Abstract
The success of growth hormone (GH) replacement in children with classical GH deficiency has led to excitement that other causes of short stature may benefit similarly. However, clinical experience has shown less consistent and generally less dramatic effects on adult height, perhaps not surprising in light of increased understanding of GH and growth plate biology. Nonetheless, clinical demand for GH treatment continues to grow. Upon the 20th anniversary of the US Food and Drug Administration's approval of GH treatment for idiopathic short stature, this review will consider the factors underlying the expansion of GH treatment, the biological mechanisms of GH action, the non-GH–deficient uses of GH as a height-promoting agent, biological constraints to GH action, and future directions.
Keywords: growth hormone, pediatric, growth plate, indications
“We want to look into growth hormone treatment for [my son]. I know some people do it just because.”
This actual chief complaint from an adolescent patient's mother on presentation to a pediatric endocrinology clinic highlights the perceptions that growth hormone (GH) treatment is effective at enhancing height and offering inherent advantages, without regard to an underlying diagnosis or the mechanisms of GH action, and with minimal to negligible safety concerns. It epitomizes the evolution of GH treatment as expansive biotechnology, spreading from treatment of an authentic disease (GH deficiency [GHD]) to treatment of conditions that blur the boundary between disease and variation (1). Upon the 20th anniversary of the US Food and Drug Administration (FDA)'s approval of GH treatment for idiopathic short stature (ISS), this review will consider the factors underlying the expansion of GH treatment, the biological mechanisms of GH action, the non-GHD uses of GH, biological constraints to GH action, and future directions. We will focus on GH use as a height-promoting agent and defer the approved non-GHD indications as an anabolic agent (HIV/AIDS-associated wasting and 4-week course for short bowel syndrome with parenteral nutrition dependence) and GH abuse (for sports performance and antiaging).
Factors Underlying the Expansion of GH Treatment
Due to the limited supplies of cadaveric GH, early experience with GH treatment was restricted to pediatric patients with severe GHD and doses based on availability (2, 3). Mean adult height of untreated individuals with isolated idiopathic GHD was −4.7 SD (range −3.9 to −6.0 SD), and mean gain in adult height was 1.5 to 2.0 SD up to 3.5 SD with 2 to 4×/week GH injections at variable and low doses by current standards (4). The dramatic responsiveness of patients with severe GHD to GH replacement fueled great excitement that many more people could be helped similarly when the advent of recombinant human GH in 1985 enabled production bound solely by demand (5). Thus, endocrinologists explored GH treatment of patients with ever milder cases of GHD and even non-GHD short stature, as well as conditions where GH's biology as a growth-promoting and anabolic agent may be beneficial.
Four broad forces sustained that expansion. Firstly, reliance on provocative GH testing to distinguish patients with GHD from those with adequate GH production despite recognition of the tests’ poor sensitivity and poor specificity means patients are frequently misclassified, with a greater tendency toward falsely diagnosing children with GHD (6, 7). Thus, the desire naturally followed to trial GH treatment for patients with “normal” responses who actually may have been misclassified or who may benefit from GH treatment anyway as a means of avoiding inadvertent harm from faulty tests. This expansive force has been opposed by the need to avoid the converse harm, also from flawed tests, of unnecessarily treating patients who do not actually have GHD or will not respond to treatment. Endocrinologists have struggled for decades to achieve balance between these opposing harms (8) by setting the appropriate test threshold value (9) even though that cannot overcome the overlapping test results between GH-deficient and GH-replete patients (6). The second force sustaining expansion of GH treatment is heightism, the prejudice that taller stature is beneficial in achieving social success (10). Although the evidence supporting a height benefit is inconsistent (11, 12) and prone to bias (13), parents of pediatric primary care patients (14) and parents of children receiving endocrine care for short stature (15) both rated concerns about psychosocial functioning, in childhood and projected into adulthood, as influencing their decision to seek height-related medical care. The third force consists of financial and professional incentives to expand GH use (16). This has been reinforced by the current consumer health care culture in the United States and elsewhere, in which consumer demands serve as the business model's primary driver and patient-reported outcomes like Press Ganey scores (Press Ganey Associates, South Bend, IA) or online reviews pressure clinicians to acquiesce to patient demands over their clinical assessment of need (17). A fourth force sustaining expansion of GH treatment is the positive reinforcement to parents and clinicians from the early growth acceleration experienced by most children treated with GH (18-21), although, as discussed later, bone age progression in this context may not confer a commensurate increase in adult height. In sum, national commercial insurance claims data showed an almost tripling of the annual prevalence of youth treated with GH in the United States from 2001 to 2016 (22).
Biological Mechanisms of GH Action
Expansion of GH treatment, involving both higher doses and new indications, drew upon the premises that more is better and that sometimes biological blockages can be “pushed” or bypassed and thereby overcome by quantity even in the absence of deficiency. GH's physiological actions as a growth promoting, anabolic, and lipolytic agent (23) made it appealing for a variety of clinical indications. For instance, in addition to stimulating somatic growth, GH promotes lean muscle mass and bone mineral accrual, more favorable lipid profiles, and cardiovascular function.
The endocrine GH/insulin-like growth factor (IGF)-I axis, extensively described elsewhere (24), seems to have originated in amphioxus, a basal chordate (25). Seminal experiments with liver-specific igf1 gene-deleted (LID) mice, which demonstrated normal postnatal growth despite 75% reduction in circulating IGF-I concentrations (26), revealed the importance of local (ie, autocrine and paracrine) vs endocrine GH/IGF production and action. This finding lent credence to the notion that our diagnostic tests measuring circulating (endocrine) GH and IGF-I levels may miss situations in which additional GH and/or IGF-I may be beneficial.
GH and IGF-I function in both endocrine and autocrine/paracrine fashions within the growth plate (27, 28) (and throughout the body (29)) and serve both overlapping and distinct roles. In the sequence of bone elongation at the growth plate, GH stimulates differentiation of reserve (progenitor) cells into chondrocytes in the resting zone (30), and IGF-I stimulates proliferation of the chondrocytes in the proliferative zone. GH induces IGF-I expression by chondrocytes in the hypertrophic zone, wherein chondrocytes enlarge and secrete extracellular matrix in the final stage of chondrogenesis and IGF-I exerts direct effects, (30, 31). The hypertrophic zone then is invaded by the resorptive front of blood vessels, osteoclasts, and osteoblasts that transforms the cartilage into bone (endochondral ossification), and both GH and IGF-I are important here, too, regulating bone marrow stem cell differentiation and proliferation (30, 31).
Non-GHD Uses of GH
FDA-Approved Indications
As the availability of GH increased, its use expanded to include children with non-GH deficient short stature. These conditions can be categorized as genetic (Turner and Noonan syndromes, and short stature homeobox–containing [SHOX] gene deficiency), secondary to chronic disease (chronic renal insufficiency [CRI]), or size determined (small for gestational age without catch-up growth [SGA] and ISS). This third category includes patients who meet certain growth criteria, without specifying an underlying diagnosis. GH also has been approved for children with Prader–Willi syndrome, caused by lack of expression from paternally inherited imprinted genes on chromosome 15q11-13 (32). Although the diagnosis of Prader–Willi syndrome suffices to qualify for GH treatment without proving GHD (33), much of the phenotype stems from hypothalamic dysfunction and a large proportion of patients with Prader–Willi syndrome have GHD, so this GH indication lies beyond the scope of this paper. Given that each of these conditions can be associated with significant short stature in adulthood, the availability of a treatment for previously untreatable short stature brought an understandable demand for GH treatment.
In order to determine the efficacy of GH treatment in increasing adult height for each of these indications, randomized controlled trials including similar patient populations who were observed until attainment of adult height would have been needed. There are obvious challenges in performing such studies in the setting of an available medication that was called “growth” hormone, and in allowing placebo control for a chronic, daily injectable medication in children. Alternative approaches were employed, including the use of historical controls, patients as their own controls using pretreatment projected adult height, or short-term (1- or 2-year) studies looking at surrogate proximate outcomes like height velocity or change in height standard deviation score (SDS).
To write this paper, a review of the literature searched for studies reporting outcome data for each indication, prioritizing first by study design and then by sample size. Randomized controlled studies with actual adult height outcomes in treated and control groups were sought. Large registry cohorts also were sought, to summarize their most recent reported height outcomes. Studies outside of these designs were included as well, prioritizing those with large patient numbers and control groups but recognizing the strengths and limitations of these study designs in assessing efficacy of GH treatment (Table 1).
Table 1.
Study design components | Strengths | Challenges and Limitations |
---|---|---|
Data collection | ||
Prospective | Data collection can be designed to answer the study question Participants can be randomized to treated and untreated cohorts Equitable recruitment of participants from different demographics is possible |
Requires more resources Participants must be recruited prospectively and retained Prolonged study duration, if both groups are followed to adult height Bone age acceleration can lead to overestimated efficacy in short-term studies |
Retrospective | Quicker (data already extant) Feasibility |
There may be selection bias, as patient characteristics (eg, shorter stature) may have influenced treatment decisions Harder to find a matched control group May be harder to cleanly define the participant characteristics, especially if assays for measuring key biomarkers have changed or if clinician diagnoses are not standardized All the desired data may not be available May be less generalizable, especially if secular changes have occurred |
Participant recruitment | ||
Randomized, controlled trial | Least prone to bias Prospective data collection |
Must recruit the right kind and matching participants Participants may not want to risk being in the control group (ie, prefer off-label treatment) |
Observational cohort | Easier to recruit participants | May be prone to selection bias |
Registry | Can accumulate a large study population → increases statistical power Can collect long-term longitudinal data Can collect “real-world” data |
May be prone to selection bias May be prone to ascertainment bias No untreated control group to determine effect size May be harder to cleanly define the participant characteristics, especially if assays for measuring key biomarkers have changed or if clinician diagnoses are not standardized |
Case series | Feasibility Can be exploratory and provide preliminary data to inform more robust studies |
Smaller sample → less power or may not be amenable to statistical analyses at all May be prone to selection bias No control group |
Primary outcome | ||
(Near)Adult height | Gold-standard outcome for assessing total height gain from GH treatment | Takes longer to collect data Requires more resources Must recruit/retain participants |
Short-term outcomes (height velocity and/or change in height SDS) | Feasibility Can more rapidly discard ineffective treatments |
Small measurement errors get amplified in calculating annualized height velocity Can overestimate the impact of GH on adult height when bone age also accelerates |
Comparator/control | ||
Untreated participants with same condition | Best group to control against confounders Prospective data collection |
May be prone to selection bias Participants may not want to risk being in the untreated group (ie, prefer off-label treatment) |
Historical controls with same condition | Data already extant | May be harder to cleanly define the participant characteristics, especially if assays for measuring key biomarkers have changed or if clinician diagnoses are not standardized Similarly affected controls may not be able to match closely Prone to confounding and less generalizability if treatment practices have changed with time |
Referent growth chart | Readily available data Provides population-level “normal” ranges Do not need additional participants as controls |
Does not take personal genetic growth potential into account |
Sex-adjusted midparental height | Growth potential that is more specific to the individual Do not need additional participants as controls |
Self- or partner-reported heights are frequently inaccurate Children often do not grow to midparental height (ie, sex-adjusted mean) exactly, especially if there is a large spread between the parents’ heights |
Self at baseline: projected adult height | Do not need additional participants as controls | Projected heights have a ± 2-inch statistical error Projected heights vary depending on projection model used |
Self at baseline: height SDS or pretreatment height velocity | Do not need additional participants as controls | Small measurement errors get amplified in calculating annualized height velocity Requires lead-in time for pretreatment data; either follow longer to measure or accept clinical height data which may not be as accurate Does not account for bone age delay at start of treatment |
Additional challenges and limitations common to all approaches include publication bias and issues related to funding source.
Abbreviations: GH, growth hormone; SDS, standard deviation score.
The greatest numbers of patients studied for GH efficacy come from proprietary postmarketing surveillance studies (a.k.a. GH registries), initially required by the FDA of the new recombinant products (5) following the Creutzfeldt–Jacob scare of cadaveric GH (2) and then voluntarily continued by the GH manufacturers. The outcome data are summarized in Table 2. The strength of the GH registries derives from their large sample sizes, long prospective longitudinal nature, and “real-world” observation of GH treatment in clinical practice internationally. However, important limitations are inherent to such study design, including the lack of untreated control groups and standardized diagnoses, as well as the potential for several types of bias (34) (Table 1).
Table 2.
KIGS (35) | NCGS (36) | GeNeSIS (19, 37) | |
---|---|---|---|
Geographic region | International | North America | United States, Germany, and France |
Dates of study | 1987-2012 | 1985-2010 | 1999-2015 |
Total number of participants with efficacy data | 55 284 | 19 422 | 2522 |
Minimum treatment duration for efficacy analyses (years) | 1 | 3 | 4 |
Total number of participants with (near) adult height data | 7911 | 8665 | 2291 |
Adult height comparison | Difference between baseline height SDS and (near) adult height SDS —provided in primary data | Difference between baseline height SDS and (near) adult height SDS—calculated here post hoc | Difference between baseline height SDSa and (near) adult height SDS—calculated here post hoc |
Height statistics | Median (IQR), according to population specific growth charts | Mean ± SD, according to CDC growth charts | Overall mean height SDS calculated from the pooled mean data reported by country |
Turner syndrome | n = 1631 | n = 1373 | n = 385 |
Baseline height | −3.2 ± 0.9 SD | −2.66 SD | |
(Near) adult height | −1.9 ± 1 SD | −1.75 SD | |
Delta height | +1.07 (0.1-2.12) SD | +1.3 SD | +0.91 SD |
Calculated delta height (cm)b | 6.96 cm | 8.45 cm | 5.92 cm |
SHOX haploinsufficiency | n = 107 | ||
Baseline height | −2.41 SD | ||
(Near) adult height | −1.54 SD | ||
Delta height | +0.87 SD | ||
Calculated delta height (cm)b | Male: 6.18 cm Female: 5.66 cm |
||
Noonan syndrome | n = 72 | ||
Baseline height | −3.4 ± 1.1 SD | ||
(Near) adult height | −2.1 ± 1.04 SD | ||
Delta height | +1.3 SD | ||
Calculated delta height (cm)b | Male: 9.23 cm Female: 8.45 cm |
||
Chronic renal insufficiency |
n = 142
(93 male, 49 female) |
n = 117 | |
Baseline height | −3.1 ± 1.3 SD | ||
(Near) adult height | −1.6 ± 1.4 SD | ||
Delta height | Male: +1.3 (−0.18, 2.46) SD Female: +1.59 (−0.14, 3.3) SD |
+1.5 SD | |
Calculated delta height (cm)b | Male: 9.23 cm Female: 10.33 cm |
Male: 10.65 cm Female: 9.75 cm |
|
Idiopathic short stature c |
n = 459
(230 male, 129 female) |
n = 1386 | |
Baseline height | −2.7 ± 0.8 SD | ||
(Near) adult height | −1.1 ± 1 SD | ||
Delta height | Male: +1.37 (0.44, 2.25) SD Female: +1.62 (0.56, 2.85) SD |
+1.6 SD | |
Calculated delta height (cm)b | Male: 9.73 cm Female: 10.53 cm |
Male: 11.36 cm Female: 10.4 cm |
|
Small for gestational age |
n = 334
(149 male, 185 female) |
n = 176 | |
Baseline height | −2.53 SD | ||
(Near) adult height | −1.48 SD | ||
Delta height | Male: +1.34 (0.52, 2.61) SD Female: +1.57 (0.59, 2.76) SD |
+1.05 SD | |
Calculated delta height (cm)b | Male: 9.51 cm Female: 10.2 cm |
Male: 7.46 cm Female: 6.83 cm |
NordiNet IOS and ANSWER studies (38, 39) were not included in this table, as adult heights were not reported.
Abbreviations: CDC, Centers for Disease Control; IQR, interquartile range; SDS, standard deviation score.
a Reported mean baseline bone age delays in the GeNeSIS study (37): Turner syndrome 1.1 year, SHOX haploinsufficiency 0.6 year, small for gestational age 0.5 year.
b Delta height was calculated from the difference between the mean baseline and mean (near) adult height SDS. This change in height should not be interpreted as GH effect as it does not account for baseline bone age delay, nor include untreated control subjects for comparison. The mean (SD) height for US adult males is 176.8 (7.1) cm and females is 163.3 (6.5) cm, according to the Centers for Disease Control and Prevention Growth Charts (40).
c Idiopathic short stature does not include children with diagnoses of known SHOX-haploinsufficiency or small for gestational age.
It is worth noting that sex-adjusted midparental height (41) and projected adult height, often used as comparators for gauging GH effectiveness, are themselves fraught with limitations. Parental heights commonly rely on self-reports or partner reports, and reported heights are notoriously inaccurate (42). Adult height projections are based on bone age radiographs, whose reading is very subjective, as measures of the proportion of skeletal maturation achieved. When computer-simulated samples of 1000 boys and 1000 girls with short stature were run through 3 commonly used algorithms for projecting adult height, the results were profoundly discrepant across algorithms (43).
Genetic Short Stature
Turner Syndrome and SHOX Haploinsufficiency
Turner syndrome is characterized by partial or complete monosomy of the X-chromosome in females, with clinical features reflecting the magnitude of X-chromosome deficit (44). The mechanism of short stature in Turner syndrome is thought to be related to haploinsufficiency of the SHOX gene (45), that encodes a transcription factor involved in signaling pathways controlling the proliferation and maturation of growth plate chondrocytes, necessary for bone elongation (46, 47). Reduced growth begins in utero, resulting in girls with Turner syndrome being slightly smaller than the general population at birth (48). This continues through childhood, is often compounded by an absent pubertal growth spurt, and an adult height about 16 cm below midparental height has been reported (49, 50).
A randomized controlled trial including 104 girls with Turner syndrome (61 GH treated, 43 untreated controls) demonstrated 7.3 cm (95% CI 5.4-9.1) taller stature in those treated with GH for 5.7 ± 1.6 years (51). A separate randomized controlled trial to adult height, including 149 girls, attributed a 0.78 ± 0.13 SD (5 cm) height increase to 7.2 ± 2.5 years of GH treatment (52). An additional 0.32 ± 0.13 SD (2.1 cm) height gain occurred with the addition of low-dose prepubertal estrogen (ethinylestradiol 25 ng/kg daily aged 5-8 years and 50 ng/kg daily aged 8-12 years) (52). Given the shared mechanisms of short stature in SHOX haploinsufficiency and Turner syndrome, it is perhaps not surprising that a small study comparing the effect of GH treatment in children with SHOX haploinsufficiency vs Turner syndrome (without GH-untreated controls) yielded similar adult height results (53).
Noonan Syndrome
Noonan syndrome is a genetically heterogeneous condition caused by germline variants in genes associated with the RAS/mitogen-activated protein kinase pathway (a.k.a. RASopathy). Half of reported cases are due to gain-of-function variants of the PTPN11 gene that encodes a ubiquitous nonreceptor tyrosine phosphatase (54, 55). Short stature is reported in approximately 70% of people with Noonan syndrome (56-58), although the mechanism for this association in unclear (59). Suggested mechanisms include impaired GH secretion (60, 61) (based on low overnight GH concentrations yet normal responses to provocative GH testing (61) or studies showing 30% to 45% of children not attaining a peak GH over 10 ng/mL on provocative testing (62-64)). However, the latter is difficult to interpret, as similar rates of “suboptimal” peak GH concentrations have been reported in healthy children without Noonan syndrome undergoing provocative testing (65-67).
Non-GHD mechanisms have been proposed for the short stature frequently associated with Noonan syndrome. The tyrosine phosphatase encoded by PTPN11, Src homology region 2-domain phosphatase-2 (SHP2), interacts with the GH receptor (68). Notably, both GH and IGF-I signal via phosphorylation cascades that can be inhibited by increased activity of a phosphatase like SHP2 (69). In 1 study demonstrating increased overnight GH secretion in children with PTPN11 variants, mild GH resistance was proposed as possibly contributing to the growth phenotype of this condition (70). In support of this mechanism, children with PTPN11 variants were found to have a blunted response to an IGF-I generation test when compared with short control participants (71). However, a difference in effect of GH treatment according to PTPN11 mutation status has not been described consistently (72). Further, the associated cardiovascular complications of Noonan syndrome have been suggested as contributing to impaired linear growth (57).
The efficacy of GH treatment in countering the short stature associated with Noonan syndrome is unclear. There have been no randomized controlled trials reporting adult height in children with Noonan syndrome (73), and a systematic review in 2015 highlighted the absence of evidence for treatment in this condition (73). A subsequent retrospective study of children with Noonan syndrome, comparing GH-treated with untreated controls, showed a slight but insignificantly greater adult height SDS in patients who were not treated with GH (−1.8 ± 1.1 vs −2.24 ± 0.89, P = .123), possibly reflecting selection bias towards shorter children with Noonan syndrome receiving GH treatment. There have been many short-term (74-78) and uncontrolled studies (76, 79, 80) demonstrating early height gains and increased height SDS at adult height when compared to baseline (0.8-1.1 SD (75-78)), but controlled studies to adult height remain absent.
Secondary to Chronic Disease
Chronic Renal Insufficiency
Poor growth is a common feature of CRI and this association is multifactorial. The severity of renal impairment, acidosis, biochemical parameters (serum phosphorus, calcium, albumin, and parathyroid hormone concentrations), age, and race are associated with short stature (81). Short stature also is associated with mortality in this condition (82), but this likely reflects the confounding effect of illness severity on growth and clinical outcomes.
While many factors contribute to the poor growth seen in children with CRI, abnormalities of the GH/IGF-I axis have been described (83). Overnight GH secretion is reduced in children with CRI (84), but reduced metabolic clearance of GH prolongs its circulating half-life by 25% to 50% (85). Uremia may induce GH resistance through decreased GH receptor expression (86), as suggested by the lower circulating concentrations of GH binding protein (the soluble ectodomain of GH receptor) in patients with CRI (86, 87). Animal studies have indicated that GH resistance also is mediated by impaired postreceptor signaling, through decreased GH-induced phosphorylation of JAK2, STAT5, and STAT3 (88, 89) and reduced IGF-I production (89) in CRI. Compounding reduced IGF-I expression, bioactive IGF-I levels may be reduced further by accumulating IGF binding protein (IGFBP) concentrations from decreased clearance in CRI (90, 91). In children with nephrotic syndrome, increased urinary losses of IGF-I, IGFBPs, and the acid labile subunit also may play a role (92, 93).
Given these multifactorial mechanisms contributing to impaired linear growth in children with CRI, it is difficult to predict the effect of GH treatment on linear growth. No randomized controlled trial to adult height is available to assess effect. A retrospective study including 214 children with CRI treated for more than 6 months prior to renal replacement therapy (dialysis or transplant) showed an early increase in height SDS during GH treatment (+0.8 SDS) (94). However, in those receiving renal replacement therapy, there was no significant difference in adult height between those treated with GH vs those who did not receive GH (+0.06 SD vs −0.04 SD, P = .46) (94). Another retrospective study demonstrated an early height increase with GH treatment of 0.56 SD at 2 years among 797 children treated with GH compared with matched controls, but similar growth rates thereafter (95). A case–control retrospective study including 32 GH-treated boys showed a 3.1 cm taller adult height compared with controls, but the effect of GH on adult height is difficult to interpret as the treated children were, on average, 8.2 cm shorter at baseline with a 2.1-year greater bone age delay (96).
Size-Determined Indications
Approved size-determined indications for GH treatment include children born SGA without catch-up growth or those categorized as having ISS. As these diagnoses are based on growth parameters rather than defined clinical diagnoses, children meeting these criteria represent a heterogeneous population with varied unidentified causes for short stature. Increasing accessibility of genetic testing and advancing understanding of the genetic determinants of height are prompting a reconsideration of the diagnostic categories of ISS and SGA. Approximately 20% to 30% of children diagnosed with SGA or ISS have an underlying skeletal dysplasia or other genetic etiology of short stature identified when extensively investigated (97-99). The majority of pathologies identified in these studies affect the growth plate, and not GH secretion or GH signaling (97, 98, 100). Other unidentified genetic diagnoses, including unrecognized Noonan syndrome, have also been demonstrated at high rates in similar studies (101, 102). Population-specific reference data are available (103-107) to assess for disproportionality in the clinic, and such screening may help increase the identification of skeletal dysplasia as a cause of “idiopathic” short stature. Further, given the aforementioned poor specificity of provocative GH testing for GHD (6, 67), many children who “fail” stimulation testing and receive a diagnosis of GHD likely have ISS.
Small for Gestational Age Without Catch-up Growth
In children born SGA, GH is approved by the FDA if there is no catch-up growth by age 2 years, and by the European Medicines Agency if height SDS remains below −2.5 at age 4 years (108). The recommendations include waiting to the specified ages to avoid unnecessarily treating the about 85% of children born SGA who enjoy spontaneous catch-up growth; because birth size is determined primarily by maternal, pregnancy, and placental health and nutrition, the prenatal growth stunting does not persist for most children postnatally (109). However, for children with Silver–Russell syndrome, an imprinting disorder associated with both prenatal and postnatal growth failure resulting in adult height around −3 SD, recommendations proposed deferring GH treatment until caloric deficits are addressed but not necessarily waiting to a certain age even though they are treated under the SGA indication for GH (110).
Few randomized controlled trials with adult height data are available to assess the effect of GH treatment on linear growth in children who had been born SGA. A systematic review of controlled studies included 391 children across 4 studies (111-114). When corrected for midparental height, they showed 0.78 SD (5.1 cm females, 5.5 cm males) increased adult height in those treated with GH compared with untreated controls; there was no difference between doses of GH (115). Similarly, when 167 Dutch children born SGA and treated with GH for a mean (SD) of 8.9 (2.5) years were compared with 50 controls from a different region, the GH-treated group was 0.8 SD (approximately 5.7 cm male, 5.2 cm female) taller as adults (116). Many cohort studies have been reported separately, analyzing only pretreatment and adult heights in children meeting SGA criteria, without untreated patients for comparison (19, 117-37).
Idiopathic Short Stature
The FDA-approved indication for GH treatment of children with ISS stipulates height below −2.25 SD, height not expected to reach adult height in the normal range if left untreated, and exclusion of conditions that should be monitored or treated by other means. As in other indications for GH treatment, short-term studies demonstrate a favorable effect of GH treatment on height in children treated under the ISS indication. For instance, a study including 334 children with ISS found a mean bone age delay of 1.7 years at baseline and a 2-year increase in height Z-score of +0.84 with GH treatment (119). In the few randomized controlled trials, GH treatment led to an adult height increase of 0.52 SD (3.4 cm females, 3.7 cm males) at doses of 0.22 mg/kg/week over a mean treatment duration of 4.4 years (120) or 0.5 to 0.8 SD (low or high dose GH) over a mean treatment duration of 5.9 years (18) when compared with controls. A retrospective analysis of 123 children with ISS treated with 0.32 ± 0.03 mg/kg/week of GH found a mean (near) adult height of −0.71 SDS (0.74 SD) (95% CI, −0.77 to −0.55) in 88 patients (27 patients were lost to follow-up, and 8 males also treated with testosterone were excluded). The study compiled adult height data from 3 randomized and 6 nonrandomized studies to calculate benefit of GH treatment (121).
Systematic reviews demonstrated a mean increase in adult height of 0.65 SD (4.2 cm females, 4.6 cm males) with GH treatment (122). However, these are all mean results, with a wide variation in responses at the individual level. This includes some children who did not increase their adult heights, reflecting the vastly heterogeneous nature of the ISS categorization. Magnitude of bone age delay at start of treatment was associated with “response” to GH treatment on multiple regression analysis (18), suggesting that perceived GH response may be realization of delayed bone age-associated growth potential.
Off-label Pediatric Uses
Off-label GH use has been widespread since the advent of recombinant GH, as evidenced in the GH registries by the many children treated for ISS both in the United States prior to the FDA approval and in Europe despite the lack of approval for the ISS indication by the European Medicines Agency. GH treatment of children with some identified conditions is based on positive results published in small case series or case reports (uncontrolled). Others are attempts to trial GH, with no supporting evidence, drawing on the ISS indication as justifying treatment irrespective of underlying diagnosis. Short stature is a common feature of many genetic disorders, and with genomic testing increasingly identifying private variants, the number of conditions proposed for off-label GH treatment is increasing as well.
Biological Constraints to GH Action
Despite the initial excitement born from GH replacement for children with classical GHD, experience with GH treatment for non-GHD indications has been less successful—often requiring higher GH doses to yield results that are not as dramatic, nor as consistent across patients. With increasing knowledge of growth biology, this should not be surprising as there are various biological constraints to GH action.
Negative Feedback Loops
Negative feedback loops tightly regulate both GH production/secretion and GH signaling to attenuate or terminate GH activity. The classic third-order system involves ultrashort feedback within the hypothalamus (GH-releasing hormone [GHRH] stimulates somatostatin production, which inhibits GHRH release), short feedback between the pituitary and hypothalamus (GH stimulates somatostatin production, which exerts a tonic negative effect on GH secretion, and inhibits GHRH) (123), and long feedback of IGF-I from target tissues (eg, the liver, for most circulating IGF-I) suppressing secretion of both pituitary GH and hypothalamic GHRH (124). Further, an intracellular negative feedback loop terminates signaling by the GH receptor, a member of the class I cytokine receptor superfamily that signals primarily through the JAK/STAT pathway; GH induces expression of various suppressors of cytokine signaling (SOCS)/cytokine-inducible SH2 (CIS)-containing protein family proteins, which in turn inhibit STAT signaling (125, 126). Thus, rather than exogenous GH treatment simply adding to endogenous GH in non-GHD conditions, it may lead to loss of at least some endogenous GH activity via the negative feedback loops before a net gain in GH activity is achieved. This is supported by a pharmacokinetic study in healthy men of recombinant 20-kDa human GH that found marked suppression of endogenous 22-kDa human GH secretion in a time-dependent manner (127).
Growth Plate Senescence
Growth plate senescence, an extensive developmental genetic program intrinsic to growth plate cartilage, has been recognized as causing the decline in growth rate, interrupted by “the growth spurt” of puberty, from birth until growth ceases with epiphyseal (growth plate) fusion (128). Growth plate senescence involves a gradual downregulation of many growth-promoting genes that results in depletion of the resting zone chondrocyte reservoir and reduction in the rate and degree of chondrocyte proliferation and hypertrophy until they fully cease, at which point final cartilage resorption and ossification occurs (128, 129). Bone age radiographs measure the proportion of growth plate senescence achieved (128).
The relative rates of linear growth vs the progression of growth plate senescence determine the ultimate achievable adult height. GH dosage affects bone age acceleration, and the presence of bone age delay at onset of GH treatment affects the long-term outcome in children with ISS (130). Short-term randomized controlled trials may overestimate the effect of GH treatment on growth if much of the early GH-induced height acceleration reflects earlier realization of the growth potential rather than a net adult height gain, especially compared with untreated controls who retain their delayed bone ages and future growth potential (20). For instance, high-dose GH treatment of children with ISS advanced the bone age by 3.6 years over 2 years, so despite higher height Z-score after 5 years of treatment, this did not differ significantly from untreated controls when adjusted for bone age at that time (20).
In children born SGA observed over 6 years of GH treatment, bone age advanced by a mean of more than 7 years across various GH dosing regimens. Data on bone age advancement in untreated controls were not available from this study, as untreated monitoring was abandoned after 2 years due to the early increased height velocity in the treated group (131). Such data would have been important, because children born SGA commonly experience bone age advancement in the peripubertal period related to premature adrenarche and early puberty. Adult height increment from enrollment was 2 ± 0.2 SD in those treated with 33 µg/kg and 2.7 ± 0.2 SD in those treated with 67 µg/kg; the study did not report on adult height in untreated controls (131). In a meta-analysis of 4 trials of children born SGA who were treated with differing regimens of GH, faster bone maturation occurred with higher GH doses and in older vs younger prepubertal children (132).
Growth plate senescence also is important in distinguishing height augmentation by GH treatment for non-GHD short stature from height outcomes in pituitary gigantism, caused by pituitary somatotropinomas or hyperplasia leading to chronic oversecretion of GH and IGF-I. In addition to GH excess, pituitary gigantism involves hypogonadism, from gonadotrope damage or dysfunction due to the large pituitary adenoma and/or from the substantial hyperprolactinemia that is particularly common in pituitary gigantism caused by AIP mutations or X-LAG syndrome (133). Hypogonadism deprives the estrogen effects on growth plate senescence (129), leaving the growth plates open for continued growth even in adulthood (133).
Defects in Other Signaling Systems in the Growth Plate
While historical contexts have focused on the endocrine GH/IGF axis as the principal mediator of height growth, the importance of local growth plate factors is being increasingly appreciated, not just in terms of senescence but as mediators and regulators of the growth process itself. A review of the various factors is beyond the scope of this paper (it is beautifully described elsewhere by Baron et al (134)), but they can be summarized as other hormones (with direct actions at the growth plate), cytokines, physical mechanisms (irradiation and mechanical compression), paracrine factors, extracellular matrix components, and intracellular pathways. Variants of these other players contribute to both “normal” variation in adult height and disorders of short and tall stature (134), and it is naïve to believe extra GH is capable of superseding all of them.
Need to Highly Regulate the GH/IGF System
The GH/IGF system is highly regulated because the stakes are high. Reduced IGF-I levels and action have been associated with increased longevity across organisms, from Caenorhabditis elegans to humans (135, 136). Loss of GH action from GH receptor deficiency (Laron syndrome or GH insensitivity syndrome) confers protection against diabetes and cancer (137, 138). Thus, beyond the negative feedback loops in the GH system, GH production/secretion and action are highly regulated systemically by multiple factors, including nutrients (139), orexigenic ghrelin (140), anorexigenic nesfatins (140), antiaging klotho (140), and other hormones (eg, thyroid, glucocorticoids and estrogen (141)). Similarly, at the cellular level, the GH/IGF system engages in cross-talk with various other signaling pathways that determine cell fate. For instance, p53, arguably the most important human tumor suppressor, suppresses IGF signaling by 4 distinct mechanisms: inhibiting transcription of the genes encoding the type 1 IGF receptor (142) and IGF-II (143), and stimulating transcription of the genes encoding the binding proteins, IGFBP-3 (144) and IGFBP-2 (145). Conversely, age-dependent accumulation of DNA damage was associated with GH expression in nontumorous colon tissue, and colonic autocrine/paracrine GH suppressed p53 and attenuated the DNA damage response (146). Thus, administering exogenous GH to individuals who are already GH replete attempts to override a highly regulated system and may yield additional effects beyond height.
Although the safety of GH treatment is beyond the scope of this paper and extensively reviewed elsewhere (147, 148), it bears keeping an open mind. With the advent of rhGH in 1985, the oldest recipients are now only in their 50s, and although the GH registries have shown GH to be safe during treatment (35), data on long-term post-treatment effects remain scarce and unclear, and changing patient and treatment characteristics divergent from physiologic replacement may yield different and unpredicted outcomes (34), either positive or negative. In any case, whatever underlying non-GHD growth defect the exogenous GH treatment is intended to “push” through, it also must overcome the body's various intrinsic regulatory mechanisms of GH system activity.
Future Directions
In summary, GH treatment has expanded from replacement for individuals with GHD to multiple FDA-approved indications and even more off-label conditions that lead to non-GHD short stature. Biological constraints to GH action result in less dramatic and less consistent adult height growth responses to GH treatment in individuals who are already GH replete than those who are deficient, although short-term growth acceleration can be seen in both. While additional conditions that may benefit from GH treatment likely await discovery, current GH treatment can be improved through better diagnostics and better definition of treatment outcomes: more precisely identifying which patients have GHD and which patients will increase adult height with GH treatment, delineating the underlying causes of growth failure currently accepted as idiopathic, deciding whether short-term growth acceleration suffices to warrant treatment or increased adult height is required, and how much height gain justifies treatment (149). Likewise, future guidelines may consider rules for discontinuation of GH therapy in poor responders, to prevent broad-blanket denials in insurance coverage that would preclude the children who are responsive to GH therapy from the chance of benefiting. Both manufacturers and regulatory agencies are accepting newer long-acting GH preparations if they can demonstrate noninferiority to current daily recombinant GH products in short-term trials, though potential risks may require further study (150).
However, growing understanding of growth plate biology urges us to look beyond circulating hormone levels, the very definition of endocrinology, to cellular and molecular actors within the growth plate. Newer technologies like genomics, proteomics, and metabolomics and maybe even, one day, growth plate imaging, may empower us not only diagnostically but even therapeutically, veering us toward non-GH agents to more specifically address the underlying defect. For instance, recombinant IGF-I treatment is important for the small number of patients whose mechanisms of primary IGF-I deficiency render GH treatment ineffective (7). The real frontier lies in therapeutics targeting defects outside the GH/IGF system altogether. The first such therapeutic, a C-type natriuretic peptide analogue that positively regulates the signaling pathway downstream of the FGFR3 gene, was approved in 2021 for treating children with achondroplasia; the approval was based on a Phase III, placebo-controlled, 1-year trial (151), and adult height data are yet to be obtained.
In conclusion, while GH replacement for classical GHD remains a joy clinically, GH augmentation for non-GHD disorders results in some patients with growth responses comparable with those seen with GHD, in others with only partial or short-term responses conferring less impressive adult height gain, and in others still, negligible to nil responses. As seemingly more and more parents bring their children to the pediatric endocrinologist seeking GH treatment to achieve a predetermined height of their choosing, 20 years’ clinical experience following approval of the ISS indication and our increasing understanding of growth and GH biology have shown us that no, that is not always possible and yes, the underlying diagnosis does matter.
Abbreviations
- CRI
chronic renal insufficiency
- FDA
Food and Drug Administration
- GH
growth hormone
- GHD
growth hormone deficiency
- IGF
insulin-like growth factor
- IGFBP
insulin-like growth factor binding protein
- ISS
idiopathic short stature
- SDS
standard deviation score
- SGA
small for gestational age
- SHOX
short stature homeobox containing
Contributor Information
Adda Grimberg, Division of Endocrinology and Diabetes, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
Colin P Hawkes, Division of Endocrinology and Diabetes, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; INFANT Research Centre, University College Cork, Cork T12 DC4A, Ireland; Department of Paediatrics and Child Health, University College Cork, Cork T12 R229, Ireland.
Funding
A.G. is funded by NIH grant 1 R01 HD097129 from the Eunice Kennedy Shriver National Institute of Child Health and Human Development.
Disclosures
A.G. received the 2020 Growth Hormone Research Competitive Grant Program Award from Pfizer, Inc., and was consultant for educational symposia for medical staff of Pfizer, Inc and Ascendis Pharma. C.P.H. has no financial relationships relevant to this article to disclose.
Data Availability
Data sharing is not applicable to this article as no datasets were generated or analyzed during the current study.
References
- 1. Grimberg A, Allen DB. Growth hormone treatment for growth hormone deficiency and idiopathic short stature: new guidelines shaped by the presence and absence of evidence. Curr Opin Pediatr. 2017;29(4):466‐471. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Laron Z. The era of cadaveric pituitary extracted human growth hormone (1958–1985): biological and clinical aspects. Pediatr Endocrinol Rev. 2018;16(Suppl 1):11‐16. [DOI] [PubMed] [Google Scholar]
- 3. Ranke MB, Wit JM. Growth hormone - past, present and future. Nat Rev Endocrinol. 2018;14(5):285‐300. [DOI] [PubMed] [Google Scholar]
- 4. Wit JM, Kamp GA, Rikken B. Spontaneous growth and response to growth hormone treatment in children with growth hormone deficiency and idiopathic short stature. Pediatr Res. 1996;39(2):295‐302. [DOI] [PubMed] [Google Scholar]
- 5. Parks JS. Gene sequence and production of recombinant MetGH/hGH. Pediatr Endocrinol Rev. 2018;16(Suppl 1):17‐27. [DOI] [PubMed] [Google Scholar]
- 6. Kamoun C, Hawkes CP, Grimberg A. Provocative growth hormone testing in children: how did we get here and where do we go now? J Pediatr Endocrinol Metab. 2021;34(6):679‐696. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. 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(6):361‐397. [DOI] [PubMed] [Google Scholar]
- 8. Allen DB. Diagnosis of growth hormone deficiency remains a judgment call - and that is good. Horm Res Paediatr. 2021;94(11-12):406‐409. [DOI] [PubMed] [Google Scholar]
- 9. Collett-Solberg PF, Ambler G, Backeljauw PF, et al. Diagnosis, genetics, and therapy of short stature in children: a growth hormone research society international perspective. Horm Res Paediatr. 2019;92(1):1‐14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Hall SS. Size Matters: How Height Affects the Health, Happiness, and Success of Boys--and the Men They Become. Houghton Mifflin Company; 2006. [Google Scholar]
- 11. Backeljauw P, Cappa M, Kiess W, et al. Impact of short stature on quality of life: a systematic literature review. Growth Horm IGF Res. 2021;57–58:101392. [DOI] [PubMed] [Google Scholar]
- 12. Cutfield WS, Albert BB. Growth hormone treatment for idiopathic short stature. Pediatr Endocrinol Rev. 2018;16(Suppl 1):113‐122. [DOI] [PubMed] [Google Scholar]
- 13. Gardner M, Scerbak T, Sandberg DE. Psychosocial aspects of short stature and rhGH treatment: implicit trends over 60+ years. Pediatr Endocrinol Rev. 2018;16(Suppl 1):129‐141. [DOI] [PubMed] [Google Scholar]
- 14. Grimberg A, Cousounis P, Cucchiara AJ, Lipman TH, Ginsburg KR. Parental concerns influencing decisions to seek medical care for a child’s short stature. Horm Res Paediatr. 2015;84(5):338‐348. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Hitt T, Ginsburg KR, Cousounis P, et al. Concerns and expectations of parents seeking subspecialist care for their child’s short stature. Horm Res Paediatr. 2019;92(5):311‐318. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Allen DB. Clinical review: lessons learned from the hGH era. J Clin Endocrinol Metab. 2011;96(10):3042‐3047. [DOI] [PubMed] [Google Scholar]
- 17. Zgierska A, Rabago D, Miller MM. Impact of patient satisfaction ratings on physicians and clinical care. Patient Prefer Adherence. 2014;8:437‐446. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Albertsson-Wikland K, Aronson AS, Gustafsson J, et al. Dose-dependent effect of growth hormone on final height in children with short stature without growth hormone deficiency. J Clin Endocrinol Metab. 2008;93(11):4342‐4350. [DOI] [PubMed] [Google Scholar]
- 19. Pfaffle R, Land C, Schonau E, et al. Growth hormone treatment for short stature in the USA, Germany and France: 15 years of surveillance in the Genetics and Neuroendocrinology of Short-Stature International Study (GeNeSIS). Horm Res Paediatr. 2018;90(3):169‐180. [DOI] [PubMed] [Google Scholar]
- 20. Kamp GA, Waelkens JJ, de Muinck Keizer-Schrama SM, et al. High dose growth hormone treatment induces acceleration of skeletal maturation and an earlier onset of puberty in children with idiopathic short stature. Arch Dis Child. 2002;87(3):215‐220. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Darendeliler F, Ranke MB, Bakker B, et al. Bone age progression during the first year of growth hormone therapy in pre-pubertal children with idiopathic growth hormone deficiency, Turner syndrome or idiopathic short stature, and in short children born small for gestational age: analysis of data from KIGS (Pfizer International Growth Database). Horm Res. 2005;63(1):40‐47. [DOI] [PubMed] [Google Scholar]
- 22. Grimberg A, Kanter GP. US growth hormone use in the idiopathic short stature era: trends in insurer payments and patient financial burden. J Endocr Soc. 2019;3(11):2023‐2031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Kamoun C, Grimberg A. Disorders of growth. In: Soeiro RT, Devon EP, eds. Netter’s Pediatrics. Elsevier; 2022:305-312. [Google Scholar]
- 24. Jorge AA, Grimberg A, Dattani MT, Baron J. Disorders of childhood growth. In: Sperling MA, Majzoub JA, Menon RK, Stratakis CA, eds. Sperling: Pediatric Endocrinology. 5th ed. Elsevier Inc.; 2021:299‐356. [Google Scholar]
- 25. Li M. The origination of growth hormone/insulin-like growth factor system: a story from ancient basal chordate amphioxus. Front Endocrinol (Lausanne). 2022;13:825722. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Yakar S, Liu JL, Stannard B, et al. Normal growth and development in the absence of hepatic insulin-like growth factor I. Proc Natl Acad Sci U S A. 1999;96(13):7324‐7329. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Schlechter NL, Russell SM, Spencer EM, Nicoll CS. Evidence suggesting that the direct growth-promoting effect of growth hormone on cartilage in vivo is mediated by local production of somatomedin. Proc Natl Acad Sci U S A. 1986;83(20):7932‐7934. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Yakar S, Werner H, Rosen CJ. Insulin-like growth factors: actions on the skeleton. J Mol Endocrinol. 2018;61(1):T115‐T137. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Perez-Ibave DC, Rodriguez-Sanchez IP, Garza-Rodriguez Mde L, Barrera-Saldana HA. Extrapituitary growth hormone synthesis in humans. Growth Horm IGF Res. 2014;24(2-3):47‐53. [DOI] [PubMed] [Google Scholar]
- 30. Hochberg Z. Clinical physiology and pathology of the growth plate. Best Pract Res Clin Endocrinol Metab. 2002;16(3):399‐419. [DOI] [PubMed] [Google Scholar]
- 31. Hallett SA, Ono W, Ono N. The hypertrophic chondrocyte: to be or not to be. Histol Histopathol. 2021;36(10):1021‐1036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Angulo MA, Butler MG, Cataletto ME. Prader-Willi syndrome: a review of clinical, genetic, and endocrine findings. J Endocrinol Invest. 2015;38(12):1249‐1263. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. 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(11):4183‐4197. [DOI] [PubMed] [Google Scholar]
- 34. Grimberg A. Cardiovascular disease in former pediatric recipients of growth hormone: another look at growth hormone safety. JAMA Pediatr. 2021;175(2):e205232. [DOI] [PubMed] [Google Scholar]
- 35. Maghnie M, Ranke MB, Geffner ME, et al. Safety and efficacy of pediatric growth hormone therapy: results from the full KIGS cohort. J Clin Endocrinol Metab. 2022;107(12):3287‐3301. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Bell JJ, Lippe B, Romano AA, Cernich JT, Swinford RD, Moawad D. National cooperative growth study: 25 years of growth hormone data, insights, and lessons for future registries. Pediatr Endocrinol Rev. 2018;16(2):240‐255. [DOI] [PubMed] [Google Scholar]
- 37. Child CJ, Zimmermann AG, Chrousos GP, et al. Safety outcomes during pediatric GH therapy: final results from the prospective GeNeSIS observational program. J Clin Endocrinol Metab. 2019;104(2):379‐389. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Savendahl L, Polak M, Backeljauw P, et al. Treatment of children with GH in the United States and Europe: long-term follow-up from NordiNet(R) IOS and ANSWER program. J Clin Endocrinol Metab. 2019;104(10):4730‐4742. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Savendahl L, Polak M, Backeljauw P, et al. Long-term safety of growth hormone treatment in childhood: two large observational studies: NordiNet IOS and ANSWER. J Clin Endocrinol Metab. 2021;106(6):1728‐1741. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Centers for Disease Control and Prevention . Data Table of Stature-for-age Charts. 2001. Accessed March 19, 2023.https://www.cdc.gov/growthcharts/html_charts/statage.htm
- 41. Tanner JM, Goldstein H, Whitehouse RH. Standards for children’s height at ages 2–9 years allowing for heights of parents. Arch Dis Child. 1970;45(244):755‐762. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Braziuniene I, Wilson TA, Lane AH. Accuracy of self-reported height measurements in parents and its effect on mid-parental target height calculation. BMC Endocr Disord. 2007;7(1):2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Topor LS, Feldman HA, Bauchner H, Cohen LE. Variation in methods of predicting adult height for children with idiopathic short stature. Pediatrics. 2010;126(5):938‐944. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Cameron-Pimblett A, La Rosa C, King TFJ, Davies MC, Conway GS. The Turner syndrome life course project: karyotype-phenotype analyses across the lifespan. Clin Endocrinol (Oxf). 2017;87(5):532‐538. [DOI] [PubMed] [Google Scholar]
- 45. Clement-Jones M, Schiller S, Rao E, et al. The short stature homeobox gene SHOX is involved in skeletal abnormalities in Turner syndrome. Hum Mol Genet. 2000;9(5):695‐702. [DOI] [PubMed] [Google Scholar]
- 46. Marchini A, Marttila T, Winter A, et al. The short stature homeodomain protein SHOX induces cellular growth arrest and apoptosis and is expressed in human growth plate chondrocytes. J Biol Chem. 2004;279(35):37103‐37114. [DOI] [PubMed] [Google Scholar]
- 47. Marchini A, Ogata T, Rappold GA. A track record on SHOX: from basic research to complex models and therapy. Endocr Rev. 2016;37(4):417‐448. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Isojima T. Revision of the growth references for Japanese girls with Turner syndrome. Pediatr Endocrinol Rev. 2017;14(Suppl 1):240‐251. [DOI] [PubMed] [Google Scholar]
- 49. Ranke MB, Grauer ML. Adult height in Turner syndrome: results of a multinational survey 1993. Horm Res. 1994;42(3):90‐94. [DOI] [PubMed] [Google Scholar]
- 50. Sempe M, Hansson Bondallaz C, Limoni C. Growth curves in untreated Ullrich-Turner syndrome: French reference standards 1–22 years. Eur J Pediatr. 1996;155(10):862‐869. [DOI] [PubMed] [Google Scholar]
- 51. Stephure DK. Canadian Growth Hormone Advisory Committee. Impact of growth hormone supplementation on adult height in Turner syndrome: results of the Canadian randomized controlled trial. J Clin Endocrinol Metab. 2005;90(6):3360‐3366. [DOI] [PubMed] [Google Scholar]
- 52. Ross JL, Quigley CA, Cao D, et al. Growth hormone plus childhood low-dose estrogen in Turner’s Syndrome. N Engl J Med. 2011;364(13):1230‐1242. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. 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‐E1392. [DOI] [PubMed] [Google Scholar]
- 54. Limal JM, Parfait B, Cabrol S, et al. Noonan syndrome: relationships between genotype, growth, and growth factors. J Clin Endocrinol Metab. 2006;91(1):300‐306. [DOI] [PubMed] [Google Scholar]
- 55. Athota JP, Bhat M, Nampoothiri S, et al. Molecular and clinical studies in 107 Noonan syndrome affected individuals with PTPN11 mutations. BMC Med Genet. 2020;21(1):50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Ranke MB, Heidemann P, Knupfer C, Enders H, Schmaltz AA, Bierich JR. Noonan syndrome: growth and clinical manifestations in 144 cases. Eur J Pediatr. 1988;148(3):220‐227. [DOI] [PubMed] [Google Scholar]
- 57. Witt DR, Keena BA, Hall JG, Allanson JE. Growth curves for height in Noonan syndrome. Clin Genet. 1986;30(3):150‐153. [DOI] [PubMed] [Google Scholar]
- 58. Lee BH, Kim J-M, Jin HY, Kim G-H, Choi J-H, Yoo H-W. Spectrum of mutations in Noonan syndrome and their correlation with phenotypes. J Pediatr. 2011;159(6):1029‐1035. [DOI] [PubMed] [Google Scholar]
- 59. Rodriguez F, Gaete X, Cassorla F. Etiology and treatment of growth delay in Noonan syndrome. Front Endocrinol (Lausanne). 2021;12:691240. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Tanaka K, Sato A, Naito T, Kuramochi K, Itabashi H, Takemura Y. Noonan syndrome presenting growth hormone neurosecretory dysfunction. Intern Med. 1992;31(7):908‐911. [DOI] [PubMed] [Google Scholar]
- 61. Noordam C, van der Burgt I, Sweep CG, Delemarre-van de Waal HA, Sengers RC, Otten BJ. Growth hormone (GH) secretion in children with Noonan syndrome: frequently abnormal without consequences for growth or response to GH treatment. Clin Endocrinol (Oxf). 2001;54(1):53‐59. [DOI] [PubMed] [Google Scholar]
- 62. Romano AA, Blethen SL, Dana K, Noto RA. Growth hormone treatment in Noonan syndrome: the National Cooperative Growth Study experience. J Pediatr. 1996;128(5):S18‐S21. [DOI] [PubMed] [Google Scholar]
- 63. Tamburrino F, Gibertoni D, Rossi C, et al. Response to long-term growth hormone therapy in patients affected by RASopathies and growth hormone deficiency: patterns of growth, puberty and final height data. Am J Med Genet A. 2015;167A(11):2786‐2794. [DOI] [PubMed] [Google Scholar]
- 64. Libraro A, D’Ascanio V, Cappa M, et al. Growth in children with Noonan syndrome and effects of growth hormone treatment on adult height. Front Endocrinol (Lausanne). 2021;12:761171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Zadik Z, Chalew SA, Kowarski A. Assessment of growth hormone secretion in normal stature children using 24-hour integrated concentration of GH and pharmacological stimulation. J Clin Endocrinol Metab. 1990;71(4):932‐936. [DOI] [PubMed] [Google Scholar]
- 66. Marin G, Domene HM, Barnes KM, Blackwell BJ, Cassorla FG, Cutler GB Jr. 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):537‐541. [DOI] [PubMed] [Google Scholar]
- 67. 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 Endocrinol Metab. 1996;81(9):3323‐3327. [DOI] [PubMed] [Google Scholar]
- 68. Stofega MR, Herrington J, Billestrup N, Carter-Su C. Mutation of the SHP-2 binding site in growth hormone (GH) receptor prolongs GH-promoted tyrosyl phosphorylation of GH receptor, JAK2, and STAT5B. Mol Endocrinol. 2000;14(9):1338‐1350. [DOI] [PubMed] [Google Scholar]
- 69. De Rocca Serra-Nedelec A, Edouard T, Treguer K, et al. Noonan syndrome-causing SHP2 mutants inhibit insulin-like growth factor 1 release via growth hormone-induced ERK hyperactivation, which contributes to short stature. Proc Natl Acad Sci U S A. 2012;109(11):4257‐4262. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Binder G, Neuer K, Ranke MB, Wittekindt NE. PTPN11 Mutations are associated with mild growth hormone resistance in individuals with Noonan syndrome. J Clin Endocrinol Metab. 2005;90(9):5377‐5381. [DOI] [PubMed] [Google Scholar]
- 71. Bertelloni S, Baroncelli GI, Dati E, et al. IGF-I generation test in prepubertal children with Noonan syndrome due to mutations in the PTPN11 gene. Hormones (Athens). 2013;12(1):86‐92. [DOI] [PubMed] [Google Scholar]
- 72. Jorge AAL, Edouard T, Maghnie M, et al. Outcomes in growth hormone-treated Noonan syndrome children: impact of PTPN11 mutation status. Endocr Connect. 2022;11(4):e210615. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Giacomozzi C, Deodati A, Shaikh MG, Ahmed SF, Cianfarani S. The impact of growth hormone therapy on adult height in Noonan syndrome: a systematic review. Horm Res Paediatr. 2015;83(3):167‐176. [DOI] [PubMed] [Google Scholar]
- 74. Ozono K, Ogata T, Horikawa R, et al. Efficacy and safety of two doses of Norditropin® (somatropin) in short stature due to Noonan syndrome: a 2-year randomized, double-blind, multicenter trial in Japanese patients. Endocr J. 2018;65(2):159‐174. [DOI] [PubMed] [Google Scholar]
- 75. Siklar Z, Genens M, Poyrazoglu S, et al. The growth characteristics of patients with Noonan syndrome: results of three years of growth hormone treatment: a nationwide multicenter study. J Clin Res Pediatr Endocrinol. 2016;8(3):305‐312. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Horikawa R, Ogata T, Matsubara Y, et al. Long-term efficacy and safety of two doses of Norditropin((R)) (somatropin) in Noonan syndrome: a 4-year randomized, double-blind, multicenter trial in Japanese patients. Endocr J. 2020;67(8):803‐818. [DOI] [PubMed] [Google Scholar]
- 77. Lee PA, Ross JL, Pedersen BT, Kotnik P, Germak JA, Christesen HT. Noonan syndrome and Turner syndrome patients respond similarly to 4 years’ growth-hormone therapy: longitudinal analysis of growth-hormone-naive patients enrolled in the NordiNet® International Outcome Study and the ANSWER Program. Int J Pediatr Endocrinol. 2015;2015(1):17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. MacFarlane CE, Brown DC, Johnston LB, et al. Growth hormone therapy and growth in children with Noonan’s syndrome: results of 3 years’ follow-up. J Clin Endocrinol Metab. 2001;86(5):1953‐1956. [DOI] [PubMed] [Google Scholar]
- 79. Ranke MB, Lindberg A, Carlsson M, Camacho-Hubner C, Rooman R. Treatment with growth hormone in Noonan syndrome observed during 25 years of KIGS: near adult height and outcome prediction. Horm Res Paediatr. 2019;91(1):46‐55. [DOI] [PubMed] [Google Scholar]
- 80. Malaquias AC, Noronha RM, Souza TTO, et al. Impact of growth hormone therapy on adult height in patients with PTPN11 mutations related to Noonan syndrome. Horm Res Paediatr. 2019;91(4):252‐261. [DOI] [PubMed] [Google Scholar]
- 81. Seikaly MG, Salhab N, Gipson D, Yiu V, Stablein D. Stature in children with chronic kidney disease: analysis of NAPRTCS database. Pediatr Nephrol. 2006;21(6):793‐799. [DOI] [PubMed] [Google Scholar]
- 82. Wong CS, Gipson DS, Gillen DL, et al. Anthropometric measures and risk of death in children with end-stage renal disease. Am J Kidney Dis. 2000;36(4):811‐819. [DOI] [PubMed] [Google Scholar]
- 83. Brown DD, Dauber A. Growth hormone and insulin-like growth factor dysregulation in pediatric chronic kidney disease. Horm Res Paediatr. 2021;94(3-4):105‐114. [DOI] [PubMed] [Google Scholar]
- 84. Schaefer F, Veldhuis JD, Stanhope R, Jones J, Scharer K. Alterations in growth hormone secretion and clearance in peripubertal boys with chronic renal failure and after renal transplantation. Cooperative Study Group of Pubertal development in Chronic Renal Failure. J Clin Endocrinol Metab. 1994;78(6):1298‐1306. [DOI] [PubMed] [Google Scholar]
- 85. Haffner D, Schaefer F, Girard J, Ritz E, Mehls O. Metabolic clearance of recombinant human growth hormone in health and chronic renal failure. J Clin Invest. 1994;93(3):1163‐1171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Tonshoff B, Eden S, Weiser E, et al. Reduced hepatic growth hormone (GH) receptor gene expression and increased plasma GH binding protein in experimental uremia. Kidney Int. 1994;45(4):1085‐1092. [DOI] [PubMed] [Google Scholar]
- 87. Tonshoff B, Cronin MJ, Reichert M, et al. Reduced concentration of serum growth hormone (GH)-binding protein in children with chronic renal failure: correlation with GH insensitivity. The European Study Group for Nutritional Treatment of Chronic Renal Failure in Childhood. The German Study Group for Growth Hormone Treatment in Chronic Renal Failure. J Clin Endocrinol Metab. 1997;82(4):1007‐1013. [DOI] [PubMed] [Google Scholar]
- 88. Schaefer F, Chen Y, Tsao T, Nouri P, Rabkin R. Impaired JAK-STAT signal transduction contributes to growth hormone resistance in chronic uremia. J Clin Invest. 2001;108(3):467‐475. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Troib A, Landau D, Kachko L, Rabkin R, Segev Y. Epiphyseal growth plate growth hormone receptor signaling is decreased in chronic kidney disease-related growth retardation. Kidney Int. 2013;84(5):940‐949. [DOI] [PubMed] [Google Scholar]
- 90. Tonshoff B, Blum WF, Wingen AM, Mehls O. Serum insulin-like growth factors (IGFs) and IGF binding proteins 1, 2, and 3 in children with chronic renal failure: relationship to height and glomerular filtration rate. The European Study Group for Nutritional Treatment of Chronic Renal Failure in Childhood. J Clin Endocrinol Metab. 1995;80(9):2684‐2691. [DOI] [PubMed] [Google Scholar]
- 91. Blum WF. Insulin-like growth factors (IGFs) and IGF binding proteins in chronic renal failure: evidence for reduced secretion of IGFs. Acta Paediatr Scand Suppl. 1991;379(s379):24‐31; discussion 32. [DOI] [PubMed] [Google Scholar]
- 92. Haffner D, Tonshoff B, Blum WF, et al. Insulin-like growth factors (IGFs) and IGF binding proteins, serum acid-labile subunit and growth hormone binding protein in nephrotic children. Kidney Int. 1997;52(3):802‐810. [DOI] [PubMed] [Google Scholar]
- 93. Feld SM, Hirschberg R. Insulin-like growth factor-I and insulin-like growth factor-binding proteins in the nephrotic syndrome. Pediatr Nephrol. 1996;10(3):355‐358. [DOI] [PubMed] [Google Scholar]
- 94. Aldridge MK, Trnka P, Francis A, McTaggart SJ. Effectiveness of growth hormone on growth and final height in paediatric chronic kidney disease. Pediatr Nephrol. 2022;37(3):651‐658. [DOI] [PubMed] [Google Scholar]
- 95. Seikaly MG, Waber P, Warady BA, Stablein D. The effect of rhGH on height velocity and BMI in children with CKD: a report of the NAPRTCS registry. Pediatr Nephrol. 2009;24(9):1711‐1717. [DOI] [PubMed] [Google Scholar]
- 96. Haffner D, Schaefer F, Nissel R, Wuhl E, Tonshoff B, Mehls O. Effect of growth hormone treatment on the adult height of children with chronic renal failure. German Study Group for Growth Hormone Treatment in Chronic Renal Failure. N Engl J Med. 2000;343(13):923‐930. [DOI] [PubMed] [Google Scholar]
- 97. Hauer NN, Popp B, Schoeller E, et al. Clinical relevance of systematic phenotyping and exome sequencing in patients with short stature. Genet Med. 2018;20(6):630‐638. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98. Flechtner I, Lambot-Juhan K, Teissier R, et al. Unexpected high frequency of skeletal dysplasia in idiopathic short stature and small for gestational age patients. Eur J Endocrinol. 2014;170(5):677‐684. [DOI] [PubMed] [Google Scholar]
- 99. Freire BL, Homma TK, Funari MFA, et al. Multigene sequencing analysis of children born small for gestational age with isolated short stature. J Clin Endocrinol Metab. 2019;104(6):2023‐2030. [DOI] [PubMed] [Google Scholar]
- 100. Dauber A. Genetic testing for the child with short stature-has the time come to change our diagnostic paradigm? J Clin Endocrinol Metab. 2019;104(7):2766‐2769. [DOI] [PubMed] [Google Scholar]
- 101. Yang L, Zhang C, Wang W, et al. Pathogenic gene screening in 91 Chinese patients with short stature of unknown etiology with a targeted next-generation sequencing panel. BMC Med Genet. 2018;19(1):212. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102. Olivieri DJ, Massingham LJ, Schwab JL, Quintos JB. Lack of catch-up growth with growth hormone treatment in a child born small for gestational age leading to a diagnosis of Noonan syndrome with a pathogenic PTPN11 variant. Case Rep Endocrinol. 2021;2021:5571524. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Hawkes CP, Mostoufi-Moab S, McCormack SE, Grimberg A, Zemel BS. Leg length and sitting height reference data and charts for children in the United States. Data Brief. 2020;32:106131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Hawkes CP, Mostoufi-Moab S, McCormack SE, Grimberg A, Zemel BS. Sitting height to standing height ratio reference charts for children in the United States. J Pediatr. 2020;226:221‐227.e15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105. Del Pino M, Orden AB, Arenas MA, Fano V. Argentine references for the assessment of body proportions from birth to 17 years of age. Arch Argent Pediatr. 2017;115(3):234‐240. [DOI] [PubMed] [Google Scholar]
- 106. Zhang Y-Q, Li H. Reference charts of sitting height, leg length and body proportions for Chinese children aged 0–18 years. Ann Hum Biol. 2015;42(3):223‐230. [DOI] [PubMed] [Google Scholar]
- 107. Fredriks AM, van Buuren S, van Heel WJ, Dijkman-Neerincx RH, Verloove-Vanhorick SP, Wit JM. Nationwide age references for sitting height, leg length, and sitting height/height ratio, and their diagnostic value for disproportionate growth disorders. Arch Dis Child. 2005;90(8):807‐812. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. Clayton PE, Cianfarani S, Czernichow P, Johannsson G, Rapaport R, Rogol A. Management of the child born small for gestational age through to adulthood: a consensus statement of the International Societies of Pediatric Endocrinology and the Growth Hormone Research Society. J Clin Endocrinol Metab. 2007;92(3):804‐810. [DOI] [PubMed] [Google Scholar]
- 109. Ong KK, Ahmed ML, Emmett PM, Preece MA, Dunger DB. Association between postnatal catch-up growth and obesity in childhood: prospective cohort study. BMJ. 2000;320(7240):967‐971. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110. 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. [DOI] [PubMed] [Google Scholar]
- 111. Carel JC, Chatelain P, Rochiccioli P, Chaussain JL. Improvement in adult height after growth hormone treatment in adolescents with short stature born small for gestational age: results of a randomized controlled study. J Clin Endocrinol Metab. 2003;88(4):1587‐1593. [DOI] [PubMed] [Google Scholar]
- 112. Dahlgren J, Wikland KA; Swedish Study Group for Growth Hormone Treatment . Final height in short children born small for gestational age treated with growth hormone. Pediatr Res. 2005;57(2):216‐222. [DOI] [PubMed] [Google Scholar]
- 113. van Dijk M, Bannink EM, van Pareren YK, Mulder PG, Hokken-Koelega AC. Risk factors for diabetes mellitus type 2 and metabolic syndrome are comparable for previously growth hormone-treated young adults born small for gestational age (SGA) and untreated short SGA controls. J Clin Endocrinol Metab. 2007;92(1):160‐165. [DOI] [PubMed] [Google Scholar]
- 114. Van Pareren Y, Mulder P, Houdijk M, Jansen M, Reeser M, Hokken-Koelega A. 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. [DOI] [PubMed] [Google Scholar]
- 115. Maiorana A, Cianfarani S. Impact of growth hormone therapy on adult height of children born small for gestational age. Pediatrics. 2009;124(3):e519‐e531. [DOI] [PubMed] [Google Scholar]
- 116. Goedegebuure WJ, van der Steen M, Smeets CCJ, Hokken-Koelega ACS. Childhood growth hormone treatment and metabolic and cardiovascular risk in adults born small for gestational age after growth hormone cessation in the Netherlands: a 12-year follow-up study. Lancet Child Adolesc Health. 2022;6(11):777‐787. [DOI] [PubMed] [Google Scholar]
- 117. Upners EN, Raket LL, Petersen JH, et al. Timing of puberty, pubertal growth, and adult height in short children born small for gestational age treated with growth hormone. J Clin Endocrinol Metab. 2022;107(8):2286‐2295. [DOI] [PubMed] [Google Scholar]
- 118. Horikawa R, Tanaka T, Nishinaga H, Nishiba Y, Yokoya S. The long-term safety and effectiveness of growth hormone treatment in Japanese children with short stature born small for gestational age. Clin Pediatr Endocrinol. 2020;29(4):159‐171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119. Lee PA, Savendahl L, Oliver I, et al. Comparison of response to 2-years’ growth hormone treatment in children with isolated growth hormone deficiency, born small for gestational age, idiopathic short stature, or multiple pituitary hormone deficiency: combined results from two large observational studies. Int J Pediatr Endocrinol. 2012;2012(1):22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120. Leschek EW, Rose SR, Yanovski JA, et al. Effect of growth hormone treatment on adult height in peripubertal children with idiopathic short stature: a randomized, double-blind, placebo-controlled trial. J Clin Endocrinol Metab. 2004;89(7):3140‐3148. [DOI] [PubMed] [Google Scholar]
- 121. Sotos JF, Tokar NJ. Growth hormone significantly increases the adult height of children with idiopathic short stature: comparison of subgroups and benefit. Int J Pediatr Endocrinol. 2014;2014(1):15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122. Deodati A, Cianfarani S. Impact of growth hormone therapy on adult height of children with idiopathic short stature: systematic review. BMJ. 2011;342:c7157. [DOI] [PubMed] [Google Scholar]
- 123. Dos Santos WO, Wasinski F, Tavares MR, et al. Ablation of growth hormone receptor in GABAergic neurons leads to increased pulsatile growth hormone secretion. Endocrinology. 2022;163(8):bqac103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124. Gusmao DO, de Sousa ME, Tavares MR, Donato J. Increased GH secretion and body growth in mice carrying ablation of IGF-1 receptor in GH-releasing hormone cells. Endocrinology. 2022;163(11):bqac151. [DOI] [PubMed] [Google Scholar]
- 125. Flores-Morales A, Greenhalgh CJ, Norstedt G, Rico-Bautista E. Negative regulation of growth hormone receptor signaling. Mol Endocrinol. 2006;20(2):241‐253. [DOI] [PubMed] [Google Scholar]
- 126. Ram PA, Waxman DJ. SOCS/CIS protein inhibition of growth hormone-stimulated STAT5 signaling by multiple mechanisms. J Biol Chem. 1999;274(50):35553‐35561. [DOI] [PubMed] [Google Scholar]
- 127. Hashimoto Y, Kamioka T, Hosaka M, et al. Exogenous 20 K growth hormone (GH) suppresses endogenous 22 K GH secretion in normal men. J Clin Endocrinol Metab. 2000;85(2):601‐606. [DOI] [PubMed] [Google Scholar]
- 128. Jee YH, Baron J. The biology of stature. J Pediatr. 2016;173:32‐38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129. Weise M, De-Levi S, Barnes KM, Gafni RI, Abad V, Baron J. Effects of estrogen on growth plate senescence and epiphyseal fusion. Proc Natl Acad Sci U S A. 2001;98(12):6871‐6876. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130. Martin DD, Wit JM, Hochberg Z, et al. The use of bone age in clinical practice - part 1. Horm Res Paediatr. 2011;76(1):1‐9. [DOI] [PubMed] [Google Scholar]
- 131. de Zegher F, Albertsson-Wikland K, Wollmann HA, et al. Growth hormone treatment of short children born small for gestational age: growth responses with continuous and discontinuous regimens over 6 years. J Clin Endocrinol Metab. 2000;85(8):2816‐2821. [DOI] [PubMed] [Google Scholar]
- 132. de Zegher F, Butenandt O, Chatelain P, et al. Growth hormone treatment of short children born small for gestational age: reappraisal of the rate of bone maturation over 2 years and metanalysis of height gain over 4 years. Acta Paediatr Suppl. 1997;423(S423):207‐212. [DOI] [PubMed] [Google Scholar]
- 133. Beckers A, Petrossians P, Hanson J, Daly AF. The causes and consequences of pituitary gigantism. Nat Rev Endocrinol. 2018;14(12):705‐720. [DOI] [PubMed] [Google Scholar]
- 134. Baron J, Savendahl L, De Luca F, et al. Short and tall stature: a new paradigm emerges. Nat Rev Endocrinol. 2015;11(12):735‐746. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135. Ma S, Gladyshev VN. Molecular signatures of longevity: insights from cross-species comparative studies. Semin Cell Dev Biol. 2017;70:190‐203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136. Vitale G, Pellegrino G, Vollery M, Hofland LJ. Role of IGF-1 system in the modulation of longevity: controversies and new insights from a centenarians’ perspective. Front Endocrinol (Lausanne). 2019;10:27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137. 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. [DOI] [PubMed] [Google Scholar]
- 138. Guevara-Aguirre J, Balasubramanian P, Guevara-Aguirre M, et al. Growth hormone receptor deficiency is associated with a major reduction in pro-aging signaling, cancer, and diabetes in humans. Sci Transl Med. 2011;3(70):70ra13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139. Caputo M, Pigni S, Agosti E, et al. Regulation of GH and GH signaling by nutrients. Cells. 2021;10(6):1376. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140. Devesa J. The complex world of regulation of pituitary growth hormone secretion: the role of ghrelin, klotho, and nesfatins in it. Front Endocrinol (Lausanne). 2021;12:636403. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141. Birzniece V, Ho KKY. Mechanisms in endocrinology: paracrine and endocrine control of the growth hormone axis by estrogen. Eur J Endocrinol. 2021;184(6):R269‐R278. [DOI] [PubMed] [Google Scholar]
- 142. Werner H, Karnieli E, Rauscher FJ, LeRoith D. Wild-type and mutant p53 differentially regulate transcription of the insulin-like growth factor I receptor gene. Proc Natl Acad Sci U S A. 1996;93(16):8318‐8323. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143. Zhang L, Kashanchi F, Zhan Q, et al. Regulation of insulin-like growth factor II P3 promotor by p53: a potential mechanism for tumorigenesis. Cancer Res. 1996;56(6):1367‐1373. [PubMed] [Google Scholar]
- 144. Buckbinder L, Talbott R, Velasco-Miguel S, et al. Induction of the growth inhibitor IGF-binding protein 3 by p53. Nature. 1995;377(6550):646‐649. [DOI] [PubMed] [Google Scholar]
- 145. Grimberg A, Coleman CM, Shi Z, et al. Insulin-like growth factor factor binding protein-2 is a novel mediator of p53 inhibition of insulin-like growth factor signaling. Cancer Biol Ther. 2006;5(10):1408‐1414. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146. Chesnokova V, Zonis S, Apostolou A, et al. Local non-pituitary growth hormone is induced with aging and facilitates epithelial damage. Cell Rep. 2021;37(11):110068. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147. Boguszewski MCS, Boguszewski CL, Chemaitilly W, et al. Safety of growth hormone replacement in survivors of cancer and intracranial and pituitary tumours: a consensus statement. Eur J Endocrinol. 2022;186(6):P35‐P52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148. Allen DB, Backeljauw P, Bidlingmaier M, et al. GH safety workshop position paper: a critical appraisal of recombinant human GH therapy in children and adults. Eur J Endocrinol. 2015;174(2):P1‐P9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149. Halas JG, Grimberg A. Dilemmas of growth hormone treatment for GH deficiency and idiopathic short stature: defining, distinguishing, and deciding. Minerva Pediatr. 2020;72(3):206‐225. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150. Christiansen JS, Backeljauw PF, Bidlingmaier M, et al. Growth Hormone Research Society perspective on the development of long-acting growth hormone preparations. Eur J Endocrinol. 2016;174(6):C1‐C8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151. Savarirayan R, Tofts L, Irving M, et al. Once-daily, subcutaneous vosoritide therapy in children with achondroplasia: a randomised, double-blind, phase 3, placebo-controlled, multicentre trial. Lancet. 2020;396(10252):684‐692. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
Data sharing is not applicable to this article as no datasets were generated or analyzed during the current study.