Growth Hormone (GH) and Growth Hormone Antagonists

Number: 0170

Table Of Contents

Policy
Applicable CPT / HCPCS / ICD-10 Codes
Background
References


Policy

Scope of Policy

This Clinical Policy Bulletin addresses growth hormone (GH) and growth hormone antagonists for commercial medical plans. For Medicare criteria, see Medicare Part B Criteria.

Note: Requires Precertification:

Precertification of Serostim or Somavert is required of all Aetna participating providers and members in applicable plan designs. For precertification of these medications, call (866) 752-7021 or fax (888) 267-3277. For Statement of Medical Necessity (SMN) precertification forms, see Specialty Pharmacy Precertification.

Somatropin Products - Genotropin, Humatrope, Norditropin, Nutropin AQ, Omnitrope, Saizen, and Zomacton

  1. Criteria for Initial Approval

    Aetna considers somatropin products: Genotropin, Humatrope, Norditropin, Nutropin AQ, Omnitrope, Saizen, and Zomacton medically necessary for the following indications:

    1. Pediatric Growth Hormone (GH) Deficiency

      When either criteria 1 or 2 below is met:

      1. Member is a neonate or was diagnosed with GH deficiency as a neonate. Medical records must be available to support the diagnosis of neonatal GH deficiency (e.g., hypoglycemia with random GH level, evidence of multiple pituitary hormone deficiency, chart notes, or magnetic resonance imaging [MRI] results); or
      2. Member meets all of the following:

        1. Member has either:

          1. Two pretreatment pharmacologic provocative GH tests with both results demonstrating a peak GH level less than 10 ng/mL, or
          2. A documented pituitary or CNS disorder (see Appendix A) and a pretreatment IGF-1 level greater than 2 standard deviations (SD) below the mean; and
        2. For members less than 2.5 years of age at initiation of treatment, the pretreatment height is greater than 2 SD below the mean and growth velocity is slow; and
        3. For members greater than or equal to 2.5 years of age at initiation of treatment:

          1. Pretreatment height is greater than 2 SD below the mean and 1-year height velocity is greater than 1 SD below the mean; or
          2. Pretreatment 1-year height velocity is greater than 2 SD below the mean; and
        4. Epiphyses are open;

        Note: Given the above criteria, further laboratory testing of children without classic GHD to diagnose "partial" GHD, or other abnormalities of GH secretion or bioactivity, is considered not medically necessary.  This includes over-night hospitalization of children for testing of spontaneous GH secretion.

        Note: Measurement of insulin-like growth factor I (IGF-I) is considered medically necessary to determine adequacy of GH therapy in adults and children. However, the diagnosis of GH deficiency should not rely solely on IGF-I measurements, but must be confirmed by provocative tests solely for GH secretion. Measurement of IGF binding protein-2 (IGFBP-2), IGF binding protein-3 (IGFBP-3), and the acid labile subunit of IGF-I are considered experimental and investigational.

    2. Small for Gestational Age (SGA)

      When all of the following criteria are met:

      1. Member meets at least one of the following:

        1. Birth weight less than 2500 g at gestational age greater than 37 weeks
        2. Birth weight or length less than 3rd percentile for gestational age
        3. Birth weight or length greater than or equal to 2 SD below the mean for gestational age; and
      2. Pretreatment age is greater than or equal to 2 years; and
      3. Member failed to manifest catch-up growth by age 2 (i.e., pretreatment height greater than 2 SD below the mean); and
      4. Epiphyses are open;
    3. Turner Syndrome

      When all of the following criteria are met:

      1. Diagnosis was confirmed by karyotyping; and
      2. Member's pretreatment height is less than the 5th percentile for age; and
      3. Epiphyses are open;
    4. Growth Failure Associated with Chronic Kidney Disease (CKD), Cerebral Palsy, Congenital Adrenal Hyperplasia, Cystic Fibrosis, or Russell-Silver Syndrome

      When all of the following criteria are met:

      1. For members less than 2.5 years of age at initiation of treatment, the pretreatment height is greater than 2 SD below the mean and growth velocity is slow; and
      2. For members greater than or equal to 2.5 years of age at initiation of treatment:

        1. Pretreatment height is greater than 2 SD below the mean and 1-year height velocity is greater than 1 SD below the mean; or
        2. Pretreatment height velocity is greater than 2 SD below the mean; and
      3. Epiphyses are open;

    5. Prader Willi syndrome

      When the diagnosis was confirmed by genetic testing demonstrating any of the following: 

      1. Deletion in the chromosomal 15q11.2-q13 region; or
      2. Maternal uniparental disomy in chromosome 15; or
      3. Imprinting defects, translocations, or inversions involving chromosome 15;
    6. Noonan Syndrome

      When all of the following criteria are met: 

      1. Pretreatment height is greater than 2 SD below the mean and 1-year height velocity is greater than 1 SD below the mean or pretreatment 1-year height velocity is greater than 2 SD below the mean; and
      2. Epiphyses are open;
    7. Short Stature Homeobox-Containing Gene (SHOX) Deficiency

      When all of the following criteria are met: 

      1. The diagnosis of SHOX deficiency is confirmed by molecular or genetic analysis; and
      2. Pretreatment height is greater than 2 SD below the mean and 1-year height velocity is greater than 1 SD below the mean or pretreatment 1-year height velocity is greater than 2 SD below the mean; and
      3. The epiphyses are open;
    8. Adult Growth Hormone (GH) Deficiency

      When any of the following criteria is met: 

      1. Member meets both of the following:

        1. Member has had 2 pretreatment pharmacologic provocative GH tests and both results demonstrated deficient GH responses defined as the following:

          1. Insulin tolerance test (ITT) with a peak GH level less than or equal to 5 ng/mL;
          2. Macrilen with a peak GH level of less than 2.8 ng/mL;
          3. Glucagon stimulation test with a peak GH level less than or equal to 3.0 ng/mL in members with a body mass index (BMI) less than or equal to 30 kg/m2 and a high pretest probability of GHD (e.g., acquired structural abnormalities) or a BMI less than 25 kg/m2
          4. Glucagon stimulation test with a peak GH level less than or equal to 1.0 ng/mL in members with a BMI of greater than or equal to 25 kg/m2 and a low pretest probability of GHD (e.g., acquired structural abnormalities) or a BMI greater than 30 kg/m2; and
        2. Member has a low pre-treatment IGF-1 (between 0 to 2 SD below the mean for age and gender); or

      2. Member meets both of the following:

        1. Member has had one pretreatment pharmacologic provocative GH test that demonstrated deficient GH responses defined as one of the following:

          1. Insulin tolerance test (ITT) with a peak GH level less than or equal to 5 ng/mL;
          2. Macrilen with a peak GH level less than or equal to 2.8 ng/mL;
          3. Glucagon stimulation test with a peak GH level less than or equal to 3.0 ng/mL in members with a body mass index (BMI) less than or equal to 30 kg/m2 and a high pretest probability of GHD (e.g., acquired structural abnormalities) or a BMI less than 25 kg/m2;
          4. Glucagon stimulation test with a peak GH level less than or equal to 1.0 ng/mL in members with a BMI of greater than or equal to 25 kg/m2 and a low pretest probability of GHD (e.g., acquired structural abnormalities) or a BMI greater than 30 kg/m2; and
        2. Member has a pretreatment IGF-1 level that is more than 2 SD below the mean for age and gender; or

      3. Member has organic hypothalamic-pituitary disease (e.g., suprasellar mass with previous surgery and cranial irradiation) with greater than or equal to 3 documented pituitary hormone deficiencies (see Appendix B) and a low pre-treatment IGF-1 more than 2 standard deviations below the mean for age and gender; or
      4. Member has a genetic or structural hypothalamic-pituitary defects (see Appendix C); or 
      5. Member has childhood-onset GH deficiency and a congenital abnormality of the CNS, hypothalamus or pituitary (see Appendix C);
    9. HIV-associated wasting / cachexia

      For HIV-associated wasting or cachexia when all of the following criteria are met:

      1. Member has trialed and experienced a suboptimal response to alternative therapies (see Appendix E) or contraindication or intolerance to alternative therapies; and
      2. Member is currently on antiretroviral therapy; and
      3. BMI is less than 18.5 kg/m2 prior to starting therapy with growth hormone (see Appendix D);
    10. Short-bowel syndrome

      For a lifetime total of 8 weeks for members with short bowel syndrome (SBS) who depend on intravenous parenteral nutrition for nutitional support when GH will be used in conjunction with optimal management of SBS.

    Aetna considers all other indications as experimental and investigational (for additional information, see Experimental and Investigational and Background sections).

  2. Continuation of Therapy

    Aetna considers continuation of somatropin products: Genotropin, Humatrope, Norditropin, Nutropin AQ, Omnitrope, Saizen, and Zomacton therapy medically necessary for the following indications:

    1. Pediatric GH Deficiency, Turner Syndrome, Noonan Syndrome, Chronic Kidney Disease (CKD), Small for Gestational Age (SGA), SHOX Deficiency, Congenital Adrenal Hyperplasia, Cerebral Palsy, Cystic Fibrosis, and Russell-Silver Syndrome

      When all of the following criteria are met:

      1. Epiphyses are open (confirmed by X-ray or X-ray is not available); and
      2. Member’s growth rate is greater than 2 cm/year unless there is a documented clinical reason for lack of efficacy (e.g., on treatment less than 1 year, nearing final adult height/late stages of puberty);
    2. Prader-Willi Syndrome

      When the member’s body composition and psychomotor function have improved or stabilized in response to GH therapy;

    3. Adult GH Deficiency

      When any of the following criteria is met: 

      1. Member meets all of the following:

        1. Member has had 2 pretreatment pharmacologic provocative GH tests and both results demonstrated deficient GH responses defined as the following:

          1. Insulin tolerance test (ITT) or another provocative GH test with a peak GH level less than or equal to 5 ng/mL;
          2. Macrilen with a peak GH level of less than 2.8 ng/mL;
          3. Glucagon stimulation test with a peak GH level less than or equal to 3.0 ng/mL in members with a body mass index (BMI) less than or equal to 30 kg/m2 and a high pretest probability of GHD (e.g., acquired structural abnormalities) or a BMI less than 25 kg/m2;
          4. Glucagon stimulation test with a peak GH level less than or equal to 1.0 ng/mL in members with a BMI of greater than or equal to 25 kg/m2 and a low pretest probability of GHD (e.g., acquired structural abnormalities) or a BMI greater than 30 kg/m2; and
        2. Member has a low pre-treatment IGF-1 (between 0 to 2 SD below the mean for age and gender); and
        3. Current IGF-1 level is not elevated for age and gender; or
      2. Member meets all of the following:

        1. Member has had 1 pretreatment pharmacologic provocative GH test that demonstrated deficient GH responses defined as one of the following:

          1. Insulin tolerance test (ITT) or another provocative GH test with a peak GH level less than or equal to 5 ng/mL;
          2. Macrilen with a peak GH level less than or equal to 2.8 ng/mL;
          3. Glucagon stimulation test with a peak GH level less than or equal to 3.0 ng/mL in members with a body mass index (BMI) less than or equal to 30 kg/m2 and a high pretest probability of GHD (e.g., acquired structural abnormalities) or a BMI less than 25 kg/m2;
          4. Glucagon stimulation test with a peak GH level less than or equal to 1.0 ng/mL in members with a BMI of greater than or equal to 25 kg/m2 and a low pretest probability of GHD (e.g., acquired structural abnormalities) or a BMI greater than 30 kg/m2; and
        2. Member has a pretreatment IGF-1 level that is more than 2 SD below the mean for age and gender; and
        3. Current IGF-1 level is not elevated for age and gender; or
      3. Member meets both of the following:

        1. Member has organic hypothalamic-pituitary disease (e.g., suprasellar mass with previous surgery and cranial irradiation) with greater than or equal to 3 documented pituitary hormone deficiencies (see Appendix B) and a low pre-treatment IGF-1 more than 2 standard deviations below the mean for age and gender; and
        2. Current IGF-1 level is not elevated for age and gender; or
      4. Member has genetic or structural hypothalamic-pituitary defects (see Appendix C) and current IGF-1 level is not elevated for age and gender; or
      5. Member has childhood-onset GH deficiency and a congenital abnormality of the CNS, hypothalamus or pituitary (see Appendix C) and current IGF-1 level is not elevated for age and gender;
    4. HIV-Associated Wasting/Cachexia

      When all of the following criteria are met:

      1. Member is diagnosed with HIV-associated wasting/cachexia; and
      2. Member is currently on antiretroviral therapy; and
      3. Member is currently receiving treatment with growth hormone excluding obtainment as samples or via manufacturer’s assistance programs; and
      4. Current BMI is less than 27 kg/m2 (see Appendix D).

Somatropin (Serostim)

  1. Criteria for Initial Approval

    HIV with wasting or cachexia

    Aetna considers somatropin (Serostim) medically necessary for treatment of HIV-associated wasting or cachexia to increase lean body mass and body weight, and improve physical endurance when all of the following criteria are met:

    1. Member is currently on antiretroviral therapy; and
    2. Trial with suboptimal response to alternative therapies (see Appendix E) or contraindication or intolerance to alternative therapies; and
    3. Body mass index (BMI) was less than 18.5 kg/m2 prior to initiating therapy with Serostim (see Appendix D).

    Aetna considers all other indications as experimental and investigational (for additional information, see Experimental and Investigational and Background sections).

  2. Continuation of Therapy

    Aetna considers continuation of somatropin (Serostim) therapy medically necessary for treatment of HIV-associated wasting or cachexia to increase lean body mass and body weight, and improve physical endurance when all of the following criteria are met:

    1. Member is currently on antiretroviral therapy; and
    2. Member is currently receiving treatment with Serostim excluding obtainment as samples or via manufacturer’s assistance programs; and
    3. Current BMI is less than 27 kg/m2 (see Appendix D).

Somapacitan-beco (Sogroya)

  1. Criteria for Initial Approval

    1. Pediatric growth hormone (GH) deficiency

      Aetna considers somapacitan-beco (Sogroya) medically necessary for members with pediatric GH deficiency 2.5 years of age or older when either criteria 1 or 2 below is met:

      1. Member was diagnosed with GH deficiency as a neonate. Medical records must be available to support the diagnosis of neonatal GH deficiency (e.g., hypoglycemia with random GH level, evidence of multiple pituitary hormone deficiency, chart notes, or magnetic resonance imaging [MRI] results); or
      2. Member meets all of the following:
        1. Member has either:

          1. Two pretreatment pharmacologic provocative GH tests with both results demonstrating a peak GH level less than 10 ng/mL; or
          2. A documented pituitary or CNS disorder (refer to Appendix A) and a pretreatment IGF-1 level greater than 2 standard deviations (SD) below the mean; and
        2. Member meets one of the following:

          1. Pretreatment height is greater than 2 SD below the mean and 1-year height velocity is greater than 1 SD below the mean; or
          2. Pretreatment 1-year height velocity is greater than 2 SD below the mean; and
        3. Epiphyses are open;

    2. Adult growth hormone deficiency

      Aetna considers somapacitan-beco (Sogroya) medically necessary for members with adult GH deficiency when any of the following criteria is met:

      1. Member meets both of the following:

        1. Member has had 2 pretreatment pharmacologic provocative GH tests and both results demonstrated deficient GH responses defined as the following:

          1. Insulin tolerance test (ITT) with a peak GH level less than or equal to 5 ng/mL;
          2. Macrilen with a peak GH level of less than 2.8 ng/mL;
          3. Glucagon stimulation test with a peak GH level less than or equal to 3.0 ng/mL in members with a body mass index (BMI) less than or equal to 30 kg/m2 and a high pretest probability of GHD (e.g., acquired structural abnormalities) or a BMI less than 25 kg/m2;
          4. Glucagon stimulation test with a peak GH level less than or equal to 1.0 ng/mL in members with a BMI of greater than or equal to 25 kg/m2 and a low pretest probability of GHD (e.g., acquired structural abnormalities) or a BMI greater than 30 kg/m2; and
        2. Member has a low pre-treatment IGF-1 (between 0 to 2 SD below the mean for age and gender); or

      2. Member meets both of the following:

        1. Member has had 1 pretreatment pharmacologic provocative GH test that demonstrated deficient GH responses defined as one of the following:

          1. Insulin tolerance test (ITT) with a peak GH level less than or equal to 5 ng/mL;
          2. Macrilen with a peak GH level of less than 2.8 ng/mL;
          3. Glucagon stimulation test with a peak GH level less than or equal to 3.0 ng/mL in members with a body mass index (BMI) less than or equal to 30 kg/m2 and a high pretest probability of GHD (e.g., acquired structural abnormalities) or a BMI less than 25 kg/m2;
          4. Glucagon stimulation test with a peak GH level less than or equal to 1.0 ng/mL in members with a BMI of greater than or equal to 25 kg/m2 and a low pretest probability of GHD (e.g., acquired structural abnormalities) or a BMI greater than 30 kg/m2; and
        2. Member has a pretreatment IGF-1 level that is more than 2 SD below the mean for age and gender; or

      3. Member has organic hypothalamic-pituitary disease (e.g., suprasellar mass with previous surgery and cranial irradiation) with greater than or equal to 3 documented pituitary hormone deficiencies (see Appendix B) and a low pre-treatment IGF-1 more than 2 standard deviations below the mean for age and gender; or
      4. Member has genetic or structural hypothalamic-pituitary defects (see Appendix C); or
      5. Member has childhood-onset GH deficiency and a congenital abnormality of the CNS, hypothalamus or pituitary (see Appendix C).

      Aetna considers all other indications as experimental and investigational (for additional information, see Experimental and Investigational and Background sections). 

  2. Continuation of Therapy

    1. Pediatric growth hormone deficiency

      Aetna considers continuation of somapacitan-beco (Sogroya) therapy medically necessary for members with pediatric GH deficiency when all of the following criteria are met:

      1. Epiphyses are open (confirmed by X-ray or X-ray is not available); and
      2. Member’s growth rate is greater than 2 cm/year unless there is a documented clinical reason for lack of efficacy (e.g., on treatment less than 1 year, nearing final adult height/late stages of puberty);
    2. Adult growth hormone deficiency

      Aetna considers continuation of somapacitan-beco (Sogroya) therapy medically necessary for members with adult GH deficiency when any of the following criteria is met:

      1. Member meets all of the following:

        1. Member has had 2 pretreatment pharmacologic provocative GH tests and both results demonstrated deficient GH responses defined as the following:

          1. Insulin tolerance test (ITT) or another provocative GH test with a peak GH level less than or equal to 5 ng/mL;
          2. Macrilen with a peak GH level of less than 2.8 ng/ml;
          3. Glucagon stimulation test with a peak GH level less than or equal to 3.0 ng/mL in members with a body mass index (BMI) less than or equal to 30 kg/m2 and a high pretest probability of GHD (e.g., acquired structural abnormalities) or a BMI less than 25 kg/m2;
          4. Glucagon stimulation test with a peak GH level less than or equal to 1.0 ng/mL in members with a BMI of greater than or equal to 25 kg/m2 and a low pretest probability of GHD (e.g., acquired structural abnormalities) or a BMI greater than 30 kg/m2; and
        2. Member has a low pre-treatment IGF-1 (between 0 to 2 SD below the mean for age and gender); and
        3. Current IGF-1 level is not elevated for age and gender; or
      2. Member meets all of the following:

        1. Member has had 1 pretreatment pharmacologic provocative GH test that demonstrated deficient GH responses defined as one of the following:

          1. Insulin tolerance test (ITT) or another provocative GH test with a peak GH level less than or equal to 5 ng/mL;
          2. Macrilen with a peak GH level less than or equal to 2.8 ng/mL;
          3. Glucagon stimulation test with a peak GH level less than or equal to 3.0 ng/mL in members with a body mass index (BMI) less than or equal to 30 kg/m2 and a high pretest probability of GHD (e.g., acquired structural abnormalities) or a BMI less than 25 kg/m2;
          4. Glucagon stimulation test with a peak GH level less than or equal to 1.0 ng/mL in members with a BMI of greater than or equal to 25 kg/m2 and a low pretest probability of GHD (e.g., acquired structural abnormalities) or a BMI greater than 30 kg/m2; and
        2. Member has a pretreatment IGF-1 level that is more than 2 SD below the mean for age and gender; and
        3. Current IGF-1 level is not elevated for age and gender; or
      3. Member meets both of the following:

        1. Member has organic hypothalamic-pituitary disease (e.g., suprasellar mass with previous surgery and cranial irradiation) with greater than or equal to 3 documented pituitary hormone deficiencies (see Appendix B) and a low pre-treatment IGF-1 more than 2 standard deviations below the mean for age and gender; and
        2. Current IGF-1 level is not elevated for age and gender; or
      4. Member has genetic or structural hypothalamic-pituitary defects (see Appendix C) and current IGF-1 level is not elevated for age and gender; or
      5. Member has childhood-onset GH deficiency and a congenital abnormality of the CNS, hypothalamus or pituitary (see Appendix C) and current IGF-1 level is not elevated for age and gender.

Lonapegsomatropin-tcgd (Skytrofa)

  1. Criteria for Initial Approval

    Aetna considers lonapegsomatropin-tcgd (Skytrofa) medically necessary for treatment of pediatric GH deficiency in members 1 year of age and older when either criteria A or B below is met:

    1. Member was diagnosed with GH deficiency as a neonate. Medical records must be available to support the diagnosis of neonatal GH deficiency (e.g., hypoglycemia with random GH level, evidence of multiple pituitary hormone deficiency, chart notes, or magnetic resonance imaging [MRI] results); or
    2. Member meets all of the following:

      1. Member has either:

        1. Two pretreatment pharmacologic provocative GH tests with both results demonstrating a peak GH level less than 10 ng/mL; or
        2. A documented pituitary or CNS disorder (see Appendix A) and a pretreatment IGF-1 level greater than 2 standard deviations (SD) below the mean; and
      2. For members less than 2.5 years of age at initiation of treatment, the pretreatment height is greater than 2 SD below the mean and growth velocity is slow; and
      3. For members greater than or equal to 2.5 years of age at initiation of treatment:
         
        1. Pretreatment height is greater than 2 SD below the mean and 1-year height velocity is greater than 1 SD below the mean; or
        2. Pretreatment 1-year height velocity is greater than 2 SD below the mean; and
      4. Epiphyses are open.

    Aetna considers all other indications as experimental and investigational

  2. Continuation of Therapy

    Aetna considers continuation of lonapegsomatropin-tcgd (Skytrofa) therapy medically necessary for pediatric GH deficiency when both of the following criteria are met:

    1. Epiphyses are open (confirmed by X-ray or X-ray is not available); and
    2. Member’s growth rate is greater than 2 cm/year unless there is a documented clinical reason for lack of efficacy (e.g., on treatment less than 1 year, nearing final adult height/late stages of puberty).

Note: GH therapy will be considered medically necessary for members who meet medical necessity criteria; even if they are also diagnosed with a co-morbid medical condition for which GH therapy is considered not medically necessary or experimental and investigational.  

Note for Idiopathic Short Stature (ISS): Aetna does not consider idiopathic short stature an illness, disease or injury.  Accordingly, coverage would not be available under most plans, which provide coverage only for treatment of illness, injury or disease.  If the benefit plan only covers treatment for disease, illness or injury and the diagnosis is idiopathic short stature, GH is not a covered plan benefit.  When GH is not a covered plan benefit, medical necessity language should not be included within the review determination rationale.  This a contractual denial and not based upon medical necessity.

Macimorelin (Macrilen)

Aetna considers orally administered macimorelin (Macrilen) stimulation test medically necessary for diagnosis of adult growth hormone deficiency (AGHD) when all of the following criteria are met:

  • Member is 18 years of age or older; and
  • Member's body mass index (BMI) is less than or equal to 40 kg/m2; and
  • Macimorelin is prescribed by an endocrinologist; and
  • Member must have a contraindication to all other diagnostic tests (insulin tolerance test, glucagon stimulation test, arginine, clonidine, levodopa, or arginine combined with levodopa) for growth hormone deficiency; and
  • Dosage does not exceed 0.5 mg/kg as single dose.

Aetna considers all other indications as experimental and investigational.

Pegvisomant (Somavert)

  1. Criteria for initial Approval

    Aetna considers pegvisomant (Somavert) medically necessary for the treatment of acromegaly when all of the following criteria are met:

    1. Member has a high pretreatment insulin-like growth factor-1 (IGF-1) level for age and/or gender based on the laboratory reference range; and
    2. Member had an inadequate or partial response to surgery or radiotherapy or there is a clinical reason why the member has not had surgery or radiotherapy.

    Aetna considers all other indications as experimental and investigational.

  2. Continuation of Therapy

    Aetna considers continuation of pegvisomant (Somavert) therapy medically necessary for acromegaly when the member's IGF-1 level has decreased or normalized since initiation of therapy.

For somatropin (Zorbtive), see Specialty Pharmacy Clinical Policy Bulletin Zorbtive 2244-A SGM.

Related Policies

Dosage and Administration

Growth Hormone or Growth Hormone Antagonist

Refer to the medication's full prescribing information for dosage and administration information.

Macimorelin (Macrilen) Oral Stimulation Test

Macimorelin (Macrilen) is available as an oral solution of 60 mg white to off-white granules in a pouch for reconstitution in 120 mL of water, resulting in a solution of 0.5 mg/mL of macimorelin.

Dosing and Administration Recommendations

  • Recommended dose is 0.5 mg/kg as a single oral dose, after fasting for at least 8 hours.
  • Discontinue therapy with strong CYP3A4 inducers, growth hormones and drugs that affect GH release for an adequate length of time before administering macimorelin.
  • Adequately replace other hormone deficiencies before administering macimorelin.

Note: Serum GH level of less than 2.8 ng/mL (i.e., at the 30, 45, 60 and 90 minute timepoints) following macimorelin administration confirms the presence of adult growth hormone deficiency.

Source: Allphamed Pharbil Arzneimittel GmbH, 2021

Experimental and Investigational

Aetna considers GH therapy to be experimental and investigational for the following indications (not an all-inclusive list) because its effectiveness for these indications has not been established:

  • Adolescent transgender transitioning to male
  • Amyotrophic lateral sclerosis
  • Anabolic therapy to enhance body mass or strength for professional, recreational or social reasons
  • Anti-aging
  • Burn injuries
  • CHARGE (Coloboma, Heart defect, Atresia choanae, Retarded growth and development, Genital hypoplasia, Ear anomalies/deafness) syndrome
  • Chiari malformations
  • Chondral defect repair
  • Chondrodystrophy
  • Chronic catabolic states, including inflammatory bowel disease, pharmacologic glucocorticoid administration, and respiratory failure
  • Chronic fatigue syndrome
  • Chronic kidney disease (other than growth failure associated with CKD)
  • Chronic pain syndromes (e.g., fibromyalgia and lower back pain)
  • Congestive heart failure
  • Constitutional delay of growth and development
  • Corticosteroid-induced pituitary ablation
  • Crohn's disease
  • Decompensated cirrhosis
  • Depression
  • Down syndrome and other syndromes associated with short stature and increased susceptibility to neoplasms (e.g., Bloom syndrome, Fanconi syndrome)
  • Fracture healing
  • Glucocorticoid-induced growth failure
  • Growth hormone insensitivity (partial or complete)
  • Growth retardation due to amphetamines (e.g., Adderall, Ritalin)
  • HIV lipodystrophy
  • Hypertension
  • Hypochondroplasia
  • Hypophosphatemia (e.g., hypophosphatemic rickets, X-linked hypophosphatemia in children)
  • Implant osseointegration
  • Improvement in healing after rotator cuff repair
  • Improvement of oral health in children with growth hormone deficiency
  • Infertility (testicular hypofunction)/in-vitro fertilization, improvement of endometrial receptivity during in-vitro fertilization, improvement of in-vitro fertilization (IVF) outcomes of poor ovarian responders
  • Injured retina
  • Insulin-like growth factor-I (IGF-1) deficiency (also known as neurosecretory defect)
  • Intra-uterine growth restriction not meeting diagnostic criteria for small for gestational age children
  • Ischemic heart disease
  • Isochromosome Yp defect 
  • Juvenile rheumatoid arthritis
  • Kabuki syndrome
  • Kearns-Sayre syndrome
  • Methadone-induced toxicity
  • Muscular dystrophy
  • Neurosecretory growth hormone dysfunction
  • Obesity/morbid obesity
  • Osteogenesis imperfecta
  • Osteoporosis
  • Post-bariatric surgery
  • Post-polio syndrome
  • Post-traumatic stress disorder
  • Precocious puberty
  • Pseudohypoparathyroidism
  • Skeletal dysplasias (e.g., achondroplasia, kyphomelic dysplasia)
  • “Somatopause” in older adults
  • Spina bifida
  • Stem cell mobilization
  • Testicular cancer survivors
  • Traumatic brain injury (other than associated with pediatric GH deficiency)
  • Treatment of thalassemia
  • Wound healing.

Table:

CPT Codes / HCPCS Codes / ICD-10 Codes

Code Code Description

Information in the [brackets] below has been added for clarification purposes.   Codes requiring a 7th character are represented by "+":

CPT codes covered if selection criteria are met:

Macimorelin (Macrilen) stimulation test - no specific code:

CPT codes not covered for indications listed in the CPB:

38205 Blood-derived hematopoetic progenitor cell harvesting for transplantation, per collection; allogenic
38206     autologous

Other CPT codes related to the CPB:

70450 - 70470 Computed tomography, head or brain; without contrast material, with contrast material(s), or without contrast material, followed by contrast material(s) and further sections
70496 Computed tomography angiography, head, with contrast material(s), including noncontrast images, if performed, and image postprocessing
70551 - 70553 Magnetic resonance (e.g., proton) imaging, brain (including brain stem); without contrast material, with contrast material(s), or without contrast material, followed by contrast material(s) and further sequences
70554 - 70555 Magnetic resonance imaging, brain, functional MRI
80418 Combined rapid anterior pituitary evaluation panel
80422 Glucagon tolerance panel; for insulinoma
80428 - 80430 Growth hormone stimulation panel (e.g., arginine infusion, l-dopa administration) or growth hormone suppression panel (glucose administration)
80434 Insulin tolerance panel; for ACTH insufficiency
80435     for growth hormone deficiency
82943 Glucagon
82946 Glucagon tolerance test
83003 Growth hormone, human (HGH) (somatotropin)
84305 Somatomedin
84436 Thyroxine; total
86277 Growth hormone, human (HGH), antibody
88271 - 88275 Molecular cytogenetics
88280 - 88289 Chromosome analysis
88291 Cytogenetics and molecular cytogenetics, interpretation and report
96372 Therapeutic, prophylactic or diagnostic injection (specify substance or drug); subcutaneous or intramuscular
99601 - 99602 Home infusion/specialty drug administration

HCPCS codes covered if selection criteria are met:

Pegvisomant (Somavert), Somapacitan-beco (Sogroya) – no specific code:

J2940 Injection, somatrem, 1 mg
J2941 Injection, somatropin, 1 mg
S9558 Home injectable therapy; growth hormone, including administrative services, professional pharmacy services, care coordination, and all necessary supplies and equipment (drugs and nursing visits coded separately), per diem

HCPCS codes not covered for indications listed in the CPB:

Q0515 Injection, semorelin acetate, 1 mcg

Other HCPCS codes related to the CPB:

B4164 - B5200 Parenteral nutrition solutions and supplies
B9004 - B9006 Parenteral nutrition infusion pump
S9364 - S9368 Home infusion therapy, total parenteral nutrition (TPN)

ICD-10 codes covered if selection criteria are met (not all-inclusive):

B20 Human immunodeficiency virus (HIV) disease
C71.0 - C71.9 Malignant neoplasm of brain
C75.1 - C75.2 Malignant neoplasm of pituitary gland and craniopharyngeal duct
C79.31 Secondary malignant neoplasm of brain
C79.49 Secondary malignant neoplasm of other parts of nervous system [spinal cord]
D33.0 - D33.2 Benign neoplasm of brain
D35.2 - D35.3 Benign neoplasm of pituitary gland and craniopharyngeal duct (pouch)
D38.1 Neoplasm of uncertain behavior of trachea, bronchus, and lung
D44.3 - D44.4 Neoplasm of uncertain behavior of pituitary gland and craniopharyngeal duct
D49.6 Neoplasm of unspecified behavior of brain
E22.0 Acromegaly and pituitary gigantism
E23.0 Hypopituitarism
E23.1, E89.3 Iatrogenic pituitary disorders
E25.0 - E25.9 Adrenogenital disorders [congenital adrenal hyperplasia]
E34.30, E34.31, E34.321, E34.322, E34.328, E34.329, E34.39 Short stature due to endocrine disorder [SHOX deficiency]
E84.0 - E84.9 Cystic fibrosis
E88.1 Lipodystrophy, not elsewhere classified [excess abdominal fat in HIV-infected persons]
G80.0 - G80.9 Cerebral palsy
K91.2 Postsurgical malabsorption, not elsewhere classified [short-bowel syndrome]
N18.1 - N18.9 Chronic kidney disease (CKD)
N25.0 Renal osteodystrophy
P05.10 - P05.19 Newborn small for gestational age
Q87.11 Prader-Willi syndrome
Q87.19 Other congenital malformation syndromes predominantly associated with short stature [Russell-Silver syndrome]
Q89.2 Congenital malformations of other endocrine glands
Q96.0 - Q96.9 Turner's syndrome
R62.52 Short stature (child) [covered for SHOX deficiency in children whose epiphyses are not closed]
R62.7 Adult failure to thrive
R64 Cachexia [AIDS-related wasting]
T66.xxx+ Radiation sickness, unspecified
Z92.3 Personal history of irradiation

ICD-10 codes not covered for indications listed in the CPB (not all-inclusive):

E20.1 Pseudohypoparathyroidism
E23.6 Other disorders of the pituitary gland, unspecified
E29.1 Testicular hypofunction
E30.1 - E30.8 Disorders of puberty, not elsewhere classified
E34.30, E34.31, E34.321, E34.322, E34.328, E34.329, E34.39 Short stature due to endocrine disorder [Laron syndrome]
E66.0 - E66.9 Overweight and obesity
E72.00 - E72.09 Disorders of amino-acid transport
E83.31 Familial hypophosphatemia
E83.32 Hereditary vitamin D-dependent rickets (type 1) (type 2)
F30.10 - F33.9
F34.8 - F34.9
Episodic mood disorders
F32.3, F33.3 Major depressive disorders
F32.9 Major depressive disorder, single episode not elsewhere classified
F34.1 Dysthymic disorder
F43.0 Acute stress reaction
F43.10 - F43.12 Post-traumatic stress disorder (PTSD)
F64.0 – F64.9 Gender identity disorders [transitioning to male]
G12.21 Amyotrophic lateral sclerosis
G14 Post-polio syndrome
G71.00 - G72.9 Muscular dystrophies and other myopathies
H33.001 – H33.8 Retinal detachments and breaks [injured retina]
H49.811 - H49.819 Kearns-Sayre syndrome
I10 - I16.2 Hypertensive diseases
I20.0 - I20.9 Ischemic heart diseases
I50.20 - I50.43 Congestive heart failure
J96.00 - J96.02 Acute respiratory failure
J96.10 - J96.12 Chronic respiratory failure
J96.20 - J96.22 Acute and chronic respiratory failure
K50.011 - K52.9 Noninfectious enteritis and colitis
K74.69 Other cirrhosis of liver [decompensated cirrhosis]
M08.00 - M08.48 Juvenile arthritis
M24.10 Other articular cartilage disorders, unspecified site [chondral defect repair]
M54.50-M54.59 Low back pain
M75.100 - M75.122 Rotator cuff tear or rupture, not specified as traumatic [healing after rotator cuff repair]
M79.7 Fibromyalgia
M81.0 - M81.8 Osteoporosis without current pathological fracture
M84.750+ - M84.759+ Atypical femoral fracture
N46.01 - N46.9 Male infertility
N97.0 - N97.9 Female infertility
P05.2 Newborn affected by fetal (intrauterine) malnutrition not light or small for gestational age [intra-uterine growth restriction]
Q05.0 - Q05.9 Spina bifida
Q07.00 – Q07.03 Arnold-Chiari syndrome [Chiari malformations]
Q14.2 Congenital malformations of optic disc
Q77.1, Q77.4, Q77.8
Q78.4, Q78.9
Chondrodystrophy
Q78.0 Osteogensis imperfecta
Q89.8 Other specified congential malformations [Charge Syndrome] [Kabuki syndrome]
Q90.0 - Q90.9 Down syndrome
Q98.6 Male with structurally abnormal sex chromosome [isochromosome Yp defect]
R53.82 Chronic fatigue, unspecified
S05.50XA – S05.62XS Penetrating wound with or without foreign body of eyeball [injured retina]
S06.0X0A - S06.A1XS Intracranial injury [traumatic brain injury] [other than associated with pediatric GH deficiency]
S42.001+ - S42.92x+
S52.001+ - S52.92x+
S62.001+ - S62.92x+
Fracture of upper limbs
S72.001+ - S72.92x+
S82.001+ - S82.92x+
S92.001+ - S92.919+
Fracture of lower limbs
T20.00x+ - T25.799+ Burns
T38.0x5+ Adverse effect of glucocorticoids and synthetic analogues
T40.3X1A - T40.3X6S Poisoning by, adverse effect of and under dosing of methadone
T43.605 Adverse effects of unspecified psychostimulants
Z31.83 Encounter for assisted reproductive fertility procedure cycle [transitioning to male]
Z33.1 Pregnant state, incidental
Z39.1 Encounter for care and examination of lactating mother
Z47.81 - Z47.89 Encounter for other orthopedic aftercare [healing after rotator cuff repair]
Z85.47 Personal history of malignant neoplasm of testis
Z87.890 Personal history of sex reassignment [transitioning to male]
Z98.84 Bariatric surgery status

Requirements for GH-Stimulation Testing in Adults:

Other CPT codes related to the CPB / CPT codes covered if selection criteria are met:

80428 - 80430 Growth hormone stimulation panel (e.g., arginine infusion, l-dopa administration) or growth hormone suppression panel (glucose administration)
80435 Insulin tolerance panel; for growth hormone deficiency
82946 Glucagon tolerance test

Lonapegsomatropin-tcgd (Skytrofa):

HCPCS codes covered if selection criteria are met:

Lonapegsomatropin-tcgd (Skytrofa) – no specific code

ICD-10 codes covered if selection criteria are met (not all-inclusive):

E23.0 Hypopituitarism

ICD-10 codes not covered for indications listed in the CPB (not all-inclusive):

D56.0 – D56.9 Thalassemia
E28.8 Other ovarian dysfunction [poor ovarian responders]
E28.9 Ovarian dysfunction, unspecified [poor ovarian responders]
N46.01 - N46.9 Azoospermia
N97.0 – N97.9 Female Infertility

Background

U.S. Food and Drug Administration (FDA)-Approved Indications

  • Somatropin Products - Genotropin, Humatrope, Norditropin, Nutropin AQ, Omnitrope, Saizen, and Zomacton

    • Pediatric patients with growth failure due to any of the following:

      • Growth hormone (GH) deficiency
      • Turner syndrome
      • Noonan syndrome
      • Small for gestational age (SGA)
      • Prader-Willi syndrome
      • Chronic kidney disease (CKD)
      • Short stature homeobox-containing gene (SHOX) deficiency

    • Adults with childhood-onset or adult-onset GH deficiency

  • Serostim (somatropin)

    Serostim is indicated for the treatment of human immunodeficiency virus (HIV) patients with wasting or cachexia to increase lean body mass and body weight, and improve physical endurance. Concomitant antiretroviral therapy is necessary.

  • Sogroya (somapacitan-beco)

    • Sogroya is indicated for the replacement of endogenous growth hormone (GH) in adults with growth hormone deficiency (GHD).
    • Sogroya is indicated for the treatment of pediatric patients aged 2.5 years and older who have growth failure due to inadequate secretion of endogenous growth hormone (GH).

  • Skytrofa (lonapegsomatropin-tcgd)

    Skytrofa is indicated for the treatment of pediatric patients 1 year and older who weigh at least 11.5 kg and have growth failure due to inadequate secretion of endogenous growth hormone.

  • Macrilen (macimorelin)

    Macrilen is a growth hormone (GH) secretagogue receptor agonist indicated for the diagnosis of adult growth hormone deficiency.

  • Somavert (pegvisomant)

    Somavert is indicated for the treatment of acromegaly in patients who have had an inadequate response to surgery or radiation therapy, or for whom these therapies are not appropriate.

Compendial Uses

Somatropin Products - Genotropin, Humatrope, Norditropin, Nutropin AQ, Omnitrope, Saizen, and Zomacton

  • Human immunodeficiency virus (HIV)-associated wasting/cachexia
  • Short bowel syndrome (SBS)
  • Growth failure associated with any of the following:

    • Cerebral palsy
    • Congenital adrenal hyperplasia
    • Cystic fibrosis
    • Russell-Silver syndrome

Growth Hormone (GH)

Growth hormone (GH) promotes linear growth; the somatotropic effects occur partially through stimulation of insulin‐like growth factor I (IGF‐I). IGF‐I produced primarily by the liver circulates throughout the body, whereas IGF‐I produced in the growth cartilage acts locally as a paracrine‐autocrine growth factor. In addition, the diverse metabolic actions of GH include its anabolic and lipolytic effects. GH also induces insulin resistance. GH has now been shown to be produced throughout adult life and to have important physiologic and metabolic effects long after final height has been reached. Short‐term administration of GH promotes lipolysis, stimulates protein synthesis, increases lean body mass, stimulates bone turnover, causes insulin antagonism, alters total body water, and promotes loss of visceral adipose tissue.

Growth hormone has been approved by the U.S. Food and Drug Administration (FDA) for treatment of GH deficiency (GHD) in both children and adults, short stature associated with chronic renal insufficiency (CRI) before renal transplantation, short stature in patients with Turner syndrome (TS), HIV-associated wasting syndrome in adults, idiopathic short stature, treatment of children with short stature associated with Noonan syndrome, short stature homeobox-containing gene deficiency, and treatment of children born small for gestational age (SGA) who fail to manifest catch-up growth.  There are several brands of GH (somatropin) on the market, and there is a lack of reliable evidence that any brand of GH is more effective than others for any indication.

GH Therapy in Adults

In members with hypothalamic‐pituitary disease, the syndrome of adult growth hormone deficiency (AGHD) characteristically presents with alterations in body composition, including reduced lean body mass and bone mineral density and increased fat mass with a preponderance of abdominal adiposity. The skin is thin and dry, and sweating is reduced. Muscle strength and exercise performance are reduced. An impaired sense of well‐being and other psychological complaints are common.

An evaluation for GH deficiency should be considered only in members with evidence of hypothalamic‐pituitary disease, subjects who have received cranial irradiation, or members with childhood onset of GH deficiency.

The diagnosis of AGHD is established by provocative testing of GH secretion. Historically, the insulin tolerance test (ITT) has been the diagnostic test of choice; this test distinguishes GH deficiency from the reduced GH secretion that accompanies normal aging and obesity. However, the ITT is an intravenous test that requires many blood draws over several hours, and because it requires the person to experience hypoglycemia to obtain an accurate result, it can be dangerous for those with coronary or cerebrovascular disease. Thus, the ITT would be considered contraindicated in members with electrocardiographic evidence or history of ischemic heart disease or in members with seizure disorder (Aeterna Zentaris, 2017; Synder, 2019). According to available guidelines, the ITT is not generally recommended for patients older than 65 years of age (NICE, 2003).

In patients in whom insulin-induced hypoglycemia is contraindicated or unsafe or where appropriate testing arrangements are unavailable, the literature states that alternatives to ITT should be used.  Information now states that intravenously administered arginine, either alone or in combination with GH-releasing hormone (GHRH), may be useful.  When only intravenously administered arginine is used, cut-off values for a normal response are similar to those expected with ITT.  When it is used in combination with GHRH, the response may be augmented and the cut-off level is somewhat higher (9 to 10 ng/ml).  Available literature suggests tests that use of glucagon, propranolol, or levodopa has a lesser established diagnostic value in comparison to ITT.  Although useful as a diagnostic procedure in children, the literature states that a test that uses clonidine is of dubious value for the diagnosis of GH deficiency in adults.  In adults with a history of hypothalamic pituitary disease or cranial irradiation, generally only 1 provocative test of GH secretion is needed (NICE, 2003).  In adults with childhood onset isolated GHD (no evidence of hypothalamic pituitary abnormality or cranial irradiation), 2 diagnostic tests should be recommended, except for those having low insulin-like growth factor-1 (1GF-1) (a marker of GH response) concentrations (standard deviation score less than -2) who may be considered for one test (NICE, 2003).

For adult members who have a contraindication to intravenous GH stimulation tests, an orally administered stimulation test has become available. On December 20, 2017, the U.S. FDA approved Macrilen (macimorelin), an orally available growth hormone (GH) secretagogue receptor agonist indicated for the diagnosis of adult growth hormone deficiency (AGHD) (Aeterna Zentaris, 2017; Garcia, 2018; Synder, 2019). Macrilen (macimorelin) stimulates the secretion of GH from the pituitary gland into the circulatory system. Stimulated GH levels are then measured in four blood samples over ninety minutes after the oral administration of Macrilen (macimorelin).

The diagnostic efficacy of macimorelin was established in a multicenter, randomized, open-label, single-dose, two-way cross-over study. Garcia et al. (2018) state that macimorelin could be used to diagnose AGHD by measuring stimulated GH levels after an oral dose. The objective of the study was to validate the efficacy and safety of single-dose oral macimorelin for AGHD diagnosis compared with the ITT. Adult subjects with high (n = 38), intermediate (n = 37), and low (n = 39) likelihood for AGHD and healthy, matched controls (n = 25) were included in the efficacy analysis. For both the ITT and the macimorelin test, serum concentrations of GH were measured at 30, 45, 60, and 90 minutes after drug administration. The test was considered positive (i.e., GH deficiency diagnosed) if the maximum serum GH level observed after stimulation was less than the pre-specified cut point value of 2.8 ng/mL for the macimorelin test or 5.1 ng/mL for the ITT. On retesting, the reproducibility was 97% for macimorelin (n = 33). In post hoc analyses, a GH cutoff of 5.1 ng/mL for both tests resulted in 94% negative agreement, 82% positive agreement, 92% sensitivity, and 96% specificity. No serious adverse events were reported for macimorelin. The authors concluded that oral macimorelin is a simple, well-tolerated, reproducible, and safe diagnostic test for AGHD with accuracy comparable to that of the ITT. This clinical study has established that a maximally stimulated serum GH level of less than 2.8 ng/mL (i.e., at the 30, 45, 60 and 90 minute timepoints) following macimorelin (Macrilen) administration confirms the presence of adult growth hormone deficiency.

The most common adverse reactions were dysgeusia, dizziness, headache, fatigue, nausea, hunger, diarrhea, upper respiratory tract infection, feeling hot, hyperhidrosis, nasopharyngitis, and sinus bradycardia.

Although there are no known contraindications for Macrilen, the labeling includes the following warnings and precautions:

  • QT prolongation, thus, the need to avoid concomitant use with drugs known to prolong QT interval,
  • Potential for false positive test results with use of strong CYP3A4 inducers,
  • Potential for false negative test results in recent onset hypothalamic disease.

A limitation for macimorelin use includes that the safety and diagnostic performance has not been established for subjects with BMI greater than 40 kg/m2.

Studies have shown that over 90% of adults diagnosed with GHD have overt pituitary disease, which is usually caused by a pituitary adenoma or by surgery or radiation therapy for a pituitary adenoma.

The syndrome of GHD characteristically manifests with deficiencies in bone density, reduction in muscle strength and exercise tolerance, decreases in vitality and energy, emotional lability, feelings of social isolation, and increases in body fat and higher serum lipid concentrations.

The usefulness of GH treatment in adults with pituitary disease who have completed their statural growth is based on the role of GH in increasing bone density and in improving mood and motivation.  There is some evidence to suggest that GH therapy improves cardiovascular risk factors and increases bone mineral density (Ball, 2002).  A Committee convened by the National Institute of Clinical Excellence (2003) concluded, however, that it was uncertain what impact GH treatment had on the longer-term clinical outcomes and mortality related to cardiovascular risk factors and changes in bone mineral density.  In addition, there are other more effective, better established and less costly therapies to reduce cardiovascular risk factors and increase bone mineral density.

The NICE Committee concluded that a trial of GH treatment could be recommended for adults with GHD who have a severe perceived impairment of QoL as demonstrated by a reported score of at least 11 in QoL-AGHDA (NICE, 2003).  The Committee agreed that the QoL-AGHDA questionnaire (see Figure 4 of Appendix) is the best available evaluation tool for the assessment of both baseline QoL and the effect of treatment in adults with GHD.  Upon examination of available evidence, the Committee found that the subgroup of adults with GHD for whom treatment may be clinically justified are those who have an improvement in QoL equivalent to an absolute change in their baseline QoL-AGHDA score of at least 7 points.  The Committee stated that re-assessment of the need for GH replacement should take place after a trial treatment period of 9 months (3 months for dose titration and 6 months for assessment of response).  For GH treatment to continue after this trial period, it should be necessary to demonstrate a sustained improvement in QoL.

NICE recommended that adults with childhood GHD must be re-tested as adults before long-term GH replacement is instituted, because some GH-deficient children are found to be GH sufficient in adulthood (NICE, 2003).

Growth hormone levels continue to decline through adulthood, and the proportion of adults who may be considered GH deficient increases with age.  Some investigators have claimed that idiopathic GHD in adults is common and that most cases of idiopathic adult-onset GHD go undetected.  These investigators have promoted GH supplementation as a “rejuvenation” treatment for aging adults with age-related declines in GH levels.  Clinical studies of elderly persons with relatively low levels of endogenous GH have shown small increases in lean body mass and bone mass, as well as improvements in plasma lipid profile with GH supplementation.  However, the long-term oncogenic effects and other potential adverse consequences of GH supplementation in adults with idiopathic GHD are unknown.  In addition, improvements in lean body mass, bone mass, and plasma lipid profile may be better achieved with other treatments in adults with idiopathic GHD.

In adults, the GH response to insulin-induced hypoglycemia is dependent on age, weight, and sex hormones, but most normal adults tested will have a peak GH secretion above 3 ng/ml (when GH is measured in a polyclonal competitive radioimmunoassay).  Thus, values less than 3 ng/ml are considered indicative of GHD.  In children and adolescents, in whom secretion may be more robust and GH effects on growth may require higher levels of secretion than in older patients, values below 10 ng/ml are considered inadequate.

Although serum IGF-I concentrations are related to GH adequacy, accepted guidelines state that the diagnosis of GHD should not rely simply on IGF-I measurements but should be confirmed by provocative tests solely for GH secretion.

In adults with GHD, the FDA-approved labeling states that the starting dosage of GH should be very low (0.1 to 0.4 mg/day).  The product labeling further states that this dose should be increased gradually on the basis of clinical and biochemical responses assessed at monthly intervals.  The biochemical marker generally relied upon for GH is the IGF-I level in serum.  Values of IGF-I should be maintained in the normal age- and sex-adjusted range.  The literature indicates that the dose may be increased, on the basis of individual patient requirements, to a maximum of 1.75 mg daily in patients younger than 35 years of age, and to a maximum of 0.875 mg daily in patients older than 35 years of age.  Of note, this dose is substantially less than GH replacement doses in children and adolescents, in whom the dose is based on weight.

The FDA has approved the use of GH (Zorbtive, Serono Inc., Rockland, MA) for the treatment of short bowel syndrome in patients receiving specialized nutritional support.  According to the FDA-approved labeling, Zorbtive should be used in conjunction with optimal management of short bowel syndrome.  Specialized nutritional support may consist of a high carbohydrate, low-fat diet, adjusted for individual patient requirements and preferences.  Nutritional supplements may be added according to the discretion of the treating physician.  Optimal management of short bowel syndrome may include dietary adjustments, enteral feedings, parenteral nutrition, fluid and micronutrient supplements, as needed.  The FDA approval of Zorbtive was based on the results of a randomized, double-blind, controlled, parallel-group phase III clinical study of GH in subjects with short bowel syndrome (SBS) who were dependent on intravenous parenteral nutrition (IPN) for nutritional support.  The primary endpoint was the change in weekly total IPN volume defined as the sum of the volumes of IPN, supplemental lipid emulsion (SLE), and intravenous hydration fluid.  Subjects received either placebo with the nutritional supplement, glutamine (n = 9), GH without glutamine (n = 16) or GH with glutamine (n = 16).  All 3 groups received a specialized diet.  Following a 2-week equilibration period, treatment was administered in a double-blind manner over a further period of 4 weeks.  The dosing of GH was approximately 0.1 mg/kg/day for 4 weeks.  Mean reductions in IPN volume in each patient group were significantly greater in both the group treated with GH (reduction of 2.1 liter per week compared to placebo plus glutamine) and the group treated with growth hormone plus glutamine (reduction of 3.9 liter per week compared to placebo plus glutamine) than in group treated with placebo plus glutamine.  According to the FDA-approved labeling, Zorbtive should be administered to patients with SBS at a dose of approximately 0.1 mg/kg subcutaneously daily to a maximum of 8 mg daily.  Administration at doses higher than 8 mg per day or for more than 4 weeks has not been adequately studied.  According to the FDA-approved labeling, injections should be administered daily for 4 weeks.  The FDA notes that the safety and effectiveness of Zorbtive in pediatric patients with SBS has not been established.

According to available guidelines, GH therapy is contraindicated in patients with active malignant disease, benign intracranial hypertension (BIH), and proliferative or pre-proliferative diabetic retinopathy.  Potential for child-bearing is not a contraindication, but accepted guidelines caution that GH therapy should be discontinued when pregnancy is confirmed.  The guidelines further caution that GH should not be used in critically ill patients who have acute catabolism.  Growth hormone therapy is also contraindicated in persons with hypersensitivity to GH or its excipients.

On February 1, 2018, Ferring Pharmaceuticals, Inc. announced the U.S. FDA approval of Zomacton (somatropin [rDNA origin]) injection for the replacement of growth hormone (GH) in adults with GH deficiency. Zomacton is already indicated to treat pediatric patients who have growth failure due to inadequate secretion of endogenous GH. Zomacton is available as 5mg and 10mg strength lyophilized powder for subcutaneous (SC) injection after reconstitution in vials (MPR, 2018). 

On August 28, 2020, the FDA approved once-weekly somapacitan-beco (Sogroya) injection for the replacement of endogenous growth hormone in adults with growth hormone deficiency (AGHD). Sogroya is the first human growth hormone (hGH) injection therapy for adults that can be taken once a week. Other FDA-approved hGH formulations for adults with GHD must be administered daily. FDA approval was based on the randomized, parallel-group, placebo-controlled (double-blind) and active-controlled (open-label) phase 3 trial, REAL 1 (Johannsson 2020; NCT02229851) to evaluate the efficacy and safety of somapacitan, a once-weekly reversible albumin-binding GH derivative, versus placebo in AGHD in a clinic-setting in 17 countries. Treatment-naïve patients with AGHD (n = 301 main study period, 272 extension period); 257 patients completed the trial.  Patients were randomized 2:2:1 to once-weekly somapacitan, daily GH, or once-weekly placebo for 34 weeks (main period). During the 52-week extension period, patients continued treatment with somapacitan or daily GH. The main outcome measures included body composition measured using dual-energy x-ray absorptiometry (DXA). The primary endpoint was change in truncal fat percentage to week 34. Insulin-like growth factor 1 (IGF-I) standard deviation score (SDS) values were used to dose titrate. At 34 weeks, somapacitan significantly reduced truncal fat percentage (estimated difference: -1.53% [-2.68; -0.38]; P = 0.0090), demonstrating superiority compared with placebo, and it improved other body composition parameters (including visceral fat and lean body mass) and IGF-I SDS. At 86 weeks, improvements were maintained with both somapacitan and daily GH. Somapacitan was well tolerated, with similar adverse events (including injection-site reactions) compared with daily GH. The authors concluded that in AGHD patients, somapacitan administered once weekly demonstrated superiority over placebo, and the overall treatment effects and safety of somapacitan were in accordance with known effects and safety of GH replacement for up to 86 weeks of treatment. Somapacitan may provide an effective alternative to daily GH in AGHD.

Adverse reactions reported in 2% or more of patients treated with somapacitan are: back pain, arthralgia, dyspepsia, sleep disorder, dizziness, tonsillitis, peripheral edema, vomiting, adrenal insufficiency, hypertension, blood creatine phosphokinase increase, weight increase, anemia. Somapacitan was well tolerated, with similar adverse events (including injection-site reactions) compared with daily GH.

Somapacitan is administered once a week by subcutaneous injection to the abdomen or thigh with regular rotation of injection sites to avoid lipohypertrophy/lipoatrophy. The initiation dose is 1.5 mg once weekly for treatment naïve patients and patients switching from daily growth hormone, increasing the weekly dosage every 2 to 4 weeks by approximately 0.5 mg to 1.5 mg until the desired response has been achieved. The dose should be titrated based on clinical response and serum insulin-like growth factor 1 (IGF-1) concentrations. The maximum recommended dosage is 8 mg once weekly. The full prescribing information contains additional details for dosage recommendations in patients aged 65 years and older, patients with hepatic impairment, and women receiving oral estrogen. 

GH Therapy in Children

Growth hormone deficiency involves abnormally short stature with normal body proportions. Growth hormone deficiency can be categorized as either congenital or acquired. An abnormally short height in childhood may occur if the pituitary gland does not produce enough growth hormone. It can be caused by a variety of genetic mutations (such as Pit‐1 gene, growth hormone receptor gene, growth hormone gene), absence of the pituitary gland, or severe brain injury, but in most cases no underlying cause of the deficiency is found.

The FDA has approved GH for use in the following pediatric conditions: GHD, Turner syndrome, chronic renal insufficiency before transplantation, and children born small for gestational age.  An advisory committee to the FDA also recommended approval of GH for children with idiopathic short stature.

An assessment conducted for the National Institute of Clinical Excellence (2001) suggests the following criteria be used to define subnormal growth in children with GHD:

  1. Decreasing growth rate combined with a predisposing condition such as previous cranial irradiation; or
  2. Evidence of other pituitary hormone deficiencies or signs of congenital GHD (hypoglycemia, microphallus); or
  3. Moderate growth retardation with height SDS for sex and chronological age between -2 and -3 SDS below the mean and decreased growth rate (growth velocity (GV) below 25th percentile for age and sex); or
  4. Severe deceleration in growth rate (GV below 3rd percentile for age and sex); or
  5. Severe growth retardation with height standard deviation score (SDS) for sex and chronological age less than 3 SDS below the mean.

In addition, retardation of bone maturation is found in most cases of subnormal growth.

Diagnosis of GHD in children is confirmed by measurements of GH secretion, commonly in several samples following stimulation by a provocative agent such as insulin or clonidine (NICE, 2001). The literature states that the standard method of assessing GH secretion in children is to measure the serum GH response to insulin and other stimuli.  Another method is to make frequent measurements of serum GH during the day and night, but this is no more effective than the standard method for detecting GHD.  Formerly, the diagnosis of GHD in children was based on a peak serum GH concentration of 5 ng/ml or less in response to a provocative test, but a peak serum GH concentration of less than 10 ng/ml is now considered abnormal.  However, because the available GH assays have not been standardized, the literature states that the cut-off value of less than 10 ng/ml is of limited usefulness, especially in borderline cases.  Instead, accepted guidelines indicate the diagnosis should be based on very short height, as defined by the standard-deviation score (more than 2.0 standard deviations below the mean height for normal children of the same age), delayed bone age, poor growth velocity (less than the 25th percentile), and predicted adult height substantially below the mean parental height.  When used in conjunction with these measures, however, the guidelines suggest a peak serum GH value of less than 10 ng/ml in response to stimulation is a reasonable definition of GHD, with values less than 5 ng/ml reflecting the most severe deficiency.

If thyroxine is insufficient, then the literature indicates tests of GH secretion should be postponed until the thyroid deficiency is adequately replaced because GH secretion may be subnormal merely as a result of the hypothyroidism.  If GHD is suspected in a peripubertal person with a growth pattern that resembles constitutional delay of growth and development, sex steroid priming before testing of GH secretion has been recommended by some investigators.

Other markers of GH secretion, such as concentrations of serum insulin-like growth factor 1 (IGF-1) and insulin-like growth factor-binding protein 3 (IGFBP-3), are not consistently abnormal in children with GHD.

Available literature states that GH stimulation tests and indirect measures of determining endogenous GH secretion (measurement of serum IGF-1 and IGFBP-3 or urinary GH) are often subject to questionable specificity, false failure rates, and lack of published age- and sex-specific normal ranges.  Due to the inadequacies of these tests, a subnormal growth velocity often becomes the deciding factor in choosing to initiate GH therapy.  However, available literature indicates that measurement of short-term growth velocity is unreliable in predicting future growth.  Growth velocity during the autumn and winter may be lower than that during the rest of the year by more than 2 cm/year, and may be normal if less than 2.5 cm/year during these cooler seasons.  Therefore, for valid measurements, accepted guidelines provide that growth velocity should be tracked over an entire year.

Because of its pronounced anabolic effects, GH is contraindicated in children with an active malignant condition.  There is controversy over whether it is safe to administer GH to children in the year or two following treatment for leukemia, medulloblastomas, ependymomas, or other tumors.

Growth hormone treatment in children with childhood-onset GHD is generally begun with a dosage of GH of 0.15 to 0.3 mg/kg per week given 6 or 7 times weekly.  A maintenance dosage of up to 0.30 mg/kg of body weight is frequently recommended.  Treatment is continued until the handicap of short stature is ameliorated, until epiphyseal closure has been recorded, or until the patient is otherwise no longer responding to GH treatment.

In August 2021, the U.S. FDA approved Skytrofa (lonapegsomatropin-tcgd) (Ascendis Pharma Inc.) for the treatment of pediatric patients one year and older who weigh at least 11.5 kg (25.4 lb) and have growth failure due to inadequate secretion of endogenous growth hormone (GH). Skytrofa is a pegylated human growth hormone (sustained release somatropin) for once-weekly subcutaneous injection. FDA approval was based on results from the phase 3 heiGHt Trial, a 52-week, global, randomized, open-label, active-controlled, parallel-group trial that compared once-weekly Skytrofa to daily somatropin (Genotropin®) in 161 treatment-naïve children (age range 3.2 to 13.1 yrs) with growth hormone deficiency (GHD);105 subjects received once-weekly Skytrofa, and 56 received daily somatropin. The dose in both arms was 0.24 mg/kg/week. The primary efficacy endpoint was annualized height velocity (AHV) at Week 52. Treatment with once-weekly Skytrofa for 52 weeks resulted in an annualized height velocity of 11.2 cm/year. Subjects treated with daily somatropin achieved an annualized height velocity of 10.3 cm/year after 52 weeks of treatment. In summary, at week 52, the treatment difference in AHV was 0.9 cm/year (11.2 cm/year for Skytrofa compared with 10.3 cm/year for daily somatropin) with a 95 percent confidence interval [0.2, 1.5] cm/year. The primary objective of non-inferiority in AHV was met for Skytrofa in this trial and further demonstrated a higher AHV at week 52 for lonapegsomatropin compared to daily somatropin, with similar safety, in treatment-naïve children with GHD. No serious adverse events or discontinuations related to Skytrofa were reported. Most common adverse reactions (≥ 5%) in pediatric patients include: infection, viral (15%), pyrexia (15%), cough (11%), nausea and vomiting (11%), hemorrhage (7%), diarrhea (6%), abdominal pain (6%), and arthralgia and arthritis (6%). In addition, both arms of the study reported low incidences of transient, non-neutralizing anti-hGH binding antibodies and no cases of persistent antibodies. Therapy with Skytrofa should be supervised by a physician who is experienced in the diagnosis and management of pediatric patients with growth failure due to GH deficiency. Skytrofa auto-injector for subcutaneous use can be administered by a trained caregiver. The recommended dose is 0.24 mg/kg body weight, given once-weekly, in which dosing is individualized and titrated based on patient response.

In April 2023, the FDA approved a label expansion for somapacitan-beco (Sogroya) injection for the treatment of pediatric patients aged 2.5 years and older who have growth failure due to inadequate secretion of endogenous growth hormone (GH). FDA approval is based on data from the phase 3, randomized, open-label, active-controlled, parallel-group REAL4 study. In the study, 200 treatment-naïve patients aged 2.5 to11 years with GHD were either given once-weekly somapacitan 0.16 mg/kg/week (n=132) or daily somatropin 0.034 mg/kg/day (n=68) for 52 weeks. The results showed that somapacitan was comparable to daily somatropin for the primary endpoint of annualized height velocity (11.2 cm/year vs. 11.7 cm/year, respectively). Adverse reactions in the REAL4 study occurring in more than 5% of patients included nasopharyngitis, headache, pyrexia, pain in extremity, and injection site reactions (Novo Nordisk, 2023a, 2023b).

Miller et al (2022) conducted a randomised, multinational, open-labeled, active-controlled parallel group phase 3 trial, comprising a 52-week main trial [REAL4] and 3-year extension in order to demonstrate efficacy and safety of somapacitan vs daily GH. The study included 200 treatment-naïve patients who were randomized 2:1 to somapacitan (0.16 mg/kg/wk) or daily GH (Norditropin; 0.034 mg/kg/d), administered subcutaneously. The primary endpoint was annualized height velocity (HV; cm/y) at week 52. Additional assessments included HV SD score (SDS), height SDS, bone age, insulin-like growth factor-1 (IGF-1) standard deviation score (SDS), patient-reported outcomes, and safety measures. The authors found that the estimated mean HV at week 52 was 11.2 and 11.7 cm/y for somapacitan and daily GH, respectively. Thus, noninferiority was confirmed. Changes in HV SDS, height SDS, bone age, and IGF-1 SDS from baseline to week 52 were similar between treatment groups. At week 52, mean IGF-1 SDS values were similar between treatment groups and within normal range (-2 to +2). Safety of somapacitan was consistent with the well-known daily GH profile. Low proportions of injection-site reactions were reported for somapacitan (5.3%) and daily GH (5.9%). Both treatments similarly reduced disease burden from baseline to week 52, whereas a greater treatment burden reduction was observed for somapacitan. The authors concluded that there was similar efficacy for somapacitan compared to daily GH was demonstrated over 52 weeks of treatment with comparable safety and mean IGF-1 SDS levels in treatment-naïve children with GHD. 

Acromegaly

Acromegaly is a potentially life-threatening disease triggered by an excess of GH.  Symptoms include headaches, profuse sweating, swelling, joint disorders, changes in facial features, as well as enlarged hands, feet and jaw.  If untreated, patients with acromegaly often have a shortened life-span because of heart and respiratory diseases, diabetes mellitus and cancer.

Acromegaly results from excessive secretion of growth hormone and elevated levels of IGF‐1. The usual cause of acromegaly is adenomas of the pituitary gland. Symptoms include headaches, profuse sweating, swelling, changes in facial features, joint disorders, and enlarged hands feet and jaw.

Goals of acromegaly management include controlling GH and IGF‐1 secretion and tumor growth, relieving central compressive effects, preserving or restoring pituitary function, treating co‐existing illnesses, preventing premature death, and preventing disease recurrence. Surgery is the preferred approach for treating most patients with acromegaly. Serum GH levels are controlled within an hour after complete removal of the GH‐secreting adenoma. Somatostatin receptor ligands (i.e. octreotide or lanreotide) are the first‐choice pharmacotherapy for treating patients who have acromegaly.

Nadir GH serum levels should be below 1 mg/L, preferably less than 0.4mg/L, in the two hours after 75‐g oral glucose load (oral glucose tolerance test [OGTT]). This criterion often is used for diagnosis of acromegaly and for follow‐up during treatment.

Pegvisomant is available as Somavert. Pegvisomant is a growth hormone receptor antagonist, a recombinant analog of human growth hormone. Pegvisomant selectively binds to growth hormone (GH) receptors on cell surfaces, where it blocks the binding of endogenous GH and thus interferes with GH signal transduction. Inhibition of GH action results in decreased serum concentrations of insulin‐like growth factor‐1 (IGF‐1), as well as other GH‐responsive serum proteins.

Somavert (pegvisomant) is indicated for the treatment of acromegaly in patients who have had an inadequate response to surgery or radiation therapy, or for whom these therapies are not appropriate. The goal of treatment is to normalize serum IGF‐I levels.

In 2003, the FDA approved pegvisomant (Somavert) for the treatment of acromegaly in patients who have had an inadequate response to existing therapies.  Pegvisomant, a polyethylene glycol derivative of human GH, is the first in a new class of drugs called GH receptor antagonists.  It competes with endogenous GH for the receptor and results in suppression of serum insulin-like growth factor (IGF-1).  Clinical studies have shown that pegvisomant normalized concentrations of IGF-I in more than 90 % of patients by blocking the effects of GH.  The most commonly reported adverse effects with pegvisomant were injection site reactions, sweating, headache and fatigue.

A pituitary MRI should be preformed every six months. Serum GH levels have been known to increase as well as persistent tumor growth in patients taking pegvisomant.

Liver function test should be preformed before and monthly during the first six months of treatment and, thereafter, every six months. Idiosyncratic chronic active hepatitis, with elevated transaminases more than three times above the upper normal range, has been reported in 9% of patients receiving pegvisomant for more than a year.

Acromegalic patients with diabetes mellitus being treated with insulin and/or oral hypoglycemic agents may require dose reductions of these therapeutic agents after the initiation of therapy with pegvisomant.

Concomitant use with a somatostatin analogue (octreotide acetate) has been shown to increase the risk of marked hepatic enzyme elevations (greater than 10 times the upper limit of normal)

Pegvisomant is available as Somavert in single‐dose vials in the following strengths: 10, 15, 20, 25, and 30mg. The recommended dose for pegvisomant is a 40 mg subcutaneous loading dose followed by daily subcutaneous injections of 10 mg. Serum IGF‐1 levels should be measured every four‐to‐six weeks at which time the dosage of pegvisomant should be adjusted in 5 mg increments if IGF‐1 levels are still elevated. The maximum daily maintenance dose should not exceed 30 mg.

AIDS-Related Wasting

Growth hormone has been approved as a treatment for AIDS wasting syndrome. AIDS wasting syndrome is defined as the involuntary loss of 10% of body weight plus 30 days of either diarrhea, or weakness and fever.

Cerebral Palsy

Children with cerebral palsy (CP) often have poor linear growth during childhood, resulting in a diminished final adult height. Coniglio, et al. (1996) reported that six of 10 children with cerebral palsy (CP) and growth failure had subnormal GH secretion consistent with GH deficiency. The authors observed that subnormal growth velocity was the best clinical predictor of GH deficiency. The authors exclaimed that "the large percentage of these children with apparent GH deficiency is surprising." The authors posited that possible mechanisms include anatomic abnormalities of the hypothalamic-pituitary axis, psychosocial deprivation, and an interaction between suboptimal nutritional status and an abnormal central nervous system.

Shim, et al (2004) reported a female with CP and short stature but without growth hormone (GH) deficiency who exhibited increased growth during treatment with GH. The authors also reported two other children with CP who were treated with GH: one female with a history of leukemia, and a male with Klinefelter syndrome. These two children were both found to be GH-deficient by insulin provocative GH testing and responded to treatment with increased growth rate. The authors reported that growth improved to a greater extent in the two children with apparent GH deficiency. The authors stated that they felt that GH therapy might be beneficial for children with CP and warrants further investigation.

Chronic Kidney Disease

Adema and colleagues (2018) stated that chronic kidney disease (CKD)-associated decline in soluble α-Klotho (α-Klotho) levels is considered detrimental.  Some studies suggested a direct induction of α-Klotho concentrations by GH.  In a prospective, single-center, open case-control, pilot study, these researchers examined the effect of exogenous GH administration on α-Klotho concentrations in a clinical cohort with mild CKD and healthy subjects.  This trial was performed involving 8 patients with mild CKD and 8 healthy controls matched for age and sex.  All participants received subcutaneous GH injections (Genotropin, 20 mcg/kg/day) for 7 consecutive days; α-Klotho concentrations were measured at baseline, after 7 days of therapy and 1 week after the intervention was stopped.  α-Klotho concentrations were not different between CKD-patients and healthy controls at baseline (554 (388 to 659) versus 547 (421 to 711) pg/ml, p = 0.38).  Overall, GH therapy increased α-Klotho concentrations from 554 (405 to 659) to 645 (516 to 754) pg/ml, p < 0.05).  This was accompanied by an increase of IGF-1 concentrations from 26.8 ± 5.0 nmol/L to 61.7 ± 17.7 nmol/L (p < 0.05).  GH therapy induced a trend toward increased α-Klotho concentrations both in the CKD group (554 (388 to 659) to 591 (358 to 742) pg/ml (p = 0.19)) and the healthy controls (547 (421 to 711) pg/ml to 654 (538 to 754) pg/ml (p = 0.13)).  The change in α-Klotho concentration was not different for both groups (p for interaction = 0.71).  α-Klotho concentrations returned to baseline levels within 1 week after the treatment (p < 0.05).  The authors concluded that GH therapy increased α-Klotho concentrations in subjects with normal renal function or stage 3 CKD.  It is unclear if this could also be accomplished in more advanced CKD.  They stated that additional studies are needed to determine whether the effect size is different between both groups or in patients with more severe CKD, and whether the increase of α-Klotho concentrations improves intermediate end-points and subsequently patient-level outcome.

The authors stated that besides the small sample size of this study (n = 8 for the GH therapy group), there were several drawbacks that need to be underlined.  First, the exclusion criteria for participants limited generalizability, in particular for patients with more advanced CKD.  Second, the specificity of the IBL-assay used to measure α-Klotho concentration was disputed.  These researchers did not use the semi-quantitative precipitation-immunoblotting technique as described by Barker et al, which probably has improved specificity.  This method awaits external validation in a different cohort and by different laboratories.  Moreover, these investigators recently found that the ELISA used in this study performs best among currently commercially available immunoassays.  Unfortunately, they were not able to assess the influence of GH therapy on membrane-bound α-Klotho due to the absence of kidney biopsies in this study.  Finally, a study of longer duration is needed to determine the more long-term effects of GH on α-Klotho concentrations in the CKD population, and establish a dose-response effect.  This study however was designed as a proof-of-concept to study the modifiability of α-Klotho by GH.

Furthermore, UpToDate reviews on “Overview of the management of chronic kidney disease in adults” (Rosenberg, 2018) and “Overview of the management of chronic kidney disease in children” (Srivastava and Warady, 2018) do not mention growth hormone as a therapeutic option.

Chronic Renal Insufficiency (CRI)

Growth delay in children with CRI may result from numerous physiologic derangements, including acidosis, secondary hyperparathyroidism, malnutrition, or zinc deficiency.  Before initiation of GH treatment in patients with CRI, existing metabolic derangements (such as acidosis, secondary hyperparathyroidism, and malnutrition) should be corrected.  Growth failure in children with CRI is thought to be due to be multi-factorial, with one of the factors being reduced sensitivity to GH rather than GH insufficiency.  The dose of GH generally recommended for children with CRI (0.045 to 0.050 mg/kg/day) is higher than that for children with classic GHD.

Congenital Adrenal Hyperplasia

In a discussion of "experimental medical therapy," an UpToDate review of treatments for congenital adrenal hyperplasia in infants and children (Merke, 2018) explained that, "because short stature commonly occurs in spite of good adrenal hormonal control during childhood and puberty, exogenous growth hormone therapy has been given to improve linear growth and adult height in patients with congenital adrenal hyperplasia. In some cases, gonadotropin-releasing hormone agonist therapy has been added to block excess androgen activity that promotes premature epiphyseal fusion."

In a discussion of "experimental therapies", Endocrine Society guidelines on CAH (2018) state that "individuals with CAH can achieve normal adult height through judicious use of standard GC [glucocorticoid] and MC [mineralocorticoid] therapies, and height-enhancing drugs are to be considered only for individuals whose heights are, or are expected to be, significantly shorter than those of peers, defined as a height of at least −2.25 SDS. We advocate further prospective, randomized, and carefully controlled studies to determine whether the use of growth-promoting drugs increases adult height in individuals with CAH."

Cystic Fibrosis

Cystic fibrosis is an inherited condition causing disease most noticeably in the lungs, digestive tract and pancreas. People with cystic fibrosis often have malnutrition and growth delay. Adequate nutritional supplementation does not improve growth optimally and hence recombinant growth hormone has been proposed as a potential intervention. Short-term acceleration of growth as a result of GH therapy has also been reported in children with cystic fibrosis. However, no studies have prospectively assessed linear growth until achievement of final height. 

Thaker, et al (2018) conducted a systematic evidence review of the effectiveness and safety of recombinant human growth hormone therapy in improving lung function, quality of life and clinical status of children and young adults with cystic fibrosis. The investigators searched the Cochrane Cystic Fibrosis and Genetic Disorders Group's Trials Register comprising references identified from comprehensive electronic database searches and handsearches of relevant journals and abstract books of conference proceedings. The investigators also searched ongoing trials registers in clinicaltrials.gov from the United States and WHO International Clinical Trials Registry Platform (ICTRP). The investigators conducted a search of relevant endocrine journals and proceedings of the Endocrinology Society meetings using Web of Science, Scopus and Proceedings First. The investigators identified randomized and quasi‐randomized controlled trials of all preparations of rhGH compared to either no treatment, or placebo, or each other at any dose (high‐dose and low‐dose) or route and for any duration, in children or young adults (aged up to 25 years) diagnosed with CF (by sweat test or genetic testing). Two authors independently screened papers, extracted trial details and assessed their risk of bias. They assessed the quality of the evidence using the Grading of Recommendations Assessment, Development and Evaluation (GRADE) system.

The investigators included eight trials (291 participants, aged between five and 23 years) in this review. Seven trials compared standard‐dose rhGH (approximately 0.3 mg/kg/week) to no treatment and one three‐arm trial (63 participants) compared placebo, standard‐dose rhGH (0.3 mg/kg/week) and high‐dose rhGH (0.5 mg/kg/week). Six trials lasted for one year and two trials for six months. They found that rhGH treatment may improve some of the pulmonary function outcomes but there was no difference between standard and high‐dose levels (low‐quality evidence, limited by inconsistency across the trials, small number of participants and short duration of therapy). The trials show evidence of improvement in the anthropometric parameters (height, weight and lean body mass) with rhGH therapy, again no differences between dose levels. They found improvement in height for all comparisons (very low‐ to low quality evidence), but improvements in weight and lean body mass were only reported for standard‐dose rhGH versus no treatment (very low‐quality evidence). There is some evidence indicating a change in the level of fasting blood glucose with rhGH therapy; however, it did not cross the clinical threshold for diagnosis of diabetes in the trials of short duration (low‐quality evidence). There is low‐ to very low‐quality evidence for improvement of pulmonary exacerbations with no further significant adverse effects, but this is limited by the short duration of trials and the small number of participants. One small trial provided inconsistent evidence on improvement in quality of life (very low‐quality evidence). There is limited evidence from three trials in improvements in exercise capacity (low‐quality evidence). None of the trials have systematically compared the expense of therapy on overall healthcare costs. The authors concluded that, when compared with no treatment, rhGH therapy is effective in improving the intermediate outcomes in height, weight and lean body mass. Some measures of pulmonary function showed moderate improvement, but no consistent benefit was seen across all trials. There was no consistent evidence that growth hormone treatment increased muscle strength or improved quality of life. The significant change in blood glucose levels, although not causing diabetes, emphasizes the need for careful monitoring of this adverse effect with therapy in a population predisposed to CF‐related diabetes. No significant changes in quality of life, clinical status or side‐effects were observed in this review due to the small number of participants. The authors stated that long‐term, well‐designed randomized controlled trials of rhGH in individuals with CF are required prior to routine clinical use of rhGH in CF. "Given these results, we are not able to identify any clear benefit of therapy and believe that more research from well-designed, adequately powered clinical trials is needed."

Prader-Willi Syndrome

Prader-Willi syndrome (PWS) consists of hypothalamic obesity, short stature, developmental delay, hypogonadotropic hypogonadism, small hands and feet, and hypotonia.  The hypothalamic disorder may result in impaired GH secretion in some patients.  Studies have shown that GH appears to have beneficial effects on growth velocity of pediatric patients with PWS.  Clinical studies have also shown that GH supplementation in PWS has a positive impact on body composition, with increases in lean mass and decreases in percent body fat.  The FDA has approved Genotropin brand of GH for the “long-term treatment of pediatric patients who have growth failure due to Prader-Willi syndrome”.  A number of randomized controlled clinical studies have reported significant increases in height velocity in PWS children treated with GH.  One uncontrolled study has reported on final height in a small group of treated children with PWS.  The study reported final height of 170 cm in males and 159 cm in females.  These heights are well within the normal range.  Presuming a treatment effect based on the change in standard deviation (SD) from the start of treatment to the completion of treatment, there was a change of 1.64 SD.  Converting this SD improvement to cm in adult height, this corresponds to treated males being approximately 11 cm and treated females being approximately 9.8 cm taller than the presumptive height of untreated children.

Children with PWS are considered to have a hypothalamic disorder, and thus GH therapy is intended to replace physiological levels of GH.  The recommended dose of GH therapy for children with PWS is 0.035 mg/kg/day.

According to the FDA-approved labeling, GH should only be used in the long-term treatment of pediatric patients with genetically confirmed PWS.  The FDA has received reports of fatalities after the initiation of somatropin therapy in pediatric patients with PWS and having one or more risk factors, including severe obesity, history of upper airway obstruction or sleep apnea, and unidentified respiratory infection.  Male sex may confer added risk to those with one or more of these risk factors.  The FDA-approved labeling of Humatrope (Eli Lilly Co.) was revised to state that GH is contraindicated in patients with PWS who are severely obese or have severe respiratory impairment.  The FDA recommends that patients with PWS be evaluated for signs of upper airway obstruction and sleep apnea prior to therapy initiation.  Treatment should be interrupted in patients showing signs of upper airway obstruction (including onset of increased snoring) and/or sleep apnea.  All patients with PWS being treated with GH should be managed effectively for weight control and monitored for signs of respiratory infection.  The FDA emphasizes the need for early diagnosis and aggressive treatment of these infections.

As of February 2018, Norditropin (somatropin) injection is FDA-approved for the treatment of pediatric patients with growth failure due to Prader-Willi syndrome (Novo Nordisk, 2018).

Short Stature Homeobox-Containing Gene (SHOX) Deficiency

SHOX is located on the distal ends of the X and Y chromosomes encoding a homeodomain transcription factor responsible for a significant proportion of long-bone growth.  Children with mutations or deletions of SHOX, including those with TS who are haplo-insufficient for SHOX have variable degrees of growth impairment, with or without a spectrum of skeletal anomalies consistent with dyschondrosteosis.

Blum et al (2007) examined the effectiveness of GH therapy in treating short stature associated with SHOX deficiency (SHOX-D).  A total of 52 pre-pubertal children (24 males, 28 females; age of 3.0 to 12.3 years) with a molecularly proven SHOX gene defect and height below the 3rd percentile for age and gender (or height below the 10th percentile and height velocity below the 25th percentile) were randomized to either a GH-treatment group (n = 27) or an untreated control group (n = 25) for 2 years.  To compare the GH treatment effect between patients with SHOX-D and those with TS, a third study group, 26 patients with TS aged 4.5 to 11.8 years, also received GH.  Between-group comparisons of 1st-year and 2nd-year height velocity, height sd score, and height gain (cm) were performed using analysis of co-variance accounting for diagnosis, sex, and baseline age.  The GH-treated SHOX-D group had a significantly greater 1st-year height velocity than the untreated control group (mean +/- se, 8.7 +/- 0.3 versus 5.2 +/- 0.2 cm/year; p < 0.001) and similar 1st-year height velocity to GH-treated subjects with TS (8.9 +/- 0.4 cm/year; p = 0.592).  Growth hormone-treated subjects also had significantly greater 2nd-year height velocity (7.3 +/- 0.2 versus 5.4 +/- 0.2 cm/year; p < 0.001), 2nd-year height sd score (-2.1 +/- 0.2 versus -3.0 +/- 0.2; p < 0.001) and 2nd-year height gain (16.4 +/- 0.4 versus 10.5 +/- 0.4 cm; p < 0.001) than untreated subjects.  The authors noted that patients with SHOX-D demonstrated marked, highly significant, GH-stimulated increases in height velocity and height SDS during the 2-year study period.  The effectiveness of GH therapy in children with SHOX-D was equivalent to that observed in children with TS. They concluded that GH is effective in improving the linear growth of patients with various forms of SHOX-D.

Small for Gestational Age (SGA)

In July 2001, Genotropin received approval as an orphan by the FDA for “long-term treatment of growth failure in children who were born small for gestational age (SGA) who fail to manifest catch-up growth by age 2”.  Studies that were presented to the FDA and published controlled clinical trials have been relatively short-term (2 to 6 years) and show, as would be expected, some normalization (“catch up”) of growth of children born small for gestational age.  Short-term clinical studies have shown that, while GH administration induces catch-up growth in SGA children, it also increases skeletal maturation, so that little or no gain in final adult height would be expected (Stanhope et al, 1991; Zeghir et al, 1996; Coutant et al, 1998; Vance and Mauras, 1999).

The first randomized controlled clinical trial of GH treatment for SGA children reporting on final adult height showed that GH supplementation had induced catch-up growth, but a relatively small increase in final adult height that was less than the child's genetic potential.  Carel et al (2003) reported on a study of 168 short children born SGA who were randomized to receive either GH supplementation until attainment of adult height or no treatment.  The investigators report that this study differs from previous published studies of GH therapy for SGA children in that this is the first published randomized controlled clinical study that reports on final adult heights.  In addition, this study differs from previous studies in that SGA children with GHD were excluded.

The investigators reported that the adult heights of GH-treated SGA patients were greater than those of control patients, with a difference of 0.6 standard deviation score (SDS) units (95 % confidence interval [CI]: 0.2 to 0.9) between groups (Carel et al, 2003).  Although the gain in height statistically significant, it is small and treated SGA children remain relatively short compared to peers of normal stature.  In this study, the difference observed between treated and control children was 2.7 cm [1.06 inches] in boys and 4.2 cm [1.65 inches] in girls.

The observed effect of GH supplementation on final adult height in patients born small for gestational age was no greater than the reported effect GH supplementation on the final adult height of patients with idiopathic short stature (Carel et al, 2003).  The investigators summarize published studies of GH supplementation in children with idiopathic short stature that show differences in adult height between treated and untreated children ranging from 0.6 SDS to 1.3 SDS.

In this study, SGA children initiated GH treatment at a mean age of 10.5 years in girls and 12.5 years in boys, and the duration of treatment varied between 6 months and 3.5 years (Carel et al, 2003).  The investigators reported that although the treatment duration was shorter than in other studies of GH supplementation for SGA children, the dose of GH supplementation was about 50 % higher than used in most other studies, the effects of GH supplementation tend to decrease with duration of therapy, and the overall results were similar to other studies of GH supplementation of SGA children.  The investigators concluded that although GH supplementation increases final adult height in SGA children, the children remain short relative to their peers, and the clinical significance of this relatively small increase in height in improving the child's functional capacity, self perception and self-esteem is unclear.

In addition, the long-term effects of GH supplementation children born small of gestational age are unknown.  Root (2002) explained that children born small for gestational age are at greatly increased risk for the development of insulin resistance and hyperinsulinism, which is associated with the “metabolic syndrome” of impaired carbohydrate tolerance progressing to type 2 diabetes mellitus, dyslipidemia, hypertension, increased mortality due to coronary artery disease, and in female hyperandrogenism and the polycystic ovarian syndrome.  Growth hormone therapy is also associated with insulin resistance and hyperinsulinism in intra-uterine growth retardation (IUGR) children and other subjects, although these effects may be reversible.  “Interestingly, the development of type 2 diabetes mellitus is not only related to subnormal fetal growth but also to increased rates of linear growth between 7 and 15 years of age.  It is precisely at this age that [growth hormone] is to be administered to subjects with IUGR to increase the rate of linear growth, potentially increasing still further their risk for development of type 2 diabetes mellitus.”

In an editorial, Silverstein and Shulman (2003) explained: "The use of [growth hormone] has only recently been approved for use in SGA children with short stature and normal growth hormone responses; hence, its use will likely increase in this population.  Its effect on long-term insulin sensitivity in an already at-risk population and later development of type 2 diabetes is not yet known.  In a study of more than 23,000 children registered in a pharmaco-epidemiological survey of GH-treated patients, Cutfield et al (2000) found an increase in type 2 diabetes (using American Diabetes Association criteria) 6-fold greater than expected for the background populations.  This risk may be even greater, more than 20-fold, when ethnically similar populations are used for comparison, and the less stringent World Health Organization criteria for abnormality are applied.  These observations are cautionary in light of the now well-established risk of adult type 2 diabetes in low-birth-weight-for-age.  Several studies have demonstrated an increased risk of insulin resistance and type 2 diabetes in adults who were small at birth.  One study of 23,000 healthy U.S. men found a 2-fold increased risk of type 2 diabetes if they were SGA".

The authors concluded that “[l]onger-term studies comparing the risk of type 2 DM and associated co-morbidities in rhGH-treated SGA children to untreated SGA children will be needed” and “[o]nly with extended follow-up can we be assured that the structural benefits outweigh the possible long-term risks” (Silverstein and Shulman, 2003).

Stanhope (2000) commented on the use of GH in children born SGA and intrauterine growth retardation (IUGR) children: "More than any other condition associated with short stature and treated with GH particular caution should be applied to the long-term sequelae of children with IUGR.  There is now convincing evidence that IUGR is a predisposing factor to the development of hypertension, diabetes and cardiovascular disease in adult life.  As the dose of administered GH needs to be pharmacological, and in the order of two or three times replacement dose, long-term follow-up of such treated children into old age will be required for absolute reassurance that high dose GH treatment throughout childhood and adolescence is safe".

Rapaport (2002) has noted, however, that the concern about insulin resistance has been shown not to result in abnormal glucose level or diabetes after as long as 6 years of treatment.  Rapaport (2002) cited the results of a study that showed that insulin sensitivity parameters adversely affected during treatment with GH revert to normal within 3 months of treatment.  Rapaport also noted that GH treatment of up to 6 years has not been shown to increase lipid levels or substantially increase blood pressure.

Root (2002) noted that the benefits of GH supplementation on the psychological well-being of person's growth small for gestational age are unknown.  “There are as yet no data demonstrating any beneficial effect of treatment on their psychological well-being, educational advancement, or vocational attainment” (Root, 2002).

According to the FDA-approved labeling for Genotropin brand of GH, the recommended dose of GH for SGA patients is 0.48mg/kg body weight per week (Pharmacia, 2003).  Van Pareren et al (2003), however, reported on the results of a randomized controlled dose-ranging study of GH in SGA children, and found no significant differences in outcomes of adult height SDS or gain in height between patients assigned to GH at the recommended dose of 0.48 mg/kg per week and patients assigned to GH therapy at half the usual recommended dose or 0.24 mg/kg per week.

The FDA has approved GH for the treatment of children with short stature associated with Noonan syndrome.  The FDA approval was based upon the results of a 2-year long prospective, open label, randomized, parallel group trial of GH in 21 children with short stature associated with Noonan syndrome.  An additional 6 children were not randomized, but did follow the protocol.  After the initial 2-year trial, children continued on Norditropin until final height.  Retrospective final height and adverse event data were collected from 18 of the 21 subjects who were originally enrolled in the trial and the 6 who had followed the protocol without randomization.  Historical reference materials of height velocity and adult height analyses of Noonan patients served as the controls.  The 24 children (12 females and 12 males) aged 3 to14 years received either 0.033 mg/kg/day or 0.066 mg/kg/day of GH subcutaneously which, after the first 2 years, was adjusted based on growth response.  In addition to a diagnosis of Noonan syndrome, key inclusion criteria included bone age determination showing no significant acceleration, prepubertal status, height SDS less than or equal to 2, and height velocity SDS less than 1 during the 12 months pre-treatment.  Exclusion criteria were previous or ongoing treatment with GH, anabolic steroids or corticosteroids, congenital heart disease or other serious disease perceived to possibly have major impact on growth, fasting plasma glucose greater than 120 mg/dL, or GHD.  Patients obtained a final height  gain from baseline of 1.5 and 1.6 SDS estimated according to the national and the Noonan reference, respectively.  A height gain of 1.5 SDS (national) corresponds to a mean height gain of 9.9 cm in boys and 9.1 cm in girls at 18 years of age, while a height gain of 1.6 SDS (Noonan) corresponds to a mean height gain of 11.5 cm in boys and 11.0 cm in girls at 18 years of age.  A comparison of height velocity between the 2 treatment groups during the first 2 years of treatment for the randomized subjects was 10.1 and 7.6 cm/year with 0.066 mg/kg/day versus 8.55 and 6.7 cm/year with 0.033 mg/kg/day, for year 1 and year 2, respectively.  Age at start of treatment was a factor for change in height SDS (national reference).  The younger the age at start of treatment, the larger the change in height SDS.  Examination of gender subgroups did not identify differences in response to GH.  The FDA-approved labeling for Norditropin brand of GH indicates that not all patients with Noonan syndrome have short stature; some will achieve a normal adult height without treatment.  Therefore, the FDA-approved labeling recommends that, prior to initiating GH for a patient with Noonan syndrome, establish that the patient does have short stature.  The FDA-approved labeling for Norditropin recommends a dosage of GH of up to 0.066 mg/kg/day for pediatric patients with short stature associated with Noonan syndrome.

An international consensus statement on the diagnosis and management of Silver-Russell syndrome (SRS) (Wakeling, et al., 2017) recommends the use of growth hormone in children with SRS. The consensus statement notes that GH treatment does not have a specific indication for SRS and is prescribed under the SGA indication (height SDS −2.5; age >2–4 years; dose 35–70 µg/kg per day). Exemptions from the current SGA licensed indication used in some centers include starting GH therapy below the age of 2 years in case of: severe fasting hypoglycemia; severe malnutrition, despite nutritional support, which will lead to gastrostomy if no improvement is seen; and severe muscular hypotonia. The consensus statement recommends starting GH at a dose of approximately 35 µg/kg per day. The consensus statement recommends using the lowest dose that results in catch-up growth. The consensus statement recommends termination of GH therapy when height velocity is less than 2 cm per year over a 6-month period and bone age is greater than 14 years (female patients) or greater than 17 years (male patients).

Turner Syndrome

Turner syndrome is a chromosomal condition that describes girls and women with common features that are caused by complete or partial absence of the second sex chromosome. The most common feature of Turner syndrome is short stature.

Turner syndrome (TS), which occurs in 1 in every 2,000 live born girls, is due to abnormalities or absence of an X chromosome and is frequently associated with short stature, which may be ameliorated with GH treatment.  Because growth in height is variable in patients with TS, literature suggest that the decision whether to treat with GH and the timing of such treatment should be made on the basis of each patient's height and growth velocity.  Treatment is often initiated when the standard deviation score for height decreases to less than 2 standard deviations below the mean.

Growth failure associated with TS is thought to be multi-factorial, with one of the factors being reduced sensitivity to GH, rather than decreased GH levels.  Therefore, supra-physiological doses of GH are required for treatment in children with TS (NICE, 2002).  According to the available literature, treatment is often initiated with GH doses higher than those used in treating GHD; the usual dose of GH for TS is 0.045 to 0.050 mg/kg/day.  Several studies suggest that statural growth may be optimized by concomitant treatment with oxandrolone in a daily dose of 0.0625 mg/kg.

Other Indications

18p Deletion Syndrome

Schober et al (1995) reported on a patient with 18p monosomy and GH deficiency due to pituitary hypoplasia, who showed an excellent response to GH-treatment.  The authors concluded that patients with this syndrome should be considered for endocrine evaluation, as they could benefit from hormonal substitution.

Turleau (2008) stated that in patients with monosomy 18p who had short stature, GH deficiency is frequently found and may justify GH treatment.  However, no specific treatment exists for deletion 18p syndrome; however, early rehabilitative and educational interventions are recommended, mainly speech therapy, since the majority of patients have major speech problems and difficulties with speech articulation.  Physical therapy for hypotonia should be advised.

Goyal et al (2017) noted that 18p deletion syndrome is characterized by the deletion of short arm of chromosome 18.  Presentation of this syndrome is quite variable with dysmorphic features, growth deficiencies, and mental retardation with poor verbal performance.  Few patients even fail to thrive when malformations involving the heart and brain are severe.  These investigators described an isolated case of 18p deletion in a 23-year-old woman who for the first time reported to the hospital for dental problems.  The patient was short statured with mental retardation and craniofacial, skeletal, dental, and endocrinal abnormalities.  Such presentation warrants prompt diagnosis for effective management.  The treatment was directed toward the specific symptoms that were apparent in each individual.  Such treatment may require the coordinated efforts of pediatricians, surgeons, physicians, orthopedic, neurologists, speech-language pathologists, and/or other health-care professionals.  The specific surgical procedures performed would depend on the severity and location of the anatomical abnormalities, their associated symptoms, and other factors.  Endocrinopathies should be taken care, and the patient should be examined for immunological deficiencies at timely intervals.  The prognosis is poor for those patients with severe brain malformations, otherwise survival up to the 6th decade has been reported.  Genetic counseling would also be of benefit for affected individuals and their families; other treatments for this disorder are symptomatic and supportive. 

Sun et al (2018) stated that 18p deletion syndrome is a rare chromosomal disease caused by deletion of the short arm of chromosome 18.  By using cytogenetic and SNP array analysis, these researchers identified a girl with 18p deletion syndrome exhibiting craniofacial anomalies, intellectual disability, and short stature.  G-banding analysis of metaphase cells revealed an abnormal karyotype 46,XX,del(18)(p10).  Furthermore, SNP array detected a 15.3-Mb deletion at 18p11.21p11.32 (chr18:12842-15375878) including 61 OMIM genes.  Genotype-phenotype correlation analysis showed that clinical manifestations of the patient were correlated with LAMA1, TWSG1, and GNAL deletions.  Her neuropsychological assessment test demonstrated delay in most cognitive functions including impaired mathematics, linguistic skills, visual motor perception, respond speed, and executive function.  In addition, her integrated visual and auditory continuous performance test (IVA-CPT) indicated a severe comprehensive attention deficit.  At age 7 and 1/12 years, her height was 110.8 cm (-2.5 SD height for age); GH treatment was initiated.  After 27 months treatment, her height was increased to 129.6 cm (-1.0 SD height for age) at 9 and 4/12 years, indicating an effective response to GH treatment.

Yang et al (2019) noted that deletion on the short arm of chromosome 18 is a rare disorder characterized by intellectual disability, growth retardation, and craniofacial malformations (such as prominent ears, microcephaly, ptosis, and a round face).  The phenotypic spectrum is wide, encompassing a range of abnormalities from minor congenital malformations to holoprosencephaly.  These investigators presented the case of a 2-year-old girl with ptosis, a round face, broad neck with low posterior hairline, short stature, and panhypopituitarism.  She Schober et al (1995) reported on a patient with 18p monosomy and GH deficiency due to pituitary hypoplasia, who showed an excellent response to GH-treatment.  The authors concluded that patients with this syndrome should be considered for endocrine evaluation, as they could benefit from hormonal substitution.

Acute Catabolism

Growth hormone is not recommended for treatment of acute catabolism, including pre-operative and post-operative treatment, critically ill patients, and burn patients.  The results of 2 clinical trials of GH therapy for critically ill patients showed a significantly higher mortality in GH-treated patients.

Amyotrophic Lateral Sclerosis (ALS)

While it has been documented that GH secretion is impaired in patients with ALS (Morselli et al, 2006), there is a lack of evidence to support the use of GH in these patients.  Based on the known trophic effects of GH on nerve and muscle, Smith et al (1993) treated 75 patients with ALS for up to 18 months with synthetic human GH (hGH) or a placebo.  The course of ALS was assessed serially using a quantitative (TQNE) neuromuscular and manual examination (MRC) and laboratory chemistries.  Average insulin-related growth factor values increased from 1.2 to 2.3 U/ml in the treated group.  Surprisingly, serum insulin levels did not increase.  Hyperglycemia was noted in only 2 patients of the 38 patients receiving hGH, and this resolved with cessation of treatment.  Over the 12-month treatment there were 11 deaths (6 controls, 5 treated).  Survival analysis, performed approximately 12 months following cessation of treatment, did not reveal a difference between the treatment and placebo group.  The TQNE scores declined inexorably in both the control and treated group.  Retrospective analysis of the TQNE data indicated a poor prognosis for patients who lost arm strength early.  A correlation between the TQNE and MRC scores was evident at early stages of motor unit loss, less so when muscle weakness was advanced.

Anti-Aging

Liu et al (2007) stated that human GH is widely used as an anti-aging therapy, although its use for this purpose has not been approved by the FDA and its distribution as an anti-aging agent is illegal in the United States.  The authors evaluated the safety and effectiveness of GH therapy in the healthy elderly.  They found that the literature published on randomized, controlled trials evaluating GH therapy in the healthy elderly is limited but suggests that it is associated with small changes in body composition and increased rates of adverse events.  The authors concluded that, based upon this evidence, GH can not be recommended as an anti-aging therapy.  Furthermore, in an editorial on the use of GH secretagogues to prevent and treat the effects of aging, Blackman (2008) stated that many questions regarding the potential utility and safety of an oral GH secretagogue in older individuals remain unanswered.  The clinical use of GH axis manipulation in the elderly should be restricted to carefully controlled clinical trials.

CHARGE (Coloboma, Heart defect, Atresia choanae, Retarded growth and development, Genital hypoplasia, Ear anomalies/deafness) Syndrome

Khadilkar et al (1999) stated that growth failure and anterior pituitary dysfunction are clinical features of the CHARGE and VATER associations.  These researchers investigated pituitary dysfunction as a potential cause of poor growth in a series of 4 and 3 patients with the CHARGE and VATER associations, respectively, who had height SDS less than-2.  Five of the 7 patients had associated subnormal growth velocity SDS.  Patients were investigated with a combination of dynamic and basal endocrine tests.  All patients were found to be normo-natremic and to have normal basal thyrotroph and stimulated corticotroph function.  The 1 peri-pubertal patient had evidence of biochemical gonadotroph dysfunction.  Although 2 patients had marginally low stimulated serum GH responses to glucagon stimulation testing, this was associated with either normal growth velocity or normal serum IGFBP-3 concentrations.  Thus, somatotroph dysfunction could not be demonstrated unequivocally in any patient.  The authors concluded that poor childhood linear growth in the CHARGE and VATER associations does not appear to be associated with pituitary dysfunction.

Chiari Malformations

Ballard and colleagues (2020) noted that the safety and efficacy of GH replacement therapy (GHRT) on pediatric patients with GHD and Chiari malformation type 1 (CM-I) are not well-investigated within the current body of literature.  With no clear indication of the effects of GHRT on CM-I disease progression, these researchers examined the effect of GHRT on tonsillar herniation and progression of CM-I symptomatology.  From a previously established database of 465 patients with radiologically confirmed CM-I defined as greater than 5 mm of tonsillar descent on head MRI, these investigators identified 20 patients who also had GHD.  Using the imaging analysis software package, ANALYZE, the degree of change in tonsillar herniation was documented between initial and final MRI measurements.  The radiologic and clinical changes over time were examined via a proportional odds model, Student's t test, Mann-Whitney test, or a mixed model corresponding to the outcomes measured either on an ordinal scale or on a quantitative scale.  Incidence of GHD in this CM-I population was 4.3 %.  There was no significant effect of GHRT on the degree of tonsillar herniation in patients with GHD and CM-I.  No patient became symptomatic, developed syringomyelia, or required surgical intervention for CM-I.  The authors concluded that based on their findings with a larger sample size, along with recent reports, the incidence of patients with CM-I and GHD reported (0.86 to 5 %) is likely more indicative of the actual incidence of GHD and CM-I than the prior findings within the literature (9.1 to 20 %).  These researchers also suggested that GHRT did not significantly affect CIM morphology or symptomatology.  Thus, neurosurgeons should have no hesitation clearing these patients for GHRT. 

Furthermore, an UpToDate review on “Chiari malformations” (Khoury, 2021) does not mention GH as a therapeutic option.

Chondral Defect Repair

Danna and co-workers (2018) stated that focal chondral defects alter joint mechanics and cause pain and debilitation.  Microfracture is a surgical technique used to treat such defects.  This technique involves penetration of subchondral bone to release progenitor cells and growth factors from the marrow to promote cartilage regeneration.  Often this results in fibrocartilage formation rather than structured hyaline cartilage.  Some reports have suggested use of GH with microfracture to augment cartilage regeneration.  These researchers examined if intra-articular (IA) GH in conjunction with microfracture, improves cartilage repair in a rabbit chondral defect model.  They hypothesized that GH would exhibit a dose-dependent improvement in regeneration.  A total of 16 New Zealand white rabbits received bilateral femoral chondral defects and standardized microfracture repair.  One group of animals (n = 8) received low-dose GH by IA injection in the left knee, and the other group (n = 8) received high-dose GH in the same manner.  All animals received IA injection of saline in the contralateral knee as control.  Serum assays, macroscopic grading, and histological analyses were used to assess any improvements in cartilage repair.  Peripheral serum GH was not elevated post-operatively (p = 0.21).  There was no improvement in macroscopic grading scores among either of the GH dosages (p = 0.83).  Scoring of safranin-O-stained sections showed no improvement in cartilage regeneration and some evidence of increased bone formation in the GH-treated knees.  The authors concluded that treatment with either low- or high-dose IA GH did not appear to enhance short-term repair in a rabbit chondral defect model.

Chronic Fatigue Syndrome

A systematic evidence review of interventions for chronic fatigue syndrome prepared by the UK National Health Service Centre for Reviews and Dissemination (2002) identified one small clinical trial of GH for chronic fatigue syndrome (Moorkens et al, 1998), and found that “no conclusions regarding the effect of [growth hormone] treatment can be drawn from this trial”.  An assessment prepared for the Agency for Healthcare Quality and Research also concluded that there is insufficient evidence of the effectiveness of GH as a treatment for chronic fatigue syndrome (Mulrow et al, 2001).

Chronic Pain Syndromes and Fibromyalgia

An UpToDate review on “Treatment of fibromyalgia in adults not responsive to initial therapies” (Goldenberg, 2013) lists GH as one of the investigational approaches for the treatment of fibromyalgia. It notes that “Growth hormone, both as monotherapy and as adjunctive therapy, improves symptoms of fibromyalgia, although cost concerns and the need for long-term efficacy and safety data are considerations limiting its use”.

Xu and colleagues (2020) noted that GH and GH-related signaling molecules play an important role in nociception and development of chronic pain.  In a systematic review, these investigators examined the potential molecular mechanisms through which GH-related signaling modulates sensory hypersensitivity in rodents, the clinical pharmacology of GH, and the clinical evidence of GH treatment for several common pain syndromes.  They carried out a search using the PubMed/Medline database, Scopus, and the Cochrane library for all reports published in English on GH in pain management from inception through May 2018.  A critical review was performed on the mechanisms of GH-related signaling and the pharmacology of GH.  The levels of clinical evidence and implications for recommendations of all of the included studies were graded.  The search yielded 379 articles, of which 201 articles were deemed irrelevant by reading the titles; 53 reports were deemed relevant after reading abstracts.  All of these 53 articles were retrieved for the analysis and discussion.  The authors concluded that dysfunction of the GH/IGF-1/ghrelin axis was linked to hyperalgesia and several common clinical pain syndromes.  Low levels of GH and IGF-1 were linked to pain hypersensitivity, whereas ghrelin appeared to provide analgesic effects.  Pre-treatment of GH reversed mechanical and thermal hypersensitivity in an animal model of inflammatory pain.  Clinical trials support GH treatment in a subgroup of patients with fibromyalgia syndrome (level of evidence: 1B+) or chronic lower back pain (LBP) syndrome (level of evidence: 2C+).  These researchers stated that the knowledge of GH-related molecules in nociception is evolving, and more research is needed to guide clinical applications in pain medicine.

Congestive Heart Failure

Le Corvoisier et al (2007) systematically reviewed and analyzed all RCTs and open studies of sustained GH treatment in patients with congestive heart failure (CHF).  A total of 12 trials were identified in 3 databases.  These researchers conducted a combined analysis of GH effects on cardiovascular parameters using the overall effect size to evaluate significance and computing the weighted mean differences with and without treatment to assess effect size.  Growth hormone treatment significantly modified morphological cardiovascular parameters [inter-ventricular septum thickness, +0.55 (S.D., 0.43) mm (p < 0.001); posterior wall thickness, +1.01 (0.44) mm (p < 0.01); left ventricle (LV) end-diastolic diameter, -2.02 (1.22) mm (p < 0.01); and LV end-systolic diameter, -5.30 (2.33) mm (p < 0.05)]; LV and systemic hemodynamics [LV end-systolic wall stress, -38.9 (13.3) dynes/cm(2) (p < 0.001); LV ejection fraction (LVEF), +5.10 (1.74) % (p < 0.05); and systemic vascular resistance, +195.0 (204.5) dyn x sec(-1) x cm(-5) (p < 0.01)]; and functional parameters [New York Heart Association (NYHA) class, -0.97 (0.23) (p < 0.01); exercise duration, +103.7 (37.6) sec (p < 0.001); and maximal oxygen uptake, +2.48 (1.76) ml/kg x min (p < 0.01)].  Subgroup analysis and meta-regression showed significant relationships between the IGF-I response and GH treatment effects.  The authors concluded that the findings of this meta-analysis suggested that GH treatment improves several relevant cardiovascular parameters in patients with CHF.  However, these results must be confirmed by a large randomized placebo-controlled trial on hemodynamic, morphological, and functional parameters during long-term high-dose GH treatment of patients with CHF.

In a meta-analysis, Tritos and Danias (2008) examined the safety effectiveness of recombinant human growth hormone (rhGH) therapy in congestive heart failure (CHF).  These investigators searched 3 literature databases (MEDLINE, EMBASE, and the Cochrane Register) for clinical studies of rhGH therapy in CHF due to systolic dysfunction.  Therapy with rhGH appears to have beneficial clinical effects (weighted mean difference [95 % CI] in CHF including improved exercise duration (1.9 mins [1.1 to 2.7]), maximum oxygen consumption (2.1 ml x kg(-1) x min(-1) [1.2 to 3.0]), and New York Heart Association class (-0.9 [-1.5 to -0.3]).  There were salutary hemodynamic effects of rhGH therapy, including increased cardiac output (0.4 L x min(-1) [0.1 to 0.6]) and decreased systemic vascular resistance (-177 dyn x s x cm(-5) [-279 to -74]).  Among rhGH-treated patients, left ventricular (LV) ejection fraction improved (4.3 % [2.2 to 6.4]).  Despite increases in LV mass and wall thickness, there were no adverse effects on diastolic function.  Subgroup analyses suggest that study design and treatment duration may influence some of the treatment effects.  Most of the beneficial effects were driven by either uncontrolled or longer duration studies.  Administration of rhGH therapy slightly increased the risk for ventricular arrhythmia; however, this finding was driven by a single small study.  The authors concluded that rhGH therapy may have beneficial cardiovascular effects in CHF caused by LV systolic dysfunction.  The possibility of pro-arrhythmia associated with rhGH therapy requires further study.  They stated that larger randomized trials with longer treatment duration are needed to fully elucidate the safety and effectiveness of rhGH therapy in this patient population.

Constitutional Delay in Growth and Development

Constitutional delay of growth is characterized by normal prenatal growth followed by growth deceleration during infancy and childhood, which is reflected by declining height percentiles at this time.  Children with constitutional delay have later timing of puberty than do their peers, allowing a longer period during which they are able to grow.  Most commonly, these patients achieve normal adult height if no treatment is given.  Although constitutional delay may be treated with GH, other effective and less costly treatments are available.  In male patients, the literature shows testosterone or anabolic steroids are effective, and in female patients, low dose estrogens may be used.

Decompensated Cirrhosis

In a randomized trial, Verma and colleagues (2018) examined the impact of multiple courses of granulocyte-colony stimulating factor (G-CSF) with or without GH in patients with decompensated cirrhosis.  A total of 65 patients with decompensated cirrhosis were randomized to standard medical therapy (SMT) plus G-CSF 3 monthly plus GH daily (group A; n = 23), or SMT plus G-CSF (group B; n = 21) or SMT alone (group C; n = 21).  The primary outcome was the transplant free survival (TFS) at 12 months.  The secondary outcomes were mobilization of CD34+ cells at day 6; the improvement in clinical scores, liver stiffness, nutrition, episodes of infection and quality of life (QoL) at 12 months.  There was significantly better 12-month TFS in groups A and B than in group C (p = 0.001).  At day 6 of therapy, CD34+ cells increased in groups A and B compared to baseline (p < 0.001).  There was a significant decrease in clinical scores, improvement in nutrition, better control of ascites, reduction in liver stiffness, lesser infection episodes and improvement in QoL scores in groups A and B, at 12 months as compared to baseline (p < 0.05).  The therapies were well-tolerated.  The authors concluded that multiple courses of G-CSF improved 12-month TFS, mobilized hematopoietic stem cells, improved disease severity scores, nutrition, fibrosis, QoL scores, ascites control, reduced infections, and the need for liver transplantation in patients with decompensated cirrhosis.  However, the use of GH was not found to have any additional benefit.

Down Syndrome and Other Syndromes associated with Short Stature and Malignant Diathesis

Because short stature is characteristic of many syndromes, GH therapy has been attempted in several conditions, including Down syndrome, Fanconi syndrome, and Bloom syndrome.  The high basal risk of malignant tumor or leukemia in these syndromes, however, has led many pediatric endocrinologists to recommend against the use of GH because the potential for GH to increase the risk of malignancy.

Glucocorticoid-Induced Growth Failure

Allen et al (1998) stated that growth failure is common during long term treatment with glucocorticoids (GC) due to blunting of GH release, IGF-I bioactivity, and collagen synthesis.  These effects could theoretically be reversed with GH therapy.  The National Cooperative Growth Study database (n = 22,005) was searched for children meeting the following criteria:
  1. pharmacological treatment with GC and GH for more than 12 months,
  2. known type and dose of GC, and
  3. height measurements for more than 12 months. 

A total of 83 patients were identified.  Monitoring of glucose, insulin, IGF-I, IGF-binding protein-3, type 1 procollagen, osteocalcin, and glycosylated hemoglobin levels was performed in a subset of patients.  Stimulated endogenous GH levels were less than 10 microg/L in 51 % of patients, and less than 7 microg/L in 37 % of patients.  The mean GC dose, expressed as prednisone equivalents, was 0.5 +/- 0.6 mg/kg day.  Baseline evaluation revealed extreme short stature (mean height SD score = -3.7 +/- 1.2), delayed skeletal maturation (mean delay of 3.1 years), and slowed growth rates (mean of 3.0 +/- 2.5 cm/yr).  After 12 months of GH therapy (mean dose of 0.29 mg/kg x weeks), mean growth rate increased to 6.3 +/- 2.6 cm/yr, and height SD score improved by 0.21 +/- 0.4 (p < 0.01).  During the second year of GH therapy (n = 44), the mean growth rate was 6.3 +/- 2.0 cm/yr.  Prednisone equivalent dose and growth response to GH therapy were negatively correlated (r = -0.264; p < 0.05).  Plasma concentrations of IGF-I, IGF-binding protein-3, procollagen, osteocalcin, and glycosylated hemoglobin increased with GH therapy, whereas glucose and insulin levels did not change.  The authors concluded that the growth-suppressing effects of GC were counter-balanced by GH therapy; the mean response is a doubling of baseline growth rate.  However, responsiveness to GH is negatively correlated with GC dose.  Glycosylated hemoglobin levels increased slightly, but glucose and insulin levels were not altered by GH therapy.

Growth Hormone for Adolescent Transgender Transitioning to Male

Growth hormone is used for the treatment of prepubertal children with short stature associated with Noonan syndrome, and short stature homeobox-containing gene (SHOX) deficiency.  However, GH for short stature per se (idiopathic short stature) is not considered an illness/disease.  Furthermore, World Professional Association for Transgender Health (WPATH) transgender guidelines (Coleman et al, 2011) have no recommendation for GH.

HIV Lipodystrophy Syndrome

Aetna considers GH treatment for HIV patients with lipodystrophy syndrome to be experimental and investigational.  Even though preliminary observations suggest that recombinant human GH may lead to partial regression of fatty Buffalo humps and to a decrease in waist size secondary to truncal obesity, there is no definitive evidence of effectiveness of GH for this indication. 

Sivakumar et al (2011) noted that HIV-associated lipodystrophy is a disorder of fat metabolism that occurs in patients with HIV infection.  It can cause metabolic derangements and negative self-perceptions of body image, and result in non-compliance with highly active anti-retroviral therapy (HAART).  Growth hormone axis drugs have been evaluated for treatment of this disorder, but no systematic review has been conducted previously.  These investigators compared the effects of GH axis drugs versus placebo in changing visceral adipose tissue (VAT), subcutaneous adipose tissue (SAT) and lean body mass (LBM) in patients with HIV-associated lipodystrophy.  They searched MEDLINE (1996 to 2009), CENTRAL (Issue 4, 2009), Web of Science, Summons, Google Scholar, the FDA website, and Clinicaltrials.gov from October 13, 2009 to June 7, 2010.  These researchers excluded newspaper articles and book reviews from the Summons search; this was the only search limitation applied.  They also manually reviewed references of included articles.  Inclusion criteria were as follows: Randomized controlled trial (RCT); study participants with HIV-associated lipodystrophy; intervention consisting of GH, GHRH, tesamorelin or IGF-1; study including at least 1 primary outcome of interest: change in VAT, SAT or LBM.  Two independent reviewers extracted data and assessed study quality using a standardized form.  The authors of one study were contacted for missing information.  The main effect was calculated as a summary of the mean differences in VAT, SAT and LBM between the intervention and placebo groups in the included studies.  Subgroup analyses were performed to assess different GH axis drug classes.  A total of 10 RCTs including 1,511 patients were included in the review.  All had a low risk of bias and passed the test of heterogeneity for each primary outcome.  Compared with placebo, GH axis treatments decreased VAT [weighted mean difference (WMD) -25.20 cm(2); 95% CI: -32.18 to -18.22 cm(2); p < 0.001] and increased LBM (WMD 1.31 kg; 95 % CI: 1.00 to 1.61 kg; p < 0.001], but had no significant effect on SAT mass (WMD -3.94 cm(2); 95 % CI: -10.88 to 3.00 cm(2); p = 0.27].  Subgroup analyses showed that GH had the most significant effects on VAT and SAT, but none on LBM.  The drugs were well-tolerated but statistically significant side effects included arthralgias and edema.  The authors concluded that the findings of this review indicated that, based on the findings of the 10 included studies, GH axis treatments were effective in reducing VAT and increasing LBM in patients with HIV-associated lipodystrophy.  However, clinicians must decide whether the attributed benefits are clinically significant, considering the costs and potential risks of GH axis treatments.  A limitation of this study was the small number of studies available of each GH axis drug class.

Also, an UpToDate review on “Treatment of HIV-associated lipodystrophy” (Glesby, 2013) states that “Recombinant human growth hormone (rhGH) is known to be lipolytic; patients with AIDS-related wasting treated with supraphysiologic doses of rhGH (Serostim®) lost body fat while gaining lean body mass.  This observation provided the rationale for studying the efficacy of rhGH in the treatment of patients with fat accumulation initially in pilot studies and ultimately in randomized, placebo-controlled trials …. The major side effects of rhGH are fluid retention, arthralgias, myalgias, and, less commonly, carpal tunnel syndrome and diabetes mellitus.  Of note, studies of rhGH for increased truncal fat have generally excluded patients with impaired fasting glucose and impaired glucose tolerance on a standard, 75 gram 2-hour oral glucose tolerance test, since these patients may be at increased risk of developing rhGH-induced hyperglycemia and diabetes.  While maintenance therapy with rhGH after an induction phase was superior to placebo in the phase III trial, the optimal strategy for maintaining visceral fat reduction that may be achieved from rhGH induction is uncertain.  rhGH is not currently indicated for the treatment of HIV-associated truncal obesity and its clinical development for this indication appears to be on hold”.

Hypochondroplasia

Kanaka-Gantenbein (2001) noted that skeletal dysplasias are genetic disorders of bone and cartilage development, mainly characterized by disproportionate short stature.  Achondroplasia is the commonest and best described form of skeletal dysplasia, leading to a mean final height of 131 +/- 5.6 cm for males and 124 +/- 5.9 cm for females.  Growth hormone has been used in different studies in patients with achondroplasia in order to ameliorate their height, and short-term results ranged from rather positive to moderate.  However, disproportionate advancement of bone age has been observed that can compromise the positive effect of such treatment.  Furthermore, concern exists about the aggravation of body disproportion necessitating a later leg-lengthening procedure in order to achieve proportionate adult stature.  In hypochondroplasia, GH treatment seems to give better results when administered at puberty.  No data on final height yet exist, however, so that more studies with greater numbers of patients need to be performed before a consensus on GH use in achondroplasia and hypochondroplasia can be reached.  Other forms of skeletal dysplasias are quite rare, so that no conclusion on GH use in such patients can be drawn.  Finally, in osteogenesis imperfecta, GH administration significantly ameliorates bone density but does not clearly seem to affect final height positively.

Francomano (2005), Chief of the Medical Genetics Branch of the National Human Genome Research Institute/National Institutes of Health stated that “Trials of growth hormone (GH) therapy in hypochondroplasia have shown mixed results.  Several reports indicate that some individuals respond well with increased proportional height velocity, others respond with increased disproportionate growth, and some do not respond [Appan et al 1990, Mullis et al 1991, Bridges et al 1991].  These differences in individual responses may result from genetic heterogeneity and indicate a need for stratification of affected individuals with regard to genetic etiology (e.g., those with FGFR3 mutations and those without).  While a response to GH has been sustained in some individuals for as long as 6 years [Bridges & Brook 1994], data about final adult height in these individuals are not yet available and the ultimate success of this approach remains uncertain.  Meyer et al [2003] emphasized the importance of considering pubertal development in assessing the response to GH stimulation testing.  Tanaka et al [2003] reported data suggesting that children with hypochondroplasia may have a greater response to GH therapy than children with achondroplasia.  Kanazawa et al [2003] also reported a response to GH among children with hypochondroplasia.  Growth hormone therapy is still considered experimental and controversial”.

In a pilot study, Rothenbuhler et al (2012) evaluated the growth promoting effect of a recombinant growth hormone (rGH) treatment protocol adjusted on IGF-1 dosing in children affected by the most severe forms of FGFR3 N540K-mutated hypochondroplasia.  This study included 6 children (mean age, 2.6 +/- 0.7 years; mean height SDS, -3.0 +/- 0.5) with the N540K mutation of FGFR3 gene who received an rGH dosage titrated to an IGF-1 level close to 1.5 SDS of the normal range.  Recombinant GH therapy was interrupted 1 day per week, 1 month per year, and 6 months every 2 years.  The mean height SDS increased by 1.9 during the 6.1 +/- 0.9-year study period, reaching -0.8 to -1.3 at age 8.7 +/- 1 years.  The mean +/- SDS baseline IGF-1 value was -1.6 =/- 0.5 before rGH treatment and 1.4 +/- 0.3 during the last year of observation.  The average cumulative rGH dose was 0.075 +/- 0.018 mg/kg/day (range of 0.059 to 0.100 mg/kg/day).  Trunk/leg disproportion was improved.  The authors concluded that IGF-1-dosing rGH treatment durably improves growth and reduces body disproportion in children with severe forms of hypochondroplasia.  The findings of this small, non-randomized study need to be validated by well-designed studies with long-term follow-up.

Hypophosphatemia (e.g., hypophasphatemic rickets, X-linked hypophosphatemia in children)

Short-term acceleration of growth as a result of GH therapy has also been reported in children with spinal cord defects and hypophosphatemic rickets; some of these children had impaired GH production. However, no studies have prospectively assessed linear growth until achievement of final height. A discordance between stimulated and spontaneous GH secretion gave rise to the belief that GH neurosecretory dysfunction might exist in children, especially in those who had received low-dose cranial irradiation. Current guidelines do not recommend GH for children with these conditions. Growth hormone treatment has been proposed for children with "partial" GH insensitivity. However, there are no established criteria for diagnosis of partial GH insensitivity, and there are no studies of the effectiveness of GH for this condition.

Smith and Remmington (2021) noted that conventional treatment of X-linked hypophosphatemia with oral phosphate and calcitriol could heal rickets; however, it does not always raise serum phosphate concentrations significantly, nor does it always normalize linear growth.  Findings of some clinical trials suggested that combining recombinant human GH therapy with conventional treatment improved growth velocity, phosphate retention, and BMD; however, other clinical trials suggested that it appeared to aggravate the pre-existent disproportionate stature of such children.  This is an updated version of a previously published Cochrane review.  These researchers examined if recombinant human GH therapy for children with X-linked hypophosphatemia is associated with changes in longitudinal growth, mineral metabolism, endocrine function, renal function, BMD, body proportions, and also with any adverse effects.  They searched the Cochrane Cystic Fibrosis and Genetic Disorders Group Trials Register which comprises references identified from comprehensive electronic database searches and hand-searches of relevant journals and abstract books of conference proceedings.  Furthermore, they searched the Cochrane Central Register of Controlled Trials, Ovid Medline and the reference lists of identified trials and other reviews.  These investigators also undertook some additional hand-searching of relevant journals and conference proceedings.  Date of the most recent search: January 12, 2021.  All RCTs or quasi-RCTs comparing GH (alone or combined with conventional treatment) with either placebo or conventional treatment alone in children with X-linked hypophosphatemia were included for analysis.  Two authors independently evaluated studies for risk of bias and extracted data from eligible studies.  GRADE criteria were used to examine the certainty of the evidence for each outcome.  These researchers included 2 studies (20 participants) in the review.  In 1 cross-over study, results showed that recombinant human GH therapy may improve the height standard deviation (SDS) score (z score), but they were unsure whether the intervention was the reason behind a transient increase in serum phosphate and tubular maximum for phosphate reabsorption.  In the 2nd, parallel study, treatment may also have improved the height SDS from baseline in the rhGH group compared to the control group, although no significant difference was observed between groups after 3 years, MD 0.50 SDS (95 % CI: -0.54 to 1.54) (low-certainty evidence).  The treatment was possibly well-tolerated during both studies with only transient adverse effects observed in 3 participants (low-certainty evidence).  These investigators were uncertain whether GH improved serum phosphate levels or change in TmP/GFR (very low-certainty evidence).  The treatment may make little or no difference to alkaline phosphatase levels (low-certainty evidence).  The authors concluded that there is insufficient high-certainty evidence to recommend the use of recombinant human GH therapy in children with X-linked hypophosphatemia.

Idiopathic Short Stature

Aetna benefit plans cover treatment of disease or injury; Aetna does not consider idiopathic short stature a disease.  A heterogeneous group of otherwise apparently normal children who are 2 or more standard deviations below the mean for height, but who have normal serum GH responses to stimuli are classified as having genetic short stature, normal-variant familial short stature if their parents are short, constitutional delay of growth if there is a delay in skeletal maturation, idiopathic short stature, or neurosecretory GH dysfunction.  Treatment of these children with GH is controversial with regard to both efficacy and ethics.  Although GH therapy initially causes growth acceleration, it also accelerates pubertal development and advances bone age so that the duration of growth during puberty is shortened.

One RCT reported near final height (NFH) in of girls with idiopathic short stature.  Two published studies reporting final height were prospective non-RCTs, one in peripubertal boys with subnormal integrated GH concentration and one in short, normal children.  Results from the RCT including NFH found that treated girls were approximately 7.5 cm taller than randomized control girls and 6 cm taller than girls who refused consent.  Other long-term studies also suggest that final height is increased by GH treatment.  However, the increase is between 2 cm to 7 cm, and treated individuals remain relatively short when compared with peers of normal stature.

Short stature does not result in disease or functional limitation.  Therefore, the use of GH for this condition considered an enhancement of human performance or appearance rather than as a medically necessary treatment of disease.  All normal and healthy populations have genetic variation that will give rise to individuals with short stature.  In a position statement, the American Academy of Pediatrics (1997) has noted that, by definition, children with short stature relative to their peers will always exist and targeting the current cohort for medical intervention will merely replace them with another cohort.

Studies have demonstrated that the use of GH in children with idiopathic short stature (ISS) increases growth rate and height and may minimally increase final height, as compared with baseline predicted values, but generally does not increase final height to normal levels.  Some argue, however, that the major criterion for the use of GH in ISS should be improvement in the individual patient's QoL, regardless of whether final height is improved or not.  But, whether short stature itself (with no pathological basis) correlates with psychosocial dysfunction of any kind is debated.  An assessment conducted by NICE stated that “Most studies concur that shortness alone does not necessarily result in negative psychological consequences.  Many studies have found no relation between degree of shortness and psychological problems”.

In addition, there is no adequate evidence from randomized prospective clinical studies demonstrating clinically significant improvements in functional status or reductions in psychological dysfunction in children with idiopathic short stature who are treated with GH.  In addition, some experts maintain, however, that psychological dysfunction may be better addressed by psychological intervention and counseling than by the use of GH.

In a position statement on the use of GH in children, the American Academy of Pediatrics (1997) has stated: “In many other instances, the use of GH has been justified on the grounds that persons with short stature (defined as more than 2 SDs below the mean for age and sex) experience stigma in an affluent society.  These children are often teased in school about their short stature; moreover, empiric evidence indicates that numerous social benefits are linked to tall stature.  In some children, short stature may be part of an acquired or inherited disorder.  For these children, growth augmentation is viewed as an avenue to normalcy.  Despite these concerns and the fairly extensive use of recombinant human GH in these patient groups, no objective current data demonstrate the psychosocial benefits of hormonal therapy in this group of children and few physiologic data demonstrate an effect on final adult height.  The above considerations have led some to question whether research on the use of human GH to attempt to increase the final adult height of non-GH-deficient children is warranted”.

“It is also unclear whether GH therapy reduces the psychosocial problems that very short children may experience.  Indeed, there is evidence that GH therapy exacerbates these problems in some children owing to unrealistic expectations concerning the therapeutic outcome and enhanced feelings that something is 'wrong' with them”.

“There is also the question of how to define treatment 'success'.  Short stature is a characteristic that must be defined relative to the general population in which people will always be of different heights.  Thus, even if GH therapy were available to and effective in all 'short' stature children, a population of short children will still exist; they will simply be a few inches taller than those in the former population”.

Root (2002) stated that “[m]any studies document the psychological good health and normal educational progress of healthy children with idiopathic short stature.  In fact, short children with behavioral problems and learning disabilities are referred more frequently for endocrine evaluation than are their normally achieving age and height peers.  Furthermore, it is quite possible that the extensive testing, daily injections, and frequent medical visits needed during [growth hormone] administration may imprint upon the child (and reinforce to the parent) a negative concept of his/her self worth”.

There is no adequate evidence that short stature, in and of itself, is associated with functional limitations.  A systematic evidence review prepared for the Agency for Healthcare Research and Quality evaluated the relationship of short stature in childhood with functional limitations, including intelligence, academic achievement, behavior, visual-motor perception, and psychomotor development (Wheeler et al, 2003).  The assessment concluded that there was no evidence that short stature in children is associated with severe functional limitations.

Poidvin et al (2014) investigated the incidence of stroke and stroke subtypes in a population-based cohort of patients in France treated with GH for short stature in childhood.  Adult morbidity data were obtained in 2008 to 2010 for 6,874 children with idiopathic isolated GH deficiency or short stature who started GH treatment between 1985 and 1996.  Cerebrovascular events were validated using medical reports and imaging data and classified according to standard definitions of subarachnoid hemorrhage, intra-cerebral hemorrhage, and ischemic stroke.  Case ascertainment completeness was estimated with capture-recapture methods.  The incidence of stroke and of stroke subtypes was calculated and compared with population values extracted from registries in Dijon and Oxford, between 2000 and 2012.  Using both Dijon and Oxford population-based registries as references, there was a significantly higher risk of stroke among patients treated with GH in childhood.  The excess risk of stroke was mainly attributable to a very substantially and significantly higher risk of hemorrhagic stroke (standardized incidence ratio from 3.5 to 7.0 according to the registry rates considered, and accounting or not accounting for missed cases), and particularly subarachnoid hemorrhage (standardized incidence ratio from 5.7 to 9.3).  The authors concluded that they reported a strong relationship between hemorrhagic stroke and GH treatment in childhood for isolated GH deficiency or childhood short stature.  They stated that patients treated with GH worldwide should be advised about this association and further studies should evaluate the potentially causal role of GH treatment in these findings.

As of February 2018, Norditropin (somatropin) injection is FDA-approved for the treatment of pediatric patients with idiopathic short stature (ISS), height standard deviation score (SDS) less than -2.25, and associated with growth rates unlikely to permit attainment of adult height in the normal range (Novo Nordisk, 2018).

Implant Osseointegration

In a systematic review and meta-analysis, Abduljabbar and colleagues (2017) examined if GH replacement therapy can enhance implant osseointegration.  These researchers performed a systematic literature search from 1982 to March 2016.  A structured search using the keywords "growth hormone", "implants", and "osseointegration" was performed to identify pre-clinical and clinical in-vivo controlled studies and was followed by a 2-phase search strategy.  Initially, 31 potentially relevant articles were identified.  After removal of duplicates and screening by title and abstract, 10 potential studies were included.  Studies were assessed for bias and data were synthesized using a random-effects meta-analysis model.  All studies were pre-clinical animal trials, and the follow-up period ranged from 2 to 16 weeks; 70 % of the included studies reported an increase in bone-to-implant contact in animals receiving GH compared with controls.  Meta-analysis showed a significant mean difference for bone to implant between GH groups versus controls (no GH supplementation) of 10.60 % (95 % CI: 3.79 % to 17.41 %) favoring GH administration.  The authors concluded that GH treatment appeared to promote osseointegration around implants in pre-clinical studies; however, these findings must be evaluated in highly controlled human clinical trials as a number of confounding factors may have influenced the outcomes of the included studies.

Improvement in Healing after Rotator Cuff Repair

In a multi-center, prospective, randomized, comparative trial, Oh and associates (2018) examined the effect of systemic injection of recombinant human GH (rhGH) on outcomes after arthroscopic rotator cuff repair.  Patients who underwent arthroscopic repair of large-sized rotator cuff tears were divided into 3 groups:
  1. rhGH 4 mg group (n = 26),
  2. rhGH 8 mg group (n = 24), and
  3. control group (n = 26).  

Sustained release rhGH was injected subcutaneously once-weekly for 3 months post-operatively.  The healing failure rate (primary end-point), fatty infiltration, and atrophy of the supraspinatus muscle, and functional scores (Constant and American Shoulder and Elbow Surgeons scores) were evaluated at 6 months.  Range of motion (ROM), pain visual analog scale (VAS), and serum IGF-1 level were measured at each follow-up.  The healing failure rate was similar between groups (rhGH 4-mg group, 30.8 %; rhGH 8-mg group, 16.7 %; and control group, 34.6 %; all p > 0.05).  The proportion of severe fatty infiltration (Goutallier grade greater than or equal to 3) was 20.8 % in the rhGH 8-mg group, 23.1 % in the rhGH 4-mg group, and 34.6 % in the control group (p > 0.05).  Functional outcomes, ROM, and pain VAS were similar between groups (all p > 0.05).  The rhGH 8-mg group showed more increased peak IGF-1 level (279.43 ng/ml) than the rhGH 4-mg group ((196.82 ng/ml) and control group (186.31 ng/ml), which was not statistically different (all p > 0.05).  No rhGH injection-related major safety issues occurred.  The authors concluded that this preliminary study showed no statistically significant improvement in healing or outcomes related to the treatment of rhGH after rotator cuff repair. However, they stated that further study with more enrolled patients after re-setting the rhGH dose or daily administration protocol would be mandatory.

Improvement of Endometrial Receptivity During In-Vitro Fertilization

Xu and colleagues (2019) noted that GH plays a critical role in cell growth, development, and metabolism throughout the body.  It can not only directly influence human oocytes and cumulus cells but also indirectly improve oocyte quality through activating synthesis of insulin-like growth factor-I (ILGF-I) or promoting follicle-stimulating hormone (FSH)-induced ovarian steroidogenesis.  Since GH can regulate female and male infertility, it has been applied in the management of infertility for many years, especially in patients with poor ovarian response or poor prognosis.  During ovarian stimulation, GH administration might improve the success rate of in-vitro fertilization (IVF) probably through the beneficial effects of GH on oocyte quality as indicated by a higher number of mature oocytes and embryos arriving at the transfer stage and a higher fertility rate in GH-treated patients.  However, there is still great controversy in the application of GH in IVF.  While some researchers showed that pregnancy, implantation and live-birth rates could be increased by ovarian pre-treatment with GH, others did not support GH as an effective adjuvant for infertility treatment because the live-birth rate was not increased.  Th authors reviewed recent advancements and potential effect of GH therapy in IVF.  The authors concluded that although GH is frequently used as an adjuvant in patients with poor ovarian response for ovulation promotion and in patients with repeated implantation failure for improving the endometrial receptivity, no clear standards have currently been set up for the indications, methods and dosages in clinical application; thus, more in-depth studies are needed to address these issues. 

Altmae and Aghajanova (2019) stated that administration of GH during ovarian stimulation has shown beneficial effects on IVF outcomes.  It is generally believed that this improvement is due to the stimulating effect of GH on oocyte quality.  However, studies are emerging that showed possible positive effect of GH administration on endometrial receptivity, thus suggesting an additional potential benefit at the level of the uterus, especially among women with recurrent implantation failure, thin endometrium, and older normal responders.  These investigators reviewed recent data on GH co-treatment effects on endometrium and endometrial receptivity among infertile women undergoing IVF, and proposed possible mechanisms of GH actions in the endometrium.  These researchers stated that whether GH administration during IVF is useful and which patient groups could benefit from it needs further investigation, but the preliminary data suggested that women suffering recurrent implantation failure, patients with thin endometrium and older normo-responders could benefit from GH treatment when undergoing ART.  The authors stated that carefully designed and sufficiently powered cohort studies, RCTs, are needed in the field in order to establish the most suitable therapeutic regimen for these patients and to clarify the confusion arisen from various studies that have shown either inconsistent or conflicting findings, used small patient cohorts and/or have been poorly designed with no blinding or placebo controls.

Shang et al (2022) stated that emerging evidence emphasizes GH-induced improvements in the endometrium; however, the findings are controversial.  In a systematic review and meta-analysis, these investigators examined if GH administration would improve endometrial function and reproductive outcomes during IVF cycles; and thus, guide clinical practice.  They carried out a literature search in the Cochrane Central Register of Controlled Trials, PubMed and Embase through November 30, 2021, without language restrictions; RCTs examining the effects of GH on IVF outcomes were included.  Risk of bias and quality of evidence (QoE) were assessed according to the Cochrane Collaboration's tool and the GRADE system; ORs and MDs with 95 % CIs were assessed by random-effects models.  A total of 25 studies with 2,424 women were included; 17 RCTs with poor responders (n = 1,723) showed that GH administration significantly increased endometrial thickness (EMT) (MD = 0.38, 95 % CI: 0.18 to 0.59; moderate QoE), which contributed to an improved LBR (OR = 1.67, 95 % CI: 1.13 to 2.49; very low QoE) and clinical pregnancy rate (CPR) (OR = 1.97, 95 % CI: 1.43 to 2.72; low QoE).  Subgroup analyses showed a dose- and time-dependent relationship between GH co-treatment and IVF outcomes; the optimal recommendation for improving CPR was consistent with that for EMT, rather than for oocytes and embryos.  Hence, GH might improve fertility via effects on the endometrium.  Administration of GH daily from the follicular phase of previous cycle until the hCG trigger with less than 5 IU/day led to a thicker endometrium and a greater chance of becoming pregnant, while 5 to 10 IU/day or administration from the luteal phase of the previous cycle until the hCG trigger resulted in higher oocyte and embryo quality.  Poor responders might benefit from co-treatment with the GnRH agonist long protocol more than other stimulation protocols.  Pooled data from 4 studies (n = 354) on women with a thin endometrium indicated that improved endometrial function might be critical for improving reproductive outcomes during GH treatment, as no improvements in embryo quality were found.  GH administration not only increased EMT (MD = 1.48, 95 % CI: 1.21 to 1.75; moderate QoE) but also promoted endometrial morphology (OR = 2.67, 95 % CI: 1.36 to 5.23; low QoE) and perfusion (OR = 5.84, 95 % CI: 1.30 to 26.17; low QoE); thus, improving the CPR (OR = 2.71, 95 % CI: 1.69 to 4.34; p < 0.0001; low QoE).  There was insufficient evidence to reach a conclusion regarding the effects of GH in normal responders (n = 80).  Due to obvious improvements in the CPR, women with a thin endometrium might be the most appropriate population to benefit from GH administration.  The authors concluded that improving endometrial function might be another vital mechanism by which GH improves IVF outcomes.  Optimal treatment should be offered to the target population according to their personal conditions and needs.  The QoE was moderate to very low, due to limited sample sizes and methodological problems; therefore, the findings of this meta-analysis should be interpreted with caution.  These researchers stated that more rigorous RCTs with large sample sizes are needed to confirm the effects and determine optimal GH protocols.

Improvement of In-Vitro Fertilization (IVF) Outcomes of Poor Ovarian Responders

In a systematic review and meta-analysis, Cozzolino and colleagues (2020) examined the effectiveness of GH supplementation in improving in-vitro fertilization (IVF) outcomes of poor ovarian responders (PORs).  Subjects were PORs undergoing conventional IVF or intracytoplasmic sperm injection (ICSI); RCTs of PORs undergoing a single IVF/ICSI cycle with GH supplementation versus conventional controlled ovarian stimulation were selected for analysis.  Primary outcome was live-birth rate (LBR); and secondary outcomes included clinical pregnancy rate, miscarriage rate, ongoing pregnancy rate, number of oocytes, number of mature (metaphase II [MII]) oocytes and the number of embryos available to transfer.  A total of 12 RCTs were included; 586 women were assigned to the intervention group and 553 to the control group.  The analysis revealed that patients receiving GH supplementation did not show an increased LBR, miscarriage rate, or ongoing pregnancy rate.  However, GH supplementation in PORs increased clinical pregnancy rate, number of oocytes retrieved (mean difference [MD] of 1.62), number of MII oocytes (MD of 2.06), and number of embryos available to transfer (MD of 0.76).  Sensitivity and subgroup analyses did not provide statistical changes to pooled results.  The authors concluded that the present meta-analysis provided evidence that GH supplementation may improve some reproductive outcomes in PORs, but not LBRs.

In a meta-analysis, Yang and co-workers (2020) examined the effect of GH supplementation in PORs undergoing IVF or ICSI.  PubMed, Medline and Cochrane Library databases were searched for the identification of relevant RCTs.  Outcome measures were LBR, clinical pregnancy rate, miscarriage rate, cycle cancelation rate, number of retrieved oocytes and total dose of gonadotropin.  A total of 15 RCTs involving 1,448 patients were eligible for the analysis.  GH supplementation improved LBR (RR, 1.74; 95 % CI: 1.19 to 2.54), clinical pregnancy rate (RR, 1.65; 95 % CI: 1.31 to 2.08) and retrieved oocytes number (SMD, 0.72; 95 % CI: 0.28 to 1.16), while reducing cancelled cycles rate (RR, 0.62; 95 % CI: 0.44 to 0.85) and dose of gonadotropin (SMD,-1.05 95 % CI: - 1.62  to -0.49) for PORs.  In additions, there was no significant difference in the miscarriage rate between GH group and control group.  The authors concluded that based on the limited available evidence, GH supplementation appeared to improve IVF/ICSI outcomes for PORs.  Moreover, these researchers stated that while GH adjuvant treatment appeared to be beneficial and relatively safe, there has been no standard protocol regarding GH addition time and dosage so far.  Studies have shown that GH may contribute to insulin resistance and may be associated with cancer.  In view of this, they stated that further RCTs with large sample sizes are needed to clarify the effect of GH adjuvant therapy in the treatment of women with poor ovarian response; in particular to establish the threshold dosage and the administration protocol of GH.

In a RCT, Mohammad et al (2021) compared the ICSI-embryo transfer (ET) outcomes in PORs who underwent ovarian stimulation by the ultrashort GnRH antagonist protocol with or without adjuvant GH injection.  This trial was carried out at Al-Azhar University from December 2018 to June 2019 in 156 patients.  All subjects received the same preparations.  After randomization, in the study group, women received GH 4 IU/day subcutaneous injection from the 2nd day of the cycle stopped 1 day before ovum pick-up.  In the control group, women received subcutaneous saline in the same dosage as in the study group.  After intervention, all procedures were the same in both groups.  The main outcome measure was the clinical pregnancy rate.  Statistical analysis was based on the intention-to-treat population.  Both groups were comparable with regard their baseline characteristics (p > 0.05).  Ovulation characteristics were comparable (p > 0.05).  The level of estradiol (E2) was significantly (p = 0.003) higher in the GH group.  The oocyte retrieved number was significantly (p < 0.001) higher in the GH group 4.94 ± 1.77 than in the control group 3.74 ± 1.82.  The mean number of MII oocytes was significantly (p < 0.001) higher in the GH group 3.3 ± 1.36 than in the control group 2.29 ± 1.24.  Fertilization characteristics, implantation rate, and pregnancy rate were comparable (p > 0.05).  The authors concluded that despite the fact that this study showed no significant increase in the clinical and chemical pregnancy rates by the addition of GH to the ultrashort antagonist protocol in PORs, the number of retrieved oocytes was significantly higher in the GH group. 

The authors stated that the main drawback of this trial was that the LBR was not reported because the follow-up of patients was not possible since they were from locations far from the hospital.  That, of course, added another drawback regarding the evaluation of the long-term safety of GH on the mothers and their children.  Furthermore, the study used a low-dose of GH that may jeopardize the effect; however, one reason for this dose was to avoid any adverse effects due to the higher doses. 

In a systematic review and meta-analysis, Liu and associates (2021) examined if additional GH treatment could improve pregnancy outcomes in POR.  This systematic review/meta-analysis was conducted in a prospective manner.  Literature search was carried out in PubMed, Embase, Web of Science, and Cochrane Library from January 2010 to June 2019, and studies before 2010 were included based on a Cochrane review published in 2010.  Only English articles and randomized clinical trial studies were included.  A total of 12 studies were included for analysis.  GH treatment in PORS significantly increased the clinical pregnancy rate (odds ratio (OR) = 1.75 (1.23 to 2.50)), and the LBR also tended to increase after GH treatment (OR = 1.51 (0.97 to 2.35)).  Other outcomes including the gonadotropin requirement, oocyte retrieval number, endometrium thickness, and the number of patients with available embryos for transfer were also improved by GH treatment (WMD = - 0.78 (- 1.23, - 0.33), 1.41 (0.72 to 2.09), 0.36 (0.18 to 0.53), OR = 2.67 (1.47, to4.68), respectively).  The authors concluded that based on the current study, GH treatment in POR could increase clinical pregnancy rate and showed a higher but not statistically significant likelihood of LBR.  The effect was likely to be mediated by improving ovarian response and endometrium thickness.  Moreover, these researchers stated that the effect of GH treatment on LBR should be tested by further studies with a larger sample size.

In a Cochrane review, Sood et al (2021) examined the safety and effectiveness of GH as an adjunct to IVF compared to standard IVF for women with infertility.  These investigators searched the following databases (to November 2020): Cochrane Gynecology and Fertility (CGF) Group specialized register, CENTRAL, Medline, Embase, CINAHL, Epistemonikos database and trial registers together with reference checking and contact with study authors and experts in the field to identify additional trials.  They included all RCTs of adjuvant GH treatment in IVF compared with no adjuvant treatment for women with infertility; and excluded trials where additional adjuvant treatments were used with GH.  These researchers also excluded trials comparing different IVF protocols.  They used standard methodological procedures recommended by Cochrane; 2 review authors independently carried out evaluation of trial risk of bias and extraction of relevant data.  The primary review outcome was LBR.  The secondary outcomes were clinical pregnancy rate, oocytes retrieved, ET, units of gonadotropin used and AEs (i.e., ectopic pregnancy, multiple pregnancy, ovarian hyperstimulation syndrome (OHSS), congenital anomalies, and edema).

These researchers included 16 RCTs (1,352 women); 2 RCTs (80 women) studied GH in routine use, and 14 RCTs (1,272 women) studied GH in poor responders.  The evidence was low to very low certainty, the main limitations being risk of bias, imprecision and heterogeneity.  Adjuvant GH compared to no adjuvant: routine use for IVF.  The evidence was very uncertain regarding the effect of GH on LBR per woman randomized for routine use in IVF (OR 1.32, 95 % CI: 0.40 to 4.43; I2 = 0 %; 2 trials, 80 participants; very low-certainty evidence).  If the chance of live birth without adjuvant GH was assumed to be 15 %, the chance of live birth with GH would be between 6 % and 43 %.  There was insufficient evidence to reach a conclusion regarding clinical pregnancy rates per woman randomized, number of women with at least 1 oocyte retrieved per woman randomized, and ET achieved per woman randomized; reported data were unsuitable for analysis.  The evidence was very uncertain regarding the effect of GH on mean number of oocytes retrieved in normal responders (MD -0.02, 95 % CI: -0.79 to 0.74; I2 = 0 %; 2 trials, 80 participants; very low-certainty evidence).  The evidence was very uncertain regarding the effect of GH on mean units of gonadotropin used in normal responders (MD 13.57, 95 % CI: -112.88 to 140.01; I2 = 0 %; 2 trials, 80 participants; very low-certainty evidence).  These investigators were uncertain of the effect of GH on AEs in normal responders.  Adjuvant GH compared to no adjuvant: use in poor responders for IVF.  The evidence was very uncertain regarding he effect of GH on LBR per woman randomized for poor responders (OR 1.77, 95 % CI: 1.17 to 2.70; I2 = 0 %; 8 trials, 737 participants; very low-certainty evidence) . If the chance of live birth without adjuvant GH was assumed to be 11 %, the chance of live birth with GH would be between 13 % and 25 %.  Adjuvant GH resulted in a slight increase in pregnancy rates in poor responders (OR 1.85, 95 % CI: 1.35 to 2.53; I2 = 15 %; 11 trials, 1,033 participants; low-certainty evidence).  The results suggested that, if the pregnancy rate without adjuvant GH was assumed to be 15 %, with GH the pregnancy rate in poor responders would be between 19 % and 31 %.  The evidence suggested that GH resulted in little to no difference in number of women with at least 1 oocyte retrieved (OR 5.67, 95 % CI: 1.54 to 20.83; I2 = 0 %; 2 trials, 148 participants; low-certainty evidence).  If the chance of retrieving at least 1 oocyte in poor responders was 81 %, with GH the chance was between 87 % and 99 %.  There was a slight increase in mean number of oocytes retrieved with the use of GH for poor responders (MD 1.40, 95 % CI: 1.16 to 1.64; I2 = 87 %; 12 trials, 1,153 participants; low-certainty evidence).  The evidence was very uncertain regarding the effect of GH on ET achieved (OR 2.32, 95 % CI: 1.08 to 4.96; I2 = 25 %; 4 trials, 214 participants; very low-certainty evidence).  If the chance of achieving ET was assumed to be 77 %, the chance with GH would be 78 % to 94 %.  Use of GH resulted in reduction of mean units of gonadotropins used for stimulation in poor responders (MD -1088.19, 95 % CI: -1,203.20 to -973.18; I2 = 91 %; 8 trials, 685 participants; low-certainty evidence).  High heterogeneity in the analyses for mean number of oocytes retrieved and units of GH used suggested that quite different effects according to differences including in trial protocols (populations, GH dose and schedule), so these results should be interpreted with caution.  These investigators were uncertain of the effect of GH on AEs in poor responders as 6 of the 14 included trials failed to report this outcome.  The authors concluded that the use of adjuvant GH in IVF treatment protocols had uncertain effect on LBR as well as mean number of oocytes retrieved in normal responders.  However, it slightly increased the number of oocytes retrieved and pregnancy rates in poor responders, while there was an uncertain effect on LBR in this group.  Moreover, these researchers stated that these findings need to be interpreted with caution, as the included trials were small and few in number, with significant bias and imprecision.  Furthermore, the dose and regimen of GH used in trials was variable; thus, further research is needed to define the role of GH as adjuvant therapy in IVF.

Improvement of Oral Health in Children with Growth Hormone Deficiency

Torlinska-Walkowiak et al (2021) stated that GH is involved in the regulation of the post-natal dental and skeletal growth; however, its effects on oral health have not been clearly defined.  These investigators reviewed current clinical knowledge of dental caries, tooth wear, developmental enamel defects, craniofacial growth and morphology, dental maturation, and tooth eruption in GH deficient (GHD) children.  They performed a systematic review using Scopus, Medline-EbscoHost and Web of Science from 2000 to May 2021.  PRISMA guidelines for reporting systematic reviews were followed.  All the selected studies involved groups under 18 years of age, covering a total of 465 GHD patients.  The studies that were selected provided reliable evidence for delayed dental maturity and orthodontic disturbances in GHD patients.  Data on dental hard tissues pathology were scarce and were limited to occurrences of dental caries.  The authors concluded that available studies indicated that children with GHD showed abnormal craniofacial morphology with reduced mandibular dimensions, with a resulting tendency to Angle’s Class II occlusion, which affected up to 31 % of the patients.  Dental age has been shown to be delayed in GHD patients by about 1 to 2 years.  Moreover, the risk of dental caries in children with GHD decreased with increasing levels of vitamin D.  The data are scarce and further studies would be valuable in evaluating the risk of various oral health problems and in organizing targeted dental care for this vulnerable group.  Moreover, these researchers stated that to gain more of an insight into the effects of this disease and its treatment on oral health and craniofacial structures, data need to be collected both before and after GH administration.  Such longitudinal studies could help clinicians to understand the complex endocrine mechanisms regulating the stomatognathic system’s development and functions, in order to provide the optimal treatment of GHD-related disturbances.

Injured Retina

Martinez-Moreno and colleagues (2019) stated that in addition to its role as an endocrine messenger, GH also acts as a neurotrophic factor in the central nervous system (CNS), whose effects are involved in neuro-protection, axonal growth, and synaptogenic modulation.  An increasing amount of clinical evidence showed a beneficial effect of GH in patients with brain trauma, stroke, spinal cord injury (SCI), impaired cognitive function, and neurodegenerative processes.  In response to injury, Muller cells trans-differentiate into neural progenitors and proliferate, which constitutes an early regenerative process in the chicken retina.  These researchers examined the long-term protective effect of GH following severe excitotoxic damage in the retina.  An acute neural injury was induced via the intravitreal injection of kainic acid (KA, 20 µg), which was followed by chronic administration of GH (10 injections [300 ng] over 21 days).  Damage provoked a severe disruption of several retinal layers.  However, in KA-damaged retinas treated with GH, these investigators observed a significant restoration of the inner plexiform layer (IPL, 2.4-fold) and inner nuclear layer (INL, 1.5-fold) thickness and a general improvement of the retinal structure.  In addition, they also observed an increase in the expression of several genes involved in important regenerative pathways, including: synaptogenic markers (DLG1, NRXN1, GAP43); glutamate receptor subunits (NR1 and GRIK4); pro-survival factors (BDNF, Bcl-2 and TNF-R2); and Notch signaling proteins (Notch1 and Hes5).  Interestingly, Muller cell trans-differentiation markers (Sox2 and FGF2) were up-regulated by this long-term chronic GH treatment.  The authors concluded that these findings were consistent with a significant increase in the number of bromodeoxyuridine-positive cells observed in the KA-damaged retina, which was induced by GH administration.  These researchers stated that these data suggested that GH was able to facilitate the early proliferative response of the injured retina and enhance the regeneration of neurite inter-connections.  They stated that this work provided additional evidence regarding the therapeutic potential of GH as a neurotrophic factor, which deserves further investigation.

Insulin-Like Growth Factor-I (IGF-1) Deficiency (Neurosecretory Defect)

Edouard et al (2009) stated that “primary IGF1 deficiency (IGFD)” is defined by low levels of IGF1 without a concomitant impairment in GH secretion in the absence of secondary cause.  These researchers evaluated the prevalence of non-GH deficient IGFD in pre-pubertal children with isolated short stature (SS) and described this population.  This retrospective study included all children with isolated SS seen in the authors’ Pediatric Endocrinology Unit from January 2005 to December 2007.  Children were included based on the following criteria:
  1. SS with current height SDS less than or equal to -2.5,
  2. age greater than or equal to 2 years, and
  3. pre-pubertal status. 

Exclusion criteria were:

  1. identified cause of SS and
  2. current or past therapy with rhGH.

IGF1-deficient children were defined as children without GH deficiency and with IGF1 levels below or equal to -2 SDS.  Among 65 children with isolated SS, 13 (20 %) had low IGF1 levels, consistent with a diagnosis of primary IGFD, 4 of which were born small for gestational age (SGA) and 9 were born appropriate for gestational age.  When compared with non-IGFD children, IGFD children had higher birth weight (-0.7 versus -1 SDS, p = 0.02) and birth height (-1.7 versus -2 SDS, p = 0.04) and more delayed bone age (2.6 versus 1.7 years, p = 0.03).  The authors concluded that the prevalence of primary IGFD was 20 % in children with isolated SS.  Concerning the pathophysiology, the findings of this study emphasized that IGFD in some children may be secondary to nutritional deficiency or to maturational delay.

Steuerman et al (2011) examined if congenital IGF1 deficiency confers protection against development of malignancies, by comparing the prevalence of malignancies in patients with congenital (secondary) deficiency of IGF1 with the prevalence of cancer in their family members.  Only patients with an ascertained diagnosis of either Laron syndrome (LS), congenital IGHD, congenital multiple pituitary hormone deficiency (cMPHD) including GH or GHRHR defect were included in this study.  In addition to the authors’ own patients, these investigators performed a worldwide survey and collected data on a total of 538 patients, 752 of their first-degree family members, of which 274 were siblings and 131 were further family members.  These researchers found that none of the 230 LS patients developed cancer and that only 1 out of 116 patients with congenital IGHD, also suffering from xeroderma pigmentosum, had a malignancy.  Out of 79 patients with GHRHR defects and out of 113 patients with congenital MPHD, these investigators found 3 patients with cancer in each group.  Among the 1st-degree family members (most heterozygotes) of LS, IGHD and MPHD, these researchers found 30 cases of cancer and 1 suspected.  In addition, 31 malignancies were reported among 131 further relatives.  The authors concluded that these findings bore heavily on the relationship between GH/IGF1 and cancer.  Homozygous patients with congenital IGF1 deficiency and insensitivity to GH such as LS seem protected from future cancer development, even if treated by IGF1.  Patients with congenital IGHD also seem protected.

Intra-Uterine Growth Restriction

Intrauterine growth restriction (IUGR) refers to a condition in which a fetus is unable to achieve its genetically determined potential size (Ross, 2013). This functional definition seeks to identify a population of fetuses at risk for modifiable but otherwise poor outcomes. This definition intentionally excludes of fetuses that are small for gestational age (SGA) but are not pathologically small. Not all fetuses that are SGA are pathologically growth restricted and, in fact, may be constitutionally small. Similarly, not all fetuses that have not met their genetic growth potential are SGA.

In a Cochrane review, Say and colleagues (2003) evaluated the effects of hormone administration for suspected impaired fetal growth and perinatal outcome.  These investigators searched the Cochrane Pregnancy and Childbirth Group trials register (November 1, 2002).  Acceptably controlled trials of hormone administration for suspected impaired fetal growth which report fetal, perinatal or maternal outcomes were selected for analysis.  Eligibility and trial quality were assessed.  No studies were included since none of the potentially relevant trials reported clinical outcomes.  The authors concluded that there is insufficient evidence to evaluate the clinical use of hormone administration for suspected impaired fetal growth.  Furthermore, an UpToDate review on “Fetal growth restriction: Evaluation and management” (Resnik, 2013) does not mention the use of GH as a management tool.

Ischemic Heart Disease

In a review on the potential of cytokines and growth factors in the treatment of ischemic heart disease, Beohar et al (2010) stated that cytokine therapy promises to provide a non-invasive treatment option for ischemic heart disease.  Several cytokines mobilize progenitor cells from the bone marrow or are involved in the homing of mobilized cells to ischemic tissue.  The recruited cells contribute to myocardial regeneration both as a structural component of the regenerating tissue and by secreting angiogenic or anti-apoptotic factors, including cytokines.  To date, randomized controlled trials (RCTs) have not reproduced the efficacy observed in pre-clinical and small-scale clinical investigations.  Nevertheless, the list of promising cytokines continues to grow, and combinations of cytokines, with or without concurrent progenitor cell therapy, warrant further investigation.  In particular, the authors noted that the effect of GH on myocardial growth, cardiac function, and IGF-1 levels in patients with  non-ischemic or ischemic cardiomyopathy, and in mixed patient populations, has been examined in several small studies.  Overall, the findings suggested that more research with GH or IGF-1 are needed, despite concerns regarding retinopathy and other potential long-term side effects.

Kabuki Syndrome

Gabrielli et al (2000) noted that Kabuki syndrome is characterized by mental retardation (mild-to-moderate), skeletal anomalies, typical facial appearance and post-natal growth deficiency.  The researchers described 2 patients with Kabuki syndrome and proven GH deficiency.  The 1st patient has been on GH replacement therapy for 4 years; the 2nd for 11 years.  There is insufficient evidence to support the use of GH for the treatment of patients with Kabuki syndrome.

Kearns-Sayre Syndrome

Quintos et al (2016) stated that Kearns-Sayre syndrome (KSS) is characterized by external ophthalmoplegia, retinal pigmentation and cardiac conduction defects due to mitochondrial DNA (mtDNA) deletions.  Short stature and GH deficiency have been reported in KSS, but data on GH treatment is limited.  These researchers described the clinical presentation, phenotype evolution, and response to GH in a patient with KSS and reported data on 8 additional KSS patients from the KIGS database.  The patient with KSS and GH deficiency achieved a final adult height at -0.8 SDS.  In the KIGS database GH treatment resulted in mean improvement in height from -3.9 to -2.9 SDS in patients with KSS.  Two patients did not show growth improvement.  These data showed improvement in height SDS in the patient and mixed results in 8 additional patients from the KIGS database after treatment with GH.  The authors concluded that heterogeneity in responsiveness may relate to presence of GH deficiency or severity of underlying mitochondrial dysfunction.

Methadone-Induced Toxicity

Nylander et al (2016) stated that human GH displays promising protective effects in the central nervous system after damage caused by various insults.  Current evidence suggests that these effects may involve N-methyl-d-aspartate (NMDA) receptor function, a receptor that also is believed to play a role in opioid-induced neurotoxicity.  These researchers examined the acute toxic effects of methadone, an opioid receptor agonist and NMDA receptor antagonist, as well as to evaluate the protective properties of rhGH on methadone-induced toxicity.  Primary cortical cell cultures from embryonic day 17 rats were grown for 7 days in-vitro.  Cells were treated with methadone for 24 hours and the 50 % lethal dose was calculated and later used for protection studies with rhGH.  Cellular toxicity was determined by measuring mitochondrial activity, lactate dehydrogenase release, and caspase activation.  Furthermore, the mRNA expression levels of NMDA receptor subunits were investigated following methadone and rhGH treatment using quantitative PCR (qPCR) analysis.  A significant protective effect was observed with rhGH treatment on methadone-induced mitochondrial dysfunction and in methadone-induced LDH release.  Furthermore, methadone significantly increased caspase-3 and -7 activation but rhGH was unable to inhibit this effect.  The mRNA expression of the NMDA receptor subunit GluN1, GluN2a, and GluN2b increased following methadone treatment, as assessed by qPCR, and rhGH treatment effectively normalized this expression to control levels.  These investigators have demonstrated that rhGH can rescue cells from methadone-induced toxicity by maintaining mitochondrial function, cellular integrity, and NMDA receptor complex expression. 

Miscellaneous Conditions in Adults

Limited data are available on the effectiveness of GH in an array of conditions in adult patients, including chronic catabolic states, older men and women, post-operative patients, those with states associated with excessive glucocorticoids, obese/morbid obese patients, osteoporosis, muscular dystrophy, and those with infertility, but no consistent benefit has been shown.  Until more data are available, however, guidelines do not recommend long-term GH therapy in these conditions.

Barake and colleagues (2018) stated that in adults, GHD has been associated with low bone mineral density (BMD), an effect counteracted by GH replacement.  Whether GH is beneficial in adults with age-related bone loss and without hypopituitarism is unclear.  These investigators conducted a systematic literature search using Medline, Embase and the Cochrane Register of Controlled Trials.  They extracted and analyzed data according to the bone outcome included [bone mineral content (BMC), BMD, and bone biomarker, fracture risk]; and performed a meta-analysis when possible.  These researchers included 8 studies; 7 randomized 272 post-menopausal women, 61 to 69 years, to GH or control, for 6 to 24 months, and the 8th was an extension trial.  Except for 1 study, all women received concurrent osteoporosis therapies.  There was no significant effect of GH, as compared to control, on BMD at the lumbar spine (WMD = -0.01 [-0.04 to 0.02]), total hip (WMD = 0 [-0.05, 0.06]) or femoral neck (WMD = 0 [-0.03, 0.04]). Similarly, no effect was seen on BMC. GH significantly increased the bone formation marker procollagen type-I carboxy-terminal propeptide (PICP) (WMD = 14.03 [2.68 to 25.38]).  Growth hormone therapy resulted in a trend for increase in osteocalcin and in bone resorption markers.  Patients who received GH had a significant decrease in fracture risk as compared to control (relative risk = 0.63 [0.46 to 0.87]).  Reported adverse events (AEs) were not major, mostly related to fluid retention.  The authors concluded that GH may not improve bone density in women with age-related bone loss but may decrease fracture risk.  Moreover, they stated that larger studies of longer duration are needed to further explore these findings in both genders, and to investigate the effect of GH on bone quality.

Non-Classic Congenital Adrenal Hyperplasia

Non-classic congenital adrenal hyperplasia (NCCAH), also termed as late onset of CAH, is a very mild form of 21-hydroxylase deficiency.  The incidence of disease is estimated at 0.1 % of population (Jesic et al, 2004).  Ghizzoni and colleagues (1996) reported that NCCAH is associated with "slight" alterations in GH secretion.  The Endocrine Society’s clinical practice guideline on “Congenital adrenal hyperplasia due to steroid 21-hydroxylase deficiency” (Speiser et al, 2010) and an UpToDate review on “Diagnosis and treatment of nonclassic (late-onset) congenital adrenal hyperplasia due to 21-hydroxylase deficiency” (Nieman, 2012) did not mention the use of GH therapy. 

Osteogenesis Imperfecta

Osteogenesis imperfecta is caused by mutations in the gene for type I collagen.  It is associated with bone demineralization and, in many instances, with retarded bone growth.  Growth hormone has not been proven to be consistently effective in improving bone growth and mineralization in patients with this condition.

Post-Bariatric Surgery

In a pilot study, Savastano et al (2009) examined if GH treatment prevents lean body mass (LBM) loss in the early post-operative period.  A total of 24 women (body mass index: 44.4 +/- 7.6 kg/m(2), aged 36.8 +/- 11.7 years) undergoing laparoscopic-adjustable silicone gastric banding (LASGB) and with GHD after LASGB was included in the study.  Group A (n = 12) included a standardized diet regimen and exercise program plus recombinant human GH (0.5 +/- 0.13 mg every day), and group B (n = 12) included a standardized diet regimen and exercise program.  The follow-up duration was 6 months.  The excess of body weight loss did not differ between groups A and B after 3 and 6 months.  At 3 months, LBM loss was lower (p < 0.0001) and fat mass (FM) loss was higher (p = 0.02) in group A than group B.  At 3 and 6 months, appendicular skeletal muscle mass loss was lower (p = 0.000) in group A than group B. At 3 (p = 0.0003 and 0.0005, respectively) and 6 months (p < 0.0001 and 0.0002, respectively), the percent changes of FM and LBM were significantly higher in group A than group B.  In both groups, fasting and post-glucose area under the plasma concentration-time curve insulin significantly reduced.  The homeostasis model assessment of insulin and insulin sensitivity indexes and total to high-density lipoprotein cholesterol ratio improved only in group A.  The authors concluded that GH therapy for 6 months after LASGB reduces loss in LBM and appendicular skeletal muscle mass during a standardized program of low-calorie diet and physical exercise program, with improvement of lipid profile and without a deterioration of glucose tolerance.  These preliminary findings need to be validated with well-designed studies.

Post-Polio Syndrome

Gupta et al (1994) stated that previous work showed low IGF-) in polio survivors compared with age-matched controls and it was hypothesized that the low IGF-I was caused by the lack of GH  secretion. These researchers examined if the nocturnal release of GH is subnormal in polio survivors and whether the low IGF-I level can be raised to the range of healthy young men (240 to 460 ng/ml) hGH treatment.  If so, what dose of hGH is required?  Does the hormone treatment affect muscle function?  A total of 11 polio survivors with evidence of post-poliomyelitis syndrome (PPS), aged 50 to 65 years, and low IGF-I levels (average IGF-I value of 170 ng/ml) were studied.  The serum level of GH was measured in the first 4 hours of sleep.  The serum IGF-I level was determined before and during hGH treatment at 0.0075, 0.015 or 0.03 mg/kg of ideal body weight (IBW), 3 times a week for successive periods of 1 month.  Before and after hGH treatment, strength was determined in knee extensor and flexor muscles and the elbow flexor and elbow extensor muscles.  Nocturnal GH was low in the polio survivors compared with healthy young men.  Serum IGF-I was raised into the target range by either 0.0075 or 0.015 mg hGH/kg 3 times a week.  After 3 months of hGH treatment, no consistent changes in muscle strength were observed in the study group.  The authors concluded that the study provided new data that the tendency for low serum IGFI level in polio survivors was caused at least in part by low endogenous GH secretion, and can be corrected by relatively low doses of hGH; however, 3 months of hormone treatment did not regularly improve muscle strength in polio survivors.  They stated that the possibility remained that treatment with hGH for longer than 3 months might be more consistently beneficial; a longer duration, double-blinded study with a larger number of participants is  needed to answer this question.

Shetty et al (1995) examined if 3  months of treatment with hGH affects muscle function in PPS subjects.  The study group consisted of 6 respondents (5 males, 1 female) from the Wisconsin Polio Resource Group who met the criteria for PPS and whose IGF-I levels were below 240 mg/ml (normal range for healthy young men is 240 to 460 ng/ml).  Serum IGF-I by RIA was performed in the authors’ laboratory.  Human growth hormone (was administered subcutaneously at 0.0075, 0.015 or 0.03 mg/kg (full replacement dose) of IBW, 3 times weekly for successive periods of 1 month.  Before and after hGH treatment evaluation of muscle strength and endurance were done in 5 subjects according to the protocol of Agre and Rodriquez.  On a separate occasion, the 6th subject who has quadriplegia and respiratory decompensation underwent pulmonary function studies before and after hGH treatment at 0.03 mg/kg thrice-weekly for 4 months.  The authors concluded that the majority of the muscle function tests showed little or no change after 3 months of hGH treatment; 3 of the 6 subjects showed improvement in a few areas.  They noted that the possibility remained that treatment with hGH for longer than 3 months might be beneficial.

Trojan et al (2001) examined if serum IGF-I levels are associated with strength, body mass index (BMI), fatigue, or quality of life in PPS.  Post-polio syndrome is likely due to a distal disintegration of enlarged post-polio motor units as a result of terminal axonal sprouting.  Age-related decline in GH and IGF-I (which support terminal axonal sprouts) has been proposed as a contributing factor.  As part of the North American Post-Poliomyelitis Pyridostigmine Study (NAPPS), baseline data on maximum voluntary isometric contraction (MVIC), BMI, subjective fatigue (fatigue severity scale, Hare fatigue symptom scale), health-related quality of life (short form health survey-36; SF-36), and serum IGF-I levels were gathered on 112 PPS patients.  Pearson correlation coefficients were calculated to evaluate the association between serum IGF-I and MVIC in 12 muscles, BMI, 2 fatigue scales, and SF-36 scale scores.  There is a significant inverse correlation of IGF-I levels with MVIC in left ankle dorsiflexors (r = -0.30, p < 0.01), and left and right knee extensors (r = -0.22, -0.25, p = < 0.01, 0.01), but no significant correlations in other muscles.  When men and women were evaluated separately, inverse correlations of IGF-I levels with MVIC were found only in men.  Insulin-like growth factor 1 correlated inversely with BMI (r = -0.32, p = 0006) and age (r = -0.32, p = 0.0005); IGF-I did not correlate with the fatigue or SF-36 scales.  The authors concluded that in this exploratory study, they found that contrary to expectations, IGF-I did not correlate positively with strength.  Insulin-like growth factor 1 correlated negatively with strength in several lower extremity muscles, BMI, and age; IGF-I is likely not an important factor in the pathogenesis of fatigue and in determining quality of life in PPS, but its role on strength should be studied further.

Furthermore, an UpToDate review on “Post-polio syndrome” (Simionescu and Jubelt, 2015) does not mention GH as a therapeutic option.

Pseudopseudohypoparathyroidism

Manfredi et al (1993) presented the case of a pre-pubertal girl with the characteristic somatic features of Albright's hereditary osteodystrophy, including severe short stature, cataracts and shortening of all metacarpals and metatarsals and of the second middle hand phalanges, whose diagnosis of pseudopseudohypoparathyroidism (PPHP) was confirmed by laboratory evaluation (normocalcemia, normophosphatemia, normal levels of circulating PTH and normal response to exogenous PTH).  Since an isolated idiopathic GH deficiency has been diagnosed at the age of 9.7 years, by an abnormal GH response to standard provocation tests, a poor spontaneous nocturnal GH secretion and a blunted response to GHRH test, the patient was treated with biosynthetic GH during a 3.5-year period.  Although a good improvement of growth velocity was obtained when comparing pre-treatment height velocity (4 cm/yr) with growth velocity evaluated during GH treatment (6.6, 6.2 and 5.9 cm/yr in the first, the second and the third year of therapy, respectively), bone age advanced more rapidly than chronological age, so that it is uncertain whether the growth acceleration promoted by GH administration really improved final height, which remained below the third centile.  This patient was the first described case of PPHP associated with idiopathic GH deficiency, and the second report of long-term GH treatment in a subject with PPHP.  They stated that further observations are needed to define the frequency and significance of GH deficiency and the role of GH replacement therapy in pseudohypoparathyroidism- and PPHP-associated short stature.

Mantovani et al (2010) noted that since the identification of GH deficiency due to resistance to GHRH in patients with pseudohypoparathyroidism type Ia (PHP-Ia), no study investigated the effects of recombinant human GH (rhGH) therapy on height velocity (HV) in these patients.  To address this question, 8 pre-pubertal PHP-Ia children with GH deficiency (7 girls and 1 boy, aged 5.8 to 12 years) underwent a 3 to 8 year treatment with rhGH.  Height and HV were measured before and at 6-month intervals during therapy.  Nine sex- and age-matched children with idiopathic GH deficiency were monitored during rhGH therapy for comparison.  In PHP-Ia children, height S.D. scores increased from -2.4 ± 0.58 to -1.8 ± 0.47 (p = 0.04) after 12 months, this increase being maintained after the second (-1.6 ± 0.6) and third (-1.15 ± 0.6) year of therapy, similarly to what recorded in children with idiopathic GH deficiency.  The HV and HV S.D. scores after 3 years maintained a significant increase from 3.5 ± 0.6 to 7.0 ± 0.9 cm/yr (p < 0.0001) and from -2.8 ± 0.8 to +2.2 ± 1.0 (p < 0.0001), respectively.  Six patients treated for 4 to 8 years had a reduced pubertal spurt and did not improve their near-adult height, with the only exception of 1 patient in whom estrogen production was blocked by GnRH analogs.  The authors concluded that this was first study on the effectiveness of rhGH replacement therapy in pre-pubertal children with PHP-Ia and provided indication that treatment of GH deficiency should be started soon due to the rather limited time window for a potentially effective therapy.

Short Stature associated with Crohn's Disease

Growth failure often complicates Crohn's disease in childhood.  Abnormalities in the GH/insulin-like growth factor-1 axis may occur.  In a randomized controlled study, Calenda et al (2005) examined the effects of administered GH on growth in these patients.  A total of 7 children (6 boys and 1 girl; age of 11.9 to 16 years) with Crohn's disease and growth failure were enrolled.  In phase 1, patients were randomized to either GH (0.05 mg/kg per day) or placebo; in phase 2, patients who received placebo during the first year received GH for various time periods.  Follow-up was every 3 months for up to 2 years.  During placebo treatment (4 patients), mean height-for-age z score (haz) increased 0.23 in the first half year and 0.55 in the second half year.  The mean improvement in haz during the first half year of GH treatment (7 patients) was 0.13; during the second half year (5 patients), haz decreased 0.01.  Effects of GH varied among patients; 2 patients grew only when nutritional supplementation was added.  Observed changes were not statistically significant.  Serum insulin-like growth factor-1 levels correlated with height velocity.  Only 2 patients later reached expected adult height.  These investigators concluded that GH treatment at the dose given did not stimulate growth in children with Crohn's disease and short stature.  Whether or not GH plus nutritional therapy would be effective in promoting sustained catch-up growth remains to be determined.

Skeletal Dysplasias

Growth hormone has been tried in several skeletal dysplasias associated with short stature, most notably achondroplasia.  Although GH treatment of patients with achondroplasia has induced some growth acceleration, the literature shows the growth velocities achieved have been insufficient to produce catch-up growth.  Thus, the height of these patients is not sufficiently altered so that it can approach the normal range for height.

Kyphomelic dysplasia is a bone dysplasia with severe rhizomelic limb shortening, bowed extremities and dimples over the bowing.  Other reported features include truncal shortening, short stature, and micrognathia.  Intelligence is normal.  There is spontaneous improvement of the bowing with growth.  There is a lack of evidence on the use of GH in kyphomelic dysplasia.

Stem Cell Mobilization

In a review on "Novel agents and approaches for stem cell mobilization in normal donors and patients", Bakanay and Demirer (2012) listed GH as one of the investigational agents.  They noted that in the future, thrombopoietin-receptor agonists may be potential adjuncts to granulocyte colony-stimulating factor in poor mobilizers.

Testicular Cancer Survivors

UpToDate reviews on “Overview of the treatment of testicular germ cell tumors” (Oh, 2015) and “Approach to the care of long-term testicular cancer survivors” (Beard and Vaughn, 2015) do not mention the use of growth hormone as a management tool.

Thalassemia

Ngim and colleagues (2020) stated that thalassemia is a blood disorder that leads to anemia of varying severity.  In individuals afflicted by the more severe forms, regular blood transfusions are needed that may result in iron-overload.  Accumulated iron from blood transfusions may be deposited in vital organs including the heart, liver and endocrine organs such as the pituitary glands that could affect GH production; GHD is one of the factors that could result in short stature, a common complication in individuals with thalassemia.  Growth hormone replacement therapy has been used in children with thalassemia who have short stature and GHD.  This review on the role of GH was originally published in September 2017 and updated in April 2020.  These investigators searched the Cochrane Haemoglobinopathies Trials Register, compiled from electronic database searches and hand-searching of journals and conference abstract books. Date of latest search was November 14, 2019.  They also searched the reference lists of relevant articles, reviews and clinical trial registries.  Date of latest search was January 6, 2020.  Randomized and quasi-randomized controlled trials comparing the use of GH therapy to placebo or standard care in individuals with thalassemia of any type or severity were selected for analysis.  Two authors independently selected trials for inclusion.  Data extraction and assessment of risk of bias were also conducted independently by 2 authors.  The certainty of the evidence was assessed using GRADE criteria. 

These researchers included 1 parallel trial conducted in Turkey.  The trial recruited 20 children with homozygous beta thalassemia who had short stature; 10 children received GH therapy administered subcutaneously on a daily basis at a dose of 0.7 IU/kg/week and 10 children received standard care.  The overall risk of bias in this trial was low except for the selection criteria and attrition bias that were unclear.  The certainty of the evidence for all major outcomes was moderate, the main concern was imprecision of the estimates due to the small sample size leading to wide CIs.  Final height (cm) (the review's pre-specified primary outcome) and change in height were not assessed in the included trial.  The trial reported no clear difference between groups in height standard deviation (SD) score after 1 year, MD of -0.09 (95 % CI: -0.33 to 0.15 (moderate-certainty evidence).  However, modest improvements appeared to be observed in the following key outcomes in children receiving GH therapy compared to control (moderate-certainty evidence): change between baseline and final visit in height SD score, MD of 0.26 (95 % CI: 0.13 to 0.39); height velocity, MD of 2.28 cm/year (95 % CI: 1.76 to 2.80); height velocity SD score, MD of 3.31 (95 % CI: 2.43 to 4.19); and change in height velocity SD score between baseline and final visit, MD of 3.41 (95 % CI: 2.45 to 4.37).  No adverse effects of treatment were reported in either group; however, while there was no clear difference between groups in the oral glucose tolerance test at 1 year, fasting blood glucose was significantly higher in the GH therapy group compared to control, although both results were still within the normal range, MD of 6.67 mg/dL (95 % CI: 2.66 to 10.68).  There were no data beyond the 1-year trial period. 

The authors concluded that a small single trial contributed evidence of moderate certainty that the use of GH for 1 year may improve height velocity of children with thalassemia via height SD score in the treatment group was similar to the control group.  There are no RCTs in adults or trials that address the use of GH therapy over a longer period and examine its effect on final height and QoL.  These researchers stated that the optimal dosage of GH and the ideal time to start this therapy remain uncertain; they stated that large, well-designed RCTs over a longer period with sufficient duration of follow-up are needed.

Traumatic Brain Injury

Mossberg et al (2017) examined the effects of rhGH replacement on physical and cognitive functioning in subjects with a moderate-to-severe traumatic brain injury (TBI) with abnormal GH secretion.  A total of 15 individuals who sustained a TBI at least 12 months prior to study enrollment were identified as having abnormal GH secretion by glucagon stimulation testing (maximum GH response less than 8 ng/ml).  Peak cardio-respiratory capacity, body composition, and muscle force testing were assessed at baseline and 1 year after rhGH replacement.  Additionally, standardized neuropsychological tests that evaluate memory, processing speed, and cognitive flexibility, as well as self-report inventories related to depression and fatigue, were administered at baseline and 1 year after rhGH replacement.  Comparison tests were performed with proper post-hoc analyses.  All analyses were carried out at α < 0.05.  Peak O2 consumption, peak oxygen pulse (estimate of cardiac stroke volume), and peak ventilation all significantly increased (p < 0.05).  Maximal isometric and isokinetic force production were not altered.  Skeletal muscle fatigue did not change but the perceptual rating of fatigue was reduced by approximately 25 % (p = 0.06).  Cognitive performance did not change significantly over time, whereas self-reported symptoms related to depression and fatigue significantly improved.  The authors concluded that the observed changes suggested that rhGH replacement has a positive impact on cardio-respiratory fitness and a positive impact on perceptual fatigue in survivors of TBI with altered GH secretion.  These preliminary findings need to be validated by well-designed studies.

Szarka and colleagues (2021) stated that one of the most devastating chronic consequences of TBI is cognitive impairment.  One of the possible underlying causes is GHD caused by TBI-induced hypopituitarism.  Currently, TBI patients are not routinely screened for pituitary function, and there are no standard therapies when GHD is diagnosed.  Furthermore, the possible positive effects of GH replacement on cognitive function and QoL following TBI are not well established.  In a systematic review, these investigators examined the evidence regarding the effect of GH therapy on cognitive function and QoL following TBI.  They carried out a literature search in PubMed, Embase, and Central databases from inception to October 2019; and extracted data on each term of severity (mild-moderate-severe) of TBI with and without GHD, time since injury, parameters of GH treatment (dosing, length), and cognitive outcomes in terms of verbal and non-verbal memory, as well as executive, emotional, and motor functions, and performed a meta-analysis on the results of a digit span test evaluating working memory.  These researchers identified 12 studies (containing 2 RCTs) with 264 mild-to-moderate-to-severe TBI patients (Glasgow Coma Score [GCS] varied between 6 and 15) with (n = 255) or without (n = 9) GHD who received GH therapy.  GH was administered subcutaneously in gradually increasing doses, monitoring serum IGF-I level.  After TBI, regardless of GCS, 6 to 12 months of GH therapy, started in the chronic phase post-TBI, induced a moderate improvement in processing speed and memory capacities, decreased the severity of depression, and led to a marked improvement in QoL.  The authors concluded that these findings indicated the need for further multi-center, controlled trials to substantiate the use of GH replacement therapy as a potential tool to alleviate TBI-related cognitive impairment and improve QoL.  These researchers stated that drawbacks of this review included the relatively low number of patients involved and the divergent neuropsychological tests used.

Provacative Testing

Arginine and Glucagon Provocative Testing for the Diagnosis of Growth Hormone Deficiency

An UpToDate review on “Diagnosis of growth hormone deficiency in children” (Richmond and Rogol, 2021) states that “If GH stimulation tests are undertaken for children with suspected isolated GHD (i.e., with no evidence of other pituitary hormone deficiencies), testing with 2 different provocative tests is recommended to establish the diagnosis.  Clonidine, arginine, and glucagon are common choices in children.  Hypothyroidism should be excluded first by performing thyroid function tests.  Provocative GH testing has a number of limitations”.


Appendix

Appendix A: Examples of Hypothalamic/Pituitary/CNS Disorders

Congenital genetic abnormalities

  • Transcription factor defects (PIT-1, PROP-1, LHX3/4, HESX-1, PITX-2)
  • Growth hormone releasing hormone (GHRH) receptor gene defects
  • GH secretagogue receptor gene defects
  • GH gene defects
  • GH receptor/post receptor defects

Congenital structural abnormalities

  • Optic nerve hypoplasia/septo-optic dysplasia
  • Agenesis of corpus callosum
  • Empty sella syndrome
  • Ectopic posterior pituitary
  • Pituitary aplasia/hypoplasia
  • Pituitary stalk defect
  • Holoprosencephaly
  • Encephalocele
  • Hydrocephalus
  • Anencephaly or prosencephaly
  • Arachnoid cyst 
  • Other mid-line facial defects (e.g., single central incisor, cleft lip/palate)
  • Vascular malformations

Acquired structural abnormalities (or causes of hypothalamic/pituitary damage)

  • CNS tumors/neoplasms (e.g., craniopharyngioma, glioma/astrocytoma, pituitary adenoma, germinoma)
  • Cysts (Rathke cleft cyst or arachnoid cleft cyst)
  • Surgery
  • Radiation
  • Chemotherapy
  • CNS infections
  • CNS infarction (e.g., Sheehan’s syndrome)
  • Inflammatory processes (e.g., autoimmune hypophysitis)
  • Infiltrative processes (e.g., sarcoidosis, histiocytosis, hemochromatosis)
  • Head trauma/traumatic brain injury
  • Aneurysmal subarachnoid hemorrhage
  • Perinatal or postnatal trauma 
  • Surgery of the pituitary or hypothalamus

Appendix B: Pituitary Hormones (Other than Growth Hormone)

  • Adrenocorticotropic hormone (ACTH)
  • Antidiuretic hormone (ADH)
  • Follicle stimulating hormone (FSH)
  • Luteinizing hormone (LH)
  • Thyroid stimulating hormone (TSH)
  • Prolactin

Appendix C: Requirements for GH-Stimulation Testing in Adults

Testing for adult GHD is not required

  • Three or more pituitary hormone deficiencies and low IGF-1
  • Congenital structural abnormalities

    • Transcription factor defects (PIT-1, PROP-1, LHX3/4, HESX-1, PITX-2
    • GHRH receptor-gene defects
    • GH-receptor/post-receptor defects
    • GH-gene defects associated with brain structural defects
    • Single central incisor
    • Cleft lip/palate

  • Acquired causes such as perinatal insults

Testing for adult GHD is required

  • Acquired

    • Skull-base lesions
    • Pituitary adenoma
    • Craniopharyngioma
    • Rathke’s cleft cyst
    • Meningioma
    • Glioma/astrocytoma
    • Neoplastic sellar and parasellar lesions
    • Chordoma
    • Hamartoma
    • Lymphoma
    • Metasteses
    • Other brain injury
    • Traumatic brain injury
    • Sports-related head trauma
    • Blast injury
    • Infiltrative/granulomatous disease
    • Langerhans cell histiocytosis\
    • Autoimmune hypophysitis (primary or secondary)
    • Sarcoidosis
    • Tuberculosus
    • Amyloidosis

  • Surgery to the sella, suprasellar, and parasellar region
  • Crainial irradiation
  • Central nervous system infections (bacteria, viruses, fungi, parasites)
  • Infarction/hemorrhage (e.g., apoplexy, Sheehan’s syndrome, subarachnoid hemorrhage, ischemic stroke, snake bite)
  • Empty sella
  • Hydrocephalus
  • Idiopathic

Appendix D: Calculation of Body Mass Index (BMI)

                  Weight (pounds) x 703               Weight (kg)

BMI =        ------------------------------     OR     -----------------

                  [Height (inches)]2                       [Height (m)]2

BMI classification

Table: BMI Classificiation
Underweight < 18.5 kg/m2
Normal weight 18.5 – 24.9 kg/m2
Overweight 25 – 29.9 kg/m2
Obesity (class 1) 30 – 34.9 kg/m2
Obesity (class 2) 35 – 39.9 kg/m2
Extreme obesity (class 3) ≥ 40 kg/m2

Appendix E: Alernative Therapies for HIV Wasting

  • Cyproheptadine
  • Marinol (dronabinol)
  • Megace (megestrol acetate)
  • Testosterone therapy if hypogonadal

Appendix F: Growth Charts

Growth charts for infants, children and adolescents are posted at the following internet sites:

Appendix G: Height Velocity Tables

Table: Height Velocity (cm/year) -- Girls
Age 3rd Percentile 10th Percentile 25th Percentile
2.0
2.5
3.0
3.5
4.0
4.5
5.0
5.5
6.0
6.5
7.0
7.5
8.0
8.5
9.0
9.5
10.0
10.5
11.0
11.5
12.0
12.5
13.0
13.5
14.0
14.5
6.4
5.8
5.5
5.2
5.0
4.8
4.6
4.5
4.4
4.3
4.3
4.3
4.2
4.1
4.1
4.2
4.4
4.8
5.8
6.1
5.2
3.5
2.4
1.3
0.3
0
7.1
6.7
6.3
6.0
5.8
5.4
5.2
5.0
5.0
4.9
4.8
4.8
4.7
4.7
4.8
4.9
5.0
4.6
6.4
6.9
6.3
4.6
3.0
1.7
0.8
0.2
8.4
7.7
7.2
6.7
6.4
6.2
5.9
5.7
5.6
5.5
5.4
5.3
5.2
5.2
5.2
5.3
5.6
6.1
7.0
7.5
6.8
5.5
3.7
2.4
1.4
0.5
Table: Height Velocity (cm/year) -- Boys
Age 3rd Percentile 10th Percentile 25th Percentile
2.0
2.5
3.0
3.5
4.0
4.5
5.0
5.5
6.0
6.5
7.0
7.5
8.0
8.5
9.0
9.5
10.0
10.5
11.0
11.5
12.0
12.5
13.0
13.5
14.0
14.5
15.0
15.5
16.0
16.5
17.0
6.3
5.7
5.4
5.1
4.9
4.8
4.6
4.4
4.3
4.2
4.1
4.0
3.8
3.8
3.8
3.8
3.7
3.7
3.7
3.8
4.0
4.9
6.1
7.1
6.0
4.3
2.3
1.1
0.3
0.0
0.0
6.7
6.4
6.1
5.8
5.7
5.5
5.3
5.1
5.0
4.9
4.7
4.6
4.5
4.4
4.4
4.3
4.2
4.2
4.2
4.2
4.7
5.5
7.3
8.0
7.2
5.1
3.5
2.2
1.2
0.5
0.1
8.0
7.4
7.0
6.6
6.3
6.1
5.9
5.7
5.7
5.4
5.3
5.2
5.0
4.9
4.8
4.7
4.7
4.6
4.6
4.8
5.1
6.2
8.0
8.7
7.5
5.6
4.0
2.6
1.6
1.0
0.5

Source: Interpolated from data from Tanner and Davies (1995).

Conversion Factor: 1 centimeter (cm) = 0.394 inches (in). 1 in = 2.54 cm.

Note: Assuming a normal distribution, 1 standard deviation below the mean is approximately equal to the 16th percentile, 2 standard deviations below the mean is equal to the 2nd percentile, and 3 standard deviations below the mean is equal to the 1/10th (0.1) percentile.

Appendix H: Insulin-like Growth Factor I (IGF-1) ELISA

IGF-1 Values 2 Standard Deviation Scores Below the Mean for Age:

It is recommended that the laboratory's own reference ranges be used in determining whether the patient's IGF-1 value is abnormal (falling 2 or more standard deviations below the mean for age and sex).  If the laboratory's own reference range is not available, the following may be used as a guide to IGF-1 values falling 2 or more standard deviations below the mean:

Table: IGF-1 Values 2 Standard Deviation Scores Below the Mean for Age
Age (years) Males (ng/ml) Females (ng/ml)
0-5
6-8
9-11
12-15
16-19
20-39
40-54
> 54
17
88
110
202
182
122
75
48
17
88
117
261
182
122
75
48

Conversion factor:

ng/ml * 0.13 = nmol/L

nmol/L * 7.7 = ng/ml

Key: ng = nanogram; ml = milliliter; nmol = nanomole; L = liter

Source: Barker, 2003; OHSU, 2003.

Appendix I: Growth Hormone QoL Assessment Instrument

Adult Growth Hormone Deficiency Assessment (QoL - AGHDA)

Instructions: Indicate whether each of the following statements below applies to you:

Table: Adult Growth Hormone Deficiency Assessment (QoL - AGHDA)
Statement Yes No
I have to struggle to finish jobs
I feel a strong need to sleep during the day
I often feel lonely even when I am with other people
I have to read things several times before they sink in
It is difficult for me to make friends
It takes a lot of effort for me to do simple tasks
I have difficulty controlling my emotions
I often lose track of what I want to say
I lack confidence
I have to push myself to do things
I often feel very tense
I feel as if I let people down
I find it hard to mix with people
I feel worn out even when I've not done anything
There are times when I feel very low
I avoid responsibility if possible
I avoid mixing with people I don't know well
I feel as if I am a burden to people
I often forget what people have said to me
I find it difficult to plan ahead
I am easily irritated by other people
I often feel too tired to do the things I ought to do
I have to force myself to do all the things that need doing
I often have to force myself to stay awake
My memory lets me down

Scoring

1 point for each "Yes" answer.

Source: McKenna et al, 1999. 

Appendix J: Weight for Gestational Age Table

Small for gestational age is generally defined as weight for gestational age below the 10th percentile at birth.  A chart indicating the 10th and 90th percentiles of weight for gestational age at birth is presented in Figure 17-1 of the International Association for Maternal and Neonatal Health's Neonatal Care Manual (Woods et al, 2005), and is available at the following website: Geneva Foundation for Medical Education and Research.

Appendix K: Normal Results of a GH Intravenous Stimulation Test

  • Normal peak value - at least 10 ng/ml
  • Indeterminate - 5 to 10 ng/ml
  • Subnormal - 5 ng/ml

Note: A normal value rules out hGH deficiency; in some laboratories, the normal level is 7 ng/ml.

Source: NIL/NLM (2019)

Appendix L: Brands of Growth Hormone and FDA-Approved Indications

Table: Brands of Growth Hormone and FDA-Approved Indications
FDA-Approved Indications Brands
Diagnosis of adult growth hormone deficiency Macrilen
Growth failure associated with chronic kidney disease Nutropin AQ
Growth failure associated with Noonan syndrome Norditropin
Growth failure associated with Prader-Willi syndrome Genotropin, Norditropin, Omnitrope
Growth failure associated with Turner syndrome Genotropin, Humatrope, Norditropin, Nutropin AQ, Omnitrope, and Zomacton
Growth failure in children due to inadequate secretion of endogenous growth hormone Genotropin, Humatrope, Norditropin, Nutropin AQ, Omnitrope, Saizen, and Zomacton
Children born small for gestational age (SGA) who fail to manifest catch-up growth  Genotropin, Humatrope, Norditropin, Omnitrope, and Zomacton 
Growth hormone deficiency in adults Genotropin, Humatrope, Norditropin, Nutropin AQ, Omnitrope, Saizen, Sogroya, and Zomacton
Idiopathic short statureFootnotes* 
Genotropin, Humatrope, Norditropin, Nutropin AQ, Omnitrope, and Zomacton
Short bowel syndrome Zorbtive
Short stature homeobox–containing gene deficiency (SHOX deficiency) Humatrope and Zomacton
Wasting or cachexia associated with HIV Serostim

Footnotes* Aetna does not cover GH for this indication (see policy section).

Source: For somatropin products: Genotropin, Humatrope, Norditropin, Nutropin AQ, Omnitrope, Saizen, Serostim, Zomacton, and Zorbtive: Adapted from Drug Facts & Comparisons, 2021. For somapacitan-beco (Sogroya): Novo Nordisk, 2020


References

The above policy is based on the following references:

  1. Abbassi V. Growth and normal puberty. Pediatrics. 1998;102(2):S507-S511.
  2. Abduljabbar T, Kellesarian SV, Vohra F, et al. Effect of growth hormone supplementation on osseointegration: A systematic review and meta-analyses. Implant Dent. 2017;26(4):613-620.
  3. Adema AY, de Roij van Zuijdewijn CLM, Hoenderop JG; NIGRAM consortium. Influence of exogenous growth hormone administration on circulating concentrations of α-klotho in healthy and chronic kidney disease subjects: A prospective, single-center open case-control pilot study. BMC Nephrol. 2018;19(1):327.
  4. Ahmad G, Brown J, Duffy JMN, et al. Growth hormone for in vitro fertilization. Cochrane Database Syst Rev. 2009;(4):CD000099. 
  5. Albanese A, Stanhope R. Growth and metabolic data following growth hormone treatment of children with intrauterine growth retardation. Hormone Res. 1993:39(1-2):8-12.
  6. Alberta Heritage Foundation for Medical Research (AHFMR). Human growth hormone for Turner's syndrome. Technote TN 15. Edmonton, AB: AHFMR; 1998:11.
  7. Albertsson-Wikland K. Growth hormone secretion and growth hormone treatment in children with intrauterine growth retardation. Acta Paediatr Scand Suppl. 1989;349:35-41.
  8. Ali O, Shim M, Fowler E, et al. Growth hormone therapy improves bone mineral density in children with cerebral palsy: A preliminary pilot study. J Clin Endocrinol Metab. 2007;92(3):932-937.
  9. Allen DB, Brook CGD, Bridges NA, et al. Therapeutic controversies: Growth hormone treatment of non-GH deficient subjects. J Clin Endo Metab. 1994;79:1239-1247.
  10. Allen DB, Julius JR, Breen TJ, Attie KM. Treatment of glucocorticoid-induced growth suppression with growth hormone. National Cooperative Growth Study. J Clin Endocrinol Metab. 1998;83(8):2824-2829.
  11. Allen NG, Bangalore Krishna K, Lee PA. Use of gonadotropin-releasing hormone analogs in children. Curr Opin Pediatr. 2021;33(4):442-448.
  12. Allphamed Pharbil Arzneimittel GmbH. Macrilen (macimorelin) for oral solution. Prescribing Information. Goettingen, Germany: Allphamed Pharbil Arzneimittel GmbH; revised July 2021.
  13. Altmae S, Aghajanova L. Growth hormone and endometrial receptivity. Front Endocrinol (Lausanne). 2019;10:653.
  14. American Academy of Pediatrics Committee on Drugs and Committee on Bioethics. Considerations related to the use of recombinant human growth hormone in children. Pediatrics. 1997;99(1):122-129.
  15. American Association of Clinical Endocrinologists (AACE) and American College of Endocrinology (ACE). AACE clinical practice guidelines for growth hormone use in adults and children. Jacksonville, FL: AACE; 1998. 
  16. American Association of Clinical Endocrinologists (AACE). Growth Hormone Usage in Short Children: American Association of Clinical Endocrinologists Position Statement. Jacksonville, FL: AACE, undated. Available at: https://www.aace.com/files/position-statements/shortchildren.pdf. Accessed January 28, 2019.
  17. American Association of Clinical Endocrinologists Acromegaly Guidelines Task Force. Medical guidelines for clinical practice for the diagnosis and treatment of acromegaly – 2011 update. Endocr Pract. 2011;17(suppl 4):1-44.
  18. American Association of Clinical Endocrinologists Growth Hormone Task Force. Medical guidelines for clinical practice for growth hormone use in adults and children 2003 Update. Endocr Pract. 2003;9(1):64-76.
  19. American Association of Clinical Endocrinologists. Medical guidelines for clinical practice for growth hormone use in growth hormone-deficient adults and transition patients 2009 update. Endocr Pract. 2009;15(2):1-28.
  20. American Society of Health-System Pharmacists, Inc. American Hospital Formulary Service Drug Information. Bethesda, MD: American Society of Health-System Pharmacists; updated periodically.
  21. Anthony D, Milne R. Growth hormone for growth hormone deficient adults. Development and Evaluation Committee Report No. 75. Wessex Institute for Health Research and Development, Development and Evaluation Service. Bristol, UK: R&D Directorate, South and West Regional Health Authority; September 1997.
  22. Anthony D, Stevens A. Growth hormone in children (for growth hormone deficiency, Turner's syndrome, chronic renal failure and idiopathic short stature). Development and Evaluation Committee Report No. 57. Wessex Institute for Health Research and Development, Development and Evaluation Service. Bristol, UK: R&D Directorate, South and West Regional Health Authority; June 1996.
  23. Antoniazzi F, Zamboni G. Central precocious puberty: Current treatment options. Paediatr Drugs. 2004;6(4):211-231.
  24. Ascendis Pharma A/S. Ascendis Pharma A/S announces U.S. Food and Drug Administration approval of Skytrofa (lonapegsomatropin-tcgd), the first once-weekly treatment for pediatric growth hormone deficiency. Press Release. Copenhagen, Denmark:Ascendis; August 25, 2021a. 
  25. Ascendis Pharma, Inc. Skytrofa (lonapegsomatropin-tcgd) for injection, for subcutaneous use. Prescribing Information. Palo Alto, CA: Ascendis; revised October 2022.
  26. Bakanay SM, Demirer T. Novel agents and approaches for stem cell mobilization in normal donors and patients. Bone Marrow Transplant. 2012;47(9):1154-1163.
  27. Ball C. Growth hormone replacement therapy for adults with growth hormone deficiency. In: STEER: Succinct and Timely Evaluated Evidence Review. DR Foxcroft, V Muthu, eds. London, UK: Wessex Institute for Health Research & Development, University of Southampton; 2002:2(1).
  28. Ballard H, Fuell W, Elwy R, et al. Effects of growth hormone therapy in pediatric patients with growth hormone deficiency and Chiari I malformation: A retrospective study. Childs Nerv Syst. 2020;36(4):835-839.
  29. Barake M, Arabi A, Nakhoul N, et al. Effects of growth hormone therapy on bone density and fracture risk in age-related osteoporosis in the absence of growth hormone deficiency: A systematic review and meta-analysis. Endocrine. 2018;59(1):39-49.
  30. Barker AN, ed. LabPLUS Electronic Handbook. Auckland District Health Board. Auckland, NZ: LabPLUS; July 16, 2003. Available at: http://www.adhb.govt.nz/LabPlusHandbook/. Accessed August 26, 2003.
  31. Baxter L, Bryant J, Cave CB, Milne R. Recombinant growth hormone for children and adolescents with Turner syndrome. Cochrane Database Syst. Rev. 2007;(1):CD003387.
  32. Beard CJ, Vaughn DJ. Approach to the care of long-term testicular cancer survivors. UpToDate [online serial]. Waltham, MA: UpToDate; reviewed December 2015.
  33. Beohar N, Rapp J, Pandya S, Losordo DW. Rebuilding the damaged heart: The potential of cytokines and growth factors in the treatment of ischemic heart disease. J Am Coll Cardiol. 2010;56(16):1287-1297.
  34. Blackman MR. Use of growth hormone secretagogues to prevent or treat the effects of aging: Not yet ready for prime time. Ann Intern Med. 2008;149(9):677-679.
  35. Blum WF, Crowe BJ, Quigley CA, et al. Growth hormone is effective in treatment of short stature associated with short stature homeobox-containing gene deficiency: two-year results of a randomized, controlled, multicenter trial. J Clin Endocrinol Metab. 2007;92:219-228. 
  36. Blum WF, Crowe BJ, Quigley CA, et al; SHOX Study Group. Growth hormone is effective in treatment of short stature associated with short stature homeobox-containing gene deficiency: Two-year results of a randomized, controlled, multicenter trial. J Clin Endocrinol Metab. 2007;92(1):219-228.  
  37. Blum WF, Ross JL, Zimmermann AG, et al. GH treatment to final height produces similar height gains in patients with SHOX deficiency and Turner syndrome: results of a multicenter trial. J Clin Endocrinol Metab. 2013;98(8):E1383-92.
  38. Boguszewski M, Albertsson-Wikland K, Aronsson S, et al. Growth hormone treatment of short children born small-for-gestational-age: The Nordic Multicentre Trial. Acta Paediatr Scand. 1998;87(3):257-263.
  39. Bramnert M, Segerlantz M, Laurila E, et al. Growth hormone replacement therapy induces insulin resistance by activating the glucose-fatty acid cycle. J Clin Endocrinol Metab. 2003;88(4):1455-1463.
  40. Breederveld RS, Tuinebreijer WE. Recombinant human growth hormone for treating burns and donor sites. Cochrane Database Syst Rev. 2014;9:CD008990.
  41. Brown TT. Approach to the human immunodeficiency virus-infected patient with lipodystrophy. J Clin Endocrinol Metab. 2008;93(8):2937-2945.
  42. Bryant J, Baxter L, Cave CB, Milne R. Recombinant growth hormone for idiopathic short stature in children and adolescents. Cochrane Database Syst Rev. 2007;(3):CD004440.
  43. Bryant J, Cavel C, Mihaylova B, et al. Clinical effectiveness and cost-effectiveness of growth hormone in children: A systematic review and economic evaluation. Health Technol Assess. 2002;6(18):1-168.
  44. Bryant J, Loveman E, Chase D, et al. Clinical effectiveness and cost-effectiveness of growth hormone in adults in relation to impact on quality of life: A systematic review and economic evaluation. Health Technol Assess. 2002;6(19):1-106.
  45. Butenandt O, Lang G. Recombinant human growth hormone in short children born small for gestational age. J Clin Endocrinol Metab. 1997;10(3):275-282.
  46. Butler MG, Miller JL, Forster JL. Prader-Willi syndrome – clinical genetics, diagnosis and treatment approaches: An update. Curr Pediatr Rev. 2019;15(4):207-244.
  47. Calenda KA, Schornagel IL, Sadeghi-Nejad A, Grand RJ. Effect of recombinant growth hormone treatment on children with Crohn's disease and short stature: A pilot study. Inflamm Bowel Dis. 2005;11(5):435-441
  48. Carel J, Ecosse E, Nicolino M, et al. Adult height after long term treatment with recombinant growth hormone for idiopathic isolated growth hormone deficiency: observational follow up study of the French population based registry. BMJ. 2002;325:70.
  49. Carr A, Samaras K, Burton S, et al. A syndrome of peripheral lipodystrophy, hyperlipidemia, and insulin resistance in patients receiving HIV protease inhibitors. AIDS. 1998;12:F51-F58.
  50. Carr A, Samaras K, Chisholm DJ, et al. Pathogenesis of HIV-1-protease inhibitor-associated peripheral lipodystrophy, hyperlipidaemia, and insulin resistance. Lancet. 1998;351(9119):1881-1883
  51. Chatelain P, Job JC, Blanchard J, et al. Dose-dependent catch-up growth after 2 years of growth hormone treatment in intrauterine growth-retarded children. J Clin Endocrinol Metab. 1994;78(6):1454-1460.
  52. Chaussain JL, Colle M, Landier F, et al. Effects of growth hormone therapy in prepubertal children with short stature secondary to intrauterine growth retardation. Acta Paediatrica Scand Suppl. 1994;399:74-76.
  53. Clemmons DR, Chihara K, Freda PU, et al. Optimizing control of acromegaly: Integrating a growth hormone receptor antagonist into the treatment algorithm. Clin Endocrinol Metab. 2003;88(10):4759-4767.
  54. Cohen M, Lahat E, Bistritzer T, et al. Evidence-based review of bone strength in children and youth with cerebral palsy. J Child Neurol. 2009;24(8):959-967.
  55. Coleman E, Adler R, Bockting W, et al. Standards of Care for the Health of Transsexual, Transgender, and Gender Nonconforming People. Version 7. Minneapolis, MN: World Professional Association for Transgender Health (WPATH); 2011.
  56. Coniglio SJ, Stevenson RD, Rogol AD. Apparent growth hormone deficiency in children with cerebral palsy. Dev Med Child Neurol. 1996;38(9):797-804.
  57. Connock M, Adi Y, Bayliss S, Moore D. The clinical effectiveness and cost-effectiveness of pegvisomant for the treatment of acromegaly: A systematic review. Midlands Health Technology Assessment Collaboration Report No. 64. Birmingham, UK: University of Birmingham, Department of Public Health and Epidemiology; 2007.
  58. Cook DM, Yuen KCJ, Biller BMK, Kemp SF, Lee Vance M. American Association of Clinical Endocrinologists. Medical guidelines for clinical practice for growth hormone use in growth hormone-deficient adults and transition patients 2009 update. Endocr  Pract. 2009;15(2):1-28.
  59. Coutant R, Carel JC, Letrait M, et al. Short stature associated with intrauterine growth retardation: Final height of untreated and growth hormone-treated children. J Clin Endocrinol Metab. 1998;83(4):1070-1074.
  60. Cozzolino M, Cecchino GN, Troiano G, Romanelli C. Growth hormone cotreatment for poor responders undergoing in vitro fertilization cycles: A systematic review and meta-analysis. Fertil Steril. 2020;114(1):97-109.
  61. Craig M, Johnson A, Cowell C. Recombinant growth hormone in Prader-Willi Syndrome (Protocol). Cochrane Database Syst Rev. 2003;(1):CD004100. 
  62. Crandall C. Combination treatment of osteoporosis: A clinical review. J Womens Health Gend Based Med. 2002;11(3):211-224.
  63. Czernichow P, Fjellestad-Paulsen A. Growth hormone in the treatment of short stature in young children with intrauterine growth retardation.Hormone Res. 1998;49 Suppl 2:23-27.
  64. Danna NR, Beutel BG, Ramme AJ, et al. The effect of growth hormone on chondral defect repair. Cartilage. 2018;9(1):63-70.
  65. Davies PS. Body composition in Prader-Willi syndrome: Assessment and effects of growth hormone administration. Acta Paediatr Suppl. 1999;88(433):105-108.
  66. de Zegher F, Maes M, Gargosky SE, et al. High-dose growth hormone treatment of short children born small for gestational age. J Clin Endocrinol Metab. 1996;81(5):1887-1892.
  67. Deal C, Hasselmann C, Pfaffle RW, et al. Associations between pituitary imaging abnormalities and clinical and biochemical phenotypes in children with congenital growth hormone deficiency: data from an international observational study. Horm Res Paediatr. 2013;79:283-292.
  68. Deal CL, Tony M, Hoybye C, et al. Growth Hormone Research Society workshop summary: Consensus guidelines for recombinant human growth hormone therapy in Prader-Willi syndrome. J Clin Endocrinol Metab. 2013;98:E1072-E1087.
  69. Donaldson MDC. Jury still out on growth hormone for normal short stature and Turner's syndrome. Lancet. 1996;348:3-4.
  70. Edouard T, Grunenwald S, Gennero I, et al. Prevalence of IGF1 deficiency in prepubertal children with isolated short stature. Eur J Endocrinol. 2009;161(1):43-50.
  71. Theratechnologies Inc. Egrifta (tesamorelin for injection) for subcutaneous use. Prescribing Information. Montreal, Québec: revised July 2018.
  72. Eiholzer U, Gisin R, Weinmann C, et al. Treatment with human growth hormone in patients with Prader-Labhart-Willi syndrome reduces body fat and increases muscle mass and physical performance. Eur J Pediatr. 1998;157(5):368-377.
  73. Eli Lilly and Company. Humatrope (somatropin) for injection, for subcutaneous use. Prescribing Information. Indianapolis, IN: Eli Lilly and Company: revised October 2019.
  74. EMD Serono Inc. Egrifta (tesamorelin for injection), for subcutaneous use. Prescribing Information. Rockland, MA: EMD Serono; revised June 2019.
  75. EMD Serono Inc. Saizen (somatropin) for injection, for subcutaneous use. Prescribing Information. Rockland, MA: Serono Inc.; revised May 2018.
  76. Evans WJ, Kotler DP, Staszewski S, et al. Effect of recombinant human growth hormone on exercise capacity in patients with HIV-associated wasting on HAART. AIDS Read. 2005;15:301-314.
  77. Falutz J, Allas S, Mamputu JC, et al. Long-term safety and effects of tesamorelin, a growth hormone-releasing factor analogue, in HIV patients with abdominal fat accumulation. AIDS. 2008 Sep 12;22(14):1719-1728.
  78. Falutz J, Mamputu JC, Potvin D, et al. Effects of tesamorelin (TH9507), a growth hormone-releasing factor analog, in human immunodeficiency virus-infected patients with excess abdominal fat: A pooled analysis of two multicenter, double-blind placebo-controlled phase 3 trials with safety extension data. J Clin Endocrinol Metab. 2010(b);95(9):4291-4304.
  79. Falutz J, Potvin D, Mamputu JC, et al. Effects of tesamorelin, a growth hormone-releasing factor, in HIV-infected patients with abdominal fat accumulation: A randomized placebo-controlled trial with a safety extension. J Acquir Immune Defic Syndr. 2010(a);53(3):311-322.
  80. Ferring Pharmaceuticals Inc. Zomacton [somatropin (rDNA origin)] for injection. Prescribing Information. Parsippany, NJ; revised July 2018.
  81. Finkelstein BS, Imperiale TF, Speroff T, et al.Effect of growth hormone therapy on height in children with idiopathic short stature: A meta-analysis. Arch Ped Adoles Med. 2002;156(3):230-240.
  82. Fjellestad-Paulsen A, Czernichow P, Brauner R, et al. Three-year data from a comparative study with recombinant human growth hormone in the treatment of short stature in young children with intrauterine growth retardation. Acta Paediatr Scand. 1998;87(5):511-517.
  83. Franklin SL, Geffner ME. Growth hormone: the expansion of available products and indications. Pediatr Clin North Am. 2011;58:1141-1165.
  84. Frasier SD. A review of growth hormone stimulation tests in children. Pediatrics. 1974;53(6):929 -937.
  85. Gabrielli O, Carloni I, Coppa GV, et al. Long-term hormone replacement therapy in two patients with Kabuki syndrome and growth hormone deficiency. Minerva Pediatr. 2000;52(1-2):47-53.
  86. Gallagher MP, Levine LS, Oberfield SE. A review of the effects of therapy on growth and bone mineralization in children with congenital adrenal hyperplasia. Growth Horm IGF Res. 2005;15 Suppl A:S26-30.
  87. Garcia JM, Biller BMK, Korbonits M, et al. Macimorelin as a diagnostic test for adult GH deficiency. J Clin Endocrinol Metab. 2018;103(8):3083-3093.
  88. Genentech, Inc.; Nutropin AQ (somatropin) injection, for subcutaneous use. South San Francisco, CA: Genentech, Inc.; revised December 2016.
  89. Gharib H, Cook DM, Saenger PH, et al. American Association of Clinical Endocrinologists medical guidelines for clinical practice for growth hormone use in adults and children--2003 update. Endocr Pract. 2003;9(1):64-76.
  90. Ghigo E, Bellone J, Aimaretti G, et al. Reliability of provocative tests to assess growth hormone secretory status. Study in 472 normally growing children. J Clin Endo Metab. 1996;81:3323-3327.
  91. Ghizzoni L, Mastorakos G, Vottero A, et al. Spontaneous cortisol and growth hormone secretion interactions in patients with nonclassic 21-hydroxylase deficiency (NCCAH) and control children. J Clin Endocrinol Metab. 1996;81(2):482-487.
  92. Glade MJ, Smith SJ. Use of recombinant human growth hormone (rhGH) in children with short stature and Noonan syndrome. Diagnostic and Therapeutic Technology Assessment (DATTA). Chicago, IL: American Medical Association; 1997.
  93. Glesby MJ. Treatment of HIV-associated lipodystrophy. UpToDate [online serial]. Waltham, MA: UpToDate; reviewed November 2013.
  94. Goldenberg DL. Treatment of fibromyalgia in adults not responsive to initial therapies. UpToDate [online serial]. Waltham, MA: UpToDate; reviewed November 2013.
  95. Goyal M, Jain M, Singhal S, Nandimath K. 18p deletion syndrome: Case report with clinical consideration and management. Contemp Clin Dent. 2017;8(4):632-636.
  96. Grimberg A, DiVall SA, Polychronakos C, et al. Guidelines for growth hormone and insulin-like growth factor-I treatment in children and adolescents: growth hormone deficiency, idiopathic short stature, and primary insulin-like growth factor-I deficiency. Horm Res Paediatr. 2016;86:361-397.
  97. Grinspoon S, Mulligan K for the Department of Health and Human Services Working Group on the Prevention and Treatment of Wasting and Weight Loss. Weight loss and wasting in patients infected with human immunodeficiency virus. Clin Infect Dis. 2003;36(Suppl 2):S69-78.
  98. Gupta KL, Shetty KR, Agre JC, et al. Human growth hormone effect on serum IGF-I and muscle function in poliomyelitis survivors. Arch Phys Med Rehabil. 1994;75(8):889-894.
  99. Hardin DS, Adams-Huet B, Brown D, et al. Growth hormone treatment improves growth and clinical status in prepubertal children with cystic fibrosis: results of a multicenter randomized controlled trial. J Clin Endocrinol Metab. 2006; 91(12):4925-9.
  100. Hardin DS, Ellis KJ, Dyson M, et al. Growth hormone improves clinical status in prepubertal children with cystic fibrosis: Results of a randomized controlled clinical trial. J Pediatr. 2001;139:636-42.
  101. Hardin DS, Rice J, Ahn C, et al. Growth hormone treatment enhances nutrition and growth in children with cystic fibrosis receiving enteral nutrition. J Pediatr. 2005;146:324-8.
  102. Hart RJ, Rombauts L, Norman RJ. Growth hormone in IVF cycles: Any hope? Curr Opin Obstet Gynecol. 2017;29(3):119-125.
  103. Health Care Insurance Board/College voor zorgverzekeringen (CVZ). Treatment with biosynthetic growth hormone in Turner syndrome- primary research. Amstelveen, The Netherlands: CVZ; 1994.
  104. Health Technology Board for Scotland (HTBS). Understanding HTBS Advice: The use of human growth hormone for children. Glasgow, Scotland, UK: Health HTBS; 2002.
  105. Hindmarsh PC, Brook CGD. Final height of short normal children treated with growth hormone. Lancet. 1996;348:13-16.
  106. Hindmarsh PC, Brook CGD. Short stature and growth hormone deficiency. Clin Endocrinol. 1995;43:133-142.
  107. Hopwood NJ, Hintz RL, Gertner JM, et al. Growth hormone response of children with non growth hormone deficiency and marked short stature during three years of growth hormone therapy. J Pediatr. 1993;123:215-222.
  108. Huiming Y, Chaomin W. Recombinant growth hormone therapy for X-linked hypophosphatemia in children. Cochrane Database Syst Rev. 2005;(1).CD004447.
  109. Isley WL. Growth hormone therapy for adults: Not ready for prime time? Ann Intern Med. 2002;137(3):190-196.
  110. Jeppesen PB, Szkudlarek J, Hoy CE, Mortensen PB. Effect of high-dose growth hormone and glutamine on body composition, urine creatinine excretion, fatty acid absorption, and essential fatty acids status in short bowel patients: A randomized, double-blind, crossover, placebo-controlled study. Scand J Gastroenterol. 2001;36(1):48-54.
  111. Jesic M, Jesic M, Sajic S, et al. Nonclassic congenital adrenal hyperplasia resulting from 21-hydroxylase deficiency. Srp Arh Celok Lek. 2004;132 Suppl 1:106-108.
  112. Job JC, Chaussain JL, Job B, et al. Follow-up of three years of treatment with growth hormone and of one post-treatment year, in children with severe growth retardation of intrauterine onset. Pediatr Res. 1996;39(2):354-359.
  113. Johannsson G, Gordon MB, Højby Rasmussen M, 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-e1376.
  114. Kanaka-Gantenbein C. Present status of the use of growth hormone in short children with bone diseases (diseases of the skeleton). J Pediatr Endocrinol Metab. 2001;14(1):17-26.
  115. Katznelson L, Laws ER, Melmed S, et al. Acromegaly: an Endocrine Society clinical practice guideline. J Clin Endocrinol Metab. 2014; 99:3933-3951.
  116. Kemp SF, Frindik JP. Emerging options in growth hormone therapy: An update. Drug Des Devel Ther. 2011;5:411-419.
  117. Khadilkar VV, Cameron FJ, Stanhope R. Growth failure and pituitary function in CHARGE and VATER associations. Arch Dis Child. 1999;80(2):167-170.
  118. Khoury C. Chiari malformations. UpToDate [online serial]. Waltham, MA: UpToDate; reviewed February 2021.
  119. Kirk J. Indications for growth hormone therapy in children. Arch Dis Child. 2012;97(1):63-68.
  120. Kohn B, Julius JR, Blethen SL. Combined use of growth hormone and gonadotropin-releasing hormone analogues: The national cooperative growth study experience. Pediatrics. 1999;104(4 Pt 2):1014-1018.
  121. Kohn DT, Kopchick JJ. Growth hormone receptor antagonists. Minerva Endocrinol. 2002;27(4):287-298.
  122. Kolibianakis EM, Venetis CA, Diedrich K, et al. Addition of growth hormone to gonadotrophins in ovarian stimulation of poor responders treated by in-vitro fertilization: A systematic review and meta-analysis. Hum Reprod Update. 2009;15(6):613-622.
  123. Kotler DP, Rosenbaum KR, Wang J, et al. Alterations in body fat distribution in HIV-infected men and women [Abstract 32173]. 12th World AIDS Conference, Geneva, Switzerland, 1998.
  124. Lafeber HN. Nutritional management and growth hormone treatment of preterm infants born small for gestational age. Acta Paediatr Scand Suppl. 1997;423:202-206.
  125. Lawson Wilkins Pediatric Endocrine Society. Guidelines for the use of growth hormone in children with short stature. A report by the Drug and Therapeutics Committee of the Lawson Wilkins Pediatric Endocrine Society. J Pediatr. 1995;127(6):857-867.
  126. Le Corvoisier P, Hittinger L, Chanson P, et al. Cardiac effects of growth hormone treatment in chronic heart failure: A meta-analysis. J Clin Endocrinol Metab. 2007;92(1):180-185.
  127. Leonart LP, Tonin FS, Ferreira VL, et al. Effectiveness and safety of pegvisomant: A systematic review and meta-analysis of observational longitudinal studies. Endocrine. 2019;63(1):18-26.
  128. Li H, Banerjee S, Dunfield L, et al. Recombinant human growth hormone for treatment of Turner Syndrome: Systematic review and economic evaluation. Technology Report. HTA No. 96. Ottawa, ON: Canadian Agency for Drugs and Technologies in Health (CADTH); 2007.
  129. Lieberman SA, Hoffman AR. Growth hormone deficiency in adults: Characteristics and response to growth hormone replacement. J Pediatrics. 1996;128(5):S58-S60.
  130. Lin D, Rieder MJ. Interventions for the treatment of decreased bone mineral density associated with HIV infection. Cochrane Database Syst Rev. 2007;(2):CD005645.
  131. Lindgren AC, Hagenas L, Muller J, et al. Effects of growth hormone treatment on growth and body composition in Prader-Willi syndrome: A preliminary report. The Swedish National Growth Hormone Advisory Group. Acta Paediatr Suppl. 1997;423:60-62.
  132. Lindgren AC, Ritzen EM. Five years of growth hormone treatment in children with Prader-Willi syndrome. Swedish National Growth Hormone Advisory Group. Acta Paediatr Suppl. 1999;88(433):109-111.
  133. Lin-Su K, Vogiatzi MG, Marshall I, et al. Treatment with growth hormone and luteinizing hormone releasing hormone analog improves final adult height in children with congenital adrenal hyperplasia. J Clin Endocrinol Metab. 2005;90:3318-3325.
  134. Liu F-T, Hu K-L, Li R, et al. Effects of growth hormone supplementation on poor ovarian responders in assisted reproductive technology: A systematic review and meta-analysis. Reprod Sci. 2021;28(4):936-948.
  135. Liu H, Bravata DM, Olkin I, et al. Systematic review: The effects of growth hormone on athletic performance. Ann Internal Med. 2008;148(10):747-758.
  136. Liu H, Bravata DM, Olkin I, et al. Systematic review: The safety and efficacy of growth hormone in the healthy elderly. Ann Intern Med. 2007;146(2):104-115.  
  137. Maison P, Chanson P. Cardiac effects of growth hormone in adults with growth hormone deficiency: A meta-analysis. Circulation. 2003;108(21):2648-2652.
  138. Manfredi R, Zucchini A, Azzaroli L, Manfredi G. Pseudopseudohypoparathyroidism associated with idiopathic growth hormone deficiency. Role of treatment with biosynthetic growth hormone. J Endocrinol Invest. 1993;16(9):709-713.
  139. Mangili A, Murman H, Zampini AM, et al. Nutrition and HIV infection: review of weight loss and wasting in the era of highly active antiretroviral therapy from the nutrition for healthy living cohort. Clin Infect Dis. 2006;42:836-42.
  140. Mantovani G, Ferrante E, Giavoli C, et al. Recombinant human GH replacement therapy in children with pseudohypoparathyroidism type Ia: First study on the effect on growth. J Clin Endocrinol Metab. 2010;95(11):5011-5017.
  141. Martinez-Moreno CG, Epardo D, Balderas-Marquez JE, et al. Regenerative effect of growth hormone (GH) in the retina after kainic acid excitotoxic damage. Int J Mol Sci. 2019;20(18).
  142. Matarese LE, Seidner DL, Steiger E. Growth hormone, glutamine, and modified diet for intestinal adaptation. J Am Diet Assoc. 2004;104(8):1265-1272.
  143. McKenna SP, Doward LC, Alonso J, et al. The QoL-AGHDA: An instrument for the assessment of quality of life in adults with growth hormone deficiency. Qual Life Res. 1999;8(4):373-383.
  144. Mekala KC, Tritos NA. Effects of recombinant human growth hormone therapy in obesity in adults: A meta analysis. J Clin Endocrinol Metab. 2009;94(1):130-137.
  145. Merke DP. Treatment of classic congenital adrenal hyperplasia due to 21-hydroxylase deficiency in infants and children. UpToDate [online serial]. Waltham, MA: UpToDate; updated October 31, 2018.
  146. Miccoli M, Bertelloni S, Massart F. Height outcome of recombinant human growth hormone treatment in achondroplasia children: A meta-analysis. Horm Res Paediatr. 2016;86(1):27-34.
  147. Miller BS, Blair JC, Rasmussen MH, et al. Weekly somapacitan is effective and well tolerated in children with GH deficiency: The randomized phase 3 REAL4 trial. J Clin Endocrinol Metab. 2022;107(12):3378-3388.
  148. Mohammad EH, El Serour AGA, Mohamed EAH, et al. Efficacy of growth hormone supplementation with ultrashort GnRH antagonist in IVF/ICSI for poor responders; randomized controlled trial. Taiwan J Obstet Gynecol. 2021;60(1):51-55.
  149. Molitch ME, Clemmons DR, Malozowski S, et al. Evaluation and treatment of adult growth hormone deficiency: an Endocrine Society clinical practice guideline. J Clin Endocrinol Metab. 2011;96:1587-1609.
  150. Monthly Prescribing Reference (MPR). Zomacton approved to treat growth hormone deficiency in adults. New York, NY: Haymarket Media; February 1, 2018. 
  151. Moore D, Meads C, Roberts L, Song F. The effectiveness and cost-effectiveness of somatostatin analogues in the treatment of acromegaly. Birmingham, UK: West Midlands Health Technology Assessment Collaboration, Department of Public Health and Epidemiology, University of Birmingham; 2002.
  152. Moorkens G, Wynants H, Abs R. Effect of growth hormone treatment in patients with chronic fatigue syndrome: A preliminary study. Growth Horm IGF Res. 1998;8 Suppl B:131-133.
  153. Morselli LL, Bongioanni P, Genovesi M, et al. Growth hormone secretion is impaired in amyotrophic lateral sclerosis. Clin Endocrinol (Oxf). 2006;65(3):385-388.
  154. Mossberg KA, Durham WJ, Zgaljardic DJ, et al. Functional changes after recombinant human growth hormone replacement in patients with chronic traumatic brain injury and abnormal growth hormone secretion. J Neurotrauma. 2017;34(4):845-852.  
  155. Mulrow CD, Ramirez G, Cornell JE, Allsup K. Defining and managing chronic fatigue syndrome. Evidence Report/Technology Assessment No. 42. Rockville, MD: Agency for Healthcare Research and Quality; 2001.
  156. Munns CF, Berry M, Vickers D, et al. Effect of 24 months of recombinant growth hormone on height and body proportions in SHOX haploinsufficiency. J Pediatr Endocrinol Metab. 2003;16(7):997-1004.
  157. Myers SE, Carrel AL, Whitman BY, et al. Physical effects of growth hormone treatment in children with Prader-Willi syndrome. Acta Paediatr Suppl. 1999;88(433):112-114.
  158. Nardo LG, El-Toukhy T, Stewart J, et al. British Fertility Society Policy and Practice Committee: Adjuvants in IVF: Evidence for good clinical practice. Hum Fertil (Camb). 2015;18(1):2-15.
  159. National Comprehensive Cancer Network (NCCN). Testicular cancer. NCCN Clinical Practice Guidelines in Oncology, Version 1.2016. Fort Washington, PA: NCCN; 2016.
  160. National Health Service, National Institute for Clinical Excellence (NICE). Guidance on use of human growth hormone (somatropin) in children with growth failure. Technology Appraisal Guidance No. 42. London, UK: NICE; May 2002.
  161. National Heart, Lung, and Blood Institute. Obesity Education Initiative: The practical guide: identification, evaluation, and treatment of overweight and obesity in adults. Bethesda, MD: US Dept of Health and Human Services, National Heart, Lung, and Blood Institute; 2000. NIH Publication No. 00-4084.
  162. National Horizon Scanning Centre (NHSC). Pegvisomant for acromegaly - horizon scanning review. Birmingham, UK: NHSC; 2002.
  163. National Institute for Clinical Excellence (NICE). Appraisal consultation document: Human growth hormone (somatropin) in adults with growth hormone deficiency. London, UK: NICE; May 2003.
  164. National Institute for Clinical Excellence (NICE). Human growth hormone (somatropin) in adults with growth hormone deficiency. Technology Appraisal 64. London, UK: NICE; August 2003.
  165. National Institute for Clinical Excellence (NICE). Guidance on the use of human growth hormone (somatropin) for the treatment of growth failure in children. London, UK: NICE; May 2010. 
  166. National Institute for Clinical Excellence: Human growth hormone (somatropin) in adults with growth hormone deficiency. August 2003.
  167. National Institutes of Health (NIH), National Heart, Lung and Blood Institute. Third Report of the Expert Panel on Detection, Evaluation, and Treatment of High Blood Cholesterol in Adults (Adult Treatment Panel III). National Cholesterol Education Program. Bethesda, MD: NIH; 2001.
  168. National Institutes of Health (NIH), National Library of Medicine (NLM). Growth hormone stimulation test. NIH/NLM: MedlinePlus [online]. Bethesda, MD: NIH; updated April 2019.
  169. National Institutes of Health (NIH). Osteoporosis prevention, diagnosis, and therapy. NIH Consens Statement. 2000;17(1):1-45.
  170. Nemechek PM, Polsky B, Gottlieb MS. Treatment Guidelines for HIV-Associated Wasting. Mayo Clin Proc. 2000;75:386-394.
  171. Ngim CF, Lai NM, Hong JY, et al. Growth hormone therapy for people with thalassaemia. Cochrane Database Syst Rev. 2020;5(5):CD012284. 
  172. NHS Centre for Reviews and Dissemination. The effectiveness of interventions used in the treatment/management of chronic fatigue syndrome and/or myalgic encephalomyelitis in adults and children. York, UK: Centre for Reviews and Dissemination; 2002.
  173. Nieman LK. Diagnosis and treatment of nonclassic (late-onset) congenital adrenal hyperplasia due to 21-hydroxylase deficiency. UpToDate [online serial]. Waltham, MA: UpToDate; reviewed December 2012.
  174. Novo Nordisk. FDA approves once-weekly Sogroya for the treatment of children living with growth hormone deficiency. Press Release. Plainsboro, NJ: Novo Nordisk. April 28, 2023a.
  175. Novo Nordisk A/S. Trial to compare the efficacy and safety of NNC0195-0092 (somapacitan) with placebo and Norditropin Flex Pro (somatropin) in adults with growth hormone deficiency. (REAL 1). ClinicalTrials.gov Identifier: NCT02229851. Bethesda, MD: National Library of Medicine; updated November 23, 2020.
  176. Novo Nordisk Inc. Norditropin (somatropin) injection, for subcutaneous use. Prescribing Information. Plainsboro, NY: Novo Nordisk: revised February 2018.
  177. Novo Nordisk, Inc. Sogroya (somapacitan-beco) injection, for subcutaneous use. Prescribing Information. Plainsboro, NJ: Novo Nordisk, Inc; April 2023b.
  178. Nylander E, Grönbladh A, Zelleroth S, et al. Growth hormone is protective against acute methadone-induced toxicity by modulating the NMDA receptor complex. Neuroscience. 2016;339:538-547.
  179. Ogata T, Onigata K, Hotsubo T, et al. Growth hormone and gonadotropin-releasing hormone analog therapy in haploinsufficiency of SHOX. Endocr J. 2001;48(3):317-322.
  180. Oh JH, Chung SW, Oh KS, et al. Effect of recombinant human growth hormone on rotator cuff healing after arthroscopic repair: Preliminary result of a multicenter, prospective, randomized, open-label blinded end point clinical exploratory trial. J Shoulder Elbow Surg. 2018;27(5):777-785.
  181. Oh WK. Overview of the treatment of testicular germ cell tumors. UpToDate [online serial]. Waltham, MA: UpToDate; reviewed December 2015.
  182. Oregon Health and Science University (OHSU), Department of Pathology, Director of Clinical Laboratory Services. OHSU Laboratory Services Manual. Portland, OR: OHSU; 2003. Available at: http://www.ohsu.edu/pathology/wardman/frame.htm. Accessed August 26, 2003.
  183. Paisley AN, Trainer P, Drake W. Pegvisomant: A novel pharmacotherapy for the treatment of acromegaly. Expert Opin Biol Ther. 2004;4(3):421-425.
  184. Parekh NR, Steiger E. Criteria for the use of recombinant human growth hormone in short bowel syndrome. Nutrition Clin Prac. 2005;20:503-508.
  185. Parkinson C, Scarlett JA, Trainer PJ. Pegvisomant in the treatment of acromegaly. Adv Drug Deliv Rev. 2003;55(10):1303-1314.
  186. Pfizer Inc. Genotropin (somatropin) fro injection, for subcutaneous use. Prescribing Information. New York, NY: Pfizer; revised April 2019.
  187. Pharmacia & Upjohn Co. Genotropin somatropin (rDNA origin) for injection. Prescribing Information. Kalamazoo, MI: Pharmacia & Upjohn; December 2016.
  188. Pharmacia & Upjohn. Genotropin somatropin (rDNA origin) for injection. Prescribing Information. 818 279 003. Kalamazoo, MI: Pharmacia; April 2003.
  189. Pharmacia & Upjohn, Division of Pfizer Inc,. Genotropin somatropin for injection. Prescribing Information. New York, NY: Pharmacia & Upjohn, Division of Pfizer Inc.; revised April 2019.
  190. Pharmacia & Upjohn, Division of Pfizer Inc,. Somavert (pegvisomant) for injection, for subcutaneous use. New York, NY: Pharmacia & Upjohn, Division of Pfizer Inc; revised August 2021.
  191. Phung OJ, Coleman CI, Baker EL, et al. Effectiveness of recombinant human growth hormone (rhGH) in the treatment of patients with cystic fibrosis. Comparative effectiveness review No. 23. (Prepared by the University of Connecticut/Hartford Evidence-based Practice Center under Contract No. 290-2007-10067-I) AHRQ Publication No. 11-EHC003. Rockville, MD: Agency for Healthcare Research and Quality. October 2010.
  192. Pichon Riviere A, Augustovski F, Cernadas C, et al. Analysis of the different available formulations of human growth hormone. Buenos Aires, Argentina: Institute for Clinical Effectiveness and Health Policy (IECS); 2003.
  193. Pichon-Riviere A, Augustovski F, Garcia Marti S, et al. Growth hormone in short children born small for gestational age [summary]. IRR No. 162. Buenos Aires, Argentina: Institute for Clinical Effectiveness and Health Policy (IECS); January 2009.
  194. Poidvin A, Touze E, Ecosse E, et al. Growth hormone treatment for childhood short stature and risk of stroke in early adulthood. Neurology. 2014;83(9):780-786.
  195. Polsky B, Kotler D, Steinhart C. HIV-associated wasting in the HAART era: guidelines for assessment, diagnosis, and treatment. AIDS Patient Care STDS. 2001;15(8):411-23.
  196. Pucarelli I, Segni M, Ortore M, et al. Effects of combined gonadotropin-releasing hormone agonist and growth hormone therapy on adult height in precocious puberty: A further contribution. J Pediatr Endocrinol Metab. 2003;16(7):1005-1010.
  197. Quintos JB, Hodax JK, Gonzales-Ellis BA, et al. Efficacy of growth hormone therapy in Kearns-Sayre syndrome: The KIGS experience. J Pediatr Endocrinol Metab. 2016;29(11):1319-1324.
  198. Quintos JB, Vogiatzi MG, Harbison MD, et al. Growth hormone therapy alone or in combination with gonadotropin-releasing hormone analog therapy to improve the height deficit in children with congenital adrenal hyperplasia. J Clin Endocrinol Metab. 2001;86(4):1511-1517.
  199. Ranke MB, Lindberg A. Growth hormone treatment of short children born small for gestational age or with Silver-Russell syndrome: Results from KIGS (Kabi International Growth Study), including the first report on final height. Acta Paediatr Suppl. 1996;417:18-26.
  200. Rapaport R. Growth hormone treatment in short children born SGA - FDA approved and recommended. Clinical Controversies: Growth Hormone for Short Children Born SGA. American Academy of Pediatrics Section on Endocrinology Newsletter. 2002;10:3-4. Available at: http://www.aap.org/sections/endocrinology/. Accessed August 26, 2003.
  201. Resnik R. Fetal growth restriction: Evaluation and management. UpToDate [online serial]. Waltham, MA: UpToDate; reviewed November 2013.
  202. Richmond EJ, Rogol AD. Diagnosis of growth hormone deficiency in children. UpToDate [online serial]. Waltham, MA: UpToDate; reviewed February 2021.
  203. Root AW. Growth hormone should not be administered routinely to children with short stature due to intrauterine growth retardation who are not classically growth hormone deficient. Clinical Controversies: Growth Hormone for Short Children Born SGA. American Academy of Pediatrics Section on Endocrinology Newsletter 2002; 10:3-4. 
  204. Rose SR, Minicchi G, Barnes KM, et al. Overnight growth hormone concentrations are usually normal in pubertal children with idiopathic short stature - A clinical research center study. J Clin Endo Metab. 1996;81:1063-1068.
  205. Rosenberg M. Overview of the management of chronic kidney disease in adults. UpToDate [online serial]. Waltham, MA: UpToDate; reviewed November 2018.
  206. Rosenfeld RG, Albertsson-Wilkand K, Frasier SD. Diagnostic controversy: The diagnosis of childhood growth hormone deficiency revisited. J Clin Endo Metab. 1995;80:1532-1540.
  207. Ross MG. Fetal growth restriction. eMedicine Obstetrics and Gynecology. New York, NY: Medscape; updated March 8, 2013.
  208. Rothenbuhler A, Linglart A, Piquard C, Bougneres P. A pilot study of discontinuous, insulin-like growth factor 1-dosing growth hormone treatment in young children with FGFR3 N540K-mutated hypochondroplasia. J Pediatr. 2012;160(5):849-853.
  209. Rubin MR, Bilezikian JP. New anabolic therapies in osteoporosis. Curr Opin Rheumatol. 2002;14(4):433-440.
  210. Saenger P. Growth hormone in growth hormone deficiency. BMJ. 2002;325(7355):58-59.
  211. Sandoz Inc. Omnitrope (somatropin) injection, for subcutaneous use. Prescribing Information. Princeton, NJ: Sandoz Inc.; revised June 2019.
  212. Sas T, de Waal W, Mulder P, et al. Growth hormone treatment in children with short stature born small for gestational age: 5-year results of a randomized, double-blind, dose-response trial. J Clin Endocrinol Metab. 1999;84(9):3064-3070.
  213. Savastano S, Di Somma C, Angrisani L, et al. Growth hormone treatment prevents loss of lean mass after bariatric surgery in morbidly obese patients: Results of a pilot, open, prospective, randomized, controlled study. J Clin Endocrinol Metab. 2009;94(3):817-826.
  214. Say L, Gülmezoglu AM, Hofmeyr GJ. Hormones for suspected impaired fetal growth. Cochrane Database Syst Rev. 2003;(1):CD000109.
  215. Schober E, Scheibenreiter S, Frisch H. 18p monosomy with GH-deficiency and empty sella: Good response to GH-treatment. Clin Genet. 1995;47(5):254-256.
  216. Schott DA, Stumpel CTRM, Klaassens M. Hypermobility in individuals with Kabuki syndrome: The effect of growth hormone treatment. Am J Med Genet A. 2019;179(2):219-223.
  217. Scolapio JS. Effect of growth hormone, glutamine, and diet on body composition in short bowel syndrome: A randomized, controlled study. JPEN J Parenter Enteral Nutr. 1999;23(6):309-313.
  218. Seguy D, Vahedi K, Kapel N, et al. Low-dose growth hormone in adult home parenteral nutrition-dependent short bowel syndrome patients: A positive study. Gastroenterology. 2003;124(2):293-302.
  219. Serono, Inc. Zorbtive (somatropin (rDNA orgin) for injection. Prescribing Information. N884O101A. Rockland, MA: Serono; May 2017.
  220. Shadid S, Jensen MD. Effects of growth hormone administration in human obesity. Obesity Res. 2003;11(2):170-175.
  221. Shang Y, Wu M, He R, et al. Administration of growth hormone improves endometrial function in women undergoing in vitro fertilization: A systematic review and meta-analysis. Hum Reprod Update. 2022;28(6):838-857.
  222. Sheridan KJ. Osteoporosis in adults with cerebral palsy. Dev Med Child Neurol. 2009;51 Suppl 4:38-51.
  223. Shetty KR, Gupta KL, Agre JC, et al.  Effect of human growth hormone on muscle function in post-polio syndrome. Ann N Y Acad Sci. 1995;753:386-389.
  224. Shim ML, Moshang T Jr, Oppenheim WL, Cohen P. Is treatment with growth hormone effective in children with cerebral palsy? Dev Med Child Neurol. 2004;46(8):569-571.
  225. Silverstein JH, Shulman D. Growth hormone for small-for-gestational-age children: Short and sweet? J Pediatr. 2003;142(2):91-92.
  226. Simionescu L, Jubelt B. Post-polio syndrome. UpToDate [online serial]. Waltham, MA: UpToDate; reviewed December 2015.
  227. Sivakumar T, Mechanic O, Fehmie DA, Paul B. Growth hormone axis treatments for HIV-associated lipodystrophy: A systematic review of placebo-controlled trials. HIV Med. 2011;12(8):453-462.
  228. Smith RA, Melmed S, Sherman B, et al. Recombinant growth hormone treatment of amyotrophic lateral sclerosis. Muscle Nerve. 1993;16(6):624-633.
  229. Smith S, Remmington T. Recombinant growth hormone therapy for X-linked hypophosphatemia in children. Cochrane Database Syst Rev. 2021;10(10):CD004447. 
  230. Smith WT, Nam TJ, Underwood LE, et al. Use of insulin like growth factor binding protein-2, IGFBP-3 and IGF-1 for assessment growth hormone status in short children. J Clin Endocrin Metab. 1993;77:1294-1299.
  231. Society for Endocrinology. Adult growth hormone (GH) replacement: The use of somatotrophin (GH) in the management of adult somatotrophin deficiency. Topical Briefing. Bristol, UK: Society for Endocrinology; revised July 22, 1999. 
  232. Sood A, Mohiyiddeen G, Ahmad G, et al. Growth hormone for in vitro fertilisation (IVF). Cochrane Database Syst Rev. 2021;11(11):CD000099. 
  233. Speiser PW, Arlt W, Auchus RJ, et al. Congenital adrenal hyperplasia due to steroid 21-hydroxylase deficiency: An Endocrine Society Clinical Practice Guideline. J Clin Endocrinol Metab. 2018;103(11):4043-4088.
  234. Speiser PW, Azziz R, Baskin LS, et al; Endocrine Society. Congenital adrenal hyperplasia due to steroid 21-hydroxylase deficiency: An Endocrine Society clinical practice guideline. J Clin Endocrinol Metab 2010;95(9):4133-4160.
  235. Srivastava T, Warady BA. Overview of the management of chronic kidney disease in children. UpToDate [online serial]. Waltham, MA: UpToDate; reviewed November 2018.
  236. Stanhope R, Ackland F, Hamill G, et al. Physiological growth hormone secretion and response to growth hormone treatment in children with short stature and intrauterine growth retardation. Acta Paediatr Scand Suppl. 1989;349:47-52.
  237. Stanhope R, Preece MA, Hamill G. Does growth hormone treatment improve final height attainment of children with intrauterine growth retardation? Arch Dis Child. 1991;66:1180-1183.
  238. State of Minnesota, Health Technology Advisory Committee (HTAC). The use of human growth hormone for children with idiopathic short stature. Technology Assessment. St. Paul, MN: HTAC; February 2000.
  239. Steuerman R, Shevah O, Laron Z. Congenital IGF1 deficiency tends to confer protection against post-natal development of malignancies. Eur J Endocrinol. 2011;164(4):485-489.
  240. Stewart PM. Pegvisomant: An advance in clinical efficacy in acromegaly. Eur J Endocrinol. 2003;148 Suppl 2:S27-S32.
  241. Sun H, Wan N, Wang X, e tal. Genotype-phenotype analysis, neuropsychological assessment, and growth hormone response in a patient with 18p deletion syndrome. Cytogenet Genome Res. 2018;154(2):71-78.
  242. Swedish Council on Technology Assessment in Health Care (SBU). Growth hormone in children with idiopathic short stature - early assessment briefs (Alert). Stockholm, Sweden: SBU; 2002.
  243. Synder PJ. Growth hormone deficiency in adults. UpToDate [online serial]. Waltham, MA: UpToDate; reviewed April 2019.
  244. Szarka N, Szellar D, Kiss S, et al. Effect of growth hormone on neuropsychological outcomes and quality of life of patients with traumatic brain injury: A systematic review. J Neurotrauma. 2021;38(11):1467-1483.
  245. Szkudlarek J, Jeppesen PB, Mortensen PB. Effect of high dose growth hormone with glutamine and no change in diet on intestinal absorption in short bowel patients: A randomised, double blind, crossover, placebo controlled study. Gut. 2000;47(2):199-205.
  246. Tanner JM, Davies PSW. Clinical longitudinal standards for height and height velocity for North American children. J Pediatr. 1985;107;317-329.
  247. Thaker V, Carter B, Putman M. Recombinant growth hormone therapy for cystic fibrosis in children and young adults. Cochrane Database Syst Rev. 2018;12:CD008901.
  248. Thaker V, Haagensen AL, Carter B, et al. Recombinant growth hormone therapy for cystic fibrosis in children and young adults. Cochrane Database Syst Rev. 2013;(6):CD008901.
  249. Thaker V, Haagensen AL, Carter B, et al. Recombinant growth hormone therapy for cystic fibrosis in children and young adults. Cochrane Database Syst Rev. 2015;(5):CD008901.
  250. Torlinska-Walkowiak N, Majewska KA, Kędzia A, Opydo-Szymaczek J. Clinical implications of growth hormone deficiency for oral health in children: A systematic review. J Clin Med. 2021;10(16):3733.
  251. Torres R, Unger KW. Treatment of dorsocervical fat pads (Buffalo hump) and truncal adiposity with Serostim (recombinant human growth hormone) in patients with AIDS maintained on HAART [Abstract 32164]. 12th World AIDS Conference, Geneva, Switzerland, 1998.
  252. Tran GT, Pagkalos J, Tsiridis E, et al. Growth hormone: Does it have a therapeutic role in fracture healing? Expert Opin Investig Drugs. 2009;18(7):887-911.
  253. Tritos NA, Biller BMK. Current concepts of the diagnosis of adult growth hormone deficiency. Rev Endocr Metab Disord. 2021;22(1):109-116.
  254. Tritos NA, Danias PG. Growth hormone therapy in congestive heart failure due to left ventricular systolic dysfunction: A meta-analysis. Endocr Pract. 2008;14(1):40-49.
  255. Trojan DA, Collet J, Pollak MN, et al. Serum insulin-like growth factor-I (IGF-I) does not correlate positively with isometric strength, fatigue, and quality of life in post-polio syndrome. J Neurol Sci. 2001;182(2):107-115.
  256. Turleau C. Monosomy 18p. Orphanet J Rare Dis. 2008;3:4.
  257. Tuvemo T, Gustafsson J, Proos LA. Growth hormone treatment during suppression of early puberty in adopted girls. Swedish Growth Hormone Advisory Group. Acta Paediatr. 1999;88(9):928-932.
  258. U.S. Food and Drug Administration (FDA). Approval of Egrifta (tesamorelin) to treat lipodystrophy. For Consumers. Rockville, MD: FDA; November 10, 2010.
  259. van der Lely AJ. Justified and unjustified use of growth hormone. Postgrad Med J. 2004;80(948):577-580.
  260. Van Pareren Y, Mulder P, Houdijk M, et al. Adult height after long-term, continuous growth hormone (GH) treatment in short children born small for gestational age: Results of a randomized, double-blind, dose-response GH trial. J Clin Endocrinol Metab. 2003;88(8):3584-3590.
  261. van Toledo-Eppinga L, Houdijk EC, Cranendonk A, et al. Effects of recombinant human growth hormone treatment in intrauterine growth-retarded preterm newborn infant on growth, body composition and energy expenditure. Acta Paediatr Scand. 1996;85(4):476-481.
  262. van Toledo-Eppinga L, Houdijk MC, Delemarre-Van De Waal HA, et al. Leucine and glucose kinetics during growth hormone treatment in intrauterine growth-retarded preterm infants. Am J Physiol. 1996;270 (3 Pt 1):E451-E455.
  263. Vance ML, Mauras N. Drug therapy: Growth hormone therapy in adults and children. N Engl J Med. 1999;341(16):1206-1216.
  264. Verma N, Kaur A, Sharma R, et al. Outcomes after multiple courses of granulocyte-colony stimulating factor and growth hormone in decompensated cirrhosis: Randomized trial. Hepatology. 2018;68(4):1559-1573.
  265. Vimalachandra D, Hodson EM, Willis NS, et al. Growth hormone for children with chronic kidney disease. Cochrane Database Syst Rev. 2006;(3):CD003264.
  266. Wakeling EL, Brioude F, Lokulo-Sodipe O, et al. Diagnosis and management of Silver-Russell syndrome: First international consensus statement. Nat Rev Endocrinol. 2017;13(2):105-124.
  267. Wales P, Nasr A, De Silva N, et al. Human growth hormone and glutamine for patients with short bowel syndrome (Protocol for Cochrane Review). Cochrane Database Syst Rev. 2007;(1):CD006321.
  268. Wallymahmed ME, Baker GA, Humphris G, et al. The development, reliability and validity of a disease specific quality of life model for adults with growth hormone deficiency. Clin Endocrinol (Oxf). 1996;44(4):403-411.
  269. Walvoord EC, Pescovitz OH. Combined use of growth hormone and gonadotropin-releasing hormone analogues in precocious puberty: Theoretic and practical considerations. Pediatrics. 1999;104(4 Pt 2):1010-1014.
  270. Wheeler P, Balk E, Cole C; Tufts-New England Medical Center EPC. Criteria for determining disability in infants and children: Short stature. AHRQ Evidence Report No. 73. Rockville, MD: Agency for Healthcare Research and Quality (AHRQ); 2003.
  271. Wheeler PG, Bresnahan K, Shephard BA, et al. Short stature and functional impairment: A systematic review. Arch Pediatr Adolesc Med. 2004;158(3):236-243.
  272. Wilson TA, Rose SR, Cohen P, et al. Update of Guidelines for the Use of Growth Hormone in Children: The Lawson Wilkins Pediatric Endocrinology Society Drug and Therapeutics Committee. J Pediatr. 2003;143:415-421.
  273. Wit JM, Balen HV, Kamp GA, Oostdijk W. Benefit of postponing normal puberty for improving final height. Eur J Endocrinol. 2004;151 Suppl 1:S41-S45.
  274. Wolters Kluwer. Somatropin injection (drug facts and comparisons). Facts & Comparisons [online serial]. Updated February 8, 2021. Available at online.lexi.com. Accessed March 10, 2021.
  275. Woods DL, Greenfield DH, Louw HH, et al. Assessing gestational age and size at birth. Unit 17. Newborn Care Manual. International Association for Maternal and Neonatal Health (IAMNH). Geneva, Switzerland: Geneva Foundation for Medical Education and Research; January 2005.
  276. Wu GH, Wu ZH, Wu ZG. Effects of bowel rehabilitation and combined trophic therapy on intestinal adaptation in short bowel patients. World J Gastroenterol. 2003;9(11):2601-2604.
  277. Xu J, Casserly E, Yin Y, Cheng J. A systematic review of growth hormone in pain medicine: From rodents to humans. Pain Med. 2020;21(1):21-31.
  278. Xu YM, Hao GM, Gao BL. Application of growth hormone in in vitro fertilization. Front Endocrinol (Lausanne). 2019;10:502.
  279. Yang A, Kim J, Cho SY, et al. A case of de novo 18p deletion syndrome with panhypopituitarism. Ann Pediatr Endocrinol Metab. 2019;24(1):60-63.
  280. Yang HM, Mao M, Wan CM, Yang F. Growth hormone treatment for short stature in children with intrauterine growth retardation (Protocol for Cochrane Review). Cochrane Database Syst Rev. 2009;(3):CD006293.
  281. Yang P, Wu R, Zhang H, et al. The effect of growth hormone supplementation in poor ovarian responders undergoing IVF or ICSI: A meta-analysis of randomized controlled trials. Reprod Biol Endocrinol. 2020;18(1):76.
  282. Yuen KCJ, Biller BMK, Radovick S, et al. American Association of Clinical Endocrinologists and American College of Endocrinology Guidelines for management of growth hormone deficiency in adults and patients transitioning from pediatric to adult care. Endocr Pract. 2019;5:1191-1232.