Archives

Hypoglycemia

CLINICAL RECOGNITION

 

Hypoglycemia is uncommon in people who are not being treated for diabetes mellitus. Low blood glucose concentrations lead to adrenergic activation and neuroglycopenia (Table 1). Symptomatic hypoglycemia is diagnosed clinically using Whipple’s triad: symptoms of hypoglycemia, plasma glucose concentration<55 mg/dl (3.0 mmol/l), and resolution of those symptoms after the plasma glucose concentration is raised. Capillary blood glucose measurements should not be used in the evaluation of hypoglycemia due to poor accuracy.

 

Table 1. Symptoms of Hypoglycemia

Adrenergic

Neuroglycopenic

Sweating
Warmth
Anxiety
Tremor
Nausea
Palpitations
Tachycardia
Hunger

Behavioral changes
Changes in vision or speech
Confusion
Dizziness
Lethargy
Seizure
Loss of consciousness
Coma

 

DIAGNOSIS AND DIFFERENTIAL DIAGNOSIS

 

Hypoglycemia in diabetes is typically the result of treatments that raise insulin levels and thus lower plasma glucose concentrations (Table 2). In adults not taking glucose-lowering drugs to treat diabetes mellitus, critical illnesses, hormone deficiencies, and islet and non-islet cell tumors should be considered.

 

Table 2. Causes of Adult-Onset Hypoglycemia

Drugs - see Table 3

Hepatic, renal or cardiac failure

Sepsis, trauma, burns

Malnutrition

Hormonal deficiencies (cortisol, glucagon, epinephrine)

Non-islet cell tumors (IGF-II secreting tumors)

Insulinoma (insulin-secreting tumors)

Non-insulinoma pancreatogenous hypoglycemia (NIPHS)

Post gastric bypass surgery

Post total pancreatectomy with islet auto-transplantation

Dumping syndrome or rapid gastric emptying

Insulin antibodies

Insulin receptor antibodies

Accidental, surreptitious or malicious including Munchausen syndrome by proxy

Adapted from: Cryer, PE, et al. Evaluation and Management of Adult Hypoglycemic Disorders: An Endocrine Society Clinical Practice Guideline. J Clin Endocrinol Metab 94:709-728, 2009.

 

Table 3. Drugs Reported to Cause Hypoglycemia

Insulin

Insulin secretagogues (especially sulfonylureas, meglitinides)

Alcohol

Cibenzoline

Glucagon (during endoscopy)

Indomethacin

Pentamidine

Sulfonamides

Quinine

Hydroxychloroquine

Artesunate/artemisin/artemether

Chloroquineoxaline

IGF-1

Lithium

Propoxyphene/dextropropoxyphene

Salicylates

The following are supported by very low-quality evidence:

Angiotensin converting enzyme inhibitors

Angiotensin receptor antagonists

Nonselective β-adrenergic receptor antagonists

Fluoroquinolones

Gabapentin

Mifepristone

Disopyramide

Trimethoprim-sulfamethoxazole

Heparin

6-Mercaptopurine

Adapted from: Cryer, PE, et al. Evaluation and Management of Adult Hypoglycemic Disorders: An Endocrine Society Clinical Practice Guideline. J Clin Endocrinol Metab 94:709-728, 2009.

 

PATHOPHYSIOLOGY

 

Glucose is an obligate fuel for the brain under physiologic conditions. In order to maintain proper brain function, plasma glucose must be maintained within a relatively narrow range. Redundant counter-regulatory mechanisms are in place to prevent or correct hypoglycemia. As glucose levels decline, major defenses include: 1) a decrease in insulin secretion; 2) an increase in glucagon secretion; 3) an increase in epinephrine secretion. Increased cortisol and growth hormone secretion also occur. If these defenses fail, plasma glucose levels will continue to fall. Symptoms, prompting food ingestion, typically develop at a plasma glucose of 55 mg/dl (3.0 mmol/liter). At glucose levels of 55 mg/dl and lower, insulin secretion is normally almost completely suppressed.

 

In longstanding type 1 and type 2 diabetes these counter-regulatory responses to hypoglycemia are impaired. This increases the risk of hypoglycemia and also contributes to hypoglycemia unawareness.

 

DIAGNOSTIC TESTS

 

If the cause of the hypoglycemia is not evident, measure plasma glucose, insulin, c-peptide, proinsulin, and beta-hydroxybutyrate concentrations and screen for oral hypoglycemic agents (sulfonylurea and meglitinide drugs) during an episode of spontaneous hypoglycemia. Glucagon, 1 mg IV, should then be administered, with a rise in glucose >25 mg/dl (1.4 mmol/L) suggesting hyperinsulinemic hypoglycemia. The diagnosis of insulinoma is supported if insulin, c-peptide and proinsulin levels are elevated, beta-hydroxybutyrate is <2.7 mmol/l, and sulfonylurea/meglitinide levels are undetectable during the hypoglycemic episode.

 

If testing cannot be performed during a spontaneous episode of hypoglycemia, a 72 hour fast or a mixed meal test, performed in a monitored setting, followed by administration of glucagon is the most useful diagnostic strategy.

 

During a 72 hour fast, patients are allowed no food but can consume non-caloric caffeine-free beverages. Insulin, c-peptide and glucose samples are obtained at the beginning of the fast and every 4-6 hours. When the plasma glucose falls to <60 mg/dl, specimens should be taken every 1-2 hours under close supervision. Patients should continue activity when they are awake. The fast continues until the plasma glucose falls below 45 mg/dl (2.5 mmol/l) [plasma glucose <55 mg/dl (3.0 mmol/l) is recommended in the Endocrine Society guidelines] and symptoms of neuroglucopenia develop, at which time insulin, glucose, c-peptide, oral insulin secretagogue, proinsulin, and beta-hydroxybutyrate levels are obtained and the fast is terminated. Additional samples for insulin antibodies, anti-insulin receptor antibodies, IGF-1/IGF-2, and plasma cortisol, glucagon or growth hormone can also be obtained at this time if a non-islet cell tumor, autoimmune etiology, or hormone deficiency is suspected. Patients are fed at the conclusion of the fast.

 

For patients with hypoglycemic symptoms several hours after meals, a mixed meal test may be performed. This test has not been well standardized. Patients eat a meal similar to one that provokes their symptoms, or a commercial mixed meal. Samples for plasma glucose, insulin, c-peptide, and proinsulin are collected prior to the meal and every 30 minutes thereafter for 5 hours. If symptoms occur prior to the end of the test then additional samples for the above are collected prior to administration of carbohydrates. If Whipple’s triad is demonstrated, testing for oral hypoglycemic drugs and testing for insulin antibodies should be done. Interpretation of test results is the same as for the 72-hour fast or spontaneous hypoglycemia (Table 4).

 

Table 4. Distinguishing Causes of Symptomatic Hypoglycemia After a Prolonged Fast

Insulin (µU/ml)

C-peptide (nmol/L)

Proinsulin (pmol/L)

Oral hypoglycemic

Interpretation

»3

<0.2

<5

No

Exogenous insulin

≥3

≥0.2

≥5

No

Endogenous insulina

≥3

≥0.2

≥5

Yes

Oral hypoglycemic drug

a- Insulinoma, non-insulinoma pancreatogenous hypoglycemia (NIPHS), post gastric bypass surgery.

Adapted from: Cryer, PE, et al. Evaluation and Management of Adult Hypoglycemic Disorders: An Endocrine Society Clinical Practice Guideline. J Clin Endocrinol Metab 94:709-728, 2009

 

In a patient with documented hypoglycemia with laboratory findings consistent with endogenous hyperinsulinism localizing studies should be done to evaluate for insulinoma. These may include computed tomography (CT) or magnetic resonance imaging, transabdominal and endoscopic ultrasonography, and, where available, new nuclear medicine scans (GLP-1 receptor imaging), somatostatin receptor imaging SPECT / PET, and 6-[fluoride-18] fluoro-levodopa-PET-CT. If the diagnosis remains unclear, selective pancreatic arterial calcium injections with measurements of hepatic venous insulin levels can be performed.

 

TREATMENT

 

Immediate treatment should be focused on reversing the hypoglycemia. If the patient is able to ingest carbohydrates 15 to 20 grams of glucose should be given every 15 minutes until the hypoglycemia has resolved. If the patient is unable to ingest carbohydrates, or if the hypoglycemic episode is severe then parenteral glucose should be administered. In a healthcare setting intravenous dextrose is used. Twenty-five-gram boluses of 50% dextrose are given until the hypoglycemia has resolved. If needed, an infusion of 10% or 20% dextrose can be used to sustain euglycemia in patients with recurrent episodes of hypoglycemia. In the outpatient setting, glucagon is used to correct hypoglycemia. Glucose gel and other forms of oral glucose should be used in impaired patients with caution and only in circumstances where no alternative is available, as they pose an aspiration risk.

 

Long-term treatment should be tailored to the specific hypoglycemic disorder, taking into account the burden of hypoglycemia on well-being and patient preferences. Offending medications should be discontinued and underlying illnesses treated, whenever possible.

 

Surgical resection can be curative for insulinomas, and can alleviate hypoglycemia in non-islet cell tumors, even if the malignancy cannot be cured. Partial pancreatectomy can be considered in patients with β-cell disorders. Medical treatment with frequent feedings, α-glucosidase inhibitors, diazoxide, or octreotide can be used if resection is not possible, or as a temporizing measure. New drugs that may be helpful include long-acting somatostatin analogs, mTOR inhibitors, and GLP-1 antagonists. Autoimmune hypoglycemic conditions may be treated with either glucocorticoids or immunosuppressants, but these disorders may be self-limited.

For adults taking insulin or insulin secretagogues for diabetes mellitus risk factors for hypoglycemia, such as advanced age and renal insufficiency, should be considered. The treatment regimen and glycemic goals should be reviewed and adjusted if needed. Patients should be instructed on how to manage hypoglycemia, either by the ingestion of carbohydrates if possible, or by parenteral glucagon or glucose. If the patient has hypoglycemia unawareness, a 2-to 3-week period of strict avoidance of hypoglycemia should be maintained, as hypoglycemia awareness will return in many patients. For individuals with type 1 diabetes and a history of serious hypoglycemia, the use of a personal continuous glucose monitoring device, sensor-augmented insulin pump therapy, or a hybrid closed loop system should be considered.

 

GUIDELINES

 

Cryer, PE, Axelrod L, Grossman AB, Heller SR, Montori VM, Seaquist ER, Service FJ. Evaluation and Management of Adult Hypoglycemic Disorders: An Endocrine Society Clinical Practice Guideline. J Clin Endocrinol Metab 94:709-728, 2009.

 

REFERENCES

 

Bansal N, Weinstock RS. Non-Diabetic Hypoglycemia. 2020 May 20. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dhatariya K, Dungan K, Hershman JM, Hofland J, Kalra S, Kaltsas G, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrère B, Levy M, McGee EA, McLachlan R, Morley JE, New M, Purnell J, Sahay R, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000–. PMID: 27099902

 

de Herder WW, Zandee WT, Hofland J. Insulinoma. 2020 Oct 25. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dhatariya K, Dungan K, Hershman JM, Hofland J, Kalra S, Kaltsas G, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrère B, Levy M, McGee EA, McLachlan R, Morley JE, New M, Purnell J, Sahay R, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000–. PMID: 25905215

 

Davis HA, Spanakis EK, Cryer PE, Davis SN. Hypoglycemia During Therapy of Diabetes. 2021 Jun 29. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dhatariya K, Dungan K, Hershman JM, Hofland J, Kalra S, Kaltsas G, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrère B, Levy M, McGee EA, McLachlan R, Morley JE, New M, Purnell J, Sahay R, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000–. PMID: 25905325

 

Inpatient Diabetes Management

CLINICAL RECOGNITION

Background

Appropriate inpatient glycemic management limits the risks of severe hypo- and hyperglycemia. Preventing and treating hyperglycemia reduces infections and minimizes fluid and electrolyte abnormalities. Specific glucose goals remain fluid. Hyperglycemia and hypoglycemia are associated with poor outcomes but the few prospective randomized studies have failed to demonstrate consistent improvements. For example, intensive glycemic control in the ICU increased mortality in one large trial. At this point, glucose goals should be thoughtful and tailored to the institution and its resources. To successfully manage inpatient diabetes, institutional infrastructure must be in place with institution specific guidelines and protocols for which all nursing staff, pharmacy staff, physicians, and others must be educated. The general guidelines below are appropriate at most institutions.

 

Check A1c level in all patients with diabetes and individuals with glucose levels greater than 140mg/dL if not performed in the prior 3 months to evaluate prior glycemic control.

 

Insulin therapy should be initiated if glucose levels are persistently ≥180 mg/dL. Once insulin therapy is started, a target glucose range of 140–180 mg/dL is recommended for most hospitalized patients but in selected patients (e.g., critically ill postsurgical patients or patients with cardiac surgery) glucose levels between 110–140 mg/dL may be targeted if they can be achieved without significant hypoglycemia. Glucose levels between 180-250 mg/dL may be acceptable in patients with severe comorbidities and in hospitals where frequent glucose monitoring or close nursing supervision is not possibility. In terminally ill patients with a short life expectancy glucose level >250 mg/dL with less aggressive insulin regimens to minimize glucosuria, dehydration, and electrolyte disturbances may be appropriate. 

Physiologic Insulin Regimen

All patients have “basal, nutritional, and correctional” requirements which they must meet with endogenous or exogenous insulin.

 

  • Basal: insulin needed even when patient is not eating (to control gluconeogenesis). Use long-acting insulins such as glargine (usually once daily in AM or at bedtime) or detemir (once daily or q 12 hours). If there are financial limitations NPH at bedtime or AM and bedtime may be used. Additionally, a continuous insulin infusion can provide basal insulin and is often employed in ICU settings.
  • Nutritional: insulin to cover carbohydrate intake from food, dextrose in IV fluid, tube feeds, TPN. Use rapid-acting insulin (aspart, lispro, or glulisine) or if financial limitations short-acting insulin (regular).
  • Correctional: insulin given to bring a high blood glucose level down to target range (with target usually below 150 mg/dL pre-meal, and below 200mg/dL at bedtime or 2am). Use rapid-acting insulin (aspart, lispro, or glulisine) or short-acting insulin (regular).

General Rules

  • A PATIENT WITH TYPE 1 DM WILL ALWAYS NEED EXOGENOUS BASAL INSULIN, EVEN IF NPO. FAILURE TO GIVE SUCH A PATIENT INSULIN WILL LEAD TO DKA.
  • Arbitrary sliding scale insulin should be avoided as it is not only ineffective but also potentially dangerous.
  • Ad hoc insulin orders should not be used. Comprehensive electronic medical record (EMR) order sets, or pre-printed order forms should only be used to order subcutaneous insulin and insulin infusions. This standardization will decrease the risks of insulin dosing and administration errors.
  • Check blood glucose (BG) before meals and at bedtime. Check BG q 4 or q 6 hours in a patient who is NPO or is receiving continuous tube feeds or TPN. Continuous glucose monitoring may be used in certain patients (for example it was used in patients with COVID-19 infections to minimize patient contact).
  • Involve the diabetes educator or nurse specialist if available.
  • On admission, begin planning discharge, especially if the discharge plan will require new outpatient insulin use. Identify whether the patient will need a new glucose meter. Prescribe insulin, insulin pens with pen needles, syringes/needles, lancets, glucose strips, glucose tablets, and glucagon kit in the discharge prescription if needed. 

Oral Hypoglycemic Agents

In general, oral diabetes medications and injectables other than insulin (e.g., GLP agonists) are inappropriate for initial management of the hyperglycemia patient. Hospitalized patients often have the potential for renal impairment, tissue hypoxia, or need IV contrast, and these are all contraindications for using metformin. Sulfonylureas should be held on admission because of current or potential NPO status resulting in a high risk of hypoglycemia. As a patient’s status improves, however, it may be appropriate to restart oral medications. DPP4 inhibitors may be useful for patients who have minimally elevated glucoses as there is a minimal risk for hypoglycemia. 

Miscellaneous Guidelines

  • Nutritional coverage: Regular insulin is given 30 min before each meal.  Lispro, aspart, or glulisine are given with each meal or immediately after eating (can base on amount eaten).
  • Infection and glucocorticoids increase insulin needs; renal insufficiency decreases insulin needs.
  • Total daily dose of insulin needed: Type 1 patients require approximately 0.4 units/kg/day; type 2 patients vary in their insulin resistance and may require from 0.5 to 2 units/kg/day.

THERAPY

Insulin Regimens

The guidelines below assist with initial determination and subsequent adjustment of insulin doses. Insulin doses must be reevaluated on a daily basis and orders should be rewritten in order to achieve goals and to adapt to the patients’ changing clinical situation.

INSULIN REGIMEN FOR A PATIENT CONTROLLED WITH DIET AT HOME BUT NEEDING INSULIN IN HOSPITAL

Day 1:  Order a correctional sliding scale for before meals and bedtime (with lispro, aspart, glulisine or regular) based on BMI – see Table 1.

Day 2:  If BG pre-meals are >150 mg/dL, add nutritional insulin (with lispro, aspart, glulisine or regular) based on appetite).  Also, if AM fasting BG is >150 mg/dL, add bedtime basal insulin (with glargine, detemir, or NPH) dosed 0.1-0.2 unit/kg.

Day 3:  Adjust insulin doses based on BG pattern: Increase or decrease basal insulin based on AM fasting BG, and adjust nutritional insulin based on pre-meal BG levels (see below for details).

 

Table 1. Correctional Insulin (lispro, aspart, glulisine or regular)

BG

(mg/dL)

Pre-meal:

Sensitive (BMI <25 or <50 units/d)

Pre-meal:

Average (BMI 25-30 or 50-90 units/d)

Pre-meal:

Resistant (BMI >30 or >90 units/d)

Bedtime

and 2 a.m.

131-150

 0 units

1 unit

2 units

0 units

151-200

1 unit

2 units

3 units

0 units

201-250

2 units

4 units

6 units

1 unit

251-300

3 units

6 units

9 units

2 units

301-350

4 units

8 units

12 units

3 units

351-400

5 units

10 units

15 units

3 units

>400

6 units

12 units

18 units

3 units

INSULIN REGIMEN FOR A PATIENT ON ORAL AGENT(S) BUT REQUIRING INSULIN IN HOSPITAL BECAUSE OF HYPERGLYCEMIA OR CONTRAINDICATIONS TO THE ORAL AGENT(S)

Day 1:  Start nutritional insulin (lispro, aspart, glulisine or regular) based on appetite – generally about 0.1-0.2 units per kg, divided between the three meals for the day.  Also, order a correctional sliding scale (lispro, aspart, glulisine or regular) based on BMI – see Table 1.

Day 2:  If AM fasting BG is >150 mg/dL, add bedtime basal (glargine, detemir or NPH) dose of 0.1-0.2 units/kg.

Day 3:  Adjust insulin doses based on BG pattern: Increase or decrease basal insulin based on AM fasting BG, and adjust nutritional insulin based on pre-meal BG levels (see below for details).

INSULIN REGIMEN FOR A PATIENT ON INSULIN AT HOME

  • If possible, consider home BG control, appetite, renal function, and risk for hypoglycemia.
  • All three components of insulin replacement must be addressed: basal, nutritional and correctional.
  • Basal requirements: Continue home regimen if patient has been well-controlled at home, but consider decreasing the total dose by 20-30% to reduce the risk of in-hospital hypoglycemia. Alternatively, start bedtime glargine, detemir or NPH at a dose of 0.2 units/kg
  • Nutritional requirements: Order nutritional insulin (lispro, aspart, glulisine or regular) based on appetite, or consider pre-meal dosing of 0.2 units/kg divided by 3 for the dose at each meal.
  • Correctional need: Order a correctional sliding scale based on total insulin dose or BMI – see Table 1.

INSULIN REGIMEN WHEN A PATIENT IS MADE NPO FOR A PROCEDURE

A patient will always require his or her basal insulin, even while NPO, and should not become hypoglycemic if that basal insulin is dosed appropriately.  For safety purposes, however:

 

  • The night before, give the usual dose of bedtime NPH, if applicable, or decrease the usual dose of bedtime glargine/detemir by 25%.
  • The morning of, if applicable, decrease the usual dose of morning NPH by 50%, or decrease the usual dose of morning glargine by 25%.
  • Do not give nutritional insulin (as patient is not eating), but continue the usual correctional insulin.
  • (An online resource to determine patient specific instructions when preparing for an NPO episode is athttp://ucsf.logicnets.com)

INSULIN REGIMEN FOR AN ICU OR SURGICAL PATIENT WHO IS NPO

 Consider insulin infusion therapy.

INSULIN REGIMEN FOR A PATIENT STARTING CONTNUOUS TUBE FEEDING

  • Consider insulin infusion therapy.
  • If moving from IV to SQ see below.
  • Basal need: The daily basal dose (glargine, detemir or total bid NPH dose) is the estimated total daily dose divided by 2.
  • Nutritional need: Divide the estimated total daily dose by 10 for the total nutritional (lispro, aspart, glulisine or regular) dose, to be given q 4 hours while tube feeding is active.
  • Correctional need: Order a correctional scale (lispro, aspart, glulisine or regular) based on total insulin dose or BMI (Table 1)
  • If not using IV insulin to start:
  • Estimate the tube feed formula’s 24-hour carbohydrate load.
  • Estimate the total daily dose (TDD) of insulin, starting with 1 unit insulin for every 10 grams carbohydrate.

INSULIN REGIMEN FOR A PATIENT RECEIVING TPN

  • Standard TPN often contains 25% glucose, which, if 100 ml/hour, yields 25 g glucose/hour.
  • Basal and nutritional needs: Adding insulin to the TPN is safest, as the unexpected discontinuation of TPN will also mean the discontinuation of the insulin.  Start with 0.1 unit per gram glucose. If patient previously needed high doses of basal insulin, divide that total daily dose by the number of TPN bottles to be administered daily, and add that to the prior calculation.
  • Correctional: Order a correctional sliding scale (lispro, aspart, glulisine or regular) based on BMI (Table 1).

INSULIN REGIMEN TO TRANSITION FROM AN INSULIN INFUSION TO SUBCUTANEOUS INSULIN

  • Calculate the patient’s total daily dose (TDD) of insulin, based on the most recent insulin infusion rate. For safety purposes, take 80% of that dose.
  • Basal need: Divide the 80% of the TDD by 2 and give half for the daily glargine, detemir, or total NPH dose.
  • Nutritional need: If the patient is eating, divide the 80% of the TDD by 6 for the pre-meal lispro, aspart, glulisine, or regular dose.  If the patient is receiving tube feeds, divide the 80% of the TDD by 10 for the nutritional (lispro, aspart, glulisine or regular) dose, to be given q 4 hours.  If the patient is not receiving nutrition, do not order nutritional insulin.
  • Correctional need: Order a q4h correctional scale (lispro, aspart, glulisine, or regular) based on total insulin dose or BMI (Table 1).
  • Give the first basal insulin SQ injection 1-2 hours before the infusion is discontinued. If the transition is being made in the morning, consider using a one-time AM NPH injection or ½ of daily glargine or detemir dose to bridge until bedtime glargine, detemir or NPH begins.

INSULIN REGIMEN FOR A PATIENT RECEIVING GLUCOCORTICOIDS

  • Glucocorticoids may dramatically increase postprandial BG levels but have little effect on gluconeogenesis (fasting glucose levels). Often, BG levels are very high during the day, then lower overnight.
  • Anticipate post-prandial hyperglycemia by increasing the nutritional insulin doses.
  • The insulin dose will typically increase by 50% from before glucocorticoid use and the total amount may be 0.5 to significantly >1 Unit/kg

DAILY INSULIN ADJUSTMENTS

There are no validated formulas for making these adjustments, but the following rules generally work well.

 

  • Basal Insulin: Generally, the basal insulin dose is adjusted based on fasting glucose levels.  For example, if FBS <140, no change.  If FBS 141-160, increase basal dose by 2-3 units.  If FBS 160-180, increase basal dose by 4-5 units. If FBS 180-200, increase basal dose by 6-7 units.  If FBS >200, increase basal dose by 8 units.  With this approach, the basal insulin can be titrated up to the patient’s actual requirement relatively quickly.
  • Nutritional Insulin: The adequacy of the nutritional insulin dose is based on the glucose level prior to the next meal. For example, the glucose level just before lunch will indicate whether the insulin given at breakfast was appropriate.  The glucose level at bedtime will indicate whether the insulin given at dinner was appropriate.  A simple approach is as follows: If there was no significant change in the glucose level from before breakfast to before lunch, then the total dose of insulin the patient received at breakfast (nutritional plus correctional) should be used as the nutritional dose for breakfast the next day.  If there was a significant increase in the glucose level from before breakfast to before lunch, then the total dose of insulin the patient received at breakfast (nutritional plus correctional) should be increased and should become the nutritional dose for breakfast the next day.  If the glucose level before breakfast was high, and the glucose level at lunch was at goal, then no change in the nutritional dose will be required for the next day.  Finally, no matter what the glucose level was at breakfast, if the glucose level after breakfast or before lunch was low, then the breakfast nutritional dose should be decreased for the next day.

Hypoglycemic Protocols      

  • BG <70 mg/dL: If patient taking po, give 20 grams of oral fast-acting carbohydrate either as glucose tablets or 6 oz. fruit juice.  If patient cannot take po, give 25 mL D50 IV push.
  • Check BG every 15 minutes and repeat above treatment until BG is ≥100 mg/dL.

Insulin Infusions

  • Use your hospital’s pre-printed order form or protocol in EMR and hospital-specific protocol for insulin infusions. Using an insulin infusion without a standardized protocol and trained providers can be unsafe.
  • Continuous glucose intake (in IV fluid or continuous TPN or tube feeds) is required during the infusion. Remember to manually adjust the infusion rate and/or the algorithm if there are changes in nutrition (e.g., if tube feeding or TPN is held) or other rapid changes in medical status.
  • When converting to SQ insulin, give the basal SQ dose 1-2 hours before discontinuing the insulin infusion.

GUIDELINE

Diabetes Care in the Hospital: Standards of Medical Care in Diabetes—2022

American Diabetes Association Professional Practice Committee. Diabetes Care December 2021, Vol.45, S244-S253.

 

Umpierrez GE, Hellman R, Korytkowski MT, Kosiborod M, Maynard GA, Montori VM, Seley JJ, Van den Berghe G; Endocrine Society. Management of hyperglycemia in hospitalized patients in non-critical care setting: an endocrine society clinical practice guideline. J Clin Endocrinol Metab. 2012 Jan;97(1):16-38.

REFERENCES

 

Society for Hospital Medicine Diabetes Resource Room:  http://www.hospitalmedicine.org/ResourceRoomRedesign/GlycemicControl.cfm

Dhatariya K, Corsino L, Umpierrez GE. Management of Diabetes and Hyperglycemia in Hospitalized Patients. 2020 Dec 30. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dhatariya K, Dungan K, Hershman JM, Hofland J, Kalra S, Kaltsas G, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrère B, Levy M, McGee EA, McLachlan R, Morley JE, New M, Purnell J, Sahay R, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000–.

PMID: 25905318

 

Guidelines for the Management of High Blood Cholesterol

ABSTRACT

 

The cholesterol hypothesis holds that high blood cholesterol is a major risk factor for atherosclerosis cardiovascular disease (ASCVD) and lowering cholesterol levels will reduce risk for ASCVD. This hypothesis is based on epidemiological evidence that both within and between populations higher cholesterol levels raise the risk for ASCVD; and conversely, randomized clinical trials (RCTs) show that lowering cholesterol levels will reduce risk. Cholesterol in the circulation is embedded in lipoproteins. The major atherogenic lipoproteins are low density lipoproteins (LDL), very low-density lipoproteins (VLDL), and remnants. Together they constitute non-high-density lipoproteins (non-HDL).  Clinically these lipoproteins are identified by their cholesterol (C) content, i.e., LDL-C, VLDL-C, and non-HDL-C.  Atherogenic lipoproteins can be reduced by both lifestyle intervention and cholesterol-lowering drugs. The efficacy of lifestyle intervention is best demonstrated in epidemiological studies, whereas efficacy of drugs is revealed through RCTs. Currently available cholesterol-lowering drugs are statins, ezetimibe, bempedoic acid, bile acid sequestrants, proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors, niacin, fibrates, and n-3 fatty acids (e.g., icosapent ethyl). The latter three generally are reserved for patients with hypertriglyceridemia; here they can be combined with statins that together lower non-HDL-C. Highest priority for cholesterol-lowering therapy goes to patients with established ASCVD (secondary prevention).  RCTs in such patients show that “lower is better” for cholesterol reduction. The greatest risk reductions are attained by reducing LDL-C concentrations by at least 50% with a high intensity statin; and if necessary, to achieve LDL-C < 55-70 mg/dL, combining a statin with ezetimibe or PCSK9 inhibitor. For primary prevention, a decision to initiate statin therapy is made on multiple factors (i.e., presence of diabetes or severe hypercholesterolemia, estimated 10-year risk or lifetime risk for ASCVD, presence of risk enhancing factors (e.g., metabolic syndrome and chronic kidney disease); and if in doubt, detection of subclinical atherosclerosis (e.g., coronary artery calcium [CAC]).  A reasonable goal for primary prevention using moderate-intensity statin therapy is an LDL-C in the range of 70-99 mg/dL. Both population epidemiology and genetic epidemiology show that low serum cholesterol throughout life will minimize lifetime risk of ASCVD.  For this reason, cholesterol-lowering intervention, preferably through lifestyle change, should be carried out as early as possible. If cholesterol concentrations are very high in younger adults, it sometimes may be judicious to introduce a cholesterol-lowering drug. 

 

INTRODUCTION

 

Atherosclerotic cardiovascular disease (ASCVD) remains the foremost cause of death among chronic diseases. Its prevalence is increasing in many countries. This increase results from aging of the population combined with atherogenic lifestyles. Even so, mortality from ASCVD has been declining in most developed countries. This decline comes from improvements in preventive measures and better clinical interventions. One of the most important advances in the cardiovascular field resulted from identifying risk factors for ASCVD.  Risk factors directly or indirectly promote atherosclerosis, or they otherwise predispose to vascular events. The major risk factors are cigarette smoking, dyslipidemia, hypertension, hyperglycemia, and advancing age. Dyslipidemia consists of elevations of atherogenic lipoproteins (LDL, VLDL, Lp(a), and remnants) and low levels of HDL. Advancing age counts as a risk factor because it reflects the impact of all risk factors over the lifespan. Several other factors, called risk enhancing factors, associate with higher risk for ASCVD (1). Lifestyle factors (for example, overnutrition and physical inactivity) contribute importantly to both major and enhancing risk factors. Hereditary factors undoubtedly contribute to the identifiable risk factors; but genetic influences also affect ASCVD risk through other ways not yet understood (2).

 

HISTORY OF THE CHOLESTEROL HYPOTHESIS AND CHOLESTEROL-LOWERING THERAPY

 

The first evidence for a connection between serum cholesterol levels and atherosclerosis came from laboratory animals (3). Feeding cholesterol to various animal species raises serum cholesterol and causes deposition of cholesterol in the arterial wall (3). The latter recapitulates early stages of human atherosclerosis. Subsequently, in humans, severe hereditary hypercholesterolemia was observed to cause premature atherosclerosis and ASCVD (3).  Later, population surveys uncovered a positive association between serum cholesterol levels and ASCVD (4,5).  Finally, clinical trials with cholesterol-lowering agents documented that lowering of serum cholesterol levels reduces the risk for ASCVD (6). These findings have convinced most investigators that the cholesterol hypothesis is proven. Moreover, the relationship between cholesterol levels and ASCVD risk is bidirectional; raising cholesterol levels increases risk, whereas reducing levels decreases risk (Figure 1).

Figure 1. The Cholesterol Hypothesis. Between the years 1955 and 1985, many epidemiologic studies showed a positive relation between cholesterol levels and atherosclerotic cardiovascular disease (ASCVD) events. Over the next 30 years, a host of randomized controlled clinical trials have demonstrated that lowering cholesterol levels will reduce the risk for ASCVD. This bidirectional relationship between cholesterol levels and ASCVD provides ample support for the cholesterol hypothesis.

 

Epidemiological Evidence

 

A relationship between cholesterol levels and ASCVD risk is observed in both developing and developed countries (4,5). Populations with the lowest cholesterol levels and LDL-C levels have the lowest rates of ASCVD. Within populations, individuals with the lowest serum cholesterol or LDL-C levels carry the least risk. In other words, “the lower, the better” for cholesterol levels holds, both between populations and for individuals within populations.

 

Pre-Statin Clinical Trial Evidence

 

Another line of evidence supporting the cholesterol hypothesis comes from randomized controlled trials (RCTs) of cholesterol-lowering therapies. Several earlier RCTs tested efficacy by reducing cholesterol through diet, bile acid sequestrants, or ileal exclusion operation (Table 1) (4). When taken individually, results from some of the smaller trials were not definitive; but meta-analysis, which combines data from all RCTs, demonstrated significant risk reduction due to cholesterol lowering. In addition, before the discovery of statins, several secondary-prevention RCTs were performed with various cholesterol-lowering drugs. Although some of these trials showed significant risk reduction, others gave equivocal results. But again when taken together, meta-analysis demonstrated ASCVD risk reduction from cholesterol reduction (7).

 

Table 1. Summary of Pre-Statin Clinical Trials of Cholesterol-Lowering Therapy

Intervention

No. trials

No. treated

Person-years

Mean cholesterol reduction (%)

CHD incidence

(% change)

CHD Mortality

(%change)

Surgery

1

421

4,084

22

-43

-30

Sequestrants

3

1,992

14,491

9

-21

-32

Diet

6

1,200

6,356

11

-24

-21

This table is derived from National Cholesterol Education Program Adult Treatment Panel III (4)

 

Statins and Clinical Trial Evidence

 

Statins were discovered in the 1970s by Endo of Japan (8). These drugs lower cholesterol by inhibiting cholesterol synthesis in the liver. They block HMG CoA reductase, a key enzyme in cholesterol synthesis. This inhibition enhances the liver’s synthesis of LDL receptors. The latter, discovered by Brown and Goldstein (9), remove LDL and VLDL from the bloodstream, which lowers serum cholesterol levels.  Statin have proven to be highly efficacious with few side effects. The development of statins as a cholesterol-lowering drug has been actively pursued by the pharmaceutical industry. Seven statins have been approved for use in clinical practice by the FDA (for a detailed discussion of statins see (10)). Over the past three decades, a series of RCTs have been carried out that documents the efficacy and safety of statin therapy. In these RCTs, statin therapy has been shown to significantly reduce morbidity and mortality from ASCVD. Although individual RCTs produced significant results, the strongest evidence of benefit comes from meta-analysis. i.e., by combining data from all the trials (6). 

 Meta-analysis has shown that for every mmol/L (39 mg/dl) reduction in LDL-C with statin therapy there is an approximate 22% reduction in ASCVD events (6,11-14). Another report (15) showed that an almost identical relationship holds when several different kinds of LDL-lowering therapy were analyzed together. This response appears to be consistent throughout all levels of LDL-C.  Individual statins vary in their intensity of cholesterol-lowering therapy at a given dose (1,10) (Table 2).  For example, per mg per day, rosuvastatin is twice as efficacious as atorvastatin, which in turn is twice as efficacious as simvastatin.   Statins are best classified according to percentage reductions in LDL-C.  As shown in Table 2, moderate- intensity statins reduce LDL-C by 30-49 %, whereas high-intensity statins reduce LDL-C by > 50%.  On average, a 35% LDL-C reduction by moderate-intensity statin reduces risk by approximately one third, whereas high-intensity statins lower risk by about one-half.  But, in fact, absolute reductions vary depending on baseline levels of LDL-C. For example, for a baseline LDL-C of 200 mg/dL, a 50% reduction in LDL-C equates to a 100 mg/dL (2.6 mmol/L) decline; this translates into a 59% reduction in 10-year risk for ASCVD events. In contrast, in a patient with a baseline LDL-C a 100 mg per dL, a 50% reduction in LDL-C equates to a 50 mg/dL (1.3 mmol/L) decline, which will reduce ASCVD risk by about 30%.  Thus, at lower and lower levels of LDL-C, progressive reductions of LDL-C produce diminishing benefit from cholesterol-lowering therapy. This modifies the aphorism "lower is better".  Whereas the statement is true, it must be kept in mind that there are diminishing benefits from intensifying cholesterol-lowering therapy when LDL-C levels are already low. One needs to balance the benefits of further reducing LDL-C levels with the risks and costs of additional therapy. 

Table 2.  Categories of Intensities of Statins

Drug

Low-Intensity

20-25% LDL-C

Moderate-Intensity

30-49% LDL-C

High Intensity

>50% LDL-C

Lovastatin

10-20 mg

40-80 mg

 

Pravastatin

10-20 mg

40-80 mg

 

Simvastatin

10 mg

20-40 mg

 

Fluvastatin

20-40 mg

80 mg

 

Pitavastatin

 

1-4 mg

 

Atorvastatin

5 mg

10-20 mg

40-80 mg

Rosuvastatin

 

5-10 mg

20-40 mg

 

Non-Statin Cholesterol-Lowering Drugs

 

Beyond statins, other agents are currently available or loom on the horizon (Table 3). Bile acid sequestrants inhibit intestinal absorption of bile acids, which like statins raise hepatic LDL receptors (10). They are moderately efficacious for reducing LDL-C concentrations. A large RCT showed that bile acid sequestrants significantly reduce risk for CHD in patients with baseline elevations in LDL-C (16). Theoretically, bile acid sequestrants could enhance risk reduction in patients with ASCVD who are treated with statins.

 

Ezetimibe blocks cholesterol absorption in the intestine and also raises hepatic LDL receptor activity (10). It moderately lowers LDL-C (15-25%). The combination of statin + ezetimibe is additive for LDL-C lowering (17).  A clinical trial (18) demonstrated that adding ezetimibe to moderate intensity statins in very high-risk patients with ASCVD is beneficial showing that combination therapy reduced risk of cardiovascular events more than a statin alone (18). In this trial, the higher the risk, the greater was risk reduction (19). Ezetimibe is a generic drug and relatively inexpensive.

 

Bempedoic acid is an adenosine triphosphate-citrate lyase (ACL) inhibitor and thereby inhibits cholesterol synthesis leading to an increase in LDL receptor activity (20). Bempedoic acid is a pro-drug and conversion to its CoA-derivative is required for activity and this occurs primarily in the liver. Bempedoic acid typically lowers LDL-C by 15-25% (10,20). The effect of bempedoic acid on cardiovascular disease is currently being evaluated in a large clinical trial.

 

Niacin and fibrates, which are primarily triglyceride-lowering drugs, have been used for many years. They modestly reduce cholesterol levels as well. Their effects on ASCVD risk vary.  Niacin used alone appears to attenuate risk, but when used in combination with high-intensity statin, any incremental benefit is minimal (21).  Like niacin, fibrates moderately reduce risk for CHD when used alone in patients with hypertriglyceridemia; risk reduction is less in those who do not have elevated triglycerides (22).  When fibrates are used in combination with statins, risk for severe myopathy is greater than for statins alone. Fenofibrate is the preferred fibrate in combination with statins because it carries the lowest risk of myopathy (23). For a detailed discussion of niacin and fibrates see the Endotext chapter on Triglyceride Lowering Drugs (24). Omega-3 fatty acids also lower serum triglyceride (24). In one notable RCT, treatment of high-risk, hypertriglyceridemic patients with statin + 2 g of the omega-3 fatty acid icosapent ethyl twice daily, compared to placebo, significantly reduced the risk of ischemic events, including cardiovascular death (25). In contrast, a recent RCT that randomized high risk hypertriglyceridemic patients on statin therapy to an omega-3 carboxylic acid formulation 4 grams per day did not observe any benefits on ASCVD (26). In both trials the reduction in triglyceride levels was similar and the explanation for the different results in these trials is uncertain. For a detailed discussion of omega-3 fatty acidssee the Endotext chapter on Triglyceride Lowering Drugs (24).   Other LDL-lowering drugs include microsomal triglyceride transfer protein (MTP) inhibitors (27) and RNA antisense drugs that block hepatic synthesis of apolipoprotein B (no longer available) (28). Both of these drugs inhibit secretion of atherogenic lipoproteins into the circulation.  At present their use is restricted to patients with severe hypercholesterolemia. Evinacumab is a human monoclonal antibody against angiopoietin-like protein 3 (ANGPTL3) that is approved for the treatment of homozygous familial hypercholesterolemia (29). Evinacumab decreases LDL-C levels by approximately 50% independent of LDL receptor activity by accelerating the clearance of VLDL thereby reducing the production of LDL (30). Another class of drugs inhibits cholesterol ester transfer protein (CETP); these agents lower LDL-C levels as well as raising HDL-C (31,32).  RCTs show their benefit is small, if any, so the pharmaceutical industry shows little interest in further development and CETP inhibitors are not FDA approved.  Finally, a class of drugs inhibits a circulating protein called proprotein convertase subtilisin/kexin type 9 (PCSK9); the PCSK9 protein promotes degradation of LDL receptors and raises LDL-C levels (10).  Inhibition of PCSK9 markedly lowers LDL-C concentrations (10,33).  Recent reports indicate that PCSK9 inhibitors reduce risk in ASCVD patients at very high risk when combined with statins (34,35).  PCSK9 inhibitors are useful for patients who are statin intolerant, those with very high baseline LDL-C, such as familial hypercholesterolemia, or patients at very high risk for additional ASCVD events.  For additional information on cholesterol and triglyceride lowering drugs see the chapters in Endotext that address these topics (10,24).  

Table 3. Non-Statin Cholesterol Lowering Drugs

Drug Class

Mechanism of Action

Effects on Plasma Lipids

LDL-C lowering

Side effects

Bile acid sequestrants

Impairs reabsorption of bile acids

Raise LDL receptor activity

Reduces LDL

Raises VLDL

Minimal effect on HDL

15-25%, depending on dose

Constipation

GI distress

Increases TG

 

Ezetimibe

Impairs absorption of cholesterol

Raises LDL receptor activity

Reduces LDL

Reduces VLDL

Minimal effect on HDL

15-25%

Rare

Bempedoic acid

Inhibitor of ATP-citrate lyase leading to decreased cholesterol synthesis and an increase in LDL receptor activity

Reduces LDL

 

15-25%

Increases uric acid leading to gout

Tendon rupture has been reported

Niacin

Reduces hepatic secretion of VLDL

 

Reduces VLDL

Reduces LDL

Raises HDL

5-20%

Flushing, rash, raise plasma glucose, hepatic dysfunction, others

Fibrates

Reduces secretion of VLDL

Enhances degradation of VLDL

Reduces VLDL

(lowers TG 25-35%)

Small effect on LDL

Raises HDL

5-15%

Myopathy (in combination with statins)

Gallstones

Uncommonly various others

MTP inhibitors

Approved for treatment of homozygous familial hypercholesterolemia

Reduces hepatic secretion of VLDL

Reduces VLDL and LDL

50+%

Fatty liver

Mipomersen

(RNA antisense)

No longer available

Reduces hepatic secretion of VLDL

Reduces VLDL and LDL

50+%

Fatty liver

CETP inhibitors

Not approved by FDA

 

Blocks transfer of cholesterol from HDL to VLDL&LDL

Raises HDL

Lowers LDL

20-30%

 

 

PCSK9 inhibitors

Recommended for ASCVD patients at high risk

Blocks effects of PCSK9 to destroy LDL receptors

Lowers LDL

45-60%

 

Evinacumab

Approved for treatment of homozygous familial hypercholesterolemia

Blocks angiopoietin-like protein 3 (ANGPTL3)

Lowers LDL

Lowers TG (~50%)

Lowers HDL (~30%)

Approx. 50%

 

 

HISTORY OF U.S. GUIDELINES FOR CHOLESTEROL MANAGEMENT

 

National Cholesterol Education Program (NCEP)

 

The most influential guidelines for cholesterol management in the United States have been those developed by the NECP. This program was sponsored by the National Heart, Lung and Blood Institute and included many health-related organizations in the United States (36).  Between 1987 and 2004, three major Adult Treatment Panel (ATP) reports (4,37,38) and one update were published (39) (Table 4).

 

Table 4. National Cholesterol Education Program’s Adult Treatment Panel (ATP) Reports

Guideline

ATP I

ATP II

ATP III

ATP III Update

Year

1987

1994

2001

2004

Thrust

Primary prevention

Secondary prevention

High-risk primary prevention

Very high risk

Drugs

Bile acid resins Nicotinic acid Fibrates

Same as ATPI   +Statins

Same as ATP II

 

Same as ATP III

Major Targets

LDL-C; HDL-C

LDL-C; HDL-C

LDL-C;                Non-HDL-C

LDL-C;         Non-HDL-C

LDL-C goal

     (mg/dL)

Low risk <190 Moderate risk <160              High risk < 130

Low risk   <160 Moderate risk <130             High risk <100

Low risk <160 Moderate risk <130          Moderately high risk <130    

High risk < 100

Low risk <160 Moderate risk <130

Moderately high    risk <130      High risk < 100   Very high risk < 70

 

ATP reports identified LDL-C as the major target of cholesterol-lowering therapy. The intensity of LDL-lowering therapy was based on aggregate knowledge from multiple sources in the cholesterol field. Priority was given to the clinical trial evidence when available. ATP I (1987) emphasized lifestyle therapy for primary prevention. Use of cholesterol-lowering drugs was down-played in ATP I.  ATP II (1993) placed more emphasis on secondary prevention; this was because a large meta-analysis of RCTs using cholesterol-lowering drugs confirmed CHD risk reduction. ATP III (2001) added more emphasis on high-risk primary prevention.  At each successive ATP report, the intensity of LDL lowering therapy was increased with lower LDL-C goals.

 

The NCEP put highest priority for cholesterol management for patients with clinical forms of atherosclerotic disease. The latter included coronary heart disease, clinical carotid artery disease, peripheral arterial disease, and abdominal aortic aneurysm. ASCVD is the inclusive term for these conditions. The 10-year risk for future cardiovascular events in patients with established ASCVD is usually > 20%.  In ATP III, the presence of ASCVD of any type warranted an LDL-C goal of < 100 mg/dL.  For high-risk patients with hypertriglyceridemia, a non-HDL-C goal of < 130 mg/dL was recommended.

 

For primary prevention, ATP III identified four levels of risk for increasing intensity of LDL-C lowering. Different LDL-C goals were set for different levels of risk (Table 4). Risk for CHD was calculated using Framingham risk scoring. Framingham risk factors included cigarette smoking, hypertension, elevated total cholesterol, low HDL-C, and advancing age. A 10-year risk > 20% for CHD was called high risk. Moderately high risk was defined as a 10-year risk of 10-19%; at this level of risk, cholesterol-lowering drugs were considered to be cost-effective.  A 10-year risk of < 10% was divided into moderate risk and low risk depending on the presence or absence of major risk factors. Moderate risk corresponds to a 10-year risk for CHD of approximately 5-9%.   Generally speaking, cholesterol-lowering drugs were not recommended for low- to- moderate risk individuals except when LDL-C levels are high.

 

In 2004, ATP III underwent an update and set an optional LDL-C goal of < 70 mg/dL for patients deemed to be at very high risk for future CHD events. This option included CHD plus other atherosclerotic conditions and/or multiple major risk factors. This progression of treatment intensity was made possible by the results of several clinical trials with statin therapy.

 

Transfer of NHLBI Guidelines to American Heart Association (AHA) and the American College of Cardiology (ACC)

 

In 2013, NHLBI made the decision to remove treatment guidelines from its agenda. This was done even though it had almost finished writing prevention guidelines. These included guidelines for high blood cholesterol, high blood pressure, obesity, and nutrition. Late in this process, the guideline process was transferred to the (American Heart Association) AHA and American College of Cardiology (ACC). Then in 2013 the NHLBI guidelines for high blood cholesterol were modified to fit the criteria for guideline development required by AHA/ACC. The 2013 cholesterol guidelines (40) adhered closely to the Institute of Medicine (National Academy of Medicine) recommendations for evidence-based guidelines (41). These recommendations advocated priority to randomized controlled trials (RCTs) as the foundation of evidence-based medicine. The NHLBI cholesterol committee carried out an extensive review of the literature and limited recommendations based largely to RCTs. Most acceptable RCTs had utilized statin therapy in middle-aged persons. Therefore, the 2013 report committee did not include detailed recommendations for younger or older adults.  Recommendations were largely limited to the age range 40-75 years. High-intensity statin therapy was recommended for patients with established ASCVD. For primary prevention, risk was stratified by use of a pool cohort equation (PCE), which are derived from five large population studies in the United States (42). The PCE was an extension of the Framingham Heart Study risk equations. 10-year risk for ASCVD was based on the following risk factors: age, gender, cigarette smoking, blood pressure, total cholesterol, HDL cholesterol, and presence or absence of diabetes.  Although the PCE was validated in another large study (43), it has been criticized by some investigators as being imprecise for many individuals or specific groups (44-48).

 

For primary prevention, an effort was made to determine what level 10-year risk is associated with efficacy of reduction of ASCVD from statin RCTs.  It was determined that statins are effective for risk reduction when 10-year risk for ASCVD is > 7.5%.  Most primary prevention trials employed moderate intensity statins, so these were recommended for most patients; but in one RCT (49), a high-intensity statin appeared to produce greater risk reduction than found with moderate-intensity statins. So high-intensity statins were considered a favorable option in patients at higher 10-year risk. Notably LDL-C goals were not emphasized. It was recognized that these recommendations may not be optimal for all patients; therefore, consideration should be given to any extenuating circumstances that could modify the translation of RCTs directly into clinical care. A clinician patient risk discussion thus was advocated for all patients to consider the pros and cons of statin therapy.

 

2018 AHA/ACC/MULTI-SOCIETY REPORT

 

2018 cholesterol guidelines were revised by AHA/ACC in collaboration with multiple other societies concerned with preventive medicine (1).  These guidelines extended those published in 2013. They expanded recommendations to include children, adolescents, young adults (20-39 years), and older patients (> 75 years). Although RCTs may be lacking in these categories, epidemiology and clinical studies indicate that high blood cholesterol is an important risk factor for future ASCVD in these age ranges. From the evidence acquired over many years related to the cholesterol hypothesis, it is reasonable to craft recommendations based on the totality of the evidence. In the following, 2018 guidelines will be highlighted in relation to general areas of cholesterol management as identified by all the previous national and international guidelines. These guidelines proposed a top-10 list of recommendations to highlight the key points. These key points will be examined.

 

Lifestyle Intervention

 

1. IN ALL INDIVIDUALS, EMPHASIZE HEART-HEALTHY LIFESTYLE ACROSS THE LIFE-COURSE.

 

There is widespread agreement in the cardiovascular field that lifestyle factors contribute to the risk for ASCVD. These factors include cigarette smoking, sedentary life habits, obesity, and an unhealthy eating pattern. The ACC/AHA strongly recommends that a healthy lifestyle be adopted throughout life. These recommendations are strongly supported by 2018 cholesterol guidelines. They are the foundation for cardiovascular prevention and should receive appropriate attention in clinical practice (50). For a detailed discussion of the effect of diet on lipid levels and atherosclerosis see the Endotext chapter The Effect of Diet on Cardiovascular Disease and Lipid and Lipoprotein Levels (51).

 

Secondary Prevention

 

2. IN PATIENTS WITH CLINICAL ASCVD, REDUCE LDL-C WITH HIGH-INTENSITY STATINS OR MAXIMALLY TOLERATED STATINS TO DECREASE ASCVD RISK. THE GOAL OF THERAPY IS TO REDUCE LDL-C BY > 50%. IF NECESSARY TO ACHIEVE THIS GOAL, CONSIDER ADDING EZETIMIBE TO MODERATE INTENSITY STATIN THERAPY.

 

The strongest evidence for efficacy of statin therapy is a meta-analysis of RTCs carried out in patients with established ASCVD.  As previously mentioned, the best fit line comparing percent ASCVD versus LDL-C in secondary prevention studies demonstrates that for every mmol/L (39mg/dL) reduction in LDL-C the risk for ASCVD is decreased by approximately 22% (11). High intensity statins typically reduce LDL-C by 50% or more; this percentage reduction occurs regardless of baseline levels of LDL-C. This explains why the guidelines set a goal for LDL-C secondary prevention to be a > 50% reduction in levels.  There are two options to achieve such reductions. RCTs give priority to use of high-intensity statins. But second, if high-intensity statins are not tolerated, similar LDL-C lowering can be attained by combining a moderate-intensity statin with ezetimibe (18). An approach to lowering LDL-C in patients with ASCVD is shown in Figure 2.

 

Figure 2. Secondary Prevention in Patients with Clinical ASCVD (1)

 

3. IN VERY HIGH-RISK PATIENTS WITH ASCVD, FIRST USE A MAXIMALLY TOLERATED STATIN + EZETIMIBE TO ACHIEVE AN LDL-C GOAL OF < 70 MG/DL (<1.8 MMOL/L). IF THIS GOAL IS NOT ACHIEVED, CONSIDER ADDING A PCSK9 INHIBITOR.

 

2018 guidelines defined very high risk of future ASCVD events as a history of multiple ASCVD events or one major event plus multiple high-risk conditions (Table 5). This definition is based in large part on subgroup analysis of the IMPROVE-IT trial (18,19).

 

Table 5. Very High Risk of Future ASCVD Events (1)

Major ASCVD Events

Recent ACS (within the past 12 months)

History of MI (other than recent acute coronary syndrome event listed above)

History of ischemic stroke

Symptomatic peripheral arterial disease (history of claudication with ABI <0.85, or previous revascularization or amputation)

High Risk Conditions

Age ≥65 y

Heterozygous familial hypercholesterolemia

History of prior coronary artery bypass surgery or percutaneous coronary intervention outside of the major ASCVD event(s)

Diabetes mellitus

Hypertension 

CKD (eGFR 15-59 mL/min/1.73 m2)

Current smoking

Persistently elevated LDL-C (LDL-C ≥100 mg/dL [≥2.6 mmol/L]) despite maximally tolerated statin therapy and ezetimibe

History of congestive heart failure

ABI indicates ankle-brachial index; CKD indicates chronic kidney disease

 

Recent RCTs have demonstrated that addition of non-statins to statin therapy can enhance risk reduction. These RCTs (and their add-on drugs) were IMPROVE-IT (ezetimibe) (18), FOURIER (evolocumab) (34), and ODYSSEY OUTCOMES (alirocumab) (35).  All RCTs were carried out in patients at very high-risk.  For IMPROVE-IT, addition of ezetimibe to statin therapy produced an additional 6% reduction in ASCVD events. In this trial, baseline LDL-C on moderate-intensity statin alone averaged about 70 mg/dL; in spite of this low level, further LDL lowering with addition of ezetimibe enhanced risk reduction. RCTs with the two PCSK9 inhibitors (evolocumab and alirocumab) restricted recruitment to patients having LDL-C > 70 mg/dL on maximally tolerated statin+ ezetimibe. In these RCTs, duration of therapy was only about 3 years. A marked additional LDL lowering was achieved. In both trials, risk for ASCVD events was reduced by 15%.

 

2018 guidelines allow consideration of PCSK9 inhibitor as an add-on drug if patients are at very high risk for future ASCVD events and have an LDL-C > 70 mg/dL during treatment with maximally tolerated statin plus ezetimibe (Figure 3). This latter threshold LDL-C was chosen because it was a recruitment criteria for PCSK9 inhibitor therapy in reported RTCs (34,35)

 

An important question about use of PCSK9 inhibitors is whether they are cost-effective   When they first became available, they were marketed at a very high cost, which was widely considered to be excessive. More recently, the cost of these drugs has declined considerably.   An analysis of cost-effectiveness has shown that at current prices in very high-risk patients PCSK9 inhibitors can be cost-effective (52).  Another analysis (53) of approximately 1 million patients with ASCVD in the Veterans Affairs system indicate that approximately 10% of patients will be classified as very high risk and having LDL-C > 70 mg/dL while taking maximal statin therapy plus ezetimibe. These later patients are potential candidates for PCSK9 inhibitors. 

 

Figure 3. Secondary Prevention in Patients with Very High-Risk ASCVD (1)

 

 Primary Prevention

 

4. IN PATIENTS WITH SEVERE PRIMARY HYPERCHOLESTEROLEMIA (LDL-C ≥190 MG/DL (≥4.9 MMOL/L)), WITHOUT CONCOMITANT ASCVD, BEGIN HIGH-INTENSITY STATIN THERAPY (OR MODERATE INTENSITY STATIN + EZETIMIBE) TO ACHIEVE IN LDL-C GOAL OF < 100 MG/DL; IF THIS GOAL IS NOT ACHIEVED, CONSIDER ADDING PCSK9 INHIBITOR IN SELECTED PATIENTS AT HIGHER RISK. MEASUREMENT OF 10-YEAR RISK FOR ASCVD IS NOT NECESSARY.

 

Patients with severe hypercholesterolemia are known to be at relatively high risk for developing ASCVD (54,55). In view of massive evidence that elevated LDL-C promotes atherosclerosis and predisposes to ASCVD, it stands to reason that such patients deserve intensive treatment with LDL-lowering drugs. RCTs with cholesterol-lowering drugs demonstrate benefit of statin therapy in patients with severe hypercholesterolemia (56,57). It is not necessary to calculate 10-year risk in such patients. Moreover, patients who have extreme elevations of LDL-C (e.g., heterozygous familial hypercholesterolemia) may be candidates for PCSK9 inhibitors if LDL-C cannot be lowered sufficiently with maximal statin therapy plus ezetimibe.

 

5. IN PATIENTS WITH DIABETES MELLITUS AGED 40 TO 75 YEARS WITH AN LDL-C ≥70 MG/DL (≥1.8 MMOL/L), WITHOUT CONCOMITANT ASCVD, BEGIN MODERATE-INTENSITY STATIN THERAPY. FOR OLDER PATIENTS (>50 YEARS), CONSIDER USING HIGH-INTENSITY STATIN (OR MODERATE INTENSITY STATIN PLUS EZETIMIBE) TO ACHIEVE A REDUCTION IN LDL-C OF > 50%. MEASUREMENT OF 10-YEAR RISK FOR ASCVD IS NOT NECESSARY.

 

Middle-aged patients with diabetes have an elevated lifetime risk for ASCVD (58). The trajectory of risk is steeper in patients with diabetes than in those without. For this reason, estimation of 10-year risk for ASCVD with pooled cohort equation (PCE) is not a reliable indicator of lifetime risk.  Meta-analysis of RCTs in middle-aged patients with diabetes treated with moderate intensity statins therapy shows significant risk reduction (14). Hence, most middle-aged patients with diabetes deserve statin therapy. It is not necessary to measure 10-year risk before initiation of statin therapy in these patients. With progression of age and accumulation of, multiple risk factors, increasing the intensity of statin therapy or adding ezetimibe seems prudent (Tables 6 and 7).

 

Table 6. Diabetes Specific Risk Enhancers That Are Independent of Other Risk Factors in Diabetes (1)

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

Albuminuria ≥30 mcg of albumin/mg creatinine

eGFR <60 mL/min/1.73 m2

Retinopathy

Neuropathy

ABI <0.9

ABI indicates ankle-brachial index

 

Table 7. ASCVD Risk Enhancers (1)

Family history of premature ASCVD

Persistently elevated LDL > 160mg/dl (>4.1mmol/L

Chronic kidney disease

Metabolic syndrome

History of preeclampsia

History of premature menopause

Inflammatory disease (especially rheumatoid arthritis, psoriasis, HIV)

Ethnicity (e.g., South Asian ancestry)

Persistently elevated triglycerides > 175mg/dl (>2.0mmol/L)

Hs-CRP > 2mg/L

Lp(a) > 50mg/dl or >125nmol/L

Apo B > 130mg/dl

Ankle-brachial index (ABI) < 0.9

 

6. INITIATION OF PRIMARY PREVENTION SHOULD BEGIN WITH A CLINICIAN-PATIENT RISK DISCUSSION.

 

This discussion is necessary to put a patient’s total risk status in perspective. The risk discussion should always begin with a review of the critical importance of lifestyle intervention. This is true for all age groups. Beyond the issue of lifestyle, the discussion can further consider the potential benefit of a cholesterol-lowering drug, especially statin therapy. When the latter may be beneficial, the provider should next review major risk factors and estimated 10-year risk for ASCVD derived from the pooled cohort equation (PCE) risk calculator (59) (https://www.acc.org/guidelines/hubs/blood-cholesterol). Estimation of lifetime risk is also useful, particularly in younger individuals. All major risk factors (e.g., cigarette smoking, elevated blood pressure, LDL-C, hemoglobin A1C [if indicated], should be discussed. In patients 40-75 years, the 10-year risk estimate is most useful. In these patients, four categories of 10-year risk for ASCVD are recognized: low risk (<5%); borderline risk (5-7.4%); intermediate risk (7.5-19.9 %), and high risk (> 20%). Estimates of lifetime risk for patients 20-39 years also are available (https://www.acc.org/guidelines/hubs/blood-cholesterol  or        https://qrisk.org/lifetime/index.php).  Three other components of the risk discussion are: risk enhancing factors (see #8), possible measurement of coronary artery calcium (CAC) (see #9), and a review of extenuating life circumstances (issues of cost and safety considerations, as well as patient motivation and preferences). The decision to initiate statin therapy should be shared between clinician and patient. All of these factors deserve a full discussion in view of the fact that statin therapy represents a lifetime commitment to taking a cholesterol-lowering drug.

 

Patients should also recognize that atherosclerosis begins early in life and progresses overtime before manifesting as clinical disease. The cumulative LDL-C levels (“LDL-C years”) strongly influence the timing of clinical manifestation (figure 4). In patients with high cholesterol levels (homozygous and heterozygous familial hypercholesterolemia) ASCVD can occur early in life whereas in patients with loss of function mutations in PCSK9 and low cholesterol level have a reduced occurrence of ASCVD.

 

Figure 4. Relationship between cumulative LDL-C exposure, age, and the development of the clinical manifestations of ASCVD. Figure from reference (60).

 

Additionally, patients should be appraised of comparisons of the reduction in ASCVD events in individuals with genetic variations resulting in life-long reductions in LDL-C levels vs. individuals treated with statins to lower LDL-C later in life. Variants in the HMG-CoA reductase, NPC1L1, PCSK9, ATP citrate lyase, and LDL receptor genes result in a lifelong decrease in LDL-C and a 10mg/dL decrease in LDL-C with any of these genetic variants was associated with a 16-18% decrease in ASCVD events (61). As noted above a 39mg/dL decrease in LDL-C in the statin trials resulted in a 22% decrease in ASCVD events. Thus, a life-long decrease in LDL-C levels results in a decrease in ASCVD events that is three to four times as great as that seen with short-term LDL-C lowering with drugs later in life suggesting that the sooner the LDL-C level is lowered the better the prevention of cardiovascular events.

 

7. IN ADULTS 40 TO 75 YEARS OF AGE WITHOUT DIABETES AND LDL-C ≥70 MG/DL (≥1.8 MMOL/L), RTC'S SHOW THAT MODERATE INTENSITY STATIN THERAPY IS EFFICACIOUS WHEN 10-YEAR RISK FOR DEVELOPING ASCVD IS > 7.5%. THEREFORE, INITIATING STATIN THERAPY SHOULD BE CONSIDERED IN THE RISK DISCUSSION.

 

A 10-year risk > 7.5% does not mandate statin therapy but indicates that moderate-intensity statins can reduce risk by 30-40% with a minimum of side effects (62). This fact alone can justify moderate intensity statin therapy, but only if other considerations noted above (#6) are taken into account in the risk discussion. An approach to lipid lowering in primary prevention patients is shown in figure 5.

 

Figure 5. Approach to Primary Prevention in Patients without LDL-C >190mg/dl or Diabetes (1)

 

8. DETERMINE PRESENCE OF RISK-ENHANCING FACTORS IN ADULTS 40 TO 75 YEARS OF AGE TO INFORM THE DECISION REGARDING INITIATION OF STATIN THERAPY.

 

If risk assessment based on PCE is equivocal or ambiguous, the presence of risk enhancing factors in patients at intermediate risk (10-year risk 7.5 to 19.9%), can tip the balance in favor of statin therapy. Risk enhancing factors are shown in Table 7.

 

9. IF A DECISION ABOUT STATIN THERAPY IS UNCERTAIN IN ADULTS 40 TO 75 YEARS OF AGE WITHOUT DIABETES MELLITUS, WITH LDL-C LEVELS ≥ 70 MG/DL, AND WITH A 10 YEAR ASCVD RISK OF ≥ 7.5% TO 19.9% (INTERMEDIATE RISK) CONSIDER MEASURING CAC.

 

CAC measurements are a safe and inexpensive method to assess severity of coronary atherosclerosis. CAC scores generally reflect lifetime exposure to coronary risk factors and therefore in young individuals (men < 40 years of age; women < 50 years of age) the long-term predictive value is limited because the CAC score is often 0. Studies show that CAC accumulation is a strong predictor of probability of ASCVD events (63). A CAC core of zero generally is accompanied by few if any ASCVD events over the subsequent decade. A CAC score of 1-100 Agatston units is associated with relatively low rates of ASCVD, both in middle-aged and older patients. In contrast, a CAC >100 Agatston units carries a risk well into the statin-benefit zone.  Data such as these led to the following recommendation of 2018 guidelines for patients at intermediate risk by PCE.

 

  1. If CAC is zero, treatment with statin therapy may be withheld or delayed, except in cigarette smokers, those with diabetes mellitus, those with a strong family history of premature ASCVD, and possibly chronic inflammatory conditions such as HIV.
  2. A CAC score of 1 to 99 Agatston units favors statin therapy in intermediate-risk patients ≥55 years of age, whereas benefit in 40-54 years is marginal (note these focuses on 10-year risk and a CAC score in this range in a younger individual is predictive of long-term risk (64)).
  3. A CAC score ≥100 Agatston units (or ≥75th percentile), strongly favors statin therapy, unless otherwise countermanded by clinician–patient risk discussion.

 

Monitoring

 

10. ASSESS ADHERENCE AND PERCENTAGE RESPONSE TO LDL-C LOWERING MEDICATIONS AND/OR LIFESTYLE CHANGES WITH REPEAT LIPID MEASUREMENT 4 TO 12 WEEKS AFTER STATIN INITIATION OR DOSE ADJUSTMENT AND EVERY 3-12 MONTHS AS NEEDED.

 

Remember that the LDL-C goal for patients with ASCVD or severe hypercholesterolemia is a > 50% reduction in LDL-C. For most such patients, this goal can be achieved by high-intensity statin therapy + ezetimibe. In ASCVD patients at very high risk, the goal is an LDL-C lowering >50% and LDL-C < 70 mg/dL. To achieve these goals, it may be necessary to combine a PCSK9 inhibitor with maximal statin therapy + ezetimibe.  For statin therapy in primary prevention, the goal is a lowering of > 35%. This goal can be achieved in most patients with a moderate intensity statin + ezetimibe

 

 2018 guidelines did not set a precise on-treatment LDL-C target of therapy, but instead, offer percent reductions as goals of therapy. Baseline levels of LDL-C can be obtained either by chart review or withholding statin therapy for about two weeks. In addition, on-treatment LDL-C can provide useful information about efficacy of treatment (Figure 6). This figure shows expected LDL-C levels for 50% or 35% reductions at different baseline levels of LDL-C.  For example, in secondary prevention, an on-treatment LDL-C of <70 mg/dL can be considered adequate treatment regardless of baseline LDL-C. On-treatment levels in the range of 70-100 mg/dL are adequate if baseline-LDL C is known to be in the range of 140- 200 mg/dL; if there is uncertainty about baseline levels, reevaluation of statin adherence and reinforcement of treatment regimen is needed. For optimal treatment, on-treatment levels in this range warrant consideration of adding ezetimibe to maximal statin therapy. If on treatment LDL-C is > 100 mg/dL, the treatment regimen is probably inadequate, and intensification of therapy is needed. For primary prevention, the LDL-C goal is a reduction > 35%, and a similar scheme for evaluating efficacy of therapy can be used.

 

Figure 6. Predicted on-treatment LDL-C compared to baseline LDL-C and suggested actions for each category of on-treatment LDL-C in secondary and primary prevention.

 

Other Issues

 

OTHER AGE GROUPS

 

2018 guidelines offered suggestions for management of high blood cholesterol in children, adolescents, young adults (20-39 years), and elderly patients > 75 years. There is no strong RCT evidence to underline cholesterol management in these populations. Instead, treatment suggestions depend largely on epidemiologic data. Lifestyle intervention is a primary method for cholesterol treatment in these age groups. However, under certain circumstances LDL-lowering drugs may be indicated. This is particularly the case for patients with familial hypercholesterolemia or similar forms of very high LDL-C. In young adults, particularly those with other risk factors, LDL lowering drug therapy (statin or ezetimibe) may be reasonable when LDL-C levels are in the range of 160-189 mg/dL or if the lifetime risk is high. Older adults having concomitant risk factors are potential candidates for initiation of statins or continuation of existing statin therapy. In all cases, clinical estimation of risk status is critical in a decision to initiate statins.

 

For details on the approach to treating hypercholesterolemia in older adults see the Endotext chapter entitled “Management of Dyslipidemia in the Elderly” (65). For details on the approach to treating hypercholesterolemia children and adolescence see the Endotext section on Pediatric Lipidology.

 

STATIN NON-ADHERENCE   

 

 In spite of proven benefit of statin therapy in high-risk patients, there is a relatively high prevalence of nonadherence to the prescribed drug (66). Some studies suggest that up to 50% of patients discontinue use of prescribed statins over the long run (67-70). This finding creates a major challenge to the health care system for prevention of ASCVD. Table 8 lists several factors that may contribute to a high prevalence of nonadherence.  

Table 8. Factors Associated with Statin Nonadherence

Healthcare system factors

Accompanying medical care costs

Lack of medical oversight and follow-up (provider therapeutic inertia)

Provider concern for side effects

Patient factors

Uncertainty of benefit

Lack of health consciousness

Lack of motivation

Lack of perceived benefit

Perceived side effects

Nocebo effects

Myalgias

Myopathy

“Brain fog”

Misattributed symptoms or syndromes (arthritis, spondylosis, neuropathy, insomnia, mental confusion and memory loss, fibromyalgia, gastrointestinal symptoms, liver dysfunction, cataract; cancer).

 When a decision is made to initiate statin therapy, the presumption is that statins are a lifetime treatment. Their use is similar to other medications, such as antihypertensive drugs, which are expected to be taken for the rest of one’s life. Such treatments imply indefinite participation in the healthcare system. This means regular ongoing visits to a prescribing clinic. Even for those with medical insurance there are usually co-pays both for the visit and for medication, not to mention cost of transportation to and from the clinic. All of these cost-related issues can be an impediment to long-term statin usage. Provider therapeutic inertia (66) can result from lack of provider education, excessive workload, and concerns about statin side effects. From the patient’s point of view, common issues are lack of understanding of the potential benefits of therapy and lack of health consciousness and motivation. A related problem is expectation of side effects because of preconditioning by information received from the news media, package inserts, Internet, family, and friends. This expectation can discourage individuals from continuation of statin therapy (nocebo effect) (71). The most common symptoms attributed to statin therapy are muscle pain and tenderness (myalgias) (10).  A complaint of statin intolerance is registered in about 5-15% of patients. If myalgias attributed to statins are due to actual pathological changes, the character of the changes is yet to be determined. In almost all cases, serum creatine kinase (CK) levels are not increased. There is no evidence for long-term muscle damage. A few reports nonetheless suggest that statins can produce a low-grade myopathy (72); such an effect has not been widely accepted. The literature is replete with case reports of other symptoms attributed to statins (66). In fact, much of the symptomology reported by patients are unrelated to statin treatment but are in fact the symptoms of other conditions. Statin therapy has been given to large numbers of people for many years without evidence of long-term muscle dysfunction. 

Still, in rare cases, especially when blood levels of statins are raised, severe myopathy (rhabdomyolysis) can occur. This proves that statins can be myotoxic. Table 9 lists conditions associated with statin-induced severe myopathy (73,74). In most such cases, severe myopathy is reversible. If the cause can be identified and eliminated, a statin can be cautiously reinstituted. Alternatively, a non-statin LDL-lowering drug (e.g., ezetimibe, bempedoic acid, or PCSK9 inhibitor) can be substituted for the offending statin (10,75).

 

Table 9. Factors Associated with Statin - Induced Rhabdomyolysis

Advanced age (>80 y)Small body frame and fragilityFemale sexAsian ethnicityPre-existing neuromuscular conditionKnown history of myopathy or family history of myopathy syndromePre-existing liver disease, kidney disease, hypothyroidismCertain rare genetic polymorphismsHigh-dose statin (?)Postoperative periodsExcessive alcohol intakeDrug interactions (gemfibrozil, antipsychotics, amiodarone, verapamil, cyclosporine, macrolide antibiotics, azole antifungals, protease inhibitors)

 

These considerations indicate that statin therapy is a much greater investment in time and effort than commonly recognized. Since statins have the potential to prevent many ASCVD events, they offer great potential in clinical management of patients at risk. Nonetheless, to achieve this benefit, the health care system must be adjusted to the requirements of statin therapy as well as other risk-reducing therapies. Unless these adjustments are made, much of the potential benefit of statin treatment will be lost. It will be necessary to address all the components of healthcare and patient factors to improve long-term adherence of statin therapy.

 

EUROPEAN GUIDELINES FOR CHOLESTEROL MANAGEMENT

 

The most influential of European guidelines for management of cholesterol and dyslipidemia are those developed by the European Society of Cardiology (ESC), the European Atherosclerosis Society (EAS), and representatives from other European organizations (76). A task force appointed by these organizations have published an update on dyslipidemia management (77). The recommendations of this report resemble in many ways those of the 2018 AHA/ACC guidelines (1). But notable differences can be identified for specific recommendations. A review of these differences may help to identify gaps in knowledge needed to format best recommendations. In the following, recommendations proposed by AHA/ACC and by ESC/EAS will be compared. These comparisons should illuminate areas of uncertainty where more information is needed for definitive recommendations. At the same time, it is important to emphasize that in many critical areas the two sets of guidelines are in strong agreement. These will be noted first.

 

Agreement Between AHA/ACC and ESC/EAS Guidelines

 

There is agreement that elevated LDL is the major atherogenic lipoprotein and that LDL-C is the primary target of treatment. Likewise, both guidelines agree that the intensity of LDL-C lowering therapy should depend on absolute risk to patients. In other words, patients who have highest risk should receive the most intensive cholesterol reduction. Both guidelines emphasize therapeutic lifestyle intervention as the foundation of risk reduction, both for elevated cholesterol and for other risk factors. The highest risk patients are those with atherosclerotic disease and are potential candidates for combined drug therapy for cholesterol-lowering. For primary prevention, the intensity of treatment depends on absolute risk as determined by population-based algorithms.  For drug therapy, statins are first-line treatment, but in highest risk patients, consideration can be given to adding non-statin drugs (e.g., ezetimibe and PCSK9 inhibitors).   Beyond population-based algorithms for primary prevention, measurement of other dyslipidemia markers or other higher risk conditions can be used as risk- enhancing factors to modify intensity of lipid-lowering therapy.  

 

Differences Between AHA/ACC and ESC/EAS Guidelines

 

DEFINITION OF VERY HIGH RISK  

 

This definition is important because it sets the stage for considering combined drug therapy for LDL-C lowering. AHA/ACC defines very high risk as a history of multiple ASCVD events or of one event + multiple high-risk conditions.  This limits the definition of very high risk to the highest risk patients among those with ASCVD. In contrast, ESC/EAS considers all patients with clinical ASCVD or ASCVD on imaging as very high risk. Additionally, ESC/EAS allows extension of the definition to highest risk patients in primary prevention, that is, to patients with multiple risk factors and/or subclinical atherosclerosis (table 10). Overall, more patients will be identified as being at very high risk by ESC/EAS guidelines. This could enlarge the usage of PCSK9 inhibitors. AHA/ACC limits the use of PCSK9 inhibitors to patients at highest risk, because of their high cost. One recent study (53) showed that only about 10% of patients with established ASCVD will be eligible for PCSK9 inhibitors by AHA/ACC recommendations.

 

Table 10. ESC/EAS Cardiovascular Risk Categories

Very High-Risk

Ø  ASCVD, either clinical or unequivocal on imaging

Ø  DM with target organ damage or at least three major risk factors or T1DM of long duration (>20 years)

Ø  Severe CKD (eGFR <30 mL/min/1.73 m2)

Ø  A calculated SCORE >10% for 10-year risk of fatal CVD.

Ø  FH with ASCVD or with another major risk factor

High-Risk

Ø  Markedly elevated single risk factors, in particular Total Cholesterol >8 mmol/L (>310mg/dL), LDL-C >4.9 mmol/L (>190 mg/dL), or BP >180/110 mmHg.

Ø  Patients with FH without other major risk factors.

Ø  Patients with DM without target organ damage, with DM duration > 10 years or another additional risk factor.

Ø  Moderate CKD (eGFR 30-59 mL/min/1.73 m2).

Ø  A calculated SCORE >5% and <10% for 10-year risk of fatal CVD.

Moderate Risk

Ø  Young patients (T1DM <35 years; T2DM <50 years) with DM duration <10 years, without other risk factors.

Ø  Calculated SCORE >1 % and <5% for 10-year risk of fatal CVD.

Low Risk

Ø  Calculated SCORE <1% for 10-year risk of fatal CVD

 

GOALS FOR LDL-C   

 

In 2013, the AHA/ACC eliminated specific numerical goals for LDL-C in both primary and secondary prevention.  Recommendations for LDL-C lowering therapy were based exclusively on RCTs of statin therapy. These recommendations have been criticized for lacking a means to evaluate efficacy of statin therapy. In 2018, AHA/ACC identified 2 goals for LDL-C lowering, namely, > 50% LDL-C reduction in secondary prevention and > 35% reduction in primary prevention. These values are based on the expected reductions achieved by high-intensity statins for secondary prevention and by moderate-intensity statins for primary prevention.  Again, no numerical targets are identified. The only exception was the recognition of an LDL-C threshold goal of 1.8 mmol/L (70 mg/dL) for consideration of PCSK9 inhibitors in very high-risk patients on maximal statin therapy + ezetimibe.

 

ESC/EAS supports the 50% reduction of LDL-C in high-risk patients but also includes a goal of <1.8 mmol/L (70 mg/dL). This goal applies to all high-risk patients, whether in primary or secondary prevention. For very high-risk patients, the goal is an LDL-C of < 1.4 mmol/L (55 mg/dL). For moderate-risk patients in primary prevention, the goal is LDL-C <2.6 mmol/L (100 mg/dL). The guideline task force presumably believed that having defined LDL-C goals facilitates cholesterol-lowering therapy in clinical practice. Additionally, following the ESC/EAS LDL-C goals will most likely result in lower LDL-C levels in many patients.  

 

Table 11. ESC/EAS LDL Cholesterol Goals

Very High Risk

LDL-C reduction of >50% from baseline and an LDL-C goal of <1.4 mmol/L (<55 mg/dL) is recommended

High Risk

LDL-C reduction of >50% from baseline and an LDL-C goal of <1.8 mmol/L (<70 mg/dL) is recommended

Moderate Risk

LDL-C goal of <2.6 mmol/L (<100 mg/dL) should be considered

Low Risk

LDL-C goal <3.0 mmol/L (<116 mg/dL) may be considered.

 

RISK ESTIMATION FOR PRIMARY PREVENTION

 

AHA/ACC employed a pooled cohort equation (PCE) developed from five large population groups in the USA to estimate 10-year risk (and lifetime risk) for ASCVD events. ESC/EAS for several years has employed a SCORE algorithm based on risk for ASCVD mortality in European populations. Both PCE and SCORE are used to define “statin eligibility” for primary prevention. A study suggests that more people are “eligible” for statin therapy using PCE compared to SCORE (78). If this finding can be confirmed, it suggests that ESC/EAS guidelines are less aggressive for reducing LDL-C in lower risk individuals (compared to AHA/ACC guidelines). In contrast, ESC/EAS appears to be more aggressive in use of non-statins for LDL lowering in higher risk patients than is AHA/ACC.

 

RISK ENHANCING FACTORS   

 

AHA/ACC proposed that several risk enhancing factors favor the decision to use statin therapy in patients at intermediate risk. Although European guidelines did not specify a list of such factors, most were considered to justify more intensive therapy. Notable among risk enhancing factors were apolipoprotein B (apoB) and lipoprotein (a) (Lp[a]). ESC/EAS seemingly placed more emphasis on these two factors for adjusting intensity of therapy; this report’s recommendations can be taken to mean that apoB and Lp(a) should be measured more frequently in risk assessment than stated by AHA/ACC.  In fact, neither guideline was highly specific as to when to exercise the option of their measurements. This option depends largely on clinical judgment.

 

SUBCLINICAL ATHEROSCLEROSIS  

 

AHA/ACC propose that CAC measurement can assist in deciding whether to use statin therapy in patients at intermediate risk. AHA/ACC in particular noted that the absence of CAC justifies delaying statin therapy. No other modalities of measurement of subclinical atherosclerosis were advocated by AHA/ACC. In contrast, ESC/EAS supported use of different modes of cardiovascular imaging to assist in decisions about intensity of LDL-C lowering therapy. Beyond this, however, recommendations for cardiovascular imaging were not highly specific.  Nonetheless, these guidelines suggest that the finding of substantial subclinical atherosclerosis in any arterial bed elevates a patient’s risk to the category of established ASCVD and can justify adding non-statin therapy to statins in such patients.

 

GUIDELINE SPECIFICITY

 

AHA/ACC guidelines place great emphasis on data from RCTs to justify its recommendations.  However, RTC’s related to specific questions typically are limited in number. AHA/ACC recommendations are highly codified and kept to a minimum. ESC/EAS in contrast bases its recommendations both on clinical trials and other types of evidence. It explores available evidence in greater detail, and many of its recommendations are more nuanced. This approach to guideline development has its advantages and disadvantages. For example, it gives the reader a broader base of information to assist in clinical decisions. On the other hand, many of its recommendations are made outside of an RCT-evidence base. Without doubt, cholesterol management in all age and gender groups with various risk factor profiles is complex. The ESC/EAS attempts to provide a rationale for management of this complexity. The AHA/ACC, on the other hand, simplifies management as much as possible; it is written specifically for the practitioner, and leaves the complexities of management to a lipid specialist. ESC/EAS delves into the complexities in more detail so that its recommendations are applicable to both practitioner and specialist.

 

KEY PRINCIPLES  

 

There are certain key principles that clinicians should remember when deciding who to treat and how aggressively to treat hypercholesterolemia.

 

  • The Sooner the Better- atherosclerosis begins early in life and progresses overtime with LDL-C levels playing a major role in the rate of development. Lowering LDL-C levels by lifestyle changes early in life will have long-term benefits. Additionally, in selected individuals initiating drug therapy sooner rather than latter will reduce ASCVD events later in life.
  • The Lower the Better- studies have clearly demonstrated that the lower the LDL-C levels the greater the decrease in ASCVD events. Clinicians need to balance the benefits of more aggressively lowering LDL-C levels with the risks and costs of high dose or additional drug therapy. It should be recognized that statins and ezetimibe are generic drugs and very inexpensive. In contrast, PCSK9 inhibitors and bempedoic acid are expensive. In many patients using high-intensity statin therapy in combination with ezetimibe can lead to marked reductions in LDL-C levels with minimal risk and at low cost.
  • The Higher the LDL-C the Greater the Benefit- if the baseline LDL-C is high the magnitude of the reduction in LDL-C will be greater leading to a larger decrease in ASCVD events. Clinicians should be more aggressive in patients with high LDL-C levels.
  • The Greater the Risk of ASCVD the Greater the Absolute Reduction in ASCVD- clinicians should identify patients at higher risk for ASCVD and more aggressively treat these patients.

 

Following these general principles will help clinicians make informed decisions in deciding on their approach to lowering LDL-C levels and will facilitate discussions with patients on the benefits and risks of treatment. For an in-depth discussion of these key principles see the following references (79,80).    

 

SUMMARY

 

Advances in the drug therapy of elevated cholesterol levels offer great potential for reducing both new-onset ASCVD and recurrent ASCVD events in those with established disease. This benefit can be enhanced by judicious use of lifestyle intervention. But among drugs, statins are first-line therapy. They are generally safe and inexpensive. They have been shown to reduce ASCVD events in both secondary and primary prevention. Ezetimibe has about half the LDL-lowering efficacy of statins; it too is generally safe, and is a generic relatively inexpensive drug. Ezetimibe can be used as an add-on drug to moderate intensity statins, especially for those who do not tolerate a high-intensity statin. PCSK9 inhibitors are powerful LDL-lowering drugs, and they appear to be largely safe. The major drawback is cost. If the cost of these inhibitors can be reduced, they too have the potential for wide usage, especially in patients who are “statin intolerant”. The major challenge for use of cholesterol-lowering drugs is the problem of long-term non-adherence. Improving adherence will require fundamental changes in the current healthcare system in which patient monitoring and follow-up is often not a high priority.

 

REFERENCES

 

  1. Grundy SM, Stone NJ, Bailey AL, Beam C, Birtcher KK, Blumenthal RS, Braun LT, de Ferranti S, Faiella-Tommasino J, Forman DE, Goldberg R, Heidenreich PA, Hlatky MA, Jones DW, Lloyd-Jones D, Lopez-Pajares N, Ndumele CE, Orringer CE, Peralta CA, Saseen JJ, Smith SC, Sperling L, Virani SS, Yeboah J. 2018 AHA/ACC/AACVPR/AAPA/ABC/ACPM/ADA/AGS/APhA/ASPC/NLA/PCNA Guideline on the Management of Blood Cholesterol: Executive Summary: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. Circulation 2019; 139:e1082-e1143
  2. Aragam KG, Natarajan P. Polygenic Scores to Assess Atherosclerotic Cardiovascular Disease Risk: Clinical Perspectives and Basic Implications. Circ Res 2020; 126:1159-1177
  3. Goldstein JL, Brown MS. A century of cholesterol and coronaries: from plaques to genes to statins. Cell 2015; 161:161-172
  4. Third Report of the National Cholesterol Education Program (NCEP) Expert Panel on Detection, Evaluation, and Treatment of High Blood Cholesterol in Adults (Adult Treatment Panel III) Final Report. Circulation 2002; 106:3143-3421
  5. Law MR, Wald NJ, Thompson SG. By how much and how quickly does reduction in serum cholesterol concentration lower risk of ischaemic heart disease? BMJ 1994; 308:367-372
  6. Cholesterol Treatment Trialists Collaborators, Mihaylova B, Emberson J, Blackwell L, Keech A, Simes J, Barnes EH, Voysey M, Gray A, Collins R, Baigent C. The effects of lowering LDL cholesterol with statin therapy in people at low risk of vascular disease: meta-analysis of individual data from 27 randomised trials. Lancet 2012; 380:581-590
  7. Rossouw JE, Lewis B, Rifkind BM. The Value of Lowering Cholesterol after Myocardial Infarction. New England Journal of Medicine 1990; 323:1112-1119
  8. Endo A. The discovery and development of HMG-CoA reductase inhibitors. Journal of Lipid Research 1992; 33:1569-1582
  9. Brown MS, Goldstein JL. A Receptor-Mediated Pathway for Cholesterol Homeostasis. Science 1986; 232:34-47
  10. Feingold KR. Cholesterol Lowering Drugs. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dhatariya K, Dungan K, Hershman JM, Hofland J, Kalra S, Kaltsas G, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, Morley JE, New M, Purnell J, Sahay R, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA)2021.
  11. Cholesterol Treatment Trialists Collaborators, Fulcher J, O'Connell R, Voysey M, Emberson J, Blackwell L, Mihaylova B, Simes J, Collins R, Kirby A, Colhoun H, Braunwald E, La Rosa J, Pedersen TR, Tonkin A, Davis B, Sleight P, Franzosi MG, Baigent C, Keech A. Efficacy and safety of LDL-lowering therapy among men and women: meta-analysis of individual data from 174,000 participants in 27 randomised trials. Lancet 2015; 385:1397-1405
  12. Cholesterol Treatment Trialists’ Collaborators, Baigent C, Blackwell L, Emberson J, Holland LE, Reith C, Bhala N, Peto R, Barnes EH, Keech A, Simes J, Collins R. Efficacy and safety of more intensive lowering of LDL cholesterol: a meta-analysis of data from 170,000 participants in 26 randomised trials. Lancet 2010; 376:1670-1681
  13. Cholesterol Treatment Trialists Collaborators. Efficacy and safety of statin therapy in older people: a meta-analysis of individual participant data from 28 randomised controlled trials. Lancet 2019; 393:407-415
  14. Cholesterol Treatment Trialists Collaborators, Kearney PM, Blackwell L, Collins R, Keech A, Simes J, Peto R, Armitage J, Baigent C. Efficacy of cholesterol-lowering therapy in 18,686 people with diabetes in 14 randomised trials of statins: a meta-analysis. Lancet 2008; 371:117-125
  15. Silverman MG, Ference BA, Im K, Wiviott SD, Giugliano RP, Grundy SM, Braunwald E, Sabatine MS. Association Between Lowering LDL-C and Cardiovascular Risk Reduction Among Different Therapeutic Interventions. JAMA 2016; 316:1289-1297
  16. The Lipid Research Clinics Coronary Primary Prevention Trial Results. JAMA 1984; 251:351-364
  17. Lakoski SG, Xu F, Vega GL, Grundy SM, Chandalia M, Lam C, Lowe RS, Stepanavage ME, Musliner TA, Cohen JC, Hobbs HH. Indices of cholesterol metabolism and relative responsiveness to ezetimibe and simvastatin. J Clin Endocrinol Metab 2010; 95:800-809
  18. Cannon CP, Blazing MA, Giugliano RP, McCagg A, White JA, Theroux P, Darius H, Lewis BS, Ophuis TO, Jukema JW, De Ferrari GM, Ruzyllo W, De Lucca P, Im K, Bohula EA, Reist C, Wiviott SD, Tershakovec AM, Musliner TA, Braunwald E, Califf RM. Ezetimibe Added to Statin Therapy after Acute Coronary Syndromes. New England Journal of Medicine 2015; 372:2387-2397
  19. Bohula EA, Morrow DA, Giugliano RP, Blazing MA, He P, Park J-G, Murphy SA, White JA, Kesaniemi YA, Pedersen TR, Brady AJ, Mitchel Y, Cannon CP, Braunwald E. Atherothrombotic Risk Stratification and Ezetimibe for Secondary Prevention. Journal of the American College of Cardiology 2017; 69:911-921
  20. Burke AC, Telford DE, Huff MW. Bempedoic acid: effects on lipoprotein metabolism and atherosclerosis. Curr Opin Lipidol 2019; 30:1-9
  21. HPS2-THRIVE Collaborative Group, Landray MJ, Haynes R, Hopewell JC, Parish S, Aung T, Tomson J, Wallendszus K, Craig M, Jiang L, Collins R, Armitage J. Effects of Extended-Release Niacin with Laropiprant in High-Risk Patients. New England Journal of Medicine 2014; 371:203-212
  22. Lee M, Saver JL, Towfighi A, Chow J, Ovbiagele B. Efficacy of fibrates for cardiovascular risk reduction in persons with atherogenic dyslipidemia: A meta-analysis. Atherosclerosis 2011; 217:492-498
  23. Guo J, Meng F, Ma N, Li C, Ding Z, Wang H, Hou R, Qin Y. Meta-Analysis of Safety of the Coadministration of Statin With Fenofibrate in Patients With Combined Hyperlipidemia. The American Journal of Cardiology 2012; 110:1296-1301
  24. Feingold KR. Triglyceride Lowering Drugs. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dhatariya K, Dungan K, Hershman JM, Hofland J, Kalra S, Kaltsas G, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, Morley JE, New M, Purnell J, Sahay R, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA)2021.
  25. Bhatt DL, Steg PG, Miller M, Brinton EA, Jacobson TA, Ketchum SB, Doyle RT, Juliano RA, Jiao L, Granowitz C, Tardif J-C, Ballantyne CM. Cardiovascular Risk Reduction with Icosapent Ethyl for Hypertriglyceridemia. New England Journal of Medicine 2019; 380:11-22
  26. Nicholls SJ, Lincoff AM, Garcia M, Bash D, Ballantyne CM, Barter PJ, Davidson MH, Kastelein JJP, Koenig W, McGuire DK, Mozaffarian D, Ridker PM, Ray KK, Katona BG, Himmelmann A, Loss LE, Rensfeldt M, Lundstrom T, Agrawal R, Menon V, Wolski K, Nissen SE. Effect of High-Dose Omega-3 Fatty Acids vs Corn Oil on Major Adverse Cardiovascular Events in Patients at High Cardiovascular Risk: The STRENGTH Randomized Clinical Trial. JAMA 2020; 324:2268-2280
  27. Cuchel M, Bloedon LT, Szapary PO, Kolansky DM, Wolfe ML, Sarkis A, Millar JS, Ikewaki K, Siegelman ES, Gregg RE, Rader DJ. Inhibition of Microsomal Triglyceride Transfer Protein in Familial Hypercholesterolemia. New England Journal of Medicine 2007; 356:148-156
  28. Stein EA, Dufour R, Gagne C, Gaudet D, East C, Donovan JM, Chin W, Tribble DL, McGowan M. Apolipoprotein B Synthesis Inhibition With Mipomersen in Heterozygous Familial Hypercholesterolemia. Circulation 2012; 126:2283-2292
  29. Raal FJ, Rosenson RS, Reeskamp LF, Hovingh GK, Kastelein JJP, Rubba P, Ali S, Banerjee P, Chan KC, Gipe DA, Khilla N, Pordy R, Weinreich DM, Yancopoulos GD, Zhang Y, Gaudet D, Elipse HoFH Investigators. Evinacumab for Homozygous Familial Hypercholesterolemia. N Engl J Med 2020; 383:711-720
  30. Adam RC, Mintah IJ, Alexa-Braun CA, Shihanian LM, Lee JS, Banerjee P, Hamon SC, Kim HI, Cohen JC, Hobbs HH, Van Hout C, Gromada J, Murphy AJ, Yancopoulos GD, Sleeman MW, Gusarova V. Angiopoietin-like protein 3 governs LDL-cholesterol levels through endothelial lipase-dependent VLDL clearance. J Lipid Res2020; 61:1271-1286
  31. Schwartz GG, Olsson AG, Abt M, Ballantyne CM, Barter PJ, Brumm J, Chaitman BR, Holme IM, Kallend D, Leiter LA, Leitersdorf E, McMurray JJV, Mundl H, Nicholls SJ, Shah PK, Tardif J-C, Wright RS. Effects of Dalcetrapib in Patients with a Recent Acute Coronary Syndrome. New England Journal of Medicine 2012; 367:2089-2099
  32. HPS3/TIMI55–REVEAL Collaborative Group, Bowman L, Hopewell JC, Chen F, Wallendszus K, Stevens W, Collins R, Wiviott SD, Cannon CP, Braunwald E, Sammons E, Landray MJ. Effects of Anacetrapib in Patients with Atherosclerotic Vascular Disease. New England Journal of Medicine 2017; 377:1217-1227
  33. Lo Surdo P, Bottomley MJ, Calzetta A, Settembre EC, Cirillo A, Pandit S, Ni YG, Hubbard B, Sitlani A, Carfí A. Mechanistic implications for LDL receptor degradation from the PCSK9/LDLR structure at neutral pH. EMBO Rep 2011; 12:1300-1305
  34. Sabatine MS, Giugliano RP, Keech AC, Honarpour N, Wiviott SD, Murphy SA, Kuder JF, Wang H, Liu T, Wasserman SM, Sever PS, Pedersen TR. Evolocumab and Clinical Outcomes in Patients with Cardiovascular Disease. New England Journal of Medicine 2017; 376:1713-1722
  35. Schwartz GG, Steg PG, Szarek M, Bhatt DL, Bittner VA, Diaz R, Edelberg JM, Goodman SG, Hanotin C, Harrington RA, Jukema JW, Lecorps G, Mahaffey KW, Moryusef A, Pordy R, Quintero K, Roe MT, Sasiela WJ, Tamby J-F, Tricoci P, White HD, Zeiher AM. Alirocumab and Cardiovascular Outcomes after Acute Coronary Syndrome. New England Journal of Medicine 2018; 379:2097-2107
  36. Cleeman JI, Lenfant C. New guidelines for the treatment of high blood cholesterol in adults from the National Cholesterol Education Program. From controversy to consensus. Circulation 1987; 76:960-962
  37. Report of the National Cholesterol Education Program Expert Panel on Detection, Evaluation, and Treatment of High Blood Cholesterol in Adults. The Expert Panel. Archives of Internal Medicine 1988; 148:36-69
  38. National Cholesterol Education Program. Second Report of the Expert Panel on Detection, Evaluation, and Treatment of High Blood Cholesterol in Adults (Adult Treatment Panel II). Circulation 1994; 89:1333-1445
  39. Grundy SM, Cleeman JI, Bairey Merz CN, Brewer HB, Clark LT, Hunninghake DB, Pasternak RC, Smith SC, Stone NJ. Implications of Recent Clinical Trials for the National Cholesterol Education Program Adult Treatment Panel III Guidelines. Journal of the American College of Cardiology 2004; 44:720-732
  40. Stone NJ, Robinson JG, Lichtenstein AH, Bairey Merz CN, Blum CB, Eckel RH, Goldberg AC, Gordon D, Levy D, Lloyd-Jones DM, McBride P, Schwartz JS, Shero ST, Smith SC, Watson K, Wilson PWF. 2013 ACC/AHA Guideline on the Treatment of Blood Cholesterol to Reduce Atherosclerotic Cardiovascular Risk in Adults. Journal of the American College of Cardiology 2014; 63:2889-2934
  41. Institute of Medicine Committee on Standards for Developing Trustworthy Clinical Practice G. In: Graham R MM, Miller Wolman D, Greenfield S, Steinberg E, eds. Clinical Practice Guidelines We Can Trust. National Academies Press; 2011.
  42. Goff DC, Jr., Lloyd-Jones DM, Bennett G, Coady S, D'Agostino RB, Sr., Gibbons R, Greenland P, Lackland DT, Levy D, O'Donnell CJ, Robinson JG, Schwartz JS, Shero ST, Smith SC, Jr., Sorlie P, Stone NJ, Wilson PWF. 2013 ACC/AHA guideline on the assessment of cardiovascular risk: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines. Journal of the American College of Cardiology 2014; 63:2935-2959
  43. Muntner P, Colantonio LD, Cushman M, Goff DC, Jr., Howard G, Howard VJ, Kissela B, Levitan EB, Lloyd-Jones DM, Safford MM. Validation of the atherosclerotic cardiovascular disease Pooled Cohort risk equations. JAMA 2014; 311:1406-1415
  44. Ridker PM, Cook NR. Statins: new American guidelines for prevention of cardiovascular disease. The Lancet2013; 382:1762-1765
  45. Kavousi M, Leening MJG, Nanchen D, Greenland P, Graham IM, Steyerberg EW, Ikram MA, Stricker BH, Hofman A, Franco OH. Comparison of Application of the ACC/AHA Guidelines, Adult Treatment Panel III Guidelines, and European Society of Cardiology Guidelines for Cardiovascular Disease Prevention in a European Cohort. JAMA 2014; 311:1416-1423
  46. DeFilippis AP, Young R, Carrubba CJ, McEvoy JW, Budoff MJ, Blumenthal RS, Kronmal RA, McClelland RL, Nasir K, Blaha MJ. An analysis of calibration and discrimination among multiple cardiovascular risk scores in a modern multiethnic cohort. Ann Intern Med 2015; 162:266-275
  47. Damen JA, Pajouheshnia R, Heus P, Moons KGM, Reitsma JB, Scholten RJPM, Hooft L, Debray TPA. Performance of the Framingham risk models and pooled cohort equations for predicting 10-year risk of cardiovascular disease: a systematic review and meta-analysis. BMC Med 2019; 17:109-109
  48. Rana JS, Tabada GH, Solomon MD, Lo JC, Jaffe MG, Sung SH, Ballantyne CM, Go AS. Accuracy of the Atherosclerotic Cardiovascular Risk Equation in a Large Contemporary, Multiethnic Population. Journal of the American College of Cardiology 2016; 67:2118-2130
  49. Ridker PM, Danielson E, Fonseca FAH, Genest J, Gotto AM, Kastelein JJP, Koenig W, Libby P, Lorenzatti AJ, MacFadyen JG, Nordestgaard BG, Shepherd J, Willerson JT, Glynn RJ. Rosuvastatin to Prevent Vascular Events in Men and Women with Elevated C-Reactive Protein. New England Journal of Medicine 2008; 359:2195-2207
  50. Arnett DK, Blumenthal RS, Albert MA, Buroker AB, Goldberger ZD, Hahn EJ, Himmelfarb CD, Khera A, Lloyd-Jones D, McEvoy JW, Michos ED, Miedema MD, Muñoz D, Smith SC, Jr., Virani SS, Williams KA, Sr., Yeboah J, Ziaeian B. 2019 ACC/AHA Guideline on the Primary Prevention of Cardiovascular Disease: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. Circulation 2019; 140:e596-e646
  51. Feingold KR. The Effect of Diet on Cardiovascular Disease and Lipid and Lipoprotein Levels. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dhatariya K, Dungan K, Hershman JM, Hofland J, Kalra S, Kaltsas G, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, Morley JE, New M, Purnell J, Sahay R, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA)2021.
  52. Fonarow GC, Keech AC, Pedersen TR, Giugliano RP, Sever PS, Lindgren P, van Hout B, Villa G, Qian Y, Somaratne R, Sabatine MS. Cost-effectiveness of Evolocumab Therapy for Reducing Cardiovascular Events in Patients With Atherosclerotic Cardiovascular Disease. JAMA Cardiol 2017; 2:1069-1078
  53. Virani SS, Akeroyd JM, Smith SC, Al-Mallah M, Maddox TM, Morris PB, Petersen LA, Ballantyne CM, Grundy SM, Stone NJ. Very High-Risk ASCVD and Eligibility for Nonstatin Therapies Based on the 2018 AHA/ACC Cholesterol Guidelines. Journal of the American College of Cardiology 2019; 74:712-714
  54. Khera AV, Won H-H, Peloso GM, Lawson KS, Bartz TM, Deng X, van Leeuwen EM, Natarajan P, Emdin CA, Bick AG, Morrison AC, Brody JA, Gupta N, Nomura A, Kessler T, Duga S, Bis JC, van Duijn CM, Cupples LA, Psaty B, Rader DJ, Danesh J, Schunkert H, McPherson R, Farrall M, Watkins H, Lander E, Wilson JG, Correa A, Boerwinkle E, Merlini PA, Ardissino D, Saleheen D, Gabriel S, Kathiresan S. Diagnostic Yield and Clinical Utility of Sequencing Familial Hypercholesterolemia Genes in Patients With Severe Hypercholesterolemia. Journal of the American College of Cardiology 2016; 67:2578-2589
  55. Warden BA, Fazio S, Shapiro MD. Familial Hypercholesterolemia: Genes and Beyond. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dhatariya K, Dungan K, Hershman JM, Hofland J, Kalra S, Kaltsas G, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, Morley JE, New M, Purnell J, Sahay R, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA)2000.
  56. Randomised trial of cholesterol lowering in 4444 patients with coronary heart disease: the Scandinavian Simvastatin Survival Study (4S). The Lancet 1994; 344:1383-1389
  57. Kashef MA, Giugliano G. Legacy effect of statins: 20-year follow up of the West of Scotland Coronary Prevention Study (WOSCOPS). Glob Cardiol Sci Pract 2016; 2016:e201635-e201635
  58. Feingold KR. Role of Glucose and Lipids in the Atherosclerotic Cardiovascular Disease of Patients with Diabetes. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dhatariya K, Dungan K, Hershman JM, Hofland J, Kalra S, Kaltsas G, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, Morley JE, New M, Purnell J, Sahay R, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA)2020.
  59. Lloyd-Jones DM, Braun LT, Ndumele CE, Smith SC, Sperling LS, Virani SS, Blumenthal RS. Use of Risk Assessment Tools to Guide Decision-Making in the Primary Prevention of Atherosclerotic Cardiovascular Disease: A Special Report From the American Heart Association and American College of Cardiology. Circulation 2019; 139:e1162-1177
  60. Horton JD, Cohen JC, Hobbs HH. PCSK9: a convertase that coordinates LDL catabolism. J Lipid Res 2009; 50 Suppl:S172-177
  61. Ference BA, Ray KK, Catapano AL, Ference TB, Burgess S, Neff DR, Oliver-Williams C, Wood AM, Butterworth AS, Di Angelantonio E, Danesh J, Kastelein JJP, Nicholls SJ. Mendelian Randomization Study of ACLY and Cardiovascular Disease. N Engl J Med 2019; 380:1033-1042
  62. Yebyo HG, Aschmann HE, Kaufmann M, Puhan MA. Comparative effectiveness and safety of statins as a class and of specific statins for primary prevention of cardiovascular disease: A systematic review, meta-analysis, and network meta-analysis of randomized trials with 94,283 participants. American Heart Journal 2019; 210:18-28
  63. Tota-Maharaj R, Blaha MJ, Blankstein R, Silverman MG, Eng J, Shaw LJ, Blumenthal RS, Budoff MJ, Nasir K. Association of coronary artery calcium and coronary heart disease events in young and elderly participants in the multi-ethnic study of atherosclerosis: a secondary analysis of a prospective, population-based cohort. Mayo Clin Proc 2014; 89:1350-1359
  64. Stone NJ, Smith SC, Jr., Orringer CE, Rigotti NA, Navar AM, Khan SS, Jones DW, Goldberg R, Mora S, Blaha M, Pencina MJ, Grundy SM. Managing Atherosclerotic Cardiovascular Risk in Young Adults: JACC State-of-the-Art Review. J Am Coll Cardiol 2022; 79:819-836
  65. Streja E, Streja DA. Management of Dyslipidemia in the Elderly. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dhatariya K, Dungan K, Hershman JM, Hofland J, Kalra S, Kaltsas G, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, Morley JE, New M, Purnell J, Sahay R, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA)2000.
  66. Banach M, Stulc T, Dent R, Toth PP. Statin non-adherence and residual cardiovascular risk: There is need for substantial improvement. International Journal of Cardiology 2016; 225:184-196
  67. Avorn J, Monette J, Lacour A, Bohn RL, Monane M, Mogun H, LeLorier J. Persistence of Use of Lipid-Lowering Medications. JAMA 1998; 279:1458
  68. Simons LA, Levis G, Simons J. Apparent discontinuation rates in patients prescribed lipid‐lowering drugs. Medical Journal of Australia 1996; 164:208-211
  69. Gamboa CM, Safford MM, Levitan EB, Mann DM, Yun H, Glasser SP, Woolley JM, Rosenson R, Farkouh M, Muntner P. Statin underuse and low prevalence of LDL-C control among U.S. adults at high risk of coronary heart disease. Am J Med Sci 2014; 348:108-114
  70. Blackburn DF, Dobson RT, Blackburn JL, Wilson TW, Stang MR, Semchuk WM. Adherence to statins, beta-blockers and angiotensin-converting enzyme inhibitors following a first cardiovascular event: a retrospective cohort study. Can J Cardiol 2005; 21:485-488
  71. Tobert JA, Newman CB. The nocebo effect in the context of statin intolerance. Journal of Clinical Lipidology2016; 10:739-747
  72. Golomb BA, Evans MA. Statin Adverse Effects: a review of the literature and evidence for a mitochondrial mechanism. American Journal of Cardiovascular Drugs 2008; 8:373-418
  73. Fitchett DH, Hegele RA, Verma S. Statin Intolerance. Circulation 2015; 131:e389-391
  74. Pasternak RC, Smith SC, Jr., Bairey-Merz CN, Grundy SM, Cleeman JI, Lenfant C, American College of C, American Heart A, National Heart L, Blood I. ACC/AHA/NHLBI Clinical Advisory on the Use and Safety of Statins. Circulation 2002; 106:1024-1028
  75. Nissen SE, Stroes E, Dent-Acosta RE, Rosenson RS, Lehman SJ, Sattar N, Preiss D, Bruckert E, Ceška R, Lepor N, Ballantyne CM, Gouni-Berthold I, Elliott M, Brennan DM, Wasserman SM, Somaratne R, Scott R, Stein EA. Efficacy and Tolerability of Evolocumab vs Ezetimibe in Patients With Muscle-Related Statin Intolerance. JAMA 2016; 315:1580-1590
  76. Catapano AL, Graham I, De Backer G, Wiklund O, Chapman MJ, Drexel H, Hoes AW, Jennings CS, Landmesser U, Pedersen TR, Reiner Ž, Riccardi G, Taskinen M-R, Tokgozoglu L, Verschuren WMM, Vlachopoulos C, Wood DA, Zamorano JL. 2016 ESC/EAS Guidelines for the Management of Dyslipidaemias. European Heart Journal 2016; 37:2999-3058
  77. Mach F, Baigent C, Catapano AL, Koskinas KC, Casula M, Badimon L, Chapman MJ, De Backer GG, Delgado V, Ference BA, Graham IM, Halliday A, Landmesser U, Mihaylova B, Pedersen TR, Riccardi G, Richter DJ, Sabatine MS, Taskinen MR, Tokgozoglu L, Wiklund O, Group ESCSD. 2019 ESC/EAS Guidelines for the management of dyslipidaemias: lipid modification to reduce cardiovascular risk. Eur Heart J 2020; 41:111-188
  78. Lee JC, Zdrojewski T, Pencina MJ, Wyszomirski A, Lachacz M, Opolski G, Bandosz P, Rutkowski M, Gaciong Z, Wyrzykowski B, Navar AM. Population Effect of Differences in Cholesterol Guidelines in Eastern Europe and the United States. JAMA Cardiol 2016; 1:700-707
  79. Feingold KR, Chait A. Approach to patients with elevated low-density lipoprotein cholesterol levels. Best Pract Res Clin Endocrinol Metab 2022:101658
  80. Feingold KR. Maximizing the benefits of cholesterol-lowering drugs. Curr Opin Lipidol 2019; 30:388-394

Adult Growth Hormone Deficiency Clinical Management

ABSTRACT

 

The clinical syndrome of GH deficiency (GHD) is characterized by non-specific features including variable presence of decreased mood and general well-being, reduced bone remodeling activity, change in body fat distribution with increased central adiposity, hyperlipidemia, and increased predisposition to atherogenesis. The goal of GH replacement therapy in adults with GH deficiency is to correct the wide spectrum of associated clinical alterations. The estimated prevalence of GHD is approximately 2-3:10,000 population. GHD is caused by structural pituitary disease or cranial irradiation, and usually occurs in the context of additional features of hypopituitarism. Pituitary adenomas are the most important cause of adult-onset GHD followed by craniopharyngiomas, which combined account for 57% of cases. Less common causes are irradiation, head injury, vascular, infiltrative, infectious. and autoimmune disease. Diagnosing patients with GHD should first of all consider who should be tested for GHD, which includes patients at relevant risk with an intention to treat, and second which stimulation test to be used including the proper diagnostic cut-off concentration of GH. The diagnosis of GHD in adults is then usually straightforward. Dosage of h-GH replacement is dependent on age, and gender with adolescents and women usually requiring an increased dosage. The dose titration is monitored by IGF-I concentrations and apart from that a number of organ end points, which may act as ‘biomarkers’ of the treatment effects. This chapter provides an update on GHD including diagnostic pitfalls, and treatment effect, safety, and monitoring.

 

INTRODUCTION

 

The term "midget" as description of a proportionate dwarf was first used in 1816, but it was not until 1912, that Harvey Cushing in “The Pituitary Gland” proposed the existence of a "hormone of growth" promoting skeletal growth in children (1). Growth hormone (GH) or somatrophin was first extracted from cadaveric pituitaries in the late 1950es, and other more metabolic actions of this hormone in humans were described soon after by Maurice Raben (2). The purified hormone was initially only used for the treatment of short stature in hypopituitary children, although Raben already in 1962 described general health improvement after injection of GH in a hypopituitary adult (2). Further, increasing knowledge of GH effects in adults was brought forward by the introduction in 1962 by Utiger et al of a radioimmunoassay for measuring GH in human serum (3).

 

The clinical syndrome of GH deficiency in adults is a consequence of decreased secretion of GH from the anterior pituitary. Until thirty years ago it was widely held that GH deficiency had little pathophysiological consequence despite the previously mentioned earlier anecdotal reports suggesting presence of GH-remediable symptoms of fatigue and decreased general well-being which responded to GH replacement (2). In retrospect, these observations of more than fifty years ago described quite precisely the later well-known classical features of the GH deficiency syndrome. However, the imitated supplies of cadaveric GH and the focus on pediatric usage resulted in delayed further elucidation of the adult GH deficiency state. The measurement of serum GH and the production of recombinant human GH (rhGH) in 1981 made studies of GH concentrations in adults as well as effects on the human body of GH deficiency possible, and clinical studies on replacement with rhGH could begin.

 

The initial pivotal trials of GH replacement therapy in adult hypopituitary patients were published in 1989 (4, 5). Numerous subsequent studies have provided compelling evidence for the existence of a syndrome of adult GH deficiency (6-8). This is characterized by the variable presence of decreased exercise tolerance, decreased mood and general well-being, reduced bone remodeling activity, change in body fat distribution with increased central adiposity, hyperlipidemia, and increased predisposition to atherogenesis. However, it is important to recognize that adult-onset GH deficiency is due to structural pituitary or hypothalamic disease or cranial irradiation for other pathologies and, therefore, usually occurs in the context of additional features of hypopituitarism (9, 10). For this reason, the clinical features attributable to GH deficiency may be compounded by, or directly related to, other pituitary deficiencies. Nonetheless, the fact that GH replacement therapy may favorably alter these clinical features provides considerable surrogate evidence for GH deficiency as a causal factor.

 

Adult GH deficiency is thus a well-recognized clinical entity. It causes abnormalities in substrate metabolism, bone remodeling, body composition, as well as physical, and psychosocial function. Since the mid 80-ies an improvement has been recognized with GH replacement, and this has gradually been incorporated in clinical routine based on the few short-term initial randomized clinical trials, which led to the first international consensus guidelines from Growth Hormone Research Society in 1997 (11) and updated in 2007 (12).

Fig 1. Growth hormone secretion varies throughout life. From: Ho KY et al. (13)

Less well recognized is the fact that the early clinical trials were based on selected groups of patients with very severe hypopituitarism and therefore had a high a priory likelihood of severe GH deficiency, there were few study participants, short-term treatment, and supraphysiological GH doses were calculated based on the experience from childhood GH deficiency. Despite knowledge of the very high influence of age on the secretion of GH (Fig 1) and subsequently on Insulin-like-Growth Factor-I (IGF-I) the initial adult doses in the studies were nevertheless chosen too high (4, 5). Most of the current recommendations and guidelines over the years have thus been based on subsequent retrospective single center experience or data from large surveillance databases run by the pharmaceutical industry as the best surrogates for efficacy and safety of GH therapy of adult GH deficiency (14). The Hypopituitary Control and Complications Study (HypoCCS) compiled data from 5,893 patients on Humatrope® and reported that significant shifts in diagnostic patterns have occurred over 10 years after approval of the adult GH deficiency indication, with a trend to less severe forms of GH deficiency (15). This was further documented in a recent publication from KIMS (Pfizer’s International Metabolic Database) where data compiled over 20 years were retrieved for a total of 6,069 patients with adult-onset GH deficiency and treated with Genotropin® from six countries (Belgium, Germany, Netherlands, Spain, Sweden, and UK). The degree of confirmed GH deficiency became less pronounced and more patients with co-morbidities and diabetes were considered for GH replacement therapy, possibly reflecting increased knowledge and confidence in GH therapy gained with time. Thus, the effects of 1 year of GH replacement were similar over the entry year periods despite changes in the patients’ baseline characteristics (16).

 

Also, less well recognized is the fact that new possible indications for testing and treatment of GH deficiency have emerged and these very likely have a lower a priori likelihood of the disease than the severely hypopituitary patients initially investigated thus challenging the diagnostic criteria laid down for severe GH deficiency. Clinicians are therefore now dealing with other patient populations as, e.g. traumatic brain injury, where neither testing nor treatment efficacy have been scrutinized sufficiently (17) and where current guidelines therefore may fail to apply correctly.

 

This chapter is an update of our chapter from 2017 which was in turn based on the previous chapter on the topic written by John Monson, Antonia Brooke and Scott Akker and the update will describe the diagnostic procedures, as well as the clinical consequences and management of adult patient with GH deficiency. The basic physiology of GH and the pathophysiology of GH deficiency in adults have been dealt with in other Endotext chapters (www.endotext.org).

 

PREVALENCE, INCIDENCE, AND ETIOLOGY OF GH DEFICIENCY IN ADULTS

 

The true prevalence and incidence rate of adult-onset GH deficiency is difficult to estimate with certainty. A reasonable estimate of the prevalence may be obtained from prevalence data for pituitary macroadenomas, which approximate to 1-2:10,000 population (20-22). Addition of cases of childhood-onset GH deficiency persisting into adult life gives an overall prevalence of 2-3:10,000 population. Incidence rates have been assessed in a Danish nationwide study based on registries (23), including 1,823 patients who were divided in males and females with childhood and adult onset GH deficiency, respectively. The average incidence rates were for childhood onset males, 2.58 (95% confidence interval (CI), 2.30-2.88), childhood onset females, 1.70 (95% CI, 1.48-1.96), adult-onset males, 1.90 (95% CI, 1.77-2.04), and adult-onset females, 1.42 (95% CI, 1.31-1.54) all per 100,000, which are slightly higher than previously reported (24, 25). The incidence rate in the Danish study was significantly higher in males compared to females in the childhood onset GH deficiency group and in the adult-onset GH deficiency group in the age ranges of 45-64 and 65+years, while there was no significant gender difference in the 18-44 years age group. The etiology spectrum of GH deficiency is summarized in figure 2.

Figure 2. Congenital and acquired causes of growth hormone deficiency

Pituitary adenomas are statistically the most important cause of adult-onset GH deficiency followed by craniopharyngiomas, which combined account for 57% of cases in the study based on data from KIMS, a multinational, pharmacoepidemiological surveillance database for adult hypopituitary patients receiving GH replacement with Genotropin® (Table 1) (26). Over a decade, there was a decrease in patients enrolled in the surveillance databases with diagnoses of pituitary adenoma (50.2 to 38.6%; P<0.001); craniopharyngioma (13.3 to 8.4%; P=0.005) and pituitary hemorrhage (5.8 to 2.8%; P=0.001); increases in idiopathic GH deficiency (13.9 to 19.3%; P<0.001) and undefined/unknown diagnosis (1.3 to 8.6%; P<0.001) in HypoCCS(15).

 

Table 1. Etiology in Patients with GH Deficiency (from the KIMS database)

Etiology category

Category components

   n

Patient-years

Pituitary adenoma

Non-functioning adenoma

5261

28 065

 

Prolactinoma

 

 

 

Gonadotropinoma

 

 

 

Thyrotropinoma

 

 

Cushing's disease

Cushing's disease

859

4814

Acromegaly

Acromegaly

239

1396

Pituitary atrophy

Congenital

2496

10 535

 

Idiopathic

 

 

 

Empty sella

 

 

Craniopharyngioma

Craniopharyngioma

1562

8392

Benign tumor/lesion

Hamartoma

462

2114

 

Cyst

 

 

 

Meningioma

 

 

 

Schwannoma

 

 

Aggressive tumor  (+hematological neoplasm)

Germ cell tumor

1135

5552

 

Glioma

 

 

 

Chordoma

 

 

 

Sarcoma

 

 

 

Astrocytoma

 

 

 

Ependymoma

 

 

 

Medulloblastoma

 

 

 

Leukemia

 

 

 

Lymphoma

 

 

Miscellaneous etiology

Traumatic brain injury

1969

8189

 

Subarachnoid hemorrhage

 

 

 

Aneurysm

 

 

 

Sheehan's syndrome

 

 

 

Hydrocephalus

 

 

 

Granulomatosis

 

 

 

Histiocytosis

 

 

 

Hypophysitis

 

 

 

Hemochromatosis

 

 

 

Missing etiology

 

 

From: Gaillard et al (26)

 

Irradiation includes both pituitary tumors but also other forms of childhood and adult cranial irradiation. Less common causes of adult hypopituitarism are head injury (27), postpartum ischemic necrosis [Sheehan’s syndrome], pituitary apoplexy, infiltrative diseases, and autoimmune lymphocytic hypophysitis. Traumatic brain injury and subarachnoid hemorrhage are increasingly recognized as a cause of hypopituitarism, in particular GH deficiency, which the recently updated guidelines from American Association of Clinical Endocrinologists and American College of Endocrinology indicated to be one of the most common causes of adult GH deficiency seen in clinical practice (18). Several anti-cancer drugs modulating the immune system and used for antineoplastic purposes may result in hypophysitis with hypofunction including GH deficiency (29, 30), and the list of drugs influencing GH secretion may be increasing, perhaps also including treatment of patient groups with autoimmune diseases.

 

Most cases of adult GH deficiency arise in adulthood, but a proportion of them are suffering from childhood onset GH deficiency thus also including congenital causes. The proportion in each clinical center will depend on referral practice. In the Danish nationwide study 27% of GH deficiency patients were of childhood origin (23). The congenital cases (figure 2) are due to structural lesions such as Rathke’s pouch cysts, pituitary hypoplasia, and midline defects, or to functionally deficient GH biosynthesis and release such as pituitary-specific transcription factors (PROP1, POU1F1, HESX1, LHX3, LHX4), and LEPR or IGSF1. Thus, childhood-onset GH deficiency due to proven genetic defects in GH synthesis is never reversible and therefore does not require retesting prior to treatment on adult indication. The reversibility of isolated idiopathic GH deficiency of childhood is on the other hand well established with normal GH responses on dynamic testing in various series being described in between 30 and 70% of subjects with confirmed GH deficiencies in childhood at completion of linear growth (31-33). Therefore, childhood-onset isolated GH deficiency should always be challenged by rigorous re-evaluation of causes and retesting at completion of final height.

Fig 3. Mean (±SEM) serum GH response to insulin hypoglycemia in normal subjects (▲) and obese subjects before (●) and after (○) weight loss. From: Rasmussen et al (34).

Isolated idiopathic GH deficiency is not accepted as de novo deficiency in adults at this point in time. This is particularly important in the assessment of non-specific symptoms in ageing or overweight persons without additional evidence of pituitary disease; body mass index of >32 kg/m2 is associated with reduced GH reserve on dynamic testing in approximately 30% of patients but this is reversible with weight loss (Fig 3) (34-37). However, combined deficiency of GH and other anterior pituitary hormones, in the absence of structural disease, may be a feature of an evolving endocrinopathy due to deficiencies of the transcription factors PIT-1 or Prop-1. These cases, and possibly also others, may account for some of the patients with isolated GH deficiency developing into multiple pituitary hormone insufficiencies in 6-65% of cases over time (Fig 4) (38-40).

Fig 4. Number of patients with GH deficiency and at least one additional pituitary deficit at baseline who developed central hypothyroidism, hypoadrenalism, hypogonadism or ADH deficiency in relation to years from baseline. From: Klose et al (38)

The sequence of loss of pituitary functions is displayed in Fig 5, demonstrating that GH deficiency usually occurs early in the progression of pituitary insufficiency, at least in pituitary adenomas (10). Normalization of GH but also other deficiencies are sometimes observed after selective adenectomy (41-43).

Fig 5. Sequence of pituitary hormone loss in relation to increasing mass effect from a pituitary tumor. From: Feldt-Rasmussen & Klose (10)

CLINICAL FEATURES OF GH DEFICIENCY IN ADULTS

 

Adult GH deficiency is associated with an extensive array of non-specific symptoms and physical signs, which are nevertheless recognized by experienced endocrinologists to justify their designation as a clinical syndrome (6-8, 44). Typical symptoms and signs are listed in Table 2.

 

Table 2. Typical Symptoms and Signs of the Adult Growth Hormone Deficiency Syndrome

Body composition

·       increased body fat, particularly central adiposity

·       decreased muscle mass

·       decreased muscle function

Cardiovascular and metabolism

·       decreased sweating and poor thermoregulation

·       decreased insulin sensitivity and increased prevalence of impaired glucose tolerance

·       increased total and LDL cholesterol and Apo B. Decreased HDL cholesterol

·       accelerated atherogenesis

·       a variable decrease in cardiac muscle mass

·       impaired cardiac function

·       decreased exercise capacity

·       decreased total and extracellular fluid volume

·       increased concentration of plasma fibrinogen and plasminogen activator inhibitor type I

Bones

·       decreased bone mineral density, associated with an increased risk of fracture

Quality of Life

·       depressed mood

·       reduced concentration

·       increased anxiety

·       fatigue

·       lack of energy levels

·       low self-esteem

·       increased sick days

·       social isolation

·       lack of positive well being

 

Body Composition and Heart

 

GH deficiency is characterized by substantial changes in body composition with increments in total fat, percentage fat, and particularly visceral fat mass (45-52). Methodologies employed for this purpose have included dual energy X-ray absorptiometry (DEXA) (53), bioelectrical impedance (6-8), CT scan of specific body parts, or the simple measurement of the ratio of waist to hip circumference (52-56) (Table 2) and there is complete concordance among all studies which have examined these aspects in hypopituitary adults. Importantly, although the prevalence of obesity is increased in hypopituitary adults, the increment in visceral fat is also evident in those patients who are non-obese (45). In parallel with changes in fat mass, lean body mass is reduced. The latter may explain the reductions in muscle strength (57-59) and exercise tolerance, which have been documented in adult GH deficiency. The degree to which lean body mass is reduced is difficult to determine because of the reduction in total body water which is also evident in the GH deficiency state; body composition measurements, particularly bioelectrical impedance, may overestimate changes in lean body mass as a consequence of alterations in tissue hydration. Furthermore, the reduction in extracellular water, which is compounded by reduced total body sodium in GH deficiency, may be a major factor underlying the reported reductions in exercise capacity (60-64). To this may be added the effect of reduced left ventricular mass and function which have been described in a number of studies (65-75), although some of these studies on cardiac function in GH deficiency have been less clear.

 

Glucose Metabolism

 

In contrast to GH deficiency occurring in children, adult GH deficiency is associated with relative insulin resistance (45, 76-79) and an increased prevalence of impaired glucose tolerance and diabetes mellitus (76). The adverse changes in insulin sensitivity are predictably most obvious in obese patients but are also evident in hypopituitary patients with normal body mass index in whom the inverse relationship between insulin sensitivity and central fat mass, which characterizes the 'metabolic syndrome' is clearly seen (45, 76-79). It is therefore likely that the changes in insulin sensitivity observed in adult GH deficiency are due predominantly to increases in central fat mass. Interestingly, adult subjects with lifetime congenital untreated isolated GH deficiency have reduced β-cell function, no evidence of insulin resistance, and a higher frequency of impaired glucose tolerance (79).Thus, lifetime, untreated isolated GH deficiency increases insulin sensitivity, but impairs β-cell function, and does not provide protection from diabetes (79, 80). It has been postulated that changes in body composition and particularly fat mass might be a consequence of unphysiological glucocorticoid replacement. Against this is the fact that the doses of glucocorticoid replacement used in primary adrenal failure, which are similar to those used in hypopituitarism, are not associated with abnormalities of body composition. However, local tissue exposure to either endogenous or exogenous cortisol may be different in secondary as opposed to primary adrenal failure. The GH/IGF-I axis is now recognized to be an important modulator of the activity of the enzyme 11b hydroxysteroid dehydrogenase Type 1 (11bHSD1) (81). This isoenzyme acts as a predominant reductase, particularly in liver and adipose tissue, increasing the net conversion of inactive cortisone to the active cortisol. The activity of the enzyme is decreased by GH and, as a consequence GH deficiency is associated with a shift in the equilibrium set point in favor of cortisol. It is therefore possible that the increase in central adiposity, which characterizes the GH deficiency state, could be compounded by enhanced exposure to cortisol within adipocytes; hepatic metabolism might be perturbed by a similar mechanism. These mechanisms would tend to increase serum cortisol concentrations in patients receiving hydrocortisone replacement, which is quite often supraphysiological doses, but not in patients with intact ACTH reserve in whom negative feedback would determine maintenance of stable circulating cortisol concentrations. However, GH is also a negative determinant of serum cortisol binding globulin and therefore comparisons of serum total cortisol concentrations between GH deficient and GH replete states are not valid.

 

Atherosclerosis Risk Factors- Lipids and Hypertension

 

Adult GH deficiency is associated with an increase in total cholesterol, LDL-cholesterol and apolipoprotein B (4-8, 82, 83). A modest decrement in HDL-cholesterol has also been described in some studies. These changes are evident in both sexes and are quantitatively greater in women. Despite GH deficiency related sodium and water depletion, an increased prevalence of hypertension in adult hypopituitarism has been documented and may be related to a reduced activity of nitric oxide synthase, and consequent increased peripheral vascular resistance, as a result of GH deficiency. The changes in lipoprotein metabolism, body composition, insulin sensitivity, and peripheral vascular resistance indicated above would predict increased atherogenesis in the GH deficiency state. Indeed, several studies have reported an increase in ultrasonographically determined intima-media thickness and plaque formation in large arteries of patients with adult-onset GH deficiency as well as in adults with childhood-onset disease (84, 85).

 

Bone Mineralization

 

Decreased bone mineral density is a recognized phenomenon in adult hypopituitary patients (53, 82, 86-92) and is associated with an increased fracture risk (Fig 6) (92-96). Measurements of markers of bone formation and bone resorption are consistent with a low bone turnover state in GH deficiency. Deficits in bone mineral content and density are more striking in adults with childhood-onset GH deficiency and this is likely to be a consequence of failure to achieve genetic potential peak bone mass either because of inadequate GH replacement in childhood or its early cessation at the time of slowing of linear growth (86). Failure to achieve peak bone mass has important implications for the future development of osteoporosis and fracture risk. In the study by Lange et al (96), it was found that patients with idiopathic childhood onset GH deficiency, who at retest in adulthood did not have GH deficiency according to adult criteria, had reduced serum IGF-I and BMD/BMC compared to controls. This observation was also made in the patients who did have persistent GH deficiency in adulthood. The findings may reflect the fact that the present diagnostic criteria for adult GH deficiency (i.e., response to the ITT) do not reflect the clinical consequences of disordered GH-IGF axis in childhood onset GH deficiency young adults who were treated with GH in childhood. Alternatively, despite seemingly adequate GH treatment in childhood an optimal peak bone mass in adolescence may never have been reached in either of the groups. Noteworthy, IGF-I levels correlated with clinical signs of the adult GH deficiency syndrome. The situation in hypopituitarism is further complicated by the frequent accompaniment of gonadal steroid deficiency, often of unknown duration, which has a documented effect on the BMD (97). In addition, glucocorticoid replacement for primary adrenal failure is associated with modest reductions in bone mineral density, but over-replacement in hypopituitary patients does accelerate bone loss (98). Thus, glucocorticoid over-replacement may increase the prevalence of vertebral fractures in patients with untreated GH deficiency. However, treatment of GH deficiency seems to protect the skeleton from the deleterious effects of glucocorticoid overtreatment in hypopituitary patients. Along the same line, data suggest that the characteristics of patients in the various diagnostic groups of hypopituitarism depend on the primary disease which resulted in GH deficiency, and that the clinical expression of GH deficiency does not differ between the groups. Patients with previous hypercortisolism showed more long-term effects of their disease, such as diabetes mellitus, hypertension, and fractures (99), and patients with former Cushing’s disease have more fractures (100). Furthermore, Lange et al reported abnormal bone collagen morphology and decreased bone strength in rats with isolated GH deficiency (101), which might provide a co-explanation for the increased fracture rate in GH deficiency. Whether similar conditions are present in patients with GH deficiency needs further study, but results from a human study of muscle and tendon biopsies from patients with GH deficiency or acromegaly indicated a collagen-stimulating role of local IGF-I in human connective tissue and add to the understanding of musculoskeletal pathology in patients with either high or low GH/IGF-I axis activity (102).

Fig. 6. Comparisons of the prevalence of (A) all fractures in the EVOS (European Vertebral Osteoporosis Study) participants and in KIMS participants over the age of 60 years and (B) all fractures in naïve and non-naïve KIMS patients and of fractures of the radius in naïve and non-naïve and in patients with adult onset (AO) and childhood onset (CO) disease. From: Wüster et al (92)

Nonetheless, available evidence indicates that qualitatively similar changes in bone mineral density are found in adult-onset isolated GH deficiency as in panhypopituitarism, therefore supporting a role for GH deficiency in the pathogenesis. Furthermore, these abnormalities in bone metabolism and bone density are favorably influenced by GH replacement (see later).

 

Quality of Life

 

Decreased psychological well-being and quality of life (QoL) are recognized as particularly important for patients with GH deficiency and from the patients' perspective they have arguably become the major indication for GH replacement therapy. In some countries such as UK, decreased QoL is a needed symptom of a certain quantitative level as measured by validated GH deficiency questionnaires before even considering testing and treatment for GH deficiency according to National Institute for Health and Care Excellence or NICE (103). QoL is also related to a number of other features of GH deficiency. Thus Chikani et al. found subnormal anaerobic capacity, which independently predicted stair-climbing capacity and QoL in adults with GH deficiency. The authors concluded that GH regulates anaerobic capacity, which determines QoL and selective aspects of physical function (Fig 7) (104).

Fig 7. Relationship between stair-climb performance and anaerobic power (A) and VO2max (B), and between daily step counts quantified by pedometry and anaerobic power (C) and VO2max (D) in the combined groups of 13 adults with GHD (●) and matched normal subjects (○). LBM, lean body mass; VO2max, maximal oxygen consumption. From: Chikani et al (104)

QoL issues have been examined using various generic measures including the Nottingham Health Profile and the Psychological General Well Being Schedule (105-108). These instruments determine various aspects of health-related and needs-based quality of life and the most prevalent findings from various studies have been deficits in the domains of mood, anxiety, and social interaction. In one of the studies the Nottingham Health profile was adapted to a 9-year follow-up study of adults with untreated GH deficiency. During this 9-year study, small but significant declines in health were observed in GH-deficient adults who remained untreated. By contrast, the patients who received GH continuously experienced improvements in energy levels while all other areas of QoL were maintained. The beneficial effects of GH on QoL are therefore maintained with long-term GH replacement and obviate the reduction in QoL seen over time in untreated GH-deficient adults (106). Although these findings are readily apparent in many patients with adult-onset GH deficiency, it has proven more difficult to discern similar phenomena in patients with childhood-onset disease. This may be due to at least two factors. Firstly, standard generic quality of life instruments may be insensitive in the investigation of young people and secondly, there may be a major element of psychological adaptation or decreased expectation when the condition has commenced early in life. In an attempt to improve the reproducibility of studies of QoL in adults with GH deficiency, questionnaires have been developed which focus on those symptoms, which are most frequently documented in hypopituitary adults during extended open interviews.

 

One such instrument, which is now widely used for the baseline and longitudinal follow-up of patients, is the Quality-of-Life Assessment in Growth Hormone Deficient Adults (QoL-AGHDA) (107-111), which is also the one required by NICE. This is a needs-based instrument consisting of 25 questions with a yes/no answer format and the final score is obtained by summating all the positive responses; a higher score, to a maximum of 25, denotes poorer quality of life. The questionnaire has been shown to be reproducible in a variety of languages and satisfies Rasch analysis criteria for unidimensionality, construct validity, and hierarchical ordering of items (108).  In the long-term KIMS study surprisingly, QoL-AGHDA scores increased, indicating worsening of QoL across the entry year periods(16). This possibly reflected a patient selection bias, due to the change in the underlying etiology of GH deficiency: for example, the increase in the proportion of patients with traumatic brain injury or other less defined diagnoses may affect QoL. Alternatively, patients with poor QoL were more likely to receive GH treatment (15).     

 

Mortality

 

Over the past decades it has been increasingly recognized that hypopituitarism is associated with premature mortality. Studies in Sweden and the UK have demonstrated a two-to-three-fold increase in standardized mortality ratio, most striking in women (112-115). Specific pituitary pathologies, especially craniopharyngioma, may convey an increased mortality rate, which is likely to be independent of specific hormonal deficiencies (114). However, bearing in mind the numerical preponderance of pituitary macroadenomas as the cause of hypopituitarism, the overall findings from these studies favor an increase in morbidity and mortality from macrovascular disease and, in one of the Swedish studies, predominantly cerebrovascular disease (113). The increase in cardiovascular mortality in the initial Swedish study (116) was paralleled by a reduction in deaths from malignant disease in males but this has not been a definite feature of subsequent observations.

 

Much debate surrounds the mechanism for increased prevalence of vascular disease. These patients were replaced with glucocorticoids, thyroxine, and in some cases gonadal steroids, which prompted the conclusion that untreated GH deficiency was the major causal factor (117). However, this interpretation assumed that replacement of adrenal and thyroid deficiency was optimal and must also take into account that estrogen deficiency may not have been replaced. In fact, replacement, particularly with hydrocortisone, was often supraphysiological (118-120), while thyroxine replacement was more likely underdosed (10, 120-123), and estrogen is often not replaced in females of the fertile age (124). Recent clinical practice has rectified this mistake and consequently hydrocortisone doses are now significantly lower and thyroxine doses significantly higher than when the original mortality data were collected. Anecdotally, a recent paper on lifelong untreated isolated GH deficiency due to a mutation in the GH releasing hormone (GHRH) receptor gene found no alteration of longevity in this highly selected genetic background (125).Nonetheless, the fact that untreated GH deficiency, in the context of varying degrees of hypopituitarism, is associated with an adverse cardiovascular risk profile provides circumstantial evidence for a causative role for GH deficiency to mediate increased rates of vascular disease (45, 54, 77, 84, 85).

 

DIAGNOSTIC PROCEDURES

 

GH is secreted in a pulsatile fashion with serum measurements varying between peaks and troughs, the latter falling below the assay detection limit of conventional radioimmunoassays. For this reason, a diagnosis of GH deficiency cannot be made by measurement of baseline serum GH concentration although a single serum GH measurement taken fortuitously at the time of a secretory peak may serve to exclude GH deficiency. Therefore, the diagnosis of GH deficiency is dependent on the demonstration of a subnormal rise in serum GH in response to one or more dynamic stimulation tests. Many diagnostic tests have been developed for GH deficiency, most of them for patients with established hypothalamo-pituitary disease with a high a priori test outcome for deficiency. The same tests are now also used for diagnosing GH deficiency in a number of other potential patient populations raising high risk of misuse and wasting of resources. Further, the technical performance of hormone assays is highly variable among different laboratories(126). Thus, diagnosing patients with GH deficiency should first of all consider who should be tested for GH deficiency, the validity of the chosen stimulation test including the proper diagnostic cut-off concentration of GH, and the availability of local resources and expertise. As stated in the 2019 ACCE/ACE paper, cases with no suggestive history such as hypothalamo-pituitary disease or cranial therapy i.e., cases with a low pretest probability or low a priori likelihood of GH deficiency (18), GH stimulation testing should not be performed.

 

The recommendations for stimulation testing of patients for adult GH deficiency are provided in the guidelines mentioned in Fig. 8 (11, 12, 18, 19). Noteworthy all guidelines mention the patients eligible for testing as having either hypothalamo-pituitary pathology, verified GH deficiency in childhood, or have had intracranial irradiation. Options include the insulin tolerance test (ITT), glucagon test, and combinations of arginine and GH releasing hormone (GHRH) or GH secretagogues. Recently, the macimorelin test was approved for the diagnosis of adult GH deficiency (127).

Fig 8. Guideline recommendations for whom to test for GH deficiency.

Fig 9. Plasma human growth hormone (HGH) and blood glucose concentrations after insulin administered intravenously. (PAR, a hypophysectomized patient; other patients, normal). From: Roth et al (128)

The first description of stimulation of GH upon hypoglycemia was published already in 1963 (128) (Fig 9), and the insulin stimulation test (ITT) is still considered the ‘gold’ standard stimulation test for GH deficiency.

 

The ITT is the best validated test, and  has been demonstrated to distinguish reliably between GH responses in patients with structural pituitary disease and those of age matched controls across the adult age range (Fig 10A) (129). A variety of serum GH cut off points have been used to define GH deficiency. However, an international consensus (convened by the Growth Hormone Research Society) has defined severe GH deficiency in adults as a peak response to ITT of <3 µg/L (11).

 

It is essential that the ITT is carried out in dedicated units under strict supervision by experienced staff and it is contraindicated in patients with epilepsy and/or ischemic heart disease. The ITT may have a questionable reproducibility, probably due to low degree of robustness to everyday life as it strongly depends on pre-test events as well as on the patient (130, 131). Further, the ITT holds a certain risk especially in inexperienced hands (132), although it is quite safe in experienced centers (133).

                          

A particular advantage of insulin and glucagon testing is the simultaneous assessment of the adequacy of ACTH reserve. Combinations of GHRH and either arginine, pyridostigmine (or GH secretagogues) are the most potent stimuli of GH secretion and normative data for these tests have been set to define GH deficiency (28, 134-136). These tests appear to be reliable and practical, with few contraindications and the GHRH + arginine test may eventually replace ITT as the diagnostic test of choice (134, 137) in European countries. However, peak GH in response to these tests as well as to the glucagon test is highly affected by BMI, and thus BMI related cut off levels are mandatory (135).

 

Choice of stimulation test may be quite difficult, and the evidence from studies is variable. Furthermore, GHRH is not available in USA, which has prompted reassessment of the use of the glucagon as alternative test when ITT is contraindicated (18, 138-141). However, the diagnostic accuracy of the glucagon stimulation test is unclear especially in patients who are overweight. Recently, the macimorelin test showed promising good overall agreement with the ITT at the same cut point, and seemed unaffected by BMI, age, and sex (127), and is now considered a good alternative.

Fig 10. Results of tests of GH deficiency in normal (○) and hypopituitary patients (●). From: Hoffman et al (129)

As a result of an age related increase in somatostatinergic tone, spontaneous GH secretion declines by approximately 14% per decade of adult life but this does not alter substantially the response to dynamic tests of GH reserve and the same cut-off GH concentrations can be used across the age range (142). However, several tests pose more risks in elderly individuals (e.g., ITT, glucagon) compared to the young (141, 143), and older people are more likely to be obese with a high waist circumference. They are therefore more at risk of getting a false positive test outcome (144, 145), at least if not lowering the cut-off GH concentration. In general, to avoid misdiagnosing hypopituitary patients with GH deficiency, the importance of using local laboratory assay and test specific cut-off concentrations cannot be overemphasized (28, 146-149).

 

Severe obesity may decrease the GH response to insulin hypoglycemia to levels suggestive of GH deficiency but this is a completely reversible phenomenon if weight loss is achieved (Fig 3) (34). Body composition is by far the most important factor that needs to be considered when GH responses are evaluated and waist corrected GH-responses may be superior to BMI-corrected cut-offs (130). However, regarding the impact of BMI or waist circumference on GH-responses, further studies are needed to establish cut-off values also considering age, sex and ethnicity. Glucocorticoid therapy, including substitution therapy, probably reduces GH-responses to the GHRH + arginine test in line with the PD–GHRH test (150). Failure to recognize the impact of obesity on stimulated GH secretion may result in a false positive diagnosis of GH deficiency (28, 34, 37, 149, 151) and it is now standard practice that the diagnosis should be made in conjunction with evidence of structural pituitary disease and/or the documentation of additional pituitary hormone deficiencies. The latter provide robust support for a diagnosis of GH deficiency because of the increasing probability of GH deficiency in the presence of one (c.80%) or more (c.90%) additional pituitary trophic hormone deficiencies (152, 153) (Fig 11). Sadly, one publication (154) documented that many centers do not comply with recommendations, since the percent of patients meeting recommended test-specific cut points varied from 32 to 100%, depending on the stimulation test used. There was no mentioning of laboratory or assay specific cut-off concentrations. The study thereby highlights the need for continued education regarding treatment guidelines for adult GH deficiency, including the testing procedures.

Fig 11. Likelihood of GH deficiency related to number of pituitary hormone deficiencies other than GH deficiency. From: Sönksen PH et al. (155)

GH secretory reserve may also be assessed by measurement of serum concentrations of the GH-dependent peptides IGF-I, IGF binding protein 3 (IGFBP3) and the acid labile subunit of the ternary complex (ALS). Of these, IGF-I is the most sensitive marker of GH action and provides a reliable test of GH reserve in childhood-onset disease. Its diagnostic value for GH deficiency is limited by the fact that between 30 and 40% of individuals with severe GH deficiency of adult-onset will demonstrate a serum IGF-I concentration in the low part of the normal age related reference range (116). Nonetheless, in the absence of liver dysfunction or malnutrition, which may secondarily reduce IGF-I generation, and if determined in the appropriate clinical context of pituitary disease and hypopituitarism, a decreased serum IGF-I provides a strong confirmatory indication for GH deficiency (129).

 

Testing of patients with suspected non-classical causes of hypopituitarism is much more challenging, since most available evidence of diagnostic reliability has been based on patients with structural hypothalamo-pituitary pathology, genetic causes of GH deficiency, documented GH deficiency in childhood, or brain irradiation. In recent years there has been considerable focus on etiologies previously considered rare causes of hypopituitarism. Published series of hypopituitarism in traumatic brain injury  and subarachnoid hemorrhage suggested prevalence rates up to 25-50% (17, 156, 157), and both traumatic brain injury  and subarachnoid hemorrhage were subsequently highlighted in the guidelines as new indication for GH testing (12, 19). Still it is notable that the large majority of these patients had isolated deficiencies and in particular isolated GH deficiency (27). Acknowledging the many aforementioned caveats for the diagnosis of GH deficiency these cases may have been strongly overemphasized also because most data were based on only single testing. The fairness of such concerns was evident in a Danish study showing a low concordance of repeated testing for GH deficiency in TBI (Fig 12) (28), which underlines the importance of stringent testing including a second confirmatory test in patients with low a-priory likelihood of GH deficiency. This approach is consistent with the ACCE/ACE 2019 guidelines (18).

Fig 12. The prevalence of insufficient test responses in the total cohort (A) and in the subgroup undergoing dual testing (B). A, Prevalence of insufficient test responses to either ITT or PD-GHRH/GHRH-arg (i.e., combined tests) in the total cohort of TBI patients (black columns) and healthy controls (white columns), respectively, as defined by either local or guideline-derived cutoffs. Whiskers indicate the 95% CI. GHD was more frequently diagnosed in TBI patients tested by a combined test as compared with ITT, and even more so if guideline cutoff values were applied instead of local cutoffs. The results from healthy controls illustrate the high false-positive rate resulting from application of guideline-derived cutoffs, which was significantly above the generally accepted 2.5% for the combined tests (P = .02). *, P < .005 compared with patients. B, Prevalence of insufficient test responses in the subgroups of patients (black columns, n = 169) and controls (white columns, n = 117) undergoing dual testing, as defined by either local cutoff values or guideline-derived cutoff values. Confirmed insufficiency was defined as a concordant positive result to both the ITT and a combined test. Whiskers indicate the 95% CI. [Reproduced with permission]. From: Klose et al. (28)

Main Conclusions

 

True GH deficiency is an important clinical entity that should be tested, treated, and managed properly. On the other hand, it is important to avoid false diagnosis, which might lead to unnecessary life-long therapy with GH replacement.

 

The diagnosis of GH deficiency is rather simple in patients with a typical structural pathology in the hypothalamo-pituitary region, especially in cases of multiple pituitary hormone deficiencies and low IGF-I concentrations, where the likelihood of GH deficiency exceeds 97% (Fig 11), and a stimulation test is rarely indicated (Fig 8). In all other cases a stimulation test is needed for the diagnosis and in some patients 2 tests should be performed in order to avoid the risk of overtreatment on a false basis. The knowledge of one’s own laboratory performance as well as own reference population data with BMI cut offs for control persons is crucial in interpretation of results. The same holds true for the testing of the other hormone axes, some of which have similar challenges in diagnosing correctly in hypothalamo-pituitary disease states.

 

New indications for GH deficiency testing should not be accepted without prior stringent evaluation of test reliability for the particular condition in question by several tests, and preferably in different laboratories, given that the classical GH deficiency phenotype such as obesity, fatigue and QoL often has other causes than GH deficiency.

 

Table 3. Strategy for Diagnosing Adult GH Deficiency for the Purpose of Replacement Therapy

  • Assess basis for hypopituitary diagnosis
  • Check if the patient is eligible for GH replacement
  • Measure IGF-I age related reference SDS – if below 0 SDS continue testing
  • Assess number of other pituitary hormone deficiencies
  • Make sure other pituitary hormone deficiencies are properly replaced
  • Choose GH stimulation test(s)
  • Perform stimulation test according to:
    • Guidelines
    • Proper local test validation
    • Own reference cut off based on local assay and normal reference population
    • BMI
    • Other confounders
  • MR/CT scan of pituitary in patients with abnormalities
  • QoL assessment (e.g., QoL-AGHDA)(requirement in UK)
  • Severe GH deficiency as defined by the respective GH stimulation tests (fig. 8)

 

GH: Growth Hormone; SDS: Standard Deviation Score; IGF-I: Insulin like Growth factor-I; BMI: Body mass index; MR: Magnetic Resonance; CT: Computer Tomography; QoL: Quality of Life; AGHDA: Adult growth hormone deficiency assessment.

 

RESPONSE TO GH REPLACEMENT IN ADULT GROWTH HORMONE DEFICIENCY

 

Quality of Life and Psychological Well-Being              

 

Potentially, the greatest immediate indication for growth hormone supplementation is in patients who are assessed as having impaired QoL, and this is in some countries, such as the UK, a prerequisite for reimbursement (table 4) (103). This recommendation from NICE is unchanged and has not been updated.

 

Table 4. NICE Recommendations for Treatment with Growth Hormone

NICE has recommended that recombinant human growth hormone should be used only for adults with severe growth hormone deficiency that is severely affecting their quality of life. To be a part of this group, NICE says a person should:

● have a peak growth hormone response of less than 9 mU/L (<3 µg/L) in the ‘insulin tolerance test’ for growth hormone deficiency or a similar low result in another reliable test, and

● have an impaired quality of life because of their growth hormone deficiency (judged using a specific questionnaire called the 'Quality of life assessment of growth hormone deficiency in adults’ designed to assess the quality of life in people with growth hormone deficiency; a person should score at least 11 in this questionnaire), and

● already be receiving replacement hormone treatment for any other deficiencies of pituitary hormones if he or she has one or more other deficiencies.

From: NICE guidelines (103)

 

The early high dose placebo controlled trials suggested that around 50% of these patients demonstrated a significant improvement and a desire to continue with replacement longer term (8). The greatest benefit was shown in patients who had severe GH deficiency and greater distress, in terms of energy and vitality, prior to commencing GH. More recent experience using lower doses with fewer side effects, also indicated clear improvement with wish to continue in >90% of patients selected on the basis of a perceived QoL deficit (53, 158). A six-month course of optimally titrated GH replacement is usually needed before the benefits can be assessed clearly, although many patients show a substantial improvement in QoL within three months. For reasons that are unclear, a small proportion of patients (<20%) may not demonstrate significant subjective benefit in QoL until 9 to 12 months after commencing treatment (159). It is important to recognize that the time taken to achieve a maintenance dose of GH may extend to 12 weeks in some patients and is longer on average in women than in men; this should be recognized in therapeutic trials of GH replacement with a finite time frame. It is clear that the time taken to derive subjective benefit from GH replacement in many patients provides strong evidence against a pure placebo effect in this respect. Furthermore, the duration of benefit in QoL, which has been observed for periods of up to 10 years, is similarly indicative of a therapeutic rather than a placebo phenomenon (106, 111, 160). Patients QoL improves most rapidly in the first 12 months of treatment, but even after this there is continued improvement towards the country specific population mean, with particular improvement in problems socializing, tenseness, and self-confidence, which normalize to the background population (161). The improvement is seen in patients with all etiologies of GH deficiency including previous acromegaly (162-164), isolated GH deficiency (56) and previous Cushing’s disease (99, 100). However, not all aspects of QoL normalize and this is particularly true in patients under 60 yrs of age.

 

The reasons for the differences in QoL outcome between the early studies and current clinical practice has been the subject of much debate and at least three factors are likely to be particularly relevant. Firstly, the initial randomized control trials utilized GH doses based on body weight or surface area and did not take account of the substantial variation in individual responsiveness to GH occurring as a result of gender and other factors. This strategy resulted in excessive GH doses in men and obese subjects and relative undertreatment of women. The adverse symptoms associated with excess GH doses included arthralgia and myalgia, due to GH-induced anti-natriuresis, and it is probable that these factors may have obscured potential subjective benefit. In addition, it is probable that the strict entry criteria inherent in any placebo-controlled study designed to prove concept may have inadvertently eliminated patients who were most likely to demonstrate a benefit in QoL (Table 3) (164). Finally, the current strategy of GH replacement is not to await the full-blown phenotype to develop, but rather to start replacement as soon as the diagnosis is made, as with any other hormone replacement.

 

These latter phenomena are readily evident when baseline indices of QoL in patients enrolled into randomized control trials are compared with those of patients commencing GH replacement selectively in the clinical practice setting (53, 158, 165), and even more so with the changed selection of patients eligible for GH replacement over the years (16).

 

The mechanism for the beneficial effect of GH on well-being and QoL remains speculative (164). GH has been shown to cross the blood brain barrier (166, 167) and to exert physiological effects in the central nervous system as evidenced by the generation of neurotransmitters (166), an effect reduced by progressive aging (168, 169). However, the effects of GH in restoring normal hydration and increasing exercise capacity are additional potential contributors to the positive effects on well-being (170).

 

Table 5. Effects of Growth Hormone Replacement Therapy on Quality of Life in Adults in Published Trials

Reference

GHD onset

(etiology)

N

Dosage per day or titration

Duration

Design (Controls)

Tests

Change in QoL in the GHD adults

Baum et al

(1998)

AO

40

2-6 μg/kg

18 m

PCDB

NHP

PGWB

GHQ

MMPI-2

Cognition tests

= cognition, QoL

Burman et al

(1995)

Mostly

AO

36

2-4 U

21 m

PCDB

NHP

PGWB

HSCL

Spousal report

↑ QoL placebo + GH

groups (HSCL)

↑QoL GH group

(NHP, spousal report)

McGauley et al (1989)

Mostly

AO

24

0.07 U/kg

6 m

PCDB

NHP

PGWB

GHQ

↑ subjective well-being

↑ QoL (NHP)

↑ QoL (PGWB)

Soares et al

(1999)

Not stated

9

0.035 U/kg

6 m

PCDB

HDS

BDI

Cognitive tests

↑ QoL, cognition

Attanasio et al(1997)

AO+ CO

173

12.5 μg/kg

18 m

6m PCDB

12 m open

NHP

=mobility, energy (6 m)

↑mobility, energy (12m)

Beshyah et al

(1995)

AO+ CO

40

0.04 U/kg

18 m

6m PCDB

12 m open

CPRS

GHQ

↑QoL 12m (CPRS)

↑QoL 6m placebo (GHQ)

Caroll et al

(1997)

Not stated

42

0.024 (6m)

0.012 (6m)

μg/kg

12 m

6m PCDB

6m open

NHP

PGWB

↑ QoL on both scales

↑ NHP score in placebo

Mahajan et al

(2004)

AO+CO

25

0.04 (1m)

0.08 (1m)

mg/kg/week, Normal IGF-I

4 m

PCDB

Cross over

NHP

HDRS

MADRS

=mobility, pain

↑energy and emotional reactions

↓social isolation, sleep disturbance

↓depression

Mardh et al

(1994)

AO

124

Not stated

12-18 m

6m PCDB

6-12m open

NHP

PGWB

↑ QoL (NHP)

↑ Well-being

Urushihara et al (2007)

AO+CO

64

0.021-0.042-0.083

mg/kg/week, Normal IGF-I

16 m

24 weeks DBPC

48 weeks Open

SF-36

↑ physical functioning and general health (AO)

↓social functioning and mental health (CO)

Wallymahmed et al (1997)

Mostly

AO

32

0.018 (1m)

0.035 (5m)

U/kg

12 m

6m PCDB

6m open

GHD-LFS

GHD-IS

NHP

HADS

SES

MFS

↑ Self esteem

↑ Energy and emotional

reaction (transient)

Bengtsson et al (1993)

AO

10

13-26 μg/kg

6 m

PCDB

Cross-over

CPRS

SCL-90

↑ QoL (CPRS)

= QoL (SCL-90)

Degerblad et al (1990)

AO

6

0.07-0.09 U/kg

3 m

PCDB

Cross-over

Mood questionnaires

Psychometric

Testing

= mood, cognition

↑ vitality, mental

alertness

Whitehead et al (1992)

AO+ CO

14

0.07 U/kg

6 m

PCDB

Cross-over

PGWB

= QoL, but no ↑ IGF-I

Cuneo et al

(1998)

Mostly

AO

166

0.018 (1 m)

0.036 (11m)

U/kg

12 m

6m PC

6m open

NHP

GHDQ

Social history

↑ QoL 12m (NHP)

= QoL (GHDQ)

Deijen et al

(1998)

CO (men)

48

1-3 U/m2

24 m

PC

Psychological

Testing

= well-being

↑ memory

Florkowski et al

(1998)

AO+ CO

20

0.035 U/kg

3m

Randomized

PC

Cross-over

DSQ

SCL-90

SAS

↑ QoL placebo + GH

groups

Giusti et al

(1998)

AO

25

0.5-1 U

6 m

Randomized

PC

HDS

KSQ

↑ QoL (HDS)

= KSQ

Miller et al

(2010)

AO (Acromegaly)

30

Normal IGF-I

6 m

Randomized PC

AGHDA

SF-36

SQ

↑ QoL (AGHDA)

↑ vitality, mental health, soc functioning, general health

↓ role limitation

Verhelst et al

(1997)

Mostly

AO

148

0.035 U/kg

24 m

6m PC

18m open

NHP

Social history

↑ QoL placebo + GH

↓ sick leave

 hospitalization

Ahmad et al

(2001)

AO

46

Normal IGF-I

3 m

Open

AGHDA

↑ QoL after 1 and 3 m

Abs et al

(2005)

AO+CO (IGHD)

1775

Not stated

12 m

Open (MPHD)

AGHDA

↑ QoL IGHD+MPHD

IGHD=MPHD

Drake et al

(1998)

AO

50

Normal IGF-I

6 m

Open

AGHDA

↑ QoL after 3 and 6 m

Follin et al.

(2010)

CO (ALL)

13

0.2-0.8 mg/d

60 m

Open

(No GH)

Symptom checklist-90

ISSI

= QoL

Gibney et al

(1999)

AO+ CO

11

0.025 U/kg

120 m

Open

(No GH)

NHP

↑ QoL (NHP), energy,

emotional reaction

Gilchrist et al

(2002)

AO+ CO

61

Not stated

108 m

Open

(No GH)

NHP

PGWB

↑ energy (NHP)

↑ vitality (PGWB)

Hernberg-Stahl

et al (2001)

AO

304

0.125-0.25 U/kg

12 m

Open

AGHDA

↑ QoL after 1 m,

higher after 3 m

Höybye et al

(2010)

AO (CD)

1070

Normal IGF-I

36 m

Open

(NFPA)

AGHDA

↑ QoL CD+NFPA

CD > NFPA

Kelestimur et al (2005)

AO (SS)

143

Normal IGF-I

24 m

Open

(NFPA)

AGHDA

↑ QoL SS+NFPA

SS=NFPA

Klose et al.

(2009)

AO (IGHD)

1152

Normal IGF-I

24 m

Open

(MPHD)

AGHDA

↑ QoL IGHD+MPHD

IGHD=MPHD

Koltowska-H et al (2006)

AO

1117

Normal IGF-I

1 – 8 yrs

Open

AGHDA

↑ QoL

Kreitschmann-Andermahr et al. (2008)

AO+CO (TBI)

41

Normal IGF-I

12 m

Open

(NFPA)

 

AGHDA

↑ QoL TBI+NFPA

GHD TBI = GHD NFPA

Link et al.

(2006)

CO (ALL)

14

Normal IGF-I

12 m

Open

Neuropsycho-

logical testing

=

Maiter et al

(2006)

AO+CO (irradiated)

1077

Normal IGF-I

12 m

24 m

Open

(non-irradiated)

AGHDA

↑ QoL irradiated+non-irradiated

irradiated=non-irradiated

Moock et al.

(2009)

Mostly AO

651

Normal IGF-I

12 m

Open

AGHDA

↑ QoL

Mukherjee et al (2005)

AO+CO

(cancer survivors)

97

Normal IGF-I

3-13 m

24-77 m

Open

(pituitary pathology)

PGWB

AGHDA

↑ QoL cancer+pit. GHD cancer survivors = pituitary pathology

Mukherjee et al (2005)

AO+CO

30

Normal IGF-I

3 m

6 m

Open

PGWB

AGHDA

↑ QoL

Murray et al

(1999)

AO + CO

65

Normal IGF-I

8 m

Open

PGWB

AGHDA

↑ QoL

Murray et al

(2001)

CO (cancer)

27

Normal IGF-I

18 m

Open

PGWB

AGHDA

↑ QoL (large, 3 m)

Rosilio et al

(2004)

AO + CO

576

Normal IGF-I

12 m

48 m

Open

QLS-H

↑ QoL

Van der Klaauw et al. (2009)

AO

(Acromegaly)

16

Normal IGF-I

12 m

Open

HADS

MFI-20

NHP

AGHDA

= QoL

Verhelst et al.

(2005)

AO (CP)

721

Normal IGF-I

24 m

Open

(NFPA)

AGHDA

↑ QoL CP+NFPA

CP = NFPA

Wiren et al

(1998)

AO + CO

71

6-12 μg/kg

20-50 m

Open

NHP

PGWB

↑ QoL

QoL, quality of life; GHD, growth hormone deficiency; AO, adult onset; CO, childhood onset; ALL, acute lymphoblastic leukemia CP, craniopharyngioma; CS, Cushing’s Disease; NFPA, non-functional pituitary adenoma; SS, Sheehan’s syndrome; TBI, traumatic brain injury; n, number of subjects; PC, placebo-controlled; PCDB, placebo-controlled, double-blind; open, open label; Tests used to quantify QoL  ↑, =, ↓, change in QoL parameter in GH-treated patients and when available compared to controls (Modified and updated from Hull and Harvey(171) with permission from the authors). From: Klose et al (164)

 

Body Composition: Fat Mass, Fat Distribution, and Lean Body Mass

 

GH replacement produces a significant redistribution of body mass, decreasing body fat, and particularly central fat, and increasing lean body mass (6-9, 172). Body fluid balance is also restored. The beneficial effects of GH on total body fat and its distribution have been examined by means of dual energy X-ray absorptiometry (DEXA), computerized tomography (CT), bioelectrical impedance, and ratio of waist to hip circumference (55, 173) (fig 13) and qualitatively similar results have been obtained with excellent concordance between virtually all reported studies. The restoration of normal total body water may result in an artefactual increment in determinations of lean body mass particularly when the latter is measured by bioelectrical impedance. The abnormal fat distribution in GH deficiency is characterized by an increase in the ratio of waist to hip circumference and during long term follow up, serial measurement of waist circumference provides a simple, rapid and reproducible means of monitoring improvement in body fat distribution.

Fig. 13. Median waist/hip ratio at 0, 6 and 12 months after commencement of GH replacement, men versus women. From: Data from Drake et al (173)

Reductions in body fat are attributed to the lipolytic effect of GH but additional indirect hormonal effects may be important. The conversion of thyroxine to triiodothyronine was shown to be enhanced by GH in early studies of GH replacement (123, 174) although this is a dose related phenomenon and is less evident with the lower doses in current use. However, levothyroxine replacement has very likely not been optimal (121, 122), and increased dosages have improved the lipids over time (121, 122). Also, the enzyme 11ßHSD1 that reduces cortisone to the active hormone cortisol shows increased activity in the GH deficient state and is normalized by low dose GH replacement(81); the consequent increase in cortisol metabolism may result in reduced tissue specific exposure to glucocorticoid in adipocytes and hepatocytes (81). The latter effect provides an additional explanation for decreased total and central fat mass during GH replacement.

 

Serum Lipoprotein Profiles

 

The effect of GH replacement on lipoprotein profiles has been examined in numerous studies using differing dose regimens. Regardless of whether the GH dose has been based on body weight or titrated against serum IGF-I the universal finding has been a reduction of serum total cholesterol, accounted for virtually entirely by a reduction in LDL-cholesterol (48, 69, 160, 175-182). The extent of this reduction is greatest in those patients with higher baseline serum cholesterol (fig 14), and independent on obesity variables (182). The median change in an unselected hypopituitary population is between 0.3 and 0.4 mmol/L (48, 175). Importantly, the improvement in LDL-cholesterol is additive to the effects of HMG CoA reductase inhibitors if the patient is receiving concurrent therapy and possibly even synergistic (173), as well as synergistic with optimization of levothyroxine therapy (120-123). The degree of reduction of serum LDL-cholesterol during GH replacement would predict an overall reduction in cardiovascular events in the range of 20%. In addition, some studies have documented an increase in serum HDL-cholesterol, but serum triglyceride levels remain unchanged. Serum lipoprotein(a) has been shown to increase in some studies in patients who demonstrated favorable changes in LDL-cholesterol (45, 183, 184) but the data remain somewhat contradictory by virtue of lipoprotein(a) assay differences; the overall significance in terms of cardiovascular risk is unclear (185).

Fig 14. Relationship between the lowering of cholesterol (∆Cholesterol) and the pretreatment serum Cholesterol concentration. Derived from data from Florakis et al (175)

Carbohydrate Metabolism and Insulin Sensitivity

 

Untreated GH deficiency of adult onset is associated with reduced insulin sensitivity, which is, at least in part, related to increased central adiposity (45, 77). The latter improves within the first 3 months of GH replacement but this does not result in an immediate improvement in insulin sensitivity (45). In fact, because of the antagonistic effects of GH on the actions of insulin mediated by hepatic effects, and the increase in circulating free fatty acids, there is on average a further decline in insulin sensitivity, which subsequently returns to baseline over the first year of GH replacement therapy (77). The decline in insulin sensitivity during GH therapy is associated with a slight elevation of fasting plasma glucose and a parallel increase in glycated hemoglobin, both within the normal reference range. Importantly, the increment in glycated hemoglobin is not evident in patients with prior abnormalities of glucose tolerance but is significantly correlated with baseline body mass index, the latter emphasizing the importance of additional dietary and lifestyle advice in these patients.

 

Reference to the KIMS database indicates that there is an increased baseline prevalence of impaired glucose tolerance and diabetes mellitus prior to commencing GH replacement but subsequently the incidence of new cases of diabetes is not increased provided the body mass index is accounted for. Thus, a recent study of data from the NordiNet® surveillance database concluded that 4 years' GH-replacement therapy did not adversely affect glucose homeostasis in the majority of adults with GH deficiency (186). Yet, the long-term effects of GH replacement on insulin sensitivity can still not be considered quite clear although they are likely to vary depending on age, duration of pituitary disease, and increase in weight/BMI/waist circumference.

 

Cardiac Function

 

The GH/IGF-I axis is a recognized modulator of cardiac function and a positive inotropic effect of GH/IGF-I occurs early in the natural history of acromegaly. In contrast, GH deficiency is associated with a reduction in left ventricular wall mass and cardiac output, which is most evident in childhood-onset disease. The variable discordance between childhood-onset and adult-onset GH deficiency in this regard is likely to be due to additional factors impacting on cardiac morphology in adult-onset, including an increased prevalence of hypertension. GH replacement results in increased left ventricular wall mass, fractional shortening, stroke volume and favorable changes in the echocardiographically determined e/a ratio reflecting improved diastolic function (8, 65-75). In some studies, in adult-onset patients, left ventricular hypertrophy has been documented during GH replacement, confirming further the heterogeneity in response to GH replacement. Importantly, GH replacement does not increase blood pressure; in fact, a modest reduction may be seen in patients with pre-existing hypertension reflecting increased generation of nitric oxide as a result of activation of nitric oxide synthase.

 

Exercise Capacity and Performance

 

Increased exercise capacity, as measured by maximal oxygen uptake, power output, and isometric muscle strength, has been observed during GH replacement in GH deficient adults (63, 64, 187, 188). A meta-analysis concluded that evidence from short-term controlled studies failed to support a benefit on muscle strength of GH replacement in GH deficient patients, which is likely to occur over a longer time-course, as seen in open-label studies (188) (Fig 15).

 

The impact of these changes for individual patients is variable and dependent on age and previous exercise requirements. It is intuitively probable that the improvements depend at least in part on improvements in lean body mass. However, restoration of normal circulating volume may also play a positive role (170). In addition, improvement in psychological well-being might be expected to enhance physical activity whilst the latter may have a reciprocal beneficial effect on well-being.

Fig 15. Per cent change from baseline in lean body mass (Panel A), fat mass (Panel B), anaerobic power (Panel C) and VO2max (Panel D) following 1 month of placebo and 1 month of GH (randomized controlled study) and 6 months of GH (open-label study) in 18 patients with GH deficiency. From: Chikani et al (170).

Indices of Bone Remodeling and Bone Mineral Density

 

GH deficiency is associated with reduced activity of bone formation and resorption. GH replacement reverses this situation rapidly resulting in increases in markers of bone formation (e.g., osteocalcin and bone specific alkaline phosphatase) and bone resorption (e.g., urine deoxypyridinoline) (Fig 16) (189).

Fig 16. Markers of bone turnover during 18 months of GH (▲) treatment in a randomized, placebo- controlled (O), double blinded study. Values are given as means (^S.E.). The P values for differences of change from baseline between GH- and placebo-treated patients are *P, 0:05; **P, 0:01; ***P, 0:001; ****P, 0:0001: Creat, creatinine. From: Sneppen et al (189).

This increase in bone metabolism eventually results in an increase in bone mineral density (BMD) but this is not evident for approximately 18 months of treatment and is preceded by a reduction attributable to an increase in the bone remodeling space (53, 93, 189-197). The fact that BMD increases under the influence of GH replacement at physiological doses provides important surrogate evidence for an etiological role for GH deficiency in mediating the reduced BMD observed in hypopituitarism. The improvement is quantitatively more obvious in men than women despite the achievement of similar serum IGF-I SD scores and therefore constitutes a genuine difference in gender susceptibility (fig 17).

Fig 17. Bone mineral density of the lumbar spine during long-term GH replacement therapy. From Drake et al (173).

Although the improvement in BMD would predict a reduction in fracture rates confirmation of this necessitates long term follow up. Evidence is now emerging supporting a lower fracture risk with GH replacement (92, 198). A prospective cohort study has shown that GH deficient patients treated with GH before the onset of osteoporosis have a lower fracture risk than those untreated, over a mean follow up of 4.6 years (SD 3.8)(199). Increased fracture risk in child onset GH deficiency women can most likely be explained by interaction between oral estrogen and the GH-IGF-I axis. The adequate substitution rate of testosterone (90%) and GH (94%) may have resulted in significantly lower fracture risk in adult-onset GH deficiency men (198). Finally, although in vitro studies have shown that GH has a direct effect on bone remodeling, present physiological concepts and the results of clinical trials from 1996 to 2008 suggest that the anabolic changes in muscle mass and strength may also contribute to changes in BMD/BMC in GH-treated adult GH deficiency patients (200).

 

GH REPLACEMENT IN ELDERLY HYPOPITUITARY PATIENTS

 

Published work indicates that the baseline characteristics and response to GH replacement in hypopituitary patients aged over 65 years are qualitatively similar to those in younger patients (52, 201-203). Importantly, GH deficiency in the elderly is distinguishable on dynamic tests from the well-recognized physiological reduction in spontaneous GH secretion with advancing age (142). It is therefore appropriate to consider older hypopituitary patients for GH replacement and to apply similar criteria to those outlined above. Elderly people with GH deficiency, in particular women, require less GH than at their earlier age, since they will be either spontaneously postmenopausal or taken off estrogen replacement.

 

TRANSITION BETWEEN PEDIATRIC AND ADULT CARE FOR CHILDHOOD ONSET GH DEFICIENCY

 

The transition from childhood into adulthood is generally a very vulnerable period in any young person’s life. It is therefore pertinent to make the transition as smooth as possible. The best way to do this is to have common transition clinics with both a pediatrician and adult endocrinologist having joint consultations to prepare the adolescent for what is going to happen. The timing can be somewhat individual but aiming at a time around final height and completion of puberty seems appropriate. The pediatrician should prepare the child for this/these consultation(s), and the adult endocrinologist taking over future follow up needs to be aware of the fact that obtaining final height and a post pubertal state does not mean that the adolescent is fully matured in a physiological as well as psychological sense.   

 

It is important to confirm persistence of GH deficiency at the time of completion of linear growth, particularly in children with isolated GH deficiency (18). In the presence of a structural lesion in childhood and multiple hormone deficiencies or some genetic causes, a low IGF-I (in the absence of poorly controlled diabetes, liver disease, or oral estrogen) is sufficient to confirm GH deficiency, without a stimulation test (19). Subsequently, decisions must be taken regarding recommencement of GH or longitudinal clinical observation off treatment. Arguments supporting continuation of GH therapy include the observation of increased accumulation of fat mass off treatment (204, 205) and continued acquisition of bone mass in young adults continuing GH in contrast to static bone mass in those discontinuing treatment at the time of completion of linear growth (206). There is no detriment seen in QoL in those patients who withdraw from GH at the completion of linear growth. There is an apparent improvement in insulin sensitivity but, as is the case during normal puberty, this may not be beneficial in the context of continuing somatic development. Given that the major indication for adult GH replacement is the impairment of QOL, then there is no clear consensus as to which patients should continue therapy seamlessly, virtually without interruption, and in which patients it may be reasonable to undertake a period of careful clinical assessment. A recent observational study has raised concern about discontinuation of GH replacement therapy in pediatrics in severely persistent GH deficiency patients, as well as about the often insufficient dose of GH in the treatment of adult patients (207). Follow-up showed improvement in lipid profile and bone mineral density in severely persistent GH deficiency patients under GH therapy. In multivariate analysis, the associated pituitary deficits seemed stronger determinant factors of metabolic and bone status than GH deficiency per se. A consensus meeting convened by The European Society for Pediatric Endocrinology suggested offering continuation of therapy (after retesting) and monitoring those who decline continuation of treatment. If therapy is continued the optimum dosing strategy has not been clearly defined although a titration approach as outlined above would seem empirically appropriate (208). The Endocrine Society Clinical practice guidelines recommend GH therapy to be continued after adult height to allow full skeletal and muscle maturation, which is often delayed in this population (18, 19).

 

INTERACTIONS WITH OTHER PITUITARY AND ADRENAL HORMONES

 

GH is known to inhibit 11ßHSD-1, therefore favoring metabolism to inactive cortisone over active cortisol (72). Hence patients who are partially ACTH deficient or on suboptimal replacement should be carefully monitored at initiation of GH replacement, which might otherwise lead to partial cortisol deficiency, and risk of Addisonian crisis by even simple infections (118, 209).

 

GH also interacts with the TSH axis (120, 123). Patients without defined TSH deficiency demonstrate a reduction in serum thyroxine (T4) after initiation of GH replacement, although maintain stable serum liothyronine (T3) (10, 210, 211), and patients on thyroxine replacement frequently require an increase in their dose (120, 121, 212).The mechanism remains unclear, but it has been postulated that GH may enhance peripheral conversion of T4 to T3 but also have a central inhibitory effect on TSH release at least in children. Clinicians should therefore be aware that the hypothalamo-pituitary-thyroid axis can very easily be both underdiagnosed and under replaced in GH deficiency, and upon commencement of GH preplacement (120, 121, 213).

 

Women require a higher GH dose than men to achieve a similar increment in IGF-I. GH sensitivity is blunted in females on oral estrogen (214-216). Transdermal estrogen reduces IGF-I generation to a lesser extent than oral estrogen. The effect of estrogen is thought to be mainly due to first pass metabolism inhibiting hepatic synthesis of IGF-I (217, 218). Testosterone stimulates GH secretion centrally, and also amplifies GH stimulation of IGF-I (216, 217). In addition to gonadal steroids, DHEA replacement has been shown to have an impact on IGF-I generation and psychological well-being (219, 220). DHEA improves psychological well-being independently of an effect on IGF-I (229). DHEA has been shown to potentiate IGF-I generation (219, 220) such that females on DHEA replacement require a lower GH dose to achieve the same IGF-I (219, 221). The mechanism is unknown, but DHEA is metabolized to testosterone and it is postulated that increased serum testosterone may be responsible, hence explaining the lack of a DHEA effect in men who are either eugonadal or are on testosterone replacement.

 

GROWTH HORMONE REPLACEMENT

 

Selecting Patients for Growth Hormone Replacement

 

The diagnosis of GH deficiency in adults is usually straightforward and consensus guidelines have been established with generalized acceptance (Fig 8). Nonetheless there is continuing debate regarding the selection of patients for GH replacement. Practice varies between countries and is undoubtedly influenced by availability of funding for treatment. In clinical practice, patients are selected for treatment on the basis of perceived need according to one or more of a number of specific criteria as outlined in Tables 3 and 4, including severe GH deficiency defined by the insulin tolerance test (ITT), glucagon test or alternative tests such as arginine plus growth hormone releasing hormone (GHRH) or the Macimorellin test.

 

Establishing the Maintenance GH Dose 

 

When the indication for GH-replacement has been ascertained, the patient is usually on a low initial dose (0.2 mg daily), but dependent on age, since adolescents during transition may benefit from higher initial doses, as will also women on estrogen therapy (replacement or oral contraceptives) a higher dose may be employed (214, 222-224). The dose titration is monitored by IGF-I concentrations (12, 154, 158), and a number of end organ end responses, which may act as ‘biomarkers’ of the treatment effects (table 6).

 

The doses used in published studies vary widely and much of the published data in this area is derived from dosing schedules established on body weight or surface area criteria which were in effect an extrapolation of earlier pediatric practice. Ongoing assessment in the routine clinical setting has indicated that patients can now be managed on much lower doses (158). Using a widely accepted clinical strategy, patients are commenced on 0.2 mg somatotrophin subcutaneously once a day initially. The dose is reviewed every two to four weeks according to clinical response, serum IGF-I levels, and any side effects and the dose is increased, if necessary, at 4 weekly intervals until the maintenance level is achieved (225). This results in a median dose requirement of 0.4 mg daily with a greater sensitivity to a given dose in male patients so that median dose requirement is lower in men. Several sustained release long-acting GH preparations are currently undergoing clinical trials (226-230) and may become an alternative, with some now being marketed in Asia, US and Europe. However, long-term surveillance of safety and efficacy of long-acting GH analogs are not available to address potential pitfalls of the sustained release preparations (231).Serum IGF-I levels may be in the lower part of the age-related reference range in approximately 40% of patients with adult-onset hypopituitarism before any GH treatment across the total age range and this becomes more likely with advancing age. An empirical strategy is to use the minimum dose of GH, which places the serum IGF-1 level between the median value and the upper limit of the age matched normal range for the individual patient. This approach minimizes the risk of overtreatment and the potential sequelae, which may ensue. Serum IGF binding protein-3 and acid-labile subunit lack sensitivity for the titration of GH replacement and are not recommended for this purpose. IGF-I, however, is regulated by several other factors than GH and changes in body composition can be seen with the addition of GH even without any alteration in the IGF-I level. For this reason other biomarkers of GH action are being sought (232).

 

Table 6. What variables and organ functions should be followed in diagnosed GH deficient adult patients treated with GH replacement?

Variables

·       IGF-I (therapy monitoring, titrate to concentration between 0 and + 2SDS)

·       Other pituitary hormone deficiencies

·       MR/CT scan of pituitary in if abnormalities present

·       Safety (adverse effect)

·       QoL assessment (AGHDA)

Metabolic variables

·       Glucose metabolism

·       Lipids

·       BMI

·       Body composition (waist-hip, fat mass, lean body mass)

·       Dexa scan of bones

·       Physical capacity

·       Cardiovascular

IGF-I: Insulin like Growth factor-I; MR: Magnetic Resonance; CT: Computer Tomography; QoL: Quality of Life; AGHDA: Adult growth hormone deficiency assessment: BMI: Body mass index                     

 

Adverse Effects

 

The main adverse effects directly attributable to GH replacement result from the correction of the sodium and water depletion present in GH deficiency patients. This manifests as arthralgia, myalgia, edema, and carpal tunnel syndrome and are usually rapidly reversible with GH dose reduction. They were predominantly a feature of the early experience when GH dose was determined by body weight rather than being based on a titration regimen commencing with a low starting dose as described above. Such adverse effects were predictably more frequent in male patients reflecting their greater sensitivity to GH. Benign intracranial hypertension is a recognized complication of GH replacement in pediatric practice but is much less likely in adult patients especially when low doses are used. However, persistent severe headache should prompt examination and investigation to exclude raised intracranial pressure. The potential mitogenic effects of IGF-I have raised concerns regarding a possible increased risk of either neoplasia or regrowth of residual pituitary and peripituitary tumors. Extensive surveillance studies based on large multinational databases, including several thousand patients on GH replacement followed longitudinally, have not demonstrated an increased incidence of de novo neoplasia and prospective magnetic resonance imaging studies have not indicated an increased risk of pituitary or parasellar tumor regrowth (233, 234). In the childhood cancer survivor study (235) there was no increased risk of recurrence over 5 years follow-up in those who received GH and on 15-year follow-up of patients with previous cranial irradiation who receive GH replacement there was no increased risk of malignancy (236). In addition, there has been no correlation between the serum IGF-I level within the normal reference range and risk of further malignancy (237).

 

Mortality

 

Definition of the precise relationship between GH deficiency and mortality must await long term observations of mortality rate in patients on GH replacement set against background mortality rates in the general population adjusted for national variations but a recent Dutch study provides some evidence that mortality is not increased by replacement and may play a role in normalizing it (particularly in men) (238).

Fig 18. Mortality in hypopituitary patients with-/without GH replacement. From: Gaillard et al (26)

The potential impact of GH replacement on the increased mortality rates described in hypopituitary patients can only be determined by long-term surveillance of treated patients in comparison with normal population data. The multinational databases designed to monitor safety of long-term GH replacement may provide useful information in this regard. Reassuringly, the mortality rates in the KIMS database were similar to the background populations (fig 18), and a later large study in a much larger population with longer follow-up did not observe any increased mortality in GH treated GH deficient patients in the KIMS database (26). Danish nationwide studies have indicated that mortality was not increased in GH treated patients with childhood onset GH deficiency (239, 240), but was highly dependent on the primary cause of GH deficiency (241), since the primary causes of childhood onset GH deficiencyand concomitant diseases severely impaired socioeconomic conditions and impacted mortality; and only the subgroup of patients with idiopathic GH deficiency conditions was similar to the background population. In two Swedish publications there was evidence that hypocortisolism during acute stress, and de novo malignant brain tumors contributed to increased mortality (242), and GH deficient men receiving GH treatment had a mortality rate no different from the background population. In women, after exclusion of high-risk patients, mortality was not different from the background population except for CVD. Mortality due to malignancies was not elevated in adults receiving GH treatment. Next to gender, the heterogeneous etiology influences mortality in GH deficient adults with GH treatment (243). In the French SAGhE study mortality rates were increased in their population of adults treated as children with recombinant GH, particularly in those who had received the highest doses. Specific effects were detected in terms of death due to bone tumors or cerebral hemorrhage but not for all cancers. These results highlight the need for additional studies of long-term mortality and morbidity after GH treatment in childhood (244). Thus, the more recent studies have been reassuring concerning GH replacement and mortality, since those groups with higher mortality seem to have been limited to patient groups with a prior higher risk due to concomitant confounding mortality risks.

 

COSTS VERSUS BENEFITS OF GH REPLACEMENT THERAPY

 

Population studies in Sweden have documented a significantly greater medical and social burden for patients with established hypopituitarism. This continuing cost occurs irrespective of the initial cost of treating the pituitary pathology and derives from issues including unemployment, early retirement, depressive illness, and requirement for disability pension. A social circumstances analysis of the KIMS database has shown that approximately 11% of males and 31% of females require assistance with activities of daily living (245) (fig 19). Additional treatment cost factors, which might be inferred from risk factor profiles in adult GH deficiency populations, include increased prevalence of ischemic heart disease and increased fracture rates. So, whilst the cost of GH replacement to the hypopituitary population is easily determined, matching this with data for economic benefit requires a quantification of long-term complications arising from surrogate markers for long-term morbidity observed in GH deficient patients. Assessments of the effectiveness of GH replacement over time is influenced by the changing characteristics of the patients, with lower doses of GH replacement being used and a shorter period of time from diagnosis of GH deficiency to treatment (181). This means that accurate assessments of cost benefit using long-term data is not yet possible, and probably never will be. A recent study based on patients enrolled into the KIMS database has demonstrated significant reductions in the numbers of patients requiring assistance with the activities of daily living, a decrease in medical consultations and a decrease in hospital in-patient stays over a period of 24 months of GH replacement (fig 20).

Fig 19. Activity of daily living in adult patient with GH deficiency before and after GH replacement therapy. From: Hernberg-Stahl et al (245)

Fig 20. Sick leave and length of hospital stays in adult patients with GH deficiency before and after GH replacement therapy. From: Hernberg-Stahl et al (245).

ADHERENCE TO MANAGEMENT OF ADULT GROWTH HORMONE DEFICIENCY IN CLINICAL PRACTICE

 

Despite 30 years of evidence, guidelines, and clinical experience with adult GH deficiency and its management, a very recently published survey within the auspices of European Society of Endocrinology (246) has indicated that despite many guidelines on safety and efficacy of GH replacement in deficient patients as well as the above health economic considerations, GH replacement is still not available or reimbursed in all European countries. It also seems that both health care professionals and health administrators need improved knowledge to optimize the care of adults with GH deficiency. The publication results were further commented upon (247) indicating that in some health care communities reimbursement has been a major issue, because the discussion of GH replacement in truly GH deficient adults has been mixed with the discussion of utilizing GH therapy for sporting enhancement or for anti-ageing purposes, both of which are strongly discouraged. Also continuous presentations of unclear indications for GH diagnosis and replacement in small cohorts of mixed etiologies also pollute the field of health care discussions of proper clear cut cases of GH deficiency according to the official guidelines (248).

 

REFERENCES

 

  1. Cushing H. The Pituitary Body and its Disorders: Clinical States produced by Disorders of the Hypophysis Cerebri. The Journal of Laryngology, Rhinology, and Otology. 1913;28(7):382-90.
  2. Raben MS. Growth hormone. 2. Clinical use of human growth hormone. N Engl J Med. 1962;266:82-6 concl.
  3. Utiger RD, Parker ML, Daughaday WH. Studies on human growth hormone. I. A radio-immunoassay for human growth hormone. The Journal of clinical investigation. 1962;41(2):254-61.
  4. Jorgensen JO, Pedersen SA, Thuesen L, Jorgensen J, Ingemann-Hansen T, Skakkebaek NE, et al. Beneficial effects of growth hormone treatment in GH-deficient adults. Lancet. 1989;1(8649):1221-5.
  5. Salomon F, Cuneo RC, Hesp R, Sonksen PH. The effects of treatment with recombinant human growth hormone on body composition and metabolism in adults with growth hormone deficiency. NEnglJMed. 1989;321(26):1797-803.
  6. Cuneo RC, Salomon F, McGauley GA, Sönksen PH. The growth hormone deficiency syndrome in adults. Clin Endocrinol (Oxf). 1992;37(5):387-97.
  7. de Boer H, Blok GJ, Van der Veen EA. Clinical aspects of growth hormone deficiency in adults. Endocrine reviews. 1995;16(1):63-86.
  8. Carroll PV, Christ ER, Bengtsson BA, Carlsson L, Christiansen JS, Clemmons D, et al. Growth hormone deficiency in adulthood and the effects of growth hormone replacement: a review. Growth Hormone Research Society Scientific Committee. The Journal of Clinical Endocrinology and Metabolism. 1998;83(2):382-95.
  9. Littley MD, Shalet SM, Beardwell CG, Ahmed SR, Applegate G, Sutton ML. Hypopituitarism following external radiotherapy for pituitary tumours in adults. The Quarterly journal of medicine. 1989;70(262):145-60.
  10. Feldt-Rasmussen U, Klose M. Central hypothyroidism and its role for cardiovascular risk factors in hypopituitary patients. Endocrine. 2016;54(1):15-23.
  11. Consensus guidelines for the diagnosis and treatment of adults with growth hormone deficiency: summary statement of the Growth Hormone Research Society Workshop on Adult Growth Hormone Deficiency. The Journal of Clinical Endocrinology and Metabolism. 1998;83(2):379-81.
  12. Ho KK. Consensus guidelines for the diagnosis and treatment of adults with GH deficiency II: a statement of the GH Research Society in association with the European Society for Pediatric Endocrinology, Lawson Wilkins Society, European Society of Endocrinology, Japan Endocrine Society, and Endocrine Society of Australia. European Journal of Endocrinology. 2007;157(6):695-700.
  13. Ho KY, Hoffman DM. Growth hormone replacement therapy in adults – pros and cons. Larsson C, Butenandt O, editors. Tel Aviv/London: Freund Publishing House; 1993.
  14. Höybye C, Beck-Peccoz P, Simsek S, Zabransky M, Zouater H, Stalla G, et al. Safety of current recombinant human growth hormone treatments for adults with growth hormone deficiency and unmet needs. Expert opinion on drug safety. 2020;19(12):1539-48.
  15. Webb SM, Strasburger CJ, Mo D, Hartman ML, Melmed S, Jung H, et al. Changing patterns of the adult growth hormone deficiency diagnosis documented in a decade-long global surveillance database. J Clin Endocrinol Metab. 2009;94(2):392-9.
  16. Höybye C, Burman P, Feldt-Rasmussen U, Hey-Hadavi J, Aydin F, Camacho-Hubner C, et al. Change in baseline characteristics over 20 years of adults with growth hormone (GH) deficiency on GH replacement therapy. Eur J Endocrinol. 2019;181(6):629-38.
  17. Klose M, Feldt-Rasmussen U. Chronic endocrine consequences of traumatic brain injury - what is the evidence? Nature reviews Endocrinology. 2018;14(1):57-62.
  18. Yuen KCJ, Biller BMK, Radovick S, Carmichael JD, Jasim S, Pantalone KM, et al. AMERICAN ASSOCIATION OF CLINICAL ENDOCRINOLOGISTS AND AMERICAN COLLEGE OF ENDOCRINOLOGY GUIDELINES FOR MANAGEMENT OF GROWTH HORMONE DEFICIENCY IN ADULTS AND PATIENTS TRANSITIONING FROM PEDIATRIC TO ADULT CARE. Endocrine practice : official journal of the American College of Endocrinology and the American Association of Clinical Endocrinologists. 2019;25(11):1191-232.
  19. Molitch ME, Clemmons DR, Malozowski S, Merriam GR, Vance ML. Evaluation and treatment of adult growth hormone deficiency: an Endocrine Society clinical practice guideline. The Journal of Clinical Endocrinology and Metabolism. 2011;96(6):1587-609.
  20. Fernandez A, Karavitaki N, Wass JA. Prevalence of pituitary adenomas: a community-based, cross-sectional study in Banbury (Oxfordshire, UK). Clin Endocrinol (Oxf). 2010;72(3):377-82.
  21. Agustsson TT, Baldvinsdottir T, Jonasson JG, Olafsdottir E, Steinthorsdottir V, Sigurdsson G, et al. The epidemiology of pituitary adenomas in Iceland, 1955-2012: a nationwide population-based study. Eur J Endocrinol. 2015;173(5):655-64.
  22. Daly AF, Beckers A. The Epidemiology of Pituitary Adenomas. Endocrinology and metabolism clinics of North America. 2020;49(3):347-55.
  23. Stochholm K, Gravholt CH, Laursen T, Jorgensen JO, Laurberg P, Andersen M, et al. Incidence of GH deficiency - a nationwide study. European Journal of Endocrinology. 2006;155(1):61-71.
  24. Parkin JM. Incidence of growth hormone deficiency. Archives of disease in childhood. 1974;49(11):904-5.
  25. Sassolas G, Chazot FB, Jaquet P, Bachelot I, Chanson P, Rudelli CC, et al. GH deficiency in adults: an epidemiological approach. Eur J Endocrinol. 1999;141(6):595-600.
  26. Gaillard RC, Mattsson AF, Akerblad AC, Bengtsson B, Cara J, Feldt-Rasmussen U, et al. Overall and cause-specific mortality in GH-deficient adults on GH replacement. Eur J Endocrinol. 2012;166(6):1069-77.
  27. Klose M, Feldt-Rasmussen U. Hypopituitarism in Traumatic Brain Injury-A Critical Note. Journal of clinical medicine. 2015;4(7):1480-97.
  28. Klose M, Stochholm K, Janukonyte J, Lehman CL, Frystyk J, Andersen M, et al. Prevalence of posttraumatic growth hormone deficiency is highly dependent on the diagnostic set-up: results from The Danish National Study on Posttraumatic Hypopituitarism. The Journal of Clinical Endocrinology and Metabolism. 2014;99(1):101-10.
  29. Barroso-Sousa R, Barry WT, Garrido-Castro AC, Hodi FS, Min L, Krop IE, et al. Incidence of Endocrine Dysfunction Following the Use of Different Immune Checkpoint Inhibitor Regimens: A Systematic Review and Meta-analysis. JAMA oncology. 2018;4(2):173-82.
  30. Torino F, Barnabei A, Paragliola RM, Marchetti P, Salvatori R, Corsello SM. Endocrine side-effects of anti-cancer drugs: mAbs and pituitary dysfunction: clinical evidence and pathogenic hypotheses. Eur J Endocrinol. 2013;169(6):R153-64.
  31. Gleeson HK, Gattamaneni HR, Smethurst L, Brennan BM, Shalet SM. Reassessment of growth hormone status is required at final height in children treated with growth hormone replacement after radiation therapy. J Clin Endocrinol Metab. 2004;89(2):662-6.
  32. Gasco V, Corneli G, Beccuti G, Prodam F, Rovere S, Bellone J, et al. Retesting the childhood-onset GH-deficient patient. Eur J Endocrinol. 2008;159 Suppl 1:S45-52.
  33. Berberoğlu M, Sıklar Z, Darendeliler F, Poyrazoğlu S, Darcan S, Işgüven P, et al. Evaluation of permanent growth hormone deficiency (GHD) in young adults with childhood onset GHD: a multicenter study. Journal of clinical research in pediatric endocrinology. 2008;1(1):30-7.
  34. Rasmussen MH, Hvidberg A, Juul A, Main KM, Gotfredsen A, Skakkebaek NE, et al. Massive weight loss restores 24-hour growth hormone release profiles and serum insulin-like growth factor-I levels in obese subjects. The Journal of Clinical Endocrinology and Metabolism. 1995;80(4):1407-15.
  35. De Marinis L, Bianchi A, Mancini A, Gentilella R, Perrelli M, Giampietro A, et al. Growth hormone secretion and leptin in morbid obesity before and after biliopancreatic diversion: relationships with insulin and body composition. J Clin Endocrinol Metab. 2004;89(1):174-80.
  36. Makimura H, Stanley T, Mun D, You SM, Grinspoon S. The effects of central adiposity on growth hormone (GH) response to GH-releasing hormone-arginine stimulation testing in men. The Journal of Clinical Endocrinology and Metabolism. 2008;93(11):4254-60.
  37. Rasmussen MH. Obesity, growth hormone and weight loss. Molecular and cellular endocrinology. 2010;316(2):147-53.
  38. Klose M, Jonsson B, Abs R, Popovic V, Koltowska-Häggström M, Saller B, et al. From isolated GH deficiency to multiple pituitary hormone deficiency: an evolving continuum - a KIMS analysis. Eur J Endocrinol. 2009;161 Suppl 1:S75-83.
  39. Blum WF, Deal C, Zimmermann AG, Shavrikova EP, Child CJ, Quigley CA, et al. Development of additional pituitary hormone deficiencies in pediatric patients originally diagnosed with idiopathic isolated GH deficiency. Eur J Endocrinol. 2014;170(1):13-21.
  40. Child CJ, Blum WF, Deal C, Zimmermann AG, Quigley CA, Drop SL, et al. Development of additional pituitary hormone deficiencies in pediatric patients originally diagnosed with isolated growth hormone deficiency due to organic causes. Eur J Endocrinol. 2016;174(5):669-79.
  41. Arafah BM, Kailani SH, Nekl KE, Gold RS, Selman WR. Immediate recovery of pituitary function after transsphenoidal resection of pituitary macroadenomas. The Journal of Clinical Endocrinology and Metabolism. 1994;79(2):348-54.
  42. Fatemi N, Dusick JR, Mattozo C, McArthur DL, Cohan P, Boscardin J, et al. Pituitary hormonal loss and recovery after transsphenoidal adenoma removal. Neurosurgery. 2008;63(4):709-18; discussion 18-9.
  43. Pedersen MB, Dukanovic S, Springborg JB, Andreassen M, Krogh J. Endocrine function after transsphenoidal surgery in patients with non-functioning pituitary adenomas: A systematic review and meta-analysis. Neuroendocrinology. 2022.
  44. Jørgensen JO, Müller J, Møller J, Wolthers T, Vahl N, Juul A, et al. Adult growth hormone deficiency. Hormone research. 1994;42(4-5):235-41.
  45. Weaver JU, Monson JP, Noonan K, John WG, Edwards A, Evans KA, et al. The effect of low dose recombinant human growth hormone replacement on regional fat distribution, insulin sensitivity, and cardiovascular risk factors in hypopituitary adults. J Clin Endocrinol Metab. 1995;80(1):153-9.
  46. Rosén T, Bosaeus I, Tölli J, Lindstedt G, Bengtsson BA. Increased body fat mass and decreased extracellular fluid volume in adults with growth hormone deficiency. Clin Endocrinol (Oxf). 1993;38(1):63-71.
  47. Li Voon Chong JS, Groves T, Foy P, Wallymahmed ME, MacFarlane IA. Elderly people with hypothalamic-pituitary disease and untreated GH deficiency: clinical outcome, body composition, lipid profiles and quality of life after 2 years compared to controls. Clin Endocrinol (Oxf). 2002;56(2):175-81.
  48. Bengtsson BA, Abs R, Bennmarker H, Monson JP, Feldt-Rasmussen U, Hernberg-Stahl E, et al. The effects of treatment and the individual responsiveness to growth hormone (GH) replacement therapy in 665 GH-deficient adults. KIMS Study Group and the KIMS International Board. J Clin Endocrinol Metab. 1999;84(11):3929-35.
  49. Gomes-Santos E, Salvatori R, Ferrão TO, Oliveira CR, Diniz RD, Santana JA, et al. Increased visceral adiposity and cortisol to cortisone ratio in adults with congenital lifetime isolated GH deficiency. J Clin Endocrinol Metab. 2014;99(9):3285-9.
  50. Fideleff HL, Boquete HR, Stalldecker G, Giaccio AV, Sobrado PG. Comparative results of a 4-year study on cardiovascular parameters, lipid metabolism, body composition and bone mass between untreated and treated adult growth hormone deficient patients. Growth hormone & IGF research : official journal of the Growth Hormone Research Society and the International IGF Research Society. 2008;18(4):318-24.
  51. Mersebach H, Feldt-Rasmussen U. Growth hormone and body composition. Frontiers of hormone research. 2005;33:185-95.
  52. Feldt-Rasmussen U, Wilton P, Jonsson P. Aspects of growth hormone deficiency and replacement in elderly hypopituitary adults. Growth HormIGFRes. 2004;14 Suppl A:S51-S8.
  53. Drake WM, Rodríguez-Arnao J, Weaver JU, James IT, Coyte D, Spector TD, et al. The influence of gender on the short and long-term effects of growth hormone replacement on bone metabolism and bone mineral density in hypopituitary adults: a 5-year study. Clin Endocrinol (Oxf). 2001;54(4):525-32.
  54. Johansson JO, Landin K, Tengborn L, Rosén T, Bengtsson BA. High fibrinogen and plasminogen activator inhibitor activity in growth hormone-deficient adults. Arteriosclerosis and thrombosis : a journal of vascular biology. 1994;14(3):434-7.
  55. Klose M, Watt T, Brennum J, Feldt-Rasmussen U. Posttraumatic hypopituitarism is associated with an unfavorable body composition and lipid profile, and decreased quality of life 12 months after injury. The Journal of Clinical Endocrinology and Metabolism. 2007;92(10):3861-8.
  56. Abs R, Mattsson AF, Bengtsson BA, Feldt-Rasmussen U, Goth MI, Koltowska-Haggstrom M, et al. Isolated growth hormone (GH) deficiency in adult patients: baseline clinical characteristics and responses to GH replacement in comparison with hypopituitary patients. A sub-analysis of the KIMS database. Growth HormIGFRes. 2005;15(5):349-59.
  57. Merola B, Sofia M, Longobardi S, Fazio S, Micco A, Esposito V, et al. Impairment of lung volumes and respiratory muscle strength in adult patients with growth hormone deficiency. Eur J Endocrinol. 1995;133(6):680-5.
  58. Janssen YJ, Doornbos J, Roelfsema F. Changes in muscle volume, strength, and bioenergetics during recombinant human growth hormone (GH) therapy in adults with GH deficiency. J Clin Endocrinol Metab. 1999;84(1):279-84.
  59. Modesto Mde J, Amer NM, Erichsen O, Hernandez S, dos Santos CD, de Carvalho J, et al. Muscle strength and body composition during the transition phase in patients treated with recombinant GH to final height. Journal of pediatric endocrinology & metabolism : JPEM. 2014;27(9-10):813-20.
  60. Johannsson G, Sverrisdóttir YB, Ellegård L, Lundberg PA, Herlitz H. GH increases extracellular volume by stimulating sodium reabsorption in the distal nephron and preventing pressure natriuresis. J Clin Endocrinol Metab. 2002;87(4):1743-9.
  61. Simpson JA, Lobo DN, Anderson JA, Macdonald IA, Perkins AC, Neal KR, et al. Body water compartment measurements: a comparison of bioelectrical impedance analysis with tritium and sodium bromide dilution techniques. Clinical nutrition (Edinburgh, Scotland). 2001;20(4):339-43.
  62. Møller J, Frandsen E, Fisker S, Jørgensen JO, Christiansen JS. Decreased plasma and extracellular volume in growth hormone deficient adults and the acute and prolonged effects of GH administration: a controlled experimental study. Clin Endocrinol (Oxf). 1996;44(5):533-9.
  63. Jørgensen JO, Vahl N, Hansen TB, Thuesen L, Hagen C, Christiansen JS. Growth hormone versus placebo treatment for one year in growth hormone deficient adults: increase in exercise capacity and normalization of body composition. Clin Endocrinol (Oxf). 1996;45(6):681-8.
  64. 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.
  65. Lombardi G, Colao A, Cuocolo A, Longobardi S, Di Somma C, Orio F, et al. Cardiological aspects of growth hormone and insulin-like growth factor-I. Journal of pediatric endocrinology & metabolism : JPEM. 1997;10(6):553-60.
  66. Colao A, Cuocolo A, Di Somma C, Cerbone G, Della Morte AM, Nicolai E, et al. Impaired cardiac performance in elderly patients with growth hormone deficiency. J Clin Endocrinol Metab. 1999;84(11):3950-5.
  67. Thuesen L, Jørgensen JO, Müller JR, Kristensen BO, Skakkebaek NE, Vahl N, et al. Short and long-term cardiovascular effects of growth hormone therapy in growth hormone deficient adults. Clin Endocrinol (Oxf). 1994;41(5):615-20.
  68. Lanes R, Gunczler P, Lopez E, Esaa S, Villaroel O, Revel-Chion R. Cardiac mass and function, carotid artery intima-media thickness, and lipoprotein levels in growth hormone-deficient adolescents. J Clin Endocrinol Metab. 2001;86(3):1061-5.
  69. Maison P, Chanson P. Cardiac effects of growth hormone in adults with growth hormone deficiency: a meta-analysis. Circulation. 2003;108(21):2648-52.
  70. Moisey R, Orme S, Barker D, Lewis N, Sharp L, Clements RE, et al. Cardiac functional reserve is diminished in growth hormone-deficient adults. Cardiovascular therapeutics. 2009;27(1):34-41.
  71. Thomas JD, Dattani A, Zemrak F, Burchell T, Akker SA, Gurnell M, et al. Characterisation of myocardial structure and function in adult-onset growth hormone deficiency using cardiac magnetic resonance. Endocrine. 2016;54(3):778-87.
  72. Mihaila S, Mincu RI, Rimbas RC, Dulgheru RE, Dobrescu R, Magda SL, et al. Growth hormone deficiency in adults impacts left ventricular mechanics: a two-dimensional speckle-tracking study. The Canadian journal of cardiology. 2015;31(6):752-9.
  73. Sneppen SB, Steensgaard-Hansen F, Feldt-Rasmussen U. Cardiac effects of low-dose growth hormone replacement therapy in growth hormone-deficient adults. An 18-month randomised, placebo-controlled, double-blind study. Hormone research. 2002;58(1):21-9.
  74. Andreassen M, Faber J, Kjaer A, Petersen CL, Kristensen L. Cardiac function in growth hormone deficient patients before and after 1 year with replacement therapy: a magnetic resonance imaging study. Pituitary. 2011;14(1):1-10.
  75. Ozdogru I, Tanriverdi F, Dogan A, Kaya MG, Tugrul Inanc M, Kalay N, et al. Impaired longitudinal myocardial velocities in patients with growth hormone deficiency improves after hormone replacement therapy. Journal of the American Society of Echocardiography : official publication of the American Society of Echocardiography. 2007;20(9):1093-9.
  76. Jørgensen JO, Krag M, Jessen N, Nørrelund H, Vestergaard ET, Møller N, et al. Growth hormone and glucose homeostasis. Hormone research. 2004;62 Suppl 3:51-5.
  77. Fowelin J, Attvall S, Lager I, Bengtsson BA. Effects of treatment with recombinant human growth hormone on insulin sensitivity and glucose metabolism in adults with growth hormone deficiency. Metabolism. 1993;42(11):1443-7.
  78. Giovannini L, Tirabassi G, Muscogiuri G, Di Somma C, Colao A, Balercia G. Impact of adult growth hormone deficiency on metabolic profile and cardiovascular risk [Review]. Endocrine journal. 2015;62(12):1037-48.
  79. Oliveira CR, Salvatori R, Barreto-Filho JA, Rocha IE, Mari A, Pereira RM, et al. Insulin sensitivity and β-cell function in adults with lifetime, untreated isolated growth hormone deficiency. J Clin Endocrinol Metab. 2012;97(3):1013-9.
  80. Vicente TA, Rocha IE, Salvatori R, Oliveira CR, Pereira RM, Souza AH, et al. Lifetime congenital isolated GH deficiency does not protect from the development of diabetes. Endocrine connections. 2013;2(2):112-7.
  81. Gelding SV, Taylor NF, Wood PJ, Noonan K, Weaver JU, Wood DF, et al. The effect of growth hormone replacement therapy on cortisol-cortisone interconversion in hypopituitary adults: evidence for growth hormone modulation of extrarenal 11 beta-hydroxysteroid dehydrogenase activity. Clin Endocrinol (Oxf). 1998;48(2):153-62.
  82. Roemmler J, Kuenkler M, Schneider HJ, Dieterle C, Schopohl J. Comparison of glucose and lipid metabolism and bone mineralization in patients with growth hormone deficiency with and without long-term growth hormone replacement. Metabolism. 2010;59(3):350-8.
  83. Elbornsson M, Götherström G, Bosæus I, Bengtsson B, Johannsson G, Svensson J. Fifteen years of GH replacement improves body composition and cardiovascular risk factors. Eur J Endocrinol. 2013;168(5):745-53.
  84. Markussis V, Beshyah SA, Fisher C, Sharp P, Nicolaides AN, Johnston DG. Detection of premature atherosclerosis by high-resolution ultrasonography in symptom-free hypopituitary adults. Lancet. 1992;340(8829):1188-92.
  85. Capaldo B, Patti L, Oliviero U, Longobardi S, Pardo F, Vitale F, et al. Increased arterial intima-media thickness in childhood-onset growth hormone deficiency. J Clin Endocrinol Metab. 1997;82(5):1378-81.
  86. Johansson AG, Burman P, Westermark K, Ljunghall S. The bone mineral density in acquired growth hormone deficiency correlates with circulating levels of insulin-like growth factor I. Journal of internal medicine. 1992;232(5):447-52.
  87. Kaufman JM, Taelman P, Vermeulen A, Vandeweghe M. Bone mineral status in growth hormone-deficient males with isolated and multiple pituitary deficiencies of childhood onset. J Clin Endocrinol Metab. 1992;74(1):118-23.
  88. Bing-You RG, Denis MC, Rosen CJ. Low bone mineral density in adults with previous hypothalamic-pituitary tumors: correlations with serum growth hormone responses to GH-releasing hormone, insulin-like growth factor I, and IGF binding protein 3. Calcified tissue international. 1993;52(3):183-7.
  89. Rosén T, Hansson T, Granhed H, Szucs J, Bengtsson BA. Reduced bone mineral content in adult patients with growth hormone deficiency. Acta endocrinologica. 1993;129(3):201-6.
  90. de Boer H, Blok GJ, van Lingen A, Teule GJ, Lips P, van der Veen EA. Consequences of childhood-onset growth hormone deficiency for adult bone mass. Journal of bone and mineral research : the official journal of the American Society for Bone and Mineral Research. 1994;9(8):1319-26.
  91. Holmes SJ, Economou G, Whitehouse RW, Adams JE, Shalet SM. Reduced bone mineral density in patients with adult onset growth hormone deficiency. J Clin Endocrinol Metab. 1994;78(3):669-74.
  92. Wuster C, Abs R, Bengtsson BA, Bennmarker H, Feldt-Rasmussen U, Hernberg-Stahl E, et al. The influence of growth hormone deficiency, growth hormone replacement therapy, and other aspects of hypopituitarism on fracture rate and bone mineral density. JBone MinerRes. 2001;16(2):398-405.
  93. Vestergaard P, Jørgensen JO, Hagen C, Hoeck HC, Laurberg P, Rejnmark L, et al. Fracture risk is increased in patients with GH deficiency or untreated prolactinomas--a case-control study. Clin Endocrinol (Oxf). 2002;56(2):159-67.
  94. Mazziotti G, Doga M, Frara S, Maffezzoni F, Porcelli T, Cerri L, et al. Incidence of morphometric vertebral fractures in adult patients with growth hormone deficiency. Endocrine. 2016;52(1):103-10.
  95. Rosén T, Wilhelmsen L, Landin-Wilhelmsen K, Lappas G, Bengtsson BA. Increased fracture frequency in adult patients with hypopituitarism and GH deficiency. Eur J Endocrinol. 1997;137(3):240-5.
  96. Lange M, Müller J, Svendsen OL, Kastrup KW, Juul A, Feldt-Rasmussen U. The impact of idiopathic childhood-onset growth hormone deficiency (GHD) on bone mass in subjects without adult GHD. Clin Endocrinol (Oxf). 2005;62(1):18-23.
  97. Tritos NA, Greenspan SL, King D, Hamrahian A, Cook DM, Jönsson PJ, et al. Unreplaced sex steroid deficiency, corticotropin deficiency, and lower IGF-I are associated with lower bone mineral density in adults with growth hormone deficiency: a KIMS database analysis. J Clin Endocrinol Metab. 2011;96(5):1516-23.
  98. Mazziotti G, Porcelli T, Bianchi A, Cimino V, Patelli I, Mejia C, et al. Glucocorticoid replacement therapy and vertebral fractures in hypopituitary adult males with GH deficiency. Eur J Endocrinol. 2010;163(1):15-20.
  99. Feldt-Rasmussen U, Abs R, Bengtsson BA, Bennmarker H, Bramnert M, Hernberg-Ståhl E, et al. Growth hormone deficiency and replacement in hypopituitary patients previously treated for acromegaly or Cushing's disease. Eur J Endocrinol. 2002;146(1):67-74.
  100. Höybye C, Ragnarsson O, Jönsson PJ, Koltowska-Häggström M, Trainer P, Feldt-Rasmussen U, et al. Clinical features of GH deficiency and effects of 3 years of GH replacement in adults with controlled Cushing's disease. Eur J Endocrinol. 2010;162(4):677-84.
  101. Lange M, Qvortrup K, Svendsen OL, Flyvbjerg A, Nowak J, Petersen MM, et al. Abnormal bone collagen morphology and decreased bone strength in growth hormone-deficient rats. Bone. 2004;35(1):178-85.
  102. Doessing S, Holm L, Heinemeier KM, Feldt-Rasmussen U, Schjerling P, Qvortrup K, et al. GH and IGF1 levels are positively associated with musculotendinous collagen expression: experiments in acromegalic and GH deficiency patients. Eur J Endocrinol. 2010;163(6):853-62.
  103. https://www.nice.org.uk/guidance/TA64?UNLID=10228138712014122893019?print=true. Human growth hormone (somatropin) in adults with growth hormone deficiency. 2017.
  104. Chikani V, Cuneo RC, Hickman I, Ho KK. Impairment of anaerobic capacity in adults with growth hormone deficiency. J Clin Endocrinol Metab. 2015;100(5):1811-8.
  105. Herschbach P, Henrich G, Strasburger CJ, Feldmeier H, Marín F, Attanasio AM, et al. Development and psychometric properties of a disease-specific quality of life questionnaire for adult patients with growth hormone deficiency. Eur J Endocrinol. 2001;145(3):255-65.
  106. Gilchrist FJ, Murray RD, Shalet SM. The effect of long-term untreated growth hormone deficiency (GHD) and 9 years of GH replacement on the quality of life (QoL) of GH-deficient adults. Clin Endocrinol (Oxf). 2002;57(3):363-70.
  107. McKenna SP, Doward LC, Alonso J, Kohlmann T, Niero M, Prieto L, et al. The QoL-AGHDA: an instrument for the assessment of quality of life in adults with growth hormone deficiency. QualLife Res. 1999;8(4):373-83.
  108. Wiren L, Whalley D, McKenna S, Wilhelmsen L. Application of a disease-specific, quality-of-life measure (QoL-AGHDA) in growth hormone-deficient adults and a random population sample in Sweden: validation of the measure by rasch analysis. Clinical Endocrinology. 2000;52(2):143-52.
  109. Koltowska-Haggstrom M, Hennessy S, Mattsson AF, Monson JP, Kind P. Quality of life assessment of growth hormone deficiency in adults (QoL-AGHDA): comparison of normative reference data for the general population of England and Wales with results for adult hypopituitary patients with growth hormone deficiency. HormRes. 2005;64(1):46-54.
  110. Webb SM. Measurements of quality of life in patients with growth hormone deficiency. Journal of endocrinological investigation. 2008;31(9 Suppl):52-5.
  111. Woodhouse LJ, Mukherjee A, Shalet SM, Ezzat S. The influence of growth hormone status on physical impairments, functional limitations, and health-related quality of life in adults. Endocrine reviews. 2006;27(3):287-317.
  112. Rosen T, Bengtsson BA. Premature mortality due to cardiovascular disease in hypopituitarism. Lancet. 1990;336(8710):285-8.
  113. Bülow B, Hagmar L, Mikoczy Z, Nordström CH, Erfurth EM. Increased cerebrovascular mortality in patients with hypopituitarism. Clin Endocrinol (Oxf). 1997;46(1):75-81.
  114. Tomlinson JW, Holden N, Hills RK, Wheatley K, Clayton RN, Bates AS, et al. Association between premature mortality and hypopituitarism. West Midlands Prospective Hypopituitary Study Group. Lancet. 2001;357(9254):425-31.
  115. Bates AS, Van't Hoff W, Jones PJ, Clayton RN. The effect of hypopituitarism on life expectancy. The Journal of Clinical Endocrinology and Metabolism. 1996;81(3):1169-72.
  116. Erfurth EM, Bülow B, Eskilsson J, Hagmar L. High incidence of cardiovascular disease and increased prevalence of cardiovascular risk factors in women with hypopituitarism not receiving growth hormone treatment: preliminary results. Growth hormone & IGF research : official journal of the Growth Hormone Research Society and the International IGF Research Society. 1999;9 Suppl A:21-4.
  117. Bengtsson BA. Untreated growth hormone deficiency explains premature mortality in patients with hypopituitarism. Growth hormone & IGF research : official journal of the Growth Hormone Research Society and the International IGF Research Society. 1998;8 Suppl A:77-80.
  118. Filipsson H, Monson JP, Koltowska-Haggstrom M, Mattsson A, Johannsson G. The impact of glucocorticoid replacement regimens on metabolic outcome and comorbidity in hypopituitary patients. J Clin Endocrinol Metab. 2006;91(10):3954-61.
  119. Ragnarsson O, Nyström HF, Johannsson G. Glucocorticoid replacement therapy is independently associated with reduced bone mineral density in women with hypopituitarism. Clin Endocrinol (Oxf). 2012;76(2):246-52.
  120. Filipsson Nyström H, Feldt-Rasmussen U, Kourides I, Popovic V, Koltowska-Häggström M, Jonsson B, et al. The metabolic consequences of thyroxine replacement in adult hypopituitary patients. Pituitary. 2012;15(4):495-504.
  121. Klose M, Marina D, Hartoft-Nielsen ML, Klefter O, Gavan V, Hilsted L, et al. Central hypothyroidism and its replacement have a significant influence on cardiovascular risk factors in adult hypopituitary patients. J Clin Endocrinol Metab. 2013;98(9):3802-10.
  122. Filipsson H, Johannsson G. GH replacement in adults: interactions with other pituitary hormone deficiencies and replacement therapies. Eur J Endocrinol. 2009;161 Suppl 1:S85-95.
  123. Feldt-Rasmussen U. Interactions between growth hormone and the thyroid gland -- with special reference to biochemical diagnosis. Current medicinal chemistry. 2007;14(26):2783-8.
  124. Olivius C, Landin-Wilhelmsen K, Olsson DS, Johannsson G, Tivesten Å. Prevalence and treatment of central hypogonadism and hypoandrogenism in women with hypopituitarism. Pituitary. 2018;21(5):445-53.
  125. Sanmartí A, Lucas A, Hawkins F, Webb SM, Ulied A. Observational study in adult hypopituitary patients with untreated growth hormone deficiency (ODA study). Socio-economic impact and health status. Collaborative ODA (Observational GH Deficiency in Adults) Group. Eur J Endocrinol. 1999;141(5):481-9.
  126. Clemmons DR. Consensus statement on the standardization and evaluation of growth hormone and insulin-like growth factor assays. ClinChem. 2011;57(4):555-9.
  127. Garcia JM, Biller BMK, Korbonits M, Popovic V, Luger A, Strasburger CJ, et al. Sensitivity and specificity of the macimorelin test for diagnosis of AGHD. Endocrine connections. 2021;10(1):76-83.
  128. Roth J, Glick SM, Yalow RS, Bersonsa. Hypoglycemia: a potent stimulus to secretion of growth hormone. Science (New York, NY). 1963;140(3570):987-8.
  129. Hoffman DM, O'Sullivan AJ, Baxter RC, Ho KK. Diagnosis of growth-hormone deficiency in adults. Lancet. 1994;343(8905):1064-8.
  130. Hoeck HC, Vestergaard P, Jakobsen PE, Laurberg P. Test of growth hormone secretion in adults: poor reproducibility of the insulin tolerance test. Eur J Endocrinol. 1995;133(3):305-12.
  131. Fisker S, Jorgensen JO, Orskov H, Christiansen JS. L-arginine and insulin-tolerance tests in the diagnosis of adult growth hormone deficiency: influence of confounding factors. Clinical Endocrinology. 1998;48(1):109-15.
  132. Shah A, Stanhope R, Matthew D. Hazards of pharmacological tests of growth hormone secretion in childhood. Bmj. 1992;304(6820):173-4.
  133. Lange M, Svendsen OL, Skakkebaek NE, Muller J, Juul A, Schmiegelow M, et al. An audit of the insulin-tolerance test in 255 patients with pituitary disease. European Journal of Endocrinology. 2002;147(1):41-7.
  134. Andersen M. The robustness of diagnostic tests for GH deficiency in adults. Growth hormone & IGF research : official journal of the Growth Hormone Research Society and the International IGF Research Society. 2015;25(3):108-14.
  135. Corneli G, Di Somma C, Baldelli R, Rovere S, Gasco V, Croce CG, et al. The cut-off limits of the GH response to GH-releasing hormone-arginine test related to body mass index. European Journal of Endocrinology. 2005;153(2):257-64.
  136. Aimaretti G, Corneli G, Razzore P, Bellone S, Baffoni C, Arvat E, et al. Comparison between insulin-induced hypoglycemia and growth hormone (GH)-releasing hormone + arginine as provocative tests for the diagnosis of GH deficiency in adults. The Journal of Clinical Endocrinology and Metabolism. 1998;83(5):1615-8.
  137. Abs R. Update on the diagnosis of GH deficiency in adults. Eur J Endocrinol. 2003;148 Suppl 2:S3-8.
  138. Yuen KC, Biller BM, Molitch ME, Cook DM. Clinical review: Is lack of recombinant growth hormone (GH)-releasing hormone in the United States a setback or time to consider glucagon testing for adult GH deficiency? J Clin Endocrinol Metab. 2009;94(8):2702-7.
  139. Diri H, Karaca Z, Simsek Y, Tanriverdi F, Unluhizarci K, Selcuklu A, et al. Can a glucagon stimulation test characterized by lower GH cut-off value be used for the diagnosis of growth hormone deficiency in adults? Pituitary. 2015;18(6):884-92.
  140. Hamrahian AH, Yuen KC, Gordon MB, Pulaski-Liebert KJ, Bena J, Biller BM. Revised GH and cortisol cut-points for the glucagon stimulation test in the evaluation of GH and hypothalamic-pituitary-adrenal axes in adults: results from a prospective randomized multicenter study. Pituitary. 2016;19(3):332-41.
  141. Tavares AB, Seixas-da-Silva IA, Silvestre DH, Paixão CM, Jr., Vaisman M, Conceição FL. Potential risks of glucagon stimulation test in elderly people. Growth hormone & IGF research : official journal of the Growth Hormone Research Society and the International IGF Research Society. 2015;25(1):53-6.
  142. Toogood AA, O'Neill PA, Shalet SM. Beyond the somatopause: growth hormone deficiency in adults over the age of 60 years. J Clin Endocrinol Metab. 1996;81(2):460-5.
  143. Yuen KC, Biller BM, Katznelson L, Rhoads SA, Gurel MH, Chu O, et al. Clinical characteristics, timing of peak responses and safety aspects of two dosing regimens of the glucagon stimulation test in evaluating growth hormone and cortisol secretion in adults. Pituitary. 2013;16(2):220-30.
  144. Micmacher E, Assumpção RP, Redorat RG, Spina LD, Cruz IC, Silva CA, et al. Growth hormone secretion in response to glucagon stimulation test in healthy middle-aged men. Arquivos brasileiros de endocrinologia e metabologia. 2009;53(7):853-8.
  145. Wilson JR, Utz AL, Devin JK. Effects of gender, body weight, and blood glucose dynamics on the growth hormone response to the glucagon stimulation test in patients with pituitary disease. Growth hormone & IGF research : official journal of the Growth Hormone Research Society and the International IGF Research Society. 2016;26:24-31.
  146. Biller BM, Samuels MH, Zagar A, Cook DM, Arafah BM, Bonert V, et al. Sensitivity and specificity of six tests for the diagnosis of adult GH deficiency. The Journal of Clinical Endocrinology and Metabolism. 2002;87(5):2067-79.
  147. Junnila RK, Strasburger CJ, Bidlingmaier M. Pitfalls of insulin-like growth factor-i and growth hormone assays. Endocrinology and metabolism clinics of North America. 2015;44(1):27-34.
  148. Pokrajac A, Wark G, Ellis AR, Wear J, Wieringa GE, Trainer PJ. Variation in GH and IGF-I assays limits the applicability of international consensus criteria to local practice. Clin Endocrinol (Oxf). 2007;67(1):65-70.
  149. Toogood A, Brabant G, Maiter D, Jonsson B, Feldt-Rasmussen U, Koltowska-Haggstrom M, et al. Similar clinical features among patients with severe adult growth hormone deficiency diagnosed with insulin tolerance test or arginine or glucagon stimulation tests. Endocrine practice : official journal of the American College of Endocrinology and the American Association of Clinical Endocrinologists. 2012;18(3):325-34.
  150. Andersen M, Støving RK, Hangaard J, Petersen PH, Hagen C. The effect of short-term cortisol changes on growth hormone responses to the pyridostigmine-growth-hormone-releasing-hormone test in healthy adults and patients with suspected growth hormone deficiency. Clin Endocrinol (Oxf). 1998;49(2):241-9.
  151. Dichtel LE, Yuen KC, Bredella MA, Gerweck AV, Russell BM, Riccio AD, et al. Overweight/Obese adults with pituitary disorders require lower peak growth hormone cutoff values on glucagon stimulation testing to avoid overdiagnosis of growth hormone deficiency. The Journal of Clinical Endocrinology and Metabolism. 2014;99(12):4712-9.
  152. Toogood AA, Beardwell CG, Shalet SM. The severity of growth hormone deficiency in adults with pituitary disease is related to the degree of hypopituitarism. Clinical Endocrinology. 1994;41(4):511-6.
  153. Aimaretti G, Corneli G, Di Somma C, Baldelli R, Gasco V, Rovere S, et al. Different degrees of GH deficiency evidenced by GHRH+arginine test and IGF-I levels in adults with pituritary disease. JEndocrinolInvest. 2005;28(3):247-52.
  154. Gordon MB, Levy RA, Gut R, Germak J. TRENDS IN GROWTH HORMONE STIMULATION TESTING AND GROWTH HORMONE DOSING IN ADULT GROWTH HORMONE DEFICIENCY PATIENTS: RESULTS FROM THE ANSWER PROGRAM. Endocrine practice : official journal of the American College of Endocrinology and the American Association of Clinical Endocrinologists. 2016;22(4):396-405.
  155. Sonksen PH. In: Adashi EY, Thorner MO, editors. The somatotrophic axis and the reproductive process in health and disease. New York: Springer-Verlag; 1995.
  156. Schneider HJ, Kreitschmann-Andermahr I, Ghigo E, Stalla GK, Agha A. Hypothalamopituitary dysfunction following traumatic brain injury and aneurysmal subarachnoid hemorrhage: a systematic review. JAMA. 2007;298(12):1429-38.
  157. Garrahy A, Sherlock M, Thompson CJ. MANAGEMENT OF ENDOCRINE DISEASE: Neuroendocrine surveillance and management of neurosurgical patients. Eur J Endocrinol. 2017;176(5):R217-r33.
  158. Murray RD, Skillicorn CJ, Howell SJ, Lissett CA, Rahim A, Shalet SM. Dose titration and patient selection increases the efficacy of GH replacement in severely GH deficient adults. Clin Endocrinol (Oxf). 1999;50(6):749-57.
  159. Wirén L, Bengtsson BA, Johannsson G. Beneficial effects of long-term GH replacement therapy on quality of life in adults with GH deficiency. Clin Endocrinol (Oxf). 1998;48(5):613-20.
  160. Gibney J, Wallace JD, Spinks T, Schnorr L, Ranicar A, Cuneo RC, et al. The effects of 10 years of recombinant human growth hormone (GH) in adult GH-deficient patients. J Clin Endocrinol Metab. 1999;84(8):2596-602.
  161. Koltowska-Haggstrom M, Mattsson AF, Monson JP, Kind P, Badia X, Casanueva FF, et al. Does long-term GH replacement therapy in hypopituitary adults with GH deficiency normalise quality of life? European Journal of Endocrinology. 2006;155(1):109-19.
  162. Miller KK, Wexler T, Fazeli P, Gunnell L, Graham GJ, Beauregard C, et al. Growth hormone deficiency after treatment of acromegaly: a randomized, placebo-controlled study of growth hormone replacement. J Clin Endocrinol Metab. 2010;95(2):567-77.
  163. Valassi E, Brick DJ, Johnson JC, Biller BM, Klibanski A, Miller KK. Effect of growth hormone replacement therapy on the quality of life in women with growth hormone deficiency who have a history of acromegaly versus other disorders. Endocrine practice : official journal of the American College of Endocrinology and the American Association of Clinical Endocrinologists. 2012;18(2):209-18.
  164. Klose M, Rasmussen AK, Feldt-Rasmussen U. Quality of Life in adult hypopituitary patiens treated for growth hormone deficiency. The Open Endocrinology Journal. 2012;6:91-102.
  165. Baum HB, Katznelson L, Sherman JC, Biller BM, Hayden DL, Schoenfeld DA, et al. Effects of physiological growth hormone (GH) therapy on cognition and quality of life in patients with adult-onset GH deficiency. The Journal of Clinical Endocrinology and Metabolism. 1998;83(9):3184-9.
  166. Nyberg F, Burman P. Growth hormone and its receptors in the central nervous system--location and functional significance. Hormone research. 1996;45(1-2):18-22.
  167. Burman P, Hetta J, Wide L, Mansson JE, Ekman R, Karlsson FA. Growth hormone treatment affects brain neurotransmitters and thyroxine [see comment]. Clinical Endocrinology. 1996;44(3):319-24.
  168. Nyberg F. Aging effects on growth hormone receptor binding in the brain. Experimental gerontology. 1997;32(4-5):521-8.
  169. Lai Z, Roos P, Zhai O, Olsson Y, Fhölenhag K, Larsson C, et al. Age-related reduction of human growth hormone-binding sites in the human brain. Brain research. 1993;621(2):260-6.
  170. Chikani V, Cuneo RC, Hickman I, Ho KK. Growth hormone (GH) enhances anaerobic capacity: impact on physical function and quality of life in adults with GH deficiency. Clin Endocrinol (Oxf). 2016;85(4):660-8.
  171. Hull KL, Harvey S. Growth hormone therapy and Quality of Life: possibilities, pitfalls and mechanisms. The Journal of endocrinology. 2003;179(3):311-33.
  172. Hazem A, Elamin MB, Bancos I, Malaga G, Prutsky G, Domecq JP, et al. Body composition and quality of life in adults treated with GH therapy: a systematic review and meta-analysis. Eur J Endocrinol. 2012;166(1):13-20.
  173. Drake WM, Coyte D, Camacho-Hübner C, Jivanji NM, Kaltsas G, Wood DF, et al. Optimizing growth hormone replacement therapy by dose titration in hypopituitary adults. J Clin Endocrinol Metab. 1998;83(11):3913-9.
  174. Jørgensen JO, Pedersen SA, Laurberg P, Weeke J, Skakkebaek NE, Christiansen JS. Effects of growth hormone therapy on thyroid function of growth hormone-deficient adults with and without concomitant thyroxine-substituted central hypothyroidism. J Clin Endocrinol Metab. 1989;69(6):1127-32.
  175. Florakis D, Hung V, Kaltsas G, Coyte D, Jenkins PJ, Chew SL, et al. Sustained reduction in circulating cholesterol in adult hypopituitary patients given low dose titrated growth hormone replacement therapy: a two year study. Clin Endocrinol (Oxf). 2000;53(4):453-9.
  176. Abrahamsen B, Nielsen TL, Hangaard J, Gregersen G, Vahl N, Korsholm L, et al. Dose-, IGF-I- and sex-dependent changes in lipid profile and body composition during GH replacement therapy in adult onset GH deficiency. Eur J Endocrinol. 2004;150(5):671-9.
  177. Lind S, Rudling M, Ericsson S, Olivecrona H, Eriksson M, Borgström B, et al. Growth hormone induces low-density lipoprotein clearance but not bile acid synthesis in humans. Arteriosclerosis, thrombosis, and vascular biology. 2004;24(2):349-56.
  178. Attanasio AF, Bates PC, Ho KK, Webb SM, Ross RJ, Strasburger CJ, et al. Human growth hormone replacement in adult hypopituitary patients: long-term effects on body composition and lipid status--3-year results from the HypoCCS Database. J Clin Endocrinol Metab. 2002;87(4):1600-6.
  179. Höybye C, Christiansen JS. Growth hormone replacement in adults - current standards and new perspectives. Best practice & research Clinical endocrinology & metabolism. 2015;29(1):115-23.
  180. van Bunderen CC, van den Dries CJ, Heymans MW, Franken AA, Koppeschaar HP, van der Lely AJ, et al. Effect of long-term GH replacement therapy on cardiovascular outcomes in isolated GH deficiency compared with multiple pituitary hormone deficiencies: a sub-analysis from the Dutch National Registry of Growth Hormone Treatment in Adults. Eur J Endocrinol. 2014;171(2):151-60.
  181. Kreitschmann-Andermahr I, Siegel S, Francis F, Buchfelder M, Schneider HJ, Kann PH, et al. Variation of the baseline characteristics and treatment parameters over time: an analysis of 15 years of growth hormone replacement in adults in the German KIMS database. Pituitary. 2012;15 Suppl 1:S72-80.
  182. Abrams P, Boquete H, Fideleff H, Feldt-Rasmussen U, Jönsson PJ, Koltowska-Häggström M, et al. GH replacement in hypopituitarism improves lipid profile and quality of life independently of changes in obesity variables. Eur J Endocrinol. 2008;159(6):825-32.
  183. Olivecrona H, Ericsson S, Berglund L, Angelin B. Increased concentrations of serum lipoprotein (a) in response to growth hormone treatment. Bmj. 1993;306(6894):1726-7.
  184. Svensson J, Jansson JO, Ottosson M, Johannsson G, Taskinen MR, Wiklund O, et al. Treatment of obese subjects with the oral growth hormone secretagogue MK-677 affects serum concentrations of several lipoproteins, but not lipoprotein(a). J Clin Endocrinol Metab. 1999;84(6):2028-33.
  185. Wieringa G, Toogood AA, Ryder WD, Anderson JM, Mackness M, Shalet SM. Changes in lipoprotein(a) levels measured by six kit methods during growth hormone treatment of growth hormone-deficient adults. Growth hormone & IGF research : official journal of the Growth Hormone Research Society and the International IGF Research Society. 2000;10(1):14-9.
  186. Weber MM, Biller BM, Pedersen BT, Pournara E, Christiansen JS, Höybye C. The effect of growth hormone (GH) replacement on blood glucose homeostasis in adult nondiabetic patients with GH deficiency: real-life data from the NordiNet(®) International Outcome Study. Clin Endocrinol (Oxf). 2017;86(2):192-8.
  187. Bollerslev J, Hallén J, Fougner KJ, Jørgensen AP, Kristo C, Fagertun H, et al. Low-dose GH improves exercise capacity in adults with GH deficiency: effects of a 22-month placebo-controlled, crossover trial. Eur J Endocrinol. 2005;153(3):379-87.
  188. Widdowson WM, Gibney J. The effect of growth hormone (GH) replacement on muscle strength in patients with GH-deficiency: a meta-analysis. Clin Endocrinol (Oxf). 2010;72(6):787-92.
  189. Sneppen SB, Hoeck HC, Kollerup G, Sørensen OH, Laurberg P, Feldt-Rasmussen U. Bone mineral content and bone metabolism during physiological GH treatment in GH-deficient adults--an 18-month randomised, placebo-controlled, double blinded trial. Eur J Endocrinol. 2002;146(2):187-95.
  190. Johannsson G, Rosén T, Bosaeus I, Sjöström L, Bengtsson BA. Two years of growth hormone (GH) treatment increases bone mineral content and density in hypopituitary patients with adult-onset GH deficiency. J Clin Endocrinol Metab. 1996;81(8):2865-73.
  191. Holmes SJ, Whitehouse RW, Swindell R, Economou G, Adams JE, Shalet SM. Effect of growth hormone replacement on bone mass in adults with adult onset growth hormone deficiency. Clin Endocrinol (Oxf). 1995;42(6):627-33.
  192. Barake M, Klibanski A, Tritos NA. Effects of recombinant human growth hormone therapy on bone mineral density in adults with growth hormone deficiency: a meta-analysis. J Clin Endocrinol Metab. 2014;99(3):852-60.
  193. Rahim A, Holmes SJ, Adams JE, Shalet SM. Long-term change in the bone mineral density of adults with adult onset growth hormone (GH) deficiency in response to short or long-term GH replacement therapy. Clin Endocrinol (Oxf). 1998;48(4):463-9.
  194. Davidson P, Milne R, Chase D, Cooper C. Growth hormone replacement in adults and bone mineral density: a systematic review and meta-analysis. Clin Endocrinol (Oxf). 2004;60(1):92-8.
  195. Clanget C, Seck T, Hinke V, Wüster C, Ziegler R, Pfeilschifter J. Effects of 6 years of growth hormone (GH) treatment on bone mineral density in GH-deficient adults. Clin Endocrinol (Oxf). 2001;55(1):93-9.
  196. Appelman-Dijkstra NM, Claessen KM, Hamdy NA, Pereira AM, Biermasz NR. Effects of up to 15 years of recombinant human GH (rhGH) replacement on bone metabolism in adults with growth hormone deficiency (GHD): the Leiden Cohort Study. Clin Endocrinol (Oxf). 2014;81(5):727-35.
  197. Tritos NA, Hamrahian AH, King D, Greenspan SL, Cook DM, Jönsson PJ, et al. Predictors of the effects of 4 years of growth hormone replacement on bone mineral density in patients with adult-onset growth hormone deficiency - a KIMS database analysis. Clin Endocrinol (Oxf). 2013;79(2):178-84.
  198. Holmer H, Svensson J, Rylander L, Johannsson G, Rosén T, Bengtsson BA, et al. Fracture incidence in GH-deficient patients on complete hormone replacement including GH. Journal of bone and mineral research : the official journal of the American Society for Bone and Mineral Research. 2007;22(12):1842-50.
  199. Mo D, Fleseriu M, Qi R, Jia N, Child CJ, Bouillon R, et al. Fracture risk in adult patients treated with growth hormone replacement therapy for growth hormone deficiency: a prospective observational cohort study. The lancet Diabetes & endocrinology. 2015;3(5):331-8.
  200. Klefter O, Feldt-Rasmussen U. Is increase in bone mineral content caused by increase in skeletal muscle mass/strength in adult patients with GH-treated GH deficiency? A systematic literature analysis. Eur J Endocrinol. 2009;161(2):213-21.
  201. Monson JP, Abs R, Bengtsson BA, Bennmarker H, Feldt-Rasmussen U, Hernberg-Stâhl E, et al. Growth hormone deficiency and replacement in elderly hypopituitary adults. KIMS Study Group and the KIMS International Board. Pharmacia and Upjohn International Metabolic Database. Clin Endocrinol (Oxf). 2000;53(3):281-9.
  202. Fernholm R, Bramnert M, Hägg E, Hilding A, Baylink DJ, Mohan S, et al. Growth hormone replacement therapy improves body composition and increases bone metabolism in elderly patients with pituitary disease. J Clin Endocrinol Metab. 2000;85(11):4104-12.
  203. Kokshoorn NE, Biermasz NR, Roelfsema F, Smit JW, Pereira AM, Romijn JA. GH replacement therapy in elderly GH-deficient patients: a systematic review. Eur J Endocrinol. 2011;164(5):657-65.
  204. Vahl N, Juul A, Jørgensen JO, Orskov H, Skakkebaek NE, Christiansen JS. Continuation of growth hormone (GH) replacement in GH-deficient patients during transition from childhood to adulthood: a two-year placebo-controlled study. J Clin Endocrinol Metab. 2000;85(5):1874-81.
  205. Carroll PV, Drake WM, Maher KT, Metcalfe K, Shaw NJ, Dunger DB, et al. Comparison of continuation or cessation of growth hormone (GH) therapy on body composition and metabolic status in adolescents with severe GH deficiency at completion of linear growth. J Clin Endocrinol Metab. 2004;89(8):3890-5.
  206. Drake WM, Carroll PV, Maher KT, Metcalfe KA, Camacho-Hübner C, Shaw NJ, et al. The effect of cessation of growth hormone (GH) therapy on bone mineral accretion in GH-deficient adolescents at the completion of linear growth. J Clin Endocrinol Metab. 2003;88(4):1658-63.
  207. Courtillot C, Baudoin R, Du Souich T, Saatdjian L, Tejedor I, Pinto G, et al. Monocentric study of 112 consecutive patients with childhood onset GH deficiency around and after transition. Eur J Endocrinol. 2013;169(5):587-96.
  208. Clayton PE, Cuneo RC, Juul A, Monson JP, Shalet SM, Tauber M. Consensus statement on the management of the GH-treated adolescent in the transition to adult care. Eur J Endocrinol. 2005;152(2):165-70.
  209. Toogood AA, Taylor NF, Shalet SM, Monson JP. Modulation of cortisol metabolism by low-dose growth hormone replacement in elderly hypopituitary patients. J Clin Endocrinol Metab. 2000;85(4):1727-30.
  210. Agha A, Walker D, Perry L, Drake WM, Chew SL, Jenkins PJ, et al. Unmasking of central hypothyroidism following growth hormone replacement in adult hypopituitary patients. Clin Endocrinol (Oxf). 2007;66(1):72-7.
  211. Jørgensen JO, Møller J, Laursen T, Orskov H, Christiansen JS, Weeke J. Growth hormone administration stimulates energy expenditure and extrathyroidal conversion of thyroxine to triiodothyronine in a dose-dependent manner and suppresses circadian thyrotrophin levels: studies in GH-deficient adults. Clin Endocrinol (Oxf). 1994;41(5):609-14.
  212. Jørgensen JO, Møller J, Skakkebaek NE, Weeke J, Christiansen JS. Thyroid function during growth hormone therapy. Hormone research. 1992;38 Suppl 1:63-7.
  213. Laurberg P, Jakobsen PE, Hoeck HC, Vestergaard P. Growth hormone and thyroid function: is secondary thyroid failure underdiagnosed in growth hormone deficient patients? Thyroidology. 1994;6(3):73-9.
  214. Span JP, Pieters GF, Sweep CG, Hermus AR, Smals AG. Gender difference in insulin-like growth factor I response to growth hormone (GH) treatment in GH-deficient adults: role of sex hormone replacement. J Clin Endocrinol Metab. 2000;85(3):1121-5.
  215. Wolthers T, Hoffman DM, Nugent AG, Duncan MW, Umpleby M, Ho KK. Oral estrogen antagonizes the metabolic actions of growth hormone in growth hormone-deficient women. American journal of physiology Endocrinology and metabolism. 2001;281(6):E1191-6.
  216. Birzniece V, Ho KKY. Sex steroids and the GH axis: Implications for the management of hypopituitarism. Best practice & research Clinical endocrinology & metabolism. 2017;31(1):59-69.
  217. Christiansen JJ, Fisker S, Gravholt CH, Bennett P, Svenstrup B, Andersen M, et al. Discontinuation of estrogen replacement therapy in GH-treated hypopituitary women alters androgen status and IGF-I. Eur J Endocrinol. 2005;152(5):719-26.
  218. Weissberger AJ, Ho KK, Lazarus L. Contrasting effects of oral and transdermal routes of estrogen replacement therapy on 24-hour growth hormone (GH) secretion, insulin-like growth factor I, and GH-binding protein in postmenopausal women. The Journal of Clinical Endocrinology and Metabolism. 1991;72(2):374-81.
  219. Brooke AM, Kalingag LA, Miraki-Moud F, Camacho-Hübner C, Maher KT, Walker DM, et al. Dehydroepiandrosterone (DHEA) replacement reduces growth hormone (GH) dose requirement in female hypopituitary patients on GH replacement. Clin Endocrinol (Oxf). 2006;65(5):673-80.
  220. Arlt W, Callies F, van Vlijmen JC, Koehler I, Reincke M, Bidlingmaier M, et al. Dehydroepiandrosterone replacement in women with adrenal insufficiency. N Engl J Med. 1999;341(14):1013-20.
  221. van Thiel SW, Romijn JA, Pereira AM, Biermasz NR, Roelfsema F, van Hemert A, et al. Effects of dehydroepiandrostenedione, superimposed on growth hormone substitution, on quality of life and insulin-like growth factor I in patients with secondary adrenal insufficiency: a randomized, placebo-controlled, cross-over trial. J Clin Endocrinol Metab. 2005;90(6):3295-303.
  222. Feldt-Rasmussen U, Brabant G, Maiter D, Jonsson B, Toogood A, Koltowska-Haggstrom M, et al. Response to GH treatment in adult GH deficiency is predicted by gender, age, and IGF1 SDS but not by stimulated GH-peak. Eur J Endocrinol. 2013;168(5):733-43.
  223. Mah PM, Webster J, Jönsson P, Feldt-Rasmussen U, Koltowska-Häggström M, Ross RJ. Estrogen replacement in women of fertile years with hypopituitarism. J Clin Endocrinol Metab. 2005;90(11):5964-9.
  224. Ho KKY, Gibney J, Johannsson G, Wolthers T. Regulating of growth hormone sensitivity by sex steroids: implications for therapy. Frontiers of hormone research. 2006;35:115-28.
  225. Fleseriu M, Hashim IA, Karavitaki N, Melmed S, Murad MH, Salvatori R, et al. Hormonal Replacement in Hypopituitarism in Adults: An Endocrine Society Clinical Practice Guideline. J Clin Endocrinol Metab. 2016;101(11):3888-921.
  226. Johannsson G, Gordon MB, Højby Rasmussen M, Håkonsson IH, Karges W, Sværke C, et al. Once-weekly Somapacitan is Effective and Well Tolerated in Adults with GH Deficiency: A Randomized Phase 3 Trial. J Clin Endocrinol Metab. 2020;105(4):e1358-76.
  227. Sävendahl L, Battelino T, Højby Rasmussen M, Brod M, Saenger P, Horikawa R. Effective GH Replacement With Once-weekly Somapacitan vs Daily GH in Children with GHD: 3-year Results From REAL 3. J Clin Endocrinol Metab. 2022;107(5):1357-67.
  228. Maniatis AK, Casella SJ, Nadgir UM, Hofman PL, Saenger P, Chertock ED, et al. Safety and Efficacy of Lonapegsomatropin in Children with Growth Hormone Deficiency: enliGHten Trial 2-Year Results. J Clin Endocrinol Metab. 2022.
  229. Deal C, Steelman J, Vlachopapadopoulou E, Stawerska R, Silverman LA, Phillip M, et al. Efficacy and safety of weekly somatrogon vs daily somatropin in children with growth hormone deficiency: a phase 3 study. J Clin Endocrinol Metab. 2022.
  230. Du H, Wu D, Yi P, Bai X, Luo Y, Yang H, et al. Evaluation of efficacy and safety of long-acting PEGylated recombinant human growth hormone (Jintrolong) for patients with growth hormone deficiency. Journal of pediatric endocrinology & metabolism : JPEM. 2022;35(4):511-7.
  231. Yuen KCJ, Miller BS, Boguszewski CL, Hoffman AR. Usefulness and Potential Pitfalls of Long-Acting Growth Hormone Analogs. Frontiers in endocrinology. 2021;12:637209.
  232. Cruz-Topete D, Jorgensen JO, Christensen B, Sackmann-Sala L, Krusenstjerna-Hafstrøm T, Jara A, et al. Identification of new biomarkers of low-dose GH replacement therapy in GH-deficient patients. J Clin Endocrinol Metab. 2011;96(7):2089-97.
  233. Frajese G, Drake WM, Loureiro RA, Evanson J, Coyte D, Wood DF, et al. Hypothalamo-pituitary surveillance imaging in hypopituitary patients receiving long-term GH replacement therapy. J Clin Endocrinol Metab. 2001;86(11):5172-5.
  234. Chung TT, Drake WM, Evanson J, Walker D, Plowman PN, Chew SL, et al. Tumour surveillance imaging in patients with extrapituitary tumours receiving growth hormone replacement. Clin Endocrinol (Oxf). 2005;63(3):274-9.
  235. Robison LL, Armstrong GT, Boice JD, Chow EJ, Davies SM, Donaldson SS, et al. The Childhood Cancer Survivor Study: a National Cancer Institute-supported resource for outcome and intervention research. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2009;27(14):2308-18.
  236. 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(9):2756-61.
  237. Popovic V, Mattsson AF, Gaillard RC, Wilton P, Koltowska-Häggström M, Ranke MB. Serum insulin-like growth factor I (IGF-I), IGF-binding proteins 2 and 3, and the risk for development of malignancies in adults with growth hormone (GH) deficiency treated with GH: data from KIMS (Pfizer International Metabolic Database). J Clin Endocrinol Metab. 2010;95(9):4449-54.
  238. van Bunderen CC, van Nieuwpoort IC, Arwert LI, Heymans MW, Franken AA, Koppeschaar HP, et al. Does growth hormone replacement therapy reduce mortality in adults with growth hormone deficiency? Data from the Dutch National Registry of Growth Hormone Treatment in adults. J Clin Endocrinol Metab. 2011;96(10):3151-9.
  239. Berglund A, Gravholt CH, Olsen MS, Christiansen JS, Stochholm K. Growth hormone replacement does not increase mortality in patients with childhood-onset growth hormone deficiency. Clin Endocrinol (Oxf). 2015;83(5):677-83.
  240. Stochholm K, Gravholt CH, Laursen T, Laurberg P, Andersen M, Kristensen L, et al. Mortality and GH deficiency: a nationwide study. Eur J Endocrinol. 2007;157(1):9-18.
  241. Stochholm K, Juul S, Christiansen JS, Gravholt CH. Mortality and socioeconomic status in adults with childhood onset GH deficiency (GHD) is highly dependent on the primary cause of GHD. Eur J Endocrinol. 2012;167(5):663-70.
  242. Burman P, Mattsson AF, Johannsson G, Höybye C, Holmer H, Dahlqvist P, et al. Deaths among adult patients with hypopituitarism: hypocortisolism during acute stress, and de novo malignant brain tumors contribute to an increased mortality. J Clin Endocrinol Metab. 2013;98(4):1466-75.
  243. Erfurth EM. Update in mortality in GH-treated patients. J Clin Endocrinol Metab. 2013;98(11):4219-26.
  244. Carel JC, Ecosse E, Landier F, Meguellati-Hakkas D, Kaguelidou F, Rey G, et al. 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(2):416-25.
  245. Hernberg-Ståhl E, Luger A, Abs R, Bengtsson BA, Feldt-Rasmussen U, Wilton P, et al. Healthcare consumption decreases in parallel with improvements in quality of life during GH replacement in hypopituitary adults with GH deficiency. J Clin Endocrinol Metab. 2001;86(11):5277-81.
  246. Martel-Duguech LM, Jorgensen JOL, Korbonits M, Johannsson G, Webb SM, Amadidou F, et al. ESE audit on management of Adult Growth Hormone Deficiency in clinical practice. Eur J Endocrinol. 2020.
  247. Yuen KCJ. Adult growth hormone deficiency guidelines: more difficult than it seems to incorporate into clinical practice universally. Eur J Endocrinol. 2021;184(4):C5-c7.
  248. Mancini A, Bruno C, Vergani E, Brunetti A, Palmisano G, Pontecorvi A. "Non-Classical" Indication for Provocative Testing of Growth Hormone: A Retrospective Cohort Study in Adult Patients Under Replacement Therapy. Endocrine, metabolic & immune disorders drug targets. 2021;21(8):1406-12.

 

Bacterial Infections in Endocrinology

ABSTRACT

 

Bacteria are microscopic organisms that are ubiquitous in the environment and human body. Some bacteria exhibit symbiotic relationship with the human body, while other bacteria are harmful and cause various diseases. Bacteria may infect the endocrine glands either by direct invasion or local or hematogenous spread. Suppurative bacterial infections can involve the pituitary, thyroid, adrenals, and gonads. In the majority of cases, specific risk factors predispose the endocrine glands to such infections. This in turn may lead to temporary or permanent endocrine dysfunction. There may also be states of hormone excess following bacterial infections. This is particularly noted in cases of bacterial thyroiditis. Permanent endocrine dysfunction following bacterial infections will warrant life-long hormone replacement therapy. In acute stages of infection, intravenous or oral antibiotics are the cornerstone of management. The choice of antibiotic is guided by culture and sensitivity report. Sometimes, however, empirical antibiotic therapy may need to be continued as no organism may be isolated on culture. Empirical therapy should provide coverage for gram positive, gram negative, and anaerobic bacteria. If there is abscess formation in any endocrine gland, it may require aspiration and drainage. In this chapter, we have discussed the risk factors, bacteriology, clinical presentation, diagnosis, and management of common bacterial infections involving endocrine glands.

INTRODUCTION

 

The incidence of bacterial infections of endocrine glands is low when compared to that in other organs of the body. The endocrine glands that may be affected by bacterial infections are: pituitary, thyroid, adrenals and gonads. Bacterial infection of parathyroid glands is extremely rare. Certain risk factors may predispose the glands for infection.

 

In general, bacteria may be classified as gram positive, gram negative, and miscellaneous categories. The classification of medically important bacteria is highlighted in another chapter of the Endotext (1). Among all the bacteria, Mycobacterium tuberculosis remains the most common agent involving the endocrine glands (2). Mycobacterium tuberculosis is a weakly gram positive highly aerobic bacterium that can cause tuberculosis in any organ of the body. This organism can affect the adrenal glands and lead to primary adrenal insufficiency. In developing countries, tuberculosis remains the most common cause of primary adrenal insufficiency. Tuberculosis can also affect pituitary, thyroid, and gonads. In this chapter, we are discussing only adrenal tuberculosis, since tuberculosis of the Endocrine system has been covered in great details in another chapter (3). Apart from Mycobacterium tuberculosis, the other common bacteria that may affect the endocrine system are Staphylococcus aureus, Streptococcus pneumoniae, Neisseria meningitides, Escherichia coli, Chlamydia trachomatis, Pseudomonas aeruginosa, Klebsiella pneumoniae, Treponema pallidum, and Yersinia enterocolitica among others. We have tried to present a spectrum of bacterial infections of various endocrine glands including their clinical presentation, investigations, management, long-term prognosis, and follow up.

 

BACTERIAL INFECTIONS OF PITUITARY

 

Infections of the pituitary gland are rare but may cause clinical problems because of the non-specific nature of the presentation (4). Among the various infectious agents, bacterial infections including Mycobacterial infections seem to be the most common. The various bacterial agents causing infection of the pituitary gland are summarized in the table 1. The common bacterial infections of the pituitary gland are described below.

 

Table 1.  Bacterial Agents Causing Infection of Pituitary-Hypothalamus

Bacterial class

Organism

Gram positive bacteria

Staphylococcus aureus, Streptococcus pneumoniae

Gram negative bacteria

E coli, Pseudomonas aeruginosa, Neisseria meningitides

Spirochaete

Treponema pallidum

Mycobacterium

Mycobacterium tuberculosis

 

Pituitary Abscess

 

EPIDEMIOLOGY AND RISK FACTORS

 

Pituitary abscesses are a very rare clinical entity and account for less than 1% of pituitary lesions (4). The first case of pituitary surgery involving an abscess was described in 1848. Since then, there have been around 300 such cases reported in the literature (4, 5). Risk factors include underlying pituitary diseases such as a pituitary adenoma, Rathke’s cyst, craniopharyngioma, lymphocytic hypophysitis, immunocompromised states (uncontrolled diabetes mellitus, tuberculosis, HIV infection, after solid organ transplantation, chemotherapy, radiotherapy), history of surgical exploration in pituitary hypothalamic region, and spread of local infection from meninges and paranasal sinuses (5-7). Rarely, abscess may develop in a normal pituitary gland (6, 8). 

 

BACTERIOLOGY

 

In the majority of cases, culture is negative in pituitary abscess, with only 19.7% cases showing growth of bacteria (9).The most common organisms isolated are Streptococci and Staphylococci. Other bacterial organisms are Escherichia coli, Mycobacteria, Neisseria, and anaerobes (6, 10). As culture is negative in most of the cases, it is important for empirical antibiotic therapy to cover gram positive, gram negative and anaerobic bacteria. Rarely, a fungal etiology is seen.

 

CLINICAL PRESENTATION

 

Clinical presentation can be classified with respect to chronicity as: acute (within days to weeks), subacute (less than a month) and chronic (more than a month). Acute and subacute abscesses have fulminant presentation while chronic abscess has a more indolent course (5). In the initial stages, patients present with headache (67%), fever, meningismus, and malaise. With progression of the disease, neurological symptoms like altered sensorium, seizures, and coma can occur.

 

Extension of infection in nearby areas can lead to visual dysfunction (45%), extra ocular movement defects, and other cranial nerve palsies (4, 8, 9, 11).

 

Both anterior and posterior pituitary hormonal hypofunction can be seen with a pituitary abscess. In the largest series of pituitary abscesses with 60 cases over 23 years, anterior pituitary hormone deficiencies were reported in 81.8% patients whereas diabetes insipidus was reported in 47.9% of the patients. In the same study, 9.3% had isolated hypogonadism, 3.7% had isolated ACTH deficiency, 1.8% had isolated hypothyroidism, and 1.8% hypothyroidism and ACTH deficiency (9).

 

DIAGNOSIS

 

The investigation of choice for the diagnosis of pituitary abscess is MRI (Magnetic Resonance Imaging) with proper sellar cuts. On T1 weighted images, pituitary abscess appears iso-intense to hypo-intense while on T2 weighted images, it is iso-intense to hyper-intense. There is a characteristic rim of enhancement after gadolinium injection around the abscess site (9, 11). Diffusion-weighted imaging (DWI) shows high signal intensity with a decrease in the apparent diffusion coefficient in the region of pus collection (9, 11).

 

MANAGEMENT

 

Trans-nasal trans-sphenoidal surgery and drainage of the abscess is the treatment of choice. The sphenoid sinus may require exploration if extrasellar invasion is suspected. Along with surgical exploration, the patient should be started on intravenous antibiotics empirically with ceftriaxone (alternatives are cefotaxime and cefepime) along with metronidazole for anaerobic coverage. In case of suspicion for Staphylococcus aureus, vancomycin should be added (9, 12). Further intensification or alteration of antibiotics is subjected to clinical improvement and culture and sensitivity reports. Microbiological etiology may not be identified in the majority of cases. Hence, it is imperative to give proper broad-spectrum coverage empirically.

 

PROGNOSIS AND FOLLOW-UP

 

With current standard of care, mortality rate is 10 % and chance of recurrence is <13%. In about 25% of cases hormonal recovery occurs. After recovering from a pituitary abscess, these patients should be followed up by serial MRI at 3, 6 and 12 months (12). Monitoring for anterior and posterior pituitary hormone deficiency should be done in any patient with a pituitary abscess. Replacement with corticosteroid, thyroid, gonadal, and growth hormone therapy may be required if the patient develops deficiency of any of these hormones. Replacement with vasopressin therapy may be required if patient develops central diabetes insipidus following a pituitary abscess.

 

Hypopituitarism Caused by Treponema Pallidum Infection

 

Syphilis caused by Treponema pallidum (a spirochete) may involve the pituitary- hypothalamic region causing syphilitic gumma with non-caseating granulomas (13, 14). It is more common in patients with underlying human immune deficiency virus (HIV) infection. Diagnosis can be made by demonstration of the spirochete in the samples of sellar tissues following trans sphenoidal surgery. Immunological diagnosis can be made by measuring titers of anti-Treponemal antibody in the serum. Treatment consists of intravenous followed by oral antibiotics (13-15). Penicillin is the drug of choice for syphilis. In patients who are allergic to penicillin, doxycycline is a good alternative.

 

BACTERIAL INFECTIONS OF THE THYROID

 

It is rare for bacteria to invade the normal thyroid gland because of the rich vascular supply, good lymphatic drainage, separation of thyroid gland from other structures by fascial planes, high iodine content, and production of hydrogen peroxide inside the gland (16). Both iodine and hydrogen peroxide have bactericidal properties.

 

Acute Suppurative Thyroiditis

 

EPIDEMIOLOGY AND RISK FACTORS      

 

Acute suppurative thyroiditis is rare and is usually due to bacterial infection of the thyroid gland. In severe cases, it can lead to abscess formation and spread to surrounding structures leading to acute obstruction of the respiratory tract. More than 90% of the patients are less than 40 years of age, with females being more commonly affected than males (17, 18). The incidence of acute suppurative thyroiditis lies between 0.1% and 0.7% of all thyroidal illnesses(19). In children acute suppurative thyroiditis is usually due to persistent pyriform sinus and almost always affects the left lobe and is often recurrent (20-22).  Risk factors for acute suppurative thyroiditis are summarized in table 2 (23).

 

Table 2. Risk Factors for Acute Suppurative Thyroiditis

Common risk factors

Pyriform sinus fistula – more common in children and young adults and associated with recurrent disease

Immunocompromised status – AIDS, blood malignancies, uncontrolled diabetes (more common risk factor overall)

Other risk factors

Thyroglossal cyst

Patent foramen cecum

Congenital brachial fistula

Spread of adjacent suppurative infection into thyroid

Anterior esophageal perforation

Underlying thyroid disorders like chronic autoimmune thyroiditis, goiter, and thyroid malignancy

Fine need aspirations/biopsy of thyroid

Dental abscess/ treatment

Systemic autoimmune disorders

 

BACTERIOLOGY

 

Although bacterial agents account for the majority of cases, acute suppurative thyroiditis can also be caused by fungal (immunosuppressive status), parasitic, and tubercular etiology. Common bacterial organisms include Staphylococcus aureus, Streptococcus pyogenes, Staphylococcus epidermidis, and Streptococcus pneumoniae. Rarely other causative bacteria include Klebsiella species, Hemophilus influenzae, Streptococcus viridans, Arcanobacterium haemolyticum, Eikenella corrodensSalmonella species, and Enterobacteriaceae. In the context of immunosuppressed states like HIV-AIDS, acute suppurative thyroiditis can be caused by Mycobacterium tuberculosis, atypical mycobacteria, Salmonella species, Nocardia species and Treponema pallidum (19, 24).

 

CLINICAL PRESENTATION

 

Acute suppurative thyroiditis due to bacterial etiology has a very rapid onset and progression of symptoms if not addressed. The common manifestations are fever, neck pain, and dysphagia. Thyroid gland may be tender on palpation and sometimes there may be swelling with fluctuation suggestive of localized pus collection (25). Very rarely infection can spread to nearby anatomical structures resulting in a more dramatic presentation with stridor due to laryngeal involvement requiring urgent tracheostomy (26). It is important to differentiate this condition from subacute thyroiditis which also presents with systemic symptoms and neck pain (Table 3) (see below).

 

DIAGNOSIS AND MANAGEMENT

 

Laboratory investigations are consistent with acute inflammation characterized by leukocytosis with shift to left, elevated erythrocyte sedimentation rate, raised C- reactive protein (CRP), and other acute inflammatory markers (23). In cases of severe disease, blood cultures may be positive. Ultrasonography of the thyroid may reveal an abscess. The latter requires aspiration and pus should be sent for microbiological diagnosis. Typical findings of acute suppurative thyroiditis on ultrasound are perithyroidal hypoechoic space, effacement of the plane between the thyroid and surrounding tissues, and unilateral presentation [Fig 1] (27). Computed Tomography (CT) offers better spatial resolution and can be used in cases where ultrasound is not showing characteristic findings or when there is involvement of nearby soft tissue structures. Barium swallow studies may be required to diagnose a pyriform sinus, especially in children when there are recurrent episodes of suppurative thyroiditis (28).

Fig 1. A. Ultrasound of the thyroid showing enlargement of the left lobe of the thyroid with heterogenous echotexture, suggestive of thyroiditis B. Ultrasound Doppler showing increased vascularity of the left lobe of the thyroid

Aspiration or surgical drainage of pus with intravenous empirical broad-spectrum antibiotics (especially in sick patients) is the cornerstone of management for acute suppurative thyroiditis. If the patient is immunocompromised, antifungal therapy should be added to initial therapy. In case of extensive involvement of nearby structures, surgical debridement of involved areas may be needed. With respect to culture sensitivity, antibiotic therapy can be modified and once clinical improvement occurs, patients can be switched to oral antibiotics. If there is presence of pyriform fistula, it should be treated either surgically (removal of entire tract with thyroidectomy) or by ablation (21, 29).

 

Subacute Thyroiditis

 

Subacute thyroiditis (also termed as granulomatous, giant cell, or deQuervain’s thyroiditis), is usually due to a viral illness following respiratory illness. Rarely, bacterial infections like Mycobacterium tuberculosis, Treponema pallidum, or Yersinia enterocolitica may cause subacute thyroiditis. Tuberculous thyroiditis is discussed in another chapter (2). Differentiating features of subacute thyroiditis and suppurative thyroiditis are presented in table 3 (19, 30, 31).

 

Table 3. Differentiating Acute Suppurative Thyroiditis and Subacute Thyroiditis

Features

Acute suppurative thyroiditis

Sub-acute thyroiditis

Etiology

Usually bacterial in origin

Usually follows viral upper respiratory tract infection

Presentation

Rapidly evolving, patient can be very toxic with extensive involvement

Presents with systemic symptoms over days to week

Age

Children, 20 to 40 years

20 to 60 years

Sex

Slight female preponderance

More common in females

Fever

 72%

54%

Neck pain

 70%

77%

Neck tenderness

Usually, unilateral (Left sided involvement due to persistent pyriform sinus)

Bilateral and migratory

Redness over skin

Common

Not present

Swelling with fluctuation suggestive of abscess formation

 Common

Not present

History of sore throat

Absent

Present

Clinical features of thyrotoxicosis

Not common

Common in the initial phase

Laboratory

 

 

Leukocytosis

82%

25 to 50 %

Raised ESR

90%

85%

Abnormal thyroid function test

44%

60%

FNAC

Pus

Giant cells, granulomas

Ultrasound Thyroid

Hypoechoic areas with abscess formation, usually unilateral

Ill-defined hypoechoic areas, usually in bilateral lobes

RAIU study

Normal

Decreased in the initial thyrotoxic phase

18 F FDG PET

Increased uptake

Increased uptake

CT scan

Useful when ultrasound is doubtful and when infection extends into peri thyroid tissue

Not useful

Treatment

Antibiotics & drainage of pus

NSAIDS, glucocorticoids in severe cases and sequential follow up of thyroid function tests.

FNAC- fine needle aspiration cytology; RAIU- radioactive iodine uptake; NSAIDS-Non steroidal anti-inflammatory drugs

 

BACTERIAL INFECTIONS OF ADRENALS

 

Tuberculosis of Adrenals

 

Tuberculosis of the adrenal glands is the most common cause of primary adrenal insufficiency in developing countries. An autoimmune etiology remains common in developed countries. Tuberculous infection of the adrenal gland occurs from hematogenous spread from pulmonary or genitourinary sites (32). Adrenals are the most common endocrine gland involved in tuberculosis (2). The symptoms are usually non-specific with generalized weakness, easy fatiguability, loss of weight, loss of appetite, pain in abdomen, and gradually progressive darkening of complexion (Fig 2). These symptoms and signs of adrenal insufficiency do not occur until more than 90% of the glands are destroyed (33). Patient can have low grade fever if the tuberculosis is active and cough and hemoptysis if associated pulmonary involvement is present. In the majority of the cases, the tuberculosis infection may not be active with only a past history of pulmonary tuberculosis (33). Untreated patients may present with adrenal crisis during times of stress. Laboratory investigations reveal low serum cortisol and high plasma adrenocorticotrophic hormone (ACTH). Sometimes, ACTH stimulation test (short synacthen test) may be needed. Adrenal insufficiency is ruled out if serum cortisol level one hour post synacthen (ACTH) stimulation is more than 500-550 nmol/L (14-20 ug/dL depending on the assay). Electrolyte abnormalities noted in adrenal insufficiency are hyponatremia and hyperkalemia. Computed tomography shows bilateral enlarged adrenal masses with areas of necrosis and caseation. In long standing cases, there may be evidence of calcifications (33). Diagnosis is confirmed by adrenal biopsy showing caseating granulomas with acid fast bacilli. Other methods like culture and molecular techniques can be used for diagnosing tuberculosis in biopsy samples. Anti-tubercular treatment (ATT) along with both glucocorticoid and mineralocorticoids remain the treatment of choice. ATT consists of isoniazid - INH (5 mg/kg /d), rifampicin (10 mg/kg /d), pyrazinamide (30 mg/kg /d), and ethambutol (20 mg/kg/d) for 3 to 6 months, subsequently isoniazid and rifampicin for 6 to 12 months (34). In case of multi drug resistant tuberculosis, ATT may be altered with respect to the pattern of resistance. It may require second line medications and longer duration of therapy. Patients usually require lifelong replacement therapy with glucocorticoids and mineralocorticoids.

 

Apart from Mycobacterium tuberculosis, in the context of HIV-AIDS and other immunocompromised states, Mycobacterium avium intracellular and Mycobacterium chelonae may also cause primary adrenal insufficiency.

Fig 2. A. Darkening of the skin in the dorsum aspect of hands in a patient with primary adrenal insufficiency due to adrenal tuberculosis B. Darkening of the palmar aspect including palmar creases of the same patient

Adrenal Abscess

 

An adrenal abscess is a rare clinical condition with very few cases reported in the literature. Organisms that are implicated are Mycobacterium, anaerobes, Salmonella, Nocardia, and E coli. Treatment includes drainage of abscess and antibiotic therapy (35-40). The choice of antibiotic is guided by culture and sensitivity report. In culture negative cases, broad spectrum antibiotics with coverage for gram positive, gram negative, and anaerobic organisms should be considered.

 

Waterhouse-Friderichsen Syndrome

 

Waterhouse-Friderichsen syndrome (WFS) or purpura fulminans is an uncommon clinical entity associated with bilateral adrenal hemorrhage in the setting of severe bacterial sepsis, which was first reported by Rupert Waterhouse and Carl Friderichsen (41). The initial version of this syndrome was classically described with Neisseria meningitidis sepsis. But later it was found that a similar clinical picture was seen with other bacterial infections such as Streptococcus pneumoniaeHemophilus influenzae, Escherichia coli, Staphylococcus aureus, Group A beta-hemolytic Streptococcus, Capnocytophaga canimorsus, Enterobacter cloacae, Pasteurella multocida, Plesiomonas shigelloides,Neisseria gonorrhoeaeMoraxella duplex, Rickettsia rickettsia, Bacillus anthracis, Treponema pallidum,and Legionella pneumophila (42-44).

 

Adrenal glands are predisposed to hemorrhage because around 50-60 small adrenal branches from 3 main adrenal arteries form a subcapsular plexus that drains into the medullary sinusoids through only a few venules (43). Therefore, an increase in adrenal venous pressure due to any cause may lead to hemorrhage. These bacteria may invade the adrenals directly or may produce endotoxins to cause adrenal necrosis and hemorrhage. There is also evidence of microthrombi within the adrenals along with disseminated intravascular coagulation (DIC). Pathologically, organisms are hardly demonstrated in the adrenal specimens (45). The patients are usually sick and present with profound adrenal crisis and shock.  A petechial rash is usually present on the trunk, lower limbs, and mucous membrane and its severity correlates with the degree of thrombocytopenia (44). Treatment involves admission to an intensive care unit and resuscitation with intravenous fluids, intravenous glucocorticoids, and appropriate antibiotics.

 

BACTERIAL INFECTIONS OF GONADS

 

Bacterial Infections of Testes

 

EPIDEMIOLOGY

 

Infection of the epididymis can occur in both children and adults. In severe cases, the inflammation can spread further into testis and present as epididymo-orchitis. If the duration of illness is less than 6 weeks, it is termed as acute epididymo-orchitis, whereas duration more than 6 weeks is termed as chronic. In children, it usually occurs between two and thirteen years of age, whereas in adults, it is common between twenty and thirty years of age (46).

 

BACTERIOLOGY

 

Causative organisms in younger males less than 35 years of age are Neisseria gonorrhoeae and Chlamydia trachomatis. In older men, causative organisms include Escherichia coli, other coliforms, and Pseudomonas. Rare bacterial causes include Ureaplasma species, Mycoplasma genitalium, Mycobacterium tuberculosis, and Brucella species (47-49). Risk factors for epididymitis include urinary tract infections, sexually transmitted diseases, bladder outlet obstruction, prostate enlargement, and urinary tract surgeries or urogenital procedures. In homosexual men, an enteric bacterial etiology is common (46, 50).

 

CLINICAL PRESENTATION

 

Acute epididymitis presents as localized testicular pain. On palpation, there may be swelling in the posterior part of the testis that represents an enlarged testis and inflamed epididymis. More advanced cases present with secondary testicular pain and swelling (epididymo-orchitis). There could be redness of scrotum and hydrocele (reactive fluid collection secondary to infection) (Fig 3). A positive Prehn sign (manual elevation of the scrotum relieves pain) is more often seen with epididymitis than testicular torsion (46).

 

Fig 3. Swelling of bilateral testes with reddening of the skin overlying the scrotum, suggestive of epidymo-orchitis

 

DIAGNOSIS AND MANAGEMENT

 

In all cases of acute epididymo-orchits, it is important to rule out acute surgical conditions like testicular torsion and Fournier’s gangrene. All patients should undergo routine urine microscopy, urine for culture and sensitivity, and a urine nucleic acid amplification test (NAAT) for N. gonorrhoeae and C. trachomatis. NAAT is helpful in diagnosing infections where urine cultures are negative (51). Management depends on the severity of illness, history suggestive of sexually transmitted diseases, and reports of NAAT (summarized in table 4) (46, 52).

 

Table 4. Management of Acute Epididymo-Orchitis

Clinical scenario

Likely organisms

Choice of empirical antibiotic therapy *

Children < 14 years

Various possibilities – secondary to anatomical issues

Treatment based on urine culture results and referral to urologist.

Individuals at risk of sexually transmitted diseases but do not practice anal intercourse

N. gonorrhoeae and C. trachomatis 

 

Single injection of ceftriaxone 500mg intramuscular and oral Doxycycline 100mg twice daily for 10 days. 

 

Alternative for doxycycline – Azithromycin

Alternative for ceftriaxone- Gentamycin

 

Individuals at risk of sexually transmitted diseases but do practice anal intercourse

N. gonorrhoeae, C. trachomatis and enteric pathogens

 

Single injection of ceftriaxone 500mg intramuscular and oral Doxycycline 100mg twice daily for 10 days plus oral levofloxacin 500 mg once daily for 10 days

 

 

Individuals at lower risk of sexually transmitted diseases

Recent urinary tract surgery or instrumentation

 

Enteric pathogens

Oral levofloxacin 500 mg once daily for 10 days

 

*Further treatment should be adjusted based culture and NAAT results; severe cases may require hospitalization and intravenous antibiotics.

 

Bacterial Infections of Ovaries

 

Isolated infection of ovaries is not common. It is usually part of pelvic inflammatory disease. In severe cases, it may present as tubo-ovarian abscess. Tubo-ovarian abscesses are often polymicrobial and typically contain a predominance of anaerobic bacteria. Common organisms include Escherichia coli, Bacteroides fragilis, other Bacteroides species, Pepto-streptococci, and anaerobic streptococci (53). Diagnosis is based on history, physical examination, ultrasound suggesting tubo -ovarian mass or abscess, and microbiological diagnosis. Treatment consists of admission, intravenous antibiotic therapy, and aspiration of abscess if needed. Patients who do not respond, will need surgical intervention (54).

 

 

Yersinia enterocolitica has been implicated in the pathogenesis of autoimmune thyroid disease (55). Immunoglobulins from patients with Yersinia infection inhibit binding of TSH to thyrocytes (56). This could be explained by structural similarity between Yersinia outer membrane proteins (YOP) and epitopes of the TSH receptor (55, 56).

 

Role of gut microbiome has recently implicated in the metabolic syndrome, obesity, and diabetes (57). Many metabolites produced by gut microbes get absorbed into the circulation. They may act on specific receptors to regulate metabolism (58, 59). Also, some bacterial components can act as endocrine factors controlling metabolism(58).

 

CONCLUSION

 

Bacterial infections of the endocrine glands are rare. Pituitary abscesses usually occur in the setting of underlying pathology of the pituitary gland.  It is commonly caused by Streptococci and Staphylococci. MRI of the sella demonstrates a characteristic rim of enhancement after gadolinium injection. Treatment of pituitary abscess is trans-sphenoidal surgery and intravenous antibiotics. Culture is positive in only 19.7% of cases. Acute suppurative thyroiditis is commonly caused by Streptococci and Staphylococci. Important risk factor for acute suppurative thyroiditis in children is pyriform fistula, whereas in adults, it is more common in immunocompromised states. Acute suppurative thyroiditis appear as hypoechoic area on ultrasound. It is treated by ultrasound guided drainage of the abscess and antibiotic therapy. Acute suppurative thyroiditis should be differentiated from subacute thyroiditis. Primary adrenal bacterial infections other than tuberculosis are rare. Waterhouse-Friderichsen syndrome (WFS) is an uncommon clinical entity associated with bilateral adrenal hemorrhage in the setting of severe bacterial sepsis. It is classically described with Neisseria meningitides, but may be associated with other bacteria as well. Toxins produced by bacteria can cause necrosis, hemorrhage, and microthrombi within the adrenal gland leading to WFS. Infection of the epididymis can occur in both children and adults. Sometimes, the inflammation spreads further into testis and presents as epididymo-orchitis. Common bacterial agents causing epididymo-orchitis are N. gonorrhoeae and C.trachomatis. Enteric pathogens should be suspected if there is history of homosexual practice. Management depends on the severity of illness, history of suggestive sexually transmitted diseases, and reports of NAAT (urine nucleic acid amplification test).

 

ACKNOWLEDGEMENTS

 

Dr. Lovekesh Bhatia, Department of Radiodiagnosis, Aadhar Health Institute, Hisar, India

Dr. Vinita Jain, Department of Pediatrics, Aadhar Health Institute, Hisar, India

 

REFERENCES

 

  1. Nagendra L, Boro H, Mannar V. Bacterial Infections in Diabetes. 2022 Apr 5. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dhatariya K, et al. editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000.
  2. Gupta S, Ansari MAM, Gupta AK, Chaudhary P, Bansal LK. Current Approach for Diagnosis and Treatment of Adrenal Tuberculosis-Our Experience and Review of Literature. Surg J (N Y). 2022 Mar 3;8(1):e92-e97.
  3. Jacob JJ, Paul PAM. Infections in Endocrinology: Tuberculosis. 2021 Mar 14. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dhatariya K, et al. editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000.
  4. Machado MJ, Ramos R, Pereira H, Barbosa MM, Antunes C, Marques O, Almeida R. Primary pituitary abscess: case report and suggested management algorithm. Br J Neurosurg. 2021 Aug 24:1-4.
  5. Pekic S, Miljic D, Popovic V. Infections of the Hypothalamic-Pituitary Region. 2021 Aug 9. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dhatariya K, et al. editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000.
  6. Cabuk B, Caklılı M, Anık I, Ceylan S, Celik O, Ustün C. Primary pituitary abscess case series and a review of the literature. Neuro Endocrinol Lett. 2019 Oct;40(2):99-104.
  7. Pekic S, Popovic V. Alternative causes of hypopituitarism: traumatic brain injury, cranial irradiation, and infections. Handb Clin Neurol. 2014;124:271-90.
  8. Adams D, Kern PA. A case of pituitary abscess presenting without a source of infection or prior pituitary pathology. Endocrinol, Diabetes & Metabol Case Reports [Internet]. 2016 Aug 16 [cited 2022 May 10];2016.
  9. Gao L, Guo X, Tian R, Wang Q, Feng M, Bao X, Deng K, Yao Y, Lian W, Wang R, Xing B. Pituitary abscess: clinical manifestations, diagnosis and treatment of 66 cases from a large pituitary center over 23 years. Pituitary. 2017 Apr;20(2):189-194.
  10. Ling X, Zhu T, Luo Z, Zhang Y, Chen Y, Zhao P, et al. A review of pituitary abscess: our experience with surgical resection and nursing care. Transl Cancer Res. 2017 Aug;6(4):852–9.
  11. Vates GE, Berger MS, Wilson CB. Diagnosis and management of pituitary abscess: a review of twenty-four cases. J Neurosurg. 2001 Aug;95(2):233-41.
  12. Bloomer ZW, Knee TS, Rubin ZS, Hoang TD. Case of an atypical pituitary abscess. BMJ Case Rep. 2021 Nov 30;14(11):e246776. 
  13. Bricaire L, Van Haecke C, Laurent-Roussel S, Jrad G, Bertherat J, Bernier M, Gaillard S, Groussin L, Dupin N. The Great Imitator in Endocrinology: A Painful Hypophysitis Mimicking a Pituitary Tumor. J Clin Endocrinol Metab. 2015 Aug;100(8):2837-40.
  14. Benzick AE, Wirthwein DP, Weinberg A, Wendel GD Jr, Alsaadi R, Leos NK, Zeray F, Sánchez PJ. Pituitary gland gumma in congenital syphilis after failed maternal treatment: a case report. Pediatrics. 1999 Jul;104(1):e4. 
  15. Spinner CD, Noe S, Schwerdtfeger C, Todorova A, Gaa J, Schmid RM, Busch DH, Neuenhahn M. Acute hypophysitis and hypopituitarism in early syphilitic meningitis in a HIV-infected patient: a case report. BMC Infect Dis. 2013 Oct 17;13:481.
  16. Har-el G, Sasaki CT, Prager D, Krespi YP. Acute suppurative thyroiditis and the branchial apparatus. Am J Otolaryngol. 1991 Jan-Feb;12(1):6-11. 
  17. Touihmi S, Rkainilham, Mehdaoui A, El boussaadni Y, Oulmaati A. Acute suppurative thyroiditis with abscess. J Pediatr Surg Case Rep. 2021 Feb;65:101757.
  18. Bukvic B, Diklic A, Zivaljevic V. Acute suppurative klebsiella thyroiditis: a case report. Acta Chir Belg. 2009 Mar-Apr;109(2):253-5.
  19. Paes JE, Burman KD, Cohen J, Franklyn J, McHenry CR, Shoham S, Kloos RT. Acute bacterial suppurative thyroiditis: a clinical review and expert opinion. Thyroid. 2010 Mar;20(3):247-55.
  20. Madana J, Yolmo D, Kalaiarasi R, Gopalakrishnan S, Saxena SK, Krishnapriya S. Recurrent neck infection with branchial arch fistula in children. Int J Pediatr Otorhinolaryngol. 2011 Sep;75(9):1181-5. 
  21. Parida PK, Gopalakrishnan S, Saxena SK. Pediatric recurrent acute suppurative thyroiditis of third branchial arch origin--our experience in 17 cases. Int J Pediatr Otorhinolaryngol. 2014 Nov;78(11):1953-7.
  22. Zhang P, Tian X. Recurrent neck lesions secondary to pyriform sinus fistula. Eur Arch Otorhinolaryngol. 2016 Mar;273(3):735-9
  23. Lafontaine N, Learoyd D, Farrel S, Wong R. Suppurative thyroiditis: Systematic review and clinical guidance. Clin Endocrinol (Oxf). 2021 Aug;95(2):253-264
  24. Falhammar H, Wallin G, Calissendorff J. Acute suppurative thyroiditis with thyroid abscess in adults: clinical presentation, treatment and outcomes. BMC Endocr Disord. 2019 Dec 3;19(1):130.
  25. Dunham B, Nicol TL, Ishii M, Basaria S. Suppurative thyroiditis. Lancet. 2006 Nov 11;368(9548):1742.
  26. Minhas SS, Watkinson JC, Franklyn J. Fourth branchial arch fistula and suppurative thyroiditis: a life-threatening infection. J Laryngol Otol. 2001 Dec;115(12):1029-31.
  27. Masuoka H, Miyauchi A, Tomoda C, Inoue H, Takamura Y, Ito Y, Kobayashi K, Miya A. Imaging studies in sixty patients with acute suppurative thyroiditis. Thyroid. 2011 Oct;21(10):1075-80.
  28. Furukawa M, Kano M, Takiguchi T, Umeda R. Piriform sinus fistula as a route of infection in acute suppurative thyroiditis. Auris Nasus Larynx. 1986;13(2):107-12.
  29. Miyauchi A, Inoue H, Tomoda C, Amino N. Evaluation of chemocauterization treatment for obliteration of pyriform sinus fistula as a route of infection causing acute suppurative thyroiditis. Thyroid. 2009 Jul;19(7):789-93. 
  30. Pearce EN, Farwell AP, Braverman LE. Thyroiditis. N Engl J Med. 2003 Jun 26;348(26):2646-55.
  31. Shrestha RT, Hennessey J. Acute and Subacute, and Riedel’s Thyroiditis. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dhatariya K, et al., editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000-.
  32. Del Borgo C, Urigo C, Marocco R, Belvisi V, Pisani L, Citton R, et al. Diagnostic and therapeutic approach in a rare case of primary bilateral adrenal tuberculosis. J Med Microbiol. 2010 Dec;59 (Pt 12):1527-1529
  33. Kelestimur F. The endocrinology of adrenal tuberculosis: the effects of tuberculosis on the hypothalamo-pituitary-adrenal axis and adrenocortical function. J Endocrinol Invest. 2004 Apr;27(4):380-6.
  34. Laway BA, Khan I, Shah BA, Choh NA, Bhat MA, Shah ZA. Pattern of adrenal morphology and function in pulmonary tuberculosis: response to treatment with antitubercular therapy. Clin Endocrinol (Oxf). 2013 Sep;79(3):321-5.
  35. Regino CA, Gómez JP, Mosquera-Klinger G. Endoscopic Ultrasound-Guided Transgastric Puncture and Drainage of an Adrenal Abscess in an Immunosuppressed Patient. Clin Endosc. 2022 Mar;55(2):302-304. 
  36. O'NEILL JA Jr, HALL WH. ISOLATED ADRENAL ABSCESS SECONDARY TO SALMONELLA. Arch Surg. 1965 Mar;90:454-6.
  37. Midiri M, Finazzo M, Bartolotta TV, Maria MD. Nocardial adrenal abscess: CT and MR findings. Eur Radiol. 1998;8(3):466-8.
  38. Yokoyama S, Sekioka A, Utsunomiya H, Hara S, Takahashi T, Yoshida A. Adrenal abscess as a complication of Escherichia coli sepsis in neonates: A case report. J Pediatr Surg Case Rep. 2013 Sep;1(9):328–30.
  39. Jin W, Miao Q, Wang M, Zhang Y, Ma Y, Huang Y, et al. A rare case of adrenal gland abscess due to anaerobes detected by metagenomic next-generation sequencing. Ann Transl Med. 2020 Mar;8(5):247. 
  40. Rumińska M, Witkowska-Sędek E, Warchoł S, Dudek-Warchoł T, Brzewski M, Pyrżak B. Adrenal abscess in a 3-week-old neonate - a case report. J Ultrason. 2015 Dec;15(63):429-37.
  41. Varon J, Chen K, Sternbach GL. Rupert Waterhouse and Carl Friderichsen: adrenal apoplexy. J Emerg Med. 1998 Jul-Aug;16(4):643-7. 
  42. Hamilton D, Harris MD, Foweraker J, Gresham GA. Waterhouse-Friderichsen syndrome as a result of non-meningococcal infection. J Clin Pathol. 2004 Feb;57(2):208-9. 
  43. Karki BR, Sedhai YR, Bokhari SRA. Waterhouse-Friderichsen Syndrome. [Updated 2021 Dec 12]. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2022 Jan-.
  44. Kalinoski T. Waterhouse-Friderichsen Syndrome with Bilateral Adrenal Hemorrhage Associated with Methicillin-Resistant Staphylococcus aureus (MRSA) Bacteremia in an Adult Patient with History of Intravenous Drug Use. Am J Case Rep. 2022 Apr 14;23:e936096.
  45. Guarner J, Paddock CD, Bartlett J, Zaki SR. Adrenal gland hemorrhage in patients with fatal bacterial infections. Mod Pathol. 2008 Sep;21(9):1113-20.
  46. McConaghy JR, Panchal B. Epididymitis: An Overview. Am Fam Physician. 2016 Nov 1;94(9):723-726. 
  47. Tracy CR, Steers WD, Costabile R. Diagnosis and management of epididymitis. Urol Clin North Am. 2008 Feb;35(1):101-8; vii. 
  48. Doble A, Taylor-Robinson D, Thomas BJ, Jalil N, Harris JR, Witherow RO. Acute epididymitis: a microbiological and ultrasonographic study. Br J Urol. 1989 Jan;63(1):90-4. 
  49. Hawkins DA, Taylor-Robinson D, Thomas BJ, Harris JR. Microbiological survey of acute epididymitis. Genitourin Med. 1986 Oct;62(5):342-4.
  50. Kaver I, Matzkin H, Braf ZF. Epididymo-orchitis: a retrospective study of 121 patients. J Fam Pract. 1990 May;30(5):548-52.
  51. Wampler SM, Llanes M. Common scrotal and testicular problems. Prim Care. 2010 Sep;37(3):613-26, x.
  52. Trojian TH, Lishnak TS, Heiman D. Epididymitis and orchitis: an overview. Am Fam Physician. 2009 Apr 1;79(7):583-7.
  53. Kairys N, Roepke C. Tubo-Ovarian Abscess. 2021 Jul 18. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2022 Jan–.
  54. Munro K, Gharaibeh A, Nagabushanam S, Martin C. Diagnosis and management of tubo-ovarian abscesses. Obstet Gynecol. 2018 Jan;20(1):11–9.

         Mar;93(3):674-6.

  1. Effraimidis G, Wiersinga WM. Mechanisms in endocrinology: autoimmune thyroid disease: old and new players. Eur J Endocrinol. 2014 Jun;170(6):R241-52. 
  2. Li R, Li Y, Li C, Zheng D, Chen P. Gut Microbiota and Endocrine Disorder. Adv Exp Med Biol. 2020;1238:143-164.
  3. Rastelli M, Cani PD, Knauf C. The Gut Microbiome Influences Host Endocrine Functions. Endocr Rev. 2019 Oct 1;40(5):1271-1284
  4. Cani PD, Knauf C. How gut microbes talk to organs: The role of endocrine and nervous routes. Mol Metab. 2016 May 27;5(9):743-52.

Hyperglycemic Hyperosmolar State

CLINICAL RECOGNITION

 

The hyperglycemic hyperosmolar state (HHS) is a life-threatening metabolic decompensation of diabetes which presents with severe hyperglycemia and profound dehydration, typically accompanied by alteration in consciousness ranging from lethargy to coma. In contrast to diabetic ketoacidosis (DKA) in which acidemia and ketonemia are key features, these are limited in HHS. Mortality in HHS ranges from 5-20% and is higher at the extremes of age and in the presence of coma. HHS is more prevalent in type 2 diabetics and in about 7-17% of cases is the initial presentation classically seen in institutionalized elderly patients with diminished thirst perception or inability to ambulate to get free water as needed. HSS is extremely rare as first presentation in patients with type 1 diabetes. Infections are the leading precipitant of HHS, but it can also be precipitated by poor medication compliance, cerebrovascular accident, myocardial infarction, pancreatitis, trauma, alcohol abuse and drugs such as corticosteroids and atypical antipsychotics.

 

PATHOPHYSIOLOGY

 

HHS and DKA represent the two ends of the spectrum of markedly decompensated diabetes, differing mainly in severity of acidosis, ketosis and dehydration. HHS usually occurs with a lesser degree of insulinopenia compared with DKA, but the pathophysiology is otherwise thought to be similar. In both entities, there is a decrease in net effective insulin action with concomitant elevation of counterregulatory hormones. In the setting of relative insulin deficiency, glucagon, catecholamines and cortisol stimulate hepatic glucose production though glycogenolysis and gluconeogenesis. High catecholamines and low insulin reduce peripheral glucose uptake. Unlike DKA, there is adequate insulin available in HHS to restrain lipolysis and ketogenesis, as well as to restrain marked elevation of counterregulatory hormones, such cortisol, glucagon and growth hormone. However, there is significant hyperglycemia with resultant glycosuria leading to loss of water and electrolytes, dehydration, decreased renal perfusion, decreased glucose clearance, and exacerbation of hyperglycemia, ultimately causing impaired level of consciousness. In HHS, the initial increase in proinflammatory cytokines, reactive oxygen species, and plasminogen activator inhibitor-1 can contribute to increased prothrombotic risk.

 

DIAGNOSIS AND DIFFERENTIAL

 

HHS usually has a slower onset than DKA, with symptoms developing over several days or weeks. Patients present with polyuria, polydipsia, weakness, and blurred vision. Altered sensorium is classic in HHS, but mental status can range from fully alert to confused, lethargic, or comatose. Seizure can occur in up to 20% of the patients. Exam reveals physical signs of dehydration, including dry mucous membranes, poor skin turgor, cool extremities, hypotension, and tachycardia. Fever may or may not be present, suggesting underlying infection, although normothermia or even hypothermia may be present due to concomitant vasodilatation.

 

The diagnostic criteria for HHS include severe hyperglycemia and hyperosmolality with preservation of near normal pH and bicarbonate, and minimal or absent serum and/or urine ketones. ADA guidelines note glucose level at presentation should be > 600 mg/dl, with pH > 7.3 and bicarbonate level > 20 mEq/L. These are common diagnostic criteria that differentiate HHS from DKA (Table 1). However, it is clear that a subpopulation of patients with type 2 diabetes can present with overlapping features of HHS and DKA. Patients with ketosis prone type 2 diabetes present with ketosis and milder acidosis than the one expects in DKA and in some cases with near normal pH and bicarbonate. More rarely, HHS can present in the setting of diabetes insipidus where patients are treated with intravenous dextrose for the severe dehydration leading to hyperglycemia and glycosuria.

 

Table 1. Diagnosis of HHS Versus DKA

 

HHS

DKA

Diagnostic criteria

pH

>7.30

≤7.30

Plasma Glucose

>600 mg/dl

>250 mg/dl

Serum bicarbonate

>15 mEq/L

<18 mEq/L

Plasma and urine ketones

None or trace

Positive

Anion gap

 

<12

>12

Serum Osmolality

>320 mOsm/kg

Variable

Glycosuria

 

++

++

Typical Deficit

Water (ml/kg)

100-200 (9L)

100 (6L)

Na+ (mEq/kg)

5-13

7-10

Cl- (mEq/kg)

5-15

3-5

K+(mEq/Kg)

4-6

3-5

P04 (mmol/kg)

3-7

5-7

Mg++& Ca++(mEq/kg)

1-2

1-2

Adapted from Kitabchi A, et al. Diabetes Care, 2006, 29: 2739-2747

 

DIAGNOSTIC TESTS NEEDED

 

Initial laboratory tests should include electrolytes with calculated anion gap, plasma glucose, blood urea nitrogen (BUN), creatinine, serum and urine ketones, osmolality, and arterial blood gas.  Evidence of infection should be sought by checking complete blood count with differential and urinalysis, with consideration of additional evaluation including chest X-ray, and culture of blood, sputum and urine. Electrocardiogram and head CT should be done if clinically indicated.  HHS produces significant loss of several electrolytes as well as a prerenal azotemia and increased hematocrit, the latter due to hemoconcentration. An increase of serum sodium in the presence of hyperglycemia indicates severe dehydration. Altered mentation appears to correlate with the degree of hyperosmolality; hence significantly diminished mentation in the setting of an osmolality of <320 mOsm/kg should prompt a search for other causes. It is notable that despite significant potassium losses, serum potassium is usually normal or even elevated on presentation because of extracellular shift in the setting of hyperosmolality and insulin deficiency. HgbA1c may be useful to discriminate chronic uncontrolled hyperglycemia from acute metabolic decompensation.   

 

THERAPY

 

There is a lack on randomized controlled trials for the treatment of HHS and the American Diabetes Association (ADA) has developed guidelines that combine the treatment of HHS and DKA. The treatment of HHS includes a four-pronged approach:

  • reestablishment of volume status with vigorous intravenous hydration;
  • electrolyte replacement;
  • correction of hyperglycemia with volume expansion and administration of intravenous insulin;
  • diagnosis and management of potential precipitants.

 

The initial emergent treatment has been summarized in table 2.

 

Fluid Replacement

 

Aggressive fluid replacement is critical in order to prevent cardiovascular collapse, with repletion of intravascular and extravascular volume and restoration of renal perfusion. The total fluid deficit should be estimated (usually 100-200 ml/kg), with the goal of replacement over 24 hours. In the absence of heart failure, 1-1.5 liters of isotonic saline should be given over the first hour. Subsequent fluid replacement depends on the hydration and electrolyte status. In patients with hypotension, aggressive isotonic saline infusion should continue until the patient is stabilized. Increased plasma sodium concentration in the setting of hyperglycemia suggests a significant water deficit; clinical practice guidelines recommend adding a correction factor of 1.6 mg/dl to the plasma sodium concentration for each 100 mg/dl of glucose above 100 mg/dl. In the normotensive patient with a corrected serum sodium level that is normal or high, fluid replacement can be continued with half normal saline given at 250-500 cc/hour, whereas if the corrected serum sodium level is low, isotonic saline should be administered at similar rate. When serum glucose reaches 200-300 mg/dl, fluid should be changed to 5% dextrose solution in half normal saline.

 

Electrolyte Replacement

 

Electrolyte replacement is the second crucial step in HHS management. Serum potassium can be normal or elevated on presentation despite total body potassium depletion. Osmotic-induced intracellular dehydration results in potassium efflux from the cells. Since insulin causes a shift of potassium into the cell, it is mandatory to correct the potassium level to >3.3 mEq/L before starting insulin therapy. If potassium is between 3.3 and 5.3 mEq/L, 20-30 mEq of potassium should be given in each liter of intravenous fluid to keep serum potassium between 4 to 5 mEq/L. The potassium should be monitored if >5.3 meq/L and potassium replacement initiated when potassium < 5.3 meq/L. Magnesium should be checked and repleted as necessary; this is important to prevent renal wasting of potassium with exacerbation of hypokalemia. Routine administration of phosphate is not recommended (17); however, careful phosphate replacement can be considered in patients with very low levels (<1 meq/L), cardiac dysfunction, or respiratory distress.

 

Insulin Therapy

 

The treatment of choice for correction of hyperglycemia is regular insulin by continuous infusion after adequate fluid and potassium replacement. While randomized controlled studies in patients with DKA have shown that insulin therapy is effective regardless of the route of administration, there is limited data supporting the use of subcutaneous or intramuscular insulin in HHS and continuous intravenous insulin administration remains the treatment of choice in patient with significant dehydration, reduced level of consciousness, and critical illness. Insulin should be given as initial bolus of 0.1 unit per kilogram body weight, followed by a drip of 0.1 unit per kilogram per hour; alternatively, 0.14 units per kilogram per hour can be given as an infusion without a bolus. If the glucose level does not decrease by 50-70 mg/dl in the first hour, the insulin dose may be doubled.  When the plasma glucose level reaches 300 mg/dl, insulin infusion may be reduced to 0.05-0.1 unit/kg/hour and dextrose can be added to the fluids to keep the glucose level between 250-300 mg/dl until hyperosmolality has resolved and the patient is alert.

 

Evaluation of Precipitant Factors

 

Evaluation for and treatment of potential precipitant factors is important. Patients with HHS have a mortality rate of about 5-20%, 10-fold higher than patients with DKA and several studies have shown that the increased mortality is likely because of the precipitating factors. For this reason, appropriate work up and treatment should be given as indicated.

 

Table 2. Initial Emergent Treatment for HHS

1--IV Fluids

a-Cardiogenic shock

b-Severe hypovolemia

c-Mild dehydration

Hemodynamic Monitoring/ Pressors/ 0.9% NaCl

0.9% NaCl (1L/hr.)

Na* low:

0.9% NaCl (250-500 ml/hr.) † 
Na* normal or high:

0.45% NaCl (250-500 ml/hr.) † 

 

When serum glucose ≤300 mg/dl, 5% dextrose/0.45% NaCl (150-250 ml/hr.)

2-IV Potassium (with adequate renal function)

 

a--K+ <3.3 mEq/L

b--K+ 3.3-5.3 mEq/L

c--K+ >5.3 mEq/L

Hold insulin, K 20-30 mEq/ hr. until K+ >3.3 mEq/L

K 20-30 mEq in each liter of IV fluid to keep K+ 4-5 mEq/L

Do not give K; monitor

3-IV Insulin

 

Bolus 0.1 unit/Kg, then 0.1 unit/Kg/hr. infusion (or 0.14 unit/kg/hr. infusion w/o bolus)

Double infusion if glucose does not decrease by 50-70 mg/dl in the first hour

When serum glucose 300 mg/dl, ↓ insulin infusion to 0.05-0.1 units/Kg/hr.

Adapted from Kitabchi A, et al. Diabetes Care, 2006, 29: 2739-2747

*Corrected serum sodium; † depending on the hydration status

 

FOLLOW-UP

 

Meticulous clinical and laboratory follow up is critical in patients with HHS. Capillary blood glucose levels should be monitored every hour to allow adjustment of the insulin infusion. Electrolytes, BUN, creatinine and plasma glucose should be checked every 2-4 hours until the patient is stable. When plasma osmolality is <315 mOsm/L, and the patient is alert and able to eat, a multidose insulin regime consisting of long-acting insulin and short/rapid acting insulin before meals may be initiated. Intravenous insulin infusion should be continued for 1-2 h after the subcutaneous insulin is given to ensure adequate plasma insulin levels are maintained.

 

It is also important to monitor for possible treatment-related complications, the most common of which are hypoglycemia and hypokalemia.  These are both usually due to overzealous treatment with insulin and can be minimized with frequent monitoring. Cerebral edema is extremely rare in patients with HHS, and usually occurs in younger adults. To reduce the risk of cerebral edema in high-risk patients, sodium, glucose, and water deficit may be more gradually corrected to avoid the rapid decline in plasma osmolality. Recurrence of HHS can be prevented by improved patient as well as caregiver education and enhanced access to medical care. For elderly nursing home residents, nursing home staff should be educated in recognition of signs and symptoms of HHS and on the importance of adequate fluid intake.

 

REFERENCES

 

            Gosmanov AR, Gosmanova EO, Kitabchi AE. Hyperglycemic Crises: Diabetic Ketoacidosis and Hyperglycemic Hyperosmolar State. 2021 May 9. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dhatariya K, Dungan K, Hershman JM, Hofland J, Kalra S, Kaltsas G, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrère B, Levy M, McGee EA, McLachlan R, Morley JE, New M, Purnell J, Sahay R, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000–. PMID: 25905280

 

Pasquel, F.J., and Umpierrez, G.E. 2014. Hyperosmolar hyperglycemic state: a historic review of the clinical presentation, diagnosis, and treatment. Diabetes Care 37:3124-3131.

 

 

Umpierrez, G., and Korytkowski, M. 2016. Diabetic emergencies - ketoacidosis, hyperglycaemic hyperosmolar state and hypoglycaemia. Nat Rev Endocrinol 12:222-232.

 

Kitabchi, A.E., Umpierrez, G.E., Miles, J.M., and Fisher, J.N. 2009. Hyperglycemic crises in adult patients with diabetes. Diabetes Care 32:1335-1343.

 

Palmer, B.F., and Clegg, D.J. 2015. Electrolyte and Acid-Base Disturbances in Diabetes Mellitus. N Engl J Med 373:2482-2483.

 

Dhatariya, K.K., and Vellanki, P. 2017. Treatment of Diabetic Ketoacidosis (DKA)/Hyperglycemic Hyperosmolar State (HHS): Novel Advances in the Management of Hyperglycemic Crises (UK Versus USA). Curr Diab Rep 17:33.

 

 

 

 

 

 

 

 

 

Initial Management of Severe Hyperglycemia in Type 2 Diabetes

CLINICAL RECOGNITION

 

Type 2 diabetes mellitus (DM) is a common disease affecting 26 million people, 8.3% of the US population.  Of these, an estimated 7 million people are undiagnosed.

 

Type 2 DM typically has two pathophysiologic defects:  an insulin secretory defect and insulin resistance.  Symptoms of uncontrolled hyperglycemia include polyuria, polydipsia, blurry vision, and possibly dehydration and weight loss. Patients may complain of thirst, sweet cravings, generalized fatigue, abdominal discomfort, and muscle cramps. They may have a history of poor wound healing and/or frequent infections. Basic metabolic laboratory tests may reveal a random blood glucose level over 200 mg/dL [11.1 mmol/L], hyper- or hyponatremia, hypokalemia, metabolic acid-base derangements, and acute renal or prerenal insufficiency. Historical clues for the diagnosis of type 2 DM might include pre-existing history of pre-diabetes, a family history of type 2 diabetes, an ethnicity at higher risk for DM (African-American, Hispanic, Native American, Pacific Islander), a history of gestational diabetes, obesity, and sedentary lifestyle. 

 

PATHOPHYSIOLOGY  

 

Table 1. Clinical Features of the Acute Presentation of Type 2 Diabetes and Pathophysiology

Hyperglycemia

Insulin resistance, insulin deficiency (pancreatic beta cell failure), increased gluconeogenesis, glycogenolysis

Dehydration, polyuria, polydipsia

Osmotic diuresis, compensatory thirst

Weight loss, sweet cravings

Glycosuric calorie loss and inadequate glucose utilization

Muscle pain and abdominal discomfort

Lactic acid accumulation, hypokalemia, electrolyte /acid-base derangements

Metabolic alkalosis and/or acidosis, electrolyte disturbances

Dehydration and ketogenesis

Ketogenesis

Insulin deficiency resulting in lipolysis yielding free fatty acids, substrate for formation of ketone bodies

 

DIAGNOSIS AND DIFFERENTIAL

 

Diabetes can be diagnosed in several ways: 1) Presence of symptoms of hyperglycemia with a random blood glucose of 200 mg/dL [11.1 mmol/L]; 2) fasting blood glucose > 126 mg/dL [7.0 mmol/L; 3) the 75-gram oral glucose tolerance test with a blood glucose > 200 mg/dL [11.1 mmol/L] at 2 hours; 4) hemoglobin A1C value > 6.5%.  If asymptomatic, the diagnosis of diabetes is confirmed with two consecutive day abnormal results from the same test or a different test or with two different tests on the same day. If using the hemoglobin A1C for diagnosis, one should be aware of several conditions (some common) making this measure un-interpretable

 

Adult patients with type 1 and type 2 DM can sometimes present similarly.  If a patient presents with hyperglycemia, ketonemia, and metabolic acidosis, distinguishing between types of diabetes is not necessary in this acute setting because initially, both type 1 and type 2 DM are treated with insulin.  Later the two diseases may be distinguished with antibody testing although this is neither completely sensitive nor specific. Type 2 DM can also present acutely with a hyperglycemic hyperosmolar state (HHS) with dehydration, altered level of consciousness, and a lesser degree of clinical ketosis than seen in diabetic ketoacidosis (DKA). Consideration for genetic syndromes and concomitant rare conditions of endocrine hormone excess (cortisol, growth hormone, epinephrine, glucagon) leading to hyperglycemia should be in the non-urgent setting for patients with new diagnoses of diabetes.

 

DIAGNOSTIC TESTS NEEDED AND SUGGESTED

 

For an acute presentation of diabetes with hyperglycemic symptoms, the patient should have a basic metabolic panel of laboratory tests including glucose, electrolytes, blood urea nitrogen, creatinine, blood and or urinary ketones, liver function tests, and urinalysis. Other testing should be guided by a patient’s history and physical exam and might include evaluation for infection or cardiac dysfunction.  A hemoglobin A1C reflects the average blood glucose over the last 90 days and is a helpful test.  Distinguishing type 1 from type 2 DM can on occasions be difficult but can be assisted with autoantibody testing [tyrosine phosphatase antibody (IA-2) or glutamic acid decarboxylase (GAD) 65 antibody]. The presence of antibody suggests an autoimmune lesion as seen in type 1 DM. In type 1 DM insulin and C-peptide levels are characteristically low, whereas they may be normal or elevated at the onset of type 2 DM.

 

TREATMENT

 

Insulin therapy is the initial management choice for patients presenting with hyperglycemia and catabolic symptoms including weight loss. If laboratory abnormalities suggest concurrent DKA or HHS, these must be treated emergently with aggressive saline rehydration, intravenous insulin, potassium and other electrolyte replacement.

 

For a severely hyperglycemic patient, with a catabolic presentation that usually includes moderate to severe volume depletion, the first therapeutic step is rehydration, usually with intravenous saline.  After adequate hydration, therapy with physiologic doses of insulin (0.3-0.4 units per kilogram body weight daily) is recommended. The ideal treatment regimen would be a combination of a long-acting basal insulin plus multiple premeal prandial “bolus” injections to manage meal-related insulin requirements and correction of pre-meal hyperglycemia, referred to as basal-bolus insulin therapy. A good starting place is to prescribe half the total daily insulin dose as basal and the other half as bolus. The combination of long-acting insulin and a rapid acting analogue are good options for basal-bolus therapy. The basal dose is given as a separate injection from the bolus injection.

 

The premeal “bolus” dose is calculated by summing the dose required to cover the carbohydrate load plus the dose to correct premeal hyperglycemia and is given as one injection 10-15 minutes before the meal. Particularly with premeal hyperglycemia but even with mealtime glucose levels within target, today’s rapid-acting analogues require time for absorption to avoid more severe postprandial hyperglycemia (this is typically called the “lag time”).  In an acute setting, and in a less sophisticated patient, it might be more appropriate to begin therapy with a twice-daily pre-mixed insulin. Even though this regimen is not ideal for many for the long-term because it does not allow for sufficient dose titration, this regimen allows approximate physiologic basal-bolus insulin coverage with fewer injections. Nevertheless, if starting with basal-bolus or premixed insulin, it is best to teach the patient to use the strategy of correcting pre-meal hyperglycemia with an additional dose of rapid acting insulin analogue, given 10-15 minutes before the meal. This adds tremendous flexibility to an otherwise rigid regimen.

 

Until more education is possible, the need to limit high glycemic-load carbohydrate intake (such as with sweetened beverages and juice) should be strongly reinforced with counseling. Certainly, arrangements for general and dietary diabetes education should be made for a newly diagnosed diabetic patient or for a patient new to insulin therapy.

 

FOLLOW-UP

 

The patient will use a glucose meter to check his/her fasting and premeal blood glucose levels.  For the patient on basal-bolus insulin therapy, he/she will increase bedtime basal insulin doses by 1-2 units every 3 days until fasting blood glucose falls into target range of 90 -130 mg/d [5 – 7.2 mmol/L]. Ideally, bedtime and fasting glucose levels are about the same at the end of the basal insulin titration. If there is a consistent reduction in bedtime to fasting glucose by more than 50 mg/dL [2.8 mmol/L], basal insulin dose is too high.

 

Adjustments for pre-meal insulin doses are most easily made with an algorithm written clearly for the patient to reference. The importance of injecting the mealtime insulin 10 -15 minutes before eating needs to be emphasized. In contrast to type 1 diabetes where carbohydrate counting is standard, most type 2 patients do well by taking the same mealtime dose or altering up or down based on the size of the meal. For example, one might take 8 units for a smaller meal and 12 units for a large one. If patients feel hypoglycemic symptoms (sweating, shaking, mental fogginess, hunger) despite concurrent blood glucoses levels in the normal range, one could use smaller insulin dose increments to lower blood glucose into the target range more gradually.  Generally, increases of insulin dose by10% are well tolerated by patients.  Late night snacks without insulin coverage may lead to morning hyperglycemia and interfere with the assessment of the adequacy of the bedtime insulin doses.  Correction doses are “trial and error” but most patients with type 2 diabetes require an “insulin sensitivity factor” of 30 (i.e., 30 mg/dL glucose reduction expected from one unit of insulin injected). For example, if additional insulin is provided for premeal glucose levels above 150 mg/dL, 1 extra unit would be given for 150-180 mg/dL, 2 units for 181-210 mg/dL, etc.  When starting insulin, it may be appropriate to use a more conservative insulin sensitivity factor such as 40 or 50.

 

Table 2.  Premeal Bolus Dose Calculation Using Rapid-Acting Insulin Analogue

Total premeal insulin dose is sum of:

Suggested Units

Meal coverage

5-8 units for smaller meal, 9-12 units for larger meal

Pre meal hyperglycemia correction

1 unit per 30-50 mg/dL above150 mg/dL

 

Initial diabetes therapy includes counseling for lifestyle and diabetic nutritional interventions.  Starting therapy with metformin could also be considered as an adjunctive therapy with insulin to reduce insulin requirements and minimize weight gain. Overtime with lifestyle changes, a decrease in glucose toxicity, and the addition of other hypoglycemic agents some patients who present with very high glucose levels may be able discontinue insulin therapy.

 

GUIDELINES

 

2019 Update to: Management of Hyperglycemia in Type 2 Diabetes, 2018. A Consensus Report by the American Diabetes Association (ADA) and the European Association for the Study of Diabetes (EASD). Buse JB, Wexler DJ, Tsapas A, Rossing P, Mingrone G, Mathieu C, D'Alessio DA, Davies MJ. Diabetes Care. 2020 Feb;43(2):487-493

 

REFERENCES

 

Donner T, Sarkar S. Insulin – Pharmacology, Therapeutic Regimens, and Principles of Intensive Insulin Therapy. 2019 Feb 23. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dhatariya K, Dungan K, Hershman JM, Hofland J, Kalra S, Kaltsas G, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrère B, Levy M, McGee EA, McLachlan R, Morley JE, New M, Purnell J, Sahay R, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000–. PMID: 25905175

 

Feingold KR. Oral and Injectable (Non-Insulin) Pharmacological Agents for the Treatment of Type 2 Diabetes. 2021 Aug 28. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dhatariya K, Dungan K, Hershman JM, Hofland J, Kalra S, Kaltsas G, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrère B, Levy M, McGee EA, McLachlan R, Morley JE, New M, Purnell J, Sahay R, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000–. PMID: 25905364

 

Gosmanov AR, Gosmanova EO, Kitabchi AE. Hyperglycemic Crises: Diabetic Ketoacidosis and Hyperglycemic Hyperosmolar State. 2021 May 9. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dhatariya K, Dungan K, Hershman JM, Hofland J, Kalra S, Kaltsas G, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrère B, Levy M, McGee EA, McLachlan R, Morley JE, New M, Purnell J, Sahay R, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000–. PMID: 25905280

 

 

Diabetic Ketoacidosis

CLINICAL RECOGNITION

 

Omission of insulin and infection are the two most common precipitants of diabetic ketoacidosis (DKA). Noncompliance may account for up to 44% of DKA presentations; while infection is less frequently observed.

 

Acute medical illnesses involving the cardiovascular system (myocardial infarction, stroke, acute thrombosis), gastrointestinal tract (bleeding, pancreatitis), endocrine axis (acromegaly, Cushing`s syndrome, hyperthyroidism) and recent surgical procedures can contribute to the development of DKA by causing dehydration, increase in insulin counterregulatory hormones, and worsening of peripheral insulin resistance.

 

Medications such as diuretics, beta-blockers, corticosteroids, second-generation anti-psychotics, anti-convulsants, sodium-glucose cotransporter-2 (SGLT-2) inhibitors, and/or immune checkpoint inhibitors may affect carbohydrate metabolism and volume status and, therefore, could precipitate DKA. SGLT-2 inhibitors have been associated with euglycemic DKA (glucose level < 250mg/dL)

 

Other factors leading to DKA include psychological problems, eating disorders, insulin pump malfunction, and drug abuse. It is well recognized that new onset T2DM can sometimes manifest with DKA. These patients are obese, mostly African Americans or Hispanics and have undiagnosed hyperglycemia, impaired insulin secretion, and impaired insulin action. A recent report suggests that cocaine abuse is an independent risk factor associated with DKA recurrence.

 

PATHOPHYSIOLOGY

 

Insulin deficiency, increased insulin counter-regulatory hormones (cortisol, glucagon, growth hormone, and catecholamines), and peripheral insulin resistance lead to hyperglycemia, dehydration, ketosis, and electrolyte imbalance which underlie the pathophysiology of DKA.

 

Hyperglycemia of DKA evolves through accelerated gluconeogenesis, glycogenolysis, and decreased glucose utilization – all due to absolute insulin deficiency. Of note, diabetes patients who developed DKA while treated with SGLT-2 inhibitors can present without hyperglycemia, i.e., with euglycemic DKA.

 

Due to increased lipolysis and decreased lipogenesis, abundant free fatty acids are converted to ketone bodies: β-hydroxybutyrate (β-OHB), acetoacetate, and acetone. Hyperglycemia-induced osmotic diuresis, if not accompanied by sufficient oral fluid intake, leads to dehydration, hyperosmolarity, electrolyte loss, and subsequent decrease in glomerular filtration. With decline in renal function, glycosuria diminishes and hyperglycemia/hyperosmolality worsens. With impaired insulin action and hyperosmolality, utilization of potassium by skeletal muscle is markedly diminished leading to intracellular potassium depletion. Also, potassium is lost via osmotic diuresis causing profound total body potassium deficiency. Therefore, DKA patients can present with broad range of serum potassium concentrations. Nevertheless, a “normal” plasma potassium concentration may indicate that potassium stores in the body are severely diminished and the institution of insulin therapy and correction of hyperglycemia will lead to hypokalemia.

 

DIAGNOSIS AND DIFFERENTIAL DIAGNOSIS

 

Diagnostic criteria for DKA are presented in Table 1.

 

Table 1. Criteria and Classification of DKA

DKA

Mild

Moderate

Severe

Plasma glucose (mg/dl)

>250 mg/dl

>250mg/dl

>250mg/dl

Arterial pH

7.25-7.30

7.00-7.24

<7.00

Serum bicarbonate (mEq/L)

15-18

10- 15

<10

Urine ketone*

+

+

+

Serum ketone*

+

+

+

Effective Serum Osmolality**

Variable

Variable

Variable

Anion Gap***

>10

>12

>12

Mental Status

Alert

Alert/drowsy

Stupor/coma

*Nitroprusside reaction method

** Serum osmolality: 2[measured Na+ (mEq/L)] + glucose (mg/dl)/18 = mOsm/kg

*** Anion Gap: [ (Na+)– (Cl- + HCO3- (mEq/L)]

 

CLINICAL PRESENTATION

 

Polyuria, polydipsia, weight loss, vomiting, and abdominal pain usually are present in patients with DKA. Abdominal pain can be closely associated with acidosis and resolves with treatment. Physical examination findings such as hypotension, tachycardia, poor skin turgor, and weakness support the clinical diagnosis of dehydration in DKA. Mental status changes may occur in DKA and are likely related to degree of acidosis and/or hyperosmolarity. A search for symptoms of precipitating causes such as infection, vascular events, or existing drug abuse should be initiated in the emergency room. Patients with hyperglycemic crises can be hypothermic because of peripheral vasodilation and decreased utilization of metabolic substrates.

 

DIFFERENTIAL DIAGNOSIS

 

Hyperglycemic hyperosmolar state is not associated with ketosis. Starvation and alcoholic ketoacidosis are not characterized by hyperglycemia >200 mg/dl and bicarbonate level <18 meq/L. With hypotension, decreased renal function, and history of metformin use, lactic acidosis (lactic acid level >7 mmol/L) should be suspected. Ingestion of methanol, isopropyl alcohol, and paraldehyde can also alter anion gap and/or osmolality and need to be investigated.

 

Table 2. Laboratory Evaluation of Causes of Acidosis

Factor Studied

DKA

HHS

Starvation

Uremic acidosis

pH

normal

normal

Mild↓

Plasma glucose

>500 mg/dl

normal

normal

Glycosuria

+ +

+ +

0

0

Total plasma ketones*

↑↑

0 or ↑

Mild↑

0

Anion gap

Normal

Mild↑

Mild↑

Osmolality

>330 mOsm/kg

normal

Normal/↑

Other

     

BUN>200 mg/dl

HHS- hyperglycemic hyperosmolar state

BUN –blood urea nitrogen

*Acetest and Ketostix (Bayer; Leverkusen, Germany) measure acetoacetic acid only; thus, misleadingly low values may be obtained because the majority of “ketone bodies” are β-hydroxybutyrate.

 

DIAGNOSTIC TESTS NEEDED

 

Initial Necessary Tests

 

Basic metabolic panel, osmolality, ketones, β-hydroxybutyrate (β-OH), complete blood count with differential, urinalysis and urine ketones by dipstick, and arterial blood gases.

 

Additional Tests

 

Electrocardiogram, chest X-ray, and various tissue cultures, if indicated, and HbA1c.

 

Caveats to Diagnostic Tests

 

Anion gap acidosis is calculated by subtracting the sum of Cl and HCO3 from measured (not corrected) Na concentration and should be corrected for hypoalbuminemia. Usually, a HCO3 level of 18-20 meq/L rules out metabolic acidosis. Arterial blood gases with pH<7.30 support the diagnosis. β-OHB is early and abundant ketoacid and indicative of ketosis. Acetoacetate but not acetone, is a product of ketone body formation and is measured by a nitroprusside reaction that is widely used but may be negative in the blood in early DKA. Effective serum osmolality can be measured directly or derived from following formula: 2 x [measured Na+(meq/L)] + glucose/18. High measured Na indicates a significant degree of dehydration. A white blood cell count >25,000 should warrant a comprehensive search for infection. Serum creatinine can be falsely elevated because of acetoacetate interference with the colorimetric creatinine assay. When patients with DKA present with mixed acid-base disorder, measurement of serum β-OHB will be required to confirm that acidosis is due to ketoacidosis.

 

THERAPY 

 

The therapeutic goals of management include optimization of:

  • volume status,
  • hyperglycemia and ketosis/acidosis,
  • electrolyte abnormalities,
  • potential precipitating factors.

 

Steps to follow in early stages of DKA management (Figures 1, 2, 3):

  • Start IV fluids after blood sample for biochemistry was sent to laboratory (Fig. 1);
  • Potassium level should be >3.3 meq/L before initiation of insulin therapy (supplement potassium intravenously if needed) (Fig. 3);
  • Initiate insulin therapy only when steps 1-2 are executed (Fig. 2).

 

Resolution of DKA:

  • Plasma glucose <200-250 mg/dl,
  • Serum bicarbonate concentration >18 meq/L,
  • Venous blood pH >7.3, and
  • Anion gap <10

 

Fluid therapy: Replace fluid deficit in DKA (~6 L) within 24-36 hours with the goal of 50% volume replacement within first 12 hours.

 

Insulin Therapy: Transition to subcutaneous insulin by giving long-acting insulin 2 hours before the discontinuation of IV insulin.

 

Bicarbonate therapy: If pH is < 7.0 or bicarbonate level is < 5 meq/L, administer 100 mmol (2 ampules) of bicarbonate in 200 ml of water with 20 meq of potassium chloride over two hours.

Figure 1. Fluid management in adult patients with DKA

 

Figure 2. Insulin management in adult patients with DKA

Figure 3. Potassium management in adult patients with DKA

 

FOLLOW UP: COMPLICATIONS AND DISCHARGE

 

Hypoglycemia and hypokalemia are the most frequent complications and can be prevented by timely adjustment of insulin dose and frequent monitoring of potassium levels.

 

Non-anion gap hyperchloremic acidosis occurs due to urinary loss of ketoanions which are needed for bicarbonate regeneration and preferential re-absorption of chloride in proximal renal tubule secondary to intensive administration of chloride-containing fluids and low plasma bicarbonate. The acidosis usually resolves and should not affect treatment course.

 

Cerebral edema is reported in young adult patients. This condition is manifested by appearance of headache, lethargy, papillary changes, or seizures. Mortality is up to 70%. Mannitol infusion and mechanical ventilation should be used to treat this condition.

 

Rhabdomyolysis is another possible complication due to hyperosmolality and hypoperfusion.

 

Pulmonary edema can develop from excessive fluid replacement in patients with CKD or CHF.

 

Discharge planning should include diabetes education, selection of appropriate insulin regimen that is understood and afforded by the patient, and preparation of set of supplies for the initial insulin administration at home.

 

REFERENCES

 

Kitabchi AE, Umpierrez GE, Fisher JN, Murphy MB, and Stentz FB. Thirty years of personal experience in hyperglycemic crises: diabetic ketoacidosis and hyperglycemic hyperosmolar state. The Journal of clinical endocrinology and metabolism 93: 1541-1552, 2008.http://www.ncbi.nlm.nih.gov/pubmed/18270259

 

Kitabchi AE, Umpierrez GE, Miles JM, and Fisher JN. Hyperglycemic crises in adult patients with diabetes. Diabetes care 32: 1335-1343, 2009.

 

Karslioglu French E, Donihi AC, Korytkowski MT. Diabetic ketoacidosis and hyperosmolar hyperglycemic syndrome: review of acute decompensated diabetes in adult patients.

BMJ. 2019 May 29;365:l1114.

 

Gosmanov AR, Gosmanova EO, Kitabchi AE. Hyperglycemic Crises: Diabetic Ketoacidosis and Hyperglycemic Hyperosmolar State. 2021 May 9. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dhatariya K, Dungan K, Hershman JM, Hofland J, Kalra S, Kaltsas G, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrère B, Levy M, McGee EA, McLachlan R, Morley JE, New M, Purnell J, Sahay R, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000–. PMID: 25905280

Diabetes Management During Ramadan

ABSTRACT

Muslims contribute to 25% of the world population and majority of them reside in the diabetes and obesity endemic Asia-Pacific region. Fasting during Ramadan is one of the five pillars of Islam and an obligatory duty for all healthy adolescents and adult Muslims. However, Islam exempts the ill and pregnant women from fasting. Despite this, many individuals with diabetes who are at high risk from fasting, fast during Ramadan. Individuals fasting during Ramadan are less likely to see their physicians before starting the fast and more likely to fast against medical advice. Hence, these individuals are at increased risk of hyperglycemia, hypoglycemia, and cardiovascular and renal complications. Management of diabetes during Ramadan needs a comprehensive and integrated planning and dissemination of knowledge through the healthcare providers and Muslim religious leaders. Diabetes care should start in the pre-Ramadan period, continue through Ramadan, and follow-up in the post-Ramadan period.

 

INTRODUCTION

 

Muslims contribute to approximately 25% of world population and are distributed across >200 countries across the globe (1,2). Of this, 61.7% of Muslims live in Asia-Pacific region, which is also a region experiencing the diabetes epidemic (3,4).

 

Ramadan is the 9th holy month of the Islamic lunar calendar. Fasting during Ramadan is one of the five pillars of Islam (5). Fasting during Ramadan is an obligatory duty for all healthy adolescents and adult Muslims aimed at spiritual and holistic wellbeing of the individual (1,5). The Holy Quran exempts the sick, medically unfit, or those traveling from fasting during the holy month (1,5).

 

 

Fasting during Ramadan involves complete abstinence from food, medication, drink (including water), or any other form of nutrition (including via a percutaneous endoscopic gastrostomy tube) from dawn to sunset (1, 2, 6).  The fasting during Ramadan is a type of intermittent fasting as it is observed for 10–21 hours depending on the geographical location and solar season and is observed daily for 29–30 consecutive days (1, 2). Individuals fasting during Ramadan take two main meals, Suhoor (pre-dawn meal) and Iftar (post sunset meal) and eat nothing from sunrise to sunset (1, 2, 6).

 

It is estimated that about 79% of Muslims with type 2 diabetes (T2D) and about 43% of them with type 1 diabetes (T1D) fast during Ramadan (7). Of those who fast during Ramadan, 64% fasted every day, and 94.2% fasted for at least 15 days (8). The medication timings of these individuals with diabetes need to be adjusted to pre-dawn and post-sunset timings (1,2). Also, many of these individuals fast against medical advice (9, 10).

 

Since a huge proportion of individuals with diabetes fast during Ramadan, and many are at risk due to fasting, management of diabetes during Ramadan and proper fasting guidance is critical (1,2). 

 

EFFECTS OF FASTING DURING RAMADAM

 

Physiological Changes

 

Fasting during Ramadan is associated with a number of physiological changes.

 

CHANGES IN FEEDING PATTERNS AND ENERGY INTAKE

 

Ramadan fasting differs from other forms of fasting as there is no consumption of any food or drink between dawn and sunset. Hence, the timing between the meals is very long, and this disrupts the normal physiology with disruption in the normal rhythm and fluctuations seen in various homeostasis and endocrine processes (Figure 1). Major changes occur in glucose homeostasis in individuals with diabetes that results in post Iftar hyperglycemia and risk of hypoglycemia during the day (Figures 2 and 3)

Figure 1. Changes in feeding patterns and energy intake during various fasting periods (11, 12). (I) normal feeding, (II) Ramadan fasting and (III) prolonged fasting and starvation.

 Figure 2. Mean continuous glucose monitoring (CGM) profiles from healthy individuals (12, 13).

Figure 3. Mean continuous glucose monitoring (CGM) profiles from individuals with diabetes fasting during Ramadan (12, 13).

DECREASE IN TOTAL SLEEP TIME

Total sleep time decreases by approximately 1 hour, with a decrease in sleep period time, rapid eye movement (REM) sleep proportion and duration. Additionally, delayed sleep and an increase in non-REM sleep proportion, sleep latency, and daytime sleepiness by1-point on the Epworth sleepiness scale is also observed (ESS) (12).

ALTERATION OF CIRCADIAN RHYTHM AND HORMONE LEVELS

Sudden alteration of circadian rhythm and hormone levels occurs due to sudden changes in sleep and wake cycles and feeding patterns. Fasting can induce epigenetic changes in genes that control the circadian rhythm (12). The change in circadian rhythm triggers many catametabolic changes, alteration in temperature, and changes in the normal rhythm of hormones like insulin, glucagon, leptin, ghrelin, cortisol, melatonin, growth hormone, and testosterone (12,14). The various changes seen are:

 

  • Insulin resistance and increased glucagon levels: excessive glycogen breakdown and increased gluconeogenesis
  • Cortisol circadian rhythm shows a shift with a blunting of the morning to evening ratio. However, serum cortisol levels do not change by end of Ramadan month.
  • Morning adiponectin levels are reduced
  • Morning and evening growth hormone levels are reduced
  • Large increases in morning leptin levels
  • No major shifts in diurnal ghrelin level

 

By the end of Ramadan significant decrease in serum levels of ghrelin, leptin, and melatonin are observed along with modest reductions in testosterone in men.

SHIFT IN FLUID BALANCE

A sudden shift in fluid balance is seen because of an absolute restriction of fluid intake between dawn and sunset. This may precipitate dehydration in a hot climate which may in turn cause hypotension and falls (6). Uncontrolled hyperglycemia can exacerbate the dehydration due to an osmotic diuresis (6). Dehydration in individuals with T2D can present as low blood pressure, lethargy, or syncope. Dehydration can also increase the risk of thrombosis and stroke due to hemoconcentration and hypercoagulability (6). Other fluid related changes are not considered a major cause of concern and include higher fluid and total water intake between sunset and dawn; urine osmolality increases significantly in the afternoon to conserve water and reduce urine output (12).

ALTERED ENERGY BALANCE

Altered energy balance is seen due to a sudden increase in food intake at Iftar. During Ramadan there is a reduction in activity and energy expenditure which is offset by the reduced time spent during sleep (12).

GUT MICROBIOTA

Intermittent fasting during Ramadan can have direct impact on the gut microbiota which could lead to positive changes in health (12).

 LIPID CHANGES

Fasting during Ramadan has been shown to be associated with a significant increase in high-density lipoprotein-cholesterol (HDL-C) and a significant decrease in total triglycerides, total cholesterol, and low-density lipoprotein-cholesterol (LDL-C) (6, 15).

Physical and Mental Wellbeing

Fasting during Ramadan can have both positive and negative effects on the physical and mental wellbeing of the individuals (Table 1) (16).

Table 1. Positive and Negative Effects on Physical and Mental Wellbeing of Individuals Fasting During Ramadan (16)

Positive benefits

Negative effects

Sense of fulfilment

Sleep deprivation and disruption of circadian rhythm leading to an increase in fatigue and reduction in cognition

More lethargy 

Improvements in:

Weight and BMI

Self-control and ability to resist temptations

Glucose excursions causing feelings of being unwell

Greater sense of:

Empathy for less fortunate

Community

Fostering relationships

Heightened feelings of fear for diabetes related complications

Participation in Sunnah practices for greater spiritual benefits

Temporary changes in weight

Reducing potentially harmful vices, such as smoking, for greater physical and mental wellbeing

Short term feelings of stress anxiety, irritability, and agitation

BMI- body mass index

 

The month long fasting during Ramadan has been associated with significant reduction in weight, waist circumference, and fat mass, especially in those who are overweight or obese (15, 17).

 RISKS OF FASTING DURING RAMADAN IN INDIVIDUALS WITH DIABETES

The various risks of fasting in individuals with diabetes who fast during Ramadan are:

  • Hyperglycemia
  • Hypoglycemia
  • Macrovascular: Cardiovascular disease (CVD) including stroke
  • Microvascular: Chronic kidney disease (CKD)
  • Dehydration

Dual Risk of Hyperglycemia and Hypoglycemia

In people with diabetes fasting during Ramadan there can be an increase in glucose variability and therefore there is increased risk of both hyperglycemia and hypoglycemia (12).

HYPERGLYCEMIA

The meals at Iftar are calorie dense and can cause a significant and rapid rise in blood glucose (BG) levels in people with diabetes (12). The EPIDIAR study showed that the hospitalization rate for severe hyperglycemia during Ramadan increased significantly in individuals with T2D (P<0.001). The hospitalization rate for severe hyperglycemia (with or without ketoacidosis) during Ramadan increased insignificantly for individuals with T1D (P = 0.1635) (6,7).

HYPOGLYCEMIA

The CREED study showed that hypoglycemia incidence before Ramadan was associated with significantly increased risk of hypoglycemia during Ramadan (18). This association between hypoglycemia incidence before and during Ramadan has been seen through multiple studies across continents (1,18,19). Similarly, the EPIDAR study7 showed that T1D and T2D patients had a 4.7-fold and a 7.5-fold increase, respectively, in severe hypoglycemia requiring hospitalization during Ramadan. Hypoglycemia during Ramadan was significantly associated with the use of sulfonylureas and insulin (18,19). Severe and non-severe hypoglycemia rates are fewer with second-generation sulfonylureas, Glucagon-like peptide-1 receptor agonists (GLP-1 RAs), insulin analogues, and sodium-glucose transporter-2 (SGLT-2) inhibitors (20-25). During the early Ramadan period, patients on sulfonylureas and those on ≥2 antidiabetic medications have significant increase in mean amplitude of glycemic excursions (26).

 

Other factors influencing the incidence of hypoglycemia during Ramadan include season, geographical location, fasting duration, time since diagnosis, gender, anthropometric measures, dietary behaviors, and pre-fasting education (1). 

Macrovascular (Cardiovascular and Cerebrovascular) and Microvascular Risk (Renal Complications)

Diabetes increases the risk of CVD and stroke, and individuals with diabetes who have pre-existing CVD or stroke are at greater risk of complications when fasting (27). Individuals with unstable CVD or stroke are also at very high risk from fasting. Individuals with diabetes and CKD stage 3 are at high-risk from fasting while those with stage 4-5 are at very high risk from fasting during Ramadan (27). Patients on dialysis or those who had a kidney transplant are also considered high risk from fasting (27).

 

Factors associated with increased risk of fasting during Ramadan are high carbohydrate intake, inadequate hydration, high activity levels, poor sleeping patterns, and missing doses of essential medicines (27).

 

All these high to very high-risk individuals should be discouraged from fasting. If they still decide to fast, then pre-Ramadan assessment and education and monitoring during Ramadan and post-Ramadan should be carried out under the expert guidance of a multidisciplinary team (diabetologist, cardiologist, neurologist, nephrologist, nutritionist etc.) (27). Weekly monitoring during Ramadan by a health care provider should be encouraged.

 

Pre-Ramadan assessment and education should begin three months prior to Ramadan and all efforts should be made to stabilize the doses of the various drugs, adjust them to morning and evening dose, and those on insulin should be taught self-titration of dose based on SMBG (27).

Patients with CKD, on dialysis or those who had a kidney transplant should be encouraged to routinely monitor electrolytes and creatinine at various time-points during Ramadan (27). A diet rich in potassium and phosphorus should be avoided (27).

 MANAGEMENT OF DIABETES DURING RAMADAN

5 R's of Ramadan Care

 

The management of diabetes during Ramadan and in general can be summarized under the mnemonic termed as the 5 R's of Ramadan care (Table 2).

 

Table 2. The 5 R's of Ramadan Care (28)

The 5 R

Significance

Respect

Respect the patient's attitudes, wishes, and needs, and consider these while planning therapy

Speak with the patient with empathy for his religious beliefs

Risk stratification

This is an essential backbone for pre-Ramadan counseling

Revision of Therapy

Diabetes therapy will need to be revised based on risk of hyperglycemia and hypoglycemia, and other risk factors

Regular Follow Up

Regular follow-up with HCP before, during and after Ramadan is necessary to ensure a safe and healthy fasting experience

Reappraisal of Strategy

Diabetes is a dynamic condition, and constant reappraisal is required during the current and next fasting period

HCP- health care provider

Pre-Ramadan Management of Diabetes

DM management in people planning to fast during Ramadan should ideally start six to eight or maximum 12 weeks before the first day of fasting (6, 29). Diabetes assessment and plan of care pre-Ramadan should ideally follow the flow chart shown in Figure 4 (6, 29).

Figure 4. Diabetes assessment and plan of care pre-, during and post Ramadan (6, 29)

 

Patient education during this time-period is necessary because many patients follow a self-management approach of diabetes during Ramadan and do not appreciate the risks and implications of fasting on DM and its medications, and fast against medical advice (6, 19). Physicians need to be sensitized about this time-period for their Muslim patients as many may not realize the religious sensitivities associated with DM management during Ramadan (6).

PRE-RAMADAN EDUCATION

The pre-Ramadan diabetes education should cover:

Risk quantification, exemptions, and removing misconceptions

  • Blood glucose monitoring
  • Fluids and dietary advice
  • Physical activity and exercise advice
  • Medication adjustment
  • When to break the fast
  • Recognition of hypoglycemia and hyperglycemia symptoms

RISK STRATIFICATION OF PEOPLE WITH DIABETES

The pre-Ramadan time period should be used to understand the individual’s risks associated with fasting, and develop an individualized treatment plan for the individual who falls in the lower risk category and can fast. The risk stratification is done based on several factors (6, 9, 10):

  • Type and duration of diabetes
  • DM treatment regime and polypharmacy with multiple glucose lowering drugs
  • Level of glycemic control
  • Risk or occurrence of hypoglycemia
  • DM self-management capability including hypoglycemia awareness, motivation for self-monitoring blood glucose (SMBG), frailty, and cognition
  • DM complications and comorbidities
  • Ongoing/recent severe illness
  • Renal impairment
  • Social determinants affecting assess and adherence to treatment including economic and education level
  • For those with T1D: access to continuous glucose monitoring (CGM) and advanced insulin technologies

 

The IDF-DAR Practical Guidelines 2021 stratify the individuals with DM who are going to fast during Ramadan into three risk categories, low, moderate and high (Figure 5) (29).

Figure 5. IDF-DAR 2021 Ramadan risk score and risk categories for fasting (2, 30)

 

DM patients who had a history of severe hypoglycemia ≤3 months before Ramadan, recurrent hypoglycemia, or of hypoglycemia unawareness are considered high risk (31). Young individuals and adolescents start fasting during Ramadan with the onset of puberty, and those with T1D are considered to be high risk for fasting and generally discouraged from fasting (9).

 

Many DM patients may need to be upgraded to high-risk category during the current coronavirus-2019 (COVID-19) pandemic and thus likely to fall under the ‘advised not to fast’ risk category (6).

 

Patients with high-risk scores are advised not to fast as fasting is not considered safe for these individuals (29). However, since fasting during Ramadan is a personal choice, the individuals deciding to fast despite being cautioned against it, should be monitored very closely during and after Ramadan (6, 19).

 

Individuals with low and moderate risk scores are educated about the risks and advised to fast with strict BG monitoring, and adjustments to diet/nutrition and medications (29). The individuals are closely monitored during Ramadan by their health care providers (HCPs) and advised to come for a post Ramadan follow-up. At this follow-up visit, the HCPs asses the glycemic control, and discuss the challenges faced to make the next Ramadan fasting more risk free. Pre-Ramadan risk assessment, education and advise is known to improve the fasting experience of individuals with DM (6).

Blood Glucose Monitoring

 

Self-monitoring blood glucose (SMBG) should be stressed upon and encouraged. Moderate to low-risk individuals with diabetes can monitor their BG once or twice daily but those at high risk of fasting should be encouraged to follow a 7-time-point guide for SMBG during Ramadan (Figure 6) (29). Additional BG check should be done if the individual experiences symptoms of hypoglycemia, hyperglycemia, or feel unwell (29).

 

Figure 6. A seven time-point blood glucose monitoring guide for people with diabetes fasting during Ramadan (29)

Fluids and Dietary Advice

Detailed diet plan for Ramadan is provided through Ramadan specific medical nutrition therapy (MNT) and Ramadan Nutrition Plan (RNP) (29,32). Adequate fluid intake should be encouraged between sunset and dawn.

Physical Activity and Exercise Advice

Individuals with diabetes who are fasting should be encouraged to carry out their normal physical activity (29). Taraweeh prayers, which involve activities such as bowing, kneeling, and rising, can be considered as part of daily exercise activities (29). Rigorous exercise/activity during the fasting period should be avoided as this can increase the risk of hypoglycemia and dehydration, especially during the last few hours of fasting (29).

Medication Adjustment

All medications (diabetes and non-diabetes) should be reviewed in the pre-Ramadan period to see which medications need a dose and time adjustment. The change should be made well before Ramadan and monitored through appropriate clinical and laboratory evaluation (29). Patients on insulin should be taught self-titration of units based on SMBG values (10).

When to Break the Fast

This is an essential component of Pre-Ramadan education. All individuals with diabetes who are fasting during Ramadan should be advised to break their fast if:

  • Blood glucose <70 mg/dL (3.9 mmol/L)
  • Advise to re-check within 1 hour if BG is between 70–90 mg/dL (3.9–5.0 mmol/L)
  • Blood glucose >300 mg/dL (16.6mmol/L)
  • Symptoms of hypoglycemia, hyperglycemia, dehydration or acute illness occur

Recognizing Symptoms of Hypoglycemia and Hyperglycemia

All individuals with diabetes who are fasting during Ramadan and their caregivers should be taught to recognize symptoms of hypoglycemia and hyperglycemia (Figure 7) (29). If they recognize these symptoms, they should be advised to break their fast.

Figure 7. Symptoms of hypoglycemia and hyperglycemia (29)

 The Medico-Religious Interplay in Ramadan

Muslims believe that Ramadan is a blessed month, and see fasting during the holy month of Ramadan as a deeply meaningful and spiritual experience (1, 30). A significant number of individuals with diabetes fast during Ramadan, even against medical advice and despite the religious exemptions available to the sick (1, 30, 33). This population also includes adolescents with T1D, who fast against medical advice (9). Individuals with diabetes who fast during Ramadan are more likely to avoid consulting their doctors (12).

 

International Diabetes Federation (IDF) and Diabetes and Ramadan (DAR) International Alliance collaborated to form the IDF-DAR Practical Guidelines 2021 to help healthcare providers (HCPs) better manage diabetes in patients fasting during the month-long holy period of Ramadan (2).

 

It is important to make these individuals with diabetes who cannot fast due to their medical condition understand that they are equally blessed even if they do not fast (30). Many individuals with diabetes who fast prefer to take fasting related advice from their holy leader (Imam). Hence, the right message and education should be disseminated by both the HCPs and the religious leaders (Table 3) (30). HCPs should avoid medical jargons, and counsel patients from a religious standpoint; and religious leaders should integrate into their counseling the value and significance of exemptions in context with the medical advice.

 

Table 3. Medical and Religious Risk Score Recommendations (30)

Risk score

Medical recommendations

Religious recommendations

LOW RISK

0-3 points

Fasting is probably safe. Ensure

1. Medical Evaluation

2. Medication adjustment

3. Strict monitoring

1. Fasting is obligatory

2. Advice not to fast is not allowed except if patient

is unable to fast due to:

3.     -  Physical burden of fasting

4.     -  Has to take medication or food or drink during the fasting hours on medical advice

MODERATE RISK

3.5-6 points

Fasting safety is uncertain

Ensure:

1. Medical Evaluation

2. Medication adjustment

3. Strict monitoring

1. Fasting is preferred but patients may choose

not to fast if they are concerned about their

health after consulting the doctor and taking

into account the full medical circumstances

and patient’s own previous experiences

2. If the patient does fast, they must follow

medical recommendations including regular blood glucose monitoring

HIGH RISK

>6 points

Fasting is probably unsafe

Advise against fasting

 

Medical Nutrition Therapy (MNT) and Ramadan Nutrition Plan (RNP) for People with Diabetes

MNT is an essential component of diabetes management and includes both meal plans and diabetes education, aimed at improving lifestyle and diabetes related behavior (4). MNT helps achieve the desired glycemic control and helps the overweight and obese individuals with T2D improve their lifestyle and lose weight (4). MNT should be appropriate and accurate for the patient’s age, comorbidities, lifestyle requirements, and other medical needs. MNT should be easily absorbed; affordable, easily accessible, acceptable (through right aroma and consider taste preferences), and attractive (visually appealing) (4). This improves adherence to the MNT (4).

 

An MNT plan for individuals with diabetes is essential for safe fasting during Ramadan (32). Structured Ramadan-specific MNT (34) has shown to improve fasting BG and triglyceride levels and pre-dawn and pre-bed SMBG values compared to patients with T2D receiving standard care (34).

 

Structured Ramadan-specific MNT includes (32, 34):

  1. Pre-Ramadan nutrition education
  2. Individualized energy and balanced macronutrient prescriptions for non-fasting period (sunset to sunrise) to prevent hypoglycemia during fasting state
  3. Well distributed carbohydrate intake to prevent post meal hyperglycemia
  4. At least one serving/day of diabetes-specific formula to be taken during Suhoor and/or pre-bed snack.
  5. Diet plan should consider other comorbidities.
  6. Ramadan toolkits:
  • Ramadan flip chart
  • 14-day menu plan
  • Ramadan Nutrition Plate (RNP)
  • Festive season nutrition plan (Syawal nutrition plan)

 

RNP “is a mobile and web-based application designed to help healthcare professionals (HCPs) individualize medical nutrition therapy (MNT) for people with diabetes” who are fasting during Ramadan (32). A well designed and customized RNP is a prerequisite to safe and confident fasting during Ramadan (32). Apart from nutrition, the platform also provides education regarding safe fasting during Ramadan. It helps individuals to safely fast who have no access to HCPs during Ramadan. Several RNPs have been developed for different countries to suit their regional customs, beliefs, and preferences. HCPs can use their country specific RNP, Ramadan Nutrition plate, and well-balanced meal (Table 4) (4, 32) as a guide to individualize the MNT during Ramadan (32).

 

Table 4. Macronutrient Meal Composition for Ramadan (4, 32)

Macronutrient

Recommended amount

Recommended sources

Sources not recommended

Carbohydrate

•                ≤130 g/day

•                Accounts for 40-45% of total caloric intake

•                Adjust as per cultural setting and individual preferences

Low glycemic index and glycemic load carbohydrates: whole grains, legumes, pulses, temperate fruits, green salad, and most vegetables

Foods rich in sugar, refined carbohydrate, processed grains, or starchy foods: sugary beverages, traditional desserts, white rice, white bread, low fiber cereal, and white potatoes

Meal

Calorie%

Carbohydrate exchange*

Suhoor

30-40

3-5

Iftar snack

10-20

1-2

Iftar meal

40-50

3-6

Healthy snack (if required)

10-20

1-2

Fiber

20-35g/day (or 14g /1000 kcal)

High fiber foods: unprocessed food, vegetables, fruits, seeds, pulses, and legumes

 

Fiber helps to provide satiety during Iftar and to delay hunger after Suhoor

-

Protein

•                ≥1.2g/kg of adjusted body weight

•                Accounts for 20-30% of the total caloric intake.

•                Protein enhances satiety and gives sensation of fullness. Also helps to maintain lean body mass

•                Fish, skinless poultry, milk and dairy products, nuts, seeds, and legumes (beans),

•                low fat milk and milk products

•                Protein with a high saturated fat content such as red meat (beef, lamb) and processed meats (increase CVD risk)

Lipids

•                Between 30–35% of the total calorie intake.

•                The type of fat is more important than the total amount of fat in reducing CVD risk.

•                Limit saturated fat to < 7%. PUFA and MUFA should comprise the rest of the fat intake.

• Limit dietary cholesterol to < 300 mg/day or < 200 mg /day if LDL cholesterol > 2.6 mmol/L

•                Consume fat from PUFA and MUFA (e.g., olive oil, vegetable oil, or blended oil (PUFA and Palm oil)). Oily fish (e.g., such as tuna, sardines, salmon, and mackerel) as a source of omega 3-fatty acids

•                Minimize saturated fat, including red meat (beef and lamb), ghee, and foods high in trans-fats (e.g., fast foods, cookies, some margarines).

* 1 Carbohydrate exchange = 15 g Carbohydrates; CVD, cardiovascular disease; MUFA, Monounsaturated fatty acids; PUFA, Polyunsaturated fatty acids

 Medical Management of Diabetes During Ramadan

Diabetes assessment and plan of care during and post Ramadan should ideally follow the flow chart shown in Figure 4 (6,29).

MEDICAL MANAGEMENT OF T1D DURING RAMADAN IN ADOLESCENTS AND YOUNG INDIVIDUALS

TID is treated with insulin replacement therapy. After the Pre-Ramadan risk stratification, adjustments are made to the patient’s dosing, timing, and type of insulin regime based on the patient’s risk level.

Insulin Regimens

There is no conclusive evidence supporting efficacy and safety of a particular insulin regime over another in adolescents with T1D who are fasting during Ramadan. The insulin regime is therefore based on affordability, access to treatment (medication, specialist and advanced technology), and cultural preferences (9).

 

Changing the insulin regime just before Ramdan is likely to result in dose errors and increase the risk of hypoglycemia. Hence, every effort should be made to continue the same regime, but with proper dose modifications and comprehensive counseling covering diet, lifestyle, physical activity, SMBG, and self-titration of insulin dose (10).

 

The most commonly used insulin regimens in adolescents are (9):

  1. Basal-bolus regimens –multiple dose injections (MDI) adjusted according to meal (preferred option)
  2. Conventional twice daily neutral protamine Hagedorn (NPH)/regular short acting (human) insulin
  3. Continuous subcutaneous insulin infusion (CSII) with or without sensors
  4. Premixed insulins (generally not recommended for T1DM)

 

Of these, MDI and CSII are closer to providing the physiological insulin secretion pattern.

Table 5 gives guidance on dose modifications of different insulin regimes.9 SMBG should be encouraged and the patients or their caregivers taught to self-titrate the insulin dose based on the BG levels (Table 6) (10).

 

Table 5. Insulin Dose Adjustments During Ramadan (10,35)

Insulin

Dose modification

Timing

Glucose monitoring

MDI (basal bolus) with analogue insulins

Basal insulin

30-40% dose reduction

Take at Iftar

5–7-point glucose monitoring*

MDI (basal bolus) with analogue insulins

RAI

Suhoor dose reduced 30-50%

Skip pre-lunch dose Iftar dose to be adjusted according to the 2hr post Iftar BG levels

Take at Iftar and Suhoor

5–7-point glucose monitoring*

MDI (basal bolus) with conventional insulins

NPH insulin

No dose modification at Iftar

50% dose reduction at Suhoor

Take at Iftar and Suhoor

5–7-point glucose monitoring or 2-3 staggered readings throughout the day*

MDI (basal bolus) with conventional insulins

Regular insulin

Suhoor dose reduced by 50%

Skip pre-lunch dose

Iftar dose unchanged unless needs to be adjusted according to the 2hr post Iftar BG levels

Take at Iftar and Suhoor

7-point glucose monitoring or 2-3 staggered readings throughout the day*

Premixed (analogue or conventional) once daily

No dose modification

Take at Iftar

At least 2-3 daily

Readings*

Premixed (analogue or conventional) twice daily

No dose modification at Iftar

50% dose reduction at Suhoor

Take at Iftar and Suhoor

At least 2-3 daily

Readings*

CSII / Insulin Pump

Basal rate adjustment

10-30% increase for the initial few hours of Iftar

20-40% decrease for the final 3-4 hours of fast

Bolus doses

Same ICR and ISF principles as followed prior to Ramadan

Reduce the dose post-Suhoor by 20%

CGM

* And whenever any symptoms of hypoglycemia/hyperglycemia develop or feeling unwell

ICR- Insulin Carbohydrate Ratio; ISF- Insulin Sensitivity Factor; RAI- rapid analogue insulin

 

Table 6. SMBG Guided Dose Titrations for Different Types of Insulin During Ramadan (10)

Fasting/pre-Iftar/pre-Suhoor blood glucose

Basal insulin

Short-acting insulin

Premixed insulin

pre-Iftar

pre-Iftar*/post-Suhoor**

pre-Iftar insulin modification

<70 mg/dL (3.9 mmol/L) or symptoms

Reduce by 4 units

Reduce by 4 units

Reduce by 4 units

<90 mg/dL (5.0 mmol/L)

Reduce by 2 units

Reduce by 2 units

Reduce by 2 units

90-126 mg/dL (5.0-7.0 mmol/L)

No change

No change

No change

>126 mg/dL (7.0 mmol/L)

Increase by 2 units

Increase by 2 units

Increase by 2 units

>200 mg/dL (16.7 mmol/L)

Increase by 4 units

Increase by 4 units

Increase by 4 units

*Reduce the insulin dose taken before Suhoor; **Reduce the insulin dose taken before Iftar

Recommendations for Insulin Regimes
  1. T1D management during Ramadan should be individualized according to patient’s need, preference, affordability, acceptability, and access to treatment (9).
  2. The basal-bolus regime is the preferred regime and consists of a long-acting insulin analogue (basal insulin) and a premeal rapid acting insulin analogue (bolus insulin) (9).
  3. Associated with a lower risk of hypoglycemia when compared to conventional, twice-daily, insulin regimens
  4. The bolus insulin dose should be dependent on the carbohydrate count of the meal. It should ideally be given 20 minutes before the meal for better post-prandial BG control.
  5. Boluses covering Suhoor and Iftar should be based on Insulin Carbohydrate Ratio (ICRs) and Insulin Sensitivity Factor (ISFs)

 

Approximately 70% of hypoglycemia occur during the last six hours of fasting. Hence, the type of basal insulin used, reduction in basal insulin dose and modification of insulin timing are the tools used to avoid hypoglycemia:

  • Ramadan fasting should be started with a reduction of basal insulin-starting with 20% and individualizing up to 40% as required
  • Basal insulin can be administered earlier in the day to minimize insulin exposure during the last few hours of fasting when BG levels are low.
  • Basal insulin can also be taken at Iftar or earlier in the evening
  • First-generation basal insulin analogues (such as glargine U-100) are more likely to cause hypoglycemia than the second generation, long-acting insulin analogues (glargine U-300 and degludec). However, the choice of insulin should be individualized based on risk of hypoglycemia.
  • Long-acting insulin analogues are preferred over intermediate acting (NPH/human insulin) as they provide a steady fall of BG towards normal levels by sunset time (9).
  • Twice daily regimens are more likely to be associated with hyperglycemia (9).
  • Twice daily regimens are usually not preferred during Ramadan, but if they are the only choice available to the patient, their timing and dose needs to be more closely monitored depending on the timing, portion size and carbohydrate content of meal (9).
  • Premixed twice daily insulin regimes are not recommended during Ramadan period as they require fixed carbohydrate intake at fixed timing, and this may be difficult for adolescents to follow (9).
  • There is emerging evidence that T1D patients can fast during Ramadan with fewer complications with the help of newer technologies such as insulin pump therapy, CGM and hybrid closed-loop systems (6).
  • CSII with insulin pumps in adolescents help achieve the targeted glycemic control with reduction in hyperglycemia and severe hypoglycemia, and provides more flexibility, improved quality of life and decreased risk of complications like diabetic ketoacidosis (9).
  • CSII allows for easier management of DM and reduces risk of complications than MDI (9)
  • The basal and bolus doses are adjusted through algorithms on the pump or through sensors and mobile applications (in more advanced versions)
  • Basal insulin is reduced by 20-35% in the last 4-5 hours before Iftar and increased by 10-30% after Iftar up to midnight
  • Prandial insulin bolus calculation is based on usual ICR and ISR
  • Bolus doses can be delivered in three different ways:
  • Standard dosing: Immediately before meals
  • Extended or square dosing: gradual dosing over a certain time period
  • Combo or dual wave bolus: combination of standard and extended
  • High fat content diet as seen during Iftar is likely to benefit from extended or combo bolus dosing (9).
  • Insulin pumps augmented with CGM provide better glycemic control and reduce complications considerably in adolescents with T1D. These sensor-augmented pumps are of two types (9):
  • Low Glucose Suspend (LGS) function pumps: The high-risk BG threshold for HE is pre-set in these pumps. The insulin administration can be automatically suspended for ≤2 hours when sensors detect BG levels below the pre-set threshold
  • Predictive Low Glucose Suspend (PLGS) pumps: Insulin administration is automatically suspended before BG reaches hypoglycemic levels (70 mg/dL [3.9 mmol/L]).
  • Automated insulin delivery (closed loop): These can suspend or increase insulin delivery based on sensor-based BG values. Thus, closed loops help increase time in range (TIR) and minimize hypoglycemia and hyperglycemia.
  • Types: Hybrid closed-loop automated insulin delivery systems; Do-It-Yourself Artificial Pancreas Systems (DIY APS)
  • However, CSII is a costly technology, has limited access in many countries, and therefore is not widely available due to cost and accessibility constraints (9).

MEDICAL MANAGEMENT OF ADULTS WITH T1D DURING RAMADAN  

Patients advised to self-monitor BG at 7-time-point points: when fasting; post-breakfast; pre-lunch; post-lunch; pre-dinner; post dinner; and midnight (9).

 

Dose adjustments for the different insulin regimes should start during the pre-Ramadan period and every attempt should be made to attain the desired glycemic goal but at low risk for hypoglycemia.

 

Short acting insulin analogues (glulisine, lispro, or aspart) are associated with less hypoglycemia and better improvement in postprandial glycemia than regular insulin. Premixed insulins are generally not preferred during Ramadan (9).

 

Table 5 provides guidance on dose modifications of different insulin regimes (9). SMBG should be encouraged and the patients taught to self-titrate the insulin dose based on the BG levels (Table 6) (10).

MEDICAL MANAGEMENT OF T2D DURING RAMADAN

Medical management of Ramadan in patients with T2D varies with wide variety of oral and injectable glucose lowering drugs (GLDs) used during Ramadan as shown in Table 9. Patients may be on one or more oral GLDs or a combination of oral and injectable GLDs.

 

Table 9. Different Types of Glucose Lowering Drugs Used by Patients with T2D During Ramadan (1,10)

Oral glucose lowering drugs

Injectable glucose lowering drugs

Sulfonylurea (gliclazide, glipizide, glimepiride, glibenclamide, or glyburide)

Long/intermediate basal insulins (insulin glargine, insulin detemir, insulin degludec or NPH)

 

Insulin: insulin pump, multiple daily injections, insulin lispro, insulin glargine, soluble human insulin, insulin detemir, and biphasic insulin

Biguanides (Metformin)

Bolus prandial rapid or short-acting insulins (lispro, glulisine, aspart or regular human insulin)

Thiazolidinediones (pioglitazone)

Premixed insulins (fixed ratio combinations of short and intermediate acting insulins)-usually not recommended during Ramadan

DPP-4 inhibitors (sitagliptin, saxagliptin, linagliptin, alogliptin, vildagliptin)

GLP-1 RA (lixisenatide, exenatide, liraglutide, dulaglutide, semaglutide)

SGLT2-I (dapagliflozin, canagliflozin, empagliflozin, and ertugliflozin)

Alpha-glucosidase inhibitor (acarbose, voglibose)

Short-acting insulin secretagogues (repaglinide and nateglinide)

Oral GLP-1 RA (semaglutide)

 

  DPP-4, dipeptidyl peptidase 4; GLP-1 RA, Glucagon-like peptide-1 receptor agonists (GLP-1 RAs); NPH, neutral protamine Hagedorn; SGLT2-I, Sodium/glucose cotransporter-2 inhibitors

 

After the Pre-Ramadan risk stratification, adjustments are made to the patient’s GLDs. Some patients may need a change of medications too depending on their risk level. Preference is given to GLDs with better glycemic control and lower risk of hypoglycemia.

Oral Glucose Lowering Drug Adjustments During Ramadan
  • In general, non-sulfonylureas GLDs are superior in lowering hypoglycemia incidence than sulfonylureas (1).
  • Metformin is the most commonly used first line oral GLD, and has minimal risk of hypoglycemia (10).
  • Sulfonylureas are the most commonly used second line oral GLDs after metformin, especially in resource limited settings (10).
  • Short-acting insulin secretagogues can be useful GLDs during Ramadan because of their short duration of action and low risk of hypoglycemia (10).
  • DPP4 inhibitors are well tolerated during fasting and have a low hypoglycemia risk (10).
  • SGLT2 inhibitors are the newest class of oral GLDs used in T2D. They have demonstrated effective glycemic control during Ramadan even in patients with cardiovascular diseases/chronic kidney disease, and have low risk of hypoglycemia (10, 36).
  • An individual should be started on a SGLT2 inhibitor at least 2-4 weeks before Ramadan for the BG levels to stabilize during the fasting time.
  • Of the thiazolidinediones, only pioglitazone is widely approved for T2DM, has low hypoglycemia risk, but clinical data on its use during Ramadan is limited (10).

 

Table 10. Oral Glucose Lowering Drugs Used During Ramadan: Dose Modifications and Timing Adjustments in Individuals with Type 2 Diabetes (10)

Oral GLD

Dose modification

Timing

Metformin once daily

No dose modification

Take at Iftar

Metformin twice daily

No dose modification

Take at Iftar and Suhoor

Metformin thrice daily

No modification to morning dose. Afternoon dose to be combined with evening dose

Take morning dose before Suhoor and evening dose at Iftar

Prolonged release metformin

No dose modification

Take at Iftar

Sulfonylureas once daily

Reduce dose in patients with well controlled BG levels

Take at Iftar

Sulfonylureas twice daily

In patients with well controlled BG levels Iftar dose remains the same. Suhoor dose is reduced

Take at Iftar

Older sulfonylureas (e.g., glibenclamide)

Avoid and replace with 2nd generation SUs such as glicazide, glicazide MR, and glimepiride

Short-acting insulin secretagogues thrice daily dosing

Reduce or re-distribute to two doses

Take before Iftar and Suhoor

DPP4 inhibitor once daily

No dose modification

Take at Iftar

DPP4 inhibitor twice daily (vildagliptin)

No dose modification

Take at Iftar and Suhoor

SGLT2 inhibitors

No dose modification

Take at Iftar

Thiazolidinedione

No dose modification

Take at Iftar

  BG, blood glucose; DPP-4, dipeptidyl peptidase 4; MR, modified release; SGLT2-I, Sodium/glucose cotransporter-2 inhibitors; SU, sulfonylurea

Injectable Glucose Lowering Drug Adjustments During Ramadan
  • Most patients with long-standing T2D eventually need insulin to manage their BG levels. Various insulin regimes are used in T2D (table 9) (1, 10) and in general, the use of insulin increases the risk of hypoglycemia during Ramadan.
  • Insulin can be given as single daily injection, MDI or as CSII through insulin pumps. The insulin regime is therefore based on affordability, access to treatment (medication, specialist and advanced technology), and cultural preferences (9).
  • Changing the insulin regime just before Ramdan is likely to result in dose errors and increase risk of hypoglycemia. Hence, every effort should be made to continue the same regime, but with proper dose modifications and comprehensive counseling covering diet, lifestyle, physical activity, SMBG, and self-titration of insulin dose (10).
  • Table 5 shows the various insulins and how their doses and timing are adjusted during Ramadan.
  • SMBG guided dose titrations for different insulin types are shown in Table 6.
  • GLP-1 RAs can be safely used with other GLDs including metformin and insulin. They have low hypoglycemia risk, but the risk could be higher if given with insulin or sulfonylureas. However, dose needs to be titrated at least 2-4 weeks prior to Ramadan (10).
Individuals on Multiple Antidiabetic Therapy

Individuals on multiple GLDs are at higher risk of hypoglycemia during Ramadan (18). The risk of hypoglycemia is highest if they are on ≥4 GLDs or on a combination of metformin, DPP4I and basal insulin (37).

 

In individuals on multiple GLDs, the risk of hypoglycemia is dependent on several factors such as type and number of GLDs, duration of diabetes, pre-Ramadan glycemic control, renal function, and presence of other comorbidities (10).

 

Individuals on ≥3 GLDs who are fasting during Ramadan should receive comprehensive counseling and advice before the start of Ramadan, and it should cover diet, lifestyle, physical activity, SMBG, and dose and time modifications of GLDs (10).

 

Individuals on a combination of insulin and SUs are at highest risk of hypoglycemia and require a dose reduction GLDs (approximately 25-50% reduction in insulin dose) during Ramadan.

 

Flash glucose monitoring, CGM, activity monitoring, risk stratification, dose adjustments, and use of artificial intelligence-based algorithms that cover one or more of these aspects are the various tools that are likely to help high-risk patients with T2D fast with fewer hypoglycemia and hyperglycemia related complications (6, 10).

MANAGEMENT OF DIABETES IN SPECIAL POPUATIONS DURING RAMADAN

As discussed, individuals who are considered high risk for fasting during Ramadan need special pre-Ramadan risk stratification, counseling, dose modification, and need to follow strict SMBG during Ramadan, and those on insulin should be capable of self-titrating their insulin dose based on their BG values.

 

This is especially true for special population considered high risk due to a high probability of harm caused by fasting during Ramadan (5). This special population of high-risk individuals with diabetes includes pregnant women, elderly, and people with CVD or CKD. All these individuals are usually advised not to fast, but many do decide to fast against medical advice (5).

Management of Diabetes/Gestational Diabetes During Ramadan

Even in healthy pregnant women, fasting during Ramadan results in biochemical changes that almost mimic the effects of prolonged fasting (35). Ramadan fasting results in an increase in triglycerides (TG), free fatty acids (FFA), and ketones in healthy pregnant women along with a decrease in glucose and insulin (35). However, data on physiological and biochemical changes caused by fasting during Ramadan in pregnant women with diabetes is largely lacking.

 

Pregnancy is an exemption from fasting. However, many pregnant women choose to fast during Ramadan. A detailed discussion regarding the potential risks of fasting must be held with them.

While healthy pregnant women can generally fast safely with no maternal or fetal risk, those with hyperglycemia need to strictly monitor their BG levels to prevent hyperglycemia, hypoglycemia, and adverse maternal and fetal outcomes (35).

 

  • Pre-Ramadan assessment should begin months before Ramadan, and apart from risk stratification, should focus on breaking general myths like ‘finger-prick testing for BG levels breaks their fast’, encourage SMBG, and educate about the maternal and fetal risks of both hypoglycemia and hyperglycemia (35).
  • Pregnant women with diabetes should maintain normal physical activity while fasting. The Taraweeh prayer they offer should be considered as exercise for which insulin doses should be adjusted as required (35).
  • Fiber rich food and drinking 2-3 liters of water a day should be encouraged. Suhoor should be taken as late as possible (35).
  • Insulin and/or metformin are the treatment of choice in pregnancy with diabetes. Though glibenclamide is also used in some patients, its use should be discouraged during Ramadan. Some women with gestational diabetes may be managed on diet and/or metformin too.
  • The metformin dose may not need any change during pregnancy but the dose of insulin should be modified as discussed in Table 5 (10, 35). Insulin dose titration should be guided by SMBG as shown in Table 6.
  • SMBG should be carried out as guided in Figure 6. Pregnant women with diabetes should strictly monitor and maintain their BG levels as follows (35):
  • Fasting between 70-95 mg/dL (3.9 – 5.3 mmol/L).
  • Post-prandial < 120 mg/dL (6.7 mmol/L).
  • Pregnant women with diabetes should break their fast if (35):
  • BG levels < 70 mg/dL (3.9 mmol/L) during fasting hours.
  • Feel unwell.
  • Feel reduced fetal movement.
  • Pregnant women with diabetes should carry out regular SMBG at the following time points (35):
  • Before the sunset meal.
  • 1-2 hours after meals
  • Once during the day while fasting, particularly in the afternoon.
  • Anytime they feel unwell.
Management of Diabetes in Elderly with Diabetes Fasting During Ramadan

Older age (≥ 65 years) by itself can be considered a high risk for fasting during Ramadan in individuals with diabetes, even though many elderly fast successfully during Ramadan (38). Older individuals with diabetes are less likely to fast than younger ones (DAR 2020 survey: 71.2% of ≥ 65 years intended to fast compared to 87.3% of those < 65 years) (39). However, fasting during Ramadan being a personal choice, many older adults with diabetes do choose to fast during Ramadan. The DAR Global Survey (2020) also showed that the elderly were more motivated to fast with 69% of those aged ≥ 65 years fasting for 30 days compared to 60% of those < 65 years (39).

 

Elderly (≥ 65 years) with diabetes were significantly more likely to break their fast than younger (<65 years) individuals with diabetes (17% vs. 11.5%; P<0.001) (39). Similarly, they were significantly more likely to break their fast due to hypoglycemia than their younger counterparts (67.7% vs.55.4%; P=0.02).

 

Fasting during Ramadan in elderly with diabetes needs special consideration and attention because:

  • Diabetes related complications are higher in elderly and they need careful BG monitoring and GLD dose adjustments, which should be started well before Ramadan (38).
  • Fasting related complications likely to be seen in elderly with diabetes can be due to both hyperglycemia and hypoglycemia, and also include impaired renal function, impaired postural balance, poor attention, and volume depletion. The risk increases with the number of days fasted (38).
  • The DAR 2020 survey showed that hypoglycemia was significantly higher in elderly as compared to younger population (17.4% vs.15.2%; P<0.001) (39).
  • 9% of those aged ≥ 65 visited the emergency department compared to 4.3% of individuals aged < 65
  • Elderly were also more likely to get hospitalized due to hypoglycemia
  • While 31.5% reduced their GLD dosing, 17% made no change to their medication dose
  • The use of SUs and insulin increases risk of hypoglycemia. 32.7% of elderly need insulin, probably due to long standing diabetes.
  • The DAR 2020 survey also showed that significantly greater number of elderly with diabetes who are fasting had hyperglycemia (BG levels > 16.6 mmol/L or 300 mg/dL) during Ramadan (19.3% vs. 15.6%; P=0.006) (39). 8.4% of the elderly had to attend the emergency department due to hyperglycemia related complications.
  • The DAR 2020 survey showed that the majority (80%) do not break their fast even if they have hyperglycemia, and 20% do not change their behavior (food intake, medication change), 25% reduced their food intake, and 21% increased their medication dose (39).
  • In the DAR 2020 Global Survey, 21% of participants with T2DM aged ≥ 65 years checked their BG levels once or less than once a week. Only around 10% checked their BG levels 3–4 times a day. There was no change in SMBG behavior during Ramadan (39).
  • Research on elderly fasting during Ramadan is largely lacking.38 Landmark trials in Ramadan like the EPIDIAR study which was used to formulate many recommendations for individuals with diabetes fasting during Ramadan, did not include the elderly.7
  • The elderly population is growing fast, and therefore there will be more individuals with diabetes who are ≥ 65 years and intend to fast (40)
  • The risk of fasting is much higher in elderly than in younger population with diabetes (38). This is because the elderly have more comorbidities (hypertension, hyperlipidemia, CVDs, CKD etc.) than the younger population (38, 39).
  • Elderly with diabetes and impaired renal functions, CVD, dementia, frailty, and/or those with risk of falls are at higher risk for complications during fasting than elderly without comorbidities (38). Therefore, risk stratification of elderly with diabetes who decide to fast during Ramadan should be based not only on age, but also on their comorbidities, functional capacity, and ability to manage medications and carry out SMBG, cognition, and social circumstances (38).

 

Hence, the elderly with diabetes are a high-risk category for fasting during Ramadan. They need proper Pre-Ramadan risk stratification, education, and support to ensure that they can fast safely with proper SMBG and medication monitoring.

 

Table 11 covers the basic recommendations for elderly who intend to fast during Ramadan.

 

Table 11. Basic Recommendations for Elderly who Intend to Fast During Ramadan

MEDICATIONS AND REGIMENS

•                Choose medications that have a lower hypoglycemia risk

•                 Make dose adjustments to lower the risk of hypoglycemia

•                 For individuals on SUs, gliclazide and glimepiride should be used instead of glibenclamide

•       SGLT2 inhibitors doses should be reviewed for benefit vs risks of adverse events especially in elderly with impaired renal function or those on diuretics

•       Insulin: dose titration based on SMBG should be taught and dose modifications carried out based on the insulin type

SMBG

•                 Increase frequency to a 5-point time scale

•                 Use CGM if available and feasible

DIET AND PHYSICAL ACTIVITY

•                 Individualized diet and activity plan

•                 Started before Ramadan and adhere during fasting days

•                 Medication doses and timings adjusted according to diet and physical activity level

•                 Adequate nutrition should be stressed and education provided

•                 Hydration ensured through proper planning

SOCIAL SUPPORT

Given that elderly may have cognition, memory, and physical deficits, adequate support should be ensured pre-Ramadan to ensure SMBG, adherence to diet and physical activity plan, insulin dose titration, and oral GLD dose modification

AWARENESS OF RISK OF COMPLICATIONS

•                 Discuss and document symptoms and events to increase awareness and recognition of complications

•                 Both patient and caregiver should be educated to recognize the symptoms of complications

  CGM, continuous glucose monitoring; GLD, glucose lowering drugs; SMBG, self-monitoring of blood glucose

Other Concerns Regarding Management of Diabetes During Ramadan

MANAGEMENT OF COMORBID HYPOTHYROIDISM

Hypothyroidism is commonly seen in patients with diabetes. Usually, thyroxine is taken half an hour before breakfast. However, during Ramadan, the breakfast time is shifted to pre-dawn. This is a time of rush and individuals may find it difficult to time the thyroxine dose half an hour before Suhoor. Similarly, if thyroxine is pushed to evening, then taking it half an hour before Iftar is usually difficult as usually this meal is taken with rest of the family and by Iftar time hunger score is high. Hence, thyroxine may be taken late (after a 4-hour gap) at night as long as no heavy meal is taken between Iftar and late night (12).

BARIATRIC SURGERY

Diabesity (co-existing diabetes and obesity) is of pandemic proportions across the world (4). Bariatric surgery is commonly performed in individuals with diabesity. Bariatric surgery poses certain concerns regarding fasting during Ramadan as these individuals cannot consume large meals and therefore absorb certain macronutrients (12).

GAPS AND WAY FORWARD

The last few decades have contributed immensely to the growing knowledge and clinical experience of health care providers regarding the clinical and metabolic complications of fasting, pre-fasting assessment, risk stratification and initiation of changes in medication dose and timing and dietary/lifestyle modifications during Ramadan (1). However, greater efforts are required to improve communication between the medical experts and religious scholars in order to ensure that medical guidance regarding safe fasting during Ramadan is best received by the public (30). Further well-designed clinical trials are required to assess the best treatment options for adolescents and adults with diabetes who fast during Ramadan. Artificial intelligence, use of RNP and other such tools need to be integrated to ensure safe fasting during Ramadan.

 ACKNOWLEDGEMENTS

All named authors for this manuscript meet the International Committee of Medical Journal Editors (ICMJE) criteria for authorship. All authors take full responsibility for the integrity of the work and have given final approval for the published version. The authors acknowledge Dr. Kokil Mathur and Dr. Punit Srivastava from Mediception Science Pvt. Ltd, Gurgaon, India for providing writing and editing assistance.

REFERENCES  

  1. Abdelrahim D, Faris ME, Hassanein M, et al. Impact of Ramadan Diurnal Intermittent Fasting on Hypoglycemic Events in Patients With Type 2 Diabetes: A Systematic Review of Randomized Controlled Trials and Observational Studies. Frontiers in Endocrinology. 2021;12. Accessed March 2, 2022. https://www.frontiersin.org/article/10.3389/fendo.2021.624423
  2. IDF-DAR Practical Guidelines. DAR practical guidelines for healthcare professionals. Published 2021. Accessed March 2, 2022. https://www.idf.org/our-activities/education/diabetes-and-ramadan/healthcare-professionals.html
  3. Jabbar A, Malek R, Hussein Z. Chapter 2. Epidemiology of diabetes and fasting during Ramadan. In: International Diabetes Federation and DAR International Alliance. Diabetes and Ramadan: Practical Guidelines. International Diabetes Federation and DAR International Alliance; 2021. Accessed March 4, 2022. https://www.idf.org/our-activities/education/diabetes-and-ramadan/healthcare-professionals.html
  4. Kapoor N, Sahay R, Kalra S, et al. Consensus on Medical Nutrition Therapy for Diabesity (CoMeND) in Adults: A South Asian Perspective. Diabetes Metab Syndr Obes. 2021;14:1703-1728. doi:10.2147/DMSO.S278928
  5. Bennakhi A, Buyukbese MA, Al Saleh Y, Almadani AA, Eliana F. Chapter 1. Introduction to the IDF-DAR ractical Guidelines. In: International Diabetes Federation and DAR International Alliance. Diabetes and Ramadan: Practical Guidelines. International Diabetes Federation and DAR International Alliance; 2021. Accessed March 4, 2022. https://www.idf.org/our-activities/education/diabetes-and-ramadan/healthcare-professionals.html
  6. Ahmed SH, Chowdhury TA, Hussain S, et al. Ramadan and Diabetes: A Narrative Review and Practice Update. Diabetes Ther. 2020;11(11):2477-2520. doi:10.1007/s13300-020-00886-y
  7. Salti I, Bénard E, Detournay B, et al. A population-based study of diabetes and its characteristics during the fasting month of Ramadan in 13 countries: results of the epidemiology of diabetes and Ramadan 1422/2001 (EPIDIAR) study. Diabetes Care. 2004;27(10):2306-2311. doi:10.2337/diacare.27.10.2306
  8. Babineaux SM, Toaima D, Boye KS, et al. Multi-country retrospective observational study of the management and outcomes of patients with Type 2 diabetes during Ramadan in 2010 (CREED). Diabet Med. 2015;32(6):819-828. doi:10.1111/dme.12685
  9. Wan Bebakar WM, Wan Mohamad RM, Hafidh K, et al. Chapter 9. Management of Type 1 diabetes when fasting during Ramadan. In: International Diabetes Federation and DAR International Alliance. Diabetes and Ramadan: Practical Guidelines. International Diabetes Federation and DAR International Alliance; 2021. Accessed March 4, 2022. https://www.idf.org/our-activities/education/diabetes-and-ramadan/healthcare-professionals.html
  10. Hanif W, Hassanein M, Elhadd TA, Mohamed NA. Chapter 10. Management of Type 2 diabetes when fasting during Ramadan. In: International Diabetes Federation and DAR International Alliance. Diabetes and Ramadan: Practical Guidelines. International Diabetes Federation and DAR International Alliance; 2021. Accessed March 4, 2022. https://www.idf.org/our-activities/education/diabetes-and-ramadan/healthcare-professionals.html
  11. Lessan N, Ali T. Energy Metabolism and Intermittent Fasting: The Ramadan Perspective. Nutrients. 2019;11(5):E1192. doi:10.3390/nu11051192
  12. Lessan N, Ezzat Faris M, Assaad-Khalil SH, Ali T. Chapter 3. What happens to the body? Physiology of fasting during Ramadan. In: International Diabetes Federation and DAR International Alliance. Diabetes and Ramadan: Practical Guidelines. International Diabetes Federation and DAR International Alliance; 2021. Accessed March 4, 2022. https://www.idf.org/our-activities/education/diabetes-and-ramadan/healthcare-professionals.html
  13. Lessan N, Hannoun Z, Hasan H, Barakat MT. Glucose excursions and glycaemic control during Ramadan fasting in diabetic patients: insights from continuous glucose monitoring (CGM). Diabetes Metab. 2015;41(1):28-36. doi:10.1016/j.diabet.2014.11.004
  14. Al-Rawi N, Madkour M, Jahrami H, et al. Effect of diurnal intermittent fasting during Ramadan on ghrelin, leptin, melatonin, and cortisol levels among overweight and obese subjects: A prospective observational study. PLoS One. 2020;15(8):e0237922. doi:10.1371/journal.pone.0237922
  15. Hassanein M, Al Awadi FF, El Hadidy KES, et al. The characteristics and pattern of care for the type 2 diabetes mellitus population in the MENA region during Ramadan: An international prospective study (DAR-MENA T2DM). Diabetes Res Clin Pract. 2019;151:275-284. doi:10.1016/j.diabres.2019.02.020
  16. Basit A, AlOzairi E, Abdelgadir E. Chapter 4. The effects of fasting during Ramadan on physical and mental wellbeing. In: International Diabetes Federation and DAR International Alliance. Diabetes and Ramadan: Practical Guidelines. International Diabetes Federation and DAR International Alliance; 2021. Accessed March 4, 2022. https://www.idf.org/our-activities/education/diabetes-and-ramadan/healthcare-professionals.html
  17. Husain R, Duncan MT, Cheah SH, Ch’ng SL. Effects of fasting in Ramadan on tropical Asiatic Moslems. Br J Nutr. 1987;58(1):41-48. doi:10.1079/bjn19870067
  18. Jabbar A, Hassanein M, Beshyah SA, Boye KS, Yu M, Babineaux SM. CREED study: Hypoglycaemia during Ramadan in individuals with Type 2 diabetes mellitus from three continents. Diabetes Res Clin Pract. 2017;132:19-26. doi:10.1016/j.diabres.2017.07.014
  19. Ba-Essa EM, Hassanein M, Abdulrhman S, Alkhalifa M, Alsafar Z. Attitude and safety of patients with diabetes observing the Ramadan fast. Diabetes Res Clin Pract. 2019;152:177-182. doi:10.1016/j.diabres.2019.03.031
  20. Mattoo V, Milicevic Z, Malone JK, et al. A comparison of insulin lispro Mix25 and human insulin 30/70 in the treatment of type 2 diabetes during Ramadan. Diabetes Res Clin Pract. 2003;59(2):137-143. doi:10.1016/s0168-8227(02)00202-4
  21. Cesur M, Corapcioglu D, Gursoy A, et al. A comparison of glycemic effects of glimepiride, repaglinide, and insulin glargine in type 2 diabetes mellitus during Ramadan fasting. Diabetes Res Clin Pract. 2007;75(2):141-147. doi:10.1016/j.diabres.2006.05.012
  22. Bakiner O, Ertorer ME, Bozkirli E, Tutuncu NB, Demirag NG. Repaglinide plus single-dose insulin glargine: a safe regimen for low-risk type 2 diabetic patients who insist on fasting in Ramadan. Acta Diabetol. 2009;46(1):63-65. doi:10.1007/s00592-008-0062-7
  23. Hassanein M, Hanif W, Malik W, et al. Comparison of the dipeptidyl peptidase-4 inhibitor vildagliptin and the sulphonylurea gliclazide in combination with metformin, in Muslim patients with type 2 diabetes mellitus fasting during Ramadan: results of the VECTOR study. Curr Med Res Opin. 2011;27(7):1367-1374. doi:10.1185/03007995.2011.579951
  24. Bashier A, Bin Hussain A, MK A. Safety and Efficacy of Liraglutide as an Add-On Therapy to Pre-Existing Anti-Diabetic Regimens during Ramadan, A Prospective Observational Trial. Journal of Diabetes & Metabolism. 2015;06. doi:10.4172/2155-6156.1000590
  25. Hassanein M, Echtay A, Hassoun A, et al. Tolerability of canagliflozin in patients with type 2 diabetes mellitus fasting during Ramadan: Results of the Canagliflozin in Ramadan Tolerance Observational Study (CRATOS). Int J Clin Pract. 2017;71(10). doi:10.1111/ijcp.12991
  26. Aldawi N, Darwiche G, Abusnana S, Elbagir M, Elgzyri T. Initial increase in glucose variability during Ramadan fasting in non-insulin-treated patients with diabetes type 2 using continuous glucose monitoring. Libyan J Med. 2019;14(1):1535747. doi:10.1080/19932820.2018.1535747
  27. Alawadi F, Mohammed K. Bashier A, Rashid F, Chowdhury TA. Chapter 13. Risks of fasting during Ramadan: Cardiovascular, Cerebrovascular and Renal complications. In: International Diabetes Federation and DAR International Alliance. Diabetes and Ramadan: Practical Guidelines. International Diabetes Federation and DAR International Alliance; 2021. Accessed March 4, 2022. https://www.idf.org/our-activities/education/diabetes-and-ramadan/healthcare-professionals.html
  28. Jawad F, Kalra S. Ramadan and diabetes management - The 5 R’s. J Pak Med Assoc. 2015;65(5 Suppl 1):S79-80.
  29. Ahmedani MY, Zainudin SB, AlOzairi E. Chapter 7. Pre-Ramadan Assessment and Education. In: International Diabetes Federation and DAR International Alliance. Diabetes and Ramadan: Practical Guidelines. International Diabetes Federation and DAR International Alliance; 2021. Accessed March 4, 2022. https://www.idf.org/our-activities/education/diabetes-and-ramadan/healthcare-professionals.html
  30. El Sayed AA, Hassanein M, Afandi B, Tayeb K, Diop SN. Chapter 6. Diabetes and Ramadan: A Medico-Religious Perspective. In: International Diabetes Federation and DAR International Alliance. Diabetes and Ramadan: Practical Guidelines. International Diabetes Federation and DAR International Alliance; 2021. Accessed March 4, 2022. https://www.idf.org/our-activities/education/diabetes-and-ramadan/healthcare-professionals.html
  31. Hassanein M, Al-Arouj M, Hamdy O, et al. Diabetes and Ramadan: Practical guidelines. Diabetes Res Clin Pract. 2017;126:303-316. doi:10.1016/j.diabres.2017.03.003
  32. Hamdy O, Mohd Yusof BN, Maher S. Chapter 8. The Ramadan Nutrition Plan (RNP) for people with diabetes. In: International Diabetes Federation and DAR International Alliance. Diabetes and Ramadan: Practical Guidelines. International Diabetes Federation and DAR International Alliance; 2021. Accessed March 4, 2022. https://www.idf.org/our-activities/education/diabetes-and-ramadan/healthcare-professionals.html
  33. Ahmed WN, Arun CS, Koshy TG, et al. Management of diabetes during fasting and COVID-19 – Challenges and solutions. Journal of Family Medicine and Primary Care. 2020;9(8):3797-3806. doi:10.4103/jfmpc.jfmpc_845_20
  34. Mohd Yusof BN, Wan Zukiman WZHH, Abu Zaid Z, et al. Comparison of Structured Nutrition Therapy for Ramadan with Standard Care in Type 2 Diabetes Patients. Nutrients. 2020;12(3):813. doi:10.3390/nu12030813
  35. Afandi B, Hassanein M, Taha Salih B, Abdo S. Chapter 11. Management of hyperglycaemia in pregnancy when fasting during Ramadan. In: International Diabetes Federation and DAR International Alliance. Diabetes and Ramadan: Practical Guidelines. International Diabetes Federation and DAR International Alliance; 2021. Accessed March 4, 2022. https://www.idf.org/our-activities/education/diabetes-and-ramadan/healthcare-professionals.html
  36. Bajaj HS, Abouhassan T, Ahsan MR, et al. Diabetes Canada Position Statement for People With Types 1 and 2 Diabetes Who Fast During Ramadan. Can J Diabetes. 2019;43(1):3-12. doi:10.1016/j.jcjd.2018.04.007
  37. Elhadd T, Dabbous Z, Bashir M, et al. Incidence of hypoglycaemia in patients with type-2 diabetes taking multiple glucose lowering therapies during Ramadan: the PROFAST Ramadan Study. J Diabetes Metab Disord. 2018;17(2):309-314. doi:10.1007/s40200-018-0374-2
  38. Shaltout I, Mohamed M, Iraqi H. Chapter 12. Management of diabetes among the elderly when fasting during Ramadan. In: International Diabetes Federation and DAR International Alliance. Diabetes and Ramadan: Practical Guidelines. International Diabetes Federation and DAR International Alliance; 2021. Accessed March 4, 2022. https://www.idf.org/our-activities/education/diabetes-and-ramadan/healthcare-professionals.html
  39. Hassanein M, Hussein Z, Shaltout I, et al. The DAR 2020 Global survey: Ramadan fasting during COVID 19 pandemic and the impact of older age on fasting among adults with Type 2 diabetes. Diabetes Res Clin Pract. 2021;173:108674. doi:10.1016/j.diabres.2021.108674
  40. IDF. IDF Diabetes Atlas 2021. International Diabetes Federation; 2021. Accessed March 11, 2022. https://diabetesatlas.org/atlas/tenth-edition/