216 results on '"Familial Hypophosphatemic Rickets drug therapy"'
Search Results
102. Clinical and genetic analysis of two Chinese families with vitamin D-dependent rickets type IA and follow-up.
- Author
-
Li Y, Yuan X, Chen R, Lin X, Shangguan H, Yang X, and Zhang Y
- Subjects
- China, Follow-Up Studies, Humans, Vitamin D, 25-Hydroxyvitamin D3 1-alpha-Hydroxylase genetics, Familial Hypophosphatemic Rickets drug therapy, Familial Hypophosphatemic Rickets genetics
- Abstract
Objective: Vitamin D-dependent rickets type IA (VDDR-IA) is a rare autosomal recessive disorder characterized by the early onset of severe rickets. The objectives of this study were twofold: (1) to analyze the clinical characteristics and therapy of two patients with VDDR-IA from two separate Chinese families, and (2) investigate the CYP27B1 gene mutations in two large pedigrees., Methods: Medical history, clinical manifestations, physical examination, radiological findings and laboratory data were analyzed from two patients with VDDR-IA. Serum 1, 25-dihydroxyvitamin D [1, 25-(OH)
2 D3 ] of the two patients and their respective families were measured by ELISA and blood samples from both families was obtained for CYP27B1 gene sequence., Results: Two patients had typical manifestations and radiological evidence of rickets. Laboratory data showed hypocalcaemia and hypophosphataemia, along with high levels of serum alkaline phosphatase, parathyroid hormone and 25-hydroxyvitamin D3. However, serum 1,25-(OH)2 D3 level were low in the patients but normal in their family members. Genetic sequence identified two patients were homozygous for a duplication mutation in exon 8 of CYP27B1 gene (c.1319_1325dupCCCACCC, p.Phe443Profs * 24). After treating with calcitriol and calcium, there was biochemical improvement with normalization of serum calcium and phosphorus, and radiographic evidence of compensatory skeletal mineralization. One patient developed nephrocalcinosis during follow-up., Conclusions: This study identified a recurrent seven-nucleotide insertion of CYP27B1 in two large pedigrees, and compared the clinical characteristics and individual therapy of two affected patients. Additionally, our experience further supports the notion that nephrocalcinosis can occur even on standard doses of calcitriol and oral calcium, and normal level of serum calcium, phosphorus, PTH and 25-(OH)D3 .- Published
- 2020
- Full Text
- View/download PDF
103. Mineralized tissues in hypophosphatemic rickets.
- Author
-
Robinson ME, AlQuorain H, Murshed M, and Rauch F
- Subjects
- Absorptiometry, Photon, Bone Development drug effects, Bone Development genetics, Bone and Bones diagnostic imaging, Bone and Bones pathology, Calcification, Physiologic drug effects, Calcification, Physiologic genetics, Calcitriol administration & dosage, Familial Hypophosphatemic Rickets diagnosis, Familial Hypophosphatemic Rickets drug therapy, Familial Hypophosphatemic Rickets genetics, Fibroblast Growth Factor-23, Fibroblast Growth Factors antagonists & inhibitors, Humans, Osteocytes metabolism, Osteomalacia diagnosis, Osteomalacia drug therapy, Osteomalacia genetics, PHEX Phosphate Regulating Neutral Endopeptidase metabolism, Paracrine Communication genetics, Phosphates administration & dosage, Phosphates blood, Renal Reabsorption drug effects, Renal Reabsorption genetics, Tooth growth & development, Tooth pathology, Treatment Outcome, Familial Hypophosphatemic Rickets pathology, Fibroblast Growth Factors metabolism, Osteomalacia pathology, PHEX Phosphate Regulating Neutral Endopeptidase genetics, Phosphates metabolism
- Abstract
Hypophosphatemic rickets is caused by renal phosphate wasting that is most commonly due to X-linked dominant mutations in PHEX. PHEX mutations cause hypophosphatemia indirectly, through the increased expression of fibroblast growth factor 23 (FGF23) by osteocytes. FGF23 decreases renal phosphate reabsorption and thereby increases phosphate excretion. The lack of phosphate leads to a mineralization defect at the level of growth plates (rickets), bone tissue (osteomalacia), and teeth, where the defect facilitates the formation of abscesses. The bone tissue immediately adjacent to osteocytes often remains unmineralized ("periosteocytic lesions"), highlighting the osteocyte defect in this disorder. Common clinical features of XLH include deformities of the lower extremities, short stature, enthesopathies, dental abscesses, as well as skull abnormalities such as craniosynostosis and Chiari I malformation. For the past four decades, XLH has been treated by oral phosphate supplementation and calcitriol, which improves rickets and osteomalacia and the dental manifestations, but often does not resolve all aspects of the mineralization defects. A newer treatment approach using inactivating FGF23 antibodies leads to more stable control of serum inorganic phosphorus levels and seems to heal rickets more reliably. However, the long-term benefits of FGF23 antibody treatment remain to be elucidated.
- Published
- 2020
- Full Text
- View/download PDF
104. Growth Curves for Children with X-linked Hypophosphatemia.
- Author
-
Mao M, Carpenter TO, Whyte MP, Skrinar A, Chen CY, San Martin J, and Rogol AD
- Subjects
- Adolescent, Child, Child, Preschool, Clinical Trials, Phase II as Topic, Clinical Trials, Phase III as Topic, Familial Hypophosphatemic Rickets drug therapy, Female, Fibroblast Growth Factor-23, Genetic Diseases, X-Linked drug therapy, Humans, Infant, Male, Phosphates administration & dosage, Retrospective Studies, Treatment Outcome, Vitamin D administration & dosage, Vitamin D analogs & derivatives, Body Height physiology, Child Development physiology, Familial Hypophosphatemic Rickets physiopathology, Genetic Diseases, X-Linked physiopathology, Growth Charts
- Abstract
Context: We characterized linear growth in infants and children with X-linked hypophosphatemia (XLH)., Objective: Provide linear growth curves for children with XLH from birth to early adolescence., Design: Data from 4 prior studies of XLH were pooled to construct growth curves. UX023-CL002 was an observational, retrospective chart review. Pretreatment data were collected from 3 interventional trials: two phase 2 trials (UX023-CL201, UX023-CL205) and a phase 3 trial (UX023-CL301)., Setting: Medical centers with expertise in treating XLH., Patients: Children with XLH, 1-14 years of age., Intervention: None., Main Outcome Measure: Height-for-age linear growth curves, including values for the 5th, 10th, 25th, 50th, 75th, 90th, and 95th percentiles for children with XLH compared to population norms., Results: A total of 228 patients (132 girls, 96 boys) with 2381 height measurements were included. Nearly all subjects (> 99%) reported prior management with supplementation therapy. Compared to the Center for Disease Control and Prevention growth curves, boys at age 3 months, 6 months, 9 months, 1 year, and 2 years had median height percentiles of 46%, 37%, 26%, 18%, and 5%, respectively; for girls the median height percentiles were 52%, 37%, 25%, 18%, and 7%, respectively. Annual growth in children with XLH fell below that of healthy children near 1 year of age and progressively declined during early childhood, with all median height percentiles < 8% between 2 and 12 years old., Conclusion: Children with XLH show decreased height gain by 1 year of age and remain below population norms thereafter. These data will help evaluate therapeutic interventions on linear growth for pediatric XLH., (© Endocrine Society 2020.)
- Published
- 2020
- Full Text
- View/download PDF
105. Alterations of bone material properties in adult patients with X-linked hypophosphatemia (XLH).
- Author
-
Fratzl-Zelman N, Gamsjaeger S, Blouin S, Kocijan R, Plasenzotti P, Rokidi S, Nawrot-Wawrzyniak K, Roetzer K, Uyanik G, Haeusler G, Shane E, Cohen A, Klaushofer K, Paschalis EP, Roschger P, Fratzl P, Zwerina J, and Zwettler E
- Subjects
- Adult, Bone Density, Bone Matrix diagnostic imaging, Bone Matrix pathology, Bone and Bones pathology, Calcitriol therapeutic use, Familial Hypophosphatemic Rickets drug therapy, Familial Hypophosphatemic Rickets genetics, Fibroblast Growth Factor-23, Genetic Diseases, X-Linked genetics, Humans, Male, PHEX Phosphate Regulating Neutral Endopeptidase genetics, Phosphates administration & dosage, Phosphates therapeutic use, Retrospective Studies, Spectroscopy, Fourier Transform Infrared, Bone and Bones diagnostic imaging, Familial Hypophosphatemic Rickets diagnostic imaging, Familial Hypophosphatemic Rickets pathology
- Abstract
X-linked hypophosphatemia (XLH) caused by PHEX mutations results in elevated serum FGF23 levels, renal phosphate wasting and low 1,25-dihydroxyvitamin D. The glycophosphoprotein osteopontin, a potent inhibitor of mineralization normally degraded by PHEX, accumulates within the bone matrix. Conventional therapy consisting of supplementation with phosphate and vitamin D analogs is burdensome and the effects on bone material poorly characterized. We analyzed transiliac bone biopsies from four adult patients, two of them severely affected due to no diagnosis and no treatment until adulthood. We used light microscopy, qBEI and FTIRI to study histology, histomorphometry, bone mineralization density distribution, properties of the organic matrix and size of hypomineralized periosteocytic lesions. Non-treatment resulted in severe osteomalacia, twice the amount of mineralized trabecular volume, multiple osteon-like perforations, continuity of lamellae from mineralized to unmineralized areas and distinctive patches of woven bone. Periosteocytic lesions were larger than in treated patients. The latter had nearly normal osteoid thicknesses, although surface was still elevated. The median calcium content of the matrix was always within normal range, although the percentage of lowly mineralized bone areas was highly increased in non-treated patients, resulting in a marked heterogeneity in mineralization. Divalent collagen cross-links were evident independently of the mineral content of the matrix. Broad osteoid seams lacked measurable pyridinoline, a mature trivalent cross-link and exhibited considerable acidic lipid content, typically found in matrix vesicles. Based on our results, we propose a model that possibly integrates the relationship between the observed mineralization disturbances, FGF23 secretion and the known osteopontin accumulation in XLH., Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
106. Burosumab in X-linked hypophosphatemia and perspective for chronic kidney disease.
- Author
-
Balani S and Perwad F
- Subjects
- Animals, Familial Hypophosphatemic Rickets metabolism, Fibroblast Growth Factor-23, Humans, Phosphates metabolism, Vitamin D metabolism, Antibodies, Monoclonal, Humanized therapeutic use, Familial Hypophosphatemic Rickets drug therapy, Fibroblast Growth Factors antagonists & inhibitors, Renal Insufficiency, Chronic metabolism
- Abstract
Purpose of Review: Perturbations in phosphate and vitamin D homeostasis impacts skeletal health in children and adults. Study of inherited and acquired hypophosphatemic syndromes led to the discovery of fibroblast growth factor 23 (FGF23) as a potent regulator of phosphate and vitamin D metabolism, and advanced our understanding of the pathophysiology of mineral and bone disorder in chronic kidney disease (CKD-MBD). Here, we review a recently approved therapy for patients with X-linked hypophosphatemia (XLH) using a novel anti-FGF23 antibody, burosumab, and discuss the implications of such targeted therapy in CKD., Recent Findings: In children and adults with XLH, burosumab treatment significantly increased renal tubular phosphate reabsorption and normalized serum phosphorus concentrations. Prolonged treatment with burosumab showed a favorable safety profile, improved healing of rickets in children, and fractures and pseudofractures in adults. FGF23 excess in CKD is independently associated with left ventricular hypertrophy and cardiovascular mortality. Research strategies to lower FGF23 in animal models of CKD are rapidly advancing and a question that remains to be answered is whether FGF23 blockade will offer a new targeted intervention for disordered mineral metabolism in CKD., Summary: Findings from recently concluded clinical trials in adults and children with XLH provide evidence for improved skeletal health with burosumab therapy with normalization of phosphate and vitamin D metabolism. Targeted anti-FGF23 antibody treatment of XLH has emerged as a novel therapeutic strategy to treat an inherited disorder of FGF23 excess.
- Published
- 2020
- Full Text
- View/download PDF
107. The international X-linked hypophosphataemia (XLH) registry (NCT03193476): rationale for and description of an international, observational study.
- Author
-
Padidela R, Nilsson O, Makitie O, Beck-Nielsen S, Ariceta G, Schnabel D, Brandi ML, Boot A, Levtchenko E, Smyth M, Jandhyala R, and Mughal Z
- Subjects
- Adult, Child, Fibroblast Growth Factor-23, Humans, Phosphates, Quality of Life, Registries, Vitamin D, Familial Hypophosphatemic Rickets drug therapy, Familial Hypophosphatemic Rickets genetics
- Abstract
Background: X-linked hypophosphataemia (XLH) is a rare, hereditary, progressive and lifelong phosphate wasting disorder characterised by pathological elevations in fibroblast growth factor (FGF) 23 concentration and activity; XLH has an incidence of approximately 1 in 20-25,000 individuals. Excess FGF23 activity leads to increased phosphate excretion in the kidneys - mediated by downregulation of renal tubular phosphate transporters - and reduced phosphate absorption in the intestines - due to impaired vitamin D activation. This results in impaired bone growth and mineralisation, short and disproportionate stature, leg bowing, musculoskeletal pain, spontaneous dental abscesses, rickets, and osteomalacia. The spectrum of manifestations differs between paediatric and adult patients. Those involved in the treatment of this condition face many challenges, including a lack of robust natural history and demographic data. This multicentre, international, rare-disease patient registry (XLH Registry) was established to address the paucity of data in XLH and to help inform future clinical practice., Results: The XLH Registry collects standard diagnostic and monitoring practice data, including (where applicable) diagnosis and disease progression history, treatment regimens and family history; the protocol does not mandate any interventions or clinical assessments. The XLH Registry aims to recruit 1200 paediatric and adult patients with XLH over 10 years, and several data analyses and peer-reviewed publications are expected to be generated throughout this period. A post-authorisation safety study for Bburosumab, for which the registry Sponsor is the marketing authorisation holder, will be nested as a sub-study within the XLH Registry via a subsequent protocol amendment., Conclusion: The data collected within this rare-disease patient registry will be utilised to synthesise real-world evidence to inform the management of XLH, to improve the quality of life and standard of care of patients living with this rare debilitating disease.
- Published
- 2020
- Full Text
- View/download PDF
108. Long-term outcomes for Asian patients with X-linked hypophosphataemia: rationale and design of the SUNFLOWER longitudinal, observational cohort study.
- Author
-
Kubota T, Fukumoto S, Cheong HI, Michigami T, Namba N, Ito N, Tokunaga S, Gibbs Y, and Ozono K
- Subjects
- Adult, Antibodies, Monoclonal, Humanized therapeutic use, Asian People, Child, Clinical Protocols, Disease Progression, Familial Hypophosphatemic Rickets pathology, Humans, Japan, Longitudinal Studies, Republic of Korea, Severity of Illness Index, Treatment Outcome, Familial Hypophosphatemic Rickets drug therapy
- Abstract
Introduction: X-linked hypophosphataemic rickets/osteomalacia (XLH) is a chronic, debilitating genetic disease characterised by skeletal abnormalities and growth disorder. The burden of XLH begins in childhood and continues throughout life. Conventional medical therapy with phosphate, active vitamin D and surgery do not address the underlying pathophysiology of the disease. While treatment during childhood may improve bone deformity and growth retardation, a large proportion of adult patients still fail to reach normal stature. Furthermore, adult patients with XLH report comorbidities associated with unresolved childhood disease, as well as newly developed disease-related complications and significantly impaired quality of life (QOL). Despite the multiple negative aspects of XLH, Asian consensus statements for diagnosis and management are lacking., Methods and Analysis: The Study of longitUdinal observatioN For patients with X-Linked hypOphosphataemic rickets/osteomalacia in collaboration With Asian partnERs study is a longitudinal observational cohort study of patients with XLH, designed to determine the medical characteristics and burdens (physical, emotional and financial) of this progressive disease and to evaluate the impact of treatment (including the use of burosumab) on clinical outcomes. The study was initiated in April 2018, and registration will remain open until 30 April 2022. The sample size planned for analyses is 160 patients, consisting of 100 patients in Japan and 60 patients in Korea. Up to 5 years of observation are planned per patient, from enrolment through to April 2023. Prospective and retrospective data will be collected to evaluate variables, including height/growth, rickets severity score, QOL, motor function and biomarkers for phosphate metabolism and bone turnover., Ethics and Dissemination: Ethics approval was obtained from the Ethics Committee of Osaka University, the Ethics Committee of Kyowa Kirin Co and by the Ethics Committee of each participating medical institution. Two interim analyses and associated publications are planned using retrospective and enrolment data at year 1 and results at year 3., Trial Registration Numbers: NCT03745521; UMIN000031605., Competing Interests: Competing interests: TK has received personal fees from Kyowa Kirin Co for the submitted work and grants from Kyowa Kirin Co outside the submitted work. SF has received grants from Teijin Pharma and Astellas Pharma; and held an endowed chair position with Chugai Pharmaceutical Co, Ono Pharmaceutical Co, Taisho Pharmaceutical Co and Kyowa Kirin Co outside the submitted work. TM has received personal fees (honorarium) from Kyowa Kirin Co for serving as a member of the advisory board during the conduct of this study. NN has received personal fees from Kyowa Kirin Co for the submitted work; and grants from Kyowa Kirin Co outside the submitted work. NI has received research grants from Kyowa Kirin Co outside the submitted work. ST and YG are the employees of Kyowa Kirin Co. KO has received lecture fees from Kyowa Kirin Co, Alexion Pharmaceuticals and Novo Nordisk Pharma outside the submitted work., (© Author(s) (or their employer(s)) 2020. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2020
- Full Text
- View/download PDF
109. Clinical Evidence for the Benefits of Burosumab Therapy for X-Linked Hypophosphatemia (XLH) and Other Conditions in Adults and Children.
- Author
-
Schindeler A, Biggin A, and Munns CF
- Subjects
- Adult, Child, Clinical Trials as Topic, Familial Hypophosphatemic Rickets pathology, Fibroblast Growth Factor-23, Humans, Nephrocalcinosis pathology, Vascular Calcification pathology, Antibodies, Monoclonal, Humanized therapeutic use, Familial Hypophosphatemic Rickets drug therapy, Nephrocalcinosis drug therapy, Vascular Calcification drug therapy
- Abstract
Burosumab (KRN23) is an FGF23 neutralizing antibody that has been the subject of several recent clinical trials principally focused on the treatment of hypophosphatemic rickets in patients with X-linked hypophosphatemia (XLH). Since the first publications in 2014, these trials have demonstrated efficacy with minimal safety concerns in both adult and pediatric cohorts. These studies have used dose-escalation to establish a dosing regimen that is well-tolerated in clinical use. This review summarizes the clinical trial data with respect to burosumab treatment in adults and children as well as noting several clinical trials currently underway. While burosumab appears transformative for the treatment of XLH, long term follow-up studies would be required to allay concerns over the potential for nephrocalcinosis and cardiac calcification. While these do not appear to be problematic in current trials, the effects of chronic or lifelong treatment have yet to be established., (Copyright © 2020 Schindeler, Biggin and Munns.)
- Published
- 2020
- Full Text
- View/download PDF
110. Complications of Phosphate and Vitamin D Treatment in X-Linked Hypophosphataemia.
- Author
-
Arango Sancho P
- Subjects
- Adult, Aged, Aged, 80 and over, Calcitriol therapeutic use, Calcium-Regulating Hormones and Agents therapeutic use, Female, Fibroblast Growth Factor-23, Humans, Male, Middle Aged, Phosphates therapeutic use, Vitamin D therapeutic use, Calcitriol adverse effects, Calcium-Regulating Hormones and Agents adverse effects, Drug-Related Side Effects and Adverse Reactions, Familial Hypophosphatemic Rickets diagnosis, Familial Hypophosphatemic Rickets drug therapy, Phosphates adverse effects, Vitamin D adverse effects
- Abstract
Conventional treatment of X-linked hypophosphataemia (XLH) consists in the oral administration of phosphate plus calcitriol supplements. Although this therapy has reduced bone deformities and even achieved adequate patient growth, overtreatment or low adherence could lead to subsequent consequences that may compromise the efficacy of the therapy. Some of the complications associated with phosphate and vitamin D treatment are abdominal discomfort, diarrhoea, hypokalaemia, hyperparathyroidism, hypercalcaemia or hypercalciuria, nephrocalcinosis or nephrolithiasis, and ectopic calcifications. Therefore, constant multidisciplinary monitoring of patients with XLH is necessary to prevent the manifestation of these complications and to deal with them as soon as they appear. The main objective of this article is to review the main complications arising from conventional treatment of XLH and how to deal with them.
- Published
- 2020
- Full Text
- View/download PDF
111. Panel Discussion: Some Aspects of the Management of Patients with X-Linked Hypophosphataemic Rickets.
- Author
-
Torregrosa JV, Sánchez Del Pozo J, Luiz Yanes MI, and Muñoz Torres M
- Subjects
- Adolescent, Child, Child, Preschool, Drug Dosage Calculations, Female, Fibroblast Growth Factor-23, Humans, Male, Antibodies, Monoclonal therapeutic use, Drug Therapy standards, Familial Hypophosphatemic Rickets diagnosis, Familial Hypophosphatemic Rickets drug therapy, Practice Guidelines as Topic
- Abstract
X-linked hypophosphataemia (XLH) rickets is a rare disease frequently misdiagnosed and mismanaged. Despite having clinical guidelines that offers some therapeutic recommendations based on the clinical experience of experts, physicians still have questions about some important aspects of the diagnosis and treatment of XLH, such as when the disease should be suspected, who should be in charge of the diagnosis, what should be done once the disease is diagnosed, or what therapeutic options are currently available. The objective of this paper is to answer some of the more frequent questions related to the management of patients with XLH by a group of experts participating in a scientific conference on XLH held in Madrid.
- Published
- 2020
- Full Text
- View/download PDF
112. Hereditary vitamin D-resistant rickets: a report of four cases with two novel variants in the VDR gene and successful use of intermittent intravenous calcium via a peripheral route.
- Author
-
Abalı S, Tamura M, Turan S, Atay Z, Isguven P, Güran T, Haliloglu B, Baş S, Isojima T, Kitanaka S, and Bereket A
- Subjects
- Child, Child, Preschool, Familial Hypophosphatemic Rickets genetics, Female, Humans, Infant, Male, Prognosis, Calcium administration & dosage, Calcium-Regulating Hormones and Agents administration & dosage, Familial Hypophosphatemic Rickets drug therapy, Familial Hypophosphatemic Rickets pathology, Mutation, Receptors, Calcitriol genetics
- Abstract
Background Hereditary vitamin D-resistant rickets (HVDRR) is caused by vitamin D receptor (VDR) defects. Patients with HVDRR do not respond to standard doses of calcitriol and oral calcium (Ca) treatment and need to be treated with intravenous Ca (IV-Ca) via a central route. However, central catheter-related complications can cause significant morbidity. Case presentation Four unrelated patients with HVDRR presenting with rickets and alopecia totalis were administered intermittent IV-Ca treatment (2-5 times/week) through a peripheral route. No complications such as infection, extravasation or arrhythmias were detected upon peripheral infusion. Peripheral 1-22 months' duration of IV-Ca normalized parathyroid hormone (PTH) and alkaline phosphatase (ALP) in all patients, after which, oral Ca of 200-400 mg/kg/day and calcitriol of 0.5 μg/kg/day were sufficient to maintain normal PTH levels. Molecular studies on the VDR gene showed a previously reported homozygous c.454C > T (p.Q152*) pathogenic variant in two patients. Two novel homozygous variants in the other two patients were detected: (1) c.756-2A > G, which affects the splice acceptor site, and (2) c.66dupG (p.I23Dfs*20) variant leading to a frameshift that results in a premature stop codon. Conclusions Peripheral IV-Ca treatment is an effective and practical alternative treatment mode that provides dramatic clinical benefit in patients with HVDRR.
- Published
- 2020
- Full Text
- View/download PDF
113. Multidisciplinary patient care in X-linked hypophosphatemic rickets: one challenge, many perspectives.
- Author
-
Raimann A, Mindler GT, Kocijan R, Bekes K, Zwerina J, Haeusler G, and Ganger R
- Subjects
- Adult, Bone and Bones, Child, Fibroblast Growth Factor-23, Fibroblast Growth Factors, Humans, Patient Care, Quality of Life, Rare Diseases, Familial Hypophosphatemic Rickets diagnosis, Familial Hypophosphatemic Rickets drug therapy, Familial Hypophosphatemic Rickets therapy
- Abstract
X‑linked hypophosphatemic rickets (XLH, OMIM #307800) is a rare genetic metabolic disorder caused by dysregulation of fibroblast-like growth factor 23 (FGF23) leading to profound reduction in renal phosphate reabsorption. Impaired growth, severe rickets and complex skeletal deformities are direct consequences of hypophosphatemia representing major symptoms of XLH during childhood. In adults, secondary complications including early development of osteoarthritis substantially impair quality of life and cause significant clinical burden. With the global approval of the monoclonal FGF23 antibody burosumab, a targeted treatment with promising results in phase III studies is available for children with XLH. Nevertheless, complete phenotypic rescue is rarely achieved and remaining multisystemic symptoms demand multidisciplinary specialist care. Coordination of patient management within the major medical disciplines is a mainstay to optimize treatment and reduce disease burden. This review aims to depict different perspectives in XLH patient care in the setting of a multidisciplinary centre of expertise for rare bone diseases.
- Published
- 2020
- Full Text
- View/download PDF
114. Sclerostin Antibody Treatment Increases Bone Mass and Normalizes Circulating Phosphate Levels in Growing Hyp Mice.
- Author
-
Carpenter KA and Ross RD
- Subjects
- Animals, Bone Density, Bone and Bones, Female, Fibroblast Growth Factor-23, Fibroblast Growth Factors, Humans, Male, Mice, Osteocytes, PHEX Phosphate Regulating Neutral Endopeptidase, Phosphates, Familial Hypophosphatemic Rickets drug therapy
- Abstract
X-linked hypophosphatemia (XLH), caused by a loss-of-function mutation in the phosphate regulating gene with homology to endopeptidase located on the X chromosome (PHEX), is the most common form of vitamin D-resistant rickets. Loss of functional PHEX results in elevated fibroblast growth factor 23 (FGF23) levels, impaired phosphate reabsorption, and inhibited skeletal mineralization. Sclerostin, a protein produced primarily in osteocytes, suppresses bone formation by antagonizing Wnt signaling and is reported to be elevated in XLH patients. This study used the Hyp mouse model to investigate sclerostin's role in the pathophysiology of XLH by evaluating the use of a monoclonal antibody to sclerostin in a mouse model of XLH, the Hyp mouse. Male and female wild-type and Hyp littermates were injected with 25 mg/kg of vehicle or sclerostin antibody (Scl-Ab) twice weekly, beginning at 4 weeks of age and euthanized at 8 weeks of age. Scl-Ab treatment increased serum phosphate levels and suppressed circulating levels of intact FGF23 in treated wild-type and Hyp mice of both sexes. Cortical area, trabecular bone volume fraction (BV/TV), metaphyseal apparent density, and the peak load increased with Scl-Ab treatment in both sexes. This short-term treatment study suggests that Scl-Ab treatment can effectively improve some of the pathologies associated with XLH, including normalization of phosphate, and that sclerostin may play a role in regulating FGF23 and phosphate metabolism in XLH. © 2019 American Society for Bone and Mineral Research., (© 2019 American Society for Bone and Mineral Research.)
- Published
- 2020
- Full Text
- View/download PDF
115. Cinacalcet treatment experience in hereditary vitamin D resistant rickets.
- Author
-
Lucas J, Badia JL, Lucas E, and Remon A
- Subjects
- Child, Preschool, Familial Hypophosphatemic Rickets genetics, Familial Hypophosphatemic Rickets pathology, Humans, Male, Prognosis, Salvage Therapy, Vitamins pharmacology, Calcium-Regulating Hormones and Agents therapeutic use, Cinacalcet therapeutic use, Drug Resistance drug effects, Familial Hypophosphatemic Rickets drug therapy, Vitamin D pharmacology
- Abstract
Background Hereditary vitamin D resistant rickets (HVDRR) is a bone disorder characterized by a phenotype of rickets with onset at early stage of life with elevated alkaline phosphatase, hypocalcemia, hypophosphatemia, hyperparathyroidism and elevated levels of 1,25-dihydroxyvitamin D (calcitriol) as a consequence of the resistance of the vitamin D receptor (VDR). Mutations in the DNA-binding domain of the VDR of the vitamin D receptor have been characterized by a lack of response to traditional treatment with calcium and calcitriol. Secondary hyperparathyroidism and hypophosphatemia are the main factors in its pathogenesis. Cinacalcet is a calciomimetic drug that reproduces the action of calcium by increasing the sensitivity of the calcium-sensitive receptors (CASR) of the parathyroid glands that regulate the secretion of the parathyroid hormone (PTH). Case presentation We describe its effectiveness and safety in a patient with HVDRR and review other published report cases in the literature. According to published experience, cinacalcet could be used as an adjunctive treatment for the HVDRR with mutations in the DNA-binding domain of the VDR refractory to traditional treatment. Due to lack of knowledge of possible effects of cinacalcet on CASR in the skeleton, long-term use should be avoided. Conclusions The optimal dose of cinacalcet for treatment of HVDRR ranges between 0.25 and 0.5 mg/kg/day. Serious side effects of cinacalcet have not been published in this type of patient, although we considered that a close monitoring is necessary in order to detect hypocalcemia.
- Published
- 2020
- Full Text
- View/download PDF
116. Iron replacement ameliorates hypophosphatemia in autosomal dominant hypophosphatemic rickets: A review of the role of iron.
- Author
-
Menon LP and Weinstein RS
- Subjects
- Adult, Calcitriol, Fibroblast Growth Factor-23, Fibroblast Growth Factors, Humans, Iron, Phosphates, Familial Hypophosphatemic Rickets complications, Familial Hypophosphatemic Rickets drug therapy, Familial Hypophosphatemic Rickets genetics, Hypophosphatemia
- Abstract
Autosomal dominant hypophosphatemic rickets (ADHR) is remarkable among the hypophosphatemic rickets syndromes for its variable age of presentation and periods of quiescence during which serum phosphate and fibroblast growth factor 23 (FGF 23) levels are normal without therapy. In contrast, hypophosphatemia in X-linked hypophosphatemic rickets (XLH) manifests soon after birth and requires lifelong therapy. This suggests that there are environmental factors which can alter FGF 23 activity in ADHR but not in XLH. We present an adult with ADHR in whom resolution of hypophosphatemia was achieved by correcting iron deficiency without the need for phosphate supplementation. Serial iron and FGF 23 levels revealed an inverse relationship (r=-0.79, p<0.04). All patients with ADHR who present with hypophosphatemia and worsening symptoms should be screened for iron deficiency. If iron deficiency is detected, therapy with a combination of calcitriol and iron supplementation should be considered without phosphate supplementation., (Copyright © 2019 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
117. Oral Iron Replacement Normalizes Fibroblast Growth Factor 23 in Iron-Deficient Patients With Autosomal Dominant Hypophosphatemic Rickets.
- Author
-
Imel EA, Liu Z, Coffman M, Acton D, Mehta R, and Econs MJ
- Subjects
- Adult, Aged, Female, Fibroblast Growth Factor-23, Fibroblast Growth Factors genetics, Humans, Iron, Male, Middle Aged, Phosphates, Prospective Studies, Familial Hypophosphatemic Rickets drug therapy, Familial Hypophosphatemic Rickets genetics
- Abstract
Autosomal dominant hypophosphatemic rickets (ADHR) is caused by mutations impairing cleavage of fibroblast growth factor 23 (FGF23). FGF23 gene expression increases during iron deficiency. In humans and mice with the ADHR mutation, iron deficiency results in increased intact FGF23 concentrations and hypophosphatemia. We conducted a prospective open label pilot clinical trial of oral iron replacement over 12 months in ADHR patients to test the hypothesis that oral iron administration would normalize FGF23 concentrations. Eligibility criteria included: FGF23 mutation; and either serum iron <50 μg/dL; or serum iron 50 to 100 μg/dL combined with hypophosphatemia and intact FGF23 >30 pg/mL at screening. Key exclusion criteria were kidney disease and pregnancy. Oral iron supplementation started at 65 mg daily and was titrated based on fasting serum iron concentration. The primary outcome was decrease in fasting intact FGF23 by ≥20% from baseline. Six adults (three male, three female) having the FGF23-R176Q mutation were enrolled; five completed the 12-month protocol. At baseline three of five subjects had severely symptomatic hypophosphatemia (phosphorus <2.5 mg/dL) and received calcitriol with or without phosphate concurrent with oral iron during the trial. The primary outcome was met by 4 of 5 (80%) subjects all by month 4, and 5 of 5 had normal intact FGF23 at month 12. Median (minimum, maximum) intact FGF23 concentration decreased from 172 (20, 192) pg/mL at baseline to 47 (17, 78) pg/mL at month 4 and 42 (19, 63) pg/mL at month 12. Median ferritin increased from 18.6 (7.7, 82.5) ng/mL at baseline to 78.0 (49.6, 261.0) ng/mL at month 12. During iron treatment, all three subjects with baseline hypophosphatemia normalized serum phosphorus, had markedly improved symptoms, and were able to discontinue calcitriol and phosphate. Oral iron repletion normalized FGF23 and phosphorus in symptomatic, iron-deficient ADHR subjects. Thus, the standard approach to ADHR should include recognition, treatment, and prevention of iron deficiency. © 2019 American Society for Bone and Mineral Research., (© 2019 American Society for Bone and Mineral Research.)
- Published
- 2020
- Full Text
- View/download PDF
118. Oral Iron for Prevention and Treatment of Rickets and Osteomalacia in Autosomal Dominant Hypophosphatemia.
- Author
-
Högler W and Kapelari K
- Subjects
- Female, Fibroblast Growth Factor-23, Fibroblast Growth Factors, Humans, Iron, Phosphates, Familial Hypophosphatemic Rickets drug therapy, Familial Hypophosphatemic Rickets genetics, Osteomalacia drug therapy, Osteomalacia prevention & control
- Published
- 2020
- Full Text
- View/download PDF
119. Burosumab Improved Histomorphometric Measures of Osteomalacia in Adults with X-Linked Hypophosphatemia: A Phase 3, Single-Arm, International Trial.
- Author
-
Insogna KL, Rauch F, Kamenický P, Ito N, Kubota T, Nakamura A, Zhang L, Mealiffe M, San Martin J, and Portale AA
- Subjects
- Adult, Antibodies, Monoclonal adverse effects, Antibodies, Monoclonal, Humanized, Biomarkers blood, Familial Hypophosphatemic Rickets blood, Female, Fibroblast Growth Factor-23, Humans, Male, Osteogenesis, Patient Reported Outcome Measures, Treatment Outcome, Antibodies, Monoclonal therapeutic use, Familial Hypophosphatemic Rickets drug therapy, Internationality, Osteomalacia drug therapy
- Abstract
In adults with X-linked hypophosphatemia (XLH), excess FGF23 impairs renal phosphate reabsorption and suppresses production of 1,25-dihydroxyvitamin D, resulting in chronic hypophosphatemia and persistent osteomalacia. Osteomalacia is associated with poor bone quality causing atraumatic fractures, pseudofractures, delayed fracture healing, and bone pain. Burosumab is a fully human monoclonal antibody against FGF23. UX023-CL304 is an ongoing, open-label, single-arm, phase 3 study investigating the efficacy of subcutaneous burosumab, 1.0 mg/kg administered every 4 weeks, in improving osteomalacia in adults with XLH who have not been treated for at least 2 years before enrollment. The primary endpoint was improvement in osteoid volume/bone volume assessed by transiliac bone biopsies obtained at baseline and week 48. Additional assessments included serum phosphorus, markers of bone turnover, fracture/pseudofracture healing, and safety. Fourteen subjects enrolled, 13 completed 48 weeks, and 11 completed paired biopsies. All osteomalacia-related histomorphometric measures improved significantly at week 48 (mean percent change: osteoid volume/bone volume, -54%, osteoid thickness, -32%, osteoid surface/bone surface, -26%, [median] mineralization lag time, -83%). Mean serum phosphorus concentration averaged across the mid-point of the dose cycle between weeks 0 and 24 was 3.3 mg/dL, a 50% increase from 2.2 mg/dL at baseline. Markers of bone formation and resorption increased at week 48 (least squares [LS] mean increase: P1NP, +77%; CTx, +36%; both p < 0.0001). All subjects had one or more treatment-emergent adverse event (AE). Most AEs were mild to moderate in severity. Two subjects experienced serious AEs (migraine; paresthesia) that were unrelated to treatment and resolved. Eleven subjects had 18 biopsy procedure-related AEs: 14 for pain, two for itch, and one each for headache and bandage irritation. No deaths or incidents of hyperphosphatemia occurred. In conclusion, by normalizing phosphate homeostasis, burosumab significantly improved osteomalacia in adults with XLH, which likely explains the improved fracture healing and amelioration of skeletal complications. © 2019 The Authors. Journal of Bone and Mineral Research published by American Society for Bone and Mineral Research., (© 2019 The Authors. Journal of Bone and Mineral Research published by American Society for Bone and Mineral Research.)
- Published
- 2019
- Full Text
- View/download PDF
120. Continued Beneficial Effects of Burosumab in Adults with X-Linked Hypophosphatemia: Results from a 24-Week Treatment Continuation Period After a 24-Week Double-Blind Placebo-Controlled Period.
- Author
-
Portale AA, Carpenter TO, Brandi ML, Briot K, Cheong HI, Cohen-Solal M, Crowley R, Jan De Beur S, Eastell R, Imanishi Y, Imel EA, Ing S, Ito N, Javaid M, Kamenicky P, Keen R, Kubota T, Lachmann R, Perwad F, Pitukcheewanont P, Ralston SH, Takeuchi Y, Tanaka H, Weber TJ, Yoo HW, Zhang L, Theodore-Oklota C, Mealiffe M, San Martin J, and Insogna K
- Subjects
- Adolescent, Adult, Aged, Antibodies, Monoclonal adverse effects, Antibodies, Monoclonal, Humanized, Double-Blind Method, Drug Administration Schedule, Female, Fibroblast Growth Factor-23, Humans, Male, Middle Aged, Placebos, Treatment Outcome, Young Adult, Antibodies, Monoclonal administration & dosage, Familial Hypophosphatemic Rickets drug therapy, Maintenance Chemotherapy
- Abstract
Burosumab, a fully human monoclonal antibody to FGF23, is the only approved treatment for X-linked hypophosphatemia (XLH), a rare genetic disorder characterized by renal phosphate wasting and substantial cumulative musculoskeletal morbidity. During an initial 24-week randomized, controlled trial, 134 adults with XLH received burosumab 1 mg/kg (n = 68) or placebo (n = 66) every 4 weeks. After 24 weeks, all subjects received open-label burosumab until week 48. This report describes the efficacy and safety of burosumab during the open-label treatment period. From weeks 24-48, serum phosphorus concentrations remained normal in 83.8% of participants who received burosumab throughout and were normalized in 89.4% who received burosumab after placebo. By week 48, 63.1% of baseline fractures/pseudofractures healed fully with burosumab, compared with 35.2% with burosumab after placebo. In both groups, burosumab was associated with clinically significant and sustained improvement from baseline to week 48 in scores for patient-reported outcomes of stiffness, pain, physical function, and total distance walked in 6 min. Rates of adverse events were similar for burosumab and placebo. There were no fatal adverse events or treatment-related serious adverse events. Nephrocalcinosis scores did not change from baseline by more than one grade at either week 24 or 48. These data demonstrate that in participants with XLH, continued treatment with burosumab is well tolerated and leads to sustained correction of serum phosphorus levels, continued healing of fractures and pseudofractures, and sustained improvement in key musculoskeletal impairments.
- Published
- 2019
- Full Text
- View/download PDF
121. Burosumab Therapy for X-Linked Hypophosphatemia and Therapeutic Implications for CKD.
- Author
-
Perwad F and Portale AA
- Subjects
- Animals, Antibodies, Monoclonal, Humanized, Fibroblast Growth Factor-23, Fibroblast Growth Factors antagonists & inhibitors, Fibroblast Growth Factors blood, Humans, Vitamin D analogs & derivatives, Vitamin D blood, Antibodies, Monoclonal therapeutic use, Familial Hypophosphatemic Rickets drug therapy, Renal Insufficiency, Chronic drug therapy
- Published
- 2019
- Full Text
- View/download PDF
122. MAPK inhibition and growth hormone: a promising therapy in XLH.
- Author
-
Fuente R, Gil-Peña H, Claramunt-Taberner D, Hernández-Frías O, Fernández-Iglesias Á, Alonso-Durán L, Rodríguez-Rubio E, Hermida-Prado F, Anes-González G, Rubio-Aliaga I, Wagner C, and Santos F
- Subjects
- Animals, Disease Models, Animal, Extracellular Signal-Regulated MAP Kinases genetics, Familial Hypophosphatemic Rickets genetics, Familial Hypophosphatemic Rickets metabolism, Familial Hypophosphatemic Rickets pathology, Fibroblast Growth Factor-23, MAP Kinase Signaling System genetics, Mice, Mice, Knockout, Extracellular Signal-Regulated MAP Kinases metabolism, Familial Hypophosphatemic Rickets drug therapy, Growth Hormone pharmacology, MAP Kinase Signaling System drug effects
- Abstract
X-linked hypophosphatemia (XLH) leads to growth retardation and bone deformities, which are not fully avoided by conventional treatment with phosphate and vitamin D analogs. Pediatric patients have been treated with growth hormone (GH), and recent findings suggest that blocking fibroblast growth factor 23 actions may be the most effective therapy, but its effects on growth are not known. We here report the effect of MAPK inhibition alone or associated with GH on growth and growth plate and bone structure of young Hyp (the XLH animal model) mice. Untreated Hyp mice were severely growth retarded and had marked alterations in both growth plate structure and dynamics as well as defective bone mineralization. GH accelerated growth and improved mineralization and the cortical bone, but it failed in normalizing growth plate and trabecular bone structures. MAPK inhibition improved growth and rickets and, notably, almost normalized the growth plate organization. The administration of a MAPK pathway inhibitor plus GH was the most beneficial treatment because of the positive synergistic effect on growth plate and bone structures. Thus, the growth-promoting effect of GH is likely linked to increased risk of bone deformities, whereas the association of GH and MAPK inhibition emerges as a promising new therapy for children with XLH.-Fuente, R., Gil-Peña, H., Claramunt-Taberner, D., Hernández-Frías, O., Fernández-Iglesias, Á., Alonso-Durán, L., Rodríguez-Rubio, E., Hermida-Prado, F., Anes-González, G., Rubio-Aliaga, I., Wagner, C., Santos, F. MAPK inhibition and growth hormone: a promising therapy in XLH.
- Published
- 2019
- Full Text
- View/download PDF
123. Experience of intravenous calcium treatment and long-term responses to treatment in a patient with hereditary vitamin D-resistant rickets resulting from a novel mutation.
- Author
-
Bayramoğlu E, Şavaş Erdeve Ş, Shi Y, Keskin M, Çetinkaya S, Kurnaz E, Muratoğlu Şahin N, and Aycan Z
- Subjects
- Administration, Intravenous, Child, Preschool, Familial Hypophosphatemic Rickets genetics, Familial Hypophosphatemic Rickets pathology, Female, Humans, Prognosis, Calcium administration & dosage, Calcium-Regulating Hormones and Agents administration & dosage, Familial Hypophosphatemic Rickets drug therapy, Mutation, Receptors, Calcitriol genetics
- Abstract
Background Vitamin D resistant rickets (HVDRR), is a rare autosomal recessive disorder caused by vitamin D receptor (VDR) gene mutations. There is no standard treatment in HVDRR. Case report The patient was a 3-year-old girl presenting with short stature, genu varum deformity, waddling gait and alopecia. She had hypocalcemia, hypophosphatemia, hyperparathyroidism and normal 1.25-(OH)2D levels. The patient was initially treated with calcitriol and high-dose oral calcium (Ca) for 22 months. The patient was treated with continuous high dose intravenous (i.v.) Ca therapy for 4 months, following initial lack of response to oral Ca and calsitriol. At the end of the 4 months, rickets was dramatically improved and did not recur for 3 years after i.v. Ca therapy. DNA sequence analyses of the VDR gene showed a homozygous novel mutation. Conclusions We identified a novel VDR gene mutation, and we concluded that i.v. Ca therapy from the central catheter is a safe treatment in HVDRR.
- Published
- 2019
- Full Text
- View/download PDF
124. Burosumab versus conventional therapy in children with X-linked hypophosphataemia: a randomised, active-controlled, open-label, phase 3 trial.
- Author
-
Imel EA, Glorieux FH, Whyte MP, Munns CF, Ward LM, Nilsson O, Simmons JH, Padidela R, Namba N, Cheong HI, Pitukcheewanont P, Sochett E, Högler W, Muroya K, Tanaka H, Gottesman GS, Biggin A, Perwad F, Mao M, Chen CY, Skrinar A, San Martin J, and Portale AA
- Subjects
- Antibodies, Monoclonal, Humanized, Body Height, Child, Child Development, Child, Preschool, Familial Hypophosphatemic Rickets diagnosis, Female, Fibroblast Growth Factor-23, Humans, Infant, Male, Severity of Illness Index, Treatment Outcome, Antibodies, Monoclonal therapeutic use, Familial Hypophosphatemic Rickets drug therapy, Immunologic Factors therapeutic use
- Abstract
Background: X-linked hypophosphataemia in children is characterised by elevated serum concentrations of fibroblast growth factor 23 (FGF23), hypophosphataemia, rickets, lower extremity bowing, and growth impairment. We compared the efficacy and safety of continuing conventional therapy, consisting of oral phosphate and active vitamin D, versus switching to burosumab, a fully human monoclonal antibody against FGF23, in paediatric X-linked hypophosphataemia., Methods: In this randomised, active-controlled, open-label, phase 3 trial at 16 clinical sites, we enrolled children with X-linked hypophosphataemia aged 1-12 years. Key eligibility criteria were a total Thacher rickets severity score of at least 2·0, fasting serum phosphorus lower than 0·97 mmol/L (3·0 mg/dL), confirmed PHEX (phosphate-regulating endopeptidase homolog, X-linked) mutation or variant of unknown significance in the patient or a family member with appropriate X-linked dominant inheritance, and receipt of conventional therapy for at least 6 consecutive months for children younger than 3 years or at least 12 consecutive months for children older than 3 years. Eligible patients were randomly assigned (1:1) to receive either subcutaneous burosumab starting at 0·8 mg/kg every 2 weeks (burosumab group) or conventional therapy prescribed by investigators (conventional therapy group). Both interventions lasted 64 weeks. The primary endpoint was change in rickets severity at week 40, assessed by the Radiographic Global Impression of Change global score. All patients who received at least one dose of treatment were included in the primary and safety analyses. The trial is registered with ClinicalTrials.gov, number NCT02915705., Findings: Recruitment took place between Aug 3, 2016, and May 8, 2017. Of 122 patients assessed, 61 were enrolled. Of these, 32 (18 girls, 14 boys) were randomly assigned to continue receiving conventional therapy and 29 (16 girls, 13 boys) to receive burosumab. For the primary endpoint at week 40, patients in the burosumab group had significantly greater improvement in Radiographic Global Impression of Change global score than did patients in the conventional therapy group (least squares mean +1·9 [SE 0·1] with burosumab vs +0·8 [0·1] with conventional therapy; difference 1·1, 95% CI 0·8-1·5; p<0·0001). Treatment-emergent adverse events considered possibly, probably, or definitely related to treatment by the investigator occurred more frequently with burosumab (17 [59%] of 29 patients in the burosumab group vs seven [22%] of 32 patients in the conventional therapy group). Three serious adverse events occurred in each group, all considered unrelated to treatment and resolved., Interpretation: Significantly greater clinical improvements were shown in rickets severity, growth, and biochemistries among children with X-linked hypophosphataemia treated with burosumab compared with those continuing conventional therapy., Funding: Ultragenyx Pharmaceutical and Kyowa Kirin International., (Copyright © 2019 Elsevier Ltd. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
125. Pharmacological management of X-linked hypophosphataemia.
- Author
-
Imel EA and White KE
- Subjects
- Antibodies, Monoclonal adverse effects, Antibodies, Monoclonal, Humanized, Biomarkers blood, Familial Hypophosphatemic Rickets diagnosis, Familial Hypophosphatemic Rickets genetics, Fibroblast Growth Factor-23, Fibroblast Growth Factors blood, Fibroblast Growth Factors immunology, Genetic Predisposition to Disease, Humans, Mutation, PHEX Phosphate Regulating Neutral Endopeptidase genetics, Phenotype, Treatment Outcome, Up-Regulation, Antibodies, Monoclonal therapeutic use, Familial Hypophosphatemic Rickets drug therapy, Fibroblast Growth Factors antagonists & inhibitors
- 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., (© 2018 The British Pharmacological Society.)
- Published
- 2019
- Full Text
- View/download PDF
126. A novel heterozygous mutation c.680A>G (p. N227S) in SLC34A1 gene leading to autosomal dominant hypophosphatemia: A case report.
- Author
-
Chen X, Xie Y, Wan S, Xu J, Cai B, Zhang Y, and Yu X
- Subjects
- Adult, Familial Hypophosphatemic Rickets diagnosis, Humans, Male, Mutation, Missense, Rickets diagnosis, Familial Hypophosphatemic Rickets drug therapy, Familial Hypophosphatemic Rickets genetics, Phosphorus therapeutic use, Rickets drug therapy, Rickets genetics, Sodium-Phosphate Cotransporter Proteins, Type IIa genetics
- Abstract
Rationale: Currently, the relationship between heterozygous mutations in SLC34A1 and hypophosphatemia is controversial. Here we report an autosomal dominant hypophosphatemia pedigree carrying a novel heterozygous mutation in SLC34A1., Patient Concerns: The proband is a 32-year old young man, presented with progressive pain and weakness in his lower extremities for more than 5 years. The proband showed persistent hypophosphatemia and low TmPO4/GFR values, indicating renal phosphate leak. His grandfather, father, and one of his uncles showed the similar symptoms., Diagnoses: Autosomal dominant hypophosphatemia., Interventions and Outcomes: Phosphorus supplement was prescribed to the proband and his affected uncle. Both their serum phosphorus levels recovered to normal and their symptoms such as back pain and lower extremity weakness were completely relieved. Whole exome sequencing was performed to identify disease-causing mutations in proband., Lessons: A novel heterozygous missense mutation c.680A>G (p. N227S) in exon 7 of SLC34A1 was found in proband by whole exome sequencing, which was also found in other 4 family members of this pedigree. Our report of an autosomal dominant hypophosphatemia pedigree with 5 mutant carriers enriches the clinical phenotype caused by the SLC34A1 mutations and further affirms the heterozygous mutations are causative for hypophosphatemia.
- Published
- 2019
- Full Text
- View/download PDF
127. Rickets severity predicts clinical outcomes in children with X-linked hypophosphatemia: Utility of the radiographic Rickets Severity Score.
- Author
-
Thacher TD, Pettifor JM, Tebben PJ, Creo AL, Skrinar A, Mao M, Chen CY, Chang T, San Martin J, and Carpenter TO
- Subjects
- Alkaline Phosphatase blood, Antibodies, Monoclonal therapeutic use, Antibodies, Monoclonal, Humanized, Child, Child, Preschool, Familial Hypophosphatemic Rickets blood, Familial Hypophosphatemic Rickets drug therapy, Fibroblast Growth Factor-23, Humans, Reproducibility of Results, Treatment Outcome, Familial Hypophosphatemic Rickets diagnostic imaging, Severity of Illness Index
- Abstract
The Rickets Severity Score (RSS) was used to evaluate X-linked hypophosphatemic rickets (XLH), a genetic disorder mediated by increased circulating FGF23. The reliability of the RSS was assessed using data from a randomized, phase 2 clinical trial that evaluated the effects of burosumab, a fully human anti-FGF23 monoclonal antibody, in 52 children with XLH ages 5 to 12 years. Bilateral knee and wrist radiographs were obtained at baseline, week 40, and week 64. We evaluated the relationships of the RSS to the Radiographic Global Impression of Change (RGI-C), serum alkaline phosphatase (ALP), height Z-score, 6-minute walk test (6MWT) percent predicted, and the Pediatric Orthopedic Society of North America Pediatric Outcomes Data Collection Instrument (POSNA-PODCI). The RSS showed moderate-to-substantial inter-rater reliability (weighted kappa, 0.45-0.65; Pearson correlation coefficient (r), 0.83-0.89) and substantial intra-rater reliability (weighted Kappa, 0.66; r = 0.91). Baseline RSS correlated with serum ALP (r = 0.47). Baseline RSS identified two subgroups (higher [RSS ≥1.5] and lower RSS [RSS <1.5]) that discriminated between subjects with greater and lesser rachitic disease. Higher RSS was associated with more severe clinical features, including impaired growth (Z-score, -2.12 vs -1.44) and walking ability (6MWT percent predicted, 77% vs 86%), more severe self-reported pain (29.9 [more severe] vs 45.3 [less severe]) and less physical function (29.6 [more severe] vs 40.9 [less severe]). During burosumab treatment, greater reductions in RSS corresponded to higher RGI-C global scores (r = -0.65). Improvements in RSS correlated with decreased serum ALP (r = 0.47). These results show the reliability of the RSS in XLH, and demonstrate that higher RSS values are associated with greater biochemical, clinical, and functional impairments in children with XLH., (Copyright © 2019 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
128. Efficacy and safety of burosumab in children aged 1-4 years with X-linked hypophosphataemia: a multicentre, open-label, phase 2 trial.
- Author
-
Whyte MP, Carpenter TO, Gottesman GS, Mao M, Skrinar A, San Martin J, and Imel EA
- Subjects
- Antibodies, Monoclonal, Humanized, Biomarkers analysis, Case-Control Studies, Child, Preschool, Female, Fibroblast Growth Factor-23, Follow-Up Studies, Humans, Infant, Male, Patient Safety, Prognosis, Antibodies, Monoclonal therapeutic use, Body Height drug effects, Familial Hypophosphatemic Rickets drug therapy, Fibroblast Growth Factors antagonists & inhibitors
- Abstract
Background: Children with X-linked hypophosphataemia have high concentrations of circulating phosphatonin fibroblast growth factor 23 (FGF23), which causes renal phosphate wasting and hypophosphataemia, rickets, skeletal deformities, and growth impairment. Burosumab, a human monoclonal antibody against FGF23, improves phosphate homoeostasis and rickets in children aged 5-12 years with X-linked hypophosphataemia. We aimed to assess the safety and efficacy of burosumab in younger children with X-linked hypophosphataemia., Methods: In this open-label, phase 2 trial at three hospitals in the USA, children (aged 1-4 years) with X-linked hypophosphataemia received burosumab (0·8 mg/kg) via subcutaneous injection every 2 weeks for 64 weeks. The dose was increased to 1·2 mg/kg if two consecutive pre-dose serum phosphorus concentrations were below 1·03 mmol/L (3·2 mg/dL), serum phosphorus had increased by less than 0·16 mmol/L (<0·5 mg/dL) from baseline, and a dose of burosumab had not been missed. Participants could continue to receive burosumab for up to an additional 96 weeks during the extension period. Key inclusion criteria were age 1-4 years at the time of informed consent; fasting serum phosphorus concentration of less than 0·97 mmol/L (3·0 mg/dL); serum creatinine 8·8-35·4 μmol/L (0·1-0·4 mg/dL); radiographic evidence of rickets (at least five participants were required to have a Thacher Rickets Severity Score of ≥1·5 at the knee); and a confirmed PHEX mutation or a variant of unknown significance in the patient or direct relative also affected with X-linked hypophosphataemia. Conventional therapy was stopped upon enrolment. The coprimary endpoints were safety and change from baseline to week 40 in fasting serum phosphorus concentrations. Changes in rickets severity from baseline to weeks 40 and 64 (assessed radiographically using Thacher Rickets Severity Score and an adaptation of the Radiographic Global Impression of Change), and recumbent length or standing height, were key secondary outcomes. This trial is registered with ClinicalTrials.gov, number NCT02750618, and is ongoing., Findings: Between May 16, 2016, and June 10, 2016, we enrolled 13 children with X-linked hypophosphataemia. All 13 children completed 64 weeks of treatment and were included in the efficacy and safety analysis; none exceeded 70 weeks of treatment at the time of analysis. Serum phosphorus least squares mean increase from baseline to week 40 of treatment was 0·31 mmol/L (SE 0·04; 95% CI 0·24-0·39; 0·96 mg/dL [SE 0·12]; p<0·0001). All patients had at least one adverse event. 14 treatment-related adverse events, mostly injection site reactions, occurred in five children. One serious adverse event considered unrelated to treatment (tooth abscess) occurred in a child with a history of tooth abscess. All other adverse events were mild to moderate, except a severe food allergy considered unrelated to treatment. No instances of nephrocalcinosis or noteworthy changes in the results of a standard safety chemistry panel emerged. Total Thacher Rickets Severity Score decreased by a least squares mean of -1·7 (SE 0·1; p<0·0001) from baseline to week 40 and by -2·0 (SE 0·1; p<0·0001) by week 64. The Radiographic Global Impression of Change score also indicated significant improvement, with a least squares mean score of +2·3 (SE 0·1) at week 40 and +2·2 (0·1) at week 64 (both p<0·0001). Mean length or standing height Z score was maintained from baseline to week 64., Interpretation: Burosumab had a favourable safety profile, increased serum phosphorus, and improved rickets and prevented early declines in growth in children aged 1-4 years with X-linked hypophosphataemia. These findings could substantially alter the treatment of young children with X-linked hypophosphataemia., Funding: Ultragenyx Pharmaceutical and Kyowa Kirin International., (Copyright © 2019 Elsevier Ltd. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
129. A genetic variant of CYP2R1 identified in a cat with type 1B vitamin D-dependent rickets: a case report.
- Author
-
Teshima T, Kurita S, Sasaki T, Matsumoto H, Niina A, Abe D, Kanno N, and Koyama H
- Subjects
- Animals, Calcitriol therapeutic use, Cat Diseases diagnosis, Cat Diseases drug therapy, Cats, Familial Hypophosphatemic Rickets diagnosis, Familial Hypophosphatemic Rickets drug therapy, Familial Hypophosphatemic Rickets genetics, Female, Frameshift Mutation genetics, Sequence Analysis, DNA veterinary, Vitamins therapeutic use, Cat Diseases genetics, Cytochrome P450 Family 2 genetics, Familial Hypophosphatemic Rickets veterinary
- Abstract
Background: Vitamin D-dependent rickets is rare in animals and humans. Several types of this condition are associated with genetic variants related to vitamin D metabolism. This is the first report of type 1B vitamin D-dependent rickets in a cat., Case Presentation: Here, we describe the case of a 3-month-old female domestic short-haired cat previously fed on commercial kitten food that presented at our clinic with seizures, lethargy, and generalized pain. Serum and ionized calcium concentrations and 1,25-dihydroxycholecalciferol in this cat were low, and radiographs showed skeletal demineralization and abnormally wide growth plates on the long bones. Initially, simple vitamin D deficiency was suspected; however, the cat's profile, which included fed a well-balanced commercial diet, together with the findings of additional laboratory tests and the cat's unresponsiveness to various treatments, raised the suspicion of vitamin D-dependent rickets. Examination of the DNA sequences of CYP2R1 and CYP27B1 genes, which are genes linked with vitamin D metabolism, showed a CYP2R1 frameshift mutation in exon 5 (where T is deleted at position c.1386). This mutation alters the amino acid sequence from position 462, while the stop codon introduced at position 481 prematurely truncates the 501 amino acid full-length protein. With this knowledge, a new treatment regime based on a standard dose of calcitriol was started and this markedly improved the cat's condition., Conclusions: To the best of our knowledge, the present case is the first description of type 1B vitamin D-dependent rickets linked with a genetic variant of CYP2R1 in a cat.
- Published
- 2019
- Full Text
- View/download PDF
130. Mutation update and long-term outcome after treatment with active vitamin D 3 in Chinese patients with pseudovitamin D-deficiency rickets (PDDR).
- Author
-
Chi Y, Sun J, Pang L, Jiajue R, Jiang Y, Wang O, Li M, Xing X, Hu Y, Zhou X, Meng X, and Xia W
- Subjects
- Adolescent, Alkaline Phosphatase blood, Body Height drug effects, Calcium-Regulating Hormones and Agents administration & dosage, Calcium-Regulating Hormones and Agents pharmacology, Child, Child, Preschool, Cholecalciferol administration & dosage, Cholecalciferol pharmacology, DNA Mutational Analysis methods, Drug Administration Schedule, Familial Hypophosphatemic Rickets blood, Familial Hypophosphatemic Rickets diagnostic imaging, Female, Humans, Male, Parathyroid Hormone blood, Pedigree, Radiography, Treatment Outcome, Young Adult, 25-Hydroxyvitamin D3 1-alpha-Hydroxylase genetics, Calcium-Regulating Hormones and Agents therapeutic use, Cholecalciferol therapeutic use, Familial Hypophosphatemic Rickets drug therapy, Familial Hypophosphatemic Rickets genetics, Mutation
- Abstract
Pseudovitamin D-deficiency rickets is a rare disease which is caused by CYP27B1. In this study, we identified 9 mutations in 7 PDDR patients. In addition, we observed the response to long-term treatment of calcitriol in 15 Chinese patients with PDDR, which showed that the biochemical abnormalities had been corrected satisfactorily after 1-year treatment., Introduction: Pseudovitamin D-deficiency rickets is a rare autosomal recessive disorder resulting from a defect in 25-hydroxyvitamin D 1α-hydroxylase, which is encoded by CYP27B1. The purpose of this study was to identify the CYP27B1 mutations and investigate the response to long-term treatment of calcitriol in Chinese patients with PDDR., Methods: We investigated CYP27B1 mutations in seven individuals from six separate families. To investigate the response to long-term (13 years) treatment with calcitriol in PDDR patients, we additionally collected clinical data of eight families from our previous report and analyzed their biochemical parameter and radiographic changes during the treatment., Results: Nine different mutations were identified: two novel missense mutations (G194R, R259L), three novel and one reported deletion mutations (c1442delA, c1504delA, c311-321del, and c. 48-60del), two novel nonsense mutations (c.85G>T, c.580G>T), and a reported insertion mutation (c1325-1332insCCCACCC). The statistical analysis revealed that parathyroid hormone (PTH) and ALP significantly decreased after 6-month and 1-year treatment with calcitriol respectively. Urine calcium was measured in all the patients without kidney stones being documented. After 6-year treatment, the radiographic abnormalities had also been improved. Two patients who had reached their final height are both with short stature (height Z-score below - 2.0)., Conclusions: We identified seven novel mutations of CYP27B1 gene in seven Chinese PDDR families. Our findings revealed after 1-year treatment of active vitamin D
3 , PTH and ALP significantly decreased. The correction of the biochemical abnormalities had not improved the final height satisfactorily.- Published
- 2019
- Full Text
- View/download PDF
131. Hereditary hypophosphatemic rickets with hypercalciuria: pathophysiology, clinical presentation, diagnosis and therapy.
- Author
-
Bergwitz C and Miyamoto KI
- Subjects
- Animals, Familial Hypophosphatemic Rickets diagnosis, Familial Hypophosphatemic Rickets drug therapy, Fibroblast Growth Factor-23, Humans, Hypercalciuria diagnosis, Hypercalciuria drug therapy, Loss of Function Mutation, Phosphates therapeutic use, Sodium-Phosphate Cotransporter Proteins, Type IIc metabolism, Vitamin D blood, Vitamin D therapeutic use, Vitamins blood, Vitamins therapeutic use, Familial Hypophosphatemic Rickets genetics, Hypercalciuria genetics, Phosphates blood, Sodium-Phosphate Cotransporter Proteins, Type IIc genetics
- Abstract
Hereditary hypophosphatemic rickets with hypercalciuria (HHRH; OMIM: 241530) is a rare autosomal recessive disorder with an estimated prevalence of 1:250,000 that was originally described by Tieder et al. Individuals with HHRH carry compound-heterozygous or homozygous (comp/hom) loss-of-function mutations in the sodium-phosphate co-transporter NPT2c. These mutations result in the development of urinary phosphate (Pi) wasting and hypophosphatemic rickets, bowing, and short stature, as well as appropriately elevated 1,25(OH)
2 D levels, which sets this fibroblast growth factor 23 (FGF23)-independent disorder apart from the more common X-linked hypophosphatemia. The elevated 1,25(OH)2 D levels in turn result in hypercalciuria due to enhanced intestinal calcium absorption and reduced parathyroid hormone (PTH)-dependent calcium-reabsorption in the distal renal tubules, leading to the development of kidney stones and/or nephrocalcinosis in approximately half of the individuals with HHRH. Even heterozygous NPT2c mutations are frequently associated with isolated hypercalciuria (IH), which increases the risk of kidney stones or nephrocalcinosis threefold in affected individuals compared with the general population. Bone disease is generally absent in individuals with IH, in contrast to those with HHRH. Treatment of HHRH and IH consists of monotherapy with oral Pi supplements, while active vitamin D analogs are contraindicated, mainly because the endogenous 1,25(OH)2 D levels are already elevated but also to prevent further worsening of the hypercalciuria. Long-term studies to determine whether oral Pi supplementation alone is sufficient to prevent renal calcifications and bone loss, however, are lacking. It is also unknown how therapy should be monitored, whether secondary hyperparathyroidism can develop, and whether Pi requirements decrease with age, as observed in some FGF23-dependent hypophosphatemic disorders, or whether this can lead to osteoporosis.- Published
- 2019
- Full Text
- View/download PDF
132. FGF23 blockade coming to clinical practice.
- Author
-
Emma F and Haffner D
- Subjects
- Antibodies, Monoclonal, Humanized, Familial Hypophosphatemic Rickets etiology, Fibroblast Growth Factor-23, Fibroblast Growth Factors physiology, Humans, Antibodies, Monoclonal therapeutic use, Familial Hypophosphatemic Rickets drug therapy, Fibroblast Growth Factors antagonists & inhibitors
- Published
- 2018
- Full Text
- View/download PDF
133. Hereditary 1,25-dihydroxyvitamin D-resistant rickets (HVDRR): clinical heterogeneity and long-term efficacious management of eight patients from four unrelated Arab families with a loss of function VDR mutation.
- Author
-
Faiyaz-Ul-Haque M, AlDhalaan W, AlAshwal A, Bin-Abbas BS, AlSagheir A, Alotaiby M, Rafiq Z, and Zaidi SHE
- Subjects
- Child, Child, Preschool, Disease Management, Familial Hypophosphatemic Rickets drug therapy, Familial Hypophosphatemic Rickets pathology, Female, Follow-Up Studies, Humans, Infant, Male, Pedigree, Prognosis, Arabs genetics, Bone Density Conservation Agents therapeutic use, Calcitriol therapeutic use, Drug Resistance, Familial Hypophosphatemic Rickets genetics, Mutation, Receptors, Calcitriol genetics
- Abstract
Background: Vitamin D regulates the concentrations of calcium and phosphate in blood and promotes the growth and remodeling of bones. The circulating active form of vitamin D, 1,25-dihydroxyvitamin D, binds to the vitamin D receptor (VDR), which heterodimerizes with the retinoid X receptor to regulate the expression of target genes. Inactivating mutations in the VDR gene cause hereditary vitamin D-resistant rickets (HVDRR), a rare disorder characterized by an early onset of rickets, growth retardation, skeletal deformities, hypocalcemia, hypophosphatemia and secondary hyperparathyroidism, and in some cases alopecia., Methods: We describe eight new HVDRR patients from four unrelated consanguineous families. The VDR gene was sequenced to identify mutations. The management of patients over a period of up to 11 years following the initial diagnosis is assessed., Results: Although all patients exhibit main features of HVDRR and carry the same c.885C>A (p.Y295*) loss of function mutation in the VDR gene, there was heterogeneity of the manifestations of HVDRR-associated phenotypes and developmental milestones. These eight patients were successfully treated over a period of 11 years. All clinical symptoms were improved except alopecia., Conclusions: The study concludes that VDR sequencing and laboratory tests are essential to confirm HVDRR and to assess the effectiveness of the treatment.
- Published
- 2018
- Full Text
- View/download PDF
134. 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.
- Author
-
Insogna KL, Briot K, Imel EA, Kamenický P, Ruppe MD, Portale AA, Weber T, Pitukcheewanont P, Cheong HI, Jan de Beur S, Imanishi Y, Ito N, Lachmann RH, Tanaka H, Perwad F, Zhang L, Chen CY, Theodore-Oklota C, Mealiffe M, San Martin J, and Carpenter TO
- Subjects
- Adult, Antibodies, Monoclonal adverse effects, Antibodies, Monoclonal pharmacology, Antibodies, Monoclonal, Humanized, Biomarkers metabolism, Bone Remodeling drug effects, Calcium metabolism, Double-Blind Method, Familial Hypophosphatemic Rickets physiopathology, Female, Fibroblast Growth Factor-23, Homeostasis, Humans, Male, Placebos, Treatment Outcome, Antibodies, Monoclonal therapeutic use, Familial Hypophosphatemic Rickets drug therapy, Fibroblast Growth Factors antagonists & inhibitors, Fibroblast Growth Factors immunology
- Abstract
In X-linked hypophosphatemia (XLH), inherited loss-of-function mutations in the PHEX gene cause excess circulating levels of fibroblast growth factor 23 (FGF23), leading to lifelong renal phosphate wasting and hypophosphatemia. Adults with XLH present with chronic musculoskeletal pain and stiffness, short stature, lower limb deformities, fractures, and pseudofractures due to osteomalacia, accelerated osteoarthritis, dental abscesses, and enthesopathy. Burosumab, a fully human monoclonal antibody, binds and inhibits FGF23 to correct hypophosphatemia. This report summarizes results from a double-blind, placebo-controlled, phase 3 trial of burosumab in symptomatic adults with XLH. Participants with hypophosphatemia and pain were assigned 1:1 to burosumab 1 mg/kg (n = 68) or placebo (n = 66) subcutaneously every 4 weeks (Q4W) and were comparable at baseline. Across midpoints of dosing intervals, 94.1% of burosumab-treated participants attained mean serum phosphate concentration above the lower limit of normal compared with 7.6% of those receiving placebo (p < 0.001). Burosumab significantly reduced the Western Ontario and the McMaster Universities Osteoarthritis Index (WOMAC) stiffness subscale compared with placebo (least squares [LS] mean ± standard error [SE] difference, -8.1 ± 3.24; p = 0.012). Reductions in WOMAC physical function subscale (-4.9 ± 2.48; p = 0.048) and Brief Pain Inventory worst pain (-0.5 ± 0.28; p = 0.092) did not achieve statistical significance after Hochberg multiplicity adjustment. At week 24, 43.1% (burosumab) and 7.7% (placebo) of baseline active fractures were fully healed; the odds of healed fracture in the burosumab group was 16.8-fold greater than that in the placebo group (p < 0.001). Biochemical markers of bone formation and resorption increased significantly from baseline with burosumab treatment compared with placebo. The safety profile of burosumab was similar to placebo. There were no treatment-related serious adverse events or meaningful changes from baseline in serum or urine calcium, intact parathyroid hormone, or nephrocalcinosis. These data support the conclusion that burosumab is a novel therapeutic addressing an important medical need in adults with XLH.© 2018 The Authors. Journal of Bone and Mineral Research Published by Wiley Periodicals, Inc., (© 2018 The Authors. Journal of Bone and Mineral Research Published by Wiley Periodicals, Inc.)
- Published
- 2018
- Full Text
- View/download PDF
135. Eldecalcitol Causes FGF23 Resistance for Pi Reabsorption and Improves Rachitic Bone Phenotypes in the Male Hyp Mouse.
- Author
-
Kaneko I, Segawa H, Ikuta K, Hanazaki A, Fujii T, Tatsumi S, Kido S, Hasegawa T, Amizuka N, Saito H, and Miyamoto KI
- Subjects
- Animals, Body Weight drug effects, Bone Density drug effects, Disease Models, Animal, Familial Hypophosphatemic Rickets blood, Familial Hypophosphatemic Rickets metabolism, Fibroblast Growth Factor-23, Fibroblast Growth Factors blood, Male, Mice, Phosphates blood, Vitamin D blood, Vitamin D metabolism, Vitamin D pharmacology, Vitamin D therapeutic use, Bone Density Conservation Agents pharmacology, Bone Density Conservation Agents therapeutic use, Familial Hypophosphatemic Rickets drug therapy, Fibroblast Growth Factors metabolism, Phosphates metabolism, Vitamin D analogs & derivatives
- Abstract
X-linked hypophosphatemia (XLH), the most common form of inheritable rickets, is caused by inactivation of phosphate-regulating gene with homologies to endopeptidases on the X chromosome (PHEX) and leads to fibroblast growth factor (FGF) 23-dependent renal inorganic phosphate (Pi) wasting. In the present study, we investigated whether maintaining Pi homeostasis with a potent vitamin D3 analog, eldecalcitol [1α,25-dihydroxy-2β-(3-hydroxypropyloxy) vitamin D3; ED71], could improve hypophosphatemic rickets in a murine model of XLH, the Hyp mouse. Vehicle, ED71, or 1,25-dihydroxyvitamin D was subcutaneously injected five times weekly in wild-type (WT) and Hyp mice for 4 weeks, from 4 to 8 weeks of age. Injection of ED71 into WT mice suppressed the synthesis of renal 1,25-dihydroxyvitamin D and promoted phosphaturic activity. In contrast, administration of ED71 to Hyp mice completely restored renal Pi transport and NaPi-2a protein levels, although the plasma-intact FGF23 levels were further increased. In addition, ED71 markedly increased the levels of the scaffold proteins, renal sodium-hydrogen exchanger regulatory factor 1, and ezrin in the Hyp mouse kidney. Treatment with ED71 increased the body weight and improved hypophosphatemia, the bone volume/total volume, bone mineral content, and growth plate structure in Hyp mice. Thus, ED71 causes FGF23 resistance for phosphate reabsorption and improves rachitic bone phenotypes in Hyp mice. In conclusion, ED71 has opposite effects on phosphate homeostasis in WT and Hyp mice. Analysis of Hyp mice treated with ED71 could result in an additional model for elucidating PHEX abnormalities.
- Published
- 2018
- Full Text
- View/download PDF
136. Three-Month Randomized Clinical Trial of Nasal Calcitonin in Adults with X-linked Hypophosphatemia.
- Author
-
Sullivan R, Abraham A, Simpson C, Olear E, Carpenter T, Deng Y, Chen C, and Insogna KL
- Subjects
- Adult, Calcitonin administration & dosage, Female, Fibroblast Growth Factor-23, Fibroblast Growth Factors metabolism, Humans, Male, Middle Aged, Phosphates metabolism, Phosphorus pharmacology, Calcitonin therapeutic use, Familial Hypophosphatemic Rickets drug therapy, Fibroblast Growth Factors drug effects, Treatment Outcome
- Abstract
Previous work has demonstrated that a single subcutaneous dose of salmon calcitonin leads to a transient decline in circulating levels of FGF23 in patients with X-linked hypophosphatemia (XLH). Since the calcitonin receptor is expressed on osteocytes, this raises the possibility that interdicting signals through that receptor could modulate circulating levels of FGF23 in XLH. In the present study, 21 subjects with XLH were randomly assigned to receive either placebo nasal spray or 400 IU of nasal salmon calcitonin daily for three months. On the first and last day of the study, serial measurements of FGF23, 1,25-dihydroxyvitamin D, and TmP/GFR were made over 27 h. At the beginning of Visit 2 (the first day of month 2) and the beginning of Visit 3 (the first day of month 3), single, first-morning, fasting measurements of these same parameters were made before the next administered dose of study drug. Following the initial or final dose of study drug, there were no differences in area under the curve, based on treatment assignment, for the three principal outcome variables. Similarly, there were no differences in the fasting measures taken at the beginning of Visit 2 or Visit 3 compared to the fasting values on either day 2 of Visit 1 or the fasting values on day 2 of Visit 4. There were also no significant changes over time in serum phosphorus, serum calcium, circulating levels of PTH, CTx, or P1NP. The reasons why nasal salmon calcitonin did not recapitulate the findings with subcutaneously administered drug may relate to the kinetics of drug delivery, the bioavailability of drug or peak drug dose achieved. It remains possible, however, that other means of altering calcitonin receptor signaling may still provide an opportunity for regulating FGF23 production.
- Published
- 2018
- Full Text
- View/download PDF
137. X-Linked Hypophosphatemia and FGF23-Related Hypophosphatemic Diseases: Prospect for New Treatment.
- Author
-
Kinoshita Y and Fukumoto S
- Subjects
- Antibodies, Monoclonal, Humanized, Familial Hypophosphatemic Rickets drug therapy, Female, Fibroblast Growth Factor-23, Fibroblast Growth Factors immunology, Humans, Hypophosphatemia drug therapy, Osteomalacia drug therapy, Paraneoplastic Syndromes drug therapy, Antibodies, Monoclonal, Familial Hypophosphatemic Rickets metabolism, Fibroblast Growth Factors metabolism, Hypophosphatemia metabolism, Osteomalacia metabolism, Paraneoplastic Syndromes metabolism
- Abstract
Phosphate plays essential roles in many biological processes, and the serum phosphate level is tightly controlled. Chronic hypophosphatemia causes impaired mineralization of the bone matrix and results in rickets and osteomalacia. Fibroblast growth factor 23 (FGF23) is a bone-derived hormone that regulates phosphate metabolism. FGF23 excess induces hypophosphatemia via impaired phosphate reabsorption in the renal proximal tubules and decreased phosphate absorption in the intestines. There are several types of genetic and acquired FGF23-related hypophosphatemic diseases. Among these diseases, X-linked hypophosphatemia (XLH), which is caused by inactivating mutations in the phosphate-regulating endopeptidase homolog, X-linked (PHEX) gene, is the most prevalent form of genetic FGF23-related hypophosphatemic rickets. Another clinically relevant form of FGF23-related hypophosphatemic disease is tumor-induced osteomalacia (TIO), a paraneoplastic syndrome associated with FGF23-producing tumors. A combination of active vitamin D and phosphate salts is the current medical therapy used to treat patients with XLH and inoperative TIO. However, this therapy has certain efficacy- and safety-associated limitations. Several measures to inhibit FGF23 activity have been considered as possible new treatments for FGF23-related hypophosphatemic diseases. In particular, a humanized monoclonal antibody for FGF23 (burosumab) is a promising treatment in patients with XLH and TIO. This review will focus on the phosphate metabolism and the pathogenesis and treatment of FGF23-related hypophosphatemic diseases.
- Published
- 2018
- Full Text
- View/download PDF
138. Burosumab Therapy in Children with X-Linked Hypophosphatemia.
- Author
-
Carpenter TO, Whyte MP, Imel EA, Boot AM, Högler W, Linglart A, Padidela R, Van't Hoff W, Mao M, Chen CY, Skrinar A, Kakkis E, San Martin J, and Portale AA
- Subjects
- Alkaline Phosphatase blood, Antibodies, Monoclonal, Humanized, Child, Child, Preschool, Familial Hypophosphatemic Rickets metabolism, Familial Hypophosphatemic Rickets physiopathology, Female, Fibroblast Growth Factor-23, Genetic Diseases, X-Linked metabolism, Genetic Diseases, X-Linked physiopathology, Growth drug effects, Humans, Kidney Tubules metabolism, Knee Joint diagnostic imaging, Male, Pain Management, Phosphorus blood, Radiography, Severity of Illness Index, Antibodies, Monoclonal therapeutic use, Familial Hypophosphatemic Rickets drug therapy, Fibroblast Growth Factors antagonists & inhibitors, Genetic Diseases, X-Linked drug therapy
- Abstract
Background: X-linked hypophosphatemia is characterized by increased secretion of fibroblast growth factor 23 (FGF-23), which leads to hypophosphatemia and consequently rickets, osteomalacia, and skeletal deformities. We investigated burosumab, a monoclonal antibody that targets FGF-23, in patients with X-linked hypophosphatemia., Methods: In an open-label, phase 2 trial, we randomly assigned 52 children with X-linked hypophosphatemia, in a 1:1 ratio, to receive subcutaneous burosumab either every 2 weeks or every 4 weeks; the dose was adjusted to achieve a serum phosphorus level at the low end of the normal range. The primary end point was the change from baseline to weeks 40 and 64 in the Thacher rickets severity total score (ranging from 0 to 10, with higher scores indicating greater disease severity). In addition, the Radiographic Global Impression of Change was used to evaluate rachitic changes from baseline to week 40 and to week 64. Additional end points were changes in pharmacodynamic markers, linear growth, physical ability, and patient-reported outcomes and the incidence of adverse events., Results: The mean Thacher rickets severity total score decreased from 1.9 at baseline to 0.8 at week 40 with every-2-week dosing and from 1.7 at baseline to 1.1 at week 40 with every-4-week dosing (P<0.001 for both comparisons); these improvements persisted at week 64. The mean serum phosphorus level increased after the first dose in both groups, and more than half the patients in both groups had levels within the normal range (3.2 to 6.1 mg per deciliter [1.0 to 2.0 mmol per liter]) by week 6. Stable serum phosphorus levels were maintained through week 64 with every-2-week dosing. Renal tubular phosphate reabsorption increased from baseline in both groups, with an overall mean increase of 0.98 mg per deciliter (0.32 mmol per liter). The mean dose of burosumab at week 40 was 0.98 mg per kilogram of body weight with every-2-week dosing and 1.50 mg per kilogram with every-4-week dosing. Across both groups, the mean serum alkaline phosphatase level decreased from 459 U per liter at baseline to 369 U per liter at week 64. The mean standing-height z score increased in both groups, with greater improvement seen at all time points with every-2-week dosing (an increase from baseline of 0.19 at week 64) than with every-4-week dosing (an increase from baseline of 0.12 at week 64). Physical ability improved and pain decreased. Nearly all the adverse events were mild or moderate in severity., Conclusions: In children with X-linked hypophosphatemia, treatment with burosumab improved renal tubular phosphate reabsorption, serum phosphorus levels, linear growth, and physical function and reduced pain and the severity of rickets. (Funded by Ultragenyx Pharmaceutical and Kyowa Hakko Kirin; ClinicalTrials.gov number, NCT02163577 ; EudraCT number, 2014-000406-35 ).
- Published
- 2018
- Full Text
- View/download PDF
139. Rickets presenting as gross motor delay in twin girls.
- Author
-
Prasad C and Cummings E
- Subjects
- Child Development, Diseases in Twins diet therapy, Familial Hypophosphatemic Rickets drug therapy, Female, Humans, Infant, Motor Skills Disorders drug therapy, Diseases in Twins genetics, Familial Hypophosphatemic Rickets genetics, Motor Skills Disorders genetics
- Abstract
Competing Interests: Competing interests: None declared.
- Published
- 2018
- Full Text
- View/download PDF
140. Burosumab: First Global Approval.
- Author
-
Lamb YN
- Subjects
- Antibodies, Monoclonal administration & dosage, Antibodies, Monoclonal adverse effects, Antibodies, Monoclonal pharmacokinetics, Antibodies, Monoclonal, Humanized, Drug Approval, Europe, Fibroblast Growth Factor-23, Fibroblast Growth Factors immunology, Humans, Neoplasms, Connective Tissue drug therapy, Nevus drug therapy, Osteomalacia, Paraneoplastic Syndromes, United States, United States Food and Drug Administration, Antibodies, Monoclonal therapeutic use, Familial Hypophosphatemic Rickets drug therapy, Fibroblast Growth Factors antagonists & inhibitors
- Abstract
Burosumab (Crysvita
® ; Kyowa Hakko Kirin Co., Ltd. and Ultragenyx Pharmaceutical Inc.) is a fully human monoclonal antibody directed at fibroblast growth factor 23 (FGF23). Excessive FGF23 production has been implicated in various hypophosphataemic diseases. Inhibition of FGF23 by burosumab results in increased renal phosphate reabsorption and increased serum levels of phosphorus and active vitamin D. In February 2018, the EMA granted subcutaneous burosumab conditional marketing authorization for the treatment of X-linked hypophosphataemia (XLH) with radiographic evidence of bone disease in children one year of age and older and adolescents with growing skeletons. In April 2018, the US FDA approved burosumab for the treatment of XLH in adults and children one year of age and older. Multinational phase III trials of burosumab are currently underway in adult and paediatric patients with XLH. Burosumab is also being evaluated in the phase II setting in adults with tumour-induced osteomalacia and epidermal nevus syndrome in the USA, as well as in Japan and Korea. This article summarizes the milestones in the development of burosumab leading to its first global approval in the EU for XLH in paediatric patients.- Published
- 2018
- Full Text
- View/download PDF
141. Effects of growth hormone treatment on adult height in severely short children with X-linked hypophosphatemic rickets.
- Author
-
Meyerhoff N, Haffner D, Staude H, Wühl E, Marx M, Beetz R, Querfeld U, Holder M, Billing H, Rabl W, Schröder C, Hiort O, Brämswig JH, Richter-Unruh A, Schnabel D, and Živičnjak M
- Subjects
- Adult, Anthropometry methods, Child, Child, Preschool, Dwarfism etiology, Familial Hypophosphatemic Rickets physiopathology, Female, Follow-Up Studies, Human Growth Hormone adverse effects, Humans, Male, Prospective Studies, Treatment Outcome, Body Height drug effects, Dwarfism drug therapy, Familial Hypophosphatemic Rickets drug therapy, Human Growth Hormone therapeutic use
- Abstract
Background: We recently showed that a 3-year growth hormone (GH) treatment improves linear growth in severely short children with X-linked hypophosphatemic rickets (XLH). It is unknown if GH therapy increases adult height in XLH patients., Methods: We carried out a follow-up analysis of a randomized controlled open-label GH study in short prepubertal children with XLH on phosphate and active vitamin D treatment. The changes in SD scores (SDS) of height, sitting height, leg and arm length, and sitting height index (i.e., the ratio between sitting height and height) were analyzed in 11 out of 16 patients followed-up until adult height., Results: At baseline, XLH patients showed disproportionately short stature with reduced standardized height (-3.2 ± 0.6), sitting height (-1.7 ± 0.6), leg (-3.7 ± 0.7) and arm (-2.5 ± 0.8) length, and markedly elevated sitting height index (3.3 ± 0.6; each p < 0.01 versus healthy children). In GH-treated patients, adult height, sitting height, leg length, and arm length exceeded baseline values by 0.7 SDS, 1.7 SDS, 0.7 SDS, and 1.2 SDS respectively, although this was only significant for sitting height. In controls, no significant changes in linear body dimensions were noted. Adult height did not statistically differ between groups (-2.4 ± 0.7 vs -3.3 ± 1.2, p = 0.082). GH did not exaggerate body disproportion., Conclusions: Growth hormone treatment did not significantly increase adult height in this group of short children with XLH, which may be at least partly due to the small number of patients included in our study.
- Published
- 2018
- Full Text
- View/download PDF
142. Hormonal Regulation of Osteocyte Perilacunar and Canalicular Remodeling in the Hyp Mouse Model of X-Linked Hypophosphatemia.
- Author
-
Tokarz D, Martins JS, Petit ET, Lin CP, Demay MB, and Liu ES
- Subjects
- Animals, Antibodies pharmacology, Antibodies therapeutic use, Biomarkers metabolism, Bone Remodeling, Calcitriol pharmacology, Calcitriol therapeutic use, Cortical Bone drug effects, Cortical Bone pathology, Disease Models, Animal, Fibroblast Growth Factor-23, Fibroblast Growth Factors immunology, Mice, Inbred C57BL, Osteocytes drug effects, Skull drug effects, Skull pathology, Tibia drug effects, Tibia pathology, Familial Hypophosphatemic Rickets drug therapy, Familial Hypophosphatemic Rickets physiopathology, Hormones therapeutic use, Osteocytes metabolism
- Abstract
Osteocytes remodel their surrounding perilacunar matrix and canalicular network to maintain skeletal homeostasis. Perilacunar/canalicular remodeling is also thought to play a role in determining bone quality. X-linked hypophosphatemia (XLH) is characterized by elevated serum fibroblast growth factor 23 (FGF23) levels, resulting in hypophosphatemia and decreased production of 1,25 dihydroxyvitamin D (1,25D). In addition to rickets and osteomalacia, long bones from mice with XLH (Hyp) have impaired whole-bone biomechanical integrity accompanied by increased osteocyte apoptosis. To address whether perilacunar/canalicular remodeling is altered in Hyp mice, histomorphometric analyses of tibia and 3D intravital microscopic analyses of calvaria were performed. These studies demonstrate that Hyp mice have larger osteocyte lacunae in both the tibia and calvaria, accompanied by enhanced osteocyte mRNA and protein expression of matrix metalloproteinase 13 (MMP13) and genes classically used by osteoclasts to resorb bone, such as cathepsin K (CTSK). Hyp mice also exhibit impaired canalicular organization, with a decrease in number and branching of canaliculi extending from tibial and calvarial lacunae. To determine whether improving mineral ion and hormone homeostasis attenuates the lacunocanalicular phenotype, Hyp mice were treated with 1,25D or FGF23 blocking antibody (FGF23Ab). Both therapies were shown to decrease osteocyte lacunar size and to improve canalicular organization in tibia and calvaria. 1,25D treatment of Hyp mice normalizes osteocyte expression of MMP13 and classic osteoclast markers, while FGF23Ab decreases expression of MMP13 and selected osteoclast markers. Taken together, these studies point to regulation of perilacunar/canalicular remodeling by physiologic stimuli including hypophosphatemia and 1,25D. © 2017 American Society for Bone and Mineral Research., (© 2017 American Society for Bone and Mineral Research.)
- Published
- 2018
- Full Text
- View/download PDF
143. Impact of Conventional Medical Therapy on Bone Mineral Density and Bone Turnover in Adult Patients with X-Linked Hypophosphatemia: A 6-Year Prospective Cohort Study.
- Author
-
Shanbhogue VV, Hansen S, Jørgensen NR, and Beck-Nielsen SS
- Subjects
- Adolescent, Adult, Aged, Female, Fibroblast Growth Factor-23, Fibroblast Growth Factors metabolism, Humans, Lumbar Vertebrae drug effects, Male, Middle Aged, Prospective Studies, Young Adult, Bone Density drug effects, Bone Density Conservation Agents therapeutic use, Bone Remodeling drug effects, Familial Hypophosphatemic Rickets drug therapy
- Abstract
X-linked hypophosphatemia (XLH) is a rare, inheritable disorder manifesting as rickets in children and osteomalacia in adults. While conventional medical treatment with oral phosphate and alfacalcidol is recommended in childhood, it is undecided whether adults should continue therapy. The aim of this 6-year prospective study was to determine the impact of conventional medical treatment on areal bone mineral density (aBMD), bone turnover markers (BTMs) and measures of calcium homeostasis in 27 adult patients with XLH, 11 of whom received medical treatment. Lumbar spine and total hip aBMD, as assessed by DXA, and biochemical measures of calcium, phosphate, PTH, 1,25 dihydroxyvitamin D
2+3 (1,25(OH)2 D), fibroblast growth factor 23 (FGF23), P1NP and CTX were measured at baseline and at follow-up. The renal tubular reabsorption of PO4 (TmPO4 /GFR) was calculated at both time points. Multilevel mixed-effects linear regression models were used for analyses. During the study period, spine and hip aBMD did not change significantly between treated and non-treated XLH patients. There was a trend towards a decrease in calcium, phosphate and TmPO4 /GFR in the treatment group (p = 0.057, p = 0.080 and p = 0.063, respectively), whereas PTH, FGF23, 1,25(OH)2 D and P1NP did not change significantly in either groups. However, CTX increased significantly in the treated compared to non-treated group (p = 0.044). Continuing conventional medical therapy in adulthood, although associated with increased bone resorption, does not promote or prevent loss of bone mass as evidenced from the stable aBMD of the hip and spine in XLH patients.- Published
- 2018
- Full Text
- View/download PDF
144. Craniofacial and dental characteristics of patients with vitamin-D-dependent rickets type 1A compared to controls and patients with X-linked hypophosphatemia.
- Author
-
Gjørup H, Beck-Nielsen SS, and Haubek D
- Subjects
- Bone Density Conservation Agents therapeutic use, Case-Control Studies, Cephalometry, Denmark, Familial Hypophosphatemic Rickets drug therapy, Female, Humans, Hydroxycholecalciferols therapeutic use, Infant, Male, Phenotype, Registries, Familial Hypophosphatemic Rickets pathology, Skull abnormalities, Tooth Abnormalities pathology
- Abstract
ᅟOBJECTIVES: Vitamin-D-dependent rickets type 1A (VDDR1A) is a rare inherited disease caused by defective activation of vitamin D. The aim of the study was to describe the craniofacial characteristics and the dental phenotype of patients with genetically confirmed VDDR1A. The VDDR1A findings were compared to findings in patients with X-linked hypophosphatemia (XLH) and healthy controls., Material and Methods: Ten patients with VDDR1A were identified. The reference group for the comparison of cephalometric findings was 49 adults without chronic disease. The reference group for the comparison of dental findings was 30 adults with XLH. Clinical examination, clinical photos, and radiographs were obtained. Cephalometric analysis was performed. Photos and radiographs were visually evaluated., Results: The depth of the posterior cranial fossa (d-p and d-s-iop) in VDDR1A adults was reduced compared to the reference group (p < 0.05). Five (83%) of six adults with VDDR1A and one (4%) of 25 adults with XLH had enamel hypoplasia on several incisors and/or canines (p < 0.001). Three (75%) of four adults with VDDR1A and none of 16 adults with XLH had several first molars with enamel hypoplasia (p = 0.004). Five of 7 (71%) adults with VDDR1A and 24 of 30 (80%) adults with XLH had endodontically affected teeth., Conclusions: The dental aberration of VDDR1A is more in line with the dental aberration of nutritional rickets than with the dental aberrations in XLH, suggesting the combination of low availability of both calcium and phosphate to be critical in periods of enamel formation., Clinical Relevance: Knowledge on craniofacial and dental aberration in patients with rare diseases, e.g., inherited rickets, is of importance to the dental practitioner, especially during diagnostics and treatment in special care units.
- Published
- 2018
- Full Text
- View/download PDF
145. Tooth Development Associated with Mutations in Hereditary Vitamin D-Resistant Rickets.
- Author
-
Hanna AE, Sanjad S, Andary R, Nemer G, and Ghafari JG
- Subjects
- Alopecia etiology, Calcium therapeutic use, Child, Child, Preschool, DNA Mutational Analysis, Familial Hypophosphatemic Rickets complications, Familial Hypophosphatemic Rickets drug therapy, Familial Hypophosphatemic Rickets physiopathology, Female, Humans, Hypocalcemia drug therapy, Male, Odontogenesis physiology, Pedigree, Phenotype, Vitamin D therapeutic use, Vitamins therapeutic use, Familial Hypophosphatemic Rickets genetics, Mutation, Odontogenesis genetics, Receptors, Calcitriol genetics
- Abstract
Hereditary vitamin D-resistant rickets (HVDRR) is a rare genetic disorder caused by mutations at the level of the vitamin D receptor ( VDR) gene. The disease is characterized by refractory hypocalcemia, elevated serum levels of 1,25-dihydroxy-vitamin D, retarded growth, sparse body hair (sometimes alopecia), premature tooth loss, enlarged pulp chambers, thin dentine, and hypoplastic enamel. The aims of this study were 1) to document the dental development of children with HVDRR in association with the mutation type within the VDR and 2) to evaluate the association between dental development and the timing of and response to HVDRR treatment. Genome analysis was performed for 4 affected children (2 y 2 mo to 6 y 8 mo) under treatment with high doses of vitamin D and calcium. Longitudinal records of clinical and radiographic data on their dental development were assessed in relation to genetic profile and response to treatment. Treatment success depended on the position of the mutation within the VDR protein: children with the p.R391S mutation had a favorable outcome but maintained alopecia totalis, while 1 child with the p.H397P mutation and normal hair had no response to very high doses of vitamin D. The primary incisors, formed prenatally and first to emerge, were missing in 3 children and mobile in 1 child; parents reported loss within months posteruption. Posterior teeth were present, having erupted after treatment initiation. Hypoplastic enamel in emerging teeth was associated with late treatment onset. Mutation type in the VDR gene appears to be related to differences in the disease phenotype and response to treatment. Dental development represents an indicator of the disease process, initially protected by maternal blood levels of calcium and later restored by therapeutic supplies that normalize these levels. Knowledge Transfer Statement: Two novel mutations were associated with different HVDRR phenotypes, one of which responded positively to treatment. Early detection of the mutation should help pediatricians forecast treatment protocol and response. The results also highlight the direct relationship between dental development and blood calcium levels, underscoring the importance of early diagnosis and treatment of HVDRR to minimize the loss of primary teeth and reduce structural abnormalities of permanent teeth.
- Published
- 2018
- Full Text
- View/download PDF
146. Spontaneous Growth and Effect of Early Therapy with Calcitriol and Phosphate in X-linked Hypophosphatemic Rickets.
- Author
-
Cagnoli M, Richter R, Böhm P, Knye K, Empting S, and Mohnike K
- Subjects
- Adolescent, Adult, Body Height, Child, Child, Preschool, Cross-Sectional Studies, Humans, Phosphates, Secondary Prevention, Sexual Maturation, Calcitriol therapeutic use, Familial Hypophosphatemic Rickets drug therapy
- Abstract
Whereas nutritional vitamin D deficient rickets affects many people world-wide, X-linked hypophosphatemic rickets (XLH, MIM 307800) has a prevalence of only 1:25.000. Like other rare diseases burden of disease in XLH and the effect of the current standard of care are inadequately described. Only few height data of untreated patients with XLH have been published. Here we report on height before start of therapy of 127 patients with XLH from 49 centres. One investigator collected all data from patient files documented at regular visits by treating physicians. Height standard deviation score (HSDS) was calculated and the geometrical mean was analysed. At birth all patients had a documented height within the healthy reference population. In this cross-sectional analysis of documented height at time of diagnosis decelerates until a mean age of 4.3 years to a nadir, i.e. lowest HSDS of -3.2 HSDS. Afterwards a spontaneous catch-up growth of +1.3 HSDS occurs until start of puberty. To assess the impact of calcitriol and phosphate supplementation on growth we analysed from a cohort of 18 patients treated at the Dept. of Paediatrics at O.-v.-Guericke-University Magdeburg. In this subgroup, size at birth and all time lowest HSDS (r=0.56 p=0.002) are correlated as well as all time low HSDS and last height during puberty (r=0.62 p=0.001). 10 of 18 patients were treated before age 18 months. Within this group the mean HSDS decelerates to -2.2 SDS at age 4.4 y. and increased to -1.4 SDS at age 9.9 years. Adult height, i.e. mean age 17.6 years was -2.4 HSDS. In conclusion, untreated children with XLR are characterized by normal length at birth, diminished growth rate compared to reference children until 4.3 years and spontaneous catch-up growth of 1.3 HSDS until start of puberty. Improved growth rate in XLR children occured by combined phosphate and calcitriol treatment before 18 months., (Copyright© of YS Medical Media ltd.)
- Published
- 2017
- Full Text
- View/download PDF
147. Therapeutic Effects of FGF23 c-tail Fc in a Murine Preclinical Model of X-Linked Hypophosphatemia Via the Selective Modulation of Phosphate Reabsorption.
- Author
-
Johnson K, Levine K, Sergi J, Chamoun J, Roach R, Vekich J, Favis M, Horn M, Cao X, Miller B, Snyder W, Aivazian D, Reagan W, Berryman E, Colangelo J, Markiewicz V, Bagi CM, Brown TP, Coyle A, Mohammadi M, and Magram J
- Subjects
- Animals, Bone Density drug effects, Bone and Bones diagnostic imaging, Bone and Bones drug effects, Bone and Bones pathology, Calcification, Physiologic drug effects, Calcitriol blood, Calcitriol pharmacology, Calcium blood, Cancellous Bone pathology, Disease Models, Animal, Familial Hypophosphatemic Rickets blood, Familial Hypophosphatemic Rickets diagnostic imaging, Fibroblast Growth Factor-23, Fibroblast Growth Factors chemistry, HEK293 Cells, Humans, Mice, Peptides pharmacology, Phosphates blood, Rats, Wistar, Recombinant Proteins pharmacology, Renal Reabsorption drug effects, Familial Hypophosphatemic Rickets drug therapy, Fibroblast Growth Factors therapeutic use
- Abstract
Fibroblast growth factor 23 (FGF23) is the causative factor of X-linked hypophosphatemia (XLH), a genetic disorder effecting 1:20,000 that is characterized by excessive phosphate excretion, elevated FGF23 levels and a rickets/osteomalacia phenotype. FGF23 inhibits phosphate reabsorption and suppresses 1α,25-dihydroxyvitamin D (1,25D) biosynthesis, analytes that differentially contribute to bone integrity and deleterious soft-tissue mineralization. As inhibition of ligand broadly modulates downstream targets, balancing efficacy and unwanted toxicity is difficult when targeting the FGF23 pathway. We demonstrate that a FGF23 c-tail-Fc fusion molecule selectively modulates the phosphate pathway in vivo by competitive antagonism of FGF23 binding to the FGFR/α klotho receptor complex. Repeated injection of FGF23 c-tail Fc in Hyp mice, a preclinical model of XLH, increases cell surface abundance of kidney NaPi transporters, normalizes phosphate excretion, and significantly improves bone architecture in the absence of soft-tissue mineralization. Repeated injection does not modulate either 1,25D or calcium in a physiologically relevant manner in either a wild-type or disease setting. These data suggest that bone integrity can be improved in models of XLH via the exclusive modulation of phosphate. We posit that the selective modulation of the phosphate pathway will increase the window between efficacy and safety risks, allowing increased efficacy to be achieved in the treatment of this chronic disease. © 2017 American Society for Bone and Mineral Research., (© 2017 American Society for Bone and Mineral Research.)
- Published
- 2017
- Full Text
- View/download PDF
148. Two-year recombinant human growth hormone (rhGH) treatment is more effective in pre-pubertal compared to pubertal short children with X-linked hypophosphatemic rickets (XLHR).
- Author
-
Rothenbuhler A, Esterle L, Gueorguieva I, Salles JP, Mignot B, Colle M, and Linglart A
- Subjects
- Child, Child, Preschool, Female, Humans, Male, Prospective Studies, Time Factors, Body Height, Familial Hypophosphatemic Rickets drug therapy, Genetic Diseases, X-Linked drug therapy, Human Growth Hormone therapeutic use, Puberty drug effects
- Abstract
Context: Twenty-five to 40% of patients with well-controlled X-linked hypophosphatemic rickets (XLHR) have a final height under -2 SDS. Previous studies have shown that recombinant human growth hormone (rhGH) treatment improves linear growth in short children with XLHR., Objective: We studied the effectiveness of rhGH treatment in children with XLHR in a larger cohort., Design: Monocentric, prospective, non-randomized trial., Setting: University hospital in France., Patients: 19 patients with XLHR and a mutation in the PHEX gene. Six male and 6 female Tanner stage 1 patients (age 6.1±2.4years) and 4 male and 3 female Tanner stage 2 patients (age 13.1±1years). At inclusion, height SDS was -2.35±0.8 SDS and growth velocity was -1.12±1.2 SDS., Intervention: 2years of treatment with 67mcg/kg/day of rhGH at initiation. Every three months rhGH dosage was adjusted using an IGF-1 dosing protocol., Main Outcome Measures: Comparison in change from baseline to year 2 in height and growth velocity., Results: Height SDS improved from -2.35±0.8 SDS at baseline, to -1.62±0.8 SDS (p=0.01) after one and to -1.2±1 SDS (p=0.04) after two years of rhGH treatment. There was a strong correlation (r
2 =0.6104, p<0.0001) between the age of onset of rhGH treatment and the number of cm gained over the study period. Pre-pubertal patients height SDS improved compared to baseline height SDS after one (-1.5±0.7, p<0.03) and two (-0.96±1, p<0.03) years of rhGH treatment. In pubertal patients there was no significant improvement in height SDS after one year (-1.75±1) and after two years (-1.7±0.8) of rhGH treatment., Conclusion: Two-year rhGH treatment is effective to treat short stature in XLHR children. Pre-pubertal children responded better to rhGH., Clinical Trial Registration Number: NCT02720770., (Copyright © 2017 Elsevier Ltd. All rights reserved.)- Published
- 2017
- Full Text
- View/download PDF
149. Cinacalcet as an alternative to phosphate therapy in X-linked hypophosphataemic rickets.
- Author
-
Alon US, Jarka D, Monachino PJ, Sebestyen VanSickle J, and Srivastava T
- Subjects
- Calcimimetic Agents administration & dosage, Child, Preschool, Cinacalcet administration & dosage, Familial Hypophosphatemic Rickets surgery, Humans, Male, Calcimimetic Agents pharmacology, Cinacalcet pharmacology, Familial Hypophosphatemic Rickets blood, Familial Hypophosphatemic Rickets drug therapy
- Published
- 2017
- Full Text
- View/download PDF
150. The Effectiveness of Cinacalcet as an Adjunctive Therapy for Hereditary 1,25 Dihydroxyvitamin D3-Resistant Rickets.
- Author
-
Akıncı A, Dündar İ, and Kıvılcım M
- Subjects
- Calcimimetic Agents therapeutic use, Child, Preschool, Female, Humans, Hyperparathyroidism, Secondary prevention & control, Hypophosphatemia prevention & control, Infant, Treatment Outcome, Cinacalcet therapeutic use, Familial Hypophosphatemic Rickets drug therapy
- Abstract
High doses of oral calcium or long-term calcium infusions are recommended to correct the hypocalcemia and secondary hyperparathyroidism in patients with hereditary 1,25 dihydroxyvitamin D3-resistant rickets (HVDRR). Preliminary studies revealed that calcimimetics may be a safe and effective therapeutic choice in children with secondary hyperparathyroidism. Our aim was to observe the efficacy of cinacalcet in the normalization of secondary hyperparathyroidism and hypophosphatemia in two siblings aged 2.5 years and 6 months with HVDRR who did not respond to traditional treatment regimes. Both patients were admitted to the hospital with severe hypocalcemia. They were treated with high doses of calcitriol and calcium infusions intravenously. Secondary hyperparathyroidism was normalized temporarily, but did not improve completely. Cinacalcet (0.25 mg/kg) once a day along with the high doses of oral calcium and calcitriol was added to the treatment schedule. After 3 months, biochemical and radiologic findings reverted to normal. Our findings indicate that cinacalcet is effective in normalizing the hyperparathyroidism and hypophosphatemia in these cases and in improving the bone pathology.
- Published
- 2017
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.