Infigratinib in children with achondroplasia: the PROPEL and PROPEL 2 studies

Ravi Savarirayan, Josep Maria De Bergua, Paul Arundel, Helen McDevitt, Valerie Cormier-Daire, Vrinda Saraff, Mars Skae, Borja Delgado, Antonio Leiva-Gea, Fernando Santos-Simarro, Jean Pierre Salles, Marc Nicolino, Massimiliano Rossi, Peter Kannu, Michael B Bober, John Phillips 3rd, Howard Saal, Paul Harmatz, Christine Burren, Garrett Gotway, Terry Cho, Elena Muslimova, Richard Weng, Daniela Rogoff, Julie Hoover-Fong, Melita Irving, Ravi Savarirayan, Josep Maria De Bergua, Paul Arundel, Helen McDevitt, Valerie Cormier-Daire, Vrinda Saraff, Mars Skae, Borja Delgado, Antonio Leiva-Gea, Fernando Santos-Simarro, Jean Pierre Salles, Marc Nicolino, Massimiliano Rossi, Peter Kannu, Michael B Bober, John Phillips 3rd, Howard Saal, Paul Harmatz, Christine Burren, Garrett Gotway, Terry Cho, Elena Muslimova, Richard Weng, Daniela Rogoff, Julie Hoover-Fong, Melita Irving

Abstract

Background: Achondroplasia is the most common short-limbed skeletal dysplasia resulting from gain-of-function pathogenic variants in fibroblast growth factor receptor 3 (FGFR3) gene, a negative regulator of endochondral bone formation. Most treatment options are symptomatic, targeting medical complications. Infigratinib is an orally bioavailable, FGFR1-3 selective tyrosine kinase inhibitor being investigated as a direct therapeutic strategy to counteract FGFR3 overactivity in achondroplasia.

Objectives: The main objective of PROPEL is to collect baseline data of children with achondroplasia being considered for future enrollment in interventional studies sponsored by QED Therapeutics. The objectives of PROPEL 2 are to obtain preliminary evidence of safety and efficacy of oral infigratinib in children with achondroplasia, to identify the infigratinib dose to be explored in future studies, and to characterize the pharmacokinetic (PK) profile of infigratinib and major metabolites.

Design: PROPEL (NCT04035811) is a prospective, noninterventional clinical study designed to characterize the natural history and collect baseline data of children with achondroplasia over 6-24 months. PROPEL 2 (NCT04265651), a prospective, phase II, open-label study of infigratinib in children with achondroplasia, consists of a dose-escalation, dose-finding, and dose-expansion phase to confirm the selected dose, and a PK substudy.

Methods and analysis: Children aged 3-11 years with achondroplasia who completed ⩾6 months in PROPEL are eligible for PROPEL 2. Primary endpoints include treatment-emergent adverse events and change from baseline in annualized height velocity. Four cohorts at ascending dose levels are planned for dose escalation. The selected dose will be confirmed in the dose-expansion phase.

Ethics: PROPEL and PROPEL 2 are being conducted in accordance with the International Conference on Harmonization Good Clinical Practice guidelines, principles of the Declaration of Helsinki, and relevant human clinical research and data privacy regulations. Protocols have been approved by local health authorities, ethics committees, and institutions as applicable. Parents/legally authorized representatives are required to provide signed informed consent; signed informed assent by the child is also required, where applicable.

Discussion: PROPEL and PROPEL 2 will provide preliminary evidence of the safety and efficacy of infigratinib as precision treatment of children with achondroplasia and will inform the design of future studies of FGFR-targeted agents in achondroplasia.

Registration: ClinicalTrials.gov: NCT04035811; NCT04265651.

Keywords: achondroplasia; clinical trial; fibroblast growth factor receptor 3; infigratinib.

Conflict of interest statement

Conflict of interest statement: The authors declared the following potential conflicts of interest with respect to the research, authorship, and/or publication of this article: RS: reports grant support from Ascendis, BioMarin, QED and Theracon, received consulting fees from BioMarin and was a paid advisory board member for Ascendis, BioMarin, QED and Sanofi. JMDB: has nothing to disclose. PA: has nothing to disclose. HM: has nothing to declare. VC-D: was a paid advisory board member for BioMarin. VS: has nothing to disclose. MS: has nothing to disclose. BD: has nothing to disclose. AL-G: has nothing to disclose. FS-S: was a paid advisory board member for BioMarin. JPS: has nothing to disclose. MN: has nothing to disclose. MR: was a paid advisory board member for BioMarin. PK: has nothing to disclose. MB: reports grant support and paid consultancies from Ascendis Pharma, BioMarin, Pfizer, QED Therapeutics. JPIII: has nothing to disclose. HS: reports grant support from Ascendis Pharma, BioMarin, Pfizer, QED Therapeutics. PH: reports paid consutancies for Audentis, Aeglea, Homology, BioMarin, Shire, Genzyme, Ultragenyx, JCR, Denali, Orphazyme, Inventiva, Paradigm, REGENXBIO, Sangamo, QED, Ascendis Pharma, contracts for research from BioMarin and Inventiva and honoraria from BioMarin, Shire, Genzyme, Ultragenyx, and Ophazyme. CB: reports research support from Amgen, Pfizer, QED Therapeutics. GG: has nothing to disclose. TC, EM, RW, DR: report that they are employees of QED therapeutics. JH-F: reports paid consultancies from Pfizer/Therachon, BioMarin, QED Therapeutics, Sanofi, Ascendis Pharma, and grant support fromPfizer/Therachon, BioMarin, Ascendis Pharma. MI: was a paid advisory board member for Ascendis Pharma, BioMarin, QED Therapeutics, Sanofi, Therachon/Pfizer.

© The Author(s), 2022.

Figures

Figure 1.
Figure 1.
Design of the PROPEL and PROPEL 2 studies. ACH, achondroplasia; PK, pharmacokinetic; TEAE, treatment-emergent adverse event.
Figure 2.
Figure 2.
PROPEL study flowchart. BL, baseline; M, month; NTEAE, non-treatment-emergent adverse event.
Figure 3.
Figure 3.
PROPEL 2 study flow chart. DRC, Data Review Committee; PK, pharmacokinetic.

References

    1. Horton WA, Hall JG, Hecht JT. Achondroplasia. Lancet 2007; 370: 162–172.
    1. Waller DK, Correa A, Vo TM, et al.. The population-based prevalence of achondroplasia and thanatophoric dysplasia in selected regions of the US. Am J Med Genet A 2008; 146a: 2385–2389.
    1. Hoover-Fong J, Cheung MS, Fano V, et al.. Lifetime impact of achondroplasia: current evidence and perspectives on the natural history. Bone 2021; 146: 115872.
    1. Pauli RM. Achondroplasia: a comprehensive clinical review. Orphanet J Rare Dis 2019; 14: 1.
    1. Unger S, Bonafé L, Gouze E. Current care and investigational therapies in achondroplasia. Curr Osteoporos Rep 2017; 15: 53–60.
    1. Hecht JT, Butler IJ. Neurologic morbidity associated with achondroplasia. J Child Neurol 1990; 5: 84–97.
    1. Hunter AG, Bankier A, Rogers JG, et al.. Medical complications of achondroplasia: a multicentre patient review. J Med Genet 1998; 35: 705–712.
    1. Hoover-Fong JE, Schulze KJ, McGready J, et al.. Age-appropriate body mass index in children with achondroplasia: interpretation in relation to indexes of height. Am J Clin Nutr 2008; 88: 364–371.
    1. Bellus GA, Hefferon TW, Ortiz de, Luna RI, et al.. Achondroplasia is defined by recurrent G380R mutations of FGFR3. Am J Hum Genet 1995; 56: 368–373.
    1. Shiang R, Thompson LM, Zhu YZ, et al.. Mutations in the transmembrane domain of FGFR3 cause the most common genetic form of dwarfism, achondroplasia. Cell 1994; 78: 335–342.
    1. Horton WA, Degnin CR. FGFs in endochondral skeletal development. Trends Endocrinol Metab 2009; 20: 341–348.
    1. Savarirayan R, Ireland P, Irving M, et al.. International Consensus Statement on the diagnosis, multidisciplinary management and lifelong care of individuals with achondroplasia. Nat Rev Endocrinol 2022; 18: 173–189.
    1. Javle MM, Roychowdhury S, Kelley RK, et al.. Final results from a phase II study of infigratinib (BGJ398), an FGFR-selective tyrosine kinase inhibitor, in patients with previously treated advanced cholangiocarcinoma harboring an FGFR2 gene fusion or rearrangement. J Clin Oncol 2021; 39: 265–265.
    1. Pal SK, Rosenberg JE, Hoffman-Censits JH, et al.. Efficacy of BGJ398, a fibroblast growth factor receptor 1-3 inhibitor, in patients with previously treated advanced urothelial carcinoma with FGFR3 alterations. Cancer Discov 2018; 8: 812–821.
    1. Komla-Ebri D, Dambroise E, Kramer I, et al.. Tyrosine kinase inhibitor NVP-BGJ398 functionally improves FGFR3-related dwarfism in mouse model. J Clin Invest 2016; 126: 1871–1884.
    1. Legeai-Mallet L, Li G, Dambkowski C, et al.. Low dose, daily or intermittent administration of infigratinib (BGJ398), a selective FGFR inhibitor, as treatment for achondroplasia in a preclinical mouse model. In: American Society of Human Genetics 69th annual meeting, Houston, TX, 15–19 October 2019.
    1. Liu S, Yuan Y. Bayesian optimal interval designs for phase I clinical trials. J R Stat Soc Ser C Appl Stat 2015; 64: 507–523.
    1. Högler W, Ward LM. New developments in the management of achondroplasia. Wien Med Wochenschr 2020; 170: 104–111.
    1. Javle M, Lowery M, Shroff RT, et al.. Phase II study of BGJ398 in patients with FGFR-altered advanced cholangiocarcinoma. J Clin Oncol 2018; 36: 276–282.
    1. Reyes M, Sinha U, Li G, et al.. SUN-098 low-dose infigratinib treatment does not lead to changes in phosphorous preclinically in mice. J Endocr Soc 2020; 4: 98.
    1. Gollust SE, Thompson RE, Gooding HC, et al.. Living with achondroplasia in an average-sized world: an assessment of quality of life. Am J Med Genet A 2003; 120a: 447–458.
    1. Lorne H, Newman CJ, Unger S. Is height important for quality of life in children with skeletal dysplasias. Eur J Med Genet 2020; 63: 103816.
    1. Savarirayan R, Irving M, Bacino CA, et al.. C-type natriuretic peptide analogue therapy in children with achondroplasia. N Engl J Med 2019; 381: 25–35.
    1. Savarirayan R, Tofts L, Irving M, et al.. Once-daily, subcutaneous vosoritide therapy in children with achondroplasia: a randomised, double-blind, phase 3, placebo-controlled, multicentre trial. Lancet 2020; 396: 684–692.

Source: PubMed

3
Suscribir