Patient-Reported Outcomes from a Randomized, Active-Controlled, Open-Label, Phase 3 Trial of Burosumab Versus Conventional Therapy in Children with X-Linked Hypophosphatemia

Raja Padidela, Michael P Whyte, Francis H Glorieux, Craig F Munns, Leanne M Ward, Ola Nilsson, Anthony A Portale, Jill H Simmons, Noriyuki Namba, Hae Il Cheong, Pisit Pitukcheewanont, Etienne Sochett, Wolfgang Högler, Koji Muroya, Hiroyuki Tanaka, Gary S Gottesman, Andrew Biggin, Farzana Perwad, Angela Williams, Annabel Nixon, Wei Sun, Angel Chen, Alison Skrinar, Erik A Imel, Raja Padidela, Michael P Whyte, Francis H Glorieux, Craig F Munns, Leanne M Ward, Ola Nilsson, Anthony A Portale, Jill H Simmons, Noriyuki Namba, Hae Il Cheong, Pisit Pitukcheewanont, Etienne Sochett, Wolfgang Högler, Koji Muroya, Hiroyuki Tanaka, Gary S Gottesman, Andrew Biggin, Farzana Perwad, Angela Williams, Annabel Nixon, Wei Sun, Angel Chen, Alison Skrinar, Erik A Imel

Abstract

Changing to burosumab, a monoclonal antibody targeting fibroblast growth factor 23, significantly improved phosphorus homeostasis, rickets, lower-extremity deformities, mobility, and growth versus continuing oral phosphate and active vitamin D (conventional therapy) in a randomized, open-label, phase 3 trial involving children aged 1-12 years with X-linked hypophosphatemia. Patients were randomized (1:1) to subcutaneous burosumab or to continue conventional therapy. We present patient-reported outcomes (PROs) from this trial for children aged ≥ 5 years at screening (n = 35), using a Patient-Reported Outcomes Measurement Information System (PROMIS) questionnaire and SF-10 Health Survey for Children. PROMIS pain interference, physical function mobility, and fatigue scores improved from baseline with burosumab at weeks 40 and 64, but changed little with continued conventional therapy. Pain interference scores differed significantly between groups at week 40 (- 5.02, 95% CI - 9.29 to - 0.75; p = 0.0212) but not at week 64. Between-group differences were not significant at either week for physical function mobility or fatigue. Reductions in PROMIS pain interference and fatigue scores from baseline were clinically meaningful with burosumab at weeks 40 and 64 but not with conventional therapy. SF-10 physical health scores (PHS-10) improved significantly with burosumab at week 40 (least-squares mean [standard error] + 5.98 [1.79]; p = 0.0008) and week 64 (+ 5.93 [1.88]; p = 0.0016) but not with conventional therapy (between-treatment differences were nonsignificant). In conclusion, changing to burosumab improved PRO measures, with statistically significant differences in PROMIS pain interference at week 40 versus continuing with conventional therapy and in PHS-10 at weeks 40 and 64 versus baseline.Trial registration: ClinicalTrials.gov NCT02915705.

Keywords: Burosumab; Patient-reported outcomes; Patient-reported outcomes measurement information system; X-linked hypophosphatemia.

Conflict of interest statement

The following authors served as clinical investigators for one or more studies, including this trial, sponsored by Ultragenyx Pharmaceutical Inc. in partnership with Kyowa Kirin International plc: RP, MPW, FHG, CFM, LMW, ON, AAP, JHS, NN, HIC, PP, ES, WH, KM, HT, GSG, AB, FP, and EAI. AAP, NN, FP, and EAI have also received honoraria for serving as advisory board members or for lectures from Ultragenyx Pharmaceutical Inc. RP has received personal fees from Ultragenyx Pharmaceutical Inc. and Kyowa Kirin International plc, a research grant, consultation fees, honoraria, and travel grants from Kyowa Kirin International plc and Alexion UK, and non-financial support from Kyowa Kirin International plc. MPW has received research grant support, honoraria, and travel from Alexion Pharmaceutical Inc. FHG has received personal fees from Kyowa Kirin International plc and research funding from Amgen and Mereo BioPharma. LMW has served as a consultant to Ultragenyx, with funds to LMW’s institution. NN has received personal fees and non-financial support from Kyowa Kirin International plc and Ultragenyx Pharmaceutical Inc. during the conduct of the study, and personal fees and non-financial support (honoraria, consulting fees, and travel support) from Alexion UK outside the submitted work. PP has received research funding from Ultragenyx Pharmaceutical Inc. and is currently an employee of Ascendis Pharma Inc. WH has received honoraria, consulting fees, and travel support from Ultragenyx Pharmaceutical Inc. and research funding, honoraria, and travel support from Kyowa Kirin. GSG has received consulting fees from Ultragenyx Pharmaceutical Inc. AW and WS are employees of Kyowa Kirin International plc. AN is an employee of Chilli Consultancy and has received consultancy fees from Kyowa Kirin International plc to support the development of this manuscript. AC and AS are employees and stockholders of Ultragenyx Pharmaceutical Inc.

Figures

Fig. 1
Fig. 1
Baseline PROMIS (a) pain interference, (b) physical function mobility, and (c) fatigue scores for patients aged ≥ 5 years (n = 35). Data show standardized PROMIS T-scores; higher T-scores indicate more pain interference, better function mobility, and worse fatigue. Reference lines at ± 1 SD of the mean are based on a population mean of 50 and SD of 10. PROMIS Patient-Reported Outcomes Measurement Information System, SD standard deviation
Fig. 2
Fig. 2
Change from baseline in PROMIS (a) pain interference, (b) physical function mobility, and (c) fatigue scores for patients aged ≥ 5 years (n = 35). Data are expressed as LS mean (standard error). *p < 0.05 for LS mean change at week 40 (burosumab–conventional therapy). †Indicates the mean change is ≥ 3-point MID from baseline. LS least-squares, MID minimally important difference, PROMIS Patient-Reported Outcomes Measurement Information System
Fig. 3
Fig. 3
Baseline SF-10 Health Survey for Children (a) PHS-10 and (b) PSS-10 for patients aged ≥ 5 years (n = 35). P.25, P.50, and P.75 are the 25th, 50th, and 75th percentiles from the general population. Higher global scores indicate better HRQoL. HRQoL health-related quality of life, PHS-10 physical health score, PSS-10 psychosocial health score, SF-10 Short Form-10
Fig. 4
Fig. 4
Changes from baseline to weeks 40 and 64 for SF-10 Health Survey for Children (a) PHS-10 and (b) PSS-10 for all patients aged ≥ 5 years (n = 35). Data are expressed as LS mean ± standard error. *p < 0.01 for change from baseline to week 64; **p < 0.001 for change from baseline to week 40. LS least-squares, PHS-10 physical health score, PSS-10 psychosocial health score, SF-10 Short Form-10

References

    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;26:1381–1388. doi: 10.1002/jbmr.340.
    1. Whyte MP, Carpenter TO, Gottesman GS, Mao M, Skrinar A, San Martin J, Imel EA. 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;3:189–199. doi: 10.1016/S2213-8587(18)30338-3.
    1. Linglart A, Biosse-Duplan M, Briot K, Chaussain C, Esterle L, Guillaume-Czitrom S, Kamenicky P, Nevoux J, Prié D, Rothenbuhler A, Wicart P, Harvengt P. Therapeutic management of hypophosphatemic rickets from infancy to adulthood. Endocr Connect. 2014;3:R13–R30. doi: 10.1530/EC-13-0103.
    1. Skrinar A, Dvorak-Ewell M, Evins A, Macica C, Linglart A, Imel EA, Theodore-Oklota C, San Martin J. The lifelong impact of X-linked hypophosphatemia: results from a burden of disease survey. J Endocr Soc. 2019;3:1321–1334. doi: 10.1210/js.2018-00365.
    1. Collins M. Burosumab: at long last, an effective treatment for FGF23-associated hypophosphatemia. J Bone Miner Res. 2018;33:1381–1382. doi: 10.1002/jbmr.3544.
    1. Carpenter TO, Imel EA, Ruppe MD, Weber TJ, Klausner MA, Wooddell MM, Kawakami T, Ito T, Zhang Z, Humphrey J, Insogna KL, Peacock M. Randomized trial of the anti-FGF23 antibody KRN23 in X-linked hypophosphatemia. J Clin Invest. 2014;124:1587–1597. doi: 10.1172/JCI72829.
    1. Imel EA, Zhang X, Ruppe MD, Weber TJ, Klausner MA, Ito T, Vergeire M, Humphrey JS, Glorieux FH, Portale AA, Insogna K, Peacock M, Carpenter TO. Prolonged correction of serum phosphorus in adults with X-linked hypophosphatemia using monthly doses of KRN23. J Clin Endocrinol Metab. 2015;100:2565–2573. doi: 10.1210/jc.2015-1551.
    1. Carpenter TO, Whyte MP, Imel EA, Boot AM, Högler W, Linglart A, Padidela R, Van't Hoff W, Mao M, Chen C-Y, Skrinar A, Kakkis E, San Martin J, Portale AA. Burosumab therapy in children with X-linked hypophosphatemia. N Engl J Med. 2018;378:1987–1998. doi: 10.1056/NEJMoa1714641.
    1. 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 C-Y, Skrinar A, San Martin J, Portale AA. Burosumab versus conventional therapy in children with X-linked hypophosphataemia: a randomised, active-controlled, open-label, phase 3 trial. Lancet. 2019;393:2416–2427. doi: 10.1016/S0140-6736(19)30654-3.
    1. Broderick JE, DeWitt EM, Rothrock N, Crane PK, Forrest CB. Advances in patient-reported outcomes: the NIH PROMIS® measures. EGEMS (Wash DC) 2013;1:1015. doi: 10.13063/2327-9214.1015.
    1. HealthMeasures (2020) Measure Development & Research. Northwestern University. . Accessed 15 Sept 2020.
    1. Nixon A, Williams A, Skrinar A, Theodore-Oklota C (2019) Psychometric validation of the PROMIS® physical function mobility, pain interference and fatigue in a cohort of paediatric X-linked hypophosphatemia (XLH) patients. Proceedings of the International Society for Pharmacoeconomics and Outcomes Research Europe, Nov 2–6, Copenhagen, Denmark.
    1. HealthMeasures (2020) PROMIS. Northwestern University. . Accessed 15 Sept 2020.
    1. Irwin DE, Stucky BD, Thissen D, DeWitt EM, Lai JS, Yeatts K, Varni JW, DeWalt DA. Sampling plan and patient characteristics of the PROMIS pediatrics large-scale survey. Qual Life Res. 2010;19:585–594. doi: 10.1007/s11136-010-9618-4.
    1. Saris-Baglama RN, DeRosa MA, Raczek AE, Bjorner JB, Turner-Bowker DM, Ware JE. The SF-10TM health survey for children: a user’s guide. Lincoln, RI, USA: QualityMetric Incorporated; 2007.
    1. Hicks CL, von Baeyer CL, Spafford PA, van Korlaar I, Goodenough B. The faces pain scale-revised: toward a common metric in pediatric pain measurement. Pain. 2001;93:173–183. doi: 10.1016/s0304-3959(01)00314-1.
    1. Thacher TD, Pettifor JM, Tebben PJ, Creo AL, Skrinar A, Mao M, Chen C-Y, Chang T, San Martin J, Carpenter TO. Rickets severity predicts clinical outcomes in children with X-linked hypophosphatemia: utility of the radiographic rickets severity score. Bone. 2019;122:76–81. doi: 10.1016/j.bone.2019.02.010.
    1. Thissen D, Liu Y, Magnus B, Quinn H, Gipson DS, Dampier C, Huang I-C, Hinds PS, Selewski DT, Reeve BB, Gross HE, DeWalt DA. Estimating minimally important difference (MID) in PROMIS pediatric measures using the scale-judgment method. Qual Life Res. 2016;25:13–23. doi: 10.1007/s11136-015-1058-8.
    1. Wyrwich KW, Norquist JM, Lenderking WR, Acaster S. Methods for interpreting change over time in patient-reported outcome measures. Qual Life Res. 2013;22:475–483. doi: 10.1007/s11136-012-0175-x.
    1. DeWalt DA, Gross HE, Gipson DS, Selewski DT, DeWitt EM, Dampier CD, Hinds PS, Huang I-C, Thissen D, Varni JW. PROMIS® pediatric self-report scales distinguish subgroups of children within and across six common pediatric chronic health conditions. Qual Life Res. 2015;24:2195–2208. doi: 10.1007/s11136-015-0953-3.
    1. Arvanitis M, DeWalt DA, Martin CF, Long MD, Chen W, Jaeger B, Sandler RS, Kappelman MD. Patient-reported outcomes measurement information system in children with Crohn's disease. J Pediatr. 2016;174:153–159.e2. doi: 10.1016/j.jpeds.2016.03.069.
    1. Brandon TG, Becker BD, Bevans KB, Weiss PF. Patient-reported outcomes measurement information system tools for collecting patient-reported outcomes in children with juvenile arthritis. Arthritis Care Res (Hoboken) 2017;69:393–402. doi: 10.1002/acr.22937.
    1. Cunningham NR, Kashikar-Zuck S, Mara C, Goldschneider KR, Revicki DA, Dampier C, Sherry DD, Crosby L, Carle A, Cook KF, Morgan EM. Development and validation of the self-reported PROMIS pediatric pain behavior item bank and short form scale. Pain. 2017;158:1323–1331. doi: 10.1097/j.pain.0000000000000914.
    1. Dampier C, Barry V, Gross HE, Lui Y, Thornburg CD, DeWalt DA, Reeve BB. Initial evaluation of the pediatric PROMIS® health domains in children and adolescents with sickle cell disease. Pediatr Blood Cancer. 2016;63:1031–1037. doi: 10.1002/pbc.25944.
    1. Dampier C, Jaeger B, Gross HE, Barry V, Edwards L, Lui Y, DeWalt DA, Reeve BB. Responsiveness of PROMIS® pediatric measures to hospitalizations for sickle pain and subsequent recovery. Pediatr Blood Cancer. 2016;63:1038–1045. doi: 10.1002/pbc.25931.
    1. Reeve BB, Edwards LJ, Jaeger BC, Hinds PS, Dampier C, Gipson DS, Selewski DT, Troost JP, Thissen D, Barry V, Gross HE, DeWalt DA. Assessing responsiveness over time of the PROMIS® pediatric symptom and function measures in cancer, nephrotic syndrome, and sickle cell disease. Qual Life Res. 2018;27:249–257. doi: 10.1007/s11136-017-1697-z.
    1. Jones JT, Carle AC, Wootton J, Liberio B, Lee J, Schanberg LE, Ying J, DeWitt EM, Brunner HI. Validation of patient-reported outcomes measurement information system short forms for use in childhood-onset systemic lupus erythematosus. Arthritis Care Res (Hoboken) 2017;69:133–142. doi: 10.1002/acr.22927.
    1. Theunissen NC, Vogels TG, Koopman HM, Verrips GH, Zwinderman KA, Verloove-Vanhorick SP, Wit JM. The proxy problem: child report versus parent report in health-related quality of life research. Qual Life Res. 1998;7:387–397. doi: 10.1023/A:1008801802877.
    1. Lyseng-Williamson KA. Burosumab in X-linked hypophosphatemia: a profile of its use in the USA. Drugs Ther Perspect. 2018;34:497–506. doi: 10.1007/s40267-018-0560-9.
    1. Veilleux LN, Cheung M, Ben Amor M, Rauch F. Abnormalities in muscle density and muscle function in hypophosphatemic rickets. J Clin Endocrinol Metab. 2012;97:E1492–E1498. doi: 10.1210/jc.2012-1336.
    1. Pesta DH, Tsirigotis DN, Befroy DE, Caballero D, Jurczak MJ, Rahimi Y, Cline GW, Dufour S, Birkenfeld AL, Rothman DL, Carpenter TO, Insogna K, Petersen KF, Bergwitz C, Shulman GI. Hypophosphatemia promotes lower rates of muscle ATP synthesis. FASEB J. 2016;30:3378–3387. doi: 10.1096/fj.201600473R.
    1. Chen YY, Kao TW, Chou CW, Wu CJ, Yang HF, Lai CH, Wu LW, Chen WL. Exploring the link between serum phosphate levels and low muscle strength, dynapenia, and sarcopenia. Sci Rep. 2018;8:3573. doi: 10.1038/s41598-018-21784-1.
    1. Allen DG, Trajanovska S. The multiple roles of phosphate in muscle fatigue. Front Physiol. 2012;3:463. doi: 10.3389/fphys.2012.00463.
    1. van Onzenoort HA, Menger FE, Neef C, Verbeck WJ, Kroon AA, de Leeuw PW, van der Kuyet PHM. Participation in a clinical trial enhances adherence and persistence to treatment: a retrospective cohort study. Hypertension. 2011;58:573–578. doi: 10.1161/HYPERTENSIONAHA.111.171074.
    1. Carls GS, Tuttle E, Tan RD, Huyuh J, Yee J, Edelman SV, Polonsky WH. Understanding the gap between efficacy in randomized controlled trials and effectiveness in real-world use of GLP-1 RA and DPP-4 therapies in patients with type 2 diabetes. Diabetes Care. 2017;40:1469–1478. doi: 10.2337/dc16-2725.
    1. Mauro MJ, Davis C, Zyczynski T, Khoury HJ. The role of observational studies in optimizing the clinical management of chronic myeloid leukemia. Ther Adv Hematol. 2015;6:3–14. doi: 10.1177/2040620714560305.
    1. Tobert JA, Newman CB. Statin tolerability: in defence of placebo-controlled trials. Eur J Prev Cardiol. 2016;23:891–896. doi: 10.1177/2047487315602861.

Source: PubMed

3
Tilaa