A Phase 3, Open-Label Study of Lumacaftor/Ivacaftor in Children 1 to Less Than 2 Years of Age with Cystic Fibrosis Homozygous for F508del-CFTR

Jonathan H Rayment, Fadi Asfour, Margaret Rosenfeld, Mark Higgins, Lingyun Liu, Molly Mascia, Hildegarde Paz-Diaz, Simon Tian, Rachel Zahigian, Susanna A McColley, Jonathan H Rayment, Fadi Asfour, Margaret Rosenfeld, Mark Higgins, Lingyun Liu, Molly Mascia, Hildegarde Paz-Diaz, Simon Tian, Rachel Zahigian, Susanna A McColley

Abstract

Rationale: Previous phase 3 trials showed that treatment with lumacaftor/ivacaftor was safe and efficacious in people aged ⩾2 years with cystic fibrosis (CF) homozygous for the F508del mutation in CFTR (CF transmembrane conductance regulator) (F/F genotype). Objectives: To assess the safety, pharmacokinetics, and pharmacodynamics of lumacaftor/ivacaftor in children aged 1 to <2 years with the F/F genotype. Methods: This open-label, phase 3 study consisted of two parts (part A [n = 14] and part B [n = 46]) in which two cohorts were enrolled on the basis of age (cohort 1, 18 to <24 mo; cohort 2, 12 to <18 mo). For the 15-day treatment period in part A, the lumacaftor/ivacaftor dose was based on weight at screening. Pharmacokinetic data from part A were used to determine dose-based weight boundaries for part B (24-wk treatment period). Measurements and Main Results: The primary endpoint of part A was pharmacokinetics, and the primary endpoint for part B was safety and tolerability. Secondary endpoints for part B were absolute change in sweat chloride concentration from baseline at Week 24 and pharmacokinetics. Analysis of pharmacokinetic data from part A confirmed the appropriateness of part B dosing. In part B, 44 children (95.7%) had adverse events, which for most were either mild (52.2% of children) or moderate (39.1% of children) in severity. The most common adverse events were cough, infective pulmonary exacerbation of CF, pyrexia, and vomiting. At Week 24, mean absolute change from baseline in sweat chloride concentration was -29.1 mmol/L (95% confidence interval, -34.8 to -23.4 mmol/L). Growth parameters (body mass index, weight, length, and associated z-scores) were normal at baseline and remained normal during the 24-week treatment period. Improving trends in some biomarkers of pancreatic function and intestinal inflammation, such as fecal elastase-1, serum immunoreactive trypsinogen, and fecal calprotectin, were observed. Conclusions: Lumacaftor/ivacaftor was generally safe and well tolerated in children aged 1 to <2 years with the F/F genotype, with a pharmacokinetic profile consistent with studies in older children. Efficacy results, including robust reductions in sweat chloride concentration, suggest the potential for CF disease modification with lumacaftor/ivacaftor treatment. These results support the use of lumacaftor/ivacaftor in this population. Clinical trial registered with www.clinicaltrials.gov (NCT03601637).

Keywords: children; cystic fibrosis; ivacaftor; lumacaftor.

Figures

Figure 1.
Figure 1.
Participant disposition diagram for part B. *This child had an AE of increased alanine aminotransferase and aspartate aminotransferase concentrations that led to treatment discontinuation. AE = adverse event.
Figure 2.
Figure 2.
Predicted areas under the curve (AUCs) for LUM (A) and IVA (B) at steady state for children in part B. In each boxplot, the median is represented by the horizontal line and the box represents the interquartile range. The whiskers represent the largest and smallest values within 1.5 times the interquartile range. Gray bars represent the adult dose exposure, with the upper line of the gray box indicating the 95th percentile of adult AUC values and the lower line of the gray box indicating the 5th percentile. The horizontal dotted lines represent the adult median AUC. Blue dots represent AUC values from individual patients. IVA = ivacaftor; L75/I94 = lumacaftor 75 mg/ivacaftor 94 mg; L100/I125 = lumacaftor 100 mg/ivacaftor 125 mg; L150/I188 = lumacaftor 150 mg/ivacaftor 188 mg; LUM = lumacaftor.
Figure 3.
Figure 3.
Mean absolute change from baseline in sweat chloride concentration by study visit in part B. Mean absolute changes from baseline are presented with 95% CIs, with numbers of children assessed at each study visit indicated on the x-axis. After the 2-week washout period (Weeks 24‒26), mean sweat chloride concentrations returned to baseline at safety follow-up visits. CI = confidence interval.
Figure 4.
Figure 4.
Changes in biomarkers of pancreatic function and intestinal inflammation by study visit in part B. Mean concentrations of fecal elastase-1 (A), serum immunoreactive trypsinogen (B), and fecal calprotectin (C) are shown by study visit. Mean values are presented with 95% CIs, with number of children assessed at each visit indicated on the x-axis. (D) Fecal elastase-1 concentrations in each child (n = 46) at each study visit. Four children who were pancreatic insufficient at baseline (<200 μg/g) had fecal elastase-1 concentrations ⩾200 μg/g at Week 24; 200 μg/g is represented by the horizontal dotted line. CI = confidence interval.

References

    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. Elborn JS. Cystic fibrosis. Lancet . 2016;388:2519–2531.
    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. Anderson MP, Gregory RJ, Thompson S, Souza DW, Paul S, Mulligan RC, et al. Demonstration that CFTR is a chloride channel by alteration of its anion selectivity. Science . 1991;253:202–205.
    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. Nguyen TT, Thia LP, Hoo AF, Bush A, Aurora P, Wade A, et al. London Cystic Fibrosis Collaboration (LCFC) Evolution of lung function during the first year of life in newborn screened cystic fibrosis infants. Thorax . 2014;69:910–917.
    1. Hoo AF, Thia LP, Nguyen TT, Bush A, Chudleigh J, Lum S, et al. London Cystic Fibrosis Collaboration Lung function is abnormal in 3-month-old infants with cystic fibrosis diagnosed by newborn screening. Thorax . 2012;67:874–881.
    1. O’Sullivan BP, Freedman SD. Cystic fibrosis. Lancet . 2009;373:1891–1904.
    1. Abu-El-Haija M, Ramachandran S, Meyerholz DK, Abu-El-Haija M, Griffin M, Giriyappa RL, et al. Pancreatic damage in fetal and newborn cystic fibrosis pigs involves the activation of inflammatory and remodeling pathways. Am J Pathol . 2012;181:499–507.
    1. Stahl M, Steinke E, Graeber SY, Joachim C, Seitz C, Kauczor HU, et al. Magnetic resonance imaging detects progression of lung disease and impact of newborn screening in preschool children with cystic fibrosis. Am J Respir Crit Care Med . 2021;204:943–953.
    1. Ranganathan SC, Hall GL, Sly PD, Stick SM, Douglas TA, Australian Respiratory Early Surveillance Team for Cystic Fibrosis (AREST-CF) Early lung disease in infants and preschool children with cystic fibrosis: what have we learned and what should we do about it? Am J Respir Crit Care Med . 2017;195:1567–1575.
    1. Van Goor F, Hadida S, Grootenhuis PD, Burton B, Cao D, Neuberger T, et al. Rescue of CF airway epithelial cell function in vitro by a CFTR potentiator, VX-770. Proc Natl Acad Sci U S A . 2009;106:18825–18830.
    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. 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. Van Goor F, Straley KS, Cao D, González J, Hadida S, Hazlewood A, et al. Rescue of ΔF508-CFTR trafficking and gating in human cystic fibrosis airway primary cultures by small molecules. Am J Physiol Lung Cell Mol Physiol . 2006;290:L1117–L1130.
    1. Wainwright CE, Elborn JS, Ramsey BW. Lumacaftor-ivacaftor in patients with cystic fibrosis homozygous for Phe508del CFTR. N Engl J Med . 2015;373:1783–1784.
    1. Konstan MW, McKone EF, Moss RB, Marigowda G, Tian S, Waltz D, et al. Assessment of safety and efficacy of long-term treatment with combination lumacaftor and ivacaftor therapy in patients with cystic fibrosis homozygous for the F508del-CFTR mutation (PROGRESS): a phase 3, extension study. Lancet Respir Med . 2017;5:107–118.
    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. 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. McNamara JJ, McColley SA, Marigowda G, Liu F, Tian S, Owen CA, et al. Safety, pharmacokinetics, and pharmacodynamics of lumacaftor and ivacaftor combination therapy in children aged 2–5 years with cystic fibrosis homozygous for F508del-CFTR: an open-label phase 3 study. Lancet Respir Med . 2019;7:325–335.
    1. Rosenfeld M, Wainwright CE, Higgins M, Wang LT, McKee C, Campbell D, et al. ARRIVAL study group Ivacaftor treatment of cystic fibrosis in children aged 12 to <24 months and with a CFTR gating mutation (ARRIVAL): a phase 3 single-arm study. Lancet Respir Med . 2018;6:545–553.
    1. Davies JC, Wainwright CE, Sawicki GS, Higgins MN, Campbell D, Harris C, et al. Ivacaftor in infants aged 4 to <12 months with cystic fibrosis and a gating mutation: results of a two-part phase 3 clinical trial. Am J Respir Crit Care Med . 2021;203:585–593.
    1. Accurso FJ, Van Goor F, Zha J, Stone AJ, Dong Q, Ordonez CL, et al. Sweat chloride as a biomarker of CFTR activity: proof of concept and ivacaftor clinical trial data. J Cyst Fibros . 2014;13:139–147.
    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. 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. Le TN, Anabtawi A, Putman MS, Tangpricha V, Stalvey MS. Growth failure and treatment in cystic fibrosis. J Cyst Fibros . 2019;18:S82–S87.
    1. Walkowiak J, Sands D, Nowakowska A, Piotrowski R, Zybert K, Herzig KH, et al. Early decline of pancreatic function in cystic fibrosis patients with class 1 or 2 CFTR mutations. J Pediatr Gastroenterol Nutr . 2005;40:199–201.
    1. Davies JC, Cunningham S, Harris WT, Lapey A, Regelmann WE, Sawicki GS, et al. KIWI Study Group Safety, pharmacokinetics, and pharmacodynamics of ivacaftor in patients aged 2–5 years with cystic fibrosis and a CFTR gating mutation (KIWI): an open-label, single-arm study. Lancet Respir Med . 2016;4:107–115.
    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. Patel S, Thompson MD, Slaven JE, Sanders DB, Ren CL. Reduction of pulmonary exacerbations in young children with cystic fibrosis during the COVID-19 pandemic. Pediatr Pulmonol . 2021;56:1271–1273.

Source: PubMed

3
Subscribe