A Phase 3 Open-Label Study of Elexacaftor/Tezacaftor/Ivacaftor in Children 6 through 11 Years of Age with Cystic Fibrosis and at Least One F508del Allele

Edith T Zemanick, Jennifer L Taylor-Cousar, Jane Davies, Ronald L Gibson, Marcus A Mall, Edward F McKone, Paul McNally, Bonnie W Ramsey, Jonathan H Rayment, Steven M Rowe, Elizabeth Tullis, Neil Ahluwalia, Chenghao Chu, Thang Ho, Samuel M Moskowitz, Sabrina Noel, Simon Tian, David Waltz, Tanya G Weinstock, Fengjuan Xuan, Claire E Wainwright, Susanna A McColley, Edith T Zemanick, Jennifer L Taylor-Cousar, Jane Davies, Ronald L Gibson, Marcus A Mall, Edward F McKone, Paul McNally, Bonnie W Ramsey, Jonathan H Rayment, Steven M Rowe, Elizabeth Tullis, Neil Ahluwalia, Chenghao Chu, Thang Ho, Samuel M Moskowitz, Sabrina Noel, Simon Tian, David Waltz, Tanya G Weinstock, Fengjuan Xuan, Claire E Wainwright, Susanna A McColley

Abstract

Rationale: Elexacaftor/tezacaftor/ivacaftor (ELX/TEZ/IVA) was shown to be efficacious and safe in patients ≥12 years of age with cystic fibrosis and at least one F508del-CFTR (cystic fibrosis transmembrane conductance regulator) allele, but it has not been evaluated in children <12 years of age. Objectives: To assess the safety, pharmacokinetics, and efficacy of ELX/TEZ/IVA in children 6 through 11 years of age with F508del-minimal function or F508del-F508del genotypes. Methods: In this 24-week open-label phase 3 study, children (N = 66) weighing <30 kg received 50% of the ELX/TEZ/IVA adult daily dose (ELX 100 mg once daily, TEZ 50 mg once daily, and IVA 75 mg every 12 h) whereas children weighing ⩾30 kg received the full adult daily dose (ELX 200 mg once daily, TEZ 100 mg once daily, and IVA 150 mg every 12 h). Measurements and Main Results: The primary endpoint was safety and tolerability. The safety and pharmacokinetic profiles of ELX/TEZ/IVA were generally consistent with those observed in older patients. The most commonly reported adverse events included cough, headache, and pyrexia; in most of the children who had adverse events, these were mild or moderate in severity. Through Week 24, ELX/TEZ/IVA treatment improved the percentage of predicted FEV1 (10.2 percentage points; 95% confidence interval [CI], 7.9 to 12.6), Cystic Fibrosis Questionnaire-Revised respiratory domain score (7.0 points; 95% CI, 4.7 to 9.2), lung clearance index2.5 (-1.71 units; 95% CI, -2.11 to -1.30), and sweat chloride (-60.9 mmol/L; 95% CI, -63.7 to -58.2); body mass index-for-age z-score increased over the 24-week treatment period when compared with the pretreatment baseline. Conclusions: Our results show ELX/TEZ/IVA is safe and efficacious in children 6 through 11 years of age with at least one F508del-CFTR allele, supporting its use in this patient population. Clinical trial registered with www.clinicaltrials.gov (NCT03691779).

Keywords: child; cystic fibrosis; elexacaftor; ivacaftor; tezacaftor.

Figures

Figure 1.
Figure 1.
Patient disposition diagram for part B. *Patient discontinued treatment because of erythematous rash.
Figure 2.
Figure 2.
Pharmacokinetic exposure simulation. For each boxplot, the median is represented by a horizontal line, and the interquartile range is represented by the box. The whiskers mark the largest and smallest values within 1.5 × interquartile range. Dots represent individual empirical Bayes AUC estimate (EBE) values. In each panel, the dashed horizontal line represents the median of the ⩾18 years AUC values, and the gray shaded area indicates the 5th and 95th percentiles of the ⩾18 years AUC values. The EBE values for 6–11 years are from part B of this study. The EBE values for ⩾12 years are from studies 445-102 and 445-103 (12, 13). AUC = area under the concentration-versus-time curve; AUC0–12 = area under the concentration-versus-time curve (0–12 h after dose); AUC0–24 = area under the concentration-versus-time curve (0–24 h after dose); ELX = elexacaftor; IVA = ivacaftor; TEZ = tezacaftor.
Figure 3.
Figure 3.
Efficacy results, by visit, in part B. (A) Absolute change from baseline at each visit in ppFEV1. (B) Absolute change from baseline at each visit in the respiratory domain score on the CFQ-R, child’s version; scores are normalized to a 100-point range, with higher scores indicating a higher patient-reported quality of life with regard to respiratory symptoms. (C) Absolute change from baseline at each visit in LCI2.5. (D) Absolute change from baseline at each visit in sweat chloride concentration; a reduction indicates improvement in CFTR (cystic fibrosis transmembrane conductance regulator) function. Data are least squares means based on a mixed-effects model for repeated measures, and error bars indicate SEMs; the dashed line indicates no change from baseline. Sample size shown under each x-axis is the number of patients at the time point with evaluable in-clinic data. CFQ-R = Cystic Fibrosis Questionnaire–Revised; LCI2.5 = lung clearance index2.5; ppFEV1 = percentage of predicted FEV1.
Figure 4.
Figure 4.
Absolute change from baseline at each visit in (A) BMI, (B) BMI-for-age z-score, (C) weight, (D) weight-for-age z-score, (E) height, and (F) height-for-age z-score in part B. Data are least squares means based on a mixed-effects model for repeated measures, and error bars indicate SEMs; the dashed line indicates no change from baseline. Sample size shown under each x-axis is the number of patients at the time point with evaluable in-clinic data. BMI = body mass index.
Figure 5.
Figure 5.
Responder analysis for sweat chloride concentration by genotype group in part B. The percentage of children in each genotype group with sweat chloride concentrations n (the number of patients with sweat chloride concentration below the indicated threshold) by N1, where N1 is the number of patients with evaluable data. Patients with missing data were considered missing at random and were not counted in the denominator. F/F = homozygous for the F508del-CFTR mutation; F/MF = heterozygous for the F508del-CFTR mutation and a minimal function CFTR mutation.

References

    1. Riordan JR, Rommens JM, Kerem B, Alon N, Rozmahel R, Grzelczak Z, et al. Identification of the cystic fibrosis gene: cloning and characterization of complementary DNA. Science. 1989;245:1066–1073.
    1. Cystic Fibrosis Foundation. Bethesda, MD: Cystic Fibrosis Foundation; 2020.
    1. Elborn JS. Cystic fibrosis. Lancet. 2016;388:2519–2531.
    1. Bell SC, Mall MA, Gutierrez H, Macek M, Madge S, Davies JC, et al. The future of cystic fibrosis care: a global perspective. Lancet Respir Med. 2020;8:65–124.
    1. Ratjen F, Bell SC, Rowe SM, Goss CH, Quittner AL, Bush A. Cystic fibrosis. Nat Rev Dis Primers. 2015;1:15010.
    1. Schlüter DK, Griffiths R, Adam A, Akbari A, Heaven ML, Paranjothy S, et al. Impact of cystic fibrosis on birthweight: a population based study of children in Denmark and Wales. Thorax. 2019;74:447–454.
    1. VanDevanter DR, Kahle JS, O’Sullivan AK, Sikirica S, Hodgkins PS. Cystic fibrosis in young children: a review of disease manifestation, progression, and response to early treatment. J Cyst Fibros. 2016;15:147–157.
    1. Farrell PM, Kosorok MR, Rock MJ, Laxova A, Zeng L, Lai HC, et al. Wisconsin Cystic Fibrosis Neonatal Screening Study Group. Early diagnosis of cystic fibrosis through neonatal screening prevents severe malnutrition and improves long-term growth. Pediatrics. 2001;107:1–13.
    1. Lopes-Pacheco M. CFTR modulators: the changing face of cystic fibrosis in the era of precision medicine. Front Pharmacol. 2020;10:1662.
    1. Boyle MP, De Boeck K. A new era in the treatment of cystic fibrosis: correction of the underlying CFTR defect. Lancet Respir Med. 2013;1:158–163.
    1. Mall MA, Mayer-Hamblett N, Rowe SM. Cystic fibrosis: emergence of highly effective targeted therapeutics and potential clinical implications. Am J Respir Crit Care Med. 2020;201:1193–1208.
    1. Heijerman HGM, McKone EF, Downey DG, Van Braeckel E, Rowe SM, Tullis E, et al. VX17-445-103 Trial Group. Efficacy and safety of the elexacaftor plus tezacaftor plus ivacaftor combination regimen in people with cystic fibrosis homozygous for the F508del mutation: a double-blind, randomised, phase 3 trial. Lancet. 2019;394:1940–1948.
    1. Middleton PG, Mall MA, Dřevínek P, Lands LC, McKone EF, Polineni D, et al. VX17-445-102 Study Group. Elexacaftor–tezacaftor–ivacaftor for cystic fibrosis with a single Phe508del allele. N Engl J Med. 2019;381:1809–1819.
    1. Davies JC, Wainwright CE, Canny GJ, Chilvers MA, Howenstine MS, Munck A, et al. VX08-770-103 (ENVISION) Study Group. Efficacy and safety of ivacaftor in patients aged 6 to 11 years with cystic fibrosis with a G551D mutation. Am J Respir Crit Care Med. 2013;187:1219–1225.
    1. Walker S, Flume P, McNamara J, Solomon M, Chilvers M, Chmiel J, et al. VX15-661-113 Investigator Group. A phase 3 study of tezacaftor in combination with ivacaftor in children aged 6 through 11 years with cystic fibrosis. J Cyst Fibros. 2019;18:708–713.
    1. Milla CE, Ratjen F, Marigowda G, Liu F, Waltz D, Rosenfeld M. VX13-809-011 Part B Investigator Group. Lumacaftor/ivacaftor in patients aged 6–11 years with cystic fibrosis and homozygous for F508del-CFTR. Am J Respir Crit Care Med. 2017;195:912–920.
    1. Ramsey BW, Davies J, McElvaney NG, Tullis E, Bell SC, Dřevínek P, et al. VX08-770-102 Study Group. A CFTR potentiator in patients with cystic fibrosis and the G551D mutation. N Engl J Med. 2011;365:1663–1672.
    1. Taylor-Cousar JL, Munck A, McKone EF, van der Ent CK, Moeller A, Simard C, et al. Tezacaftor-ivacaftor in patients with cystic fibrosis homozygous for Phe508del. N Engl J Med. 2017;377:2013–2023.
    1. Wainwright CE, Elborn JS, Ramsey BW, Marigowda G, Huang X, Cipolli M, et al. TRAFFIC Study Group; TRANSPORT Study Group. Lumacaftor-ivacaftor in patients with cystic fibrosis homozygous for Phe508del CFTR. N Engl J Med. 2015;373:220–231.
    1. Ratjen F, Hug C, Marigowda G, Tian S, Huang X, Stanojevic S, et al. VX14-809-109 investigator group. Efficacy and safety of lumacaftor and ivacaftor in patients aged 6-11 years with cystic fibrosis homozygous for F508del-CFTR: a randomised, placebo-controlled phase 3 trial. Lancet Respir Med. 2017;5:557–567.
    1. Frauchiger BS, Binggeli S, Yammine S, Spycher B, Krüger L, Ramsey KA, et al. Longitudinal course of clinical lung clearance index in children with cystic fibrosis. Eur Respir J. doi: 10.1183/13993003.02686-2020.
    1. Bessonova L, Volkova N, Higgins M, Bengtsson L, Tian S, Simard C, et al. Data from the US and UK cystic fibrosis registries support disease modification by CFTR modulation with ivacaftor. Thorax. 2018;73:731–740.
    1. Sawicki GS, McKone EF, Pasta DJ, Millar SJ, Wagener JS, Johnson CA, et al. Sustained Benefit from ivacaftor demonstrated by combining clinical trial and cystic fibrosis patient registry data. Am J Respir Crit Care Med. 2015;192:836–842.
    1. Konstan MW, Butler SM, Wohl ME, Stoddard M, Matousek R, Wagener JS, et al. Investigators and Coordinators of the Epidemiologic Study of Cystic Fibrosis. Growth and nutritional indexes in early life predict pulmonary function in cystic fibrosis. J Pediatr. 2003;142:624–630.
    1. Stallings VA, Stark LJ, Robinson KA, Feranchak AP, Quinton H. Clinical Practice Guidelines on Growth and Nutrition Subcommittee; Ad Hoc Working Group. Evidence-based practice recommendations for nutrition-related management of children and adults with cystic fibrosis and pancreatic insufficiency: results of a systematic review. J Am Diet Assoc. 2008;108:832–839.
    1. Wilschanski M, Dupuis A, Ellis L, Jarvi K, Zielenski J, Tullis E, et al. Mutations in the cystic fibrosis transmembrane regulator gene and in vivo transepithelial potentials. Am J Respir Crit Care Med. 2006;174:787–794.
    1. Farrell PM, White TB, Ren CL, Hempstead SE, Accurso F, Derichs N, et al. Diagnosis of cystic fibrosis: consensus guidelines from the Cystic Fibrosis Foundation J Pediatr 2017181SS4–S15, e1.. [Published erratum appears in J Pediatr 184:243.]
    1. McKone EF, Velentgas P, Swenson AJ, Goss CH. Association of sweat chloride concentration at time of diagnosis and CFTR genotype with mortality and cystic fibrosis phenotype. J Cyst Fibros. 2015;14:580–586.
    1. Davies JC, Sermet-Gaudelus I, Naehrlich L, Harris RS, Campbell D, Ahluwalia N, et al. VX16-661-115 Investigator Group. A phase 3, double-blind, parallel-group study to evaluate the efficacy and safety of tezacaftor in combination with ivacaftor in participants 6 through 11 years of age with cystic fibrosis homozygous for F508del or heterozygous for the F508del-CFTR mutation and a residual function mutation. J Cyst Fibros. 2021;20:68–77.
    1. McGarry ME, McColley SA. Cystic fibrosis patients of minority race and ethnicity less likely eligible for CFTR modulators based on CFTR genotype. Pediatr Pulmonol. 2021;56:1496–1503.

Source: PubMed

3
S'abonner