Sustained Efficacy and Safety of Burosumab, a Monoclonal Antibody to FGF23, in Children With X-Linked Hypophosphatemia

Agnès Linglart, Erik A Imel, Michael P Whyte, Anthony A Portale, Wolfgang Högler, Annemieke M Boot, Raja Padidela, William Van't Hoff, Gary S Gottesman, Angel Chen, Alison Skrinar, Mary Scott Roberts, Thomas O Carpenter, Agnès Linglart, Erik A Imel, Michael P Whyte, Anthony A Portale, Wolfgang Högler, Annemieke M Boot, Raja Padidela, William Van't Hoff, Gary S Gottesman, Angel Chen, Alison Skrinar, Mary Scott Roberts, Thomas O Carpenter

Abstract

Purpose: In X-linked hypophosphatemia (XLH), excess fibroblast growth factor-23 causes hypophosphatemia and low calcitriol, leading to musculoskeletal disease with clinical consequences. XLH treatment options include conventional oral phosphate with active vitamin D, or monotherapy with burosumab, a monoclonal antibody approved to treat children and adults with XLH. We have previously reported outcomes up to 64 weeks, and here we report safety and efficacy follow-up results up to 160 weeks from an open-label, multicenter, randomized, dose-finding trial of burosumab for 5- to 12-year-old children with XLH.

Methods: After 1 week of conventional therapy washout, patients were randomized 1:1 to burosumab every 2 weeks (Q2W) or every 4 weeks (Q4W) for 64 weeks, with dosing titrated based on fasting serum phosphorus levels between baseline and week 16. From week 66 to week 160, all patients received Q2W burosumab.

Results: Twenty-six children were randomized initially into each Q2W and Q4W group and all completed treatment to week 160. In 41 children with open distal femoral and proximal tibial growth plates (from both treatment groups), total Rickets Severity Score significantly decreased by 0.9 ± 0.1 (least squares mean ± SE; P < 0.0001) from baseline to week 160. Fasting serum phosphorus increases were sustained by burosumab therapy throughout the study, with an overall population mean (SD) of 3.35 (0.39) mg/dL, within the pediatric normal range (3.2-6.1 mg/dL) at week 160 (mean change from baseline P < 0.0001). Most adverse events were mild to moderate in severity.

Main conclusions: In children with XLH, burosumab administration for 160 weeks improved phosphate homeostasis and rickets and was well-tolerated. Long-term safety was consistent with the reported safety profile of burosumab.

Clinicaltrials.gov: NCT02163577.

Keywords: calcitriol; growth; hypophosphatemia; osteomalacia; phosphate; rickets.

© The Author(s) 2021. Published by Oxford University Press on behalf of the Endocrine Society.

Figures

Figure 1.
Figure 1.
UX023-CL201 CONSORT diagram.
Figure 2.
Figure 2.
Tanner stage at baseline and week 160.
Figure 3.
Figure 3.
(A) Sustained improvement in serum phosphorus with burosumab—note that data from week 0 to week 64 were previously reported (13). (B) Sustained improvement in TmP/GFR with burosumab—note that data from week 0 to week 64 were previously reported (13). (C) Sustained improvement in serum 1,25(OH)2D with burosumab—note that data from week 0 to week 64 were previously reported (13).
Figure 4.
Figure 4.
(A) RSS total score decreased from baseline indicating improved rickets. (B) RGI-C global score increased indicating improved rickets. (C) Sustained healing of rickets in an 8-year-old boy. (D) RGI-C lower limb deformity score increased indicating improvement throughout the study. (E) Sustained improvement in serum alkaline phosphatase (ALP) with burosumab—note that data from week 0 to week 64 were previously reported (13).
Figure 5.
Figure 5.
(A) Change in standing height z-score from baseline. (B) Change in percent of predicted 6MWT from baseline (least square mean ± SE). (C) POSNA-PODCI normative score (mean ± SE).
Figure 6.
Figure 6.
(A) No clinically meaningful changes in serum calcium. (B) No clinically meaningful changes in 24-hour urine calcium excretion. (C) No clinically meaningful changes in serum PTH (iPTH).

References

    1. Albright F, Butler AM, Bloomberg E. Rickets resistant to vitamin D therapy. Am J Dis Child. 1937;54(3):529-547.
    1. Francis F, Hennig S, Meitinger T, et al. . A gene (PEX) with homologies to endopeptidases is mutated in patients with X–linked hypophosphatemic rickets. Nat Genet. 1995;11(2):130-136.
    1. Carpenter TO, Imel EA, Holm IA, Jan de Beur SM, Insogna KL. A clinician’s guide to X-linked hypophosphatemia. J Bone Miner Res. 2011;26(7):1381-1388.
    1. Cagnoli M, Richter R, Böhm P, Knye K, Empting S, Mohnike K. Spontaneous growth and effect of early therapy with calcitriol and phosphate in X-linked hypophosphatemic rickets. Pediatr Endocrinol Rev. 2017;15(Suppl 1):119-122.
    1. Linglart A, Dvorak-Ewell M, Marshall A, et al. . Impaired mobility and pain significantly impact the quality of life of children with X-linked hypophosphatemia. Bone Abstracts. 2015;4:198.
    1. McWhorter AG, Seale NS. Prevalence of dental abscess in a population of children with vitamin D-resistant rickets. Pediatr Dent. 1991;13(2):91-96.
    1. Vakharia JD, Matlock K, Taylor HO, Backeljauw PF, Topor LS. Craniosynostosis as the presenting feature of X-linked hypophosphatemic rickets. Pediatrics. 2018;141(Suppl 5):S515-S519.
    1. Glorieux FH, Marie PJ, Pettifor JM, Delvin EE. Bone response to phosphate salts, ergocalciferol, and calcitriol in hypophosphatemic vitamin D-resistant rickets. N Engl J Med. 1980;303(18):1023-1031.
    1. Rafaelsen S, Johansson S, Ræder H, Bjerknes R. Hereditary hypophosphatemia in Norway: a retrospective population-based study of genotypes, phenotypes, and treatment complications. Eur J Endocrinol. 2016;174(2):125-136.
    1. Linglart A, Biosse-Duplan M, Briot K, et al. . Therapeutic management of hypophosphatemic rickets from infancy to adulthood. Endocr Connect. 2014;3(1):R13-R30.
    1. Lamb YN. Burosumab: first global approval. Drugs. 2018;78(6):707-714.
    1. Whyte MP, Carpenter TO, Gottesman GS, et al. . Efficacy and safety of burosumab in children aged 1-4 years with X-linked hypophosphataemia: a multicentre, open-label, phase 2 trial. Lancet Diabetes Endocrinol. 2019;7(3):189-199.
    1. Carpenter TO, Whyte MP, Imel EA, et al. . Burosumab therapy in children with X-linked hypophosphatemia. N Engl J Med. 2018;378(21):1987-1998.
    1. Imel EA, Glorieux FH, Whyte MP, et al. . Burosumab versus conventional therapy in children with X-linked hypophosphataemia: a randomised, active-controlled, open-label, phase 3 trial. Lancet. 2019;393(10189):2416-2427.
    1. Whyte MP, Imel E, Portale AA, et al. . Burosumab, a Fully Human Anti-FGF23 Monoclonal Antibody, for X-Linked Hypophosphatemia (XLH): Results from Two Phase 2 Trials in Affected Children 1-12 Years Old 2018. Chicago, IL: Endocrine Society Annual Meeting (ENDO; ).
    1. Whyte MP, Fujita KP, Moseley S, Thompson DD, McAlister WH. Validation of a novel scoring system for changes in skeletal manifestations of hypophosphatasia in newborns, infants, and children: the radiographic global impression of change scale. J Bone Miner Res. 2018;33(5):868-874.
    1. Geiger R, Strasak A, Treml B, et al. . Six-minute walk test in children and adolescents. J Pediatr. 2007;150(4):395-9, 399.e1.
    1. Daltroy LH, Liang MH, Fossel AH, Goldberg MJ. The POSNA pediatric musculoskeletal functional health questionnaire: report on reliability, validity, and sensitivity to change. Pediatric Outcomes Instrument Development Group. Pediatric Orthopaedic Society of North America. J Pediatr Orthop. 1998;18(5):561-571.
    1. Imel EA, Zhang X, Ruppe MD, et al. . Prolonged correction of serum phosphorus in adults with X-linked hypophosphatemia using monthly doses of KRN23. J Clin Endocrinol Metab. 2015;100(7):2565-2573.
    1. Kruse K, Kracht U, Göpfert G. Renal threshold phosphate concentration (TmPO4/GFR). Arch Dis Child. 1982;57(3):217-223.
    1. Burosumab (CRYSVITA®) prescribing information. Ultragenyx Pharmaceutical Inc. 06/2020. . Accessed October 20, 2021.

Source: PubMed

3
購読する