Scolaris Content Display Scolaris Content Display

Tratamiento con hormona del crecimiento recombinante para la hipofosfatemia ligada al cromosoma X en niños

Contraer todo Desplegar todo

Antecedentes

El tratamiento convencional de la hipofosfatemia ligada al cromosoma X con fosfato y calcitriol oral puede curar el raquitismo, pero no siempre eleva de forma significativa las concentraciones de fosfato sérico, ni tampoco normaliza siempre el crecimiento lineal. Algunos ensayos clínicos indican que la combinación del tratamiento con hormona de crecimiento humana recombinante con el tratamiento convencional mejora la velocidad de crecimiento, la retención del fosfato y la densidad mineral ósea, pero algunos ensayos clínicos indican que parece agravar la estatura desproporcionada preexistente de estos niños. Esta es una versión actualizada de una revisión publicada anteriormente.

Objetivos

Determinar si el tratamiento con hormona de crecimiento humana recombinante en los niños con hipofosfatemia ligada al cromosoma X se asocia con cambios en el crecimiento longitudinal, el metabolismo mineral, la función endocrina, la función renal, la densidad mineral ósea, las proporciones corporales y también con cualquier efecto adverso.

Métodos de búsqueda

Se realizaron búsquedas en el Registro de ensayos del Grupo Cochrane de Fibrosis quística y enfermedades genéticas (Cochrane Cystic Fibrosis and Genetic Disorders Group), que incluye referencias identificadas a partir de búsquedas exhaustivas en bases de datos electrónicas, así como búsquedas manuales en revistas relevantes y en los libros de resúmenes de congresos. Además, se realizaron búsquedas en el Registro Cochrane central de ensayos controlados (Cochrane Central Register of Controlled Trials), en Ovid MEDLINE y en las listas de referencias de los ensayos identificados y de otras revisiones. También se realizaron algunas búsquedas manuales en revistas y resúmenes de congresos relevantes.

Fecha de la búsqueda más reciente: 12 de enero de 2021

Criterios de selección

Todos los estudios controlados aleatorizados o cuasialeatorizados que compararan la hormona de crecimiento (sola o combinada con el tratamiento convencional) con placebo o con el tratamiento convencional solo, en niños con hipofosfatemia ligada al cromosoma X.

Obtención y análisis de los datos

Dos autores, de forma independiente, evaluaron los estudios en cuanto al riesgo de sesgo y extrajeron los datos de los estudios elegibles. Se utilizó el método GRADE para evaluar la certeza de la evidencia de cada desenlace.

Resultados principales

En la revisión se incluyeron dos estudios (20 participantes). En un estudio cruzado (cross‐over), los resultados mostraron que el tratamiento con hormona de crecimiento humana recombinante podría mejorar la puntuación de la desviación estándar (DE) de altura (puntuación z), pero no se sabe si la intervención fue la razón de un aumento transitorio del fosfato sérico y del máximo tubular de reabsorción del fosfato. En el segundo estudio, paralelo, el tratamiento también podría haber mejorado la DE de la estatura desde el inicio en el grupo de rhGH en comparación con el grupo control, aunque no se observaron diferencias significativas entre los grupos después de tres años, DM 0,50 DE (IC del 95%: ‐0,54 a 1,54) (evidencia de certeza baja). El tratamiento posiblemente fue bien tolerado durante ambos estudios, con sólo efectos adversos transitorios observados en tres participantes (evidencia de certeza baja). No está claro si la hormona de crecimiento mejora los niveles de fosfato sérico o el cambio en la TmP/TFG (evidencia de certeza muy baja). El tratamiento podría dar lugar a poca o ninguna diferencia en los niveles de fosfatasa alcalina (evidencia de certeza baja).

Conclusiones de los autores

No se cuenta con suficiente evidencia de certeza alta para recomendar el uso del tratamiento con hormona de crecimiento humana recombinante en niños con hipofosfatemia ligada al cromosoma X.

PICO

Population
Intervention
Comparison
Outcome

El uso y la enseñanza del modelo PICO están muy extendidos en el ámbito de la atención sanitaria basada en la evidencia para formular preguntas y estrategias de búsqueda y para caracterizar estudios o metanálisis clínicos. PICO son las siglas en inglés de cuatro posibles componentes de una pregunta de investigación: paciente, población o problema; intervención; comparación; desenlace (outcome).

Para saber más sobre el uso del modelo PICO, puede consultar el Manual Cochrane.

Resumen en términos sencillos

Hormona de crecimiento humana sintética para el tratamiento de la hipofosfatemia ligada al cromosoma X (o raquitismo resistente a la vitamina D) en niños

Pregunta de la revisión

¿La hormona de crecimiento humana puede mejorar los desenlaces clínicos (como el aumento del crecimiento y la densidad mineral ósea, así como mejorar la función renal y hormonal) sin efectos secundarios en niños con hipofosfatemia ligada al cromosoma X?

Antecedentes

La hipofosfatemia ligada al cromosoma X es un trastorno genético que provoca niveles anormales de fosfato en el organismo. Esto puede dar lugar a una baja estatura y a raquitismo. El tratamiento estándar de la hipofosfatemia ligada al cromosoma X puede curar el raquitismo, pero no siempre eleva el nivel de fosfato en la sangre ni restituye los niveles de crecimiento a la normalidad. No está claro si la combinación del tratamiento con hormona de crecimiento humana con el tratamiento estándar mejora los niveles de fosfato, las tasas de crecimiento y la densidad mineral ósea.

Fecha de la búsqueda

La evidencia está actualizada hasta el: 12 de enero de 2021

Características de los estudios

En esta revisión se incluyeron dos estudios pequeños con un total de 20 niños de entre 2,5 y nueve años de edad. Había el mismo número de niños y niñas. Ambos ensayos fueron aleatorizados, de modo que los participantes tenían las mismas posibilidades de ser asignados al grupo de la hormona del crecimiento o al de control (los niños del grupo control no recibieron tratamiento adicional o recibieron un tratamiento placebo [simulado]). Un ensayo (paralelo) comparó a los niños a los que se les administró la hormona del crecimiento con los que no recibieron tratamiento durante tres años. El segundo ensayo fue un ensayo cruzado, de modo que, para empezar, un grupo de niños recibió el tratamiento con hormona de crecimiento humana y el segundo grupo recibió un placebo durante un año y luego los grupos recibieron el tratamiento opuesto durante otro año.

Resultados

El ensayo paralelo no encontró diferencias en las puntuaciones de estatura tras el tratamiento con hormona de crecimiento en comparación con ningún tratamiento adicional. No se observaron efectos secundarios graves en los ensayos. En el ensayo cruzado, el tratamiento con hormona de crecimiento humana mejoró la puntuación de la desviación estándar de la estatura (puntuación z) y aumentó temporalmente los niveles de fosfato en sangre.

No se encontró evidencia concluyente que muestre que el tratamiento con hormona de crecimiento humana funciona en esta enfermedad. No se han realizado suficientes estudios sobre el tratamiento con hormona del crecimiento humana en esta enfermedad y se necesitan más estudios de investigación. Esta es una versión actualizada de una revisión publicada anteriormente.

Calidad de la evidencia

En su mayoría, la certeza de la evidencia se consideró baja o muy baja. Esto se debe a que los ensayos eran muy pequeños y sólo incluían a unos pocos niños. También preocupaba que pudiera haber algún sesgo en los resultados debido al diseño del ensayo cruzado más pequeño.

Authors' conclusions

Implications for practice

The data are too few and of insufficient quality to provide recommendations for practice. Although the theory suggests that  recombinant human growth hormone (rhGH) may be beneficial to people with X‐linked hypophosphatemia (XLH), clinical use of rhGH in children with XLH cannot be supported on the basis of the two small included studies.

Implications for research

There have not been sufficient randomized controlled studies of rhGH therapy in children with XLH. We were unable to find any conclusive evidence to show whether the use of rhGH therapy in children with XLH is associated with changes in longitudinal growth, mineral metabolism, endocrine, renal function, bone mineral density, body proportions, and also with any adverse effects. We need more multicentre, adequately‐powered and well‐designed randomised controlled studies to address these important issues.

Summary of findings

Open in table viewer
Summary of findings 1. Summary of findings ‐ recombinant growth hormone compared with placebo or no intervention for children with X‐linked hypophosphataemia

Recombinant growth hormone compared with placebo/no intervention for X‐linked hypophosphataemia

Patient or population: children with X‐linked hypophosphataemia

Settings: outpatient

Intervention: rhGH (0.08 mg/kg/day given daily by SI (Seikaly 1997); 0.4 mg/kg/week by SI (Živičnjak 2011))

Comparison: placebo SI (Seikaly 1997) or no treatment (Živičnjak 2011)

Outcomes

Illustrative comparative risks* (95% CI)

Relative effect
(95% CI)

No of Participants
(studies)

Quality of the evidence
(GRADE)

Comments

Assumed risk

Corresponding risk

Control

rhGH

Longitudinal growth: change in height SDS

 

Follow‐up: 1 to 3 years

Both studies reported within‐group differences and found height SDS score to be significantly improved from baseline in the rhGH groups compared with no change in the control groups.

Between group analysis of the study results from the larger parallel study showed there to be no difference between groups after one year of treatment, MD 0.00 SDS (95 % CI ‐1.04 to 1.04) and a slight improvement at 2 years, MD 0.30 SDS (95 % CI ‐0.74 to 1.34) and 3 years, MD 0.50 SDS (95 % CI ‐0.54 to 1.54), but this was not statistically significant (Živičnjak 2011).

20
(2)

⊕⊕⊝⊝
low1,2

Due to risk of bias and imprecision

Both studies reported within‐group differences which may enhance the effects seen (Seikaly 1997Živičnjak 2011)

Measures of mineral metabolism: change in serum phosphate mmol/L

 

Follow‐up: 1 to 3 years

One study reported no significant change in serum phosphate levels in either group by the end of the study (Seikaly 1997).

Serum phosphate was significantly increased after 3 years in the second study, MD 0.18 mmol/L (95 % CI 0.08 to 0.28) (P = 0.0007) (Živičnjak 2011).

20
(2)

⊕⊝⊝⊝
very low1,2,3

Due to risk of bias, imprecision and inconsistency

The Seikaly studiy reports within‐group differences only.

Results appear to be opposing for this outcome but the Seikaly study was only reported at one year (Seikaly 1997) The Živičnjak study was 3 years duration and showed an effect at the three year time point (Živičnjak 2011).

Measures of endocrine function function: change in levels of alkaline phosphatase IU/L

 

Follow‐up: 1 to 3 years

No difference was reported in levels of alkaline phosphatase at the end of the studies (Seikaly 1997Živičnjak 2011).

20
(2)

⊕⊕⊝⊝
low1,2

 

There was a transient increase in ALP after one year of treatment compared to control in the Živičnjak study, MD 91.00 IU/L (95 % CI 25.21 to 156.79) (Živičnjak 2011)

Measures of renal function: change in TmP/GFR

 

Follow‐up: 1 to 3 years

There was no statistically significant difference in TmP/GFR between the rhGH and control groups at either 6 months, MD ‐0.05 mmol/L (95 % CI ‐0.15 to 0.05); 1 year, MD 0.01 mmol/L (95 % CI ‐0.16 to 0.18); 2 years, MD 0.12 mmol/L (95 % CI ‐0.14 to 0.38); or 3 years, MD 0.00 mmol/L (95 % CI ‐0.25 to 0.25) (Seikaly 1997Živičnjak 2011).

20
(2)

⊕⊝⊝⊝
very low1,2,4

Due to risk of bias, imprecision and publication bias

Both studies reported transient increases in TmP/GFR in the rhGH treated group compared to baseline (within‐group difference) (Seikaly 1997Živičnjak 2011). Seikaly found an increase in TmP/GFR at 3 months but not thereafter, whilst Živičnjak found a transient increase in the rhGH group and not in the control group.

 

No results were presented in the published paper for the control group in the Seikaly study (Seikaly 1997).

 

Body proportions: sitting height SDS

 

 

Follow‐up: 3 years

There was no difference in sitting height SDS after 1 year, MD ‐0.15 SDS (95 % CI ‐1.03 to 0.73) and a slight but non‐significant improvement in sitting height favouring the rhGH treated group after 3 years (Živičnjak 2011).

15
(1)

⊕⊕⊕⊝
moderate2

Due to imprecision

This outcome was only reported in one of the studies (Živičnjak 2011).

 

Adverse effects

 

Follow‐up: 1 to 3 years

rhGH was well tolerated in both studies with one study reporting three participants with adverse effects (Seikaly 1997Živičnjak 2011). One participant developed transient glucosuria that was not associated with hyperglycemia, one participant developed transient splenomegaly and one participant developed muscular protruberance.

The Živičnjak study reported that there were no side effects observed.

20
(2)

⊕⊕⊝⊝
low1,2

Due to risk of bias and imprecision

 

*The basis for the assumed risk (e.g. the median control group risk across studies) is provided in footnotes. The corresponding risk (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).
ALP: ; CI: confidence interval; RR: risk ratio; MD: mean difference; rhGH: recombinant human growth hormone; SDS: standard deviation score; SI: subcutaneous injection; TmP/GFR: ratio of the maximum rate of tubular phosphate reabsorption to the glomerular filtration rate

GRADE Working Group grades of evidence
High certainty: further research is very unlikely to change our confidence in the estimate of effect.
Moderate certainty: further research is likely to have an important impact on our confidence in the estimate of effect and may change the estimate.
Low certainty: further research is very likely to have an important impact on our confidence in the estimate of effect and is likely to change the estimate.
Very low certainty: we are very uncertain about the estimate.

1. High risk of bias due to one of the studies being a small cross‐over study where the results have been reported as if it were a parallel trial and where the risk of bias is unclear due to the randomisation process not being reported.

2. Imprecision due to very small numbers of participants in both studies.

3. Inconsistency due to studies reporting opposing results for this outcome

4. Selective reporting of results for this outcome as no data were given in the published paper for the control group

Background

Description of the condition

X‐linked hypophosphatemia (XLH) is an inherited disorder of phosphate homeostasis (internal balance) and is the commonest cause of inherited phosphate wasting. The incidence of XLH is 3.9 per 100,000 live births and the prevalence ranges from 1.7 per 100,000 children to 4.8 per 100,000 people (children and adults) (Haffner 2019). The condition is characterised by disproportionate short stature, rickets and osteomalacia, hypophosphatemia, abnormal phosphate reabsorption and disturbance of vitamin D metabolism (Rasmussen 1995). XLH is caused by mutations in genes, i.e. PHEX, which encodes a membrane‐bound endopeptidase. PHEX is expressed in bones and teeth but not in the kidneys and efforts are underway to clarify how PHEX function relates to the mutant phenotype (Jan de Beur 2002).

The diagnosis of XLH is based on a consistent medical history and physical examination, radiological evidence of rachitic disease, normal blood calcium levels, hypophosphatemia caused by selective renal phosphate wasting for which no other cause is found, a family history consistent with multigenerational occurrence of XLH, and demonstration of PHEX mutations (Dixon 1998); a significant number of sporadic cases (i.e. nonfamilial) appears to be due to de novo PHEX mutations.

Affected individuals have normal intelligence, but they can suffer significant limitations due to the short stature and bone deformities. Conventional treatment of XLH with oral phosphate and calcitriol can heal rickets, although it does not raise serum phosphate concentrations significantly, and it does not always normalize linear growth and many individuals fail to reach normal adult height (Friedman 1993). Furthermore, the phosphate therapy is often limited by adverse gastrointestinal symptoms (such as nausea, vomiting, and so on) and often leads to the development of hyperparathyroidism, and the high doses of calcitriol can lead to hypercalciuria (abnormally high concentration of calcium in the urine) and nephrocalcinosis (an abnormal condition in which calcium salts are deposited in kidney) (Goodyer 1987).

Description of the intervention

Growth hormone increases glomerular filtration rate and effective renal plasma flow in people with hypothalamic growth hormone deficiency (Hirschberg 1988) and it has been found to increase the rate of renal phosphate reabsorption in immature rats (Haramati 1990). 

How the intervention might work

Administration of growth hormone to hypophysectomized rats on a phosphate‐restricted diet results in a significant increase in 1,25‐dihydroxyvitamin D levels (Halloran 1988). In vivo recombinant human growth hormone (rhGH) treatment stimulates osteoblasts and activates bone remodelling (Brixen 1990), which suggest that growth hormone therapy may be beneficial in the treatment of people with XLH. Some observations that administration of exogenous growth hormone increases renal phosphate reabsorption and calcitriol levels (Brixen 1992Baroncelli 2001) have led to speculation regarding the role of growth hormone in phosphate homeostasis and the possible beneficial effects of growth hormone therapy in XLH.

Why it is important to do this review

Combining rhGH with conventional treatment offers theoretical advantages in poorly growing children with XLH. It is suggested that rhGH improves growth velocity, phosphate retention, and bone mineral density, but some studies suggest that it appears to aggravate the pre‐existent disproportionate stature of such children (Haffner 1995). In addition, rhGH is expensive, with the lifetime incremental cost of treating one child ranging from GBP 43,100 (USD 67,700) to GBP 53,400 (USD 83,900) (Bryant 2002). In light of this it is important to evaluate it's effectiveness and adverse effects. This is an updated version of a previously published review (Yang 2005).

Objectives

To determine whether the use of rhGH therapy in children with XLH is associated with changes in longitudinal growth, mineral metabolism, endocrine function, renal function, bone mineral density, body proportions, and also with any adverse effects.

Methods

Criteria for considering studies for this review

Types of studies

Randomized controlled stuides or quasi‐randomized controlled studies (both published and unpublished). Studies in which quasi‐randomized methods such as alternation were used wwould have been included if there was sufficient evidence that the treatment and control groups were similar at baseline.

Types of participants

Children (from 0 to 18 years old) with XLH defined by clinical, laboratory, or molecular criteria.

Types of interventions

Recombinant human growth hormone (given by subcutaneous injection, at any dose, any frequency and any duration) only or combined with conventional treatment (calcitriol and oral phosphate) compared with either placebo or conventional treatment alone.

Types of outcome measures

We planned to assess the following outcomes.

Primary outcomes

  1. Measures of longitudinal growth (growth velocity z score, height z score)

Secondary outcomes

  1. Measures of mineral metabolism (serum values of phosphate and calcium)

  2. Measures of endocrine function (serum values of 1,25‐dihydroxyvitamin D, insulin‐like growth factor I (IGF‐1), alkaline phosphatase, osteocalcin, growth hormone)

  3. Measures of renal function (urinary calcium to creatinine ratio, maximum rate of renal tubular reabsorption of phosphate normalized to the glomerular filtration rate (TmP/GFR))

  4. Bone mineral density

  5. Body proportions (sitting height z score, subischial leg length z score)

  6. Any adverse effects reported (e.g. transient glucosuria, transient splenomegaly and muscular prominence)

Search methods for identification of studies

Electronic searches

We searched the Cochrane Cystic Fibrosis and Genetic Disorders Group's Inborn Errors of Metabolism trials Register using the term: hypophosphatemia.

The Coagulopathies Trials Register is compiled from electronic searches of the Cochrane Central Register of Controlled Trials (CENTRAL) (updated each new issue of the Cochrane Library) and weekly searches of MEDLINE and the prospective handsearching of one journal ‐ Haemophilia. Unpublished work is identified by searching the abstract books of major conferences: the European Haematology Association conference; the American Society of Hematology conference; the British Society for Haematology Annual Scientific Meeting; the Congress of the World Federation of Hemophilia; the European Association for Haemophilia and Allied Disorders, the American Society of Gene and Cell Therapy and the International Society on Thrombosis and Haemostasis. For full details of all searching activities for the register, please see the relevant section of the Cochrane Cystic Fibrosis and Genetic Disorders Group's website.

Date of most recent search: 12 January 2021

We searched for trials on the Cochrane Central Register of Controlled Trials (searched 2011 to 18th January 2021) and Ovid MEDLINE 2011 to 18th January 2021 (searched 18th January 2021). For the full search strategies, please refer to the appendices (Appendix 1Appendix 2).

We also searched for trial registration entries on clinicaltrials.gov (https://www.clinicaltrials.gov/) and WHO ICTRP (https://www.who.int/clinical-trials-registry-platform). The search strategies used are given in an appendix (Appendix 3). Date of most recent search: 18th January 2021.

Searching other resources

The previous author team did not identify any further studies from the searches they carried out of the Journal of Bone and Mineral Research (1986 to 2003) and the proceedings of the American Society for Bone and Mineral Research Annual Meeting (1st to 24th). The original author team also contacted companies that market growth hormone for information on unpublished studies, however, they did not identify any studies. The current author team did not contact them again for this update.

Data collection and analysis

Selection of studies

Two authors (SS and TR for this update; previously YH and WC ) independently selected the studies to be included in the review. There was no disagreement on the suitability of the studies for inclusion.

Data extraction and management

Each author used standard data acquisition forms to independently extract data. We used Covidence to independently extract the data and then used the compare function to reach an agreed data extraction form (Covidence 2019). This form was electronically uploaded into Revman 5.

We planned to group outcome data into those measured at baseline, 6, 12, 18, 24 months and annually thereafter. If outcome data were recorded at other time periods, we would have considered examining those as well.

Assessment of risk of bias in included studies

We assessed all randomized controlled studies using the Cochrane 'Risk of bias’ tool described in the Cochrane Handbook for Systematic Reviews of Interventions (Higgins 2017). Two review authors (SS, TR) independently assessed each element of potential bias listed below as high, low, or unclear risk of bias:

  • selection bias (random sequence generation and allocation concealment);

  • performance bias (blinding of participants and personnel);

  • detection bias (blinding of outcome assessment);

  • attrition bias (incomplete outcome data);

  • reporting bias (selective reporting);

  • other bias.

We briefly described the rationale for the judgements of potential bias in the 'Characteristics of included studies' table. We ensured that a consensus on the degree of risk of bias was met through a comparison of the review authors' statements and where necessary, through consultation with a third person from the Cochrane Cystic Fibrosis and Genetic Disorders Group's editoral base.

Measures of treatment effect

For continuous outcomes, where possible we used the  mean change from baseline or post treatment values with standard deviations (SD) to calculate a summary statistic for the mean difference between groups with corresponding 95% confidence intervals (CIs); for the cross‐over trial, we describe the results in the text of the review. If future updates provide more studies presenting suitable data for analysis we will carry out a pooled estimate of effect by calculating the mean difference and 95 % confidence intervals (CIs).

If binary outcome data, e.g. adverse event data, are included in future updates of the review we will analyse these using risk ratio (RR) and 95 % CIs.

Where data were reported narratively in the original papers or the analysis wasn't appropriate to include in our own analyses, we have reported treatment effect narratively.

Unit of analysis issues

One of our included studies used a cross‐over design. Ideally when conducting a meta‐analysis combining results from cross‐over studies we would have liked to use the methods that are recommended by Elbourne (Elbourne 2002). However, due to restrictions on the available data, the only method that we would have been able to use would be to treat the cross‐over studies as if they were parallel trials. Elbourne says that this approach will produce conservative results as it does not take into account within‐patient correlation (Elbourne 2002). Also each participant will appear in both the treatment and control group, so the two groups will not be independent. Since we believe the data will have been analysed more appropriately by the primary authors, we have reported the results that were obtained from the primary paper and not entered the data from the cross‐over study into Data and analyses. For future updates of this review, when suitable data are available for inclusion, we will aim to use the most appropriate method available to analyse these data.

Dealing with missing data

We have reported the numbers and reasons for dropout or withdrawal from the studies in both the intervention and control groups. In order to allow an intention‐to‐treat analysis, we sought data on the number of participants by allocated treatment group, irrespective of compliance and whether or not the participant was later thought to be ineligible or otherwise excluded from treatment or follow‐up.

Assessment of heterogeneity

We have not been able to combine any outcome data in a meta‐analysis, but if this becomes possible in a future update we plan to quantify the impact of statistical and clinical heterogeneity in the meta‐analysis using a measure of the degree of inconsistency in the studies' results (Higgins 2017). This measure (I2) describes the percentage of total variation across studies that isdue to heterogeneity rather than chance (Higgins 2003). The values of I2 lie between 0% and 100%, and a simplified categorization of heterogeneity that we plan to use is of low (I2 value of 25%), moderate (I2 value of 50%), and high (I2 value of 75%) (Higgins 2017).

Assessment of reporting biases

If we had been able to include sufficient studies in the review (at least 10), we planned to look for asymmetry in a funnel plot for the primary outcome measures as an indication for the presence of publication bias. We did however describe any selective reporting by comparing the study protocols or methods sections with the results presented in the papers (or both).

Data synthesis

We were unable to combine study data into a meta‐analysis as outcomes were reported differently and at different timepoints in the two included studies. If we are able to combine data in a future update we plan to carry out a meta‐analysis taking into account the amount of heterogeneity present. Where heterogeneity is low we will use a fixed‐effects model (I2 less than 40 %), but if heterogeneity is greater than 40 % we will use a random‐effects model (Deeks 2021).

Subgroup analysis and investigation of heterogeneity

We were unable to carry out a subgroup analysis as we only had a small amount of data to add to the analyses. If in a future update we are able to carry out a meta‐analysis we will investigate possible causes of any heterogeneity by carrying out a subgroup analysis.

Sensitivity analysis

When possible, in future versions of this review, we plan to perform a sensitivity analysis based on the methodological quality of the studies by including and excluding studies that were at high risk of bias.

Summary of findings and assessment of the certainty of the evidence

In accordance with current Cochrane guidance we have included a summary of findings table for the comparison of rhGH versus control (summary of findings Table 1). We have selected the following outcomes to present in the summary of findings table based on their clinical importance.

  1. Longitudinal growth: height SDS

  2. Adverse effects

  3. Measures of mineral metabolism: serum phosphate

  4. Measures of endocrine function: alkaline phosphatase

  5. Measures of renal function TmP/GFR

  6. Body proportions: sitting height SDS

We used GRADE methodology to assess the quality of the evidence for each outcome based on the risk of bias within the studies, relevance to our population of interest (indirectness), unexplained heterogeneity or inconsistency (inconsistency), imprecision of the results or high risk of publication bias. The certainty of the evidence was deemed to be high before assessment. After assessment, the certainty of evidence was downgraded once for serious risk and twice for very serious risk.

Results

Description of studies

Results of the search

We have presented the results of our searches in a PRISMA diagram (Figure 1).


Study flow diagram.

Study flow diagram.

Eleven studies were identified from the searches (Baroncelli 2001Canete 2014Charron 2003Glorieux 1980NCT02720770Reusz 1997Rothenbuhler 2017Seikaly 1997Seikaly 2008Wilson 1991Živičnjak 2011). Two were included (Seikaly 1997Živičnjak 2011), and the remainder were excluded (Baroncelli 2001Canete 2014Charron 2003Glorieux 1980NCT02720770Reusz 1997Rothenbuhler 2017Seikaly 2008Wilson 1991).

Included studies

Only two studies met the predefined inclusion criteria and included 20 children (Seikaly 1997Živičnjak 2011).

Trial design

The Seikaly study is a randomised, double‐blind (both the participants and the physicians were blinded) cross‐over study, performed throughout a 24‐month period in five children with XLH (Seikaly 1997). The Živičnjak  study is a randomised controlled parallel group study carried out over three years in 15 children.

Participants

Both studies included small numbers of children with XLH; with a mean (standard error (SE)) age of 5.6 (1.4) years (range: 2.5 to 9.0 years) in the Seikaly study and mean (SD) age of 7.3 (1.7) years in the Živičnjak study. Both studies had a fairly equal gender split with 40% males in the Seikaly study and 47% males in the Živičnjak study (Seikaly 1997Živičnjak 2011). All participants were prepubertal at enrolment.

Interventions

In the Seikaly study, participants were randomized to receive either placebo or rhGH (0.08 mg/kg given daily by subcutaneous injection) for the first 12 months, after which the participants were crossed‐over to the other arm of the study for an additional 12 months. The dose was adjusted every three months according to changes in body weight. No washout period was described (Seikaly 1997).

In the Živičnjak study, participants in the intervention group were given 0.4 mg/kg body weight rhGH (Genotropin (Pfizer, Berlin, Germany)) via subcutaneous injection once a week for three years whilst the control group received no additional treatment and no placebo (Živičnjak 2011).

Outcomes

The primary outcome in both studies was height standard deviation score (SDS) against standardised scores for age and gender, although the Živičnjak study also reported on sitting height, arm length, leg length and sitting height index. Both studies also reported on TmP/GFR (Seikaly 1997Živičnjak 2011).

The Seikaly study additionally reported on mineral metabolism, glucose and lipid metabolism, hemoglobin, thyroid and parathyroid function, serum 1,25‐dihydroxyvitamin D, osteocalcin, growth hormone, urinary calcium, phosphate, nephrocalcinosis, renal function, and bone density after 12 months of either placebo or rhGH. They also measured fasting serum phosphate concentration after 3, 6, 9, and 12 months of either placebo control or rhGH therapy; changes in bone width, mass, and density after 12 months of either placebo control or rhGH therapy; biochemical data immediately and after 12 months of either placebo or rhGH therapy including: GH, IGF‐I, IGF‐BP3, TSH, T4, T3, 1,25‐dihydroxyvitamin D, calcium, alkaline phosphatase, urinary calcium to creatinine ratio, 24‐hour urinary albumin excretion and osteocalcin (Seikaly 1997).

Živičnjak also reported measures of mineral metabolism at three monthly intervals throughout the study including: serum calcium; serum phosphate; serum alkaline phosphatase activity; and serum IgF (Živičnjak 2011).

Excluded studies

We excluded 100 references on the basis of title and abstract alone and we have not described these any further. Of the remaining 11 studies, nine (11 references) were excluded after full‐text screening and reasons are given in the tables (Characteristics of excluded studies). Six studies were not randomised controlled studies (Baroncelli 2001Canete 2014NCT02720770Reusz 1997Rothenbuhler 2017Wilson 1991) and in three studies the treatment was not rhGH (Charron 2003Glorieux 1980Seikaly 2008).

Risk of bias in included studies

The risk of bias in the individual studies is presented in a figure (Figure 2).


Risk of bias summary: review authors' judgements about each risk of bias item for each included study.

Risk of bias summary: review authors' judgements about each risk of bias item for each included study.

Allocation

Generation of allocation sequence

In the Seikaly study, no information was provided about the generation of the randomization sequence and this was therefore judged as having an unclear risk of bias (Seikaly 1997). The Živičnjak study reported that randomisation was performed by an independent statistician via an in‐house computer program which randomised participants to the intervention and control arms in a 1:1 ratio and we judged the study to be at low risk of bias for this domain (Živičnjak 2011).

Concealment of allocation

In the Seikaly study, participants received a daily subcutaneous injection of placebo provided by Genetech during the blinded control period. The pharmacist assigned to the study prepared and dispensed blinded solutions. The pharmacist retained coded records of solutions dispensed. The codes were disclosed only at the end of the study and the pharmacist did not participate in the analysis of the data (Seikaly 1997). The method of allocation concealment was therefore judged to have a low risk of bias.

We also deemed the Živičnjak study to be at low risk of bias for this domain as randomisation was done via a computer program (Živičnjak 2011).

Blinding

The included cross‐over study was reported as double‐blind with a low risk of bias (Seikaly 1997). The parallel study was an open‐label study, so there was no blinding of participants, study personnel or outcome assessors (Živičnjak 2011). As the outcomes in this study are objective measures, we do not feel that being aware of treatment allocation will affect the results except for the outcome assessor. It is possible that knowledge of the intervention could affect how they carry out the measurements of height and limb length and therefore we feel that the risk of bias for this study and for this domain is unclear (Živičnjak 2011).

Incomplete outcome data

For the Seikaly study, there was no mention of withdrawals and no discussion of whether an intention‐to‐treat analysis had been performed, we therefore judged there to be an unclear risk of bias (Seikaly 1997). The Živičnjak study lost one participant to follow‐up in the second year of the study due to social reasons and they were removed from the analysis. The remaining 15 participants completed the study and we deemed the study to be at low risk of attrition bias (Živičnjak 2011).

Selective reporting

Both of our included studies were at low risk of reporting bias as outcome measures stated in the methods section were reported fully in the results (Seikaly 1997Živičnjak 2011). The Živičnjak study, however, only reported within‐group differences, which may have over or underrepresented the true effect of the treatment (Živičnjak 2011).

Other potential sources of bias

No other sources of bias were identified.

Effects of interventions

See: Summary of findings 1 Summary of findings ‐ recombinant growth hormone compared with placebo or no intervention for children with X‐linked hypophosphataemia

The certainty of the evidence has been graded for those outcomes included in the summary of findings table; for the definitions of these gradings, please refer to the summary of findings table (summary of findings Table 1).

As detailed within the 'Methods' section, we were unable to enter any data into the analyses from the cross‐over Seikaly study, as the information required to undertake the most appropriate analysis was not available in the primary paper (Seikaly 1997). We will request further information from the primary investigators and in the interim, have reported data qualitatively, not quantitatively. We have entered the results of the Živičnjak study into the analyses where appropriate (Data and analyses).

Primary outcomes

Please see the additional tables for a summary of the means and standard errors (SEs) reported (Table 1).

Open in table viewer
Table 1. Primary outcomes (mean (standard error))

longitudinal growth

baseline

3 months

12 months

height z score (treatment)

‐2.66 (0.21)

‐2.02 (0.25)

‐1.46 (0.28)

height z score (control)

‐2.27 (0.30)

‐2.22 (0.16)

growth velocity (treatment)

4.04 (1.50)

growth velocity (control)

‐1. 90 (0.40)

1. Measures of longitudinal growth
Height SDS

The Seikaly study reported within‐group differences and found that rhGH therapy improved the height SDS (z score) from a mean (SE) baseline of ‐2.66 (0.21) to ‐2.02 (0.25) P < 0.05, and to ‐1.46 (0.28) P < 0.05 after 3 and 12 months, respectively. At the start of the control period the height z score (mean (SE)) was ‐2.27 (0.30) compared with ‐2.22 (0.16) after 12 months of placebo administration. The growth velocity SDS (mean (SE)) was 4.04 (1.50) during the 12 months of rhGH therapy and ‐1. 90 (0.40) during the 12 months of placebo administration (Seikaly 1997).

The Živičnjak study also reports on within‐group differences and highlights an increase in height SDS of +1.1 SDS from baseline in the rhGH treated group compared to no significant change in the control group (P < 0.01). We entered the data into our analysis (using SEMs estimated from the plots provided in the paper) and found no difference in SDS between rhGH treated group and the control group at one year, MD 0.00 SDS (95 % CI ‐1.04 to 1.04) and a slight improvement in the rhGH treated group after two years, MD 0.30 SDS (95 % CI ‐0.74 to 1.34) and three years MD 0.50 SDS (95 % CI ‐0.54 to 1.54), but this was using endpoint values and therefore does not take account of baseline differences (Analysis 1.1) (Živičnjak 2011).

We graded the certainty of this evidence as low.

Secondary outcomes

Please see the additional tables for a summary of the means and SEs reported (Table 2).

Open in table viewer
Table 2. Secondary outcomes (mean (standard error))

Outcome

Baseline

3 months

12 months

Serum phosphate (mmol/L) (treatment)

0.88 (0.07)

1.17 (0.14)

Serum calcium (mmol/L) (treatment)

2.4 (0.1)

2.3 (0.1)

Serum calcium (mmol/L) (control)

2.3 (0.1)

2.3 (0.1)

Insulin‐like growth factor 1 (ng/mL) (treatment)

114.0 (25.0)

354.0 (51.0)

Insulin‐like growth factor 1 (ng/mL) (control)

224.0 (60.0)

157.0 (32.0)

Urinary calcium/creatinine ratio (treatment)

0.10 (0.06)

0.17 (0.02)

Urinary calcium/creatinine ratio (control)

0.15 (0.03)

0.20 (0.04)

Tubular maximum for phosphate reabsorption (mg/dL) (treatment)

2.12 (0.15)

3.41 (0.25)

24 hr urinary albumin excretion (mg) (treatment)

4.0 (1.0)

5.0 (2.0)

24 hr urinary albumin excretion (mg) (control)

2.0 (1.0)

3.0 (1.0)

Change of bone mass (g/cm) (treatment)

0.04 (0.02)

Change of bone mass (g/cm) (control)

0.02 (0.01)

Change of bone width (cm) (treatment)

0.18 (0.06)

Change of bone width (cm) (control)

0.02 (0.08)

Change of bone density (g/cm2) (treatment)

0.04 (0.06)

Change of bone density (g/cm2) (control)

0.01 (0.01)

1. Measures of mineral metabolism
Serum phosphate

Seikaly reported within‐group differences only and reported that mean (SE) serum phosphate increased from 0.88 (0.07) mmol/L to 1.17 (0.14) mmol/L after three months (P < 0.05); the changes from baseline were not statistically significant after 6, 9, and 12 months of rhGH therapy (P > 0.05). The change  from baseline in serum phosphate was also not statistically significant during the placebo period (P > 0.05) (Seikaly 1997). 

In the Živičnjak study serum phosphate was found to be significantly increased with rhGH treatment compared to the control group at the three‐year point, MD 0.18 mmol/L (95 % CI 0.08 to 0.28), although there was no difference between the groups at either the six months, MD 0.04 mmol/L (95 % CI ‐0.09 to 0.17); one year, MD 0.07 mmol/L (95 % CI ‐0.09 to 0.23); or two years, MD 0.09 mmol/L (95 % CI ‐0.13 to 0.31) (Analysis 1.2) (Živičnjak 2011). We graded the certainty of evidence as very low. 

Serum calcium

Both of the included studies reported only within‐group differences for serum calcium. In the cross‐over study the mean (SE) serum calcium level did not show a statistically significant change from baseline after 12 months of rhGH therapy, 2.4 (0.1) versus 2.3 (0.1) (P = 0.27), or after placebo 2.3 (0.1) versus 2.3 (0.1) (P = 0.90) (Seikaly 1997). 

In the parallel study, there was an increase in serum calcium in the control group at the two‐year timepoint compared to baseline (P < 0.05), but no change in the rhGH treated group at any time point (Živičnjak 2011).

2. Measures of endocrine function
Serum 1,25‐dihydroxyvitamin D

This outcome was only measured in the cross‐over study, which showed there to be no significant effect of either rhGH or placebo (Seikaly 1997).

IGF‐1

In the Seikaly study within‐group differences showed mean (SE) IGF‐1 increased from 114 (25) ng/mL to 354 (51) ng/mL after 12 months of rhGH therapy (P = 0.03), but did not exceed normal serum concentration. After 12 months, the mean (SE) IGF‐1 did not change significantly from baseline after rhGH treatment (224 (60) ng/mL) or after placebo administration (157 (32) ng/mL) (P = 0.32) (Seikaly 1997).

The Živičnjak study reported an increase in serum IGF‐1 in the GH‐treated group and in three out of the eight participants, the increase was greater than 2 SDS which was not seen in the control group. IGF levels were significantly higher in the GH treated group at all time points compared to control; six months MD 1.38 SDS (95 % CI 0.19 to 2.57), one year MD 1.95 SDS (95 % CI 0.97 to 2.93), two years MD 1.95 SDS (95 % CI 0.79 to 3.11), three years MD 2.70 SDS (95 % CI 1.29 to 4.11) (Analysis 1.4) (Živičnjak 2011).

Alkaline phosphatase

Neither therapy with rhGH nor with placebo had a statistically significant effect on alkaline phosphatase in the cross‐over study (within‐group differences only reported) (Seikaly 1997).

In the Živičnjak study, the authors reported a transient increase in alkaline phosphatase activity after one year of rhGH treatment which was significant compared to control, MD 91.00 IU/L (95 % CI 25.21 to 156.79). There was no difference between groups at two years or three years, MD 16 IU/L (95 % CI ‐80.86 to 112.86) and MD 25 IU/L (95 % CI ‐105.06 to 155.06), respectively (Analysis 1.3) (Živičnjak 2011). We graded the certainty of this evidence as low.

Osteocalcin and growth hormone

The Seikaly study reported no statistically significant effect of either rhGH or placebo on osteocalcin or growth hormone (within‐group differences) (P > 0.05) (Seikaly 1997)

The Živičnjak study did not measure either osteocalcin or growth hormone.

3. Measures of renal function
Urinary calcium to creatinine ratio

Neither of our included studies reported a significant change in urinary calcium to creatinine ratio in either the rhGH treated or control groups (within‐group differences only). In the Seikaly study urinary calcium to creatinine ratio did not change significantly after 12 months of placebo, mean (SE) 0.15 (0.03) versus 0.20 (0.04) (P = 0.35) or rhGH therapy, 0.10 (0.06) versus 0.17 (0.02) (P = 0.28) (Seikaly 1997). Živičnjak found no significant change in urinary calcium/creatinine ratio in either the rhGH or control group (Živičnjak 2011).

Maximum rate of TmP/GFR

In the cross‐over Seikaly study (within‐group differences only), mean (SE) TmP/GFR increased from  2.12 (0.15) mg/dL to 3.41 (0.25) mg/dL after three months, but the changes after 6, 9, and 12 months of rhGH therapy were not statistically significant from baseline. The TmP/GFR did not change significantly from baseline during the placebo administration, but the actual figures were not reported in the paper (Seikaly 1997).

Živičnjak found a transient increase in TmP/GFR in the rhGH treated group but not in the control group. When we entered the study data into our analysis there was no statistically significant difference in TmP/GFR between the rhGH and control groups at either six months, MD ‐0.05 mmol/L (95 % CI ‐0.15 to 0.05); one year, MD 0.01 mmol/L (95 % CI ‐0.16 to 0.18); two years, MD 0.12 mmol/L (95 % CI ‐0.14 to 0.38); or three years, MD 0.00 mmol/L (95 % CI ‐0.25 to 0.25) (Analysis 1.5) (Živičnjak 2011). We assessed the certainty of this evidence is very low.

Urinary albumin excretion

In the Seikaly study, after 12 months therapy the mean (SE) 24‐hour urinary albumin excretion level did not change significantly from baseline with rhGH, 4 (1) mg versus 5 (2) mg (P = 0.63) or with placebo 2 (1) mg versus 3 (1) mg (P = 0.50) (within‐group differences) (Seikaly 1997).

This outcome was not measured in the parallel Živičnjak study (Živičnjak 2011).

4. Bone mineral density

Both studies reported on a measure of bone density. Seikaly reported within‐group differences and showed that after 12 months of rhGH therapy there was a mean (SE) increase from baseline in bone mass 0.04 (0.02) g/cm (P = 0.049) and width, 0.18 (0.06) cm (P = 0.049), but not density, 0.04 (0.06) g/cm2 (P = 0.220).  In the placebo group there was no statistically significant change from baseline on bone mass ( P = 0.12), width (P = 0.70), or density (P = 0.07) after 12 months. The authors did not carry out a between‐group analysis (Seikaly 1997).

Živičnjak reported that mean (SD) bone age tended to increase more rapidly in rhGH participants (3.6 (5) years) than in control participants (3.1 (5) years) per calendar year (P = 0.18), MD 0.5 years (95 % CI ‐0.01 to 1.01) (Analysis 1.6) (Živičnjak 2011).

5. Body proportions: sitting height z score or subischial leg length z score

These outcomes were not reported in the Seikaly study (Seikaly 1997). The Živičnjak study reported on sitting height, arm length, leg length and sitting height index (Živičnjak 2011).

Sitting height SDS

Results from the Živičnjak study showed that after one year there was no difference in sitting height SDS score between the rhGH‐treated group and the control group, MD ‐0.15 SDS (95 % CI ‐1.03 to 0.73). There was a slight improvement favouring the rhGH treated group at the end of year two, MD 0.40 SDS (95 % CI ‐0.37 to 1.17) and year three, MD 0.35 SDS (95 % CI ‐0.14 to 0.84), but these were not statistically significant (Analysis 1.7) (Živičnjak 2011). We graded the certainty of the evidence for this outcome as moderate.

Arm length SDS

Živičnjak found no difference between the intervention group and control group at any time point: at one year, MD 0.00 SDS (95 % CI ‐0.69 to 0.69); two years, MD 0.25 SDS (95 % CI ‐0.58, to 1.08); or three years, MD 0.15 SDS (95 % CI ‐0.69 to 0.99) (Analysis 1.8) (Živičnjak 2011).

Leg length SDS

Results from the Živičnjak study showed no difference in SDS between the rhGH‐treated and control groups at any time point: one year, MD ‐0.20 SDS (95 % CI ‐0.75 to 0.35); two years, MD 0.20 SDS (95 % CI ‐0.78 to 1.18); and three years, MD 0.25 SDS (95 % CI ‐0.93 to 1.43) (Analysis 1.9) (Živičnjak 2011).

Sitting height index

The Živičnjak study measured change in sitting height index from baseline but did not report the SE of the change and so we have not entered this into an analysis. Mean sitting height index increased slightly in both rhGH‐treated and control group participants, but results were not statistically significant, 0.4 SDS versus 0.6 SDS (GH versus control) (P = 0.43) (Živičnjak 2011).

6. Adverse effects

In the Seikaly cross‐over study (n = 5), one participant developed transient glucosuria that was not associated with hyperglycemia (abnormally high concentration of glucose in the blood), one participant developed transient splenomegaly (enlargement of the spleen), and one participant developed muscular prominence (protuberance) (Seikaly 1997).

In the Živičnjak parallel study the authors reported that rhGH was well‐tolerated and no severe side effects were observed (Živičnjak 2011).

Discussion

Summary of main results

Two studies met the inclusion criteria for this review with 20 children participating. One study was a cross‐over study with only five participants (Seikaly 1997) and the second study was a three‐year parallel study involving 15 participants (Živičnjak 2011). Both studies reported within‐group differences from baseline and found the height SDS score to be significantly improved from baseline in the groups receiving rhGH. Our analyses found there to be no difference in height SDS between the rhGH group and the control group at three years (low‐certainty evidence). The treatment was well‐tolerated during both studies with only transient adverse effects seen in three participants from the small cross‐over study (low‐certainty evidence). RhGH may slightly improve height but again, the certainty of evidence is low. We are uncertain whether growth hormone improves serum phosphate levels or change in TmP/GFR (very low‐certainty evidence). The treatment may make little or no difference to alkaline phosphatase levels (low‐certainty evidence).

Overall completeness and applicability of evidence

We were only able to include two studies with a very small number of participants which limits the completeness of the evidence for rhGH. The studies were different in design and duration, one study being a cross‐over design carried out over one year (Seikaly 1997) and the other a parallel study which ran for three years (Živičnjak 2011). This means that we were unable to combine data in meta‐analyses and reported the results narratively. The Seikaly study used a cross‐over design in five participants and it is unclear whether there was a difference at baseline between those participants that were randomised to rhGH first and those that were randomised to placebo first. The studies used a different dose of rhGH given to a different time schedule; Seikaly used 0.08 mg/kg/day given daily (Seikaly 2008) whilst the Živičnjak study used 0.4 mg/kg per week by subcutaneous injection (Živičnjak 2011). Both of the studies reported only within‐group differences which may have over or underestimated the true effect of rhGH.

Quality of the evidence

The certainty of the evidence across outcomes ranged from very low to moderate and was largely downgraded due to the very small sample sizes, risk of bias within the study design and inconsistency. The cross‐over study did not report their methods of randomisation and so the risk of bias for that domain was deemed to be unclear. Blinding to the intervention was not possible in the Živičnjak study as the control was 'no treatment'. We do not, however, feel that this would have affected the outcomes which were objective measures or laboratory tests, where knowledge of the intervention would have little impact.

Potential biases in the review process

We are confident that we have identified all the eligible studies from the comprehensive searches we carried out. Data were extracted using Covidence which allowed comparison of independent data extraction forms and the resulting forms were imported electronically into RevMan to reduce data entry errors.

Agreements and disagreements with other studies or reviews

We were unable to find any other randomised controlled studies looking at the effects of rhGH in children with XLH which may be due to the linear nature of growth and the difficulties in ensuring groups are similar at baseline. A cohort study of 19 children with XLH who were given 67 mcg/kg/day of rhGH (adjusted every three months) for two years reported a significant improvement in height SDS after one (P = 0.01) and two years (P = 0.04) (Rothenbuhler  2017). They also found a strong correlation between the age at onset of treatment and the number of centimetres gained with prepubertal children responding better. Without a comparison group however, it is not possible to say whether these results would have occurred without rhGH treatment.

Study flow diagram.

Figuras y tablas -
Figure 1

Study flow diagram.

Risk of bias summary: review authors' judgements about each risk of bias item for each included study.

Figuras y tablas -
Figure 2

Risk of bias summary: review authors' judgements about each risk of bias item for each included study.

Comparison 1: Growth hormone (GH) versus control (no treatment), Outcome 1: Height (SDS)

Figuras y tablas -
Analysis 1.1

Comparison 1: Growth hormone (GH) versus control (no treatment), Outcome 1: Height (SDS)

Comparison 1: Growth hormone (GH) versus control (no treatment), Outcome 2: Measures of mineral metabolism: serum phosphate (mmol/L)

Figuras y tablas -
Analysis 1.2

Comparison 1: Growth hormone (GH) versus control (no treatment), Outcome 2: Measures of mineral metabolism: serum phosphate (mmol/L)

Comparison 1: Growth hormone (GH) versus control (no treatment), Outcome 3: Measures of endocrine function: serum alkaline phosphatase activity (IU/L)

Figuras y tablas -
Analysis 1.3

Comparison 1: Growth hormone (GH) versus control (no treatment), Outcome 3: Measures of endocrine function: serum alkaline phosphatase activity (IU/L)

Comparison 1: Growth hormone (GH) versus control (no treatment), Outcome 4: Measures of endocrine function: serum IGF (SDS)

Figuras y tablas -
Analysis 1.4

Comparison 1: Growth hormone (GH) versus control (no treatment), Outcome 4: Measures of endocrine function: serum IGF (SDS)

Comparison 1: Growth hormone (GH) versus control (no treatment), Outcome 5: Measures of renal function: TmP/GFR (mmol/L)

Figuras y tablas -
Analysis 1.5

Comparison 1: Growth hormone (GH) versus control (no treatment), Outcome 5: Measures of renal function: TmP/GFR (mmol/L)

Comparison 1: Growth hormone (GH) versus control (no treatment), Outcome 6: BMD: determination of bone age (years) (change from baseline)

Figuras y tablas -
Analysis 1.6

Comparison 1: Growth hormone (GH) versus control (no treatment), Outcome 6: BMD: determination of bone age (years) (change from baseline)

Comparison 1: Growth hormone (GH) versus control (no treatment), Outcome 7: Body proportions: sitting height (SDS)

Figuras y tablas -
Analysis 1.7

Comparison 1: Growth hormone (GH) versus control (no treatment), Outcome 7: Body proportions: sitting height (SDS)

Comparison 1: Growth hormone (GH) versus control (no treatment), Outcome 8: Body proportions: arm length (SDS)

Figuras y tablas -
Analysis 1.8

Comparison 1: Growth hormone (GH) versus control (no treatment), Outcome 8: Body proportions: arm length (SDS)

Comparison 1: Growth hormone (GH) versus control (no treatment), Outcome 9: Body proportions: leg length (SDS)

Figuras y tablas -
Analysis 1.9

Comparison 1: Growth hormone (GH) versus control (no treatment), Outcome 9: Body proportions: leg length (SDS)

Summary of findings 1. Summary of findings ‐ recombinant growth hormone compared with placebo or no intervention for children with X‐linked hypophosphataemia

Recombinant growth hormone compared with placebo/no intervention for X‐linked hypophosphataemia

Patient or population: children with X‐linked hypophosphataemia

Settings: outpatient

Intervention: rhGH (0.08 mg/kg/day given daily by SI (Seikaly 1997); 0.4 mg/kg/week by SI (Živičnjak 2011))

Comparison: placebo SI (Seikaly 1997) or no treatment (Živičnjak 2011)

Outcomes

Illustrative comparative risks* (95% CI)

Relative effect
(95% CI)

No of Participants
(studies)

Quality of the evidence
(GRADE)

Comments

Assumed risk

Corresponding risk

Control

rhGH

Longitudinal growth: change in height SDS

 

Follow‐up: 1 to 3 years

Both studies reported within‐group differences and found height SDS score to be significantly improved from baseline in the rhGH groups compared with no change in the control groups.

Between group analysis of the study results from the larger parallel study showed there to be no difference between groups after one year of treatment, MD 0.00 SDS (95 % CI ‐1.04 to 1.04) and a slight improvement at 2 years, MD 0.30 SDS (95 % CI ‐0.74 to 1.34) and 3 years, MD 0.50 SDS (95 % CI ‐0.54 to 1.54), but this was not statistically significant (Živičnjak 2011).

20
(2)

⊕⊕⊝⊝
low1,2

Due to risk of bias and imprecision

Both studies reported within‐group differences which may enhance the effects seen (Seikaly 1997Živičnjak 2011)

Measures of mineral metabolism: change in serum phosphate mmol/L

 

Follow‐up: 1 to 3 years

One study reported no significant change in serum phosphate levels in either group by the end of the study (Seikaly 1997).

Serum phosphate was significantly increased after 3 years in the second study, MD 0.18 mmol/L (95 % CI 0.08 to 0.28) (P = 0.0007) (Živičnjak 2011).

20
(2)

⊕⊝⊝⊝
very low1,2,3

Due to risk of bias, imprecision and inconsistency

The Seikaly studiy reports within‐group differences only.

Results appear to be opposing for this outcome but the Seikaly study was only reported at one year (Seikaly 1997) The Živičnjak study was 3 years duration and showed an effect at the three year time point (Živičnjak 2011).

Measures of endocrine function function: change in levels of alkaline phosphatase IU/L

 

Follow‐up: 1 to 3 years

No difference was reported in levels of alkaline phosphatase at the end of the studies (Seikaly 1997Živičnjak 2011).

20
(2)

⊕⊕⊝⊝
low1,2

 

There was a transient increase in ALP after one year of treatment compared to control in the Živičnjak study, MD 91.00 IU/L (95 % CI 25.21 to 156.79) (Živičnjak 2011)

Measures of renal function: change in TmP/GFR

 

Follow‐up: 1 to 3 years

There was no statistically significant difference in TmP/GFR between the rhGH and control groups at either 6 months, MD ‐0.05 mmol/L (95 % CI ‐0.15 to 0.05); 1 year, MD 0.01 mmol/L (95 % CI ‐0.16 to 0.18); 2 years, MD 0.12 mmol/L (95 % CI ‐0.14 to 0.38); or 3 years, MD 0.00 mmol/L (95 % CI ‐0.25 to 0.25) (Seikaly 1997Živičnjak 2011).

20
(2)

⊕⊝⊝⊝
very low1,2,4

Due to risk of bias, imprecision and publication bias

Both studies reported transient increases in TmP/GFR in the rhGH treated group compared to baseline (within‐group difference) (Seikaly 1997Živičnjak 2011). Seikaly found an increase in TmP/GFR at 3 months but not thereafter, whilst Živičnjak found a transient increase in the rhGH group and not in the control group.

 

No results were presented in the published paper for the control group in the Seikaly study (Seikaly 1997).

 

Body proportions: sitting height SDS

 

 

Follow‐up: 3 years

There was no difference in sitting height SDS after 1 year, MD ‐0.15 SDS (95 % CI ‐1.03 to 0.73) and a slight but non‐significant improvement in sitting height favouring the rhGH treated group after 3 years (Živičnjak 2011).

15
(1)

⊕⊕⊕⊝
moderate2

Due to imprecision

This outcome was only reported in one of the studies (Živičnjak 2011).

 

Adverse effects

 

Follow‐up: 1 to 3 years

rhGH was well tolerated in both studies with one study reporting three participants with adverse effects (Seikaly 1997Živičnjak 2011). One participant developed transient glucosuria that was not associated with hyperglycemia, one participant developed transient splenomegaly and one participant developed muscular protruberance.

The Živičnjak study reported that there were no side effects observed.

20
(2)

⊕⊕⊝⊝
low1,2

Due to risk of bias and imprecision

 

*The basis for the assumed risk (e.g. the median control group risk across studies) is provided in footnotes. The corresponding risk (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).
ALP: ; CI: confidence interval; RR: risk ratio; MD: mean difference; rhGH: recombinant human growth hormone; SDS: standard deviation score; SI: subcutaneous injection; TmP/GFR: ratio of the maximum rate of tubular phosphate reabsorption to the glomerular filtration rate

GRADE Working Group grades of evidence
High certainty: further research is very unlikely to change our confidence in the estimate of effect.
Moderate certainty: further research is likely to have an important impact on our confidence in the estimate of effect and may change the estimate.
Low certainty: further research is very likely to have an important impact on our confidence in the estimate of effect and is likely to change the estimate.
Very low certainty: we are very uncertain about the estimate.

1. High risk of bias due to one of the studies being a small cross‐over study where the results have been reported as if it were a parallel trial and where the risk of bias is unclear due to the randomisation process not being reported.

2. Imprecision due to very small numbers of participants in both studies.

3. Inconsistency due to studies reporting opposing results for this outcome

4. Selective reporting of results for this outcome as no data were given in the published paper for the control group

Figuras y tablas -
Summary of findings 1. Summary of findings ‐ recombinant growth hormone compared with placebo or no intervention for children with X‐linked hypophosphataemia
Table 1. Primary outcomes (mean (standard error))

longitudinal growth

baseline

3 months

12 months

height z score (treatment)

‐2.66 (0.21)

‐2.02 (0.25)

‐1.46 (0.28)

height z score (control)

‐2.27 (0.30)

‐2.22 (0.16)

growth velocity (treatment)

4.04 (1.50)

growth velocity (control)

‐1. 90 (0.40)

Figuras y tablas -
Table 1. Primary outcomes (mean (standard error))
Table 2. Secondary outcomes (mean (standard error))

Outcome

Baseline

3 months

12 months

Serum phosphate (mmol/L) (treatment)

0.88 (0.07)

1.17 (0.14)

Serum calcium (mmol/L) (treatment)

2.4 (0.1)

2.3 (0.1)

Serum calcium (mmol/L) (control)

2.3 (0.1)

2.3 (0.1)

Insulin‐like growth factor 1 (ng/mL) (treatment)

114.0 (25.0)

354.0 (51.0)

Insulin‐like growth factor 1 (ng/mL) (control)

224.0 (60.0)

157.0 (32.0)

Urinary calcium/creatinine ratio (treatment)

0.10 (0.06)

0.17 (0.02)

Urinary calcium/creatinine ratio (control)

0.15 (0.03)

0.20 (0.04)

Tubular maximum for phosphate reabsorption (mg/dL) (treatment)

2.12 (0.15)

3.41 (0.25)

24 hr urinary albumin excretion (mg) (treatment)

4.0 (1.0)

5.0 (2.0)

24 hr urinary albumin excretion (mg) (control)

2.0 (1.0)

3.0 (1.0)

Change of bone mass (g/cm) (treatment)

0.04 (0.02)

Change of bone mass (g/cm) (control)

0.02 (0.01)

Change of bone width (cm) (treatment)

0.18 (0.06)

Change of bone width (cm) (control)

0.02 (0.08)

Change of bone density (g/cm2) (treatment)

0.04 (0.06)

Change of bone density (g/cm2) (control)

0.01 (0.01)

Figuras y tablas -
Table 2. Secondary outcomes (mean (standard error))
Comparison 1. Growth hormone (GH) versus control (no treatment)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1.1 Height (SDS) Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

1.1.1 1 year

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

1.1.2 2 years

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

1.1.3 3 years

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

1.2 Measures of mineral metabolism: serum phosphate (mmol/L) Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

1.2.1 6 months

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

1.2.2 1 year

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

1.2.3 2 years

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

1.2.4 3 years

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

1.3 Measures of endocrine function: serum alkaline phosphatase activity (IU/L) Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

1.3.1 6 months

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

1.3.2 1 year

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

1.3.3 2 years

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

1.3.4 3 years

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

1.4 Measures of endocrine function: serum IGF (SDS) Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

1.4.1 6 months

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

1.4.2 1 year

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

1.4.3 2 years

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

1.4.4 3 years

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

1.5 Measures of renal function: TmP/GFR (mmol/L) Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

1.5.1 6 months

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

1.5.2 1 year

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

1.5.3 2 years

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

1.5.4 3 years

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

1.6 BMD: determination of bone age (years) (change from baseline) Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

1.6.1 3 years

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

1.7 Body proportions: sitting height (SDS) Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Subtotals only

1.7.1 1 year

1

15

Mean Difference (IV, Fixed, 95% CI)

‐0.15 [‐1.03, 0.73]

1.7.2 2 years

1

15

Mean Difference (IV, Fixed, 95% CI)

0.40 [‐0.37, 1.17]

1.7.3 3 years

1

15

Mean Difference (IV, Fixed, 95% CI)

0.35 [‐0.14, 0.84]

1.8 Body proportions: arm length (SDS) Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

1.8.1 1 year

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

1.8.2 2 years

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

1.8.3 3 years

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

1.9 Body proportions: leg length (SDS) Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

1.9.1 1 year

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

1.9.2 2 years

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

1.9.3 3 years

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 1. Growth hormone (GH) versus control (no treatment)