Archives

The Management of Type 1 Diabetes

ABSTRACT

 

Type 1 diabetes (T1D) is an autoimmune disease characterized by progressive pancreatic beta-cell loss resulting in insulin deficiency and hyperglycemia. Exogenous insulin therapy is essential to prevent fatal complications from hyperglycemia. The Diabetes Control and Complications Trial and its long-term follow up, the Epidemiology of Diabetes and its Complications study, demonstrated that stringent glycemic control with intensive insulin therapy can prevent or postpone progression of microvascular disease and reduce risk for macrovascular disease and all-cause mortality. In addition, data obtained from the T1D Exchange, a registry of T1D patients founded in 2010, has become an invaluable resource for scientists worldwide, facilitating collaboration and accelerating understanding of prevailing diabetes practices. Insulin therapy using rapid- and long-acting insulin analogs is the mainstay of management of T1D. Insulin delivery is achieved subcutaneously using multiple daily injections or subcutaneous insulin infusion using insulin pumps. Effective management also involves use of self-monitoring of blood glucose using improved blood glucose meters, continuous glucose monitoring (CGM) devices, and newer insulin pumps with integrated sensor-augmented systems. Addressing psychosocial aspects of T1D plays a crucial role in effective disease management. Strategies to manage T1D are rapidly evolving. In addition to newer insulins, adjunctive non-insulin therapies such as use of incretin agents and SGLT-2 and combination SGLT-1/2 inhibitors are being actively pursued. CGM technology combined with glucose prediction algorithms has allowed for the development of artificial pancreas delivery systems. Cellular replacement options include pancreas and islet cell transplantation which can restore euglycemia but are limited by donor availability and the need for chronic immunosuppression. Newer strategies under development include islet cell encapsulation techniques, which might obviate the need for immunosuppression. Smart-insulin delivery systems, capable of releasing insulin depending on ambient glucose, are also being evaluated.

HISTORY OF TYPE 1 DIABETES TREATMENTS

 

Insulin Therapy

 

The discovery of insulin in 1921-22 was one of the greatest medical breakthroughs in history (1) (Figure 1). Initial work at the University of Toronto allowed for pancreatic extracts to be used to decrease blood glucoses in diabetic dogs. Developments by the pharmaceutical industry allowed for the large-scale commercial insulin production in 1923 (2). Individuals, mostly children with type 1 diabetes (T1D), whose life expectancies were measured in months were now able to prevent fatal ketoacidosis by taking injections of crude “soluble” (later known as regular) insulin. However, problems were soon noted. Hypoglycemia, occasionally life-threatening, was encountered as diabetes monitoring by urine testing for glycosuria was crude at best during those first decades after the discovery of insulin. The insulin itself was often impure and varied in potency from lot to lot. Allergic reactions were common and occasionally anaphylaxis would occur. Even more concerning was the appreciation that these patients often succumbed to chronic vascular complications which either dramatically reduced quality of life or resulted in a fatal cardiovascular event.

 

Tools to manage individuals with T1D improved over the decades since the discovery of insulin. Initial insulins were manufactured from bovine or porcine pancreata and production techniques became more efficient. Insulins with longer duration of action were first introduced in the 1930s, and over time purity and consistency of potency of these insulins improved (3). Nevertheless, “standard” animal insulins prior to 1980 contained 300-10000 parts per million of impurities, and elicited local and systemic effects when injected. Present day insulins sold in the United States today all contain less than 1 part per million of impurities.

 

Major improvements in insulin were developed in the late 1970s and early 1980s. First, not only was “purified” insulin introduced, but in 1982 the first human insulin was marketed both by Eli Lilly (recombinant DNA technology) and Novo (semi-synthetic methodology).  These insulins were available as short-acting (regular) and longer-acting (Neutral protamine Hagedorn (NPH), lente, and ultralente) preparations. The other major advance with insulin therapy was with the delivery by the first continuous subcutaneous insulin infusion (CSII) pumps. While pumps were initially touted as providing less variable insulin absorption, the use of CSII had a greater impact: both patients and clinicians used this tool to teach themselves how to best use “basal bolus” insulin therapy, a strategy that would become a standard of care after the beginning of the next century with the development of insulin analogs.

Figure 1. Time line of the evolution of insulin therapy. Figure source ref 3.

Monitoring Tools

 

At the same time as the development of human insulin and insulin pumps, improvements in glucose monitoring were introduced. Although there was initial skepticism if home blood glucose monitoring would be accepted by patients with diabetes, history has confirmed that this technology has revolutionized diabetes management and has allowed patients to titrate blood glucose to normal or near-normal levels. While self-monitoring of blood glucose (SMBG) allowed immediate evaluation of diabetes management, the introduction of hemoglobin A1c (HbA1c, or glycated hemoglobin, A1C) around the same time was used as a marker of objective longer-term (about 90 days) glucose control. When hemoglobin is exposed to glucose in the bloodstream, the glucose slowly becomes nonenzymatically bound to the hemoglobin in a concentration-dependent manner. The percentage of hemoglobin molecules that are glycated (have glucose bound to it) indicates what the average blood glucose concentration has been over the life of the red blood cell. Perhaps as importantly, A1C made it possible for researchers to study the effects of long-term glucose control and the development of vascular complications. New students of diabetes may now find it difficult to appreciate that one of the greatest medical controversies between the discovery of insulin and the early 1990s was the relationship between glucose control and diabetes complications. Improved insulins, pumps, SMBG, and A1C finally allowed this question to be properly studied.

 

THE DIABETES CONTROL AND COMPLICATIONS TRIAL

 

In 1993, all controversy regarding the impact of glucose control and vascular complications was dramatically answered with the publication of the Diabetes Control and Complications Trial (DCCT) (4). The trial showed definitively that stringent blood glucose control (for an average of 6.5 years) could slow or postpone the progression of retinal, renal, and neurological complications in individuals with T1D (Figure 2). In patients treated with “intensive therapy”—that is, therapy aimed at maintaining blood glucose levels as close to normal as possible—the risk of developing diabetic retinopathy was reduced by 76%, diabetic neuropathy by 60%, and diabetic nephropathy by 54%, compared with conventionally treated patients. Other benefits of intensive diabetes management include improved lipid profiles, reduced risk factors for macrovascular disease, and better maternal and fetal health.

 

Since the DCCT was completed in 1993, the research subjects have been followed in an observational study calledEpidemiology of Diabetes and its Complications (EDIC) (5).  It was soon observed that the impact of this improved diabetes therapy for an average of 6.5 years (maintaining a A1C of approximately 7% with multiple injections or CSII compared to once or twice daily insulin and a A1C of approximately 9%) had long-lasting effects. Termed “metabolic memory”, there continued to be improvements in microvascular complications four years after the DCCT ended (Figure 3) (6-8).  Despite the fact that A1C levels remained about 8% for both groups after the DCCT, the risk reduction for nonfatal myocardial infarction, stroke, or death were reduced by 57% eleven years after the conclusion of the formal study. The conclusions of this are profound since this was the first study to report a reduction of macrovascular disease with glucose control. Furthermore, these data confirmed the need to control blood glucose as meticulously as possible early in the course of the disease (9).

Figure 2. Relationship between microvascular complications and A1C in T1D

Figure 3. Cumulative incidence of further 3-step progression of retinopathy from DCCT closeout to EDIC study year 10 (adjusted for retinopathy level at DCCT end, cohort, entry A1C, baseline diabetes duration). From reference (10).

TYPE 1 DIABETES EXCHANGE 

 

Compared with treatment methods used in the DCCT over 20 years ago, many new tools and technologies have now become available that enable patients and clinicians to attain target A1C levels more safely. Rapid- and long-acting insulin analogs, improved blood glucose meters, newer insulin pumps with integrated sensor-augmented systems and with automatic threshold suspend capabilities and continuous glucose monitoring (CGM) devices now play an integral part of T1D management. To evaluate how these advances in diabetes technology have impacted glycemic control in T1D, a broad-based, large-scale, multisite registry that includes patients at all ages across the life span in the U.S. was established in 2010 through a grant from the Leona M. and Harry B. Helmsley Charitable Trust. Called the T1D Exchange, this registry aims to provide an expansive data set to address important clinical and public health issues related to T1D. It comprises three complementary sections: i) a clinic network of adult and pediatric diabetes clinics; ii) a Web site called Glu, serving as an online community for patients; and iii) a biobank to store biological human samples for use by researchers. A statistical resource center provides statistical support to the Exchange as well as other T1D researchers. The data have provided information about various aspects of T1D, including metabolic control and management, in the United States and the opportunity to compare this data with registries from Europe and Australia (11). The clinic registry has provided valuable information regarding the state of T1D management and outcomes and allowed for addressing important clinical and public health issues. Registry data also have helped identify knowledge gaps leading to further advancements in clinical trials and epidemiologic research with over 47 publications as of March 2019 (12).

 

Currently there are over 35,000 patients enrolled in the registry, ranging in age from 1 - 93 years, with a duration of diabetes ranging from 1.5 to 83 years, 50% female, 82% were non-Hispanic white (13). Most recent data from the registry revealed that mean A1C in adults over age 30 ranged from 7.5-7.8%, which is lower than the value of 8% observed in the DCCT (14). However mean A1C levels increased in teens and emerging adults from 8.5% to 9.3%. Insulin pump use was observed in 63% of individuals. CGM use increased exponentially from 2010-12 to 2016-18 from 7% to 30%, with most participants using the Dexcom system (77%).  CGM use increased significantly in the pediatric population. Many patients in the registry were able to achieve target A1C levels without an increase in the frequency of serious hypoglycemia as was observed in the DCCT. Use of adjunctive non-insulin glucose-lowering therapies was low overall and primarily included metformin, in 6% of adult participants over age 26 years.

 

CURRENT TECHNOLOGY IN TYPE 1 DIABETES

 

Glucose Meters

 

Current blood glucose monitoring systems (BGMS) are small electronic devices capable of analyzing glucose levels in capillary whole blood. To test blood glucose levels, patients are required to prick a finger using a lancing device to obtain a small drop of blood. The patient then places the drop of blood onto a glucose test strip, which has been previously inserted into the glucose meter. Typically, just a few seconds are required for the device to provide a blood glucose value.

 

BGMS use enzymatic reactions to provide estimates of blood glucose levels and the enzymes utilized include glucose oxidase, glucose dehydrogenase and hexokinase. The specific enzyme is usually packaged in a dehydrated form in a glucose test strip. Once blood is applied to the test strip, glucose in the patient’s blood sample rehydrates the enzyme activating a reaction. The product of this reaction can then be detected and measured by the glucose meter (15).

 

Notably, the advent of point-of-care BGMS has revolutionized diabetes care by allowing patients and practitioners to obtain real-time estimates of blood glucose values. These portable devices enabled patients to perform self-monitoring of blood glucose (SMBG), an integral component of effective diabetes self-management. The benefits of SMBG were confirmed during the DCCT which showed that intensive insulin therapy, requiring SMBG≥4 times/day with concomitant insulin dose titration, delayed the onset and slowed the progression of microvascular complications (4). Later, it was shown in the T1D Exchange that a higher frequency of testing (up to 10 times daily) is inversely associated with A1C levels in all age groups (16).

 

SMBG allows patients to guide management decisions (e.g., adjusting food intake, insulin therapy, and exercise) and determine whether glucose targets are being achieved. Further, it can help patients in monitoring and preventing asymptomatic hypoglycemia (17).

 

Patients with T1D should perform SMBG at a minimum of 4 times a day (before meals and at bedtime), as this will allow adjustments to prandial and basal insulin doses. In addition, SMBG should be considered prior to snacks, before and at completion of exercise, in the event of symptoms suggestive of hypoglycemia, and after treating hypoglycemia until blood glucose levels have normalized. Lastly, patients should test their blood glucose before performing critical tasks such as driving a motor vehicle or operating heavy machinery. Ultimately, frequency of SMBG will largely depend on patients’ individual needs (17).

 

An important point to make, however, is that patients should also be educated on avoiding “overuse” of SMBG. Testing too frequently may lead to administration of multiple correction doses within short periods of time, particularly if patients are anxious about their glucose levels not returning to target “fast enough”, leading to insulin “stacking” and resulting in iatrogenic hypoglycemia.

 

The technology of BGMS has evolved over the years and current devices are relatively easy to use and require minimal amounts of blood (Figure 4). Some instruments are able to capture events affecting glucose control (e.g., exercise, meals, insulin administration), provide customized reports, and calculate insulin bolus needs according to glycemia and intake of carbohydrate based on pre-established settings (i.e., insulin sensitivity factor and insulin-to-carbohydrate ratios). However, despite these unique advances in self-monitoring of blood glucose, independent analytic testing has shown that various BGMS do not fulfill the accuracy requirements set by the International Organization for Standardization (ISO) 151917 which requires for ≥95% of results to fall within ± 15 mg/dL of the reference result for samples with glucose concentrations <100 mg/dL and ±15% for samples with glucose concentrations ≥100 mg/dL (18). In addition, the FDA has stated that the ISO 15197 criteria are not sufficient to adequately protect lay-users of SMBGs because, for example, the standard does not adequately address the performance of over-the-counter blood glucose testing systems in the hypoglycemic range or across test strip lots. In view of this, the FDA has developed the “Self-Monitoring Blood Glucose Test Systems for Over-the-Counter use” guidance document which is intended to guide manufacturers in conducting appropriate performance studies and preparing 510(k) submissions for these device types (https://www.fda.gov/regulatory-information/search-fda-guidance-documents/self-monitoring-blood-glucose-test-systems-over-counter-use). Thus, there is a pressing need for high quality standards to ensure improved accuracy and precision from BGMS.

 

SMBG has important drawbacks since blood is only sampled intermittently and therefore only glimpses of blood glucose concentrations are provided. SMBG does not offer information on glucose fluctuations even if performed frequently. Thus, there is potential for missing episodes of hyperglycemia and hypoglycemia.

Figure 4. Examples of a few blood glucose monitoring systems.

Glucose Downloads

 

The vast majority of currently available BGMS allow the generation of downloadable reports. These reports are a unique component of the patients’ evaluation allowing the identification of areas that require special attention in diabetes management. However, technical difficulties often compromise the usefulness of these data. For instance, it is not unusual for the date and/or time of the glucose meters to be inaccurate. Simple errors such as these have a huge impact on patient management as the data downloaded becomes largely uninterpretable. In addition, as each glucose meter usually has its own proprietary software, if a clinic does not have the specific software installed on their local computers, then the data may not be downloaded. The clinician is left with trying to review the data directly from the device, which is time consuming and does not offer the detailed overview from a customized printable report. There are platforms that are currently available which allow downloading various glucose meters, insulin pumps and CGM data and provide standardized reports (e.g., Clinipro®, Diasend®, Carelink®, Glooko®). However, there needs to be a unified effort by BGMS, insulin pump, and CGM companies in order to generate a universal download protocol as this would simplify data analysis and interpretation by practitioners (19).

 

Continuous Glucose Monitoring

 

Perhaps the most innovative technology for the treatment of T1D is the introduction of CGM (Figure 5). CGM technology allows for the measurement of glucose concentrations in the interstitial fluid (ISF) which correlates with plasma glucose values. However, when interpreting CGM values it is important to understand that ISF glucose consistently lags plasma glucose. A study in healthy adults analyzing glucose tracers following an overnight fast showed that it takes 5-6 minutes for glucose to be transported from the vascular to the interstitial space (physiological delay) (20). This is particularly relevant when glucose levels are trending up or down quickly as CGM data will not be as reliable in such scenarios and thus patients should confirm the direction of their glucose concentration by SMBG.

 

The components of CGM consist of a sensor that is inserted subcutaneously, a small electronic device that serves as the platform for the sensor, a transmitter, and a receiver device, which can be a standalone device or a smartphone (Figure 5). CGM Sensors can measure glucose levels up to every minute allowing for a glucose tracing to be generated and displayed in real-time (RT-CGM) on a receiver device, greatly improving the understanding of patients’ glucose profiles. Further, with the exception of the GuardianTM Connect system (Medtronic) which is pending approval, currently available CGM devices have obtained FDA approval for non-adjunctive use which means that patients can rely on their CGM values in order to guide management decisions (21).

 

Patients can customize alarms to activate for hypoglycemia or hyperglycemia. Understanding the trend allows patients to decide whether an increase or decrease in mealtime insulin dose is necessary. CGM thus also allows patients to intercept hypoglycemia (or hyperglycemia) prior to it occurring. Patients can also “flag” events thereby improving interpretation of glucose control associated with meals, insulin administration, and exercise. Also, most CGM devices allow users to share their RT-CGM data with others (e.g., family members or friends) which can then be monitored on a smartphone or other internet-enabled devices. This is of particular interest in the pediatric population as it allows parents to remotely monitor their child’s glucose profile when away from home or while exercising (e.g., participating in sports). Features of currently available CGM devices are listed in Figure 6.

 

Based on how the CGM data is delivered to the user, current CGM devices fall under 2 categories: Flash glucose monitoring (or intermittently scanned glucose monitoring) and Real-time glucose monitoring.

 

FLASH GLUCOSE MONITORING

 

Flash glucose monitoring requires the user to hold a reader device (which can be a smart phone) close to the subcutaneously inserted sensor (the patient “scans” the sensor with the reader) to have the real-time interstitial glucose value displayed. During a scan, the reader displays the real time glucose value, glucose alerts, a historic glucose trend of values recorded and a trend arrow indicating the glucose direction (22). There are currently 2 approved Flash CGM devices for patient use, the FreeStyle Libre 14 day and the FreeStyle Libre 2 (Figure 5). The Libre 14 days allows for real-time data sharing but is limited by the lack of alarms in case glucose values are dangerously high or low. Nonetheless, this device may be appealing to those patients who want to minimize capillary blood glucose measurements and complain of CGM sensor alarm fatigue (23, 24). On the other hand, the Libre 2 has optional real-time glucose alarms but currently it requires a dedicated stand-alone receiver (data cannot be sent to a smartphone) and it does not have the capability of real-time data sharing.

 

REAL-TIME GLUCOSE MONITORING

 

Real-time glucose monitoring allows for data to be continuously sent to a receiver device and apart from viewing the display to check glucose levels and the direction of glucose profile, no additional action is required by the patient. Further, real-time CGM systems provide real-time alerts which can be customized to prevent or treat hyper or hypoglycemia. In addition, all currently approved real-time CGMs allow for data sharing.

 

Another advantage of CGM is the amount of data that can be generated and downloaded in customizable reports (Figure 7). Health care professionals are not only able to download daily glucose profiles in a graphic display but can also obtain several statistics including means, medians, standard deviations, interquartile ranges, and minimum and maximum values. This provides a better assessment of glycemic variability (Figure 8). Most importantly, time in glucose ranges can be identified and evaluated. This is particularly helpful in patients who have hypoglycemia unawareness and allows for adjusting the treatment plan by both the patient and practitioners to eliminate occurrence of hypoglycemia.

 

KEY CGM METRICS

 

Key CGM metrics include: Time in target range (TIR) defined as the percentage of readings and time per day within the recommended target glucose range of 70-180 mg/dL; time below target glucose range (TBR); and time above target glucose range (TAR) (see Figures 7 and 9 for examples). Current recommendations are to achieve TIR >70% (>16 h, 48 min), TBR <4% (<1 h) and TAR <25% (<6 h). However, recommendations are different for older adults/high-risk populations and during pregnancy (25). In addition to time in glucose ranges, CGM data has also allowed to generate a formula to estimate the laboratory A1C based on CGM mean glucose levels. This estimated A1C has been named “Glucose Management Indicator” and offers the advantage of being unaltered by limitations inherent to the laboratory A1C measurement (e.g., anemia, iron deficiency, glycation abnormalities, drug interference).  The enormous amount of data generated by CGMs can be overwhelming and difficult to follow and interpret and the need for a standardized report is critical for data interpretation and medical decision making. The Ambulatory Glucose Profile is a standardized report which incorporates all the core CGM metrics and recommended targets along with a 14-day composite glucose profile and is the recommended report by the International Consensus on Time in Range (Figure 9) (25, 26).

Figure 5. Examples of real-time continuous glucose monitoring systems.

Figure 6. Features of currently approved CGM devices in the United States.

Figure 7. A 14-day DEXCOM CGM overview report showing sensor glucose data over a 24-hour period including mean (dotted line), standard deviation, glucose management indicator, interquartile range (grey bars), upper and lower glucose thresholds (orange and red lines, set by the user), percent time in range, sensor usage, top patterns, and average daily calibrations.

Figure 8. A 7-day DEXCOM CGM overlay report showing daily profiles allowing for the identification of trends and patterns.

Figure 9. Ambulatory Glucose Profile (AGP) sample.

INSURANCE COVERAGE AND BILLING OF CGM DEVICES

 

Insurance coverage in the United States for devices is highly variable and challenging to navigate, and maybe unaffordable for some patients due to high copays or coverage issues. (These coverage requirements vary depending upon geographic area; practitioners are urged to follow guidelines in their country of practice). Understanding requirements for prescribing any CGM device is necessary and appropriate documentation is necessary. For individuals on Medicare to receive approval for a CGM device, documentation must include the following (as of 2021):

 

  1. The patient has diabetes mellitus and requires a therapeutic CGM.
  2. The patient is performing SMBG at least 4 times daily (Medicare only provides 3 test strips daily).
  3. The patient is treated with insulin and is injecting insulin at least 3 times daily or is on an insulin pump.
  4. The patient’s insulin treatment regimen requires frequent dose adjustment based on SMBG/CGM results.
  5. The patient had an in-person visit within 6 months prior to ordering the CGM with the treating practitioner to evaluate their diabetes and determine that criteria 1 to 4 are met. Subsequently, the patient must have an in-person visit every 6 months following the initial prescription to assess adherence to CGM and diabetes treatment plan.

 

There are billing codes for analyzing data from CGM devices. The patient visit should include certain key elements that need to be clearly documented in the chart as follows:

 

  1. A brief statement or narrative that the glucose sensor data were evaluated
  2. What patterns were noted
  3. Action steps and plan based on data interpretation provided to the patient
  4. Electronic or print of data report should be attached to the patient chart

 

CGM Integrated Insulin Pumps

 

As seen in Figure 10, some sensors are already integrated with insulin pumps (“sensor-augmented pumps”) so that the pump and receiver are in the same device. In addition, development of an integrated sensor and infusion set is currently being pursued, as this will simplify the incorporation of sensor technology into insulin pumps. Eventually, it is expected that all insulin pumps will be integrated with sensors. Yet, it should be appreciated that CGM is an equally important tool for MDI patients, and probably a more important diabetes management tool than using an insulin pump (21). Even after short periods of time, many patients can learn how to best use this technology to improve both mean glucose and glycemic variability. In a meta-analysis, comparing SMBG with RT-CGM, the latter achieved a lower A1C (between-group difference of change, -0.26%, (95% CI, -0.33% to -0.19%)) without increasing hypoglycemia (27). In the Juvenile Diabetes Research Foundation’s CGM trial, those individuals starting with baseline A1C levels under 7% overall had less hypoglycemia with CGM (28). A recent analysis of the T1D registry data suggests that CGM users, irrespective of insulin delivery method – i.e. multiple daily injections vs. pump therapy – had lower A1C levels than non-CGM users even after adjustment for confounding factors (29).

 

The American Association of Clinical Endocrinologists and American College of Endocrinology recommend the use of CGM for patients with T1D particularly for those with a history of severe hypoglycemia, hypoglycemia unawareness, and to assist in correction of hyperglycemia in patients not at goal. It may also be considered in pregnancy as it can help fine-tune insulin dosing, monitor for overnight hypoglycemia or hyperglycemia, and assess occurrence of postprandial hyperglycemia (30). The Endocrine Society guidelines on CSII Therapy and Continuous Glucose Monitoring in Adults recommend the use of RT-CGM for adult patients with T1D who either have A1C levels above target or well-controlled T1D and are willing and able to use these devices on a nearly daily basis (31).

Figure 10. Examples of modern-day insulin pumps.

OVERVIEW OF THERAPY FOR TYPE 1 DIABETES

 

Glycemic Targets

 

A1C is a measure of average glycemia over ~3 months and is a strong predictor of complications of diabetes (32). Current glycemic targets for adults from the American Diabetes Association (ADA) include a target A1C of <7%. However, it should be noted that this recommendation is a general target and the goal for the individual patient is as close to normal as possible (A1C of < 6%) without significant hypoglycemia. In addition, patients with T1D and hypoglycemia unawareness, long duration (> 25-30 years) of disease, limited life expectancies, very young children, or those with co-morbid conditions will require higher A1C targets. Individualized A1C targets need to be reviewed with each patient (17).

 

Thus, A1C testing should be performed routinely in all patients with diabetes as part of ongoing care. Frequency of A1C testing is determined based on the clinical situation, the treatment regimen used, and the clinician’s judgment. A1C measurements every 3 months help in the assessment of whether a patient’s glycemic targets have been reached. Although convenient, there are drawbacks to A1C measurements, as glycation rates may vary with patients’ race/ethnicity. Similarly, in patients with hemoglobinopathies, hemolytic anemia or other conditions that shorten the red blood cell life span, the A1C may not accurately reflect glycemic control or correlate with SMBG testing results. In such conditions, fructosamine may be considered as a substitute measure of long-term (average over 4 weeks) glycemic control. Clinicians should routinely compare downloaded SMBG or CGM averages with A1C as there are many reasons A1C may be altered due to a non-glycemic etiology and thus fructosamine or the downloaded glucose data itself would be a better metric to follow (33).

 

Non-Glycemic Treatment Targets

 

It should also be pointed out that in addition to glycemic targets, specific non-glycemic targets have also been recommended (34). Non-glycemic targets should also be tailored according to the individual with less stringent treatment goals for individuals with multiple coexisting illnesses and/or poor health and limited life expectancy. Recent real-world data from the T1D Exchange revealed that the incidence of cardiovascular disease (CVD) over 4.6 years was ~3.7% (35). Age, longer duration of diabetes, glycemic control, obesity, hypertension, dyslipidemia, and diabetic nephropathy were all associated with increased risk for CVD.

 

BLOOD PRESSURE

 

Good quality data to guide blood pressure management in T1D is lacking and most data are extrapolated from type 2 diabetes (T2D) clinical trials. The ADA recommends treatment to a goal of <140/90 mmHg for individuals with diabetes and hypertension at lower risk for CVD. Lower targets of <130/80 mmHg, should be considered for individuals who have higher cardiovascular risk or pre-existing ASCVD. Antihypertensive therapy should be initiated using a drug class that has demonstrated cardiovascular benefit such as angiotensin converting enzyme (ACE) inhibitors, angiotensin receptor blockers (ARBs), thiazide-like diuretics, or dihydropyridine calcium channel blockers. ACE inhibitors or ARBs are the preferred first line treatment for individuals with albuminuria.

 

LIPIDS

 

Very limited data exists for lipid management in patients with T1D of any age. Limited evidence suggests that primary prevention with lipid-lowering medications decreases the incidence of CVD (36). The ADA has adopted the approach of the 2018 American College of Cardiology/ American Heart Association multi-society cholesterol guidelines and recommends similar statin approaches for individuals with T1D (34). All patients with T1D and CVD should be treated with high intensity statins. Addition of non-statin therapies such as ezetimibe and PCSK9 inhibitors should be considered based on overall risk and achieved LDL-C thresholds. Patients with T1D over the age of 40 should be offered statin therapy. In individuals younger than age 40 with T1D and additional risk factors (such as albuminuria, HTN, strong family history, long duration of diabetes >20 years), moderate intensity statin therapy should be considered after clinical discussion. Recently, a prediction model for CVD events in T1D to help decision making for primary prevention that has been developed and shows promise but needs further validation (37). There is new evidence of the contribution of cardiac autoimmunity to CVD in T1D in the DCCT/EDIC cohort that warrants further investigation (38). 

 

INSULIN THERAPY

 

Insulin therapy is the cornerstone of management of T1D as beta cell dysfunction or destruction progressively leads to absolute insulin deficiency. Physiologic insulin replacement that aims to mimic normal pancreatic insulin secretion is the preferred method of treatment of T1D patients. Basal insulin is the background insulin required to suppress hepatic glucose production overnight and between meals. Prandial (bolus or meal-time) insulin replacement, provides enough insulin to dispose of glucose after eating. Such a therapeutic insulin regimen providing both basal and bolus insulin allows flexibility of dosing. Older twice-daily combination of regular and NPH regimens generally should not be used in T1D as they are less effective since the time-action profile of these two standard insulins do not readily allow for the clear separation of basal and prandial insulin action. However, it may be necessary to use such regimens in patients who cannot otherwise afford insulin. It also should be pointed out that for newly diagnosed patients with T1D, transient use of once- or twice-daily basal injections is sometimes adequate.

 

Principles of Management of T1DM

 

Management of T1D involves a multidisciplinary framework that includes the following:

 

  1. Physiologic insulin replacement using basal-bolus therapy, either as MDI or CSII
  2. Blood glucose monitoring with SMBG and/or CGM with development of individualized A1c goals
  • Patient education
  1. A supportive team of providers including endocrinologists, nurses, certified diabetes care and education specialists (CDCES)s, pharmacists, psychologists, dietitians, social workers, other specialists such as cardiologists, nephrologists, psychiatrists as well as family members, social support groups etc.

 

Types of Insulin

 

Selecting the appropriate insulin depends largely on the desired time course of insulin action. Table 1 shows the pharmacokinetic characteristics—time to onset of action, time of peak action, effective duration of action, and maximum duration of action—of currently available insulins; however, these can vary considerably among individuals.

 

Insulin products are categorized according to their action profiles:

 

  • Rapid-acting: e.g., insulin lispro, insulin aspart, and insulin glulisine (genetically engineered insulin analogs)
  • Short-acting: regular (soluble) insulin
  • Intermediate-acting: NPH (isophane)
  • Long-acting, e.g., insulin glargine, insulin detemir, and insulin degludec (genetically engineered insulin analogs)
  • Pre-mixed insulin
  • Inhaled insulin

 

Insulin analogs are insulin molecules modified by genetic engineering and recombinant DNA technology. The amino acid structure of insulin is altered to change the properties of insulin – i.e., time to onset, peak, and duration of action, compared to human regular insulin. However, the biological properties and stability of the insulin molecule are intact. A general principle to bear in mind is the longer the time to peak, the broader the peak and the longer the duration of action. Additionally, the breadth of the peak and the duration of action will be extended with increasing dose. Figure 11 should therefore be considered a conceptual representation of insulin action curves.

 

Mealtime (Prandial) Insulins

RAPID-ACTING INSULIN

 

These are insulin analogs with a rapid onset in 15-30 minutes, peak in 30-90 minutes, and an effective duration of 4 to 5 hours when injected subcutaneously.  They have a shorter time action profile compared to human (regular) insulin because they do not self-aggregate in solution. All rapid-acting insulin analogs have a 1 - 2 amino acid difference from the primary structure of human insulin. Insulin lispro differs from human insulin by an amino acid exchange of lysine and proline at positions B28 and B29 (39). The substitution of aspartic acid for proline at position B28 characterizes insulin aspart (40). Insulin glulisine differs from human insulin in that the B3 asparagine is replaced by lysine, and B29 lysine is replaced by glutamic acid (41). These modifications in the primary structure of human insulin increase the rapidity of breakdown of insulin hexamers in the analogs and thus result in more rapid absorption. When administered before meals, rapid-acting insulins used as part of multiple daily injections (Figure 11) or with CSII, resemble physiologic insulin increases stimulated by food. Doses can be adjusted proportionate to food consumed; in patients with gastroparesis or poor appetite, insulin can be injected halfway through or after the meal. A follow-on biologic to insulin lispro (biosimilar lispro) is now available as Admelog. 

 

 Ultra-rapid acting insulin aspart (Fiasp) available since 2018 is insulin aspart with added niacinamide. This results in quicker absorption with faster onset of action after injection and therefore can be injected right before the start of a meal (or within 20 minutes after the start of a meal). This allows for some flexibility of dosing. Safety and efficacy data in adults and children is similar to insulin aspart (42). Fiasp has recently also been approved for use in insulin pumps. Data in pregnant women is lacking. Recently, ultra-rapid acting lispro (lispro-aabc) has become available in several countries including the United States. This insulin has been shown to appear in the bloodstream within 1 minute of injection (43). Ultra-rapid acting lispro was found to be non-inferior to rapid-acting lispro and superior for postprandial blood glucose control in T1D and T2D (44, 45).

 

INHALED INSULIN

 

Currently, one form of inhaled insulin is available in the market. Afrezza was approved by the FDA in 2014. This is a drug-device combination that contains powdered human insulin in single use dose cartridges delivered via a small inhaler. When inhaled, it dissolves immediately on contact with the alveolar surface of the lung and is rapidly absorbed into the systemic circulation, reaching a peak within 15 minutes. Thus, Afrezza acts similar to rapid-acting insulin analogs but with a much faster peak of action, and shorter duration of action. Prior to initiation of its use, patients should be screened for underlying lung disease with spirometry. Follow-up spirometry is recommended after 6 months’ use, and annually thereafter. The main advantages of inhaled insulin are avoidance of injections, faster onset of action, less weight gain, and less hypoglycemia (46). Dosing is not flexible as cartridges are available in fixed doses (4, 8 and 12 units). Afrezza is contraindicated in patients with chronic lung disease such as asthma or chronic obstructive pulmonary disease (COPD).

 

SHORT-ACTING INSULIN

 

Regular insulin is structurally similar to endogenous human insulin. It consists of dissolved zinc-insulin crystals which self-aggregate in the subcutaneous tissue and results in a delayed onset of action of 30 to 60 minutes, a peak of 2 to 3 hours, and an effective duration of 6 to 8 hours. Proper use requires injection at least 20 to 30 minutes prior to meals to match insulin availability and carbohydrate absorption. Use of regular insulin is associated with greater hypoglycemia risk (47). Regular insulin acts almost instantly when injected intravenously.

 

Basal Insulins

 

INTERMEDIATE-ACTING INSULIN

 

Neutral protamine Hagedorn (NPH) insulin, developed in the 1950s, is a combination of recombinant human insulin with protamine which results in crystal formation. When injected subcutaneously, precipitated crystals of NPH insulin are released slowly resulting in a longer duration of action compared to regular insulin. Action of NPH varies quite widely within the same patient as well as between patients.  Its onset of action occurs 2 to 4 hours from the time of injection, with a peak effect lasting 6 to 10 hours, and an effective duration of 10 to 16 hours.  Due to this peak effect, NPH insulin acts as a basal and a prandial insulin, necessitating that patients eat a meal at the time the insulin is peaking. NPH typically requires twice a day dosing (48).

 

LONG-ACTING INSULIN ANALOGS

 

Long acting insulin analogs were created by modifying the amino acid sequence on the beta chain of insulin (49). They exhibit much improved pharmacokinetics and pharmocodynamics without a peak effect and maintain a longer duration of action. Improved absorption rates result in significantly decreased inter-individual and intra-individual variability with improvement in glycemic control and reduced hypoglycemia risk. 

 

Insulin glargine is a modified human insulin produced by the substitution of glycine for asparagine at position A21 of the insulin molecule and by the addition of two arginine molecules at position B30 (48). These changes result in an insulin molecule that is less soluble at the injection site forming a precipitate in the subcutaneous tissue to form a depot from which insulin is slowly released after injection and is slowly released into the circulation. It has no pronounced peak and a longer duration of action of about 20 to 24 hours in most patients, allowing for once daily dosing. In clinical practice, many patients with T1DM may benefit from twice-daily injections.  Insulin glargine is solubilized in acidic pH and should not be mixed with rapid-acting insulins as the kinetics of both insulins will be altered. Insulin glargine shows a greater reduction in A1C and decreased hypoglycemia in patients with T1DM compared to NPH insulin (50).

 

Insulin detemir is a soluble basal insulin analog. It is covalently acylated with fatty acids on the lysine at position B29, which allows for reversible binding to albumin (51). This delays its absorption from subcutaneous tissue and prolongs its time in the circulation. Although the mean duration of action of insulin detemir has been shown to be 24h, one study showed shorter duration of action (about 17h), which suggests that most patients with T1D may require twice-daily dosing of insulin detemir (52).

 

ULTRALONG-ACTING INSULIN ANALOGS

 

Insulin degludec is an ultra-long acting basal insulin available in the US since 2015 that has the same amino acid sequence as human insulin, apart from the deletion of the threonine amino acid residue at B30 and the addition of a fatty acid to the lysine at B29 (53).  The fatty acid moiety causes self-aggregation of insulin molecules into soluble multihexamers. Slow dissociation of zinc from the insulin allows for gradual and stable absorption of insulin monomers resulting in a long half-life and a prolonged duration of action of 42 hours at steady state. In patients with T1D, similar A1C reduction with lower rates of nocturnal hypoglycemia have been reported with insulin degludec compared with insulin glargine (54, 55). The extended duration of insulin degludec allows for more flexibility of day-to-day dose timing without compromising glycemic control or safety (56).

 

U-300 glargine (Gla-300) is a formulation of insulin glargine that delivers the same number of insulin units as insulin glargine 100 units/mL (Gla-100), but in a third of the volume. The compact depot renders a smaller surface area of insulin glargine for a given dose, leading to a slower release of insulin glargine over time. This translates into a more constant PK/PD profile, with a prolonged duration of action (up to 30 hours) with Gla-300 compared with Gla-100 in patients with T1DM (57). Gla-300 has been shown to provide similar glucose control compared to Gla-100 with less weight gain and hypoglycemia (58).

 

Pre-Mixed Insulins

 

Premixed insulins are mixtures of prandial and intermediate acting insulins (the same prandial insulin attached to protamine so that it becomes intermediate acting). Insulin mixtures are available as human insulin mixtures (NPH and regular mixture) as well as analog mixtures. In the US, insulin lispro protamine mixtures are available in two forms: 75% insulin lispro protamine suspension and 25% insulin lispro injection (75/25) and 50% insulin lispro protamine suspension and 50% insulin lispro injection (50/50). Available preparations of insulin aspart protamine mixtures include 50/50 and 70/30 suspensions. A variety of other ratios are available in Europe. There is only one mixture of analog-analog without protamine (aspart 30% +degludec 70%, Ryzodeg). These insulin mixtures are typically administered before breakfast and dinner. This alleged twice daily dosing is the primary advantage of these insulins. In general, use of premixed insulins restricts adjustment of doses and meal timing. Therefore, premixed insulins are not recommended for adult patients with T1D, where intensive regimens with ability to make adjustments in the premeal short-acting insulin bolus are better suited for glycemic control. Premixed insulin in T1D could have benefit for some patients who do not adhere to an intensive insulin regimen, and with consistent food intake and timing of meals.

 

Concentrated Insulins

 

U-500 INSULIN

 

U-500 insulin is highly concentrated regular insulin, administered 2-3 times a day without basal insulin. Due to its concentration, the action is prolonged and variable. In T1D, use is primarily limited to individuals with significant insulin resistance (requiring >200 units of insulin a day). Caution should be used while prescribing this insulin as confusion may occur among clinicians, pharmacists, nurses, and patients who are unfamiliar with its use. U-500 insulin is also available in a pen delivery system allowing patients to administer insulin by 5 units increments up to a maximum of 300 units at a time. Units to be delivered are clearly readable through the pen “dose window” which should minimize or eliminate confusion when administering this highly concentrated insulin formulation.

 

CONCENTRATED INSULIN ANALOGS

 

U-200 formulations of insulin lispro and insulin degludec are also available and allow for delivery of lower volumes and therefore better absorption. U-300 glargine is available in pen form and holds up to 900 units of insulin with dosing capability up to 160 units per dose.

 

CONVERSION FROM U-100 TO CONCENTRATED INSULIN

 

Switching from U-100 insulin to concentrated insulin may occasionally be necessary in the setting of severe insulin resistance and use of large amount of U-100 insulin. U-200 lispro is bioequivalent to U-100 lispro, and U-200 degludec is bioequivalent to U-100 degludec. This means that the dose can be converted 1:1 on a unit basis when switching from U-100 to U-200 formulation. The insulin is delivered at 50% less volume. U-300 glargine, on the other hand is not bioequivalent to U-100 glargine. Individuals with T1D often require 15-20% higher dose of U-300 glargine. Similarly, a dose reduction of 20% is essential when switching from U-300 glargine back to U-100 glargine to avoid hypoglycemia. When initiating U-500R, dosing should be determined based on current and targeted glycemic goals to optimize efficacy and safety. U-500R provides mealtime coverage and its extended duration of action provides basal coverage also.

 

Biosimilar Insulins/Follow-on Biologics

 

According to the FDA, a “biosimilar” is a biological product that is highly similar to a US-licensed reference biological product not withstanding minor differences in clinically inactive components, and for which there are no clinically meaningful differences between the biological product and the reference product in terms of the safety, purity, and potency of the product. As of 2020, there are 4 follow-on biologics approved. These include Basaglar (US, Europe - insulin glargine), Basalin (China- insulin glargine), Semglee (EU, Australia, insulin glargine) and

 

Table 1. Currently Available Insulin Preparations

Insulin Preparation

Onset of action (h)

Peak          Action (h)

Effective duration of action (h)

Maximum duration(h)

Rapid-acting analogs

 

 

 

 

     Insulin lispro (Humalog, Admelog) 

¼ - ½  

½-1 ½

3-4      

4-6

     Insulin aspart (NovoLog)

¼ - ½

½ -1 ¼

3-4                   

4-6

     Insulin glulisine (Apidra)

¼ - ½

½ -1 ¼

3-4         

4-6

     Insulin aspart (Fiasp)

¼ -1/3

 1.5-2.5

3-4

5-7

     Insulin lispro-aabc (Lyumjev)

1/8

2

 

4-6

Inhaled insulin (Afrezza)

seconds

12-17 min

2-3

2-3

Short-acting

 

 

 

 

     Regular (soluble)

½ - 1

2-3

3-6

6-8

Intermediate-acting

 

 

 

 

     NPH (isophane)

2-4

6-10

10-16

14-16

Long-acting analog

 

 

 

 

     Insulin glargine (Lantus, Basaglar)

0.5-1.5

8-16

18-20

20-24

     Insulin glargine U-300 (Toujeo)

0.5-1.5

none

24

30

     Insulin detemir (Levemir)

0.5-1.5

6-8

14

~20

     Insulin degludec (Tresiba)

0.5-1.5

none

24

40

 

Figure 11. Available basal insulins and duration of action. Figure source Ref (59).

Factors Influencing Insulin Absorption

 

Insulin absorption variability is one of the greatest obstacles to replicating physiologic insulin secretion. Among the many factors that affect insulin absorption and availability (Table 2) are injection site, the timing, type or dose of insulin used, and physical activity. Day-to-day intra-individual variation in insulin absorption is approximately 25%, and the variation between patients may be as high as 50%. This occurs more commonly with larger doses of human insulin which form a depot and can unpredictably prolong duration of action; however, this is less of an issue with rapid-acting insulin analogs. In general, any strategy that increases the consistency of delivery should decrease glucose fluctuations; and insulin regimens that emphasize rapid-acting insulin are more reproducible in their effects on blood glucose levels. Insulin pumps using a rapid-acting insulin analog can significantly reduce glucose variability. Like multiple-injection regimens, use of an insulin pump requires frequent blood glucose monitoring. In addition, pump users need a back-up method of insulin administration, and attention to mechanical and injection site issues.

 

Reducing Variability of Insulin Absorption

 

INJECTION SITES

 

Subcutaneous insulin is absorbed most rapidly when injected into the abdomen, followed by the arms, buttocks and thighs. These differences are likely due to variations in regional blood flow. A single region should be utilized for injections without rotation between regions, as this may result in day-to-day variation of insulin absorption. However, while using a region, site rotation (i.e. – rotating injections systematically within the abdomen) is important to avoid development of lipohypertrophy or atrophy due to repeated injections at the same site. Injection into lipohypertrophic areas results in erratic, slower absorption of insulin. Exercise increases the rate of absorption from injection sites, likely by increasing blood flow to the skin; local effects may also be involved.

 

TIMING OF PRE-MEAL INJECTIONS

 

Gauging the appropriate interval between preprandial injections and eating, known as the “lag time,” is essential for coordinating insulin availability with glycemic excursions following meals. The timing of the injections should also be adapted to the level of premeal glycemia. Insulin lispro, insulin aspart, and insulin glulisine have rapid onset of action and, ideally, should be given approximately 10-20 minutes before mealtime when blood glucose is in the target range, keeping in mind that if the meal is delayed, hypoglycemia may ensue. When blood glucose levels are above a patient’s target range, the lag time should be increased to permit the insulin to begin to have an effect sooner. In this case, rapid-acting acting insulin analogs can be given 20-30 minutes before the meal, depending upon the degree of hyperglycemia. If premeal blood glucose levels are below target range, administration of rapid-acting insulin should be postponed until after some carbohydrates have been consumed. Use of frequent home glucose monitoring or CGM can assist in determining appropriate lag times. It is important to emphasize the effect of administering prandial insulin up to 20 minutes before a meal. Pre-bolusing has been shown to reduce post-prandial glucose spike by up to 50 mg/dL.

 

OTHER FACTORS

 

Exercise, as discussed earlier, results in increased blood flow to muscle groups and can increase rate of insulin absorption. Heat can also increase the rate at which insulin is absorbed from the skin. For example, being out in the sun or injection before going into a hot tub may lead to hypoglycemia. Intra-muscular injections result in a more rapid onset of action compared to subcutaneous tissue. This route can be utilized under certain situations such as ketoacidosis, insulin pump failure or in the event of profound hyperglycemia.

 

Table 2. Factors Affecting the Bioavailability and Absorption Rate of Subcutaneously Injected Insulin

Factor

Effects

Site of injection

Abdominal injection (particularly if above the umbilicus) results in the quickest absorption; arm injection results in quicker absorption than thigh or hip injection.

Depth of injection

Intramuscular injections are absorbed more rapidly than subcutaneous

injections.

Insulin concentration

U-40 insulin (40 units per mL) is absorbed more rapidly than U-100 insulin (100 units per mL).

U-40 insulin is an old insulin formulation not available in the United States for patient use. Currently, it is used for treating canine and feline diabetes mellitus

Insulin dose

Higher doses have prolonged duration of action compared with lower doses.

Insulin mixing

Regular insulin maintains its potency and time-action profile when it is

mixed with NPH insulin

Exercise

Exercising a muscle group before injecting insulin into that area

Increases the rate of insulin absorption.

Heat application or Massage

Local application of heat or massage after an insulin injection increases

the rate of insulin absorption.

 

Role of Insulin Analogs in Management of T1D

 

Most of the problems of insulin replacement in T1D arise from the fact that subcutaneous injection or pump infusion remains a relatively poor route of administration. From the subcutaneous site of injection, insulin is absorbed into the systemic, not portal circulation. More importantly, subcutaneous injection leads to variable absorption from one injection to another, due largely to the non-physiologic pharmacokinetics of standard insulins. Insulin analogs were developed to overcome this problem.

 

Currently there are three rapid-acting insulin analogs: insulin lispro, insulin aspart, and insulin glulisine, all of which have a rapid onset of action and peak, thereby improving 1- to 2-hour postprandial blood glucose control compared with regular insulin. These rapid-acting analogs must be used in conjunction with a basal insulin to improve overall glycemic control (Figures 11 and 12). Importantly, the rapid-acting analogs have consistently outperformed regular insulin in terms of post-absorptive hypoglycemia. This finding should not be surprising since the duration of regular insulin is much longer than the gut absorption of a typical mixed meal.

Figure 12. Idealized insulin curves for prandial insulin with a rapid-acting analog (RAA) with basal insulin glargine or insulin detemir. Each insulin preparation is responsible for either the prandial or basal component. Many patients find the basal insulins do not last the entire 24 hours and they give the basal insulin twice daily. B=breakfast; L=lunch; S=supper; HS=bedtime

Clinical trials have demonstrated lower fasting glucose levels and less nocturnal hypoglycemia with insulin glargine than with NPH insulin, advantages that are especially relevant in patients aiming for meticulous control (A1C <7%) or those with hypoglycemia unawareness. Trials with T1D have shown similar results with insulin detemir which compared with NPH insulin was equally effective in maintaining glycemic control, although detemir was administered at a higher molar dose. The newest basal insulin preparations, insulin degludec and U-300 insulin glargine are claimed to show less nocturnal hypoglycemia than insulin glargine or insulin detemir.  In general, hypoglycemia is reduced with any of these basal analog insulins compared to NPH insulin. Since hypoglycemia is clearly one of the treatment-limiting aspects of T1D therapy, the use of these analogs has gained wide-spread acceptance.

 

Multiple Daily Injection (MDI) Insulin Therapy

 

A simpler conceptual approach preferred by most patients with T1D is using a prandial insulin analog for each meal (i.e., insulin lispro, insulin aspart, or insulin glulisine) and a separate basal insulin analog (i.e., insulin glargine, insulin detemir, or insulin degludec). Although these true basal-prandial regimens require more shots than conventional twice-daily regimens, they are considerably more flexible, allowing greater freedom to skip meals or change mealtimes. Moreover, use of the long-acting basal and rapid-acting insulin analogs, allows strategies to achieve individual, defined blood glucose targets more easily. Such modifications might include changing the timing of insulin injections in relation to meals, changing the portions or content of food to be consumed, or adjusting insulin doses or supplements for premeal hyperglycemia.

 

The basic treatment principles of insulin dosing include establishing a total daily dose, an insulin to carbohydrate ratio and an insulin sensitivity or correction factor. 

 

ESTABLISHING A TOTAL DAILY DOSE (TDD) OF INSULIN

 

This is the first step in starting treatment in a patient with newly diagnosed diabetes. This dose can vary based on the individual and can range from 0.3- 1.5 units/kg/day.  A good starting dose is ~0.5 units/kg/day. Once the TDD is determined, this number is divided by half to establish the basal and bolus requirements.  As a general rule of thumb, half the insulin is used as basal insulin, while the other half is used as prandial or mealtime insulin.For example, in a person weighing 75 kg, a typical total daily insulin dose might be 75 kg X 0.7 units/kg = roughly 37 units/day. The basal insulin dose would be roughly 18 units and bolus insulin total would be 18 units (divided amongst meals, see below).

 

Long-acting insulin analogs U-100 glargine and detemir can be administered once or twice daily. Insulin degludec or U-300 insulin glargine can be administered once a day.

 

USING PRANDIAL INSULIN

 

Establishing an Insulin to Carbohydrate (Carb) Ratio

 

Patients with T1D derive the greatest therapeutic benefit when basal and prandial analogs are used together, because the physiologic pharmacokinetics and pharmacodynamics of these analogs make separating the basal and prandial components of insulin replacement easier. In general, administering the appropriate amount of pre-meal insulin requires that the patient know at least their current blood glucose level and the estimated amount of carbohydrates for a meal. Initially, the amount of prandial insulin can be determined by approximating the percentage of calories consumed at each meal. As patients become more educated, however, they may alter the prandial dose by estimating the carbohydrate component of each meal or snack. As patients become more sophisticated, they may note that the same carbohydrate quantity may have a different effect on their blood glucose level depending upon the specific type of meal consumed.

 

The carb ratio provides the dose of rapid acting insulin (lispro, aspart, glulisine) to cover the carbohydrate content of a meal. A typical starting point in patients with T1D is to give 1 unit of rapid acting insulin for every 15 grams of carbohydrates. This ratio is variable ranging from 1 unit for every 5g to 30 g of carbohydrate. To estimate the carb ratio, the “500 rule” can be used:

 

500/total daily dose (TDD) = grams of carbohydrate covered by 1 unit of insulin.

 

Example: A person who takes a total of 50 units of insulin per day (both basal and prandial combined) will need 1 unit of rapid acting prandial insulin for every 10g carbohydrate (500/50 = 10g of carbohydrate covered by 1 unit of insulin, using above formula).

 

Alternative way to calculate the carb ratio – Add all carbohydrates consumed in a day and divide this by the total units of prandial insulin taken that day, using an average over 3 days.

 

Prandial insulin may be reduced/skipped when:

 

  • Extra carbohydrates are used to raise low blood sugars or cover increased physical activity
  • Recent dose of correction insulin within past 1-2 h
  • Nausea or vomiting preventing oral intake

 

Determining the Correction Dose or “Insulin Sensitivity Factor” (ISF)

 

In addition to covering the carbohydrate load of a meal, individuals will also need to correct hyperglycemia, called the “correction dose”. The method commonly used for this is the “1800 Rule”. This estimates the point drop in glucose for every unit of rapid-acting insulin administered:

 

1800/TDD = Point drop in glucose for 1 unit of rapid-acting insulin

This ISF (also called the correction factor) can be used for between-meal elevations in blood glucose. Thus, in general this correction dose can be utilized anytime provided the patient has not taken an injection of rapid acting insulin over the past 2-4 hours (insulin on board, Figure 12).  

 

Target glucose: The ISF enables achieving appropriate individualized blood glucose targets.

 

For example: A person who takes a total of 60 units of insulin per day will require 1 unit of rapid acting insulin to drop the glucose by 30 points. If the patient’s glucose is 180 mg/dL and the glucose target has been set at 120 mg/dL, a correction dose of 2 units would be required to bring the glucose down to target:

 

  1. ISF = 1800/60 (TDD) = 30; 1 unit of rapid-acting insulin will decrease glucose by 30 points
  2. 180 mg/dL (actual glucose level) – 120 mg/dL (target glucose level) = 60; this is the excess glucose, that is, the value that is above target and that needs to be corrected
  3. 60/30 (ISF) = 2; dividing the excess glucose by the ISF will provide the amount of correction insulin units that are required to bring down the glucose to target, in this case it will be 2 units.

 

Putting it All Together - Combining the Carb Ratio and ISF

 

Combining the carbohydrate load and ISF will enable patients to appropriately target their pre-meal glucose. 

 

For example: An individual with a carb ratio of 1:15 and ISF of 1 unit/50mg/dL, prior to a meal of 60g carbohydrates and a pre-meal blood glucose of 220mg/dL and target of 120mg/dL would take the following steps to administer the appropriate amount of prandial insulin as follows:

 

  1. To cover carbohydrate intake: 60g/15g per unit =4 units
  2. Correction dose: 220 mg/dL (actual glucose)– 120mg/dL (target glucose) = 100mg/dL. ISF is 100/50 = 2 units to correct.
  3. Total amount of prandial insulin: 4+2= 6 units

 

Insulin Titration and Pattern Adjustments

 

Reviewing blood glucoses and recognizing patterns is one of the most important aspects of diabetes management, allowing for timely and appropriate adjustments in insulin dose, food intake, and managing physical activity. Pattern management is aided by valuable tools such as SMBG with information obtained through download software (see above) or logbooks and CGM data. These tools can be used in order of priority, for assessment of hypoglycemia, hyperglycemia, glycemic variability, frequency of SMBG readings, etc.

Figure 13. The appearance of insulin into the blood stream (pharmacokinetics) is different than the measurement of insulin action (pharmacodynamics). This figure is a representation of timing of insulin action for insulin aspart from euglycemic clamp data (0.2 U/kg into the abdomen). Using this graph assists patients to avoid “insulin stacking”. For example, 3 hours after administration of 10 units of insulin aspart, one can estimate that there is still 40% X 10 units, or 4 units of insulin remaining. By way of comparison, the pharmacodynamics of regular insulin is approximately twice that of insulin aspart or insulin lispro. Currently used insulin pumps keep track of this “insulin-on-board” to avoid insulin stacking. Adapted from reference (40).

INSULIN DELIVERY SYSTEMS

 

Significant improvement in pharmacokinetics and pharmacodynamics of insulin analogs and advances in technology has allowed for insulin delivery systems to resemble endogenous insulin secretion as closely as possible.

 

Insulin Pens

 

Insulin pens were first introduced in 1981 as injection devices. These pens contain a cartridge holding insulin which is injected into the subcutaneous tissue through a fine, replaceable needle. Insulin pens are convenient, portable and are widely used as a part of MDI therapy. Currently, insulin pens are available as disposable pens containing prefilled cartridges or reusable insulin pens with replaceable insulin cartridges. Several insulin pens allow the convenience of ½ unit dosing, a critical need for pediatric patients and those adults with high insulin sensitivity and low insulin requirements.

 

Insulin smart pens - The first insulin smart pen was approved for use in the United States (InPen, Companion Medical, California, USA) in 2017 (Figure 14). Smart pens can record timing and amount of each administered insulin dose, display the last dose and insulin onboard and also make dosing recommendations based on pre-specified information (Figure 15). This information is wirelessly transmitted via Bluetooth to a dedicated mobile application on a smartphone device. Other similar devices are in development including the NovoPen 6 and NovoPen Echo Plus reusable insulin pens equipped with near-field communication technology (Novo Nordisk, Denmark), recently approved in the European Union.

Figure 14. InPen Smart Insulin Pen

Figure 15. InPen Insight report. Report provides missed doses, bolus calculator dosage, long-acting insulin assessment and CGM data (if paired with the InPen app).

Continuous Subcutaneous Insulin Infusion Therapy (CSII)

 

While not a new tool, insulin pump therapy remains the gold standard of insulin delivery for T1D (Figure 9). CSII is the most precise way to mimic normal insulin secretion because basal insulin infusion rates can be programmed throughout a 24-hour period. Essentially, the CSII pump may be thought of as a computerized mechanical syringe automatically delivering insulin in physiologic fashion. Patients can accommodate metabolic changes related to eating, exercise, illness, or varying work and travel schedules by modifying insulin availability. Basal rates can be adjusted to match lower insulin demands at night (between approximately 11 PM and 4 AM) and higher requirements between 3 AM or 4 AM and 9 AM.

 

Various studies comparing glycemic control during CSII versus intensive insulin injection regimens have been published. A meta-analysis of 12 randomized controlled trials of CSII versus multiple injection regimens showed a weighted mean difference in blood glucose concentration of 16 mg/dL (95% CI 9-22) and a difference in A1C of 0.5% (0.2-0.7) favoring CSII (60). The slightly but significantly better control in patients on CSII was accomplished with a 14% average reduction in daily insulin dose.

 

A meta-analysis funded by the Agency for Healthcare Research and Quality showed that in adults with T1D A1C levels decreased more with CSII than multiple injections, but one study heavily influenced this finding  (27). For both children and adults, there was no difference in severe hypoglycemia. The common misconception that CSII leads to more hypoglycemia is not valid.

 

Modern insulin pumps are much smaller and easier to use than the pumps of the past (Figure 10).

 

With the exception of insulin lispro-aabc (Lyumjev), all rapid-acting analogs are approved in the United States for use in insulin pumps. The basal rate of the insulin pump replaces the use of daily injections of basal insulin. The boluses given before each meal are essentially the same as normal insulin injections of rapid acting insulin. The pump allows programming of several different basal infusion rates at increments that can range from 0.025 up to 35.0 units/hour (usually ranging from 0.4 to 2.0 units/hour) to meet non-prandial insulin demands, though it is unlikely that the average patient will require more than 2 or 3 different rates (Figure 16). As with MDI, correction doses can be provided before or between meals. Figures 17 and 18 show data that is typically downloaded from a pump.

Figure 16. Idealized insulin curves for CSII with either insulin lispro, insulin aspart, or insulin glulisine. Note the basal insulin component can be altered based on changing basal insulin requirements. Typically, insulin rates need to be lowered between midnight and 0400 h (predawn phenomenon) and raised between 0400 h and 0800 h (dawn phenomenon). The basal rate the rest of the day is usually intermediate to the other two. Modern-day pumps can calculate prandial insulin dose by the patient entering the blood glucose concentration and the anticipated amount of carbohydrate to be consumed. The pump calculates how much previous prandial insulin is still active and provides the patient a final suggested dose which the patient may activate or override.

There are many fundamental differences between CSII and MDI. These include:

 

TITRATION OF BASAL RATES

 

From a practical point of view, the first and most important insulin dose to provide in a correct amount is the basal rate. If the basal dose is set incorrectly, neither the bolus doses nor the correction doses will be appropriate. A common mistake observed in CSII therapy is that the basal dose is set too high, making the administration of even small insulin correction doses result in hypoglycemia. The greatest advantage of CSII is it allows more flexibility and titration of the basal doses.

 

The basal dose can be titrated throughout the day to meet patients’ individual needs and this should be done in a systematic manner by performing “basal checks.” Prior to starting a basal rate assessment (basal check), the following conditions should be met for the day of the test: last meal and/or insulin bolus should have occurred at least 4 hours prior to starting the assessment; last meal should preferentially be low in fat and not have too much protein; avoid exercise and alcohol; do not perform the assessment if hypoglycemia has occurred earlier in the day or there is an inter-current illness. Of note, it is recommended to repeat the assessment on several occasions to identify a pattern prior to making adjustments to the basal rate. 

 

Nighttime Basal Rate

 

It is usually best to start by addressing the overnight basal rate. An overnight basal assessment is performed on a night the patient has a bedtime glucose level within target. The patient is asked not to have anything to eat during the assessment. The patient then measures glucose levels at bedtime, midnight, 3AM and upon awakening to assess for changes in glucose profile (the use of a CGM obviously makes this exercise much easier). Glucose should also be checked in case of hypoglycemic symptoms. If hypoglycemia ensues or glucose level rises above target, the assessment is stopped and the patient treats the glucose level accordingly. Rises or falls of ≤ 30 mg/dl from bedtime to morning (upon awakening) are usually acceptable. By contrast, glucose changes > 30 mg/dl will require adjustments in basal rates usually consisting of 10-20% changes in insulin dose (as deemed clinically appropriate) starting 2 hours before the observed rise or fall in glucose levels. In general, a change in a basal dose takes two to four hours to result in a change in blood glucose.

 

Daytime Basal Rates

 

Daytime basal rates are checked by assessing the glucose profile across a skipped-meal time segment (i.e., pre-breakfast to pre-lunch, pre-lunch to pre-dinner, and pre-dinner to bedtime). To check the “pre-breakfast to pre-lunch” time segment, breakfast is skipped and glucose level is checked at 1-2 hour intervals for the duration of the time segment (prior to lunch). Glucose levels should also be checked in the event of hypoglycemic symptoms. The same recommendations regarding changes in glycemic levels requiring insulin dose adjustments described for the overnight basal assessment apply here. 

 

TRACKING OF INSULIN-ON-BOARD

 

Another major difference between CSII and MDI is the pump can accurately track the insulin-on-board for safer use of correction doses (Figure 13). As noted above, doing this accurately can have a major impact in preventing insulin stacking.

 

INSULIN DOSE CALCULATOR

 

Insulin-to-carbohydrate ratios and insulin sensitivity factors with corresponding target glucose values can be set and modified as needed in insulin pumps. Patients are only required to enter their glucose level and/or anticipated carbohydrate amount to be consumed and the insulin pump will calculate the insulin dose and recommend a bolus dose. So, the complicated mathematics to best utilize MDI are done automatically with CSII.

 

MODIFICATIONS TO BOLUS DELIVERY

 

Pumps can be programmed for individual boluses to be administered over an extended period of time (“extended” or “square wave” bolus). This feature may be particularly helpful for very high-fat meals or those patients with delayed gastric emptying, seen with gastroparesis or in those receiving pramlintide (see below).

 

TEMPORARY BASAL RATES

 

The other major advantage of CSII is that it allows the use of “temporary basal rates.” This is extremely helpful in situations where metabolic demands have “temporarily” changed such as during illness (requiring an increase in insulin dose) or during exercise (requiring a dose reduction). Again, due to the time action of the rapid-acting analogs, sufficient time must be incorporated when using a temporary basal rate.

 

DOWNLOAD CAPABILITY

 

Pump data can be downloaded, and the data obtained is extremely helpful in understanding patients’ glycemic responses to an established insulin regimen (Figure 17). Also, it can assist in evaluating patients’ behaviors pertaining to their glucose management. Downloads provide information regarding the total daily insulin use broken down into percentages corresponding to basal and bolus delivery. This allows determining if patients are consistently administering boluses or whether they are essentially “running on basal.” Some of the additional data that can be downloaded includes average glucose levels, frequency of glucose monitoring, days between site changes, amount of time patients are suspending the pump or using temporary basal rates, frequency of boluses (which allows to identify non-compliance or insulin stacking behaviors), and average daily carbohydrates consumed (Figure 18).

Figure 17. A patient’s insulin pump download showing comprehensive data for one day including basal rates, boluses and use of bolus calculator, glucose monitoring, carbohydrate intake, and percentage of glucose at target.

Figure 18. A patient’s pump download showing glucose measurements, bolus events, fill events (denoting frequency of site and set changes), as well as insulin pump suspension duration for a 14-day period).

However, despite the multiple benefits of CSII therapy there are also several risks. The first is an abrupt stoppage of insulin delivery either from an occlusion or dislodging of the catheter. For most patients who measure glucose levels at least 4 times daily the problem can be discovered and rectified quickly. However, for the occasional patient who tests infrequently or misses several glucose tests the discontinuation of the insulin infusion can result in ketoacidosis. Fortunately, this is rare. When glucose levels are found to be elevated for no apparent reason, it is appropriate to bolus the appropriate correction dose and if after 1 to 2 hours glucose levels are not improved, an injection of insulin is recommended, and the infusion site should be changed.

 

Another potential complication is infection, often an abscess, at the infusion site. This is also rare and can be minimized with meticulously cleaning the pump site prior to insertion. Although not as severe, inflammation from pump sites can be problematic. This can be improved by changing the infusion set every 24 to 72 hours and rotating pump sites. Similarly, some patients develop lipohypertrophy from infusing the insulin in the same area. This can result in extreme variability in insulin absorption. Again, frequent rotation of pump sites can alleviate this problem which is under-reported. Clinicians should therefore make pump site observation a part of every clinic visit.

 

CLASSIFICATION OF INSULIN PUMPS

 

Insulin pumps can be classified by the way insulin is delivered into:

 

Pumps with Tubing

 

These insulin pumps require an infusion set for insulin delivery. They house an insulin-filled cartridge connected to a tubing with a prespecified length, allowing patients to select the length that better accommodates to their needs.  At the end of the tubing is a needle or soft Teflon cannula that can be inserted into the subcutaneous tissue at a 30- to 45- or 90-degree angle, depending on the type of infusion set used. The abdomen is the preferred infusion site because placement of the catheter there is convenient and comfortable and insulin absorption is most consistent in this region. However, the upper outer quadrant of the buttocks, upper thighs, and triceps fat pad of the arms may also be used.

 

Infusion sets allow removal of the insertion needle, leaving only the soft cannula in place subcutaneously. Patients who experience frequent soft cannula kinking or those with Teflon allergies can opt for infusion sets that use a small stainless-steel needle to infuse insulin instead of a Teflon cannula. Infusion sets have a quick-release mechanism, allowing them to be temporarily disconnected from the insertion site. This quick-release feature makes dressing, swimming, showering, and other activities more convenient.

 

Tubeless Pump

 

Patients may also choose the convenience of a tubeless or patch pump. This pump consists of disposable “pods” which are discarded every three days. The pods are essentially small self-contained insulin pumps with an internal insulin cartridge, an insertion needle and cannula, and the necessary hardware required for insulin administration. Insulin is infused directly from the pod through a catheter without the use of any tubing. Both basal and bolus insulin dosing is communicated to the pod through either audio frequency or Bluetooth technology via a separate “personal diabetes manager” device.

 

Artificial Pancreas Device Systems

 

Improvements in CGM sensor technology have allowed for the integration of CGM systems with insulin pumps and the development of artificial pancreas device systems (APDS), also known as closed-loop (CL) systems. An APDS consists of an insulin pump, a CGM device, and an insulin infusion algorithm designed for safety and glucose control optimization.

 

In 2009, the JDRF developed an artificial pancreas road map defining 6 stages of APDS technology based on the level of automation (61):

  • First generation:
    • Stage 1: Very-Low-Glucose Insulin Off Pump. Pump shuts off when user not responding to low-glucose alarm.
    • Stage 2: Hypoglycemia Minimizer. Predictive hypoglycemia causes alarms, followed by reduction or cessation of insulin delivery before blood glucose gets low.
    • Stage 3: Hypoglycemia/Hyperglycemia Minimizer. Same product as #2 but with added feature allowing insulin dosing above high threshold.
  • Second Generation:
    • Stage 4: Automated Basal/Hybrid Closed Loop. Closed loop at all times with mealtime manual assist bolus.
    • Stage 5: Fully Automated Insulin Closed Loop. Manual mealtime bolus eliminated.
  • Third Generation
    • Stage 6: Fully Automated Multihormone Closed Loop.

 

First generation devices focused primarily on prevention of hypoglycemia. Second generation devices have introduced automation of basal insulin delivery with or without automatic correction boluses. Lastly, third generation devices are expected to fully close the loop while providing a multi-hormonal (e.g., insulin, glucagon, amylin) delivery approach.

 

It is important to note that the development of any of these specific stages is not dependent on the previous one being completed and can occur in tandem.

 

APDS can also be classified according to the type of control algorithm used to determine insulin delivery (62):

 

  • Proportional Integral Derivative (PID). This algorithm responds to measured glucose levels where: “proportional” refers to the difference between the measured sensor glucose and the target glucose; “integral” refers to how long the sensor glucose has been away from the target; and “derivative” refers to how rapidly the sensor glucose is changing.
  • Model Predictive Control (MPC). This algorithm allows prediction of glucose levels at a specific point in the future and based on this data, modulation of insulin delivery.
  • Fuzzy logic. The calculation of insulin doses is similar to what a diabetes specialist would recommend based on CGM data.
  • Bio-inspired. Uses a mathematical model of beta cell insulin production in response to changes in blood glucose.

 

A list of currently approved APDS and features is listed in Figure 19 (63-67)

Figure 19. Features and CGM outcomes from pivotal studies on currently available artificial pancreas device systems.

ADJUNCTIVE NON-INSULIN THERAPIES IN TYPE 1 DIABETES

 

Intensive insulin therapy for T1D is associated with increased risk of hypoglycemia. Additionally, glycemic variability and weight gain with resultant non-adherence to insulin are commonly encountered. Weight gain also contributes to increased cardiometabolic risk such as hypertension, dyslipidemia, and atherosclerotic cardiovascular disease. Insulin therapy also does not address glucagon excess and altered gastric emptying that is seen in patients with T1D. Hence adjunctive therapies could be of potential benefit in management of T1D.

 

Amylin Analog - Pramlintide

 

Amylin is a neuroendocrine hormone co-secreted with insulin by the pancreatic beta cells in a fixed ratio (68); T1D is a state of deficiency. Amylin reduces postprandial hyperglycemia by reducing mealtime glucagon secretion. It also delays gastric emptying, increases satiety and enables weight loss. Overall, amylin complements the action of insulin by targeting postprandial hyperglycemia.

 

Pramlintide is an injectable amylin analog approved for use in T1D as an adjunct to prandial insulin. Pramlintide has similar physiological effects as amylin, such as decreased food intake, and decreases mean A1C by 0.3-0.5% with modest weight loss (69). A recent crossover study of pramlintide infusion co-administered with human regular insulin via a pump over 24h improved glycemic variability and postprandial hyperglycemia in adults with T1D (70). Pramlintide is injected just prior to meals at an initial dose of 15 mcg and increased as tolerated to a final dose of 60 mcg. It should be administered only prior to major meals consisting of 250 calories or 30 grams of carbohydrate. Prandial insulin doses of insulin (in MDI or CSII therapy) should be reduced as food intake decreases and gastric emptying is delayed. For those receiving insulin via a pump, using an “extended bolus” (see above) works best to avoid postprandial hypoglycemia. For those using MDI, some patients administer their insulin just prior to eating (without a lag time) or after eating. Use of pramlintide is limited by nausea, often mild and self-limited. Severe insulin-induced hypoglycemia has also been noted with the use of pramlintide if insulin doses are not sufficiently reduced on initiation of pramlintide therapy.  However widespread use of pramlintide as a therapeutic adjunct in T1D has been limited due to concerns of nausea, hypoglycemia and additional injection burden. Long-term use of pramlintide is unclear at this time.

 

Metformin

 

Metformin, a biguanide, is used as first-line therapy in patients with T2D. It decreases hepatic gluconeogenesis and improves insulin sensitivity (71). Metformin may have some benefit in reducing insulin doses and possibly improve metabolic control in obese/overweight individuals as observed in small studies in patients with T1D. An early meta-analysis of 5 studies suggested that addition of metformin resulted in a decrease in insulin requirement (6.6 units/day), and a decrease in weight with minimal change in A1C (72). A randomized placebo-controlled trial in 140 overweight adolescents with T1D evaluated the addition of metformin to insulin (73). There was no improvement in glycemic control after 6 months but use of metformin resulted in decreased insulin dose and improved measures of adiposity, despite increased gastrointestinal adverse events. A meta-analysis of 19 RCTs suggests short term improvement in A1C that is not sustained after 3 months and associated with higher incidence of GI side effects (74). Although metformin has been shown to decrease CVD morbidity in T2D, data in T1D is lacking.  Recent evidence suggests that metformin decreases insulin resistance and improves vascular health in adolescents with T1D (75). The REMOVAL trial assessed benefit of metformin in T1D and cardiovascular risk and showed no evidence of sustained A1C reduction, and no benefit in carotid intima-media thickness (the study’s primary endpoint); however, reductions in body weight, LDL-C and total insulin requirements was observed (76).  Therefore, based on current evidence, concomitant use of metformin in patients with T1D and is not recommended in current published guidelines.

 

Sodium Glucose Cotransporter 2 (SGLT2) Inhibitors

 

SGLT2 is a protein expressed in the proximal convoluted tubule (PCT) of the kidney and is responsible for re-absorption of filtered glucose.  Inhibition of SGLT2 prevents glucose reabsorption in the PCT and increases glucose excretion by the kidney. SGLT1 is the major intestinal glucose transporter. SGLT1 inhibition also increases postprandial release of the gastrointestinal hormones GLP-1 and polypeptide YY, probably by increasing delivery of glucose to the distal small intestine, thereby regulating glucose and appetite control. Notably, the action of these agents is insulin-independent, therefore this class of drugs has potential as adjunctive therapy for T1D. Additionally recent clinical trials have also demonstrated improvements in cardiovascular outcomes trials as well as reductions in renal outcomes in T2D; therefore, there is significant interest for use in T1D. Early small studies of SGLT2 inhibitors in T1D showed promising results with evidence of decreased total daily insulin dosage, improvement in fasting glucose and A1C, measures of glycemic variability, rates of hypoglycemia and body weight (77-79).

 

Common side effects associated with this class of drugs include genital and urinary infections. Euglycemic diabetic ketoacidosis has been recognized in patients with T1D due to glycosuria masking hyperglycemia but with a catabolic state (due to insulin deficiency and hyperglucagonemia) with ketonemia (80, 81).

 

A dual inhibitor of SGLT1 and 2 sotagliflozin is under development and shows promise in T1D patients (82). Currently in the US, SGLT2 inhibitors are approved for use in T2D only. SGLT2 and mixed SGLT1/2 inhibitors are approved for use in T1D by the European Medicines Agency.

 

All four available SGLT2 inhibitors have been studied in T1D. When added to insulin therapy, all SGLT2 inhibitors appear to decrease A1C levels, averaging 0.35-0,5% within 6 months of initiation; however, this effect does not appear to be sustained at 1 year in clinical trials and effects appear to wane with time (83). Insulin dosing should be adjusted with caution to avoid hypoglycemia. There is no data on efficacy comparing the different agents currently. It is estimated that these agents increase risk of diabetic ketoacidosis by 8-fold, and therefore are not approved for use in T1D in the US.

 

Incretin Therapies

 

Endogenous glucagon-like peptide-1 (GLP-1) is secreted from L cells (present in the small and large intestine) in response to food ingestion. GLP-1 enhances glucose-induced insulin secretion, inhibits glucagon secretion, delays gastric emptying, and induces satiety. GLP-1 secretion in T1D patients is similar to that seen in healthy individuals. In vitro studies suggest that incretin-based therapies can expand beta cell mass, stimulate beta cell proliferation and inhibit beta cell apoptosis, although this has not been demonstrated in humans. Thus, due to their putative effects on beta cell integrity and function, GLP-1 receptor agonists and oral dipeptidyl peptidase-4 (DPP-4) inhibitors are of interest in T1D.  GLP-1 receptor agonists delay gastric emptying, suppress postprandial glucagon secretion, and increase satiety. Studies suggest that these agents may decrease insulin requirements and facilitate weight loss (84, 85). Early RCTs of liraglutide in T1D revealed weight loss and some A1C lowering benefit (85, 86). Recent data suggests benefit of liraglutide 1.8 mg in individuals with T1D and higher BMI in decreasing A1C, weight and no increased hypoglycemia risk (87). However, these effects may not be sustained, based on results from a weekly exenatide study (88). At this time, GLP-1 receptor agonists are not a recommended treatment option in T1D.

 

The DPP-4 enzyme degrades endogenous GLP-1 and removes it from the circulation. DPP-4 inhibitors lower blood glucose by preventing breakdown of endogenous GLP-1, thereby increasing concentration in the circulation. In patients with T2D, DPP-4 inhibitors potentiate glucose-dependent insulin secretion and inhibit glucagon release without effect on gastric emptying or bodyweight. Patients with T1D have inappropriately raised glucagon secretion and DPP-4 inhibitors added to insulin could potentially enhance insulin secretion in patients with residual endogenous insulin secretion and improve glycemic control.  However, observed effects in patients with T1D are limited with modest improvements in A1C that are short-term and not sustained (89). Therefore, these agents cannot be recommended for use in T1D.           

 

Bariatric Surgery

 

Bariatric and other metabolic surgeries are effective weight loss treatments in severe obesity. In T1D individuals with morbid obesity, bariatric surgery has been shown to result in significant weight loss, decrease in insulin requirements and an overall improvement in metabolic profile. However, DKA and hypoglycemia occur in the post-operative period. Longer term and larger studies are required to further evaluate the role of bariatric surgery in T1D (90).

 

OTHER ASPECTS OF MANAGEMENT

 

Psychosocial Aspects

 

Assessment and management of psychosocial issues are an important component of care in individuals with T1D throughout their life span (91). While the individual patient is the focus of care, family support should be encouraged when appropriate. Evaluation and discussion of psychosocial issues and screening for depression screening should be included as part of each clinic visit. Many patients experience “diabetes distress” related to the multitude of self-care responsibilities to optimize glycemic control. Diabetes distress is frequently associated with suboptimal glycemic control, low self-efficacy and reduced self-care. Depression, anxiety from fear of hypoglycemia, and eating disorders can develop and are associated with poor glycemic control. In young adults, comprehensive management of diabetes that addresses these psychosocial issues can improve glycemic control and reduce hospitalization due to diabetic ketoacidosis. Strategic interventions such as cognitive restructuring, goal setting and problem solving can help individuals particularly adolescents and young adults reduce diabetes distress (92). Thus, early identification and treatment including referral to a mental health specialist can help aid management of diabetes.

 

Management in Exercise

 

The benefits of exercise and physical activity in patients with type 1 diabetes have been well documented (93, 94). However, achieving adequate glycemic control during and after completion of exercise remains a rather challenging aspect of type 1 diabetes management. Glycemia at the initiation of exercise, sensor glucose trend (if using a CGM), timing from the previous meal, carbohydrate content in the meal preceding exercise, type and duration of exercise, are all but a few of the factors that need to be considered to ensure that glycemic control remains stable during and after cessation of exercise.

 

In 2017, an international consensus statement for exercise management in type 1 diabetes was published (95). This consensus is a unique resource which provides detailed glucose management strategies. Recommended adjustments to basal and prandial insulin, for both insulin pump and multiple daily insulin injection users, as well as carbohydrate intake requirements depending on the intensity and duration of activity are clearly presented. Quite importantly, the consensus also covers factors that would preclude exercise including the presence of elevated ketones, recent hypoglycemia, and diabetes-related complications which may be exacerbated in the context of vigorous exercise and/or competitive endurance events. We encourage the reader to refer to this publication for additional guidance.

 

For those patients on hybrid closed loop systems, a way to minimize the occurrence of exercise-induced hypoglycemia is the use of a higher glucose target for exercise. For the Medtronic 670G, the standard Auto-Mode target is 120 mg/dL which can be temporarily changed to 150 mg/dL. For the Tandem X2 with Control IQ, the standard target for regular activity is between 112.5 and 160 mg/dL and can be temporarily changed to 140-160 mg/dL.

 

A study in open loop insulin pump users found that a basal rate reduction starting 90 min before exercise was superior to pump suspension at exercise onset for reduction of hypoglycemia risk during exercise and did not compromise the post-exercise meal glycemic control (96).

 

Another strategy that may be more effective than basal rate reduction for prevention of exercise induced hypoglycemia is the use of a subcutaneously administered mini-dose of glucagon. A small study including 15 subjects with type 1 diabetes on insulin pump therapy who exercised in the fasting state in the morning for 45 min, found that a dose of 150 µg of subcutaneous glucagon, compared to a 50% basal insulin reduction or 40-g oral glucose tablets, resulted in no hypoglycemia (vs. basal insulin reduction) and no hyperglycemia (vs. oral glucose tablets) (97). However, larger and long-term studies are required before determining if a mini-dose of glucagon is safe and effective for prevention of exercise induced hypoglycemia in subjects with type 1 diabetes. 

 

Management of Special Populations

 

OLDER ADULTS

 

Adults with T1D now span a very large age spectrum—from 18 to 100 years of age and beyond. These individuals are unique in that they usually have lived with a complex disease for many years (91).  An understanding of each individual’s circumstances is vital and management often requires assessment of medical, functional, mental, and social domains. The ADA emphasizes that glycemic targets should be individualized with the goal of achieving the best possible control while minimizing the risk of severe hyperglycemia and hypoglycemia (98).

 

Glycemic goals in older adults vary. Most older adults with T1D have long-standing disease (unlike individuals with T2DM where diabetes can be long-standing or new onset). Additionally, there is a wide range of health in older individuals, with some patients enjoying good functional status and no comorbid conditions, while others are limited by multiple comorbidities as well as physical or cognitive impairments. Older T1D patients may develop diabetes related complications which pose a challenge in disease management. Insulin dosing errors, hypoglycemia unawareness, and inability to manage hypoglycemia when it occurs may result from physical and cognitive decline. Special attention should be focused on meal planning and physical activities in this population.  Severe hyperglycemia can lead to dehydration and hyperglycemic crises (91). Issues related to self-care capacity, mobility, and autonomy should be promptly addressed.

 

Thus, treatment goals should be reassessed and individualized based on patient factors. Older patients with long life expectancy and little comorbidity should have treatment targets similar to those of middle-aged or younger adults. In patients with multiple comorbid conditions, treatment targets may be relaxed, while avoiding symptomatic hyperglycemia or the risk of diabetic ketoacidosis (91). Therefore, it is important to assess the clinical needs of the patient, setting specific goals and expectations that may differ quite significantly between a healthy 24-year-old and a frail 82-year-old with retinopathy and cardiovascular disease.

 

There are few long-term studies in older adults demonstrating the benefits of intensive glycemic, blood pressure, and lipid control (98). As with younger adults, glycemic control should be assessed based on frequent SMBG levels (and CGM data, if available) as well as A1C to help direct changes in therapy. More stringent A1C goals (~6.5-7%) can be recommended in select older adults if this can be achieved without hypoglycemia or other adverse effects. This is appropriate for older individuals with anticipated long-life expectancy, hypoglycemia awareness and no CVD. Less stringent A1C goals (for example A1C<8.5%) may be appropriate for patients with a history of severe hypoglycemia, hypoglycemia unawareness, limited life expectancy, advanced microvascular/macrovascular complications, or extensive comorbid conditions (91, 99).  

 

INPATIENT MANAGEMENT AND OUTPATIENT PROCEDURES

 

The challenges involved in management of individuals with T1D in the hospital and in preparation for scheduled outpatient procedures include difficulties associated with fasting, maintaining a consistent source of carbohydrate, and facilitating inpatient blood glucose management while modifying scheduled insulin therapy. Individuals with T1D may have difficulty fasting for long periods of time (more than 10 h) prior to a procedure. Patients with T1D should be prepared with a treatment plan for insulin dose adjustments and oral glucose intake prior to any procedure that requires alterations in dietary intake and/or fasting.

 

In general, goals for blood glucose levels in individuals with T1D are the same as for people with T2D or hospital-related hyperglycemia (100) . It is imperative that the entire health care team, including anesthesiologists and surgeons as well as other specialists who perform procedures, understands T1D and how it factors into the comprehensive delivery of care. First, the diagnosis of T1D should be clearly identified in the patient’s record.  Second, the awareness that people with T1D will be at high risk for hypoglycemia during prolonged fasting and are at risk for ketosis if insulin is inappropriately withheld. Under anesthesia, individuals with T1D must be carefully monitored for hypoglycemia and hyperglycemia. Third, a plan for preventing and treating hypoglycemia should be established for each patient.

 

SMBG should be ordered to fit the patient’s usual insulin regimen with modifications as needed based on clinical status. Self-management in the hospital may be appropriate for some individuals with T1D including those who successfully manage their disease at home, have cognitive skills to perform necessary tasks such as administer insulin and perform SMBG, count carbohydrates and have a good understanding of their condition (100). For some individuals, once the most acute phase of an illness has resolved or improved, patients may be able to self-administer their prior multiple-dose or CSII insulin regimen under the guidance of hospital personnel who are knowledgeable in glycemic management.  Individuals managed with insulin pumps and/or multiple-dose regimens with carbohydrate counting and correction dosing may be allowed to manage their own diabetes if this is what they desire, once they are capable of doing so.

 

The need for uninterrupted basal insulin to prevent hyperglycemia and ketoacidosis is important to recognize. Insulin dosing adjustments should also be made in the perioperative period and inpatient setting with consideration of oral intake and blood glucose trends.

 

The use of CGM in the inpatient setting is an area of ongoing research. Currently, the Endocrine Society recommends against the use of real-time CGM (RT-CGM) alone in the intensive care unit or operating room settings due to limited available data on accuracy (101). A study in T2D patients on basal bolus insulin therapy admitted to the general ward evaluated the use of retrospective CGM versus point of care capillary glucose testing for inpatient glycemic control (102). Although average daily glucose levels were comparable between CGM and capillary blood glucose testing, CGM detected a higher number of hypoglycemic episodes (55 vs 12, P < 0.01) suggesting that CGM may be beneficial for identification of hypoglycemia in the general ward particularly in patients with hypoglycemia unawareness. We feel it is reasonable to allow T1D patients who already benefit from use of RT-CGM to continue the use of this technology in the non-ICU inpatient setting under the supervision of the care team. Large prospective randomized trials will be required to establish benefit or lack thereof of RT-CGM use on inpatient glycemic control.

 

BETA-CELL REPLACEMENT STRATEGIES

 

Pancreas Transplantation

 

Pancreas transplantation is a currently available therapeutic option for patients with diabetes who meet specific clinical criteria. Patients with end-stage renal disease are eligible to undergo simultaneous pancreas kidney (SPK) transplantation. Also, pancreas transplantation may be offered as a separate procedure after a patient has already received a kidney transplant (pancreas after kidney (PAK)). In addition, solitary pancreas transplantation may also be offered to those individuals presenting with severe metabolic complications attributed to poor glycemic control (pancreas transplant alone (PTA)). Pancreas transplantation procedures have been performed since the 1960’s. A 2011 update on Pancreas Transplantation from the International Pancreas Transplant Registry reported improvements in patient survival and graft function over a course of 24 years of pancreas transplantation (103). These improved outcomes were related to changes in surgical technique and immunosuppressive regimens as well as tighter donor selection criteria. At 5-years post-transplantation, pancreas graft survival is now reported at ~70% for SPK and at ~ 50% for PAK and PTA. Further, patient survival at 10 years exceeds 70% with the highest survival rate observed in PTA recipients (82%).

 

Islet Transplantation

 

Islet transplantation provides a less invasive surgical alternative for beta-cell replacement in patients with labile diabetes and has the potential to restore normoglycemia, eliminate severe hypoglycemia and restore hypoglycemia awareness. However, this procedure is still considered experimental in the United States. Marked improvements have also been noted in the field of islet transplantation over the past decade which have led to insulin independence rates at 5 years being comparable to pancreas transplantation outcomes (104). A pivotal study of islet transplantation in patients with T1D showed that at 1-year post transplant, 87% of study participants achieved the primary endpoint of a A1C <7.0% and freedom from severe hypoglycemia (from day 28 to 365) (105). Further details about islet transplantation can be found in the Endotext chapter on this topic.

 

FUTURE DIRECTIONS IN MANAGEMENT OF TYPE 1 DIABETES

 

Artificial Pancreas Device Systems - Closed Loop Systems

 

In addition to insulin-only CL-systems, bi-hormonal closed loop systems are also being actively explored. Additional manufacturers utilizing insulin-only CL-systems are expected to launch their devices in the near future. The introduction of faster-acting insulins (biochaperone lispro and faster-acting insulin aspart (FIAsp)) could potentially make these strategies more effective. As this technology advances, we are getting closer to the goal of a fully automated device which will be able to predict with high accuracy changes in glucose profiles and respond accordingly with stringent modulation of infusion of hormones (e.g., insulin, glucagon, amylin) to maintain glycemia within normal ranges.   

 

Implantation of Encapsulated Islets

 

Some of the limitations of islet transplantation currently include the limited availability of donors and the need for long term immunosuppression to prevent rejection of the transplanted graft. Protecting the islets from the immunologic environment may allow both the use of non-human islets for transplantation and minimize or eliminate the need for systemic immunosuppression. Thus, the encapsulation of islets to attain these goals has been sought for several years but unfortunately this technology is still not at the stage to make it to the clinical arena. Although initial attempts at encapsulation of islets resulted in damage of the capsule by local tissue responses, newer techniques allowing for conformal coating of human islets have shown promising results in pre-clinical models and are currently being explored (106).

 

Islet Xenotransplantation

 

An alternative to human pancreas and islet transplantation which is currently being explored is the use of pig islets. Pig islets have major physiologic similarities to human islets. Notably, pig insulin differs from human insulin by only one amino acid. Donor pigs may be genetically engineered to be protected from the human immune system thus reducing the need for potent immunosuppression. Studies in non-human primates using encapsulated pig islets have resulted in graft survival for more than 6 months (107). Research in this field in actively ongoing.

Stem Cell Based Therapies

 

Stem cell research has allowed the generation of insulin-producing pancreatic β-cells from human pluripotent stem cells (108). Further, scientists can now also generate alpha and delta cells from stem cells therefore more closely mimicking a fully functional human islet. This technology has the potential to generate vast amounts of glucose-responsive β-cells and allow for the development of customizable islets containing predetermined amounts of specific cell lines. Results in preclinical models are encouraging and a clinical trial is expected in 2021.

Glucose Responsive Insulins (Smart Insulins)

 

Another area of ongoing research is the development of “smart” drug delivery systems able to respond to environmental or external triggers greatly improving therapeutic performance. Conceptually, “smart” insulins should be able to respond to changes in ambient glucose which would dictate activation or cessation of insulin delivery.  Several efforts have been made to generate glucose-responsive insulin delivery systems and some have shown promising results in pre-clinical studies including the utilization of enzymatic triggers, glucose-binding proteins, and synthetic molecules able to bind to glucose. However, current limitations include the potential for immunogenicity and poor glucose selectivity (109). Continued progress in this field in the coming years to reduce the burden of diabetes is anticipated. 

 

CONCLUSIONS

 

No disease has had such an evolution of therapy in the past 100 years as T1D. From certain death to the discovery of insulin, from impure animal insulin preparations to purified human insulins, from once daily long-acting insulin to CSII, from urine glucose testing to real-time continuous glucose sensors and closed loop insulin pumps, treatments continue to emerge that improve the lives of people with T1D. Our current challenges remain teaching the providers how to best use these new tools, directing our medical systems to allow us to best utilize these therapies, and perhaps most importantly, transferring diabetes technologies to the patients who can best apply them. Although the future is exciting, we need to continually master the use of our current tools before we can successfully move forward. Hopefully, soon the successful management of T1D will become a reality for all with this disease.

 

REFERENCES

 

  1. Rosenfeld, L., Insulin: discovery and controversy. Clin Chem, 2002. 48(12): p. 2270-88.
  2. Hirsch, I.B., et al., The Evolution of Insulin and How it Informs Therapy and Treatment Choices. Endocr Rev, 2020. 41(5).
  3. Lauritzen, T., O.K. Faber, and C. Binder, Variation in 125I-insulin absorption and blood glucose concentration. Diabetologia, 1979. 17(5): p. 291-5.
  4. 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. 977-86.
  5. 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. 2643-53.
  6. Martin, C.L., et al., Neuropathy among the diabetes control and complications trial cohort 8 years after trial completion. Diabetes Care, 2006. 29(2): p. 340-4.
  7. Retinopathy and nephropathy in patients with type 1 diabetes four years after a trial of intensive therapy. The Diabetes Control and Complications Trial/Epidemiology of Diabetes Interventions and Complications Research Group. N Engl J Med, 2000. 342(6): p. 381-9.
  8. Writing Team for the Diabetes, C., I. Complications Trial/Epidemiology of Diabetes, and G. Complications Research, Sustained effect of intensive treatment of type 1 diabetes mellitus on development and progression of diabetic nephropathy: the Epidemiology of Diabetes Interventions and Complications (EDIC) study. JAMA, 2003. 290(16): p. 2159-67.
  9. Genuth, S., Insights from the diabetes control and complications trial/epidemiology of diabetes interventions and complications study on the use of intensive glycemic treatment to reduce the risk of complications of type 1 diabetes. Endocr Pract, 2006. 12 Suppl 1: p. 34-41.
  10. White, N.H., et al., Prolonged effect of intensive therapy on the risk of retinopathy complications in patients with type 1 diabetes mellitus: 10 years after the Diabetes Control and Complications Trial. Arch Ophthalmol, 2008. 126(12): p. 1707-15.
  11. Beck, R.W., et al., The T1D Exchange clinic registry. J Clin Endocrinol Metab, 2012. 97(12): p. 4383-9.
  12. Beck, R.W., K.M. Miller, and N.C. Foster, The T1D Exchange Clinic Network and Registry: 10 Years of Enlightenment on the State of Type 1 Diabetes in the United States. Diabetes Technol Ther, 2019. 21(6): p. 310-312.
  13. Foster, N.C., et al., State of Type 1 Diabetes Management and Outcomes from the T1D Exchange in 2016-2018. Diabetes Technol Ther, 2019. 21(2): p. 66-72.
  14. 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. 971-8.
  15. Tonyushkina, K. and J.H. Nichols, Glucose meters: a review of technical challenges to obtaining accurate results. J Diabetes Sci Technol, 2009. 3(4): p. 971-80.
  16. Miller, K.M., et al., Evidence of a strong association between frequency of self-monitoring of blood glucose and hemoglobin A1c levels in T1D exchange clinic registry participants. Diabetes Care, 2013. 36(7): p. 2009-14.
  17. American Diabetes, A., 6. Glycemic Targets: Standards of Medical Care in Diabetes-2021. Diabetes Care, 2021. 44(Suppl 1): p. S73-S84.
  18. Freckmann, G., et al., System accuracy evaluation of 43 blood glucose monitoring systems for self-monitoring of blood glucose according to DIN EN ISO 15197. J Diabetes Sci Technol, 2012. 6(5): p. 1060-75.
  19. Bergenstal, R.M., et al., Recommendations for standardizing glucose reporting and analysis to optimize clinical decision making in diabetes: the Ambulatory Glucose Profile (AGP). Diabetes Technol Ther, 2013. 15(3): p. 198-211.
  20. Basu, A., et al., Time lag of glucose from intravascular to interstitial compartment in humans. Diabetes, 2013. 62(12): p. 4083-7.
  21. Castle, J.R. and P.G. Jacobs, Nonadjunctive Use of Continuous Glucose Monitoring for Diabetes Treatment Decisions. J Diabetes Sci Technol, 2016. 10(5): p. 1169-73.
  22. Bailey, T., et al., The Performance and Usability of a Factory-Calibrated Flash Glucose Monitoring System. Diabetes Technol Ther, 2015. 17(11): p. 787-94.
  23. New, J.P., et al., Continuous glucose monitoring in people with diabetes: the randomized controlled Glucose Level Awareness in Diabetes Study (GLADIS). Diabet Med, 2015. 32(5): p. 609-17.
  24. Shivers, J.P., et al., "Turn it off!": diabetes device alarm fatigue considerations for the present and the future. J Diabetes Sci Technol, 2013. 7(3): p. 789-94.
  25. Battelino, T., et al., Clinical Targets for Continuous Glucose Monitoring Data Interpretation: Recommendations From the International Consensus on Time in Range. Diabetes Care, 2019. 42(8): p. 1593-1603.
  26. Bergenstal, R.M., et al., Recommendations for standardizing glucose reporting and analysis to optimize clinical decision making in diabetes: the ambulatory glucose profile. J Diabetes Sci Technol, 2013. 7(2): p. 562-78.
  27. Yeh, H.C., et al., Comparative effectiveness and safety of methods of insulin delivery and glucose monitoring for diabetes mellitus: a systematic review and meta-analysis. Ann Intern Med, 2012. 157(5): p. 336-47.
  28. Juvenile Diabetes Research Foundation Continuous Glucose Monitoring Study, G., et al., The effect of continuous glucose monitoring in well-controlled type 1 diabetes. Diabetes Care, 2009. 32(8): p. 1378-83.
  29. Foster, N.C., et al., Continuous Glucose Monitoring in Patients With Type 1 Diabetes Using Insulin Injections. Diabetes Care, 2016. 39(6): p. e81-2.
  30. Bailey, T.S., et al., American Association of Clinical Endocrinologists and American College of Endocrinology 2016 Outpatient Glucose Monitoring Consensus Statement. Endocr Pract, 2016. 22(2): p. 231-61.
  31. Peters, A.L., et al., Diabetes Technology-Continuous Subcutaneous Insulin Infusion Therapy and Continuous Glucose Monitoring in Adults: An Endocrine Society Clinical Practice Guideline. J Clin Endocrinol Metab, 2016: p. jc20162534.
  32. Sacks, D.B., et al., Position statement executive summary: guidelines and recommendations for laboratory analysis in the diagnosis and management of diabetes mellitus. Diabetes Care, 2011. 34(6): p. 1419-23.
  33. Rubinow, K.B. and I.B. Hirsch, Reexamining metrics for glucose control. JAMA, 2011. 305(11): p. 1132-3.
  34. American Diabetes, A., 10. Cardiovascular Disease and Risk Management: Standards of Medical Care in Diabetes-2021. Diabetes Care, 2021. 44(Suppl 1): p. S125-S150.
  35. Shah, V.N., et al., Risk Factors for Cardiovascular Disease (CVD) in Adults with Type 1 Diabetes: Findings from Prospective Real-life T1D Exchange Registry. J Clin Endocrinol Metab, 2020. 105(5).
  36. Schofield, J., J. Ho, and H. Soran, Cardiovascular Risk in Type 1 Diabetes Mellitus. Diabetes Ther, 2019. 10(3): p. 773-789.
  37. Vistisen, D., et al., Prediction of First Cardiovascular Disease Event in Type 1 Diabetes Mellitus: The Steno Type 1 Risk Engine. Circulation, 2016. 133(11): p. 1058-66.
  38. Sousa, G.R., et al., Glycemic Control, Cardiac Autoimmunity, and Long-Term Risk of Cardiovascular Disease in Type 1 Diabetes Mellitus. Circulation, 2019. 139(6): p. 730-743.
  39. Howey, D.C., et al., (Lys(B28), Pro(B29))-human insulin. A rapidly absorbed analogue of human insulin. Diabetes, 1994. 43(3): p. 396-402.
  40. Mudaliar, S.R., et al., Insulin aspart (B28 asp-insulin): a fast-acting analog of human insulin: absorption kinetics and action profile compared with regular human insulin in healthy nondiabetic subjects. Diabetes Care, 1999. 22(9): p. 1501-6.
  41. Garg, S.K., S.L. Ellis, and H. Ulrich, Insulin glulisine: a new rapid-acting insulin analogue for the treatment of diabetes. Expert Opin Pharmacother, 2005. 6(4): p. 643-51.
  42. Davis, A., J. Kuriakose, and J.N. Clements, Faster Insulin Aspart: A New Bolus Option for Diabetes Mellitus. Clin Pharmacokinet, 2019. 58(4): p. 421-430.
  43. Linnebjerg, H., et al., Pharmacokinetics and Glucodynamics of Ultra Rapid Lispro (URLi) versus Humalog((R)) (Lispro) in Younger Adults and Elderly Patients with Type 1 Diabetes Mellitus: A Randomised Controlled Trial. Clin Pharmacokinet, 2020. 59(12): p. 1589-1599.
  44. Blevins, T., et al., Randomized Double-Blind Clinical Trial Comparing Ultra Rapid Lispro With Lispro in a Basal-Bolus Regimen in Patients With Type 2 Diabetes: PRONTO-T2D. Diabetes Care, 2020. 43(12): p. 2991-2998.
  45. Klaff, L., et al., Ultra rapid lispro improves postprandial glucose control compared with lispro in patients with type 1 diabetes: Results from the 26-week PRONTO-T1D study. Diabetes Obes Metab, 2020. 22(10): p. 1799-1807.
  46. Bode, B.W., et al., Inhaled Technosphere Insulin Compared With Injected Prandial Insulin in Type 1 Diabetes: A Randomized 24-Week Trial. Diabetes Care, 2015. 38(12): p. 2266-73.
  47. Borgono, C.A. and B. Zinman, Insulins: past, present, and future. Endocrinol Metab Clin North Am, 2012. 41(1): p. 1-24.
  48. Hirsch, I.B., Insulin analogues. N Engl J Med, 2005. 352(2): p. 174-83.
  49. Switzer, S.M., et al., Intensive insulin therapy in patients with type 1 diabetes mellitus. Endocrinol Metab Clin North Am, 2012. 41(1): p. 89-104.
  50. Ratner, R.E., et al., Less hypoglycemia with insulin glargine in intensive insulin therapy for type 1 diabetes. U.S. Study Group of Insulin Glargine in Type 1 Diabetes. Diabetes Care, 2000. 23(5): p. 639-43.
  51. Havelund, S., et al., The mechanism of protraction of insulin detemir, a long-acting, acylated analog of human insulin. Pharm Res, 2004. 21(8): p. 1498-504.
  52. Heller, S., C. Koenen, and B. Bode, Comparison of insulin detemir and insulin glargine in a basal-bolus regimen, with insulin aspart as the mealtime insulin, in patients with type 1 diabetes: a 52-week, multinational, randomized, open-label, parallel-group, treat-to-target noninferiority trial. Clin Ther, 2009. 31(10): p. 2086-97.
  53. Keating, G.M., Insulin degludec and insulin degludec/insulin aspart: a review of their use in the management of diabetes mellitus. Drugs, 2013. 73(6): p. 575-93.
  54. Mathieu, C., et al., Efficacy and safety of insulin degludec in a flexible dosing regimen vs insulin glargine in patients with type 1 diabetes (BEGIN: Flex T1): a 26-week randomized, treat-to-target trial with a 26-week extension. J Clin Endocrinol Metab, 2013. 98(3): p. 1154-62.
  55. Russell-Jones, D., et al., Insulin degludec results in lower rates of nocturnal hypoglycaemia and fasting plasma glucose vs. insulin glargine: A meta-analysis of seven clinical trials. Nutr Metab Cardiovasc Dis, 2015. 25(10): p. 898-905.
  56. Meneghini, L.F., Insulin therapy for type 2 diabetes. Endocrine, 2013. 43(3): p. 529-34.
  57. Dailey, G. and F. Lavernia, A review of the safety and efficacy data for insulin glargine 300 units/ml, a new formulation of insulin glargine. Diabetes Obes Metab, 2015. 17(12): p. 1107-14.
  58. Home, P.D., et al., New Insulin Glargine 300 Units/mL Versus Glargine 100 Units/mL in People With Type 1 Diabetes: A Randomized, Phase 3a, Open-Label Clinical Trial (EDITION 4). Diabetes Care, 2015. 38(12): p. 2217-25.
  59. Pettus, J., et al., The past, present, and future of basal insulins. Diabetes Metab Res Rev, 2016. 32(6): p. 478-96.
  60. Hirsch, I.B., et al., Continuous subcutaneous insulin infusion (CSII) of insulin aspart versus multiple daily injection of insulin aspart/insulin glargine in type 1 diabetic patients previously treated with CSII. Diabetes Care, 2005. 28(3): p. 533-8.
  61. Kowalski, A.J., Can we really close the loop and how soon? Accelerating the availability of an artificial pancreas: a roadmap to better diabetes outcomes. Diabetes Technol Ther, 2009. 11 Suppl 1: p. S113-9.
  62. Trevitt, S., S. Simpson, and A. Wood, Artificial Pancreas Device Systems for the Closed-Loop Control of Type 1 Diabetes: What Systems Are in Development? J Diabetes Sci Technol, 2016. 10(3): p. 714-23.
  63. Bergenstal, R.M., et al., Safety of a Hybrid Closed-Loop Insulin Delivery System in Patients With Type 1 Diabetes. JAMA, 2016. 316(13): p. 1407-1408.
  64. Breton, M.D., et al., A Randomized Trial of Closed-Loop Control in Children with Type 1 Diabetes. N Engl J Med, 2020. 383(9): p. 836-845.
  65. Brown, S.A., et al., Six-Month Randomized, Multicenter Trial of Closed-Loop Control in Type 1 Diabetes. N Engl J Med, 2019. 381(18): p. 1707-1717.
  66. Forlenza, G.P., et al., Safety Evaluation of the MiniMed 670G System in Children 7-13 Years of Age with Type 1 Diabetes. Diabetes Technol Ther, 2019. 21(1): p. 11-19.
  67. Garg, S.K., et al., Glucose Outcomes with the In-Home Use of a Hybrid Closed-Loop Insulin Delivery System in Adolescents and Adults with Type 1 Diabetes. Diabetes Technol Ther, 2017. 19(3): p. 155-163.
  68. Lebovitz, H.E., Adjunct therapy for type 1 diabetes mellitus. Nat Rev Endocrinol, 2010. 6(6): p. 326-34.
  69. Edelman, S., et al., A double-blind, placebo-controlled trial assessing pramlintide treatment in the setting of intensive insulin therapy in type 1 diabetes. Diabetes Care, 2006. 29(10): p. 2189-95.
  70. Riddle, M.C., et al., Control of Postprandial Hyperglycemia in Type 1 Diabetes by 24-Hour Fixed-Dose Coadministration of Pramlintide and Regular Human Insulin: A Randomized, Two-Way Crossover Study. Diabetes Care, 2018. 41(11): p. 2346-2352.
  71. Schimmack, G., R.A. Defronzo, and N. Musi, AMP-activated protein kinase: Role in metabolism and therapeutic implications. Diabetes Obes Metab, 2006. 8(6): p. 591-602.
  72. Vella, S., et al., The use of metformin in type 1 diabetes: a systematic review of efficacy. Diabetologia, 2010. 53(5): p. 809-20.
  73. Libman, I.M., et al., Effect of Metformin Added to Insulin on Glycemic Control Among Overweight/Obese Adolescents With Type 1 Diabetes: A Randomized Clinical Trial. JAMA, 2015. 314(21): p. 2241-50.
  74. Liu, Y.S., et al., Vascular and metabolic effects of metformin added to insulin therapy in patients with type 1 diabetes: A systematic review and meta-analysis. Diabetes Metab Res Rev, 2020. 36(6): p. e3334.
  75. Bjornstad, P., et al., Metformin Improves Insulin Sensitivity and Vascular Health in Youth With Type 1 Diabetes Mellitus. Circulation, 2018. 138(25): p. 2895-2907.
  76. Petrie, J.R., et al., Cardiovascular and metabolic effects of metformin in patients with type 1 diabetes (REMOVAL): a double-blind, randomised, placebo-controlled trial. Lancet Diabetes Endocrinol, 2017. 5(8): p. 597-609.
  77. Henry, R.R., et al., Efficacy and Safety of Canagliflozin, a Sodium-Glucose Cotransporter 2 Inhibitor, as Add-on to Insulin in Patients With Type 1 Diabetes. Diabetes Care, 2015. 38(12): p. 2258-65.
  78. Perkins, B.A., et al., Sodium-glucose cotransporter 2 inhibition and glycemic control in type 1 diabetes: results of an 8-week open-label proof-of-concept trial. Diabetes Care, 2014. 37(5): p. 1480-3.
  79. Pieber, T.R., et al., Empagliflozin as adjunct to insulin in patients with type 1 diabetes: a 4-week, randomized, placebo-controlled trial (EASE-1). Diabetes Obes Metab, 2015. 17(10): p. 928-35.
  80. Bode, B.W. and S.K. Garg, The Emerging Role of Adjunctive Noninsulin Antihyperglycemic Therapy in the Management of Type 1 Diabetes. Endocr Pract, 2016. 22(2): p. 220-30.
  81. Janssens, B., S. Caerels, and C. Mathieu, SGLT inhibitors in type 1 diabetes: weighing efficacy and side effects. Ther Adv Endocrinol Metab, 2020. 11: p. 2042018820938545.
  82. Sands, A.T., et al., Sotagliflozin, a Dual SGLT1 and SGLT2 Inhibitor, as Adjunct Therapy to Insulin in Type 1 Diabetes. Diabetes Care, 2015. 38(7): p. 1181-8.
  83. Taylor, S.I., et al., SGLT2 inhibitors as adjunctive therapy for type 1 diabetes: balancing benefits and risks. Lancet Diabetes Endocrinol, 2019. 7(12): p. 949-958.
  84. Dejgaard, T.F., et al., Efficacy and safety of liraglutide for overweight adult patients with type 1 diabetes and insufficient glycaemic control (Lira-1): a randomised, double-blind, placebo-controlled trial. Lancet Diabetes Endocrinol, 2016. 4(3): p. 221-32.
  85. Ahren, B., et al., Efficacy and Safety of Liraglutide Added to Capped Insulin Treatment in Subjects With Type 1 Diabetes: The ADJUNCT TWO Randomized Trial. Diabetes Care, 2016. 39(10): p. 1693-701.
  86. Mathieu, C., et al., Efficacy and Safety of Liraglutide Added to Insulin Treatment in Type 1 Diabetes: The ADJUNCT ONE Treat-To-Target Randomized Trial. Diabetes Care, 2016. 39(10): p. 1702-10.
  87. Dejgaard, T.F., et al., Liraglutide reduces hyperglycaemia and body weight in overweight, dysregulated insulin-pump-treated patients with type 1 diabetes: The Lira Pump trial-a randomized, double-blinded, placebo-controlled trial. Diabetes Obes Metab, 2020. 22(4): p. 492-500.
  88. Herold, K.C., et al., Exenatide extended release in patients with type 1 diabetes with and without residual insulin production. Diabetes Obes Metab, 2020. 22(11): p. 2045-2054.
  89. Ellis, S.L., et al., Effect of sitagliptin on glucose control in adult patients with Type 1 diabetes: a pilot, double-blind, randomized, crossover trial. Diabet Med, 2011. 28(10): p. 1176-81.
  90. Kirwan, J.P., et al., Bariatric Surgery in Obese Patients With Type 1 Diabetes. Diabetes Care, 2016. 39(6): p. 941-8.
  91. Chiang, J.L., et al., Type 1 diabetes through the life span: a position statement of the American Diabetes Association. Diabetes Care, 2014. 37(7): p. 2034-54.
  92. Hagger, V., et al., Diabetes Distress Among Adolescents with Type 1 Diabetes: a Systematic Review. Curr Diab Rep, 2016. 16(1): p. 9.
  93. Chimen, M., et al., What are the health benefits of physical activity in type 1 diabetes mellitus? A literature review. Diabetologia, 2012. 55(3): p. 542-51.
  94. Sluik, D., et al., Physical Activity and Mortality in Individuals With Diabetes Mellitus: A Prospective Study and Meta-analysis. Arch Intern Med, 2012. 172(17): p. 1285-95.
  95. Riddell, M.C., et al., Exercise management in type 1 diabetes: a consensus statement. Lancet Diabetes Endocrinol, 2017. 5(5): p. 377-390.
  96. Zaharieva, D.P., et al., Improved Open-Loop Glucose Control With Basal Insulin Reduction 90 Minutes Before Aerobic Exercise in Patients With Type 1 Diabetes on Continuous Subcutaneous Insulin Infusion. Diabetes Care, 2019. 42(5): p. 824-831.
  97. Rickels, M.R., et al., Mini-Dose Glucagon as a Novel Approach to Prevent Exercise-Induced Hypoglycemia in Type 1 Diabetes. Diabetes Care, 2018. 41(9): p. 1909-1916.
  98. American Diabetes, A., 12. Older Adults: Standards of Medical Care in Diabetes-2021. Diabetes Care, 2021. 44(Suppl 1): p. S168-S179.
  99. Kirkman, M.S., et al., Diabetes in older adults. Diabetes Care, 2012. 35(12): p. 2650-64.
  100. American Diabetes, A., 15. Diabetes Care in the Hospital: Standards of Medical Care in Diabetes-2021. Diabetes Care, 2021. 44(Suppl 1): p. S211-S220.
  101. Gomez, A.M. and G.E. Umpierrez, Continuous glucose monitoring in insulin-treated patients in non-ICU settings. J Diabetes Sci Technol, 2014. 8(5): p. 930-6.
  102. Gomez, A.M., et al., Continuous Glucose Monitoring Versus Capillary Point-of-Care Testing for Inpatient Glycemic Control in Type 2 Diabetes Patients Hospitalized in the General Ward and Treated With a Basal Bolus Insulin Regimen. J Diabetes Sci Technol, 2016. 10(2): p. 325-9.
  103. Gruessner, A.C., 2011 update on pancreas transplantation: comprehensive trend analysis of 25,000 cases followed up over the course of twenty-four years at the International Pancreas Transplant Registry (IPTR). Rev Diabet Stud, 2011. 8(1): p. 6-16.
  104. Bellin, M.D., et al., Potent induction immunotherapy promotes long-term insulin independence after islet transplantation in type 1 diabetes. Am J Transplant, 2012. 12(6): p. 1576-83.
  105. Hering, B.J., et al., Phase 3 Trial of Transplantation of Human Islets in Type 1 Diabetes Complicated by Severe Hypoglycemia. Diabetes Care, 2016. 39(7): p. 1230-40.
  106. Tomei, A.A., et al., Device design and materials optimization of conformal coating for islets of Langerhans. Proc Natl Acad Sci U S A, 2014. 111(29): p. 10514-9.
  107. Park, C.G., R. Bottino, and W.J. Hawthorne, Current status of islet xenotransplantation. Int J Surg, 2015. 23(Pt B): p. 261-6.
  108. Pagliuca, F.W., et al., Generation of functional human pancreatic beta cells in vitro. Cell, 2014. 159(2): p. 428-39.
  109. Webber, M.J. and D.G. Anderson, Smart approaches to glucose-responsive drug delivery. J Drug Target, 2015. 23(7-8): p. 651-5.

Endocrinology of The Gut and the Regulation of Body Weight and Metabolism

ABSTRACT

 

Obesity prevalence continues to increase globally, leading to ill-health and reduced life expectancy in those affected and an urgent need for effective preventative and therapeutic strategies. Until recently obesity was viewed simplistically as an imbalance between energy expenditure and consumption that could be easily corrected by lifestyle changes. However, obesity is now recognized to be a chronic progressive disease, with bodyweight controlled by a complex interplay between the central nervous system, peripheral signals of energy balance from adipose tissue and the gastrointestinal tract, environmental food cues, and a powerful biological drive to defend the highest weight achieved. Currently, bariatric surgery represents the most effective treatment for people with severe obesity, leading to marked sustained weight loss as a consequence of altered eating behavior with improved health and life expectancy. Bariatric surgical procedures were initially envisaged to limit calorie intake by physically restricting food passage and inducing malabsorption. However, it is now clear that the success of bariatric surgery lies rather in the impact of these procedures on the biological regulation of energy homeostasis.  In this review we summarize the complex bi-directional communication system known as the gut-brain axis with special focus on gut hormones, bile acids and gut microbiota. We discuss the impact of obesity, lifestyle interventions and bariatric surgery upon the gut-brain axis. Finally, we discuss the progress being made to pharmacologically mimic the beneficial hormonal milieu of bariatric surgery.

 

INTRODUCTION

 

Obesity, defined as the accumulation of excess adipose tissue that impairs health, is now recognized as a chronic progressive disease. Its prevalence continues to increase unabated (1). Globally in 2016, approximately 39% of the adult population were overweight (1.9 billion) and 13% had obesity (> 650 million) (1). Increased fat deposition is the result of an imbalance between energy expenditure and consumption, which in turn is due to an alteration of the homeostatic and/or hedonic systems that regulate energy homeostasis (2). This simplistic definition does not consider how complex obesity is, being the consequence of interactions between genetic, environmental, dietary, psychological and socio-economic factors (3, 4). Eating behavior is governed by specific brain areas that integrate peripheral signals regarding nutrient intake and energy stores (5).

 

The obese state is a very difficult condition to treat because of the coexistence of low-grade inflammation, dysbiosis, hormonal and neurogenic imbalances (3, 4, 6) (Figure 1). These factors also make major contributions to obesity-related diseases, such as type 2 diabetes (T2D), cardiovascular disease, and some types of cancer, impacting adversely upon health, socio-economic factors and life expectancy (1, 7, 8). Weight loss can improve these co-morbidities and increase life expectancy. However, current treatments that emphasize dietary (especially low-calorie diets) or lifestyle approaches for obesity lack long-term efficacy. A meta-analysis of weight loss clinical trials mediated by lifestyle interventions showed an average weight loss of 5% to 9% in the first 6 months, which back-tracked to 3% to 6% in those studies where 48-month data were available (9). Another review assessing the long-term outcomes of calorie-restricted diets showed that up to two-thirds of dieters regain more weight than they lost during their weight loss programs (10). The data for the impact of anti-obesity medication (AOM) on total weight loss percentage after 1 year are highly variable, ranging from 3% with lorcaserin to 9.4% with phentermine/topiramate (11).

Figure 1. Schematic diagram comparing the simplistic definition of obesity, thought to be the result of an imbalance between energy expenditure and consumption (1), with the very complex physiopathology of the obese state (2). This is the result of genetic, inflammatory, microbiota, endocrine, neurogenic and other factors. This pathophysiological complexity underlies the difficulty in finding effective treatments to combat obesity.

Bariatric/metabolic procedures are currently the most effective treatments for people with severe obesity both in terms of weight loss amount and sustainability and the resolution of complications (12). The mechanisms behind the success of bariatric/metabolic surgeries remain to be fully elucidated but post-surgical changes in gut-derived hormonal peptides, bile acids (BA), gut microbiota, and vagal tone are suggested to be involved (13, 14). Importantly, research studies undertaken in animal models and patients with obesity undergoing bariatric surgery have significantly advanced our understanding of the important interplay between the central nervous system (CNS) and the gastrointestinal (GI) tract in regulating energy and glucose homeostasis. Several brain regions integrate continuous information provided by chemical messengers and neural networks arising from the periphery that reflect nutrient availability, the body's energy status, and play a key role in regulating energy homeostasis. The GI tract is responsible for generating the majority of inputs communicated to the CNS regarding both the quality and quantity of a meal. This complex bi-directional communication system between the GI tract and the CNS has come to be known as the gut-brain axis (4, 15) (Figure 2).

Figure 2. Schematic diagram illustrating the gut-brain axis. The entire gastrointestinal tract (GIT) is responsible for generating multiple signals that inform the central nervous system (CNS) regarding quality and quantity of a meal. Key components include neural signals, gut hormones, bile acids, and gut microbiota.

In this review we will explore the gut-brain axis in detail, focusing on the role of gut hormones, Bas, and gut microbiota. We will concentrate our attention on the perturbations of the gut-brain axis in the obese state, and the compensatory response to weight loss induced by lifestyle interventions. Finally, we will discuss the impact of bariatric surgery upon gut hormones, Bas, and gut microbiota and the evidence supporting a role for these factors in mediating the beneficial weight and metabolic effects of bariatric surgeries.

 

THE PHYSIOLOGY OF BODY WEIGHT REGULATION AND THE GUT—BRAIN AXIS

 

During the majority of human evolution food has been scarce. It is therefore not a surprise that endogenous systems have evolved to prioritize food-seeking behaviours when necessary to ensure adequate nutrition for reproduction and survival.  Neuronal circuits within the brain control energy homeostasis, integrating peripheral signals of energy availability originating from the GI tract, adipose tissue mass, muscle mass, and bone density, together with information from higher cognitive centers and external environmental food cues (16). Upon food consumption, sensory information reflecting nutrient availability is transferred from the GI vagal and/or somatosensory (spinal) afferent fibers to the nucleus tractus solitarius (NTS) that, in turn, are integrated and transferred to several other brain centers, including the hypothalamus (17).

Figure 3. Schematic diagram illustrating the central effects of hormones that control eating. Leptin and insulin are secreted in proportions to body fat mass and decrease appetite by inhibiting neurons that produce the NPY and AgRP, while stimulating melanocortin-producing neurons in the ARC region of the hypothalamus, near the third ventricle of the brain. NPY and AgRP stimulate eating, and melanocortins inhibit eating, via higher-order neurons. Activation of NPY/AgRP-expressing neurons inhibits melanocortin-producing neurons. The gastric hormone acyl-ghrelin stimulates appetite by activating the NPY/AgRP-expressing neurons. Gut hormones released from the GI tract in response to eating, including PYY, inhibit these neurons and thereby suppress appetite and decrease energy intake. Abbreviations: AgRP, agouti-related peptide; ARC, arcuate nucleus; CART, cocaine and amphetamine-regulated transcript; NPY, neuropeptide Y, PVN, paraventricular nucleus; PYY, peptide tyrosine-tyrosine 3-36; POMC, pro-opiomelanocortin: Lepr, Leptin receptor; GHSR, Ghrelin receptor, MC3R, Melanocortin 3 receptor, MC4R, Melanocortin 4 receptor, Y1r, NPY receptor; Y2r, NPY/PYY3-36 receptor.

The hypothalamus is the key integrative brain site that governs reciprocal orexigenic and anorexigenic behavioral responses, as well as adaptive metabolic changes in response to alterations in food availability and activity levels (18)(Figure 3). The arcuate (ARC), paraventricular (PVN), ventromedial and dorsomedial nuclei, as well as the lateral hypothalamus, are the most important hypothalamic areas involved in energy homeostasis (19). The ARC responds to peripheral and central signals reflecting nutrient availability and energy expenditure by releasing neurotransmitters from two separate and reciprocally connected neuronal populations: pro-opiomelanocortin (POMC)/cocaine-and-amphetamine-regulated transcript (CART) and neuropeptide Y (NPY)/agouti-related protein (AgRP) neurons. NPY/AgRP neurons are situated in the medial ARC and release AgRP and NPY, which stimulate hunger, appetite, and decrease energy expenditure (17, 19-21). Neighboring POMC and CART-containing neurons located in the lateral ARC release α-melanocortin-stimulating hormone (α-MSH) and CART respectively (17). These neurotransmitters are antagonistic of AgRP and NPY and act via the melanocortin-4 receptor (MC4R) to decreased hunger and increased energy expenditure (22) (Figure 3). In addition to vagal signaling, gut hormones can also directly influence these hypothalamic circuits. For example, injected peptide YY (PYY) can inhibit food intake by binding to Y2 receptors localized to the ARC (23) (Figure 3).

Figure 4. Schematic diagram illustrating the mechanisms involved in regulating feeding behavior. Nutrient entry into the GI tract causes gastric and intestinal distension, secretion of pancreatic enzymes and BA, altered enteric and vagal nerve signaling and exposure of EECs to nutrients with altered circulating gut hormone levels (e.g. decrease in orexigenic hormone acyl-ghrelin and increase in anorectic hormones PYY3-36 and GLP-1). Gut-derived signals (nutrients, hormones, and neural) and adipokines (e.g. leptin and others) act directly and indirectly upon the brainstem and hypothalamic areas (see Figure 3 for a detailed description of hypothalamic nuclei controlling energy homeostasis). All of these factors are involved in the regulation of homeostatic hunger. Social factors, emotion, reward, pleasure, increased food availability and sensory cues can influence brain reward and higher cognitive brain regions leading to altered feeding behavior (hedonic influences on hunger and appetite control). Taste and olfactory signals can also influence energy intake acting on both homeostatic and brain reward systems. Insulin leptin, GLP-1, PYY and ghrelin are present in saliva with cognate receptors on taste buds and olfactory neurons. Abbreviations: AgRP, agouti-related peptide; ARC, arcuate nucleus; CART, cocaine and amphetamine-regulated transcript; EEC’s, enteroendocrine cells; FGF-19, fibroblast growth factor-19; GLP-1, glucagon-like peptide 1; LHA, lateral hypothalamic area; NPY, neuropeptide Y, peripheral nervous system, PNS; PVN, paraventricular nucleus; PYY, peptide tyrosine-tyrosine 3-36; POMC, pro-opiomelanocortin, sympathetic nervous system, SNS.

Several other brain regions have key roles in energy homeostasis. `The area postrema (AP) is proximally located to NTS and, along with the ARC, are unique in that they have incomplete blood-brain-barriers, thus allowing them to be directly accessed and influenced by blood-borne gut hormones and other circulating factors (4). In animal models, AP lesions have been shown to result in diminished central effect of  several gut hormones (24). Signals from the GI tract also interact with the brain reward systems that constitute dopaminergic neurons located in the ventral tegmental area, nucleus accumbens, and other sites.  These neuronal pathways are thought to mediate effects of exposure to hedonic food cues present in obesogenic environments of Western societies, possibly contributing to the creation of the desire to eat in the absence of an energy requirement in what has been termed “hedonic obesity” (18) (Figure 4).

 

More broadly, the gut-brain axis includes bidirectional communication between the CNS and the enteric nervous system (ENS), autonomic nervous system (ANS) (with sympathetic and parasympathetic branches), neuroendocrine system, gut immune system, BAs, and gut microbiota (25, 26). Both acute and chronic alterations in these systems can arise in response in changes in energy expenditure and consumption (6). Peripheral energy-regulating signals are traditionally classified as long-term signals of energy balance, such as leptin and insulin levels reflective of body adipose stores (“adiposity signals”), and short-term signals, which convey information regarding nutrient and meal-derived energy availability (5) (“satiety signals”), whereas the role of the immune system of the GI tract is still under investigation  (Figure 4).

Figure 5. Schematic diagram illustrating the regulation of an L-cell, one of the several EECs present in the GI tract. Nutrients and their interaction with gut microbiota and BAs in the intestinal lumen activate luminal receptors located on the apical cell membrane, which then activate intracellular metabolism leading to calcium influx to induce the synthesis and release of gut hormones into the sub-epithelial space. Luminal receptors includes receptors for short-chain fatty acids (SCFAs) (e.g., GPR41, GPR43), long chain fatty acids (e.g., GPR40, GPR120), proteolytic products (e.g., CaSR) and BAs (e.g., TGR5). Various gut-derived hormones glucagon-like peptide 1 (GLP-1), peptide YY (PYY), and oxyntomodulin (OXM) are synthesized, secreted and released from L-cells systemically to induce an effect on various tissues throughout the body such as the brain. These hormones can act systemically or cooperate with the enterocytes via local paracrine action. Their systemic effects could be endocrine but also neural through the activation of afferent neurons located in the GI wall. The secretion of gut hormones can be stimulated, in turn, by circulating hormones and glucose or by stimulation from enteric nerves.

Most of these communications related to energy homeostasis involve hormonal and neural signals, which are quite substantial given that the GI tract releases more than 100 different hormonally active peptides (27) and contains approximately 500 million neurons (28). In response to nutrient ingestion, stretching of stomach mechanoreceptors generates the first ENS feedback signals to the CNS mediating meal cessation (4, 29). Subsequently, digested luminal nutrients come in contact with the microvilli of apical cell membranes of enteroendocrine cells (EECs) located in the epithelium of the GI tract to stimulate gut hormone release. Therefore, the majority of signaling and communication within the gut-brain axis is initiated in response to pre-absorptive nutrients (15) (Figure 5 illustrates in detail the biology of an L-cell as a model of an EEC). Following gut hormone receptor activation, nutrient-derived signals exert local control over intestinal function and are conveyed directly and indirectly to the brain via vagal and spinal afferents (30-32) (Figure 5).

 

Digestion and absorption take place predominantly in the stomach and small intestine where dense innervation originating from splanchnic and vagal nerves transmit neurological signals generated during the process of nutrient sensing (29). Additional roles of the ENS include regulation of gastric emptying by vagal activation (17, 33) and mediation of GI endocrine signaling via vagal nerve afferents that project into the lamina propria of the gut ending at the basolateral cell membrane of EECs. It is through these nutrient-specific sensory signals that the GI tract informs the CNS about a meal’s energy and macronutrient content (34).

 

LINKING THE GUT AND BRAIN: GUT HORMONES, BILE ACIDS AND THE GUT MICROBIOTA

 

Gut Hormones

 

As previously described, the gut-brain neuronal-signaling axis is initiated by nutrient-induced gut hormone secretion (32). EECs distributed throughout the entire GI tract length respond to luminal nutrients and release a panoply of gut hormones that act as endocrine, autocrine and paracrine regulators of energy and glucose homeostasis (35) (Figure 5). Although nutrient ingestion triggers the secretion of numerous gut hormones, in this review we will focus on glucagon-like peptide 1 (GLP-1), oxyntomodulin (OXM), and PYY (all of which are released from L-cells and have central appetite-suppressing effects (35, 36)), pancreatic polypeptide (PP), cholecystokinin (CCK), ghrelin, and anandamide. 

 

The enteroendocrine L-cells responsible for secreting PYY, GLP-1 and OXM reside throughout the GI tract with the highest concentrations in the ileum and colon. In response to nutrient intake, circulating PYY, GLP-1 and OXM  levels show a biphasic increase with an initial early peak within 15 minutes and then a later peak around 90 minutes post-ingestion (4, 37, 38). The early increase is thought to be mediated by neural (vagal) and/or hormonal mechanisms whilst the later peak, which is in proportion to the energy intake, is thought to result from the direct contact of nutrients with L-cells located in the distal small intestine and large intestine (39).

 

GLUCAGON-LIKE PEPTIDE 1

 

Glucagon-like peptide 1 results from post-translational processing of the preproglucagon gene (37, 40). Glucagon-like peptide 1 exerts its metabolic effects by activating the GLP-1 receptor, which is widely expressed in the GI tract, pancreas, and CNS (41, 42). Actions mediated by the GLP-1 receptor include enhancing glucose-dependent insulin release (incretin effect) (43), inhibition of glucagon secretion (44), and stimulation of  satiety centers in the ARC, NTS and AP leading to decreased hunger and increased satiation (45). In addition, GLP-1 limits energy intake by reducing the rate of gastric emptying, which in turn increases gastric distension (46). As a result of these functions, GLP-1 receptor agonists are currently used for the treatment of both T2D and obesity (47).

 

Glucagon-like peptide 1 is rapidly inactivated by the enzyme dipeptidyl peptidase-IV (DPP-IV) with only about 10% of GLP-1 reaching the systemic circulation (48-50). Thus, GLP-1 is thought to mainly act in a paracrine fashion. The vagus nerve is particularly important for the action of GLP-1 as demonstrated by vagotomy studies altering the effects of this hormone (51). Glucagon-like peptide 1 also stimulates the brainstem to enhance motor output and/or thermogenesis (52).

 

OXYNTOMODULIN

 

Like GLP-1, OXM is synthesized by post-translational processing of the preproglucagon gene (53, 54). Other similarities of OXM to GLP-1 include binding to GLP-1 receptors within the GI tract, the pancreas, and the ARC; subsequent reductions in gastric acid secretion, blood glucose concentrations and food intake (53, 55, 56); and degradation by DPP-IV (57). In addition, OXM administration enhances satiety and increases energy expenditure in both animal models and humans (53, 54).

 

PEPTIDE YY

 

Circulating levels of PYY are low in the fasted state (58, 59) and increase during nutrient ingestion in proportion to the caloric content (60), exhibit differential responses according to the specific macronutrient composition of the meal (36, 59), and remain elevated for several hours after a meal with sustained endocrine effects (61).

 

Peptide YY circulates in two native forms: PYY1−36 (minor form) and PYY3−36 (major form) (36, 59).  Peptide YY3−36 results from the N-terminal cleavage of PYY1−36 by the enzyme DPP-IV (59). Interestingly, PYY1−36 and PYY3−36 have divergent actions on appetite, glucose homeostasis and differential binding affinities of each form for the five neuropeptide Y receptor (YR) subtypes (59). Y2 receptors are located on the vagus nerve, in the NTS and in the ARC (23, 36, 60). Peptide YY1−36 has equivalent affinities for Y1R and Y2R, whereas PYY3−36 is a high-affinity ligand for Y2R (59).  By binding to the Y2 receptor, PYY3-36 decreases energy intake by inhibiting the orexigenic effects of NPY neurons and activating the anorexigenic POMC neurons in the ARC (4, 20, 62) (Figure 3), physiological effects supported by studies showing that PYY knockout mice become hyperphagic and obese (36, 60). Increased PYY levels have been associated with prolonged appetite loss and food aversion during exogenous administration and following bariatric surgery (6, 63-65).

 

PANCREATIC POLYPEPTIDE

 

Pancreatic polypeptide is secreted by specialized F-cells located in the islets of Langerhans (66) during the pre-absorptive (cephalic) phase of nutrient metabolism and for up to 6 hours post-prandially in proportion to energy intake (67, 68). Pancreatic polypeptide acts centrally upon the Y4 receptor within the AP, NTS, and the ARC, reducing energy intake. Peripherally, it induces gallbladder relaxation, inhibits pancreatic secretion and delays gastric emptying thus inducing satiety (69, 70). Pancreatic polypeptide is a potent appetite suppressant (71) and studies have demonstrated a difference in PP concentrations in anorexic and obesogenic states, where it is increased and diminished respectively (61). Moreover, studies in people with Prader-Willi syndrome and obesity suggest that circulating post-prandial PP levels are reduced in comparison to healthy individuals. Furthermore, intravenous PP injection to these patients led to a significant decline in energy intake (72).

 

CHOLECYSTOKININ

 

Cholecystokinin is secreted from EECs located mainly in the duodenum and jejunum (73). Cholecystokinin release is stimulated by fat and protein ingestion and its circulating concentrations increase within 15 minutes after meal ingestion (6, 74). Cholecystokinin has a short half-life and it acts upon CCK-1 and CCK-2 receptors located throughout tissues of the GI tract and the CNS, including the vagal nerve, NTS and hypothalamus (6). Cholecystokinin increases gallbladder and GI motility and secretion but also has an active role in controlling food intake, energy expenditure and glucose utilization (17). Cholecystokinin reduces energy intake in a dose-dependent manner and it is a specific mediator of fat and protein satiation (6, 75). It has been suggested that it acts synergistically with leptin and amylin, a pancreatic hormone co-secreted with insulin (17). However, repeated doses can induce tolerance to CCK (6), potentially explaining why attempts to use CCK-derivatives as a medication to induce weight loss have failed (76).

 

GHRELIN

 

Ghrelin is a 28-amino-acid orexigenic hormone and is secreted by P/D1 cells located primarily in the gastric fundus in the absence of nutrient intake, leading to increased appetite and food intake (77). Ghrelin is also produced by the pituitary gland (77) and within the ARC and PVN area of the hypothalamus. Ghrelin is secreted as the inactive des-acyl-ghrelin. The active orexigenic form, acyl-ghrelin, is synthesized by the action of ghrelin O-acyltransferase enzyme (GOAT) (77) and can bind to growth hormone secretagogue receptor (GHS-R) to increase food intake in rodents (77)and humans (77, 78). The extremely complex ghrelin/GOAT/GHS-R system has a crucial role in the regulation of energy and metabolism as well as in the adaptation of energy homeostasis to environmental changes (77).

 

Acyl-ghrelin administration to humans has been used as an orexigenic agent in patients with anorexia that accompanies cachexia (79). A rising circulating ghrelin level precedes nutrient ingestion and decreases rapidly after a meal (78) which has led to the speculation that this is the first discovered “hunger hormone” (80). Plasma levels of ghrelin increase after diet induced weight loss, thought to be part of the body’s homeostatic adaptation response that restores lost weight, and are very high in patients with anorexia nervosa (81). Nutrient intake but not water ingestion is the main regulator of ghrelin leading to a decrease of its plasma levels (82). Peripheral ghrelin exerts its orexigenic actions through the stimulation of NPY/AgRP co-expressing neurons (83).

 

ANANDAMIDE

 

Anandamide and other bioactive lipids belonging to the endocannabinoid system contribute to the gut-brain axis. These molecules are secreted in the GI tract and activate endocannabinoid receptors 1 and 2 (CB1/CB2) (3, 4) which are expressed in the CNS, peripheral nervous system, liver, pancreas, adipose tissue, and immune cells (84). Exogenous cannabinoids convey orexigenic effects and so it is not a surprise that the endocannabinoid regulates gut motility and appetite (3, 84). Endocannabinoid receptor 1 antagonists were used to induce weight-loss in subjects with obesity before being withdrawn for their severe psychological side-effects (3, 85), including increased suicidality (86).

 

Bile Acids

 

Bile acids (BAs) are endogenous steroid molecules synthesized from cholesterol in the liver, stored in the gallbladder and secreted into the duodenum upon nutrient ingestion. These amphipathic molecules facilitate micelle formation, promoting the digestion of dietary fat and fat-soluble vitamins. More recently, BAs have also been shown to play a role in regulating glucose and lipid metabolism and energy expenditure via the activation of BA receptors in the liver, gut, and peripheral tissues (87, 88). Interactions between BAs, their receptors, and the gut microbiota determine synthesis, metabolism, and distribution of bile acids in the body (88).

 

There is complex cross talk between BAs, gut hormones and the microbiome (Figure 6). Bile acids stimulate GLP-1 secretion via activating G protein‐coupled receptors (TGR5) on L-cells and fasting total circulating BAs levels are positively correlated with post-prandial GLP-1 levels (89). TGR5 receptors are also located on skeletal muscle and in brown adipose tissue where they increase energy expenditure by promoting the conversion of inactive thyroxine (T4) into active thyroid hormones (T3) (90). Bile acids have been shown to act on farnesoid X receptors (FXR). During BA binding of FXR on pancreatic β cells, insulin release is increased (91). Bile acid activation of intestinal FXR-containing cells stimulates the secretion of fibroblast growth factor-19 (FGF-19), a protein that contributes to improved peripheral glucose disposal and lipid homeostasis, increased metabolic rate, and reduced weight (92, 93). In animal studies, BA supplementation has been shown to reduce weight gain (90). In humans, postprandial BA levels are also inversely related with body fat mass (94). Thus, the physiologic effects of BA likely extend beyond the gut and pancreas to include actions that improve body weight and glucolipid metabolism.

Figure 6. Schematic diagram illustrating the complex cross talk between BAs, gut hormones, FGF-19, and the microbiome. BAs are important regulators of energy balance and glucose metabolism, primarily via the FXR and the TGR5. The trans-intestinal BAs flux activates intestinal FXR, inducing synthesis and secretion into the circulation of the ileal-derived enterokine FGF-19. FGF-19 can improve glucose tolerance by regulating insulin-independent glucose efflux and hepatic glucose production. FGF-19 can also increase energy expenditure with its central and peripheral effect in the adipose tissue. BAs acting via TGR5 stimulate L-cell secretion of GLP-1 (and PYY) then enhancing insulin secretion acting on β-cells. TGR5 activation in muscle and brown adipose tissue promotes the conversion of inactive thyroxine into active thyroid hormone inducing thermogenesis. BAs can reduce food intake centrally through FGF-19 and anorectic gut hormones. BAs also regulate gut microbiota composition. BAs are actively reabsorbed from the terminal ileum and returned via the portal circulation to the liver. A small percentage of BAs are deconjugated by gut bacteria, forming secondary BAs, which are reabsorbed or excreted in feces. Red dotted lines: FGF-19 effects. Blue dotted lines: IGF-1 effects. Green lines: BAs circulation.

The Gut Microbiota

 

The healthy human gut hosts trillions of microorganisms with a ratio of bacterial-to-human cells of 1.3:1, comprising a complex ecosystem referred to as the gut microbiota (95, 96).  These microbes exist within a symbiotic relationship with their human host, who provides a nutrient-rich environment. The microbiota, in turn, provides metabolic processing of these nutrients that the host's genome does not possess (97, 98). With more than a thousand different bacterial species, the diversity and function of the microbiota is dynamic depending on the host’s diet, antibiotic exposure and other environmental factors (98, 99).

 

The extent of the symbiotic relationship between the host and the microbiota is highlighted by studies showing that mice lacking a microbiota (germ-free) have reduced adiposity, energy intake, and energy extraction from a standard rodent diet compared to conventionally-raised mice (100). More recent studies including germ-free rats transplanted with the microbiota of either obesity-prone or obesity-resistant rats confirmed the importance of the microbiome for the production of enzymes involved in energy harvesting from indigested carbohydrates (75). Both the Westernized-diet and obesity fecal transplant models are associated with an increased ratio of bacteria belonging to the Firmicutes phylum compared to the Bacteroidetes phylum, which is reversed upon surgical and dietary interventions (29, 101, 102).

 

Gut microbiota have been demonstrated to affect adiposity and weight-gain through several pathways.  A typical Western diet contains indigestible carbohydrates, such as resistant starches and plant cell wall polysaccharides that are hydrolyzed by gut microbiota generating small-chain fatty acids (SCFA) (3). Short chain fatty acids in the form of butyrate, acetate, and propionate provide approximately 10% of the host's daily energy requirements (103). In the obesogenic state, feces contain an increased quantity of SCFA, especially propionate (103). Short-chain fatty acids are not always correlated with increased weight-gain as some may possess beneficial properties (4, 29). In animal models, prebiotics (indigestible compounds that can modulate the composition and activity of the gut microbiota) and oral or intestinal SCFA infusions lead to a reduction in food consumption and a decrease in body weight. This occurs when prebiotics and supplements promote the growth of favorable microbial species or when SCFA activates signaling pathways that ultimately increase gut hormone synthesis (104-106). For example, SCFA’s have also been shown to activate free fatty-acid receptors 2 and 3 (FFAR2/FFAR3) in the GI tract, immune cells, liver and adipose tissue (107). Intestinal FFAR2/FFAR3 receptors are expressed by EECs, in particular, L-cells and, when activated, can facilitate release of gut hormones, such as GLP-1 and PYY (107). Moreover, FFAR3 expressed within the ENS and ANS can stimulate the sympathetic tone in the adipose tissue regulating fat storage as well as glucose utilization in muscle and liver tissues (3, 108). The gut microbiota may also moderate the endocannabinoid tone affecting colonic CB1 expression and anandamide concentrations (3, 109). Finally, gut microbiota can enhance energy expenditure by intracellular thyroid hormone activation via FXR signaling (3, 90, 99), which may help to explain the observation that germ-free mice are resistance to adiposity despite an increased food intake (110).  

 

Obesity is characterized by the presence of chronic low-grade inflammation. In another pathogenic pathway involving the microbiota, high-energy dense diets can lead to obesity and obesity-related diseases through changes in bacterial species composition that favors an increase in systemic lipopolysaccharide (LPS) concentrations (111, 112). Lipopolysaccharide is the pro-inflammatory component located in the cell wall of gram-negative bacteria that can enter the circulation when the permeability of intestinal epithelium is altered in a process called metabolic endotoxemia (111, 112).  Leakage of LPS into the systemic circulation is a proposed trigger of a cascade of pro-inflammatory events in adipose tissue and throughout the whole body  mediated by LPS stimulation of the toll-like receptor 4 (TLR4), which enhances the synthesis of inflammatory cytokines linked with reduced host insulin sensitivity (113, 114). Lipopolysaccharide can also inhibit the interstitial cells of Cajal, which are responsible for smooth muscle contraction in the gut and regulation of the ENS. This inhibition has been associated with disorders of both GI motility and gut hormones (4, 115).

 

The gut microbiota may also directly influence CNS-mediated stress and anxiety behaviours and the regulation of energy homeostasis (4).  For example, germ-free mice have been found to have a resistance to adiposity despite an increased food intake (127). Germ-free mice have been found to have higher gene expression of food intake-regulating peptides like GLP1 and OXM within the brainstem and hypothalamus, when compared to normally-reared mice (116).   On the other hand, a reduced leptin-mediated suppression of orexigenic peptides NPY and AgRP in the conventionally-raised mice has been noted, suggesting how the gut-microbiota could directly affect energy homeostasis leading to an increase in adiposity (116, 117).  Further studies are needed, however, to understand if specific manipulations of the gut microbiota phenotype could be used as obesity treatments (Figure 7).

Figure 7: Schematic diagram illustrating the possible causative links between an altered gut microbiota and obesity. The hydroxylation of indigestible carbohydrates and the altered intestinal permeability could lead to increased energy harvest and weight gain. The production of certain types of SCFA can reduce the sympathetic tone favoring fat accumulation in the adipose tissue and dysregulation of glucose utilization in muscles and the liver. Gut microbiota can induce a leakage of LPS into the systemic circulation (endotoxemia) and chronic low-grade inflammation. This is in turn responsible of insulin resistance and weight gain. The increased endocannabinoid tone could induce food intake. SCFAs, inflammation and BAs perturbations may all lead to a reduced activation of anorectic pathways. All these mechanisms could be responsible for weight gain, inflammation and obesity-related comorbidities. Abbreviations: FGF-19, fibroblast growth factor-19; GLP-1, glucagon-like peptide 1; PYY3-36, peptide tyrosine-tyrosine 3-36; SCFA; short chain fatty acid; LPS, Lipopolysaccharide; FFAR2 and FFAR3, free fatty-acid receptors 2 and 3; T2D, type 2 diabetes.

THE COMPLEXITIES OF ENERGY HOMEOSTASIS

 

The Physiology of Weight Regulation

 

As mentioned above (Figure 3), the adipokine leptin acts as a signal of long-term energy availability (fat mass), promoting satiety via its inhibitory action on orexigenic neurons located in the ARC of the hypothalamus (118). A recent report of a patient with leptin deficiency highlights key interactions between leptin and gut hormones. Leptin supplementation resulted in significant rises in meal-stimulated insulin, GLP-1, and PYY levels (61). In the same study, ghrelin levels were decreased, highlighting the regulatory effect of leptin on ghrelin secretion and the interplay between leptin, GLP-1 and PYY.

 

An important physiologic insight that has implications for pharmacological weight management is that gut hormones act synergistically. For example, GLP-1 and PYY in combination are more potent in reducing energy intake compared to either of the two hormones alone (119, 120). Oxyntomodulin, CCK and other gut hormones also act synergistically with GLP-1 to enhance its effects on appetite behaviours (120-123). An additional layer of complexity is added when considering that MC4Rs have been localized on L- and P/D1-cells and could, in turn, regulate GLP-1, PYY and ghrelin secretion (124). Furthermore, gut hormones influence energy homeostasis through interactions with the microbiome and BAs.

 

Hedonic factors can generate meaningful physiological responses that interact with homeostatic signals of energy availability in the regulation of body weight. This could lead to excess energy intake with possible weight gain. In humans, brain functional imaging studies, have shown that several gut hormones modulate neural activity in brain reward regions altering the reward value of food (2, 58, 125) by food cues, memory and social factors, and strongly influencing eating behavior (18). Exposure to food-related stimuli can induce changes in circulating gut hormone levels. Those in turn act upon brain reward pathways, either decreasing in the case of PYY or, increasing in the case of ghrelin, the reward value of food (23, 126).  Those hormones are also present in saliva and their cognate receptors are present on taste buds and the olfactory bulb (127, 128).  The taste and smell of food are key influencers of food selection with impacts on energy intake (127).

 

Adding to the complexity of the gut-brain regulation of body weight, studies have demonstrated that energy expenditure can increase without changes in activity but through the action of gut-derived neurohumoral signals that increase thermogenesis and basal metabolic rate (4). The existence of the gut-brain-brown adipose tissue axis has been hypothesized after studies showing how intestinal lipid-sensing activates vagal afferent fibers to enhance brown adipose tissue thermogenesis through a CCK-dependent pathway (129). Table 1 summarizes key gut hormone actions including their perturbations in the obese state and the changes induced by bariatric surgery procedures.

 

Table 1. GI Tract Hormones Involved in the Control of Energy Balance and Changes in their Circulating Levels Induced by Obesity and Bariatric Surgery

Hormone

EEC (Type)

Location

Receptor

Food

Intake

Other Effects

Obese State

Bariatric

Surgery

PYY

Ileum

(L cell)

Y2-R

↓ Gastric acid secretion and emptying

↓ Pancreatic and intestinal secretion

↓ Gastrointestinal motility

↑ Insulin secretion and vagus stimulation

GLP-1

Ileum

(L cell)

GLP-1R

↑ Insulin secretion

↑ β-cell proliferation and gene expression

↓ β-cell Apoptosis

↓ Gastric acid secretion and emptying

Ghrelin

Stomach

(P/D1cell)

GHS-R

↑ Growth-hormone secretion

↑ Gastric acid secretion and emptying

↑ Vasodilatation

↓ Insulin secretion

CCK

Duodenum, jejunum and pancreas

(I/L cell)

CCK 1, 2

↓ Gastric emptying

↑ Pancreatic secretion

↑ Gallbladder contraction

?

PP

Pancreas

(F-cell)

Y4, Y5

↓ Gastric emptying

↓ Leptin levels

↑ Insulin secretion

↓ β-cell Apoptosis

GIP

Duodenum and

jejunum

(K-cell)

GIP-R

?

↑ Insulin secretion and β-cell proliferation

↓ β-cell Apoptosis

↑ Lipoprotein lipase activity and fat deposition

↑ Fatty acid synthesis

↓?

OXM

Ileum

(L cell)

GLP-1R

↓ Gastric emptying and acid secretion

↓ Blood glucose

↑ Insulin secretion

↑ Energy expenditure

↑?

Glucagon

Pancreas

(α-cell)

GCGR

↑ Energy expenditure

↑ Blood glucose

?

Amylin

Pancreas

(β-cell)

AMY1-3

↓ Gastric emptying and acid secretion

↓ Postprandial glucagon secretion

↓ Glucose elevation

Insulin

Pancreas

(β-cell)

Insulin receptor

↑ Absorption

↑ Glycogen synthesis

↓ Blood glucose

↑ Lipid synthesis

↓ Lipolysis and proteolysis

Leptin

Adipose Tissue and gastric EECs

Leptin (Ob-R)

↓Glucose production and steatosis in the liver

↑ Glucose uptake and fatty acid oxidation in muscles

↓ Insulin and glucagon secretion

↑ Sympathetic nervous system tone

↑Thyroid hormones

Modulates immunity and fertility

FGF-19

Ileum

(FXR activation from BA)

FGFR 1, 2, 3, 4

Regulates glucose and lipid metabolism,

↑Hepatic protein and glycogen synthesis

↑ energy expenditure

NT

Jejunum

(L-cell)

NTR1, NTR2, NTR3

↓ Reduces GI motility and gastric secretion,

↑ Pancreatic and biliary secretion

?

↑?

Abbreviations: FGF-19, fibroblast growth factor-19; GLP-1, glucagon-like peptide 1; PYY3-36, peptide tyrosine-tyrosine 3-36; PP, pancreatic polypeptide, GIP, gastric inhibitory polypeptide; OXM, Oxyntomodulin; BA, Bile Acids: NT, Neurotensin. ? = effect not certain or not valid for every bariatric procedure.

 

The Obese State: Pathophysiologic Changes

 

Obesity is the result of a period of uncompensated chronic positive energy balance (130) when energy intake exceeds energy requirements. Dysregulation of the metabolic mechanisms controlling energy homeostasis includes an impaired gut hormone secretion response to nutrient ingestion (131). People with obesity have reduced circulating baseline and meal-stimulated levels of PYY and GLP-1 levels compared to lean subjects (131-133).  Lower circulating ghrelin levels and a reduced suppression of circulation ghrelin levels after nutrient intake have been demonstrated in people with obesity, suggesting that dynamic changes more than absolute values are important in appetite regulation (7, 40). Animal models with diet-induced obesity show reduced circulating concentrations together with impaired circadian secretion profiles of PYY and GLP-1 (134), in addition to an increase in ghrelin-producing cells (135). However, reduced diurnal variability in circulating ghrelin is thought to contribute to the lack of a regular meal pattern and the frequent snacking behavior often observed in individuals with obesity (77, 136).

 

The directionality of the association between obesity and altered gut hormone profile remains to be fully elucidated. For example, high energy intake per se may affect gut hormone responsiveness to ingested nutrients. Moreover, intestinal EEC population differentiation and responsivity is reduced in people with obesity, which may underlie their blunted gut hormone secretion (137). Obesity has also been shown to blunt the rise in circulating post-prandial BAs levels (33). Paradoxically, most studies have found increased total BAs levels in subjects with obesity suggesting that BAs composition could shift unfavorably with detrimental metabolic effects (138). Interestingly, while obesity is thought to be due to resistance to the effects of insulin and leptin within key weight regulatory centers in the hypothalamus, sensitivity to the effects of PYY, GLP-1 and OXM during exogenous administration is preserved, suggesting these hormones and their receptor systems offer a viable therapeutic target for the treatment of obesity (139).

 

As detailed in the previous section, a dysbiotic relationship between host and gut microbiota has been suggested to contribute to the development of obesity (140), with profound differences found between the composition of the gut microbiota of obese and lean individuals (141). Obesity is associated with the relative increase or reduction of certain bacterial species and the importance of the relative proportions of those species remains an area of active investigation. Transplantation of gut bacteria from obese mice to normal-weight germ-free mice results in weight gain in the recipients (142). Conversely, fecal transplantation from lean human donors to recipient patients with metabolic syndrome led to improvements in insulin sensitivity (143). A dysbiotic relationship may affect host energy and nutrient metabolism by altering intestinal mucosal permeability, and promoting increased fat storage in adipose tissue (110). The mechanism for this could be by enhancing the absorption of SCFA derived by otherwise indigestible luminal polysaccharides and by triggering inflammatory responses through a process referred to as “metabolic endotoxemia” (144, 145) (Figure 7). Altered neural responses to food cues in people with obesity compared to people with normal weight have been confirmed by brain-imaging studies showing an increased stimulation of central reward pathways in response to eating or food cues (2). In addition, there is evidence that eating behavior in people with obesity becomes dissociated from perceptions of satiety and hunger (146, 147). Furthermore, in the obese state, enhanced endocannabinoid tone, CB1 expression, and plasma and adipose tissue endocannabinoid concentrations coexist (3). All these complex pathophysiological changes create an internal environment conducive to expression of unwanted weight gain, maintenance of the obese state, and resistance to diet-induced weight loss, providing an explanation as to why treating people with obesity can be challenging (Figure 7).

 

BARIATRIC/METABOLIC SURGERY

 

Bariatric/metabolic surgery is recognized as the most effective weight loss treatment for people with severe obesity (148).  Procedures with the best outcomes involve surgical modifications of the anatomy of the GI tract that alter nutrient flow, thus affecting GI tract biology (83). Many clinical trials have demonstrated the superiority of bariatric surgery in terms of sustainability of weight loss and resolution of obesity-related comorbidities, especially diabetes, when compared with intensive medical interventions (12, 149, 150). Mechanisms other than restriction and/or malabsorption are responsible for this superiority and this has resulted in a marked increase in the number of procedures undertaken worldwide (151). Currently, the most commonly performed bariatric/metabolic procedures globally are sleeve gastrectomy (SG) and Roux-en-Y gastric bypass (RYGB), whereas purely restrictive procedures, like gastric banding, are now performed less frequently (151) (Figure 8). However, post-operative weight loss can be highly variable (152), an important consideration total amount of weight loss plays a major role in determining post-operative remission of comorbidities (153).

Figure 8. Schematic diagram illustrating the normal upper GI anatomy (a) and the two most commonly performed bariatric surgical procedures. The metabolic procedures: (b) RYGB and (c) SG (Refer to the main text for a detailed description of surgical techniques). Abbreviations: RYGB, Roux-en-Y gastric bypass, SG, Sleeve gastrectomy.

Roux-en-Y gastric bypass involves division of the stomach into two parts, generating a small gastric pouch (20-30 mL), which is then anastomosed with the mid-jejunum, creating the alimentary limb or Roux limb. Nutrients bypass most of the stomach, duodenum, and the proximal jejunum. In the common limb, after the anastomosis of the biliopancreatic limb with the jejunum, nutrients, BAs and pancreatic secretions mix and the absorption of nutrients occurs (154). In a SG, a transection along the greater curvature is performed, removing the fundus and body and creating a tube-like stomach (155). The transit of gastric contents into the duodenum is rapid. The SG was initially performed as a first-stage procedure followed by a second more invasive malabsorptive step (biliopancreatic diversion), but the significant weight loss results observed with this procedure led to its adoption as a standalone approach. Because it is a simpler operation compared with RYGB and has fewer complications with similar short-term weight-loss, SG have become the most common bariatric procedure worldwide (151).

 

Biological Changes Favoring Sustained Weight Loss and Metabolic Improvement Following Bariatric/Metabolic Surgery

 

A negative energy balance is a key component of many lifestyle interventions. Unfortunately, weight regain is very common after initial weight loss. Multiple powerful adaptive biological changes occur in response to weight loss from lifestyle alone that lead to increased hunger, enhanced neural responses to food cues and heightened drive to consume energy-dense foods. These include decreased total energy expenditure secondary to reduced lean muscle mass, sympathetic activity (156), circulating leptin, GLP-1 and PYY levels, along with increased ghrelin levels (147). Other changes following lifestyle-induced weight loss that have been described and may contribute to weight recidivism include impaired circulating BAs levels, an altered gut microbiome, and decreased vagal signal transmission (10, 157).

Figure 9. Schematic diagram illustrating the different biological changes induced by weight loss achieved through dieting (upper part) compared to bariatric/metabolic surgery (lower part). Powerful compensatory biological changes contribute to the high rate of weight recidivism observed following lifestyle intervention weight management. Many homeostatic mechanisms act to restore a higher body weight and these includes hormonal alterations and a decreased energy expenditure leading to increased hunger and energy consumption. By contrast, bariatric surgery leads to a favorable biology that includes increased satiety hormones, reduced ghrelin, enhanced BA secretion and a “lean” microbiota. Together these mechanisms lead to reduced hunger and a shift towards healthier food options with a resetting of body weight “set point” to a lower level facilitating meaningful and sustained weight loss. References for this figure: (149, 158, 159). Abbreviations: GLP-1, glucagon-like peptide 1; PYY3-36; peptide tyrosine-tyrosine 3-36. *Suggestion that leptin sensitivity may improve

Weight loss following RYGB and SG are the result of multifactorial mechanisms and not from malabsorption or restricted stomach size alone (160, 161) (Figures 9 and 10). Reduced energy intake, as a result of altered eating behavior, is recognized as the main driver for weight loss following these procedures, and increased exposure of EECs to ingested nutrients is thought to play a key mediating role in the expression of these appetitive behaviours (83) (Table 1 and Figure 10). In contrast to lifestyle approaches to weight loss, favorable changes in these behaviours following bariatric/metabolic procedures include reduced hunger and neural responsiveness to food cues. Multiple studies have shown that bariatric surgery causes marked elevations in nutrient-stimulated levels of several anorectic hormones including PYY and GLP-1, along with decreased ghrelin levels, which have been reported post-RYGB but are more pronounced post-SG (162, 163). Following RYGB, increased nutrient-stimulated circulating levels of PYY and GLP-1 are most likely as a result of increased nutrient stimulation of L-cells as a consequence of anatomical rearrangement. Sleeve gastrectomy leads to rapid gastric emptying and enhanced exposure of L-cells to nutrients with increased nutrient-stimulated PYY and GLP-1 levels, but to a lesser extent than following RYGB. Sleeve gastrectomy leads to sustained and greater reduction in circulating acyl-ghrelin levels than RYGB because of the removal of the fundus of the stomach where most ghrelin-producing cells are located (164). These changes are present immediately after surgery and sustained up to 10 years post-operatively (165, 166). Oxyntomodulin levels are increased after RYGB (167) and a rise in CCK levels has been demonstrated following both RYGB and SG (163). Emerging evidence also suggest that the number of EECs changes after bariatric surgery. The total numbers of EECs in the stomach and duodenum of people with obesity are reduced when compared to lean individuals (31) and this has been found to normalize 3 months post-SG (158).

Figure 10. Schematic diagram illustrating RYGB and SG and the mechanisms leading to weight loss and resolution of comorbidities. For every mechanism the effect of the procedure is represented with a “” when stimulating or “” when suppressing. A “+” means that the proposed mechanism is present only after surgery when compared to the pre-operative period. When the effect is stronger for one of the two procedures there is a double arrow compared with a single one. When the effect is missing for one procedure it means that the mechanism is procedure specific. Abbreviations: RYGB, Roux-en-Y gastric bypass; SG, Sleeve gastrectomy; GLP-1, glucagon-like peptide 1; PYY3-36; peptide tyrosine-tyrosine 3-36; GIP, gastric inhibitory polypeptide; FGF-19, fibroblast growth factor-19, CCK; cholecystokinin.

Variability in EEC secretion response may underlie differences in weight loss responses to bariatric/metabolic procedures. Profound anorexia and excessive weight loss post-SG have been associated with markedly elevated circulating fasted and post-meal PYY levels (65). Patients with poor weight loss after surgery have been found to have increased appetite coupled with lower meal-stimulated GLP-1 and PYY and higher ghrelin levels when compared with good responders (168). Additional support for the importance of EEC in weight loss responsiveness in the post-op period comes from data showing that administration of octreotide (a general inhibitor of EEC secretion), or selectively blocking GLP-1 and PYY, promotes appetite and weight gain (14, 65, 169).

 

Following SG and RYGB, food becomes less rewarding and there is a shift in preference from energy dense food rich in fat and sugar to healthier options enabling patients to adopt more favorable eating behaviours (158). These changes in eating behavior are the result of multiple mechanisms, some of which are common to both SG and RYGB and others that are procedure specific (Figure 10).

 

Studies of the physiological changes following bariatric/metabolic have also elucidated novel effectors of changes in weight and metabolism, many of which are gut-related and discussed above. For example, following RYGB and SG, changes in circulating BAs levels and composition are reported that may contribute to weight loss and improved glucose metabolism. Despite their anatomical differences, RYGB and SG exert similar effects on BA composition and circulating concentrations, although the changes observed following SG are more modest (87, 170). The exact mechanism responsible for elevated BAs following RYGB and SG is unclear, but animal studies suggests that an accelerated nutrient flow to the distal small intestine is a key mechanism (171). Indeed, in animal models, rerouting bile to the distal small bowel by transposing the common bile duct increases plasma BA levels similarly those seen after RYGB and results in weight loss, improved glucose metabolism, and reduced hepatic steatosis. The rise in circulating BAs appears even greater several months post-operation and may be due to intestinal cellular adaptations (172), increased hepatic synthesis, altered enterohepatic recirculation of bile, or a combination of these possibilities. Post-surgically increased BAs diversity might also impact on GLP-1 secretion and energy expenditure.  Binding of BAs to TGR5 receptors in skeletal muscle and brown adipose tissue may contribute to enhanced action of thyroid hormones, thereby increasing energy expenditure (173). Therefore, BAs could contribute to weight loss and metabolic improvements after bariatric surgery through direct and indirect regulatory mechanisms.

 

Weight‐loss surgery can also affect the interplay between BAs and gut microbiota, which can have favorable metabolic effects in the post-operative period (174) (175). For example, in RYGB subjects, bacterial overgrowth in the biliopancreatic limb may generate secondary BAs species with altered affinity for FXR or TGR5 receptors (176). In rodent models, the importance of the FXR receptor in mediating weight loss and metabolic improvements after SG was demonstrated when FXR knockout mice regained lost weight following this procedure (173), although whether FXR signaling and/or FGF-19 contributes to the beneficial effects of bariatric surgery in humans is uncertain at present. Finally, a study that measured serum BAs levels before and after bariatric surgery showed that they were significantly increased only at one-year post‐surgery, whereas, the substantial increase in PYY and GLP-1 levels could be observed as soon as 1-week post-surgery. This finding suggest that increased plasma BAs may be less important in early metabolic improvements observed after bariatric surgery (170) but more so for long-term effects.

 

Alterations in intestinal microbiome following RYGB and SG have been described. Animal studies of fecal transplants from RYGB-treated to germ-free mice showed significantly greater weight loss in the germ-free mice, suggesting that the altered microbiome per se contributes to weight loss (177). RYGB can produce greater and more favorable changes in gut microbiota functional capacity and species than SG despite similar weight loss (178) (179, 180). Although the specific procedure-related mechanisms responsible for post-surgery gut microbiota changes remain to be delineated (181), potential explanations include differences in the physical manipulation of the GI tract and final anatomy, dietary changes, and weight loss differences between procedures. In rodents, these changes can be detected as early as 7 days after RYGB (175), with similar patterns observed in humans (102). Because of significant differences between the rodents and the humans, it is not possible to firmly conclude that gut bacteria are essential for the effects of metabolic procedures. However, it is evident in the rodent models that changes in gut microbiota induced by RYGB are sufficient to produce weight loss (174).

 

Other appetite-related post-surgery effects that may influence weight loss include changes in taste and smell that could, in turn, influence food preference (83). Interestingly, early data suggest that RYGB and SG may differently impact subjective changes in appetite, taste, olfaction and food aversion post-operatively (182). Finally, neurophysiological studies suggest that vagal nerve signaling also increases post-RYGB (157) and these changes may affect food intake in a procedure-specific fashion (183, 184).

 

Developing “Knifeless Surgery”

A multitude of compounds mimicking gut hormone actions are currently under development, opening a new era of pharmacotherapy for obesity. At present, GLP-1 analogues are broadly used in the management of people with T2DM and obesity (185). The longer acting GLP-1 analogue semaglutide has shown promising results for weight loss in early phase studies with both a weekly subcutaneous injection (186) and an oral compound form (187, 188). Intravenous administration of supra-physiological levels of native gut hormones like PYY, GLP-1 and others lead to reduced appetite and decreased energy intake (23, 189-191).  Strategies aimed at reducing acyl-ghrelin and/or increasing des-acyl-ghrelin are also being developed and show promise. The inhibition of GOAT has been shown to reduce energy intake and bodyweight (192).

 

In order to mimic bariatric/metabolic procedures, the effects of combinations of hormones are under investigation with the aim of circumventing compensatory adaptive changes associated with energy restriction. For example, the co-infusion of GLP-1, PYY and OXM induced a 32% reduction in energy intake when compared to placebo (193). Animal models suggest a potential role of CCK as an adjunct to GLP-1 based therapies (194) or monomeric GLP-1/GIP/glucagon triagonism to reduce food intake and obesity (195).

 

CONCLUSION

 

Obesity is a complex disease where genetic, environmental, dietary, psychological and socio-economic factors interact complicating treatments for this life-threatening condition. Peripheral signals such as gut hormones, BAs and gut microbiota inform the CNS regarding the quality and the quantity of any ingested meal and are part of the complex bi-directional communication system known as the gut-brain axis. During the recent years many studies have identified perturbations of this system as a cause of weight gain. Current lifestyle approaches to weight loss lack efficacy because multiple powerful adaptive biological responses promote weight regain. Bariatric surgery, which reduces energy intake as a consequence of favorable gut-brain axis signaling, is currently the most successful treatment for people with severe obesity, leading to marked sustained weight loss and improved health.  Understanding the hidden mechanisms responsible for this success is an exciting area of current research and holds promise to identify novel effective obesity pharmacotherapies.

 

ACKNOWLEDGEMENTS

 

The authors would like to thank Chiara Bullo for the illustrations, Janine Makaronidis and Cormac McGee for their critical review of the manuscript.

 

REFERENCES

 

  1. Lauby-Secretan, B., et al., Body fatness and cancer—viewpoint of the IARC Working Group. New England Journal of Medicine, 2016. 375(8): p. 794-798.
  2. Makaronidis, J.M. and R.L. Batterham, Obesity, body weight regulation and the brain: insights from fMRI. Br J Radiol, 2018: p. 20170910.
  3. Moran, C.P. and F. Shanahan, Gut microbiota and obesity: role in aetiology and potential therapeutic target. Best Pract Res Clin Gastroenterol, 2014. 28(4): p. 585-97.
  4. Bauer, P.V., S.C. Hamr, and F.A. Duca, Regulation of energy balance by a gut-brain axis and involvement of the gut microbiota. Cell Mol Life Sci, 2016. 73(4): p. 737-55.
  5. Miras, A.D. and C.W. le Roux, Mechanisms underlying weight loss after bariatric surgery. Nat Rev Gastroenterol Hepatol, 2013. 10(10): p. 575-84.
  6. Buhmann, H., C.W. le Roux, and M. Bueter, The gut-brain axis in obesity. Best Pract Res Clin Gastroenterol, 2014. 28(4): p. 559-71.
  7. Trends in adult body-mass index in 200 countries from 1975 to 2014: a pooled analysis of 1698 population-based measurement studies with 19.2 million participants. Lancet, 2016. 387(10026): p. 1377-1396.
  8. Afshin, A., et al., Health effects of overweight and obesity in 195 countries over 25 years. The New England journal of medicine, 2017. 377(1): p. 13-27.
  9. Franz, M.J., et al., Weight-loss outcomes: a systematic review and meta-analysis of weight-loss clinical trials with a minimum 1-year follow-up. J Am Diet Assoc, 2007. 107(10): p. 1755-67.
  10. Mann, T., et al., Medicare's search for effective obesity treatments: diets are not the answer. Am Psychol, 2007. 62(3): p. 220-33.
  11. Pilitsi, E., et al., Pharmacotherapy of obesity: Available medications and drugs under investigation. Metabolism, 2018.
  12. Schauer, P.R., et al., Bariatric Surgery versus Intensive Medical Therapy for Diabetes - 5-Year Outcomes. N Engl J Med, 2017. 376(7): p. 641-651.
  13. Pucci, A. and R.L. Batterham, Mechanisms underlying the weight loss effects of RYGB and SG: similar, yet different. J Endocrinol Invest, 2018.
  14. Santo, M.A., et al., Weight Regain After Gastric Bypass: Influence of Gut Hormones. Obes Surg, 2016. 26(5): p. 919-25.
  15. Gribble, F.M. and F. Reimann, Enteroendocrine Cells: Chemosensors in the Intestinal Epithelium. Annu Rev Physiol, 2016. 78: p. 277-99.
  16. Dokken, B.B. and T.-S. Tsao, The Physiology of Body Weight Regulation: Are We Too Efficient for Our Own Good? Diabetes Spectrum, 2007. 20(3): p. 166-170.
  17. Suzuki, K., C.N. Jayasena, and S.R. Bloom, Obesity and appetite control. Exp Diabetes Res, 2012. 2012: p. 824305.
  18. Berthoud, H.R., Metabolic and hedonic drives in the neural control of appetite: who is the boss? Curr Opin Neurobiol, 2011. 21(6): p. 888-96.
  19. Cone, R.D., et al., The arcuate nucleus as a conduit for diverse signals relevant to energy homeostasis. Int J Obes Relat Metab Disord, 2001. 25 Suppl 5: p. S63-7.
  20. Dryden, S., et al., Increased neuropeptide Y secretion in the hypothalamic paraventricular nucleus of obese (fa/fa) Zucker rats. Brain Res, 1995. 690(2): p. 185-8.
  21. Ollmann, M.M., et al., Antagonism of central melanocortin receptors in vitro and in vivo by agouti-related protein. Science, 1997. 278(5335): p. 135-8.
  22. Schwartz, M.W. and G.J. Morton, Obesity: keeping hunger at bay. Nature, 2002. 418(6898): p. 595.
  23. Batterham, R.L., et al., Gut hormone PYY 3-36 physiologically inhibits food intake. Nature, 2002. 418(6898): p. 650.
  24. van der Kooy, D., Area postrema: site where cholecystokinin acts to decrease food intake. Brain Res, 1984. 295(2): p. 345-7.
  25. Grenham, S., et al., Brain-gut-microbe communication in health and disease. Front Physiol, 2011. 2: p. 94.
  26. Mayer, E.A., K. Tillisch, and A. Gupta, Gut/brain axis and the microbiota. J Clin Invest, 2015. 125(3): p. 926-38.
  27. Rehfeld, J.F., A centenary of gastrointestinal endocrinology. Horm Metab Res, 2004. 36(11-12): p. 735-41.
  28. Monje, M., Settling a Nervous Stomach: The Neural Regulation of Enteric Cancer. Cancer Cell, 2017. 31(1): p. 1-2.
  29. Bliss, E.S. and E. Whiteside, The Gut-Brain Axis, the Human Gut Microbiota and Their Integration in the Development of Obesity. Front Physiol, 2018. 9: p. 900.
  30. Dockray, G.J., Enteroendocrine cell signalling via the vagus nerve. Curr Opin Pharmacol, 2013. 13(6): p. 954-8.
  31. Amato, A., et al., Peripheral motor action of glucagon-like peptide-1 through enteric neuronal receptors. Neurogastroenterol Motil, 2010. 22(6): p. 664-e203.
  32. Costa, M., S.J. Brookes, and G.W. Hennig, Anatomy and physiology of the enteric nervous system. Gut, 2000. 47 Suppl 4: p. iv15-9; discussion iv26.
  33. Cooke, A.R. and E.D. Clark, Effect of first part of duodenum on gastric emptying in dogs: response to acid, fat, glucose, and neural blockade. Gastroenterology, 1976. 70(4): p. 550-5.
  34. Hamr, S.C., et al., Does nutrient sensing determine how we "see" food? Curr Diab Rep, 2015. 15(6): p. 604.
  35. Latorre, R., et al., Enteroendocrine cells: a review of their role in brain–gut communication. Neurogastroenterology & Motility, 2016. 28(5): p. 620-630.
  36. Batterham, R.L., et al., Critical role for peptide YY in protein-mediated satiation and body-weight regulation. Cell Metab, 2006. 4(3): p. 223-33.
  37. Elliott, R.M., et al., Glucagon-like peptide-1 (7-36)amide and glucose-dependent insulinotropic polypeptide secretion in response to nutrient ingestion in man: acute post-prandial and 24-h secretion patterns. J Endocrinol, 1993. 138(1): p. 159-66.
  38. Svendsen, B., et al., An analysis of cosecretion and coexpression of gut hormones from male rat proximal and distal small intestine. Endocrinology, 2015. 156(3): p. 847-57.
  39. Oesch, S., et al., Effect of gastric distension prior to eating on food intake and feelings of satiety in humans. Physiol Behav, 2006. 87(5): p. 903-10.
  40. Adam, T.C. and M.S. Westerterp-Plantenga, Glucagon-like peptide-1 release and satiety after a nutrient challenge in normal-weight and obese subjects. Br J Nutr, 2005. 93(6): p. 845-51.
  41. Bliss, E.S. and E. Whiteside, The Gut-Brain Axis, the Human Gut Microbiota and Their Integration in the Development of Obesity. Frontiers in Physiology, 2018. 9(900).
  42. Yamato, E., et al., Tissue-specific and glucose-dependent expression of receptor genes for glucagon and glucagon-like peptide-1 (GLP-1). Horm Metab Res, 1997. 29(2): p. 56-9.
  43. Holst, J.J. and C. Orskov, The incretin approach for diabetes treatment: modulation of islet hormone release by GLP-1 agonism. Diabetes, 2004. 53 Suppl 3: p. S197-204.
  44. Manning, S., A. Pucci, and R.L. Batterham, GLP-1: a mediator of the beneficial metabolic effects of bariatric surgery? Physiology (Bethesda), 2015. 30(1): p. 50-62.
  45. Larsen, P.J., M. Tang-Christensen, and D.S. Jessop, Central administration of glucagon-like peptide-1 activates hypothalamic neuroendocrine neurons in the rat. Endocrinology, 1997. 138(10): p. 4445-55.
  46. Edwards, C.M., et al., Glucagon-like peptide 1 has a physiological role in the control of postprandial glucose in humans: studies with the antagonist exendin 9-39. Diabetes, 1999. 48(1): p. 86-93.
  47. Pucci, A. and N. Finer, New medications for treatment of obesity: metabolic and cardiovascular effects. Can J Cardiol, 2015. 31(2): p. 142-52.
  48. Holst, J.J., The physiology of glucagon-like peptide 1. Physiol Rev, 2007. 87(4): p. 1409-39.
  49. Kuhre, R.E., et al., Measurement of the incretin hormones: glucagon-like peptide-1 and glucose-dependent insulinotropic peptide. J Diabetes Complications, 2015. 29(3): p. 445-50.
  50. Vilsboll, T., et al., Similar elimination rates of glucagon-like peptide-1 in obese type 2 diabetic patients and healthy subjects. J Clin Endocrinol Metab, 2003. 88(1): p. 220-4.
  51. Ruttimann, E.B., et al., Intrameal hepatic portal and intraperitoneal infusions of glucagon-like peptide-1 reduce spontaneous meal size in the rat via different mechanisms. Endocrinology, 2009. 150(3): p. 1174-81.
  52. Graaf, C., et al., Glucagon-Like Peptide-1 and Its Class B G Protein-Coupled Receptors: A Long March to Therapeutic Successes. Pharmacol Rev, 2016. 68(4): p. 954-1013.
  53. Baggio, L.L., et al., Oxyntomodulin and glucagon-like peptide-1 differentially regulate murine food intake and energy expenditure. Gastroenterology, 2004. 127(2): p. 546-58.
  54. Cohen, M.A., et al., Oxyntomodulin suppresses appetite and reduces food intake in humans. J Clin Endocrinol Metab, 2003. 88(10): p. 4696-701.
  55. Maida, A., et al., The glucagon-like peptide-1 receptor agonist oxyntomodulin enhances beta-cell function but does not inhibit gastric emptying in mice. Endocrinology, 2008. 149(11): p. 5670-8.
  56. Pocai, A., Action and therapeutic potential of oxyntomodulin. Mol Metab, 2014. 3(3): p. 241-51.
  57. Druce, M.R., et al., Investigation of structure-activity relationships of Oxyntomodulin (Oxm) using Oxm analogs. Endocrinology, 2009. 150(4): p. 1712-22.
  58. Batterham, R.L., et al., PYY modulation of cortical and hypothalamic brain areas predicts feeding behaviour in humans. Nature, 2007. 450(7166): p. 106-9.
  59. Manning, S. and R.L. Batterham, The role of gut hormone peptide YY in energy and glucose homeostasis: twelve years on. Annu Rev Physiol, 2014. 76: p. 585-608.
  60. le Roux, C.W., et al., Attenuated peptide YY release in obese subjects is associated with reduced satiety. Endocrinology, 2006. 147(1): p. 3-8.
  61. Batterham, R.L., et al., Inhibition of food intake in obese subjects by peptide YY3-36. N Engl J Med, 2003. 349(10): p. 941-8.
  62. Broberger, C., et al., Subtypes Y1 and Y2 of the neuropeptide Y receptor are respectively expressed in pro-opiomelanocortin- and neuropeptide-Y-containing neurons of the rat hypothalamic arcuate nucleus. Neuroendocrinology, 1997. 66(6): p. 393-408.
  63. Di Francesco, V., et al., Delayed postprandial gastric emptying and impaired gallbladder contraction together with elevated cholecystokinin and peptide YY serum levels sustain satiety and inhibit hunger in healthy elderly persons. J Gerontol A Biol Sci Med Sci, 2005. 60(12): p. 1581-5.
  64. El-Salhy, M., et al., The role of peptide YY in gastrointestinal diseases and disorders (review). Int J Mol Med, 2013. 31(2): p. 275-82.
  65. Pucci, A., et al., A case of severe anorexia, excessive weight loss and high peptide YY levels after sleeve gastrectomy. Endocrinol Diabetes Metab Case Rep, 2015. 2015: p. 150020.
  66. Khandekar, N., et al., The role of pancreatic polypeptide in the regulation of energy homeostasis. Mol Cell Endocrinol, 2015. 418 Pt 1: p. 33-41.
  67. Adrian, T.E., et al., Distribution and release of human pancreatic polypeptide. Gut, 1976. 17(12): p. 940-44.
  68. Teff, K., Nutritional implications of the cephalic-phase reflexes: endocrine responses. Appetite, 2000. 34(2): p. 206-13.
  69. Lin, S., et al., Critical role of arcuate Y4 receptors and the melanocortin system in pancreatic polypeptide-induced reduction in food intake in mice. PLoS One, 2009. 4(12): p. e8488.
  70. Parker, R.M. and H. Herzog, Regional distribution of Y-receptor subtype mRNAs in rat brain. Eur J Neurosci, 1999. 11(4): p. 1431-48.
  71. Batterham, R.L., et al., Pancreatic polypeptide reduces appetite and food intake in humans. J Clin Endocrinol Metab, 2003. 88(8): p. 3989-92.
  72. Berntson, G.G., et al., Pancreatic polypeptide infusions reduce food intake in Prader-Willi syndrome. Peptides, 1993. 14(3): p. 497-503.
  73. Gibbs, J., R.C. Young, and G.P. Smith, Cholecystokinin elicits satiety in rats with open gastric fistulas. Nature, 1973. 245(5424): p. 323-5.
  74. Lieverse, R.J., et al., Satiety effects of a physiological dose of cholecystokinin in humans. Gut, 1995. 36(2): p. 176-9.
  75. Duca, F.A. and J.T. Yue, Fatty acid sensing in the gut and the hypothalamus: in vivo and in vitro perspectives. Mol Cell Endocrinol, 2014. 397(1-2): p. 23-33.
  76. Castillo, E.J., et al., Effect of oral CCK-1 agonist GI181771X on fasting and postprandial gastric functions in healthy volunteers. Am J Physiol Gastrointest Liver Physiol, 2004. 287(2): p. G363-9.
  77. Muller, T.D., et al., Ghrelin. Mol Metab, 2015. 4(6): p. 437-60.
  78. Cummings, D.E., et al., A preprandial rise in plasma ghrelin levels suggests a role in meal initiation in humans. (0012-1797 (Print)).
  79. Neary, N.M., et al., Ghrelin increases energy intake in cancer patients with impaired appetite: acute, randomized, placebo-controlled trial. J Clin Endocrinol Metab, 2004. 89(6): p. 2832-6.
  80. Cummings, D.E., et al., Plasma ghrelin levels and hunger scores in humans initiating meals voluntarily without time- and food-related cues. (0193-1849 (Print)).
  81. Cummings, D.E., et al., Plasma ghrelin levels after diet-induced weight loss or gastric bypass surgery. (1533-4406 (Electronic)).
  82. Callahan, H.S., et al., Postprandial suppression of plasma ghrelin level is proportional to ingested caloric load but does not predict intermeal interval in humans. J Clin Endocrinol Metab, 2004. 89(3): p. 1319-24.
  83. Makaronidis, J.M., et al., Reported appetite, taste and smell changes following Roux-en-Y gastric bypass and sleeve gastrectomy: Effect of gender, type 2 diabetes and relationship to post-operative weight loss. Appetite, 2016. 107: p. 93-105.
  84. Mackie, K., Cannabinoid receptors: where they are and what they do. J Neuroendocrinol, 2008. 20 Suppl 1: p. 10-4.
  85. Aronne, L.J., et al., A clinical trial assessing the safety and efficacy of taranabant, a CB1R inverse agonist, in obese and overweight patients: a high-dose study. Int J Obes (Lond), 2010. 34(5): p. 919-35.
  86. Topol, E.J., et al., Rimonabant for prevention of cardiovascular events (CRESCENDO): a randomised, multicentre, placebo-controlled trial. Lancet, 2010. 376(9740): p. 517-23.
  87. Seeley, R.J., A.P. Chambers, and D.A. Sandoval, The role of gut adaptation in the potent effects of multiple bariatric surgeries on obesity and diabetes. Cell Metab, 2015. 21(3): p. 369-78.
  88. Schaap, F.G., M. Trauner, and P.L. Jansen, Bile acid receptors as targets for drug development. Nat Rev Gastroenterol Hepatol, 2014. 11(1): p. 55-67.
  89. Thomas, C., et al., TGR5-mediated bile acid sensing controls glucose homeostasis. Cell Metab, 2009. 10(3): p. 167-77.
  90. Watanabe, M., et al., Bile acids induce energy expenditure by promoting intracellular thyroid hormone activation. Nature, 2006. 439(7075): p. 484-9.
  91. Dufer, M., et al., Bile acids acutely stimulate insulin secretion of mouse beta-cells via farnesoid X receptor activation and K(ATP) channel inhibition. Diabetes, 2012. 61(6): p. 1479-89.
  92. Lundasen, T., et al., Circulating intestinal fibroblast growth factor 19 has a pronounced diurnal variation and modulates hepatic bile acid synthesis in man. J Intern Med, 2006. 260(6): p. 530-6.
  93. Fang, S., et al., Intestinal FXR agonism promotes adipose tissue browning and reduces obesity and insulin resistance. Nat Med, 2015. 21(2): p. 159-65.
  94. Suzuki, T., et al., Correlation between postprandial bile acids and body fat mass in healthy normal-weight subjects. Clin Biochem, 2014. 47(12): p. 1128-31.
  95. Turnbaugh, P.J., et al., The human microbiome project. Nature, 2007. 449(7164): p. 804-10.
  96. Sender, R., S. Fuchs, and R. Milo, Revised Estimates for the Number of Human and Bacteria Cells in the Body. PLoS Biol, 2016. 14(8): p. e1002533.
  97. Wang, H.X. and Y.P. Wang, Gut Microbiota-brain Axis. Chin Med J (Engl), 2016. 129(19): p. 2373-80.
  98. Qin, J., et al., A human gut microbial gene catalogue established by metagenomic sequencing. Nature, 2010. 464(7285): p. 59-65.
  99. Martinez, K.B., J.F. Pierre, and E.B. Chang, The Gut Microbiota: The Gateway to Improved Metabolism. Gastroenterol Clin North Am, 2016. 45(4): p. 601-614.
  100. Wostmann, B.S., et al., Dietary intake, energy metabolism, and excretory losses of adult male germfree Wistar rats. Lab Anim Sci, 1983. 33(1): p. 46-50.
  101. De Filippo, C., et al., Impact of diet in shaping gut microbiota revealed by a comparative study in children from Europe and rural Africa. Proc Natl Acad Sci U S A, 2010. 107(33): p. 14691-6.
  102. Furet, J.P., et al., Differential adaptation of human gut microbiota to bariatric surgery-induced weight loss: links with metabolic and low-grade inflammation markers. Diabetes, 2010. 59(12): p. 3049-57.
  103. Schwiertz, A., et al., Microbiota and SCFA in lean and overweight healthy subjects. Obesity (Silver Spring), 2010. 18(1): p. 190-5.
  104. Bomhof, M.R., et al., Combined effects of oligofructose and Bifidobacterium animalis on gut microbiota and glycemia in obese rats. Obesity (Silver Spring), 2014. 22(3): p. 763-71.
  105. Lin, H.V., et al., Butyrate and propionate protect against diet-induced obesity and regulate gut hormones via free fatty acid receptor 3-independent mechanisms. PLoS One, 2012. 7(4): p. e35240.
  106. Pan, X.D., et al., Prebiotic oligosaccharides change the concentrations of short-chain fatty acids and the microbial population of mouse bowel. J Zhejiang Univ Sci B, 2009. 10(4): p. 258-63.
  107. Kasubuchi, M., et al., Dietary gut microbial metabolites, short-chain fatty acids, and host metabolic regulation. Nutrients, 2015. 7(4): p. 2839-49.
  108. Nohr, M.K., et al., Expression of the short chain fatty acid receptor GPR41/FFAR3 in autonomic and somatic sensory ganglia. Neuroscience, 2015. 290: p. 126-37.
  109. Muccioli, G.G., et al., The endocannabinoid system links gut microbiota to adipogenesis. Mol Syst Biol, 2010. 6: p. 392.
  110. Backhed, F., et al., The gut microbiota as an environmental factor that regulates fat storage. Proc Natl Acad Sci U S A, 2004. 101(44): p. 15718-23.
  111. Cani, P.D., et al., Metabolic endotoxemia initiates obesity and insulin resistance. Diabetes, 2007. 56(7): p. 1761-72.
  112. de La Serre, C.B., et al., Propensity to high-fat diet-induced obesity in rats is associated with changes in the gut microbiota and gut inflammation. Am J Physiol Gastrointest Liver Physiol, 2010. 299(2): p. G440-8.
  113. Caesar, R., et al., Crosstalk between Gut Microbiota and Dietary Lipids Aggravates WAT Inflammation through TLR Signaling. Cell Metab, 2015. 22(4): p. 658-68.
  114. Vaure, C. and Y. Liu, A comparative review of toll-like receptor 4 expression and functionality in different animal species. Front Immunol, 2014. 5: p. 316.
  115. Zuo, D.C., et al., Inhibition of pacemaker activity in interstitial cells of Cajal by LPS via NF-kappaB and MAP kinase. World J Gastroenterol, 2013. 19(8): p. 1210-8.
  116. Schele, E., et al., The gut microbiota reduces leptin sensitivity and the expression of the obesity-suppressing neuropeptides proglucagon (Gcg) and brain-derived neurotrophic factor (Bdnf) in the central nervous system. Endocrinology, 2013. 154(10): p. 3643-51.
  117. Schele, E., et al., Regulation of body fat mass by the gut microbiota: Possible mediation by the brain. Peptides, 2016. 77: p. 54-9.
  118. Rosenbaum, M. and R.L. Leibel, 20 years of leptin: role of leptin in energy homeostasis in humans. J Endocrinol, 2014. 223(1): p. T83-96.
  119. De Silva, A., et al., The gut hormones PYY3-36 and GLP-17-36 amide reduce food intake and modulate brain activity in appetite centers in humans. Cell metabolism, 2011. 14(5): p. 700-706.
  120. Schmidt, J.B., et al., Effects of PYY3–36 and GLP-1 on energy intake, energy expenditure, and appetite in overweight men. American Journal of Physiology-Endocrinology and Metabolism, 2014. 306(11): p. E1248-E1256.
  121. De Silva, A., et al., The gut hormones PYY 3-36 and GLP-1 7-36 amide reduce food intake and modulate brain activity in appetite centers in humans. Cell Metab, 2011. 14(5): p. 700-6.
  122. Neary, N.M., et al., Peptide YY3-36 and glucagon-like peptide-17-36 inhibit food intake additively. Endocrinology, 2005. 146(12): p. 5120-7.
  123. Field, B.C., et al., PYY3-36 and oxyntomodulin can be additive in their effect on food intake in overweight and obese humans. Diabetes, 2010. 59(7): p. 1635-9.
  124. Panaro, B.L., et al., The melanocortin-4 receptor is expressed in enteroendocrine L cells and regulates the release of peptide YY and glucagon-like peptide 1 in vivo. Cell metabolism, 2014. 20(6): p. 1018-1029.
  125. Hayes, M.R. and H.D. Schmidt, GLP-1 influences food and drug reward. Current Opinion in Behavioral Sciences, 2016. 9: p. 66-70.
  126. Kroemer, N.B., et al., Fasting levels of ghrelin covary with the brain response to food pictures. Addict Biol, 2013. 18(5): p. 855-62.
  127. Cummings, D.E., Taste and the regulation of food intake: it's not just about flavor. Am J Clin Nutr, 2015. 102(4): p. 717-8.
  128. Zolotukhin, S., Metabolic hormones in saliva: origins and functions. Oral diseases, 2013. 19(3): p. 219-229.
  129. Blouet, C. and G.J. Schwartz, Duodenal lipid sensing activates vagal afferents to regulate non-shivering brown fat thermogenesis in rats. PLoS One, 2012. 7(12): p. e51898.
  130. Piaggi, P., et al., Energy expenditure in the etiology of human obesity: spendthrift and thrifty metabolic phenotypes and energy-sensing mechanisms. J Endocrinol Invest, 2018. 41(1): p. 83-89.
  131. Lean, M.E. and D. Malkova, Altered gut and adipose tissue hormones in overweight and obese individuals: cause or consequence? Int J Obes (Lond), 2016. 40(4): p. 622-32.
  132. Steinert, R.E., et al., Ghrelin, CCK, GLP-1, and PYY (3–36): Secretory controls and physiological roles in eating and glycemia in health, obesity, and after RYGB. Physiological reviews, 2016. 97(1): p. 411-463.
  133. Auguet, T., et al., Low Circulating Levels of Neurotensin in Women with Nonalcoholic Fatty Liver Disease Associated with Severe Obesity. Obesity (Silver Spring), 2018. 26(2): p. 274-278.
  134. Moghadam, A.A., T.H. Moran, and M.J. Dailey, Alterations in circadian and meal-induced gut peptide levels in lean and obese rats. Exp Biol Med (Maywood), 2017. 242(18): p. 1786-1794.
  135. Francois, M., et al., High-fat diet increases ghrelin-expressing cells in stomach, contributing to obesity. Nutrition, 2016. 32(6): p. 709-15.
  136. Briggs, D.I., et al., Diet-Induced Obesity Causes Ghrelin Resistance in Arcuate NPY/AgRP Neurons. Endocrinology, 2010. 151(10): p. 4745-4755.
  137. Wölnerhanssen, B.K., et al., Deregulation of transcription factors controlling intestinal epithelial cell differentiation; a predisposing factor for reduced enteroendocrine cell number in morbidly obese individuals. Scientific reports, 2017. 7(1): p. 8174.
  138. Chavez-Talavera, O., et al., Bile Acid Control of Metabolism and Inflammation in Obesity, Type 2 Diabetes, Dyslipidemia, and Nonalcoholic Fatty Liver Disease. Gastroenterology, 2017. 152(7): p. 1679-1694.e3.
  139. Behary, P., et al., Combined GLP-1, Oxyntomodulin, and Peptide YY Improves Body Weight and Glycemia in Obesity and Prediabetes/Type 2 Diabetes: A Randomized, Single-Blinded, Placebo-Controlled Study. Diabetes Care, 2019. 42(8): p. 1446-1453.
  140. Aron-Wisnewsky, J. and K. Clement, The effects of gastrointestinal surgery on gut microbiota: potential contribution to improved insulin sensitivity. Curr Atheroscler Rep, 2014. 16(11): p. 454.
  141. Turnbaugh, P.J., et al., A core gut microbiome in obese and lean twins. Nature, 2009. 457(7228): p. 480-4.
  142. Turnbaugh, P.J., et al., The effect of diet on the human gut microbiome: a metagenomic analysis in humanized gnotobiotic mice. Sci Transl Med, 2009. 1(6): p. 6ra14.
  143. Aron-Wisnewsky, J., K. Clement, and M. Nieuwdorp, Fecal Microbiota Transplantation: a Future Therapeutic Option for Obesity/Diabetes? Curr Diab Rep, 2019. 19(8): p. 51.
  144. Tambo, A., M.H. Roshan, and N.P. Pace, The Microbial Hypothesis: Contributions of Adenovirus Infection and Metabolic Endotoxaemia to the Pathogenesis of Obesity. Int J Chronic Dis, 2016. 2016: p. 7030795.
  145. Turnbaugh, P.J., et al., An obesity-associated gut microbiome with increased capacity for energy harvest. Nature, 2006. 444(7122): p. 1027-31.
  146. Barkeling, B., et al., Characterization of obese individuals who claim to detect no relationship between their eating pattern and sensations of hunger or fullness. Int J Obes (Lond), 2007. 31(3): p. 435-9.
  147. Sumithran, P., et al., Long-term persistence of hormonal adaptations to weight loss. N Engl J Med, 2011. 365(17): p. 1597-604.
  148. Gloy, V.L., et al., Bariatric surgery versus non-surgical treatment for obesity: a systematic review and meta-analysis of randomised controlled trials. Bmj, 2013. 347: p. f5934.
  149. Sjostrom, L., Review of the key results from the Swedish Obese Subjects (SOS) trial - a prospective controlled intervention study of bariatric surgery. J Intern Med, 2013. 273(3): p. 219-34.
  150. Radaelli, M.G., et al., NAFLD/NASH in patients with type 2 diabetes and related treatment options. J Endocrinol Invest, 2017.
  151. Angrisani, L., et al., Bariatric Surgery and Endoluminal Procedures: IFSO Worldwide Survey 2014. Obes Surg, 2017. 27(9): p. 2279-2289.
  152. Manning, S., et al., Early postoperative weight loss predicts maximal weight loss after sleeve gastrectomy and Roux-en-Y gastric bypass. Surg Endosc, 2015. 29(6): p. 1484-91.
  153. Pucci, A., et al., Type 2 diabetes remission 2 years post Roux-en-Y gastric bypass and sleeve gastrectomy: the role of the weight loss and comparison of DiaRem and DiaBetter scores. Diabet Med, 2018. 35(3): p. 360-367.
  154. Olbers, T., et al., Laparoscopic gastric bypass: development of technique, respiratory function, and long-term outcome. Obes Surg, 2003. 13(3): p. 364-70.
  155. Abu-Jaish, W. and R.J. Rosenthal, Sleeve gastrectomy: a new surgical approach for morbid obesity. Expert Rev Gastroenterol Hepatol, 2010. 4(1): p. 101-19.
  156. Leibel, R.L. and J. Hirsch, Diminished energy requirements in reduced-obese patients. Metabolism, 1984. 33(2): p. 164-70.
  157. Browning, K.N., S.R. Fortna, and A. Hajnal, Roux-en-Y gastric bypass reverses the effects of diet-induced obesity to inhibit the responsiveness of central vagal motoneurones. J Physiol, 2013. 591(9): p. 2357-72.
  158. Manning, S., A. Pucci, and R.L. Batterham, Roux-en-Y gastric bypass: effects on feeding behavior and underlying mechanisms. J Clin Invest, 2015. 125(3): p. 939-48.
  159. Leibel, R.L., et al., Biologic Responses to Weight Loss and Weight Regain: Report From an American Diabetes Association Research Symposium. Diabetes, 2015. 64(7): p. 2299-309.
  160. Ionut, V. and R.N. Bergman, Mechanisms responsible for excess weight loss after bariatric surgery. J Diabetes Sci Technol, 2011. 5(5): p. 1263-82.
  161. Odstrcil, E.A., et al., The contribution of malabsorption to the reduction in net energy absorption after long-limb Roux-en-Y gastric bypass. Am J Clin Nutr, 2010. 92(4): p. 704-13.
  162. Yousseif, A., et al., Differential effects of laparoscopic sleeve gastrectomy and laparoscopic gastric bypass on appetite, circulating acyl-ghrelin, peptide YY3-36 and active GLP-1 levels in non-diabetic humans. Obesity surgery, 2014. 24(2): p. 241-252.
  163. Peterli, R., et al., Metabolic and hormonal changes after laparoscopic Roux-en-Y gastric bypass and sleeve gastrectomy: a randomized, prospective trial. Obes Surg, 2012. 22(5): p. 740-8.
  164. Yousseif, A., et al., Differential effects of laparoscopic sleeve gastrectomy and laparoscopic gastric bypass on appetite, circulating acyl-ghrelin, peptide YY3-36 and active GLP-1 levels in non-diabetic humans. Obes Surg, 2014. 24(2): p. 241-52.
  165. le Roux, C.W., et al., Gut hormones as mediators of appetite and weight loss after Roux-en-Y gastric bypass. Annals of surgery, 2007. 246(5): p. 780-785.
  166. Jirapinyo, P., et al., A Meta-Analysis of GLP-1 After Roux-En-Y Gastric Bypass: Impact of Surgical Technique and Measurement Strategy. Obes Surg, 2017.
  167. Scott, W.R. and R.L. Batterham, Roux-en-Y gastric bypass and laparoscopic sleeve gastrectomy: understanding weight loss and improvements in type 2 diabetes after bariatric surgery. Am J Physiol Regul Integr Comp Physiol, 2011. 301(1): p. R15-27.
  168. Dirksen, C., et al., Gut hormones, early dumping and resting energy expenditure in patients with good and poor weight loss response after Roux-en-Y gastric bypass. International journal of obesity, 2013. 37(11): p. 1452-1460.
  169. Svane, M.S., et al., Peptide YY and glucagon-like peptide-1 contribute to decreased food intake after Roux-en-Y gastric bypass surgery. Int J Obes (Lond), 2016. 40(11): p. 1699-1706.
  170. Steinert, R.E., et al., Bile acids and gut peptide secretion after bariatric surgery: a 1-year prospective randomized pilot trial. Obesity (Silver Spring), 2013. 21(12): p. E660-8.
  171. Pournaras, D.J., et al., The role of bile after Roux-en-Y gastric bypass in promoting weight loss and improving glycaemic control. Endocrinology, 2012. 153(8): p. 3613-9.
  172. Ahmad, N.N., A. Pfalzer, and L.M. Kaplan, Roux-en-Y gastric bypass normalizes the blunted postprandial bile acid excursion associated with obesity. Int J Obes (Lond), 2013. 37(12): p. 1553-9.
  173. Ryan, K.K., et al., FXR is a molecular target for the effects of vertical sleeve gastrectomy. Nature, 2014. 509(7499): p. 183-8.
  174. Liu, H., et al., Role of gut microbiota, bile acids and their cross-talk in the effects of bariatric surgery on obesity and type 2 diabetes. J Diabetes Investig, 2018. 9(1): p. 13-20.
  175. Liou, A.P., et al., Conserved shifts in the gut microbiota due to gastric bypass reduce host weight and adiposity. Sci Transl Med, 2013. 5(178): p. 178ra41.
  176. Midtvedt, T., A. Norman, and K. Nygaard, Bile acid transforming micro-organisms in rats with an intestinal blind segment. Acta Pathol Microbiol Scand, 1969. 77(1): p. 162-6.
  177. Tremaroli, V., et al., Roux-en-Y Gastric Bypass and Vertical Banded Gastroplasty Induce Long-Term Changes on the Human Gut Microbiome Contributing to Fat Mass Regulation. Cell Metab, 2015. 22(2): p. 228-38.
  178. Murphy, R., et al., Differential Changes in Gut Microbiota After Gastric Bypass and Sleeve Gastrectomy Bariatric Surgery Vary According to Diabetes Remission. Obes Surg, 2017. 27(4): p. 917-925.
  179. Medina, D.A., et al., Distinct patterns in the gut microbiota after surgical or medical therapy in obese patients. PeerJ, 2017. 5: p. e3443.
  180. Guo, Y., et al., Gut microbiota after Roux-en-Y gastric bypass and sleeve gastrectomy in a diabetic rat model: Increased diversity and associations of discriminant genera with metabolic changes. Diabetes Metab Res Rev, 2017. 33(3).
  181. Dethlefsen, L. and D.A. Relman, Incomplete recovery and individualized responses of the human distal gut microbiota to repeated antibiotic perturbation. Proc Natl Acad Sci U S A, 2011. 108 Suppl 1: p. 4554-61.
  182. Zakeri, R. and R.L. Batterham, Potential mechanisms underlying the effect of bariatric surgery on eating behaviour. Curr Opin Endocrinol Diabetes Obes, 2018. 25(1): p. 3-11.
  183. Berthoud, H.R., Vagal and hormonal gut-brain communication: from satiation to satisfaction. Neurogastroenterol Motil, 2008. 20 Suppl 1: p. 64-72.
  184. Berthoud, H.R., A.C. Shin, and H. Zheng, Obesity surgery and gut-brain communication. Physiol Behav, 2011. 105(1): p. 106-19.
  185. Pi-Sunyer, X., et al., A Randomized, Controlled Trial of 3.0 mg of Liraglutide in Weight Management. New England Journal of Medicine, 2015. 373(1): p. 11-22.
  186. Marso, S.P., et al., Semaglutide and Cardiovascular Outcomes in Patients with Type 2 Diabetes. N Engl J Med, 2016. 375(19): p. 1834-1844.
  187. Husain, M., et al., Oral Semaglutide and Cardiovascular Outcomes in Patients with Type 2 Diabetes. N Engl J Med, 2019. 381(9): p. 841-851.
  188. Aroda, V.R., et al., PIONEER 1: Randomized Clinical Trial of the Efficacy and Safety of Oral Semaglutide Monotherapy in Comparison With Placebo in Patients With Type 2 Diabetes. Diabetes Care, 2019. 42(9): p. 1724-1732.
  189. Bagger, J.I., et al., Effect of oxyntomodulin, glucagon, GLP-1, and combined glucagon+ GLP-1 infusion on food intake, appetite, and resting energy expenditure. The Journal of Clinical Endocrinology & Metabolism, 2015. 100(12): p. 4541-4552.
  190. Wynne, K., et al., Oxyntomodulin increases energy expenditure in addition to decreasing energy intake in overweight and obese humans: a randomised controlled trial. International journal of obesity, 2006. 30(12): p. 1729.
  191. Reda, T.K., A. Geliebter, and F.X. Pi‐Sunyer, Amylin, food intake, and obesity. Obesity research, 2002. 10(10): p. 1087-1091.
  192. Allas, S., et al., AZP-531, an unacylated ghrelin analog, improves food-related behavior in patients with Prader-Willi syndrome: A randomized placebo-controlled trial. PloS one, 2018. 13(1): p. e0190849.
  193. Tan, T., et al., The effect of a subcutaneous infusion of GLP-1, OXM, and PYY on energy intake and expenditure in obese volunteers. The Journal of Clinical Endocrinology & Metabolism, 2017. 102(7): p. 2364-2372.
  194. Pathak, V., P.R. Flatt, and N. Irwin, Cholecystokinin (CCK) and related adjunct peptide therapies for the treatment of obesity and type 2 diabetes. Peptides, 2018. 100: p. 229-235.
  195. Finan, B., et al., A rationally designed monomeric peptide triagonist corrects obesity and diabetes in rodents. Nature medicine, 2015. 21(1): p. 27.

Ultrasonography of the Thyroid

ABSTRACT

Thyroid ultrasonography (US) is the most common, extremely useful, safe, and cost-effective way to image the thyroid gland and its pathology. US has largely replaced the need for thyroid scintigraphy except to detect iodine-avid thyroid metastases after thyroidectomy and to identify hyper-functioning (toxic) thyroid nodules. This chapter reviews the literature; discusses the science and method of performing US; examines it’s clinical utility to assess thyroid goiters, nodules, cancers, post-operative remnants, cervical lymph nodes, and metastases; presents it’s practical value to enhance US-guided aspiration biopsy of thyroid lesions (FNA); and endorses it’s importance in medical education. US reveals, with good sensitivity but only fair specificity very important and diagnostically useful clues to the clinician and surgeon about the likelihood that a thyroid nodule is malignant. Color flow Doppler enhancement of the US images, that delineates the vasculature, is essential. Comprehensive understanding of the local anatomy, the specific disease process, technical skill and experience are essential for proper interpretation of the US images. Features that favor the presence of a malignant nodule include decreased echogenicity, microcalcifications, central hypervascularity, irregular margins, an incomplete halo, a tall rather than wide shape (larger in the anteroposterior axis compared to the horizontal axis, the nodule is growing in one direction and not growing concentrically), documented enlargement of the solid portion of the nodule and associated lymphadenopathy. Several of these attributes enhance the diagnostic probability. The patient’s history, physical examination, and comorbidities refine the diagnosis. FNA and cytological examination of thyroid nodules and adenopathy in adults, children, and adolescents has become a major, specific, and highly diagnostic tool that is safe and inexpensive. In addition, the aspirate may be analyzed by biochemical measurements and especially by evolving molecular genetic methods.

 

INTRODUCTION

 

Ultrasonography (US) is the most common and most useful way to image the thyroid gland and its pathology, as recognized in guidelines for managing thyroid disorders published by the American Thyroid Association (1) and other authoritative bodies. In addition to facilitating the diagnosis of clinically apparent nodules, the widespread use of US has resulted in uncovering a multitude of clinically imperceptible thyroid nodules, the overwhelming majority of which are benign. The high sensitivity for nodules but inadequate specificity for cancer has posed a management and economic problem. This chapter will address the method and utility of clinically effective thyroid US to assess the likelihood of cancer, to enhance fine needle aspiration biopsy and cytology (FNA), to facilitate other thyroid diagnoses, and to teach thyroidology.

 

Previously, imaging of the thyroid required scintiscanning to provide a map of those areas of the thyroid that accumulate and process radioactive iodine or other nucleotides. The major premise of thyroid scanning was that thyroid cancers concentrate less radioactive iodine than healthy tissue and therefore provided triage in the selection for thyroid surgery. Unfortunately, however, since benign nodules also concentrate radioactive iodine poorly, the selection process is too inefficient to be cost-effective. Although, scintiscanning remains of primary importance in patients who are hyperthyroid or for detection of iodine-avid tissue after thyroidectomy for thyroid cancer, US has largely replaced nuclear scanning for the majority of patients because of its higher resolution, superior correlation of true thyroid dimensions with the image, smaller expense, greater simplicity, and lack of need for radioisotope administration. The other imaging methods, computerized tomography (CT), magnetic resonance imaging (MRI), and 18F-FDG positron emission tomography (PET) are more costly than US, are not as efficient in detecting small lesions, and are best used selectively when US is inadequate to elucidate a clinical problem (2,3).

 

 

Medical testing must be cost-effective. There is documentation that in a hospital or emergency department setting, the expense of thyroid ultrasound is quite low (5).

 

Although sonography can supply very important and clinically useful clues about the nature of a thyroid lesion, it does not reliably differentiate benign lesions and cancer. However, it can help significantly. US can:

 

  1. Depict accurately the anatomy of the neck in the thyroid region,
  2. Help the student and clinician to learn thyroid palpation,
  3. Elucidate cryptic findings on physical examination,
  4. Assess the comparative size of nodules, lymph nodes, or goiters in patients who are under observation or therapy,
  5. Detect a non-palpable thyroid lesion in a patient who was exposed to therapeutic irradiation,
  6. Give very important and clinically useful clues about the likelihood of malignancy,
  7. Identify the solid component of a complex nodule,
  8. Guide and facilitate fine needle aspiration biopsy of a nodule,
  9. Evaluate for recurrence of a thyroid mass after surgery,
  10. Monitor thyroid cancer patients for residual disease or early evidence of reappearance of malignancy in the thyroid bed or lymphadenopathy,
  11. Identify patients who have ultrasonic thyroid patterns that suggest diagnoses such as thyroiditis.
  12. Perhaps refine the management of patients on therapy such as antithyroid drugs,
  13. Facilitate delivery of medication or physical high-energy therapy precisely into a lesion and spare the surrounding tissue,
  14. Monitor in utero the fetal thyroid for size, ultrasonic texture, and vascularity,
  15. Scrutinize the neonatal thyroid for size and location,
  16. Screen the thyroid during epidemiologic investigation in the field.

 

TECHNICAL ASPECTS

 

Sonography depicts the internal structure of the thyroid gland and the regional anatomy and pathology without using ionizing radiation or iodine containing contrast medium (6,7). Rather, high frequency sound waves in the megahertz range (ultrasound), are used to produce an image. The procedure is safe, does not cause damage to tissue and is less costly than any other imaging procedure. The patient remains comfortable during the test, which typically takes only a few minutes unless there is a need to evaluate the lateral neck, does not require discontinuation of any medication, or preparation of the patient. The procedure is usually done with the patient reclining with the neck hyperextended but it can be done in the seated position. A probe that contains a piezoelectric crystal called a transducer is applied to the neck but since air does not transmit ultrasound, it must be coupled to the skin with a liquid medium or a gel. This instrument rapidly alternates as the generator of the ultrasound and the receiver of the signal that has been reflected by internal tissues. The signal is organized electronically into numerous shades of gray and is processed electronically to produce an image instantaneously (real-time). Although each image is a static picture, rapid sequential frames are processed electronically to depict motion. Two-dimensional images have been standard and 3-dimentional images are an improvement in certain circumstances (8). There is considerable potential for improving ultrasound images of the thyroid by using ultrasound contrast agents. These experimental materials include gas-filled micro-bubbles with a mean diameter less than that of a red blood corpuscle and Levovist, an agent consisting of granules that are composed of 99.9% galactose and 0.1% palmitic acid. They are injected intravenously, enhance the echogenicity of the blood, and increase the signal to noise ratio (9,10). Contrast-enhanced thyroid US features such as heterogeneous enhancement, slow ”wash in”, ill-defined enhancement of the border a the nodule, and fast “wash out” seem to be associated with increased association with malignancy (11).

 

Dynamic information such as blood flow can be added to the standard US signal by employing a physics principle called the Doppler effect. The frequency of a sound wave increases when it approaches a listener (the ear or, in the case of ultrasonography, a transducer) and decreases as it departs. The Doppler signals, which are superimposed on real time gray scale images, are extremely bright in black and white images and may be color coded to reveal the velocity (frequency shift) and direction of blood flow (phase shift) as well as the degree of vascularity of an organ (12,13). Flow in one direction is made red and in the opposite direction, blue. The shade and intensity of color can correlate with the velocity of flow. Thus, in general terms, venous and arterial flow can be depicted by assuming that flow in these two kinds of blood vessels is parallel, but in opposite directions. Since portions of blood vessels may be tortuous, modifying orientation to the probe, different colors are displayed within the same blood vessel even if the true direction of blood flow has not changed. Thus, an analysis of flow characteristics requires careful observations and cautious interpretations. The absence of flow in a fluid-filled structure can differentiate a cystic structure and a blood vessel.

 

Blood flow within anatomic structures can also be depicted by non-Doppler technology. This technique is called B-flow ultrasonic imaging (BFI). It is accomplished by transmitting precisely separated adjacent ultrasound beams and analyzing with a computer, the reflected echo pairs (14).

 

Various anatomic features and tissues result in different ultrasound characteristics (2,6). The air-filled trachea does not transmit the ultrasound. Calcified tissues such as bone and sometimes cartilage and calcific deposits in other anatomic structures block the passage of ultrasound resulting in a very bright signal and a linear echo-free shadow distally. Most tissues transmit the ultrasound to varying degrees and interfaces between tissues reflect portions of the sound waves. Fluid-filled structures have a uniform echo-free appearance whereas fleshy structures and organs have a ground glass appearance that may be uniform or heterogeneous depending on the characteristics of the structure.

 

The depth penetration and resolving power of ultrasound depends greatly on frequency (7). Depth penetration is inversely related and spatial resolution is directly related to the frequency of the ultrasound. For thyroid, a frequency of 7.5 to 10 - 15 megahertz is generally optimal for all but the largest goiters. Using these frequencies, nodules as small as two to three millimeters can be identified.

 

Performance and interpretation of thyroid sonograms are quite subjective and reflect probabilities, not certainty. Both overaggressive and excessively timid interpretation can be misleading. Routine protocols for sonography are not always optimal. Although some technologists become extremely proficient after specific training and experience, supervision and participation by a knowledgeable and interested physician-sonographer is usually required to obtain a precise and pertinent answer to a specific problem that has been posed by the clinician. For instance, one group has reported accurate, surgically proven preoperative identification of non-recurrent inferior laryngeal nerves (15). It is not that the ultrasound images depict an inferior laryngeal nerve. Rather, the diagnosis is suggested when, while performing the sonogram, the surgeon asks a specific, direct question about the anatomic region where the nerve should be located. Thereafter, a series of images are obtained with and without Doppler interrogation that reveal the presence of a small, linear structure that, on Doppler interrogation, is associated with blood vessels, allowing a probable answer to the inquiry. The surgeon is then in a position to operate, minimizing the risk of adverse consequences.

 

Standard sonographic reports may provide considerable information about major anatomic features, but are suboptimal unless the specific clinical concern is explored and answered. Indeed, because some radiologists may not address the clinical issue adequately, and for convenience, numerous thyroidologists and surgeons perform their own ultrasound examinations, in their office or clinic (point of service). However, it is essential that non-dedicated ultrasonographers have state-of-the-art equipment (might not be cost-effective) and that they are willing to expend a considerable amount of time for a complete study, in particular if there is a need to evaluate the lateral neck compartments. Technical ingenuity, electronic enhancements such as Doppler capability, and even artistry are frequently required. Special maneuvers, various degrees of hyperextension of the neck, swallowing to facilitate elevation of the lower portions of the thyroid gland above the clavicles, swallowing water to identify the esophagus, and a Valsalva maneuver to distend the jugular veins may enhance the value of the images. Nevertheless, sonography is rather difficult to interpret in the upper portion of the jugular region and in the areas adjacent to the trachea. Aiming the transducer obliquely may permit exploration of the region behind the trachea. Sonography is generally not useful below the clavicles.

 

To orient the imager, it can be useful to survey the entire neck and thyroid gland with a low-energy transducer before proceeding to 10-15 megahertz equipment. Protocols have been devised to assemble a montage of images to encompass an unusually large lobe or goiter. For an overview, panoramic ultrasound, which is a variation of conventional ultrasound allows one to produce images with a large anatomic field of view, displaying both lobes of the thyroid gland on a single image (16).

 

There may be considerable differences between sonographers in estimating the size of large goiters or nodules (17). One investigation has reported that curved-array transducers may avoid significant inter-observer variation that may occur when linear-array equipment is employed, especially when the gland is very large (18). The inter-observer variation may be almost 50% even among experienced ultrasonographers, because it is difficult to reproduce a two-dimensional image plane for multiple studies (19). Accuracy in volume estimation becomes most important when one uses ultrasound measurements to calculate an isotope dose or to compare changes over time in the size of a nodule or a goiter. Indeed, it has been suggested even for well-defined nodules, a change of less than 1 cm in size should not be accepted as a real change (17). The important aspect is that the clinician must be guided by the constellation of risk factors, local anatomy, and intervening events, when making a management decision. Stability of size is one factor, but not a major one.

 

Using planimetry from three-dimensional images reportedly has lower intra-observer variability (3.4%) and higher repeatability (96.5%) than the standard ellipsoid model for nodules and lobes, with 14.4% variability and 84.8% repeatability (p < 0.001) (20).

 

Importantly, for autonomous nodules, US-evidence of growth does not indicate a likelihood of malignancy. Rather, it usually reflects cystic or hemorrhagic degeneration, which correlates well with prior experience by pathologists and the literature. In contrast, growth of a non-functional follicular adenoma can be of concern and the lesion needs to be carefully evaluated for other suspicious signs (21). There may be imperfect concordance between the ultrasonic dimensions of large thyroid nodules compared with intraoperative findings (22).

 

SONOGRAPHY OF THE NORMAL THYROID AND ITS REGION

 

The anterior neck is depicted rather well with standard gray scale sonography (Figure 1). The thyroid gland is slightly more echo-dense than the adjacent structures because of its high iodine content. It has a homogenous ground glass appearance. Each lobe has a smooth globular-shaped contour and is no more than 3 - 4 centimeters in height, 1 - 1.5 cm in width, and 1 centimeter in depth. The isthmus is identified, anterior to the trachea as a uniform structure that is approximately 0.5 cm in height and 2 - 3 mm in depth. The pyramidal lobe is not seen unless it is significantly enlarged. In the female, the upper pole of each thyroid lobe may be seen at the level of the thyroid cartilage, whereas it is lower in the male. The surrounding muscles are of lower echogenicity than the thyroid and tissue planes between muscles are usually identifiable. The air-filled trachea does not transmit the ultrasound. Only the anterior portions of the cartilaginous rings are represented by dense, bright echoes. The carotid artery and other blood vessels are echo-free unless they are calcified. The jugular vein is usually in a collapsed condition and it distends with a Valsalva maneuver. There are frequently 1-2 mm echo-free zones on the surface and within the thyroid gland that represent blood vessels. The vascular nature of all of these echo-less areas can be demonstrated by color Doppler imaging to differentiate them from cystic structures (12,13). Lymph nodes may be observed. Nerves are generally not seen. However, a keen understanding of the local anatomy may permit critical interpretation of a series of gray scale US and Doppler images to gain useful insights into the probable presence or absence of an expected neurovascular bundle. Meticulous preoperative analysis that may include lymph node mapping can benefit surgical management (15). The parathyroid glands are observed only when they are enlarged and are less dense ultrasonically than thyroid tissue because of the absence of iodine. The esophagus may be demonstrated behind the medial part of the left thyroid lobe, especially if a sip of water distends it (Figure 2).

Figure 1. Sonogram of the neck in the transverse plane showing a normal right thyroid lobe and isthmus. L = small thyroid lobe in a patient who is taking suppressive amounts of L-thyroxine, I = isthmus, T = tracheal ring (the dense white arc represents calcification, distal to it reflects artifact), C = carotid artery (note the enhanced echoes deep to the fluid-filled blood vessel), J = jugular vein, S = sternocleidomastoid muscle, m = strap muscle.

Figure 2. Sonogram of the left lobe of the thyroid gland in the transverse plane showing a rounded lobe of a goiter. L = enlarged lobe, I = widened isthmus, T = trachea, C = carotid artery (note the enhanced echoes deep to the fluid-filled blood vessel), J = jugular vein, S = sternocleidomastoid muscle, m = strap muscles, E = esophagus.

GENERAL THOUGHTS ABOUT SONOGRAPHY

 

Thyroid US may play a useful role in the management of patients even when the thyroid examination is normal but it is debatable if the procedure is cost-effective as a screening test (1). Many thyroidologists/endocrinologists advocate routine use of US at the time of physical examination to discover subclinical, non-palpable thyroid abnormalities, which will be discussed presently, and to enhance the sensitivity and accuracy of palpation. This practice is called “point of service” US.

 

Whether US is performed at the point of service or in an US laboratory by ultrasonographers/radiologists, it is important to employ thyroid sonography selectively to supplement or confirm a physical examination especially when clinical perception is confused by obesity, great muscularity, distortion by abnormal adjacent structures, tortuous regional blood vessels, a prominent thyroid cartilage, metastatic tumor, lymphadenopathy, or prior surgery.

 

In practice, US may be used to supplement an examination when there is uncertainty about the palpation. It is important, however, to comprehend that the optimal clinical value of US depends on the quality of the examination, including the experience of the examiner and the characteristics of the equipment. Grossly misleading results may occur with quick, incomplete studies, insensitive US machines or substandard interpretations. “Routine” sonography in a medical office, clinic or in a radiology facility by an incompletely trained clinician or general radiologist can be misleading. Without study, training and practice, there are likely to be unacceptable results and adverse outcomes. Furthermore, the efficacy of US when performed in sub-optimal conditions has yet to be critically examined.

 

In the academic situation, sonography is useful to teach palpation of the thyroid gland.

There are claims that US can offer insights into thyroid function. For instance, among 4649 randomly selected adult subjects one investigation found that there was correlation between thyroid hypoechogenicity and higher than average levels of serum TSH, even in subjects without overt thyroid disease (23). One group reported TSH elevations in 26 patients with autoimmune thyroiditis when there was a well-defined, approximately 10 mm triangular area of low echogenicity, between the lateral margin of one or both thyroid lobes, the medial wall of the carotid artery, and, posteriorly, the pre-vertebral muscles. Euthyroid patients (71) with thyroiditis and controls (154) did not demonstrate a hypoechoic “triangle” (24). In contrast, the question arises how accurately a normal thyroid sonogram will predict normal thyroid function? In one study of normal-appearing US, TSH was normal in 41/48 (85.4%) subjects but was elevated in 7 individuals (14.6%) (p<0.001) and anti-thyroid antibodies were detected in 5 patients (10.4%) (25). Therefore, a normal sonogram does not preclude hypothyroidism or Hashimoto’s thyroiditis.

 

SONOGRAPHY USED TO FACILITATE AN UNCONVENTIONAL SURGICAL APPROACH TO THYROIDECTOMY OR REMOVAL OF METASTASES

 

US maybe used to guide a surgeon who performs a trans-axillary or sub-mammary approach to thyroidectomy, thereby avoiding a neck scar. Retro-pharyngeal thyroid metastases can be managed via trans-oral robotic and surgeon-performed US to localize and excise lymphadenopathy (26).

 

SONOGRAPHY IN THE PATIENT WITH AN ENLARGED THYROID GLAND (GOITER)

 

Thyroid sonography probably is not cost-effective in evaluating the average patient with thyroid enlargement. Since thyroid goiters are common and rarely associated with malignancy, there is little useful purpose to sonographic documentation of the size, shape, or uniformity of a goiter. However, US may be used in a goiter to identify non-palpable thyroid nodules for biopsy, or those that have enlarged or become harder. Importantly, US permits one to characterize nodules and estimate the risk of malignancy. The value of aspirating a selected nodule in a goiter is under current scrutiny. At this time, the data seems persuasive that the incidence of cancer in a particular nodule in a goiter is independent of the number of sonographically identified nodules, in distinction to prior belief. Therefore, this practice seems worthwhile (27,28).

 

At times, it will be useful to know the ultrasonic appearance of a dominant nodule in a goiter, a tender spot, a region of focal hardness because it might give a clue about pathology (Figure 2) (1). For example, sonography can identify one region in a goiter whose echo pattern is distinct from the rest of the goiter suggesting a second type of pathology, especially if the region is surrounded by an incomplete and irregular sonolucent rim, has punctate microcalcifications, or Doppler examination reveals internal vascularity. The significance of these ultrasonic features will be discussed below. Among the lesions that have been demonstrated in goiters using US are neoplasms and lymphoma. Other uses of sonography in goitrous patients include: differentiation of thyroid enlargement from adipose tissue or muscle, identifying a large unilateral mass in distinction to an asymmetric goiter, confirming substernal extension, providing the correct interpretation to varying clinical impressions among several examiners, and objectively documenting volume changes in response to suppressive therapy with thyroid hormone, which may be particularly useful information when patients change physicians.

 

An interesting public health use of US in underdeveloped countries has been to objectively identify goiter as a screen for iodine deficiency. Furthermore, in the epidemiological setting, with proper ultrasound equipment, assessment of thyroid volume and prevalence of thyroid nodules, but not echogenicity or echographic pattern, are comparable among different observers (29).

Sonography is useful to monitor patients undergoing long-term treatment with lithium for bipolar, major depressive, and schizoaffective disease. Their total thyroid volume in one investigation was significantly greater (23.7 ml vs. 13.6 ml) in the lithium-treated group (30) than among controls (96 sex- and age-matched control subjects). Furthermore, US detects thyroid enlargement with greater accuracy and sensitivity than palpation (31).

 

SONOGRAPHY WITH THYROIDITIS AUTONOMOUS NODULES, AND GRAVES' DISEASE

 

Routine sonography can be useful in distinguishing thyroiditis or Graves' disease, but it is uncertain whether this is cost-effective. Several publications have shown that the ultrasound pattern correlates with the presence of autoimmune thyroid disease and can predict thyroid dysfunction as will be discussed below (32). In subacute thyroiditis, the severely inflamed thyroid reflects very low intensity echoes, which is generally not seen with any other thyroid disorder (33). In the inflamed portions of the thyroid gland there is no increased vascular flow pattern on Doppler examination. The non-involved regions demonstrate normal vascularity and hemodynamics. In the recovery phase of subacute thyroiditis, the thyroid regains isoechogenicity and a Doppler study may show slightly increased vascularity (33-37). Hashimoto's thyroiditis and Graves' disease show moderately heterogeneous, reduced echogenicity (38-43). The diagnostic precision of this US pattern was compared to that of anti-thyroid peroxidase antibody (TPOAb) concentrations in 451 ambulatory patients with unknown thyroid status, excluding those with suspected hyperthyroidism or on drugs known to cause hypothyroidism. There was high intra-observer and inter-observer agreement on the abnormal thyroid ultrasound patterns, which were judged highly indicative of autoimmune thyroiditis and allowed the detection of thyroid dysfunction by other means with 96% probability (44). It has been reported that among 55 patients with hyperthyroidism (29 Graves' disease and 26 toxic nodules), color flow Doppler examination was useful to differentiate the etiology. Increased blood flow was successful in differentiating untreated Graves' disease from Hashimoto’s thyroiditis, both of which had similar gray scale findings (p < 0.001), and from controls (p < 0.001). “Hot”, autonomous nodules could also be differentiated from “cold” nodules because of more prominent vascular patterns and significantly higher peak systolic velocity values (p < 0.001) (45). The gray-scale US features of nontoxic autonomous nodules are similar to those of toxic autonomous nodules (46).

 

Investigation of patients with postpartum thyroiditis who had both high levels of antithyroid peroxidase antibody and a hypoechogenic thyroid gland also had a high risk of long-term thyroid dysfunction (47). In 119 patients with postpartum thyroiditis and 97 normal postpartum women as the control group, thyroid hypoechogenecity was present in 98.5% of patients and 7% of the control group (p <0.001). Initially, mean thyroid volume in the patients with thyroiditis was 77% greater than in the control group. After remission, mean thyroid volume decreased by 25% in the thyroiditis patients. Twelve months after delivery, hypoechogenicity persisted in 4 patients (48).

 

It has been reported that in children US findings of Hashimoto’s thyroiditis are present in only a third at the time of diagnosis and half of the Hashimoto’s children with normal initial thyroid sonography develop changes within 7 months. In some cases, characteristic Hashimoto’s findings may not develop for over 4 years (49).

 

Especially in Graves' disease, color Doppler imaging of the thyroid can demonstrate diffuse hyperemia of the thyroid gland (50) that has been called a "thyroid inferno" (51). In patients with amiodarone-induced thyrotoxicosis, Doppler flow sonography has been reported to differentiate two types of disorder with implications for therapy (52-56). Patients with moderate to high vascular flow had underlying thyroid disease, such as latent Graves' disease or nodular goiter. The latter are at risk of amiodarone-induced thyrotoxicosis type I (AIT I), which is caused by the organification of the high amounts of iodine in amiodarone. In contrast, AIT II is caused by a destructive thyroiditis caused by the drug and there is typically no demonstrable vascular flow. The clinical value of this observation is that the Type II patients seem to respond to treatment with glucocorticoids. In contrast, AIT I patients tend to respond to a combined regimen with methimazole and potassium perchlorate (52). Although this conclusion was originally based on a small number of patients, the observations were confirmed in a retrospective case-note audit of 37 patients (53). Interestingly, in that study, euthyroid amiodarone-treated patients failed to show hyperactive flow (52). In another investigation, looking at the data from the perspective of patients who had been treated for amiodarone-induced thyrotoxicosis, in a retrospective study of 24 patients, responsiveness to prednisolone correlated poorly with the absence of enhanced blood flow in the thyroid glands, but the presence of enhanced flow appeared to correlate with non-response to prednisolone (55). Interleukin 6 (IL-6) levels correlated with the ultrasound classification in one study (52), but not in another (53).

 

A report has successfully validated excessive mean peak systolic velocity of the superior thyroidal artery in Graves’ disease but not in patients with destructive thyroiditis (57).

An important application of standard US in patients with thyroiditis or Graves' disease is to assess those thyroid glands that have focal firm consistency or are enlarged or painful for coincidental tumor or lymphoma (1). In one report, patients with Hashimoto's thyroiditis had sonography to detect nodules and then had ultrasound-guided aspiration biopsy to elucidate the nature of the lesion. Two of 24 patients (8.3%) had papillary thyroid cancer (58).

 

In patients with thyrotoxicosis, US can assess the size of the thyroid gland to facilitate I-131 dosimetry. The size of each lobe is measured in the sagittal and transverse planes to provide the length (L), anterior-posterior depth (D), and transverse width (W), respectively. The volume of each lobe is calculated using the formula for a prolate ellipse: (volume = 0.5 {L x D x W}). 3D echography may improve the accuracy of assessment of thyroid volume (20).

 

Doppler sonography may become a useful tool for the clinical endocrinologist in the management of patients with Graves’ disease if observations are confirmed in large populations. It has been suggested that color-flow mapping of the thyroid gland may have a role in the selection of an optimal dose of methimazole needed to maintain a euthyroid state in patients with Graves' disease (59). Another study has characterized Doppler ultrasound data from patients with Graves' disease, Hashimoto's disease, and goiter to obtain a "hemodynamic index" to ascertain when antithyroid drugs should be withdrawn or ablative therapy given in patients with Graves' disease. The hemodynamics in the thyroid was significantly different between untreated thyrotoxic and medically well-controlled patients but there were no significant differences between untreated or medically poorly controlled patients. It would be interesting to ascertain whether the hemodynamics permit an identification of a subset of well-controlled patients who will relapse after a course of therapy (60). Furthermore, Doppler sonography has provided data from 40 patients with Graves' disease showing significantly increased thyroid blood flow in euthyroid patients who presented early in relapse after withdrawal of antithyroid drug therapy when compared with 16 age-matched normal control subjects. Conversely, there were no significant differences in euthyroid patients who remained in remission when compared with normal controls (61). The value of quantifying thyroid blood flow at the time of diagnosis has been assessed in 24 patients with Graves' disease, using percutaneous spectral Doppler recordings from the thyroid arteries, in an attempt to predict the likelihood of remission following withdrawal of antithyroid drug therapy. The mean duration of treatment was 14 months and follow-up in 13 women was at least 18 months (range: 18 - 39 months) after antithyroid drug withdrawal. Mean peak systolic velocity and volume flow rate values as well as thyroid volume measured at the time of diagnosis were significantly higher (139 cm/s, SD 46; 195 ml/min, SD 170; 52 ml, SD 18) in patients who relapsed after drug treatment compared with patients in remission (71 cm/s, SD 27; 67 ml/min, SD 61; 25 ml, SD 13) (62). Thyroid hypoechogenicity at onset of Graves’ disease is probably not a reliable prognostic index of relapse after medical treatment. However, the absence of thyroid hypoechogenicity after methimazole treatment seems to be a favorable prognosticator of remission (63). In another investigation, Doppler ultrasound determined increased peak systolic velocity in the inferior thyroid artery in untreated hyperthyroid patients with Graves' disease was significantly and positively associated with the maintenance dose of methimazole needed to keep TSH normal (64). Normoechoic Graves' hyperthyroid glands seem to be more resistant to therapy with I-131 than hypoechoic thyroids (65).

 

Another example of the value of Doppler ultrasound relates to the administration of iodide solutions that have been used traditionally prior to thyroid surgery for Graves' disease because it was thought that they reduce the vascularity of the thyroid gland. Doppler echography has demonstrated a significant decrease in thyroid vascularity in patients with Graves' disease after seven days of Lugol's solution, confirming the rationale of this form of treatment (66). Preoperative treatment with Lugol’s solution decreased the rate of thyroid blood flow and vascularity, as assessed by Doppler evaluation. Lugol’s solution also decreased intraoperative blood loss during thyroidectomy in another investigation (67). In contrast, US has also shown that preoperative iodide may increase the size of the thyroid gland, which could complicate surgery when a Graves’ thyroid is very large before the Lugol’s solution is administered (68).

 

Doppler examination has been used trans-vaginally in pregnant women with Graves’ disease to depict and assess the size of the fetal thyroid gland. Clinical benefits might include facilitating adjustment of the mother’s dose of antithyroid drug and anticipating or preventing fetal and neonatal hypothyroidism. The authors suggested that when reduction of the medication does not result in decrease in the size of the fetal goiter, trans-placental passage of thyroid stimulating immunoglobulin should be suspected (69).

 

SONOGRAPHY OF LYMPHOMA

 

In the author’s experience, the value of US to predict lymphoma is very limited. However, the sonographic patterns of thyroid lymphoma have been classified into three types based on internal echoes within the suspected lesion, the border of the lesion, and the intensity of the echoes behind (deep to) the lesion. The echoes behind the lesion in each type of lymphoma are increased, presumably because of enhanced transmission of the ultrasound through the lesion. In the nodular type of lymphoma, the internal echoes within a nodule are uniform and hypoechoic (may be sufficiently hypoechoic to be pseudocystic). The border between lymphoma and non-lymphomatous tissue is well-defined and the borderline is described as “broccoli-like or coastline-like” irregularity. In the diffuse type of lymphoma internal echoes are also exceedingly hypoechoic but the border between lymphoma and non-lymphomatous tissues is not distinct. It is difficult to differentiate the diffuse type lymphoma from chronic thyroiditis. The mixed type lymphoma shows multiple, patchy hypoechoic lesions, each with enhanced posterior echoes (70).

 

SONOGRAPHY OF THE THYROID NODULE

 

The most frequent use of US is to refine the diagnosis of a thyroid nodule. US can identify thyroid nodules, even when they are too small to palpate. Sonography can demonstrate nodules that have an enhanced risk of malignancy with the best sensitivity of any non-invasive technique, but with only fair specificity. In addition, FNA of thyroid nodules should be performed under US guidance whenever possible.

 

Thyroid nodules can be identified by sonography because they distort the uniform shape or echo pattern of the thyroid gland. Thyroid nodules may be large or small. They may distort the surrounding thyroid architecture or may dwell within a lobe and be unobtrusive. They may be solid tissue or consist of solid areas interspersed with echo-free zones that represent fluid-filled hemorrhagic or straw-colored degenerative zones (Figure 3). A smooth, globular area without echoes generally represents an epithelial-lined cyst, which is a rare benign lesion (Figure 4) (71). Most thyroid nodules have a less dense ultrasound appearance than normal thyroid tissue and some are more echo-dense (6). A sonolucent rim, which is called a halo, may be present around a nodule. A halo represents a capsule or another interface, such as inflammation or edema, segregating the nodule and the rest of the gland. Doppler technique may demonstrate increased vascularity within a nodule or in a halo (Figure 5) (12). “Nodules” are not a single disease but are a manifestation of different diseases including adenomas, carcinomas, inflammations, cysts, fibrotic areas, vascular regions, and accumulations of colloid.

Figure 3. Sonograms showing longitudinal (left panel) and transverse (right panel) images of the left lobe containing a degenerated thyroid nodule. Note the thick wall and irregularity. N = nodule, H = hemorrhagic degenerated region.

Figure 4. The left panel shows an anterior scintiscan of a euthyroid patient who had a firm nodule in the left thyroid lobe. The nodule is "cold". * * * = nodule. The right panel shows a sonogram of the neck in the longitudinal plane revealing that the nodule is a smooth-walled cystic structure without internal echoes. between the + + symbols. Note the dark dense echoes distal to the cyst. C = cyst, L = thyroid lobe.

Figure 5. Sonogram of the neck in the longitudinal plane showing a hypoechogenic nodule that was surrounded by an echo free rim, called a halo. Doppler examination demonstrates great vascularity in the halo, identified as bright spots. Small blood vessels are also seen elsewhere. N = nodule, L = heterogeneous thyroid lobe, m = muscle.

The ultrasonic appearance of a thyroid nodule does not reliably differentiate a benign thyroid lesion and cancer (1,6)but it does offer strong clues that help the clinician in the process of triage. Nevertheless, sonography cannot identify a specific kind of tumor such as a Hürthle cell lesion (72). The most reliable sonographic indicator that a nodule is malignant is observing vascular invasion by tumor, which is rarely seen. However, there are distinctions in echo-density, calcifications, distortions of the rim, and vascularity that favor a benign or malignant diagnosis (73,74). These characteristics are summarized in Tables 1 & 2. But it is important to understand that the features described reflect statistical probabilities and not dependable criteria.

 

Table 1. Ultrasound Characteristics Associated with an Increased Thyroid Cancer Risk

1. Hypoechoic

2. Microcalcifications

3. Central vascularity

4. Irregular margins

5. Incomplete halo

6. Taller-than-wide

7. Significant enlargement of a nodule

 

Table 2. Ultrasound Characteristics Associated with a Low Thyroid Cancer Risk

1. Hyperechoic

2. Large, coarse calcifications (except medullary thyroid cancer)

3. Peripheral vascularity

4. No hyper-vascular center

5. Spongiform appearance (puff pastry appearance)

6. Comet-tail shadowing

 

An important aspect is that single or even a few US features may be inadequate to select some nodules for FNA or to reliably assess the risk of thyroid cancer. In contrast, selections based on multiple characteristics that are associated with elevated cancer risk are more dependable indicators of probable malignancy. Nevertheless, certain single features should prompt for FNA including microcalcifications, a taller-than-wide shape, or irregular margins. Absence of elasticity-will probably identify nodules with a clinically meaningful increased, perhaps even high risk for malignancy (75).

 

  • ECHOGENICITY: Thyroid malignancies tend to be hypoechoic when compared with the rest of the thyroid (71,76-79). Since many benign thyroid nodules, which are far more common than malignancies, are also hypoechoic, this finding is not particularly useful except that it is reasonably safe to conclude that hyper-dense nodules are probably not cancerous. One group of investigators has concluded that hyperechogenic lesions occurring in thyroiditis-affected thyroid glands bear no-clinical relevance. Therefore, they advocate that aspiration biopsy of these nodules is not advisable (80) and many clinicians follow that practice.
  • CALCIFICATIONS: The presence of calcification is also not a straightforward diagnostic aid. Microcalcifications are relatively more common in malignant than in benign lesions and may represent psammoma bodies. Microcalcifications have been reported as demonstrating a 95.2% specificity for thyroid cancer, but a low sensitivity of 59.3 % and a diagnostic accuracy of 83.8% (77). B-flow ultrasonic imaging may be particularly sensitive in detecting microcalcifications by demonstrating “twinkling” in some nodules (14). However, large coarse calcifications and calcifications along the rim of nodule are common in all types of nodules and reflect previous hemorrhage and degenerative changes. Thus, since some cancers may have been chronic and have undergone degenerative change, they may demonstrate peripheral or coarse internal calcification. Therefore, diagnostic FNA biopsy may be appropriate even when there are large, coarse, or eggshell calcifications to avoid missing a cancer (79). Indeed, in one investigation, among 64 thyroid nodules with peripheral calcifications 19 (30%) were benign, and 45 (70%) were malignant. Interruption and thickening of peripheral calcifications and decreased internal echogenicity of a thyroid nodule with peripheral calcifications were associated with malignancy in this study (81). In our estimation, considerably more than 30% of such nodules are benign; thyroid calcifications that are greater than pin-point size provides little practical help in identifying cancer in the individual patient. In one study, the highest incidence of calcification was found in thyroid cancer (54%), followed by multinodular goiter (40%), solitary nodular goiter (14%), and follicular adenomas (12%). The authors reported that calcifications in a "solitary" nodule in a person younger than 40 years should raise a strong suspicion of malignancy: relative cancer risk of 3.8 versus 2.5 in patients older than 40 years (82). In contrast to the prior statements, it is important to note that large calcifications are seen with increased frequency in medullary thyroid carcinoma (83).
  • HALO: A halo around the nodule may be seen with benign or malignant conditions. It suggests that there is an acoustic interface around the nodule that does not reflect the ultrasound. It implies that there are two different types of histology in the region: the nodule and the surrounding thyroid (6,71,84). Some observers have suggested that cancer should be suspected when the periphery of a halo has a blurred appearance. We have not found that characteristic reliable. Since adenomas are more common than carcinomas, the finding of a halo is, in our opinion, more often seen with adenomas than carcinomas.
  • NODULE BORDER: There have been investigations of a possible correlation between the degree of definition of the border of a nodule and the likelihood of malignancy and even of the predictability of aggressive characteristics of a papillary cancer. In one series of 155 cases, poor definition of a nodule’s edge was observed in 21.5% of patients, all of whom showed worse disease-free survival (p = 0.0477) than those with a well-defined edge. Furthermore, this finding was directly linked to US-diagnosed lateral node metastasis (p = 0.0001) (85). Ultra-high frequency thyroid ultrasound (12–18 MHz) may reveal jagged edges and lobulated borders in 1 to 3 cm thyroid nodules. These findings have been reported to correlate with papillary thyroid cancer with a sensitivity of about 60 to 70% (86).
  • HEMODYNAMIC CHARACTERISTICS: Increased blood flow in the central part of a nodule is more likely associated with cancer than when the vascularity is along the periphery. An analysis of the hemodynamic characteristics of a nodule by high resolution pulsed and power Doppler ultrasonography also may offer valuable preoperative diagnostic insights. For example, one study of 25 follicular adenomas and 10 follicular carcinomas compared the vascular pattern and the velocimetric parameters (such as peak systolic velocity), end-diastolic velocity, pulsatility index or resistance index. Eight of 10 patients with follicular carcinomas showed moderate increase of intra-nodular vascularity using “Power Doppler”. In contrast, the 21 out of 25 follicular adenomas showed only a peripheral rim of color flow. Furthermore, the velocimetric analyses were significantly higher in the patients with cancer than those with adenomas (80).

 

Bayes' mathematical theorem has been used to evaluate the cancer diagnostic value of enhanced intranodular blood flow by Doppler analysis in determining the probability of cancer in thyroid nodules that demonstrate follicular cytology. The sensitivity of enhanced intranodular flow by Doppler analysis for detection of thyroid carcinoma was 80%-86% and the specificity of indicating cancer ranged from 85% to 89%. In contrast, the probability that a nodule is thyroid cancer before a Doppler test was estimated at 12%-14%. After Doppler examination, the probability of thyroid cancer increased to nearly 50% in the presence of intranodular flow but declined to 3% when there was no central intranodular flow (87). In one investigation of 230 patients, 203 of whom were treated surgically, the addition of color flow Doppler imaging to conventional sonography increased the screening sensitivity and accuracy in identifying 36 malignant thyroid nodules from 71.9% to 83.3% (88). Thus, Doppler ultrasound may be a particularly useful predictor of the risk of malignancy in thyroid nodules (89).

 

Doppler sonography employing ultrasound contrast medium may further enhance the diagnosis of thyroid cancer. In one investigation, carcinomas showed a significantly earlier arrival time of Levovist in the nodule than nodular hyperplastic benign nodules or adenomas (90).

  • SHAPE: There have been observations that some cancers tend to have a non-globular, “tall” shape, as if growing in one plane (i.e. the depth of the nodule is larger than the width in the horizontal plane). Nodules that are tall rather than wide should be viewed with considerable suspicion. This observation has recently been validated in 500 patients with thyroid microcarcinomas (91).
  • CYSTIC SPACES: Especially large benign or malignant thyroid nodules tend to undergo hemorrhagic or cystic degenerative changes. It has been reported that features associated with cancer in a cystic thyroid nodule include more than 50% solid tissue, eccentricity of the cystic space, and micro-calcifications (92).
  • MISCELLANEOUS CHARACTERISTICS: Ultrasonographers have observed that colloid nodules, which are benign with high probability, have a more or less characteristic appearance of a “stack of pancakes”, “puff pastry like a Napoleon”, or sponge. In one publication, among 201 nodules, no malignancies were found when the US appearance was greater than 75% “sponge-like” (spongiform) (93). There may be a small, echogenic, bright spot with “comet-tail shadowing” associated with colloid that must be differentiated from a pin-point bright spot. There seems to be triage-merit to these characteristics, which will require critical scrutiny. Furthermore, it is important to be aware that a cancer may co-occur in an otherwise nodular or colloid goiter. This issue will be further discussed below in the section on thyroid biopsy.

 

To summarize, the cancer-predictive value of ultrasonic characteristics varies considerably but is acceptable when multiple characteristics are considered together. The most supportive data we have found is that there was a 97.2% positive predictive value for cytologically diagnosed cancer and 96.1% predictive value for benign disease among 1,244 nodules in 900 patients who were stratified according to ultrasound characteristics on a scale of 1-5 assessing cancer-risk (85).

 

In contrast, the sensitivity for cancer, however, is lower as shown in the following studies. Retrospective examination of 849 nodules (360 malignant, 489 benign) revealed that statistically significant (P <0.05) sonographic characteristics of malignancy included: a taller-than-wide shape (sensitivity 40.0%; specificity 91.4%), a spiculated margin (sensitivity 48.3%; specificity 91.8%), marked hypoechogenicity (sensitivity 41.4%; specificity 92.2%), microcalcification (sensitivity 44.2%; specificity 90.8%), and macrocalcification (sensitivity 9.7%; specificity 96.1%). The US findings for benign nodules were isoechogenicity (sensitivity 56.6%; specificity 88.1%; P <0.001) and a spongiform appearance (sensitivity 10.4%; specificity 99.7%; P <0.001). The presence of at least one malignant US finding had a sensitivity of 83.3%, a specificity of 74.0%, and a diagnostic accuracy of 78.0% (74). In an iodine-deficient geographic region where there is endemic goiter and thyroid nodules are frequent, among 2,642 consecutive patients (3,645 nodules) a numeric score was assigned to nodules based on ultrasonic high-risk of cancer. Nodules with a score of over 5.5 out of 10 had a 66% sensitivity and a 76% specificity for cancer, both of which were much higher values than when the scores were below 5 (94).

 

It is noteworthy that the results of sonography may influence a management decision even when the results of needle biopsy are only “suspicious”. In one study, 303 patients who had thyroid nodules with an aspiration biopsy reading of merely suspicious for papillary thyroid cancer had surgery anyway. The pre-surgery ultrasound examination had a positive predictive value of 94.9%, and negative predictive value of 80.9% (95).

 

The use of a Bayesian classifier to differentiate benign and malignant thyroid nodules by using sonographic features is under investigation (96).

 

Preoperative US of a nodule that turns out to be thyroid carcinoma has a very limited ability to predict postoperative staging. In one study, the sensitivity of depicting metastases to lymph nodes was 36.7%, invasion of the muscles 77.8%, trachea involvement 42.9%, and esophagus 28.6% (97).

 

Postoperatively, sonographic features of nodules in a thyroid bed cannot reliably distinguish recurrent thyroid cancer and benign thyroid remnants (98). However, in remnants, increased vascularity, and microcalcifications of a lesion that is larger than 6 mm in size should be viewed with suspicion.

 

Perhaps more objective, computerized triage of ultrasound features of thyroid nodules will become possible. In one investigation an artificial neural network and binary logistic regression was significantly better than two experienced radiologists in distinguishing benign and malignant thyroid nodules based on 8 ultrasonographic parameters: size, shape, margin, echogenicity, cystic change, microcalcification, macrocalcification, and halo. The study included 109 pathologically proven thyroid lesions (49 malignant and 60 benign) in 96 patients (99).

It is important to note that there may be significant inter-observer variation in interpretation. The inter-observer variation in the interpretation of thyroid ultrasonograms among 4 experienced readers reviewing 144 patients, varied according to the characteristic examined. Echogenicity showed slight agreement (kappa = 0.34); composition, margin, calcification, and final assessment had fair agreement (kappa = 0.59, 0.42, 0.58, and 0.54, respectively); shape and vascularity showed substantial agreement (kappa = 0.61 and 0.64, respectively). Intra-observer variability showed better agreement (kappa > 0.61). For the four radiologists, the overall sensitivity was 88.2%, specificity 78.7%, positive predictive value 76.2%, negative predictive value 89.6%, and accuracy 82.8% (100).

 

There have been investigations into the differences in the biologic behavior of thyroid cancer based on preoperative US features. One study in patients with follicular variant of papillary thyroid cancer showed more aggressive cancer behavior when there were preoperative US characteristics that suggested malignancy when compared with those without such features (101).

 

In children, there is no consensus about the value of US characteristics as predictors of malignancy. One group is not enthusiastic (102). Another group of investigators who also did molecular genetics on aspirated thyroid nodules offered a more positive view (103).

 

SONOGRAPHY OF A PALPABLE DOMINANT NODULE IN AN ENLARGED OR NODULAR THYROID

 

We now know that a so-called “solitary nodule” in an otherwise normal thyroid gland often is a nodule in a gland that has sub-clinical nodules (see below). Even more frequently, clinicians encounter patients with a “dominant” nodule in an enlarged or nodular thyroid. It is generally agreed that for a dominant thyroid nodule FNA is the best test to assess malignancy. Furthermore, a diagnostic strategy using initial FNA was found to be more cost-effective than starting with ultrasonography or scintigraphy (104). Evidence is mounting in support of US for patients with palpable uninodular thyroid disease and goiter because non-palpable nodules are common and a few of these are cancerous. In many countries, US is being employed more often than previously especially when palpation is uncertain or skills are tentative. US has been reported to provide information to the clinician that importantly alters management in 63% (109/173) of patients who were referred to a tertiary endocrine group. Sonography showed an indication for needle aspiration or demonstrated that the procedure is not necessary. Among 114 patients who were referred because of a solitary thyroid nodule, US detected additional nonpalpable thyroid nodules that were at least 1 cm in diameter in 27 patients and no nodules in 23 subjects. Thus, among 50 patients US lead to an almost equal number of additional aspirations or no biopsy. Among 59 patients who were referred because of goiter, US showed no nodule in 20, thus avoiding biopsy, and revealed nodules at least 1 cm in diameter in 39 patients that required aspiration (27).

 

THE NON-PALPABLE THYROID NODULE OR INCIDENTALOMA

 

Sonography demonstrates micronodules (incidentalomas) of the thyroid that are less than 1 cm in diameter, non-palpable, common, and of questionable clinical significance (105) (Figure 6). Whereas palpable thyroid nodules occur in 1.5 - 6.4 % of the general population (106), the incidence of non-palpable nodules is at least ten fold greater when the population is screened by US (107). Non-palpable nodules increase with age to involve approximately 50% of older adults, especially women. The risk of malignancy among palpable nodules is approximately 10% and in micronodules had been thought to be considerably smaller (108). However, investigations reported a similar incidence of cancer in palpable and non-palpable thyroid nodules (109-111). One study actually reported a higher incidence of malignancy among incidentally discovered nodules than among clinically detected lesions (112). However, most microcarcinomas are clinically indolent. Yet, among 317 incidentalomas that were aspirated from 267 patients the rate of malignancy was 12% in a retrospective analysis. In addition, in this subgroup, 69% (25/36) of patients had either extra-thyroidal extension or regional node involvement and 39% had multifocal tumors at surgery, suggesting that the small size alone does not guarantee a low risk in incidentally found thyroid cancers (113). Therefore, the clinical impact of incidentalomas is quite small but they cannot be ignored. Rather, they should be monitored at intervals with US for suspicious characteristics, size, adenopathy, other clinical features, and - perhaps – a thyroglobulin measurement.

 

How useful are the sonographic characteristics of non-palpable nodules as an index of malignancy? Some insight to this question has been gained from a study performed on 16,352 self-referred patients in a health care center. Among 1,325 non-palpable thyroid nodules in 1,009 patients, marked hypoechogenicity, an irregular shape, a taller-than-wide shape, a well-defined spiculated margin, microcalcification, and an entirely solid nature were significant predictors for malignancy (P < .05) (114).

Figure 6. Sonograms of the right thyroid lobe in the longitudinal plane showing a 2.7 x 3.2 mm hypoechoic nodule that is delineated in the lower panel by the xx and ++ symbols. Note the linear hypoechoic structure below that (arrow). In the upper panel the bright structure is a Doppler signal and indicates a blood vessel below the nodule. The nodule is not vascular.

Non-palpable nodules or those that have escaped detection on examination are often discovered incidental to imaging of the neck for vascular or neurological reasons. They may be discovered during upper GI endoscopy (115). These thyroid lesions should be managed like other “Incidentalomas”, with observation, dedicated thyroid US, aspiration biopsy, or even surgery, as indicated by the data and mature judgment. This opinion is supported by an investigation in which thyroid nodules were found in 9.4% (116) of 2,004 consecutive patients undergoing carotid duplex ultrasonography. There was high correlation of the nodules with standard thyroid ultrasonography (presence of nodules, 97% (64 of 66) and size, r = 0.95, P<.001). Twenty-one (32%) of the nodules were smaller than 1 cm. Only two patients with unilateral masses noted on carotid duplex had a normal thyroid sonogram. Twenty-nine of the 66 (44%) were selected for fine-needle aspiration biopsy due to cancer-risk criteria. These results lead to surgery in 13 of the 66 (19.7%); pathology included 5 patients with cancer (3 with papillary cancer, 2 with follicular cancer), 4 patients with a follicular adenoma, and 2 with lymphocytic thyroiditis (117).

 

How successful is ultrasound-guided cytological diagnosis of non-palpable nodules? Intuitively, it is generally believed that success varies inversely with nodule size but the data are not conclusive. The diagnostic yield with nodules as small as 10 mm has been reported as comparable to that of aspirating larger nodules (110). Adequate material for cytological analysis reportedly was obtained in 64% of 0.7-cm lesions and 86.7% of 1.1 cm nodules. For nodules ≥1 cm, the sensitivity was 35.8% and false-negative results were seen in 49.3% (118). In contrast, a study of aspirates from 317 nodules in 267 patients reported that the size of impalpable nodules (0.9 +/- 0.3 cm, a range of 0.2 cm to 1.5 cm) was not related to the probability of getting an adequate specimen for cytological diagnosis (105). Of 201 thyroid nodules that were 5 mm or smaller in size, in 180 patients, investigators reported that were 162 adequate specimens (81%) (115). Personally, we generally do not routinely aspirate nodules smaller than 8 mm but have had limited diagnostic success in sampling incidentalomas as small as 5 mm. Based on a review of the literature, Mazzaferri et al. have concluded that thyroid nodules 5 mm or smaller have a high rate of false positive ultrasound findings and often yield inadequate cytology on fine needle aspiration biopsy. Therefore, they advise that nodules of this size with no other suspicious clinical findings should not undergo routine needle biopsy, even if they appear ultrasonographically suspicious (119). In contrast, more optimistic results have been reported. When ultrasound-guided FNA was done on 5 mm or smaller nodules, surgical confirmation was obtained in 62 nodules and there were 34 (55%) true positives, 0 (0%) false positives, 23 (37%) true negatives, and five (8%) false negative results for malignancy (sensitivity 87%, specificity 100%, positive predictive value 100%, negative predictive value 82%, accuracy 92%, false positive rate 0%, and false negative rate 8%) (120). However, considering the minimal clinical impact of thyroid microcarcinomas, the clinical value of aspirating nodules this small is uncertain. Importantly, the American Thyroid Association guidelines recommend avoiding cytological evaluation of nodules less than 1 cm in size (1). A selected, population base study of 485 thyroid nodules suggested that this advice would not miss any thyroid cancers with high risk features (121).

 

US has changed our clinical perception of what is a normal thyroid gland and has advanced medical practice. Current high-resolution ultrasonography of the thyroid has permitted the clinical detection of nodules that are as small as 2 mm. It frequently demonstrates that what appears to be a normal gland, actually contains a non-palpable nodule or is a subclinical nodular goiter (78,108). It may show that a solitary nodule on palpation really is a clinically palpable nodule in a gland that is subclinically multinodular. Pathologists have long known about the ubiquitous nature of thyroid micronodules and the relative frequency of occult thyroid carcinomas, which are rarely of clinical consequence. Now the clinician is often confronted with the challenge that micronodules are discovered as a consequence of investigations for orthopedic, neurological, vascular pathology or other pathologies, or together with a palpable thyroid nodule. As a rule, their discovery often results in needless expense, concern, and therapy because it is not known which of the myriad nodules that have been revealed is, or will progress to become a cancer with clinical impact.

 

It remains for future investigation to determine the appropriate management for micronodules. Because it is rare for one of these lesions to represent an occult thyroid cancer and rarer still for one to become a clinically significant malignancy, non-selective surgery, which has an exceedingly small yield of cancer and is not risk-free, seems ill advised. Also inappropriate is dismissal of the problem as unimportant. Rather, to this author, periodic sonographic reassessment for possible growth of the nodule or change in characteristics appears preferable. The role of ultrasound guided needle biopsy in the management of these patients, especially when there is a history of exposure to therapeutic x-ray will be discussed below.

 

Not all "incidentalomas" in the neck are thyroid in origin. Parathyroid adenomas have been observed within the thyroid gland or in the usual parathyroid anatomic location when ultrasonography was performed to evaluate thyroid nodules (122,123). An example of a misidentified lesion that demonstrates the extent of the lack of specificity of a “sonographic nodule” is an esophageal tumor that was erroneously characterized as thyroid (124).

 

SONOGRAPHY OF LYMPHADENOPATHY

 

Even in a patient with thyroid cancer, enlarged benign thyroid lymph nodes are more common than malignant ones. Nevertheless, US may be useful to diagnose and if appropriate, periodically reassess lymphadenopathy in the patient with a history of thyroid cancer, or if there is a history of exposure to therapeutic radiation in childhood or adolescence. A high-resolution ultrasound system equipped with a high-energy linear probe, a 12 -15 MHz transducer, B-Mode and Doppler capability, experience, and diligence are required to detect lymphadenopathy.

 

NORMAL LYMPH NODES: Normal lymph nodes are depicted by sonography as approximately 1 X 3 mm, well-defined, elliptical, uniform structures that are slightly less echo-dense than normal thyroid tissue and that have an echo-dense central hilum. Lymphadenopathy that is reactive to infection may be larger but the lymph nodes tend to maintain an oval shape; in contrast, malignant nodes more often have a "plump" rounded shape (125) (Figures 7, 8).

Figure 7. Sonogram in the longitudinal plane of the left side of the neck after thyroidectomy showing a small, elliptical benign appearing lymph node in the jugular region. It is delineated by the xx and ++ symbols.

Figure 8. Sonogram in the transverse plane after thyroidectomy for cancer, from a muscular man. There was no palpable mass. The image shows a rounded lymph node that was cancer. C=carotid artery, m=muscle, ++ marks the node.

Especially in children, inflammatory lymphadenopathy is common, which may complicate a search for cancerous nodes. Tuberculous cervical lymphadenitis can mimic metastatic lymph nodes from papillary thyroid carcinoma (126). Indeed, especially in a region where tuberculosis is endemic, even when a patient is known to have papillary thyroid cancer, adenopathy reportedly is more commonly due to tuberculosis than to thyroid cancer (127).

 

A source of confusion in diagnosing lymphadenopathy especially in the elderly and obese subjects is fatty change in a node that may mimic a macro-metastasis at palpation. US can offer useful insight. In one study, of 110 selected patients with a total of 247 nodes, the central “fatty”, hyperechoic hilum was quite large, extending more than one third of the transverse diameter. The ratio of the long to short axes of the node and the parenchyma to fat (P:F) were obtained. Differences between mean P:F ratio in diabetic and nondiabetic patients were significant (p=0.045). The mean P:F ratio was negatively related to body mass index (BMI) (r=0.62, p=0.015) and age (r=0.54, p=0.024). All of the nodes examined with a mean P:F ratio ≤ 1.2 (58) were found in patients older than 72 years and with a BMI higher than 27.8 (30).

 

CHARACTERISTICS OF MALIGNANT LYMPHADENOPATHY: There are ultrasonic characteristics of lymphadenopathy that correlate in a clinically useful fashion with metastases from thyroid cancer. The features that correlate most highly include microcalcifications, a spherical shape, a large cystic space, loss of the hilum, and neo-vascularization that is characterized by blood vessels penetrating the node from its periphery rather than its hilum.

The results of investigations are reasonably confirmatory. In one study of 19 patients who were referred for lymph node dissection, 578 nodes were removed, 103 of which were ultrasonically detected. The authors analyzed only the 56 nodes (28 benign and 28 malignant) that were unequivocally matched for US and pathology. The authors reported that the major criteria of malignancy were: cystic appearance, hyperechoic punctations, loss of hilum, and peripheral vascularization. If there was only cystic appearance or hyperechoic punctations, the risk of malignancy was lower but still suspicious of malignancy. They were of the opinion that nodes with “a hyperechoic hilum should be considered as benign, that peripheral vascularization has the best sensitivity-specificity compromise, and that round shape, hypoechogenicity, and the loss of hilum taken as single criterion are not specific enough to suspect malignancy”. The reported sensitivity and specificity of these criteria were 46 and 64% for round shape (long to short axis ratio < 2), 100 and 29% for the loss of fatty hyperechoic hilum, 39 and 18% for hypoechogenicity, 11 and 100% for cystic appearance, 46 and 100% for hyperechoic punctations, and 86 and 82% for peripheral vascularization (128). In several other investigations, the two most useful diagnostic characteristics are the ratio of the longitudinal to the transverse diameter of a lymph node ( L/T ratio) and the absence of a central echogenic hilum (125,129-131). In one study, the L/T ratio was less than 1.5 in 62% of metastatic nodes and greater than two 2 in 79% of reactive nodes (132). A wide cortex or narrow hilum was observed in 90% of malignant lesions, but only 45% of benign nodes. The absence of a hilum was observed in 44% of malignant lesions, but in only 8% of benign nodes. In this study the size and uniformity of a lymph node was not helpful in differentiating benign or malignant nodes.

The location of adenopathy in proximity to the thyroid in the central compartment of the neck may also be indicative of thyroid cancer. Multivariate analysis in an investigation addressing this question showed that only central location (odds ratio, 4.07; 95% confidence interval (CI), 1.64 to 10.10) and size (odds ratio, 5.14; 95% CI, 1.64 to 16.06) remained as significant corollaries of cancer (57).

 

It is not clear whether additional information about the nature of lymphadenopathy may be offered by color and spectral Doppler investigation. Although one group of investigators found that malignant nodes (29/32) more often than benign ones (6/16) demonstrate enhanced color flow signals (133), another group observed abundant color flow signals in all enlarged lymph nodes (134). There may be some diagnostic value to examining the ratio of systolic and a diastolic blood flow in a lymph node, which is called the resistive index. It has been reported that cancerous lymph nodes have a high resistive index (mean 0.92) while reactive nodes have a considerably lower value (<0.6) (134). Another investigator reported that metastatic nodes from papillary carcinoma frequently demonstrate prominent hilar vascularity similar to reactive nodes (135).

 

Among abnormal nodes that had cystic spaces, one study showed a high likelihood of papillary thyroid cancer as assessed by FNA. Cystic changes were not seen in 43 of 63 pathologic nodes that were either metastatic from other malignancies (22 patients) or benign reactive lymphadenopathy (21 patients) (136). Since cystic spaces due to necrotic material may be seen in tuberculous nodes, caution is warranted when one interprets the clinical meaning of this finding. An important diagnostic aspect of cystic masses that are lateral to the thyroid is demonstrated by one report that showed that among 37 adults (age 16-59 years), 10.8% of cervical cysts were lymphatic metastases from occult thyroid carcinoma (137). Others have reported similar observations and the point has been made that in younger patients, the lymph nodes might appear purely cystic, thereby mimicking branchial cysts (138).

 

In some studies, the addition of CT of the neck to US was found to be slightly superior to sonography alone for the detection of metastatic papillary thyroid cancer lymph nodes in the lateral compartment of the neck but not in the central compartment (139). Another investigation suggested that high-resolution ultrasound is accurate in preoperative evaluation for extra-thyroidal tumor extension and lateral lymph node metastasis. In contrast however, in this study, CT had greater sensitivity than ultrasound alone in the detection of central lymph node metastases (140).

 

In patients with suspected recurrent thyroid cancer, however, a combination of diagnostic techniques maybe necessary to differentiate a true recurrence and noncancerous images, called cryptic findings (141). Fused I-131 whole body scan SPECT when coupled with CT or PET can elucidate the nature of such images. Many of these findings prove to be inflammatory in nature, thereby avoiding unnecessary treatment with I-131 (142). Precisely directed US has been reported to enhance the specificity while maintaining sensitivity, especially in the neck and superior mediastinum (143).

 

Cytological, immunocytological, and biochemical (thyroglobulin) analysis of enlarged cervical lymph nodes, using the ultrasound-guided aspiration biopsy technique described below, can differentiate thyroid cancer metastases and inflammatory lymphadenopathy (144). It is important to add that is not necessary to require a classical cytological diagnosis of thyroid cancer in a lymph node aspirate. Any evidence of thyroid cells or the detection of thyroglobulin in the node is adequate proof of cancer; thyroid cells or thyroglobulin do not belong in non-cancerous nodes.

 

WHAT A THYROID ULTRASOUND REPORT SHOULD INCLUDE

 

The thyroid ultrasound report must answer the question that has been posed by the clinician and not be just a routine recitation. The ultrasonographer or the thyroidologist who interprets the images should note and record in the report the features listed in Table 3 and call specific attention to the features that reveal a higher than average risk of malignancy.

 

TABLE 3. ESSENTIAL ELEMENTS OF A THYROID ULTRASOUND REPORT

1. Each lobe and isthmus

A. Dimensions of Lobes (cm)

B. Shape of Lobes, (conventional shape or indentations and where they are)

C. Echogenicity of Lobes

·       Hyperechoic

·       Hypoechoic

·       Isoechoic

·       Heterogeneous

D. Vascularity of Lobes

·       Physiologic

·       Increased

·       Decreased

·       Avascular

E. Nodule(s) in Each Lobe or Isthmus

·       Location

·       Number of Nodules (1 or 2, a few, multinodular)

·       Do all nodules have uniform characteristics?

·       Does one nodule have noteworthy characteristics? *

·       Margins

o   Distinct

o   Ill-defined

·       Halo

o   Continuous

o   Discontinuous

·       Echogenicity of nodule

o   Hyperechoic

o   Hypoechoic *

o   Isoechoic *

·       Composition

o   Solid

o   Cystic

o   Complex (solid with cystic component)

·       Shape

o   Globular

o   Irregular

o   Taller-than-wide *

·       Vascularity

o   Physiologic

o   Decreased

o   Avascular

o   Increased

o   Peripheral

o   Central *

·       Calcifications

o   Punctate *

o   Coarse

o   Egg-shell

·       Other features

o   Puff-pastry “Napoleon-like” layers that are alternatingly echo-dense and echo-poor

o   Spongiform

o   Bright spot with “comet tail shadowing”

2. Lymph nodes *

·       Location

o   Ipsilateral to nodule

o   Contralateral to nodule

o   Standard levels or relation to another anatomic structure

·       Shape

o   Oval, elliptical

o   Globular *

·       Hilum

o   Fatty

o   Vascular

o   Absent *

·       Margin

o   Well-defined

o   Ill-defined *

·       Vascularity

o   Increased

o   Physiologic

o   Blood-flow from periphery rather than hilum *

·       Calcifications

o   Punctate *

o   Coarse

o   Egg-shell

·       Composition

o   Solid

o   Complex with cystic component *

·       Impact on surrounding structures

o   Deforming or infiltrating *

o   No impact

3. Extra-thyroid bed mass

·       Anatomic site (thyroglossal? sub-lingual?)

·       Ultrasonic characteristics

4. Comparison with prior examination, prior date, _____________

Comparison based on _____report or _____images?

·       Technically comparable? _____Yes  _____ No

·       Compare characteristics of lobes

·       Compare characteristics of nodules

·       Compare characteristics of nodes

*Enhanced risk of thyroid cancer

 

It is both logical and useful to separate a report into: 1) a brief statement of the reason for the US in the context of the history including pathology if any, 2) an objective narration of the findings, which represents the anatomy as defined by ultrasound, and 3) a brief, subjective, summary and conclusion or opinion. Mixing concepts 2 and 3 can be confusing to the clinician by mistaking what the interpreter sees in distinction to what he/she thinks, which may lead to variance in management.

 

There have been several attempts to codify thyroid ultrasound reports and stratify cancer-risk. An example is the Thyroid Imaging Reporting and Data Systems (TIRADS). One of them has, for example, been correlated with needle-biopsy results in 1959 thyroid nodules. The classifications were expressed as 1-5 with the following percentages of malignancy: TIRADS 2 (0% malignancy), TIRADS 3 (<5% malignancy), TIRADS 4 (5-80% malignancy), and TIRADS 5 (>80% malignancy). In a sample of 1097 nodules (benign: 703; follicular lesions: 238; and carcinoma: 156), the sensitivity specificity positive predictive value, negative predictive value, and accuracy were 88, 49, 49, 88, and 94%, respectively. The major problems of this approach are that the classifications are subjective and, as we shall see below, environmental and other factors may influence ultrasound appearance of nodules. Nevertheless, uniform, reproducible, and relevant reporting should facilitate clinical management and help the clinician to select nodules for aspiration biopsy, surgery or observation (145).

 

In one investigation, the American College of Radiology TIRADS structured reporting improved the “quality” of thyroid ultrasound reports. The authors reported an improved description of features that were predictors of malignancy. In addition, there was an increased number of definitive management recommendations that resulted in reducing the number of biopsies. In this author’s opinion, the value of management advice and reduced biopsies based on image appearance alone needs to be established on more firm evidence than is currently available (146,147). Attempts are in progress to unify thyroid ultrasound reporting features and recommendations, as well as the various TIRADS systems, across the various medical specialty societies and also internationally (148).

 

Several novel computer-based approaches taking advantage of developments in artificial intelligence for malignancy risk assessment of thyroid nodules in ultrasound images have been suggested. Local echogenic variance and boundary features are utilized to incorporate information associated with local echo distribution. Analysis of variance is performed utilizing feature vectors derived from all combinations of the characteristics under study. The classification results are evaluated with the use of receiver operating characteristics that are capable of discriminating between medium-risk and high-risk nodules (149). This promising field Is it in its infancy.

 

SONOGRAPHY IN THE PATIENT WITH A HISTORY OF HEAD AND NECK THERAPEUTIC IRRADIATION IN YOUTH

 

In the patient with a history of therapeutic irradiation to the head and neck in youth, the thyroid cancer risk may be as high as 30%. Since thyroid nodules may be detected with ultrasound before they become large enough to be palpable, sonography has been employed to screen irradiated people for tiny nodules. This selection process is quite inefficient because in the process, many more benign nodules are found than malignant ones. Furthermore, as has been observed after the nuclear event in Fukushima, Japan, the detected papillary microcarcinomas tend to be indolent (150). Consequently, some clinicians prefer not to detect micronodules contending that they are clinically irrelevant. In contrast, the author prefers to obtain a potentially useful baseline sonogram, but not to act on the presence of a micronodule unless a repeat sonogram after an interval of time demonstrates its growth or other circumstances that heighten the suspicion of malignancy. It is this author’s practice to obtain a thyroglobulin level when a micronodule is detected and again a year later in order to assess whether it has risen significantly. However, this conjecture and its validity have not been studied rigorously.

 

SONOGRAPHY TO MONITOR CHANGES IN THYROID OR NODULE SIZE

 

Changes in the size of a nodule may be clinically important, but difficult to perceive clinically. However, sonography can accurately and objectively assess changes in the volume of thyroid nodule(s) and the thyroid gland over a period of time. This is especially important during the course of therapy with thyroid hormone, in patients with a history of exposure to therapeutic irradiation, and when there is a history of thyroid cancer. Interval studies on such patients may be performed without discontinuing thyroid suppressive therapy, administering recombinant human TSH, or any preparation of the patient. Consequently, it is a simple matter to compare serial records, which may lead to changes in thyroid management earlier than palpation alone would warrant. Furthermore, since most patients tend to change doctors and residence over a period of years, an objective assessment of the size and volume of the thyroid gland or nodules will greatly facilitate the continuity of care.

 

Caution is warranted in interpreting the meaning of changes in the volume of thyroid nodules shortly after fine-needle aspiration has been performed. Bi-directional volume changes after the biopsy have been reported (151). Therefore, it is appropriate to assess nodule size at least weeks after FNA. For the same reason, to assess nodule size after a period of observation or suppressive therapy, the US should be done before another FNA is performed.

 

A Downside to Serial Sonography to Monitor Changes in Thyroid or Nodule Size/Volume               

 

Although it makes intuitive sense to repeat sonograms at some interval to detect early evidence of growth or malignant change, there is a downside to this practice too. There is indication that frequent screening with serial neck US is more likely to identify false positive abnormalities rather than significant disease. Therefore, coupling repeat US with clinically suspicious events or examination is warranted (152).

 

SONOGRAPHY IN THE PATIENT WHO HAD THYROID CANCER

 

Sonography has become a most useful imaging procedure in patients who have had either partial or complete thyroidectomy (Figure 8) (153). Sonography is done without interrupting the therapy with thyroid hormone, which is used universally in the thyroid cancer patient.

 

One study, in which 110 patients who had partial or total thyroidectomy for thyroid cancer were examined every 1-2 years, showed that ultrasonography is the most sensitive and important way to image postsurgical recurrences of thyroid carcinomas and lymphadenopathy in the neck (154). This observation is most important because recurrence in the neck is by far the most common location of reappearance of thyroid cancer. The authors suggest routine use of US in these patients.

 

Furthermore, a five-year observational study of 80 patients investigated the optimal initial follow-up strategy for patients who had near total thyroidectomy for papillary thyroid microcarcinomas (155). Sonography identified lymph node metastases not only in two thyroglobulin-positive patients but also in one thyroglobulin-negative patient. Importantly, after observation for 32 +/- 13 months after surgery, all US node-negative patients had undetectable thyroglobulin levels while on suppressive therapy and US remained negative. In contrast, whole body scanning showed no “pathological” uptake in any patient and was essentially useless, probably because differentiation of postoperative gland-remnants and tumor was not possible. Yet, radioiodine uptake in the region of the thyroid bed did correlate with recombinant human TSH (rhTSH)-stimulated thyroglobulin levels: 1 ng/ml or less in 45 patients without uptake and more than 1 ng/ml in 35 patients with uptake (r = 0.40, P < 0.0001). The authors concluded that in their population, the thyroglobulin probably derived mainly from small normal tissue remnants rather than cancer. Therefore, they contend that mild elevations of thyroglobulin are also of limited diagnostic value.

 

Sonography can detect post-operative thyroid remnants in the thyroid bed and thyroglossal region even when surgeons report a total thyroidectomy. One investigation found US remnants in 34 of 102 cases (156). This author believes that the frequency of remnants is highly experience- and surgeon-dependent.

 

It is important to appreciate that sonography may yield clinically erroneous or misleading results if it is performed during the initial several months following the surgery. During this time there may be abundant lymph nodes and heterogeneous, sono-dense regions that probably reflect postoperative changes such as edema and inflammation.

 

Sonography may serve to uncover unsuspected disease. After less than total thyroidectomy, sonography will detect nodules in the thyroid remnant, post-operative thyroid bed or in the contra-lateral thyroid lobe, which could be benign tissue or tumor. After total thyroidectomy but not following partial thyroidectomy, nodules and adenopathy are more likely to represent cancer when the concentration of thyroglobulin is elevated. Sonography may detect this disease even before it has grown sufficiently large to be palpable.

 

In patients in whom thyroid carcinoma has been diagnosed as the result of metastases to bone, lung or cervical nodes, sonography can detect an occult thyroid primary cancer even if the thyroid gland is normal to palpation.

 

One investigation has shown that rhTSH-stimulated serum thyroglobulin measurements combined with neck ultrasonography has the highest sensitivity in monitoring differentiated thyroid carcinoma in children, and many investigators believe in adults also (157). One group of investigators has reported that even when thyroglobulin levels remain low or undetectable after stimulation with rhTSH, sonography may identify lymph node metastases from thyroid cancer (158).

 

It may be difficult to differentiate a suture granuloma from recurrent thyroid cancer. A case report demonstrated a nodule that mimicked recurrent thyroid cancer on sonography and 2-{fluorine-18}-fluoro-2-deoxy-D-glucose positron emission tomography, but the diagnosis of a suture granuloma was confirmed by a US-guided fine needle aspiration biopsy (159). The ultrasonic appearance of suture granulomas includes echogenic foci larger than 1 mm in diameter (p<0.05) that are paired (p<0.05), and usually are clustered centrally or on near the middle of the nodule, unlike those in recurrent carcinomas (p<0.05) (160).

 

US is useful in the operating room during surgery. Intra-operative ultrasonography may enhance the ability to locate and resect recurrent thyroid cancer that does not accumulate radioactive iodine. Experience in seven patients suggests that sonography was particularly helpful after external beam radiotherapy to identify tumor nodules of 20 mm or less that were invasive or adherent to the airway (161). One investigation reported that intra-operative ultrasound performed by the surgeon influenced the management in 57 percent (41/72) of patients by identifying non-palpable adenopathy (162). However, one wonders if resection of non-palpable or even larger deposits of differentiated thyroid cancer will affect outcomes since historically even bilateral radical neck dissection was not associated with enhanced results when compared with thyroidectomy alone. Nevertheless, excision of non-palpable nodules that are in proximity to a vital structure could be palliative if the cancer is removed before it invades. In this author’s opinion intra-operative US could become standard to look for and remove undetected nodes after the surgeon has completed a thyroidectomy for cancer, even after a compartmental node dissection, before “closing”.

 

Preoperative sonography in the cancer patient may be associated with decreased recurrences. One group of investigators studied 275 patients who underwent pre-operative US and had a median follow-up of 41 months. They reported that patients who have had recurrence of papillary thyroid cancer were at an increased risk for subsequent recurrence of the tumor in the neck. US before the initial operation and followed by compartment-oriented surgery based on the US was related to decreased subsequent recurrence rates (162).

 

With respect to the lateral compartment of the neck, preoperative US is an excellent predictor of outcome for disease-free interval. Furthermore, a surgical approach based on preoperative US provides excellent long-term regional control (163).

 

SONOGRAPHY IN CONJUNCTION WITH NEEDLE BIOPSY

 

Fine needle aspiration biopsy of thyroid nodules and adenopathy in adults, children, and adolescents has become a major diagnostic tool that is safe and inexpensive (133,164-169). Major untoward effects are very uncommon and include bleeding (especially in patients who use anticoagulants or antiplatelet agents or those who have a bleeding diathesis), hoarseness, and infection. Many authorities, however, contend that it is reasonably safe to continue anticoagulants, including the newer novel agents, in patients who been taking these medications when performing an FNA (170). Having seen a few patients who have experienced excessive bleeding as a result of FNA while using aspirin or anticoagulants, it is this author’s practice to discontinue any agent that interferes with coagulation of blood prior to performing and FNA. Actually, there have been very occasional reports of fatal cervical hemorrhage related to FNA (171). Furthermore, even if there is no increased risk of significant hemorrhage due to FNA, the specimen maybe diluted by unnecessarily abundant red blood cells, complicating cytological interpretation. Seeding the needle track with thyroid cancer is a remote consideration (172,173).

 

Indications

 

The major indications for ultrasound-guided FNA are summarized in Tables 4 and 5. Ultrasound has made placement of the needle more accurate especially for small or complex nodules or nodes. Cytopathological interpretation is usually clinically satisfactory and promises to improve with tissue marker analysis of specimens (174). However, the accuracy of the puncture varies considerably depending on factors that are related not only to the operator and the cytologist, but also to the patient. The latter conditions include the size, homogeneity and vascularity of the nodule or node, its location in the neck, sampling errors, and the habitus of the patient. These issues affect biopsy technique.

 

Table 4. Needle Biopsy with Ultrasound Guidance is Generally Reserved For:

1. A small nodule in an obese, muscular, or large framed patient.

2. Nodules that are barely palpable or non-palpable

3. Nodule size less than one centimeter.

4. A nodule that is located in the posterior portions of the thyroid gland.

5. A dominant or suspicious nodule within a goiter.

6. All nodules that yielded non-diagnostic results on a free-hand biopsy.

7. Complex degenerated nodules if a prior biopsy without ultrasound guidance has not been diagnostic.

8. Incidentalomas that have been detected ultrasonically in patients with high risk factors for thyroid cancer such as exposure to therapeutic x-ray.

9. Small lymphadenopathy.

 

Table 5. Features That Warrant Percutaneous Fine-Needle Aspiration Biopsy of a “Solitary” Nodule or a “Special” Nodule in A Goiter

1. Clinical Features

 a. History of head and neck irradiation in youth

 b. Family history of medullary (or signs & symptoms) or less so papillary thyroid cancer

 c. Unusual firmness without calcification

 d. Growth of nodule especially during suppressive therapy

 e. Lymphadenopathy

2. Ultrasonic Features (at least two “suspicious” ultrasound features)

 a. Hypoechoic nodules with one or more of the following

  i. Irregular margins

  ii. Enhanced intranodular vascular spots (central vascularity)

  iii. Microcalcifications (punctate calcifications)

  iv. Blurred margins

  v. Taller-than-wide nodule shape

  vi. Enlargement of a nodule when compared to prior examination

 b. Lymphadenopathy (palpable or ultrasonographic)

3. In a goiter, biopsy the nodule that has “suspicious” ultrasonographic features rather than the largest nodule.

4. The size or number of nodules in a gland does not correlate with risk factors

 

Methods

 

Thyroid nodules or lymph nodes that are palpable are often biopsied directly. In some cases, correlation of the palpable anatomy with a sonographic film or screen image may be useful. In such cases, for small, complex, or deep nodules, or when a palpation-guided biopsy has resulted in an insufficient specimen, ultrasound-guided fine needle biopsy is employed (27,175), but with added cost ($289 by one estimate (176) and some inconvenience. Direct, real-time ultrasound guidance improves accuracy in puncturing the nodule. Ultrasound-guided biopsy is always required for impalpable incidentalomas and even then, it is difficult to reliably sample lesions smaller than 10 mm, as discussed previously.

 

Two methods for ultrasound-guided needle biopsy have been suggested: 1) A sonographer manipulates the transducer to locate the nodule and a second physician inserts the needle under direct vision. With practice, the assistance of a second operator is usually not required. 2) A special clamp is used to hold the transducer and fix the direction of insertion of the needle. Both require hand-eye coordination and experience is necessary to identify the spot on the skin over the target nodule to insert the needle. In our practice a dimple is produced on the skin with a blunt 1 mm wooden dowel directly over the nodule as the transducer identifies it. We have not found it appropriate to employ a "permanent marker" for this purpose, as has been suggested (177). Furthermore, this author finds the holder cumbersome and restrictive and prefers the free hand approach. With the free-hand method, the needle may be inserted parallel to, or at an angle to the ultrasound beam and at a short distance from the transducer, aiming at the nodule. The parallel approach may be technically challenging but is "comforting" to the operator because the image of the needle shaft may be viewed as it traverses the neck and into the nodule. Nevertheless, many experienced operators prefer an oblique to a perpendicular approach because of its simplicity and relatively fewer complications. The needle shaft is not imaged with this technique but its tip is seen as a very bright spot when it crosses the plane of the scan. The tip of the needle must be within the nodule during aspiration. However, even with ultrasound guidance, it is rather difficult to be certain that the tip of the needle is actually within a small nodule at the instant of aspiration, particularly if it is less than 7 or 8 mm in diameter (Figure 9).

Figure 9. Sonogram from an ultrasound guided fine needle aspiration biopsy showing a hypoechoic small nodule. The bright spot (above the arrows) is the tip of the needle within the nodule at the instant of aspiration. N=nodule.

Employing Doppler technique to identify and avoid puncturing blood vessels in the region of a nodule provides a distinct advantage of ultrasound-guided aspiration over palpation-guided biopsy. This precaution reduces the amount of blood in the aspirate and facilitates interpretation of the cytology (178). The same purpose is served by discontinuing antiplatelet and anticoagulant medication prior to a biopsy.

 

Samples of thyroid nodules and adenopathy may be obtained in either of two ways. One may aspirate the material with a syringe, employing a to and fro motion to produce a large quantity that frequently contains excessive blood, and complicates cytological examination. This author prefers the capillary technique that is done with a 25 or smaller gauge needle (without a syringe) using minimal trauma. The utility of a 27-gauge needle has been validated (179). Capillary action achieves a small, concentrated sample that remains in the needle shaft. The specimen is then expelled with an air-filled syringe quickly and gently on to a microscope slide (180). The diagnostic accuracy of the two methods is equivalent (181). One group has reported that the non-aspiration technique produces specimens of better quality and reduces inadequate results (182).

 

In this author’s experience, the capillary action aspiration method results in a superior cytological yield and the syringe/larger needle aspiration should be reserved for low-yield or fibrotic lesions.

 

Microscopic assessment of aspirates onsite for adequate cells by a cytologist at the time of the biopsy significantly reduces the number of non-diagnostic reports especially when the operator is not optimally experienced (183). It is likely that on-site assessment of cytopathologic adequacy of aspirates would help reduce the costs of needle biopsy, reportedly, by as much as 35.5%, by reducing unsatisfactory specimens that are sent to off-site cytologists (184). Furthermore, in some centers cytologists actually do the aspirations (185).

Ultrasound-guided FNA is an accurate method for identifying suspected recurrence of thyroid cancer in enlarged lymph nodes or in the thyroid bed.

 

Specimens

 

Obtaining material that is sufficient for a reliable cytological diagnosis involves competing realities. It is often necessary to do multiple punctures of a thyroid nodule to obtain enough cells even when ultrasound guided aspiration is employed. Yet, the first puncture is likely to be associated with less blood than subsequent samplings and may therefore be the best one for the cytologist to interpret. Especially for small nodules and those that are very vascular, gentle technique and point of service examination of the aspirate with a microscope to assess adequacy are important factors. In some cases when a hematoma has been produced it may be prudent to delay completing the aspiration until another day when the blood has been resorbed. Furthermore, especially when there are only a few benign-looking cells, the clinician should not be convinced that a nodule has been sampled adequately. Rather, a repeat biopsy after an interval of time may be prudent. In contrast, high suspicion is warranted when there are even a few cells that have features that are associated with malignancy. Sometimes cytology cannot suitably assess the pathological potential of a nodule. Such nodules are referred to as Atypia of Unknown Significance (AUS) or Follicular Lesions of Unknown Significance (FLUS), which will be discussed elsewhere. Caution is appropriate in accepting a report of negative cytology when the aspiration was done because a nodule grew during the course of suppressive therapy. Occasionally, when the specimen is inadequate, a better specimen maybe obtained with a needle with a larger lumen (186).

Effectiveness

 

One investigation retrospectively evaluated the effectiveness of ultrasound-guided fine-needle aspiration, in 37 patients previously treated for thyroid cancer, in identifying as cancer those cervical nodules that were suspicious of recurrence. There were 29 true positives, 6 true negatives, 1 false negative, and 1 inadequate biopsy. Therefore, US-guided biopsy had a sensitivity of 96.7%, a specificity of 100%, and an overall accuracy of 97.2% in detecting recurrence (187).

 

Caution with Respect to Negative Cytology in Children and Adults When the US is Suspicious

 

In a retrospective investigation of 35 children and adolescents, the global accuracy of FNA was 83%, with a sensitivity of 75%, and a specificity of 94%. Fourteen FNAs suggested malignancy (40%), only 1 of which was a false positive (7%). In significant contrast, 5 of the 21 FNAs suggesting benign lesions were false negatives (24%). These 5 cases had US findings suggestive of malignancy (188). Thus, a cautious approach is warranted especially in children when US findings suggest malignancy even if the cytology is benign.

 

In the postoperative thyroid bed, ultrasound-guided FNA may be particularly useful. In one series, among 21 cases there were 15 recurrent cancers, 5 benign nodules such as a parathyroid gland or regenerated normal thyroid, and 1 false positive (189).

 

There is limited ability to reliably aspirate and accurately diagnose a non-palpable nodule or node even with ultrasound-guidance (190). Ultrasound-guided cytological diagnosis of non-palpable nodules depends on the size of the lesion. One study suggested that the diagnostic yield of aspirating incidentally discovered, non-palpable 10 mm or larger thyroid nodules was high (99). Another study found that sampling of material that is adequate for cytological analysis was 64% for a 0.7-cm lesion and it increased to 86.7% when a nodule was 1.1 cm. For nodules that are 1 cm or smaller, the sensitivity was 35.8% and false-negative results were seen in 49.3% (108). In contrast, similar success has been reported in aspirating nodules that were 4 to 10 mm in size when compared with larger ones (191).

 

We have had mixed diagnostic success in sampling nodules or nodes as small as 5 mm. A few micro-cancers have been discovered in this way. The cost-effectiveness of aspirating nodules this small is uncertain considering the small (if any) clinical significance of thyroid microcarcinomas. We biopsy small lymph nodes that are “plump”. Generally, the width/depth must be almost 1 cm to yield adequate cells.

 

The cancer-predictive value of measuring thyroglobulin in the wash-out obtained from a cell-poor aspirate of nodes has been mentioned. Assaying thyroglobulin in aspirates from a thyroid nodule is not useful as an index of malignancy.

 

Suspicious Nodules in Goiters

 

It has been reported from a goiter zone in Italy that as many as 52% of histological malignant nodules in goiters were found only with the aid of ultrasound-guided FNAB. Therefore, the authors concluded that ultrasound-guided aspiration should be used in areas where multinodular goiter is endemic to assess nodules that are deemed suspicious by virtue of a hypoechoic pattern, a "blurred halo", micro-calcifications, or an intranodular color Doppler signal (192). In another report of patients with endemic goiter, 44 were selected for surgery based on suspicious ultrasonography and among 24 of them who had a “cold” nodule, aspiration biopsy revealed 2 with papillary cancer and surgery disclosed 2 more cases of papillary cancer and one case of insular cancer (193).

 

Predictors

 

One group has investigated the predictors and optimal follow-up strategy for initial non-diagnostic ultrasound-guided FNAs of thyroid nodules. Among 1,128 patients with 1,458 nodules that were biopsied over a 6-yr period, 1,269 aspirations (950 patients) were diagnostic, and 189 (178 patients) were non-diagnostic. The authors reported that the only significant independent predictor of non-diagnostic cytology (P < 0.001) was a cystic content of each nodule and the fraction of specimens with initial non-diagnostic cytology increased with greater cystic space. As emphasized above, for pathologic lymph nodes, in distinction to thyroid nodules, cystic degeneration is typical of thyroid cancer metastases. For example, diagnostic ultrasound-guided FNA was obtained on the first repeat biopsy in 63% of nodules and was inversely related to increasing cystic content of each nodule (P = 0.03). One hundred and nineteen patients with 127 nodules returned for follow-up as advised, and malignancy was documented in 5% (194).

 

For non-palpable thyroid nodules, the relative importance of sonographic features as risk factors of malignancy and the use of ultrasound-guided aspiration cytology was studied in 494 consecutive patients with nodules between 8-15 mm. It is noteworthy that 92 patients (19%) had inadequate cytology and were excluded from the study. Cancers were observed in 18 of 195 (9.2%) solitary thyroid nodules and in 13 of 207 (6.3%) multinodular goiters. The prevalence of cancer was similar in nodules greater or smaller than 10 mm (9.1 vs. 7.0%). The authors recommended that ultrasound-guided FNA should be performed on all 8-15 mm hypoechoic nodules with irregular margins, intranodular vascular spots or microcalcifications (194). In another study, among 402 patients with 8 mm to 15 mm non-palpable nodules, the cancers were most likely to be hypoechoic and solid, and have microcalcifications, irregular borders, or central blood flow. Since 125 (31 %) of nodules met those criteria, biopsies could be avoided in 69 percent of nodules, incurring a risk of missing 13 percent of the cancers (111).

 

It would appear that that no single parameter satisfactorily identifies the subset of patients whose nodule should be subjected to biopsy. In one investigation of 6,136 nodules in 4,495 patients, the best compromise between missing cancers and cost-benefit was achieved with at least two “suspicious” ultrasound features. The most useful were nodule shape (taller-than-wide), microcalcifications, blurred margins, and a hypoechoic pattern (195). Enhanced intranodular blood flow on Doppler examination also was reported as a helpful criterion (81). Another investigation of 1,141 nodules reported that logistic regression analysis showed that the size of the nodule affected the utility of ultrasonic characteristics of nodules in assessing cancer risk and selection for needle biopsy. In nodules smaller than 15 mm in size, hypoechogenicity (odds ratios, OR: 3.18), microcalcifications (OR: 19.12), solitary occurrence (OR: 3.29) and height-to-width ratio ≥1 (OR: 8.57) were independent risk factors for malignancy. The authors concluded that all lesions presenting at least one of the above-mentioned features should be biopsied (sensitivity 98%, specificity 44%). With nodules larger than 1.5 cm, the mentioned selection criteria were less sensitive than for smaller nodules. Useful features included, hypoechogenicity, taller than wide or microcalcifications (sensitivity 84%, specificity 72%) (196).

 

It is difficult to decide which nodule in a multinodular goiter to biopsy. Guidelines include selection by size, the ultrasound characteristics mentioned above, and most importantly nodules that are clinically suspicious. Perhaps one may be reassured that the pathology is likely benign when there are very many nodules in a goiter rather than a few. In one investigation of thyroid nodules that underwent ultra-sound-guided FNA, the authors found that the cancer risk is similar for patients with one or two nodules (over 1 cm) and decreases with three or more thyroid nodules (197).

 

It is particularly difficult to effectively select nodules for biopsy in an endemic goiter zone where nodules are ubiquitous. In one investigation in an iodine deficient region, a numeric score was assigned to nodules based on ultrasonic high-risk of cancer. Among 2,642 consecutive patients (3,645 nodules), nodules with a score of over 5.5 out of 10 had a 66% sensitivity and a 76% specificity for cancer, which was much higher than for those with lower scores. The data strongly facilitated the decision of which nodules to biopsy (94).

 

Combining the results of cytology and the tumor marker thyroglobulin after a patient has had a total thyroidectomy may enhance the accuracy of either single predictor of residual/recurrent thyroid cancer. One investigation reported that among 340 consecutive patients with differentiated thyroid carcinoma, who had been treated with near-total thyroidectomy, 131-I thyroid ablation, and TSH suppressive doses of L-thyroxine, rhTSH-stimulated thyroglobulin alone had a diagnostic sensitivity of 85% for detecting active disease and a negative predictive value of 98.2%. After adding the results of neck ultrasound, the sensitivity increased to 96.3%, and the negative predictive value to 99.5% (198). However, in one study, US and FNA did not seem useful to detect recurrent papillary thyroid cancer when the serum thyroglobulin level was undetectable (199).

 

One should be somewhat more suspicious that an incidentaloma could be cancerous when the patient has another non-thyroid cancer. In one investigation of 41 patients who had another cancer and who had an incidentally discovered thyroid nodule, surgical pathology revealed 4 papillary thyroid cancers, 4 microscopic papillary thyroid cancers, 2 metastatic cancers, and 7 benign lesions (200).

 

Not Biopsying Nodules that are Not Likely Malignant by US Criteria

 

Several studies have recognized sonographic morphological patterns that correlate with benign thyroid disease. The authors advise not biopsying these nodules or goiters in the interest of cost-effectiveness (201-203). In one study, 650 patients were identified for whom both a pathology report and ultrasound images were available. From an alphabetized list, the first 500 nodules were reviewed retrospectively. Most of the diagnoses were based on cytological rather than histological findings. Four patterns associated with benign disease were identified and seemingly attributed to colloid: spongiform configuration, cyst (cystic), a “giraffe pattern” (light blocks separated by black bands), and diffuse hyperechogenicity (201). One characteristic has borne the test of time: thyroid cancer is rarely if ever hyperechogenic.

 

It is useful to know that one group has reported that not performing a biopsy on nodules less than 5 mm in size seemed safe because when they underwent later surgery because of a 2 or 3 mm enlargement, there was no evidence of distant metastases or fatalities (204).

 

The rationale supplied by the authors (201,204) for not biopsying these nodules is that fewer biopsies, will lead to less delay of “necessary” biopsies and less false-positives. This author completely agrees with not biopsying non-suspicious nodules unless there are other factors that indicate cancer-risk. Fortunately, small low risk nodules generally do not adversely affect quality of life or survival. However, the practical outcome of this “leave the nodule alone” philosophy may result in a mind-set that, in this author’s opinion, should be avoided. The difference between focusing clinical attention on biopsying suspicious nodules and confidently dismissing nodules that “can be left alone” may result in a difference in the risk of missing a cancer. The outcome of the difference is similar to misjudging that a dog may bite, and giving it wide berth to avoid getting bitten, and mistaking that a dog does not bite and getting mauled.

 

Thus, I feel that we should not use tentative data from limited investigation to make a pivotal decision not to biopsy certain thyroid nodules and selection against surgery. A simple, logical, safe, inexpensive, and more reliable clinical attitude is employing sonography to enhance the efficiency and accuracy of biopsying ultrasonically suspicious nodules and nodules that have clinical or historical features that are associated with higher than average cancer-risk, and paying reasonable but not invasive attention to the rest of the nodules and the gland as a whole.

 

If the pattern approach to selecting nodules not to biopsy is employed by ultrasonographers, they should be cognizant that cancerous thyroid nodules in a radiation-exposed population may often not exhibit the classic ultrasonic features of malignancy. Rather, benign characteristics are more often encountered. Therefore in this setting especially, benign-looking nodules should be biopsied and not “left alone” (205).

 

Repeat Aspiration Biopsy in Patients with a Previously Benign Result

 

There is no consensus about how often FNA should be repeated after previous aspirations have indicated benign disease. Considerations that enter the decision include periodic US, historical risk factors, changes in physical examination, and even the patient’s or the physician’s level of anxiety. It seems reasonable that growth of the volume of the nodule, the emergence of adenopathy, or symptoms that suggest pressure on cervical structures such as hoarseness or dysphasia should be viewed with suspicion. Sometimes the observations of an ENT consultant may influence management. In this author’s experience routine re-aspiration rarely results in a discovery of malignancy. In a retrospective review of records of patients seen at the Mayo Clinic between January 2003 and December 2013 of 334 nodules with benign FNA, 85.3% were benign, 7.2% suspicious, 5.7% non-diagnostic, and 1.8% malignant. Importantly, the repeat FNA altered clinical management in only 9.5% of cases (206).

 

Non-Cytologic Examination of Aspirates

 

Ultrasound-guided aspiration can facilitate biochemical analysis. Needle washings of adenopathy (not applicable to thyroid nodules) may contain Tg, revealing papillary thyroid cancer even when there are insufficient or inadequate cells. It is noteworthy that assay of Tg in tissue is reportedly not effected by serum anti-thyroglobulin antibodies (207). Furthermore, the aspirate of nodules or lymph nodes may contain calcitonin in medullary cancer, a tumor marker such as galectin-3 (208) in papillary thyroid cancer, or lead to a non-neoplastic diagnosis such as tuberculosis (209) or amyloidosis (210). One anticipates that one day aspirates may be studied routinely for biochemical products, sub-cellular components, and, bacteriologic, fungal, or viral material. Examination for molecular genetic tumor markers will be discussed elsewhere.

 

Core Biopsy

 

There is also interest in sonographically-guided core biopsy of thyroid nodules. One group has concluded that percutaneous acquisition of tissue for histological rather than cytological evaluation is an accurate and safe alternative to aspiration biopsy in the assessment of thyroid nodules (211). However, one needs to be aware that there may be greater risk from core biopsy, including an occasional fatal case, in contrast to fine needle aspiration biopsy (170). Other investigators have reported on the use of an ultrasound-guided special compound needle that can accomplish both aspiration and core biopsy and suggest its use when prior aspiration has been unsuccessful (212).

 

SONOGRAPHY BY THE THYROID SURGEON

 

Although preoperative thyroid ultrasonography is not essential for successful surgery, many surgeons have come to recognize that it may be useful to identify pre-operatively suspicious lymph nodes in patients with biopsy-proven papillary thyroid cancer. Indeed, respected surgical authorities assert that ultrasound is an essential modality in the evaluation of thyroid malignancy and that surgeon-performed ultrasound has proved invaluable in the preoperative, intraoperative and postoperative setting (213-215). It has become increasingly popular for surgeons personally to perform a pre-operative sonogram since metastatic disease may not be clinically apparent to them intra-operatively. Preoperative identification of metastatic disease by cervical ultrasound may result in altering the surgical approach in as many as 40 percent of patients (27,28,97,192,216,217). Furthermore, pre-operative thyroid ultrasonography followed by compartment-oriented surgery may decrease recurrence rates in patients if performed before their primary operation (162). It is noteworthy that ultrasound guided FNA for thyroid nodules has been incorporated into some general surgery residency programs. (218).

 

Preoperative Labeling Lymph Nodes or Intraoperative US may Facilitate Intraoperative Identification and Removal of Adenopathy

 

It may be difficult for a surgeon to identify at surgery a small node that was discovered by preoperative ultrasonography. Insertion of a hook 20-gauge needle into a US-suspicious lymph node pre-operatively facilitates identification and removal of the pathological lesion (219-221). Alternatively, pre-incision, ultrasound-guided injection of blue dye into abnormal lymph nodes was very useful in the re-operative neck to facilitate their safe and efficient removal in one study (222). Other investigations have employed ultrasound-guided, preoperative injection of charcoal suspensions to tattoo the lesion. The rate of success is reportedly as high as 84-96% in small studies. However, in 1 case the charcoal was found several centimeters away from the lesion, tattooing a lesion behind a large blood vessel has not been achieved, and in 2 of 55 patients a charcoal dot remained in the skin after the procedure. There were no reported serious adverse effects (223,224). In strong contrast to this approach, other surgeons eschew selective removal of nodes in favor of classical compartmental dissection.

 

Intraoperative sonography may be very useful (161,225,226). In 26 of 31 patients with papillary thyroid cancer who had preoperative sonographic identification of adenopathy, intraoperative palpation did not locate adenopathy but intraoperative ultrasonography located and facilitated removal of the lesions (smaller than 10 mm in diameter) in all patients (225).

 

A method that may help find a thyroid sentinel node preoperatively has been reported in a porcine experimental model. US contrast agent and methylene blue dye were injected trans-cutaneously into the thyroid glands of pigs and draining lymphatic channels and sentinel lymph nodes were identified ultrasonically. Subsequently, a sentinel node biopsy was conducted; bilateral neck and upper mediastinal dissection was performed. The lympho-sonography of the thyroid gland in this porcine model correlated well with blue dye-guided sentinel node surgical biopsy. If applied to humans, this technique might potentially enable a detailed analysis of thyroidal lymphatic drainage and enhance thyroid cancer surgery (227).

 

SONOGRAPHY IN CONJUNCTION WITH PERCUTANEOUS THERAPEUTIC INTERVENTION

 

After an aspiration and cytology have demonstrated that a nodule is benign, ultrasound-guided puncture of a nodule may have a role in therapy to deliver medication or other therapy precisely into the lesion and to spare the surrounding tissue.

 

Percutaneous injection of ethanol has been used to reduce the function of autonomous thyroid nodules (228). One investigation has observed 34 patients, for up to three years, who had percutaneous ethanol injection of autonomous thyroid nodules. The patients required 1-11 sessions of 3-14 ml of ethanol injection (total amount of ethanol per patient: 20-125 ml). The authors report recovery of extra-nodular uptake on isotope scan and normalization of TSH levels within 3 months from the end of the treatment in 30/34 patients and an average reduction in nodule volume of 62.9%. 4/34 patients were refractory to the treatment, 3 of whom had had nodule volumes > 60 ml. There were no recurrences during 6 to 36 months of observation (229). Another study examined 20 patients with autonomous thyroid nodules for 763 +/- 452 days after ethanol injection. A mean of 2.85 +/- 1.1 injections per patient, and a mean volume of 4.63 ml of ethanol were required (nodule volume-dependent). After a mean time of 50 +/- 23 days TSH normalized and was maintained in 16 patients (80%), whose nodular volume reduced 60.8%. Four patients (20%) did not completely respond to the treatment (230). Less impressive but “clinically acceptable” results have also been observed in a study that reported a "complete cure" in only 22 of 42 patients (52%), mainly in small nodules, and little or no hormonal response in 4 patients (9%). However, nodule volume decreased in all cases and there were no recurrences or serious adverse effects (231). In the reported series, "mild to moderate" local pain often occurred after the injections and lasted a day or two. Local hematomas were seen. Major complications like permanent dysphonia or vascular thrombosis seem to be very uncommon. However, transient paralysis of the recurrent laryngeal nerve may occur. Thus this technique may be an option for large, but not very large autonomous nodules that cannot or should not be treated surgically or with I-131 (232).

 

Percutaneous injection of ethanol has also been used to treat toxic nodular goiter (231,233) and thyroid masses that are recurrent after non-toxic nodular goiters have been treated surgically (233), with results that are similar to those described above.

 

Recurrent cysts, and cystic spaces in a degenerated solid lesion have been obliterated in this fashion (234,235). Perhaps the procedure will have use in cosmetically unacceptable or very large structures. Prospective studies will be required to ascertain if ultrasound-guided placement of medication will reduce the intensity or duration of pain after the injection and improve success over palpation-directed injection.

 

Sonographically guided percutaneous ethanol injection is a treatment option for patients with cervical nodal metastases from papillary thyroid cancer that are not amenable to further surgical or radioiodine therapy. In a study of 21 metastatic nodes in 14 patients, all treated lymph nodes decreased in volume, some impressively. No major complications occurred in this series (116). Yet, in other studies severe untoward effects have been reported including necrosis of the larynx and adjacent skin due to ethyl alcohol (236). It seems that this option may be palliative when there are large nodes that threaten to impact on surrounding structures. However, since ethanol-treated nodes may increase in size due to inflammation, caution is warranted especially when there are bulky nodes in the thoracic inlet or adjacent to vital structures.

 

Greater use of percutaneous administration of ethanol for a variety of benign and malignant conditions seems likely. However, prudence dictates that the injection should only be used when essential and not as an optional therapy to reduce the size of routine cysts, euthyroid nodules and goiters, or even non-threatening malignant nodules.

 

Thermal ablation techniques for benign thyroid nodules, toxic adenomas and even for papillary microcarcinomas are increasingly used, particularly in Korea and Europe (237). Both radiofrequency ablation (RFA) and laser ablation are used and have been shown to be efficient, cost-effective, and to have a low rate of complications (238,239). For example, one investigation evaluated the efficacy of ultrasound -guided laser thermal ablation in reducing the volume of hypofunctioning benign thyroid lesions that caused local compression symptoms or patient-concern in 20 patients, when the patients refused or were ineligible for surgical treatment. A 75-mm, 21-gauge spinal needle was inserted into the thyroid gland under ultrasound-guidance, and a flat-tipped 300-micron quartz fiberoptic guide was placed into the tissue that was to be destroyed with a 1.064-micron continuous-wave neodymium yttrium-aluminum-garnet laser for 10 minutes. Ultrasonograms were used to assess the decrease in nodule volume at 1 month and 6 months after therapy. The mean nodule volume decreased from a baseline value of 24.1 +/- 15.0 mL to 13.3 +/- 7.7 mL at 1 month (43.8 +/- 8.1%) and to 9.6 +/- 6.6 mL at 6 months (63.8 +/- 8.9%). Untoward effects included burning cervical pain, which rapidly decreased after the laser energy was turned off and treatment with betamethasone for 48 hours in 3 patients. No patient had local bruising, cutaneous burning, or dysphonia (240).

 

In a multicenter study on 44 patients with toxic adenomas or autonomous nodules and a follow-up of 19.9±12.6 months showed that the mean  nodule volume decreased from an initial volume of 18.5±30.1 ml to 11.8±26.9 ml at 1 month and to 4.5±9.8 ml at the last month (241). The thyroid function tests improved significantly and 35 of the hot nodules became cold or normal when followed by scintigraphy, and 9 had a decreased uptake. There were no major complications.

 

Several studies have shown that radiofrequency ablation may be an alternative to active surveillance for papillary thyroid microcarcinomas (PTMC). For example, in a study involving 107 patients, the mean volume reduction ratio at 18 months was 0.999 ± 0.002 (range: 0.992-1) at 12 months (242). Thyroid function tests remained normal, and there was no tumor regrowth, local recurrence, or distant metastases during follow-up visits. In an Korean study with 152 biopsy-proven PTMCs from 133 patients complete disappearance was found in 91.4% (139/152) of ablated tumors (mean follow-up 39 months) (243). All patients were either of high surgical risk or refused to undergo surgery. In the 13 tumors that did not show complete disappearance, none of the PTMC showed regrowth of the residual ablated lesion during the follow-up period, and there were no local recurrences, lymph node or distant metastases in any of the patients. The complication rate was 3% (4/133), including one voice change.

 

SONOGRAPHY TO DISCOVER PELVIC THYROID TISSUE

 

Trans-vaginal and trans-abdominal pelvic sonography has been employed to identify a 16 cm mass in the right adnexa that was a cystic teratoma, a struma ovarii, containing a 5 mm focus of papillary cancer within the thyroid tissue (244).

 

SONOGRAPHY OF THE FETAL THYROID

 

Ultrasonography in pregnancy can be become a helpful tool to assess thyroid status in utero. Gestational age-dependent and age-independent nomograms for fetal thyroid size have been developed by performing ultrasonograms in 200 fetuses between 16 and 37 weeks of gestation (245). Fetal hyperthyroidism can be detected by the presence of increased blood flow within a goiter in contrast to peripheral vascularity when goiters are associated with hypothyroidism (246). Fetal goiters and hypothyroidism have been studied, and successful treatment has been reported (247). It is thought that intrauterine recognition and treatment of congenital goitrous hypothyroidism may reduce obstetric complications and improve the prognosis for normal growth and mental development of affected fetuses. One report cited a fetal goiter diagnosed at 29 weeks of gestation during routine ultrasound examination. Fetal blood sampling performed at this time documented fetal hypothyroidism and treatment was given using a series of intra-amniotic injections of tri-iodothyronine and subsequently, thyroxine. Following birth, neonatal serum TSH levels were within the reference range (248). A case of fetal goitrous hypothyroidism associated with high-output cardiac failure was diagnosed at 32 weeks of gestation based on ultrasound examination. The fetus' thyroid function was examined by amniocentesis and cordocentesis. The fetus was treated by injection of L-thyroxine into the amniotic fluid at 33 weeks of gestation. Thereafter, the goiter decreased in size, and the high-output cardiac failure improved (249). Similarly, fetal goiter and hypothyroidism that resulted from the treatment of maternal Graves’ disease with propylthiouracil was diagnosed with trans-vaginal US and treated by levothyroxine injection into the amniotic fluid. Successful ultrasound-facilitated treatment of fetal goitrous hypothyroidism has been reported in 12 cases (250). Assessing the fetal thyroid size ultrasonically may also be beneficial in adjusting the dose of antithyroid drug in mothers with Graves’ disease and in preventing fetal and neonatal goiter and hypothyroidism, as discussed before (67). In addition, determining fetal thyroid size with ultrasonography in mothers with a history of Graves' disease has been reported to facilitate achieving normal fetal thyroid function (251).

 

SONOGRAPHY OF THE THYROID IN THE NEWBORN

 

There are several uses of ultrasonography in newborn infants. Normative data in 100 (49 male) healthy term Scottish neonates showed a mean thyroid length of 1.94 cm (SD 0.24, range 0.9-2.5), width of 0.88 cm (SD 0.16, range 0.5-1.4), depth of 0.96 cm (SD 0.17, range 0.6-2.0), and volume of 0.81 ml (SD 0.24, range 0.3-1.7) (252). There was considerable variation (-0.8 to + 0.7 ml) between the two lobes in individual babies. Another investigation revealed that the ratio of thyroid width to tracheal width is a simple, practical parameter for estimating the size of the thyroid gland in neonates and small children (253).

 

In permanent primary congenital hypothyroidism, ultrasonography has been reported to identify 66 instances where the thyroid gland was not located in the usual anatomical position and hemiagenesis in one case. The diagnosis was confirmed by scintigraphy. The authors concluded that sonography might be used as the first imaging tool in patients with congenital hypothyroidism, but scintigraphy should be used to distinguish agenesis from ectopia (254).

 

EPIDEMIOLOGICAL USE OF ULTRASONOGRAPHY

 

Ultrasonography has been used effectively even in the field in undeveloped areas to evaluate thyroid anatomy and size in iodine-deficient regions or to search for cancer in radiation-exposed populations. Inter-observer agreement on estimates of thyroid volume has been good in epidemiologic studies but agreement on echogenicity has been poor (29). One study has revealed that in the Chernobyl population thyroid cancers often exhibit benign ultrasound characteristics, that malignant features are uncommonly encountered, and as many nodules as is feasible should be biopsied (205). Correlation of age, body size and thyroid volume in endemic goiter areas have been reported (255). Data for thyroid volumes that are specific to a geographic region, iodine status, sex, and pubertal stage may be more appropriate than a single age-specific international reference (256,257). Systematic ultrasound screening has been found useful in Belarus for the early detection of thyroid carcinoma in children 4-14 years of age who were exposed to radioactive fallout due to the Chernobyl accident (258).

 

After the nuclear accident in the Fukushima Daiichi Nuclear Power Plant, large-scale ultrasound screening has been implemented (for review see (259)). This led to a high rate of detection of thyroid cancer in younger individuals within the studied cohort of approximately 300,000 subjects in Fukushima prefecture. This observation resulted in significant concerns in the population because it was felt that these cancers might have been caused by radiation. The current evidence indicates, however, that these findings are largely explained by the effect of screening.

 

Ultrasonography has also shown that the prevalence of thyroid cancer has not increased in a population exposed to the accidental release of I-131 in Hanford, Washington during 1944-1957 (260). Ultrasonography has also been used to monitor thyroid nodule development among workers in nuclear power plants (261).

 

The value of ultrasonographic mass screening to uncover thyroid carcinoma depends on the cancer-risk status of the population. In a population with average cancer risk the value of screening is controversial because of the presumed low benefit/cost of the screening as contrasted with subsequent discovery of the small number of tumors that will progress to palpable, clinical, but low-virulence tumor. One group studied 1401 women who were scheduled to undergo a breast examination. Thyroid nodules were detected in 25.2% and thyroid cancer in 2.6% of all subjects. The size of the tumors was significantly smaller in the ultrasound-studied group than that of a clinically detected cancer group (P < 0.05) (262). Another group studied thyroid sonography in 5549 patients who were undergoing breast sonography. Forty-two (0.76%) thyroid cancers were found; all were papillary carcinomas. The incidence of thyroid cancer was significantly higher in the group with breast cancer than in the group who did not have breast cancer (37). In contrast, epidemiologic investigation of the long-term risk of developing thyroid cancer has been useful in a population with a higher risk of cancer such as irradiated people. In a prospective ultrasound examination of 2637 atomic-bomb survivors the hazard ratio for cancer development was significantly high at 23.6 (95% confidence interval, 7.6-72.8) and even higher, 40.2 (95%, confidence interval, 9.4-173.0) in 31 people who initially had cytologically benign solid nodules. The hazard ratio was only 2.7 (95% confidence interval, 0.3-22.2) in 121 subjects who had thyroid “cysts”. Importantly, sex, age, TSH level, thyroglobulin level, radiation dose, nodule volume, and increase in nodule volume did not predict cancer development in the solid nodule group but sonography did reveal the risk of cancer (263).

 

ELASTOGRAPHY

 

Ultrasonography can estimate the rigidity or stiffness of tissue, which is called elastography. The deformability of a tissue may be assessed from a change in Doppler signal in response to externally applied pressure or vibrations, or by tracking shear wave propagation. This phenomenon may correlate with palpable consistency and cytology of a nodule or goiter. The technique may enhance the cancer-predictive value of sonography of non-cystic non-calcified thyroid nodules (75,264-267). However, it is premature to judge the clinical value of the test and the literature contains controversial data. One retrospective investigation revealed that among 16 malignant and 20 benign thyroid nodules elastography correlated with FNA in a sensitivity of 100% and specificity of 75.6% in detecting malignant thyroidnodules (267). Other investigators reported that elastography is not able to select cancers among follicular lesions of indeterminate significance (268). Elastography was not very useful in detecting thyroid cancer in patients affected by Hashimoto’s thyroiditis (269). Elastography may be useful in the diagnosis of inflammatory conditions of the thyroid like sub-acute thyroiditis (270).

 

OTHER USES OF ULTRASONOGRAPHY

 

There have been other novel and inventive applications of ultrasound to thyroidology and the list grows. Just as medical practices have evolved as a result of sonography, surgical techniques may change as well, as was discussed previously. Intra-operative diagnostic sonography is already used in the patient with thyroid cancer and one suspects that it will impact favorably on surgical methods, complications and outcome. Another example of the potential is a recent report that used ultrasonography to demonstrate that routine insertion of drains into the thyroid bed to prevent formation of hematoma or seroma following thyroid surgery may not be necessary. The authors contended that not draining the wound did not adversely influence the volume of the sequestered fluid (p = 0.313) and actually was beneficial by reducing morbidity and decreasing hospital stay (p = 0.007) (271).

 

Thyroid sonography may also be useful before neck surgery for non-thyroid disease. In a retrospective study of 1200 consecutive patients who were treated surgically for primary head and neck tumors and who had routine preoperative neck ultrasound by the surgeon, 47%, (477/1195) of the patients had coexisting thyroid disease. Preoperative fine-needle biopsy of sonographically detected thyroid nodules was performed in 20%, which was cost-effective in limiting concomitant thyroid surgery to fewer patients (6%; 21/350) (272).

 

Ultrasonic energy can be used therapeutically to destroy tissue, as discussed previously, and also to activate mechanical equipment. An example of the latter is ultrasonically activated shears for thyroidectomy that have been reported not to increase complications, shorten operative time, improve cosmetic results, and reduce the patient’s pain, without greater expense than conventional methods (273). An ultrasonically activated scalpel significantly improved bleeding control during thyroid resections and may also be beneficial with respect to cost containment by reducing operative time (274). Ultrasound-guided percutaneous interventional procedures to deliver medications, enzymes, recombinant materials such as RNAS, monoclonal antibodies, or energetic forces to the thyroid gland, nodules, or nodes also challenge the imagination. US-guided high-intensity, focused ultrasound maybe use to ablate benign thyroid nodules (275).

 

REFERENCES

 

  1. Cooper DS, Doherty GM, Haugen BR, Kloos RT, Lee SL, Mandel SJ, Mazzaferri EL, McIver B, Pacini F, Schlumberger M, Sherman SI, Steward DL, Tuttle RM. Revised American Thyroid Association management guidelines for patients with thyroid nodules and differentiated thyroid cancer. Thyroid : official journal of the American Thyroid Association. 2009;19(11):1167-1214.
  2. Blum M. Evaluation of thyroid function; sonography, computed tomography and magnetic resonance imaging. Principles and Practice of Endocrinology and Metabolism. 1990:289-293.
  3. Ishigaki S, Shimamoto K, Satake H, Sawaki A, Itoh S, Ikeda M, Ishigaki T, Imai T. Multi-slice CT of thyroid nodules: comparison with ultrasonography. Radiation medicine. 2004;22(5):346-353.
  4. The thyroid nodule. Annals of internal medicine. 1982;96(2):221-232.
  5. Parikh K, Davenport MS. Net Revenue Analysis of Inpatient and Emergency Department Thyroid Ultrasound at a US Quaternary Care Center From 2012 to 2015. Journal of the American College of Radiology : JACR. 2018;15(1 Pt A):75-81.
  6. Butch RJ, Simeone JF, Mueller PR. Thyroid and parathyroid ultrasonography. Radiologic clinics of North America. 1985;23(1):57-71.
  7. Leopold GR. Ultrasonography of superficially located structures. Radiologic clinics of North America. 1980;18(1):161-173.
  8. Rago T, Bencivelli W, Scutari M, Di Cosmo C, Rizzo C, Berti P, Miccoli P, Pinchera A, Vitti P. The newly developed three-dimensional (3D) and two-dimensional (2D) thyroid ultrasound are strongly correlated, but 2D overestimates thyroid volume in the presence of nodules. Journal of endocrinological investigation. 2006;29(5):423-426.
  9. Jakobsen JA. Ultrasound contrast agents: clinical applications. European radiology. 2001;11(8):1329-1337.
  10. Menna S, Di Virgilio MR, Burke P. Ultrasonography contrast media Levovist and power Doppler in the study of the breast. Methodology, vascular morphology and automatic enhancement quantification with wash-in and wash-out curves. La Radiologia medica. 1999;97(6):472-478.
  11. Wang Y, Nie F, Liu T, Yang D, Li Q, Li J, Song A. Revised Value of Contrast-Enhanced Ultrasound for Solid Hypo-Echoic Thyroid Nodules Graded with the Thyroid Imaging Reporting and Data System. Ultrasound in medicine & biology. 2018;44(5):930-940.
  12. Clark KJ, Cronan JJ, Scola FH. Color Doppler sonography: anatomic and physiologic assessment of the thyroid. Journal of clinical ultrasound : JCU. 1995;23(4):215-223.
  13. Foley WD, Erickson SJ. Color Doppler flow imaging. AJR American journal of roentgenology. 1991;156(1):3-13.
  14. Brunese L, Romeo A, Iorio S, Napolitano G, Fucili S, Biondi B, Vallone G, Sodano A. A new marker for diagnosis of thyroid papillary cancer: B-flow twinkling sign. Journal of ultrasound in medicine : official journal of the American Institute of Ultrasound in Medicine. 2008;27(8):1187-1194.
  15. Iacobone M, Citton M, Pagura G, Viel G, Nitti D. Increased and safer detection of nonrecurrent inferior laryngeal nerve after preoperative ultrasonography. The Laryngoscope. 2015;125(7):1743-1747.
  16. Shapiro RS. Panoramic ultrasound of the thyroid. Thyroid : official journal of the American Thyroid Association. 2003;13(2):177-181.
  17. Choi YJ, Baek JH, Hong MJ, Lee JH. Inter-observer variation in ultrasound measurement of the volume and diameter of thyroid nodules. Korean journal of radiology. 2015;16(3):560-565.
  18. Peeters EY, Shabana WM, Verbeek PA, Osteaux MM. Use of a curved-array transducer to reduce interobserver variation in sonographic measurement of thyroid volume in healthy adults. Journal of clinical ultrasound : JCU. 2003;31(4):189-193.
  19. Brauer VF, Eder P, Miehle K, Wiesner TD, Hasenclever H, Paschke R. Interobserver variation for ultrasound determination of thyroid nodule volumes. Thyroid : official journal of the American Thyroid Association. 2005;15(10):1169-1175.
  20. Lyshchik A, Drozd V, Reiners C. Accuracy of three-dimensional ultrasound for thyroid volume measurement in children and adolescents. Thyroid : official journal of the American Thyroid Association. 2004;14(2):113-120.
  21. Nakamura H, Hirokawa M, Ota H, Kihara M, Miya A, Miyauchi A. Is an Increase in Thyroid Nodule Volume a Risk Factor for Malignancy? Thyroid : official journal of the American Thyroid Association. 2015;25(7):804-811.
  22. Deveci MS, Deveci G, LiVolsi VA, Gupta PK, Baloch ZW. Concordance between thyroid nodule sizes measured by ultrasound and gross pathology examination: effect on patient management. Diagnostic cytopathology. 2007;35(9):579-583.
  23. Vejbjerg P, Knudsen N, Perrild H, Laurberg P, Pedersen IB, Rasmussen LB, Ovesen L, Jorgensen T. The association between hypoechogenicity or irregular echo pattern at thyroid ultrasonography and thyroid function in the general population. European journal of endocrinology. 2006;155(4):547-552.
  24. Solivetti FM, Papini E, Misischi I, Palermo A, Pantano AL, Bizzarri G, Papini L, Guglielmi R. "Hypoechoic triangle": a new sonographic sign or marker of advanced autoimmune thyroiditis. Thyroid : official journal of the American Thyroid Association. 2011;21(3):285-289.
  25. Trimboli P, Rossi F, Condorelli E, Laurenti O, Ventura C, Nigri G, Romanelli F, Guarino M, Valabrega S. Does normal thyroid gland by ultrasonography match with normal serum thyroid hormones and negative thyroid antibodies? Experimental and clinical endocrinology & diabetes : official journal, German Society of Endocrinology [and] German Diabetes Association. 2010;118(9):630-632.
  26. Goepfert RP, Liu C, Ryan WR. Trans-oral robotic surgery and surgeon-performed trans-oral ultrasound for intraoperative location and excision of an isolated retropharyngeal lymph node metastasis of papillary thyroid carcinoma. American journal of otolaryngology. 2015;36(5):710-714.
  27. Marqusee E, Benson CB, Frates MC, Doubilet PM, Larsen PR, Cibas ES, Mandel SJ. Usefulness of ultrasonography in the management of nodular thyroid disease. Annals of internal medicine. 2000;133(9):696-700.
  28. Tollin SR, Mery GM, Jelveh N, Fallon EF, Mikhail M, Blumenfeld W, Perlmutter S. The use of fine-needle aspiration biopsy under ultrasound guidance to assess the risk of malignancy in patients with a multinodular goiter. Thyroid : official journal of the American Thyroid Association. 2000;10(3):235-241.
  29. Knudsen N, Bols B, Bulow I, Jorgensen T, Perrild H, Ovesen L, Laurberg P. Validation of ultrasonography of the thyroid gland for epidemiological purposes. Thyroid : official journal of the American Thyroid Association. 1999;9(11):1069-1074.
  30. Giovagnorio F, Drudi FM, Fanelli G, Flecca D, Francioso A. Fatty changes as a misleading factor in the evaluation with ultrasound of superficial lymph nodes. Ultrasound in medicine & biology. 2005;31(8):1017-1022.
  31. Bauer M, Blumentritt H, Finke R, Schlattmann P, Adli M, Baethge C, Bschor T, Muller-Oerlinghausen B, Berghofer A. Using ultrasonography to determine thyroid size and prevalence of goiter in lithium-treated patients with affective disorders. Journal of affective disorders. 2007;104(1-3):45-51.
  32. Pedersen OM, Aardal NP, Larssen TB, Varhaug JE, Myking O, Vik-Mo H. The value of ultrasonography in predicting autoimmune thyroid disease. Thyroid : official journal of the American Thyroid Association. 2000;10(3):251-259.
  33. Blum M, Passalaqua AM, Sackler JP, Pudlowski R. Thyroid echography of subacute thyroiditis. Radiology. 1977;125(3):795-798.
  34. Hiromatsu Y, Ishibashi M, Miyake I, Soyejima E, Yamashita K, Koike N, Nonaka K. Color Doppler ultrasonography in patients with subacute thyroiditis. Thyroid : official journal of the American Thyroid Association. 1999;9(12):1189-1193.
  35. Kunz A, Blank W, Braun B. De Quervain's subacute thyroiditis -- colour Doppler sonography findings. Ultraschall in der Medizin. 2005;26(2):102-106.
  36. Lu CP, Chang TC, Wang CY, Hsiao YL. Serial changes in ultrasound-guided fine needle aspiration cytology in subacute thyroiditis. Acta cytologica. 1997;41(2):238-243.
  37. Park JS, Oh KK, Kim EK, Chang HS, Hong SW. Sonographic screening for thyroid cancer in females undergoing breast sonography. AJR American journal of roentgenology. 2006;186(4):1025-1028.
  38. Espinasse P. Thyroid echography in chronic autoimmune lymphocytic thyroiditis. Journal de radiologie. 1983;64(10):537-544.
  39. Gutekunst R, Hafermann W, Mansky T, Scriba PC. Ultrasonography related to clinical and laboratory findings in lymphocytic thyroiditis. Acta endocrinologica. 1989;121(1):129-135.
  40. Hayashi N, Tamaki N, Konishi J, Yonekura Y, Senda M, Kasagi K, Yamamoto K, Iida Y, Misaki T, Endo K, et al. Sonography of Hashimoto's thyroiditis. Journal of clinical ultrasound : JCU. 1986;14(2):123-126.
  41. Jayaram G, Marwaha RK, Gupta RK, Sharma SK. Cytomorphologic aspects of thyroiditis. A study of 51 cases with functional, immunologic and ultrasonographic data. Acta cytologica. 1987;31(6):687-693.
  42. Marcocci C, Vitti P, Cetani F, Catalano F, Concetti R, Pinchera A. Thyroid ultrasonography helps to identify patients with diffuse lymphocytic thyroiditis who are prone to develop hypothyroidism. The Journal of clinical endocrinology and metabolism. 1991;72(1):209-213.
  43. Rago T, Chiovato L, Grasso L, Pinchera A, Vitti P. Thyroid ultrasonography as a tool for detecting thyroid autoimmune diseases and predicting thyroid dsfunction in apparently healthy subjects. Journal of endocrinological investigation. 2001;24(10):763-769.
  44. Raber W, Gessl A, Nowotny P, Vierhapper H. Thyroid ultrasound versus antithyroid peroxidase antibody determination: a cohort study of four hundred fifty-one subjects. Thyroid : official journal of the American Thyroid Association. 2002;12(8):725-731.
  45. Erdogan MF, Anil C, Cesur M, Baskal N, Erdogan G. Color flow Doppler sonography for the etiologic diagnosis of hyperthyroidism. Thyroid : official journal of the American Thyroid Association. 2007;17(3):223-228.
  46. Dirikoc A, Polat SB, Kandemir Z, Aydin C, Ozdemir D, Dellal FD, Ersoy R, Cakir B. Comparison of ultrasonography features and malignancy rate of toxic and nontoxic autonomous nodules: a preliminary study. Annals of nuclear medicine. 2015;29(10):883-889.
  47. Premawardhana LD, Parkes AB, Ammari F, John R, Darke C, Adams H, Lazarus JH. Postpartum thyroiditis and long-term thyroid status: prognostic influence of thyroid peroxidase antibodies and ultrasound echogenicity. The Journal of clinical endocrinology and metabolism. 2000;85(1):71-75.
  48. Shahbazian HB, Sarvghadi F, Azizi F. Ultrasonographic characteristics and follow-up in post-partum thyroiditis. Journal of endocrinological investigation. 2005;28(5):410-412.
  49. Vlachopapadopoulou E, Thomas D, Karachaliou F, Chatzimarkou F, Memalai L, Vakaki M, Kaldrymides P, Michalacos S. Evolution of sonographic appearance of the thyroid gland in children with Hashimoto's thyroiditis. Journal of pediatric endocrinology & metabolism : JPEM. 2009;22(4):339-344.
  50. Hodgson KJ, Lazarus JH, Wheeler MH, Woodcock JP, Owen GM, McGregor AM, Hall R. Duplex scan-derived thyroid blood flow in euthyroid and hyperthyroid patients. World journal of surgery. 1988;12(4):470-475.
  51. Ralls PW, Mayekawa DS, Lee KP, Colletti PM, Radin DR, Boswell WD, Halls JM. Color-flow Doppler sonography in Graves disease: "thyroid inferno". AJR American journal of roentgenology. 1988;150(4):781-784.
  52. Bogazzi F, Bartalena L, Brogioni S, Mazzeo S, Vitti P, Burelli A, Bartolozzi C, Martino E. Color flow Doppler sonography rapidly differentiates type I and type II amiodarone-induced thyrotoxicosis. Thyroid : official journal of the American Thyroid Association. 1997;7(4):541-545.
  53. Eaton SE, Euinton HA, Newman CM, Weetman AP, Bennet WM. Clinical experience of amiodarone-induced thyrotoxicosis over a 3-year period: role of colour-flow Doppler sonography. Clinical endocrinology. 2002;56(1):33-38.
  54. Loy M, Perra E, Melis A, Cianchetti ME, Piga M, Serra A, Pinna G, Mariotti S. Color-flow Doppler sonography in the differential diagnosis and management of amiodarone-induced thyrotoxicosis. Acta radiologica. 2007;48(6):628-634.
  55. Macedo TA, Chammas MC, Jorge PT, Souza LP, Farage L, Watanabe T, Santos VA, Cerri GG. Differentiation between the two types of amiodarone-associated thyrotoxicosis using duplex and amplitude Doppler sonography. Acta radiologica. 2007;48(4):412-421.
  56. Wong R, Cheung W, Stockigt JR, Topliss DJ. Heterogeneity of amiodarone-induced thyrotoxicosis: evaluation of colour-flow Doppler sonography in predicting therapeutic response. Internal medicine journal. 2003;33(9-10):420-426.
  57. Kim TK, Lee EJ. The value of the mean peak systolic velocity of the superior thyroidal artery in the differential diagnosis of thyrotoxicosis. Ultrasonography. 2015;34(4):292-296.
  58. Su DH, Liao KM, Hsiao YL, Chang TC. Determining when to operate on patients with Hashimoto's thyroiditis with nodular lesions: the role of ultrasound-guided fine needle aspiration cytology. Acta cytologica. 2004;48(5):622-629.
  59. Saleh A, Furst G, Feldkamp J, Godehardt E, Grust A, Modder U. Estimation of antithyroid drug dose in Graves' disease: value of quantification of thyroid blood flow with color duplex sonography. Ultrasound in medicine & biology. 2001;27(8):1137-1141.
  60. Wang CY, Chang TC. Thyroid Doppler ultrasonography and resistive index in the evaluation of the need for ablative or antithyroid drug therapy in Graves' hyperthyroidism. Journal of the Formosan Medical Association = Taiwan yi zhi. 2001;100(11):753-757.
  61. Varsamidis K, Varsamidou E, Mavropoulos G. Doppler ultrasonography in predicting relapse of hyperthyroidism in Graves' disease. Acta radiologica. 2000;41(1):45-48.
  62. Saleh A, Cohnen M, Furst G, Modder U, Feldkamp J. Prediction of relapse after antithyroid drug therapy of Graves' disease: value of color Doppler sonography. Experimental and clinical endocrinology & diabetes : official journal, German Society of Endocrinology [and] German Diabetes Association. 2004;112(9):510-513.
  63. Zingrillo M, D'Aloiso L, Ghiggi MR, Di Cerbo A, Chiodini I, Torlontano M, Liuzzi A. Thyroid hypoechogenicity after methimazole withdrawal in Graves' disease: a useful index for predicting recurrence? Clinical endocrinology. 1996;45(2):201-206.
  64. Nagasaki T, Inaba M, Kumeda Y, Fujiwara-Ueda M, Hiura Y, Nishizawa Y. Significance of thyroid blood flow as a predictor of methimazole sensitivity in untreated hyperthyroid patients with Graves' disease. Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie. 2007;61(8):472-476.
  65. Markovic V, Eterovic D. Thyroid echogenicity predicts outcome of radioiodine therapy in patients with Graves' disease. The Journal of clinical endocrinology and metabolism. 2007;92(9):3547-3552.
  66. Ansaldo GL, Pretolesi F, Varaldo E, Meola C, Minuto M, Borgonovo G, Derchi LE, Torre GC. Doppler evaluation of intrathyroid arterial resistances during preoperative treatment with Lugol's iodide solution in patients with diffuse toxic goiter. Journal of the American College of Surgeons. 2000;191(6):607-612.
  67. Erbil Y, Ozluk Y, Giris M, Salmaslioglu A, Issever H, Barbaros U, Kapran Y, Ozarmagan S, Tezelman S. Effect of lugol solution on thyroid gland blood flow and microvessel density in the patients with Graves' disease. The Journal of clinical endocrinology and metabolism. 2007;92(6):2182-2189.
  68. Yabuta T, Ito Y, Hirokawa M, Fukushima M, Inoue H, Tomoda C, Higashiyama T, Kihara M, Uruno T, Takamura Y, Kobayashi K, Miya A, Matsuzuka F, Miyauchi A. Preoperative administration of excess iodide increases thyroid volume of patients with Graves' disease. Endocrine journal. 2009;56(3):371-375.
  69. Cohen O, Pinhas-Hamiel O, Sivan E, Dolitski M, Lipitz S, Achiron R. Serial in utero ultrasonographic measurements of the fetal thyroid: a new complementary tool in the management of maternal hyperthyroidism in pregnancy. Prenatal diagnosis. 2003;23(9):740-742.
  70. Ota H, Ito Y, Matsuzuka F, Kuma S, Fukata S, Morita S, Kobayashi K, Nakamura Y, Kakudo K, Amino N, Miyauchi A. Usefulness of ultrasonography for diagnosis of malignant lymphoma of the thyroid. Thyroid : official journal of the American Thyroid Association. 2006;16(10):983-987.
  71. Simeone JF, Daniels GH, Mueller PR, Maloof F, vanSonnenberg E, Hall DA, O'Connell RS, Ferrucci JT, Jr., Wittenberg J. High-resolution real-time sonography of the thyroid. Radiology. 1982;145(2):431-435.
  72. Maizlin ZV, Wiseman SM, Vora P, Kirby JM, Mason AC, Filipenko D, Brown JA. Hurthle cell neoplasms of the thyroid: sonographic appearance and histologic characteristics. Journal of ultrasound in medicine : official journal of the American Institute of Ultrasound in Medicine. 2008;27(5):751-757; quiz 759.
  73. Cappelli C, Castellano M, Pirola I, Cumetti D, Agosti B, Gandossi E, Agabiti Rosei E. The predictive value of ultrasound findings in the management of thyroid nodules. QJM : monthly journal of the Association of Physicians. 2007;100(1):29-35.
  74. Moon WJ, Jung SL, Lee JH, Na DG, Baek JH, Lee YH, Kim J, Kim HS, Byun JS, Lee DH, Thyroid Study Group KSoN, Head, Neck R. Benign and malignant thyroid nodules: US differentiation--multicenter retrospective study. Radiology. 2008;247(3):762-770.
  75. Remonti LR, Kramer CK, Leitao CB, Pinto LC, Gross JL. Thyroid ultrasound features and risk of carcinoma: a systematic review and meta-analysis of observational studies. Thyroid : official journal of the American Thyroid Association. 2015;25(5):538-550.
  76. Scheible W, Leopold GR, Woo VL, Gosink BB. High-resolution real-time ultrasonography of thyroid nodules. Radiology. 1979;133(2):413-417.
  77. Solbiati L, Cioffi V, Ballarati E. Ultrasonography of the neck. Radiologic clinics of North America. 1992;30(5):941-954.
  78. Solbiati L, Volterrani L, Rizzatto G, Bazzocchi M, Busilacci P, Candiani F, Ferrari F, Giuseppetti G, Maresca G, Mirk P, et al. The thyroid gland with low uptake lesions: evaluation by ultrasound. Radiology. 1985;155(1):187-191.
  79. Yoon DY, Lee JW, Chang SK, Choi CS, Yun EJ, Seo YL, Kim KH, Hwang HS. Peripheral calcification in thyroid nodules: ultrasonographic features and prediction of malignancy. Journal of ultrasound in medicine : official journal of the American Institute of Ultrasound in Medicine. 2007;26(10):1349-1355.
  80. Solivetti FM, Bacaro D, Cecconi P, Baldelli R, Marandino F. Small hyperechogenic nodules in thyroiditis: usefulness of cytological characterization. Journal of experimental & clinical cancer research : CR. 2004;23(3):433-435.
  81. Park M, Shin JH, Han BK, Ko EY, Hwang HS, Kang SS, Kim JH, Oh YL. Sonography of thyroid nodules with peripheral calcifications. Journal of clinical ultrasound : JCU. 2009;37(6):324-328.
  82. Kakkos SK, Scopa CD, Chalmoukis AK, Karachalios DA, Spiliotis JD, Harkoftakis JG, Karavias DD, Androulakis JA, Vagenakis AG. Relative risk of cancer in sonographically detected thyroid nodules with calcifications. Journal of clinical ultrasound : JCU. 2000;28(7):347-352.
  83. Gorman B, Charboneau JW, James EM, Reading CC, Wold LE, Grant CS, Gharib H, Hay ID. Medullary thyroid carcinoma: role of high-resolution US. Radiology. 1987;162(1 Pt 1):147-150.
  84. Propper RA, Skolnick ML, Weinstein BJ, Dekker A. The nonspecificity of the thyroid halo sign. Journal of clinical ultrasound : JCU. 1980;8(2):129-132.
  85. Ito Y, Amino N, Yokozawa T, Ota H, Ohshita M, Murata N, Morita S, Kobayashi K, Miyauchi A. Ultrasonographic evaluation of thyroid nodules in 900 patients: comparison among ultrasonographic, cytological, and histological findings. Thyroid : official journal of the American Thyroid Association. 2007;17(12):1269-1276.
  86. Siebert SM, Gomez AJ, Liang T, Tahvildari AM, Desser TS, Jeffrey RB, Kamaya A. Diagnostic Performance of Margin Features in Thyroid Nodules in Prediction of Malignancy. AJR American journal of roentgenology. 2018;210(4):860-865.
  87. Levine RA. Value of Doppler ultrasonography in management of patients with follicular thyroid biopsy specimens. Endocrine practice : official journal of the American College of Endocrinology and the American Association of Clinical Endocrinologists. 2006;12(3):270-274.
  88. Appetecchia M, Solivetti FM. The association of colour flow Doppler sonography and conventional ultrasonography improves the diagnosis of thyroid carcinoma. Hormone research. 2006;66(5):249-256.
  89. Bakhshaee M, Davoudi Y, Mehrabi M, Layegh P, Mirsadaee S, Rad MP, Leyegh P. Vascular pattern and spectral parameters of power Doppler ultrasound as predictors of malignancy risk in thyroid nodules. The Laryngoscope. 2008;118(12):2182-2186.
  90. Spiezia S, Farina R, Cerbone G, Assanti AP, Iovino V, Siciliani M, Lombardi G, Colao A. Analysis of color Doppler signal intensity variation after levovist injection: a new approach to the diagnosis of thyroid nodules. Journal of ultrasound in medicine : official journal of the American Institute of Ultrasound in Medicine. 2001;20(3):223-231.
  91. Huang K, Gao N, Zhai Q, Bian D, Wang D, Wang X. The anteroposterior diameter of nodules in the risk assessment of papillary thyroid microcarcinoma. Medicine. 2018;97(10):e9712.
  92. Lee MJ, Kim EK, Kwak JY, Kim MJ. Partially cystic thyroid nodules on ultrasound: probability of malignancy and sonographic differentiation. Thyroid : official journal of the American Thyroid Association. 2009;19(4):341-346.
  93. Kim JY, Jung SL, Kim MK, Kim TJ, Byun JY. Differentiation of benign and malignant thyroid nodules based on the proportion of sponge-like areas on ultrasonography: imaging-pathologic correlation. Ultrasonography. 2015;34(4):304-311.
  94. Cavaliere A, Colella R, Puxeddu E, Gambelunghe G, Falorni A, Stracci F, d'Ajello M, Avenia N, De Feo P. A useful ultrasound score to select thyroid nodules requiring fine needle aspiration in an iodine-deficient area. Journal of endocrinological investigation. 2009;32(5):440-444.
  95. Kwak JY, Kim EK, Kim MJ, Hong SW, Choi SH, Son EJ, Oh KK, Park CS, Chung WY, Kim KW. The role of ultrasound in thyroid nodules with a cytology reading of "suspicious for papillary thyroid carcinoma". Thyroid : official journal of the American Thyroid Association. 2008;18(5):517-522.
  96. Liu YI, Kamaya A, Desser TS, Rubin DL. A Bayesian classifier for differentiating benign versus malignant thyroid nodules using sonographic features. AMIAAnnual Symposium proceedings AMIA Symposium. 2008:419-423.
  97. Shimamoto K, Satake H, Sawaki A, Ishigaki T, Funahashi H, Imai T. Preoperative staging of thyroid papillary carcinoma with ultrasonography. European journal of radiology. 1998;29(1):4-10.
  98. Shin JH, Han BK, Ko EY, Kang SS. Sonographic findings in the surgical bed after thyroidectomy: comparison of recurrent tumors and nonrecurrent lesions. Journal of ultrasound in medicine : official journal of the American Institute of Ultrasound in Medicine. 2007;26(10):1359-1366.
  99. Lim KJ, Choi CS, Yoon DY, Chang SK, Kim KK, Han H, Kim SS, Lee J, Jeon YH. Computer-aided diagnosis for the differentiation of malignant from benign thyroid nodules on ultrasonography. Academic radiology. 2008;15(7):853-858.
  100. Choi SH, Kim EK, Kwak JY, Kim MJ, Son EJ. Interobserver and intraobserver variations in ultrasound assessment of thyroid nodules. Thyroid : official journal of the American Thyroid Association. 2010;20(2):167-172.
  101. Rhee SJ, Hahn SY, Ko ES, Ryu JW, Ko EY, Shin JH. Follicular variant of papillary thyroid carcinoma: distinct biologic behavior based on ultrasonographic features. Thyroid : official journal of the American Thyroid Association. 2014;24(4):683-688.
  102. Al Nofal A, Gionfriddo MR, Javed A, Haydour Q, Brito JP, Prokop LJ, Pittock ST, Murad MH. Accuracy of thyroid nodule sonography for the detection of thyroid cancer in children: systematic review and meta-analysis. Clinical endocrinology. 2016;84(3):423-430.
  103. Buryk MA, Simons JP, Picarsic J, Monaco SE, Ozolek JA, Joyce J, Gurtunca N, Nikiforov YE, Feldman Witchel S. Can malignant thyroid nodules be distinguished from benign thyroid nodules in children and adolescents by clinical characteristics? A review of 89 pediatric patients with thyroid nodules. Thyroid : official journal of the American Thyroid Association. 2015;25(4):392-400.
  104. Khalid AN, Hollenbeak CS, Quraishi SA, Fan CY, Stack BC, Jr. The cost-effectiveness of iodine 131 scintigraphy, ultrasonography, and fine-needle aspiration biopsy in the initial diagnosis of solitary thyroid nodules. Archives of otolaryngology--head & neck surgery. 2006;132(3):244-250.
  105. Tan GH, Gharib H. Thyroid incidentalomas: management approaches to nonpalpable nodules discovered incidentally on thyroid imaging. Annals of internal medicine. 1997;126(3):226-231.
  106. 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. Annals of internal medicine. 1968;69(3):537-540.
  107. Mazzaferri EL. Management of a solitary thyroid nodule. The New England journal of medicine. 1993;328(8):553-559.
  108. Ridgway EC. Clinical review 30: Clinician's evaluation of a solitary thyroid nodule. The Journal of clinical endocrinology and metabolism. 1992;74(2):231-235.
  109. Berker D, Aydin Y, Ustun I, Gul K, Tutuncu Y, Isik S, Delibasi T, Guler S. The value of fine-needle aspiration biopsy in subcentimeter thyroid nodules. Thyroid : official journal of the American Thyroid Association. 2008;18(6):603-608.
  110. Hagag P, Strauss S, Weiss M. Role of ultrasound-guided fine-needle aspiration biopsy in evaluation of nonpalpable thyroid nodules. Thyroid : official journal of the American Thyroid Association. 1998;8(11):989-995.
  111. Papini E, Guglielmi R, Bianchini A, Crescenzi A, Taccogna S, Nardi F, Panunzi C, Rinaldi R, Toscano V, Pacella CM. Risk of malignancy in nonpalpable thyroid nodules: predictive value of ultrasound and color-Doppler features. The Journal of clinical endocrinology and metabolism. 2002;87(5):1941-1946.
  112. Liebeskind A, Sikora AG, Komisar A, Slavit D, Fried K. Rates of malignancy in incidentally discovered thyroid nodules evaluated with sonography and fine-needle aspiration. Journal of ultrasound in medicine : official journal of the American Institute of Ultrasound in Medicine. 2005;24(5):629-634.
  113. Nam-Goong IS, Kim HY, Gong G, Lee HK, Hong SJ, Kim WB, Shong YK. Ultrasonography-guided fine-needle aspiration of thyroid incidentaloma: correlation with pathological findings. Clinical endocrinology. 2004;60(1):21-28.
  114. Kim JY, Lee CH, Kim SY, Jeon WK, Kang JH, An SK, Jun WS. Radiologic and pathologic findings of nonpalpable thyroid carcinomas detected by ultrasonography in a medical screening center. Journal of ultrasound in medicine : official journal of the American Institute of Ultrasound in Medicine. 2008;27(2):215-223.
  115. Vila JJ, Jimenez FJ, Irisarri R, Vicuna M, Ruiz-Clavijo D, Gonzalez de la Higuera B, Fernandez-Urien I, Borda F. Prospective observational study of the incidental findings on endoscopic ultrasonography: should a complete exploration always be performed? Scandinavian journal of gastroenterology. 2009;44(9):1139-1145.
  116. Lewis BD, Hay ID, Charboneau JW, McIver B, Reading CC, Goellner JR. Percutaneous ethanol injection for treatment of cervical lymph node metastases in patients with papillary thyroid carcinoma. AJR American journal of roentgenology. 2002;178(3):699-704.
  117. Steele SR, Martin MJ, Mullenix PS, Azarow KS, Andersen CA. The significance of incidental thyroid abnormalities identified during carotid duplex ultrasonography. Archives of surgery. 2005;140(10):981-985.
  118. Carpi A, Nicolini A, Casara D, Rubello D, Rosa Pelizzo M. Nonpalpable thyroid carcinoma: clinical controversies on preoperative selection. American journal of clinical oncology. 2003;26(3):232-235.
  119. Mazzaferri EL, Sipos J. Should all patients with subcentimeter thyroid nodules undergo fine-needle aspiration biopsy and preoperative neck ultrasonography to define the extent of tumor invasion? Thyroid : official journal of the American Thyroid Association. 2008;18(6):597-602.
  120. Kim DW, Park AW, Lee EJ, Choo HJ, Kim SH, Lee SH, Eom JW. Ultrasound-guided fine-needle aspiration biopsy of thyroid nodules smaller than 5 mm in the maximum diameter: assessment of efficacy and pathological findings. Korean journal of radiology. 2009;10(5):435-440.
  121. Brito JP, Singh-Ospina N, Gionfriddo MR, Maraka S, Espinosa De Ycaza A, Rodriguez-Gutierrez R, Morris JC, Montori VM, Tuttle RM. Restricting ultrasound thyroid fine needle aspiration biopsy by nodule size: which tumors are we missing? A population-based study. Endocrine. 2016;51(3):499-505.
  122. Kwak JY, Kim EK, Moon HJ, Kim MJ, Ahn SS, Son EJ, Sohn YM. Parathyroid incidentalomas detected on routine ultrasound-directed fine-needle aspiration biopsy in patients referred for thyroid nodules and the role of parathyroid hormone analysis in the samples. Thyroid : official journal of the American Thyroid Association. 2009;19(7):743-748.
  123. Sung JY. Parathyroid ultrasonography: the evolving role of the radiologist. Ultrasonography. 2015;34(4):268-274.
  124. Sierra M, Sebag F, De Micco C, Loudot C, Misso C, Calzolari F, Henry JF. Abrikossoff tumor of the proximal esophagus misdiagnosed as a thyroid nodule. Annales de chirurgie. 2006;131(3):219-221.
  125. Solbiati L, Rizzatto G, Bellotti E, Montali G, Cioffi V, Croce F. High-resolution sonography of cervical lymph nodes in head and neck cancer: criteria for differentiation of reactive versus malignant nodes. Radiology. 1988;169:113.
  126. Choi EC, Moon WJ, Lim YC. Case report. Tuberculous cervical lymphadenitis mimicking metastatic lymph nodes from papillary thyroid carcinoma. The British journal of radiology. 2009;82(982):e208-211.
  127. Iqbal M, Subhan A, Aslam A. Papillary thyroid carcinoma with tuberculous cervical lymphadenopathy mimicking metastasis. Journal of the College of Physicians and Surgeons--Pakistan : JCPSP. 2011;21(4):207-209.
  128. Leboulleux S, Girard E, Rose M, Travagli JP, Sabbah N, Caillou B, Hartl DM, Lassau N, Baudin E, Schlumberger M. Ultrasound criteria of malignancy for cervical lymph nodes in patients followed up for differentiated thyroid cancer. The Journal of clinical endocrinology and metabolism. 2007;92(9):3590-3594.
  129. Kuna SK, Bracic I, Tesic V, Kuna K, Herceg GH, Dodig D. Ultrasonographic differentiation of benign from malignant neck lymphadenopathy in thyroid cancer. Journal of ultrasound in medicine : official journal of the American Institute of Ultrasound in Medicine. 2006;25(12):1531-1537; quiz 1538-1540.
  130. Rosario PW, de Faria S, Bicalho L, Alves MF, Borges MA, Purisch S, Padrao EL, Rezende LL, Barroso AL. Ultrasonographic differentiation between metastatic and benign lymph nodes in patients with papillary thyroid carcinoma. Journal of ultrasound in medicine : official journal of the American Institute of Ultrasound in Medicine. 2005;24(10):1385-1389.
  131. Vassallo P, Wernecke K, Roos N, Peters PE. Differentiation of benign from malignant superficial lymphadenopathy: the role of high-resolution US. Radiology. 1992;183(1):215-220.
  132. Alzahrani AS, Alsuhaibani H, Salam SA, Al Sifri SN, Mohamed G, Al Sobhi S, Sulaiman O, Akhtar M. Diagnostic accuracy of high-resolution neck ultrasonography in the follow-up of differentiated thyroid cancer: a prospective study. Endocrine practice : official journal of the American College of Endocrinology and the American Association of Clinical Endocrinologists. 2005;11(3):165-171.
  133. Chang DB, Yuan A, Yu CJ, Luh KT, Kuo SH, Yang PC. Differentiation of benign and malignant cervical lymph nodes with color Doppler sonography. AJR American journal of roentgenology. 1994;162(4):965-968.
  134. Choi MY, Lee JW, Jang KJ. Distinction between benign and malignant causes of cervical, axillary, and inguinal lymphadenopathy: value of Doppler spectral waveform analysis. AJR American journal of roentgenology. 1995;165(4):981-984.
  135. Ahuja AT, Ying M, Yuen HY, Metreweli C. Power Doppler sonography of metastatic nodes from papillary carcinoma of the thyroid. Clinical radiology. 2001;56(4):284-288.
  136. Kessler A, Rappaport Y, Blank A, Marmor S, Weiss J, Graif M. Cystic appearance of cervical lymph nodes is characteristic of metastatic papillary thyroid carcinoma. Journal of clinical ultrasound : JCU. 2003;31(1):21-25.
  137. Seven H, Gurkan A, Cinar U, Vural C, Turgut S. Incidence of occult thyroid carcinoma metastases in lateral cervical cysts. American journal of otolaryngology. 2004;25(1):11-17.
  138. Wunderbaldinger P, Harisinghani MG, Hahn PF, Daniels GH, Turetschek K, Simeone J, O'Neill MJ, Mueller PR. Cystic lymph node metastases in papillary thyroid carcinoma. AJR American journal of roentgenology. 2002;178(3):693-697.
  139. Kim E, Park JS, Son KR, Kim JH, Jeon SJ, Na DG. Preoperative diagnosis of cervical metastatic lymph nodes in papillary thyroid carcinoma: comparison of ultrasound, computed tomography, and combined ultrasound with computed tomography. Thyroid : official journal of the American Thyroid Association. 2008;18(4):411-418.
  140. Choi JS, Kim J, Kwak JY, Kim MJ, Chang HS, Kim EK. Preoperative staging of papillary thyroid carcinoma: comparison of ultrasound imaging and CT. AJR American journal of roentgenology. 2009;193(3):871-878.
  141. Blum M, Tiu S, Chu M, Goel S, Friedman K. I-131 SPECT/CT elucidates cryptic findings on planar whole-body scans and can reduce needless therapy with I-131 in post-thyroidectomy thyroid cancer patients. Thyroid : official journal of the American Thyroid Association. 2011;21(11):1235-1247.
  142. Garger YB, Winfeld M, Friedman K, Blum M. In Thyroidectomized Thyroid Cancer Patients, False-Positive I-131 Whole Body Scans Are Often Caused by Inflammation Rather Than Thyroid Cancer. Journal of investigative medicine high impact case reports. 2016;4(1):2324709616633715.
  143. Biermann M, Krakenes J, Brauckhoff K, Haugland HK, Heinecke A, Akslen LA, Varhaug JE, Brauckhoff M. Post-PET ultrasound improves specificity of 18F-FDG-PET for recurrent differentiated thyroid cancer while maintaining sensitivity. Acta radiologica. 2015;56(11):1350-1360.
  144. Pisani T, Vecchione A, Sinopoli NT, Drusco A, Valli C, Giovagnoli MR. Cytological and immunocytochemical analysis of laterocervical lymph nodes in patients with previous thyroid carcinoma. Anticancer research. 1999;19(4C):3527-3530.
  145. Horvath E, Majlis S, Rossi R, Franco C, Niedmann JP, Castro A, Dominguez M. An ultrasonogram reporting system for thyroid nodules stratifying cancer risk for clinical management. The Journal of clinical endocrinology and metabolism. 2009;94(5):1748-1751.
  146. Griffin AS, Mitsky J, Rawal U, Bronner AJ, Tessler FN, Hoang JK. Improved Quality of Thyroid Ultrasound Reports After Implementation of the ACR Thyroid Imaging Reporting and Data System Nodule Lexicon and Risk Stratification System. Journal of the American College of Radiology : JACR. 2018;15(5):743-748.
  147. Hoang JK, Middleton WD, Farjat AE, Langer JE, Reading CC, Teefey SA, Abinanti N, Boschini FJ, Bronner AJ, Dahiya N, Hertzberg BS, Newman JR, Scanga D, Vogler RC, Tessler FN. Reduction in Thyroid Nodule Biopsies and Improved Accuracy with American College of Radiology Thyroid Imaging Reporting and Data System. Radiology. 2018;287(1):185-193.
  148. Grani G, Lamartina L, Ascoli V, Bosco D, Biffoni M, Giacomelli L, Maranghi M, Falcone R, Ramundo V, Cantisani V, Filetti S, Durante C. Reducing the Number of Unnecessary Thyroid Biopsies While Improving Diagnostic Accuracy: Toward the "Right" TIRADS. The Journal of clinical endocrinology and metabolism. 2019;104(1):95-102.
  149. Savelonas M, Maroulis D, Sangriotis M. A computer-aided system for malignancy risk assessment of nodules in thyroid US images based on boundary features. Computer methods and programs in biomedicine. 2009;96(1):25-32.
  150. Suzuki S, Bogdanova TI, Saenko VA, Hashimoto Y, Ito M, Iwadate M, Rogounovitch T, Tronko MD, Yamashita S. Histopathological analysis of papillary thyroid carcinoma detected during ultrasound screening examinations in Fukushima. Cancer science. 2018.
  151. Gordon DL, Flisak M, Fisher SG. Changes in thyroid nodule volume caused by fine-needle aspiration: a factor complicating the interpretation of the effect of thyrotropin suppression on nodule size. The Journal of clinical endocrinology and metabolism. 1999;84(12):4566-4569.
  152. Peiling Yang S, Bach AM, Tuttle RM, Fish SA. Frequent screening with serial neck ultrasound is more likely to identify false-positive abnormalities than clinically significant disease in the surveillance of intermediate risk papillary thyroid cancer patients without suspicious findings on follow-up ultrasound evaluation. The Journal of clinical endocrinology and metabolism. 2015;100(4):1561-1567.
  153. Simeone JF, Daniels GH, Hall DA, McCarthy K, Kopans DB, Butch RJ, Mueller PR, Stark DD, Ferrucci JT, Jr., Wang CA. Sonography in the follow-up of 100 patients with thyroid carcinoma. AJR American journal of roentgenology. 1987;148(1):45-49.
  154. Arora P, Blum M. Utility of ultrasonography in post surgical management of patients with thyroid carcinoma. Paper presented at: American Thyroid Association: Seventy-fourth Meeting2001; Washington DC
  155. Torlontano M, Crocetti U, Augello G, D'Aloiso L, Bonfitto N, Varraso A, Dicembrino F, Modoni S, Frusciante V, Di Giorgio A, Bruno R, Filetti S, Trischitta V. Comparative evaluation of recombinant human thyrotropin-stimulated thyroglobulin levels, 131I whole-body scintigraphy, and neck ultrasonography in the follow-up of patients with papillary thyroid microcarcinoma who have not undergone radioiodine therapy. The Journal of clinical endocrinology and metabolism. 2006;91(1):60-63.
  156. D'Andrea V, Cantisani V, Catania A, Di Matteo FM, Sorrenti S, Greco R, Kyriacou KA, Menichini G, Marotta E, De Stefano M, Palermo S, Di Marco C, De Antoni E. Thyroid tissue remnants after "total thyroidectomy". Il Giornale di chirurgia. 2009;30(8-9):339-344.
  157. Antonelli A, Miccoli P, Fallahi P, Ferrari SM, Grosso M, Boni G, Berti P. Thyrotropin-stimulated serum thyroglobulin combined with neck ultrasonography has the highest sensitivity in monitoring differentiated thyroid carcinoma in children. Surgery. 2006;140(6):1035-1041; discussion 1041-1032.
  158. Torlontano M, Crocetti U, D'Aloiso L, Bonfitto N, Di Giorgio A, Modoni S, Valle G, Frusciante V, Bisceglia M, Filetti S, Schlumberger M, Trischitta V. Serum thyroglobulin and 131I whole body scan after recombinant human TSH stimulation in the follow-up of low-risk patients with differentiated thyroid cancer. European journal of endocrinology. 2003;148(1):19-24.
  159. Chung YE, Kim EK, Kim MJ, Yun M, Hong SW. Suture granuloma mimicking recurrent thyroid carcinoma on ultrasonography. Yonsei medical journal. 2006;47(5):748-751.
  160. Kim JH, Lee JH, Shong YK, Hong SJ, Ko MS, Lee DH, Choi CG, Kim SJ. Ultrasound features of suture granulomas in the thyroid bed after thyroidectomy for papillary thyroid carcinoma with an emphasis on their differentiation from locally recurrent thyroid carcinomas. Ultrasound in medicine & biology. 2009;35(9):1452-1457.
  161. Karwowski JK, Jeffrey RB, McDougall IR, Weigel RJ. Intraoperative ultrasonography improves identification of recurrent thyroid cancer. Surgery. 2002;132(6):924-928; discussion 928-929.
  162. Marshall CL, Lee JE, Xing Y, Perrier ND, Edeiken BS, Evans DB, Grubbs EG. Routine pre-operative ultrasonography for papillary thyroid cancer: effects on cervical recurrence. Surgery. 2009;146(6):1063-1072.
  163. Moreno MA, Agarwal G, de Luna R, Siegel ER, Sherman SI, Edeiken-Monroe BS, Clayman GL. Preoperative lateral neck ultrasonography as a long-term outcome predictor in papillary thyroid cancer. Archives of otolaryngology--head & neck surgery. 2011;137(2):157-162.
  164. Boland GW, Lee MJ, Mueller PR, Mayo-Smith W, Dawson SL, Simeone JF. Efficacy of sonographically guided biopsy of thyroid masses and cervical lymph nodes. AJR American journal of roentgenology. 1993;161(5):1053-1056.
  165. Gharib H, Goellner JR, Johnson DA. Fine-needle aspiration cytology of the thyroid. A 12-year experience with 11,000 biopsies. Clinics in laboratory medicine. 1993;13(3):699-709.
  166. Matalon TA, Silver B. US guidance of interventional procedures. Radiology. 1990;174(1):43-47.
  167. Rizzatto G, Solbiati L, Croce F, Derchi LE. Aspiration biopsy of superficial lesions: ultrasonic guidance with a linear-array probe. AJR American journal of roentgenology. 1987;148(3):623-625.
  168. Seiberling KA, Dutra JC, Gunn J. Ultrasound-guided fine needle aspiration biopsy of thyroid nodules performed in the office. The Laryngoscope. 2008;118(2):228-231.
  169. Takashima S, Sone S, Nomura N, Tomiyama N, Kobayashi T, Nakamura H. Nonpalpable lymph nodes of the neck: assessment with US and US-guided fine-needle aspiration biopsy. Journal of clinical ultrasound : JCU. 1997;25(6):283-292.
  170. Lyle MA, Dean DS. Ultrasound-guided fine-needle aspiration biopsy of thyroid nodules in patients taking novel oral anticoagulants. Thyroid : official journal of the American Thyroid Association. 2015;25(4):373-376.
  171. Kakiuchi Y, Idota N, Nakamura M, Ikegaya H. A Fatal Case of Cervical Hemorrhage After Fine Needle Aspiration and Core Needle Biopsy of the Thyroid Gland. The American journal of forensic medicine and pathology. 2015;36(3):207-209.
  172. Hor T, Lahiri SW. Bilateral thyroid hematomas after fine-needle aspiration causing acute airway obstruction. Thyroid : official journal of the American Thyroid Association. 2008;18(5):567-569.
  173. Leung AM, Farwell AP. Unsatisfactory consequences from fine-needle aspiration biopsy of thyroid nodules. Thyroid : official journal of the American Thyroid Association. 2008;18(5):491-492.
  174. Lee MJ, Ross DS, Mueller PR, Daniels GH, Dawson SL, Simeone JF. Fine-needle biopsy of cervical lymph nodes in patients with thyroid cancer: a prospective comparison of cytopathologic and tissue marker analysis. Radiology. 1993;187(3):851-854.
  175. Danese D, Sciacchitano S, Farsetti A, Andreoli M, Pontecorvi A. Diagnostic accuracy of conventional versus sonography-guided fine-needle aspiration biopsy of thyroid nodules. Thyroid : official journal of the American Thyroid Association. 1998;8(1):15-21.
  176. Khalid AN, Quraishi SA, Hollenbeak CS, Stack BC, Jr. Fine-needle aspiration biopsy versus ultrasound-guided fine-needle aspiration biopsy: cost-effectiveness as a frontline diagnostic modality for solitary thyroid nodules. Head & neck. 2008;30(8):1035-1039.
  177. Brenta G, Schnitman M, Bonnahon L, Besuschio S, Zuk C, De Barrio G, Peruzzotti C, Saubidet G. Evaluation of innovative skin-marking technique performed before thyroid ultrasound-guided fine-needle aspiration biopsies. Endocrine practice : official journal of the American College of Endocrinology and the American Association of Clinical Endocrinologists. 2002;8(1):5-9.
  178. Rausch P, Nowels K, Jeffrey RB, Jr. Ultrasonographically guided thyroid biopsy: a review with emphasis on technique. Journal of ultrasound in medicine : official journal of the American Institute of Ultrasound in Medicine. 2001;20(1):79-85.
  179. Zhang L, Liu Y, Tan X, Liu X, Zhang H, Qian L. Comparison of Different-Gauge Needles for Fine-Needle Aspiration Biopsy of Thyroid Nodules. Journal of ultrasound in medicine : official journal of the American Institute of Ultrasound in Medicine. 2018;37(7):1713-1716.
  180. Tublin ME, Martin JA, Rollin LJ, Pealer K, Kurs-Lasky M, Ohori NP. Ultrasound-guided fine-needle aspiration versus fine-needle capillary sampling biopsy of thyroid nodules: does technique matter? Journal of ultrasound in medicine : official journal of the American Institute of Ultrasound in Medicine. 2007;26(12):1697-1701.
  181. Schoedel KE, Tublin ME, Pealer K, Ohori NP. Ultrasound-guided biopsy of the thyroid: a comparison of technique with respect to diagnostic accuracy. Diagnostic cytopathology. 2008;36(11):787-789.
  182. Romitelli F, Di Stasio E, Santoro C, Iozzino M, Orsini A, Cesareo R. A comparative study of fine needle aspiration and fine needle non-aspiration biopsy on suspected thyroid nodules. Endocrine pathology. 2009;20(2):108-113.
  183. Ghofrani M, Beckman D, Rimm DL. The value of onsite adequacy assessment of thyroid fine-needle aspirations is a function of operator experience. Cancer. 2006;108(2):110-113.
  184. Borget I, Vielh P, Leboulleux S, Allyn M, Iacobelli S, Schlumberger M, de Pouvourville G. Assessment of the cost of fine-needle aspiration cytology as a diagnostic tool in patients with thyroid nodules. American journal of clinical pathology. 2008;129(5):763-771.
  185. Lieu D. Cytopathologist-performed ultrasound-guided fine-needle aspiration and core-needle biopsy: a prospective study of 500 consecutive cases. Diagnostic cytopathology. 2008;36(5):317-324.
  186. Ucler R, Kaya C, Cuhaci N, Tam AA, Usluogullari CA, Balkan F, Ersoy R, Cakir B. Thyroid Nodules with 2 Prior Inadequate Fine-Needle Aspiration Results: Effect of Increasing the Diameter of the Needle. Endocrine practice : official journal of the American College of Endocrinology and the American Association of Clinical Endocrinologists. 2015;21(6):595-603.
  187. Breslin M, Lawrance JA, Desai M, Ryder WD, Allan E. The role of ultrasound-guided fine-needle aspiration biopsy in the previously treated patient with thyroid cancer. Clinical otolaryngology and allied sciences. 2004;29(2):146-148.
  188. Saavedra J, Deladoey J, Saint-Vil D, Boivin Y, Alos N, Deal C, Van Vliet G, Huot C. Is ultrasonography useful in predicting thyroid cancer in children with thyroid nodules and apparently benign cytopathologic features? Hormone research in paediatrics. 2011;75(4):269-275.
  189. Krishnamurthy S, Bedi DG, Caraway NP. Ultrasound-guided fine-needle aspiration biopsy of the thyroid bed. Cancer. 2001;93(3):199-205.
  190. Leenhardt L, Hejblum G, Franc B, Fediaevsky LD, Delbot T, Le Guillouzic D, Menegaux F, Guillausseau C, Hoang C, Turpin G, Aurengo A. Indications and limits of ultrasound-guided cytology in the management of nonpalpable thyroid nodules. The Journal of clinical endocrinology and metabolism. 1999;84(1):24-28.
  191. Accurso A, Rocco N, Palumbo A, Leone F. Usefulness of ultrasound-guided fine-needle aspiration cytology in the diagnosis of non-palpable small thyroid nodules. Tumori. 2005;91(4):355-357.
  192. Deandrea M, Mormile A, Veglio M, Motta M, Pellerito R, Gallone G, Grassi A, Torchio B, Bradac R, Garberoglio R, Fonzo D. Fine-needle aspiration biopsy of the thyroid: comparison between thyroid palpation and ultrasonography. Endocrine practice : official journal of the American College of Endocrinology and the American Association of Clinical Endocrinologists. 2002;8(4):282-286.
  193. Gurleyik E, Coskun O, Aslaner A. Clinical importance of solitary solid nodule of the thyroid in endemic goiter region. Indian journal of medical sciences. 2005;59(9):388-395.
  194. Alexander EK, Heering JP, Benson CB, Frates MC, Doubilet PM, Cibas ES, Marqusee E. Assessment of nondiagnostic ultrasound-guided fine needle aspirations of thyroid nodules. The Journal of clinical endocrinology and metabolism. 2002;87(11):4924-4927.
  195. Cappelli C, Castellano M, Pirola I, Gandossi E, De Martino E, Cumetti D, Agosti B, Rosei EA. Thyroid nodule shape suggests malignancy. European journal of endocrinology. 2006;155(1):27-31.
  196. Popowicz B, Klencki M, Lewinski A, Slowinska-Klencka D. The usefulness of sonographic features in selection of thyroid nodules for biopsy in relation to the nodule's size. European journal of endocrinology. 2009;161(1):103-111.
  197. Barroeta JE, Wang H, Shiina N, Gupta PK, Livolsi VA, Baloch ZW. Is fine-needle aspiration (FNA) of multiple thyroid nodules justified? Endocrine pathology. 2006;17(1):61-65.
  198. Pacini F, Molinaro E, Castagna MG, Agate L, Elisei R, Ceccarelli C, Lippi F, Taddei D, Grasso L, Pinchera A. Recombinant human thyrotropin-stimulated serum thyroglobulin combined with neck ultrasonography has the highest sensitivity in monitoring differentiated thyroid carcinoma. The Journal of clinical endocrinology and metabolism. 2003;88(8):3668-3673.
  199. Epstein S, McEachern R, Khot R, Padia S, Patrie JT, Itri JN. Papillary Thyroid Carcinoma Recurrence: Low Yield of Neck Ultrasound With an Undetectable Serum Thyroglobulin Level. Journal of ultrasound in medicine : official journal of the American Institute of Ultrasound in Medicine. 2018;37(10):2325-2331.
  200. Wilhelm SM, Robinson AV, Krishnamurthi SS, Reynolds HL. Evaluation and management of incidental thyroid nodules in patients with another primary malignancy. Surgery. 2007;142(4):581-586; discussion 586-587.
  201. Bonavita JA, Mayo J, Babb J, Bennett G, Oweity T, Macari M, Yee J. Pattern recognition of benign nodules at ultrasound of the thyroid: which nodules can be left alone? AJR American journal of roentgenology. 2009;193(1):207-213.
  202. Moon WJ, Kwag HJ, Na DG. Are there any specific ultrasound findings of nodular hyperplasia ("leave me alone" lesion) to differentiate it from follicular adenoma? Acta radiologica. 2009;50(4):383-388.
  203. Reading CC, Charboneau JW, Hay ID, Sebo TJ. Sonography of thyroid nodules: a "classic pattern" diagnostic approach. Ultrasound quarterly. 2005;21(3):157-165.
  204. Moon HJ, Lee HS, Kim EK, Ko SY, Seo JY, Park WJ, Park HY, Kwak JY. Thyroid nodules </= 5 mm on ultrasonography: are they "leave me alone" lesions? Endocrine. 2015;49(3):735-744.
  205. Drozd VM, Lushchik ML, Polyanskaya ON, Fridman MV, Demidchik YE, Lyshchik AP, Biko J, Reiners C, Shibata Y, Saenko VA, Yamashita S. The usual ultrasonographic features of thyroid cancer are less frequent in small tumors that develop after a long latent period after the Chernobyl radiation release accident. Thyroid : official journal of the American Thyroid Association. 2009;19(7):725-734.
  206. Singh Ospina N, Sebo TJ, Morris JC, Castro MR. The Value of Repeat Thyroid Fine-Needle Aspiration Biopsy in Patients with a Previously Benign Result: How Often Does It Alter Management? Thyroid : official journal of the American Thyroid Association. 2015;25(10):1121-1126.
  207. Boi F, Baghino G, Atzeni F, Lai ML, Faa G, Mariotti S. The diagnostic value for differentiated thyroid carcinoma metastases of thyroglobulin (Tg) measurement in washout fluid from fine-needle aspiration biopsy of neck lymph nodes is maintained in the presence of circulating anti-Tg antibodies. The Journal of clinical endocrinology and metabolism. 2006;91(4):1364-1369.
  208. Pisani T, Vecchione A, Giovagnolii MR. Galectin-3 immunodetection may improve cytological diagnosis of occult papillary thyroid carcinoma. Anticancer research. 2004;24(2C):1111-1112.
  209. Chung SY, Oh KK, Chang HS. Sonographic findings of tuberculous thyroiditis in a patient with Behcet's syndrome. Journal of clinical ultrasound : JCU. 2002;30(3):184-188.
  210. Basaria S, Ayala AR, Westra WH, Cooper DS. Amyloidosis: role of fine-needle aspiration. Thyroid : official journal of the American Thyroid Association. 2003;13(3):313-314.
  211. Harvey JN, Parker D, De P, Shrimali RK, Otter M. Sonographically guided core biopsy in the assessment of thyroid nodules. Journal of clinical ultrasound : JCU. 2005;33(2):57-62.
  212. Strauss EB, Iovino A, Upender S. Simultaneous fine-needle aspiration and core biopsy of thyroid nodules and other superficial head and neck masses using sonographic guidance. AJR American journal of roentgenology. 2008;190(6):1697-1699.
  213. Lew JI, Rodgers SE, Solorzano CC. Developments in the use of ultrasound for thyroid cancer. Current opinion in oncology. 2010;22(1):11-16.
  214. Sakorafas GH, Christodoulou S, Lappas C, Safioleas M. Preoperative detection of cervical lymph node metastases in papillary thyroid cancer: a surgical perspective. Onkologie. 2009;32(12):762-766.
  215. Monteiro R, Han A, Etiwy M, Swearingen A, Krishnamurthy V, Jin J, Shin JJ, Berber E, Siperstein AE. Importance of surgeon-performed ultrasound in the preoperative nodal assessment of patients with potential thyroid malignancy. Surgery. 2018;163(1):112-117.
  216. Kouvaraki MA, Shapiro SE, Fornage BD, Edeiken-Monro BS, Sherman SI, Vassilopoulou-Sellin R, Lee JE, Evans DB. Role of preoperative ultrasonography in the surgical management of patients with thyroid cancer. Surgery. 2003;134(6):946-954; discussion 954-945.
  217. Stulak JM, Grant CS, Farley DR, Thompson GB, van Heerden JA, Hay ID, Reading CC, Charboneau JW. Value of preoperative ultrasonography in the surgical management of initial and reoperative papillary thyroid cancer. Archives of surgery. 2006;141(5):489-494; discussion 494-486.
  218. Davis JR, Hale AL, Ewing JA, Lokey JS. Instituting Ultrasound-Guided FNA for Thyroid Nodules into a General Surgery Residency Program: What We Learned. Journal of surgical education. 2018;75(3):594-600.
  219. Duprez R, Lebas P, Marc OS, Mongeois E, Emy P, Michenet P. Preoperative US-guided hook-needle insertion in recurrent lymph nodes of papillary thyroid cancer: a help for the surgeon. European journal of radiology. 2010;73(1):40-42.
  220. Triponez F, Poder L, Zarnegar R, Goldstein R, Roayaie K, Feldstein V, Lee J, Kebebew E, Duh QY, Clark OH. Hook needle-guided excision of recurrent differentiated thyroid cancer in previously operated neck compartments: a safe technique for small, nonpalpable recurrent disease. The Journal of clinical endocrinology and metabolism. 2006;91(12):4943-4947.
  221. Chami L, Hartl D, Leboulleux S, Baudin E, Lumbroso J, Schlumberger M, Travagli JP. Preoperative localization of neck recurrences from thyroid cancer: charcoal tattooing under ultrasound guidance. Thyroid : official journal of the American Thyroid Association. 2015;25(3):341-346.
  222. Sippel RS, Elaraj DM, Poder L, Duh QY, Kebebew E, Clark OH. Localization of recurrent thyroid cancer using intraoperative ultrasound-guided dye injection. World journal of surgery. 2009;33(3):434-439.
  223. Hartl DM, Chami L, Al Ghuzlan A, Leboulleux S, Baudin E, Schlumberger M, Travagli JP. Charcoal suspension tattoo localization for differentiated thyroid cancer recurrence. Annals of surgical oncology. 2009;16(9):2602-2608.
  224. Kang TW, Shin JH, Han BK, Ko EY, Kang SS, Hahn SY, Kim JS, Oh YL. Preoperative ultrasound-guided tattooing localization of recurrences after thyroidectomy: safety and effectiveness. Annals of surgical oncology. 2009;16(6):1655-1659.
  225. Lucchini R, Puxeddu E, Calzolari F, Burzelli F, Monacelli M, D'Ajello F, Macaluso R, Giammartino C, Ragusa M, De Feo P, Cavaliere A, Avenia N. Recurrences of thyroid well differentiated cancer: ultrasonography-guided surgical treatment. Minerva chirurgica. 2008;63(4):257-260.
  226. Solorzano CC, Carneiro DM, Ramirez M, Lee TM, Irvin GL, 3rd. Surgeon-performed ultrasound in the management of thyroid malignancy. The American surgeon. 2004;70(7):576-580; discussion 580-572.
  227. Curry JM, Ezzat WH, Merton DA, Goldberg BB, Cognetti DM, Rosen D, Pribitkin EA. Thyroid lymphosonography: a novel method for evaluating lymphatic drainage. The Annals of otology, rhinology, and laryngology. 2009;118(9):645-650.
  228. Ozdemir H, Ilgit ET, Yucel C, Atilla S, Isik S, Cakir N, Gokcora N. Treatment of autonomous thyroid nodules: safety and efficacy of sonographically guided percutaneous injection of ethanol. AJR American journal of roentgenology. 1994;163(4):929-932.
  229. Del Prete S, Russo D, Caraglia M, Giuberti G, Marra M, Vitale G, Lupoli G, Abbruzzese A, Capasso E. Percutaneous ethanol injection of autonomous thyroid nodules with a volume larger than 40 ml: three years of follow-up. Clinical radiology. 2001;56(11):895-901.
  230. Janowitz P, Ackmann S. Long-term results of ultrasound-guided ethanol injections in patients with autonomous thyroid nodules and hyperthyroidism. Medizinische Klinik. 2001;96(8):451-456.
  231. Brkljacic B, Sucic M, Bozikov V, Hauser M, Hebrang A. Treatment of autonomous and toxic thyroid adenomas by percutaneous ultrasound-guided ethanol injection. Acta radiologica. 2001;42(5):477-481.
  232. Guglielmi R, Pacella CM, Bianchini A, Bizzarri G, Rinaldi R, Graziano FM, Petrucci L, Toscano V, Palma E, Poggi M, Papini E. Percutaneous ethanol injection treatment in benign thyroid lesions: role and efficacy. Thyroid : official journal of the American Thyroid Association. 2004;14(2):125-131.
  233. Solymosi T, Gal I. Treatment of recurrent nodular goiters with percutaneous ethanol injection: a clinical study of twelve patients. Thyroid : official journal of the American Thyroid Association. 2003;13(3):273-277.
  234. Cho YS, Lee HK, Ahn IM, Lim SM, Kim DH, Choi CG, Suh DC. Sonographically guided ethanol sclerotherapy for benign thyroid cysts: results in 22 patients. AJR American journal of roentgenology. 2000;174(1):213-216.
  235. Valcavi R, Frasoldati A. Ultrasound-guided percutaneous ethanol injection therapy in thyroid cystic nodules. Endocrine practice : official journal of the American College of Endocrinology and the American Association of Clinical Endocrinologists. 2004;10(3):269-275.
  236. Mauz PS, Maassen MM, Braun B, Brosch S. How safe is percutaneous ethanol injection for treatment of thyroid nodule? Report of a case of severe toxic necrosis of the larynx and adjacent skin. Acta oto-laryngologica. 2004;124(10):1226-1230.
  237. Chung SR, Suh CH, Baek JH, Park HS, Choi YJ, Lee JH. Safety of radiofrequency ablation of benign thyroid nodules and recurrent thyroid cancers: a systematic review and meta-analysis. International journal of hyperthermia : the official journal of European Society for Hyperthermic Oncology, North American Hyperthermia Group. 2017;33(8):920-930.
  238. Ha EJ, Baek JH, Kim KW, Pyo J, Lee JH, Baek SH, Dossing H, Hegedus L. Comparative efficacy of radiofrequency and laser ablation for the treatment of benign thyroid nodules: systematic review including traditional pooling and bayesian network meta-analysis. The Journal of clinical endocrinology and metabolism. 2015;100(5):1903-1911.
  239. Pacella CM, Mauri G, Cesareo R, Paqualini V, Cianni R, De Feo P, Gambelunghe G, Raggiunti B, Tina D, Deandrea M, Limone PP, Mormile A, Giusti M, Oddo S, Achille G, Di Stasio E, Misischi I, Papini E. A comparison of laser with radiofrequency ablation for the treatment of benign thyroid nodules: a propensity score matching analysis. International journal of hyperthermia : the official journal of European Society for Hyperthermic Oncology, North American Hyperthermia Group. 2017;33(8):911-919.
  240. Papini E, Guglielmi R, Bizzarri G, Pacella CM. Ultrasound-guided laser thermal ablation for treatment of benign thyroid nodules. Endocrine practice : official journal of the American College of Endocrinology and the American Association of Clinical Endocrinologists. 2004;10(3):276-283.
  241. Sung JY, Baek JH, Jung SL, Kim JH, Kim KS, Lee D, Kim WB, Na DG. Radiofrequency ablation for autonomously functioning thyroid nodules: a multicenter study. Thyroid : official journal of the American Thyroid Association. 2015;25(1):112-117.
  242. Wang L, Xu D, Yang Y, Li M, Zheng C, Qiu X, Huang B. Safety and efficacy of ultrasound-guided percutaneous thermal ablation in treating low-risk papillary thyroid microcarcinoma: A pilot and feasibility study. Journal of cancer research and therapeutics. 2019;15(7):1522-1529.
  243. Lim HK, Cho SJ, Baek JH, Lee KD, Son CW, Son JM, Baek SM. US-Guided Radiofrequency Ablation for Low-Risk Papillary Thyroid Microcarcinoma: Efficacy and Safety in a Large Population. Korean journal of radiology. 2019;20(12):1653-1661.
  244. Makani S, Kim W, Gaba AR. Struma Ovarii with a focus of papillary thyroid cancer: a case report and review of the literature. Gynecologic oncology. 2004;94(3):835-839.
  245. Ranzini AC, Ananth CV, Smulian JC, Kung M, Limbachia A, Vintzileos AM. Ultrasonography of the fetal thyroid: nomograms based on biparietal diameter and gestational age. Journal of ultrasound in medicine : official journal of the American Institute of Ultrasound in Medicine. 2001;20(6):613-617.
  246. Huel C, Guibourdenche J, Vuillard E, Ouahba J, Piketty M, Oury JF, Luton D. Use of ultrasound to distinguish between fetal hyperthyroidism and hypothyroidism on discovery of a goiter. Ultrasound in obstetrics & gynecology : the official journal of the International Society of Ultrasound in Obstetrics and Gynecology. 2009;33(4):412-420.
  247. Khater C, Ceccaldi PF, Poujade O, Banige M, Ottenwalter A, Luton D. Foetal thyroid dysfunction: treat the mother first! Hormone research in paediatrics. 2015;83(2):136-140.
  248. Agrawal P, Ogilvy-Stuart A, Lees C. Intrauterine diagnosis and management of congenital goitrous hypothyroidism. Ultrasound in obstetrics & gynecology : the official journal of the International Society of Ultrasound in Obstetrics and Gynecology. 2002;19(5):501-505.
  249. Morine M, Takeda T, Minekawa R, Sugiyama T, Wasada K, Mizutani T, Suehara N. Antenatal diagnosis and treatment of a case of fetal goitrous hypothyroidism associated with high-output cardiac failure. Ultrasound in obstetrics & gynecology : the official journal of the International Society of Ultrasound in Obstetrics and Gynecology. 2002;19(5):506-509.
  250. Ribault V, Castanet M, Bertrand AM, Guibourdenche J, Vuillard E, Luton D, Polak M, French Fetal Goiter Study G. Experience with intraamniotic thyroxine treatment in nonimmune fetal goitrous hypothyroidism in 12 cases. The Journal of clinical endocrinology and metabolism. 2009;94(10):3731-3739.
  251. Luton D, Le Gac I, Vuillard E, Castanet M, Guibourdenche J, Noel M, Toubert ME, Leger J, Boissinot C, Schlageter MH, Garel C, Tebeka B, Oury JF, Czernichow P, Polak M. Management of Graves' disease during pregnancy: the key role of fetal thyroid gland monitoring. The Journal of clinical endocrinology and metabolism. 2005;90(11):6093-6098.
  252. Perry RJ, Hollman AS, Wood AM, Donaldson MD. Ultrasound of the thyroid gland in the newborn: normative data. Archives of disease in childhood Fetal and neonatal edition. 2002;87(3):F209-211.
  253. Yasumoto M, Inoue H, Ohashi I, Shibuya H, Onishi T. Simple new technique for sonographic measurement of the thyroid in neonates and small children. Journal of clinical ultrasound : JCU. 2004;32(2):82-85.
  254. Kreisner E, Camargo-Neto E, Maia CR, Gross JL. Accuracy of ultrasonography to establish the diagnosis and aetiology of permanent primary congenital hypothyroidism. Clinical endocrinology. 2003;59(3):361-365.
  255. Semiz S, Senol U, Bircan, Gumuslu S, Bilmen S, Bircan I. Correlation between age, body size and thyroid volume in an endemic area. Journal of endocrinological investigation. 2001;24(8):559-563.
  256. Kaloumenou I, Alevizaki M, Ladopoulos C, Antoniou A, Duntas LH, Mastorakos G, Chiotis D, Mengreli C, Livadas S, Xekouki P, Dacou-Voutetakis C. Thyroid volume and echostructure in schoolchildren living in an iodine-replete area: relation to age, pubertal stage, and body mass index. Thyroid : official journal of the American Thyroid Association. 2007;17(9):875-881.
  257. Yazici B, Simsek E, Erdogmus B, Bahcebasi T, Aktas A, Buyukkaya R, Uzun H, Safak AA. Evaluation of the thyroid blood flow with Doppler ultrasonography in healthy school-aged children. European journal of radiology. 2007;63(2):286-289.
  258. Drozd V, Polyanskaya O, Ostapenko V, Demidchik Y, Biko I, Reiners C. Systematic ultrasound screening as a significant tool for early detection of thyroid carcinoma in Belarus. Journal of pediatric endocrinology & metabolism : JPEM. 2002;15(7):979-984.
  259. Yamashita S, Suzuki S, Suzuki S, Shimura H, Saenko V. Lessons from Fukushima: Latest Findings of Thyroid Cancer After the Fukushima Nuclear Power Plant Accident. Thyroid : official journal of the American Thyroid Association. 2018;28(1):11-22.
  260. Kopecky KJ, Onstad L, Hamilton TE, Davis S. Thyroid ultrasound abnormalities in persons exposed during childhood to 131I from the Hanford nuclear site. Thyroid : official journal of the American Thyroid Association. 2005;15(6):604-613.
  261. Kindler S, Roser M, Below H, Hoffmann W, Kohlmann T, Kramer A, Kirsch G, Volzke H. Thyroid disorders in employees of a nuclear power plant. Thyroid : official journal of the American Thyroid Association. 2006;16(10):1009-1017.
  262. Chung WY, Chang HS, Kim EK, Park CS. Ultrasonographic mass screening for thyroid carcinoma: a study in women scheduled to undergo a breast examination. Surgery today. 2001;31(9):763-767.
  263. Imaizumi M, Usa T, Tominaga T, Akahoshi M, Ashizawa K, Ichimaru S, Nakashima E, Ishii R, Ejima E, Hida A, Soda M, Maeda R, Nagataki S, Eguchi K. Long-term prognosis of thyroid nodule cases compared with nodule-free controls in atomic bomb survivors. The Journal of clinical endocrinology and metabolism. 2005;90(9):5009-5014.
  264. Garra BS. Imaging and estimation of tissue elasticity by ultrasound. Ultrasound quarterly. 2007;23(4):255-268.
  265. Hong Y, Liu X, Li Z, Zhang X, Chen M, Luo Z. Real-time ultrasound elastography in the differential diagnosis of benign and malignant thyroid nodules. Journal of ultrasound in medicine : official journal of the American Institute of Ultrasound in Medicine. 2009;28(7):861-867.
  266. Rago T, Santini F, Scutari M, Pinchera A, Vitti P. Elastography: new developments in ultrasound for predicting malignancy in thyroid nodules. The Journal of clinical endocrinology and metabolism. 2007;92(8):2917-2922.
  267. Luo S, Kim EH, Dighe M, Kim Y. Thyroid nodule classification using ultrasound elastography via linear discriminant analysis. Ultrasonics. 2011;51(4):425-431.
  268. Lippolis PV, Tognini S, Materazzi G, Polini A, Mancini R, Ambrosini CE, Dardano A, Basolo F, Seccia M, Miccoli P, Monzani F. Is elastography actually useful in the presurgical selection of thyroid nodules with indeterminate cytology? The Journal of clinical endocrinology and metabolism. 2011;96(11):E1826-1830.
  269. Cappelli C, Pirola I, Gandossi E, Formenti A, Agosti B, Castellano M. Elastography Evaluation of Benign Thyroid Nodules in Patients Affected by Hashimoto's Thyroiditis. International journal of endocrinology. 2015;2015:367054.
  270. Ruchala M, Szczepanek E, Sowinski J. Sonoelastography in de Quervain thyroiditis. The Journal of clinical endocrinology and metabolism. 2011;96(2):289-290.
  271. Khanna J, Mohil RS, Chintamani, Bhatnagar D, Mittal MK, Sahoo M, Mehrotra M. Is the routine drainage after surgery for thyroid necessary? A prospective randomized clinical study [ISRCTN63623153]. BMC surgery. 2005;5:11.
  272. Milas M, Mensah A, Alghoul M, Berber E, Stephen A, Siperstein A, Weber CJ. The impact of office neck ultrasonography on reducing unnecessary thyroid surgery in patients undergoing parathyroidectomy. Thyroid : official journal of the American Thyroid Association. 2005;15(9):1055-1059.
  273. Casadei R, Perenze B, Vescini F, Piccoli L, Zanini N, Minni F. Usefulness of the ultrasonically activated shears in total thyroidectomy. Chirurgia italiana. 2004;56(6):843-848.
  274. Witzel K, von Rahden BH, Stein HJ. The effect of ultrasound dissection in thyroid surgery. European surgical research Europäische chirurgische Forschung Recherches chirurgicales europeennes. 2009;43(2):241-244.
  275. Kovatcheva RD, Vlahov JD, Stoinov JI, Zaletel K. Benign Solid Thyroid Nodules: US-guided High-Intensity Focused Ultrasound Ablation-Initial Clinical Outcomes. Radiology. 2015;276(2):597-605.

Adipose Tissue: Physiology to Metabolic Dysfunction

ABSTRACT

 

Like the obesity epidemic, our understanding of adipocytes and adipose tissue is expanding. Just in the past decade, substantial advances have led to new insights into the contributions of adipose tissue to normal physiology and obesity-related complications, which places adipocyte biology at the epicenter of a global pandemic of metabolic diseases. In addition to detailing the types, locations, and functions of different adipose tissue depots, this chapter will review the secretory capacities of adipose tissue. Arguably one of the most significant discoveries in the last two decades of adipocyte research is that not only do adipocytes release endocrine hormones, but fat cells and adipose tissue secrete a variety of effectors, including exosomes, miRNA, lipids, inflammatory cytokines, and peptide hormones that act in both paracrine and endocrine capacities to impact local and systemic metabolic responses. The origins of adipocytes via progenitor cells and the process of adipocyte development are discussed. Inflammation, metabolically healthy fat, and adipose tissue expansion are also considered. Finally, several emerging research areas in fat cell biology with therapeutic potential in the management patients who are overweight and have obesity are summarized.

 

INTRODUCTION: AN HISTORICAL PERSPECTIVE ON ADIPOSE TISSUE BIOLOGY

 

The first published citation referencing adipose tissue (AT) dates to 1837. Subsequent sporadic single AT citations appeared in in the literature until the 1940’s, including a 1933 publication in Biochemical Journal examining the degree of fatty acid unsaturation in human AT in relation to its depth from the skin surface (1). The first year in which two AT-related citations were recorded was in 1942. In 1947, nearly ten AT citations appeared. Adipose tissue remained understudied for decades due to the misconception that it was simply an inert energy storage depot, but recent discoveries of AT’s wider role in cell and whole-body signaling have created a scientific renaissance in this field. As of early 2019, over 139,000 citations involving adipocytes or AT are now discoverable.

 

The earliest recognized function of adipocytes was the storage of energy in the form of triacylglycerols (TAGs). It was not until the mid-1980s that the secretory functions of AT and the production of adipocyte-specific proteins were revealed. At that time, a serine protease named adipsin was shown to be secreted from cultured adipocytes and reported to be reduced in mouse models of obesity compared to lean littermates (2). Acylation stimulating protein, a member of the alternative complement family, was also revealed to be produced by AT (3) and implicated in lipid storage (4). Although the functions of these AT secretory products remain poorly understood, their discovery revealed adipocytes and AT to be significant sources of a variety of protein products, including many endocrine hormones. Arguably one of the most important of these discoveries was leptin (5), a bona-fide adipocyte-derived hormone that clearly acts not only as an afferent “adipostat” signal of fat mass to central brain centers in the regulation of body weight (5) but also has peripheral actions that impact glucose metabolism (6) and immune function (7).

 

In addition, adipocytes are also highly sensitive to insulin and involved in the regulation of blood glucose levels. Insulin action on fat cells stimulates glucose uptake and modulates lipid metabolism by increasing the accumulation and decreasing the breakdown of TAGs (and subsequent release of free fatty acids into the circulation) within the adipocyte. The importance of each of these 3 fat cell functions (Figure 1) – lipid storage, secretory function, and insulin sensitivity – is underscored by the demonstration that disruption of any one role has profound systemic ramifications in mice and man that can contribute to a variety of obesity-related metabolic disease states (8).

 

The first CDC statistics reporting obesity rates over 20% in many US states also appeared in the late 1990’s, as did literature from a variety of disciplines showing that obesity, or excess adipose tissue, enhanced the risk of metabolic diseases, particularly type 2 diabetes (T2D). This was a substantial shift in thinking from the previous two decades when AT was not considered to have much importance or relevance to T2D. In addition to metabolic diseases, obesity is associated with increased risk of 13 types of cancer that account for ~40% of all cancers diagnosed in the United States (9).

 

Today, obesity and accompanying epidemics of co-morbidities have become global problems.  While in 2015–2016 the prevalence of obesity was 39.8% in adults and 18.5% in youth in the USA (10), the World Health Organization (WHO) reports that obesity has nearly tripled across the world since 1975, and in 2016 more than 1.9 billion adults were overweight and over 650 million were obese. Today, with most of the world's population living in countries where overweight and obesity account for more deaths than malnutrition (underweight), excess AT presents a major challenge to chronic disease prevention and health across the planet. This global epidemic can be attributed to advancing economies and the adoption of mechanized transport, urbanization, commercial growth, industrialization, a progressively more sedentary lifestyle, and a nutritional transition to processed foods and high calorie diets over the last 30 years (11). Besides preventing obesity by promoting a healthy lifestyle through diet and exercise, one of the best ways for modern-day physicians and scientists to combat the global menace of obesity is to better understand AT.

Figure 1. Physiological characteristics of adipocytes. Disruption of any one of these fat cell functions may lead to the development of systemic metabolic dysfunction.

ADIPOCYTE PHYSIOLOGY

 

Adipocyte Hues – White, Brown, Beige and Pink

 

Adipose tissue has historically been classified into two types, white adipose tissue (WAT) and brown adipose tissue (BAT), which are visibly distinguishable based on tissue color. The white and brown adipocytes comprising these depots exhibit physiological differences, which give rise to specialized tissue functions. White adipose tissue, which is critical for energy storage, endocrine communication, and insulin sensitivity, comprises the largest AT volume in most mammals including humans. In contrast, BAT is largely present in mammals postnatally and during hibernation. Brown adipose tissue uses energy for non-shivering heat production, which is critical for body temperature maintenance. While BAT was originally thought to only be present in infant humans, imaging studies have revealed metabolically active BAT in the supraclavicular and thoracic regions of adults (12–14). Although women have increased BAT mass and activity over men (14,15), the chance of detecting BAT activity in either sex has been shown to be inversely correlated with age and body mass index (BMI) (14). Seasonal correlations have also been observed with BAT activity being higher in the winter and lower in the summer, possibly due to either the temperature or, more likely, the photoperiod (14,15). In healthy humans, BAT activity contributes to whole-body fat oxidation and diet-induced thermogenesis (16), supporting a physiological role for this AT depot in adults.

 

Brown and white adipocytes differ in shape, size, and the intracellular structure of their organelles (Figure 2). White adipocytes are generally spherical in shape and each contains a large, single lipid droplet that pushes all other organelles, including the nucleus, to the cell’s periphery. Brown adipocytes contain multiple lipid droplets dispersed throughout a more ellipsoidal-shaped cell that is enriched with iron-containing mitochondria, giving the cell (and the BAT as a whole) a brownish hue. The thermogenic activity of brown adipocytes is conferred by the presence of its numerous mitochondria containing uncoupling protein 1 (UCP-1), a proton transporter that short-circuits the ATP (energy)-generating proton gradient and allows for concurrent heat production as protons flow back into the mitochondrial matrix (17). Brown fat cells typically grow to 15 to 50 µm, while white fat cells have a larger capacity for lipid storage and can expand to nearly 100 µm in diameter (18). The capacity of white adipocytes to expand in number and size is depot-dependent and is discussed in more detail in the Adipose Tissue Expandability and Metabolic Health section.

Figure 2. Adipocyte types are described by color hues. The primary characteristic of an adipocyte is its ability to store lipid; white, brown, beige, and pink adipocytes all share this property. However, each type of fat cell is somewhat specialized and has a distinct intracellular distribution of organelles and gene expression profile. All fat cells have Golgi and endoplasmic reticulum, but these organelles make up a more significant portion of pink adipocytes than other adipocyte types.

Recently, two additional adipocyte hues – beige and pink – have been described. Beige adipocytes display characteristics of both brown and white fat cells (Figure 2) and typically develop within subcutaneous WAT from a distinct subset of preadipocytes (19) or via the  

transdifferentiation of existing white adipocytes (20,21). However, gene expression analyses indicate that beige fat cells represent a distinct type of thermogenic fat cell (19). Beige adipocytes were originally observed to arise in response to cold exposure in rodents (22,23); however, many studies have since identified that diet (24), exercise (25), pre- and post-biotics (26), pharmaceutical agents, numerous plant-based bioactives, and even adipokines (27)can also induce “beiging” or “browning” of WAT, which may protect against obesity and associated metabolic dysfunction. The “beiging” of WAT is inducible in both mice and humans (28), but this process is more highly observed in mice.

 

Pink adipocytes were first described in 2014, arising in the subcutaneous WAT of female mice during days 17-18 of pregnancy and persisting throughout lactation. These fat cells appear to derive from white adipocytes that take on epithelial-like features to form milk-secreting alveoli, giving the tissue a pink hue (29). Pink adipocytes are characterized by compartmentalized lipid droplets, cytoplasmic projections, and abundant organelles including mitochondria, peroxisomes, and rough endoplasmic reticulum, that show a structure more typical of epithelial cells. While reversible transdifferentiation appears to be responsible for the development and disappearance of pink adipocytes during pregnancy, lactation, and post-lactation in rodents (30), it remains uncertain whether or not pink adipocytes form in humans. Notably, loss of a key adipogenic transcription factor within the mammary secretory epithelium creates a pro-breast tumorigenic environment and indicates that the reversible white-to-pink transition might reveal insights into breast cancer biology (29,31). Further investigations into adipocyte plasticity might therefore identify novel therapeutic targets to combat obesity and its pathological consequences, as well as cancer. However, since WAT makes up the largest AT volume in the human body and undergoes the most expansion during obesity, in this chapter we will focus on the roles that white adipocytes and WAT play in normal physiology and metabolic dysfunction.

 

Adipose Tissue in the Regulation of Lipid Metabolism

 

Adipose tissue stores body fat as neutral TAGs and represents the chief energy reservoir within mammals. Although many diverse cell types are found in whole AT, adipocytes constitute the largest cell volumes and are the defining AT cell type. White adipocytes are characterized by their large unilocular central lipid droplets (cLDs). However, the biogenesis of unilocular LDs in adipocytes is poorly understood due to the fragile nature of WAT.

 

Using live-cell imaging combined with fluorescent labeling techniques, the cytoarchitecture of unilocular adipocytes (Figure 3) and spatiotemporal dynamics of lipid droplet formation have been investigated (32). As shown in Figure 3, cytoplasmic nodules containing micro LDs (mLDs; small green fluorescent protein (GFP)-negative spheres within the cytoplasm) appear on the surface of fat cells, pushed to the edges by the large cLD. Surprisingly, the cytoplasm and organelles do not distribute uniformly around the edge of the cell, but instead form numerous, discrete cytoplasmic nodules connected via a thin layer of GFP-positive cytoplasm. The largest nodule also contains the nucleus, which is surrounded by a thicker layer of cytoplasm. The electron micrograph (Figure 3F) shows the close contacts between mLDs and mitochondria. Furthermore, additional nascent lipid droplets can be visualized budding off from the smooth ER (sER). Studies using a fluorescent-labeled free fatty acid (FFA) analog revealed that exogenously added lipids were rapidly taken up by the fat cell and concurrently esterified to TAG and absorbed by mLDs prior to packaging within the cLD. The lipid transfer followed a unidirectional path from mLD to cLD and provides insight into adipose tissue growth via fat cell hypertrophy (32).  

Figure 3. Architecture of primary unilocular adipocytes. Figure adapted from (32). The cytoplasm and nuclei of adipocytes and stromovascular cells were labeled by infecting visceral WAT explants from nonhuman primates with an adenoviral vector encoding enhanced green fluorescent protein (eGFP). Two days after infection, live explants were examined by for GFP expression using confocal microscopy. Cellular and subcellular features are labeled: cLD, central lipid droplet; Cyt, cytoplasm; LDM, lipid droplet membrane; mLD, micro-LD; N, nucleus; PM, plasma membrane; sER, smooth ER. (A) GFP-positive unilocular adipocytes (spheres) and stromovascular cells (asterisks) residing in WAT. The image represents the sum of all confocal slices. Bar, 10 um. (B) Single confocal section of the image in A. Enhanced magnification of adipocytes containing cytoplasmic nodules (C) and perinuclear cytoplasm (D). (E) Schematic representation a unilocular adipocyte demonstrates that the cLD is a sphere tightly fitted within the cell, whereas the cytoplasm collects in multiple organelle- and mLD-containing nodules. (F) Electron micrograph of a unilocular adipocyte from a visceral WAT explant that was fixed and processed for electron microscopy. Asterisks mark contact sites between mitochondria and mLDs, whereas arrowheads point towards vesicles budding off the ER tubules. Bar, 500 nm.

Adipocytes store TAG under conditions of energy surplus and release fatty acids to supply to other tissues during fasting or times of high energy demand. As such, AT is central to the regulation of systemic lipid metabolism, and nutritional and hormonal cues serve to balance lipid storage and breakdown within the fat cell (Figure 4).

 

Figure 4. A critical balance between lipogenesis and lipolysis within adipocytes must be established to maintain whole body insulin sensitivity and energy homeostasis. Lipogenesis is shown on the left (gray arrows mark the pathway), whereas lipolysis is shown on the right and is marked by black arrows. Nutritional and hormonal cues regulate both processes. Lipid droplet associated proteins, such as perilipin and comparative gene identification-58 (CGI-58) are not shown but play important roles in lipolysis. CD36 (cluster of differentiation 36) is a fatty acid transporter that facilitates entry of free fatty acids (FFAs) into the cell. Insulin stimulates glucose uptake into fat cells by increasing the localization of the insulin responsive glucose transporter, GLUT4, within the plasma membrane. Other abbreviations: VLDL-TG – triglyceride-containing very low density lipoprotein; LPL – lipoprotein lipase; ACC - acetyl-CoA carboxylase 1; FAS – fatty acid synthase; G3P – glycerol 3 phosphate; DGAT - diacylglycerol acyltransferase; β-AR – β-adrenergic receptor; NA – noradrenaline; AC – adenylyl cyclase; PKA – protein kinase A; ATGL - adipocyte triglyceride lipase; HSL - hormone sensitive lipase; MGL - monoacylglycerol lipase; TAG – triacylglycerol; DAG – diacylglycerol; MAG – monoacylglycerol.

LIPOGENESIS

 

Adipocytes accumulate lipid via one of two processes (Figure 4).  In the first process, under normal daily feeding conditions adipocytes take up dietary lipids from the circulation in the form of FFA’s liberated from circulating TAGs via the action of lipoprotein lipase (LPL) (33). Adipocytes secrete LPL, which is transported to the adjacent capillary lumen to catalyze the hydrolysis of FFA’s from circulating triglyceride-containing lipoproteins (34,35), such as chylomicrons produced in the small intestine and very low density lipoproteins (VLDLs) synthesized by the liver (36). Adipocytes also take up glucose, which is converted to glycerol and serves as the backbone for the sequential esterification of fatty acids for form TAG. The final step in TAG synthesis, re-esterification of circulating free fatty acids, mediated by diacylglycerol acyltransferase (DGAT) (37,38). The second process is by de novo lipogenesis (DNL) within the adipocytes themselves. Lipogenesis comprises both de novo synthesis of fatty acids from acetyl-coenzyme A (acetyl-CoA) and the esterification of these fatty acids to a glycerol backbone producing TAGs (Figure 4). De novolipogenesis can occur in the fasting and fed states (36). Following a meal, especially one high in carbohydrates, excess glucose oxidation yields elevated levels of acetyl-CoA that become substrate to generate fatty acids.  This occurs through actions of the DNL enzymes acetyl-CoA carboxylase 1 (ACC1) and fatty acid synthase (FAS) to convert acetyl-CoA to palmitate, which can then be elongated and desaturated to form other fatty acid species (39).

 

Surprisingly, in rodents DNL is relatively low in WAT compared to BAT and liver, and it plays an even lesser role in WAT lipid storage in humans under physiological conditions (40,41). Typically, hepatic DNL activity exceeds that of AT and is a more substantial contributor DNL-generated circulating lipids. However, in humans fed high-carbohydrate diets, liver DNL contributes only a small portion of total de novo fat biosynthesis, suggesting that AT contributes significantly to whole body DNL when there is a carbohydrate surplus (39,42). Under this condition, adipocyte DNL is usually quite low but has been shown to be important for whole body substrate metabolism (43,44) as inhibition of WAT DNL is associated with insulin resistance (45).

 

A primary transcriptional regulator of adipocyte DNL is carbohydrate response element-binding protein (ChREBP) (39). Mice lacking AT ChREBP have decreased DNL and insulin resistance (46). The other major DNL regulator in AT is sterol regulatory element-binding protein 1 (SREBP1). Mice with whole body knockout of SREBP1 do not display decreased lipogenic gene expression in AT (45,47), thus supporting ChREBP as the primary lipogenic transcription factor driving AT DNL. However, a new mouse model of inducible, overexpression of insulin-induced gene 1 (Insig1), an inhibitor of SREBP1 activation and transcriptional activity, demonstrated that several acute and chronic white adipocyte-specific compensatory mechanisms are activated to restore adipocyte DNL in the absence of SREBP1 activity (44). Decreased SREBP1 activity prior to this compensation and during conditions where compensation was inactivated result in decreased lipogenic gene expression, impaired whole body glucose tolerance, and elevated lipid clearance (44) suggesting that both SREBP1 and ChREBP play important roles in adipocyte DNL.

 

Enhanced AT DNL can produce favorable lipid species that may be therapeutically advantageous in the context of obesity and insulin resistance (48). Adipocytes synthesize and secrete a novel family of bioactive lipids, known as the branched fatty acid esters of hydroxyl fatty acids (FAHFAs). Although FAHFAs are found in many tissues, the highest levels are in white and brown AT, and their production is likely dependent on AT lipogenesis as disruption of adipocyte DNL impairs their synthesis (39,49). Over 1000 structurally distinct FAHFAs have been predicted based on in silicoanalyses and at least 20 FAHFA families have already been identified in mammalian tissues (50). The serum and subcutaneous AT levels of one FAHFA family, palmitic acid esters of hydroxyl steric acids (PAHSAs) have been shown to be higher in insulin-sensitive compared to insulin-resistant individuals (51). In animal models, PAHSAs have been shown to decrease inflammation and enhance whole body insulin sensitivity (39,49). Recent evidence from a mouse model of high-fat diet (HFD)-induced insulin resistance demonstrates that PAHSAs act via both direct and indirect mechanisms to improve insulin sensitivity in multiple metabolic tissues, such glycolytic skeletal muscle, heart, liver, and AT. In WAT explants, PAHSAs directly inhibit lipolysis and enhance insulin’s ability to suppress lipolysis. While PASHAs can also directly inhibit endogenous glucose production (EGP) in isolated hepatocytes, the decreased AT lipolysis indirectly attenuates EGP because of reduced glycerol (gluconeogenic substrate) delivery to the liver (50).

 

Additional evidence from humans support a role for increased DNL and ChREBP activity in maintaining metabolic health. These include restoration of DNL in WAT following as bariatric surgery-induced weight loss (52) and reported observations of elevated WAT DNL in other metabolically favorable states including caloric restriction and adaptive thermogenesis (53,54). Collectively, these studies in mice and man support a potential role of WAT DNL in metabolic health.

 

LIPOLYSIS

 

Under physiological conditions when metabolic fuels are low and/or energy demand is high, such as fasting, exercise, and cold exposure, adipocytes mobilize their TAG stores via the catabolic process of lipolysis to supply fuel to peripheral tissues (55). Lipolysis is a highly regulated biochemical process that generates glycerol and FFAs from the enzymatic cleavage of TAGs by lipases (36) and can occur in all tissues, although it is most prevalent in AT where the bulk of TAG is stored. As shown in Figure 4, TAGs are broken down into diacylglycerols (DAGs) and monoacylglycerols (MAGs) by the sequential action of adipocyte triglyceride lipase (ATGL), hormone sensitive lipase (HSL), and monoacylglycerol lipase (MGL). At each step a single FFA is released, and in the final step MGL releases the glycerol backbone from the last FFA. These breakdown products can be re-esterified within the adipocyte or released into circulation to be used by other tissues (36,55), including by the liver for gluconeogenesis (glycerol) and for oxidative phosphorylation by muscle or other oxidative tissues (56).

 

Lipolysis is controlled by sympathetic nervous system (SNS) input as well as a variety of hormones (55). The best understood of these regulators is the catecholamine, noradrenaline (NA), also known as norepinephrine. Noradrenaline stimulates β-adrenergic receptors (Figure 4), which, in turn, stimulate protein kinase A (PKA) via adenylyl cylase (AC)-mediated production of cyclic AMP (cAMP). PKA activates the lipolytic action of ATGL and HSL by different mechanisms. Several lipid droplet-associated proteins, such as perilipin 1 (PLIN1) and comparative gene identification-58 (CGI-58) are also important in regulating lipolysis (57,58). Whereas PKA can directly phosphorylate and activate HSL (59–62), it primarily stimulates ATGL activity indirectly by phosphorylating PLIN1. This phosphorylation releases CGI-58 to potently activate ATGL (58,63,64). Non-adrenergic lipolytic stimuli include glucocorticoids, natriuretic peptides, growth hormone, and tumor necrosis factor alpha (TNFα) (58). These hormones are typically less potent lipolytic inducers than β-adrenergic stimulation and the molecular mechanisms responsible for their lipolytic abilities have not been clearly elucidated. However, some of these hormones clearly utilize different pathways than β-adrenergic signaling with additive or synergistic affects to increase lipolysis (55,58).

 

After a meal, the post-prandial increase in circulating insulin readily suppresses lipolysis (65) by increasing the activity of phosphodiesterase 3 (PDE3B) and decreasing cAMP levels (58). In the fasting state, insulin levels drop and NA is released, thus promoting lipolysis (66). Physiologically, exercise is another major pro-lipolytic stimulus in humans (58). Growth hormone, along with NA, adrenaline, and cortisol increase with exercise intensity, while insulin levels decrease. These changes culminate in an overall lipolytic response, the magnitude of which depends on exercise intensity and duration (58,67).

 

When AT becomes insulin resistant, as occurs in patients with diabetes and may also be present in patients with obesity, insulin’s ability to inhibit adipocyte lipolysis and reduce serum levels of FFA and glycerol are impaired. As a result, excessive lipolysis leads to increased FFA levels in both the fasted and fed state. Constant exposure of the liver and muscle to these high FFA levels is thought to promote the uptake and ectopic storage of lipids in these tissues (68). Ectopic lipids have been shown to impair insulin signaling, and thus insulin resistance at the level of adipocyte via increased lipolysis may be a major contributor to whole body insulin resistance (69). In addition to impaired insulin responsiveness in fat cells, elevated lipolysis in obesity may be mediated by decreased expression of adipocyte lipid droplet proteins such as PLIN1 and Fsp27/Cidec (fat-specific protein 27/cell death-inducing DFFA-like effector c) (70). These proteins coat the lipid droplet and promote TAG retention via the inhibition of lipolysis, and mice or humans deficient for PLIN1 (71) or Fsp27 (72,73) exhibit lipodystrophy and insulin resistance (70). Interestingly, adipocyte-selective gene deletions or transgenic overexpression mouse models of proteins involved in insulin signaling, glucose and lipid metabolism demonstrate parallel modulation of adipocyte insulin action and systemic insulin sensitivity or glucose tolerance.

 

In addition to potent anti-lipolytic action (58), insulin also stimulates lipogenesis (74) by activating LPL (activation) and increasing the transcription of lipogenic enzymes (74). Growth hormone antagonizes insulin by promoting lipolysis and inhibiting lipogenesis (36,58). The insulin-sensitizing, anti-inflammatory lipids (PAHSAs) generated during AT DNL and the excess basal lipolysis associated with ectopic lipid deposition and insulin resistance make both AT lipogenesis and lipolysis attractive targets for pharmaceutical intervention. On the other hand, the balance between lipid storage, mobilization, and utilization is homeostatically regulated through a complex interaction of often redundant hormonal signaling, neurological input, and nutrient flow. These intricacies complicate attempts to develop therapies targeting one aspect of lipid metabolism since disrupting the balance between lipolysis and lipogenesis may, in turn, have unanticipated effects on insulin sensitivity and whole-body energy homeostasis.

 

Figure 5. The Adipocyte Secretome. Fat cells express and release numerous protein, lipid, and nucleic acid factors that can act on other nearby or distant tissues within the body in a paracrine or endocrine manner. Leptin, adiponectin, and resistin are highlighted here because they are exclusively secreted from mouse adipocytes, while the other factors can also be secreted from other cell types. The arrow-headed line representing secretion of resistin is dashed since in humans, macrophages, and not adipocytes, primarily produce this adipokine. Abbreviations are RBP4 – retinol binding protein 4, BMPs – bone morphogenetic proteins, PAI-1 – plasminogen activator inhibitor 1, miRNA – microRNA, FFA – free fatty acid, FAHFA - fatty acid esters of hydroxyl fatty acids, PAHSA – palmitic-acid-hydroxy-steric-acid, FGF21 – fibroblast growth factor 21.

 

Endocrine Properties of Adipose Tissue

 

Adipocytes and other AT cells secrete a variety of mediators, including exosomes, miRNA, lipids, inflammatory cytokines, and peptide hormones that act in both paracrine and endocrine modes (Figure 5) (75). Although adipocytes secrete a large variety of bioactive molecules with widespread systemic effects contributing to numerous physiological and pathological processes, the autocrine and paracrine actions of these molecules are highly complex, and our understanding of these processes is likely rudimentary. However, substantial progress has been made studying three endocrine hormones that are almost exclusively produced in adipocytes and function to regulate food intake, the reproductive axis, insulin sensitivity, and immune responses. These hormones are leptin, adiponectin, and resistin, and we review their expression in obesity, their receptors, and effects in target tissues including metabolic actions (Figure 6). While not produced in human adipocytes directly but secreted instead by AT macrophages, resistin has similar functions in mouse and man. The dysregulation of any one of these hormones can contribute to systemic metabolic dysfunction, as well as to the pathogenesis of chronic metabolic diseases and some types of cancer.

Figure 6. Summary of adipocyte-specific adipokines, and their actions on other tissues. Abbreviations: TLR4 - Toll-like receptor 4; CAP1 - adenylyl cyclase-associated protein 1; AdipoR1 & R2 - Adiponectin receptors 1 and 2; CNS - Central nervous system; FAO – fatty acid oxidation; EE – energy expenditure.

LEPTIN

 

The first discovered endocrine hormone of adipocyte origin was leptin (5). In 1949 spontaneously occurring obese offspring in a Jackson Laboratories’ non-obese mouse colony were determined to be homozygous for a recessive mutation, termed “obese” (ob) (76). These ob/ob mice appear normal at birth, but soon begin gaining excess fat mass and displaying hyperglycemia and hyperinsulinemia (77). In the 1950s, ob/ob mice and their non-obese littermates underwent parabiosis experiments, where two animals are surgically joined (usually by peritoneum or a long bone of the leg) to allow for the exchange of whole blood between them (78). Weight gain was inhibited in ob/ob mice parabiosed with non-obese littermates, providing evidence that the ob/ob gene product was a circulating factor transferred from the blood of the lean littermate. In 1972, a similar study demonstrated that parabiosis with lean animals not only reduced weight gain in ob/ob mice, but also improved hyperglycemia, hyperinsulinemia, and insulin sensitivity (6). Finally, in the 1990s, positional cloning studies identified the product of the ob gene, dubbed leptin, which was derived from the Greek word “leptos” meaning to be thin. Further characterization of leptin revealed that adipocytes were its predominant source (5). Following this discovery, the first directly observed function of leptin was its effect on food intake (79) followed shortly thereafter by demonstration that leptin levels in mice and men strongly correlate with fat mass and play a key role in body-weight (energy) homeostasis as described below. 

 

Leptin Receptor and Signaling

 

Another spontaneously arising mutant mouse that developed obesity and type 2 diabetes is the diabetes or db/dbmouse (80). In contrast to ob/ob mice, parabiosis of db/db mice to wild type littermates did not improve body weight or diabetes, but instead resulted in unhealthy weight loss in the lean littermates, leading investigators to deduce that mice with the db mutation lacked a functioning receptor for the ob gene but still manufactured a circulating protein that crossed over to the lean littermates to induce anorexia (81). Further confirmation of these hypotheses came when parabiosis of ob/ob with db/db mice induced weight loss in the ob/ob mouse while the obese state was preserved in the db/db mouse (81–83). Although the db gene was cloned in 1990 (84), it was not until almost 5 years later (85)following the identification of leptin that the db gene was identified to encode the leptin receptor (LR).

 

The LR is a class 1 cytokine receptor with substantial homology to glycoprotein 130, a plasma membrane receptor that mediates the actions of many cytokines. Unlike other plasma membrane receptors, such as the insulin receptor, the LR lacks intrinsic kinase activity and signals via Janus kinases (JAKs). Six LR isoforms exist, designated LRa-LRf, with LRb being the best characterized. It is the longest LR isoform that is capable of full signaling via the JAK/STAT pathway (86).

 

Leptin-regulated circuits involved in energy homeostasis have been mapped to distinct yet diverse brain regions (87)expressing the long form of the LR (88). Increased central leptin signaling inhibits food intake and elevates energy expenditure, while leptin deficiency (such as during fasting or starvation) has opposite effects.  Expression of the LR has also been detected in peripheral tissues, but the exclusivity of the central leptin circuits to modulate energy intake and expenditure is supported by studies showing that deletion of the long form LR in peripheral tissues had no effects on these processes (89). Leptin levels strongly correlate with fat mass in mice and men (90,91), and as such leptin acts as a sensor of energy stores signaling the availability of body fat to the brain and regulating adipose reserves. However, during obesity the negative feedback loop between increasing leptin levels that signal high energy availability and inhibit food intake becomes disrupted due to the development of leptin resistance (92) — the inability to respond to leptin despite having sufficient or excess levels in circulation during accumulation of excess adipose stores. Although the physiological causes of leptin resistance are not well understood, it has been shown that hyperleptinemia is required for the development of leptin resistance during obesity. When leptin levels of mice are clamped to low levels (similar to lean mice), these clamped mice still develop obesity on HFD, but they do not become leptin resistant (93). The inability to overcome leptin resistance by giving supplemental doses has precluded leptin’s use as an anti-obesity therapeutic. Interestingly, leptin resistance that accompanies obesity appears to result from selective impairment of leptin’s ability to reduce food intake, while preserving its other capacity to raise energy expenditure (94). The molecular basis for this phenomenon has not yet been elucidated and remains under active investigation. 

 

Leptin also has central nervous system effects not directly related to energy balance, including modulation of reproduction and thermoregulation. Additionally, research into the role of leptin to mediate anxiety and depression is currently ongoing (95). Leptin can also act peripherally on hepatocytes and pancreatic β-cells to regulate glucose and lipid metabolism independently of its central effects (96). Leptin has also been shown to affect innate and adaptive immunity (7), bone formation (97), bone metabolism (98,99), angiogenesis, and wound healing (88). Skeletal muscle, liver, and intestines have been described as targets for leptin action (100), and some evidence suggests that leptin may also act in an autocrine manner on AT (101). How leptin mediates responses in peripheral tissues is poorly understood and complicated by the existence of its six receptor isoforms, their differential expression across tissues, the pleiotropic nature of leptin’s effects, the demonstration of “selective” or tissue-specific leptin resistance, and the complexity of the signaling pathways involved.  

 

Leptin and Cancer

 

Given the elevated risk for many cancers in patients with obesity in whom leptin levels are also high, it is not surprising that leptin has been implicated in tumorigenesis. Indeed, leptin levels or leptin signaling has been found to be dysregulated in breast, thyroid, endometrial, and gastrointestinal malignancies (102). Ectopic leptin expression in colorectal adenomas increases during the progression to colorectal cancer (103,104) yet associates with a favorable prognosis of the cancer (103). In papillary thyroid cancer, increased circulating leptin levels occur independently of body mass index (BMI), coincide with elevated LR expression on the tumor cells, and associate with aggressive carcinogenesis and poor prognosis (105,106). In contrast, reported associations between leptin levels and endometrial cancer are not maintained when adjusted for BMI, suggesting that leptin is not likely a causative factor in the development of this cancer (107–109).

 

Studies show that postmenopausal women with obesity have a 20-40% greater risk of developing breast cancer compared to normal weight women (110). In breast cancer, particularly in high-grade tumors, overexpression of both leptin and LR is associated with cancer progression and poor patient survival. Leptin’s ability to stimulate angiogenesis, regulate endothelial cell proliferation, and crosstalk with insulin and human epidermal growth factor receptor 2 (HER2) signaling pathways represent a few of the possible mechanisms by which leptin plays a role in breast cancer (111). As obesity rates continue to rise, it is likely that studies examining the relationship between leptin and cancer will become even more relevant. 

 

ADIPONECTIN

 

Adiponectin is a unique and extensively studied adipocyte-derived hormone with complex biology. Efforts to identify genes regulated during adipogenesis led to the discovery of adiponectin in 1995 (112) and 1996 by three separate research groups employing different approaches (113–115). Secreted by adipocytes, adiponectin is characterized by its remarkably high circulating levels reaching plasma concentrations in humans of 2-20 ug/ml (116), values that are more than 1000-fold higher than most other secreted factors (Figure 7). Unlike leptin, adiponectin levels decease as a function of increasing fat mass in both rodents and humans with obesity (115); thus, they are lower in patients with obesity than those who are lean. Adiponectin’s widely reported anti-hyperglycemic, anti-atherogenic, and anti-inflammatory effects have made it an attractive therapeutic target for the treatment of obesity and insulin resistance. However, efforts to develop therapies targeting adiponectin function have been impeded by its complex structure and regulation (117). 

 

Figure 7. Typical circulating concentrations of select adipokines and insulin for normal weight, healthy humans. Adipokine levels in the blood are several orders of magnitude higher than that of insulin.

Adipocytes secrete different forms of adiponectin: low-molecular weight (LMW) trimers (the most basic form), medium-molecular weight (MMW) hexamers, and high-molecular weight (HMW) oligomers (118), as well as globular adiponectin, a proteolytic fragment of the protein (119,120). In humans, the MMW and HMW oligomers make up most of the circulating adiponectin while the LMW trimer constitutes less than 30% of serum adiponectin. The HMW oligomer is most closely associated with enhanced insulin sensitivity and reduced glucose levels (121).

 

Adiponectin signaling is complex and incompletely understood. Three adiponectin receptors have been identified. Adiponectin receptor-1 and -2, referred to as AdipoR1 and AdipoR2, bind the LMW and globular forms (122). T-cadherin binds HMW adiponectin (123). Both AdipoR1 and AdipoR2 can modulate insulin sensitivity and metabolic gene expression in insulin-responsive tissues, and both receptors have demonstrated roles in the pathophysiology of insulin resistance and T2D (124,125). T-cadherin, which is expressed in a variety of tissues including the liver (126), belongs to a family of cell surface proteins involved in cell-cell interactions (127). Mice lacking T-cadherin accumulate adiponectin in circulation and have a similar cardiovascular phenotype to adiponectin knockout mice (128), suggesting that T-cadherin is the primary effector of cardioprotection by adiponectin (129–131).

 

Adiponectin enhances fatty acid oxidation through activation of AMP-activated kinase (AMPK) (132,133), a cellular energy sensor, which then inhibits acetyl CoA carboxylase, a rate limiting enzyme in DNL (134). This, in turn, reduces malonyl-CoA production and enhances fatty acid oxidation. Adiponectin can activate AMPK through two independent pathways, and can also modulate lipid metabolism by increasing mitochondrial density and mitochondrial DNA content (135,136). Adiponectin has diverse effects in many tissues, including bone and cartilage (137), and can act in an autocrine or paracrine manner in AT and other tissues (138). Adiponectin also appears to modulate a wide range of biological processes, including reproduction and embryonic development (139,140). The heart, liver, and skeletal muscle are considered the primary targets for adiponectin action, and adiponectin’s prominent insulin-sensitizing effects have been most fully characterized at the mechanistic level in liver and muscle (117) (Figure 6).

 

The liver performs a critical function in maintaining normal blood glucose levels by releasing glucose (i.e. hepatic glucose output) into circulation in conditions such as fasting, exercise, and pregnancy. Conversely, the ability of the liver to reduce its glucose output when demand is low, as in the fed state, is also crucial to preventing hyperglycemia, and this process is often impaired with obesity and insulin resistance. Adiponectin can robustly reduce plasma glucose levels predominantly by inhibition of hepatic glucose production as opposed to effects on whole-body glucose uptake into cells and glycolysis (141). The importance of adiponectin in regulating glucose output in the liver is underscored by studies showing that mouse models with genetic deletion (142) or overexpression (143) of adiponectin have impaired or enhanced hepatic insulin sensitivity, respectively. Adiponectin levels are increased bythiazolidinediones (TZDs), which is thought to be the predominant mechanism of action that improves insulin sensitivity and glucose tolerance with this class of medications (142–144). Adiponectin also promotes hepatocyte survival, inhibits hepatic fibrosis and inflammation, stimulates fatty acid oxidation (133,145) and modulates fatty acid uptake and metabolism (146). In patients with nonalcoholic fatty liver disease (NAFLD) who are insulin resistant, low plasma adiponectin levels are associated with the progression of NAFLD and non-alcoholic steatohepatitis (147,148). In summary, adiponectin has beneficial effects in the liver, where it protects against metabolic dysfunction and hepatic diseases (Figure 6).

 

Skeletal muscle is responsible for up to 80% of insulin-mediated glucose uptake in healthy individuals (149,150). Adiponectin can promote glucose uptake (151,152), enhance fatty acid oxidation (152,153), and enhance insulin sensitivity (154) in cultured muscle cell lines and mouse skeletal muscle. Adiponectin administration to obese, insulin-resistant adiponectin-knockout mice improves skeletal muscle insulin sensitivity (146,155,156). In human myotubes, adiponectin promotes fat oxidation via AMPK activation; this response is impaired in myotubes from patients with T2D and obesity (157). Thus, adiponectin has an important role in skeletal muscle metabolism in humans as well as rodents, and defective adiponectin signaling in skeletal muscle may contribute to insulin resistance.

 

Finally, in addition to its insulin-sensitizing and glucose-lowering effects in liver and skeletal muscle, adiponectin is also cardioprotective. Low circulating adiponectin levels correlate significantly and independently with coronary artery disease (158), and are considered a risk factor for cardiovascular diseases (CVD) such as hypertension, coronary artery disease, and restenosis (159). The vascular endothelium is believed to mediate some of the cardioprotective effects of adiponectin via AMPK activation and subsequent activation of eNOS (endothelial nitric oxide synthase) (160).

 

In light of these beneficial functions, adiponectin has significant therapeutic potential in the treatment of T2D, CVD, and NAFLD. Several years ago, small molecule screening efforts produced the first small molecule AdipoR agonist.  “AdipoRon”, as it was named, not only recapitulated adiponectin’s effects on AdipoR signaling pathways but also had profound anti-hyperglycemia effects in both diet-induced obese mice and a genetic mouse model for diabetes (161). A more recent study has now shown that AdipoRon can also decrease ceramides and lipotoxicity, and mitigate diabetic nephropathy (162). Hence, small molecule activators of adiponectin signaling show promise in the management of obesity-associated metabolic diseases like insulin resistance, NAFLD, and T2D.

 

RESISTIN

 

Resistin, the most recently discovered of the major adipocyte-derived hormones, was independently identified by two laboratories. In one case, the gene coding for this novel endocrine factor was identified in a screen for genes inhibited by TZD drugs and was named “resistin” because it induced insulin resistance (163). Another group identified the same gene in a screen for genes expressed exclusively in adipocytes and induced during adipogenesis; they named it ADSF, for adipose tissue-specific secretory factor. In this study, the product of the gene was shown to inhibit differentiation of adipocytes in vitro (164), later confirmed in a separate study (165).

 

Elucidating resistin’s role in physiology has been challenging. While resistin is expressed in both white and brown fat in mice, the various WAT depots (inguinal, gonadal, retroperitoneal, and mesenteric) and BAT exhibit distinct patterns of resistin expression (166). In addition, circulating levels of resistin are directly proportional to its gene expression in some conditions, but inversely proportional in others (167,168). Remarkably, while resistin produces similar metabolic and inflammatory effects in humans and mice, human resistin is predominantly secreted from macrophages, not adipocytes (169–171). The complex regulation of resistin expression and the fundamental differences in resistin biology between species are significant obstacles to fully understanding this hormone’s functions and mechanisms of action in humans.

 

Resistin interacts with two known receptors: the toll-like receptor 4 (TLR4) and adenylyl cyclase-associated protein 1 (CAP1) (172,173). Resistin signaling through TLR4 contributes to monocyte recruitment and chemokine expression, and is involved in inflammatory responses in atherosclerosis and acute lung injury (135,174). Both knockout and overexpression studies of CAP1 indicate that this receptor can also mediate proinflammatory effects of resistin (173).  Overall, the similarities, differences, and tissue specificity of resistin signaling through TLR4 versus CAP1 remains poorly understood.

 

Resistin has also been shown to regulate fasting blood glucose levels in mice (175). Elevated levels of circulating resistin are reported in genetic and diet-induced mouse models of obesity (163). Anti-resistin antibody administration improves insulin sensitivity in diet-induced obese mice, and conversely, resistin injection impairs glucose tolerance in normal mice; supporting a causative role of resistin in mediating insulin resistance in mouse models (176). Moreover, both human and mouse resistin have been shown to impair insulin-stimulated glucose uptake in cultured murine myocytes in vitro (177). Other studies have shown similar insulin desensitizing effects of resistin in liver and brain(178,179).

 

The finding that human resistin originates not in adipocytes but in mononuclear lymphocytes raised the possibility that the hormone may have distinct roles in the two species. An elegant mouse model was generated to address this issue, the so-called humanized resistin mouse.  In these mice, the endogenous resistin gene (normally expressed in adipocytes) was deleted, and the macrophage-expressed human resistin gene was inserted (180). Data from this study revealed that like murine adipocyte-derived resistin, the humanized resistin induced systemic insulin resistance, adipose tissue inflammation, and elevated circulating free fatty acids in high-fat diet (HFD)-fed mice.

 

In humans, epidemiological, genetic, and clinical data support a role for resistin in dysfunctional metabolism and related pathologies (181). As in mouse models, serum resistin levels are elevated during human obesity (182,183). Furthermore, high circulating resistin concentrations in humans have been associated with atherosclerosis, coronary heart disease, congestive heart failure, as well as inflammatory conditions including systemic lupus erythematosus, inflammatory bowel disease, and rheumatoid arthritis (184–188). Whether the relationship between resistin and insulin resistant states is merely correlative and whether interventions to antagonize resistin action will be of therapeutic value in the treatment of metabolic or cardiovascular disease in humans remains undetermined.

 

Cell Types in Adipose Tissue

 

Besides adipocytes, AT is comprised of endothelial cells, blood cells, fibroblasts, pericytes, preadipocytes, macrophages, and several types of immune cells (189). These non-adipocyte cell types are commonly referred to as the AT stromal vascular fraction (SVF) (Figure 8). Our understanding of the complexity of the cell types present in the SVF and how this milieu is altered by metabolic disease states is an area of active investigation. Cells in the SVF produce hormones and cytokines that can act in a paracrine manner on adjacent adipocytes. In the early 1990s, it was shown that TNF alpha production was increased in AT during metabolic disease states, in particular, T2D (190).Yet, it wasn’t for another ten years that adipose tissue macrophages (ATMs) were identified as the primary cellular source of AT TNF alpha (191). It is now largely accepted that in conditions of obesity and T2D, TNF alpha is produced in ATMs and acts on adjacent adipocytes within AT to promote insulin resistance. Hence, it is important to consider the presence and dynamic interactions of the SVF cells, especially when determining the cellular sources of AT-derived paracrine and endocrine hormones.

 

Figure 8. Constituents of adipose tissue (AT). Left: Along with mature, functional adipocytes and precursor cells, many cell types related to vasculature and immune function reside within AT. They perform both physiological and pathophysiological functions by communicating with the adipocytes via secreted factors and scavenging lipid from dying fat cells. The number and diversity of these cell types increases with developing obesity and metabolic dysfunction. Right: The non-adipocyte cells are collectively referred to as the stromal vascular fraction (SVF), and the SVF can be separated from lipid-containing adipocytes by digesting the extracellular matrix (ECM) and centrifuging the cellular mixture. The SVF will form a pellet at the bottom of the tube, while the adipocytes will float and form a visible lipid layer at the top of the aqueous medium. This separation technique is critical to studying the cellular composition of adipose tissue and gaining insight regarding the individual functions of these diverse and distinct cell types under physiological and pathophysiological conditions.

Adipogenesis

 

To understand how adipocytes contribute to systemic metabolic regulation, it is important to understand their development. Adipogenesis refers to the process by which precursor cells differentiate and become committed to storing lipid and maintaining energy homeostasis as adipocytes. Adipogenesis is regulated by hundreds of factors, including nutrients, cellular signaling pathways, miRNAs, cytoskeletal proteins, and endocrine hormones such as growth hormone, insulin-like growth factor 1, insulin, and several steroid hormones, as well as cytokines. Generally, pro-inflammatory cytokines inhibit adipogenesis (192), although some cytokines within the same family exert opposing effects (192). Cytoskeletal proteins (193), ECM proteins and their regulators (194), microRNAs (miRNAs) (195), and long noncoding RNAs (lncRNAs) (196) differentially modulate adipogenesis. Dozens of different transcription factors, briefly described below, also regulate adipogenesis (Figure 9).

Figure 9. Transcriptional regulation of adipogenesis as determined in vitro in a fibroblast-like preadipocyte clonal cell line. Preadipocytes are grown to confluence and become growth arrested. Following induction of differentiation, they re-enter the cell cycle and undergo several rounds of proliferation, a process known as mitotic clonal expansion. At the end of this short proliferative phase, preadipocytes terminally differentiate into adipocytes as they begin synthesizing lipid and assume characteristics of mature fat cells. Numerous transcription factors have been determined to promote (green arrows) or inhibit (orange horizontal ended line) adipogenesis either during clonal expansion or at later stages of terminal differentiation. The timing of activation (i.e. when each transcription factor is turned on and off) is critical to the progression of adipocyte differentiation.

 

PROMOTORS OF ADIPOGENESIS

 

The transcription factor peroxisome proliferator activated receptor gamma (PPARg) is considered the principal adipogenesis regulator (197). Its discovery substantially enhanced our understanding of the adipocyte and its role in metabolic disease. For example, mice with adipocyte-specific PPARg deletion have decreased AT mass and are insulin resistant (198). In humans, PPARg gene mutations can also cause lipodystrophy (partial or generalized loss of fat in the body) and severe insulin resistance (199–201). The discovery of PPARg as the functional receptor for the insulin-sensitizing TZDs resulted in a significant effort to understand PPARg action and identify additional agonists.Synthetic TZDs induce weight gain in humans and rodents by increasing fat mass, more so in the subcutaneous adipose depot, which is associated with improved metabolic outcomes. However, this weight gain is also considered as a negative side effect of TZD treatment, especially in the typical patient who has pre-existing obesity. Other adverse side effects of TZDs, such as bone fractures and heart failure, have spawned the search for structurally distinct PPARg ligands capable of inducing unique receptor-ligand conformations with signature affinities for diverse co-regulators (202). Several selective PPARg modulators (SPPARMs) with fewer side effects have been identified. These act as partial PPARg agonists, alter specific post-translational modifications of PPARg, and preserve anti-hyperglycemia effects while minimizing or eliminating the adipogenic effect that leads to increased fat mass via activation of distinct gene profiles that may be cell and tissue specific (203,204). Interestingly, the TZD, rosiglitazone, is capable of improving glucose homeostasis even in the absence of PPARg in mature adipocytes (205), suggesting that its adipogenic effects (in addition to its non-adipogenic ones) may also be important for its anti-hyperglycemic action.

 

In addition to TZDs, PPARg binds endogenous lipophilic molecules, including: long chain fatty acids (LCFAs), oxidized or nitrated FAs, prostaglandins, and arachidonic acid derivatives (206). Interestingly, serotonin (5-hydroxytryptamine, 5-HT) has also been shown to be a high affinity agonist for PPARg (207). Many of the endogenous PPARg ligands enhance adipocyte differentiation and regulate fat cell functions such as lipolysis, glucose uptake, and lipogenesis through PPARg-dependent and independent methods (208–214). Overall, these endogenous ligands have low affinity and limited subtype selectivity for PPARg relative to other PPARs, suggesting that much remains to be understood regarding this critical adipogenesis regulator. While there is no question that PPARg is essential for adipogenesis and lipid accumulation within fat cells, a better mapping of its gene expression profiles in discrete cell and tissue types and with endogenous and synthetic ligands will improve our understanding of AT development and function under both physiological and pathophysiological conditions.   

 

The CCAAT/enhancer-binding proteins (C/EBPs) are widely expressed transcription factors that regulate proliferation and differentiation of various cell types in mammals. Studies in vivo and in vitro have identified C/EBP isoforms α, β and δ as important regulators of adipogenesis (215). C/EBPs β and δ work together in early adipogenesis to promote fat cell differentiation by inducing expression of C/EBPα and PPARg (216). Additionally, the transcription factors Krox20 and ZNF638 can modulate adipogenesis by affecting C/EBPβ function (217,218).

 

The Signal Transducer and Activator of Transcription (STAT) family of transcription factors was first identified over 20 years ago (219). Both the protein expression of STATs and their ability to regulate gene expression are tissue-specific (220). In AT, STATs regulate gene expression during adipogenesis, and the expression of STATs 1, 3, 5A, and 5B is induced during differentiation of murine and human preadipocytes (221,222). Notably, the ability of STAT5 proteins to promote adipogenesis has been documented by over a dozen independent laboratories using both in vitro and in vivoapproaches (17).

 

Of the three isoforms of Sterol Response Element Binding Proteins (SREBP-1a, SREBP-1c, and SREBP-2), SREBP-1c is the predominant form expressed in white AT (223,224) and is an important regulator of lipogenesis genes, while SREBP-2 regulates the expression of cholesterol biosynthesis genes (225). Intriguingly, two miRNAs (miR-33a and miR-33b) located within the SREBP genes are highly induced during adipogenesis (226). Although SREBP-1 clearly plays a promoting role in adipogenesis in vitro, in vivo studies suggest that SREBP-1 is not critical for AT development and/or expansion, perhaps due to compensatory SREBP-2 overexpression (47,227).

 

Members of the early B-cell factor (EBF) family of transcription factors are characterized for their ability to modulate islet beta-cell maturation and neural development. Three primary members of this family (EBFs 1, 2, and 3) are expressed in fat cells. EBFs 1 and 2 can promote adipogenesis (228,229), and EBF2 can also play roles in determining brown versus white adipocyte identity in vivo (230) and the beiging process of adipose tissue in mice (231).  

 

INHIBITORS OF ADIPOGENESIS

 

The interferon-regulatory factor (IRF) family of transcription factors has functionally diverse roles in the immune system, but also plays a role in adipocyte development. All nine IRF family members are regulated to different degrees during adipogenesis in vitro, and some members can repress adipogenesis (232) and contribute to insulin resistance (233). For example, knockdown of IRF3, whose is expression is elevated in visceral and subcutaneous AT of obese mice as well as in subcutaneous AT from humans with obesity and diabetes decreases fat mass and prevents insulin resistance in high fat diet-fed mice (233).

 

Wingless-related integration site (Wnt) proteins regulate development and cell fate through both autocrine and paracrine signaling (234) by using three well-characterized pathways: the canonical Wnt signaling and the planar cell polarity and Wnt/calcium pathways, which are non-canonical. The canonical pathway is dependent upon the transcription factor, β-catenin (235). Wnt10b is the best studied member of the Wnt signaling family in terms of adipocyte development. In the presence of Wnt10b, β-catenin translocates to the nucleus where it inhibits PPARγ and C/EBPα activity, thereby impeding adipogenesis (236,237). On the other hand, extracellular antagonists of Wnt/β-catenin signaling have been reported to promote adipocyte differentiation (238,239). 

 

The GATA family of transcription factors were named based on their ability to bind the DNA sequence GATA (240). Only GATAs 2 and 3 are expressed in preadipocytes residing in white AT (241), and both are repressed during adipogenesis. In fact, GATA2 can directly bind to the PPARγ promoter to suppress its activity (241). In addition to inhibition of PPARγ expression, GATAs 2 and 3 can also associate with C/EBPs to disrupt their transcriptional activity (242). GATA3 expression is driven by the canonical Wnt signaling pathway (243,244). Collectively, these studies demonstrate that two GATA proteins can attenuate adipocyte development via multiple transcriptional and signaling pathways.

 

TRANSCRIPTION FACTOR FAMILIES THAT CAN EITHER PROMOTE OR INHIBIT ADIPOGENESIS

 

The Krüppel-like transcription factors (KLFs) include 17 members that can either activate or repress transcription. In relation to adipocyte development, KLFs 4, 5, 6, 9 and 15 can promote adipogenesis, while KLFs 2, 3 and 7 repress adipocyte development. Most studies on the roles of KLFs in adipogenesis have been performed in vitro using a variety of cell culture models, and have demonstrated that KLFs act in concert with other transcription factors modulate adipogenesis (245).

 

The transcription factor activator protein 1 (AP-1) consists of Jun proteins (c-Jun, JunB, and JunD), Fos proteins (c-Fos, FosB, Fra1, and Fra2), ATF and JDP family members, several of which are induced during adipogenesis (222). In humans, a mutation in the c-fos gene that is associated with lipodystrophy has been shown to reduce c-fos activity and adipocyte development (246). Many in vitro and in vivo studies demonstrate that, like KLFs, AP-1 transcription factors can positively and negatively regulate adipogenesis.

 

Many of the zinc finger proteins (ZFPs) function as transcription factors with several contributing to adipocyte determination and/or adipogenesis. Zfp423 and Zfp467 can promote adipocyte differentiation by enhancing PPARγ expression and activity (247,248). In addition to stimulating adipogenesis, Zfp423 can suppress ‘beige-like’ properties in white adipocytes that are typically associated with improved metabolic health (249). Zfp521 can inhibit Zfp423 to reduce adipocyte development and is also considered a critical regulator of the commitment to either osteogenic or adipogenic lineages (250,251).

 

The transforming growth factor beta (TGF-β) superfamily encompasses a large number of proteins, including bone morphogenetic proteins (BMPs) (252). BMPs and TGF-β have been reported to be involved in both adipocyte commitment and differentiation (253–255). Specifically, BMPs 2 and BMP4 can promote adipogenesis via the Smad signaling pathway (256) to regulate transcription of target genes such as PPARγ (257,258). While BMPs are known to promote adipogenesis, in vitro and in vivo studies demonstrated that TGF-β primarily inhibits fat cell differentiation.

 

HORMONAL REGULATION OF ADIPOGENESIS

 

Steroids are prominent regulators of AT development and distribution, and adipocytes express high levels of many steroid hormone receptors. These lipophilic hormones diffuse through plasma membranes, dimerize, and bind to their specific receptors to impart both genomic and non-genomic responses (259,260). Since steroid-bound receptors act as transcription factors, their capabilities should be fully considered in the transcriptional regulation of adipogenesis.

 

Two types of estrogen receptors, ERα and ERβ, are expressed in rat and human preadipocytes, mature adipocytes, and in other AT cells (261–263). Although many studies describing the role of estrogens in AT are contradictory, most investigations indicate that estrogen inhibits adipocyte differentiation (245) and the adipogenic action of PPARγ (264). Aromatase is an enzyme found in several tissues, including AT, that aromatizes androgens into estrogens. Both ERα-and aromatase-knockout mice have increased adiposity, suggesting that both estrogen and its receptor can reduce adipocyte development (265,266). Mice lacking ERα have enhanced visceral AT deposition and increased weight gain compared with wild-type mice (267).

 

Androgen receptors (AR) are also expressed in rodent (268,269) and human AT (270).  Similar to estrogen, many studies report contradictory actions of androgens on the differentiation and function of adipocytes. These inconsistent results highlight the importance of accounting for sex-, depot- and organism-specific effects. In studies of human AT, testosterone and the non-aromatizable androgen, dihydrotestosterone, inhibit differentiation of preadipocytes obtained from subcutaneous and omental depots of both men and women, although the magnitude of the inhibitory effect may differ between the sexes  (271,272). Overall, most studies indicate that androgens exert inhibitory effects on adipogenesis.  

 

Glucocorticoids (GCs) are well-known promoters of adipocyte development. GCs also promote adipocyte hypertrophy and differentiation of central fat depots that can lead to abdominal obesity and insulin resistance (273). In vitroadipogenesis studies include the wide use of the synthetic GC, dexamethasone. Although the mechanisms of action and target genes of GCs involved in adipocyte differentiation are not completely clear, it is known that GCs induce expression of C/EBPs beta  and δ and that GC-induced C/EBPδ coordinates with C/EBPβ to induce PPARg expression and adipogenesis (274).

 

To understand the actions of GCs via the glucocorticoid receptor (GR), it is important consider the enzyme that affects circulating levels of cortisol, the active form of GR’s endogenous ligand. 11β-hydroxysteroid dehydrogenase type 1 (11 beta HSD1) is an enzyme highly expressed in AT and liver that in AT converts inactive cortisone to the active hormone cortisol. Hence, it is not surprising that 11 beta HSD1 mRNA expression and activity is essential for the induction of human adipogenesis and that adipocyte development can be blocked with a 11 beta HSD1 specific inhibitor (275). In addition to inducing the expression of early adipogenic transcription factors, GCs promote adipocyte development by mechanisms that include suppression of anti-adipogenic factors (Pref-1 and Runx2); anti-proliferative effects on preadipocytes; and sensitizing or ‘priming’ of human preadipocytes to insulin action (276). Recent attention has focused on the potential contributions of environmental pollutants known as endocrine disrupting chemicals (EDCs) in the development of metabolic diseases. Studies reveal that EDCs can promote adipogenesis through GR activation (277), thereby implicating these compounds in the rising rates of obesity and diabetes.

 

In addition to regulating water and salt homeostasis, the mineralocorticoid aldosterone and its receptor (MR) have also been shown to play a role in the regulation of adipocyte development. This is important since MR is a high-affinity receptor for both mineralocorticoids and GCs. Aldosterone promotes adipogenesis in an MR-dependent manner (278)and a MR antagonist can inhibit adipogenesis (279). Although GRs and MRs are expressed in AT and thought to mediate cortisol’s actions on AT, the levels of GR are several hundred-fold higher than MR in both human preadipocytes and adipocytes (280). Loss of GR, but not MR, blocks the adipogenic capabilities of cortisol in human preadipocytes (280). However, MR expression is higher in omental than in subcutaneous AT, so there could potentially be depot differences in the relative importance of MR and GR in cortisol-induced adipogenesis (280). There could also be differences in the contribution of MR to adipogenesis during obesity when MR and 11 beta HSD1 expression levels are increased, while the GR and 11 beta HSD2 (the enzyme that deactivates cortisol) levels do not increase accordingly (280). Most of the current evidence suggests that the ability of aldosterone to modulate adipogenesis in vitro is largely dependent on MR. Additional studies are needed to determine if MR plays a role in adipocyte development in vivo.  

 

Vitamin D is another steroid hormone with strong experimental evidence that it can regulate adipogenesis. Unlike most of the water-soluble vitamins that are excreted via urine when in excess, Vitamin D, along with the other fat-soluble vitamins (A, E, and K), can be stored within fat-laden adipose tissue. The vitamin D receptor (VDR) and 1α-hydroxylase (CYP27B1), the enzyme that activates vitamin D, are expressed in human AT, primary preadipocytes, and newly-differentiated adipocytes (281). The most active form of Vitamin D, 1, 25-Dihydroxyvitamin D, represses adipocyte differentiation (282,283) and the VDR can block adipogenesis by inhibiting C/EBPβ expression (284). Vitamin D-induced inhibition of adipogenesis also involves direct suppression of C/EBPα and PPARg (285). Vitamin D and VDR also play a role in the inhibition of adipogenesis of bone marrow stromal cells (286), in part by suppressing the expression of inhibitors of the canonical Wnt/β-catenin signaling pathway (287). Although vitamin D inhibits adipogenesis in the widely used murine and bone marrow-derived cells, both 25-hydroxyvitamin D and 1,25-dihydroxyvitamin D3 can promote the differentiation of human subcutaneous preadipocytes (281). Overall, a case could be made that concentrations of vitamin D as well as the type of adipocyte precursor determine whether this hormone exerts pro- or anti-adipogenic actions via the VDR.

 

On the other hand, evidence regarding Vitamin D’s role in adipocyte development in humans is controversial and contradictory. According to a systematic review and meta-analysis of 23 studies between 2002 and 2014, overweight or obese subjects exhibit a higher prevalence of Vitamin D deficiency (288). In two double-blind, placebo-controlled randomized clinical trials, Vitamin D-supplemented individuals with healthy overweight or obesity lost significantly more fat mass than the placebo group when fed either a calorie-restriction (289) or weight-maintenance (290) diet for 12 weeks. While decreased fat mass may result from Vitamin-D induced inhibition of adipogenesis, this hypothesis was not directly tested in the studies, and two other longer term studies demonstrated no change in fat mass with Vitamin D supplementation between 14,000 and 20,000 IU per week (291,292).  

 

The relationship between adipocyte development and thyroid hormones has been recognized since 1888 when a report on myxedema proposed that obesity was a requirement for a diagnosis of hypothyroidism (293). The most biologically active form of thyroid hormone, T3, can induce brown adipocyte differentiation (294). Hyperthyroidism in rodents induces adipocyte hyperplasia, whereas hypothyroidism impedes AT development (295). Overall, studies on the involvement of thyroid hormones in AT development are controversial. While the induction of adipogenesis is differentially regulated by various thyroid hormone receptor (TR) isoforms, studies largely indicate that TRs promote adipogenesis in the majority of model systems (245).   

 

Adipocyte Progenitors

 

In AT, pools of adipose stem/progenitor cells (APs) exist that can differentiate into mature adipocytes (296,297). At least two distinct progenitor populations give rise to adipocytes:  developmental APs and adult APs (296,298). Our understanding of the molecular characteristics of APs has dramatically increased in recent years as discussed below.

 

APS IN ADIPOSE DEVELOPMENT

 

AT organogenesis in mice and humans begins during embryogenesis, and ends in the postnatal period for mice and just before birth in humans (296,297,299). AT is widely accepted to be of mesodermal origin (297). However, some of the spatiotemporal and molecular differences observed in formation of different AT depots suggest diverse developmental origins (297,300). Further, white and brown adipocytes, once considered to have common APs are now known to have different origins (297,301,302).

 

In the generation of white adipocytes, developmental APs express the master adipogenesis regulator PPARg but have distinct functional and molecular properties compared to adult APs (298,302). Developmental APs do not contain lipid but express the mature adipocyte markers perilipin and adiponectin, are able to replicate, and are located along the vasculature in developing white adipose tissue (298,302,303). Brown adipocytes can arise from myogenic Myf5-expressing precursors that also give rise to skeletal myocytes (297,302,304). Interestingly, brown-like adipocytes, known as beige adipocytes, emerge in white adipose from Myf5-negative precursors in response to cold or adrenergic stimuli, which suggests that the developmental origins of brown adipocytes and beige adipocytes are different (297,304). Collectively, these findings highlight the complex developmental heterogeneity of APs observed among adipose tissue depots in animals and humans.

 

APS IN ADULT ADIPOSE TISSUE

 

The notion that we are born with all the fat cells we will ever have is now considered archaic and inaccurate. Adipose tissue continues to generate new adipocytes throughout the lifespan, with a median adipocyte turnover rate of 8.3 years (302,305). Adult APs have been found in the SVF of AT depots in both rodents and humans (302,306–308) and are thought to represent an AP pool that contributes to this adipocyte renewal. Flow cytometry techniques that use a variety of cell surface and stem cell markers, have helped identify stromal cells that can undergo adipogenesis (302,306,307,309). These adult APs arise from tissue-resident mesenchymal stem cells, and are a major source of new adipocytes in AT (297,310). Bone marrow-derived APs from the myeloid lineage can also be recruited to AT where they become adipocytes (Figure 10). Bone marrow-derived adipocytes (BMDAs) are more abundant in female mice and are more frequent in visceral depots (297,311,312). Though BMDAs have been observed in human AT and are increased in patients with obesity, the processes and factors involved in BMDA recruitment to AT remain unclear. Compared to normal adipocytes, BMDAs have reduced expression of lipid metabolism genes and increased pro-inflammatory gene expression, suggesting that they may have negative metabolic effects (297,311,313).

 

Figure 10. Adipocytes are derived from both resident mesenchymal cells in the stromal-vascular fraction of adipose tissue and hematopoietic progenitors that reside in the bone marrow. In addition to adipocytes, mesenchymal progenitors can form other connective tissue cells, such as myocytes and osteocytes. Myeloid progenitors derived from hematopoietic progenitors in bone marrow give rise to adipocytes as well as neutrophils, macrophages, dendritic cells, and granulocytes.

Most of the information regarding AP proliferation in obesity comes from rodent models. In mice fed high-fat diets to induce obesity, APs form new adipocytes primarily in the visceral depot (299,302). Although limited data report decreased AP proliferation and differentiation capacity from humans with obesity compared to lean individuals (314), convincing evidence for depot-specific AP populations in humans has emerged. Subcutaneous APs were shown to have a higher growth rate and adipogenic potential than visceral APs, giving rise to more functional adipocytes (315,316). Increasingly sophisticated methods for assessing APs in mice will help facilitate the identification of the origins of all APs for each adipose depot as well as the niches in which they reside.

 

Adipose Extracellular Matrix: From Normal Development to Fibrosis

 

An underappreciated influence on AT physiology is the adipose extracellular matrix (ECM). The dynamics and composition of the ECM are critical for proper adipocyte development and function (317). During adipogenesis, there is increased synthesis of laminar ECM constituents and maintenance of peri-adipocyte fibrillar collagens that ultimately allows the adipocyte to embed itself in the basal lamina (317). In the growth phase of adipogenesis, adipocytes require ECM-mediated traction forces to properly accumulate lipids and increase in size. A number of inhibitors, enzymes, and modifiers contribute to adipocyte ECM maintenance and renewal; these reactions consume a large amount of energy in the mature fat cell (317). In obesity, the ECM expands to accommodate the adipocyte hypertrophy and hyperplasia, and subsequent tissue growth, induced by the increased demand for lipid storage (317–321). This process appears to occur in a similar fashion in both animal models and humans.

Figure 11. Differences in AT between lean and obese mammals. The AT extracellular matrix (ECM) is important for normal tissue function but can also contribute to its dysfunction. In obesity, accumulation of ECM components can restrict AT expansion, promote inflammation by recruiting immune cells, and impair adipogenesis. These combined effects can worsen insulin resistance.

Adipose tissue expansion during obesity, coupled with immune cell accumulation and hypoxia, can lead to AT fibrosis (Figure 11) (317,319,322). Fibrosis is the excessive accumulation of ECM components, such as collagens, that typically results from an imbalance of the synthesis and degradation of ECM components (319,323). Ultimately, adipocyte dysfunction will result from the decreased ECM flexibility conferred by the accumulation of fibrillar ECM components (317,320,323). Abnormal ECM collagen deposition is associated with immune cell infiltration, which can worsen fibrosis and contribute to AT dysfunction that often occurs in obesity (319,323). The removal of collagen VI, a major AT ECM component, improves adipocyte function and metabolism in obese mice by both decreasing AT immune cell infiltration and “weakening” the ECM, which allows uninhibited adipocyte hypertrophy (317,318,323). In humans, AT collagen VI expression is increased in obesity, and subjects with higher collagen VI have increased macrophage content and AT inflammation (324). Endotrophin, an adipocyte-derived cleavage product of collagen VI, directly stimulates AT fibrosis and macrophage accumulation, and can lead to systemic insulin resistance (325). Endotrophin can also cause fibrosis and endothelial cell migration in mammary tumors, leading to tumor expansion and the enhancement of metastatic growth (325,326).

 

Accumulation of ECM components and increased ECM-receptor signaling are associated with insulin resistance in obesity thought to be mediated by several possible mechanisms. In addition to physically restricting AT expansion, excess ECM components can also increase AT inflammation by interactions with their cell surface receptors (CD44, CD36, and integrins) (320). These ECM-receptor interactions can induce adipocyte death, inhibit angiogenesis, and promote macrophage infiltration and inflammation in adipose tissue, thereby driving insulin resistance (320). Interestingly, these downstream effector pathways of ECM-receptor signaling are similar to those involved in tumor growth and pulmonary fibrosis development. 

 

The ECM has clear roles in the normal development and function of adipocytes, but in excess can also play roles in obesity development and metabolic dysfunction. Our understanding of the adipose ECM has deepened in recent years, but more research is necessary to better delineate how ECM components and their interactions can directly influence AT physiology and pathophysiology. Since many AT cell types produce ECM components, studies to determine the specific contributions of adipocyte-derived ECM components to normal AT function as well as dysfunction will be required.

 

Rodent versus Human Adipose Depots

 

Much of the knowledge about the depot-specific characteristics and metabolic profiles of AT has been obtained from rodents. However, the validity of translating studies conducted in rodent fat to humans remains controversial. Relative to humans, rodents have substantially more BAT and rely heavily on this highly-inducible depot to stimulate thermogenesis (327). While BAT activation in rodents has been shown to elicit beneficial effects, including improvements in glucose and lipid metabolism (328,329), BAT function in humans is more controversial. Overall, the majority of studies have reported that the amount of active BAT in humans appears insufficient to induce meaningful changes in energy metabolism and, thus, is not thought to impact whole-body physiology and metabolic control in humans (330) as described in rodents.

 

With regard to white AT, notable differences exist with respect to fat depot structure and function between species (18). Humans have subcutaneous depots primarily in the abdominal and gluteal-femoral regions; whereas rodents have subcutaneous fat pads located anteriorly and posteriorly (Figure 12). With regards to location, the inguinal (posterior) fat pad in rodents is considered comparable to the gluteal and femoral depots in humans. Human subcutaneous

abdominal AT can be categorized as superficial SAT or deep SAT (331), which are  morphologically and metabolically different. Deep SAT has been reported to be closely related  to the pathophysiology of obesity-related metabolic complications, while superficial SAT is more closely related to the protective lower-body SAT (332–334). However, these subcutaneous layers are not present in rodents. In humans, intra-abdominal fat refers to visceral AT, which surrounds the inner organs, and includes omental, mesenteric, retroperitoneal, gonadal, and pericardial depots (335). For most purposes, however, when used in reference to human studies, visceral AT refers to omental and mesenteric depots that are quantified by abdominal computed tomography or MRI scans. On the other hand, visceral fat pads in rodents are classified as perigonadal (epididymal in males and periovarian in females), retroperitoneal, and mesenteric. While the mesenteric fat pad is most analogous to abdominal (visceral) AT in humans, it is not often studied in rodents due to surgical limitations. The perigonadal fat pads are the largest and most the readily assessable fat in rodents; hence, they are most frequently used in mouse studies and cited the most often in the literature as surrogates for human visceral AT. However, humans do not have an AT depot analogous to the rodent perigonadal fat pads. In addition, the omental depot is clearly defined in humans, but in mice it is difficult to detect. Overall, striking anatomical differences in AT distribution exist between rodents and humans, and these differences should be considered when interpreting rodent studies and potentially translating these observations to humans.

 

Figure 12. Rodent versus Human AT depots. Several differences exist between rodent and human subcutaneous (SubQ) and visceral AT depots. In the figure SubQ depots are colored as beige, while visceral depots are white, and BAT or BAT-like depots are brown.

It is well-established that the various adipose depots display metabolic heterogeneity and are intrinsically different within each species. In humans, fat deposition in the upper body, mainly the visceral but also the subcutaneous abdominal depot, is linked to a higher risk of metabolic dysfunction; while lower body adiposity in the subcutaneous gluteal and femoral regions is associated with lower risk and may even be protective (336). Rodent studies reveal that surgical removal of visceral fat pads improves insulin action, glucose tolerance, and longevity (337,338), while the removal of subcutaneous fat pads can cause metabolic syndrome (339). In addition, subcutaneous, but not visceral, donor AT transplanted into the visceral region of recipient mice improves glucose metabolism (340). In contrast, human studies have shown that the removal of small amounts of omental AT in individuals with obesity provided no metabolic health benefits (341). Likewise, liposuction (~10 kg) of subcutaneous AT in humans neither harmed nor improved the cardio-metabolic profile (342,343). Nevertheless, fat is redistributed from the subcutaneous to visceral depots during aging (344) in conjunction with increasing prevalence of chronic diseases such as hypertension, T2DM, and cardiovascular disease, suggesting that subcutaneous AT may be metabolically beneficial in humans as has been extensively reported in rodents.

 

Studies of depot-specific expression patterns have enhanced our understanding of the mechanisms underlying abdominal versus gluteal and femoral adiposity (345–347). Unique expression patterns in different adipose tissue depots in mice indicate substantial difference in the expression of homeobox (HOX) developmental genes (348). Not surprisingly, HOX genes exhibit differential expression patterns in human compared to mouse fat depots (346,347). In contrast, structural and hormonal regulators, including collagen VI (349,350) and glucocorticoids (351,352), respectively, that influence fat distribution are similarly associated with AT expansion in both rodents and humans.

 

Similar to humans (353), female rodents have a higher percentage fat mass relative to males, yet remain more insulin sensitive (354). However, there are many notable sex differences in rodent versus human depots. The inguinal depots of female mice contain mammary glands and the gonadal fat pad is near reproductive tissue, which is not the case in humans. In addition, high-fat diet-induced obesity affect men and women alike, but in many strains of mice females are resistant HFD obesity, unlike male mice (355,356). Furthermore, the periovarian (visceral) fat pad in female mice has been shown to be more insulin sensitive than the inguinal fat pad (354), which is contrary to human data that indicates in women the gluteal and femoral depots are more insulin sensitive relative to the visceral AT (357).

 

Current literature suggests that the secretion patterns of adipokines (including leptin, interleukin-6, and tumor necrosis factor α) in the visceral versus subcutaneous depots of humans are relatively similar to that of rodents. Interestingly, lower body AT has been shown to secrete more metabolically favorable adipokines such as adiponectin (358). These observations are similar in rodents studies (340).

 

While lipolysis can be stimulated in rodents and humans under similar physiological conditions, important biological differences in AT lipolysis among these species have been suggested. The β1 and β2 adrenergic receptors (AR) are ubiquitously expressed in rodents and humans, while β3-AR expression is confined to white AT in rodents and only marginally expressed in human adipocytes (359). The α2-ARs are highly expressed in the subcutaneous AT of humans and act to inhibit lipolysis (360), but are absent in rodent adipocytes. Though common factors, including catecholamines, growth hormone, and cortisol, are similar among species in regulating lipolysis, differences in the response to other lipolytic agents have also been reported. Natriuretic peptide induces lipolysis in humans, but not in rodents (361), while adrenocorticotropic hormone and alpha-melanocyte-stimulating hormone modulate lipolysis in rodent but not human adipocytes (362,363). Therefore, it is important to account for these differences and commonalities in AT lipolysis among species.

 

Rodent studies are essential to expand our understanding of pathways underlying the associations between fat distribution and metabolic health and disease. Fortunately, there are many shared traits among rodent fat pads and human fat depots. However, given the clear differences in adipose depot location and physiology between the species, interpretation of experimental data and the extrapolation of conclusions drawn from rodent data to humans should be conducted with appropriate caution and caveats.

 

Dermal Adipose Tissue

 

A thick layer of adipocytes, historically referred to as subcutaneous AT, underlies the reticular dermis in both rodents and humans (364). Recent studies have revealed major differences between the adipocytes from this dermal layer and more typical subcutaneous adipocytes found in other locations (364–366). Today, dermal adipose tissue (dWAT) is considered a separate adipose depot that is distinguishable from subcutaneous fat (364). Two unique features of dermal adipocytes in this regard are that they can alter their cellular characteristics and have high turnover rates (366). An additional distinguishing factor for dWAT is its organization. In rodents, dWAT forms several adipocyte layers between the dermis and muscle layer (panniculus carnosus) (367). Human dWAT is present as individual units referred to as dermal cones. These cones are concentrated around pilosebaceous units that functionally interact with each other to form the dWAT structure (366,368). Interestingly, only body regions prone to scarring contain dermal cones (368), indicating a potential role for dWAT in scarring and wound healing. Also, dWAT can regenerate after injury. Following injury, adipogenesis is activated in the proliferative phase of wound healing and dermal adipocytes repopulate the wound (366,367). This is a critical event, as mouse models lacking mature adipocytes cannot recruit the fibroblasts required for wound healing (369–371).

 

Other identified roles for dWAT include insulation (372), barrier protection from skin infection (373), and hair follicle cycling (374). It is well known that brown adipose tissue (BAT) rapidly responds to cold temperature challenges by mobilizing lipids for heat generation (adaptive thermogenesis), yet dWAT slowly responds to these challenges by thickening/expanding over days to provide an effective layer of insulation (367,372). Mouse models lacking adequate dWAT undergo chronic activation of BAT since the dWAT cannot provide adequate mitigation of body temperature (367). Conversely, obese mice with excess dWAT undergo minimal adaptive thermogenesis (367). The dWAT thickening observed with cold exposure also occurs with bacterial exposure. Adipocytes in dWAT differentiate and become hypertrophic and result in a thicker dWAT layer in response to epidermal Staphylococcus aureus. This dWAT adipocyte reaction is also critical for immune response to bacterial invasion (373). Hair follicles go through repeated rounds of death and regrowth, referred to as the hair follicle cycle (367). Robust dWAT expansion is characterized by increased adipogenesis and dermal adipocyte hypertrophy that accompanies the regrowth of hair follicles (374). Conversely, inhibiting adipogenesis impedes hair follicle regeneration. In several species of mammals, a thickening of the hair coat accompanies dWAT expansion in response to cold exposure (367). In summary, dWAT has distinct roles from subcutaneous AT. Thus far, unlike other AT depots, the contribution of dWAT to metabolic health has not been investigated. Nonetheless, there is clear evidence that dWAT has distinct structures and functions and plays a role in variety of physiological processes.

 

Epicardial AT

 

Epicardial AT (EAT) has recently emerged as an important player in the development of cardiovascular disorders (375,376). Notably, EAT is distinct from pericardial fat. While pericardial AT surrounds the pericardium, EAT lies between the visceral pericardium and the myocardium and shares a blood supply with the coronary arteries (375–378). The adipocytes in EAT are smaller than those in other visceral or subcutaneous depots and are outnumbered by preadipocytes; this is thought to be related to the high energy requirement of the heart, which normally favors oxidation of fatty acids over other substrates (376,379). Furthermore, the gene expression and adipokine secretion profiles of EAT are unique from those of other depots (376,380,381).

 

In normal physiological conditions, EAT behaves like BAT and serves to protect the coronary vessels and myocardium against hypothermia (376,382). In pathologies such as coronary artery disease and type 2 diabetes, EAT can display an extensive pro-inflammatory signature (383–385). Macrophages and mast cells have been shown to infiltrate EAT, undergo activation, and through a cascade of signaling events facilitate lipid accumulation in atherosclerotic plaques (376,384). Pro-inflammatory adipokine secretion from EAT has also been shown to induce atrial fibrosis (381). Further, insulin sensitivity and EAT thickness are inversely correlated, whereas fasting glucose and EAT size are positively correlated, with enlarged EAT depots often found in individuals with type 2 diabetes (376,386,387). These data suggest that EAT functions as a distinct fat depot with important physiological and pathological roles.

 

METABOLIC DYSFUNCTION ASSOCIATED WITH ADIPOSE TISSUE

 

Adipose Tissue Expandability and Metabolic Health

 

White AT retains the ability to expand during adult life to accommodate chronic excess caloric intake. AT expansion is characterized by adipocytes accumulating lipid and growing in size (hypertrophy) or number (hyperplasia or adipogenesis) or increasing in both size and number. Evidence suggests that the capacity of subcutaneous AT to expand as well as the manner of expansion (hypertrophy vs. hyperplasia) can influence cardiometabolic health. This mechanism is thought to underlie the benefits of thiazolidinedione (TZD) medications, which are approved for the treatment of type 2 diabetes (388,389). These PPARg agonists stimulate preadipocyte differentiation and the proliferation of adipocytes (390,391), especially in subcutaneous depots as compared to visceral adipose tissue (392), which leads to increased adiponectin levels and improved insulin sensitivity (393,394). Hence, there is a clear rationale to further characterize the mechanisms of AT expansion through adipocyte proliferation in humans that may inform future effective drug therapies.

 

On the other hand, the presence of enlarged, hypertrophic adipocytes, a lack of hyperplasia, and development of AT inflammation and fibrosis reflect impaired AT expansion and is associated with metabolic derangements (395–398). These observations support the “AT expandability hypothesis”, which postulates that a lack of adipogenesis (or hyperplasia) results in the limited capacity of AT to expand and store lipid, causing ectopic fat accretion and “lipotoxicity” in non-adipose tissues such as skeletal muscle and liver (399–401). The degree of ectopic lipid deposition in the liver and skeletal muscle is a significant determinant of metabolic syndrome (MetS) and the development of T2D and CVD (402).

 

Other findings do not support the AT expandability hypothesis and indicate that higher adipogenesis does not necessarily denote improvements in metabolic health. These studies report a higher population of small adipocytes (a measure of hyperplasia) in the AT of individuals with insulin resistance and T2D (403–406) and in those with more visceral AT and liver fat (406,407). Experimental overfeeding intervention studies have shown that individuals with smaller adipocytes at baseline have poorer metabolic health outcomes (i.e. impaired insulin sensitivity) in response to substantial weight gain than those with larger adipocytes (408,409). In addition, one in vivo analysis in humans demonstrated that increased hyperplastic expansion correlated with an increased number of metabolic syndrome components (410). Collectively, these data imply an alternative model of impaired AT expansion, as compared to the mechanisms proposed by the AT expandability hypothesis, and suggest that there is not a deficiency in hyperplasia but an abundance of adipocytes with a limited capacity to adequately expand and accommodate lipid, whether large or small. This inability to store excess lipid in AT is thought to be a key feature that leads to metabolic dysfunction.

 

Although the mechanisms of adipose expansion and its precise role in promoting glucolipid dysregulation remain a matter of debate, all of the aforementioned studies support the view that AT’s capacity to expand is intimately related to metabolic homeostasis, as the failure to store excess lipid appropriately in AT can contribute to many obesity-related complications.

 

AT Inflammation

 

A variety of cell types from both the innate and adaptive immune systems have been found in AT (411–413). Though resident AT immune cells are critical to normal adipocyte function in healthy individuals, AT inflammation, as mentioned in several preceding sections, is considered a major contributor to the metabolic dysfunction associated with obesity (413,414).

 

During nutrient excess as AT expandability reaches its limit, a strong association exists between adipocyte size and adipocyte death (415). In response to adipocyte death, pro-inflammatory macrophages surround dead and dying cells and remove debris from the damaged area. During this process, macrophages acutely produce inflammatory cytokines (413,416). In obesity, this cytokine production often fails to resolve, becomes chronic, and leads to impaired adipocyte insulin signaling, further inflammation, and a continued worsening of AT dysfunction (413,416,417). In a field that is rapidly changing, it is worth mentioning that some degree of inflammatory signaling might be required for normal AT function. The pro-inflammatory cytokines TNF alpha and oncostatin M have been shown to be required for proper AT expansion and maintenance of insulin sensitivity in mice (414,418–420). Although AT inflammation clearly has detrimental effects in obesity, evidence also indicates adaptive and homeostatic roles for pro-inflammatory signaling in AT expansion and function.

 

Metabolically Healthy (MHO) versus Metabolically Unhealthy (MUO) Obesity

 

An estimated 10-30% of individuals with obesity are considered to have “metabolically healthy obesity” (MHO) with favorable metabolic profiles (421). Although there is currently no consensus for parameters used to classify MHO,these individuals are characterized by normal insulin sensitivity, normal fasting glucose levels, low incidence of hypertension, and blood lipid profiles in the healthy range (422,423) (Figure 13). In contrast, individuals with “metabolically unhealthy obesity” (MUO) have comparable body mass indices (BMI) but develop metabolic aberrations. Factors that distinguish individuals with MHO from MUO (Figure 13) highlight the premise that metabolic health risk is not solely dependent on body weight and are described in more detail below. Understanding these characteristics and potential mechanisms underlying the MHO and the perceived healthy metabolic state of these individuals is an important area of ongoing research.

 

Figure 13. Clinical and biological factors thought to distinguish metabolically healthy obesity (MHO) from metabolically unhealthy obesity (MUO). Abbreviations: VAT – Visceral AT, SubQ AT – Subcutaneous AT, EMCL - extramyocellular lipid; IMCL – intramyocellular lipid; HDL – high density lipoprotein.

Evidence suggests that WAT plays a critical role in the development of MHO vs MUO, as its properties, location, and function are closely linked with cardiometabolic risk. Fat distribution (422), as well as changes associated with AT expansion, including the capacity for adipocyte differentiation (403) and parameters related to ECM remodeling (424), may also contribute to the MHO phenotype. In addition, adipose-derived circulating factors that impact whole-body metabolism have been implicated in MHO vs MUO differences (425).  However, studies have shown that the location of AT, rather than overall obesity, may be a stronger predictor of metabolic health risks (336). The accumulation of upper-body fat, namely visceral AT (VAT) but also subcutaneous abdominal (scABD) adipose tissue, confers a higher risk of obesity-related disorders (426), while lower-body fat (subcutaneous gluteal and femoral) may be metabolically protective (427). The preserved metabolic function of individuals with MHO may be attributed to significantly lower accumulation of VAT relative to MUO (422,428,429). As described in the previous section, enlarged adipocyte size, independent of adiposity, is positively correlated with the development of insulin resistance and impaired metabolic health (396). MUO individuals have been shown to have larger adipocytes than their MHO counterparts (430,431). Hypertrophic adipocytes may represent the failure of subcutaneous AT to expand and store excess fat, which can ultimately lead to ectopic lipid deposition in non-adipose tissues such as the liver and skeletal muscle (402).

 

Ectopic lipid accumulation in both the liver and skeletal muscle is of pathophysiological significance as part of the “lipotoxicity” hypothesis and may also impact the varying health risk of MHO vs MUO. Extramyocellular lipid (EMCL) and intramyocellular lipid (IMCL) are postulated to cause defects in insulin signaling and reduce insulin-stimulated skeletal muscle glucose uptake (432). These lipid stores are strong correlates of insulin resistance and are increased in individuals with T2D (402). Paradoxically, increased IMCL is also observed in ‘insulin sensitive’ athletes, which may be attributed to the oxidative capacity of skeletal muscle (433) and increased glucose transport in trained muscle (434). Intrahepatic lipid accumulation strongly associates with impaired insulin-induced suppression of hepatic glucose production, even independently of visceral AT amount, and the development of T2D (435). Ectopic fat in both the liver and skeletal muscle has been shown to be lower in MHO than MUO individuals (422,436,437) (refer to Figure 13).

 

The differential secretion of pro-inflammatory adipokines has also been proposed as a mechanism underlying the MHO phenotype (438–440) by some investigators, although others have reported conflicting results (441). Nevertheless, studies show reduced macrophage infiltration in MHO (442,443), supporting a reduced inflammatory state in these individuals. Intriguing data implicating potential genetic differences among MHO vs. MUO indicate that specific polymorphisms in genes, including the adiponectin receptor 1 and hepatic lipase, may be associated with the MHO phenotype (436). In addition, genes encoding some proinflammatory cytokines can be more highly expressed in the adipose tissue of MUO compared with MHO individuals (444,445).

 

A lingering question that remains unanswered is whether MHO subjects will sustain a healthy metabolic state throughout their lifespan or if they will eventually become MUO. An additional question is if a healthy lifestyle can help to maintain a favorable profile and prevent the transition to MUO. Indeed, longitudinal data clearly show that not all MHO individuals remain metabolically healthy, as up to 30% progress to MUO over a 5-10 year time frame (446–448). Of note, the length of time for follow-up assessments of MHO individuals is an important factor that may have considerable effects on the observed outcomes, because the total number of years as obese and aging can independently increase mortality risk. A major obstacle in advancing the understanding of the MHO phenotype is the manner by which metabolic health is described, including the parameters used to define insulin sensitivity and metabolic syndrome (449). Defining metabolic outcomes based on differing criteria can result in a broad range of reported prevalence, discrepancies regarding the observed characteristics, varied interpretations of health and mortality risks, and disagreement concerning the implications of therapeutic interventions. In addition, the use of the “healthy” descriptor may be misrepresentative of the true medical risks to these individuals, as long-term adverse health outcomes have been observed in individuals with MHO during follow-up years, thus no longer characterizing them as “metabolically healthy” (450,451). Additional long-term prospective studies are necessary to assess features of the MHO phenotype and to observe how the factors discussed above are altered over time. In addition, these studies may reveal if WAT function is a cause or consequence of the MHO and MUO phenotypes.

 

Lipodystrophy

 

While excessive adiposity, or obesity, can have adverse health consequences, deficiency of AT mass, as seen in lipodystrophy, can also lead to derangements in glucolipid metabolism. Lipodystrophy encompasses a group of rare, heterogeneous, genetic or acquired disorders characterized by varying degrees of severe reduction or absence of body fat (452) . Anatomically, this disease can present as a partial (i.e. localized to certain body areas) or generalized lack of AT. The combined overall prevalence of lipodystrophy is estimated to be 1 in 1,000,000 individuals, with ~1000 patients reported with genetic forms (453). Lipodystrophy associated with highly active antiretroviral therapy for HIV is one of the most common acquired forms worldwide (454). The diagnosis of this disorder mostly relies on clinical criteria. In most cases of generalized lipodystrophy, standard physical examination is sufficient to establish this diagnosis. In contrast, partial lipodystrophy may be represented by mild physical abnormalities and can sometimes be misdiagnosed as common forms of central (abdominal) obesity, suggesting that this form of lipodystrophy may be an underestimated condition (455). Although the pathological basis of most lipodystrophies remains unclear, it is well-accepted that AT dysfunction is a primary determinant of the resulting health consequences in these patients. Limited development and non-expandability of AT and failure of AT to accommodate excess lipid leads to the redistribution and storage of fat ectopically in the liver and skeletal muscle and the development of non-alcoholic fatty liver disease, often severe insulin resistance and type 2 diabetes, hypertriglyceridemia, and associated diseases (456–458).

 

Markedly reduced levels of leptin and adiponectin may also contribute to the pathology of lipodystrophy. As described above, leptin plays an important role in the regulation of body weight and energy metabolism, and leptin deficiency is common in lipodystrophic patients, due to the lack of AT (452). Low leptin levels can not only impact glucose metabolism (459) but also contribute to increased appetite and excessive caloric intake in these patients (460). Transgenic animal models shed light on the pathology of lipodystrophy and confirm the importance of AT in normal physiology. Fatless rodents, created via AT ablation, display hypertriglyceridemia and ectopic lipid accumulation, along with severe insulin resistance (457,461). In addition, several groups have successfully treated the metabolic derangements in these fatless mice by transplanting AT from wild-type animals (461–464). However, transplantation of AT from leptin-deficient mice did not improve the metabolic abnormalities in fatless mice (465), while leptin administration in the fatless mice ameliorated insulin resistance and hepatic steatosis (466). In humans with total lipodystrophy, leptin treatment also markedly improves the severe hypertriglyceridemia and insulin resistance that accompanies this disorder (467).  These studies confirm that both AT and leptin deficiency play a central role in lipodystrophy-associated pathologies.

 

Adiponectin has insulin-sensitizing and anti-inflammatory effects, and low levels of this adipokine have also been observed in patients with lipodystrophies (468). Recombinant adiponectin, adiponectin analogues (i.e. osmotin), and compounds that upregulate endogenous adiponectin (i.e. TZDs) have all been proposed as treatment approaches for lipodystrophy (469). In a fatless mouse model, treatment with the globular domain of adiponectin significantly improved the hyperglycemia and hyperinsulinemia characteristic of these lipoatrophic diabetic mice (146). Interestingly, the insulin resistance observed in these mice was completely ameliorated by treatment with both adiponectin and leptin, but only partially by either adiponectin or leptin alone (146), suggesting that both adiponectin and leptin deficiency may contribute to the insulin resistance in humans with lipodystrophy.

 

Studies to date support the premise that too little AT, as seen in lipodystrophy, appears to be just as deleterious as too much AT. Emerging data reveal that patients with lipodystrophy may have reduced survival and high mortality at an early age, predominantly due to cardiometabolic complications (470–472). Lipodystrophy has no cure; therefore, the primary treatment option is to improve metabolic outcomes via physical activity and dietary and pharmacological interventions. Conventional insulin-sensitizing agents, such as metformin and TZDs, are often used (453,473), and leptin replacement is also an approved therapy for total congenital lipodystrophy (467). Future investigations to better understand the pathogenesis and the clinical manifestation of lipodystrophy syndromes are essential for the development of improved therapeutics.

 

Adipose Tissue and Reproduction

 

While many studies have primarily examined the influence of white AT on the metabolic consequences associated with obesity, less frequently mentioned is the interplay between AT and reproductive health. Nevertheless, it is well established that AT is important for the normal function of the reproductive system, including the production and regulation of sex and reproductive hormones, pubertal development, and the maintenance of pregnancy and lactation (474).

 

Leptin and adiponectin are the most investigated adipokines as mediators of reproductive health and pathology. Receptors for both leptin and adiponectin have been identified in all major reproductive tissues, including the testes, placenta, ovaries, oviducts and endometrium (475). Obese mice that are deficient in leptin or the leptin receptor are unable to reproduce (476). Although rare, humans with leptin mutations have been identified, and studies in these individuals have validated the infertility findings in rodents (477).

 

Leptin administration in rodents was shown to increase luteinizing hormone (LH), follicle stimulating hormone (FSH), and ovarian and uterine weights in females, and testosterone, testicular weights and sperm counts in males (478,479). During human pubertal development, there is a steady increase in leptin, stimulating a rise in testosterone levels and fat-free mass in boys and in estradiol and fat mass in girls (480,481). Adiponectin administration was shown to inhibit gonadotrophin releasing hormone (GnRH), LH, and FSH (482,483) in pigs and increase estrogen and progesterone (484) in rats, while circulating levels in humans have been shown to be associated with serum levels of sex hormones (primarily estrogens), though this correlation was largely mediated by body weight (485).

 

In humans, leptin levels increase during pregnancy and rapidly fall in the post-partum period (486), and other reports have suggested that adiponectin may influence the amount of gestational weight gain and weight maintenance post-partum, even after adjusting for the sum of skinfold thickness and BMI (487,488). The effects of leptin on fetal development continue to be investigated, and have been suggested to correlate with fetal growth, birth weight, and organogenesis (489). Adiponectin plasma levels were shown to be significantly lower in overweight patients than normal weight women during pregnancy and negatively correlated with progressive gestational age and weight gain (490). In addition, women with low adiponectin concentrations experienced a significantly increased risk of gestational diabetes mellitus (491,492), and large reductions in adiponectin levels during pregnancy may also predict large-for-gestational-age offspring and increased birth weight (493). Interestingly, several studies have shown that higher adiponectin levels are associated with increased conception success in women undergoing assisted reproduction approaches (494). Overall, these studies are consistent with adipose-derived leptin and adiponectin having critical roles in reproductive function.

 

Many lines of evidence also demonstrate that either insufficient or excessive AT can have detrimental effects on reproductive health. Women with lipodystrophy disorders (see above), are characterized by AT and leptin deficiency and are frequently infertile (495,496). Anorexia nervosa is an eating disorder characterized by very low AT mass that is often accompanied by amenorrhea (absence of at least three menstrual periods in a row) (497). It is estimated that ~38% of women affected by anorexia experience infertility (498). Leptin deficiency is common in these patients (499)and may lead to disruptions in downstream neuroendocrine signaling (500).  This was tested when leptin replacement to women with hypogonadotrophic hypogonadism due to anorexia nervosa or excessive exercise was found to restore normal periods (501). Estrogen deficiency in these women results in major implications for bone health, ultimately contributing to increased osteopenia or osteoporosis (502). 

 

Body weight has been shown to predict testosterone levels in men (503); and obesity, specifically central adiposity, is associated with low testosterone levels (504). Increased AT also leads to elevated estradiol, resulting in reduced circulating testosterone through feed-back inhibition of gonadotrophs (504,505). A common medical condition in women at the crossroads of dysfunctional AT and reproduction is polycystic ovary syndrome (PCOS), which in roughly 50% of affected women is associated with increased central obesity and metabolic health risk. PCOS is commonly defined using the consensus of Rotterdam, which requires two of three criteria: polycystic ovaries on ultrasonography, hyperandrogenism, or amenorrhea. Studies of PCOS generally show that adiponectin levels are lower in these patients (506). Another burgeoning area of research is the study of excessive AT and reproductive malignancies, as obesity is known to increase the risk of breast, uterine, cervical, and prostate cancers (507). Studies have reported inverse relationships between leptin and adiponectin levels with breast, endometrial, ovarian, and prostate cancers (508,509).

 

EMERGING AREAS IN ADIPOCYTE BIOLOGY

 

Critical considerations in the study of fat tissue are its cellular complexity and heterogeneity. AT depots can exist in close association with other organs and act physiologically as metabolic “sinks” that store excess energy as lipid in a protective manner, or they can promote systemic metabolic dysfunction by secreting excess lipid or inflammatory adipokines. As the recognition of distinct AT depots increases, so does our understanding of their diversity. A recent review considers the locations and functions of several depots, ranging from facial AT to cardiovascular AT as well as the presence of adipocytes in bone marrow, within and between muscle beds, and joints (510). Currently, we are experiencing a new and exciting period in AT research with the focus shifting toward recognizing neglected AT depots, the expanding types of adipocytes, and the complex developmental and sex-regulated origins of adipocytes. Adipocytes are critical secretory cells that contribute a variety of circulating proteins, including endocrine hormones. Of course, adipocytes also produce lipids and can release genetic material that can have profound systemic functions.

 

Much remains to be discovered about the types of nerves present in fat tissue and how they vary according to AT type and location. How these AT nerves act to regulate metabolic homeostasis is a current focus of fat cell biology. Recent advances in whole tissue AT imaging and studies on brain-adipose communication suggest we are just beginning to uncover the capabilities and function of AT nerves, and there are many unanswered questions in this field (511). Research on the molecular pathways that connect AT innervation to insulin action in obesity and diabetes may provide insight into our understanding of the pathogenesis of metabolic disease states.

 

Another developing area of fat cell biology is the effects of exercise on adipocyte function. Recent studies have shown that transplantation of subcutaneous AT from exercise-trained mice improves glucose tolerance and insulin sensitivity in recipient, non-exercised mice (512), and strongly suggest that exercise favorably remodels AT to improve systemic metabolic health. Recently, an AT-derived lipid was shown to increase fatty acid uptake in skeletal muscle (513). The importance of AT to whole-body energy metabolism is well established; yet, the impacts of different types of endurance or resistance exercise on adipose tissue dynamics remains largely understudied, particularly in the context of obesity and other metabolic disease states.  

 

A newly discovered pathway shows that lipids can be released by adipocytes in the form of exosome-sized, lipid-filled vesicles (514). This process occurs independently of canonical lipolytic pathways, and adipocyte exosomes deliver excess lipid to local macrophages in obesity (514). Other novel pathways of paracrine regulation have also been demonstrated in AT. These paradigm-shifting observations demonstrated that extracellular vesicles (EV) from endothelial cells in adipose tissue can provide lipids and proteins to adjacent adipocytes. This EV communication between endothelial cells and adipocytes within AT is bi-directional and is regulated by fasting/refeeding and in conditions of obesity (515). These very recent observations reveal the highly complex signaling mechanisms that exist in AT.  

 

Though it was once considered a mere energy storage site, AT is now considered an important endocrine organ and site of inflammatory cell signaling that governs not only survival but also plays critical roles in reproduction and in glucometabolic homeostasis. As scientific methods for the study of AT continue to rapidly evolve, so does our understanding regarding the metabolic, biomechanical, immune, and secretory functions of AT in normal physiology and metabolic disease.

 

           ACKNOWLEDGEMENTS

 

The authors are grateful to Anik Boudreau and Christina Zunica for their assistance in editing and referencing the chapter. This work was supported by National Institutes of Health Grant R01 DK052968.

 

           REFERENCES

 

  1. Cuthbertson D, Tompsett S. The degree of unsaturation of the fats of human adipose tissue in relation to depth from skin surface. Biochem. J. 1933;27(4):1103–1106.
  2. Cook KS, Min HY, Johnson D, Chaplinsky RJ, Flier JS, Hunt CR, Spiegelman BM. Adipsin: a circulating serine protease homolog secreted by adipose tissue and sciatic nerve. Science 1987;237(4813):402–405.
  3. Cianflone K, Maslowska M, Sniderman AD. Acylation stimulating protein (ASP), an adipocyte autocrine: new directions. Semin. Cell Dev. Biol. 1999;10(1):31–41.
  4. Saleh J, Al-Wardy N, Farhan H, Al-Khanbashi M, Cianflone K. Acylation stimulating protein: a female lipogenic factor? Obes. Rev. 2011;12(6):440–448.
  5. Zhang Y, Proenca R, Maffei M, Barone M, Leopold L, Friedman JM. Positional cloning of the mouse obese gene and its human homologue. Nature 1994;372(6505):425–432.
  6. Chlouverakis C. Insulin resistance of parabiotic obese-hyperglycemic mice (obob). Horm. Metab. Res.1972;4(3):143–148.
  7. Cava A La, Matarese G. The weight of leptin in immunity. Nat. Rev. Immunol. 2004;4(5):371–379.
  8. Carobbio S, Pellegrinelli V, Vidal-Puig A. Adipose tissue function and expandability as determinants of lipotoxicity and the metabolic syndrome. In: Obesity and Lipotoxicity. Springer, Cham; 2017:161–196.
  9. Cancers Associated with Overweight and Obesity Make up 40 percent of Cancers Diagnosed in the United States | CDC Online Newsroom | CDC. Available at: https://www.cdc.gov/media/releases/2017/p1003-vs-cancer-obesity.html. Accessed January 21, 2019.
  10. Hales CM, Carroll MD, Fryar CD, Ogden CL.Prevalence of obesity among adults and youth: United States, 2015-2016. NCHS data brief, no 288. Hyattsville, MD: National Center for Health Statistics; 2017. Available at: https://www.cdc.gov/nchs/products/databriefs/db288.htm. Accessed February 27, 2019.
  11. Hruby A, Hu FB. The epidemiology of obesity: A big picture. Pharmacoeconomics 2015;33(7):673–689.
  12. Nedergaard J, Bengtsson T, Cannon B. Unexpected evidence for active brown adipose tissue in adult humans. Am. J. Physiol. Metab. 2007;293(2):E444–E452.
  13. Zingaretti MC, Crosta F, Vitali A, Guerrieri M, Frontini A, Cannon B, Nedergaard J, Cinti S. The presence of UCP1 demonstrates that metabolically active adipose tissue in the neck of adult humans truly represents brown adipose tissue. FASEB J. 2009;23(9):3113–3120.
  14. Cypess AM, Lehman S, Williams G, Tal I, Rodman D, Goldfine AB, Kuo FC, Palmer EL, Tseng Y-H, Doria A, Kolodny GM, Kahn CR. Identification and importance of brown adipose tissue in adult humans. N. Engl. J. Med.2009;360(15):1509–1517.
  15. Au-Yong ITH, Thorn N, Ganatra R, Perkins AC, Symonds ME. Brown adipose tissue and seasonal variation in humans. Diabetes 2009;58(11):2583–7.
  16. Hibi M, Oishi S, Matsushita M, Yoneshiro T, Yamaguchi T, Usui C, Yasunaga K, Katsuragi Y, Kubota K, Tanaka S, Saito M. Brown adipose tissue is involved in diet-induced thermogenesis and whole-body fat utilization in healthy humans. Int. J. Obes. (Lond). 2016;40(11):1655–1661.
  17. de Sá PM, Richard AJ, Hang H, Stephens JM. Transcriptional regulation of adipogenesis. In: Comprehensive Physiology.Vol 7. Hoboken, NJ, USA: John Wiley & Sons, Inc.; 2017:635–674.
  18. Cinti S. The adipose organ at a glance. Dis. Model. Mech. 2012;5(5):588–594.
  19. Wu J, Boström P, Sparks LM, Ye L, Choi JH, Giang A-H, Khandekar M, Virtanen KA, Nuutila P, Schaart G, Huang K, Tu H, van Marken Lichtenbelt WD, Hoeks J, Enerbäck S, Schrauwen P, Spiegelman BM. Beige adipocytes are a distinct type of thermogenic fat cell in mouse and human. Cell 2012;150(2):366–376.
  20. Barbatelli G, Murano I, Madsen L, Hao Q, Jimenez M, Kristiansen K, Giacobino JP, De Matteis R, Cinti S. The emergence of cold-induced brown adipocytes in mouse white fat depots is determined predominantly by white to brown adipocyte transdifferentiation. Am. J. Physiol. Metab. 2010;298(6):E1244–E1253.
  21. Himms-Hagen J, Melnyk A, Zingaretti MC, Ceresi E, Barbatelli G, Cinti S. Multilocular fat cells in WAT of CL-316243-treated rats derive directly from white adipocytes. Am. J. Physiol. Physiol. 2000;279(3):C670–C681.
  22. Young P, Arch JRS, Ashwell M. Brown adipose tissue in the parametrial fat pad of the mouse. FEBS Lett.1984;167(1):10–14.
  23. Loncar D, Afzelius BA, Cannon B. Epididymal white adipose tissue after cold stress in rats. I. Nonmitochondrial changes. J. Ultrastruct. Mol. Struct. Res. 1988;101(2–3):109–122.
  24. Wang S, Pan M-H, Hung W-L, Tung Y-C, Ho C-T. From white to beige adipocytes: therapeutic potential of dietary molecules against obesity and their molecular mechanisms. Food Funct. 2019;10:1263–1279.
  25. Mika A, Macaluso F, Barone R, Di Felice V, Sledzinski T. Effect of exercise on fatty acid metabolism and adipokine secretion in adipose tissue. Front. Physiol. 2019;10(26):1–7.
  26. Reynés B, Palou M, Rodríguez AM, Palou A. Regulation of adaptive thermogenesis and browning by prebiotics and postbiotics. Front. Physiol. 2018;9:1–15.
  27. Kaisanlahti A, Glumoff T. Browning of white fat: agents and implications for beige adipose tissue to type 2 diabetes. J. Physiol. Biochem. 2019;75(1):1–10.
  28. Ikeda K, Maretich P, Kajimura S. The common and distinct features of brown and beige adipocytes. Trends Endocrinol. Metab. 2018;29(3):191–200.
  29. Giordano A, Smorlesi A, Frontini A, Barbatelli G, Cinti S. MECHANISMS IN ENDOCRINOLOGY: White, brown and pink adipocytes: the extraordinary plasticity of the adipose organ. Eur. J. Endocrinol. 2014;170(5):R159–R171.
  30. Cinti S. Pink Adipocytes. Trends Endocrinol. Metab. 2018:1–16.
  31. Apostoli AJ, Skelhorne-Gross GEA, Rubino RE, Peterson NT, Di Lena MA, Schneider MM, SenGupta SK, Nicol CJB. Loss of PPARγ expression in mammary secretory epithelial cells creates a pro-breast tumorigenic environment. Int. J. Cancer 2014;134(5):1055–1066.
  32. Chu M, Sampath H, Cahana DY, Kahl CA, Somwar R, Cornea A, Roberts CT, Varlamov O. Spatiotemporal dynamics of triglyceride storage in unilocular adipocytes. Gruenberg JE, ed. Mol. Biol. Cell 2014;25(25):4096–4105.
  33. Kersten S. Physiological regulation of lipoprotein lipase. Biochim. Biophys. Acta - Mol. Cell Biol. Lipids2014;1841(7):919–933.
  34. Fielding BA, Frayn KN. Lipoprotein lipase and the disposition of dietary fatty acids. Br. J. Nutr. 1998;80(6):495–502.
  35. Frayn K. Adipose tissue as a buffer for daily lipid flux. Diabetologia 2002;45(9):1201–1210.
  36. Luo L, Liu M. Adipose tissue in control of metabolism. J. Endocrinol. 2016;231(3):R77–R99.
  37. Harris CA, Haas JT, Streeper RS, Stone SJ, Kumari M, Yang K, Han X, Brownell N, Gross RW, Zechner R, Farese R V, Jr. DGAT enzymes are required for triacylglycerol synthesis and lipid droplets in adipocytes. J. Lipid Res. 2011;52(4):657–667.
  38. Smith SJ, Cases S, Jensen DR, Chen HC, Sande E, Tow B, Sanan DA, Raber J, Eckel RH, Farese R V.Obesity resistance and multiple mechanisms of triglyceride synthesis in mice lacking Dgat. Nat. Genet.2000;25(1):87–90.
  39. Song Z, Xiaoli AM, Yang F. Regulation and metabolic significance of de novo lipogenesis in adipose tissues. Nutrients 2018;10(1383):1–22.
  40. Swierczynski J, Goyke E, Wach L, Pankiewicz A, Kochan Z, Adamonis W, Sledzinski Z, Aleksandrowicz Z.Comparative study of the lipogenic potential of human and rat adipose tissue. Metabolism. 2000;49(5):594–599.
  41. Letexier D, Pinteur C, Large V, Fréring V, Beylot M. Comparison of the expression and activity of the lipogenic pathway in human and rat adipose tissue. J. Lipid Res. 2003;44(11):2127–2134.
  42. Aarsland A, Chinkes D, Wolfe RR. Hepatic and whole-body fat synthesis in humans during carbohydrate overfeeding. Am. J. Clin. Nutr. 1997;65(6):1774–1782.
  43. Kawahito S, Kitahata H, Oshita S. Problems associated with glucose toxicity: Role of hyperglycemia-induced oxidative stress. World J. Gastroenterol. 2009;15(33):4137.
  44. Crewe C, Zhu Y, Paschoal VA, Joffin N, Ghaben AL, Gordillo R, Oh DY, Liang G, Horton JD, Scherer PE.SREBP-regulated adipocyte lipogenesis is dependent on substrate availability and redox modulation of mTORC1. JCI Insight 2019;4(15). doi:10.1172/JCI.INSIGHT.129397.
  45. Herman MA, Peroni OD, Villoria J, Schön MR, Abumrad NA, Blüher M, Klein S, Kahn BB. A novel ChREBP isoform in adipose tissue regulates systemic glucose metabolism. Nature 2012;484(7394):333–338.
  46. Vijayakumar A, Aryal P, Wen J, Syed I, Vazirani RP, Moraes-Vieira PM, Camporez JP, Gallop MR, Perry RJ, Peroni OD, Shulman GI, Saghatelian A, McGraw TE, Kahn BB. Absence of carbohydrate response element binding protein in adipocytes causes systemic insulin resistance and impairs glucose transport. Cell Rep.2017;21(4):1021–1035.
  47. Shimano H, Shimomura I, Hammer RE, Herz J, Goldstein JL, Brown MS, Horton JD. Elevated levels of SREBP-2 and cholesterol synthesis in livers of mice homozygous for a targeted disruption of the SREBP-1 gene. J. Clin. Invest. 1997;100(8):2115–2124.
  48. Lodhi IJ, Wei X, Semenkovich CF. Lipoexpediency: de novo lipogenesis as a metabolic signal transmitter. Trends Endocrinol. Metab. 2011;22(1):1–8.
  49. Smith U, Kahn BB. Adipose tissue regulates insulin sensitivity: role of adipogenesis, de novo lipogenesis and novel lipids. J. Intern. Med. 2016;280(5):465–475.
  50. Zhou P, Santoro A, Peroni OD, Nelson AT, Saghatelian A, Siegel D, Kahn BB. PAHSAs enhance hepatic and systemic insulin sensitivity through direct and indirect mechanisms. J. Clin. Invest. 2019;129(10):4138–4150.
  51. Yore MM, Syed I, Moraes-Vieira PM, Zhang T, Herman MA, Homan EA, Patel RT, Lee J, Chen S, Peroni OD, Dhaneshwar AS, Hammarstedt A, Smith U, McGraw TE, Saghatelian A, Kahn BB. Discovery of a class of endogenous mammalian lipids with anti-diabetic and anti-inflammatory effects. Cell 2014;159(2):318–32.
  52. Eissing L, Scherer T, Tödter K, Knippschild U, Greve JW, Buurman WA, Pinnschmidt HO, Rensen SS, Wolf AM, Bartelt A, Heeren J, Buettner C, Scheja L. De novo lipogenesis in human fat and liver is linked to ChREBP-β and metabolic health. Nat. Commun. 2013;4:1528.
  53. Bruss MD, Khambatta CF, Ruby MA, Aggarwal I, Hellerstein MK. Calorie restriction increases fatty acid synthesis and whole body fat oxidation rates. Am. J. Physiol. Metab. 2010;298(1):E108–E116.
  54. Mottillo EP, Balasubramanian P, Lee Y-H, Weng C, Kershaw EE, Granneman JG. Coupling of lipolysis and de novo lipogenesis in brown, beige, and white adipose tissues during chronic β3-adrenergic receptor activation. J. Lipid Res. 2014;55(11):2276–2286.
  55. Braun K, Oeckl J, Westermeier J, Li Y, Klingenspor M. Non-adrenergic control of lipolysis and thermogenesis in adipose tissues. J. Exp. Biol. 2018;221:1–14.
  56. Kuriyama H, Shimomura I, Kishida K, Kondo H, Furuyama N, Nishizawa H, Maeda N, Matsuda M, Nagaretani H, Kihara S, Nakamura T, Tochino Y, Funahashi T, Matsuzawa Y. Coordinated regulation of fat-specific and liver-specific glycerol channels, aquaporin adipose and aquaporin 9. Diabetes 2002;51(10):2915–2921.
  57. Sztalryd C, Brasaemle DL. The perilipin family of lipid droplet proteins: Gatekeepers of intracellular lipolysis. Biochim. Biophys. acta. Mol. cell Biol. lipids 2017;1862(10 Pt B):1221–1232.
  58. Nielsen TS, Jessen N, Jorgensen JOL, Moller N, Lund S. Dissecting adipose tissue lipolysis: molecular regulation and implications for metabolic disease. J. Mol. Endocrinol. 2014;52(3):R199–R222.
  59. Greenberg AS, Egan JJ, Wek SA, Garty NB, Blanchette-Mackie EJ, Londos C. Perilipin, a major hormonally regulated adipocyte-specific phosphoprotein associated with the periphery of lipid storage droplets. J. Biol. Chem. 1991;266(17):11341–11346.
  60. Strålfors P, Björgell P, Belfrage P. Hormonal regulation of hormone-sensitive lipase in intact adipocytes: identification of phosphorylated sites and effects on the phosphorylation by lipolytic hormones and insulin. Proc. Natl. Acad. Sci. U. S. A. 1984;81(11):3317–3321.
  61. Garton AJ, Campbell DG, Cohen P, Yeaman SJ. Primary structure of the site on bovine hormone-sensitive lipase phosphorylated by cyclic AMP-dependent protein kinase. FEBS Lett. 1988;229(1):68–72.
  62. Anthonsen MW, Rönnstrand L, Wernstedt C, Degerman E, Holm C. Identification of novel phosphorylation sites in hormone-sensitive lipase that are phosphorylated in response to isoproterenol and govern activation properties in vitro. J. Biol. Chem. 1998;273(1):215–221.
  63. Lass A, Zimmermann R, Haemmerle G, Riederer M, Schoiswohl G, Schweiger M, Kienesberger P, Strauss JG, Gorkiewicz G, Zechner R. Adipose triglyceride lipase-mediated lipolysis of cellular fat stores is activated by CGI-58 and defective in Chanarin-Dorfman Syndrome. Cell Metab. 2006;3(5):309–319.
  64. Granneman JG, Moore H-PH, Krishnamoorthy R, Rathod M. Perilipin controls lipolysis by regulating the interactions of AB-hydrolase containing 5 (Abhd5) and adipose triglyceride lipase (Atgl). J. Biol. Chem.2009;284(50):34538–34544.
  65. Jensen MD. Gender differences in regional fatty acid metabolism before and after meal ingestion. J. Clin. Invest. 1995;96(5):2297–303.
  66. Gjedsted J, Gormsen LC, Nielsen S, Schmitz O, Djurhuus CB, Keiding S, Ørskov H, Tønnesen E, Møller N.Effects of a 3-day fast on regional lipid and glucose metabolism in human skeletal muscle and adipose tissue. Acta Physiol. 2007;191(3):205–216.
  67. Moro C, Pillard F, de Glisezinski I, Crampes F, Thalamas C, Harant I, Marques M-A, Lafontan M, Berlan M. Sex differences in lipolysis-regulating mechanisms in overweight subjects: effect of exercise intensity*. Obesity2007;15(9):2245–2255.
  68. Morigny P, Houssier M, Mouisel E, Langin D. Adipocyte lipolysis and insulin resistance. Biochimie2016;125:259–266.
  69. Czech MP. Mechanisms of insulin resistance related to white, beige and brown adipocytes. Mol. Metab. 2020;in press. doi:https://doi.org/10.1016/j.molmet.2019.12.014.
  70. Czech MP. Insulin action and resistance in obesity and type 2 diabetes. Nat. Med. 2017;23(7):804–814.
  71. Gandotra S, Le Dour C, Bottomley W, Cervera P, Giral P, Reznik Y, Charpentier G, Auclair M, Delépine M, Barroso I, Semple RK, Lathrop M, Lascols O, Capeau J, O’Rahilly S, Magré J, Savage DB, Vigouroux C.Perilipin deficiency and autosomal dominant partial lipodystrophy. N. Engl. J. Med. 2011;364(8):740–8.
  72. Rubio-Cabezas O, Puri V, Murano I, Saudek V, Semple RK, Dash S, Hyden CSS, Bottomley W, Vigouroux C, Magré J, Raymond-Barker P, Murgatroyd PR, Chawla A, Skepper JN, Chatterjee VK, Suliman S, Patch A-M, Agarwal AK, Garg A, Barroso I, Cinti S, Czech MP, Argente J, O’Rahilly S, Savage DB, LD Screening Consortium LS. Partial lipodystrophy and insulin resistant diabetes in a patient with a homozygous nonsense mutation in CIDEC. EMBO Mol. Med. 2009;1(5):280–7.
  73. Zhou L, Park S-Y, Xu L, Xia X, Ye J, Su L, Jeong K-H, Hur JH, Oh H, Tamori Y, Zingaretti CM, Cinti S, Argente J, Yu M, Wu L, Ju S, Guan F, Yang H, Choi CS, Savage DB, Li P. Insulin resistance and white adipose tissue inflammation are uncoupled in energetically challenged Fsp27-deficient mice. Nat. Commun. 2015;6:5949.
  74. Czech MP, Tencerova M, Pedersen DJ, Aouadi M. Insulin signalling mechanisms for triacylglycerol storage. Diabetologia 2013;56(5):949–964.
  75. Fasshauer M, Blüher M. Adipokines in health and disease. Trends Pharmacol. Sci. 2015;36(7):461–470.
  76. INGALLS AM, DICKIE MM, SNELL GD. Obese, a new mutation in the house mouse. J. Hered.1950;41(12):317–318.
  77. Lindström P. The physiology of obese-hyperglycemic mice [ob/ob mice]. ScientificWorldJournal. 2007;7:666–85.
  78. Hausberger FX. Parabiosis and transplantation experiments in hereditary obese mice. Anat Rec 1958;130:313 abstr.
  79. Friedman JM, Halaas JL. Leptin and the regulation of body weight in mammals. Nature 1998;395(6704):763–770.
  80. Hummel KP, Dickie MM, Coleman DL. Diabetes, a new mutation in the mouse. Science 1966;153(3740):1127–1128.
  81. Coleman DL. Obese and diabetes: two mutant genes causing diabetes-obesity syndromes in mice. Diabetologia 1978;14(3):141–148.
  82. Harris RB. Loss of body fat in lean parabiotic partners of ob/ob mice. Am. J. Physiol. 1997;272(6 Pt 2):R1809–R1815.
  83. Harris RBS. Parabiosis between db/db and ob/ob or db/+ mice. Endocrinology 1999;140(1):138–145.
  84. Bahary N, Leibel RL, Joseph L, Friedman JM. Molecular mapping of the mouse db mutation. Proc. Natl. Acad. Sci. U. S. A. 1990;87(21):8642–8646.
  85. Tartaglia LA, Dembski M, Weng X, Deng N, Culpepper J, Devos R, Richards GJ, Campfield LA, Clark FT, Deeds J, Muir C, Sanker S, Moriarty A, Moore KJ, Smutko JS, Mays GG, Wool EA, Monroe CA, Tepper RI.Identification and expression cloning of a leptin receptor, OB-R. Cell 1995;83(7):1263–1271.
  86. Peelman F, Zabeau L, Moharana K, Savvides SN, Tavernier J. 20 years of leptin: insights into signaling assemblies of the leptin receptor. J. Endocrinol. 2014;223(1):T9-23.
  87. Münzberg H, Morrison CD. Structure, production and signaling of leptin. Metabolism. 2015;64(1):13–23.
  88. Wauman J, Tavernier J. Leptin receptor signaling: pathways to leptin resistance. Front. Biosci. (Landmark Ed.2011;16:2771–2793.
  89. Guo K, McMinn JE, Ludwig T, Yu Y-H, Yang G, Chen L, Loh D, Li C, Chua S, Zhang Y. Disruption of peripheral leptin signaling in mice results in hyperleptinemia without associated metabolic abnormalities. Endocrinology2007;148(8):3987–3997.
  90. Frederich RC, Hamann A, Anderson S, Löllmann B, Lowell BB, Flier JS. Leptin levels reflect body lipid content in mice: Evidence for diet-induced resistance to leptin action. Nat. Med. 1995;1(12):1311–1314.
  91. Maffei M, Halaas J, Ravussin E, Pratley RE, Lee GH, Zhang Y, Fei H, Kim S, Lallone R, Ranganathan S, Kern PA, Friedman JM. Leptin levels in human and rodent: Measurement of plasma leptin and ob RNA in obese and weight-reduced subjects. Nat. Med. 1995;1(11):1155–1161.
  92. Enriori PJ, Evans AE, Sinnayah P, Jobst EE, Tonelli-Lemos L, Billes SK, Glavas MM, Grayson BE, Perello M, Nillni EA, Grove KL, Cowley MA. Diet-induced obesity causes severe but reversible leptin resitance in arcuate melanocortin neurons. Cell Metab. 2007;5(3):181–194.
  93. Knight ZA, Hannan KS, Greenberg ML, Friedman JM. Hyperleptinemia Is Required for the Development of Leptin Resistance. Stadler K, ed. PLoS One 2010;5(6):e11376.
  94. Balland E, Cowley MA. New insights in leptin resistance mechanisms in mice. Front. Neuroendocrinol.2015;39:59–65.
  95. Friedman J. 20 years of leptin: leptin at 20: an overview. J. Endocrinol. 2014;223(1):T1-8.
  96. Denroche HC, Huynh FK, Kieffer TJ. The role of leptin in glucose homeostasis. J. Diabetes Investig.2012;3(2):115–129.
  97. Ducy P, Amling M, Takeda S, Priemel M, Schilling AF, Beil FT, Shen J, Vinson C, Rueger JM, Karsenty G. Leptin inhibits bone formation through a hypothalamic relay: a central control of bone mass. Cell 2000;100(2):197–207.
  98. Dalamaga M, Chou SH, Shields K, Papageorgiou P, Polyzos SA, Mantzoros CS. Leptin at the intersection of neuroendocrinology and metabolism: current evidence and therapeutic perspectives. Cell Metab.2013;18(1):29–42.
  99. Chen XX, Yang T. Roles of leptin in bone metabolism and bone diseases. J. Bone Miner. Metab.2015;33(5):474–485.
  100. Sáinz N, Barrenetxe J, Moreno-Aliaga MJ, Martínez JA. Leptin resistance and diet-induced obesity: central and peripheral actions of leptin. Metabolism 2015;64(1):35–46.
  101. Harris RBS. Direct and indirect effects of leptin on adipocyte metabolism. Biochim. Biophys. Acta2014;1842(3):414–423.
  102. Ghosh S, Mukhopadhyay P, Pandit K, Chowdhury S, Dutta D. Leptin and cancer: pathogenesis and modulation. Indian J. Endocrinol. Metab. 2012;16(9):596.
  103. Paik SS, Jang S-M, Jang K-S, Lee KH, Choi D, Jang SJ. Leptin expression correlates with favorable clinicopathologic phenotype and better prognosis in colorectal adenocarcinoma. Ann. Surg. Oncol.2009;16(2):297–303.
  104. Kim HH, Kim YS, Kang YK, Moon JS. Leptin and peroxisome proliferator-activated receptor γ expression in colorectal adenoma. World J. Gastroenterol. 2012;18(6):557–562.
  105. Akinci M, Kosova F, Cetin B, Aslan S, Ari Z, Cetin A. Leptin levels in thyroid cancer. Asian J. Surg.2009;32(4):216–223.
  106. Uddin S, Bavi P, Siraj AK, Ahmed M, Al-Rasheed M, Hussain AR, Ahmed M, Amin T, Alzahrani A, Al-Dayel F, Abubaker J, Bu R, Al-Kuraya KS. Leptin-R and its association with PI3K/AKT signaling pathway in papillary thyroid carcinoma. Endocr. Relat. Cancer 2010;17(1):191–202.
  107. Petridou E, Belechri M, Dessypris N, Koukoulomatis P, Diakomanolis E, Spanos E, Trichopoulos D. Leptin and body mass index in relation to endometrial cancer risk. Ann. Nutr. Metab. 2002;46(3–4):147–151.
  108. Yuan S-SF, Tsai K-B, Chung Y-F, Chan T-F, Yeh Y-T, Tsai L-Y, Su J-H. Aberrant expression and possible involvement of the leptin receptor in endometrial cancer. Gynecol. Oncol. 2004;92(3):769–775.
  109. Cymbaluk A, Chudecka-Głaz A, Rzepka-Górska I. Leptin levels in serum depending on body mass index in patients with endometrial hyperplasia and cancer. Eur. J. Obstet. Gynecol. Reprod. Biol. 2008;136(1):74–77.
  110. Munsell MF, Sprague BL, Berry DA, Chisholm G, Trentham-Dietz A. Body mass index and breast cancer risk according to postmenopausal estrogen-progestin use and hormone receptor status. Epidemiol. Rev.2014;36(1):114–136.
  111. Artac M, Altundag K. Leptin and breast cancer: an overview. Med. Oncol. 2012;29(3):1510–1514.
  112. Scherer PE, Williams S, Fogliano M, Baldini G, Lodish HF. A novel serum protein similar to C1q, produced exclusively in adipocytes. J. Biol. Chem. 1995;270(45):26746–26749.
  113. Nakano Y, Tobe T, Choi-Miura N-H, Mazda T, Tomita M. Isolation and characterization of GBP28, a novel gelatin-binding protein purified from human plasma. J. Biochem. 1996;120(4):803–812.
  114. Maeda K, Okubo K, Shimomura I, Funahashi T, Matsuzawa Y, Matsubara K. cDNA cloning and expression of a novel adipose specific collagen-like factor, apM1 (adiposemost abundant gene transcript 1). Biochem. Biophys. Res. Commun. 1996;221(2):286–289.
  115. Hu E, Liang P, Spiegelman BM. AdipoQ is a novel adipose-specific gene dysregulated in obesity. J. Biol. Chem.1996;271(18):10697–10703.
  116. Turer AT, Scherer PE. Adiponectin: Mechanistic insights and clinical implications. Diabetologia2012;55(9):2319–2326.
  117. Ruan H, Dong LQ. Adiponectin signaling and function in insulin target tissues. J. Mol. Cell Biol. 2016;8(2):101–109.
  118. Wang Z V., Scherer PE. Adiponectin, the past two decades. J. Mol. Cell Biol. 2016;8(2):93–100.
  119. Waki H, Yamauchi T, Kamon J, Kita S, Ito Y, Hada Y, Uchida S, Tsuchida A, Takekawa S, Kadowaki T.Generation of globular fragment of adiponectin by leukocyte elastase secreted by monocytic cell line THP-1. Endocrinology 2005;146(2):790–796.
  120. Fruebis J, Tsao TS, Javorschi S, Ebbets-Reed D, Erickson MR, Yen FT, Bihain BE, Lodish HF. Proteolytic cleavage product of 30-kDa adipocyte complement-related protein increases fatty acid oxidation in muscle and causes weight loss in mice. Proc. Natl. Acad. Sci. 2001;98(4):2005–2010.
  121. Pajvani UB, Hawkins M, Combs TP, Rajala MW, Doebber T, Berger JP, Wagner JA, Wu M, Knopps A, Xiang AH, Utzschneider KM, Kahn SE, Olefsky JM, Buchanan TA, Scherer PE. Complex distribution, not absolute amount of adiponectin, correlates with thiazolidinedione-mediated improvement in insulin sensitivity. J. Biol. Chem. 2004;279(13):12152–12162.
  122. Yamauchi T, Kamon J, Ito Y, Tsuchida A, Yokomizo T, Kita S, Sugiyama T, Miyagishi M, Hara K, Tsunoda M, Murakami K, Ohteki T, Uchida S, Takekawa S, Waki H, Tsuno NH, Shibata Y, Terauchi Y, Froguel P, Tobe K, Koyasu S, Taira K, Kitamura T, Shimizu T, Nagai R, Kadowaki T. Cloning of adiponectin receptors that mediate antidiabetic metabolic effects. Nature 2003;423(6941):762–769.
  123. Hug C, Wang J, Ahmad NS, Bogan JS, Tsao T-S, Lodish HF. T-cadherin is a receptor for hexameric and high-molecular-weight forms of Acrp30/adiponectin. Proc. Natl. Acad. Sci. 2004;101(28):10308–10313.
  124. Yamauchi T, Nio Y, Maki T, Kobayashi M, Takazawa T, Iwabu M, Okada-Iwabu M, Kawamoto S, Kubota N, Kubota T, Ito Y, Kamon J, Tsuchida A, Kumagai K, Kozono H, Hada Y, Ogata H, Tokuyama K, Tsunoda M, Ide T, Murakami K, Awazawa M, Takamoto I, Froguel P, Hara K, Tobe K, Nagai R, Ueki K, Kadowaki T. Targeted disruption of AdipoR1 and AdipoR2 causes abrogation of adiponectin binding and metabolic actions. Nat. Med.2007;13(3):332–339.
  125. Yamauchi T, Iwabu M, Okada-Iwabu M, Kadowaki T. Adiponectin receptors: a review of their structure, function and how they work. Best Pract. Res. Clin. Endocrinol. Metab. 2014;28(1):15–23.
  126. Chan DW, Lee JMF, Chan PCY, Ng IOL. Genetic and epigenetic inactivation of T‐cadherin in human hepatocellular carcinoma cells. Int. J. Cancer 2008;123(5):1043–1052.
  127. Vestal DJ, Ranscht B. Glycosyl phosphatidylinositol--anchored T-cadherin mediates calcium-dependent, homophilic cell adhesion. J. Cell Biol. 1992;119(2):451–461.
  128. Matsuda K, Fujishima Y, Maeda N, Mori T, Hirata A, Sekimoto R, Tsushima Y, Masuda S, Yamaoka M, Inoue K, Nishizawa H, Kita S, Ranscht B, Funahashi T, Shimomura I. Positive feedback regulation between adiponectin and t-cadherin Iimpacts adiponectin levels in tissue and Plasma of male mice. Endocrinology 2015;156(3):934–946.
  129. Fujishima Y, Maeda N, Matsuda K, Masuda S, Mori T, Fukuda S, Sekimoto R, Yamaoka M, Obata Y, Kita S, Nishizawa H, Funahashi T, Ranscht B, Shimomura I. Adiponectin association with T-cadherin protects against neointima proliferation and atherosclerosis. FASEB J. 2017;31(4):1571–1583.
  130. Denzel MS, Scimia M-C, Zumstein PM, Walsh K, Ruiz-Lozano P, Ranscht B. T-cadherin is critical for adiponectin-mediated cardioprotection in mice. J. Clin. Invest. 2010;120(12):4342–4352.
  131. Parker-Duffen JL, Nakamura K, Silver M, Kikuchi R, Tigges U, Yoshida S, Denzel MS, Ranscht B, Walsh K. T-cadherin Is Essential for Adiponectin-mediated Revascularization. J. Biol. Chem. 2013;288(34):24886–24897.
  132. Brooks NL, Trent CM, Raetzsch CF, Flurkey K, Boysen G, Perfetti MT, Jeong Y-C, Klebanov S, Patel KB, Khodush VR, Kupper LL, Carling D, Swenberg JA, Harrison DE, Combs TP. Low utilization of circulating glucose after food withdrawal in Snell dwarf mice. J. Biol. Chem. 2007;282(48):35069–35077.
  133. Combs TP, Marliss EB. Adiponectin signaling in the liver. Rev. Endocr. Metab. Disord. 2014;15(2):137–147.
  134. Towler MC, Hardie DG. AMP-activated protein kinase in metabolic control and insulin signaling. Circ. Res.2007;100(3):328–341.
  135. Pirvulescu MM, Gan AM, Stan D, Simion V, Calin M, Butoi E, Manduteanu I. Subendothelial resistin enhances monocyte transmigration in a co-culture of human endothelial and smooth muscle cells by mechanisms involving fractalkine, MCP-1 and activation of TLR4 and Gi/o proteins signaling. Int. J. Biochem. Cell Biol. 2014;50:29–37.
  136. Kadowaki T, Yamauchi T, Waki H, Iwabu M, Okada-Iwabu M, Nakamura M. Adiponectin, adiponectin receptors, and epigenetic regulation of adipogenesis. Cold Spring Harb. Symp. Quant. Biol. 2011;76(0):257–265.
  137. Ruscica M, Steffani L, Magni P. Adiponectin interactions in bone and cartilage biology and disease. In: Vitamins and hormones.Vol 90.; 2012:321–339.
  138. Dadson K, Liu Y, Sweeney G. Adiponectin action: a combination of endocrine and autocrine/paracrine effects.Front. Endocrinol. (Lausanne). 2011;2. doi:10.3389/fendo.2011.00062.
  139. Santos E Dos, Pecquery R, Mazancourt P de, Dieudonné M-N. Adiponectin and Reproduction. In: Vitamins and hormones.Vol 90.; 2012:187–209.
  140. Čikoš Š. Adiponectin and its receptors in preimplantation embryo development. In: Vitamins and hormones.Vol 90.; 2012:211–238.
  141. Combs TP, Berg AH, Obici S, Scherer PE, Rossetti L. Endogenous glucose production is inhibited by the adipose-derived protein Acrp30. J. Clin. Invest. 2001;108(12):1875–1881.
  142. Nawrocki AR, Rajala MW, Tomas E, Pajvani UB, Saha AK, Trumbauer ME, Pang Z, Chen AS, Ruderman NB, Chen H, Rossetti L, Scherer PE. Mice lacking adiponectin show decreased hepatic insulin sensitivity and reducedresponsiveness to proliferator-activated receptor γ agonists. J. Biol. Chem. 2006;281(5):2654–2660.
  143. Combs TP, Pajvani UB, Berg AH, Lin Y, Jelicks LA, Laplante M, Nawrocki AR, Rajala MW, Parlow AF, Cheeseboro L, Ding Y-Y, Russell RG, Lindemann D, Hartley A, Baker GRC, Obici S, Deshaies Y, Ludgate M, Rossetti L, Scherer PE. A transgenic mouse with a deletion in the collagenous domain of adiponectin displays elevated circulating adiponectin and improved insulin sensitivity. Endocrinology 2004;145(1):367–383.
  144. Kubota N, Terauchi Y, Kubota T, Kumagai H, Itoh S, Satoh H, Yano W, Ogata H, Tokuyama K, Takamoto I, Mineyama T, Ishikawa M, Moroi M, Sugi K, Yamauchi T, Ueki K, Tobe K, Noda T, Nagai R, Kadowaki T.Pioglitazone ameliorates insulin resistance and diabetes by b adiponectin-dependent and -independent pathways. J. Biol. Chem. 2006;281(13):8748–8755.
  145. Yan W, Zhang H, Liu P, Wang H, Liu J, Gao C, Liu Y, Lian K, Yang L, Sun L, Guo Y, Zhang L, Dong L, Lau WB, Gao E, Gao F, Xiong L, Wang H, Qu Y, Tao L. Impaired mitochondrial biogenesis due to dysfunctional adiponectin-AMPK-PGC-1α signaling contributing to increased vulnerability in diabetic heart. Basic Res. Cardiol.2013;108(3):329.
  146. Yamauchi T, Kamon J, Waki H, Terauchi Y, Kubota N, Hara K, Mori Y, Ide T, Murakami K, Tsuboyama-Kasaoka N, Ezaki O, Akanuma Y, Gavrilova O, Vinson C, Reitman ML, Kagechika H, Shudo K, Yoda M, Nakano Y, Tobe K, Nagai R, Kimura S, Tomita M, Froguel P, Kadowaki T. The fat-derived hormone adiponectin reverses insulin resistance associated with both lipoatrophy and obesity. Nat. Med. 2001;7(8):941–946.
  147. Pagano C, Soardo G, Esposito W, Fallo F, Basan L, Donnini D, Federspil G, Sechi LA, Vettor R. Plasma adiponectin is decreased in nonalcoholic fatty liver disease. Eur. J. Endocrinol. 2005;152(1):113–118.
  148. Buechler C, Wanninger J, Neumeier M. Adiponectin, a key adipokine in obesity related liver diseases. World J. Gastroenterol. 2011;17(23):2801–2811.
  149. Thiebaud D, Jacot E, DeFronzo RA, Maeder E, Jequier E, Felber JP. The effect of graded doses of insulin on total glucose uptake, glucose oxidation, and glucose storage in man. Diabetes 1982;31(11):957–963.
  150. Pendergrass M, Bertoldo A, Bonadonna R, Nucci G, Mandarino L, Cobelli C, DeFronzo RA. Muscle glucose transport and phosphorylation in type 2 diabetic, obese nondiabetic, and genetically predisposed individuals. Am. J. Physiol. Metab. 2007;292(1):E92–E100.
  151. Ceddia RB, Somwar R, Maida A, Fang X, Bikopoulos G, Sweeney G. Globular adiponectin increases GLUT4 translocation and glucose uptake but reduces glycogen synthesis in rat skeletal muscle cells. Diabetologia2005;48(1):132–139.
  152. Yamauchi T, Kamon J, Minokoshi Y, Ito Y, Waki H, Uchida S, Yamashita S, Noda M, Kita S, Ueki K, Eto K, Akanuma Y, Froguel P, Foufelle F, Ferre P, Carling D, Kimura S, Nagai R, Kahn BB, Kadowaki T. Adiponectin stimulates glucose utilization and fatty-acid oxidation by activating AMP-activated protein kinase. Nat. Med.2002;8(11):1288–1295.
  153. Yoon MJ, Lee GY, Chung J-J, Ahn YH, Hong SH, Kim JB. Adiponectin increases fatty acid oxidation in skeletal muscle cells by sequential activation of AMP-activated protein kinase, p38 mitogen-activated protein kinase, and peroxisome proliferator-activated receptor alpha. Diabetes 2006;55(9):2562–2570.
  154. Wang C, Mao X, Wang L, Liu M, Wetzel MD, Guan K-L, Dong LQ, Liu F. Adiponectin sensitizes insulin signaling by reducing p70 S6 kinase-mediated serine phosphorylation of IRS-1. J. Biol. Chem. 2007;282(11):7991–7996.
  155. Yano W, Kubota N, Itoh S, Kubota T, Awazawa M, Moroi M, Sugi K, Takamoto I, Ogata H, Tokuyama K, Noda T, Terauchi Y, Ueki K, Kadowaki T. Molecular mechanism of moderate insulin resistance in adiponectin-knockout mice. Endocr. J. 2008;55(3):515–522.
  156. Kandasamy AD, Sung MM, Boisvenue JJ, Barr AJ, Dyck JRB. Adiponectin gene therapy ameliorates high-fat, high-sucrose diet-induced metabolic perturbations in mice. Nutr. Diabetes 2012;2(9):e45–e45.
  157. Chen MB, McAinch AJ, Macaulay SL, Castelli LA, O’Brien PE, Dixon JB, Cameron-Smith D, Kemp BE, Steinberg GR. Impaired activation of AMP-kinase and fatty acid oxidation by globular adiponectin in cultured human skeletal muscle of obese Type 2 diabetics. J. Clin. Endocrinol. Metab. 2005;90(6):3665–3672.
  158. Kumada M, Kihara S, Sumitsuji S, Kawamoto T, Matsumoto S, Ouchi N, Arita Y, Okamoto Y, Shimomura I, Hiraoka H, Nakamura T, Funahashi T, Matsuzawa Y, Osaka CAD Study Group. Coronary artery disease.Association of hypoadiponectinemia with coronary artery disease in men. Arterioscler. Thromb. Vasc. Biol.2003;23(1):85–89.
  159. Ding M, Rzucidlo EM, Davey JC, Xie Y, Liu R, Jin Y, Stavola L, Martin KA. Adiponectin in the heart and vascular system. In: Vitamins and hormones.Vol 90.; 2012:289–319.
  160. Nanayakkara G, Kariharan T, Wang L, Zhong J, Amin R. The cardio-protective signaling and mechanisms of adiponectin. Am. J. Cardiovasc. Dis. 2012;2(4):253–266.
  161. Okada-Iwabu M, Yamauchi T, Iwabu M, Honma T, Hamagami K, Matsuda K, Yamaguchi M, Tanabe H, Kimura-Someya T, Shirouzu M, Ogata H, Tokuyama K, Ueki K, Nagano T, Tanaka A, Yokoyama S, Kadowaki T. A small-molecule AdipoR agonist for type 2 diabetes and short life in obesity. Nature 2013;503(7477):493–499.
  162. Choi SR, Lim JH, Kim MY, Kim EN, Kim Y, Choi BS, Kim Y-S, Kim HW, Lim K-M, Kim MJ, Park CW. Adiponectin receptor agonist AdipoRon decreased ceramide, and lipotoxicity, and ameliorated diabetic nephropathy. Metabolism 2018. doi:10.1016/j.metabol.2018.02.004.
  163. Steppan CM, Bailey ST, Bhat S, Brown EJ, Banerjee RR, Wright CM, Patel HR, Ahima RS, Lazar MA. The hormone resistin links obesity to diabetes. Nature 2001;409(6818):307–312.
  164. Kim KH, Lee K, Moon YS, Sul HS. A cysteine-rich adipose tissue-specific secretory factor inhibits adipocyte differentiation. J. Biol. Chem. 2001;276(14):11252–11256.
  165. Blagoev B, Kratchmarova I, Nielsen MM, Fernandez MM, Voldby J, Andersen JS, Kristiansen K, Pandey A, Mann M. Inhibition of adipocyte differentiation by resistin-like molecule α. J. Biol. Chem. 2002;277(44):42011–42016.
  166. Oliver P, Picó C, Serra F, Palou A. Resistin expression in different adipose tissue depots during rat development. Mol. Cell. Biochem. 2003;252(1/2):397–400.
  167. Rajala MW, Qi Y, Patel HR, Takahashi N, Banerjee R, Pajvani UB, Sinha MK, Gingerich RL, Scherer PE, Ahima RS. Regulation of resistin expression and circulating levels in obesity, diabetes, and fasting. Diabetes2004;53(7):1671–1679.
  168. Lefterova MI, Mullican SE, Tomaru T, Qatanani M, Schupp M, Lazar MA. Endoplasmic reticulum stress regulates adipocyte resistin expression. Diabetes 2009;58(8):1879–1886.
  169. Patel L, Buckels AC, Kinghorn IJ, Murdock PR, Holbrook JD, Plumpton C, Macphee CH, Smith SA. Resistin is expressed in human macrophages and directly regulated by PPARγ activators. Biochem. Biophys. Res. Commun. 2003;300(2):472–476.
  170. Qatanani M, Szwergold NR, Greaves DR, Ahima RS, Lazar MA. Macrophage-derived human resistin exacerbates adipose tissue inflammation and insulin resistance in mice. J. Clin. Invest. 2009;119(3):531–539.
  171. Savage DB, Sewter CP, Klenk ES, Segal DG, Vidal-Puig A, Considine R V, O’Rahilly S. Resistin / Fizz3 expression in relation to obesity and peroxisome proliferator-activated receptor-gamma action in humans. Diabetes 2001;50(10):2199–2202.
  172. Tarkowski A, Bjersing J, Shestakov A, Bokarewa MI. Resistin competes with lipopolysaccharide for binding to toll-like receptor 4. J. Cell. Mol. Med. 2010;14(6B):1419–1431.
  173. Lee S, Lee HC, Kwon YW, Lee SE, Cho Y, Kim J, Lee S, Kim JY, Lee J, Yang HM, Mook-Jung I, Nam KY, Chung J, Lazar MA, Kim HS. Adenylyl cyclase-associated protein 1 is a receptor for human resistin and mediates inflammatory actions of human monocytes. Cell Metab. 2014;19(3):484–497.
  174. Jiang S, Park DW, Tadie J-M, Gregoire M, Deshane J, Pittet JF, Abraham E, Zmijewski JW. Human resistin promotes neutrophil proinflammatory activation and neutrophil extracellular trap formation and increases severity of acute lung injury. J. Immunol. 2014;192(10):4795–4803.
  175. Banerjee RR, Rangwala SM, Shapiro JS, Rich AS, Rhoades B, Qi Y, Wang J, Rajala MW, Pocai A, Scherer PE, Steppan CM, Ahima RS, Obici S, Rossetti L, Lazar MA. Regulation of fasted blood glucose by resistin. Science2004;303(5661):1195–1198.
  176. Steppan CM, Brown EJ, Wright CM, Bhat S, Banerjee RR, Dai CY, Enders GH, Silberg DG, Wen X, Wu GD, Lazar MA. A family of tissue-specific resistin-like molecules. Proc. Natl. Acad. Sci. 2001;98(2):502–506.
  177. Graveleau C, Zaha VG, Mohajer A, Banerjee RR, Dudley-Rucker N, Steppan CM, Rajala MW, Scherer PE, Ahima RS, Lazar MA, Abel ED. Mouse and human resistins impair glucose transport in primary mouse cardiomyocytes, and oligomerization Is required for this biological action. J. Biol. Chem. 2005;280(36):31679–31685.
  178. Qi Y, Nie Z, Lee Y-S, Singhal NS, Scherer PE, Lazar MA, Ahima RS. Loss of resistin improves glucose homeostasis in leptin deficiency. Diabetes 2006;55(11):3083–3090.
  179. Singhal NS, Lazar MA, Ahima RS. Central resistin induces hepatic insulin resistance via neuropeptide Y. J. Neurosci. 2007;27(47):12924–12932.
  180. Qatanani M, Szwergold NR, Greaves DR, Ahima RS, Lazar MA. Macrophage-derived human resistin exacerbates adipose tissue inflammation and insulin resistance in mice. J Clin Invest 2009;119(3):531–539.
  181. Schwartz DR, Lazar MA. Human resistin: found in translation from mouse to man. Trends Endocrinol. Metab.2011;22(7):259–265.
  182. Silha J V., Murphy LJ. Serum resistin (FIZZ3) protein is increased in obese humans. J. Clin. Endocrinol. Metab.2004;89(4):1977–1977.
  183. Degawa-Yamauchi M, Bovenkerk JE, Juliar BE, Watson W, Kerr K, Jones R, Zhu Q, Considine R V. Serum resistin (FIZZ3) protein is increased in obese humans. J. Clin. Endocrinol. Metab. 2003;88(11):5452–5455.
  184. BAKER JF, MORALES M, QATANANI M, CUCCHIARA A, NACKOS E, LAZAR MA, TEFF K, VON FELDT JM.Resistin levels in lupus and associations with disease-specific measures, insulin resistance, and coronary calcification. J. Rheumatol. 2011;38(11):2369–2375.
  185. Reilly MP, Lehrke M, Wolfe ML, Rohatgi A, Lazar MA, Rader DJ. Resistin is an inflammatory marker of atherosclerosis in humans. Circulation 2005;111(7):932–939.
  186. Filková M, Haluzík M, Gay S, Šenolt L. The role of resistin as a regulator of inflammation: Implications for various human pathologies. Clin. Immunol. 2009;133(2):157–170.
  187. Ding Q, White SP, Ling C, Zhou W. Resistin and cardiovascular disease. Trends Cardiovasc. Med.2011;21(1):20–27.
  188. Lee SE, Kim H-S. Human resistin in cardiovascular disease. J. Smooth Muscle Res. 2012;48(1):27–35.
  189. Saetang J, Sangkhathat S. Role of innate lymphoid cells in obesity and metabolic disease (review). Mol. Med. Rep. 2018;17(1):1403–1412.
  190. Hotamisligil GS, Shargill NS, Spiegelman BM. Adipose expression of tumor necrosis factor-alpha: direct role in obesity-linked insulin resistance. Science 1993;259(5091):87–91.
  191. Weisberg SP, McCann D, Desai M, Rosenbaum M, Leibel RL, Ferrante AW. Obesity is associated with macrophage accumulation in adipose tissue. J. Clin. Invest. 2003;112(12):1796–1808.
  192. Sarjeant K, Stephens JM. Adipogenesis. Cold Spring Harb. Perspect. Biol. 2012;4(9):1–19.
  193. Mathieu PS, Loboa EG. Cytoskeletal and focal adhesion influences on mesenchymal stem cell shape, mechanical properties, and differentiation down osteogenic, adipogenic, and chondrogenic pathways. Tissue Eng. Part B. Rev. 2012;18(6):436–444.
  194. Poulos SP, Dodson M V, Culver MF, Hausman GJ. The increasingly complex regulation of adipocyte differentiation. Exp. Biol. Med. (Maywood). 2016;241(5):449–456.
  195. Engin A. The pathogenesis of obesity-associated adipose tissue inflammation. In: Advances in experimental medicine and biology.Vol 960.; 2017:221–245.
  196. Chen C, Cui Q, Zhang X, Luo X, Liu Y, Zuo J, Peng Y. Long non-coding RNAs regulation in adipogenesis and lipid metabolism: Emerging insights in obesity. Cell. Signal. 2018;51:47–58.
  197. Rosen ED, Sarraf P, Troy AE, Bradwin G, Moore K, Milstone DS, Spiegelman BM, Mortensen RM. PPARγ is required for the differentiation of adipose tissue in vivo and in vitro. Mol. Cell 1999;4(4):611–617.
  198. He W, Barak Y, Hevener A, Olson P, Liao D, Le J, Nelson M, Ong E, Olefsky JM, Evans RM. Adipose-specific peroxisome proliferator-activated receptor gamma knockout causes insulin resistance in fat and liver but not in muscle. Proc. Natl. Acad. Sci. U. S. A. 2003;100(26):15712–15717.
  199. Barroso I, Gurnell M, Crowley VEF, Agostini M, Schwabe JW, Soos MA, Maslen GL, Williams TDM, Lewis H, Schafer AJ, Chatterjee VKK, O’Rahilly S. Dominant negative mutations in human PPARγ associated with severe insulin resistance, diabetes mellitus and hypertension. Nature 1999;402(6764):880–883.
  200. Doney ASF, Fischer B, Leese G, Morris AD, Palmer CNA. Cardiovascular risk in type 2 diabetes is associated with variation at the PPARG locus. Arterioscler. Thromb. Vasc. Biol. 2004;24(12):2403–2407.
  201. Monajemi H, Zhang L, Li G, Jeninga EH, Cao H, Maas M, Brouwer CB, Kalkhoven E, Stroes E, Hegele RA, Leff T. Familial partial lipodystrophy phenotype resulting from a single-base mutation in deoxyribonucleic acid-binding domain of peroxisome proliferator-activated receptor-γ. J. Clin. Endocrinol. Metab. 2007;92(5):1606–1612.
  202. Ahmadian M, Suh JM, Hah N, Liddle C, Atkins AR, Downes M, Evans RM. PPARγ signaling and metabolism: the good, the bad and the future. Nat. Med. 2013;19(5):557–566.
  203. Rangwala SM, Lazar MA. The dawn of the SPPARMs? Sci. STKE 2002;2002(121):pe9.
  204. Higgins LS, Depaoli AM. Selective peroxisome proliferator-activated receptor γ (PPARγ) modulation as a strategy for safer therapeutic PPARγ activation. In: American Journal of Clinical Nutrition.; 2010. doi:10.3945/ajcn.2009.28449E.
  205. Wang QA, Zhang F, Jiang L, Ye R, An Y, Shao M, Tao C, Gupta RK, Scherer PE. Peroxisome Proliferator-Activated Receptor γ and Its Role in Adipocyte Homeostasis and Thiazolidinedione-Mediated Insulin Sensitization. Mol. Cell. Biol. 2018;38(10). doi:10.1128/MCB.00677-17.
  206. Itoh T, Fairall L, Amin K, Inaba Y, Szanto A, Balint BL, Nagy L, Yamamoto K, Schwabe JWR. Structural basis for the activation of PPARgamma by oxidized fatty acids. Nat. Struct. Mol. Biol. 2008;15(9):924–931.
  207. Waku T, Shiraki T, Oyama T, Maebara K, Nakamori R, Morikawa K. The nuclear receptor PPARγ individually responds to serotonin- and fatty acid-metabolites. EMBO J. 2010;29(19):3395–3407.
  208. Yanting C, Yang QY, Ma GL, Du M, Harrison JH, Block E. Dose- and type-dependent effects of long-chain fatty acids on adipogenesis and lipogenesis of bovine adipocytes. J. Dairy Sci. 2018;101(2):1601–1615.
  209. Kobayashi T, Fujimori K. Very long-chain-fatty acids enhance adipogenesis through coregulation of Elovl3 and PPARγ in 3T3-L1 cells. Am. J. Physiol. Metab. 2012;302(12):E1461–E1471.
  210. Lee Y-H, Kim S-N, Kwon H-J, Maddipati KR, Granneman JG. Adipogenic role of alternatively activated macrophages in β-adrenergic remodeling of white adipose tissue. Am. J. Physiol. Regul. Integr. Comp. Physiol.2016;310(1):R55-65.
  211. Baker PRS, Lin Y, Schopfer FJ, Woodcock SR, Groeger AL, Batthyany C, Sweeney S, Long MH, Iles KE, Baker LMS, Branchaud BP, Chen YE, Freeman BA. Fatty acid transduction of nitric oxide signaling: multiple nitrated unsaturated fatty acid derivatives exist in human blood and urine and serve as endogenous peroxisome proliferator-activated receptor ligands. J. Biol. Chem. 2005;280(51):42464–75.
  212. Fujimori K, Urade Y. Transcriptional Regulation in Adipogenesis Through PPARγ-Dependent and -Independent Mechanisms by Prostaglandins. In: Humana Press, New York, NY; 2014:177–196.
  213. Fujimori K. Prostaglandins as PPARγ Modulators in Adipogenesis. PPAR Res. 2012;2012:527607.
  214. Madsen L, Petersen RK, Kristiansen K. Regulation of adipocyte differentiation and function by polyunsaturated fatty acids. Biochim. Biophys. Acta - Mol. Basis Dis. 2005;1740(2):266–286.
  215. Hamm JK, Park BH, Farmer SR. A role for C/EBPβ in regulating peroxisome proliferator-activated receptor γ activity during adipogenesis in 3T3-L1 preadipocytes. J. Biol. Chem. 2001;276(21):18464–18471.
  216. Cao Z, Umek RM, McKnight SL. Regulated expression of three C/EBP isoforms during adipose conversion of 3T3-L1 cells. Genes Dev. 1991;5(9):1538–1352.
  217. Chen Z, Torrens JI, Anand A, Spiegelman BM, Friedman JM. Krox20 stimulates adipogenesis via C/EBPβ-dependent and -independent mechanisms. Cell Metab. 2005;1(2):93–106.
  218. Du C, Ma X, Meruvu S, Hugendubler L, Mueller E. The adipogenic transcriptional cofactor ZNF638 interacts with splicing regulators and influences alternative splicing. J. Lipid Res. 2014;55(9):1886–1896.
  219. Stark GR, Darnell JE, Jr. The JAK-STAT pathway at twenty. Immunity 2012;36(4):503–514.
  220. Darnell JE. STATs and gene regulation. Science 1997;277(5332):1630–1635.
  221. Harp JB, Franklin D, Vanderpuije AA, Gimble JM. Differential expression of signal transducers and activators of transcription during human adipogenesis. Biochem. Biophys. Res. Commun. 2001;281(4):907–912.
  222. Stephens JM, Morrison RF, Pilch PF. The expression and regulation of STATs during 3T3-L1 adipocyte differentiation. J. Biol. Chem. 1996;271(18):10441–104444.
  223. Hua X, Wu J, Goldstein JL, Brown MS, Hobbs HH. Structure of the human gene encoding sterol regulatory element binding protein-1 (SREBF1) and localization of SREBF1 and SREBF2 to chromosomes 17p11.2 and 22q13. Genomics 1995;25(3):667–673.
  224. Shimomura I, Shimano H, Horton JD, Goldstein JL, Brown MS. Differential expression of exons 1a and 1c in mRNAs for sterol regulatory element binding protein-1 in human and mouse organs and cultured cells. J. Clin. Invest. 1997;99(5):838–845.
  225. Horton JD, Shimomura I, Brown MS, Hammer RE, Goldstein JL, Shimano H. Activation of cholesterol synthesis in preference to fatty acid synthesis in liver and adipose tissue of transgenic mice overproducing sterol regulatory element-binding protein-2. J. Clin. Invest. 1998;101(11):2331–2339.
  226. Price NL, Holtrup B, Kwei SL, Wabitsch M, Rodeheffer M, Bianchini L, Suárez Y, Fernández-Hernando C.SREBP-1c/MicroRNA 33b genomic loci control adipocyte differentiation. Mol. Cell. Biol. 2016;36(7):1180–1193.
  227. Yahagi N, Shimano H, Hasty AH, Matsuzaka T, Ide T, Yoshikawa T, Amemiya-Kudo M, Tomita S, Okazaki H, Tamura Y, Iizuka Y, Ohashi K, Osuga J-I, Harada K, Gotoda T, Nagai R, Ishibashi S, Yamada N. Absence of sterol regulatory element-binding protein-1 (SREBP-1) ameliorates fatty livers but not obesity or insulin resistance in Lep(ob)/Lep(ob) mice. J. Biol. Chem. 2002;277(22):19353–19357.
  228. Åkerblad P, Månsson R, Lagergren A, Westerlund S, Basta B, Lind U, Thelin A, Gisler R, Liberg D, Nelander S, Bamberg K, Sigvardsson M. Gene expression analysis suggests that EBF-1 and PPARγ2 induce adipogenesis of NIH-3T3 cells with similar efficiency and kinetics. Physiol. Genomics 2005;23(2):206–216.
  229. Jimenez MA, Akerblad P, Sigvardsson M, Rosen ED. Critical role for Ebf1 and Ebf2 in the adipogenic transcriptional cascade. Mol. Cell. Biol. 2007;27(2):743–757.
  230. Rajakumari S, Wu J, Ishibashi J, Lim H-W, Giang A-H, Won K-J, Reed RR, Seale P. EBF2 determines and maintains brown adipocyte identity. Cell Metab. 2013;17(4):562–574.
  231. Stine RR, Shapira SN, Lim H-W, Ishibashi J, Harms M, Won K-J, Seale P. EBF2 promotes the recruitment of beige adipocytes in white adipose tissue. Mol. Metab. 2016;5(1):57–65.
  232. Eguchi J, Yan Q-W, Schones DE, Kamal M, Hsu C-H, Zhang MQ, Crawford GE, Rosen ED. Interferon regulatory factors are transcriptional regulators of adipogenesis. Cell Metab. 2008;7(1):86–94.
  233. Kumari M, Wang X, Lantier L, Lyubetskaya A, Eguchi J, Kang S, Tenen D, Roh HC, Kong X, Kazak L, Ahmad R, Rosen ED. IRF3 promotes adipose inflammation and insulin resistance and represses browning. J. Clin. Invest. 2016;126(8):2839–54.
  234. Kennell JA, MacDougald OA. Wnt signaling inhibits adipogenesis through beta-catenin-dependent and -independent mechanisms. J. Biol. Chem. 2005;280(25):24004–24010.
  235. Rao TP, Kühl M. An updated overview on wnt signaling pathways. Circ. Res. 2010;106(12):1798–1806.
  236. Cadigan KM, Nusse R. Wnt signaling: a common theme in animal development. Genes Dev. 1997;11(24):3286–2305.
  237. Chen N, Wang J. Wnt/β-catenin signaling and obesity. Front. Physiol. 2018;9:792.
  238. Christodoulides C, Scarda A, Granzotto M, Milan G, Dalla Nora E, Keogh J, De Pergola G, Stirling H, Pannacciulli N, Sethi JK, Federspil G, Vidal-Puig A, Farooqi IS, O’Rahilly S, Vettor R. WNT10B mutations in human obesity. Diabetologia 2006;49(4):678–684.
  239. Lagathu C, Christodoulides C, Virtue S, Cawthorn WP, Franzin C, Kimber WA, Nora ED, Campbell M, Medina-Gomez G, Cheyette BNR, Vidal-Puig AJ, Sethi JK. Dact1, a nutritionally regulated preadipocyte gene, controls adipogenesis by coordinating the Wnt/beta-catenin signaling network. Diabetes 2009;58(3):609–19.
  240. Weiss MJ, Orkin SH. GATA transcription factors: key regulators of hematopoiesis. Exp. Hematol.1995;23(2):99–107.
  241. Tong Q, Dalgin G, Xu H, Ting CN, Leiden JM, Hotamisligil GS. Function of GATA transcription factors in preadipocyte-adipocyte transition. Science 2000;290(5489):134–138.
  242. Tong Q, Tsai J, Tan G, Dalgin G, Hotamisligil GS. Interaction between GATA and the C/EBP family of transcription factors is critical in GATA-mediated suppression of adipocyte differentiation. Mol. Cell. Biol.2005;25(2):706–715.
  243. Chen C, Xiang H, Peng Y, Peng J, Jiang S. Mature miR-183, negatively regulated by transcription factor GATA3, promotes 3T3-L1 adipogenesis through inhibition of the canonical Wnt/β-catenin signaling pathway by targeting LRP6. Cell. Signal. 2014;26(6):1155–1165.
  244. Wang L, Di L. Wnt/β-Catenin mediates AICAR effect to increase GATA3 expression and inhibit adipogenesis. J. Biol. Chem. 2015;290(32):19458–19468.
  245. de Sá PM, Richard AJ, Hang H, Stephens JM. Transcriptional regulation of adipogenesis. In: Comprehensive Physiology.Vol 7. Hoboken, NJ, USA: John Wiley & Sons, Inc.; 2017:635–674.
  246. Knebel B, Kotzka J, Lehr S, Hartwig S, Avci H, Jacob S, Nitzgen U, Schiller M, März W, Hoffmann MM, Seemanova E, Haas J, Muller-Wieland D. A mutation in the c-fos gene associated with congenital generalized lipodystrophy. Orphanet J. Rare Dis. 2013;8:11–12.
  247. Gupta RK, Arany Z, Seale P, Mepani RJ, Ye L, Conroe HM, Roby YA, Kulaga H, Reed RR, Spiegelman BM.Transcriptional control of preadipocyte determination by Zfp423. Nature 2010;464(7288):619–623.
  248. Quach JM, Walker EC, Allan E, Solano M, Yokoyama A, Kato S, Sims NA, Gillespie MT, Martin TJ. Zinc finger protein 467 is a novel regulator of osteoblast and adipocyte commitment. J. Biol. Chem. 2011;286(6):4186–4198.
  249. Shao M, Ishibashi J, Kusminski CM, Wang QA, Hepler C, Vishvanath L, MacPherson KA, Spurgin SB, Sun K, Holland WL, Seale P, Gupta RK. Zfp423 maintains white adipocyte identity through suppression of the beige cell thermogenic gene program. Cell Metab. 2016;23(6):1167–1184.
  250. Kang S, Akerblad P, Kiviranta R, Gupta RK, Kajimura S, Griffin MJ, Min J, Baron R, Rosen ED. Regulation of early adipose commitment by Zfp521. PLoS Biol. 2012;10(11):1–9.
  251. Correa D, Hesse E, Seriwatanachai D, Kiviranta R, Saito H, Yamana K, Neff L, Atfi A, Coillard L, Sitara D, Maeda Y, Warming S, Jenkins NA, Copeland NG, Horne WC, Lanske B, Baron R. Zfp521 is a target gene and key effector of parathyroid hormone-related peptide signaling in growth plate chondrocytes. Dev. Cell2010;19(4):533–546.
  252. Zamani N, Brown CW. Emerging roles for the transforming growth factor-{beta} superfamily in regulating adiposity and energy expenditure. Endocr. Rev. 2011;32(3):387–403.
  253. Wang EA, Israel DI, Kelly S, Luxenberg DP. Bone morphogenetic protein-2 causes commitment and differentiation in C3H10T1/2 and 3T3 cells. Growth Factors 1993;9(1):57–71.
  254. Butterwith SC, Wilkie RS, Clinton M. Treatment of pluripotential C3H 10T1/2 fibroblasts with bone morphogenetic protein-4 induces adipocyte commitment. Biochem. Soc. Trans. 1996;24(2):163S.
  255. Ignotz RA, Massagué J. Type beta transforming growth factor controls the adipogenic differentiation of 3T3 fibroblasts. Proc. Natl. Acad. Sci. U. S. A. 1985;82(24):8530–8534.
  256. Huang H, Song T-J, Li X, Hu L, He Q, Liu M, Lane MD, Tang Q-Q. BMP signaling pathway is required for commitment of C3H10T1/2 pluripotent stem cells to the adipocyte lineage. Proc. Natl. Acad. Sci. U. S. A.2009;106(31):12670–12675.
  257. Hata K, Nishimura R, Ikeda F, Yamashita K, Matsubara T, Nokubi T, Yoneda T. Differential roles of Smad1 and p38 kinase in regulation of peroxisome proliferator-activating receptor gamma during bone morphogenetic protein 2-induced adipogenesis. Mol. Biol. Cell 2003;14(2):545–55.
  258. Margoni A, Fotis L, Papavassiliou AG. The transforming growth factor-beta/bone morphogenetic protein signalling pathway in adipogenesis. Int. J. Biochem. Cell Biol. 2012;44(3):475–479.
  259. Björntorp P. Adipose tissue distribution and function. Int. J. Obes. 1991;15 Suppl 2:67–81.
  260. Wade GN, Gray JM, Bartness TJ. Gonadal influences on adiposity. Int. J. Obes. 1985;9 Suppl 1:83–92.
  261. Price TM, O’Brien SN. Determination of estrogen receptor messenger ribonucleic acid (mRNA) and cytochrome P450 aromatase mRNA levels in adipocytes and adipose stromal cells by competitive polymerase chain reaction amplification. J. Clin. Endocrinol. Metab. 1993;77(4):1041–1045.
  262. Crandall DL, Busler DE, Novak TJ, Weber R V., Kral JG. Identification of estrogen receptor β RNA in human breast and abdominal subcutaneous adipose tissue. Biochem. Biophys. Res. Commun. 1998;248(3):523–526.
  263. Dieudonné MN, Leneveu MC, Giudicelli Y, Pecquery R. Evidence for functional estrogen receptors α and β in human adipose cells: regional specificities and regulation by estrogens. Am. J. Physiol. Physiol.2004;286(3):C655–C661.
  264. Jeong S, Yoon M. 17β-Estradiol inhibition of PPARγ-induced adipogenesis and adipocyte-specific gene expression. Acta Pharmacol. Sin. 2011;32(2):230.
  265. Heine PA, Taylor JA, Iwamoto GA, Lubahn DB, Cooke PS. Increased adipose tissue in male and female estrogen receptor-alpha knockout mice. Proc. Natl. Acad. Sci. U. S. A. 2000;97(23):12729–12734.
  266. Jones MEE, Thorburn AW, Britt KL, Hewitt KN, Wreford NG, Proietto J, Oz OK, Leury BJ, Robertson KM, Yao S, Simpson ER. Aromatase-deficient (ArKO) mice have a phenotype of increased adiposity. Proc. Natl. Acad. Sci. 2000;97(23):12735–12740.
  267. Pedram A, Razandi M, Blumberg B, Levin ER. Membrane and nuclear estrogen receptor α collaborate to suppress adipogenesis but not triglyceride content. FASEB J. 2016;30(1):230–240.
  268. De Pergola G, Xu XF, Yang SM, Giorgino R, Bjorntorp P. Up-regulation of androgen receptor binding in male rat fat pad adipose precursor cells exposed to testosterone: study in a whole cell assay system. J. Steroid Biochem. Mol. Biol. 1990;37(4):553–558.
  269. Dieudonne M-N, Pecquery R, Leneveu M-C, Jaubert A-M, Giudicelli Y. Androgen receptors in cultured rat adipose precursor cells during proliferation and differentiation: regional specificities and regulation by testosterone. Endocrine 1995;3(7):537–541.
  270. Dieudonne MN, Pecquery R, Boumediene A, Leneveu MC, Giudicelli Y. Androgen receptors in human preadipocytes and adipocytes: regional specificities and regulation by sex steroids. Am. J. Physiol. 1998;274(6 Pt 1):C1645–C1652.
  271. Blouin K, Veilleux A, Luu-The V, Tchernof A. Androgen metabolism in adipose tissue: Recent advances. Mol. Cell. Endocrinol. 2009;301(1–2):97–103.
  272. Blouin K, Nadeau M, Perreault M, Veilleux A, Drolet R, Marceau P, Mailloux J, Luu-The V, Tchernof A. Effects of androgens on adipocyte differentiation and adipose tissue explant metabolism in men and women. Clin. Endocrinol. (Oxf). 2010;72(2):176–188.
  273. Vegiopoulos A, Herzig S. Glucocorticoids, metabolism and metabolic diseases. Mol. Cell. Endocrinol.2007;275(1–2):43–61.
  274. Wu Z, Bucher NL, Farmer SR. Induction of peroxisome proliferator-activated receptor gamma during the conversion of 3T3 fibroblasts into adipocytes is mediated by C/EBPbeta, C/EBPdelta, and glucocorticoids. Mol. Cell. Biol. 1996;16(8):4128–4136.
  275. Bujalska IJ, Gathercole LL, Tomlinson JW, Darimont C, Ermolieff J, Fanjul AN, Rejto PA, Stewart PM. A novel selective 11beta-hydroxysteroid dehydrogenase type 1 inhibitor prevents human adipogenesis. J. Endocrinol.2008;197(2):297–307.
  276. Lee M-J, Pramyothin P, Karastergiou K, Fried SK. Deconstructing the roles of glucocorticoids in adipose tissue biology and the development of central obesity. Biochim. Biophys. Acta 2014;1842(3):473–81.
  277. Sargis RM, Johnson DN, Choudhury RA, Brady MJ. Environmental endocrine disruptors promote adipogenesis in the 3T3-L1 cell line through glucocorticoid receptor activation. Obesity (Silver Spring). 2010;18(7):1283–1288.
  278. Caprio M, Fève B, Claës A, Viengchareun S, Lombès M, Zennaro M-C. Pivotal role of the mineralocorticoid receptor in corticosteroid-induced adipogenesis. FASEB J. 2007;21(9):2185–2194.
  279. Caprio M, Antelmi A, Chetrite G, Muscat A, Mammi C, Marzolla V, Fabbri A, Zennaro M-C, Fève B.Antiadipogenic effects of the mineralocorticoid receptor antagonist drospirenone: potential implications for the treatment of metabolic syndrome. Endocrinology 2011;152(1):113–125.
  280. Lee M-J, Fried SK. The glucocorticoid receptor, not the mineralocorticoid receptor, plays the dominant role in adipogenesis and adipokine production in human adipocytes. Int. J. Obes. (Lond). 2014;38(9):1228–1233.
  281. Nimitphong H, Holick MF, Fried SK, Lee M-J. 25-hydroxyvitamin D₃ and 1,25-dihydroxyvitamin D₃ promote the differentiation of human subcutaneous preadipocytes. Makishima M, ed. PLoS One 2012;7(12):e52171.
  282. Ji S, Doumit ME, Hill RA. Correction: regulation of adipogenesis and key adipogenic gene expression by 1, 25-dihydroxyvitamin D in 3T3-L1 cells. PLoS One 2015;10(7):e0134199.
  283. Ji S, Doumit ME, Hill RA. Regulation of adipogenesis and key adipogenic gene expression by 1, 25-dihydroxyvitamin D in 3T3-L1 cells. PLoS One 2015;10(6):1–29.
  284. Blumberg JM, Tzameli I, Astapova I, Lam FS, Flier JS, Hollenberg AN. Complex role of the vitamin D receptor and its ligand in adipogenesis in 3T3-L1 cells. J. Biol. Chem. 2006;281(16):11205–11213.
  285. Kong J, Li YC. Molecular mechanism of 1,25-dihydroxyvitamin D 3 inhibition of adipogenesis in 3T3-L1 cells. Am. J. Physiol. Metab. 2006;290(5):E916–E924.
  286. Kelly KA, Gimble JM. 1,25-Dihydroxy vitamin D 3 inhibits adipocyte differentiation and gene expression in murine bone marrow stromal cell clones and primary cultures. Endocrinology 1998;139(5):2622–2628.
  287. Cianferotti L, Demay MB. VDR-mediated inhibition of DKK1 and SFRP2 suppresses adipogenic differentiation of murine bone marrow stromal cells. J. Cell. Biochem. 2007;101(1):80–88.
  288. Pereira-Santos M, Costa PRF, Assis AMO, Santos CAST, Santos DB. Obesity and vitamin D deficiency: a systematic review and meta-analysis. Obes. Rev. 2015;16(4):341–349.
  289. Lotfi-Dizaji L, Mahboob S, Aliashrafi S, Vaghef-Mehrabany E, Ebrahimi-Mameghani M, Morovati A. Effect of vitamin D supplementation along with weight loss diet on meta-inflammation and fat mass in obese subjects with vitamin D deficiency: A double-blind placebo-controlled randomized clinical trial. Clin. Endocrinol. (Oxf).2019;90(1):94–101.
  290. Salehpour A, Hosseinpanah F, Shidfar F, Vafa M, Razaghi M, Dehghani S, Hoshiarrad A, Gohari M. A 12-week double-blind randomized clinical trial of vitamin D3supplementation on body fat mass in healthy overweight and obese women. Nutr. J. 2012;11(1):78.
  291. Mason C, Xiao L, Imayama I, Duggan C, Wang C-Y, Korde L, McTiernan A. Vitamin D3 supplementation during weight loss: a double-blind randomized controlled trial. Am. J. Clin. Nutr. 2014;99(5):1015–1025.
  292. Sneve M, Figenschau Y, Jorde R. Supplementation with cholecalciferol does not result in weight reduction in overweight and obese subjects. Eur. J. Endocrinol. 2008;159(6):675–684.
  293. Mariash CN. Thyroid hormone and the adipocyte. J. Clin. Endocrinol. Metab. 2003;88(12):5603–5604.
  294. Klaus S, Choy L, Champigny O, Cassard-Doulcier AM, Ross S, Spiegelman B, Ricquier D. Characterization of the novel brown adipocyte cell line HIB 1B. Adrenergic pathways involved in regulation of uncoupling protein gene expression. J. Cell Sci. 1994;107 ( Pt 1:313–319.
  295. Levacher C, Sztalryd C, Kinebanyan MF, Picon L. Effects of thyroid hormones on adipose tissue development in Sherman and Zucker rats. Am. J. Physiol. 1984;246(1 Pt 1):C50–C56.
  296. Berry DC, Jiang Y, Graff JM. Emerging roles of adipose progenitor cells in tissue development, homeostasis, expansion and thermogenesis. Trends Endocrinol. Metab. 2016;27(8):574–585.
  297. Lee Y-H, Mottillo EP, Granneman JG. Adipose tissue plasticity from WAT to BAT and in between. Biochim. Biophys. Acta 2014;1842(3):358–369.
  298. Yuwei Jiang, Daniel C. Berry WT, Graff JM. Independent stem cell lineages regulate adipose organogenesis and adipose homeostasis. Cell Rep. 2014;9(3):1007–1022.
  299. Wang QA, Tao C, Gupta RK, Scherer PE. Tracking adipogenesis during white adipose tissue development, expansion and regeneration. Nat. Med. 2013;19(10):1338–1344.
  300. Macotela Y, Emanuelli B, Mori MA, Gesta S, Schulz TJ, Tseng Y-H, Kahn CR. Intrinsic differences in adipocyte precursor cells from different white fat depots. Diabetes 2012;61(7):1691–1699.
  301. Sarjeant K, Stephens JM. Adipogenesis. Cold Spring Harb. Perspect. Biol. 2012;4(9):a008417–a008417.
  302. Wang W, Seale P. Control of brown and beige fat development. Nat. Rev. Mol. Cell Biol. 2016;17(11):691–702.
  303. Hong KY, Bae H, Park I, Park D-Y, Kim KH, Kubota Y, Cho E-S, Kim H, Adams RH, Yoo O-J, Koh GY. Perilipin+ embryonic preadipocytes actively proliferate along growing vasculatures for adipose expansion. Development2015;142(15):2623–2632.
  304. Seale P, Bjork B, Yang W, Kajimura S, Chin S, Kuang S, Scimè A, Devarakonda S, Conroe HM, Erdjument-Bromage H, Tempst P, Rudnicki MA, Beier DR, Spiegelman BM. PRDM16 controls a brown fat/skeletal muscle switch. Nature 2008;454(7207):961–967.
  305. Spalding KL, Arner E, Westermark PO, Bernard S, Buchholz BA, Bergmann O, Blomqvist L, Hoffstedt J, Näslund E, Britton T, Concha H, Hassan M, Rydén M, Frisén J, Arner P. Dynamics of fat cell turnover in humans. Nature 2008;453(7196):783–787.
  306. Rodeheffer MS, Birsoy K, Friedman JM. Identification of white adipocyte progenitor cells in vivo. Cell2008;135(2):240–249.
  307. Tang W, Zeve D, Suh JM, Bosnakovski D, Kyba M, Hammer RE, Tallquist MD, Graff JM. White fat progenitor cells reside in the adipose vasculature. Science 2008;322(5901):583–586.
  308. Zimmerlin L, Donnenberg VS, Pfeifer ME, Meyer EM, Péault B, Rubin JP, Donnenberg AD. Stromal vascular progenitors in adult human adipose tissue. Cytom. Part A 2009;9999A(1):NA-NA.
  309. Berry R, Rodeheffer MS. Characterization of the adipocyte cellular lineage in vivo. Nat. Cell Biol.2013;15(3):302–308.
  310. Cawthorn WP, Scheller EL, MacDougald OA. Adipose tissue stem cells meet preadipocyte commitment: going back to the future. J. Lipid Res. 2012;53(2):227–246.
  311. Majka SM, Fox KE, Psilas JC, Helm KM, Childs CR, Acosta AS, Janssen RC, Friedman JE, Woessner BT, Shade TR, Varella-Garcia M, Klemm DJ. De novo generation of white adipocytes from the myeloid lineage via mesenchymal intermediates is age, adipose depot, and gender specific. Proc. Natl. Acad. Sci.2010;107(33):14781–14786.
  312. Majka SM, Miller HL, Sullivan T, Erickson PF, Kong R, Weiser-Evans M, Nemenoff R, Moldovan R, Morandi SA, Davis JA, Klemm DJ. Adipose lineage specification of bone marrow-derived myeloid cells. Adipocyte2012;1(4):215–229.
  313. Rydén M, Uzunel M, Hård JL, Borgström E, Mold JE, Arner E, Mejhert N, Andersson DP, Widlund Y, Hassan M, Jones C V., Spalding KL, Svahn B-M, Ahmadian A, Frisén J, Bernard S, Mattsson J, Arner P. Transplanted bone marrow-derived cells contribute to human adipogenesis. Cell Metab. 2015;22(3):408–417.
  314. Roldan M, Macias-Gonzalez M, Garcia R, Tinahones FJ, Martin M. Obesity short-circuits stemness gene network in human adipose multipotent stem cells. FASEB J. 2011;25(12):4111–4126.
  315. Tchkonia T, Giorgadze N, Pirtskhalava T, Tchoukalova Y, Karagiannides I, Forse RA, DePonte M, Stevenson M, Guo W, Han J, Waloga G, Lash TL, Jensen MD, Kirkland JL. Fat depot origin affects adipogenesis in primary cultured and cloned human preadipocytes. Am. J. Physiol. Integr. Comp. Physiol. 2002;282(5):R1286–R1296.
  316. Baglioni S, Cantini G, Poli G, Francalanci M, Squecco R, Di Franco A, Borgogni E, Frontera S, Nesi G, Liotta F, Lucchese M, Perigli G, Francini F, Forti G, Serio M, Luconi M. Functional differences in visceral and subcutaneous fat pads originate from differences in the adipose stem cell. Gimble JM, ed. PLoS One2012;7(5):e36569.
  317. Mariman ECM, Wang P. Adipocyte extracellular matrix composition, dynamics and role in obesity. Cell. Mol. Life Sci. 2010;67(8):1277–1292.
  318. Khan T, Muise ES, Iyengar P, Wang Z V, Chandalia M, Abate N, Zhang BB, Bonaldo P, Chua S, Scherer PE.Metabolic dysregulation and adipose tissue fibrosis: role of collagen VI. Mol. Cell. Biol. 2009;29(6):1575–1591.
  319. Sun K, Tordjman J, Clément K, Scherer PE. Fibrosis and adipose tissue dysfunction. Cell Metab.2013;18(4):470–477.
  320. Lin D, Chun T-H, Kang L. Adipose extracellular matrix remodelling in obesity and insulin resistance. Biochem. Pharmacol. 2016;119:8–16.
  321. Halberg N, Wernstedt-Asterholm I, Scherer PE. The adipocyte as an endocrine cell. Endocrinol. Metab. Clin. North Am. 2008;37(3):753–768, x–xi.
  322. Rutkowski JM, Davis KE, Scherer PE. Mechanisms of obesity and related pathologies: the macro- and microcirculation of adipose tissue. FEBS J. 2009;276(20):5738–5746.
  323. Sun K, Kusminski CM, Scherer PE. Adipose tissue remodeling and obesity. J. Clin. Invest. 2011;121(6):2094–2101.
  324. Pasarica M, Gowronska-Kozak B, Burk D, Remedios I, Hymel D, Gimble J, Ravussin E, Bray GA, Smith SR.Adipose tissue collagen VI in obesity. J. Clin. Endocrinol. Metab. 2009;94(12):5155–5162.
  325. Sun K, Park J, Gupta OT, Holland WL, Auerbach P, Zhang N, Goncalves Marangoni R, Nicoloro SM, Czech MP, Varga J, Ploug T, An Z, Scherer PE. Endotrophin triggers adipose tissue fibrosis and metabolic dysfunction. Nat. Commun. 2014;5:3485.
  326. Park J, Scherer PE. Adipocyte-derived endotrophin promotes malignant tumor progression. J. Clin. Invest.2012;122(11):4243–4256.
  327. Nedergaard J, Bengtsson T, Cannon B. New Powers of Brown Fat: Fighting the Metabolic Syndrome. Cell Metab. 2011;13(3):238–240.
  328. Bartelt A, Bruns OT, Reimer R, Hohenberg H, Ittrich H, Peldschus K, Kaul MG, Tromsdorf UI, Weller H, Waurisch C, Eychmüller A, Gordts PLSM, Rinninger F, Bruegelmann K, Freund B, Nielsen P, Merkel M, Heeren J. Brown adipose tissue activity controls triglyceride clearance. Nat. Med. 2011;17(2):200–206.
  329. Ravussin Y, Xiao C, Gavrilova O, Reitman ML. Effect of Intermittent Cold Exposure on Brown Fat Activation, Obesity, and Energy Homeostasis in Mice. Aguila MB, ed. PLoS One 2014;9(1):e85876.
  330. Marlatt KL, Ravussin E. Brown Adipose Tissue: an Update on Recent Findings. Curr. Obes. Rep.2017;6(4):389–396.
  331. Johnson D, Dixon AK, Abrahams PH. The abdominal subcutaneous tissue: computed tomographic, magnetic resonance, and anatomical observations. Clin. Anat. 1996;9(1):19–24.
  332. Kelley DE, Thaete FL, Troost F, Huwe T, Goodpaster BH. Subdivisions of subcutaneous abdominal adipose tissue and insulin resistance. Am. J. Physiol. Metab. 2000;278(5):E941–E948.
  333. Smith SR, Lovejoy JC, Greenway F, Ryan D, deJonge L, de la Bretonne J, Volafova J, Bray GA. Contributions of total body fat, abdominal subcutaneous adipose tissue compartments, and visceral adipose tissue to the metabolic complications of obesity. Metabolism 2001;50(4):425–435.
  334. Golan R, Shelef I, Rudich A, Gepner Y, Shemesh E, Chassidim Y, Harman-Boehm I, Henkin Y, Schwarzfuchs D, Ben Avraham S, Witkow S, Liberty IF, Tangi-Rosental O, Sarusi B, Stampfer MJ, Shai I. Abdominal superficial subcutaneous fat: a putative distinct protective fat subdepot in type 2 diabetes. Diabetes Care 2012;35(3):640–7.
  335. Bjørndal B, Burri L, Staalesen V, Skorve J, Berge RK. Different adipose depots: their role in the development of metabolic syndrome and mitochondrial response to hypolipidemic agents. J. Obes. 2011;2011:490650.
  336. Karpe F, Pinnick KE. Biology of upper-body and lower-body adipose tissue—link to whole-body phenotypes. Nat. Rev. Endocrinol. 2015;11(2):90–100.
  337. Gabriely I, Ma XH, Yang XM, Atzmon G, Rajala MW, Berg AH, Scherer P, Rossetti L, Barzilai N. Removal of visceral fat prevents insulin resistance and glucose intolerance of aging: an adipokine-mediated process? Diabetes 2002;51(10):2951–2958.
  338. Muzumdar R, Allison DB, Huffman DM, Ma X, Atzmon G, Einstein FH, Fishman S, Poduval AD, McVei T, Keith SW, Barzilai N. Visceral adipose tissue modulates mammalian longevity. Aging Cell 2008;7(3):438–440.
  339. Weber R V, Buckley MC, Fried SK, Kral JG. Subcutaneous lipectomy causes a metabolic syndrome in hamsters. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2000;279(3):R936–R943.
  340. Tran TT, Yamamoto Y, Gesta S, Kahn CR. Beneficial effects of subcutaneous fat transplantation on metabolism. Cell Metab. 2008;7(5):410–420.
  341. Fabbrini E, Tamboli RA, Magkos F, Marks-Shulman PA, Eckhauser AW, Richards WO, Klein S, Abumrad NN.Surgical removal of omental fat does not improve insulin sensitivity and cardiovascular risk factors in obese adults. Gastroenterology 2010;139(2):448–455.
  342. Klein S, Fontana L, Young VL, Coggan AR, Kilo C, Patterson BW, Mohammed BS. Absence of an effect of liposuction on insulin action and risk factors for coronary heart disease. N. Engl. J. Med. 2004;350(25):2549–2557.
  343. Mohammed BS, Cohen S, Reeds D, Young VL, Klein S. Long-term effects of large-volume liposuction on metabolic risk factors for coronary heart disease. Obesity (Silver Spring). 2008;16(12):2648–2651.
  344. Tchkonia T, Morbeck DE, Von Zglinicki T, Van Deursen J, Lustgarten J, Scrable H, Khosla S, Jensen MD, Kirkland JL. Fat tissue, aging, and cellular senescence. Aging Cell 2010;9(5):667–684.
  345. Vohl M-C, Sladek R, Robitaille J, Gurd S, Marceau P, Richard D, Hudson TJ, Tchernof A. A survey of genes differentially expressed in subcutaneous and visceral adipose tissue in men. Obes. Res. 2004;12(8):1217–1222.
  346. Gesta S, Blüher M, Yamamoto Y, Norris AW, Berndt J, Kralisch S, Boucher J, Lewis C, Kahn CR. Evidence for a role of developmental genes in the origin of obesity and body fat distribution. Proc. Natl. Acad. Sci. U. S. A.2006;103(17):6676–6681.
  347. Karastergiou K, Fried SK, Xie H, Lee M-J, Divoux A, Rosencrantz MA, Chang RJ, Smith SR. Distinct developmental signatures of human abdominal and gluteal subcutaneous adipose tissue depots. J. Clin. Endocrinol. Metab. 2013;98(1):362–371.
  348. Yamamoto Y, Gesta S, Lee KY, Tran TT, Saadatirad P, Ronald Kahn C. Adipose depots possess unique developmental gene signatures. Obesity 2010;18(5):872–878.
  349. Khan T, Muise ES, Iyengar P, Wang Z V, Chandalia M, Abate N, Zhang BB, Bonaldo P, Chua S, Scherer PE.Metabolic dysregulation and adipose tissue fibrosis: role of collagen VI. Mol. Cell. Biol. 2009;29(6):1575–1591.
  350. Chandalia M, Lin P, Seenivasan T, Livingston EH, Snell PG, Grundy SM, Abate N. Insulin resistance and body fat distribution in South Asian men compared to Caucasian men. Maedler K, ed. PLoS One 2007;2(8):e812.
  351. Masuzaki H, Paterson J, Shinyama H, Morton NM, Mullins JJ, Seckl JR, Flier JS. A transgenic model of visceral obesity and the metabolic syndrome. Science 2001;294(5549):2166–2170.
  352. Rask E, Olsson T, Söderberg S, Andrew R, Livingstone DE, Johnson O, Walker BR. Tissue-specific dysregulation of cortisol metabolism in human obesity. J. Clin. Endocrinol. Metab. 2001;86(3):1418–1421.
  353. Sjöström L, Smith U, Krotkiewski M, Björntorp P. Cellularity in different regions of adipose tissue in young men and women. Metabolism. 1972;21(12):1143–1153.
  354. Macotela Y, Boucher J, Tran TT, Kahn CR. Sex and depot differences in adipocyte insulin sensitivity and glucose metabolism. Diabetes 2009;58(4):803–82.
  355. Nishikawa S, Yasoshima A, Doi K, Nakayama H, Uetsuka K. Involvement of sex, strain and age factors in high fat diet-induced obesity in C57BL/6J and BALB/cA mice. Exp. Anim. 2007;56(4):263–272.
  356. Hwang L-L, Wang C-H, Li T-L, Chang S-D, Lin L-C, Chen C-P, Chen C-T, Liang K-C, Ho I-K, Yang W-S, Chiou L-C. Sex differences in high-fat diet-induced obesity, metabolic alterations and learning, and synaptic plasticity deficits in mice. Obesity (Silver Spring). 2010;18(3):463–469.
  357. Goodpaster BH, Thaete FL, Simoneau JA, Kelley DE. Subcutaneous abdominal fat and thigh muscle composition predict insulin sensitivity independently of visceral fat. Diabetes 1997;46(10):1579–1585.
  358. Pinnick KE, Neville MJ, Fielding BA, Frayn KN, Karpe F, Hodson L. Gluteofemoral adipose tissue plays a major role in production of the lipokine palmitoleate in humans. Diabetes 2012;61(6):1399–1403.
  359. Jaworski K, Sarkadi-Nagy E, Duncan RE, Ahmadian M, Sul HS. Regulation of triglyceride metabolism. IV. Hormonal regulation of lipolysis in adipose tissue. Am. J. Physiol. Gastrointest. Liver Physiol. 2007;293(1):G1–G4.
  360. Langin D. Adipose tissue lipolysis as a metabolic pathway to define pharmacological strategies against obesity and the metabolic syndrome. Pharmacol. Res. 2006;53(6):482–491.
  361. Sengenès C, Zakaroff-Girard A, Moulin A, Berlan M, Bouloumié A, Lafontan M, Galitzky J. Natriuretic peptide-dependent lipolysis in fat cells is a primate specificity. Am. J. Physiol. Integr. Comp. Physiol. 2002;283(1):R257–R265.
  362. Kiwaki K, Levine JA. Differential effects of adrenocorticotropic hormone on human and mouse adipose tissue. J. Comp. Physiol. B Biochem. Syst. Environ. Physiol. 2003;173(8):675–678.
  363. Bousquet-Mélou A, Galitzky J, Lafontan M, Berlan M. Control of lipolysis in intra-abdominal fat cells of nonhuman primates: comparison with humans. J. Lipid Res. 1995;36(3):451–461.
  364. Driskell RR, Jahoda CAB, Chuong C-M, Watt FM, Horsley V. Defining dermal adipose tissue. Exp. Dermatol.2014;23(9):629–631.
  365. Wojciechowicz K, Gledhill K, Ambler CA, Manning CB, Jahoda CAB. Development of the mouse dermal adipose layer occurs independently of subcutaneous adipose tissue and is marked by restricted early expression of FABP4. Schneider MR, ed. PLoS One 2013;8(3):e59811.
  366. Kruglikov IL, Scherer PE. Dermal adipocytes: from irrelevance to metabolic targets? Trends Endocrinol. Metab.2016;27(1):1–10.
  367. Alexander CM, Kasza I, Yen C-LE, Reeder SB, Hernando D, Gallo RL, Jahoda CAB, Horsley V, MacDougald OA. Dermal white adipose tissue: a new component of the thermogenic response. J. Lipid Res.2015;56(11):2061–2069.
  368. Matsumura H, Engrav LH, Gibran NS, Yang TM, Grant JH, Yunusov MY, Fang P, Reichenbach DD, Heimbach DM, Isik FF. Cones of skin occur where hypertrophic scar occurs. Wound Repair Regen. 2001;9(4):269–277.
  369. Driskell RR, Lichtenberger BM, Hoste E, Kretzschmar K, Simons BD, Charalambous M, Ferron SR, Herault Y, Pavlovic G, Ferguson-Smith AC, Watt FM. Distinct fibroblast lineages determine dermal architecture in skin development and repair. Nature 2013;504(7479):277–281.
  370. Schmidt BA, Horsley V. Intradermal adipocytes mediate fibroblast recruitment during skin wound healing. Development 2013;140(7):1517–1527.
  371. Stepp MA, Gibson HE, Gala PH, Iglesia DDS, Pajoohesh-Ganji A, Pal-Ghosh S, Brown M, Aquino C, Schwartz AM, Goldberger O, Hinkes MT, Bernfield M. Defects in keratinocyte activation during wound healing in the syndecan-1-deficient mouse. J. Cell Sci. 2002;115(Pt 23):4517–4531.
  372. Kasza I, Suh Y, Wollny D, Clark RJ, Roopra A, Colman RJ, MacDougald OA, Shedd TA, Nelson DW, Yen M-I, Yen C-LE, Alexander CM. Syndecan-1 is required to maintain intradermal fat and prevent cold stress. Barsh GS, ed. PLoS Genet. 2014;10(8):e1004514.
  373. Zhang L -j., Guerrero-Juarez CF, Hata T, Bapat SP, Ramos R, Plikus M V., Gallo RL. Dermal adipocytes protect against invasive Staphylococcus aureus skin infection. Science 2015;347(6217):67–71.
  374. Festa E, Fretz J, Berry R, Schmidt B, Rodeheffer M, Horowitz M, Horsley V. Adipocyte lineage cells contribute to the skin stem cell niche to drive hair cycling. Cell 2011;146(5):761–771.
  375. Suffee N, Moore-Morris T, Farahmand P, Rücker-Martin C, Dilanian G, Fradet M, Sawaki D, Derumeaux G, LePrince P, Clément K, Dugail I, Puceat M, Hatem SN. Atrial natriuretic peptide regulates adipose tissue accumulation in adult atria. Proc. Natl. Acad. Sci. U. S. A. 2017;114(5):E771–E780.
  376. Iacobellis G, Barbaro G. Epicardial adipose tissue feeding and overfeeding the heart. Nutrition 2019;59:1–6.
  377. Iacobellis G. Epicardial and pericardial fat: close, but very different. Obesity 2012;17(4):625–625.
  378. Pabon MA, Manocha K, Cheung JW, Lo JC. Linking arrhythmias and adipocytes: insights, mechanisms, and future directions. Front. Physiol. 2018;9:1–12.
  379. Marchington JM, Mattacks CA, Pond CM. Adipose tissue in the mammalian heart and pericardium: Structure, foetal development and biochemical properties. Comp. Biochem. Physiol. Part B Comp. Biochem.1989;94(2):225–232.
  380. McAninch EA, Fonseca TL, Poggioli R, Panos AL, Salerno TA, Deng Y, Li Y, Bianco AC, Iacobellis G. Epicardial adipose tissue has a unique transcriptome modified in severe coronary artery disease. Obesity2015;23(6):1267–1278.
  381. Venteclef N, Guglielmi V, Balse E, Gaborit B, Cotillard A, Atassi F, Amour J, Leprince P, Dutour A, Clément K, Hatem SN. Human epicardial adipose tissue induces fibrosis of the atrial myocardium through the secretion of adipo-fibrokines. Eur. Heart J. 2015;36(13):795–805.
  382. Sacks HS, Fain JN, Holman B, Cheema P, Chary A, Parks F, Karas J, Optican R, Bahouth SW, Garrett E, Wolf RY, Carter RA, Robbins T, Wolford D, Samaha J. Uncoupling protein-1 and related messenger ribonucleic acids in human epicardial and other adipose tissues: epicardial fat functioning as brown fat. J. Clin. Endocrinol. Metab.2009;94(9):3611–3615.
  383. Greulich S, Maxhera B, Vandenplas G, de Wiza DH, Smiris K, Mueller H, Heinrichs J, Blumensatt M, Cuvelier C, Akhyari P, Ruige JB, Ouwens DM, Eckel J. Secretory products from epicardial adipose tissue of patients with type 2 diabetes mellitus induce cardiomyocyte dysfunction. Circulation 2012;126(19):2324–2334.
  384. Mazurek T, Zhang L, Zalewski A, Mannion JD, Diehl JT, Arafat H, Sarov-Blat L, O’Brien S, Keiper EA, Johnson AG, Martin J, Goldstein BJ, Shi Y. Human epicardial adipose tissue is a source of inflammatory mediators. Circulation 2003;108(20):2460–2466.
  385. Packer M. Epicardial adipose tissue may mediate deleterious effects of obesity and inflammation on the myocardium. J. Am. Coll. Cardiol. 2018;71(20):2360–2372.
  386. Cetin M, Cakici M, Polat M, Suner A, Zencir C, Ardic I. Relation of epicardial fat thickness with carotid intima-media thickness in patients with type 2 diabetes mellitus. Int. J. Endocrinol. 2013;2013:769175.
  387. Iacobellis G, Barbaro G, Gerstein HC. Relationship of epicardial fat thickness and fasting glucose. Int. J. Cardiol. 2008;128(3):424–426.
  388. Inzucchi SE, Bergenstal RM, Buse JB, Diamant M, Ferrannini E, Nauck M, Peters AL, Tsapas A, Wender R, Matthews DR. Management of hyperglycaemia in type 2 diabetes, 2015: a patient-centred approach. Update to a Position Statement of the American Diabetes Association and the European Association for the Study of Diabetes. Diabetologia 2015;58(3):429–442.
  389. Palmer SC, Mavridis D, Nicolucci A, Johnson DW, Tonelli M, Craig JC, Maggo J, Gray V, De Berardis G, Ruospo M, Natale P, Saglimbene V, Badve S V., Cho Y, Nadeau-Fredette A-C, Burke M, Faruque L, Lloyd A, Ahmad N, Liu Y, Tiv S, Wiebe N, Strippoli GFM. Comparison of Clinical Outcomes and Adverse Events Associated With Glucose-Lowering Drugs in Patients With Type 2 Diabetes. JAMA 2016;316(3):313.
  390. Sandouk T, Reda D, Hofmann C. Antidiabetic agent pioglitazone enhances adipocyte differentiation of 3T3-F442A cells. Am. J. Physiol. 1993;264(6 Pt 1):C1600-8.
  391. Hallakou S, Doaré L, Foufelle F, Kergoat M, Guerre-Millo M, Berthault MF, Dugail I, Morin J, Auwerx J, Ferré P.Pioglitazone induces in vivo adipocyte differentiation in the obese Zucker fa/fa rat. Diabetes 1997;46(9):1393–9.
  392. Smith SR, de Jonge L, Volaufova J, Li Y, Xie H, Bray GA. Effect of pioglitazone on body composition and energy expenditure: a randomized controlled trial. Metabolism 2005;54(1):24–32.
  393. Chiquette E, Ramirez G, DeFronzo R. A Meta-analysis Comparing the Effect of Thiazolidinediones on Cardiovascular Risk Factors. Arch. Intern. Med. 2004;164(19):2097.
  394. McLaughlin TM, Liu T, Yee G, Abbasi F, Lamendola C, Reaven GM, Tsao P, Cushman SW, Sherman A.Pioglitazone Increases the Proportion of Small Cells in Human Abdominal Subcutaneous Adipose Tissue. Obesity 2010;18(5):926–931.
  395. Paolisso G, Tataranni PA, Foley JE, Bogardus C, Howard B V, Ravussin E. A high concentration of fasting plasma non-esterified fatty acids is a risk factor for the development of NIDDM. Diabetologia 1995;38(10):1213–1217.
  396. Weyer C, Foley JE, Bogardus C, Tataranni PA, Pratley RE. Enlarged subcutaneous abdominal adipocyte size, but not obesity itself, predicts Type II diabetes independent of insulin resistance. Diabetologia2000;43(12):1498–1506.
  397. Okuno A, Tamemoto H, Tobe K, Ueki K, Mori Y, Iwamoto K, Umesono K, Akanuma Y, Fujiwara T, Horikoshi H, Yazaki Y, Kadowaki T. Troglitazone increases the number of small adipocytes without the change of white adipose tissue mass in obese Zucker rats. J. Clin. Invest. 1998;101(6):1354–1361.
  398. Arner E, Westermark PO, Spalding KL, Britton T, Rydén M, Frisén J, Bernard S, Arner P. Adipocyte turnover: relevance to human adipose tissue morphology. Diabetes 2010;59(1):105–109.
  399. Danforth E. Failure of adipocyte differentiation causes type II diabetes mellitus? Nat. Genet. 2000;26(1):13–13.
  400. Slawik M, Vidal-Puig AJ. Adipose tissue expandability and the metabolic syndrome. Genes Nutr. 2007;2(1):41–45.
  401. Virtue S, Vidal-Puig A. Adipose tissue expandability, lipotoxicity and the Metabolic Syndrome — An allostatic perspective. Biochim. Biophys. Acta - Mol. Cell Biol. Lipids 2010;1801(3):338–349.
  402. Heilbronn L, Smith SR, Ravussin E. Failure of fat cell proliferation, mitochondrial function and fat oxidation results in ectopic fat storage, insulin resistance and type II diabetes mellitus. Int. J. Obes. Relat. Metab. Disord.2004;28 Suppl 4(S4):S12-521.
  403. McLaughlin T, Sherman A, Tsao P, Gonzalez O, Yee G, Lamendola C, Reaven GM, Cushman SW. Enhanced proportion of small adipose cells in insulin-resistant vs insulin-sensitive obese individuals implicates impaired adipogenesis. Diabetologia 2007;50(8):1707–1715.
  404. Pasarica M, Xie H, Hymel D, Bray G, Greenway F, Ravussin E, Smith SR. Lower total adipocyte number but no evidence for small adipocyte depletion in patients with type 2 diabetes. Diabetes Care 2009;32(5):900–902.
  405. McLaughlin T, Lamendola C, Coghlan N, Liu TC, Lerner K, Sherman A, Cushman SW. Subcutaneous adipose cell size and distribution: relationship to insulin resistance and body fat. Obesity (Silver Spring). 2014;22(3):673–680.
  406. Michaud A, Laforest S, Pelletier M, Nadeau M, Simard S, Daris M, Lebœuf M, Vidal H, Géloën A, Tchernof A.Abdominal adipocyte populations in women with visceral obesity. Eur. J. Endocrinol. 2016;174(2):227–239.
  407. Kursawe R, Eszlinger M, Narayan D, Liu T, Bazuine M, Cali AMG, D’Adamo E, Shaw M, Pierpont B, Shulman GI, Cushman SW, Sherman A, Caprio S. Cellularity and adipogenic profile of the abdominal subcutaneous adipose tissue from obese adolescents: association with insulin resistance and hepatic steatosis. Diabetes2010;59(9):2288–2296.
  408. Johannsen DL, Tchoukalova Y, Tam CS, Covington JD, Xie W, Schwarz J-M, Bajpeyi S, Ravussin E. Effect of 8 weeks of overfeeding on ectopic fat deposition and insulin sensitivity: testing the “adipose tissue expandability” hypothesis. Diabetes Care 2014;37(10):2789–2797.
  409. McLaughlin T, Craig C, Liu L-F, Perelman D, Allister C, Spielman D, Cushman SW. Adipose Cell Size and Regional Fat Deposition as Predictors of Metabolic Response to Overfeeding in Insulin-Resistant and Insulin-Sensitive Humans. Diabetes 2016;65(5):1245–1254.
  410. White UA, Fitch MD, Beyl RA, Hellerstein MK, Ravussin E. Association of in vivo adipose tissue cellular kinetics with markers of metabolic health in humans. J. Clin. Endocrinol. Metab. 2017;102(7):2171–2178.
  411. Winer DA, Winer S, Shen L, Wadia PP, Yantha J, Paltser G, Tsui H, Wu P, Davidson MG, Alonso MN, Leong HX, Glassford A, Caimol M, Kenkel JA, Tedder TF, McLaughlin T, Miklos DB, Dosch H-M, Engleman EG. B cells promote insulin resistance through modulation of T cells and production of pathogenic IgG antibodies. Nat. Med.2011;17(5):610–617.
  412. Schipper HS, Prakken B, Kalkhoven E, Boes M. Adipose tissue-resident immune cells: key players in immunometabolism. Trends Endocrinol. Metab. 2012;23(8):407–415.
  413. Crewe C, An YA, Scherer PE. The ominous triad of adipose tissue dysfunction: inflammation, fibrosis, and impaired angiogenesis. J. Clin. Invest. 2017;127(1):74.
  414. DiSpirito JR, Mathis D. Immunological contributions to adipose tissue homeostasis. Semin. Immunol.2015;27(5):1–17.
  415. Strissel KJ, Stancheva Z, Miyoshi H, Perfield JW, DeFuria J, Jick Z, Greenberg AS, Obin MS. Adipocyte death, adipose tissue remodeling, and obesity complications. Diabetes 2007;56(12):2910–2918.
  416. Cinti S, Mitchell G, Barbatelli G, Murano I, Ceresi E, Faloia E, Wang S, Fortier M, Greenberg AS, Obin MS.Adipocyte death defines macrophage localization and function in adipose tissue of obese mice and humans. J. Lipid Res. 2005;46(11):2347–2355.
  417. Lumeng CN, Deyoung SM, Saltiel AR. Macrophages block insulin action in adipocytes by altering expression of signaling and glucose transport proteins. Am. J. Physiol. Endocrinol. Metab. 2007;292(1):E166–E174.
  418. Wernstedt Asterholm I, Tao C, Morley TS, Wang QA, Delgado-Lopez F, Wang Z V, Scherer PE. Adipocyte inflammation is essential for healthy adipose tissue expansion and remodeling. Cell Metab. 2014;20(1):103–118.
  419. Elks CM, Zhao P, Grant RW, Hang H, Bailey JL, Burk DH, McNulty MA, Mynatt RL, Stephens JM. Loss of oncostatin M signaling in adipocytes induces insulin resistance and adipose tissue inflammation in vivo. J. Biol. Chem. 2016;291(33):17066–17076.
  420. Stephens JM, Bailey JL, Hang H, Rittell V, Dietrich MA, Mynatt RL, Elks CM. Adipose tissue dysfunction occurs independently of obesity in adipocyte‐specific oncostatin receptor knockout mice. Obesity 2018;26(9):1439–1447.
  421. Smith GI, Mittendorfer B, Klein S. Metabolically healthy obesity: facts and fantasies. J. Clin. Invest.2019;129(10):3978–3989.
  422. Primeau V, Coderre L, Karelis AD, Brochu M, Lavoie M-E, Messier V, Sladek R, Rabasa-Lhoret R.Characterizing the profile of obese patients who are metabolically healthy. Int. J. Obes. 2011;35(7):971–981.
  423. Magkos F. Metabolically healthy obesity: what’s in a name? Am. J. Clin. Nutr. 2019;110(3):533–539.
  424. Lackey DE, Burk DH, Ali MR, Mostaedi R, Smith WH, Park J, Scherer PE, Seay SA, McCoin CS, Bonaldo P, Adams SH. Contributions of adipose tissue architectural and tensile properties toward defining healthy and unhealthy obesity. Am. J. Physiol. Endocrinol. Metab. 2014;306(3):E233–E246.
  425. Karelis AD, Rabasa-Lhoret R. Can inflammatory status define metabolic health? Nat. Rev. Endocrinol.2013;9(12):694–695.
  426. Tchernof A, Després J-P. Pathophysiology of human visceral obesity: An update. Physiol. Rev. 2013;93(1):359–404.
  427. Manolopoulos KN, Karpe F, Frayn KN. Gluteofemoral body fat as a determinant of metabolic health. Int. J. Obes. 2010;34(6):949–959.
  428. Brochu M, Tchernof A, Dionne IJ, Sites CK, Eltabbakh GH, Sims EAH, Poehlman ET. What are the physical characteristics associated with a normal metabolic profile despite a high level of obesity in postmenopausal women? J. Clin. Endocrinol. Metab. 2001;86(3):1020–1025.
  429. Messier V, Karelis AD, Prud’homme D, Primeau V, Brochu M, Rabasa-Lhoret R. Identifying metabolically healthy but obese individuals in sedentary postmenopausal women. Obesity 2010;18(5):911–917.
  430. Klöting N, Fasshauer M, Dietrich A, Kovacs P, Schön MR, Kern M, Stumvoll M, Blüher M. Insulin-sensitive obesity. Am. J. Physiol. Metab. 2010;299(3):E506–E515.
  431. O’Connell J, Lynch L, Cawood TJ, Kwasnik A, Nolan N, Geoghegan J, McCormick A, O’Farrelly C, O’Shea D.The relationship of omental and subcutaneous adipocyte size to metabolic disease in severe obesity. PLoS One2010;5(4):e9997.
  432. Shulman GI. Cellular mechanisms of insulin resistance. J. Clin. Invest. 2000;106(2):171–176.
  433. Goodpaster BH, He J, Watkins S, Kelley DE. Skeletal Muscle Lipid Content and Insulin Resistance: Evidence for a Paradox in Endurance-Trained Athletes. J. Clin. Endocrinol. Metab. 2001;86(12):5755–5761.
  434. Etgen GJ, Jensen J, Wilson CM, Hunt DG, Cushman SW, Ivy JL. Exercise training reverses insulin resistance in muscle by enhanced recruitment of GLUT-4 to the cell surface. Am. J. Physiol. 1997;272(5 Pt 1):E864-9.
  435. Seppälä-Lindroos A, Vehkavaara S, Häkkinen A-M, Goto T, Westerbacka J, Sovijärvi A, Halavaara J, Yki-Järvinen H. Fat accumulation in the liver is associated with defects in insulin suppression of glucose production and serum free fatty acids independent of obesity in normal men. J. Clin. Endocrinol. Metab. 2002;87(7):3023–3028.
  436. Stefan N, Kantartzis K, Machann J, Schick F, Thamer C, Rittig K, Balletshofer B, Machicao F, Fritsche A, Häring H-U. Identification and characterization of metabolically benign obesity in humans. Arch. Intern. Med.2008;168(15):1609.
  437. Ogorodnikova AD, Khan UI, McGinn AP, Zeb I, Budoff MJ, Harman SM, Miller VM, Brinton EA, Manson JE, Hodis HN, Merriam GR, Cedars MI, Taylor HS, Naftolin F, Lobo RA, Santoro N, Wildman RP. Ectopic fat and adipokines in metabolically benign overweight/obese women: the kronos early estrogen prevention study. Obesity (Silver Spring). 2013;21(8):1726–1733.
  438. Shin M-J, Hyun YJ, Kim OY, Kim JY, Jang Y, Lee JH. Weight loss effect on inflammation and LDL oxidation in metabolically healthy but obese (MHO) individuals: low inflammation and LDL oxidation in MHO women. Int. J. Obes. 2006;30(10):1529–1534.
  439. Koster A, Stenholm S, Alley DE, Kim LJ, Simonsick EM, Kanaya AM, Visser M, Houston DK, Nicklas BJ, Tylavsky FA, Satterfield S, Goodpaster BH, Ferrucci L, Harris TB, Health ABC Study. Body fat distribution and inflammation among obese older adults with and without metabolic syndrome. Obesity (Silver Spring).2010;18(12):2354–2361.
  440. Phillips CM, Perry IJ. Does inflammation determine metabolic health status in obese and nonobese adults? J. Clin. Endocrinol. Metab. 2013;98(10):E1610–E1619.
  441. Wildman RP, Kaplan R, Manson JE, Rajkovic A, Connelly SA, Mackey RH, Tinker LF, Curb JD, Eaton CB, Wassertheil-Smoller S. Body size phenotypes and inflammation in the women’s health initiative observational study. Obesiity 2011;19(7):1482–1491.
  442. Blüher M. The distinction of metabolically “healthy” from “unhealthy” obese individuals. Curr. Opin. Lipidol.2010;21(1):38–43.
  443. van Beek L, Lips MA, Visser A, Pijl H, Ioan-Facsinay A, Toes R, Berends FJ, Willems van Dijk K, Koning F, van Harmelen V. Increased systemic and adipose tissue inflammation differentiates obese women with T2DM from obese women with normal glucose tolerance. Metabolism 2014;63(4):492–501.
  444. Xu XJ, Gauthier M-S, Hess DT, Apovian CM, Cacicedo JM, Gokce N, Farb M, Valentine RJ, Ruderman NB.Insulin sensitive and resistant obesity in humans: AMPK activity, oxidative stress, and depot-specific changes in gene expression in adipose tissue. J. Lipid Res. 2012;53(4):792–801.
  445. Hardy OT, Perugini RA, Nicoloro SM, Gallagher-Dorval K, Puri V, Straubhaar J, Czech MP. Body mass index-independent inflammation in omental adipose tissue associated with insulin resistance in morbid obesity. Surg. Obes. Relat. Dis. 2011;7(1):60–67.
  446. Appleton SL, Seaborn CJ, Visvanathan R, Hill CL, Gill TK, Taylor AW, Adams RJ, North West Adelaide Health Study Teamon behalf of the NWAHS. Diabetes and cardiovascular disease outcomes in the metabolically healthy obese phenotype: a cohort study. Diabetes Care 2013;36(8):2388–2394.
  447. Chang Y, Ryu S, Suh B-S, Yun KE, Kim C-W, Cho S-I. Impact of BMI on the incidence of metabolic abnormalities in metabolically healthy men. Int. J. Obes. 2012;36(9):1187–1194.
  448. Soriguer F, Gutiérrez-Repiso C, Rubio-Martín E, García-Fuentes E, Almaraz MC, Colomo N, Esteva de Antonio I, de Adana MSR, Chaves FJ, Morcillo S, Valdés S, Rojo-Martínez G. Metabolically healthy but obese, a matter of time? Findings from the prospective pizarra study. J. Clin. Endocrinol. Metab. 2013;98(6):2318–2325.
  449. Samocha-Bonet D, Dixit VD, Kahn CR, Leibel RL, Lin X, Nieuwdorp M, Pietiläinen KH, Rabasa-Lhoret R, Roden M, Scherer PE, Klein S, Ravussin E. Metabolically healthy and unhealthy obese--the 2013 Stock Conference report. Obes. Rev. 2014;15(9):697–708.
  450. Lin H, Zhang L, Zheng R, Zheng Y. The prevalence, metabolic risk and effects of lifestyle intervention for metabolically healthy obesity: a systematic review and meta-analysis: A PRISMA-compliant article. Medicine (Baltimore). 2017;96(47):e8838.
  451. Mongraw-Chaffin M, Foster MC, Anderson CAM, Burke GL, Haq N, Kalyani RR, Ouyang P, Sibley CT, Tracy R, Woodward M, Vaidya D. Metabolically Healthy Obesity, Transition to Metabolic Syndrome, and Cardiovascular Risk. J. Am. Coll. Cardiol. 2018;71(17):1857–1865.
  452. Akinci B, Sahinoz M, Oral E.Lipodystrophy Syndromes: Presentation and Treatment. MDText.com, Inc.; 2000. Available at: http://www.ncbi.nlm.nih.gov/pubmed/29989768. Accessed March 20, 2020.
  453. Garg A. Lipodystrophies: genetic and acquired body fat disorders. J. Clin. Endocrinol. Metab.2011;96(11):3313–3325.
  454. Villarroya F, Domingo P, Giralt M. Lipodystrophy associated with highly active anti-retroviral therapy for HIV infection: the adipocyte as a target of anti-retroviral-induced mitochondrial toxicity. Trends Pharmacol. Sci.2005;26(2):88–93.
  455. Rother KI, Brown RJ. Novel forms of lipodystrophy: why should we care? Diabetes Care 2013;36(8):2142–2145.
  456. Huang-Doran I, Sleigh A, Rochford JJ, O’Rahilly S, Savage DB. Lipodystrophy: metabolic insights from a rare disorder. J. Endocrinol. 2010;207(3):245–255.
  457. Savage DB. Mouse models of inherited lipodystrophy. Dis. Model. Mech. 2009;2(11–12):554–562.
  458. Vernochet C, Damilano F, Mourier A, Bezy O, Mori MA, Smyth G, Rosenzweig A, Larsson N-G, Kahn CR.Adipose tissue mitochondrial dysfunction triggers a lipodystrophic syndrome with insulin resistance, hepatosteatosis, and cardiovascular complications. FASEB J. 2014;28(10):4408–4419.
  459. Oral E, Chan J. Rationale for leptin-replacement therapy for severe lipodystrophy. Endocr. Pract.2010;16(2):324–333.
  460. McDuffie JR, Riggs PA, Calis KA, Freedman RJ, Oral EA, DePaoli AM, Yanovski JA. Effects of exogenous leptin on satiety and satiation in patients with lipodystrophy and leptin insufficiency. J. Clin. Endocrinol. Metab.2004;89(9):4258–4263.
  461. Moitra J, Mason MM, Olive M, Krylov D, Gavrilova O, Marcus-Samuels B, Feigenbaum L, Lee E, Aoyama T, Eckhaus M, Reitman ML, Vinson C. Life without white fat: a transgenic mouse. Genes Dev. 1998;12(20):3168–3181.
  462. Reitman ML, Mason MM, Moitra J, Gavrilova O, Marcus-Samuels B, Eckhaus M, Vinson C. Transgenic mice lacking white fat: models for understanding human lipoatrophic diabetes. Ann. N. Y. Acad. Sci. 1999;892:289–296.
  463. Gavrilova O, Marcus-Samuels B, Graham D, Kim JK, Shulman GI, Castle AL, Vinson C, Eckhaus M, Reitman ML. Surgical implantation of adipose tissue reverses diabetes in lipoatrophic mice. J. Clin. Invest.2000;105(3):271–278.
  464. Zhang Z, Turer E, Li X, Zhan X, Choi M, Tang M, Press A, Smith SR, Divoux A, Moresco EMY, Beutler B. Insulin resistance and diabetes caused by genetic or diet-induced KBTBD2 deficiency in mice. Proc. Natl. Acad. Sci. U. S. A. 2016;113(42):E6418–E6426.
  465. Colombo C, Cutson JJ, Yamauchi T, Vinson C, Kadowaki T, Gavrilova O, Reitman ML. Transplantation of adipose tissue lacking leptin is unable to reverse the metabolic abnormalities associated with lipoatrophy. Diabetes 2002;51(9):2727–2733.
  466. Shimomura I, Hammer RE, Ikemoto S, Brown MS, Goldstein JL. Leptin reverses insulin resistance and diabetes mellitus in mice with congenital lipodystrophy. Nature 1999;401(6748):73–76.
  467. Oral EA, Simha V, Ruiz E, Andewelt A, Premkumar A, Snell P, Wagner AJ, DePaoli AM, Reitman ML, Taylor SI, Gorden P, Garg A. Leptin-replacement therapy for lipodystrophy. N. Engl. J. Med. 2002;346(8):570–578.
  468. Haque WA, Shimomura I, Matsuzawa Y, Garg A. Serum Adiponectin and Leptin Levels in Patients with Lipodystrophies. J. Clin. Endocrinol. Metab. 2002;87(5):2395–2395.
  469. Polyzos SA, Perakakis N, Mantzoros CS. Fatty liver in lipodystrophy: A review with a focus on therapeutic perspectives of adiponectin and/or leptin replacement. Metabolism. 2019;96:66–82.
  470. Akinci B, Onay H, Demir T, Ozen S, Kayserili H, Akinci G, Nur B, Tuysuz B, Nuri Ozbek M, Gungor A, Yildirim Simsir I, Altay C, Demir L, Simsek E, Atmaca M, Topaloglu H, Bilen H, Atmaca H, Atik T, Cavdar U, Altunoglu U, Aslanger A, Mihci E, Secil M, Saygili F, Comlekci A, Garg A. Natural history of congenital generalized lipocity: A nationwide study from turkey. J. Clin. Endocrinol. Metab. 2016;101(7):2759–2767.
  471. Akinci B, Onay H, Demir T, Savas-Erdeve Ş, Gen R, Simsir IY, Keskin FE, Erturk MS, Uzum AK, Yaylali GF, Ozdemir NK, Atik T, Ozen S, Yurekli BS, Apaydin T, Altay C, Akinci G, Demir L, Comlekci A, Secil M, Oral EA.Clinical presentations, metabolic abnormalities and end-organ complications in patients with familial partial lipodystrophy. Metabolism 2017;72:109–119.
  472. Lima JG, Nobrega LHC, Lima NN, Dos Santos MCF, Silva PHD, Baracho M de FP, Lima DN, de Melo Campos JTA, Ferreira LC, Freire Neto FP, Mendes-Aguiar C de O, Jeronimo SMB. Causes of death in patients with Berardinelli-Seip congenital generalized lipodystrophy. PLoS One 2018;13(6):e0199052.
  473. Simha V, Rao S, Garg A. Prolonged thiazolidinedione therapy does not reverse fat loss in patients with familial partial lipodystrophy, Dunnigan variety. Diabetes, Obes. Metab. 2008;10(12):1275–1276.
  474. Farooqi IS. Leptin and the onset of puberty: insights from rodent and human genetics. Semin. Reprod. Med.2002;20(2):139–144.
  475. Kawwass JF, Summer R, Kallen CB. Direct effects of leptin and adiponectin on peripheral reproductive tissues: a critical review. Mol. Hum. Reprod. 2015;21(8):617–632.
  476. Moschos S, Chan JL, Mantzoros CS. Leptin and reproduction: a review. Fertil. Steril. 2002;77(3):433–444.
  477. O’Rahilly S, Farooqi IS, Yeo GSH, Challis BG. Minireview: human obesity—lessons from monogenic disorders. Endocrinology 2003;144(9):3757–3764.
  478. Hausman GJ, Barb CR. Adipose tissue and the reproductive axis: biological aspects. In: Adipose Tissue Development.Vol 19. Basel: KARGER; 2010:31–44.
  479. Barash IA, Cheung CC, Weigle DS, Ren H, Kabigting EB, Kuijper JL, Clifton DK, Steiner RA. Leptin is a metabolic signal to the reproductive system. Endocrinology 1996;137(7):3144–3147.
  480. Mantzoros CS, Moschos S, Avramopoulos I, Kaklamani V, Liolios A, Doulgerakis DE, Griveas I, Katsilambros N, Flier JS. Leptin concentrations in relation to body mass index and the tumor necrosis factor-α system in humans 1. J. Clin. Endocrinol. Metab. 1997;82(10):3408–3413.
  481. APTER D. The role of leptin in female adolescence. Ann. N. Y. Acad. Sci. 2003;997(1):64–76.
  482. Wen J-P, Lv W-S, Yang J, Nie A-F, Cheng X-B, Yang Y, Ge Y, Li X-Y, Ning G. Globular adiponectin inhibits GnRH secretion from GT1-7 hypothalamic GnRH neurons by induction of hyperpolarization of membrane potential. Biochem. Biophys. Res. Commun. 2008;371(4):756–761.
  483. Kiezun M, Smolinska N, Maleszka A, Dobrzyn K, Szeszko K, Kaminski T. Adiponectin expression in the porcine pituitary during the estrous cycle and its effect on LH and FSH secretion. Am. J. Physiol. Metab.2014;307(11):E1038–E1046.
  484. Chabrolle C, Tosca L, Dupont J. Regulation of adiponectin and its receptors in rat ovary by human chorionic gonadotrophin treatment and potential involvement of adiponectin in granulosa cell steroidogenesis. Reproduction 2007;133(4):719–731.
  485. Tworoger SS, Mantzoros C, Hankinson SE. Relationship of plasma adiponectin with sex hormone and insulin-like growth factor levels*. Obesity 2007;15(9):2217–2224.
  486. Henson MC, Castracane VD. Leptin in pregnancy: an update. Biol. Reprod. 2006;74(2):218–229.
  487. Stein TP, Scholl TO, Schluter MD, Schroeder CM. Plasma leptin influences gestational weight gain and postpartum weight retention. Am. J. Clin. Nutr. 1998;68(6):1236–1240.
  488. Sámano R, Martínez-Rojano H, Chico-Barba G, Godínez-Martínez E, Sánchez-Jiménez B, Montiel-Ojeda D, Tolentino M. Serum concentration of leptin in pregnant adolescents correlated with gestational weight gain, postpartum weight retention and newborn weight/length. Nutrients 2017;9(10):1–16.
  489. Schubring C, Englaro P, Siebler T, Blum WF, Demirakca T, Kratzsch J, Kiess W. Longitudinal analysis of maternal serum leptin levels during pregnancy, at birth and up to six weeks after birth: relation to body massindex, skinfolds, sex steroids and umbilical cord blood leptin levels. Horm. Res. Paediatr. 1998;50(5):276–283.
  490. Nien JK, Mazaki-Tovi S, Romero R, Erez O, Kusanovic JP, Gotsch F, Pineles BL, Gomez R, Edwin S, Mazor M, Espinoza J, Yoon BH, Hassan SS. Plasma adiponectin concentrations in non-pregnant, normal and overweight pregnant women. J. Perinat. Med. 2007;35(6):522–531.
  491. Williams MA, Qiu C, Muy-Rivera M, Vadachkoria S, Song T, Luthy DA. Plasma adiponectin concentrations in early pregnancy and subsequent risk of gestational diabetes mellitus. J. Clin. Endocrinol. Metab.2004;89(5):2306–2311.
  492. Thyfault JP, Hedberg EM, Anchan RM, Thorne OP, Isler CM, Newton ER, Dohm GL, DeVente JE. Gestational diabetes is associated with depressed adiponectin levels. J. Soc. Gynecol. Investig. 2005;12(1):41–45.
  493. Lekva T, Roland MCP, Michelsen AE, Friis CM, Aukrust P, Bollerslev J, Henriksen T, Ueland T. Large reduction in adiponectin during pregnancy is associated with large-for-gestational-age newborns. J. Clin. Endocrinol. Metab. 2017;102(7):2552–2559.
  494. Michalakis KG, Segars JH. The role of adiponectin in reproduction: from polycystic ovary syndrome to assisted reproduction. Fertil. Steril. 2010;94(6):1949–1957.
  495. Musso C, Cochran E, Javor E, Young J, Depaoli AM, Gorden P. The long-term effect of recombinant methionyl human leptin therapy on hyperandrogenism and menstrual function in female and pituitary function in male and female hypoleptinemic lipodystrophic patients. Metabolism. 2005;54(2):255–263.
  496. Maguire M, Lungu A, Gorden P, Cochran E, Stratton P. Pregnancy in a woman with congenital generalized lipodystrophy: leptin’s vital role in reproduction. Obstet. Gynecol. 2012;119(2 Pt 2):452–455.
  497. Ahima RS. Body fat, leptin, and hypothalamic amenorrhea. N. Engl. J. Med. 2004;351(10):959–962.
  498. Mathew H, Castracane VD, Mantzoros C. Adipose tissue and reproductive health. Metabolism 2018;86:18–32.
  499. Ruscica M, Macchi C, Gandini S, Morlotti B, Erzegovesi S, Bellodi L, Magni P. Free and bound plasma leptin in anorexia nervosa patients during a refeeding program. Endocrine 2016;51(2):380–383.
  500. Mantzoros CS, Magkos F, Brinkoetter M, Sienkiewicz E, Dardeno TA, Kim S-Y, Hamnvik O-PR, Koniaris A.Leptin in human physiology and pathophysiology. Am. J. Physiol. Endocrinol. Metab. 2011;301(4):E567–E584.
  501. 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.
  502. Lambrinoudaki I, Papadimitriou D. Pathophysiology of bone loss in the female athlete. Ann. N. Y. Acad. Sci.2010;1205(1):45–50.
  503. Mantzoros CS, Georgiadis EI. Body mass and physical activity are important predictors of serum androgen concentrations in young healthy men. Epidemiology 1995;6(4):432–5.
  504. Kelly DM, Jones TH. Testosterone and obesity. Obes. Rev. 2015;16(7):581–606.
  505. Cohen PG. The hypogonadal-obesity cycle: role of aromatase in modulating the testosterone-estradiol shunt--a major factor in the genesis of morbid obesity. Med. Hypotheses 1999;52(1):49–51.
  506. Toulis KA, Goulis DG, Farmakiotis D, Georgopoulos NA, Katsikis I, Tarlatzis BC, Papadimas I, Panidis D.Adiponectin levels in women with polycystic ovary syndrome: a systematic review and a meta-analysis. Hum. Reprod. Update 2009;15(3):297–307.
  507. Calle EE, Kaaks R. Overweight, obesity and cancer: epidemiological evidence and proposed mechanisms. Nat. Rev. Cancer 2004;4(8):579–591.
  508. Nagaraju GP, Rajitha B, Aliya S, Kotipatruni RP, Madanraj AS, Hammond A, Park D, Chigurupati S, Alam A, Pattnaik S. The role of adiponectin in obesity-associated female-specific carcinogenesis. Cytokine Growth Factor Rev. 2016;31:37–48.
  509. Mantzoros C, Petridou E, Dessypris N, Chavelas C, Dalamaga M, Alexe DM, Papadiamantis Y, Markopoulos C, Spanos E, Chrousos G, Trichopoulos D. Adiponectin and breast cancer risk. J. Clin. Endocrinol. Metab.2004;89(3):1102–1107.
  510. Zwick RK, Guerrero-Juarez CF, Horsley V, Plikus M V. Anatomical, physiological, and functional diversity of adipose tissue. Cell Metab. 2018;27(1):68–83.
  511. Blaszkiewicz M, Willows JW, Johnson CP, Townsend KL, Blaszkiewicz M, Willows JW, Johnson CP, Townsend KL. The importance of peripheral nerves in adipose tissue for the regulation of energy balance. Biology (Basel).2019;8(1):10.
  512. Stanford KI, Middelbeek RJW, Townsend KL, Lee M-Y, Takahashi H, So K, Hitchcox KM, Markan KR, Hellbach K, Hirshman MF, Tseng Y-H, Goodyear LJ. A novel role for subcutaneous adipose tissue in exercise-induced improvements in glucose homeostasis. Diabetes 2015;64(6):2002–2014.
  513. Stanford KI, Lynes MD, Takahashi H, Baer LA, Arts PJ, May FJ, Lehnig AC, Middelbeek RJW, Richard JJ, So K, Chen EY, Gao F, Narain NR, Distefano G, Shettigar VK, Hirshman MF, Ziolo MT, Kiebish MA, Tseng Y-H, Coen PM, Goodyear LJ. 12,13-diHOME: an exercise-induced lipokine that increases skeletal muscle fatty acid uptake. Cell Metab. 2018;27(5):1111–1120.
  514. Flaherty SE, Grijalva A, Xu X, Ables E, Nomani A, Ferrante AW. A lipase-independent pathway of lipid release and immune modulation by adipocytes. Science 2019;363(6430):989–993.
  515. Crewe C, Joffin N, Rutkowski JM, Kim M, Zhang F, Towler DA, Gordillo R, Scherer PE. An endothelial-to-adipocyte extracellular vesicle axis governed by metabolic state. Cell 2018;175(3):695–708.

Nephrolithiasis

ABSTRACT

 

Kidney stones are concretions of different mineral salts mixed with an organic matrix that form in the upper urinary tract. As a stone moves from the kidney to the ureter, it can present with renal colic symptoms, and may cause urinary tract obstruction and/or infection. In fact, acute passage of a kidney stone is one of the leading reasons for visits to an emergency room. Over the past four decades, the lifetime prevalence of nephrolithiasis has more than doubled in the United States (and several developed countries), afflicting around 11% of men and 7% of women. Unless the underlying etiology of stone formation is adequately addressed, kidney stones can recur at a rate of around 50% ten years after initial presentation. The evaluation of a kidney stone former requires an extensive medical history (to identify environmental, metabolic, and/or genetic factors contributing to stone formation), imaging studies to evaluate and track stone burden, and laboratory studies (serum and urinary chemistries, stone composition analysis) to guide lifestyle and pharmacological therapy. The majority of kidney stones are composed of calcium (calcium oxalate and/or calcium phosphate), either pure or in combination with uric acid. Calcium oxalate stones can be caused by hypercalciuria, hyperoxaluria, hyperuricosuria, hypocitraturia, and/or low urine volume. Calcium phosphate stones occur in patients with hypercalciuria, hypocitraturia, an elevated urine pH, and/or low urine volume. In addition to lifestyle changes (increasing fluid intake, reduction in salt intake, moderation of calcium and animal protein intake), pharmacological therapy directed at the underlying metabolic abnormality (thiazides for hypercalciuria, potassium citrate for hypocitraturia, xanthine oxidase inhibitors for hyperuricosuria) can significantly reduce calcium stone recurrence rate. Pure uric acid stones account for 8-10% of all stones, although their prevalence is significantly greater in stone formers with type 2 diabetes and/or the metabolic syndrome. Uric acid stones are primarily caused by an excessively acidic urine, and urinary alkalinization with medications such as potassium citrate can dissolve uric acid stones and prevent recurrent uric acid nephrolithiasis. Cystine stones result from inactivating mutations in genes that encode renal tubular transporters that reabsorb the amino acid cysteine, typically present in childhood, are highly recurrent, and require aggressive control of cystinuria with specific pharmacological therapy. Infection (struvite) stones often present as staghorn, and require careful surgical removal of all of the stone material.

 

OVERVIEW

 

Urinary stone disease is a common clinical condition that afflicts 1 in 11 individuals in the United States, and is increasing in incidence. It typically presents with renal colic symptoms following the passage of a calculus from the kidney to the ureter. The pathophysiology of kidney stone formation is diverse, and includes a combination of genetic and environmental factors. In fact, kidney stone formation should not be viewed as a diagnosis but rather a symptom of an underlying abnormality. Evaluation of a kidney stone former with a detailed history, and appropriate laboratory and imaging studies helps to identify risk factors for stone formation and provides an opportunity to institute therapy that reduces the risk of stone recurrence. This chapter reviews the epidemiology of kidney stone disease, discusses the pathophysiology underlying the most commonly encountered stone types, and details the evaluation and management of patients with nephrolithiasis.

 

EPIDEMIOLOGY AND NATURAL HISTORY

 

Epidemiology

 

Nephrolithiasis is a common clinical condition encountered in both developed and developing countries. Its prevalence in the United States has more than doubled over the past 4 decades from 3.8% in 1976-1980 to 5.2% in 1988-1994 to 8.8% in 2007-2010 (1, 2). Similar increases in the prevalence of nephrolithiasis have been reported in other developed countries (3). Factors underlying this rising prevalence include changes in diet and fluid intake, greater use of medications and procedures that predispose to stone formation, the association of stone disease with the rampant epidemics of obesity and type 2 diabetes, climate change, and increased use of abdominal imaging (2). In the United States, stone disease afflicts Caucasians at a greater frequency than Hispanics (Odds Ratio: 0.60 vs. Caucasians) and African Americans (Odds Ratio: 0.37 vs. Caucasians) (2). It is also more prevalent in men than women, although recent reports suggest a narrowing in this gender gap, with the greatest increase in stone incidence in recent years occurring in younger women (4). A marked geographic variability in the prevalence of nephrolithiasis is also reported, with 20-50% higher prevalence of stone disease in the U.S. Southeast (“stone belt”) than the Northwest, primarily due to differences in exposure to temperature, humidity, and sunlight (5). Recent predictions suggest a likely northward expansion of the present-day U.S. “stone belt” as an unanticipated result of global warming (6). In 2000, the total cumulative costs for caring for patients with urolithiasis were estimated at US $2.1 billion (7). The rising prevalence of obesity and diabetes, together with population growth, is projected to contribute to dramatic increases in the cost of urolithiasis by an additional $1.24 billion/year estimated by 2030(8).

 

Natural History

 

RECURRENCE

 

The natural history of stone disease was studied in detail in all validated cases presenting with incident kidney stones in Olmsted County, Minnesota between 1984 to 2003 and followed for stone recurrence through 2012 (9). For the first episode, 48% of patients passed their stone spontaneously with confirmation, 33% required surgery for stone removal, 8% presumably spontaneously passed their stone (without confirmation), and 12% had no documentation of passage (9). This cohort was followed for a median of 11.2 years with recurrence occurring in 11%, 20%, 31%, and 39% at 2, 5, 10, and 15 years, respectively. The stone recurrence rates per 100 person-years were 3.4 after the first stone episode, 7.1 after the second episode, 12.1 after the third episode, and 17.6 after the fourth or higher episode (10). Independent risk factors for incident stone recurrence include younger age; male sex; higher body mass index; family history of stones; pregnancy; history of a brushite, struvite, or uric acid stone; number of kidney stones on imaging; and diameter of the largest kidney stone on imaging (10). These studies have led to the development and refinement of the Recurrence Of Kidney Stone (ROKS) tool to predict the risk of symptomatic recurrence by using readily available clinical characteristics of patients with kidney stones (9, 10).

 

POTENTIAL COMPLICATIONS

 

Urinary Tract Infection

 

There is a bi-directional relationship between urinary tract infections (UTI) and nephrolithiasis, as chronic UTIs lead to the formation of struvite stones, and stone disease increases the risk of UTI. Struvite stones typically occur in patients infected with urea-splitting organisms, and their pathogenesis and management are described in more detail in a later section. Gram-negative bacilli are the most common pathogen in UTI in patients with urolithiasis. Independent risk factors for UTI among patients with kidney stones include female gender, older age, presence of obstruction, and higher number of kidney stones (11). Low fluid intake is a reversible risk factor for both UTI and nephrolithiasis (12). Infectious (13) as well as non-infectious stones (14) can harbor bacteria inside, making the bacteria resistant to antimicrobial therapy. In patients with recurrent UTIs, removal of non-struvite non-obstructing stones is associated with elimination of further UTI recurrence in nearly 90% of cases (15).

 

Chronic Kidney Disease and End-Stage Renal Disease

 

Loss of renal function in patients with kidney stones may occur as a complication of obstruction by a stone lodged in the ureter, a complication of the urological procedure to remove a stone, or from the disordered pathophysiology underlying some stones. Staghorn stones caused by uric acid nephrolithiasis, cystinuria, renal tubular acidosis (RTA), or chronic infection are well-recognized causes of decreased renal function. Additional risk factors for the development of chronic kidney disease in stone formers include a solitary kidney, ileal conduit, neurogenic bladder, and development of hydronephrosis (16). In the Olmsted County cohort, end-stage renal disease incidence in patients with recurrent symptomatic kidney stones was twice that of the general non-stone forming population even after adjusting for baseline hypertension, diabetes mellitus, dyslipidemia, gout, obesity, and chronic kidney disease (17). Still, the absolute risk of ESRD from kidney stone disease was low.

 

Stone Disease in Pregnancy

 

Pregnancy-related mechanical and physiological changes alter risk factors for kidney stone formation, and management of acute nephrolithiasis in pregnancy is significantly more complicated than in non-pregnant women, at least in part due to imaging limitations and treatment restrictions (18). In an observational study, stones in pregnancy were associated with recurrent abortions, mild preeclampsia, chronic hypertension, gestational diabetes mellitus, and cesarean deliveries (19). Urinary tract infections and pyelonephritis and signs of ureteral obstruction including hydroureter and hydronephrosis were common, while premature rupture of membranes and preterm delivery were not more frequent. The newborns also were affected by perinatal complications including low birth weight, lower Apgar scores, and perinatal mortality. The majority of stones during pregnancy are calcium phosphate with a lesser number of calcium oxalate, in contrast to non-pregnant women in whom calcium oxalate stones are most common (20).

 

ASSOCIATION WITH OTHER CONDITIONS

 

Traditionally, nephrolithiasis was thought of as a condition caused by poor diet and abnormal renal handling of electrolytes, with complications limited to the kidneys and the urinary tract. However, recent investigations suggest that kidney stones may in fact be a systemic disorder associated with serious disorders including osteoporosis and greater fracture risk (21), metabolic syndrome features including diabetes, hypertension, and dyslipidemia (22), and greater incidence of cardiovascular disease (23).

 

CLINICAL MANIFESTATIONS

 

Symptoms and Signs

 

A classical episode of renal colic has a sudden onset, with fluctuation and intensification over 15 to 45 minutes. The pain then becomes steady and unbearable and often is accompanied by nausea and emesis. As the stone passes down the ureter toward the bladder, flank pain changes in a downward direction toward the groin. As the stone lodges at the ureterovesical junction, urinary frequency and dysuria appear. The pain may clear as the stone moves into the bladder or from the calyceal system into the ureter. Hematuria, generally microscopic but occasionally frank, frequently accompanies stone passage. The presence of bleeding alone does not predict a more severe outcome. Episodes of rapid onset of pain, bleeding, and then rapid clearing, often called ‘passing gravel’, is the result of passing a large amount of crystals of calcium oxalate, uric acid, or cystine. “Non-classical” presentations of kidney stones include dull low back pain, gastrointestinal symptoms such as diarrhea, isolated microscopic hematuria, asymptomatic urinary obstruction with renal insufficiency, recurrent urinary tract obstruction, or incidental discovery on abdominal imaging.

 

Likelihood of Passage

 

The size, number, and metabolic composition of new stones strongly influence the natural history and complication rates. Smaller stone size and more distal location in the ureter at presentation predict greater likelihood of spontaneous stone passage. Furthermore, the clinical presentation can be in part classified by metabolic type (Table 1). Spontaneous stone passage may occur with calcium oxalate, calcium phosphate, uric acid, and cystine stones. Rarely does a struvite stone or a staghorn stone of other composition (cystine, uric acid) pass spontaneously.

 

Table 1. Clinical Manifestations of Stones by Composition

Clinical feature

Calcium

Uric acid

Struvite

Cystine

Crystalluria

+

+

-

+

Stone passage

+

+

-

+

Small discrete stones

+

+

-

+

Sludge and obstruction

-

+

-

+

Radiodense

+

-

+

+

Staghorn

-

+

+

+

Nephrocalcinosis

+

-

-

-

 

EVALUATION

 

History

 

Evaluation of kidney stones starts with a detailed history focusing on medical conditions associated with higher risk of kidney stones (e.g. primary hyperparathyroidism, gastrointestinal disorders or surgeries, frequent urinary tract infections, gout, metabolic syndrome, etc.), family history suggestive of genetic causes of kidney stones (e.g.cystinuria, idiopathic hypercalciuria, young age at onset, etc.), dietary history (e.g. intake of fluid, salt, protein, dairy products, oxalate-rich foods (Table 2) (24), etc.), medications associated with increased risk of kidney stones (e.g.topiramate, zonisamide, excessive vitamin C, etc.)  (25, 26), and medications that directly precipitate and form stones (e.g. indinavir, triamterene, etc.) (Table 3).  This history can provide guidance on the biochemical evaluation and management of kidney stones. 

 

Table 2. Oxalate content of foods (24)

Food category

High in oxalate

Low in oxalate

Fruits

Figs, raspberries, dates

Apples, oranges, peaches, raisins, mango

Vegetables

Spinach, okra, beans, beets

Lettuce, asparagus, carrots, avocado, corn

Grains

Whole grain products

White rice, pasta, white bread

Nuts

All nuts (peanuts, almonds...)

 

Other

Chocolate, cocoa, black tea

Coffee, milk products, meat products

 

 

Table 3. Drugs associated with kidney stones (25, 26)

Drugs associated with increased risk of stones

Drugs that precipitate and form stones

Hypercalciuria (predispose to calcium stones):

Excessive calcium and vitamin D supplement

Loop diuretics

Glucocorticoids

 

Hypocitraturia and high urine pH (predispose to calcium phosphate stones):

Carbonic anhydrase inhibitors (acetazolamide, topiramate, zonisamide)

 

Hyperoxaluria (predispose to calcium oxalate stones):

Frequent use of antibacterial agents

Excessive vitamin C supplement 

 

Hyperuricosuria (predispose to uric acid stones and calcium oxalate stones):

Uricosuric drugs (probenecid, losartan)

 

High urine ammonium (predispose to ammonium urate stones):

Laxative abuse

Anti-viral:

Indinavir

Atazanavir

Nelfinavir

Tenofovir disoproxil fumarate

Raltegravir

Efavirenz

 

Antibiotics:

Sulfadiazine

Sulfamethoxazole

Ciprofloxacin

Amoxicillin

Ampicillin

Ceftriaxone

 

Others:

Triamterene

Sulfasalazine

Allopurinol

Guaifenesin / Ephedrine

Magnesium trisilicate

 

Laboratory Testing

 

The extent of biochemical evaluation depends on the risk of stone recurrence and patients’ interest.  For first-time stone formers, a basic evaluation including urinalysis, urine culture, and basic metabolic panel should be obtained.  For high risk stone formers (Table 4) and interested first-time stone formers, a thorough evaluation (Table 5) is warranted which includes 24-hour urine stone risk profile (27-29).

 

BLOOD

 

The recommended blood testing of kidney stone formers includes assessment of renal function, serum electrolytes (including potassium, calcium, phosphorus, and magnesium), and serum uric acid. Assessment of serum PTH may be needed in patients with suspected primary hyperparathyroidism.

 

Table 4. High Risk Stone Formers (27-29)

Recurrent stone formers:

·       Recurrent kidney stones

·       Bilateral or multiple kidney stones

 

History suggestive of genetic causes of kidney stones:

·       Early onset of kidney stones (age < 18 years)

·       Family history of kidney stones

 

Stone types that are more commonly associated with metabolic abnormalities:

·       Pure calcium phosphate stones

·       Non-calcium stones (uric acid, cystine, struvite, and magnesium ammonium phosphate stones) 

·       Any stone requiring percutaneous nephrolithotomy

 

History of medical conditions associated with increased recurrence risk: 

·       Hyperparathyroidism

·       Sarcoidosis

·       Bowel disease (e.g. inflammatory bowel disease, chronic pancreatitis, chronic diarrhea)

·       Bowel resection (e.g. colon resection, ileostomy, small bowel resection, Roux-en-Y gastric bypass)

·       Cystic fibrosis

·       Metabolic syndrome (e.g. obesity, type 2 diabetes mellitus, dyslipidemia, hypertension)

·       Gout

·       Spinal cord injury

·       Neurogenic bladder

·       Recurrent urinary tract infection

·       Osteoporosis

·       Renal tubular acidosis

·       Polycystic kidney disease

·       Cystinuria

·       Primary hyperoxaluria

·       Anatomical abnormalities that result in impaired urine flow (e.g. medullary sponge kidney, ureteral stricture, horseshoe kidney, etc.)

 

At high risk for complications of kidney stones:

·       Solitary kidney (functional or anatomical)

·       Chronic kidney disease

·       Complicated stone episodes that resulted in severe acute kidney injury, sepsis, or complicated hospitalization

 

At high risk occupations (e.g. pregnancy, pilots, police officer, military personnel, firemen, etc.)

 

Table 5. Biochemical Evaluation for High Risk Stone Formers and Interested First Time Stone Formers (27-29)

Serum: Basic metabolic panel, albumin, phosphate, magnesium, uric acid, PTH

 

Urinalysis and urine culture

 

24-hour urine: Volume, creatinine, pH, calcium, citrate, oxalate, uric acid, sodium, potassium, magnesium, chloride, sulfate, phosphate, ammonium and cystine (if cystine stone is confirmed or suspected)

 

Stone analysis

 

URINE

 

The initial 24-hour urine sample should be collected on the patient’s typical random diet.  It is controversial whether one or two 24-hour urine collections should be obtained (30-34).  At least one collection is needed, but two collections are preferred (27-29) . It is important to provide detailed instructions to patients to ensure an adequate collection and proper storage of urine sample.  Patients start urine collection after their first morning void and end collection with the first morning void the next day.  Storage of urine sample varies according to the instructions of the urine collection kits.  Urine sample should be sent to a reliable lab for 24-hour urine stone risk profile analysis.  24-hour urine stone risk profile should be repeated at 8-16 weeks after dietary changes or if pharmacotherapy is initiated to monitor response to therapy and allow dose adjustment as needed (28). Once the therapeutic target is achieved, 24-hour urine stone risk profile is repeated annually (28).  

 

24-hour urine stone risk profile typically provides information on 24-hour urine volume, creatinine, pH, calcium, citrate, oxalate, uric acid, sodium, potassium, magnesium, sulfate, phosphate, ammonium and cystine (if requested).  In addition, relative supersaturations with respect to calcium oxalate, calcium phosphate and uric acid are reported. Relative supersaturations are calculated accounting for multiple factors including promotors and inhibitors associated with crystallization (35). Higher relative supersaturation is associated with higher likelihood of being stone formers and correlate with stone composition (36, 37).

 

STONE ANALYSIS

 

Knowledge of stone composition may help direct the appropriate choice of urological procedures, evaluation of potential underlying metabolic abnormalities, and medical interventions to prevent stone recurrence (27-29, 38, 39). Current guidelines recommend obtaining stone analysis when feasible for all first-time stone formers (27-29). Stone composition may change in the same individual over time (40, 41)  Discordant stone compositions may also coexist in the same individual with bilateral kidney stones (42, 43).  Therefore, repeat stone analysis should be obtained in recurrence under pharmacological treatment, early recurrence after urological intervention and late recurrence after a prolonged stone-free period (28).

 

Multiple analytical techniques are available for stone analysis.  The currently preferred methods are X-ray diffraction and Fourier transform infrared spectroscopy (44).  X-ray diffraction uses monochromic X-rays to create a unique diffraction pattern of the crystalline structure of the stone (45).  Fourier transform infrared spectroscopy uses infrared radiation to create a unique energy absorption band pattern of the molecular structure of the stone (45).  These patterns can then be matched to a reference database to determine the stone composition.  Both X-ray diffraction and Fourier transform infrared spectroscopy are very accurate in identifying pure stones; however, the majority of kidney stones in clinical practice are mixed stones (44, 45).  Both methods have limitations in identification of certain mixed stone compositions.  X-ray diffraction cannot identify non-crystalline structures thus is prone to high rates of error in the detection of apatite component in mixed stones which is mostly pseudo-amorphous (44).  It is also time-consuming and expensive which limit its broad use in clinical practice (45).  Fourier transform infrared spectroscopy is quick and less expensive, but it cannot reliably detect small amounts of components in certain mixed stones (e.g. whewellite (hydrated calcium oxalate) in whewellite/uric acid stones and struvite in struvite/apatite stones) (44-47).  The accuracy of stone analysis by Fourier transform infrared spectroscopy depends on the quality of the reference database and trained personnel (46, 47).  While pure stones are reliably identified, there is variability in reporting the components of mixed stones in commercial laboratories which needs to be kept in mind when interpreting stone analysis results (47, 48). 

 

Imaging

 

A number of imaging modalities are available to evaluate stone number, size, and location in patients with nephrolithiasis (Table 6). Abdominal computed tomography (CT) without contrast is the initial imaging test of choice for suspected stone disease due to its high sensitivity and specificity, along with its widespread availability and the rapidity of scan time. One downside to CT scan use is exposure to ionizing radiation, which may increase long-term cancer risk. Lower radiation doses are effective in the diagnosis of nephrolithiasis in most patients, leading to greater recent adoption of “low-dose” and “ultra-low dose” CT scan protocols for evaluation of stone disease (49, 50). Use of ultrasonography as the initial test in patients with suspected nephrolithiasis in the emergency department (ED) may reduce cumulative radiation exposure without significantly increasing subsequent serious adverse events, pain scores, return visits to the ED, or hospitalizations (51). Abdominal X-rays (KUB) may be used as an alternative to CT scan and ultrasonography for follow-up of stone burden, although this modality misses radiolucent stones such as uric acid stones. Magnetic resonance imaging is capable of identifying kidney stones, but cost and limited availability make it a less attractive imaging modality for nephrolithiasis.

 

Table 6. Comparison of Different Imaging Modalities in the Assessment of Nephrolithiasis

 

Availability

Cost

Ionizing Radiation

Other Advantages

Other Drawbacks

CT Scan

Wide

Moderate

Highest

-Detects extra-renal pathology

-Useful in identifying uric acid stone composition

-Major drawback is radiation

Ultrasound

Wide

Moderate

None

-Portable US available

-Use in children, pregnancy

-Does not visualize ureteral stones

-Large body habitus limits visualization

X-ray

Wide

Low

Low

-Useful in follow-up of known radiopaque stones

-Visualizes kidneys, ureters, and bladder

-Misses radiolucent and/or small stones

-Overlying bowel gas, and extra-renal calcification impact stone visualization

MRI

Limited

High

None

-Use in children, pregnancy

-Contrast risk in CKD patients

-Cannot distinguish stone from blood clot

IVP

Limited

Moderate

High

-Occasional use in

preoperative planning

-Contrast use

 

SURGICAL MANAGEMENT

 

An obstructive stone in the setting of urinary infection is a urological emergency and requires urgent decompression with a ureteral stent or nephrostomy tube (28, 38). Patients should have urine and, if appropriate, blood cultures obtained and be started on broad spectrum intravenous antibiotics until culture results are available. These patients often require fluid resuscitation and monitoring in an intensive care setting. Definitive stone treatment is delayed until infection resolves. 

 

Patients who present with a ureteral stone up to 10mm can be offered a trial of passage if they have no signs or symptoms of urinary tract infection, their renal function is at their baseline, and their pain is well controlled. The likelihood of ureteral stone passage is influenced by stone size and location with smaller, more distal stones having the highest chance of passage (52).  Furthermore, smaller stones tend to pass quicker than larger stones (53).  For those patients attempting spontaneous passage, a trial of medical expulsive therapy with pain control and a-blocker for 4-6 weeks can be offered in uncomplicated cases (i.e. in the absence of infection, uncontrolled pain, obstruction, renal insufficiency, or renal anatomy associated with low likelihood of spontaneous stone passage) (28, 38, 54-56).  Nonsteroidal anti-inflammatory drugs (NSAIDs) including intravenous ketorolac are the treatment of choice for pain control. (57)  Opioids are used as rescue therapy for pain refractory to NSAIDs. (58)  In emergency room setting, IV lidocaine is a useful non-opioid option for pain control with close cardiac monitoring if there are no contraindications. (59, 60)  Alpha-blockers inhibit basal tone and decrease peristaltic frequency and amplitude in the lower ureters, decrease intraureteral pressure and increase fluid transport, thus they are proposed to be useful in stone expulsion. (61)  However, the effectiveness of a-blockers as medical expulsive therapy remains controversial.  A recent meta-analysis showed the use of a-blockers is associated with increased stone clearance and decreased time to stone passage with little major adverse events compared to standard therapy without a-blockers, but the quality of evidence is low. (62)  The benefit of a-blockers was mainly demonstrated in individuals with ureteral stones with sizes 6-10mm.  Little effect was found in stones measuring 5mm or smaller likely because these stones frequently pass spontaneously even without medical expulsive therapy. (62)  Potential side effects of a-blockers include orthostatic hypotension, dizziness, tachycardia, palpitations, headache, and abnormal ejaculation in males.  In large clinical trials, these were not found to be more frequent in individuals treated with a-blockers than placebo with the exception of ejaculatory dysfunction in males. (62, 63)   

 

Outpatient referral to urology is indicated for stones larger than 10mm, stones smaller than 10mm that fail to pass with medical expulsive therapy, and stones causing obstruction at the ureteropelvic junction, renal pelvis, or renal calyces especially in symptomatic patients and those at high risk for potential complications. Urological referral should also be considered for high risk stone formers (Table 4). Surgical intervention for stone treatment depends on symptoms, stone composition, size, and location (Table 7) (28, 38, 64).

 

Table 7. Surgical Management of Kidney Stones (28, 38, 64)

Stone

Surgical procedure 

Urological emergency:

 

Obstructive stone with infected urine

·       Obtain urinalysis, urine culture and start empiric antibiotics and IV fluid resuscitation

·       May need ICU care

·       Urgent decompression (ureteral stent or percutaneous nephrostomy tube)

·       Delay definitive stone treatment until infection resolves

Ureteral stones:

 

Uncomplicated ureteral stone £ 10mm

·       Trial of medical expulsive therapy with pain control (NSAIDS with narcotics if needed) and a-blocker for up to 4-6 weeks with reassessment of pain control, renal function and stone passage

Proximal ureteral stone < 10mm

·       SWL*

·       URS# if cystine stone or uric acid stone

Proximal ureteral stone > 10mm

·       URS

Mid ureteral stone of any size

·       URS

Distal ureteral stone of any size

·       URS

Renal stones:

 

Asymptomatic renal stone < 15 mm

·       Conservative therapy with surveillance of symptoms and imaging at 6 months and then annually for stone growth and new stone formation

Symptomatic non-lower pole renal stone £20mm

·       SWL or URS

Symptomatic non-lower pole renal stone > 20mm

·       PCNL

Symptomatic lower pole renal stone £ 10mm

·       SWL or URS

Symptomatic lower pole renal stone > 10mm

·       URS or PCNL

Staghorn stones

·       PCNL

Involved kidney with negligible function

·       Consider nephrectomy if recurrent infection or pain

     

#URS: ureteroscopy;

†PCNL: percutaneous nephrolithotomy

*SWL: Shockwave lithotripsy.  Calcium oxalate monohydrate, brushite and cystine stones are hard and resistant to fragmentation by SWL, thus alternative methods of stone removal are considered (28).  SWL has a lower rate of complications and morbidity, but a lower stone free rate in a single procedure than URS.

 

MEDICAL MANAGEMENT

 

General Measures for All Patients with Kidney Stones

 

Some risk factors (e.g. low urine volume, hypocitraturia, high sodium intake, and high animal protein intake) are shared among different types of stones. General dietary measures (summarized in Table 8) targeting these risk factors can be recommended for stone prevention. These can be especially useful when stone analysis and/or 24-hour urine stone risk profile are not available. Results from a 24-hour urine collection (Table 9) can further refine these recommendations.

 

Low urine volume is a risk factor for nephrolithiasis. High urine volume leads to urinary dilution of lithogenic constituents and reduced crystallization of calcium oxalate, calcium phosphate and uric acid (65, 66).  Several prospective studies have demonstrated high urine volume achieved with high fluid intake is associated with reduction in incident stones and recurrent stones (67-69).  Fluid intake of 2.5 to 3 liters per day or achieving a urine volume of at least 2-2.5 liters per day is recommended (27-29). Regarding the types of fluid other than water, orange juice, lemonade, coffee (caffeinated and decaffeinated), tea and alcohol have been associated with reduced risk of stone formation although with some controversial results (5, 68, 70-72).  Cola and grapefruit juice have been associated with increased risk of stone formation (70).

 

A high dietary sodium intake is associated with increased risk of nephrolithiasis likely by causing increased urinary calcium and decreased urinary citrate (28, 66, 73, 74).  For every 100 mmol/day increase in dietary sodium intake, urinary calcium increases by an average of 40 mg/day in non-stone forming adults and by up to 80 mg/day in hypercalciuric stone formers (66, 74-76).  A low sodium diet reduced urinary calcium and recurrent stones in hypercalciuric stone formers (76, 77).  Stone formers are therefore recommended to limit their dietary sodium intake to less than 2300 mg/day (or 100 mmol/day) which is equivalent to 5.9 grams of salt (sodium chloride) (27-29).

 

A high dietary animal protein intake (meat, fish and poultry) is a risk factor for nephrolithiasis in general (69, 78).  It is associated with increased urinary calcium, uric acid, phosphate and reduced urinary citrate and pH (79). On average, urinary calcium increases by 1 mg/day for every 1 g/day increase in dietary animal protein intake (80, 81).  In a randomized clinical trial of recurrent calcium oxalate stone formers, a diet with limited animal protein (52 g/day) and sodium (50 mmol/day) but normal calcium (1,200 mg/day) reduced stone recurrence by about 50% at 5 years when compared to a low-calcium diet (400mg mg/day) in hypercalciuric stone formers (77).  It is recommended to limit dietary animal protein intake to 0.8 to 1.0 g/kg weight per day (28, 82).

 

Urinary calcium excretion increases with increased dietary calcium intake which can be more pronounced in individuals with hyperabsorptive idiopathic hypercalciuria (66, 83).  However, a diet restrictive in calcium has not been demonstrated to prevent nephrolithiasis.  On the contrary, several studies showed a lower dietary calcium intake is associated with a higher risk of both incident and recurrent stones than a higher dietary calcium intake in men and women (69, 73, 77, 84).  A restricted calcium diet increases enteric absorption of oxalate and urinary oxalate which increases supersaturation of calcium oxalate (85, 86).  In addition, a low calcium diet may lead to negative calcium balance and bone loss. Therefore, a normal calcium diet with 1,000-1,200 mg/day is recommended as a dietary measure for stone prevention (27-29). Dietary sources of calcium are preferred.  However, if supplemental calcium is needed, it is best taken in divided doses with meals to reduce enteric absorption of oxalate (27-29).

 

A diet rich in fruits and vegetables is associated with a decreased risk of incident kidney stones (87) and current guidelines on medical management of nephrolithiasis also recommend a diet rich in fruits and vegetables for prevention of stone recurrence (27-29).  In normal individuals, elimination of dietary fruits and vegetables decreased urinary potassium, magnesium, citrate and oxalate, and increased urinary calcium, ammonium and relative supersaturation of calcium oxalate and calcium phosphate (88). In hypocitraturic stone formers, introduction of fruits and vegetables in the diet increased urinary potassium, magnesium, citrate, volume and pH, and decreased relative supersaturation of calcium oxalate and uric acid (88). 

 

Table 8. General Dietary Measures for All Stone Formers (27-29)

Dietary measures

Targeted risk factors

Fluid intake:

·       Fluid intake of 2.5 to 3 liters per day

·       Achieving urine output of 2-2.5 liters per day

 

Low urine volume

Salt intake:

·       Sodium intake less than 100 mEq (2,300 mg) per day

 

Hypercalciuria

Animal protein intake:

·       0.8-1.0 grams / kilogram body weight per day

 

Hypercalciuria

Hyperuricosuria

Hyperphosphaturia

Hypocitraturia

Low urine pH

Calcium intake:

·       Calcium intake of 1,000-1,200 mg per day divided into 2 doses taken with meals (prefer dietary source over supplemental calcium)

 

Hyperoxaluria

Fibers, vegetables and fruits:

Hypocitraturia

Low urine pH

 

Table 9. 24-hour Urine Stone Risk Profile Interpretation (89)

Urine parameter 

Reference range

Risk factor for stone types

Interpretation

Volume

> 2-2.5 L/day

All

·       Reflect fluid intake and extra-renal fluid loss

·       Goal is above 2-2.5 L/day

Creatinine

·       Male: 20-25 mg/kg body weight/day

·       Female: 15-20 mg/kg body weight/day

---

·       Assess adequacy of urine collection

pH

5.7-6.3

·       High pH: calcium phosphate and struvite

·       Low pH: uric acid and cystine

·       High pH: distal renal tubular acidosis (dRTA) or UTIs

·       Low pH: excessive animal protein intake, chronic diarrhea, or idiopathic

Calcium

·       Male: < 300 mg/day

·       Female: < 250 mg/day

·       Either sex: < 4 mg/kg body weight/day

Calcium oxalate

Calcium phosphate

·       Hypercalciuria (see details in “Hypercalciuria”) 

Oxalate

< 45 mg/day

Calcium oxalate

·       Enteric hyperoxaluria

·       > 100 mg/day, consider primary hyperoxaluria

Citrate

> 320 mg/day

Calcium oxalate

Calcium phosphate

·       Hypocitraturia (see details in “Hypocitraturia”) 

Uric acid

< 700 mg/day

Calcium oxalate

Uric acid

·       High purine intake or production

Phosphorus

< 1,100 mg/day

Calcium phosphate

·       Excessive protein intake

Sodium

< 200 mmol/day

Calcium oxalate

Calcium phosphate

·       Excessive salt intake

Chloride

< 200 mmol/day

Calcium oxalate

Calcium phosphate

·       Varies with sodium and potassium intake

Sulfate

< 40 mmol/day

Calcium oxalate

Calcium phosphate

Uric acid

·       Excessive animal protein intake

Ammonium

< 40 mmol/day

---

·       Excessive animal protein intake

·       Non-dietary acid load (e.g.diarrhea)

Potassium

> 40 mmol/day

---

Low urine potassium:

·       Low alkaline intake

·       Potassium loss (e.g.diarrhea)

Magnesium

> 80 mg/day

---

Low urine magnesium:

·       Low magnesium intake

·       Malabsorption

Cystine

< 40 mg/day

Cystine stone

·       Cystinuria 

 

Calcium Stones

 

Approximately 80% of kidney stones are calcium stones (calcium oxalate and/or calcium phosphate) (44). The initiating events of stone formation are controversial (90-94), but there are three proposed pathways of stone formation: 1) Randall’s plaque (interstitial calcium phosphate deposit at the renal papilla) grows and erodes the urothelium and becomes a nidus for crystal growth in urine supersaturated with respect to calcium oxalate; 2) Randall’s plug formed by fixed particle mechanism in which a crystal nidus is attached to the apical epithelium of the collecting duct and allows crystal growth in urine supersaturated with respect to the constituents of the stone; 3) Randall’s plug formed by free particle mechanism in which a crystal nidus forms through homogenous nucleation in the lumen of the nephron in the supersaturated environment (92).  Randall’s plaque is a prominent feature in idiopathic calcium oxalate stone formers and patients with primary hyperparathyroidism; although plugging is also observed (93, 94).  Randall’s plug formed by fixed particle mechanism is seen in brushite stone formers and patients with dRTA and primary hyperparathyroidism.  Randall’s plug formed by free particle mechanism is seen in cystinuric stone formers and intestinal bypass patients (92).

 

Calcium stones can be idiopathic or associated with systemic diseases (see Table 10) (89, 95).  Idiopathic calcium stones formers may exhibit various urinary risk factors for calcium oxalate and calcium phosphate stones (Table 11) (89).

 

Table 10. Systemic Conditions Associated with Calcium Stones (89, 95)

Systemic diseases:

·       Primary hyperparathyroidism

·       Sarcoidosis

·       Bone diseases (e.g. fractures, multiple myeloma)

·       Immobilization

·       Hyperthyroidism

·       Distal renal tubular acidosis

·       Polycystic kidney disease

·       Bowel disease (e.g. inflammatory bowel disease, chronic pancreatitis, chronic diarrhea)

·       Bowel resection (e.g. colon resection, ileostomy, small bowel resection, Roux-en-Y gastric bypass)

·       Cystic fibrosis

·       Primary hyperoxaluria

·       Gout

·       Anatomical abnormalities that impair urine flow (e.g. medullary sponge kidney, ureteral stricture, horseshoe kidney, etc.)

 

Medications

·       Carbonic anhydrase inhibitor (e.g. topiramate, acetazolamide, zonisamide)

·       Calcium and vitamin D supplements

·       Vitamin C supplement

·       Loop diuretics

·       Uricosuric agents (e.g. probenecid, benzbromarone)

 

Table 11. Risk Factors for Calcium Stones (89)

Risk factors for calcium oxalate stones

Risk factors for calcium phosphate stones

Low urine volume

Low urine volume

High urine calcium

High urine calcium

Low urine citrate

Low urine citrate

High urine oxalate

---

---

High urine pH

High urine uric acid

---

 

PATHOGENESIS AND RISK FACTORS

 

Hypercalciuria

 

Hypercalciuria is the most common risk factor of calcium stones and found in 30-60% of calcium stone formers (96). Hypercalciuria is classically defined as 24-hour urine calcium greater than 300 mg/day in men, greater than 250 mg/day in women, greater than 4 mg/kg body weight/day in either sex, or urine calcium > 140 mg calcium/gram creatinine/day (75).  Although threshold values are provided to define hypercalciuria, there is no threshold value that predicts risk of stone incidence or recurrence. Rather, risk of stone incidence and recurrence increases progressively with higher urinary calcium excretion (97). 

 

Environmental (diet, supplement, and medications) and metabolic disorders can contribute to hypercalciuria.  One way to determine causes of hypercalciuria is to divide it into three broad categories: hypercalcemic hypercalciuria, normocalcemic hypercalciuria, and hypocalcemic hypercalciuria (Table 12). 

 

Table 12. Causes of Hypercalciuria

Hypercalcemic hypercalciuria

·             PTH-dependent causes

Primary hyperparathyroidism

o   Lithium-induced hyperparathyroidism

·             PTH-independent causes

o   Granulomatous diseases (e.g. sarcoidosis, tuberculosis, histoplasmosis, coccidoimycosis, lymphoma)

o   Vitamin D toxicity

o   Low level or activity of vitamin D 24-hydroxylase

o   Hypercalcemia of malignancy (e.g. bone metastases, lymphoma, PTHrP, multiple myeloma) 

o   Immobilization

o   Hyperthyroidism

o   Paget’s disease of bone

o   Vitamin A toxicity

o   Milk alkali syndrome

 

Normocalcemic hypercalciuria

·             Absorptive

o   Excessive calcium intake (diet and/or supplement)

o   Excessive animal protein intake

o   Sarcoidosis

o   Idiopathic hypercalciuria

·             Resorptive

o   Excessive animal protein intake

o   Hyperthyroidism

o   Immobilization

o   Paget’s disease of bone

o   Osteoporosis

o   Glucocorticoid excess

o   Distal renal tubular acidosis

o   Malignant tumors

o   Idiopathic hypercalciuria

·       Renal leak

o   Excessive salt intake

o   Loop diuretics

o   Mineralocorticoid excess

o   Glucocorticoid excess

o   Distal renal tubular acidosis

o   Idiopathic hypercalciuria

 

Hypocalcemic hypercalciuria

·       Autosomal dominant hypocalcemia (activating mutation in CaSR or GNA11)

 

Idiopathic Hypercalciuria

 

Idiopathic hypercalciuria is found in up to 50% of idiopathic calcium stone formers (98).  It appears to be familial which suggests a genetic basis (99). It presents with a pattern of variable inheritance, and is likely polygenic in most stone formers, with described base changes in some candidate genes including CaSR, VDR, TRPV5, TRPV6, CLCN5, ADCY10, and CLDN14 (75, 96). The pathophysiology of idiopathic hypercalciuria involves increased intestinal calcium absorption, renal leak of calcium, and increased bone resorption especially when challenged with a restricted calcium diet (75, 96).

 

Intestinal calcium hyperabsorption is the most common abnormality in idiopathic hypercalciuria (100). It can be 1,25(OH)2D-dependent or independent. In patients with 1,25(OH)2D-dependent absorptive hypercalciuria, there is generally an increased production of 1,25(OH)2D when compared to normal individuals (101), although some patient with CYP24A1 mutations exhibit reduced 1,25(OH)2D catabolism. The exact mechanism leading to increased 1,25(OH)2D production remains unclear. There was a suggestion that renal tubular phosphate handling may play a role; however, others found that regulators of 1,25(OH)2D production (PTH, serum phosphorus and renal tubular reabsorption of phosphate) in idiopathic hypercalciuric stone formers were comparable to non-stone formers (102-105). In patients with 1,25(OH)2D-independent absorptive hypercalciuria, intestinal absorption of calcium remains elevated despite a normal 1,25(OH)2D level. Again, the mechanism is unclear.  Animal studies demonstrated an increased abundance and half-life of vitamin D receptors (VDR) in the intestines of genetic hypercalciuric rats (106, 107).  In male calcium oxalate stone formers with idiopathic hypercalciuria, an elevated level of VDR in monocytes was found when compared to non-stone formers (108).  Both suggest increased tissue VDR may contribute to absorptive hypercalciuria in individuals with a normal 1,25(OH)2D level.

 

Idiopathic hypercalciuric stone formers also display abnormal renal calcium handling with a lower postprandial renal calcium reabsorption than normal individuals without a difference in filtered load (109). There is evidence of defective renal calcium reabsorption in both proximal tubule and distal nephron (110-112). The reduced renal calcium reabsorption could not be explained by sodium excretion and PTH levels (109). The underlying mechanism of decreased renal calcium reabsorption remains to be elucidated. 

 

Hypercalciuric stone formers were found to have lower bone mineral density (BMD) than non-stone formers even in those with absorptive hypercalciuria (21, 113). The decreased BMD is more pronounced in trabecular bone than cortical bone (21, 113).  Nephrolithiasis was associated with an increased risk of vertebral fractures in both men and women in a population-based retrospective cohort study (114), and with an increased risk of prevalent vertebral and wrist fractures in men in a cross-sectional study in NHANES III (115).  Prior bone histomorphometry studies demonstrated increased bone resorption in fasting hypercalciuria and decreased bone formation in absorptive hypercalciuria (113). The pathophysiology underlying bone loss in idiopathic hypercalciuria is not exactly clear; however, several risk factors have been associated with bone loss in this population. A restricted calcium diet sometimes used by patients or physicians to reduce urine calcium may generate negative calcium balance leading to increased bone resorption and bone loss without reducing risk of kidney stones (77, 116, 117). High dietary salt and protein intake increase urinary calcium excretion and create a subtle metabolic acidosis both of which may contribute to bone loss (21, 75, 113, 118).  Inflammatory cytokines such as IL1, IL6, TNF-α and GM-CSF have been associated with hypercalciuria and bone loss by increased bone resorption (113).  Idiopathic hypercalciuric stone formers were also found to have increased bone expression of RANKL and decreased expression of TGF-β which may be the mediators for increased bone resorption and decreased bone formation and mineralization, respectively (119). High 1,25(OH)2D and/or increased expression of VDR found in idiopathic hypercalciuria may also increase bone resorption and decrease bone formation (21).

 

Hypocitraturia

 

Urinary citrate is an endogenous inhibitor of calcium stone formation.  It forms a more soluble calcium citrate complex than calcium oxalate and calcium phosphate (89, 96).  It reduces urinary supersaturation with respect to calcium oxalate and calcium phosphate (89, 96).  Hypocitraturia is generally defined as urine citrate less than 320 mg/day and is a well-described reversible risk factor that is present in 20-60% of calcium stone formers (89, 96). Extracellular and intracellular pH affects renal citrate excretion. Systemic acidosis increases urinary citrate reabsorption and leads to hypocitraturia (120).  Intracellular acidosis increases intracellular citrate metabolism in the cytosol and mitochondrial via the TCA cycle (120). Thus, hypocitraturia occurs mainly in conditions with extracellular or intracellular acidosis (89).  Table 13 summarizes common causes of hypocitraturia (89, 96, 120). 

 

Table 13. Causes of Hypocitraturia (89, 96, 120)

Systemic diseases:

·       Complete dRTA (hypocitraturia, frank metabolic acidosis, hypokalemia, hypercalciuria, high urine pH)

·       Incomplete dRTA (hypocitraturia without frank metabolic acidosis, high urine pH)

·       Hypokalemia

·       Chronic diarrhea

·       Chronic kidney disease

·       Primary hyperaldosteronism

·       Idiopathic hypocitraturia

Dietary:

·       High animal protein intake

·       High sodium intake

·       Low fruit / vegetable intake

Medications:

·       Carbonic anhydrase inhibitor (e.g. topiramate, acetazolamide, zonisamide)

·       Angiotensin converting enzyme inhibitors

·       Angiotensin II receptor blockers

·       Diuretic-induced hypokalemia

 

Hyperoxaluria

 

High urinary oxalate increases urinary supersaturation with respect to calcium oxalate (96). Hyperoxaluria is generally defined as urinary oxalate greater than 45 mg/day (0.5 mmol/day) (89).  It is encountered in 8-50% of calcium stone formers (121). Etiologies of hyperoxaluria can be divided into three categories: 1) increased endogenous oxalate production due to inborn error of metabolism, 2) increased intake of foods rich in oxalate or its precursors (Table 2) or increased intestinal bioavailability of oxalate, and 3) increased intestinal oxalate absorption (89, 121).  Table 14summarizes causes of hyperoxaluria.

 

Primary hyperoxalurias (PH) are a group of rare autosomal recessive disorders involving overproduction of oxalate which results in markedly increased urinary oxalate excretion. There are three genetic forms: PH1 due to mutations in AGXT (encodes for a pyridoxal-5’-phosphate-dependent hepatic peroxisomal alanine-glyoxylate aminotransferase, AGT), PH2 due to mutations in GRHPR (encodes for glyoxylate reductase and hydroxypyruvate reductase, GRHPR) and PH3 due to mutations in HOGA1 (encodes for hepatic mitochondrial 4-hydroxy-2-oxoglutarate aldolase, HOGA) (122, 123).  Deficiency in AGT in PH1 results in decreased conversion of glyoxylate to glycine. The accumulated glyoxylate is in turn converted to oxalate by lactate dehydrogenase (LDH) leading to increased production of oxalate (123). Deficiency in GRHPR in PH2 results in decreased conversion of glyoxylate and hydroxypyruvate to glycolate and D-glycerate, respectively. The accumulated glyoxylate and hydroxypyruvate are converted to oxalate and L-glycerate respectively by LDH (123). Mutations in HOGA1 in PH3 result in decreased enzymatic activity of HOGA which converts 4-hydroxy-2-oxoglutarate (HOG) to pyruvate and glyoxylate. This results in accumulation of HOG which inhibits mitochondrial GRHPR (which is the deficient enzyme in PH2) activity, thus increasing oxalate production (122, 124).  Primary hyperoxalurias should be suspected if urinary oxalate is greater than 90 mg/day (1 mmol/day) (125). Measurement of other urinary metabolites including glyoxylate and L-glycerate can be helpful, but genetic testing is required for definitive diagnosis of primary hyperoxaluria (123).

 

Dietary oxalate is estimated to range between 50 and 1,000 mg/day (121).  Oxalate is absorbed mainly in the small intestine and to a lesser extent in the colon (121).  Intestinal absorption of oxalate varies between 10% and 72% (121). On a normal calcium diet (1,000 mg/day calcium), urinary oxalate increases by 2.7 mg/day with every 100 mg/day increase in dietary oxalate between 50mg to 750mg/day (126). Table 2 provides a list of foods rich in oxalate and some alternative options with low oxalate content (24).  Although there is considerable variation in the reported oxalate content in foods among the available online sources, the simplest strategy is to avoid the foods with high oxalate content (127).  Attention should be paid to portion size even with foods with low to moderate oxalate contents.  Excessive intake of vitamin C more than 1,000 mg/day is associated with increased urine oxalate because vitamin C is metabolized into oxalate in the body (128, 129).

 

A restricted calcium diet increases enteric oxalate availability for absorption and results in increased urinary oxalate (85, 86). Patients with chronic diarrhea, pancreatic insufficiency, inflammatory bowel diseases, or small bowel resections may have malabsorption of bile acids and/or fatty acids which can complex with luminal calcium in the intestine, resulting in increased bioavailability of oxalate to be absorbed and subsequently excreted in the urine (89, 96). This is also termed enteric hyperoxaluria.  Increased bioavailability of luminal oxalate can also result from decreased colonization by Oxalobacter formigenes which is a Gram-negative obligate anaerobe that utilizes oxalate as the sole energy source. O. formigenes also increases secretion of endogenous oxalate from plasma to the gut lumen which results in decreased urinary oxalate (126). Colonization by O. formigenes is associated with decreased bioavailability of intestinal oxalate for absorption, decreased urinary oxalate, and reduced risk of calcium oxalate stones (130, 131).  Use of certain antibiotics to which O. formigenes are sensitive (macrolides, tetracyclines, chloramphenicol, rifampin and metronidazole) within the past 5 years is associated with a reduction in colonization when compared to non-users (132), and has been separately associated with greater incidence of stone disease (133). Furthermore, patients with cystic fibrosis or inflammatory bowel disease who receive frequent antibiotic courses were found to have lower prevalence of colonization by O. formigenes, which may contribute to their higher oxalate excretion and increased kidney stone formation (134, 135). 

 

Table 14. Causes of Hyperoxaluria

Primary hyperoxaluria

·       Primary hyperoxaluria type 1 (mutations in AGXT gene)

·       Primary hyperoxaluria type 2 (mutations in GRHPR gene)

·       Primary hyperoxaluria type 3 (mutations in HOGA1 gene)

Secondary hyperoxaluria

·       High intake of oxalate rich foods (See Table 2)

·       Vitamin C intake > 1,000 mg/day

·       Low calcium intake

·       Pancreatic insufficiency

·       Inflammatory bowel disease (Crohn’s disease)

·       Small bowel surgeries

·       Cystic fibrosis

·       Decreased colonization by Oxalobacter formigenes

 

Elevated Urine pH

 

Urine pH higher than 6.7 is a risk factor for calcium phosphate stones. The pKa for monohydrogen phosphate (HPO42-) is ~ 6.7.  At a pH higher than the pKa, there is an increased abundance of HPO42- which complexes with divalent cation calcium (Ca2+) to form brushite (CaHPO4.2H2O) and eventually to hydroxyapatite [Ca10(PO4)6(OH)2](96). Table 15 summarizes potential causes of high urine pH.

 

Table 15. Causes of High Urine pH (89, 96)

·       Distal renal tubular acidosis (RTA)

·       Urinary tract infections

·       Primary hyperparathyroidism

·       Carbonic anhydrase inhibitors

·       Alkali therapy 

 

Hyperuricosuria

 

Hyperuricosuria is defined as urinary uric acid greater than 800 mg/day in men and 750 mg/day in women (136).  It is found in 40% of calcium stone formers and associated with increased risk of calcium oxalate stones (96). There are three main proposed mechanisms by which hyperuricosuria promotes calcium oxalate crystallization: 1) calcium oxalate precipitation on monosodium urate crystals through heterogeneous nucleation (137, 138), 2) removal of calcium oxalate crystallization inhibitors by colloidal urate particles (139), and 3) increased urate concentration decreases the solubility of calcium oxalate and leads to precipitation of calcium oxalate from solution by a salting-out mechanism (140). Hyperuricosuria is generally caused by increased purine intake, increased production of uric acid, or increased urinary excretion primarily from acquired conditions, although inherited causes of hyperuricosuria are also described (Table 16) (89, 136).

 

Table 16. Causes of Hyperuricosuria (89, 136)

Acquired Conditions:

Increased intake:

·       High purine rich diet (e.g. red meat, fish and poultry)

Increased production:

·       Gout

·       Myeloproliferative and neoplastic disorders

Increased urinary excretion:

·       Uricosuric drugs

 

Inherited Conditions (rare):

Disorders of uric acid metabolism:

·       Lesch-Nyhan syndrome

·       Glycogen storage disease type 1A

Disorders of renal uric acid reabsorption:

·       Renal hypouricemia

 

MANAGEMENT OF CALCIUM STONES

 

Lifestyle Measures

 

A diet high in fluid (fluid intake of 2.3-3 liters per day or achieving urine volume of at least 2-2.5 liters per day), rich in fruits and vegetables, low in sodium (less than 2300 mg/day or 100 mmol/day), animal protein (limit to 0.8 to 1.0 g/kg body weight/day) and oxalate (less than 100 mg/day) and normal in calcium (1,000 to 1,200 mg/day preferably from dietary source) is recommended for calcium stone prevention (27-29, 95, 96).  These were previously addressed in section “General measures for all patients with kidney stones” (Table 8). 

 

Pharmacotherapy

 

Thiazide and thiazide-like diuretics:

 

Hypercalciuria is the most common risk factor for calcium stones. Thiazide (hydrochlorothiazide or HCTZ) and thiazide-like diuretics (indapamide and chlorthalidone) can reduce urinary calcium by two proposed mechanisms: 1) blockage of NaCl symporter in distal convoluted tubule leads to decreased distal sodium and water reabsorption and volume contraction, which results in increased sodium and water reabsorption in the renal proximal tubule, resulting in increased calcium reabsorption by passive transport (141), and 2) increased distal tubular calcium absorption by increased abundance of transport proteins TRVP5 and calbindins (142, 143). The hypocalciuric dose-response to HCTZ has been studied in six healthy adults which demonstrated hypocalciuric effect at 12.5mg, 25mg, and 50mg daily; however, it was subtherapeutic for 12.5mg and 25mg daily when compared to 50mg daily (144). Several randomized controlled trials on thiazide diuretics with an average follow up of ~ 3 years showed reduction in the risk of recurrent stones in both hypercalciuric and normocalciuric stone formers (145). This underscores the lack of threshold effect of urinary calcium in predicting stone risk. Rather, the risk of stone formation increases progressively with increasing urinary calcium excretion even within the “normal range” (97).  It also supports the empiric use of thiazide diuretics in recurrent calcium stone formers, even those with normocalciuria (27, 146). The doses used in these trials were HCTZ 25mg BID, 50mg and 100mg daily, indapamide 2.5mg daily and chlorthalidone 25mg and 50mg daily (145).  A recent retrospective cohort study suggests lower doses of thiazide diuretics (HCTZ or chlorthalidone ≤ 12.5 mg daily or indapamide ≤ 1.25 mg daily) appear to have a similar protective effect against stone formation as higher doses in older adults (147).

 

Thiazide diuretics have also been shown to improve bone health. They reduce urinary calcium excretion resulting in positive calcium balance and reduced PTH which may reduce bone turnover (96). They have also been shown to stimulate osteoblast differentiation and function and inhibit osteoclast differentiation in vitro (21, 115, 148).  HCTZ 50mg daily improved cortical BMD in healthy postmenopausal women without baseline hypercalciuria (149, 150). In postmenopausal women with osteoporosis and hypercalciuria, addition of indapamide 2.5mg daily to alendronate 70mg weekly resulted in a reduction in urinary calcium and an additional increase in BMD at the lumbar spine over 12 months compared to alendronate single therapy (151). A meta-analysis of five observational cohort studies showed thiazide diuretics use was associated with a reduction in risk of hip fractures (152). Although there is no randomized placebo-controlled trial available, a secondary analysis of the Antihypertensive and Lipid-Lowering Treatment to Prevent Heart Attack Trial (ALLHAT) showed a reduction in hip and pelvic fracture risk in patients on chlorthalidone 12.5mg to 25mg daily compared to those on lisinopril or amlodipine (153).

 

Thiazide diuretics use may be associated with hypokalemia which may induce hypocitraturia, thus potassium supplements are often needed. Potassium citrate is superior to potassium chloride given its ability to increase urinary citrate and pH and to further lower urine calcium excretion (154). Combination of thiazide diuretics with a potassium sparing diuretic (e.g. amiloride) can also be considered.

 

In summary, thiazide diuretics are recommended to patients with recurrent calcium stones with and without hypercalciuria (27).  

 

Alkali therapy:

 

Potassium citrate treatment increases urine pH and urine citrate, decreases urine calcium, and decreases urinary supersaturation with respect to calcium oxalate (155). Several placebo-controlled randomized trials demonstrated potassium citrate and potassium magnesium citrate treatment reduced recurrent stone events in calcium stone formers with and without hypocitraturia (156-158).  

 

Potassium citrate treatment may also prevent bone loss.  Potassium citrate treatment increased BMD at the spine in idiopathic calcium stone formers (159) and increased BMD at the spine, femoral neck, and total hip in healthy elderly men and postmenopausal women without osteoporosis (160).

 

The proposed mechanisms by which potassium citrate improves BMD include systemic alkalization, increased osteoblastic activity, and reduced osteoclastic activity demonstrated by bone turnover markers, and positive calcium balance created by reduced urinary calcium excretion (21, 160, 161).

 

Currently, potassium citrate therapy is recommended for patients with recurrent calcium stones with and without hypocitraturia (27).

 

Xanthine oxidase inhibitors: Allopurinol and Febuxostat:

 

There is only one published randomized placebo-controlled trial on allopurinol in prevention of calcium nephrolithiasis. In this study, calcium oxalate stone formers with hyperuricosuria and normocalciuria treated with allopurinol 100mg TID had a lower rate of stone events than those treated with placebo over 24 months (162).  Allopurinol is recommended for patients with recurrent calcium oxalate stones with hyperuricosuria and normocalciuria (27).

 

Febuxostat was studied in a randomized controlled trial comparing febuxostat 80mg daily with allopurinol 300mg daily or placebo on the effect of stone prevention in calcium stone formers (calcium oxalate and/or calcium phosphate) with hyperuricosuria and normocalciuria over 6 months (163). Febuxostat led to a greater reduction in urinary uric acid than allopurinol or placebo, but percent change in stone size was similar to allopurinol or placebo. This study was not powered to detect difference in stone events in the three groups.  Currently, there is insufficient evidence to support the routine use of febuxostat for stone prevention in hyperuricosuric calcium stone patients, except in those who may be allopurinol-intolerant (29).

 

Pyridoxine:

 

Pyridoxine (vitamin B6) supplementation is helpful in primary hyperoxaluria type 1 (PH1) with specific mutations, namely Gly170Arg, Phe152Ile and Ile244Thr.  A trial of pyridoxine for 3 months with a starting dose of 5 mg/kg body weight/day titrated to a maximum of 20 mg/kg body weight/day can be attempted in patients with suspected primary hyperoxaluria.  Response to therapy is defined as more than 30% reduction in urinary oxalate from baseline (123).

 

Uric Acid Stones

 

Uric acid stones generally represent around 10% of all stones analyzed, although their prevalence has markedly increased in recent years, in parallel with the diabetes and obesity epidemics (164, 165). In a series of 2,464 calculi, the proportion of uric acid stones was 35.7% in patients with type 2 diabetes and 11.3% in patients without type 2 diabetes (166). Reciprocally, the proportion of patients with type 2 diabetes was significantly higher among uric acid than among calcium stone formers (27.8 versus 6.9%) (166).  In fact, several epidemiological and metabolic studies have reported an association of uric acid stone disease with various features of the metabolic syndrome including obesity, type 2 diabetes, hypertension, dyslipidemia, hyperglycemia, hepatic steatosis, and greater visceral adiposity (167).

 

PATHOGENESIS AND RISK FACTORS

 

The three main factors implicated in the development of uric acid nephrolithiasis are low urine pH, hyperuricosuria, and low urine volume (Table 17) (168). Of these, low urine pH is the primary determinant of uric acid nephrolithiasis, as acidic urine favors the protonation of urate, forming relatively insoluble uric acid which precipitates in this overly acidic urinary environment. In fact, a decline in urine pH from 6.0 to 5.0 increases urinary uric acid concentration six-fold, whereas states of increased urate production typically increase urate excretion two-fold. Therefore, uric acid stone formation is more determined by pH than by urine volume or urine uric acid concentrations. Low urine pH may result from excessive intake of animal proteins (81), gastrointestinal alkali loss (from chronic diarrhea or laxative abuse), or may be idiopathic as frequently observed in patients with obesity, type 2 diabetes, and/or the metabolic syndrome (164, 168). Human metabolic studies have identified greater acid excretion and reduced urinary buffering by ammonia as the two culprits of aciduria in uric acid nephrolithiasis (169). Hyperuricosuria is less frequently encountered in patients with uric acid nephrolithiasis, and may result from inherited and/or acquired conditions (Tables 16 and 17). Finally, low urine volume due to extra-renal fluid losses contributes to increased urinary saturation with respect to uric acid, leading to stone formation.

 

Table 17. Risk Factors and Etiological Conditions Associated with Uric Acid Nephrolithiasis

Urinary Risk Factor

Type of Abnormality

Etiological conditions

Low urine pH

Inherited Conditions

Inherited uric acid lithiasis (unknown genetic abnormality)

Acquired Conditions

Metabolic syndrome, obesity, diabetes, CKD, high animal protein intake, gastrointestinal alkali loss

Medications

Laxatives

Hyperuricosuria

Inherited Conditions

Disorders of uric acid metabolism (e.g. Lesch-Nyhan);

Disorders of uric acid excretion (e.g. renal hypouricemia);

Glycogen storage disorder type 1A (glucose-6-phosphatase deficiency)

Acquired Conditions

Gout, myeloproliferative disorders, hemolytic disorders, high purine intake

Medications

Uricosuric agents: Losartan, Probenecid, Benzbromarone

Low urine volume

Acquired Conditions

Chronic diarrhea, excessive perspiration, low fluid intake

Medications

Laxatives

 

MANAGEMENT

 

Since uric acid stone formation is more determined by urine pH than by urine volume or urine uric acid concentrations, the cornerstone of therapy is urinary alkalinization.

 

Lifestyle Changes

 

Dietary restriction of animal protein intake is helpful in decreasing ingestion of proton sources, which reduces aciduria and aids with urinary alkalinization. At the same time, animal protein restriction also lowers uric acid excretion through a reduction in purine intake. Conversely, greater ingestion of alkali-rich fruits and vegetables aids in raising urine pH. Finally, higher fluid intake in general aids in raising urine volume, while intake of certain fruit juices such as orange juice can increase urine pH (along with the concomitant rise in urine volume). Still, one should be cautious about the sugar load imparted by fruit juices in uric acid stone formers with pre-diabetes or frank diabetes.

 

Pharmacological Therapy

 

Medical dissolution therapy of uric acid stones with alkali therapy (potassium citrate, to raise 24-hour urine pH to 6.0 to 6.5) is the cornerstone of uric acid management. Alkali therapy is well-tolerated by most uric acid stone formers and effectively dissolves stones, potentially avoiding the morbidity of urological interventions (170, 171). In patients who cannot tolerate potassium citrate, alternative alkali regimens include sodium bicarbonate and potassium bicarbonate. Occasionally, xanthine oxidase inhibitors (allopurinol or febuxostat) are added to potassium citrate in patients with hyperuricosuria whose uric acid stones recur despite alkali therapy. A recent study has suggested that the thiazolidinedione pioglitazone may aid in raising urine pH in uric acid stone formers (172), although the risk/benefit ratio of this medication needs to be considered.

 

Cystine Stones

 

PATHOGENESIS AND RISK FACTORS

 

Cystine represent around 1-2% of stones in adult patients, but account for 5-8% of stones in pediatric patients. Cystine stones result from inactivating mutations in genes that encode renal tubular transporters that reabsorb the amino acid cysteine (173). The complexation of two molecules of the dibasic amino acid cysteine results in the formation of cystine which is relatively insoluble. Cystine normally appears in urine in small amounts that are insufficient to cause supersaturation, crystalluria, or stone formation. Due to defects in renal cysteine reabsorption, patients with cystinuria exhibit greater than a 10-fold increase in urine cystine excretion (as well as greater excretion of the other dibasic amino acids lysine, ornithine, and arginine). As a result, the solubility limit of cystine in the urine (250 mg/L) is exceeded. Homozygous inheritance results in more severe phenotype, whereas heterozygous inheritance is associated with variable increases in amino acid excretion and an intermediate increase in cystinuria. Cystine stones form in the upper urinary tract as early as the first decade of life, and tend to be large, staghorn, bilateral, and highly recurrent (173). Stone formation may manifest as obstruction, infection, hematuria, and renal failure. Cystine stones are visible on standard abdominal radiographs because of the relative density of the sulfur constituent of cysteine (Table 6).

 

MANAGEMENT

 

The goal of therapy in cystinuria is to reduce cystine excretion and increase urinary cystine solubility (173, 174). This is accomplished using a combination of lifestyle changes and pharmacological interventions.

 

Lifestyle Changes

 

Large urine volumes of 3-4 liters per day may be effective at reducing cystine concentration and reducing stone recurrence in some patients, although this is difficult to institute in children and even adult patients. Dietary protein restriction to around 1 g protein/kg body weight/day reduces cysteine intake, and may cause small decreases in cystine synthesis (175), although this should be avoided in growing children and adolescents. A low sodium intake can also contribute to reduced cystine excretion (176).

 

Pharmacological Therapy

 

When fluid and dietary therapy fail, then pharmacologic therapy may be effective. Alkaline pH in the 7.0-7.5 range will reduce cystine solubility and can be achieved by the addition of alkali therapy such as potassium citrate (177). Tiopronin (Thiola®) and D-penicillamine reduce cystine formation in urine by preventing cysteine-cysteine complexation and the formation of more soluble thiol-cysteine disulfides that are more readily excreted in the urine. Both agents however have potentially serious side effects (proteinuria, abnormal LFTs, others) and therefore they are not used as first-line treatment (174).

 

Struvite (Infection) Stones

 

PATHOGENESIS AND RISK FACTORS

 

Struvite (magnesium ammonium phosphate) stones form only in the presence of bacteria that produce urease. Common urease-producing bacteria that may populate the urinary tract are proteus, klebsiella, pseudomonas, and enterococci. Urease-mediated splitting of urea and the generation of ammonium results in an alkaline urine. Urine pH above 7.0 normally is associated with very low urine ammonium levels of less than 10 mEq/day. However, urine ammonium excretion exceeding 30 mEq/day along with 24-hour urine pH > 7.0 virtually make the diagnosis of struvite stones. Other constituents of the stone may include calcium carbonate and brushite (calcium phosphate), which form crystals in the very alkaline urine. Patients who form struvite stones do not pass them spontaneously, but rather are at high risk for bleeding, obstruction, and decreased renal function. Some infection stones begin as calcium oxalate stones that become infected with a urease-producing bacterium. Spread of infection to the contralateral kidney may occur.

 

MANAGEMENT

 

Because untreated staghorn calculi will require nephrectomy in 50% of patients, definitive treatment is indicated (178). Growth of infection stones and their progressive damage to kidney tissue may be limited by shockwave lithotripsy and percutaneous nephrolithotomy (PCNL); however definitive treatment of struvite stones is surgical removal. Extended antibiotic therapy has proven ineffective in eradicating the infection and does not substitute for complete removal of even the smallest particulate of the stone (178). Management with PCNL followed by careful follow-up and medical management minimizes stone recurrence and maintains kidney function in the majority of patients (179). Larger stone burden pre-operatively, residual stones after surgery, and presence of medical comorbidities are independent risk factors for stone recurrence or residual stone-related events (179). Acetohydroxamic acid inhibits urease produced by the bacteria and has been shown to be effective in eradicating chronic infection of struvite stones (180). Use of the drug has been limited, however, as it is associated with potentially serious side effects such as hemolytic anemia and venous thromboembolic disease.

 

CONCLUSIONS

 

In conclusion, urinary stones are common, morbid, and highly recurrent. The pathophysiology of kidney stone formation is diverse, and includes a combination of genetic and environmental factors. Several endocrinological disorders increase the risk of stone formation. Metabolic evaluation of patients with kidney stones helps to identify the underlying etiological factors and provides an opportunity to institute preventive lifestyle and/or pharmacologic measures to reduce stone recurrence risk.

 

REFERENCES

 

  1. Stamatelou KK, Francis ME, Jones CA, Nyberg LM, Curhan GC. Time trends in reported prevalence of kidney stones in the United States: 1976-1994. Kidney Int. 2003;63(5):1817-23.
  2. Scales CD, Jr., Smith AC, Hanley JM, Saigal CS. Prevalence of kidney stones in the United States. Eur Urol. 2012;62(1):160-5.
  3. Ziemba JB, Matlaga BR. Epidemiology and economics of nephrolithiasis. Investig Clin Urol. 2017;58(5):299-306.
  4. Kittanamongkolchai W, Vaughan LE, Enders FT, Dhondup T, Mehta RA, Krambeck AE, et al. The Changing Incidence and Presentation of Urinary Stones Over 3 Decades. Mayo Clin Proc. 2018;93(3):291-9.
  5. Soucie JM, Coates RJ, McClellan W, Austin H, Thun M. Relation between geographic variability in kidney stones prevalence and risk factors for stones. Am J Epidemiol. 1996;143(5):487-95.
  6. Brikowski TH, Lotan Y, Pearle MS. Climate-related increase in the prevalence of urolithiasis in the United States. Proc Natl Acad Sci U S A. 2008;105(28):9841-6.
  7. Pearle MS, Calhoun EA, Curhan GC. Urologic diseases in America project: urolithiasis. J Urol. 2005;173(3):848-57.
  8. Antonelli JA, Maalouf NM, Pearle MS, Lotan Y. Use of the National Health and Nutrition Examination Survey to calculate the impact of obesity and diabetes on cost and prevalence of urolithiasis in 2030. Eur Urol. 2014;66(4):724-9.
  9. Rule AD, Lieske JC, Li X, Melton LJ, 3rd, Krambeck AE, Bergstralh EJ. The ROKS nomogram for predicting a second symptomatic stone episode. J Am Soc Nephrol. 2014;25(12):2878-86.
  10. Vaughan LE, Enders FT, Lieske JC, Pais VM, Rivera ME, Mehta RA, et al. Predictors of Symptomatic Kidney Stone Recurrence After the First and Subsequent Episodes. Mayo Clin Proc. 2019;94(2):202-10.
  11. Yongzhi L, Shi Y, Jia L, Yili L, Xingwang Z, Xue G. Risk factors for urinary tract infection in patients with urolithiasis-primary report of a single center cohort. BMC Urol. 2018;18(1):45.
  12. Lotan Y, Daudon M, Bruyere F, Talaska G, Strippoli G, Johnson RJ, et al. Impact of fluid intake in the prevention of urinary system diseases: a brief review. Curr Opin Nephrol Hypertens. 2013;22 Suppl 1:S1-10.
  13. Rocha H, Santos LC. Relapse of urinary tract infection in the presence of urinary tract calculi: the role of bacteria within the calculi. J Med Microbiol. 1969;2(3):372-6.
  14. Barr-Beare E, Saxena V, Hilt EE, Thomas-White K, Schober M, Li B, et al. The Interaction between Enterobacteriaceae and Calcium Oxalate Deposits. PLoS One. 2015;10(10):e0139575.
  15. Agarwal DK, Krambeck AE, Sharma V, Maldonado FJ, Westerman ME, Knoedler JJ, et al. Treatment of non-obstructive, non-struvite urolithiasis is effective in treatment of recurrent urinary tract infections. World J Urol. 2019.
  16. Keddis MT, Rule AD. Nephrolithiasis and loss of kidney function. Curr Opin Nephrol Hypertens. 2013;22(4):390-6.
  17. Dhondup T, Kittanamongkolchai W, Vaughan LE, Mehta RA, Chhina JK, Enders FT, et al. Risk of ESRD and Mortality in Kidney and Bladder Stone Formers. Am J Kidney Dis. 2018;72(6):790-7.
  18. Semins MJ, Matlaga BR. Kidney stones during pregnancy. Nat Rev Urol. 2014;11(3):163-8.
  19. Swartz MA, Lydon-Rochelle MT, Simon D, Wright JL, Porter MP. Admission for nephrolithiasis in pregnancy and risk of adverse birth outcomes. Obstet Gynecol. 2007;109(5):1099-104.
  20. Ross AE, Handa S, Lingeman JE, Matlaga BR. Kidney stones during pregnancy: an investigation into stone composition. Urol Res. 2008;36(2):99-102.
  21. Sakhaee K, Maalouf NM, Kumar R, Pasch A, Moe OW. Nephrolithiasis-associated bone disease: pathogenesis and treatment options. Kidney Int. 2011;79(4):393-403.
  22. Sakhaee K. Nephrolithiasis as a systemic disorder. Curr Opin Nephrol Hypertens. 2008;17(3):304-9.
  23. Devarajan A. Cross-talk between renal lithogenesis and atherosclerosis: an unveiled link between kidney stone formation and cardiovascular diseases. Clin Sci (Lond). 2018;132(6):615-26.
  24. Oxalate contents of foods 2017 [Available from: https://regepi.bwh.harvard.edu/health/Oxalate/files.
  25. Daudon M, Frochot V, Bazin D, Jungers P. Drug-Induced Kidney Stones and Crystalline Nephropathy: Pathophysiology, Prevention and Treatment. Drugs. 2018;78(2):163-201.
  26. Matlaga BR, Shah OD, Assimos DG. Drug-induced urinary calculi. Rev Urol. 2003;5(4):227-31.
  27. Pearle MS, Goldfarb DS, Assimos DG, Curhan G, Denu-Ciocca CJ, Matlaga BR, et al. Medical management of kidney stones: AUA guideline. J Urol. 2014;192(2):316-24.
  28. Turk C, Petrik A, Sarica K, Seitz C, Skolarikos A, Straub M, et al. EAU Guidelines on Diagnosis and Conservative Management of Urolithiasis. Eur Urol. 2016;69(3):468-74.
  29. Dion M, Ankawi G, Chew B, Paterson R, Sultan N, Hoddinott P, et al. CUA guideline on the evaluation and medical management of the kidney stone patient - 2016 update. Can Urol Assoc J. 2016;10(11-12):E347-E58.
  30. Healy KA, Hubosky SG, Bagley DH. 24-hour urine collection in the metabolic evaluation of stone formers: is one study adequate? J Endourol. 2013;27(3):374-8.
  31. Nayan M, Elkoushy MA, Andonian S. Variations between two 24-hour urine collections in patients presenting to a tertiary stone clinic. Can Urol Assoc J. 2012;6(1):30-3.
  32. Pak CY, Peterson R, Poindexter JR. Adequacy of a single stone risk analysis in the medical evaluation of urolithiasis. J Urol. 2001;165(2):378-81.
  33. Parks JH, Goldfisher E, Asplin JR, Coe FL. A single 24-hour urine collection is inadequate for the medical evaluation of nephrolithiasis. J Urol. 2002;167(4):1607-12.
  34. Castle SM, Cooperberg MR, Sadetsky N, Eisner BH, Stoller ML. Adequacy of a single 24-hour urine collection for metabolic evaluation of recurrent nephrolithiasis. J Urol. 2010;184(2):579-83.
  35. Werness PG, Brown CM, Smith LH, Finlayson B. EQUIL2: a BASIC computer program for the calculation of urinary saturation. The Journal of urology. 1985;134(6):1242-4.
  36. Parks JH, Coward M, Coe FL. Correspondence between stone composition and urine supersaturation in nephrolithiasis. Kidney Int. 1997;51(3):894-900.
  37. Prochaska M, Taylor E, Ferraro PM, Curhan G. Relative Supersaturation of 24-Hour Urine and Likelihood of Kidney Stones. J Urol. 2018;199(5):1262-6.
  38. Assimos D, Krambeck A, Miller NL, Monga M, Murad MH, Nelson CP, et al. Surgical Management of Stones: American Urological Association/Endourological Society Guideline, PART II. J Urol. 2016;196(4):1161-9.
  39. Pak CY, Poindexter JR, Adams-Huet B, Pearle MS. Predictive value of kidney stone composition in the detection of metabolic abnormalities. Am J Med. 2003;115(1):26-32.
  40. Lee TT, Elkoushy MA, Andonian S. Are stone analysis results different with repeated sampling? Can Urol Assoc J. 2014;8(5-6):E317-22.
  41. Mandel N, Mandel I, Fryjoff K, Rejniak T, Mandel G. Conversion of calcium oxalate to calcium phosphate with recurrent stone episodes. The Journal of urology. 2003;169(6):2026-9.
  42. Kadlec AO, Fridirici ZC, Acosta-Miranda AM, Will TH, Sakamoto K, Turk TM. Bilateral urinary calculi with discordant stone composition. World J Urol. 2014;32(1):281-5.
  43. Rivera ME, Nottingham CU, Borofsky MS, Kissel SM, Maniar V, Dauw CA, et al. Variability in stone composition and metabolic correlation between kidneys in patients with bilateral nephrolithiasis. Int Urol Nephrol. 2019.
  44. Hesse A, Kruse R, Geilenkeuser WJ, Schmidt M. Quality control in urinary stone analysis: results of 44 ring trials (1980-2001). Clin Chem Lab Med. 2005;43(3):298-303.
  45. Basiri A, Taheri M, Taheri F. What is the state of the stone analysis techniques in urolithiasis? Urol J. 2012;9(2):445-54.
  46. Mandel NS, Mandel IC, Kolbach-Mandel AM. Accurate stone analysis: the impact on disease diagnosis and treatment. Urolithiasis. 2017;45(1):3-9.
  47. Siener R, Buchholz N, Daudon M, Hess B, Knoll T, Osther PJ, et al. Quality Assessment of Urinary Stone Analysis: Results of a Multicenter Study of Laboratories in Europe. PLoS One. 2016;11(6):e0156606.
  48. Krambeck AE, Khan NF, Jackson ME, Lingeman JE, McAteer JA, Williams JC, Jr. Inaccurate reporting of mineral composition by commercial stone analysis laboratories: implications for infection and metabolic stones. J Urol. 2010;184(4):1543-9.
  49. Poletti PA, Platon A, Rutschmann OT, Schmidlin FR, Iselin CE, Becker CD. Low-dose versus standard-dose CT protocol in patients with clinically suspected renal colic. AJR Am J Roentgenol. 2007;188(4):927-33.
  50. Wilhelm K, Schoenthaler M, Hein S, Adams F, Schlager D, Kuehhas FE, et al. Focused Dual-energy CT Maintains Diagnostic and Compositional Accuracy for Urolithiasis Using Ultralow-dose Noncontrast CT. Urology. 2015;86(6):1097-102.
  51. Smith-Bindman R, Aubin C, Bailitz J, Bengiamin RN, Camargo CA, Jr., Corbo J, et al. Ultrasonography versus computed tomography for suspected nephrolithiasis. N Engl J Med. 2014;371(12):1100-10.
  52. Hubner WA, Irby P, Stoller ML. Natural history and current concepts for the treatment of small ureteral calculi. Eur Urol. 1993;24(2):172-6.
  53. Miller OF, Kane CJ. Time to stone passage for observed ureteral calculi: a guide for patient education. J Urol. 1999;162(3 Pt 1):688-90; discussion 90-1.
  54. NICE Guideline - Renal and ureteric stones: assessment and management: NICE (2019) Renal and ureteric stones: assessment and management. BJU Int. 2019;123(2):220-32.
  55. Parmar MS. Kidney stones. BMJ. 2004;328(7453):1420-4.
  56. Segura JW, Preminger GM, Assimos DG, Dretler SP, Kahn RI, Lingeman JE, et al. Ureteral Stones Clinical Guidelines Panel summary report on the management of ureteral calculi. The American Urological Association. J Urol. 1997;158(5):1915-21.
  57. Afshar K, Jafari S, Marks AJ, Eftekhari A, MacNeily AE. Nonsteroidal anti-inflammatory drugs (NSAIDs) and non-opioids for acute renal colic. Cochrane Database Syst Rev. 2015(6):CD006027.
  58. Gottlieb M, Long B, Koyfman A. The evaluation and management of urolithiasis in the ED: A review of the literature. Am J Emerg Med. 2018;36(4):699-706.
  59. Minhaj FS, Hoang-Nguyen M, Tenney A, Bragg A, Zhang W, Foster J, et al. Evaluation of opioid requirements in the management of renal colic after guideline implementation in the emergency department. Am J Emerg Med. 2019.
  60. Soleimanpour H, Hassanzadeh K, Vaezi H, Golzari SE, Esfanjani RM, Soleimanpour M. Effectiveness of intravenous lidocaine versus intravenous morphine for patients with renal colic in the emergency department. BMC Urol. 2012;12:13.
  61. Sterrett SP, Nakada SY. Medical expulsive therapy. Curr Opin Urol. 2008;18(2):210-3.
  62. Campschroer T, Zhu X, Vernooij RW, Lock MT. Alpha-blockers as medical expulsive therapy for ureteral stones. Cochrane Database Syst Rev. 2018;4:CD008509.
  63. Meltzer AC, Burrows PK, Wolfson AB, Hollander JE, Kurz M, Kirkali Z, et al. Effect of Tamsulosin on Passage of Symptomatic Ureteral Stones: A Randomized Clinical Trial. JAMA Intern Med. 2018;178(8):1051-7.
  64. Ziemba JB, Matlaga BR. Guideline of guidelines: kidney stones. BJU Int. 2015;116(2):184-9.
  65. Pak CY, Sakhaee K, Crowther C, Brinkley L. Evidence justifying a high fluid intake in treatment of nephrolithiasis. Ann Intern Med. 1980;93(1):36-9.
  66. Borghi L, Meschi T, Maggiore U, Prati B. Dietary therapy in idiopathic nephrolithiasis. Nutr Rev. 2006;64(7 Pt 1):301-12.
  67. Borghi L, Meschi T, Amato F, Briganti A, Novarini A, Giannini A. Urinary volume, water and recurrences in idiopathic calcium nephrolithiasis: a 5-year randomized prospective study. J Urol. 1996;155(3):839-43.
  68. Littlejohns TJ, Neal NL, Bradbury KE, Heers H, Allen NE, Turney BW. Fluid Intake and Dietary Factors and the Risk of Incident Kidney Stones in UK Biobank: A Population-based Prospective Cohort Study. Eur Urol Focus. 2019.
  69. Curhan GC, Willett WC, Rimm EB, Stampfer MJ. A prospective study of dietary calcium and other nutrients and the risk of symptomatic kidney stones. N Engl J Med. 1993;328(12):833-8.
  70. Curhan GC, Willett WC, Rimm EB, Spiegelman D, Stampfer MJ. Prospective study of beverage use and the risk of kidney stones. Am J Epidemiol. 1996;143(3):240-7.
  71. Kang DE, Sur RL, Haleblian GE, Fitzsimons NJ, Borawski KM, Preminger GM. Long-term lemonade based dietary manipulation in patients with hypocitraturic nephrolithiasis. J Urol. 2007;177(4):1358-62; discussion 62; quiz 591.
  72. Wabner CL, Pak CY. Effect of orange juice consumption on urinary stone risk factors. J Urol. 1993;149(6):1405-8.
  73. Sorensen MD, Kahn AJ, Reiner AP, Tseng TY, Shikany JM, Wallace RB, et al. Impact of nutritional factors on incident kidney stone formation: a report from the WHI OS. J Urol. 2012;187(5):1645-9.
  74. Sakhaee K, Harvey JA, Padalino PK, Whitson P, Pak CY. The potential role of salt abuse on the risk for kidney stone formation. J Urol. 1993;150(2 Pt 1):310-2.
  75. Worcester EM, Coe FL. New insights into the pathogenesis of idiopathic hypercalciuria. Semin Nephrol. 2008;28(2):120-32.
  76. Nouvenne A, Meschi T, Prati B, Guerra A, Allegri F, Vezzoli G, et al. Effects of a low-salt diet on idiopathic hypercalciuria in calcium-oxalate stone formers: a 3-mo randomized controlled trial. Am J Clin Nutr. 2010;91(3):565-70.
  77. Borghi L, Schianchi T, Meschi T, Guerra A, Allegri F, Maggiore U, et al. Comparison of two diets for the prevention of recurrent stones in idiopathic hypercalciuria. N Engl J Med. 2002;346(2):77-84.
  78. Robertson WG, Peacock M, Marshall DH. Prevalence of urinary stone disease in vegetarians. Eur Urol. 1982;8(6):334-9.
  79. Breslau NA, Brinkley L, Hill KD, Pak CY. Relationship of animal protein-rich diet to kidney stone formation and calcium metabolism. J Clin Endocrinol Metab. 1988;66(1):140-6.
  80. Kerstetter JE, O'Brien KO, Insogna KL. Low protein intake: the impact on calcium and bone homeostasis in humans. J Nutr. 2003;133(3):855S-61S.
  81. Maalouf NM, Moe OW, Adams-Huet B, Sakhaee K. Hypercalciuria associated with high dietary protein intake is not due to acid load. J Clin Endocrinol Metab. 2011;96(12):3733-40.
  82. Monsen ER. The 10th edition of the Recommended Dietary Allowances: what's new in the 1989 RDAs? J Am Diet Assoc. 1989;89(12):1748-52.
  83. Bleich HL, Moore MJ, Lemann J, Jr., Adams ND, Gray RW. Urinary calcium excretion in human beings. N Engl J Med. 1979;301(10):535-41.
  84. Taylor EN, Stampfer MJ, Curhan GC. Dietary factors and the risk of incident kidney stones in men: new insights after 14 years of follow-up. J Am Soc Nephrol. 2004;15(12):3225-32.
  85. von Unruh GE, Voss S, Sauerbruch T, Hesse A. Dependence of oxalate absorption on the daily calcium intake. J Am Soc Nephrol. 2004;15(6):1567-73.
  86. Zarembski PM, Hodgkinson A. Some factors influencing the urinary excretion of oxalic acid in man. Clin Chim Acta. 1969;25(1):1-10.
  87. Sorensen MD, Hsi RS, Chi T, Shara N, Wactawski-Wende J, Kahn AJ, et al. Dietary intake of fiber, fruit and vegetables decreases the risk of incident kidney stones in women: a Women's Health Initiative report. J Urol. 2014;192(6):1694-9.
  88. Meschi T, Maggiore U, Fiaccadori E, Schianchi T, Bosi S, Adorni G, et al. The effect of fruits and vegetables on urinary stone risk factors. Kidney Int. 2004;66(6):2402-10.
  89. Maalouf N. Approach to the Adult Kidney Stone Former. Clin Rev Bone Miner Metab. 2012;10(1):38-49.
  90. Bird VY, Khan SR. How do stones form? Is unification of theories on stone formation possible? Arch Esp Urol. 2017;70(1):12-27.
  91. Bushinsky DA. Nephrolithiasis: site of the initial solid phase. J Clin Invest. 2003;111(5):602-5.
  92. Evan AP. Physiopathology and etiology of stone formation in the kidney and the urinary tract. Pediatr Nephrol. 2010;25(5):831-41.
  93. Evan AP, Lingeman JE, Coe FL, Parks JH, Bledsoe SB, Shao Y, et al. Randall's plaque of patients with nephrolithiasis begins in basement membranes of thin loops of Henle. J Clin Invest. 2003;111(5):607-16.
  94. Linnes MP, Krambeck AE, Cornell L, Williams JC, Jr., Korinek M, Bergstralh EJ, et al. Phenotypic characterization of kidney stone formers by endoscopic and histological quantification of intrarenal calcification. Kidney Int. 2013;84(4):818-25.
  95. Worcester EM, Coe FL. Clinical practice. Calcium kidney stones. N Engl J Med. 2010;363(10):954-63.
  96. Sakhaee K, Maalouf NM, Sinnott B. Clinical review. Kidney stones 2012: pathogenesis, diagnosis, and management. J Clin Endocrinol Metab. 2012;97(6):1847-60.
  97. Parvin M, Shakhssalim N, Basiri A, Miladipour AH, Golestan B, Mohammadi Torbati P, et al. The most important metabolic risk factors in recurrent urinary stone formers. Urol J. 2011;8(2):99-106.
  98. Coe FL, Worcester EM, Evan AP. Idiopathic hypercalciuria and formation of calcium renal stones. Nat Rev Nephrol. 2016;12(9):519-33.
  99. Coe FL, Parks JH, Moore ES. Familial idiopathic hypercalciuria. N Engl J Med. 1979;300(7):337-40.
  100. Pak CY, Oata M, Lawrence EC, Snyder W. The hypercalciurias. Causes, parathyroid functions, and diagnostic criteria. J Clin Invest. 1974;54(2):387-400.
  101. Insogna KL, Broadus AE, Dreyer BE, Ellison AF, Gertner JM. Elevated production rate of 1,25-dihydroxyvitamin D in patients with absorptive hypercalciuria. J Clin Endocrinol Metab. 1985;61(3):490-5.
  102. D.E. Barilla JEZ, C.Y.C. Pak A critical evaluation of the role of phosphate in the pathogenesis of absorptive hypercalciuria. Mineral and Electrolyte Metabolism. 1979;2(6):302-9.
  103. Van Den Berg CJ, Kumar R, Wilson DM, Heath H, 3rd, Smith LH. Orthophosphate therapy decreases urinary calcium excretion and serum 1,25-dihydroxyvitamin D concentrations in idiopathic hypercalciuria. J Clin Endocrinol Metab. 1980;51(5):998-1001.
  104. Dhayat NA, Luthi D, Schneider L, Mattmann C, Vogt B, Fuster DG. Distinct phenotype of kidney stone formers with renal phosphate leak. Nephrol Dial Transplant. 2019;34(1):129-37.
  105. Negri AL, Spivacow R, Del Valle E, Fradinger E, Marino A, Zanchetta JR. Renal phosphate leak in patients with idiopathic hypercalciuria and calcium nephrolithiasis. Urol Res. 2003;31(6):378-81.
  106. Karnauskas AJ, van Leeuwen JP, van den Bemd GJ, Kathpalia PP, DeLuca HF, Bushinsky DA, et al. Mechanism and function of high vitamin D receptor levels in genetic hypercalciuric stone-forming rats. J Bone Miner Res. 2005;20(3):447-54.
  107. Li XQ, Tembe V, Horwitz GM, Bushinsky DA, Favus MJ. Increased intestinal vitamin D receptor in genetic hypercalciuric rats. A cause of intestinal calcium hyperabsorption. J Clin Invest. 1993;91(2):661-7.
  108. Favus MJ, Karnauskas AJ, Parks JH, Coe FL. Peripheral blood monocyte vitamin D receptor levels are elevated in patients with idiopathic hypercalciuria. J Clin Endocrinol Metab. 2004;89(10):4937-43.
  109. Worcester EM, Gillen DL, Evan AP, Parks JH, Wright K, Trumbore L, et al. Evidence that postprandial reduction of renal calcium reabsorption mediates hypercalciuria of patients with calcium nephrolithiasis. Am J Physiol Renal Physiol. 2007;292(1):F66-75.
  110. Sakhaee K, Nicar MJ, Brater DC, Pak CY. Exaggerated natriuretic and calciuric responses to hydrochlorothiazide in renal hypercalciuria but not in absorptive hypercalciuria. J Clin Endocrinol Metab. 1985;61(5):825-9.
  111. Sutton RA, Walker VR. Responses to hydrochlorothiazide and acetazolamide in patients with calcium stones. Evidence suggesting a defect in renal tubular function. N Engl J Med. 1980;302(13):709-13.
  112. Worcester EM, Coe FL, Evan AP, Bergsland KJ, Parks JH, Willis LR, et al. Evidence for increased postprandial distal nephron calcium delivery in hypercalciuric stone-forming patients. Am J Physiol Renal Physiol. 2008;295(5):F1286-94.
  113. Zerwekh JE. Bone disease and idiopathic hypercalciuria. Semin Nephrol. 2008;28(2):133-42.
  114. Melton LJ, 3rd, Crowson CS, Khosla S, Wilson DM, O'Fallon WM. Fracture risk among patients with urolithiasis: a population-based cohort study. Kidney Int. 1998;53(2):459-64.
  115. Lauderdale DS, Thisted RA, Wen M, Favus MJ. Bone mineral density and fracture among prevalent kidney stone cases in the Third National Health and Nutrition Examination Survey. J Bone Miner Res. 2001;16(10):1893-8.
  116. Fuss M, Pepersack T, Bergman P, Hurard T, Simon J, Corvilain J. Low calcium diet in idiopathic urolithiasis: a risk factor for osteopenia as great as in primary hyperparathyroidism. Br J Urol. 1990;65(6):560-3.
  117. Hess B. Low calcium diet in hypercalciuric calcium nephrolithiasis: first do no harm. Scanning Microsc. 1996;10(2):547-54; discussion 54-6.
  118. Frings-Meuthen P, Baecker N, Heer M. Low-grade metabolic acidosis may be the cause of sodium chloride-induced exaggerated bone resorption. J Bone Miner Res. 2008;23(4):517-24.
  119. Gomes SA, dos Reis LM, Noronha IL, Jorgetti V, Heilberg IP. RANKL is a mediator of bone resorption in idiopathic hypercalciuria. Clin J Am Soc Nephrol. 2008;3(5):1446-52.
  120. Zuckerman JM, Assimos DG. Hypocitraturia: pathophysiology and medical management. Rev Urol. 2009;11(3):134-44.
  121. Sakhaee K. Recent advances in the pathophysiology of nephrolithiasis. Kidney Int. 2009;75(6):585-95.
  122. Fargue S, Milliner DS, Knight J, Olson JB, Lowther WT, Holmes RP. Hydroxyproline Metabolism and Oxalate Synthesis in Primary Hyperoxaluria. J Am Soc Nephrol. 2018;29(6):1615-23.
  123. Cochat P, Rumsby G. Primary hyperoxaluria. N Engl J Med. 2013;369(7):649-58.
  124. Riedel TJ, Knight J, Murray MS, Milliner DS, Holmes RP, Lowther WT. 4-Hydroxy-2-oxoglutarate aldolase inactivity in primary hyperoxaluria type 3 and glyoxylate reductase inhibition. Biochim Biophys Acta. 2012;1822(10):1544-52.
  125. Hoppe B. An update on primary hyperoxaluria. Nat Rev Nephrol. 2012;8(8):467-75.
  126. Mitchell T, Kumar P, Reddy T, Wood KD, Knight J, Assimos DG, et al. Dietary oxalate and kidney stone formation. Am J Physiol Renal Physiol. 2019;316(3):F409-F13.
  127. Attalla K, De S, Monga M. Oxalate content of food: a tangled web. Urology. 2014;84(3):555-60.
  128. Baxmann AC, De OGMC, Heilberg IP. Effect of vitamin C supplements on urinary oxalate and pH in calcium stone-forming patients. Kidney Int. 2003;63(3):1066-71.
  129. Traxer O, Huet B, Poindexter J, Pak CY, Pearle MS. Effect of ascorbic acid consumption on urinary stone risk factors. J Urol. 2003;170(2 Pt 1):397-401.
  130. Kaufman DW, Kelly JP, Curhan GC, Anderson TE, Dretler SP, Preminger GM, et al. Oxalobacter formigenes may reduce the risk of calcium oxalate kidney stones. J Am Soc Nephrol. 2008;19(6):1197-203.
  131. Siener R, Bangen U, Sidhu H, Honow R, von Unruh G, Hesse A. The role of Oxalobacter formigenes colonization in calcium oxalate stone disease. Kidney Int. 2013;83(6):1144-9.
  132. Kelly JP, Curhan GC, Cave DR, Anderson TE, Kaufman DW. Factors related to colonization with Oxalobacter formigenes in U.S. adults. J Endourol. 2011;25(4):673-9.
  133. Tasian GE, Jemielita T, Goldfarb DS, Copelovitch L, Gerber JS, Wu Q, et al. Oral Antibiotic Exposure and Kidney Stone Disease. J Am Soc Nephrol. 2018;29(6):1731-40.
  134. Kumar R, Ghoshal UC, Singh G, Mittal RD. Infrequency of colonization with Oxalobacter formigenes in inflammatory bowel disease: possible role in renal stone formation. J Gastroenterol Hepatol. 2004;19(12):1403-9.
  135. Sidhu H, Hoppe B, Hesse A, Tenbrock K, Bromme S, Rietschel E, et al. Absence of Oxalobacter formigenes in cystic fibrosis patients: a risk factor for hyperoxaluria. Lancet. 1998;352(9133):1026-9.
  136. Wiederkehr MR, Moe OW. Uric Acid Nephrolithiasis: A Systemic Metabolic Disorder. Clin Rev Bone Miner Metab. 2011;9(3-4):207-17.
  137. Coe FL, Lawton RL, Goldstein RB, Tembe V. Sodium urate accelerates precipitation of calcium oxalate in vitro. Proc Soc Exp Biol Med. 1975;149(4):926-9.
  138. Pak CY, Arnold LH. Heterogeneous nucleation of calcium oxalate by seeds of monosodium urate. Proc Soc Exp Biol Med. 1975;149(4):930-2.
  139. Zerwekh JE, Holt K, Pak CY. Natural urinary macromolecular inhibitors: attenuation of inhibitory activity by urate salts. Kidney Int. 1983;23(6):838-41.
  140. Grover PK, Marshall VR, Ryall RL. Dissolved urate salts out calcium oxalate in undiluted human urine in vitro: implications for calcium oxalate stone genesis. Chem Biol. 2003;10(3):271-8.
  141. Nijenhuis T, Vallon V, van der Kemp AW, Loffing J, Hoenderop JG, Bindels RJ. Enhanced passive Ca2+ reabsorption and reduced Mg2+ channel abundance explains thiazide-induced hypocalciuria and hypomagnesemia. J Clin Invest. 2005;115(6):1651-8.
  142. Jang HR, Kim S, Heo NJ, Lee JH, Kim HS, Nielsen S, et al. Effects of thiazide on the expression of TRPV5, calbindin-D28K, and sodium transporters in hypercalciuric rats. J Korean Med Sci. 2009;24 Suppl:S161-9.
  143. Lee CT, Shang S, Lai LW, Yong KC, Lien YH. Effect of thiazide on renal gene expression of apical calcium channels and calbindins. Am J Physiol Renal Physiol. 2004;287(6):F1164-70.
  144. Martins MC, Meyers AM, Whalley NA, Margolius LP, Buys ME. Indapamide (Natrilix): the agent of choice in the treatment of recurrent renal calculi associated with idiopathic hypercalciuria. Br J Urol. 1996;78(2):176-80.
  145. Reilly RF, Peixoto AJ, Desir GV. The evidence-based use of thiazide diuretics in hypertension and nephrolithiasis. Clin J Am Soc Nephrol. 2010;5(10):1893-903.
  146. Goldfarb DS. Empiric therapy for kidney stones. Urolithiasis. 2019;47(1):107-13.
  147. Alexander RT, McArthur E, Jandoc R, Welk B, Fuster DG, Garg AX, et al. Thiazide Diuretic Dose and Risk of Kidney Stones in Older Adults: A Retrospective Cohort Study. Can J Kidney Health Dis. 2018;5:2054358118787480.
  148. Dvorak MM, De Joussineau C, Carter DH, Pisitkun T, Knepper MA, Gamba G, et al. Thiazide diuretics directly induce osteoblast differentiation and mineralized nodule formation by interacting with a sodium chloride co-transporter in bone. J Am Soc Nephrol. 2007;18(9):2509-16.
  149. Bolland MJ, Ames RW, Horne AM, Orr-Walker BJ, Gamble GD, Reid IR. The effect of treatment with a thiazide diuretic for 4 years on bone density in normal postmenopausal women. Osteoporos Int. 2007;18(4):479-86.
  150. Reid IR, Ames RW, Orr-Walker BJ, Clearwater JM, Horne AM, Evans MC, et al. Hydrochlorothiazide reduces loss of cortical bone in normal postmenopausal women: a randomized controlled trial. Am J Med. 2000;109(5):362-70.
  151. Giusti A, Barone A, Pioli G, Girasole G, Siccardi V, Palummeri E, et al. Alendronate and indapamide alone or in combination in the management of hypercalciuria associated with osteoporosis: a randomized controlled trial of two drugs and three treatments. Nephrol Dial Transplant. 2009;24(5):1472-7.
  152. Aung K, Htay T. Thiazide diuretics and the risk of hip fracture. Cochrane Database Syst Rev. 2011(10):CD005185.
  153. Puttnam R, Davis BR, Pressel SL, Whelton PK, Cushman WC, Louis GT, et al. Association of 3 Different Antihypertensive Medications With Hip and Pelvic Fracture Risk in Older Adults: Secondary Analysis of a Randomized Clinical Trial. JAMA Intern Med. 2017;177(1):67-76.
  154. Nicar MJ, Peterson R, Pak CY. Use of potassium citrate as potassium supplement during thiazide therapy of calcium nephrolithiasis. The Journal of urology. 1984;131(3):430-3.
  155. Sakhaee K, Nicar M, Hill K, Pak CY. Contrasting effects of potassium citrate and sodium citrate therapies on urinary chemistries and crystallization of stone-forming salts. Kidney Int. 1983;24(3):348-52.
  156. Barcelo P, Wuhl O, Servitge E, Rousaud A, Pak CY. Randomized double-blind study of potassium citrate in idiopathic hypocitraturic calcium nephrolithiasis. J Urol. 1993;150(6):1761-4.
  157. Ettinger B, Pak CY, Citron JT, Thomas C, Adams-Huet B, Vangessel A. Potassium-magnesium citrate is an effective prophylaxis against recurrent calcium oxalate nephrolithiasis. J Urol. 1997;158(6):2069-73.
  158. Soygur T, Akbay A, Kupeli S. Effect of potassium citrate therapy on stone recurrence and residual fragments after shockwave lithotripsy in lower caliceal calcium oxalate urolithiasis: a randomized controlled trial. J Endourol. 2002;16(3):149-52.
  159. Pak CY, Peterson RD, Poindexter J. Prevention of spinal bone loss by potassium citrate in cases of calcium urolithiasis. J Urol. 2002;168(1):31-4.
  160. Jehle S, Hulter HN, Krapf R. Effect of potassium citrate on bone density, microarchitecture, and fracture risk in healthy older adults without osteoporosis: a randomized controlled trial. J Clin Endocrinol Metab. 2013;98(1):207-17.
  161. Moseley KF, Weaver CM, Appel L, Sebastian A, Sellmeyer DE. Potassium citrate supplementation results in sustained improvement in calcium balance in older men and women. J Bone Miner Res. 2013;28(3):497-504.
  162. Ettinger B, Tang A, Citron JT, Livermore B, Williams T. Randomized trial of allopurinol in the prevention of calcium oxalate calculi. N Engl J Med. 1986;315(22):1386-9.
  163. Goldfarb DS, MacDonald PA, Gunawardhana L, Chefo S, McLean L. Randomized controlled trial of febuxostat versus allopurinol or placebo in individuals with higher urinary uric acid excretion and calcium stones. Clin J Am Soc Nephrol. 2013;8(11):1960-7.
  164. Sakhaee K, Maalouf NM. Metabolic syndrome and uric acid nephrolithiasis. Semin Nephrol. 2008;28(2):174-80.
  165. Xu LHR, Adams-Huet B, Poindexter JR, Maalouf NM, Moe OW, Sakhaee K. Temporal Changes in Kidney Stone Composition and in Risk Factors Predisposing to Stone Formation. J Urol. 2017;197(6):1465-71.
  166. Daudon M, Traxer O, Conort P, Lacour B, Jungers P. Type 2 diabetes increases the risk for uric acid stones. Journal of the American Society of Nephrology : JASN. 2006;17(7):2026-33.
  167. Tran TVMM, N.M. Uric Acid Stone Disease: Lessons from Recent Human Physiologic Studies. Curr Opin Nephrol Hypertens. 2020;29(4):in press.
  168. Maalouf NM, Sakhaee K, Parks JH, Coe FL, Adams-Huet B, Pak CY. Association of urinary pH with body weight in nephrolithiasis. Kidney Int. 2004;65(4):1422-5.
  169. Bobulescu IA, Park SK, Xu LHR, Blanco F, Poindexter J, Adams-Huet B, et al. Net Acid Excretion and Urinary Organic Anions in Idiopathic Uric Acid Nephrolithiasis. Clin J Am Soc Nephrol. 2019;14(3):411-20.
  170. Gridley CM, Sourial MW, Lehman A, Knudsen BE. Medical dissolution therapy for the treatment of uric acid nephrolithiasis. World J Urol. 2019.
  171. Canales BK, Sharma N, Yuzhakov SV, Bozorgmehri S, Otto BJ, Bird VG. Long-term Recurrence Rates in Uric Acid Stone Formers With or Without Medical Management. Urology. 2019;131:46-52.
  172. Maalouf NM, Poindexter JR, Adams-Huet B, Moe OW, Sakhaee K. Increased production and reduced urinary buffering of acid in uric acid stone formers is ameliorated by pioglitazone. Kidney Int. 2019;95(5):1262-8.
  173. Sakhaee K. Pathogenesis and medical management of cystinuria. Semin Nephrol. 1996;16(5):435-47.
  174. Andreassen KH, Pedersen KV, Osther SS, Jung HU, Lildal SK, Osther PJ. How should patients with cystine stone disease be evaluated and treated in the twenty-first century? Urolithiasis. 2016;44(1):65-76.
  175. Rodman JS, Blackburn P, Williams JJ, Brown A, Pospischil MA, Peterson CM. The effect of dietary protein on cystine excretion in patients with cystinuria. Clin Nephrol. 1984;22(6):273-8.
  176. Jaeger P, Portmann L, Saunders A, Rosenberg LE, Thier SO. Anticystinuric effects of glutamine and of dietary sodium restriction. N Engl J Med. 1986;315(18):1120-3.
  177. Fjellstedt E, Denneberg T, Jeppsson JO, Tiselius HG. A comparison of the effects of potassium citrate and sodium bicarbonate in the alkalinization of urine in homozygous cystinuria. Urol Res. 2001;29(5):295-302.
  178. Preminger GM, Assimos DG, Lingeman JE, Nakada SY, Pearle MS, Wolf JS, Jr. Chapter 1: AUA guideline on management of staghorn calculi: diagnosis and treatment recommendations. J Urol. 2005;173(6):1991-2000.
  179. Iqbal MW, Youssef RF, Neisius A, Kuntz N, Hanna J, Ferrandino MN, et al. Contemporary Management of Struvite Stones Using Combined Endourologic and Medical Treatment: Predictors of Unfavorable Clinical Outcome. J Endourol. 2016;30(7):771-7.
  180. Silverman DE, Stamey TA. Management of infection stones: the Stanford experience. Medicine (Baltimore). 1983;62(1):44-51.

Performance Enhancing Hormone Doping in Sport

ABSTRACT

 

Sport is the organized playing of competitive games according to rules. Hence doping represent drug cheating, a fraud on competitors, the sport, and the public. The charter of the World Anti-doping Agency (WADA) forms a harmonized Code that authorizes an annually updated list of prohibited doping substances and methods as well as accrediting national anti-doping labs around the world. Sports performance has 4 major components: skill, strength, endurance, and recovery, with each sport employing a distinct combination of these elements. These performance characteristics also correspond to the most potent and effective forms of doping. Sports requiring explosive power are most susceptible to androgen doping through their effect on increasing muscle mass and strength whereas sports that require endurance are most enhanced by hemoglobin (blood) doping which increases oxygen delivering capacity to exercising tissues. Performance in contact sports and those involving intense physical activity or training may also be enhanced by growth hormone and its secretagogues through speeding of tissue recovery from injury. Hormones remain the most potent and widely detected doping agents being responsible for about 2/3 of anti-doping rule violations detected by increasingly sophisticated detection methods. At present, the vast majority of positives are still due to a wide variety of androgens, including marketed and illicit nutraceutical, designer, specific androgen receptor modulator (SARM)) synthetic androgens as well as exogenous natural androgens, while the peptide hormones (erythropoiesis stimulating agents, growth hormone and its secretagogues) and autologous blood transfusion remain difficult to detect.

 

INTRODUCTION

 

Across the world, sport is a ubiquitous human social activity that forms an unique intersection of health, recreation, entertainment, and industry (1). It is both a major economic activity as well as a profound influence on social behavior of individuals at home, work, and play. One practical and concise definition of sport is the organized playing of competitive games according to rules. In that context, rule breaking is cheating to achieve an unfair competitive advantage whether it involves using illegal equipment, match fixing, banned drugs, or any other prohibited means.

 

The illicit use of banned drugs (doping) to influence the outcome of a sporting contest, constitutes a fraud against competitors, spectators, sport, sponsors, and the public no different from other personal, professional, or commercial frauds. While performance enhancement is almost invariably the intent of cheating, impairing performance is also well known in horse racing and even, rarely, in human elite sports (e.g. drink-spiking of banned drugs, injurious physical assaults). Rules of sporting contest may change by agreement, but once set, represents the boundaries of fair competition. Nevertheless, fairness is an elastic, socially constructed concept which may change gradually over time. For example, a century ago deliberate training itself was considered an ungentlemanly breach of fairness as competition was then envisaged as a contest based solely on natural endowments. Similarly, some sports once maintained a distinction between amateurs and professionals. The philosophical foundations of the concept of fairness is a deep and complex issue (2,3) where the focus has been mainly on distributive justice with an implicit goal of equality of outcomes. Less attention has been given to the philosophical basis of fair competition in sport where the prior distribution of talent and training and the outcome of contest are intended to provide equality of opportunity, but not of outcome, between contestants.

 

Naïve arguments are made that deny doping is cheating, or unsafe or violates the spirit of sport and asserting that drugs should be freely available or under medical supervision (4-6). However, removing prohibition on doping would immediately render drug taking as pervasive as training in elite sport extending to promising underage and sub-elite athletes. Ensuing demands on doctors to prescribe excessive, often massive, drug doses without medical indications would be unprofessional, unethical, and unsafe. This could convert sporting participation into a potentially dangerous rather than a healthful activity. In practice, creating enforceable boundaries for drugs in sport is unavoidable whether it is prohibition or, even under the most idealistic libertarian scenarios, by age or dosage. Within the limitations of unverifiable self-report regarding an illicit activity (7,8), surveys indicate athletes support antidoping testing mainly to prevent cheating but also to promote safety (9-11). Motivating factors for, and routes by which athletes get involved in doping are complex but include the use of non-banned nutritional supplements as a gateway to doping (12) and the suspicion of athletes or their entourage that their rivals may be using illicit drugs, the so-called “false consensus belief” (7,13-15). These philosophical issues are not considered further here and, recognizing that sport requires concrete, practical decisions, the establishment and enforcement of agreed rules is the basis of fair competition. An excellent discussion of the logic and morality of a decisive antidoping approach from an ethicist with extensive experience in sports anti-doping is recommended (16).

 

It is well understood that individual human genetic endowments are unequal and, among these, sporting prowess is at least partly genetically determined (17). However, little is still known of the genotype-phenotype correlations that underlie beneficial genetic endowments for sports performance. Natural genetic advantages are recognized in height (tallness for basketball, shortness for jockeys and motor-cycle riders) and hereditary erythrocytosis where a high circulating hemoglobin due to a high affinity erythropoietin (EPO) receptor (18)) for endurance sports, or conversely genetic disadvantage such as the common α-actinin-3 deletion genetic polymorphism which limits anaerobic, explosive power (19). More examples of genetic (dis)advantages for sports performance are likely to be identified as genomics continues to expand our understanding of the biological basis of health, including natural human sporting prowess. In the context of sports doping, however, a person’s genetic endowment is a given creating a natural boundary whereby the use of exogenous drugs or chemicals (including DNA) may constitute drug cheating or doping.

 

WORLD ANTI-DOPING AGENCY (WADA) AND THE GLOBAL ANTI-DOPING REGULATORY ENVIRONMENT

 

Cheating is as old as sport itself, yet the present endemic of doping using pharmaceutical drugs to boost sports performance is largely a Cold War legacy. Eastern European national doping programs were established by governments aiming to achieve a short-cut propaganda victory over their Western rivals, a challenge quickly reciprocated and then taken up by individual coaches and athletes. Starting with power sports (20), the epidemic became entrenched as an endemic in sufficiently affluent circles. In 1967, following the introduction of anti-doping rules by some sports federations, the International Olympic Committee (IOC) established its Medical Commission, which published their first list of prohibited substances. During the 1970’s the IOC Medical Commission took an increasingly active role by banning androgens which required developing standardized, valid methods to detect and deter androgen doping. After discarding alternatives such as immunoassays and blood sampling, in the 1980s mass spectrometry (MS)-based tests became (21) and remain the standard for detecting synthetic androgens in urine.

 

In 1999, the IOC established the WADA based in Montreal to be equally supported by governments and sporting organizations with its charter, the WADA Code, representing a harmonized set of global anti-doping rules introduced in 2004, revised in 2009 and 2015 and will be again revised in 2021(22). WADA also publishes an annually updated Prohibited List of Substances and Methods, accredits national anti-doping labs together with their operational anti-doping testing framework, and established the Court for Arbitration in Sport (CAS) to settle anti-doping legal disputes as sport’s “Supreme Court”. The WADA Code has been adopted by over 660 sporting organizations including all Olympic and Paralympic organizations and National Anti-Doping Organizations as well as most non-Olympic International and National Sports Federations. The WADA Code prohibits substances or methods which meet 2 of 3 criteria comprising:

(i) enhance performance (cheating),

(ii) harmful to health (safety) or

(iii) violate the spirit of sport (unsporting).

 

Although the primacy of penalizing cheating is widely understood, implementing these criteria encounter ethical and practical difficulties in proving ergogenic effects of increasing numbers of illicit and/or non-approved substances. These substances have unknown safety so that human testing is not feasible making athlete safety an important consideration. Crucially, the Code imposes strict liability on individual athletes so that a positive anti-doping test (including refusal or avoidance of testing or possession, attempts, trading and tampering with banned drugs) constitutes an anti-doping rule violation (ADRV), regardless of intent or negligence. Sanctions involve suspension from any elite competitive sport and extend to support personnel and teams. Suspensions, once 2 years are now 4 years since adoption of the 2015 Code. This is generally believed to be longer than the ergogenic benefits of doping, although recent evidence suggests that androgen effects on muscle may create durable or even permanent effects (23) which might argue for much longer or permanent banning of androgen doping violators.

 

The Prohibited List bans, at any time either in or out of competition, the use of performance enhancing hormones, including androgens, EPO and growth hormone and related substances or drugs which stimulate endogenous production of these hormones (Table 1). Among the 15 categories of prohibited substances (12) and methods (3), hormones feature prominently in S1 (anabolic agents, mainly androgens), S2 (peptide hormones, growth factors, related substances and mimetics), S4 (hormone and metabolic modulators), and S9 (glucocorticoids) with S1 and S2 having important “catch-all” provision for unnamed but related substances “with similar chemical structure or biological effects”. In addition, the S0 category bans non-approved substances, those without current regulatory approval for human therapeutic use. The prominence of hormones is reinforced by the WADA laboratory statistics for anti-doping tests where hormones remain the most frequently detected banned drugs (Table 2). In 2017, of over 322,000 anti-doping tests ~1.5% were positive with 61% due to hormones, the vast majority (~99%) due to androgens. These findings confirm that the detection of androgen doping is effective whereas the low rate of detection of hemoglobin or growth hormone doping may reflect the limitations of available tests for peptides and peptide hormones which require blood rather than conventional urine sampling and feature low sensitivity and brief windows of detection, rather than their lack of abuse. Further use of out-of-competition testing and blood samples together with more sensitive detection tests with longer windows of detection are required particularly for peptide hormones.

 

Table 2. Performance Enhancing Hormone Tests in WADA Labs

 

2005

2009

2013

2017

ACCREDITED LABS

33

35

33

31

TOTAL TESTS

183,337

277,928

269,878

322,050

POSITIVES

(% OF TOTAL)

3,909

(2.1%)

5,610

(2.0%)

5962

(2.2%)

4756

(1.5%)

HORMONES

(% OF +VE)

55%

73%

57%

61%

ANDROGENS

3893

5541

3352

1813

BLOOD/EPO

16

68

63

85

GH/PEPTIDES

0

1

0

19

Source: WADA website report on laboratory testing figures. See http://www.wada-ama.org/en/Science-Medicine/Anti-Doping-Laboratories/Laboratory-Testing-Figures/

 

The prevalence of doping in elite sports as an illicit activity with drastic consequences for athletes admitting guilt remains difficult to quantify using laboratory-based testing, inference from performance, or self-report questionnaires (24). The most promising methods appear to be questionnaires using the unrelated question random response methodology (25). This methodology was developed to estimate the prevalence of sensitive, disapproved, or illicit activities by asking the sensitive personal questions masked by mixing them with unrelated non-sensitive questions in an anonymized framework. This provides overall prevalence self-report estimates of the target activities without allowing for individual identification of answers. One study of two elite athletic competitions provided estimates of 43.6% and 57.1% for recent (last year) and 70.1% of ever use of banned doping methods (26). However, another study using the same methodology found markedly lower prevalence estimates of 0.7% to 11.9% for recent use of banned doping (27). The discrepancies between these prevalence estimates requires further clarification. A significant limitation of these methods is their reliance on athlete’s perception of banned methods. For example, whether “banned drugs” are interpreted as including the widely used (but non-banned) nutritional supplements which athletes are urged to avoid for fear of adulteration with unlabeled banned substances.

 

Therapeutic Use Exemption (TUE)

 

In rare cases, an elite athlete with a genuine medical need for therapeutic use of a prohibited drug may be granted a TUE (28). This exempts the athlete from the Code’s strict liability provision and permits them to compete during ongoing necessary medical treatment. WADA provides medical guidelines that standardize the evaluation and management of TUE applications for a range of medical illnesses. A TUE is granted by a national anti-doping organization based on an independent, expert review of valid, documented diagnosis, appropriate clinical indications and dose for hormonal treatment with a view to facilitating essential medical treatment but avoiding unjustified use or over-dosage. After stringent review TUE’s may be granted for treatment with testosterone, glucocorticoids, and insulin but there are very rarely any valid medical indications for EPO or, in adults, for growth hormone or IGF-1 in elite athletes. For example, TUE’s are usually justified for young male athletes with genuine androgen deficiency, occurring in ~1:200 men (29),  due to organic pituitary-testicular disorders with an established pathological basis (e.g. bilateral orchidectomy, severe mumps orchitis, Klinefelter’s syndrome, etc.) who require life-long testosterone replacement therapy (30). The TUE will approve, subject to regular review, a standard testosterone replacement regimen, including dosage and monitoring, with changes to regimen requiring approval. TUEs are not granted for men with functional decreases in blood T due to non-reproductive disorders including stress (“over-training”) or ageing (“andropause”, “LowT” “late-onset hypogonadism”), or for women.

 

In principle, detection of prohibited substances is ideally aimed at identifying a xenobiotic substance or its distinctive chemical signature(s) which do not occur naturally in the body, thereby distinguishing it categorically from normal body constituents. Such identification of a non-natural substance that can’t be of endogenous origin is congruent with the strict liability onus in proving an anti-doping rule violation (ADRV). Proving an ADRV is more difficult to achieve with administration of natural hormones or their analogs which must be distinguished from their endogenous counterparts. In this situation, the alternative requires developing valid biomarkers to prove the use of banned substances through their distinctive effects on the body and tissues. It is a formidable challenge to validate an indirect biomarker as proof of an ADRV capable of withstanding vigorous medico-legal challenge when a proven ADRV would prevent an athlete from pursuing their profession. Proof of an ADRV based on a doping detection test requires rigorous standardization and harmonization of every stage of the anti-doping tests from sample collection, chain-of-custody, storage, and analysis including accounting for any fixed (genetic, gender, age, ethnicity) or variable (exercise, hydration, masking vulnerabilities) factors which may impact on proposed test metrics.

 

COMPONENTS OF SPORTS PERFORMANCE AND DOPING

 

Sports performance has 4 major dimensions – skill, strength, endurance and recovery (Figure 1). High performance in any sport requires a characteristic blend of these dimensions although individual sports differ widely in that balance. Similarly, the major ergogenic drug classes have distinctive effects aligned predominantly along one of these dimensions so that the most effective ergogenic drug classes used in doping are dictated by these dimensions of sports performance (Figure 2). While every sport requires an acquired skill, some are largely or solely based on skill and concentration (e.g. board games, target shooting, car driving, and motor-cycle riding) and may benefit from drugs that reduce anxiety, tremor, inattention or fatigue. Sports that are highly dependent on explosive, short-term anaerobic power (sprinting, throwing, boxing, wrestling), typically ones which favor a stocky, muscular build, are most susceptible to androgen-induced increases in muscle mass and strength. Other sports with an emphasis on aerobic effort and endurance (e.g. long distance or duration events), characteristically favored by a lean build, may be boosted by hemoglobin doping (blood transfusion, erythropoietin (EPO) and its analogs or mimetics. Finally, sports that depend on recovery from major injury or recurrent minor injury during intensive training, notably contact sports, may benefit from tissue proliferative and remodeling effects of growth hormone and various growth factors.

Figure 1. Components of Sports Performance

Figure 2. Drugs that Enhance Sports Performance

ANDROGENS

 

Although the ergogenic effects of androgens were discovered empirically soon after the identification of testosterone as the principal male androgen of testicular origin in 1935 (31), their applications to elite sport performance were mainly developed during the Cold War by trial and error experiments undertaken on unknowing elite athletes (20,32,33); however, the scientific basis of androgen doping was only objectively proven in the 1990’s. Until that time, the settled consensus was that exogenous androgens had no effect in eugonadal men whose androgen receptors were already saturated by endogenous testosterone (T) (20,34,35). The then alleged benefits of androgen doping were misattributed placebo responses together with training and nutritional effects. Using an exemplary placebo-controlled, randomized clinical trial design with a wide range of testosterone doses, Bhasin et al showed that T increased muscle mass and strength in eugonadal young men to a similar extent as exercise alone and with additive effects when combined with exercise (36) (figure 3). Subsequent dose-response studies showed that administration of T increased muscle mass and strength by 10% without and 20-37% with exercise (where exercise alone increased them by 10-20%) together with additive effects from 3% increase in circulating hemoglobin. These benefits extended from below to well above physiological T doses or blood levels without evidence of plateau (37,38) and regardless of age (39).

Figure 3. Biological Basis of Androgen Doping

Androgen doping may be either direct or indirect (Table 3, figure 4). Direct androgen doping involves administration of testosterone, natural or synthetic androgens whereas indirect androgen doping includes a variety of non-androgenic drugs which increase endogenous T. Direct androgen doping originally involved all pharmaceutically marketed natural (T, DHT, nandrolone) and synthetic androgens but has extended to non-marketed designer and nutraceutical androgens as well as pro-androgens (androstenedione, DHEA) and the new class of non-steroidal androgens (selective androgen receptor modulators, SARM (40)). Indirect androgen doping involves use of hCG, LH, anti-estrogens (estrogen receptor blockers, aromatase inhibitors), opioid antagonists, and neurotransmitters involved in neuroendocrine regulation of endogenous LH and T secretion (41-44).

Table 3– Direct and Indirect Androgen Doping and Detection Methods

 

Substance

Detection method

Direct

 

Synthetic androgens

L/GC-MS

Natural androgens

L/GC-MS, T/E, CIRMS

Designer & nutraceutical androgens

L/GC-MS (bioassay)

Indirect

 

hCG (urinary or recombinant)

hCG immunoassay or LC-MS

hLH (recombinant)

hLH immunoassay or LC-MS

Anti-estrogens

L/GC-MS

GnRH analogs

L/C-MS

Opioid antagonists & neurotransmitters

L/C-MS

 

Figure 4. Direct and Indirect Androgen Doping

Detection of direct androgen doping using steroids of known chemical structure is highly effective using gas or liquid chromatography MS (45-47). Traces of synthetic androgens or their metabolites may remain detectable for periods up to months after last administration (48). Recent developments including the identification of long-term metabolites has further widened the detection windows for synthetic androgens (49-55). Challenges to detection of synthetic androgens have included the development of non-marketed designer and nutraceutical androgens, the use of natural androgens, and pro-androgens, masking methods, restricting use to out-of-competition training or micro-dosing. Designer and nutraceutical androgens are typically non-marketed synthetic androgens based on structures and synthesis methods recovered from expired patent literature of the 1960-70’s. These are now synthesized by unregulated chemical manufacturers without Good Manufacturing Practice (GMP) licensing advertising and sold over the internet or over-the-counter as nutritional supplements, which may contain undeclared steroids (56). However, once the chemical structures of any synthetic androgens are known, they are easily detectable although the sheer profusion of such chemicals represents an ongoing challenge. Nevertheless, despite their novelty, there is little evidence designer androgens have been used after they are discovered so that there is a high likelihood of detection. As a result, virtually all ongoing androgen ADRVs are still due to conventional marketed synthetic androgens.

 

Distinguishing Between the Exogenous and Endogenous Steroids

 

Administration of natural androgens (T or DHT) or pro-androgens (androstenedione, DHEA), raises the problem of distinguishing between the exogenous and endogenous steroids. Exogenous T administration can be detected by the urine T/E ratio, the ratio in urine of T to its 17α-epimer epitestosterone (E), operating as a sensitive screening test. In males, both T and E are co-secreted by Leydig cells and excreted in urine consistently so that the urine T/E is usually stable for any individual over time, being typically around 1. Administration of exogenous T, which is not converted to E, increases the urine T/E ratio and, when it exceeds a specified threshold, is evidence for administration of exogenous T. The urine T/E ratio thresholds were originally population-based, set initially at 6 and then subsequently lowered to 4. However, the urine T/E ratio is not an effective screening test for testosterone doping in females (57)because, unlike males whose circulating testosterone originates from as single source subject to strong negative hypothalamic feedback, circulating testosterone in females originates from three steroidogenic sources (adrenal, ovary, extraglandular conversion) none of which are subject to strong negative feedback by testosterone. Furthermore, the possibility of false negatives and false positives of population-based thresholds are limitations which may require further analysis to confirm or refute T doping in individual cases. These considerations have led to establishment of the steroid module of the Athletes Biological Passport (ABP), a compendium of serial observation of any individual’s tests which creates adaptive individual-specific T/E ratio threshold (58). This substitution of an individual’s own person-specific, in place of the population-based, thresholds allows for more sensitive and accurate detection of individual deviations in urine T/E ratio as evidence of T doping.

 

One limitation of the urine T/E ratio is a genetic polymorphism of the uridine 5'-diphospho-glucuronosyltransferase(UGT) 2B17 gene which encodes a phase II hepatic enzyme that glucuronidates T rendering it more hydrophilic to facilitate urinary excretion. This polymorphism comprises a genetic deletion which, in homozygotes, produces a non-functional enzyme that reduces urinary T (but not E) excretion to near zero producing an extremely low T/E ratio (<0.1). While this genetic polymorphism has no apparent biological effect on T action, it is unevenly distributed geographically being much more frequent in South East Asian populations (59). This biological false negative means that administration of exogenous T will be greatly reduced and may not exceed the usual population-based T/E ratio thresholds (60). On the other hand, it will exceed any individual’s own specific urine T/E ratio threshold so that genotyping and/or Bayesian profiling of serial T/E ratio in an ABP program provide complementary evidence (61-63).

 

Administration of exogenous T may also be identified by carbon isotope ratio MS (CIRMS) that can distinguish endogenous from exogenous T according to the C13/C12 ratio of urinary T (64,65). Commercially, steroids are manufactured from plant sterols produced by photosynthesis that exhibit distinctly lower C13/C12 ratio (typically, -26‰ to -36‰ relative to the global standard) compared with mammalian T biosynthesis (between -16‰ to -26‰) (66,67). Hence, a significantly lowered (“depleted”) C13/C12 ratio of urinary T, exceeding 3‰ relative to endogenous reference steroids, indicates that urinary T originates at least partly from exogenous chemical manufacture from plant sterols. CIRMS can also be applied to detect administration of other natural androgens or pro-androgens including DHT and DHEA (68), androstenedione, or even attempted masking by administering E (to lower urine T/E ratio) (66). A few T products (<5% (69)) have recently emerged with a lower, more mammalian-like C13/C12 ratio for urine T (70) creating a challenge for CIRMS detection. Nevertheless, extended isotope profiling of other steroid precursors and metabolites provides additional complementary reference biomarkers (71). A longitudinal application of CIRMS along the lines of another module of the ABP has been proposed (72). Furthermore, development of hydrogen ion ratio mass spectrometry has further enhanced the ability to distinguish between endogenous and exogenous steroids even when the carbon isotope ratio is non-informative (73-75). Suppression of urine (or serum) LH excretion may also provide corroborative evidence for the use of exogenous T or other synthetic androgens (63,76-78).

 

While MS is highly effective for detecting specific androgens, it requires knowledge of the chemical structure to be detected and otherwise cannot be applied. This principle applies to never-marketed designer or nutraceutical androgens sold over the internet or in unregulated over-the-counter nutritional supplements with unlabeled steroid content. A potential solution is the modern in vitro androgen bioassay that incorporates the human androgen receptor together with a convenient transactivation chemical read-out signal into a host yeast or mammalian cell (79). This has the generic capacity to detect all bioactive androgens regardless of structure due to their direct activation of the androgen receptor. Constructed in vitro androgen bioassays feature a sensitive dose-response signal proportional to the potency of the bioactive androgen (80-83). Yeast host cells have high specificity for detecting androgens but are less sensitive than mammalian cells, which express native steroid mechanisms including steroidogenic enzymes and/or other steroid receptors. Mammalian in vitro androgen bioassays can also detect pro-androgens, steroids lacking intrinsic androgenic bioactivity but which are converted into androgens within the mammalian cell. Hence, while mammalian host cells sacrifice specificity for higher sensitivity, they can also detect pro-androgens (79). Hence yeast and mammalian in vitro androgen bioassays are complementary in detecting both androgens and pro-androgens. The limitations of in vitro androgen bioassays are their susceptibility to matrix effects and difficulties in standardizing bioassay-based test. Consequently. they are best deployed to characterize products and substances for androgens or pro-androgen content rather than to detect androgens in complex biological samples. Hence the yeast androgen bioassay was decisive in the first conviction for use of a designer androgen by proving that tetrahydrogestrinone (THG) was a potent androgen (84) and has also been used to screen synthetic progestins to show that, unlike the original androgen-derived progestins, the modern generation of progestins are not androgenic (85).

 

Additional underutilized options to detect androgen doping is the use of alternative biological matrices such as hair, skin or nails as well as saliva and exhaled breath (86). Hair has the advantages of minimally invasive sampling with simple, convenient storage and the potential for very long window of detection, according hair growth rates (87). MS-based methods have been reported to detect exogenous (88-102) and endogenous (91,103-106) androgens in human hair following long-term, but not single dose (107), exposure. However, hair analysis tests have yet to undergo the rigorous standardization and validation required to become acceptable anti-doping tests in their medicolegal context. Problems that remain to be fully overcome include matrix effects, low recovery and limited sensitivity as well as the impact of age, hair color, alopecia, and shaving or passive chemical (cosmetic) contamination of hair.  Additionally nails and skin could also provide analogous information on past androgen exposure with relatively long windows of detection but suitably rigorous tests are yet to be convincingly developed (108). Saliva sampling has also been considered (109,110) for anti-doping application analogous to the use of salivary cortisol measurement for diagnosis of hypercortisolism (111). While potentially applicable to xenobiotic drugs, salivary testosterone immunoassay is not sufficiently accurate (112) and is not suitable to detect testosterone doping because even microscopic blood contamination (e.g. gingivitis, chewing hard food, tooth brushing) produces anomalous high readings. The existence of these renders salivary testosterone testing for antidoping purposes as unreliable by providing opportunity for claims of false positive for any adverse findings. Exhaled breath testing has also been investigated for certain small molecular weight chemicals (113). In theory, androgen-induced gene expression in circulating leukocytes might provide an additional biomarker of androgen action if specific and reproducible signatures can be defined (114); however, as direct detection of androgens is feasible and preferable for proving an ADRV, a role for gene expression biomarkers of androgen action remains to be established for anti-doping.

 

Indirect Androgen Doping

 

This doping strategy aims to increase endogenous T production and thereby evades detection by routine screening tests for exogenous T such as urine T/E ratio or CIRMS. Urine hCG is detected by commercial hCG immunoassays using immunoassays specific for intact heterodimeric hCG (including its nicked variant) which, if positive by exceeding a detection threshold (>5 IU/L), requires confirmation by a second immunoassay for intact heterodimeric hCG which is required to prove hCG use. A highly sensitive LC-MS method to detect urine hCG (115) is more specific than immunoassays (116) and has a lower threshold for a positive result in male athletes (117). A key issue is to distinguish a positive hCG urine test, presumptively indicating hCG doping, from early trophoblastic tumor or immunoassay artefacts. As hCG doping is not effective in women and urine hCG screening can detect early pregnancy, an unwarranted privacy intrusion, hCG testing is restricted to male athletes (43). Although direct LH doping is an implausible doping threat (118), suppressed (63,76-78,118) or elevated urine LH may be useful for confirming direct or indirect androgen doping (42,43,76,119). Anti-estrogens (estrogen receptor antagonists) or aromatase inhibitors, which can cause reflex increases in serum and urine LH and testosterone (42), are detected by MS-based chemical detection methods.

 

Overall, detection of direct androgen doping is now so effective that in WADA-compliant elite competitions it is restricted to the ill-informed, often using counterfeit or unlabeled products (120). Yet the potency of androgen doping in power sports continues to prompt development of novel androgen doping strategies. These will include use of undocumented synthetic androgens, novel indirect androgen doping methods and micro-dosing of natural androgens during out of competition training. The retreat to using micro-dosing inherently reduces the dose-dependent ergogenic benefits of doping while maintaining the risk of detection and disqualification. There remains a need to maintain deterrence by effective detection methods for evolving new androgen doping threats.

 

HEMOGLOBIN (BLOOD) DOPING

 

Hemoglobin doping involves either direct blood transfusion or indirect methods of increasing hemoglobin via stimulating erythropoiesis by administration of erythropoietin, its analogs or mimetics (see excellent reviews (121,122)) (Table 4). Boosting hemoglobin is advantageous in aerobic, endurance sports such as road cycling, distance running and cross-country skiing. Maximal oxygen consumption (Vo2), a rate-limiting factor in aerobic exercise, principally determined by cardiac output and blood oxygen transfer with a lesser contribution from tissue oxygen transfer (123). Experiments on exercise tolerance and blood transfusion were first reported in 1945 (124,125)but the scientific basis of hemoglobin doping via enhanced tissue oxygen transfer was firmly established in 1972 by the work of Ekblom et al reporting experiments in healthy volunteers who underwent venesection and/or re-transfusion of 1, 2 or 3 units (400 mL) of blood with repeated testing of maximal exercise-induced oxygen consumption before and after each procedure (126). This proved unequivocally that the maximal oxygen consumption was highly correlated with acute changes in hemoglobin (figure 5). Subsequently, during the 1970-80’s before its banning in 1988, blood transfusion became a prevalent surreptitious practice in road cycling and cross-country skiing and the apparently low prevalence among distance runners may be an underestimate (127). Modelling of historical performance in European road cycling from 1993 onwards shows a unique progression averaging an improvement of 6.4% corresponding closely with the performance enhancement (6-7%) due to rhEPO administration, which is sustained for at least 4 weeks after administration (128-130).

Table 4. Direct and Indirect Hemoglobin Doping and Detection Tests

Doping Mechanism

Detection

Direct (Blood transfusion)

 

Heterologous

Flow cytometry: bimodal population of blood group antigens

Autologous

No direct detection tests.

Athletes Biological Passport Biomarkers:

Urine phthalate excretion

Total hemoglobin mass

Indirect (Erythropoiesis stimulation)

Direct

 

rhEpo & biosimilars (>100) Epo analogs

Urine double immunoblot, (LC-MS)

Indirect

 

Hypoxia altitude training, hypoxic sleep area

Not banned

Hypoxia-mimetics: hypoxia-inducible factor & stabilizers, iron chelation, cobalt, 2,3 DPG analogs

LC-MS/MS

Artificial O2 carriers: HbOC, perfluorocarbons

LC-MS/MS

 

Figure 5. Direct and Indirect Hemoglobin Doping

Blood Transfusion

 

Transfusion may involve either another person’s (homologous) or the athlete’s own (autologous) blood administered prior to a contest to acutely increase circulating hemoglobin. Homologous blood can be transfused at any convenient time to enhance performance in competition but when performed by untrained personal in non-clinical environments risks transfusion reaction, blood-borne infectious disease, and iron overload. By contrast, autologous transfusion reduces health risks but requires complex coordination as venesection itself is detrimental to performance, and it requires balancing recovery from blood withdrawal and loss of erythrocyte viability during long-term cryostorage with training and competition schedules. Although blood transfusion was first banned by the IOC in 1986, the first practical approach to banning blood doping was the introduction of hematocrit testing in 1997 by the international skiing and cycling federations. These regulations excluded athletes on health grounds from entering competition on the day if their hematocrit exceed a safety threshold (0.50). However, this encouraged hematocrit titration to just below threshold and only prevented competing until hematocrit returned under that threshold, which could be quickly accomplished by venesection. The first ADRV’s for blood manipulation involving hematocrit threshold and titration were in 2001.

 

Homologous blood transfusion creates a bimodal population of blood group antigens which is detectable by flow cytometry using a panel of 12 minor blood group antigens (131), from the wider array of blood group antigens (132), which can detect a <5% contamination of exogenous erythrocytes. Subsequent refinements simplified and improved test sensitivity so that a panel of 8 antigens can detect contamination comprising a minor admixture population of 0.3-2.0% with no false positives but high sensitivity (~80%), the latter depending on the magnitude of the minor contaminating mixture (133,134). Alternatives based on genotyping for the admixture population of leukocytes have also been proposed (135,136). As a test proving unequivocally the presence of non-endogenous erythrocytes in the circulation, this method is definitive if performed to the required standard. A remotely hypothetical defense against a positive test, based on stable marrow chimerism from a vanished twin, was raised by a cyclist who subsequently admitted transfusion (135). Based on risk of detection as well as to health risks, homologous transfusion has now largely disappeared in favor of autologous transfusion (137).

 

Autologous Transfusion

 

The biggest gap in current anti-doping tests is the lack of a specific test to detect autologous transfusion (138). Research to identify robust physico-chemical or biological markers for direct identification of a subpopulation of ex-vivo aged erythrocytes is underway using flow cytometry  (139) but the dilution and rapid clearance of effete erythrocytes make for challenging detection problems (140). In the interim, other indirect methods have been developed. These include measuring urinary excretion of phthalates, plasticizers that leach out from the polyvinylchloride blood packs used to store venesected blood (141). This test has brief window of detection (2 day) so will detect auto-transfusion during or immediately before events (characteristic in road cycling, according to convicted dopers) but may miss earlier auto-transfusion. Furthermore, the ubiquity of low-level environmental phthalate exposure requires establishing detection thresholds and non-plastic blood containers can be used. An alternative is the measurement of total hemoglobin mass (142), a measure with good stability and reproducibility even during exercise and circumvents influence of variations in plasma volume such as due to dehydration or dilutional masking (142,143). However, as this requires inhalation of carbon monoxide, which has transient detrimental effects on performance, it is not ideal for routine anti-doping use and its sensitivity may be insufficient to detect all EPO micro-dosing (144,145). Nevertheless, alternative methods for serial measurement of total hemoglobin mass remain attractive. Other hypothetical methods include the detection of microRNA (146) or immune reactions to transfusion (147) but the sensitivity and specificity of these proposed tests remains to be fully evaluated.

 

The best detection test for autologous hemoglobin doping at present is the hematological module of the ABP introduced in 2009 (148). Conceptually, it is a biomarker test which adopts a Bayesian approach of creating serially-adaptive, person-specific reference limits, based on using all prior testing, to supplant population-based thresholds. Combining all of an individual’s previously collected hematological data creates a probabilistic test of whether any new result deviates significantly from that individual’s personal reference limits (149). These person-specific thresholds allow for ongoing refinement and reinforcement by further testing. The thresholds are calculated by a variety of algorithms incorporating routine hematological parameters, notably hematocrit and reticulocyte counts. Those were developed over the last two decades to create the ABP hematological model which is sensitive to both direct and indirect hemoglobin doping (150). The first attempts to regulate hemoglobin doping in the late 1990’s sought to prevent road cyclists or cross-country skiing athletes competing on health risk grounds when their hematocrit exceeded pre-determined, population-based safety criteria (e.g. hematocrit 0.50 or hemoglobin 170 g/L for cycling). However, while this excluded extreme hemoglobin doping only until the short period when the safety threshold was no longer exceeded, it allowed an increase in an athlete’s natural hematocrit, typically averaging ~0.45, up to the permitted ceiling threshold which fostered titrated hemoglobin doping and manipulations like hemodilution by saline or plasma volume expander infusions to avoid detection (151). More sophisticated hematological algorithms were then developed to detect hemoglobin doping initially for the Sydney 2000 Olympics (152,153), the first generation of algorithms developing validated tests for ongoing and for recent cessation of hemoglobin doping, using a combination of biochemical variables related to erythropoiesis physiology. This approach was simplified by a second generation algorithm using only routine hematological parameters (hemoglobin, reticulocytes) (154), and was subsequently combined with the concept of a sequential development of individual-specific reference ranges (155)  into a third generation algorithms (156,157) which were refined for the ABP (148,149). The hematological module of the ABP currently employs an algorithm involving 8 parameters derived from routine hematological profile (hemoglobin, hematocrit, erythrocyte count, reticulocyte count and percentage, mean corpuscular volume, mean corpuscular hemoglobin, mean corpuscular hemoglobin concentration) (158). This is capable of detecting any form of hemoglobin doping, whether direct or indirect, with good but imperfect sensitivity (143-145) and using only routine hematological tests. The reported increasing use of very low EPO doses (“micro-dosing”) would markedly reduce the magnitude of any dose-dependent ergogenic benefits (145) while still carrying risks of detection, disqualification, and disgrace.

 

Stimulation of Erythropoiesis

 

Indirect methods to increase hemoglobin include administration of recombinant human EPO or its analogs as well as hypoxia-mimetic drugs (hypoxia-inducible factor stabilizers, iron chelation, cobalt, 2,3 diphosphoglycerate analogs) or artificial oxygen carriers (perfluorocarbons, hemoglobin-based oxygen carriers). Related but non-banned methods include altitude training or its simulation by sleeping in hypoxic rooms which are less effective than hemoglobin doping (129).

 

The identification of the human EPO gene in 1985 led to the marketing of recombinant human EPO (rhEPO) between 1987-9. Despite the IOC’s prohibition of EPO’s use in sports in 1990, the commercial availability of rhEPO created powerful new opportunities for indirect hemoglobin doping which were soon proven experimentally (159). A drug, which circulates for hours to days, but with potent and long-lasting ergogenic effects after its disappearance due to the 4-month lifespan of erythrocytes, is both attractive for doping and a challenge to anti-doping testing. Expiry of the rhEPO patent in 2004 allowed marketing of a profusion of generic EPO (“biosimilar”) products, estimated globally at over 80 (160), as well as modified EPO analogs (darbepoeitin, pegylated EPO, peginesatide, EPO fusion proteins). A fatal cluster involving deaths of 18 Dutch and Belgian road cyclists, presumably due inadvertent over-dosage during empirical attempts to maximize ergogenic effects of illicit rhEPO, was reported (161), although difficult to verify (162). A similar excess of unexpected deaths of road cyclists was also reported again in 2003-5 when novel EPO analogs and EPO biosimilars were marketed.

 

Detection of EPO in urine is difficult because of the prevailing low concentrations and need to distinguish exogenous recombinant from endogenous EPO. The first effective method for rhEPO in urine was a double immunoblot (163,164)which was capable of detecting urinary excretion of a variety of exogenous EPO products and analogs according to their differences in glycosylation side-chains, and differences in primary amino acid sequence where they exist, while distinguishing them from endogenous EPO. Although further refined (165) and extended to other EPO analogs (166), the immune-electrophoresis test is sensitive but relatively laborious and provides only a short window of detection of up to a week post-administration (167). More sensitive methods based on proteomics (for EPO analogs with differences in primary structure) together with glycomics (for biosimilars and analogs which have host-cell specific variations in side-chain glycosylation but unchanged natural EPO primary structure (168)) are possible but not yet approved. Additional applications to detect EPO and analogs using dried blood spots have been reported (169). Similarly, preliminary investigations have proposed a EPO-induced gene expression signatures as a biomarker to detect EPO administration but specificity relative to exercise and other physiological effects remain to be clarified (170).

 

Other EPO mimetics such as hypoxia mimetic drugs including hypoxia-inducible factor (HIF) stabilizers and related small molecules represent growing threats as potential indirect hemoglobin doping agents (171). These non-peptide chemicals interfere with various steps of the molecular oxygen sensing mechanism to mimic renal hypoxia and thereby induce EPO secretion resulting in increased blood hemoglobin. As a convenient orally active alternative to the lucrative pharmaceutical market for injectable erythropoiesis-stimulating peptides (~$7-8 billion (172)) to counteract anemias of chronic renal failure or marrow failure due to myeloproliferative disease or cytotoxic cancer therapy, they constitute a very active area of pre-clinical patent-based clinical drug development (171). Experience suggests that such innovator products can enter the doping black market before marketing approval (150,171). Despite the profusion of pre-clinical leads, they represent families of related chemical structures disclosed in patents for which LC and/or GC-MS detection tests should, in principle, be effective. Understanding the metabolism of these drugs when they come to market may identify long-lasting metabolites that can extend the windows of detection. Coupled with evidence from the ABP, manipulation of the EPO pathway may be detected in conjunction with corroborative measurement of inappropriately suppressed or elevated endogenous EPO for the prevailing hemoglobin level.

 

HIF is a key generic biological mechanism for tissue sensing of hypoxia and triggering local (neovascularization, angiogenesis) and systemic (EPO) defensive reactions. The promoter of the EPO gene contains enhancer and inhibitor regions with the hypoxia-responsive element which binds HIF and a GATA binding site which enhance and inhibit, respectively, EPO gene transcription. HIF is a heterodimer formed by constitutively expressed subunits with the β subunit in excess and availability of α subunit limiting formation of bioactive HIF. The 3 HIFα subunit isoforms are subject to hydroxylation of specific proline residues by prolyl hydroxylase enzymes which inactivate HIFα by ubiquitination, a tag which targets it to proteasomal degradation. HIFα subunit inactivation is strongly dependent on tissue oxygenation being active during normoxia but reduced during hypoxia when persistence of HIFα stabilizes the HIF heterodimer. Notably, during hypoxia the expression of HIFα in renal cortical cells stimulates EPO gene expression so that HIF stabilization by prolyl hydroxylase inhibitors leads to increased EPO secretion and circulating hemoglobin. Hence inhibiting prolyl hydroxylase activity via blocking its required cofactors (ascorbate, ketoglutarate, iron) using cobalt, nickel, iron chelation, ketoglutarate analogs or mechanism-based chemical inhibitors can result in increased hemoglobin via stimulation of EPO secretion (171). Similarly, small molecule GATA inhibitors potently stimulate circulating EPO, hemoglobin and performance in mice (173) although none have yet been marketed so their human efficacy and safety remain to be determined.

 

Another approach to increase oxygen delivery to muscle has been to exploit the ability of 2,3 diphophoglycerate (2,3 DPG), whose binding to hemoglobin reduces its affinity for oxygen with the left-shift of its oxygen dissociation curve as an oxygen unloading mechanism in tissues. 2,3 DPG analogs, developed as radiation sensitizers for hypoxic radio-resistant tumors, enhance tissue oxygen delivery in vivo (174,175) but would feature only short-term, acute effects readily detectable by mass spectrometry (176,177).

 

Adverse effects from use of rhEPO or its analogs are well known in medicine but poorly recognized in doping. They include immunogenicity (with risk of EPO autoantibody mediated pure red cell aplasia) (178,179), cardiovascular complications (including venous thromboembolism, stroke, hypertension and myocardial infarction) and premature death (180-183). In routine clinical use of EPO to correct renal anemia, the goal is a gradual increase to subnormal hemoglobin targets so that the excessive and/or rapid rises in hematocrit and blood viscosity (184) may explain the excess unexplained deaths among young European road cyclists in the late 1980s. In addition, use of rhEPO may deplete iron stores which limits hemoglobin synthesis so that athletes may also use oral or injectable iron supplements, which carry their own risks such as iv iron supplementation’s potentially adverse effects in enhanced tissue oxidative damage and excess mortality in chronic kidney disease (185). Although clinical safety experience with ESAs is restricted to patients with serious medical disorders, there is evidence from the general community that higher natural hematocrit is associated with worse long-term cardiovascular health outcomes (186-188).

 

GROWTH HORMONE

 

Growth hormone (GH) is a tissue growth promoter in children but after puberty it is predominantly a metabolic hormone although latent tissue growth promoting effects may be unleashed under non-physiological circumstances, such as during recovery from tissue injury. There is consistent anecdotal evidence that GH has been used in elite sports for decades (189). Nevertheless, ergogenic effects of GH remain unproven and largely speculative as discussed in excellent recent reviews (190-192). Claims of GH benefits in sport have included increases in muscle mass and strength, especially in conjunction with androgens, and/or improved tissue healing with more rapid recovery from either major injuries or minor repetitive injuries, such as from intense physical training allowing for more effective training. The biological basis of ergogenic effects of GH have been tested in these two different scenarios with largely inconclusive findings.

 

Evidence for direct enhancement of athletic performance by GH has been investigated in two well controlled RCTs with a primary focus on athletic performance. In one study, 96 recreational sub-elite athletes (63 male, 33 female, mean age 28 years) were administered 8 weeks of daily sc injections of GH or placebo with the men also having weekly im injections of T enanthate or saline placebo for the last 5 weeks (193). GH increased lean (muscle) mass (by +2.7 kg) and reduced fat mass (by -1.4 kg) while T increased lean mass (alone by +2.4 kg, by +5.8 kg with GH). The effects of GH were marginally significant for anaerobic sprint capacity (by +3.9%, p=0.05) when pooling male and female participants but this was due to significant effects in men only (by +5.5% alone and +8.3% with GH). However, there were no significant effects on maximal Vo2 consumption, dead lift, or jump height (193). A second study involved 30 healthy non-athletes (15 male, 15 female, mean age 25 years) who were administered daily sc injections of GH at high (4.6 mg/day) or low (2.3 mg/day) doses or placebo (194). There was no significant effect on muscle mass or maximal Vo2 consumption. Additional controlled studies of GH effects but with less focus on athletic performance have also shown that (a) a single dose of GH (~0.8 mg) in 9 recreational athletes did not affect maximal Vo2 or power output in repeated 30 min bursts of bicycle ergometry (195), (b) short term (6 days), low dose GH (~1.7 mg/day) treatment of 48 male androgen abusers withdrawn from androgens for 12 weeks significantly increased maximal Vo2more than placebo (196), (c) daily sc injections of a GH receptor antagonist (pegvisomant) or placebo for 16 days to 20 sedentary men did not change maximal Vo2 although time to exhaustion at 90% maximal Vo2 was reduced (197)and (d) 4 weeks of daily sc injections of GH (~5 mg/day) increased whole body protein synthesis (198), lipolysis and glucose uptake (199) with uncertain significance for athletic performance. Overall, these studies indicate that GH has, at most, a modest ergogenic effect in men only and through enhancing T effects. That is consistent with the fact that young women have markedly greater growth hormone secretion than young men so that growth hormone cannot explain the sex differences in athletic performance (200).

 

It is also claimed that GH may enhance injury healing, thereby facilitating more intensive training and/or recovery from muscle, connective tissue or bone injury, notably in contact sports. This claim is difficult to evaluate and no well controlled studies of recovery from sports injuries or tolerance of training intensity in elite athletes are reported. The most germane surrogate evidence available arises from investigations on the use of GH in recovery from injuries due to burns, fracture, or for wound healing. A recent Cochrane meta-analysis review of GH treatment effects on recovery from burns injury and healing of donor skin graft sites suggests that GH has a small benefit in skin healing with large burns and reduced hospital stay but there was no benefit in reducing mortality or scarring and adverse effects, notably hyperglycemia, were increased (201). In practice, the increased mortality due to administration of high dose GH in critical illness (202) has led to GH treatment not being widely adopted in clinical practice of treatment of burns. Similarly, the only well controlled study of GH effects on bone healing from fracture reported that, among over 400 patients with tibial fractures treated for up to 16 weeks with GH (1, 2 or 4 mg/day) or placebo, there was no benefit of GH for overall healing (203). Finally, while there are numerous experimental studies of GH or growth factors on wound healing in animal models a wide variety of findings are reported with detrimental, neutral, or beneficial effects but no well-controlled human studies are available. In summary, the available evidence for improved tissue repair or regeneration is minimal.

 

Important caveats on interpreting these few well designed studies are that the effects of higher GH and T doses, as used in doping, have not been studied so that more potent higher dose and/or interactive effects cannot be excluded in the absence of well controlled, high dose, placebo-controlled studies. Nevertheless, the hypothesis that high dose GH exposure would enhance muscular function is inconsistent with the experience of acromegaly in which patients experience much higher (25-100 times) growth hormone exposure than doses that can be ethically administered to healthy human volunteers (204), yet characteristically display muscular weakness rather than increased muscle size or strength (205). Anti-doping science history suggests that caution is required before rejecting evidence for claimed ergogenic effects without investigations replicating the pharmacological doses used.

 

Furthermore, safety analysis is not feasible based on the few, small, short-term studies of GH’s potential ergogenic effects; however, there are significant safety concerns about the long-term risk of cancer following GH administration. Even standard therapeutic GH doses administered to GH deficient children are associated with increased risk of second cancers in some (206-208) but not all (209) follow-up studies although these risks appear largely confined to survivors of childhood cancers and its treatment which render them GH deficient (210-213). Although the significant cancer risk based on uncontrolled observational cohort data using standard GH doses remains contentious (214,215), the long-term risks of much higher GH doses used illicitly by athletes must be viewed with significant concern.

 

Detection of GH doping remains difficult (216). A major challenge is the non-glycosylated primary structure of recombinant and endogenous 22 kDa GH, that lack the distinctive side-chain carbohydrate differences of exogenous glycoproteins EPO or hCG which provide a convenient basis for sensitive molecular detection tests. Nevertheless, minor infidelities in commercial manufacturing of GH may incorporate distinctive non-natural chemical features proving an exogenous origin (217-219) although these findings have not been developed into detection tests. Challenges to the detection of GH doping arise from the physiological pattern of endogenous GH secretion with its intermittent, pulsatile pattern subject to prominent influence of exercise, stress, and nutritional effects together with GH’s brief circulating half-life and low urine concentrations (220,221). Like other major doping classes, there are both direct and indirect forms of GH doping, involving either direct administration of GH or IGF-I or their analogs and indirect GH doping involving drugs that aim to increase endogenous GH and IGF-I secretion (Table 5).

 

Table 5. Growth Factors, Growth Hormone Related and Other Peptides

Growth Factors

Growth Hormone Related Peptides

Other Peptides

 

GHRH analogs

Ghrelin analogs

Other

 

FGFs

GHRH

Lenomorelin (ghrelin)

IGF-1 & analogs (MGF, long R3IGF-1)

Thymosinß4

HGF

CJC-1295

GHRP-1

IGF-2

 

MGF

CJC-1293

GHRP-2 (pralmorelin)

Insulin & analogs

 

PDGF

Sermorelin

GHRP-3

AOD-9604

 

VEGF

Tesamorelin

GHRP-4

hGH 176-191

 

 

 

GHRP-5

 

 

 

 

GHRP-6

 

 

 

 

Hexarelin

 

 

 

 

Ipamorelin

 

 

 

 

Alexamorelin

 

 

 

 

Anamorelin

 

 

 

 

Macimorelin

 

 

 

 

Tabimorelin

 

 

 

 

Examorelin

 

 

 

The first test to detect administration of exogenous GH, the 22kD recombinant form of human GH, was based on blood sampling to measure the ratio of circulating isoforms of GH recognizing the fact that the pituitary secretes not only the major 22 kD isoform (65-80%) but also a variety of minor isoforms including a wide variety of minor isoforms and their multimeric variants (222). Administration of exogenous GH suppresses endogenous pituitary GH secretion leading to a predominance of circulating 22 kD GH. This is the basis for the GH isoform ratio test whereby a serum sample is measured by two different GH immunoassays, one with predominant 22 kD GH specificity (“rec” assay) and the other recognizing the broad spectrum of pituitary GH isoforms (“pit” assay) and the ratio of results (“rec”/”pit” ratio) is an index to detect administration of exogenous recombinant GH (220,223). This ratio test then serves to detect administration of exogenous recombinant human 22kD GH analogous to detection of exogenous T by the urine T/E ratio and exogenous insulin by analysis of serum C peptide (224). The differential GH isoform ratio test has undergone extensive validation involving standardization of the two GH immunoassays with distinctive immunoreactivities to quantify 20kD and 22kD epitopes as well as its application to various populations of elite athletes and evaluating physiological factors which might impact on the validity of test read-out. A strength of this test is that it is aimed at the exogenous doping agent itself, although it cannot definitively distinguish it from its endogenous counterpart. The major limitations of this differential isotope ratio test are its narrow window of detection (24-36 hr post administration) and its inability to detect indirect GH doping. While pituitary-derived human GH might not be detected, human pituitary GH, once obtained from national scale pituitary collection and purification programs, has not been available since 1985 when its risks of Creutzfeldt-Jakob disease were identified (225,226) with recombinant human GH replacing pituitary-extracted GH worldwide. This differential isoform test was first introduced for the 2004 Olympics (227) and led in 2010 to the first successful detection of out of competition GH doping (228).

 

A complementary detection test with a wider window of detection has been developed based on biomarkers of GH action. This uses two serum biomarkers of tissue GH effects, circulating IGF-1 as a short-term marker of hepatic GH action, and N-terminal peptide of procollagen type III (PIII-NP) as a long-term marker of GH-dependent collagen synthesis. In a study of 102 recreational athletes (53 male, 49 female, mean age 25 years, from 4 different European cities) randomly assigned to self-inject 2.7 mg or 5.4 mg GH or placebo once daily, measurement of serum IGF-1 and PIII-NP by specific immunoassays were able to correctly classify 86% of samples from males and 60% of samples from female using an empirical linear discriminant analysis of log-transformed serum IGF-1 and PIII-NP at the specificity of 1:10,000 required for a WADA biomarker threshold (229). Subsequent studies have shown that additional collagen biomarkers, N-terminal propeptide and C-terminal telopeptide of type I collagen, further widen the window of detection for GH administration (230,231). This multiplex biomarker test, based on using standardized immunoassay antibodies, requires establishment of reliable reference range with specificity (false positive detection rate) of no more than 1:10,000 incorporating the impact of gender and age, although exercise, injury, ethnicity and sports type appear not to be confounding influences but is not yet in routine use by WADA anti-doping labs. The two GH doping test, the differential isoform and biomarker approaches, are considered ultimately complementary (232).

 

IGF and Insulin Doping

 

IGF-1 is a circulating marker of hepatic GH effects and mediator of GH action so the marketing in 2005 of recombinant human IGF-I alone, and later with its major binding protein recombinant human IGF binding protein 3 (IGF-BP3) (233), for treatment of diabetes, insulin or GH insensitivity or motor neuron disease, together with the availability of IGF-1 analogs for laboratory use, creates the possibility of IGF doping (234). Time-series analysis of elite sports performance (235) is consistent with the occurrence of IGF-1 doping but its prevalence is unknown (56). As the biological basis for ergogenic effects of IGFs is due to its GH-like effects, this remains largely speculative and accompanied by the same safety concerns. IGF-1, IGF-2 and their analogs (236) as well as insulin and its analogs (237) are all readily detectable by LC-tandem MS and preliminary evidence suggests that biomarkers for IGF-1 administration (IGF-2, IGFBP2) may widen the window of detection (238). However, a specific test to detect IGF doping remains to be established (239).

 

MGF is a splice variant of IGF-I which, although not known to appear in the circulation, have any pharmacological effects, or be approved for human use (240), is advertised on the black-market and internet (241) for alleged anabolic or tissue repair/regeneration benefits. Like other short peptide with known structure, it is readily detectable using LC- tandem MS (241).

 

Insulin has long been used in doping and was prohibited in sports since 1999 (242,243). Other than its proper medical use in diabetics, the use of insulin and its analogs for doping is based solely on its easy availability coupled with anecdotal information from other drug users. There are no clinical studies showing any ergogenic effects of insulin or its analogs in non-diabetic individuals. The doping folklore appears to arise from the classification of insulin as being “anabolic”, in a loose generalization and mistaken analogy to androgens. In healthy non-diabetic individuals, insulin and its synthetic analogs stimulate weight gain via hypoglycemia and increasing appetite, but produce fat rather than muscle gain. The adverse effects include hypoglycemia, hypokalemia, injection-related infections and weight (fat) gain. Doping detection tests for insulin and its analogs continue to evolve and focus on highly sensitive and specific quantitative MS-based proteomics (244).

 

Growth Factors, GH Releasing and Other Peptides

 

For the unscrupulous in pursuit of the unlawful, the increasingly stringent detection of androgen and hemoglobin doping, the two most potent classes of ergogenic drugs, has led to new, highly speculative form of doping involving peptide growth factors and GH releasing peptides. These are within the size range of automated bulk custom peptide synthesis and are marketed cheaply by chemical manufacturers. While notionally sold solely for laboratory research, these unregulated products are available for purchase over the internet. Promoted by speculative fantasies on their mode of action coupled with testimonials to their efficacy but without objective testing or assurance of safety in humans, they are believed to be widely used by gullible and/or desperate athletes and their trainers. As unregistered drugs, this growing range of peptides appears to constitute a greater threat to athlete’s health than a risk of effective cheating.

 

The S2 category of Prohibited Substances lists, in addition to GH and IGF-1, GH fragments and releasing peptides, a wide array of growth factors and modulators and, crucially, a generic catch-all provision for unnamed growth factors and peptides which may affect connective, vascular, muscular, or regenerative tissues or energy utilization and other substances with similar chemical structure or biological effects.

 

The major category of oligopeptides used for doping is the class of GH releasing peptides analogs of the endogenous GH releasing peptides, GHRH and ghrelin, whereby their analogs aim to increase endogenous GH secretion and are therefore banned (table 5). Most of these peptide were developed in the pharmaceutical industry from the 1990s aiming to provide cheaper, orally active, non-peptide agonists with capacity for sustained stimulation of endogenous GH secretion to “rejuvenate the GH/IGF-1 axis” (245), an unusually explicit acknowledgment of the regular nexus between hormonal rejuvenation and doping (246). However, none of these hormonal peptides have been registered for human therapeutic use with only one (pralmorelin) registered for single-dose, diagnostic use (for GH deficiency) in Japan and unacetylated cyclin ghrelin marketed in Europe for Prader-Willi syndrome. Although they may stimulate GH release initially, many failed to achieve sustained GH release due to desensitization and none achieved meaningful clinical improvements in any target diseases. If their unproven ergogenic benefits are due to sustained GH release this renders them unlikely to be beneficial; nevertheless, the caveat on not accepting negative conclusions without direct testing are also relevant to this class of peptides. Like other short peptides, once chemical structures are known, detection is readily feasible using LC-MS (247,248). The illicit nature of this market raises the risks of counterfeit and unsafe products with attendant risks of infection and residual toxic contaminants unlike the purity pharmaceutical product manufacturers are required to demonstrate by batch release testing.

 

PROGRESS, GAPS, AND FUTURE PROSPECTS

 

Anti-doping science continues to make major progress over recent decades especially since the advent of WADA with its harmonization and focus on deterrence through standardized testing. Progress from improved MS-based testing methodologies and instrumentation, summarized annually by the editor (M Thevis) of the major antidoping science journal, Drug Testing and Analysis (249), is evident from the increasing numbers of ADRV findings among frozen stored urine samples now banked for 10 years from previous Olympics. Like any efforts to combat human malfeasance, the quest for drug-free and safe sport requires ongoing vigilance and continual renewal of intelligence-based detection testing. While great progress has been made in the two canonical forms of doping, androgen and hemoglobin doping, human ingenuity continually finds way to challenge the testing just as traditional frauds are supplanted by cyber-crime and ingenious computer hacking. It is important to bear in mind that the winning margin (defined as the difference in performance between gold and silver medals, getting a medal or not, making a final or not in the Olympic athletic or swimming events) is <1% (200) so even small systematic advantages may be important motives and unfair advantages for doping.

 

The major gaps remaining in anti-doping science are (a) the lack of a definitive test for autologous blood transfusion, (b) need for more sensitive detection tests for peptide doping with wider windows of detection and (c) more economical, affordable and robust sample handling and storage procedures including dried blood spot sampling. These challenges must be met by adapting novel technologies such as quantitative proteomics, genomics, and metabolomics as well as implementing more out of competition and blood testing. Such progress depends on innovative applied research which is supported by WADA, Partnership for Clean Competition and certain national anti-doping organizations together with regular peer-review research granting agencies. Finally, the development of effective forensic intelligence investigations, a slow, complex and costly process but which can have salutary effects (e.g. for road cycling in the Lance Armstrong case), is proving a valuable complementary approach as an adjunct to effective laboratory testing.

 

REFERENCES

 

  1. Handelsman DJ, Gooren LJ. Hormones and sport: physiology, pharmacology and forensic science. Asian J Androl 2008; 10:348-350
  2. Rawls J. Justice as Fairness: A Restatement. Cambridge, MA: Belknap, Harvard University Press.
  3. Ryan A. Fairness and philosopy. Social Research 2006; 73:597-606
  4. Wiesing U. Should performance-enhancing drugs in sport be legalized under medical supervision? Sports Med 2011; 41:167-176
  5. Shuster S, Devine JW. The banning of sportsmen and women who fail drug tests is unjustifiable. The Journal of the Royal College of Physicians of Edinburgh 2013; 43:39-43
  6. Savulescu J, Creaney L, Vondy A. Should athletes be allowed to use performance enhancing drugs? BMJ 2013; 347:f6150
  7. Uvacsek M, Nepusz T, Naughton DP, Mazanov J, Ranky MZ, Petroczi A. Self-admitted behavior and perceived use of performance-enhancing vs psychoactive drugs among competitive athletes. Scandinavian journal of medicine & science in sports 2011; 21:224-234
  8. Petroczi A, Uvacsek M, Nepusz T, Deshmukh N, Shah I, Aidman EV, Barker J, Toth M, Naughton DP. Incongruence in doping related attitudes, beliefs and opinions in the context of discordant behavioural data: in which measure do we trust? PLoS One 2011; 6:e18804
  9. Dunn M, Thomas JO, Swift W, Burns L, Mattick RP. Drug testing in sport: the attitudes and experiences of elite athletes. Int J Drug Policy 2010; 21:330-332
  10. Bloodworth AJ, Petroczi A, Bailey R, Pearce G, McNamee MJ. Doping and supplementation: the attitudes of talented young athletes. Scandinavian journal of medicine & science in sports 2012; 22:293-301
  11. Lamberti N, Malagoni AM, Felisatti M, Caracciolo S, Resch N, Litmanen H, Dalfollo D, Jeannier P, Zhukovskaja L, Carrabre JE, Manfredini F. Antidoping attitudes among elite athletes: a cross sectional study in biathlon using a suitably developed questionnaire. J Sports Med Phys Fitness 2017; 57:610-623
  12. Backhouse SH, Whitaker L, Petroczi A. Gateway to doping? Supplement use in the context of preferred competitive situations, doping attitude, beliefs, and norms. Scandinavian journal of medicine & science in sports 2013; 23:244-252
  13. Morente-Sanchez J, Zabala M. Doping in sport: a review of elite athletes' attitudes, beliefs, and knowledge. Sports Med 2013; 43:395-411
  14. Petroczi A, Mazanov J, Nepusz T, Backhouse SH, Naughton DP. Comfort in big numbers: Does over-estimation of doping prevalence in others indicate self-involvement? J Occup Med Toxicol 2008; 3:19
  15. Dunn M, Thomas JO, Swift W, Burns L. Elite athletes' estimates of the prevalence of illicit drug use: evidence for the false consensus effect. Drug Alcohol Rev 2012; 31:27-32
  16. Thomas H. Murray. Good Sport. Oxford University Press.
  17. Eynon N, Ruiz JR, Oliveira J, Duarte JA, Birk R, Lucia A. Genes and elite athletes: a roadmap for future research. J Physiol 2011; 589:3063-3070
  18. de la Chapelle A, Traskelin AL, Juvonen E. Truncated erythropoietin receptor causes dominantly inherited benign human erythrocytosis. Proc Natl Acad Sci U S A 1993; 90:4495-4499
  19. Berman Y, North KN. A gene for speed: the emerging role of alpha-actinin-3 in muscle metabolism. Physiology (Bethesda) 2010; 25:250-259
  20. Wade N. Anabolic Steroids: Doctors Denounce Them, but Athletes Aren't Listening. Science 1972; 176:1399-1403
  21. Ljungqvist A. Half a century of challenges. Bioanalysis 2012; 4:1531-1533
  22. WADA. World Anti-Doping Code. Montreal: https://www.wada-ama.org/en/resources/the-code/world-anti-doping-code;2015.
  23. Egner IM, Bruusgaard JC, Eftestol E, Gundersen K. A cellular memory mechanism aids overload hypertrophy in muscle long after an episodic exposure to anabolic steroids. J Physiol 2013; 591:6221-6230
  24. de Hon O, Kuipers H, van Bottenburg M. Prevalence of doping use in elite sports: a review of numbers and methods. Sports Med 2015; 45:57-69
  25. Lensveldt-Mulders GJL, Hox JJ, van der Heijden PGM, Maas CJM. Meta-analysis of randomized response research. Socieological Methods & Research 2005; 33:319-348
  26. Ulrich R, Pope HG, Jr., Cleret L, Petroczi A, Nepusz T, Schaffer J, Kanayama G, Comstock RD, Simon P. Doping in Two Elite Athletics Competitions Assessed by Randomized-Response Surveys. Sports Med 2018; 48:211-219
  27. Schroter H, Studzinski B, Dietz P, Ulrich R, Striegel H, Simon P. A Comparison of the Cheater Detection and the Unrelated Question Models: A Randomized Response Survey on Physical and Cognitive Doping in Recreational Triathletes. PLoS One 2016; 11:e0155765
  28. WADA. International Standard for Therapeutic Use Exemptions. 2014. https://wada-main-prod.s3.amazonaws.com/resources/files/WADA-2015-ISTUE-Final-EN.pdf. Accessed Oct 2014
  29. Allan CM, Couse JF, Simanainen U, Spaliviero J, Jimenez M, Rodriguez K, Korach KS, Handelsman DJ. Estradiol induction of spermatogenesis is mediated via an estrogen receptor-{alpha} mechanism involving neuroendocrine activation of follicle-stimulating hormone secretion. Endocrinology 2010; 151:2800-2810
  30. WADA. Medical Information to Support the Decisions of TUECs - Androgen Deficiency-Male Hypogonadism. Montreal: WADA; April 20015 2015.
  31. David K, Dingemanse E, Freud J, Laqueur E. Uber krystallinisches mannliches Hormon aus Hoden (Testosteron), wirksamer als aus Harn oder aus Cholestrin bereitetes Androsteron. Hoppe Seylers Zeischrift Physiologische Chemie 1935; 233:281-282
  32. Yesalis CE, Courson SP, Wright JE. History of anabolic steroid use in sport and exercise. In: Yesalis CE, ed. Anabolic Steroids in Sports and Exercise. Champaign, IL: Human Kinetics; 2000:51-71.
  33. Franke WW, Berendonk B. Hormonal doping and androgenization of athletes: a secret program of the German Democratic Republic government. Clin Chem 1997; 43:1262-1279
  34. Ryan AJ. Anabolic steroids are fool's gold. Fed Proc 1981; 40:2682-2688
  35. Elashoff JD, Jacknow AD, Shain SG, Braunstein GD. Effects of anabolic-androgenic steroids on muscular strength. Ann Intern Med 1991; 115:387-393
  36. Bhasin S, Storer TW, Berman N, Callegari C, Clevenger B, Phillips J, Bunnell TJ, Tricker R, Shirazi A, Casaburi R. The effects of supraphysiologic doses of testosterone on muscle size and strength in normal men. N Engl J Med 1996; 335:1-7
  37. Woodhouse LJ, Reisz-Porszasz S, Javanbakht M, Storer TW, Lee M, Zerounian H, Bhasin S. Development of models to predict anabolic response to testosterone administration in healthy young men. Am J Physiol Endocrinol Metab 2003; 284:E1009-1017
  38. Storer TW, Magliano L, Woodhouse L, Lee ML, Dzekov C, Dzekov J, Casaburi R, Bhasin S. Testosterone dose-dependently increases maximal voluntary strength and leg power, but does not affect fatigability or specific tension. J Clin Endocrinol Metab 2003; 88:1478-1485
  39. Bhasin S, Woodhouse L, Casaburi R, Singh AB, Mac RP, Lee M, Yarasheski KE, Sinha-Hikim I, Dzekov C, Dzekov J, Magliano L, Storer TW. Older men are as responsive as young men to the anabolic effects of graded doses of testosterone on the skeletal muscle. J Clin Endocrinol Metab 2005; 90:678-688
  40. Thevis M, Schanzer W. Detection of SARMs in doping control analysis. Mol Cell Endocrinol 2018; 464:34-45
  41. Stenman UH, Hotakainen K, Alfthan H. Gonadotropins in doping: pharmacological basis and detection of illicit use. Br J Pharmacol 2008; 154:569-583
  42. Handelsman DJ. Indirect androgen doping by oestrogen blockade in sports. Br J Pharmacol 2008; 154:598-605
  43. Handelsman DJ. Clinical review: The rationale for banning human chorionic gonadotropin and estrogen blockers in sport. J Clin Endocrinol Metab 2006; 91:1646-1653
  44. Mauras N, Rogol AD, Veldhuis JD. Appraising the instantaneous secretory rates of luteinizing hormone and testosterone in response to selective mu opiate receptor blockade in late pubertal boys. J Androl 1987; 8:203-209
  45. Kicman AT. Pharmacology of anabolic steroids. Br J Pharmacol 2008; 154:502-521
  46. Thevis M, Schanzer W. Synthetic anabolic agents: steroids and nonsteroidal selective androgen receptor modulators. Handb Exp Pharmacol 2010:99-126
  47. Ponzetto F, Boccard J, Baume N, Kuuranne T, Rudaz S, Saugy M, Nicoli R. High-resolution mass spectrometry as an alternative detection method to tandem mass spectrometry for the analysis of endogenous steroids in serum. J Chromatogr B Analyt Technol Biomed Life Sci 2017; 1052:34-42
  48. Palonek E, Ericsson M, Garevik N, Rane A, Lehtihet M, Ekstrom L. Atypical excretion profile and GC/C/IRMS findings may last for nine months after a single dose of nandrolone decanoate. Steroids 2016; 108:105-111
  49. Sobolevsky T, Rodchenkov G. Detection and mass spectrometric characterization of novel long-term dehydrochloromethyltestosterone metabolites in human urine. J Steroid Biochem Mol Biol 2012; 128:121-127
  50. Sobolevsky T, Rodchenkov G. Mass spectrometric description of novel oxymetholone and desoxymethyltestosterone metabolites identified in human urine and their importance for doping control. Drug Test Anal 2012; 4:682-691
  51. Thevis M, Piper T, Beuck S, Geyer H, Schanzer W. Expanding sports drug testing assays: mass spectrometric characterization of the selective androgen receptor modulator drug candidates RAD140 and ACP-105. Rapid Commun Mass Spectrom 2013; 27:1173-1182
  52. Rzeppa S, Viet L. Analysis of sulfate metabolites of the doping agents oxandrolone and danazol using high performance liquid chromatography coupled to tandem mass spectrometry. J Chromatogr B Analyt Technol Biomed Life Sci 2016; 1029-1030:1-9
  53. Liu J, Chen L, Joseph JF, Nass A, Stoll A, de la Torre X, Botre F, Wolber G, Parr MK, Bureik M. Combined chemical and biotechnological production of 20betaOH-NorDHCMT, a long-term metabolite of Oral-Turinabol (DHCMT). J Inorg Biochem 2018; 183:165-171
  54. Esquivel A, Alechaga E, Monfort N, Ventura R. Sulfate metabolites improve retrospectivity after oral testosterone administration. Drug Test Anal 2019; 11:392-402
  55. Esquivel A, Alechaga E, Monfort N, Yang S, Xing Y, Moutian W, Ventura R. Evaluation of sulfate metabolites as markers of intramuscular testosterone administration in Caucasian and Asian populations. Drug Test Anal 2019; 11:1218-1230
  56. Kohler M, Thomas A, Geyer H, Petrou M, Schanzer W, Thevis M. Confiscated black market products and nutritional supplements with non-approved ingredients analyzed in the Cologne Doping Control Laboratory 2009. Drug Test Anal 2010; 2:533-537
  57. Handelsman DJ, Bermon S. Detection of testosterone doping in female athletes. Drug Test Anal 2019; 11:1566-1571
  58. Saugy M, Lundby C, Robinson N. Monitoring of biological markers indicative of doping: the athlete biological passport. Br J Sports Med 2014; 48:827-832
  59. Xue Y, Sun D, Daly A, Yang F, Zhou X, Zhao M, Huang N, Zerjal T, Lee C, Carter NP, Hurles ME, Tyler-Smith C. Adaptive evolution of UGT2B17 copy-number variation. Am J Hum Genet 2008; 83:337-346
  60. Schulze JJ, Lundmark J, Garle M, Skilving I, Ekstrom L, Rane A. Doping test results dependent on genotype of uridine diphospho-glucuronosyl transferase 2B17, the major enzyme for testosterone glucuronidation. J Clin Endocrinol Metab 2008; 93:2500-2506
  61. Schulze JJ, Lundmark J, Garle M, Ekstrom L, Sottas PE, Rane A. Substantial advantage of a combined Bayesian and genotyping approach in testosterone doping tests. Steroids 2009; 74:365-368
  62. Sottas PE, Saugy M, Saudan C. Endogenous steroid profiling in the athlete biological passport. Endocrinol Metab Clin North Am 2010; 39:59-73, viii-ix
  63. Martin-Escudero P, Munoz-Guerra JA, Garcia-Tenorio SV, Garde ES, Soldevilla-Navarro AB, Galindo-Canales M, Prado N, Fuentes-Ferrer ME, Fernandez-Perez C. Impact of the UGT2B17 polymorphism on the steroid profile. Results of a crossover clinical trial in athletes submitted to testosterone administration. Steroids 2019; 141:104-113
  64. Shackleton CH, Roitman E, Phillips A, Chang T. Androstanediol and 5-androstenediol profiling for detecting exogenously administered dihydrotestosterone, epitestosterone, and dehydroepiandrosterone: potential use in gas chromatography isotope ratio mass spectrometry. Steroids 1997; 62:665-673
  65. Piper T, Thevis M. Applications of Isotope Ratio Mass Spectrometry in Sports Drug Testing Accounting for Isotope Fractionation in Analysis of Biological Samples. Methods Enzymol 2017; 596:403-432
  66. Piper T, Mareck U, Geyer H, Flenker U, Thevis M, Platen P, Schanzer W. Determination of 13C/12C ratios of endogenous urinary steroids: method validation, reference population and application to doping control purposes. Rapid Commun Mass Spectrom 2008; 22:2161-2175
  67. Cawley AT, Trout GJ, Kazlauskas R, Howe CJ, George AV. Carbon isotope ratio (delta13C) values of urinary steroids for doping control in sport. Steroids 2009; 74:379-392
  68. Van Renterghem P, Van Eenoo P, Sottas PE, Saugy M, Delbeke F. Subject-based steroid profiling and the determination of novel biomarkers for DHT and DHEA misuse in sports. Drug Test Anal 2010; 2:582-588
  69. Brooker L, Cawley A, Drury J, Edey C, Hasick N, Goebel C. Stable carbon isotope ratio profiling of illicit testosterone preparations--domestic and international seizures. Drug Test Anal 2014; 6:996-1001
  70. Cawley A, Collins M, Kazlauskas R, Handelsman DJ, Heywood R, Longworth M, Arenas-Queralt A. Stable isotope ratio profiling of testosterone preparations. Drug Test Anal 2010; 2:557-567
  71. Van Renterghem P, Van Eenoo P, Sottas PE, Saugy M, Delbeke F. A pilot study on subject-based comprehensive steroid profiling: novel biomarkers to detect testosterone misuse in sports. Clin Endocrinol (Oxf) 2011;
  72. Jardines D, Botre F, Colamonici C, Curcio D, Procida G, de la Torre X. Longitudinal evaluation of the isotope ratio mass spectrometric data: towards the 'isotopic module' of the athlete biological passport? Drug Test Anal 2016; 8:1212-1221
  73. Piper T, Emery C, Saugy M. Recent developments in the use of isotope ratio mass spectrometry in sports drug testing. Anal Bioanal Chem 2011; 401:433-447
  74. Piper T, Emery C, Thomas A, Saugy M, Thevis M. Combination of carbon isotope ratio with hydrogen isotope ratio determinations in sports drug testing. Anal Bioanal Chem 2013; 405:5455-5466
  75. Thevis M, Piper T, Horning S, Juchelka D, Schanzer W. Hydrogen isotope ratio mass spectrometry and high-resolution/high-accuracy mass spectrometry in metabolite identification studies: detecting target compounds for sports drug testing. Rapid Commun Mass Spectrom 2013; 27:1904-1912
  76. Kicman AT, Brooks RV, Collyer SC, Cowan DA, Nanjee MN, Southan GJ, Wheeler MJ. Criteria to indicate testosterone administration. Br J Sports Med 1990; 24:253-264
  77. Palonek E, Gottlieb C, Garle M, Bjorkhem I, Carlstrom K. Serum and urinary markers of exogenous testosterone administration. J Steroid Biochem Mol Biol 1995; 55:121-127
  78. Goebel C, Howe CJ, Ho KK, Nelson A, Kazlauskas R, Trout GJ. Screening for testosterone abuse in male athletes using the measurement of urinary LH, a revision of the paradigm. Drug Test Anal 2009; 1:511-517
  79. Akram ON, Bursill C, Desai R, Heather AK, Kazlauskas R, Handelsman DJ, Lambert G. Evaluation of androgenic activity of nutraceutical-derived steroids using mammalian and yeast in vitro androgen bioassays. Anal Chem 2011; 83:2065-2074
  80. Handelsman DJ, Heather A. Androgen abuse in sports. Asian J Androl 2008; 10:403-415
  81. Zierau O, Lehmann S, Vollmer G, Schanzer W, Diel P. Detection of anabolic steroid abuse using a yeast transactivation system. Steroids 2008; 73:1143-1147
  82. Houtman CJ, Sterk SS, van de Heijning MP, Brouwer A, Stephany RW, van der Burg B, Sonneveld E. Detection of anabolic androgenic steroid abuse in doping control using mammalian reporter gene bioassays. Anal Chim Acta 2009; 637:247-258
  83. Cadwallader AB, Lim CS, Rollins DE, Botre F. The androgen receptor and its use in biological assays: looking toward effect-based testing and its applications. J Anal Toxicol 2011; 35:594-607
  84. Death AK, McGrath KC, Kazlauskas R, Handelsman DJ. Tetrahydrogestrinone is a potent androgen and progestin. J Clin Endocrinol Metab 2004; 89:2498-2500
  85. McRobb L, Handelsman DJ, Kazlauskas R, Wilkinson S, McLeod MD, Heather AK. Structure-activity relationships of synthetic progestins in a yeast-based in vitro androgen bioassay. Journal of Steroid Biochemistry and Molecular Biology 2008; 110:39-47
  86. Thevis M, Geyer H, Tretzel L, Schanzer W. Sports drug testing using complementary matrices: Advantages and limitations. J Pharm Biomed Anal 2016; 130:220-230
  87. Kintz P. Testing for anabolic steroids in hair: a review. Legal medicine 2003; 5 Suppl 1:S29-33
  88. Deng XS, Kurosu A, Pounder DJ. Detection of anabolic steroids in head hair. J Forensic Sci 1999; 44:343-346
  89. Kintz P, Cirimele V, Sachs H, Jeanneau T, Ludes B. Testing for anabolic steroids in hair from two bodybuilders. Forensic Sci Int 1999; 101:209-216
  90. Kintz P, Cirimele V, Jeanneau T, Ludes B. Identification of testosterone and testosterone esters in human hair. J Anal Toxicol 1999; 23:352-356
  91. Hold KM, Borges CR, Wilkins DG, Rollins DE, Joseph RE, Jr. Detection of nandrolone, testosterone, and their esters in rat and human hair samples. J Anal Toxicol 1999; 23:416-423
  92. Gaillard Y, Vayssette F, Balland A, Pepin G. Gas chromatographic-tandem mass spectrometric determination of anabolic steroids and their esters in hair. Application in doping control and meat quality control. J Chromatogr B Biomed Sci Appl 1999; 735:189-205
  93. Gaillard Y, Vayssette F, Pepin G. Compared interest between hair analysis and urinalysis in doping controls. Results for amphetamines, corticosteroids and anabolic steroids in racing cyclists. Forensic Sci Int 2000; 107:361-379
  94. Cirimele V, Kintz P, Ludes B. Testing of the anabolic stanozolol in human hair by gas chromatography-negative ion chemical ionization mass spectrometry. J Chromatogr B Biomed Sci Appl 2000; 740:265-271
  95. Kintz P, Cirimele V, Ludes B. Discrimination of the nature of doping with 19-norsteroids through hair analysis. Clin Chem 2000; 46:2020-2022
  96. Kintz P, Cirimele V, Dumestre-Toulet V, Ludes B. Doping control for nandrolone using hair analysis. J Pharm Biomed Anal 2001; 24:1125-1130
  97. Dumestre-Toulet V, Cirimele V, Ludes B, Gromb S, Kintz P. Hair analysis of seven bodybuilders for anabolic steroids, ephedrine, and clenbuterol. J Forensic Sci 2002; 47:211-214
  98. Kintz P, Cirimele V, Dumestre-Toulet V, Villain M, Ludes B. Doping control for methenolone using hair analysis by gas chromatography-tandem mass spectrometry. J Chromatogr B Analyt Technol Biomed Life Sci 2002; 766:161-167
  99. Bresson M, Cirimele V, Villain M, Kintz P. Doping control for metandienone using hair analyzed by gas chromatography-tandem mass spectrometry. J Chromatogr B Analyt Technol Biomed Life Sci 2006; 836:124-128
  100. Gambelunghe C, Sommavilla M, Ferranti C, Rossi R, Aroni K, Manes N, Bacci M. Analysis of anabolic steroids in hair by GC/MS/MS. Biomedical chromatography : BMC 2007; 21:369-375
  101. Deshmukh N, Hussain I, Barker J, Petroczi A, Naughton DP. Analysis of anabolic steroids in human hair using LC-MS/MS. Steroids 2010; 75:710-714
  102. Strano-Rossi S, Castrignano E, Anzillotti L, Odoardi S, De-Giorgio F, Bermejo A, Pascali VL. Screening for exogenous androgen anabolic steroids in human hair by liquid chromatography/orbitrap-high resolution mass spectrometry. Anal Chim Acta 2013; 793:61-71
  103. Kintz P, Cirimele V, Ludes B. Physiological concentrations of DHEA in human hair. J Anal Toxicol 1999; 23:424-428
  104. Kintz P, Cirimel V, Devaux M, Ludes B. Dehydroepiandrosterone (DHEA) and testosterone concentrations in human hair after chronic DHEA supplementation. Clin Chem 2000; 46:414-415
  105. Rambaud L, Monteau F, Deceuninck Y, Bichon E, Andre F, Le Bizec B. Development and validation of a multi-residue method for the detection of a wide range of hormonal anabolic compounds in hair using gas chromatography-tandem mass spectrometry. Anal Chim Acta 2007; 586:93-104
  106. Deshmukh NI, Barker J, Petroczi A, Naughton DP. Detection of testosterone and epitestosterone in human hair using liquid chromatography-tandem mass spectrometry. J Pharm Biomed Anal 2012; 67-68:154-158
  107. Segura J, Pichini S, Peng SH, de la Torre X. Hair analysis and detectability of single dose administration of androgenic steroid esters. Forensic Sci Int 2000; 107:347-359
  108. Brown HG, Perrett D. Detection of doping in sport: detecting anabolic-androgenic steroids in human fingernail clippings. The Medico-legal journal 2011; 79:67-69
  109. Anizan S, Huestis MA. The potential role of oral fluid in antidoping testing. Clin Chem 2014; 60:307-322
  110. Schonfelder M, Hofmann H, Schulz T, Engl T, Kemper D, Mayr B, Rautenberg C, Oberhoffer R, Thieme D. Potential detection of low-dose transdermal testosterone administration in blood, urine, and saliva. Drug Test Anal 2016; 8:1186-1196
  111. Chiodini I, Ramos-Rivera A, Marcus AO, Yau H. Adrenal Hypercortisolism: A Closer Look at Screening, Diagnosis, and Important Considerations of Different Testing Modalities. J Endocr Soc 2019; 3:1097-1109
  112. Welker KM, Lassetter B, Brandes CM, Prasad S, Koop DR, Mehta PH. A comparison of salivary testosterone measurement using immunoassays and tandem mass spectrometry. Psychoneuroendocrinology 2016; 71:180-188
  113. Thevis M, Krug O, Geyer H, Schanzer W. Expanding analytical options in sports drug testing: Mass spectrometric detection of prohibited substances in exhaled breath. Rapid Commun Mass Spectrom 2017; 31:1290-1296
  114. Schonfelder M, Hofmann H, Anielski P, Thieme D, Oberhoffer R, Michna H. Gene expression profiling in human whole blood samples after controlled testosterone application and exercise. Drug Test Anal 2011; 3:652-660
  115. Woldemariam GA, Butch AW. Immunoextraction-tandem mass spectrometry method for measuring intact human chorionic gonadotropin, free beta-subunit, and beta-subunit core fragment in urine. Clin Chem 2014; 60:1089-1097
  116. Butch AW, Woldemariam GA. Urinary human chorionic gonadotropin isoform concentrations in doping control samples. Drug Test Anal 2016; 8:1147-1151
  117. Butch AW, Ahrens BD, Avliyakulov NK. Urine reference intervals for human chorionic gonadotropin (hCG) isoforms by immunoextraction-tandem mass spectrometry to detect hCG use. Drug Test Anal 2018; 10:956-960
  118. Handelsman DJ, Goebel C, Idan A, Jimenez M, Trout G, Kazlauskas R. Effects of recombinant human LH and hCG on serum and urine LH and androgens in men. Clinical Endocrinology 2009; 71:417-428
  119. Handelsman DJ, Idan A, Grainger J, Goebel C, Turner L, Conway AJ. Detection and effects on serum and urine steroid and LH of repeated GnRH analog (leuprolide) stimulation. J Steroid Biochem Mol Biol 2014; 141:113-120
  120. Graham MR, Ryan P, Baker JS, Davies B, Thomas NE, Cooper SM, Evans P, Easmon S, Walker CJ, Cowan D, Kicman AT. Counterfeiting in performance- and image-enhancing drugs. Drug Test Anal 2009; 1:135-142
  121. Elliott S. Erythropoiesis-stimulating agents and other methods to enhance oxygen transport. Br J Pharmacol 2008; 154:529-541
  122. Reichel C, Gmeiner G. Erythropoietin and analogs. Handb Exp Pharmacol 2010:251-294
  123. di Prampero PE, Ferretti G. Factors limiting maximal oxygen consumption in humans. Respiration physiology 1990; 80:113-127
  124. Pace N, Consolazio WV, Lozner EL. The effect of transfusions of red blood cells on the hypoxia tolerance of normal men. Science 1945; 102:589-591
  125. Pace N, Lozner EL, et al. The increase in hypoxia tolerance of normal men accompanying the polycythemia induced by transfusion of erythrocytes. Am J Physiol 1947; 148:152-163
  126. Ekblom B, Goldbarg AN, Gullbring B. Response to exercise after blood loss and reinfusion. J Appl Physiol 1972; 33:175-180
  127. Hoberman J. History and prevalence of doping in the marathon. Sports Med 2007; 37:386-388
  128. Birkeland KI, Stray-Gundersen J, Hemmersbach P, Hallen J, Haug E, Bahr R. Effect of rhEPO administration on serum levels of sTfR and cycling performance. Med Sci Sports Exerc 2000; 32:1238-1243
  129. Ashenden MJ, Hahn AG, Martin DT, Logan P, Parisotto R, Gore CJ. A comparison of the physiological response to simulated altitude exposure and r-HuEpo administration. J Sports Sci 2001; 19:831-837
  130. Russell G, Gore CJ, Ashenden MJ, Parisotto R, Hahn AG. Effects of prolonged low doses of recombinant human erythropoietin during submaximal and maximal exercise. Eur J Appl Physiol 2002; 86:442-449
  131. Nelson M, Popp H, Sharpe K, Ashenden M. Proof of homologous blood transfusion through quantification of blood group antigens. Haematologica 2003; 88:1284-1295
  132. Cartron JP, Colin Y. Structural and functional diversity of blood group antigens. Transfusion clinique et biologique : journal de la Societe francaise de transfusion sanguine 2001; 8:163-199
  133. Giraud S, Robinson N, Mangin P, Saugy M. Scientific and forensic standards for homologous blood transfusion anti-doping analyses. Forensic Sci Int 2008; 179:23-33
  134. Voss SC, Thevis M, Schinkothe T, Schanzer W. Detection of homologous blood transfusion. Int J Sports Med 2007; 28:633-637
  135. Manokhina I, Rupert JL. A DNA-based method for detecting homologous blood doping. Anal Bioanal Chem 2013;
  136. Stampella A, Di Marco S, Pirri D, de la Torre X, Botre F, Donati F. Application of DNA-based forensic analysis for the detection of homologous transfusion of whole blood and of red blood cell concentrates in doping control. Forensic Sci Int 2016; 265:204-210
  137. Giraud S, Sottas PE, Robinson N, Saugy M. Blood transfusion in sports. Handb Exp Pharmacol 2010:295-304
  138. Segura J, Monfort N, Ventura R. Detection methods for autologous blood doping. Drug Test Anal 2012; 4:876-881
  139. Donati F, Acciarini R, De Benedittis I, de la Torre X, Pirri D, Prete M, Stampella A, Vernucci E, Botre F. Detecting Autologous Blood Transfusion in Doping Control: Biomarkers of Blood Aging and Storage Measured by Flow Cytofluorimetry. Curr Pharm Biotechnol 2018; 19:124-135
  140. Doctor A, Spinella P. Effect of processing and storage on red blood cell function in vivo. Seminars in perinatology 2012; 36:248-259
  141. Monfort N, Ventura R, Balcells G, Segura J. Determination of five di-(2-ethylhexyl)phthalate metabolites in urine by UPLC-MS/MS, markers of blood transfusion misuse in sports. J Chromatogr B Analyt Technol Biomed Life Sci 2012; 908:113-121
  142. Pottgiesser T, Umhau M, Ahlgrim C, Ruthardt S, Roecker K, Schumacher YO. Hb mass measurement suitable to screen for illicit autologous blood transfusions. Med Sci Sports Exerc 2007; 39:1748-1756
  143. Morkeberg J, Sharpe K, Belhage B, Damsgaard R, Schmidt W, Prommer N, Gore CJ, Ashenden MJ. Detecting autologous blood transfusions: a comparison of three passport approaches and four blood markers. Scandinavian journal of medicine & science in sports 2011; 21:235-243
  144. Pottgiesser T, Echteler T, Sottas PE, Umhau M, Schumacher YO. Hemoglobin mass and biological passport for the detection of autologous blood doping. Med Sci Sports Exerc 2012; 44:835-843
  145. Ashenden M, Gough CE, Garnham A, Gore CJ, Sharpe K. Current markers of the Athlete Blood Passport do not flag microdose EPO doping. Eur J Appl Physiol 2011; 111:2307-2314
  146. Kannan M, Atreya C. Differential profiling of human red blood cells during storage for 52 selected microRNAs. Transfusion 2010; 50:1581-1588
  147. Pottgiesser T, Schumacher YO, Funke H, Rennert K, Baumstark MW, Neunuebel K, Mosig S. Gene expression in the detection of autologous blood transfusion in sports--a pilot study. Vox sanguinis 2009; 96:333-336
  148. Sottas PE, Robinson N, Saugy M. The athlete's biological passport and indirect markers of blood doping. Handb Exp Pharmacol 2010:305-326
  149. Sottas PE, Robinson N, Rabin O, Saugy M. The athlete biological passport. Clin Chem 2011; 57:969-976
  150. Pottgiesser T, Schumacher YO. Current strategies of blood doping detection. Anal Bioanal Chem 2013;
  151. Cazzola M. A global strategy for prevention and detection of blood doping with erythropoietin and related drugs. Haematologica 2000; 85:561-563
  152. Parisotto R, Gore CJ, Emslie KR, Ashenden MJ, Brugnara C, Howe C, Martin DT, Trout GJ, Hahn AG. A novel method utilising markers of altered erythropoiesis for the detection of recombinant human erythropoietin abuse in athletes. Haematologica 2000; 85:564-572
  153. Parisotto R, Wu M, Ashenden MJ, Emslie KR, Gore CJ, Howe C, Kazlauskas R, Sharpe K, Trout GJ, Xie M. Detection of recombinant human erythropoietin abuse in athletes utilizing markers of altered erythropoiesis. Haematologica 2001; 86:128-137
  154. Gore CJ, Parisotto R, Ashenden MJ, Stray-Gundersen J, Sharpe K, Hopkins W, Emslie KR, Howe C, Trout GJ, Kazlauskas R, Hahn AG. Second-generation blood tests to detect erythropoietin abuse by athletes. Haematologica 2003; 88:333-344
  155. Malcovati L, Pascutto C, Cazzola M. Hematologic passport for athletes competing in endurance sports: a feasibility study. Haematologica 2003; 88:570-581
  156. Sharpe K, Ashenden MJ, Schumacher YO. A third generation approach to detect erythropoietin abuse in athletes. Haematologica 2006; 91:356-363
  157. Sottas PE, Robinson N, Giraud S, Tarioni F, Kamber M, Mangin P, Saugy M. Statistical classificiation of abnormal blood profiles inathletes. International Journal of Biostatistics 2006; 2:3
  158. WADA. Athlete Biological Passport: Operating guidelines and compilation of required elements. Verson 3.1. World Anti-Doping Agency;2012.
  159. Ekblom B, Berglund B. Effect of erythropoeitin administration on maximal aerobic power in man. Scandanavian Journal of Medicine and Science in Sport 1991; 1:88-93
  160. Jelkmann W. Biosimilar recombinant human erythropoietins ("epoetins") and future erythropoiesis-stimulating treatments. Expert Opin Biol Ther 2012; 12:581-592
  161. Catlin DH, Fitch KD, Ljungqvist A. Medicine and science in the fight against doping in sport. J Intern Med 2008; 264:99-114
  162. Lopez B. The invention of a 'drug of mass destruction': deconstructing the EPO myth. Sport in History 2011; 31:84-109
  163. Lasne F, de Ceaurriz J. Recombinant erythropoietin in urine. Nature 2000; 405:635
  164. Lasne F, Martin L, Crepin N, de Ceaurriz J. Detection of isoelectric profiles of erythropoietin in urine: differentiation of natural and administered recombinant hormones. Anal Biochem 2002; 311:119-126
  165. Lasne F, Thioulouse J, Martin L, de Ceaurriz J. Detection of recombinant human erythropoietin in urine for doping analysis: interpretation of isoelectric profiles by discriminant analysis. Electrophoresis 2007; 28:1875-1881
  166. Leuenberger N, Reichel C, Lasne F. Detection of erythropoiesis-stimulating agents in human anti-doping control: past, present and future. Bioanalysis 2012; 4:1565-1575
  167. Breidbach A, Catlin DH, Green GA, Tregub I, Truong H, Gorzek J. Detection of recombinant human erythropoietin in urine by isoelectric focusing. Clin Chem 2003; 49:901-907
  168. Wickramasinghe S, Medrano JF. Primer on genes encoding enzymes in sialic acid metabolism in mammals. Biochimie 2011; 93:1641-1646
  169. Reverter-Branchat G, Ventura R, Ezzel Din M, Mateus J, Pedro C, Segura J. Detection of erythropoiesis-stimulating agents in a single dried blood spot. Drug Test Anal 2018; 10:1496-1507
  170. Durussel J, Haile DW, Mooses K, Daskalaki E, Beattie W, Mooses M, Mekonen W, Ongaro N, Anjila E, Patel RK, Padmanabhan N, McBride MW, McClure JD, Pitsiladis YP. Blood transcriptional signature of recombinant human erythropoietin administration and implications for antidoping strategies. Physiol Genomics 2016; 48:202-209
  171. Beuck S, Schanzer W, Thevis M. Hypoxia-inducible factor stabilizers and other small-molecule erythropoiesis-stimulating agents in current and preventive doping analysis. Drug Test Anal 2012; 4:830-845
  172. La Merie Publishing. Blockbuster Biologics 2012. La Merie Business Intlligence;2013.
  173. Imagawa S, Matsumoto K, Horie M, Ohkoshi N, Nagasawa T, Doi T, Suzuki N, Yamamoto M. Does k-11706 enhance performance and why? Int J Sports Med 2007; 28:928-933
  174. Richardson RS, Tagore K, Haseler LJ, Jordan M, Wagner PD. Increased VO2 max with right-shifted Hb-O2 dissociation curve at a constant O2 delivery in dog muscle in situ. Journal of applied physiology (Bethesda, Md : 1985) 1998; 84:995-1002
  175. Pagel PS, Hettrick DA, Montgomery MW, Kersten JR, Steffen RP, Warltier DC. RSR13, a synthetic modifier of hemoglobin-oxygen affinity, enhances the recovery of stunned myocardium in anesthetized dogs. J Pharmacol Exp Ther 1998; 285:1-8
  176. Breidbach A, Catlin DH. RSR13, a potential athletic performance enhancement agent: detection in urine by gas chromatography/mass spectrometry. Rapid Commun Mass Spectrom 2001; 15:2379-2382
  177. Thevis M, Krug O, Schanzer W. Mass spectrometric characterization of efaproxiral (RSR13) and its implementation into doping controls using liquid chromatography-atmospheric pressure ionization-tandem mass spectrometry. J Mass Spectrom 2006; 41:332-338
  178. Casadevall N, Nataf J, Viron B, Kolta A, Kiladjian JJ, Martin-Dupont P, Michaud P, Papo T, Ugo V, Teyssandier I, Varet B, Mayeux P. Pure red-cell aplasia and antierythropoietin antibodies in patients treated with recombinant erythropoietin. N Engl J Med 2002; 346:469-475
  179. Macdougall IC, Roger SD, de Francisco A, Goldsmith DJ, Schellekens H, Ebbers H, Jelkmann W, London G, Casadevall N, Horl WH, Kemeny DM, Pollock C. Antibody-mediated pure red cell aplasia in chronic kidney disease patients receiving erythropoiesis-stimulating agents: new insights. Kidney Int 2012; 81:727-732
  180. Unger EF, Thompson AM, Blank MJ, Temple R. Erythropoiesis-stimulating agents--time for a reevaluation. N Engl J Med 2010; 362:189-192
  181. Bennett CL, Silver SM, Djulbegovic B, Samaras AT, Blau CA, Gleason KJ, Barnato SE, Elverman KM, Courtney DM, McKoy JM, Edwards BJ, Tigue CC, Raisch DW, Yarnold PR, Dorr DA, Kuzel TM, Tallman MS, Trifilio SM, West DP, Lai SY, Henke M. Venous thromboembolism and mortality associated with recombinant erythropoietin and darbepoetin administration for the treatment of cancer-associated anemia. JAMA 2008; 299:914-924
  182. Strippoli GF, Navaneethan SD, Craig JC. Haemoglobin and haematocrit targets for the anaemia of chronic kidney disease. Cochrane Database Syst Rev 2006:CD003967
  183. Tonia T, Mettler A, Robert N, Schwarzer G, Seidenfeld J, Weingart O, Hyde C, Engert A, Bohlius J. Erythropoietin or darbepoetin for patients with cancer. Cochrane Database Syst Rev 2012; 12:CD003407
  184. Rampling MW. Hyperviscosity as a complication in a variety of disorders. Semin Thromb Hemost 2003; 29:459-465
  185. Kovesdy CP, Kalantar-Zadeh K. Iron therapy in chronic kidney disease: current controversies. Journal of renal care 2009; 35 Suppl 2:14-24
  186. Gagnon DR, Zhang TJ, Brand FN, Kannel WB. Hematocrit and the risk of cardiovascular disease--the Framingham study: a 34-year follow-up. Am Heart J 1994; 127:674-682
  187. Danesh J, Collins R, Peto R, Lowe GD. Haematocrit, viscosity, erythrocyte sedimentation rate: meta-analyses of prospective studies of coronary heart disease. Eur Heart J 2000; 21:515-520
  188. Braekkan SK, Mathiesen EB, Njolstad I, Wilsgaard T, Hansen JB. Hematocrit and risk of venous thromboembolism in a general population. The Tromso study. Haematologica 2010; 95:270-275
  189. Holt RI, Erotokritou-Mulligan I, Sonksen PH. The history of doping and growth hormone abuse in sport. Growth Horm IGF Res 2009; 19:320-326
  190. Liu H, Bravata DM, Olkin I, Friedlander A, Liu V, Roberts B, Bendavid E, Saynina O, Salpeter SR, Garber AM, Hoffman AR. Systematic review: the effects of growth hormone on athletic performance. Ann Intern Med 2008; 148:747-758
  191. Baumann GP. Growth hormone doping in sports: a critical review of use and detection strategies. Endocr Rev 2012; 33:155-186
  192. Holt RIG, Ho KKY. The Use and Abuse of Growth Hormone in Sports. Endocr Rev 2019; 40:1163-1185
  193. Meinhardt U, Nelson AE, Hansen JL, Birzniece V, Clifford D, Leung KC, Graham K, Ho KK. The effects of growth hormone on body composition and physical performance in recreational athletes: a randomized trial. Ann Intern Med 2010; 152:568-577
  194. Berggren A, Ehrnborg C, Rosen T, Ellegard L, Bengtsson BA, Caidahl K. Short-term administration of supraphysiological recombinant human growth hormone (GH) does not increase maximum endurance exercise capacity in healthy, active young men and women with normal GH-insulin-like growth factor I axes. J Clin Endocrinol Metab 2005; 90:3268-3273
  195. Irving BA, Patrie JT, Anderson SM, Watson-Winfield DD, Frick KI, Evans WS, Veldhuis JD, Weltman A. The effects of time following acute growth hormone administration on metabolic and power output measures during acute exercise. J Clin Endocrinol Metab 2004; 89:4298-4305
  196. Graham MR, Baker JS, Evans P, Kicman A, Cowan D, Hullin D, Davies B. Short-term recombinant human growth hormone administration improves respiratory function in abstinent anabolic-androgenic steroid users. Growth Horm IGF Res 2007; 17:328-335
  197. Goto K, Doessing S, Nielsen RH, Flyvbjerg A, Kjaer M. Growth hormone receptor antagonist treatment reduces exercise performance in young males. J Clin Endocrinol Metab 2009; 94:3265-3272
  198. Healy ML, Gibney J, Russell-Jones DL, Pentecost C, Croos P, Sonksen PH, Umpleby AM. High dose growth hormone exerts an anabolic effect at rest and during exercise in endurance-trained athletes. J Clin Endocrinol Metab 2003; 88:5221-5226
  199. Healy ML, Gibney J, Pentecost C, Croos P, Russell-Jones DL, Sonksen PH, Umpleby AM. Effects of high-dose growth hormone on glucose and glycerol metabolism at rest and during exercise in endurance-trained athletes. J Clin Endocrinol Metab 2006; 91:320-327
  200. Handelsman DJ, Hirschberg AL, Bermon S. Circulating Testosterone as the Hormonal Basis of Sex Differences in Athletic Performance. Endocr Rev 2018; 39:803-829
  201. Breederveld RS, Tuinebreijer WE. Recombinant human growth hormone for treating burns and donor sites. Cochrane Database Syst Rev 2012; 12:CD008990
  202. Takala J, Ruokonen E, Webster NR, Nielsen MS, Zandstra DF, Vundelinckx G, Hinds CJ. Increased mortality associated with growth hormone treatment in critically ill adults N Engl J Med 1999; 341:785-792
  203. Raschke M, Rasmussen MH, Govender S, Segal D, Suntum M, Christiansen JS. Effects of growth hormone in patients with tibial fracture: a randomised, double-blind, placebo-controlled clinical trial. Eur J Endocrinol 2007; 156:341-351
  204. van den Berg G, Frolich M, Veldhuis JD, Roelfsema F. Growth hormone secretion in recently operated acromegalic patients. J Clin Endocrinol Metab 1994; 79:1706-1715
  205. Fuchtbauer L, Olsson DS, Bengtsson BA, Norrman LL, Sunnerhagen KS, Johannsson G. Muscle strength in patients with acromegaly at diagnosis and during long-term follow-up. Eur J Endocrinol 2017; 177:217-226
  206. Sklar CA, Mertens AC, Mitby P, Occhiogrosso G, Qin J, Heller G, Yasui Y, Robison LL. Risk of disease recurrence and second neoplasms in survivors of childhood cancer treated with growth hormone: a report from the Childhood Cancer Survivor Study. J Clin Endocrinol Metab 2002; 87:3136-3141
  207. Ergun-Longmire B, Mertens AC, Mitby P, Qin J, Heller G, Shi W, Yasui Y, Robison LL, Sklar CA. Growth hormone treatment and risk of second neoplasms in the childhood cancer survivor. J Clin Endocrinol Metab 2006; 91:3494-3498
  208. Carel JC, Ecosse E, Landier F, Meguellati-Hakkas D, Kaguelidou F, Rey G, Coste J. Long-term mortality after recombinant growth hormone treatment for isolated growth hormone deficiency or childhood short stature: preliminary report of the French SAGhE study. J Clin Endocrinol Metab 2012; 97:416-425
  209. Mo D, Hardin DS, Erfurth EM, Melmed S. Adult mortality or morbidity is not increased in childhood-onset growth hormone deficient patients who received pediatric GH treatment: an analysis of the Hypopituitary Control and Complications Study (HypoCCS). Pituitary 2013;
  210. Swerdlow AJ, Higgins CD, Adlard P, Preece MA. Risk of cancer in patients treated with human pituitary growth hormone in the UK, 1959-85: a cohort study. Lancet 2002; 360:273-277
  211. Wilton P, Mattsson AF, Darendeliler F. Growth hormone treatment in children is not associated with an increase in the incidence of cancer: experience from KIGS (Pfizer International Growth Database). J Pediatr 2010; 157:265-270
  212. Mackenzie S, Craven T, Gattamaneni HR, Swindell R, Shalet SM, Brabant G. Long-term safety of growth hormone replacement after CNS irradiation. J Clin Endocrinol Metab 2011; 96:2756-2761
  213. Woodmansee WW, Zimmermann AG, Child CJ, Rong Q, Erfurth EM, Beck-Peccoz P, Blum WF, Robison LL. Incidence of second neoplasm in childhood cancer survivors treated with GH: an analysis of GeNeSIS and HypoCCS. Eur J Endocrinol 2013; 168:565-573
  214. Jenkins PJ, Mukherjee A, Shalet SM. Does growth hormone cause cancer? Clin Endocrinol (Oxf) 2006; 64:115-121
  215. Holly J, Perks C. Growth hormone and cancer: are we asking the right questions?*. Clin Endocrinol (Oxf) 2006; 64:122-124
  216. Ho KK, Nelson AE. Growth hormone in sports: detecting the doped or duped. Hormone research in paediatrics 2011; 76 Suppl 1:84-90
  217. Hepner F, Cszasar E, Roitinger E, Lubec G. Mass spectrometrical analysis of recombinant human growth hormone (Genotropin(R)) reveals amino acid substitutions in 2% of the expressed protein. Proteome science 2005; 3:1
  218. Hepner F, Csaszar E, Roitinger E, Pollak A, Lubec G. Mass spectrometrical analysis of recombinant human growth hormone Norditropin reveals amino acid exchange at M14_V14 rhGH. Proteomics 2006; 6:775-784
  219. Jiang H, Wu SL, Karger BL, Hancock WS. Mass spectrometric analysis of innovator, counterfeit, and follow-on recombinant human growth hormone. Biotechnology progress 2009; 25:207-218
  220. Bidlingmaier M, Strasburger CJ. Growth hormone. Handb Exp Pharmacol 2010:187-200
  221. Bosch J, Luchini A, Pichini S, Tamburro D, Fredolini C, Liotta L, Petricoin E, Pacifici R, Facchiano F, Segura J, Garaci E, Gutierrez-Gallego R. Analysis of urinary human growth hormone (hGH) using hydrogel nanoparticles and isoform differential immunoassays after short recombinant hGH treatment: preliminary results. J Pharm Biomed Anal 2013; 85:194-197
  222. Bidlingmaier M, Suhr J, Ernst A, Wu Z, Keller A, Strasburger CJ, Bergmann A. High-sensitivity chemiluminescence immunoassays for detection of growth hormone doping in sports. Clin Chem 2009; 55:445-453
  223. Bidlingmaier M, Manolopoulou J. Detecting growth hormone abuse in athletes. Endocrinol Metab Clin North Am 2010; 39:25-32, vii
  224. Marks V. Murder by insulin: suspected, purported and proven-a review. Drug Test Anal 2009; 1:162-176
  225. Will RG. Acquired prion disease: iatrogenic CJD, variant CJD, kuru. Br Med Bull 2003; 66:255-265
  226. Brown P, Preece M, Brandel JP, Sato T, McShane L, Zerr I, Fletcher A, Will RG, Pocchiari M, Cashman NR, d'Aignaux JH, Cervenakova L, Fradkin J, Schonberger LB, Collins SJ. Iatrogenic Creutzfeldt-Jakob disease at the millennium. Neurology 2000; 55:1075-1081
  227. Barroso O, Schamasch P, Rabin O. Detection of GH abuse in sport: Past, present and future. Growth Horm IGF Res 2009; 19:369-374
  228. Travis J. Pharmacology. Growth hormone test finally nabs first doper. Science 2010; 327:1185
  229. Powrie JK, Bassett EE, Rosen T, Jorgensen JO, Napoli R, Sacca L, Christiansen JS, Bengtsson BA, Sonksen PH, Group GHPS. Detection of growth hormone abuse in sport. Growth Horm IGF Res 2007; 17:220-226
  230. Nelson AE, Meinhardt U, Hansen JL, Walker IH, Stone G, Howe CJ, Leung KC, Seibel MJ, Baxter RC, Handelsman DJ, Kazlauskas R, Ho KK. Pharmacodynamics of growth hormone abuse biomarkers and the influence of gender and testosterone: a randomized double-blind placebo-controlled study in young recreational athletes. Journal of Clinical Endocrinology and Metabolism 2008; 93:2213-2222
  231. Krusenstjerna-Hafstrom T, Rasmussen MH, Raschke M, Govender S, Madsen J, Christiansen JS. Biochemical markers of bone turnover in tibia fracture patients randomly assigned to growth hormone (GH) or placebo injections: Implications for detection of GH abuse. Growth Horm IGF Res 2011; 21:331-335
  232. Barroso O, Handelsman DJ, Strasburger C, Thevis M. Analytical challenges in the detection of peptide hormones for anti-doping purposes. Bioanalysis 2012; 4:1577-1590
  233. Williams RM, McDonald A, Savage MO, Dunger DB. Mecasermin rinfabate: rhIGF-I/rhIGFBP-3 complex: iPLEX. Expert Opin Drug Metab Toxicol 2008; 4:311-324
  234. Guha N, Dashwood A, Thomas NJ, Skingle AJ, Sonksen PH, Holt RI. IGF-I abuse in sport. Current drug abuse reviews 2009; 2:263-272
  235. Ernst S, Simon P. A quantitative approach for assessing significant improvements in elite sprint performance: has IGF-1 entered the arena? Drug Test Anal 2013; 5:384-389
  236. Thomas A, Kohler M, Schanzer W, Delahaut P, Thevis M. Determination of IGF-1 and IGF-2, their degradation products and synthetic analogues in urine by LC-MS/MS. Analyst 2011; 136:1003-1012
  237. Hess C, Thomas A, Thevis M, Stratmann B, Quester W, Tschoepe D, Madea B, Musshoff F. Simultaneous determination and validated quantification of human insulin and its synthetic analogues in human blood serum by immunoaffinity purification and liquid chromatography-mass spectrometry. Anal Bioanal Chem 2012; 404:1813-1822
  238. Guha N, Erotokritou-Mulligan I, Nevitt SP, Francis M, Bartlett C, Cowan DA, Bassett EE, Sonksen PH, Holt RI. Biochemical markers of recombinant human insulin-like growth factor-I (rhIGF-I)/rhIGF binding protein-3 (rhIGFBP-3) misuse in athletes. Drug Test Anal 2013;
  239. Guha N, Cowan DA, Sonksen PH, Holt RI. Insulin-like growth factor-I (IGF-I) misuse in athletes and potential methods for detection. Anal Bioanal Chem 2013; 405:9669-9683
  240. Matheny RW, Jr., Nindl BC, Adamo ML. Minireview: Mechano-growth factor: a putative product of IGF-I gene expression involved in tissue repair and regeneration. Endocrinology 2010; 151:865-875
  241. Esposito S, Deventer K, Van Eenoo P. Characterization and identification of a C-terminal amidated mechano growth factor (MGF) analogue in black market products. Rapid Commun Mass Spectrom 2012; 26:686-692
  242. Thevis M, Thomas A, Schanzer W. Insulin. Handb Exp Pharmacol 2010:209-226
  243. Anderson LJ, Tamayose JM, Garcia JM. Use of growth hormone, IGF-I, and insulin for anabolic purpose: Pharmacological basis, methods of detection, and adverse effects. Mol Cell Endocrinol 2018; 464:65-74
  244. Thomas A, Thevis M. Recent advances in the determination of insulins from biological fluids. Adv Clin Chem 2019; 93:115-167
  245. Smith RG. Development of growth hormone secretagogues. Endocr Rev 2005; 26:346-360
  246. Hersch EC, Merriam GR. Growth hormone (GH)-releasing hormone and GH secretagogues in normal aging: Fountain of Youth or Pool of Tantalus? Clin Interv Aging 2008; 3:121-129
  247. Thomas A, Walpurgis K, Krug O, Schanzer W, Thevis M. Determination of prohibited, small peptides in urine for sports drug testing by means of nano-liquid chromatography/benchtop quadrupole orbitrap tandem-mass spectrometry. J Chromatogr A 2012; 1259:251-257
  248. Thomas A, Delahaut P, Krug O, Schanzer W, Thevis M. Metabolism of growth hormone releasing peptides. Anal Chem 2012; 84:10252-10259
  249. Thevis M, Kuuranne T, Geyer H. Annual banned-substance review: Analytical approaches in human sports drug testing. Drug Test Anal 2019; 11:8-26

Laboratory Assessment of Testicular Function

ABSTRACT

 

Since the symptoms of hypogonadism are nonspecific, and the signs of testosterone deficiency can be subtle and slow to develop, the assessment of testicular function relies heavily on laboratory testing. The laboratory diagnosis of hypogonadism is based on a consistent and unequivocally low serum total testosterone level measured in blood samples obtained in the early morning, but normal ranges vary with different methods and among laboratories. Moreover, many men who present with adult onset testosterone deficiency have a low level of sex hormone-binding globulin (SHBG) associated with obesity, insulin resistance, and type 2 diabetes. In these men, tests for the free (or non-SHBG/bioavailable) testosterone fraction testosterone) are helpful for an accurate diagnosis. If testosterone deficiency is confirmed, the next step is to differentiate between primary and secondary hypogonadism by measuring LH and FSH.  With many disorders, however, both the testes and the hypothalamic-pituitary unit are affected. Other tests such as estradiol, inhibin-B, and Mullerian inhibitory hormone, and provocative endocrine tests using hCG, GnRH or its analogs, or antiandrogens or antiestrogens, and semen analysis in the subfertile male are discussed.   

 

INTRODUCTION

 

The evaluation of men for suspected hypogonadism begins with a detailed medical history and a careful physical examination. Laboratory tests are an essential component of almost all evaluations, and proper interpretation of the results obtained requires an understanding of methodology as well as an awareness of the impact of endocrine rhythms, age, race, body composition, drug exposure, co-morbidities, other illnesses, and nutrition on tests of endocrine testicular function. Endocrinologists rely on clinical laboratories to provide accurate and precise, and in some cases highly sensitive assays for accurate diagnoses. We expect the reference ranges to be based on large normal control populations, and for some assays, normative data must be stratified by age. Unfortunately, these expectations are not always fulfilled. Endocrinologists should examine the protocols for each assay, and discuss them with the laboratory director. In recognition of these potential shortcomings, the U.S. Endocrine Society has spearheaded efforts towards standardizing the methods by which testosterone assays are validated (1), and several societies have provided guidelines and recommendations to physicians who order and interpret the results of androgen assays (2-4). The College of American Pathologists and the U.S. Centers for Disease Control havevoluntary Hormone Standardization Programs to help clinical, research, and public health laboratories maintain and enhance the quality and comparability of their results.

 

TESTOSTERONE

 

Testosterone, the major androgen in men, is necessary for fetal male sexual differentiation, pubertal development, and the maintenance of adult secondary sex characteristics and spermatogenesis. Testosterone regulates gene expression in most extra-genital tissues, including muscle and bone, and modulates the immune system. The testes are the source of more than 95% of the circulating testosterone in men although the adrenal cortex produces large amounts of the testosterone precursor steroids, dehydroepiandrosterone (DHEA) and androstenedione.

 

Following birth, there is a period of activation of the gonadotropin-releasing hormone (GnRH) pulse generator that stimulates testosterone secretion to peak levels of 200-300 ng/dl (7-10.5 nmol/L) at ages 1-2 months and lasts until age 4-6 months, and is often called mini-puberty (5). Thereafter, GnRH secretion declines, causing a fall in testosterone to very low levels until puberty begins.  LH and testosterone testing at mini-puberty provides an opportunity to confirm the diagnosis of hypogonadotropic hypogonadism (6).

 

In early puberty, LH and testosterone secretion increase dramatically during sleep which gradually transitions to sustained secretion throughout the day and night (7). The nocturnal rise in testosterone levels in early puberty can be used clinically to evaluate boys with delayed puberty because it may precede pubertal testis growth, and indicates that puberty has begun.  Figure 1 illustrates age-specific median values and ranges for testosterone in morning blood samples from 138 boys (8).  

Figure 1. Serum testosterone levels (median and range) by LC-MS in leftover samples from blood checks before minor surgery or for the exclusion of endocrine diseases. Data from Kulle et al (8). To convert total testosterone levels to nmol/L, multiply by 0.0347

Hypothalamic neurons that express kisspeptin, neurokinin-B, and dynorphin, are thought to activate and up-regulate GnRH and initiate pubertal development (9).  Adult testosterone levels are usually achieved by age 16 years, and generally range from 300-1000 ng/dL (10-35 nmol/L). 

 

The secretion of testosterone is periodic with oscillations occurring over hours, days, and months. Frequent sampling of peripheral blood in adult men reveals small frequent moment-to-moment fluctuations (10) whereas spermatic venous blood sampling reveals robust episodes of testosterone secretion occurring about once per hour (11).  Presumably because of this rapid pulse frequency and a plasma half-life of 60-90 min, only small testosterone fluctuations are generally observed in peripheral blood. Therefore. a single blood sample is usually an adequate assessment of testosterone production on a given day.  There is a substantial diurnal variation in testosterone levels in adult men, however, with highest levels in the early morning, followed by a progressive decline throughout the day, reaching lowest levels in the evening (Figure 2). Thus, the time of day of blood sampling is an important consideration, and a blood sample drawn in the morning between 0800 and 1000h is recommended because reference ranges are generally based on morning values. Nadir evening levels  in young men are generally 15-20% lower than morning values although the differences can be as great as 50% (12). The diurnal testosterone rhythm is blunted as men grow older (13) as well as in young men with primary testicular failure. On the other hand, the diurnal variation in testosterone is exaggerated in hypogonadal men with hyperprolactinemia (14), much like normal adolescents. There may be a seasonable variation in testosterone as well (15).

Figure 2. 24-hour testosterone concentration profiles in eugonadal men (n=5), untreated hypogonadal men with PRL-producing pituitary adenomas, and men with testicular failure (n=5). Adapted from Winters, SJ (14). To convert total testosterone levels to nmol/L, multiply by 0.0347.

There is also a slight decline in testosterone levels following a meal (16), so that some authors suggest obtaining a fasting level.  In general, the results of one abnormal test should be confirmed. In one study in which multiple blood samples were taken from middle-aged men over one year, there was a relatively good correlation (r=0.85) between the plasma testosterone level in the first sample and the mean value of seven subsequent samples; however, as many as 15% of men were misclassified, with either low or normal values that were not reproducible (17).

 

Testosterone deficiency can be an obvious clinical diagnosis, and laboratory tests are often merely confirmatory; however, the diagnosis of hypogonadism is sometimes less straightforward. Experts and international guidelines state that the diagnosis of hypogonadism requires the presence of symptoms of hypogonadism as well as a low serum testosterone level. Symptoms of hypogonadism lack specificity, however, and are subjective. Thus, the diagnosis by laboratory testing may be more objective and less controversial. On the other hand, as discussed below, laboratory test results may be inconsistent and suffer from methodological flaws.  While men with low testosterone levels tend to have symptoms of hypogonadism (18), many studies have shown that symptoms are often unrelated to testosterone levels, and many men with low testosterone levels do not complain of symptoms (19, 20).

 

The level of total testosterone in serum is the best single test to screen for hypogonadism because methodology has been optimized and normative data are widely available.  While early assays extracted plasma steroids into organic solvents and separated them by column chromatography, those research methods generated radioactive waste and are too costly for clinical purposes.  Currently most hospital laboratories use automated platforms with either a competitive or proportional two-site format. These platforms use small sample volumes to avoid matrix effects, and a monoclonal antibody with a chemi-luminescent label for detection. The accuracy of the result is highly dependent on the affinity and specificity of the antibody used, and the survey result summarized in Figure 3 demonstrates that platform immunoassays continue to provide variable results with a coefficient of variation (SD/mean) among methods of 12% at a potency of 256 ng/dL which is close to the lower limit of the usual reference range for normal men. Nevertheless, when the manufacturer provides a reference range which is based on a large number of samples, and results are age-adjusted, automated assays are probably sufficient for most clinical purposes in adult men. Assay modifications have reduced the impact of hemolysis, icterus, and lipemia on measured results.

Figure 3. Total testosterone levels (±SD) in a proficiency sample (Y-04) distributed by the College of American Pathologists, and assayed using 14 different instruments in 1556 participating laboratories in 2018. The mean (±SD) value was 256 ± 51 ng/dL. The most frequent method (#6) was used by 353 laboratories. Method #4 is mass spectrometry, and was 7% lower than the mean of all methods. To convert total testosterone levels to nmol/L multiply by 0.0347

There is a positive bias at low values in most immunoassays so that automated assays are not recommended for clinical purposes in women and children. Reference laboratories are increasingly employing liquid chromatography (LC) - tandem mass spectrometry (MS) methods (21, 22). This approach combines the resolution of chromatography with the specificity of MS, and is now viewed as a gold standard.  Larger aliquots of serum are extracted with organic solvents, steroid hormones are separated by chromatography, in some assays following derivatization, and values are determined by peak area integration of testosterone-containing fractions of the column eluate. Accuracy is gained by adding an internal standard as a stable isotope of the same compound being measured in order to correct for procedural losses.  LC-MS accurately detects testosterone concentrations as low as 1.0 ng/dL (23, 24). In the 2018 CAP survey (Figure 3), the coefficient of variation for the high range sample (419 ng/dL) was 10%, and was a very respectable 11% for both the mid- range (245 ng/dL) and low-range (72 ng/dL) samples among those laboratories (n=35) that used LC-MS methodology. The equipment needed is costly, however, and each LC-MS assay requires substantial analytical development and optimization, as well as highly trained personnel.

 

While one study of men ages 40-80 who self-reported good or excellent health found no age-related change in total testosterone levels (25),  most cross-sectional and longitudinal studies have found that testosterone levels peak in the third decade, and decline as men grow older (e.g. (26, 27). Therefore, most reference laboratories present results for testosterone as a function of age. Small differences based on race and ethnicity have been found (28). One reference range for total testosterone levels in normal volunteers assayed by high-turbulence flow liquid chromatography(HTLC)-MS (22) is shown in Table 1. The decline in total testosterone in this study was roughly 0.4% per year beyond age 20, and mean T levels were 31% lower in men older than age 80 compared to men in their 20’s

 

Table 1. Impact of Age on Total Testosterone by HPLC–MS/MS in Normal Men  

 Age (years)

20-29

30-39

40-49

50-59

60-69

70-79

>80

Mean (ng/dl)

590

546

573

534

559

417

404

SD

232

206

209

194

226

177

234

 95th percentile

1052

910

901

909

928

755

716

5th percentile

283

319

310

296

290

168

92

N

61

76

55

51

19

44

19

Results from Quest Diagnostics, San Juan Capistrano, CA, courtesy of M.P. Caulfield. To convert total testosterone levels to nmol/L multiply by 0.0347

 

Table 2 shows a second reference range with combined values from the Framingham Heart Study analyzed by LC-MS, and the European Male Aging Study and Osteoporotic Fractures in Men Study analyzed by GS-MS (29). Mean levels for men in their 80s were 20% lower than for men in their 20s representing a similar decrease of 0.33% per year. Overall values are, however, 20-40% higher than from the reference laboratory in Table 1. Thus age-dependent reference ranges are method-specific, and no generalizable “cut-point” for the diagnosis of testosterone deficiency seems possible.

 

Table 2. Impact of Age on Total Testosterone Levels Measured by LC-MS or GC-MS) from Bhasin et al (29)

Age (years)

20-29

30-39

40-49

50-59

60-69

70-79

80+

Mean (ng/dl)

713

656

617

611

569

567

570

95thpercentile

1104

1084

1001

1059

965

1097

1079

5th percentile

375

343

304

286

276

254

238

N

220

660

872

788

493

289

26

 To convert total testosterone levels to nmol/L multiply by 0.0347

 

SEX HORMONE BINDING GLOBULIN (SHBG)

 

Sex hormone-binding globulin (SHBG) is a glycoprotein of molecular weight 90-100,000 KDa that is produced by the liver. SHBG binds testosterone and other steroids, prolongs their metabolic clearance, and regulates their access to target tissues (24). SHBG had been measured indirectly by radio-ligand binding assays, but two site immuno-radiometric and enzyme-linked assay kits are now widely available, and automated versions have been developed. There is little effect of meals or time-of-day on SHBG

Figure 4. SHBG levels from birth to old age in males and females. Redrawn from Elmlinger et al (30).

A cross-sectional analysis of the impact of age on SHBG levels in males and females is shown in Figure 4. There is an increase following birth to peak values at ages 4-5 years followed by a decline that is greater in males than females (30). SHBG levels in adult male plasma range from 20-100 nmol/L, and rise slightly in old age (28); analysis by race revealed higher levels among younger AA males (31).

 

In normal men, SHBG binds 40-60% of the circulating testosterone with high affinity, and the level of SHBG is a major determinant of the total testosterone level (Figure 5). Single nucleotide polymorphisms in the SHBG gene influence the affinity of SHBG for testosterone and affect the level in plasma (32).

 

Figure 5. Levels of SHBG and total testosterone in 28 normal men whose BMI ranged from 23-40 kg/m2. From Winters SJ et al (33).

Various clinical conditions are associated with reduced or increased SHBG levels, and thereby tend to lower or increase total testosterone concentrations (Table 3).  High SHBG levels in men with HepC or hyperthyroidism may lead to very high total testosterone levels, and can be a diagnostic dilemma. Serum SHBG levels are low in obesity (34), in T2DM or the Metabolic Syndrome (MetS) (35). In fact, low SHBG is frequently used as a biomarker in epidemiology studies to predict the development of the MetS as well as type 2 diabetes mellitus (36).  SHBG levels rise with weight loss (37) and when insulin resistance improves even without weight loss, e.g. with exercise. The molecular mechanism linking low levels of SHBG with insulin resistance and obesity is partly through hyperinsulinemia (38, 39) and through liver fat and cytokines which together regulate the transcription factor HNF4 alpha, a proximate activator of the SHBG promoter (40, 41) and at least 50% of genes expressed in liver.

 

Table 3. Factors that Influence the Level of SHBG in Plasma

Increase

Decrease

hyperthyroidism

hypothyroidism

hepatitis C

insulin resistance/NAFLD

GH deficiency

growth hormone excess

alcoholic cirrhosis

glucocorticoids

acute intermittent porphyria

androgens

first generation anticonvulsants

progestins

estrogens, certain SERMS

nephrotic syndrome

mitotane

genetic polymorphisms

thinness

obesity

genetic polymorphisms

 

 

FREE AND NON-SHBG (BIOAVAILABLE) TESTOSTERONE

 

The free hormone hypothesis proposes that the biological activity of a hormone is engendered by its unbound (free) but not its high affinity protein-bound concentration in plasma. The hypothesis remains controversial, and was recently reviewed for testosterone (42).

 

While some authors propose that there is little extra benefit in measuring free testosterone, my view is that the total testosterone level is sometimes inadequate to determine whether testosterone deficiency or excess is present. This occurs with a borderline value, or when the clinical findings and the total testosterone concentration do not agree. I encounter this conundrum most often in obese men, those with type 2 diabetes, NAFLD, or the Metabolic Syndrome among whom total testosterone levels are generally low, and who present because they have symptoms consistent with hypogonadism. In the Boston Area Community Health (BACH) Survey, 57% of men younger than age 50 with low total testosterone levels reported “normal” sexual function compared to 33% of those with low free testosterone levels (19). Likewise, the European Male Aging Study found that men age 40-79 with low free but normal total testosterone levels more often had symptoms and signs of sexual dysfunction while men with low total but normal free testosterone concentrations were more obese but had physical or sexual symptom scores that were similar to men with normal values (20). Thus, free testosterone seemed to be more closely aligned with the symptoms and signs of adult hypogonadism than was the total testosterone level.

 

In older men, on the other hand, the total testosterone level may be within the normal range while free testosterones decline more dramatically. The total testosterone level may also be elevated, as in men with hyperthyroidism, hepatitis-C, and patients treated with first generation anticonvulsants. Each of these situations occurs primarily because the primary condition is associated with a decrease or increase in the circulating SHBG level. 

 

Therefore, an assessment of free or non-SHBG bound testosterone may assist in the accurate diagnosis of androgen deficiency. This approach stems from the hypothesis that the bioactivity of circulating testosterone is due to the small percentage (1-4%) of total testosterone that circulates unbound (free testosterone) as well as the 40-50% of testosterone that circulates loosely bound to albumin and is often designated as “bioavailable-testosterone” (non-SHBG testosterone). The dissociation of albumin-bound testosterone is very rapid, and a short dissociation time is thought to allow the albumin-bound fraction to be available for uptake by cells (43). However, there is considerable debate as to which method for estimating free testosterone most accurately reflects testosterone production (42, 44).

 

Equilibrium dialysis is the benchmark reference method for measuring free testosterone. With the two-step approach, the free testosterone concentration is calculated from the product of the total testosterone level and the percentage that is determined to be free. H3-testosterone is added to the serum sample, and is allowed to partition between two compartments, one containing the tracer and serum sample, and the second containing buffer or an albumin solution. The compartments are separated by a semi-permeable membrane with a low-molecular weight cut-off. The protein-bound testosterone is retained while unbound testosterone, including H3-testosterone, crosses the membrane. It may take several hours to reach equilibrium, and using centrifugal ultrafiltration accelerates dialysis by forcing the unbound steroid thorough the membrane. Overall, the result is dependent on the accuracy of the total testosterone assay, and there is potential error due to temperature effects, sample dilution, and tracer impurities, among other problems. This assay is limited to reference laboratories.

Liquid chromatograph tandem mass spectrometry (LC-MS) can reliably measure hormone concentrations as low 1 pg/mL, and has been adapted for the measurement of free testosterone.  With this approach, equilibrium dialysis of undiluted serum is performed against buffer overnight at 37C, or more rapidly using ultracentrifugation cartridges, and (free) testosterone  is measured directly in the low molecular weight dialysate by LC-MS (45). Many reference laboratories have embraced this approach but it remains too complex and costly for most hospital laboratories.  

 

A published reference range using the two-step equilibrium dialysis method (Table 4) reveals that free testosterone levels decline as men grow older with a difference of 36% between the youngest and oldest reference groups. Thus, the percentage change in free testosterone by this method with age (0.6%/year) exceeds the rate of decline in total testosterone (0.3-0.4%/year).

 

Table 4. Reference for Free Testosterone by Equilibrium Dialysis in Normal Men

 Age (years)

20-29

30-39

40-49

50-59

60-69

70-79

>80

Mean (pg/ml)

102

86

74

84

84

80

65

SD

29

33

22

30

22

27

26

95.00%

148

144

114

136

111

131

101

5.00%

57

45

37

35

55

47

34

N

49

55

48

36

14

36

17

Results from Quest Diagnostics, San Juan Capistrano, CA, courtesy of M.P. Caulfield. 3H-testosterone was added to a sample diluted 1:5 in assay buffer and incubated in a dialysis chamber for 20 h at 37 °C to allow the tracer to reach equilibrium with the endogenous testosterone and binding proteins. To convert total testosterone levels to nmol/L multiply by 0.0347.

 

Calculated Free Testosterone

 

A technically simpler approach used by many hospital laboratories is to calculate the free testosterone level from the levels of total testosterone and SHBG, using experimentally determined testosterone binding constants. The KD for testosterone for SHBG is most often defined as 1 x 109 L/M, and for albumin 3 x 104 L/M.  These are estimates, however, and different constants will produce different results (46). Differences in the level of albumin in the sample have little impact on the calculated free testosterone, and are usually ignored. Free and non-SHBG bound testosterone levels can be computed using an internet program (www.issam.ch/freetesto.htm). There is an excellent correlation between the level of free testosterone obtained by equilibrium dialysis and the calculated free testosterone level (47), and between the calculated free testosterone and non-SHBG testosterone levels. Calculated free testosterone concentrations vary with the binding constants and algorithms employed, however, and SNPs of the SHBG gene may influence the affinity of SHBG for testosterone (32), and thereby the accuracy of the calculated free testosterone level. It has also been suggested that ligand binding to the SHBG dimer is allosteric such that occupancy of one site by a ligand alters the affinity of the second site. Furthermore, because the result is calculated from the levels of testosterone and SHBG, measurement error for either assay impacts directly on the calculated free and non-SHBG testosterone levels. A reference range (21) for free testosterone based on calculation revealed a decline of 61% from young to old adult men (Table 5), representing a rate of decline of 1%/yr.  In a second study (48) the difference between young and old was only 26%, but again far exceeded the fall in total testosterone with aging which was 1.2%.

 

Table 5. Influence of Age on Free Testosterone Levels in Community Men

Age (years)

20-29

30-39

 

40-49

 

50-59

 

60-69

 

70-79

 

>80

 

Mean (pg/ml)

148

132

116

99

82

72

58

5th Percentile

79

70

61

50

45

33

29

95thPercentile

229

212

199

164

135

132

93

n

220

660

872

788

493

289

26

Total testosterone was measured in community dwelling men in Framingham, MA using LC-MS.  SHBG was measured using a two-site immunofluorometric assay from DELFIA-Wallac, Inc. Free testosterone was calculated by the law-of-mass-action equation using of K SHBG-T of 0.998 × 109 L/mol and a K Alb-T of 3.57 × 104 L/mol. Data from Bhasin et al (29). To convert total testosterone levels to nmol/L multiply by 0.0347.

 

Bioavailable Testosterone

 

Non-SHBG-testosterone is called "bio-available” (BAT) because adding SHBG to an androgen-containing sample reduces its androgen receptor binding activity (49). This approach assumes that the testosterone bound with low affinity to albumin is active. The non-SHBG-testosterone (bioavailable) level can be determined by adding a tracer amount of 3H-testosterone to the serum sample, and selectively precipitating the SHBG-bound 3H-testosterone by adding 50% ammonium sulfate or concanavalin-A Sepharose. The 3H-testosterone that remains in the supernatant is presumed to be either free or albumin-bound, and is counted. The percentage of 3H-counts added that is in the supernatant is multiplied by the total testosterone level in order to determine the non-SHBG (bioavailable) testosterone. The assay is not readily automated, requires purified 3H-testosterone, and the complete separation of SHBG from albumin is presumed, but not verified.

 

Free Testosterone Index

 

The free testosterone index (FAI, free androgen index) represents the ratio: total testosterone/SHBG (both in units of nmol/L).  This value is easy to calculate, and may be valid in serum samples from women but is not valid in men (50)because most of the SHBG in men is bound to testosterone. Like the calculated free testosterone, and BAT, the FAI is dependent on accurate values for testosterone and SHBG

 

Direct Free Testosterone

 

The direct free testosterone assay was developed as a single-step, non-extraction method in which an125I-labeled testosterone analog competes with unbound testosterone in plasma for binding to a testosterone-specific antiserum that has been immobilized on a polypropylene assay tube. The basis for the test is that the analog has a low affinity for SHBG and for albumin.   Values for normal men with this method, as a percentage of the total testosterone (0.2-0.64%), are substantially lower than the 1.0-4.0% determined by other methods. While this difference alone does not cause a problem if adequate reference ranges are available, it immediately prompted speculation concerning the accuracy of the method.  Subsequent studies revealed that analog free testosterone assay results are positively correlated with the level of SHBG, much like total testosterone (33), and that free testosterone is un-measurable by analog assays in a dialysate of normal adult male serum (51).  Thus, the free testosterone level determined with analog assays appears to provide essentially the same information as the total testosterone level, is often misleading (tends to over diagnose hypogonadism), and is not recommended. The 2018 College of American Pathologists survey revealed that only 7% of participating laboratories continue to use this method, and it is has nearly disappeared from research articles related to testosterone.

 

Testosterone in Saliva   

 

The level of testosterone in saliva is positively correlated with the plasma free testosterone concentration. Salivary samples are easily collected, usually by a non-stimulated drool.  Both extraction and non-extraction immunoassay methods are available although LC-MS is being increasingly employed (44).   Usual values in adult men are 150-500 pmol/L (40-145 pg/ml).   Salivary testosterone assays are a useful research tool for field studies and other settings in which blood sampling is impossible or impractical.  While androgens are stable for a few days in untreated saliva, method artifacts may occur, and careful assay validation and quality control are essential.  Salivary testosterone assays have not been recommended for clinical purposes.

 

Cell-Based Reporter Bioassays

 

Cell-based reporter bioassays have been developed to analyze androgen bioactivity in biological samples. A stable cell line is created by transfection with plasmids encoding the human androgen receptor and a reporter system containing an androgen-responsive gene such as the mouse mammary tumor virus (MMTV)-luciferase reporter. When cells are stimulated with androgens, luciferase activity is increased dose-dependently (52), These assays remain investigational.

 

DIHYDROTESTOSTERONE

 

5 alpha reduction of testosterone to the more potent androgen dihydrotestosterone (DHT) is essential for fetal male genital development. T is converted to DHT by at least two steroid 5α-reductase (5AR) isoenzymes, 5AR types 1 and 2.   5AR-1 is found in liver, skin, brain, ovary, prostate, and testis whereas 5AR-2 is expressed in prostate, seminal vesicle, epididymis and skin. Approximately 20% of the circulating DHT in men is secreted by the testes, and the remainder is derived from the bioconversion of testosterone in tissues.  Because of a high level of expression of 5AR-2 in prostate, testosterone is effectively converted to DHT in that tissue, in which the level of DHT exceeds the peripheral blood concentration by 5-10-fold. The concentration of DHT in adult male serum is only about 10% of the value for testosterone, however. Therefore, an assay with negligible cross-reactivity with testosterone is needed for an accurate result.  An LC-MS method is now used by most reference laboratories. With gradient elution, the separation is totally complete (53).  Patients with 5α-reductase deficiency type 2 have ambiguous genitalia, and are generally detected as neonates, although a few patients have only microphallus or cryptorchidism.  A rise in testosterone but not DHT following hCG stimulation, producing a ratio of more than 10:1, is characteristic in most of these patients (54).  However, DHT production by 5AR1 can reduce the reliability of the ratio for diagnosis (55). The diagnosis is most often made by urinary steroid profiling by GC-MS, together with mutational analysis. DHT levels are often measured in epidemiological studies and in clinical research on prostate cancer and its treatments, and testosterone treatment results in a dose-dependent increase in serum DHT concentrations (56). Yet DHT levels are probably not useful in most clinical situations.

 

ESTRADIOL AND ESTRONE

 

Estrogens are important male hormones. They regulate the hypothalamic-pituitary-testicular axis, influence the function of  the testes and prostate, increase growth hormone and IGF-1 secretion, modify lipid metabolism and other hepatic proteins, and play an important role in male skeletal health, body fat, and perhaps sexual functioning  (57, 58).  Serum levels of estradiol and estrone are often measured in men with gynecomastia (59) or with unexplained gonadotropin deficiency. These conditions are rarely, but occasionally, due to estrogen-producing tumors, or to acquired (60) or genetic (61)   abnormalities in which estrogen production is increased. Moreover, the accurate measurement of estradiol may be helpful when SERMs, aromatase inhibitors, or hCG are used are used to increase testosterone levels, and in research on the role of estrogens in males (58, 62).  Estradiol is produced from testosterone, and estrone is produced from androstenedione, by aromatase P450, the product of the CYP19 gene. This enzyme is expressed in Leydig cells and in the adrenal cortex, as well as in adipose- and skin-stromal cells, aortic smooth muscle cells, kidney, skeletal muscle cells, and the brain. The promoter sequences of the P450 aromatase genes are tissue-specific, but the translated protein appears to be the same in all tissues. Increased aromatase expression in adipose and skin stroma with obesity is the most common cause for mild estrogen excess in men.  Interestingly, most studies of the age-associated decline in testosterone levels do not find a parallel fall in plasma estradiol levels perhaps because of increasing aromatase activity and fat mass as men grow older (63).

 

Because of low levels in males, traditional immunoassays for estrogens employed large volumes of plasma (2-5 ml) that were extracted with organic solvents. Because those assays are time-consuming and expensive to perform, non-radioactive automated methods that were optimized for the higher values normally found in pre-menopausal women were employed. However, the very small sample volumes used in automated assays may produce unexpectedly high values.  Results for estradiol in male plasma determined in platform assays are unreliable. The lack of agreement between laboratories is shown in Figure 6 which depicts the results for the low potency proficiency sample (mean 199 pg/mL) distributed in 2018 by the College of American Pathologists to 1450 participating laboratories. While many assays produced similar results, overall values ranged from 151 to 415 pg/ml (554-1523 pmol/L), with a coefficient of variation of 33%, compared to the goal of ±12.5%.  The mean value of the sample far exceeds normal levels in males, however, and testing at a lower potency would no doubt have produced even more dramatic differences.

Figure 6. Estradiol levels (±SD) in a mid-range proficiency sample (Y-05) distributed by the College of American Pathologists assayed using 15 different instruments in 1499 participating laboratories in 2018. The most frequently used (#6) was used by 341 laboratories. Mass spectrometry assays (n=11) are represented in column 4.

Mass spectrometry assays for estradiol and estrone, with a limit of detection of 1 pg/ml, have replaced conventional radio-immunoassays in most reference laboratories (24), and are highly recommended. The result with LC-MS is lower than with many immunoassays which presumably also detect interfering substances. Moreover, the between laboratory coefficient of variation for laboratories using MS methods (column 4) was also acceptably low at 10.3%.

 

Most of the circulating estradiol in men is loosely bound to albumin or is unbound (64), and only about 20% is thought to be bound to SHBG. Therefore, the serum level of SHBG was not predicted to appreciably influence the actions of estradiol. Measurement of free estradiol may be useful, however (65). In a series of studies, calculated non-SHBG-bound estradiol levels correlated more strongly with low bone mineral density and with indexes of high bone turnover in older men than did levels of total estradiol (66). On the other hand, increased mortality over 12 years of follow-up was seen among those with either low or high total as well as calculated free estradiol levels at baseline when compared to those in the middle tertile (67).  Akin to testosterone, non-SHBG (bioavailable) estradiol levels can be determined by ammonium sulfate precipitation using 3H-estradiol, or by equilibrium dialysis with or without using LC-MS.  Estimated levels are < 1 pg/mL. Calculated values require an accurate total estradiol assay, affinity constants are less certain than for testosterone, and the influence of testosterone and other steroids that also bind SHBG on the calculated estradiol level is no doubt important.  Age-specific free estradiol levels were reported but total estradiol was measured using an electro-chemiluminescence immunoassay (68); results using LC-MS methods are needed.  

 

GONADOTROPINS

 

FSH and LH, together with TSH and hCG, form a closely related family of heterodimeric glycoprotein hormones. Each consists of a common α-subunit that is non-covalently linked to a specific β-subunit. The α–β dimer is held together by a ‘seatbelt’ structure formed by the C-terminal amino acids of the β-subunit wrapped around the α-subunit.  Both subunits have asparagine-linked carbohydrate chains (2 for human α-subunit, 2 for FSH-β, and 1 for LH-β). The oligosaccharides project from the peptide skeleton, and by shielding of epitopes and altering the tertiary structure of the hormone, the sugars may impact receptor activation and bioactivity as well as antibody binding. Glycosylation also prolongs hormone clearance.

 

Most laboratories utilize fully automated, commercial assay systems for peptide hormones including LH and FSH.  Detection is generally by chemiluminescence, avoiding the use of radioactive tracers. Many assays achieve specificity by utilizing a biotinylated monoclonal antibody to the α-subunit as a capture antibody.  A second monoclonal antibody to the β-subunit is labeled with an organic ester that produces chemiluminescence in the presence of hydrogen peroxide (indicator antibody). Various pituitary and recombinant preparations are used for the standard curve but most assays are calibrated in terms of IU/L of International Reference Preparations (IRP) of highly purified human LH and FSH. The various standards have differing sugar sequences and branch patterns producing some variation in results between laboratories and assays, however, differences are relatively small.  When defined as a level of precision of replicate determinations of <10%, the sensitivity of these assays approximates 0.1 U/L.

Figure 7. Serum LH and FSH levels in normal males measured by an ICMA assay. Redrawn from Resende et al. (69).

Figure 7 shows mean levels of LH and FSH across the stages of pubertal development in boys measured with an automated ICMA (69). Values with an immunofluometric assay were slightly higher. FSH levels tend to exceed LH before puberty in boys, and both gonadotropins rise progressively during puberty with substantial overlap among the various pubertal stages.  

Prepubertal children have low amplitude but discrete pulsatile patterns of LH secretion that are amplified during sleep as puberty begins (70).  In adult men, LH is released in robust pulses every 1-2 h throughout the day and night with within subject variation in pulse height and between-pulse interval (Figure 8). In part because of pulsatility, the normal range for LH is wide, with typical ranges for adult men in terms of 2nd IRP of 1.6–8.0 IU/L for LH and 1.3–8.4 IU/L for FSH. Because of this pulsatility, pooling of 3 samples taken 20–30 min apart may provide a more accurate estimate of a person’s LH value than does a single sample.

 

Figure 8. Pulsatile patterns of LH secretion. Blood samples were drawn every 10 min for 12h starting at 0800 h from a 32-year-old normal man whose testosterone level was 474 ng/dL. Winters, SJ (unpublished).

Men with elevated levels of LH and FSH generally have testicular damage. Testes are usually small, there is oligo or azoospermia, and testosterone levels are low. Circulating estradiol levels are generally normal, however, in part because testicular aromatase is stimulated by the high level of LH. In some men with high LH and FSH levels, testosterone levels remain normal, sometimes because of elevated SHBG. The term “mild” or “compensated” hypogonadism is often applied (71). The diagnosis of gonadotroph adenoma is sometimes entertained, but long-standing infertility and small testes are a clue to the correct diagnosis. Much like the controversy surrounding thyroid hormone replacement in individuals with “subclinical” hypothyroidism, men with compensated hypogonadism who are not seeking fertility, mighty be viewed as candidates for testosterone replacement even though testosterone levels are within the reference range. This is often the case in teenagers with Klinefelter syndrome (72).

 

As men grow older, there is a decrease in sperm quality, the testes tend to be smaller, and there are fewer germ cells with more hyalinization and thickening of the tunica albuginea. Inhibin-B levels fall and FSH levels are often elevated. Mean LH levels also rise, but most often remain within the reference range (73).

 

Men treated with SERMs or aromatase inhibitors (74), and the rare men with inactivating mutations of the estrogen receptor-α (75) often have elevated LH and normal or elevated testosterone levels. These findings imply that estradiol is a major mediator of testicular negative feedback control of GnRH-LH secretion. Elevated LH and testosterone levels are also characteristic of men with androgen insensitivity syndromes (76) and men treated with anti-androgens (77), and reveal that there is additional negative feedback control of gonadotropins through the  androgen receptor.

 

FSH levels may be increased selectively because FSH production is regulated not only by GnRH and gonadal steroids but also by a paracrine control mechanism that involves pituitary activin and follistatin, and gonadal inhibin (see below).  When Sertoli cells fail to function normally, inhibin-B production declines, and the paracrine effects of pituitary activin stimulation of FSH-β gene expression is unopposed. Most men with high FSH levels have small testes and oligo- or azoospermia (78); however, FSH levels may sometimes be within the reference range (note that in some instances the reference range for FSH may be too high and some experts feel that the upper level for FSH is 8IU/L) in infertile men with severe damage to the germinal epithelium (79)  who may have either normal sized or small testes. In these cases, testicular biopsy is sometimes needed for a definitive diagnosis which is most often maturation arrest at the primary spermatocyte stage.

 

An elevated serum FSH level may rarely indicate an FSH-producing pituitary adenoma (80).  These tumors are generally macroadenomas that present with headache and a visual disturbance, and often co-secrete free α-subunit. The testes may occasionally  enlarge (81). The LH level is usually suppressed even when the tumors are LH-β immunoreactive, and testosterone levels are generally low.  LH secretion by a pituitary tumor is very rare (82, 83).  Most pituitary adenomas that are immunoreactive for LH or FSH are clinically silent with little or no hormone secretion.

 

The diagnosis of hypogonadotropic hypogonadism (HH) is based upon finding a low serum testosterone level with inappropriately low/normal LH and FSH concentrations. It is important to verify testosterone deficiency since a low SHBG level may result in the misdiagnosis of testosterone deficiency, and low LH may overlap with the reference range because of the pulsatile nature of LH secretion. Thus, the diagnosis of secondary hypogonadism (84) is sometimes problematic. The differential diagnosis of true hypogonadotropic hypogonadism is broad, and includes congenital HH due to a variety of genetic mutations including Kallmann syndrome (85), and acquired disorders such as prolactinoma, other sellar and suprasellar tumors or cysts, infiltrative diseases and vascular causes, iron overload, head trauma and others. HH in childhood limits pubertal development while in adults HH results in the regression of secondary sexual characteristics, albeit slowly.    

 

Isolated deficiency of FSH due to inactivating mutation in the FSH-β gene is a rare disorder (86-88). Affected men have been azoospermic, with borderline or low testosterone and increased LH levels. Pathological examination revealed narrowed seminiferous tubules, decreased Sertoli cell numbers, absence or aplasia of germ cells, and Leydig cell hyperplasia. Most mutations disrupt the ability of the mutant FSH- β to combine with the alpha-subunit. The low testosterone and increased LH levels may be explained by impaired signaling from the dysfunctional seminiferous tubules to Leydig cells (89). A few hypogonadal men with inactivating mutations of the LH-β gene have also been described (90-92).  Immunoreactive LH may be present or absent in serum depending on the nature of the mutation, but if present is bioinactive (92). Testosterone is low, and FSH is increased.  As placental hCG, rather than LH, stimulates male sexual differentiation, affected males have a normal male phenotype at birth while bioactive LH deficiency results in testosterone deficiency, and prevents normal puberty.

 

Two-site assays can occasionally be "too specific".  Polymorphisms that affect immunoassay detection by monoclonal antibodies may lead to misdiagnoses. For example, there is a relatively common polymorphism in the LH-β gene that is characterized by two point mutations in codons 8 and 15 resulting in two amino acid substitutions and an extra glycosylation site (88). Even though men and women with the LH variant appear to be normal and fertile, the serum LH level is low or undetectable using certain monoclonal antibodies whereas the result with other assays is normal. Clearly, when a man with a low or undetectable LH with a normal testosterone level is encountered, or a disparity between LH and FSH levels cannot be readily explained, a second assay method should be used.   

 

Various glycoforms of LH and FSH with structurally heterogeneous glycans are found in the pituitary and in the circulation, and glycosylation has been shown to influence hormone clearance and biological activity (93). Both sex steroids and activin have been reported to affect gonadotropin glycosylation. Highly sialylated glycoforms, with an acidic pH, tend to have a longer circulating half-life whereas the more alkaline forms tend to exhibit greater bioactivity in vitro. There is evidence that LH and FSH glycosylation is physiologically important. The ideal immunoassay would detect accurately only the total bioactive LH or FSH in the sample, but clearly this goal is difficult to accomplish. As an alternative approach, in vitro bioassays for LH and FSH can be used to assess the function of the gonadotropins. An in vitro bioassay based on the production of testosterone by cultured mouse or rat Leydig cells (94) was developed to assess LH function, and the production of estradiol by rat granulosa cells or immature Sertoli cells can be used to assay FSH bioactivity (95). Bioassays based on cAMP production by cell lines stably expressing gonadotropin receptors, with quantification using cAMP-responsive promoters linked to a luciferase reporter, have also been developed (34).  While useful for the study of the biological properties of recombinant or purified proteins, the clinical use of these assays is limited by the nonspecific effects of serum. In fact, many findings reported in patients using in vitro LH bioassays were found subsequently to be methodological artifacts.

 

Glycoprotein α-Subunit

 

Glycoprotein α-subunit is secreted in bursts that coincide with LH secretory episodes, implying pre-eminent regulation by GnRH (96) Accordingly, serum α-subunit levels increase at puberty (97) and are elevated in men with testicular failure and in postmenopausal women.  α-Subunit is also expressed in thyrotrophs, and levels are increased in patients with primary hypothyroidism (98).

Figure 9. Pulsatile patterns of α-subunit secretion. Blood samples were drawn every 10 min for 12h starting at 0800 h from a 32-year-old normal man whose testosterone level was 474 ng/dL. Winters, SJ (unpublished)

α-Subunit levels are low, but measurable, in normal children and in patients with congenital hypogonadotropic hypogonadism.  Because peak α-subunit levels after GnRH stimulation tend to be lower in IHH patients than in prepubertal boys, this test has been used to distinguish between these two patient groups, although some overlap occurs (99). α-Subunit is produced by as many as 20% of pituitary adenomas (100) often together with FSH or sometimes GH. Because it is cleared by renal excretion, α-subunit levels are high in patients with renal failure (101) .

 

INSULIN-LIKE FACTOR 3

 

Insulin-like factor-3 (INSL3) is a peptide hormone member of the relaxin-insulin hormone family that is secreted by Leydig cells following LH stimulation. INSL3 affects testicular descent through effects on the gubernaculum (102) and plays a role in spermatogonial differentiation. At the time of puberty, INSL3 levels rise in parallel with testosterone (103).  INSL3 concentrations in the blood of normal adult men are approximately 1 ng/mL (0.4-1.5 ng/mL).   Patients with Leydig cell dysfunction have lower levels, and the very low concentrations in men with hypogonadotropic hypogonadism increase following hCG administration (104) but are unaffected by testosterone treatment (105).

 

INHIBIN-B

 

Inhibin, a glycoprotein hormone produced by the testes as well as the ovaries, is responsible for the selective negative feedback control of FSH secretion, and functions as an intra-gonadal regulator (106). Most evidence supports the idea that inhibin decreases FSH-β mRNA levels by blocking pituitary activin-stimulated FSH-b transcription (107).  Inhibin is a 32-kDa heterodimer composed of an α-subunit, and one of two β subunits, βAor βB. Inhibin-B (α-βB) is the form produced by testicular Sertoli cells; inhibin-A (α-βA), produced by the corpus luteum and placenta, is undetectable in adult male plasma. Higher molecular weight forms of the uncombined inhibin α-subunit that lack bioactivity are released into the circulation in excess of dimeric inhibin-B.

 

The Gen II two site inhibin-B ELISA assay uses a capture antibody raised to a peptide from the βB-subunit, and a biotinylated detection antibody raised to a peptide from the inhibin α-subunit (108).  Wells are then incubated with a streptavidin-labeled horseradish peroxidase followed by the substrate tetramethylbenzidine which produces a colorometric signal that is proportional to the amount of inhibin-B in the sample. The assay is not automated and no performance statistics could be located.  Older assays were more complex, and added methionine and an oxidation step with hydrogen peroxide to facilitate capture, and heating with sodium dodecyl sulfate solution to enhance specificity.

 

Inhibin-B is produced by the fetal testis and is measurable in serum at term. Levels increase substantially in newborns coincident with the rise in gonadotropins and testosterone (mini-puberty), remain elevated for 2-4 months and then decline (109).  In contrast to barely detectable levels of gonadotropins and testosterone during childhood, however, circulating inhibin-B is readily measurable in sera from prepubertal boys, implying that production is partly gonadotropin-independent. Serum inhibin-B levels increase to adult values at the time of puberty, and decline as men grow older (110).  Serum inhibin levels are partly determined by gonadotropin stimulation (111) but based on studies in monkeys, also reflect Sertoli cell number (112). With the updated assay, results for normospermic men were (2.5-95thpercentile) 32-416 with a median value of 174 pg/mL (113).

Figure 10. Inhibin B levels in males in relation to age. Reprinted from Andersson A-M (114) with permission of the publisher (Elsevier).

Although there is no remarkable pulsatile fluctuation in circulating inhibin-B levels, there is a diurnal variation in adult men with the highest values in the morning and nadir values approximately 35% lower in the evening. This diurnal pattern parallels that of testosterone (115).

 

Inhibin has been extensively studied as a biomarker of spermatogenesis. In keeping with its function to suppress FSH production, circulating inhibin-B levels are inversely correlated with FSH levels in adult men, and are more strongly correlated inversely when values from men with primary testicular failure are included in the analysis (111).  Inhibin-B levels are higher in fertile than infertile men but there is substantial overlap between the groups (113, 116). There is also a demonstrable, albeit weak, positive correlation with sperm count (117), and with the germ cell score in testicular biopsy specimens among infertile men (118). Inhibin-B levels are low in men with testicular failure, and are very low in men with Klinefelter syndrome in whom seminiferous tubules are hyalinized and Sertoli cells are essentially absent (119). Low but measurable inhibin-B levels in men following chemotherapy (120) or testicular irradiation (121), some of whom develop germinal cell aplasia, suggest that germ cell factors regulate SC inhibin production. Low inhibin-B levels predict persistent azoospermia in men with testicular cancer who undergo orchidectomy, chemotherapy and irradiation (122). Serum inhibin levels changed little in normal men who participated in a male contraceptive clinical trial of testosterone together with a progestin, and developed azoospermia or severe oligospermia (123).

 

Inhibin-B levels have been studied as a biomarker in the male partners of infertile couples undergoing testicular spermextraction (TESE) as a predictor of success of intra-cytoplasmic sperm injection (ICSI). While mean inhibin B levels tend to be lower (and FSH higher) in men with no sperm found at TESE, there is no level of inhibin-B that reproducibly predicts either the presence or absence of spermatozoa in TESE samples, or successful in vitro fertilization (124). Thus, measurement of inhibin-B is not recommended in the decision making for fertility potential among azoospermic men undergoing TESE and ICSI. Further, very large doses of FSH are needed to increase circulating inhibin-B levels in men, so that an FSH stimulation test for inhibin-B is also not helpful in a clinical evaluation of hypospermatogenesis.

 

Inhibin-B levels are reduced in gonadotropin-deficient men (125).  In those with the complete form of congenital hypogonadotropic hypogonadism, values are lower than in men with partial gonadotropin deficiency who have some spontaneous pubertal development. This quantitative difference is partly due to a presumed larger mass of Sertoli cells (as reflected by larger testicular size) in the latter group of men. Low inhibin-B levels have been reported to differentiate boys with constitutional delay of puberty (CDP) from those with congenital hypogonadoptropic hypogonadism (CHH) with 80-100% sensitivity and specificity (126). While plasma inhibin B concentrations in CHH and CDP may overlap, in one study of boys age 14-18 years with delayed puberty, among those who were genital stage 1 (testis volume ≤ 3ml), inhibin-B levels <35 pg/ml predicted persistent hypogonadism (testosterone levels < 3 nmol/L), and presumably the diagnosis of CHH, after 2 years of follow-up (127).  Inhibin-B is undetectable in most boys with congenital anorchia as in castrates, and is therefore a useful test to help distinguish these patients from boys with intra-abdominal testes (128).

 

High inhibin-B levels have been reported in a few boys with FSH-producing pituitary tumors (129), in patients with Sertoli cell tumors (130) and in boys with McCune Albright syndrome with macro-orchidism and autonomous function of Sertoli cells (131).  Adrenal tumors often express the inhibin α-subunit, and there is one report of high serum dimeric inhibin level in an adult man with an adrenal neoplasm (132).

 

ANTI- MULLERIAN HORMONE

 

Anti-Mullerian hormone (AMH, also known as Mullerian inhibitory hormone) is a 140 KDa homodimeric member of the TGF-β family of growth and differentiation factors (133). It is produced by fetal Sertoli cells and causes regression of Mullerian structures during male sexual development (134). AMH production increases in response to FSH and is inhibited by androgens. AMH is readily detectable in the serum of prepubertal boys in concentrations of 10-70 ng/ml, and declines to levels of 2-5 ng/ml with entry into adolescence (135). AMH remains detectable at low values throughout adulthood (104) but how AMH functions in males beyond fetal life is uncertain. AMH levels are elevated in untreated men with congenital hypogonadotropic hypogonadism and decline following treatment with hCG (136). AMH is absent from the plasma of most prepubertal boys with congenital anorchia but is generally detectable in boys with bilateral cryptorchidism (134). Therefore, measuring AMH is useful in evaluating boys with non-palpable gonads. AMH levels are low in men with seminiferous tubular failure but overlap with normal in men with oligospermia, and do not predict testicular sperm retrieval for ICSI (137).  Levels tend be low in obesity (138),

Figure 11. Serum concentrations of AMH in healthy males from birth (cord blood) to age 69 years. From Aksglaede L et al (139). Red dots are values from men with bilateral anorchia. Republished with permission of The Endocrine Society.

FUNCTIONAL TESTS

 

hCG Stimulation

 

Human chorionic gonadotropin (hCG) can be used as a test agent to examine Leydig cell steroidogenesis in prepubertal boys who secrete little or no endogenous gonadotropins. A variety of protocols have been used. In one study, serum testosterone levels rose to greater than 300 ng/dL (10.5 nmol/L) in healthy prepubertal boys administered hCG 1500 IU intramuscularly every other day for seven doses. The effect of hCG to increase circulating levels of testosterone, precursor steroids and DHT can be used in conjunction with mutation analysis to help evaluate patients with a 46,XY karyotype who have ambiguous genitalia which may be due to androgen insensitivity, defects in testosterone biosynthesis, or 5α-reductase deficiency (140). When the basal and hCG-stimulated profile indicates accumulation of steroid precursors upstream of an enzymatic defect, sequencing can be performed to identify the disorder. A raised serum testosterone to DHT ratio would suggest the diagnosis of 5α-reductase deficiency type 2. However, DHT production by 5α-RD1 can reduce the reliability of the ratio for diagnosis (55).

 

Testosterone levels are generally unaffected by hCG in boys with congenital bilateral anorchia but increase in boys with bilateral intra-abdominal testes (141). Therefore, this test, together with measurement of inhibin-B and AMH, help establish which boys have intra-abdominal testes and should undergo laparoscopy, and orchidopexy or orchiectomy (128).

 

Adult men with primary testicular failure have elevated endogenous serum LH concentrations, and hCG will predictably increase serum testosterone levels less in these men than in eugonadal men. In gonadotropin-deficient men, the testosterone response to short-term administration of hCG is also blunted because Leydig cell steroidogenic enzymes are down-regulated. Overall, hCG testing nowadays provides little clinically useful information in either group of adult men.

 

Blockade of Steroid Hormone Biosynthesis and Action

 

Lowering circulating sex steroid levels with pharmacological inhibitors, or blocking steroid hormone action with receptor antagonists, can be used to assess the integrity of the hypothalamic-pituitary-testicular unit as a research tool, and clinically as a treatment for adult men with adult onset hypogonadism (142).

 

Ketoconazole, a competitive inhibitor of cytochrome P450 cholesterol side chain cleavage (P450scc) and C17/20 lyase in the biosynthetic pathway to testosterone is used in the treatment of endemic mycoses and off-label in other conditions including Cushing syndrome, but causes a dose-dependent reduction in circulating testosterone and estradiol levels. (143). Because ketoconazole also lowers cortisol production, glucocorticoids are co-administered with ketoconazole to prevent the symptoms of cortisol deficiency.

 

Estrogens produced by the testes, peripheral tissues, and the CNS play an important role in the physiological feedback regulation of gonadotropin secretion in men. Selective estrogen response modulators (SERMs) bind to estrogen receptors (ER-α and/or ER-β) and exert estrogen-like effects, or inhibit estrogen effects, in a tissue-specific manner. SERMs also have non-classical extra-nuclear (membrane) signaling mechanisms in certain cells. By blocking estradiol negative feedback, most SERMS, such as clomiphene (144) and tamoxifen, increase plasma LH and FSH levels. Similarly, aromatase inhibitors (145) such as anastrazole, reduce circulating estradiol levels, and increase LH in men.  Dosages of 25 mg/day of clomiphene produced a mean two-fold increase in serum testosterone levels in adult men (Helo 2015). (142)., and while higher doses are more effective, estradiol levels may increase, and gynecomastia and galactorrhea may occur (144).  Two-fold increments in LH were produced by anastrazole 10 mg daily within 3-4 days.  Both blocking estrogen negative feedback and decreasing estradiol production increased LH pulse frequency indicating an effect of estradiol on the GnRH pulse generator. A normal response implies functional integrity of GnRH-LH-testosterone pathways; however, responses among normal subjects are variable, and diagnostic tests using SERMS are not thought to be useful clinically.  So far, these agents are not FDA-approved for any use in men.

 

Nonsteroidal antiandrogens increase serum LH levels in men by blocking the androgen negative feedback effect on GnRH secretion (146) and can be used to test GnRH-LH integrity.

 

GnRH Test

 

GnRH is used as a research tool to examine the responsiveness of gonadotrophs to their physiological stimulus. In normal adult men, the intravenous administration of 100 µg of GnRH increases serum LH levels three- to- six-fold while serum FSH levels rise by about 50%. Generally, the total and incremental release of LH and FSH following GnRH administration is directly proportional to the basal hormone level, although exceptions do occur.

 

The GnRH test was introduced as a method to diagnose hypogonadism, and to distinguish hypogonadism due to hypothalamic from pituitary disorders. The LH and FSH response to GnRH is subnormal when gonadotrophs are damaged or destroyed by pituitary tumors or by other pathologies. However, the gonadotropin response to stimulation with GnRH is also diminished in patients with GnRH deficiency since GnRH up-regulates its receptor on gonadotrophs, as well as the expression level of each of the gonadotropin subunit genes and thereby LH and FSH production. Thus, the LH response to GnRH stimulation is attenuated in these patients even though the pituitary is essentially normal. Therefore, when evaluating gonadotropin deficient adult men, the GnRH test generally provides little information beyond that of the basal testosterone, LH and FSH levels, and is not recommended for clinical purposes.

 

GnRH analog (naferelin, leuprolide, buserelin, triptorelin) testing may help distinguish prepubertal boys with constitutional delay of puberty (CDP) from those with congenital hypogonadotropic hypogonadism (147-149). These agents bind the GnRH receptor with higher affinity and have a longer circulating half-life than does native GnRH. The LH and testosterone responses at 4h and 24h in boys with CDP generally exceed those of HH.  In one study, prepubertal boys age 13.7-17.5 years with testicular volume ≤ 4ml whose LH level at 4h following 0.1 mg sc triptorelin exceeded 5.3 U/L all progressed to testicular enlargement, testis size ≥ 8 ml, over 18 months. Those boys with lower LH basal ≤ 2U/L and stimulated ≤5.3 mIU/ml values did not progress to this level. The CHH group also had low levels of inhibin-B (<111 pg/ml) at baseline (127). Thus, the test seems to be of use in evaluating prepubertal boys with delayed puberty. In patients with CHH who are homozygotes or compound heterozygotes for mutations of the GnRH receptor gene, GnRH binding to its receptor may be either absent or reduced in affinity, or receptor signal transduction may be impaired, and the LH response to GnRH stimulation in these patients may be absent or reduced. The LH response in those with partial hypogonadotropic hypogonadism overlaps with the normal response (150), and while these tests are helpful so far there is no gold-standard diagnostic test to fully differentiate boys with CHH from those with CDP.

 

SEMEN ANALYSIS

 

Laboratory testing for men who present for an evaluation of infertility begins with the semen analysis (151). Most laboratories report the semen volume, the sperm density (million/mL), motility (% with progressive motion on the microscopic slide), and the sperm morphology (% with oval-shaped sperm heads), and follow WHO guidelines in their methods and reports (152). The test helps predict fertility potential but is not a test of fertility since there is substantial overlap between results for fertile and infertile men (153). Moreover, lack of complete adherence to standardized methods, and insufficiently skilled laboratory staff, may substantially limit the diagnostic value of the semen analysis. The sample can be collected by masturbation, or by interrupted intercourse, generally after 2-3 days of abstinence from ejaculation. Most of the sperm are in the first portion of the ejaculate so it is important to collect a complete sample. Condom collection is less desirable since condoms contain spermicides, but if so, the sample should be immediately placed into a sterile jar. It is important to analyze the sample within 60 min of collection, as motility decreases over time. In cold climates, the sample should be kept warm in transit by holding it within outer clothing adjacent to the body. Even with optimized collection methods, there is a physiological day-to-day variation in sperm production, so 2-3 samples may provide more insight that a single sample. A man can be considered to be sterile if there are no sperm in the ejaculate (azoospermia).

 

Table 6. WHO Guidelines for an Adequate Semen Analysis

Semen volume

 ≥1.4 mL

Sperm concentration

≥15 million/mL

Total sperm output

≥40 million sperm

Sperm progressive (total) motility

≥32 (≥40%)

Sperm morphology

≥4%

 

GENETIC TESTING

 

Assisted reproductive techniques have enabled azoospermic and severely oligospermic (<5 million/mL) men to reproduce. Therefore, there is a need to identify and discuss the consequences of possible mutations with the infertile couples. Genetic abnormalities are found in 3% to 5% of men with severe oligosperma, and 14% to 19% of men with non-obstructive azoospermia (NOA). The peripheral blood karyotype should be analyzed in these men unless there is a likely cause for hypospermatogenesis (e.g. cancer chemotherapy or orchitis). Of those with azoospermia, 2/3 have Klinefelter syndrome (47,XXY and mosaics) (154). Rarely, azoospermic men have a 46,XX chromosome constitution with Y-chromosome material detectable on the X-chromosome by FISH analysis using an antibody to the transcription factor encoded by the SRY gene. In studies of various ethnic groups, 4-15% of men with idiopathic non-obstructive azoospermic and 1–10% of men with severe oligospermia have microdeletions of the AZF regions of the Y chromosome (155, 156). Thresholds of 0.5-5 million sperm/mL have been suggested for genetic testing for higher specificity and overall lower cost (157) although some authors would like to see all infertile men tested.  Microdeletions (undetectable in standard karyotypes; YCMD) are detected by PCR and may partially or completely eliminate the azoospermia factor (AZF)a or the AZFb/c region including genes that are important for spermatogenesis. YCMD are detected by PCR. Couples are counselled that male offspring will inherit the AZF microdeletion, and will be at risk for infertility. In azoospermic men Y deletion testing is essential prior to considering surgical sperm retrieval because AZFa , AZFb or AZFB+c deletion are associated with very severe spermatogenic defects such as germ cell arrest or Sertoli cell only syndrome with an essentially zero prospect of success (160). On the other hand, even azoospermic men with AZFc deletions have ~ 50% chance of surgical sperm recovery (160).

 

Obstructive azoospermia may be recognized by a low-volume ejaculate with low pH. The testes of these men are generally normal in size, and FSH and inhibin-B levels are in the reference range. Obstruction can be demonstrated by ultrasound (158). If bilateral absence of the vas deferens (CBAVD) is found, approximately 75% of men will have cystic fibrosis, sometimes with little respiratory or pancreatic disease (159). When Cystic Fibrosis is diagnosed by genetic testing, the next step is to screen the female partner for Cystic Fibrosis gene abnormalities in order to assess the risk to any offspring.  

 

REFERENCES

 

  1. Rosner, W., Vesper, H., Endocrine, S., American Association for Clinical, C., American Association of Clinical, E., Androgen Excess, P.S., American Society for, B., Mineral, R., American Society for Reproductive, M., American Urological, A., et al. (2010). Toward excellence in testosterone testing: a consensus statement. The Journal of clinical endocrinology and metabolism 95, 4542-4548.
  2. Khera, M., Adaikan, G., Buvat, J., Carrier, S., El-Meliegy, A., Hatzimouratidis, K., McCullough, A., Morgentaler, A., Torres, L.O., and Salonia, A. (2016). Diagnosis and Treatment of Testosterone Deficiency: Recommendations From the Fourth International Consultation for Sexual Medicine (ICSM 2015). J Sex Med 13, 1787-1804.
  3. Bhasin, S., Brito, J.P., Cunningham, G.R., Hayes, F.J., Hodis, H.N., Matsumoto, A.M., Snyder, P.J., Swerdloff, R.S., Wu, F.C., and Yialamas, M.A. (2018). Testosterone Therapy in Men With Hypogonadism: An Endocrine Society Clinical Practice Guideline. The Journal of clinical endocrinology and metabolism 103, 1715-1744.
  4. Mulhall, J.P., Trost, L.W., Brannigan, R.E., Kurtz, E.G., Redmon, J.B., Chiles, K.A., Lightner, D.J., Miner, M.M., Murad, M.H., Nelson, C.J., et al. (2018). Evaluation and Management of Testosterone Deficiency: AUA Guideline. J Urol 200, 423-432.
  5. Forest, M.G., Sizonenko, P.C., Cathiard, A.M., and Bertrand, J. (1974). Hypophyso-gonadal function in humans during the first year of life. 1. Evidence for testicular activity in early infancy. The Journal of clinical investigation 53, 819-828.
  6. Grumbach, M.M. (2005). A window of opportunity: the diagnosis of gonadotropin deficiency in the male infant. The Journal of clinical endocrinology and metabolism 90, 3122-3127.
  7. Boyar, R.M., Rosenfeld, R.S., Kapen, S., Finkelstein, J.W., Roffwarg, H.P., Weitzman, E.D., and Hellman, L. (1974). Human puberty. Simultaneous augmented secretion of luteinizing hormone and testosterone during sleep. The Journal of clinical investigation 54, 609-618.
  8. Kulle, A.E., Riepe, F.G., Melchior, D., Hiort, O., and Holterhus, P.M. (2010). A novel ultrapressure liquid chromatography tandem mass spectrometry method for the simultaneous determination of androstenedione, testosterone, and dihydrotestosterone in pediatric blood samples: age- and sex-specific reference data. The Journal of clinical endocrinology and metabolism 95, 2399-2409.
  9. Plant, T.M. (2019). The neurobiological mechanism underlying hypothalamic GnRH pulse generation: the role of kisspeptin neurons in the arcuate nucleus. F1000Res 8.
  10. Veldhuis, J.D., King, J.C., Urban, R.J., Rogol, A.D., Evans, W.S., Kolp, L.A., and Johnson, M.L. (1987). Operating characteristics of the male hypothalamo-pituitary-gonadal axis: pulsatile release of testosterone and follicle-stimulating hormone and their temporal coupling with luteinizing hormone. The Journal of clinical endocrinology and metabolism 65, 929-941.
  11. Winters, S.J., and Troen, P. (1986). Testosterone and estradiol are co-secreted episodically by the human testis. The Journal of clinical investigation 78, 870-873.
  12. Spratt, D.I., O'Dea, L.S., Schoenfeld, D., Butler, J., Rao, P.N., and Crowley, W.F., Jr. (1988). Neuroendocrine-gonadal axis in men: frequent sampling of LH, FSH, and testosterone. The American journal of physiology 254, E658-666.
  13. Brambilla, D.J., Matsumoto, A.M., Araujo, A.B., and McKinlay, J.B. (2009). The effect of diurnal variation on clinical measurement of serum testosterone and other sex hormone levels in men. The Journal of clinical endocrinology and metabolism 94, 907-913.
  14. Winters, S.J. (1991). Diurnal rhythm of testosterone and luteinizing hormone in hypogonadal men. Journal of andrology 12, 185-190.
  15. Andersson, A.M., Carlsen, E., Petersen, J.H., and Skakkebaek, N.E. (2003). Variation in levels of serum inhibin B, testosterone, estradiol, luteinizing hormone, follicle-stimulating hormone, and sex hormone-binding globulin in monthly samples from healthy men during a 17-month period: possible effects of seasons. The Journal of clinical endocrinology and metabolism 88, 932-937.
  16. Caronia LM, Dwyer AA, Hayden D, Amati F, Pitteloud N, and Hayes FJ. (2013). Abrupt decrease in serum testosterone levels after an oral glucose load in men: implications for screening for hypogonadism. Clin Endocrinol (Oxf). 78(2):291-6.
  17. Vermeulen, A., and Verdonck, G. (1992). Representativeness of a single point plasma testosterone level for the long term hormonal milieu in men. The Journal of clinical endocrinology and metabolism 74, 939-942.
  18. Wu, F.C., Tajar, A., Beynon, J.M., Pye, S.R., Silman, A.J., Finn, J.D., O'Neill, T.W., Bartfai, G., Casanueva, F.F., Forti, G., et al. (2010). Identification of late-onset hypogonadism in middle-aged and elderly men. The New England journal of medicine 363, 123-135.
  19. Araujo, A.B., Esche, G.R., Kupelian, V., O'Donnell, A.B., Travison, T.G., Williams, R.E., Clark, R.V., and McKinlay, J.B. (2007). Prevalence of symptomatic androgen deficiency in men. The Journal of clinical endocrinology and metabolism 92, 4241-4247.
  20. Antonio, L., Wu, F.C., O'Neill, T.W., Pye, S.R., Ahern, T.B., Laurent, M.R., Huhtaniemi, I.T., Lean, M.E., Keevil, B.G., Rastrelli, G., et al. (2016). Low Free Testosterone Is Associated with Hypogonadal Signs and Symptoms in Men with Normal Total Testosterone. The Journal of clinical endocrinology and metabolism 101, 2647-2657.
  21. Vesper, H.W., Bhasin, S., Wang, C., Tai, S.S., Dodge, L.A., Singh, R.J., Nelson, J., Ohorodnik, S., Clarke, N.J., Salameh, W.A., et al. (2009). Interlaboratory comparison study of serum total testosterone (corrected) measurements performed by mass spectrometry methods. Steroids 74, 498-503.
  22. Salameh, W.A., Redor-Goldman, M.M., Clarke, N.J., Reitz, R.E., and Caulfield, M.P. (2010). Validation of a total testosterone assay using high-turbulence liquid chromatography tandem mass spectrometry: total and free testosterone reference ranges. Steroids 75, 169-175.
  23. Cawood, M.L., Field, H.P., Ford, C.G., Gillingwater, S., Kicman, A., Cowan, D., and Barth, J.H. (2005). Testosterone measurement by isotope-dilution liquid chromatography-tandem mass spectrometry: validation of a method for routine clinical practice. Clinical chemistry 51, 1472-1479.
  24. Kushnir, M.M., Rockwood, A.L., Roberts, W.L., Pattison, E.G., Bunker, A.M., Fitzgerald, R.L., and Meikle, A.W. (2006). Performance characteristics of a novel tandem mass spectrometry assay for serum testosterone. Clinical chemistry 52, 120-128.
  25. Sartorius, G., Spasevska, S., Idan, A., Turner, L., Forbes, E., Zamojska, A., Allan, C.A., Ly, L.P., Conway, A.J., McLachlan, R.I., et al. (2012). Serum testosterone, dihydrotestosterone and estradiol concentrations in older men self-reporting very good health: the healthy man study. Clinical endocrinology 77, 755-763.
  26. Harman, S.M., Metter, E.J., Tobin, J.D., Pearson, J., Blackman, M.R., and Baltimore Longitudinal Study of, A. (2001). Longitudinal effects of aging on serum total and free testosterone levels in healthy men. Baltimore Longitudinal Study of Aging. The Journal of clinical endocrinology and metabolism 86, 724-731.
  27. Travison, T.G., Araujo, A.B., Kupelian, V., O'Donnell, A.B., and McKinlay, J.B. (2007). The relative contributions of aging, health, and lifestyle factors to serum testosterone decline in men. The Journal of clinical endocrinology and metabolism 92, 549-555.
  28. Richard, A., Rohrmann, S., Zhang, L., Eichholzer, M., Basaria, S., Selvin, E., Dobs, A.S., Kanarek, N., Menke, A., Nelson, W.G., et al. (2014). Racial variation in sex steroid hormone concentration in black and white men: a meta-analysis. Andrology 2, 428-435.
  29. Bhasin, S., Pencina, M., Jasuja, G.K., Travison, T.G., Coviello, A., Orwoll, E., Wang, P.Y., Nielson, C., Wu, F., Tajar, A., et al. (2011). Reference ranges for testosterone in men generated using liquid chromatography tandem mass spectrometry in a community-based sample of healthy nonobese young men in the Framingham Heart Study and applied to three geographically distinct cohorts. The Journal of clinical endocrinology and metabolism 96, 2430-2439.
  30. Elmlinger, M.W., Kuhnel, W., Wormstall, H., and Doller, P.C. (2005). Reference intervals for testosterone, androstenedione and SHBG levels in healthy females and males from birth until old age. Clin Lab 51, 625-632.
  31. Mazur, A. (2009). The age-testosterone relationship in black, white, and Mexican-American men, and reasons for ethnic differences. Aging Male 12, 66-76.
  32. Wu, T.S., and Hammond, G.L. (2014). Naturally occurring mutants inform SHBG structure and function. Mol Endocrinol 28, 1026-1038.
  33. Winters, S.J., Kelley, D.E., and Goodpaster, B. (1998). The analog free testosterone assay: are the results in men clinically useful? Clinical chemistry 44, 2178-2182.
  34. Albanese, C., Christin-Maitre, S., Sluss, P.M., Crowley, W.F., and Jameson, J.L. (1994). Development of a bioassay for FSH using a recombinant human FSH receptor and a cAMP responsive luciferase reporter gene. Molecular and cellular endocrinology 101, 211-219.
  35. Brand, J.S., van der Tweel, I., Grobbee, D.E., Emmelot-Vonk, M.H., and van der Schouw, Y.T. (2011). Testosterone, sex hormone-binding globulin and the metabolic syndrome: a systematic review and meta-analysis of observational studies. Int J Epidemiol 40, 189-207.
  36.  Laaksonen, D.E., Niskanen, L., Punnonen, K., Nyyssonen, K., Tuomainen, T.P., Valkonen, V.P., Salonen, R., and Salonen, J.T. (2004). Testosterone and sex hormone-binding globulin predict the metabolic syndrome and diabetes in middle-aged men. Diabetes care 27, 1036-1041.
  37. Hammoud, A., Gibson, M., Hunt, S.C., Adams, T.D., Carrell, D.T., Kolotkin, R.L., and Meikle, A.W. (2009). Effect of Roux-en-Y gastric bypass surgery on the sex steroids and quality of life in obese men. The Journal of clinical endocrinology and metabolism 94, 1329-1332.
  38. Pasquali, R., Macor, C., Vicennati, V., Novo, F., De lasio, R., Mesini, P., Boschi, S., Casimirri, F., and Vettor, R. (1997). Effects of acute hyperinsulinemia on testosterone serum concentrations in adult obese and normal-weight men. Metabolism: clinical and experimental 46, 526-529.
  39. Xie, X., Liao, H., Dang, H., Pang, W., Guan, Y., Wang, X., Shyy, J.Y., Zhu, Y., and Sladek, F.M. (2009). Down-regulation of hepatic HNF4alpha gene expression during hyperinsulinemia via SREBPs. Mol Endocrinol 23, 434-443.
  40. Simo, R., Saez-Lopez, C., Barbosa-Desongles, A., Hernandez, C., and Selva, D.M. (2015). Novel insights in SHBG regulation and clinical implications. Trends Endocrinol Metab 26, 376-383.
  41. Winters, S.J., Gogineni, J., Karegar, M., Scoggins, C., Wunderlich, C.A., Baumgartner, R., and Ghooray, D.T. (2014). Sex hormone-binding globulin gene expression and insulin resistance. The Journal of clinical endocrinology and metabolism 99, E2780-2788.
  42. Goldman, A.L., Bhasin, S., Wu, F.C.W., Krishna, M., Matsumoto, A.M., and Jasuja, R. (2017). A Reappraisal of Testosterone's Binding in Circulation: Physiological and Clinical Implications. Endocr Rev 38, 302-324.
  43. Pardridge, W.M. (1986). Serum bioavailability of sex steroid hormones. Clinics in endocrinology and metabolism 15, 259-278.
  44. Keevil, B.G., and Adaway, J. (2019). Assessment of free testosterone concentration. The Journal of steroid biochemistry and molecular biology 190, 207-211.
  45. Chen, Y., Yazdanpanah, M., Wang, X.Y., Hoffman, B.R., Diamandis, E.P., and Wong, P.Y. (2010). Direct measurement of serum free testosterone by ultrafiltration followed by liquid chromatography tandem mass spectrometry. Clinical biochemistry 43, 490-496.
  46. Ly, L.P., Sartorius, G., Hull, L., Leung, A., Swerdloff, R.S., Wang, C., and Handelsman, D.J. (2010). Accuracy of calculated free testosterone formulae in men. Clinical endocrinology 73, 382-388.
  47. Vermeulen, A., Verdonck, L., and Kaufman, J.M. (1999). A critical evaluation of simple methods for the estimation of free testosterone in serum. The Journal of clinical endocrinology and metabolism 84, 3666-3672.
  48. Watts, E.L., Appleby, P.N., Albanes, D., Black, A., Chan, J.M., Chen, C., Cirillo, P.M., Cohn, B.A., Cook, M.B., Donovan, J.L., et al. (2017). Circulating sex hormones in relation to anthropometric, sociodemographic and behavioural factors in an international dataset of 12,300 men. PloS one 12, e0187741.
  49. Damassa, D.A., Lin, T.M., Sonnenschein, C., and Soto, A.M. (1991). Biological effects of sex hormone-binding globulin on androgen-induced proliferation and androgen metabolism in LNCaP prostate cells. Endocrinology 129, 75-84.
  50. Kapoor, P., Luttrell, B.M., and Williams, D. (1993). The free androgen index is not valid for adult males. The Journal of steroid biochemistry and molecular biology 45, 325-326.
  51. Fritz, K.S., McKean, A.J., Nelson, J.C., and Wilcox, R.B. (2008). Analog-based free testosterone test results linked to total testosterone concentrations, not free testosterone concentrations. Clinical chemistry 54, 512-516.
  52. Need, E.F., O'Loughlin, P.D., Armstrong, D.T., Haren, M.T., Martin, S.A., Tilley, W.D., Florey Adelaide Male Aging, S., Wittert, G.A., and Buchanan, G. (2010). Serum testosterone bioassay evaluation in a large male cohort. Clinical endocrinology 72, 87-98.
  53. van der Veen, A., van Faassen, M., de Jong, W.H.A., van Beek, A.P., Dijck-Brouwer, D.A.J., and Kema, I.P. (2019). Development and validation of a LC-MS/MS method for the establishment of reference intervals and biological variation for five plasma steroid hormones. Clinical biochemistry 68, 15-23.
  54. Maimoun, L., Philibert, P., Cammas, B., Audran, F., Bouchard, P., Fenichel, P., Cartigny, M., Pienkowski, C., Polak, M., Skordis, N., et al. (2011). Phenotypical, biological, and molecular heterogeneity of 5alpha-reductase deficiency: an extensive international experience of 55 patients. The Journal of clinical endocrinology and metabolism 96, 296-307.
  55. Chan, A.O., But, B.W., Lee, C.Y., Lam, Y.Y., Ng, K.L., Tung, J.Y., Kwan, E.Y., Chan, Y.K., Tsui, T.K., Lam, A.L., et al. (2013). Diagnosis of 5alpha-reductase 2 deficiency: is measurement of dihydrotestosterone essential? Clinical chemistry 59, 798-806.
  56. Thirumalai, A., Cooper, L.A., Rubinow, K.B., Amory, J.K., Lin, D.W., Wright, J.L., Marck, B.T., Matsumoto, A.M., and Page, S.T. (2016). Stable Intraprostatic Dihydrotestosterone in Healthy Medically Castrate Men Treated With Exogenous Testosterone. The Journal of clinical endocrinology and metabolism 101, 2937-2944.
  57. Russell, N., and Grossmann, M. (2019). MECHANISMS IN ENDOCRINOLOGY: Estradiol as a male hormone. Eur J Endocrinol 181, R23-R43.
  58. Finkelstein, J.S., Lee, H., Burnett-Bowie, S.A., Pallais, J.C., Yu, E.W., Borges, L.F., Jones, B.F., Barry, C.V., Wulczyn, K.E., Thomas, B.J., et al. (2013). Gonadal steroids and body composition, strength, and sexual function in men. The New England journal of medicine 369, 1011-1022.
  59. Narula, H.S., and Carlson, H.E. (2014). Gynaecomastia--pathophysiology, diagnosis and treatment. Nature reviews. Endocrinology 10, 684-698.
  60. Gandhi, G.Y., Basu, R., Dispenzieri, A., Basu, A., Montori, V.M., and Brennan, M.D. (2007). Endocrinopathy in POEMS syndrome: the Mayo Clinic experience. Mayo Clinic proceedings 82, 836-842.
  61. Shozu, M., Fukami, M., and Ogata, T. (2014). Understanding the pathological manifestations of aromatase excess syndrome: lessons for clinical diagnosis. Expert review of endocrinology & metabolism 9, 397-409.
  62. Hess, R.A., and Cooke, P.S. (2018). Estrogen in the male: a historical perspective. Biology of reproduction 99, 27-44.
  63. Wu, A., Shi, Z., Martin, S., Vincent, A., Heilbronn, L., and Wittert, G. (2018). Age-related changes in estradiol and longitudinal associations with fat mass in men. PloS one 13, e0201912.
  64. Dunn, J.F., Nisula, B.C., and Rodbard, D. (1981). Transport of steroid hormones: binding of 21 endogenous steroids to both testosterone-binding globulin and corticosteroid-binding globulin in human plasma. The Journal of clinical endocrinology and metabolism 53, 58-68.
  65. Rosner, W. (2015). Free estradiol and sex hormone-binding globulin. Steroids 99, 113-116.
  66. Khosla, S. (2008). Estrogen and bone: insights from estrogen-resistant, aromatase-deficient, and normal men. Bone 43, 414-417.
  67. Laouali, N., Brailly-Tabard, S., Helmer, C., Ancelin, M.L., Tzourio, C., Elbaz, A., Guiochon-Mantel, A., and Canonico, M. (2018). Oestradiol level, oestrogen receptors, and mortality in elderly men: The three-city cohort study. Clinical endocrinology 89, 514-525.
  68. Chadid, S., Barber, J.R., Rohrmann, S., Nelson, W.G., Yager, J.D., Kanarek, N.F., Bradwin, G., Dobs, A.S., McGlynn, K.A., and Platz, E.A. (2019). Age-Specific Serum Total and Free Estradiol Concentrations in Healthy Men in US Nationally Representative Samples. J Endocr Soc 3, 1825-1836.
  69. Resende, E.A., Lara, B.H., Reis, J.D., Ferreira, B.P., Pereira, G.A., and Borges, M.F. (2007). Assessment of basal and gonadotropin-releasing hormone-stimulated gonadotropins by immunochemiluminometric and immunofluorometric assays in normal children. The Journal of clinical endocrinology and metabolism 92, 1424-1429.
  70. Goji, K., and Tanikaze, S. (1993). Spontaneous gonadotropin and testosterone concentration profiles in prepubertal and pubertal boys: temporal relationship between luteinizing hormone and testosterone. Pediatric research 34, 229-236.
  71. Ventimiglia, E., Ippolito, S., Capogrosso, P., Pederzoli, F., Cazzaniga, W., Boeri, L., Cavarretta, I., Alfano, M., Vigano, P., Montorsi, F., et al. (2017). Primary, secondary and compensated hypogonadism: a novel risk stratification for infertile men. Andrology 5, 505-510.
  72. Pacenza, N., Pasqualini, T., Gottlieb, S., Knoblovits, P., Costanzo, P.R., Stewart Usher, J., Rey, R.A., Martinez, M.P., and Aszpis, S. (2012). Clinical Presentation of Klinefelter's Syndrome: Differences According to Age. International journal of endocrinology 2012, 324835.
  73. Kaufman, J.M., Lapauw, B., Mahmoud, A., T'Sjoen, G., and Huhtaniemi, I.T. (2019). Aging and the Male Reproductive System. Endocr Rev 40, 906-972.
  74. Awouters, M., Vanderschueren, D., and Antonio, L. (2019). Aromatase inhibitors and selective estrogen receptor modulators: Unconventional therapies for functional hypogonadism? Andrology.
  75. Bulun, S.E. (2014). Aromatase and estrogen receptor alpha deficiency. Fertility and sterility 101, 323-329.
  76. Arnhold, I.J., Melo, K., Costa, E.M., Danilovic, D., Inacio, M., Domenice, S., and Mendonca, B.B. (2011). 46,XY disorders of sex development (46,XY DSD) due to androgen receptor defects: androgen insensitivity syndrome. Adv Exp Med Biol 707, 59-61.
  77. Verhelst, J., Denis, L., Van Vliet, P., Van Poppel, H., Braeckman, J., Van Cangh, P., Mattelaer, J., D'Hulster, D., and Mahler, C. (1994). Endocrine profiles during administration of the new non-steroidal anti-androgen Casodex in prostate cancer. Clinical endocrinology 41, 525-530.
  78. de Kretser, D.M., Burger, H.G., and Hudson, B. (1974). The relationship between germinal cells and serum FSH levels in males with infertility. The Journal of clinical endocrinology and metabolism 38, 787-793.
  79. Sikaris, K., McLachlan, R.I., Kazlauskas, R., de Kretser, D., Holden, C.A., and Handelsman, D.J. (2005). Reproductive hormone reference intervals for healthy fertile young men: evaluation of automated platform assays. The Journal of clinical endocrinology and metabolism 90, 5928-5936.
  80. Ntali, G., Capatina, C., Grossman, A., and Karavitaki, N. (2014). Clinical review: Functioning gonadotroph adenomas. The Journal of clinical endocrinology and metabolism 99, 4423-4433.
  81. Heseltine, D., White, M.C., Kendall-Taylor, P., De Kretser, D.M., and Kelly, W. (1989). Testicular enlargement and elevated serum inhibin concentrations occur in patients with pituitary macroadenomas secreting follicle stimulating hormone. Clinical endocrinology 31, 411-423.
  82. Chamoun, R., Layfield, L., and Couldwell, W.T. (2013). Gonadotroph adenoma with secondary hypersecretion of testosterone. World Neurosurg 80, 900 e907-911.
  83. Thakkar, A., Kannan, S., Hamrahian, A., Prayson, R.A., Weil, R.J., and Faiman, C. (2014). Testicular "hyperstimulation" syndrome: a case of functional gonadotropinoma. Case reports in endocrinology 2014, 194716.
  84. Tajar, A., Forti, G., O'Neill, T.W., Lee, D.M., Silman, A.J., Finn, J.D., Bartfai, G., Boonen, S., Casanueva, F.F., Giwercman, A., et al. (2010). Characteristics of secondary, primary, and compensated hypogonadism in aging men: evidence from the European Male Ageing Study. The Journal of clinical endocrinology and metabolism 95, 1810-1818.
  85. Young, J., Xu, C., Papadakis, G.E., Acierno, J.S., Maione, L., Hietamaki, J., Raivio, T., and Pitteloud, N. (2019). Clinical Management of Congenital Hypogonadotropic Hypogonadism. Endocr Rev 40, 669-710.
  86. Berger, K., Souza, H., Brito, V.N., d'Alva, C.B., Mendonca, B.B., and Latronico, A.C. (2005). Clinical and hormonal features of selective follicle-stimulating hormone (FSH) deficiency due to FSH beta-subunit gene mutations in both sexes. Fertility and sterility 83, 466-470.
  87. Zheng, J., Mao, J., Cui, M., Liu, Z., Wang, X., Xiong, S., Nie, M., and Wu, X. (2017). Novel FSHbeta mutation in a male patient with isolated FSH deficiency and infertility. European journal of medical genetics 60, 335-339.
  88. Lamminen, T., and Huhtaniemi, I. (2001). A common genetic variant of luteinizing hormone; relation to normal and aberrant pituitary-gonadal function. European journal of pharmacology 414, 1-7.
  89. Winters, S.J., Moore, J.P., Jr., and Clark, B.J. (2018). Leydig cell insufficiency in hypospermatogenesis: a paracrine effect of activin-inhibin signaling? Andrology 6, 262-271.
  90. Basciani, S., Watanabe, M., Mariani, S., Passeri, M., Persichetti, A., Fiore, D., Scotto d'Abusco, A., Caprio, M., Lenzi, A., Fabbri, A., et al. (2012). Hypogonadism in a patient with two novel mutations of the luteinizing hormone beta-subunit gene expressed in a compound heterozygous form. The Journal of clinical endocrinology and metabolism 97, 3031-3038.
  91. Yang, X., Ochin, H., Shu, L., Liu, J., Shen, J., Liu, J., Lin, C., and Cui, Y. (2018). Homozygous nonsense mutation Trp28X in the LHB gene causes male hypogonadism. Journal of assisted reproduction and genetics 35, 913-919.
  92. Weiss, J., Axelrod, L., Whitcomb, R.W., Harris, P.E., Crowley, W.F., and Jameson, J.L. (1992). Hypogonadism caused by a single amino acid substitution in the beta subunit of luteinizing hormone. The New England journal of medicine 326, 179-183.
  93. Campo, S., Andreone, L., Ambao, V., Urrutia, M., Calandra, R.S., and Rulli, S.B. (2019). Hormonal Regulation of Follicle-Stimulating Hormone Glycosylation in Males. Frontiers in endocrinology 10, 17.
  94. Dufau, M.L., Pock, R., Neubauer, A., and Catt, K.J. (1976). In vitro bioassay of LH in human serum: the rat interstitial cell testosterone (RICT) assay. The Journal of clinical endocrinology and metabolism 42, 958-969.
  95. Dahl, K.D., and Stone, M.P. (1992). FSH isoforms, radioimmunoassays, bioassays, and their significance. Journal of andrology 13, 11-22.
  96. Winters, S.J., and Troen, P. (1985). Pulsatile secretion of immunoreactive alpha-subunit in man. The Journal of clinical endocrinology and metabolism 60, 344-348.
  97. Styne, D.M., Kaplan, S.L., and Grumbach, M.M. (1980). Plasma glycoprotein hormone alpha-subunit in the neonate and in prepubertal and pubertal children: effects of luteinizing hormone-releasing hormone. The Journal of clinical endocrinology and metabolism 50, 450-455.
  98. Kourides, I.A., Weintraub, B.D., Ridgway, E.C., and Maloof, F. (1975). Pituitary secretion of free alpha and beta subunit of human thyrotropin in patients with thyroid disorders. The Journal of clinical endocrinology and metabolism 40, 872-885.
  99. Mainieri, A.S., and Elnecave, R.H. (2003). Usefulness of the free alpha-subunit to diagnose hypogonadotropic hypogonadism. Clinical endocrinology 59, 307-313.
  100. Oppenheim, D.S., Kana, A.R., Sangha, J.S., and Klibanski, A. (1990). Prevalence of alpha-subunit hypersecretion in patients with pituitary tumors: clinically nonfunctioning and somatotroph adenomas. The Journal of clinical endocrinology and metabolism 70, 859-864.
  101. Medri, G., Carella, C., Padmanabhan, V., Rossi, C.M., Amato, G., De Santo, N.G., Beitins, I.Z., and Beck-Peccoz, P. (1993). Pituitary glycoprotein hormones in chronic renal failure: evidence for an uncontrolled alpha-subunit release. Journal of endocrinological investigation 16, 169-174.
  102. Bay, K., Main, K.M., Toppari, J., and Skakkebaek, N.E. (2011). Testicular descent: INSL3, testosterone, genes and the intrauterine milieu. Nat Rev Urol 8, 187-196.
  103. Johansen, M.L., Anand-Ivell, R., Mouritsen, A., Hagen, C.P., Mieritz, M.G., Soeborg, T., Johannsen, T.H., Main, K.M., Andersson, A.M., Ivell, R., et al. (2014). Serum levels of insulin-like factor 3, anti-Mullerian hormone, inhibin B, and testosterone during pubertal transition in healthy boys: a longitudinal pilot study. Reproduction (Cambridge, England) 147, 529-535.
  104. Ferlin, A., Arredi, B., Zuccarello, D., Garolla, A., Selice, R., and Foresta, C. (2006). Paracrine and endocrine roles of insulin-like factor 3. Journal of endocrinological investigation 29, 657-664.
  105. Trabado, S., Maione, L., Bry-Gauillard, H., Affres, H., Salenave, S., Sarfati, J., Bouvattier, C., Delemer, B., Chanson, P., Le Bouc, Y., et al. (2014). Insulin-like peptide 3 (INSL3) in men with congenital hypogonadotropic hypogonadism/Kallmann syndrome and effects of different modalities of hormonal treatment: a single-center study of 281 patients. The Journal of clinical endocrinology and metabolism 99, E268-275.
  106. Makanji, Y., Zhu, J., Mishra, R., Holmquist, C., Wong, W.P., Schwartz, N.B., Mayo, K.E., and Woodruff, T.K. (2014). Inhibin at 90: from discovery to clinical application, a historical review. Endocr Rev 35, 747-794.
  107. Gray, P.C., Bilezikjian, L.M., and Vale, W. (2002). Antagonism of activin by inhibin and inhibin receptors: a functional role for betaglycan. Molecular and cellular endocrinology 188, 254-260.
  108. Ludlow, H., Muttukrishna, S., Hyvonen, M., and Groome, N.P. (2008). Development of a new antibody to the human inhibin/activin betaB subunit and its application to improved inhibin B ELISAs. J Immunol Methods 329, 102-111.
  109. Andersson, A.M., Toppari, J., Haavisto, A.M., Petersen, J.H., Simell, T., Simell, O., and Skakkebaek, N.E. (1998). Longitudinal reproductive hormone profiles in infants: peak of inhibin B levels in infant boys exceeds levels in adult men. The Journal of clinical endocrinology and metabolism 83, 675-681.
  110. Mahmoud, A.M., Goemaere, S., De Bacquer, D., Comhaire, F.H., and Kaufman, J.M. (2000). Serum inhibin B levels in community-dwelling elderly men. Clinical endocrinology 53, 141-147.
  111. Anawalt, B.D., Bebb, R.A., Matsumoto, A.M., Groome, N.P., Illingworth, P.J., McNeilly, A.S., and Bremner, W.J. (1996). Serum inhibin B levels reflect Sertoli cell function in normal men and men with testicular dysfunction. The Journal of clinical endocrinology and metabolism 81, 3341-3345.
  112. Abdelrahaman, E., Raghavan, S., Baker, L., Weinrich, M., and Winters, S.J. (2005). Racial difference in circulating sex hormone-binding globulin levels in prepubertal boys. Metabolism: clinical and experimental 54, 91-96.
  113. Barbotin, A.L., Ballot, C., Sigala, J., Ramdane, N., Duhamel, A., Marcelli, F., Rigot, J.M., Dewailly, D., Pigny, P., and Mitchell, V. (2015). The serum inhibin B concentration and reference ranges in normozoospermia. Eur J Endocrinol 172, 669-676.
  114. Andersson, A.M. (2000). Inhibin B in the assessment of seminiferous tubular function. Baillieres Best Pract Res Clin Endocrinol Metab 14, 389-397.
  115. Crofton, P.M., Evans, A.E., Groome, N.P., Taylor, M.R., Holland, C.V., and Kelnar, C.J. (2002). Inhibin B in boys from birth to adulthood: relationship with age, pubertal stage, FSH and testosterone. Clinical endocrinology 56, 215-221.
  116. Myers, G.M., Lambert-Messerlian, G.M., and Sigman, M. (2009). Inhibin B reference data for fertile and infertile men in Northeast America. Fertility and sterility 92, 1920-1923.
  117. Pierik, F.H., Vreeburg, J.T., Stijnen, T., De Jong, F.H., and Weber, R.F. (1998). Serum inhibin B as a marker of spermatogenesis. The Journal of clinical endocrinology and metabolism 83, 3110-3114.
  118. von Eckardstein, S., Simoni, M., Bergmann, M., Weinbauer, G.F., Gassner, P., Schepers, A.G., and Nieschlag, E. (1999). Serum inhibin B in combination with serum follicle-stimulating hormone (FSH) is a more sensitive marker than serum FSH alone for impaired spermatogenesis in men, but cannot predict the presence of sperm in testicular tissue samples. The Journal of clinical endocrinology and metabolism 84, 2496-2501.
  119. Christiansen, P., Andersson, A.M., and Skakkebaek, N.E. (2003). Longitudinal studies of inhibin B levels in boys and young adults with Klinefelter syndrome. The Journal of clinical endocrinology and metabolism 88, 888-891.
  120. Wallace, E.M., Groome, N.P., Riley, S.C., Parker, A.C., and Wu, F.C. (1997). Effects of chemotherapy-induced testicular damage on inhibin, gonadotropin, and testosterone secretion: a prospective longitudinal study. The Journal of clinical endocrinology and metabolism 82, 3111-3115.
  121. Petersen, P.M., Andersson, A.M., Rorth, M., Daugaard, G., and Skakkebaek, N.E. (1999). Undetectable inhibin B serum levels in men after testicular irradiation. The Journal of clinical endocrinology and metabolism 84, 213-215.
  122. Isaksson, S., Eberhard, J., Stahl, O., Cavallin-Stahl, E., Cohn-Cedermark, G., Arver, S., Lundberg Giwercman, Y., and Giwercman, A. (2014). Inhibin B concentration is predictive for long-term azoospermia in men treated for testicular cancer. Andrology 2, 252-258.
  123. Martin, C.W., Riley, S.C., Everington, D., Groome, N.P., Riemersma, R.A., Baird, D.T., and Anderson, R.A. (2000). Dose-finding study of oral desogestrel with testosterone pellets for suppression of the pituitary-testicular axis in normal men. Human reproduction (Oxford, England) 15, 1515-1524.
  124. Mitchell, V., Boitrelle, F., Pigny, P., Robin, G., Marchetti, C., Marcelli, F., and Rigot, J.M. (2010). Seminal plasma levels of anti-Mullerian hormone and inhibin B are not predictive of testicular sperm retrieval in nonobstructive azoospermia: a study of 139 men. Fertility and sterility 94, 2147-2150.
  125. Seminara, S.B., Boepple, P.A., Nachtigall, L.B., Pralong, F.P., Khoury, R.H., Sluss, P.M., Lecain, A.E., and Crowley, W.F., Jr. (1996). Inhibin B in males with gonadotropin-releasing hormone (GnRH) deficiency: changes in serum concentration after shortterm physiologic GnRH replacement--a clinical research center study. The Journal of clinical endocrinology and metabolism 81, 3692-3696.
  126. Coutant, R., Biette-Demeneix, E., Bouvattier, C., Bouhours-Nouet, N., Gatelais, F., Dufresne, S., Rouleau, S., and Lahlou, N. (2010). Baseline inhibin B and anti-Mullerian hormone measurements for diagnosis of hypogonadotropic hypogonadism (HH) in boys with delayed puberty. The Journal of clinical endocrinology and metabolism 95, 5225-5232.
  127. Binder, G., Schweizer, R., Blumenstock, G., and Braun, R. (2015). Inhibin B plus LH vs GnRH agonist test for distinguishing constitutional delay of growth and puberty from isolated hypogonadotropic hypogonadism in boys. Clinical endocrinology 82, 100-105.
  128. Brauner, R., Neve, M., Allali, S., Trivin, C., Lottmann, H., Bashamboo, A., and McElreavey, K. (2011). Clinical, biological and genetic analysis of anorchia in 26 boys. PloS one 6, e23292.
  129. Clemente, M., Caracseghi, F., Gussinyer, M., Yeste, D., Albisu, M., Vazquez, E., Ortega, A., and Carrascosa, A. (2011). Macroorchidism and panhypopituitarism: two different forms of presentation of FSH-secreting pituitary adenomas in adolescence. Horm Res Paediatr 75, 225-230.
  130. Bergada, I., Del Toro, K., Katz, O., Chemes, H., and Campo, S. (2000). Serum inhibin B concentration in a prepubertal boy with gynecomastia and Peutz-Jeghers syndrome. J Pediatr Endocrinol Metab 13, 101-103.
  131. Coutant, R., Lumbroso, S., Rey, R., Lahlou, N., Venara, M., Rouleau, S., Sultan, C., and Limal, J.M. (2001). Macroorchidism due to autonomous hyperfunction of Sertoli cells and G(s)alpha gene mutation: an unusual expression of McCune-Albright syndrome in a prepubertal boy. The Journal of clinical endocrinology and metabolism 86, 1778-1781.
  132. Fragoso, M.C., Kohek, M.B., Martin, R.M., Latronico, A.C., Lucon, A.M., Zerbini, M.C., Longui, C.A., Mendonca, B.B., and Domenice, S. (2007). An inhibin B and estrogen-secreting adrenocortical carcinoma leading to selective FSH suppression. Hormone research 67, 7-11.
  133. Xu, H.Y., Zhang, H.X., Xiao, Z., Qiao, J., and Li, R. (2019). Regulation of anti-Mullerian hormone (AMH) in males and the associations of serum AMH with the disorders of male fertility. Asian journal of andrology 21, 109-114.
  134. Josso, N., Rey, R.A., and Picard, J.Y. (2013). Anti-mullerian hormone: a valuable addition to the toolbox of the pediatric endocrinologist. International journal of endocrinology 2013, 674105.
  135. Sharpe, R.M., McKinnell, C., Kivlin, C., and Fisher, J.S. (2003). Proliferation and functional maturation of Sertoli cells, and their relevance to disorders of testis function in adulthood. Reproduction (Cambridge, England) 125, 769-784.
  136. Young, J., Rey, R., Couzinet, B., Chanson, P., Josso, N., and Schaison, G. (1999). Antimullerian hormone in patients with hypogonadotropic hypogonadism. The Journal of clinical endocrinology and metabolism 84, 2696-2699.
  137. Tuttelmann, F., Dykstra, N., Themmen, A.P., Visser, J.A., Nieschlag, E., and Simoni, M. (2009). Anti-Mullerian hormone in men with normal and reduced sperm concentration and men with maldescended testes. Fertility and sterility 91, 1812-1819.
  138. Andersen, J.M., Herning, H., Aschim, E.L., Hjelmesaeth, J., Mala, T., Hanevik, H.I., Bungum, M., Haugen, T.B., and Witczak, O. (2015). Body Mass Index Is Associated with Impaired Semen Characteristics and Reduced Levels of Anti-Mullerian Hormone across a Wide Weight Range. PloS one 10, e0130210.
  139. Aksglaede, L., Sorensen, K., Boas, M., Mouritsen, A., Hagen, C.P., Jensen, R.B., Petersen, J.H., Linneberg, A., Andersson, A.M., Main, K.M., et al. (2010). Changes in anti-Mullerian hormone (AMH) throughout the life span: a population-based study of 1027 healthy males from birth (cord blood) to the age of 69 years. The Journal of clinical endocrinology and metabolism 95, 5357-5364.
  140. Bertelloni, S., Russo, G., and Baroncelli, G.I. (2018). Human Chorionic Gonadotropin Test: Old Uncertainties, New Perspectives, and Value in 46,XY Disorders of Sex Development. Sexual development : genetics, molecular biology, evolution, endocrinology, embryology, and pathology of sex determination and differentiation 12, 41-49.
  141. McEachern, R., Houle, A.M., Garel, L., and Van Vliet, G. (2004). Lost and found testes: the importance of the hCG stimulation test and other testicular markers to confirm a surgical declaration of anorchia. Hormone research 62, 124-128.
  142. Wheeler, K.M., Sharma, D., Kavoussi, P.K., Smith, R.P., and Costabile, R. (2019). Clomiphene Citrate for the Treatment of Hypogonadism. Sexual medicine reviews 7, 272-276.
  143. Glass, A.R., and Eil, C. (1986). Ketoconazole-induced reduction in serum 1,25-dihydroxyvitamin D. The Journal of clinical endocrinology and metabolism 63, 766-769.
  144. Winters, S.J., and Troen, P. (1985). Evidence for a role of endogenous estrogen in the hypothalamic control of gonadotropin secretion in men. The Journal of clinical endocrinology and metabolism 61, 842-845.
  145. Hayes, F.J., Seminara, S.B., Decruz, S., Boepple, P.A., and Crowley, W.F., Jr. (2000). Aromatase inhibition in the human male reveals a hypothalamic site of estrogen feedback. The Journal of clinical endocrinology and metabolism 85, 3027-3035.
  146. Veldhuis, J.D., Zwart, A., Mulligan, T., and Iranmanesh, A. (2001). Muting of androgen negative feedback unveils impoverished gonadotropin-releasing hormone/luteinizing hormone secretory reactivity in healthy older men. The Journal of clinical endocrinology and metabolism 86, 529-535.
  147. Ghai, K., Cara, J.F., and Rosenfield, R.L. (1995). Gonadotropin releasing hormone agonist (nafarelin) test to differentiate gonadotropin deficiency from constitutionally delayed puberty in teen-age boys--a clinical research center study. The Journal of clinical endocrinology and metabolism 80, 2980-2986.
  148. Wilson, D.A., Hofman, P.L., Miles, H.L., Unwin, K.E., McGrail, C.E., and Cutfield, W.S. (2006). Evaluation of the buserelin stimulation test in diagnosing gonadotropin deficiency in males with delayed puberty. J Pediatr 148, 89-94.
  149. Segal, T.Y., Mehta, A., Anazodo, A., Hindmarsh, P.C., and Dattani, M.T. (2009). Role of gonadotropin-releasing hormone and human chorionic gonadotropin stimulation tests in differentiating patients with hypogonadotropic hypogonadism from those with constitutional delay of growth and puberty. The Journal of clinical endocrinology and metabolism 94, 780-785.
  150. Harrington, J., and Palmert, M.R. (2012). Clinical review: Distinguishing constitutional delay of growth and puberty from isolated hypogonadotropic hypogonadism: critical appraisal of available diagnostic tests. The Journal of clinical endocrinology and metabolism 97, 3056-3067.
  151. Barratt, C.L.R., Bjorndahl, L., De Jonge, C.J., Lamb, D.J., Osorio Martini, F., McLachlan, R., Oates, R.D., van der Poel, S., St John, B., Sigman, M., et al. (2017). The diagnosis of male infertility: an analysis of the evidence to support the development of global WHO guidance-challenges and future research opportunities. Hum Reprod Update 23, 660-680.
  152. Organization, W.H. WHO Laboratory Manual for the Examination and Processing of Human Semen, (Cambridge Univeristy Press, New York).
  153. Guzick, D.S., Overstreet, J.W., Factor-Litvak, P., Brazil, C.K., Nakajima, S.T., Coutifaris, C., Carson, S.A., Cisneros, P., Steinkampf, M.P., Hill, J.A., et al. (2001). Sperm morphology, motility, and concentration in fertile and infertile men. The New England journal of medicine 345, 1388-1393.
  154. Yatsenko, A.N., Yatsenko, S.A., Weedin, J.W., Lawrence, A.E., Patel, A., Peacock, S., Matzuk, M.M., Lamb, D.J., Cheung, S.W., and Lipshultz, L.I. (2010). Comprehensive 5-year study of cytogenetic aberrations in 668 infertile men. J Urol 183, 1636-1642.
  155. Rives, N. (2014). Y chromosome microdeletions and alterations of spermatogenesis, patient approach and genetic counseling. Ann Endocrinol (Paris) 75, 112-114.
  156. Zhang, F., Li, L., Wang, L., Yang, L., Liang, Z., Li, J., Jin, F., and Tian, Y. (2013). Clinical characteristics and treatment of azoospermia and severe oligospermia patients with Y-chromosome microdeletions. Mol Reprod Dev 80, 908-915.
  157. Johnson, M., Raheem, A., De Luca, F., Hallerstrom, M., Zainal, Y., Poselay, S., Mohammadi, B., Moubasher, A., Johnson, T.F., Muneer, A., et al. (2019). An analysis of the frequency of Y-chromosome microdeletions and the determination of a threshold sperm concentration for genetic testing in infertile men. BJU Int 123, 367-372.
  158. Mittal, P.K., Little, B., Harri, P.A., Miller, F.H., Alexander, L.F., Kalb, B., Camacho, J.C., Master, V., Hartman, M., and Moreno, C.C. (2017). Role of Imaging in the Evaluation of Male Infertility. Radiographics 37, 837-854.
  159. de Souza, D.A.S., Faucz, F.R., Pereira-Ferrari, L., Sotomaior, V.S., and Raskin, S. (2018). Congenital bilateral absence of the vas deferens as an atypical form of cystic fibrosis: reproductive implications and genetic counseling. Andrology 6, 127-135.
  160. Krausz C, Hoefsloot L, Simoni M, Tüttelmann F; European Academy of Andrology; European Molecular Genetics Quality Network. EAA/EMQN best practice guidelines for molecular diagnosis of Y-chromosomal microdeletions: state-of-the-art 2013. Andrology. 2014 Jan;2(1):5-19

Overview of Endocrine Hypertension

ABSTRACT

Endocrine hypertension typically is referred to disorders of the adrenal gland including primary aldosteronism, glucocorticoid excess, and the pheochromocytoma-paraganglioma syndromes. Rare conditions in patients with congenital adrenal hyperplasia and glucocorticoid resistance (Chrousos syndrome) can also lead to hypertension. Nonadrenal endocrine disorders, such as growth hormone excess or deficiency, thyroid dysfunction, primary hyperparathyreoidism, testosterone deficiency, vitamin D deficiency, obesity-associated hypertension, insulin resistance and metabolic syndrome are also linked to hypertension. In this chapter, we provide an overview of endocrine hypertension including rare syndromes of mineralocorticoid excess.

INTRODUCTION

Hypertension is the most common diagnosis in USA as it affects approximately 31% of Americans (1,2) and approx. 33% of the Mozambican population using a blood pressure cutoff

of 139/89 mm Hg (3). The assignment of a diagnosis of hypertension is dependent on the appropriate measurement of blood pressure, the level of blood pressure (BP) elevation, and the duration of follow-up (4). The secondary causes of hypertension include mostly renal as well as endocrine diseases. An accurate diagnosis of endocrine hypertension offers clinicians the chance to achieve an optimal treatment with either specific pharmacologic or surgical therapy (5). Herein, the different causes of endocrine hypertension with a focus on prevalence, clinical presentation, and currently diagnostic tools.

How to Measure BP

Manual measurement using a mercury sphygmomanometer and a stethoscope remains the Gold Standard. However due to environmental issues regarding mercury, this technique tends to be abandoned. Automatic devices have substituted them, but a standardised procedure of obtaining comparable measurements is poor and their validity in clinical practice is limited (6). The device should have an upper arm cuff and should be properly validated and calibrated. A correct cuff size that encircles 75%–100% of the arm should be used. Blood pressure assessment should be based on the mean of 2 or more properly measured seated BP readings on each of 2 or more office visits. Optimally, the measurement of the blood pressure can take place in the office, with the patient seated comfortably with legs uncrossed or in supine position for 3–5 minutes without talking or moving around. It is recommended to avoid caffeine, smoking as well as exercise before the measurement. Clothes covering the cuff location of the upper arm should be removed (7). At the first visit, BP should be recorded in both arms and the higher reading must be considered and repeated measurements after 1-2 minutes can be done. During the measurement, the patient’s arm needs to be supported, and upper arm must be at the level of right atrium. Regarding auscultatory determinations, radial pulse obliteration can be palpated to estimate systolic blood pressure. Korotkoff sounds must be recorded, with readings of SBP and DBP at the onset of the first and the last audible sound, respectively (7).

Classification of BP

Based on recommendations of the Seventh Report of the Joint National Committee on Prevention, Detection, Evaluation, and Treatment of High Blood Pressure (JNC 7)(8), the classification of BP for adults aged 18 years or older has been as follows:

  • Normal: Systolic lower than 120 mm Hg, diastolic lower than 80 mm Hg
  • Prehypertension: Systolic 120-139 mm Hg, diastolic 80-89 mm Hg
  • Stage 1: Systolic 140-159 mm Hg, diastolic 90-99 mm Hg
  • Stage 2: Systolic 160 mm Hg or greater, diastolic 100 mm Hg or greater

 

The 2017 ACC/AHA guidelines eliminate the classification of prehypertension and divides it into two levels (9):

  • Elevated blood pressure with a systolic pressure between 120- and 129-mm Hg and diastolic pressure less than 80 mm Hg
  • Stage 1 hypertension, with a systolic pressure of 130 to 139 mm Hg or a diastolic pressure of 80 to 89 mm Hg

Figure 1 provides an overview of classification of BP for adults 18 years and older.

Figure 1. Classification of Hypertension. AHA, American Heart Association; ACC, American College of Cardiology; ESC, European Society of Cardiology; ESH, European Society of Hypertension; DHL, German Hypertension League; NICE, National Institute for Health and Care Excellence of the United Kingdom. DBP, diastolic blood pressure; SBP, systolic blood pressure. Modified from: Jordan J, Kurschat C, Reuter H. Arterial hypertension. Dtsch Arztebl Int. 2018 Aug 20;115(33-34):557-568(10)

Prevalence of Hypertension

Several studies have previously reported prevalence of hypertension among different populations worldwide, but these data depend on different classification systems used. Hypertension affects 28.6% of adults in United States Data from the National Health and Nutrition Examination Survey 2011-2012 showed an increase in the prevalence of hypertension in all age groups compared to 1991 (11). Among adults with hypertension in that survey, 52% achieved a BP of less than 140/90 mm Hg with 76% taking antihypertensives, and with 83% being aware of their hypertension. The prevalence of hypertension increases with age and most individuals with hypertension are diagnosed with primary (essential) hypertension. Hypertension is a major risk factor for stroke, ischemic heart disease, and cardiac failure. It is the second most common reason for office visits to physicians in the United States. Analysis of the Framingham study data suggested that individuals from age 40 to 69 years have an increasing risk of stroke or coronary artery disease mortality with every 20 mm Hg increment in SBP.

Prevalence of Secondary Hypertension

In most people, hypertension is primary, but approximately 15-30% of hypertensive population has secondary hypertension (12). Among children presenting with hypertension, 50% have a secondary cause (13). Young adults (<40 years old), are reported to have a prevalence of secondary hypertension 30% (14). The secondary causes of hypertension include primarily causes such as primary renal disease, oral contraceptive use, sleep apnea syndrome, congenital or acquired cardiovascular disease (i.e. coarctation of the aorta) and excess hormonal secretion. Endocrine Hypertension was previously reported to account for approx. Recent studies suggest an overall prevalence of >5% and possibly > 10% for endocrine hypertension among the hypertensive population (15,16), but several authors have suggested that this prevalence is probably underestimated. The most common causes of endocrine hypertension are excess production of mineralocorticoids (i.e. primary hyperaldosteronism), catecholamines (pheochromocytoma), thyroid hormone, and glucocorticoids (Cushing syndrome) (17). Table 1 lists the most common causes of Secondary hypertension.

Table 1. Etiology of Secondary Hypertension

Endocrine Causes

Other Causes

Adrenal-dependent causes 

Renal causes (2.5-6%)

Pheochromocytoma and sympathetic paraganglioma 

Polycystic kidney disease

Primary aldosteronism 

Chronic kidney disease

Hyperdeoxycorticosteronism 

Urinary tract obstruction

Congenital adrenal hyperplasia 

Renin-producing tumor

 11β-Hydroxylase deficiency 

Liddle syndrome

 17α-Hydroxylase deficiency 

Renovascular causes

 Deoxycorticosterone-producing tumor 

renal artery stenosis fibromuscular dysplasia or atherosclerosis

 Chrousos syndrome

Vascular causes 

   Cushing syndrome 

Coarctation of aorta

   Apparent mineralocorticoid excess/11β- hydroxysteroid dehydrogenase deficiency 

Vasculitis

Parathyroid-dependent causes 

Collagen vascular disease

  Hyperparathyroidism 

Neurogenic causes 

Pituitary-dependent causes 

Brain tumor

 Acromegaly 

Autonomic dysfunction

 Cushing disease 

Sleep apnea

Secondary hyperaldosteronism 

Intracranial hypertension

 Renovascular hypertension 

Drugs and toxins

Thyroid-dependent causes 

Alcohol

 Hypothyroidism 

Cocaine

 Hyperthyroidism 

Cyclosporine, tacrolimus

Vitamin D deficiency

NSAIDs

 

Erythropoietin

Adrenergic medications

Decongestants containing ephedrine

Herbal remedies containing licorice or ephedrine

Nicotine

 

Prevalence of Resistant Hypertension and Typical Causes

The prevalence of resistant hypertension is high: 53% of patients in NHANES had a BP < 140/90 mm Hg vs. 48% in the Framingham Heart Study. In NHANES participants with chronic kidney disease, 37% had a BP < 130/80 mm Hg. In ALLHAT, 34% of patients

remained uncontrolled after 5-year follow-up on at least 2 antihypertensive drugs (5).

One important question in this regard is when to screen for secondary causes. Some patients with hypertension, but without primary aldosteronism, demonstrate ACTH-dependent aldosterone hypersecretion by stress (18). The clinician should carefully screen for cardinal signs and symptoms of Cushing syndrome, hyper- or hypothyroidism, acromegaly, insulin resistance (acanthosis nigricans), or pheochromocytoma (flushing and excessive sweating). Hypertension in young patients and refractory hypertension (characterized by poorly controlled blood pressure on > 3 antihypertensive drugs) should alert the physician to screen for secondary causes (14). The importance of endocrine-mediated hypertension resides in the fact that in most cases, the cause is clear and can be traced to the actions of a hormone, often produced in excess by a tumor, such as an aldosteronoma, in a patient with hypertension due to primary aldosteronism. More importantly, once the diagnosis is made, a disease-specific targeted antihypertensive therapy can be implemented, and, in some cases, surgical intervention may result in complete cure, obviating the need for life-long antihypertensive treatment.

As in other causes of hypertension, the clinician should question the patient about dietary habits (salt intake etc.), weight fluctuations, use of over the counter drugs and health supplements including teas and herbal preparations, recreational drugs, and oral contraceptives. Moreover, a detailed family history may provide valuable insights into familial forms of endocrine hypertension. The review of systems should include disease-specific questions. Many patients harboring a pheochromocytoma are symptomatic. Symptoms may include headaches, palpitations, anxiety-like attacks and profuse sweating, similar to symptoms of hyperthyroidism. The triad headache, palpitations, and sweating in a hypertensive patient was initially found to have a sensitivity of 91% and specificity of 94% for pheochromocytoma (19). More recent studies suggest that this typical triad of symptoms is found much less frequently, for instance, in only 10% of cases (20). Ten or more percent of patients with pheochromocytoma may not have any clinical symptoms and may be normotensive (19,21,22).

Patients with Cushing’s syndrome often complain of weight gain, insomnia, depression, easy bruising and fatigue. Acne and hirsutism (in women) can also be observed. The challenge these days is to recognize patients with evolving Cushing’s syndrome amongst the many obese and often poorly controlled diabetic individuals. An Endocrine Society Clinical Practice Guideline can assist in this task (23). Primary hyperaldosteronism is manifested by mild to severe hypertension. Hypokalemia can be present, but it is not a universal finding and there is normokalemic and normotensive primary aldosteronism (24,25). Polyuria, myopathy and cardiac dysrhythmias may occur in cases of severe hypokalemia. A thorough physical exam with attention to evidence of target organ injury and features of secondary hypertension should be conducted.

Low renin is often associated with several causes of hypertension. Figure 2 lists conditions with low renin levels.

Figure 2. Low Renin Conditions

Despite the increasing understanding of the pathophysiology of hypertension, control of the disease is often difficult and far from optimal. Recent large meta-analyses and genotype studies have identified some “risk genes” for hypertension (15). Surendran and colleagues found a low frequency nonsense variant in the gene ENPEP, which codes for the enzyme aminopeptidase A that converts angiotensin II into angiotensin III and therefore being part of the regulation of the renin-angiotensin-aldosterone system  (26). Liu and colleagues observed associations for the aggregation of rare and low frequency missense variants in the genes NPR1, DBH, and PTPMT1 (27). The gene DBH codes for the enzyme dopamine beta-hydroxylase, which catalyzes the conversion of dopamine into noradrenaline and, thereby, influences the autonomic nervous system. The gene PTPMT1 codes for the mitochondrial protein tyrosine phosphatase 1, which influences insulin production (27).

CLINICAL DIAGNOSIS OF ENDOCRINE HYPERTENSION

The first step when evaluating a patient with suspected endocrine-related hypertension is to exclude other causes of secondary hypertension, particularly renal disorders. A detailed medical history and review of systems should be obtained. The onset of hypertension and the response to previous anti-hypertensive treatment should be determined. Consideration of adherence to prescribed antihypertensive regimen should be given. A history of target organ damage (i.e. retinopathy, nephropathy, claudication, heart disease, abdominal or carotid artery disease) and the overall cardiovascular risk status should also be explored in detail (28).

The prevalence of resistant hypertension is high: 53% of patients in NHANES had a BP < 140/90 mm Hg vs. 48% in the Framingham Heart Study. In NHANES participants with chronic kidney disease, 37% had a BP < 130/80 mm Hg. In ALLHAT, 34% of patients remained uncontrolled after 5-year follow-up on at least 2 antihypertensive drugs (16). Table 2 presents clinical history, physical exam findings, and routine labs that suggest specific endocrine causes of hypertension.

Table 2: Endocrine Causes of Hypertension. Clinical Presentation. Diagnostic Tools

Etiology

Clinical presentation

Diagnostic tools

Adrenal-dependent causes 

Pheochromocytoma and sympathetic paraganglioma 

Headaches, palpitations, anxiety-like attacks, and profuse sweating

Free plasma or fractionated urinary metanephrines

Primary aldosteronism 

Polyuria, myopathy, and cardiac dysrhythmias may occur in cases of severe hypokalemia

Increased Aldosterone/Renin Ratio. Suppressed PRA, Increased aldosterone. Low potassium.

Congenital adrenal hyperplasia 

 11β-Hydroxylase deficiency 

Androgen production is increased and may lead to prenatal virilization with resulting pseudohermaphroditism in females. Males may develop pseudoprecocious puberty, short stature, and sometimes prepubertal gynecomastia

Increased 17 OH PRG, DOC, 11-deoxycortisol, androstenedionetestosterone, and DHEA-S

Germline mutation testing

 17α-Hydroxylase deficiency 

Pseudohermaphroditism in XY males, and sexual infantilism and primary amenorrhea in females

Low/low normal blood levels of androstenedione, testosterone, DHEA-S, 17-hydroxyprogesterone, aldosterone, and cortisol

Germline mutation testing

Deoxycorticosterone-producing tumor 

Hypertension, adrenal tumors usually large and malignant. Women may present virilization and men feminization.

Low renin and low/normal aldosterone. Increased DOC

Chrousos syndrome

Children may present with ambiguous genitalia and precocious puberty. In women, hirsutism and oligo-amenorrhea. Men may be infertile and/or oligospermic. No features of Cushing’s syndrome. Hypertension.

Hypokalemic alkalosis

Increased DOC, costisol, ACTH. Increased adrenal androgen secretion.

Liddle syndrome

Hypertension and spontaneous hypokalemia

Low potassium and low levels of aldosterone and renin

Cushing syndrome 

Weight gain, insomnia, depression, easy bruising, fatigue, acne, hirsutism, hyperglycemia

24-h urinary free cortisol excretion on at least 2 occasions. Suppressed ACTH

1mg overnight dexamethasone suppression test

Apparent mineralocorticoid excess/11β-hydroxysteroid dehydrogenase deficiency 

Congenital: Growth retardation/short stature, hypertension, hypokalemia, diabetes insipidus renalis, and nephrocalcinosis polyuria and polydipsia.

Acquired form is  attributed to licorice root ingestion and presents with hypertension and hypokalemia

Hypokalemia, metabolic alkalosis, low renin, low aldosterone, normal plasma cortisol levels

Abnormal urinary cortisol-cortisone metabolite profile

Parathyroid-dependent causes 

  Hyperparathyroidism 

Hypercalcemia, hypercalciuria, nephrocalcinosis, cortical bone loss, proximal myopathy, weakness and easy fatigability, depression, inability to concentrate

PTH intact, increased serum calcium concentration

Pituitary-dependent causes 

 Acromegaly 

Enlargement of the lower lip and nose, prognathism, mild hirsutism (in women), sweating, oily skin, diabetes mellitus, acanthosis nigrigans

IGF-1

 Cushing's disease

Weight gain, insomnia, depression, easy bruising, fatigue, acne, hirsutism, hyperglycemia

24-h urinary free cortisol excretion on at least 2 occasions

High normal/increased plasma ACTH

1mg overnight dexamethasone suppression test

MRI pituitary

Thyroid-dependent causes 

 Hypothyroidism 

Fatigue, weight gain, bradycardia, loss of appetite

Increased TSH

Low FT3, FT4

 Hyperthyroidism 

Nervousness, anxiety, palpitations, hyperactivity, weight loss, tachycardia

Low TSH

Increased FT3, FT4

 

PRIMARY ALDOSTERONISM

Prevalence of Primary Aldosteronism

In a community-based study (Framingham Offspring) comprising 1688 nonhypertensive participants, increased plasma aldosterone concentrations within the physiologic range predisposed persons to the development of hypertension (29). Previous studies have reported a prevalence of primary aldosteronism (PA) of 1-2 %. Newer data suggest an overall prevalence of >5% and possibly > 10% among the hypertensive population (15,16). In patients with mild to moderate hypertension without hypokalemia, the prevalence of PA has been reported to be 3% (30). In patients with resistant hypertension, the prevalence ranges between 17 and 23 % (31). In a study involving 1616 patients with resistant hypertension, 21% (338 pts) had an Aldosterone/Renin Ratio of > 65 with concomitant plasma aldosterone concentrations of > 416 pmol/L (15 ng) (25). After salt suppression testing, only 11% (182 pts) of these patients had primary aldosteronism (25). In patients with adrenal incidentaloma and hypertension, the prevalence of aldosteronism is low at 2% (31). Many (up to 63%) patients with PA may not present with hypokalemia but are rather normokalemic (31,32). Low renin hypertension is not always easy to differentiate from PA (33). Born-Frontsberg and colleagues found that 56% of 553 patients with primary aldosteronism had hypokalemia and 16% had cardio-and cerebrovascular comorbidities (32). In addition to the patient group with resistant hypertension, screening for primary aldosteronism is recommended for those patients with diuretic-induced or spontaneous hypokalemia, those with hypertension and a family history of early-onset hypertension or cerebrovascular accident at young age, and those with hypertension and an adrenal incidentaloma (27,34).

Etiology of Primary Aldosteronism

PA can be a sporadic or familial condition. Many cases of sporadic PA are caused by an aldosterone-producing adrenal adenoma. However, bilateral zona glomerulosa hyperplasia is much more common in apparently sporadic primary hyperaldosteronism than previously thought and is an important differential diagnosis, since it is treated medically with aldosterone antagonists, rather than by adrenalectomy (35). Selective use of adrenal venous sampling is helpful in this setting (36,37). Very rarely, PA can be caused by an adrenal carcinoma, or unilateral adrenal cortex hyperplasia (also called primary adrenal hyperplasia) (36).

Familial aldosteronism is estimated to affect 2% of all patients with primary hyperaldosteronism and is classified as type 1, type 2, type 3, and type 4 (38–40). Patients with familial aldosteronism type 3 produce amounts of 18-OHF and 18-oxoF 10-1,000 times higher than patients with familial aldosteronism type 1 (approx. 20 times normal) or patients with familial aldosteronism type 2 or sporadic aldosteronism (41). Patients with familial aldosteronism type 3 have a paradoxical rise of aldosterone after dexamethasone, atrophy of the zona glomerulosa, diffuse hyperplasia of the zona fasciculata, and severe hypertension in early childhood (around age 7 years) that is resistant to drug therapy but curable by bilateral adrenalectomy (42).

In familial hyperaldosteronism type 1, an autosomal dominantly inherited chimeric gene defect in CYP11B1/CYPB2 (coding for 11beta-hydroxylase/aldosterone synthase) causes ectopic expression of aldosterone synthase activity in the cortisol-producing zona fasciculata, making mineralocorticoid production regulated by corticotropin (24,43). The hybrid gene has been identified on chromosome 8. Under normal conditions, aldosterone secretion is mainly stimulated by hyperkalemia and angiotensin II. An increase of serum potassium of 0.1 mmol/L increases aldosterone by 35%. In familial hyperaldosteronism type 1 or glucocorticoid-remediable aldosteronism, urinary hybrid steroids 18-oxocortisol and 18-hydroxycortisol are approx. 20-fold higher than in sporadic aldosteronomas. Intracranial aneurysms and hemorrhagic stroke are clinical features frequently associated with familial hyperaldosteronism type 1 (35). The diagnosis is made by documenting dexamethasone suppression of serum aldosterone using the Liddle’s Test (dexamethasone 0.5 mg q 6h for 48h should reduce plasma aldosterone to nearly undetectable levels (below 4 ng/dl) or by genetic testing (Southern Blot or PCR) (44). In contrast, familial hyperaldosteronism type 2 is not glucocorticoid-remediable and is caused by mutations in the inwardly rectifying chloride channel CLCN2 (42).

Familial aldosteronism type 3 is caused by heterozygous gain-of-function mutation in the potassium channel GIRK4 (encoded by KCNJ5) leading to an increase in aldosterone synthase expression and production of aldosterone (35). Familial aldosteronism type 4 results from germline mutations in the T-type calcium channel subunit gene CACNA1H (45). Germline mutations in CACNA1D (encoding a subunit of L-type voltage-gated calcium channel CaV1.3) are found in patients with primary aldosteronism sometimes associated with seizures, and neurological abnormalities (46) . Table 3 shows genetic and clinical characteristics of familial aldosteronism.

Table 3. Classification of Familial Hyperaldosteronism

Type

Gene Mutation

Treatment

Clinical manifestations

FH-1

CYP11B2/CYP11B1 Chimeric

Low-dose dexamethasone

Intracranial aneurysms and hemorrhagic stroke

FH-2

CLCN2 (R172Q, M22K, G24D, S865R, Y26N)

MRA

Primary aldosteronism

FH-3

KCNJ5 (T158A, I157S, E145Q)

Bilateral adrenalectomy

Primary aldosteronism

 

KCNJ5 (G151E, Y152C)

MRA

Primary aldosteronism

FH-4

CACNA1H (M1549V, S196L, P2083L, V1951E)

MRA

Primary aldosteronism

 

Diagnosis – Screening and Confirming Tests

Primary aldosteronism is screened for by measuring plasma aldosterone (PA) and plasma renin activity (PRA) or direct renin concentration. There are various assays for measuring aldosterone, which can prove to be problematic(47,48). Measuring PRA is complicated and includes generating angiotensin from endogenous angiotensinogen. Quantification of renin’s conversion of angiotensinogen to angiotensin is performed utilizing radioimmunoassays for PRA, which are not standardized among laboratories. Measuring plasma renin molecules directly by an automated chemiluminescence immnoassay as direct renin concentration also is feasible. A PA/PRA-ratio > 30 with a concomitant PA > 20 ng/dl has a sensitivity of 90% and specificity of 91% for primary aldosteronism. Because low renin hypertension can be difficult to distinguish from PA, an upright plasma aldosterone of at least 15 ng/dl may be helpful (30).

As hypokalemia can reduce aldosterone secretion, it should be corrected before further diagnostic work-up. Also, if a patient with hypertension treated with an ACE inhibitor or ARB, calcium channel blocker, and a diuretic (all of which should increase PRA, thereby lower the PA/PRA-ratio or ARR), still has a suppressed renin and 2-digit plasma aldosterone level, primary aldosteronism is likely. False-positive ARRs may occur in premenopausal women during the luteal phase of the menstrual cycle as well as in those who are on medication with estrogen-containing contraceptive agents (14). Because of medication interference, it is commonly recommended to withdraw betablockers, ACE inhibitors, ARBs (angiotensin receptor blockers), renin inhibitors, dihydropyridine calcium channel blockers, nonsteroidal anti-inflammatory drugs, and central alpha 2-agonists approx. 2 weeks before PA/PRA-ratio or ARR testing, and to hold spironolactone, eplerenone, amiloride, and triamterene, and loop diuretics approx. 4 weeks before ARR testing. Licorice root products should also be withheld 4 weeks prior to testing (49). Confirmatory testing can be done by different techniques (31,50) [Table 4(50)]. A study including 148 hypertensive patients found that a new overnight diagnostic test using pharmaceutical renin-angiotensin-aldosterone system blockade with dexamethasone, captopril and valsartan, has low cost, is rapid, safe and easy to perform with an estimated sensitivity of 98% and specificity of 100% (51).

To clinically distinguish hyperplasia from unilateral adenoma, imaging with computed tomography and magnetic resonance imaging are helpful.

Table 4. ConfirmatoryTests (50)

Confirmation Method

Protocol

Interpretation of Results

Oral Salt Suppression Test

·Increase sodium intake for 3-4 days via supplemental tablets or dietary sodium to >200 mmol/day
· Monitor blood pressure
· Provide potassium supplementation to ensure normal serum levels
· Measure 24h urinary aldosterone excretion and urinary sodium on 3rd or 4th day

· PA confirmed: if 24h urinary aldosterone excretion >12 mcg in setting of 24h sodium balance >200 mmol
· PA unlikely: if 24h urinary aldosterone excretion <10mcg

Intravenous Saline Infusion Test

· Being infusion of 2L of normal saline after patient lies supine for 1 hour.
· Infuse 2L of normal saline over 4 hours (500 mL/h)
· Monitor blood pressure, heart rate, potassium
· Measure plasma renin and serum aldosterone at time=0h and time=4h

· PA confirmed: 4h aldosterone level > 10 ng/dL
· PA unlikely: 4h aldosterone level < 5 ng/dL

Captopril Challenge Test

· Administer 25-50mg of captopril in the seated position
· Measure renin and aldosterone at time=0h and again at time=2h
· Monitor blood pressure

· PA confirmed: serum aldosterone high and renin suppressed*
· PA unlikely: renin elevated and aldosterone suppressed*
*varying interpretations without specific validated cut-offs

Fludrocortisone Suppression Test

· Administer 0.1 mg fludrocortisone q6h for 4 days
· Supplement 75-100 mmol of NaCl daily to ensure a urinary sodium excretion rate of 3 mmol/kg/body weight
· Monitor blood pressure
· Provide potassium supplementation to ensure normal serum levels
· Measure plasma renin and serum aldosterone in the morning of day 4 while seated

· PA confirmed: Seated serum aldosterone > 6 ng/dL on day 4 with PRA< 1ng/mL/h
· PA unlikely: suppressed aldosterone < 6 ng/dL

Oral Salt Suppression Test

·Increase sodium intake for 3-4 days via supplemental tablets or dietary sodium to >200 mmol/day
· Monitor blood pressure
· Provide potassium supplementation to ensure normal serum levels
· Measure 24h urinary aldosterone excretion and urinary sodium on 3rd or 4th day

· PA confirmed: if 24h urinary aldosterone excretion >12 mcg in setting of 24h sodium balance >200 mmol
· PA unlikely: if 24h urinary aldosterone excretion <10mcg

Intravenous Saline Infusion Test

· Being infusion of 2L of normal saline after patient lies supine for 1 hour.
· Infuse 2L of normal saline over 4 hours (500 mL/h)
· Monitor blood pressure, heart rate, potassium
· Measure plasma renin and serum aldosterone at time=0h and time=4h

· PA confirmed: 4h aldosterone level > 10 ng/dL
· PA unlikely: 4h aldosterone level < 5 ng/dL

 

Localization

Despite imaging studies, adrenal venous sampling (AVS) with cosyntropin (ACTH) infusion is often essential if the patient desires surgery in case of a unilateral adenoma: cutoff for unilateral adenoma > 4 “cortisol-corrected” aldosterone ratio (adenoma side aldosterone/cortisol: normal adrenal gland aldosterone/cortisol); cutoff for bilateral hyperplasia < 3 “cortisol-corrected” aldosterone ratio (high-side aldosterone/cortisol: low-side aldosterone/cortisol) (36,52).

Medical Treatment

The 2016 Endocrine Society clinical practice guideline for the management of primary aldosteronism suggests that patients with hypertension, spontaneous hypokalemia, undetectable renin, and a plasma aldosterone concentration above 20 ng/dl (550 pmol/L) may not need to undergo further confirmatory testing but instead proceed with further imaging and/or adrenal vein sampling or (if unable or unwilling to undergo surgery/adrenalectomy) treatment with a mineralocorticoid antagonist (36). Bilateral adrenal hyperplasia is treated with spironolactone, eplerenone, and/or amiloride (50).

Spironolactone is a nonselective, competitive mineralocorticoid receptor antagonist and is generally considered first-line therapy for patients with BAH at doses ranging between 12.5-400 mg/d (usually 12.5-50 mg/d). It also acts as antagonist of the androgen receptor, a weak antagonist of the glucocorticoid receptor, and an agonist of the progesterone receptor. These actions are associated with adverse effects, including hyperkalemia, hyponatremia, gynecomastia, menstrual disturbances and breast tenderness and decreased libido in women, and gynecomastia in men, occuring in a dose-dependent manner.

Eplerenone, is a more expensive but selective mineralocorticoid receptor blocker with fewer antiandrogenic effects, but also with lower affinity for the mineralocorticoid receptor and less effectiveness than spironolactone with respect to BP lowering in patients with moderate hypertension (53); Generally, higher doses of eplerenone are prescribed for similar effects as spironolactone (usually 25-50 mg twice daily) (50).

Currently under investigation are aldosterone synthase inhibitors, which may not have any nongenomic/non-mineralocorticoid receptor-mediated adverse effects (54). In cases of familial hyperaldosteronism type 1, dexamethasone is effective in suppressing ACTH and, hence, aldosterone overproduction (55).

Surgical Treatment

Adrenal adenomas producing aldosterone should be removed. Nearly all patients with such endocrine hypertension have improved blood pressure control and up to 60% are cured (normotensive without antihypertensive therapy) from hypertension (56–59). This outcome is influenced by various factors including age, duration of hypertension, coexistence of renal insufficiency, use of more than 2 antihypertensive drugs preoperatively, family history of hypertension, and others. Parameters of insulin sensitivity can be restored to normal with treatment of PA (60). A cross-sectional study including 460 pts with primary aldosteronism and 1363 controls with essential hypertension found no significant difference between pre- and postoperative levels of fasting plasma glucose and serum lipids (61). This topic has been extensively reviewed from a pro and contra perspective. If a patient does not desire surgery/adrenalectomy for a unilateral aldosteronoma/hyperplasia (Figure 3), medical therapy should be initiated (54). AVS and CT/MRI of the adrenal glands show a unilateral abnormality in 60.5% and 56%, respectively, but were congruent on the involved side in the same patient in only 37% in a recent systematic review (62). If a patient is older than age 40 years, the risk for an adrenal incidentaloma increases (62). Unilateral adrenalectomy can be helpful in some patients with primary aldosteronism and bilateral adrenal hyperplasia (56).

Figure 3. Conn adenoma. Appearance of a 1 cm right-sided adrenal nodule (arrow) on contrast-enhanced computed tomography in a middle-aged man with hypertension treated for 20 years, initially only with a betablocker before becoming medically refractory and hypokalemic with inappropriate kaliuresis. After laparoscopic right adrenalectomy, the patient required only one antihypertensive drug to control his blood pressure.

PHEOCHROMOCYTOMA (PPGLS)

Prevalence

Pheochromocytomas are rare neoplasms, with an estimated occurrance of approximately 0.2 percent of patients with hypertension. It has been reported that the annual incidence of pheochromocytoma is nearly 0.8 per 100,000 person-years (63). Pheochromocytomas may occur at any age, however they are commonly presented  in the fourth to fifth decade (64) .

Etiology

Pheochromocytoma and paragangliomas (PPGLs) rare neuroendocrine tumors are composed of chromaffin tissue containing neurosecretory granules (65). Most pheochromocytomas are sporadic but as of known today, approx. 40% of patients with pheochromocytoma or paraganglioma irrespective of age at onset and family history harbor a germline mutation (66,67). At present, there are 10 currently clinically relevant syndromes known: multiple endocrine neoplasia type 2, von Hippel-Lindau syndrome, neurofibromatosis type 1, paraganglioma syndromes 1 through 5, caused by mutations of the succinate dehydrogenase genes SDHD (syndrome 1), SDHAF2 (syndrome 2), SDHC (syndrome 3), SDHB (syndrome 4), and SDHA (syndrome 5), and the hereditary pheochromocytoma syndromes resulting from germline mutations in the genes coding transmembrane protein 127 (TMEM127) and MYC-associated factor X (MAX). Further susceptibility genes include EGLN1 (PHD2), EGLN2 (PHD1), DNMT3A, IDH1, FH, MDH2, SLC25A11, KIF1B, and HIF2A (68–70). There is controversy when genetic testing should be obtained in patients with pheochromocytoma, especially considering cost effectiveness.

Clinical Features

The clinical presentation of patients with PPGLs shows a wide variety from no or minor symptoms, to dramatic life-threatening manifestations. Asymptomatic patients present mostly incidentally discovered adrenal masses. Normotensive patients may also have sporadic pheochromocytomas (71). It appears that approx. 15% of patients with pheochromocytoma are normotensive (19,21). The classic triad of pounding headache, profuse sweating, and palpitations occurs sporadically with a duration from several minutes to 1 hour. Paroxysmal hypertension occurs commonly in 35-50% of patients. The patients show a complete relief of symptoms between episodes. The high BP surges and the other symptoms are associated with the underlying tumoral catecholamine release, which is the major cause for the high prevalence of cardiovascular emergencies, such as myocardial infarction, stroke, and heart failure. Pheochromocytoma (PHEO) and PPGLs may be the prevalent cause of acute Takotsubo-like catecholamine cardiomyopathy (TLC) (72–75). This association has been reported in in up to 3% of patients with secreting PPGL. The real prevalence of PPGL in TTC remains to be determined. The biochemical profile of pheochromocytomas associated with the a forementioned hereditary syndromes varies (76). Patients with MEN 2 and VHL syndrome may have clinically “silent” pheochromocytomas. Blood pressure does not correlate with circulating catecholamines in patients with pheochromocytoma. Sipple syndrome for multiple endocrine neoplasia type 2 first described by Max Schottelius and Felix Fraenkel in 1886 (77).

Diagnosis – Screening

The diagnosis can be established by measuring free plasma or fractionated urinary metanephrines (metanephrine and normetanephrine) (22). When plasma free metanephrines cannot be measured by HPLC with electrochemical detection or high-throughput automated liquid–chromatography-tandem mass spectrometry (LC-MS/MS), measuring plasma free metanephrines by RIA or measuring plasma chromogranin A may represent good markers for pheochromocytoma. In rare circumstances, pheochromocytomas release large O-methylated dopamine metabolite methoxytyramine, which can be elevated in extra-adrenal tumor location (in particular, neck and skull-base paragangliomas) and the presence of metastatic disease (78). In patients with renal failure, plasma concentrations of free metanephrines can be increased several folds (79). For optimal diagnostic accuracy, established reference values for plasma free and 24-hour urinary fractionated metanephrines should be btained, according to age and sex. The upper cutoff level of plasma free normetanephrine, but not for metanephrine or methoxytyramine is higher in older patients (80).

Several medications can cause false-positive biochemical testing. Thus, plasma normetanephrine levels may increase in patients treated with tricyclic antidepressants, antipsychotics, buspirone, MAO inhibitors, sympathomimetics, cocaine, levodopa, phenoxybenzamine, acetaminophen, alpha- methyldopa, and sulphasalazine. Plasma metanephrine levels may increase in patients treated with buspirone, MAO inhibitors, sympathomimetics, cocaine, and levodopa. Urine normetanephrine levels may be higher in patients receiving all the above-mentioned substances, as well as labetalol and sotalol. Urine normetanephrine levels may be increased by buspirone, MAO inhibitors, sympathomimetics, cocaine, levodopa, labetalol and sotalol (14). People who eat biogenic amines may have false-positive urinary metanephrine results. However, for measuring plasma free metanephrines and the O-methylated dopamine metabolite methoxytyramine, no specific dietary requirements are needed, but fasting state (14,81).

The 2014 Endocrine Society clinical practice guideline recommends that all patients with pheochromocytoma-paraganglioma should be engaged in shared decision making for genetic testing (22). All patients with paraganglioma should undergo testing for succinate dehydrogenase (SDH) mutations and those patients with metastatic disease should be tested for SDHB mutations. Recognizing the distinct genotype-phenotype presentations of patients with hereditary tumors, the guideline recommends a personalized approach to patient management. Of note is that SDHD and SDHAF2 are maternally imprinted and therefore one or more generations can be skipped. During the first 2 decades of life (before the age of 20 years), the most common hereditary pheochromocytoma-paraganglioma syndromes found are related to von Hippel Lindau disease, paraganglioma syndrome type 4 (SDHB), and neurofibromatosis type 1. Pheochromocytomas related to multiple endocrine neoplasia type 2 occur most frequently between the third and fifth decade of life and should first be considered in a patient presenting with bilateral pheochromocytomas. The mean penetrance of pheochromocytoma or paraganglioma in individuals carrying a RET germline mutation is 50% by the age of 44 years (82).

Approximately 35% of extra-adrenal pheochromocytomas are considered “malignant” (metastasizing) as opposed to approximately 10% of those arising in the adrenal gland. The 2017 WHO classification of endocrine tumors replaced the term “malignant” with “metastatic”. The risk for metastases increases when the tumor exceeds 5 cm in size and when there is a germline mutation in the SDHB gene (83,84).

Localization

CT or MR imaging can localize the tumor in approx. 95 % of cases. For metastatic pheochromocytomas, 18F-Fluorodopamine and 18F-FDG PET appears to be more helfpul than 123I-MIBG or 131I-MIBG scintigraphy (85,86). In fact, MIBG scintigraphy should nowadays only been used in selected patients (85,87). Many medications can interfere with 123I-MIBG or 131I-MIBG uptake (for instance, calcium channel blockers, antipsychotics) and should be discontinued before the scan/imaging. The 2014 Endocrine Society guideline recommends the use of 123I-MIBG in patients with metastatic pheochromocytoma-paraganglioma when radiotherapy with 131I-MIBG is planned and occasionally in some patients with an increased risk for metastatic disease (large tumor size, extra-adrenal tumor, multifocal or recurrent disease) (22). For patients with head and neck paragangliomas, 111In-octreotide has a very good sensitivity (88). Newer functional imaging techniques such as 68Ga-labeled 1,4,7,10-tetraazacylododecane-1,4,7,10-tetraacetic acid-octreotate (DOTATATE) of 18F-labeled L-dihydroxyphenylalanine (L-DOPA) have excellent resolution in detecting pheochromocytomas and paragangliomas.

Medical Treatment

The Endocrine Society, the American Association for Clinical Chemistry, and the European Society of Endocrinology have released clinical practice guidelines recommended preoperative blockade of hormonally functional PPGL to prevent cardiovascular complications, along with medication for normalization of blood pressure as well as heart rate. Alpha-adrenergic blockade (i.e., doxazosin, prazosin or terazosin) followed by a β-adrenergic blockade (i.e., propranolol, atenolol) is recommended for preoperative preparation (89). It is also suggested to administer high-sodium diet and fluid intake to prevent low blood pressure after surgery. Approx. 50% of patients with metastatic pheochromocytomas respond to 131I-MIBG therapy by partial remission or at least stable disease. Selective alpha1 blocking agents, such as prazosin (Minipress), terazosin (Hytrin), and doxazosin (Cardura), have more favorable adverse effect profiles and are used when long-term therapy is required (metastatic pheochromocytoma). Newer therapy options of metastatic pheochromocytoma-paraganglioma include 90Y-DOTATATE and 177Lu-DOTATATE. Chemotherapy is usually administered according to the so-called Averbuch protocol from 1988. New therapies may include tyrosine kinase inhibitors in selected patients (90).

Surgical Treatment

For tumors exceeding 5 cm in size, open adrenalectomy has long been considered the suggested procedure for tumor removal rather than laparoscopic or retroperitoneoscopic minimally invasive tumor removal, to ensure complete tumor resection, prevent tumor (capsule) rupture, and avoid local recurrence (22) (Figure 4 and 5)  

Figure 4. Computed tomography showing recurrence of a right adrenal pheochromocytoma. unpublished observation in a patient with MEN2-related bilateral pheochromocytomas and unilateral tumor recurrence 11 years after bilateral adrenalectomy, photo: courtesy of Prof. Andrea Tannapfel).

Figure 5. Macroscopic photo of a right adrenal pheochromocytoma removed from the above patient with multiple endocrine neoplasia type 2.

For pheochromocytomas less than 6 cm, a recent cohort study from a multicenter consortium-based registry for 625 patients treated for bilateral pheochromocytomas between 1950 and 2018 compared patients undergoing total vs. cortical-sparing adrenalectomy and found that patients undergoing cortical-sparing adrenalectomy did not demonstrate decreased survival, despite development of recurrent pheochromocytoma in 13%. The authors recommend cortical-sparing adrenalectomy should be considered in all patients with hereditary pheochromocytoma (91). A retrospective, multicenter, international study in patients carrying the Met918Thr RET variant with no age restrictions who were followed from 1970 to 2016 based on registry data from 48 centers globally, found that adrenal-sparing surgery in multiple endocrine neoplasia type 2B can preserve normal adrenal function. In that study, three (10%) of the 31 patients in whom adrenal-sparing surgery had been performed, developed long-term recurrence, while normal adrenal function was mantained in 16 (62%) of patients (92). Apparently one third of one functioning adrenal gland is sufficient for normal glucocorticoid and mineralocorticoid secretion (93).

Cushing’s Syndrome

Hypercortisolemia is associated with hypertension in approximately 80% of adult cases and half of children (94,95). A workshop consensus paper attempted to rationalize the treatment of hypertension in patients with Cushing’s syndrome (95). In patients with Cushing’s disease, night-time blood pressure decline is significantly lower than that in patients with essential hypertension (96). After cure of Cushing’s syndrome, approximately 30% of patients have persistent hypertension (97). In children and adolescents, blood pressure normalization occurs in most patients within a year and seems to be dependent on the degree and duration of presurgical hypercortisolemia (94). In patients with Cushing’s disease, renin and DOC levels are usually normal, whereas in ectopic corticotropin syndrome, hypokalemia is common and related to an increased mineralocorticoid activity with suppressed renin and elevated DOC levels (98).

There are several mechanisms of blood pressure elevation in Cushing’s syndrome: increased hepatic production of angiotensinogen and cardiac output by glucocorticoids, reduced production of prostaglandins via inhibition of phospholipase A, increased insulin resistance, and oversaturation of 11beta-Hydroxysteroid dehydrogenase activity with increased mineralocorticoid effect through stimulation of the mineralocorticoid receptor (99). Screening studies for Cushing’s syndrome include measuring 24-h urinary free cortisol excretion on at least 2 occasions, performing a 1 mg dexamethasone suppression test, checking a midnight salivary cortisol and diurnal rhythm of cortisol secretion, and others listed in the recent Endocrine Society Clinical Practice Guideline (100). Therapy should be directed at removing glucocorticoid excess (101). Hypokalemia (especially in patients with ectopic ACTH production) can be treated with mineralocorticoid receptor antagonists such as spironolactone or eplerenone. Thiazide diuretics may also be helpful.

Given the increasing improvement in imaging and laboratory (assays etc.) techniques/modalities, one can expect an increasing number of incidentally discovered tumors and nodules in various organs including the adrenal glands. The future challenge will be when and to which extent to test individuals for disease conditions (102,103). For those individuals with adrenal incidentalomas but clearly lack of clinical features of Cushing’s syndrome, subclinical hypercortisolism may be detected biochemically depending upon which cutoff values and assays will be used (104). For the latter population, the American Association of Clinical Endocrinologists recommend using a cutoff for (8 AM) serum cortisol of 5 mcg/dl after 1 mg overnight (11 PM) dexamethasone which reveals approx. 58% sensitivity at a 100% specificity (105). A lower cutoff for serum cortisol suppression, i.e. 1.8 mcg/dl, usually rules out Cushing’s syndrome (102). A prospective, randomized study including 45 patients with subclinical hypercortisolism and adrenal incidentalomas was divided into 23 pts who underwent adrenalectomy and 22 pts under surveillance. Monitoring included glycemic control, blood pressure, lipid profile, obesity, and bone mineral density. In the surgical group, diabetes mellitus improved in 62% and hypertension in 67% of pts, whereas the conservative group showed worsening of glycemic control, blood pressure and lipid profiles (37).

To better understand the sequelae of disturbed adrenal hormone synthesis, please refer to Figure 6 and related Endotext chapters (106,107).

Figure 6. Adrenal Steroid Synthesis. Z Glom = zona glomerulosa; Z Fas = zona fasciculata; Z Ret = zona reticularis; 19-H = 19-Hydroxylase; HSD = Hydroxysteroid dehydrogenase; P450aro = aromatase; 5alpha-Red = 5alpha-Reductase. The 3 adrenal cortex zones Z Glom, Z Fas, and Z Ret stand above the “column” of hormones that are produced in the respective zone. The steroidogenic enzymes on the left starting with P450scc (Desmolase) are listed in order for “vertical and horizontal reading”, i.e. Desmolase converts cholesterol to pregnenolone, 3beta-OH-Steroid Dehydrogenase I/II convert pregnenolone to progesterone, 17-OH-Pregnenolone to 17-OH-Progesterone, and P450c11 converts deoxycorticosterone to 18-OH-Corticosterone and 11-Deoxycortisol to cortisol, etc. (modified from ref. 35: Koch CA. Encyclopedia of Endocrine Disease, 2004)

GLUCOCORTICOID RESISTANCE (CHROUSOS SYNDROME)

This autosomal recessive or dominant inherited disorder is rare and caused by inactivating mutations of the glucocorticoid receptor gene (108,109). Cortisol and ACTH are elevated but there are no clinical features of Cushing syndrome. Permanent elevation of ACTH can lead to stimulation of adrenal compounds with mineralocorticoid activity (corticosterone, DOC), along with elevated cortisol secretion may lead to stimulation of the mineralocorticoid receptor, resulting in hypertension. In women, hirsutism and oligo-amenorrhea may develop through stimulation of androgens (androstendione, DHEA, 5-androstendiol). Clinically, children may present with ambiguous genitalia and precocious puberty. Men may be infertile and/or oligospermic. Women may have acne, excessive hair, menstrual irregularities with oligo- anovulation, as well as infertility (108–110).

Treatment entails suppression of ACTH secretion with high doses of dexamethasone (1-3 mg/day). Mineralocorticoid receptor-dependent hypertension may be treated with blockade of the receptor, with spironolactone or eplerenone.

Congenital Adrenal Hyperplasia

11Beta-Hydroxylase Deficiency

The most common cause of congenital adrenal hyperplasia (CAH) is 21-hydroxylase deficiency. Hypertension per se has not been regarded as a component of this syndrome. Recent data have suggested that hypertension may be more prevalent in this patient population than previously thought (111–113).

Approx. 5% of all cases of CAH care caused by 11beta-hydroxylase deficiency. 11beta-hydroxylase is responsible for the conversion of deoxycorticosterone (DOC) to corticosterone (precursor of aldosterone) and 11-deoxycortisol to cortisol. In approximately 2/3 of individuals affected by a deficiency of this enzyme, monogenic low renin hypertension with low aldosterone levels occurs caused by accumulation of 11-deoxycortisol and DOC (114,115). The earliest age of onset of hypertension was reported at birth (116). The inheritance mode is autosomal recessive. The responsible gene CYP11B1 is located on chromosome 8 and is mutated (40,117,118). Since corticotropin (ACTH) is chronically elevated and precursors such as 17-OH progesterone and androstendione accumulate, androgen production is increased and may lead to prenatal virilization with resulting pseudohermaphroditism in females. Males may develop pseudoprecocious puberty, short stature, and sometimes prepubertal gynecomastia (119,120). Usually, glucocorticoid replacement reduces hypertension in these patients. In selected patients, bilateral adrenalectomy may be safe and effective in managing high blood pressure (121).

17Alpha-Hydroxylase Deficiency

This enzyme deficiency is rare and leads to diminished production of cortisol and sex steroids. Chronic elevation of ACTH causes an increased production of DOC and corticosterone with subsequent hypertension, hypokalemia, low aldosterone concentrations with suppressed renin as well as pseudohermaphroditism in XY males (122), and sexual infantilism and primary amenorrhea in females (123,124). Diagnosis may be delayed until puberty. Plasma adrenal androgen levels are low as are cortisol, aldosterone, plasma renin activity, and 17alpha-hydroxyprogesterone. DOC, corticosterone, and 18-hydroxycorticosterone are elevated. Blood pressure is reduced by glucocorticoid replacement. The responsible gene for cytochrome P450C17 is located on chromosome 10q24.

Deoxycorticosterone-Producing Tumor

Deoxycorticosterone-producing tumors are rare adrenal tumors presented mostly large and malignant (125). Along with deoxycortisone, androgens and estrogens may be cosecreted. Women may present virilization and men feminization. Hypertension and hypokalemia may manifestate with rapid onset. Renin and aldosterone are often low.

Apparent Mineralocorticoid Excess

Low-renin hypertension (undetectable aldosterone, hypokalemia) can present in various forms, one of them is apparent mineralocorticoid excess (AME), an autosomal recessive disorder caused by deficiency of the 11beta-hydroxysteroid dehydrogenase type 2 (11beta-HSD2) enzyme (49,126,127). This enzyme converts cortisol to the inactive cortisone in renal tubular cells.

In 1977, New et al. (128) first described this syndrome and in 1995 Wilson et al. (129) first reported that mutations in the 11beta-HSD2 gene located on chromosome 16q22 cause AME. The 11beta-HSD2 enzyme is co-expressed with the mineralocorticoid receptor in renal tubular cells and leads to conversion of cortisol to cortisone (130,131). Cortisone does not bind to the mineralocorticoid receptor. Cortisol and aldosterone bind with equal affinity to the mineralocorticoid receptor, but normal circulating concentrations of cortisol are 100 to 1000 fold higher than those of aldosterone (132). If 11beta-HSD2 is oversaturated or defective, more cortisol will be available to bind to the mineralocorticoid receptor (133). Diminished 11beta-HSD2 activity may be hereditary or acquired. Acquired deficiency of this enzyme may result from inhibition by glycyrrhhetinic acid which may occur with use of licorice, chewing tobacco, and carbenoloxone. In childhood, AME often causes growth retardation/short stature, hypertension, hypokalemia, diabetes insipidus renalis, and nephrocalcinosis. Diminished 11beta-HSD2 activity may play a role in the pathogenesis of preeclampsia (134). The diagnosis of AME can be established by measuring free unconjugated steroids in urine (free cortisol/free cortisone ratio), and/or steroid metabolites (tetrahydrocortisol + allotetrahydrocortisol/tetrahydrocortisone) (135). Affected individuals have low renin and aldosterone levels, normal plasma cortisol levels, and hypokalemia. Treatment of AME consists of spironolactone, eplerenone, triamterene, or amiloride. Renal transplant is an option for patients with advanced renal insufficiency.

Constitutive Activation of The Mineralocorticoid Receptor (Geller Syndrome)

The Mineralocorticoid (MC) receptor can be mutated leading to the onset of hypertension before age 20 (136). In vitro experiments demonstrate that progesterone and spironolactone, usually antagonists of the (MC) receptor, become agonists in Geller syndrome, suggesting “gain of function” mutations in the MC gene on chromosome 4q31. The inheritance pattern is autosomal-dominant.

Liddle Syndrome

In 1963, Liddle (137) described patients with severe hypertension, hypokalemia, and metabolic alkalosis, who had low plasma aldosterone levels and plasma renin activity. An improvement of the hypertension occurred after salt restriction and triamterene therapy. Spironolactone is ineffective in this autosomal-dominant inherited syndrome. So-called “gain of function” mutations in the genes coding for the beta- or gamma-subunit of the renal epithelial sodium channel, located at chromosome 16p13, lead to constitutive activation of renal sodium reabsorption and subsequent volume expansion. The 24-h urine cortisone/cortisol ratio is normal.

Pseudohypaldosteronism Type 2

Pseudohypoaldosteronism type 2 or Gordon’s syndrome (138) is a rare Mendelian disorder, transmitted in an autosomal dominant fashion, and can cause low renin hypertension (139). It has an unknown prevalence, since many patients remain undiagnosed. Published families with this condition (hypertension, hyperkalemia, metabolic acidosis, normal renal function, low/normal aldosterone levels) are predominantly from Australia or the United States (138). Hypertension in these patients may develop as a consequence of increased renal salt reabsorption, and hyperkalemia ensues as a result of reduced renal K excretion despite normal glomerular filtration and aldosterone secretion (140). The reduced renal secretion of potassium makes this condition look like an aldosterone-deficient state, thus the term “pseudohypoaldosteronism”.

These features are chloride dependent. Infusion of sodium chloride instead of sodium bicarbonate corrects the abnormalities, as does the administration of thiazide diuretics, which inhibit salt reabsorption in the distal nephron. Gordon and coworkers found that all features could be reversed by very strict dietary salt restriction (138). Gordon syndrome is an autosomal, dominantly inherited disorder with genes mapping to chromosomes 1, 12, and 17 (141,142). Mutations have been identified in WNK kinases WNK1 and WNK4 on chromosomes 12 and 17, respectively (141,143). Abnormalities such as activating mutations in the amiloride-sensitive sodium channel of the distal renal tubule are responsible for the clinical phenotype (144,145). Severe dietary salt restriction, antihypertensives, with preferably use of thiazide diuretics, can control the hypertension in this syndrome. Interestingly, common variants in WNK1 contribute to blood pressure variation in the general population (146).

Insulin Resistance

The metabolic syndrome is characterized by hypertension, abdominal/visceral obesity, dyslipidemia, and insulin resistance (147). At least 24% of adults in the United States meet the criteria for the diagnosis of metabolic syndrome, and this number may even be higher for individuals over the age of 50 years (148). Insulin resistance is significantly associated with hypertension in Hispanics and can cause vascular dysfunction (16,149). Patients with essential hypertension often are insulin resistant (150). Interestingly, not all insulin resistant patients are obese. Excess weight gain, however, accounts for as much as 70% of the risk for essential hypertension and also increases the risk for end stage renal disease (16). In insulin-sensitive tissues, insulin can directly stimulate the calcium pump leading to calcium loss from the cell (151). In an adipocyte, elevated cytosolic calcium concentrations can induce insulin resistance. In a cell resistant to insulin, the insulin-induced calcium loss from cells would be decreased. With the subsequent increase in intracellular calcium, vascular smooth muscle cells respond more eagerly to vasoconstrictors and thus lead to rising blood pressure. Other mechanisms possibly explaining the association of insulin resistance and hypertension are increased sodium retention and increased activity of the adrenergic nervous system. In obesity, increased production of most adipokines (bioactive peptides secreted by adipose tissue) impacts on multiple functions including insulin sensitivity, blood pressure, lipid metabolism, and others (152,153).

Primary Hyperparathyroidism

Parathyroid hormone levels in hypertensive patients usually are in the normal range and appropriate for the serum calcium concentration. However, patients with essential hypertension excrete more calcium compared to normotensive people, suggesting an enhanced parathyroid gland function (154). When infused, PTH is a vasodilator, although chronic infusion of PTH raises blood pressure in healthy subjects (155,156). High-calcium intake may lower blood pressure (157,158). However, hypercalcemia is associated with an increased incidence of hypertension (1). In patients with primary hyperparathyroidism, hypertension is observed in approximately 40% of cases. The mechanisms of these observations/associations are unclear. Hypertension is usually not cured or better controlled after parathyroidectomy (159). In patients with asymptomatic primary hyperparathyroidism, surgery/parathyroidectomy did not show any benefit regarding blood pressure or quality of life when compared to medical management (160). On the other hand, severe hypertension may improve in patients with primary hyperparathyroidism who undergo parathyroidectomy. Arterial stiffness measured in the radial artery seems to be increased in patients with mild primary hyperparathyroidism (161). Patients with primary hyperparathyroidism have carotid vascular abnormalities (162). In normotensive patients with primary hyperparathyroidism, SBP variability is increased and is reduced by parathyroidectomy (163,164). Furthermore, parathyroidectomy in patients with primary hyperparathyroidism may decrease risk of cardiovascular diseases by lowering total cholesterol levels, although ambulatory diastolic BP increases in response to surgery (165). Another contributory factor to hypertension in patients with primary HPT may be endothelial dysfunction (166). In MEN syndromes, hypertension in patients with hyperparathyroidism may be related to an underlying pheochromocytoma or primary aldosteronism. Criteria for parathyroidectomy have recently been revisited at the Fourth International Workshop on the management of asymptomatic primary hyperparathyroidism, including now skeletal and/or renal involvement (nephrocalcinosis on imaging) (167).

Hyperthyroidism

Hyperthyroidism increases systolic blood pressure by increasing heart rate, decreasing systemic vascular resistance, and raising cardiac output (168–171). In thyrotoxicosis, patients usually are tachycardic and have high cardiac output with an increased stroke volume and elevated systolic blood pressure (172,173). Approx. one third of patients with hyperthyroidism have hypertension which often resolves after achieving euthyroidism (174). Subclinical hyperthyroidism may contribute to left ventricular hypertrophy and thereby lead to hypertension , although it has not yet been found to be associated with hypertension (174).

Hypothyroidism

Hypothyroid patients have impaired endothelial function, increased systemic vascular resistance, extracellular volume expansion, and an increased diastolic blood pressure (89,171,175). Hypothyroid patients have higher mean 24-h systolic blood pressure and BP variability on 24-h ambulatory BP monitoring (176). In 32% of hypertensive hypothyroid patients, replacement therapy with thyroxine leads to a fall in diastolic blood pressure to 90 mm Hg or less (177). There is a positive association between serum TSH and blood pressure within the normal serum TSH range, statistically significant for diastolic hypertension (177). Subclinical hypothyroidism may or may not to be associated with hypertension. Hypothyroidism can lead to volume-dependent blood pressure elevation with low plasma renin concentrations (178–180).

Acromegaly

The prevalence of hypertension in patients with growth hormone excess is approximately 46% and more frequent than in the general population (181,182). Growth hormone has antinatriuretic actions and may lead to sodium retention and volume expansion (181,182). Increased systolic output and high heart rate as manifestations of a hyperkinetic syndrome may lead to congestive heart failure (181,183). Blood pressure values are increased in patients with acromegaly associated with reduced glucose tolerance or diabetes compared to those with normal glucose tolerance (181). The RAAS system appears to be implicated in the pathogenesis of hypertension in patients with growth hormone excess (181,183–185). Comorbidities in acromegalics, such as hypertension, hyperlipidemia, diabetes mellitus, and cardiomyopathy, all may improve even with partial biochemical control of growth hormone excess (184,186). However, in some patients, hypertension and diabetes mellitus may persist after attempting biochemical cure/remission (187).

Other Potential Endocrine Conditions Causing Endocrine Hypertension

There is accumulating evidence that vitamin D deficiency may be linked to an increased cardiovascular risk and hypertension (188). Potential mechanisms in this setting are concurrent insulin resistance and direct vitamin D action through the renin-angiotensin-aldosterone system (Figure 7).

Figure 7. Pathway of vitamin D metabolism and its relationship with PTH and the renin-angiotensin-aldosterone system (modified from Ullah et al., 2009)(188)

Testosterone deficiency is frequently identified in obese individuals and those with diabetes mellitus and/or metabolic syndrome including hypertension. Replacement therapy in selected patients may be beneficial not only related to their symptomatology of androgen deficiency such as low libido, poor erections, fatigue, and others, but also in regards to their metabolic profile and blood pressure (189,190).

Similarly, individuals with growth hormone deficiency may be at risk for developing hypertension, mostly because of their body composition being more “fat” and “inflamed” when compared to subjects with growth hormone sufficiency, as assessed by serum IFG-1 levels matched to gender and age. The key in such patients will be to replace them with growth hormone individually to an IGF-1 level at which no features of growth hormone excess develop and to increase physical activity. In obese subjects who are willing to take on major lifestyle changes with the goal to lose weight, eat and live healthier, temporary medication assistance (phentermine, topiramate, liraglutide, lorcaserin, orlistat, naltrexone-bupropion) including administration of growth hormone may be acceptable (191–193).

Individual tissue-dependent sensitivity of the glucocorticoid receptor and actions of endogenous glucocorticoids may play a major role in the development of hypertension, obesity, and diabetes mellitus (119,194,195).

Similarly, individuals with growth hormone deficiency may be at risk for developing hypertension, mostly because of their body composition being more “fat” and “inflamed” when compared to subjects with growth hormone sufficiency, as assessed by serum IFG-1 levels matched to gender and age. The key in such patients will be to replace them with growth hormone individually to an IGF-1 level at which no features of growth hormone excess develop and to increase physical activity. In obese subjects who are willing to take on major lifestyle changes with the goal to lose weight, eat and live healthier, temporary medication assistance (phentermine, topiramate, liraglutide, lorcaserin, orlistat, naltrexone-bupropion) including administration of growth hormone may be acceptable (191–193).

Individual tissue-dependent sensitivity of the glucocorticoid receptor and actions of endogenous glucocorticoids may play a major role in the development of hypertension, obesity, and diabetes mellitus (119,194,195).

REFERENCES

  1. Fields LE, Burt VL, Cutler JA, Hughes J, Roccella EJ, Sorlie P. The burden of adult hypertension in the United States 1999 to 2000: A rising tide. Hypertension 2004;44(4):398–404.
  2. Cutler JA, Sorlie PD, Wolz M, Thom T, Fields LE, Roccella EJ. Trends in hypertension prevalence, awareness, treatment, and control rates in United States adults between 1988-1994 and 1999-2004. Hypertension 2008;52(5):818–827.
  3. Damasceno A, Azevedo A, Silva-Matos C, Prista A, Diogo D, Lunet N. Hypertension prevalence, awareness, treatment, and control in mozambique: Urban/rural gap during epidemiological transition. Hypertension 2009;54(1):77–83.
  4. Hemmelgarn BR, McAllister FA, Myers MG, McKay DW, Bolli P, Abbott C, Schiffrin EL, Grover S, Honos G, Lebel M, Mann K, Wilson T, Penner B, Tremblay G, Tobe SW, Feldman RD, Canadian Hypertension Education Program. The 2005 Canadian Hypertension Education Program recommendations for the management of hypertension: part 1- blood pressure measurement, diagnosis and assessment of risk. Can. J. Cardiol. 2005;21(8):645–56.
  5. O’Shea PM, Griffin TP, Fitzgibbon M. Hypertension: The role of biochemistry in the diagnosis and management. Clin. Chim. Acta. 2017;465:131–143.
  6. Vischer AS, Burkard T. Principles of blood pressure measurement – current techniques, office vs ambulatory blood pressure measurement. In: Advances in Experimental Medicine and Biology.Vol 956. Springer New York LLC; 2017:85–96.
  7. Muntner P, Shimbo D, Carey RM, Charleston JB, Gaillard T, Misra S, Myers MG, Ogedegbe G, Schwartz JE, Townsend RR, Urbina EM, Viera AJ, White WB, Wright JT. Measurement of blood pressure in humans: A scientific statement from the american heart association. Hypertension 2019;73(5):E35–E66.
  8. National High Blood Pressure Education Program B. The Seventh Report of the Joint National Committee on Prevention, Detection, Evaluation, and Treatment of High Blood Pressure.; 2004. Available at: http://www.ncbi.nlm.nih.gov/pubmed/20821851. Accessed December 22, 2019.
  9. Whelton PK, Carey RM, Aronow WS, Casey DE, Collins KJ, Himmelfarb CD, DePalma SM, Gidding S, Jamerson KA, Jones DW, MacLaughlin EJ, Muntner P, Ovbiagele B, Smith SC, Spencer CC, Stafford RS, Taler SJ, Thomas RJ, Williams KA, Williamson JD, Wright JT. 2017 ACC/AHA/AAPA/ABC/ACPM/AGS/APhA/ASH/ ASPC/NMA/PCNA guideline for the prevention, detection, evaluation, and management of high blood pressure in adults: Executive summary: A report of the American college of cardiology/American Heart Association task force on clinical practice guidelines. Hypertension 2018;71(6):1269–1324.
  10. Jordan J, Kurschat C, Reuter H. Arterial hypertension-diagnosis and treatment. Dtsch. Arztebl. Int. 2018;115(33–34):557–558.
  11. Nwankwo T, Yoon SS u., Burt V, Gu Q. Hypertension among adults in the United States: National Health and Nutrition Examination Survey, 2011-2012. NCHS Data Brief 2013;(133):1–8.
  12. Camelli S, Bobrie G, Postel-Vinay N, Azizi M, Plouin PF, Amar L. LB01.11. J. Hypertens. 2015;33:e47.
  13. Gupta-Malhotra M, Banker A, Shete S, Hashmi SS, Tyson JE, Barratt MS, Hecht JT, Milewicz DM, Boerwinkle E. Essential hypertension vs. secondary hypertension among children. Am. J. Hypertens. 2015;28(1):73–80.
  14. Young WF, Calhoun DA, Lenders JWM, Stowasser M, Textor SC. Screening for endocrine hypertension: An endocrine society scientific statement. Endocr. Rev. 2017;38(2):103–122.
  15. Rossi GP, Bernini G, Caliumi C, Desideri G, Fabris B, Ferri C, Ganzaroli C, Giacchetti G, Letizia C, Maccario M, Mallamaci F, Mannelli M, Mattarello MJ, Moretti A, Palumbo G, Parenti G, Porteri E, Semplicini A, Rizzoni D, Rossi E, Boscaro M, Pessina AC, Mantero F. A Prospective Study of the Prevalence of Primary Aldosteronism in 1,125 Hypertensive Patients. J. Am. Coll. Cardiol. 2006;48(11):2293–2300.
  16. Koch CA, Chrousos GP. Endocrine hypertension : underlying mechanisms and therapy. Humana Press; 2013.
  17. Plouin PF, Degoulet P, Tugayé A, Ducrocq MB, Ménard J. [Screening for phaeochromocytoma : in which hypertensive patients? A semiological study of 2585 patients, including 11 with phaeochromocytoma (author’s transl)]. Nouv. Presse Med. 1981;10(11):869–72.
  18. Markou A, Sertedaki A, Kaltsas G, Androulakis II, Marakaki C, Pappa T, Gouli A, Papanastasiou L, Fountoulakis S, Zacharoulis A, Karavidas A, Ragkou D, Charmandari E, Chrousos GP, Piaditis GP. Stress-induced Aldosterone Hyper-Secretion in a Substantial Subset of Patients With Essential Hypertension. J. Clin. Endocrinol. Metab. 2015;100(8):2857–64.
  19. Agarwal A, Gupta S, Mishra AK, Singh N, Mishra SK. Normotensive pheochromocytoma: Institutional experience. World J. Surg. 2005;29(9):1185–1188.
  20. Bhansali A, Rajput R, Behra A, Rao KLN, Khandelwal N, Radotra BD. Childhood sporadic pheochromocytoma: Clinical profile and outcome in 19 patients. J. Pediatr. Endocrinol. Metab. 2006;19(5):749–756.
  21. Otsuka F, Ogura T, Nakagawa M, Hayakawa N, Kataoka H, Oishi T, Makino H. Normotensive bilateral pheochromocytoma with Lindau disease: case report. Endocr. J. 1996;43(6):719–23.
  22. Lenders JWM, Duh QY, Eisenhofer G, Gimenez-Roqueplo AP, Grebe SKG, Murad MH, Naruse M, Pacak K, Young WF. Pheochromocytoma and paraganglioma: An endocrine society clinical practice guideline. J. Clin. Endocrinol. Metab. 2014;99(6):1915–1942.
  23. Nieman LK, Biller BMK, Findling JW, Newell-Price J, Savage MO, Stewart PM, Montori VM, Edwards H. The diagnosis of Cushing’s syndrome: An endocrine society clinical practice guideline. J. Clin. Endocrinol. Metab. 2008;93(5):1526–1540.
  24. Fardella CE, Mosso L, Gómez-Sánchez C, Cortés P, Soto J, Gómez L, Pinto M, Huete A, Oestreicher E, Foradori A, Montero J. Primary Hyperaldosteronism in Essential Hypertensives: Prevalence, Biochemical Profile, and Molecular Biology 1 . J. Clin. Endocrinol. Metab. 2000;85(5):1863–1867.
  25. Douma S, Petidis K, Doumas M, Papaefthimiou P, Triantafyllou A, Kartali N, Papadopoulos N, Vogiatzis K, Zamboulis C. Prevalence of primary hyperaldosteronism in resistant hypertension: a retrospective observational study. Lancet (London, England) 2008;371(9628):1921–6.
  26. Surendran P, Drenos F, Young R, Warren H, Cook JP, Manning AK, Grarup N, Sim X, Barnes DR, Witkowska K, Staley JR, Tragante V, Tukiainen T, Yaghootkar H, Masca N, Freitag DF, Ferreira T, Giannakopoulou O, Tinker A, Harakalova M, Mihailov E, Liu C, Kraja AT, Fallgaard Nielsen S, Rasheed A, Samuel M, Zhao W, Bonnycastle LL, Jackson AU, Narisu N, Swift AJ, Southam L, Marten J, Huyghe JR, Stančáková A, Fava C, Ohlsson T, Matchan A, Stirrups KE, Bork-Jensen J, Gjesing AP, Kontto J, Perola M, Shaw-Hawkins S, Havulinna AS, Zhang H, Donnelly LA, Groves CJ, Rayner NW, Neville MJ, Robertson NR, Yiorkas AM, Herzig K-H, Kajantie E, Zhang W, Willems SM, Lannfelt L, Malerba G, Soranzo N, Trabetti E, Verweij N, Evangelou E, Moayyeri A, Vergnaud A-C, Nelson CP, Poveda A, Varga T V, Caslake M, de Craen AJ, Trompet S, Luan J, Scott RA, Harris SE, Liewald DC, Marioni R, Menni C, Farmaki A-E, Hallmans G, Renström F, Huffman JE, Hassinen M, Burgess S, Vasan RS, Felix JF, CHARGE-Heart Failure Consortium, Uria-Nickelsen M, Malarstig A, Reily DF, Hoek M, Vogt T, Lin H, Lieb W, EchoGen Consortium, Traylor M, Markus HF, METASTROKE Consortium, Highland HM, Justice AE, Marouli E, GIANT Consortium, Lindström J, Uusitupa M, Komulainen P, Lakka TA, Rauramaa R, Polasek O, Rudan I, Rolandsson O, Franks PW, Dedoussis G, Spector TD, EPIC-InterAct Consortium, Jousilahti P, Männistö S, Deary IJ, Starr JM, Langenberg C, Wareham NJ, Brown MJ, Dominiczak AF, Connell JM, Jukema JW, Sattar N, Ford I, Packard CJ, Esko T, Mägi R, Metspalu A, de Boer RA, van der Meer P, van der Harst P, Lifelines Cohort Study, Gambaro G, Ingelsson E, Lind L, de Bakker PI, Numans ME, Brandslund I, Christensen C, Petersen ER, Korpi-Hyövälti E, Oksa H, Chambers JC, Kooner JS, Blakemore AI, Franks S, Jarvelin M-R, Husemoen LL, Linneberg A, Skaaby T, Thuesen B, Karpe F, Tuomilehto J, Doney AS, Morris AD, Palmer CN, Holmen OL, Hveem K, Willer CJ, Tuomi T, Groop L, Käräjämäki A, Palotie A, Ripatti S, Salomaa V, Alam DS, Shafi Majumder A Al, Di Angelantonio E, Chowdhury R, McCarthy MI, Poulter N, Stanton A V, Sever P, Amouyel P, Arveiler D, Blankenberg S, Ferrières J, Kee F, Kuulasmaa K, Müller-Nurasyid M, Veronesi G, Virtamo J, Deloukas P, Wellcome Trust Case Control Consortium, Elliott P, Understanding Society Scientific Group, Zeggini E, Kathiresan S, Melander O, Kuusisto J, Laakso M, Padmanabhan S, Porteous D, Hayward C, Scotland G, Collins FS, Mohlke KL, Hansen T, Pedersen O, Boehnke M, Stringham HM, EPIC-CVD Consortium, Frossard P, Newton-Cheh C, CHARGE+ Exome Chip Blood Pressure Consortium, Tobin MD, Nordestgaard BG, T2D-GENES Consortium, GoT2DGenes Consortium, ExomeBP Consortium, CHD Exome+ Consortium, Caulfield MJ, Mahajan A, Morris AP, Tomaszewski M, Samani NJ, Saleheen D, Asselbergs FW, Lindgren CM, Danesh J, Wain L V, Butterworth AS, Howson JM, Munroe PB. Trans-ancestry meta-analyses identify rare and common variants associated with blood pressure and hypertension. Nat. Genet. 2016;48(10):1151–1161.
  27. Liu C, Kraja AT, Smith JA, Brody JA, Franceschini N, Bis JC, Rice K, Morrison AC, Lu Y, Weiss S, Guo X, Palmas W, Martin LW, Chen YDI, Surendran P, Drenos F, Cook JP, Auer PL, Chu AY, Giri A, Zhao W, Jakobsdottir J, Lin LA, Stafford JM, Amin N, Mei H, Yao J, Voorman A, Larson MG, Grove ML, Smith A V., Hwang SJ, Chen H, Huan T, Kosova G, Stitziel NO, Kathiresan S, Samani N, Schunkert H, Deloukas P, Li M, Fuchsberger C, Pattaro C, Gorski M, Kooperberg C, Papanicolaou GJ, Rossouw JE, Faul JD, Kardia SLR, Bouchard C, Raffel LJ, Uitterlinden AG, Franco OH, Vasan RS, O’Donnell CJ, Taylor KD, Liu K, Bottinger EP, Gottesman O, Daw EW, Giulianini F, Ganesh S, Salfati E, Harris TB, Launer LJ, Dörr M, Felix SB, Rettig R, Völzke H, Kim E, Lee WJ, Lee I Te, Sheu WHH, Tsosie KS, Edwards DRV, Liu Y, Correa A, Weir DR, Völker U, Ridker PM, Boerwinkle E, Gudnason V, Reiner AP, Van Duijn CM, Borecki IB, Edwards TL, Chakravarti A, Rotter JI, Psaty BM, Loos RJF, Fornage M, Ehret GB, Newton-Cheh C, Levy D, Chasman DI. Meta-analysis identifies common and rare variants influencing blood pressure and overlapping with metabolic trait loci. Nat. Genet. 2016;48(10):1162–1170.
  28. Melcescu E, Koch CA. Endocrine Hypertension.; 2000. Available at: http://www.ncbi.nlm.nih.gov/pubmed/25905183. Accessed December 21, 2019.
  29. Vasan RS, Evans JC, Larson MG, Wilson PWF, Meigs JB, Rifai N, Benjamin EJ, Levy D. Serum aldosterone and the incidence of hypertension in nonhypertensive persons. N. Engl. J. Med. 2004;351(1):33-41+111.
  30. Seiler L, Rump LC, Schulte-Mönting J, Slawik M, Borm K, Pavenstädt H, Beuschlein F, Reincke M. Diagnosis of primary aldosteronism: value of different screening parameters and influence of antihypertensive medication. Eur. J. Endocrinol. 2004;150(3):329–37.
  31. Streeten DHP, Tomycz N, Anderson GH. Reliability of screening methods for the diagnosis of primary aldosteronism. Am. J. Med. 1979;67(3):403–413.
  32. Born-Frontsberg E, Reincke M, Rump LC, Hahner S, Diederich S, Lorenz R, Allolio B, Seufert J, Schirpenbach C, Beuschlein F, Bidlingmaier M, Endres S, Quinkler M, Participants of the German Conn’s Registry. Cardiovascular and cerebrovascular comorbidities of hypokalemic and normokalemic primary aldosteronism: results of the German Conn’s Registry. J. Clin. Endocrinol. Metab. 2009;94(4):1125–30.
  33. Ulick S, Blumenfeld JD, Atlas SA, Wang JZ, Vaughan ED. The unique steroidogenesis of the aldosteronoma in the differential diagnosis of primary aldosteronism. J. Clin. Endocrinol. Metab. 1993;76(4):873–8.
  34. Young WF, Stanson AW, Thompson GB, Grant CS, Farley DR, Van Heerden JA. Role for adrenal venous sampling in primary aldosteronism. Surgery 2004;136(6):1227–1235.
  35. Asbach E, Williams TA, Reincke M. Recent Developments in Primary Aldosteronism. Exp. Clin. Endocrinol. Diabetes 2016;124(6):335–41.
  36. Funder JW, Carey RM, Mantero F, Murad MH, Reincke M, Shibata H, Stowasser M, Young WF. The management of primary aldosteronism: Case detection, diagnosis, and treatment: An endocrine society clinical practice guideline. J. Clin. Endocrinol. Metab. 2016;101(5):1889–1916.
  37. Toniato A, Bernante P, Rossi GP, Pelizzo MR. The role of adrenal venous sampling in the surgical management of primary aldosteronism. World J. Surg. 2006;30(4):624–627.
  38. Gordon RD, Stowasser M. Familial forms broaden the horizons for primary aldosteronism. Trends Endocrinol. Metab. 1998;9(6):220–227.
  39. Stowasser M, Gordon RD. Primary aldosteronism: From genesis to genetics. Trends Endocrinol. Metab. 2003;14(7):310–317.
  40. Melcescu E, Phillips J, Moll G, Subauste JS, Koch CA. 11Beta-hydroxylase deficiency and other syndromes of mineralocorticoid excess as a rare cause of endocrine hypertension. Horm. Metab. Res. 2012;44(12):867–78.
  41. Grim CE, Weinberger MH. Familial, dexamethasone-suppressible, normokalemic hyperaldosteronism. Pediatrics 1980;65(3):597–604.
  42. Lafferty AR, Torpy DJ, Stowasser M, Taymans SE, Lin JP, Huggard P, Gordon RD, Stratakis CA. A novel genetic locus for low renin hypertension: familial hyperaldosteronism type II maps to chromosome 7 (7p22). J. Med. Genet. 2000;37(11):831–5.
  43. Charmandari E, Sertedaki A, Kino T, Merakou C, Hoffman DA, Hatch MM, Hurt DE, Lin L, Xekouki P, Stratakis CA, Chrousos GP. A novel point mutation in the KCNJ5 gene causing primary hyperaldosteronism and early-onset autosomal dominant hypertension. J. Clin. Endocrinol. Metab. 2012;97(8):E1532-9.
  44. Weinberger MH, Fineberg NS. The diagnosis of primary aldosteronism and separation of two major subtypes. Arch. Intern. Med. 1993;153(18):2125–9.
  45. Wulczyn K, Perez-Reyes E, Nussbaum RL, Park M. Primary aldosteronism associated with a germline variant in CACNA1H. BMJ Case Rep. 2019;12(5). doi:10.1136/bcr-2018-229031.
  46. Seidel E, Schewe J, Scholl UI. Genetic causes of primary aldosteronism. Exp. Mol. Med. 2019;51(11). doi:10.1038/s12276-019-0337-9.
  47. Schirpenbach C, Seiler L, Maser-Gluth C, Beuschlein F, Reincke M, Bidlingmaier M. Automated chemiluminescence-immunoassay for aldosterone during dynamic testing: comparison to radioimmunoassays with and without extraction steps. Clin. Chem. 2006;52(9):1749–55.
  48. Stowasser M, Gordon RD. Aldosterone assays: an urgent need for improvement. Clin. Chem. 2006;52(9):1640–2.
  49. Palermo M, Quinkler M, Stewart PM. Apparent mineralocorticoid excess syndrome: an overview. Arq. Bras. Endocrinol. Metabol. 2004;48(5):687–96.
  50. Vaidya A, Dluhy R. Hyperaldosteronism. MDText.com, Inc; 2000.
  51. Tsiavos V, Markou A, Papanastasiou L, Kounadi T, Androulakis II, Voulgaris N, Zachaki A, Kassi E, Kaltsas G, Chrousos GP, Piaditis GP. A new highly sensitive and specific overnight combined screening and diagnostic test for primary aldosteronism. Eur. J. Endocrinol. 2016;175(1):21–8.
  52. Stowasser M, Gordon RD. Primary aldosteronism - Careful investigation is essential and rewarding. In: Molecular and Cellular Endocrinology.Vol 217.; 2004:33–39.
  53. Fourkiotis V, Vonend O, Diederich S, Fischer E, Lang K, Endres S, Beuschlein F, Willenberg HS, Rump LC, Allolio B, Reincke M, Quinkler M. Effectiveness of eplerenone or spironolactone treatment in preserving renal function in primary aldosteronism. Eur. J. Endocrinol. 2013;168(1):75–81.
  54. Jansen PM, van den Meiracker AH, Jan Danser AH. Aldosterone synthase inhibitors: pharmacological and clinical aspects. Curr. Opin. Investig. Drugs 2009;10(4):319–26.
  55. Stowasser M, Gordon RD. Primary aldosteronism: Learning from the study of familial varieties. J. Hypertens. 2000;18(9):1165–1176.
  56. Sukor N, Gordon RD, Yee KK, Jones M, Stowasser M. Role of unilateral adrenalectomy in bilateral primary aldosteronism: A 22-year single center experience. J. Clin. Endocrinol. Metab. 2009;94(7):2437–2445.
  57. Letavernier E, Peyrard S, Amar L, Zinzindohoué F, Fiquet B, Plouin P-F. Blood pressure outcome of adrenalectomy in patients with primary hyperaldosteronism with or without unilateral adenoma. J. Hypertens. 2008;26(9):1816–23.
  58. Meyer A, Brabant G, Behrend M. Long-term follow-up after adrenalectomy for primary aldosteronism. World J. Surg. 2005;29(2):155–9.
  59. Rossi GP. Surgically correctable hypertension caused by primary aldosteronism. Best Pract. Res. Clin. Endocrinol. Metab. 2006;20(3):385–400.
  60. Catena C, Lapenna R, Baroselli S, Nadalini E, Colussi GL, Novello M, Favret G, Melis A, Cavarape A, Sechi LA. Insulin sensitivity in patients with primary aldosteronism: A follow-up study. J. Clin. Endocrinol. Metab. 2006;91(9):3457–3463.
  61. Matrozova J, Steichen O, Amar L, Zacharieva S, Jeunemaitre X, Plouin PF. Fasting plasma glucose and serum lipids in patients with primary aldosteronism a controlled cross-sectional study. Hypertension 2009;53(4):605–610.
  62. Kempers MJE, Lenders JWM, Van Outheusden L, Van Der Wilt GJ, Kool LJS, Hermus ARMM, Deinum J. Systematic review: Diagnostic procedures to differentiate unilateral from bilateral adrenal abnormality in primary aldosteronism. Ann. Intern. Med. 2009;151(5):329–337.
  63. Beard CM, Sheps SG, Kurland LT, Carney JA, Lie JT. Occurrence of pheochromocytoma in Rochester, Minnesota, 1950 through 1979. Mayo Clin. Proc. 1983;58(12):802–804.
  64. Guerrero MA, Schreinemakers JMJ, Vriens MR, Suh I, Hwang J, Shen WT, Gosnell J, Clark OH, Duh Q-Y. Clinical spectrum of pheochromocytoma. J. Am. Coll. Surg. 2009;209(6):727–32.
  65. Erlic Z, Rybicki L, Peczkowska M, Golcher H, Kann PH, Brauckhoff M, Müssig K, Muresan M, Schäffler A, Reisch N, Schott M, Fassnacht M, Opocher G, Klose S, Fottner C, Forrer F, Plöckinger U, Petersenn S, Zabolotny D, Kollukch O, Yaremchuk S, Januszewicz A, Walz MK, Eng C, Neumann HPH, European-American Pheochromocytoma Study Group. Clinical predictors and algorithm for the genetic diagnosis of pheochromocytoma patients. Clin. Cancer Res. 2009;15(20):6378–85.
  66. Jiménez C, Cote G, Arnold A, Gagel RF. Review: Should patients with apparently sporadic pheochromocytomas or paragangliomas be screened for hereditary syndromes? J. Clin. Endocrinol. Metab. 2006;91(8):2851–8.
  67. Eisenhofer G, Lenders JWM, Goldstein DS, Mannelli M, Csako G, Walther MM, Brouwers FM, Pacak K. Pheochromocytoma catecholamine phenotypes and prediction of tumor size and location by use of plasma free metanephrines. Clin. Chem. 2005;51(4):735–744.
  68. Eisenhofer G, Walther MM, Huynh TT, Li ST, Bornstein SR, Vortmeyer A, Mannelli M, Goldstein DS, Linehan WM, Lenders JWM, Pacak K. Pheochromocytomas in von Hippel-Lindau syndrome and multiple endocrine neoplasia type 2 display distinct biochemical and clinical phenotypes. J. Clin. Endocrinol. Metab. 2001;86(5):1999–2008.
  69. Neumann HPH, Vortmeyer A, Schmidt D, Werner M, Erlic Z, Cascon A, Bausch B, Januszewicz A, Eng C. Evidence of MEN-2 in the original description of classic pheochromocytoma. N. Engl. J. Med. 2007;357(13):1311–5.
  70. Bravo EL, Tagle R. Pheochromocytoma: State-of-the-art and future prospects. Endocr. Rev. 2003;24(4):539–553.
  71. Lu Y, Li P, Gan W, Zhao X, Shen S, Feng W, Xu Q, Bi Y, Guo H, Zhu D. Clinical and Pathological Characteristics of Hypertensive and Normotensive Adrenal Pheochromocytomas. Exp. Clin. Endocrinol. Diabetes 2016;124(6):372–379.
  72. Prejbisz A, Lenders JWM, Eisenhofer G, Januszewicz A. Cardiovascular manifestations of phaeochromocytoma. J. Hypertens. 2011;29(11):2049–2060.
  73. Giavarini A, Chedid A, Bobrie G, Plouin PF, Hagège A, Amar L. Acute catecholamine cardiomyopathy in patients with phaeochromocytoma or functional paraganglioma. Heart 2013;99(19):1438–1444.
  74. Y-Hassan S. Clinical Features and Outcome of Pheochromocytoma-Induced Takotsubo Syndrome: Analysis of 80 Published Cases. Am. J. Cardiol. 2016;117(11):1836–1844.
  75. Gagnon N, Mansour S, Bitton Y, Bourdeau I. TAKOTSUBO-LIKE CARDIOMYOPATHY in A LARGE COHORT of PATIENTS with PHEOCHROMOCYTOMA and PARAGANGLIOMA. Endocr. Pract. 2017;23(10):1178–1192.
  76. Miehle K, Kratzsch J, Lenders JWM, Kluge R, Paschke R, Koch CA. Adrenal incidentaloma diagnosed as pheochromocytoma by plasma chromogranin A and plasma metanephrines. J. Endocrinol. Invest. 2005;28(11):1040–2.
  77. Bausch B, Tischler AS, Schmid KW, Leijon H, Eng C, Neumann HPH. Max Schottelius: Pioneer in Pheochromocytoma. J. Endocr. Soc. 2017;1(7):957–964.
  78. Eisenhofer G, Lenders JWM, Siegert G, Bornstein SR, Friberg P, Milosevic D, Mannelli M, Linehan WM, Adams K, Timmers HJ, Pacak K. Plasma methoxytyramine: A novel biomarker of metastatic pheochromocytoma and paraganglioma in relation to established risk factors of tumour size, location and SDHB mutation status. Eur. J. Cancer 2012;48(11):1739–1749.
  79. Niculescu DA, Ismail G, Poiana C. Plasma free metanephrine and normetanephrine levels are increased in patients with chronic kidney disease. Endocr. Pract. 2014;20(2):139–44.
  80. Eisenhofer G, Lattke P, Herberg M, Siegert G, Qin N, Därr R, Hoyer J, Villringer A, Prejbisz A, Januszewicz A, Remaley A, Martucci V, Pacak K, Ross HA, Sweep FCGJ, Lenders JWM. Reference intervals for plasma free metanephrines with an age adjustment for normetanephrine for optimized laboratory testing of phaeochromocytoma. Ann. Clin. Biochem. 2013;50(1):62–69.
  81. De Jong WHA, Eisenhofer G, Post WJ, Muskiet FAJ, De Vries EGE, Kema IP. Dietary influences on plasma and urinary metanephrines: Implications for diagnosis of catecholamine-producing tumors. J. Clin. Endocrinol. Metab. 2009;94(8):2841–2849.
  82. Neumann HPH, Young WF, Eng C. Pheochromocytoma and Paraganglioma. N. Engl. J. Med. 2019;381(6):552–565.
  83. Majumdar S, Friedrich CA, Koch CA, Megason GC, Fratkin JD, Moll GW. Compound heterozygous mutation with a novel splice donor region DNA sequence variant in the succinate dehydrogenase subunit B gene in malignant paraganglioma. Pediatr. Blood Cancer 2010;54(3):473–5.
  84. Timmers HJLM, Gimenez-Roqueplo A-P, Mannelli M, Pacak K. Clinical aspects of SDHx-related pheochromocytoma and paraganglioma. Endocr. Relat. Cancer 2009;16(2):391–400.
  85. Bhatia KSS, Ismail MM, Sahdev A, Rockall AG, Hogarth K, Canizales A, Avril N, Monson JP, Grossman AB, Reznek RH. 123I-metaiodobenzylguanidine (MIBG) scintigraphy for the detection of adrenal and extra-adrenal phaeochromocytomas: CT and MRI correlation. Clin. Endocrinol. (Oxf). 2008;69(2):181–8.
  86. Lenders JWM, Pacak K, Walther MM, Marston Linehan W, Mannelli M, Friberg P, Keiser HR, Goldstein DS, Eisenhofer G. Biochemical diagnosis of pheochromocytoma: Which test is best? J. Am. Med. Assoc. 2002;287(11):1427–1434.
  87. Koch CA. Should 123I-MIBG scintigraphy be part of the workup for pheochromocytomas? Nat. Clin. Pract. Endocrinol. Metab. 2009;5(2):76–7.
  88. Koopmans KP, Jager PL, Kema IP, Kerstens MN, Albers F, Dullaart RPF. 111In-octreotide is superior to 123I-metaiodobenzylguanidine for scintigraphic detection of head and neck paragangliomas. J. Nucl. Med. 2008;49(8):1232–7.
  89. Mazza A, Beltramello G, Armigliato M, Montemurro D, Zorzan S, Zuin M, Rampin L, Marzola MC, Grassetto G, Al-Nahhas A, Rubello D. Arterial hypertension and thyroid disorders: What is important to know in clinical practice? Ann. Endocrinol. (Paris). 2011;72(4):296–303.
  90. Jimenez C, Cabanillas ME, Santarpia L, Jonasch E, Kyle KL, Lano EA, Matin SF, Nunez RF, Perrier ND, Phan A, Rich TA, Shah B, Williams MD, Waguespack SG. Use of the tyrosine kinase inhibitor sunitinib in a patient with von Hippel-Lindau disease: targeting angiogenic factors in pheochromocytoma and other von Hippel-Lindau disease-related tumors. J. Clin. Endocrinol. Metab. 2009;94(2):386–91.
  91. Neumann HPH, Tsoy U, Bancos I, Amodru V, Walz MK, Tirosh A, Kaur RJ, McKenzie T, Qi X, Bandgar T, Petrov R, Yukina MY, Roslyakova A, Van Der Horst-Schrivers ANA, Berends AMA, Hoff AO, Castroneves LA, Ferrara AM, Rizzati S, Mian C, Dvorakova S, Hasse-Lazar K, Kvachenyuk A, Peczkowska M, Loli P, Erenler F, Krauss T, Almeida MQ, Liu L, Zhu F, Recasens M, Wohllk N, Corssmit EPM, Shafigullina Z, Calissendorff J, Grozinsky-Glasberg S, Kunavisarut T, Schalin-Jäntti C, Castinetti F, Vlček P, Beltsevich D, Egorov VI, Schiavi F, Links TP, Lechan RM, Bausch B, Young WF, Eng C. Comparison of Pheochromocytoma-Specific Morbidity and Mortality among Adults with Bilateral Pheochromocytomas Undergoing Total Adrenalectomy vs Cortical-Sparing Adrenalectomy. JAMA Netw. Open 2019. doi:10.1001/jamanetworkopen.2019.8898.
  92. Castinetti F, Waguespack SG, Machens A, Uchino S, Hasse-Lazar K, Sanso G, Else T, Dvorakova S, Qi XP, Elisei R, Maia AL, Glod J, Lourenço DM, Valdes N, Mathiesen J, Wohllk N, Bandgar TR, Drui D, Korbonits M, Druce MR, Brain C, Kurzawinski T, Patocs A, Bugalho MJ, Lacroix A, Caron P, Fainstein-Day P, Borson Chazot F, Klein M, Links TP, Letizia C, Fugazzola L, Chabre O, Canu L, Cohen R, Tabarin A, Spehar Uroic A, Maiter D, Laboureau S, Mian C, Peczkowska M, Sebag F, Brue T, Mirebeau-Prunier D, Leclerc L, Bausch B, Berdelou A, Sukurai A, Vlcek P, Krajewska J, Barontini M, Vaz Ferreira Vargas C, Valerio L, Ceolin L, Akshintala S, Hoff A, Godballe C, Jarzab B, Jimenez C, Eng C, Imai T, Schlumberger M, Grubbs E, Dralle H, Neumann HP, Baudin E. Natural history, treatment, and long-term follow up of patients with multiple endocrine neoplasia type 2B: an international, multicentre, retrospective study. lancet. Diabetes Endocrinol. 2019;7(3):213–220.
  93. Brauckhoff M, Gimm O, Thanh PN, Bär A, Ukkat J, Brauckhoff K, Bönsch T, Dralle H, McHenry CR, Thompson GB, Duh QY. Critical size of residual adrenal tissue and recovery from impaired early postoperative adrenocortical function after subtotal bilateral adrenalectomy. In: Surgery.Vol 134. Mosby Inc.; 2003:1020–1027.
  94. Lodish MB, Sinaii N, Patronas N, Batista DL, Keil M, Samuel J, Moran J, Verma S, Popovic J, Stratakis CA. Blood pressure in pediatric patients with Cushing syndrome. J. Clin. Endocrinol. Metab. 2009;94(6):2002–8.
  95. Isidori AM, Graziadio C, Paragliola RM, Cozzolino A, Ambrogio AG, Colao A, Corsello SM, Pivonello R, ABC Study Group. The hypertension of Cushing’s syndrome: controversies in the pathophysiology and focus on cardiovascular complications. J. Hypertens. 2015;33(1):44–60.
  96. Zacharieva S, Orbetzova M, Stoynev A, Shigarminova R, Yaneva M, Kalinov K, Nachev E, Elenkova A. Circadian blood pressure profile in patients with Cushing’s syndrome before and after treatment. J. Endocrinol. Invest. 2004;27(10):924–30.
  97. Baid S, Nieman LK. Glucocorticoid excess and hypertension. Curr. Hypertens. Rep. 2004;6(6):493–499.
  98. Juszczak A, Sulentic P, Grossman A. Cushing’s Syndrome.; 2000. Available at: http://www.ncbi.nlm.nih.gov/pubmed/25905314. Accessed January 16, 2020.
  99. Torpy DJ, Mullen N, Ilias I, Nieman LK. Association of hypertension and hypokalemia with Cushing’s syndrome caused by ectopic ACTH secretion: a series of 58 cases. Ann. N. Y. Acad. Sci. 2002;970:134–44.
  100. Nieman LK, Biller BMK, Findling JW, Newell-Price J, Savage MO, Stewart PM, Montori VM. Endo Soc Cushing’s. J. Clin. Endocrinol. Metab. 2008;93(5):1526–40.
  101. Nieman LK, Biller BMK, Findling JW, Murad MH, Newell-Price J, Savage MO, Tabarin A, Endocrine Society. Treatment of Cushing’s Syndrome: An Endocrine Society Clinical Practice Guideline. J. Clin. Endocrinol. Metab. 2015;100(8):2807–31.
  102. Fassnacht M, Arlt W, Bancos I, Dralle H, Newell-Price J, Sahdev A, Tabarin A, Terzolo M, Tsagarakis S, Dekkers OM. ESEC 2016 Guideline incidentaloma adrenal. Eur. J. Endocrinol. 2016. doi:10.1530/EJE-16-0467.
  103. Lopez D, Luque-Fernandez MA, Steele A, Adler GK, Turchin A, Vaidya A. “Nonfunctional” adrenal Tumors and the risk for incident diabetes and cardiovascular outcomes: A cohort study. Ann. Intern. Med. 2016;165(8):533–542.
  104. Chatzellis E, Kaltsas G. Adrenal Incidentalomas.; 2000. Available at: http://www.ncbi.nlm.nih.gov/pubmed/25905250. Accessed January 16, 2020.
  105. Zeiger MA, Thompson GB, Duh Q-Y, Hamrahian AH, Angelos P, Elaraj D, Fishman E, Kharlip J, American Association of Clinical Endocrinologists, American Association of Endocrine Surgeons. American Association of Clinical Endocrinologists and American Association of Endocrine Surgeons Medical Guidelines for the Management of Adrenal Incidentalomas: executive summary of recommendations. Endocr. Pract. 15(5):450–3.
  106. Hannah-Shmouni F, Melcescu E, Koch CA. Testing for Endocrine Hypertension.; 2000. Available at: http://www.ncbi.nlm.nih.gov/pubmed/25905199. Accessed December 21, 2019.
  107. Gläsker S, Neumann HPH, Koch CA, Vortmeyer A. Von Hippel-Lindau Disease.; 2000. Available at: http://www.ncbi.nlm.nih.gov/pubmed/25905347. Accessed January 16, 2020.
  108. Kino T, Vottero A, Charmandari E, Chrousos GP. Familial/sporadic glucocorticoid resistance syndrome and hypertension. Ann. N. Y. Acad. Sci. 2002;970:101–11.
  109. Chrousos GP, Vingerhoeds A, Brandon D, Eil C, Pugeat M, DeVroede M, Loriaux DL, Lipsett MB. Primary cortisol resistance in man. A glucocorticoid receptor-mediated disease. J. Clin. Invest. 1982;69(6):1261–1269.
  110. Charmandari E, Kino T, Ichijo T, Chrousos GP. Generalized glucocorticoid resistance: Clinical aspects, molecular mechanisms, and implications of a rare genetic disorder. J. Clin. Endocrinol. Metab. 2008;93(5):1563–1572.
  111. Nebesio TD, Eugster EA. Observation of hypertension in children with 21-hydroxylase deficiency: a preliminary report. Endocrine 2006;30(3):279–82.
  112. Finkielstain GP, Kim MS, Sinaii N, Nishitani M, Van Ryzin C, Hill SC, Reynolds JC, Hanna RM, Merke DP. Clinical characteristics of a cohort of 244 patients with congenital adrenal hyperplasia. J. Clin. Endocrinol. Metab. 2012;97(12):4429–38.
  113. Kim MS, Merke DP. Cardiovascular disease risk in adult women with congenital adrenal hyperplasia due to 21-hydroxylase deficiency. Semin. Reprod. Med. 2009;27(4):316–21.
  114. Zachmann M, Tassinari D, Prader A. Clinical and biochemical variability of congenital adrenal hyperplasia due to 11 beta-hydroxylase deficiency. A study of 25 patients. J. Clin. Endocrinol. Metab. 1983;56(2):222–9.
  115. New MI, Geller DS, Fallo F, Wilson RC. Monogenic low renin hypertension. Trends Endocrinol. Metab. 2005;16(3):92–97.
  116. Mimouni M, Kaufman H, Roitman A, Morag C, Sadan N. Hypertension in a neonate with 11 β-hydroxylase deficiency. Eur. J. Pediatr. 1985;143(3):231–233.
  117. Speiser PW, Azziz R, Baskin LS, Ghizzoni L, Hensle TW, Merke DP, Meyer-Bahlburg HFL, Miller WL, Montori VM, Oberfield SE, Ritzen M, White PC. Congenital adrenal hyperplasia due to steroid 21-hydroxylase deficiency: An Endocrine Society clinical practice guideline. J. Clin. Endocrinol. Metab. 2010;95(9):4133–4160.
  118. Speiser PW, White PC. Congenital adrenal hyperplasia. N. Engl. J. Med. 2003;349(8):776–88.
  119. Melcescu E, Griswold M, Xiang L, Belk S, Montgomery D, Bray M, Del Ben KS, Uwaifo GI, Marshall GD, Koch CA. Prevalence and cardiometabolic associations of the glucocorticoid receptor gene polymorphisms N363S and BclI in obese and non-obese black and white Mississippians. Hormones (Athens). 11(2):166–77.
  120. Hochberg Z, Even L, Zadik Z. Mineralocorticoids in the mechanism of gynecomastia in adrenal hyperplasia caused by 11β-hydroxylase deficiency. J. Pediatr. 1991;118(2):258–260.
  121. Kacem M, Moussa A, Khochtali I, Nabouli R, Morel Y, Zakhama A. Bilateral adrenalectomy for severe hypertension in congenital adrenal hyperplasia due to 11β-hydroxylase deficiency: Long term follow-up. Ann. Endocrinol. (Paris). 2009;70(2):113–118.
  122. New MI. Male pseudohermaphroditism due to 17 alpha-hydroxylase deficiency. J. Clin. Invest. 1970;49(10):1930–41.
  123. Wong S-L, Shu S-G, Tsai C-R. Seventeen alpha-hydroxylase deficiency. J. Formos. Med. Assoc. 2006;105(2):177–81.
  124. Costa-Santos M, Kater CE, Auchus RJ, Brazilian Congenital Adrenal Hyperplasia Multicenter Study Group. Two prevalent CYP17 mutations and genotype-phenotype correlations in 24 Brazilian patients with 17-hydroxylase deficiency. J. Clin. Endocrinol. Metab. 2004;89(1):49–60.
  125. Müssig K, Wehrmann M, Horger M, Maser-Gluth C, Häring HU, Overkamp D. Adrenocortical carcinoma producing 11-deoxycorticosterone: A rare cause of mineralocorticoid hypertension. J. Endocrinol. Invest. 2005;28(1):61–65.
  126. Carvajal CA, Gonzalez AA, Romero DG, González A, Mosso LM, Lagos ET, Hevia M del P, Rosati MP, Perez-Acle TO, Gomez-Sanchez CE, Montero JA, Fardella CE. Two homozygous mutations in the 11 beta-hydroxysteroid dehydrogenase type 2 gene in a case of apparent mineralocorticoid excess. J. Clin. Endocrinol. Metab. 2003;88(6):2501–7.
  127. Lin-Su K, Zhou P, Arora N, Betensky BP, New MI, Wilson RC. In vitro expression studies of a novel mutation delta299 in a patient affected with apparent mineralocorticoid excess. J. Clin. Endocrinol. Metab. 2004;89(5):2024–7.
  128. New MI, Levine LS, Biglieri EG, Pareira J, Ulick S. Evidence for an unidentified steroid in a child with apparent mineralocorticoid hypertension. J. Clin. Endocrinol. Metab. 1977;44(5):924–33.
  129. Wilson RC, Krozowski ZS, Li K, Obeyesekere VR, Razzaghy-Azar M, Harbison MD, Wei JQ, Shackleton CH, Funder JW, New MI. A mutation in the HSD11B2 gene in a family with apparent mineralocorticoid excess. J. Clin. Endocrinol. Metab. 1995;80(7):2263–2266.
  130. Stewart PM, Krozowski ZS, Gupta A, Milford D V., Howie AJ, Sheppard MC, Whorwood CB. Hypertension in the syndrome of apparent mineralocorticoid excess due to mutation of the 11β-hydroxysteroid dehydrogenase type 2 gene. Lancet 1996;347(8994):88–91.
  131. Stewart PM, Corrie JE, Shackleton CH, Edwards CR. Syndrome of apparent mineralocorticoid excess. A defect in the cortisol-cortisone shuttle. J. Clin. Invest. 1988;82(1):340–9.
  132. Arriza JL, Weinberger C, Cerelli G, Glaser TM, Handelin BL, Housman DE, Evans RM. Cloning of human mineralocorticoid receptor complementary DNA: Structural and functional kinship with the glucocorticoid receptor. Science (80-. ). 1987;237(4812):268–275.
  133. Funder JW, Pearce PT, Smith R, Smith AI. Mineralocorticoid action: Target tissue specificity is enzyme, not receptor, mediated. Science (80-. ). 1988;242(4878):583–585.
  134. Heilmann P, Buchheim E, Wacker J, Ziegler R. Alteration of the activity of the 11beta-hydroxysteroid dehydrogenase in pregnancy: relevance for the development of pregnancy-induced hypertension? J. Clin. Endocrinol. Metab. 2001;86(11):5222–6.
  135. Quinkler M, Oelkers W, Diederich S. In Vivo Measurement of Renal 11β-Hydroxysteroid Dehydrogenase Type 2 Activity . J. Clin. Endocrinol. Metab. 2000;85(12):4921–4922.
  136. Geller DS, Farhi A, Pinkerton N, Fradley M, Moritz M, Spitzer A, Meinke G, Tsai FTF, Sigler PB, Lifton RP. Activating mineralocorticoid receptor mutation in hypertension exacerbated by pregnancy. Science (80-. ). 2000;289(5476):119–123.
  137. Liddle GW, Bledsoe T, Coppage WS. Hypertension reviews. J. Tenn. Med. Assoc. 1974;67(8):669.
  138. Gordon RD, Geddes RA, Pawsey CG, O’Halloran MW. Hypertension and severe hyperkalaemia associated with suppression of renin and aldosterone and completely reversed by dietary sodium restriction. Australas. Ann. Med. 1970;19(4):287–94.
  139. Geller DS. Mineralocorticoid resistance. Clin. Endocrinol. (Oxf). 2005;62(5):513–520.
  140. Klemm SA, Gordon RD, Tunny TJ, Thompson RE. The syndrome of hypertension and hyperkalemia with normal GFR (Gordon’s syndrome): is there increased proximal sodium reabsorption? Clin. Invest. Med. 1991;14(6):551–8.
  141. Wilson FH, Disse-Nicodème S, Choate KA, Ishikawa K, Nelson-Williams C, Desitter I, Gunel M, Milford D V, Lipkin GW, Achard JM, Feely MP, Dussol B, Berland Y, Unwin RJ, Mayan H, Simon DB, Farfel Z, Jeunemaitre X, Lifton RP. Human hypertension caused by mutations in WNK kinases. Science 2001;293(5532):1107–12.
  142. Mansmann G, Lau J, Balk E, Rothberg M, Miyachi Y, Bornstein SR. The clinically inapparent adrenal mass: Update in diagnosis and management. Endocr. Rev. 2004;25(2):309–340.
  143. Sakoh T, Sekine A, Mori T, Mizuno H, Kawada M, Hiramatsu R, Hasegawa E, Hayami N, Yamanouchi M, Suwabe T, Sawa N, Ubara Y, Fujimaru T, Sohara E, Shinichi U, Hoshino J, Takaichi K. A familial case of pseudohypoaldosteronism type II (PHA2) with a novel mutation (D564N) in the acidic motif in WNK4. Mol. Genet. genomic Med. 2019;7(6):e705.
  144. Xie J, Craig L, Cobb MH, Huang C-L. Role of with-no-lysine [K] kinases in the pathogenesis of Gordon’s syndrome. Pediatr. Nephrol. 2006;21(9):1231–6.
  145. Kahle KT, Wilson FH, Leng Q, Lalioti MD, O’Connell AD, Dong K, Rapson AK, MacGregor GG, Giebisch G, Hebert SC, Lifton RP. WNK4 regulates the balance between renal NaCl reabsorption and K+ secretion. Nat. Genet. 2003;35(4):372–6.
  146. Tobin MD, Raleigh SM, Newhouse S, Braund P, Bodycote C, Ogleby J, Cross D, Gracey J, Hayes S, Smith T, Ridge C, Caulfield M, Sheehan NA, Munroe PB, Burton PR, Samani NJ. Association of WNK1 gene polymorphisms and haplotypes with ambulatory blood pressure in the general population. Circulation 2005;112(22):3423–9.
  147. Koch CA, Bornstein SR, Birkenfeld AL. Introduction to Hanefeld Symposium: 40+ years of metabolic syndrome. Rev. Endocr. Metab. Disord. 2016;17(1):1–4.
  148. Haffner SM, Ruilope L, Dahlöf B, Abadie E, Kupfer S, Zannad F. Metabolic syndrome, new onset diabetes, and new end points in cardiovascular trials. J. Cardiovasc. Pharmacol. 2006;47(3):469–75.
  149. Saad MF, Rewers M, Selby J, Howard G, Jinagouda S, Fahmi S, Zaccaro D, Bergman RN, Savage PJ, Haffner SM. Insulin resistance and hypertension: The insulin resistance atherosclerosis study. Hypertension 2004;43(6):1324–1331.
  150. Ferrannini E, Buzzigoli G, Bonadonna R, Giorico MA, Oleggini M, Graziadei L, Pedrinelli R, Brandi L, Bevilacqua S. Insulin resistance in essential hypertension. N. Engl. J. Med. 1987;317(6):350–7.
  151. Levy J, Gavin JR, Hammerman MR, Avioli L V. Ca2+-Mg2+-ATPase Activity in Kidney Basolateral Membrane in Non-Insulin-Dependent Diabetic Rats: Effect of Insulin. Diabetes 1986;35(8):899–905.
  152. Bogaert YE, Linas S. The role of obesity in the pathogenesis of hypertension. Nat. Clin. Pract. Nephrol. 2009;5(2):101–11.
  153. Graessler J, Schwudke D, Schwarz PEH, Herzog R, Shevchenko A, Bornstein SR. Top-down lipidomics reveals ether lipid deficiency in blood plasma of hypertensive patients. PLoS One 2009;4(7):e6261.
  154. McCarron DA, Pingree PA, Rubin RJ, Gaucher SM, Molitch M, Krutzik S. Enhanced parathyroid function in essential hypertension: A homeostatic response to a urinary calcium leak. Hypertension 1980;2(2):162–168.
  155. Bukoski RD, Ishibashi K, Bian K. Vascular actions of the calcium-regulating hormones. Semin. Nephrol. 1995;15(6):536–49.
  156. Hulter HN, Melby JC, Peterson JC, Cooke CR. Chronic continuous PTH infusion results in hypertension in normal subjects. J. Clin. Hypertens. 1986;2(4):360–70.
  157. Bucher HC, Cook RJ, Guyatt GH, Lang JD, Cook DJ, Hatala R, Hunt DL. Effects of dietary calcium supplementation on blood pressure. A meta-analysis of randomized controlled trials. JAMA 1996;275(13):1016–22.
  158. Allender PS, Cutler JA, Follmann D, Cappuccio FP, Pryer J, Elliott P. Dietary calcium and blood pressure: A meta-analysis of randomized clinical trials. Ann. Intern. Med. 1996;124(9):825–831.
  159. Bollerslev J, Rosen T, Mollerup CL, Nordenström J, Baranowski M, Franco C, Pernow Y, Isaksen GA, Godang K, Ueland T, Jansson S. Effect of surgery on cardiovascular risk factors in mild primary hyperparathyroidism. J. Clin. Endocrinol. Metab. 2009;94(7):2255–2261.
  160. Bollerslev J, Jansson S, Mollerup CL, Nordenström J, Lundgren E, Tørring O, Varhaug J-E, Baranowski M, Aanderud S, Franco C, Freyschuss B, Isaksen GA, Ueland T, Rosen T. Medical observation, compared with parathyroidectomy, for asymptomatic primary hyperparathyroidism: a prospective, randomized trial. J. Clin. Endocrinol. Metab. 2007;92(5):1687–92.
  161. Rubin MR, Maurer MS, McMahon DJ, Bilezikian JP, Silverberg SJ. Arterial stiffness in mild primary hyperparathyroidism. J. Clin. Endocrinol. Metab. 2005;90(6):3326–30.
  162. Walker MD, Fleischer J, Rundek T, McMahon DJ, Homma S, Sacco R, Silverberg SJ. Carotid vascular abnormalities in primary hyperparathyroidism. J. Clin. Endocrinol. Metab. 2009;94(10):3849–56.
  163. Concistrè A, Grillo A, La Torre G, Carretta R, Fabris B, Petramala L, Marinelli C, Rebellato A, Fallo F, Letizia C. Ambulatory blood pressure monitoring-derived short-term blood pressure variability in primary hyperparathyroidism. Endocrine 2018;60(1):129–137.
  164. Storvall S, Ryhänen EM, Heiskanen I, Sintonen H, Roine RP, Schalin-Jäntti C. Surgery Significantly Improves Neurocognition, Sleep, and Blood Pressure in Primary Hyperparathyroidism: A 3-Year Prospective Follow-Up Study. Horm. Metab. Res. 2017;49(10):772–777.
  165. Ejlsmark-Svensson H, Rolighed L, Rejnmark L. Effect of Parathyroidectomy on Cardiovascular Risk Factors in Primary Hyperparathyroidism: A Randomized Clinical Trial. J. Clin. Endocrinol. Metab. 2019;104(8):3223–3232.
  166. Ekmekci A, Abaci N, Colak Ozbey N, Agayev A, Aksakal N, Oflaz H, Erginel-Unaltuna N, Erbil Y. Endothelial function and endothelial nitric oxide synthase intron 4a/b polymorphism in primary hyperparathyroidism. J. Endocrinol. Invest. 2009;32(7):611–6.
  167. Bilezikian JP, Brandi ML, Eastell R, Silverberg SJ, Udelsman R, Marcocci C, Potts JT. Guidelines for the management of asymptomatic primary hyperparathyroidism: Summary statement from the fourth international workshop. In: Journal of Clinical Endocrinology and Metabolism.Vol 99. Endocrine Society; 2014:3561–3569.
  168. Danzi S, Klein I. Thyroid hormone and blood pressure regulation. Curr. Hypertens. Rep. 2003;5(6):513–520.
  169. Prisant LM, Gujral JS, Mulloy AL. Hyperthyroidism: a secondary cause of isolated systolic hypertension. J. Clin. Hypertens. (Greenwich). 2006;8(8):596–599.
  170. Kempf T, Wollert KC. Risk stratification in critically ill patients: GDF-15 scores in adult respiratory distress syndrome. Crit. Care 2013;17(4):173.
  171. Berta E, Lengyel I, Halmi S, Zrínyi M, Erdei A, Harangi M, Páll D, Nagy E V, Bodor M. Hypertension in Thyroid Disorders. Front. Endocrinol. (Lausanne). 2019;10:482.
  172. Fountoulakis S, Tsatsoulis A. Molecular genetic aspects and pathophysiology of endocrine hypertension. Hormones (Athens). 5(2):90–106.
  173. Iglesias P, Acosta M, Sánchez R, Fernández-Reyes MJ, Mon C, Díez JJ. Ambulatory blood pressure monitoring in patients with hyperthyroidism before and after control of thyroid function. Clin. Endocrinol. (Oxf). 2005;63(1):66–72.
  174. Völzke H, Alte D, Dörr M, Wallaschofski H, John U, Felix SB, Rettig R. The association between subclinical hyperthyroidism and blood pressure in a population-based study. J. Hypertens. 2006;24(10):1947–53.
  175. Biondi B, Cooper DS. The clinical significance of subclinical thyroid dysfunction. Endocr. Rev. 2008;29(1):76–131.
  176. Streeten DHP, Anderson GH, Howland T, Chiang R, Smulyan H. Effects of thyroid function on blood pressure. Recognition of hypothyroid hypertension. Hypertension 1988;11(1):78–83.
  177. Iqbal A, Schirmer H, Lunde P, Figenschau Y, Rasmussen K, Jorde R. Thyroid stimulating hormone and left ventricular function. J. Clin. Endocrinol. Metab. 2007;92(9):3504–3510.
  178. Stabouli S, Papakatsika S, Kotsis V. Hypothyroidism and hypertension. Expert Rev. Cardiovasc. Ther. 2010;8(11):1559–65.
  179. Ittermann T, Thamm M, Wallaschofski H, Rettig R, Völzke H. Serum thyroid-stimulating hormone levels are associated with blood pressure in children and adolescents. J. Clin. Endocrinol. Metab. 2012;97(3):828–34.
  180. Cai Y, Ren Y, Shi J. Blood pressure levels in patients with subclinical thyroid dysfunction: a meta-analysis of cross-sectional data. Hypertens. Res. 2011;34(10):1098–105.
  181. Colao A, Ferone D, Marzullo P, Lombardi G. Systemic Complications of Acromegaly: Epidemiology, Pathogenesis, and Management. Endocr. Rev. 2004;25(1):102–152.
  182. Rizzoni D, Porteri E, Giustina A, De Ciuceis C, Sleiman I, Boari GEM, Castellano M, Muiesan ML, Bonadonna S, Burattin A, Cerudelli B, Agabiti-Rosei E. Acromegalic patients show the presence of hypertrophic remodeling of subcutaneous small resistance arteries. Hypertens. (Dallas, Tex. 1979) 2004;43(3):561–5.
  183. Lombardi G, Galdiero M, Auriemma RS, Pivonello R, Colao A. Acromegaly and the cardiovascular system. Neuroendocrinology 2006;83(3–4):211–7.
  184. Colao A, Terzolo M, Bondanelli M, Galderisi M, Vitale G, Reimondo G, Ambrosio MR, Pivonello R, Lombardi G, Angeli A, degli Uberti EC. GH and IGF-I excess control contributes to blood pressure control: results of an observational, retrospective, multicentre study in 105 hypertensive acromegalic patients on hypertensive treatment. Clin. Endocrinol. (Oxf). 2008;69(4):613–20.
  185. Bielohuby M, Roemmler J, Manolopoulou J, Johnsen I, Sawitzky M, Schopohl J, Reincke M, Wolf E, Hoeflich A, Bidlingmaier M. Chronic growth hormone excess is associated with increased aldosterone: a study in patients with acromegaly and in growth hormone transgenic mice. Exp. Biol. Med. (Maywood). 2009;234(8):1002–9.
  186. Sardella C, Urbani C, Lombardi M, Nuzzo A, Manetti L, Lupi I, Rossi G, Del Sarto S, Scattina I, Di Bello V, Martino E, Bogazzi F. The beneficial effect of acromegaly control on blood pressure values in normotensive patients. Clin. Endocrinol. (Oxf). 2014;81(4):573–81.
  187. González B, Vargas G, de Los Monteros ALE, Mendoza V, Mercado M. Persistence of Diabetes and Hypertension After Multimodal Treatment of Acromegaly. J. Clin. Endocrinol. Metab. 2018;103(6):2369–2375.
  188. Ullah MI, Uwaifo GI, Nicholas WC, Koch CA. Does vitamin d deficiency cause hypertension? Current evidence from clinical studies and potential mechanisms. Int. J. Endocrinol. 2010;2010:579640.
  189. Kazi M, Geraci SA, Koch CA. Considerations for the diagnosis and treatment of testosterone deficiency in elderly men. Am. J. Med. 2007;120(10):835–40.
  190. Shabsigh R, Arver S, Channer KS, Eardley I, Fabbri A, Gooren L, Heufelder A, Jones H, Meryn S, Zitzmann M. The triad of erectile dysfunction, hypogonadism and the metabolic syndrome. Int. J. Clin. Pract. 2008;62(5):791–8.
  191. Sattler FR, Castaneda-Sceppa C, Binder EF, Schroeder ET, Wang Y, Bhasin S, Kawakubo M, Stewart Y, Yarasheski KE, Ulloor J, Colletti P, Roubenoff R, Azen SP. Testosterone and growth hormone improve body composition and muscle performance in older men. J. Clin. Endocrinol. Metab. 2009;94(6):1991–2001.
  192. Widdowson WM, Gibney J. The effect of growth hormone replacement on exercise capacity in patients with GH deficiency: a metaanalysis. J. Clin. Endocrinol. Metab. 2008;93(11):4413–7.
  193. Mekala KC, Tritos NA. Effects of recombinant human growth hormone therapy in obesity in adults: a meta analysis. J. Clin. Endocrinol. Metab. 2009;94(1):130–7.
  194. Zhang J, Ge R, Matte-Martone C, Goodwin J, Shlomchik WD, Mamula MJ, Kooshkabadi A, Hardy MP, Geller D. Characterization of a novel gain of function glucocorticoid receptor knock-in mouse. J. Biol. Chem. 2009;284(10):6249–59.
  195. Michailidou Z, Carter RN, Marshall E, Sutherland HG, Brownstein DG, Owen E, Cockett K, Kelly V, Ramage L, Al-Dujaili EAS, Ross M, Maraki I, Newton K, Holmes MC, Seckl JR, Morton NM, Kenyon CJ, Chapman KE. Glucocorticoid receptor haploinsufficiency causes hypertension and attenuates hypothalamic-pituitary-adrenal axis and blood pressure adaptions to high-fat diet. FASEB J. 2008;22(11):3896–907.

 

 

 

 

 

Existing and Emerging Molecular Targets for The Pharmacotherapy of Obesity

ABSTRACT

 

Obesity is pandemic and a multidisciplinary approach is critical for its management. Anti-obesity treatment includes lifestyle modifications combined with anti-obesity medications. Anti-obesity drugs target either central nervous system pathways, which regulate sensations of satiety and fullness, or peripheral modulators of digestion, metabolism and lipogenesis. Combined anti-obesity agents is a novel, promising field, especially the co-administration of gut hormone analogues with centrally acting molecules. Consequently, it is hoped that in the near future, individualized pharmacological management of obesity could be meaningfully achieved by targeting different pathways governing energy homeostasis and weight regulation.  This chapter reviews potential molecular targets of the energy homeostasis system along with new anti-obesity drugs currently under investigation.

INTRODUCTION                                                                                                                      

 

The pathophysiology that leads to obesity is considered a novel field for research. Understanding human metabolism and the homeostatic mechanisms of weight regulation includes comprehension of the interaction between central nervous system and peripheral modulators of weight maintenance. Current anti-obesity molecular pharmacotherapy is based on single molecule anti-obesity drugs that act either via enhancement of satiety feeling, inhibition of hunger, or triggering of catabolism. However, on average, the weight-lowering effects of these medications are modest at best and side effects are common.

 

According to current clinical practice guidelines for pharmacological management of obesity published in 2015 by The Endocrine Society, if a patient’s weight is not responsive to lifestyle intervention, weight loss pharmacotherapy can be offered for a BMI ≥27kg/m2 when an obesity-related comorbidity is present, or when the BMI is ≥30kg/m2 (1). In fact, pharmacologic weight management should be considered in patients who meet these weight criteria and have any of a number of chronic conditions in which obesity is considered to play a major role, including type 2 diabetes mellitus (T2DM), cardiovascular disease, hypertension, dyslipidemia, obstructive sleep apnea, nonalcoholic fatty liver disease, certain cases of malignancies (i.e. endometrial, breast, colon) (2), osteoarthritis, depression (3), and infertility (4).

 

Currently, there are six anti-obesity medications that have received US Food and Drug Administration (FDA) approval: orlistat, phentermine, phentermine/topiramate extended release (ER), lorcaserin, naltrexone sustained release (SR)/bupropion SR, and liraglutide (the only injectable formulation). At the same time, the European Medicines Agency (EMA) has approved only three of these: orlistat, bupropion/naltrexone and liraglutide.

 

Considering the extent to which obesity impairs health alone or through expression of one or more of these comorbidities, the need for new molecular pharmaceutic agents is crucial. As detailed below, future weight-loss medications will be based on our knowledge of key regulatory sites of weight regulation and energy homeostasis so as to achieve greater efficacy while minimizing off-target side effects, characteristics that are necessary for approval by both American and European drug regulatory agencies.

 

TARGETS OF PHARMACOTHERAPY IN THE MANAGEMENT OF OBESITY

 

Novel insights provided by pathophysiology indicate the presence of a complex homeostatic system in which information about the energy reserve status and the meal quality and content is relayed from the periphery (gastrointestinal tract, pancreas, and adipose tissue) via specific orexigenic and anorexigenic peptides and hormones to the central nervous system (CNS). Peripheral peptide hormones are released postprandially and travel in the circulation to bind to their receptors in the homeostatic regulatory centers in the CNS, notably the arcuate nucleus (ARC) of the hypothalamus and the dorsal vagal complex (DVC) in the brainstem medulla. The ARC contains neurons expressing key orexigenic neurotransmitters, agouti-related peptide (AgRP) and neuropeptide Y (NPY), as well as anorexigenic neurotransmitters, proopiomelanocortin (POMC) and cocaine- and amphetamine-regulated transcript (CART). Food intake is thus modulated by complementary mechanisms so as to maintain energy and weight homeostasis. New drug therapies have begun to focus on combination therapy using medications that target more than one of these central pathways, thereby achieving more favorable weight loss outcomes. In addition, combining treatments may provide a better safety profile given that lower doses of each drug when used together may achieve better weight loss than higher doses of a single agent (see Figure 1 below).

 

Factors That Influence Appetite

 

The regulation of satiety and appetite depends on the interaction of three major factors: biological systems, modern macro-environmental exposures, and micro-environmental influences. Biological systems are shaped by genetic and epigenetic influences from early-life events that govern development of orexigenic and anorexigenic neuro-hormonal pathways involved in the pathophysiology of obesity. Modern macroenvironment (food production, consumption, availability, social structure, weather influencing physical activity, television and technology, cultural norms, endocrine disruptors) and microenvironment (nutrition, exercise, sleep, stressful lifestyle, circadian rhythm) play an important role in the conformational development of cognitive and emotional brain regions, thus predisposing to the obese phenotype.

 

Genetic Factors of Physical Activity

 

Specific genes predict to what extent adults remain active. This is evidenced in a study examining identical twins in which environmental factors shared by children at age 13 accounted for 78% to 84% of sport participation, whereas genetic differences provided no contribution at all. At the age of 17 to 18 the genetic influences represented 36% of the variance in the level of participation in sports, and by age 18 to 20, genetic factors were responsible for almost all (85%) of the differences in participation in sports.

Figure 1. Sites of Action of the Most Important Anti-Obesity Drugs

CENTRALLY-ACTING ANTI-OBESITY DRUGS

 

Monoamine Neurotransmitter Modulators

 

With the exception of the glucagon-like peptide 1 (GLP-1) receptor agonist liraglutide, currently available weight loss medications act on the central nervous system to enhance dopamine, norepinephrine, and serotonin action to enhance satiety, diminish hunger, and consequently affect weight loss. Drug combinations have opened new horizons as they use multiple neural pathways, leading to better results with less adverse events. Recently, a review of fifty reports involving 43,443 subjects compared the efficacy of the central acting anti-obesity drugs lorcaserin (5HT2c receptor agonist), naltrexone-bupropion (opioid receptor antagonist combined with a norepinephrine releasing agent that stimulates POMC neuronal firing), phentermine-topiramate (a norepinephrine and dopamine modulator plus a carbonate anhydrase inhibitor), and liraglutide. It was found that the maximal mean weight loss relative to placebo was -3.06, -6.15, -7.45, and -5.5kg after 1 year with mean weight regain +0.48kg, +0.91kg, +1.27kg, +0.43kg the following year, respectively.  In these studies, the one-year drop-out rate was 40.9%, 49.1%, 34.9%, 24.3%, respectively (5).

 

Leptin, Leptin Analogues and Leptin Sensitizers

 

Leptin is a protein secreted primarily by white adipose tissue (WAT). It directly stimulates anorexigenic POMC neurons and inhibits adjacent orexigenic NPY neurons in the ARC of the hypothalamus, thus promoting satiety, increasing energy expenditure, and resulting in weight loss (6). Circulating levels of leptin increase with adiposity and decline following body weight reduction; the latter might be implicated in the total and resting energy expenditure reduction seen after weight loss. The discovery of leptin in 1994 was a seminal event in obesity research. It helped to establish that body weight should be viewed as a disorder with a strong biological basis rather than simply the result of poor lifestyle choices. Studies with congenitally leptin-deficient, severely obese subjects revealed that administration of physiological doses of leptin decreased food intake and body weight (7). Obese individuals, however, are leptin-resistant and have increased circulating leptin levels. Whether administration of leptin could overcome leptin resistance and exert an anti-obesity effect was tested in a placebo-controlled study with 47 obese men and women given varying doses of recombinant human leptin (0.03 mg/kg and 0.30 mg/kg, respectively) for 24 weeks and advised to eat 500 kcal less than body requirements each day. A dose-dependent decrease in body weight was shown, ranging from -1.3 kg in the placebo group to -1.4 kg in the 0.03 mg/kg leptin-treated group, and to -7.1 kg in the 0.30 mg/kg leptin-treated group (8). These results suggested that leptin resistance can be overcome with high doses of leptin but resulting in only modest weight loss similar to currently approved medications.  In addition, whether these effects can be sustained long-term is not known. Reports were similar from animal studies testing the effect of leptin sensitizers targeting the protein tyrosine phosphatase-1B (PTP1B)(9)(10) or the chemical chaperones that repair ER stress, including 4-phenyl butyric acid (PBA) and tauroursodeoxycholic acid (TUDCA) (11), each of which demonstrated reduced food intake and body weight. Like leptin treatment, sustainability of these anti-obesity effects is still not clear.

 

Weight loss is associated with reduction in energy expenditure, which makes long term weight loss maintenance difficult (12). Furthermore, 6 days of high fat diet in mice suffice to dramatically decrease the levels of phosphorylated signal transducer and activator of transcription 3 (p-STAT3) in the arcuate nucleus (13) while short term overfeeding of normal weight mice can lead to an increase of leptin resistance (14). Besides the inefficiency of leptin analogues as monotherapy, combinations of leptin with amylin (15), fibroblast growth factor 21(FGF21), exendin4, (16), or a GLP-1/glucagon co-agonist (17) were proposed. Only the combination with the GLP-1/glucagon co-agonist has shown improvement of leptin sensitivity (18). Apart from diet, stress of endoplasmic reticulum contributes to leptin resistance (19). Several plant-derived substances, such as celastrol (20) and withaferin (21) have been tested in diet-induced obese rodents for improvement of this pathway that leads to leptin resistance.

 

METRELEPTIN

 

Metreleptin (MYALEPT) is an injectable human recombinant leptin analogue approved in Japan for metabolic disorders including lipodystrophy and in USA as first-line treatment for non-HIV related forms of generalized lipodystrophy (leptin deficiency, congenital/acquired lipodystrophy) (22). A previous indication for hypothalamic amenorrhea has been withdrawn (23). (see Table 1)

 

Table 1. Metreleptin (MYALEPT)

FDA approved/Phase

Approved in Japan for lipodystrophy disorders and in USA for non-HIV lipodystrophy

Mechanism of action

Human recombinant leptin injectable analogue

Clinical Benefits

↓blood glucose, triglycerides, hepatic fatty steatosis

Adverse events

Headache, hypoglycemia, decreased weight, abdominal pain

-previous indication for hypothalamic amenorrhea discontinued

 

PRAMLINTIDE/METRELEPTIN

 

The combination of amylin-leptin (pramlintide-metreleptin) has been shown to be effective in the treatment of obesity. The anti-obesity properties of the combined treatment with pramlintide and metreleptin (pramlintide/metreleptin) were tested and showed a significant weight reduction of 12.7 ± 0.9% (11.5 ± 0.9 kg) without plateauing in obese patients during a 20-week trial period (24). The sponsors subsequently announced positive results from a 28-week proof-of-concept study with pramlintide and metreleptin combination treatment in overweight or obese subjects. The combination treatment reduced body weight on average by 12.7%, significantly more than treatment with pramlintide alone (8.4%), which is interpreted as 10 pounds more weight loss with the combined treatment. Remarkably, subjects receiving pramlintide/metreleptin continued to lose weight until the end of the study, compared to those treated with pramlintide alone, whose weight loss had stabilized towards the end of the study. The magnitude of weight loss was found to be dose-dependent and baseline BMI-dependent. Patients with a starting BMI less than 35 kg/m2 experienced the best weight loss efficacy with the combined treatment. A year later, the results of the 52-week blinded, placebo-controlled Phase II extension study of pramlintide/metreleptin were announced. The results indicated sustained and robust weight loss through the combined treatment; again, the most robust efficacy was seen in patients with a BMI less than 35 kg/m2 (25). Although the pramlintide/metreleptin combination seemed to be the next promising anti-obesity drug to be marketed, the sponsors discontinued its development in 2011, following commercial reassessment of the program (26).

 

Melanocortin-4 Receptor Agonists

 

The melanocortin system has a highly significant role in the hypothalamic regulation of body weight and energy expenditure. Leptin inhibits the release of the orexigenic neuropeptides orexin and melanocortin-concentrating hormone (MCH) in the lateral hypothalamic area (LHA) through the release of CART and melanocyte-stimulating hormone (α-MSH). The latter derives from the cleavage of POMC by prohormone convertase-1 and acts via melanocortin-3 and -4 receptors (MC3R, MC4R) activation. α-MSH emerged as a promising novel anti-obesity drug, and intranasal administration of the melanocortin sequence MSH/ACTH4-10 to normal-weight subjects was shown to acutely increase subcutaneous WAT lipolysis (27) and decrease body fat by 1.7 kg, when administered for six weeks (28). It eventually proved not to induce any significant reduction in body weight or body fat when compared with placebo in a 12-week study of 23 overweight men.

 

In preclinical studies, obese primates treated for eight weeks with the MC4R agonist RM-493 (Setmelanotide) lost an average of 13.5% of their body weight, with significant improvements in both insulin sensitivity and cardiovascular function. In June 2014, the results from the first human Phase II trial were released, testing the hypothesis that an MC4R agonist increases resting energy expenditure in obese subjects. A total of 12 obese but otherwise healthy individuals were randomized and completed both RM-493 and placebo periods in this double-blind, placebo-controlled, two-period crossover study. Analysis of the data indicates that short-term treatment with RM-493 increased resting energy expenditure significantly (by 6.4% vs placebo), thus suggesting RM-493 may be clinically effective for treating obesity. In 2015, administration of Setmelanotide to obese individuals for a limited time increased resting energy expenditure (REE) by 6.4% and shifted substrate oxidation to fat (29). Currently, Setmelanotide is being tested as a therapeutic option for rare genetic disorders of obesity such as POMC deficiency, heterozygous deficiency obesity, and POMC epigenetic disorders (30-32). (see Table 2)

 

Table 2. Setmelanotide (RM-493)

FDA approved/Phase

Phase II

Mechanism of action

MC4R-agonist

Weight loss vs placebo

13.5%

Clinical Benefits

↑insulin sensitivity, cardiovascular function, energy expenditure, ↓ body weight

-tested for POMC deficiency, heterogenous deficiency obesity, POMC epigenetic disorders

Adverse events

Headache, arthralgia, nausea, spontaneous penile erection, female genital sensitivity

 

Melanin-Concentrating Hormone (MCH) Antagonists

 

The melanocortin-concentrating hormone (MCH) is an important orexigenic neuropeptide in the LHA. Its release is stimulated by NPY and inhibited by leptin, exerting its orexigenic effects through the MCH1 receptor (MCHR1) (33). Like NPY, MCH exerts pleiotropic effects on locomotor activity, sensory processing, anxiety, aggression, and learning. Thus, despite the role of MCH in hunger stimulation, MCHR1 blockade as an anti-obesity target is questionable because such inhibition could elicit undesirable side effects. In animal models, MCH antagonists have consistently demonstrated efficacy in reducing food intake acutely and in inhibiting body weight gain when given chronically (34). Five compounds have reached testing in human subjects. Although they were reported as well-tolerated, none has proceeded to Phase II studies. A major issue with many lead compounds is increased cardiovascular risk due to drug-induced QTc prolongation (35). Among others, the MCHR1 antagonist AMG 076 entered Phase I safety and tolerability testing in 2004, but there have been no subsequent reports of its status since 2005. The MCHR1 antagonist GW-856464 also entered Phase I studies in 2004; however, in 2010 it was reported that low bioavailability precluded further development. The MCHR1 antagonist NGD-4715 was safe and well-tolerated in a Phase I clinical trial, but its development ceased in 2013. Similarly, despite the reported tolerability and indication of efficacy of the MCHR1 antagonist ALB-127158, its development was terminated before the initiation of Phase II studies. Finally, the longest (28-day) Phase I study with BMS-830216, a pharmacological antagonist of MCH signaling (36) produced no indications of weight loss or reduced food intake and the compound did not proceed to Phase II studies.

 

Subtype-Selective Serotonin-Receptor Agonists

 

Central serotonin participates in feeding behavior and energy balance modulation, reducing food intake in animals and human beings.  This finding was supported by reports of two selective serotonin reuptake inhibitors (SSRIs) developed to treat depression, fluoxetine and sertraline, being associated with non-sustained weight loss in obese subjects. Thus, agonists to appropriate serotonin receptors are potentially valuable drugs. The serotonin (5-HT) system directly modulates the hypothalamic POMC (anorexigenic) and NPY (orexigenic) networks, enhancing satiety and causing hypophagia. These effects are mediated by 5-HT2C and 5-HT1B receptors, located on hypothalamic POMC and NPY neurons, respectively. Through the 5-HT1B receptors, serotonin inhibits the NPY/Agrp neurons, thereby decreasing the GABAergic inhibitory input to POMC cells; while through the 5-HT2C receptors it directly activates the anorexigenic POMC neurons. Via these actions, serotonin increases α-MSH and decreases AgRP release into the hypothalamic melanocortin system, promoting satiety. Between 1973 and 2000 there was an explosion in the pharmaceutic industry regarding central acting anti-obesity drugs. Three non-selective serotonin-receptor agonists were approved by FDA: fenfluramine (1973-1997), the combination phentermine-fenfluramine (1992-1997), and dexfenfluramine (1996-1997). These were all 5-HT1b agonists characterized for their ability to inhibit food consumption, but also had effects on other serotonin receptors that lead to unacceptable side effects (cardiac valvular thickening) and were voluntarily withdrawn from the market.

 

In 1997, when fenfluramine and dexfenfluramine were discontinued by the manufacturer, sibutramine, a serotonin and norepinephrine reuptake inhibitor emerged. Sibutramine has only little clinical relevance as an antidepressant but enhances weight loss due to an increase in energy expenditure and inhibition of food intake (37). In addition to weight loss, sibutramine was found to improve fasting levels of insulin, triglycerides, and high-density lipoprotein cholesterol. Sibutramine was also associated with increase of blood pressure, cardiovascular events, and cardiac arrhythmias (38). For these reasons, FDA withdrew it in 2010.

 

LORCASERIN

 

As activation of the 5-HT1B receptor has been implicated in both primary pulmonary hypertension (39) and valvopathy (40), the 5-HT2C receptor subtype has been proposed as a target for therapeutic intervention to allow weight loss. Several potent and selective 5-HT2Creceptor agonists proved to be effective in suppressing food intake and inducing weight loss in rodents, including WAY-163909 (41), CP-809101 (42), and vabicaserin (43). However, only lorcaserin (APD356) moved into clinical testing. Lorcaserin (Belviq) is a selective 5-HT2c receptor agonist, which belongs in the third generation of 5-HT-based anti-obesity drugs (44). It activates hypothalamic POMC neurons to induce satiety and decrease food intake but does not affect energy expenditure. Through actions on midbrain dopaminergic tone, it has been shown to suppress binge- food behaviors. Its action in addictive disorders is currently under investigation (45). Based on the outcome of the BLOOM (46) and BLOSSOM trials (47), in 2012 the FDA approved lorcaserin as an addition to a reduced-calorie diet and exercise for eligible patients (48). The efficacy of lorcaserin appears similar to that of orlistat (mean difference in weight loss between active and placebo treated groups approximately 3 to 4 kg) and perhaps slightly less than that of phentermine-topiramate. The impact of lorcaserin in patients with T2DM and BMI: 27-45kg/m2 was examined in the BLOOM-DM trial which showed a reduction of body weight by approximately 5kg versus 1.6kg in the placebo group, as well as significant decreases in heart rate, HDL levels, and waist circumference. Valvopathy was shown not to occur in excess with treatment and lorcaserin was generally well tolerated, with a low incidence of side effects such as headache, dizziness, fatigue, nausea. After the results of BLOOM-DM trial, a potential combination of GLP-1RA and 5-HT2A/C is now under investigation (49).

 

In a multicenter, randomized, double-blind, placebo-controlled, parallel-group study involving12,000 overweight and obese patients with cardiovascular disease or multiple cardiovascular risk factors (CAMELLIA-TIMI 61), the effect of long-term treatment with lorcaserin on major cardiovascular events and conversion to T2DM over a 5-year period were examined. After one year of treatment, 5% weight loss was observed in 38.7% and 17.4% in the lorcaserin and the placebo groups, respectively. Regarding cardiac risk, the lorcaserin group was non-inferior to the placebo group with slightly better values in cardiac risk factors (blood pressure, heart rate, glycemic control, lipid profile). Adverse events were rare in both groups, apart from the incidence of serious hypoglycemia in the lorcaserin group in those with diabetes managed using insulin or sulfonylureas (50, 51). In addition, lorcaserin administration decreased the incidence of T2DM by 19% in patients with prediabetes and by 23% in patients without diabetes. In patients with T2DM, lorcaserin resulted in a reduction of 0.33% in HbA1c compared with placebo at 1 year from a mean baseline of 7.0%. (see Table 3, 4)

 

Table 3. Lorcaserin (Belviq)

FDA approved/Phase

2012

Mechanism of action

Selective Serotonin 2C agonist

Weight loss vs placebo

3-4kg

Clinical Benefits

↓food intake, heart rate, HDL levels, waist circumference, HbA1c

Adverse events

Headache, dizziness, fatigue, nausea, dry mouth, constipation, heart valvopathy

-In diabetics: hypoglycemia, headache, back pain, cough, fatigue, risk of serotonin syndrome/neuroleptic malignant syndrome, valvular heart disease

 

 

Table 4. Clinical Trials of Lorcaserin

Clinical trial

Patients

Dose

Treatment, placebo from baseline

% of patients losing ≥5% of baseline weight

Comment

 

BLOSSOM

1-year randomized, double-blind, placebo-controlled trial

(2011)

4008 patients (18-65 y.o., BMI- 30-45kg/m2 or 27-29.9kg/m2 with comorbidity) randomized in a 2:1:2 ratio

i.10mg x2 po

 

ii.10mg x1 po

 

iii.placebo

i.-5.8kg

 

 

ii.-4.7kg

 

 

iii.-2.9kg

i.47.2%

 

 

ii.40.2%

 

 

iii.25%

Exclusion criteria: recent cardiovascular events, diabetes mellitus, BP >150/95mmHg

BLOOM

2-year randomized, double-blind, placebo-controlled trial

(2010)

 

3182 adults (mean BMI-36.2kg/m2) randomized to lorcaserin twice daily or placebo group. After 52 weeks, the placebo group continued placebo and lorcaserin group selected placebo or lorcaserin for 52 weeks

i.10mg x2 po

 

ii. placebo

i.-5.8kg

 

 

ii.-2.2kg

i.47.5%

 

 

ii.20.3%

Weight loss was greater in the group which continued lorcaserin for the second year

BLOOM-DM

1-year randomized, double-blind, placebo-controlled trial

(2012)

604 patients (HbA1c: 7-10%, BMI-27-45kg/m2, treatment with metformin, sulfonylurea or both)

i.10mg x2 po

 

ii.10mg x1 po

 

iii.placebo

i.-4.7kg

 

 

ii.-5.0kg

 

 

iii.-1.6kg

i.37.5%

 

 

ii.44.7%

 

 

iii.16.1%

↓heart rate, HDL levels, waist circumference in lorcaserin treated groups

NO valvopathy was statistically significant

CAMELLIA-TIMI 61

3.3-year randomized, placebo-controlled trial

(2018)

 

12,000 patients overweight/obese-three subgroups

A. diabetes

B. prediabetes

C. normoglycemic

i.10mg x2/day

 

ii. placebo

At 1 year the mean treatment difference:

 A: -2.6kg

 B: -2.8kg

 C: -3.3kg

 

 

At 1 year compared with placebo:

A: 37.4%

B: 39.7%

C: 42.3%

↓ BMI, waist circumference, waist-to-hip ratio, HbA1c, reduced microvascular complications

 

Bupropion

 

Bupropion is a dopamine and norepinephrine-reuptake inhibitor that has been marketed as an anti-depressant and for smoking cessation. Previous animal studies have clearly shown a dose-dependent satiety effect of bupropion following intraperitoneal injection (52). The acute effects of dopamine and noradrenaline reuptake inhibition on energy homeostasis demonstrated their additive effects on short-term food intake (53). Bupropion increases dopamine activity and POMC neuronal activation, thereby reducing appetite and increasing energy expenditure (54). Whether the acute meal terminating effects of bupropion documented in animal studies could be translated into long-term weight loss efficacy in humans was addressed by three clinical trials with overweight and obese adults (55, 56, 57) using different treatment doses (100 to 400 mg/d) and duration (up to 24 weeks). They have all shown bupropion to have dose-dependent modest weight reducing efficacy, plus a safe profile. One study that assessed the anti-obesity efficacy of bupropion over two years reported maintenance of weight loss during the continuation phase, while another demonstrated its efficacy even in depressed patients. Although the weight loss effect of bupropion was superior in non-depressed patients compared to those suffering from depression, the fact that bupropion was well-tolerated and effective in this group of patients provides a potential valuable adjunctive therapy to elevate mood in depressed subjects in whom weight gain secondary to antidepressant therapy is an issue. Cardiovascular effects, such as a rise in blood pressure and tachycardia, were usually mild, while the risk of seizure, which was high with the original bupropion formulation, has been significantly reduced with the advent of bupropion-SR and bupropion-ER.

 

An interesting finding of the previous studies was that the rather modest weight loss effect of bupropion reached a plateau by 24 weeks of treatment. This could be explained by the molecular pathophysiology of the weight reducing effects of bupropion, which directly stimulates the hypothalamic POMC neurons that in turn release α-MSH and β-endorphin. α-MSH mediates the anorectic effect of POMC activation, whereas β-endorphin exerts negative feedback on POMC neurons via opioid receptors (58). The latter possibly points to one of the compensatory mechanisms that limits long-term efficacy of bupropion and other weight loss modalities.

 

Naltrexone

 

Naltrexone is an opioid receptor antagonist. By blocking opioid receptors on the POMC neurons, feedback inhibition is prevented further increasing POMC activity. Monotherapy with opioid antagonists to decrease short-term food intake has been tested (59). Naltrexone failed to produce consistent or clinically meaningful weight loss, even at large doses (300 mg/d) (60), implying that a single opioid mechanism is unlikely to explain all aspects of ingestive behavior.

 

Bupropion/Naltrexone Sustained Release (SR)

 

The combined bupropion/naltrexone (NB) therapy induced significantly greater weight loss on a diet and exercise program over 56 weeks compared to monotherapy and placebo (61). In 2014, the FDA approved this combination (Contrave, Mysimba) for body weight management in adults who are overweight and obese. This combined therapy of opioid antagonist and aminoketone antidepressant is titrated over four weeks to the maximum dose. NB has shown remarkable benefit in patients with binge-eating disorder (BED) and concomitant alcohol abuse, but this result needs further evaluation (62). Four major 56-week phase III randomized, double-blind, placebo-controlled trials have shown the therapeutic effect of ΝΒ SR (COR-I, COR-II, COR-BMOD, COR-DIABETES) in different dosage combinations (see Table 6). In COR-I, the weight loss ratio on NB 16/360mg, NB 32/360mg or placebo was -5.0%, -6.1%, -1.3% (P<0.00) respectively. In COR-II, the weight loss ratio on NB 32/360mg or placebo was -6.4%, -1.2% (P<0.001) (63). In COR-BMOD, NB SR 32/360mg plus intensive behavioral modification was compared with the behavioral modification alone as a therapeutic option. The weight loss ratio was -11.5% versus -7.3% (P<0.001), respectively (64). Recently, COR-Diabetes has included patients with T2DM with or without antidiabetic treatment. The NB SR 32/360mg treatment resulted in -5.1% weight loss versus -1.8% in the placebo group (P<0.001). NB treatment resulted in a HbA1c reduction, cardiovascular benefit, and lipid profile improvement (65). Due to FDA request for further investigation of the effect of NB on major cardiovascular events, the LIGHT trial was created. Unfortunately, this trial terminated early following recommendation by the academic leadership of the study because confidential interim data were publicly released by the sponsor (66). (See Table 5, 6)

 

Table 5. Bupropion/Naltrexone Sustained Release (Contrave, Mysimba+

FDA approved/Phase

2014

Mechanism of action

Aminoketone antidepressant/Opioid antagonist

Weight loss vs placebo

4.8kg

Clinical Benefits

↓ appetite

Adverse events

Nausea, constipation, headache, vomiting, dizziness, insomnia, dry mouth, suicidal ideation, increase blood pressure/heart rate, hepatotoxicity, angle-closure glaucoma Uncontrolled hypertension, seizures, anorexia nervosa/bulimia, chronic opioid use, coadministration with MAO inhibitors

 

Table 6: Clinical trials of Naltrexone/Bupropion SR

Clinical trial

Patients

Dose

Treatment, placebo from baseline

% of patients losing ≥5% of baseline weight

Comment

 

COR I

1-year randomized, double-blind, placebo-controlled trial

(2010)

1742 patients randomly categorized in a 1:1:1 ratio

i.16/360mg po

 

ii.32/360mg po

 

iii. placebo

i.-5.0%

 

 

ii.-6.1%

 

 

iii.-1.3%

 

i.39%

 

 

ii.48%

 

 

iii.16%

 

COR II

1-year randomized, double-blind, placebo-controlled trial

(2013)

1496 patients randomly categorized in a 2:1 ratio to NB 32/360mg or placebo; patients on NB with <5% weight loss in 28-44 week were reassigned to continue 32/360mg or increase daily dose to NB 48/360mg

i.32/360mg (or increased daily dose 48/360mg)

 

ii.placebo

i.-6.4%

 

 

 

 

 

ii.-1.2%

 

i.50.5%

 

 

 

 

 

ii.17.1%

Random reassignment to higher dose did not change weight loss results

COR-BMOD

1-year randomized, double-blind, placebo-controlled trial

(2011)

793 patients with obesity randomly categorized in a 1:3 ratio

i. BMOD+ NB (32/350mg)

 

ii. BMOD+ placebo

i.-11.5%

 

 

 

ii.-7.3%

i.66.4%

 

 

 

ii.42.5%

The efficacy of NB is obvious, and a lifestyle change can increase weight loss

COR-DIABETES

1-year randomized, double-blind, placebo-controlled trial

(2013)

505 patients overweight/obese and T2DM with/without oral anti-hypoglycemic agents randomly categorized in a 2:1 ratio

i.32/360mg

 

ii. placebo

i.-5.0%

 

ii.-1.8%

i.44.5%

ii.18.9%

↓HbA1c, certain improvements in CVD risk factors.

↑ nausea, constipation, vomiting

 

 

Zonisamide

 

Given the pathophysiology behind the anti-obesity efficacy of the selective serotonin-receptor agonists and the dopamine-reuptake inhibitors, an ideal drug would combine serotonergic and dopaminergic activity. This is exactly the case of Zonisamide, a marketed antiepileptic drug that exerts dose-dependent biphasic dopaminergic (67) and serotonergic (68) activity. Its weight loss efficacy was investigated by a double-blind, placebo-controlled trial which reported a 32-week mean weight loss of 9.2 kg (1.7 kg) (9.4% loss) for the Zonisamide group (dose administered up to 600 mg/d) compared with 1.5 kg (0.7 kg) (1.8% loss) for the placebo group (P<0.001); Zonisamide was generally well-tolerated with only a few adverse effects (69). The findings were similar when the long-term effectiveness and tolerability of Zonisamide for weight control was examined in psychiatric outpatients using various psychotropic medications; the mean BMI reduction achieved was 0.8±1.7 kg/m2 and ranged from -2.9 kg/m2 to 4.7 kg/m2 (p<0.001), while the drug was generally safe and well-tolerated (70). Zonisamide was also assessed in the treatment of binge-eating (BE) disorder where it proved to be effective in reducing binge-eating frequency, severity of illness, and weight; however, the reports regarding its tolerability were conflicting (71). (see Table 7).

 

Table 7. Zonisamide

Mechanism of action

Selective serotonin-receptor agonist and dopamine-reuptake inhibitor

Weight loss vs placebo

7.8kg

Clinical Benefits

Assess in the treatment of binge-eating disorder

Adverse events

Nausea, headache, insomnia

 

Zonisamide/Bupropion SR

 

Whether the anti-obesity efficacy of Zonisamide is increased when combined with bupropion (dopamine and norepinephrine -reuptake inhibitor) has been evaluated in a few Phase II clinical trials with different combined doses; the bupropion SR/Zonisamide SR combination is marketed under the trade name Empatic. In its 24-week, double-blind, placebo-controlled Phase IIb trial (72), patients completing 24 weeks of bupropion SR 360 mg/Zonisamide SR 360 mg therapy lost 9.9% of their baseline body weight, or 22 pounds, compared to 1.7% for placebo patients (p<0.001). Of patients who completed 24 weeks of therapy, 82.6% lost at least 5% of their baseline body weight and 47.7% lost at least 10% of their baseline body weight compared to 18.9% and 5.7% of placebo patients, respectively (p<0.001 for both). Patients experienced significant weight loss as early as by their first post-baseline visit at week four. Importantly, patients continued to lose weight until the end of the trial period with no evidence of a weight loss plateau. Early results showed that patients lost an average of 14% over 48 weeks. Improvements were observed in key markers of cardiometabolic risk such as waist circumference, triglycerides, fasting insulin, and blood pressure. The most commonly reported adverse events for all patients were headache, insomnia, and nausea. The most common adverse events leading to discontinuation were insomnia, headache, and urticaria (hives). There were no serious adverse events attributed by investigators to the study drug. There were no statistically or clinically meaningful differences between the drug and placebo on measures of cognitive function, depression, suicidality or anxiety. These reports revealed a significant weight-reduction effect for the combination Bupropion/Zonisamide. However, the safety concerns (73) will need to be addressed in the upcoming Phase III studies before firm conclusions about its safety profile can be drawn. (see Table 8)

 

Table 8. Zonisamide/Bupropion (Empatic)

FDA approved/Phase

Phase II completed

Mechanism of action

Selective serotonin-receptor agonist and dopamine-reuptake inhibitor/dopamine and norepinephrine reuptake inhibitor

Weight loss vs placebo

9.9% of their baseline weight

Clinical Benefits

↓cardiometabolic risk

Adverse events

Headache, insomnia, nausea, urticaria

 

Topiramate

 

Topiramate is another anticonvulsant agent associated with weight loss. It is a sulphamate-substituted fructose that is approved as an antiepileptic/antimigraine agent and has multiple effects on the CNS, including action on the orexigenic GABA systems causing appetite suppression (74). A 6-month dose-ranging study in obese human subjects addressing its anti-obesity efficacy at doses of 64, 96, 192, and 384 mg/day (in divided twice-daily dosing) concluded that all doses produced significantly greater weight loss compared to placebo, and that weight loss in the 192 mg/day group was similar to the 384 mg/day group (75). This is important as topiramate has been associated with several neuropsychiatric effects, especially when administered at high doses (of 192 mg/d or more). Another study investigating the weight loss efficacy and safety of topiramate doses of 96, 192, and 256 mg/day over a 1-year period in obese subjects using the immediate release form tablets (before the development of the controlled-release formulation). Clinically significant weight loss (7.0, 9.1, and 9.7% of their baseline body weight for the doses of 96, 192, and 256 mg/day, respectively), was reported compared to 1.7% body weight loss in the placebo group (P<0.001) plus improvements in blood pressure and glucose tolerance (76). Finally, several other studies investigated the therapeutic effect of topiramate in patients with BED and bulimia (77) that are both associated with obesity; the results were very promising regarding control of symptoms in both disorders. (see Table 9)

 

Table 9. Topiramate

FDA approved/Phase

Phase II completed

Mechanism of action

Sulphamate-substituted fructose acts on GABA system

Weight loss vs placebo

7.0%(96mg),9.1%(192mg), 9.7% (256mg/day)

Clinical Benefits

Assess in the treatment of binge-eating, bulimia

Adverse events

Headache, insomnia, nausea, urticaria

 

Phentermine

 

Phentermine is a sympathomimetic amine, which has anorexigenic action, that also releases insignificant quantities of dopamine. Thus, it is characterized by lower abuse potential (78). Its main mechanism of action involves catecholamine release in the hypothalamus resulting in enhanced satiety feeling and reduction of food intake (79). The most common side effects of phentermine as a sympathomimetic drug is heart rate increase, hypertension, dizziness, dry mouth, insomnia, irritability, and gastrointestinal disorders (80). Phentermine was the first FDA approved anti-obesity drug in 1959 for those aged >16 years old, but for only short-term use (maximum 3 months). The reason for the time limit is because the pharmaceutic industry had not updated labeling since 1959. In 1968, in a double-blind, placebo-controlled trial, 108 overweight or obese women were categorized into three groups that received continuously or intermittently (every 4 weeks) dosed phentermine or placebo, respectively. The weight loss was -12.2kg, -13.0kg or -4.8kg, respectively (81).

 

Currently, the off-label long-term use of phentermine is indicated only if there is clinical benefit, stable blood pressure and pulse rate in the absence of cardiovascular history or substance abuse disorders. In a recently published retrospective cohort study, 13,972 patients were observed for 6, 12 and 24 months after phentermine initiation. They were categorized in five groups based on the time of phentermine administration: short-term use, short-term intermittent use, medium-term continuous use, medium-term intermittent use, long-term continuous use. Weight-loss, changes in blood pressure, heart rate, and incidence of cardiovascular events (myocardial infarction, stroke, angina, coronary artery bypass grafting, carotid artery intervention, death) were examined. Weight loss was greater among off-label groups than referent group of short-term use with results depending on the duration of phentermine initiation. Specifically, at six months, short-term intermittent patients lost 1.8% further body weight relative to short-term single patients and medium-term intermittent patients lost 5.6% further body weight relative to short-term single patients. At twelve months, the medium-term intermittent use group lost further 5.6% body weight relatively to the short-term use group. At twenty-four months, long-term the continuous use group lost 7.4% additional body weight in comparison with the short-term use group. Concerning safety of phentermine, changes in heart rate and diastolic blood pressure were insignificant at six, twelve, and twenty-four months. Interestingly, although the referent group showed a slight increase in systolic blood pressure (+0.5-3.2 mmHg) at twenty-four months, all groups had slightly lower systolic blood pressure than the referent group at twelve- and twenty-four-months follow-up period. Lastly, the incidence of major cardiovascular events was low. So, it was shown that the off-label over three months therapy with phentermine was superior to short–term administration with greater weight-loss effect and cardiovascular safety. More studies with fewer limitations should follow in order to support these findings (82). In 2013, a clinical trial comparing phentermine as monotherapy or as part of a combined therapy, took place and resulted in a weight loss of 5.1% at 28 weeks follow-up period in favor of the combined phentermine/topiramate group.(see Table 10)

 

Table 10. Phentermine

FDA approved/Phase

1959

Mechanism of action

Norepinephrine release and minor dopamine release

Weight loss vs placebo

0.23kg/week

Clinical Benefits

Lower abuse potential

Adverse events

Stimulation, insomnia, dry mouth, constipation, primary pulmonary hypertension

Contraindicated in cardiovascular disease, coadministration with MAO inhibitors, hyperthyroidism, glaucoma, drug abuse

 

Phentermine/Topiramate ER

 

Because of dose-related side effects seen with topiramate treatment including suicidality, metabolic acidosis, acute myopia, and secondary angle closure glaucoma, a lower dose of topiramate was used (in a special controlled release formulation) in a novel anti-obesity drug called Qsymia. The main mechanism of action of Phentermine/Topiramate extended release(ER) is possibly the alteration of various neurotransmitters, including inhibition of voltage-dependent sodium channels, glutamate receptors, and carbonic anhydrase as well as potentiation of γ-aminobutyrate activity (83).Two large randomized, double-blind, placebo-controlled trials took place (EQUIP and CONQUER) followed by a 2-year extension trial (SEQUEL). In the EQUIP trial 1,267 patients with BMI>35kg/m2were allocated in two groups and received phentermine/topiramate ER 3.75/23mg and 15/92mg, respectively, once daily. With 59.9% of the patients discontinuing, this trial found no statistically significant difference between the two groups regarding weight reduction (84). In the CONQUER trial 2,487 patients were allocated in three groups and received phentermine/topiramate ER 7.5/46mg, phentermine/topiramate ER 15/92mg, and placebo, respectively. The results were in favor of the combined therapy while the greater dosage resulted in greater weight loss with mean weight loss -7.8kg, -9.8kg, and -1.2kg in the three respective groups (85). Patients who completed the CONQUER trial entered the SEQUEL trial for 52 weeks. The weight loss as percentage of the initial weight was -9.3%, -10.5% and -1.8% in the three respective groups. A statistically significant improvement of lipid profile, glycemic control, and waist circumference in the phentermine/topiramate ER groups was reported (86). Based on the positive results from three Phase III studies, in 2012 FDA approved topiramate/phentermine extended-release as an adjunct to a reduced-calorie diet and increased physical activity for chronic weight management in eligible adults. Meanwhile however, approval was denied by European regulatory authorities, who cited potential risk to the heart and blood vessels, psychiatric side effects, and cognitive side effects in explaining their decision (see Table 11, 12).

 

Table 11. Topiramate/Phentermine Extended Release (ER) (Qsymia)

FDA approved/Phase

2012

Mechanism of action

Norepinephrine release, GABA modulation, voltage-gated ion channel modulation, stop of AMPA/kainite excitatory glutamate receptors and carbonic anhydrase

Weight loss vs placebo

6,6kg

Clinical Benefits

↓ lipid profile, HbA1c, waist circumference

Adverse events

Paresthesia, dizziness, dysgeusia, insomnia, constipation, dry mouth, fetal toxicity, metabolic acidosis, cognitive impairment

Contraindicated in: Glaucoma, hyperthyroidism, coadministration with MAO inhibitors

 

Table 12. Clinical Trials of Phentermine/Topiramate ER

Clinical trial

Administration

N

Treatment, placebo from baseline

% of patients losing ≥5% of baseline weight

Comment

 

CONQUER

Double-blind, placebo-controlled trial over 1 year

(2011)

4-week titration+ 52 weeks of treatment:

15/92mg po

or 7.5/46mg po

2487 patients (BMI:27-45kg/m2 with 2+ risk factors

i.15/92mg

 

ii.7.5/46mg

 

iii. placebo

 

 

 

 

 

 

i.-9.8kg

 

ii.-7.8kg

 

iii.-1.2kg

 

 

 

 

 

 

i.70%

 

ii.62%

 

iii.21%

↑improvement in blood pressure, waist circumference, lipid levels, fasting glucose and insulin

SEQUEL

2-year study overall;1-year extension of CONQUER

(2012)

227 patients completed the original blinded treatment

 

 

i.15/92mg

 

ii.7.5/46mg

 

iii. placebo

i.-10.9kg

 

ii.-9.6kg

 

iii.-2.1kg

i.79.3%

 

ii.75.2%

 

iii.30%

 

 

Neuropeptide Y (NPY) Inhibitors

 

The ARC NPY neurons inhibit the anorexigenic POMC neurons (via NPY Y1 and Y5 receptors) and promote the release of the orexigenic neuropeptides orexin and MCH in the LHA, thus promoting food intake. Therefore, NPY blockade could be a promising target for body weight management. Animal experiments (in mice) have shown that pharmacologic blockade or genetic deletion of the Y1- and Y5-receptors reduces food intake and weight, with Y1-receptor signaling appearing to be the major mediator of the orexigenic effects of NPY. However, NPY is the most abundant central neuropeptide and regulates many functions beyond feeding; thus, targeting NPY neurons/Y receptors specifically for obesity is not easy and could result in unacceptable side effects. In addition, experimental medical blockade of NPY signaling with the Y5-receptor antagonist MK-0577 failed to cause any significant weight loss in a 1-year clinical trial (87). On the other hand, the oral, once-daily, centrally acting selective Y5-receptor antagonist velneperit, previously known as S-2367, induced a mean placebo-adjusted weight loss of 5.0% from initial weight (p <0.0001) over 54 weeks of therapy and was accompanied by improvement of lipid profile and waist circumference reduction (88).Nevertheless, velneperit did not proceed in markets due to disappointing results in phase IIb trials. However, the combined Y1/Y5-receptor antagonism may prove more effective, though we are not aware of any Y1/Y5-receptor antagonist in development to date. In contrast to Y1 and Y5, the Y2- and Y4-receptors are the targets of the satiety hormones PYY and pancreatic polypeptide (PP), respectively, and, as mentioned below, two drugs, a Y2/Y4-receptor agonist (obinepitide and a selective Y4-receptor agonist (TM30339)) are in Phase I/II clinical trials and are yielding results that appear quite promising as regards weight loss. A combined anti-obesity medication of velneperit/orlistat is under way (phase II clinical trial), also with promising results (89).

 

Dopamine antagonists

 

The mesolimbic dopamine system was proven to play a critical role in compulsive overeating or binge eating, which is one of the main reasons why people become overweight or obese. There is some evidence that blocking the action of dopamine in animals can reduce food intake, particularly of foods that are high in fat and sugar. GSK 598809 is a D3 antagonist that blocks dopamine. Preliminary data from human studies failed to show any significant effect on body weight control (90).

 

Tesofensine

 

Tesofensine (TE) is a presynaptic inhibitor of norepinephrine, dopamine, and serotonin. Like sibutramine, it suppresses appetite and may result in significant weight loss, as this was shown when given for the treatment of Parkinson’s disease, but also in a multi-dose, dose-ranging trial where 203 obese patients were randomly assigned to Tesofensine (0.25, 0.5, and 1.0 mg) or placebo once daily. Phase II testing of the drug has been completed. After 24 weeks, mean weight reduction was greater in the Tesofensine groups (-6.7, -11.3, -12.8 kg, for the three doses, respectively) compared with placebo (-2.2 kg). Additionally, an improvement in lipid profile and glycemic control was observed. A dose-dependent increase in blood pressure was observed along with a 7.4bpm increase in pulse rate in the 0.5mg/day group. Adverse events such as headache and mood alterations were also present in all groups especially in the 1mg/day group (91). In another trial, 32 males were allocated in two groups and received 2mg/day Tesofensine and placebo, respectively. The interesting point in this trial was that the patients were free to consume their usual amounts levels of food and exercise as usual. However, in the Tesofensine group they lost 1.8kg over 2 weeks because Tesofensine increased visual analog scale ratings of satiety and 24h fat oxidation in comparison with placebo. Even if an increase in total energy expenditure was not observed, an increase in sleeping energy expenditure was found. Altogether, Tesofensine induces weight loss by promoting the satiety feeling and slightly increasing metabolic rate (92). The effect of Tesofensine in appetite sensations was evaluated in another phase II trial, in which patients were allocated in 4 groups and received 0.25mg, 0.5mg, 1mg and placebo, respectively, for 24 weeks. For the first 12 weeks, a dose-dependent increase in the satiety feeling was noticed even though this feeling faded away as the trial was in progress (93). In 2010, a study on the abuse effect of Tesofensine, bupropion, atomoxetine, and placebo in comparison to d-amphetamine took place and concluded that the studied substances had no abusive action (94). Tesofensine has been shown to increase both blood pressure and pulse rate. In 2018, a phase III clinical trial was powered by the pharmaceutic industry producing Tesofensine. In this study 372 patients were allocated in three groups and received Tesofensine 0.25mg, 0.5mg and placebo. Furthermore, a combination of Tesofensine/metoprolol is recently being examined against hypothalamic injury-induced obesity and Prader-Willi syndrome (95). (see Table 13)

 

Table 13. Tesofensine

FDA approved/Phase

Phase III

Mechanism of action

Triple monoamine reuptake inhibitor of dopamine, norepinephrine, serotonin

Weight loss vs placebo

4.5-10.6%

Clinical Benefits

Pharmacological similar to sibutramine

↓ appetite, body weight, lipid profile, blood glucose

Adverse events

Headache, mood alterations, potentially increase heart rate, blood pressure, psychiatric disorders

.

Lisdexamfetamine dimesylate

 

Another sympathomimetic, Lisdexamfetamine dimesylate, at certain doses appears effective in decreasing binge-eating days in patients with BED compared with placebo, according to a study published online by JAMA Psychiatry (96). The study included 259 and 255 adults with BED in safety and intention-to-treat analyses, respectively. Patients received lisdexamfetamine 30, 50 or 70 mg/day or placebo. BE days per week decreased in the 50 mg and 70 mg groups but not in the 30 mg group compared with placebo. Confirmation of these findings in ongoing clinical trials may result in improved pharmacologic treatment for moderate to severe BED.

 

Cannabinoid-1 Receptor (CB1) Antagonists

 

Among other neurotransmitter systems, the cannabinoid system modulates the hypothalamic melanocortin and NPY feeding networks. It has been shown that administration of cannabinoid-1 receptor (CB1) agonists and antagonists induces hyperphagia and hypophagia, respectively. These observations led to development of rimonabant, a cannabinoid-1 receptor antagonist, for the treatment of obesity, which was shown quite effective in promoting weight loss; however, it increased the incidence of mood-related disorders (97). As a result, in 2009, rimonabant was withdrawn from the market and the development of other cannabinoid-1 receptor antagonists for the treatment of obesity has also been discontinued. Before withdrawal, rimonabant was shown to have advantages in glycemic control and cardiovascular events (98). In 2010, another CB1 antagonist (AM6545) was found to have less psychological side effects and to induce satiety feeling and weight loss in animal studies (99). (see Table 14)

 

Table 14. Cannabinoid Type-1 Receptor Antagonists (SR141716, AM251, AM6545)

Mechanism of action

Antagonism of cannabinoid type-1 receptors stimulates anorexigenic signaling

Clinical Benefits

↓ body weight, blood glucose, cardiovascular events

-AM6545: has limited CNS penetration

Adverse events

Mood alterations

 

Human Chorionic Gonadotropin (hCG)

 

Human chorionic gonadotropin (hCG) in the form of subcutaneous injection and oral or sublingual diet drops has been advertised as aiding in weight loss of one to two pounds daily, absence of hunger, and maintenance of muscle tone. Clinical trials, however, failed to support this claim (100). In fact, FDA recommended avoiding buying over-the-counter weight loss products which contain hCG. One might ask why the hCG diet has so many enthusiastically supporting it. The reason may be that this diet needs to be accompanied by severe calorie restriction, to only 500-800 calories per day. Anyone following such recommendations is bound to lose weight, if only short-term. Most crucially, though, since hCG has been reported to induce serious side effects, this drug should not be used for the treatment of obesity. In addition, very low-calorie diets have not been shown to be superior to conventional diets for long-term weight loss, plus they have risks, such as gallstone formation, irregular heartbeat, and an imbalance of electrolytes. Therefore, if weight loss is the goal, there are safer ways to make it happen.

 

Nesfatin-1

 

Nesfatin-1 is a satiety molecule, which was first described in rats and is derived from its precursor molecule nucleobindin2 (NUCB2) (101). It is expressed both centrally in hypothalamic food intake-regulatory nuclei, the nucleus paraventricular and the arcuate nucleus, and peripherally, in the stomach, pancreas, adipose tissue, and testis. In the gastric oxyntic mucosa, nesfatin-1 is co-expressed with the orexigenic peptide ghrelin in X/A-like cell in rats and humans. The anorexigenic action of nesfatin-1 is based on its ability to cross the blood-brain barrier. It is notable that NUCB2/nesfatin-1 not only decreases food intake, gastric emptying, and small intestine motility, but also reduces glucose and increases insulin levels (102). Intracerebroventricular (icv) injection of full length nesfatin-1 caused a significant reduction of food intake in rats and mice (103). These findings suggest that downstream signaling might be altered, a hypothesis to be further investigated. The fact that nesfatin-1 acts in a leptin-independent way, indicates that it might be a new molecular target in the pharmacotherapy of obesity. The identification of the yet unknown nesfatin-1 receptor will allow the development of nesfatin-1 agonists and antagonists. Whether peripheral nesfatin-1 is primarily involved in the regulation of food intake is questionable and should be further investigated.

 

GASTROINTESTINAL AND PANCREATIC PEPTIDES THAT REGULATE FOOD INTAKE

 

The gut-brain axis plays an important role in food consumption regulation. During food intake, information regarding meal quality and content and short-term alterations in nutrient status is relayed from the gastrointestinal (GI) tract and pancreas to the brain which in turn determines meal size. Apart from feeding, a few satiation signals optimize these processes by influencing gastrointestinal motility and secretion. Several peptides have been identified that mediate this GI system-brain communication including satiety signals such as gastrin releasing peptide (GRP), cholecystokinin (CCK), peptide YY (PYY), glucagon-like peptide-1 (GLP-1), pancreatic polypeptide, glucagon, and amylin, as well as the orexigenic peptide ghrelin. While the anorexigenic peptides are secreted during feeding, ghrelin is secreted before meals and acts to increase hunger and meal initiation. Some of the GI and pancreatic peptides implicated in the regulation of food intake act directly on regions of the brain involved in the regulation of food intake, including the ARC in the hypothalamus and the area postrema, while others act outside of the CNS.  For example, modulating the activity of neurons such as the vagus nerve, which projects to the nucleus of the solitary tract in the brain stem.

 

CCK and CCK1R Agonists

 

CCK is the first described intestinal satiation peptide (104). It is produced by the mucosal I cell (105) of the duodenum and jejunum, and the enteric nervous system, in response to luminal nutrients, especially lipids and proteins. Through endocrine and/or neural mechanisms, CCK regulates numerous GI functions, including satiation, by acting on two CCK-specific receptors: the CCK receptor 1 (CCK1R), expressed mainly in the GI system, and the CCK2R that predominates in the brain. The vagus nerve plays a critical role in CCK-induced satiation as it contains CCK1R, indicating the afferent pathway through which CCK relays satiation signals from the GI to the hindbrain region. Corroborating this hypothesis is the well-documented attenuation of CCK-induced satiation following abdominal subdiaphragmatic vagotomy (106). In addition, CCK inhibits gastric emptying, thereby augmenting gastric distention and mechanoreceptor stimulation, which in turn augments the anorectic effects of CCK (107). Despite the satiety effect of CCK, its potential as an anti-obesity target is questionable. Human studies with intravenously infused CCK carboxy-terminal octapeptide (CCK-8) have shown decreases in meal size and duration in a dose-dependent manner (108). However, the CCK satiating effects were very short-lived, usually not lasting more than 30 minutes, which raises issues as to its importance in long-term body weight regulation. In an animal study, chronic CCK administration with up to 20 peripheral injections per day, although reducing meal size, was associated with increased meal frequency, leaving body weight unaffected (109). Finally, the reports from trials testing CCK1R agonists as potential anti-obesity drugs were disappointing (110). It is currently suggested that there might be a role for CCK in body weight regulation not as a monotherapy but possibly as an adjunctive/synergistic therapy to long-term adiposity signals, such as leptin (111).

 

Glucagon-Like Peptide-1 Analogues

 

The dominant role of GI in satiation (112) is mediated not only by the gastric mechanoreceptors and upper intestinal neuropeptides such as CCK, but also by gut satiation peptides that are secreted from lower-intestine enteroendocrine cells in response to ingested food. They in turn diffuse through interstitial fluids to activate nearby nerve fibers and/or enter the bloodstream to function as hormones and augment the perception of GI fullness by acting in specific parts of the CNS. There is a well-defined duodenal-ileal communication (the ileal brake) via which the proximal intestine informs the distal intestine as to meal quality and content so that the latter modulates/restricts feeding duration, proximal GI motility, and gastric emptying, while it also regulates metabolic responses to nutrient intake. GLP-1 appears to engage such a mechanical and behavioral brake effect on eating and is produced primarily by L cells in the distal small intestine and colon. Along with glucagon and oxyntomodulin, GLP-1 is cleaved from proglucagon, which is expressed in the gut, pancreas, and brain. The GLP-1 equipotent bioactive forms GLP17–36 and GLP17-37 are rapidly inactivated in the circulation by dipeptidyl peptidase-4 (DPP4). Therefore, GLP-1 analogues that have a slightly different molecular structure, but a significantly longer duration of action compared to wild GLP-1 have been used for therapeutic interventions in patients with diabetes, in whom they significantly improved glycemic control, fasting plasma glucose, β-cell function, and probably β-cell regeneration. Currently, the GLP-1 analogues used in clinical practice for diabetes control are exenatide, lixisenatide, dulaglutide, liraglutide, and semaglutide. Beyond the improved glycemic control achieved, clinical studies have also demonstrated anorectic effects and significant weight loss via these agents (113, 114). Although the exact mechanisms by which GLP1 induces anorexia are not yet fully known, it is suggested that vagal and possibly direct central pathways are involved (115). The GLP-1 receptor R (GLP1R) is the principle mediator of the anorectic effects of GLP-1 (116) and is expressed by the gut, pancreas, brainstem, hypothalamus, and vagal-afferent nerves (117).

 

LIRAGLUTIDE

 

Its mechanism of action is both central and peripheral targeting satiety centers of the brain and regulating glucose metabolism. It is the only injectable medication for obesity and is titrated from 0.6mg to 3.0mg over 4 weeks. The most common side effects of liraglutide and generally of GLP1 analogues are gastrointestinal (nausea, diarrhea, constipation, vomiting, dyspepsia, abdominal pain) and rarely pancreatitis. The product has a boxed warning stating that thyroid C-cell tumors have been seen in rodents but the relevance of this in humans is uncertain. The drug should not be used in patients with a personal or family history of medullary thyroid carcinoma (MTC) or in patients with multiple endocrine neoplasia syndrome type 2. Three major trials, SCALE-Obesity, Prediabetes, SCALE-Diabetes, SCALE-Maintenance, have established the therapeutic benefit of liraglutide for weight loss. The SCALE-Obesity, Prediabetes evaluated liraglutide in patients who were overweight and obese but did not have diabetes. The study included 3,731 individuals who were assigned to treatment with liraglutide 3 mg or a placebo. Patients were also counseled on diet and exercise. At the end of the 56-week trial, the liraglutide group lost an average 8% (7.2kg) of their body weight compared to 2.6% (2.8kg) in the placebo group (118); net weight loss was 4.4kg. In the SCALE-Diabetes trial, 846 adults who were overweight or obese and had T2DM were allocated to receive either daily 3.0mg liraglutide or placebo, with mean weight loss -6.0% and -2.0%, respectively (119). In the SCALE-Maintenance trial, 422 adults who were overweight or obese and had lost >5% of initial body weight with a calorie restriction diet were allocated to receive either liraglutide or placebo, respectively, with mean weight loss -6.2% and -0.2%, respectively (120).Recently, Saxenda (liraglutide 3.0mg) has asked for a label update based on the results of the LEADER trial, which studied the effects of the lower dose version of liraglutide (1.8 mg) used to treat diabetes. According to this trial, which examined a population with T2DM and established cardiovascular disease,1.8mg liraglutide daily showed statistically significant reduction of cardiovascular death, of non-fatal myocardial infarction (heart attack), and of non-fatal stroke by 13% versus placebo, when added to standard care. (121) (see Table 15, 16).

 

Table 15. Liraglutide (Saxenda)

FDA approved/Phase

2014

Mechanism of action

Glucagon-like peptide-1 agonist

Weight loss vs placebo

4.4kg

Clinical Benefits

↓cardiovascular death, non-fatal myocardial infarction, non-fatal stroke

Adverse events

Nausea, hypoglycemia (serious if co-administrated with insulin), gastrointestinal disorders, fatigue, dizziness, abdominal pain, increased lipase, acute pancreatitis, acute gallbladder disease, increase heart rate, suicidal ideation, thyroid c-cell tumors seen in mice

Contraindicated in: History of medullary thyroid carcinoma or multiple endocrine neoplasia 2

 

Table 16. Clinical Trials of Liraglutide

Clinical trial

Patients

Dose

Treatment, placebo from baseline

% of patients losing ≥5% of baseline weight

Comment

 

SCALE-Obesity+Prediabetes

1-year randomized, double-blind, placebo-controlled trial

(2015)

3731 patients overweight/obese without DM (61.2% had prediabetes) randomly divided into two groups

i.3.0mg sc once daily

 

ii. placebo

i.-8.4kg

 

 

ii.-2.8kg

i.63.2%

 

 

ii.27.1%

Improvement of body weight, glycemic index, blood pressure, waist circumference

SCALE-Diabetes

1-year randomized, double-blind, placebo-controlled trial

(2015)

846 adults with T2DM overweight/obese

i.3.0mg sc once daily

 

ii. placebo

i.-6.0%

 

 

ii.-2.0%

i.54.2%

 

 

ii.21.4%

More GI disorders in the liraglutide group. No pancreatitis was reported

SCALE-Maintenance

1-year randomized, double-blind, placebo-controlled trial

(2013)

422 adults overweight/obese who had lost ≥5% of initial body weight during a calorie-restriction period were randomized

i.3.0mg sc once daily

 

 

ii. placebo

i.-6.2%

 

 

 

ii.-0.2%

i.81.4%

 

 

 

ii.48.9%

A combination of liraglutide, diet, exercise induced further weight loss and improvement in certain cardiovascular risk factors

 

SEMAGLUTIDE

 

Semaglutide is a novel long-acting GLP1 analogue indicated for T2DM and awaiting approval for obesity at higher doses. The efficacy of this anti-obesity drug has been proven by the SUSTAIN 1-6 trials. In these trials, patients who were overweight or obese, with and without T2DM, with or without antidiabetic medications, were allocated in groups which received semaglutide in two different dosages (0.5mg or 1.0mg) or placebo or another anti-diabetic therapy. The superiority of semaglutide 1.0mg against semaglutide 0.5mg or placebo or another anti-diabetic agent was obvious (122). In SUSTAIN 7, Semaglutide administered in subcutaneous injections once weekly was compared with Dulaglutide. Mean weight loss was greater in the group which received 1.0mg semaglutide (-4.9kg) vs the groups that received 0.5mg semaglutide (-3.6kg), 1.5mg Dulaglutide (-3kg), and0.75mg Dulaglutide (-2.3kg). Additionally, oral semaglutide is currently approved for the treatment of T2DM. In order to avoid malabsorption, semaglutide is administrated 30 minutes before breakfast. Apart from semaglutide, other oral GLP-1 agonists, such as TTP054/TTP-054 and ZYOG1, are under investigation (122). Two other trials, STEP, which studies the effects of semaglutide in patients with obesity, and SELECT, which investigates the cardiovascular effects of semaglutide in patients with obesity are currently underway (123). PIONEER, which examines the cardiovascular safety of oral administration of semaglutide in patients with T2DM, recently showed the non-inferiority of this medication to placebo (124). (see Table 17).

 

Table 17. Clinical Trials of Semaglutide

Clinical trial

Study Design

Dose

Treatment, placebo from baseline

% of patients losing ≥5% of baseline weight

SUSTAIN 1

Double-blinded

For 30 weeks

i.0.5mg sc once weekly

ii.1.0mg sc once weekly

iii. placebo

i. -3.7kg

ii. -4.5kg

iii. -1.0kg

i.37%

ii.45%

iii.7%

SUSTAIN 2

Double-blinded

Duration: 56 weeks

i.0.5mg sc once weekly

ii.1.0mg sc once weekly

iii. sitagliptin 100mg per po once daily

i.-4.3kg

ii.-6.1kg

iii.-1.9kg

i.46%

ii.62%

iii.18%

SUSTAIN 3

Open-label

Duration:56 weeks

i.1.0mg sc once weekly

ii. exenatide extended release 2.0mg

i.-5.6kg

ii.-1.9kg

i.52%

ii.17%

SUSTAIN 4

Open-label

Duration: 30 weeks

i.0.5mg sc once weekly

ii.1.0mg sc once weekly

iii. insulin glargine

i.-3.5kg

ii.-5.2kg

iii.+1.2kg

i.37%

ii.51%

iii.5%

SUSTAIN 5

Double-blinded

Duration:30 weeks

ii.0.5mg sc once weekly

ii.1.0mg sc once weekly

iii. placebo

i.-3.7kg

ii.-6.4kg

iii.-1.4kg

i.42%

ii.66%

iii.11%

SUSTAIN 7

Open-label

Duration: 40 weeks

i.0.5mg sc once weekly

ii.0.75mg dulaglutide sc once weekly

iii.1.0mg sc once weekly

iv.1.5mg dulaglutide sc once weekly

i.-4.6kg

ii.-2.3kg

 

iii.-6.5kg

iv.-3.0kg

i.44%

ii.23%

 

iii.63%

iv.30%

SUSTAIN 6

(CVD outcomes)

Double-blinded

Duration:104 weeks

i. 0.5mg sc once weekly

ii.1.0mg sc once weekly

iii. placebo 0.5mg

iv. placebo 1.0mg

i.-3.6kg

ii.-4.9kg

iii.-0.7kg

iv.-0.5kg

Non inferior

SUSTAIN 8

Phase 3b

Semaglutide vs canagliflozin

 

 

 

SUSTAIN 9

Semaglutide as an add-on to SGLT2 monotherapy or in combination with either metformin or sulfonylurea

 

 

 

 

OTHER LONG-ACTING GLP-1 ANALOGUES

 

Other long-acting GLP-1 analogues are currently being investigated for weight loss in addition to diabetes treatment. Once-daily 13-week treatment with 20 μg or 30 μg of lixisenatide reduced body weight significantly more compared to placebo (-3 kg for lixisenatide 20 μg; p<0.01, -3.47 kg for lixisenatide 30 μg; p<.01, -1.94 kg for placebo) (125). Current findings regarding CJC-1134-PC, which is a conjugate of exendin-4 and recombinant human albumin and represents a once-weekly glucagon-like peptide-1 receptor agonist, suggest that it provides similar reduction in body weight compared with exenatide twice-daily. It may have a more favorable adverse event profile which might improve patient compliance and probably total weight loss in the long-term (126). Finally, albiglutide and taspoglutide are two novel GLP-1 analogues currently being investigated. A recent review that examined the efficacy, safety, and perspective for the future of the once-weekly GLP-1 receptor agonists exenatide, taspoglutide, albiglutide, LY2189265 and CJC-1134-PC, and compared them to the currently available agonists, exenatide BID and liraglutide QD, concluded that the long-acting agonists are not superior compared to the currently used exenatide BID and liraglutide QD regarding weight loss (127).In a separate development, an orally administered PYY3-36 and GLP-1 combination has been formulated using a sodium N-[8-(2-hydroxybenzoyl) amino] caprylate (SNAC) carrier (127). Early studies revealed that the neuropeptides delivered orally in this way had a pharmacodynamic profile consistent with the reported pharmacology, were rapidly absorbed by the gastrointestinal tract, and reached concentrations several-fold higher than those seen naturally postprandially (128). Oral GLP-1 (2-mg tablet) alone and in combination with PYY3-36 (1-mg tablet) showed enhanced fullness at meal onset and induced a significant reduction in energy intake. Exenatide-CCK (129) and Liraglutide-Setmelanotide (130) have been also introduced as different combined anti-obesity therapies which act synergistically on POMC-deficient patients.

 

Single Molecule Multi-Agonists

 

The main therapeutic idea of this category is based on the concept that a single molecule could target multiple receptors (at least two; multi-agonist), thus allowing synergistic action of both pharmaceutical agents.

 

GLUCAGON-LIKE PEPTIDE 1/GLUCAGON

 

As mentioned before, GLP-1 analogues are effective anti-obesity medications and improve glucose intolerance. Glucagon has direct action on the liver by stimulating gluconeogenesis and glycogenolysis (131). It can even result in hyperglycemia and T2DM. Of note, patients with T2DM are characterized by impaired glucagon secretion. However, glucagon in CNS decreases food intake, increases energy expenditure via brown fat thermogenesis, decreases fat accumulation via lipolysis and lipid synthesis inhibition, improves cardiac performance, inhibits gastric motility, and stimulates autophagy. In 2009, the first human study announced that low-dose co-infusion of GLP-1 and glucagon could decrease food intake and increase energy expenditure (132). Therapy with a GLP-1/glucagon multi-agonist was created when amino acids 17, 18, 20, 21, 23 of glucagon were substituted in the glucagon molecule by the respective GLP-1 residues (133). The alanine at position 2 of the peptide was substituted with Aminoisobuturic acid (Aib) to protect the molecule from DDP-IV inactivation, and a lactam bridge was introduced between amino acids 16 and 20 to stabilize the secondary structure to ensure glucagon receptor potency. Once weekly administration of this pharmaceutical agent, for 4 weeks, in diet-induced obesity in mice, resulted in improvement of obesity, hepatic steatosis, glucose control, and lipid profile. Increase in energy expenditure was observed only with the multi-agonist therapy, but not with the glucagon monotherapy. Moreover, it was found that therapy with the multi-agonist improved leptin sensitivity in DIO mice (134). Different GLP-1/glucagon multi-agonists are currently under investigation (135). Interestingly, an oxyntomodulin multi-agonist was under investigation concurrently with the GLP1/glucagon multi-agonist.

 

OXYNTOMODULIN

 

Oxyntomodulin (OXM) is a 37-amino acid anorexigenic peptide hormone produced in the L-cells of the distal small intestine and colon, where it co-localizes with GLP-1 and PYY. Animal studies have shown weight reduction and improved glucose metabolism following chronic OXM injections beyond that explained by food intake restriction, suggesting an additional effect of OXM on energy expenditure. Just like GLP1, OXM is a product of proglucagon gene believed to modulate energy homeostasis at least in part via GLP1R, although its GLP1R binding affinity is about 100 times lower than that of GLP1 (136). Centrally however, GLP1 and OXM have different targets, as OXM activates neurons in the hypothalamus (137), whereas GLP1 acts in the hindbrain and other autonomic control areas (138). In human studies, acute anorectic effect of OXM was demonstrated by intravenously infused OXM (139). A reduction in food intake was also seen and retained during chronic administration in a 4-week trial with OXM injections three times a day 30 minutes before meals in a group of overweight and obese volunteers (n = 14). OXM reduced nutrient intake (35% ± 9%) resulting in significant weight loss compared to placebo (2.3 ± 0.4 kg vs 0.5 ± 0.5 kg, respectively). The findings of another study with twelve overweight or obese human volunteers who underwent a randomized, double-blinded, placebo-controlled study were similar; an ad libitum test meal was used to measure energy intake during intravenous infusions of either PYY3-36 or OXM or combined PYY3-36/OXM. Again, OXM significantly reduced energy intake compared to placebo, although the combined treatment had superior effects compared to PYY3-36 or OXM monotherapy. Human studies have also clearly demonstrated the direct effect of OXM on energy expenditure (140); this effect was later confirmed by indirect calorimetry (141). These modest but favorable results suggest significant promise for OXM-based therapies for obesity. In addition to the established action of OXM on appetite, another mechanism that potentially plays a role in energy intake and glucose metabolism is gastric emptying. Intravenous infusion of OXM reduced gastric emptying in humans (142). Whether reduction in gastric emptying is involved in the acute and long-term metabolic effects of OXM is not yet clear. Nevertheless, the immediate future will reveal OXM’s role in obesity management. However, as for other peptide hormones, their clinical application is limited by their short circulatory half-life, a major component of which is cleavage by DPP-IV. Therefore, structurally modified analogues with an altered OXM pharmacological profile have been produced with longer duration of action, good safety profile, and positive effects on body weight (and glucose metabolism) management in animal studies (143). These findings bring closer their usage in human clinical trials. Furthermore, the crystal structure of OXM has been determined, and this advance should facilitate the rational design of oxyntomodulin peptidomimetics to be tested as oral anti-obesity pharmaceuticals. Even so, despite the promising weight reduction efficacy of OXM, only a small number of development projects appears to be at an advanced stage. TKS1225 is an OXM analogue. The present status of this molecule is unknown. OXY-RPEG has been engineered via its proprietary reversible pegylation technology to increase its half-life and increased potency. In preclinical testing, OXY-RPEG was significantly superior to twice daily injections of OXM in the reduction of food intake and the degree and durability of weight loss. In 2009, an oxyntomodulin-based multi-agonist peptide with glucagon and GLP-1 agonistic actions were created. This multi-agonist had advanced action comparing to the one that Day et al had introduced at the same year (144). A 2-weeks trial in DIO mice showed weight loss and glucose control improvement. This beneficial action was obvious even in mice without GLP1 or glucagon receptor confirming the superiority of this analogue. Oxyntomodulin functions endogenously as a physiologic co-agonist, but regarding its small bioactivity, it is mainly characterized by its function as biosynthetic precursor to glucagon.

 

GLUCAGON-LIKE PEPTIDE 1/AMYLIN

 

In 2010, salmon calcitonin-exendin-4 combined therapy achieved reduction of food intake and weight in non-human primates (145). Of note, the human amylin receptor subtypes consist of calcitonin receptor and receptor activity-modifying proteins. This observation was the first step in the development of multi-agonist molecules targeting GLP-1 and Amylin (146). Two of these peptide hybrids (phybrids) had a C-terminally truncated Exenatide, which was covalently linked to the N-terminus of an amylin analogue (Davalintide) through either a repeat β-ala-β-ala dipeptide, or through triple-glycine linear repeat. Administration of phybrids resulted in greater weight loss in non-human primates than monotherapy, although similar to that achieved by a physical commixture of the single hormones. Another GLP1/Amylin phybrid was introduced, which used a full-length Exenatide sequence linked to Davalintide viaan intervening 40-kDa PEG. This phybrid reduced both blood glucose and body weight in a dose-dependent fashion.    

 

GLUCAGON-LIKE PEPTIDE 1/GLUCOSE-DEPENDENT INSULINOTROPIC POLYPEPTIDE

 

This single-molecule multi-agonist was quite controversial. Glucose-Dependent Insulinotropic Polypeptide (GIP), is a 42-amino acid peptide, produced by K-cells in the duodenum and jejunum and released into the general circulation upon stimulation by dietary lipids (147). The investigation following the discovery of this new peptide, showed that GIP is the first incretin hormone. It acts directly on the pancreas augmenting glucose-stimulated insulin secretion (148). It is worth mentioning that GIP has the ability to enhance both insulin secretion in hyperglycemia and glucagon release in hypoglycemia (149). A few years later, the role of GIP in obesity development became apparent. GIP acts on adipocytes enhancing adipogenesis, inhibition of lipolysis, stimulation of de novo lipogenesis (150) and on chylomicrons stimulating triglyceride release. It also affects adipocyte glucose and fatty acid uptake and adipocyte lipoprotein lipase enzyme activity (151). It is remarkable that although GIP was regarded as an obesogenic hormone, mice overexpressing GIP showed improved β-cell function and improved glycemic control and were resistant to DIO (152). Additionally, in studies with mice, it was shown that the chronic GIP agonist administration improves glucose metabolism without body weight changes (153). In 2013, two single-molecule multi-agonists GLP1/GIP were introduced, whose action was based on the insulinotropic action of both components (153). GIP agonist enhanced GLP1 action upon glucose metabolism and GLP1 could mitigate obesogenic effect of GIP via its anorectic effect. The biochemical structure of multi-peptide was similar to GLP1/glucagon multi-agonist i.e. a single peptide with potency at both receptors (GIP residues were introduced in the median and the C-terminal part of peptide; certain modifications that increased activity on the glucagon receptor were removed; the C-terminus of the peptide ended with the nine amino acid extension found in exendin-4 and an Aib was added at position 2 to protect against DPP-IV inactivation) (154). Several clinical trials in mice, rodent models, non-human primates and humans were performed, concluding that the GLP1/GIP multi-agonist therapy reduced food intake, and consequently body weight, improved glycemic control, lipid profile and lipolysis but without any improvement in energy expenditure.

 

GLUCAGON LIKE PEPTIDE 1/GLUCAGON/GLUCOSE-DEPENDENT INSULINOTROPIC POLYPEPTIDE

 

The creation of this single-molecule multi-agonist was based on the biochemical structure of GLP1/GIP and GLP1/glucagon multi-peptides. An Aib at position 2 both protected the molecule from DPP-IV inactivation and decreased its potency at the glucagon receptor; an amino acid lysine at position 10 was fatty-acylated via a γ-glutamic acid linker to palmitic acid; amino acids at positions 16,17, 20, 27, 28 replaced balanced glucagon bioactivity; a C-terminal exendin-4 extension sequence (CEX) succeeds agonism at all three receptors 10-fold greater than native hormones (155). The main mechanism of action is based on the combination of the anorectic effect of GLP-1, the lipolytic and thermogenic characteristics of glucagon, and the action of GIP on β-cell function and glycemic control. Contrary to GLP-1/GIP multi-agonist, which doesn’t affect energy expenditure, this triple agonist increases energy expenditure. (see Table 18)

 

Table 18. Single Molecule Multi-Agonists

Drug name

Clinical benefits

Adverse events

Glucagon-like peptide 1/glucagon

oxyntomodulin, MED10382, G530S (Glucagon analogue/Semaglutide), GC-co-agonist 1177

↓ food intake, obesity, hepatic steatosis, HbA1c, lipid profile

↑energy expenditure

 

Glucagon-like peptide1/amylin co-agonism

↓ blood glucose and body weight dose-dependently

 

Glucagon-like peptide 1/glucose-dependent insulinotropic polypeptide

↓ blood glucose, lipid profile, food intake, body weight,

↑ lipolysis

No improvement in energy expenditure

Glucagon-like peptide 1/glucagon/glucose-dependent insulinotropic polypeptide

↓body weight, HbA1c, hepatosteatosis, cholesterol, ↑energy expenditure, lipolysis

 

 

Peptide-Mediated Delivery of Nuclear Hormones

 

The use of nuclear hormones as an agent of GLP-1 and Glucagon is a novel promising therapy in the treatment of obesity. Nuclear hormones are characterized by high potency and pleiotropic action as well as unwanted adverse effects. The basic idea involves a linkage of a nuclear hormone to a peptide, usually through a linker that would allow metabolism of the nuclear hormone only within the targeted cell reducing the undesirable effects in other tissues. However, in the cell types that possess the specific peptide receptor, its activation should lead to internalization of the ligand-nuclear hormone receptor complex. In this case, the peptide receptor plays the role of a gateway into the cell. Upon internalization, biological processing of a suitably designed linker would release the nuclear hormone and allow activation of its intracellular receptor. Although a promising option, not all nuclear hormones can be used as peptide-mediated agents. They should have high tissue selectivity, ability to be internalized and compatibility to peptide wanted. Estrogens, tri-iodothyronine, and dexamethasone are the nuclear hormones that have been tested.

 

GLUCAGON-LIKE PEPTIDE 1-MEDIATED DELIVERY OF ESTROGEN

 

Glucagon-Like Peptide 1-mediated delivery of estrogen was first introduced in 2012. The use of estrogens was indicated by the fact that estrogen replacement therapy in postmenopausal women improved multiple cardio metabolic parameters (156). Furthermore, estrogens have anabolic, insulinotropic, and anorectic effects (157). The combination of estrogen and GLP-1 was found to improve body weight and glycemic control in rodent models with the metabolic syndrome (158). The weight-lowering effect was due to appetite suppression, while the GLP-1/E2 combination showed greater potency comparing to GLP-1 analog or E2 alone. Further clinical trials enhanced this finding showing an influence on feeding behavior. Additive contribution of GLP-1/E2 on pancreatic islet function, cytoarchitecture and protection from deleterious insults such as lipotoxicity was found in 2015 (159). Despite the powerful metabolic benefits associated with estrogen action, effects on the reproductive endocrine system and oncogenic potential have restricted their clinical use in postmenopausal women. Furthermore, many aspects of molecular pharmacology and mechanism of action remain unresolved. Specifically, neither the precise intracellular processing of the GLP-1/E2 conjugate, which results in active estrogen cargo release, nor the molecular identity that delivers estrogen activity, have been determined. It is possible that estrogens enhance brain penetration and alter the bio-distribution of the conjugate to more privileged sites for central nervous action.

 

GLUCAGON-MEDIATED DELIVERY OF TRI-IODOTHYRONINE

 

Glucagon and thyroid hormone can separately lower body weight and LDL cholesterol in humans. Thyroid hormones act both on liver, regulating hepatic lipid metabolism and hepatosteatosis, and in adipose cells, increasing energy expenditure and enhancing lipolysis (160, 161). On the other hand, they can cause cardiac hypertrophy, tachycardia, muscle catabolism, and bone deterioration. Glucagon receptors are highly concentrated not only in the liver, which is the preferred site for T3 action, but also in adipose tissues, kidney, and the cardiovascular system resulting in metabolic enhancement along with toxicity risk. Considering all of the above, a glucagon/T3 conjugate was created. A native T3 combined with a DPP4-protected C-terminally extended glucagon analog via a peptide spacer (162). Several control compounds were also generated to permit appropriate pharmacological comparisons. These additional peptides included a conjugate with selective chemical substitution in the peptide to suppress glucagon activity, a compound with a linker that proved metabolically stable and was incapable of intracellular T3 release, and a third control conjugate that carried a metabolically-inert thyroid hormone. Finan found that the conjugate glucagon/T3 corrected lipid metabolism in rodent models with dietary-induced metabolic syndrome. The above findings showed that the body-weight effect of the conjugate can partially be governed by actions in adipose depots because glucagon receptors exist in rodent adipocytes, less than in liver. Moreover, the glucagon/T3 conjugate effect is supported by the uncoupling protein 1-mediated thermogenesis, enhanced FGF21 secretion and biased by PGC-1 cofactor signaling. Interestingly, the combination of glucagon/T3 seems to decrease arterial plaque area in LDL receptor -/- mice and fibrosis in mice with advanced fatty liver disease.  Although the above data demonstrate the cardiovascular benefit of this conjugate, further chemical improvements should be made in order to be safe for chronic use in higher mammals and especially humans.

 

GLUCAGON-LIKE PEPTIDE 1-MEDIATED DELIVERY OF DEXAMETHASONE

 

It is widely known that dietary-induced obesity causes chronic peripheral and central inflammation (163). Glucocorticoids are widely known for their anti-inflammatory characteristics, but due to their ubiquitous action profile, their therapeutic use can lead to off-target effects. In 2017, DiMarchi and Tschop created a GLP1/dexamethasone conjugate which managed to improve body weight in DIO mice, in a superior way to GLP1 or dexamethasone alone.  This combination improved hypothalamic inflammation, astrocytosis, microgliosis, and insulin sensitivity. The targeted delivery of dexamethasone to GLP1R-positive cells prevented typical dexamethasone off–target effects on glucose metabolism, bone density, and the hypothalamus-pituitary-adrenal axis activity (164). (see Table 19)

 

Table 19. Peptide-Mediated Delivery of Nuclear Hormones

Drug name

Clinical benefits

Glucagon-like peptide 1/estrogen

↓ food intake, body weight, HbA1c

Glucagon/tri-iodothyronine

↓ lipid profile, arterial plaque and fibrosis in advanced fatty liver disease

Glucagon-like peptide 1/dexamethasone

↓ hypothalamic inflammation, astrocytosis, microgliosis, ↑insulin sensitivity

 

Peptide Y (PYY)

 

PYY is a 36-amino acid anorexigenic peptide with a hairpin-like U-shaped fold secreted from the entero-endocrine L-cells of the ileum and colon in response to feeding. PYY presents in two major forms, PYY1-36 and PYY3-36. More specifically, PYY is a member of the pancreatic polypeptide-fold (PP-fold) family which also includes NPY and PP and interacts with a family of receptors (mainly Y2R). It is produced postprandially, in response and proportionally to caloric load, by the distal-intestinal L cells along with oxyntomodulin (OXM) and GLP-1. Just like GLP-1 and OXM, PYY1-36 is rapidly proteolyzed by DPP4. However, unlike the other two neuropeptides, the cleaved product PYY3-36, is bioactive. Human studies have shown that PYY delays gastric emptying and promotes satiety (165), while short-term intravenous administration of PYY3-36 , at doses generating physiologic postprandial blood excursions, was shown to decrease calorie intake by approximately 30% in lean and obese subjects, without causing nausea, affecting food palatability, or altering fluid intake, nor was it followed by compensatory hyperphagia (166). Another study confirmed the above findings, reporting dose-dependent reductions of food intake (maximal inhibition, 35%; P<0.001 vs control) and calorie intake (32%; P<0.001) after intravenous infusions of several different concentrations of PYY3-36 (167). Sloth et al. first showed the significantly higher energy expenditure following PYY3-36 intravenous infusion compared with PYY1-36 or control. In a recent study, the effect of infused PYY3-36 on energy intake was compared to that of OXM or the combined PYY3-36/OXM treatment; the results demonstrated that energy intake was significantly less with the combined treatment compared to PYY3-36 or OXM monotherapy (168). Whether these findings pointed to a weight loss efficacy of PYY was evaluated in a 12-week trial of 133 obese patients who were randomly assigned to intranasal PYY3-36 (200 or 600 mcg three times a day before meals) or placebo, in conjunction with diet and exercise. At the 200 mcg dose, PYY3-36 failed to reduce body weight, while 60% of patients treated with the high PYY3-36 dose (600 mcg three times a day) dropped out due to nausea and vomiting, so that no meaningful inference could be drawn from the few patients who completed the study on 600 mcg. These findings contrast with those in rodents (169, 170) and nonhuman primates (171) where PYY3-36 preparations reduce body weight. One suggested explanation is that the PYY3-36 effect is critically modulated by the time of injection. As the main anorexigenic effect of PYY is by Y2R-mediated NPY inhibition, PYY is obviously more effective at times that the orexigenic NPY is increased. In accordance with this theory is the reported weight loss effect of PYY3-36 when injected in rodents in the fasting state or in the early dark cycle — times when NPY is naturally induced (172).

 

PYY3-36 is structurally similar to pancreatic polypeptide (PP); PYY3-36 acts mainly through Y2R, while PP acts through Y4R. Obinepitide (TM30338), a synthetic dual-analogue of PYY3-36 and PP that stimulates both Y2/Y4-receptors, has been developed. Pre-clinical studies have shown that obinepitide efficiently reduces weight in obese mice. Furthermore, initial studies in humans have shown that once-a-day subcutaneous administration of obinepitide in obese human subjects inhibited food intake, at a statistically significant level, up to at least nine hours after dosing (173). Various PYY analogues have been created including intravenous, oral or nasal formulations. Interestingly, the combined therapy of PYY3-36 and GLP-1 receptor agonist (exendin-4) was found to decrease food intake and body weight in an additive manner in animal models and humans. Specifically, this synergistic result was attributed to the enhancement of c-fos reactivity in special cerebral nuclei (174). (see Table 20)

 

Table 20. PYY

Mechanism of action

Anorexigenic peptide which decreases gastric motility, increases satiety, inhibits NPY receptors

Clinical Benefits

↓ appetite, decreases food intake, ghrelin levels

Adverse events

Short-time action

 

Ghrelin Vaccines and Ghrelin Inhibitors

 

Ghrelin is a 28-amino acid peptide produced primarily by the stomach and proximal small intestine (175). It is the only known circulating orexigenic hormone and signals both on vagal afferents and in the arcuate nucleus where it powerfully enhances NPY orexigenic signaling (176, 177). Its levels increase before meals and are suppressed by ingested nutrients, with carbohydrates being the most effective ones (compared to proteins and lipids). Ghrelin’s suppression results from neutrally transmitted (non-vagal) intestinal signals, augmented by insulin. An experimental ghrelin vaccine, CYT009-GhrQb, was discontinued in 2006 as it did not have the expected effects on weight loss. A novel one conjugated to the hapten, keyhole limpet hemocyanin (KLH), tested in rodent models, was shown to decrease feeding and induce weight loss (178). NOX-B11 is a ghrelin-neutralizing RNA spiegelmer that attaches to the active form of ghrelin and blocks its ability to bind to its receptor thus blocking the orexigenic activity of exogenously administrated ghrelin in rats (179). However, NOX-B11 did not affect basal food intake in nonfood-deprived rats, thus this treatment may only be efficacious when plasma ghrelin levels are high, such as before a meal or during times of food restriction (dieting).Since the discovery that the effects of ghrelin are primarily mediated by the GH secretagogue receptor (GHSR) 1a, there have been multiple potent, selective, and orally bioavailable ghrelin antagonists produced with good pharmacokinetic (PK) profiles that are currently in preclinical testing. An amide derivative 13d (Ca2+ flux IC50 = 188 nM, [brain]/[plasma] = 0.97 @ 8 h in rat), for example, showed a 10% decrease in 24-hour food intake in rats, and over 5% body weight reduction after 14-day oral treatment in diet-induced obese (DIO) mice (180).

 

Moreover, the discovery of ghrelin O-acyltransferase (GOAT) as the enzyme that catalyzes ghrelin octanoylation, revealed several therapeutic possibilities including the design of drugs that inhibit GOAT and block the attachment of the octanoyl group to the ghrelin third serine residue; such GOAT inhibitors could potentially prevent or treat obesity (181). Octanolyation of ghrelin by GOAT on its third amino acid (serine-3) is necessary for the hormone’s biological functions. Octanoylated ghrelin enhances hyperphagia and increases gastrointestinal motility. Furthermore, it reduces insulin secretion causing glucose dysfunction, enhances thermogenesis, adipogenesis and liver lipogenesis, limiting lipolysis at the same time (182). So, inhibiting GOAT could impede the production of acyl-ghrelin and increase desacyl-ghrelin, thus improving glucose homeostasis. In 2010, GO-CoA-Tat was created. A peptide-based bi-substrate analog which inhibited GOAT activity. The chronic treatment with GO-CoA-Tat, resulted in body weight stabilization in vehicle-treated mice fed MCT-rich HFD. Additionally, a decrease of fat mass was shown, but not of lean mass (183). Another study on Siberian hamsters also resulted in improvement in ingestive behavior. Remarkably, after 48h food deprivation, GO-CoA-Tat attenuated food foraging, food intake, and food hoarding post-refeeding relative to animals treated with saline. GO-CoA-Tat treated mice improved their blood glucose (184).

 

Another promising anti-obesity agent against ghrelin is a brain penetrant CAMKK2 inhibitor. Generally, CAMKK2 has been identified as the hypothalamic AMPK kinase that transduces Ca2+-mediated ghrelin signaling, inhibiting selectively hypothalamic AMPK and NPY’s downstream orexigenic effect. 4t, a 2,4-diaryl 7-azaindole, was created in order to inhibit AMPK phosphorylation in a hypothalamus-derived cell line. When this agent was tested in rodents, it managed to reduce ghrelin-induced food intake (185) (see Table 21).

 

Table 21. Ghrelin Vaccine (NOX-B11)

Mechanism of action

Ghrelin vaccine

Clinical Benefits

↓ food intake, hypothalamic orexigenic signals, ↑energy expenditure

Adverse events

No weight loss seen in human trials

 

Fat-Specific Satiation Peptides

 

ENTEROSTATIN AND APOLIPOPROTEIN A-IV

 

Enterostatin and apolipoprotein A-IV appear to be GI peptides that are specifically stimulated by fat ingestion and subsequently regulate intake and/or metabolism of lipids. Although peripheral and central enterostatin administration decreases dietary fat intake in animals (while enterostatin-receptor antagonists did the opposite) (186), its administration to humans has shown no effects on food intake, appetite, energy expenditure, or body weight (187). Similarly, apolipoprotein A-IV, which is synthesized and secreted exclusively by the small intestine (primarily by the jejunum, but also by the duodenum and ileum), acts as a satiety factor that is downregulated by leptin (188) and upregulated by insulin and PYY in both rodents and humans (189). Although exogenous administration of apolipoprotein A-IV was quite effective concerning meal size, food intake, and weight gain reduction in rats (190), data is lacking regarding apo A-IV therapeutic administration in humans and its effects on body weight.

 

Pancreatic Satiation Peptides

 

PANCREATIC POLYPEPTIDE (PP)

 

Pancreatic polypeptide (PP) is a 36-amino acid peptide that is structurally similar to PYY. It is primarily produced in the pancreas in response to ingestion of food and in proportion to caloric load (191). Animal studies have shown that peripheral administration of PP decreases feeding (through Y4R in the area postrema), whereas centrally administrated PP increases it (through Y5R deeper in the brain) (192). In humans, intravenous infusion of PP (10 pmol/kg/min) (supra-physiological levels of PP) in ten healthy volunteers (men and women of normal body weight) caused a sustained decrease in both appetite and cumulative 24-hour energy intake by 25.3 +/- 5.8% (193). The findings of another study studying the anorexigenic effect of a lower infusion rate of PP (5 pmol/kg/min) in lean fasted volunteers were similar, holding promise for potential use as an anti-obesity agent (194). Another trial studying whether combined treatment with PP/PYY3-36 is superior regarding weight loss compared to either agent alone concluded that PP and PYY3-36 do not inhibit feeding additively in humans (195). Again, this study was conducted on lean subjects. Conversely, as previously mentioned, a synthetic analogue (TM30338) of both PYY3-36 and PP, which acts as an agonist of both the Y2 and Y4 receptors, yielded very promising results as concerns early meal termination when administered once-a-day subcutaneously in obese human subjects. Similarly, initial reports of a selective Y4-receptor agonist (TM30339) currently under development were also quite promising inducing reduction of food intake and promoting weight loss.

 

AMYLIN AND AMYLIN ANALOGUES

 

Amylin is a 37-amino acid neuroendocrine peptide hormone co-secreted postprandially with insulin by pancreatic β-cells. Among other properties, amylin is characterized by centrally mediated glucoregulatory and anorexigenic actions (196). It inhibits gastric emptying and glucagon secretion as well as decreases meal size and calorie intake (fat specific) (197) in a dose-dependent manner. These are vagus-independent actions and are exerted via binding to specific amylin receptors in the hindbrain area postrema (198), which is in contrast with the peripheral neural mechanisms engaged by most other gut peptides involved in energy homeostasis system regulation. The anorectic efficacy of amylin along with its glucoregulatory actions were investigated in human studies with the usage of pramlintide, a subcutaneous injectable amylin analogue which differs from amylin by only three amino acids. Studies in patients with type 1 and type 2 diabetes have shown great improvement in glycemic control plus sustained reductions in food intake and meal size, as well as mild progressive weight loss, following acute and long-term adjunctive pramlintide treatment (120 μg) (199). The most common adverse event associated with pramlintide usage was transient, mild-to-moderate nausea. This weight loss is noteworthy because it occurred in subjects with type 2 diabetes, on concomitant insulin therapy, and in the face of a significant A1C reduction, factors that all favor weight gain. Similar to the GLP-1 analogues discussed previously, pramlintide is currently approved for the treatment of type 1 and type 2 diabetes.

 

Whether pramlintide could constitute a potent anti-obesity agent was investigated in well-designed trials addressing this issue. In such a study (16-week randomized, double-blind, placebo-controlled), 204 individuals with obesity but not diabetes were treated with self-administered subcutaneous injections of pramlintide (nonforced dose escalation ≤ 240 μg) or placebo three times a day, 15 minutes before meals without concomitant lifestyle intervention (200). Pramlintide was generally well-tolerated and approximately 90% of the pramlintide-treated subjects were able to escalate to the highest dose of 240 μg three times a day. In contrast to the placebo-treated subjects who experienced minimal changes in body weight over the 16-week treatment period, the pramlintide-treated subjects attained significant weight loss from baseline as early as week 2, which was progressive up to week 16, with no evidence of a plateau. At week 16, the placebo-corrected reduction in body weight after pramlintide treatment was statistically significant compared with placebo (3.7 ± 0.5%, P < 0.001; 3.6 ± 0.6 kg, P < 0.001). Furthermore, the reduction in weight in pramlintide-treated subjects was accompanied by a significant reduction in waist circumference compared with placebo-treated subjects after 16 weeks of treatment (evaluable 4.3 ± 0.6 vs. 0.7 ± 0.9 cm, P < 0.01). At the end of the 16-week trial, 31% of the subjects treated with pramlintide achieved ≥ 5% weight loss compared to just 2% of the placebo group (P < 0.001). Interestingly, 8 weeks after treatment cessation, the pramlintide-treated subjects had on average regained one third of the overall weight loss observed by week 16. These findings constitute a proof of concept that pramlintide may have therapeutic use as an anti-obesity agent. Remarkably, at this higher dose (240 μg three times a day), the mean reduction in body weight with pramlintide treatment over 16 weeks was approximately twice that previously observed over a similar time-frame in insulin-treated subjects with type 2 diabetes who were treated with lower pramlintide doses (120 μg). This could suggest that higher doses of pramlintide might be necessary to achieve significant weight loss, although it is not yet clear whether concurrent insulin treatment was the main cause of that difference.

 

AMYLIN/PRAMLINTIDE COMBINATIONS

 

Previous animal studies have shown that amylin treatment significantly enhanced hypothalamic anorexigenic leptin signaling, while the combination treatment with amylin and leptin led to marked, synergistic reductions in food intake (up to 45%) and fat-specific weight loss (up to 15%). Recently, the weight-lowering effect of combined amylin/leptin agonism in human obesity was evaluated using the analogues pramlintide/metreleptin, respectively. As previously discussed, (see leptin), three trials addressing the weight loss efficacy of the combined treatment over 20, 28, and 52 weeks, respectively) reported sustained and robust weight loss by the combined treatment. Development was discontinued following commercial reassessment of the program. A Phase II study of davalintide, a second-generation analogue of amylin, for the treatment of obesity has also completed. In this study however, the weight loss efficacy and tolerability profile of davalintide was not superior to pramlintide, and was inferior to the pramlintide/metreleptin combination, thus resulting in deciding to halt further development of davalintide.

 

The anti-obesity effect of the combined treatment amylin/PYY3-36 was evaluated in an animal study, given that they both may have the potential for short-term signals of meal termination with anorexigenic and weight-reducing effects (201, 202). Statistical analyses revealed that food intake suppression with the combined treatment was synergistic, whereas body weight reduction was additive; this combination has not yet been studied in humans.  Additional preclinical studies looking at the safety and efficacy of the combined treatment with pramlintide/phentermine and pramlintide/sibutramine was evaluated in a randomized placebo-controlled study with 244 obese or overweight nondiabetic subjects (203). The results suggested that the weight loss achieved at week 24 with either combination treatment was greater than with pramlintide alone or placebo (P < 0.001; 11.1 +/- 1.1% with pramlintide + sibutramine, 11.3 +/- 0.9% with pramlintide + phentermine, -3.7 +/- 0.7% with pramlintide; -2.2 +/- 0.7% with placebo; mean +/- s.e.), without any major adverse events.

 

As mentioned above, the human amylin receptor subtypes consist of calcitonin receptor and receptor activity-modifying proteins. Because of their mechanism of action, amylin mimetics coupled with calcitonin receptor agonists, are known as dual action amylin and calcitonin receptor agonists (DACRA). DACRA KBP-088 showed greater efficacy relative to davalintide regarding in vitro receptor pharmacology and in vivo efficacy of food intake and body weight (204). DACRA KBP-088 and KBP-042 improved body weight, glycemic control and adipose hypertrophy in high-fat diet-fed rats (205). A long acting amylin analogue is also in phase I clinical trial as a once daily anti-obesity treatment (206). (Table 22)

 

Table 22. Amylin/Pramlintide Combinations

Drug name

FDA approved/Phase

Mechanism of action

Clinical Benefits

Adverse events

pramlintide

Approved for DM1, DM2

Amylin analogue

-in DM1, DM2: ↓ blood glucose, food intake, body weight, waist circumference

Nausea

Davalintide (AC2307)

Phase II

Amylin analogue

↓food intake, body weight, HbA1c

hypoglycemia

DACRA KBP-088, KBP-042

 

Dual amylin and calcitonin receptor agonist

↓body weight, glycemic control, adipose hypertrophy

 

 

PERIPHERAL MODULATORS OF THE EFFICIENCY OF DIGESTION, METABOLISM, AND LIPOGENESIS

 

Lipase Inhibitors

 

Apart from early termination of food intake augmented by the centrally acting appetite suppressants, another potential therapeutic anti-obesity approach is the induction of a negative energy balance through the inhibition of nutrient, particularly fat, absorption. Lipase inhibitors inhibit gastric and pancreatic lipases in the lumen of the gastrointestinal tract that decrease systemic absorption of dietary fat. Orlistat is currently the only marketed anti-obesity drug of this category licensed for the treatment of obesity (including weight loss and weight maintenance). Additionally, it has been proven to improve glucose metabolism and nonalcoholic fatty liver disease. The most common adverse events are gastrointestinal system and include oily spotting, flatus with discharge, diarrhea, fecal urgency, and vitamin malabsorption (207).

 

The only other pancreatic and gastrointestinal lipase inhibitor currently in clinical development is Cetilistat (ATL-962). A short-term (12-week) randomized, placebo-controlled study of weight reduction addressing the efficacy, safety, and tolerability of Cetilistat in obese patients reported that Cetilistat produced a clinically and statistically significant weight loss in obese patients to similar extents at all doses examined compared to placebo (60 mg t.i.d. 3.3 kg, P<0.03; 120 mg t.i.d. 3.5 kg, P=0.02; 240 mg t.i.d. 4.1 kg, P<0.001), plus it significantly improved other obesity-related parameters including waist circumference, serum cholesterol and low-density lipoprotein cholesterol levels. Cetilistat treatment was also well-tolerated and the common orlistat-induced GI adverse events, such as flatus with discharge and oily spotting, occurred in only 1.8-2.8% of subjects in the Cetilistat-treated group (208). The combined results from three Phase I clinical studies designed to investigate the efficacy, pharmacodynamics, and tolerability of a range of Cetilistat doses [50 mg t.i.d. (n = 7), 60 mg t.i.d. (n = 9), 100 mg t.i.d. (n = 7), 120 mg t.i.d. (n = 9), 150 mg t.i.d. (n = 16), 240 mg t.i.d. (n = 9) and 300 mg t.i.d. (n = 9)] compared with placebo or orlistat [120 mg t.i.d. (n = 9)] in healthy volunteers were published (209). They reported that Cetilistat is equipotent with orlistat regarding fecal fat excretion; it however achieves a much better tolerance profile, as the number of episodes of steatorrhea per subject in the orlistat group (4.11) was 2.5-fold greater than that in the Cetilistat-treated group. The different tolerance profile between the two lipase inhibitors, seems to be related to the physical form of the fat in the intestine (rather than the amount of fat) resulting from each medication. Thus, Cetilistat acts more like a detergent, whereas orlistat may promote the coalescence of micelles, leading to oil-drops and increased gastrointestinal adverse events. Finally, a 12-week trial compared the efficacy and safety of Cetilistat (40, 80 or 120 mg three times daily) and orlistat (120 mg t.i.d.) relative to placebo in obese patients with type 2 diabetes on metformin (210). In this study similar reductions in body weight were observed in patients receiving Cetilistat (80 or 120 mg t.i.d.) or orlistat; these reductions were significant compared to placebo (3.85 kg, P = 0.01; 4.32 kg, P = 0.0002; 3.78 kg, P = 0.008). Furthermore, treatment with Cetilistat (80 or 120 mg t.i.d.) or with orlistat significantly improved glycemic control relative to placebo; again, Cetilistat was well-tolerated and showed fewer discontinuations due to adverse events than in the placebo and orlistat groups. Based on the above findings, this novel lipase inhibitor is currently at the furthest stage in the clinical development of new drugs of this class (see Table 23).

 

Table 23. Lipase Inhibitors

Drug name

FDA approved

Mechanism of action

Weight loss vs placebo

Clinical Benefits

Adverse events

Orlistat (Xenical)

1999

Lipase inhibitor

2.6%

↓ HbA1c, nonalcoholic fatty liver disease

Gastrointestinal side effects, vitamin malabsorption

Contraindicated in:Chronic malabsorption syndrome, cholestasis

Cetilistat (ATL-962)

 

Pancreatic and gastric lipase inhibitor

 

↓body weight, lipid profile, waist circumference

Gastrointestinal (less than orlistat)

 

Growth Hormone (GH) and GH Lipolytic Domain Synthetic Analogues

 

Besides its growth effects, GH also possesses significant metabolic properties, including lipolysis induction. On the other hand, GH dynamics change with increasing adiposity and GH circulating levels and response to stimuli are repressed in obesity (211, 212). Taken together, it could be hypothesized that GH administration is an effective therapeutic option for weight loss and fat mass reduction in obese individuals. However, the majority of the 16 clinical trials of GH administration in obesity indicated little or no beneficial effects of GH treatment on body weight (213). There is a report from an Australia-based biotechnology company of the development of a modified fragment of amino acids 177-191 of GH (hGH177-191) (AOD-9604) that mimics the lipolytic effects of GH without producing growth effects. AOD-9604 however failed to induce significant weight loss in a 24-week trial of 536 subjects and its development as an anti-obesity agent was terminated (214). In 2018, it was announced that GH not only promotes lipolysis, but also enhances the creation of beige adipose tissue through activation of STAT5 and induction of ADRB3. Consequently, it promotes the adrenergic action of WAT.

 

β3-Adrenoreceptor Agonists

 

The β3-adrenergic receptor is expressed in adipocytes; its activation by cognate β-agonists cause lipolysis and increase thermogenesis. Thyroid hormones increase thermogenesis via the thyroid hormone receptor β subtype; however, to date, every attempt to develop selective thyroid hormone receptor agonists which are effective in adipose tissue without systemic side-effects has failed. In 2000, a selective human β3-agonist, L-796568, was developed (215). Although its acute (4-hour period) administration in overweight human subjects was associated with significant increase in energy expenditure (by ~8%) (216), a 28-day clinical trial investigating the efficacy of chronic use of L-796568 in overweight and obese non-diabetic men receiving the drug (350 mg/d) failed to display any significant changes in body composition or 24-hour energy expenditure (217). The ineffectiveness of β3-adrenreceptor activation to induce significant and sustained lipolysis in humans may be explained by the fact that human WAT expresses minimal levels of β3-adrenoreceptors; similarly, their expression is also low within human brown adipose tissue.

 

11β-Hydroxysteroid Dehydrogenase Type 1 Inhibitors

 

Previous studies have shown enhanced conversion of inactive cortisone to active cortisol through the expression of 11β-hydroxysteroid dehydrogenase type 1 (11βHSD1) in cultured omental adipose stromal cells (218); the autocrine action of cortisol may be crucial in the pathogenesis of central obesity and features of the metabolic syndrome, such as insulin resistance. The reports relating to effectiveness of carbenoxolone (nonselective 11β-HSD inhibitor) in reducing central obesity are conflicting (219). Currently, several pharmaceutical companies are developing selective 11β-HSD1 inhibitors that are effective in adipose tissue and may be more effective in improving insulin sensitivity and reducing body weight. Preliminary data from animal studies evaluating the weight-loss benefit of T-BVT, a new 11β-HSD1 pharmacological inhibitor with specificity for WAT, are very promising regarding its anti-obesity effectiveness and amelioration of multiple metabolic syndrome parameters (220). CNX-010-49, is another selective tissue-acting 11β-HSD1 inhibitor under investigation. Animal studies showed that this inhibitor acts on glucocorticoids and isoproterenol resulting in lipolysis in mature 3T3-L1 adipocytes. It not only enhances muscle glucose oxidation and mitochondrial biogenesis, but also reduces proteolysis and gluconeogenesis in primary mouse hepatocytes. As a result, it improves glucose control, lipid metabolism, and inhibits body weight gain without affecting feed consumption. A potential cardiovascular benefit was found because of the action of CNX-010-49 on plasminogen activator inhibitor-1 (PAI-1), interleukin-6 (IL-6), and fetuin-A (221). (see Table 24)

 

Table 24. 11β-Hydroxysteroid Dehydrogenase Type 1 Inhibitors

Drug name

Mechanism of action

Clinical Benefits

T-BVT

Selective to white adipose tissue 11β-HSD1

 

CNX-010-49

Selective to white adipose tissue 11β-HSD1

↑lipolysis,

↓ HbA1c, lipid metabolism, inhibits body weight gain without affecting feed consumption

 

Angiogenesis Inhibitors

 

Increasing adiposity is associated with expansion of the adipose capillary bed. Several vascular growth factors are produced by enlarged adipocytes, for example, vascular endothelial growth factor (VEGF), hepatocyte growth factor (HGF), and angiogenin, which may in turn facilitate the expansion of adipose tissue. Thus, anti-angiogenesis may eventually participate in the treatment of obesity. This hypothesis is strengthened by studies where the experimental administration of anti-angiogenic agents in mice from different obesity models resulted in significant weight reduction and adipose tissue loss (222). Remarkably, there were benefits on food intake, metabolic rate, and preferred energy substrate. These findings appeared to modulate fat tissue by altering vasculature. Although there are many foods and beverages containing naturally occurring inhibitors of angiogenesis (e.g. green tea, oranges, strawberries, lemons, red wine, ginseng, garlic, tomato, olive oil, etc.), no convincing clinical trials have been conducted investigating their anti-obesity effect so far. Currently, a Phase II trial using the anti-angiogenic/anti-MMP drug ALS-L1023 for the treatment of obesity is underway (223). Similarly, endostatin was found to have both anti-adipogenic and anti-angiogenic action protecting mice against dietary-induced obesity (224).

 

Sirtuin 1 (SIRT1) Activators

 

Sirtuin 1 (SIRT1) is a member of the Sirtuin family of proteins that comprises seven members in mammals (SirT1-T7). Sirtuin proteins have gained considerable attention due to their importance as physiological targets for treating diseases associated with aging. They contribute to cellular regulation interacting with metabolic pathways and may serve as entry points for drugs. SIRT1 has gained popularity as it has been linked with the French Paradox and the calorie restriction-mediated longevity and delayed incidence of several diseases associated with aging, such as cancer, atherosclerosis, and diabetes. The calorie restriction-induced modulations have been demonstrated in organisms ranging from yeast to mammals. White adipose tissue seems to be a primary factor in the longevity brought about through calorie restriction, as mice engineered to have reduced levels of WAT live longer (224). Corroborating this, it was found that food withdrawal is followed by SIRT1 binding and repression of genes controlled by the fat regulator PPAR-γ (peroxisome proliferator-activated receptor-γ), including genes mediating fat storage. This, in turn, activates fat mobilization and lipolysis and reduces WAT mass (225). In addition to PPAR-γ, SIRT1 also interacts with PGC-1α, inducing the expression of mitochondrial genes involved in oxidative metabolism and fatty acid oxidation, while it also enhances leptin sensitivity by repressing PTP1B. The weight restricting effects of SIRT1 were further supported by experiments with resveratrol (RSV), a potent allosteric SIRT1 activator, which was shown to protect mice from diet-induced obesity (226). Furthermore, mice treated with SRT1720, a potent, selective synthetic activator of SIRT1, were resistant to diet-induced obesity due to enhanced oxidative metabolism in skeletal muscle, liver, and brown adipose tissue, indicating the positive metabolic consequences of specific SIRT1 activation (227). Currently, several pharmaceutical companies are investigating specific SIRT1 activators in Phase I and Phase II trials for the treatment of type II diabetes and obesity (228) to define their utility in the treatment of obesity and metabolic diseases.

 

Cyclic-GMP Signaling in Anti-Obesity Pharmacotherapy

 

Cyclic nucleotides, including 3-5-cyclic guanosine monophosphate (cGMP) and 3-5-cyclic adenosine monophosphate (cAMP), are second messengers important in many biological processes. Knowledge of the role of cAMP in the regulation of energy homeostasis has been extended, thanks to its intimate relationship with AMPK (AMP-activated protein kinase) signaling; intracellular cAMP activates the AMPK signaling pathway. AMPK regulates energy balance at both cellular and whole-body levels (229). Activation of AMPK facilitates fatty acid oxidation and mitochondria biogenesis, which promotes energy expenditure (230). Interestingly, activation of AMPK in the hypothalamus promotes food intake behavior (231). e.g. physiologic processes in the same direction and induces weight loss by mutual reinforcement. Moreover, off-the-shelf approaches might be possible, given the existence of an established market for medications targeting cGMP pathways, with FDA- and EMA-approved drugs such as sildenafil and linaclotide. Sildenafil acts on adipocytes, possibly through cGMP-dependent protein kinase I and mechanistic/mammalian target of rapamycin (mTOR) signaling pathways, browning subcutaneous white fat, thus increasing energy expenditure (232).

 

Beloranib

 

Beloranib is an analogue of the natural chemical compound fumagillin and is a methionine aminopeptidase 2 (MetAP2) inhibitor acting to reduce production of new fatty acid molecules by the liver and converting stored fats into useful energy (233). It was first tested in 31 obese women, who were divided into four groups (0.1mg, 0.3mg, 0.9mg, or placebo twice weekly). A dose-dependent weight loss was shown after four weeks of 0.9mg Beloranib administration with mean 3.8kg loss vs 0.6kg in the placebo group. It also improved lipid metabolism and lowered C-reactive protein and adiponectin. A phase II double-blinded, randomized clinical trial examined the efficacy and safety of Beloranib administration (234).147 obese patients were divided into four groups: 0.6, 1.2, 2.4 mg subcutaneous injection or placebo. After twelve weeks of administration, a dose-dependent weight loss of -5.5, -6.9, -10kg, respectively, was reported, vs -0.4kg in the placebo group. The main adverse events were sleep disturbance and gastrointestinal abnormalities. Beloranib may also cause robust weight loss and hypophagia in rats with hypothalamic and genetic obesity (235). In 2015, however, a phase III clinical trial for Prader-Willi was stopped after a second patient death (236). (see Table 25)

 

Table 25. Beloranib

FDA approved/Phase

Phase III aborted in 2015 after second patient death in Prader-Willi trial

Mechanism of action

Fumagillin analogue with methionine aminopeptidase 2 inhibition that reduced fatty acid synthesis in the liver and converted stored fat into useful energy; originally designed as an angiogenesis inhibitor

Clinical Benefits

↑ weight loss, hypophagia,

↓ lipid metabolism, CRP, adiponectin, cardiovascular factors

Adverse events

Sleep disturbance, gastrointestinal abnormalities

 

Fibroblast Growth Factor (FGF21)

 

Fibroblast growth factor (FGF) 21, expressed primarily in the liver, but also found in adipose tissue, skeletal muscle, and pancreas, is a member of the FGF family and acts as a metabolic regulator of body weight, glucose metabolism, and lipid metabolism (237). In WAT, FGF21 induces glucose uptake and adiponectin secretion with browning of white adipose tissue. In brown adipose tissue, it stimulates glucose uptake and thermogenesis, thus increasing energy expenditure. In the liver, it blocks GH signaling, regulates fatty acid oxidation both in the fasted state and in mice consuming high-fat, low-carbohydrate ketogenic diet and it maintains lipid homeostasis (238). FGF21 is characterized by anti-inflammatory, anti-oxidative stress properties with its circulating concentration increasing during periods of muscle activity or critical stress (239). Although, it is an attractive anti-obesity and anti-diabetes target, FGF21 levels are increased in obese ob/ob and db/db mice and correlate positively with BMI in humans. Exogenous administration of FGF21 in DIO in mice show virtually no beneficial effects on glucose tolerance and lipid metabolism, suggesting that the obesity state is FGF21-resistant (240).

 

ALTERNATIVE AND COMPLEMENTARY TYPES OF TREATMENT OF OBESITY

 

Gut Microbiota

 

Recently, a major shift in research has occurred towards the investigation of gut microbiota effects on energy expenditure and metabolism. Gut microbiota are responsible for a significant amount of the interaction between the host and the nutritional environment. Soluble fiber such as galacto-oligosaccharides and fructo-oligosaccharides (FOS), are fermented by the gut microbiota into short-chain fatty acids (SCFAs) acetate, propionate and butyrate (241). This mechanism provides to host 10% of its daily energy requirement (242). These SCFAs are an energy source for colonic epithelium, liver, and peripheral tissues (243). By fermenting nondigestible dietary fibers, host metabolism is enhanced. In mice with DIO, SCFAs improved glucose metabolism, insulin resistance, and obesity. In other animal studies, butyrate-producing bacteria (F. prausnitzii) induced secretion of glucagon-like peptide 1 (GLP1) from colonic L cells through the fatty acid receptor FFAR2(244). Furthermore, butyrate and propionate activate intestinal gluconeogenesis. Butyrate, through a cAMP-dependent mechanism, promotes the gene expression involved in intestinal gluconeogenesis. Propionate, itself a substrate for intestinal gluconeogenesis, activates its expression viaa gut-brain neural circuit involving the fatty acid receptor FFAR3 (245).

 

Given the key role played by microbiota in host nutrient processing and metabolism, it is not surprising that data points to a strong relation between gut microbiota and obesity and diabetes in humans. A reduced gut microbial diversity and altered microbiota composition is observed in obese individuals. There is also a low rate of gut microbial richness and specific bacterial groups are enriched or decreased in obese patients in comparison with lean people (246). Moreover, chronic diseases, such as obesity, diabetes, and HIV are associated with chronic low-grade inflammation. Gut microbiota regulates this inflammation through several mechanisms. Lipopolysaccharides (LPSs) from the outer membrane of Gram-negative bacteria may translocate through the intestinal border and cause subsequent systemic inflammation (247). Indeed, the intestinal barrier of obese patients is more permeable compared with that of lean individuals. Bile acids are characterized by a strong relation with gut microbiota affecting host’s body-weight homeostasis. Bile acids are microbially altered metabolites that are first endogenously produced by the liver and further metabolized by the gut microbiota (248). FXR signaling is an important pathway connecting gut microbiota and bile acids.

 

Based on the above knowledge, several interventions involving manipulation of the microbiome have been proposed as anti-obesity treatment. A diet which contains soluble fiber, prebiotics and/or probiotics could enhance the growth of beneficial gut microbiota and boost host metabolism. Lately, there has been interest in berberine administration in T1D, T2D, gestational diabetes, and prediabetes. The early reports of interventions using probiotics appear successful (249). Fecal microbiota transplant (FMT), the transfer of fecal suspension from a healthy (lean) donor into the gastrointestinal tract of an individual with disease (obesity) in order to restore a healthy gut is a potentially novel option to treat obesity.  However, there is not enough data about the safety of this method, that is why it is only FDA approved for recurrent Clostridium difficile infection.

 

Anti-Obesity Vaccines (Ghrelin, Somatostatin, Ad36)

 

The idea of a vaccination against obesity is also intriguing. The main action of these vaccines would be based on suppressing appetite-stimulating hormones or blocking food absorption. Three vaccines have been tested so far:

 

  1. An anti-ghrelin vaccine was found not only to reduce appetite by decreasing hypothalamic orexigenic signals but also to increase energy expenditure in rodent and pigs (250). Despite the promising results in rodents, clinical trials in humans showed no weight loss despite the development of ghrelin autoantibodies after four injections of anti-ghrelin vaccine (251). Another study, however, showed that IgG anti-ghrelin autoantibodies could protect ghrelin from degradation, suggesting that an autoimmune response may be involved in the orexigenic effects of ghrelin (252).

 

  1. An anti-somatostatin vaccine. Somatostatin is a peptide hormone which is produced, mainly, in the hypothalamus as well as other tissues, such as the gastrointestinal system. Somatostatin has the ability to suppress GH and insulin-like growth factor 1 (IGF-1) secretion. Reduced GH is associated with obesity and increased adiposity. So, the somatostatin vaccine could increase the secretion of GH and IGF-1(253). However, clinical trials in mice failed to reduce food intake, though a 10% improvement of body weight was observed (254).

 

  1. A live adenovirus 36 (Ad36) vaccine. Adenovirus 36 increases the risk of obesity in humans, characterized by increased inflammation and adiposity (255). Mice were injected with live Ad36 vaccine and compared to the control group (unvaccinated) after 14 weeks. The control group had 17% greater body weight and 20% more epididymal fats versus the vaccinated group, which also had decreased inflammatory cytokines and macrophages in fat tissue (256). (see Table 26)

 

Table 26. Anti-Obesity Vaccines

Drug name

Mechanism of action

Weight loss vs placebo

Clinical Benefits

Anti-obesity vaccine: somatostatin vaccine

Increases the secretion of GH, IGF-1

10%

 

Adenovirus 36

Live adenovirus36

 

Decreases body weight, epididymal fat in mice, inflammatory cytokines and macrophages

 

Nanomedicine

 

The introduction of nanomedicine in the field of obesity treatment is highly novel (257). Nanoparticles can achieve targeted drug delivery along with minimized side effects. The poor water-solubility of anti-obesity drugs can be overcome via nano-encapsulation. More specifically, nanoemulsion of orlistat has been tried in order to overcome its high lipophilicity, to improve its dissolution and to avoid the pancreatic lipase inhibition caused by this pharmaceutical agent in vivo (258). Additionally, a conjugated polymer-nanocarrier was created in order to reduce the side effects of orlistat (259). In 2014, the ability of mesoporous silica particles to reduce body weight was investigated (260). They found that the silica particles embedded in food could sequestrate lipase in their small pores through a lipase-specific interaction, leading to decreased absorption of fat.

 

Appetite suppression is an alternative method to decrease food intake and impact energy homeostasis (261). As mentioned above, however, anti-ghrelin vaccine was formed using virus-like particles for obesity treatment. The passive delivery of anti-ghrelin antibodies did not lead to long-term inhibition of food intake. So, to solve this problem, investigators immunoconjugated ghrelin with virus proteins to create a vaccine that was able to trigger an immune response leading to generation of specific anti-ghrelin antibodies. This anti-ghrelin vaccine played an important role in maintaining energy homeostasis in a DIO murine model.

 

In other examples, nanomedicine has enhanced the action of antiangiogenic agents in the treatment of obesity. Detailed above, antiangiogenic therapy inhibits the progression of adipocyte hyperplasia and reduces weight gain. A targeting nanoparticle was created in order to enhance the accumulation of the antiangiogenic drug in WATs by delivering it to vascular endothelial cells. Unlike WAT, brown adipose tissue (BAT) is full of mitochondria and a robust vascular structure helps to induce thermogenesis, increasing energy expenditure, and decreasing body weight. Thus, two nanoparticle platforms delivering browning agents to adipose tissue vasculature were formed (262). PPARγ nuclear receptor agonists (including rosiglitazone) have been shown to be characterized by anti-inflammatory properties against obesity and atherosclerosis. However, they are associated with severe side effects that limit their therapeutic use (263). In another, a mitochondria-targeted nanoparticle delivers the proposed anti-obesity compound PLGA-bPEG-triphenylphosphonium (TPP) polymer (264). The PEG shell extends the circulation time of nanoparticles, and TPP could facilitate the internalization into the matrix space of mitochondria to achieve targeted drug delivery.

 

Instead of targeted delivery, a localized and sustained release of a browning agent is a promising alternative for facilitation of WAT browning. Two nanoparticles, one injectable (265) and one in a painless microneedle array patch (266) were introduced. In vivo studies revealed successful delivery of the model drug into the human adipose tissue followed by ~15% decrease of weight gain after a four-week treatment.

 

CONCLUSION

 

The field of anti-obesity molecular pharmacotherapy is expanding. The homeostasis of body weight and metabolism are tightly linked to the central nervous system. The latter is characterized by centers that send orexigenic and anorexigenic signals regulating starvation and satiety, reducing and increasing energy expenditure, respectively. Pharmaceutical multi-agents in single compounds containing active portions of two or more drugs may allow for simultaneous effects on several synergistic pathways affecting appetite control and energy expenditure. Such medications could achieve increased weight loss with fewer side effects. Furthermore, the possibility of improved formulations (e.g., injectable forms of anti-obesity drugs and or once weekly verses daily administration) serve to enhance compliance. Considering that obesity is a multifactorial disease, it needs multimodal treatment. In an era where a variety of different therapeutic options is the norm for the management of chronic diseases such as type 2 diabetes and hypertension, the hope is that this process will led to better personalized anti-obesity treatments, focusing on the special characteristics, needs, and comorbidities of each patient and the effectiveness and safety of the recommended therapy. Thus, before starting any therapy, it will be important to record the detailed medical profile of the patient. Hereditary or acquired diseases, lifestyle parameters, and psychiatric history have to be taken into account when anti-obesity treatment is tailored for each patient. Further on the therapeutic horizon and still in much need of research are the place for altering gut microbiota balance and development of anti-obesity vaccines, novel peptide-mediated delivery of nuclear hormones, single molecular multi-agonists, and nanotechnologies that improve drug delivery and hold promise in the future of molecular pharmacotherapy of obesity.

 

REFERENCES

 

  1. Apovian CM, Aronne LJ, Bessesen DH, McDonnell ME, Murad MH, Pagotto U, et al. Pharmacological Management of Obesity: An Endocrine Society Clinical Practice Guideline. The Journal of Clinical Endocrinology & Metabolism. 2015;100(2):342-62.
  2. Kopelman P. Health risks associated with overweight and obesity. Obesity Reviews. 2007;8(s1):13-7.
  3. de Wit L, Luppino F, van Straten A, Penninx B, Zitman F, and Cuijpers P. Depression and obesity: A meta-analysis of community-based studies. Psychiatry Research. 2010;178(2):230-5.
  4. Pories WJ, Swanson MS, MacDonald KG, Long SB, Morris PG, Brown BM, et al. Who would have thought it? An operation proves to be the most effective therapy for adult-onset diabetes mellitus. Ann Surg. 1995;222(3):339-52.
  5. Dong Z, Xu L, Liu H, Lv Y, Zheng Q, and Li L. Comparative efficacy of five long-term weight loss drugs: quantitative information for medication guidelines. Obes Rev. 2017;18(12):1377-85.
  6. Ahima RS, and Flier JS. Leptin. Annu Rev Physiol. 2000;62:413-37.
  7. Montague CT, Farooqi IS, Whitehead JP, Soos MA, Rau H, Wareham NJ, et al. Congenital leptin deficiency is associated with severe early-onset obesity in humans. Nature. 1997;387(6636):903-8.
  8. Heymsfield SB, Greenberg AS, Fujioka K, Dixon RM, Kushner R, Hunt T, et al. Recombinant leptin for weight loss in obese and lean adults: a randomized, controlled, dose-escalation trial. Jama. 1999;282(16):1568-75
  9. Elchebly M, Payette P, Michaliszyn E, Cromlish W, Collins S, Loy AL, et al. Increased insulin sensitivity and obesity resistance in mice lacking the protein tyrosine phosphatase-1B gene. Science. 1999;283(5407):1544-8
  10. Picardi PK, Calegari VC, Prada PO, Moraes JC, Araujo E, Marcondes MC, et al. Reduction of hypothalamic protein tyrosine phosphatase improves insulin and leptin resistance in diet-induced obese rats. Endocrinology. 2008;149(8):3870-80.
  11. Ozcan L, Ergin AS, Lu A, Chung J, Sarkar S, Nie D, et al. Endoplasmic reticulum stress plays a central role in development of leptin resistance. Cell Metab. 2009;9(1):35-51.
  12. Vasselli JR. The Role of Dietary Components in Leptin Resistance. Advances in Nutrition. 2012;3(5):736-8.
  13. Munzberg H, Flier JS, and Bjorbaek C. Region-specific leptin resistance within the hypothalamus of diet-induced obese mice. Endocrinology. 2004;145(11):4880-9.
  14. Wang J, Obici S, Morgan K, Barzilai N, Feng Z, and Rossetti L. Overfeeding rapidly induces leptin and insulin resistance. Diabetes. 2001;50(12):2786-91.
  15. Roth JD, Roland BL, Cole RL, Trevaskis JL, Weyer C, Koda JE, et al. Leptin responsiveness restored by amylin agonism in diet-induced obesity: evidence from nonclinical and clinical studies. Proc Natl Acad Sci U S A. 2008;105(20):7257-62.
  16. Müller TD, Sullivan LM, Habegger K, Yi C-X, Kabra D, Grant E, et al. Restoration of leptin responsiveness in diet-induced obese mice using an optimized leptin analog in combination with exendin-4 or FGF21. Journal of Peptide Science. 2012;18(6):383-93.
  17. Clemmensen C, Chabenne J, Finan B, Sullivan L, Fischer K, Kuchler D, et al. GLP-1/glucagon coagonism restores leptin responsiveness in obese mice chronically maintained on an obesogenic diet. Diabetes. 2014;63(4):1422-7.
  18. Trevaskis JL, Wittmer C, Athanacio J, Griffin PS, Parkes DG, and Roth JD. Amylin/leptin synergy is absent in extreme obesity and not restored by calorie restriction-induced weight loss in rats. Obes Sci Pract. 2016;2(4):385-91.
  19. Ozcan L, Ergin AS, Lu A, Chung J, Sarkar S, Nie D, et al. Endoplasmic reticulum stress plays a central role in development of leptin resistance. Cell Metab. 2009;9(1):35-51.
  20. Liu J, Lee J, Salazar Hernandez MA, Mazitschek R, and Ozcan U. Treatment of obesity with celastrol. Cell. 2015;161(5):999-1011.
  21. Lee J, Liu J, Feng X, Salazar Hernandez MA, Mucka P, Ibi D, et al. Withaferin A is a leptin sensitizer with strong antidiabetic properties in mice. Nat Med. 2016;22(9):1023-32.
  22. Meehan CA, Cochran E, Kassai A, Brown RJ, and Gorden P. Metreleptin for injection to treat the complications of leptin deficiency in patients with congenital or acquired generalized lipodystrophy. Expert Rev Clin Pharmacol. 2016;9(1):59-68.
  23. Gordon CM, Ackerman KE, Berga SL, Kaplan JR, Mastorakos G, Misra M, et al. Functional Hypothalamic Amenorrhea: An Endocrine Society Clinical Practice Guideline. The Journal of Clinical Endocrinology & Metabolism. 2017;102(5):1413-39.
  24. Roth JD, Roland BL, Cole RL, Trevaskis JL, Weyer C, Koda JE, et al. Leptin r     responsiveness restored by amylin agonism in diet-induced obesity: evidence from nonclinical and clinical studies. Proc Natl Acad Sci U S A. 2008;105(20):7257-62.
  25. Aronne LJ, Halseth AE, Burns CM, Miller S, and Shen LZ. Enhanced weight loss following coadministration of pramlintide with sibutramine or phentermine in a multicenter trial. Obesity (Silver Spring). 2010;18(9):1739-46.
  26. Amylin and Takeda Discontinue Development of Pramlintide/Metreleptin Combination Treatment for Obesity Following Commercial Reassessment of the Program. 2011.
  27. Wellhoner P, Horster R, Jacobs F, Sayk F, Lehnert H, and Dodt C. Intranasal application of the melanocortin 4 receptor agonist MSH/ACTH(4-10) in humans causes lipolysis in white adipose tissue. Int J Obes (Lond). 2012;36(5):703-8.
  28. Fehm HL, Smolnik R, Kern W, McGregor GP, Bickel U, and Born J. The melanocortin melanocyte-stimulating hormone/adrenocorticotropin(4-10) decreases body fat in humans. J Clin Endocrinol Metab. 2001;86(3):1144-8.
  29. Chen KY, Muniyappa R, Abel BS, Mullins KP, Staker P, Brychta RJ, et al. RM-493, a melanocortin-4 receptor (MC4R) agonist, increases resting energy expenditure in obese individuals. J Clin Endocrinol Metab. 2015;100(4):1639-45.
  30. Low MJ. Neuroendocrinology: New hormone treatment for obesity caused by POMC-deficiency. Nat Rev Endocrinol. 2016;12(11):627-8.
  31. Kühnen P, Clément K, Wiegand S, Blankenstein O, Gottesdiener K, Martini LL, et al. Proopiomelanocortin Deficiency Treated with a Melanocortin-4 Receptor Agonist. New England Journal of Medicine. 2016;375(3):240-6.
  32. An investigational, melanocortin-4 receptor (MC4R) agonist in clinical development for the treatment of rare genetic disorders of obesity. https://www.rhythmtx.com/our-pipeline/.
  33. Pissios P, Bradley RL, and Maratos-Flier E. Expanding the scales: The multiple roles of MCH in regulating energy balance and other biological functions. Endocr Rev. 2006;27(6):606-20.
  34. Ito M, Ishihara A, Gomori A, Egashira S, Matsushita H, Mashiko S, et al. Melanin-concentrating hormone 1-receptor antagonist suppresses body weight gain correlated with high receptor occupancy levels in diet-induced obesity mice. Eur J Pharmacol. 2009;624(1-3):77-83.
  35. Mendez-Andino JL, and Wos JA. MCH-R1 antagonists: what is keeping most research programs away from the clinic? Drug Discov Today. 2007;12(21-22):972-9.
  36. trials.gov C. Safety, Pharmacokinetics and Pharmacodynamics Study to Evaluate BMS-830216 in Obese Subjects. https://clinicaltrials.gov/ct2/show/NCT00909766?term=BMS-830216&draw=2&rank=1.
  37. Heal DJ, Aspley S, Prow MR, Jackson HC, Martin KF, and Cheetham SC. Sibutramine: a novel anti-obesity drug. A review of the pharmacological evidence to differentiate it from d-amphetamine and d-fenfluramine. Int J Obes Relat Metab Disord. 1998;22 Suppl 1:S18-28; discussion S9.
  38. Wooltorton E. Obesity drug sibutramine (Meridia): hypertension and cardiac arrhythmias. CMAJ. 2002;166(10):1307-8.
  39. Launay JM, Herve P, Peoc'h K, Tournois C, Callebert J, Nebigil CG, et al. Function of the serotonin 5-hydroxytryptamine 2B receptor in pulmonary hypertension. Nat Med. 2002;8(10):1129-35.
  40. Fitzgerald LW, Burn TC, Brown BS, Patterson JP, Corjay MH, Valentine PA, et al. Possible role of valvular serotonin 5-HT(2B) receptors in the cardiopathy associated with fenfluramine. Mol Pharmacol. 2000;57(1):75-81.
  41. Dunlop J, Sabb AL, Mazandarani H, Zhang J, Kalgaonker S, Shukhina E, et al. WAY-163909 [(7bR, 10aR)-1,2,3,4,8,9,10,10a-octahydro-7bH-cyclopenta-[b][1,4]diazepino[6,7,1hi]indol e], a novel 5-hydroxytryptamine 2C receptor-selective agonist with anorectic activity. J Pharmacol Exp Ther. 2005;313(2):862-9.
  42. Siuciak JA, Chapin DS, McCarthy SA, Guanowsky V, Brown J, Chiang P, et al. CP-809,101, a selective 5-HT2C agonist, shows activity in animal models of antipsychotic activity. Neuropharmacology. 2007;52(2):279-90.
  43. John Dunlop SW, James E. Barrett, Joseph Coupet, Boyd Harrison, Hossein, Mazandarani SN, Menelas N. Pangalos, Siva Ramamoorthy, Lee, Schechter DS, Gary Stack, Jean Zhang, Guoming Zhang and Sharon, and Rosenzweig-Lipson. Characterization of Vabicaserin (SCA-136), a Selective 5-HT2C Receptor Agonist ASPET Journals. 2011,March 14.
  44. Burke LK, and Heisler LK. 5-Hydroxytryptamine Medications for the Treatment of Obesity. Journal of Neuroendocrinology. 2015;27(6):389-98.
  45. Higgins GA, and Fletcher PJ. Therapeutic Potential of 5-HT2C Receptor Agonists for Addictive Disorders. ACS Chem Neurosci. 2015;6(7):1071-88.
  46. Smith SR, Weissman NJ, Anderson CM, Sanchez M, Chuang E, Stubbe S, et al. Multicenter, placebo-controlled trial of lorcaserin for weight management. N Engl J Med. 2010;363(3):245-56.
  47. Fidler MC, Sanchez M, Raether B, Weissman NJ, Smith SR, Shanahan WR, et al. A one-year randomized trial of lorcaserin for weight loss in obese and overweight adults: the BLOSSOM trial. J Clin Endocrinol Metab. 2011;96(10):3067-77.
  48. FDA approves Belviq to treat some overweight or obese adults. Home Healthc Nurse. 2012;30(8):443-4.
  49. Anderberg RH, Richard JE, Eerola K, Lopez-Ferreras L, Banke E, Hansson C, et al. Glucagon-Like Peptide 1 and Its Analogs Act in the Dorsal Raphe and Modulate Central Serotonin to Reduce Appetite and Body Weight. Diabetes. 2017;66(4):1062-73.
  50. Bohula EA, Wiviott SD, McGuire DK, Inzucchi SE, Kuder J, Im K, et al. Cardiovascular Safety of Lorcaserin in Overweight or Obese Patients. New England Journal of Medicine. 2018;379(12):1107-17.
  51. Bohula EA, Scirica BM, Inzucchi SE, McGuire DK, Keech AC, Smith SR, et al. Effect of lorcaserin on prevention and remission of type 2 diabetes in overweight and obese patients (CAMELLIA-TIMI 61): a randomised, placebo-controlled trial. Lancet. 2018;392(10161):2269-79.
  52. Zarrindast MR, and Hosseini-Nia T. Anorectic and behavioural effects of bupropion. Gen Pharmacol. 1988;19(2):201-4.
  53. Billes SK, and Cowley MA. Inhibition of dopamine and norepinephrine reuptake produces additive effects on energy balance in lean and obese mice. Neuropsychopharmacology. 2007;32(4):822-34.
  54. Greenway FL, Whitehouse MJ, Guttadauria M, Anderson JW, Atkinson RL, Fujioka K, et al. Rational design of a combination medication for the treatment of obesity. Obesity (Silver Spring). 2009;17(1):30-9.
  55. Anderson JW, Greenway FL, Fujioka K, Gadde KM, McKenney J, and O'Neil PM. Bupropion SR enhances weight loss: a 48-week double-blind, placebo- controlled trial. Obes Res. 2002;10(7):633-41.
  56. Jain AK, Kaplan RA, Gadde KM, Wadden TA, Allison DB, Brewer ER, et al. Bupropion SR vs. placebo for weight loss in obese patients with depressive symptoms. Obes Res. 2002;10(10):1049-56.
  57. Gadde KM, Parker CB, Maner LG, Wagner HR, 2nd, Logue EJ, Drezner MK, et al. Bupropion for weight loss: aninvestigation of efficacy and tolerability in overweight and obese women. Obes Res. 2001;9(9):544-51.
  58. Cowley MA, Smart JL, Rubinstein M, Cerdan MG, Diano S, Horvath TL, et al. Leptin activates anorexigenic POMC neurons through a neural network in the arcuate nucleus. Nature. 2001;411(6836):480-4.
  59. Yeomans MR, and Gray RW. Opioid peptides and the control of human ingestive behaviour. Neurosci Biobehav Rev. 2002;26(6):713-28.
  60. Mitchell JE, Morley JE, Levine AS, Hatsukami D, Gannon M, and Pfohl D. High-dose naltrexone therapy and dietary counseling for obesity. Biol Psychiatry. 1987;22(1):35-42.
  61. Greenway FL, Fujioka K, Plodkowski RA, Mudaliar S, Guttadauria M, Erickson J, et al. Effect of naltrexone plusbupropion on weight loss in overweight and obese adults (COR-I): a multicentre, randomised, double-blind, placebo-controlled, phase 3 trial. Lancet. 2010;376(9741):595-605.
  62. Calderone A, Calabro PF, Lippi C, Jaccheri R, Vitti J, and Santini F. Psychopathological Behaviour and Cognition in Morbid Obesity. Recent Pat Endocr Metab Immune Drug Discov. 2017;10(2):112-8.
  63. Apovian CM, Aronne L, Rubino D, Still C, Wyatt H, Burns C, et al. A randomized, phase 3 trial of naltrexone SR/bupropion SR on weight and obesity-related risk factors (COR-II). Obesity (Silver Spring). 2013;21(5):935-43.
  64. Wadden TA, Foreyt JP, Foster GD, Hill JO, Klein S, O'Neil PM, et al. Weight loss with naltrexone SR/bupropion SR combination therapy as an adjunct to behavior modification: the COR-BMOD trial. Obesity (Silver Spring). 2011;19(1):110-20.
  65. Hollander P, Gupta AK, Plodkowski R, Greenway F, Bays H, Burns C, et al. Effects of naltrexone sustained-release/bupropion sustained-release combination therapy on body weight and glycemic parameters in overweight and obese patients with type 2 diabetes. Diabetes Care. 2013;36(12):4022-9.
  66. Nissen SE, Wolski KE, Prcela L, Wadden T, Buse JB, Bakris G, et al. Effect of Naltrexone-Bupropion on Major Adverse Cardiovascular Events in Overweight and Obese Patients with Cardiovascular Risk Factors: A Randomized Clinical Trial. Jama. 2016;315(10):990-1004.
  67. Okada M, Kaneko S, Hirano T, Mizuno K, Kondo T, Otani K, et al. Effects of zonisamide on dopaminergic system. Epilepsy Res. 1995;22(3):193-205.
  68. Okada M, Hirano T, Kawata Y, Murakami T, Wada K, Mizuno K, et al. Biphasic effects of zonisamide on serotonergic system in rat hippocampus. Epilepsy Res. 1999;34(2-3):187-97.
  69. Gadde KM, Franciscy DM, Wagner HR, 2nd, and Krishnan KR. Zonisamide for weight loss in obese adults: a randomized controlled trial. Jama. 2003;289(14):1820-5.
  70. Lim J, Ko YH, Joe SH, Han C, Lee MS, and Yang J. Zonisamide produces weight loss in psychotropic drug-treated psychiatric outpatients. Prog Neuropsychopharmacol Biol Psychiatry. 2011;35(8):1918-21.
  71. McElroy SL, Kotwal R, Hudson JI, Nelson EB, and Keck PE. Zonisamide in the treatment of binge-eating disorder: an open-label, prospective trial. J Clin Psychiatry. 2004;65(1):50-6.
  72. Gadde KM, Yonish GM, Foust MS, and Wagner HR. Combination therapy of zonisamide and bupropion forweight reduction in obese women: a preliminary, randomized, open-label study. J Clin Psychiatry.2007;68(8):1226-9.
  73. Ohtahara S, and Yamatogi Y. Safety of zonisamide therapy: prospective follow-up survey. Seizure. 2004;13:S50-S5.
  74. Rosenfeld WE. Topiramate: a review of preclinical, pharmacokinetic, and clinical data. Clin Ther. 1997;19(6):1294-308.
  75. Bray GA, Hollander P, Klein S, Kushner R, Levy B, Fitchet M, et al. A 6-month randomized, placebo-controlled, dose-ranging trial of topiramate for weight loss in obesity. Obes Res. 2003;11(6):722-33.
  76. Wilding J, Van Gaal L, Rissanen A, Vercruysse F, and Fitchet M. A randomized double-blind placebo-controlledstudy of the long-term efficacy and safety of topiramate in the treatment of obese subjects. Int J Obes Relat Metab Disord. 2004;28(11):1399-410.
  77. McElroy SL, Arnold LM, Shapira NA, Keck PE, Jr., Rosenthal NR, Karim MR, et al. Topiramate in the treatmentof binge eating disorder associated with obesity: a randomized, placebo-controlled trial. Am J Psychiatry. 2003;160(2):255-61.
  78. Hendricks EJ, Srisurapanont M, Schmidt SL, Haggard M, Souter S, Mitchell CL, et al. Addiction potential of phentermine prescribed during long-term treatment of obesity. Int J Obes (Lond). 2014;38(2):292-8.
  79. Samanin R, and Garattini S. Neurochemical mechanism of action of anorectic drugs. Pharmacol Toxicol. 1993;73(2):63-8
  80. Yanovski SZ, and Yanovski JA. Long-term drug treatment for obesity: a systematic and clinical review. Jama. 2014;311(1):74-86.
  81. Munro JF, MacCuish AC, Wilson EM, and Duncan LJ. Comparison of continuous and intermittent anorectic therapy in obesity. Br Med J. 1968;1(5588):352-4.
  82. Lewis KH, Fischer H, Ard J, Barton L, Bessesen DH, Daley MF, et al. Safety and Effectiveness of Longer-Term Phentermine Use: Clinical Outcomes from an Electronic Health Record Cohort. Obesity (Silver Spring). 2019;27(4):591-602.
  83. Antel J, and Hebebrand J. Weight-reducing side effects of the antiepileptic agents topiramate and zonisamide. Handb Exp Pharmacol. 2012(209):433-66.
  84. Allison DB, Gadde KM, Garvey WT, Peterson CA, Schwiers ML, Najarian T, et al. Controlled-release phentermine/topiramate in severely obese adults: a randomized controlled trial (EQUIP). Obesity (Silver Spring). 2012;20(2):330-42.
  85. Gadde KM, Allison DB, Ryan DH, Peterson CA, Troupin B, Schwiers ML, et al. Effects of low-dose, controlled-release, phentermine plus topiramate combination on weight and associated comorbidities in overweight and obese adults (CONQUER): a randomised, placebo-controlled, phase 3 trial. Lancet. 2011;377(9774):1341-52.
  86. Garvey WT, Ryan DH, Look M, Gadde KM, Allison DB, Peterson CA, et al. Two-year sustained weight loss and metabolic benefits with controlled-release phentermine/topiramate in obese and overweight adults (SEQUEL): a randomized, placebo-controlled, phase 3 extension study. Am J Clin Nutr. 2012;95(2):297-308.
  87. Erondu N, Gantz I, Musser B, Suryawanshi S, Mallick M, Addy C, et al. Neuropeptide Y5 receptor antagonism does not induce clinically meaningful weight loss in overweight and obese adults. Cell Metab. 2006;4(4):275-82.
  88. Sargent BJ, and Moore NA. New central targets for the treatment of obesity. Br J Clin Pharmacol. 2009;68(6):852-60.
  89. Double-Blind, Multi-Center, Randomized Study to Assess the Efficacy and Safety of Velneperit (S-2367) and Orlistat Administered Individually or Combined with a Reduced Calorie Diet (RCD) in Obese Subjects. https://clinicaltrials.gov/ct2/show/NCT01126970.
  90. Nathan PJ, O'Neill BV, Mogg K, Bradley BP, Beaver J, Bani M, et al. The effects of the dopamine D(3) receptor antagonist GSK598809 on attentional bias to palatable food cues in overweight and obese subjects. Int J Neuropsychopharmacol. 2012;15(2):149-61.
  91. Astrup A, Madsbad S, Breum L, Jensen TJ, Kroustrup JP, and Larsen TM. Effect of tesofensine on bodyweight loss, body composition, and quality of life in obese patients: a randomised, double-blind, placebo-controlled trial. Lancet. 2008;372(9653):1906-13.
  92. Sjodin A, Gasteyger C, Nielsen AL, Raben A, Mikkelsen JD, Jensen JK, et al. The effect of the triple monoamine reuptake inhibitor tesofensine on energy metabolism and appetite in overweight and moderately obese men. Int J Obes (Lond). 2010;34(11):1634-43.
  93. Gilbert JA, Gasteyger C, Raben A, Meier DH, Astrup A, and Sjodin A. The effect of tesofensine on appetite sensations. Obesity (Silver Spring). 2012;20(3):553-61.
  94. Schoedel KA, Meier D, Chakraborty B, Manniche PM, and Sellers EM. Subjective and objective effects of the novel triple reuptake inhibitor tesofensine in recreational stimulant users. Clin Pharmacol Ther. 2010;88(1):69-78.
  95. Effect of Tesofensine on Weight Reduction in Patients with Obesity. . Updated April 22, 2013.
  96. McElroy SL, Mitchell JE, Wilfley D, Gasior M, Ferreira-Cornwell MC, McKay M, et al. LisdexamfetamineDimesylate Effects on Binge Eating Behaviour and Obsessive-Compulsive and Impulsive Features in Adults with Binge Eating Disorder. Eur Eat Disord Rev. 2016;24(3):223-31.
  97. Greenway FL, and Bray GA. Human chorionic gonadotropin (HCG) in the treatment of obesity: a critical assessment of the Simeons method. West J Med. 1977;127(6):461-3.
  98. Despres JP, Golay A, and Sjostrom L. Effects of rimonabant on metabolic risk factors in overweight patients with dyslipidemia. N Engl J Med. 2005;353(20):2121-34.
  99. Randall PA, Vemuri VK, Segovia KN, Torres EF, Hosmer S, Nunes EJ, et al. The novel cannabinoid CB1 antagonist AM6545 suppresses food intake and food-reinforced behavior. Pharmacol Biochem Behav. 2010;97(1):179-84.
  100. Bosch B, Venter I, Stewart RI, and Bertram SR. Human chorionic gonadotrophin and weight loss. A double-blind, placebo-controlled trial. S Afr Med J. 1990;77(4):185-9.
  101. Oh IS, Shimizu H, Satoh T, Okada S, Adachi S, Inoue K, et al. Identification of nesfatin-1 as a satiety molecule in the hypothalamus. Nature. 2006;443(7112):709-12.
  102. Gonzalez R, Reingold BK, Gao X, Gaidhu MP, Tsushima RG, and Unniappan S. Nesfatin-1 exerts a direct, glucose-dependent insulinotropic action on mouse islet beta- and MIN6 cells. J Endocrinol. 2011;208(3):R9-r16.
  103. Prinz P, Teuffel P, Lembke V, Kobelt P, Goebel-Stengel M, Hofmann T, et al. Nesfatin-130-59 Injected Intracerebroventricularly Differentially Affects Food Intake Microstructure in Rats Under Normal Weight and Diet-Induced Obese Conditions. Front Neurosci. 2015;9:422.
  104. Gibbs J, Young RC, and Smith GP. Cholecystokinin elicits Satiety in Rats with Open Gastric Fistulas. Nature. 1973;245(5424):323-5.
  105. Moran TH, and Kinzig KP. Gastrointestinal satiety signals II. Cholecystokinin. Am J Physiol Gastrointest Liver Physiol. 2004;286(2):G183-8.
  106. Moran TH, Baldessarini AR, Salorio CF, Lowery T, and Schwartz GJ. Vagal afferent and efferent contributions to the inhibition of food intake by cholecystokinin. Am J Physiol. 1997;272(4 Pt 2):R1245-51.
  107. Kissileff HR, Carretta JC, Geliebter A, and Pi-Sunyer FX. Cholecystokinin and stomach distension combine to reduce food intake in humans. Am J Physiol Regul Integr Comp Physiol. 2003;285(5):R992-8.
  108. Muurahainen N, Kissileff HR, Derogatis AJ, and Pi-Sunyer FX. Effects of cholecystokinin-octapeptide (CCK-8) on food intake and gastric emptying in man. Physiol Behav. 1988;44(4-5):645-9.
  109. West DB, Fey D, and Woods SC. Cholecystokinin persistently suppresses meal size but not food intake in free-feeding rats. Am J Physiol. 1984;246(5 Pt 2):R776-87.
  110. Castillo EJ, Delgado-Aros S, Camilleri M, Burton D, Stephens D, O'Connor-Semmes R, et al. Effect of oral CCK-1 agonist GI181771X on fasting and postprandial gastric functions in healthy volunteers. Am J Physiol Gastrointest Liver Physiol. 2004;287(2):G363-9.
  111. Morton GJ, Cummings DE, Baskin DG, Barsh GS, and Schwartz MW. Central nervous system control of food intake and body weight. Nature. 2006;443(7109):289-95.
  112. Woods SC, Lutz TA, Geary N, and Langhans W. Pancreatic signals controlling food intake; insulin, glucagon and amylin. Philos Trans R Soc Lond B Biol Sci. 2006;361(1471):1219-35.
  113. Verdich C, Flint A, Gutzwiller JP, Naslund E, Beglinger C, Hellstrom PM, et al. A meta-analysis of the effect of glucagon-like peptide-1 (7-36) amide on ad libitum energy intake in humans. J Clin Endocrinol Metab. 2001;86(9):4382-9.
  114. Buse JM, Leigh; Stonehouse, Anthony; Guan, Xuesong; Malone, James; Okerson, Ted; Maggs, David; Kim, Dennis. Exenatide Maintained Glycemic Control with Associated Weight Reduction Over Three Years in Patients with Type 2 Diabetes. Diabetes. June 2007; Vol. 56, pA73.
  115. Abbott CR, Monteiro M, Small CJ, Sajedi A, Smith KL, Parkinson JR, et al. The inhibitory effects of peripheral administration of peptide YY(3-36) and glucagon-like peptide-1 on food intake are attenuated by ablation of the vagal-brainstem-hypothalamic pathway. Brain Res. 2005;1044(1):127-31.
  116. Baggio LL, Huang Q, Brown TJ, and Drucker DJ. Oxyntomodulin and glucagon-like peptide-1 differentially regulate murine food intake and energy expenditure. Gastroenterology. 2004;127(2):546-58.
  117. Drucker DJ. The biology of incretin hormones. Cell Metab. 2006;3(3):153-65.
  118. Pi-Sunyer X, Astrup A, Fujioka K, Greenway F, Halpern A, Krempf M, et al. A Randomized, Controlled Trial of 3.0 mg of Liraglutide in Weight Management. New England Journal of Medicine. 2015;373(1):11-22.
  119. Davies MJ, Bergenstal R, Bode B, Kushner RF, Lewin A, Skjoth TV, et al. Efficacy of Liraglutide for Weight Loss Among Patients with Type 2 Diabetes: The SCALE Diabetes Randomized Clinical Trial. Jama. 2015;314(7):687-99.
  120. Wadden TA, Hollander P, Klein S, Niswender K, Woo V, Hale PM, et al. Weight maintenance and additional weight loss with liraglutide after low-calorie-diet-induced weight loss: the SCALE Maintenance randomized study. Int J Obes (Lond). 2013;37(11):1443-51.
  121. Aroda VR, Ahmann A, Cariou B, Chow F, Davies MJ, Jodar E, et al. Comparative efficacy, safety, and cardiovascular outcomes with once-weekly subcutaneous semaglutide in the treatment of type 2 diabetes: Insights from the SUSTAIN 1-7 trials. Diabetes Metab. 2019;45(5):409-18.
  122. Husain M, Birkenfeld AL, Donsmark M, Dungan K, Eliaschewitz FG, Franco DR, et al. Oral Semaglutide and Cardiovascular Outcomes in Patients with Type 2 Diabetes. N Engl J Med. 2019;381(9):841-51.
  123. Fosgerau K, and Hoffmann T. Peptide therapeutics: current status and future directions. Drug Discov Today. 2015;20(1):122-8.
  124. Semaglutide Effects on Heart Disease and Stroke in Patients with Overweight or Obesity (SELECT). https://clinicaltrials.gov/ct2/show/NCT03574597. Updated 2019.
  125. Barnett AH. Lixisenatide: evidence for its potential use in the treatment of type 2 diabetes. Core Evid. 2011;6:67-79.
  126. Pinelli NR, and Hurren KM. Efficacy and safety of long-acting glucagon-like peptide-1 receptor agonistscompared with exenatide twice daily and sitagliptin in type 2 diabetes mellitus: a systematic review and meta-analysis. Ann Pharmacother. 2011;45(7-8):850-60.
  127. Madsbad S, Kielgast U, Asmar M, Deacon CF, Torekov SS, and Holst JJ. An overview of once-weekly glucagon-like peptide-1 receptor agonists-available efficacy and safety data and perspectives for the future. Diabetes Obes Metab. 2011;13(5):394-407
  128. Steinert RE, Poller B, Castelli MC, Drewe J, and Beglinger C. Oral administration of glucagon-like peptide 1 orpeptide YY 3-36 affects food intake in healthy male subjects. Am J Clin Nutr. 2010;92(4):810-7.
  129. Trevaskis JL, Sun C, Athanacio J, D'Souza L, Samant M, Tatarkiewicz K, et al. Synergistic metabolic benefits of an exenatide analogue and cholecystokinin in diet-induced obese and leptin-deficient rodents. Diabetes Obes Metab. 2015;17(1):61-73.
  130. Clemmensen C, Finan B, Fischer K, Tom RZ, Legutko B, Sehrer L, et al. Dual melanocortin-4 receptor and GLP-1 receptor agonism amplifies metabolic benefits in diet-induced obese mice. EMBO Mol Med. 2015;7(3):288-98.
  131. Muller TD, Finan B, Clemmensen C, DiMarchi RD, and Tschop MH. The New Biology and Pharmacology of Glucagon. Physiol Rev. 2017;97(2):721-66.
  132. Cegla J, Troke RC, Jones B, Tharakan G, Kenkre J, McCullough KA, et al. Coinfusion of low-dose GLP-1 and glucagon in man results in a reduction in food intake. Diabetes. 2014;63(11):3711-20.
  133. Day JW, Ottaway N, Patterson JT, Gelfanov V, Smiley D, Gidda J, et al. A new glucagon and GLP-1 co-agonist eliminate obesity in rodents. Nat Chem Biol. 2009;5(10):749-57.
  134. Clemmensen C, Chabenne J, Finan B, Sullivan L, Fischer K, Kuchler D, et al. GLP-1/glucagon coagonism restores leptin responsiveness in obese mice chronically maintained on an obesogenic diet. Diabetes. 2014;63(4):1422-7.
  135. Brandt SJ, Gotz A, Tschop MH, and Muller TD. Gut hormone polyagonists for the treatment of type 2 diabetes. Peptides. 2018;100:190-201.
  136. 136. Dakin CL, Gunn I, Small CJ, Edwards CM, Hay DL, Smith DM, et al. Oxyntomodulin inhibits food intake in the rat. Endocrinology. 2001;142(10):4244-50.
  137. Dakin CL, Small CJ, Batterham RL, Neary NM, Cohen MA, Patterson M, et al. Peripheral oxyntomodulin reduces food intake and body weight gain in rats. Endocrinology. 2004;145(6):2687-95.
  138. Yamamoto H, Lee CE, Marcus JN, Williams TD, Overton JM, Lopez ME, et al. Glucagon-like peptide-1 receptor stimulation increases blood pressure and heart rate and activates autonomic regulatory neurons. J Clin Invest. 2002;110(1):43-52.
  139. Cohen MA, Ellis SM, Le Roux CW, Batterham RL, Park A, Patterson M, et al. Oxyntomodulin suppresses appetite and reduces food intake in humans. J Clin Endocrinol Metab. 2003;88(10):4696-701.
  140. Wynne K, Park AJ, Small CJ, Meeran K, Ghatei MA, Frost GS, et al. Oxyntomodulin increases energy expenditure in addition to decreasing energy intake in overweight and obese humans: a randomised controlled trial. Int J Obes (Lond). 2006;30(12):1729-36.
  141. Baggio LL, Huang Q, Brown TJ, and Drucker DJ. Oxyntomodulin and glucagon-like peptide-1 differentially regulate murine food intake and energy expenditure. Gastroenterology. 2004;127(2):546-58.
  142. Schjoldager B, Mortensen PE, Myhre J, Christiansen J, and Holst JJ. Oxyntomodulin from distal gut. Role in regulation of gastric and pancreatic functions. Dig Dis Sci. 1989;34(9):1411-9.
  143. Santoprete A, Capito E, Carrington PE, Pocai A, Finotto M, Langella A, et al. DPP-IV-resistant, long-actingoxyntomodulin derivatives. J Pept Sci. 2011;17(4):270-80.
  144. Pocai A, Carrington PE, Adams JR, Wright M, Eiermann G, Zhu L, et al. Glucagon-like peptide 1/glucagon receptor dual agonism reverses obesity in mice. Diabetes. 2009;58(10):2258-66.
  145. Bello NT, Kemm MH, Ofeldt EM, and Moran TH. Dose combinations of exendin-4 and salmon calcitonin produce additive and synergistic reductions in food intake in nonhuman primates. Am J Physiol Regul Integr Comp Physiol. 2010;299(3):R945-52.
  146. Sun C, Trevaskis JL, Jodka CM, Neravetla S, Griffin P, Xu K, et al. Bifunctional PEGylated exenatide-amylinomimetic hybrids to treat metabolic disorders: an example of long-acting dual hormonal therapeutics. J Med Chem. 2013;56(22):9328-41.
  147. Takeda J, Seino Y, Tanaka K, Fukumoto H, Kayano T, Takahashi H, et al. Sequence of an intestinal cDNA encoding human gastric inhibitory polypeptide precursor. Proc Natl Acad Sci U S A. 1987;84(20):7005-8
  148. Dupre J, Ross SA, Watson D, and Brown JC. Stimulation of insulin secretion by gastric inhibitory polypeptide in man. J Clin Endocrinol Metab. 1973;37(5):826-8.
  149. Pederson RA, and Brown JC. Interaction of gastric inhibitory polypeptide, glucose, and arginine on insulin and glucagon secretion from the perfused rat pancreas. Endocrinology. 1978;103(2):610-5.
  150. Oben J, Morgan L, Fletcher J, and Marks V. Effect of the entero-pancreatic hormones, gastric inhibitory polypeptide and glucagon-like polypeptide-1(7-36) amide, on fatty acid synthesis in explants of rat adipose tissue. J Endocrinol. 1991;130(2):267-72.
  151. Eckel RH, Fujimoto WY, and Brunzell JD. Gastric inhibitory polypeptide enhanced lipoprotein lipase activity in cultured preadipocytes. Diabetes. 1979;28(12):1141-2.
  152. Kim SJ, Nian C, Karunakaran S, Clee SM, Isales CM, and McIntosh CH. GIP-overexpressing mice demonstrate reduced diet-induced obesity and steatosis, and improved glucose homeostasis. PLoS One. 2012;7(7):e40156.
  153. Martin CM, Irwin N, Flatt PR, and Gault VA. A novel acylated form of (d-Ala(2))GIP with improved antidiabetic potential, lacking effect on body fat stores. Biochim Biophys Acta. 2013;1830(6):3407-13.
  154. Finan B, Ma T, Ottaway N, Muller TD, Habegger KM, Heppner KM, et al. Unimolecular dual incretins maximize metabolic benefits in rodents, monkeys, and humans. Sci Transl Med. 2013;5(209):209ra151.
  155. Finan B, Yang B, Ottaway N, Smiley DL, Ma T, Clemmensen C, et al. A rationally designed monomeric peptide triagonist corrects obesity and diabetes in rodents. Nat Med. 2015;21(1):27-36.
  156. Mauvais-Jarvis F, Manson JE, Stevenson JC, and Fonseca VA. Menopausal Hormone Therapy and Type 2 Diabetes Prevention: Evidence, Mechanisms, and Clinical Implications. Endocr Rev. 2017;38(3):173-88.
  157. Gao Q, Mezei G, Nie Y, Rao Y, Choi CS, Bechmann I, et al. Anorectic estrogen mimics leptin's effect on the rewiring of melanocortin cells and Stat3 signaling in obese animals. Nat Med. 2007;13(1):89-94.
  158. Finan B, Yang B, Ottaway N, Stemmer K, Muller TD, Yi CX, et al. Targeted estrogen delivery reverses the metabolic syndrome. Nat Med. 2012;18(12):1847-56.
  159. Schwenk RW, Baumeier C, Finan B, Kluth O, Brauer C, Joost HG, et al. GLP-1-oestrogen attenuates hyperphagia and protects from beta cell failure in diabetes-prone New Zealand obese (NZO) mice. Diabetologia. 2015;58(3):604-14.
  160. Angelin B, and Rudling M. Lipid lowering with thyroid hormone and thyromimetics. Curr Opin Lipidol. 2010;21(6):499-506.
  161. Lopez M, Varela L, Vazquez MJ, Rodriguez-Cuenca S, Gonzalez CR, Velagapudi VR, et al. Hypothalamic AMPK and fatty acid metabolism mediate thyroid regulation of energy balance. Nat Med. 2010;16(9):1001-8.
  162. Finan B, Clemmensen C, Zhu Z, Stemmer K, Gauthier K, Muller L, et al. Chemical Hybridization of Glucagon and Thyroid Hormone Optimizes Therapeutic Impact for Metabolic Disease. Cell. 2016;167(3):843-57.e14.
  163. Hotamisligil GS. Inflammation and metabolic disorders. Nature. 2006;444(7121):860-7.
  164. Quarta C, Clemmensen C, Zhu Z, Yang B, Joseph SS, Lutter D, et al. Molecular Integration of Incretin and Glucocorticoid Action Reverses Immunometabolic Dysfunction and Obesity. Cell Metab. 2017;26(4):620-32.e6.
  165. Pironi L, Stanghellini V, Miglioli M, Corinaldesi R, De Giorgio R, Ruggeri E, et al. Fat-induced ileal brake in humans: a dose-dependent phenomenon correlated to the plasma levels of peptide YY. Gastroenterology. 1993;105(3):733-9.
  166. Batterham RL, Cohen MA, Ellis SM, Le Roux CW, Withers DJ, Frost GS, et al. Inhibition of food intake in obese subjects by peptide YY3-36. N Engl J Med. 2003;349(10):941-8.
  167. Degen L, Oesch S, Casanova M, Graf S, Ketterer S, Drewe J, et al. Effect of peptide YY3-36 on food intake in humans. Gastroenterology. 2005;129(5):1430-6.
  168. Field BC, Wren AM, Peters V, Baynes KC, Martin NM, Patterson M, et al. PYY3-36 and oxyntomodulin can beadditive in their effect on food intake in overweight and obese humans. Diabetes. 2010;59(7):1635-9.
  169. Talsania T, Anini Y, Siu S, Drucker DJ, and Brubaker PL. Peripheral exendin-4 and peptide YY(3-36) synergistically reduce food intake through different mechanisms in mice. Endocrinology. 2005;146(9):3748-56.
  170. Scott V, Kimura N, Stark JA, and Luckman SM. Intravenous peptide YY3-36 and Y2 receptor antagonism in the rat: effects on feeding behaviour. J Neuroendocrinol. 2005;17(7):452-7.
  171. Moran TH, Smedh U, Kinzig KP, Scott KA, Knipp S, and Ladenheim EE. Peptide YY(3-36) inhibits gastric emptying and produces acute reductions in food intake in rhesus monkeys. Am J Physiol Regul Integr Comp Physiol. 2005;288(2):R384-8.
  172. Batterham RL, Cowley MA, Small CJ, Herzog H, Cohen MA, Dakin CL, et al. Gut hormone PYY(3-36) physiologically inhibits food intake. Nature. 2002;418(6898):650-4.
  173. 7TM Pharma Initiates Phase II Clinical Study with the Drug Candidate Obinepitide for the Treatment of Obesity. https://www.biospace.com/article/releases/7tm-pharma-initiates-phase-ii-clinical-study-with-the-drug-candidate-obinepitide-for-the-treatment-of-obesity-/.
  174. Kjaergaard M, Salinas CBG, Rehfeld JF, Secher A, Raun K, and Wulff BS. PYY(3-36) and exendin-4 reduce food intake and activate neuronal circuits in a synergistic manner in mice. Neuropeptides. 2019;73:89-95.
  175. Cummings DE, Foster-Schubert KE, and Overduin J. Ghrelin and energy balance: focus on current controversies. Curr Drug Targets. 2005;6(2):153-69.
  176. Date Y, Murakami N, Toshinai K, Matsukura S, Niijima A, Matsuo H, et al. The role of the gastric afferent vagalnerve in ghrelin-induced feeding and growth hormone secretion in rats. Gastroenterology. 2002;123(4):1120-8.
  177. Wren AM, Seal LJ, Cohen MA, Brynes AE, Frost GS, Murphy KG, et al. Ghrelin enhances appetite and increases food intake in humans. J Clin Endocrinol Metab. 2001;86(12):5992.
  178. Zorrilla EP, Iwasaki S, Moss JA, Chang J, Otsuji J, Inoue K, et al. Vaccination against weight gain. Proc Natl Acad Sci U S A. 2006;103(35):13226-31.
  179. Moran TH, and Dailey MJ. Minireview: Gut peptides: targets for antiobesity drug development? Endocrinology. 2009;150(6):2526-30.
  180. Xin Z, Serby MD, Zhao H, Kosogof C, Szczepankiewicz BG, Liu M, et al. Discovery and pharmacologicalevaluation of growth hormone secretagogue receptor antagonists. J Med Chem. 2006;49(15):4459-69.
  181. Yang J, Zhao TJ, Goldstein JL, and Brown MS. Inhibition of ghrelin O-acyltransferase (GOAT) by octanoylated pentapeptides. Proc Natl Acad Sci U S A. 2008;105(31):10750-5.
  182. Li Z, Mulholland M, and Zhang W. Ghrelin O-acyltransferase (GOAT) and energy metabolism. Science China Life Sciences. 2016;59(3):281-91.
  183. Barnett BP, Hwang Y, Taylor MS, Kirchner H, Pfluger PT, Bernard V, et al. Glucose and weight control in mice with a designed ghrelin O-acyltransferase inhibitor. Science. 2010;330(6011):1689-92.
  184. Teubner BJ, Garretson JT, Hwang Y, Cole PA, and Bartness TJ. Inhibition of ghrelin O-acyltransferase attenuates food deprivation-induced increases in ingestive behavior. Horm Behav. 2013;63(4):667-73.
  185. Price DJ, Drewry DH, Schaller LT, Thompson BD, Reid PR, Maloney PR, et al. An orally available, brain-penetrant CAMKK2 inhibitor reduces food intake in rodent model. Bioorg Med Chem Lett. 2018;28(10):1958-63.
  186. Okada S, York DA, Bray GA, Mei J, and Erlanson-Albertsson C. Differential inhibition of fat intake in two strains of rat by the peptide enterostatin. Am J Physiol. 1992;262(6 Pt 2):R1111-6.
  187. Kovacs EM, Lejeune MP, and Westerterp-Plantenga MS. The effects of enterostatin intake on food intake and energy expenditure. Br J Nutr. 2003;90(1):207-14.
  188. Doi T, Liu M, Seeley RJ, Woods SC, and Tso P. Effect of leptin on intestinal apolipoprotein AIV in response to lipid feeding. Am J Physiol Regul Integr Comp Physiol. 2001;281(3):R753-9.
  189. Attia N, Touzani A, Lahrichi M, Balafrej A, Kabbaj O, and Girard-Globa A. Response of apolipoprotein AIV and lipoproteins to glycaemic control in young people with insulin-dependent diabetes mellitus. Diabet Med. 1997;14(3):242-7.
  190. Fujimoto K, Machidori H, Iwakiri R, Yamamoto K, Fujisaki J, Sakata T, et al. Effect of intravenous administration of apolipoprotein A-IV on patterns of feeding, drinking and ambulatory activity of rats. Brain Res. 1993;608(2):233-7.
  191. Katsuura G, Asakawa A, and Inui A. Roles of pancreatic polypeptide in regulation of food intake. Peptides. 2002;23(2):323-9.
  192. Asakawa A, Inui A, Ueno N, Fujimiya M, Fujino MA, and Kasuga M. Mouse pancreatic polypeptide modulatesfood intake, while not influencing anxiety in mice. Peptides. 1999;20(12):1445-8.
  193. Batterham RL, Le Roux CW, Cohen MA, Park AJ, Ellis SM, Patterson M, et al. Pancreatic polypeptide reduces appetite and food intake in humans. J Clin Endocrinol Metab. 2003;88(8):3989-92.
  194. Jesudason DR, Monteiro MP, McGowan BM, Neary NM, Park AJ, Philippou E, et al. Low-dose pancreaticpolypeptide inhibits food intake in man. Br J Nutr. 2007;97(3):426-9.
  195. Neary NM, McGowan BM, Monteiro MP, Jesudason DR, Ghatei MA, and Bloom SR. No evidence of an additive inhibitory feeding effect following PP and PYY 3-36 administration. Int J Obes (Lond). 2008;32(9):1438-40.
  196. Lutz TA, Althaus J, Rossi R, and Scharrer E. Anorectic effect of amylin is not transmitted by capsaicin-sensitive nerve fibers. Am J Physiol. 1998;274(6):R1777-82.
  197. Roth JD, Hughes H, Kendall E, Baron AD, and Anderson CM. Antiobesity effects of the beta-cell hormone amylin in diet-induced obese rats: effects on food intake, body weight, composition, energy expenditure, and gene expression. Endocrinology. 2006;147(12):5855-64.
  198. Rushing PA, Hagan MM, Seeley RJ, Lutz TA, and Woods SC. Amylin: a novel action in the brain to reduce body weight. Endocrinology. 2000;141(2):850-3.
  199. Chapman I, Parker B, Doran S, Feinle-Bisset C, Wishart J, Strobel S, et al. Effect of pramlintide on satiety and food intake in obese subjects and subjects with type 2 diabetes. Diabetologia. 2005;48(5):838-48.
  200. Aronne L, Fujioka K, Aroda V, Chen K, Halseth A, Kesty NC, et al. Progressive reduction in body weight after treatment with the amylin analog pramlintide in obese subjects: a phase 2, randomized, placebo-controlled, dose-escalation study. J Clin Endocrinol Metab. 2007;92(8):2977-83.
  201. Roth JD, Roland BL, Cole RL, Trevaskis JL, Weyer C, Koda JE, et al. Leptin responsiveness restored by amylinagonism in diet-induced obesity: evidence from nonclinical and clinical studies. Proc Natl Acad Sci U S A. 2008;105(20):7257-62.
  202. Trevaskis JL, Coffey T, Cole R, Lei C, Wittmer C, Walsh B, et al. Amylin-mediated restoration of leptinresponsiveness in diet-induced obesity: magnitude and mechanisms. Endocrinology. 2008;149(11):5679-87.
  203. Aronne LJ, Halseth AE, Burns CM, Miller S, and Shen LZ. Enhanced weight loss following coadministration ofpramlintide with sibutramine or phentermine in a multicenter trial. Obesity (Silver Spring). 2010;18(9):1739-46.
  204. Gydesen S, Andreassen KV, Hjuler ST, Christensen JM, Karsdal MA, and Henriksen K. KBP-088, a novel DACRA with prolonged receptor activation, is superior to davalintide in terms of efficacy on body weight. Am J Physiol Endocrinol Metab. 2016;310(10):E821-7.
  205. Hjuler ST, Andreassen KV, Gydesen S, Karsdal MA, and Henriksen K. KBP-042 improves bodyweight and glucose homeostasis with indices of increased insulin sensitivity irrespective of route of administration. Eur J Pharmacol. 2015;762:229-38.
  206. Long-acting amylin analog (obesity / diabetes). https://www.zealandpharma.com/longacting-amylin-analog
  207. Drent ML, Larsson I, William-Olsson T, Quaade F, Czubayko F, von Bergmann K, et al. Orlistat (Ro 18-0647), a lipase inhibitor, in the treatment of human obesity: a multiple dose study. Int J Obes Relat Metab Disord. 1995;19(4):221-6
  208. Kopelman P, Bryson A, Hickling R, Rissanen A, Rossner S, Toubro S, et al. Cetilistat (ATL-962), a novel lipase inhibitor: a 12-week randomized, placebo-controlled study of weight reduction in obese patients. Int J Obes (Lond). 2007;31(3):494-9.
  209. Bryson A, de la Motte S, and Dunk C. Reduction of dietary fat absorption by the novel gastrointestinal lipase inhibitor cetilistat in healthy volunteers. Br J Clin Pharmacol. 2009;67(3):309-15.
  210. Kopelman P, Groot Gde H, Rissanen A, Rossner S, Toubro S, Palmer R, et al. Weight loss, HbA1c reduction, and tolerability of cetilistat in a randomized, placebo-controlled phase 2 trial in obese diabetics: comparison with orlistat (Xenical). Obesity (Silver Spring). 2010;18(1):108-15.
  211. Nelson CN, List EO, Ieremia M, Constantin L, Chhabra Y, Kopchick JJ, et al. Growth hormone activated STAT5is required for induction of beige fat in vivo. Growth Horm IGF Res. 2018;42-43:40-51.
  212. Scacchi M, Pincelli AI, and Cavagnini F. Growth hormone in obesity. Int J Obes Relat Metab Disord. 1999;23(3):260-71.
  213. Shadid S, and Jensen MD. Effects of growth hormone administration in human obesity. Obes Res. 2003;11(2):170-5.
  214. Valentino MA, Lin JE, and Waldman SA. Central and peripheral molecular targets for antiobesity pharmacotherapy. Clin Pharmacol Ther. 2010;87(6):652-62.
  215. Mathvink RJ, Tolman JS, Chitty D, Candelore MR, Cascieri MA, Colwell LF, Jr., et al. Discovery of a potent, orally bioavailable beta(3) adrenergic receptor agonist, (R)-N-[4-[2-[[2-hydroxy-2-(3-pyridinyl)ethyl]amino]ethyl]phenyl]-4-[4 -[4-(trifluoromethyl)phenyl]thiazol-2-yl]benzenesulfonamide. J Med Chem. 2000;43(21):3832-6.
  216. van Baak MA, Hul GB, Toubro S, Astrup A, Gottesdiener KM, DeSmet M, et al. Acute effect of L-796568, a novel beta 3-adrenergic receptor agonist, on energy expenditure in obese men. Clin Pharmacol Ther. 2002;71(4):272-9.
  217. Larsen TM, Toubro S, van Baak MA, Gottesdiener KM, Larson P, Saris WH, et al. Effect of a 28-d treatment with L-796568, a novel beta(3)-adrenergic receptor agonists, on energy expenditure and body composition in obese men. Am J Clin Nutr. 2002;76(4):780-8.
  218. Bujalska IJ, Kumar S, and Stewart PM. Does central obesity reflect "Cushing's disease of the omentum"? Lancet. 1997;349(9060):1210-3.
  219. Sandeep TC, Andrew R, Homer NZ, Andrews RC, Smith K, and Walker BR. Increased in vivo regeneration of cortisol in adipose tissue in human obesity and effects of the 11beta-hydroxysteroid dehydrogenase type 1 inhibitor carbenoxolone. Diabetes. 2005;54(3):872-9.
  220. Liu J, Wang L, Zhang A, Di W, Zhang X, Wu L, et al. Adipose tissue-targeted 11beta-hydroxysteroid dehydrogenase type 1 inhibitor protects against diet-induced obesity. Endocr J. 2011;58(3):199-209.
  221. Anil TM, Dandu A, Harsha K, Singh J, Shree N, Kumar VS, et al. A novel 11beta-hydroxysteroid dehydrogenase type1 inhibitor CNX-010-49 improves hyperglycemia, lipid profile and reduces body weight in diet induced obese C57B6/J mice with a potential to provide cardio protective benefits. BMC Pharmacol Toxicol. 2014;15:43.
  222. Kim YM, An JJ, Jin YJ, Rhee Y, Cha BS, Lee HC, et al. Assessment of the anti-obesity effects of the TNP-470 analog, CKD-732. J Mol Endocrinol. 2007;38(4):455-65.
  223. 223. The Company is developing an anti-abdominal obesity botanical drug, ALS-L1023, which is in phase III clinical trial in Korea. The company has already commercialized anti-abdominal obesity dietary supplement (Ob-X). https://www.pharmalicensing.com/detail.php?uid=64551.
  224. Bluher M, Kahn BB, and Kahn CR. Extended longevity in mice lacking the insulin receptor in adipose tissue. Science. 2003;299(5606):572-4.
  225. Picard F, Kurtev M, Chung N, Topark-Ngarm A, Senawong T, Machado De Oliveira R, et al. Sirt1 promotes fat mobilization in white adipocytes by repressing PPAR-gamma. Nature. 2004;429(6993):771-6.
  226. Lagouge M, Argmann C, Gerhart-Hines Z, Meziane H, Lerin C, Daussin F, et al. Resveratrol improves mitochondrial function and protects against metabolic disease by activating SIRT1 and PGC-1alpha. Cell. 2006;127(6):1109-22.
  227. Feige JN, Lagouge M, Canto C, Strehle A, Houten SM, Milne JC, et al. Specific SIRT1 activation mimics low energy levels and protects against diet-induced metabolic disorders by enhancing fat oxidation. Cell Metab. 2008;8(5):347-58.
  228. Huynh FK, Hershberger KA, and Hirschey MD. Targeting sirtuins for the treatment of diabetes. Diabetes Manag (Lond). 2013;3(3):245-57.
  229. Hardie DG, and Frenguelli BG. A neural protection racket: AMPK and the GABA(B) receptor. Neuron. 2007;53(2):159-62.
  230. Narkar VA, Downes M, Yu RT, Embler E, Wang YX, Banayo E, et al. AMPK and PPARdelta agonists are exercise mimetics. Cell. 2008;134(3):405-15.
  231. Minokoshi Y, Alquier T, Furukawa N, Kim YB, Lee A, Xue B, et al. AMP-kinase regulates food intake by responding to hormonal and nutrient signals in the hypothalamus. Nature. 2004;428(6982):569-74.
  232. Li S, Li Y, Xiang L, Dong J, Liu M, and Xiang G. Sildenafil induces browning of subcutaneous white adipose tissue in overweight adults. Metabolism. 2018;78:106-17.
  233. Chun E, Han CK, Yoon JH, Sim TB, Kim YK, and Lee KY. Novel inhibitors targeted to methionine aminopeptidase 2 (MetAP2) strongly inhibit the growth of cancers in xenografted nude model. Int J Cancer. 2005;114(1):124-30.
  234. Kim DD, Krishnarajah J, Lillioja S, de Looze F, Marjason J, Proietto J, et al. Efficacy and safety of beloranib for weight loss in obese adults: a randomized controlled trial. Diabetes Obes Metab. 2015;17(6):566-72.
  235. Elfers CT, and Roth CL. Robust Reductions of Excess Weight and Hyperphagia by Beloranib in Rat Models of Genetic and Hypothalamic Obesity. Endocrinology. 2017;158(1):41-55.
  236. McCandless SE, Yanovski JA, Miller J, Fu C, Bird LM, Salehi P, et al. Effects of MetAP2 inhibition on hyperphagia and body weight in Prader-Willi syndrome: A randomized, double-blind, placebo-controlled trial. Diabetes, obesity & metabolism. 2017;19(12):1751-61.
  237. Giralt M, Gavalda-Navarro A, and Villarroya F. Fibroblast growth factor-21, energy balance and obesity. Mol Cell Endocrinol. 2015;418 Pt 1:66-73.
  238. Fisher FM, and Maratos-Flier E. Understanding the Physiology of FGF21. Annu Rev Physiol. 2016;78:223-41.
  239. Gomez-Samano MA, Grajales-Gomez M, Zuarth-Vazquez JM, Navarro-Flores MF, Martinez-Saavedra M, Juarez-Leon OA, et al. Fibroblast growth factor 21 and its novel association with oxidative stress. Redox Biol. 2017;11:335-41.
  240. Fisher FM, Chui PC, Antonellis PJ, Bina HA, Kharitonenkov A, Flier JS, et al. Obesity is a fibroblast growth factor 21 (FGF21)-resistant state. Diabetes. 2010;59(11):2781-9.
  241. Flint HJ, Scott KP, Louis P, and Duncan SH. The role of the gut microbiota in nutrition and health. Nat Rev Gastroenterol Hepatol. 2012;9(10):577-89.
  242. Rosenbaum M, Knight R, and Leibel RL. The gut microbiota in human energy homeostasis and obesity. Trends Endocrinol Metab. 2015;26(9):493-501.
  243. Rooks MG, and Garrett WS. Gut microbiota, metabolites and host immunity. Nat Rev Immunol. 2016;16(6):341-52.
  244. Christiansen CB, Gabe MBN, Svendsen B, Dragsted LO, Rosenkilde MM, and Holst JJ. The impact of short-chain fatty acids on GLP-1 and PYY secretion from the isolated perfused rat colon. Am J Physiol Gastrointest Liver Physiol. 2018;315(1):G53-g65.
  245. De Vadder F, Kovatcheva-Datchary P, Goncalves D, Vinera J, Zitoun C, Duchampt A, et al. Microbiota-generated metabolites promote metabolic benefits via gut-brain neural circuits. Cell. 2014;156(1-2):84-96.
  246. Cotillard A, Kennedy SP, Kong LC, Prifti E, Pons N, Le Chatelier E, et al. Dietary intervention impact on gut microbial gene richness. Nature. 2013;500(7464):585-8.
  247. Cani PD, Possemiers S, Van de Wiele T, Guiot Y, Everard A, Rottier O, et al. Changes in gut microbiota control inflammation in obese mice through a mechanism involving GLP-2-driven improvement of gut permeability. Gut. 2009;58(8):1091-103.
  248. de Aguiar Vallim TQ, Tarling EJ, and Edwards PA. Pleiotropic roles of bile acids in metabolism. Cell Metab. 2013;17(5):657-69.
  249. Stefanaki C, Peppa M, Mastorakos G, and Chrousos GP. Examining the gut bacteriome, virome, and mycobiome in glucose metabolism disorders: Are we on the right track? Metabolism. 2017;73:52-66.
  250. Altabas V, and Zjacic-Rotkvic V. Anti-ghrelin antibodies in appetite suppression: recent advances in obesity pharmacotherapy. Immunotargets Ther. 2015;4:123-30.
  251. Colon-Gonzalez F, Kim GW, Lin JE, Valentino MA, and Waldman SA. Obesity pharmacotherapy: what is next? Mol Aspects Med. 2013;34(1):71-83.
  252. Takagi K, Legrand R, Asakawa A, Amitani H, Francois M, Tennoune N, et al. Anti-ghrelin immunoglobulins modulate ghrelin stability and its orexigenic effect in obese mice and humans. Nat Commun. 2013;4:2685.
  253. Monteiro MP. Obesity vaccines. Hum Vaccin Immunother. 2014;10(4):887-95.
  254. Haffer KN. Effects of novel vaccines on weight loss in diet-induced-obese (DIO) mice.J Anim Sci Biotechnol. 2012;3(1):21.
  255. Yamada T, Hara K, and Kadowaki T. Association of adenovirus 36 infection with obesity and metabolic markers in humans: a meta-analysis of observational studies. PLoS One. 2012;7(7):e42031.
  256. Na HN, and Nam JH. Proof-of-concept for a virus-induced obesity vaccine; vaccination against the obesity agent adenovirus 36. Int J Obes (Lond). 2014;38(11):1470-4.
  257. Zhang Y, Yu J, Qiang L, and Gu Z. Nanomedicine for obesity treatment. Sci China Life Sci. 2018;61(4):373-9.
  258. Sangwai M, Sardar S, and Vavia P. Nanoemulsified orlistat-embedded multi-unit pellet system (MUPS) with improved dissolution and pancreatic lipase inhibition. Pharm Dev Technol. 2014;19(1):31-41.
  259. Chen YL, Zhu S, Zhang L, Feng PJ, Yao XK, Qian CG, et al. Smart conjugated polymer nanocarrier for healthy weight loss by negative feedback regulation of lipase activity. Nanoscale. 2016;8(6):3368-75.
  260. Kupferschmidt N, Csikasz RI, Ballell L, Bengtsson T, and Garcia-Bennett AE. Large pore mesoporous silica induced weight loss in obese mice. Nanomedicine (Lond). 2014;9(9):1353-62.
  261. Mun EC, Blackburn GL, and Matthews JB. Current status of medical and surgical therapy for obesity. Gastroenterology. 2001;120(3):669-81.
  262. Xue Y, Xu X, Zhang XQ, Farokhzad OC, and Langer R. Preventing diet-induced obesity in mice by adipose tissue transformation and angiogenesis using targeted nanoparticles. Proc Natl Acad Sci U S A. 2016;113(20):5552-7.
  263. Di Mascolo D, C JL, Aryal S, Ramirez MR, Wang J, Candeloro P, et al. Rosiglitazone-loaded nanospheres for modulating macrophage-specific inflammation in obesity. J Control Release. 2013;170(3):460-8.
  264. Marrache S, and Dhar S. Engineering of blended nanoparticle platform for delivery of mitochondria-acting therapeutics. Proceedings of the National Academy of Sciences of the United States of America. 2012;109(40):16288-93.
  265. Jiang C, Cano-Vega MA, Yue F, Kuang L, Narayanan N, Uzunalli G, et al. Dibenzazepine-Loaded Nanoparticles Induce Local Browning of White Adipose Tissue to Counteract Obesity. Mol Ther. 2017;25(7):1718-29.
  266. Yu J, Zhang Y, Ye Y, DiSanto R, Sun W, Ranson D, et al. Microneedle-array patches loaded with hypoxia-sensitive vesicles provide fast glucose-responsive insulin delivery. Proc Natl Acad Sci U S A. 2015;112(27):8260-5.

Subcutaneous Adipose Tissue Diseases: Dercum Disease, Lipedema, Familial Multiple Lipomatosis and Madelung Disease

ABSTRACT

 

Subcutaneous adipose tissue diseases involving adipose tissue and its fascia, also known as adipofascial disorders, represent variations in the spectrum of obesity. The adipofascia diseases discussed in this chapter can be localized or generalized and include a common disorder primarily affecting women, lipedema, and four rare diseases, familial multiple lipomatosis, angiolipomatosis, Dercum disease, and multiple symmetric lipomatosis. The fat in adipofascial disorders is difficult to lose by standard weight loss approaches, including lifestyle (diet, exercise), pharmacologic therapy, and even bariatric surgery, due in part to tissue fibrosis. In the management of obesity, healthcare providers should be aware of this difficulty and be able to provide appropriate counseling and care of these conditions. Endocrinologists and primary care providers alike will encounter these conditions and should consider their occurrence during workup for bariatric surgery or hypothyroidism (lipedema) and in those that manifest, or are referred for, dyslipidemia or diabetes (Dercum disease). People with angiolipomas should be worked up for Cowden’s disease where a mutation in the gene PTEN increases their risk for thyroid and breast cancer. This chapter provides details on the pathophysiology, prevalence, genetics and treatments for these adipofascial disorders along with recommendations for the care of people with these diseases. 

 

INTRODUCTION

 

People with subcutaneous adipose tissue (SAT) diseases have fat within this compartment that grows abnormally in amount or structure, often causing pain and other discomfort.  Subcutaneous adipose tissue is loose connective tissue, or adipofascia, which is the most common type of connective tissue in vertebrates. The focus of this chapter is on abnormal SAT that has within it changes in blood vessels, lymphatic vessels, immune cells, mesenchymal stem cells, fascia, interstitial matrix organ, or other components that make up loose connective tissue. 

 

The SAT diseases discussed here include lipedema, which commonly occurs in women, and four rare adipose tissue diseases (RAD): familial multiple lipomatosis, angiolipomatosis, Dercum disease, and Madelung disease (1). Adipose tissue in SAT diseases is resistant to loss by usual measures including extreme dietary changes (both hypocaloric and in macronutrient content) and exercise. Because of this, it is often referred to persistent fat tissue. People with diabetes and/or obesity may have a mixture of normal and persistent fat, making the understanding of SAT diseases by clinicians important in the care of these patients. Persistent fat may also be found in conditions where adipose tissue proliferation occurs, such as during infection, in autoimmune diseases, in those with hypermobile joint disorders, or with exposure to environmental toxins. Information on subcutaneous adipose tissue diseases not discussed here can be found in recent reviews and other Endotext chapters, including those covering lipodystrophies (2-4), cellulite (5), obesity (6,7), and other fat depots such visceral fat (abdominal, perirenal, pericardial), and perivascular fat (8).

 

Along with the gut (9), subcutaneous adipose tissue is thought to be one of the largest endocrine organs in the body (10). Subcutaneous adipose tissue houses immune cells including monocytes/macrophages, mast cells, and lymphocytes, which produce some of the hormones secreted by fat tissue (11,12). Greater amounts of fat and immune cells result in an inflammatory process that can lead to insulin resistance and slow intrinsic pumping of lymphatic vessels, which, in turn, may prolong inflammation in this tissue (13).   

 

Patients, most often women with swelling, have slow blood flow in, and lymph flow out of, depots of increased fat on the abdomen (14,15) or the gynoid area (hips, thighs and buttocks) (16). Poor blood and lymph flow through fat tissue results in accumulation of fluid, cell waste material, proteins, cells and other metabolic products in the extracellular matrix (ECM) around adipocytes and other components of adipofascia, resulting in a hypoxic environment, especially in adipocytes furthest from their nutrient and oxygen sources. These adipocytes then send signals that recruit more immune cells, resulting in a state of sustained inflammation and tissue degradation. Connective tissue then replaces degraded tissue in a process called fibroplasia or fibrosis. When tissue ischemia occurs or ECM accumulation outpaces its degradation, fibrosis may become unchecked and lead to widespread pathological remodeling of the ECM culminating in permanent scar tissue that completely inhibits flow through the adipose tissue (17).

 

Obesity is a main cause of densification of fascia and fibrosis development in loose connective tissue (18). The result is a fibrotic mesh around adipocytes and fat lobules which has been well described (19). This sick fat, or adisopathy, increases the risk of metabolic disease (20). In addition, more fibrotic adipose tissue is less responsive to mobilization and reduction attempts through diet, exercise, use of weight loss medications, or bariatric surgery (19,21,22). All the SAT disorders in this chapter have a component of fibrosis in the tissue that prevents loss by usual measures. An important goal is to determine why the loose connective tissue in SAT diseases becomes fibrotic and adisopathic to prevent its occurrence and progression and treat when identified.

 

LIPEDEMA

 

Lipedema is a common SAT disease that was first described in 1940 at the Mayo clinic by Drs. Allen and Hines (23). A second seminal paper in 1951 provided a description of lipedema that is still commonly used for clinical diagnosis. Lipedema occurs almost exclusively in women but has been reported rarely in men (24-26).

 

Lipedema fat is located just under the skin on the limbs including upper arms, hips, buttocks, thighs, lower legs, generally sparing the trunk and feet. It feels nodular when palpated, may be painful to touch, and often has prominent superficial veins. Lipedema tissue can be found under the umbilicus and in some woman, a deeper nodular adipofascia is found in the lateral abdomen. This inner nodularity may reflect changes in the ECM that may be a precursor to lipedema fat if abdominal obesity develops. Disease defining key questions and physical examination characteristics can help to establish the diagnosis of lipedema (Figure 1).

Figure 1. Characteristics of lipedema that aid in establishment of diagnosis are listed, and many can be seen in the accompanying photo. This patient was diagnosed with Stage 3, type III and IV lipedema without lymphedema (see below). A quick and easy set of questions and exam findings are included to help in diagnosis of women with lipedema.

Classification of Lipedema

 

Lipedema is classified by stages and types (27). In Stage 1 (Figure 2) the skin surface is normal over an enlarged hypodermis that already has palpable pea-sized nodules in the fat. These pea-sized nodules represent enlargement of and fibrosis in the ECM and in the connective tissue surrounding the fat lobule. Stage 2 skin is uneven with indentations (like cellulite) representing thickening and contraction of underlying connective tissue fibers over increased fat with small to larger hypodermal masses. Lobular extrusions of skin, fat and fascia tissue in Stage 3 represent significant loss of elasticity in the adipofascia drastically inhibiting mobility; blood flow in and lymph flow out of the lobules is reduced resulting in inflammation followed by fibrosis; it is in this stage that fibrosis in the tissue becomes prominent and clearly palpable; fibrosis may or may not affect the skin and the skin may develop thinning and a looser connection to the underlying hypodermis (Figure 1). Modifications of diagnostic criteria for lipedema have been suggested (28).

 

Lymphedema can occur at any stage, but is more often found in women with Stage 3 lipedema when it is often called lipo-lymphedema or Stage 4 (26). Rather than use this combined term, however, it is preferable to identify the lipemia stage and state whether lymphedema is also present or not. Lymphedema can be identified in women with lipedema by visible swelling of the hands or feet, pitting edema, asymmetry between the size of one limb versus the other, and/or a positive Stemmer sign. A positive Stemmer sign occurs when edema in the limb makes it difficult to pinch skin on the great toe, top of the foot, finger, or dorsal hand. A negative Stemmer sign occurs when only skin is pinched. Other differences between lipedema and lymphedema include symmetry (lipedema tissue occurs symmetrically versus lymphedema, which is usually unilateral), sparing of the hands and feet in lipedema, and report by patients of pain in lipedematous but not lymphedematous tissue.

Figure 2. Three stages of legs of women with lipedema with subcategories of types. In Stage 1, the skin is smooth, and the legs can appear normal but there is pain, easy bruising and a nodular feel to the fat tissue. In Stage 2, the lipedema fat exhibits a mattress-like pattern indicating fibrosis under the skin that tethers on the skin that can be found on the upper legs (Type II) or extend down to the ankles (Type III). In lipedema Stage 3, there are folds of tissue and the lipedema fat usually extends down to the ankles. For description of the types of lipedema, see Figure 3.

The type of lipedema is defined by its anatomical location (29). Women with Type I lipedema have affected fat on the pelvis, buttocks and hips (saddle bag phenomenon). Women with Type II have affected fat from the buttocks to knees with formation of a tender deposits of fat around the inner side of and below the knee. Women with Type III lipedema have affected fat from the buttocks to ankles where a prominent “cuff,” or ledge, of fat tissue develops. Women with Type IV lipedema fat have affected arms and women with Type V lipedema, a rare type, have only affected lower legs. The most common phenotype of women with lipedema are combinations of II and IV or III and IV (Figure 3).

Figure 3. Types of lipedema fat. Lipedema fat may be located from the umbilicus down to the bottom of the hips (Type I), down to the medial knees usually including a pad of fat on the inner knee and below the knee (Type II), and down to the ankle (Type III) where a “cuff” of fat develops but spares the dorsal foot. Rarely only the lower legs are affected (Type V). Lipedema affecting the arms alone is rare (Type IV) and, instead, usually is found in combination with Type II or III lipedema. The arms can be variably affected with nodular lipedema fat around the cubit nodes, over the brachioradialis, down the medial arm to the wrist in line with the thumb or 5th digit, the entire lower arm, or the entire arm.

Prevalence of Lipedema

 

From one specialty lymphedema/obesity clinic in Germany, the prevalence of lipedema in women was estimated to be 11%. Estimates from similar clinics reported prevalence rates of lipedema ranging from 15 to 18.8% (30,31). The prevalence of lipedema in children in the United States in a vascular clinic was reported to be 6.5% (32). Examination of 62 women outside of clinics in Germany found a prevalence rate for all types of lipedema of 39% (33).  Using the lowest prevalence estimate in adults of 11%, over 16 million women in the US may be affected with lipedema.  

 

Genetics of Lipedema

 

The gene or genes underlying lipedema are not known, but another affected immediate family member has been reported in up to 60% of women (34-37). This is compatible with an autosomal dominant inheritance with incomplete penetrance (38) in which an affected parent has a 50% chance of passing lipedema to their child. Supportive of a genetic component, lipedema has been reported in children (32) and as early as infancy by some women. The phenotypic expression of lipedema may vary amongst affected females, especially if there is also obesity in the family. Males that carry the putative gene do not generally exhibit the phenotype, even the fathers of affected daughters. 

 

A mutation in the POU1F1/PIT-1 gene has been shown to cause multiple anterior pituitary deficiencies including thyroxine and growth hormone (GH) deficiency. A PIT-1 mutation was associated with the phenotypic presentation of lipedema in a short mother but not her short son who also carried the mutation, and not in her normal height daughter who did not carry the mutation (39). In mice with GH receptor (GHR) antagonism or lacking GH function, subcutaneous adipose tissue is increased more than other fat depots similar to lipedema in humans (40). No other cases of women with lipedema and a mutation in PIT-1 have been reported to date and women with lipedema are not known to be more likely to have short stature. Classic features of lipedema can be found in people with Williams syndrome caused by a chromosomal 7q11.23 microdeletion of ~1.6 million base pairs, which includes the elastin gene ELN (41). Loss of elasticity results in the reduction of the ability of tissue to contract back after being stretched. Changes in elasticity may therefore allow more fat to grow.  These reports suggest there may be more than one genetic mutation causing expression of the lipedema phenotype. Additional genes that may be important in the development of lipedema have been reviewed (42).

 

Pathophysiology of Lipedema

 

The cause of lipedema remains unknown. Given the predominance of occurrence in women, it is tempting to consider sex steroids, especially estrogen, as major triggers or contributors of this condition. That knee laxity in women peaks when estrogen levels decline between ovulation and post-ovulation is indictive of estrogen’s effect on connective tissue (43). Other observations that support this effect include reports that lipedema is often first noticed at the time of puberty in young girls and occasionally following pregnancy, when multiple hormone levels are high, including estrogen. Although data in men is sparse, those reported to have lipedema tend to have low testosterone or liver disease, both of which are associated with a relative increase in estrogen levels and therefore a higher estrogen to testosterone ratio (21). While higher levels of testosterone in women with polycystic ovarian syndrome are not protective against lipedema, the adipose tissue in women with this condition may be predisposed to lipedema due to abdominal obesity and inflammation associated with pre-diabetes or diabetes. A causative role for estrogen in the expression of lipedema remains speculative until well-controlled studies are conducted that quantify sex hormone levels, sex hormone receptors, tissue effects, metabolism or sex hormone driven pathways in men and women with lipedema. 

 

PROPOSED THEORIES TO EXPLAIN LIPEDEMA

 

Theory 1: Abnormal Blood Microvasculature

 

We and others (36,44,45) have advanced the theory that increased compliance from structural changes in connective tissue results in the ability to hold on to fluids, proteins and other constituents within the ECM and is causally important in the development of lipedema. As in lymphedema, changes in lipedema tissue may occur globally but are likely to also be found regionally in the same limb (46). Over 50% of women with lipedema are thought to have some kind of joint hypermobility consistent with a connective tissue disorder (25). Most women with lipedema and hypermobility fall into the Ehlers Danlos spectrum of disorders, the gene for which is not known (47,48). This hypothesis is consistent with loss of elasticity in tissue resulting in classic features of lipedema in Williams syndrome. Structures in loose connective tissue that contain elastin include blood vessels, lymph nodes, and connective tissue fascia that helps loose connective tissue hold its shape. Initial lymphatic vessels do not have elastic fibers, but elastic fibers support lymphatic vessels to open and close in response to pressure changes in the tissue; loss of elasticity could reduce the ability of lymphatic vessels to open with increased pressure in the ECM. Capillaries do not have elastic fibers but the loose connective tissue surrounding them does; as loose connective tissue enlarges due to growth of adipocytes, capillaries are at risk for dilation and distortion. Dilated and/or distorted capillaries may release their contents into tissues at a higher rate and/or amount in lipedema which initially, lymphatic vessels promptly pump out. Over time, due to compliance in fascia surrounding cells, malfunction of lymphatic vessels, and increased infiltrate leaving altered capillaries fill the ECM, with protein dense and salt-rich (49) deposits that accumulate in the interstitial space clogging flow through the tissue resulting in hypoxia. Hypoxia results in the secretion of hypoxia inducible factor (HIF)-1 by local adipocytes, which stimulates hypoxia response elements on a number of genes including the vascular endothelial growth factor (VEGF) gene and inducing proliferation of stem cells in the adipose tissue (50). Levels of VEGF have been shown to be elevated in women with lipedema (n=9) compared to women without lipedema (n=5) (51), supporting an underlying role for hypoxia in the pathogenesis of lipedema. 

 

Lymph fluid induces adipogenesis when added to adipocytes (52). Therefore, accumulation of pre-lymph fluid in the ECM may be a stimulating factor in adipogenesis. Mesenchymal stem cells isolated from lipedema stromal vascular fraction (SVF), a  heterogeneous collection of cells surrounding adipocytes within adipose tissue, contained the connective tissue cell marker CD90+ and endothelial/pericytic marker CD146+ (53). With ~50% of cells in the SVF expressing +CD146+, perivascular cells (pericytes) play a physiological role in vascular development and homeostasis (52). The presence of such high numbers of pericytes is consistent with a chronic capillary injury in lipedema leading to increased need for repair and increased protection of vessels.

 

As overworked lymphatic vessels eventually lose function, microaneurysms form in the lymphatic vessels in lipedemadous tissue, becoming high risk for breakage and leakage analogous to what happens in lymphedema (54,55). Adipokines, especially large adipokines such as leptin and monocyte chemoattractant protein (MCP)-1, become secreted primarily into the lymphatic system rather than blood capillaries (56). It would then follow that adipokine flow out of lipedema loose connective tissue would be diminished and systemic levels lower (e.g., leptin levels), leading to larger quantities of adipose tissue necessary to achieve weight homeostasis. 

 

Other potential contributors to expression of lipedema include release of lipids from leaky lymphatic vessels in the gut and tissue level (57), which could mediate induction of adipogenesis and have an important role in the development of the loose connective tissue in lipedema (58). Finally, inflammation in response to hypoxia or injuries could facilitate the development of fibrosis within loose connective tissue, not only reducing flow through the tissue further, but also impeding fat loss during weight reduction (19,21,22).

 

Theory 2: Abnormal Lymphatic Vasculature

 

Another theory posits that fluid accumulation in the ECM results from a primary defect in lymphatic vessels. Such accumulation enhances permeability issues in surrounding blood microvessels (59). In support, of this hypothesis, one study of women with lipedema and obesity noted a mismatch in the number of lymphatic vessels and the increased numbers of blood vessels in affected tissue (60). Instead there was an increase in the size (area) and area/perimeter ratio of the lymphatic vessels. Increased angiogenesis but fewer numbers and dilated lymphatic vessels has also been reported in a diet-induced obesity model in mice (61).  In another supportive study of lipedemadous tissue free of lymphedema, an expansion in the size of lymphatic vessels but no significant changes in transport in of lymphatic fluid was reported (62). However, against this hypothesis of a primary defect in lymphatic tissue as the proximal cause of lipedema is that lymphatic vessel function as determined by lymphangioscintigraphy appears normal in many women with early stages of lipedema and only later can reductions in lymphatic flow rate or function be detected in many women with late stage lipedema (54,63,64).  

 

Markers of Obesity, Cardiometabolic Health, and Aortic Disease in Women with Lipedema

 

Hypertrophic adipocytes, a marker of an inflammatory environment at risk for insulin resistance and other metabolic dysfunction, are reported in loose connective tissue in lipedema from women regardless of whether they were obese or not (58,65). Adipogenesis has also been identified in lipedema loose connective tissue (58) as has hypertrophy and hyperplasia of adipocytes in people who developed obesity after lifestyle changes. Unhealthy hypertrophic adipocytes undergo necrosis and become surrounded by macrophages that phagocytize the dead adipocytes forming crown like structures on histological exams of tissue. Crown-like structures have also been found in the loose connective tissue of women with lipedema (53,58). 

 

Adipose tissue stem cells collected from subcutaneous adipose tissue from people with obesity have reduced adipogenic potential and proliferative ability (66). The same reduction in adipogenic potential including a reduced capacity to produce leptin by cells in culture was found for adipose tissue removed by tumescent liposuction from women with lipedema compared to women without lipedema (67). These data support the possibility that even in early stages of lipedema when BMI is in the non-obese range, lipedema fat tissue shares characteristics of adipose tissue taken from people with obesity. Thus, even though femoral adipose tissue is known to be cardioprotective (68), this association weakens in later stages of lipedema. The later the stage, the greater the obesity and metabolic risk, including lower high density lipoprotein (HDL) cholesterol levels, higher diastolic and systolic blood pressures, higher reported history of hypertension, and higher percentages of pre-diabetes (69).  As such women with advanced lipedema should be closely monitored for these conditions as part of their ongoing care.

 

Women with lipedema are thought to have a connective tissue disease along the spectrum of hypermobile Ehlers Danlos. Transthoracic 2D echocardiography (2DE) and Doppler imaging revealed that women with Stage 2 lipedema in their early 40s with BMI ~30 kg/m2 had impaired left ventricular apical rotation and left ventricular twist compared to people with lymphedema and those without either disease (70). Another paper by the same group used 2DE and Doppler imaging demonstrated enlarged ascending aortic systolic and diastolic diameters resulting in aortic stiffness in women with lipedema compared to controls (44). Individuals with Williams syndrome with loss of elasticity and features of lipedema also have aortic stiffness (71). These cardiac changes may reflect an underlying connective tissue disorder in women with lipedema and the possible need for cardiovascular screening even if lipid levels and other markers of metabolic syndrome are normal.

 

Imaging of Lipedema

There are currently no imaging exams that can be used to definitively differentiate lipedema fat from non-lipedemadous adipose tissue.  However, some imaging studies may be useful.  Nuclear medicine lymphangioscintigraphy (NM LAS) may be helpful in differentiating the presence of lymphedema in patients with lipedema. Flow of Technetium-99m-sulfur colloid injected dermally and taken up by the lymphatic vessels starting at the toe or finger webbing can be normal in lipedema (72), or the lymphatics can be tortuous especially below the knee (73). Other authors found slower lymphatic flow and a marked asymmetry of the lymphatic system in women with lipedema as compared with women without lipedema (74,75).          

 

Dual energy X-ray absorptiometry scans (DEXA) can be used for assessing whole body composition including regional fat mass and lean body mass in addition to bone mineral density; some scanners also estimate visceral fat mass. One study suggested that DEXA can be used to strengthen the confirmation of a diagnosis of lipedema in women, differentiating them from women without lipedema by a cutoff value of 0.46 for fat mass in the legs (kg) adjusted for BMI (76). Even though many women with lipedema also have obesity, the authors assert this cutoff value allows for a separation of lipedema of the legs from women without lipedema regardless of obesity.

 

Ultrasound of lipedema tissue compared to control tissue or tissue from women with lymphedema demonstrates thinner skin in agreement with previous data (77), and increased thickness and hypoechogenicity of the subcutaneous fat throughout the lower limb suggesting a diffuse increase in aqueous material (78). The hypoechogenicity was most significant in the distal extremity (medial calf) and may provide support to a clinical diagnosis when found.  Another group found no difference between ultrasonographic features of women with and without lipedema including compressibility and echogenicity (79). The control women were reported to have obesity or lipohypertrophy, an enlargement of the legs that is phenotypically similar to lipedema but painless. The definition of lipohypertrophy is unclear in the literature where authors have stated that symptoms of lipohypertrophy resolve with elevation suggesting a fluid component associated with the fat tissue (80). In personal communication with the authors, they state the relief of symptoms is due to lowering of pressure on the venous system suggesting that venous disease is important in the diagnosis of lipohypertrophy. Other authors state that lipohypertrophy is a precursor to lipedema which may explain why the latter ultrasound data showed no differences. Clearly, better means of distinguishing lipedema from those with larger legs but no lipedema is needed.

 

Finally, widening of lymphatic vessels up to 2 mm has been found by magnetic resonance imaging (MRI) of the legs of women with lipedema; women with lipedema and lymphedema had lymphatic vessel enlargement >3 mm (81). If this dilation of lymphatic vessels is consistent with lymphostatic decompensation (failure of lymphatic vessel function) in lipedema as the authors suggest, then salt should be found in the skin of women with lipedema as lymphatic vessels regulate Na+, Cl– and water in the skin, where reduced lymphatic vessel numbers are paralleled by increased blood pressure (82). Indeed Crescenzi et al. found increased salt in the skin and loose connective tissue of women with lipedema compared to women without lipedema, even in earlier Stage 1 lipedema where women tend to not have obesity (49).

 

Conditions Associated with Lipedema

 

OBESITY

 

Women with lipedema are often are often thought of as having common obesity whether or not they meet BMI criteria for this condition. The two striking differences between women with lipedema and women with obesity are that women with lipedema often have tenderness of the affected tissue and/or easy bruising of the skin overlying the lipedema fat, which is not found in women with common obesity (Table 1). However, some women with lipedema do not have pain in their tissues.  It is unclear if women with lipedema with pain have the same disease as women without pain. Of note, women with lipedema may have no pain in their lipedema fat tissue when they are well-controlled under treatment regimens, however, they should still be considered to have lipedema. For example, women with lipedema who eat low inflammatory foods, avoiding processed starch and sugar, who exercise most days of the week and wear compression garments on their legs can have minimal to no pain. Therefore, a good history is important to identify a history of pain in the tissue that would be indicate the presence of lipedema. As described above, helpful measures to differentiate women with lipedema from women with common obesity include the disproportionate distribution of their adipose tissue between the trunk and legs, any family history of lipedema, as well as a historical inability to lose much fat from the lipedema-affected areas. On the other hand, when women with obesity and lipedema lose more substantial weight through medical or surgical interventions, they can lose some fat from the areas with lipedema, which can then leave them with rolls of excess skin along in the areas of remaining lipedema fat tissue (Figure 4). 

 

Table 1. Clinical Similarities and Differences Between Lipedema and Obesity

Sign/Symptom

Lipedema

Obesity

Sex affected

Females

Females and Males

Onset

Puberty

Any age

Increased fat

Common

Common

Gynoid disproportion

Common

Possible

Influenced by lifestyle

No

Yes

Tenderness of the tissue

Common

Absent

Easy bruising

Common

Absent

Pitting edema

Uncommon

Uncommon

Stemmer sign

Negative

Negative

Able to lose fat from the legs/hips

Minimal

Common

 

Obesity (especially abdominal/visceral obesity) and/or polycystic ovary syndrome can worsen lipedema severity. This is thought to be mediated by increases in adipokines, tumor necrosis factor alpha (TNFa) and leptin that often accompany these conditions, which are associated with venous disease (83). Venous dysfunction that can lead to leakage of fluid back into tissue due to reflux may also be an important contributor to worsening of both lipedema and lymphedema, when present (84,85). 

Figure 4. Fat due to obesity and fat due to lipedema can be intermixed on the legs. With weight loss, the obesity fat can be lost resulting in excess skin and lipedema fat tissue remaining on the legs.

LYMPHEDEMA

 

Women with lipedema are at risk for developing lymphedema, which may happens in lipedema Stage 3 > Stage 2 > Stage 1 (26). The presence of lymphatic disease or lymphedema increases the risk of cellulitis and wounds, which can be difficult to manage and disfiguring. Women with heavy limbs and swelling should be considered for manual lymphatic drainage and deeper tissue therapies such as instrument assisted soft tissue therapies (e.g., Astym therapy, Graston technique) or manual therapies (e.g., myofascial therapies or other deep tissue therapies (86)), followed by compression plus reduction of obese adipofascia to reduce the risk of developing lymphedema.

 

PSYCHOSOCIAL

 

Psychosocial issues are prominent in women with lipedema including appearance-related distress and depression (87), which can result in eating disorders (88). This is not surprising in the United States where very thin women or photos of women that have been photoshopped to accentuate the appearance of leanness are posted on the internet and on television. Imagine how a woman in today’s society might feel who developed lipedema at puberty and is told to diet and exercise by friends, family and healthcare providers to lose weight, something that she has done for years to no avail. One author polled women in Germany and found a high rate of suicide attempts in women with lipedema (89). Lower mobility associated with lipedema and obesity were also found to affect quality of life in women living with lipedema (87) and may contribute to social isolation. Prevention and management of obesity in women with lipedema becomes paramount to maintain their quality of life. High anxiety is associated with hypermobile joint disorders (90), and because hypermobile joint disorders are associated with lipedema (25), anxiety should be assessed and treated to help women living with lipedema.

 

DERCUM DISEASE (PAINFUL LIPOMAS)

 

Lipedema can be present in the same individual who also has Dercum disease (see below), and in this instance, would be considered a mixed disorder. Authors have tried to differentiate women with lipedema from those with Dercum disease by examining populations and finding that people with Dercum disease tend to have other pain disorders including higher pain scores, fibromyalgia, abdominal pain, and migraines, and more often have lipomas, cognitive dysfunction and shortness of breath; whereas women with lipedema have more often fibrotic tissue, easy bruising, hypermobile joints, venous disease and edema of the feet (25).

 

Numerous other conditions apart from those described above have been associated with lipedema (Table 2). Of note, hypothyroidism is found in 27% of women with lipedema (25,26).  In one case, a woman with lipedema was described as having lymphedema and multiple symmetric lipomatosis (91).

 

Table 2. Co-Morbidities and Complications Associated with Lipedema (60, 92-94)

Musculoskeletal

Soft Tissue

Vascular

Other

Gait disturbance

Obesity; fat deposits

Lymphedema/Idiopathic Edema

Pain

Change in posture (e.g., lordosis)

Loss of skin elasticity

Dilated Capillaries Microangiopathy

Psychological distress/anxiety

Genu valgum and arthritis of the knees

Thinning of the skin

Bruising

Shortness of breath

Ankle pronation

Lipomas

Varicose veins

Venous insufficiency

Cellulitis

Hypermobile joints (Hypermobile Ehlers Danlos?)

Cellulite; fibrosis

Cherry angiomas

Slow metabolic rate

 

Clinical Care of Women with Lipedema

 

Depending on whether the astute clinician makes the diagnosis of lipedema during the course of taking a history and physical, affected patients may more typically seek care for an associated co-morbidity. For example, women with lipedemia who have thyroid disease and/or obesity may regularly be referred by their primary care provider to an Endocrinologist. Endocrinologists should be clued in to a possible diagnosis of lipedema in women who present with difficulty losing weight from their hips, buttocks and legs, and who are convinced they have a thyroid issue, but thyroid labs are normal. In desperation, patients with lipedema may ask for a “complete set of thyroid labs” including thyroid stimulating hormone (TSH), free T4, free T3, reverse T3 and thyroid peroxidase (TPO) antibodies to ensure that there is no thyroid issue, which may cause tension during a clinical visit when a provider chooses not to order all these laboratories. Other times, women with joint hypermobility are often cared for by rheumatologists and orthopedic surgeons, and those with lipedema and venous disease are often followed by vascular surgeons, physical therapists and lymphedema specialists.

 

DIAGNOSIS

 

Once the possibility of lipedema is considered, a good medical history will include an assessment of the food eaten, patterns of exercise, and a timeline of development of lipedema signs and symptoms with special attention to hormonal transitions in women including puberty, pregnancy, or menopause. Additionally, helpful findings on history include pain and easy bruising in affected areas and a family history of similar traits in other female members. The upper arms and legs should then be examined for physical manifestations of lipedema as described in more detail below. In the author’s experience, diagnosing lipedema in a woman who presents thinking they have another condition, such as thyroid disease but with normal thyroid function tests, and providing education and treatment recommendations can be transformative for the patient’s life and greatly enhance the patient-physician relationship. 

 

The physical exam to diagnose lipedema can be performed quickly if a woman can be seen in her underwear after donning a gown.  Visual inspection to establish disproportionality between the upper and lower body fat should be done initially and include a measure of the waist and hip ratio, which is also helpful in diagnosing central obesity.  Following this, examination (both visually and by palpation of fat tissues) should be performed with special attention to characteristics described in Table 3.

 

Table 3. Examination of Subcutaneous Fat for Lipedema, With or Without Obesity

No Obesity

Head

Normal

Neck

Normal

Arms

Normal (nodular fat tissue may be found around cubital nodes)

Wrist

Normal

Hands

Normal; Stemmer sign negative (no edema)

Abdomen

Normal (nodules may be found deep laterally or under the umbilicus)

Buttocks

Increased loose connective tissue; may be nodular and heavy

Hips

Increased loose connective tissue; may be nodular and heavy

Thighs

Increased loose connective tissue; may be nodular and heavy

Medial knee

Nodular or enlarged fat pad; usually tender

Under knee

Fat pad; may be nodular

Shin

May be covered in fat making the shin hard to palpate

Lateral malleolus

May have fat pad underneath

Ankle

Cuff may be very small but present in Type III lipedema

Feet

Normal; stemmer sign negative

Skin

Bruising; livedo reticularis; may see peau d’orange with long-standing disease

With Obesity

Head

May have hair loss and increased fat

Neck

May have filling of the supraclavicular fossae

Arms

Nodular fat tissue on upper and/or lower arms and around cubital nodes; hanging fat on upper arm that may be heavy

Wrist

A cuff of fat may be present; bend the hand back to easily see the cuff

Hands

Fat may be found at the base of the thumb, between the MCP joints or over the hand

Abdomen

Increased deposit of fat above and/or below umbilicus

Buttocks

Increased loose connective tissue; may be nodular and usually heavy

Hips

Increased loose connective tissue; may be nodular and heavy

Thighs

Increased loose connective tissue; may be nodular and usually heavy

Medial knee

Nodular fat pad; usually tender

Under knee

Fat pad; may be nodular

Shin

Usually covered in fat making the shin hard to palpate

Lateral malleolus

May have fat pad underneath

Ankle

Cuff present in Type III lipedema

Feet

May have increased fat; Stemmer sign negative (no edema) when no lymphedema

Skin

Bruising; livedo reticularis; may see peau d’orange with long-standing disease

There is a wide variation in the phenotype of lipedema (see Figure 2 Types) therefore lack of one or more physical exam finding does not negative the presence of lipedema.

 

At present, lipedema does not have an International Classification of Disease (ICD)-10 code but an ICD-11 code of EF02.2 has been proposed.  In the meantime, other ICD-10 codes useful when caring for patients with lipedema are listed in Table 4.

.

Table 4. ICD-10 Codes for Clinical Visits for Patients with Lipedema

Sign/Symptom

ICD-10 Code

Lymphedema/Swelling (may be non-pitting)

I89.0

Edema unspecified

R60.9

Lipomatosis not elsewhere classified

E88.2

Chronic pain

G89.4

Venous insufficiency

I87.2

Varicose veins

I83.10

Overweight

E66.3

Other Obesity

E66.8

Obesity (ICD-10 code varies by BMI)

Z68

 

Treatments for Lipedema

 

FOOD PLANS

 

Many women with lipedema bring along family members that can attest to their healthy or minimal eating and beneficial exercise patterns as they tend not to be initially believed by healthcare providers. There is very little data on the use of diets to reduce lipedema fat.  Although poorly studied, it is generally accepted that lipedema fat is resistant to weight loss mediated through lifestyle, which compounds patients’ frustrations when weight loss expectations are not met. In the absence of specific recommendations, dietary counseling can focus on establishing healthy eating patterns for overall health improvement and weight management.

 

Food plans are important in helping manage obesity that accompanies lipedema with a minimal goal of stabilizing weight and a maximal goal of losing obesity weight. The most successful food plans are those with low processed carbohydrates including added sugars that reduce insulin levels and inflammation and, therefore, reduce adipogenesis (95); fasting between meals (no snacking) has been suggested (96). One group used a 1200 calorie diet along with complete decongestive therapy to reduce volume in the legs of women with lipedema (97), but evidence for long-term weight loss maintenance by this approach is lacking.

 

EXERCISE

 

Exercise is important for women living with lipedema as the muscle action helps pump blood and lymph fluid through the limbs. However, women with lipedema have ~67% of the normative value for quadriceps muscle strength compared to women without this condition matched for age and BMI (98). One theory is that fibrosis from the fat tissue extends into and reduces muscle function. The Dutch guidelines for lipedema recommend graded exercise programs aimed at strength training and conditioning for women with unhealthy lifestyles or physical limitations, although they recognize that the body parts affected by lipedema tend to increase in tissue volume despite activity (80). These authors also state that exercise and heat can increase swelling and pain in the lipedematous areas. Anecdotally, women with lipedema appear to benefit greatly from water exercises, which may be in part due to the compressive effect of water on the body that helps mobilize fluid and soften fibrotic tissues, as well as from water jets that may also help reduce fluid in the adipofascia. Some women with lipedema have a concern about showing their bodies in public due to the commonality of public body shaming (99). Cropped pant, swim tights and other swimwear coverings have enabled more women with lipedema to feel comfortable during public swimming.  Garments with compression are generally recommended for women with lipedema to wear during land-based exercises especially, when using Nordic poles that improve the adipofasica of the arms and legs.

 

COMPRESSION GARMENTS

 

Compression garments are usually worn on the legs with a high waist (to treat fat on the abdomen) and on the arms as needed. Compression can be lower in millimeters of mercury (mm Hg) for lipedema than for lymphedema. For example, 15-25 mm Hg or Class I 20-30 mm Hg, compared to Class II 30-40 mm Hg or Class III 40-50 mm Hg. The type of knit for a lower pressure garment can be circular knit, which means it is seamless and is knitted on a round cylinder. Circular knit garments have more stretch and are best suited for women with lipedema that have less lymphedema or swelling. They can also stretch to fit any shape and size. Flat knit garments have less stretch and therefore provide better edema control. Flat knit is recommended especially for women with lipedema who have an ankle cuff or unusual shape requiring a custom fit and usually have a seam. A durable medical equipment (DME) order can be provided to patients to take to a medical supply store if they are able to get insurance coverage for compression garments. Therapists treating patients with lipedema can provide guidance on compression wear.

 

VENOUS DISEASE

 

Venous insufficiency has been documented in 25% of women with lipedema (92,100). When pitting edema is present, venous insufficiency should be investigated in women with lipedema by a venous duplex ultrasound of the legs. These studies are performed in a vascular lab and should specify to look at the greater and lesser veins of the legs to evaluate for venous insufficiency and not just thrombus. Care should be taken to treat venous disease conservatively first as there is no data showing correction of venous insufficiency by surgical means will improve lipedema. Anecdotally in reports from women with or without lipedema, lymphedema can occur after surgical treatment of venous insufficiency of the greater saphenous vein (101).

 

BARIATRIC SURGERY

 

Women with lipedema without some upper body obesity may respond poorly to bariatric surgery with regard to weight loss (102) and often feel like failures or are mistakenly told (directly or indirectly) by their providers that it was their fault, with devastating psychological impact.  Indeed, women that lose minimal weight from their lower abdomen, hips, and legs after bariatric surgery should be examined for the presence of lipedema. Even with less-than-expected weight loss, patients with lipedema should still be considered candidates for bariatric surgery as several procedures (e.g., laparoscopic gastric bypass and sleeve gastrectomy) have shown weight-independent benefits on glucometabolic outcomes, especially prediabetes and diabetes, and cardiovascular risk. When women do lose weight and it includes a portion of their lipedema-affected regions, it often results in accentuation of the “saddle bag” look (See Figure 4) and may worsen their body image anxieties.  Optimally, women with lipedema should be identified prior to bariatric surgery, counseled on their condition and how it might influence their overall weight-loss response, and be offered complete decongestive therapy and compression garments to reduce the risk of developing lymphedema after bariatric surgery and to improve weight loss success.  In addition, pre-surgery is a good time to initiate consultation with a plastic surgeon regarding options of removal of excess skin removal once weight stability is established post-operatively (usually between 1 and 3 years).

 

LIPOSUCTION

 

Women with lipedema typically have several medically necessary reasons for undergoing liposuction to remove lipedema fat, including:

  • Loss of mobility
  • Reduced quality of life
  • Joint damage or altered gait
  • Chronic pain
  • Failure to improve signs and symptoms associated with lipedema despite conservative therapy

 

Complete decongestive therapy including manual lymphatic drainage, compression garments, a healthy eating plan, and as much activity as allowed or possible are important before liposuction to improve outcomes. Due to the increased vascularity of the lipedematous tissue and blood loss with liposuction, post-procedure anemia is not uncommon. Therefore, labs prior to surgery should include a complete blood count (CBC) with platelet level as well as coagulation labs to include activated prothrombin time (aPTT), prothrombin time (PT), thrombin time (TT), andfibrinogen.  People with normal coagulation labs and easy bruising can have hereditary and acquired platelet defects, hereditary disorders of vascular and perivascular tissues including Ehlers Danlos Syndrome, and other disorders of blood clotting.  Any woman with lipedema and a personal or family history of bleeding or clotting should work with a healthcare provider to determine if additional testing is needed before liposuction surgery (103).

 

Removal of lipedema fat by liposuction that spares lymphatic vessels (wet, not dry, technique) has been performed primarily in Europe, especially in Germany, since the 1990s (104-107).  The fat is saturated with Klein solution which includes saline or lactated Ringers solution, an anesthetic such as lidocaine or prilocaine, epinephrine, sodium bicarbonate buffer(108), usually without steroid (109).  This tumescent technique provides turgor to the tissue allowing blunt microcannula to slide through the fat tissue avoiding creation of shearing forces and tissue damage. When power assisted, tiny, rapid vibrations of the microcannula break up fat which is then suctioned out of the tissue. Water jet assisted liposuction (WAL) uses jets of saline and Klein solution to release fat for suction with minimal damage to cells and vessels(106) without the waiting period required to tumesce the tissue. Laser assisted tumescent liposuction is another technique which some reserve for fibrotic areas such as the posterior thighs.

 

Most affected women undergo liposuction in stages, involving removal of an area of lipedema fat from the lower body and arms followed by a period of recovery and healing before returning to remove an adjacent region. The average number of surgeries for a women with Stage II lipedema ranged between two and three (110), but some had more than five (37). Patients are either awake during the liposuction procedure with or without conscious sedation (104,111,112), or general anesthesia, the former allowing for rapid recovery (111). Some medications used in general anesthesia reduce the pumping activity of lymphatic vessels (113-116). Prior to undergoing liposuction by a qualified surgeon, therefore, a patient should have a thorough understanding of the surgeon’s technique, whether the surgeon uses general anesthesia along with the type of analgesia, the number of surgeries performed by the surgeon and outcomes and their complication rate. After liposuction, the surgically treated areas may be quite tender and uncomfortable for days to weeks.

 

Most studies on liposuction are from surgeons performing the procedure, are not randomized or controlled, and do not include external oversight of data collection. Nevertheless, current data are compelling for benefit. Twenty-five women with lipedema had significant improvements in pain, tension in the legs, excessive warmth, muscle cramps, leg heaviness, tired legs, swelling, itching, general involvement of the skin, difficulty walking, quality of life, and appearance of the legs six months post-liposuction surgery (104). A larger study of 85 women from the same clinic demonstrated significant improvements six months after surgery for all complaints with the greatest improvement in quality of life (110). In a longer study from a different clinic, 21 women over an average of 3.7 years after their first liposuction procedure and 2.9 years after the second liposuction showed improvement in the parameters of body disproportion, swelling, edema and quality of life, except for bruising which improved in all but two of the women (105).  A retrospective study of women with Stage I or II lipedema from the same clinic, four, and eight years after liposuction, showed sustained improvements during follow-up for parameters including pain, sensitivity to pressure, edema, bruising, restriction of movement, cosmetic impairment, reduction of overall quality of life and overall impairment (117). The most interesting data was the reduced need for combined decongestive therapy four years after liposuction, which decreased further after eight years (37). 

 

Any surgery, including liposuction, requires that efficacy of the procedure and the medical necessity be demonstrated to the insurance company.  What are currently needed are well conducted randomized, controlled trials of sufficient numbers of patients with lipedema to determine which patients do and do not benefit from liposuction. In the meantime, documenting patient baseline characteristics and outcomes by surgeons in the United States will be important to understand the benefits of liposuction for lipedema in the US population compared to reports from other countries (e.g., Germany). It is notable that surgeons agree that quality of life is strongly and consistently improved by liposuction (104,110,117,118). 

 

COMPLETE DECONGESTIVE THERAPY

 

Complete decongestive therapy (CDT) is commonly recommended for the treatment of lymphedema and includes skin care, education on home exercise programs, manual lymphatic drainage (MLD) therapy, wrapping as needed to reduce fluid build-up, and skin care recommendations performed by physical and occupational therapists and licensed massage therapists that have undergone additional training. Many women with lipedema benefit from CDT with reduced pain, limb volume and capillary fragility (119-121). Near-infrared fluorescence lymphatic imaging (NIRFLI) has added additional techniques to MLD including the “Fill and Flush” method (122). Complete decongestive therapy also improves lymph flow in brain lymphatic vessels (123). Deeper tissue therapies to reduce fibrosis in the lipedema tissues may also be beneficial for patients with lipedema. 

 

PNEUMATIC COMPRESSION DEVICES

 

Studies have shown the benefit of advanced pneumatic compression devices (PCDs) in the treatment of lymphedema. There are also studies on the benefits of PCDs in the treatment of lipedema (119,124). Important for the distorted and dilated capillaries in lipedema (36,88), PCDs decrease capillary fragility (120), improving vessel quality. Along with manual therapy to improve flow of fluid through lipedema tissue, PCDs are also recommended in conjunction with liposuction surgery for lipedema (110). If a woman with lipedema responds well to manual therapy, or she tries a PCD and has a reduction in tissue volume, she should be offered PCD therapy to continue treatment at home when insurance will no longer cover CDT or when distance or commitments prevent regular professional visits. The PCD should ideally be an E0652 device with a segmented, multi-ported pump that allows for individual pressure calibration at each port. This allows the patient to alter pressure in areas of severe pain or for different shaped tissue. Pump garments should wrap around and treat the abdomen and pelvis when the legs are pumped, and the chest when the arms are pumped.  If basic compression pumps are prescribed (E0650; E0651), compression garments to protect the abdomen, pelvis, chest and/or head should be worn during pumping. Without these compression garments, fluid is pushed up the leg into the abdominal and pelvic area where it accumulates due to lymphatic dysfunction. As this fluid sits in the tissue with all its nutrients and protein, evidence suggests it may stimulate further adipogenesis (125). With an E0652 pump, the abdomen is treated along with the leg and the chest is treated along with the arm preventing pooling of lymph fluid. PCDs can be easily ordered by writing a prescription for durable medical equipment with multiple suppliers.

 

DEEP TISSUE THERAPY

 

Women with lipedema treated with deep tissue manual therapy have reduced pain, fat tissue on the legs, tissue volume, tissue fibrosis and leaky or fibrotic vessels (86,126). This deep tissue therapy is in the spectrum of meridian massage shown to reduce body weight (127) and is thought to  improve lymphatic flow through lipedema fat tissue. Massage also reduced fat in preterm infants (128). Instrument-assisted soft tissue (IAST) therapy has cc in lipedema fat tissue with noted reduction in palpable fibrosis after treatment. Instrument-assisted soft tissue  techniques include Astym therapy, which increased fibroblast activation and number, production of fibronectin, movement, and decreased pain in patients with fibrosis (129) and Graston technique which reduces pain and improves movement (130), and are performed by physical therapists who can be located on websites for these techniques. Traditional Chinese gua sha tools or bian stones have been used to improve pain and function (131) as has cupping (132).  Pressure required to occlude lymphatic function in the upper limb was found to be 86 mm Hg, suggesting that deeper treatment into the tissue is safe, for example at pressures ranging from 15 to 25 mm Hg used to reduce scars (133), and will not damage lymphatic vessels (122).

 

PSYCHOLOGICAL SUPPORT

 

Women with lipedema have often spent years looking for answers and help for their condition.  Healthcare providers often hold strong negative attitudes and stereotypes about people with obesity, which may reduce the quality of care they provide to women with lipedema (134). Poor quality of life associated with mobility and appearance-related stress associated with lipedema can result in depression (87). For many patients, they experience a huge sense of relief when they finally get a diagnosis of lipedema after trying a myriad of diets and exercise programs, even bariatric surgery to lose the lipedema fat. In addition to treatment recommendations in this chapter, there are a number of things a healthcare provider can do to help improve the lives of people living with lipedema: 1) Reduce focus on body weight in lipedema and provide education on improving metabolism, reducing inflammation and improving quality of the lipedema fat tissue (reducing fibrosis); 2) Use motivational interviewing focusing on strides made to improve markers of health including healthy eating, activity, metabolic lab markers, and social interactions; 3) Ensure that the clinic environment is welcoming with tables and chairs that allow women with larger lower bodies to be comfortable; 4) Ensure that patients with lipedema have identity safety in clinic situations and encourage healthy social interactions at home and in on-line social groups that also provide safe affiliations known to improve satisfaction of life for women with lipedema (135); and 5) Ensure that the continuum of care includes adequate referral resources for counselling, physical therapy and message, and when indicated pain management specialists. Including providers that understand lipedema and the physical and the psychological burden this diagnosis carries for patients is especially important (134).

 

MEDICATIONS AND SUPPLEMENTS

 

There are no medications and supplements specifically for lipedema.  Instead, recommendations regarding use of medications and supplements for the treatment of lipedema should focus on reducing tissue inflammation, fibrosis, swelling, pain, and pharmacologic weight loss management for those who are overweight or have obesity.  Supplements used for lipedema are, in part, based on literature for lymphedema and venous disease, both complications of lipedema.  Some medications exacerbate symptoms in lipedema and should be avoided (Table 5). 

 

Sympathomimetic Amines

 

Sympathomimetic amines (SA) such as phentermine and amphetamine are approved by the food and drug administration (FDA) for the treatment of obesity. Sympathomimetic amines bind to adrenergic receptors (AR) located on adipocytes to induce lipolysis, reducing the storage of fat.  Adrenergic receptors are also located on blood vessels and lymphatic vessels. Activating AR on blood vessels induces vasoconstriction. Activating AR on lymphatic vessels improves the efficiency of lymphatic pumping by increasing the force of contraction (136); medications or supplements that improve lymphatic pumping are lymphagogues. Amphetamine and dextroamphetamine alone or in combination are also FDA-approved for the treatment of attention deficit disorder (ADD), attention deficit hyperactivity disorder (ADHD), and narcolepsy.  The use of SA for treatment of lipedema may be beneficial in reducing fat and improving lymphatic pumping. A retrospective questionnaire study found that low dose sympathomimetic amines improved quality of life, reduced weight, clothing size, pain and leg heaviness in women with lipedema (137). Contraindications of sympathomimetic amines include advanced arteriosclerosis, symptomatic cardiovascular disease, moderate to severe hypertension, hyperthyroidism, known hypersensitivity or idiosyncrasy to the sympathomimetic amines, and glaucoma.

 

Diosmin

 

Diosmin is a bioflavonoid found in the rind of citrus fruit and is traditionally prescribed for the treatment of inflammation associated with chronic venous insufficiency. Diosmin was shown to reduce oxidative stress markers in people with chronic venous insufficiency (138). Diosmin also functions as a lymphagogue, and in combination with its anti-inflammatory activity, reduces edema (138). Women with lipedema who have a feeling of heaviness in their legs, obvious edema, chronic venous insufficiency or Stage II and III lipedema report feeling less pain and improved swelling on diosmin based on the author’s experience. Diosmin can be found over the counter or ordered by prescription as a medical food.  Placing lemons, limes or other citrus in water to soak before drinking is a way to intake diosmin throughout the day.

 

Metformin

 

There are no current medications that can be used to reduce fibrosis already present in lipedema fat tissue, for which liposuction and deep tissue therapy are better modalities.  Metformin and resveratrol have been shown to reduce the development of hypoxia-inducible factor (HIF)-1 inflammation and fibrosis in mice fed a high fat diet (139). Metformin also prevented fibrosis and restored glucose uptake in fat after insulin stimulation, although it did not prevent side effects of doxorubicin that included tissue loss and inflammatory response (140).  Metformin should be considered early in women with obesity and lipedema Stages II and III where fibrosis in the fat tissue is prominent, as well as in women who have signs of metabolic syndrome (69).

 

Selenium

 

Selenium is a mineral found in the soil and in high concentration in Brazil nuts (Bertholletia excelsa). Selenium has been demonstrated to have anti-inflammatory effects on multiple levels of the inflammatory cascade (141-144).  Edema was significantly decreased after selenium intake in two placebo-controlled trials for people with lymphedema (145,146) and improved complete decongestive therapy while reducing the incidence of erysipelas infections in patients with chronic lymphedema (145). Each Brazil nut contains approximately 200 mcg of selenium with a no observed adverse effects for dietary intake of selenium up to 800 mcg daily (147).  Care must be taken to follow blood selenium levels as selenium deficiency and excess can both adversely affect glucose and lipid metabolism and potentiate the risk of development of type 2 diabetes in several animal studies, with less clear associations in human studies (148). One case report of a woman with lipedema showed reduced leg volume with a combination of selenium and Butcher’s broom (149).

 

Table 5: Medications and Supplements to Avoid When Treating People with Lipedema

Medication

Used for

Reason to avoid

Thiazolidinediones

Diabetes

Increases subcutaneous fat tissue; fluid retention

Calcium channel blockers

Hypertension

Fluid retention

Oral Corticosteroids*

Reduce inflammation

Weaken tissue; fluid retention; rebound inflammation

NSAIDs

Pain

Fluid retention

Sex hormones

Hormone replacement

Fluid retention; implicated to effect development of lipedema

Beta blockers

Cardiac health

Fluid retention

Clonidine

Hypertension

Fluid retention

Gabapentin

Pain

Fluid retention

Furosemide**

Edema

Concentrates protein in the interstitial organ eventually halting fluid flux

*Nasal or inhaled corticosteroids have less effect; oral corticosteroids should be used when medically necessary

**Aldactone and hydrochlorothiazide have less adverse effects in women with lipedema

NSAIDs: Non-steroidal anti-inflammatory drugs

 

Concluding Remarks on Lipedema

 

Lipedema is a common disease mostly in women resulting in an enlargement of the adipofascia on the limbs due to excess fibrosis in the tissue that typically defies expectations for loss by lifestyle, weight-loss medications, and bariatric/metabolic surgical interventions. The presence of fibrosis, especially in the interstitial spaces where it may serve to restrict blood and lymph out flow, is thought to contribute to the resistance of this tissue to weight loss. Women with lipedema should be recognized prior to weight loss efforts so that expectations can be discussed, and manual therapies and other treatments can be considered to improve outcomes.  Medications and supplements can be tried, but liposuction should be considered for women with lipedema who fail conservative measures and following weight loss with medications and/or bariatric surgery. There is a wide variety of presentations of lipedema in women due to co-morbidities and other genetic and environmental influences. Therefore, every affected woman should be considered on a spectrum and treatments personalized.

 

FAMILIAL MULTIPLE LIPOMATOSIS

 

Familial multiple lipomatosis (FML) is a rare adipose disorder (RAD) of multiple lipomas in subcutaneous fat (OMIM 151900).  Some members in an FML family may have only a few lipomas whereas others may have hundreds to thousands; it is not understood why there is unequal penetrance in families. Lipomas usually are not painful or tender to the touch except while growing; they may also cause a slight feeling of itching or burning when forming. Some lipomas can be tender if they develop in areas of pressure such as on the back of the legs, the lower back (pressure from a chair), or the lateral wrist due to repetitive stress such as comes from using a computer mouse (150,151). Another example of trauma-induced lipomatous growth includes movement of the xiphoid process (152).  

 

According to older FML literature (153), "pain may suddenly develop in one of the lipomas (called lipoma dolorosa), and will gradually extend to involve more and more of the discrete lipomas.” The authors state that lipoma dolorosa syndrome in families with FML is not the same as Dercum disease (see below) (154). This is confusing as individuals with painful lipomas in an FML family have been described as having Dercum disease. While painful lipomas in a person with FML may also be on the spectrum of Dercum disease, a more precise name is FML with painful lipomas, especially when a family history of FML is known.

 

It is interesting that by observation in some families with FML, the men will develop lipomas and the women often develop obesity in line with lipedema. This suggests an overlap between the development of one fat disorder (FML) and another (lipedema) and should prompt more detailed questions regarding other potentially affected family members.

 

Prevalence of FML

 

Familial multiple lipomatosis is considered to be a rare disease with an estimated prevalence of 1/50,000 (155).

 

Genetics of FML

 

Familial multiple lipomatosis is usually inherited in an autosomal dominant manner with males and females equally affected. The gene High Mobility Group AT-Hook 2 (HMGA2; 12q15) has been implicated in FML but is not thought to be causative. A mutation in partner and localizer of breast cancer (BRCA2, DNA-repair associated gene), called PALB2, was described in a family with FML (156). PALB2 is an intranuclear protein that anchors BRCA2 to nuclear structures.  PALB2 mutations are associated with a 2-fold increased risk of breast cancer, a Fanconi anemia subset, pancreatic cancer and ovarian cancer (156).

 

Conditions Associated with FML

 

In case reports, FML has been associated other rare or unusual disorders (Table 6).  Because multiple lipomas are often linked with mutations in tumor suppressor genes, FML can be considered clinically to be a marker for the presence of an underlying tumor suppression gene mutation and affected patients and their families should be appropriately screened.

 

For example, in MEN-1, lipomas have  been reported in association with a recessive mutation in a tumor suppressor gene (157).  In a family with retinoblastoma and multiple lipomas, the lipomas were present in people with a gene mutation in the RB1 gene who did not develop retinoblastomas (158). Multiple lipomas in Cowden's disease can be due to a germline inactivation of PTEN/MMAC1 that renders a person susceptible to thyroid and breast malignancies (159). Other genes including other tumor suppressor genes have been implicated in the growth of lipomas (160). For example, a mutation was found in the tumor suppression gene PALB2 in a family with multiple lipomas suggestive of a diagnosis of FML (156).  And finally, lipomatosis like that of FML has also been reported in two cases after chemotherapy (161,162), a treatment known to be associated with an increased risk of cancer development.  Because of these associations, people with multiple lipomas should be considered at increased risk for cancers and a referral to a geneticist considered.

 

Table 6. Disorders Found in Association with Multiple Lipomas

Atypical mole syndrome (163)

Gastroduodenal lipomatosis (no gastroduodenal lipomatosis in proband’s mother) (164)

Celiac disease (165)

Cowden’s disease (159)

Gastrointestinal stromal tumor (166)

Interhemispheric brain lipoma with corpus callosum hypoplasia and the malformation of cortical development in a young woman with refractory epilepsy (167)

Neurofibromatosis (NF1) (168)

Multiple endocrine neoplasia (MEN)-1 (169)

Retinoblastoma (Rb1) (158)

Legius Syndrome (SPRED 1); autosomal dominant; multiple café-au-lait macules and skin fold freckling, ± macrocephaly, a Noonan-like appearance, learning difficulties and/or attention deficit in children and lipomas in adults (170)

 

Pathophysiology of FML

 

The pathophysiology of lipoma growth in FML is not known. Single lipomas of subcutaneous fat tissue are the most common benign tumor growths in humans and may be induced by genetic changes, trauma, inflammation, or other causes.  As detailed above, multiple lipomas tend to be linked with tumor suppression genes. People with FML are known to be insulin sensitive, therefore an insulin-resistant metabolic cause of FML is unlikely (171). Additionally, the presence of the lipomas themselves do not confer insulin resistance.

 

Imaging of Lipomas in FML

 

Lipomas in FML are identified by palpation as connected to skin, surrounded by fat or connected to other structures such as muscle or solid fascial structures. Localized pain can assist in finding smaller lipomas. Silky or tight clothing can also assist in palpation. Sonographic evaluation is the best most inexpensive means to identify lipomas other than palpation, but the average sensitivity for three Radiologists when retrospectively reviewing sonographic exams of lipomas was only 48%, and average accuracy was 59% (172). Magnetic resonance imaging without contrast can be used to find lipomas (173), but small lipomas, lipomas without a capsule, and lipomas with minimal fibrosis or surrounding edema remain difficult to identify.  Radiologists were able to render the correct diagnosis for lipoma versus liposarcoma in 69% of cases (174). Computed tomography (CT) scans have been used to differentiate lipomas from liposarcomas (175) but should be used after sonography and MRI to avoid excess radiation exposure.

 

Evaluation of the Patient With FML

 

The initial workup for people with FML includes a family history of lipomas and cancer, and any associated conditions such as nevi or neuropathy. The exam incudes assessment for multiple lipomas usually located on the trunk, lower back, arms, and thighs; rarely on the upper back or calves. Skin should be examined for nevi and cherry angiomas, the latter seen commonly with multiple lipomatosis (176). Due to the associated cancer risk, the exam includes examination of the thyroid and breasts for nodules. Reflexes should be checked along with monofilament and/or vibration assessments for peripheral neuropathy. Cancer screening as appropriate for sex and age should be advised, and appropriate labs ordered (Table 7).  Although there is no definitive association of FML with dyslipidemia, statin therapy may be helpful in lowering lipoma size (177) and so a lipid panel is also appropriate.

 

Table 7. The FML Workup

Family history

Lipomas; cancer; nevi; celiac; neuropathy

Medical history

Lipomas; cancer; nevi; celiac; neuropathy

Physical Exam

Lipomas

Trunk, arms, low back, flanks, abdomen, thigh.

Attached to skin, muscle, other.

Skin

Nevi; cherry angiomas

Thyroid

Nodules

Nervous system

Reflexes; skin sensory exam (monofilament)

Laboratory Studies

Thyroid

TSH

Blood fats

Lipid panel; other per family history

Fibrin clot (found in angiolipoma)

D-dimer

Food/gluten intolerance

Celiac panel

 

Treatment of FML

 

The current management of FML includes screening for associated conditions such as cancer (Table 7) and consideration of a referral to genetics for tumor suppressor gene workups as needed. A healthy diet and an exercise plan to avoid or reduce obesity is important as obesity in families with FML can be associated with pain (154) and, anecdotally, triathletes notice a reduction in lipoma size during high intensity training. A statin has been shown to reduce a lipoma in a case report (177). Painful lipomas or those that interfere with activities of daily living can be excised as needed but these procedures can cause numerous scars (Figure 5).  Massive amounts of lipomas can occur on the arms, hips/flanks, buttocks and thighs. Therefore, this condition can be psychologically devastating and people with severe FML do not consider it benign.

 

Liposuction is an option to excision of lipomas in people with FML as it provides good results in terms of skin appearance, and there is reported lack of recurrence or growth or development of other lipomas in the same area for at least 12 months (178). Injections of collagenase have been shown to shrink or destroy lipomas with minimal pain and good cosmetic result in a published abstract (179). Similar data were found for the detergent, deoxycholic acid (180), but anecdotally care should be taken not to inject too much detergent that can remain in the tissue requiring excision to remove. Additional treatments such as cryotherapy have been suggested and reviewed (181). More data is needed for the efficacy of injections and other therapies for the lipomas in FML as they are preferable to more invasive and scarring surgical techniques.

Figure 5. Multiple scars after excision of lipomas in FML.

Concluding Remarks on FML

 

Familial multiple lipomatosis is a rare disease of multiple lipomas often associated with mutations in a tumor suppressor gene. Therefore, people identified with FML should be assessed for cancer. Liposuction should be considered to remove symptomatic lipomas and is preferable to surgical excision as multiple excisions leave many scars.

 

ANGIOLIPOMATOSIS

 

Angiolipomatosis also known as angiolipoma microthromboticum (OMIM 206550) is a rare disease of multiple angiolipomas and connective tissue that occurs commonly in men and usually begins after puberty; one case of a child with an angiolipoma in a family in which the father also had angiolipomatosis has been reported (182). Angiolipomas have been described as vascular malformations or vascular lipomas where blood vessels occupy between 10-90% of the angiolipoma. In families with familial angioliopomatosis, lipomas and angiolipomas can exist in the same person. Subcutaneous angiolipomas usually occur on the trunk and limbs, rarely on the head, hands, or feet (183). The angiolipomas can be the size of a rice grain, pea, a marble or much larger and are tender to the touch and can be associated with intense pain.  Angiolipomas may or may not be visible and may be palpable or non-palpable depending on their location and size. Numerous case reports describe epidural or extradural spinal angiolipomas, and rare cases report colonic (184), bronchial (185), joint (186), and testicular angiolipomas (187). Angiolipomas are known to be painful, although not always, and should be distinguished from other painful neoplasms (188). One case of angiolipomatosis was reported to occur after treatment with corticosteroids (189).

 

The loose connective tissue of angiolipomas contains adipose cells, fibrotic tissue, vessels with fibrin clots, and mast cells as salient features (Figure 6). Due to the large number of vessels, angiolipomas are bluish in color through the skin. Interestingly, the vessels in angiolipomas can grow from the dermis into the territory of the epidermis making the vessels palpable as small raised areas on the skin (Figure 7). A plethora of capillary “cherry” angiomas where a capillary grows and dilates through the epidermis may be found on the skin in areas of angiolipomas (Figure 8).

Figure 6. Angiolipoma with mast cell with enlarged multiple vessel lumens and degraded tissue. The black arrow points to a classic fried egg appearance of a mast cell stained with Alcian Blue in angiolipoma tissue. Red arrows point to small fat cell remnants likely non-functional as evidenced by the absence of nuclei. Blood vessels are numerous and large for location. The green arrow demonstrates the remnant of a capillary. Connective tissue is evident especially in the area surrounding the mast cell as bluish fibers. Magnification 100X.

Figure 7. Multiple cherry angiomas present on the legs and arms of a woman with angiolipomatosis.

Figure 8. Histological features of angiolipomas. A. Small area of hypervascularity in an angiolipoma (40X). B. Blood vessels in an angiolipoma grow up and through the epidermis and are palpable on the skin (40X). C. Empty and presumed dead and non-functional vessel on the left containing an eosinophil next to two functional blood vessel lumens containing red blood cells (100X). Microthrombi can be seen as pale areas especially between the right side of the dead vessel and the lumen of the active vessel. Dead vessels may result in hypoxia and ischemia causing pain. D. Non-functioning blood vessel to the right and smaller fat cells surrounded by an enlarged interstitial organ (40X).

Prevalence of Angiolipomatosis

The prevalence of angiolipomatosis is unknown but it is considered to be a rare disease (190,191). 

 

Genetics of Angiolipomatosis

 

Angiolipomatosis most often occurs sporadically, but a family history can be identified in a minority of cases as autosomal dominant (192) or autosomal recessive (193,194). There are no known genes identified to date for angiolipomatosis.

Pathophysiology of Angiolipomatosis

Angiolipomas likely arise from fascia and therefore may also be painful because fascia is highly innervated, and when inflamed, is a likely source of pain (195). Inflamed fascia has robust angiogenesis (196) and may be important in the initial development of angiolipomas as resident mesenchymal cells in fascia can develop into adipocytes (197). It is thought that microthrombi in angiolipomas leads to necrosis of blood vessels, adipocytes and other components of adipofascia. Other hypotheses regarding pain include nerve damage from limited blood flow and tethering by fibrotic tissue. 

 

Subcutaneous angiolipomas are assumed to be congenital in origin where pubertal hormones may induce differentiation of adult adipose-derived stromal adipogenic precursors that reside in adipofascia; these precursors develop into adipocytes in intimate association with blood vessels (197). Vascular proliferation is thought to occur after repeated trauma to the fascia resulting in the development of an angiolipoma. However, there is a question of whether angiolipomas can become autonomous as a cancer. Two of three cases of angiolipomas in one publication suggest a neoplastic nature for these tumors due to deletion of parts of chromosome 13, a region containing the retinoblastoma gene, a tumor suppressor gene (198). The neoplastic nature of angiolipomas should be considered in individuals with significant numbers of angiolipomas and anti-neoplastic treatments considered when other conservative therapies fail.

Imaging of Angiolipomatosis

Identification of angiolipomas in tissue by Ga-PSMA PET/CT (199), magnetic resonance imaging (200), and ultrasound (201) allows surgeons to identify superficial and deeper angiolipomas targeted for removal. 

 

Treatment of Angiolipomatosis

 

SURGICAL

 

The only definitive treatment of angiolipomas to date is individual resection by excision or liposuction (202). Angiolipomas are typically removed if they are painful or restrict movement. A surgical emergency may occur to prevent hemorrhage of angiolipomas which can compress the spinal cord (203,204). Karyotypes of DNA from the angiolipomas should be assessed to determine the neoplastic nature of the angiolipomas so as to prepare the patient for the potential of multiple resections throughout life (198).

 

A concern with resection of lipomas is that inflammation is often a sequela of the removal process. As fascia plays an important role in the pathophysiology of angiolipomas, generation of inflammation in the fascia by surgical techniques has been anecdotally noted to incite a pain crisis. Removal of angiolipomas must therefore be considered carefully and manual or IAST therapies for the fascia should be considered after any surgery to speed recovery and reduce pain.

 

PAIN MANAGEMENT

 

The necrosis of tissue in angiolipomas and inflammation of fascia with all of its nerve endings can cause severe pain in individuals with angiolipomatosis.  In a case report from Germany, the systemic administration of acetylsalicylic acid, diclofenac, ketotifen, ranitidine, tramadol, or tilidine combined with naloxone did not provide adequate pain relief. In contrast, the antidepressant doxepin, which also has antihistaminergic effects to control the release of mast cell mediators, demonstrated good therapeutic efficiency for the pain from angiolipomas (205).  Depending on the mast cell burden in angiolipomas and their systemic effects including flushing, itching, nausea, diarrhea, angioedema, pain and a cadre of other signs and symptoms (206,207), individuals with angiolipomatosis may be considered to have mast cell disease or mast cell activation disease. Treatments to reduce the burden of mast cells in angiolipomas such as histamine 1 and/or histamine 2 receptor blockers, montelukast, non-steroidal anti-inflammatory drugs, antihistaminergic bioflavonoids such as quercetin or pycnogenol, amphetamines, and possibly stronger immunosuppressants that have been used for mastocytosis such as sunitinib (208) or mast cell activation syndrome such as tofacitinib (209) or imatinib (210) may provide benefit for pain and growth of angiolipomas. Non-neoplastic therapy for mast cell activation disease should be considered prior to the use of antineoplastic agents, which have been described extensively (211). Patients with angiolipomatosis can have a poor quality of life due to extreme pain and fatigue and consider suicide.  In these individuals, use of anti-neoplastic agents should be considered early. 

 

Many individuals with angiolipomatosis require opioid pain management and should be under the care of pain management specialists. Unfortunately, opioids can activate mast cells requiring concurrent treatment of mast cell signs and symptoms (212). Opioids should not be withheld during a pain crisis, and in fact may need to be escalated before weaning back down after the pain crisis has resolved.

 

Concluding Remarks on Angiolipomatosis

 

People with angiolipomas have severe pain that can be out of proportion to the outward appearance of the individual. Treatment with mast cell stabilizers, pain medications, and surgical treatments of angiolipomas are all important in management.  More research is needed for this rare disease to enable individuals with angiolipomas to live a full and active life.

 

DERCUM DISEASE

 

Dercum disease (DD; OMIM 103200) is a term used to describe extremely painful adipofascial tissue that is resistant to loss by diet and exercise and poorly responsive to analgesics. Other names include adiposis dolorosa (a term that is also used to describe women with lipedema) and Morbus Dercum. While Dercum disease is defined as painful fatty masses accompanied by other signs and symptoms of a chronic healing cycle disorder (25) (213), there remains a lot of confusion in the literature as to what exactly Dercum disease is. One review article stated that people with Dercum disease have obesity and chronic pain (214), which can easily be confused with people who have obesity and chronic pain for a variety of reasons including fibromyalgia. The old classification of Dercum disease and a new classification remain inadequate to differentiate the overlapping disorders that are bundled together as Dercum disease (65,215,216) because they describe only the phenotype and not the history (Table 8).

 

Table 8. Comparison of Outdated Classifications of Dercum Disease.

Older Classification

Previous Recent Classification

Type I: Diffuse Type. Widespread occurrence of painful lipomas in a diffuse manner

Diffuse Type. Diffusely painful adipose tissue that may present as painful folds of fats containing fat nodules that feel like pearls located around lymph node beds. Mostly resembles lipedema but extends or start in the trunk which differentiates it from lipedema

Type II: Generalized Nodular

Nodular type. Intense pain in and around grape-like clustered lipomas of variable size most commonly on the arms, legs, lower back or thorax; can include angiolipomas. Most resembles familial multiple lipomatosis

Type III. Localized Nodular

Nodular type. Intense pain in and around grape-like clustered lipomas of variable size in defined areas; can include angiolipomas

Type IV: Juxta-articular

NA

NA

Mixed Type: Combination of diffuse and nodular

 

A better classification of painful adipofascia considers the history of the disease (Table 9). For example, women with lipedema who become obese and/or develop lymphedema can metabolically become toxic or ill leading to the growth of painful masses in fat tissue. The etiology of these masses is likely due to the presence of inflammation known to slow lymphatic pumping leaving more pre-lymph fluid in the ECM, inducing adipogenesis, as lymph (even pre-lymph) makes fat grow. Women with lipedema, obesity, and painful fatty masses have dominated some studies on Dercum disease leading the authors to describe women with Dercum disease as having obesity and chronic pain (214). The masses that develop in women with lipedema and metabolic syndrome are similar to those that develop on the abdomens of people who have obesity and do not have lipedema. These tender masses resolve with weight loss and have been called Ander’s disease or adiposis tuberosa simplex (217). A good history taken from a woman with lipedema and the label Dercum disease, may reveal the development of lipedema earlier in life and additional weight gain later in life with development of tender masses, allowing treatment to be focused on lipedema and obesity rather than on Dercum disease.

 

Table 9. Conditions with Painful Adipofascia which Have Been Labeled as Dercum Disease

Types

Comments

Obesity-associated

Non-painful lipomas resolve with weight loss (Ander’s disease)

Lipedema with obesity and/or lymphedema

Painful lipomas resolve with weight loss; lipedema fat tissue remains

Familial multiple lipomatosis (FML) with obesity

Lipomas may get smaller and pain reduce with weight loss

Angiolipomas with or without obesity

Weight loss does not affect angiolipomas but is important to reduce inflammation

Localized due to trauma (218)

Multiple lipomas in an area of trauma and not just a single lipoma; likely due to injury of the fascia as with angiolipomas

Toxic/infectious; present around lymph node beds or diffuse with or without obesity

Likely due to a healing cycle disorder, infection, methylation deficiency, high oxalate or other toxin overload.

 

Signs and symptoms of Dercum disease include chronic pain, fatigue, brain fog, insomnia, cardiac arrhythmia most often tachycardia (palpitations), gastrointestinal distress often similar to irritable bowel syndrome, muscle weakness, tremor or jerking of muscles (myoclonus), joint pains, insulin resistance and diabetes, hypothyroidism, and other autoimmune disorders (219).  The signs and symptoms of Dercum disease have been suggested to be in the spectrum of fibromyalgia (220).

 

Dr. Dercum’s first patient was a woman with obesity with fat similar to a woman with Stage 3 lipedema (221). Dr. Dercum and his medical resident described rapid changes in fat tissue shape and size in real time suggestive of edema or fluid shifts. Therefore, involvement of the lymphatic system is likely in Dercum disease. Irregular and thickened lymphatic vessels have been described in Dercum disease suggesting that an altered lymphatic system can contribute to changes in the adipofascial tissue that found by palpation (222). Once people with Dercum disease are more accurately described by phenotype, there will be a better chance of finding a gene or biomarkers.

 

Prevalence of Dercum Disease

 

Many women with lipedema have been miscategorized as having Dercum disease when actually they have lipedema and metabolic syndrome, making the prevalence estimate of 1/1000 for Dercum disease in Sweden too high (223). There are no other prevalence studies of Dercum disease although the angiolipomatosis type is considered rare and the obesity- or lipedema-associated types are likely common, but these individuals are better described with what they have, angiolipomas, obesity, or lipedema respectively, with metabolic disease rather than lumping them all together as Dercum disease simply due to the presence of pain in the tissue.

 

Genetics of Dercum Disease

 

There is(are) currently no known gene(s) for Dercum disease.  Continuing to include people with angiolipomas, FML, and lipedema under the same moniker of Dercum disease will make it very difficult to discover genes important for these diseases when examining populations. The study of genes for specific families may be more helpful to find gene mutations that can then be assessed in individuals with painful adipofascia.

 

One family with familial multiple lipomatosis was found to have members that developed pain in the lipomas consistent with Dercum disease (adiposis dolorosa). While many of the family members with FML and pain also had obesity, not all were. Therefore the authors concluded that “adiposis dolorosa may in fact be an expression of familial multiple lipomas” (154). It remains unclear, however, why some individuals would develop pain and some not in a family with FML. Fascia can become inflamed for a variety of reasons including surgery, trauma, infection, toxin or drug exposure, and development of obesity. Lipomas in people with FML are often connected by a tail of connective tissue to solid fascial structures in the body, and fascia is a source of preadipocytes (Figure 9).  It may be that lipomas in FML are a marker of fascial disease and that pain in and around the lipomas depends on the amount and extent of inflammation present.  Other genes may modify susceptibility to prolonged inflammation including those yet to be identified in fibromyalgia (224).

Figure 9. Lipomas with fascial component. A. Lipoma with obvious tail of connective tissue. Removal of the fascia is important along with the lipoma to reduce additional growth in the area of removal. B. Long piece of connective tissue weaving amongst multiple lipomas during resection.

Pathophysiology of Dercum Disease

 

The pathophysiology of Dercum disease needs to be determined by type, something that very few papers have done so accurately. In mostly women with lipedema type Dercum disease, substance P was lower in the spinal fluid compared to controls (225), confirming a strong pain component is present when women with lipedema develop obesity and metabolic syndrome. In another study, interleukin-6 levels were elevated in the fat from women with Dercum disease compared to women without lipedema supporting Dercum disease as an inflammatory disorder (226). Weight stabilization and when possible, weight loss in patients with obesity should be a focus for women who have developed metabolic disease, in addition to caring for their lipedema.

 

The juxta-articular type of Dercum disease where nodules in the adipofascial tissue are present around joints had been associated with rheumatoid arthritis (227). Lymph nodes are present around many joints including the elbow (cubit nodes), knees (popliteal nodes), and hips (femoral nodes), and in these locations adipofacial nodules have been found. Cases of juxta-articular Dercum disease suggest that inflammation in the adipofascia around joints may reduce lymphatic pumping in these areas resulting in a backup of fluid in the interstitial body leading to densification of fascia and eventually fibrosis around lobules of fat making them palpable as nodules. These nodules are tender due to inflammation of the fascia and nerves. As an example, a woman with rheumatoid arthritis was treated with tocilizumab, a humanized monoclonal antibody of class IgG1, targeting interleukin-6 receptors, and developed painful fatty masses of her knees documented by MRI (228). A similar pathophysiology would be likely for the trauma-induced Dercum disease. 

 

Familial multiple lipomatosis and angiolipomas have been previously discussed including why pain develops in angiolipomas. It is unclear why a person with FML would suddenly develop pain in and around lipomas qualifying for a diagnosis of Dercum disease (FML type with pain).  The presence of inflammation occurring in the body of a person with FML such as from obesity, trauma, hypermobile joint spectrum disorders, arthritis and any other inflammatory condition that includes the fascia in the inflammatory process likely accounts for the development of pain in FML. Resolving the inflammation in the fascia may reduce the pain and return the diagnosis back to FML alone.

 

One theory on the origin of Dercum Disease is based on the work of Robert Naviaux in which a failure of healing of inflammation occurs (213). According to Naviaux, a normal healing cycle includes normal wakefulness, restorative sleep, fitness and healthy aging. The cell “danger response” is an evolutionarily conserved cellular metabolic response activated when a cell encounters a threat that could injure or kill it, examples of which can be microbial, chemical, physical, or psychological in nature. Chronic disease occurs when cells fail to heal or contain inflammation, and a toxic repeating loop of incomplete recovery and re-injury occurs. Chronic pain disorders are included by Naviaux as a healing disorder and the author feels many people with Dercum disease fall into this category.

 

Imaging of Dercum Disease

 

The most inexpensive means to document lipomas in the adipofascia of people with Dercum disease is by ultrasound. Ultrasound findings include a hyperechogeneity (higher density) to the lipoma suggesting fibrotic tissue and no increased Doppler signal (minimal blood flow) (229).

 

Magnetic resonance imaging of the tissue of people with Dercum disease has found lymphedema in a woman and multiple lipomas in a man  (230). Nodular type lipomas have also been visualized by MRI in the tissue of people with Dercum disease (229). The lipomas were multiple, oblong, fatty lesions in the superficial subcutaneous adipose tissue, mostly < 2 cm in long axis diameter. A nodular ("blush-like") fluid signal was also found without the presence of contrast. According to Richard Semelka, MD, gadolinium contrast should not be used in people with Dercum disease unless absolutely necessary to avoid any risk for development of gadolinium deposition disease (231,232). MRI images demonstrate variability in the tissue of people with Dercum disease, from lymphedema to distinct lipomas, and exemplify the different phenotypes under the moniker of Dercum disease. To date there are no confirmed connections between multiple lipomas as in FML or trauma-induced Dercum disease and development of lymphedema.

 

Conditions Associated with Dercum Disease

 

Dercum disease has been associated with many conditions such as disrupted sleep cycle, headaches, cognitive difficulties, tachycardia, shortness of breath, and gastrointestinal symptoms (219). Many of these symptoms are consistent with mast cell activation disease (MCAD).  Therefore, MCAD is considered an associated condition in Dercum disease. Diabetes is common in Dercum disease (25,219) and cardiovascular disease should be evaluated for and treated in any person with Dercum disease especially if blood markers of inflammation are high, such as C-reactive protein. A woman with Dercum disease had a dysfunctional arteriolar venous reflex in her arm suggesting a blood vascular or nerve problem in Dercum disease (233).  Another case of a woman with the FML type of painful lipomas was described who had dizziness followed by left sided sensory-motor deficit suggestive of a vascular origin (234).  Anecdotally, some of the author’s patients with Dercum disease also have postural orthostatic tachycardia syndrome (POTS). Fibromyalgia is often an accompanying diagnosis in people with Dercum disease as are other pain syndromes such as migraines (25).

 

Many people with Dercum disease become concerned that the painful lipomas can spread throughout the body as with cancer.  Once case of a lipoma on the uterus of a woman with Dercum disease is known to the author, and two women with Dercum disease had invasive calcaneal lipomas that were resected.  Other lipomas in people with Dercum disease have been identified in the gastrointestinal system but these need to be verified. Lipomatous hypertrophy of the interatrial septum was found in one person with Dercum disease (235), but this type of fat can also be associated with obesity (236). Altered lymphatics were found in a few cases of Dercum disease (222). One family had Dercum disease with dysarthria, visual pursuit defect and progressive dystonia (237).

 

General swelling, a sensation of a “heaviness” in tissue, increased pain in one limb, or aching limbs all suggest that a lymphatic dysfunction may be present, which can be evaluated using lymphangioscintigraphy, which is performed in most nuclear medicine departments, or near infrared lymphatic imaging using indocyanine green, which is not yet to the point of being readily clinically available. Finding altered lymphatic function can change clinical management by steering practitioners towards prescribing manual lymphatic drainage therapy and compression garments to contain and support lymphatic flow.

 

Treatment of Dercum Disease

 

To maintain a healthy weight or lose excess adipofascial tissue, people with Dercum disease should be encouraged to eat a healthy diet such as Mediterranean, DASH, low processed sugar, plant-based low inflammatory foods, or foods that are low in histamine if mast cell activation disease is present or suspected; they should also undertake a graded exercise program.

 

PAIN MANAGEMENT OF DERCUM DISEASE

 

Signs and symptoms common in people with Dercum disease, including pain. should be treated symptomatically (Table 10). Opioids are often used for pain treatment for Dercum disease, but doses can escalate over time and care should be taken to try and find additional alternative treatments.

 

Table 10. Medications Used to Treat Pain and Other Symptoms in People with Dercum Disease

Medication

Comments

Deoxycholic acid (238)

Injection of deoxycholic acid reduced pain and size of lipomas in a man with FML type of Dercum disease

Doxepin (205)

Has antihistaminergic activity therefore useful for pain, depression and mast cell activation disease

Intravenous lidocaine (239)

Ketamine is often used in addition to or in place of lidocaine if not effective

Topical lidocaine (240)

Often combined with other medications in topical form such as EMLA (241)

Metformin (242)

Useful for metabolic disease and when inflammatory markers are high

Mexiletine and amitriptyline (243)

Mexilitine has been described as oral lidocaine and offers an alternative to opioids

Low dose naltrexone (244)

Effective for fibromyalgia pain, a condition often present in Dercum disease

Pregabalin (245)

Gabapentin has also been shown to reduce pain (246) but may increase edema

Sympathomimetic amines (137)

Phentermine, dextroamphetamine, amphetamine; sympathomimetic amines resolved lipomas and liver fat in two cases of Dercum disease.

 

NON-MEDICATION TREATMENT OF DERCUM DISEASE

 

People with Dercum disease should be offered manual or IAST therapies or pool/water therapy to reduce pain, improve mobility and impede progression of the disease. Manual lymphatic drainage combined with pregabalin improved weight and pain in a woman with Dercum disease  (245). It has also been reported that fascia improved, pain reduced, and fat was lost after women with lipedema and Dercum disease received deep tissue therapy (86,126).

 

Liposuction has been used as treatment for Dercum disease (247), reducing pain by one point on a visual analogue scale (248) and improving insulin sensitivity (249). Surgeons removing lipomas by liposuction must have experience in the removal of fibrotic tissue and manual or IAST therapies should be performed before and after any surgery to keep inflammation levels at a minimum.

 

Transcutaneous frequency rhythmic electrical modulation system (FREMS) reduced pain and the size of lipomas in one case of Dercum disease (250). Cycling hypobaric air around ten people with Dercum disease improved pain and mental quality of life after five days of therapy (251). Cycling air around the body by sequential pneumatic compression pump therapy is also useful for people with Dercum disease due to the presence of lymphatic dysfunction (222,229).

 

Concluding Remarks on Dercum Disease

 

People with Dercum disease have painful lipomas and other signs and symptoms of a healing disorder. The different types of Dercum disease need to be delineated before any gene or biomarker can be found. The pain and associated signs and symptoms of Dercum disease should be treated to improve quality of life. People with Dercum disease may be at high risk for cardiovascular disease and cardiovascular risk factors should be closely monitored and treated when appropriate.

 

MULTIPLE SYMMETRIC LIPOMATOSIS

 

Multiple symmetric lipomatosis (MSL; OMIM#151800) also known as Madelung disease, Launois-Bensaudesyndrome, cephalothoracic lipodystrophy, and benign symmetric lipomatosis is a rare disease first described by Brodie in 1846.This disorder is clearly not benign. Madelung reported data on 33 cases, but the classical description of the disease is attributed to Launois and Bensaude who published a detailed account of 65 cases in 1898. The literature on MSL was initially dominated by research on men with alcoholism; however, people who do not consume alcohol (252), women, and children are also affected (253).

 

There are different types of MSL described initially by two different groups and reclassified in 2018 based on a German cohort of 45 patients (Table 11 and Figure 10) (91). 

 

Table 11. Types of Multiple Symmetric Lipomatosis (Locations of Abnormal Fat Tissue)

Types

Old Classification (254)

Old Classification (255)

New German Classification (91)

I

Neck, shoulders, supraclavicular triangle, and proximal upper limbs

Neck, upper back, shoulder girdle,

and upper arms

Ia: Neck

Ib: Neck, shoulder girdle, upper arms

Ic: Neck, shoulder girdle, upper arms, chest, abdomen, upper and lower back

II

Abdomen and thighs

Shoulder girdle, deltoid region, upper arms, and thorax

Hips, bottom, and upper legs

III

Thigh or female type similar to lipedema

Gynecoid type: Thighs

and medial side of the knees

General distribution skipping head, forearms, and lower legs

IV

NA

Abdominal type: Abdomen

NA

Women tend to have Type II MSL and the authors state that it is difficult to differentiate women with lipedema from women with MSL type II. Criteria the authors used to distinguish the two are “the hips and bottom are affected [in Type II MSL] which are not affected in patients suffering from lipedema.”  Lipedema, however, does affect the hips and buttocks in Types I-III lipedema (Figure 3). Finding a gene or biomarker for lipedema and MSL will be ultimately be helpful in distinguishing these adipofascial disorders.

Figure 10. Two different presentations of MSL. The man has MSL with a Charcot Marie Tooth presentation with increased fat on the upper body even after multiple resections, and the woman on the right has increased fat on the arms and upper back consistent with the old classification of MSL Type II.

In rare cases, MSL SAT can invade the muscles of the tongue (256,257), vocal cords (258), and periorbital area (259). Tracheal or esophageal compression can occur resulting in superior vena cava syndrome (260).

 

Prevalence of MSL

 

Multiple symmetric lipomatosis is considered rare occurring 1:25,000 in a primarily male Italian population (261) and 1:25,000 in a German population where females outnumbered men 2.5:1 (91).

 

Genetics of MSL

 

Multiple deletions of mitochondrial DNA, and the myoclonus epilepsy and ragged red fibers (MERRF) tRNA(Lys) A>G(8344) mutation have been found in some cases of MSL (262,263) but not in others (264,265). Chalk et al. found no mitochondrial pathology or mutations in four siblings with MSL with a pattern favoring autosomal recessive (265). Another study examined individuals with mitochondrial mutations and found that MSL was a rare sign of mitochondrial disease with a strong association between multiple lipomas and lysine tRNA mutations (266). If triglyceride cannot be mobilized from fat, then along with adipogenesis (via microRNAs), fat would be expected to increase. Indeed, a mutation in the LIPE gene coding for hormone sensitive lipase was found to be mutated in a family with MSL and lipodystrophy (267).

 

Mutations in the MFN2 gene coding for mitofusin 2 have been found to cause MSL with Charcot Marie Tooth Disease (268). Mitofusin 2 helps to regulate the morphology of mitochondria by controlling the fusion process. Individuals with mutations in MFN2 have increased fat on the upper part of the body and a lipodystrophy or lack of fat on other aspects of the body. These data support pathophysiology of MSL hypothesis 1 above for the development of MSL but also support hypothesis 2 in that alcohol may cause widespread damage to mitochondria.

 

Pathophysiology of MSL

 

HYPOTHESIS 1: BROWN ADIPOSE TISSUE

 

The dorsocervical fat pad (buffalo hump) is thought to be a location of brown adipose tissue found both in MSL (264,269,270) and HIV-associated lipodystrophy (271-274) suggesting that the abnormal fat tissue in both of these conditions arises from brown adipocytes (275).  Uncoupling protein (UCP)-1 has been shown to be activated in HIV-associated lipodystrophy and in agreement, calcyphosine-like (CAPSL), important in early adipogenesis, was down-regulated and uncoupling protein (UCP)-1 upregulated in eleven individuals, one with familial MSL and ten with sporadic MSL disease (276). Stromal vascular cells grown out of MSL fat tissue resulted in multiloculated adipocytes consistent with brown adipocytes (277,278). These data suggest that altered pre-adipocyte mesenchymal stem cells, adipogenesis and energy metabolism are important in development of MSL fat. In support, microRNAs miR-125a-3p and miR-483-5p are significantly increased in the fat of patients with MSL. These microRNAs promote adipogenesis through regulating the RhoA/ROCK1/ERK1/2 pathway (279). Finally, stem cells from MSL tissue showed significantly higher proliferative activity (280) suggesting a defect in regulation of adipogenesis. That brown fat was not found by 18 F-fluorodeoxyglucose (18 F-FDG) uptake using PET/CT in areas of MSL tissue (281) does not rule out the brown fat hypothesis as there may be browning of MSL fat rather than as strict replication of brown adipocytes.

 

HYPOTHESIS 2: INFLAMMATION, ALCOHOL AND THE LYMPHATIC SYSTEM

 

Further research is needed to determine the exact pathophysiology involved in the development of MSL fat tissue, but the fact that alcohol is damaging to many tissues in the body suggests that inflammation may play a role. Interleukin-6 levels were elevated in MSL tissue compared to unaffected tissue (280), and ethanol intake increases CYP2E1 activity in adipose tissue, leading to apoptosis of adipocytes through activation of the pro-apoptotic Bcl-2 family protein Bid, resulting in activation of complement via C1q, and adipose tissue inflammation (282).

 

The liver produces over 50% of lymphatic fluid that enters the thoracic lymphatic ducts in the great veins in the neck (283).  When the liver is fatty or cirrhotic, the liver produces even more lymphatic fluid (284). That many men with men with MSL develop fat around the neck in the location of the thoracic ducts or abdomen where the digestive tract transports approximately 2/3 of lymphatic fluid, becomes intriguing and may suggest involvement of the lymphatic system.  Women have more developed vasculature, including lymphatics, in subcutaneous adipose tissue and therefore, if lymphatic vessels are important in the pathophysiology of MSL, they could be expected to have a different phenotype than men.  Rats provided acute alcohol intoxication were found to have mesenteric lymphatic hyperpermeability (thoracic duct was not examined), a peri-lymphatic adipose tissue inflammatory response, and an altered systemic adipokine profile (285). When lymphatic vessels leak, fat grows (52). Alcohol and other mediators of lymphatic vessel leakage may therefore play a role in MSL.

 

Disorders Associated with MSL

 

Associated disorders include liver disease, dyslipidemia, metabolic syndrome, hypertriglyceridemia, hypothyroidism, diabetes mellitus, and peripheral and autonomic neuropathy (Figure 11).

Figure 11. Disorders often associated with MSL (Madelung disease). Copyright © 2018 Szewc et al. (286). This work is published and licensed by Dove Medical Press Limited. Full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution – Non Commercial (unported, v3.0) License (http://creativecommons.org/licenses/by-nc/3.0/).

Morbidity and mortality in MSL is thought to be high with sudden non-coronary death accounting for a large percentage of deaths in one series of primarily men (254). The neuropathology of MSL is a distal axonal demyelination different from that associated with alcohol intake and impairment of autonomic function has been suggested as a possible cause of sudden death; this impairment seems to prevalently involve the autonomic nervous system and not related to a high alcohol intake.

 

Treatment of MSL

 

Anyone with MSL should be encouraged to stop intake of alcohol. The only definitive treatment of MSL is liposuction or excision of the MSL tissue. The advantages of lipectomy is more complete removal of MSL tissue and better control of iatrogenic damage to nearby structures. Liposuction, however, achieves good cosmetic results and is simpler and less invasive than lipectomy (287). Multiple symmetric lipomatosis tissue tends to recur after liposuction and even excision. Therefore, other treatments are needed to slow down the progression of this disease to improve quality of life. Some believe that combining excision with liposuction can reduce recurrence (288).

 

Mesotherapy is a procedure that involves injections of multiple substances such as pharmaceuticals and/or vitamins into subcutaneous fat to reduce the fat tissue or cellulite. Such substances include phosphatidylcholine, multivitamins, pentoxifylline, aminophylline, hyaluronic acid, yohimbine, collagenase and others. Mesotherapy has been used to treat MSL but the injections can cause fibrosis which can make excision or liposuction difficult (289).

 

Concluding Remarks on MSL

 

Multiple symmetric lipomatosis is a rare adipofasial disorder associated with alcohol use, but not always. The pathophysiology is unknown but may involve early adipogenesis, mitochondrial dysfunction, and brown adipose tissue formation. Women with MSL may have lipedema and vice versa, therefore a gene or biomarker is needed to identify people with different types of MSL. Surgical treatment remains the only therapy for MSL.

 

OVERALL CONCLUSIONS ON ADIPOFASCIAL DISEASES

 

Aipofascial diseases occur when there is an increase in adipofascial tissue on the body that becomes fibrotic and is resistant to loss by lifestyle change. Until such time that better understanding of the pathophysiology of these disorder hints at other treatment modalities, these disorders often require removal by surgical means. Many of the diseases overlap, making identification difficult and will remain so until additional genes or biomarkers are clinically available. 

 

A comparison table of the five adipofascial disorders presented in this chapter can be helpful (Table 12).

 

Table 12.  Comparison of Adipofascial Diseases

Characteristic

Lipedema

DD

MSL

FML

Angiolipomas

Fat Location

Limbs

Global

Upper body

Trunk, arms, thighs

Global

Diet-resistant fat

Yes

Yes

Yes

Yes

Yes

Lipomas

+

+++

+++

+++

+++

Time SAT change

Puberty

Adult

Adult

Child, adult

Young adult

Painful SAT

Yes

Yes

Not usually

Not usually

Yes

Sex

Female

Female

Male

Male, female

Male, female

Lymphatic dysfunction

Yes

Yes

Yes

Possible

Unknown

Prevalence

Common

Rare

Rare

Rare

Rare

Associated conditions

Lymphedema

Autoimmune; diabetes

Neuropathy

Moles; neuropathy

Unknown

Inheritance Pattern

Autosomal dominant; incomplete penetrance

Autosomal dominant; sex-specific influence

Autosomal dominant or recessive

Autosomal dominant

Autosomal dominant; spontaneous

Gene

None

None

LIPE (267)

MFN2 (268)

tRNALys(266)

PALB2(156)

None

Biomarkers

None

None

miRNA (279)

None

None

Abbreviations: miRNA: microRNA; PALB2; Partner and localizer of BRCA2

 

REFERENCES

 

  1. Herbst KL. Rare adipose disorders (RADs) masquerading as obesity. Acta Pharmacol Sin 2012; 33:155-172. doi: 110.1038/aps.2011.1153.
  2. Luong Q, Huang J, Lee KY. Deciphering White Adipose Tissue Heterogeneity. Biology (Basel) 2019; 8(2).biology8020023. doi: 8020010.8023390/biology8020023.
  3. Hussain I, Patni N, Garg A. Lipodystrophies, dyslipidaemias and atherosclerotic cardiovascular disease. Pathology 2019; 51:202-212. doi: 210.1016/j.pathol.2018.1011.1004. Epub 2018 Dec 1027.
  4. Akinci B, Sahinoz M, Oral E. Lipodystrophy Syndromes: Presentation and Treatment. In: Feingold KR AB, Boyce A, Chrousos G, Dungan K, Grossman A, Hershman JM, Kaltsas G, Koch C, Kopp P, Korbonits M, McLachlan R, Morley JE, New M, Perreault L, Purnell J, Rebar R, Singer F, Trence DL, Vinik A, Wilson DP, ed. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000-2018.
  5. Hogan S, Velez MW, Kaminer MS. Updates on the understanding and treatment of cellulite. Semin Cutan Med Surg 2018; 37:242-246. doi: 210.12788/j.sder.12018.12056.
  6. van der Valk ES, van den Akker ELT, Savas M, Kleinendorst L, Visser JA, Van Haelst MM, Sharma AM, van Rossum EFC. A comprehensive diagnostic approach to detect underlying causes of obesity in adults. Obes Rev 2019; 20:795-804. doi: 710.1111/obr.12836. Epub 12019 Mar 12831.
  7. Kyrou I, Randeva HS, Tsigos C, Kaltsas G, Weickert MO. Clinical Problems Caused by Obesity. In: Feingold KR AB, Boyce A, Chrousos G, Dungan K, Grossman A, Hershman JM, Kaltsas G, Koch C, Kopp P, Korbonits M, McLachlan R, Morley JE, New M, Perreault L, Purnell J, Rebar R, Singer F, Trence DL, Vinik A, Wilson DP, ed. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000-2018.
  8. Gruzdeva O, Borodkina D, Uchasova E, Dyleva Y, Barbarash O. Localization of fat depots and cardiovascular risk. Lipids Health Dis 2018; 17:218. doi: 210.1186/s12944-12018-10856-12948.
  9. Ahlman H, Nilsson. The gut as the largest endocrine organ in the body. Ann Oncol 2001; 12:S63-68.
  10. Coelho M, Oliveira T, Fernandes R. Biochemistry of adipose tissue: an endocrine organ. Archives of medical science : AMS 2013; 9:191-200
  11. Chung KJ, Nati M, Chavakis T, Chatzigeorgiou A. Innate immune cells in the adipose tissue. Rev Endocr Metab Disord 2018; 19:283-292. doi: 210.1007/s11154-11018-19451-11156.
  12. Del Corno M, Conti L, Gessani S. Innate Lymphocytes in Adipose Tissue Homeostasis and Their Alterations in Obesity and Colorectal Cancer. Front Immunol 2018; 9:2556.:10.3389/fimmu.2018.02556. eCollection 02018.
  13. Chen Y, Rehal S, Roizes S, Zhu HL, Cole WC, von der Weid PY. The pro-inflammatory cytokine TNF-alpha inhibits lymphatic pumping via activation of the NF-kappaB-iNOS signaling pathway. Microcirculation 2017; 24(3).10.1111/micc.12364.
  14. Arngrim N, Simonsen L, Holst JJ, Bulow J. Reduced adipose tissue lymphatic drainage of macromolecules in obese subjects: a possible link between obesity and local tissue inflammation? Int J Obes (Lond) 2013; 37:748-750. doi: 710.1038/ijo.2012.1098. Epub 2012 Jul 1033.
  15. Blaak EE, van Baak MA, Kemerink GJ, Pakbiers MT, Heidendal GA, Saris WH. Beta-adrenergic stimulation and abdominal subcutaneous fat blood flow in lean, obese, and reduced-obese subjects. Metabolism 1995; 44:183-187.
  16. L'Hermitte F, Behar A, Paries J, Cohen-Boulakia F, Attali JR, Valensi P. Impairment of lymphatic function in women with gynoid adiposity and swelling syndrome. Metabolism 2003; 52:805-809.
  17. Kumar V, Abbas AK, Fausto N. Tissue renewal and repair: regeneration, healing, and fibrosis. In: Abbas VKAK, Fausto N, eds. Pathologic Basis of Disease. Philadelphia, PA, USA: Elsevier Saunders; 2005.
  18. Usunier B, Benderitter M, Tamarat R, Chapel A. Management of fibrosis: the mesenchymal stromal cells breakthrough. Stem Cells Int 2014; 2014:340257.:10.1155/2014/340257. Epub 342014 Jul 340214.
  19. Sun K, Tordjman J, Clement K, Scherer PE. Fibrosis and adipose tissue dysfunction. Cell Metab 2013; 18:470-477. doi: 410.1016/j.cmet.2013.1006.1016. Epub 2013 Aug 1015.
  20. Crewe C, An YA, Scherer PE. The ominous triad of adipose tissue dysfunction: inflammation, fibrosis, and impaired angiogenesis. J Clin Invest 2017; 127:74-82. doi: 10.1172/JCI88883. Epub 82017 Jan 88883.
  21. Chabot K, Gauthier MS, Garneau PY, Rabasa-Lhoret R. Evolution of subcutaneous adipose tissue fibrosis after bariatric surgery. Diabetes Metab 2017; 43:125-133. doi: 110.1016/j.diabet.2016.1010.1004. Epub 2016 Nov 1011.
  22. Bel Lassen P, Charlotte F, Liu Y, Bedossa P, Le Naour G, Tordjman J, Poitou C, Bouillot JL, Genser L, Zucker JD, Sokolovska N, Aron-Wisnewsky J, Clement K. The FAT Score, a Fibrosis Score of Adipose Tissue: Predicting Weight-Loss Outcome After Gastric Bypass. J Clin Endocrinol Metab 2017; 102:2443-2453. doi: 2410.1210/jc.2017-00138.
  23. Allen EV, Hines EAJ. Lipedema of the legs: A syndrome characterised by fat legs and orthostatic edema. Proc Staff Meet Mayo Clin 1940; 15:184-187
  24. Fife CE, Maus EA, Carter MJ. Lipedema: a frequently misdiagnosed and misunderstood fatty deposition syndrome. Adv 2010; 23:81-92; quiz 93-84.
  25. Beltran K, Herbst KL. Differentiating lipedema and Dercum's disease. Int J Obes (Lond) 2017; 41:240-245.
  26. Herbst K, Mirkovskaya L, Bharhagava A, Chava Y, Te CH. Lipedema Fat and Signs and Symptoms of Illness, Increase with Advancing Stage. Archives of Medicine 2015; 7:1-8
  27. Meier-Vollrath I, Schmeller W. Lipoedema — current status, new perspectives. . Journal der Deutschen Dermatologischen Gesellschaft 2004; 2:181-186
  28. Wold LE, Hines EA, Jr., Allen EV. Lipedema of the legs; a syndrome characterized by fat legs and edema. Ann Intern Med 1951; 34:1243-1250
  29. Coppel T, Cunneen J, Fetzer S, Gordon K, Hardy D, Jones J, McCarroll A, O'Neill C, Smith S, White C, Williams A. Best Practice Guidelines: The management of lipoedema. Wounds UK 2017;13(1). http://www.wounds-uk.com/best-practice-statements/best-practice-guidelines-the-management-of-lipoedema.
  30. Herpertz U. Krankheitsspektrum des Lipödems an einer Lymphologischen Fachklinik - Erscheinungsformen, Mischbilder und Behandlungsmöglichkeiten. vasomed 1997:301-307
  31. Forner-Cordero I, Szolnoky G, Forner-Cordero A, Kemény L. Lipedema: an overview of its clinical manifestations, diagnosis and treatment of the disproportional fatty deposition syndrome - systematic review. Clinical Obesity 2012; 2:86-95
  32. Schook CC, Mulliken JB, Fishman SJ, Alomari AI, Grant FD, Greene AK. Differential diagnosis of lower extremity enlargement in pediatric patients referred with a diagnosis of lymphedema. Plast Reconstr Surg 2011; 127:1571-1581. doi: 1510.1097/PRS.1570b1013e31820a31864f31823.
  33. Marshall M, Schwahn-Schreiber C. Prävalenz des Lipödems bei berufstätigen Frauen in Deutschland (Lipödem-3-Studie). Phlebologie 2011; 3:127-134
  34. Schmeller W, Meier-Vollrath I. Lipödem-aktuelles zu einem weitgehend unbekannter Krankheitsbild. Aktuelle Dermatologie 2007; 33:1-10
  35. Greer KE. Lipedema of the legs. Cutis 1974; 14:98
  36. Földi E, Földi M. Das Lipödem. In: Földi M, Földi E, Kubik S, eds. Lehrbuch der Lymphologie für Mediziner, Masseure und Physiotherapeuten. Munich Elsevier, Urban&Fischer; 2005:443-453.
  37. Schmeller W, Hueppe M, Meier-Vollrath I. Tumescent liposuction in lipoedema yields good long-term results. Br J Dermatol 2012; 166:161-168. doi: 110.1111/j.1365-2133.2011.10566.x. Epub 12011 Nov 10517.
  38. Child AH, Gordon KD, Sharpe P, Brice G, Ostergaard P, Jeffery S, Mortimer PS. Lipedema: an inherited condition. Am J Med Genet A 2010; 152A:970-976. doi: 910.1002/ajmg.a.33313.
  39. Bano G, Mansour S, Brice G, Ostergaard P, Mortimer PS, Jeffery S, Nussey S. Pit-1 mutation and lipoedema in a family. Exp Clin Endocrinol Diabetes 2009; 118:377-380

 

  1. Berryman DE, List EO, Coschigano KT, Behar K, Kim JK, Kopchick JJ. Comparing adiposity profiles in three mouse models with altered GH signaling. Growth Horm IGF Res 2004; 14:309-318. doi: 310.1016/j.ghir.2004.1002.1005.
  2. Waxler JL, Guardino C, Feinn RS, Lee H, Pober BR, Stanley TL. Altered body composition, lipedema, and decreased bone density in individuals with Williams syndrome: A preliminary report.
  3. Paolacci S, Precone V, Acquaviva F, Chiurazzi P, Fulcheri E, Pinelli M, Buffelli F, Michelini S, Herbst KL, Unfer V, Bertelli M. Genetics of lipedema: new perspectives on genetic research and molecular diagnoses. European Review for Medical and Pharmacological Sciences 2019; 23:5581-5594
  4. Zazulak BT, Paterno M, Myer GD, Romani WA, Hewett TE. The effects of the menstrual cycle on anterior knee laxity: a systematic review. Sports Med 2006; 36:847-862. doi: 810.2165/00007256-200636100-200600004.
  5. Szolnoky G, Nemes A, Gavaller H, Forster T, Kemeny L. Lipedema is associated with increased aortic stiffness. Lymphology 2012; 45:71-79.
  6. Jagtman BA, Kuiper JP, Brakkee AJ. [Measurements of skin elasticity in patients with lipedema of the Moncorps "rusticanus" type]. Phlebologie 1984; 37:315-319.
  7. Stanton AW, Svensson WE, Mellor RH, Peters AM, Levick JR, Mortimer PS. Differences in lymph drainage between swollen and non-swollen regions in arms with breast-cancer-related lymphoedema. Clin Sci (Lond) 2001; 101:131-140.
  8. Kumar B, Lenert P. Joint Hypermobility Syndrome: Recognizing a Commonly Overlooked Cause of Chronic Pain. Am J Med 2017; 130:640-647. doi: 610.1016/j.amjmed.2017.1002.1013. Epub 2017 Mar 1010.
  9. Tinkle B, Castori M, Berglund B, Cohen H, Grahame R, Kazkaz H, Levy H. Hypermobile Ehlers-Danlos syndrome (a.k.a. Ehlers-Danlos syndrome Type III and Ehlers-Danlos syndrome hypermobility type): Clinical description and natural history. Am J Med Genet C Semin Med Genet 2017; 175:48-69. doi: 10.1002/ajmg.c.31538. Epub 32017 Feb 31531.
  10. Crescenzi R, Marton A, Donahue PMC, Mahany HB, Lants SK, Wang P, Beckman JA, Donahue MJ, Titze J. Tissue Sodium Content is Elevated in the Skin and Subcutaneous Adipose Tissue in Women with Lipedema. Obesity (Silver Spring) 2018; 26:310-317. doi: 310.1002/oby.22090. Epub 22017 Dec 22027.
  11. Kakudo N, Morimoto N, Ogawa T, Taketani S, Kusumoto K. Hypoxia Enhances Proliferation of Human Adipose-Derived Stem Cells via HIF-1a Activation. PLoS One 2015; 10:e0139890. doi: 0139810.0131371/journal.pone.0139890. eCollection 0132015.
  12. Siems W, Grune T, Voss P, Brenke R. Anti-fibrosclerotic effects of shock wave therapy in lipedema and cellulite. Biofactors 2005; 24:275-282.
  13. Harvey NL, Srinivasan RS, Dillard ME, Johnson NC, Witte MH, Boyd K, Sleeman MW, Oliver G. Lymphatic vascular defects promoted by Prox1 haploinsufficiency cause adult-onset obesity. Nat Genet 2005; 37:1072-1081. Epub 2005 Sep 1018.
  14. Priglinger E, Wurzer C, Steffenhagen C, Maier J, Hofer V, Peterbauer A, Nuernberger S, Redl H, Wolbank S, Sandhofer M. The adipose tissue-derived stromal vascular fraction cells from lipedema patients: Are they different? Cytotherapy 2017; 19:849-860. doi: 810.1016/j.jcyt.2017.1003.1073. Epub 2017 Apr 1025.
  15. Amann-Vesti BR, Franzeck UK, Bollinger A. Microlymphatic aneurysms in patients with lipedema. Lymphology 2001; 34:170-175
  16. Stallworth JM, Hennigar GR, Jonsson HT, Jr., Rodriguez O. The chronically swollen painful extremity. A detailed study for possible etiological factors. Journal of the American Medical Association 1974; 228:1656-1659.
  17. Miller NE, Michel CC, Nanjee MN, Olszewski WL, Miller IP, Hazell M, Olivecrona G, Sutton P, Humphreys SM, Frayn KN. Secretion of adipokines by human adipose tissue in vivo: partitioning between capillary and lymphatic transport. American journal of physiology Endocrinology and metabolism 2011; 301:E659-667
  18. Homan EA, Kim YG, Cardia JP, Saghatelian A. Monoalkylglycerol ether lipids promote adipogenesis. J Am Chem Soc 2011; 133:5178-5181. doi: 5110.1021/ja111173c. Epub 112011 Mar 111123.
  19. Suga H, Araki J, Aoi N, Kato H, Higashino T, Yoshimura K. Adipose tissue remodeling in lipedema: adipocyte death and concurrent regeneration. J Cutan Pathol 2009; 3:3
  20. Szel E, Kemeny L, Groma G, Szolnoky G. Pathophysiological dilemmas of lipedema. Med Hypotheses 2014; 83:599-606. doi: 510.1016/j.mehy.2014.1008.1011. Epub 2014 Aug 1023.
  21. AL-Ghadban S, Cromer W, Allen M, Ussery C, Badowski M, Harris D, Herbst KL. Dilated Blood and Lymphatic Microvessels, Angiogenesis, Increased Macrophages, and Adipocyte Hypertrophy in Lipedema Thigh Skin and Fat Tissue. Journal of Obesity 2019;
  22. Weitman ES, Aschen SZ, Farias-Eisner G, Albano N, Cuzzone DA, Ghanta S, Zampell JC, Thorek D, Mehrara BJ. Obesity impairs lymphatic fluid transport and dendritic cell migration to lymph nodes. PLoS One 2013; 8:e70703. doi: 70710.71371/journal.pone.0070703. eCollection 0072013.
  23. Brautigam P, Foldi E, Schaiper I, Krause T, Vanscheidt W, Moser E. Analysis of lymphatic drainage in various forms of leg edema using two compartment lymphoscintigraphy. Lymphology 1998; 31:43-55
  24. Witte CL, Witte MH, Unger EC, Williams WH, Bernas MJ, McNeill GC, Stazzone AM. Advances in imaging of lymph flow disorders. Radiographics 2000; 20:1697-1719.
  25. Gould DJ, El-Sabawi B, Goel P, Badash I, Colletti P, Patel KM. Uncovering Lymphatic Transport Abnormalities in Patients with Primary Lipedema. J Reconstr Microsurg 2019; 23:0039-1697904
  26. Brorson H, Fagher B. [Dercum's disease. Fatty tissue rheumatism caused by immune defense reaction?]. Lakartidningen 1996; 93:1430, 1433-1436
  27. Roldan M, Macias-Gonzalez M, Garcia R, Tinahones FJ, Martin M. Obesity short-circuits stemness gene network in human adipose multipotent stem cells. FASEB J 2011; 25:4111-4126. doi: 4110.1096/fj.4110-171439. Epub 172011 Aug 171416.
  28. Bauer AT, D VL, Lossagk K, Hopfner U, Kirsch M, Moog P, Bauer H, Machens HG, Schmauss D. Adipose stem cells from lipedema and control adipose tissue respond differently to adipogenic stimulation in vitro. Plast Reconstr Surg 2019; 6:0000000000005918
  29. Piche ME, Vasan SK, Hodson L, Karpe F. Relevance of human fat distribution on lipid and lipoprotein metabolism and cardiovascular disease risk. Curr Opin Lipidol 2018; 29:285-292. doi: 210.1097/MOL.0000000000000522.
  30. Torre YS, Wadeea R, Rosas V, Herbst KL. Lipedema: friend and foe. Horm Mol Biol Clin Investig 2018; 33(1)./j/hmbci.ahead-of-print/hmbci-2017-0076/hmbci-2017-0076.xml. doi: 2010.1515/hmbci-2017-0076.
  31. Nemes A, Kormanyos A, Domsik P, Kalapos A, Kemeny L, Forster T, Szolnoky G. Left ventricular rotational mechanics differ between lipedema and lymphedema: Insights from the three-dimensional speckle tracking echocardiographic MAGYAR-path study. Lymphology 2018; 51:102-108.
  32. Kozel BA, Danback JR, Waxler JL, Knutsen RH, de Las Fuentes L, Reusz GS, Kis E, Bhatt AB, Pober BR. Williams syndrome predisposes to vascular stiffness modified by antihypertensive use and copy number changes in NCF1. Hypertension 2014; 63:74-79. doi: 10.1161/HYPERTENSIONAHA.1113.02087. Epub 02013 Oct 02014.
  33. Witte CL, Witte MH, Unger EC, Williams WH, Bernas MJ, McNeill GC, Stazzone AM. Advances in Imaging of Lymph Flow Disorders. RadioGraphics 2000; 20:1697–1719
  34. Forner-Cordero I, Olivan-Sasot P, Ruiz-Llorca C, Munoz-Langa J. Lymphoscintigraphic findings in patients with lipedema. Rev Esp Med Nucl Imagen Mol 2018; 37:341-348. doi: 310.1016/j.remn.2018.1006.1008. Epub 2018 Aug 1028.
  35. Bilancini S, Lucchi M, Tucci S, Eleuteri P. Functional lymphatic alterations in patients suffering from lipedema. Angiology 1995; 46:333-339
  36. Tiedjen KV, Knorz S. Different Methods of diagnostic imaging in lymphedema, lipedema and venous disorders: Indirect lymphography, xeroradiography, CT and isotope lymphography. In: Cluzan RV, Pecking AP, Lokiec FM, eds. Progress in lymphology: XIII, Int. Congress of Lymphology. Amsterdam: Elsevier Science Publishers B.V.; 1992.
  37. Dietzel R, Reisshauer A, Jahr S, Calafiore D, Armbrecht G. Body composition in lipoedema of the legs using Dual-Energy-X-Ray-Absorptiometry - a case control study. Br J Dermatol 2015; 1:13697
  38. Dimakakos PB, Stefanopoulos T, Antoniades P, Antoniou A, Gouliamos A, Rizos D. MRI and ultrasonographic findings in the investigation of lymphedema and lipedema. Int Surg 1997; 82:411-416
  39. Iker E, Mayfield CK, Gould DJ, Patel KM. Characterizing Lower Extremity Lymphedema and Lipedema with Cutaneous Ultrasonography and an Objective Computer-Assisted Measurement of Dermal Echogenicity. Lymphat Res Biol 2019; 7
  40. Hirsch T, Schleinitz J, Marshall M, Faerber G. Is the differential diagnosis of lipoedema by means of high-resolution ultrasonography possible? Phlebologie 2018; 47
  41. Halk AB, Damstra RJ. First Dutch guidelines on lipedema using the international classification of functioning, disability and health. Phlebology 2016; 12:0268355516639421
  42. Lohrmann C, Foeldi E, Langer M. MR imaging of the lymphatic system in patients with lipedema and lipo-lymphedema. Microvasc Res 2009; 77:335-339. Epub 2009 Jan 2027.
  43. Machnik A, Neuhofer W, Jantsch J, Dahlmann A, Tammela T, Machura K, Park JK, Beck FX, Muller DN, Derer W, Goss J, Ziomber A, Dietsch P, Wagner H, van Rooijen N, Kurtz A, Hilgers KF, Alitalo K, Eckardt KU, Luft FC, Kerjaschki D, Titze J. Macrophages regulate salt-dependent volume and blood pressure by a vascular endothelial growth factor-C-dependent buffering mechanism. Nat Med 2009; 15:545-552. doi: 510.1038/nm.1960. Epub 2009 May 1033.
  44. Allison MA, Cushman M, Callas PW, Denenberg JO, Jensky NE, Criqui MH. Adipokines are associated with lower extremity venous disease: the San Diego population study. J Thromb Haemost 2010; 8:1912-1918. doi: 1910.1111/j.1538-7836.2010.03941.x.
  45. Greene AK. Diagnosis and Management of Obesity-Induced Lymphedema. Plast Reconstr Surg 2016; 138:111e-118e. doi: 110.1097/PRS.0000000000002258.
  46. Mortimer PS. Implications of the lymphatic system in CVI-associated edema. Angiology 2000; 51:3-7. doi: 10.1177/000331970005100102.
  47. Ibarra M, Eekema A, Ussery C, Neuhardt D, Garby K, Herbst KL. Subcutaneous adipose tissue therapy reduces fat by dual X-ray absorptiometry scan and improves tissue structure by ultrasound in women with lipoedema and Dercum disease. Clin Obes 2018; 8:398-406. doi: 310.1111/cob.12281. Epub 12018 Sep 12224.
  48. Dudek JE, Bialaszek W, Ostaszewski P, Smidt T. Depression and appearance-related distress in functioning with lipedema. Psychol Health Med 2018; 3:1-8
  49. Foldi E, Foldi M. Lipedema. In: Foldi M, Foldi E, eds. Foldi's Textbook of Lymphology. Munich, Germany: Elsevier GmbH; 2006:420.
  50. Stutz JJ. All about lipedema. human med AG 2015;
  51. Bulbena A, Baeza-Velasco C, Bulbena-Cabre A, Pailhez G, Critchley H, Chopra P, Mallorqui-Bague N, Frank C, Porges S. Psychiatric and psychological aspects in the Ehlers-Danlos syndromes. Am J Med Genet C Semin Med Genet 2017; 175:237-245. doi: 210.1002/ajmg.c.31544. Epub 32017 Feb 31510.
  52. Schiltz D, Anker A, Ortner C, Tschernitz S, Koller M, Klein S, Felthaus O, Schreml J, Schreml S, Prantl L. Multiple Symmetric Lipomatosis: New Classification System Based on the Largest German Patient Cohort. Plast Reconstr Surg Glob Open 2018; 6:e1722. doi: 1710.1097/GOX.0000000000001722. eCollection 0000000000002018 Apr.
  53. Wold LE, Hines EA, Jr., Allen EV. Lipedema of the legs; a syndrome characterized by fat legs and edema. Ann Intern Med 1951; 34:1243-1250.
  54. Hodson S, Eaton SE. Lipoedema management: Gaps in our knowledge. Journal of Lymphoedema 2013; 8:30-34
  55. Partsch H, Stoberl C, Urbanek A, Wenzel-Hora BI. Clinical use of indirect lymphography in different forms of leg edema. Lymphology 1988; 21:152-160
  56. Ehrlich C, Iker E, Herbst KL, Kahn LA, Sears DD, Kenyon M. Lymphedema and Lipedema Nutrition Guide.Foods, vitamins, minerals, and supplements. San Francisco: Lymph Notes.
  57. Reich-Schupke S, Schmeller W, Brauer WJ, Cornely ME, Faerber G, Ludwig M, Lulay G, Miller A, Rapprich S, Richter DF, Schacht V, Schrader K, Stucker M, Ure C. S1 guidelines: Lipedema. J Dtsch Dermatol Ges 2017; 15:758-767. doi: 710.1111/ddg.13036.
  58. Khalaf MM, Ashem HA. Suggested physical therapy protocol for reduction of lipomatosis dolorosa of the legs. Egyptian Journal of Medical Human Genetics 2013; 14:103-108
  59. van Esch-Smeenge J, Damstra RJ, Hendrickx AA. Muscle strength and functional exercise capacity in patients with lipoedema and obesity: a comparative study. Journal of Lymphoedema 2017; 12:27-31
  60. Westermann S, Rief W, Euteneuer F, Kohlmann S. Social exclusion and shame in obesity. Eat Behav 2015; 17:74-6.:10.1016/j.eatbeh.2015.1001.1001. Epub 2015 Jan 1010.
  61. Sandhofer M, Schauer P, Sandhofer M, Anderhuber F. Lipödem. Journal für Ästhetische Chirurgie 2017; 10:61-70
  62. Heitink MV, Schurink GWH, de Pont CDJM, van Kroonenburgh MJPG, Veraart JCJM. Lymphedema after Greater Saphenous Vein Surgery. EJVES Extra 2009; 18:41-43
  63. Bast JH, Ahmed L, Engdahl R. Lipedema in patients after bariatric surgery. Surg Obes Relat Dis 2016; 12:1131-1132. doi: 1110.1016/j.soard.2016.1104.1013. Epub 2016 Apr 1114.
  64. Neutze D, Roque J. Clinical Evaluation of Bleeding and Bruising in Primary Care. Am Fam Physician 2016; 93:279-286.
  65. Rapprich S, Dingler A, Podda M. Liposuction is an effective treatment for lipedema-results of a study with 25 patients. J Dtsch Dermatol Ges 2011; 9:33-40. doi: 10.1111/j.1610-0387.2010.07504.x. Epub 02010 Sep 07507.
  66. Schmeller W, Meier-Vollrath I. Tumescent liposuction: a new and successful therapy for lipedema. J Cutan Med Surg 2006; 10:7-10
  67. Stutz JJ, Krahl D. Water jet-assisted liposuction for patients with lipoedema: histologic and immunohistologic analysis of the aspirates of 30 lipoedema patients. Aesthetic Plast Surg 2009; 33:153-162. doi: 110.1007/s00266-00008-09214-y. Epub 02008 Jul 00229.
  68. Wollina U, Goldman A, Heinig B. Microcannular tumescent liposuction in advanced lipedema and Dercum's disease. G 145:151-159.
  69. Gadelha Alcidarta dR, de Miranda Leão TL. Rule of four: a simple and safe formula for tumescent anesthesia in dermatologic surgical procedures. Surgical & Cosmetic Dermatology 2009; 1:99-102
  70. Sattler G, Rapprich S, Hagedorn M. Tumeszenz-Lokalanästhesie – Untersuchung zur Pharmakokinetik von Prilocain. Zeitschrift fur Hautkrankheiten 1997; 7:522-525
  71. Rapprich S, Baum S, Kaak I, Kottmann T, Podda M. Treatment of lipoedema using liposuction. Results of our own surveys. Phlebologie 2015; 44:121-132
  72. Pollock H, Forman S, Pollock T, Raccasi M. Conscious sedation/local anesthesia in the office-based surgical and procedural facility. Clin Plast Surg 2013; 40:383-388. doi: 310.1016/j.cps.2013.1004.1014. Epub 2013 May 1023.
  73. Amron D. 2016 Liposuction Panel. Living with Lipedema and Dealing with Dercum's Disease,; 2016; St. Louis, Missouri.
  74. Hattori J, Yamakage M, Seki S, Okazaki K, Namiki A. Inhibitory effects of the anesthetics propofol and sevoflurane on spontaneous lymphatic vessel activity in rats. Anesthesiology 2004; 101:687-694.
  75. Takeshita T, Morio M, Kawahara M, Fujii K. Halothane-induced changes in contractions of mesenteric lymphatics of the rat. Lymphology 1988; 21:128-130.
  76. McHale NG, Thornbury KD. The effect of anesthetics on lymphatic contractility. Microvasc Res 1989; 37:70-76.
  77. Quin JW, Shannon AD. The effect of anaesthesia and surgery on lymph flow, protein and leucocyte concentration in lymph of the sheep. Lymphology 1975; 8:126-135.
  78. Baumgartner A, Hueppe M, Schmeller W. Long-term benefit of liposuction in patients with lipoedema.A follow-up study after an average of 4 and 8 years. Br J Dermatol 2015; 17:14289
  79. Rapprich S, Loehnert M, Hagedorn M. Therapy of lipoedema syndrome by liposuction under tumescent local anaesthesia. Ann Dermatol Venereol 2002; 129:1S711-
  80. Szolnoky G, Borsos B, Barsony K, Balogh M, Kemeny L. Complete decongestive physiotherapy with and without pneumatic compression for treatment of lipedema: a pilot study. Lymphology 2008; 41:40-44
  81. Szolnoky G, Nagy N, Kovacs RK, Dosa-Racz E, Szabo A, Barsony K, Balogh M, Kemeny L. Complex decongestive physiotherapy decreases capillary fragility in lipedema. Lymphology 2008; 41:161-166.
  82. Szolnoky G, Varga E, Varga M, Tuczai M, Dosa-Racz E, Kemeny L. Lymphedema treatment decreases pain intensity in lipedema. Lymphology 2011; 44:178-182.
  83. Belgrado JP, Vandermeeren L, Vankerckhove S, Valsamis JB, Malloizel-Delaunay J, Moraine JJ, Liebens F. Near-Infrared Fluorescence Lymphatic Imaging to Reconsider Occlusion Pressure of Superficial Lymphatic Collectors in Upper Extremities of Healthy Volunteers. Lymphat Res Biol 2016; 14:70-77. doi: 10.1089/lrb.2015.0040. Epub 2016 May 1011.
  84. Iliff JJ, Wang M, Liao Y, Plogg BA, Peng W, Gundersen GA, Benveniste H, Vates GE, Deane R, Goldman SA, Nagelhus EA, Nedergaard M. A paravascular pathway facilitates CSF flow through the brain parenchyma and the clearance of interstitial solutes, including amyloid beta. Sci Transl Med 2012; 4:147ra111. doi: 110.1126/scitranslmed.3003748.
  85. Macdonald JM, Sims N, Mayrovitz HN. Lymphedema, lipedema, and the open wound: the role of compression therapy. Surg Clin North Am 2003; 83:639-658.
  86. Schneider M, Conway EM, Carmeliet P. Lymph makes you fat. Nat Genet 2005; 37:1023-1024.
  87. Herbst KL, Ussery C, Eekema A. Pilot study: whole body manual subcutaneous adipose tissue (SAT) therapy improved pain and SAT structure in women with lipedema. LID - 10.1515/hmbci-2017-0035 [doi] LID - /j/hmbci.ahead-of-print/hmbci-2017-0035/hmbci-2017-0035.xml [pii]. Horm Mol Biol Clin Investig 2017;
  88. Yan B-h, Peng Q-s, Wei Q-h, Feng F. The effect of meridian massage on 8M, 8MI, WC and HC in simple obesity patients: a randomized controlled trial. World Journal ofAcupuncture-Moxibustion (WJAM) 2014; 24:6-9, 50
  89. Moyer-Mileur LJ, Haley S, Slater H, Beachy J, Smith SL. Massage Improves Growth Quality by Decreasing Body Fat Deposition in Male Preterm Infants. The Journal of pediatrics 2013; 162:490-495
  90. Sevier TL, Stegink-Jansen CW. Astym treatment vs. eccentric exercise for lateral elbow tendinopathy: a randomized controlled clinical trial. PeerJ 2015; 3:e967.:10.7717/peerj.7967. eCollection 2015.
  91. Lee JH, Lee DK, Oh JS. The effect of Graston technique on the pain and range of motion in patients with chronic low back pain. J Phys Ther Sci 2016; 28:1852-1855. doi: 1810.1589/jpts.1828.1852. Epub 2016 Jun 1828.
  92. Braun M, Schwickert M, Nielsen A, Brunnhuber S, Dobos G, Musial F, Ludtke R, Michalsen A. Effectiveness of traditional Chinese "gua sha" therapy in patients with chronic neck pain: a randomized controlled trial. Pain Med 2011; 12:362-369. doi: 310.1111/j.1526-4637.2011.01053.x. Epub 02011 Jan 01028.
  93. Mehta P, Dhapte V. Cupping therapy: A prudent remedy for a plethora of medical ailments. J Tradit Complement Med 2015; 5:127-134. doi: 110.1016/j.jtcme.2014.1011.1036. eCollection 2015 Jul.
  94. Ai JW, Liu JT, Pei SD, Liu Y, Li DS, Lin HM, Pei B. The effectiveness of pressure therapy (15-25 mmHg) for hypertrophic burn scars: A systematic review and meta-analysis. Sci Rep 2017; 7:40185.:10.1038/srep40185.
  95. Phelan SM, Burgess DJ, Yeazel MW, Hellerstedt WL, Griffin JM, van Ryn M. Impact of weight bias and stigma on quality of care and outcomes for patients with obesity. Obes Rev 2015; 16:319-326. doi: 310.1111/obr.12266. Epub 12015 Mar 12265.
  96. Dudek JE, Bialaszek W, Ostaszewski P. Quality of life in women with lipoedema: a contextual behavioral approach. Qual Life Res 2016; 25:401-408. doi: 410.1007/s11136-11015-11080-x. Epub 12015 Jul 11128.
  97. McHale NG, Allen JM, Iggulden HL. Mechanism of alpha-adrenergic excitation in bovine lymphatic smooth muscle. Am J Physiol 1987; 252:H873-878.
  98. Herbst KL, Abu-Zaid L, Fazel MT. Question-Based Self-Reported Experience of Patients with Subcutaneous Adipose Tissue (SAT) Disease Prescribed Sympathomimetic Amines. Medical Research Archives 2019; 7:1-17
  99. Feldo M, Wozniak M, Wojciak-Kosior M, Sowa I, Kot-Wasik A, Aszyk J, Bogucki J, Zubilewicz T, Bogucka-Kocka A. Influence of Diosmin Treatment on the Level of Oxidative Stress Markers in Patients with Chronic Venous Insufficiency. Oxid Med Cell Longev 2018; 2018:2561705.:10.1155/2018/2561705. eCollection 2562018.
  100. Li X, Li J, Wang L, Li A, Qiu Z, Qi LW, Kou J, Liu K, Liu B, Huang F. The role of metformin and resveratrol in the prevention of hypoxia-inducible factor 1alpha accumulation and fibrosis in hypoxic adipose tissue. Br J Pharmacol 2016; 173:2001-2015. doi: 2010.1111/bph.13493. Epub 12016 May 13415.
  101. Biondo LA, Batatinha HA, Souza CO, Teixeira AAS, Silveira LS, Alonso-Vale MI, Oyama LM, Alves MJ, Seelaender M, Neto JCR. Metformin Mitigates Fibrosis and Glucose Intolerance Induced by Doxorubicin in Subcutaneous Adipose Tissue. Front Pharmacol 2018; 9:452.:10.3389/fphar.2018.00452. eCollection 02018.
  102. Conley SM, Bruhn RL, Morgan PV, Stamer WD. Selenium's effects on MMP-2 and TIMP-1 secretion by human trabecular meshwork cells. Invest Ophthalmol Vis Sci 2004; 45:473-479.
  103. Lewin MH, Arthur JR, Riemersma RA, Nicol F, Walker SW, Millar EM, Howie AF, Beckett GJ. Selenium supplementation acting through the induction of thioredoxin reductase and glutathione peroxidase protects the human endothelial cell line EAhy926 from damage by lipid hydroperoxides. Biochim Biophys Acta 2002; 1593:85-92. doi: 10.1016/s0167-4889(1002)00333-00336.
  104. Horvathova M, Jahnova E, Gazdik F. Effect of selenium supplementation in asthmatic subjects on the expression of endothelial cell adhesion molecules in culture. Biol Trace Elem Res 1999; 69:15-26
  105. Kiremidjian-Schumacher L, Roy M, Glickman R, Schneider K, Rothstein S, Cooper J, Hochster H, Kim M, Newman R. Selenium and immunocompetence in patients with head and neck cancer. Biol Trace Elem Res 2000; 73:97-111
  106. Micke O, Bruns F, Schäfer U, Kisters K, Hesselmann S, Willich N. Selenium in the treatment of acute and chronic lymphedema. Trace Elements and Electrolytes 2000; 17:206-209
  107. Kasseroller RG, Schrauzer GN. Treatment of secondary lymphedema of the arm with physical decongestive therapy and sodium selenite: a review. Am J Ther 2000; 7:273-279
  108. Medicine Io. Dietary Reference Intakes for Vitamin C, Vitamin E, Selenium, and Carotenoids. Washington, DC: The National Academies Press.
  109. Prabhu KS, Lei XG. Selenium. Adv Nutr 2016; 7:415-417. doi: 410.3945/an.3115.010785. Print 012016 Mar.
  110. Nourollahi S, Mondry TE, Herbst KL. Bucher’s Broom and Selenium Improve Lipedema: A Retrospective Case Study Alternative and Integrative Medicine 2013; 2:1-7
  111. Rabbiosi G, Borroni G, Scuderi N. Familial multiple lipomatosis. Acta Derm Venereol 1977; 57:265-267.
  112. Mohar N. Familial multiple lipomatosis. Acta Derm Venereol 1980; 60:509-513.
  113. Tana C, Tchernev G. Images in clinical medicine. Familial multiple lipomatosis. N Engl J Med 2014; 371:1237. doi: 1210.1056/NEJMicm1316241.
  114. Pack GT, Ariel IM. Tumors of the Soft Somatic Tissues: A Clinical Treatise. New York: Hoeber-Harper.
  115. Campen R, Mankin H, Louis DN, Hirano M, Maccollin M. Familial occurrence of adiposis dolorosa. J Am Acad Dermatol 2001; 44:132-136
  116. Ware R, Mane A, Saini S, Saini N. Familial multiple lipomatosis—a rare syndrome diagnosed on FNAC. International Journal of Medical Science and Public Health 2016; 5:367-369
  117. Reddy N, Malipatil B, Kumar S. A rare case of familial multiple subcutaneous lipomatosis with novel PALB2 mutation and increased predilection to cancers. Hematol Oncol Stem Cell Ther 2016; 9:154-156. doi: 110.1016/j.hemonc.2016.1001.1001. Epub 2016 Jan 1027.
  118. Singh G, Jialal I. Multiple Endocrine Neoplasia Type 1 (MEN I, Wermer Syndrome). 2019;
  119. Genuardi M, Klutz M, Devriendt K, Caruso D, Stirpe M, Lohmann DR. Multiple lipomas linked to an RB1 gene mutation in a large pedigree with low penetrance retinoblastoma. Eur J Hum Genet 2001; 9:690-694.
  120. Liaw D, Marsh DJ, Li J, Dahia PL, Wang SI, Zheng Z, Bose S, Call KM, Tsou HC, Peacocke M, Eng C, Parsons R. Germline mutations of the PTEN gene in Cowden disease, an inherited breast and thyroid cancer syndrome. Nat Genet 1997; 16:64-67. doi: 10.1038/ng0597-1064.
  121. Yehia L, Ni Y, Sesock K, Niazi F, Fletcher B, Chen HJL, LaFramboise T, Eng C. Unexpected cancer-predisposition gene variants in Cowden syndrome and Bannayan-Riley-Ruvalcaba syndrome patients without underlying germline PTEN mutations. PLoS Genet 2018; 14:e1007352. doi: 1007310.1001371/journal.pgen.1007352. eCollection 1002018 Apr.
  122. Bracaglia R, D'Ettorre M, Gentileschi S, Mingrone G, Tambasco D. Multiple lipomatosis after stem cell trasplant and chemotherapy: a case report. Eur Rev Med Pharmacol Sci 2014; 18:413-415.
  123. Cronin PA, Myers E, Redmond HP, O'Reilly S, Kirwan WO. Lipomatosis: an unusual side-effect of cytotoxic chemotherapy? Acta Derm Venereol 2010; 90:303-304. doi: 310.2340/00015555-00010823.
  124. Tsao H, Sober AJ. Multiple lipomatosis in a patient with familial atypical mole syndrome. Br J Dermatol 1998; 139:1118-1119.
  125. Djuric-Stefanovic A, Ebrahimi K, Sisevic J, Saranovic D. Gastroduodenal Lipomatosis in Familial Multiple Lipomatosis. Med Princ Pract 2017; 26:189-191. doi: 110.1159/000454714. Epub 000452016 Nov 000454724.
  126. Sayar I, Demirtas L, Gurbuzel M, Isik A, Peker K, Gulhan B. Familial multiple lipomas coexisting with celiac disease: a case report. J Med Case Rep 2014; 8:309.:10.1186/1752-1947-1188-1309.
  127. Arabadzhieva E, Yonkov A, Bonev S, Bulanov D, Taneva I, Ivanova V, Dimitrova V. A rare combination between familial multiple lipomatosis and extragastrointestinal stromal tumor. Int J Surg Case Rep 2015; 14:117-20.:10.1016/j.ijscr.2015.1007.1027. Epub 2015 Jul 1031.
  128. Nunes JC, Martins RF, Bastos A, Claudino LS, Guarnieri R, Lima Dde C, Schroeder HK, Lin K, Walz R. Brain lipoma, corpus callosum hypoplasia and polymicrogyria in familial multiple lipomatosis. Clin Neurol Neurosurg 2013; 115:1157-1159. doi: 1110.1016/j.clineuro.2012.1109.1023. Epub 2012 Oct 1130.
  129. Oktenli C, Gul D, Deveci MS, Saglam M, Upadhyaya M, Thompson P, Consoli C, Kocar IH, Pilarski R, Zhou XP, Eng C. Unusual features in a patient with neurofibromatosis type 1: multiple subcutaneous lipomas, a juvenile polyp in ascending colon, congenital intrahepatic portosystemic venous shunt, and horseshoe kidney. Am J Med Genet A 2004; 127:298-301
  130. Morelli A, Falchetti A, Weinstein L, Fabiani S, Tomassetti P, Enzi G, Carraro R, Bordi C, Tonelli F, Brandi ML. RFLP analysis of human chromosome 11 region q13 in multiple symmetric lipomatosis and multiple endocrine neoplasia type 1-associated lipomas. Biochem Biophys Res Commun 1995; 207:363-368. doi: 310.1006/bbrc.1995.1196.
  131. Bianchi M, Saletti V, Micheli R, Esposito S, Molinaro A, Gagliardi S, Orcesi S, Cereda C. Legius Syndrome: two novel mutations in the SPRED1 gene. Hum Genome Var 2015; 2:15051.:10.1038/hgv.2015.1051. eCollection 2015.
  132. D'Ettorre M, Gniuli D, Guidone C, Bracaglia R, Tambasco D, Mingrone G. Insulin sensitivity in Familial Multiple Lipomatosis. Eur Rev Med Pharmacol Sci 2013; 17:2254-2256.
  133. Inampudi P, Jacobson JA, Fessell DP, Carlos RC, Patel SV, Delaney-Sathy LO, van Holsbeeck MT. Soft-tissue lipomas: accuracy of sonography in diagnosis with pathologic correlation. Radiology 2004; 233:763-767. doi: 710.1148/radiol.2333031410. Epub 2333032004 Oct 2333031414.
  134. Coran A, Ortolan P, Attar S, Alberioli E, Perissinotto E, Tosi AL, Montesco MC, Rossi CR, Tropea S, Rastrelli M, Stramare R. Magnetic Resonance Imaging Assessment of Lipomatous Soft-tissue Tumors. In Vivo 2017; 31:387-395. doi: 310.21873/invivo.11071.
  135. O'Donnell PW, Griffin AM, Eward WC, Sternheim A, White LM, Wunder JS, Ferguson PC. Can Experienced Observers Differentiate between Lipoma and Well-Differentiated Liposarcoma Using Only MRI? Sarcoma 2013; 2013:6
  136. Wolfe SW, Bansal M, Healey JH, Ghelman B. Computed tomographic evaluation of fatty neoplasms of the extremities. A clinical, radiographic, and histologic review of cases. Orthopedics 1989; 12:1351-1358.
  137. Cherry hemangiomas and lipomas with a peculiar distribution. Journal of the American Academy of Dermatology 2017; 76:AB68
  138. Self TH, Akins D. Dramatic reduction in lipoma associated with statin therapy. Journal of the American Academy of Dermatology 2008; 58:S30-S31
  139. Al-basti HA, El-Khatib HA. The use of suction-assisted surgical extraction of moderate and large lipomas: long-term follow-up. Aesthetic Plast Surg 2002; 26:114-117.
  140. Gerut ZE. Abstract: The Enzymatic Dissolution of Human Fat. Plast Reconstr Surg Glob Open 2017; 5:66-67
  141. Mlosek RK, Malinowska S, Wozniak W. Lipoma removal using a high-frequency ultrasound-guided injection of a Class III CE-marked device-Empirical findings. J Cosmet Dermatol 2019; 18:469-473. doi: 410.1111/jocd.12681. Epub 12018 Jul 12619.
  142. Boyer M, Monette S, Nguyen A, Zipp T, Aughenbaugh WD, Nimunkar AJ. A review of techniques and procedures for lipoma treatment. Clincial Dermatology 2015; 3:105-112
  143. Kanamori Y, Takezoe T, Takahashi M, Nakano N, Matsuoka K, Osaku M. A case of an 8-year-old boy who was strongly suspected of suffering from familial angiolipomatosis. Journal of Pediatric Surgery Case Reports 2015; 3:255-256
  144. Yeo ED, Chung BM, Kim EJ, Kim WT. Infiltrating angiolipoma of the foot: magnetic resonance imaging features and review of the literature. Skeletal Radiol 2018; 47:859-864. doi: 810.1007/s00256-00017-02870-00258. Epub 02018 Jan 00210.
  145. Nissen A, Yi F. Rare cause of colonic intussusception in an adult. BMJ Case Rep 2017; 2017.:bcr-2017-221976. doi: 221910.221136/bcr-222017-221976.
  146. Wu Z, Wan H, Shi M, Li M, Wang Z, Yang C, Gao W, Li Q. Bronchoscopic resection of bronchial angiolipoma: A rare case report. Mol Clin Oncol 2016; 5:850-852. doi: 810.3892/mco.2016.1069. Epub 2016 Nov 3891.
  147. Ilyas G, Turgut A, Ayaz D, Kalenderer O. Intraarticular Giant Size Angiolipoma of the Knee Causing Lateral Patellar Dislocation. Balkan Med J 2016; 33:691-694. doi: 610.5152/balkanmedj.2016.141269. Epub 142016 Nov 141261.
  148. Srivastava A, Gupta N, Joice GA, Wright EJ. Management and Excision of a 15 cm Paratesticular Angiolipoma. Urol Case Rep 2017; 15:8-10.:10.1016/j.eucr.2017.1007.1004. eCollection 2017 Nov.
  149. Naversen DN, Trask DM, Watson FH, Burket JM. Painful tumors of the skin: "LEND AN EGG". J Am Acad Dermatol 1993; 28:298-300.
  150. Clayman E, King K, Harrington MA. Corticosteroid-Associated Angiolipomatosis. Eplasty 2017; 17:ic9. eCollection 2017.
  151. Namba M, Kohda M, Mimura S, Nakagawa S, Ueki H. Angiolipoma in brothers. J Dermatol 1977; 4:255-257.
  152. Howard WR, Helwig EB. Angiolipoma. Arch Dermatol 1960; 82:924-931.
  153. Garib G, Siegal GP, Andea AA. Autosomal-dominant familial angiolipomatosis. Cutis 2015; 95:E26-29.
  154. Abbasi NR, Brownell I, Fangman W. Familial multiple angiolipomatosis Dermatology Online Journal 2007;13(1):3. http://dermatology.cdlib.org/131/cases/NYUcases/101805_5.html#9#9. Accessed 11/19/2007
  155. Hapnes SA, Boman H, Skeie SO. Familial angiolipomatosis. Clin Genet 1980; 17:202-208.
  156. Wilke J, Schleip R, Klingler W, Stecco C. The Lumbodorsal Fascia as a Potential Source of Low Back Pain: A Narrative Review. Biomed Res Int 2017:1-6
  157. Yoshida K, Ito H, Furuya K, Ukichi T, Noda K, Kurosaka D. Angiogenesis and VEGF-expressing cells are identified predominantly in the fascia rather than in the muscle during the early phase of dermatomyositis. Arthritis Res Ther 2017; 19:272. doi: 210.1186/s13075-13017-11481-z.
  158. Su X, Lyu Y, Wang W, Zhang Y, Li D, Wei S, Du C, Geng B, Sztalryd C, Xu G. Fascia Origin of Adipose Cells. Stem Cells 2016; 34:1407-1419. doi: 1410.1002/stem.2338. Epub 2016 Mar 1407.
  159. Panagopoulos I, Gorunova L, Andersen K, Lobmaier I, Bjerkehagen B, Heim S. Consistent Involvement of Chromosome 13 in Angiolipoma. Cancer Genomics Proteomics 2018; 15:61-65. doi: 10.21873/cgp.20065.
  160. Dekker I, van der Leest M, van Rijk MC, Gerritsen WR, Arens AIJ. 68Ga-PSMA Uptake in Angiolipoma. Clin Nucl Med 2018; 43:757-758. doi: 710.1097/RLU.0000000000002236.
  161. Kitagawa Y, Miyamoto M, Konno S, Makino A, Maruyama G, Takai S, Higashi N. Subcutaneous angiolipoma: magnetic resonance imaging features with histological correlation. J Nippon Med Sch 2014; 81:313-319.
  162. Bang M, Kang BS, Hwang JC, Weon YC, Choi SH, Shin SH, Kwon WJ, Hwang CM, Lee SY. Ultrasonographic analysis of subcutaneous angiolipoma. Skeletal Radiol 2012; 41:1055-1059. doi: 1010.1007/s00256-00011-01309-x. Epub 02011 Nov 00258.
  163. Kaneko T, Tokushige H, Kimura N, Moriya S, Toda K. The treatment of multiple angiolipomas by liposuction surgery. J Dermatol Surg Oncol 1994; 20:690-692.
  164. Horiuchi K, Yamada T, Sakai K, Okawa A, Arai Y. Hemorrhagic Sudden Onset of Spinal Epidural Angiolipoma. Case Rep Orthop 2018; 2018:5231931.:10.1155/2018/5231931. eCollection 5232018.
  165. Lacour M, Gilard V, Marguet F, Curey S, Perez A, Derrey S. Sudden paraplegia due to spontaneous bleeding in a thoracic epidural angiolipoma and literature review. Neurochirurgie 2018; 64:73-75. doi: 10.1016/j.neuchi.2017.1008.1003. Epub 2018 Feb 1019.
  166. Behrendt H, Faes J, Ruzicka T. [Multiple angiolipomas--analgesics therapy with doxepin]. Hautarzt 1992; 43:139-142.
  167. Afrin L, Molderings G. A concise, practical guide to diagnostic assessment for mast cell activation disease. World Journal of Hematology 2014; 3:1-17
  168. Afrin LB, Pohlau D, Raithel M, Haenisch B, Dumoulin FL, Homann J, Mauer UM, Harzer S, Molderings GJ. Mast cell activation disease: An underappreciated cause of neurologic and psychiatric symptoms and diseases. Brain Behav Immun 2015; 50:314-21.:10.1016/j.bbi.2015.1007.1002. Epub 2015 Jul 1018.
  169. Molderings GJ, Afrin LB, Hertfelder HJ, Brettner S. Case Report: Treatment of systemic mastocytosis with sunitinib. 1000Res 2017; 6:2182.:10.12688/f11000research.13343.12681. eCollection 12017.
  170. Afrin LB, Fox RW, Zito SL, Choe L, Glover SC. Successful targeted treatment of mast cell activation syndrome with tofacitinib. Eur J Haematol 2017; 99:190-193. doi: 110.1111/ejh.12893. Epub 12017 May 12893.
  171. Malik F, Ali N, Jafri SIM, Ghani A, Hamid M, Boigon M, Fidler C. Continuous diphenhydramine infusion and imatinib for KIT-D816V-negative mast cell activation syndrome: a case report. J Med Case Rep 2017; 11:119. doi: 110.1186/s13256-13017-11278-13253.
  172. Molderings GJ, Haenisch B, Brettner S, Homann J, Menzen M, Dumoulin FL, Panse J, Butterfield J, Afrin LB. Pharmacological treatment options for mast cell activation disease. Naunyn Schmiedebergs Arch Pharmacol 2016; 389:671-694. doi: 610.1007/s00210-00016-01247-00211. Epub 02016 Apr 00230.
  173. Baldo BA, Pham NH. Histamine-releasing and allergenic properties of opioid analgesic drugs: resolving the two. Anaesth Intensive Care 2012; 40:216-235. doi: 210.1177/0310057X1204000204.
  174. Naviaux RK. Metabolic features and regulation of the healing cycle-A new model for chronic disease pathogenesis and treatment. Mitochondrion 2018; 9:30105-30103
  175. Hansson E, Svensson H, Brorson H. Review of Dercum's disease and proposal of diagnostic criteria, diagnostic methods, classification and management. Orphanet J Rare Dis 2012; 7:23.:10.1186/1750-1172-1187-1123.
  176. Giudiceandrea V. L’adiposis dolorosa (malattia di Dercum). Riv Patol Nerv Ment 1900; V:289-304
  177. Roux J, Vitaut M. Maladie de Dercum (Adiposis dolorosa). Revue Neurol (Paris) 1901; 9:881-888
  178. Anders JM. Adiposis tuberosa simplex. The American Journal of the Medical Sciences (1827-1924) 1908; 135:325-333
  179. Hao D, Olugbodi A, Udechukwu N, Donato AA. Trauma-induced adiposis dolorosa (Dercum's disease). BMJ Case Rep 2018; 2018.:bcr-2017-223869. doi: 223810.221136/bcr-222017-223869.
  180. Herbst KL, Asare-Bediako S. Adiposis Dolorosa is More than Painful Fat. The Endocrinologist 2007; 17:326-344
  181. Stormorken H, Brosstad F, H. S. The fibromyalgia syndrome: A member of the painful lipo[mato]sis family? In: Pederson JA, ed. New Research on Fibromyalgia. New York: Nova Science Publishers, Inc.; 2006.
  182. Dercum FX. A subcutaneous connective-tissue dystrophy of the arms and back, associated with symptoms resembling myxoedema. University Medical Magazine Philadelphia. Vol 11888:140-150.
  183. Rasmussen JC, Herbst KL, Aldrich MB, Darne CD, Tan IC, Zhu B, Guilliod R, Fife CE, Maus EA, Sevick-Muraca EM. An abnormal lymphatic phenotype is associated with subcutaneous adipose tissue deposits in Dercum's disease. Obesity 2014; 9:20836
  184. Fagher B, Monti M, Nilsson-Ehle P, Akesson B. Fat-cell heat production, adipose tissue fatty acids, lipoprotein lipase activity and plasma lipoproteins in adiposis dolorosa. Clin Sci (Lond) 1991; 81:793-798
  185. Bordoni B, Marelli F, Morabito B, Cavallaro F, Lintonbon D. Fascial preadipocytes: another missing piece of the puzzle to understand fibromyalgia? Open Access Rheumatol 2018; 10:27-32.:10.2147/OARRR.S155919. eCollection 152018.
  186. Hansson E, Manjer J, Svensson H, Aberg M, Fagher B, Ekman R, Brorson H. Neuropeptide levels in Dercum's disease (adiposis dolorosa). Reumatismo 2012; 64:134-141. doi: 110.4081/reumatismo.2012.4134.
  187. Herbst KL, Coviello AD, Chang A, Boyle DL. Lipomatosis-associated inflammation and excess collagen may contribute to lower relative resting energy expenditure in women with adiposis dolorosa. Int J Obes (Lond) 2009; 33:1031-1038. Epub 2009 Jul 1021.
  188. Weinberger A Fau - Wysenbeec AJ, Wysenbeec Aj Fau - Pinkhas J, Pinkhas J. Juxta-articular adiposis dolorosa associated with rheumatoid arthritis. Report of 2 cases with good response to local corticosteroid injection.
  189. Baudart P, Cesini J, Marcelli C. Atypical juxta-articular form of Dercum's disease in a patient treated with tocilizumab for rheumatoid arthritis. Joint Bone Spine 2017; 2:30202-30206
  190. Tins BJ, Matthews C, Haddaway M, Cassar-Pullicino VN, Lalam R, Singh J, Tyrrell PN. Adiposis dolorosa (Dercum's disease): MRI and ultrasound appearances. Clin Radiol 2013; 25:00199-00192
  191. Petscavage-Thomas JM, Walker EA, Bernard SA, Bennett J. Imaging findings of adiposis dolorosa vs. massive localized lymphedema. Skeletal Radiol 2015; 13:13
  192. Semelka RC, Ramalho J, Vakharia A, AlObaidy M, Burke LM, Jay M, Ramalho M. Gadolinium deposition disease: Initial description of a disease that has been around for a while. Magn Reson Imaging 2016; 34:1383-1390. doi: 1310.1016/j.mri.2016.1307.1016. Epub 2016 Aug 1313.
  193. Semelka RM. The Odd Fellow Collection of Modern Diseases: Fibromyalgia, Dercum's Disease, Multiple Chemical Sensitivity, Gadolinium Deposition Disease. RICHARD SEMELKA, MD. CONSULTING. Vol 2019. https://www.richardsemelka.com/single-post/2019/02/13/The-Odd-Fellow-Collection-of-Modern-Diseases-Fibromyalgia-Dercums-Disease-Multiple-Chemical-Sensitivity-Gadolinium-Deposition-Disease2019.
  194. Skagen K, Petersen P, Kastrup J, Norgaard T. The regulation of subcutaneous blood flow in patient with Dercum's disease. Acta Derm Venereol 1986; 66:337-339
  195. Cantone M, Lanza G, Pennisi M, Bella R, Schepis C, Siragusa M, Barone R, Ferri R. Prominent neurological involvement in Dercum disease. J Neurol 2017; 264:796-798. doi: 710.1007/s00415-00017-08415-00411. Epub 02017 Feb 00413.
  196. Miraglia E, Visconti B, Bianchini D, Calvieri S, Giustini S. An uncommon association between lipomatous hypertrophy of the interatrial septum (LHIS) and Dercum's disease. Eur J Dermatol 2013; 19:19
  197. Laura DM, Donnino R, Kim EE, Benenstein R, Freedberg RS, Saric M. Lipomatous Atrial Septal Hypertrophy: A Review of Its Anatomy, Pathophysiology, Multimodality Imaging, and Relevance to Percutaneous Interventions. J Am Soc Echocardiogr 2016; 29:717-723. doi: 710.1016/j.echo.2016.1004.1014. Epub 2016 Jun 1017.
  198. Kyllerman M, Brandberg G, Wiklund LM, Mansson JE. Dysarthria, progressive parkinsonian features and symmetric necrosis of putamen in a family with painful lipomas (Dercum disease variant). Neuropediatrics 2002; 33:69-72. doi: 10.1055/s-2002-32366.
  199. Wipf A, Lofgreen S, Miller DD, Farah RS. Novel Use of Deoxycholic Acid for Adiposis Dolorosa (Dercum Disease). Dermatol Surg 2019; 14:0000000000001800
  200. Atkinson RL. Intravenous lidocaine for the treatment of intractable pain of adiposis dolorosa. Int J Obes 1982; 6:351-357.
  201. Desai MJ, Siriki R, Wang D. Treatment of pain in Dercum's disease with Lidoderm (lidocaine 5% patch): a case report. Pain Med 2008; 9:1224-1226. Epub 2008 Mar 1211.
  202. Reggiani M, Errani A, Staffa M, Schianchi S. Is EMLA effective in Dercum's disease? Acta Derm Venereol 1996; 76:170-171
  203. Labuzek K, Liber S, Suchy D, Okopiea BA. A successful case of pain management using metformin in a patient with adiposis dolorosa. Int J Clin Pharmacol Ther 2013; 51:517-524. doi: 510.5414/CP201878.
  204. Tiesmeier J, Warnecke H, Schuppert F. [An uncommon cause of recurrent abdominal pain in a 63-year-old obese woman]. Dtsch Med Wochenschr 2006; 131:434-437.
  205. Parkitny L, Younger J. Reduced Pro-Inflammatory Cytokines after Eight Weeks of Low-Dose Naltrexone for Fibromyalgia. Biomedicines 2017; 5(2).biomedicines5020016. doi: 5020010.5023390/biomedicines5020016.
  206. Lange U, Oelzner P, Uhlemann C. Dercum's disease (Lipomatosis dolorosa): successful therapy with pregabalin and manual lymphatic drainage and a current overview. Rheumatol Int 2008; 29:17-22. Epub 2008 Jul 2005.
  207. Landheer JA, Toonstra J. Dercum's disease: Successful treatment with gabapentin. Nederlands Tijdschrift voor Dermatologie en Venereologie 2009; 19(5):265-267:265-267
  208. Wollina U, Goldman A, Heinig B. Microcannular tumescent liposuction in advanced lipedema and Dercum's disease. G Ital Dermatol Venereol 2010; 145:151-159.
  209. Hansson E, Svensson H, Brorson H. Liposuction may reduce pain in Dercum's disease (adiposis dolorosa). Pain Med 2011; 12:942-952. doi: 910.1111/j.1526-4637.2011.01101.x. Epub 02011 Apr 01111.
  210. Berntorp E, Berntorp K, Brorson H, Frick K. Liposuction in Dercum's disease: impact on haemostatic factors associated with cardiovascular disease and insulin sensitivity. J Intern Med 1998; 243:197-201
  211. Martinenghi S, Caretto A, Losio C, Scavini M, Bosi E. Successful Treatment of Dercum's Disease by Transcutaneous Electrical Stimulation: A Case Report. Medicine (Baltimore) 2015; 94:e950. doi: 910.1097/MD.0000000000000950.
  212. Herbst KL, Rutledge T. Pilot study: rapidly cycling hypobaric pressure improves pain after 5 days in adiposis dolorosa. Journal of Pain Research 2010; 3:147–153
  213. El Ouahabi H, Doubi S, Lahlou K, Boujraf S, Ajdi F. Launois-bensaude syndrome: A benign symmetric lipomatosis without alcohol association. Ann Afr Med 2017; 16:33-34. doi: 10.4103/1596-3519.202082.
  214. Kratz C, Lenard HG, Ruzicka T, Gartner J. Multiple symmetric lipomatosis: an unusual cause of childhood obesity and mental retardation. Eur J Paediatr Neurol 2000; 4:63-67
  215. Enzi G, Busetto L, Ceschin E, Coin A, Digito M, Pigozzo S. Multiple symmetric lipomatosis: clinical aspects and outcome in a long-term longitudinal study. Int J Obes Relat Metab Disord 2002; 26:253-261
  216. Donhauser G, Vieluf D, Ruzicka T, Braun-Falco O. [Benign symmetric Launois-Bensaude type III lipomatosis and Bureau-Barriere syndrome]. Hautarzt 1991; 42:311-314
  217. Ettl T, Gaumann A, Ehrenberg R, Reichert TE, Driemel O. Encapsulated lipomas of the tongue in benign symmetric lipomatosis. J Dtsch Dermatol Ges 2009; 7:441-444. Epub 2009 Jan 2019.
  218. Lopez-Ceres A, Aguilar-Lizarralde Y, Villalobos Sanchez A, Prieto Sanchez E, Valiente Alvarez A. Benign symmetric lipomatosis of the tongue in Madelung's disease. J Craniomaxillofac Surg 2006; 34:489-493. Epub 2006 Dec 2008.
  219. Birnholz JC, Macmillan AS, Jr. Advanced laryngeal compression due to diffuse, symmetric lipomatosis (Madelung's disease). Br J Radiol 1973; 46:245-249.
  220. Laure B, Sury F, Tayeb T, Corre P, Goga D. Launois-Bensaude syndrome involving the orbits. J 39:21-23. Epub 2010 Aug 2014.
  221. Enzi G. Multiple symmetric lipomatosis: an updated clinical report. Medicine (Baltimore) 1984; 63:56-64
  222. Enzi G, Biondetti PR, Fiore D, Mazzoleni F. Computed tomography of deep fat masses in multiple symmetrical lipomatosis. Radiology 1982; 144:121-124
  223. Klopstock T, Naumann M, Schalke B, Bischof F, Seibel P, Kottlors M, Eckert P, Reiners K, Toyka KV, Reichmann H. Multiple symmetric lipomatosis: abnormalities in complex IV and multiple deletions in mitochondrial DNA. Neurology 1994; 44:862-866
  224. Berkovic SF, Andermann F, Shoubridge EA, Carpenter S, Robitaille Y, Andermann E, Melmed C, Karpati G. Mitochondrial dysfunction in multiple symmetrical lipomatosis. Ann Neurol 1991; 29:566-569
  225. Klopstock T, Naumann M, Seibel P, Shalke B, Reiners K, Reichmann H. Mitochondrial DNA mutations in multiple symmetric lipomatosis. Molecular and Cellular Biochemistry 1997; 174:271-275
  226. Chalk CH, Mills KR, Jacobs JM, Donaghy M. Familial multiple symmetric lipomatosis with peripheral neuropathy. Neurology 1990; 40:1246-1250
  227. Musumeci O, Barca E, Lamperti C, Servidei S, Comi GP, Moggio M, Mongini T, Siciliano G, Filosto M, Pegoraro E, Primiano G, Ronchi D, Vercelli L, Orsucci D, Bello L, Zeviani M, Mancuso M, Toscano A. Lipomatosis Incidence and Characteristics in an Italian Cohort of Mitochondrial Patients. Front Neurol 2019; 10:160.:10.3389/fneur.2019.00160. eCollection 02019.
  228. Zolotov S, Xing C, Mahamid R, Shalata A, Sheikh-Ahmad M, Garg A. Homozygous LIPE mutation in siblings with multiple symmetric lipomatosis, partial lipodystrophy, and myopathy. Am J Med Genet A 2017; 173:190-194. doi: 110.1002/ajmg.a.37880. Epub 32016 Nov 37811.
  229. Sawyer SL, Cheuk-Him Ng A, Innes AM, Wagner JD, Dyment DA, Tetreault M, Majewski J, Boycott KM, Screaton RA, Nicholson G. Homozygous mutations in MFN2 cause multiple symmetric lipomatosis associated with neuropathy. Hum Mol Genet 2015; 24:5109-5114. doi: 5110.1093/hmg/ddv5229. Epub 2015 Jun 5117.
  230. Kodish ME, Alsever RN, Block MB. Benign symmetric lipomatosis: functional sympathetic denervation of adipose tissue and possible hypertrophy of brown fat. Metabolism 1974; 23:937-945
  231. Busetto L, Strater D, Enzi G, Coin A, Sergi G, Inelmen EM, Pigozzo S. Differential clinical expression of multiple symmetric lipomatosis in men and women. Int J Obes Relat Metab Disord 2003; 27:1419-1422
  232. Carr A, Miller J, Law M, Cooper DA. A syndrome of lipoatrophy, lactic acidaemia and liver dysfunction associated with HIV nucleoside analogue therapy: contribution to protease inhibitor-related lipodystrophy syndrome. Aids 2000; 14:F25-32
  233. Engelson ES. HIV lipodystrophy diagnosis and management. Body composition and metabolic alterations: diagnosis and management. AIDS Read 2003; 13:S10-14
  234. Lo JC, Mulligan K, Tai VW, Algren H, Schambelan M. "Buffalo hump" in men with HIV-1 infection. Lancet 1998; 351:867-870
  235. Heath KV, Hogg RS, Chan KJ, Harris M, Montessori V, O'Shaughnessy MV, Montanera JS. Lipodystrophy-associated morphological, cholesterol and triglyceride abnormalities in a population-based HIV/AIDS treatment database. Aids 2001; 15:231-239
  236. Urso R, Gentile M. Are 'buffalo hump' syndrome, Madelung's disease and multiple symmetrical lipomatosis variants of the same dysmetabolism? Aids 2001; 15:290-291
  237. Lindner A, Marbach F, Tschernitz S, Ortner C, Berneburg M, Felthaus O, Prantl L, Kye MJ, Rappl G, Altmüller J, Thiele H, Schreml S, Schreml J. Calcyphosine-like (CAPSL) is regulated in Multiple Symmetric Lipomatosis and is involved in Adipogenesis. Scientific Reports 2019; 9:8444
  238. Zancanaro C, Sbarbati A, Morroni M, Carraro R, Cigolini M, Enzi G, Cinti S. Multiple symmetric lipomatosis. Ultrastructural investigation of the tissue and preadipocytes in primary culture. Lab Invest 1990; 63:253-258
  239. Cinti S, Enzi G, Cigolini M, Bosello O. Ultrastructural features of cultured mature adipocyte precursors from adipose tissue in multiple symmetric lipomatosis. Ultrastruct Pathol 1983; 5:145-152
  240. Chen K, He H, Xie Y, Zhao L, Zhao S, Wan X, Yang W, Mo Z. miR-125a-3p and miR-483-5p promote adipogenesis via suppressing the RhoA/ROCK1/ERK1/2 pathway in multiple symmetric lipomatosis. Sci Rep 2015; 5:11909.:10.1038/srep11909.
  241. Prantl L, Schreml J, Gehmert S, Klein S, Bai X, Zeitler K, Schreml S, Alt E, Gehmert S, Felthaus O. Transcription Profile in Sporadic Multiple Symmetric Lipomatosis Reveals Differential Expression at the Level of Adipose Tissue-Derived Stem Cells. Plast Reconstr Surg 2016; 137:1181-1190. doi: 1110.1097/PRS.0000000000002013.
  242. Moonen MPB, Nascimento EBM, van Kroonenburgh M, Brandjes D, van Marken Lichtenbelt WD. Absence of (18) F-fluorodeoxyglucose uptake using Positron Emission Tomography/Computed Tomography in Madelung's disease: A case report. Clin Obes 2019; 9:e12302. doi: 12310.11111/cob.12302. Epub 12019 Feb 12327.
  243. Sebastian BM, Roychowdhury S, Tang H, Hillian AD, Feldstein AE, Stahl GL, Takahashi K, Nagy LE. Identification of a cytochrome P4502E1/Bid/C1q-dependent axis mediating inflammation in adipose tissue after chronic ethanol feeding to mice. J Biol Chem 2011; 286:35989-35997. doi: 35910.31074/jbc.M35111.254201. Epub 252011 Aug 254219.
  244. Dumont AE, Mulholland JH. Flow rate and composition of thoracic-duct lymph in patients with cirrhosis. N Engl J Med 1960; 263:471-4.:10.1056/NEJM196009082631001.
  245. Ludwig J, Linhart P, Baggenstoss AH. Hepatic lymph drainage in cirrhosis and congestive heart failure. A postmortem lymphangiographic study. Arch Pathol 1968; 86:551-562.
  246. Souza-Smith FM, Siggins RW, Molina PE. Mesenteric Lymphatic-Perilymphatic Adipose Crosstalk: Role in Alcohol-Induced Perilymphatic Adipose Tissue Inflammation. Alcohol Clin Exp Res 2015; 39:1380-1387. doi: 1310.1111/acer.12796. Epub 12015 Jul 12794.
  247. Szewc M, Sitarz R, Moroz N, Maciejewski R, Wierzbicki R. Madelung's disease - progressive, excessive, and symmetrical deposition of adipose tissue in the subcutaneous layer: case report and literature review. Diabetes Metab Syndr Obes 2018; 11:819-825.:10.2147/DMSO.S181154. eCollection 182018.
  248. Chen CY, Fang QQ, Wang XF, Zhang MX, Zhao WY, Shi BH, Wu LH, Zhang LY, Tan WQ. Madelung's Disease: Lipectomy or Liposuction? Biomed Res Int 2018; 2018:3975974.:10.1155/2018/3975974. eCollection 3972018.
  249. Constantinidis J, Steinhart H, Zenk J, Gassner H, Iro H. Combined surgical lipectomy and liposuction in the treatment of benign symmetrical lipomatosis of the head and neck. Scand J Plast Reconstr Surg Hand Surg 2003; 37:90-96.
  250. Andou E, Komoto M, Hasegawa T, Mizuno H, Hayashi A. Surgical excision of madelung disease using bilateral cervical lymphnode dissection technique-its effect and the influence of previous injection lipolysis. Plast Reconstr Surg Glob Open 2015; 3:e375. doi: 310.1097/GOX.0000000000000337. eCollection 0000000000002015 Apr.