You are on page 1of 11

British Journal of Clinical Br J Clin Pharmacol (2019) 85 1188–1198 1188

Pharmacology

REVIEW-THEMED ISSUE
Pharmacological management of X-linked
hypophosphataemia
Correspondence Erik A. Imel, MD, Gatch Hall, Suite 380 F, 1120 W. Michigan St. Indianapolis, IN 46202-5111. Tel.: +1 317 274 1339;
Fax: +1 317 278 0658; E-mail: eimel@iu.edu

Received 17 July 2018; Revised 5 September 2018; Accepted 5 September 2018

Erik A. Imel1,2 and Kenneth E. White3,4


1
Department of Medicine, Division of Endocrinology and Metabolism, Indiana University School of Medicine, Indianapolis, IN, USA, 2Department of
Pediatrics, Section of Endocrinology and Diabetology, Indiana University School of Medicine, Indianapolis, IN, USA, 3Department of Medical and
Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA, and 4Department of Medicine, Division of Nephrology, Indiana
University School of Medicine, Indianapolis, IN, USA

Keywords burosumab-twza, fibroblast growth factor 23, PHEX, X-linked hypophosphataemia

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.

Introduction Genetic cause


XLH is a rare bone disease affecting an estimated 1:20–
X-linked hypophosphataemia (XLH) is a rare metabolic 25 000 persons [1, 2] (OMIM no. 307800). Deleterious mu-
bone disease with significant clinical consequences in both tations in the phosphate regulating gene with homologies
children and adults, due largely to fibroblast growth to endopeptidases on the X chromosome (PHEX) [3] cause
factor 23 (FGF23) excess and resulting chronic this X-linked dominant condition. The female to male ratio
hypophosphataemia. In the past, treatment has been re- is about 2:1. Sporadic cases frequently occur. Other genes
stricted to administering phosphorus and activated vitamin having different inheritance patterns cause phenotypically
D, with limitations in effectiveness, and significant risk for similar disorders. PHEX mutations cause increased plasma
important adverse events. Over the past 2 decades research FGF23 concentrations, leading to impaired renal reabsorp-
into XLH mechanisms has generated novel potential targets tion of phosphorus, hypophosphataemia and impaired acti-
for treatment, including the recent regulatory approvals of vation of vitamin D [2, 4].
burosumab-twza/CRYSVITA (henceforth referred to just
as burosumab) for clinical use in patients with XLH. This pa-
per will review the clinical consequences of XLH, the patho- Clinical manifestations
physiology involved and various approaches to its medical XLH typically presents in early childhood with manifesta-
management. tions of rickets, although family history may lead to testing

DOI:10.1111/bcp.13763 © 2018 The British Pharmacological Society


Pharmacological management of X-linked hypophosphataemia

before visible features appear. Unfortunately, clinical labora- formation. The pathophysiology of enthesopathy in XLH is
tories often fail to report age-appropriate normal ranges, lead- uncertain, although it may be due to direct effects of FGF23.
ing to missed diagnoses, since adult normal phosphorus Enthesopathy occurs in multiple hypophosphataemic mouse
values are much lower than paediatric (especially infant) models having FGF23 excess [23, 24]. FGF receptor 3 and
values [2]. Both children and adults with XLH have low phos- klotho expression in fibrocartilage of the entheses make di-
phorus concentrations for age, and impaired renal tubular rect signalling at least theoretically possible [24].
phosphorus reabsorption, typically assessed as a low Enthesopathy causes joint stiffness and limitations in
transport maximum for phosphorus adjusted for glomerular range of motion which may be extreme. When involving
filtration rate (TmP/GFR) [2, 4]. Serum alkaline phosphatase the anterior and posterior spinal ligaments, patients lose flex-
activity is typically increased for age in children, although ion, extension and rotational movement of the neck and
this is more variable in adults [2, 4]. spine. Sometimes patients develop spinal nerve or spinal cord
At birth, length is usually normal and legs are not usually compression requiring surgical intervention, although the
bowed [5–7]. Bowing and impaired linear growth become incidence of this is uncertain. Enthesopathy is more common
more evident with weight bearing, especially between ages 1 in men, and with increasing age and BMI [19], affecting be-
and 2 years [2, 5, 6, 8, 9]. Childhood features include bowing tween 30 and 100% of adults [10, 11, 19, 21, 22, 24].
of the femora and tibiae leading to genu varus or sometimes Osteoarthritis is probably a consequence of chronic ab-
valgus appearance, widening of the growth plates at the end normalities in joint alignment and gait causing degenerative
of long bones, abnormalities of the skull shape, sometimes changes in joint cartilage and bony articulation surfaces. Os-
bone pain and occasionally delays in gross motor milestones teoarthritis might be exacerbated by enthesopathy. Joint re-
(such as difficulties in walking or running) [2, 9, 10]. placements are common surgeries in XLH patients,
Radiographic growth plate abnormalities are similar to nutri- although the frequency of such procedures is uncertain [25].
tional rickets, along with diaphyseal bowing, often with me- Up to 82% of adults with XLH complain of joint pain and
dial cortical thickening, and torsion of weight-bearing long stiffness, and about 50% report using pain medications
bones. Skeletal severity and the response to medical weekly to daily [19, 22].
management are highly variable [2, 6, 10]. Patients often re- Muscle weakness, a feature of hypophosphataemia, is
quire corrective surgical procedures to straighten lower ex- present in both the Hyp mouse and patients with XLH
tremities. Short stature and gait abnormalities persist into [22, 26, 27]. However, in XLH patients it is difficult to
adulthood [2, 6, 8, 10, 11]. separate the effects on mobility from skeletal deforma-
Cranial bones are also affected in XLH, most notably with tion, joint abnormalities, and bone pain from those due
frontal bossing and dolicocephally [12–14]. Flattening of the to muscle weakness.
cranial base leads to decreased depth of the posterior fossa Overall, the lifetime burden of illness is high in XLH,
predisposing to Chiari malformations [13], occurring in up complicated often by a lack of understanding of the disease
to 44% of XLH patients [12]. For some patients, craniosynos- and its management by both patients and clinical providers.
tosis of the sagittal suture requires craniotomy [14]. Not surprisingly, with the growth abnormalities, skeletal de-
XLH predisposes to dental abscesses due to a combination formities, muscle weakness, and bone and joint pain, quality
of intrinsic effects of Phex deficiency and associated of life impairments are documented in both children and
hypophosphataemia leading to under-mineralized dentin adults with XLH [28–30].
and cementum [15–18]. Severe dental disease (abscesses and
periodontitis) affects 61–78% of patients with XLH [19, 20].
Retrospective studies suggest that treating XLH with phos- Molecular pathophysiology
phate salts and active vitamin D decreases the occurrence of XLH is fully penetrant with highly variable severity. The
dental abscesses and periodontitis [19, 20]. Hyp Consortium determined that XLH is caused by
XLH does not negatively influence lifespan, so one would inactivating mutations in the PHEX gene [3]. PHEX en-
expect at least three quarters of XLH patients to be adults. As codes a protein that structurally resembles the M13 family
such, although commonly thought of as a paediatric bone of membrane-bound metalloproteases. Members of this en-
disease, adults bear a considerable burden of the conse- zyme class include single transmembrane proteins with
quences of this lifelong condition. The most debilitating fea- large extracellular domains such as neutral endopeptidase
tures of XLH are experienced by adults: bone pain, and endothelin converting enzymes 1 and 2 (ECE-1 and
pseudofractures, enthesopathy and osteoarthritis [11, 21, 22]. ECE-2) [3, 31]. This family is known to cleave small pep-
Osteomalacic bone pain is common [22] and debilitating. tides, although the endogenous PHEX protein’s substrate
Although complete fractures are not more common in XLH, has yet to be fully confirmed. Close to 300 inactivating
patients with XLH are prone to pseudofractures [11]. PHEX mutations have been described from XLH patients,
Pseudofractures occasionally occur in children, but are seen including variations that lead to missense, nonsense, frame
in up to half of adults in cross sectional studies [21, 22], shift, and exon splicing alterations. PHEX shows the
although the lifetime incidence might be higher. highest expression in bone cells including osteoblasts and
Pseudofractures may progress to complete fractures and may osteocytes, and odontoblasts in teeth, as well as lower ex-
require surgical intervention. Skeletal healing after fracture or pression in the parathyroid glands, lung, brain and skeletal
surgery may be delayed. muscle [31, 32].
Enthesopathy involves the calcification of tendons and Although the PHEX mutations are predicted to cause loss
ligaments, typically beginning near the bony attachment of function, the mechanisms whereby this leads to changes
sites, and progressing to enthesophyte or osteophyte in bone function remain elusive. The Hyp mouse model of

Br J Clin Pharmacol (2019) 85 1188–1198 1189


E. A. Imel and K. E. White

XLH, has a 30 deletion in PHEX, and has been a very useful Medical management
tool for the in vivo study of the XLH phenotype [31]. Similar
to XLH patients, this model has approximately a 10-fold in-
crease in serum FGF23 (although in patients with XLH, serum
Phosphate salts and active vitamin D
For about 4 decades, children and adults with XLH have been
FGF23 is typically less severely elevated), and manifests the
medically managed using pharmacological doses of phos-
XLH biochemical syndrome including hypophosphataemia
with inappropriately normal 1,25-dihydroxyvitamin D3 phate salts and active vitamin D, attempting to counter the
[1,25(OH)2D] and normocalcaemia [33]. Like patients, the effects of FGF23 excess. Doses reported vary widely in the
Hyp mice display growth retardation and bone mineraliza- literature [2]. A US-based group of experts recommended tar-
tion defects. Interestingly, loss of Phex protein function get doses of calcitriol 20–30 ng kg–1 daily and phosphate
was associated with a differentiation defect in osteocytes 20–40 mg kg–1 daily, each in divided doses [2]. European
from Hyp mice [34]. This mouse line had altered develop- experts recommended 1–3 μg day–1 of alfacalcidiol or
mental transitions from osteoblasts to osteocytes, with in- 0.5–1.5 μg day–1 of calcitriol, and 40–60 mg kg–1 daily of
appropriate expression of cell matrix genes [34]. Indeed phosphate [10]. However, no study has systematically evalu-
Hyp bones had over-expression of type I collagen, as well ated the optimal target dose ranges. Clinicians start at lower
as altered expression of matrix proteins such as bone doses and titrate to target doses primarily based on gastroin-
sialoprotein [34]. Fgf23 mRNA is elevated in Hyp bone testinal tolerance, with frequent laboratory monitoring to
[33] and in isolated Hyp osteoblasts/osteocytes [35]. These avoid hypercalciuria, hypercalcaemia, hyperphosphataemia
cultures also fail to mineralize fully, consistent with an in- or impairments in renal function. This strategy does not ef-
trinsic cell defect. However, the cellular pathophysiological fectively normalize the serum phosphorus. In fact, attempts
mechanisms caused by Phex deficiency that result in al- to target normal phosphorus concentrations might be a con-
tered FGF23 gene expression and sustained FGF23 protein tributing reason for the high risk of hyperparathyroidism and
elevation remain elusive. nephrocalcinosis with these agents [2]. Monitoring patients
on treatment is complicated by many factors, and most labo-
ratory monitoring addresses safety (serum calcium, phospho-
FGF23 control of phosphate and active rus, creatinine, PTH, urine calcium, urine creatinine).
vitamin D Treatment with active vitamin D and phosphate decreases
The primary kidney transport protein responsible for phos- alkaline phosphatase and, in bone biopsy studies, improves
phate reabsorption in the proximal tubule is the type II the osteomalacia of XLH [47, 48]. However, the skeletal re-
sodium-phosphate cotransporter, or NaPi-IIa. Transgenic sponse varies widely. Some children straighten leg deformi-
FGF23 mice had dramatic decreases in protein expression of ties and improve their growth, while others persist in
NaPi-IIa and a related transporter, NaPi-IIc [36]. The Hyp skeletal deformities and short stature. The reason for good re-
mouse has a 50% reduction in proximal tubule NaPi-IIa ex- sponders vs. poor responders is not solely an issue of compli-
pression [37]. In normal individuals, phosphate depletion is ance [49]. Treating XLH with vitamin D analogues and
a strong stimulus for increasing serum 1,25(OH)2D [38]. phosphate also increases plasma FGF23 concentrations [50–
However, in XLH patients, hypophosphataemia is accom- 52], which theoretically could blunt therapeutic effective-
panied by low or inappropriately normal 1,25(OH)2D ness, although the true consequences remain unknown.
concentrations, due to effects of FGF23. In mice implanted Many children with XLH have a variable but progressive
with cells expressing FGF23, or injected with FGF23 itself, decline in height Z-scores, often worsening during puberty
mRNA and protein expression levels of the activating enzyme despite therapy with vitamin D analogues and phosphate
25-hydroxyvitamin D3 1-α-hydroxylase (Cyp27b1) was de- [8]. Studies suggest that beginning treatment prior to age
creased, while the catabolic 1,25-(OH)2D 24-hydroxylase 1 year improves height outcomes [6].
(Cyp24a1) is increased with high FGF23 [39–41]. Thus, the When leg deformities persist, the optimal timing of cor-
effects of FGF23 on the renal vitamin D metabolic enzymes is rective surgery remains uncertain. In a retrospective study,
responsible for the reductions in serum 1,25(OH)2D concen- 29% of XLH patients had recurrence of deformity after their
trations observed in XLH patients. first corrective surgery [53]. Although the differences were
Patients with autosomal dominant hypophosphataemic not significant, those having earlier corrective procedures
rickets (ADHR) also have elevated FGF23 but some patients were more likely to require additional procedures. This find-
display waxing and waning of the disease symptoms [42]. ing may be confounded by more severely affected patients
Studying a large ADHR kindred, we documented that the having earlier surgeries.
ADHR disease state and circulating levels of intact FGF23 cor- Unlike nutritional rickets, which is truly cured by adequate
related strongly with iron deficiency and were reciprocal to vitamin D repletion, the homeostatic defect in phosphorus
serum iron concentrations [43], which was borne out in an handling is lifelong in XLH. However, because of the known
ADHR knock in animal model to be due to increased bone risks of XLH therapy with active vitamin D and phosphate,
Fgf23 mRNA production during experimental iron depletion general practice has been to stop treatment at the end of
[44, 45]. However, intact FGF23 levels in XLH patients were growth, and restart if developing clinical issues associated
unrelated to serum iron concentration [46]. A clinical trial is with active osteomalacia [2]. This has led to some confusion
underway to place ADHR patients on low-dose oral iron over whether or not adults benefit from treatment, and mis-
repletion (ClinicalTrials.gov NCT02233322), thus this regi- interpretation of XLH as a childhood disease. Although some
men may be the optimal treatment for ADHR, but not XLH adults tolerate stopping treatment with few symptoms, at
(see below). least for a time, many develop active osteomalacic symptoms,

1190 Br J Clin Pharmacol (2019) 85 1188–1198


Pharmacological management of X-linked hypophosphataemia

bone pain, muscle weakness and pseudofractures, which secretion. Calcitonin decreased serum FGF23 in a patient
demonstrate varying degrees of improvement during treat- with tumour-induced osteomalacia (TIO) [70]. A single
ment with active vitamin D and phosphate [22]. calcitonin injection decreased serum FGF23 and increased
Important side effects limit this therapy. Doses of phos- serum phosphorus transiently in patients with XLH,
phate may be limited by gastrointestinal symptoms, although not in healthy controls [71]. To make the
nephrocalcinosis, ectopic calcification and hyperparathy- calcitonin story more confusing, case reports indicate
roidism, while active vitamin D may also contribute to calcitonin increases urinary phosphorus excretion in
nephrocalcinosis or other ectopic calcification. Nephro- hyperphosphatemic tumoural calcinosis (a condition of
calcinosis is reported in 50–80% of XLH patients receiving FGF23 deficiency rather than excess) [72], which would be
active vitamin D and phosphate, and appears to relate to opposite of the goal of XLH management. A 3-month-long
episodes of hypercalciuria [54–57]. Most nephrocalcinosis is blinded randomized controlled clinical trial of monotherapy
mild, but occasionally patients develop chronic kidney dis- with nasal calcitonin 400 units daily for XLH failed to
ease (CKD), the prevalence of which is uncertain, but progres- demonstrate improvements in serum phosphorus, TmP/GFR
sion to end-stage renal disease is rare. Hypertension has been or FGF23 [73]. Given the current lack of data indicating
reported in 27% of XLH patients and may relate in part to benefit, calcitonin should not be used for XLH.
nephrocalcinosis and CKD [58].
Hyperparathyroidism in XLH is complex. Nearly half of
treatment naïve children with XLH have elevated serum
Fibroblast growth factor receptor antagonism in
PTH [59], which often decreases after starting active vitamin TIO
D. Serum PTH correlates with FGF23 concentrations in un- TIO (OMIM 605380) is another syndrome of excess fibro-
treated patients [52]. However, phosphate doses also lead to blast growth factor receptor (FGF23) [39, 74]. These tu-
secondary hyperparathyroidism, probably through transient mours are classified under the collective term of
decreases in serum calcium following every dose. Some pa- phosphaturic mesenchymal tumour, mixed connective tissue var-
tients with secondary hyperparathyroidism progress to ter- iant (PMTMCT) or phosphaturic mesenchymal tumour [75].
tiary hyperparathyroidism with multigland hyperplasia and Complete tumour resection is the most straight forward
require surgical intervention to control hypercalcaemia [60, cure for TIO. For tumour localization, imaging techniques
61]. Prior to the development of parathyroid autonomy, ade- using radiolabelled octreotide [76] as well as magnetic reso-
quate doses of active vitamin D and lowering doses of phos- nance imaging [77], computed tomography [78], whole
phate may combat the rise of PTH. body sestamibi scanning [79], and Ga68-DOTA-octreotide
positron emission tomography/computed tomography imag-
ing [80] may be useful. Selective venous sampling for FGF23
Novel targets levels has been attempted to locate tumours as well [77, 81].
Given the limitations of effect and the significant risks of Unfortunately, many PMTMCTs are small in size and remain
treating XLH with active vitamin D and phosphate salts, the difficult to localize, necessitating medical management.
need for improved treatment options has led to development In a group of 15 TIO tumours analysed through next-
of other strategies. We will focus on those targets that have generation sequencing, a fibronectin (FN1)-FGFR1 fusion
some clinical/translational data in humans. gene was detected in 60% of the tested samples [82]. The fu-
sion protein is predicted to express portions of the three ex-
Calcimimetics. PTH stimulates FGF23 expression in tracellular FGF-binding (Ig-like) domains [82], therefore
osteocytes [62], which could exacerbate ligand-activated receptor signalling could perhaps occur. It
hypophosphataemia in XLH. Since hyperparathyroidism is not established however, whether the fusion gene is causa-
also complicates XLH treatment, cinacalcet, a tive of the TIO tumour or is a consequence of the
calcimimetic, has been used to manage hyperparathyroidism tumourigenesis. Activating FGFR1 mutations are associated
in cases of XLH [63, 64]. Short term studies of cinacalcet in with osteoglophonic dysplasia, a disease of dwarfism as well
children with XLH demonstrated suppression of the PTH as craniosynostosis [83]. Some osteoglophonic dysplasia
surge that follows doses of phosphate [65], and improved patients also have significantly elevated FGF23 and
TmP/GFR and serum phosphorus. In a case report, one hypophosphataemia. Thus, inhibitors of FGF binding or
child developed a severe aversion to, and refusal of, his FGFR activity could be useful pharmacological treatments
oral phosphate doses. Although he did not have for TIO. Such inhibitors may block FGFR auto- and ligand-
hyperparathyroidism, treatment with cinacalcet, calcitriol and Klotho-dependent FGFR dimerization, and may reduce
and calcium led to improvements in serum phosphorus signalling through FGFRs along with directly inhibiting
and rickets, although serum FGF23 still increased [66]. FGF23 bioactivity. However, the ubiquitous nature of FGFR1
Further studies are needed to determine what role could lead to off-target effects. Considering the difficulty with
cinacalcet should play, although it may be useful in targeting FGFR inhibitors to a specific tissue, another plausi-
tertiary hyperparathyroidism. ble approach for treating TIO patients until the tumour is
found may be monotherapy with anti-FGF23 antibody (see
Calcitonin. Calcitonin stimulates 1α-hydroxylase in the below) as is currently being tested in TIO and XLH
proximal renal tubule in Hyp mice [67], and transiently (ClinicalTrials.gov NCT02304367).
increases 1,25(OH)2D in subjects with XLH after a single
injection [68]. Osteocytes also express the calcitonin FGF23-blocking antibodies. Investigators at Kirin Pharma
receptor [69], and as such, calcitonin could influence FGF23 developed murine anti-FGF23 monoclonal antibodies that

Br J Clin Pharmacol (2019) 85 1188–1198 1191


E. A. Imel and K. E. White

bind separate isotopes on either the C-terminal or N-terminal generally stable through the 12 month extension, except for
portions of FGF23 [84], providing information on the the peak of 1,25(OH)2D, which decreased in magnitude over
structural interactions of FGF23 with its receptor/co- later doses [87]. Most subjects at least transiently achieved
receptor. Both antibodies interfered with FGFR signalling as normal serum phosphorus concentrations.
indicated by declines in Egr-1 reporter activity (a molecular Despite the transient increase in 1,25(OH)2D during each
marker for MAPK activation). The N-terminal antibody cycle, there was no systematic change in serum or urine cal-
interfered with binding of FGF23 to the FGFR, while the C- cium or in PTH. Kidney function also remained stable. Ten
terminal antibody interfered with binding to the coreceptor subjects had baseline nephrocalcinosis or nephrolithiasis,
klotho. Injecting either antibody into mice produced and no subject had worsening nephrocalcinosis [87].
generally similar effects: transient increases in 1,25(OH)2D Although this first multidose trial did not assess clinical
beginning within 3–5 hours, and in TmP/GFR and serum bone outcomes, the bone biomarkers P1NP, osteocalcin and
phosphorus after 16 h [84]. Combining the antibodies was bone alkaline phosphatase increased during the study period.
synergistic. Quality of life instruments (SF-36v2 and WOMAC) indicated
Hyp mice receiving subcutaneous injection of these anti- baseline impairments in bodily pain, physical function and
bodies in combination demonstrated a similar biochemical role limitations, along with stiffness. After 4 months there
response and time course after a single dose [85]. When juve- were improvements in the patients’ perception of their phys-
nile, 4-week-old Hyp mice were treated with weekly injections ical functioning and stiffness [28].
for 5 weeks, renal sodium phosphate cotransporter expres- In a recently published, open-label, phase 2 trial, 52 chil-
sion increased on immunohistochemistry, and serum phos- dren aged 5–12 years with XLH in Europe and the USA were
phorus improved in a dose-dependent manner along with randomized to receive burosumab subcutaneously every
increases in Cyp27b1 mRNA expression and serum 2 weeks or every 4 weeks for 64 weeks [29]. Patients were
1,25(OH)2D. As a result, mice had bone histomorphometric mostly prepubertal, although Tanner stage 2 subjects were
improvements, including decrease in osteoid thickness and allowed. All patients stopped phosphate and vitamin D ana-
volume toward normal, as well as increased mineral apposi- logues 2 weeks prior to screening. Since this was the first trial
tion rate, bone formation rate, and bone volume/total vol- conducted in children, the initial cohort started at very low
ume (BV/TV). This translated into improvements in the burosumab doses and titrated upwards targeting serum phos-
rachitic growth plate morphology, the growth and shape of phorus levels between 3.2 and 6.1 mg dl–1 (1.0–2.0 mmol l–1).
their tails, femora and tibiae, and in overall size of the mice Rickets was assessed using a rickets severity score (RSS) that
[26, 85]. Treating adult Hyp mice with anti-FGF23 antibodies rated radiographic appearance at the wrist and knee on a scale
also improved osteomalacia on histology, and increased mus- from 0–10 (10 being the worst). Differences between baseline
cle grip strength toward normal without changing muscle and follow-up radiographs were also assessed using a seven-
weight [26]. point ordinal Radiographic Global Impression of Change
Based on successes in the Hyp mouse, a fully human IgG1 (RGI-C) scale (ranging from –3 severe worsening, to 0 no
monoclonal antibody to bind FGF23 was developed for hu- change, to +3 complete healing) [89].
man trials (KRN23, now called burosumab or CRYSVITA). One limitation of this paediatric study was its lack of a
Table 1 summarizes key findings from the published clinical control group. However, all but one subject had been receiv-
trials. In the initial dose finding study, single doses of intrave- ing standard treatment with active vitamin D analogues and
nous or subcutaneous burosumab (or placebo) were adminis- phosphate salts up to the time of enrolment (for a mean of
tered to adults with XLH after stopping phosphate or active 6.9 years prior treatment). Thus, the baseline rickets severity
vitamin D [86]. Burosumab increased TmP/GFR, serum phos- at enrolment (ranging from RSS 0–4.5, mean 1.8 ± 1.1) repre-
phorus and 1,25(OH)2D in a dose dependent manner. The sents residual rickets despite prior treatment. Any improve-
half-life was longer for subcutaneous dosing (13–19 days) ments seen in trial occurred in the setting of switching from
than with intravenous dosing (8–12 days). Baseline FGF23 prior standard therapy to burosumab.
concentration did not predict AUC changes for any pharma- At week 40 the mean burosumab dose had been titrated to
codynamic parameter. 0.98 mg kg–1 every 2 weeks or 1.5 mg kg–1 every 4 weeks [29].
Twenty-eight adults with XLH stopped all XLH medica- Children receiving every 4 weeks dosing had pharmacody-
tions and enrolled in a Phase 2 dose-escalating multidose trial namic profiles of serum phosphorus, 1,25(OH)2D and
of burosumab administered subcutaneously every 4 weeks, TmP/GFR similar to that seen in adults [29]. However, the ev-
with doses ranging from 0.05 to 0.6 mg kg–1 and 22 subjects ery 2 weeks dosing group had more sustained improvements
continued in a 12-month extension with doses ranging from in these parameters, with less pronounced biochemical
0.1–1 mg kg–1 [87]. During the multidose trial, burosumab ex- trough effects, and serum phosphorus values more consis-
hibited first-order absorption and elimination kinetics, simi- tently in the normal range. Alkaline phosphatase is com-
lar to other monoclonal antibody therapies, with an monly elevated in XLH patients (mean 459 ± 105 units l–1 at
elimination half-life of 17.8 days [88]. baseline of this trial), which decreased by 20% at week 64.
The peak value of 1,25(OH)2D and serum phosphorus oc- Both groups demonstrated improvements in rickets by week
curred about 3–7 days after injection, while peak TmP/GFR 40 that were sustained at week 64. Rickets improvements
occurred about 7 days after injection [87]. After each peak, were numerically greater in those receiving every 2 weeks in-
the TmP/GFR, serum phosphorus and 1,25(OH)2D decreased jections and also in those having worse baseline rickets sever-
toward a trough level 4 weeks after injection that remained ity. Total RSS score decreased from mean 1.9 to 0.8 after every
generally above the baseline values. Peak and trough values 2 weeks dosing, and from 1.7 to 1.1 after every 4 weeks dos-
increased during the dose escalation phase, then remained ing. The mean total RGI-C was +1.57 at the end of study,

1192 Br J Clin Pharmacol (2019) 85 1188–1198


Pharmacological management of X-linked hypophosphataemia

Table 1
Key findings from the primary burosumab clinical trial publications in X-linked hypophosphataemia (XLH) to datea

Imel et al. JCEM 2015 [87],


Carpenter et al. Ruppe et al. Carpenter et al. Insogna et al.
Study [ref] / JCI 2011 [86] Bone Reports 2016 [28] NEJM 2018 [29] JBMR 2018 [21]
ClinicalTials.gov number NCT00830674 NCT01340482, NCT01571596 NCT02163577 NCT02526160

Population Adults with XLH Adults with XLH Children with XLH, Adults with XLH
ages 5–12 years at and Brief Pain Inventory
enrollment worst pain score ≥ 4

Treatment arms Single dose Multidose for 16 months: Multidose for 64 weeks, Multidose for 24 weeks:
with dose titration period c
Intravenous 4-month dose escalation phase Every 2 weeks burosumab Every 4 weesk burosumab
–1
burosumab Every 4 weeks burosumab (mean dose at week 40 was 1 mg kg (n = 68)
–1 –1
0.003–0.3 mg kg (n = 17) 0.05–0.6 mg kg
–1
(n = 28), 0.98 mg kg ) (n = 26)
vs. placebo (n = 5)
or followed by or or

Subcutaneous 12-month extension phase Every Every 4 weeks burosumab Every 4 weeks placebo
burosumab 4 weeks burosumab (mean dose at week 40 was (n = 66)
–1 –1
0.1–1 mg kg (n = 12) 0.1–1 mg kg
–1
(n = 22) b 1.5 mg kg ; n = 26)
vs. placebo (n = 4)
Primary outcome Safety and tolerability Proportion of subjects achieving Change in RSS from Percent of subjects achieving
maximum fasting serum Pi in baseline to week 40 and 64 mean serum Pi in the normal
normal range range across the midpoint of
dosing intervals

Biochemical findings Increased serum Pi, serum Increased serum Pi, serum Increased serum Pi, serum Increased serum Pi, serum
1,25(OH)2D3, and TmP/ 1,25(OH)2D3, and TmP/GFR 1,25(OH)2D3, and TmP/ 1,25(OH)2D3, and TmP/GFR
GFR GFR;
decreased serum alkaline
phosphatase.

Radiographic outcomes Improved rickets by RSS Improved healing of active


and RGI-C. fractures/pseudofractures
Greater changes seen with from baseline:
every 2-week dosing, and in burosumab (43.1% of
those with higher baseline fractures healed) compared
RSS to placebo (7.7%)

Other outcomes At baseline: Increased standing height WOMAC:


Impairment of physical function z-score; Decreased stiffness scores

Over 4 months of treatment: Among those with baseline


Improved role limitations due to impairments in 6MWT, the
physical health (SF-36v2), distance walked improved
physical functioning and stiffness
(WOMAC) On PODCI, among those
with impairments,
burosumab improved:
Sports and physical
functioning domain,
Pain and comfort domain,
Global functioning
a
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
b –1
During the extension phase over 80% of subjects received doses of 0.6–1 mg kg [87]
c
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

Br J Clin Pharmacol (2019) 85 1188–1198 1193


E. A. Imel and K. E. White

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 im-
provements 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 impair-
ments 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 im-
prove 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 ac-
tive comparator treatment), after which all subjects received
burosumab for the subsequent 24 weeks [21]. Subjects were re-
quired 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 multi-
ple pseudofractures. The primary endpoint (the percent of pa-
tients achieving mean serum phosphorus concentrations in
the normal range across the midpoint of dosing intervals) Figure 1
was achieved in 94% of subjects after burosumab vs. 7.6% af- Data from a 24-week randomized, placebo-controlled trial of
ter placebo (Figure 1A). Trough serum phosphorus remained –1
burosumab 1 mg kg vs. placebo in adults with X-linked
in the normal range in 67.6% of burosumab treated subjects. hypophosphataemia are shown. (A) Mean serum phosphorus is
The burosumab group had improvements at week 24 in within the normal range at the midpoint of dosing interval during
the brief pain inventory worst pain score, and WOMAC phys- treatment with burosumab, compared with placebo treated sub-
ical function and stiffness subscales compared to placebo, but jects, whose phosphorus remained low. (B) Active fractures or
only stiffness remained significant after adjustment for multi- pseudofractures were more likely to heal during 24 weeks of
burosumab compared with placebo. Adapted and reproduced from
ple comparisons. Most importantly 43.1% of baseline
Figures 1A and 3A of Insogna et al. J Bone Miner Res 2018;33:1383–
fractures/pseudofractures were healed after 24 weeks in the
1393; (Reference [21]); Wiley Publishers, https://doi.org/10.1002/
burosumab group vs. 7.7% in the placebo group (Figure 1B). jbmr.3475. Creative Commons Attribution License (CC BY 4.0,
Regarding safety, the most common drug-related adverse https://creativecommons.org/licenses/by/4.0)
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 treated subjects in the placebo controlled adult study (8/68
occurred in 57.7% of children [29] and 11.8% of adults [21], or 11.8%) but was also present to some degree in the placebo
although, interestingly, injection site reactions also occurred arm (5/66 or 7.6%) [21]. No serious adverse events were attrib-
in 12.1% of placebo treated adults. Additional adverse events uted to burosumab itself.
occurring in ≥25% of children included headache, vomiting, There has been no systematic evidence of change in PTH,
pyrexia, extremity pain and decrease in vitamin D. Addi- serum calcium or urine calcium excretion in any trials.
tional adverse events occurring in ≥10% of adults included Nephrocalcinosis was present at baseline in 35% of children
back pain, tooth abscess, nasopharyngitis, headaches, nausea in the Phase 2 trial [29] and 54% of adults in the Phase 3 trial
and dizziness. However, most listed adverse events were not [21]. So far, there has been no clear indication of either wors-
considered drug related, while some are consistent with ening or improvement of nephrocalcinosis overall with
symptoms common to XLH patients. Restless legs syndrome burosumab treatment. In fact, similar numbers of patients
was noted in the initial multidose trial (5/28 or 17.9%) [87]. had increases in nephrocalcinosis score in the burosumab
Restless legs syndrome occurred more often in burosumab- and placebo groups, while similar numbers also had decreases

1194 Br J Clin Pharmacol (2019) 85 1188–1198


Pharmacological management of X-linked hypophosphataemia

in nephrocalcinosis scores in each group [21]. No significant Conclusion


changes in echocardiograms were reported.
An advantage of burosumab is that it achieves normalization XLH is a complex musculoskeletal condition, requiring
of serum phosphorus through improvements in TmP/GFR, in careful medical and surgical management. Recent approval
contrast to standard therapy with active vitamin D and of burosumab represents a substantial advancement in
phosphate salts. One hopes that this will translate into lower management of XLH, directed at its pathophysiology of
long-term risk of nephrocalcinosis and hyperparathyroidism, FGF23 excess. Further studies are needed to fully characterize
provided that episodes of hyperphosphataemia are avoided. In long-term risks and benefits of this drug, especially regarding
the phase 3 placebo controlled adult trial, five patients (7.4%) disease features that take years to develop.
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 Nomenclature of targets and ligands
trial on smaller doses. Thus, some patients may respond well to Key protein targets and ligands in this article are hyperlinked
smaller doses to manage XLH. to corresponding entries in http://www.guidetophar-
Conceptually, anti-drug antibodies could bind and inter- macology.org, the common portal for data from the
fere with the action of burosumab. The initial multidose IUPHAR/BPS Guide to PHARMACOLOGY [94], and are
trial in adults, and the Phase 2 trial in children did not find permanently archived in the Concise Guide to PHARMA-
anti-drug antibody development [29, 87]. However, one COLOGY 2017/18 [95, 96].
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 Competing Interests
development of antibodies during trial participation, was
E.A.I. receives research funding that contributes to this work
not seen. To date, there is no evidence for neutralizing
from the NIH/NIAMS 1P30AR072581. K.E.W. would like to
anti-drug antibodies attenuating effect of burosumab in
acknowledge support by NIH grants DK063934, DK095784,
human subjects.
and AR059278. E.A.I. has received research funding and fees
Patients with moderate to severe CKD were excluded from
for consulting with Kyowa Hakko Kirin and Ultragenyx,
the burosumab trials. In a rat model of severe CKD, treatment
Pharmaceuticals. K.E.W. receives royalties from Kyowa
with a different FGF23 antibody hastened death, probably
Hakko Kirin Co. Ltd for licensing FGF23.
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
References
European Medicines Agency (EMA; conditionally) and in 1 Beck-Nielsen SS, Brock-Jacobsen B, Gram J, Brixen K, Jensen TK.
2018 by the FDA (fully). The EMA conditionally approved Incidence and prevalence of nutritional and hereditary rickets in
use of burosumab in children aged 1 year and older with ra- southern Denmark. Eur J Endocrinol 2009; 160: 491–7.
diographic evidence of bone disease, through adolescence 2 Carpenter TO, Imel EA, Holm IA, Jan de Beur SM, Insogna KL. A
during skeletal growth. The EMA approved starting dose is clinician’s guide to X-linked hypophosphatemia. J Bone Miner
0.4 mg kg–1 subcutaneously every 2 weeks in children with Res 2011; 26: 1381–8.
0.8 mg kg–1 as typical maintenance dosing, and a maximum 3 Hyp Consortium. A gene (PEX) with homologies to
dose of 90 mg [93]. The FDA approved starting dose is endopeptidases is mutated in patients with X-linked
0.8 mg kg–1 every 2 weeks in children aged 1 year and older, hypophosphatemic rickets. The HYP consortium. Nat Genet
and 1 mg kg–1 every 4 weeks in adults with a maximum dose 1995; 11: 130–6.
of 90 mg [91]. Doses are titrated on the basis of peak and
4 Imel EA, Econs MJ. Approach to the hypophosphatemic patient.
trough serum phosphorus concentrations. J Clin Endocrinol Metab 2012; 97: 696–706.
Several questions remain. A true active comparator study in
growing children aged 1–12 years is underway (ClinicalTrials. 5 McNair SL, Stickler GB. Growth in familial hypophosphatemic
vitamin-D-resistant rickets. N Engl J Med 1969; 281: 512–6.
gov NCT02915705). This trial will identify the effect of 64 weeks
of burosumab vs. phosphate and active vitamin D on multiple 6 Makitie O, Doria A, Kooh SW, Cole WG, Daneman A, Sochett E.
clinical parameters with a primary outcome of differences in Early treatment improves growth and biochemical and
healing of radiographic rickets. The effect on adult height in radiographic outcome in X-linked hypophosphatemic rickets.
growing children is unknown. However, this will take years to J Clin Endocrinol Metab 2003; 88: 3591–7.
evaluate, given the mean ages of children enrolled in the clinical 7 Harrison HE, Harrison HC, Lifshitz F, Johnson AD. Growth
trials. Since enthesopathy and other joint related complications disturbance in hereditary hypophosphatemia. Am J Dis Child
develop and progress slowly, many years of treatment with 1966; 112: 290–7.
burosumab will be necessary to interpret whether or not these 8 Zivicnjak M, Schnabel D, Billing H, Staude H, Filler G, Querfeld U,
outcomes are altered. Similarly, years of treatment are likely to et al. Age-related stature and linear body segments in children
be necessary to determine whether the chronic risk of with X-linked hypophosphatemic rickets. Pediatr Nephrol 2011;
nephrocalcinosis changes. 26: 223–31.

Br J Clin Pharmacol (2019) 85 1188–1198 1195


E. A. Imel and K. E. White

9 Kruse K, Hinkel GK, Griefahn B. Calcium metabolism and growth 25 Mills E, Iorio L, Macica CM. Assessing outcomes of joint
during early treatment of children with X-linked replacement in patients with X-linked hypophosphatemia. In:
hypophosphataemic rickets. Eur J Pediatr 1998; 157: 894–900. ASBMR 2017 Annual Meeting, Denver, Colorado: ASBMR, 2017:
SA0339.
10 Linglart A, Biosse-Duplan M, Briot K, Chaussain C, Esterle L,
Guillaume-Czitrom S, et al. Therapeutic management of 26 Aono Y, Hasegawa H, Yamazaki Y, Shimada T, Fujita T, Yamashita
hypophosphatemic rickets from infancy to adulthood. Endocr T, et al. Anti-FGF-23 neutralizing antibodies ameliorate muscle
Connect 2014; 3: R13–30. weakness and decreased spontaneous movement of Hyp mice.
J Bone Miner Res 2011; 26: 803–10.
11 Beck-Nielsen SS, Brusgaard K, Rasmussen LM, Brixen K, Brock-
Jacobsen B, Poulsen MR, et al. Phenotype presentation of 27 Veilleux LN, Cheung M, Ben Amor M, Rauch F. Abnormalities in
hypophosphatemic rickets in adults. Calcif Tissue Int 2010; 87: muscle density and muscle function in hypophosphatemic
108–19. rickets. J Clin Endocrinol Metab 2012; 97: E1492–8.
12 Caldemeyer KS, Boaz JC, Wappner RS, Moran CC, Smith RR, 28 Ruppe MD, Zhang X, Imel EA, Weber TJ, Klausner MA, Ito T, et al.
Quets JP. Chiari I malformation: association with Effect of four monthly doses of a human monoclonal anti-FGF23
hypophosphatemic rickets and MR imaging appearance. antibody (KRN23) on quality of life in X-linked
Radiology 1995; 195: 733–8. hypophosphatemia. Bone Rep 2016; 5: 158–62.
13 Gjorup H, Kjaer I, Sonnesen L, Haubek D, Beck-Nielsen SS, Hintze 29 Carpenter TO, Whyte MP, Imel EA, Boot AM, Hogler W,
H, et al. Craniofacial morphology in patients with Linglart A, et al. Burosumab therapy in children with X-linked
hypophosphatemic rickets: a cephalometric study focusing on hypophosphatemia. N Engl J Med 2018; 378: 1987–98.
differences between bone of cartilaginous and intramembranous
30 Che H, Roux C, Etcheto A, Rothenbuhler A, Kamenicky P, Linglart
origin. Am J Med Genet A 2011; 155A: 2654–60.
A, et al. Impaired quality of life in adults with X-linked
14 Vega RA, Opalak C, Harshbarger RJ, Fearon JA, Ritter AM, Collins JJ, hypophosphatemia and skeletal symptoms. Eur J Endocrinol
et al. Hypophosphatemic rickets and craniosynostosis: a multicenter 2016; 174: 325–33.
case series. J Neurosurg Pediatr 2016; 17: 694–700.
31 Beck L, Soumounou Y, Martel J, Krishnamurthy G, Gauthier C,
15 Onishi T, Umemura S, Shintani S, Ooshima T. Phex mutation causes Goodyer CG, et al. PEX/PEX tissue distribution and evidence for a
overexpression of FGF23 in teeth. Arch Oral Biol 2008; 53: 99–104. deletion in the 30 region of the Pex gene in X-linked
16 Boukpessi T, Hoac B, Coyac BR, Leger T, Garcia C, Wicart P, et al. hypophosphatemic mice. J Clin Invest 1997; 99: 1200–9.
Osteopontin and the dento-osseous pathobiology of X-linked 32 Meyer MH, Meyer RA Jr. MRNA expression of Phex in mice
hypophosphatemia. Bone 2017; 95: 151–61. and rats: the effect of low phosphate diet. Endocrine 2000; 13:
17 Chaussain-Miller C, Sinding C, Wolikow M, Lasfargues JJ, Godeau 81–7.
G, Garabedian M. Dental abnormalities in patients with familial 33 Liu S, Zhou J, Tang W, Jiang X, Rowe DW, Quarles LD. Pathogenic
hypophosphatemic vitamin D-resistant rickets: prevention by role of Fgf23 in Hyp mice. Am J Physiol Endocrinol Metab 2006;
early treatment with 1-hydroxyvitamin D. J Pediatr 2003; 142: 291: E38–49.
324–31.
34 Miao D, Bai X, Panda D, McKee M, Karaplis A, Goltzman D.
18 Fong H, Chu EY, Tompkins KA, Foster BL, Sitara D, Lanske B, et al. Osteomalacia in hyp mice is associated with abnormal PHEX
Aberrant cementum phenotype associated with expression and with altered bone matrix protein expression and
the hypophosphatemic hyp mouse. J Periodontol 2009; 80: 1348–54. deposition. Endocrinology 2001; 142: 926–39.
19 Connor J, Olear EA, Insogna KL, Katz L, Baker S, Kaur R, et al. 35 Liu S, Guo R, Simpson LG, Xiao Z-S, Burnham CE, Quarles LD.
Conventional therapy in adults with X-linked Regulation of fibroblastic growth factor 23 expression but not
hypophosphatemia: effects on enthesopathy and dental disease. degradation by PHEX. J Biol Chem 2003; 278: 37419–26.
J Clin Endocrinol Metab 2015; 100: 3625–32.
36 Larsson T, Marsell R, Schipani E, Ohlsson C, Ljunggren O,
20 Biosse Duplan M, Coyac BR, Bardet C, Zadikian C, Rothenbuhler A, Tenenhouse HS, et al. Transgenic mice expressing fibroblast
Kamenicky P, et al. Phosphate and vitamin D prevent periodontitis growth factor 23 under the control of the alpha1(I) collagen
in X-linked hypophosphatemia. J Dent Res 2017; 96: 388–95. promoter exhibit growth retardation, osteomalacia, and
21 Insogna KL, Briot K, Imel EA, Kamenicky P, Ruppe MD, Portale disturbed phosphate homeostasis. Endocrinology 2004; 145:
AA, et al. A randomized, double-blind, placebo-controlled, phase 3087–94.
3 trial evaluating the efficacy of burosumab, an anti-FGF23 37 Tenenhouse HS, Beck L. Renal na(+)-phosphate cotransporter
antibody, in adults with X-linked hypophosphatemia: week 24 gene expression in X-linked Hyp and Gy mice. Kidney Int 1996;
primary analysis. J Bone Miner Res 2018; 33: 1383–93. 49: 1027–32.
22 Reid IR, Hardy DC, Murphy WA, Teitelbaum SL, Bergfeld MA, 38 Burnett SM, Gunawardene SC, Bringhurst FR, Juppner H, Lee H,
Whyte MP. X-linked hypophosphatemia: a clinical, biochemical, Finkelstein JS. Regulation of C-terminal and intact FGF-23 by
and histopathologic assessment of morbidity in adults. Medicine dietary phosphate in men and women. J Bone Miner Res 2006; 21:
(Baltimore) 1989; 68: 336–52. 1187–96.
23 Karaplis AC, Bai X, Falet JP, Macica CM. Mineralizing 39 Shimada T, Mizutani S, Muto T, Yoneya T, Hino R, Takeda S, et al.
enthesopathy is a common feature of renal phosphate-wasting Cloning and characterization of FGF23 as a causative factor of
disorders attributed to FGF23 and is exacerbated by standard tumor-induced osteomalacia. Proc Natl Acad Sci U S A 2001; 98:
therapy in hyp mice. Endocrinology 2012; 153: 5906–17. 6500–5.
24 Liang G, Katz LD, Insogna KL, Carpenter TO, Macica CM. Survey 40 Shimada T, Hasegawa H, Yamazaki Y, Muto T, Hino R, Takeuchi Y,
of the enthesopathy of X-linked hypophosphatemia and its et al. FGF-23 is a potent regulator of vitamin D metabolism and
characterization in Hyp mice. Calcif Tissue Int 2009; 85: 235–46. phosphate homeostasis. J Bone Miner Res 2004; 19: 429–35.

1196 Br J Clin Pharmacol (2019) 85 1188–1198


Pharmacological management of X-linked hypophosphataemia

41 Bai XY, Miao D, Goltzman D, Karaplis AC. The autosomal 56 Taylor A, Sherman NH, Norman ME. Nephrocalcinosis in X-
dominant hypophosphatemic rickets R176Q mutation in linked hypophosphatemia: effect of treatment versus disease.
fibroblast growth factor 23 resists proteolytic cleavage and Pediatr Nephrol 1995; 9: 173–5.
enhances in vivo biological potency. J Biol Chem 2003; 278:
57 Verge CF, Lam A, Simpson JM, Cowell CT, Howard NJ, Silink M.
9843–9.
Effects of therapy in X-linked hypophosphatemic rickets. N Engl J
42 Econs MJ, McEnery PT. Autosomal dominant hypophosphatemic Med 1991; 325: 1843–8.
rickets/osteomalacia: clinical characterization of a novel renal
58 Nakamura Y, Takagi M, Takeda R, Miyai K, Hasegawa Y.
phosphate-wasting disorder. J Clin Endocrinol Metab 1997; 82:
Hypertension is a characteristic complication of X-linked
674–81.
hypophosphatemia. Endocr J 2017; 64: 283–9.
43 Imel EA, Peacock M, Gray AK, Padgett LR, Hui SL, Econs MJ. Iron
59 Carpenter TO, Mitnick MA, Ellison A, Smith C, Insogna KL.
modifies plasma FGF23 differently in autosomal dominant
Nocturnal hyperparathyroidism: a frequent feature of X-linked
hypophosphatemic rickets and healthy humans. J Clin
hypophosphatemia. J Clin Endocrinol Metab 1994; 78: 1378–83.
Endocrinol Metab 2011; 96: 3541–9.
60 Makitie O, Kooh SW, Sochett E. Prolonged high-dose phosphate
44 Farrow EG, Yu X, Summers LJ, Davis SI, Fleet JC, Allen MR, et al.
treatment: a risk factor for tertiary hyperparathyroidism in
Iron deficiency drives an autosomal dominant X-linked hypophosphatemic rickets. Clin Endocrinol (Oxf) 2003;
hypophosphatemic rickets (ADHR) phenotype in fibroblast 58: 163–8.
growth factor-23 (Fgf23) knock-in mice. Proc Natl Acad Sci U S A
2011; 108: E1146–55. 61 Rivkees SA, el-Hajj-Fuleihan G, Brown EM, Crawford JD. Tertiary
hyperparathyroidism during high phosphate therapy of familial
45 Clinkenbeard EL, Farrow EG, Summers LJ, Cass TA, Roberts JL, hypophosphatemic rickets. J Clin Endocrinol Metab 1992; 75:
Bayt CA, et al. Neonatal iron deficiency causes abnormal 1514–8.
phosphate metabolism by elevating FGF23 in normal and ADHR
mice. J Bone Miner Res 2014; 29: 361–9. 62 Rhee Y, Bivi N, Farrow E, Lezcano V, Plotkin LI, White KE, et al.
Parathyroid hormone receptor signaling in osteocytes increases
46 Imel EA, Gray AK, Padgett LR, Econs MJ. Iron and fibroblast the expression of fibroblast growth factor-23 in vitro and in vivo.
growth factor 23 in X-linked hypophosphatemia. Bone 2014; 60: Bone 2011; 49: 636–43.
87–92.
63 Yavropoulou MP, Kotsa K, Gotzamani Psarrakou A, Papazisi A,
47 Harrell RM, Lyles KW, Harrelson JM, Friedman NE, Drezner MK. Tranga T, Ventis S, et al. Cinacalcet in hyperparathyroidism
Healing of bone disease in X-linked hypophosphatemic secondary to X-linked hypophosphatemic rickets: case report and
rickets/osteomalacia. Induction and maintenance with brief literature review. Hormones (Athens) 2010; 9: 274–8.
phosphorus and calcitriol. J Clin Invest 1985; 75: 1858–68.
64 Raeder H, Shaw N, Netelenbos C, Bjerknes R. A case of X-linked
48 Glorieux FH, Marie PJ, Pettifor JM, Delvin EE. Bone response to hypophosphatemic rickets: complications and the therapeutic
phosphate salts, ergocalciferol, and calcitriol in use of cinacalcet. Eur J Endocrinol 2008; 159 (Suppl. 1): S101–5.
hypophosphatemic vitamin D-resistant rickets. N Engl J Med
1980; 303: 1023–31. 65 Alon US, Levy-Olomucki R, Moore WV, Stubbs J, Liu S, Quarles
LD. Calcimimetics as an adjuvant treatment for familial
49 Petersen DJ, Boniface AM, Schranck FW, Rupich RC, Whyte MP. hypophosphatemic rickets. Clin J Am Soc Nephrol 2008; 3:
X-linked hypophosphatemic rickets: a study (with literature 658–64.
review) of linear growth response to calcitriol and phosphate
therapy. J Bone Miner Res 1992; 7: 583–97. 66 Alon US, Jarka D, Monachino PJ, Sebestyen VanSickle J, Srivastava
T. Cinacalcet as an alternative to phosphate therapy in X-linked
50 Imel EA, DiMeglio LA, Hui SL, Carpenter TO, Econs MJ. hypophosphataemic rickets. Clin Endocrinol (Oxf) 2017; 87:
Treatment of X-linked hypophosphatemia with calcitriol and 114–6.
phosphate increases circulating fibroblast growth factor 23
67 Nesbitt T, Lobaugh B, Drezner MK. Calcitonin stimulation of
concentrations. J Clin Endocrinol Metab 2010; 95: 1846–50.
renal 25-hydroxyvitamin D-1 alpha-hydroxylase activity in
51 Liu S, Tang W, Zhou J, Stubbs JR, Luo Q, Pi M, et al. Fibroblast hypophosphatemic mice. Evidence that the regulation of
growth factor 23 is a counter-regulatory phosphaturic hormone calcitriol production is not universally abnormal in X-linked
for vitamin D. J Am Soc Nephrol 2006; 17: 1305–15. hypophosphatemia. J Clin Invest 1987; 79: 15–9.
52 Carpenter TO, Insogna KL, Zhang JH, Ellis B, Nieman S, Simpson 68 Econs MJ, Lobaugh B, Drezner MK. Normal calcitonin
C, et al. Circulating levels of soluble klotho and FGF23 in X-linked stimulation of serum calcitriol in patients with X-linked
hypophosphatemia: circadian variance, effects of treatment, and hypophosphatemic rickets. J Clin Endocrinol Metab 1992; 75:
relationship to parathyroid status. J Clin Endocrinol Metab 2010; 408–11.
95: E352–7.
69 Plotkin LI, Weinstein RS, Parfitt AM, Roberson PK, Manolagas SC,
53 Gizard A, Rothenbuhler A, Pejin Z, Finidori G, Glorion C, de Billy Bellido T. Prevention of osteocyte and osteoblast apoptosis by
B, et al. Outcomes of orthopedic surgery in a cohort of 49 patients bisphosphonates and calcitonin. J Clin Invest 1999; 104:
with X-linked hypophosphatemic rickets (XLHR). Endocr 1363–74.
Connect 2017; 6: 566–73.
70 van Boekel G, Ruinemans-Koerts J, Joosten F, Dijkhuizen P, van
54 Goodyer PR, Kronick JB, Jequier S, Reade TM, Scriver CR. Sorge A, de Boer H. Tumor producing fibroblast growth factor 23
Nephrocalcinosis and its relationship to treatment of hereditary localized by two-staged venous sampling. Eur J Endocrinol 2008;
rickets. J Pediatr 1987; 111: 700–4. 158: 431–7.
55 Reusz GS, Hoyer PF, Lucas M, Krohn HP, Ehrich JH, Brodehl J. X 71 Liu ES, Carpenter TO, Gundberg CM, Simpson CA, Insogna KL.
linked hypophosphataemia: treatment, height gain, and Calcitonin administration in X-linked hypophosphatemia. N
nephrocalcinosis. Arch Dis Child 1990; 65: 1125–8. Engl J Med 2011; 364: 1678–80.

Br J Clin Pharmacol (2019) 85 1188–1198 1197


E. A. Imel and K. E. White

72 Folsom LJ, Imel EA. Hyperphosphatemic familial tumoral 85 Aono Y, Yamazaki Y, Yasutake J, Kawata T, Hasegawa H, Urakawa
calcinosis: genetic models of deficient FGF23 action. Curr I, et al. Therapeutic effects of anti-FGF23 antibodies in
Osteoporos Rep 2015; 13: 78–87. hypophosphatemic rickets/osteomalacia. J Bone Miner Res 2009;
24: 1879–88.
73 Sullivan R, Abraham A, Simpson C, Olear E, Carpenter T, Deng Y,
et al. Three-month randomized clinical trial of nasal calcitonin in 86 Carpenter TO, Imel EA, Ruppe MD, Weber TJ, Klausner MA,
adults with X-linked hypophosphatemia. Calcif Tissue Int 2018; Wooddell MM, et al. Randomized trial of the anti-FGF23 antibody
102: 666–70. KRN23 in X-linked hypophosphatemia. J Clin Invest 2014; 124:
1587–97.
74 White KE, Jonsson KB, Carn G, Hampson G, Spector TD,
Mannstadt M, et al. The autosomal dominant hypophosphatemic 87 Imel EA, Zhang X, Ruppe MD, Weber TJ, Klausner MA, Ito T, et al.
rickets (ADHR) gene is a secreted polypeptide overexpressed by Prolonged correction of serum phosphorus in adults with
tumors that cause phosphate wasting. J Clin Endocrinol Metab X-linked hypophosphatemia using monthly doses of KRN23.
2001; 86: 497–500. J Clin Endocrinol Metab 2015; 100: 2565–73.
75 Folpe AL, Fanburg-Smith JC, Billings SD, Bisceglia M, Bertoni F, 88 Zhang X, Peyret T, Gosselin NH, Marier JF, Imel EA, Carpenter
Cho JY, et al. Most osteomalacia-associated mesenchymal tumors TO. Population pharmacokinetic and pharmacodynamic
are a single histopathologic entity - an analysis of 32 cases and a analyses from a 4-month intradose escalation and its
comprehensive review of the literature. American Journal of subsequent 12-month dose titration studies for a human
Surgical Pathology 2004; 28: 1–30. monoclonal anti-FGF23 antibody (KRN23) in adults with
76 Seufert J, Ebert K, Muller J, Eulert J, Hendrich C, Werner E, et al. X-linked hypophosphatemia.
Octreotide therapy for tumor-induced osteomalacia. N Engl J Med J Clin Pharmacol 2016; 56: 429–38.
2001; 345: 1883–8. 89 Whyte MP, Fujita KP, Moseley S, Thompson DD, McAlister WH.
77 Nasu T, Kurisu S, Matsuno S, Tatsumi K, Kakimoto T, Kobayashi Validation of a novel scoring system for changes in skeletal
M, et al. Tumor-induced hypophosphatemic osteomalacia manifestations of Hypophosphatasia in newborns, infants, and
diagnosed by the combinatory procedures of magnetic resonance children: the radiographic global impression of change scale.
imaging and venous sampling for FGF23. Intern Med 2008; 47: J Bone Miner Res 2018; 33: 868–74.
957–61.
90 Whyte M, Imel EA, Portale AA, Boot AM, Hogler W, Linglart A,
78 Hesse E, Moessinger E, Rosenthal H, Laenger F, Brabant G, Petrich et al. Burosumab, a fully human anti-FGF23 monoclonal
T, et al. Oncogenic osteomalacia: exact tumor localization by antibody, for X-linked hypophosphatemia (XLH): results from
co-registration of positron emission and computed tomography. two phase 2 trials in affected children 1–12 years old. In: ENDO
J Bone Miner Res 2007; 22: 158–62. 2018, Chicago, Il, 2018: Abstract MON-473.

79 Hodgson SF, Clarke BL, Tebben PJ, Mullan BP, Cooney WP 3rd, 91 FDA. CRYSVITA® (burosumab-twza) injection, for subcutaneous
Shives TC. Oncogenic osteomalacia: localization of underlying use; Initial U.S. Approval: 2018. In, edFDA. Available at https://
peripheral mesenchymal tumors with use of Tc 99m sestamibi www.accessdata.fda.gov/drugsatfda_docs/nda/2018/
scintigraphy. Endocr Pract 2006; 12: 35–42. 761068Orig1s000TOC.cfm (last accessed 18 October 2018).

80 Khadgawat R, Singh Y, Kansara S, Tandon N, Bal C, Seith A, 92 Shalhoub V, Shatzen EM, Ward SC, Davis J, Stevens J, Bi V, et al.
et al. PET/CT localisation of a scapular haemangiopericytoma FGF23 neutralization improves chronic kidney disease-associated
with tumour-induced osteomalacia. Singapore Med J 2009; 50: hyperparathyroidism yet increases mortality. J Clin Invest 2012;
e55–7. 122: 2543–53.

81 Andreopoulou P, Dumitrescu CE, Kelly MH, Brillante BA, Cutler 93 EMA. Crysvita: EPAR - Product Information In. Available at
Peck CM, Wodajo FM, et al. Selective venous catheterization for http://www.ema.europa.eu/ema/index.jsp?curl=pages/
the localization of phosphaturic mesenchymal tumors. J Bone medicines/human/medicines/004275/human_med_002224.
Miner Res 2011; 26: 1295–302. jsp&mid=WC0b01ac058001d124 (last accessed 18 October
2018).
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 94 Harding SD, Sharman JL, Faccenda E, Southan C, Pawson AJ,
event in phosphaturic mesenchymal tumour. J Pathol 2015; 235: Ireland S, et al. The IUPHAR/BPS Guide to PHARMACOLOGY
539–45. in 2018: updates and expansion to encompass the new
guide to IMMUNOPHARMACOLOGY. Nucl Acids Res 2018; 46:
83 White KE, Cabral JM, Davis SI, Fishburn T, Evans WE, Ichikawa S,
D10–106.
et al. Mutations that cause osteoglophonic dysplasia define novel
roles for FGFR1 in bone elongation. Am J Hum Genet 2005; 76: 95 Alexander SPH, Fabbro D, Kelly E, Marrion NV, Peters JA,
361–7. Faccenda E, et al. The Concise Guide to PHARMACOLOGY 2017/
18: Enzymes. Br J Pharmacol 2017; 174: S272–359.
84 Yamazaki Y, Tamada T, Kasai N, Urakawa I, Aono Y, Hasegawa H,
et al. Anti-FGF23 neutralizing antibodies show the physiological 96 Alexander SPH, Kelly E, Marrion NV, Peters JA, Faccenda E,
role and structural features of FGF23. J Bone Miner Res 2008; 23: Harding SD, et al. The Concise Guide to PHARMACOLOGY 2017/
1509–18. 18: Transporters. Br J Pharmacol 2017; 174: S360–446.

1198 Br J Clin Pharmacol (2019) 85 1188–1198

You might also like