Abstract
The most common heritable disorder of renal phosphate wasting, X‐linked hypophosphataemia (XLH), was discovered to be caused by inactivating mutations in the phosphate regulating gene with homology to endopeptidases on the X‐chromosome (PHEX) gene in 1995. Although the exact molecular mechanisms by which PHEX mutations cause disturbed phosphate handling in XLH remain unknown, focus for novel therapies has more recently been based upon the finding that the bone‐produced phosphaturic hormone fibroblast growth factor‐23 is elevated in XLH patient plasma. Previous treatment strategies for XLH were based upon phosphate repletion plus active vitamin D analogues, which are difficult to manage, fail to address the primary pathogenesis of the disease, and can have deleterious side effects. A novel therapy for XLH directly targeting fibroblast growth factor‐23 via a humanized monoclonal antibody (burosumab‐twza/CRYSVITA, henceforth referred to just as burosumab) has emerged as an effective, and recently approved, pharmacological treatment for both children and adults. This review will provide an overview of the clinical manifestations of XLH, the molecular pathophysiology, and summarize its current treatment.
Keywords: burosumab‐twza, fibroblast growth factor 23, PHEX, X‐linked hypophosphataemia
Introduction
X‐linked hypophosphataemia (XLH) is a rare metabolic bone disease with significant clinical consequences in both children and adults, due largely to fibroblast growth factor 23 (FGF23) excess and resulting chronic hypophosphataemia. In the past, treatment has been restricted to administering phosphorus and activated vitamin D, with limitations in effectiveness, and significant risk for important adverse events. Over the past 2 decades research into XLH mechanisms has generated novel potential targets for treatment, including the recent regulatory approvals of burosumab‐twza/CRYSVITA (henceforth referred to just as burosumab) for clinical use in patients with XLH. This paper will review the clinical consequences of XLH, the pathophysiology involved and various approaches to its medical management.
Genetic cause
XLH is a rare bone disease affecting an estimated 1:20–25 000 persons 1, 2 (OMIM no. 307800). Deleterious mutations in the phosphate regulating gene with homologies to endopeptidases on the X chromosome (PHEX) 3 cause this X‐linked dominant condition. The female to male ratio is about 2:1. Sporadic cases frequently occur. Other genes having different inheritance patterns cause phenotypically similar disorders. PHEX mutations cause increased plasma FGF23 concentrations, leading to impaired renal reabsorption of phosphorus, hypophosphataemia and impaired activation of vitamin D 2, 4.
Clinical manifestations
XLH typically presents in early childhood with manifestations of rickets, although family history may lead to testing before visible features appear. Unfortunately, clinical laboratories often fail to report age‐appropriate normal ranges, leading to missed diagnoses, since adult normal phosphorus values are much lower than paediatric (especially infant) values 2. Both children and adults with XLH have low phosphorus concentrations for age, and impaired renal tubular phosphorus reabsorption, typically assessed as a low transport maximum for phosphorus adjusted for glomerular filtration rate (TmP/GFR) 2, 4. Serum alkaline phosphatase activity is typically increased for age in children, although this is more variable in adults 2, 4.
At birth, length is usually normal and legs are not usually bowed 5, 6, 7. Bowing and impaired linear growth become more evident with weight bearing, especially between ages 1 and 2 years 2, 5, 6, 8, 9. Childhood features include bowing of the femora and tibiae leading to genu varus or sometimes valgus appearance, widening of the growth plates at the end of long bones, abnormalities of the skull shape, sometimes bone pain and occasionally delays in gross motor milestones (such as difficulties in walking or running) 2, 9, 10. Radiographic growth plate abnormalities are similar to nutritional rickets, along with diaphyseal bowing, often with medial cortical thickening, and torsion of weight‐bearing long bones. Skeletal severity and the response to medical management are highly variable 2, 6, 10. Patients often require corrective surgical procedures to straighten lower extremities. Short stature and gait abnormalities persist into adulthood 2, 6, 8, 10, 11.
Cranial bones are also affected in XLH, most notably with frontal bossing and dolicocephally 12, 13, 14. Flattening of the cranial base leads to decreased depth of the posterior fossa predisposing to Chiari malformations 13, occurring in up to 44% of XLH patients 12. For some patients, craniosynostosis of the sagittal suture requires craniotomy 14.
XLH predisposes to dental abscesses due to a combination of intrinsic effects of Phex deficiency and associated hypophosphataemia leading to under‐mineralized dentin and cementum 15, 16, 17, 18. Severe dental disease (abscesses and periodontitis) affects 61–78% of patients with XLH 19, 20. Retrospective studies suggest that treating XLH with phosphate salts and active vitamin D decreases the occurrence of dental abscesses and periodontitis 19, 20.
XLH does not negatively influence lifespan, so one would expect at least three quarters of XLH patients to be adults. As such, although commonly thought of as a paediatric bone disease, adults bear a considerable burden of the consequences of this lifelong condition. The most debilitating features of XLH are experienced by adults: bone pain, pseudofractures, enthesopathy and osteoarthritis 11, 21, 22. Osteomalacic bone pain is common 22 and debilitating. Although complete fractures are not more common in XLH, patients with XLH are prone to pseudofractures 11. Pseudofractures occasionally occur in children, but are seen in up to half of adults in cross sectional studies 21, 22, although the lifetime incidence might be higher. Pseudofractures may progress to complete fractures and may require surgical intervention. Skeletal healing after fracture or surgery may be delayed.
Enthesopathy involves the calcification of tendons and ligaments, typically beginning near the bony attachment sites, and progressing to enthesophyte or osteophyte formation. The pathophysiology of enthesopathy in XLH is uncertain, although it may be due to direct effects of FGF23. Enthesopathy occurs in multiple hypophosphataemic mouse models having FGF23 excess 23, 24. FGF receptor 3 and klotho expression in fibrocartilage of the entheses make direct signalling at least theoretically possible 24.
Enthesopathy causes joint stiffness and limitations in range of motion which may be extreme. When involving the anterior and posterior spinal ligaments, patients lose flexion, extension and rotational movement of the neck and spine. Sometimes patients develop spinal nerve or spinal cord compression requiring surgical intervention, although the incidence of this is uncertain. Enthesopathy is more common in men, and with increasing age and BMI 19, affecting between 30 and 100% of adults 10, 11, 19, 21, 22, 24.
Osteoarthritis is probably a consequence of chronic abnormalities in joint alignment and gait causing degenerative changes in joint cartilage and bony articulation surfaces. Osteoarthritis might be exacerbated by enthesopathy. Joint replacements are common surgeries in XLH patients, although the frequency of such procedures is uncertain 25. Up to 82% of adults with XLH complain of joint pain and stiffness, and about 50% report using pain medications weekly to daily 19, 22.
Muscle weakness, a feature of hypophosphataemia, is present in both the Hyp mouse and patients with XLH 22, 26, 27. However, in XLH patients it is difficult to separate the effects on mobility from skeletal deformation, joint abnormalities, and bone pain from those due to muscle weakness.
Overall, the lifetime burden of illness is high in XLH, complicated often by a lack of understanding of the disease and its management by both patients and clinical providers. Not surprisingly, with the growth abnormalities, skeletal deformities, muscle weakness, and bone and joint pain, quality of life impairments are documented in both children and adults with XLH 28, 29, 30.
Molecular pathophysiology
XLH is fully penetrant with highly variable severity. The Hyp Consortium determined that XLH is caused by inactivating mutations in the PHEX gene 3. PHEX encodes a protein that structurally resembles the M13 family of membrane‐bound metalloproteases. Members of this enzyme class include single transmembrane proteins with large extracellular domains such as neutral endopeptidase and endothelin converting enzymes 1 and 2 (ECE‐1 and ECE‐2) 3, 31. This family is known to cleave small peptides, although the endogenous PHEX protein's substrate has yet to be fully confirmed. Close to 300 inactivating PHEX mutations have been described from XLH patients, including variations that lead to missense, nonsense, frame shift, and exon splicing alterations. PHEX shows the highest expression in bone cells including osteoblasts and osteocytes, and odontoblasts in teeth, as well as lower expression in the parathyroid glands, lung, brain and skeletal muscle 31, 32.
Although the PHEX mutations are predicted to cause loss of function, the mechanisms whereby this leads to changes in bone function remain elusive. The Hyp mouse model of XLH, has a 3′ deletion in PHEX, and has been a very useful tool for the in vivo study of the XLH phenotype 31. Similar to XLH patients, this model has approximately a 10‐fold increase in serum FGF23 (although in patients with XLH, serum FGF23 is typically less severely elevated), and manifests the XLH biochemical syndrome including hypophosphataemia with inappropriately normal 1,25‐dihydroxyvitamin D3 [1,25(OH)2D] and normocalcaemia 33. Like patients, the Hyp mice display growth retardation and bone mineralization defects. Interestingly, loss of Phex protein function was associated with a differentiation defect in osteocytes from Hyp mice 34. This mouse line had altered developmental transitions from osteoblasts to osteocytes, with inappropriate expression of cell matrix genes 34. Indeed Hyp bones had over‐expression of type I collagen, as well as altered expression of matrix proteins such as bone sialoprotein 34. Fgf23 mRNA is elevated in Hyp bone 33 and in isolated Hyp osteoblasts/osteocytes 35. These cultures also fail to mineralize fully, consistent with an intrinsic cell defect. However, the cellular pathophysiological mechanisms caused by Phex deficiency that result in altered FGF23 gene expression and sustained FGF23 protein elevation remain elusive.
FGF23 control of phosphate and active vitamin D
The primary kidney transport protein responsible for phosphate reabsorption in the proximal tubule is the type II sodium‐phosphate cotransporter, or NaPi‐IIa. Transgenic FGF23 mice had dramatic decreases in protein expression of NaPi‐IIa and a related transporter, NaPi‐IIc 36. The Hyp mouse has a 50% reduction in proximal tubule NaPi‐IIa expression 37. In normal individuals, phosphate depletion is a strong stimulus for increasing serum 1,25(OH)2D 38. However, in XLH patients, hypophosphataemia is accompanied by low or inappropriately normal 1,25(OH)2D concentrations, due to effects of FGF23. In mice implanted with cells expressing FGF23, or injected with FGF23 itself, mRNA and protein expression levels of the activating enzyme 25‐hydroxyvitamin D3 1‐α‐hydroxylase (Cyp27b1) was decreased, while the catabolic 1,25‐(OH)2D 24‐hydroxylase (Cyp24a1) is increased with high FGF23 39, 40, 41. Thus, the effects of FGF23 on the renal vitamin D metabolic enzymes is responsible for the reductions in serum 1,25(OH)2D concentrations observed in XLH patients.
Patients with autosomal dominant hypophosphataemic rickets (ADHR) also have elevated FGF23 but some patients display waxing and waning of the disease symptoms 42. Studying a large ADHR kindred, we documented that the ADHR disease state and circulating levels of intact FGF23 correlated strongly with iron deficiency and were reciprocal to serum iron concentrations 43, which was borne out in an ADHR knock in animal model to be due to increased bone Fgf23 mRNA production during experimental iron depletion 44, 45. However, intact FGF23 levels in XLH patients were unrelated to serum iron concentration 46. A clinical trial is underway to place ADHR patients on low‐dose oral iron repletion (ClinicalTrials.gov NCT02233322), thus this regimen may be the optimal treatment for ADHR, but not XLH (see below).
Medical management
Phosphate salts and active vitamin D
For about 4 decades, children and adults with XLH have been medically managed using pharmacological doses of phosphate salts and active vitamin D, attempting to counter the effects of FGF23 excess. Doses reported vary widely in the literature 2. A US‐based group of experts recommended target doses of calcitriol 20–30 ng kg–1 daily and phosphate 20–40 mg kg–1 daily, each in divided doses 2. European experts recommended 1–3 μg day–1 of alfacalcidiol or 0.5–1.5 μg day–1 of calcitriol, and 40–60 mg kg–1 daily of phosphate 10. However, no study has systematically evaluated the optimal target dose ranges. Clinicians start at lower doses and titrate to target doses primarily based on gastrointestinal tolerance, with frequent laboratory monitoring to avoid hypercalciuria, hypercalcaemia, hyperphosphataemia or impairments in renal function. This strategy does not effectively normalize the serum phosphorus. In fact, attempts to target normal phosphorus concentrations might be a contributing reason for the high risk of hyperparathyroidism and nephrocalcinosis with these agents 2. Monitoring patients on treatment is complicated by many factors, and most laboratory monitoring addresses safety (serum calcium, phosphorus, creatinine, PTH, urine calcium, urine creatinine).
Treatment with active vitamin D and phosphate decreases alkaline phosphatase and, in bone biopsy studies, improves the osteomalacia of XLH 47, 48. However, the skeletal response varies widely. Some children straighten leg deformities and improve their growth, while others persist in skeletal deformities and short stature. The reason for good responders vs. poor responders is not solely an issue of compliance 49. Treating XLH with vitamin D analogues and phosphate also increases plasma FGF23 concentrations 50, 51, 52, which theoretically could blunt therapeutic effectiveness, although the true consequences remain unknown.
Many children with XLH have a variable but progressive decline in height Z‐scores, often worsening during puberty despite therapy with vitamin D analogues and phosphate 8. Studies suggest that beginning treatment prior to age 1 year improves height outcomes 6.
When leg deformities persist, the optimal timing of corrective surgery remains uncertain. In a retrospective study, 29% of XLH patients had recurrence of deformity after their first corrective surgery 53. Although the differences were not significant, those having earlier corrective procedures were more likely to require additional procedures. This finding may be confounded by more severely affected patients having earlier surgeries.
Unlike nutritional rickets, which is truly cured by adequate vitamin D repletion, the homeostatic defect in phosphorus handling is lifelong in XLH. However, because of the known risks of XLH therapy with active vitamin D and phosphate, general practice has been to stop treatment at the end of growth, and restart if developing clinical issues associated with active osteomalacia 2. This has led to some confusion over whether or not adults benefit from treatment, and misinterpretation of XLH as a childhood disease. Although some adults tolerate stopping treatment with few symptoms, at least for a time, many develop active osteomalacic symptoms, bone pain, muscle weakness and pseudofractures, which demonstrate varying degrees of improvement during treatment with active vitamin D and phosphate 22.
Important side effects limit this therapy. Doses of phosphate may be limited by gastrointestinal symptoms, nephrocalcinosis, ectopic calcification and hyperparathyroidism, while active vitamin D may also contribute to nephrocalcinosis or other ectopic calcification. Nephrocalcinosis is reported in 50–80% of XLH patients receiving active vitamin D and phosphate, and appears to relate to episodes of hypercalciuria 54, 55, 56, 57. Most nephrocalcinosis is mild, but occasionally patients develop chronic kidney disease (CKD), the prevalence of which is uncertain, but progression to end‐stage renal disease is rare. Hypertension has been reported in 27% of XLH patients and may relate in part to nephrocalcinosis and CKD 58.
Hyperparathyroidism in XLH is complex. Nearly half of treatment naïve children with XLH have elevated serum PTH 59, which often decreases after starting active vitamin D. Serum PTH correlates with FGF23 concentrations in untreated patients 52. However, phosphate doses also lead to secondary hyperparathyroidism, probably through transient decreases in serum calcium following every dose. Some patients with secondary hyperparathyroidism progress to tertiary hyperparathyroidism with multigland hyperplasia and require surgical intervention to control hypercalcaemia 60, 61. Prior to the development of parathyroid autonomy, adequate doses of active vitamin D and lowering doses of phosphate may combat the rise of PTH.
Novel targets
Given the limitations of effect and the significant risks of treating XLH with active vitamin D and phosphate salts, the need for improved treatment options has led to development of other strategies. We will focus on those targets that have some clinical/translational data in humans.
Calcimimetics
PTH stimulates FGF23 expression in osteocytes 62, which could exacerbate hypophosphataemia in XLH. Since hyperparathyroidism also complicates XLH treatment, cinacalcet, a calcimimetic, has been used to manage hyperparathyroidism in cases of XLH 63, 64. Short term studies of cinacalcet in children with XLH demonstrated suppression of the PTH surge that follows doses of phosphate 65, and improved TmP/GFR and serum phosphorus. In a case report, one child developed a severe aversion to, and refusal of, his oral phosphate doses. Although he did not have hyperparathyroidism, treatment with cinacalcet, calcitriol and calcium led to improvements in serum phosphorus and rickets, although serum FGF23 still increased 66. Further studies are needed to determine what role cinacalcet should play, although it may be useful in tertiary hyperparathyroidism.
Calcitonin
Calcitonin stimulates 1α‐hydroxylase in the proximal renal tubule in Hyp mice 67, and transiently increases 1,25(OH)2D in subjects with XLH after a single injection 68. Osteocytes also express the calcitonin receptor 69, and as such, calcitonin could influence FGF23 secretion. Calcitonin decreased serum FGF23 in a patient with tumour‐induced osteomalacia (TIO) 70. A single calcitonin injection decreased serum FGF23 and increased serum phosphorus transiently in patients with XLH, although not in healthy controls 71. To make the calcitonin story more confusing, case reports indicate calcitonin increases urinary phosphorus excretion in hyperphosphatemic tumoural calcinosis (a condition of FGF23 deficiency rather than excess) 72, which would be opposite of the goal of XLH management. A 3‐month‐long blinded randomized controlled clinical trial of monotherapy with nasal calcitonin 400 units daily for XLH failed to demonstrate improvements in serum phosphorus, TmP/GFR or FGF23 73. Given the current lack of data indicating benefit, calcitonin should not be used for XLH.
Fibroblast growth factor receptor antagonism in TIO
TIO (OMIM 605380) is another syndrome of excess fibroblast growth factor receptor (FGF23) 39, 74. These tumours are classified under the collective term of phosphaturic mesenchymal tumour, mixed connective tissue variant (PMTMCT) or phosphaturic mesenchymal tumour 75. Complete tumour resection is the most straight forward cure for TIO. For tumour localization, imaging techniques using radiolabelled octreotide 76 as well as magnetic resonance imaging 77, computed tomography 78, whole body sestamibi scanning 79, and Ga68‐DOTA‐octreotide positron emission tomography/computed tomography imaging 80 may be useful. Selective venous sampling for FGF23 levels has been attempted to locate tumours as well 77, 81. Unfortunately, many PMTMCTs are small in size and remain difficult to localize, necessitating medical management.
In a group of 15 TIO tumours analysed through next‐generation sequencing, a fibronectin (FN1)‐FGFR1 fusion gene was detected in 60% of the tested samples 82. The fusion protein is predicted to express portions of the three extracellular FGF‐binding (Ig‐like) domains 82, therefore ligand‐activated receptor signalling could perhaps occur. It is not established however, whether the fusion gene is causative of the TIO tumour or is a consequence of the tumourigenesis. Activating FGFR1 mutations are associated with osteoglophonic dysplasia, a disease of dwarfism as well as craniosynostosis 83. Some osteoglophonic dysplasia patients also have significantly elevated FGF23 and hypophosphataemia. Thus, inhibitors of FGF binding or FGFR activity could be useful pharmacological treatments for TIO. Such inhibitors may block FGFR auto‐ and ligand‐ and Klotho‐dependent FGFR dimerization, and may reduce signalling through FGFRs along with directly inhibiting FGF23 bioactivity. However, the ubiquitous nature of FGFR1 could lead to off‐target effects. Considering the difficulty with targeting FGFR inhibitors to a specific tissue, another plausible approach for treating TIO patients until the tumour is found may be monotherapy with anti‐FGF23 antibody (see below) as is currently being tested in TIO and XLH (ClinicalTrials.gov NCT02304367).
FGF23‐blocking antibodies
Investigators at Kirin Pharma developed murine anti‐FGF23 monoclonal antibodies that bind separate isotopes on either the C‐terminal or N‐terminal portions of FGF23 84, providing information on the structural interactions of FGF23 with its receptor/co‐receptor. Both antibodies interfered with FGFR signalling as indicated by declines in Egr‐1 reporter activity (a molecular marker for MAPK activation). The N‐terminal antibody interfered with binding of FGF23 to the FGFR, while the C‐terminal antibody interfered with binding to the coreceptor klotho. Injecting either antibody into mice produced generally similar effects: transient increases in 1,25(OH)2D beginning within 3–5 hours, and in TmP/GFR and serum phosphorus after 16 h 84. Combining the antibodies was synergistic.
Hyp mice receiving subcutaneous injection of these antibodies in combination demonstrated a similar biochemical response and time course after a single dose 85. When juvenile, 4‐week‐old Hyp mice were treated with weekly injections for 5 weeks, renal sodium phosphate cotransporter expression increased on immunohistochemistry, and serum phosphorus improved in a dose‐dependent manner along with increases in Cyp27b1 mRNA expression and serum 1,25(OH)2D. As a result, mice had bone histomorphometric improvements, including decrease in osteoid thickness and volume toward normal, as well as increased mineral apposition rate, bone formation rate, and bone volume/total volume (BV/TV). This translated into improvements in the rachitic growth plate morphology, the growth and shape of their tails, femora and tibiae, and in overall size of the mice 26, 85. Treating adult Hyp mice with anti‐FGF23 antibodies also improved osteomalacia on histology, and increased muscle grip strength toward normal without changing muscle weight 26.
Based on successes in the Hyp mouse, a fully human IgG1 monoclonal antibody to bind FGF23 was developed for human trials (KRN23, now called burosumab or CRYSVITA). Table 1 summarizes key findings from the published clinical trials. In the initial dose finding study, single doses of intravenous or subcutaneous burosumab (or placebo) were administered to adults with XLH after stopping phosphate or active vitamin D 86. Burosumab increased TmP/GFR, serum phosphorus and 1,25(OH)2D in a dose dependent manner. The half‐life was longer for subcutaneous dosing (13–19 days) than with intravenous dosing (8–12 days). Baseline FGF23 concentration did not predict AUC changes for any pharmacodynamic parameter.
Table 1.
Key findings from the primary burosumab clinical trial publications in X‐linked hypophosphataemia (XLH) to datea
| Study [ref] /ClinicalTials.gov number | Carpenter et al. JCI 2011 86 NCT00830674 |
Imel et al. JCEM 2015 87, Ruppe et al. Bone Reports 2016 28
NCT01340482, NCT01571596 |
Carpenter et al. NEJM 2018 29
NCT02163577 |
Insogna et al. JBMR 2018 21
NCT02526160 |
|---|---|---|---|---|
| Population | Adults with XLH | Adults with XLH |
Children with XLH, ages 5–12 years at enrollment |
Adults with XLH and Brief Pain Inventory worst pain score ≥ 4 |
| Treatment arms |
Single dose
Intravenous burosumab 0.003–0.3 mg kg–1 (n = 17) vs. placebo (n = 5) or Subcutaneous burosumab 0.1–1 mg kg–1 (n = 12) vs. placebo (n = 4) |
Multidose for 16 months: 4‐month dose escalation phase Every 4 weeks burosumab 0.05–0.6 mg kg–1 (n = 28), followed by 12‐month extension phase Every 4 weeks burosumab 0.1–1 mg kg–1 (n = 22) b |
Multidose for 64 weeks, with dose titration period c Every 2 weeks burosumab (mean dose at week 40 was 0.98 mg kg–1) (n = 26) or Every 4 weeks burosumab (mean dose at week 40 was 1.5 mg kg–1; n = 26) |
Multidose for 24 weeks:
Every 4 weesk burosumab 1 mg kg–1 (n = 68) or Every 4 weeks placebo (n = 66) |
| Primary outcome | Safety and tolerability | Proportion of subjects achieving maximum fasting serum Pi in normal range | Change in RSS from baseline to week 40 and 64 | Percent of subjects achieving mean serum Pi in the normal range across the midpoint of dosing intervals |
| Biochemical findings | Increased serum Pi, serum 1,25(OH)2D3, and TmP/GFR | Increased serum Pi, serum 1,25(OH)2D3, and TmP/GFR |
Increased serum Pi, serum 1,25(OH)2D3, and TmP/GFR; decreased serum alkaline phosphatase. |
Increased serum Pi, serum 1,25(OH)2D3, and TmP/GFR |
| Radiographic outcomes |
Improved rickets by RSS and RGI‐C. Greater changes seen with every 2‐week dosing, and in those with higher baseline RSS |
Improved healing of active fractures/pseudofractures from baseline: burosumab (43.1% of fractures healed) compared to placebo (7.7%) |
||
| Other outcomes |
At baseline: Impairment of physical function Over 4 months of treatment: Improved role limitations due to physical health (SF‐36v2), physical functioning and stiffness (WOMAC) |
Increased standing height z‐score; Among those with baseline impairments in 6MWT, the distance walked improved On PODCI, among those with impairments, burosumab improved: Sports and physical functioning domain, Pain and comfort domain, Global functioning |
WOMAC: Decreased stiffness scores |
In each of these trials, subjects had a washout period from phosphate salts and active vitamin D analogues, and burosumab (or placebo) was given as monotherapy. Dose titration was based on serum phosphorus concentration, targeting normal serum phosphorus ranges
During the extension phase over 80% of subjects received doses of 0.6–1 mg kg–1 87
Children in this trial started initially at very low doses, which were titrated based on serum phosphorus concentration, and sequential entry cohorts began at higher doses 29
Pi, phosphorus; PODCI, Pediatric Outcomes Data Collection Instrument; RGI‐C, Radiographic Global Impression of Change scale for rachitic features (positive scores indicate improvement); RSS, Thacher Rickets Severity Score; Sf‐36v2, Medical Outcomes Study Short Form Health Survey version 2; TmP/GFR, transport maximum phosphorus adjusted for glomerular filtration rate; WOMAC, Western Ontario and McMaster Osteoarthritis Index; 1,25(OH)2D3 1,25‐dihydroxyvitamin D3; 6MWT, 6‐min walking test
Twenty‐eight adults with XLH stopped all XLH medications and enrolled in a Phase 2 dose‐escalating multidose trial of burosumab administered subcutaneously every 4 weeks, with doses ranging from 0.05 to 0.6 mg kg–1 and 22 subjects continued in a 12‐month extension with doses ranging from 0.1–1 mg kg–1 87. During the multidose trial, burosumab exhibited first‐order absorption and elimination kinetics, similar to other monoclonal antibody therapies, with an elimination half‐life of 17.8 days 88.
The peak value of 1,25(OH)2D and serum phosphorus occurred about 3–7 days after injection, while peak TmP/GFR occurred about 7 days after injection 87. After each peak, the TmP/GFR, serum phosphorus and 1,25(OH)2D decreased toward a trough level 4 weeks after injection that remained generally above the baseline values. Peak and trough values increased during the dose escalation phase, then remained generally stable through the 12 month extension, except for the peak of 1,25(OH)2D, which decreased in magnitude over later doses 87. Most subjects at least transiently achieved normal serum phosphorus concentrations.
Despite the transient increase in 1,25(OH)2D during each cycle, there was no systematic change in serum or urine calcium or in PTH. Kidney function also remained stable. Ten subjects had baseline nephrocalcinosis or nephrolithiasis, and no subject had worsening nephrocalcinosis 87.
Although this first multidose trial did not assess clinical bone outcomes, the bone biomarkers P1NP, osteocalcin and bone alkaline phosphatase increased during the study period. Quality of life instruments (SF‐36v2 and WOMAC) indicated baseline impairments in bodily pain, physical function and role limitations, along with stiffness. After 4 months there were improvements in the patients' perception of their physical functioning and stiffness 28.
In a recently published, open‐label, phase 2 trial, 52 children aged 5–12 years with XLH in Europe and the USA were randomized to receive burosumab subcutaneously every 2 weeks or every 4 weeks for 64 weeks 29. Patients were mostly prepubertal, although Tanner stage 2 subjects were allowed. All patients stopped phosphate and vitamin D analogues 2 weeks prior to screening. Since this was the first trial conducted in children, the initial cohort started at very low burosumab doses and titrated upwards targeting serum phosphorus levels between 3.2 and 6.1 mg dl–1 (1.0–2.0 mmol l–1). Rickets was assessed using a rickets severity score (RSS) that rated radiographic appearance at the wrist and knee on a scale from 0–10 (10 being the worst). Differences between baseline and follow‐up radiographs were also assessed using a seven‐point ordinal Radiographic Global Impression of Change (RGI‐C) scale (ranging from –3 severe worsening, to 0 no change, to +3 complete healing) 89.
One limitation of this paediatric study was its lack of a control group. However, all but one subject had been receiving standard treatment with active vitamin D analogues and phosphate salts up to the time of enrolment (for a mean of 6.9 years prior treatment). Thus, the baseline rickets severity at enrolment (ranging from RSS 0–4.5, mean 1.8 ± 1.1) represents residual rickets despite prior treatment. Any improvements seen in trial occurred in the setting of switching from prior standard therapy to burosumab.
At week 40 the mean burosumab dose had been titrated to 0.98 mg kg–1 every 2 weeks or 1.5 mg kg–1 every 4 weeks 29. Children receiving every 4 weeks dosing had pharmacodynamic profiles of serum phosphorus, 1,25(OH)2D and TmP/GFR similar to that seen in adults 29. However, the every 2 weeks dosing group had more sustained improvements in these parameters, with less pronounced biochemical trough effects, and serum phosphorus values more consistently in the normal range. Alkaline phosphatase is commonly elevated in XLH patients (mean 459 ± 105 units l–1 at baseline of this trial), which decreased by 20% at week 64. Both groups demonstrated improvements in rickets by week 40 that were sustained at week 64. Rickets improvements were numerically greater in those receiving every 2 weeks injections and also in those having worse baseline rickets severity. Total RSS score decreased from mean 1.9 to 0.8 after every 2 weeks dosing, and from 1.7 to 1.1 after every 4 weeks dosing. The mean total RGI‐C was +1.57 at the end of study, indicating healing of rickets. Substantial healing of rickets (RGI‐C ≥ +2) occurred in approximately half the patients in each group, but in 94% of those with baseline RSS ≥1.5 that received burosumab every 2 weeks. In addition, small improvements in standing height Z‐score were observed (+0.15 ± 0.04). Additional functional improvements were noted. Impairments in 6‐min walking distance were present at baseline in 46% of patients, and among these the 6‐min walking distance improved by 10% of the predicted normal range for age. Similarly, baseline scores indicated impairments for sports and physical functioning, and in pain and comfort domains in 54% of the children using the paediatric outcomes data collection instrument, which were seen to improve during treatment.
In a Phase 2 trial, 13 children with XLH, age 1–4 years, were enrolled to receive open‐label burosumab 0.8 mg kg–1 every 2 weeks. This trial indicated similar improvements in serum phosphorus, alkaline phosphatase and rickets severity to that seen in the 5–12‐year‐old children 90, 91. Some details of this trial have been presented at scientific meetings and are indicated in the Food and Drug Administration (FDA) drug label 90, 91, but have not yet been fully published.
The pivotal Phase 3 adult trial randomized 134 adults with XLH to receive burosumab at doses of 1 mg kg–1 (maximum 90 mg) every 4 weeks vs. a placebo for 24 weeks (without an active comparator treatment), after which all subjects received burosumab for the subsequent 24 weeks 21. Subjects were required to have a brief pain inventory worst pain ≥4 to enrol. Nearly all subjects (99%) had radiographic evidence of enthesopathy, half had nephrocalcinosis, while 47% in the burosumab group and 57% in the placebo group had pseudofractures/fractures with several subjects having multiple pseudofractures. The primary endpoint (the percent of patients achieving mean serum phosphorus concentrations in the normal range across the midpoint of dosing intervals) was achieved in 94% of subjects after burosumab vs. 7.6% after placebo (Figure 1A). Trough serum phosphorus remained in the normal range in 67.6% of burosumab treated subjects.
Figure 1.

Data from a 24‐week randomized, placebo‐controlled trial of burosumab 1 mg kg–1 vs. placebo in adults with X‐linked hypophosphataemia are shown. (A) Mean serum phosphorus is within the normal range at the midpoint of dosing interval during treatment with burosumab, compared with placebo treated subjects, whose phosphorus remained low. (B) Active fractures or pseudofractures were more likely to heal during 24 weeks of burosumab compared with placebo. Adapted and reproduced from Figures 1A and 3A of Insogna et al. J Bone Miner Res 2018;33:1383–1393; (Reference [21]); Wiley Publishers, https://doi.org/10.1002/jbmr.3475. Creative Commons Attribution License (CC BY 4.0, https://creativecommons.org/licenses/by/4.0)
The burosumab group had improvements at week 24 in the brief pain inventory worst pain score, and WOMAC physical function and stiffness subscales compared to placebo, but only stiffness remained significant after adjustment for multiple comparisons. Most importantly 43.1% of baseline fractures/pseudofractures were healed after 24 weeks in the burosumab group vs. 7.7% in the placebo group (Figure 1B).
Regarding safety, the most common drug‐related adverse events have been local injection site reactions that were often characterized as welts, and usually lasted a few to several hours, resolving without treatment. Injection site reactions occurred in 57.7% of children 29 and 11.8% of adults 21, although, interestingly, injection site reactions also occurred in 12.1% of placebo treated adults. Additional adverse events occurring in ≥25% of children included headache, vomiting, pyrexia, extremity pain and decrease in vitamin D. Additional adverse events occurring in ≥10% of adults included back pain, tooth abscess, nasopharyngitis, headaches, nausea and dizziness. However, most listed adverse events were not considered drug related, while some are consistent with symptoms common to XLH patients. Restless legs syndrome was noted in the initial multidose trial (5/28 or 17.9%) 87. Restless legs syndrome occurred more often in burosumab‐treated subjects in the placebo controlled adult study (8/68 or 11.8%) but was also present to some degree in the placebo arm (5/66 or 7.6%) 21. No serious adverse events were attributed to burosumab itself.
There has been no systematic evidence of change in PTH, serum calcium or urine calcium excretion in any trials. Nephrocalcinosis was present at baseline in 35% of children in the Phase 2 trial 29 and 54% of adults in the Phase 3 trial 21. So far, there has been no clear indication of either worsening or improvement of nephrocalcinosis overall with burosumab treatment. In fact, similar numbers of patients had increases in nephrocalcinosis score in the burosumab and placebo groups, while similar numbers also had decreases in nephrocalcinosis scores in each group 21. No significant changes in echocardiograms were reported.
An advantage of burosumab is that it achieves normalization of serum phosphorus through improvements in TmP/GFR, in contrast to standard therapy with active vitamin D and phosphate salts. One hopes that this will translate into lower long‐term risk of nephrocalcinosis and hyperparathyroidism, provided that episodes of hyperphosphataemia are avoided. In the phase 3 placebo controlled adult trial, five patients (7.4%) had dose reductions according to protocol, due to serum phosphorus above the target range, four of which were classified as hyperphosphataemic 21. These subjects maintained in the trial on smaller doses. Thus, some patients may respond well to smaller doses to manage XLH.
Conceptually, anti‐drug antibodies could bind and interfere with the action of burosumab. The initial multidose trial in adults, and the Phase 2 trial in children did not find anti‐drug antibody development 29, 87. However, one published trial (and the drug labels) notes that anti‐drug antibodies have been detected in small numbers of subjects at baseline before burosumab administration 21, 91. Anti‐drug antibodies persisted in some subjects, but further development of antibodies during trial participation, was not seen. To date, there is no evidence for neutralizing anti‐drug antibodies attenuating effect of burosumab in human subjects.
Patients with moderate to severe CKD were excluded from the burosumab trials. In a rat model of severe CKD, treatment with a different FGF23 antibody hastened death, probably due to the frank hyperphosphataemia that developed 92. Thus, the use of burosumab in severe kidney disease is not recommended, and hyperphosphataemia should be avoided. The safety of burosumab (or for that matter of phosphate and active vitamin D) in pregnancy has not been established.
Burosumab was approved as monotherapy in 2017 by the European Medicines Agency (EMA; conditionally) and in 2018 by the FDA (fully). The EMA conditionally approved use of burosumab in children aged 1 year and older with radiographic evidence of bone disease, through adolescence during skeletal growth. The EMA approved starting dose is 0.4 mg kg–1 subcutaneously every 2 weeks in children with 0.8 mg kg–1 as typical maintenance dosing, and a maximum dose of 90 mg 93. The FDA approved starting dose is 0.8 mg kg–1 every 2 weeks in children aged 1 year and older, and 1 mg kg–1 every 4 weeks in adults with a maximum dose of 90 mg 91. Doses are titrated on the basis of peak and trough serum phosphorus concentrations.
Several questions remain. A true active comparator study in growing children aged 1–12 years is underway (ClinicalTrials.gov NCT02915705). This trial will identify the effect of 64 weeks of burosumab vs. phosphate and active vitamin D on multiple clinical parameters with a primary outcome of differences in healing of radiographic rickets. The effect on adult height in growing children is unknown. However, this will take years to evaluate, given the mean ages of children enrolled in the clinical trials. Since enthesopathy and other joint related complications develop and progress slowly, many years of treatment with burosumab will be necessary to interpret whether or not these outcomes are altered. Similarly, years of treatment are likely to be necessary to determine whether the chronic risk of nephrocalcinosis changes.
Conclusion
XLH is a complex musculoskeletal condition, requiring careful medical and surgical management. Recent approval of burosumab represents a substantial advancement in management of XLH, directed at its pathophysiology of FGF23 excess. Further studies are needed to fully characterize long‐term risks and benefits of this drug, especially regarding disease features that take years to develop.
Nomenclature of targets and ligands
Key protein targets and ligands in this article are hyperlinked to corresponding entries in http://www.guidetopharmacology.org, the common portal for data from the IUPHAR/BPS Guide to PHARMACOLOGY 94, and are permanently archived in the Concise Guide to PHARMACOLOGY 2017/18 95, 96.
Competing Interests
E.A.I. receives research funding that contributes to this work from the NIH/NIAMS 1P30AR072581. K.E.W. would like to acknowledge support by NIH grants DK063934, DK095784, and AR059278. E.A.I. has received research funding and fees for consulting with Kyowa Hakko Kirin and Ultragenyx, Pharmaceuticals. K.E.W. receives royalties from Kyowa Hakko Kirin Co. Ltd for licensing FGF23.
Imel E. A., and White K. E. (2019) Pharmacological management of X‐linked hypophosphataemia, Br J Clin Pharmacol, 85, 1188–1198. doi: 10.1111/bcp.13763.
References
- 1. Beck‐Nielsen SS, Brock‐Jacobsen B, Gram J, Brixen K, Jensen TK. Incidence and prevalence of nutritional and hereditary rickets in southern Denmark. Eur J Endocrinol 2009; 160: 491–497. [DOI] [PubMed] [Google Scholar]
- 2. Carpenter TO, Imel EA, Holm IA, Jan de Beur SM, Insogna KL. A clinician's guide to X‐linked hypophosphatemia. J Bone Miner Res 2011; 26: 1381–1388. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Hyp Consortium . A gene (PEX) with homologies to endopeptidases is mutated in patients with X‐linked hypophosphatemic rickets. The HYP consortium. Nat Genet 1995; 11: 130–136. [DOI] [PubMed] [Google Scholar]
- 4. Imel EA, Econs MJ. Approach to the hypophosphatemic patient. J Clin Endocrinol Metab 2012; 97: 696–706. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. McNair SL, Stickler GB. Growth in familial hypophosphatemic vitamin‐D‐resistant rickets. N Engl J Med 1969; 281: 512–516. [DOI] [PubMed] [Google Scholar]
- 6. Makitie O, Doria A, Kooh SW, Cole WG, Daneman A, Sochett E. Early treatment improves growth and biochemical and radiographic outcome in X‐linked hypophosphatemic rickets. J Clin Endocrinol Metab 2003; 88: 3591–3597. [DOI] [PubMed] [Google Scholar]
- 7. Harrison HE, Harrison HC, Lifshitz F, Johnson AD. Growth disturbance in hereditary hypophosphatemia. Am J Dis Child 1966; 112: 290–297. [DOI] [PubMed] [Google Scholar]
- 8. Zivicnjak M, Schnabel D, Billing H, Staude H, Filler G, Querfeld U, et al Age‐related stature and linear body segments in children with X‐linked hypophosphatemic rickets. Pediatr Nephrol 2011; 26: 223–231. [DOI] [PubMed] [Google Scholar]
- 9. Kruse K, Hinkel GK, Griefahn B. Calcium metabolism and growth during early treatment of children with X‐linked hypophosphataemic rickets. Eur J Pediatr 1998; 157: 894–900. [DOI] [PubMed] [Google Scholar]
- 10. Linglart A, Biosse‐Duplan M, Briot K, Chaussain C, Esterle L, Guillaume‐Czitrom S, et al Therapeutic management of hypophosphatemic rickets from infancy to adulthood. Endocr Connect 2014; 3: R13–R30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Beck‐Nielsen SS, Brusgaard K, Rasmussen LM, Brixen K, Brock‐Jacobsen B, Poulsen MR, et al Phenotype presentation of hypophosphatemic rickets in adults. Calcif Tissue Int 2010; 87: 108–119. [DOI] [PubMed] [Google Scholar]
- 12. Caldemeyer KS, Boaz JC, Wappner RS, Moran CC, Smith RR, Quets JP. Chiari I malformation: association with hypophosphatemic rickets and MR imaging appearance. Radiology 1995; 195: 733–738. [DOI] [PubMed] [Google Scholar]
- 13. Gjorup H, Kjaer I, Sonnesen L, Haubek D, Beck‐Nielsen SS, Hintze H, et al Craniofacial morphology in patients with hypophosphatemic rickets: a cephalometric study focusing on differences between bone of cartilaginous and intramembranous origin. Am J Med Genet A 2011; 155A: 2654–2660. [DOI] [PubMed] [Google Scholar]
- 14. Vega RA, Opalak C, Harshbarger RJ, Fearon JA, Ritter AM, Collins JJ, et al Hypophosphatemic rickets and craniosynostosis: a multicenter case series. J Neurosurg Pediatr 2016; 17: 694–700. [DOI] [PubMed] [Google Scholar]
- 15. Onishi T, Umemura S, Shintani S, Ooshima T. Phex mutation causes overexpression of FGF23 in teeth. Arch Oral Biol 2008; 53: 99–104. [DOI] [PubMed] [Google Scholar]
- 16. Boukpessi T, Hoac B, Coyac BR, Leger T, Garcia C, Wicart P, et al Osteopontin and the dento‐osseous pathobiology of X‐linked hypophosphatemia. Bone 2017; 95: 151–161. [DOI] [PubMed] [Google Scholar]
- 17. Chaussain‐Miller C, Sinding C, Wolikow M, Lasfargues JJ, Godeau G, Garabedian M. Dental abnormalities in patients with familial hypophosphatemic vitamin D‐resistant rickets: prevention by early treatment with 1‐hydroxyvitamin D. J Pediatr 2003; 142: 324–331. [DOI] [PubMed] [Google Scholar]
- 18. Fong H, Chu EY, Tompkins KA, Foster BL, Sitara D, Lanske B, et al Aberrant cementum phenotype associated with the hypophosphatemic hyp mouse. J Periodontol 2009; 80: 1348–1354. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Connor J, Olear EA, Insogna KL, Katz L, Baker S, Kaur R, et al Conventional therapy in adults with X‐linked hypophosphatemia: effects on enthesopathy and dental disease. J Clin Endocrinol Metab 2015; 100: 3625–3632. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Biosse Duplan M, Coyac BR, Bardet C, Zadikian C, Rothenbuhler A, Kamenicky P, et al Phosphate and vitamin D prevent periodontitis in X‐linked hypophosphatemia. J Dent Res 2017; 96: 388–395. [DOI] [PubMed] [Google Scholar]
- 21. Insogna KL, Briot K, Imel EA, Kamenicky P, Ruppe MD, Portale AA, et al A randomized, double‐blind, placebo‐controlled, phase 3 trial evaluating the efficacy of burosumab, an anti‐FGF23 antibody, in adults with X‐linked hypophosphatemia: week 24 primary analysis. J Bone Miner Res 2018; 33: 1383–1393. [DOI] [PubMed] [Google Scholar]
- 22. Reid IR, Hardy DC, Murphy WA, Teitelbaum SL, Bergfeld MA, Whyte MP. X‐linked hypophosphatemia: a clinical, biochemical, and histopathologic assessment of morbidity in adults. Medicine (Baltimore) 1989; 68: 336–352. [PubMed] [Google Scholar]
- 23. Karaplis AC, Bai X, Falet JP, Macica CM. Mineralizing enthesopathy is a common feature of renal phosphate‐wasting disorders attributed to FGF23 and is exacerbated by standard therapy in hyp mice. Endocrinology 2012; 153: 5906–5917. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Liang G, Katz LD, Insogna KL, Carpenter TO, Macica CM. Survey of the enthesopathy of X‐linked hypophosphatemia and its characterization in Hyp mice. Calcif Tissue Int 2009; 85: 235–246. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Mills E, Iorio L, Macica CM. Assessing outcomes of joint replacement in patients with X‐linked hypophosphatemia. In: ASBMR 2017 Annual Meeting, Denver, Colorado: ASBMR, 2017: SA0339.
- 26. Aono Y, Hasegawa H, Yamazaki Y, Shimada T, Fujita T, Yamashita T, et al Anti‐FGF‐23 neutralizing antibodies ameliorate muscle weakness and decreased spontaneous movement of Hyp mice. J Bone Miner Res 2011; 26: 803–810. [DOI] [PubMed] [Google Scholar]
- 27. Veilleux LN, Cheung M, Ben Amor M, Rauch F. Abnormalities in muscle density and muscle function in hypophosphatemic rickets. J Clin Endocrinol Metab 2012; 97: E1492–E1498. [DOI] [PubMed] [Google Scholar]
- 28. Ruppe MD, Zhang X, Imel EA, Weber TJ, Klausner MA, Ito T, et al Effect of four monthly doses of a human monoclonal anti‐FGF23 antibody (KRN23) on quality of life in X‐linked hypophosphatemia. Bone Rep 2016; 5: 158–162. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Carpenter TO, Whyte MP, Imel EA, Boot AM, Hogler W, Linglart A, et al Burosumab therapy in children with X‐linked hypophosphatemia. N Engl J Med 2018; 378: 1987–1998. [DOI] [PubMed] [Google Scholar]
- 30. Che H, Roux C, Etcheto A, Rothenbuhler A, Kamenicky P, Linglart A, et al Impaired quality of life in adults with X‐linked hypophosphatemia and skeletal symptoms. Eur J Endocrinol 2016; 174: 325–333. [DOI] [PubMed] [Google Scholar]
- 31. Beck L, Soumounou Y, Martel J, Krishnamurthy G, Gauthier C, Goodyer CG, et al PEX/PEX tissue distribution and evidence for a deletion in the 3′ region of the Pex gene in X‐linked hypophosphatemic mice. J Clin Invest 1997; 99: 1200–1209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Meyer MH, Meyer RA Jr. MRNA expression of Phex in mice and rats: the effect of low phosphate diet. Endocrine 2000; 13: 81–87. [DOI] [PubMed] [Google Scholar]
- 33. Liu S, Zhou J, Tang W, Jiang X, Rowe DW, Quarles LD. Pathogenic role of Fgf23 in Hyp mice. Am J Physiol Endocrinol Metab 2006; 291: E38–E49. [DOI] [PubMed] [Google Scholar]
- 34. Miao D, Bai X, Panda D, McKee M, Karaplis A, Goltzman D. Osteomalacia in hyp mice is associated with abnormal PHEX expression and with altered bone matrix protein expression and deposition. Endocrinology 2001; 142: 926–939. [DOI] [PubMed] [Google Scholar]
- 35. Liu S, Guo R, Simpson LG, Xiao Z‐S, Burnham CE, Quarles LD. Regulation of fibroblastic growth factor 23 expression but not degradation by PHEX. J Biol Chem 2003; 278: 37419–37426. [DOI] [PubMed] [Google Scholar]
- 36. Larsson T, Marsell R, Schipani E, Ohlsson C, Ljunggren O, Tenenhouse HS, et al Transgenic mice expressing fibroblast growth factor 23 under the control of the alpha1(I) collagen promoter exhibit growth retardation, osteomalacia, and disturbed phosphate homeostasis. Endocrinology 2004; 145: 3087–3094. [DOI] [PubMed] [Google Scholar]
- 37. Tenenhouse HS, Beck L. Renal na(+)‐phosphate cotransporter gene expression in X‐linked Hyp and Gy mice. Kidney Int 1996; 49: 1027–1032. [DOI] [PubMed] [Google Scholar]
- 38. Burnett SM, Gunawardene SC, Bringhurst FR, Juppner H, Lee H, Finkelstein JS. Regulation of C‐terminal and intact FGF‐23 by dietary phosphate in men and women. J Bone Miner Res 2006; 21: 1187–1196. [DOI] [PubMed] [Google Scholar]
- 39. Shimada T, Mizutani S, Muto T, Yoneya T, Hino R, Takeda S, et al Cloning and characterization of FGF23 as a causative factor of tumor‐induced osteomalacia. Proc Natl Acad Sci U S A 2001; 98: 6500–6505. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Shimada T, Hasegawa H, Yamazaki Y, Muto T, Hino R, Takeuchi Y, et al FGF‐23 is a potent regulator of vitamin D metabolism and phosphate homeostasis. J Bone Miner Res 2004; 19: 429–435. [DOI] [PubMed] [Google Scholar]
- 41. Bai XY, Miao D, Goltzman D, Karaplis AC. The autosomal dominant hypophosphatemic rickets R176Q mutation in fibroblast growth factor 23 resists proteolytic cleavage and enhances in vivo biological potency. J Biol Chem 2003; 278: 9843–9849. [DOI] [PubMed] [Google Scholar]
- 42. Econs MJ, McEnery PT. Autosomal dominant hypophosphatemic rickets/osteomalacia: clinical characterization of a novel renal phosphate‐wasting disorder. J Clin Endocrinol Metab 1997; 82: 674–681. [DOI] [PubMed] [Google Scholar]
- 43. Imel EA, Peacock M, Gray AK, Padgett LR, Hui SL, Econs MJ. Iron modifies plasma FGF23 differently in autosomal dominant hypophosphatemic rickets and healthy humans. J Clin Endocrinol Metab 2011; 96: 3541–3549. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Farrow EG, Yu X, Summers LJ, Davis SI, Fleet JC, Allen MR, et al Iron deficiency drives an autosomal dominant hypophosphatemic rickets (ADHR) phenotype in fibroblast growth factor‐23 (Fgf23) knock‐in mice. Proc Natl Acad Sci U S A 2011; 108: E1146–E1155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Clinkenbeard EL, Farrow EG, Summers LJ, Cass TA, Roberts JL, Bayt CA, et al Neonatal iron deficiency causes abnormal phosphate metabolism by elevating FGF23 in normal and ADHR mice. J Bone Miner Res 2014; 29: 361–369. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Imel EA, Gray AK, Padgett LR, Econs MJ. Iron and fibroblast growth factor 23 in X‐linked hypophosphatemia. Bone 2014; 60: 87–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Harrell RM, Lyles KW, Harrelson JM, Friedman NE, Drezner MK. Healing of bone disease in X‐linked hypophosphatemic rickets/osteomalacia. Induction and maintenance with phosphorus and calcitriol. J Clin Invest 1985; 75: 1858–1868. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Glorieux FH, Marie PJ, Pettifor JM, Delvin EE. Bone response to phosphate salts, ergocalciferol, and calcitriol in hypophosphatemic vitamin D‐resistant rickets. N Engl J Med 1980; 303: 1023–1031. [DOI] [PubMed] [Google Scholar]
- 49. Petersen DJ, Boniface AM, Schranck FW, Rupich RC, Whyte MP. X‐linked hypophosphatemic rickets: a study (with literature review) of linear growth response to calcitriol and phosphate therapy. J Bone Miner Res 1992; 7: 583–597. [DOI] [PubMed] [Google Scholar]
- 50. Imel EA, DiMeglio LA, Hui SL, Carpenter TO, Econs MJ. Treatment of X‐linked hypophosphatemia with calcitriol and phosphate increases circulating fibroblast growth factor 23 concentrations. J Clin Endocrinol Metab 2010; 95: 1846–1850. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Liu S, Tang W, Zhou J, Stubbs JR, Luo Q, Pi M, et al Fibroblast growth factor 23 is a counter‐regulatory phosphaturic hormone for vitamin D. J Am Soc Nephrol 2006; 17: 1305–1315. [DOI] [PubMed] [Google Scholar]
- 52. Carpenter TO, Insogna KL, Zhang JH, Ellis B, Nieman S, Simpson C, et al Circulating levels of soluble klotho and FGF23 in X‐linked hypophosphatemia: circadian variance, effects of treatment, and relationship to parathyroid status. J Clin Endocrinol Metab 2010; 95: E352–E357. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Gizard A, Rothenbuhler A, Pejin Z, Finidori G, Glorion C, de Billy B, et al Outcomes of orthopedic surgery in a cohort of 49 patients with X‐linked hypophosphatemic rickets (XLHR). Endocr Connect 2017; 6: 566–573. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Goodyer PR, Kronick JB, Jequier S, Reade TM, Scriver CR. Nephrocalcinosis and its relationship to treatment of hereditary rickets. J Pediatr 1987; 111: 700–704. [DOI] [PubMed] [Google Scholar]
- 55. Reusz GS, Hoyer PF, Lucas M, Krohn HP, Ehrich JH, Brodehl J. X linked hypophosphataemia: treatment, height gain, and nephrocalcinosis. Arch Dis Child 1990; 65: 1125–1128. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Taylor A, Sherman NH, Norman ME. Nephrocalcinosis in X‐linked hypophosphatemia: effect of treatment versus disease. Pediatr Nephrol 1995; 9: 173–175. [DOI] [PubMed] [Google Scholar]
- 57. Verge CF, Lam A, Simpson JM, Cowell CT, Howard NJ, Silink M. Effects of therapy in X‐linked hypophosphatemic rickets. N Engl J Med 1991; 325: 1843–1848. [DOI] [PubMed] [Google Scholar]
- 58. Nakamura Y, Takagi M, Takeda R, Miyai K, Hasegawa Y. Hypertension is a characteristic complication of X‐linked hypophosphatemia. Endocr J 2017; 64: 283–289. [DOI] [PubMed] [Google Scholar]
- 59. Carpenter TO, Mitnick MA, Ellison A, Smith C, Insogna KL. Nocturnal hyperparathyroidism: a frequent feature of X‐linked hypophosphatemia. J Clin Endocrinol Metab 1994; 78: 1378–1383. [DOI] [PubMed] [Google Scholar]
- 60. Makitie O, Kooh SW, Sochett E. Prolonged high‐dose phosphate treatment: a risk factor for tertiary hyperparathyroidism in X‐linked hypophosphatemic rickets. Clin Endocrinol (Oxf) 2003; 58: 163–168. [DOI] [PubMed] [Google Scholar]
- 61. Rivkees SA, el‐Hajj‐Fuleihan G, Brown EM, Crawford JD. Tertiary hyperparathyroidism during high phosphate therapy of familial hypophosphatemic rickets. J Clin Endocrinol Metab 1992; 75: 1514–1518. [DOI] [PubMed] [Google Scholar]
- 62. Rhee Y, Bivi N, Farrow E, Lezcano V, Plotkin LI, White KE, et al Parathyroid hormone receptor signaling in osteocytes increases the expression of fibroblast growth factor‐23 in vitro and in vivo . Bone 2011; 49: 636–643. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Yavropoulou MP, Kotsa K, Gotzamani Psarrakou A, Papazisi A, Tranga T, Ventis S, et al Cinacalcet in hyperparathyroidism secondary to X‐linked hypophosphatemic rickets: case report and brief literature review. Hormones (Athens) 2010; 9: 274–278. [DOI] [PubMed] [Google Scholar]
- 64. Raeder H, Shaw N, Netelenbos C, Bjerknes R. A case of X‐linked hypophosphatemic rickets: complications and the therapeutic use of cinacalcet. Eur J Endocrinol 2008; 159 (Suppl. 1): S101–S105. [DOI] [PubMed] [Google Scholar]
- 65. Alon US, Levy‐Olomucki R, Moore WV, Stubbs J, Liu S, Quarles LD. Calcimimetics as an adjuvant treatment for familial hypophosphatemic rickets. Clin J Am Soc Nephrol 2008; 3: 658–664. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Alon US, Jarka D, Monachino PJ, Sebestyen VanSickle J, Srivastava T. Cinacalcet as an alternative to phosphate therapy in X‐linked hypophosphataemic rickets. Clin Endocrinol (Oxf) 2017; 87: 114–116. [DOI] [PubMed] [Google Scholar]
- 67. Nesbitt T, Lobaugh B, Drezner MK. Calcitonin stimulation of renal 25‐hydroxyvitamin D‐1 alpha‐hydroxylase activity in hypophosphatemic mice. Evidence that the regulation of calcitriol production is not universally abnormal in X‐linked hypophosphatemia. J Clin Invest 1987; 79: 15–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Econs MJ, Lobaugh B, Drezner MK. Normal calcitonin stimulation of serum calcitriol in patients with X‐linked hypophosphatemic rickets. J Clin Endocrinol Metab 1992; 75: 408–411. [DOI] [PubMed] [Google Scholar]
- 69. Plotkin LI, Weinstein RS, Parfitt AM, Roberson PK, Manolagas SC, Bellido T. Prevention of osteocyte and osteoblast apoptosis by bisphosphonates and calcitonin. J Clin Invest 1999; 104: 1363–1374. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. van Boekel G, Ruinemans‐Koerts J, Joosten F, Dijkhuizen P, van Sorge A, de Boer H. Tumor producing fibroblast growth factor 23 localized by two‐staged venous sampling. Eur J Endocrinol 2008; 158: 431–437. [DOI] [PubMed] [Google Scholar]
- 71. Liu ES, Carpenter TO, Gundberg CM, Simpson CA, Insogna KL. Calcitonin administration in X‐linked hypophosphatemia. N Engl J Med 2011; 364: 1678–1680. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Folsom LJ, Imel EA. Hyperphosphatemic familial tumoral calcinosis: genetic models of deficient FGF23 action. Curr Osteoporos Rep 2015; 13: 78–87. [DOI] [PubMed] [Google Scholar]
- 73. Sullivan R, Abraham A, Simpson C, Olear E, Carpenter T, Deng Y, et al Three‐month randomized clinical trial of nasal calcitonin in adults with X‐linked hypophosphatemia. Calcif Tissue Int 2018; 102: 666–670. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. White KE, Jonsson KB, Carn G, Hampson G, Spector TD, Mannstadt M, et al The autosomal dominant hypophosphatemic rickets (ADHR) gene is a secreted polypeptide overexpressed by tumors that cause phosphate wasting. J Clin Endocrinol Metab 2001; 86: 497–500. [DOI] [PubMed] [Google Scholar]
- 75. Folpe AL, Fanburg‐Smith JC, Billings SD, Bisceglia M, Bertoni F, Cho JY, et al Most osteomalacia‐associated mesenchymal tumors are a single histopathologic entity ‐ an analysis of 32 cases and a comprehensive review of the literature. American Journal of Surgical Pathology 2004; 28: 1–30. [DOI] [PubMed] [Google Scholar]
- 76. Seufert J, Ebert K, Muller J, Eulert J, Hendrich C, Werner E, et al Octreotide therapy for tumor‐induced osteomalacia. N Engl J Med 2001; 345: 1883–1888. [DOI] [PubMed] [Google Scholar]
- 77. Nasu T, Kurisu S, Matsuno S, Tatsumi K, Kakimoto T, Kobayashi M, et al Tumor‐induced hypophosphatemic osteomalacia diagnosed by the combinatory procedures of magnetic resonance imaging and venous sampling for FGF23. Intern Med 2008; 47: 957–961. [DOI] [PubMed] [Google Scholar]
- 78. Hesse E, Moessinger E, Rosenthal H, Laenger F, Brabant G, Petrich T, et al Oncogenic osteomalacia: exact tumor localization by co‐registration of positron emission and computed tomography. J Bone Miner Res 2007; 22: 158–162. [DOI] [PubMed] [Google Scholar]
- 79. Hodgson SF, Clarke BL, Tebben PJ, Mullan BP, Cooney WP 3rd, Shives TC. Oncogenic osteomalacia: localization of underlying peripheral mesenchymal tumors with use of Tc 99m sestamibi scintigraphy. Endocr Pract 2006; 12: 35–42. [DOI] [PubMed] [Google Scholar]
- 80. Khadgawat R, Singh Y, Kansara S, Tandon N, Bal C, Seith A, et al PET/CT localisation of a scapular haemangiopericytoma with tumour‐induced osteomalacia. Singapore Med J 2009; 50: e55–e57. [PubMed] [Google Scholar]
- 81. Andreopoulou P, Dumitrescu CE, Kelly MH, Brillante BA, Cutler Peck CM, Wodajo FM, et al Selective venous catheterization for the localization of phosphaturic mesenchymal tumors. J Bone Miner Res 2011; 26: 1295–1302. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Lee JC, Jeng YM, Su SY, Wu CT, Tsai KS, Lee CH, et al Identification of a novel FN1‐FGFR1 genetic fusion as a frequent event in phosphaturic mesenchymal tumour. J Pathol 2015; 235: 539–545. [DOI] [PubMed] [Google Scholar]
- 83. White KE, Cabral JM, Davis SI, Fishburn T, Evans WE, Ichikawa S, et al Mutations that cause osteoglophonic dysplasia define novel roles for FGFR1 in bone elongation. Am J Hum Genet 2005; 76: 361–367. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Yamazaki Y, Tamada T, Kasai N, Urakawa I, Aono Y, Hasegawa H, et al Anti‐FGF23 neutralizing antibodies show the physiological role and structural features of FGF23. J Bone Miner Res 2008; 23: 1509–1518. [DOI] [PubMed] [Google Scholar]
- 85. Aono Y, Yamazaki Y, Yasutake J, Kawata T, Hasegawa H, Urakawa I, et al Therapeutic effects of anti‐FGF23 antibodies in hypophosphatemic rickets/osteomalacia. J Bone Miner Res 2009; 24: 1879–1888. [DOI] [PubMed] [Google Scholar]
- 86. Carpenter TO, Imel EA, Ruppe MD, Weber TJ, Klausner MA, Wooddell MM, et al Randomized trial of the anti‐FGF23 antibody KRN23 in X‐linked hypophosphatemia. J Clin Invest 2014; 124: 1587–1597. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. Imel EA, Zhang X, Ruppe MD, Weber TJ, Klausner MA, Ito T, et al Prolonged correction of serum phosphorus in adults with X‐linked hypophosphatemia using monthly doses of KRN23. J Clin Endocrinol Metab 2015; 100: 2565–2573. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Zhang X, Peyret T, Gosselin NH, Marier JF, Imel EA, Carpenter TO. Population pharmacokinetic and pharmacodynamic analyses from a 4‐month intradose escalation and its subsequent 12‐month dose titration studies for a human monoclonal anti‐FGF23 antibody (KRN23) in adults with X‐linked hypophosphatemia. J Clin Pharmacol 2016; 56: 429–438. [DOI] [PubMed] [Google Scholar]
- 89. Whyte MP, Fujita KP, Moseley S, Thompson DD, McAlister WH. Validation of a novel scoring system for changes in skeletal manifestations of Hypophosphatasia in newborns, infants, and children: the radiographic global impression of change scale. J Bone Miner Res 2018; 33: 868–874. [DOI] [PubMed] [Google Scholar]
- 90. Whyte M, Imel EA, Portale AA, Boot AM, Hogler W, Linglart A, et al Burosumab, a fully human anti‐FGF23 monoclonal antibody, for X‐linked hypophosphatemia (XLH): results from two phase 2 trials in affected children 1–12 years old. In: ENDO 2018, Chicago, Il, 2018: Abstract MON‐473.
- 91. FDA . CRYSVITA® (burosumab‐twza) injection, for subcutaneous use; Initial U.S. Approval: 2018. In, edFDA. Available at https://www.accessdata.fda.gov/drugsatfda_docs/nda/2018/761068Orig1s000TOC.cfm (last accessed 18 October 2018).
- 92. Shalhoub V, Shatzen EM, Ward SC, Davis J, Stevens J, Bi V, et al FGF23 neutralization improves chronic kidney disease‐associated hyperparathyroidism yet increases mortality. J Clin Invest 2012; 122: 2543–2553. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. EMA . Crysvita: EPAR ‐ Product Information In. Available at http://www.ema.europa.eu/ema/index.jsp?curl=pages/medicines/human/medicines/004275/human_med_002224.jsp&mid=WC0b01ac058001d124 (last accessed 18 October 2018).
- 94. Harding SD, Sharman JL, Faccenda E, Southan C, Pawson AJ, Ireland S, et al The IUPHAR/BPS Guide to PHARMACOLOGY in 2018: updates and expansion to encompass the new guide to IMMUNOPHARMACOLOGY. Nucl Acids Res 2018; 46: D1091–106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Alexander SPH, Fabbro D, Kelly E, Marrion NV, Peters JA, Faccenda E, et al The Concise Guide to PHARMACOLOGY 2017/18: Enzymes. Br J Pharmacol 2017; 174: S272–S359. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Alexander SPH, Kelly E, Marrion NV, Peters JA, Faccenda E, Harding SD, et al The Concise Guide to PHARMACOLOGY 2017/18: Transporters. Br J Pharmacol 2017; 174: S360–S446. [DOI] [PMC free article] [PubMed] [Google Scholar]
