Skip to main content

MINI REVIEW article

Front. Endocrinol., 09 June 2022
Sec. Pediatric Endocrinology
This article is part of the Research Topic Controversies in Growth Hormone Treatment and Diagnosis View all 17 articles

Growth Hormone Stimulation Testing: To Test or Not to Test? That Is One of the Questions

  • Division of Pediatric Endocrine and Diabetes, Mount Sinai Kravis Children’s Hospital, New York, NY, United States

The evaluation of children with short stature includes monitoring over a prolonged period to establish a growth pattern as well as the exclusion of chronic medical conditions that affect growth. After a period of monitoring, evaluation, and screening, growth hormone stimulation testing is considered when the diagnosis of growth hormone deficiency (GHD) is entertained. Though flawed, growth hormone stimulation tests remain part of the comprehensive evaluation of growth and are essential for the diagnosis of growth hormone (GH) deficiency. Variables including testing length, growth hormone assay and diagnostic cut off affect results. Beyond the intrinsic issues of testing, results of GH stimulation testing can be influenced by patient characteristics. Various factors including age, gender, puberty, nutritional status and body weight modulate the secretion of GH.

Introduction

Concern about poor growth is the leading reason for referrals to a pediatric endocrinologist (1). The evaluation of children with short stature includes monitoring over a prolonged period to establish a growth pattern as well as the exclusion of chronic medical conditions that affect growth. After a period of monitoring, evaluation, and screening, growth hormone stimulation testing is considered when the diagnosis of growth hormone deficiency (GHD) is entertained.

Lack of puberty at an expected age is another common reason patients are referred. Typically, 95% of girls present at least one sign of puberty by 13 years of age and boys by 14 years of age. These conditions of growth and pubertal delay can be difficult to diagnose because the rate of growth appears to decelerate as they cross growth percentiles around the time of the anticipated pubertal growth spurt. Thus, the growth deceleration due to constitutional delay of puberty and growth hormone deficiency can be difficult to ascertain with certainty (1).

Measuring GH concentrations at random times is unreliable because growth hormone (GH) is secreted from the anterior pituitary in a pulsatile fashion and is mainly stimulated by the release of hypothalamic growth hormone-releasing hormone (GHRH). Instead, patients are usually screened for GH deficiency by measuring serum IGF-1 and IGF-BP3 levels which have longer half-life and no pulsatility. Though flawed, growth hormone stimulation tests remain part of the comprehensive evaluation of growth and are essential for the diagnosis of growth hormone (GH) deficiency (24). For GH stimulation testing, two agents provoke GH secretion from the pituitary (L-dopa, clonidine, arginine, glucagon). These provocative agents are not physiological and do not replicate normal secretory dynamics. The insulin tolerance test which is considered the gold standard for diagnosis of GHD is used to assess GH secretion in response to hypoglycemia. Given the risks associated with hypoglycemia, it is performed less frequently in the outpatient setting. Serial blood samples are taken to detect the point of maximal serum concentration of GH (1). Peak growth hormone (PGH) response to provocative testing is a vital determinant of the clinical response to GH therapy. In a study by Cohen et al. of prepubertal children characterized as GH deficient or idiopathic short stature based on GH stimulation testing without sex hormone priming, those with idiopathic short stature required doses nearly 2 times higher to reach an IGF-1 target of 2 SDS (5). The stimulated PGH level response to two pharmacologic stimuli that distinguishes between GH deficient and sufficient patients is unclear and likely exists on a continuum with levels of 5, 7 and 10 ng/mL having been proposed, each without adequate data for substantiation. The currently agreed upon peak GH cutoff is 10 ng/mL (1, 6, 7). Stimulation testing with arginine and levodopa with samples obtained for 3 hours is perhaps most frequently used in many large multicenter studies (3, 6, 8, 9).

Intrinsic Test Factors

Test Length

Some studies of various GH ST protocols suggest that sampling or duration can be reduced from 3 hours, whilst preserving the accuracy of diagnosis (1013). Data from our single center experience of provocative GH testing using an identical protocol on a large cohort of 315 pediatric patients with short stature and/or growth failure showed peak GH response was reached by 2 hours in 97.8% of those tested. This study indicated that the GH ST with arginine and levodopa can be terminated at 2 hours without compromising its diagnostic value based on the currently accepted peak growth hormone response cutoff of 10 ng/mL, as exclusion of the 3 hour sample did not alter the GH sufficiency status in any of the 315 patients (9).

Growth Hormone Assay

There are several practical and logistical limitations to stimulation testing. Results are often often vary depending on which assay is used to analyze blood samples. Historically, GH was measured by a wide variety of approaches including bioassays, radio receptor assays, immunoassays and mass spectrometry (14). Currently, immunoassays are used most commonly to measure serum GH concentrations in clinical settings. Endogenous GH in serum exists in numerous isoforms with the majority being the isoform of 22 kDa molecular weight. However, approximately 10% circulates as the 20 kDa isoform and other isoforms and growth hormone fragments circulate in smaller portions (15). Different immunoassays can detect different spectrums of total GH isoforms. In an effort to standardize across isoforms, current consensus guidelines recommend assay calibration with a highly purified preparation of the 22 kDa recombinant human GH isoform of GH (2, 16). Additionally, GH immunoassays transitioned from using polyclonal antibodies that targeted multiple epitopes on varying GH isoforms to monoclonal antibodies targeting one isoform (14). With these changes, current assays have a narrower target.

On the new assays, GH concentrations yields are lower than on older assays. Cutoffs for peak growth hormone response to GH stimulation testing may need to be revisited with the adoption of newer assays with lower reported GH concentrations. Since only small changes in isoform ratios have been reported in certain states such has pituitary tumors and exercise, concentrations of 22 kDa GH accurately reflect total GH secretion (14). Still, inter-assay differences between immunoassays occur due to differences variations in the type of immunoassay, antibody specificity, and interference from GH binding proteins (14).

Peak Growth Hormone Cut Off

As recombinant growth hormone became more widely available, less stringent criteria for the diagnosis of GHD were implemented with increase in peak GH cut off levels. With the renewed interest in oral GH secretagogues, a reassessment of peak GH cut offs may be helpful. The studies by Bright et al. and Blum et al. suggest that a partially intact pituitary axis is needed for GH secretagogues to be effective (17, 18). Individuals with “moderate” growth hormone deficiency who may respond to GH secretagogues need to be differentiated from those with “severe” growth hormone deficiency who require growth hormone therapy (9).

Traditionally, the interpretation of GH stimulation testing results was binary with the adherence to pass/fail diagnostic GH cutoffs. Perhaps, instead, the results should be interpreted on a continuum that spans severe GHD requiring GH therapy to moderate or provisional GHD for which alternative therapies and further monitoring of growth should be considered (19). There is increasing evidence supporting the need to revisit cutoffs for peak GH after stimulation based on the assay used to measure serum GH concentrations (20, 21). Lower cutoffs for peak GH levels based on specific assays have been proposed. The establishment of method-specific clinical evidence-based GH cutoff limits would help ensure adequate clinical diagnoses.

Supporting MRI Findings

In GHD, brain MRI may show congenital pituitary abnormalities such as anterior pituitary dysplasia/hypoplasia, pituitary stalk interruption syndrome, and developmental cyst but also tumoral lesions (22). Neuroimaging is a crucial study in the diagnostic process of GHD. With only partial integrity of the hypothalamic pituitary connections, growth hormone secretion was able to be stimulated by growth hormone releasing hormone plus Arginine (23). In children with congenital GHD but less severe impairment of the pituitary stalk, the GH response to stimulation may be sufficient but pituitary GH reserve deteriorates with a GH response of < 10 ug/L after 20 yr of age (23). MRI may be helpful in differentiating those with moderate or provisional GHD. Findings of pituitary abnormalities support decisions on GH treatment in such cases of moderate GHD (peak GH of 7-10 ng/ml), as GHD is expected to evolve.

Patient Factors

Beyond the intrinsic issues of testing, results of GH stimulation testing can be influenced by patient characteristics. GH secretion is influenced by several factors including age, gender, puberty, nutritional status and body weight (2426).

BMI

Obesity has been associated with decreased spontaneous and stimulated GH secretion in both adults (2730), and children (31) and weight reduction has been followed by increased GH secretion (3135). Though the exact neuroendocrine mechanism causing the blunted GH response in obesity is unknown, proposed mechanisms include high circulating levels of insulin which can suppress GH synthesis and release and adipocyte-secreted leptin affecting GH regulation (3639). It has been demonstrated that PGH response to stimulation testing with A-LD decreased with higher BMI SDS in a large cohort of normal weight healthy children with a range of BMI that approximated a normal distribution (mean BMI SDS of -0.3 ± 1.0). This finding suggests that the inverse relationship between BMI and PGH is not isolated to obesity and is evident in the normal weight children (9). Still, BMI is not currently consistently considered in the interpretation of the peak GH response in children.

Puberty and Sex Hormones

During puberty, there is a normal increase in growth hormone concentrations due to a larger mass of GH released per pituitary secretory episode resulting from a higher maximal rate of GH secretion per secretory burst (40). Due to the physiologic rise of GH during puberty, there is a debate as to whether prepubertal children should be “primed” with sex hormones before GH provocative testing (1). In addition to endogenous sex hormones, short term administration of exogenous sex hormones can modulate growth hormone secretion (41). Priming leads to increased peak GH levels and decreases the false positive rate for diagnosing GH deficiency in healthy controls (4244). In the study by Marin et. al, a subset of 11 prepubertal normal children were primed with 2 days of estrogen. Peak GH response rose to levels seen in subjects at pubertal stages 4 and 5 (45). In a later study by Muller et al. of 26 boys primed with a single dose of testosterone, 77% increased their peak GH level to > 10 ng/ml (46). In a study of 315 patients undergoing GH stimulation testing, there was no difference in rates of GHD in prepubertal and pubertal patients (9).

In 2016, the Pediatric Endocrine Society updated their guidelines to support priming with sex hormones in prepubertal children (boys > 11 years old and girls > 10 years old) (2). The stated reason for this recommendation of priming was to avoid unnecessary GH treatment of children with constitutional delay of growth and puberty (2). Yet, the practice of priming remains controversial in Europe (47). In a study of 8 European countries and the US performed after these guidelines were published, priming was recommended in 5 out of 9 countries (48).

This hormonal milieu of puberty is not sustained after priming. On these supraphysiologic testosterone levels, endogenous growth hormone secretion may be overestimated. Will these children who responded to exogenous sex hormones be able to secrete enough GH at the time of puberty? Would peripubertal children who have lower peak GH levels without priming benefit from exogenous GH therapy? This overestimation can lead to false negative results and deny eligible children required treatment with growth hormone. It is unclear whether children diagnosed with GHD with or without priming respond differently to GH treatment. However, constitutional delay and GHD can be difficult to differentiate and priming should be considered in delayed puberty (49). Short term adverse side effects of priapism and testicular pain were reported in approximately 3% of prepubertal boys primed with short courses of testosterone (50).

Altering a patient’s baseline characteristics is not recommended with any other stimulation testing to diagnose a hormonal deficiency. As an alternative to priming, normative values of peak growth hormone response should be further explored to develop cut off limits based on pubertal stage. This was first proposed by Rose et al. when they found that mean spontaneous night time growth hormone levels rose during pubertal development in both boys and girls, with the highest levels at mid-puberty (51). Currently, distinct cut offs are only defined for children and adults. Cut offs based on pubertal staging would bridge the continuum. Reassessment of the GH/IGF-I axis when a child treated with growth hormone peripubertally enters puberty has been proposed as another alternative to priming (19). Though it is common in practice to continue growth hormone therapy once diagnosed with GHD until the epiphyses close, GH therapy could be paused at onset of puberty to repeat the GH stimulation test and determine if continued therapy is necessary. We recommend obtaining pubertal hormone levels at time of GH stimulation testing to correlate GH response to pubertal status. In our clinical experience, we have cared for patients with low peak GH response to stimulation without priming in whom we elected not to treat that later demonstrated adequate growth velocity and adult height. This demonstrates the importance of the clinician’s interpretation of clinical findings in combination with stimulation testing results.

Discussion

Given its flaws, one should enter GH ST with a high predictive value. The Pediatric Endocrine Society recommends against the use of GH stimulation testing as the sole diagnostic criterion of GHD (2). The decision to proceed with growth hormone (GH ST) stimulation testing should be reached only after careful consideration and only when the result will significantly contribute to the diagnostic process (9). If one combines stimulation testing result with the patient’s anthropometric measurements, height velocity, physical findings, screening tests, and IGF-1 and IGF-BP3 levels, more complete clinical picture is capture that allows for proper individualized diagnosis and treatment (1). While by itself growth hormone stimulation testing is unreliable, within the overall picture of a patient with short stature, decreased growth velocity, and low IGF-1 level, the results of growth hormone stimulation testing may complete a picture that the astute clinician can properly utilize to decide on interventions such as growth hormone therapy.

In conclusion we recommend careful, long term observation of patients with growth failure. The decision to undertake growth hormone stimulation should be reserved for those in whom its results would be the last and deciding parameter for therapeutic intervention. If still unclear, additional observation and evaluations such as genetic testing and perhaps repeat stimulation testing should be considered.

Author Contributions

MY contributed with substantial contributions to the conception or design of the work; or the acquisition, analysis or interpretation of data for the work, and drafting the work or revising it critically for important intellectual content. RR contributed by providing approval for publication of the content and analysis or interpretation of data for the work, and drafting the work or revising it critically for important intellectual content. All authors contributed to the article and approved the submitted version.

Conflict of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s Note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

1. Graber E, Rapaport R. Growth and Growth Disorders in Children and Adolescents. Pediatr Ann (2012) 41(4):e1–9. doi: 10.3928/00904481-20120307-07

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Grimberg A, DiVall SA, Polychronakos C, Allen DB, Cohen LE, Quintos JB, 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(6):361–97. doi: 10.1159/000452150

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Cohen P, Rogol AD, Deal CL, Saenger P, Reiter EO, Ross JL, et al. Consensus Statement on the Diagnosis and Treatment of Children With Idiopathic Short Stature: A Summary of the Growth Hormone Research Society, the Lawson Wilkins Pediatric Endocrine Society, and the European Society for Paediatric Endocrinology Workshop. J Clin Endocrinol Metab (2008) 93(11):4210–7. doi: 10.1210/jc.2008-0509

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Chesover AD, Dattani MT. Evaluation of Growth Hormone Stimulation Testing in Children. Clin Endocrinol (Oxf) (2016) 84(5):708–14. doi: 10.1111/cen.13035

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Cohen P, Germak J, Rogol AD, Weng W, Kappelgaard AM, Rosenfeld RG, et al. Variable Degree of Growth Hormone (GH) and Insulin-Like Growth Factor (IGF) Sensitivity in Children With Idiopathic Short Stature Compared With GH-Deficient Patients: Evidence From an IGF-Based Dosing Study of Short Children. J Clin Endocrinol Metab (2010) 95(5):2089–98. doi: 10.1210/jc.2009-2139

PubMed Abstract | CrossRef Full Text | Google Scholar

6. van Vught AJ, Nieuwenhuizen AG, Gerver WJ, Veldhorst MA, Brummer RJ, Westerterp-Plantenga MS, et al. Pharmacological and Physiological Growth Hormone Stimulation Tests to Predict Successful GH Therapy in Children. J Pediatr Endocrinol Metab (2009) 22(8):679–94. doi: 10.1515/JPEM.2009.22.8.679

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Gandrud LM, Wilson DM. Is Growth Hormone Stimulation Testing in Children Still Appropriate? Growth Horm IGF Res (2004) 14(3):185–94. doi: 10.1016/j.ghir.2003.11.003

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Cohen P. Statement 1: A Serum Insulin-Like Growth Factor I (IGF-I) Level Should be Part of the Evaluation of Children With Short Stature. Pediatr Endocrinol Rev (2008) 5 Suppl 3:834–6.

PubMed Abstract | Google Scholar

9. Yau M, Chacko E, Regelmann MO, Annunziato R, Wallach EJ, Chia D, et al. Peak Growth Hormone Response to Combined Stimulation Test in 315 Children and Correlations With Metabolic Parameters. Horm Res Paediatr (2019) 92(1):36–44. doi: 10.1159/000502308

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Muster L, Zangen DH, Nesher R, Hirsch HJ, Muster Z, Gillis D, et al. Arginine and Clonidine Stimulation Tests for Growth Hormone Deficiency Revisited–do We Really Need So Many Samples? J Pediatr Endocrinol Metab (2009) 22(3):215–23. doi: 10.1515/JPEM.2009.22.3.215

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Jaruratanasirikul S, Leethanaporn K, Sriplung H. Should the Duration of the Insulin Tolerance Test be Shortened to 90 Minutes? J Pediatr Endocrinol Metab (2004) 17(8):1105–9. doi: 10.1515/JPEM.2004.17.8.1105

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Galluzzi F, Stagi S, Parpagnoli M, Losi S, Pagnini I, Favelli F, et al. Oral Clonidine Provocative Test in the Diagnosis of Growth Hormone Deficiency in Childhood: Should We Make the Timing Uniform? Horm Res (2006) 66(6):285–8. doi: 10.1159/000095781

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Strich D, Terespolsky N, Gillis D. Glucagon Stimulation Test for Childhood Growth Hormone Deficiency: Timing of the Peak Is Important. J Pediatr (2009) 154(3):415–9. doi: 10.1016/j.jpeds.2008.08.044

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Ribeiro de Oliveira Longo Schweizer J, Ribeiro-Oliveira A Jr, Bidlingmaier M. Growth Hormone: Isoforms, Clinical Aspects and Assays Interference. Clin Diabetes Endocrinol (2018) 4:18. doi: 10.1186/s40842-018-0068-1

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Baumann GP. Growth Hormone Isoforms. Growth Horm IGF Res (2009) 19(4):333–40. doi: 10.1016/j.ghir.2009.04.011

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Clemmons DR. Consensus Statement on the Standardization and Evaluation of Growth Hormone and Insulin-Like Growth Factor Assays. Clin Chem (2011) 57(4):555–9. doi: 10.1373/clinchem.2010.150631

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Bright GM, Do MT, McKew JC, Blum WF, Thorner MO. Development of a Predictive Enrichment Marker for the Oral GH Secretagogue LUM201 in Pediatric Growth Hormone Deficiency. J Endocrine Soc (2021) 5(6):bvab030. doi: 10.1210/jendso/bvab030

CrossRef Full Text | Google Scholar

18. Blum WF, Bright GM, Do MT, McKew JC, Chen H, Thorner MO, et al. Corroboration of Height Velocity Prediction Markers for rhGH With an Oral GH Secretagogue Treatment in Children With GHD. J Endocrine Soc (2021) 5(6):bvab029. doi: 10.1210/jendso/bvab029

CrossRef Full Text | Google Scholar

19. Allen DB. The Diagnosis of Growth Hormone Deficiency Remains a Judgment Call - and That Is Good. Horm Res Paediatr (2021) 94:406–9. doi: 10.1159/000521628

PubMed Abstract | CrossRef Full Text | Google Scholar

20. Wagner IV, et al. Clinical Evidence-Based Cutoff Limits for GH Stimulation Tests in Children With a Backup of Results With Reference to Mass Spectrometry. Eur J Endocrinol (2014) 171(3):389–97.

PubMed Abstract | Google Scholar

21. Lotierzo M, et al. Comparative Study of Human Growth Hormone Measurements: Impact on Clinical Interpretation. Clin Chem Lab Med (2022) 60(2):191–7.

PubMed Abstract | Google Scholar

22. Xu C, Zhang X, Dong L, Zhu B, Xin T. MRI Features of Growth Hormone Deficiency in Children With Short Stature Caused by Pituitary Lesions. Exp Ther Med (2017) 13(6):3474–8. doi: 10.3892/etm.2017.4377

PubMed Abstract | CrossRef Full Text | Google Scholar

23. Maghnie M, Salati B, Bianchi S, Rallo M, Tinelli C, Autelli M, et al. Relationship Between the Morphological Evaluation of the Pituitary and the Growth Hormone (GH) Response to GH-Releasing Hormone Plus Arginine in Children and Adults With Congenital Hypopituitarism. J Clin Endocrinol Metab (2001) 86(4):1574–9. doi: 10.1210/jcem.86.4.7394

PubMed Abstract | CrossRef Full Text | Google Scholar

24. Lee HS, Hwang JS. Influence of Body Mass Index on Growth Hormone Responses to Classic Provocative Tests in Children With Short Stature. Neuroendocrinology (2011) 93(4):259–64. doi: 10.1159/000326838

PubMed Abstract | CrossRef Full Text | Google Scholar

25. Ho KY, Evans WS, Blizzard RM, Veldhuis JD, Merriam GR, Samojlik E, et al. Effects of Sex and Age on the 24-Hour Profile of Growth Hormone Secretion in Man: Importance of Endogenous Estradiol Concentrations. J Clin Endocrinol Metab (1987) 64(1):51–8. doi: 10.1210/jcem-64-1-51

PubMed Abstract | CrossRef Full Text | Google Scholar

26. Iranmanesh A, Lizarralde G, Veldhuis JD. Age and Relative Adiposity Are Specific Negative Determinants of the Frequency and Amplitude of Growth Hormone (GH) Secretory Bursts and the Half-Life of Endogenous GH in Healthy Men. J Clin Endocrinol Metab (1991) 73(5):1081–8. doi: 10.1210/jcem-73-5-1081

PubMed Abstract | CrossRef Full Text | Google Scholar

27. Makimura H, Feldpausch MN, Rope AM, Hemphill LC, Torriani M, Lee H, et al. Metabolic Effects of a Growth Hormone-Releasing Factor in Obese Subjects With Reduced Growth Hormone Secretion: A Randomized Controlled Trial. J Clin Endocrinol Metab (2012) 97(12):4769–79. doi: 10.1210/jc.2012-2794

PubMed Abstract | CrossRef Full Text | Google Scholar

28. Radack JA, White PC, Adams-Huet B, Oden JD. Stimulated Growth Hormone Concentrations in Obese Pediatric Patients With Mild and Severe Insulin Resistance: A Pilot Study. J Pediatr Endocrinol Metab (2010) 23(4):355–61. doi: 10.1515/jpem.2010.056

PubMed Abstract | CrossRef Full Text | Google Scholar

29. Qu XD, Gaw Gonzalo IT, Al Sayed MY, Cohan P, Christenson PD, Swerdloff RS, et al. Influence of Body Mass Index and Gender on Growth Hormone (GH) Responses to GH-Releasing Hormone Plus Arginine and Insulin Tolerance Tests. J Clin Endocrinol Metab (2005) 90(3):1563–9. doi: 10.1210/jc.2004-1450

PubMed Abstract | CrossRef Full Text | Google Scholar

30. Stanley TL, Feldpausch MN, Murphy CA, Grinspoon SK, Makimura H. Discordance of IGF-1 and GH Stimulation Testing for Altered GH Secretion in Obesity. Growth Horm IGF Res (2014) 24(1):10–5. doi: 10.1016/j.ghir.2013.11.001

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Loche S, Guzzetti C, Pilia S, Ibba A, Civolani P, Porcu M, et al. Effect of Body Mass Index on the Growth Hormone Response to Clonidine Stimulation Testing in Children With Short Stature. Clin Endocrinol (Oxf) (2011) 74(6):726–31. doi: 10.1111/j.1365-2265.2011.03988.x

PubMed Abstract | CrossRef Full Text | Google Scholar

32. Rasmussen MH, Hvidberg A, Juul A, Main KM, Gotfredsen A, Skakkebaek NE, et al. Massive Weight Loss Restores 24-Hour Growth Hormone Release Profiles and Serum Insulin-Like Growth Factor-I Levels in Obese Subjects. J Clin Endocrinol Metab (1995) 80(4):1407–15. doi: 10.1210/jcem.80.4.7536210

PubMed Abstract | CrossRef Full Text | Google Scholar

33. Argente J, Caballo N, Barrios V, Muñoz MT, Pozo J, Chowen JA, et al. Multiple Endocrine Abnormalities of the Growth Hormone and Insulin-Like Growth Factor Axis in Patients With Anorexia Nervosa: Effect of Short- and Long-Term Weight Recuperation. J Clin Endocrinol Metab (1997) 82(7):2084–92. doi: 10.1210/jcem.82.7.4090

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Williams T, Berelowitz M, Joffe SN, Thorner MO, Rivier J, Vale W, et al. Impaired Growth Hormone Responses to Growth Hormone-Releasing Factor in Obesity. A Pituitary Defect Reversed With Weight Reduction. N Engl J Med (1984) 311(22):1403–7. doi: 10.1056/NEJM198411293112203

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Tanaka K, Inoue S, Numata K, Okazaki H, Nakamura S, Takamura Y, et al. Very-Low-Calorie Diet-Induced Weight Reduction Reverses Impaired Growth Hormone Secretion Response to Growth Hormone-Releasing Hormone, Arginine, and L-Dopa in Obesity. Metabolism (1990) 39(9):892–6. doi: 10.1016/0026-0495(90)90296-O

PubMed Abstract | CrossRef Full Text | Google Scholar

36. Luque RM, Kineman RD. Impact of Obesity on the Growth Hormone Axis: Evidence for a Direct Inhibitory Effect of Hyperinsulinemia on Pituitary Function. Endocrinology (2006) 147(6):2754–63. doi: 10.1210/en.2005-1549

PubMed Abstract | CrossRef Full Text | Google Scholar

37. Lee EJ, Kim KR, Lee HC, Cho JH, Nam MS, Nam SY, et al. Acipimox Potentiates Growth Hormone Response to Growth Hormone-Releasing Hormone by Decreasing Serum Free Fatty Acid Levels in Hyperthyroidism. Metabolism (1995) 44(11):1509–12. doi: 10.1016/0026-0495(95)90154-X

PubMed Abstract | CrossRef Full Text | Google Scholar

38. Kok P, Buijs MM, Kok SW, Van Ierssel IH, Frölich M, Roelfsema F, et al. Acipimox Enhances Spontaneous Growth Hormone Secretion in Obese Women. Am J Physiol Regul Integr Comp Physiol (2004) 286(4):R693–8. doi: 10.1152/ajpregu.00595.2003

PubMed Abstract | CrossRef Full Text | Google Scholar

39. Coutant R, Lahlou N, Bouvattier C, Bougnères P. Circulating Leptin Level and Growth Hormone Response to Stimulation Tests in Obese and Normal Children. Eur J Endocrinol (1998) 139(6):591–7. doi: 10.1530/eje.0.1390591

PubMed Abstract | CrossRef Full Text | Google Scholar

40. Martha PM Jr, Gorman KM, Blizzard RM, Rogol AD, Veldhuis JD. Endogenous Growth Hormone Secretion and Clearance Rates in Normal Boys, as Determined by Deconvolution Analysis: Relationship to Age, Pubertal Status, and Body Mass. J Clin Endocrinol Metab (1992) 74(2):336–44. doi: 10.1210/jcem.74.2.1730812

PubMed Abstract | CrossRef Full Text | Google Scholar

41. Leung KC, Johannsson G, Leong GM, Ho KKY. Estrogen Regulation of Growth Hormone Action. Endocr Rev (2004) 25(5):693–721. doi: 10.1210/er.2003-0035

PubMed Abstract | CrossRef Full Text | Google Scholar

42. Growth Hormone Research S. Consensus Guidelines for the Diagnosis and Treatment of Growth Hormone (GH) Deficiency in Childhood and Adolescence: Summary Statement of the GH Research Society. GH Research Society. J Clin Endocrinol Metab (2000) 85(11):3990–3. doi: 10.1210/jcem.85.11.6984

PubMed Abstract | CrossRef Full Text | Google Scholar

43. Murray PG, Dattani MT, Clayton PE. Controversies in the Diagnosis and Management of Growth Hormone Deficiency in Childhood and Adolescence. Arch Dis Child (2016) 101(1):96–100. doi: 10.1136/archdischild-2014-307228

PubMed Abstract | CrossRef Full Text | Google Scholar

44. Sato T, Kusakawa M, Ichihashi Y, Ishii T, Hasegawa T. Testosterone Priming Increased Growth Hormone Peak Levels in the Stimulation Test and Suppressed Gonadotropin Secretion in Three Japanese Adolescent Boys. Clin Pediatr Endocrinol (2020) 29(3):119–21. doi: 10.1297/cpe.29.119

PubMed Abstract | CrossRef Full Text | Google Scholar

45. Marin G, Domené HM, Barnes KM, Blackwell BJ, Cassorla FG, Cutler GB Jr, et al. The Effects of Estrogen Priming and Puberty on the Growth Hormone Response to Standardized Treadmill Exercise and Arginine-Insulin in Normal Girls and Boys. J Clin Endocrinol Metab (1994) 79(2):537–41. doi: 10.1210/jcem.79.2.8045974

PubMed Abstract | CrossRef Full Text | Google Scholar

46. Müller G, Keller A, Reich A, Hoepffner W, Kratzsch J, Buckler JM, et al. Priming With Testosterone Enhances Stimulated Growth Hormone Secretion in Boys With Delayed Puberty. J Pediatr Endocrinol Metab (2004) 17(1):77–83. doi: 10.1515/JPEM.2004.17.1.77

PubMed Abstract | CrossRef Full Text | Google Scholar

47. Hage C, Gan HW, Ibba A, Patti G, Dattani M, Loche S, et al. Advances in Differential Diagnosis and Management of Growth Hormone Deficiency in Children. Nat Rev Endocrinol (2021) 17(10):608–24. doi: 10.1038/s41574-021-00539-5

PubMed Abstract | CrossRef Full Text | Google Scholar

48. Binder G, Reinehr T, Ibáñez L, Thiele S, Linglart A, Woelfle J, et al. GHD Diagnostics in Europe and the US: An Audit of National Guidelines and Practice. Horm Res Paediatr (2019) 92(3):150–6. doi: 10.1159/000503783

PubMed Abstract | CrossRef Full Text | Google Scholar

49. Lazar L, Phillip M. Is Sex Hormone Priming in Peripubertal Children Prior to Growth Hormone Stimulation Tests Still Appropriate? Horm Res Paediatr (2010) 73(4):299–302. doi: 10.1159/000284396

PubMed Abstract | CrossRef Full Text | Google Scholar

50. Albrecht A, Penger T, Marx M, Hirsch K, Dörr HG. Short-Term Adverse Effects of Testosterone Used for Priming in Prepubertal Boys Before Growth Hormone Stimulation Test. J Pediatr Endocrinol Metab (2018) 31(1):21–4. doi: 10.1515/jpem-2017-0280

PubMed Abstract | CrossRef Full Text | Google Scholar

51. Rose SR, Municchi G, Barnes KM, Kamp GA, Uriarte MM, Ross JL, et al. Spontaneous Growth Hormone Secretion Increases During Puberty in Normal Girls and Boys. J Clin Endocrinol Metab (1991) 73(2):428–35. doi: 10.1210/jcem-73-2-428

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: sex hormone priming, IGF – I, short stature, growth hormone deficiency, growth hormone – secretion

Citation: Yau M and Rapaport R (2022) Growth Hormone Stimulation Testing: To Test or Not to Test? That Is One of the Questions. Front. Endocrinol. 13:902364. doi: 10.3389/fendo.2022.902364

Received: 23 March 2022; Accepted: 04 May 2022;
Published: 09 June 2022.

Edited by:

Valentino Cherubini, Azienda Ospedaliero Universitaria Ospedali Riuniti, Italy

Reviewed by:

Sandro Loche, Ospedale Microcitemico, Italy

Copyright © 2022 Yau and Rapaport. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Mabel Yau, mabel.yau@mssm.edu

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.