Warning: mkdir(): Permission denied in /home/virtual/lib/view_data.php on line 86 Warning: fopen(/home/virtual/pfmjournal/journal/upload/ip_log/ip_log_2024-04.txt): failed to open stream: No such file or directory in /home/virtual/lib/view_data.php on line 88 Warning: fwrite() expects parameter 1 to be resource, boolean given in /home/virtual/lib/view_data.php on line 89 Prader-Willi syndrome and growth hormone therapy: exploring the precise management of hypothalamic short stature: A review

Prader-Willi syndrome and growth hormone therapy: exploring the precise management of hypothalamic short stature: A review

Article information

Precis Future Med. 2023;7(3):107-116
Publication date (electronic) : 2023 September 25
doi : https://doi.org/10.23838/pfm.2023.00079
Department of Pediatrics, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea
Corresponding author: Aram Yang Department of Pediatrics, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, 29 Saemunan-ro, Jongno-gu, Seoul 03181, Korea Tel: +82-2-2001-1980 E-mail: aram.yang@samsung.com
Received 2023 August 30; Revised 2023 September 5; Accepted 2023 September 7.

Abstract

Prader-Willi syndrome (PWS) is a rare genetic disorder characterized by various clinical features linked to hypothalamic/pituitary gland abnormalities. Growth hormone deficiency is a prominent feature of PWS that results in poor linear growth and delayed development. This review discusses the evaluation and effects of growth hormone therapy (GHT) in PWS. Heterogeneity in growth hormone secretion patterns based on genotype and the potential for personalized GHT were explored. The benefits of GHT, including improvements in motor and cognitive development, growth, and body composition, are discussed in detail. Safety considerations for GHT initiation and response to GHT in adults with PWS are discussed, along with ongoing debates regarding the efficacy and safety. Although controversies persist, an evolving understanding of the long-term effects and safety of GHT underscores the need for comprehensive research in this field.

INTRODUCTION

Prader-Willi syndrome (PWS) is a rare genetic disorder caused by a lack of paternally expressed genes on chromosome 15q11-q13. Approximately 65% to 75% of PWS cases are caused by deletion of the paternal chromosome in that region, whereas maternal disomy 15 accounts for 20% to 30% of cases, and genomic imprinting defects cause 1% to 3% of cases. PWS presents a unique constellation of features, including hypotonia in infancy, hyperphagia, early onset morbid obesity, intellectual disabilities, hypogonadism, and behavioral abnormalities [1-4]. These symptoms appear to be associated with hypothalamic/pituitary gland abnormalities, and manifestations based on deficient pituitary hormones include the following [5-7]. (1) Growth hormone deficiency (GHD): insufficient production of growth hormone (GH) leads to poor linear growth and delayed physical development; (2) central hypothyroidism: reduced production of thyroid-stimulating hormone affects thyroid hormone release, leading to symptoms similar to those of primary hypothyroidism; (3) central adrenal insufficiency: adrenal hormone deficiencies can cause weakness, fatigue, low blood pressure, and a decreased stress response; (4) central hypogonadism: impaired production of gonadotropin-releasing hormone production leads to delayed sexual development and infertility; and (5) central diabetes insipidus: insufficient antidiuretic hormone production causes excessive thirst and frequent urination.

Early diagnosis and appropriate treatment are crucial for managing hormonal imbalances and improving the overall health and well-being of affected individuals. Among these symptoms, hypothalamic short stature with a lack of a pubertal growth spurt is particularly significant in PWS because of a deficiency of both GH and insulin-like growth factor 1 (IGF-1) [3,8,9].

In this review, short stature in PWS was explored, along with an exploration of ways to determine the effects, side effects, and duration of growth hormone therapy (GHT).

GROWTH HORMONE DEFICIENCY AND PRADER-WILLI SYNDROME

GHD is well recognized in PWS. Previously, GH stimulation tests were conducted to evaluate GHD in PWS. However, given the inherent nature of reduced GH secretion in the syndrome, once PWS is diagnosed, GHT is typically indicated after a certain age. Currently, in Korea, GHT for patients with PWS is covered by insurance, starting from the age of 2 years. Furthermore, most experts agree on initiating GHT before the onset of obesity, often setting the target age at < 2 years of age [10,11]. Although there may be slight variations across different studies, spontaneous impairment of GH secretion and low IGF-1 levels have been observed in nearly all patients with PWS. Approximately 80% of patients show a significant reduction in GHD levels, compared to the general pediatric population, through stimulation tests [11-15]. However, the frequency of GHD varies from 40% to 100% [12,15]. This discrepancy can be attributed to factors such as the type of stimulation test, non-standardized methods, country-specific diagnostic cut-offs, and the influence of stimulation test outcomes based on obesity [13,16-19]. In addition, it is important to consider the diverse normal ranges of GH levels according to age. Therefore, in PWS, comprehensive combined pituitary function and GH stimulation tests for hypothalamic-pituitary deficiency are essential. However, GHD may not align with the diagnostic cut-offs and could yield inconclusive results in PWS. Even in such cases, owing to the inherent nature of the disorder, GH supplementation should take precedence [11]. Indications and guidelines for GHT in PWS patients include comparisons between domestic, U.S. Food and Drug Administration, and GH Research Society recommendations (see Table 1 for details).

Indications for growth hormone injection therapy in Prader-Willi syndrome

THE ADVANTAGES OF GROWTH HORMONE THERAPY IN PRADER-WILLI SYNDROME

The advantages of GHT in PWS are as follows and have been demonstrated in numerous observational studies and RCTs [20-26]. (1) Enhanced motor development: a meta-analysis of five randomized trials involving 154 infants and children with PWS (aged 1.3 to 3 years) found that those receiving recombinant human growth hormone (rhGH) treatment demonstrated higher scores on standardized motor development tests (standard mean difference, 0.71; 95% confidence interval [CI], 0.38 to 1.03) [25]. One representative trial, focusing on infants and toddlers (29 participants aged 6 months to 3 years), revealed that rhGH therapy led to improved motor development during the initial year of treatment [27]. Standardized motor development scores on the Bayley Scales of Infant Development (BSID) increased by an average of 11.2% in the rhGH group, while the control group showed a decrease of 18.5%. (2) Enhanced cognitive development: in the same trial involving infants and toddlers, rhGH therapy also demonstrated improvements in cognitive development during the first year of treatment [26]. Standardized BSID scores for mental development increased by an average of 9.3% in the rhGH group, compared to a 2.9% decrease in the control group. However, in the earlier meta-analysis, which included six trials with 165 infants, children, and adolescents with PWS, cognitive scores were found to be similar between rhGH-treated children and untreated controls (standard mean difference, 0.2; 95% CI, –0.1 to 0.5) [25]. Notably, two of the trials in this analysis involved older children (average ages, 7 and 17 years, respectively), which may explain the lack of cognitive benefits in the overall analysis. (3) Improved growth and body composition: previous studies consistently show that rhGH therapy enhances linear growth and positively impacts body composition and obesity, regardless of whether treatment starts in infancy or later in childhood. A meta-analysis of nine randomized trials involving 328 patients demonstrated improvements in height and body mass index (BMI) in the rhGH group compared to the control group (mean difference in Z-score for height, 1.67 [95% CI, 1.54 to 1.81]; mean difference in Z-score for BMI, –0.67 [95% CI, –0.87 to –0.47]) [24]. Long-term data are needed to determine if rhGH therapy can increase adult height and potentially improve lipid profiles [28].

Observational studies and clinical trials have consistently reported similar benefits of rhGH therapy in infants and young children [22,23,29,30]. In a randomized trial involving 22 participants aged 5 to 32 months, the combination of rhGH and physical therapy increased muscle thickness, which correlated with improved muscle strength and motor performance compared to physical therapy alone in a control group [22,23]. Another trial (29 participants aged 4 to 37 months) showed that rhGH treatment resulted in improved mobility skill acquisition and body composition [30]. Starting rhGH treatment before 18 months of age has been linked to accelerated development of mobility skills, in line with expert recommendations to commence rhGH therapy at the time of PWS diagnosis, ideally within the first year of life [31].

The most substantial response to rhGH therapy in PWS children typically occurs within the initial 12 months of treatment [32]. However, sustained enhancements in height, bone density, and body composition have been observed with extended rhGH treatment, spanning up to 5 years [33]. Nevertheless, it is essential to emphasize that, despite extended treatment, full normalization of body composition may not be attainable. The chronological summary of various studies demonstrating the effects of rhGH treatment in pediatric Prader-Willi syndrome can be found in Table 2 [24,26,30,33-48].

Previous literature on the effects of growth hormone treatment in pediatric Prader-Willi syndrome

PRECAUTIONS FOR GROWTH HORMONE THERAPY IN PRADER-WILLI SYNDROME

Although GH replacement therapy has demonstrated general tolerability and has been extensively documented over the last 20 years, fatal events within the initial months of GH treatment have been reported in young patients with PWS. Such events are often associated with respiratory infections [49]. Despite the apparent improbability of a direct causal link between GHT and these fatalities, further investigations are warranted [11]. To ensure the safety of early initiation of GHT, thorough pretreatment assessments, including sleep studies, evaluations of the ear-nose-throat region, and thyroid function tests, are necessary. Regular polysomnography and continuous monitoring of the ear, nose, and throat area during therapy are recommended [50]. Adenoidectomy or tonsillectomy should be considered for issues related to lymphoid tissue. In addition, weight gain commences after the age of 2 years; therefore, strict monitoring for diabetes and blood glucose regulation is essential during GHT. Lifestyle adjustments are particularly necessary in cases of rapid weight gain [51]. GH treatment is not recommended for individuals with severe obesity, uncontrolled diabetes, or pre-existing severe scoliosis. Vigilant monitoring is essential in cases where treatment may exacerbate these risk conditions, alongside a comprehensive understanding of these potential risks [52,53].

RESPONSE TO GROWTH HORMONE THERAPY ACCORDING TO GENOTYPE

Differences exist in various clinical aspects between chromosome 15 deletion and maternal uniparental disomy (UPD) genotypes, which are genetic types representative of PWS [1,9]. These differences are also evident in the GH secretory pattern in response to GH stimulation. Regarding the GH secretory pattern, adults with PWS with del15q11-q13 exhibited higher growth hormone-releasing hormone (GHRH)+arginine-induced GH peaks than those with UPD15 [54,55]. Similar findings have been observed in pediatric age groups through various stimulation tests, such as clonidine, arginine, and insulin tolerance tests, which showed higher GH peaks in microdeletion cases than in UPD cases [13,56]. Furthermore, patients with the UPD15 genotype had a higher incidence of GHD than those with the del15q11-q13 genotype (80% vs. 25%). In line with these trends, both children and adults with the UPD genotype demonstrated better responses to GH stimulation than those with deletions [55].

However, it is unclear whether the growth-promoting and metabolic effects of GHT vary according to genotype. As there are differences in GH secretion and response to stimulation tests based on genotype in PWS, the response to and effects of GHT may also vary. However, the number of studies on this topic is limited. In a study comparing the effects of GH injection therapy in 65 Korean patients with PWS (21 UPD and 44 deletion types), no significant differences were found between the two genotype groups [57]. However, some studies have suggested that patients with specific genetic mutations may exhibit varying responses to GHT [8,13,54,56]. Therefore, further extensive research and investigation in this regard is imperative. In the future, GHT may evolve into a more personalized precision medicine tailored to individuals based on their genetic profiles.

GROWTH HORMONE TREATMENT FOR ADULTS WITH PRADER-WILLI SYNDROME

GH treatment for adults with PWS offers potential benefits, such as improved body composition, muscle strength, metabolic health, cognition, and quality of life [34,58]. However, safety concerns have been raised. Some studies have raised doubts regarding the efficacy and safety of long-term GHT in adult patients with PWS [11]. This debate centers on whether the gains in body composition from GHT are substantial enough to outweigh potential long-term uncertainties [59]. Critics argue that individuals with PWS are relatively protected from adverse metabolic effects and that intensive nutritional interventions can maintain healthy levels even without GH. GHT does not significantly reduce obesity-related issues [60-62]. Concerns have also arisen from elevated IGF-1 levels due to GHT, potentially raising cancer risk. Researchers have proposed that dietary and exercise interventions are more cost-effective for adult patients with PWS. Proponents of GHT assert that the natural persistence of hypothalamic dysfunction in PWS justifies continuous GH supplementation for consistent improvement in body composition, physical performance, and psychosocial function. They argued that an increase in IGF-1 levels is a biological effect without clear long-term consequences. The efficacy and safety of GHT in adult patients with PWS remain controversial, with research suggesting its benefits in various areas. Further studies are needed for a comprehensive understanding of the long-term effects and safety, necessitating patience to obtain conclusive results. The findings from the last 3 years are presented in Table 3 [63-65].

Recent studies on the effects of growth hormone treatment in adult Prader-Willi syndrome

INNOVATIVE APPROACHES AND RESEARCH DIRECTIONS

Ongoing research on PWS explores innovative treatment modalities beyond GHT. Interventions targeting appetite regulation, metabolic pathways, and behavioral aspects are being investigated to complement GHT and enhance the overall management of PWS. Collaborative efforts among clinicians, researchers, and geneticists are pivotal in driving these innovative approaches and expanding our understanding of the pathophysiology of PWS.

CONCLUSION

PWS is a complex disorder characterized by hypothalamic/pituitary abnormalities that lead to GHD, among other clinical manifestations. GH injection therapy has demonstrated beneficial effects on motor and cognitive development, growth, and body composition in individuals with PWS. However, the efficacy and safety of GHT in adults remains debatable. While some researchers have expressed doubts about its long-term benefits, others have emphasized the potential improvements in body composition and psychosocial function. A consensus on the benefits and risks of GHT in adults with PWS requires extensive long-term research. Future studies should address the divergence of opinions and provide a comprehensive understanding of the impact of GHT on this unique population. This evolving research landscape is expected to shed light on the optimal use of GHT and its potential contribution to the well-being of individuals with PWS.

Notes

No potential conflict of interest relevant to this article was reported.

AUTHOR CONTRIBUTIONS

Conception or design: AY.

Acquisition, analysis, or interpretation of data: AY.

Drafting the work or revising: AY.

Final approval of the manuscript: AY.

References

1. Cassidy SB, Schwartz S, Miller JL, Driscoll DJ. Prader-Willi syndrome. Genet Med 2012;14:10–26.
2. Cheon CK. Genetics of Prader-Willi syndrome and Prader-Will-Like syndrome. Ann Pediatr Endocrinol Metab 2016;21:126–35.
3. Driscoll DJ, Miller JL, Cassidy SB. Prader-Willi syndrome. In: Adam MP, Mirzaa GM, Pagon RA, Wallace SE, Bean LJH, Gripp KW, et al., editors. GeneReviews. University of Washington, Seattle; 2023 [cited 2023 Sep 6]. Available from: https://www.ncbi.nlm.nih.gov/books/NBK1330.
4. Sohn YB. Genetic obesity: an update with emerging therapeutic approaches. Ann Pediatr Endocrinol Metab 2022;27:169–75.
5. Kim SJ, Cho SY, Jin DK. Prader-Willi syndrome: an update on obesity and endocrine problems. Ann Pediatr Endocrinol Metab 2021;26:227–36.
6. Emerick JE, Vogt KS. Endocrine manifestations and management of Prader-Willi syndrome. Int J Pediatr Endocrinol 2013;2013:14.
7. Miller JL, Goldstone AP, Couch JA, Shuster J, He G, Driscoll DJ, et al. Pituitary abnormalities in Prader-Willi syndrome and early onset morbid obesity. Am J Med Genet A 2008;146A:570–7.
8. Grugni G, Crino A, Pagani S, Meazza C, Buzi F, De Toni T, et al. Growth hormone secretory pattern in non-obese children and adolescents with Prader-Willi syndrome. J Pediatr Endocrinol Metab 2011;24:477–81.
9. Goldstone AP, Holland AJ, Hauffa BP, Hokken-Koelega AC, Tauber M, ; speakers contributors at the Second Expert Meeting of the Comprehensive Care of Patients with PWS. Recommendations for the diagnosis and management of Prader-Willi syndrome. J Clin Endocrinol Metab 2008;93:4183–97.
10. Corripio R, Tubau C, Calvo L, Brun C, Capdevila N, Larramona H, et al. Safety and effectiveness of growth hormone therapy in infants with Prader-Willi syndrome younger than 2 years: a prospective study. J Pediatr Endocrinol Metab 2019;32:879–84.
11. Deal CL, Tony M, Hoybye C, Allen DB, Tauber M, Christiansen JS, 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:E1072–87.
12. Tauber M, Cutfield W. KIGS highlights: growth hormone treatment in Prader-Willi syndrome. Horm Res 2007;68 Suppl 5:48–50.
13. Di Giorgio G, Grugni G, Fintini D, Bocchini S, Spera S, Cuttini M, et al. Growth hormone response to standard provocative stimuli and combined tests in very young children with Prader-Willi syndrome. Horm Res Paediatr 2014;81:189–95.
14. Diene G, Mimoun E, Feigerlova E, Caula S, Molinas C, Grandjean H, et al. Endocrine disorders in children with Prader-Willi syndrome: data from 142 children of the French database. Horm Res Paediatr 2010;74:121–8.
15. Burman P, Ritzen EM, Lindgren AC. Endocrine dysfunction in Prader-Willi syndrome: a review with special reference to GH. Endocr Rev 2001;22:787–99.
16. Casamitjana L, Gimenez-Palop O, Corripio R, Pareja R, Berlanga E, Rigla M, et al. Glucagon stimulation test to assess growth hormone status in Prader-Willi syndrome. J Endocrinol Invest 2021;44:621–9.
17. Cohen M, Harrington J, Narang I, Hamilton J. Growth hormone secretion decreases with age in paediatric Prader-Willi syndrome. Clin Endocrinol (Oxf) 2015;83:212–5.
18. Yoon JY, Cheon CK, Lee JH, Kwak MJ, Kim HJ, Kim YJ, et al. Response to growth hormone according to provocation test results in idiopathic short stature and idiopathic growth hormone deficiency. Ann Pediatr Endocrinol Metab 2022;27:37–43.
19. Lee HS. The effects of growth hormone treatment on height in short children. Ann Pediatr Endocrinol Metab 2022;27:1–2.
20. Bakker NE, Kuppens RJ, Siemensma EP, Tummers-de Lind van Wijngaarden RF, Festen DA, Bindels-de Heus GC, et al. Eight years of growth hormone treatment in children with Prader-Willi syndrome: maintaining the positive effects. J Clin Endocrinol Metab 2013;98:4013–22.
21. Bakker NE, Lindberg A, Heissler J, Wollmann HA, Camacho-Hubner C, Hokken-Koelega AC, et al. Growth hormone treatment in children with Prader-Willi syndrome: three years of longitudinal data in prepubertal children and adult height data from the KIGS Database. J Clin Endocrinol Metab 2017;102:1702–11.
22. Reus L, Pillen S, Pelzer BJ, van Alfen-van der Velden JA, Hokken-Koelega AC, Zwarts M, et al. Growth hormone therapy, muscle thickness, and motor development in Prader-Willi syndrome: an RCT. Pediatrics 2014;134:e1619–27.
23. Reus L, Pelzer BJ, Otten BJ, Siemensma EP, van Alfen-van der Velden JA, Festen DA, et al. Growth hormone combined with child-specific motor training improves motor development in infants with Prader-Willi syndrome: a randomized controlled trial. Res Dev Disabil 2013;34:3092–103.
24. Passone CG, Franco RR, Ito SS, Trindade E, Polak M, Damiani D, et al. Growth hormone treatment in Prader-Willi syndrome patients: systematic review and meta-analysis. BMJ Paediatr Open 2020;4e000630.
25. Luo Y, Zheng Z, Yang Y, Bai X, Yang H, Zhu H, et al. Effects of growth hormone on cognitive, motor, and behavioral development in Prader-Willi syndrome children: a meta-analysis of randomized controlled trials. Endocrine 2021;71:321–30.
26. Yang A, Choi JH, Sohn YB, Eom Y, Lee J, Yoo HW, et al. Effects of recombinant human growth hormone treatment on growth, body composition, and safety in infants or toddlers with Prader-Willi syndrome: a randomized, active-controlled trial. Orphanet J Rare Dis 2019;14:216.
27. Festen DA, Wevers M, Lindgren AC, Bohm B, Otten BJ, Wit JM, et al. Mental and motor development before and during growth hormone treatment in infants and toddlers with Prader-Willi syndrome. Clin Endocrinol (Oxf) 2008;68:919–25.
28. Carrel AL, Myers SE, Whitman BY, Eickhoff J, Allen DB. Long-term growth hormone therapy changes the natural history of body composition and motor function in children with Prader-Willi syndrome. J Clin Endocrinol Metab 2010;95:1131–6.
29. Eiholzer U, L’allemand D, Schlumpf M, Rousson V, Gasser T, Fusch C. Growth hormone and body composition in children younger than 2 years with Prader-Willi syndrome. J Pediatr 2004;144:753–8.
30. Carrel AL, Moerchen V, Myers SE, Bekx MT, Whitman BY, Allen DB. Growth hormone improves mobility and body composition in infants and toddlers with Prader-Willi syndrome. J Pediatr 2004;145:744–9.
31. Duis J, van Wattum PJ, Scheimann A, Salehi P, Brokamp E, Fairbrother L, et al. A multidisciplinary approach to the clinical management of Prader-Willi syndrome. Mol Genet Genomic Med 2019;7e514.
32. Carrel AL, Myers SE, Whitman BY, Allen DB. Benefits of long-term GH therapy in Prader-Willi syndrome: a 4-year study. J Clin Endocrinol Metab 2002;87:1581–5.
33. Obata K, Sakazume S, Yoshino A, Murakami N, Sakuta R. Effects of 5 years growth hormone treatment in patients with Prader-Willi syndrome. J Pediatr Endocrinol Metab 2003;16:155–62.
34. Kuppens RJ, Bakker NE, Siemensma EP, Donze SH, Stijnen T, Hokken-Koelega AC. Metabolic health profile in young adults with Prader-Willi syndrome: results of a 2-year randomized, placebo-controlled, crossover GH trial. Clin Endocrinol (Oxf) 2017;86:297–304.
35. Hauffa BP. One-year results of growth hormone treatment of short stature in Prader-Willi syndrome. Acta Paediatr Suppl 1997;423:63–5.
36. Lindgren AC, Hagenas L, Muller J, Blichfeldt S, Rosenborg M, Brismar T, 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;423:60–2.
37. Lindgren AC, Hagenas L, Ritzen EM. Growth hormone treatment of children with Prader-Willi syndrome: effects on glucose and insulin homeostasis. Swedish National Growth Hormone Advisory Group. Horm Res 1999;51:157–61.
38. Carrel AL, Myers SE, Whitman BY, Allen DB. Growth hormone improves body composition, fat utilization, physical strength and agility, and growth in Prader-Willi syndrome: a controlled study. J Pediatr 1999;134:215–21.
39. Myers SE, Carrel AL, Whitman BY, Allen DB. Physical effects of growth hormone treatment in children with Prader-Willi syndrome. Acta Paediatr Suppl 1999;88:112–4.
40. Myers SE, Carrel AL, Whitman BY, Allen DB. Sustained benefit after 2 years of growth hormone on body composition, fat utilization, physical strength and agility, and growth in Prader-Willi syndrome. J Pediatr 2000;137:42–9.
41. Carrel AL, Myers SE, Whitman BY, Allen DB. Sustained benefits of growth hormone on body composition, fat utilization, physical strength and agility, and growth in Prader-Willi syndrome are dose-dependent. J Pediatr Endocrinol Metab 2001;14:1097–105.
42. Myers SE, Whitman BY, Carrel AL, Moerchen V, Bekx MT, Allen DB. Two years of growth hormone therapy in young children with Prader-Willi syndrome: physical and neurodevelopmental benefits. Am J Med Genet A 2007;143A:443–8.
43. Lin HY, Lin SP, Tsai LP, Chao MC, Chen MR, Chuang CK, et al. Effects of growth hormone treatment on height, weight, and obesity in Taiwanese patients with Prader-Willi syndrome. J Chin Med Assoc 2008;71:305–9.
44. Meinhardt U, Christiansen JS, Farholt S, Lammer C, Ostergaard JR, Schmidt F, et al. The efficacy and safety of long-term Norditropin treatment in children with Prader-Willi syndrome. Horm Metab Res 2013;45:532–6.
45. Kuppens RJ, Bakker NE, Siemensma EP, Tummers-de Lind van Wijngaarden RF, Donze SH, Festen DA, et al. Beneficial effects of GH in young adults with Prader-Willi syndrome: a 2-year crossover trial. J Clin Endocrinol Metab 2016;101:4110–6.
46. Donze SH, Kuppens RJ, Bakker NE, van Alfen-van der Velden JA, Hokken-Koelega AC. Bone mineral density in young adults with Prader-Willi syndrome: a randomized, placebo-controlled, crossover GH trial. Clin Endocrinol (Oxf) 2018;88:806–12.
47. Angulo M, Abuzzahab MJ, Pietropoli A, Ostrow V, Kelepouris N, Tauber M. Outcomes in children treated with growth hormone for Prader-Willi syndrome: data from the ANSWER Program and NordiNet International Outcome Study. Int J Pediatr Endocrinol 2020;2020:20.
48. Grootjen LN, Trueba-Timmermans DJ, Damen L, Mahabier EF, Kerkhof GF, Hokken-Koelega AC. Long-term growth hormone treatment of children with PWS: the earlier the start, the better the outcomes? J Clin Med 2022;11:2496.
49. Tauber M, Diene G, Molinas C, Hebert M. Review of 64 cases of death in children with Prader-Willi syndrome (PWS). Am J Med Genet A 2008;146A:881–7.
50. Berini J, Spica Russotto V, Castelnuovo P, Di Candia S, Gargantini L, Grugni G, et al. Growth hormone therapy and respiratory disorders: long-term follow-up in PWS children. J Clin Endocrinol Metab 2013;98:E1516–23.
51. Crino A, Di Giorgio G, Manco M, Grugni G, Maggioni A. Effects of growth hormone therapy on glucose metabolism and insulin sensitivity indices in prepubertal children with Prader-Willi syndrome. Horm Res 2007;68:83–90.
52. Murakami N, Obata K, Abe Y, Oto Y, Kido Y, Itabashi H, et al. Scoliosis in Prader-Willi syndrome: effect of growth hormone therapy and value of paravertebral muscle volume by CT in predicting scoliosis progression. Am J Med Genet A 2012;158A:1628–32.
53. Kim MS, Kim J, Cho J, Cho SY, Jin DK. Tailored management of life-threatening complications related to severe obesity in a young adult with Prader-Willi syndrome. Ann Pediatr Endocrinol Metab 2022;27:148–52.
54. Grugni G, Marzullo P, Ragusa L, Sartorio A, Trifiro G, Liuzzi A, et al. Impairment of GH responsiveness to combined GH-releasing hormone and arginine administration in adult patients with Prader-Willi syndrome. Clin Endocrinol (Oxf) 2006;65:492–9.
55. Grugni G, Giardino D, Crino A, Malvestiti F, Ballarati L, Di Giorgio G, et al. Growth hormone secretion among adult patients with Prader-Willi syndrome due to different genetic subtypes. J Endocrinol Invest 2011;34:493–7.
56. Oto Y, Obata K, Matsubara K, Kozu Y, Tsuchiya T, Sakazume S, et al. Growth hormone secretion and its effect on height in pediatric patients with different genotypes of Prader-Willi syndrome. Am J Med Genet A 2012;158A:1477–80.
57. Kwun C, Cho SY, Maeng SH, Jung YJ, Jin DK. Growth hormone treatment and its effect on height in pediatric patients with different genotypes of Prader-Willi syndrome. Ann Pediatr Endocrinol Metab 2012;17:175–8.
58. Lafortuna CL, Minocci A, Capodaglio P, Gondoni LA, Sartorio A, Vismara L, et al. Skeletal muscle characteristics and motor performance after 2-year growth hormone treatment in adults with Prader-Willi syndrome. J Clin Endocrinol Metab 2014;99:1816–24.
59. Hirsch HJ, Gross-Tsur V. Growth hormone treatment for adults with Prader-Willi syndrome: another point of view. Orphanet J Rare Dis 2021;16:337.
60. Hoybye C, Holland AJ, Driscoll DJ, ; Clinical and Scientific Advisory Board of The International Prader-Willi Syndrome Organisation. Time for a general approval of growth hormone treatment in adults with Prader-Willi syndrome. Orphanet J Rare Dis 2021;16:69.
61. Butler MG, Manzardo AM, Heinemann J, Loker C, Loker J. Causes of death in Prader-Willi syndrome: Prader-Willi Syndrome Association (USA) 40-year mortality survey. Genet Med 2017;19:635–42.
62. Tauber M, Hoybye C. Endocrine disorders in Prader-Willi syndrome: a model to understand and treat hypothalamic dysfunction. Lancet Diabetes Endocrinol 2021;9:235–46.
63. Grugni G, Sartorio A, Soranna D, Zambon A, Grugni L, Zampino G, et al. Long-term effects of GH therapy in adult patients with Prader-Willi syndrome: a longitudinal study. Front Endocrinol (Lausanne) 2023;14:1198616.
64. Frixou M, Vlek D, Lucas-Herald AK, Keir L, Kyriakou A, Shaikh MG. The use of growth hormone therapy in adults with Prader-Willi syndrome: a systematic review. Clin Endocrinol (Oxf) 2021;94:645–55.
65. Rosenberg AG, Passone CG, Pellikaan K, Damiani D, van der Lely AJ, Polak M, et al. Growth hormone treatment for adults with Prader-Willi syndrome: a meta-analysis. J Clin Endocrinol Metab 2021;106:3068–91.

Article information Continued

Table 1.

Indications for growth hormone injection therapy in Prader-Willi syndrome

South Korea United States FDA GH Research Society [11]
Age of treatment initiation Reimbursement for ages 2 and above Unspecified age reimbursement No consensus on rhGH start age, but agreed on benefits of treating before obesity onset, often by age 2
Duration of treatment Stop in children with <1 cm growth rate & before epiphyseal closure Stop in children with almost closed epiphyses Until reaching an adult height or near adult height
Dosage 0.24 mg/kg/week 0.24 mg/kg/week Initial dose: 0.5 mg/m2/day, adjustable to approximately 1 mg/m2/day
Approved somatropin injection Genotropin (approved Mar. 2004) Genotropin (approved Jun. 2000)
Scitropin Aa) Omnitropea)
Eutropin (approved Dec. 2019)
Contraindications for PWS Severe obesity or severe respiratory disorders Severe obesity, history of upper airway obstruction or sleep apnea, or unidentified respiratory infection Severe obesityb), uncontrolled diabetes, untreated severe obstructive sleep apnea, active cancer, and active psychosis

FDA, Food and Drug Administration; GH, growth hormone; rhGH, recombinant human growth hormone; PWS, Prader-Willi syndrome.

a)

Biosimilars;

b)

Considered severe if BMI >95th percentile with obesity-related complications, such as sleep apnea, fatty liver, or metabolic issues.

Table 2.

Previous literature on the effects of growth hormone treatment in pediatric Prader-Willi syndrome

Study Study design Mean age at GH start No. of patients (female %) GHT duration GH dose Country/Ethnicity Outcomes
Hauffa (1997) [35] RCT 8.25 years 17 (41) 1 year 0.15 IU/kg/day NA (Germany research team) Increase height velocity.
(1) Lindgren et al. (1997) [36] RCT 3–12 years 27 (n/a) 2 years Group A: 0.1 IU/kg/day The Swedish National Growth Hormone Advisory Group (1) Increased height SDS, reduced the percentage body fat and increased the muscle area of the thigh.
(2) Lindgren et al. (1999) [37] Group B: 0.2 IU/kg/day (2) Increased insulin blood levels, unchanged fasting glucose and HbA1c but decreased glucose elimination rate after an intravenous glucose test.
(1) Carrel et al. (1999) [38] RCT 9.8 years 54 (48) control, 19; GH group, 35 12 months, 24 months [40] 1 mg/m2/day NA (USA research team) (1, 2) Increased height velocity Z-scores; decreased percent body fat; and improved respiratory muscle function, physical strength, and agility. However, resting energy expenditure did not change.
(2) Myers et al. (1999) [39]
(3) Myers et al. (2000) [40] (3) Sustained increase in lean body mass, decrease in body fat percentage, improvements in physical strength and agility, and increased fat oxidation after 24 months.
Carrel et al. (2001) [41] RCT 10.9 years (5–16) 46 (48) 36 months 1 mg/m2/day (24 months) NA (USA research team) Decrease in fat mass and increase in lean body mass, growth velocity, and resting energy expenditure occurred with the standard 1.5 mg/m2/day, but not with lower doses of GH.
0.3–1.5 mg/m2/day (25–36 months)
Obata et al. (2003) [33] Retrospective 6 months–14 years 37 (49) 6 months-5 years 0.5 IU/kg/week Japanese Improved height velocity, height SDS, final height, and the degree of obesity.
Carrel et al. (2004) [30] RCT 15±9 months (4–37) 29 (45) 12 months 1 mg/m2/day NA (USA research team) Decreased percent body fat, increased lean body mass, increased height velocity Z-scores, and patients <18 months old showed higher mobility skill acquisition.
Myers et al. (2007) [42] RCT Mean 15±9 months (154–37) 25 (n/a) 2 years GHT (1 mg/m2/day) or 1 year without GH and then placed on GH (1.5 mg/m2/day) for 1 year NA (USA research team) Normalization of length/height SDS, faster head growth, increased lean body mass accrual, and decreased percent body fat, as well as improved language and cognitive quotient Z-scores.
Lin et al. (2008) [43] Retrospective 4.3±3.0 years 27 (70) 3 years 0.1 IU/kg/day Taiwanese Increased height SDS, decreased BMI SDS, decreased Rohrer index.
Meinhardt et al. (2013) [44] Retrospective 3.8±3.0 years 41 (n/a) 4.1 years 0.03–0.06 mg/kg/day Switzerland, Denmark, and Germany Significant improvements in height and body composition.
(1) Kuppens et al. (2016) [45] RCT, crossover study on young adults with PWS who were treat- ed with GH from childhood until attainment of AH 17.2 years (at the initiation of GH in this study) 27 (70) 2 years 0.67 mg/m2/day NA (Dutch research team) (1) Compared with GH treatment, 1 year of placebo after attainment of AH did not deteriorate BMD SDS in young adults with PWS. GH did not prevent the decline in BMD SDS in hypogonadal young adults with PWS, unless combined with sex steroid replacement therapy.
(2) Kuppens et al. (2017) [34] 8.5 years (at the onset of previous GH therapy) (2) Compared with placebo, GH treatment resulted in similar glucose and insulin levels during oral glucose tolerance tests. GH treatment had no adverse effects on metabolic health profile.
(3) Donze et al. (2018) [46] (3) Lower fat mass and higher lean body mass compared to placebo group.
Yang et al. (2019) [26] RCT 6.3 months (2.3–24) 34 (62) 52 weeks 0.24mg/kg/week South Korea Increased height SDS and lean body mass, decreased fat mass (%), improved motor and cognitive develop- ments.
Angulo et al. (2020) [47] Observational 4.42 years 129 (50) 2.48 years 0.03mg/kg/day ANSWER Program (USA), NordiNet IOS (Europe and the Middle East) Increased height SDS, earlier treatment resulted in a greater increase in height, and a longer treatment peri- od resulted in better outcomes for both height and BMI.
Passone et al. (2020) [24] Meta-analysis (16 RCTs and 20 NRCTs) NA NA NA NA Sweden (3), Netherland (17), USA (8), Denmark (2), Australia (2), Finland (1), Taiwan (1), France (2), Germany (1), Switzerland (1) Improvement in height (1.67 SD), BMI Z-scores (–0.67 SDS) and fat mass proportion (–6.5% SDS).
Grootjen et al. (2022) [48] Open-label, prospective study 1.92 years 104 (NA) 8 years 1 mg/m2/day Netherland Better body composition and tall height in treated group. Especially, lower trunk/peripheral fat ratio, lower fat mass and higher IQ in group A.
Group A: <1 Control: 22
Group B: 2–5 GH group: 82

GH, growth hormone; GHT, growth hormone therapy; RCT, randomized controlled trial; NA, not available; SDS, standard deviation score; HbA1c, hemoglobin A1c; BMI, body mass index; PWS, Prader-Willi syndrome; AH, adult height; BMD, bone mineral density; NRCT, non-randomized controlled trial; SD, standard deviation; IQ, intelligence quotient.

Table 3.

Recent studies on the effects of growth hormone treatment in adult Prader-Willi syndrome

Study Study design Age at the time of study No. of patients (female %) GHT duration GH dose Country/ Ethnicity Outcomes
Grugni et al. (2023) [63] Longitudinal Median of 27.1 years 12 (42) 17 years 0.35 mg/day Caucasian Lower waist circumference, reduction of fat mass (%), a slight impairment of glucose homeostasis (an increase in only the fasting glucose).
Frixou et al. (2021) [64] Meta-analysis (20 papers) Median of 26.2 years 424 (49) Median of 1 year Median of 0.8 mg/day Japan (1), Netherlands (3), Italy (5), Sweden (8), Norway (3), Denmark (3), USA (2) Increase in LBM and reductions in FM (%). The impact on cholesterol, echocardiography, and BMD remains unclear.
Rosenberg et al. (2021) [65] Meta-analysis (9 RCTs and 20 NRCTs) The median or average age is not provided RCTs (the sample size per study ranged from 26 to 27 individuals) From a minimum of 1 year to a maximum of 15 years Individually titrated NA With over 12 months of GH treatment, body composition improved with increased lean body mass and reduced fat mass. However, no significant changes were observed in BMI, cholesterol levels, glucose levels, or bone mineral density.
NRCTs (the sample size per study ranged from 10 to 1,067 individuals)

GHT, growth hormone therapy; GH, growth hormone; LBM, lean body mass; FM, fat mass; BMD, bone mineral density; RCT, randomized controlled trial; NRCT, non-randomized controlled trial; NA, not available; BMI, body mass index.