have beneficial effects on scalp hair loss, body hair growth, sebaceous glands, and skin consistency” (48). Side effects include sexual dysfunction and decreased muscle (which may be perceived as a risk or benefit in this population), anhedonia, and trouble concentrating (116).

 

Overall, the selection of which agent alone or in combination with estradiol depends on many factors including patient age, country, insurance coverage, cost, goals of care, and tolerability of side effects (e.g. severe and fatal hepatotoxicity has been reported with cyproterone acetate and bicalutamide (117)). Further studies are needed to determine superiority for relevant patient outcomes including body composition, breast development, facial and body hair (104).

 

Masculinizing Hormone Therapy

 

TESTOSTERONE  

 

For eligible adolescents, the Endocrine Society recommends a gradually increasing dose schedule of testosterone (typically injectable IM or SQ) (11). This will cause masculinization of the body, with expected effects including skin oiliness/acne, facial/body hair growth, scalp hair loss, increased muscle mass/strength, body fat redistribution, cessation of menses, clitoral enlargement, vaginal atrophy, and deepened voice with onset occurring one to several months after treatment with maximum effects generally about 2-5 years or more into treatment (11,48). For younger individuals, the Endocrine Society recommends starting injectable testosterone esters at a dose of 25 mg/m2 IM or SQ every 2 weeks and increasing every 6 months up to an adult dose of 100-200 mg every 2 weeks (11). In post-pubertal individuals, the starting dose may be higher and titrated more quickly (start at 75 mg every 2 weeks for 6 months and increase to 125 mg every 2 weeks) (11). Subcutaneous testosterone is gaining in popularity, and has shown to be effective and preferred by patients (118-120). Pharmacokinetic studies of weekly subcutaneous testosterone injections show that steady state is approached after the third dose, and that serum concentrations stay relatively constant throughout the week between doses (121). Finally, SQ testosterone doses may be lower than those delivered IM, with two studies reporting doses of 50-80 mg/week to achieve target testosterone concentrations in adults or older adolescents (118,119). During induction of puberty, it is recommended to measure height, weight, sitting height, blood pressure, and Tanner stages every 3-6 months, and measure hemoglobin/hematocrit, lipids, testosterone, and 25OH vitamin D every 6-12 months (11). Additionally, DXA and bone age (if clinically indicated or a growing patient) is recommended every 1-2 years (11).

 

The most common adverse effect of testosterone is erythrocytosis/polycythemia (hematocrit >50%) (11). Other risks as outlined in the Endocrine Society guidelines include liver dysfunction, coronary artery disease, cerebrovascular disease, hypertension, and breast or uterine cancer (11).

 

In adults, studies using three large cohorts have shown conflicting results. In Europe, transgender men on testosterone therapy have a higher risk of myocardial infarction than cisgender women (but not men) and no increased risk of stroke or venous thromboembolism compared to reference populations (91). Transgender men in the 2015 U.S. BRFSS survey had no increased risk of hypertension, myocardial infarction, stroke, angina/coronary heart disease compared to cisgender men or women (92). However, another analysis of BRFSS data (years 2014-2017) reported a >2-fold increase risk of myocardial infarction compared to cisgender men and 4-fold increase compared to cisgender women (93). In the Kaiser STRONG cohort, there was no increased risk of type 2 diabetes among transgender men compared to cisgender men (95). In a meta-analysis, testosterone therapy in transgender men was associated with increases in serum triglycerides and low-density lipoprotein cholesterol (LDL-C) concentrations and decreases in high-density lipoprotein cholesterol (HDL) (96). Testosterone therapy in transgender men is known to result in increased body weight and lean mass and decreased body fat (97). In meta-analyses, testosterone therapy is not associated with significant changes in bone mineral density (98,122). There is a recent position statement from the International Society for Clinical Densitometry on bone densitometry in TGD individuals (123).

 

Among TGD youth starting testosterone therapy, there is an increase in BMI and decrease in HDL (124). The decrease in HDL is exacerbated by obesity (101). Other studies have found that testosterone treatment in TGD youth is associated with statistical, but not clinically significant increases in triglycerides, alanine aminotransferase, potassium, and hemoglobin (99).

 

Non-Binary Care

 

Non-binary or gender non-conforming individuals represent a growing proportion of patients presenting to gender clinics and may have additional challenges accessing healthcare (125). Limited studies have reported worse mental and physical health among individuals who identify as gender non-conforming compared to matched controls (92). An individualized approach to understand the individual’s gender identity, sources of dysphoria (if any), and gender goals are important. Some individuals may desire reversible interventions such as menstrual suppression, others may request certain hormones and/or surgical interventions as a part of their gender goals. The 8th version of the WPATH standards of care will include a chapter on non-binary care.

 

Menstrual Management  

 

Many transmasculine and non-binary individuals who are designated female at birth seek medical attention or desire interventions for menstrual management (126). Some also utilize these methods for contraception. It is important to ask individuals about their individual goals, as well as their sexual orientation, partners (including sex assigned at birth and what body parts they currently have), and types of sex they are engaging in. These factors can guide choice of intervention for menstrual management and/or contraception. An overview of options is in Table 5.  Progestin-only methods, including norethindrone or depo medroxyprogesterone are particularly popular choices among this population (126). Review of options for menstrual management and contraceptive options for transgender individuals was recently published (127). For those patients wishing to and eligible for testosterone therapy, menses suppression typically is achieved within 6-12 months of the start of testosterone therapy (128).

 

Table 5. Options for Menstrual Suppression/Management

Combined hormonal contraceptives (pills, patch, ring)

Progestins

   Norethindrone acetate (5-15 mg/day orally)

   Medroxyprogesterone acetate (150 mg IM every 3 months)

   Etonogestrel implant

   Levonorgestrel IUD

IUD: intrauterine device

 

SURGICAL MANAGEMENT

 

Surgeries that impact fertility are generally not available until the individual has reached the age of majority in their country. There are a wide variety of surgical options for transgender adults (and some options, primarily chest surgery, for adolescents), and this has recently been reviewed (129). Physicians (including surgeons and non-surgeons) and behavioral health providers should be aware of the criteria needed for each surgical procedure, including whether social transition is recommended, whether hormonal therapy is needed (and length), and how many referral letters are needed and by whom (48,130). In general, a documentation of persistent gender dysphoria by a qualified mental health provider is a requirement for surgery (48). Additionally, guidelines may change over time (the 8th version of the WPATH standards of care are coming soon) and may vary by location (country/state) and insurance coverage. Table 6 summarizes the various gender affirming surgical options. Genital surgery or removal of the gonads is generally not performed until the individual is the age of majority in a given country (age 18 years or older in the U.S.). Individuals younger than 18 may be eligible for chest/breast surgery, with consent from medical decision-makers. The WPATH Standards of Care state for mastectomy/chest masculinizing surgery or for breast augmentation surgery, the individual must have “(1) persistent, well-documented gender dysphoria, (2) capacity to make s fully informed decision and to consent for treatment, (3) age of majority in a given country, (4) if significant medical or mental health concerns are present, they must be reasonably well controlled.” Although masculinizing hormone therapy (testosterone) is not a prerequisite for chest masculinizing surgery in the WPATH guidelines, it is recommended (although not an explicit criterion) that individuals on feminizing hormone therapy be on for a minimum of 12 months prior to breast augmentation surgery for better aesthetic results (48). However, the Endocrine Society Guidelines recommend 2 years of testosterone therapy prior to mastectomy/chest masculinizing surgery. Neither the WPATH or Endocrine Society guidelines recommend a specific age cutoff, but “suggest that clinicians determine the timing of breast surgery for transgender males based upon the physical and mental health status of the individual” (11).

 

Table 6. Gender Affirming Surgical Options

Feminizing surgeries

Breast augmentation

Increasing the size of the breasts

Facial feminization surgery

May include: forehead feminization, rhinoplasty, periorbital rejuvenation, rhytidectomy (face lift), cheek augmentation, rhinoplasty, lip feminization, gonial angle shave, genioplasty

Genital surgery/vaginoplasty

May include penectomy, orchiectomy, surgical creation of a vagina (penile inversion, intestinal conduit), clitoroplasty, labiaplasty

Orchiectomy

Removal of testes

Tracheal shave

Thyroid cartilage shave

Masculinizing surgeries

Chest masculinizing surgery (mastectomy)

Removal of breast tissue

Facial masculinization surgery

Rhinoplasty, gonial implants, genioplasty

Hysterectomy, salpingectomy, oophorectomy

Removal of uterus and/or fallopian tubes, and/or ovaries

Metoidioplasty

Creation of a phallus using existing genital tissue 

Phalloplasty

Construction of phallus, glansplasty, urethroplasty, erectile prosthesis, scrotoplasty, testicular implants

 

MENTAL HEALTH

 

Recently studies have demonstrated a strikingly high prevalence of behavioral health diagnoses among youth diagnosed with gender dysphoria (up to 60%) (131,132). Studies evaluating behavioral health outcomes among TGD youth have most frequently demonstrated disproportionate anxiety (132-134), depression (133-136), suicidality (19,132-134,137), self-harm (132-135), and substance use problems (19). Large surveys of TGD individuals in the U.S. have shown that 40% of adults (138) and 35% of youth (19) have attempted suicide. Poor behavioral health outcomes may be conceptualized as the result of complex and layered socio-cultural and political factors that impact TGD youth (15,139). Risk factors that are likely to impact overall mental health for TGD individuals include minority stress (e.g., victimization, discrimination) (19,140), gender dysphoria and appearance congruence (141), feelings of isolation, inadequate family support (142), emotional/social isolation (143), lack of autonomy over decision making (143), barriers to accessing gender affirming care (143-145), employment discrimination (143), and limited financial resources (143). In the Youth Risk Behavior Survey, TGD youth were two to six times more likely to be victimized, including experiencing sexual dating violence, experiencing physical dating violence, being bullied at school, being electronically bullied, feeling unsafe during travel to or from school, and being forced to have sexual intercourse (19).

 

The co-occurrence of autism spectrum disorders (ASD) and gender dysphoria is a growing area of interest (132,146-150). A recent meta-analysis found that the prevalence of ASD among those with GD has ranged from 6% to 68% depending on the methodology of the study (151). In a large 2020 study, TGD individuals were 3.0 to 6.4 times more likely to be diagnosed with ASD than their cisgender counterparts (136). Other samples have shown that youth with gender dysphoria are about 2-3x as likely to have a diagnosis of ASD than their matched cisgender counterparts (131,132). The exact link between GD and ASD is not known, but factors contributing may include: symptom overlap between the two diagnoses, misclassification due to symptom overlap, children with ASD may be more likely to express their gender identity and dysphoria, or they may be more likely to be referred to care to be diagnosed with either GD or ASD (152).

 

Protective factors including social support (153,154), parental support/affirmation of gender identity (155,156), higher self-esteem (153), resiliency (153,157), and access to affirming care (144,158,159) have improved well-being and decreased mental health distress. Access to gender-affirming interventions, including hormone therapy and surgery, has been shown to improve gender dysphoria, psychological symptoms and quality of life in small samples and meta-analyses (28). Recent studies have shown that those who were older at presentation have worse mental health than those who presented to care at a younger age (160) and those who had access to GnRH agonists had lower lifetime odds of suicidal ideation than those who did not have access (144).

 

Finally, there are many other important topics that impact the care of transgender individuals that are beyond the scope of this chapter including dermatologic considerations and hair loss (161-163), chest binding (164), sexual health (79), HIV prevention and treatment (165-167), fertility (81), sleep (168), athletic performance and sports participation (169), eating disorders (170,171), homelessness, the impact of family support, and the underpinnings of links between gender diversity and neurodiversity (172).

 

CONCLUSION

 

An improved understanding of the variety of individual gender trajectories is needed, as well as how best to individualize care, how to improve mental health, and minimize risks of medical intervention. Large, multi-center, prospective cohorts, as are currently established in the U.S. (94,173) and Europe (174), will help answer some of these important questions. And community-based participatory research and hearing the voices of individuals from the community about their own research priorities and dissemination of results is of utmost importance. There is also much to be learned about the impact of early GnRH agonist therapy on growth, bone health, physical development, long-term health, mental health, cognitive development, and overall wellbeing. Finally, an improved understanding on the impact of other stressors including minority stress and depression on overall health (175,176) for TGD persons is needed. The American Heart Association published a scientific statement with recommendations to assess and address cardiovascular health among TGD people (177).

 

REFERENCES  

 

  1. David K, Dingemanse E, Freud J, Laqueur E. On crystalline male hormone from testicles (testosterone). Hoppe Seylers Z Physiol Chem 1935; 233:281
  2. Veler CD, Thayer S, Doisy EA. The preparation of the crystalline follicular ovarian hormone: theelin. Journal of Biological Chemistry 1930; 87:357-371
  3. Butenandt A. Über „Progynon “ein krystallisiertes weibliches Sexualhormon. Naturwissenschaften 1929; 17:879-879
  4. Slotta K, Ruschig H, Fels E. Reindarstellung der Hormone aus dem Corpus luteum (II. Mitteil.). Berichte der deutschen chemischen Gesellschaft (A and B Series) 1934; 67:1624-1626
  5. Wintersteiner O, Allen WM. Crystalline progestin. Journal of Biological Chemistry 1934; 107:321-336
  6. Elbe L, Jacobsen ELH. Man into Woman: An Authentic Record of a Change of Sex. The True Story of the Miraculous Transformation of the Danish Painter, Einar Wegener (Andreas Sparre). Jarrolds.
  7. Huelke H. Ein Transvestit. Z Kriminalistische Wissen 1949; 3:91-92
  8. Hamburger C, Stürup GK, Dahl-Iversen E. Transvestism: Hormonal, psychiatric, and surgical treatment. Journal of the American Medical Association 1953; 152:391-396
  9. Benjamin H. The transsexual phenomenon. Ace Publishing Company.
  10. Hembree WC, Cohen-Kettenis P, Delemarre-van de Waal HA, Gooren LJ, Meyer WJ, III, Spack NP, Tangpricha V, Montori VM. Endocrine treatment of transsexual persons: an Endocrine Society clinical practice guideline. J Clin Endocrinol Metab 2009; 94:3132-3154
  11. Hembree WC, Cohen-Kettenis PT, Gooren L, Hannema SE, Meyer WJ, Murad MH, Rosenthal SM, Safer JD, Tangpricha V, T'Sjoen GG. Endocrine Treatment of Gender-Dysphoric/Gender-Incongruent Persons: An Endocrine Society Clinical Practice Guideline. J Clin Endocrinol Metab 2017; 102:3869-3903
  12. Gillig TK, Rosenthal EL, Murphy ST, Folb KL. More than a media moment: The influence of televised storylines on viewers’ attitudes toward transgender people and policies. Sex Roles 2018; 78:515-527
  13. Equality Federation.https://www.equalityfederation.org/state-legislation. Accessed 1/14/22.
  14. Rafferty J. Ensuring Comprehensive Care and Support for Transgender and Gender-Diverse Children and Adolescents. Pediatrics 2018; 142:e20182162
  15. Association AP. Guidelines for psychological practice with transgender and gender nonconforming people. American Psychologist 2015; 70:832-864
  16. Skordis N, Butler G, de Vries MC, Main K, Hannema SE. ESPE and PES International Survey of Centers and Clinicians Delivering Specialist Care for Children and Adolescents with Gender Dysphoria. Horm Res Paediatr 2018; 90:326-331
  17. Korpaisarn S, Safer JD. Gaps in transgender medical education among healthcare providers: A major barrier to care for transgender persons. Reviews in Endocrine and Metabolic Disorders 2018; 19:271-275
  18. Herman JL, Flores, A.R., Brown, T.N.T., Wilson, B.D.M., & Conron, K.J. . Age of individuals who identify as transgender in the United States. . Los Angeles, CA: The Williams Institute 2017;
  19. Johns MM, Lowry R, Andrzejewski J, Barrios LC, Demissie Z, McManus T, Rasberry CN, Robin L, Underwood JM. Transgender Identity and Experiences of Violence Victimization, Substance Use, Suicide Risk, and Sexual Risk Behaviors Among High School Students - 19 States and Large Urban School Districts, 2017. MMWR Morbidity and mortality weekly report 2019; 68:67-71
  20. Association AP. Diagnostic and statistical manual of mental disorders (DSM-5®). American Psychiatric Pub.
  21. Perry DG, Pauletti RE, Cooper PJ. Gender identity in childhood: A review of the literature. International Journal of Behavioral Development 2019; 43:289-304
  22. Fausto-Sterling A. Sex/gender: Biology in a social world. Routledge.
  23. Berenbaum SA, Meyer-Bahlburg HF. Gender development and sexuality in disorders of sex development. Horm Metab Res 2015; 47:361-366
  24. Dessens AB, Slijper FM, Drop SL. Gender dysphoria and gender change in chromosomal females with congenital adrenal hyperplasia. Arch Sex Behav 2005; 34:389-397
  25. Meyer-Bahlburg HF, Dolezal C, Baker SW, Ehrhardt AA, New MI. Gender development in women with congenital adrenal hyperplasia as a function of disorder severity. Arch Sex Behav 2006; 35:667-684
  26. Meyer-Bahlburg HF, Dolezal C, Baker SW, Carlson AD, Obeid JS, New MI. Prenatal androgenization affects gender-related behavior but not gender identity in 5-12-year-old girls with congenital adrenal hyperplasia. Arch Sex Behav 2004; 33:97-104
  27. Heylens G, De Cuypere G, Zucker KJ, Schelfaut C, Elaut E, Vanden Bossche H, De Baere E, T'Sjoen G. Gender identity disorder in twins: a review of the case report literature. The journal of sexual medicine 2012; 9:751-757
  28. Fernández R, Esteva I, Gómez-Gil E, Rumbo T, Almaraz MC, Roda E, Haro-Mora JJ, Guillamón A, Pásaro E. Association study of ERβ, AR, and CYP19A1 genes and MtF transsexualism. The journal of sexual medicine 2014; 11:2986-2994
  29. Henningsson S, Westberg L, Nilsson S, Lundström B, Ekselius L, Bodlund O, Lindström E, Hellstrand M, Rosmond R, Eriksson E, Landén M. Sex steroid-related genes and male-to-female transsexualism. Psychoneuroendocrinology 2005; 30:657-664
  30. Hare L, Bernard P, Sánchez FJ, Baird PN, Vilain E, Kennedy T, Harley VR. Androgen receptor repeat length polymorphism associated with male-to-female transsexualism. Biol Psychiatry 2009; 65:93-96
  31. Lombardo F, Toselli L, Grassetti D, Paoli D, Masciandaro P, Valentini F, Lenzi A, Gandini L. Hormone and genetic study in male to female transsexual patients. J Endocrinol Invest 2013; 36:550-557
  32. Ujike H, Otani K, Nakatsuka M, Ishii K, Sasaki A, Oishi T, Sato T, Okahisa Y, Matsumoto Y, Namba Y, Kimata Y, Kuroda S. Association study of gender identity disorder and sex hormone-related genes. Prog Neuropsychopharmacol Biol Psychiatry 2009; 33:1241-1244
  33. Kohlberg LA. A cognitive-developmental analysis of children's sex role concepts and attitudes. The development of sex differences 1966:82-173
  34. Eichstedt JA, Serbin LA, Poulin-Dubois D, Sen MG. Of bears and men: Infants’ knowledge of conventional and metaphorical gender stereotypes. Infant Behav Dev 2002; 25:296-310
  35. Diamond LM, Butterworth M. Questioning gender and sexual identity: Dynamic links over time. Sex roles 2008; 59:365-376
  36. Carver PR, Yunger JL, Perry DG. Gender identity and adjustment in middle childhood. Sex roles 2003; 49:95-109
  37. Egan SK, Perry DG. Gender identity: a multidimensional analysis with implications for psychosocial adjustment. Developmental psychology 2001; 37:451
  38. Zucker K, Aitken M. Sex ratio of transgender adolescents: A meta-analysis. Paper presented at: 3rd biennal EPATH Conference Inside Matters. On Law, Ethics and Religion2019
  39. Arnoldussen M, Steensma TD, Popma A, van der Miesen AI, Twisk JW, de Vries AL. Re-evaluation of the Dutch approach: are recently referred transgender youth different compared to earlier referrals? European child & adolescent psychiatry 2020; 29:803-811
  40. Aitken M, Steensma TD, Blanchard R, VanderLaan DP, Wood H, Fuentes A, Spegg C, Wasserman L, Ames M, Fitzsimmons CL. Evidence for an altered sex ratio in clinic‐referred adolescents with gender dysphoria. The journal of sexual medicine 2015; 12:756-763
  41. Erikson EH. Identity: Youth and crisis. WW Norton & company.
  42. Marcia JE. Development and validation of ego-identity status. Journal of personality and social psychology 1966; 3:551
  43. Steensma TD, Kreukels BP, de Vries AL, Cohen-Kettenis PT. Gender identity development in adolescence. Horm Behav 2013; 64:288-297
  44. Drummond KD, Bradley SJ, Peterson-Badali M, Zucker KJ. A follow-up study of girls with gender identity disorder. Dev Psychol 2008; 44:34
  45. Zucker KJ. On the “natural history” of gender identity disorder in children. J Am Acad Child Adolesc Psychiatry 2008; 47:1361-1363
  46. Wallien MSC, Cohen-Kettenis PT. Psychosexual outcome of gender-dysphoric children. J Am Acad Child Adolesc Psychiatry 2008; 47:1413-1423
  47. Steensma TD, Biemond R, de Boer F, Cohen-Kettenis PT. Desisting and persisting gender dysphoria after childhood: A qualitative follow-up study. Clin Child Psychol Psychiatry 2011:1359104510378303
  48. Coleman E, Bockting W, Botzer M, Cohen-Kettenis P, DeCuypere G, Feldman J, Fraser L, Green J, Knudson G, Meyer WJ, Monstrey S, Adler RK, Brown GR, Devor AH, Ehrbar R, Ettner R, Eyler E, Garofalo R, Karasic DH, Lev AI, Mayer G, Meyer-Bahlburg H, Hall BP, Pfäfflin F, Rachlin K, Robinson B, Schechter LS, Tangpricha V, van Trotsenburg M, Vitale A, Winter S, Whittle S, Wylie KR, Zucker K. Standards of Care for the Health of Transsexual, Transgender, and Gender-Nonconforming People. World Professional Association for Transgender Health (WPATH).
  49. Keo-Meier CE, Ehrensaft DE. The gender affirmative model: An interdisciplinary approach to supporting transgender and gender expansive children. American Psychological Association.
  50. Marshall WA, Tanner JM. Variations in the pattern of pubertal changes in boys. Arch Dis Child 1970; 45:13-23
  51. Marshall WA, Tanner JM. Variations in pattern of pubertal changes in girls. Arch Dis Child 1969; 44:291-303
  52. Harries ML, Walker JM, Williams DM, Hawkins S, Hughes I. Changes in the male voice at puberty. Archives of disease in childhood 1997; 77:445-447
  53. Nielsen CT, SKAKKEBÆK NE, Richardson DW, Darling JA, Hunter WM, Jørgensen M, Nielsen A, Ingerslev O, Keiding N, Müller J. Onset of the release of spermatozia (supermarche) in boys in relation to age, testicular growth, pubic hair, and height. The Journal of Clinical Endocrinology & Metabolism 1986; 62:532-535
  54. Kleigman RM, Behrman RE, Jenson HB, Stanton B. Nelson textbook of pediatrics. Stanton, chapter 2007;
  55. Abbassi V. Growth and normal puberty. Pediatrics 1998; 102:507-511
  56. Mansfield MJ, Beardsworth DE, Loughlin JS, Crawford JD, Bode HH, Rivier J, Vale W, Kushner DC, Crigler JF, Jr., Crowley WF, Jr. Long-term treatment of central precocious puberty with a long-acting analogue of luteinizing hormone-releasing hormone. Effects on somatic growth and skeletal maturation. N Engl J Med 1983; 309:1286-1290
  57. Cohen-Kettenis PT, Van Goozen S. Pubertal delay as an aid in diagnosis and treatment of a transsexual adolescent. Eur Child Adolesc Psychiatry 1998; 7:246-248
  58. Roth CL, Brendel L, Rückert C, Hartmann K. Antagonistic and agonistic GnRH analogue treatment of precocious puberty: tracking gonadotropin concentrations in urine. Horm Res 2005; 63:257-262
  59. Stevens J, Gomez-Lobo V, Pine-Twaddell E. Insurance coverage of puberty blocker therapies for transgender youth. Pediatrics 2015; 136:1029-1031
  60. Panagiotakopoulos L, Chulani V, Koyama A, Childress K, Forcier M, Grimsby G, Greenberg K. The effect of early puberty suppression on treatment options and outcomes in transgender patients. Nat Rev Urol 2020; 17:626-636
  61. Rew L, Young CC, Monge M, Bogucka R. Review: Puberty blockers for transgender and gender diverse youth-a critical review of the literature. Child Adolesc Ment Health 2021; 26:3-14
  62. Mahfouda S, Moore JK, Siafarikas A, Zepf FD, Lin A. Puberty suppression in transgender children and adolescents. Lancet Diabetes Endocrinol 2017; 5:816-826
  63. Mejia-Otero JD, White P, Lopez X. Effectiveness of Puberty Suppression with Gonadotropin-Releasing Hormone Agonists in Transgender Youth. Transgender Health 2021; 6:31-35
  64. de Vries ALC, Steensma TD, Doreleijers TAH, Cohen‐Kettenis PT. Puberty Suppression in Adolescents With Gender Identity Disorder: A Prospective Follow‐Up Study. J Sex Med 2011; 8:2276-2283
  65. de Vries ALC, McGuire JK, Steensma TD, Wagenaar ECF, Doreleijers TAH, Cohen-Kettenis PT. Young adult psychological outcome after puberty suppression and gender reassignment. Pediatrics 2014; 134:696-704
  66. Harris RM, Tishelman AC, Quinn GP, Nahata L. Decision making and the long-term impact of puberty blockade in transgender children. The American Journal of Bioethics 2019; 19:67-69
  67. Schagen SE, Cohen-Kettenis PT, Delemarre-van de Waal HA, Hannema SE. Efficacy and Safety of Gonadotropin-Releasing Hormone Agonist Treatment to Suppress Puberty in Gender Dysphoric Adolescents. The journal of sexual medicine 2016; 13:1125-1132
  68. Klaver M, de Mutsert R, Wiepjes CM, Twisk JWR, den Heijer M, Rotteveel J, Klink DT. Early Hormonal Treatment Affects Body Composition and Body Shape in Young Transgender Adolescents. The journal of sexual medicine 2018; 15:251-260
  69. Nokoff NJ, Scarbro SL, Moreau KL, Zeitler P, Nadeau KJ, Reirden D, Juarez-Colunga E, Kelsey MM. Body Composition and Markers of Cardiometabolic Health in Transgender Youth on Gonadotropin-Releasing Hormone Agonists. Transgend Health 2021; 6:111-119
  70. Cutler GB, Jr. The role of estrogen in bone growth and maturation during childhood and adolescence. J Steroid Biochem Mol Biol 1997; 61:141-144
  71. Sasano H, Uzuki M, Sawai T, Nagura H, Matsunaga G, Kashimoto O, Harada N. Aromatase in human bone tissue. Journal of Bone and Mineral Research 1997; 12:1416-1423
  72. Schulmeister C, Millington K, Kaufman M, Finlayson C, Olson-Kennedy J, Garofalo R, Chan Y-M, Rosenthal SM. Growth in Transgender/Gender-Diverse Youth in the First Year of Treatment With Gonadotropin-Releasing Hormone Agonists. Journal of Adolescent Health 2021;
  73. Vlot MC, Klink DT, den Heijer M, Blankenstein MA, Rotteveel J, Heijboer AC. Effect of pubertal suppression and cross-sex hormone therapy on bone turnover markers and bone mineral apparent density (BMAD) in transgender adolescents. Bone 2017; 95:11-19
  74. Lee JY, Finlayson C, Olson-Kennedy J, Garofalo R, Chan Y-M, Glidden DV, Rosenthal SM. Low bone mineral density in early pubertal transgender/gender diverse youth: Findings from the Trans Youth Care Study. Journal of the Endocrine Society 2020; 4:bvaa065
  75. Joseph T, Ting J, Butler G. The effect of GnRH analogue treatment on bone mineral density in young adolescents with gender dysphoria: findings from a large national cohort. J Pediatr Endocrinol Metab 2019; 32:1077-1081
  76. Klink D, Caris M, Heijboer A, van Trotsenburg M, Rotteveel J. Bone mass in young adulthood following gonadotropin-releasing hormone analog treatment and cross-sex hormone treatment in adolescents with gender dysphoria. J Clin Endocrinol Metab 2015; 100:E270-275
  77. Chen D, Strang JF, Kolbuck VD, Rosenthal SM, Wallen K, Waber DP, Steinberg L, Sisk CL, Ross J, Paus T, Mueller SC, McCarthy MM, Micevych PE, Martin CL, Kreukels BPC, Kenworthy L, Herting MM, Herlitz A, Haraldsen I, Dahl R, Crone EA, Chelune GJ, Burke SM, Berenbaum SA, Beltz AM, Bakker J, Eliot L, Vilain E, Wallace GL, Nelson EE, Garofalo R. Consensus Parameter: Research Methodologies to Evaluate Neurodevelopmental Effects of Pubertal Suppression in Transgender Youth. Transgend Health 2020; 5:246-257
  78. Holmberg M, Arver S, Dhejne C. Supporting sexuality and improving sexual function in transgender persons. Nat Rev Urol 2019; 16:121-139
  79. Stephenson R, Riley E, Rogers E, Suarez N, Metheny N, Senda J, Saylor KM, Bauermeister JA. The sexual health of transgender men: a scoping review. The Journal of Sex Research 2017; 54:424-445
  80. van de Grift TC, van Gelder ZJ, Mullender MG, Steensma TD, de Vries ALC, Bouman MB. Timing of Puberty Suppression and Surgical Options for Transgender Youth. Pediatrics 2020; 146
  81. Nahata L, Chen D, Moravek MB, Quinn GP, Sutter ME, Taylor J, Tishelman AC, Gomez-Lobo V. Understudied and under-reported: fertility issues in transgender youth—a narrative review. The Journal of pediatrics 2019; 205:265-271
  82. Rivarola MA, Camacho AM, Migeon CJ. Effect of treatment with medroxyprogesterone acetate (Provera) on testicular function. The Journal of Clinical Endocrinology & Metabolism 1968; 28:679-684
  83. KUPPERMAN HS, Epstein JA. Medroxyprogesterone acetate in the treatment of constitutional sexual precocity. The Journal of Clinical Endocrinology & Metabolism 1962; 22:456-458
  84. Lee PA. Medroxyprogesterone therapy for sexual precocity in girls. American Journal of Diseases of Children 1981; 135:443-445
  85. Collipp PJ, Kaplan SA, Boyle DC, Plachte F, Kogut MD. CONSTITUTIONAL ISOSEXUAL PRECOCIOUS PUBERTY. EFFECTS OF MEDROXYPROGESTERONE ACETATE THERAPY. Am J Dis Child 1964; 108:399-405
  86. Hahn HB, Jr., Hayles AB, Albert A. MEDROXYPROGESTERONE AND CONSTITUTIONAL PRECOCIOUS PUBERTY. Mayo Clin Proc 1964; 39:182-190
  87. Schoen EJ. Treatment of idiopathic precocious puberty in boys. J Clin Endocrinol Metab 1966; 26:363-370
  88. Grundy V, Columbus G. Selecting and monitoring hormonal contraceptives: an overview of available products. US Pharm 2006; 6:62-70
  89. Merrin PK, Alexander WD. Cushing's syndrome induced by medroxyprogesterone. BMJ: British Medical Journal 1990; 301:345
  90. Lynch MM, Khandheria MM, Meyer III WJ. Retrospective study of the management of childhood and adolescent gender identity disorder using medroxyprogesterone acetate. International Journal of Transgenderism 2015; 16:201-208
  91. Nota NM, Wiepjes CM, de Blok CJM, Gooren LJG, Kreukels BPC, den Heijer M. Occurrence of Acute Cardiovascular Events in Transgender Individuals Receiving Hormone Therapy. Circulation 2019; 139:1461-1462
  92. Nokoff NJ, Scarbro S, Juarez-Colunga E, Moreau KL, Kempe A. Health and Cardiometabolic Disease in Transgender Adults in the United States: Behavioral Risk Factor Surveillance System 2015. J Endocr Soc 2018; 2:349-360
  93. Alzahrani T, Nguyen T, Ryan A, Dwairy A, McCaffrey J, Yunus R, Forgione J, Krepp J, Nagy C, Mazhari R, Reiner J. Cardiovascular Disease Risk Factors and Myocardial Infarction in the Transgender Population. Circ Cardiovasc Qual Outcomes 2019; 12:e005597
  94. Quinn VP, Nash R, Hunkeler E, Contreras R, Cromwell L, Becerra-Culqui TA, Getahun D, Giammattei S, Lash TL, Millman A. Cohort profile: Study of Transition, Outcomes and Gender (STRONG) to assess health status of transgender people. BMJ open 2017; 7:e018121
  95. Islam N, Nash R, Zhang Q, Panagiotakopoulos L, Daley T, Bhasin S, Getahun D, Haw JS, McCracken C, Silverberg MJ, Tangpricha V, Vupputuri S, Goodman M. Is There a Link Between Hormone Use and Diabetes Incidence in Transgender People? Data from the STRONG Cohort. J Clin Endocrinol Metab 2021;
  96. Maraka S, Singh Ospina N, Rodriguez-Gutierrez R, Davidge-Pitts CJ, Nippoldt TB, Prokop LJ, Murad MH. Sex Steroids and Cardiovascular Outcomes in Transgender Individuals: A Systematic Review and Meta-Analysis. J Clin Endocrinol Metab 2017; 102:3914-3923
  97. Klaver M, Dekker M, de Mutsert R, Twisk JWR, den Heijer M. Cross-sex hormone therapy in transgender persons affects total body weight, body fat and lean body mass: a meta-analysis. Andrologia 2017; 49
  98. Singh-Ospina N, Maraka S, Rodriguez-Gutierrez R, Davidge-Pitts C, Nippoldt TB, Prokop LJ, Murad MH. Effect of Sex Steroids on the Bone Health of Transgender Individuals: A Systematic Review and Meta-Analysis. J Clin Endocrinol Metab 2017; 102:3904-3913
  99. Olson-Kennedy J, Okonta V, Clark LF, Belzer M. Physiologic Response to Gender-Affirming Hormones Among Transgender Youth. J Adolesc Health 2018; 62:397-401
  100. Nokoff NJ, Scarbro SL, Moreau KL, Zeitler P, Nadeau KJ, Juarez-Colunga E, Kelsey MM. Body composition and markers of cardiometabolic health in transgender youth compared to cisgender youth. J Clin Endocrinol Metab 2019;
  101. Millington K, Finlayson C, Olson-Kennedy J, Garofalo R, Rosenthal SM, Chan YM. Association of High-Density Lipoprotein Cholesterol With Sex Steroid Treatment in Transgender and Gender-Diverse Youth. JAMA Pediatr 2021; 175:520-521
  102. Lee JY, Finlayson C, Olson-Kennedy J, Garofalo R, Chan YM, Glidden DV, Rosenthal SM. Low Bone Mineral Density in Early Pubertal Transgender/Gender Diverse Youth: Findings From the Trans Youth Care Study. Journal of the Endocrine Society 2020; 4:bvaa065
  103. Mamoojee Y, Seal LJ, Quinton R. Transgender hormone therapy: understanding international variation in practice. The Lancet Diabetes & Endocrinology 2017; 5:243-246
  104. Angus LM, Nolan BJ, Zajac JD, Cheung AS. A systematic review of antiandrogens and feminization in transgender women. Clinical endocrinology 2021; 94:743-752
  105. Bonne C, Raynaud JP. Mode of spironolactone anti-androgenic action: inhibition of androstanolone binding to rat prostate androgen receptor. Mol Cell Endocrinol 1974; 2:59-67
  106. Fagart J, Hillisch A, Huyet J, Bärfacker L, Fay M, Pleiss U, Pook E, Schäfer S, Rafestin-Oblin ME, Kolkhof P. A new mode of mineralocorticoid receptor antagonism by a potent and selective nonsteroidal molecule. J Biol Chem 2010; 285:29932-29940
  107. Kossor DC, Kominami S, Takemori S, Colby HD. Role of the steroid 17 alpha-hydroxylase in spironolactone-mediated destruction of adrenal cytochrome P-450. Mol Pharmacol 1991; 40:321-325
  108. Stripp B, Taylor AA, Bartter FC, Gillette JR, Loriaux DL, Easley R, Menard RH. Effect of spironolactone on sex hormones in man. J Clin Endocrinol Metab 1975; 41:777-781
  109. Prior JC, Vigna YM, Watson D. Spironolactone with physiological female steroids for presurgical therapy of male-to-female transsexualism. Arch Sex Behav 1989; 18:49-57
  110. Yemisci A, Gorgulu A, Piskin S. Effects and side‐effects of spironolactone therapy in women with acne. Journal of the European Academy of Dermatology and Venereology 2005; 19:163-166
  111. Helfer EL, Miller JL, Rose LI. Side-effects of spironolactone therapy in the hirsute woman. The Journal of Clinical Endocrinology & Metabolism 1988; 66:208-211
  112. Weill A, Cadier B, Nguyen P, Coste J. Exposition prolongée à de fortes doses d’acétate de cyprotérone et risque de méningiome chez la femme. Etude pharmaco-épidémiologique de cohorte à partir des données du SNDS 2019;
  113. Nota NM, Wiepjes CM, De Blok CJ, Gooren LJ, Peerdeman SM, Kreukels BP, Den Heijer M. The occurrence of benign brain tumours in transgender individuals during cross-sex hormone treatment. Brain 2018; 141:2047-2054
  114. Regidor P, Speer K, Regidor M, Schindler E. Long-term side-effects following cyproterone acetate containing therapy in gynecology. Zentralblatt fur Gynakologie 2000; 122:268-273
  115. Neyman A, Fuqua JS, Eugster EA. Bicalutamide as an androgen blocker with secondary effect of promoting feminization in male-to-female transgender adolescents. Journal of Adolescent Health 2019; 64:544-546
  116. Chiriacò G, Cauci S, Mazzon G, Trombetta C. An observational retrospective evaluation of 79 young men with long‐term adverse effects after use of finasteride against androgenetic alopecia. Andrology 2016; 4:245-250
  117. Thole Z, Manso G, Salgueiro E, Revuelta P, Hidalgo A. Hepatotoxicity induced by antiandrogens: A review of the literature. Urol Int 2004; 73:289-295
  118. Spratt DI, Stewart II, Savage C, Craig W, Spack NP, Chandler DW, Spratt LV, Eimicke T, Olshan JS. Subcutaneous injection of testosterone is an effective and preferred alternative to intramuscular injection: demonstration in female-to-male transgender patients. The Journal of Clinical Endocrinology & Metabolism 2017; 102:2349-2355
  119. Olson J, Schrager SM, Clark LF, Dunlap SL, Belzer M. Subcutaneous testosterone: an effective delivery mechanism for masculinizing young transgender men. LGBT health 2014; 1:165-167
  120. Wilson DM, Kiang TK, Ensom MH. Pharmacokinetics, safety, and patient acceptability of subcutaneous versus intramuscular testosterone injection for gender-affirming therapy: a pilot study. The Bulletin of the American Society of Hospital Pharmacists 2018; 75:351-358
  121. Kaminetsky J, Jaffe JS, Swerdloff RS. Pharmacokinetic profile of subcutaneous testosterone enanthate delivered via a novel, prefilled single‐use autoinjector: a phase ii study. Sexual medicine 2015; 3:269-279
  122. Rothman MS, Iwamoto SJ. Bone Health in the Transgender Population. Clin Rev Bone Miner Metab 2019; 17:77-85
  123. Rosen HN, Hamnvik O-PR, Jaisamrarn U, Malabanan AO, Safer JD, Tangpricha V, Wattanachanya L, Yeap SS. Bone densitometry in transgender and gender non-conforming (TGNC) individuals: 2019 ISCD official position. Journal of Clinical Densitometry 2019; 22:544-553
  124. Valentine A, Nokoff N, Bonny A, Chelvakumar G, Indyk J, Leibowitz S, Nahata L. Cardiometabolic Parameters Among Transgender Adolescent Males on Testosterone Therapy and Body Mass Index-Matched Cisgender Females. Transgender health 2021; 6:369-373
  125. Clark BA, Veale JF, Townsend M, Frohard-Dourlent H, Saewyc E. Non-binary youth: Access to gender-affirming primary health care. International Journal of Transgenderism 2018; 19:158-169
  126. Kanj RV, Conard LAE, Corathers SD, Trotman GE. Hormonal contraceptive choices in a clinic-based series of transgender adolescents and young adults. Int J Transgend 2019; 20:413-420
  127. Krempasky C, Harris M, Abern L, Grimstad F. Contraception across the transmasculine spectrum. American journal of obstetrics and gynecology 2020; 222:134-143
  128. Ahmad S, Leinung M. The response of the menstrual cycle to initiation of hormonal therapy in transgender men. Transgender health 2017; 2:176-179
  129. Colebunders B, Brondeel S, D'Arpa S, Hoebeke P, Monstrey S. An Update on the Surgical Treatment for Transgender Patients. Sex Med Rev 2017; 5:103-109
  130. Berli JU, Knudson G, Fraser L, Tangpricha V, Ettner R, Ettner FM, Safer JD, Graham J, Monstrey S, Schechter L. What Surgeons Need to Know About Gender Confirmation Surgery When Providing Care for Transgender Individuals: A Review. JAMA Surg 2017; 152:394-400
  131. Nunes-Moreno M, Buchanan C, Cole FS, Davis S, Dempsey A, Dowshen N, Furniss A, Kazak AE, Kerlek AJ, Margolis P, Pyle L, Razzaghi H, Reirden DH, Schwartz B, Sequeira GM, Nokoff NJ. Behavioral Health Diagnoses in Youth with Gender Dysphoria Compared with Controls: A PEDSnet Study. J Pediatr 2021;
  132. Becerra-Culqui TA, Liu Y, Nash R, Cromwell L, Flanders WD, Getahun D, Giammattei SV, Hunkeler EM, Lash TL, Millman A, Quinn VP, Robinson B, Roblin D, Sandberg DE, Silverberg MJ, Tangpricha V, Goodman M. Mental Health of Transgender and Gender Nonconforming Youth Compared With Their Peers. Pediatrics 2018; 141
  133. Connolly MD, Zervos MJ, Barone CJ, 2nd, Johnson CC, Joseph CL. The Mental Health of Transgender Youth: Advances in Understanding. J Adolesc Health 2016; 59:489-495
  134. Reisner SL, Vetters R, Leclerc M, Zaslow S, Wolfrum S, Shumer D, Mimiaga MJ. Mental health of transgender youth in care at an adolescent urban community health center: a matched retrospective cohort study. J Adolesc Health 2015; 56:274-279
  135. Almeida J, Johnson RM, Corliss HL, Molnar BE, Azrael D. Emotional distress among LGBT youth: the influence of perceived discrimination based on sexual orientation. J Youth Adolesc 2009; 38:1001-1014
  136. Warrier V, Greenberg DM, Weir E, Buckingham C, Smith P, Lai MC, Allison C, Baron-Cohen S. Elevated rates of autism, other neurodevelopmental and psychiatric diagnoses, and autistic traits in transgender and gender-diverse individuals. Nat Commun 2020; 11:3959
  137. Grossman AH, D'Augelli AR. Transgender youth and life-threatening behaviors. Suicide Life Threat Behav 2007; 37:527-537
  138. James SE, Herman, J. L., Rankin, S., Keisling, M., Mottet, L., & Anafi, M. The Report of the 2015 U.S. Transgender Survey. Washington, DC: National Center for Transgender Equality 2016;
  139. Hendricks ML, Testa RJ. A conceptual framework for clinical work with transgender and gender nonconforming clients: An adaptation of the Minority Stress Model. Professional Psychology: Research and Practice 2012; 43:460-467
  140. Chodzen G, Hidalgo MA, Chen D, Garofalo R. Minority stress factors associated with depression and anxiety among transgender and gender-nonconforming youth. Journal of Adolescent Health 2019; 64:467-471
  141. Kozee HB, Tylka TL, Bauerband LA. Measuring transgender individuals’ comfort with gender identity and appearance: Development and validation of the Transgender Congruence Scale. Psychology of Women Quarterly 2012; 36:179-196
  142. Durwood L, McLaughlin KA, Olson KR. Mental Health and Self-Worth in Socially Transitioned Transgender Youth. J Am Acad Child Adolesc Psychiatry 2017; 56:116-123 e112
  143. Singh AA, Meng SE, Hansen AW. “I am my own gender”: Resilience strategies of trans youth. Journal of Counseling & Development 2014; 92:208-218
  144. Turban JL, King D, Carswell JM, Keuroghlian AS. Pubertal Suppression for Transgender Youth and Risk of Suicidal Ideation. Pediatrics 2020; 145
  145. Rider GN, McMorris BJ, Gower AL, Coleman E, Eisenberg ME. Health and Care Utilization of Transgender and Gender Nonconforming Youth: A Population-Based Study. Pediatrics 2018; 141
  146. Skagerberg E, Di Ceglie D, Carmichael P. Brief Report: Autistic Features in Children and Adolescents with Gender Dysphoria. J Autism Dev Disord 2015; 45:2628-2632
  147. Hisle-Gorman E, Landis CA, Susi A, Schvey NA, Gorman GH, Nylund CM, Klein DA. Gender Dysphoria in Children with Autism Spectrum Disorder. LGBT Health 2019; 6:95-100
  148. Strang JF, Kenworthy L, Dominska A, Sokoloff J, Kenealy LE, Berl M, Walsh K, Menvielle E, Slesaransky-Poe G, Kim KE, Luong-Tran C, Meagher H, Wallace GL. Increased gender variance in autism spectrum disorders and attention deficit hyperactivity disorder. Arch Sex Behav 2014; 43:1525-1533
  149. van der Miesen AIR, de Vries ALC, Steensma TD, Hartman CA. Autistic Symptoms in Children and Adolescents with Gender Dysphoria. J Autism Dev Disord 2018; 48:1537-1548
  150. de Vries AL, Noens IL, Cohen-Kettenis PT, van Berckelaer-Onnes IA, Doreleijers TA. Autism spectrum disorders in gender dysphoric children and adolescents. J Autism Dev Disord 2010; 40:930-936
  151. Thrower E, Bretherton I, Pang KC, Zajac JD, Cheung AS. Prevalence of autism spectrum disorder and attention-deficit hyperactivity disorder amongst individuals with gender dysphoria: a systematic review. Journal of autism and developmental disorders 2020; 50:695-706
  152. Shumer DE, Reisner SL, Edwards-Leeper L, Tishelman A. Evaluation of Asperger Syndrome in Youth Presenting to a Gender Dysphoria Clinic. LGBT Health 2016; 3:387-390
  153. Grossman AH, D'augelli AR, Frank JA. Aspects of psychological resilience among transgender youth. Journal of LGBT youth 2011; 8:103-115
  154. Watson RJ, Veale JF, Saewyc EM. Disordered eating behaviors among transgender youth: Probability profiles from risk and protective factors. Int J Eat Disord 2017; 50:515-522
  155. Simons L, Schrager SM, Clark LF, Belzer M, Olson J. Parental support and mental health among transgender adolescents. J Adolesc Health 2013; 53:791-793
  156. Olson KR, Durwood L, DeMeules M, McLaughlin KA. Mental Health of Transgender Children Who Are Supported in Their Identities. Pediatrics 2016; 137:e20153223
  157. Delozier AM, Kamody RC, Rodgers S, Chen D. Health Disparities in Transgender and Gender Expansive Adolescents: A Topical Review From a Minority Stress Framework. Journal of Pediatric Psychology 2020; 45:842-847
  158. Turban JL, King D, Carswell JM, Keuroghlian AS. Pubertal Suppression for Transgender Youth and Risk of Suicidal Ideation. Pediatrics 2020; 145:e20191725
  159. van der Miesen AIR, Steensma TD, de Vries ALC, Bos H, Popma A. Psychological Functioning in Transgender Adolescents Before and After Gender-Affirmative Care Compared With Cisgender General Population Peers. J Adolesc Health 2020; 66:699-704
  160. Sorbara JC, Chiniara LN, Thompson S, Palmert MR. Mental Health and Timing of Gender-Affirming Care. Pediatrics 2020:e20193600
  161. Gao Y, Maurer T, Mirmirani P. Understanding and Addressing Hair Disorders in Transgender Individuals. Am J Clin Dermatol 2018; 19:517-527
  162. Yeung H, Kahn B, Ly BC, Tangpricha V. Dermatologic conditions in transgender populations. Endocrinology and Metabolism Clinics 2019; 48:429-440
  163. Ginsberg B. Dermatologic care of the transgender patient. Vol 3: Elsevier; 2017:65-67.
  164. Julian JM, Salvetti B, Held JI, Murray PM, Lara-Rojas L, Olson-Kennedy J. The Impact of Chest Binding in Transgender and Gender Diverse Youth and Young Adults. J Adolesc Health 2021; 68:1129-1134
  165. Baral SD, Poteat T, Strömdahl S, Wirtz AL, Guadamuz TE, Beyrer C. Worldwide burden of HIV in transgender women: a systematic review and meta-analysis. Lancet Infect Dis 2013; 13:214-222
  166. Herbst JH, Jacobs ED, Finlayson TJ, McKleroy VS, Neumann MS, Crepaz N. Estimating HIV prevalence and risk behaviors of transgender persons in the United States: a systematic review. AIDS Behav 2008; 12:1-17
  167. Becasen JS, Denard CL, Mullins MM, Higa DH, Sipe TA. Estimating the Prevalence of HIV and Sexual Behaviors Among the US Transgender Population: A Systematic Review and Meta-Analysis, 2006-2017. Am J Public Health 2019; 109:e1-e8
  168. Bowen AE, Staggs S, Kaar J, Nokoff N, Simon SL. Short sleep, insomnia symptoms, and evening chronotype are correlated with poorer mood and quality of life in adolescent transgender males. Sleep Health 2021; 7:445-450
  169. Jones BA, Arcelus J, Bouman WP, Haycraft E. Sport and Transgender People: A Systematic Review of the Literature Relating to Sport Participation and Competitive Sport Policies. Sports Med 2017; 47:701-716
  170. Hartman-Munick SM, Silverstein S, Guss CE, Lopez E, Calzo JP, Gordon AR. Eating disorder screening and treatment experiences in transgender and gender diverse young adults. Eat Behav 2021; 41:101517
  171. Coelho JS, Suen J, Clark BA, Marshall SK, Geller J, Lam PY. Eating Disorder Diagnoses and Symptom Presentation in Transgender Youth: a Scoping Review. Curr Psychiatry Rep 2019; 21:107
  172. Glidden D, Bouman WP, Jones BA, Arcelus J. Gender Dysphoria and Autism Spectrum Disorder: A Systematic Review of the Literature. Sex Med Rev 2016; 4:3-14
  173. Olson-Kennedy J, Chan YM, Rosenthal S, Hidalgo MA, Chen D, Clark L, Ehrensaft D, Tishelman A, Garofalo R. Creating the Trans Youth Research Network: A Collaborative Research Endeavor. Transgender health 2019; 4:304-312
  174. Reardon S. The largest study involving transgender people is providing long-sought insights about their health. Nature 2019; 568:446-450
  175. Flentje A, Heck NC, Brennan JM, Meyer IH. The relationship between minority stress and biological outcomes: A systematic review. Journal of Behavioral Medicine 2019:1-22
  176. Ditmars HL, Logue MW, Toomey R, McKenzie RE, Franz CE, Panizzon MS, Reynolds CA, Cuthbert KN, Vandiver R, Gustavson DE. Associations between depression and cardiometabolic health: A 27-year longitudinal study. Psychological Medicine 2021:1-11
  177. Streed Jr CG, Beach LB, Caceres BA, Dowshen NL, Moreau KL, Mukherjee M, Poteat T, Radix A, Reisner SL, Singh V. Assessing and addressing cardiovascular health in people who are transgender and gender diverse: A scientific statement from the American heart association. Circulation 2021; 144:e136-e148
  178. Rosenthal SM. Approach to the Patient: Transgender Youth: Endocrine Considerations. J Clin Endocrinol Metab. 2014 2014;99:4379-4389.

Thyroid Hormones in Brain Development and Function

ABSTRACT

 

Thyroid hormones are essential for normal brain development. They influence neurogenesis, neuronal and glial cell differentiation and migration, synaptogenesis, and myelination. Thyroid hormone deficiency may severely affect the brain during fetal and postnatal development, causing retarded maturation, intellectual deficits, and neurological impairment. Neural cells express the thyroid hormone nuclear receptors THRA and THRB, which mediate most actions of T3, the active hormone. Brain T3 derives in part from the circulation, and part from type-2 deiodinase-mediated 5’-deiodination of T4 in glial cells. Type 3 deiodinase inactivates T4 and T3 by 5-deiodination in neurons. Membrane transporters facilitate the passage of T4 and T3 across the brain barriers. The main transporters are the monocarboxylate transporter 8 (MCT8) and the organic anion transporter polypeptide 1C1 (OATP1C1). MCT8 facilitates T4 and T3 transport whereas OATP1C1 transports T4 but not T3. T3 regulates the expression of a large number of genes in the brain, mostly during developmental stages, but also in the adult. Rodent models of disease have provided most of our knowledge on thyroid hormone action in the brain. However, species-specific differences in brain maturation and organization make it difficult sometimes to extrapolate the data obtained in rodent models to the human. This review will present a summary of the main concepts developed from rodent studies, with a focus on the human brain.

 

INTRODUCTION

 

Thyroid hormones are crucial for brain development, and influence brain function throughout life. In adults, hypothyroidism causes lethargy, hyporeflexia, and poor motor coordination (1,2), is associated with bipolar affective disorders, depression, or loss of cognitive functions (3,4). Subclinical hypothyroidism is often associated with memory impairment. Conversely, hyperthyroidism causes hyperreflexia, irritability, and anxiety among other symptoms (5). Hypo- or hyperthyroidism can lead to mood disorders, dementia, confusion, and personality changes. Most of these disorders are usually reversible with proper treatment, indicating that adult-onset thyroid hormone alterations affect neural function but do not leave permanent structural defects.

 

The actions of thyroid hormones during mammalian brain maturation are qualitatively different. They influence many developmental processes, usually during limited time windows.  They are required for the timely synchronization of independent events and facilitate the transition between fetal and postnatal stages (6,7), similarly to promoting amphibian metamorphosis (8,9). Thyroid hormone deficiency during critical transition periods may lead to irreversible brain damage, the consequences of which depend on the severity and duration of the deficiency, and most importantly its time of onset (10-14).

 

Until recently, the rat was the most widely used animal model in the study of thyroid physiology and the actions of thyroid hormones in the brain. However, for more than 20 years, the mouse is the preferred animal model, and the use of knockout and knockin mice has facilitated our understanding. However, it is important to be aware of species specificities which make it difficult to extrapolate the results to the human situation. The timing of development in relation to birth among mammals presents substantial differences, even if the sequence of events might be similar (15-17). The web resource www.translatingtime.org (18) provides tools to compare neurodevelopmental time across species. As an approximation, the newborn rat may be compared with a second-trimester human fetus, and the maturation of a newborn human cerebral cortex to that of a 12-13-day old rat pup (19). For integrative reviews on molecular and evolutionary aspects of the cerebral cortex and cerebellar development and the effects of thyroid hormones see (20-23).

 

Whenever possible, this chapter focuses on observations in humans. Gene and protein notations follow the HUGO nomenclature (http://www.genenames.org): for human genes, names are in italics and in capital letters, protein names are written in non-italic capital letters irrespective of species.

 

STRUCTURAL DEFECTS CAUSED BY THYROID HORMONE DEFICIENCY

 

As an approach to understanding how thyroid hormones influence brain development, the changes caused by thyroid hormone deprivation are informative. In rats, perinatal hypothyroidism causes reduced myelination and diverse structural defects (for a classical review on this topic see (24). A reduction of the neuropil causes increased cell density in the cerebral cortex (25,26). Reduction in total cell numbers of regions with significant neurogenesis during the postnatal period, such as the olfactory bulb and the granular layers of the hippocampus and cerebellum (26-28). Transient structures show retarded disappearance. An important example is the subplate, a transient structure of the cortex involved in the organization of thalamic afferents to the cortex (29). In the cerebellum, regression of the external granular, or germinal layer, is retarded by a few days (30). The Cajal-Retzius cells, formed early during cortical development and involved in the regulation of the inside-out migration of neurons have delayed appearance (Fig. 1 left panel) (31). The GABAergic interneurons have altered distribution and connectivity, and the parvalbumin subclass is reduced in number (32-35). The maturation of several types of neurons is compromised, with stunted dendritic and/or axonal growth and maturation, for example cholinergic cells (36), cerebellar Purkinje cells (Fig. 1 central panel) (37,38), and cortex layer V pyramidal cells (39,40). Changes in dendritic spine number are also observed in the cortex and hippocampus after adult-onset hypothyroidism and are reversible with thyroxine treatment (41,42). Hypothyroidism also causes delayed and poor deposition of myelin (43-46) whereas hyperthyroidism accelerates myelination (47).  After prolonged neonatal hypothyroidism, the number of myelinated axons in adult rats is abnormally low in hypothyroid animals although most of the myelinated axons appear to have a normal thickness of the myelin sheath (Fig. 1 right panel).

Fig. 1. Examples of the effects of hypothyroidism on developmental timing, cell differentiation, and cell migration. Left panel: In the cortical plate, hypothyroidism of fetal onset reduced the number of Cajal-Retzius cells, Rln mRNA (reelin), and reelin protein at P0. Normal amounts of reelin were present in the cortex at P5, indicating that thyroid hormones control the timing of Rln expression (from (31)). Central panel: Neonatal hypothyroidism causes arrested Purkinje cell differentiation (upper panels), and delayed disappearance of the external (germinal) granular layer due to delayed migration of granular cells. The right panel shows a permanent myelin deficit in hypothyroidism. Myelin staining of the anterior commissure of adult rats with fetal/neonatal-onset hypothyroidism and their respective euthyroid controls shows a thinner and paler commissure in the hypothyroid rats.  Electron microscope imaging reveals that a higher proportion of axons of hypothyroid rats were of lower diameter and unmyelinated (from (48)).

 

Less information is available on how hypothyroidism affects the structure of the human brain. Post mortem examination in two cases of deficient thyroid hormone transport to the brain caused by MCT8 mutations revealed anatomical changes compatible with thyroid hormone deficiency (49) (Fig. 2): delayed maturation of the neocortex and cerebellum, delayed myelination, altered neuronal differentiation with lower expression of neurofilaments, and reduced synaptogenesis with reduced synaptophysin expression. Specific cellular changes included a reduced number of Cajal-Retzius cells and parvalbumin interneurons in the cortex. Cerebellar Purkinje cells had a normal morphology in contrast to the usual finding in hypothyroid rodents. These findings indicate that it is possible to extrapolate, but with caution, the brain lesions observed in hypothyroid rodents to humans.

Fig. 2. MCT8 mutations cause anatomical changes compatible with cerebral hypothyroidism already during fetal stages. The left upper panels show staining of the cortex with neurofilament (brown color) in a control fetus (a) and a fetus with a MCT8 mutation (b). The normal cortex shows staining of Cajal-Retzius cells, which were absent in the MCT8 fetus. The left lower panels show staining of the cerebellum with anti-myelin basic protein. The normal fetus (c) shows immunoreactivity, which is not present in the MCT8 fetus (d). The right panels correspond to an 11-years old boy with MCT8 mutation (panels f, h, k l, n) and control of the same age (panels e, g, i, j, m). Panels e, f: cross-section of the pyramidal track stained with neurofilament; in the MCT8-mutated patient the axonal diameter is smaller than in the control. Panels g, h: staining of the cortex with anti-parvalbumin antibody shows that parvalbumin interneurons are present in the normal cortex but not in the MCT8 cortex. Panels i-l: cerebellar staining for myelin basic protein (i, j) and myelin (k, l). Panels m. n: cerebellar staining for synaptophysin showing the presence of synaptic boutons around the body of a Purkinje cell in the control (m) but not in the patient (n). Data from (49).

 

THYROID HORMONES IN THE DEVELOPING BRAIN

 

Maternal and Fetal Thyroid Hormones

 

The relative roles of the maternal and fetal thyroid hormones on brain development is a most important question. Fetal brain thyroid hormones depend on the transplacental passage of the maternal hormones and the onset of fetal thyroid function. In rodents, the onset of active fetal thyroid secretion occurs at E17.5, but thyroid hormones are present earlier in the embryos, a few days after implantation. This hormone is of maternal origin, as T4 and T3 cross the placenta (50-55). In normal rats at term, the maternal T4 accounts for about 17.5% of the total extra-thyroidal fetal thyroxine pool (56). However, in the fetal brain the concentrations of T4 and T3 are low before the onset of fetal thyroid function, and rapidly increase several-fold from E18 to E21 (57).

 

In humans, Vulsma et al. (58) demonstrated the transplacental passage of T4 by showing that in neonates with thyroid agenesis or a total organification defect, T4 was present in cord serum at 30-50% of the normal concentration. The only possible origin of this T4 was maternal. Transfer of T4 from the mother to the fetus protects the fetal brain in congenital hypothyroidism, preventing neurological damage before birth, and making it possible that early postnatal treatment is effective (59). The importance of maternal hormones becomes apparent in situations where protection does not take place. This occurs in the combined maternal and fetal thyroid failure, as in feto-maternal PIT-1 deficiency (60), or presence of high titers of thyroid stimulation blocking antibodies (61).  In these cases, mothers and neonates had extremely low thyroid hormones. The infants suffered profound developmental delays, permanent sensorineural deafness, and irreversible neuromotor impairment.

 

STUDIES IN RODENTS

 

If thyroid hormones from the mother offer protection to the brain in cases of fetal thyroid failure, one could then ask about the relevance of maternal thyroid hormones for brain development in the presence of a normal fetal thyroid gland, or before the gland becomes functional. If maternal thyroid hormones contribute to fetal brain development, then alterations of maternal thyroid physiology might have consequences on the fetal brain. A relevant question is whether the isolated failure of the mother’s thyroid gland causes thyroid hormone deficiency in the fetal brain. Thyroidectomy in pregnant rats, a situation of maternal hypothyroidism with intact fetal thyroid, causes a reduction in the total extra-thyroidal T4 and T3 of the fetuses and developmental delays, but at E21 there was no reduction of T4 and T3 in the brain compared to fetuses from control dams (62). In these experiments, fetal thyroid activity compensated for any possible reduction of brain thyroid hormones caused by maternal thyroidectomy. More recent studies (63), confirmed that thyroidectomy of the dams did not alter the concentrations of T4 and T3 in the fetal cerebral cortex at E21, whereas dams treated with methyl-mercapto-imidazole (methimazole), which causes maternal and fetal hypothyroidism, had greatly reduced brain T4 and T3 concentrations. Microarray analysis of the fetal cerebral cortex successfully identified many genes with altered expression in the combined maternal and fetal hypothyroidism methimazole-induced model but with unchanged fetal expression in the offspring of thyroidectomized mothers.  The conclusion emanating from the comparison of these two models is that fetal brain gene expression in the rat at term is under predominant control of the fetal thyroid gland.

 

In contrast to these studies, the offspring from rat dams thyroidectomized at day 16 of pregnancy showed altered cortical lamination and other structural defects when analyzed at P40 (29). The bulk microarray studies of the cortex might have failed to identify subtle, layer-specific changes of gene expression requiring time to transduce into structural alterations. Other studies support the effect of maternal thyroid hormones before the onset of fetal thyroid function. Transient maternal hypothyroidism of pregnant rats, from E12 to E15, caused the displacement of cells in the neocortex and hippocampus of the offspring, associated with audiogenic seizures when analyzed at 40 days of age, (64). Moderate thyroid hormone deficiency during pregnancy caused neuronal ectopias in the corpus callosum of the progeny (65). Thus, although the fetal thyroid gland exerts the main control on fetal brain development, there is experimental support for subtle actions of maternal thyroid hormone before the onset of fetal thyroid function. These actions do not have yet a correlate in terms of defined molecular events, and the possibility remains that they reflect indirect effects of systemic hypothyroidism rather than direct actions of the hormones on neural targets (66).

 

STUDIES IN HUMANS  

 

Early studies on the human fetal thyroid gland development showed that colloid formation, iodide concentration, and synthesis of thyroglobulin and T4 could be demonstrated by the 11th week of gestation (67,68). Recent gene expression studies showed that the genes encoding thyroid transcription factors, thyroglobulin, thyroid peroxidase, and the TSH receptor are expressed already by week 7, and that the sodium-iodide symporter is strongly upregulated by the 10th week (69).  From the 11th week, serum total and free T4 and T3 increase with time (Fig 3) (70,71), and T4 reaches maternal concentrations by the 36th week (72). The increased T4 and T3 observed in Fig. 3 are clearly due to fetal thyroid gland activity, and it is uncertain to what extent the maternal hormones contribute to the fetal hormone pool in the presence of a normally active fetal thyroid gland. Supporting the contribution of maternal hormones in a situation of normality, its interruption might explain the relative hypothyroxinemia of premature babies (73). Early in gestation, T4 is present in low amounts in the coelomic fluid from the 5th-6thgestational weeks, and the amniotic fluid contains T4 and T3 from the 10th-12th week (74,75).

Fig. 3. Developmental events in the human fetal cortex and changes of thyroid hormone concentrations and nuclear receptor. The lower part of the figure shows the approximate timing of developmental events, the formation of the neural tube; the start of neurogenesis on GW5-5; the formation of the preplate, i.e. the formation of the first neurons, the Cajal-Retzius cells in the marginal zone, and the subplate cells; the preplate split, when the first projection neurons migrate and start filling the space between the Cajal-Retzius and the subplate cells, leading to the formation of the cortical plate; the appearance of tight junctions between vascular endothelial cells, and the blood-brain barrier (BBB), neuronal migration, and gliogenesis with the appearance of astroglial cells and oligodendrocytes. The vertical gray band marks the onset of thyroid gland function. Data on thyroid hormone concentrations in serum are from (71). Data on thyroid hormone concentrations in the cortex are from (76). Data on T3 nuclear receptor are from (77).

 

The recognition of the transplacental passage of thyroid hormones, described above, raised the question of the role of maternal T4 on fetal brain development (78). The implication is that maternal T4 exerts actions on the fetal brain before onset of the fetal thyroid gland, and even complements the fetal hormones after midgestation (79). Reports are indicating adverse effects of hypothyroxinemia on cognition and behavior of the progeny (80-82), or no effects (83). The issue is unsettled, mainly due to disparities in methodological approaches and the presence of compounding factors. Among these, the definition of hypothyroxinemia, the presence or not of hypothyroidism, iodine deficiency as the main cause of hypothyroxinemia (84), environmental pollutants, and indirect effects of hypothyroxinemia through complications of pregnancy (85-88). Clinical trials involving treatment of hypothyroxinemic pregnant women with T4 gave no clear answers yet (89-91).

 

Transport of Thyroid Hormones into the Brain

 

Other chapters of this book describe in detail the issue of thyroid hormone transport (92). This section contains only a few specificities concerning transport in the brain. The crucial role of transporters in the brain is to facilitate that T4 and T3 from the circulation cross the brain barriers (93-97). There are two main barriers in the mature brain: the blood-brain barrier (BBB) and the blood-cerebrospinal-spinal fluid barrier (98,99). The presence of tight junctions between endothelial vascular cells, or choroid plexus epithelial cells form barriers that hamper paracellular transport. Crossing these barriers requires transporters at the opposite sides of the plasma membrane facilitating the influx and efflux of the transported solutes. The gradient concentration of the free solutes determines the flux direction. The BBB surface is 5000-fold larger than that of the blood-CSF barrier (100). The larger exchange surface and the short distance of individual neurons from microvessels (8-20 µm (98)), makes the BBB the most relevant site of transport from the blood to the brain parenchyma.

Tight junctions between vascular endothelial cells, i.e., the BBB proper, are present in rodents by E15-E16 (101,102), and in humans by GW12 (99) (Fig. 3).

Fig. 4. Thyroid hormone transport and metabolism in the rodent brain. MCT8 in the blood-brain barrier facilitates the transport of circulating T4 and T3 to the brain parenchyma and OATP1C1 the transfer of T4. T4 in the astrocytes is converted to T3 by DIO2, providing additional T3 to nearby neural cells. DIO3 in neurons degrades T3 to T2 and T4 to rT3. Modified with permission from (103).

 

Further maturation and consolidation require the presence of pericytes and astrocytes in the vascular unit (104). Lopez-Espindola et al. (96) found immunoreactive MCT8 at GW12 in the human fetal brain endothelial cells, possibly highlighting the importance of MCT8-mediated transport at a time when the BBB starts limiting the entry of solutes to the brain. MCT8 was also present in other barriers such as the blood-cerebrospinal fluid barrier, the transient outer cerebrospinal fluid-brain barrier, and the ependymocytes. These findings suggest the presence of alternative routes by which T4 and T3 could reach the brain parenchyma in the fetus.

 

There is a fundamental difference between rodents and humans in the transport of thyroid hormones through the BBB. The rodent BBB contains MCT8 and OATP1C1 (Fig. 4). MCT8 transports T4 and T3 whereas OATP1C1 transports T4, reverse T3 (rT3), and T4 sulfate. In humans, as in other primates, the BBB contains MCT8 but lacks OATP1C1 (93,105). One explanation why MCT8 mutations cause profound neurological impairment in patients but not in mice is that T4 transport through OATP1C1 compensates similar mutations in mice by making T4 available as a DIO2 substrate. There is experimental support for this mechanism as follows: i. MCT8-deficient mice lack neurological impairment (106), with only slight alterations of brain gene expression (107), contrary to what would be expected; ii. DIO2 activity and T3 generation increase in the brain of MCT8-deficient mice (108,109); and iii. To achieve cerebral hypothyroidism in mice, a compound deficiency of MCT8 and DIO2, or MCT8 and OATP1C1 is needed, i.e., a knockout of Slc16a2 and Dio2 or Slc16a2 and Slco1c1 (66,107,110,111).

 

Role of Deiodinases

 

The deiodinases present in the cerebrum are DIO2 and DIO3 (112). The adult mouse cerebellum also contains significant DIO1 activity (113). Different cells express DIO2 and DIO3 in the brain. Glial cells and some interneurons express DIO2, and excitatory neurons express DIO3 (114-118). The glial cells expressing DIO2 include radial glial cells, astrocytes, and tanycytes (Fig. 5) (114,116,118). Dio2 expression occurs in most brain areas, and increase in hypothyroidism (Fig. 5). Dio3 shows a restricted high expression at P0 in discrete nuclei, related to sexual differentiation of the brain, but the physiological relevance of this expression has not been studied (Fig. 5 (119)).

Fig. 5. Dio2 and Dio3 expression by in situ hybridization. Panels A, E, Dio2: in euthyroid rats (panel A) Dio2is distributed all over the brain especially in the upper layers of the cortex, the hippocampus, the thalamic ventromedial nucleus, with increased expression in hypothyroid rats (panel B). The median eminence, infundibulum, and walls of the 3rd ventricle show high expression in tanycytes, a specialized type of glial cells (panel C). Dio2 increases in hypothyroidism also in this region (panel D). In the rest of the brain, Dio2is expressed in the astrocytes as shown in panel E after GFAP counterstaining of the in situ hybridization. Panels F-H; Dio3 expression in the mouse at P0: Selective expression in the accumbens nucleus and the anterior pole of the bed nucleus of stria terminalis (Acb/BST, panel F), BST, median and medial preoptic nuclei (MnPO and MPO, panel G) and amygdala (panel H).  Panels A, B, and E (114,115). Panels C and D are adapted from (116). Panels F,G,H are from (119).

 

Astrocytes, which exceed by ten-fold the number of neurons supply T3 to other neural cells by DIO2 deiodination of T4 (Fig. 3) (114,115,120). In rodents, the majority of astrocytes arise during the first postnatal week, and DIO2 expression and activity increase accordingly. Tanycytes are specialized glial cells lining the walls of the 3rd ventricle and the cause for the high hypothalamic DIO2 activity (121). Tanycytes in the median eminence form a blood-hypothalamus barrier, modulated in response to metabolic factors, which control the access of blood-borne substances to the arcuate nucleus (122). These cells are linked to the central control of feeding, body weight, and energy balance, and may act as stem cells to produce hypothalamic neurons (123). The hypothalamic paraventricular nucleus does not express DIO2, and T3 generated in astrocytes or tanycytes might control TRH production in this nucleus (114).

 

It is estimated that in DIO2-expressing tissues, such as the brain, brown adipose tissue, and pituitary, 50% or more of T3 derives from local T4 deiodination (124-127). In the adult rat brain, as much as 80% of nuclear-bound T3 is formed locally from T4 (128). Through DIO2 and DIO3 expression, the concentrations of T3 meet the local requirements of the particular developmental stage independently of fluctuations of circulating T3. Some discrete examples are the roles of Dio3 (129) and Dio2 (130) in the mouse cochlea with a peak of DIO2 activity on P7 before the onset of hearing, and the sequential expression of Dio3 and Dio2 in the control of retinal cone specification (131).

 

The balance between DIO2 and DIO3 activity during development is critical to ensure adequate amounts of T3 in neural tissue. Extremely high levels of Dio3 expression and activity are present in the uterine implantation site, and later on in the uterine epithelium, preventing that inadequate amounts of T4 and T3 reach the embryos at early stages (132). By the end of pregnancy in the rat, DIO2 activity increases markedly in the fetal brain, in parallel with a 10-fold increase of T4 and 28-fold increase of T3 (133,134). The increase in DIO2 activity occurs after the start of gliogenesis and generation of astrocytes by E17 (135).

 

The fetal brain produces most T3 locally from circulating T4. Circulating T3 has poor access to the fetal brain. In a classical experiment, Morreale de Escobar and coworkers (136) showed that generation of T3 from T4 provides a means to regulate the concentrations of T3 within narrow limits (Fig. 6).

Fig. 6. T3 concentrations in the fetal brain after infusion of pregnant rats with increasing doses of T4 or T3. MMI-treated pregnant rats were infused with T4 or T3 using osmotic pumps from gestational day 15 through 21. The doses shown correspond to the starting dose on E15 and remained constant during the infusion period. Infusion of T4, but not T3, increased T3 in the brain. Data from (136).

 

In this experiment, they infused increasing doses of T4 or T3 to hypothyroid pregnant rats and then measured the resulting concentrations of T3 in the hypothyroid fetal brain. After T4 infusion to the mother, the fetal brain T3 concentration increased, reaching a normal concentration with a relatively low dose of T4. In contrast, it was difficult to increase T3 in the fetal brain after T3 infusion only over a wide dose range. DIO2-dependent generation of T3 in the fetal brain from maternal T4 also occurs in euthyroid pregnant mice. Administration of T3 to the mothers increased T3 in the fetal brain, not by direct transfer, but by stimulating Dio2 gene expression and DIO2 activity (137).

 

The reason why the fetal rodent brain is not permeable to T3 is unknown. One possibility would be lack of expression of the T4 and T3 transporter MCT8, but the fetal brain expresses high concentrations of MCT8, even larger than in the postnatal period (138). One possible mechanism would be selective T3 degradation by DIO3 after crossing the brain barriers. Different cellular transport routes for T4 and T3 could be involved in the selectivity of this mechanism. Knocking out Dio3 increases the sensitivity of the fetal and postnatal mouse brain to thyroid hormones (139-141).

 

STUDIES IN HUMANS

 

In the developing human cerebral cortex, DIO2 mRNA and activity are present already by GW7-GW8 (142). The relative expression of DIO2 and DIO3 is an important determinant of the T3 concentrations as development proceeds and has a strong regional component, as shown by Kester et al. (76) (Fig. 7).

Fig. 7. T3 concentrations in the fetal cortex and cerebellum respectively at different gestation weeks. Data extracted from (76).

 

These authors measured T4 and T3 concentrations and deiodinase activities in several brain regions from GW12-GW20 fetuses. The cerebral cortex had a progressive increase in T3 concentrations (Fig. 3), which correlated with an increased concentration of T4 and DIO2 activity. The parallel increase of T4 and DIO2 activity indicated an increased DIO2 transcription since T4 inhibits DIO2 activity. DIO3 activity was very low, near the limit of detection. The cerebellum showed a completely different picture: T3 was very low, and D3 activity was the highest among all regions. The difference between the cortex and the cerebellum (Fig. 7) supports the notion that the relative expression of DIO2 and DIO3 regulate the tissue response to thyroid hormones during development.

 

These changes occur in parallel to the accumulation of the nuclear receptor (77), indicating an increased sensitivity of the cortex to thyroid hormones from GW12 onwards (Fig. 3). This occurs after the preplate split, i.e., when the first migrating neurons start the formation of the cortical plate overlapping the period of neurogenesis and cell migration. Gliogenesis and the generation of astrocytes, the main cells expressing DIO2, occur around GW25 in the cortex (143).

 

The cells expressing DIO2 before astrocytes are formed are the radial glial cells (96,118), especially the outer radial glial cells (Fig. 8). This is most surprising because the outer radial glial cells, the universal stem cells of the cortex, are especially abundant in the cortex of primates, including humans. They are responsible for the enlargement of the subventricular zone and cortex expansion in these species (144). Furthermore, there is tightly correlated coexpression of DIO2 and the T4 transporter SLCO1C1 (OATP1C1) (Fig. 9) (118), indicating that the outer radial glia is the origin of the T3 formed locally in the human cortex at midgestation.

Fig. 8. A scheme representing a cross-section of the human fetal cerebral cortex at 18 -19 weeks of gestation. MZ, marginal zone; CP, cortical plate; IZ, intermediate zone; OSVZ, outer subventricular zone; ISVZ, inner subventricular zone; VZ, ventricular zone. In red, the ventricular or apical radial glia, with their bodies located in the ventricular zone, extending an apical process to the ventricular Surface and a basal process to the Surface of the brain. In green, the outer radial glia, lacking the apical process. Migrating neurons arriving at the cortical plate along the radial glial processes. In yellow, intermediate progenitors in the internal subventricular zone. Drawing reproduced with permission from (144).

 

The locally produced T3 could be involved in neurogenesis or could act on the newly formed neurons, which express THRA (145), or interneurons expressing THRB (118) to regulate their migration or terminal differentiation. T4 could reach the radial glia from the circulation. Alternatively, T4 could reach the apical processes of the ventricular radial glia by crossing the neuroepithelium from the inner CSF, or their basal processes from the outer CSF in the subarachnoid space (146). After the onset of gliogenesis, astrocytes express DIO2 (96,118). In contrast to the specific expression of SLCO1C1/OATP1C1, SLC16A2/MCT8 is widely expressed in most cell types (Fig. 9).

Fig. 9. Single-cell transcriptome profiling of DIO2, SLCO1C1, SLC16A2, THRA, and THRB expression (data from reference 118). In color are bulk cell clusters obtained using Uniform Manifold Approximation and Projection plots from the single-cell RNA sequencing datasets of human fetuses at gestational weeks 17–18 by Polioudakis et al. (147). Endo: endothelial cells, DIV: proliferating cells, ExM-U: excitatory neurons upper cortex enriched, InCGE: interneurons from the caudal ganglionic eminence, InMGE: interneurons from the medial ganglionic eminence, IP: intermediate progenitors, Mg: microglia, Neu: excitatory projection neurons, OPC: oligodendrocyte precursor cells, oRG: outer radial glial cells, Per: pericytes, vRG: ventricular radial glial cells. Blue dots represent single cells. DIO2 and SLCO1C1 coexpress in the outer radial glial cells. DIO2 is also expressed in some interneurons.

 

THYROID HORMONE RECEPTORS IN THE BRAIN

 

Flamant and coworkers have reviewed the relative roles of the receptor isoforms on developing and adult brain, using mouse genetic models (148,149). Two different genes THRA and THRB encode two receptor types, alpha (THRA1) and beta (THRB1 and THRB2), respectively. THRA also encodes THRA2, which does not bind T3 and is not a T3 receptor.

 

The cellular and regional complexity of the brain and its asynchronous maturation requires a previous understanding of the ontogeny and patterns of expression of the receptors to understand their physiological role. The low number of receptor molecules per nucleus, and the relative lack of specificity of receptor antibodies have precluded their use to analyze receptor expression by conventional immunohistochemistry. Analysis of the regional and cellular expression requires in situ hybridization histochemistry. This technique provides data on receptor mRNA expression that may not reflect accurately the concentration of the receptor protein (150,151). 

 

Studies on receptor protein concentration used T3 binding assays in nuclei isolated from whole brain or brain regions, with a sensitivity of about 100-200 molecules/nucleus. Binding assays detect indistinctly the THRA1 and THRB subtypes, but discrimination between THRA1 and THRB may be possible to some extent using a combination of T3 binding and immunoprecipitation, or by competitive binding assays using the acetic acid derivative triac. THRA1 has the same affinity for triac as for T3, and THRB up to ten-fold higher affinity for triac than for T3 (152). Receptor expression studies have also used knock-in mice in which the receptor protein was tagged with green fluorescent protein (GFP) (145), or with hemagglutinin epitopes (151).

 

There are discrepancies between the cellular abundance of receptor-encoded mRNAs and the corresponding receptor proteins (150). At the mRNA level, all receptor isoforms are present in the brain. Apart from the highly abundant, non-receptor encoding, Thra2 transcript, the predominant receptor isoform is Thra1, widely distributed in the CNS from E14 to adulthood (151,153-155). From E19 to P0, Thra1 is present in the outer part of the cerebral cortex and hippocampal CA1 field. During the late fetal stage, Thra1 is present in the piriform cortex, superior colliculus, pyramidal cell layer of the hippocampus, and the granular layer of the dentate gyrus. In adult rodents, Thra1 expression is prominent in the cerebral cortex, cerebellum, hippocampus, striatum, and olfactory bulb. Knock-in mice studies with green fluorescent protein-tagged THRA1 (145) showed that most neurons express the THRA1-GFP as they become postmitotic in tanycytes of the third ventricle and very low expression in astrocytes. In the cerebellum, the conjugated protein was present in migrating granule cells of the molecular layer and mature granule cells of the internal granule cell layer but not in the proliferating cells of the external germinal layer (also known as the external granular layer). Juvenile, but not adult Purkinje cells expressed THRA1-GFP. In the study by Minakhina et al. (151) the hemagglutinin-tagged THRA1 protein was the predominant receptor subtype in the mouse brain, and THRA2 concentration was 5-10-fold higher.

 

In cultured cells T3 receptors are present in neurons, astrocytes, oligodendrocytes, sensory neurons, Schwann cells, and microglia (156-160). Primary cultures of mouse astrocytes, which respond transcriptionally to T3, express Thra and Thrb (7). Receptor-specific actions can be demonstrated on some genes such as Dio3, which is specific for THRA1 (161-164). Some cells express specific receptor isoforms, for example, the mouse retina expresses Thra1 widely whereas immature photoreceptor cells selectively express Thrb1 (165,166).

 

Thrb1 expression during rodent development is low during the fetal period and increases during the postnatal period and through adulthood.  Between E17 and E20, only low levels are present in the brain, especially in the hippocampal pyramidal layer. On P0, an increase occurs in the accumbens, striatum, and hippocampus. From around P7 Thrb1 appears in the cerebral cortex. The patterns of expression of Thra1 and Thrb1 overlap (Fig. 10), but in some cells, one of the isoforms is predominant.

Fig. 10. T3 receptor mRNA expression in the mouse brain, by in situ hybridization with Thra1 and Thrb1-specific probes. In the cerebrum (left panels) there is an overlapping distribution of both receptor subtypes, with some differences in the hippocampus, amygdala, and hypothalamus. In the cerebellum (right panels) Thra1 is expressed in the granular layer (left upper panel), whereas Thrb1 is expressed in the Purkinje cell layer.

 

As an example, in the cerebellum, differentiated granular cells express Thra1 while Purkinje cells express Thrb1. Thrb2, which is abundant in the pituitary, is also expressed in the retina and the hypothalamus (166,167). During fetal stages, low levels of Thrb2 are present in the striatum (153). The web resource of the Allan Brain Institute (https://celltypes.brain-map.org/rnaseq/mouse_ctx-hip_smart-seq) provides a tool to explore the specific distribution of Thra1 and Thrb during mouse development.

 

Ontogeny of Thyroid Hormone Receptors in the Developing Brain

 

T3 binding assays detect the T3 receptor for the first time in the rat brain at E13.5-E14.5 and then increase, reaching stable concentrations from E17 to P0 (168). In close agreement, studies in knock-in mice expressing a conjugated TRα1-green fluorescent protein detect the receptor for the first time at E13.5 in the cortical plate (145). This date marks the onset of brain responsiveness to thyroid hormones (141). The receptor increases at birth and the highest concentrations are reached at P6 (169,170), but receptor occupancy is maximal at P15 (171).

 

In the human brain, the receptor protein quantified by nuclear binding assays, and receptor mRNAs by PCR, are detected during the first trimester (77,142,172). The receptor protein, with a binding profile typical of THRB, is present at low levels in the fetus around GW10 and increases in concentration 10-fold up to GW18-GW18 (77) (Fig. 3), during neurogenesis and neuron migration. At these stages, subsets of CALB2 (calretinin) and SST(somatostatin) interneurons selectively express THRB (Fig. 9) (118), but its contribution to the total receptor protein measured by binding assays is unknown.

 

Occupancy of receptors with the ligand in the brain antecedes the occupancy in other tissues. In the human brain, the T3 ligand is present from GW10, at concentrations enough to result in about 25% occupancy of receptor (77,173), and when no T3 is detected in other organs. As stated above, this is due to the early presence of DIO2 activity (76).  A similar situation occurs in fetal lambs at gestational day 100. The brain had a 74% receptor occupancy compared with 10% in the liver and lung (174). In rats total receptor occupancy by the hormone increases in parallel with the postnatal increase in plasma and cytosol total and free T3, and reach a maximum of 50-60% at postnatal day 15 (171).

 

THYROID HORMONE ACTION ON BRAIN GENE EXPRESSION

 

Aporeceptor and Holoreceptor

 

Thyroid hormones regulate the expression of many genes in the rodent brain, mainly during the postnatal period, but the brain is also sensitive in the fetus and in adult animals (6,7,107,175-182). T3 interacting with the nuclear receptors, i.e., the holoreceptor, has a modulatory role on the receptor transcriptional activity, upregulating or downregulating gene expression. In the absence of the hormone, the aporeceptor has intrinsic transcriptional activity, relevant in events such as amphibian metamorphosis (183), PC12 cell differentiation (184) or development of the inner ear in mice (185). To some extent, the hypothyroid phenotype is the consequence of aporeceptor activity (30,186). Similarly, mutations of receptors that abolish T3 binding cause developmental abnormalities (166,187,188) (see the chapter on the syndromes of resistance to thyroid hormones).

 

Non-genomic Actions

 

It is unclear to what extent non-genomic actions might also mediate effects of T4 and T3 in the brain (189-191). Interaction of T4 with integrin αvβ3 might have a role in the expansion of progenitors in the neocortex (192). Proposed non-genomic actions of T3 include the maturation and plasticity of hippocampal pyramidal neurons (193), and the regulation of the onset and duration of the sensitive period of imprinting in chicks (194).

 

For many genes, the role of thyroid hormones during development is to regulate the timing of gene expression with a strong regional specificity, for example on the myelin genes (46) (see also Fig. 1). In the absence of DIO3, developmental expression of some T3 target genes is accelerated (141). The response of many target genes is region-dependent and shows partial overlap in different brain areas (9). Even within the same area, the same gene may be responsive only in a fraction of cells, as shown by Nrgn (RC3/neurogranin (175)). Transcription of thyroid hormone target genes is likely the result of the combinatorial activity of transcription factors, which include the T3 receptor whose relative weight may depend on the specific region and developmental period. It also reflects the diversity existing within apparently homogeneous cell classes.

 

Thyroid Hormone-Responsive Elements

 

Early studies showed the presence of thyroid hormone-responsive elements in the promoter or intronic regions of thyroid hormone-dependent brain genes. To name a few, the myelin basic protein (195); the Purkinje cell-specific gene PCP2 (196); the calmodulin-binding and PKC substrate RC3 (197); the prostaglandin D2 synthetase (198,199); the transcription factor Hairless (200); the neuronal cell adhesion molecule NCAM (201); and the early response gene NGFI-A (202). More recently, Chromatin immunoprecipitation assays identified receptor binding sites in the developing mouse cerebellum (203) or cerebellar cell lines expressing THRA1 or THRB1 (162). Other targets are regulated at the levels of mRNA stability (acetylcholinesterase), protein translation (MAP2 (204)) or mRNA splicing (TAU (205)). Regulation of splicing might be due to a primary action on the transcription of splicing regulators (206).

 

Genes Responsive to Thyroid Hormones in the Fetal and Postnatal Rodent Brain

 

Thyroid hormone regulation of several genes occurs in the rodent fetal brain in vivo (207-209). In the fetal cerebral cortex at the end of gestation, thyroid hormone controls the expression of genes involved in the biogenesis of the cytoskeleton, cell migration, and branching of neurites. In some studies, a large percentage of the thyroid hormone-dependent genes were related to Camk4 signaling pathways (63,210). Camk4 is regulated by T3 in cultured primary neurons (211,212), but is not in the postnatal rat brain in vivo (213). Results of extensive analysis by RNA-Seq of primary embryonal cerebrocortical cultures disclosed many thyroid hormone targets with roles during cortical development (6,20). The responsiveness of the fetal brain to thyroid hormones increases in Dio3 knockout mice (139,141).

 

Many of the thyroid hormone-regulated genes identified during the postnatal period in the rodent brain are sensitive to the hormone only during a time window that spans the first 2-3 weeks after birth. Many of these genes are not dependent on thyroid hormone in the fetal or in the adult brain, possibly due to a DIO3-related mechanism (141). Chatonnet et al. (9), performed an analysis of the published data on thyroid hormone action on gene expression in the brain and cultured cells and arrived at a list of at least 37 genes consistently found in different studies as targets of the T3 receptors. Some of them are candidates for transcriptional regulation because they contain a thyroid hormone-responsive element. These genes include Adamtsl4, Dbp, Fos, Hr, Kcna1, Klf9, Scd1, Stat5a, and Txnip. Additional genes found regulated transcriptionally by T3 in several independent studies are Shh, Hr, Dbp, Gbp3, and Nrgn.

 

In our studies, employing RNA-Seq of primary cerebrocortical cells (6) we found that T3 up-or down-regulated up to 7.7% of expressed genes 24 hours after treatment. T3 was active in the presence of cycloheximide on about 30% of these genes indicating an effect on transcription. No response to T3 occurred in similar cells from Thra and Thrbknockout mice, as expected for receptor-mediated actions. Additionally, a large proportion of the T3-responsive genes in the presence of cycloheximide contained T3-responsive elements (162). The estimation is that T3 regulates around 2.5% of all expressed cellular genes and at least 1% at the transcriptional level through a T3 responsive element.

 

Brain deprivation of thyroid hormones caused by systemic hypothyroidism causes larger changes of gene expression than compound inactivation of the MCT8 and OATP1C1 transporters, or inactivation of MCT8 and DIO2. In the MCT8 plus OATP1C1 deficiency, the transport of T4 and T3 is severely compromised, resulting in isolated brain hypothyroidism with low tissue T4 and T3 (66). In the MCT8 plus DIO2 deficiency, the transport of T3 and the generation of local T3 from T4 are blocked, leading also to brain hypothyroidism buy with normal T4 and low T3. Contrary to what would be expected, the changes in gene expression are not equivalent in these two models of brain hypothyroidism, which are also different from systemic hypothyroidism. The latter causes altered expression of about the double number of genes in the cerebral cortex and the striatum than the cerebral hypothyroidism of the double knockouts. One reason is that systemic hypothyroidism causes many effects on gene expression that are secondary to other actions of T3 elsewhere in the body. When transcriptionally-regulated genes were compared, similar profiles were found in the three conditions. In systemic hypothyroidism, among the transcriptional targets of T3, the most affected downregulated genes in the cortex were Kcnj10, Hr, Ky, Cyp11a1, Nefm, Npt, Stac2, Hcrtr1, Shh, Prlr, Sema7a, and upregulated, Aldh1a3, Adamts18, Ntf3, Mc4r, Dio2, Trhr. In the striatum, the most affected downregulated genes were Prlr, Cyp11a1, Cnr1, Sema7a, Gls2, Shh, Igsf9, Enpp2, Gdf10, Arg2, Nefm, and upregulated, Cyp26b1, Syt10, Trhr, Mc4r, Dio2, Gabra5.

 

Interactions with Glucocorticoids and Retinoids

 

The interactions of thyroid hormones with other hormonal systems (214) might be relevant to their actions on neural cells. In primary cerebrocortical cultures, one of the most significant processes regulated by T3 was the control of G-protein-coupled receptor activity (164), estimated to be important in the transition from fetal type to adult-type gene expression. A detailed analysis of these interactions is beyond the scope of this chapter, and only one example is mentioned related to the crosstalk with glucocorticoids and retinoid signaling pathways. T3 controls the expression of several enzymes involved in retinoic acid (RA) metabolism: the RA synthesizing enzymes ALDH1A1 and ALDH1A3, and the degrading enzyme CYP26B1 (9,163). The final effect on RA concentrations depends on the relative expression of each of the enzymes, which have developmental and regional variations, and on glucocorticoid signaling. Aldh1a1 is upregulated by T3, preferentially through TRα1, and glucocorticoids potentiated the effect of T3. Additionally, T3 controls the expression of Nr3c1, the glucocorticoid receptor. Aldh1a3 is downregulated by T3 in primary cells and increases in expression in hypothyroidism as indicated in the previous paragraph. On the other hand, Cyp26b1 is upregulated by T3 in primary cells and is downregulated in the hypothyroid cortex but upregulated in the hypothyroid striatum. By increasing ALDH1A1, especially in the presence of glucocorticoids, T3 will increment RA concentrations, whereas acting on ALDH1A3 and CYP26B1, T3 will reduce RA concentrations. A recent study on Dio3 knockout mice has found evidence for these interactions (141).

 

Actions on Gene Expression in the Adult Brain

 

In adult subjects, thyroid hormones influence mood and behavior, and thyroid dysfunction affects neurotransmitter systems (215) often leading to psychiatric disorders (216). High doses of T4 improve mood in bipolar depression (217,218). In the adult rat striatum, administration of a large single T3 dose leads to up-regulation of 149 genes and down-regulation of 88 genes (177). Physiological doses of T3 given for several days to hypothyroid animals led to up-regulation of 18 genes, and down-regulation of just one gene. Therefore, acute large doses of thyroid hormone cause large changes in gene expression, with more modest changes with lower doses. Some of the regulated genes are related to circadian rhythms and wakefulness, with one of them (Dbp or D-site binding protein) proposed as a candidate gene in bipolar disorders (219), and likely to be regulated directly by TRα1 (176). Many other genes were involved in striatal physiology as components of several signaling pathways. Fig. 11 shows a putative model of T3 action on the adult striatum.

Fig. 11. Regulation of gene expression by thyroid hormones in the adult striatum. Signaling pathways are schematically represented and the main groups of regulated genes are shown in numerals. 1: G-protein coupled receptor signaling (Cnr1, Rgs9, Rasd2, Rasgrp1). 2: Ca2+/calmodulin pathway (Nrgn); MAPK pathways (Map2k3, Fos). 4: Early genes (Nr4a1, Arc, Dusp1, Egr1, Homer). 5: Ion channels (Scn4b). Abbreviations: VDCC, voltage-dependent sodium channels, NMDA, N-methyl-D-aspartate, D1 and D2, dopamine receptors 1 and 2.  From (177).

 

ACTIONS OF THYROID HORMONES ON SPECIFIC DEVELOPMENTAL EVENTS

 

There are several possible approaches for studying the role of thyroid hormones on brain development. One way would be to analyze the effects of hypothyroidism and thyroid hormone treatment on the development of the cerebral cortex, the striatum, the cerebellum, the hippocampus, and other regions. Another way is to analyze common events occurring in the different brain regions, such as neurogenesis, cell migration, and differentiation, among others. In this review, we have opted for this second approach, with a focus on the T3 regulation of genes involved in these processes. Reviews on the cerebral cortex, and the cerebellum are available (20,21,23).

 

Neurogenesis

 

Generation of neurons, or neurogenesis, starts in humans around the fifth gestational week (GW5, Fig. 3), and in mice around embryonic day 10 (E10). The bulk of neurogenesis takes place until GW25 or E16/17 respectively, partially overlapping gliogenesis. The granular cell layers of the hippocampus, olfactory bulb, and cerebellum continue accumulating neurons postnatally, a reason why they are especially sensitive to thyroid function. At the onset of neurogenesis, the neuroepithelial cells sequentially express Pax6, Neurog1/2, and NeuroD, undergoing glutamatergic identity, and under the influence of FGF10 undergo a fast transition to radial glia (20,220). These cells remain attached by an apical process to the ventricular surface and elongate as the embryonic brain epithelium thickens, adopting a bipolar morphology (Fig. 8). In the cortex, the elongated basal process reaches the pia providing a scaffold for cell migration (221,222). In addition to this structural and supportive function, the radial glia is the universal cortical stem cell that generates all neurons and glia, directly or through intermediate precursors. (220). In primates, enlargement of the cortex is due to the accumulation of a population of radial glial cells that lose the apical process and accumulate in the outer part of an enlarged subventricular zone (Fig. 8). Human-specific changes occur with further enlargement of the cortex over the great apes (223).

 

The role of thyroid hormones on proliferation and differentiation of neural precursors in the embryonic neurogenic areas has been shown during tadpole premetamorphosis (224) and concerning the effects of maternal thyroid hormones (192,225,226). NeuroD, mentioned above, is sensitive to thyroid hormones in the cerebellum (227), and THRA1 mutations cause abnormal proliferation and adhesion of human cortical progenitors (228). Iodine deficiency in rats affects hippocampal radial glial cells (229). The first neurons originating in the cortex, the Cajal-Retzius cells, and the subplate cells, are sensitive to thyroid hormones in rodents and in humans (31,49,210).

 

Limited neurogenesis also occurs in the adult brain, related in humans to neuropsychiatric conditions, cognitive deficits, and depression. Adult neurogenesis takes place in two structures: the subventricular zone, located underneath the surface of the lateral ventricles, and the subgranular zone, adjacent to the granular layer of the hippocampal dentate gyrus. The subventricular zone generates olfactory bulb interneurons in adult rodents, and in humans provides new interneurons to the adjacent striatum (230). The subgranular zone generates dentate gyrus granular neurons in adult rodents and humans (231). Hypothyroidism and thyroid hormones influence neurogenesis in the rodent subventricular zone and subgranular zone (224,225,232-241).

 

Neural Cell Differentiation

 

Thyroid hormone controls the expression of many genes with roles on terminal cell differentiation, such as cell cycle regulators, cytoskeletal proteins, neurotrophins, and neurotrophin receptors, and extracellular matrix proteins. Among the cell cycle regulators, E2F1, p53, cyclins, and cyclin-dependent kinase inhibitors are regulated by thyroid hormone in cell culture (242-244).

 

Neural cell shape is determined by the cytoskeleton, which consists of microtubules (tubulin), microfilaments (actin), and intermediate filaments, specific for neurons (neurofilaments), glia (glial fibrillary acidic protein), or maturing cells (vimentin, nestin). Tubulins α1 and α2 are downregulated by thyroid hormone, and tubulin β4 is upregulated (245,246). Microtubule-associated proteins (MAPs) are also under thyroid hormone control at a posttranscriptional level. For example, thyroid hormone regulates Map2 protein distribution in the Purkinje cell dendritic tree (204), and conversion of immature forms of the microtubule-associated protein TAU (MAPT) to mature forms by alternative splicing of the MAPT mRNA (205). The neurofilament genes Nefh, and Nefm are also under thyroid hormone control in the fetal and postnatal cerebral cortex (63,107).

 

Astrocytes (247,248) cerebellar Golgi epithelial cells (249), and microglia (160) are thyroid hormone targets. Thyroid hormones influence the in vivo expression of astroglial genes encoding tenascin C, laminin, and L1 adhesion molecule, with effects on neuronal migration and differentiation, and axonal fasciculation (250-252) . In vitro, β-adrenergic receptor antagonists block the effect of T3 on astrocyte differentiation (253). In another studies T3 upregulates Arrb1 (arrestin beta1), facilitating endocytosis of β2 adrenergic receptor (ADRB2) and ERK activation (254).

 

The control of neurotrophin expression might mediate some of the effects of thyroid hormone on differentiation and survival. Interactions between thyroid hormone and NGF are important for the growth and maintenance of cholinergic neurons in the basal forebrain (36). Unliganded THRA1 in PC12 cells expressing Thra1 blocks NGF-induced differentiation and the blockade is released by T3 (184), suggesting a possible mechanism for the control of differentiation timing. Changes in NGF, NTRK, P75NTR, BDNF, and NTF3 after hypothyroidism have been described (255-257).

 

Thyroid hormones influence myelination directly through effects on oligodendrocyte differentiation (258,259), and may also have indirect effects by stimulating axonal maturation, which is impaired in hypothyroidism (Fig. 1 right panel (260,261)). A lower axon diameter prevents reaching the critical size to become myelinated (262). Low axon diameter also occurred in a patient with MCT8 mutation (Fig. 3 (49)), which might contribute to the hypomyelination of these patients.

 

As thyroid hormones are required for terminal differentiation of oligodendroglial cells, they influence the expression of practically all myelin protein genes, but only during the myelination period. In the rat, this period extends from about the end of the first postnatal week up to the end of the first month, with a strong regional component in parallel with the wave of myelination (46,263). The control of myelin gene expression is transient, and in hypothyroidism, there is a developmental delay, but the effect on the myelin content is permanent (Fig. 1).

 

Early studies showed that T3 inhibits proliferation and promotes differentiation of oligodendrocyte precursor cells (OPC) (264,265) through repression of the E2F1 transcription factor (266). Transcriptomic analysis identified the universal T3-target gene Klf9 (Krüppel-like factor 9) as a mediator of the effect of T3 on OPC differentiation (267). The transcriptional repressors NCoR and HDAC repress the oligodendrocyte differentiation pathway. T3 relieves this repression and induces Sox10, needed for the maintenance of the differentiated state (268). Oligodendrocyte precursor cells express Thra1 and Thrb (269-271), but Thra1 is the predominant receptor gene expressed in the newly formed and myelinating oligodendrocytes.

 

Neural Cell Migration

 

Thyroid hormone also influences neuronal migration in the cerebral cortex, hippocampus, and cerebellum. Thyroid hormone deficiency during cortical development leads to less than the normal definition of cortical layers (260,272,273). Thyroid hormone influences the maturation of the radial glia, the path along which radial migration occurs in the cerebral cortex and the hippocampus (229).

 

An important cellular target of thyroid hormone is the Cajal-Retzius cell. These cells are located in the marginal zone, the future cortex layer 1, and are required for proper migration of neurons in the cerebral cortex and the hippocampus, cortical lamination, and establishment of synaptic connections (274-276).  These cells secrete reelin (RLN), an extracellular matrix protein under thyroid hormone control (31). RLN function is essential for the inside-out pattern of cerebral cortex development. The protein disabled (DAB), a component of the RLN signaling pathway is also under thyroid hormone control. The Rln and Dab1 genes are not regulated by T3 at the transcriptional level, but other genes expressed in Cajal-Retzius cells are transcriptional targets of T3 in primary neurons: these include Rgs4, Npnt, Ephb6, Clstn2, and Dnmt3a (6,20). Cajal-Retzius cells, therefore, appear to be important and selective cellular targets of thyroid hormones during development. Their number is low in perinatal hypothyroidism in rat pups (277), and in a human embryo with mutated MCT8 (49).

 

There are many extracellular matrix proteins regulated by thyroid hormones, with actions on cell migration and neuronal differentiation, synaptic plasticity, etc. The extracellular matrix proteins are heterogeneous, comprising laminin, fibronectin, collagen, neurotrophic factors, adhesion molecules, hyaluronan proteins, proteoglycans, and other components. As already mentioned, thyroid hormones regulate negatively tenascin C, laminin, and L1 (250-252), and the adhesion molecule NCAM (201,278). In addition to these proteins, regulated by T3 in vivo, T3 regulates many other genes of this heterogeneous group in cultured primary cerebrocortical cells (6) twenty-five of them at the transcriptional level. Among these, seven have thyroid hormone receptor responsive elements: Adamts2, Lingo3, Mfap3l, Bmp1, Megf10, Nav2, and Crim1. The function of these genes and the significance of their regulation has been discussed (20).

 

In the rodent cerebellum (279,280) thyroid hormones are involved in the late phase of granular cell migration from the external germinal layer (EGL) to the internal granular layer (IGL). This process takes place postnatally in rodents ending by P20 with the complete disappearance of the EGL. A characteristic feature of the hypothyroid cerebellum is a delay in the migration of granule cells and the persistence of the EGL beyond P20 (Fig. 1) (281). It is unclear how the absence of thyroid hormone interferes with granule cell migration. Cell migration in Thra1 knock-out mice proceeds normally, and there is no effect of hypothyroidism (30). This suggests that the migration defect in hypothyroid mice represents a non-physiological action of the unliganded receptor. On the other hand, these cells express Thra1 during the migration period (P7-P19) and not before (145), which might indicate an action of the receptor on granule cell maturation during migration. Several groups have described T3-regulated genes involved in different processes (9,163,282).

 

CONCLUSIONS

 

Thyroid hormone actions in the brain are extremely complex with a continuously changing landscape as development proceeds. Brain maturation involves continuous changes in cell composition, i.e., the target organ of thyroid hormone is under constant change. This is reflected in the changing regulated gene network. Genes that are transcriptional targets of T3 at a certain developmental time may be refractory at another time. The importance of thyroid hormone for the brain requires tightly controlled mechanisms of thyroid hormone delivery to the brain and cellular interactions in the metabolism of thyroid hormones, with crucial roles of DIO2 and DIO3 regulating the cellular concentration of T3.

 

 Disruption of these mechanisms results in syndromes of profound neurological impairment. The challenge is to understand in detail the mechanisms of action of thyroid hormones at different stages of development in the human brain, and not merely extrapolating from rodent models, for a better understanding of thyroid hormone action defects.

 

REFERENCES

 

  1. Joffe RT, Sokolov STH. Thyroid hormones, the brain, and affective disorders. Crit Rev Neurobiol. 1994; 8:45-63.
  2. Laureno R. Neurologic manifestations of thyroid disease. The Endocrinologist. 1996; 6:467-473.
  3. Ganguli M, Burmeister LA, Seaberg EC, Belle S, DeKosky ST. Association between dementia and elevated TSH: a community-based study. Biol Psychiatr. 1996; 40:714-725.
  4. Joffe RT, Pearce EN, Hennessey JV, Ryan JJ, Stern RA. Subclinical hypothyroidism, mood, and cognition in older adults: a review. Int J Geriatr Psychiatry. 2013; 28:111-118.
  5. Craske MG, Stein MB. Anxiety. Lancet. 2016; 388:3048-3059.
  6. Gil-Ibanez P, Garcia-Garcia F, Dopazo J, Bernal J, Morte B. Global Transcriptome Analysis of Primary Cerebrocortical Cells: Identification of Genes Regulated by Triiodothyronine in Specific Cell Types. Cereb Cortex. 2017; 27:706-717.
  7. Morte B, Gil-Ibanez P, Bernal J. Regulation of Gene Expression by Thyroid Hormone in Primary Astrocytes: Factors Influencing the Genomic Response. Endocrinology. 2018; 159:2083-2092.
  8. Laudet V. The origins and evolution of vertebrate metamorphosis. Curr Biol 2011; 21:R726–R737.
  9. Chatonnet F, Flamant F, Morte B. A temporary compendium of thyroid hormone target genes in brain. Biochim Biophys Acta. 2015; 1849:122-129.
  10. Anderson GW, Schoonover CM, Jones SA. Control of thyroid hormone action in the developing rat brain. Thyroid. 2003; 13:1039-1056.
  11. Bernal J. Thyroid hormones and brain development. Vitam Horm. 2005; 71:95-122.
  12. Bernal J. Thyroid hormone receptors in brain development and function. Nat Clin Pract Endocrinol Metab. 2007; 3:249-259.
  13. Koibuchi N, Iwasaki T. Regulation of brain development by thyroid hormone and its modulation by environmental chemicals. Endocr J. 2006; 53:295-303.
  14. Morreale de Escobar G, Obregon MJ, Escobar del Rey F. Role of thyroid hormone during early brain development. Eur J Endocrinol. 2004; 151 Suppl 3:U25-37.
  15. Fisher DA, Polk DH. Maturation of thyroid hormone actions. In: Delange F, Fisher DA, Glinoer D, eds. Research in congenital hypothyroidism. New York: Plenum Press; 1988:61-77.
  16. Holt AB, Renfree MB, Chek DB. Comparative aspects of brain growth: a critical evaluation of mammalian species used in brain growth research with emphasis on the Tammar wallaby. In: Hetzel BS, Smith RM, eds. Fetal brain disorders. Recent approaches to the problem of mental deficiency. Amsterdam: Elsevier/North Holland; 1981:17-43.
  17. Morreale de Escobar G, Escobar del Rey F, Ruiz-Marcos A. Thyroid hormone and the developing brain. In: Dussault JH, Walker P, eds. Congenital Hypothyroidism. New York: Marcel Decker, Inc.; 1983:85-126.
  18. Workman AD, Charvet CJ, Clancy B, Darlington RB, Finlay BL. Modeling transformations of neurodevelopmental sequences across mammalian species. J Neurosci. 2013; 33:7368-7383.
  19. Romijn HJ, Hofman MA, Gramsbergen A. At what age is the developing cerebral cortex of the rat comparable to that of the full-term newborn human baby? Early Hum Dev. 1991; 26:61-67.
  20. Bernal J. Thyroid hormone regulated genes in cerebral cortex development. J Endocrinol. 2017; 232:R83-R97.
  21. Leto K, Arancillo M, Becker EB, Buffo A, Chiang C, Ding B, Dobyns WB, Dusart I, Haldipur P, Hatten ME, Hoshino M, Joyner AL, Kano M, Kilpatrick DL, Koibuchi N, Marino S, Martinez S, Millen KJ, Millner TO, Miyata T, Parmigiani E, Schilling K, Sekerkova G, Sillitoe RV, Sotelo C, Uesaka N, Wefers A, Wingate RJ, Hawkes R. Consensus Paper: Cerebellar Development. Cerebellum. 2016; 15:789-828.
  22. Chatonnet F, Picou F, Fauquier T, Flamant F. Thyroid hormone action in cerebellum and cerebral cortex development. J Thyroid Res. 2011; 2011:145762.
  23. Berbel P, Navarro D, Roman GC. An evo-devo approach to thyroid hormones in cerebral and cerebellar cortical development: etiological implications for autism. Front Endocrinol (Lausanne). 2014; 5:146.
  24. Legrand J. Effects of thyroid hormones on Central Nervous System. In: Yanai J, ed. Neurobehavioral Teratology. Amsterdam: Elsevier Science Publishers; 1984:331-363.
  25. Eayrs JT. Developmental relationships between brain and thyroid. In: Michael RP, ed. Endocrinol. Hum. Behav. London: Oxford University Press; 1968:239-255.
  26. Madeira MD, Paula-Barbosa M, Cadete-Leite A, Tavares MA. Unbiased estimate of hippocampal granule cell numbers in hypothyroid and in sex-age-matched control rats. J Hirnforsch. 1988; 29:643-650.
  27. Madeira MD, Cadete-Leite A, Andrade JP, Paula-Barbosa MM. Effects of hypothyroidism upon the granular layer of the dentate gyrus in male and female adult rats: a morphometric study. J Comp Neurol. 1991; 314:171-186.
  28. Madeira MD, Pereira A, Cadete-Leite A, Paula-Barbosa MM. Estimates of volumes and pyramidal cell numbers in the prelimbic subarea of the prefrontal cortex in experimental hypothyroid rats. J Anat. 1990; 171:41-56.
  29. Berbel P, Navarro D, Auso E, Varea E, Rodriguez AE, Ballesta JJ, Salinas M, Flores E, Faura CC, de Escobar GM. Role of late maternal thyroid hormones in cerebral cortex development: an experimental model for human prematurity. Cereb Cortex. 2010; 20:1462-1475.
  30. Morte B, Manzano J, Scanlan T, Vennstrom B, Bernal J. Deletion of the thyroid hormone receptor alpha 1 prevents the structural alterations of the cerebellum induced by hypothyroidism. Proc Natl Acad Sci U S A. 2002; 99:3985-3989.
  31. Alvarez-Dolado M, Ruiz M, del Rio JA, Alcantara S, Burgaya F, Sheldon M, Nakajima K, Bernal J, Howell BW, Curran T, Soriano E, Muñoz A. Thyroid hormone regulates reelin and dab1 expression during brain development. J Neurosci. 1999; 19:6979-6973.
  32. Berbel P, Marco P, Cerezo JR, DeFelipe J. Distribution of parvalbumin immunoreactivity in the neocortex of hypothyroid adult rats. Neurosci Lett. 1996; 204:65-68.
  33. Gilbert ME, Sui L, Walker MJ, Anderson W, Thomas S, Smoller SN, Schon JP, Phani S, Goodman JH. Thyroid hormone insufficiency during brain development reduces parvalbumin immunoreactivity and inhibitory function in the hippocampus. Endocrinology. 2007; 148:92-102.
  34. Manzano J, Cuadrado M, Morte B, Bernal J. Influence of thyroid hormone and thyroid hormone receptors in the generation of cerebellar GABAergic interneurons from precursor cells. Endocrinology. 2007;
  35. Richard S, Guyot R, Rey-Millet M, Prieux M, Markossian S, Aubert D, Flamant F. A Pivotal Genetic Program Controlled by Thyroid Hormone during the Maturation of GABAergic Neurons. iScience. 2020; 23:100899.
  36. Gould E, Butcher LL. Developing cholinergic basal forebrain neurons are sensitive to thyroid hormone. J Neurosci. 1989; 9:3347-3358.
  37. Dusart I, Flamant F. Profound morphological and functional changes of rodent Purkinje cells between the first and the second postnatal weeks: a metamorphosis? Front Neuroanat. 2012; 6:11.
  38. Legrand J. Analyse de l'action morphogénétic des hormones thyroïdiennes sur le cervelet du jeune rat. Arch Anat Microsc Morphol Exp. 1967; 56:205-244.
  39. Ruiz-Marcos A, Sanchez-Toscano F, Obregón MJ, Escobar del Rey F, Morreale de Escobar G. Reversible morphological alterations of cortical neurons in juvenile and adult hypothyroidism in the rat. Brain Res. 1980; 185:91-102.
  40. Ruiz-Marcos A, Sanchez-Toscano F, Obregón MJ, Escobar del Rey F, Morreale de Escobar G. Thyroxine treatment and recovery of hypothyroidism-induced pyramidal cell damage. Brain Res. 1982; 239:559-574.
  41. Gould E, Allan MD, McEwen BS. Dendritic spine density of adult hippocampal pyramidal cells is sensitive to thyroid hormone. Brain Res. 1990; 525:327-329.
  42. Ruiz-Marcos A, Cartagena P, García A, Escobar del Rey F, Morreale de Escobar G. Rapid effects of adult-onset hypothyroidism on dendritic spines of pyramidal cells of the rat cerebral cortex. Exp Brain Res. 1988; 73:583-588.
  43. Balazs R, Brooksbank BWL, Davison AN, Eayrs JT, Wilson DA. The effect of neonatal thyroidectomy on myelination in the rat brain. Brain Res. 1969; 15:219-232.
  44. Malone MJ, Rosman NP, Szoke M, Davis D. Myelination of brain in experimental hypothyroidism. An electron-microscopic and biochemical study of purified myelin isolates. J Neurol Sci. 1975; 26:1-11.
  45. Noguchi T, Sugisaki T. Hypomyelination in the cerebrum of the congenitally hypothyroid mouse (hyt). J Neurochem. 1984; 42:891-893.
  46. Rodriguez-Pena A, Ibarrola N, Iniguez MA, Munoz A, Bernal J. Neonatal hypothyroidism affects the timely expression of myelin- associated glycoprotein in the rat brain. J Clin Invest. 1993; 91:812-818.
  47. Adamo AM, Aloise PA, Soto EF, Pasquini JM. Neonatal Hyperthyroidism in the Rat Produces an Increase in the Activity of Microperoxisomal Marker Enzymes Coincident with Biochemical Signs of Accelerated Myelination. J Neurosci Res. 1990; 25:353-359.
  48. Guadaño-Ferraz A, Escobar del Rey F, Morreale de Escobar G, Innocenti GM, Berbel P. The development of the anterior commissure in normal and hypothyroid rats. Dev Brain Res. 1994; 81:293-308.
  49. Lopez-Espindola D, Morales-Bastos C, Grijota-Martinez C, Liao XH, Lev D, Sugo E, Verge CF, Refetoff S, Bernal J, Guadano-Ferraz A. Mutations of the thyroid hormone transporter MCT8 cause prenatal brain damage and persistent hypomyelination. J Clin Endocrinol Metab. 2014; 99:E2799-2804.
  50. Morreale de Escobar G, Obregón MJ, Escobar del Rey F. Fetal and maternal thyroid hormones. Horm Res. 1987; 26:12-27.
  51. Morreale de Escobar G, Pastor R, Obregón MJ, Escobar del Rey F. Effects of maternal hypothyroidism on the weight and thyroid hormone content of rat embryonic tissues. Endocrinology. 1985; 117:1890-1901.
  52. Nataf B, Sfez M. Debut du fonctionnement de la thyroide foetale du rat. Soc Biol. 1961; 156:1235-1238.
  53. Obregón MJ, Mallol J, Pastor R, Morreale de Escobar G, Escobar del Rey G. L-thyroxine and 3,3',5-triiodo-L-thyronine in rat embryos before onset of fetal thyroid function. Endocrinology. 1984; 114:305-307.
  54. Porterfield SP, Hendrich CE. Tissue iodothyronine levels in fetuses of control and hypothyroid rats at 13 and 16 days gestation. Endocrinology. 1992; 131:195-200.
  55. Woods RJ, Sinha AK, Ekins RP. Uptake and metabolism of thyroid hormones by the rat foetus early in pregnancy. Clin Sci. 1984; 67:359-363.
  56. Morreale de Escobar G, Calvo R, Obregón MJ, Escobar del Rey F. Contribution of maternal thyroxine to fetal thyroxine pools in normal rats near term. Endocrinology. 1990; 126:2765-2767.
  57. Obregón MJ, Ruiz de Oña C, Calvo R, Escobar del Rey F, Morreale de Escobar G. Outer ring iodothyronine deiodinases and thyroid hormone economy: responses to iodine deficiency in the rat fetus and neonate. Endocrinology. 1991; 129:2663-2673.
  58. Vulsma T, Gons MH, de Vijlder J. Maternal-fetal transfer of thyroxine in congenital hypothyroidism due to a total organification defect or thyroid dysgenesis. N Eng J Med. 1989; 321:13-16.
  59. van Trotsenburg P, Stoupa A, Leger J, Rohrer T, Peters C, Fugazzola L, Cassio A, Heinrichs C, Beauloye V, Pohlenz J, Rodien P, Coutant R, Szinnai G, Murray P, Bartes B, Luton D, Salerno M, de Sanctis L, Vigone M, Krude H, Persani L, Polak M. Congenital Hypothyroidism: A 2020-2021 Consensus Guidelines Update-An ENDO-European Reference Network Initiative Endorsed by the European Society for Pediatric Endocrinology and the European Society for Endocrinology. Thyroid. 2021; 31:387-419.
  60. de Zegher F, Pernasetti F, Vanhole C, Devlieger H, Van den Berghe G, Martial JA. The prenatal role of thyroid hormone evidenced by fetomaternal Pit-1 deficiency. J Clin Endocrinol Metab. 1995; 80:3127-3130.
  61. Yasuda T, Ohnishi H, Wataki K, Minagawa M, Minamitami K, Niimi H. Outcome of a baby born from a mother with acquired juvenile hypothyroidism having undetectable thyroid hormone concentrations. J Clin Endocrinol Metabol. 1999; 84:2630-2632.
  62. Morreale de Escobar G, Pastor R, Obregon MJ, Escobar del Rey F. Effects of maternal hypothyroidism on the weight and thyroid hormone content of rat embryonic tissues, before and after onset of fetal thyroid function. Endocrinology. 1985; 117:1890-1900.
  63. Morte B, Diez D, Auso E, Belinchon MM, Gil-Ibanez P, Grijota-Martinez C, Navarro D, de Escobar GM, Berbel P, Bernal J. Thyroid hormone regulation of gene expression in the developing rat fetal cerebral cortex: prominent role of the Ca2+/calmodulin-dependent protein kinase IV pathway. Endocrinology. 2010; 151:810-820.
  64. Ausó E, Lavado-Autric R, Cuevas E, Del Rey FE, Morreale De Escobar G, Berbel P. A moderate and transient deficiency of maternal thyroid function at the beginning of fetal neocorticogenesis alters neuronal migration. Endocrinology. 2004; 145:4037-4047.
  65. Goodman JH, Gilbert ME. Modest thyroid hormone insufficiency during development induces a cellular malformation in the corpus callosum: a model of cortical dysplasia. Endocrinology. 2007; 148:2593-2597.
  66. Morte B, Gil-Ibanez P, Heuer H, Bernal J. Brain Gene Expression in Systemic Hypothyroidism and Mouse Models of MCT8 Deficiency: The Mct8-Oatp1c1-Dio2 Triad. Thyroid. 2021; 31:985-993.
  67. Shepard TH. Onset of function of the human fetal thyroid: biochemical and autoradiographic studies from organ culture. J Clin Endocrinol Metabol. 1967; 27:945-.
  68. Olin P, Vecchio G, Ekholm R, Almqvist S. Human fetal thyroglobulin: characterization and in vitro biosynthesis studies. Endocrinology. 1970; 86:1041-1048.
  69. Szinnai G, Lacroix L, Carre A, Guimiot F, Talbot M, Martinovic J, Delezoide AL, Vekemans M, Michiels S, Caillou B, Schlumberger M, Bidart JM, Polak M. Sodium/iodide symporter (NIS) gene expression is the limiting step for the onset of thyroid function in the human fetus. J Clin Endocrinol Metab. 2007; 92:70-76.
  70. Greenberg AH, Czernichow P, Reba RC, Tyson J, Blizzard RM. Observations on the maturation of thyroid function in early fetal life. J Clin Invest. 1970; 49:1790-1803.
  71. Thorpe-Beeston JG, Nicolaides KH, Felton CV, Butler J, McGregor AM. Maturation of the secretion of thyroid hormone and thyroid stimulating hormone in the fetus. N Eng J Med. 1991; 324:532-536.
  72. Thorpe-Beeston JG, Nicolaides KH. Fetal thyroid function. Fetal Diagn Ther. 1993; 8:60-72.
  73. Morreale de Escobar G, Ares S. The hypothyroxinemia of prematurity. J Clin Endocrinol Metabol. 1998; 83:713-715.
  74. Contempré B, Jauniaux E, Calvo R, Jurkovic D, Campbell S, Morreale de Escobar G. Detection of thyroid hormones in human embryonic cavities during the first trimester of pregnancy. J Clin Endocrinol Metabol. 1993; 77:1719-1722.
  75. Calvo RM, Jauniaux E, Gulbis B, Asuncion M, Gervy C, Contempre B, Morreale de Escobar G. Fetal tissues are exposed to biologically relevant free thyroxine concentrations during early phases of development. J Clin Endocrinol Metab. 2002; 87:1768-1777.
  76. Kester MH, Martinez de Mena R, Obregon MJ, Marinkovic D, Howatson A, Visser TJ, Hume R, Morreale de Escobar G. Iodothyronine levels in the human developing brain: major regulatory roles of iodothyronine deiodinases in different areas. J Clin Endocrinol Metab. 2004; 89:3117-3128.
  77. Bernal J, Pekonen F. Ontogenesis of the nuclear 3,5,3'-triiodothyronine receptor in the human fetal brain. Endocrinology. 1984; 114:677-679.
  78. Morreale de Escobar G, Obregón MJ, Escobar del Rey F. Is neuropsychological development related to maternal hypothyroidism, or to maternal hypothyroxinemia? J Clin Endocrinol Metabol. 2000; 85:3975-3987.
  79. de Escobar GM, Obregon MJ, del Rey FE. Maternal thyroid hormones early in pregnancy and fetal brain development. Best Pract Res Clin Endocrinol Metab. 2004; 18:225-248.
  80. Berbel P, Mestre JL, Santamaria A, Palazon I, Franco A, Graells M, Gonzalez-Torga A, de Escobar GM. Delayed neurobehavioral development in children born to pregnant women with mild hypothyroxinemia during the first month of gestation: the importance of early iodine supplementation. Thyroid. 2009; 19:511-519.
  81. Henrichs J, Bongers-Schokking JJ, Schenk JJ, Ghassabian A, Schmidt HG, Visser TJ, Hooijkaas H, de Muinck Keizer-Schrama SM, Hofman A, Jaddoe VV, Visser W, Steegers EA, Verhulst FC, de Rijke YB, Tiemeier H. Maternal thyroid function during early pregnancy and cognitive functioning in early childhood: the generation R study. J Clin Endocrinol Metab. 2010; 95:4227-4234.
  82. Ghassabian A, El Marroun H, Peeters RP, Jaddoe VW, Hofman A, Verhulst FC, Tiemeier H, White T. Downstream effects of maternal hypothyroxinemia in early pregnancy: nonverbal IQ and brain morphology in school-age children. J Clin Endocrinol Metab. 2014; 99:2383-2390.
  83. Momotani N, Iwama S, Momotani K. Neurodevelopment in children born to hypothyroid mothers restored to normal thyroxine (T(4)) concentration by late pregnancy in Japan: no apparent influence of maternal T(4) deficiency. J Clin Endocrinol Metab. 2011; 97:1104-1108.
  84. Glinoer D. The regulation of thyroid function in pregnancy: pathways of endocrine adaptation from physiology to pathology. Endocr Rev. 1997; 18:401-433.
  85. Henrichs J, Ghassabian A, Peeters RP, Tiemeier H. Maternal hypothyroxinemia and effects on cognitive functioning in childhood: how and why? Clin Endocrinol (Oxf). 2013; 79:152-162.
  86. Ramezani Tehrani F, Nazarpour S, Behboudi-Gandevani S. Isolated maternal hypothyroxinemia and adverse pregnancy outcomes: A systematic review. J Gynecol Obstet Hum Reprod. 2021; 50:102057.
  87. Thompson W, Russell G, Baragwanath G, Matthews J, Vaidya B, Thompson-Coon J. Maternal thyroid hormone insufficiency during pregnancy and risk of neurodevelopmental disorders in offspring: A systematic review and meta-analysis. Clin Endocrinol (Oxf). 2018; 88:575-584.
  88. Dosiou C, Medici M. MANAGEMENT OF ENDOCRINE DISEASE: Isolated maternal hypothyroxinemia during pregnancy: knowns and unknowns. Eur J Endocrinol. 2017; 176:R21-R38.
  89. Casey BM, Thom EA, Peaceman AM, Varner MW, Sorokin Y, Hirtz DG, Reddy UM, Wapner RJ, Thorp JM, Jr., Saade G, Tita AT, Rouse DJ, Sibai B, Iams JD, Mercer BM, Tolosa J, Caritis SN, VanDorsten JP, Eunice Kennedy Shriver National Institute of Child H, Human Development Maternal-Fetal Medicine Units N. Treatment of Subclinical Hypothyroidism or Hypothyroxinemia in Pregnancy. N Engl J Med. 2017; 376:815-825.
  90. Lazarus JH, Bestwick JP, Channon S, Paradice R, Maina A, Rees R, Chiusano E, John R, Guaraldo V, George LM, Perona M, Dall'Amico D, Parkes AB, Joomun M, Wald NJ. Antenatal thyroid screening and childhood cognitive function. N Engl J Med. 2012; 366:493-501.
  91. Cooper DS, Pearce EN. Subclinical Hypothyroidism and Hypothyroxinemia in Pregnancy - Still No Answers. N Engl J Med. 2017; 376:876-877.
  92. Visser WE, Friesema EC, Visser TJ. Minireview: thyroid hormone transporters: the knowns and the unknowns. Mol Endocrinol. 2011; 25:1-14.
  93. Roberts LM, Woodford K, Zhou M, Black DS, Haggerty JE, Tate EH, Grindstaff KK, Mengesha W, Raman C, Zerangue N. Expression of the thyroid hormone transporters monocarboxylate transporter-8 (SLC16A2) and organic ion transporter-14 (SLCO1C1) at the blood-brain barrier. Endocrinology. 2008; 149:6251-6261.
  94. Ceballos A, Belinchon MM, Sanchez-Mendoza E, Grijota-Martinez C, Dumitrescu AM, Refetoff S, Morte B, Bernal J. Importance of Monocarboxylate transporter 8 (Mct8) for the blood-brain barrier dependent availability of 3,5,3’-Triiodo-L-Thyronine (T3). Endocrinology. 2009; In Press
  95. Bernal J, Guadano-Ferraz A, Morte B. Thyroid hormone transporters-functions and clinical implications. Nat Rev Endocrinol. 2015; 11:406-417.
  96. Lopez-Espindola D, Garcia-Aldea A, Gomez de la Riva I, Rodriguez-Garcia AM, Salvatore D, Visser TJ, Bernal J, Guadano-Ferraz A. Thyroid hormone availability in the human fetal brain: novel entry pathways and role of radial glia. Brain Struct Funct. 2019; 224:2103-2119.
  97. Vatine GD, Al-Ahmad A, Barriga BK, Svendsen S, Salim A, Garcia L, Garcia VJ, Ho R, Yucer N, Qian T, Lim RG, Wu J, Thompson LM, Spivia WR, Chen Z, Van Eyk J, Palecek SP, Refetoff S, Shusta EV, Svendsen CN. Modeling Psychomotor Retardation using iPSCs from MCT8-Deficient Patients Indicates a Prominent Role for the Blood-Brain Barrier. Cell Stem Cell. 2017; 20:831-843 e835.
  98. Abbott NJ, Ronnback L, Hansson E. Astrocyte-endothelial interactions at the blood-brain barrier. Nat Rev Neurosci. 2006; 7:41-53.
  99. Abbott NJ, Patabendige AA, Dolman DE, Yusof SR, Begley DJ. Structure and function of the blood-brain barrier. Neurobiol Dis. 2010; 37:13-25.
  100. Pardridge WM. Brain metabolism: a perspective from the blood-brain barrier. Physiol Rev. 1983; 63:1481-1535.
  101. Ben-Zvi A, Lacoste B, Kur E, Andreone BJ, Mayshar Y, Yan H, Gu C. Mfsd2a is critical for the formation and function of the blood-brain barrier. Nature. 2014; 509:507-511.
  102. Haddad-Tovolli R, Dragano NRV, Ramalho AFS, Velloso LA. Development and Function of the Blood-Brain Barrier in the Context of Metabolic Control. Front Neurosci. 2017; 11:224.
  103. Rinholm JE, Bergersen LH. Neuroscience: The wrap that feeds neurons. Nature. 2012; 487:435-436.
  104. Zhao Z, Nelson AR, Betsholtz C, Zlokovic BV. Establishment and Dysfunction of the Blood-Brain Barrier. Cell. 2015; 163:1064-1078.
  105. Ito K, Uchida Y, Ohtsuki S, Aizawa S, Kawakami H, Katsukura Y, Kamiie J, Terasaki T. Quantitative membrane protein expression at the blood-brain barrier of adult and younger cynomolgus monkeys. J Pharm Sci. 2011; 100:3939-3950.
  106. Wirth EK, Roth S, Blechschmidt C, Holter SM, Becker L, Racz I, Zimmer A, Klopstock T, Gailus-Durner V, Fuchs H, Wurst W, Naumann T, Brauer A, de Angelis MH, Kohrle J, Gruters A, Schweizer U. Neuronal 3',3,5-triiodothyronine (T3) uptake and behavioral phenotype of mice deficient in Mct8, the neuronal T3 transporter mutated in Allan-Herndon-Dudley syndrome. J Neurosci. 2009; 29:9439-9449.
  107. Morte B, Ceballos A, Diez D, Grijota-Martinez C, Dumitrescu AM, Di Cosmo C, Galton VA, Refetoff S, Bernal J. Thyroid hormone-regulated mouse cerebral cortex genes are differentially dependent on the source of the hormone: a study in monocarboxylate transporter-8- and deiodinase-2-deficient mice. Endocrinology. 2010; 151:2381-2387.
  108. Dumitrescu AM, Liao XH, Weiss RE, Millen K, Refetoff S. Tissue-specific thyroid hormone deprivation and excess in monocarboxylate transporter (mct) 8-deficient mice. Endocrinology. 2006; 147:4036-4043.
  109. Trajkovic M, Visser TJ, Mittag J, Horn S, Lukas J, Darras VM, Raivich G, Bauer K, Heuer H. Abnormal thyroid hormone metabolism in mice lacking the monocarboxylate transporter 8. J Clin Invest. 2007; 117:627-635.
  110. Liao XH, Di Cosmo C, Dumitrescu AM, Hernandez A, Van Sande J, St Germain DL, Weiss RE, Galton VA, Refetoff S. Distinct roles of deiodinases on the phenotype of Mct8 defect: a comparison of eight different mouse genotypes. Endocrinology. 2011; 152:1180-1191.
  111. Mayerl S, Muller J, Bauer R, Richert S, Kassmann CM, Darras VM, Buder K, Boelen A, Visser TJ, Heuer H. Transporters MCT8 and OATP1C1 maintain murine brain thyroid hormone homeostasis. J Clin Invest. 2014; 124:1987-1999.
  112. Gereben B, Zavacki AM, Ribich S, Kim BW, Huang SA, Simonides WS, Zeold A, Bianco AC. Cellular and molecular basis of deiodinase-regulated thyroid hormone signaling. Endocr Rev. 2008; 29:898-938.
  113. Barez-Lopez S, Bosch-Garcia D, Gomez-Andres D, Pulido-Valdeolivas I, Montero-Pedrazuela A, Obregon MJ, Guadano-Ferraz A. Abnormal motor phenotype at adult stages in mice lacking type 2 deiodinase. PLoS One. 2014; 9:e103857.
  114. Guadaño-Ferraz A, Obregón MJ, St-Germain D, Bernal J. The type 2 iodothyronine deiodinase is expressed primarily in glial cells in the neonatal rat brain. Proc Natl Acad Sci USA. 1997; 94:10391-10396.
  115. Guadaño-Ferraz A, Escámez MJ, Rausell E, Bernal J. Expression of type 2 iodothyronine deiodinase in hypothyroid rat brain indicates an important role of thyroid hormone in the development of specific primary sensory systems. J Neurosci. 1999; 19:3430-3439.
  116. Tu HM, Kim SW, Salvatore D, Bartha T, Legradi G, Larsen PR, Lechan RM. Regional distribution of type 2 thyroxine deiodinase messenger ribonucleic acid in rat hypothalamus and pituitary and its regulation by thyroid hormone. Endocrinology. 1997; 138:3359-3368.
  117. Tu HM, Legradi G, Bartha T, Salvatore D, Lechan RM, Larsen PR. Regional expression of the type 3 iodothyronine deiodinase messenger ribonucleic acid in the rat central nervous system and its regulation by thyroid hormone. Endocrinology. 1999; 140:784-790.
  118. Diez D, Morte B, Bernal J. Single-cell transcriptome profiling of thyroid hormone effectors in the human fetal neocortex: expression of SLCO1C1, DIO2, and THRB in specific cell types. Thyroid. 2021;
  119. Escámez MJ, Guadaño-Ferraz A, Cuadrado A, Bernal J. Type 3 iodothyronine deiodinase is selectively expressed in areas related to sexual differentiation in the newborn rat brain. Endocrinology. 1999; 140:5443-5446.
  120. Freitas BC, Gereben B, Castillo M, Kallo I, Zeold A, Egri P, Liposits Z, Zavacki AM, Maciel RM, Jo S, Singru P, Sanchez E, Lechan RM, Bianco AC. Paracrine signaling by glial cell-derived triiodothyronine activates neuronal gene expression in the rodent brain and human cells. J Clin Invest. 2010; 120:2206-2217.
  121. Riskind PN, Kolodny JM, Larsen PR. The regional distribution of type II 5'-monodeiodinase in euthyroid and hypothyroid rats. Brain Res. 1987; 420:194-198.
  122. Langlet F, Levin BE, Luquet S, Mazzone M, Messina A, Dunn-Meynell AA, Balland E, Lacombe A, Mazur D, Carmeliet P, Bouret SG, Prevot V, Dehouck B. Tanycytic VEGF-A boosts blood-hypothalamus barrier plasticity and access of metabolic signals to the arcuate nucleus in response to fasting. Cell Metab. 2013; 17:607-617.
  123. Bolborea M, Dale N. Hypothalamic tanycytes: potential roles in the control of feeding and energy balance. Trends Neurosci. 2013; 36:91-100.
  124. Larsen PR, Silva JR, Kaplan MM. Relationships between circulating and intracellular thyroid hormones. Physiological and clinical implications. Endocr Rev. 1981; 2:87-102.
  125. van Doorn J, Roelfsema F, van der Heide D. Contribution from local conversion of thyroxine to 3,5,3'-triiodothyronine to intracellular 3,5,3'-triiodothyronine in several organs in hypothyroid rats at isotope equilibrium. Acta Endocrinol (Copenh). 1982; 101:386-396.
  126. Galton VA, Wood ET, St Germain EA, Withrow CA, Aldrich G, St Germain GM, Clark AS, St Germain DL. Thyroid hormone homeostasis and action in the type 2 deiodinase-deficient rodent brain during development. Endocrinology. 2007; 148:3080-3088.
  127. Morte B, Bernal J. Thyroid hormone action: astrocyte-neuron communication. Front Endocrinol (Lausanne). 2014; 5:82.
  128. Crantz FR, Silva JE, Larsen PR. Ananalysis of the sources and quantity of 3,5,3'-triiodothyronine specifically bound to nuclear receptors in rat cerebral cortex and cerebellum. Endocrinology. 1982; 110:367-375.
  129. Ng L, Hernandez A, He W, Ren T, Srinivas M, Ma M, Galton VA, St Germain DL, Forrest D. A protective role for type 3 deiodinase, a thyroid hormone-inactivating enzyme, in cochlear development and auditory function. Endocrinology. 2009; 150:1952-1960.
  130. Campos-Barros A, Amma LL, Faris JS, Shailam R, Kelley MW, Forrest D. Type 2 iodothyronine deiodinase expression in the cochlea before the onset of hearing. Proc Natl Acad Sci USA. 2000; 97:1287-1292.
  131. Eldred KC, Hadyniak SE, Hussey KA, Brenerman B, Zhang PW, Chamling X, Sluch VM, Welsbie DS, Hattar S, Taylor J, Wahlin K, Zack DJ, Johnston RJ, Jr. Thyroid hormone signaling specifies cone subtypes in human retinal organoids. Science. 2018; 362
  132. Galton VA, Martínez E, Hernández A, St. Germain EA, Bates JM, St.Germain DL. Pregnant rat uterus expresses high levels of the type 3 iodothyronine deiodinase. J Clin Invest. 1999; 103:979-987.
  133. Bates JM, St.Germain DL, Galton VA. Expression profiles of the three iodothyronine deiodinases D1, D2 and D3, in the developing rat. Endocrinology. 1999; 140:844-851.
  134. Ruiz de Oña C, Obregón MJ, Escobar del Rey F, Morreale de Escobar G. Developmental changes in rat brain 5'-deiodinase and thyroid hormones during the fetal period: the effects of fetal hypothyroidism and maternal thyroid hormones. Pediatr Res. 1988; 24:588-594.
  135. Zuchero JB, Barres BA. Glia in mammalian development and disease. Development. 2015; 142:3805-3809.
  136. Calvo R, Obregón MJ, Ruiz de Oña C, Escobar del Rey F, Morreale de Escobar G. Congenital hypothyroidism, as studied in rats. Crucial role of maternal thyroxine but not of 3,5,3'-triiodothyronine in the protection of the fetal brain. J Clin Invest. 1990; 86:889-899.
  137. Barez-Lopez S, Obregon MJ, Bernal J, Guadano-Ferraz A. Thyroid Hormone Economy in the Perinatal Mouse Brain: Implications for Cerebral Cortex Development. Cereb Cortex. 2018; 28:1783-1793.
  138. Grijota-Martinez C, Diez D, Morreale de Escobar G, Bernal J, Morte B. Lack of action of exogenously administered T3 on the fetal rat brain despite expression of the monocarboxylate transporter 8. Endocrinology. 2011; 152:1713-1721.
  139. Hernandez A, Morte B, Belinchon MM, Ceballos A, Bernal J. Critical role of types 2 and 3 deiodinases in the negative regulation of gene expression by T(3)in the mouse cerebral cortex. Endocrinology. 2012; 153:2919-2928.
  140. Hernandez A, Quignodon L, Martinez ME, Flamant F, St Germain DL. Type 3 deiodinase deficiency causes spatial and temporal alterations in brain T3 signaling that are dissociated from serum thyroid hormone levels. Endocrinology. 2010; 151:5550-5558.
  141. Martinez ME, Hernandez A. The Type 3 Deiodinase Is a Critical Modulator of Thyroid Hormone Sensitivity in the Fetal Brain. Front Neurosci. 2021; 15:703730.
  142. Chan S, Kachilele S, McCabe CJ, Tannahill LA, Boelaert K, Gittoes NJ, Visser TJ, Franklyn JA, Kilby MD. Early expression of thyroid hormone deiodinases and receptors in human fetal cerebral cortex. Brain Res Dev Brain Res. 2002; 138:109-116.
  143. Zhong S, Zhang S, Fan X, Wu Q, Yan L, Dong J, Zhang H, Li L, Sun L, Pan N, Xu X, Tang F, Zhang J, Qiao J, Wang X. A single-cell RNA-seq survey of the developmental landscape of the human prefrontal cortex. Nature. 2018; 555:524-528.
  144. Johnson MB, Wang PP, Atabay KD, Murphy EA, Doan RN, Hecht JL, Walsh CA. Single-cell analysis reveals transcriptional heterogeneity of neural progenitors in human cortex. Nat Neurosci. 2015; 18:637-646.
  145. Wallis K, Dudazy S, van Hogerlinden M, Nordstrom K, Mittag J, Vennstrom B. The thyroid hormone receptor alpha1 protein is expressed in embryonic postmitotic neurons and persists in most adult neurons. Mol Endocrinol. 2010; 24:1904-1916.
  146. Saunders NR, Dziegielewska KM, Mollgard K, Habgood MD. Physiology and molecular biology of barrier mechanisms in the fetal and neonatal brain. J Physiol. 2018; 596:5723-5756.
  147. Polioudakis D, de la Torre-Ubieta L, Langerman J, Elkins AG, Shi X, Stein JL, Vuong CK, Nichterwitz S, Gevorgian M, Opland CK, Lu D, Connell W, Ruzzo EK, Lowe JK, Hadzic T, Hinz FI, Sabri S, Lowry WE, Gerstein MB, Plath K, Geschwind DH. A Single-Cell Transcriptomic Atlas of Human Neocortical Development during Mid-gestation. Neuron. 2019; 103:785-801 e788.
  148. Flamant F, Gauthier K, Richard S. Genetic Investigation of Thyroid Hormone Receptor Function in the Developing and Adult Brain. Curr Top Dev Biol. 2017; 125:303-335.
  149. Flamant F, Gauthier K. Thyroid hormone receptors: the challenge of elucidating isotype-specific functions and cell-specific response. Biochim Biophys Acta. 2013; 1830:3900-3907.
  150. Ercan-Fang S, Schwartz HL, Oppenheimer JH. Isoform specific 3,5,3'-triiodothyronine receptor binding capacity and messenger ribonucleic acid content in rat adenohypophysis: Effect of thyroidal state and comparison with extrapituitary tissues. Endocrinology. 1996; 137:3228-3233.
  151. Minakhina S, Bansal S, Zhang A, Brotherton M, Janodia R, De Oliveira V, Tadepalli S, Wondisford FE. A Direct Comparison of Thyroid Hormone Receptor Protein Levels in Mice Provides Unexpected Insights into Thyroid Hormone Action. Thyroid. 2020; 30:1193-1204.
  152. Messier N, Langlois MF. Triac regulation of transcription is T(3) receptor isoform- and response element-specific. Mol Cell Endocrinol. 2000; 165:57-66.
  153. Bradley DJ, Towle HC, Young WS. Spatial and temporal expression of a- and b- thyroid hormone receptor mRNAs, including the b2-subtype, in the developing mammalian nervous system. J Neurosci. 1992; 12:2288-2302.
  154. Bradley DJ, Young III WS, Weinberger C. Differential expression of alpha and beta thyroid hormone receptor genes in rat brain and pituitary. Proc Natl Acad Sci USA. 1989; 86:72507254.
  155. Mellström B, Naranjo JR, Santos A, González AM, Bernal J. Independent expression of the a and b c-erbA genes in developing rat brain. Mol Endocrinol. 1991; 5:1339-1350.
  156. Luo M, Faure R, Dussault JH. Ontogenesis of nuclear T3 receptors in primary cultured astrocytes and neurons. Brain Res. 1986; 381:275-280.
  157. Yusta B, Besnard F, Ortiz-Caro J, Pascual A, Aranda A, Sarlieve L. Evidence for the presence of nuclear 3,5,3'-triiodothyronine receptors in secondary cultures of pure rat oligodendrocytes. Endocrinology. 1988; 122:2278-2284.
  158. Glauser L, Barakat-Walter I. Differential distribution of thyroid hormone receptor isoform in rat dorsal root ganglia and sciatic nerve in vivo and in vitro. J Neuroendocrinol. 1997; 9:217-227.
  159. Barakat-Walter I, Droz B. Nuclear and cytoplasmic triiodothyronine binding sites in primary sensory neurons and Schwann cells: radioautographic study during development. J Neuroendocrinol. 1995; 7:127-136.
  160. Lima FR, Gervais A, Colin C, Izembart M, Neto VM, Mallat M. Regulation of microglial development: a novel role for thyroid hormone. J Neurosci. 2001; 21:2028-2038.
  161. Barca-Mayo O, Liao XH, Alonso M, Di Cosmo C, Hernandez A, Refetoff S, Weiss RE. Thyroid hormone receptor alpha and regulation of type 3 deiodinase. Mol Endocrinol. 2011; 25:575-583.
  162. Chatonnet F, Guyot R, Benoit G, Flamant F. Genome-wide analysis of thyroid hormone receptors shared and specific functions in neural cells. Proc Natl Acad Sci U S A. 2013; 110:E766-775.
  163. Gil-Ibanez P, Bernal J, Morte B. Thyroid hormone regulation of gene expression in primary cerebrocortical cells: role of thyroid hormone receptor subtypes and interactions with retinoic acid and glucocorticoids. PLoS One. 2014; 9:e91692.
  164. Gil-Ibanez P, Morte B, Bernal J. Role of thyroid hormone receptor subtypes alpha and beta on gene expression in the cerebral cortex and striatum of postnatal mice. Endocrinology. 2013; 154:1940-1947.
  165. Ng L, Hurley JB, Dierks B, Srinivas M, Salto C, Vennstrom B, Reh TA, Forrest D. A thyroid hormone receptor that is required for the development of green cone photoreceptors. Nat Genet. 2001; 27:94-98.
  166. Jones I, Srinivas M, Ng L, Forrest D. The thyroid hormone receptor beta gene: structure and functions in the brain and sensory systems. Thyroid. 2003; 13:1057-1068.
  167. Cook CB, Kakucska I, Lechan RM, Koenig RJ. Expression of thyroid hormone receptor beta 2 in rat hypothalamus. Endocrinology. 1992; 130:1077-1079.
  168. Pérez-Castillo A, Bernal J, Ferreiro B, Pans T. The early ontogenesis of thyroid hormone receptor in the rat fetus. Endocrinology. 1985; 117:2457-2461.
  169. Schwartz HL, Oppenheimer JH. Ontogenesis of 3,5,3'-triiodothyronine receptors in neonatal rat brain, dissociation between receptor concentration and stimulation of oxygen consumption by 3,5,3'-triiodothyronine. Endocrinology. 1978; 103:943-948.
  170. Strait KA, Schwartz HL, Perezcastillo A, Oppenheimer JH. Relationship of C-erbA Messenger RNA Content to Tissue Triiodothyronine Nuclear Binding Capacity and Function in Developing and Adult Rats. J BiolChem. 1990; 265:10514-10521.
  171. Ferreiro B, Pastor R, Bernal J. T3 receptor occupancy and T3 levels in plasma and cytosol during rat brain development. Acta Endocrinol (Copenh). 1990; 123:95-99.
  172. Iskaros J, Pickard M, Evans I, Sinha A, Hardiman P, Ekins R. Thyroid hormone receptor gene expression in first trimester human fetal brain. J Clin Endocrinol Metabol. 2000; 85:2620-2623.
  173. Ferreiro B, Bernal J, Goodyer CG, Branchard CL. Estimation of nuclear thyroid hormone receptor saturation in human fetal brain and lung during early gestation. J Clin Endocrinol Metabol. 1988; 67:853-856.
  174. Ferreiro B, Bernal J, Morreale de Escobar G, Potter BJ. Preferential saturation of brain 3,5,3'-triiodothyronine receptor during development in fetal lambs. Endocrinology. 1988; 122:438-443.
  175. Iñiguez MA, De Lecea L, Guadaño-Ferraz A, Morte B, Gerendasy D, Sutcliffe JG, Bernal J. Cell-specific effects of thyroid hormone on RC3/neurogranin expression in rat brain. Endocrinology. 1996; 137:1032-1041.
  176. Chatonnet F, Guyot R, Picou F, Bondesson M, Flamant F. Genome-wide search reveals the existence of a limited number of thyroid hormone receptor alpha target genes in cerebellar neurons. PLoS One. 2012; 7:e30703.
  177. Diez D, Grijota-Martinez C, Agretti P, De Marco G, Tonacchera M, Pinchera A, de Escobar GM, Bernal J, Morte B. Thyroid hormone action in the adult brain: gene expression profiling of the effects of single and multiple doses of triiodo-L-thyronine in the rat striatum. Endocrinology. 2008; 149:3989-4000.
  178. Dong H, Wade M, Williams A, Lee A, Douglas GR, Yauk C. Molecular insight into the effects of hypothyroidism on the developing cerebellum. Biochem Biophys Res Commun. 2005; 330:1182-1193.
  179. Martel J, Cayrou C, Puymirat J. Identification of new thyroid hormone-regulated genes in rat brain neuronal cultures. Neuroreport. 2002; 13:1849-1851.
  180. Miller LD, McPhie P, Suzuki H, Kato Y, Liu ET, Cheng SY. Multi-tissue gene-expression analysis in a mouse model of thyroid hormone resistance. Genome Biol. 2004; 5:R31.
  181. Poguet AL, Legrand C, Feng X, Yen PM, Meltzer P, Samarut J, Flamant F. Microarray analysis of knockout mice identifies cyclin D2 as a possible mediator for the action of thyroid hormone during the postnatal development of the cerebellum. Dev Biol. 2003; 254:188-199.
  182. Quignodon L, Grijota-Martinez C, Compe E, Guyot R, Allioli N, Laperriere D, Walker R, Meltzer P, Mader S, Samarut J, Flamant F. A combined approach identifies a limited number of new thyroid hormone target genes in post-natal mouse cerebellum. J Mol Endocrinol. 2007; 39:17-28.
  183. Sato Y, Buchholz DR, Paul BD, Shi YB. A role of unliganded thyroid hormone receptor in postembryonic development in Xenopus laevis. Mech Dev. 2007; 124:476-488.
  184. Munoz A, Wrighton C, Seliger B, Bernal J, Beug H. Thyroid hormone receptor/c-erbA: control of commitment and differentiation in the neuronal/chromaffin progenitor line PC12. J Cell Biol. 1993; 121:423-438.
  185. Winter H, Braig C, Zimmermann U, Engel J, Rohbock K, Knipper M. Thyroid hormone receptor alpha1 is a critical regulator for the expression of ion channels during final differentiation of outer hair cells. Histochem Cell Biol. 2007; 128:65-75.
  186. Flamant F, Poguet AL, Plateroti M, Chassande O, Gauthier K, Streichenberger N, Mansouri A, Samarut J. Congenital hypothyroid Pax8(-/-) mutant mice can be rescued by inactivating the TRalpha gene. Mol Endocrinol. 2002; 16:24-32.
  187. Hashimoto K, Curty FH, Borges PP, Lee CE, Abel ED, Elmquist JK, Cohen RN, Wondisford FE. An unliganded thyroid hormone receptor causes severe neurological dysfunction. Proc Natl Acad Sci U S A. 2001; 98:3998-4003.
  188. Roberts MR, Srinivas M, Forrest D, Morreale de Escobar G, Reh TA. Making the gradient: thyroid hormone regulates cone opsin expression in the developing mouse retina. Proc Natl Acad Sci U S A. 2006; 103:6218-6223.
  189. Cheng SY, Leonard JL, Davis PJ. Molecular aspects of thyroid hormone actions. Endocr Rev. 2010; 31:139-170.
  190. Davis PJ, Davis FB, Cody V. Membrane receptors mediating thyroid hormone action. Trends Endocrinol Metab. 2005; 16:429-435.
  191. Davis PJ, Leonard JL, Davis FB. Mechanisms of nongenomic actions of thyroid hormone. Front Neuroendocrinol. 2008; 29:211-218.
  192. Stenzel D, Wilsch-Brauninger M, Wong FK, Heuer H, Huttner WB. Integrin alphavbeta3 and thyroid hormones promote expansion of progenitors in embryonic neocortex. Development. 2014; 141:795-806.
  193. Martin NP, Marron Fernandez de Velasco E, Mizuno F, Scappini EL, Gloss B, Erxleben C, Williams JG, Stapleton HM, Gentile S, Armstrong DL. A rapid cytoplasmic mechanism for PI3 kinase regulation by the nuclear thyroid hormone receptor, TRbeta, and genetic evidence for its role in the maturation of mouse hippocampal synapses in vivo. Endocrinology. 2014; 155:3713-3724.
  194. Yamaguchi S, Aoki N, Kitajima T, Iikubo E, Katagiri S, Matsushima T, Homma KJ. Thyroid hormone determines the start of the sensitive period of imprinting and primes later learning. Nat Commun. 2012; 3:1081.
  195. Farsetti A, Desvergne B, Hallenbeck P, Robbins J, Nikodem V. Characterization of myelin basic protein thyroid hormone response element and its function in the context of native and heterologous promoter. J Biol Chem. 1992; 267:15784-15788.
  196. Zou L, Hagen SC, Strait KA, Oppenheimer JH. Identification of thyroid hormone response elements in rodent Pcp-2, a developmentally regulated gene of cerebellar Purkinje cells. J Biol Chem. 1994; 269:13346-13352.
  197. Martinez de Arrieta C, Morte B, Coloma A, Bernal J. The human RC3 gene homolog, NRGN contains a thyroid hormone-responsive element located in the first intron. Endocrinology. 1999; 140:335-343.
  198. García-Fernández LF, Urade Y, Hayaishi O, Bernal J, Muñoz A. Identification of a thyroid hormone response element in the promoter region of the rat lipocalin-type prostaglandin D synthase (beta-trace) gene. Mol Brain Res. 1998; 55:321-330.
  199. White DM, Takeda T, DeGroot LJ, Stefansson K, Arnason BG. Beta-trace gene expression is regulated by a core promoter and a distal thyroid hormone response element. J Biol Chem. 1997; 272:14387-14393.
  200. Thompson CC, Bottcher MC. The product of a thyroid hormone-responsive gene interacts with thyroid hormone receptors. Proc Natl Acad Sci U S A. 1997; 94:8527-8532.
  201. Iglesias T, Caubin J, Stunnenberg HG, Zaballos A, Bernal J, Muñoz A. Thyroid hormone-dependent transcriptional repression of neural cell adhesion molecule during brain maturation. EMBO J. 1996; 15:4307-4316.
  202. Ghorbel MT, Seugnet I, Hadj-Sahraoui N, Topilko P, Levi G, Demeneix B. Thyroid hormone effects on Krox-24 transcription in the post-natal mouse brain are developmentally regulated but are not correlated with mitosis. Oncogene. 1999; 18:917-924.
  203. Dong H, Yauk CL, Rowan-Carroll A, You SH, Zoeller RT, Lambert I, Wade MG. Identification of thyroid hormone receptor binding sites and target genes using ChIP-on-chip in developing mouse cerebellum. PLoS One. 2009; 4:e4610.
  204. Silva JE, Rudas P. Effects of congenital hypothyroidism on microtubule-associated protein-2 expression in the cerebellum of the rat. Endocrinology. 1990; 126:1276-1282.
  205. Aniello F, Couchie D, Bridoux AM, Gripois D, Nunez J. Splicing of juvenile and adult tau mRNA variants is regulated by thyroid hormone. Proc Natl Acad Sci U S A. 1991; 88:4035-4039.
  206. Cuadrado A, Garcia-Fernandez LF, Imai T, Okano H, Munoz A. Regulation of tau RNA maturation by thyroid hormone is mediated by the neural RNA-binding protein musashi-1. Mol Cell Neurosci. 2002; 20:198-210.
  207. Dowling AL, Iannacone EA, Zoeller RT. Maternal Hypothyroidism Selectively Affects the Expression of Neuroendocrine-Specific Protein A Messenger Ribonucleic Acid in the Proliferative Zone of the Fetal Rat Brain Cortex. Endocrinology. 2001; 142:390-399.
  208. Sampson D, Pickard MR, Sinha AK, Evans IM, Leonard AJ, Ekins RP. Maternal thyroid status regulates the expression of neuronal and astrocytic cytoskeletal proteins in the fetal brain. J Endocrinol. 2000; 167:439-445.
  209. Sinha RA, Pathak A, Mohan V, Bandyopadhyay S, Rastogi L, Godbole MM. Maternal thyroid hormone: a strong repressor of neuronal nitric oxide synthase in rat embryonic neocortex. Endocrinology. 2008; 149:4396-4401.
  210. Navarro D, Alvarado M, Morte B, Berbel D, Sesma J, Pacheco P, Morreale de Escobar G, Bernal J, Berbel P. Late maternal hypothyroidism alters the expression of Camk4 in neocortical subplate neurons: a comparison with Nurr1 labeling. Cereb Cortex. 2014; 24:2694-2706.
  211. Krebs J, Honegger P. Calmodulin kinase IV: expression and function during rat brain development. Biochim Biophys Acta. 1996; 1313:217-222.
  212. Liu YY, Brent GA. Thyroid hormone-dependent gene expression in differentiated embryonic stem cells and embryonal carcinoma cells: identification of novel thyroid hormone target genes by deoxyribonucleic acid microarray analysis. Endocrinology. 2005; 146:776-783.
  213. Schwartz HL, Ross ME, Oppenheimer JH. Lack of effect of thyroid hormone on late fetal rat brain development. Endocrinology. 1997; 138:3119-3124.
  214. Bagamasbad PD, Espina JEC, Knoedler JR, Subramani A, Harden AJ, Denver RJ. Coordinated transcriptional regulation by thyroid hormone and glucocorticoid interaction in adult mouse hippocampus-derived neuronal cells. PLoS One. 2019; 14:e0220378.
  215. Bauer M, Heinz A, Whybrow PC. Thyroid hormones, serotonin and mood: of synergy and significance in the adult brain. Mol Psychiatry. 2002; 7:140-156.
  216. Simon NM, Blacker D, Korbly NB, Sharma SG, Worthington JJ, Otto MW, Pollack MH. Hypothyroidism and hyperthyroidism in anxiety disorders revisited: new data and literature review. J Affect Disord. 2002; 69:209-217.
  217. Bauer M, London ED, Rasgon N, Berman SM, Frye MA, Altshuler LL, Mandelkern MA, Bramen J, Voytek B, Woods R, Mazziotta JC, Whybrow PC. Supraphysiological doses of levothyroxine alter regional cerebral metabolism and improve mood in bipolar depression. Mol Psychiatry. 2005; 10:456-469.
  218. Baumgartner A, Bauer M, Hellweg R. Treatment of intractable non-rapid cycling bipolar affective disorder with high-dose thyroxine: an open clinical trial. Neuropsychopharmacology. 1994; 10:183-189.
  219. Niculescu AB, 3rd, Segal DS, Kuczenski R, Barrett T, Hauger RL, Kelsoe JR. Identifying a series of candidate genes for mania and psychosis: a convergent functional genomics approach. Physiol Genomics. 2000; 4:83-91.
  220. Kriegstein A, Alvarez-Buylla A. The glial nature of embryonic and adult neural stem cells. Annu Rev Neurosci. 2009; 32:149-184.
  221. Hatten ME. Riding the glial monorail: a common mechanism for glial-guided neuronal migration in different regions of the developing mammalian brain. Trends Neurosci. 1990; 13:179-184.
  222. Rakic P. Mode of cell migration to the superficial layers of fetal monkey neocortex. J Comp Neurol. 1972; 145:61-83.
  223. Benito-Kwiecinski S, Giandomenico SL, Sutcliffe M, Riis ES, Freire-Pritchett P, Kelava I, Wunderlich S, Martin U, Wray GA, McDole K, Lancaster MA. An early cell shape transition drives evolutionary expansion of the human forebrain. Cell. 2021; 184:2084-2102 e2019.
  224. Denver RJ, Hu F, Scanlan TS, Furlow JD. Thyroid hormone receptor subtype specificity for hormone-dependent neurogenesis in Xenopus laevis. Dev Biol. 2009; 326:155-168.
  225. Chen C, Zhou Z, Zhong M, Zhang Y, Li M, Zhang L, Qu M, Yang J, Wang Y, Yu Z. Thyroid hormone promotes neuronal differentiation of embryonic neural stem cells by inhibiting STAT3 signaling through TRalpha1. Stem Cells Dev. 2012; 21:2667-2681.
  226. Mohan V, Sinha RA, Pathak A, Rastogi L, Kumar P, Pal A, Godbole MM. Maternal thyroid hormone deficiency affects the fetal neocorticogenesis by reducing the proliferating pool, rate of neurogenesis and indirect neurogenesis. Exp Neurol. 2012; 237:477-488.
  227. Chantoux F, Francon J. Thyroid hormone regulates the expression of NeuroD/BHF1 during the development of rat cerebellum. Mol Cell Endocrinol. 2002; 194:157-163.
  228. Krieger TG, Moran CM, Frangini A, Visser WE, Schoenmakers E, Muntoni F, Clark CA, Gadian D, Chong WK, Kuczynski A, Dattani M, Lyons G, Efthymiadou A, Varga-Khadem F, Simons BD, Chatterjee K, Livesey FJ. Mutations in thyroid hormone receptor alpha1 cause premature neurogenesis and progenitor cell depletion in human cortical development. Proc Natl Acad Sci U S A. 2019; 116:22754-22763.
  229. Martinez-Galan JR, Pedraza P, Santacana M, Escobar del Ray F, Morreale de Escobar G, Ruiz-Marcos A. Early effects of iodine deficiency on radial glial cells of the hippocampus of the rat fetus. A model of neurological cretinism. J Clin Invest. 1997; 99:2701-2709.
  230. Ernst A, Alkass K, Bernard S, Salehpour M, Perl S, Tisdale J, Possnert G, Druid H, Frisen J. Neurogenesis in the striatum of the adult human brain. Cell. 2014; 156:1072-1083.
  231. Spalding KL, Bergmann O, Alkass K, Bernard S, Salehpour M, Huttner HB, Bostrom E, Westerlund I, Vial C, Buchholz BA, Possnert G, Mash DC, Druid H, Frisen J. Dynamics of hippocampal neurogenesis in adult humans. Cell. 2013; 153:1219-1227.
  232. Kapoor R, Fanibunda SE, Desouza LA, Guha SK, Vaidya VA. Perspectives on thyroid hormone action in adult neurogenesis. J Neurochem. 2015; 133:599-616.
  233. Remaud S, Gothie JD, Morvan-Dubois G, Demeneix BA. Thyroid hormone signaling and adult neurogenesis in mammals. Front Endocrinol (Lausanne). 2014; 5:62.
  234. Ambrogini P, Cuppini R, Ferri P, Mancini C, Ciaroni S, Voci A, Gerdoni E, Gallo G. Thyroid hormones affect neurogenesis in the dentate gyrus of adult rat. Neuroendocrinology. 2005; 81:244-253.
  235. Desouza LA, Ladiwala U, Daniel SM, Agashe S, Vaidya RA, Vaidya VA. Thyroid hormone regulates hippocampal neurogenesis in the adult rat brain. Mol Cell Neurosci. 2005; 29:414-426.
  236. Lemkine GF, Raj A, Alfama G, Turque N, Hassani Z, Alegria-Prevot O, Samarut J, Levi G, Demeneix BA. Adult neural stem cell cycling in vivo requires thyroid hormone and its alpha receptor. Faseb J. 2005; 19:863-865.
  237. Montero-Pedrazuela A, Venero C, Lavado-Autric R, Fernandez-Lamo I, Garcia-Verdugo JM, Bernal J, Guadano-Ferraz A. Modulation of adult hippocampal neurogenesis by thyroid hormones: implications in depressive-like behavior. Mol Psychiatry. 2006; 11:361-371.
  238. Fanibunda SE, Desouza LA, Kapoor R, Vaidya RA, Vaidya VA. Thyroid Hormone Regulation of Adult Neurogenesis. Vitam Horm. 2018; 106:211-251.
  239. Kapoor R, Desouza LA, Nanavaty IN, Kernie SG, Vaidya VA. Thyroid hormone accelerates the differentiation of adult hippocampal progenitors. J Neuroendocrinol. 2012; 24:1259-1271.
  240. Lopez-Juarez A, Remaud S, Hassani Z, Jolivet P, Pierre Simons J, Sontag T, Yoshikawa K, Price J, Morvan-Dubois G, Demeneix BA. Thyroid hormone signaling acts as a neurogenic switch by repressing Sox2 in the adult neural stem cell niche. Cell Stem Cell. 2012; 10:531-543.
  241. Gothie JD, Sebillot A, Luongo C, Legendre M, Nguyen Van C, Le Blay K, Perret-Jeanneret M, Remaud S, Demeneix BA. Adult neural stem cell fate is determined by thyroid hormone activation of mitochondrial metabolism. Mol Metab. 2017; 6:1551-1561.
  242. Pérez-Juste G, Aranda A. The cyclin-dependent kinase inhibitor p27(Kip1) is involved in thyroid hormone-mediated neuronal differentiation. J Biol Chem. 1999; 19:5026-5031.
  243. Qi JS, Yuan Y, Desai-Yajnik V, Samuels HH. Regulation of the mdm2 oncogene by thyroid hormone receptor. Mol Cell Biol. 1999; 19:864-872.
  244. Wood WM, Sarapura VD, Dowding JM, Woodmansee WW, Haakinson DJ, Gordon DF, Ridgway EC. Early gene expression changes preceding thyroid hormone-induced involution of a thyrotrope tumor. Endocrinology. 2002; 143:347-359.
  245. Aniello F, Couchie D, Gripois D, Nunez J. Regulation of five tubulin isotypes by thyroid hormone during brain development. J Neurochem. 1991; 57:1781-1786.
  246. Lorenzo PI, Menard C, Miller FD, Bernal J. Thyroid hormone-dependent regulation of Ta1 a-tubulin during brain development. Mol Cell Neurosci. 2002; 19:333-343.
  247. Lima FR, Goncalves N, Gomes FC, de Freitas MS, Moura Neto V. Thyroid hormone action on astroglial cells from distinct brain regions during development. Int J Dev Neurosci. 1998; 16:19-27.
  248. Manzano J, Bernal J, Morte B. Influence of thyroid hormones on maturation of rat cerebellar astrocytes. Int J Dev Neurosci. 2007; 25:171-179.
  249. Clos J, Legrand C, Legrand J. Effects of thyroid state on the formation and early morphological development of bergmann glia in the developing rat cerebellum. Dev Neurosci. 1980; 3:199-208.
  250. Alvarez-Dolado M, Cuadrado A, Navarro-Yubero C, Sonderegger P, Furley AJ, Bernal J, Muñoz A. Regulation of the L1 cell adhesion molecule by thyroid hormone in the developing brain. Mol Cell Neurobiol. 2000; 499-514
  251. Alvarez-Dolado M, Gonzalez-Sancho JM, Bernal J, Muñoz A. Developmental expression of tenascin-C is altered by hypothyroidism in the rat brain. Neurosci. 1998; 84:309-322.
  252. Farwell AP, Dubord-Tomasetti SA. Thyroid hormone regulates the expression of laminin in the developing rat cerebellum. Endocrinology. 1999; 140:4221-4227.
  253. Gharami K, Das S. Thyroid hormone-induced morphological differentiation and maturation of astrocytes are mediated through the beta-adrenergic receptor. J Neurochem. 2000; 75:1962-1969.
  254. Das M, Ghosh M, Das S. Thyroid Hormone-Induced Differentiation of Astrocytes is Associated with Transcriptional Upregulation of beta-arrestin-1 and beta-adrenergic Receptor-Mediated Endosomal Signaling. Mol Neurobiol. 2016; 53:5178-5190.
  255. Alvarez-Dolado M, Iglesias T, Rodríguez-Peña A, Bernal J, Muñoz A. Expression of neurotrophins and the trk family of neurotrophin receptors in normal and hypothyroid rat brain. Mol Brain Res. 1994; 27:249-257.
  256. Koibuchi N, Yamaoka S, Chin WW. Effect of altered thyroid status on neurotrophin gene expression during postnatal development of the mouse cerebellum. Thyroid. 2001; 11:205-210.
  257. Lindholm D, Castren E, Tsoulfas P, Kolbeck R, Penha Berzaghi M, Leingartner A, Heisenberg CP, Tesarollo L, parada LF, Thoenen H. Neurotrophin-3 induced by tri-iodothyronine in cerebellar granullar cells promotes purkinje cell differentiation. J Cell Biol. 1993; 122:443-450.
  258. Schoonover CM, Seibel MM, Jolson DM, Stack MJ, Rahman RJ, Jones SA, Mariash CN, Anderson GW. Thyroid hormone regulates oligodendrocyte accumulation in developing rat brain white matter tracts. Endocrinology. 2004; 145:5013-5020.
  259. Lee JY, Petratos S. Thyroid Hormone Signaling in Oligodendrocytes: from Extracellular Transport to Intracellular Signal. Mol Neurobiol. 2016; 53:6568-6583.
  260. Berbel P, Guadaño-Ferraz A, Angulo A, Cerezo JR. Role of thyroid hormones in the maturation of interhemispheric connections in rat. Behav Brain Res. 1994; 64:9-14.
  261. Gravel C, Sasseville R, Hawkes R. Maturation of the corpus callosum of the rat: II. Influence of thyroid hormones on the number and maturation of axons. J Comp Neurol. 1990; 291:147-161.
  262. Notterpek LM, Rome LH. Functional evidence for the role of axolemma in CNS myelination. Neuron. 1994; 13:473-485.
  263. Ibarrola N, Rodriguez-Peña A. Hypothyroidism coordinately and transiently affects myelin proein gene expression in most rat brain regions during postnatal development. Brain Res. 1997; 752:285-293.
  264. Johe KK, Hazel TG, Muller T, Dugich-Djordjevic MM, McKay RD. Single factors direct the differentiation of stem cells from the fetal and adult central nervous system. Genes Dev. 1996; 10:3129-3140.
  265. Gao FB, Apperly J, Raff M. Cell-intrinsic timers and thyroid hormone regulate the probability of cell-cycle withdrawal and differentiation of oligodendrocyte precursor cells. Dev Biol. 1998; 197:54-66.
  266. Nygard M, Wahlstrom GM, Gustafsson MV, Tokumoto YM, Bondesson M. Hormone-Dependent Repression of the E2F-1 Gene by Thyroid Hormone Receptors. Mol Endocrinol. 2003; 17:79-92.
  267. Dugas JC, Ibrahim A, Barres BA. The T3-induced gene KLF9 regulates oligodendrocyte differentiation and myelin regeneration. Mol Cell Neurosci. 2012; 50:45-57.
  268. Castelo-Branco G, Lilja T, Wallenborg K, Falcao AM, Marques SC, Gracias A, Solum D, Paap R, Walfridsson J, Teixeira AI, Rosenfeld MG, Jepsen K, Hermanson O. Neural stem cell differentiation is dictated by distinct actions of nuclear receptor corepressors and histone deacetylases. Stem Cell Reports. 2014; 3:502-515.
  269. Billon N, Jolicoeur C, Tokumoto Y, Vennstrom B, Raff M. Normal timing of oligodendrocyte development depends on thyroid hormone receptor alpha 1 (TRalpha1). Embo J. 2002; 21:6452-6460.
  270. Billon N, Tokumoto Y, Forrest D, Raff M. Role of thyroid hormone receptors in timing oligodendrocyte differentiation. Dev Biol. 2001; 235:110-120.
  271. Baxi EG, Schott JT, Fairchild AN, Kirby LA, Karani R, Uapinyoying P, Pardo-Villamizar C, Rothstein JR, Bergles DE, Calabresi PA. A selective thyroid hormone beta receptor agonist enhances human and rodent oligodendrocyte differentiation. Glia. 2014; 62:1513-1529.
  272. Berbel P, Auso E, Garcia-Velasco JV, Molina ML, Camacho M. Role of thyroid hormones in the maturation and organisation of rat barrel cortex. Neuroscience. 2001; 107:383-394.
  273. Berbel P, Guadano-Ferraz A, Martinez M, Quiles JA, Balboa R, Innocenti GM. Organization of auditory callosal connections in hypothyroid adult rats. Eur J Neurosci. 1993; 5:1465-1478.
  274. Marin-Padilla M. Three-dimensional structural organization of layer I of the human cerebral cortex: a Golgi study. J Comp Neurol. 1990; 299:89-105.
  275. Del Rio JA, Heimrich B, Borrell V, Forster E, Drakew A, Alcantara S, Nakajima K, Miyata T, Ogawa M, Mikoshiba K, Derer P, Frotscher M, Soriano E. A role for Cajal-Retzius cells and reelin in the development of hippocampal connections. Nature. 1997; 385:70-74.
  276. Rakic P. Evolution of the neocortex: a perspective from developmental biology. Nat Rev Neurosci. 2009; 10:724-735.
  277. García-Fernández LF, Rausell E, Urade Y, Hayaishi O, Bernal J, Muñoz A. Hypothyroidism alters the expression of prostaglandin D2 synthase/beta trace in specific areas of the developing rat brain. Eur J Neurosci. 1997; 9:1566-1573.
  278. Laslett AL, Li LH, Jester WF, Jr., Orth JM. Thyroid hormone down-regulates neural cell adhesion molecule expression and affects attachment of gonocytes in Sertoli cell-gonocyte cocultures. Endocrinology. 2000; 141:1633-1641.
  279. Avci HX, Lebrun C, Wehrle R, Doulazmi M, Chatonnet F, Morel MP, Ema M, Vodjdani G, Sotelo C, Flamant F, Dusart I. Thyroid hormone triggers the developmental loss of axonal regenerative capacity via thyroid hormone receptor alpha1 and kruppel-like factor 9 in Purkinje cells. Proc Natl Acad Sci U S A. 2012; 109:14206-14211.
  280. Fauquier T, Chatonnet F, Picou F, Richard S, Fossat N, Aguilera N, Lamonerie T, Flamant F. Purkinje cells and Bergmann glia are primary targets of the TRalpha1 thyroid hormone receptor during mouse cerebellum postnatal development. Development. 2014; 141:166-175.
  281. Nicholson JL, Altman J. The effects of early hypo- and hyperthyroidism on the development of the rat cerebellar cortex. I. Cell proliferation and differentiation. Brain Res. 1972; 44:13-23.
  282. Desouza LA, Sathanoori M, Kapoor R, Rajadhyaksha N, Gonzalez LE, Kottmann AH, Tole S, Vaidya VA. Thyroid hormone regulates the expression of the sonic hedgehog signaling pathway in the embryonic and adult Mammalian brain. Endocrinology. 2011; 152:1989-2000.

 

Thyroid Nodules and Cancer in the Elderly

ABSTRACT

 

Thyroid nodules and thyroid cancer are common in elderly patients and demonstrate age-specific prevalence, malignancy risk, and clinical behavior. Improved risk stratification by ultrasound characteristics and molecular testing of thyroid nodules pre-operatively has reduced the need for diagnostic surgery in many individuals. In patients with differentiated thyroid cancer, total thyroidectomy and radioactive iodine followed by thyroid stimulating hormone (TSH) suppression remain the mainstays of therapy. However, newer approaches of active surveillance and thyroid lobectomy have expanded treatment options for patients with low risk differentiated thyroid cancer. Co-morbid conditions and patient preference should inform management of thyroid nodules and thyroid cancer in the elderly, with particular attention to the risks of surgery and medication adverse effects. Furthermore, the mechanisms underlying the distinct clinical behavior of thyroid cancer found in older patients, including the drivers of more advanced stage at presentation, higher recurrence risk, and greater mortality, remain poorly understood. Patients with advanced thyroid cancer may benefit from recently developed targeted and immune therapies.

 

THYROID NODULES IN THE ELDERLY

 

Thyroid nodules are common in clinical practice and present unique management issues in elderly patients. The reported prevalence of thyroid nodules in iodine sufficient regions is 1-6% as detected by palpation, or as high as 19-68% when detected by ultrasound imaging (1-5). Evaluation of thyroid nodules is increasingly a concern for general internists and endocrinologists in the context of an aging population, increased use of imaging in clinical practice, and rising obesity.

 

Thyroid nodules are more frequent in elderly patients, with a linear increase with age in both the presence of nodules and the absolute number of nodules per patient (6). Approximately 50% of individuals aged 65 years have thyroid nodules detected by ultrasonography (7). A cross-sectional survey of asymptomatic adults in Germany using ultrasonography to detect thyroid nodules demonstrated an even higher prevalence of 80% in women and 74% in men over 60 years old (4). In a prospective study of 6,391 patients referred for thyroid nodules at a large academic center, Kwong et al. showed a linear increase in the number of thyroid nodules per patient with age, rising from an average of 1.55 nodules ≥1 cm in patients age 20–29 years old to a mean of 2.21 nodules ≥1 cm in patients ≥70 years old, demonstrating a 1.6% annual increased risk for multinodularity (6).

 

Another potential contributor to this rising prevalence of thyroid nodules is the increased use of high-frequency ultrasound, CT, and MR imaging in routine clinical care, leading to the detection of asymptomatic, or incidental, thyroid nodules (4,5,7,8). Lastly, changes in population demographics over time, specifically increased rates of obesity, may contribute. Data from several ethnically diverse cohorts has identified parameters independently associated with the development of thyroid nodules, including obesity, female sex, radiation exposure, iodine deficiency, and smoking. These factors should be noted when evaluating elderly patients for potential thyroid nodules (9).

 

THYROID NODULE EVALUATION  

 

Once identified, thyroid nodules should be evaluated to determine appropriate management. The differential diagnosis of thyroid nodularity includes benign and malignant solitary nodules, multinodular goiter, autonomous functioning nodules, cysts, and inflammation or thyroiditis (10). Nodules causing thyroid dysfunction, compressive symptoms, or harboring malignancy require attention.

 

In the presence of biochemical and/or clinical signs of hyperthyroidism, a radioiodine uptake and scan should be pursued to distinguish autonomous nodules. Adjunctive data to support a diagnosis of inflammation or autoimmune destruction may include thyroid autoantibodies [anti-thyroid peroxidase (TPO) and anti-thyroglobulin (Tg)]. In addition, the presence of thyroid stimulating immunoglobulins suggest a diagnosis of Graves’ disease in the presence of goiter (11).

 

Nodules without associated thyroid function abnormalities should be further evaluated to determine or exclude the presence of cancer. Guidelines from the American Thyroid Association summarize the management of non-functional thyroid nodules based upon imaging and patient characteristics (12). A general approach to evaluation of thyroid nodules is shown in Figure 1.

Figure 1. Approach to evaluation of thyroid nodules. Evaluation always includes measurement of thyroid function tests, including thyroid stimulating hormone (TSH), and thyroid ultrasound. If hyperthyroidism is present, a thyroid uptake to exclude an autonomous functioning nodule (i.e. hyperfunctioning thyroid follicular tissue producing thyroid hormone excess). Cold nodules refer to nodules without autonomous production of thyroid hormone. Systems developed for malignancy risk stratification of thyroid nodules on ultrasound include the 2015 American Thyroid Association guidelines (ATA) and the American College of Radiology (ACR-TIRADS). Thyroid fine needle aspiration (FNA) biopsy should be considered for nodules with intermediate or high risk of malignancy based upon size and patient specific factors.

Thyroid ultrasound is the most important imaging modality in the assessment of thyroid nodules. Multiple systems have been developed to stratify thyroid nodules by their malignancy risk based upon ultrasound findings and provide recommendations for FNA biopsy (12-15). A systematic review and meta-analysis of eight studies including 13,092 thyroid nodules compared the diagnostic performance of four commonly used ultrasound-based risk stratification systems: the American College of Radiology Thyroid Imaging and Reporting System (ACR-TIRADS), the American Thyroid Association (ATA), the Korean Thyroid Imaging and Reporting System (K-TIRADS) and European Thyroid Imaging and Reporting System (EU-TIRADS) (12-16). This analysis found that the pooled rate of unnecessary FNA biopsies (i.e., those with a benign cytology result) was significantly lower with ACR-TIRADS (25%) when compared to ATA (51%, p<0.001) and K-TIRADS (55%, p<0.001), and not statistically different from EU-TIRADS (38%, p=0.087) (16). The diagnostic odds ratios among these four systems was similar (16). Features of thyroid nodules commonly associated with a higher risk of malignancy across these systems include solid and hypoechogenic appearance, irregular margins, microcalcifications, taller than wide shape, and evidence of extrathyroidal extension (12-15). Iso- or hyperechogenic appearance, smooth margins, and spongiform or partially cystic composition are features less associated with malignancy (12-15). Based upon ultrasound evaluation, nodules with highest risk for malignancy are recommended to have further evaluation by thyroid biopsy.

 

THYROID NODULE BIOPSY

 

Fine needle aspiration (FNA) biopsy is the recommended modality for sampling thyroid nodules. Cytology specimens collected by FNA are classified traditionally by the Bethesda System for Reporting Thyroid Cytopathology (17) across six categories: (i) non-diagnostic or unsatisfactory; (ii) benign; (iii) atypia of undetermined significance (AUS) or follicular lesion of undetermined significance (FLUS); (iv) follicular neoplasm or suspicious for a follicular neoplasm; (v) suspicious for malignancy; and (vi) malignant (Table 1). The risk of malignancy determined by surgical pathology is estimated across each category and used to guide decisions about continued clinical observation or treatment with surgical resection (17).

 

Table 1. Bethesda System for Reporting Thyroid Cytopathology and Associated Estimated Risk of Malignancy.

Bethesda category

Cytopathology

Cytologic descriptions

Malignancy risk

  Cancer = NIFTP

  Cancer ≠ NIFTP

Typical management

I

Non-diagnostic

 

Acellular specimen

Cyst fluid only

Obscuring factors

5-10%

5-10%

Repeat FNA

 

Ii

Benign

 

Benign follicular nodule

Chronic lymphocytic thyroiditis

Granulomatous thyroiditis

0-3%

0-3%

Clinical and ultrasound follow-up

Iii

Atypia of undetermined significance (AUS) or follicular lesion

of undetermined significance (FLUS)

Atypia: Cytologic (focal nuclear changes, extensive but mild nuclear changes, atypical cyst lining cells, or ‘‘histiocytoid’’ cells) and/or architectural (predominantly microfollicles, sparsely cellular); Hurthle cells

6-18%

10-30%

Repeat FNA, molecular testing, or diagnostic lobectomy

Iv

Follicular neoplasm or suspicious for a follicular neoplasm

 

Follicular-patterned cases with mild nuclear changes (increased nuclear size, nuclear contour irregularity, and/or chromatin clearing), and lacking true papillae and intranuclear pseudo-inclusions

10-40%

25-40%

Molecular testing or diagnostic lobectomy

V

Suspicious for malignancy

 

Features suspicious for PTC, MTC, lymphoma, or other malignancy

45-60%

50-75%

Total thyroidectomy or lobectomy

Vi

Malignant

 

 

Features conclusive for malignancy:

PTC (true papillae, psammoma bodies, nuclear pseudo-inclusions)

MTC

Poorly differentiated / ATC

Non-endocrine malignancy (squamous cell, lymphoma, metastatic)

 

94-96%

97-99%

Total thyroidectomy or lobectomy

PTC, papillary thyroid carcinoma. MTC, medullary thyroid cancer. ATC, anaplastic thyroid cancer. NIFTP, noninvasive follicular thyroid neoplasm with papillary-like nuclear features.

 

In situations of non-diagnostic FNA results or indeterminate cytology (i.e., Bethesda iii or iv), repeat FNA biopsy is recommended. Additionally, three molecular tests are now available for further cancer risk stratification and can reduce the number of thyroid surgeries performed for ultimately benign lesions (18). The ThyroSeq v3 multigenomic classifer (University of Pittsburgh Medical Center and CBL PATH, Pittsburgh, PA) is a DNA based assay that detects the presence of high-risk cancer mutations and was initially developed as a rule-in test for thyroid cancer (19). The current version, ThyroSeq v3, incorporates 112 genes associated with thyroid cancer and has a reported sensitivity of 98% and specificity of 81% for detection of thyroid cancer from FNA samples (20). The Afirma genomic sequencing classifier (GSC) (Veracyte, San Francisco, CA, USA) evaluates mRNA expression associated with benign or malignant profiles and detects thyroid cancer-associated mutations (21, 22). The Afirma GSC has a reported sensitivity of 91% (95% CI, 79-98) and a specificity of 68% (95% CI, 60-76) for thyroid cancer (21). A recent randomized clinical trial compared the diagnostic performance between the Thyroseq v3 and Afirma GSC assays in 346 patients with 427 cytologically indeterminate nodules (median age, 55 years) (18). This study found that both molecular tests showed high specificity for thyroid cancer with no significant differences in diagnostic performance, leading to 49% of patients avoiding diagnostic surgery. Lastly, the combined ThyraMIR microRNA Classifier and ThyGenX Oncogene Panel (Interpace Diagnostics, Parsippany, NJ) is a cancer rule-in test that uses multiplex PCR to identify cancer-associated gene mutations and translocations, done in tandem with evaluation of microRNA expression. The test estimated negative predictive value and positive predictive value are 94% and 74%, respectively (23). As molecular testing continues to evolve, clinicians and patients will have additional tools to aid in treatment decisions.

 

AGE-SPECIFIC NODULE PREVALENCE AND MALIGNANCY RISK

 

Several studies have specifically addressed thyroid cancer risk and nodule management across the age spectrum. Kwong et al. (6) reported the rate of malignancy in a cohort of 6,391 patients referred to a large academic center who underwent thyroid ultrasound and FNA of 12,115 nodules (all ≥1 cm). With advancing age, the prevalence of clinically relevant (>1 cm) thyroid nodules increased, whereas the risk that such nodules were malignant decreased. For patients ages 20–29, 30–39, 40–49, 50–59, 60–69, and >70 years, the cancer prevalence was 22.9, 21.8, 17.1, 13.0, 13.7, and 12.6%, respectively (p<0.001). When the malignancy rate was analyzed “per-nodule,” the youngest cohort (20–29 years) demonstrated a 14.8% malignant risk per nodule at diagnosis in comparison to 5.6% in the oldest cohort (>70 y; p<0.01). Between the ages of 20 and 60 years, each advancing year was associated with a 2.2% reduction in the relative risk that any newly evaluated thyroid nodule was malignant (OR 0.972; p<0.001), and this risk of malignancy stabilized after age 60 years. However, this study also found that despite a lower likelihood of malignancy for nodules in elderly patients, these cancers were more likely to have aggressive phenotypes (6).

 

Further addressing the burden and risk of thyroid nodule evaluation in older patients, Angell and colleagues analyzed a large cohort of elderly patients (age 70 years and older) who underwent thyroid nodule evaluation over a 20-year period (24). In this study, 1,129 patients over the age of 70 years with 2,527 nodules ≥1 cm were evaluated. Thyroid cancer-specific mortality was observed in 8% of thyroid cancer patients. All such patients could be recognized during initial evaluation based on the presence of invasive tumor, extensive lymph node metastases, or distant metastases. While FNA was a safe procedure in this age-group and a benign result was obtained in two-thirds of samples, FNA led to surgery in 208 patients, of whom 93 (44.7%) had benign histopathology. These data suggest that while an identifiable group of older patients are at risk for mortality from thyroid cancer warranting aggressive treatment, many patients ≥70 years old derive little benefit or are even harmed by thyroid nodule therapy.

 

Judicious use of FNA biopsy, improved stratification of nodule cancer risk by ultrasound characteristics, and molecular testing have improved pre-operative determination of malignancy risk in patients with thyroid nodules and reduced the need for diagnostic surgery. However, a significant number of patients who undergo thyroid nodule resection for suspicious nodules are still ultimately found to have benign lesions on surgical histopathology. Particularly in elderly patients with a greater burden of co-morbid medical disease, the risk of unnecessary thyroid surgery is an important consideration.

 

DIFFERENTIATED THYROID CANCER IN THE ELDERLY

 

While thyroid nodules are relatively common in elderly patients and the vast majority are benign (24), thyroid cancer is identified in a subset. Patients and their families are often concerned about the implications of this diagnosis and disease outcomes. Several subtypes of thyroid cancer are frequently encountered and increasing information about the underlying biology of these malignancies is now available. Most thyroid cancers are identified incidentally on imaging rather than by palpation on physical examination. Rarely, symptoms of thyroid cancer can include lymphadenopathy, hoarseness from laryngeal nerve involvement, dysphagia, airway compression from mass effect, or pain; when present, these symptoms portend more advanced disease and worse clinical prognosis (25, 26). When thyroid cancer is identified, a combination of surgical, radioactive iodine, and surveillance strategies are employed and tailored to the individual patient and disease characteristics.

 

Incidence and Prevalence of Thyroid Cancer

 

Thyroid cancer currently accounts for 2.3% of all new cancers, with an estimated 44,280 new cases in 2021, but only 0.4% of cancer deaths, in the United States annually (27). In the general population, the peak occurrence is between ages 51 and 60 years (28). Thyroid cancer is more common in women than men and among those with a family history of thyroid disease (27).

 

The incidence of thyroid rose over the past few decades, from an incidence of approximately 5 new cases per 100,000 persons per year in 1975 to a peak of 15 new cases per 100,000 in 2014 (27). The rate of thyroid cancer more recently has remained near 13 to 14 cases per 100,000 (27). Notably, small (<2 cm) papillary thyroid cancers account for the majority of this increase (29), and despite a much higher incidence, the death rate from thyroid cancer has remained stable (27, 30), likely reflecting greater detection of early disease associated with a good prognosis.

 

Classification of Thyroid Cancer

 

Thyroid follicular cell-derived cancer is subdivided into several histopathologic types: papillary thyroid carcinoma (PTC), follicular thyroid carcinoma (FTC), Hurthle cell carcinoma (HCC), and poorly differentiated or anaplastic thyroid cancer (31-33). Other malignancies encountered in the thyroid include medullary thyroid cancer arising from thyroid gland C-cells (discussed below), lymphoma, and secondary metastasis of other primary cancers.

 

Papillary thyroid cancer (PTC) is the most common type of differentiated thyroid cancer (DTC) accounting for approximately 80 to 85% of all cases (27, 31-33). It has a bimodal frequency, with the peak incidence being in the third and sixth decades, and it affects women three times more often than men. These carcinomas arise from the thyroid follicular cells and frequently harbor BRAF V600E mutations, produce thyroglobulin (Tg), and express the sodium-iodide symporter (NIS) with resultant radio-iodine avidity (31). A history of radiation exposure increases the risk of PTC (34-36). PTC frequently spreads via the lymphatics to the regional lymph nodes, and bilateral involvement is present in approximately one-third of the cases at diagnosis. In rare cases, metastatic disease occurs in the lungs, brain, and bone (31).

 

Micropapillary thyroid cancer, defined as a PTC less than 1 cm in diameter and confined to the thyroid, is likely to be of minimal clinical significance (37). A prospective, observation study of papillary thyroid microcarcinoma in Japan, found that in patients less than age 40 the microcarcinoma progressed to clinical disease, (defined as significant growth, size >1.2 cm, or lymph node metastases), in contrast to those over age 60, whose disease remained static (38), suggesting that in most elderly patients these lesions can be safely observed.

 

Follicular thyroid cancer (FTC) is the second most common type of DTC and constitutes approximately 10 to 15% of all thyroid cancers (27, 31, 32). Risk factors include iodine deficiency and female sex (27, 39, 40). Compared to PTC, FTC less often has cervical lymph node spread but shows a predilection for vascular invasion and distant metastasis (41). Mutations of RAS, an activator of the mitogen-activated protein kinase and PI3K-AKT pathways, and rearrangements of PPAR-γ (e.g., PAX8-PPAR-γ translocation) have been implicated in the tumorigenesis of follicular adenomas and FTC (41, 42).

 

Hurthle cell carcinomas (HCCs) account for 5% of DTC and are characterized by an abundance of dysfunctional mitochondria (>75% of cell volume) and tendency for vascular invasion (43, 44). These malignancies are more often radio-iodine refractory and aggressive in clinical behavior. Unique genetic drivers of HCC have been reported, namely widespread loss of heterozygosity, a high burden of disruptive mutations to protein-coding and tRNA-encoding regions of the mitochondrial genome, and recurrent mutations in DAXX, TP53, NF1, CDKN1A, ARHGAP35, TERT promoter, and the RTK/RAS/AKT/ mTOR pathway (45, 46).

 

Anaplastic thyroid cancer is rare and discussed separately.

 

Variation in Histopathology and Tumor Extent by Age

 

Several studies have shown variance in histopathology distribution with rising age. Lin et al. (47) conducted a retrospective analysis of 204 thyroid cancer patients aged 60 years and older; 142 (70%) thyroid cancers were well differentiated and of those 68% were PTC, 30% FTC, and 2% Hurthle cell carcinoma. Fifty-nine (29%) of the thyroid cancers were poorly differentiated (39 anaplastic thyroid, 9 metastatic cancers to the thyroid, 7 lymphoma, 4 squamous cell carcinomas, and 4 without enough cells for interpretation) and 3 (2%) were medullary thyroid cancer. This pattern is significant for fewer PTC and more FTC in elderly patients, as well as more poorly-differentiated tumors.

 

Girardi et al. conducted a retrospective study of thyroid cancer in 596 adults from 2000-2010; their results similarly showed a lower frequency of PTC among elderly patients, with a complementary increase in the frequency of FTC, poorly differentiated and anaplastic thyroid carcinoma (28). This study also demonstrated variability in other presenting features of thyroid cancer in elderly patients (age ≥ 65 years) compared to middle-aged cohorts (25-44 years or 45-64 years); specifically, there was larger primary tumor size (median 2.1 cm for elderly versus 1.5 cm in 25-44 years and 1.1 cm in 45-64 years) and higher rates of extrathyroidal disease (mean 43% for elderly versus 25.3% in 25-44 years and 28.6% in 45-64 years) (28). Lymph node metastasis was greatest at the extremes of age (<24 and >70 years).

 

Similarly, Chereau et al. evaluated histopathology and extent of disease at diagnosis in elderly (65-75 years old) and very elderly (>75 years old) patients compared to younger patients in 3,835 patients treated at an academic center from 1978 to 2014 (48). These data were notable for significantly increased primary tumor size, tumor number, extra-capsular invasion, advanced TNM stage, and lymph node and distant metastasis in the very old group (48). Collectively these studies show a pattern of more widespread disease at presentation in elderly patients and a relative increase in the frequency of more aggressive histologic subtypes.

 

Relation of Age to Mortality and Risk of Recurrence

 

Numerous studies have demonstrated increased recurrence and mortality in thyroid cancer with rising age (49-54). Indeed, age is incorporated into current clinical staging systems for differentiated thyroid cancer, including the American Joint Committee on Cancer (AJCC) 8th edition (55); Metastasis, Age, Completeness of resection, Invasion, Size (MACIS) model (56); Age, Grade, Extent, Size (AGES) score; and the Age, Metastasis, Extent, Size (AMES) score (57). In all of these staging systems, advanced age is included as a risk factor predicting worse prognosis.

 

Historic studies by Halnan (58) and Cady et al. (59) established a positive correlation between advanced age and worse prognosis in patients with DTC, later corroborated by Ito et al. (60) in a study of 1,740 patients with PTC and by Sugino et al. (61) in 134 patients with FTC. In many of these studies, worse prognosis has been defined variably as recurrence, decreased disease- or metastasis-free survival, cause-specific mortality, and/or overall mortality. Other reports have shown that the presence of lymph node involvement and extrathyroidal extension may portend a more ominous outcome in older compared to younger patients (59, 62-64). Extrathyroidal disease in older patients increased recurrence to 67% and death rates to 60% compared to those with intrathyroidal disease, while in younger patients the relative increases were 12% and 4%, respectively (59). Additionally, the risk of death with distant metastasis is greater in older compared to younger patients (96% versus 63%) (59).

 

Recently, this well-accepted tenet of thyroid cancer has been modified in two important ways, namely that age likely modifies prognosis in a continuous rather than dichotomous manner and that age itself may not be as relevant to thyroid cancer behavior as the accompanying changes in accumulated cell mutations, immune senescence, and hormone changes that accompany it (65).

 

With the 8th edition of AJCC staging for differentiated thyroid cancer, the age threshold for increased risk was raised from 45 to 55 years, based upon several reports suggesting that this increased validity for staging (66, 67). More recent data suggest that thyroid cancer mortality and recurrence prediction is more robust when age is modeled as a continuous variable, leading some to suggest the elimination of a specific age cutoff from staging completely (65).

 

In a study of 3,664 patients with differentiated thyroid cancer, Ganly et al. found that disease-specific mortality increased progressively with advancing age, without a threshold age (54). Similarly, evaluation of over 30,000 patients in the SEER database by Orosco et al. demonstrated a linear association with age and thyroid cancer death (53).

 

A review by Haymart et al. summarizes possible biologic mechanisms underlying the clinical observations of worse thyroid cancer prognosis in the elderly (51). Briefly, mortality findings may be confounded by greater comorbid nonthyroidal diseases with older age. Higher baseline levels of thyroid-stimulating hormone (TSH) may accelerate tumor cell growth via stimulation of the TSH-receptor. If one presumes that thyroid cancers detected in elderly patients have had a longer time of subclinical growth and evolution compared to cancers detected in younger patients, then such tumors might have had greater opportunity to acquire genetic mutations facilitating cell cycle escape, loss of differentiated features (e.g., loss of sodium-iodine symporter and radioiodine avidity), and metastasis. In summary, there is significant observational evidence that older patients with thyroid cancer have worse clinical outcomes, though the precise effect of increasing age and the etiology of this distinct clinical behavior remain incompletely understood.

 

Treatment of Differentiated Thyroid Cancer

 

Historically, differentiated thyroid cancers were treated with complete surgical resection of the thyroid gland combined with thyroid hormone suppression of TSH and radioactive iodine adjuvant therapy. More recently, recognition of the overall good prognosis and low disease specific survival in patients with DTC (68) has shifted management toward greater consideration for partial thyroid surgery, reduced use of radioactive iodine (RAI) in patients with low risk of recurrent disease, and active surveillance of some cancers. Treatment strategies for progressive or metastatic disease include repeat surgery, RAI ablation, or systemic therapies (12). A general approach to the treatment of differentiated thyroid cancers is presented in Figure 2.

Figure 2. Approach to the treatment of differentiated thyroid cancers (DTC). RAI, radioactive iodine. TSH, thyroid stimulating hormone.

Management will be influenced by patient characteristics, such as age, comorbid conditions, and preference for invasive or conservative therapy, and modalities available at the treating center. Two competing facts in older patients must be considered in selecting appropriate therapy for each individual. First, as discussed above, thyroid cancer in elderly patients is associated with more aggressive histologic features and greater lymph node spread at diagnosis. On the other hand, thyroid-cancer related mortality remains very low and treatment-associated morbidity may pose a greater risk to the elderly patient (68, 69).

 

Papaleontio and colleagues recently demonstrated that competing causes of death in older patients (>65 years) with DTC contributed more to patient mortality than the underlying diagnosis of thyroid cancer (69). Among 21,509 elderly patients with thyroid cancer identified in the SEER-Medicare database, 4168 (19.4%) died of other causes versus 2644 (12.3%) died of thyroid cancer during the study period from 2000 to 2015, with median follow-up of 50 months. Specifically for DTC patients, the likelihood of dying from other causes exceeded the likelihood of dying from thyroid cancer. A competing risks hazards regression analysis showed that heart disease [HR 1.34; CI (1.25–1.44)], chronic lower respiratory disease [HR 1.25; CI (1.17–1.34)], and diabetes mellitus [HR 1.14; CI (1.06-1.21)] were associated with death from other causes. Increased probability of death from thyroid cancer was associated with non-papillary histology [e.g., FTC HR 1.29; CI (1.12-1.48), or anaplastic HR 5.51; CI (4.82-6.31)], larger tumor size [ >4cm HR 3.35; CI (2.71-4.15)], and regional or distant metastatic disease [HR 4.59; CI (3.98-5.31) and HR 12.65; CI (10.91-14.66), respectively]. Progressively advancing age was associated with an increased probability of death from both other causes and thyroid cancer. In summary, this suggests that in elderly patients a diagnosis of thyroid cancer may not be the most significant factor influencing life expectancy. A careful discussion of treatment options, including expected benefits and risks in the context of disease burden and comorbid conditions, is warranted with each patient.

 

SURGERY

 

Patients diagnosed with DTC by thyroid FNA, or with a nodule highly suspicious for malignancy, may be referred to a surgeon for thyroid resection. Total thyroidectomy aims to remove the primary tumor and normal thyroid tissue and remains the primary initial treatment for DTC (12). Thyroid lobectomy is a more limited surgery that removes only the primary tumor and ipsilateral normal thyroid lobe. Additional exploration and removal of central and lateral neck lymph nodes suspicious for cancer metastases may be done concurrently with either procedure, as guided by preoperative imaging or intraoperative findings. The decisions to pursue surgery and the extent of surgery (i.e., total thyroidectomy versus lobectomy) in an elderly patient require individual evaluation of co-morbid illnesses and life expectancy.

 

The most common complications of thyroidectomy include hypoparathyroidism, recurrent laryngeal nerve injury, hematoma, and wound infection; high-volume thyroid surgeons have minimal to no increase in the risk of surgical complications with increasing age (70-75). However, elderly patients are more likely to receive thyroidectomy at community and low-volume sites (76) where the rate of surgical complications may be higher. In population-based studies of thyroidectomy, which may reflect more accurately the experience of many elderly patients, increasing age is associated with longer hospital length of stay (76) and readmissions after thyroidectomy (77). In the cohort of elderly and very elderly patients studied by Chereau et al. (48), the authors found no increase in thyroidectomy-specific complications (i.e., permanent hypocalcemia and recurrent laryngeal nerve palsy) with increasing age, but did find an increase in medical complications surrounding surgery, 2.3-2.7% in those over 65 years of age compared to 0.6% in those under 65 years old.

 

LOBECTOMY

 

Thyroid lobectomy may be considered in select patients with low-risk disease (12, 70). The ATA guidelines revised in 2015 suggest that lobectomy is appropriate for DTC with a primary tumor size <4 cm and without extrathyroidal extension or clinical evidence of lymph node metastasis (12). Potential advantages of lobectomy over total thyroidectomy are lower rates of surgical complications from hypoparathyroidism and recurrent laryngeal nerve damage (78). In addition, some patients do not require thyroid hormone replacement after lobectomy to achieve the recommended low normal target TSH range of 0.5-2mIU/L (79).

 

On the other hand, patients who initially undergo lobectomy, but are found to have aggressive disease features (e.g.,extrathyroidal extension, lymph node metastasis) on surgical pathology, are encouraged to undergo completion thyroidectomy. This facilitates monitoring of disease with serum thyroglobulin (Tg) and treatment with adjuvant RAI (discussed below). Because elderly patients more often have aggressive disease features and higher rates of local recurrence requiring re-operation, some recommend initial total thyroidectomy in this population (73).

 

ACTIVE SURVEILLANCE  

 

Although surgery is the accepted initial management for most DTC, active surveillance may be an alternative strategy to immediate surgery for an appropriately selected group of patients (80). Multiple international retrospective studies with long term follow-up suggest that many small (<1-1.5cm), well-differentiated PTCs, without evidence of extrathyroidal extension or metastases, have low rates of growth and progression (38, 81, 82). In a cohort of 291 US patients with low-risk PTC followed by serial ultrasonography, Tuttle and colleagues (74e) showed significant growth in only a minority of patients over a median follow-up of 25 months: volume increase of greater than 50% in only 36 (12%) patients, and size increase greater than 3 mm in 11 (3.8%) patients. Ito and colleagues (38) similarly showed in a cohort of 1235 Japanese patients with small PTCs followed with active ultrasonographic surveillance for a median of 75 months that, by 5 and 10 years, only 4.9% and 8% of patients experienced tumor growth of more than 3 mm, and 1.7% and 3.8% of patients experienced new lymph node metastases, respectively. Importantly, in a subset of patients who ultimately underwent thyroid surgery in this study, none were found to have distant metastases and no patient died of PTC, suggesting that delaying intervention until the time of growth or detection of lymph node spread did not adversely affect mortality. Active surveillance for low-risk PTC may avoid unwarranted surgery, surgical complications, RAI administration, and lifelong thyroid hormone replacement therapy and should be considered for appropriate patients, particularly those with reliable follow-up, high surgical risk, shorter life expectancy, or with concomitant medical issues that need to be addressed before surgery (80, 83).

 

RADIOACTIVE IODINE ABLATION  

 

Based upon the extent of primary disease noted on surgical pathology (i.e., tumor size, extrathyroidal extension, lymph node and vascular spread), patients can be stratified by their risk for recurrent disease. Adjuvant therapy with radioactive iodine (I131; usual dose100-150mCi) is recommended for patients with a high risk of recurrence after total thyroidectomy, and RAI should be considered for patients with an intermediate risk of recurrence (12). An analysis of 21,870 patients with intermediate-risk PTC found that adjuvant RAI therapy was associated with a 29% reduced risk of death overall with clear benefit in those over 65 years of age (84). RAI may also be used as a treatment modality in patients with persistent or recurrent RAI-avid disease who are not surgical candidates, usually requiring doses of 150mCi or higher. Finally, lower doses of RAI (30mCi) may be used to ablate remnant normal thyroid tissue and improve the utility of serum Tg tumor marker monitoring even in patients with a lower risk of recurrence. Two multicenter studies showed that an ablative dose of 30 mCi (1.1 MBq) I131 was as effective as 100 mCi (3.7 MBq); both doses were 90% effective for ablation of residual thyroid tissue (85, 86). A long-term follow-up of one of these studies (median 4.5 years) showed that the radioiodine dose selected for remnant ablation did not affect recurrence rate (87).

 

Treatment benefits of RAI should be weighed against side effects. The adverse effects of RAI therapy are increasingly recognized and include transient neck pain and swelling, decreased fertility, dry mouth and eyes, and secondary malignancy and are correlated with higher I131 doses (88).

 

In patients initially treated with lobectomy or active surveillance for low-risk DTC, surgical removal of remaining normal thyroid tissue is recommended prior to RAI use.

 

THYROID STIMULATING HORMONE (TSH) SUPPRESSION

 

Following surgery, and RAIA if indicated, patients are treated with thyroid hormone, usually with a dose of levothyroxine that suppresses serum TSH to subnormal levels. Several special considerations for the goals of thyroid hormone therapy following thyroid cancer arise in elderly patients.

 

Thyroid hormone replacement is titrated to levels sufficient to suppress pituitary secretion of TSH, which is considered a growth-promoting factor for follicular cell-derived thyroid cancers. Revised guidelines from the ATA (12) suggest individualized targets for TSH suppression in thyroid cancer, generally targeting a low to low-normal range TSH. Greater TSH suppression in more aggressive disease is balanced with greater cardiac and bone complications in elderly patients.

 

Older patients are more likely to have co-morbid cardiac disease, including arrhythmias, coronary artery disease, and heart failure, which can place them at increased risk for complications from thyroid hormone excess. A population-based study of patients taking levothyroxine for any cause, found a significantly higher risk of cardiac arrhythmias [HR 1.6 (1.10–2.33)] and cardiovascular admission or death [1.37 (1.17–1.60)] in those with a suppressed serum TSH (≤0.03 mU/L) compared to those with TSH in the normal reference interval (89). Notably, increased cardiovascular risk was not observed in patients with a low but not fully suppressed TSH (TSH 0.04 – 0.4 mU/L). Specifically, in thyroid cancer patients treated with levothyroxine with modestly suppressed TSH (mean TSH <0.35 mU/L), atrial fibrillation was common (17.5% prevalence) in those patients ≥60 years old (89).

 

Longstanding hyperthyroidism is associated with osteoporotic fractures and loss of bone mineral density. Specifically, post-menopausal women (≥65yo) with suppressed TSH levels (0.1 mU/L) due to endogenous or exogenous thyroid hormone had significantly higher rates of new hip (OR 3.6, 95% CI 1.0-12.9) and vertebral fractures (OR 4.5, 95% CI 1.3 -15.6) compared to comparable women with normal TSH levels over a 3.7 years follow-up (90). In adult patients on levothyroxine therapy, a suppressed TSH (≤0.03 mU/L) was associated with a two-fold increase in risk [HR 2.02 (1.55–2.62)] of new osteoporotic fracture compared to similar patients treated with levothyroxine with a TSH maintained in the normal reference interval (89). Studies evaluating thyroid cancer patients are limited in outcome evaluation of bone mineral density (BMD) rather than fracture incidence, but generally support similar conclusions regarding lower BMD with suppressive-dose levothyroxine therapy (91-93). In elderly patients receiving TSH-suppression therapy, dual-emission X-ray absorptiometry (DEXA) monitoring of BMD should be considered based upon age and other risk factors for osteoporosis. There are no guidelines to suggest the optimal interval for DEXA screening; osteoporosis once identified should be treated using standard therapies (such as bisphosphonates or RANKL inhibitor) unless otherwise contraindicated (94).

 

Peripheral metabolism of thyroid hormone and clearance decreases with advanced age so that a lower medication dose is needed to achieve comparable serum levels (95, 96). Levothyroxine therapy is complicated further by polypharmacy in elderly patients, where commonly prescribed medications (e.g., calcium, iron) can decrease gut absorption of levothyroxine (97) or change drug metabolism (e.g., rifampicin, phenytoin, carbamazepine, amiodarone) (98). In summary, as suggested by society guidelines (12), TSH goals in thyroid cancer should be individualized and re-evaluated over time.

 

POSTOPERATIVE SURVEILLANCE

 

Recommended follow-up of DTC includes biochemical surveillance with measurement of serum thyroglobulin (Tg) and Tg antibody (Tg Ab) concentrations and structural surveillance with neck ultrasonography at clinically appropriate intervals (12).

 

Tg is a thyroid-specific protein that can be measured in blood to monitor for the presence of thyroid cancer, as well as remnant normal thyroid tissue. As such, serum Tg is used as a tumor marker for follicular cell-derived thyroid cancers, including PTC, FTC, and Hurthle cell thyroid cancer. Serum Tg levels are initially checked 4-6 weeks after total thyroidectomy, and then at intervals of 6-12 months (12). The trends of serum Tg overtime are most informative, with a rising Tg concerning for disease recurrence. While Tg levels are usually monitored in the context of a suppressed TSH, a stimulated Tg measurement may provide a more sensitive evaluation for persistent or recurrent disease (99). Finally, the measurement of serum Tg is confounded by the presence of Tg antibodies (Tg Ab), which occurs in approximately 20% of patients with DTC and can mask recurrent or persistent disease by causing falsely low or undetectable serum Tg levels (100). Evidence from retrospective studies suggests that increasing TgAb levels (measured using validated assays), compared with stable or decreasing titers, can be used as a surrogate tumor marker in these patients (100, 101). A suppressed Tg level >0.2 ng/mL after total thyroidectomy and RAI, a stimulated Tg level >2- 5 ng/mL, a rising Tg level, or the persistence of Tg antibodies, is concerning for further evaluation (12, 100). A persistently elevated (>0.2ng/mL Tg with TSH suppression or >2ng/mL stimulated Tg) or rising serum Tg or Tg Ab level should prompt concern for persistent or recurrent disease (12).

 

In patients who have not received RAI after thyroidectomy, residual normal thyroid tissue may contribute to a higher baseline Tg level after thyroidectomy. In these patients, a suppressed Tg >1ng/mL or rising Tg level should prompt concern for disease recurrence. Similarly, because of the significant volume of normal thyroid tissue remaining in patients who have undergone thyroid lobectomy, the optimal use of Tg in these patients remains uncertain.

 

DYNAMIC RISK STRATIFICATION

 

The process by which serum Tg (or Tg Ab) levels and imaging surveillance data are combined to make an evolving assessment of disease status in DTC patients is called dynamic risk stratification (12, 102). Patients are classified across the spectrum from no biochemical or structural evidence of disease to definite persistent or recurrent thyroid cancer.

 

Patients with reassuring or low risk continue on the current regimen of tumor surveillance or are relaxed to a more conservative approach. In contrast, patients in whom DRS shows an increasing risk of thyroid cancer recurrence or progression are recommended to undergo further diagnostic evaluations to localize disease and/or additional treatment. In addition, levothyroxine therapy may be adjusted for greater TSH suppression and the interval of monitoring with serum tumor markers and imaging may be shortened.

 

Specifically, patients with no biochemical or structural evidence of disease are deemed to have an “excellent response” (12, 102). TSH suppression is relaxed to the low normal range (0.5-2mIU/L) to mitigate long term adverse effects of iatrogenic hyperthyroidism and annual tumor marker surveillance with or without thyroid ultrasound usual. A “biochemical incomplete” response indicates persistent abnormal Tg values or increasing TgAb levels in the absence of localizable disease (12, 102). An “indeterminate response” is defined as nonspecific biochemical or structural findings that cannot be confidently classified as representing malignant disease (12, 102). Patients with a “biochemical incomplete’ or “indeterminate response” have a TSH goal of 0.1 - 0.5mIU/L. In patients with clear recurrent or persistent disease on imaging and elevated Tg or TgAb serum markers are classified as structural incomplete response” (12, 102). TSH is maintained suppressed below 0.1mIU/L. In these patients, and some with “incomplete biochemical” or “indeterminate response,” additional evaluation with neck ultrasonography, whole-body RAI scanning, and/or PET/CT depending on level of clinical suspicion, is recommended to localize residual thyroid tissue/cancer. Identification of abnormal lymph nodes or tumor mass can then be evaluated for possible further treatment with RAI, surgery, or targeted therapy.

 

SYSTEMIC THERAPY

 

Patients with advanced and symptomatic DTC that cannot be treated with further surgery or RAI may benefit from systemic therapy. Older cytotoxic drugs have shown little benefit for progressive, advanced, or metastatic papillary or follicular thyroid cancer while causing significant side effects. Improved understanding of the pathogenesis of these cancers is leading to the development of new agents aimed at specific oncogenic mechanisms (e.g., RET, BRAF). Currently three tyrosine kinase inhibitors (sorafenib, lenvatinib, and cabozantinib) are approved for therapy of metastatic, RAI-resistant DTC.

 

Sorafenib, an oral multi-kinase inhibitor, inhibits vascular endothelial growth factor receptors (VEGFR-1, VEGFR-2, and VEGFR-3), RET kinase (including RET/PTC), BRAF V600E, and platelet-derived growth factor receptor (PDGFR) beta. In the DECISION phase 3 multicenter placebo-controlled trial of 416 patients, 409 had distant metastases: 86% in the lungs, 51% in lymph nodes, and 27% in bone (103). The group treated with sorafenib had longer progression-free survival (10.8 months) compared to the placebo group (5.8 months). At disease progression, 71% of patients in the placebo group crossed over to receive open-label sorafenib; as a consequence, overall survival did not differ between the two groups. Twenty percent of patients in the sorafenib group received other cancer therapy after the trial. The most frequent adverse events in the active drug group were palmar-plantar erythrodysesthesia, diarrhea, alopecia, rash, weight loss, hypertension, anorexia, oral mucositis and pruritus. Side effects were relieved by dose reduction.

 

Lenvatinib is a tyrosine kinase inhibitor of the VEGFRs 1, 2, and 3; fibroblast derived growth factor receptor (FGFR)s 1 through 4; PDGFRα; RET; and KIT signaling pathways. The SELECT phase 3 trial randomly assigned 261 patients to receive lenvatinib and 131 patients to receive placebo; the median age of patients in the trial was over 60 years (104). The median duration of follow-up was 17 months; 114 patients assigned placebo had progression, and 109 of them elected to receive lenvatinib. Disease progression occurred in 36% in the lenvatinib group compared to 83% in the placebo group. Median progression free survival was 18.3 months with lenvatinib versus 3.6 months with placebo. Disease response rate was 66% with lenvatinib compared with 1.5% with placebo. The benefit appeared in all subgroups, including all histologic types of tumor. Adverse events occurred in 97% of patients taking lenvatinib and in 60% taking placebo; the main adverse events were hypertension, diarrhea, fatigue, decreased appetite, palmar-plantar erythrodysesthesia, proteinuria, renal failure, and thromboembolic events.

 

Cabozantinib, another kinase inhibitor, was approved for use in metastatic, RAI-refractory DTC based upon the findings of the COSMIC-311 trial (105). This study showed longer progression free survival with cabozantinib treatment (n=125) compared to placebo (n=62): median not reached (96% CI 5.7 months-not estimable [NE]) versus 1.9 months (CI 1.8-3.6); hazard ratio 0·22 (96% CI 0.13-0.36; p<0·0001). Of note, patients included in this trial must have previously progressed on prior sorafenib and/or lenvatinib therapy, and those in the placebo arm were allowed to cross over to cabozantinib therapy on disease progression. Side effects of cabozantinib were similar to other kinase inhibitors, including palmar-plantar erythrodysesthesia, hypertension, and fatigue.

 

In 2020, two RET inhibitors, selpercatinib and pralsetinib, were approved for the treatment of advanced or metastatic RET-altered thyroid cancers, including medullary and follicular cell-derived. Selpercatinib (LIBRETTO-001) showed an overall response rate of 79% (95% CI, 54 to 94), and 1-year progression-free survival of 64% (95% CI, 37 to 82) in 19 patients with previously treated RET fusion-positive thyroid cancer (inclusive of PTC, Hurthle cell, poorly differentiated and analplastic) (106). The most common high grade (3-4) adverse events were hypertension, hepatotoxicity, hyponatremia, and diarrhea (106). A recently published update of LIBRETTO-001 outcomes (107), showed a persistent overall response rate of 77.3% (95% CI 54.6-92.2) with a median duratrion of response of 18.4 months (95% CI 10.1-not reached) and 68.6% (95% CI 42.8-84.6) of patients showing continued progression free survival >12 months. The efficacy evaluation for pralsetinib in patients with RET fusion–positive thyroid cancer was based on the phase I/II ARROW study (108) evaluating nine patients with RAI-refractory PTC; 56% had received prior lenvatinib and/or sorafenib and 22% had received cabozantinib and/or vandetanib. The overall response rate was 89% (95% CI 52-100), with all eight patients demonstrating partial response to treatment and effects lasting >6 months. Notable adverse events seen in thyroid cancer patients included hypertension, fatigue, cytopenia, and pneumonitis (108). Finally, larotrectinib and entrectinib are approved for metastatic or unresectable thyroid cancers with NTRK gene fusions and no alternative treatments (109, 110).

 

While not without side effects, these targeted kinase and RET inhibitors demonstrated efficacy in prolonging disease free survival in patients with metastatic, RAI-refractory DTC and should be considered in the symptomatic elderly patient with sufficient performance status and potential benefit. For differentiated thyroid cancer that progresses despite these therapies, additional treatment with external beam radiation, off label use of BRAF inhibitors, and clinical trials of immune checkpoint inhibitor therapies are sometimes utilized. These modalities are discussed below in the context of anaplastic thyroid cancer.

 

REDIFFERENTIATION THERAPY

 

Another treatment approach that has been explored in advanced, RAI-resistant follicular cell-derived thyroid cancers is the use of MEK or BRAF inhibitors for redifferentiation and restoration of RAI sensitivity. Several trials have been pursued based upon promising preclinical evidence showing increased sodium-iodine symporter expression and radioiodine uptake reinduction by modulation of the MAPK signaling pathway (111). An early phase study by Ho et al. (112) showed that in 20 patients with RAI-refractory advanced DTC, treatment with MEK inhibitor selumetinib for 4 weeks increased sensitivity to RAI uptake in 12 (60%) subjects (including 4/9 with BRAF and 5/5 with NRASmutations). Eight of 12 patients reached the pre-defined dosimetry threshold for repeat RAI therapy, of which five had an objective response and three had stable disease (112).

 

The subsequent phase III ASTRA study (113) was done to evaluate whether selumetinib given with initial RAI therapy in DTC patients with a high risk of primary treatment failure (i.e. ATA high risk of recurrence) would improve complete remission at 18 months and decrease the need for additional therapy. The addition of selumetinib to radioiodine did not improve the complete remission rate (40% vs. 38.5% in the placebo group). Another multi-center phase II prospective trial evaluating selumetinib in RAI-refractory DTC patients is currently underway (SEL-I-METRY, trial ISRCTN17468602) (114).

 

Several small studies have examined other BRAF (e.g. drabafenib) and MEK inhibitors (e.g. trametinib), alone or in combination, as reviewed recently by Buffet et al. (114). For example, Rothenberg et al. (115) showed resensitization in six of 10 BRAFV600E+ RAI-resistant thyroid cancer patients following six weeks of drabafenib therapy. In addition, following RAI therapy with 150mCi, two of the six patients showed a partial response and the remaining four had stable disease at three months. Another group, Dunn et al. (116) also evaluated vemurafenib in 10 patients with a BRAF-mutated-PTC or poorly differentiated thyroid cancer. After 4 weeks of vemurafenib, RAI uptake increased in 6/10 patients, and of four patients retreated with RAI therapy, two showed partial response and two had stable disease at six months. While a potentially promising adjuvant strategy for these challenging tumors, additional prospective evaluation is needed before this strategy can be considered within the standard of care.

 

ANAPLASTIC THYROID CANCER

 

Anaplastic thyroid carcinoma (ATC) is a rare and aggressive subtype of thyroid cancer that accounts for <1% of all thyroid cancers (27, 31). It more commonly affects the elderly, with a mean age at diagnosis of 65 years and more than 90% patients with ATC are over age 50 (31). Despite recent advances, the median overall survival remains poor, around 3–5 months, with a 1-year survival of approximately 20% (117). Aldinger et al. reported a five-year survival rate of only 7.1% with a mean survival period of 6.2 months from the time of tissue diagnosis and 11.8 months from the time of onset of symptoms (118).

 

The most frequent presenting complaint in patients with ATC is a rapidly growing mass with tightness in the neck (118). Patients may also complain of dysphagia, hoarseness, dyspnea, neck pain, sore throat, and cough. Examination of the neck usually reveals a fixed, large, firm mass, which may impair the ability to detect lymphadenopathy on clinical examination. Hemorrhage and necrosis within the tumor may result in soft, fluctuant masses. Rarely, patients with massive tumor extension into the mediastinum or lungs may present with superior vena cava syndrome or dyspnea.

 

Unfortunately, most patients with ATC present with advanced stage disease. In a retrospective study of thyroid cancers in 204 elderly (age >60 years) patients by Lin et al. (47), 75% of patients diagnosed with ATC had distant metastases to the lung, bone, mediastinum, and peritoneum at presentation. Similarly, in the cohort reported by Aldinger et al., 78 of 84 (93%) patients with ATC presented with advanced stage III and stage IV disease (118). Additional patient factors associated with worse prognosis in ATC include advanced age (>60–70 years), male gender, presence of leukocytosis (>10,000), and symptoms related to tumor mass effect, such as neck pain, dysphagia, rapidly growing neck mass. Regarding older age as a poor prognostic factor, in a cohort of 516 patients with ATC, Kebebew et al. reported a 28% greater mortality in patients over 60 years of age compared to those less than 60 years determined by multivariate analysis (117).

 

ATC often, but not always, arises from pre-existing differentiated thyroid cancer, with 20% of patients with antecedent DTC and another 20-30% with concurrent DTC (co-existent on histopathology). There is also a higher incidence of ATC in patients with endemic goiter. These associations are relevant for the treatment of ATC because driver mutations such as BRAF and RAS may be retained in the anaplastic tumor cells and can be targeted with therapy (31, 118).

 

Treatment of Anaplastic Thyroid Cancer

 

While the prognosis of ATC remains poor, treatment options to slow the progression of disease, palliate symptoms, and, in rare cases, attempt cure, are available as approved therapies and in clinical trials.

 

EXTERNAL RADIATION  

 

External radiation to the neck region is appropriate for patients with aggressive cancers that cannot be completely resected surgically (12). Schwartz et al. reported limited success in the treatment of RAI-refractory patients with extrathyroidal spread, positive surgical margins, or gross residual disease with a mean of 60 Gy (38-72 Gy); survival was less in patients with high-risk pathology, metastases, and gross residual disease (119). In the context of ATC, disease is often assumed to be radioiodine refractory, and external beam radiation may be used for preservation of vital neck structures.

 

TARGETED SYSTEMIC THERAPY AND IMMUNOTHERAPY

 

Most patients with ATC have rapidly progressive disease and should be evaluated for clinical trials when feasible as new treatments continue to be developed. Targeted therapy with inhibitors to specific gene mutations and fusions has shown some success and is the focus of numerous ongoing clinical trials. Therapies include inhibitors of BRAF, MEK, NTRK, RET, and ALK. Combination treatment with BRAF inhibitor dabrafenib and MEK inhibitor trametinib was recently approved for the treatment of BRAFV600E mutated, unresectable/locally advanced ATC, following a 69% overall response rate in a phase II open label trial of 16 patients with ATC (120).

 

Immunotherapy reagents target the impaired immune responses and immune suppression that arise in cancer allowing malignant cells to grow and spread. Immune checkpoint inhibitors are a kind of immunotherapy that block immune regulatory pathways with the goal of increasing anti-tumor immune responses and producing tumor killing by host leukocytes. Two primary classes of immunotherapy being evaluated for advanced thyroid cancer are inhibitors of cytotoxic T lymphocyte A (CTLA)-4 (such as ipilimumab) and inhibitors of programmed cell death (PD) receptor/ligand interactions (nivolumab, pembrolizumab, atezolizumab). Currently, immune checkpoint inhibitors are being evaluated alone and in combination with targeted therapies for ATC (120).

 

MEDULLARY THYROID CANCER

 

Medullary thyroid cancer (MTC) constitutes approximately 2-5% of all thyroid malignancies, but it is responsible for up to 13.4% of all deaths from thyroid cancer (30, 121). It is a well-differentiated type of tumor that arises from the parafollicular C cells of the thyroid gland, and therefore it is categorized as a neuroendocrine tumor. In 80% of patients, medullary thyroid cancer occurs sporadically, but in about 20% of patients there is a family history of medullary carcinoma. Familial MTC is inherited in an autosomal dominant pattern with nearly complete penetrance. A germline mutation in the RET proto-oncogene, which encodes a transmembrane tyrosine kinase receptor, predisposes individuals to develop hereditary MTC. In the sporadic form, the tumor occurs as a result of a mutation involving only the somatic cells. Sporadic forms of MTC are more common in older patients (mean age at presentation 47 years), while the hereditary forms of MTC are more common in younger patients (121). The prevalence of MTC is nearly equal in males and females.

 

Parafollicular cells secrete calcitonin, and in MTC this protein is greatly elevated and serum level correlates directly with the burden of disease (122). Other neuroendocrine cell products, including histamine, serotonin, prolactin, vasoactive intestinal polypeptide, and prostaglandin, can be elevated in patients with MTC and lead to systemic symptoms such as diarrhea or flushing (122). In some cases, Cushing’s syndrome may develop as a result of ectopic adrenocorticotrophic hormone (ACTH) secretion from the tumor. The typical presentation of MTC is a palpable nodule in the upper part of the thyroid lobe, and the presence of systemic symptoms is almost universally associated with distant metastases (37). In the retrospective report of 104 patients with MTC by Kebebew et al., 74% of the patients in the sporadic group presented with a thyroid mass, 16% had local symptoms (dysphagia, dyspnea, or hoarseness), and 10% had systemic symptoms (bone pain, flushing, and/or diarrhea) attributable to the cancer (121).

 

Within MTC, older age at diagnosis has been associated with a worse prognosis. Kebebew et al. followed patients with MTC for a mean time of 8.6 years and found that advanced age and stage at diagnosis were independent predictors of worse survival (121). The 5-year survival rates by stage were 100% (stage I), 90% (stage II), 86% (stage III), and 55% (stage IV). The highest survival was seen in female patients under age 45 with MTC confined to the thyroid (121). Saad et al. similarly reported that patients younger than 40 years old at diagnosis had a significantly better survival rate in MTC (122). Scopsi et al. reported a worse prognosis in patients with sporadic MTC who had extrathyroidal tumor invasion, distant metastases, or age greater than 60 years at the time of diagnosis (123). Interestingly, a more recent study that adjusted for baseline age-related mortality in the general population found no significant association with age and prognosis in MTC (124). This raises similar questions to those posed recently for differentiated thyroid cancer as to whether age truly has an independent role in prognosis for these thyroid cancers apart from the general increase in morbidity and mortality with aging.

 

Treatment of Medullary Thyroid Cancer

 

The standard treatment for MTC is surgical resection (total thyroidectomy) with regional lymph node dissection, with routine bilateral central neck dissection and consideration of lateral neck dissection in patients with large primary tumors (>1 cm) or pre-operative imaging with involved nodes. Successful complete surgical resection is associated with improved prognosis. In patients with disease restricted to the thyroid gland and without nodal involvement, the risk of recurrence and mortality is very low, compared to those with nodal disease at presentation (125).

 

Serum calcitonin and CEA levels are trended post-operatively to monitor for residual or recurrent disease, beginning around 2-3 months after surgery. A rise in either tumor marker should prompt imaging to look for recurrent disease. Radioactive iodine is not indicated in the treatment of MTC as parafollicular cells do not express NIS or concentrate iodine. Additionally, thyroid hormone replacement is required following thyroidectomy, with TSH targeted to the normal range rather than suppression (126). TSH does not stimulate the growth of parafollicular cells.

 

In patients with progressive or metastatic disease not amenable to surgery, tyrosine kinase inhibitors vandetanib and cabozantinib may be used. Vandetanib is an oral inhibitor that targets VEGFR, RET, and epidermal growth factor receptor (EGFR). In the international, randomized controlled phase III ZETA trial of vandetanib 300 mg per day that included over 300 patients with unresectable, locally advanced or metastatic sporadic or hereditary MTC, progression-free survival was significantly greater for patients treated with vandetanib (hazard ratio 0.46, 95% CI 0.31-0.69 versus placebo) (127). Adverse events occurred more commonly with vandetanib compared to placebo, including diarrhea, nausea, palmar-plantar erythrodysesthesia, hypertension, and headache.

 

Cabozantinib (128) is another oral tyrosine kinase inhibitor targeting MET, VEGFR2, and RET signaling pathways. The phase III international, randomized controlled EXAM trial evaluated cabozantinib versus placebo in the treatment of 330 patients with progressive, metastatic MTC, with a primary outcome of progression free survival (PFS). Median PFS was 11.2 months for cabozantinib versus 4.0 months for placebo (hazard ratio, 0.28; 95% CI, 0.19 to 0.40; P <0.001), with benefit seen across all subgroups including age, prior TKI treatment, and RET mutation status (hereditary or sporadic). Response rate was 28% for cabozantinib and 0% for placebo. Common cabozantinib-associated adverse events noted in the trial included diarrhea, palmar-plantar erythrodysesthesia, decreased weight, nausea, and fatigue.

 

As discussed above, two targeted RET-fusion inhibitors have recently been approved for use in MTC: selpercatinib (106) and pralsetinib (108). Selpercatinib showed a 69% (95% CI 55-81) overall response rate and 82% (95% CI 69-90) one year progression free survival in 55 patients with RET-mutated MTC patients who previously had failed treatment with vandetanib, cabozantinib, or both (106). In addition, in 88 patients RET-mutated MTC but without prior systemic therapy, the study found a 73% (95% CI 62-82) overall response and 92% (95% CI 82-97) one year progression free survival (106). Similarly, in the phase I/II study of pralsetinib, the overall response rates were 60% (95% CI 47-63) for patients with RET-mutated MTC patients who had previously received cabozantinib or vandetanib, or both, and 71% (95% CI 48–89) in patients with treatment-naive RET-mutated MTC (108). Side effects for both RET inhibitors were similar to those seen in follicular cell-derived thyroid cancer patients discussed above.

 

Given the poor prognosis of MTC, continued development of new treatment strategies is needed and management at a center experienced with this type of cancer is recommended.

 

CONCLUSION

 

In summary, thyroid nodules and cancer are common in elderly patients and demonstrate age-specific prevalence, malignancy risk, and clinical behavior. Co-morbid conditions and patient preference should inform management of these entities in the elderly, with particular attention to the risks of surgery and medication adverse effects. More research is needed to understand the mechanisms underlying the distinct clinical behavior of thyroid cancer found in older patients, including the drivers of more advanced stage at presentation, higher recurrence risk, and greater mortality.

 

REFERENCES

 

  1. Vander JB, Gaston EA, Dawber TR. The significance of nontoxic thyroid nodules. Final report of a 15- year study of the incidence of thyroid malignancy. Ann Intern Med. 1968;69:537–540. [PubMed: 5673172]
  2. Tunbridge WM, Evered DC, Hall R, Appleton D, Brewis M, Clark F, Evans JG, Young E, Bird T, Smith PA. The spectrum of thyroid disease in a community: the Whickham survey. Clin Endocrinol (Oxf). 1977;7:481–493. [PubMed: 598014]
  3. Tan GH, Gharib H. Thyroid incidentalomas: management approaches to nonpalpable nodules discovered incidentally on thyroid imaging. Ann Intern Med. 1997;126:226–231. [PubMed: 9027275]
  4. Guth S, Theune U, Aberle J, Galach A, Bamberger CM. Very high prevalence of thyroid nodules detected by high frequency (13 MHz) ultrasound examination. Eur J Clin Invest. 2009;39:699–706. [PubMed: 19601965]
  5. Dean DS, Gharib H. Epidemiology of thyroid nodules. Best Pract Res Clin Endocrinol Metab. 2008;22:901–911. [PubMed: 19041821]
  6. Kwong N, Medici M, Angell TE, Liu X, Marqusee E, Cibas ES, Krane JF, Barletta JA, Kim MI, Larsen PR, Alexander EK. The Influence of Patient Age on Thyroid Nodule Formation, Multinodularity, and Thyroid Cancer Risk. J Clin Endocrinol Metab. 2015 Dec;100(12):4434–40. [PubMed: 26465395]
  7. Mazzaferri E. Management of a solitary thyroid nodule. N Engl J Med. 1993;328:553–559. [PubMed: 8426623]
  8. Ezzat S, Sarti DA, Cain DR, et al. Thyroid incidentalomas prevalence by palpation and ultrasonography. Arch Intern Med. 1994;154:1838–1840. [PubMed: 8053752]
  9. Dauksiene D, Petkeviciene J, Klumbiene J, Verkauskiene R, Vainikonyte-Kristapone J, Seibokaite A, Ceponis J, Sidlauskas V, Daugintyte-Petrusiene L, Norkus A, Zilaitiene B. Factors Associated with the Prevalence of Thyroid Nodules and Goiter in Middle-Aged Euthyroid Subjects. Int J Endocrinol. 2017;2017:8401518. [PubMed: 28356911]
  10. Diez JL. Goiter in adult patients aged 55 years and older: etiology and clinical features in 634 patients. J Gerontol A Biol Sci Med Sci. 2005;60:920–923. [PubMed: 16079218]
  11. Schlumberger M, Filetti S, Hay I. Nontoxic Diffuse and Nodular Goiter and Thyroid Neoplasia. In: Melmed S, Williams RH, editors. Williams Textbook of Endocrinology. 12th ed. Philadelphia: Elsevier/Saunders; 2011. p 440-449.
  12. Haugen BR, Alexander EK, Bible KC, Doherty GM, Mandel SJ, Nikiforov YE, Pacini F, Randolph GW, Sawka AM, Schlumberger M, Schuff KG, Sherman SI, Sosa JA, Steward DL, Tuttle RM, Wartofsky L. 2015 American Thyroid Association Management Guidelines for Adult Patients with Thyroid Nodules and Differentiated Thyroid Cancer: The American Thyroid Association Guidelines Task Force on Thyroid Nodules and Differentiated Thyroid Cancer. Thyroid. 2016 Jan;26(1):1–133. [PubMed: 26462967]
  13. Tessler FN, Middleton WD, Grant EG, Hoang JK, Berland LL, Teefey SA, Cronan JJ, Beland MD, Desser TS, Frates MC, Hammers LW, Hamper UM, Langer JE, Reading CC, Scoutt LM, Stavros AT. ACR Thyroid Imaging, Reporting and Data System (TI-RADS): White Paper of the ACR TI-RADS Committee. J Am Coll Radiol. 2017;14(5):587–595. [PubMed: 28372962]
  14. Russ G, Bonnema SJ, Erdogan MF, Durante C, Ngu R, Leenhardt L. European Thyroid Association Guidelines for Ultrasound Malignancy Risk Stratification of Thyroid Nodules in Adults: The EU-TIRADS. Eur Thyroid J. 2017;6(5):225–237. [PubMed: 29167761]
  15. Shin JH, Baek JH, Chung J, Ha EJ, Kim JH, Lee YH, Lim HK, Moon WJ, Na DG, Park JS, Choi YJ, Hahn SY, Jeon SJ, Jung SL, Kim DW, Kim EK, Kwak JY, Lee CY, Lee HJ, Lee JH, Lee JH, Lee KH, Park SW, Sung JY, Korean Society of Thyroid R, Korean Society of R. Ultrasonography Diagnosis and Imaging-Based Management of Thyroid Nodules: Revised Korean Society of Thyroid Radiology Consensus Statement and Recommendations. Korean J Radiol. 2016;17(3):370–395. [PubMed: 27134526]
  16. Kim PH, Suh CH, Baek JH, Chung SR, Choi YJ, Lee JH. Unnecessary thyroid nodule biopsy rates under four ultrasound risk stratification systems: a systematic review and meta-analysis. Eur Radiol. 2021 May;31(5):2877-2885. [PubMed: 33057762]
  17. Cibas ES, Ali SZ. The 2017 Bethesda System for Reporting Thyroid Cytopathology. Thyroid. 2017 Nov;27(11):1341–1346. [PubMed: 29091573]
  18. Livhits MJ, Zhu CY, Kuo EJ, Nguyen DT, Kim J, Tseng CH, Leung AM, Rao J, Levin M, Douek ML, Beckett KR, Cheung DS, Gofnung YA, Smooke-Praw S, Yeh MW. Effectiveness of Molecular Testing Techniques for Diagnosis of Indeterminate Thyroid Nodules: A Randomized Clinical Trial. JAMA Oncol. 2021 Jan 1;7(1):70-77. [PubMed: 33300952]
  19. Nikiforov YE, Carty SE, Chiosea SI, et al. Highly accurate diagnosis of cancer in thyroid nodules with follicular neoplasm/suspicious for a follicular neoplasm cytology by ThyroSeq v2 next-generation sequencing assay. Cancer. 2014;120:3627–34. [PubMed: 25209362]
  20. Nikiforova MN, Mercurio S, Wald AI, Barbi de Moura M, Callenberg K, Santana-Santos L, Gooding WE, Yip L, Ferris RL, Nikiforov YE. Analytical performance of the ThyroSeq v3 genomic classifier for cancer diagnosis in thyroid nodules. Cancer. 2018 Apr 15;124(8):1682–1690. [PubMed: 29345728]
  21. Alexander EK, Kennedy GC, Baloch ZW, et al. Preoperative diagnosis of benign thyroid nodules with indeterminate cytology. N Engl J Med. 2012;367:705–15. [PubMed: 22731672]
  22. Patel KN, Angell TE, Babiarz J, Barth NM, Blevins T, Duh QY, Ghossein RA, Harrell RM, Huang J, Kennedy GC, Kim SY, Kloos RT. LiVolsi VA, Randolph GW, Sadow PM, Shanik MH, Sosa JA, Traweek ST, Walsh PS, Whitney D, Yeh MW, Ladenson PW. Performance of a Genomic Sequencing Classifier for the Preoperative Diagnosis of Cytologically Indeterminate Thyroid Nodules. JAMA Surg. 2018 Sep 1;153(9):817–824. [PubMed: 29799911]
  23. Labourier E, Shifrin A, Busseniers AE, Lupo MA, Manganelli ML, Andruss B, Wylie D, Beaudenon-Huibregtse S. Molecular Testing for miRNA, mRNA, and DNA on Fine-Needle Aspiration Improves the Preoperative Diagnosis of Thyroid Nodules With Indeterminate Cytology. J Clin Endocrinol Metab. 2015 Jul;100(7):2743–50. [PubMed: 25965083]
  24. Wang Z, Vyas CM, Van Benschoten O, Nehs MA, Moore FD Jr, Marqusee E, Krane JF, Kim MI, Heller HT, Gawande AA, Frates MC, Doubilet PM, Doherty GM, Cho NL, Cibas ES, Benson CB, Barletta JA, Zavacki AM, Larsen PR, Alexander EK, Angell TE. Quantitative Analysis of the Benefits and Risk of Thyroid Nodule Evaluation in Patients ≥70 Years Old. Thyroid. 2018 Apr;28(4):465–471. [PubMed: 29608439]
  25. Smith SA, Hay ID, Goellner JR, et al. Mortality from papillary thyroid carcinoma: a case-control study of 56 lethal cases. Cancer. 1988;62:1381–1388. [PubMed: 3416277]
  26. Tollefsen HR, DeCosse JJ, Hutter RVP. Papillary carcinoma of the thyroid: a clinical and pathological study of 70 fatal cases. Cancer. 1964;17:1035–1044. [PubMed: 14202591]
  27. Noone AM, Howlader N, Krapcho M, Miller D, Brest A, Yu M, Ruhl J, Tatalovich Z, Mariotto A, Lewis DR, Chen HS, Feuer EJ, Cronin KA (eds). SEER Cancer Statistics Review, 1975-2015, National Cancer Institute. Bethesda, MD, https://seer​.cancer.gov/csr/1975_2015/, based on November 2017 SEER data submission, posted to the SEER web site, April 2018.
  28. Girardi FM. Thyroid Carcinoma Pattern Presentation According to Age. Int Arch Otorhinolaryngol. 2017 Jan;21(1):38–41. [PubMed: 28050206]
  29. Davies L, Welch HG. Increasing incidence of thyroid cancer in the United States, 1973-2002. JAMA. 2006;295:2164–2167. [PubMed: 16684987]
  30. Lim H, Devesa SS, Sosa JA, Check D, Kitahara CM. Trends in Thyroid Cancer Incidence and Mortality in the United States, 1974-2013. JAMA. 2017 Apr 4;317(13):1338–1348. [PubMed: 28362912]
  31. Lebastchi AH, Callender GG. Thyroid cancer. Curr Probl Cancer. 2014 Mar-Apr;38(2):48–74. [PubMed: 24951026]
  32. Cabanillas ME, McFadden DG, Durante C. Thyroid cancer. Lancet. 2016 Dec 3;388(10061):2783–2795. [PubMed: 27240885]
  33. Gupta KL. Neoplasm of the thyroid gland. Clin Geriatr Med. 1995;11:271–290. [PubMed: 7606696]
  34. Sarne D, Schneider AB. External radiation and thyroid neoplasia. Endocrinol Metab Clin North Am. 1996;25(1):181–195. [PubMed: 8907686]
  35. Ron E, Kleinerman RA, Boice JD Jr. LiVolsi VA, Flannery JT, Fraumeni JF Jr. A population-based case-control study of thyroid cancer. J Natl Cancer Inst. 1987;79(1):1–12. [PubMed: 3474436]
  36. Ron E, Lubin JH, Shore RE, et al. Thyroid cancer after exposure to external radiation: a pooled analysis of seven studies. 1995. Radiat Res. 2012;178(2):AV43–AV60. [PubMed: 22870979]
  37. Lee YS , Lim H , Chang HS , Park CS. Papillary thyroid microcarcinomas are different from latent papillary thyroid carcinomas at autopsy. J Korean Med Sci. 2014;29:676–679.  [PubMed: 24851024]
  38. Ito Y, Miyauchi A, Kihara M, Higashiyama T, Kobayashi K, Miya A.  Patient age is significantly related to the progression of papillary microcarcinoma of the thyroid under observation. Thyroid. 2014;24:27–34.  [PubMed: 24001104]
  39. Grebe SK, Hay ID. Follicular thyroid cancer. Endocrinol Metab Clin North Am. 1995;24:761. [PubMed: 8608779]
  40. Brennan MD, Bergstralh EJ, Van Heerden JA, et al. Follicular thyroid cancer treated at the Mayo Clinic, 1946 through 1970: initial manifestations, pathologic findings, therapy, and outcome. Mayo Clin Proc. 1991;66:11–22. [PubMed: 1988751]
  41. Grani G, Lamartina L, Durante C, Filetti S, Cooper DS. Follicular thyroid cancer and Hürthle cell carcinoma: challenges in diagnosis, treatment, and clinical management. Lancet Diabetes Endocrinol. 2018 Jun;6(6):500–514. [PubMed: 29102432]
  42. Xing M. Molecular pathogenesis and mechanisms of thyroid cancer. Nat Rev Cancer. 2013 Mar;13(3):184–99. [PubMed: 23429735]
  43. Hundahl SA, Fleming ID, Fremgen AM, Menck HR. A National Cancer Data Base report on 53,856 cases of thyroid carcinoma treated in the U.S., 1985-1995. Cancer. 1998 Dec 15;83(12):2638–48. [PubMed: 9874472]
  44. Máximo V, Lima J, Prazeres H, Soares P, Sobrinho-Simões M. The biology and the genetics of Hürthle cell tumors of the thyroid. Endocr Relat Cancer. 2016 Dec;23(12):X2. [PubMed: 27807063]
  45. Ganly I, Makarov V, Deraje S, Dong Y, Reznik E, Seshan V, Nanjangud G, Eng S, Bose P, Kuo F, Morris LGT, Landa I, Carrillo Albornoz PB, Riaz N, Nikiforov YE, Patel K, Umbricht C, Zeiger M, Kebebew E, Sherman E, Ghossein R, Fagin JA, Chan TA. Integrated Genomic Analysis of Hürthle Cell Cancer Reveals Oncogenic Drivers, Recurrent Mitochondrial Mutations, and Unique Chromosomal Landscapes. Cancer Cell. 2018 Aug 13;34(2):256–270.e5. [PubMed: 30107176]
  46. Gopal RK, Kübler K, Calvo SE, Polak P, Livitz D, Rosebrock D, Sadow PM, Campbell B, Donovan SE, Amin S, Gigliotti BJ, Grabarek Z, Hess JM, Stewart C, Braunstein LZ, Arndt PF, Mordecai S, Shih AR, Chaves F, Zhan T, Lubitz CC, Kim J, Iafrate AJ, Wirth L, Parangi S, Leshchiner I, Daniels GH, Mootha VK, Dias-Santagata D, Getz G, McFadden DG. Widespread Chromosomal Losses and Mitochondrial DNA Alterations as Genetic Drivers in Hürthle Cell Carcinoma. Cancer Cell. 2018 Aug 13;34(2):242–255.e5. [PubMed: 30107175]
  47. Lin JD, Chao TC, Chen ST, et al. Characteristics of thyroid carcinomas in aging patients. Eur J Clin Invest. 2000;30:147–153. [PubMed: 10651840]
  48. Chereau N, Trésallet C, Noullet S, Godiris-Petit G, Tissier F, Leenhardt L, Menegaux F. Prognosis of papillary thyroid carcinoma in elderly patients after thyroid resection: A retrospective cohort analysis. Medicine (Baltimore). 2016 Nov;95(47):e5450. [PubMed: 27893690]
  49. Lerch H, Schober O, Kuwert T, et al. Survival of differentiated thyroid carcinoma studied in 500 patients. J Clin Onc. 1997;15:2067–2075. [PubMed: 9164219]
  50. Shi LY, Liu J, Yu LJ, Lei YM, Leng SX, Zhang HY. Clinic-pathologic Features and Prognostic Analysis of Thyroid Cancer in the Older Adult: A SEER Based Study. J Cancer. 2018 Jul 1;9(15):2744–2750. [PubMed: 30087716]
  51. Haymart MR. Understanding the relationship between age and thyroid cancer. Oncologist. 2009 Mar;14(3):216–21. [PubMed: 19270027]
  52. Adam MA, Pura J, Goffredo P, Dinan MA, Reed SD, Scheri RP, Hyslop T, Roman SA, Sosa JA. Presence and Number of Lymph Node Metastases Are Associated With Compromised Survival for Patients Younger Than Age 45 Years With Papillary Thyroid Cancer. J Clin Oncol. 2015 Jul 20;33(21):2370–5. [PubMed: 26077238]
  53. Orosco RK, Hussain T, Brumund KT, Oh DK, Chang DC, Bouvet M. Analysis of age and disease status as predictors of thyroid cancer-specific mortality using the Surveillance, Epidemiology, and End Results database. Thyroid. 2015 Jan;25(1):125–32. [PubMed: 25369076]
  54. Ganly I, Nixon IJ, Wang LY, Palmer FL, Migliacci JC, Aniss A, Sywak M, Eskander AE, Freeman JL, Campbell MJ, Shen WT, Vaisman F, Momesso D, Corbo R, Vaisman M, Shaha A, Tuttle RM, Shah JP, Patel SG. Survival from Differentiated Thyroid Cancer: What Has Age Got to Do with It? Thyroid. 2015 Oct;25(10):1106–14. [PubMed: 26148759]
  55. Brierley, J.D.; Gospodarowicz, M.K.; Wittekind, C. TNM Classification of Malignant Tumours, 8th ed.; John Wiley & Sons: Weinheim, Germany, 2017; pp. 69–71.
  56. Hay ID, Bergstralh EJ, Goellner JR, Ebersold JR, Grant CS. Predicting outcome in papillary thyroid carcinoma: development of a reliable prognostic scoring system in a cohort of 1779 patients surgically treated at one institution during 1940 through 1989. Surgery. 1993 Dec;114(6):1050–7. [PubMed: 8256208]
  57. Davis NL, Bugis SP, McGregor GI, Germann E. An evaluation of prognostic scoring systems in patients with follicular thyroid cancer. Am J Surg. 1995 Nov;170(5):476–80. [PubMed: 7485736]
  58. Halnan KE. Influence of age and sex on incidence and prognosis of thyroid cancer. Cancer. 1966;19:1534–1536. [PubMed: 5925260]
  59. Cady B, Sedgwick CE, Meissner WA, et al. Risk factor analysis in differentiated thyroid cancer. Cancer. 1979;43:810–820. [PubMed: 427722]
  60. Ito Y, Miyauchi A, Jikuzono T, Higashiyama T, Takamura Y, Miya A, Kobayashi K, Matsuzuka F, Ichihara K, Kuma K. Risk factors contributing to a poor prognosis of papillary thyroid carcinoma: validity of UICC/AJCC TNM classification and stage grouping. World J Surg. 2007 Apr;31(4):838–48. [PubMed: 17347900]
  61. Sugino K, Ito K, Nagahama M, Kitagawa W, Shibuya H, Ohkuwa K, Yano Y, Uruno T, Akaishi J, Kameyama K, Ito K. Prognosis and prognostic factors for distant metastases and tumor mortality in follicular thyroid carcinoma. Thyroid. 2011 Jul;21(7):751–7. [PubMed: 21615311]
  62. Voutilainen PE, Multanen MM, Leppaniemi AK, et al. Prognosis after lymph node recurrence in papillary thyroid carcinoma depends on age. Thyroid. 2001;11:953. [PubMed: 11716043]
  63. Zaydfundim V, Feurer ID, Griffin MR, et al. The impact of lymph node involvement on survival in patients with papillary and follicular thyroid carcinoma. Surgery. 2008;144:1070–1077. [PubMed: 19041020]
  64. Ito Y, Kudo T, Takamura Y, Kobayashi K, Miya A, Miyauchi A. Prognostic factors of papillary thyroid carcinoma vary according to sex and patient age. World J Surg. 35:2684-2690, 2011. [PubMed: 21953129]
  65. Ylli D, Burman KD, Van Nostrand D, Wartofsky L. Eliminating the Age Cutoff in Staging of Differentiated Thyroid Cancer: The Safest Road? J Clin Endocrinol Metab. 2018 May 1;103(5):1813–1817. [PubMed: 29741712]
  66. Nixon IJ, Wang LY, Migliacci JC, Eskander A, Campbell MJ, Aniss A, Morris L, Vaisman F, Corbo R, Momesso D, Vaisman M, Carvalho A, Learoyd D, Leslie WD, Nason RW, Kuk D, Wreesmann V, Morris L, Palmer FL, Ganly I, Patel SG, Singh B, Tuttle RM, Shaha AR, Gönen M, Pathak KA, Shen WT, Sywak M, Kowalski L, Freeman J, Perrier N, Shah JP. An International Multi-Institutional Validation of Age 55 Years as a Cutoff for Risk Stratification in the AJCC/UICC Staging System for Well-Differentiated Thyroid Cancer. Thyroid. 2016 Mar;26(3):373–80. [PubMed: 26914539]
  67. Nixon IJ, Kuk D, Wreesmann V, Morris L, Palmer FL, Ganly I, Patel SG, Singh B, Tuttle RM, Shaha AR, Gönen M, Shah JP. Defining a Valid Age Cutoff in Staging of Well-Differentiated Thyroid Cancer. Ann Surg Oncol. 2016 Feb;23(2):410–5. [PubMed: 26215199]
  68. Lechner MG, Bernardo AC, Lampe A, Praw SS, Tam SH, Angell TE. Changes in Stage Distribution and Disease-Specific Survival in Differentiated Thyroid Cancer with Transition to American Joint Committee on Cancer 8th Edition: A Systematic Review and Meta-Analysis. Oncologist. 2021 Feb;26(2):e251-e260. [PubMed: 32864832]
  69. Papaleontiou M, Norton EC, Reyes-Gastelum D, Banerjee M, Haymart MR. Competing Causes of Death in Older Adults with Thyroid Cancer. Thyroid. 2021 Sep;31(9):1359-1365. [PubMed: 33764188]
  70. McLeod DS, Carruthers K, Kevat DA. Optimal differentiated thyroid cancer management in the elderly. Drugs Aging. 2015 Apr;32(4):283–94. [PubMed: 25825123]
  71. Randolph GW, Doherty GM. Carcinoma of the follicular thyroid: surgical therapy. In: Braverman LE, Cooper DS, editors. Werner & Ingbar’s the thyroid: a fundamental and clinical text. 10th ed. Philadelphia: Lippincott Williams & Wilkins; 2013. p. 702–11.
  72. Bliss R, Patel N, Guinea A, et al. Age is no contraindication to thyroid surgery. Age Ageing. 1999;28:363–6. [PubMed: 10459789]
  73. Passler C, Avanessian R, Kaczirek K, et al. Thyroid surgery in the geriatric patient. Arch Surg. 2002;137:1243–8. [PubMed: 12413310]
  74. Seybt MW, Khichi S, Terris DJ. Geriatric thyroidectomy: safety of thyroid surgery in an aging population. Arch Otolaryngol Head Neck Surg. 2009;135:1041–4. [PubMed: 19841346]
  75. Raffaelli M, Bellantone R, Princi P, et al. Surgical treatment of thyroid diseases in elderly patients. Am J Surg. 2010;200:467–72. [PubMed: 20887839]
  76. Sosa JA, Mehta PJ, Wang TS, et al. A population-based study of outcomes from thyroidectomy in aging Americans: at what cost? J Am Coll Surg. 2008;206:1097–105. [PubMed: 18501806]
  77. Tuggle CT, Park LS, Roman S, et al. Rehospitalization among elderly patients with thyroid cancer after thyroidectomy are prevalent and costly. Ann Surg Oncol. 2010;17:2816–23. [PubMed: 20552406]
  78. Kandil E, Krishnan B, Noureldine SI, Yao L, Tufano RP. Hemithyroidectomy: a meta-analysis of postoperative need for hormone replacement and complications. ORL J Otorhinolaryngol Relat Spec. 2013;75(1):6-17. [Pubmed: 23486083].
  79. Schumm MA, Lechner MG, Shu ML, Ochoa JE, Kim J, Tseng CH, Leung AM, Yeh MW. Frequency of Thyroid Hormone Replacement After Lobectomy for Differentiated Thyroid Cancer. Endocr Pract. 2021 Jul;27(7):691-697. [PubMed: 33642257]
  80. Tuttle RM, Zhang L, Shaha A. A clinical framework to facilitate selection of patients with differentiated thyroid cancer for active surveillance or less aggressive initial surgical management. Expert Rev Endocrinol Metab. 2018 Mar;13(2):77-85. [PubMed: 30058863]
  81. Sugitani I, Fujimoto Y, Yamada K. Association between serum thyrotropin concentration and growth of asymptomatic papillary thyroid microcarcinoma. World J Surg. 2014 Mar;38(3):673-8. [Pubmed: 24233662].
  82. Tuttle RM, Fagin JA, Minkowitz G, Wong RJ, Roman B, Patel S, Untch B, Ganly I, Shaha AR, Shah JP, Pace M, Li D, Bach A, Lin O, Whiting A, Ghossein R, Landa I, Sabra M, Boucai L, Fish S, Morris LGT. Natural History and Tumor Volume Kinetics of Papillary Thyroid Cancers During Active Surveillance. JAMA Otolaryngol Head Neck Surg. 2017 Oct 1;143(10):1015-1020. [Pubmed: 28859191]
  83. Tufano RP, Shindo M, Shaha AR. New Recommendations for Extent of Thyroidectomy and Active Surveillance for the Treatment of Differentiated Thyroid Cancer. JAMA Otolaryngol Head Neck Surg. 2016 Jul 1;142(7):625-6. [PubMed: 27196622].
  84. Ruel E , Thomas S , Dinan M , Perkins JM , Roman SA , Sosa JA. Adjuvant radioactive iodine therapy is associated with improved survival for patients with intermediate-risk papillary thyroid cancer. J Clin Endocrinol Metab. 2015;100:1529–1536.  [PubMed: 25642591]
  85. Schlumberger M, Catargi B, Borget I, et al. .Strategies of radioiodine ablation in patients with low-risk thyroid cancer. N Engl J Med. 2012;366:1663–1673 [PubMed: 22551127]
  86. Mallick U, Harmer C, Yap B, et al. Ablation with low-dose radioiodine and thyrotropin alfa in thyroid cancer. N Engl J Med. 2012;366:1674–1685. [PubMed: 22551128]
  87. Schlumberger M, Leboulleux S, Catargi B, et al. Outcome after ablation in patients with low-risk thyroid cancer (ESTIMABL1): 5-year follow-up results of a randomised, phase 3, equivalence trial. Lancet Diabetes Endocrinol. 2018;6:618–626. [PubMed: 29807824]
  88. Andresen NS, Buatti JM, Tewfik HH, Pagedar NA, Anderson CM, Watkins JM. Radioiodine Ablation following Thyroidectomy for Differentiated Thyroid Cancer: Literature Review of Utility, Dose, and Toxicity. Eur Thyroid J. 2017 Jul;6(4):187–196. [PubMed: 28868259]
  89. Flynn RW, Bonellie SR, Jung RT, et al. Serum thyroid stimulating hormone concentration and morbidity from cardiovascular disease and fractures in patients on long-term thyroxine therapy. J Clin Endocrinol Metab. 2010;95:186–93. [PubMed: 19906785]
  90. Bauer DC, Ettinger B, Nevitt MC, et al. Risk for fracture in women with low serum levels of thyroid-stimulating hormone. Ann Intern Med. 2001;134:561–8. [PubMed: 12803168]
  91. Kung AW, Yeung SS. Prevention of bone loss induced by thyroxine suppressive therapy in postmenopausal women: the effect of calcium and calcitonin. J Clin Endocrinol Metab. 1996;81:1232–6. [PubMed: 8772604]
  92. Sugitani I, Fujimoto Y. Effect of postoperative thyrotropin suppressive therapy on bone mineral density in patients with papillary thyroid carcinoma: a prospective controlled study. Surgery. 2011;150:1250–7. [PubMed: 22136848]
  93. Wang LY, Smith AW, Palmer FL, Tuttle RM, Mahrous A, Nixon IJ, Patel SG, Ganly I, Fagin JA, Boucai L. Thyrotropin suppression increases the risk of osteoporosis without decreasing recurrence in ATA low- and intermediate-risk patients with differentiated thyroid carcinoma. Thyroid. 2015 Mar;25(3):300–7. [PubMed: 25386760]
  94. Cosman F, de Beur SJ, LeBoff MS, Lewiecki EM, Tanner B, Randall S, Lindsay R., National Osteoporosis Foundation. Clinician's Guide to Prevention and Treatment of Osteoporosis. Osteoporos Int. 2014 Oct;25(10):2359–81. [PubMed: 25182228]
  95. Gregerman RI, Gaffney GW, Shock NW, et al. Thyroxine turnover in euthyroid man with special reference to changes with age. J Clin Invest. 1962;41:2065–74. [PubMed: 13950233]
  96. Rosenbaum RL, Barzel US. Levothyroxine replacement dose for primary hypothyroidism decreases with age. Ann Intern Med. 1982;96:53–5. [PubMed: 7053703]
  97. Singh N, Weisler SL, Hershman JM. The acute effect of calcium carbonate on the intestinal absorption of levothyroxine. Thyroid. 2001 Oct;11(10):967–71. [PubMed: 11716045]
  98. Kim M, Ladenson P. Thyroid. In: Goldman L, Schafer AI, editors. Goldman’s Cecil medicine. 24th ed. Philadelphia: Elsevier Saunders; 2012. p 1450–63.
  99. Soroushyari A, Do D, Langton J, et al. Partial withdrawal of levothyroxine to stimulate serum thyroglobulin for thyroid cancer monitoring. Thyroid. 2004;14:1105–1107. [PubMed: 15666427]
  100. Spencer C, Fatemi S. Thyroglobulin antibody (TgAb) methods - Strengths, pitfalls and clinical utility for monitoring TgAb-positive patients with differentiated thyroid cancer. Best Pract Res Clin Endocrinol Metab. 2013 Oct;27(5):701–12. [PubMed: 24094640]
  101. Pedrazzini L, Baroli A, Lomuscio G, Marzoli L. Prevalence, clinical significance and prognostic value of anti-thyroglobulin antibodies in the follow-up of patients with differentiated thyroid carcinoma: a retrospective study. Minerva Endocrinol. 2009 Sep;34(3):195-203. [PubMed: 19859043].
  102. Momesso DP, Vaisman F, Yang SP, Bulzico DA, Corbo R, Vaisman M, Tuttle RM. Dynamic Risk Stratification in Patients with Differentiated Thyroid Cancer Treated Without Radioactive Iodine. J Clin Endocrinol Metab. 2016 Jul;101(7):2692-700. [Pubmed: 27023446]
  103. Brose MS, Nutting CM, Jarzab B, et al. Sorafenib in radioactive iodine-refractory, locally advanced or metastatic differentiated thyroid cancer: a randomised, double-blind, phase 3 trial. Lancet. 2014;384:319–328. [PubMed: 24768112]
  104. Schlumberger M, Tahara M, Wirth LJ, et al. Lenvatinib versus placebo in radioiodine-refractory thyroid cancer. N Engl J Med. 372:621-30, 2015. [PubMed: 25671254]
  105. Brose MS, Robinson B, Sherman SI, Krajewska J, Lin CC, Vaisman F, Hoff AO, Hitre E, Bowles DW, Hernando J, Faoro L, Banerjee K, Oliver JW, Keam B, Capdevila J. Cabozantinib for radioiodine-refractory differentiated thyroid cancer (COSMIC-311): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol. 2021 Aug;22(8):1126-1138. [PubMed: 34237250].
  106. Wirth LJ, Sherman E, Robinson B, Solomon B, Kang H, Lorch J, Worden F, Brose M, Patel J, Leboulleux S, Godbert Y, Barlesi F, Morris JC, Owonikoko TK, Tan DSW, Gautschi O, Weiss J, de la Fouchardière C, Burkard ME, Laskin J, Taylor MH, Kroiss M, Medioni J, Goldman JW, Bauer TM, Levy B, Zhu VW, Lakhani N, Moreno V, Ebata K, Nguyen M, Heirich D, Zhu EY, Huang X, Yang L, Kherani J, Rothenberg SM, Drilon A, Subbiah V, Shah MH, Cabanillas ME. Efficacy of Selpercatinib in RET-Altered Thyroid Cancers. N Engl J Med. 2020 Aug 27;383(9):825-835. doi: 10.1056/NEJMoa2005651. [PubMed: 32846061]
  107. Sherman EJ, Wirth LJ, Shah MH, et al. Selpercatinib efficacy and safety in patients with RET-altered thyroid cancer: A clinical trial update. Journal of Clinical Oncology 2021 39:15_suppl, 6073-6073
  108. Subbiah V, Hu MI, Wirth LJ, Schuler M, Mansfield AS, Curigliano G, Brose MS, Zhu VW, Leboulleux S, Bowles DW, Baik CS, Adkins D, Keam B, Matos I, Garralda E, Gainor JF, Lopes G, Lin CC, Godbert Y, Sarker D, Miller SG, Clifford C, Zhang H, Turner CD, Taylor MH. Pralsetinib for patients with advanced or metastatic RET-altered thyroid cancer (ARROW): a multi-cohort, open-label, registrational, phase 1/2 study. Lancet Diabetes Endocrinol. 2021 Aug;9(8):491-501. doi: 10.1016/S2213-8587(21)00120-0. [PubMed: 34118198].
  109. Hong DS, DuBois SG, Kummar S, Farago AF, Albert CM, Rohrberg KS, et al. . Larotrectinib in Patients With TRK Fusion-Positive Solid Tumours: A Pooled Analysis of Three Phase 1/2 Clinical Trials. Lancet Oncol (2020) 21(4):531–40. 10.1016/S1470-2045(19)30856-3;
  110. Doebele RC, Drilon A, Paz-Ares L, Siena S, Shaw AT, Farago AF, et al. . Entrectinib in Patients With Advanced or Metastatic NTRK Fusion-Positive Solid Tumours: Integrated Analysis of Three Phase 1-2 Trials. Lancet Oncol (2020) 21(2):271–82. 10.1016/S1470-2045(19)30691-6)
  111. Chakravarty D, Santos E, Ryder M, Knauf JA, Liao XH, West BL, Bollag G, Kolesnick R, Thin TH, Rosen N, Zanzonico P, Larson SM, Refetoff S, Ghossein R, Fagin JA. Small-molecule MAPK inhibitors restore radioiodine incorporation in mouse thyroid cancers with conditional BRAF activation. J Clin Invest. 2011 Dec;121(12):4700-11. [PubMed: 22105174]
  112. Ho AL, Grewal RK, Leboeuf R, Sherman EJ, Pfister DG, Deandreis D, Pentlow KS, Zanzonico PB, Haque S, Gavane S, Ghossein RA, Ricarte-Filho JC, Domínguez JM, Shen R, Tuttle RM, Larson SM, Fagin JA. Selumetinib-enhanced radioiodine uptake in advanced thyroid cancer. N Engl J Med. 2013 Feb 14;368(7):623-32. doi: 10.1056/NEJMoa1209288. [PubMed: 23406027]
  113. Ho A, Dedecjus M, Wirth LJ, et al. ASTRA: a phase III, randomized, placebo-controlled study evaluating complete remission rate (CRR) with short-course selumetinib plus adjuvant radioactive iodine (RAI) in patients (pts) with differentiated thyroid cancer (DTC). Presented at: the 88 Annual Meeting of the American Thyroid Association; Washington, D.C.: October 3-7, 2018. Abstract short call oral 8.
  114. Buffet C, Wassermann J, Hecht F, Leenhardt L, Dupuy C, Groussin L, Lussey-Lepoutre C. Redifferentiation of radioiodine-refractory thyroid cancers. Endocr Relat Cancer. 2020 May;27(5):R113-R132. [Pubmed: 32191916].
  115. Rothenberg SM, McFadden DG, Palmer EL, Daniels GH, Wirth LJ. Redifferentiation of iodine-refractory BRAF V600E-mutant metastatic papillary thyroid cancer with dabrafenib. Clin Cancer Res. 2015 Mar 1;21(5):1028-35. [PubMed: 25549723].
  116. Dunn LA, Sherman EJ, Baxi SS, Tchekmedyian V, Grewal RK, Larson SM, Pentlow KS, Haque S, Tuttle RM, Sabra MM, Fish S, Boucai L, Walters J, Ghossein RA, Seshan VE, Ni A, Li D, Knauf JA, Pfister DG, Fagin JA, Ho AL. Vemurafenib Redifferentiation of BRAF Mutant, RAI-Refractory Thyroid Cancers. J Clin Endocrinol Metab. 2019 May 1;104(5):1417-1428. [PubMed: 30256977]
  117. Kebebew E, Greenspan FS, Clark OH, et al. Anaplastic thyroid carcinoma treatment outcome and prognostic factors. Cancer. 2005;103:1330–1335. [PubMed: 15739211]
  118. Aldinger KA, Samaan NA, Ibanez M, et al. Anaplastic carcinoma of the thyroid a review of 84 cases of spindle and giant cell carcinoma of the thyroid. Cancer. 1978;41:2267–2275. [PubMed: 657091]
  119. Schwartz DL, Lobo ML, Ang KK, et al. Postoperative external beam radiotherapy for differentiated thyroid cancer: outcomes and morbidity with conformal treatment. Int J Radiat Oncol Biol Phys. 2009;75:1–9. [PubMed: 19095376]
  120. Rao SN, Cabanillas ME. Navigating systemic therapy in advanced thyroid carcinoma: From standard care to personalized therapy and beyond. J Endocr Soc. 2018 Oct 1;2(10):1109–1130. [PubMed: 30250937]
  121. Kebebew E, Ituarte PHG, Siperstein AE, et al. Medullary thyroid carcinoma clinical characteristics, treatment, prognostic factors, and a comparison of staging systems. Cancer. 2000;88:1139–1147. [PubMed]
  122. Saad MF, Ordonez NG, Rashid RK, et al. Medullary carcinoma of the thyroid: a study of the clinical features and prognostic factors in 161 patients. Medicine (Baltimore). 1984;63:319. [PubMed]
  123. Scopsi L, Sampietro G, Boracchi P, et al. Multivariate analysis of prognostic factors in sporadic medullary carcinoma of the thyroid: a retrospective study of 109 consecutive patients. Cancer. 1996;78:2173–2183. [PubMed]
  124. deGroot JW, Plukker JT, Wolffenbuttel BH, et al. Determinants of life expectancy in medullary thyroid cancer: age does not matter. Clin Endocrinol (Oxf). 2006;65:729. [PubMed]
  125. Machens A, Hauptmann S, Dralle H. Increased risk of lymph node metastasis in multifocal hereditary and sporadic medullary thyroid cancer. World J Surg. 2007;31:1960–1965. [PubMed]
  126. Roy M, Chen H, Sippel RS. Current understanding and management of medullary thyroid cancer. The Oncologist. 2013;18:1093–1100. [PubMed]
  127. Wells SA, Robinson BG, Gagel RF, et al. Vandetanib in patients with locally advanced or metastatic medullary thyroid cancer: a randomized, double blind phase III trial. J Clin Oncol. 2012;30:134. [PubMed]
  128. Elisei R, Schlumberger MJ, Müller SP, Schöffski P, Brose MS, Shah MH, Licitra L, Jarzab B, Medvedev V, Kreissl MC, Niederle B, Cohen EE, Wirth LJ, Ali H, Hessel C, Yaron Y, Ball D, Nelkin B, Sherman SI. Cabozantinib in progressive medullary thyroid cancer. J Clin Oncol. 2013 Oct 10;31(29):3639–46. [PubMed]

 

Changing the Course of Disease in Type 1 Diabetes

ABSTRACT

 

In the U.S. alone, more than one million people are living with type 1 diabetes (TID) and approximately 80 people per day, or 30,000 individuals per year, are newly diagnosed (1, 2). Recent epidemiological studies demonstrate that the global T1D incidence is increasing at a rate of approximately 3-4% per year, notably among younger children (3, 4). Despite improvements in insulins, insulin delivery methods, and home glucose monitoring, the vast majority of those with T1D do not achieve recommended levels of glycemic control.  This is particularly true in childhood and adolescence, where a recent U.S. study reported mean HbA1c values exceeding 9.5%, and a high frequency of both DKA and severe hypoglycemia (5). In addition to the increased risk of morbidity and mortality, TID places significant emotional and financial burdens on individuals, families, and society. These realities highlight the need for both better TID therapies and the continued push towards the prevention of TID. In recent decades, research efforts have described the natural history of type 1 diabetes and expanded the ability to identify individuals at risk for the disease even before clinical onset, via the recognition of genetic markers or TID-specific autoantibodies. The increasing ability to identify the at-risk population affords researchers the opportunity to intervene at progressively earlier stages in the disease.  With the understanding that established islet autoimmunity, confirmed by the presence of multiple T1D autoantibodies, inevitably leads to clinical TID, investigative efforts are shifting towards the prevention or modification of autoimmunity.  Furthermore, with the mounting evidence that any amount of residual C-peptide improves long term clinical outcomes in TID, some therapies aim to preserve remaining beta cell function in those with clinical disease. In this chapter, we review the epidemiology of TID, genetic and environmental risk factors, the scientific underpinnings of previous and current approaches towards disease-modifying therapy, and future directions of clinical trials. 

 

EPIDEMIOLOGY OF DIABETES

 

T1D, or autoimmune diabetes, represents 5-10% of diabetes, and like autoimmunity in general, TID is increasing worldwide. The increase likely is attributable to environmental factors or epigenetic changes, as genetic changes don’t occur rapidly enough to explain such a dramatic increase. The SEARCH for Diabetes in Youth Study is a multicenter observational study investigating trends in incidence and prevalence of diabetes in American youth < age 20.  SEARCH data suggests that the prevalence of TID among non-Hispanic white youth is ~1/300 in the US by age 20 years (6). Between 2002 and 2009, the incidence of TID among non-Hispanic white youth < age 20 years increased by an average of 2.7% per year (7). Similarly, the EURODIAB study evaluated TID incidence trends in 17 European countries from 1989-2003 in youth < age 15 years, and found an average annual incidence increase of 3.9%. This trend predicts a 70% increase in TID prevalence between 2005-2020 among European youth < 15 years old (8) with the peak of diagnosis between ages 10-14 (9). While incidence and prevalence are well documented in children, TID occurs in adults as well, at a frequency that is less certain; estimates are that 25-50% of all TID cases are diagnosed in adulthood. The uncertainty likely is due to a less dramatic clinical presentation than is typically seen in children who present with TID. The incidence of TID varies tremendously by geographic location, with higher rates generally seen in countries located farther from the equator. Worldwide incidence data was reported in 2000 by the DIAMOND project (10), a WHO-sponsored effort to address the public health implications of TID. The incidence of TID between 1990 and 1994 in 50 countries is shown in Figure 1. Between 1990 and 1994, the incidence of TID in individuals aged 0-14 years in both Finland and Sardinia was 37/100,000 individuals, whereas the incidence in both China and Venezuela was 0.1/100,000 individuals, a 350-fold difference. The increased incidence coupled with reduced early mortality has contributed to the increasing prevalence of disease. 

 

Figure 1. Worldwide incidence of TID 1990-1994, used with permission from International Diabetes Federation.

 

 

WHAT IS THE RISK OF TYPE 1 DIABETES?

 

As is true for Cindy, 85% of individuals who develop TID have no family history of TID; nonetheless, a family history of the disease does increase an individual’s relative risk.  The prevalence of TID in the US non-Hispanic white population by age 20 is ~0.3%, as compared with ~5% of those with a relative with TID, a 15-fold increase in relative risk.   This relative risk is depicted in Figure 2.

 

Figure 2. Among 300 people without a family member with diabetes, 1 will have TID. Among 300 people with a family member with diabetes, 15 will have TID

 

The risk of TID among family members varies depending on who the affected family member is, as shown in Table 1.  

 

Table 1. Prevalence of TID in Individuals with a Family History of TID.

Relative with TID

Prevalence at age 20

Reference

Mother

2%

(11, 12)

Father

6%

(11, 12)

Non-twin sibling

6%

(13)

Dizygotic (fraternal) twin

10%

(13, 14)

Monozygotic (identical) twin

>50%

(15)

 

The heritability pattern suggests that both genes and environment contribute to risk.  Curiously, the risk of TID in offspring is higher if the father has TID (~6%) as compared to if the mother has TID (~2%) (11, 12). Moreover, the risk to a dizygotic twin is slightly higher (~10%) than is the risk to a non-twin sibling with similar HLA risk genes (~6%) (13, 14)  suggesting that the intrauterine environment and/or similar early life exposures may be important. Lastly, the risk to a monozygotic twin is upwards of ~50%; surprisingly the second twin’s diagnosis may occur many decades after the index twin, highlighting the complexities of gene and environmental interactions that underlie the disease (15).

 

 

 

THE NATURAL HISTORY TYPE 1 DIABETES

 

It is now understood that TID is an immune-mediated disease that begins in the setting of genetic predisposition and then progresses along a predictable path: early islet autoimmunity (one autoantibody), established islet autoimmunity (two or more autoantibodies), abnormal glucose tolerance, clinical TID with some remaining beta cell function, and finally, little or no remaining beta cell function. This understanding comes from decades of effort by multiple investigators and from participation by thousands of patients with TID and their family members.  George Eisenbarth’s description of TID as a chronic autoimmune disease, manifested by autoimmunity and a gradual linear fall in beta cell function until there is insufficient beta cell mass to suppress symptomatic hyperglycemia, has served for decades as the TID natural history paradigm (16). The “Eisenbarth” model has undergone refinements in recent years; namely, although autoimmunity and beta cell dysfunction do appear prior to diagnosis, these changes are often step-wise and non-linear.  Furthermore, beta cell destruction may not be absolute.  Nonetheless, the paradigm is largely correct and serves as the underlying rationale for TID trials. 

 

The long pre-symptomatic natural history of TID presents an opportunity to intervene earlier than is done currently.   Diabetes-specific autoantibodies can appear many years before clinical diagnosis and may reliably be used to predict disease progression.  In 2015, JDRF, the Endocrine Society, and the American Diabetes Association proposed a new TID staging system which underscores that TID begins with islet autoimmunity rather than with symptomatic hyperglycemia (17). Stage 1 TID is defined as the presence of 2 or more autoantibodies with normoglycemia; stage 2 TID is 2 or more autoantibodies, impaired glucose tolerance and no symptoms; stage 3 TID is clinical disease. The staging system is depicted in figure 3.  

Figure 3. New staging classification of Type 1 diabetes. Stages of Type 1 Diabetes. Adapted from internet image. https://beyondtype1.org/clinical-trials-and-the-type-1-diabetes-cure/final-trialnet-stages-of-diabetes-graph-2/ Used with permission.

 

HOW TO DETERMINE RISK OF TID

 

Risk of TID may be determined by the identification of autoantibodies, usually in those identified as having genetic risk through HLA testing or by family history. Autoantibodies are detectable years before the onset of clinical TID. 

 

Determining Risk: Genes

 

With the knowledge that TID runs in families and with advances in technology, investigators have described the genetic risk of TID.  TID risk is strongly linked to HLA class II DR3 and DR4 haplotypes, with the highest risk in those with the DR3/DR4 genotype.  The importance of HLA genes to TID risk highlights the role of the adaptive immune system in the development of autoimmunity.  Newer studies have discovered multiple other genes that also contribute to TID risk (18). They are largely genes known also to impact immune function; however, their contribution is dwarfed by the impact of HLA genes.  Interestingly, recent work suggests that HLA genes primarily contribute to development of autoantibodies, while non-HLA genes and environmental factors may be more important in the progression from autoantibodies to clinically overt disease (19, 20). The description of non-HLA risk genes (such as the genes for insulin, a major TID autoantigen) highlights other potential pathways to disease and potential therapies. 

Although the contribution of HLA class II risk genes overwhelms the contribution of non-HLA risk genes, the HLA contribution may be decreasing as the overall incidence of TID increases.  This suggests that in a population with non-HLA genetic susceptibility, the environment may have become more conducive to the development of TID. This was reported in a 2004 Lancet article by Gillespie, et al., in which the investigators compared the frequency of HLA class II haplotypes in a UK cohort of 194 individuals diagnosed with TID between 1922-1946 (the Golden Years cohort) to a cohort of 582 individuals diagnosed between 1985-2002 (the BOX cohort) (21). In this comparison, shown in Figure 4, 47% of individuals in the Golden Years cohort were positive for the highest risk genotype DR3-DQ2/DR4-DQ8, compared to 35% of individuals in the BOX cohort.

 

Figure 4. Decreased contribution of high-risk HLA haplotypes over time. HLA class II haplotypes in Golden Years and BOX cohorts, adapted from Gillespie et.al Lancet 2004 (21).

 

Determining Risk: Family History and Islet Cell Autoantibodies

 

Natural history studies of relatives such as Diabetes Prevention Trial (DPT-1) and Diabetes TrialNet Pathway to Prevention have helped define the risk of TID in those with a family history of TID.  Since 2000, Diabetes TrialNet has screened over 200,000 relatives of people with TID, aiming to enroll at-risk individuals in prevention trials.  Among relatives of people with TID, ~5% will have at least one of five islet autoantibodies (22). TrialNet screens for islet cell antibodies (ICA), autoantibodies to insulin (IAA or mIAA), antibodies to a tyrosine phosphatase (IA-2; previously ICA512), antibodies to glutamic acid decarboxylase (GAD), and antibodies to a zinc transporter (ZnT8).  With each additional autoantibody, the risk of TID increases predictably. Unsurprisingly, those with islet autoimmunity and abnormal glucose tolerance are at an even further increased risk of symptomatic T1D. The TrialNet strategy to identify islet autoimmunity among relatives of individuals with TID is shown in Figure 5. There are many other screening efforts ongoing outside of TrialNet. (23-25)

 

Figure 5. Diabetes TrialNet process for identifying relatives with islet autoimmunity.

 

Natural history studies have shown not only that islet autoimmunity predicts TID risk, but also that islet autoantibodies usually appear early in life; 64% of babies destined to develop T1D before puberty will have antibodies by age 2 and 95% by age 5 (26). Furthermore, the data from both prospective birth cohort studies (27) and cross-sectional studies(28) (29) (30, 31) is remarkably consistent and suggests that the risk of progression from established autoimmunity to clinical TID is in the range of 40% after 5 years, 70% after 10 years, and 85% after 15 years. This risk over time is depicted in Figure 6. The key understanding from natural history studies is that essentially all individuals with confirmed islet autoimmunity will eventually develop clinical T1D at a rate of 11% per year.

 

 

Figure 6. Established islet autoimmunity inevitably progresses to clinical T1D. Extrapolated data from multiple studies in genetically at-risk individuals; Ziegler et al. JAMA 2013; DPT-1 Study Group Diabetes 1997; Sosenko et al. Diabetes Care 2014; Mahon et al. Pediatric Diabetes 2009

 

Identifying individuals with islet autoimmunity has two potential benefits; namely, the opportunity to monitor closely for disease progression, conferring a reduced risk of morbidity and mortality at the time of TID diagnosis, and the identification of individuals who are eligible for prevention trials.  It is perhaps underappreciated that there is potentially a direct clinical benefit to identifying those with islet autoimmunity.  Individuals with islet autoimmunity followed regularly until clinical diagnosis present with lower HbA1c and experience less DKA than those diagnosed in the community (Table 2) (32-36). For this reason, since 2009, the ADA has recommended that all individuals with a relative with T1D be counseled about the opportunity to be screened for diabetes autoantibodies in the context of a clinical research trial (37).

 

Table 2.  Individuals Diagnosed with T1D While Enrolled in a Clinical Trial have Less Morbidity at the Time of Diagnosis. (32-36)

 

STUDY

HbA1c at time of TID diagnosis

% with DKA at time of TID diagnosis

 

Enrolled in study

Usual care

Enrolled in study

Usual care

SEARCH

 

 

 

25.5%

BABYDIAB

8.6%

11.0%

3.3%

29.1%

DPT-1

6.4%

 

3.7%

 

DAISY

7.2%

10.9%

< 4%

 

TEDDY < age 5

 

 

13.1%

 

SEARCH < age 5

 

 

 

36.4%

BABYDIAB < age 5

 

 

 

32.3%

 

STRATEGIES TO BRING SCREENING FOR RISK TO CLINICAL PRACTICE

 

Screening relatives does identify a population of those at risk for clinical T1D; however, at least 85% who get T1D have no relatives with disease.  Thus, to truly prevent all T1D, testing of the general population would have to occur.  This could be done with current technology by testing all babies for genetic (HLA) risk at birth and then following with antibody testing.  The Population Level Estimate of type 1 Diabetes risk Genes in children (PLEDGE) study enrolls newborns from the general population and offers one-time genetic testing and follow-up autoantibody testing at 2 and 4 years of age (38). The study aims to demonstrate feasibility and to develop evidence to support eventual inclusion of a T1D screening program in standard primary care.

 

Other studies, such as The Environmental Determinants of Diabetes in the Young (TEDDY) study, the Diabetes Autoimmunity Study in the Young (DAISY), and the Global Platform for the Prevention of Autoimmune Diabetes (GPPAD) are exploring similar methodologies to screen and monitor for risk (24, 39, 40).  However, with an increasing number of individuals developing T1D even without the high-risk HLA types, such approaches may still miss some destined to develop disease. 

 

An alternative risk detection strategy for those without a family history may be to perform point-of-care antibody testing in a routine pediatric visit.  Since almost all who will develop diabetes before puberty will have antibodies by age 5; such testing could be done at age 4-5 and perhaps once again in the teenage years.  This method will still miss those who develop T1D before this age, but would likely be a cost-effective approach to finding those at risk.  If these at-risk subjects are monitored regularly until development of clinical disease they would benefit from reduced morbidity at time of diagnosis even if a prevention therapy were not yet available.

 

There are many ongoing projects aimed at screening members of the general population for diabetes autoantibodies even without prior HLA testing (23, 25, 41, 42).

 

As risk-screening programs employ varying assays and recruit from different populations, interpretation and translation of results is unclear. It is not yet known whether those found to be autoantibody positive through one program will experience the same rates of T1D progression and/or benefit from the same therapies as individuals who have participated in other screening and intervention efforts.

Source: (37)
 
 

PRENATAL INFLUENCES  

 

The prenatal environment can have profound effects on the developing fetus. With the recognition that antibodies often develop early in life and that essentially all those with established islet autoimmunity (two or more autoantibodies) will eventually develop TID, investigators have looked to the prenatal period to search for factors that could contribute to disease development in utero.  As shown in Table 3, decades of observational studies have yielded inconsistent results.  Yet this remains an important area of investigation and one that may lead to primary prevention strategies for T1D. The Environmental Determinants of Islet Autoimmunity (ENDIA) study is an ongoing prospective birth cohort study in Australia that enrolled infants and unborn infants of first degree relatives with T1D. Biologic samples including blood, stool, and saliva will be collected longitudinally for investigation of factors including viral exposures during pregnancy and early childhood, maternal and fetal microbiome, delivery method, maternal and early infant nutrition, pregnancy and early childhood body weight, and both innate and adaptive immune function. In 2018, the ENDIA study completed target enrollment of ~1500 subjects, who will be followed regularly until the development of islet autoimmunity (43).

 

Table 3.  Potential Prenatal Influences on TID Risk

Pre-natal or intrauterine exposure

Relative risk to offspring

Reference

Maternal age

Inconsistent data

(44-46)

Birth weight > 2 SD above norm (~4000g)

Inconsistent data

(47-51)

Birth weight < 2 SD below norm (~2500g)

Inconsistent data

(49-51)

Birth order: second and later borns

Inconsistent data

(46, 52, 53)

Birth interval < 3 years

Inconsistent data

(46, 54)

Caesarean delivery

Inconsistent data

(51, 55, 56)

Pre-eclampsia

Inconsistent data

(51, 57)

Pre-term delivery (<37 weeks gestation)

Inconsistent data

(51, 58)

Maternal vitamin D supplementation

Inconsistent data

(59-62)

Maternal antibiotic use

No association

(53, 63)

maternal BMI/pregnancy weight gain

No association

(51, 64)

Maternal omega 3 fatty acid supplementation

No association

(60, 65, 66)

 

Source: (67)

Investigators also have studied the early childhood period for clues to the causes of islet autoimmunity and TID; these have included both observational studies and randomized clinical trials. Such influences might be divided into early nutritional exposures and early microbial/infectious exposures, both of which can affect development of the normal immune system.

 

The inconsistent findings relating to environmental factors reported from observational studies and clinical trials led to the design and implementation of a large international comprehensive evaluation of genetically at-risk babies using cutting edge technologies to study genetics, genomics (gene function), metabolomics, and the microbiome. The Environmental Determinants of Diabetes in the Young (TEDDY) is an international prospective birth cohort study that recruited almost 8,000 babies at increased risk for TID (based on HLA and family history) from Finland, Germany, Sweden, and the US from 2004-2010.  Information on environmental exposures such as diet (including breastfeeding history), infections, vaccinations, and psychosocial stressors will be collected. Participants will be followed until the age of 15 for the development of islet autoimmunity or TID. The wealth of data from this study will provide a foundation for future randomized clinical trials (24). One interesting finding reported in December 2019 is that there are subtle differences in the gut microbiome—such as, persistent stool enterovirus B species--in children who develop islet autoimmunity compared to children who do not develop autoimmunity (68).

 

EARLY NUTRITIONAL EXPOSURES

 

Breastfeeding

 

The hypothesis that human breastmilk may protect against future TID development was presented as early as 1984 (69). Since then, there have been several prospective cohort studies to suggest that breastmilk lowers the risk of islet autoimmunity and TID, including the German BABYDIAB/BABYDIET study (70), the Colorado-based DAISY study (71), and the Norwegian MIDIA study (72), but others show no effect (73).  Although the data on whether breastmilk is protective against TID isn’t clear, it certainly isn’t harmful.  Given the well-established general benefits of breastfeeding, patients may safely be advised to follow the American Academy of Pediatrics’ guidelines related to infant feeding. The mechanism by which breastmilk may lower the risk of TID is uncertain, but one theory suggests that breastmilk has positive effects on the infant microbiome. The microbiome is discussed in greater detail below.  

 

Cow’s Milk and Bovine Insulin Exposure

 

In contrast to considering breastfeeding as potentially beneficial in protecting against autoimmunity, it was hypothesized that early introduction of cow’s milk or cow protein might accelerate disease.  This concept was tested in the Trial to Reduce IDDM in the Genetically at Risk (TRIGR) which asked whether weaning to hydrolyzed casein (which is free of bovine proteins including insulin) formula (n=1081) instead of regular cow’s milk formula (n=1078) in genetically at-risk infants could prevent or delay TID.  Though the TRIGR pilot study was suggestive of benefit, no benefit was seen in the fully powered study (74) (75). Similarly, The Finnish Dietary Intervention Trial for the Prevention of Type 1 Diabetes of (FINDIA) suggested that weaning to hydrolyzed cow’s milk formula was not effective in reducing the appearance of autoantibodies, though they did report that a patented cow’s milk formula specifically removing bovine insulin appeared to be beneficial in this pilot study (76).  While additional studies may be informative, current data does not support that weaning to hydrolyzed cow’s milk formula is protective against islet autoimmunity. 

 

Gluten Exposure

 

Both BABYDIAB (77) and DAISY (78) were observational studies that suggested an association between introduction of gluten and islet autoimmunity.  However, these studies had different results as to the timing of gluten introduction. Similarly, no effect was found in the BABYDIET study; a randomized controlled trial that asked whether delayed introduction of gluten to 6 vs 12 months would affect the risk of diabetes autoimmunity (79, 80).

 

Vitamin D and/or Omega 3 Fatty Acids

 

Vitamin D is an important component of a normal immune response; moreover, the higher incidence of TID in northern climates suggests that vitamin D deficiency could contribute to autoimmunity and TID.  However, data from observational studies is mixed on whether vitamin D and/or omega 3 supplementation is beneficial or not (60, 81-86). A pilot randomized trial of omega 3 supplementation to pregnant mothers and infants failed to demonstrate a profound immunologic effect of treatment (87). With routine vitamin D supplementation recommended for infants (88), it is unlikely that a fully powered randomized trial would be feasible to assess the impact on autoimmunity. 

 

MICROBIAL EXPOSURES

 

The Hygiene Hypothesis

 

Parallel to the rising incidence of TID and other autoimmune diseases, there has been a worldwide trend towards urbanization, increased standard of living, smaller family sizes, less crowded living conditions, safer water and food supplies, less cohabitation with animals, wide use of antibiotics, childhood vaccination, etc.  While these trends are generally considered improvements in human existence, the so-called “hygiene hypothesis,” proposed by Strachan in 1989 (89) suggests a possible downside; that is, that early microbial exposures might have a protective effect via the early education of the immune system and the development of normal tolerance to self-antigens.   Data cited in support of the hygiene hypothesis comes from comparisons between eastern Finland and Russian Karelia (Figure 7) (90-92).

 

Figure 7. Border between Finland and Russian Karelia, with a 6-fold difference in the incidence of TID, from "Karelia today”. The countries share a common border and ancestry and thus have similar geography, climate, vitamin D levels, and prevalence of HLA risk haplotypes. However, Finland has 6-fold higher incidence of TID. This markedly higher rate of TID is accompanied by a much lesser rate of infectious disease. In Finland as compared to Karelia 2% vs 24% had hepatitis A; 5% vs 24% had toxoplasma gondii; and 5% vs. 73% for helicobacter pylori. There is an ongoing study aiming to better understand the mechanisms that may underlie these differences.

 

The Microbiome

 

Another possible interface between microbial exposure and human disease is through the microbiome; that is the gut flora established within the first 3 years of life (93).  It has been hypothesized that perturbations in normal early microbiome development might pre-dispose to disease whether through direct modulation of innate immunity or via alteration of intestinal permeability and the downstream effects on adaptive immunity.   Interestingly, it appears that the gut microbiome is less diverse and less “protective” in individuals with islet autoimmunity or recent onset TID (94-96).  Whether this difference is cause, effect, or correlation isn’t known.   Nonetheless, multiple factors might affect the early intestinal microbiome, some of which also have been shown to correlate with risk of islet autoimmunity and TID.  For example, breastfeeding can alter the intestinal microbiome of the infant by increasing the number and diversity of beneficial microbiota (97, 98). As previously discussed, multiple prospective observational studies suggest that breastfeeding protects against future development of islet autoimmunity and TID, but there’s no evidence to connect this directly to the infant microbiome.  

 

Viral Infections

 

A viral etiology for initiation of autoimmunity is an attractive idea; a beta cell trophic virus could contribute to disease by directly killing beta cells, by leading to a chronic infection which triggers an immune response, or by molecular mimicry in which self-antigens are erroneously recognized as viral epitopes targeted for destruction.  Notably, these possible mechanisms would not necessarily point to a particular virus; any virus widespread in a population could theoretically lead to autoimmunity in genetically susceptible individuals if encountered at a vulnerable time in immune system or beta cell development.  With the notable exception of congenital rubella which is associated with type 1 diabetes (99), other data relating viruses to initiation of autoimmunity is less conclusive.  While some studies have reported viral “footprints” in islets from individuals who have died from TID, these have not been consistently confirmed.  Similarly, many studies have focused on enteroviruses, including coxsackie B, due to observations suggesting seasonal variation in antibody development that is reminiscent of the timing of such infections (100) (101), yet this remains controversial.  Aside from a viral role in the initiation of autoimmunity, others have proposed that acute viral infections may impact the transition from islet autoimmunity to clinical TID due to increased insulin demand during infections.  Patients commonly report an acute viral illness preceding the diagnosis of TID, and the clinical onset of TID more commonly presents in the fall and winter months in both the northern and southern hemispheres (102); but this does not imply a causal relationship.

 

Vaccinations

 

In recent decades, an increasing number of parents in Western countries have declined routine childhood vaccination of their children, which has created a situation with significant personal and public health consequences.  Multiple high-quality studies have thoroughly investigated vaccinations and TID, and none have found any association with islet autoimmunity or TID (103-107)

 

Sources: (88, 103-108)

DISEASE-MODIFYING THERAPY FOR PRECLINICAL TID

 

As previously discussed, the ability to recognize autoimmunity (via the detection of autoantibodies) in subjects even before the clinical onset of T1D affords the possibility of designing trials specifically for the high-risk population. One might consider established islet autoimmunity not only a marker of impending T1D, but a condition in its own right.  Just as hypertension warrants treatment to prevent stroke and myocardial infarction, in the future, TID may be treated in its earliest stages to prevent symptomatic hyperglycemia.  Some potential strategies are discussed in the following section. 

 

Many TID studies have tested antigen-based therapies.  With this type of therapy, the concept is that administration of a specific antigen could shift the immune response towards tolerance of the antigen.  For example, in allergy desensitization therapy, small amounts of antigen are repeatedly administered to ‘teach’ the immune system to be tolerant of the foreign protein so that the immune system no longer reacts.  In TID, the aim is to administer self-antigens in order to tolerize the immune system to beta-cell-derived proteins and downregulate the immune attack. Theoretically this can be done through oral, nasal, subcutaneous, or parenteral administration of antigen, with or without repeated dosing. Conceptually, antigen therapy should be more effective early in the disease process (i.e., to prevent progression from islet immunity to symptomatic disease rather than in those already clinically diagnosed) and thus most studies have targeted the at-risk population. 

 

Perhaps the most rigorously tested antigen therapy for pre-clinical T1D is insulin, as in the GGAP-03 POInt, DPT-1, TrialNet oral insulin, DIPP, and INIT II trials, described next. The JDRF-funded GGAP-03 POInT Trial, a primary intervention dose-finding study, is evaluating whether or not early exposure to oral insulin, even before those with high genetic risk develop autoantibodies, may confer greater benefit. Results are expected in early 2025. Preliminary results from the pre-POINT pilot trial suggest that higher doses of oral insulin may elicit greater immunologic response (109). In the Diabetes Prevention Trial (DPT-1), 372 family members of T1D probands who were positive for both ICA and mIAA were assigned to receive either daily oral insulin or placebo (110).  While this trial did not meet its primary endpoint, post-hoc analysis showed a delay in disease onset in participants with the highest levels of insulin autoantibodies. Specifically, those with a mIAA titer ≥80nU/ml showed a 4.5 year delay in disease onset and those with a mIAA titer ≥300nU/ml showed a 10 year delay in disease onset (111, 112). In response to these intriguing findings, Diabetes TrialNet launched a larger study to determine whether or not these results could be replicated  While the fully-powered TrialNet study showed no benefit to oral insulin in the primary cohort of more than 300 individuals, an independently-randomized cohort of 55 antibody positive individuals who had low first phase insulin response at baseline had a significant delay in disease progression in those treated with oral insulin (113). This intriguing finding raised the possibility that oral insulin may benefit those who are closer to clinical diagnosis; that is, those with more active disease.

 

In addition to studying oral insulin, the DPT-1 evaluated the effect of parenteral insulin on individuals who were considered to have the highest risk for T1D. These participants were ICA positive with abnormal beta-cell function (dysglycemia on an OGTT or low first phase insulin response on IVGTT). These 339 high risk participants were assigned to either close observation or low dose subcutaneous ultra-Lente insulin in addition to annual four-day continuous insulin infusions. While the therapy was found to be ineffective in preventing the progression to T1D, there was no excessive hypoglycemia, and a subset analysis found a temporary decrease in the immune response to beta cell proteins (114).

 

To date, trials with intranasal insulin have proven safe but ineffective in preserving insulin secretion. The Type 1 Diabetes Prediction and Prevention Study (DIPP), a randomized controlled trial evaluating the effects of intranasal insulin in children with high-risk genotypes and autoantibody positivity, was negative. When intranasal insulin was administered soon after the detection of autoantibodies, there was no delay in the progression to T1D (115). Similarly, the Intranasal Insulin Trial II (INIT II), which tested a different dose and dosing schedule of nasal insulin in a phase II prevention trial, showed that intranasal insulin was safe and induced an immune response, but this did not alter the progression to T1D. Participants were first-degree relatives of T1D probands with autoantibody positivity  (116, 117).

 

Another approach to antigen therapy is to use a plasmid to transfer DNA into cells, where it encodes for a given antigen, a technique that should decrease the anti-inflammatory response from intravenous, subcutaneous, oral, or nasal antigen delivery. This technique is being tested in the TrialNet TOPPLE T1D Study, a phase 1 trial launched in 2021 to evaluate the safety of a plasmid therapy called NNC0361-0041 in adults with recent-onset T1D. NNC0361-0041 encodes for four different human proteins: pre-proinsulin (PPI), transforming growth factor β1 (TGF-β1), interleukin-10 (IL-10), and interleukin-2 (IL-2) (118). In preclinical trials in NOD mice, the treatment was well-tolerated and led to beta cell preservation. If this phase 1 trial shows no safety concerns, then a larger study of the same treatment is planned to assess whether or not NNC0361-0041 can slow disease progression in the at-risk human population.

 

Antigen therapy may be more effective in both new-onset and at-risk populations when combined with other immune-modulating agents. Such combination trials are currently underway. In late 2020, enrollment was completed for a phase 1b/2a study assessing the safety and tolerability of different doses of an oral therapy called AG019 administered alone or in association with teplizumab infusions (see below) in individuals with recent-onset T1D. AG019 consists of live Lactococcus lactis bacteria, genetically modified to secrete human proinsulin and human interleukin 10. Results are pending (119).

 

While some trials have tested antigen-based therapies to treat islet immunity and prevent progression to clinical disease, others are building on successful studies of immunomodulating therapy in individuals with recently diagnosed TID. Examples include abatacept (Orencia; CTLA4 Ig) and teplizumab (Anti-CD3), both of which have been shown to slow loss of beta cell function post diagnosis. (See Recent Clinical Trials with Compelling Results and Figure 8). TrialNet recently completed enrollment of a placebo controlled trial testing abatacept in individuals with Stage 1 TID with results expected in late 2021 (120). In 2019, TrialNet published results of its placebo-controlled trial testing teplizumab in 76 individuals with Stage 2 TID. The trial demonstrated that a two-week course of teplizumab delayed the onset of clinical type 1 diabetes by two years and halved the rate of clinical diagnoses (121). This trial was highly significant in that it was the first ever to show that clinical type 1 diabetes can be delayed in children and adults at high risk. The latest findings from this trial, published in March of 2021, show ongoing delay of diabetes in the teplizumab treated group, with a median time to diagnosis of approximately 60 months (5 years) vs. approximately 27 months (2.3 years) in the placebo group (122).  Teplizumab has been granted Breakthrough Therapy Designation by the FDA, and the manufacturer of teplizumab is pursuing full FDA approval. 

 

Table 4. Clinical Preconceptions are Not Always Correct

AGE OF DIAGNOSIS: TID IS DIAGNOSED IN CHILDHOOD AND T2D IS DIAGNOSED IN ADULTHOOD.

At least 25% of people with TID are diagnosed as adults.  T1D is not “juvenile” diabetes.

WEIGHT: PEOPLE WITH TID ARE THIN, AND PEOPLE WITH T2D ARE OVERWEIGHT.

At least 50% of people living with TID in the US are overweight or obese, a statistic which mirrors the general US population.   Excess weight doesn’t prevent autoimmunity! 

CLINICAL PRESENTATION: THE ONSET OF TID IS DRAMATIC, AND INSULIN IS IMMEDIATELY REQUIRED FOR TREATMENT.

While this is generally true, the presentation of TID tends to be less abrupt in adults (in whom beta cell destruction is more gradual).  Moreover, insulin isn’t always required immediately, especially in adults or in overweight individuals, where treatments to improve insulin sensitivity such as weight loss and/or metformin, may be sufficient to control blood glucose for a limited period of time.

RESIDUAL INSULIN SECRETION: PEOPLE WITH TID HAVE AN ABSOLUTE INSULIN DEFICIENCY.

At the time of diagnosis, essentially all people with TID have clinically significant amounts of C-peptide.  Furthermore, among those with > 40 years of TID, 6-16% have a non-fasting C-peptide level ≥0.017 nmol/L.

AUTOIMMUNITY: IF YOU DON’T FIND ANTIBODIES, IT’S NOT TID.

There are five well-characterized antibodies associated with TID; most commercial laboratories don’t measure all five, so the results can be misleading.  In addition, up to 10% of those with newly-diagnosed TID may not have antibodies.  While these individuals may have a monogenic form of diabetes (http://monogenicdiabetes.uchicago.edu), it is also possible that they have autoimmunity not detectable with current antibody measurements.

Sources: (5, 123, 124)

 

IMPORTANCE OF BETA CELL PRESERVATION IN LIGHT OF RISKS OF THERAPY

 

The preservation of residual beta cell function, as measured by C-peptide, has repeatedly been demonstrated to be clinically important in those with T1D, warranting ongoing efforts to develop therapies to prevent beta cell destruction both in individuals with islet autoimmunity and in those with new-onset disease. In addition to its primary finding that intensive insulin therapy results in better outcomes (125, 126), the landmark Diabetes Control and Complications Trial (DCCT) showed that among intensively treated subjects, those who had ≥ 0.20 nmol/l stimulated C-peptide initially or sustained over a year had fewer complications, including 79% risk reduction in progression of retinopathy (127, 128).  Importantly, these benefits were seen in the face of markedly less severe hypoglycemia. Subjects in the intensive insulin therapy group with ≥ 0.20 nmol/l C-peptide had about the same frequency of severe hypoglycemia as those in the standard care group; a 62% relative reduction as compared to those who received intensive therapy without this level of C-peptide. Subsequent analyses have demonstrated that even lower levels of preserved beta cell function in DCCT subjects were protective against complications (129).  Importantly, a beneficial effect of preserved insulin secretion was also recently reported in those with type 2 diabetes. Endogenous insulin deficiency was strongly associated with hypoglycemia and a limited ability to control HbA1c in Type 2 subjects in the ACCORD study (130). Together, these data strongly support the concept that preserved insulin secretion coupled with intensive insulin therapy can reduce diabetes complications while averting the severe hypoglycemia that has been a limiting factor in attaining glycemic control.              

Islet transplant studies confirm a positive association between C-peptide secretion and a lower risk of hypoglycemia.  Subjects eligible for islet transplantation are largely individuals suffering from severe hypoglycemic unawareness.  Vantyghem et al. showed that while significant beta cell function was required to improve mean glucose, lower glucose excursions, and result in insulin independence in transplant patients, only minimal beta cell function was needed to abrogate severe hypoglycemic events (131). 

 

Additionally, post islet-cell transplant patients with higher as compared to absent or minimal C-peptide levels are more likely to maintain fasting blood glucose values in the 60-140mg/dL (3.3 – 7.8 mmol/l) range, HbA1c values <6.5% (47.4 mmol/mol), and insulin independence after transplantation (132). The DCCT showed similar metabolic benefits in those with residual C-peptide. In this trial, patients with C-peptide ≥ 0.2nmol/l had lower fasting glucose and HbA1c values. A 9-year longitudinal analysis showed that for every 1 nmol/l increase in baseline stimulated C-peptide, there was an associated 1% reduction in HbA1c among intensively treated DCCT participants (133). Such positive clinical outcomes in those with preserved C-peptide reinforce the significance of efforts to protect beta cell function.

 

Of course, the benefits of beta cell preservation must be weighed against the intrinsic risks of therapies used to preserve C-peptide. Two therapies in particular highlight the challenges of balancing benefits with risk.  First, one of the initial immunomodulatory therapies used in T1D was cyclosporine, a general immunosuppressant. Treatment with cyclosporine induced remission from insulin dependence in children with recently diagnosed TID, with half of participants not requiring insulin after a full year of treatment (134). Unfortunately, the risks of using this drug were deemed to outweigh the benefits. Continuous effectiveness required continuous therapy, which induced nephrotoxicity (134).

 

More recently, studies with autologous hematopoietic stem cell transplant (HSCT) in the new onset population have further highlighted the risks of more aggressive approaches to treatment.  Although the pooled data from HSCT trials suggests that this therapy imparts a high diabetes remission rate, the remission is not durable, and there are significant risks associated with the treatment, including neutropenic fever, serious infection, gonadal failure, and even death (135).

 

Importantly, there are dozens of immunotherapeutic agents or combinations of agents that are safely used in current clinical practice in other autoimmune diseases.  For example, adults and children with juvenile idiopathic arthritis (JIA) are routinely treated with immunotherapy, an approach that has markedly transformed the lives of many living with this disease. Similarly, the aim for T1D is to use disease modifying therapies prudently and safely to truly improve the lives of those living with T1D. Possible approaches may include short term therapy aimed at inducing a long-term effect (tolerance), intermittent therapy, or limited doses of chronic therapy.  Some of these methodologies are described below.

 

CLINICAL TRIALS WITH COMPELLING RESULTS IN NEW-ONSET T1D

 

Selecting therapies for clinical trials is based on multiple factors.  We can now take advantage of the tremendous advances in understanding the disease process and basic and applied immunology.  As illustrated in Figure 8, there are now therapies that target specific mechanisms underlying disease. Trials are considered in the context of what is known about safety of the therapy and efficacy in animal models, pilot studies, and other autoimmune diseases.  Using these approaches, we have succeeded in altering disease course without the excessive risk previously described. 

Figure 8. Major pathways leading to beta cell destruction and potential mechanisms underlying the use of selected therapies. Both CD4 and CD8 T effector cells infiltrate and impair/destroy beta cells along with inflammatory cytokines such as IL 21, IL-1 and IL12/23. Anti-IL21/Liraglutide, Golimumab, Ustekinumab, Anakinra, and Canakinumab are aimed at blocking these inflammatory pathways. Activation of Teff cells depends upon presentation of antigen to naïve T cells which result in both Teff turning the immune response “on” and Treg cells turning the immune response “off”. Rituximab decreases B cells and therefore decreases the presentation of antigen to the immune system. Abatacept blocks co-stimulation and oral insulin (and other antigen therapy including the use of antigen specific dendritic cells) alters the response to self-antigen. The aim in both cases is to deviate the response to Treg cells or keep Teff cells from fully activating. ATG and anti-CD3 agents modulate and/or deplete activated T cells. Alefacept has a similar mechanism although primarily aimed at memory T cells. By blocking IL-6, Tocilizumab should change the balance of immune activation towards T regulatory cells. Similarly, GSCF, IL-2 (at the “right dose”), and infusion of Treg cells should preferentially increase Treg cells.

It is well established that T1D is the result of an immune-cell mediated destruction of the pancreatic beta cells. Many research efforts have thus targeted T-cells as well as the cells with which they interact.  As in secondary prevention trials, anti-inflammatory agents, antigen therapies, and immunomodulatory drugs have all been used in tertiary prevention studies, which are designed to stop further beta cell destruction in the new onset population, therefore preventing complications. In addition, cellular therapies have been tested in this population.  Excitingly, several therapies have now been shown to safely alter the disease course, particularly in the period soon after drug administration, allowing treated subjects to retain more C-peptide than controls 1-4 years later (Figure 9). Thus, while not yet ready for clinical use by endocrinologists, it is likely that immunotherapy with these or other agents will become a part of T1D new onset clinical care in the future.

 

Otelixizumab and Teplizumab (anti-CD3)

 

Some success in beta cell preservation has been shown with Teplizumab (hOKT3gl Ala-Ala) and Otelixizumab (ChAglyCD3), both of which are humanized Anti-CD3 monoclonal antibodies directed against the CD3 portion of the T-cell receptor. These drugs are distinct from OKT3, an anti-T cell agent with significant short term adverse effects. A study with Otelixizumab showed preserved insulin secretion for up to four years after 80 new-onset participants were treated with a single 6-day course of drug (136, 137). At 6, 12, and 18 months, the treatment group showed more residual beta cell function and a delay in the rise in insulin requirements as compared to the placebo group.

 

Similarly, in 2002, Herold et al. reported that a single 14-day course of Teplizumab given within the first 2 months of diagnosis resulted in more residual beta cell function at 12 months as compared to untreated individuals (138). While the effect of the therapy appeared most pronounced early on, follow-up of study participants continued to show differences in insulin production between treated and control subjects at 2 and 5 years after drug administration (139).In the AbATE Trial, a second course of Teplizumab was given 12 months after the first. In this study, C-peptide loss was delayed by an average of 15.9 months in treated subjects versus control subjects at 2 years (140). Finally, the Protégé Trial was a large phase III, placebo controlled randomized trial.  While this study failed to meet its primary endpoint, post-hoc analysis found preserved beta cell function in a subset of the recent onset individuals who received Teplizumab as compared with placebo (141). As previously discussed, TrialNet  found that 14 consecutive daily infusions of Teplizumab successfully delayed the progression from  stage 2 T1D  to stage 3 TID in family members by up to 3 years (122). Additionally, in [i]2019 Provention Bio launched Recent-Onset Type 1 Diabetes Trial Evaluating Efficacy and Safety of Teplizumab (PROTECT), a phase 3 trial (n=300) comparing two courses of 12 daily infusions of either teplizumab or placebo. The two courses are administered either 6 or 12 months apart.  Results of the PROTECT study will provide additional safety and efficacy data for use of teplizumab in T1D.

 

Rituximab (anti-CD20)

 

In addition to anti-T cell therapies, investigators have studied anti-B-cell agents. A placebo controlled, double masked, randomized trial with Rituximab (anti-CD20) found that a single course of drug preserved C-peptide for 8.2 months in the drug-treated group compared to the placebo-treated group (142). The precise mechanism of action of Rituximab remains unclear, although it is believed that this therapy may reduce the production of pro-inflammatory cytokines or inhibit B lymphocyte antigen presentation, thus inhibiting the cascade of events leading to T-lymphocyte activation.  Other anti-B-cell agents are being considered for study.

 

ATG-GCSF

 

In 2019, TrialNet completed a 3-arm study (n=82) of ATG compared to ATG and granulocyte colony stimulating factor (GCSF) compared to placebo. GCSF was combined with ATG to test whether GCSF may facilitate the return of T-regs following ATG-induced lymphocyte depletion. The 2-year C-peptide AUC was significantly higher in ATG treated subjects compared to placebo treated. Interestingly, GCSF did not provide additional benefit compared to ATG alone (143). Given the demonstrated benefit of low-dose ATG in stage 3 T1D, TrialNet may study this therapy in those with earlier stage disease.

 

Abatacept (CTLA4 Ig)

 

Abatacept works through co-stimulatory blockade; that is, the interruption of the interactions between different components of the immune system that propagate an immune response.  A placebo-controlled, double-masked, randomized trial in the new onset population showed that when Abatacept therapy was provided continuously over 2 years, treated individuals benefited from a 9.6-month delay in beta cell destruction (144). Like the anti-B cell and anti-T cell therapies, the effect of Abatacept therapy on insulin secretion was most pronounced soon after initiation of drug.  Importantly, while continued loss of beta cell function occurred over the remaining treatment period, when the drug was withdrawn, no acceleration of disease progression was seen (145). These findings set the stage for testing a shorter course of therapy in those with early stage T1D (stage 1 or stage 2).  TrialNet is now studying Abatacept therapy in this population with the aim to prevent or slow onset of clinical disease (120).

 

Alefacept (LFA-3 Ig)

 

The T1Dal study assessed the use of Alefacept (LFA-3 Ig) in the new onset population in a placebo-controlled, double-masked, randomized trial.  It was expected that Alefacept would target the memory cells of the immune response and mechanistic studies indicated that this was the case.  Unfortunately, there was insufficient drug available to fully complete the study.  As such, while there was a trend, the difference in C-peptide secretion measured at 2h between treated and control subjects was not statistically significant at 1 year.  However, Alefacept therapy did preserve the 4h C-Peptide AUC at 1 year with lower insulin use, and also reduced hypoglycemic events, suggesting at least some efficacy (146). Moreover, further data found a positive effect of therapy 2 years after randomization (147).

 

Cytokine and Anti-cytokine Therapies

 

IL-1: It has been recognized for many years that the cytokine IL-1, a key factor in the inflammatory response, can injure beta cells.  However, in recently diagnosed patients, two Phase 2 trials with different anti-IL-1 therapies (Anakinra and Canakinumab) failed to preserve beta cell function (148).

 

IL-2: IL-2 is necessary for immune cell proliferation, but the amount of IL-2 needed to promote T regulatory cells differs from that needed to promote T effector cells. A pilot study using IL-2 in T1D subjects aimed to exploit this difference and even exaggerate it by combining the therapy with Rapamycin, which selectively blocks T effector cells, thus resulting in an augmentation of T regulatory cells.  Indeed, a marked increase in T regulatory cells was seen.  Unfortunately, a transient decrease in beta cell function was also observed, leading to the trial’s early termination (149). It was suggested that (150) the decrease in beta cell function may have been due to IL-2 simulation of eosinophils and natural killer cells and it has thus been postulated that giving a lower dose or alternative form of IL-2 may more selectively augment Tregs. This was suggested by a small (n=24) study which defined an IL-2 dose range that was both safe and able to induce Treg expansion (151).

 

IL-6 is another important cytokine in the immune cascade.  It promotes a particular type of T effector cell (Th17 cells), and some patients with T1D have an exaggerated response to IL-6.  Tocilizumab blocks the IL-6 receptor and is effective (and approved for clinical use) in adult and pediatric arthritis patients. The Tocilizumab (TCZ) in New-onset Type 1 Diabetes (EXTEND) trial was a randomized trial in adults and children (n=136) with new onset T1D, completed in 2020. While the study confirmed the safety of tocilizumab, it did not demonstrate efficacy in new onset T1D, as measured by 2-hour C-peptide AUC in response to standardized MMTT (150).

 

IL12 and IL23 may indirectly contribute to the etiopathology of T1D, as they are involved in the production of IFN λ and IL-17, key cytokines in the generation of Th1 and Th17 effector cells. Ustekinumab is a monoclonal antibody that blocks a subunit common to IL12 and IL23 and is currently approved for treatment of psoriasis, psoriatic arthritis, ulcerative colitis and Crohn’s disease. Its efficacy to preserve C-peptide is being tested in a Canadian Phase 2/3 study in adults with recently diagnosed T1D (152, 153).

 

Anti-TNFα: The results of the T1GER Study, which assessed the effects of the anti-TNFα medication golimumab on beta cell function in 84 youth with new-onset T1D, were published in November, 2020.  Participants aged 6-21 received either subcutaneous golimumab or placebo via injection in a 2:1 randomization for 52 weeks. At week 52, endogenous insulin production was significantly higher in the treatment group (0.64±0.42 pmol per milliliter vs. 0.43±0.39 pmol per milliliter, P<0.001) and exogenous insulin use was significantly lower. There was no significant difference in mean HbA1c or number of hypoglycemic events between groups, although there were more hypoglycemic events that met adverse event criteria in the treatment group. The promising results of this trial may warrant further investigation of anti-TNFα agents (154).

 

Anti-IL-21: A recent trial funded by Novo Nordisk investigated combination therapy with anti-interleukin (IL)-21 antibody and liraglutide (to improve β-cell function) as a means of enabling β-cell survival. 308 participants were randomly assigned to receive either anti-IL-21 plus liraglutide, anti-IL-21, liraglutide, or placebo (77 assigned to each group). Compared with placebo (ratio to baseline 0·61, 39% decrease), the decrease in MMTT-stimulated C-peptide concentration from baseline to week 54 was significantly smaller with combination treatment (0·90, 10% decrease; estimated treatment ratio 1·48, 95% CI 1·16-1·89; p=0·0017), but not with anti-IL-21 alone (1·23, 0·97-1·57; p=0·093) or liraglutide alone (1·12, 0·87-1·42; p=0·38). It is important to note, however, that 26 weeks after cessation of therapy, both the liraglutide monotherapy group and the combination therapy group showed increased C-peptide loss, perhaps suggesting that while liraglutide may transiently augment insulin secretion in the peri-diagnostic period, it is not beneficial to long-term beta cell function or survival (155). 

 

OTHER APPROACHES

 

Cellular Therapy

 

Several clinical trials have tested administration of cells as compared to pharmaceutical agents with the aim of preserving beta cells.  These include administration of antigen specific dendritic cells which are thought to restore immune tolerance by exploiting the role of dendritic cells in presenting antigen to the immune system (156).  Autologous mesenchymal stromal cells (MSCs) are considered to have immunomodulatory properties and have also been examined and shown  preliminary safety and proof of concept information in a pilot study (157). Other investigators have infused participants with T-regulatory cells (Tregs).  These cells, which can come from saved umbilical cord blood or by expanding the patient’s own cells, should increase the number of Tregs, thereby altering the immune balance with T-effector cells and preventing further beta cell injury.  Small studies to date have had conflicting results (158-160);

 

Therapies Directed at Components of the Innate Immune System

 

General anti-inflammatory agents have been tested as single agents in stage 3 TID and may be used in combination with other therapies in the future. For example, alpha-1-antitrypsin (A1AT) is a serum protease inhibitor that broadly suppresses pro-inflammatory cytokines such as IL-1, TNF-α, and IL-6.  It has been tested in stage 3 TID, where it appears safe and well-tolerated (161). Bacillus Calmette-Guerin (BCG) has been proposed as a “vaccine” for those with T1D, citing the concept that BCG stimulation of innate immunity would alter the cytokine attack on beta cells. Notably, BCG is widely used, particularly in Europe, as a vaccine to prevent tuberculosis. Despite this broad usage, there is no epidemiological evidence that BCG administration has impacted the incidence of T1D. Moreover, a large, placebo controlled randomized trial demonstrated that BCG has no effect on insulin secretion, insulin requirements, or HbA1c in individuals with new onset T1D (162). Finally, the tyrosine kinase inhibitor imatinib (Gleevac), developed to treat leukemia, has several effects supporting its use in autoimmunity and T1D. The initial proposed mechanism of action is that the therapy reduces innate inflammation (163). However, other studies suggest it may also directly improve beta cell secretion (164). In a recent multicenter, randomized, double-blind, placebo-controlled study, 64 newly diagnosed adults were treated with either a 26-week course of imatinib or placebo in a 2:1 ratio. The study met its primary endpoint, showing preserved c-peptide secretion in the treatment group at 12 months. However, this effect was not sustained out to 24 months. Additionally, during the 24-month follow-up, 71% of participants who received imatinib had a grade 2 severity or worse adverse event. Imatinib might offer a novel means to alter the course of type 1 diabetes, but care must be taken to monitor for toxicities. Further trials to define an ideal dose and duration of therapy and to evaluate safety and efficacy in children or the at-risk population should be considered (165).  

 

LESSONS FROM TRIALS WITH DISEASE MODIFYING THERAPIES

 

The trials that have successfully altered the course of disease by changing the rate of loss of C-peptide, even if for a brief period of time, have taught us much about the immune system and the natural history of T1D. First, it appears that the time of administration in the course of T1D may determine the effectiveness of a therapy as there appears to be a window during which agents may elicit the greatest effect upon the autoimmune process. Interestingly, in the cases of rituximab, otelixizumab/teplizumab, alefacept, ATG, golimumab, anti-IL-21, and abatacept, each of which has a different mechanism of action, treatment effected a marked delay in beta cell destruction/dysfunction initially, but thereafter, rates of decline in C-peptide paralleled those of the placebo groups (136, 140, 142-144, 155, 165, 166). Collectively, these observations suggest a difference in immune activity soon after diagnosis as compared with later on in the disease course (see Figure 9).

Figure 9. Stylized representation of selected new onset clinical trial results. Studies with positive outcomes, whether using a single course of treatment (136, 140, 142) or continuous therapy (144) appeared to have the most pronounced effects early after treatment started. See text for details.

Because of the time-dependent nature of the therapeutic response, the traditional approach of testing therapies in those with new-onset T1D before moving them “upstream” for use in treating autoimmunity may not be optimal.   Several medications or combinations of medications are more likely to be effective earlier in disease.  Thus, demonstration of efficacy in new onset trials should not be required before testing whether therapies can effectively treat islet autoimmunity. 

 

The results of several trials have demonstrated that not all T1D patients are alike, and they vary in their response to therapy.  For instance, in the Abate trial, 45% of subjects treated with teplizumab appeared to respond to the drug, showing almost no change in C-peptide secretion at two years, whereas 55% were deemed “non-responders” as their C-peptide secretion was not distinguishable from controls. Post-hoc analysis suggests that responders had lower A1C levels, less exogenous insulin use, and fewer Th-1-like T cells than non-responders (140). Next, post-hoc analysis from the Protégé trial revealed that C-peptide preservation was better in teplizumab treated patients who were aged 8-17, randomized within 6 weeks of diagnosis, had mean C-peptide AUC > 0.2nmol/l, A1c< 7.5%, and insulin dose < 0.4 units/kg/day (167). Last, as previously discussed, upon initial analysis of DPT-1 data, oral insulin did not appear to prevent T1D in the at-risk population. However, subsequent analysis showed a marked delay in diabetes development among those participants who had high titer anti-insulin autoantibodies (111). These results suggest that individualized therapies, which take into account a patient’s unique characteristics, are not only a possibility, but may be a necessity.

 

Participant age also appears to play a role in response to therapy, suggesting that optimal disease modifying agents may differ between pediatric and adult populations. Pre-teen children have less C-peptide at diagnosis than older children and adults.  All age groups of children have a markedly different rate of fall of C-peptide than adults in the first year after diagnosis (168). Additionally, prior to diagnosis, children progress much faster through the preclinical stages of disease.  Specifically, children with early autoimmunity (1 antibody) are more likely to develop established autoimmunity (2+antibodies) than adults; and children with established autoimmunity with or without abnormal glucose tolerance progress more rapidly to clinical diabetes than adults (169). Historically, the FDA has required that therapies first be tested in the adult population before they may be approved for use in the pediatric population.  However, this approach may prevent researchers from identifying therapies that may only be viable in pediatric populations.  Changing this paradigm was the focus of a recent American Diabetes Association consensus conference on disease modifying therapy (169). 

 

In the next few years, not only will new agents be tested, but the community will build on these results by using them in selected individuals (personalized medicine), in combination trials, and at different stages of disease.  Each step takes us closer to clinical use of a disease modifying agent.

 

RESIDUAL INSULIN SECRETION

 

Traditional teaching holds that all subjects with T1D will eventually lose all of their beta cells.  This statement is no longer true; multiple lines of research demonstrate that a proportion of those even with longstanding T1D may have residual beta cell function.  The Joslin Medalist study showed that 67% of 411 T1D subjects at least 50 years from diagnosis had at least minimal (0.03 nmol/l) random serum C-peptide levels. Of these individuals, 2.6% had random serum C-peptide ≥ 0.20 nmol/l. Post-mortem analysis of pancreata from these same subjects revealed that insulin positive cells were noted in 9/9 pancreases studied (170).  Since many of the Joslin Medalists were diagnosed at a time when life expectancy was markedly reduced in those with T1D, it was felt that this was a unique population, not representative of the majority of people with T1D and that the preservation of C-peptide itself may have contributed to their long-term survival.  However, multiple studies have now confirmed that C-peptide is present in a significant proportion of individuals with T1D.  At the time of diagnosis, essentially all individuals (both youth and adults) have clinically significant levels of C-peptide (123, 168, 171). Two years after diagnosis, more than 66% of individuals retain these high levels (168). Unfortunately, with increasing duration of disease, the proportion of those with detectable C-peptide falls (124, 168, 170). However, as recently reported by Davis et al. (124), about 6-7% of those even more than 40 years from diagnosis have measurable C-peptide and more sensitive assays can actually detect C-peptide in a greater proportion of individuals.  Moreover, like the pancreata from the Joslin cohort, studies from those who have had T1D for at least 4 years have shown that residual (insulin-positive) β-cells can be found in ~ 40% of T1D pancreases upon autopsy (172).  Careful studies of post-mortem samples using new technologies have suggested that insulin-positive cells may be scattered in the exocrine tissue, raising the tantalizing possibility that new beta cells could emerge.  Longitudinal studies of those long from diagnosis with low levels of C-peptide are underway to better understand variation over time.

 

There are two important take-aways from these new data. First, the presence of C-peptide does NOT rule out a T1D diagnosis.   Yet, this data should not be over-interpreted; most individuals will eventually lose essentially all of their C-peptide secretion.  The Davis study showed that 93% of those diagnosed as children had absent or extremely low levels of C-peptide >20 years from diagnosis (124).

 

To date, there are no therapies that have regenerated beta cells in humans.  It is abundantly clear that mouse and human beta cells are markedly different, and therapies touted to grow cells in mice have not had such effects in humans.  Instead of regeneration, replacement of dead or dysfunctional beta cells may be a viable option.  Beta cell replacement is currently done through either whole pancreas or islet transplantation in conjunction with immune therapies to suppress the alloimmune (tissue rejection) and autoimmune (initial disease process) response.  While outside the scope of this chapter, it has been recently recommended that those with severe hypoglycemic unawareness be referred for islet transplant (173). Other efforts to replace beta cells include placing them in capsules to allow viability and function while blocking immune cells from entering the capsules.  These efforts remain experimental. 

FUTURE CONSIDERATIONS

 

Despite advances in glucose monitoring and insulin delivery, the daily psychological and financial burden of disease on individuals, their families, and society together with the persistence of complications and reduced life span demand a paradigm shift.

 

As of 2021, we know much about the natural history of disease.  We know that antibodies can develop early in life and that essentially all of those with established islet autoimmunity will develop clinically overt disease.  We also know that identifying these individuals is of significant clinical benefit.  Those with islet autoimmunity followed carefully until diagnosis have markedly less morbidity at the time of diagnosis and lower HbA1c values. Family members of T1D probands should be made aware of their disease risk and should be offered autoantibody screening and enrollment in monitoring trials. Correspondingly, patients with TID should be informed of the opportunity to have their relatives screened for TID risk in the setting of a clinical research study.

 

While the interaction of humans with their environment must contribute to disease; how this occurs is still being elucidated.  It is likely that there are many different paths by which individual gene/environment interactions result in T1D; suggesting that dissecting this heterogeneity will provide better insights and therapies.

 

Whatever the primary cause, we know that the immune system is involved in disease progression. There have been successes in delaying beta cell destruction. Looking ahead, we will likely see the development of more targeted immunotherapies as well as more trials with combination therapies. Advances in treating childhood cancers have relied upon combining multiple approaches; this will be mimicked in T1D as well.  More studies will be done in those with islet autoimmunity and variations in dose and route of administration of drugs will be tested in the search for greater efficacy.  With newer and safer drugs, studies are likely to test chronic intermittent treatment for both islet cell autoimmunity and in new-onset TID to prevent further beta cell loss.  Future studies will reflect the heterogeneity of TID.  As medicine in general becomes more personalized, TID disease modifying therapies will target those most likely to benefit, whether because they are more likely to respond to therapy, or because their underlying disease is predicted to be worse. 

 

Yet, there are non-scientific barriers to the use of disease modifying therapies for either islet cell autoimmunity or new-onset TID.  One barrier is the lack of familiarity with these therapies amongst clinicians.  Immune-modulating medications are used routinely by rheumatologists; whereas endocrinologists and others who care for people with TID are generally less comfortable with these therapies.  This lack of familiarity exaggerates the risks and minimizes the benefits of immune-modulating medications.  If we consider islet cell autoimmunity a silent disease in the same way that we consider hypertension a silent disease, then it makes sense to prevent the consequences of that disease, such as hyperglycemia in the case of islet cell autoimmunity, or cardiovascular disease in the case of hypertension.  Similarly, if we consider new-onset TID in the same way we consider JIA, our goal in TID is to preserve beta cell function, just as in JIA, the goal is to preserve joint function. 

 

With a shift in mindset and training, and in anticipation of successful clinical trials, one can envision a not-too-distant future in which endocrinologists might use immune modulating therapies to treat their patients who have islet cell autoimmunity and/or new-onset TID.   

 

Table 5. How to Keep Informed About Research Opportunities

TrialNet

http://www.trialnet.org/

Offers free autoantibody screening to relatives of individuals with type 1 diabetes. If autoantibody positive, participants may be eligible for a diabetes prevention or preservation trial.

Offers New-onset trials to preserve beta cell function in those with new onset T1D (typically within 100 days of diagnosis)

ClinicalTrials.gov

https://clinicaltrials.gov/

Offers a complete registry of clinical trials being conducted in the US and worldwide. Provides an online search tool that allows users to search for clinical trials for which they might be eligible.

JDRF’s Clinical Trial Finder

 

Clinical Trials

JDRF is a global organization funding T1D research aimed at improving the lives of those living with the disease. JDRF has created a search tool that matches potential participants with enrolling trials.

Immune Tolerance Network

 

http://www.immunetolerance.org/

Offers clinical trials aimed at developing new therapeutic approaches for many immune-mediated diseases, including T1D.

 

REFERENCES

 

  1. JDRF Fact Sheet(accessed 2021 Dec 14) Available from: https://www.jdrf.org/t1d-resources/about/facts/.
  2. Centers for Disease Control and Prevention, National Diabetes Statistics Report, 2014. 2014; (accessed 2021 Dec 14) Available from: http://www.cdc.gov/diabetes/pubs/statsreport14/national-diabetes-report-web.pdf.
  3. Patterson, C., et al., Trends in childhood type 1 diabetes incidence in Europe during 1989-2008:Evidence of non-uniformity over time in rates of increase. Diabetologia, 2012. 55(8): p. 2142-2147.
  4. Bell, R., et al., Diabetes in non-Hispanic white youth:prevalence, incidence, and clinical characteristics: the SEARCH for diabetes in youth study.Diabetes Care, 2009. 32(Suppl 2): p. S102-S111.
  5. Miller, K.M., et al., Current State of Type 1 Diabetes Treatment in the U.S.: Updated Data From the T1D Exchange Clinic Registry. Diabetes Care, 2015. 38(6): p. 971-978.
  6. Liese, A.D., R.B. D'Agostino, Jr., and R. Hamman, The burden of diabetes mellitus among US youth; prevalence estimates from the SEARCH for diabetes in youth study. Pediatrics, 2006. 118(4): p. 1510-18.
  7. Lawrence, J., et al., Trends in incidence of type 1 diabetes among non-Hispanic white youth in the U.S., 2002-2009 Diabetes, 2014. 63(11): p. 3938-3945.
  8. Patterson, C., et al., Incidence trends for childhood type 1 diabetes in Europe during 1989-2003 and predicted new cases 2005-20: a multicentre prospective registration study. Lancet, 2009. 373(9680): p. 2027-2033.
  9. Incidence and trends of childhood Type 1 diabetes worldwide 1990-1999. Diabet. Med, 2006. 23(8): p. 857-866.
  10. Karvonen, M., et al., Incidence of childhood type 1 diabetes worldwide. Diabetes Mondiale (DiaMond) Project Group. Diabetes Care, 2000. 23(10): p. 1516-1526.
  11. Warram, J.H., et al., Differences in risk of insulin-dependent diabetes in offspring of diabetic mothers and diabetic fathers. N Engl J Med, 1984. 311(3): p. 149-52.
  12. Pociot, F., et al., A nationwide population-based study of the familial aggregation of type 1 (insulin-dependent) diabetes mellitus in Denmark. Danish Study Group of Diabetes in Childhood. Diabetologia, 1993. 36(9): p. 870-875.
  13. Nistico, L., et al., Emerging effects of early environmental factors over genetic background for type 1 diabetes susceptibility: evidence from a Nationwide Italian Twin Study. J Clin Endocrinol Metab, 2012. 97(8): p. E1483-E1491.
  14. Kyvik, K., A. Green, and H. Beck-Nielsen, Concordance rates of insulin dependent diabetes mellitus: apopulation based study of young Danish twins. BMJ, 1995. 311(7010): p. 913-917.
  15. Redondo, M., et al., Concordance for islet autoimmunity among monozygotic twins. N Engl J Med, 2008. 359(26): p. 2849-2850.
  16. Eisenbarth, G.S., Type I diabetes mellitus. A chronic autoimmune disease. N. Engl. J. Med, 1986. 314(21): p. 1360-1368.
  17. Insel, R.A., et al., Staging presymptomatic type 1 diabetes: a scientific statement of JDRF, the Endocrine Society, and the American Diabetes Association. Diabetes Care, 2015. 38(10): p. 1964-74.
  18. Concannon, P., S.S. Rich, and G.T. Nepom, Genetics of type 1A diabetes. N. Engl. J. Med, 2009. 360(16): p. 1646-1654.
  19. Knip, M., Can we predict type 1 diabetes in the general population? Diabetes Care, 2002. 25(3): p. 623-5.
  20. Steck, A.K., et al., Predictors of Progression From the Appearance of Islet Autoantibodies to Early Childhood Diabetes: The Environmental Determinants of Diabetes in the Young (TEDDY). Diabetes Care, 2015. 38(5): p. 808-13.
  21. Gillespie, K.M., et al., The rising incidence of childhood type 1 diabetes and reduced contribution of high-risk HLA haplotypes. Lancet, 2004. 364(9446): p. 1699-1700.
  22. Mahon, J.L., et al., The TrialNet Natural History Study of the Development of Type 1 Diabetes: objectives, design, and initial results. Pediatr. Diabetes, 2008.
  23. JDRF. T1Detect: Learn why you should be screened.(accessed 2021 Dec 14); Available from: https://www.jdrf.org/t1d-resources/t1detect/.
  24. Hagopian, W.A., et al., TEDDY--The Environmental Determinants of Diabetes in the Young: an observational clinical trial. Ann N Y Acad Sci, 2006. 1079: p. 320-6.
  25. Combined Antibody Screening for Celiac and Diabetes Evaluation (CASCADE). 2020.
  26. Parikka V., N.-S.K., Saarinen M, Simell T, Ilonen J, Hyöty H, Veijola R, Knip M, Simell O., Early seroconversion and rapidly increasing autoantibody concentrations predict prepubertal manifestation of type 1 diabetes in children at genetic risk. Diabetologia, 2012. 55(7): p. 1926-36.
  27. Vehik, K., et al., Methods, quality control and specimen management in an international multicentre investigation of type 1 diabetes: TEDDY. Diabetes Metab Res Rev, 2013. 29(7): p. 557-67.
  28. Ziegler, A.G., et al., Seroconversion to Multiple Islet Autoantibodies and Risk of Progression to Diabetes in Children. Journal of the American Medical Association, 2013. 309(23): p. 2473-9.
  29. DPT-1 Study Group, Demographics of relatives screened and ICA positive in the diabetes prevention trial-type 1 diabetes (DPT-1). Diabetes, 1997. 46 (Suppl 1): p. 142A.
  30. Sosenko, J., et al., Use of the Diabetes Prevention Trial- Type 1 Risk Score (DPTRS) for improving the accuracy of the risk classification of type 1 diabetes. Diabetes Care, 2014. 37(4): p. 979-984.
  31. Mahon, J., et al., The TrialNet Natural History Study of the Development of Type 1 Diabetes: Objectives, design, and initial results. Pediatr Diabetes, 2009. 10(2): p. 97-104.
  32. Winkler, C., et al., Markedly reduced rate of diabetic ketoacidosis at onset of type 1 diabetes in relatives screened for islet autoantibodies. Pediatric Diabetes, 2012. 13(4): p. 308-313.
  33. Triolo, T., et al., Diabetic subjects diagnosed through the Diabetes Prevention Trial-Type 1 (DPT-1) are often asymptomatic with normal A1C at diabetes onset. Diabetes Care, 2009. 32: p. 769-773.
  34. Elding Larsson, H., et al., Reduced prevalence of diabetic ketoacidosis at diagnosis of type 1 diabetes in young children participating in longitudinal follow-up. Diabetes Care, 2011. 34(11): p. 2347-52.
  35. Rewers, A., et al., Presence of diabetic ketoacidosis at diagnosis of diabetes mellitus in youth: the Search for Diabetes in Youth Study. Pediatrics, 2008. 121: p. e1258-e1266.
  36. Barker, J., et al., Clinical characteristics of children diagnosed with type 1 diabetes through intensive screening and follow-up. Diabetes Care, 2004. 27(6): p. 1399-404.
  37. American Diabetes Association, I., American Diabetes Association Standards of Medical Care in Diabetes- 2021. Diabetes Care, 2021. 44 (Supplement 1): p. S18.
  38. General Population Level Estimation for Type 1 Diabetes Risk in Children 0-5 Years Old During Routine Care Delivery (PLEDGE). 2021 October 27, 2020 (accessed 2021 Dec 14); Available from: https://clinicaltrials.gov/ct2/show/NCT04477928?term=PLEDGE+study&cond=t1d&draw=2&rank=1.
  39. Rewers, M., et al., Newborn screening for HLA markers associated with IDDM: diabetes autoimmunity study in the young (DAISY). Diabetologia, 1996. 39(7): p. 807-12.
  40. Munchen, T.U., GPPAD-POInT (Global Platform of Autoimmune Diabetes - Primary Oral Insulin Trial).
  41. Barbara Davis Center for Diabetes, Autoimmunity Screening for Kids. 2019, March 13 (accessed 2021 Dec 14); Available from: https://www.askhealth.org/.
  42. Puff, R., et al., (Early diagnosis, early care--"Fr1da" screening of children for type 1 diabetes). MMW Fortschr Med, 2016. 158(4): p. 65-6.
  43. Megan AS Penno, J.J.C., Maria E Craig, et al. ENDIA Study Group, Environmental determinants of islet autoimmunity (ENDIA): a pregnancy to early life cohort study in children at-risk of type 1 diabetes, BMC Pediatr, 2013. 13-124: p. 1471-2431.
  44. Cardwell, C., et al., Maternal age at birth and childhood type 1 diabetes: a pooled analysis of 30 observational studies. Diabetes, 2010. 59(2): p. 486-494.
  45. Flood, T., S. Brink, and R. Gleason, Increased incidence of type 1 diabetes in children of older mothers. Diabetes Care, 1982. 5(6): p. 571-573.
  46. Warram, J.H., B.C. Martin, and A.S. Krolewski, Risk of IDDM in children of diabetic mothers decreases with increasing maternal age at pregnancy. Diabetes, 1991. 40(12): p. 1679-1684.
  47. Cardwell, C., et al., Birthweight and the risk of childhood-onset type 1 diabetes: a meta-analysis of observational studies using individual patient data. Diabetologia, 2010. 53(4): p. 641-651.
  48. Stene, L., et al., Birth weight and childhood onset type 1 diabetes: population-based cohort study. BMJ, 2001. 322(7291): p. 889-892.
  49. Dahlquist, G., S. Bennich, and B. Kallen, Intrauterine growth pattern and risk of childhood onset insulin dependent (type 1) diabetes: population based case-control study. BMJ, 1996. 313(7066): p. 1174-1177.
  50. Harder, T., et al., Birth weight, early weight gain, and subsequent risk of type 1 diabetes: systematic review and meta-analysis. Am J Epidemiol, 2009. 169(12): p. 1428-1436.
  51. Robertson, L. and K. Harrild, Maternal and neonatal risk factors for childhood type 1 diabetes: a Matched case-control study. BMC Public Health, 2010. 10: p. 281.
  52. Cardwell, C., et al., Birth order and childhood type 1 diabetes risk: a pooled analysis of 31 observational studies. Int J Epidemiol, 2011. 40(2): p. 363-374.
  53. Virtanen, S.M., et al., Microbial exposure in infancy and subsequent appearance of type 1 diabetes mellitus-associated autoantibodies: a cohort study. JAMA Pediatr, 2014. 168(8): p. 755-63.
  54. Cardwell, C., et al., Interbirth interval is associated with childhood type 1 diabetes risk. Diabetes, 2012. 61(3): p. 702-707.
  55. Cardwell, C., et al., Caesarean section is associated with an increased risk of childhood-onset type 1 diabetes mellitus: a meta-analysis of observational studies. Diabetologia, 2008. 51(5): p. 726-735.
  56. Khashan, A., et al., Mode of obstetrical delivery and type 1 diabetes: a sibling design study. Pediatrics, 2014. 134(3): p. e806-e813.
  57. Dahlquist, G. and B. Kallen, Maternal-child blood goup incompatibility and other perinatal events increase the risk for early-onset type 1 (insulin-dependent) diabetes mellitus. Diabetologia, 1992. 35(7): p. 671-675.
  58. Zhang, L., et al., Preterm birth and risk of type 1 and type 2 diabetes: systematic review and meta-analysis. Obes. Rev., 2014. 15(10): p. 804-811.
  59. Granfors, M., et al., No association between use of multivitamin supplement containing vitamin D during pregnancy and risk of Type 1 Diabetes in the child. Pediatr Diabetes, 2016. 17(7): p. 525-530.
  60. Stene, L.C. and G. Joner, Use of cod liver oil during the first year of life is associated with lower risk of childhood-onset type 1 diabetes: a large, population-based, case-control study. Am. J. Clin. Nutr, 2003. 78(6): p. 1128-1134.
  61. Fronczak, C., et al., In utero dietary exposures and risk of islet autoimmunity in children. Diabetes Care, 2003. 26(12): p. 3237-3242.
  62. Marjamaki, L., et al., Maternal intake of vitamin D during pregnancy and risk of advanced beta cell autoimmunity and type 1 diabetes in offspring. Diabetologia, 2010. 53(8): p. 1599-607.
  63. Kikkinen, A., et al., Use of antimicrobials and risk of type 1 diabetes in a population-based mother-child cohort. Diabetalogia, 2006. 49(1): p. 66-70.
  64. Arkkola, T., et al., Relationship of maternal weight status and weight gain rate during pregnancy to the development of advanced beta cell autoimmunity in the offspring: a prospective birth cohort study. Pediatr Diabetes, 2011. 12(5): p. 478-84.
  65. Niinisto, S., et al., Fatty acid status in infancy is associated with the risk of type 1 diabetes-associated autoimmunity. Diabetologia, 2017. 60(7): p. 1223-1233.
  66. Sorenson, I., et al., Serum long chain n-3 fatty acids (EPA and DHA) in the pregnant mother are independent of tisk of type 1 diabetes in the offspring. Diabetes Metab Res Rev, 2012. 28(5): p. 431-438.
  67. Ross, A., et al., The 2011 report on dietary reference intakes for calcium and vitamin D from the Institute of Medicine: what clinicians need to know. J Clin Endocrinol Metab, 2011. 96(1): p. 53-38.
  68. Vehik. Kendra; Lynch, K.W., Matthew; et al.TEDDY Study Group, Prospective virome analyses in young children at increased genetic risk for type 1 diabetes. Nature Medicine, 2019. 12.
  69. Borch-Johnsen, K., et al., Relation between breast-feeding and incidence rates of insulin-dependent diabetes mellitus: A hypothesis. Lancet, 1984. 2(8411): p. 1083-1086.
  70. Chimel, R., et al., Early infant feeding and risk of developing islet autoimmunity and type 1 diabetes. Acta Diabetol, 2014.
  71. Frederiksen, B., et al., Infant exposures and development of type 1 diabetes mellitus: The Diabetes Autoimmunity Study in the Young (DAISY). JAMA Pediatr, 2013. 167(9): p. 808-815.
  72. Lund-Blix, N., et al., Infant feeding in relation to islet autoimmunity and type 1 diabetes in genetically susceptible children: the MIDIA Study. Diabetes Care, 2015. 38(2): p. 257-263.
  73. Couper, J.J., et al., Lack of association between duration of breast-feeding or introduction of cow's milk and development of islet autoimmunity. Diabetes, 1999. 48(11): p. 2145-2149.
  74. Knip, M., et al., Hydrolyzed infant formula and early beta-cell autoimmunity: a randomized clinical trial. JAMA, 2014. 311(22): p. 2279-87.
  75. Knip, M., Writing group for teh TRIGR Study Group, Effect of Hydrolyzed Infant Formula vs Conventional Formula on Risk of Type 1 Diabetes. JAMA, 2018 Jan. 319(1): p. 38-48.
  76. Vaarala, O., et al., Removal of Bovine Insulin From Cow's Milk Formula and Early Initiation of Beta-Cell Autoimmunity in the FINDIA Pilot Study. Arch Pediatr Adolesc Med, 2012. 166(7): p. 608-14.
  77. Ziegler, A.G., et al., Early infant feeding and risk of developing type 1 diabetes-associated autoantibodies. JAMA, 2003. 290(13): p. 1721-1728.
  78. Norris, J.M., et al., Timing of initial cereal exposure in infancy and risk of islet autoimmunity. JAMA, 2003. 290(13): p. 1713-1720.
  79. Hummel S, P.M., Hummel M, Primary dietary intervention study to reduce the risk of islet autoimmunity in children at increased risk for type 1 diabetes: the BABYDIET study. Diabetes Care, 2011. 34(6): p. 1302-1305.
  80. Beyerlein, A., et al., Timing of gluten introduction and islet autoimmunity in young children: updated results from the BABYDIET study. Diabetes Care, 2014. 37(9): p. e194-e195.
  81. Brekke, H. and J. Ludvigsson, Vitamin D supplementation and diabetes-related autoimmunity in the ABIS study. Pediatr Diabetes, 2007. 8(1): p. 11-14.
  82. Vitamin D supplement in early childhood and risk for Type I (insulin-dependent) diabetes mellitus. The EURODIAB Substudy 2 Study Group. Diabetologia, 1999. 42(1): p. 51-54.
  83. Hypponen, E., et al., Intake of vitamin D and risk of type 1 diabetes: a birth-cohort study. Lancet, 2001. 358(9292): p. 1500-1503.
  84. Zipitis, C. and A. Akobeng, Vitamin D supplementation in early childhood and risk of type 1 diabetes: a systematic review and meta-analysis. Arch Dis Child, 2008. 93(6): p. 512-517.
  85. Simpson, M., et al., No association of vitamin D intake or 25-hydroxyvitamin D levels in childhood with risk of islet autoimmunity and type 1 diabetes: the Diabetes Autoimmunity Study in the Young (DAISY). Diabetologia, 2011. 54(11): p. 2779-2788.
  86. Norris, J., et al., Omega-3 polyunsaturated fatty acid intake and islet autoimmunity in children at increased risk for type 1 diabetes. JAMA, 2007. 298(12): p. 1420-1428.
  87. Chase, H., et al., Effect of docosahexaenoic acid supplementation on inflammatory cytokine levels in infants at high risk for type 1 diabetes. Pediatr Diabetes, 2014.
  88. Wagner, C. and F. Greer, Prevention of rickets and vitamin D deficiency in infants, children, and adolescents. Pediatrics, 2008. 122(5): p. 1142-1152.
  89. Strachan, D.P., Hay fever, hygiene, and household size. BMJ, 1989. 299(6710): p. 1259-1260.
  90. Karelia map. 2015; (accessed 2021 Dec 14) Available from: https://commons.wikimedia.org/wiki/File:Karelia_today.png#/media/File:Karelia_today.png.
  91. Kondrashova, A., et al., The 'Hygiene hypothesis' and the sharp gradient in the incidence of autoimmune and allergic diseases between Russian Karelia and Finland. APMIS, 2013. 121(6): p. 478-93.
  92. Seiskari, T., et al., Allergic senstization and microbial load--a comparison between Finland and Russian Karelia. Clin Exp Immunol, 2007. 148(1): p. 47-52.
  93. Yatsunenko, T., et al., Human gut microbiome viewed across age and geography. Nature, 2012. 486(7402): p. 222-227.
  94. Giongo, A., et al., Toward defining the autoimmune microbiome for type 1 diabetes. ISME J, 2011. 5(1): p. 82-91.
  95. Brown, C.T., et al., Gut microbiome metagenomics analysis suggests a functional model for the development of autoimmunity for type 1 diabetes. PLoS One, 2011. 6(10): p. e25792.
  96. de Goffau, M.C., et al., Fecal microbiota composition differs between children with beta-cell autoimmunity and those without. Diabetes, 2013. 62(4): p. 1238-44.
  97. Bezirtzoglou, E., A. Tsiotsias, and G.W. Welling, Microbiota profile in feces of breast- and formula-fed newborns by using flourescence in situ hybridization (FISH). Anaerobe, 2011. 2011(17): p. 6.
  98. Stark, P. and A. Lee, The microbial ecology of the large bowel of breast-fed and formula-fed infants during the first year of life. J Med Microbiol, 1982. 15(2): p. 189-203.
  99. Menser, M., J. Forrest, and R. Bransby, Rubella infection and diabetes mellitus. Lancet, 1978. 1: p. 57-60.
  100. Oikarinen, S., et al., Enterovirus RNA in blood is linked to the development of type 1 diabetes. Diabetes, 2011. 60(1): p. 276-9.
  101. Kimpimaki, T., et al., The first signs of beta-cell autoimmunity appear in infancy in genetically susceptible children from the general population: the Finnish Type 1 Diabetes Prediction and Prevention Study. J Clin Endocrinol Metab, 2001. 86(10): p. 4782-8.
  102. Moltchanova, E., et al., Seasonal variation of diagnosis of type 1 diabetes mellitus in children worldwide. Diabet Med, 2009. 26(7): p. 673-678.
  103. Duderstadt, S., et al., Vaccination and risk of type 1 diabetes mellitus in active component U.S. military, 2002-2008. Vaccine, 2012. 30(4): p. 813-819.
  104. Graves, P., et al., Lack of association between early childhood immunizations and beta-cell autoimmunity. Diabetes Care, 1999. 22(10): p. 1694-1697.
  105. DeStefano, F., et al., Childhood vaccinations, vaccination timing, and risk of type 1 diabetes mellitus. Pediatrics, 2001. 108(6): p. E112.
  106. Hviid, A., et al., Childhood vaccination and type 1 diabetes. N Engl J Med, 2004. 350(14): p. 1398-1404.
  107. Elding Larsson, H., et al., Pandemrix(R) vaccination is not associated with increased risk of islet autoimmunity or type 1 diabetes in the TEDDY study children. Diabetologia, 2018. 61(1): p. 193-202.
  108. Breastfeeding and the use of human milk. Pediatrics, 2012. 129(3): p. e827-e841.
  109. Bonifacio, E., et al., Effects of high-dose oral insulin on immune responses in children at high risk for type 1 diabetes: the Pre-POINT randomized clinical trial. JAMA, 2015. 313(15): p. 1541-9.
  110. Skyler JS, K.J., Wolfsdorf J et al., Effects of oral insulin in relatives of patients with type 1 diabetes: The Diabetes Prevention Trial--Type 1. Diabetes Care, 2005. 28(5).
  111. Skyler, J., Update on worldwide efforts to prevent type 1 diabetes. Ann N Y Acad Sci, 2008. 1150: p. 190.
  112. Vehik, K., et al., Long-term outcome of individuals treated with oral insulin: diabetes prevention trial-type 1 (DPT-1) oral insulin trial. Diabetes Care, 2011. 34(7): p. 1585-1590.
  113. Writing Committee for the Type 1 Diabetes TrialNet Oral Insulin Study, G., et al., Effect of Oral Insulin on Prevention of Diabetes in Relatives of Patients With Type 1 Diabetes: A Randomized Clinical Trial. JAMA, 2017. 318(19): p. 1891-1902.
  114. Greenbaum, C.J., et al., Parenteral insulin suppresses T cell proliferation to islet antigens. Pediatr Diabetes, 2011. 12(3 Pt 1): p. 150-5.
  115. Nanto-Salonen, K., et al., Nasal insulin to prevent type 1 diabetes in children with HLA genotypes and autoantibodies conferring increased risk of disease: a double-blind, randomised controlled trial. Lancet, 2008. 372(9651): p. 1746-55.
  116. Health, M., Trial of Intranasal Insulin in Children and Young Adults at Risk of Type 1 Diabetes (INITII). ClinicalTrialsgov (Internet) Bethesda (MD): National Library of Medicine (US) 2000- (accessed 2021 Dec 14) Available from: http://clinicaltrials.gov/show/NCT00336674 NLM Identifier: NCT00336674.
  117. Team, P.E. ADC 2018: Update on the Intranasal Insulin Trial II (INIT II) to Prevent Type 1 Diabetes. 2018(accessed 2021 Dec 14); Available from: https://www.practiceupdate.com/content/adc-2018-update-on-the-intranasal-insulin-trial-ii-init-ii-to-prevent-type-1-diabetes/73021#:~:text=The%20estimated%205%2Dyear%20risk,people%20(38%25)%20developed%20diabetes.
  118. Clinicaltrials.gov. A multiple ascending dose trial investigating safety, tolerability and pharmacokinetics of NNC0361-0041 (TOPPLE T1D); NCT04279613. 2020 (accessed 2021 Dec 14); Available from: https://clinicaltrials.gov/ct2/show/NCT04279613?term=TOPPLE&draw=2&rank=1.
  119. ClinicalTrials.gov. A study to assess the safety and tolerability of different doses of AG019 administered alone or in combination with teplizumab in participants with recently diagnosed type 1 diabetes mellitus (T1D); NCT03751007, 2018 (accessed 2021 Dec 14); Available from: https://clinicaltrials.gov/ct2/show/NCT03751007?term=AG019&draw=2&rank=1.
  120. Clinicaltrials.gov. CTLA4-Ig (Abatacept)for Prevention of Abnormal Glucose Tolerance and Diabetes in Relatives At -Risk for Type 1; NCT01773707. 2017; Available from: https://clinicaltrials.gov/ct2/show/NCT01773707?term=NCT01773707&draw=2&rank=1.
  121. Herold, K.C., et al., An Anti-CD3 Antibody, Teplizumab, in Relatives at Risk for Type 1 Diabetes. N Engl J Med, 2019. 381(7): p. 603-613.
  122. Sims, E.K., et al., Teplizumab improves and stabilizes beta cell function in antibody-positive high-risk individuals. Science Translational Medicine, 2021. 13(583).
  123. Greenbaum, C.J., et al., Preservation of beta-cell function in autoantibody-positive youth with diabetes. Diabetes Care, 2009. 32(10): p. 1839-44.
  124. Davis, A.K., et al., Prevalence of detectable C-Peptide according to age at diagnosis and duration of type 1 diabetes. Diabetes Care, 2015. 38(3): p. 476-81.
  125. DCCT Group, The effect of intensive treatment of diabetes on the development and progression of long-term complications in insulin-dependent diabetes mellitus. The Diabetes Control and Complications Trial Research Group. N. Engl. J. Med, 1993. 329(14): p. 977-986.
  126. Nathan, D.M., et al., Intensive diabetes treatment and cardiovascular disease in patients with type 1 diabetes. N Engl J Med, 2005. 353(25): p. 2643-2653.
  127. Steffes, M.W., et al., Beta-cell function and the development of diabetes-related complications in the diabetes control and complications trial. Diabetes Care, 2003. 26(3): p. 832-836.
  128. Lachin, J.M., et al., Impact of C-peptide preservation on metabolic and clinical outcomes in the Diabetes Control and Complications Trial. Diabetes, 2014. 63(2): p. 739-48.
  129. Jeyam, A., et al., Clinical Impact of Residual C-Peptide Secretion in Type 1 Diabetes on Glycemia and Microvascular Complications. Diabetes Care, 2021. 44(2): p. 390-398.
  130. Chow, L., et al., Biomarkers related to severe hypoglycemia and lack of good glycaemic control in ACCORD. Diabetologia, 2015. 58(6): p. 1160-1166.
  131. Vantyghem, M., et al., Continuous glucose monitoring after islet transplantation in type 1 diabetes: An excellent graft function (B-score greater than 7) is required to abrogate severe hypoglycemia, whereas minimal function is necessary to suppress severe hypoglycemia (B-score greater than 3). J Clin Endocrinol Metab, 2012. 97(11): p. E2078-E2083.
  132. Barton, F.B., et al., Improvement in outcomes of clinical islet transplantation: 1999-2010. Diabetes Care, 2012. 35(7): p. 1436-1445.
  133. Palmer, J.P., et al., C-peptide is the appropriate outcome measure for type 1 diabetes clinical trials to preserve beta-cell function: report of an ADA workshop, 21-22 October 2001. Diabetes, 2004. 53(1): p. 250-264.
  134. Bougneres, P.F., et al., Factors associated with early remission of type I diabetes in children treated with cyclosporine. N Engl J Med, 1988. 318(11): p. 663-70.
  135. D'Addio F, V.V., Ben NM et al., Autologous nonmyeloablative hematopoietic stem cell transplantation in new-onset type 1 diabetes: a multicenter analysis. Diabetes, 2014. 63(9).
  136. Keymeulen, B., et al., Insulin needs after CD3-antibody therapy in new-onset type 1 diabetes. N. Engl. J. Med, 2005. 352(25): p. 2598-2608.
  137. Keymeulen, B., et al., Four-year metabolic outcome of a randomised controlled CD3-antibody trial in recent-onset type 1 diabetic patients depends on their age and baseline residual beta cell mass. Diabetologia, 2010. 53(4): p. 614-623.
  138. Herold, K.C., et al., Anti-CD3 monoclonal antibody in new-onset type 1 diabetes mellitus. N. Engl. J. Med, 2002. 346(22): p. 1692-1698.
  139. Herold, K.C., et al., A single course of anti-CD3 monoclonal antibody hOKT3gamma1(Ala-Ala) results in improvement in C-peptide responses and clinical parameters for at least 2 years after onset of type 1 diabetes. Diabetes, 2005. 54(6): p. 1763-1769.
  140. Herold, K.C., et al., Teplizumab (anti-CD3 mAb) treatment preserves C-peptide responses in patients with new-onset type 1 diabetes in a randomized controlled trial: metabolic and immunologic features at baseline identify a subgroup of responders. Diabetes, 2013. 62(11): p. 3766-74.
  141. Sherry, N., et al., Teplizumab for treatment of type 1 diabetes (Protege study): 1-year results from a randomised, placebo-controlled trial. Lancet, 2011. 378(9790): p. 487-97.
  142. Pescovitz, M.D., et al., Rituximab, B-Lymphocyte Depletion, and Preservation of Beta-Cell Function. N. Engl. J. Med, 2009. 361(22): p. 2143-2152.
  143. Haller, M.J., et al., Low-Dose Anti-Thymocyte Globulin Preserves C-Peptide, Reduces HbA1c, and Increases Regulatory to Conventional T-Cell Ratios in New-Onset Type 1 Diabetes: Two-Year Clinical Trial Data. Diabetes, 2019. 68(6): p. 1267-1276.
  144. Orban, T., et al., Co-stimulation modulation with abatacept in patients with recent-onset type 1 diabetes: a randomised, double-blind, placebo-controlled trial. Lancet, 2011. 378(9789): p. 412-9.
  145. Orban, T., et al., Costimulation modulation with abatacept in patients with recent-onset type 1 diabetes: follow-up 1 year after cessation of treatment. Diabetes Care, 2014. 37(4): p. 1069-75.
  146. Rigby, M.R., et al., Targeting of memory T cells with alefacept in new-onset type 1 diabetes (T1DAL study): 12 month results of a randomised, double-blind, placebo-controlled phase 2 trial. Lancet Diabetes Endocrinol, 2013. 1(4): p. 284-94.
  147. Rigby, M.R., et al., Alefacept provides sustained clinical and immunological effects in new-onset type 1 diabetes patients. J Clin Invest, 2015. 125(8): p. 3285-96.
  148. Moran, A., et al., Interleukin-1 antagonism in type 1 diabetes of recent onset: two multicentre, randomised, double-blind, placebo-controlled trials. Lancet, 2013. 381(9881): p. 1905-15.
  149. Long, S.A., et al., Rapamycin/IL-2 combination therapy in patients with type 1 diabetes augments Tregs yet transiently impairs beta-cell function. Diabetes, 2012. 61(9): p. 2340-8.
  150. Greenbaum, C., Sanda, S for the ITN058AI EXTEND Study Team, IL-6 receptor blockade does not slow β cell loss in new-onset type 1 diabetes. JCI Insight, 2021. 6(21).
  151. Hartemann A, e.a., Low-dose interleukin 2 in patients with type 1 diabetes: a phase 1/2 randomised, double-blind, placebo-controlled trial. Lancet Diabetes Endocrinol. 2013 Dec;1(4):295-305, 2013.
  152. Clinicaltrials.gov. Pilot Clinical Trial of Ustekinumab in Patients With New-onset T1D (USTID); NCT02117765. 2016; (accessed 2021 Dec 2014) Available from: https://clinicaltrials.gov/ct2/show/NCT02117765.
  153. Clinicaltrials.gov. Clinical phase II/III trial of ustekinumab to treat type 1 diabetes (UST1D2) (UST1D2); NCT03941132. 2019; (accessed 2021 Dec 14) Available from: https://clinicaltrials.gov/ct2/show/NCT03941132?term=NCT03941132&draw=2&rank=1
  154. Quattrin, T.H., M.J., Steck, A.K., Felner, E.I., Li, Y., Xia, Y., Leu, ,J.H., Zoka, R., Hedrick, J.A., Rigby, M.R., Vercruysse, F.; T1GER Study Investigators Golimumab and beta-cell function in youth with new-onset type 1 diabetes. NEJM, 2020. 383(21): p. 2007-2017.
  155. von Herrath, M., et al., Anti-interleukin-21 antibody and liraglutide for the preservation of beta-cell function in adults with recent-onset type 1 diabetes: a randomised, double-blind, placebo-controlled, phase 2 trial. Lancet Diabetes Endocrinol, 2021. 9(4): p. 212-224.
  156. Giannoukakis, N., et al., Phase I (safety) study of autologous tolerogenic dendritic cells in type 1 diabetic patients. Diabetes Care, 2011. 34(9): p. 2026-2032.
  157. Carlsson, P., et al., Preserved beta-cell function in type 1 diabetes by mesenchymal stromal cells. Diabetes, 2015. 64(2): p. 587-592.
  158. Haller, M., et al., Autologous umbilical cord blood transfusion in young children with type 1 diabetes fails to preserve c-peptide. Diabetes Care, 2011. 34(12): p. 2567-2569.
  159. Marek-Trzonkowska, N., et al., Administration of CD4+CD25highC. Diabetes Care, 2012. 35(9): p. 1817-1820.
  160. Marek-Trzonkowska, N., et al., Therapy of type 1 diabetes with CD4(+)CD25(high)CD127-regulatory T cells prolongs survival of pancreatic islets - results of one year follow-up Clin Immunol, 2014. 153(1): p. 23-30.
  161. Gottlieb PA, A.A., Michels AW α1−Antitrypsin therapy downregulates toll-like receptor-induced IL-1β responses in monocytes and myeloid dendritic cells and may improve islet function in recently diagnosed patients with type 1 diabetes. J Clin Endocrinol Metab, 2014. 99: p. E1418–E1426.
  162. Allen, H.F., et al., Effect of Bacillus Calmette-Guerin vaccination on new-onset type 1 diabetes. A randomized clinical study. Diabetes Care, 1999. 22(10): p. 1703-1707.
  163. Louvet C1, S.G., Lang J, Lee MR, Martinier N, Bollag G, Zhu S, Weiss A, Bluestone JA, Tyrosine kinase inhibitors reverse type 1 diabetes in nonobese diabetic mice. Proc Natl Acad Sci U S A, 2008. 105(48): p. 18895-900.
  164. Hagerkvist, R., et al., Amelioration of diabetes by imatinib mesylate (Gleevec): Role of beta-cell NF-kappaB activation and anti-apoptotic preconditioning. FASEB J, 2007. 21(2): p. 618-628.
  165. Gitelman, S., et al. , Imatinib therapy for patients with recent-onset type 1 diabetes: a multicentre, randomised, double-blind, placebo-controlled, phase 2 trial. Lancet Diabetes and Endocrinology, 2021. 9(8): p. 502-514.
  166. Pescovitz, M.D., et al., Rituximab, B-lymphocyte depletion, and preservation of beta-cell function. N Engl J Med, 2009. 361(22): p. 2143-52.
  167. Hagopian, W., et al., Teplizumab preserves C-peptide in recent-onset type 1 diabetes: two-year results from the randomized, placebo-controlled Protege trial. Diabetes, 2013. 62(11): p. 3901-8.
  168. Greenbaum, C.J., et al., Fall in C-peptide during first 2 years from diagnosis: evidence of at least two distinct phases from composite Type 1 Diabetes TrialNet data. Diabetes, 2012. 61(8): p. 2066-73.
  169. Wherrett, D.K., et al., Defining pathways for development of disease-modifying therapies in children with type 1 diabetes: a consensus report. Diabetes Care, 2015. 38(10): p. 1975-85.
  170. Keenan, H.A., et al., Residual insulin production and pancreatic ss-cell turnover after 50 years of diabetes: Joslin Medalist Study. Diabetes, 2010. 59(11): p. 2846-53.
  171. Dabelea, D., et al., Clinical evolution of beta cell function in youth with diabetes: the SEARCH for Diabetes in Youth study. Diabetologia, 2012. 55(12): p. 3359-68.
  172. Gianani, R., et al., Dimorphic histopathology of long-standing childhood-onset diabetes. Diabetologia, 2010. 53: p. 690-698.
  173. Choudhary, P., et al., Evidence-informed clinical practice recommendations for treatment of type 1 diabetes complicated by problematic hypoglycemia. Diabetes Care, 2015. 38(6): p. 1016-1029.

 

Risk of Fasting and Non-Fasting Hypertriglyceridemia in Coronary Vascular Disease and Pancreatitis

ABSTRACT

 

Cardiovascular disease (CVD) remains a major cause of mortality in the Western world and in spite of the reduction of CVD risk by the use of lipid lowering agents per current treatment goals there remains substantial residual and absolute risk of CVD in high-risk populations. Focus on elevated triglyceride (TG) levels deserves renewed attention, particularly as one-third of all adults in the United States suffer from elevated TG and a growing number of people are diagnosed with metabolic syndrome or type 2 diabetes mellitus (T2DM). The dyslipidemia of metabolic syndrome and T2DM is characterized by low high-density lipoprotein cholesterol (HDL-c) concentrations and marked elevations in triglyceride rich lipoproteins (TRL). There has been growing data that points towards an association of fasting and non-fasting triglycerides with CVD, including a number of genetic studies suggesting causality. However, the association of TG as an independent risk faster in CVD is confounded by its inverse metabolic relationship with high density lipoprotein (HDL-c) and the heterogeneity of TG lipoproteins. Current guidelines suggest diagnosis of hypertriglyceridemia based on fasting levels where length of fast is recommended to be 9-12 hours. Although non-fasting TG levels may be a better indicator of risk, the lack of standardization of non-fasting TG measurements, lack of specific reference ranges, and the variability of postprandial lipid measurements have hampered their routine clinical use. Current guidelines focus mainly on LDL-c levels; however, lowering TG may provide additional benefit for CVD prevention. Lifestyle changes including dietary changes and exercise play an important role in treatment of hyperlipidemia. Pharmacological agents used in treatment of hypertriglyceridemia include niacin, fibrates, fish oil and statins. Most guidelines recommend treating elevated TG for prevention of pancreatitis. This article will discuss the role of elevated TG in pancreatitis and CVD risk.

 

INTRODUCTION

 

Cardiovascular disease (CVD) remains a major cause of mortality in the Western world, especially in individuals with obesity, metabolic syndrome, and type 2 diabetes mellitus (T2DM). With increasing incidence of metabolic syndrome and T2DM worldwide, the global burden of CVD will also increase (1). In spite of the reduction of CVD risk by 25-35% with the use of lipid lowering agents, especially statins, there remains substantial residual and absolute risk of CVD in high-risk populations such as T2DM (2). Elevated low-density lipoprotein (LDL-c) is a well-established CVD risk factor and has been the primary target for lipid lowering treatment. However, growing evidence suggests that an elevated triglyceride (TG) level is also an independent risk factor (34)  Borderline high TGs or high TGs defined by National Cholesterol Education Program (NCEP) Adult Treatment Panel III (ATP III) as TG concentration (150 – 199 mg/dl) and (200 – 499 mg/dl) respectively are present in 30% of the US adult population (5) and these levels have been associated with increased CVD. The dyslipidemia of metabolic syndrome and T2DM is characterized by raised TG concentrations, low high-density lipoprotein cholesterol (HDL-c) concentrations and marked elevations in triglyceride rich lipoproteins (TRL). This triad is termed mixed or atherogenic dyslipidemia (6). Impaired metabolism of TRLs in the postprandial state have been observed in insulin resistant states such as visceral obesity, metabolic syndrome, and T2DM and this has been linked to the development of atherosclerosis (7).

 

Severe hypertriglyceridemia and very severe hyperlipidemia defined by Endocrine Society Clinical Practice Guideline on Evaluation and Treatment of Hypertriglyceridemia as TG concentration (1000 – 1999 mg/dl) and (>2000 mg/dl) carries an increased risk for pancreatitis (8). Case series and uncontrolled studies have shown that severely elevated TG levels are associated with the chylomicronemia syndrome and an increased risk of pancreatitis. Serum TG levels of 1000 mg/dl and higher have been observed in 12% to 38% of patients with acute pancreatitis (9). Hence, understanding the role of hypertriglyceridemia in CVD, chylomicronemia syndrome and risk of pancreatitis is important.

 

METABOLISM OF TRIGLYCERIDE RICH LIPOPROTEINS AND TRIGLYCERIDEMIA

 

Triglyceride Rich Lipoprotein Metabolism

 

Triglyceride rich lipoproteins (TRL) consist of chylomicrons carrying triglycerides from the diet, VLDLs synthesized in the liver, and their respective remnant particles. After a fatty meal, dietary triglycerides are hydrolyzed in the intestine to free fatty acids and monoglycerides. Fatty acids and monoglycerides are then absorbed by enterocytes and resynthesized to form triglycerides. Triglycerides within the intestinal enterocytes are assembled with apolipoprotein (apo) B-48 into large chylomicrons which are released from the cells into the lymphatic system. They access the plasma via the thoracic duct and are rapidly metabolized by lipoprotein lipase (LPL) to yield chylomicron remnants. These are taken up by remnant receptors and by LDL receptors in the liver. Free fatty acids liberated by the action of LPL are available to adipose tissue for storage and to other tissues (e.g., skeletal muscle, heart) for use as energy substrates. Lipids derived from chylomicron remnants, from de novo lipid synthesis, and from lipolysis of adipose tissue are reassembled in the liver as very-low-density lipoprotein (VLDL) particles, which are secreted into the plasma. VLDL particles are metabolized by LPL to yield intermediate density lipoprotein (IDL) particles, which are metabolized by LPL and hepatic lipase to yield low density lipoprotein (LDL) particles. IDL can be taken up by the liver through an apo E-dependent process, and LDL is taken up by the liver through the binding of apoB100 to LDL receptors. Small VLDL particles, IDL particles, and LDL particles may be taken up by peripheral tissues to deliver nutrients, cholesterol, and fat-soluble vitamins (1011). For more details see (12).

 

Hypertriglyceridemia 

 

Hypertriglyceridemia is a normal physiological state that occurs post ingestion of a meal where lipids undergo the above-mentioned metabolism. In insulin- resistant states there is an exaggerated lipid response leading to pathological hypertriglyceridemia which is thought to be atherogenic. In insulin-resistant states there is an increase in the production of VLDL by the liver and decreased hepatic uptake of VLDL, IDL and LDL. There is a reduction in LPL activity resulting in high triglyceride concentrations, especially in the postprandial state. The over secretion of VLDL, which competes with chylomicron remnants for clearance through the common pathway, can exacerbate the post prandial response. The large amount of TRLs and their prolonged residence time in the circulation leads to increased exchange of cholesteryl ester for triglycerides between TRL and LDL or HDL particles mediated by cholesteryl ester transfer protein (CETP). This process enriches LDL and HDL with triglyceride, and these particles are subsequently more readily hydrolyzed by hepatic lipase resulting in smaller, denser LDL particles and lower concentrations of HDL (13). These abnormalities result in a characteristic dyslipidemia in insulin resistant states, which is now recognized to be atherogenic.

 

Apo C-III

 

Apolipoprotein C-III (APOC3) has a key role in lipoprotein metabolism and regulation of triglyceride levels. APOC3 is synthesized in the liver and transported on triglyceride-rich lipoproteins. It inhibits LPL mediated hydrolysis of triglycerides, and at high concentrations can also inhibit hepatic lipase activity. In addition, APOC3 impairs the hepatic uptake of triglyceride rich lipoproteins by remnant receptors. Thus increased APOC3 levels are an independent risk factor for CVD (14-16); genetic variants of APOC3 leading to lower levels are associated with reduced risk for CVD (17-19).

 

ANGPTL

 

Angiopoietin-like proteins (ANGPTL) are regulators of lipoprotein metabolism. ANGPT3 and ANGPTL4 are natural inhibitors of LPL. Loss of function variants in these proteins have been associated with decreased triglyceride levels and decreased CVD. Murine studies have found that suppression of ANGPTL3 decreases atherosclerosis (20).

 

Severe Hypertriglyceridemia 

 

In severe or very severe TG levels (> 1000 mg/dl), which occur as a result of defective lipolysis or excessive production of endogenous triglyceride, the LPL removal system is saturated. There is decreased degradation of dietary TGs incorporated into chylomicrons and rapid increase of TG levels post fat-rich meals (worsened by dietary simple sugars, fructose, and alcohol) in susceptible individuals causing pancreatitis. The mechanism by which hypertriglyceridemia causes pancreatitis is not understood, but could include local accumulation of free fatty acids and serum hyperviscosity (821).

 

HYPERTRIGLYCERIDEMIA AND RISK OF PANCREATITIS AND CHYLOMICRONEMIA SYNDROME

 

Severe hypertriglyceridemia (> 1000 mg/dl) is an infrequent laboratory finding and is generally associated with genetic disorders of lipid metabolism or TG levels exacerbated by secondary causes. Familial chylomicronemia is a rare monogenic disorder than can cause severe hypertriglyceridemia.(109) It is defined as presence of chylomicronemia (TG > 1000 mg/dl) along with one or more of the following: eruptive xanthomas, lipemia retinalis, or abdominal findings of pain, acute pancreatitis and/or hepatosplenomegaly (22). Multifactorial chylomicronemia syndrome (MFCS) which is a much more common cause of severe hypertriglyceridemia is caused by the accumulation of genetic, non-genetic and environmental factors (109). Individuals with severe hypertriglyceridemia may present with these classic findings and pancreatitis or may be asymptomatic. The mechanisms by which hypertriglyceridemia may lead to acute pancreatitis are not known. Possible mechanisms include intra-pancreatic hydrolysis of high triglycerides by pancreatic lipase leading to accumulation of fatty acids in the pancreas which may be toxic and lead to inflammation and ischemia. Another proposed mechanism is increased viscosity by high chylomicron levels leading to ischemia (23). A study looking at the frequency of signs and symptoms of hypertriglyceridemia including pancreatitis found that the incidence of pancreatitis and eruptive xanthomas was low unless TG levels were significantly elevated, e.g. > 1700 mg/dl (20 mmol/l); patients with extreme hypertriglyceridemia had a combination of primary and secondary factors (T2DM, obesity, alcohol intake, pregnancy) contributing to their high TG levels (24). Murphy et al. in their cohort study estimated the risk of pancreatitis with differing degrees of TG elevations and showed that the crude incidence of pancreatitis was 0.91 per 1000 person years (95% CI, 0.76 – 1.09) in individuals with TG levels <150 mg/dl, 1.24 (95% CI, 1.07 – 1.44) with TG levels 150 – 499 mg/dl and 2.48 (95% CI, 1.79 – 3.42) with TG levels >500 mg/dl (25). Increased incidence is seen with increased TG levels. The level of TG at which pancreatitis can be attributed to hypertriglyceridemia is not well defined nor is the level of TG reduction that is associated with reduced risk known. A study by Lindkist et al. (23) looked at the association of moderately elevated serum TG levels and acute pancreatitis. In this study, 33,260 individuals were followed for median 25.7 years where overall incidence of acute pancreatitis in the cohort was 35.5/100,000-person years. There was a statistically significant association between TG levels and risk of pancreatitis with adjusted HR for pancreatitis of 1.21 (95% CI, 1.07 – 1.36) per 1 mmol/l (~88.5 mg/dl) increment in TG and a significant increased risk for acute pancreatitis in individuals with TG levels > 1.64 mmol/l (145 mg/dl). The analysis in this study was restricted to individuals with TG levels < 6 mmol/l (530 mg/dl) producing statistically significant results and showing that TG levels much lower than previously believed can be associated with increased risk of acute pancreatitis. Another study evaluated the association between lower follow-up TG levels and the incidence of recurrent clinical events for patients with severe hypertriglyceridemia (>500 mg/dl). This study included 41,210 individuals with < 1% having a history of pancreatitis. Individuals with severe hypertriglyceridemia with follow up TG levels <200 mg/dl experienced a lower rate of recurrent pancreatitis episodes, with an adjusted rate ratio of 0.45 (95% CI, 0.34 – 0.60) compared to those with TG levels >500 mg/dl (26). There is an increased risk of pancreatitis with severe hypertriglyceridemia and in individuals with elevated dietary TG levels and in some cases pharmacological intervention is necessary to prevent severe complications such as pancreatitis.

 

HYPERTRIGLYCERIDEMIA AS A CARDIOVASCULAR RISK FACTOR

 

Epidemiological Data Supporting TG as a CVD Risk Factor

 

The association of elevated TG values with CVD remains controversial. Establishing TG level as an independent risk factor for CVD is confounded by its inverse metabolic relationship with HDL-c and the heterogeneity of TG risk lipoproteins. Growing evidence suggests that an elevated TG level is an independent risk factor for CVD and represents an important biomarker of CVD risk because of their association with atherogenic remnant particles. A meta-analysis by Hokanson and Austin (4), showed increased plasma TG levels are associated with a significant increase in risk of CVD independent of HDL-c level. An overall relative risk (RR) for CVD of 1.32 for men and 1.76 for women per 1 mmol/L (~88.5 mg/dl) increase in TGs was noted. However, this analysis was limited to Caucasian study subjects. A non-overlapping meta-analysis involving data from 26 prospective studies in Asian and Pacific populations reported a RR for CVD of 1.8 (95% CI, 1.49 – 2.19), comparing subjects in the top fifth with the bottom fifth of triglyceride levels (27). Sawar et al. (28) reported data from two prospective cohort studies: the Reykjavik study and the European Prospective Investigational of Cancer (EPIC) - Norfolk study, which together comprised 44,237 western middle-aged men and women and total of 3582 incident cases of CVD. Comparing individuals with TGs in the top tertile with the bottom tertile, the adjusted odds ratio for CVD was 1.76 (95% CI, 1.39 – 2.21) in the Reykjavik study and 1.57 (95% CI, 1.10 – 2.24) in the EPIC-Norfolk study. However, adjustment for HDL-c substantially attenuated the magnitude of association of TG level with CVD. They also performed an updated meta-analysis of the Western population studies adding information to include a total of >10,000 CVD cases from 29 Western prospective studies involving a total of > 260,000 participants, and report an adjusted odds ratio of 1.72 (95% CI, 1.56 – 1.90) comparing top and the bottom tertiles of TG values (28). A more recent meta-analysis by Murad, et al. (9), included 35 studies with total of 927,218 subjects who suffered 132,460 deaths and 72,654 cardiac events, myocardial infarctions or pancreatitis; with odds ratio of 1.80 (95% CI, 1.31 – 2.49) for cardiac events, 1.31 (95% CI, 1.15 – 1.49) for myocardial infarctions and 3.96 (95%, CI 1.27 – 12.34) for pancreatitis.

 

Genetic Data Linking TG to CVD

 

Recent human genetic studies show that elevated TGs and TRLs are causal risk factors for CVD. A Mendelian randomization study based on several genetic variants affecting remnant cholesterol and/or HDL showed that a 1 mmol/l (39 mg/dl) increase in non-fasting remnant cholesterol is associated with a 2.8-fold causal risk for ischemic heart disease, independent of reduced HDL cholesterol (29). A meta-analysis of 46 lipid genome-wide-association studies (GWAS) together comprising >100,000 individuals of European descent identified four novel loci associated with CVD that were related to HDL-c and TG levels suggesting elevated TG metabolism may also be associated with CVD risk (30). Another large Mendelian randomization study based on a single APOA5 variant (-1131T>C) that regulates TG showed an association with CVD risk. The odds ratio for coronary heart disease was 1·18 (95% CI 1·11–1·26; p=2·6×10−7) per C allele, which was concordant with the hazard ratio of 1·10 (95% CI 1·08–1·12) per 16% higher triglyceride concentration recorded in prospective studies (31). This finding is similar to that seen in the study by Jorgensen et al. (32), where doubling of genetically raised remnant cholesterol and TG levels due to APOA5 genetic variants was associated with an increased risk of myocardial infarctions. In addition, a study using individuals from the Copenhagen City Heart Study with genetic variants in lipoprotein lipase (LPL), tested whether low concentrations of non-fasting triglycerides were associated with reduced all-cause mortality in observational analyses (n = 13,957). The results showed that each genetically-derived 1 mmol/l (~88.5 mg/dl) reduction in TG levels was associated with a halved risk of all-cause mortality (33). Two large studies examining the relationship between the gene encoding apolipoprotein C3 (APOC3) found that loss of function mutations in APOC3 were associated with low levels of triglycerides and reduced risk of CVD (1719). ANGPTLs have also been linked to CVD. A large human study found that individuals with loss of function variants in ANGPTL3 had lower triglyceride levels, as well as lower levels of HDL-c and LDL-c compared to control subjects, and decreased odds of CVD (20). Similarly, individuals with loss of function variants of ANGPTL4 also had lower triglyceride levels and decreased risk for CVD (34). A study by Ference et al did a randomization analysis of a total of 654,783 participants to determine if there was any association with risk of CVD per unit of change in ApoB from either LDL-C lowering variants in the LDL receptor gene (LDLR) or triglyceride-lowering variants in the lipoprotein lipase gene.  It showed that there was a similar decrease in risk of CVD per unit difference in ApoB from either triglyceride-lowering LPL variants or LDL-C lowering LDLR variants. For each 10-mg/dL lower level of ApoB-containing lipoproteins, the LPL score was associated with 69.9-mg/dL lower plasma triglyceride levels while the same 10-mg/dL lower level of ApoB-containing lipoproteins, the LDLR score was associated with only a 14.2-mg/dL lower plasma LDL-C level and 1.9-mg/dL. This suggests that one will need to lower plasma triglyceride levels by a much greater degree to achieve the same benefits on cardiovascular disease as lowering LDL-C levels and could explain why studies of triglyceride lowering drugs have given ambiguous results. To be noted, the results from this study are from genetic variants and not lipid lowering therapies (106). These studies strongly point to a causal effect of elevated TG and TRLs with CVD.

 

Clinical Trial Evidence Supporting Lowering TG Reduces CVD

 

Results with clinical outcomes trials of fibrate therapy have been variable but primarily indicate a reduction in CV events. Post hoc analysis of several of these trials provides consistent evidence showing a clinical benefit in subgroups with elevated TG levels. A meta-analysis of the effect of TG lowering in 18 trials providing data for 45,058 participants showed that fibrate therapy produced a 10% RR reduction (95% CI 0-18) for major cardiovascular events (p=0.048) and a 13% RR reduction (95% CI 7-19) for coronary events (p<0.0001) (35). The Helsinki Heart Study (HHS) , a primary prevention trial showed that in an average follow up of 5 years, there was a 34% (95% CI 8.2-52.6, P =0.02) RR reduction in CVD in those treated with gemfibrozil compared to placebo (36). In an 18 year follow up of this study, individuals randomized to gemfibrozil had a 24% adjusted RR reduction (p=0.05) in CVD, and individuals with elevated TG and body mass index (BMI) showed significant benefit from treatment with gemfibrozil. Those with TG level in the highest tertiles had a 71% lower RR of CVD mortality (p <0.001) (37). The Bezafibrate Infarction Prevention (BIP) study assessed the role of fibrates in secondary prevention.  The initial reports showed no significant RR reductions in CVD outcomes in bezafibrate treated vs. placebo-treated subjects. However, a post-hoc analysis of individuals with TG >200 mg/dl demonstrated significant RR reduction by 39.5% (p=0.02); there was no significant RR in those with TG values < 200 mg/dl (3839). In a subgroup analysis of patients in the BIP study by Tenebaum at al. (40), patients with CVD, metabolic syndrome and TG > 150 mg/dl experienced significant benefits from the treatment with bezafibrate. Bezafibrate was associated with a reduced risk of any MI and nonfatal MI with HRs of 0.71 (95% CI, 0.54-0.95) and 0.67 (95% CI, 0.49-0.91), respectively. The cardiac mortality risk tended to be lower in patients taking bezafibrate (HR, 0.74; 95% CI, 0.54-1.03). The Veterans Affairs High-Density Lipoprotein Intervention (VA-HIT) involved 2531 males with CVD with low HDL-c and relatively low LDL-c who were treated with gemfibrozil or placebo and monitored for 5.1 years. Gemfibrozil safely reduced the risk of death from CVD or nonfatal myocardial infarction by 22 percent (41). For every 100 mg/dl increment in baseline TG there was a 14% increase in coronary risk (p=0.045). Further, those with highest tertile of TG values (>180 mg/dl) exhibited a more marked decrease in coronary risk with gemfibrozil compared with those in lower tertiles (42). In the Action to Control Cardiovascular Risk in Diabetes (ACCORD) lipid study using statin + fibrate combination therapy, fenofibrate + simvastatin had no effect on the primary outcome vs. simvastatin alone for all patients. However, in the fenofibrate + simvastatin group, there was a 31% reduction in CV risk in the subgroup with baseline TG levels in the upper tertile vs. simvastatin monotherapy (1).

 

These studies demonstrate that fibrate therapy leading to a reduction in TG levels prevents coronary events. Many of these studies also show improvements in HDL-c levels which may contribute to the improvements in CVD seen; however, recent HDL-c raising studies (using CETP inhibitors) have not found improved cardiovascular benefits suggesting that the decrease in TG levels contributed to the reduction of CVD seen in the fibrate studies.

 

Post-Prandial TG as a CVD Risk Factor

 

There is also growing evidence that postprandial hypertriglyceridemia may be a better indicator of the presence or development of CVD than fasting hypertriglyceridemia.  In the Women’s Health Study, a prospective cohort of 26,330 initially healthy women with over 11 years of follow up, it was observed that higher non-fasting TG levels were strongly associated with an increased risk of future cardiovascular events independent of baseline cardiac risk factors, levels of other lipids, and markers of insulin resistance. The concentrations of lipids and apolipoproteins differed minimally when measurements were performed on non-fasting compared to fasting blood samples, except for TG, which were higher when non-fasting. There was a > 4-fold increased risk of a cardiovascular event among individuals with postprandial TG concentrations peaking at 2-4 hours following a meal. This study showed that HDL-c, TG, total cholesterol/HDL-c ratio, and apolipoprotein B predict CVD when measured in non-fasting samples. By contrast, total cholesterol, LDL, and non-HDL cholesterol, in addition to apolipoprotein B-100 and B-100/A-I ratio, may provide less useful CVD risk information when measured non-fasting (4344). In a Norwegian study which included 42,600 women and 43,641 men ages 20 – 50 years at inclusion, with a mean follow-up of 27 years, non-fasting TG were positively associated with CVD death in both genders, with hazard ratios being higher in women than in men. However, after adjustment for cholesterol, systolic blood pressure and smoking, and in a sub-sample also HDL-c, the associations were distinctly attenuated (45). In another study, the Copenhagen City Heart Study, a prospective cardiovascular study of the Danish general population initiated in 1976, 7581 women and 6391 men who had lipids measured at baseline in 1976-1978, were followed for up to 31 years without losses to follow-up, and most were not taking lipid-lowering therapy. The study found that the cumulative incidence of myocardial infarction, ischemic heart disease, and death increased with increasing levels of non-fasting TG levels. Non-fasting TG level were a better predictor of coronary heart disease in women whereas non-fasting cholesterol level was a better predictor in men. However, non-fasting cholesterol levels were not found to be associated with total mortality (4647). A Japanese study which included 4,988 participants with diabetes already on statin therapy, evaluated the relationship between fasting and non-fasting triglycerides and cardiovascular events.  It showed that cardiac events were associated with elevated fasting and non-fasting TG.   However, the study found that non-fasting TG was more helpful for risk assessment for future CVD instead of fasting TG (103). Data from these studies provide evidence for a link between non-fasting TG and cardiovascular disease and support the concept that non-fasting TG levels may strongly predict the risk of cardiovascular events.

 

Mechanisms by Which TG are a CVD Risk Factor

 

The exact mechanism by which TG may promote vascular disease remains to be elucidated. A possible explanation for TG being associated with increased CVD is that elevated levels of postprandial TG may indicate a high content of TRLs derived from chylomicrons and VLDL. Given their relatively small size, these TRLs can enter the arterial wall, and contribute to the formation of foam cells and thus cause atherosclerosis.  The remnant particles under normal conditions are rapidly taken up by the liver. However, in people with the metabolic syndrome or T2DM, hepatic clearance of remnant particles can be delayed and thus there is a predisposition towards increased production of remnant particles and small dense LDL and HDL particles. Thus, increased production along with prolonged exposure of circulating remnant particles enhances the possibility for the particles to be trapped in the arterial wall. Accordingly, remnant lipoproteins have been shown to increase the risk of atherosclerotic heart disease. This suggests a need to direct attention towards diagnosis and treatment of high TG levels in conjunction with treating high cholesterol levels (48). These studies also draw importance to further investigate independent association of fasting and non-fasting hypertriglyceridemia in CVD.

 

PREVALENCE AND ASSESMENT OF HYPERTRIGLYCERIDEMIA

 

Prevalence of Hypertriglyceridemia

 

There is high prevalence of hypertriglyceridemia in the US which necessitates periodic assessment of TG levels, especially in individuals with increased risk. A study looking at 5680 subjects, greater than or equal to 20 years of age who participated in the National Health and Nutrition Examination Survey from 2001 and 2006 evaluated the epidemiology of adults with hypertriglyceridemia. This study reports about 67.8% of the study participants had a normal TG level (<150 mg/dl), 14.2% had borderline high TG levels (150 – 200 mg/dl) and 16.3% had high TG levels (200 - 500 mg/dl). The prevalence of severe high TG (500 – 2000 mg/dl) was noted to be 1.7% equating to about 2.4 million Americans. Three participants were noted to have TG levels > 2000 mg/dl. The participants with severe high TG tended to be men (75.3%), non-Hispanic whites (70.1%), and aged 40 to 59 years (58.5%), and more than 14% of those reported having diabetes mellitus, and 31.3% reported having hypertension (49). A study published in 2018 surveyed 9593 American adults between 2007-2014 to determine the TG levels in patients taking and not taking a statin.  It showed almost one-third of people taking statins have unsatisfactory TG levels, as well as one-fourth of overall US adults (104).

 

Assessment of Hypertriglyceridemia

 

Plasma lipids and lipoproteins are generally measured in the fasting state and guidelines for therapy for CVD prevention are based on these measurements. The Endocrine Society clinical practice guidelines on evaluation and treatment of hyperlipidemia suggest diagnosis of hypertriglyceridemia based on fasting levels where length of fast is recommended to be 12 hours (8). In insulin resistant states postprandial TG may be more relevant to CVD risk. To assess postprandial TG there is a need to identify an accurate and standardized methodology to measure postprandial triglycerides and TRLs. Currently, the lack of standardization of non-fasting TG measurements, lack of specific reference ranges and the variability of postprandial lipid measurements have hampered their routine clinical use (50). A Fat Tolerance Test (FTT) has been used to assess post prandial lipoproteins.  An expert panel suggests that individuals with fasting TG concentrations between 1-2 mmol/l (89-180 mg/dl) would have better risk assessment by being tested with a FTT than with just fasting TG.  Individuals with fasting TG concentration of less than 1 mmol/l (88.5 mg/dl) commonly do not have exaggerated and delayed response of TGs to a FTT, whereas individuals with elevated fasting TG values above 2 mmol/l (180 mg/dl) are expected to have an exaggerated and delayed response of TG to a FTT. These two patient populations would not benefit from a FTT for better risk assessment (51).

 

Fat Tolerance Testing 

 

Given that humans spend most of their awake time in a post prandial state, various factors including fasting concentrations of serum TGs, time of the day when test is undertaken, the fat content and quality of FTT need to be considered. An expert panel statement recommends measuring total TGs to evaluate the post prandial lipemia response 4 hours after a standardized FTT performed after an 8 hour fast. There has been significant variability in the fat- rich meal used for FTT ranging from dairy products, eggs, oils to liquid formulations. An expert panel suggests a FTT meal consisting of 75 g fat including both saturated and unsaturated fatty acids (51). ApoB-48 is an alternative marker for the assessment of post prandial hypertriglyceridemia as it measures the number of circulating chylomicrons and their remnants after a meal (there is one ApoB-48 per chylomicron particle). The level of ApoB-48 is very low compared to ApoB-100 in the fasting state but it increases after a FTT. However, the lack of internationally recognized standardized assays and reference ranges, limited availability of the ApoB-48 assay, and high costs limit the utilization of ApoB-48 in clinical settings (5052).

 

Secondary Causes of Hypertriglyceridemia

 

Individuals found to have any elevation of fasting TG should be evaluated for secondary causes including endocrine conditions and medications (Table 1) (5354). Patients with untreated diabetes, obesity, and insulin resistant states commonly have elevated TG levels (5556). Other endocrine disorders such as hypothyroidism, Cushing’s disease, and growth hormone deficiency can also be associated with elevated TG levels (8). TG levels can also significantly increase during pregnancy owing to estrogen-induced stimulation of the secretion of hepatic TRLs (57). In women with underlying disorders of TG metabolism, this increase in TG levels during pregnancy can be associated with pancreatitis and fetal loss. Alcohol intake increases hepatic fatty acid synthesis and decreases breakdown resulting in increased hepatic VLDL secretion and hypertriglyceridemia. Lipodystrophies, either primary or as seen in HIV treated patients or with other diseases is also associated with hypertriglyceridemia (8). There are several monogenic autosomal recessive disorders that lead to hypertriglyceridemia (table 1). LPL deficiency, apo CII deficiency and GPIHBP1 loss of function mutations are associated with impaired LPL activity and present in young patients with increased risk of chylomicronemia and pancreatitis. Additional genetic syndromes in the differential diagnosis of hypertriglyceridemia include mixed or familial combined hyperlipidemia (FCHL), type III dysbetalipoproteinemia and familial hypertriglyceridemia (FHTG) (5859). Hypertriglyceridemia can also be polygenic. Many patients have multiple genetic variants and combined with environmental factors, can cause hypertriglyceridemia (107). However a study with 563 patients with severe hypertriglyceridemia showed that 14.4% had heterozygous rare variants known to cause hypertriglyceridemia while 3.8% of the control group had these variants as well indicating that these variants are incompletely or partly penetrant. Polygenic risk can now be assessed by a polygenic score.  An elevated score increases the probability of developing hypertriglyceridemia, but it is not an absolute predictor of developing hypertriglyceridemia (107). Many drugs also raise triglyceride levels (table 1). Oral estrogens increase the hepatic secretion of VLDL causing an increase in serum TG levels (60). Other medications include Tamoxifen/Raloxifene, retinoids, beta blockers, thiazide inhibitors, corticosteroids, immunosuppressants, antipsychotics and antiretroviral protease inhibitors (8). If possible, individuals with secondary hypertriglyceridemia should have the secondary cause addressed, and such individuals may then not need primary, TG-lowering therapy. However, secondary causes of hypertriglyceridemia cannot always be addressed, in which case providers should consider TG-lowering therapy.

 

Table 1. Causes of Hypertriglyceridemia

Disorders

Drugs

Monogenic*

Hypothyroidism

Uncontrolled Diabetes

Obesity

Chronic renal failure

Nephrotic syndrome

Pregnancy

HIV

Cushing’s syndrome

Lipodystrophy

Inflammatory disease – rheumatoid arthritis, lupus, psoriasis, etc.

Alcohol

Estrogens

Beta blockers

Tamoxifen/Raloxifene

Glucocorticoids

Atypical anti-psychotics

Cyclosporine

Protease inhibitors

Lipoprotein lipase deficiency

Apolipoprotein CII deficiency

Apolipoprotein AV deficiency

GPIHBP1 deficiency

Lipase Maturation factor 1 (LMF1)

*autosomal recessive disorders

 

GUIDELINES FOR TRIGLYCERIDE EVALUTION AND MANAGEMENT

 

The Endocrine Society Clinical Guidelines

 

The Endocrine Society Guidelines recommends that the diagnosis of hypertriglyceridemia be based on fasting serum triglyceride levels and defines TG levels of 150 to 199 mg per dL as mild hypertriglyceridemia; 200 to 999 mg per dL as moderate; 1,000 to 1,999 mg per dL as severe; and 2,000 mg per dL or greater as very severe hypertriglyceridemia. The screening for elevated triglyceride levels for all adults is recommended as part of a lipid panel at least every five years. These guidelines recommend against the routine measurement of lipoprotein particle heterogeneity. The guidelines also recommend screening patients with hypertriglyceridemia for secondary causes (medications, alcohol use, endocrine diseases, renal disease, liver disease) and that patients with primary hyperlipidemia be evaluated for family history of dyslipidemia and CVD. The guidelines recommend the use of non-HDL-c (goal 30 mg/dl higher than the LDL-c goal) for both risk stratification and as a target for therapy in patients with moderate hypertriglyceridemia.  Initial treatment of patients with mild to moderate hypertriglyceridemia should include lifestyle therapy. For patients with severe to very severe hypertriglyceridemia, dietary modifications in combination with drug treatment should be considered. A fibrate is recommended as a first-line agent in patients with severe or very severe hyperlipidemia (8).

 

American Association Of Clinical Endocrinologists (AACE) Guidelines

 

Similar to other guidelines, the AACE clinical practice guidelines recommend evaluating all adults >20 years of age for dyslipidemia every 5 years for risk assessment with a fasting (9 to 12-hour fast) lipid profile. In addition, it recommends more frequent assessments for patients with a family history of premature CVD. However, unlike other guidelines, AACE also recommends Apo B measurements to assess for residual risk in patients with increased TG levels (>150 mg/dl) or low HDL-c levels (< 40 mg/dl). TG levels less than 150 mg/dl are defined as normal, 150 – 199 mg/dl as borderline high, 200 – 499 mg/dl as high, and levels >500 mg/dl or greater as very high, and AACE recommends maintaining TG levels less than 150 mg/dl.  Fibrates are recommended for the treatment of severe hypertriglyceridemia (>500 mg/dl) and lifestyle changes including physical activity, weight loss, and smoking cessation are recommended as first line therapy in moderate hypertriglyceridemia (61).

 

American Heart Association (AHA) Statement on Triglycerides and Cardiovascular Disease

 

In 2011, AHA published a scientific statement addressing TG and CVD; however, this statement is not intended to serve as a specific guideline. In this statement optimal fasting TG level is defined as < 100 mg/dl as a parameter of metabolic health and they suggest screening for non-fasting TG for those with high fasting TG levels. A non-fasting level of <200 mg/dl corresponds with a normal (<150 mg/dl) or optimal (<100 mg/dl) fasting TG level and requires no further testing. The AHA statement uses fasting TG levels to define levels between 150 – 199 mg/dl as borderline high, 200 – 499 mg/dl as high and levels >500 mg/dl as very high. Intensive lifestyle changes are recommended to be most crucial in the management of hypertriglyceridemia and reductions of 50% or more in TG levels may be attained through intensive therapeutic lifestyle change (58).

 

National Lipid Association (NLA)

 

The National Lipid Association guidelines recommend obtaining a fasting or a non-fasting lipoprotein profile in all adults (>20 years) every 5 years. It defines TG level of <150 mg/dl as normal, 150 – 199 mg/dl borderline high, 200 – 499 mg/dl as high and levels of >500 mg/dl as very high. The NLA Expert Panel views non-HDL-c as a better primary target for medication than LDL-c and recommends levels of non-HDL-c < 130 mg/dl as the desirable level of atherogenic cholesterol for primary prevention of CVD and non-HDL-c <100 mg/dL for high risk patients or patients with ASCVD.  An elevated TG level is not a target of therapy, except when very high (>500 mg/dl). NLA recommends that when TG levels are between 200 – 499 mg/dl, the targets of therapy are non-HDL-c and LDL-c to reduce risk of CVD events and when TG levels are very high (>500 mg/dl, and especially if >1000 mg/dl), reducing the concentration to <500 mg/dl to prevent pancreatitis becomes the primary goal. The NLA recommends lifestyle interventions as first step in efforts to reduce triglycerides. If drug therapy is indicated NLA guidelines recommend using fibric acids, omega-3 fatty acids, or nicotinic acid as first line agents if fasting TG level is >1000 mg/dl. For patients with TG levels of 500 – 999 mg/dl a triglyceride-lowering agent or a statin is considered reasonable and for TG level between 200 – 499 mg/dl a statin generally is considered fist-line drug therapy with addition of a  triglyceride-lowering agent if non-HDL-c is not at goal post initiation of statin (62).

 

European Society of Cardiology (ESC)/European Atherosclerosis Society (EAS) Guidelines

 

Similar to the above-mentioned guidelines, ESC/EAS guidelines also recommend checking lipid levels in the fasting state. The specific target for non-HDL-c is recommended to be 0.8 mmol/L (~30 mg/dl) higher than the corresponding LDL-c target and TG level < 1.7 mmol/L (~ 150 mg/dl). TG levels above 10 mmol/L (~ 880 mg/dl) are considered to be clinically significant for the risk of pancreatitis and treatment is recommended. In patients with hypertriglyceridemia the ESC/EAS guidelines recommend that the first step is to consider possible causes of hypertriglyceridemia and to evaluate for total CVD risk; the primary goal is to achieve LDL-c target based on the total CVD risk. The use of pharmacologic agents is recommended in patients with TG levels > 2.3 mmol/L (~ 200 mg/dl) who cannot lower them by lifestyle changes and/or if the subject is at high risk of CVD. Statins are recommended as the first choice to reduce both total CVD risk and moderately elevated TG levels. In high-risk patients with TG levels between 1.5-5.6 mmol/L (135-499 mg/dL) while already on statin therapy, use of icosapent ethyl 2x2 g/day can be considered (63). Patients who present with ACS and those whose LDL-C are not at goal on maximally tolerated statin and ezetimibe, addition of PCSK9 inhibitor should be considered.

 

TG Assessment Strategies

 

The different guidelines in general recommend screening for lipids using a fasting lipid profile, screening for secondary causes of dyslipidemia, and focusing on lifestyle interventions as the first approach to lower elevated TG. A practical approach is to request fasting lipid panels on patients, but to obtain non-fasting (random) panels if fasting samples cannot be provided. For any patient with a TG level that is elevated (for example, > 200 mg/dl), screen for secondary causes and address these if possible. For significantly elevated TG (e.g., > 500 mg/dl) consider the addition of TG-lowering therapy, with the goal of preventing any further elevations increasing the risk for pancreatitis. For individuals with moderately elevated TG (e.g., 150-500mg/dl) then we recommend consideration of TG-lowering therapy on an individual basis. For example, individuals with existing CVD or at high CVD risk, or those with very low HDL levels might be candidates for therapy; whereas in those with low CVD risk, or desirable HDL levels, additional focus on lifestyle interventions to lower TG may be the appropriate first line of therapy. The use of non-HDL cholesterol levels can help guide decisions.

 

MANANGEMENT OF HYPERTRIGLYCERIDEMIA

 

Lifestyle Intervention

 

Studies have shown that the consumption of a Western diet which includes highly processed, calorie-dense and nutrient poor foods leads to an exaggerated lipemia. In addition, factors such as physical inactivity, cigarette smoking, excessive alcohol intake, and obesity worsen lipemia (51). Hence, the control of secondary factors and lifestyle changes are considered to be the first line approach of the clinical management of both fasting hypertriglyceridemia and post prandial hyperlipidemia. Appropriate dietary changes include limiting fat content, caloric restriction resulting in weight loss, restriction of alcohol intake, and increased exercise are fundamental for management of hypertriglyceridemia (5051). The type of carbohydrate consumed may affect serum triglycerides and a diet rich in simple carbohydrates and sugar-sweetened beverages is associated with hypertriglyceridemia. As compared with starches, sugars, particularly sucrose and fructose, tend to increase serum triacylglycerol concentrations by about 60%. Because fructose bypasses a major rate-determining step in glycolysis, a high influx of fructose to the liver promotes triacylglycerol synthesis and VLDL production (64). The effects of sucrose or fructose on fasting TG may be more pronounced in men, sedentary overweight individuals, or those with the metabolic syndrome. Sucrose and fructose also increase postprandial TG levels and may augment the lipemia associated with fat-containing meals (65).  There is mounting evidence that physical activity lowers risk for CVD (66). Mestek et al. (67)reported that aerobic exercise lowered the postprandial triglyceride response to a high fat meal in subjects with metabolic syndrome. The effects of exercise in reducing postprandial lipemia are seen both in an acute setting right after exercise as well as delayed effects through the next day. Additionally, exercise does not need to be a single continuous bout but instead could be spread out throughout the day. Accumulated physical activity appears to be as effective in lowering postprandial TGs concentrations (68) as a single bout. The mechanisms leading to decreased triglyceride levels post meals are not completely understood and need further investigation.

 

Statins

 

Statins are the most widely used lipid lowering agents and have beneficial effects on cardiovascular morbidity and mortality. Statins are effective in lowering non-HDL-c, mainly because of their LDL lowering action and to a certain extent lowering TG levels. The higher the baseline TG levels, the greater the TG lowering effect. Available data also indicate that statins can reduce postprandial TG values (51). Statins inhibit HMG-CoA reductase, hence up-regulate the LDL receptor due to the intracellular depletion of cholesterol in the liver. Increased numbers of LDL receptors may improve removal of TRL remnants in postprandial state. It is also postulated that statins inhibit VLDL synthesis (70).  Parhofer et al (71)showed that 10 mg of atorvastatin per day for 4 weeks improves, but does not normalize, post prandial lipoprotein metabolism in hypertriglyceridemic patients. Other studies have also shown that atorvastatin improved fasting as well as postprandial lipemia (7273).

 

Fibrates

 

Fibrates have the most pronounced effect on lowering plasma TG levels of currently available lipid lowering therapies. Through activation of peroxisomal proliferator activated receptor (PPAR) alpha, fibrates decrease triglycerides by increasing LPL activity and decreasing apolipoprotein CIII production leading to increased lipolysis. Fibrates also increase fatty acid oxidation in the liver leading to a decrease in VLDL secretion (51). The Endocrine Society Clinical Practice Guidelines on Evaluation and Treatment of Hypertriglyceridemia recommend that a fibrate be used as a first line agent for reduction of TGs in patients at risk for triglyceride- induced pancreatitis (8). The ACCORD trial evaluated the benefit of adding fenofibrate to simvastatin therapy concluded that the addition of fenofibrate in patients with diabetes did not reduce the rate of CVD events. However, in the fenofibrate + simvastatin group there was a significant reduction in cardiovascular risk in the subgroup with clinically significant dyslipidemia marked by elevated TG levels and low HDL levels (74). Rosenson et al. reported that fenofibrate treatment for 6 weeks significantly decreased both postprandial hypertriglyceridemia and the inflammatory response after the ingestion of a test meal consisting of a milkshake including standardized fat content (68% of energy) that was adjusted to body surface area (50 g/m2) in patients with hypertriglyceridemia and the metabolic syndrome (75). In a small study (n = 10), bezafibrate was shown to significantly decrease postprandial endothelial dysfunction and elevations of both exogenous and endogenous triglycerides in patients with metabolic syndrome (76). The effects of fibrates in decreasing postprandial TRLs may play a role in their vascular protective effects.

 

Niacin

 

Niacin decreases TG levels and has pronounced effects on increasing HDL concentration. The mechanism of action of niacin remains unclear, but it is proposed that niacin decreases triglyceride synthesis and hepatic secretion of VLDL. The Coronary Drug Project was a randomized controlled trial that looked at the role of immediate-release niacin as a solo agent for coronary prevention. The Coronary Drug Project showed that niacin was associated with a significant reduction in cardiovascular events (7778).  Studies have shown that both immediate-release and extended-release niacin suppress postprandial hypertriglyceridemia (7980). The Atherothrombosis Intervention in Metabolic Syndrome with Low HDL/High Triglycerides: Impact on Global Health Outcomes (AIM-HIGH) trial showed that the addition of niacin to statin therapy in patients with CVD and LDL cholesterol levels of less than 70 mg per deciliter had no incremental clinical benefit during a 36-month follow-up period, despite significant improvements in HDL cholesterol and triglyceride levels (2). However, a trend towards benefit (hazard ratio 0.74; p=0.073) was found for the subset of patients with both the highest TG levels and lowest HDL levels (>198 and <33mg/dl respectively) (81). Lipids in this study were measured in fasting state. Similarly, the Heart Protection Study 2-Treatment of HDL to Reduce the Incidence of Vascular Events (HPS2 –THRIVE) study, which compared niacin + laropiprant (a prostaglandin D2 receptor antagonist used as an anti-flushing agent) + statin vs statin alone did not find added benefit of niacin. However, this lack of additional benefit may be related to the patient population studied which did not have elevated TG levels (82)and a possible benefit may be seen for subjects with both elevated TG and low HDL. Further studies are needed to access the effects of niacin on hypertriglyceridemia in metabolic syndrome and patients with T2DM.

 

Ezetimibe

 

Ezetimibe is a cholesterol lowering agent that inhibits the intestinal absorption of cholesterol (83). Recent studies show that ezetimibe alone or in conjunction with statins also reduces postprandial hypertriglyceridemia. Masuda, et al. showed that ezetimibe significantly decreased triglycerides in the fasting state along with a decrease in postprandial elevations of cholesterol and TG levels in the chylomicrons (CM) size range, suggesting that the postprandial production of CM particles was suppressed by ezetimibe (84). In a study by Olijhoek et al, combination therapy with low dose simvastatin and ezetimibe was shown to preserve post-fat load endothelial function when compared to treatment with high-dose simvastatin monotherapy in male metabolic syndrome patients (85). The Improved Reduction of Outcomes: Vytorin Efficacy International trial (IMPROVE-IT), a multicenter, randomized, double blind trial of 18,144 moderate-high risk patients stabilized following ACS, was conducted to investigate if the addition of ezetimibe to a statin improves cardiovascular outcomes relative to statin monotherapy in these patients. The results from this study suggest that the addition of ezetimibe to statin therapy improves cardiovascular outcomes, but likely via further LDL-c lowering (86).

 

Fish Oil

 

Omega 3 polyunsaturated fatty acids (PUFAs) have dose dependent TG lowering effects resulting from variety of mechanisms including decreased VLDL secretion and improved VLDL TG clearance (87). In the Japan EPA Lipid Intervention Study (JELIS) trial, 18,645 patients in Japan were recruited between 1996 and 1999 and assigned to receive either 1800 mg of eicosapentaenoic acid (EPA) daily with statin or statin only.  A 19% relative reduction in major coronary events (p = 0.011) was seen in patients in the EPA group. Unstable angina and non-fatal coronary events were significantly reduced; however, sudden cardiac death and coronary death did not differ between the groups (88). Two recent trials, REDUCE-IT and STRENGTH studied the effects of high dose omega-3 fatty acids and had differing results. The REDUCE-IT trail with 8,179 patients showed a decrease in major cardiac events with high dose icosapent ethyl (4g/d EPA), while the STRENGTH trial with 13,086 patients showed no effects on cardiac events with high dose EPA/DHA carboxylic acid (4g/d). (98,99) A difference in the two studies could be due to the EPA levels achieved in each study. REDUCE-IT achieved an EPA level of 144 (mg/mL) while the STRENGTH trial achieved an EPA level of 89.6 (mg/mL) (105). Another issue is that in the REDUCE-IT trial, mineral oil was used as the placebo in the control group which resulted in higher atherogenic lipoproteins in that arm. This raised concerns that both the negative effects from the mineral oil in the control group and the positive effects of EPA in the treatment group underlies the observed reduction in major cardiac events seen in the trial.  However, Olshansky et al reviewed eight studies that used mineral oil as a placebo which showed no evidence that mineral oil in the dosage used in the REDUCE-IT trial had any effect on clinical outcomes. (108) Given the opposite results of the two trials, further investigation is needed in regards to EPA levels and drug formulations. A secondary analysis of the STREGNTH trial showed no cardiovascular benefits at the highest levels of DHA or EPA (99). A meta-analysis which includes the REDUCE-IT and STRENGTH trails show that there was a higher risk of atrial fibrillation in groups treated with omega-3 fatty acids than placebo. (100). This necessitates further evaluation in the potential adverse effects of omega 3 fatty acids.

 

 

A few studies have examined the effects of fish oil supplementation on postprandial lipemia and found that fish oil use decreases fasting and postprandial triglyceride levels (8990). A study looking at the effect of fish oil, exercise and the combined treatments on fasting and postprandial chylomicron metabolism showed that combining fish oil with chronic exercise, reduced the plasma concentration of pro-atherogenic chylomicron remnants; in addition it reduced the fasting and postprandial TG response in viscerally obese insulin resistant subjects (91).

 

For additional information on drugs to treat hyperlipidemia see the chapters on triglyceride lowering drugs and cholesterol lowering drugs in Endotext (9293).

 

New Therapies on the Horizon

 

APOC3 is a CVD risk factor due to its association with increased triglyceride levels. Antisense oligonucleotides (ASOs) are novel therapeutic agents that bind mRNA leading to its degradation. An ASO to APOC3 was found to lower APOC3 and triglyceride levels. A RCT evaluating this ASO in patients with hypertriglyceridemia (fasting triglyceride levels between 350-2000 mg/dl if not on triglyceride-lowering therapy, or 225-2000 mg/dl if on a fibrate) found that it led to reductions in triglyceride levels of 30-71% over the 13-week trial period. After the ASO was discontinued triglyceride levels returned towards baseline levels over the next 13 weeks. There were no safety concerns in this trial (94). In the APPROACH trial 66 patients with monogenic chlyomicronemia were randomly given a placebo or volanesorsen (300mg once weekly). The group treated with volanesorsen had TG levels lowered to less than 750 mg/dl in 77% of the patients at 3 months compared to 10% in patients treated with the placebo (101). A common side effect with volanesorsen is thrombocytopenia and it is currently only approved for use in Europe. (101) Studies using this approach to target triglycerides as a CVD risk factor are ongoing. Furthermore, inhibition of APOC3 may be a therapeutic option in individuals with LPL deficiency (95); at present there are no effective therapies except for extreme dietary restrictions for these individuals.

 

ANGPTL3 is another potential target for triglyceride lowering. Genetic causes of decreased activity are associated with lower triglyceride, HDL, and LDL levels as well as a decreased risk for CVD. A small trial using a human monoclonal antibody to target ANGPTL3 reported decreases in triglycerides up to 76% (20).  A recent study by Harada-Shiba et al, a phase 1 study took a total of 96 Caucasian and Japanese patients and randomized them to receive varying doses and routes of evinacumab or placebo. In the evinacumab cohorts, reduced triglycerides were rapidly seen in a dose-dependent manner. The study showed no serious or severe treatment emergent adverse events (102). Further clinical trials using either monoclonal antibody or antisense technology are ongoing.

 

Other Treatments

 

Emerging evidence suggests that incretin-based therapies not only improve post prandial glucose levels in diabetic patients, but may also pay a role in postprandial lipid metabolism (96). Recent trials have found cardiovascular protection for some of these agents (see (97). Improved understanding of the physiology and mechanism thorough which postprandial hypertriglyceridemia leads to increased cardiovascular events will help identify new targets in future.

 

CONCLUSION

 

Recent data strongly indicate that fasting as well as non-fasting hypertriglyceridemia is a risk factor for atherosclerosis and CVD. Current treatment goals aimed at lowering LDL-c still do not eliminate residual risk of CVD. Current guidelines focus mainly on LDL-c levels and correction of hypertriglyceridemia is not the aim of current treatment. However, focus on elevated hypertriglyceridemia deserves renewed attention, particularly as one-third of all adults in the United States suffer from elevated TG and growing number of people are diagnosed with metabolic syndrome or T2DM. There is need for more studies specifically testing the benefits of lowering hypertriglyceridemia. Additionally, the usefulness of “fat tolerance test” using a standardized meal, analogous to a glucose tolerance test, warrants further evaluation as potential indicator of a metabolic state identifying individuals at higher risk for cardiovascular events. Given the association with CVD, elevated postprandial TGs levels may represent a particularly attractive therapeutic target and further studies particularly looking at effect of various lipid lowering agents on postprandial along with fasting TGs are necessary.

 

REFERENCES

 

  1. Chapman, M.J., et al., Triglyceride-rich lipoproteins and high-density lipoprotein cholesterol in patients at high risk of cardiovascular disease: evidence and guidance for management. Eur Heart J, 2011. 32(11): p. 1345-61.
  2. Boden, W.E., et al., Niacin in Patients with Low HDL Cholesterol Levels Receiving Intensive Statin Therapy. New England Journal of Medicine, 2011. 365(24): p. 2255- 2267.
  3. Talayero, B.G. and F.M. Sacks, The role of triglycerides in atherosclerosis. Curr Cardiol Rep, 2011. 13(6): p. 544-52.
  4. Hokanson, J.E. and M.A. Austin, Plasma triglyceride level is a risk factor for cardiovascular disease independent of high-density lipoprotein cholesterol level: a meta- analysis of population-based prospective studies. J Cardiovasc Risk, 1996. 3(2): p. 213- 9.
  5. Expert Panel on Detection, E. and A. Treatment of High Blood Cholesterol in, Executive Summary of The Third Report of The National Cholesterol Education Program (NCEP) Expert Panel on Detection, Evaluation, And Treatment of High Blood Cholesterol In Adults (Adult Treatment Panel III). JAMA, 2001. 285(19): p. 2486-97.
  6. Abdel-Maksoud, M., et al., Effects of modifying triglycerides and triglyceride-rich lipoproteins on cardiovascular outcomes, in J Cardiovasc Pharmacol. 2008. p. 331-51.
  7. Zilversmit, D.B., Atherogenesis: a postprandial phenomenon. Circulation, 1979. 60(3): p. 473-85.
  8. Berglund, L., et al., Evaluation and treatment of hypertriglyceridemia: an Endocrine Society clinical practice guideline. J Clin Endocrinol Metab, 2012. 97(9): p. 2969-89.
  9. Murad, M.H., et al., The association of hypertriglyceridemia with cardiovascular events and pancreatitis: a systematic review and meta-analysis. BMC Endocr Disord, 2012. 12: p. 2.
  10. Champe, P., et al., Cholesterol and Steroid Metabolism, in Lippincott's Illustrated Reviews: Biochemistry 3rd Ed. 2005, Lippincott Williams & Wilkins: Philadelphia. p. 217- 24.
  11. Melmed, S., et al., Disorder of Lipid Metabolism, in Williams Textbook of Endocrinology. 2011, Elsevier/Saunders: Philidelphia. p. 1633-74.
  12. Feingold, K.R. Introduction to Lipids and Lipoproteins. 2021 Jan 19. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dhatariya K, Dungan K, Hershman JM, Hofland J, Kalra S, Kaltsas G, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrère B, Levy M, McGee EA, McLachlan R, Morley JE, New M, Purnell J, Sahay R, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000–.
  13. Adiels, M., et al., Overproduction of very low-density lipoproteins is the hallmark of the dyslipidemia in the metabolic syndrome. Arteriosclerosis Thrombosis and Vascular Biology, 2008. 28(7): p. 1225-1236.
  14. Mendivil, C.O., et al., Low-density lipoproteins containing apolipoprotein C-III and the risk of coronary heart disease. Circulation, 2011. 124(19): p. 2065-72.
  15. Ooi, E.M., et al., Apolipoprotein C-III: understanding an emerging cardiovascular risk factor. Clin Sci (Lond), 2008. 114(10): p. 611-24.
  16. Sacks, F.M., et al., VLDL, apolipoproteins B, CIII, and E, and risk of recurrent coronary events in the Cholesterol and Recurrent Events (CARE) trial. Circulation, 2000. 102(16): p. 1886-92.
  17. Jorgensen, A.B., et al., Loss-of-function mutations in APOC3 and risk of ischemic vascular disease. N Engl J Med, 2014. 371(1): p. 32-41.
  18. Pollin, T.I., et al., A null mutation in human APOC3 confers a favorable plasma lipid profile and apparent cardioprotection. Science, 2008. 322(5908): p. 1702-5.
  19. Tg, et al., Loss-of-function mutations in APOC3, triglycerides, and coronary disease. N Engl J Med, 2014. 371(1): p. 22-31.
  20. Dewey, F.E., et al., Genetic and Pharmacologic Inactivation of ANGPTL3 and Cardiovascular Disease. N Engl J Med, 2017. 377(3): p. 211-221.
  21. Streja, D., Hypertriglyceridemic Pancreatitis, in Endotext, L.J. De Groot, et al., Editors. 2017: South Dartmouth (MA).
  22. Leaf, D.A., Chylomicronemia and the chylomicronemia syndrome: a practical approach to management. Am J Med, 2008. 121(1): p. 10-2.
  23. Lindkvist, B., et al., A prospective cohort study on risk of acute pancreatitis related to serum triglycerides, cholesterol and fasting glucose. Pancreatology, 2012. 12(4): p. 317- 24.
  24. Sandhu, S., et al., Incidence of pancreatitis, secondary causes, and treatment of patients referred to a specialty lipid clinic with severe hypertriglyceridemia: a retrospective cohort study. Lipids Health Dis, 2011. 10: p. 157.
  25. Murphy, M.J., et al., Hypertriglyceridemia and acute pancreatitis. JAMA Intern Med, 2013. 173(2): p. 162-4.
  26. Christian, J.B., et al., Determining triglyceride reductions needed for clinical impact in severe hypertriglyceridemia. Am J Med, 2014. 127(1): p. 36-44 e1.
  27. Patel, A., et al., Serum triglycerides as a risk factor for cardiovascular diseases in the Asia-Pacific region. Circulation, 2004. 110(17): p. 2678-2686.
  28. Sarwar, N., et al., Triglycerides and the risk of coronary heart disease - 10 158 incident cases among 262 525 participants in 29 Western prospective studies. Circulation, 2007. 115(4): p. 450-458.
  29. Varbo, A., et al., Remnant Cholesterol as a Causal Risk Factor for Ischemic Heart Disease. Journal of the American College of Cardiology, 2013. 61(4): p. 427-436.
  30. Teslovich, T.M., et al., Biological, clinical and population relevance of 95 loci for blood lipids. Nature, 2010. 466(7307): p. 707-713.
  31. Sarwar, N., et al., Triglyceride-mediated pathways and coronary disease: collaborative analysis of 101 studies. Lancet, 2010. 375(9726): p. 1634-1639.
  32. Jorgensen, A.B., et al., Genetically elevated non-fasting triglycerides and calculated remnant cholesterol as causal risk factors for myocardial infarction. European Heart Journal, 2013. 34(24): p. 1826-+.
  33. Thomsen, M., et al., Low Nonfasting Triglycerides and Reduced All-Cause Mortality: A Mendelian Randomization Study. Clinical Chemistry, 2014. 60(5): p. 737-746.
  34. Myocardial Infarction, G., et al., Coding Variation in ANGPTL4, LPL, and SVEP1 and the Risk of Coronary Disease. N Engl J Med, 2016. 374(12): p. 1134-44.
  35. Jun, M., et al., Effects of fibrates on cardiovascular outcomes: a systematic review and meta-analysis. Lancet, 2010. 375(9729): p. 1875-84.
  36. Frick, M.H., et al., Helsinki Heart Study: primary-prevention trial with gemfibrozil in middle-aged men with dyslipidemia. Safety of treatment, changes in risk factors, and incidence of coronary heart disease. N Engl J Med, 1987. 317(20): p. 1237-45.
  37. Tenkanen, L., et al., Gemfibrozil in the treatment of dyslipidemia: an 18-year mortality follow-up of the Helsinki Heart Study. Arch Intern Med, 2006. 166(7): p. 743-8.
  38. Haim, M., et al., Elevated serum triglyceride levels and long-term mortality in patients

          with coronary heart disease: the Bezafibrate Infarction Prevention (BIP) Registry.

         Circulation, 1999. 100(5): p. 475-82.

  1. Bezafibrate Infarction Prevention, s., Secondary prevention by raising HDL cholesterol

         and reducing triglycerides in patients with coronary artery disease. Circulation, 2000.

         102(1): p. 21-7.

  1. Tenenbaum, A., et al., Bezafibrate for the secondary prevention of myocardial infarction

          in patients with metabolic syndrome. Arch Intern Med, 2005. 165(10): p. 1154-60.

  1. Rubins, H.B., et al., Gemfibrozil for the secondary prevention of coronary heart disease

          in men with low levels of high-density lipoprotein cholesterol. Veterans Affairs High- Density Lipoprotein Cholesterol Intervention Trial Study Group. N Engl J Med, 1999. 341(6): p. 410-8.

  1. Miller, M., B. Cosgrove, and S. Havas, Update on the role of triglycerides as a risk factor for coronary heart disease. Curr Atheroscler Rep, 2002. 4(6): p. 414-8.
  2. Mora, S., et al., Fasting compared with nonfasting lipids and apolipoproteins for predicting incident cardiovascular events. Circulation, 2008. 118(10): p. 993-1001.
  3. Bansal, S., et al., Fasting compared with nonfasting triglycerides and risk of cardiovascular events in women. JAMA, 2007. 298(3): p. 309-16.
  4. Lindman, A.S., et al., Nonfasting triglycerides and risk of cardiovascular death in men and women from the Norwegian Counties Study. Eur J Epidemiol, 2010. 25(11): p. 789- 98.
  5. Langsted, A., et al., Nonfasting cholesterol and triglycerides and association with risk of myocardial infarction and total mortality: the Copenhagen City Heart Study with 31 years of follow-up. J Intern Med, 2011. 270(1): p. 65-75.
  6. Nordestgaard, B.G., et al., Nonfasting triglycerides and risk of myocardial infarction, ischemic heart disease, and death in men and women. Jama, 2007. 298(3): p. 299-308.
  7. Nordestgaard, B.G., A. Langsted, and J.J. Freiberg, Nonfasting hyperlipidemia and cardiovascular disease. Curr Drug Targets, 2009. 10(4): p. 328-35.
  8. Christian, J.B., et al., Prevalence of severe (500 to 2,000 mg/dl) hypertriglyceridemia in United States adults. Am J Cardiol, 2011. 107(6): p. 891-7.
  9. Boren, J., et al., Postprandial hypertriglyceridemia as a coronary risk factor. Clinica Chimica Acta, 2014. 431: p. 131-142.
  10. Kolovou, G.D., et al., Assessment and clinical relevance of non-fasting and postprandial triglycerides: an expert panel statement. Curr Vasc Pharmacol, 2011. 9(3): p. 258-70.
  11. Smith, D., et al., Post-prandial chylomicron response may be predicted by a single measurement of plasma apolipoprotein B48 in the fasting state. European Journal of Clinical Investigation, 1999. 29(3): p. 204-209.
  12. Feingold KR, Brinton EA, Grunfeld C. The Effect of Endocrine Disorders on Lipids and Lipoproteins. 2020 Mar 9. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dhatariya K, Dungan K, Hershman JM, Hofland J, Kalra S, Kaltsas G, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrère B, Levy M, McGee EA, McLachlan R, Morley JE, New M, Purnell J, Sahay R, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000–.
  13. Herink, M. and Ito MK. Medication Induced Changes in Lipid and Lipoproteins. 2018 May 10. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dhatariya K, Dungan K, Hershman JM, Hofland J, Kalra S, Kaltsas G, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrère B, Levy M, McGee EA, McLachlan R, Morley JE, New M, Purnell J, Sahay R, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000–.
  14. Feingold, K.R. Dyslipidemia in Diabetes. 2020 Aug 10. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dhatariya K, Dungan K, Hershman JM, Hofland J, Kalra S, Kaltsas G, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrère B, Levy M, McGee EA, McLachlan R, Morley JE, New M, Purnell J, Sahay R, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000–.
  15. Feingold, K.R. Obesity and Dyslipidemia. 2020 Nov 2. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dhatariya K, Dungan K, Hershman JM, Hofland J, Kalra S, Kaltsas G, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrère B, Levy M, McGee EA, McLachlan R, Morley JE, New M, Purnell J, Sahay R, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000–.
  16. Grimes, S.B. and Wild R. Effect of Pregnancy on Lipid Metabolism and Lipoprotein Levels. 2018 Feb 20. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dhatariya K, Dungan K, Hershman JM, Hofland J, Kalra S, Kaltsas G, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrère B, Levy M, McGee EA, McLachlan R, Morley JE, New M, Purnell J, Sahay R, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000–.
  17. Miller, M., et al., Triglycerides and cardiovascular disease: a scientific statement from the American Heart Association. Circulation, 2011. 123(20): p. 2292-333.
  18. Chait, A., S. Subramanian, and J.D. Brunzell, Genetic Disorders of Triglyceride Metabolism, in Endotext, L.J. De Groot, et al., Editors. 2015: South Dartmouth (MA).
  19. Kissebah, A.H., P. Harrigan, and V. Wynn, Mechanism of hypertriglyceridaemia associated with contraceptive steroids. Horm Metab Res, 1973. 5(3): p. 184-90.
  20. Jellinger, P.S., et al., American Association of Clinical Endocrinologists' Guidelines for Management of Dyslipidemia and Prevention of Atherosclerosis. Endocr Pract, 2012. 18 Suppl 1: p. 1-78.
  21. Jacobson, T.A., et al., National Lipid Association recommendations for patient-centered management of dyslipidemia: part 1 - executive summary. J Clin Lipidol, 2014. 8(5): p. 473-88.
  22. Catapano, A.L., et al., ESC/EAS Guidelines for the management of dyslipidaemias The Task Force for the management of dyslipidaemias of the European Society of Cardiology (ESC) and the European Atherosclerosis Society (EAS). Atherosclerosis, 2011. 217(1): p. 3-46.
  23. Fried, S.K. and S.P. Rao, Sugars, hypertriglyceridemia, and cardiovascular disease. Am J Clin Nutr, 2003. 78(4): p. 873S-880S.
  24. Johnson, R.K., et al., Dietary sugars intake and cardiovascular health: a scientific statement from the American Heart Association. Circulation, 2009. 120(11): p. 1011-20.
  25. Li, J. and J. Siegrist, Physical activity and risk of cardiovascular disease--a meta- analysis of prospective cohort studies. Int J Environ Res Public Health, 2012. 9(2): p. 391-407.
  26. Mestek, M.L., et al., Aerobic exercise and postprandial lipemia in men with the metabolic syndrome. Med Sci Sports Exerc, 2008. 40(12): p. 2105-11.
  27. Maraki, M.I. and L.S. Sidossis, The latest on the effect of prior exercise on postprandial lipaemia. Sports Med, 2013. 43(6): p. 463-81.
  28. Enkhmaa, B., et al., Lifestyle Changes: Effect of Diet, Exercise, Functional Food, and Obesity Treatment, on Lipids and Lipoproteins, in Endotext, L.J. De Groot, et al., Editors. 2015: South Dartmouth (MA).
  29. Karpe, F., Postprandial lipemia--effect of lipid-lowering drugs. Atheroscler Suppl, 2002. 3(1): p. 41-6.
  30. Parhofer, K.G., E. Laubach, and P.H. Barrett, Effect of atorvastatin on postprandial lipoprotein metabolism in hypertriglyceridemic patients. J Lipid Res, 2003. 44(6): p. 1192-8.
  31. Parhofer, K.G., P .H. Barrett, and P . Schwandt, Atorvastatin improves postprandial lipoprotein metabolism in normolipidemlic subjects. J Clin Endocrinol Metab, 2000. 85(11): p. 4224-30.
  32. Schaefer, E.J., et al., Effects of atorvastatin on fasting and postprandial lipoprotein subclasses in coronary heart disease patients versus control subjects. Am J Cardiol, 2002. 90(7): p. 689-96.
  33. Tenenbaum, A. and E.Z. Fisman, "If it ain't broke, don't fix it": a commentary on the positive-negative results of the ACCORD Lipid study. Cardiovascular Diabetology, 2010. 9.
  34. Rosenson, R.S., et al., Fenofibrate therapy ameliorates fasting and postprandial lipoproteinemia, oxidative stress, and the inflammatory response in subjects with hypertriglyceridemia and the metabolic syndrome. Diabetes Care, 2007. 30(8): p. 1945- 51.
  35. Ohno, Y., et al., Bezafibrate improves postprandial hypertriglyceridemia and associated endothelial dysfunction in patients with metabolic syndrome: a randomized crossover study. Cardiovasc Diabetol, 2014. 13: p. 71.
  36. Clofibrate and Niacin in Coronary Heart-Disease. Jama-Journal of the American Medical Association, 1975. 231(4): p. 360-381.
  37. Canner, P.L., et al., Fifteen year mortality in Coronary Drug Project patients: long-term benefit with niacin. J Am Coll Cardiol, 1986. 8(6): p. 1245-55.
  38. King, J.M., et al., Evaluation of effects of unmodified niacin on fasting and postprandial plasma lipids in normolipidemic men with hypoalphalipoproteinemia. Am J Med, 1994. 97(4): p. 323-31.
  39. Usman, M.H., et al., Extended-release niacin acutely suppresses postprandial triglyceridemia. Am J Med, 2012. 125(10): p. 1026-35.
  40. Guyton, J.R., et al., Relationship of lipoproteins to cardiovascular events: the AIM-HIGH Trial (Atherothrombosis Intervention in Metabolic Syndrome With Low HDL/High Triglycerides and Impact on Global Health Outcomes). J Am Coll Cardiol, 2013. 62(17): p. 1580-4.
  41. HPS2-THRIVE Collaborative Group, et al., Effects of extended-release niacin with laropiprant in high-risk patients. N Engl J Med, 2014. 371(3): p. 203-12.
  42. Nutescu, E.A. and N.L. Shapiro, Ezetimibe: a selective cholesterol absorption inhibitor. Pharmacotherapy, 2003. 23(11): p. 1463-74.
  43. Masuda, D., et al., Ezetimibe improves postprandial hyperlipidaemia in patients with type IIb hyperlipidaemia. Eur J Clin Invest, 2009. 39(8): p. 689-98.
  44. Olijhoek, J.K., et al., The effects of low-dose simvastatin and ezetimibe compared to high-dose simvastatin alone on post-fat load endothelial function in patients with metabolic syndrome: a randomized double-blind crossover trial. J Cardiovasc Pharmacol, 2008. 52(2): p. 145-50.
  45. Cannon, C.P., et al., Ezetimibe Added to Statin Therapy after Acute Coronary Syndromes. N Engl J Med, 2015. 372(25): p. 2387-97.
  46. Harris, W.S., et al., Omega-3 fatty acids and coronary heart disease risk: clinical and mechanistic perspectives. Atherosclerosis, 2008. 197(1): p. 12-24.
  47. Yokoyama, M., et al., Effects of eicosapentaenoic acid on major coronary events in hypercholesterolaemic patients (JELIS): a randomised open-label, blinded endpoint analysis. Lancet, 2007. 369(9567): p. 1090-8.
  48. Tinker, L.F., et al., (n-3) fatty acid supplementation in moderately hypertriglyceridemic adults changes postprandial lipid and apolipoprotein B responses to a standardized test meal. J Nutr, 1999. 129(6): p. 1126-34.
  49. Park, Y. and W.S. Harris, Omega-3 fatty acid supplementation accelerates chylomicron triglyceride clearance. J Lipid Res, 2003. 44(3): p. 455-63.
  50. Slivkoff-Clark, K.M., A.P. James, and J.C. Mamo, The chronic effects of fish oil with exercise on postprandial lipaemia and chylomicron homeostasis in insulin resistant viscerally obese men. Nutr Metab (Lond), 2012. 9: p. 9.
  51. Feingold KR. Triglyceride Lowering Drugs. 2021 Apr 1. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dhatariya K, Dungan K, Hershman JM, Hofland J, Kalra S, Kaltsas G, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrère B, Levy M, McGee EA, McLachlan R, Morley JE, New M, Purnell J, Sahay R, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000–.
  52. Feingold, K.R. Cholesterol Lowering Drugs. 2021 Mar 30. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dhatariya K, Dungan K, Hershman JM, Hofland J, Kalra S, Kaltsas G, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrère B, Levy M, McGee EA, McLachlan R, Morley JE, New M, Purnell J, Sahay R, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000–
  53. Gaudet, D., et al., Antisense Inhibition of Apolipoprotein C-III in Patients with Hypertriglyceridemia. N Engl J Med, 2015. 373(5): p. 438-47.
  54. Gaudet, D., et al., Targeting APOC3 in the familial chylomicronemia syndrome. N Engl J Med, 2014. 371(23): p. 2200-6.
  55. Ansar, S., J. Koska, and P.D. Reaven, Postprandial hyperlipidemia, endothelial dysfunction and cardiovascular risk: focus on incretins. Cardiovasc Diabetol, 2011. 10: p. 61.
  56. Feingold, K.R. Role of Glucose and Lipids in the Atherosclerotic Cardiovascular Disease of Patients with Diabetes. 2020 Aug 4. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dhatariya K, Dungan K, Hershman JM, Hofland J, Kalra S, Kaltsas G, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrère B, Levy M, McGee EA, McLachlan R, Morley JE, New M, Purnell J, Sahay R, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000–.
  57. Bhatt DL, Steg PG, Miller M, Brinton EA, Jacobson TA, Ketchum SB, Doyle RT Jr, Juliano RA, Jiao L, Granowitz C, Tardif JC, Ballantyne CM; REDUCE-IT Investigators. Cardiovascular Risk Reduction with Icosapent Ethyl for Hypertriglyceridemia. N Engl J Med. 2019 Jan 3;380(1):11-22.
  58. Nissen SE, Lincoff AM, Wolski K, Ballantyne CM, Kastelein JJP, Ridker PM, Ray KK, McGuire DK, Mozaffarian D, Koenig W, Davidson MH, Garcia M, Katona BG, Himmelmann A, Loss LE, Poole M, Menon V, Nicholls SJ. Association Between Achievedω-3 Fatty Acid Levels and Major Adverse Cardiovascular Outcomes in Patients With High Cardiovascular Risk: A Secondary Analysis of the STRENGTH Trial. JAMA Cardiol. 2021 May 16;6(8):1-8
  59. Lombardi M, Carbone S, Del Buono MG, Chiabrando JG, Vescovo GM, Camilli M, Montone RA, Vergallo R, Abbate A, Biondi-Zoccai G, Dixon DL, Crea F. Omega-3 fatty acids supplementation and risk of atrial fibrillation: an updated meta-analysis of randomized controlled trials. Eur Heart J Cardiovasc Pharmacother. 2021 Jul 23;7(4):e69-e70.
  60. Witztum JL, Gaudet D, Freedman SD, Alexander VJ, Digenio A, Williams KR, Yang Q, Hughes SG, Geary RS, Arca M, Stroes ESG, Bergeron J, Soran H, Civeira F, Hemphill L, Tsimikas S, Blom DJ, O'Dea L, Bruckert E. Volanesorsen and Triglyceride Levels in Familial Chylomicronemia Syndrome. N Engl J Med. 2019 Aug 8;381(6):531-542.
  61. Harada-Shiba M, Ali S, Gipe DA, Gasparino E, Son V, Zhang Y, Pordy R, Catapano AL. A randomized study investigating the safety, tolerability, and pharmacokinetics of evinacumab, an ANGPTL3 inhibitor, in healthy Japanese and Caucasian subjects. Atherosclerosis. 2020 Dec;314:33-40.
  62. Tada H, Nomura A, Yoshimura K, Itoh H, Komuro I, Yamagishi M, Takamura M, Kawashiri MA. Fasting and Non-Fasting Triglycerides and Risk of Cardiovascular Events in Diabetic Patients Under Statin Therapy. Circ J. 2020 Feb 25;84(3):509-515
  63. Fan W, Philip S, Granowitz C, Toth PP, Wong ND. Hypertriglyceridemia in statin-treated US adults: the National Health and Nutrition Examination Survey. J Clin Lipidol. 2019 Jan-Feb;13(1):100-108.
  64. Mason RP, Eckel RH. Mechanistic Insights from REDUCE-IT STRENGTHen the Case Against Triglyceride Lowering as a Strategy for Cardiovascular Disease Risk Reduction. Am J Med. 2021 Sep;134(9):1085-1090.
  65. Ference BA, Kastelein JJP, Ray KK, Ginsberg HN, Chapman MJ, Packard CJ, Laufs U, Oliver-Williams C, Wood AM, Butterworth AS, Di Angelantonio E, Danesh J, Nicholls SJ, Bhatt DL, Sabatine MS, Catapano AL. Association of Triglyceride-Lowering LPL Variants and LDL-C-Lowering LDLR Variants With Risk of Coronary Heart Disease. JAMA 2019 Jan 29;321(4):364-373.
  66. Dron JS, Hegele RA. Genetics of Hypertriglyceridemia. Front Endocrinol (Lausanne). 2020 Jul 24;11:455
  67. Olshansky B, Chung MK, Budoff MJ, Philip S, Jiao L, Doyle RT Jr, Copland C, Giaquinto A, Juliano RA, Bhatt DL. Mineral oil: safety and use as placebo in REDUCE-IT and other clinical studies. Eur Heart J Suppl. 2020 Oct 6;22(Suppl J):J34-J48
  68. Chait A, Subramanian S. Hypertriglyceridemia: Pathophysiology, Role of Genetics, Consequences, and Treatment. 2019 Apr 23. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dhatariya K, Dungan K, Hershman JM, Hofland J, Kalra S, Kaltsas G, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrère B, Levy M, McGee EA, McLachlan R, Morley JE, New M, Purnell J, Sahay R, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000–.

 

 

 

 

 

 

Physiology of GnRH and Gonadotropin Secretion

ABSTRACT

 

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

 

INTRODUCTION

 

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

 

The HPG axis anatomically comprises of:

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

 

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

 

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

 

The Discovery of GnRH

 

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

 

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

 

Neuroanatomy of GnRH Neurons

 

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

 

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

 

GnRH Secretion and Pulsatility

 

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

 

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

 

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

 

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

 

Differential Regulation of LH and FSH

 

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

 

Biological and Clinical Relevance of GnRH Pulsatility

 

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

 

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

 

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

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

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

 

Whilst the central role attributed to GnRH remains undisputed, its effective function requires input from other neuronal networks. For instance, the absence of estrogen receptor alpha (ER-alpha) expression on GnRH neurons suggests the need for an intermediate signaling pathway to mediate gonadal steroid feedback (1). The discovery of kisspeptin signaling in neuroendocrine regulation of human reproduction revolutionized the current understanding of the HPG axis. Kisspeptin signaling pathway is increasingly recognized as essential for normal puberty, gonadotropin secretion, and regulation of reproduction (65-67). Other relevant kisspeptin roles have been identified such as regulation of sexual and social behavior, emotional brain processing, mood, audition, olfaction, metabolism, body composition, cardiac function, among others (68-73).

 

Discovery of KNDy Neuronal Network

 

KiSS1, the gene encoding kisspeptins, was first described in 1996 as a suppressor of metastasis in human malignant melanoma (74,75).  This gene was discovered in Hershey and named in accordance to the famous chocolates ‘Hershey’s Kisses’; the inclusion of ‘SS’ is indicative of ‘suppressor sequence’. The KiSS1 gene maps to chromosome 1q32 and includes four exons of which the first two are not translated (76). The gene encodes the precursor 145 amino acid peptide, which is cleaved down to a 54 amino acid peptide. This peptide can be truncated to 14, 13 and 10 amino-acid peptides, all sharing the C-terminal sequence (77,78). These peptides are collectively referred to as kisspeptins - and Kp-10, Kp-13, Kp-14 and Kp-54 are suggested abbreviations for human kisspeptins (79). In 2001, kisspeptins were identified as ligands for the orphan G–protein receptor 54 (GPR54) (80-82), currently named KISS1R (79). KISS1R is localized to human chromosome 19p13.3 and it has five exons, encoding a 398-amino acid protein with seven trans-membrane domains (78,81). Upon binding by kisspeptin, KISS1R activates phospholipase C and recruits intracellular messengers, inositol triphosphate and diacylglycerol, which in turn lead to the release of calcium and activation of protein kinase C (81-83).

 

A reproductive role for kisspeptin in humans became apparent from patients with pubertal disorders which were associated with KISS1R mutations (84,85). A number of inactivating mutations of Kiss1 and Kiss1r have since been reported in animal models with phenotypes characterized by pubertal delay (86). An activating mutation in KISS1Rhas been described in a girl with precocious puberty: when compared to cells with wild-type transfected GPR54, cells with this mutation showed prolonged inositol phosphate accumulation and phosphorylation of extracellular signal–regulated kinase, suggesting extended activation of intracellular signaling by the mutant GPR54 (87). Missense mutations have also been reported in KISS1 gene in three unrelated children with central precocious puberty (88). Functional studies of these mutant peptides demonstrated higher resistance to in vitro degradation but normal affinity to KISS1R, thus suggestive of increased bioavailability as the mechanism by which these abnormal kisspeptins induce precocious puberty (88).

 

A role for neurokinin B in the hypothalamic regulation was also demonstrated when genetic studies in patients from consanguineous families with hypogonadotropic hypogonadism were found to have missense mutations in TAC3 (encodes neurokinin B) and TACR3 (encodes neurokinin B receptor) (89). Other cases have been reported since (90-93).

 

There is also long-standing evidence for the role of opioid system in reproduction. In 1980, Wilkes reported the localization of β endorphin in the human hypothalamus (94). Studies involving the administration of naloxone and naltrexone (opioid antagonists) to humans showed stimulatory effects on LH secretion (95,96), and other studies supported the notion that endogenous opioids play a role in the control of HPG axis (97-101). In 2007, it was demonstrated that dynorphin and kisspeptin are co-localized along with neurokinin B in the same hypothalamic neuronal population in sheep, therefore termed KNDy (Kisspeptin-Neurokinin B-Dynorphin) neurons, highlighting the possible interconnection between these neuropeptides in the control of GnRH and gonadotropin secretion (102,103). The co-localization of kisspeptin, neurokinin B and dynorphin has also been demonstrated in humans (104).

Kisspeptin neurons have also other important neuroanatomical relationships, such as with neuronal nitric oxide synthase neurons as demonstrated in prepubertal female sheep (105), or with somatostatin neurons in the rat hypothalamus (106).

 

Neuroanatomy of KNDy Neuronal Network

 

In humans, kisspeptin neurons are distributed in the rostral Pre-optic Area (POA) and in the infundibular nucleus in the hypothalamus (Figure 2) (104,107). In both male and female autopsy samples, the majority of kisspeptin cell bodies are identified in the infundibular nucleus, and a second dense population of kisspeptin neurons in the rostral POA (104). The infundibular nucleus (arcuate nucleus in non-human species) is similar across species but the rostral region is more species specific (104,108,109). In rodents, the rostral population is located in the anteroventral periventricular nucleus (AVPV) and the periventricular nucleus, the continuum of this region named as the rostral periventricular region of the third ventricle (RP3V) (108,110). Humans and ruminants lack this well-defined RP3V population of kisspeptin neurons, which are more scattered within the preoptic region (109,111).

 

Kisspeptin axons form dense plexuses in the human infundibular stalk, where the secretion of GnRH occurs (104). Axo-somatic, axo-dendritic and axo-axonal contacts between kisspeptin and GnRH axons were demonstrated at this level, showing that kisspeptin and GnRH networks are in close proximity (104,112). Moreover, GnRH neurons express Kiss1r mRNA, reinforcing the notion of kisspeptin involvement in GnRH secretion (113-115).

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

Three-quarters of kisspeptin-immunoreactive cells in the human infundibular nucleus of the hypothalamus co-express neurokinin B and dynorphin (KNDy neurons) (104,116). KNDy neurons in rodents and ruminants are localized in the arcuate nucleus of the hypothalamus. However, neurokinin B and dynorphin are absent from kisspeptin neurons in the hypothalamic POA (Figure 2) (67,111). This differential expression of neuropeptides may reflect distinct functions of these two kisspeptin populations.

 

Significant kisspeptin expression was also demonstrated in extra-hypothalamic sites, including in limbic and paralimbic brain regions, such as medial amygdala, cingulate, globus pallidus, hippocampus, putamen and thalamus, key areas of neurobiological control of sexual and emotional behaviors (reviewed in detail in (117)), as well as peripherally in organs like ovary, testis, uterus and placenta where the kisspeptin system may also play a part in reproduction function (118,119).

 

Interactions Between Kisspeptin, Neurokinin B and Dynorphin

 

KDNy neurons act synergistically to induce coordinated and pulsatile GnRH secretion by regulating the neuroactivity of other KDNy cells. This is supported by the existence of neurokinin B and kappa opioid peptide receptors (receptor for dynorphin) within the KNDy cells, but not kisspeptin receptors, which are predominantly expressed on GnRH neurons (116,120,121). Neuron-neuron and neuron-glia communications via gap junctions contribute for the synchronized activities among KNDy neurons (122).  

 

Neurokinins (A and B) are members of the tachykinin family of peptides, which stimulate three related GPCRs (encoded by TACR1, TACR2 and TACR3) (123). Neurokinin B acts predominantly on TACR3. Neurokinin B stimulates kisspeptin neurons, which in turn lead to GnRH secretion (67,124). Neurokinin B signaling regulates GnRH/LH secretion in healthy women, and it is crucial for the mediation of the estrogenic positive and negative feedback on LH secretion (125-127). There is rapidly increasing interest in the therapeutic value of neurokinin antagonists in several indications in reproductive health, recently reviewed in (128).

 

In women with polycystic ovary syndrome, the administration of neurokinin 3 receptor antagonists markedly reduced serum LH concentration and pulse frequency, as well as serum testosterone (129-131). A recent study confirmed a complex crosstalk between neurokinin B and kisspeptin pathways in the regulation of GnRH secretion in polycystic ovary syndrome. In this study, kisspeptin-10 infusion given to women with polycystic ovary syndrome increased LH secretion with a direct relationship to estradiol exposure. Neurokinin 3 receptor antagonism reduced LH secretion and pulsatility, and whilst LH response to kisspeptin-10 was preserved, its relationship with circulating estradiol was not. More interestingly, although kisspeptin-10 increased LH pulse frequency, changes in other parameters of LH secretory pattern were prevented when co-administered with neurokinin 3 receptor antagonists (131).

 

In postmenopausal women,  seven day treatment with neurokinin 3 receptor antagonist decreased LH secretion, but not FSH secretion, as well as lead to a remarkable reduction in hot flushes (132). Neurokinin 3 receptor antagonism efficiency in treating menopausal hot flushes has been also demonstrated in other clinical trials (133,134), thus supporting its therapeutical role in menopausal vasomotor symptoms (135-137). In healthy men, neurokinin B signaling display a central role for the reproductive function, and this is functionally upstream of kisspeptin-mediated GnRH secretion: LH, FSH and testosterone secretion decreased during the administration of a neurokinin 3 receptor antagonist, while kisspeptin-10 administration restored LH secretion to the same degree before and during neurokinin 3 receptor antagonist treatment (138).

 

An increase in the expression of Kiss1 in the hypothalamic neurons was observed following senktide (agonist of neurokinin B) administration (139), and its stimulatory effects were abolished in Gpr54 knock-out male (140). In ovariectomized goats, neurokinin B stimulated LH secretion through electrical multi-unit activity corresponded to LH secretion, suggesting a hypothalamic site for this GnRH pulse generation (141). GnRH antagonists abolished the stimulatory effect of neurokinin B, demonstrating its site of action to be functionally higher than the GnRH receptor (142,143).

 

Studies involving the administration of opioid antagonists to humans have shown stimulatory effects on LH secretion in late follicular and mid-luteal phase (95,96), and together with other studies (97-101), highlight the inhibitory input by dynorphins on kisspeptin signaling, and consequently on GnRH/gonadotropin secretion. Through the stimulatory effects of neurokinin B and kisspeptin, and the inhibitory action of dynorphin, these neuropeptides coordinate pulsatile GnRH and LH secretion (Figure 2) (144,145).

 

Kisspeptin-mediated GnRH secretion is sex steroid dependent. Estrogen and progesterone modulate kisspeptin activity though the sex-steroid receptors expressed on kisspeptin neurons at both AVPV and the arcuate nucleus (146-148). Furthermore, two distinct populations of kisspeptin neurons, the infundibular/arcuate region of which interacts with neurokinin B and dynorphin, appear to mediate distinct sex-steroid pathways (discussed in more detail in sections 4.1-4.4). Briefly, in humans, KNDy neurons in the infundibular nucleus alone are involved in negative and positive sex-steroid feedback, whereas in rodents positive sex-steroid feedback seems to be mediated via kisspeptin neurons in the AVPV region and negative sex-steroid feedback via the arcuate KNDy neurons (Figure 2) (67,107,148,149).

 

Stimulatory Effect of Kisspeptin on GnRH and Gonadotropin Secretion

 

Kisspeptin is a potent stimulator of the HPG axis – and in fact, it is the most potent GnRH secretagogue currently known. Kisspeptin signals directly to the hypothalamic GnRH neurons via kisspeptin receptor to release GnRH into the portal circulation, which in turn stimulates the anterior pituitary gonadotropes to produce LH and FSH (124,150).

 

The stimulatory effects of kisspeptin on LH secretion have been documented in animal models (151-154). This is consistent with human studies, where kisspeptin increases both LH and FSH secretion with the preferential stimulatory effect on the former (67,155-165). Kissppetin-54 was first administered in healthy men as an intravenous infusion with dose-dependent rise in LH secretion (157). Since then kisspeptin was administered in different isoforms (kisspeptin-54 and kisspeptin-10), different routes (subcutaneous and intravenous), different types of exposure (continuous and bolus), to healthy men and women and in endocrine disease models with low gonadotropin output, all showing stimulatory effect of kisspeptin on LH secretion (fully reviewed in (67)).

 

Pulsatile GnRH secretion correlates with LH pulsatility, prompting investigation of the effect of kisspeptin on regulating LH pulse frequency. LH pulse frequency and amplitude were increased following intravenous infusion of kisspeptin-10 in healthy men (160), and subcutaneous bolus of kisspeptin-54 in healthy women (162). The hypothalamic response to kisspeptin-54 and the pituitary response to GnRH are preserved in healthy older men (166). Kisspeptin also stimulates LH pulse frequency in reproductive endocrine disorders of low LH pulsatility, including hypothalamic amenorrhea, defects in the neurokinin B pathway and hypogonadal men with diabetes (93,167,168). Indeed, kisspeptin-54 and kisspeptin-10, as well as kisspeptin agonists like MVT-602 (previously known as TAK-448) are able to stimulate physiological reproductive hormone secretion in individuals with functional hypogonadism related to deficient GnRH secretion, such as in hypothalamic amenorrhea or polycystic ovary syndrome (169,170).

 

Kisspeptin regulates GnRH and subsequently gonadotropin secretion through Kiss1r, as suggested by Messager who demonstrated no detectable LH levels in response to kisspeptin in Kiss1r knockout mice (115). The prevention of the stimulatory effect of kisspeptin on LH secretion by GnRH antagonists indicate that kisspeptin action is GnRH-mediated (114,152,171-173). This is further supported by the observation that kisspeptin cause depolarization of GnRH neurons (113) and stimulate GnRH release from hypothalamic explants (174,175). The expression of GnRH mRNA is upregulated in GnRH neurons following kisspeptin administration (176). Moreover, in patients with impaired functional capacity of GnRH neurons (idiopathic hypogonadotropic hypogonadism), the same dose of kisspeptin failed to induce LH response seen in healthy men and women (177). In female rats, ablation of KNDy neurons resulted in hypogonadotropic hypogonadism, confirming its role in the maintenance of normal LH levels and to estrous cyclicity (178).

 

Some investigators have demonstrated a direct stimulatory effect of kisspeptin on gonadotropes, but this direct stimulatory action of kisspeptin on gonadotropes remains debatable (179-183). Kiss1 and Kiss1r gene expression has been shown in gonadotropes, and gonadotropin secretion from the pituitary explants was observed following exposure to kisspeptin (77,179-182). Moreover, LHβ and FSHβ gene expression was upregulated in the primary pituitary cells treated with kisspeptin. Whilst kisspeptin can directly regulate gonadotropins at the transcriptional level, it appears to be less relevant than the GnRH-mediated action (67,182,183).

 

Desensitization Effect of Chronic or Continuous Exposure to Kisspeptin

 

Continuous administration of GnRH desensitizes the HPG axis by downregulation of GnRH receptors and desensitization of gonadotropes, following an initial stimulatory effect (39). It is therefore important to ascertain the effects of continuous exposure to kisspeptin on the HPG axis. Efforts have been made to assess the impact of continuous infusions of kisspeptin in a number of animal experiments (115,184-187). In adult rats, continuous administration of kisspeptin-54 increased serum LH and free testosterone on day one, but this stimulatory effect was lost after 2 days, indicative of kisspeptin receptor desensitization (187). In rhesus monkeys, the continuous administration of kisspeptin-10 resulted in suppression of LH secretion, indicating desensitization of kisspeptin receptor (185). The kisspeptin receptor has been shown to desensitize in vitro (184). In sheep, infusion of kisspeptin-10 resulted in acute increase in serum LH levels, which declined by the end of 4 hour infusion, while GnRH remained elevated following the discontinuation of kisspeptin-10 administration. This suggests that desensitization to GnRH could be occurring at the level of pituitary gonadotropes (115).  

 

Consistent with animal studies, Jayasena et al. demonstrated that in women with hypothalamic amenorrhea an initial increase in LH and FSH secretion was not sustained following twice daily subcutaneous kisspeptin-54 administration for two weeks (164). Other studies in humans employing continuous or repeated kisspeptin administration provide conflicting evidence for kisspeptin-mediated desensitization and appear to be dose-related (160,168). High doses of kisspeptin may induce desensitization, but this is not apparent at lower doses (67). Sustained LH secretion and increased LH pulsatility was demonstrated with lower dose of kisspeptin-54 (0.01-1nmol/kg/h) infusion for 8 hours in women with hypothalamic amenorrhea (168) and kisspeptin-10 (3.1 nmol/kg/h) infusion for 22.5 hours in healthy men (160). In contrast, LH secretion was not maintained in three healthy men during the 24 hour infusion of kisspeptin-10 at 9.2 nmol/kg/h (the highest dose used in humans so far), although serum LH did not fall to the castrate levels and remained well above baseline at end of infusion (188).

 

Kisspeptin receptor agonist analogues, TAK-488 and TAK-683, induce desensitization when administered to healthy men (189,190). However, the ability of natural kisspeptin fragments to downregulate the HPG axis in humans remains to be established, and is to date complicated by differences in study protocols, in terms of isoform of kisspeptin used, duration (8 hours-2 weeks), mode and route of kisspeptin administration, lower doses of kisspeptin in human studies compared to animal, and the endocrine profile of the study participants (men versus women versus hypothalamic amenorrhea). 

 

Sexual Dimorphism in Kisspeptin Signaling

 

The response to kisspeptin is different in men and women. In men, kisspeptin potently stimulates the release of LH, but in women the effect of kisspeptin is variable and dependent on the phase of menstrual cycle (67). Whilst men respond to the modest doses of kisspeptin, LH response to kisspeptin in healthy women is minimal and inconsistent in the early follicular phase but greatest in the pre-ovulatory phase of the menstrual cycle (157-159,165). This indicates that in addition to the fluctuations in sex-steroid milieu, other mechanisms, such as changes in pituitary sensitivity to GnRH or GnRH network responsiveness to kisspeptin regulate the sensitivity to kisspeptin throughout the menstrual cycle (67,121,191).

 

Not only there is sexual dimorphism in gonadotropin response to kisspeptin, but there are also anatomical differences. Female hypothalami have significantly more kisspeptin fibers and kisspeptin cell bodies than men (161). Only a few kisspeptin cell bodies are present in the male infundibular nucleus and none in the rostral periventricular nucleus, which is on contrary to the female hypothalami with abundant kisspeptin network in both of these hypothalamic nuclei (104). These sex differences in kisspeptin neurons appear to be established early during perinatal development through the action of sex steroids (121,192).

 

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

 

Kisspeptin, GnRH and Puberty

 

Kisspeptin is crucial for normal pubertal development, the discovery of which formed the basis for the obligate role of kisspeptin signaling in the control of reproductive function (193). More than a decade ago two independent groups identified ‘inactivating’ mutations in KISS1R in patients with hypogonadotropic hypogonadism presenting with pubertal delay (84,85). Recently, a male patient with a biallelic loss-of-function KISS1R mutation was described who had undergone a normal and timely puberty, although as a child he had presented with microphallus and bilateral cryptorchidism. This suggests different levels of dependence of the hypothalamic-pituitary-gonadal axis on kisspeptin signaling during the reproductive life span, with the mini-puberty of infancy appearing more dependent on the kisspeptin system than is adolescent puberty (194). On the other hand, activating mutations in KISS1R and KISS1were then described in children with central precocious puberty (87,88).

 

Hypothalamic expression of Kiss1 and Kiss1R mRNA is upregulated at puberty (113,153,195), and the percentage of GnRH neurons depolarizing in response to kisspeptin increases from juvenile (25%) to pubertal (50%) and to adult mice (>90%) (113), suggesting that GnRH neurons may acquire sensitivity to kisspeptin across puberty. In monkeys, kisspeptin-54 secretion and pulsatility increased at the onset of puberty (196). Moreover, the exogenous administration of kisspeptin resulted in earlier puberty in rats and monkeys (195,197), whereas kisspeptin antagonists delayed puberty in rats (173) and inhibited GnRH release in pubertal monkeys (198). In other study, daily injections of a synthetic kisspeptin analog has been shown to significantly advance puberty in prepubertal female mice (199). GnRH neuron-specific Kiss1r knockout mouse showed a delay in pubertal onset, abnormal estrous cyclicity in female and abnormal external genitalia in male (microphallus, decreased anogenital distance associated with failure of preputial gland separation) (200).

 

Exogenous kisspeptin stimulated GnRH-induced LH secretion in patients with hypogonadism resulted in a spontaneous and permanent activation of their hypothalamic-pituitary-gonadal axis, whereas patients with idiopathic hypogonadotropic hypogonadism and no spontaneous LH pulsatility did not respond to kisspeptin, suggesting that the reversal of hypogonadism, sexual maturation and puberty may well be associated with the acquisition of kisspeptin responsiveness which in turn signals the emergence of reproductive endocrine activity (201). A recent study, 15 children with delayed puberty were administered intravenous kisspeptin and displayed divergent responses, with seven subjects having no response to kisspeptin, whereas others having either robust response (comparable to those of adults) or intermediate responses as perceived in one case (202).

 

GnRH release during puberty appears to require a cooperative mechanism between the kisspeptin and NKB networks. Agonists and antagonists of kisspeptin and NKB were administered into the stalk-median eminence (region with high concentration of GnRH, kisspeptin and NKB neuroterminal fibers), and it was found that both kisspeptin-10 and the NK3R agonist senktide stimulated GnRH release in a dose-responsive manner in prepubertal and pubertal monkeys. However, senktide-induced GnRH release was blocked in the presence of a KISS1R antagonist and the kisspeptin-induced GnRH release was blocked in the presence of NK3R antagonist in pubertal monkeys, leading to the notion that a reciprocal signaling mechanism between kisspeptin and NKB exists and is possibly necessary for a normal puberty (203).

 

These data together emphasizes that disrupted kisspeptin-GPR54-NKB signaling leads to hypogonadotropic hypogonadism, reinforcing the critical role of kisspeptin in puberty.

 

REGULATION OF GnRH AND GONADOTROPIN SECRETION

 

Development and maintenance of normal reproductive function requires a coordinated interplay between neuroendocrine, metabolic and environmental factors. The GnRH-gonadotropin system plays a central role in the regulation of reproduction by integrating different signals and factors (Figure 3) (121,191). 

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

Overview of Sex Steroid Feedback

 

A crucial role for sex steroids in the regulation of GnRH neurons and/or gonadotropes in humans was initially proposed as serial blood sampling and gonadotropin assays in women through phases of menstrual cycle showed an uneven distribution, with a clear mid-cycle surge in LH and FSH. Two mechanisms were proposed to mediate this effect: first, GnRH secretion is altered in response to the steroid milieu; second, sensitivity of the gonadotropes to a GnRH input is sex-steroid dependent, although the exact mechanism remains controversial due to inter-species variation (204).

 

Hypothalamic secretion of GnRH increases during proestrus in rats (205), sheep (206) and non-human primates (207). Pulsatile once hourly administration of exogenous GnRH restored ovulation in Rhesus monkeys with hypothalamic lesions which abolished GnRH secretion, suggesting that it was the ‘ebb and flow’ of ovarian estrogen feedback acting directly on the pituitary which triggered an LH surge (208). In humans, endogenous GnRH secretion is potentially diminished during the pre-ovulatory LH surge and the suppression of gonadotropin secretion is greater with lower doses of a GnRH receptor antagonist during the mid-cycle surge in comparison to the other phases of the menstrual cycle (209). This suggests that pituitary gonadotrope sensitivity to GnRH is enhanced during the mid-cycle surge. Administration of exogenous estradiol or testosterone in men with hypogonadotropic hypogonadism receiving pulsatile GnRH therapy, decreased gonadotropin concentrations, demonstrating inhibitory effects of sex-steroids at the level of pituitary (210). A direct effect of estrogen on gonadotropes is further demonstrated by the inhibition of LH secretion from rat pituitary gonadotropes in vitro (211). Literature to date suggests that there is a dual-site sex-steroid feedback in the regulation of gonadotropin secretion, occurring at the level of both pituitary and hypothalamus (212-217).

 

Estrogen Feedback

 

Patterns of GnRH and LH secretion across the menstrual cycle are modulated by estradiol feedback. A biphasic effect of estradiol on gonadotropin secretion has long been established and it is essential for normal menstrual cycle, with an initial negative feedback (greater suppression of FSH) and a subsequent positive feedback (more prominent for LH) (32). However, the basis for estrogen feedback has been long unclear. GnRH neurons do not express estrogen receptor alpha (ER-alpha) (218,219), and therefore a mediator between gonads and hypothalamus was missed. Recent evidence suggests that kisspeptin and neurokinin B (126) appears to be providing this “missing link” as a key regulator of both negative and positive estrogen feedback (67,121).

 

KNDy neurons in the infundibular nucleus in humans and the arcuate nucleus in other mammals mediate negative estrogen feedback. Estrogen suppresses kisspeptin and neurokinin B release from KNDy neurons, which reduce their stimulatory input to GnRH neurons. Simultaneously, there is a relative deficiency in dynorphin signaling as part of this negative feedback, releasing the inhibitory action on kisspeptin signaling (Figure 2) (67). Immunohistochemical staining of the postmenopausal female hypothalami showed up-regulated expression of KISS1 mRNA and hypertrophy of kisspeptin neurons in the infundibular nucleus when compared to the premenopausal women (107). These hypertrophied kisspeptin neurons co-localized with ER-alpha, had increased expression of neurokinin B and decreased levels of prodynorphin mRNA (220-222). The above evidence for the involvement of the infundibular KNDy system in mediating negative estrogen feedback in humans is consistent with animal studies. Kisspeptin neurons in the arcuate nucleus show frequent co-localization with ER-alpha (148,223). In ovariectomized animals, the expression of Kiss1 and neurokinin B mRNA was up-regulated but prodynorphin mRNA reduced in the arcuate nucleus (equivalent to the infundibular nucleus in humans), and this was reversed by estrogen replacement (99,111,116,224-228). Postmenopausal women are resistant to the stimulatory effect of kisspeptin on LH secretion (132,229), but postmenopausal women receiving estradiol replacement therapy are only resistant to kisspeptin initially and then they do demonstrate a remarkable increase in LH pulse amplitude with direct correlation to the circulating levels of estradiol and duration of kisspeptin administration (229). However, neurokinin B regulates gonadotropin secretion in postmenopausal women, and antagonizing the neurokinin 3 receptor modestly decreases LH secretion in this context (132). Interestingly, the use of fezolinetant (a neurokinin 3 receptor antagonist) has been shown to effectively reduce the menopause-related vasomotor symptoms owing to its inhibitory effect in the hypothalamic thermoregulatory center, and thus presenting a potential non-hormonal treatment option for menopausal women (134).

 

Negative estrogen feedback switches to positive feedback in the late follicular phase of menstrual cycle, in order to induce the pre-ovulatory LH surge. Recent evidence support the role of kisspeptin in generating the LH surge: during an assisted conception cycle, kisspeptin-54, used instead of a routinely administered human chorionic gonadotropin, induced an LH surge, and oocyte maturation, with a subsequent live term birth (227). Repeated twice-daily administration of kisspeptin-54 shortened the menstrual cycle, suggesting that the onset of LH surge was advanced (161). This is further supported by antagonistic studies in animal models, where the administration of kisspeptin antiserum or antagonists blunt/prevent LH peak, whilst kisspeptin advances LH surge (198,230,231). However, kisspeptin-mediated positive estrogen feedback has marked anatomical variations between humans and other species. In rodents, positive estrogen feedback is mediated via the AVPV nucleus, which is absent in humans, other primates and sheep (Figure 2). The expression of Kiss1 mRNA in the AVPV nucleus is low following an ovariectomy, but is dramatically increased with estrogen treatment and at the time of LH surge (148,149). In sheep, positive estrogen feedback is mediated though the arcuate nucleus, where the expression of Kiss1 mRNA is the greatest at the pre-ovulatory LH surge (182). There are no studies looking at the anatomical region of estrogen mediating positive feedback in humans. Although there does not appear to be two distinct anatomical populations of kisspeptin neurons to relay negative and positive sex-steroid feedback in humans, it is possible that separate signaling pathways exists to mediate gonadal steroid feedback.

 

Whilst it is clear that kisspeptin is involved in estrogen-induced mid-cycle gonadotropin surge, the role of KNDy neurons in positive estrogen feedback is less obvious. In sheep, the expression of neurokinin B mRNA was increased during the LH surge, and neurokinin B receptor agonist senktide induced LH secretion mimicking its mid-cycle surge (232,233). However, this has not been reproduced in other species, including humans (168). In summary, KNDy neurons mediate negative estrogen feedback in the infundibular nucleus in humans and the arcuate nucleus in other species. Positive estrogen feedback is mediated via kisspeptin neurons, which show marked inter-species anatomical variation.

 

In addition to the gonads, the brain is one of the major organs producing estradiol, and recently a number of studies demonstrated that estradiol is synthesized and released in the hypothalamus (i.e. neuroestradiol) contributing to the regulation of GnRH release, particularly regarding its positive feedback effect during the preovulatory GnRH/LH surge (234).

 

Progesterone Feedback

 

Progesterone reduces LH pulse frequency in healthy women. LH secretory pattern in women exposed to exogenous progesterone was comparable to LH profile observed in the mid-luteal phase, demonstrating that progesterone plays a central role in the luteal phase of menstrual cycle (235). These inhibitory effects of progesterone on gonadotropin secretion are mediated by the progesterone receptor (PR) (236). The suppressive effect of progesterone on LH secretion was diminished in the context of estrogen deficiency, while co-administration of estradiol restored it (236), suggesting an interplay between these sex steroids. However, the presence of PR on only a small subset of GnRH neurons (237-239) led to the notion that intermediaries are involved in mediating inhibitory progesterone signal to GnRH neurons.

 

There is evidence that KNDy neurons play a role in mediating progesterone feedback on GnRH through dynorphin signaling (Figure 2) (99,116). PR have been demonstrated to be co-localized with dynorphin in the KNDy neurons (147) and progesterone increased dynorphin concentrations (240). Moreover, the number of preprodynorphin mRNA expressing cells decreased in postmenopausal women (222) and in ovariectomized ewes, but normalized with exogenous progesterone to luteal levels (240).

 

Testosterone Feedback

 

Testosterone exerts negative feedback on gonadotropin secretion. Early studies verified that LH and FSH pulse frequency are enhanced in hypogonadal men and exogenous testosterone decreases gonadotropin secretion, suggesting that testosterone have an inhibitory effect on GnRH secretion (216,241).

 

Few GnRH neurons express androgen receptors (AR) (242). GnRH neurons were thus considered to be reliant on an intermediary neuronal population to mediate testosterone feedback. A key role for KNDy neurons in this mediation has been suggested, as these neurons express AR which directly mediate the androgen feedback. The androgen feedback may also rely on the aromatization of testosterone, as testosterone-induced suppression of Kiss1 mRNA in the rodent arcuate nucleus is identical to that observed with estradiol, but more than that observed with dihydrotestosterone administration (243). The cross-talk between AR and ER was suggested from animal studies: AR expression was downregulated in the prostate following neonatal estrogen exposure (244), and AR transcription was modulated following a co-transfection of AR and ER (245).

 

Navarro has described a role for KNDy neurons in mediating the negative testosterone feedback on GnRH secretion, and provided evidence that neurokinin B released from KNDy neurons is part of an auto-feedback loop that generates the pulsatile secretion of Kiss1 and GnRH in male mice: Kiss1 and dynorphin mRNA are regulated by testosterone through estrogen and androgen receptor-dependent pathways; KNDy neurons express neurokinin B receptor whereas GnRH neurons do not, and senktide (an agonist for the neurokinin B receptor) activates KNDy neurons leading to gonadotropin secretion but has no discernible effect on GnRH neurons (246). Other studies demonstrated that the suppression of gonadotropin secretion using testosterone is associated with a reduction of Kiss1 mRNA in the hypothalamus (114,195,247). Moreover, post-orchidectomy rise in LH in rodents can be blocked by kisspeptin antagonists, further suggesting that kisspeptin system mediates the hypothalamic androgen feedback (173).

 

Stress and Glucocorticoids

 

Physical and psychological stress is associated with hypothalamic amenorrhea, possibly though the activation of hypothalamic-pituitary-adrenal (HPA) axis (248,249). Experimental evidence points towards a cortisol-mediated suppression of gonadotropin secretion as the main key pathway to explain stress-induced gonadotropin suppression(55,250-257). The negative effect of cortisol on HPG axis is recognized to occur at both pituitary and hypothalamic levels. There are also data suggesting that upstream factors in the HPA axis, such as Corticotropin Releasing Hormone (CRH) and vasopressin may play a mediatory role (258,259).

 

Cortisol secretion in women with hypothalamic amenorrhea is elevated (251), and evening adrenocorticotropic hormone (ACTH) and cortisol concentrations are higher in excessive exercise (250,254). Administration of exogenous glucocorticoids to eugonadal women was associated with a decrease in LH pulse frequency, suggesting that glucocorticoids have a negative action on GnRH secretion (257). In ovine portal blood, cortisol administration led to a decrease in GnRH pulse frequency (256). Inferences of cortisol effects on gonadotropin secretion were also derived from observations in women and men with Cushing’s syndrome (condition associated with excessive cortisol secretion), where exogenous GnRH preferentially stimulates FSH whilst LH remains unchanged (252,255). The resolution of male hypogonadotropic hypogonadism was also observed in men with remission of Cushing’s disease (255).   

 

This negative input of cortisol on the HPG axis may be modulated by sex-steroid hormones, and kisspeptin signaling has also been implicated in the process. Cortisol alone had no impact on GnRH pulsatility in ovariectomized ewes, but the co-administration of estradiol and progesterone led to a 70% decrease in GnRH secretion (256). Decreased hypothalamic Kiss1 mRNA expression has been observed during exposure to stress or exogenous glucocorticoids. The role of kisspeptin in mediating stress inputs is further supported by the expression of glucocorticoid receptor on murine kisspeptin neurons (260).

 

Hypothalamic CRH neurons, important regulators of the stress response, also directly modulate GnRH excitability in a dose-dependent and receptor-specific manner, and the GnRH response to CRH is influenced by estrogens (261). Intracerebroventricular administration of CRH in female rats suppressed LH pulsatility and the LH surge, and this suppression was enhanced by estrogens (262).

 

Prolactin

 

Prolactin is a well-known inhibitor of GnRH release and a suppressor of the HPG axis. The association between hyperprolactinemia and reproductive dysfunction has long been established, accounting for 14% of secondary amenorrhea and hypogonadism cases (263) and for a third of women presenting with infertility (264,265). Hyperprolactinemia is evident in 16% of men with erectile dysfunction and in 11% of men with oligospermia (266). The decreased pulsatility of LH in hyperprolactinemia responds to bromocriptine (267). GnRH therapy has restored ovulation and normal luteal function in bromocriptine resistant hyperprolactinemic women (268,269), suggesting that prolactin exerts inhibition through direct reduction of GnRH secretion.

 

The neuroendocrine pathway by which prolactin inhibits GnRH pulse frequency remains to be fully elucidated. A direct action of prolactin on the GnRH neuronal network is possible (270,271). Prolactin has also been demonstrated to influence other systems, including GABA (272), β endorphins (273), neuropeptide Y (274) and dopaminergic systems (275). Recent data suggest that kisspeptin signaling may be involved too, as kisspeptin neurons express prolactin receptors (276). In rodent models, kisspeptin neurons in the arcuate nucleus modulate dopamine release from dopaminergic neurons, thereby regulating prolactin secretion (277). Kiss1 expression is decreased in lactation, a physiological state associated with hyperprolactinemia (278). Prolactin-sensitive GABA and kisspeptin neurons were identified in regions of the rat hypothalamus (276). Moreover, in a mouse model of anovulatory hyperprolactinemia (induced by a continuous infusion of prolactin), Kiss1 mRNA levels were diminished and peripheral administration of kisspeptin restored gonadotropin secretion and ovarian cyclicity (279). There are also other animal studies reporting an inhibitory effect of prolactin on Kiss1 expression (280,281). This data suggests that kisspeptin is a possible link between hyperprolactinemia and GnRH deficiency. The administration of kisspeptin-10 reactivated the gonadotropin secretion in women with hyperprolactinemia-induced hypogonadotropic amenorrhea, suggesting that GnRH deficiency in the context of hyperprolactinemia is, at least in part, mediated by an impaired hypothalamic kisspeptin secretion (282).

 

On the other hand, kisspeptins appears to have a stimulatory effect on prolactin release, as demonstrated in a recent study in ovariectomized rats which had intracerebroventricular injections of kisspeptin-10 with subsequent increase in prolactin release, and this required the estrogen receptor-alpha and was potentiated by progesterone via progesterone receptor activation (283).

 

Nutrition and Metabolism

 

A link between energy balance and reproductive function enables organisms to survive to reproductive maturity and to withstand the energy needs of parturition, lactation and other parental behaviors. This link optimizes reproductive success under fluctuating metabolic conditions (284). Kisspeptin signaling may link nutrition/metabolic status and reproduction by sensing energy stores and translate this information into GnRH secretion (285). These relations elucidate further associations between reproductive dysfunction and metabolic disturbances, such as diabetes, obesity or anorexia nervosa (67,286,287).

 

Food deprivation impairs GnRH and gonadotropin secretion, and leptin (a satiety hormone secreted by adipose tissue, the levels of which drop in response to fasting) plays a role in this inter-regulation by stimulating LH release (67,288-290). Periods of fasting and calorie restriction decrease LH pulse frequency and increase pulse amplitude (284,291-293). Administration of recombinant leptin increased LH pulse frequency in women with hypothalamic amenorrhea (294) and prevented fasting-induced drop in testosterone and LH pulsatility in healthy men (295). Moreover, humans with mutations in leptin or in leptin receptor show hypogonadism (296). Thus, the crosstalk between kisspeptin and leptin is relevant for reproduction and fertility (71), including in the setting of assisted reproduction techniques (297).

 

Kisspeptin neurons may have a role in mediating the metabolic signals of leptin on the control of HPG axis, as 40% of the arcuate kisspeptin neurons express leptin receptors in contrast to the GnRH neurons, where leptin receptors are absent (298-301). Food deprivation is associated with a decrease in kisspeptin, and subsequent reduction in gonadotropin secretion (302-305). Levels of low Kiss1 mRNA expression in the leptin-deficient mice are partially upregulated by leptin (149). Moreover, exogenous kisspeptin restored vaginal opening (marker of sexual maturation) in malnourished rodents (302). Animal models of type 1 diabetes, characterized by insulin deficiency and impaired cellular nutrition, had hypogonadotropic hypogonadism and decreased Kiss1 mRNA expression. Repeated administration of kisspeptin to these rodents increased prostate and testis weight (306). It is plausible that a relative deficiency of kisspeptin secretion is a mechanism for hypogonadotropic hypogonadism in patients with obesity and diabetes (167). In hypogonadal men with type 2 diabetes, kisspeptin-10 increased LH secretion and pulse frequency(167). Although early studies appeared to suggest a direct link between kisspeptin and leptin, it seems that the neuronal pathway whereby leptin modulates GnRH is far more complex (307,308). Only partial restoration in Kiss1mRNA in leptin-deficiency and normal pubertal development and fertility observed in selective leptin receptor deletion from kisspeptin neurons suggest that kisspeptin may link reproduction and metabolism through other ways than leptin (149,309). Proopiomelanocortin (POMC), agouti-related peptide, neuropeptide Y, ghrelin, and cocaine- and amphetamine-regulated transcript (CART) expressing neurons have been linked to this process (285,301). Kisspeptin neurons communicate with POMC and neuropeptide Y neurons and are able to modulate the expression of relevant genes in these cells (298). Several studies have suggested that ghrelin can interact directly with hypothalamic neurons leading to suppression of gonadotropins release, and thus impairing fertility, an effect that is dependent of the estradiol milieu (285,310-312).

 

GABA (Gamma-Amino Butyric Acid)

 

GABA has also been implicated as a regulator of GnRH secretion. Although GABA is classically an inhibitory neurotransmitter in the central nervous system, most mature GnRH neurons are stimulated by GABA, which has attributed to GABA an excitatory action in HPG axis. The precise physiology of this mechanism is still unclear (313-317), but it may be related to the bidirectional interactions between GABA and kisspeptin pathways, as well as between these and GnRH neurons, in a variety of ways throughout development (318). In early development, GABA seems to increase KISS1 expression in embryonic phase and early postnatally, while in the absence of GABAergic input the expression of KISS1 declines (318,319). In the prepubertal period, the central restraint on GnRH secretion seems to be mediated by GABA possibly acting directly via kisspeptin neurons (318). In the peri-pubertal phase, the antagonism of GABA and the intrinsic disinhibition of kisspeptin neurons seem to be critical in puberty initiation and development (320,321). In the adulthood, the interactions between GnRH-GABA-kisspeptin become more complex with HPG axis function critically dependent on such interactions. For instance, the preovulatory surge does not occur in the absence of GABA signaling, thus neurons co-expressing GABA and kisspeptin seem crucial in providing double excitatory input to GnRH neurons at the time of ovulation (318,322).

 

Other Neuropeptides

 

In addition to KNDy system and GABA, other peptides and neurotransmitters have been shown to influence GnRH-gonadotrope system: vasoactive intestinal polypeptide (VIP), vasopressin, catecholamines, nitric oxide, neurotensin, gonadotropin-inhibitory hormone (GnIH) /RFamide related peptide-3 (RFRP-3) (317), nucleobindin-2/nesfatin-1 (323). Excitatory inputs to the HPG axis may be mediated by VIP, catecholamines, glutamate and possibly vasopressin, whereas GnIH in birds, or its mammalian homolog RFRP-3, provide inhibitory inputs (324-328). RFRP neuronal populations have been detected mainly in the hypothalamic dorsomedial nucleus or adjacent regions, and they have projections to several hypothalamic areas including the arcuate nucleus, paraventricular nucleus, ventromedial nucleus and the lateral hypothalamus, all areas with major roles in the regulation of reproduction and energy balance (329,330). RFRP-3, encoded by the gene Rfrp, inhibits the electric firing of GnRH and kisspeptin neurons (325,331), which results in a suppression of GnRH-induced gonadotropin release with consequent inhibition of the reproductive axis (332). This RFRP-3 inhibitory input on the gonadotropin release is influenced by estrogens, and may well be involved in their negative feedback. Estrogens reduce RFRP-3 expression and RFRP-3 neuronal activation (333,334).

 

SUMMARY

 

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

 

REFERENCES

 

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

 

Hyperprolactinemia

ABSTRACT

Hyperprolactinemia is the most common hypothalamic-pituitary dysfunction, being an important cause of irregular menses and infertility amongst young women. Clinical and laboratory investigation is crucial to determine hyperprolactinemia etiology and to indicate the proper treatment. Prolactinoma is the most common cause of pathological hyperprolactinemia. Physiological and pharmacological causes must be ruled out. Macroprolactinemia is a laboratorial pitfall and must be ruled out in asymptomatic hyperprolactinemic individuals. Usually, treatment is not necessary. Hook effect is another laboratory pitfall that may underestimate prolactin levels confounding the differential diagnosis between macroprolactinomas and pseudoprolactinomas. Clinical treatment with dopamine agonists is effective in 80 to 90% of hyperprolactinemic patients leading to normal serum prolactin levels and tumor reduction. Normoprolactinemia after dopamine agonist (DA) withdrawal is possible in around 30% of cases. Hypogonadism and infertility are usually reversed upon prolactin level normalization due to DA treatment, allowing pregnancy in most patients. In micro and intrasellar macroprolactinomas, DA can be withdrawal after pregnancy confirmation. Dopamine agonists are usually well tolerated. Nevertheless, valvopathy and psychiatric side effects should be actively evaluated. Surgical treatment may be indicated in resistant/intolerant patients and symptomatic apoplectic tumors. Radiotherapy is rarely performed and must be reserved to control tumors growing in an aggressive fashion. Temozolomide is an alternative treatment for resistant/aggressive prolactinomas not responding to high doses of dopamine agonists, multiple surgeries, and radiotherapy.

 

INTRODUCTION

 

Prolactin secreting pituitary tumors are the most common type of hormone secreting pituitary tumors (1). They are also the only pituitary tumors which can be effectively managed primarily by medical means (2). Therefore, their identification and diagnosis are imperative to avoid unnecessary pituitary surgery.

 

PROLACTIN SECRETION

 

Prolactin is secreted by the lactotrophs in the anterior pituitary gland. Prolactin secretion is regulated by the hypothalamus. Unlike the other anterior pituitary hormones, however, the hypothalamic influence is predominantly of tonic inhibition (3).

 

The hypothalamus secretes prolactin-release-inhibiting factors (PIF) and prolactin-releasing factors (PRF). PIF is predominantly exert by dopamine, with GABA having a minor role (4). Prolactin controls its own secretion through a short loop negative feedback, stimulating tuberoinfundibular dopamine (TIDA) cells.  Nevertheless, the nature of the physiological PRF is unclear. Thyrotropin-releasing hormone (TRH), vasoactive intestinal peptide (VIP), serotonin, histamine, oxytocin, and estrogens can act as a PRF. Other neurotransmitters and neuropeptides can also modulate prolactin secretion including endothelin, TGF beta1, angiotensin, somatostatin, substance P, neurotensin, calcitonin, EGF, natriuretic atrial peptide, bombesin, cholecystokinin, acetylcholine and vasopressin (5).

 

CAUSES OF HYPERPROLACTINEMIA

 

Disorders causing elevated prolactin levels is shown in Table 1. The causes of hyperprolactinemia may be considered, in a simplified fashion, as resulting from four basic abnormalities. In some patients, however, it is not possible to elucidate the cause of hyperprolactinemia, discussed below (6). Also, stress from venipuncture can cause slight increase in serum prolactin levels in asymptomatic individuals and a normal serum prolactin measured after resting for 30 minutes can rule out this condition (7). Nevertheless, routinely resting before venipuncture is usually not recommended (8).  In addition, prolactin should not be measured after a seizure, which could increase hormonal level (6).

 

­Table 1. Etiology of Hyperprolactinemia

Pituitary Disease

Prolactinomas

Acromegaly 

Clinically nonfunctioning pituitary adenomas 

Empty Sella syndrome            

Hypophysitis

Hypothalamic Disease        

Craniopharyngiomas

Meningiomas 

Germinomas 

Other tumors 

Sarcoidosis 

Langerhans cell histiocytosis

Neuroaxis irradiation

Vascular 

Pituitary Stalk Sec­tion

Medications

Phenothiazines

Butyrophenones

Atypical Antipsychotics

Tricyclic Antidepressants

Serotonin Reuptake Inhibitors

Reserpine

Methyldopa

Verapamil

Metoclopramide

Neurogenic

Chest wall/Breast lesions

Spinal Cord lesions

Other

Pregnancy

Breast-feeding

Hypothyroidism

Renal Insufficiency

Adrenal Insufficiency

Ectopic prolactin production

Familial hyperprolactinemia (mutated prolactin receptor)

Untreated phenylketonuria

Macroprolactinoma

Idiopathic

 

Hypothalamic Dopamine Deficiency

 

Diseases of the hypothalamus, such as tumors, arterio-venous malformations, and inflammatory processes such as sarcoidosis result in either diminished synthesis or release of dopamine. Furthermore, certain drugs (e.g., alpha-methyldopa and reserpine) are capable of depleting the central dopamine stores.

 

Defective Transport Mechanisms

 

Transection of the pituitary stalk results in impaired transport of dopamine from the hypothalamus to the lactotrophs. Pituitary or stalk tumors with abnormal blood supplies, or their pressure effects, may interfere with the circulatory pathway from the hypothalamus down the pituitary stalk to the normal lactotrophs, or a tumor, producing effective dopamine deficiency due to a functional stalk section. Besides tumors, infections, and inflammatory diseases such as hypophysitis, sarcoidosis, and tuberculosis can also cause pituitary stalk disconnection.

 

Lactotroph Insensitivity to Dopamine

 

Dopamine receptors have been found on human pituitary lactotroph adenoma cells. Receptor sensitivity to dopamine could be diminished, which would explain the lack of response to increased endogenous dopamine stimulation. However, an obvious response of the receptors to pharmacologic dopamine agonists (DA) makes this possibility less likely. Certain drugs act as dopamine-receptor-blocking agents, including phenothiazines (e.g., chlorpromazine), butyrophenones (haloperidol), and benzamides (metoclopramide, sulpiride, and domperidone). These drugs block the effects of endogenous dopamine and thus release lactotrophs from their hypothalamic inhibition. This sequence of events results in hyperprolactinemia. Table 2 summarizes the most common drugs causing hyperprolactinemia (5,9).

 

Table 2. Drugs Related to Hyperprolactinemia

Drug

Causing hyperprolactinemia

Not causing hyperprolactinemia

Antipsychotics

 

 

typical

Haloperidol Chlorpromazine,

Thioridazine, Thiothixene

 

atypical

Risperidone, quetiapine, olanzapine

Clozapine, aripiprazole, ziprasidone

Antidepressants

 

 

 

Tryclicic: clomipramine

 

 

Monoamine oxidase inhibitors: pargyline, clorgyline

 

 

Selective serotonin reuptake inhibitors: Sertraline, Fluoxetine, Paroxetine

 

Other psychotropics

Buspirone, Alprazolam

 

Other substances

Opiates and cocaine

 

Antihypertensive medications

Verapamil, methyldopa

 

Gastrointestinal medications

Metolopramide, domperidone

 

Sexual steroids

Estrogens

 

 

Stimulation of Lactotrophs

 

Hypothyroidism may be associated with hyperprolactinemia. If hypothyroidism results in increased TRH production, then TRH (which can act as a PRF) could lead to hyperprolactinemia. Estrogens act directly at the pituitary level, causing stimulation of lactotrophs, and thus enhance prolactin secretion. Furthermore, estrogens increase the mitotic activity of lactotrophs, increasing cell numbers. Pregnancy and lactation are physiological causes of hyperprolactinemia. Injury to the chest wall can also lead to hyperprolactinemia. This results from abnormal stimulation of the reflex associated with the rise in prolactin that is seen normally in lactating women during suckling (6).

 

CLINICAL MANIFESTATIONS OF HYPERPROLACTINEMIA

 

The symptoms associated with hyperprolactinemia may be due to several factors: the direct effects of excess prolactin, such as the induction of galactorrhea (10) or hypogonadism (11), the effects of the structural lesion causing the disorder (i.e. the pituitary tumor), leading to, for example, headaches, visual field defects, or external ophthalmoplegia (12); or associated dysfunction of the secretion of other anterior pituitary hormones.

 

The incidence of galactorrhea in hyperprolactinemic patients is between 30% and 80%, depending on the skill of the examiner and the degree of estrogen deficiency. Approximately 50% of women with galactorrhea, however, have normal prolactin levels. As mentioned below, it is particularly those patients with very high prolactin levels, i.e., greater than 100ng/mL (2000mU/L), who often have no galactorrhea. Thus, galactorrhea is an inconsistent marker of hyperprolactinemia (10).

 

Women with hyperprolactinemia usually present with menstrual abnormalities – amenorrhea or oligomenorrhea – or regular cycles with infertility. Occasionally, patients may present with menorrhagia. Menstrual disorders are often not seen with mild hyperprolactinemia but it is unusual not having  menstrual problems if serum prolactin is greater than 180 ng/mL (3,600 mU/L) (13).

 

In contrast, men often present late in the course of the disease with symptoms of expansion of their pituitary tumor (i.e., headaches, visual defects, and external ophthalmoplegia) or symptoms from secondary adrenal or thyroid failure. Nevertheless, these men can present with sexual impairment for many years before their diagnosis. It is unknown if macroprolactinomas are more commonly seen in men due to these delayed diagnosis and/or if prolactinoma´s pathogenesis is different in men (14). In contrast to women in whom microprolactinomas are most commonly seen, macroprolactinomas are usually found in men and the serum prolactin levels are usually much higher than those in women (15).

 

Occasionally, the syndrome may occur in prepubertal or peripubertal children, when it may present with delayed or arrested puberty or with headache and/or visual field defects or with growth arrest. Children and adolescents often present with aggressive prolactinomas, especially in boys (16). Genetic conditions such as MEN 1 and familial isolated pituitary adenomas (FIPA) should be considered (17).

 

DIFFERENTIAL DIAGNOSIS

 

It is important to exclude other causes of hyperprolactinemia: pregnancy, lactation, hypothyroidism, use of drugs that either deplete central dopamine or block dopamine receptors, and renal or hepatic failure. Ruling out these important causes, and any hypothalamic lesion, three common diagnostic possibilities remain: the presence of a microadenoma, macroadenoma, or no visible tumor at all. If patients do not harbor an identifiable tumor, they are described as having idiopathic hyperprolactinemia. It is likely, however, that patients with this condition may harbor small microprolactinomas, which were undetected with less sensitive imaging tools used in the past, and even with magnetic resonance imaging (MRI) (3).

 

A microadenoma is described as having a maximum diameter of up to 10mm (the maximal diameter of the normal pituitary gland) while a macroadenoma has a diameter larger than 10 mm. Giant adenomas are defined as the presence of the largest diameter of the tumor being larger than 4 cm (18). A microadenoma is often visualized using MRI. Usually, the serum prolactin level is below 200ng/mL (4000mU/L) in patients with microadenomas. A macroadenoma that secretes prolactin is usually associated with a serum prolactin level of more than 200ng/mL (4000mU/L). If the patient has a macroadenoma and a serum prolactin level of less than 200ng/mL (4000mU/L), consideration should be given to the possibility that a nonfunctioning pituitary adenoma (pseudo-prolactinoma) is present, the hyperprolactinemia resulting from deprivation of some lactotrophs of dopaminergic inhibition (3). However, a laboratory artifact may lead to a wrong differential diagnosis between macroprolactinomas and pseudoprolactinomas. When serum prolactin is evaluated by two-site immunometric assays, large amounts of prolactin saturate both the capture and the signal antibodies, impairing their binding, causing serum prolactin to be underestimated (the so-called “high-dose hook effect”). Therefore, patients bearing macroprolactinomas with extremely high serum prolactin levels (generally >1,000 ng/ml [>180,000 mU/L]) may present falsely low levels, e.g., 30-120 ng/ml (600-2,400 mU/L) range, causing the patient to be misdiagnosed as harboring a nonfunctioning pituitary adenoma. In order to avoid unnecessary surgery (treatment of choice for nonfunctioning tumors), prolactin assays with serum dilution are recommended in patients with macroadenomas who may harbor a prolactinomas (19). Recent assays do not present hook effect at  PRL concentrations as  high as 295,000 mIU/L (around 14,750 ng/mL) (20).

 

Another condition that interferes in the parallelism of serum PRL concentrations and prolactinoma dimensions is cystic prolactinomas, defined if 45% of lesion is the predominantly cystic sellar lesions (>50% of the volume being cystic) (21). In these lesions, serum prolactin levels are lower, but usually greater than 94 ng/mL. Differential diagnosis is important to determine the correct therapeutic intervention and includes Rathke’s cleft cysts, non-prolactin secreting cystic adenomas, craniopharyngiomas, and arachnoid cysts (22).

 

Another laboratory pitfall concerns the presence of high serum prolactin levels in subjects with few or no symptoms related to prolactin excess. Human prolactin circulates as monomeric prolactin and as larger forms, which are indistinguishable by routine assays. Monomeric prolactin is the most common form, but serum prolactin can be elevated due to the presence of aggregates with low biological activity, such as big-big prolactin, leading to so-called macroprolactinemia. The presence of molecular aggregates with low biological activity, macroprolactin, should be suspected when high serum prolactin levels are detected in patients without or with few signs and symptoms related to hyperprolactinemia. Precipitation with polyethylene glycol (PEG) is an excellent screening method. Chromatography confirms the presence of macroprolactin but is an expensive and time-consuming method; it is performed only when PEG precipitation results are inconclusive. Macroprolactinemia is a common finding, some reporting it as frequently as 8 to 42% of all cases; other centers find that it is extremely rare. Big-big prolactin biological activity is still controversial in the literature (23). Studies in vitro with rat Nb2 cell bioassays show either the presence or the absence of biological activity. A recent study using a human prolactin receptor-mediated assay compared with rat Nb2 cell assay showed that the activity displayed by macroprolactin toward the rat receptor may be inappropriate because it is not observed in the human prolactin receptor-mediated assay, consistent with the apparent absence of bioactivity in vivo (24). Most patients with macroprolactinemia do not manifest clinical features related to hyperprolactinemia, and do not need any treatment. Therefore, in order to avoid unnecessary medical or even surgical procedures, macroprolactin screening is important to consider when clinical features and serum prolactin assay results are not consonant with one another. Moreover, standardization of monomeric prolactin levels after PEG precipitation is crucial to evaluate conditions that could be associated with macroprolactinemia, as prolactinomas. Therefore, monomeric prolactin levels point to real hyperprolactinemia, even in the presence of macroprolactinemia (25).

 

Enlargement of the pituitary fossa on a skull X-ray may represent the expansion of the fossa by the macroadenoma, but care should be exercised to exclude the possibility of cisternal herniation (a partially empty fossa) as a cause for the enlargement. CT and MRI scans are useful and will also demonstrate any hypothalamic-pituitary pathologies, including solid and cystic lesions (26).   

 

CHANGES IN THE BREAST DUE TO PROLACTIN

 

A woman with amenorrhea due to hyperprolactinemia does not develop the breast atrophy seen in postmenopausal women or women with amenorrhea who are gonadotropin-deficient or have primary ovarian failure. On examination, the breast and areola are well developed and the Montgomery tubercles are hyperplastic. If the breast is correctly examined, first by expressing it from the periphery towards the areola to empty milk ducts, followed by squeezing and lifting the areola (rather than the nipple itself) to empty the milk sinuses, galactorrhea can usually be found.

 

In patients with extremely high prolactin levels and hypogonadism, galactorrhea may not be found, as minimum estrogen levels are necessary for this physical sign to occur. In male patients with hyperprolactinemia, there is usually no gynecomastia, but milk may be expressed from an entirely normal-sized male breast. The incidence of galactorrhea in men with hyperprolactinemia is low, less than 30% (i.e., it is much less common than in women). Nevertheless, the presence of galactorrhea in a man with a pituitary mass is an important clinical clue to the presence of hyperprolactinemia and possible prolactinomas (3).

 

HYPERPROLACTINEMIC HYPOGONADISM

 

The pathogenesis of the hypogonadal state in hyperprolactinemia is poorly understood. Results from an animal model study suggest that hyperprolactinemia inhibits gonadotropin-releasing hormone (GnRH) pulsatility by reducing kisspeptin input, considered the major controlling point of reproduction (27).  Moreover, kisspeptin administration restored hypothalamic-pituitary-ovarian in two hyperprolactinemic women (28).

 

In men, testosterone levels are usually low but can occasionally be normal, while in women, a hypoestrogenic state may occur, with loss of ovulation. The clinical features in hyperprolactinemic women, however, differ from those in the postmenopausal state since breast atrophy is absent and gonadotropin levels are not elevated.

 

Suppression of Gonadal Function in Hyperprolactinemia

 

In addition to GnRH inhibition, via kisspeptin,  suppression of gonadotropin secretion through inhibition of positive estrogen feedback on luteinizing hormone (LH) secretion in women (29), an increase in adrenal androgen secretion (30), and  blockade of the effects of gonadotropins at the gonadal level contribute to suppression of gonadal function in hyperprolactinemia (31,32) . Reduction in the normal LH pulsatility, essential for normal gonadal function, also occurs. Prolactin may interfere with LH and FSH action at the gonad, blocking progesterone synthesis, and may stimulate adrenal androgen secretion (29-32).

 

IMAGING OF THE PITUITARY

 

The anatomy of the pituitary is optimally assessed by contrast-enhanced MRI. MRI allows imaging of the optic chiasm, the cavernous sinuses, the pituitary (both the normal gland and tumors), and its stalk. In addition, aneurysms of the carotid are immediately obvious. Thus, MRI allows accurate measurement of the size of the pituitary and of any tumor and its relationship to the optic chiasm and cavernous sinuses. Cisternal herniation is also readily seen. If MRI is not available, CT scanning is also helpful but the resolution is less good and it is less satisfactory for delineating the relationship of the diaphragm sellae with the optic chiasm. There is little place for routine skull X-ray other than for delineating bony structures (26). For additional information see the chapter on the radiology of the pituitary.

 

TREATMENT OF HYPERPROLACTINEMIA

 

Therapeutic strategy must consider several aspects, such as the patient’s clinical presentation, the differences between microadenomas and macroadenomas concerning their natural history, the desire for pregnancy, and the patient’s treatment preference, if applicable. Medical treatment with dopamine agonist (DA) drugs is currently the gold standard approach both for microprolactinomas and macroprolactinomas. Pituitary surgery, usually by the transsphenoidal approach is generally reserved for prolactinomas resistant to DA drugs. For microadenomas, the results in the hands of most experienced surgeons are similar, with about 80% having serum prolactin normalization. However, approximately 25% develop recurrence of hyperprolactinemia by five years after surgery even with the most experienced transsphenoidal surgeons. Surgical results in macroprolactinomas are much poorer, mainly in big and/or invasive tumors. Radiotherapy for prolactinomas generally brings poor results, especially regarding normoprolactinemia restoration, and is currently reserved only for macroadenomas refractory both to medical and surgical treatment (3).

 

Dopamine Agonist Drug Therapy

 

The first DA ergot compound to be used in clinical practice was bromocriptine, a peptide ergot. It was introduced in the early 1970s in Europe and thus there is more than 40 years of experience of the use of such compounds in the treatment of hyperprolactinemia. Bromocriptine has the advantage of having a long duration of action compared to dopamine itself or oral compounds such a levo-dopa (33).

 

Bromocriptine has a similar mode of action to dopamine in stimulating dopamine receptors on the prolactin-secreting pituitary cells – D2 receptors. Stimulation of these receptors leads to inhibition of both prolactin secretion and synthesis. Figure 1 illustrates a successful case of prolactinoma treatment with bromocriptine. Subsequently a variety of other compounds have been developed which are useful additions. These include quinagolide and cabergoline (3).

Figure 1. A 34-year-old patient with amenorrhea and hyperprolactinemia (PRL 1630 ng/ml) had a sella CT imaging depicting a pituitary mass on the left (A). Bromocriptine was introduced and after 30 days on 7.5 mg a day, PRL dropped to 32 ng/mL with menses restoration. A repeat CT demonstrated a decrease in tumor size (B).

Cabergoline has an extremely long biological half-life and thus, generally only needs to be administered either once or twice per week, with a weekly dose of 0.5 to 2.0 mg. Doses higher than 2 mg per week may be used in refractory cases. In addition to its long biological half-life, cabergoline is generally better tolerated than bromocriptine, increasing the patient’s adherence. Therefore, cabergoline is currently considered the first-choice drug for the treatment of prolactinomas, except for patients wishing pregnancy in the short-term (see below). In a study comparing bromocriptine (2.5 to 5.0 mg twice daily) to cabergoline (0.5 to 1.0 mg twice weekly) in 459 hyperprolactinemic women with amenorrhea, stable normoprolactinemia was achieved in 83% of patients on cabergoline and in 59% patients on bromocriptine. Ovulatory cycles or pregnancy occurred in 72% of cases on cabergoline and in 52% of cases on bromocriptine. Drug withdrawal due to adverse effects was reported in 3% of patients on cabergoline and in 12% of patients on bromocriptine. Regarding tumor size, a decrease in at least 50% was obtained in 64% of patients on bromocriptine and in 93% of patients on cabergoline (as demonstrated in Figure 2). This important beneficial effect of DA treatment can rapidly relieve mass effects symptoms such as visual impairment, without the need of surgical decompression (34).

Figure 2. A 32 yrs-old female patient with hyperprolactinemia (PRL 80 ng/mL), irregular menses and galactorrhea had a sellar MR showing a pituitary lesion of 0.8 cm on maximal diameter (A). After cabergoline introduction (0.5 mg/week), PRL levels normalized, paralleled with menses restoration and remission of galactorrhea. Tumoral dimensions reduction are shown: tumoral maximal diameter of 0.6 cm after two years of treatment (B) and no lesion visualization after seven years on cabergoline (C).

Surgical therapy of large prolactin-secreting pituitary tumors is unsatisfactory since it is capable of normalizing serum prolactin levels or gonadal function in fewer than 20% of patients, particularly those with high prolactin levels, so the problem should be solved by another therapeutic approach.

 

SIDE EFFECTS

 

Side effects of DA therapy usually occur at the start of treatment and frequently disappear with continued therapy. If treatment is started with full doses or increased too quickly, dizziness, nausea, and postural hypotension may occur. To avoid such effects, DA must always be taken during a meal. Administration should be started at night, with a snack, when the patient retires to bed. Doses can be gradually increased afterwards.

 

Cabergoline and pergolide were associated with a higher risk of cardiac valvopathy in patients with Parkinson´s disease. These DA also have an agonist effect on serotonin receptor 5HT2B, present in fibroblast of cardiac valves and chordae tendineae. Fibroblast’s proliferation occurs after this receptor activation, leading to valve insufficiency, especially of tricuspid and pulmonary valves. This proposed mechanism was already described in carcinoid syndrome. Nevertheless, mean cabergoline dose for Parkinson patients is 3 mg a day, much higher than the usual dose for hyperprolactinemia.  Stiles et al in 2018 published a meta-analysis including 836 cabergoline-treated hyperprolactinemic patients and 1388 healthy controls from 13 published studies and there was an increase in tricuspid regurgitation of any degree (35). Although moderate and severe tricuspid regurgitation was heavily influenced by data from one center (36), data from this meta-analysis could not rule out an effect on cardiac valvular dysfunction of cabergoline used at “endocrine” dosages to treat hyperprolactinemia. British Society of Echocardiography, the British Heart Valve Society and the Society for Endocrinology recommend that a standard transthoracic echocardiogram should be performed before a patient starts DA therapy for hyperprolactinemia, repeating this exam at 5 years after starting cabergoline in patients taking a total weekly dose less than or equal to 2 mg, or annually if the dose is greater than 2 mg a week(37).

 

Quinagolide is the only non-ergot derived DA and is only available in Europe, while pergolide has now been withdrawn.

 

Recently, impulse control disorders (ICD) have been associated with DA treatment, due to the dopamine receptor type 3. There are some case reports of patients harboring prolactinomas who presented ICD during DA treatment, with different doses and length of time and an active approach to screen psychiatry disorders before and during DA treatment is recommended (38). In five series of cases, summing up 543 patients with prolactinoma, ICD frequency varied from 8 to 61%, with hypersexuality being the most common ICD and associated with male gender (39).  DA withdrawal led to an improvement of psychiatric symptoms. Patients must be evaluated by psychiatry and DA treatment should be reevaluated on an individualized basis (40).

 

CAN A DOPAMINE AGONIST DRUG BE WITHDRAWN WITHOUT RECURRENCE?    

 

One of the drawbacks of medical treatment of prolactinomas is the need for long-term therapy in the majority of the cases. As a matter of fact, treatment with bromocriptine and other DA drugs generally is considered as “symptomatic”, since DA discontinuation often leads to recurrence of hyperprolactinemia and to tumor regrowth in most patients at least after short-term use.

 

Nevertheless, remission and normoprolactinemia after DA withdrawal can occur, especially after long-term treatment. Concerning long-term therapy with bromocriptine, a  retrospective study showed that 25.8% of 62 patients with microprolactinomas and 15.9% of 69 patients with macroprolactinomas treated with bromocriptine for a median time of 47 months had persistent normoprolactinemic after a median time of 44 months after drug withdrawal (41). Another study encompassed a large cohort of hyperprolactinemic patients on cabergoline. The drug was discontinued in patients who attained normoprolactinemia, with at least 50% of tumor reduction or disappearance on image, with at least 2 years of follow-up after cabergoline withdrawal. Serum prolactin remained normal in 76%, 70% and 64% of patients with “idiopathic” hyperprolactinemia, microprolactinomas, and macroprolactinomas, respectively (42). This great discrepancy between results with bromocriptine and cabergoline was not confirmed by a meta-analysis, including 743 patients from nineteen studies: the pooled proportion of patients with remission was 21%. Stratifying those results, remission was obtained in 32% for idiopathic hyperprolactinemia, 21% for micro, and 16% for macroprolactinomas. The probability of success was higher when DA treatment lasted at least two years. A trend of cabergoline superiority over bromocriptine was observed albeit with no statistical significance (43). Hu et al performed a meta-analysis about prolactinoma remission, including 637 patients (492 micro and 123 macroprolactinomas treated with CAB, and found the pooled proportion of patients with recurrence of 65 % in a random effects model. Patients who received the lowest CAB dose and presented a significant reduction in tumor size before withdrawal were more likely to achieve the best success (44). More recently, a third meta-analysis included 1106 patients, 727 harboring microprolactinoma and 306 macroprolactinoma, treated with BRC or CAB. The overall success rate after DA withdrawal reached 36.6% (95% CI 29.4–44.2%).  Better remission rate was related to CAB treatment, especially in patients with a duration of treatment longer than two years, to low-dose CAB maintenance, and to a significant reduction in tumor size before withdrawal. (45)

 A second attempt to DA withdrawal was also described, with lower rates of success (46).

 

Although the exact mechanism of prolactinomas remission is not completely understood, it could also be linked to the natural history of the disease. Periodic withdrawal of DA is recommended, especially in cases with normal serum PRL levels and tumor reduction. Prolactinoma remission is also described after pregnancy. It is suggested that estrogen-induced necrosis could lead to tumor size and PRL decrease after delivery (3).

 

PROLACTINOMAS RESISTANT TO DOPAMINE AGONISTS

 

About 15% of patients with prolactinomas are resistant to DA therapy. The main mechanism is reduction of D2R tumor expression (47,48), although D2R polymorphism (49,50) and filamin A (51) low expression could also be implied. If a patient has been responsive and then becomes unresponsive, a pituitary carcinoma should be ruled out. The approach to the resistant prolactinoma includes pituitary surgery, radiotherapy and drugs such as temozolomide (52)and pasireotide, which has been used in only a few reported cases (53). Pituitary surgery, usually by transsphenoidal approach, aims for complete tumor removal or at least a vast debulking, which may lead to serum prolactin normalization with DA reintroduction in partially resistant cases. Surgery is more effective in microadenomas and non-invasive macroadenomas. In a meta-analysis, a surgical remission rate of 74.7% in micro and 34% in macroadenomas was shown. Nevertheless, the recurrence rate was 18% and 23% in micro and macroadenomas, respectively, leading to an even lower long-term remission rate (54,55). Radiotherapy is indicated in aggressive cases not controlled by surgery or drugs (54,56). The alkylating agent temozolomide has efficacy in aggressive pituitary adenomas and carcinomas, mainly the prolactin secreting ones. Complete and partial response was obtained in 56% of 32 cases reviewed in the literature (52). Figure 3 illustrates sellar MR of a young man with an invasive/aggressive macroprolactinoma, resistant do dopamine agonist, multiple surgeries and radiotherapy.

Figure 3. 26 yrs-old man complaining of visual disturbance was submitted to a sellar MRI (A): sellar mass with 5 cm in the maximal diameter with supra, infra and left parasellar invasion. Serum PRL levels were above 1000 ng/mL. After one year on cabergoline, 0.5 mg/day, there was no tumor reduction and PRL levels were around 800 ng/mL. He was then submitted to transsphenoidal surgery and radiotherapy in another medical service. Sellar MRI (B) two months after the surgery showed a pituitary mass with 3.1 cm in the maximal diameter. Despite chiasmal decompression, visual dysfunction was not reversed and, during his follow-up, anterior pituitary function was lost. PRL levels on cabergoline, 1.5 mg/week, were 250 ng/ml. After two more years, sellar MRI depicted a lesion of 2.9 cm. in the maximal diameter (C). There was a progressive rise of PRL levels despite increased cabergoline doses (0.5 mg/d) and another sellar MRI (D), after two years, depicted a lesion of 3.1 cm in the maximal diameter. Another surgery was indicated.

Fertility in Women

 

The efficacy of hyperprolactinemia/prolactinoma´s treatment allow fertility and pregnancy in many women. There are, however, several important considerations that must be recognized by both the physician and patient. It includes tumor growth risk and possible teratogenic sequelae of fetal exposure to bromocriptine and other drugs (57).

 

There is little doubt that patients with pituitary tumors run a small, but significant, risk of expansion of the tumor during pregnancy. It is very difficult, however, to assess the absolute risk. With microadenomas, which did not undergo previous surgery or radiotherapy, the incidence seems to be 2.5%. In patients with macroadenomas not operated on or irradiated, the incidence is higher, 18.1% (58,59). However, the risk of complications may be lower in women previously operated or irradiated. This risk is unrelated to bromocriptine therapy prior to pregnancy but may occur when fertility is induced with other drugs, including exogenous gonadotropins and clomiphene, and even when no drug therapy has been employed in patients with pre-existing pituitary adenomas.

 

In practice, the problem of pregnancy is not great, since the vast majority of women who present with hyperprolactinemia only have microadenomas. To avoid major problems, it is extremely important that patients undergo careful endocrine, neuroradiologic, and neuro-ophthalmologic evaluation prior to treatment. If there is no suprasellar extension, and if the patient harbors only a microadenoma, then the risk of clinically significant swelling of the pituitary is extremely small; it is therefore suggested that the patient be evaluated clinically in every trimester throughout pregnancy, with no routine serum prolactin assessment. If the patient has a macroadenoma and suprasellar extension, transsphenoidal decompression can be considered, mainly for resistant cases. However, in those patients with a good response to DA in terms of prolactin normalization and tumor shrinkage within sellar boundaries, at least one year before pregnancy, the drug can be withdrawn and reintroduced if tumor re-growth is observed. If such an approach fails, pituitary surgery or premature delivery, if feasible, would be indicated. Additionally, in the case of tumor apoplexy, high-dose dexamethasone may improve clinical symptoms and also may reduce the chances of fetal respiratory distress if premature delivery is needed (2,59).

 

In recent years, pregnancies have been described in patients using other DA such as quinagolide and cabergoline. Although there is no evidence of increased frequency of abortions or malformations in cabergoline-induced pregnancies, the drug’s long action, which persists up to three weeks after its withdrawal, associated with fewer (albeit increasing) data when compared to bromocriptine (around 1000 versus over 6,000 pregnancies), limit the confidence that it can be used for patients who wish to conceive or its use during pregnancy. In the United States, bromocriptine is the only FDA approved drug for inducing pregnancy in hyperprolactinemic women. Cabergoline’s label does not recommend use during pregnancy. Nevertheless, experience of pregnancy induced by cabergoline is accumulating. Incidence of premature delivery, multiple births, and malformations is not different from women who used bromocriptine or from the general population (60). However, the issue of spontaneous abortion is still under debate (60,61). Quinagolide use was related to abortions and malformations (58).

 

Fertility in Men

 

Hyperprolactinemic men exhibit reduced quality of seminal fluid, including the total sperm count, the sperm kinetic index, and sperm nuclear DNA integrity. Cabergoline significantly increased those indexes, mainly after 12 months of treatment (62).

 

Interestingly enough, it was shown that clomiphene citrate administration can increase testosterone levels in men, even when serum prolactin levels were elevated. Fertility restoration is an additional advantage of this approach, compared to testosterone replacement (63).

 

Conclusion

 

Dopamine-agonist therapy for hyperprolactinemia leads to a reversal of the hyperprolactinemic hypogonadal state without risk of the development of pituitary insufficiency, thus allowing pregnancy in former infertile patients. Dopamine-agonist therapy is effective not only in patients with microadenomas but also in the majority of patients with large prolactin-secreting tumors in reducing tumor size. Moreover, emerging evidences point to the possibility of drug withdrawal after long-term treatment. Surgery, radiotherapy and antiblastic drugs, as temozolomide, should be reserved for resistant/aggressive cases.

 

REFERENCES

 

  1. Daly AF, Beckers A. The Epidemiology of Pituitary Adenomas. Endocrinol Metab Clin North Am 2020; 49:347-355
  2. Melmed S, Casanueva FF, Hoffman AR, Kleinberg DL, Montori VM, Schlechte JA, Wass JA, Society E. Diagnosis and treatment of hyperprolactinemia: an Endocrine Society clinical practice guideline. J Clin Endocrinol Metab 2011; 96:273-288
  3. BRONSTEIN, M.D. Disorders of Prolactin Secretion and Prolactinomas. In: De, LJ G, JL J, eds. Endocrinology. Vol 1. Philadelphia: Elsevier Saunders; 2010:485-510.
  4. Grattan DR. 60 YEARS OF NEUROENDOCRINOLOGY: The hypothalamo-prolactin axis. J Endocrinol 2015; 226:T101-122
  5. Torre DL, Falorni A. Pharmacological causes of hyperprolactinemia. Ther Clin Risk Manag 2007; 3:929-951
  6. ME M. Disorders of prolactin secretion. Endocrinol Metab Clin North Am 2001; 30:585-610
  7. Jacobs S, Brown SA, Mason J, Wahby V, Kasl S, Ostfeld A. Psychological distress, depression and prolactin response in stressed persons. J Human Stress 1986; 12:113-118
  8. Ferriani RA, Silva de Sá MF. Effect of venipuncture stress on plasma prolactin levels. Int J Gynaecol Obstet 1985; 23:459-462
  9. Molitch ME. Drugs and prolactin. Pituitary 2008; 11:209-218
  10. Kleinberg DL, Noel GL, Frantz AG. Galactorrhea: a study of 235 cases, including 48 with pituitary tumors. N Engl J Med 1977; 296:589-600
  11. Franks S, Murray MA, Jequier AM, Steele SJ, Nabarro JD, Jacobs HS. Incidence and significance of hyperprolactinaemia in women with amenorrhea. Clin Endocrinol (Oxf) 1975; 4:597-607
  12. Abe T, Matsumoto K, Kuwazawa J, Toyoda I, Sasaki K. Headache associated with pituitary adenomas. Headache 1998; 38:782-786
  13. Mah PM, Webster J. Hyperprolactinemia: etiology, diagnosis, and management. Semin Reprod Med 2002; 20:365-374
  14. Duskin-Bitan H, Shimon I. Prolactinomas in males: any differences? Pituitary 2020; 23:52-57
  15. Ciccarelli A, Guerra E, De Rosa M, Milone F, Zarrilli S, Lombardi G, Colao A. PRL secreting adenomas in male patients. Pituitary 2005; 8:39-42
  16. Arya VB, Aylwin SJB, Hulse T, Ajzensztejn M, Kalitsi J, Kalogirou N, Bodi I, Thomas N, Hampton T, Kapoor RR, Buchanan CR. Prolactinoma in childhood and adolescence-Tumour size at presentation predicts management strategy: Single centre series and a systematic review and meta-analysis. Clin Endocrinol (Oxf) 2021; 94:413-423
  17. Barry S, Korbonits M. Update on the Genetics of Pituitary Tumors. Endocrinol Metab Clin North Am 2020; 49:433-452
  18. Maiter D, Delgrange E. Therapy of endocrine disease: the challenges in managing giant prolactinomas. Eur J Endocrinol 2014; 170:R213-227
  19. Frieze TW, Mong DP, Koops MK. "Hook effect" in prolactinomas: case report and review of literature. Endocr Pract 2002; 8:296-303
  20. Fahie-Wilson M, Bieglmayer C, Kratzsch J, Nusbaumer C, Roth HJ, Zaninotto M, Plebani M, Hubbuch A, Schneider E. Roche Elecsys Prolactin II assay: reactivity with macroprolactin compared with eight commercial assays for prolactin and determination of monomeric prolactin by precipitation with polyethylene glycol. Clin Lab 2007; 53:485-492
  21. Faje A, Chunharojrith P, Nency J, Biller BM, Swearingen B, Klibanski A. Dopamine Agonists Can Reduce Cystic Prolactinomas. J Clin Endocrinol Metab 2016; 101:3709-3715
  22. Nakhleh A, Shehadeh N, Hochberg I, Zloczower M, Zolotov S, Taher R, Daoud Naccache D. Management of cystic prolactinomas: a review. Pituitary 2018; 21:425-430
  23. Hattori N. Macroprolactinemia: a new cause of hyperprolactinemia. J Pharmacol Sci 2003; 92:171-177
  24. Glezer A, Soares CR, Vieira JG, Giannella-Neto D, Ribela MT, Goffin V, Bronstein MD. Human macroprolactin displays low biological activity via its homologous receptor in a new sensitive bioassay. J Clin Endocrinol Metab 2006; 91:1048-1055
  25. Bronstein MD. Editorial: is macroprolactinemia just a diagnostic pitfall? Endocrine 2012; 41:169-170
  26. Rennert J, Doerfler A. Imaging of sellar and parasellar lesions. Clin Neurol Neurosurg 2007; 109:111-124
  27. Sonigo C, Bouilly J, Carré N, Tolle V, Caraty A, Tello J, Simony-Conesa FJ, Millar R, Young J, Binart N. Hyperprolactinemia-induced ovarian acyclicity is reversed by kisspeptin administration. J Clin Invest 2012; 122:3791-3795
  28. Millar RP, Sonigo C, Anderson RA, George J, Maione L, Brailly-Tabard S, Chanson P, Binart N, Young J. Hypothalamic-Pituitary-Ovarian Axis Reactivation by Kisspeptin-10 in Hyperprolactinemic Women With Chronic Amenorrhea. J Endocr Soc 2017; 1:1362-1371
  29. Glass, MR SR, Butt WR, Edwards RL and London DR. An abnormality of osterogen feedback in amenorrhoea-galactorrhoea. BMJ 1975; 3:274-275
  30. Feitus, FA GBaMM. Stat5-mediated regulation of the human type II 3beta-hydrosteroid dehydrogenase/delat-5-delta4isomerase gene activation by prolactin. Molecular Endocrinology 1999; 13:1084-1093
  31. Demura R, Ono M, Demura H, Shizume K, Oouchi H. Prolactin directly inhibits basal as well as gonadotropin-stimulated secretion of progesterone and 17 beta-estradiol in the human ovary. J Clin Endocrinol Metab 1982; 54:1246-1250
  32. Dorrington JH, Gore-Langton RE. Antigonadal action of prolactin: further studies on the mechanism of inhibition of follicle-stimulating hormone-induced aromatase activity in rat granulosa cell cultures. Endocrinology 1982; 110:1701-1707
  33. Thorner MO, McNeilly AS, Hagan C, Besser GM. Long-term treatment of galactorrhoea and hypogonadism with bromocriptine. Br Med J 1974; 2:419-422
  34. Webster J, Piscitelli G, Polli A, Ferrari CI, Ismail I, Scanlon MF. A comparison of cabergoline and bromocriptine in the treatment of hyperprolactinemic amenorrhea. Cabergoline Comparative Study Group. N Engl J Med 1994; 331:904-909
  35. Stiles CE, Tetteh-Wayoe ET, Bestwick J, Steeds RP, Drake WM. A meta-analysis of the prevalence of cardiac valvulopathy in hyperprolactinemic patients treated with Cabergoline. J Clin Endocrinol Metab 2018;
  36. Colao A, Galderisi M, Di Sarno A, Pardo M, Gaccione M, D'Andrea M, Guerra E, Pivonello R, Lerro G, Lombardi G. Increased prevalence of tricuspid regurgitation in patients with prolactinomas chronically treated with cabergoline. J Clin Endocrinol Metab 2008; 93:3777-3784
  37. Steeds R, Stiles C, Sharma V, Chambers J, Lloyd G, Drake W. Echocardiography and monitoring patients receiving dopamine agonist therapy for hyperprolactinaemia: A joint position statement of the British Society of Echocardiography, the British Heart Valve Society and the Society for Endocrinology. Clin Endocrinol (Oxf) 2019; 90:662-669
  38. Noronha S, Stokes V, Karavitaki N, Grossman A. Treating prolactinomas with dopamine agonists: always worth the gamble? Endocrine 2016; 51:205-210
  39. De Sousa SMC, Baranoff J, Rushworth RL, Butler J, Sorbello J, Vorster J, Thompson T, McCormack AI, Inder WJ, Torpy DJ. Impulse Control Disorders in Dopamine Agonist-Treated Hyperprolactinemia: Prevalence and Risk Factors. J Clin Endocrinol Metab 2020; 105
  40. Ioachimescu AG, Fleseriu M, Hoffman AR, Vaughan Iii TB, Katznelson L. Psychological effects of dopamine agonist treatment in patients with hyperprolactinemia and prolactin-secreting adenomas. Eur J Endocrinol 2019; 180:31-40
  41. Passos VQ, Souza JJ, Musolino NR, Bronstein MD. Long-term follow-up of prolactinomas: normoprolactinemia after bromocriptine withdrawal. J Clin Endocrinol Metab 2002; 87:3578-3582
  42. Colao A, Di Sarno A, Cappabianca P, Di Somma C, Pivonello R, Lombardi G. Withdrawal of long-term cabergoline therapy for tumoral and nontumoral hyperprolactinemia. N Engl J Med 2003; 349:2023-2033
  43. Dekkers OM, Lagro J, Burman P, Jørgensen JO, Romijn JA, Pereira AM. Recurrence of hyperprolactinemia after withdrawal of dopamine agonists: systematic review and meta-analysis. J Clin Endocrinol Metab 2010; 95:43-51
  44. Hu J, Zheng X, Zhang W, Yang H. Current drug withdrawal strategy in prolactinoma patients treated with cabergoline: a systematic review and meta-analysis. Pituitary 2015; 18:745-751
  45. Xia MY, Lou XH, Lin SJ, Wu ZB. Optimal timing of dopamine agonist withdrawal in patients with hyperprolactinemia: a systematic review and meta-analysis. Endocrine 2018; 59:50-61
  46. Vilar L, Albuquerque JL, Gadelha PS, Rangel Filho F, Siqueira AM, da Fonseca MM, Viana KF, Gomes BS, Lyra R. Second Attempt of Cabergoline Withdrawal in Patients with Prolactinomas after a Failed First Attempt: Is it Worthwhile? Front Endocrinol (Lausanne) 2015; 6:11
  47. Passos VQ, Fortes MA, Giannella-Neto D, Bronstein MD. Genes differentially expressed in prolactinomas responsive and resistant to dopamine agonists. Neuroendocrinology 2009; 89:163-170
  48. Molitch ME. Pharmacologic resistance in prolactinoma patients. Pituitary 2005; 8:43-52
  49. Filopanti M, Lania AG, Spada A. Pharmacogenetics of D2 dopamine receptor gene in prolactin-secreting pituitary adenomas. Expert Opin Drug Metab Toxicol 2010; 6:43-53
  50. Bueno C, Trarbach EB, Bronstein MD, Glezer A. Cabergoline and prolactinomas: lack of association between DRD2 polymorphisms and response to treatment. Pituitary 2017; 20:295-300
  51. Peverelli E, Mantovani G, Vitali E, Elli FM, Olgiati L, Ferrero S, Laws ER, Della Mina P, Villa A, Beck-Peccoz P, Spada A, Lania AG. Filamin-A is essential for dopamine d2 receptor expression and signaling in tumorous lactotrophs. J Clin Endocrinol Metab 2012; 97:967-977
  52. Chen C, Yin S, Zhang S, Wang M, Hu Y, Zhou P, Jiang S. Treatment of aggressive prolactinoma with temozolomide: A case report and review of literature up to date. Medicine (Baltimore) 2017; 96:e8733
  53. Sari R, Altinoz MA, Ozlu EBK, Sav A, Danyeli AE, Baskan O, Er O, Elmaci I. Treatment Strategies for Dopamine Agonist-Resistant and Aggressive Prolactinomas: A Comprehensive Analysis of the Literature. Horm Metab Res 2021; 53:413-424
  54. Gillam MP, Molitch ME, Lombardi G, Colao A. Advances in the treatment of prolactinomas. Endocr Rev 2006; 27:485-534
  55. Primeau V, Raftopoulos C, Maiter D. Outcomes of transsphenoidal surgery in prolactinomas: improvement of hormonal control in dopamine agonist-resistant patients. Eur J Endocrinol 2012; 166:779-786
  56. Ježková J, Hána V, Kosák M, Kršek M, Liščák R, Vymazal J, Pecen L, Marek J. Role of gamma knife radiosurgery in the treatment of prolactinomas. Pituitary 2019; 22:411-421
  57. Glezer A, Bronstein MD. Prolactinomas: how to handle prior to and during pregnancy? Minerva Endocrinol 2017;
  58. Huang W, Molitch ME. Pituitary Tumors in Pregnancy. Endocrinol Metab Clin North Am 2019; 48:569-581
  59. Glezer A, Bronstein MD. Prolactinomas in pregnancy: considerations before conception and during pregnancy. Pituitary 2019;
  60. Sant' Anna BG, Musolino NRC, Gadelha MR, Marques C, Castro M, Elias PCL, Vilar L, Lyra R, Martins MRA, Quidute ARP, Abucham J, Nazato D, Garmes HM, Fontana MLC, Boguszewski CL, Bueno CB, Czepielewski MA, Portes ES, Nunes-Nogueira VS, Ribeiro-Oliveira A, Francisco RPV, Bronstein MD, Glezer A. A Brazilian multicentre study evaluating pregnancies induced by cabergoline in patients harboring prolactinomas. Pituitary 2020; 23:120-128
  61. Hurault-Delarue C, Montastruc JL, Beau AB, Lacroix I, Damase-Michel C. Pregnancy outcome in women exposed to dopamine agonists during pregnancy: a pharmacoepidemiology study in EFEMERIS database. Arch Gynecol Obstet 2014; 290:263-270
  62. De Rosa M, Zarrilli S, Di Sarno A, Milano N, Gaccione M, Boggia B, Lombardi G, Colao A. Hyperprolactinemia in men: clinical and biochemical features and response to treatment. Endocrine 2003; 20:75-82
  63. Ribeiro RS, Abucham J. Recovery of persistent hypogonadism by clomiphene in males with prolactinomas under dopamine agonist treatment. Eur J Endocrinol 2009; 161:163-169