1
|
Edwards SJ, Farrar BG, Ennis K, Downes N, Wakefield V, Mackenzie I, Walters A, Jhita T. Ivacaftor-tezacaftor-elexacaftor, tezacaftor-ivacaftor and lumacaftor-ivacaftor for treating cystic fibrosis: a systematic review and economic evaluation. Health Technol Assess 2025; 29:1-111. [PMID: 40418577 DOI: 10.3310/cpld8546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2025] Open
Abstract
Background Cystic fibrosis is a life-limiting genetic condition that affects over 9000 people in England. Cystic fibrosis is usually diagnosed through newborn screening and causes symptoms throughout the body, including the lungs and digestive system. Around 90% of individuals with cystic fibrosis have at least one copy of the F508del mutation on the cystic fibrosis transmembrane conductance regulator gene. Objectives To appraise the clinical effectiveness and cost-effectiveness of elexacaftor-tezacaftor-ivacaftor, tezacaftor-ivacaftor and lumacaftor-ivacaftor within their expected marketing authorisations for treating people with cystic fibrosis and at least one F508del mutation, compared with each other and with established clinical management before these treatments. Methods A de novo systematic literature review (search date February 2023) was conducted searching electronic databases (MEDLINE, EMBASE, Cochrane Central Register of Controlled Trials), bibliographies of relevant systematic literature reviews, clinical trial registers, recent conferences and evidence provided by Vertex Pharmaceuticals (Boston, MA, USA). Data on the following outcomes were summarised: acute change in per cent predicted forced expiratory volume in 1 second (change in weight-for-age z-score; and change in pulmonary exacerbation frequency requiring intravenous antibiotics. Network meta-analyses were conducted where head-to-head data were not available. Data from clinical trials and real-world evidence were examined to assess long-term effectiveness. A patient-level simulation model was developed to assess the cost-effectiveness of the three modulator treatments. The model employed a lifetime horizon and was developed from the perspective of the National Health Service. Results Data from 19 primary studies and 7 open-label extension studies were prioritised in the systematic literature review. Elexacaftor/tezacaftor/ivacaftor was associated with a statistically significant increase in predicted forced expiratory volume in 1 second and weight-for-age z-score and a reduction in pulmonary exacerbations compared with established clinical management, lumacaftor/ivacaftor and tezacaftor/ivacaftor, and also led to a reduction in the rate of predicted forced expiratory volume in 1 second decline relative to established clinical management, although the magnitude of this decrease was uncertain. Lumacaftor/ivacaftor and tezacaftor/ivacaftor were also associated with a statistically significant increase in predicted forced expiratory volume in 1 second and reduction in pulmonary exacerbations relative to established clinical management, but with a smaller effect size than elexacaftor/tezacaftor/ivacaftor. There was some evidence that tezacaftor/ivacaftor reduced the rate of predicted forced expiratory volume in 1 second decline relative to established clinical management, but little evidence that lumacaftor/ivacaftor reduced the rate of predicted forced expiratory volume in 1 second decline relative to established clinical management. The incremental cost-effectiveness ratios from the economic analysis were confidential. However, for all genotypes studied the incremental cost-effectiveness ratios were above what would be considered cost-effective based on the National Institute for Health and Care Excellence threshold of £20,000-30,000 per quality-adjusted life-year gained. Conclusions Despite the improved clinical benefits observed, none of the cystic fibrosis transmembrane conductance regulator gene modulators assessed would be considered cost-effective based on the National Institute for Health and Care Excellence threshold of £20,000-30,000 per quality-adjusted life-year gained. This is largely driven by the high acquisition costs of cystic fibrosis transmembrane conductance regulator gene modulator treatments. Study registration This study is registered as PROSPERO CRD42023399583. Funding This award was funded by the National Institute for Health and Care Research (NIHR) Evidence Synthesis programme (NIHR award ref: NIHR135829) and is published in full in Health Technology Assessment; Vol. 29, No. 19. See the NIHR Funding and Awards website for further award information.
Collapse
|
2
|
Pastor-Vivero MD, Costa I Colomer J, Martín de Vicente C, Vicente-Santamaria S, Garcia Romero R, González Jiménez D, Luna Paredes C. Advances in the treatment of cystic fibrosis: CFTR modulators. An Pediatr (Barc) 2025; 102:503857. [PMID: 40374426 DOI: 10.1016/j.anpede.2025.503857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Accepted: 04/04/2025] [Indexed: 05/17/2025] Open
Abstract
Cystic fibrosis is a severe genetic disease caused by variants in the CFTR gene. Although it is a multisystem disease, respiratory involvement is the main cause of morbidity and mortality. Cystic fibrosis transmembrane conductance regulator modulator (CFTRm) therapies have advanced the treatment of this disease by improving function of this protein. Ivacaftor, the first CFTRm, has been found to significantly improve lung function and quality of life in patients with certain gating variants. However, only a small percentage of patients in Spain are eligible for this treatment. Combinations of correctors and potentiators, such as lumacaftor-ivacaftor or tezacaftor-ivacaftor, have been developed for treatment of patients with the most frequent variant (F508del), although with limited benefits. Triple therapy with elexacaftor-tezacaftor-ivacaftor has been found to significantly improve respiratory, gastrointestinal and nutritional outcomes as well as quality of life, thus changing the management of CF in eligible patients. The impact of triple therapy is also reflected in an increase in life expectancy and a decrease in mortality and lung transplantation. As regards hepatic and pancreatic involvement, while CFTR modulators have exhibited promising effects, further research is required. The use of CFTR modulators has also shifted nutritional status trends in patients with CF, reducing the risk of undernutrition but increasing the risk of obesity. The use of these drugs for treatment of less frequent variants and for potential antenatal treatment is currently being investigated. Despite these advances, there is still a subset of patients who are ineligible for treatment with modulators or highly effective therapy.
Collapse
Affiliation(s)
- Maria Dolores Pastor-Vivero
- Sección de Neumología Pediátrica, Unidad Multidisciplinar de Fibrosis Quística, Hospital Universitario Cruces. Barakaldo. Bizkaia. Spain; Instituto de Investigación Sanitaria Biobizkaia. Barakaldo. Bizkaia. Spain.
| | - Jordi Costa I Colomer
- Servicio de Neumología Pediátrica y Unidad de Fibrosis Quística, Hospital Sant Joan de Déu, Esplugues de Llobregat. Barcelona, Spain
| | | | - Saioa Vicente-Santamaria
- Unidad de Fibrosis Quística, Gastroenterología, Hepatología y Nutrición Pediátrica, Hospital Universitario Ramón y Cajal. Madrid. Spain
| | - Ruth Garcia Romero
- Unidad de Gastroenterología y Nutrición Pediátrica, Hospital Miguel Servet. Zaragoza. Spain
| | - David González Jiménez
- Unidad de Fibrosis Quística, Area de Gestión Clínica de la Infancia y Adolescencia, Hospital Universitario Central de Asturias. Oviedo. Spain
| | - Carmen Luna Paredes
- Sección de Neumología Pediátrica, Unidad Multidisciplinar de Fibrosis Quística, Hospital Universitario 12 de Octubre. Madrid. Spain
| |
Collapse
|
3
|
Blaisonneau E, Le Daré B, Mercerolle M, Bacle A, Triquet L, Osmont MN, Belleguic C, Polard E. [Adverse effects of the tezacaftor/ivacaftor/elexacaftor combination that may lead to discontinuation: About a series of 10 cases]. Therapie 2025; 80:279-293. [PMID: 39174453 DOI: 10.1016/j.therap.2024.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 05/31/2024] [Accepted: 06/27/2024] [Indexed: 08/24/2024]
Abstract
INTRODUCTION Cystic fibrosis transmembrane regulator (CFTR) channel modulators (ivacaftor, lumacaftor, tezacaftor and elexacaftor) represent a major advance in the management of cystic fibrosis. However, few data are available on the real-life safety profile of these medications, in particular on adverse events that may lead to their discontinuation. The aim of this study is to describe the characteristics and evolution of adverse reactions to the tezacaftor/ivacaftor/elexacaftor combination that led to discontinuation and were reported to the Centre régional de pharmacovigilance (CRPV) in Rennes (France). MATERIALS AND METHODS A retrospective study was conducted from December 2021 to May 2023, focusing on cases of discontinuation of the tezacaftor/ivacaftor/elexacaftor combination due to the occurrence of one or more adverse effects, and reported to the CRPV of Rennes, France. RESULTS Ten cases of drug discontinuation were reported to the Rennes CRPV (6 women/4 men). Adverse effects mainly involved neuropsychiatric disorders (n=6), followed by liver disorders (n=2), ear, nose and throat disorders (n=1), and digestive disorders (n=1). The average duration of treatment at discontinuation was 339.8 [39-668] days. The drug was reintroduced in 7 patients on average 48.7 [7-123] days after discontinuation, with a dosage adjustments (n=4) consisting of changes in dosing times or a reduction in daily doses, with varying success in alleviating adverse symptoms depending on the case. CONCLUSION This small case series suggests that neuropsychiatric adverse effects may occur more frequently than initially described after initiation of tezacaftor/ivacaftor/elexacaftor, and should be carefully screened and monitored. Dosage or administration schedule modifications may be considered for patients experiencing these adverse effects. Further pharmacovigilance studies are needed to better understand the adverse effect profiles of "caftors", their possible risk factors, and the impact of adjusting dosing modalities.
Collapse
Affiliation(s)
- Elora Blaisonneau
- Pôle pharmacie, centre hospitalier universitaire de Rennes, 35000 Rennes, France
| | - Brendan Le Daré
- Pôle pharmacie, centre hospitalier universitaire de Rennes, 35000 Rennes, France; Inserm, INRAE, UMR_A 1341, UMR_S 1317, NuMeCan Institute (Nutrition, Metabolisms and Cancer), CHU de Rennes, université de Rennes, 35000 Rennes, France.
| | - Marion Mercerolle
- Pôle pharmacie, centre hospitalier universitaire de Rennes, 35000 Rennes, France
| | - Astrid Bacle
- Pôle pharmacie, centre hospitalier universitaire de Rennes, 35000 Rennes, France; Inserm, EHESP, Irset-UMR_S 1085, CHU de Rennes, université de Rennes, 35000 Rennes, France
| | - Louise Triquet
- Centre régional de pharmacovigilance, centre hospitalier universitaire de Rennes, 35000 Rennes, France
| | - Marie-Noëlle Osmont
- Centre régional de pharmacovigilance, centre hospitalier universitaire de Rennes, 35000 Rennes, France
| | - Chantal Belleguic
- CRCM adulte, service de pneumologie, centre hospitalier universitaire de Rennes, 35000 Rennes, France
| | - Elisabeth Polard
- Centre régional de pharmacovigilance, centre hospitalier universitaire de Rennes, 35000 Rennes, France
| |
Collapse
|
4
|
Sheikh S, Holtzlander M, Eisner M, Gushue C, Palacios S, Kotha K, Imran S, McCoy KS. Real-life impact of genotype and severity of lung disease on efficacy of elexacaftor-tezacaftor-ivacaftor in people with cystic fibrosis. Pulm Pharmacol Ther 2025; 88:102345. [PMID: 39978537 DOI: 10.1016/j.pupt.2025.102345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 02/14/2025] [Accepted: 02/16/2025] [Indexed: 02/22/2025]
Abstract
BACKGROUND Elexacaftor-tezacaftor-ivacaftor (ETI) therapy has shown improvement in lung function, BMI and reduction in pulmonary exacerbations but the impact of genotype and severity of lung disease on heterogeneity of ETI efficacy in real life is not known. METHODS This is a prospective observational study. Clinical data at baseline and at one-year of therapy were compared for the total cohort and for two subgroups; genotype [homozygous vs. heterozygous for F508del], and severity of lung disease at ETI initiation (ppFEV1 <80 % vs. ≥80 %). RESULTS Among the total cohort of 115 pwCF, median age of 23 (17, 32) years, 66 (58 %) were homozygous, 76 (66 %) had ppFEV1 <80 %. Significant increases in mean ppFEV1 and mean BMI and decrease in MRSA and Pa culture positivity on sputum/throat swab were observed at one year of ETI therapy in the total cohort, and in groups based on either genotype or disease severity (p < 0.05 in all). Comparing one-year prior to one-year on ETI therapy, significant improvements were noted in pulmonary exacerbations, hospital admissions, antibiotic courses, number of pwCF receiving daily chest therapy, dornase alfa and hypertonic saline in the total cohort and in all four subgroups (p < 0.05 for all). Though improvements were not dependent on genotype, we noted larger mean differences in ppFEV1, BMI, pulmonary exacerbations and antibiotic use in the group with more severe lung disease (ppFEV1 <80 %) after one year of ETI therapy. CONCLUSION ETI therapy improved clinical outcomes in pwCF which were impacted by severity of underlying lung disease.
Collapse
Affiliation(s)
- Shahid Sheikh
- Departments of Pediatrics, Ohio State University College of Medicine, Columbus, OH, USA; Division of Pulmonary Medicine, Columbus, OH, USA; Nationwide Children's Hospital, Columbus, OH, USA.
| | - Melissa Holtzlander
- Departments of Pediatrics, Ohio State University College of Medicine, Columbus, OH, USA; Division of Pulmonary Medicine, Columbus, OH, USA; Nationwide Children's Hospital, Columbus, OH, USA
| | - Mariah Eisner
- Nationwide Children's Hospital, Columbus, OH, USA; Biostatistics Resource, Columbus, OH, USA
| | - Courtney Gushue
- Departments of Pediatrics, Ohio State University College of Medicine, Columbus, OH, USA; Division of Pulmonary Medicine, Columbus, OH, USA; Nationwide Children's Hospital, Columbus, OH, USA
| | - Sabrina Palacios
- Departments of Pediatrics, Ohio State University College of Medicine, Columbus, OH, USA; Division of Pulmonary Medicine, Columbus, OH, USA; Nationwide Children's Hospital, Columbus, OH, USA
| | - Kavitha Kotha
- Departments of Pediatrics, Ohio State University College of Medicine, Columbus, OH, USA; Division of Pulmonary Medicine, Columbus, OH, USA; Nationwide Children's Hospital, Columbus, OH, USA
| | - Sehyr Imran
- Departments of Pediatrics, Ohio State University College of Medicine, Columbus, OH, USA; Division of Pulmonary Medicine, Columbus, OH, USA; Nationwide Children's Hospital, Columbus, OH, USA
| | - Karen S McCoy
- Departments of Pediatrics, Ohio State University College of Medicine, Columbus, OH, USA; Division of Pulmonary Medicine, Columbus, OH, USA; Nationwide Children's Hospital, Columbus, OH, USA
| |
Collapse
|
5
|
Fawcett LK, Waters SA, Jaffe A. The equitable challenges to quality use of modulators for cystic fibrosis in Australia. Med J Aust 2025; 222:4-9. [PMID: 39552247 DOI: 10.5694/mja2.52527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 10/14/2024] [Indexed: 11/19/2024]
Affiliation(s)
- Laura K Fawcett
- Sydney Children's Hospital Randwick, Sydney, NSW
- University of New South Wales, Sydney, NSW
| | - Shafagh A Waters
- Sydney Children's Hospital Randwick, Sydney, NSW
- University of New South Wales, Sydney, NSW
| | - Adam Jaffe
- Sydney Children's Hospital Randwick, Sydney, NSW
- University of New South Wales, Sydney, NSW
| |
Collapse
|
6
|
Zwitserloot AM, Aziz SZ, Chen Y, Bannier MAGE, Janssens HM, Merkus PFJM, Nuijsink M, Terheggen‐Lagro SWJ, Tiddens HAWM, Zomer‐ van Ommen DD, Vonk JM, de Winter‐ de Groot KM, Willemse BWM, Koppelman GH. Introduction of Ivacaftor/Lumacaftor in Children With Cystic Fibrosis Homozygous for F508del in the Netherlands: A Nationwide Real-Life Study. Pediatr Pulmonol 2025; 60:e27473. [PMID: 39785291 PMCID: PMC11715133 DOI: 10.1002/ppul.27473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 12/10/2024] [Accepted: 12/23/2024] [Indexed: 01/12/2025]
Abstract
INTRODUCTION Lumacaftor/ivacaftor (lum/iva) was introduced in the Netherlands in 2017. We investigated 1-year efficacy of lum/iva on lung function and small airway and structural lung disease evaluated by multiple breath nitrogen washout and CT scan. Additionally, we investigated effects of lum/iva on exacerbations, anthropometry, sweat chloride and safety in children with CF in the Netherlands. METHODS Children with CF aged 6-18 years and homozygous for F508del treated in one of the seven Dutch CF centers for at least 12 months were eligible. Data were extracted from the Dutch CF Registry and electronic patient records. Primary outcome was change in percent predicted FEV1 (ppFEV1) after 12 months. RESULTS Nationwide, 247 children with CF were eligible for lum/iva. Eight patients discontinued lum/iva due to side effects. In total, 223/247 children (90.3%) were evaluated. Mean (range) age at baseline was 11.0 (6.0-17.1) years. There was no change in FEV1 after 12 months of lum/iva. In a subgroup, markers of small airway function and structural lung disease, such as LCI (n = 28), mean change (SD) -10.0% (15.8), and bronchus-artery (BA) analysis on CT scan (n = 81), showed significant improvement (p < 0.01). Moreover, BMI (n = 192), exacerbations (n = 219) and sweat chloride measurements (n = 105) improved. CONCLUSION Lum/iva was generally well tolerated in a real-life, nationwide pediatric cohort. The efficacy of lum/iva was comparable to phase 3 studies in children. LCI and BA analysis as markers of small airway and structural lung disease showed significant improvement which indicates the importance of these parameters to evaluate treatment effects of CF modulators in children.
Collapse
Affiliation(s)
- A. M. Zwitserloot
- Beatrix Children's Hospital Department of Pediatric Pulmonology and Pediatric AllergyUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC)GroningenThe Netherlands
| | - S. Z. Aziz
- Beatrix Children's Hospital Department of Pediatric Pulmonology and Pediatric AllergyUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC)GroningenThe Netherlands
| | - Y. Chen
- Department of Pediatrics, div of Respiratory Medicine and AllergologyErasmus University Medical Center, Sophia Children's HospitalRotterdamThe Netherlands
- Department of Radiology and Nuclear MedicineErasmus University Medical Center, Sophia Children's HospitalRotterdamThe Netherlands
| | - M. A. G. E. Bannier
- Department of Pediatric Pulmonology, MosaKids Children's HospitalMaastricht University Medical CenterMaastrichtThe Netherlands
| | - H. M. Janssens
- Department of Pediatrics, div of Respiratory Medicine and AllergologyErasmus University Medical Center, Sophia Children's HospitalRotterdamThe Netherlands
| | - P. F. J. M. Merkus
- Department of Pediatric Pulmonology, Amalia Children's HospitalRadboud University Medical CenterNijmegenThe Netherlands
| | - M. Nuijsink
- Juliana Children's Hospital, Haga Teaching HospitalThe HagueThe Netherlands
| | - S. W. J. Terheggen‐Lagro
- Department of Pediatric Pulmonology, Emma Children's HospitalAmsterdam University Medical CentersAmsterdamThe Netherlands
| | - H. A. W. M. Tiddens
- Department of Pediatrics, div of Respiratory Medicine and AllergologyErasmus University Medical Center, Sophia Children's HospitalRotterdamThe Netherlands
- Department of Radiology and Nuclear MedicineErasmus University Medical Center, Sophia Children's HospitalRotterdamThe Netherlands
- ThironaNijmegenThe Netherlands
| | | | - J. M. Vonk
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC)GroningenThe Netherlands
- Department of EpidemiologyUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | - K. M. de Winter‐ de Groot
- Department of Pediatric Pulmonology, Wilhelmina Children's HospitalUniversity Medical CenterUtrecht UniversityUtrechtThe Netherlands
| | - B. W. M. Willemse
- Beatrix Children's Hospital Department of Pediatric Pulmonology and Pediatric AllergyUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC)GroningenThe Netherlands
| | - G. H. Koppelman
- Beatrix Children's Hospital Department of Pediatric Pulmonology and Pediatric AllergyUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC)GroningenThe Netherlands
| |
Collapse
|
7
|
Moiceanu ES, Stan IV, Moşescu SE, Leucuţa DC, Iacobescu M, Niţescu GV, Vivisenco IC, Petran EM, Dumitraşcu DL. Impact of CFTR modulatory therapies on liver function and fibrosis indices in cystic fibrosis patients: a retrospective analysis from two Romanian medical centers. Med Pharm Rep 2025; 98:29-35. [PMID: 39949910 PMCID: PMC11817584 DOI: 10.15386/mpr-2806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 07/31/2024] [Accepted: 08/24/2024] [Indexed: 02/16/2025] Open
Abstract
Background Patients with cystic fibrosis (CF) frequently require modulatory therapies such as Lumacaftor/Ivacaftor (LI) and Elexacaftor/Tezacaftor/Ivacaftor (ETI) to manage their condition. Given the potential hepatic complications associated with CF, it is critical to understand the impact of these therapies on liver function and fibrosis indices. This study aimed to evaluate the changes in liver function markers and fibrosis indices in CF patients undergoing LI and ETI therapies, with a specific focus on the influence of underlying hepatic disease. Methods In this retrospective analysis, liver function markers and fibrosis indices were assessed in CF patients receiving ETI (n=24), LI (n=4), or LI transitioned to ETI (LI/ETI, n=8). Key liver function markers, including alanine aminotransferase (ALT), aspartate aminotransferase (AST), bilirubin, platelet count, and fibrosis indices (APRI and FIB-4), were measured at baseline and at various time points up to 12 months. Results In patients receiving LI therapy, ALT and AST levels demonstrated a slight but non-significant decrease over six months, accompanied by significant fluctuations in total bilirubin levels. Among those receiving ETI therapy, ALT and AST levels initially increased but stabilized over time, while total bilirubin levels significantly increased from baseline to 12 months. No significant differences were observed in liver function markers between patients with and without hepatic disease under ETI therapy. Trends in fibrosis indices (APRI and FIB-4) were modest and largely non-significant across both therapies. Conclusions ETI therapy appears to be safe for CF patients, including those with pre-existing hepatic disease, with no significant deterioration in liver function over a 12-month period. However, the observed fluctuations in bilirubin levels underscore the necessity for ongoing monitoring. Further research is warranted to investigate the long-term hepatic effects of LI and ETI therapies.
Collapse
Affiliation(s)
- Elena-Simona Moiceanu
- Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Pediatric Poison Centre, Grigore Alexandrescu Clinical Emergency Hospital for Children, Bucharest, Romania
| | - Iustina Violeta Stan
- Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
- Alessandrescu-Rusescu National Institute for Mother and Child Health, Bucharest, Romania
| | - Simona Elena Moşescu
- Department of Pediatrics, Grigore Alexandrescu Clinical Emergency Hospital for Children, Bucharest, Romania
| | - Daniel-Corneliu Leucuţa
- Medical Informatics and Biostatistics Department, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Maria Iacobescu
- Institute of Medical Research and Life Sciences – MEDFUTURE, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Gabriela Viorela Niţescu
- Pediatric Poison Centre, Grigore Alexandrescu Clinical Emergency Hospital for Children, Bucharest, Romania
- Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Iolanda Cristina Vivisenco
- Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
- Department of Pediatrics, Grigore Alexandrescu Clinical Emergency Hospital for Children, Bucharest, Romania
| | - Elena Mădălina Petran
- Pediatric Poison Centre, Grigore Alexandrescu Clinical Emergency Hospital for Children, Bucharest, Romania
- Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Dan Lucian Dumitraşcu
- 2nd Department of Internal Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| |
Collapse
|
8
|
Graham E, Heltshe SL, Magaret AS. Baseline-dependent improvement in CF studies, plausibility of bias. Contemp Clin Trials Commun 2024; 42:101378. [PMID: 39678155 PMCID: PMC11639362 DOI: 10.1016/j.conctc.2024.101378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 09/13/2024] [Accepted: 09/25/2024] [Indexed: 12/17/2024] Open
Abstract
Background It has been commonly reported that therapeutic treatments in cystic fibrosis (CF) have ceiling effects, such that their efficacy is diminished for persons with high pre-treatment health (Montgomery et al., 2012 and Newsome et al., 2019). Floor effects have also been reported where decline is of lower magnitude in those with below-average pre-treatment health (Harun et al., 2016; Konstan et al., 2012 and Szczesniak et al., 2017). When measurement error is present, the statistical literature has warned of exaggerated or spurious associations between pre-treatment measures and subsequent change (Chambless and Davis, 2003 and Yanez et al., 1998). Measurement error, equivalently described as day-to-day variation, has been described to occur in CF outcome measurements such as forced expiratory volume in 1 s taken by spirometry (FEV1 pp) (Magaret et al., 2024; Stanojevic et al., 2020 and Thornton et al., 2023). Methods We conducted a simulation study to assess the potential for spurious floor or ceiling effects in studies of CF therapeutics. We considered uncontrolled or single-arm studies, and evaluated estimated association between pre-treatment FEV1 pp and treatment-induced change: post-versus pre-treatment. Results When day-to-day variation was present in FEV1 pp, at levels equivalent to those reported in large studies measuring spirometry both at home and in clinic, naive analytic approaches found spurious associations of change with baseline (Paynter et al., 2022 and Saiman et al., 2003). Type I error ranged from 31.9% to 98.3% for day-to-day variation as high as 3% to 15% relative to biological variation. Incorporating known day-to-day variation, the regression calibration approach corrected bias and controlled type I error (Chambless and Davis, 2003). Conclusion Exaggerated ceiling effects are possible. Further studies could provide meaningful confirmation of ceiling effects in CF, perhaps reducing day-to-day variation by incorporating multiple pre- and post-treatment measurements.
Collapse
Affiliation(s)
- Ellen Graham
- MS University of Washington, Department of Biostatistics, United States of America
| | - Sonya L. Heltshe
- University of Washington, Departments of Pediatrics and Biostatistics Seattle Children’s Research Institute, Center for Respiratory Biology and Therapeutics, United States of America
| | - Amalia S. Magaret
- University of Washington, Departments of Pediatrics and Biostatistics Seattle Children’s Research Institute, Center for Respiratory Biology and Therapeutics, United States of America
| |
Collapse
|
9
|
Escobar NS, Ratjen F. An update on multiple breath washout in children with cystic fibrosis. Expert Rev Respir Med 2024; 18:1061-1071. [PMID: 39709582 DOI: 10.1080/17476348.2024.2445683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 12/18/2024] [Indexed: 12/23/2024]
Abstract
INTRODUCTION Cystic fibrosis (CF) is an autosomal recessive disorder caused by mutations in the CF transmembrane regulator (CFTR) gene, leading to progressive lung disease and systemic complications. Lung disease remains the primary cause of morbidity and mortality, making early detection of lung function decline crucial. The Lung Clearance Index (LCI), derived from the multiple breath washout (MBW) test, has emerged as a sensitive measure for identifying early airway disease. AREAS COVERED This review examines the technical aspects and clinical relevance of LCI, its advantages over traditional lung function tests, and its application in CF clinical trials. A focused literature review highlights LCI's utility in evaluating treatment efficacy and its potential integration into routine CF care. EXPERT OPINION LCI is more sensitive than spirometry for detecting early lung function decline and is predominantly used in pediatric settings. Its use is expanding in adult CF populations as advances in treatment allow adults to maintain stable lung function. In clinical trials, LCI is widely recognized as an outcome measure. While implemented into clinical care in many centers in Europe, this is not yet the case in North America. Faster testing protocols and point-of-care interpretation tools will support LCI's integration into routine CF monitoring.
Collapse
Affiliation(s)
- Natalia S Escobar
- Division of Respiratory Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Felix Ratjen
- Division of Respiratory Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Pediatrics, University of Toronto, Toronto, Ontario, Canada
- Translational Medicine, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada
| |
Collapse
|
10
|
Chatterjee P, Moss CT, Omar S, Dhillon E, Hernandez Borges CD, Tang AC, Stevens DA, Hsu JL. Allergic Bronchopulmonary Aspergillosis (ABPA) in the Era of Cystic Fibrosis Transmembrane Conductance Regulator (CFTR) Modulators. J Fungi (Basel) 2024; 10:656. [PMID: 39330416 PMCID: PMC11433030 DOI: 10.3390/jof10090656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 09/11/2024] [Accepted: 09/13/2024] [Indexed: 09/28/2024] Open
Abstract
Allergic bronchopulmonary aspergillosis (ABPA) is a hypersensitivity disease caused by Aspergillus fumigatus (Af), prevalent in persons with cystic fibrosis (CF) or asthma. In ABPA, Af proteases drive a T-helper cell-2 (Th2)-mediated allergic immune response leading to inflammation that contributes to permanent lung damage. Corticosteroids and antifungals are the mainstays of therapies for ABPA. However, their long-term use has negative sequelae. The treatment of patients with CF (pwCF) has been revolutionized by the efficacy of cystic fibrosis transmembrane conductance regulator (CFTR) modulator therapy. Pharmacological improvement in CFTR function with highly effective elexacaftor/tezacaftor/ivacaftor (ETI) provides unprecedented improvements in lung function and other clinical outcomes of pwCF. The mechanism behind the improvement in patient outcomes is a continued topic of investigation as our understanding of the role of CFTR function evolves. As ETI therapy gains traction in CF management, understanding its potential impact on ABPA, especially on the allergic immune response pathways and Af infection becomes increasingly crucial for optimizing patient outcomes. This literature review aims to examine the extent of these findings and expand our understanding of the already published research focusing on the intersection between ABPA therapeutic approaches in CF and the rapid impact of the evolving CFTR modulator landscape. While our literature search yielded limited reports specifically focusing on the role of CFTR modulator therapy on CF-ABPA, findings from epidemiologic and retrospective studies suggest the potential for CFTR modulator therapies to positively influence pulmonary outcomes by addressing the underlying pathophysiology of CF-ABPA, especially by decreasing inflammatory response and Af colonization. Thus, this review highlights the promising scope of CFTR modulator therapy in decreasing the overall prevalence and incidence of CF-ABPA.
Collapse
Affiliation(s)
- Paulami Chatterjee
- Division of Pulmonary, Allergy and Critical Care Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; (P.C.); (S.O.); (E.D.)
| | - Carson Tyler Moss
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94304, USA
| | - Sarah Omar
- Division of Pulmonary, Allergy and Critical Care Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; (P.C.); (S.O.); (E.D.)
| | - Ekroop Dhillon
- Division of Pulmonary, Allergy and Critical Care Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; (P.C.); (S.O.); (E.D.)
| | | | - Alan C. Tang
- Department of Medicine, Keck School of Medicine, Los Angeles, CA 90089, USA;
| | - David A. Stevens
- Division of Infectious Diseases and Geographic Medicine, Stanford University Medical School, Stanford, CA 94305, USA;
| | - Joe L. Hsu
- Division of Pulmonary, Allergy and Critical Care Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; (P.C.); (S.O.); (E.D.)
| |
Collapse
|
11
|
Kleinfelder K, Melotti P, Hristodor AM, Fevola C, Taccetti G, Terlizzi V, Sorio C. CFTR modulators response of S737F and T465N CFTR variants on patient-derived rectal organoids. Orphanet J Rare Dis 2024; 19:343. [PMID: 39272186 PMCID: PMC11401437 DOI: 10.1186/s13023-024-03334-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 08/21/2024] [Indexed: 09/15/2024] Open
Abstract
BACKGROUND Predictions based on patient-derived materials of CFTR modulators efficacy have been performed lately in patient-derived cells, extending FDA-approved drugs for CF patients harboring rare variants. Here we developed intestinal organoids from subjects carrying S737F- and T465N-CFTR in trans with null alleles to evaluate their functional impact on CFTR protein function and their restoration upon CFTR modulator treatment. The characterization of S737F-CFTR was performed in two subjects recently assessed in nasal epithelial cells but not in colonoids. RESULTS Our functional analysis (Ussing chamber) confirmed that S737F-CFTR is a mild variant with residual function as investigated in colonoids of patients with S737F/Dele22-24 and S737F/W1282X genotypes. An increase of current upon Elexacaftor/Tezacaftor/Ivacaftor (ETI) treatment was recorded for the former genotype. T465N is a poorly characterized missense variant that strongly impacts CFTR function, as almost no CFTR-mediated anion secretion was registered for T465N/Q39X colonoids. ETI treatment substantially improved CFTR-mediated anion secretion and increased the rescue of mature CFTR expression compared to either untreated colonoids or to dual CFTR modulator therapies. CONCLUSIONS Our study confirms the presence of a residual function of the S737F variant and its limited response to CFTR modulators while predicting for the first time the potential clinical benefit of Trikafta® for patients carrying the rare T465N variant.
Collapse
Affiliation(s)
- Karina Kleinfelder
- Department of Medicine, Division of General Pathology, Cystic Fibrosis Laboratory "D. Lissandrini", University of Verona, 37134, Verona, Italy
| | - Paola Melotti
- Cystic Fibrosis Centre, Azienda Ospedaliera Universitaria Integrata Verona, 37126, Verona, Italy
| | - Anca Manuela Hristodor
- Cystic Fibrosis Centre, Azienda Ospedaliera Universitaria Integrata Verona, 37126, Verona, Italy
| | - Cristina Fevola
- Department of Pediatric Medicine, Meyer Children's Hospital IRCCS, Cystic Fibrosis Regional Reference Center, Florence, Italy
| | - Giovanni Taccetti
- Department of Pediatric Medicine, Meyer Children's Hospital IRCCS, Cystic Fibrosis Regional Reference Center, Florence, Italy
| | - Vito Terlizzi
- Department of Pediatric Medicine, Meyer Children's Hospital IRCCS, Cystic Fibrosis Regional Reference Center, Florence, Italy.
| | - Claudio Sorio
- Department of Medicine, Division of General Pathology, Cystic Fibrosis Laboratory "D. Lissandrini", University of Verona, 37134, Verona, Italy.
| |
Collapse
|
12
|
Stahl M, Dohna M, Graeber SY, Sommerburg O, Renz DM, Pallenberg ST, Voskrebenzev A, Schütz K, Hansen G, Doellinger F, Steinke E, Thee S, Röhmel J, Barth S, Rückes-Nilges C, Berges J, Hämmerling S, Wielpütz MO, Naehrlich L, Vogel-Claussen J, Tümmler B, Mall MA, Dittrich AM. Impact of elexacaftor/tezacaftor/ivacaftor therapy on lung clearance index and magnetic resonance imaging in children with cystic fibrosis and one or two F508del alleles. Eur Respir J 2024; 64:2400004. [PMID: 38901883 PMCID: PMC11375515 DOI: 10.1183/13993003.00004-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Accepted: 05/28/2024] [Indexed: 06/22/2024]
Abstract
BACKGROUND We recently demonstrated that elexacaftor/tezacaftor/ivacaftor (ETI) improves the lung clearance index (LCI) and abnormalities in lung morphology detected by magnetic resonance imaging (MRI) in adolescent and adult patients with cystic fibrosis (CF). However, real-world data on the effect of ETI on these sensitive outcomes of lung structure and function in school-age children with CF have not been reported. The aim of this study was therefore to examine the effect of ETI on the LCI and the lung MRI score in children aged 6-11 years with CF and one or two F508del alleles. METHODS This prospective, observational, multicentre, post-approval study assessed the longitudinal LCI up to 12 months and the lung MRI score before and 3 months after initiation of ETI. RESULTS A total of 107 children with CF including 40 heterozygous for F508del and a minimal function mutation (F/MF) and 67 homozygous for F508del (F/F) were enrolled in this study. Treatment with ETI improved the median (interquartile range (IQR)) LCI in F/MF (-1.0 (-2.0- -0.1); p<0.01) and F/F children (-0.8 (-1.9- -0.2); p<0.001) from 3 months onwards. Further, ETI improved the median (IQR) MRI global score in F/MF (-4.0 (-9.0-0.0); p<0.01) and F/F children (-3.5 (-7.3- -0.8); p<0.001). CONCLUSIONS ETI improves early abnormalities in lung ventilation and morphology in school-age children with CF and at least one F508del allele in a real-world setting. Our results support early initiation of ETI to reduce or even prevent lung disease progression in school-age children with CF.
Collapse
Affiliation(s)
- Mirjam Stahl
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, and Cystic Fibrosis Center, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, Berlin, Germany
- German Center for Lung Research (DZL), Associated Partner Site, Berlin, Germany
- M. Stahl, M. Dohna, S.Y. Graeber and O. Sommerburg contributed equally as first authors
| | - Martha Dohna
- Department for Radiology, Hannover Medical School, Hannover, Germany
- M. Stahl, M. Dohna, S.Y. Graeber and O. Sommerburg contributed equally as first authors
| | - Simon Y Graeber
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, and Cystic Fibrosis Center, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, Berlin, Germany
- German Center for Lung Research (DZL), Associated Partner Site, Berlin, Germany
- M. Stahl, M. Dohna, S.Y. Graeber and O. Sommerburg contributed equally as first authors
| | - Olaf Sommerburg
- Division of Pediatric Pulmonology and Allergy, and Cystic Fibrosis Center, Department of Pediatrics, University of Heidelberg, Heidelberg, Germany
- Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), University of Heidelberg, Heidelberg, Germany
- M. Stahl, M. Dohna, S.Y. Graeber and O. Sommerburg contributed equally as first authors
| | - Diane M Renz
- Department for Radiology, Hannover Medical School, Hannover, Germany
| | - Sophia T Pallenberg
- Department for Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
- German Center for Lung Research, Biomedical Research in Endstage and Obstructive Lung Disease (BREATH), Hannover Medical School, Hannover, Germany
| | | | - Katharina Schütz
- Department for Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
- German Center for Lung Research, Biomedical Research in Endstage and Obstructive Lung Disease (BREATH), Hannover Medical School, Hannover, Germany
| | - Gesine Hansen
- Department for Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
- German Center for Lung Research, Biomedical Research in Endstage and Obstructive Lung Disease (BREATH), Hannover Medical School, Hannover, Germany
- Cluster of Excellence RESIST (EXC 2155), German Research Foundation (DFG), Hannover Medical School, Hannover, Germany
| | - Felix Doellinger
- Department of Radiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Eva Steinke
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, and Cystic Fibrosis Center, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, Berlin, Germany
- German Center for Lung Research (DZL), Associated Partner Site, Berlin, Germany
| | - Stephanie Thee
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, and Cystic Fibrosis Center, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, Berlin, Germany
- German Center for Lung Research (DZL), Associated Partner Site, Berlin, Germany
| | - Jobst Röhmel
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, and Cystic Fibrosis Center, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, Berlin, Germany
- German Center for Lung Research (DZL), Associated Partner Site, Berlin, Germany
| | - Sandra Barth
- Department of Pediatrics, Justus Liebig University Giessen, Giessen, Germany
- Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Giessen, Germany
| | - Claudia Rückes-Nilges
- Department of Pediatrics, Justus Liebig University Giessen, Giessen, Germany
- Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Giessen, Germany
| | - Julian Berges
- Division of Pediatric Pulmonology and Allergy, and Cystic Fibrosis Center, Department of Pediatrics, University of Heidelberg, Heidelberg, Germany
- Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), University of Heidelberg, Heidelberg, Germany
| | - Susanne Hämmerling
- Division of Pediatric Pulmonology and Allergy, and Cystic Fibrosis Center, Department of Pediatrics, University of Heidelberg, Heidelberg, Germany
| | - Mark O Wielpütz
- Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), University of Heidelberg, Heidelberg, Germany
- Department of Diagnostic and Interventional Radiology, University of Heidelberg, Heidelberg, Germany
| | - Lutz Naehrlich
- Department of Pediatrics, Justus Liebig University Giessen, Giessen, Germany
- Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Giessen, Germany
- L. Naehrlich, J. Vogel-Claussen, B. Tümmler, M.A. Mall and A-M. Dittrich contributed equally as senior authors
| | - Jens Vogel-Claussen
- Department for Radiology, Hannover Medical School, Hannover, Germany
- German Center for Lung Research, Biomedical Research in Endstage and Obstructive Lung Disease (BREATH), Hannover Medical School, Hannover, Germany
- L. Naehrlich, J. Vogel-Claussen, B. Tümmler, M.A. Mall and A-M. Dittrich contributed equally as senior authors
| | - Burkhard Tümmler
- Department for Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
- German Center for Lung Research, Biomedical Research in Endstage and Obstructive Lung Disease (BREATH), Hannover Medical School, Hannover, Germany
- L. Naehrlich, J. Vogel-Claussen, B. Tümmler, M.A. Mall and A-M. Dittrich contributed equally as senior authors
| | - Marcus A Mall
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, and Cystic Fibrosis Center, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, Berlin, Germany
- German Center for Lung Research (DZL), Associated Partner Site, Berlin, Germany
- L. Naehrlich, J. Vogel-Claussen, B. Tümmler, M.A. Mall and A-M. Dittrich contributed equally as senior authors
| | - Anna-Maria Dittrich
- Department for Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
- German Center for Lung Research, Biomedical Research in Endstage and Obstructive Lung Disease (BREATH), Hannover Medical School, Hannover, Germany
- L. Naehrlich, J. Vogel-Claussen, B. Tümmler, M.A. Mall and A-M. Dittrich contributed equally as senior authors
| |
Collapse
|
13
|
Smith AD, Schwartzman G, Lyons CE, Flowers H, Albon D, Greer K, Lonabaugh K, Zlotoff BJ. Cutaneous manifestations of cystic fibrosis. J Am Acad Dermatol 2024; 91:490-498. [PMID: 38697219 DOI: 10.1016/j.jaad.2024.04.052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 04/19/2024] [Accepted: 04/22/2024] [Indexed: 05/04/2024]
Abstract
Cystic fibrosis (CF) is caused by a mutation in the Cystic fibrosis transmembrane conductance regulator (CFTR) gene, and features recurrent sinus and pulmonary infections, steatorrhea, and malnutrition. CF is associated with diverse cutaneous manifestations, including transient reactive papulotranslucent acrokeratoderma of the palms, nutrient deficiency dermatoses, and vasculitis. Rarely these are presenting symptoms of CF, prior to pulmonary or gastrointestinal sequelae. Cutaneous drug eruptions are also highly common in patients with CF (PwCF) given frequent antibiotic exposure. Finally, CFTR modulating therapy, which has revolutionized CF management, is associated with cutaneous side effects ranging from acute urticaria to toxic epidermal necrolysis. Recognition of dermatologic clinical manifestations of CF is important to appropriately care for PwCF. Dermatologists may play a significant role in the diagnosis and management of CF and associated skin complications.
Collapse
Affiliation(s)
- Aaron D Smith
- University of Virginia School of Medicine, Charlottesville, Virginia.
| | | | - Catherine E Lyons
- University of Virginia School of Medicine, Charlottesville, Virginia
| | - Hal Flowers
- Department of Dermatology, University of Virginia, Charlottesville, Virginia
| | - Dana Albon
- Department of Pulmonology and Critical Care Medicine, University of Virginia, Charlottesville, Virginia
| | - Kenneth Greer
- Department of Dermatology, University of Virginia, Charlottesville, Virginia
| | - Kevin Lonabaugh
- Department of Pulmonology and Critical Care Medicine, University of Virginia, Charlottesville, Virginia
| | - Barrett J Zlotoff
- Department of Dermatology, University of Virginia, Charlottesville, Virginia
| |
Collapse
|
14
|
Radomsky T, Anderson RC, Millar RP, Newton CL. Restoring function to inactivating G protein-coupled receptor variants in the hypothalamic-pituitary-gonadal axis 1. J Neuroendocrinol 2024; 36:e13418. [PMID: 38852954 DOI: 10.1111/jne.13418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 03/30/2024] [Accepted: 05/15/2024] [Indexed: 06/11/2024]
Abstract
G protein-coupled receptors (GPCRs) are central to the functioning of the hypothalamic-pituitary-gonadal axis (HPG axis) and include the rhodopsin-like GPCR family members, neurokinin 3 receptor, kappa-opioid receptor, kisspeptin 1 receptor, gonadotropin-releasing hormone receptor, and the gonadotropin receptors, luteinizing hormone/choriogonadotropin receptor and follicle-stimulating hormone receptor. Unsurprisingly, inactivating variants of these receptors have been implicated in a spectrum of reproductive phenotypes, including failure to undergo puberty, and infertility. Clinical induction of puberty in patients harbouring such variants is possible, but restoration of fertility is not always a realisable outcome, particularly for those patients suffering from primary hypogonadism. Thus, novel pharmaceuticals and/or a fundamental change in approach to treating these patients are required. The increasing wealth of data describing the effects of coding-region genetic variants on GPCR function has highlighted that the majority appear to be dysfunctional as a result of misfolding of the encoded receptor protein, which, in turn, results in impaired receptor trafficking through the secretory pathway to the cell surface. As such, these intracellularly retained receptors may be amenable to 'rescue' using a pharmacological chaperone (PC)-based approach. PCs are small, cell permeant molecules hypothesised to interact with misfolded intracellularly retained proteins, stabilising their folding and promoting their trafficking through the secretory pathway. In support of the use of this approach as a viable therapeutic option, it has been observed that many rescued variant GPCRs retain at least a degree of functionality when 'rescued' to the cell surface. In this review, we examine the GPCR PC research landscape, focussing on the rescue of inactivating variant GPCRs with important roles in the HPG axis, and describe what is known regarding the mechanisms by which PCs restore trafficking and function. We also discuss some of the merits and obstacles associated with taking this approach forward into a clinical setting.
Collapse
Affiliation(s)
- Tarryn Radomsky
- Centre for Neuroendocrinology, Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
- Department of Physiology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Ross C Anderson
- Centre for Neuroendocrinology, Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
- Department of Physiology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Robert P Millar
- Centre for Neuroendocrinology, Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
- Deanery of Biomedical Sciences, University of Edinburgh, Edinburgh, UK
- Faculty of Health Sciences, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa
- School of Medicine, University of St Andrews, St Andrews, UK
| | - Claire L Newton
- Centre for Neuroendocrinology, Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
- Deanery of Biomedical Sciences, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
15
|
Sellers ZM, Assis DN, Paranjape SM, Sathe M, Bodewes F, Bowen M, Cipolli M, Debray D, Green N, Hughan KS, Hunt WR, Leey J, Ling SC, Morelli G, Peckham D, Pettit RS, Philbrick A, Stoll J, Vavrina K, Allen S, Goodwin T, Hempstead SE, Narkewicz MR. Cystic fibrosis screening, evaluation, and management of hepatobiliary disease consensus recommendations. Hepatology 2024; 79:1220-1238. [PMID: 37934656 PMCID: PMC11020118 DOI: 10.1097/hep.0000000000000646] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 10/11/2023] [Indexed: 11/09/2023]
Abstract
Cystic fibrosis (CF) may cause a spectrum of hepatobiliary complications, including portal hypertension, multilobular cirrhosis, and liver failure. Current guidelines on the detection and monitoring of hepatobiliary complications in CF were published in 1999. The CF Foundation assembled a committee to evaluate research advances and formulate revised guidelines for CF-associated liver disease. A committee of hepatologists, gastroenterologists, pulmonologists, pharmacists, nurses, dietitians, individuals with CF, and the parents of a child with CF devised "population, intervention, comparison, and outcome" questions regarding hepatobiliary disease in CF. PubMed literature searches were performed for each population, intervention, comparison, and outcome question. Recommendations were voted on with 80% agreement required to approve a recommendation. Public comment on initial recommendations was solicited prior to the formulation of final recommendations. Thirty-one population, intervention, comparison, and outcome questions were assembled, 6401 manuscripts were title screened for relevance, with 1053 manuscripts undergoing detailed full-text review. Seven recommendations were approved for screening, 13 for monitoring of existing disease, and 14 for treatment of CF-associated hepatobiliary involvement or advanced liver disease. One recommendation on liver biopsy did not meet the 80% threshold. One recommendation on screening ultrasound was revised and re-voted on. Through a multidisciplinary committee and public engagement, we have assembled updated recommendations and guidance on screening, monitoring, and treatment of CF-associated hepatobiliary involvement and advanced liver disease. While research gaps remain, we anticipate that these recommendations will lead to improvements in CF outcomes through earlier detection and increased evidence-based approaches to monitoring and treatment.
Collapse
Affiliation(s)
- Zachary M. Sellers
- Department of Pediatrics, Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Stanford University, Palo Alto, California, USA
| | - David N. Assis
- Department of Medicine, Section of Digestive Diseases, Yale School of Medicine, New Haven, Connecticut, USA
| | - Shruti M. Paranjape
- Division of Pediatric Pulmonology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Meghana Sathe
- Department of Pediatrics, Division of Pediatric Gastroenterology and Nutrition, UT Southwestern, Dallas, Texas, USA
| | - Frank Bodewes
- Department of Pediatric Gastroenterology, University Medical Center Groningen, Groningen, The Netherlands
| | - Melissa Bowen
- Department of Advanced Lung Disease and Lung Transplant, Inova Fairfax Hospital, Falls Church, Virginia, USA
| | - Marco Cipolli
- Cystic Fibrosis Center, Azienda Ospedaliera Universitaria Integrata, Verona, Italy
| | - Dominique Debray
- Pediatric Hepatology Unit, AP-HP, HôpitalNecker-Enfants malades, Paris, France
| | - Nicole Green
- Department of Pediatrics, Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Seattle Children’s Hospital and University of Washington, Seattle, Washington State, USA
| | - Kara S. Hughan
- Department of Pediatrics, Division of Pediatric Endocrinology and Metabolism, UPMC Children’s Hospital of Pittsburgh and University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - William R. Hunt
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep, Emory University, Atlanta, Georgia, USA
| | - Julio Leey
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Florida, Gainesville, Florida, USA
| | - Simon C. Ling
- Department of Paediatrics, Division of Gastroenterology, Hepatology & Nutrition, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Giuseppe Morelli
- Department of Medicine, Division of Gastroenterology, Hepatology, and Nutrition, University of Florida, Gainesville, Florida, USA
| | - Daniel Peckham
- Leeds Institute of Medical Research at St. James’s, University of Leeds, Leeds, UK
| | - Rebeca S. Pettit
- Riley Hospital for Children at IU Health, Indianapolis, Indiana, USA
| | - Alexander Philbrick
- Department of Specialty Pharmacy, Northwestern Medicine, Chicago, Illinois, USA
| | - Janis Stoll
- Department of Pediatrics, Division of Gastroenterology, Hepatology, and Nutrition, Washington University School of Medicine, St Louis, Missouri, USA
| | - Kay Vavrina
- University of Texas, Health Science Center, San Antonio, Texas, USA
| | - Stacy Allen
- CF Parent Community Advisor to Cystic Fibrosis Foundation, USA
| | - Tara Goodwin
- CF Parent Community Advisor to Cystic Fibrosis Foundation, USA
| | | | - Michael R. Narkewicz
- Department of Pediatrics, Section of Pediatric Gastroenterology, Hepatology, and Nutrition, Digestive Health Institute, Children’s Hospital Colorado, University of Colorado School of, Aurora, Colorado, USA
| |
Collapse
|
16
|
Corrao F, Kelly-Aubert M, Sermet-Gaudelus I, Semeraro M. Unmet challenges in cystic fibrosis treatment with modulators. Expert Rev Respir Med 2024; 18:145-157. [PMID: 38755109 DOI: 10.1080/17476348.2024.2357210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 05/15/2024] [Indexed: 05/18/2024]
Abstract
INTRODUCTION 'Highly effective' modulator therapies (HEMTs) have radically changed the Cystic Fibrosis (CF) therapeutic landscape. AREAS COVERED A comprehensive search strategy was undertaken to assess impact of HEMT in life of pwCF, treatment challenges in specific populations such as very young children, and current knowledge gaps. EXPERT OPINION HEMTs are prescribed for pwCF with definite genotypes. The heterogeneity of variants complicates treatment possibilities and around 10% of pwCF worldwide remains ineligible. Genotype-specific treatments are prompting theratyping and personalized medicine strategies. Improvement in lung function and quality of life increase survival rates, shifting CF from a pediatric to an adult disease. This implies new studies addressing long-term efficacy, side effects, emergence of adult co-morbidities and possible drug-drug interactions. More sensitive and predictive biomarkers for both efficacy and toxicity are warranted. As HEMTs cross the placenta and are found in breast milk, studies addressing the potential consequences of treatment during pregnancy and breastfeeding are urgently needed. Finally, although the treatment and expected outcomes of CF have improved dramatically in high- and middle-income countries, lack of access in low-income countries to these life-changing medicines highlights inequity of care worldwide.
Collapse
Affiliation(s)
- Federica Corrao
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Palermo, Italy
- INSERM, Institut Necker Enfants Malades, Paris, France
| | | | - Isabelle Sermet-Gaudelus
- INSERM, Institut Necker Enfants Malades, Paris, France
- Centre de Référence Maladies Rares Mucoviscidose et maladies apparentées. Site constitutif, Université de Paris, Paris, France
- European Reference Lung Center, Frankfurt, Germany
- Université Paris Cité, Paris, France
| | - Michaela Semeraro
- Université Paris Cité, Paris, France
- Centre Investigation Clinique, Hôpital Necker Enfants Malades, Paris, France
| |
Collapse
|
17
|
Teper A, Lubovich S, Rodríguez V, Zaragoza S, Rodríguez E, Bournissen FG. Real-life experience with a generic formulation of lumacaftor-ivacaftor in patients with cystic fibrosis homozygous for the Phe508del CFTR mutation. Pediatr Pulmonol 2023; 58:3560-3565. [PMID: 37712606 DOI: 10.1002/ppul.26690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 08/11/2023] [Accepted: 09/05/2023] [Indexed: 09/16/2023]
Abstract
INTRODUCTION Cystic fibrosis (CF) is the most frequent recessive autosomal disorder in the Caucasian population. It is caused by mutations that result in a deficient or dysfunctional cystic fibrosis transmembrane conductance regulator (CFTR) protein activity. Among CFTR modulators, potentiator compounds increase channel opening, whereas corrector compounds increase CFTR quantity in the cell surface. OBJECTIVE To report real-life effects of a generic formulation of lumacaftor-ivacaftor use in patients with CF homozygous for the Phe508del CFTR mutation. PATIENTS AND METHODS Clinical variables (body mass index [BMI], pulmonary exacerbations, sweat test, and pulmonary function) were analyzed in 30 CF patients homozygous for the Phe508del CFTR mutation, treated with lumacaftor-ivacaftor for 12 months, at the Respiratory Center of Hospital de Niños Ricardo Gutiérrez. These clinical variables were compared with those before the use of modulators. RESULTS A total of 30 patients with CF homozygous for the Phe508del CFTR mutation receiving lumacaftor-ivacaftor therapy were included in this study. The median (interquartile range [IQR]) age at the start of treatment was 10.79 (7.08-14.05) years. Nineteen patients were male. Before treatment, median (IQR) sweat chloride concentration was 80 (72-92) mEq/L, and it had decreased to 74 (68-78) mEq/L (p = .05) 12 months after treatment. Median (IQR) BMI z-score improved from -0.33 (-0.86 to 0.21) to -0.13 (-0.66 to 0.54) (p = .003). A spirometry was performed in 28 of 30 patients. Median (IQR) ppFEV1 was 83.5 (71-91) before treatment and 86.5 (67-103) after treatment (p = .38), 73.3% of patients referred decreased sputum production and 40% reported improvement in their dyspnea at 12 months. Severe pulmonary exacerbations significantly decreased from 60% in the year before treatment, to 30% at 12 months after treatment (p = .037); 13 patients showed an improvement in their exacerbation rates, 2 showed an increased rate, and 15 showed no change. CONCLUSIONS The use of a generic formulation of lumacaftor-ivacaftor in patients homozygous for the Phe508del CFTR mutation was associated with improvement in nutritional status and respiratory symptoms, and a significant reduction in severe pulmonary exacerbations.
Collapse
Affiliation(s)
- Alejandro Teper
- Division of Respiratory, Hospital de Niños Ricardo Gutiérrez, Ciudad de Buenos Aires, Argentina
| | - Silvina Lubovich
- Division of Respiratory, Hospital de Niños Ricardo Gutiérrez, Ciudad de Buenos Aires, Argentina
| | - Viviana Rodríguez
- Division of Respiratory, Hospital de Niños Ricardo Gutiérrez, Ciudad de Buenos Aires, Argentina
| | - Silvina Zaragoza
- Division of Respiratory, Hospital de Niños Ricardo Gutiérrez, Ciudad de Buenos Aires, Argentina
| | - Ezequiel Rodríguez
- Division of Respiratory, Hospital de Niños Ricardo Gutiérrez, Ciudad de Buenos Aires, Argentina
| | - Facundo García Bournissen
- Department of Paediatrics, Schulich School of Medicine and Dentistry Western University, London, Ontario, Canada
| |
Collapse
|
18
|
Pócsi M, Fejes Z, Bene Z, Nagy A, Balogh I, Amaral MD, Macek M, Nagy B. Human epididymis protein 4 (HE4) plasma concentration inversely correlates with the improvement of cystic fibrosis lung disease in p.Phe508del-CFTR homozygous cases treated with the CFTR modulator lumacaftor/ivacaftor combination. J Cyst Fibros 2023; 22:1085-1092. [PMID: 37087300 DOI: 10.1016/j.jcf.2023.04.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 03/30/2023] [Accepted: 04/06/2023] [Indexed: 04/24/2023]
Abstract
BACKGROUND We previously documented that elevated HE4 plasma concentration decreased in people with CF (pwCF) bearing the p.Gly551Asp-CFTR variant in response to CFTR modulator (CFTRm) ivacaftor (IVA), and this level was inversely correlated with the FEV1% predicted values (ppFEV1). Although the effectiveness of lumacaftor (LUM)/IVA in pwCF homozygous for the p.Phe508del-CFTR variant has been evaluated, plasma biomarkers were not used to monitor treatment efficacy thus far. METHODS Plasma HE4 concentration was examined in 68 pwCF drawn from the PROSPECT study who were homozygous for the p.Phe508del-CFTR variant before treatment and at 1, 3, 6 and 12 months after administration of LUM/IVA therapy. Plasma HE4 was correlated with ppFEV1 using their absolute and delta values. The discriminatory power of delta HE4 was evaluated for the detection of lung function improvements based on ROC-AUC analysis and multiple regression test. RESULTS HE4 plasma concentration was significantly reduced below baseline following LUM/IVA administration during the entire study period. The mean change of ppFEV1 was 2.6% (95% CI, 0.6 to 4.5) by 6 months of therapy in this sub-cohort. A significant inverse correlation between delta values of HE4 and ppFEV1 was observed especially in children with CF (r=-0.7053; p<0.0001). Delta HE4 predicted a 2.6% mean change in ppFEV1 (AUC: 0.7898 [95% CI 0.6823-0.8972]; P < 0.0001) at a cut-off value of -10.7 pmol/L. Moreover, delta HE4 independently represented the likelihood of being a responder with ≥ 5% delta ppFEV1 at 6 months (OR: 0.89, 95% CI: 0.82-0.95; P = 0.001). CONCLUSIONS Plasma HE4 level negatively correlates with lung function improvement assessed by ppFEV1 in pwCF undergoing LUM/IVA CFTRm treatment.
Collapse
Affiliation(s)
- Marianna Pócsi
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary; Laki Kálmán Doctoral School, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zsolt Fejes
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zsolt Bene
- Department of Pediatrics, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Attila Nagy
- Department of Preventive Medicine, Faculty of Public Health, University of Debrecen, Debrecen, Hungary
| | - István Balogh
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary; Division of Clinical Genetics, Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Margarida D Amaral
- BioISI - Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisboa, Lisboa, Portugal
| | - Milan Macek
- Department of Biology and Medical Genetics, 2nd Faculty of Medicine of Charles University and Motol University Hospital, Prague, Czech Republic
| | - Béla Nagy
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.
| |
Collapse
|
19
|
Davis MD, Zein JG, Carraro S, Gaston B. Defining and Promoting Pediatric Pulmonary Health: Developing Biomarkers for Pulmonary Health. Pediatrics 2023; 152:e2023062292C. [PMID: 37656025 PMCID: PMC10484306 DOI: 10.1542/peds.2023-062292c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/16/2023] [Indexed: 09/02/2023] Open
Abstract
Children with inherited and/or acquired respiratory disorders often arrive in adolescence and adulthood with diminished lung function that might have been detected and prevented had better mechanisms been available to identify and to assess progression of disease. Fortunately, advances in genetic assessments, low-cost diagnostics, and minimally- invasive novel biomarkers are being developed to detect and to treat respiratory diseases before they give rise to loss of life or lung function. This paper summarizes the Developing Biomarkers for Pulmonary Health sessions of the National Heart, Lung, and Blood Institute- sponsored 2021 Defining and Promoting Pediatric Pulmonary Health workshop. These sessions discussed genetic testing, pulse oximetry, exhaled nitric oxide, and novel biomarkers related to childhood lung diseases.
Collapse
Affiliation(s)
- Michael D. Davis
- Wells Center for Pediatric Research and Division of Pulmonology, Allergy, and Sleep Medicine, Riley Hospital for Children at Indiana University, Indianapolis, Indiana
| | - Joe G. Zein
- Department of Pulmonary Medicine, Respiratory Institute and Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Silvia Carraro
- Unit of Pediatric Allergy and Respiratory Medicine, Women’s and Children’s Health Department, University of Padova, Padova, Italy
| | - Benjamin Gaston
- Wells Center for Pediatric Research and Division of Pulmonology, Allergy, and Sleep Medicine, Riley Hospital for Children at Indiana University, Indianapolis, Indiana
| |
Collapse
|
20
|
Westhoff J, Barth S, Naehrlich L. Drug desensitization to lumacaftor/ivacaftor: A fast lane to drug tolerance. J Cyst Fibros 2023; 22:941-943. [PMID: 37045685 DOI: 10.1016/j.jcf.2023.03.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 03/26/2023] [Accepted: 03/26/2023] [Indexed: 04/14/2023]
Abstract
We present the case of a girl (now 11 years and 9 months old) with cystic fibrosis (F508del homozygote), who developed pruritic rash and urticaria six days after the first dose of the CFTR modulators lumacaftor/ivacaftor. The treatment was paused and had to be interrupted due to an immediate recurrence of the urticarial rash after rechallenge. We developed a drug desensitization protocol, aligned to protocols used for desensitization against oral antibiotics. In contrast to other published protocols, it was performed by rapidly increasing the dose of lumacaftor/ivacaftor granulate at 15 min intervals. The medication was continued without interruption, the rash did not reappear during follow-up of two years. This drug desensitization protocol provides a potential new therapeutic option for patients with drug hypersensitivity reactions to CFTR modulators, especially when there are no alternative treatments. Lumacaftor/ivacaftor is available as granulate, doses can be titrated during desensitization and used for long-term treatment.
Collapse
Affiliation(s)
- J Westhoff
- Department of Pediatrics, Pediatric Pulmonology, Justus-Liebig-University Giessen, Germany.
| | - S Barth
- Department of Pediatrics, Pediatric Pulmonology, Justus-Liebig-University Giessen, Germany
| | - L Naehrlich
- Department of Pediatrics, Pediatric Pulmonology, Justus-Liebig-University Giessen, Germany
| |
Collapse
|
21
|
McNally P, Linnane B, Williamson M, Elnazir B, Short C, Saunders C, Kirwan L, David R, Kemner-Van de Corput MPC, Tiddens HAWM, Davies JC, Cox DW. The clinical impact of Lumacaftor-Ivacaftor on structural lung disease and lung function in children aged 6-11 with cystic fibrosis in a real-world setting. Respir Res 2023; 24:199. [PMID: 37568199 PMCID: PMC10416528 DOI: 10.1186/s12931-023-02497-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 07/21/2023] [Indexed: 08/13/2023] Open
Abstract
BACKGROUND Data from clinical trials of lumacaftor-ivacaftor (LUM-IVA) demonstrate improvements in lung clearance index (LCI) but not in FEV1 in children with Cystic Fibrosis (CF) aged 6-11 years and homozygous for the Phe508del mutation. It is not known whether LUM/IVA use in children can impact the progression of structural lung disease. We sought to determine the real-world impact of LUM/IVA on lung structure and function in children aged 6-11 years. METHODS This real-world observational cohort study was conducted across four paediatric sites in Ireland over 24-months using spirometry-controlled CT scores and LCI as primary outcome measures. Children commencing LUM-/IVA as part of routine care were included. CT scans were manually scored with the PRAGMA CF scoring system and analysed using the automated bronchus-artery (BA) method. Secondary outcome measures included rate of change of ppFEV1, nutritional indices and exacerbations requiring hospitalisation. RESULTS Seventy-one participants were recruited to the study, 31 of whom had spirometry-controlled CT performed at baseline, and after one year and two years of LUM/IVA treatment. At two years there was a reduction from baseline in trapped air scores (0.13 to 0.07, p = 0.016), but an increase from baseline in the % bronchiectasis score (0.84 to 1.23, p = 0.007). There was no change in overall % disease score (2.78 to 2.25, p = 0.138). Airway lumen to pulmonary artery ratios (AlumenA ratio) were abnormal at baseline and worsened over the course of the study. In 28 participants, the mean annual change from baseline LCI2.5 (-0.055 (-0.61 to 0.50), p = 0.85) measurements over two years were not significant. Improvements from baseline in weight (0.10 (0.06 to 0.15, p < 0.0001), height (0.05 (0.02 to 0.09), p = 0.002) and BMI (0.09 (0.03 to 0.15) p = 0.005) z-scores were seen with LUM/IVA treatment. The mean annual change from baseline ppFEV1 (-2.45 (-4.44 to 2.54), p = 0.66) measurements over two years were not significant. CONCLUSION In a real-world setting, the use of LUM/IVA over two years in children with CF aged 6-11 resulted in improvements in air trapping on CT but worsening in bronchiectasis scores. Our results suggest that LUM/IVA use in this age group improves air trapping but does not prevent progression of bronchiectasis over two years of treatment.
Collapse
Affiliation(s)
- Paul McNally
- Respiratory Department, Children's Health Ireland, Crumlin, Dublin, Ireland
- RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Barry Linnane
- University of Limerick School of Medicine, Limerick, Ireland
| | - Michael Williamson
- Respiratory Department, Children's Health Ireland, Crumlin, Dublin, Ireland
| | - Basil Elnazir
- Respiratory Department, Children's Health Ireland, Crumlin, Dublin, Ireland
- Trinity College, Dublin, Ireland
| | - Christopher Short
- NHLI, Imperial College, London, UK
- Royal Brompton and Harefield Hospitals, Guy's and St Thomas' Trust, London, UK
| | - Clare Saunders
- NHLI, Imperial College, London, UK
- Royal Brompton and Harefield Hospitals, Guy's and St Thomas' Trust, London, UK
| | - Laura Kirwan
- Cystic Fibrosis Registry of Ireland, Dublin, Ireland
| | - Rea David
- Respiratory Department, Children's Health Ireland, Crumlin, Dublin, Ireland
| | - Mariette P C Kemner-Van de Corput
- Department of Paediatric Pulmonology and Allergology, Department of Radiology and Nuclear Medicine, Erasmus Medical Centre - Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Harm A W M Tiddens
- Respiratory Department, Children's Health Ireland, Crumlin, Dublin, Ireland
| | - Jane C Davies
- NHLI, Imperial College, London, UK
- Royal Brompton and Harefield Hospitals, Guy's and St Thomas' Trust, London, UK
| | - Des W Cox
- Respiratory Department, Children's Health Ireland, Crumlin, Dublin, Ireland.
- University College Dublin, Dublin, Ireland.
| |
Collapse
|
22
|
Wojsyk-Banaszak I, Stachowiak Z, Więckowska B, Andrzejewska M, Tąpolska-Jóźwiak K, Szczepankiewicz A, Sobkowiak P, Bręborowicz A. How Does the Corrected Exhalyzer Software Change the Predictive Value of LCI in Pulmonary Exacerbations in Children with Cystic Fibrosis? Diagnostics (Basel) 2023; 13:2336. [PMID: 37510079 PMCID: PMC10377908 DOI: 10.3390/diagnostics13142336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 05/29/2023] [Accepted: 05/30/2023] [Indexed: 07/30/2023] Open
Abstract
Aim: Recently, the most commonly used for multiple breath washout device, the Exhalyzer D, has been shown to overestimate lung clearance index (LCI) results due to a software error. Our study aimed to compare the predictive values of LCI in the CF pulmonary exacerbations (PE) calculated with the updated (3.3.1) and the previous (3.2.1) version of the Spiroware software. Materials and Methods: The measurements were performed during 259 visits in CF pediatric patients. We used 39ΔPE pairs (PE preceded by stable visit) and 138ΔS pairs (stable visit preceded by stable visit) to compare the LCI changes during PE. The areas under the receiver operating curves (AUCROC) and odds ratios were calculated based on the differences between ΔPEs and ΔSs. The exacerbation risk was estimated using a logistic regression model with generalized estimating equations (GEE). Results: There were statistically significant differences in LCI 2.5% median values measured using the two versions of the software in the stable condition but not during PE. The AUCROC for changes between the two consecutive visits for LCI did not change significantly using the updated Spiroware software. Conclusions: Despite the lower median values, using the recalculated LCI values does not influence the diagnostic accuracy of this parameter in CF PE.
Collapse
Affiliation(s)
- Irena Wojsyk-Banaszak
- Department of Paediatric Pulmonology, Allergy and Clinical Immunology, Poznan University of Medical Sciences, 60-572 Poznan, Poland
| | - Zuzanna Stachowiak
- Molecular and Cell Biology Unit, Department of Paediatric Pulmonology, Allergy and Clinical Immunology, Poznan University of Medical Sciences, 60-572 Poznan, Poland
| | - Barbara Więckowska
- Department of Computer Science and Statistics, Poznan University of Medical Sciences, 60-806 Poznan, Poland
| | - Marta Andrzejewska
- Department of Paediatric Pulmonology, Allergy and Clinical Immunology, Poznan University of Medical Sciences, 60-572 Poznan, Poland
| | - Katarzyna Tąpolska-Jóźwiak
- Department of Paediatric Pulmonology, Allergy and Clinical Immunology, Poznan University of Medical Sciences, 60-572 Poznan, Poland
| | - Aleksandra Szczepankiewicz
- Molecular and Cell Biology Unit, Department of Paediatric Pulmonology, Allergy and Clinical Immunology, Poznan University of Medical Sciences, 60-572 Poznan, Poland
| | - Paulina Sobkowiak
- Department of Paediatric Pulmonology, Allergy and Clinical Immunology, Poznan University of Medical Sciences, 60-572 Poznan, Poland
| | - Anna Bręborowicz
- Department of Paediatric Pulmonology, Allergy and Clinical Immunology, Poznan University of Medical Sciences, 60-572 Poznan, Poland
| |
Collapse
|
23
|
Berges J, Graeber SY, Hämmerling S, Yu Y, Krümpelmann A, Stahl M, Hirtz S, Scheuermann H, Mall MA, Sommerburg O. Effects of lumacaftor-ivacaftor therapy on cystic fibrosis transmembrane conductance regulator function in F508del homozygous patients with cystic fibrosis aged 2-11 years. Front Pharmacol 2023; 14:1188051. [PMID: 37324488 PMCID: PMC10266342 DOI: 10.3389/fphar.2023.1188051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 05/18/2023] [Indexed: 06/17/2023] Open
Abstract
Rationale: Lumacaftor/ivacaftor was approved for the treatment of patients with cystic fibrosis who are homozygous for F508del aged 2 years and older following positive results from phase three trials. However, the improvement in CFTR function associated with lumacaftor/ivacaftor has only been studied in patients over 12 years of age, while the rescue potential in younger children is unknown. Methods: In a prospective study, we aimed to evaluate the effect of lumacaftor/ivacaftor on the CFTR biomarkers sweat chloride concentration and intestinal current measurement as well as clinical outcome parameters in F508del homozygous CF patients 2-11 years before and 8-16 weeks after treatment initiation. Results: A total of 13 children with CF homozygous for F508del aged 2-11 years were enrolled and 12 patients were analyzed. Lumacaftor/ivacaftor treatment reduced sweat chloride concentration by 26.8 mmol/L (p = 0.0006) and showed a mean improvement in CFTR activity, as assessed by intestinal current measurement in the rectal epithelium, of 30.5% compared to normal (p = 0.0015), exceeding previous findings of 17.7% of normal in CF patients homozygous for F508del aged 12 years and older. Conclusion: Lumacaftor/ivacaftor partially restores F508del CFTR function in children with CF who are homozygous for F508del, aged 2-11 years, to a level of CFTR activity seen in patients with CFTR variants with residual function. These results are consistent with the partial short-term improvement in clinical parameters.
Collapse
Affiliation(s)
- Julian Berges
- Division of Pediatric Pulmonology and Allergology and Cystic Fibrosis Center, Department of Pediatrics, University of Heidelberg, Heidelberg, Germany
- Department of Translational Pulmonology, Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), University of Heidelberg, Heidelberg, Germany
| | - Simon Y. Graeber
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine and Cystic Fibrosis Center, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health at Charité—Universitätsmedizin Berlin, Berlin, Germany
- German Center for Lung Research (DZL), Associated Partner Site, Berlin, Germany
| | - Susanne Hämmerling
- Division of Pediatric Pulmonology and Allergology and Cystic Fibrosis Center, Department of Pediatrics, University of Heidelberg, Heidelberg, Germany
- Department of Translational Pulmonology, Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), University of Heidelberg, Heidelberg, Germany
| | - Yin Yu
- Division of Pediatric Pulmonology and Allergology and Cystic Fibrosis Center, Department of Pediatrics, University of Heidelberg, Heidelberg, Germany
- Department of Translational Pulmonology, Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), University of Heidelberg, Heidelberg, Germany
| | - Arne Krümpelmann
- Division of Pediatric Pulmonology and Allergology and Cystic Fibrosis Center, Department of Pediatrics, University of Heidelberg, Heidelberg, Germany
- Department of Translational Pulmonology, Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), University of Heidelberg, Heidelberg, Germany
| | - Mirjam Stahl
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine and Cystic Fibrosis Center, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health at Charité—Universitätsmedizin Berlin, Berlin, Germany
- German Center for Lung Research (DZL), Associated Partner Site, Berlin, Germany
| | - Stephanie Hirtz
- Division of Pediatric Pulmonology and Allergology and Cystic Fibrosis Center, Department of Pediatrics, University of Heidelberg, Heidelberg, Germany
- Department of Translational Pulmonology, Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), University of Heidelberg, Heidelberg, Germany
| | - Heike Scheuermann
- Division of Pediatric Pulmonology and Allergology and Cystic Fibrosis Center, Department of Pediatrics, University of Heidelberg, Heidelberg, Germany
- Department of Translational Pulmonology, Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), University of Heidelberg, Heidelberg, Germany
| | - Marcus A. Mall
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine and Cystic Fibrosis Center, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health at Charité—Universitätsmedizin Berlin, Berlin, Germany
- German Center for Lung Research (DZL), Associated Partner Site, Berlin, Germany
| | - Olaf Sommerburg
- Division of Pediatric Pulmonology and Allergology and Cystic Fibrosis Center, Department of Pediatrics, University of Heidelberg, Heidelberg, Germany
- Department of Translational Pulmonology, Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
24
|
Olivier M, Kavvalou A, Welsner M, Hirtz R, Straßburg S, Sutharsan S, Stehling F, Steindor M. Real-life impact of highly effective CFTR modulator therapy in children with cystic fibrosis. Front Pharmacol 2023; 14:1176815. [PMID: 37229253 PMCID: PMC10203630 DOI: 10.3389/fphar.2023.1176815] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 04/17/2023] [Indexed: 05/27/2023] Open
Abstract
Introduction: Recently, cystic fibrosis transmembrane regulator modulator therapy with elexacaftor/tezacaftor/ivacaftor has become available for children with cystic fibrosis (CF) carrying at least one F508del mutation. Objective: To assess the intermediate term effects of elexacaftor/tezacaftor/ivacaftor in children with cystic fibrosis in a real-world setting. Methods: We performed a retrospective analysis of records of children with cystic fibrosis, who started elexacaftor/tezacaftor/ivacaftor between 8/2020 and 10/2022. Pulmonary function tests, nutritional status, sweat chloride and laboratory data were assessed before, 3 and 6 months after the start of elexacaftor/tezacaftor/ivacaftor respectively. Results: Elexacaftor/tezacaftor/ivacaftor was started in 22 children 6-11 years and in 24 children 12-17 years. Twenty-seven (59%) patients were homozygous for F508del (F/F) and 23 (50%) patients were transitioned from ivacaftor/lumacaftor (IVA/LUM) or tezacaftor/ivacaftor (TEZ/IVA) to elexacaftor/tezacaftor/ivacaftor. Overall, mean sweat chloride concentration decreased by 59.3 mmol/L (95% confidence interval: -65.0 to -53.7 mmol/L, p < 0.0001) under elexacaftor/tezacaftor/ivacaftor. Sweat chloride concentration also decreased significantly after transition from IVA/LUM or TEZ/IVA to elexacaftor/tezacaftor/ivacaftor (-47.8 mmol/l; 95% confidence interval: -57.6 to -37.8 mmol/l, n = 14, p < 0.0001). Sweat chloride reduction was more marked in children with the F/F than in those with the F/MF genotype (69.4 vs 45.9 mmol/L, p < 0.0001). At 3 months follow-up, body-mass-index-z-score increased by 0.31 (95% CI, 0.2-0.42, p < 0.0001) with no further increase at 6 months. BMI-for-age-z-score was more markedly improved in the older group. Overall pulmonary function (percent predicted FEV1) at 3 months follow-up increased by 11.4% (95% CI: 8.0-14.9, p < 0.0001) with no further significant change after 6 months. No significant differences were noted between the age groups. Children with the F/MF genotype had a greater benefit regarding nutritional status and pulmonary function tests than those with the F/F genotype. Adverse events led to elexacaftor/tezacaftor/ivacaftor dose reduction in three cases and a temporary interruption of therapy in four cases. Conclusion: In a real-world setting, elexacaftor/tezacaftor/ivacaftor therapy had beneficial clinical effects and a good safety profile in eligible children with cystic fibrosis comparable to previously published data from controlled clinical trials. The positive impact on pulmonary function tests and nutritional status seen after 3 months of elexacaftor/tezacaftor/ivacaftor therapy was sustained at 6 months follow-up.
Collapse
Affiliation(s)
- Margarete Olivier
- Pediatric Pulmonology and Sleep Medicine, Cystic Fibrosis Center, Children’s Hospital, University of Duisburg-Essen, Essen, Germany
| | - Alexandra Kavvalou
- Pediatric Pulmonology and Sleep Medicine, Cystic Fibrosis Center, Children’s Hospital, University of Duisburg-Essen, Essen, Germany
| | - Matthias Welsner
- Department of Pulmonary Medicine, Adult Cystic Fibrosis Center, University Hospital Essen—Ruhrlandklinik, University of Duisburg-Essen, Essen, Germany
| | - Raphael Hirtz
- Pediatric Endocrinology, Children’s Hospital, University of Duisburg-Essen, Essen, Germany
| | - Svenja Straßburg
- Department of Pulmonary Medicine, Adult Cystic Fibrosis Center, University Hospital Essen—Ruhrlandklinik, University of Duisburg-Essen, Essen, Germany
| | - Sivagurunathan Sutharsan
- Department of Pulmonary Medicine, Adult Cystic Fibrosis Center, University Hospital Essen—Ruhrlandklinik, University of Duisburg-Essen, Essen, Germany
| | - Florian Stehling
- Pediatric Pulmonology and Sleep Medicine, Cystic Fibrosis Center, Children’s Hospital, University of Duisburg-Essen, Essen, Germany
| | - Mathis Steindor
- Pediatric Pulmonology and Sleep Medicine, Cystic Fibrosis Center, Children’s Hospital, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
25
|
Appelt D, Steinkamp G, Sieber S, Ellemunter H. Early and sustained improvements of lung clearance index from two to sixteen weeks of elexacaftor/tezacaftor/ivacaftor therapy in patients with cystic fibrosis-a real world study. Front Pharmacol 2023; 14:1125853. [PMID: 36969845 PMCID: PMC10030732 DOI: 10.3389/fphar.2023.1125853] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 02/21/2023] [Indexed: 03/11/2023] Open
Abstract
Since the introduction of CFTR modulator therapies, longitudinal real-life data of lung clearance index (LCI) during treatment is scarce. In this single-centre, post-approval setting, we report data of 51 patients with different stages of lung disease, age 2-52 years with repeated measurements of forced expiratory volume as a percentage of the predicted value (ppFEV₁) and LCI after 2, 4, and 16 weeks of CFTR modulator treatment and at baseline. In 25 patients during elexacaftor/tezacaftor/ivacaftor (ELX/TEZ/IVA) treatment, significant improvements of LCI (median -1.4) and ppFEV₁ (median +8.3%) were observed after only 2 weeks, and were maintained after 4 and 16 weeks of treatment (LCI: -2.0, -2.2; ppFEV₁: +7.2%, +11.8%). We observed a significant correlation between LCI improvement at week 16 and lower baseline LCI. In 26 younger and healthier patients receiving lumacaftor/ivacaftor (LUM/IVA) treatment, no significant changes of LCI and ppFEV₁ occured. With ELX/TEZ/IVA, our data shows rapid, significant improvements of LCI and ppFEV₁ already after 2 weeks. Early LCI measurements can help to assess the patient's response to this high-cost therapy.
Collapse
Affiliation(s)
- Dorothea Appelt
- Cystic Fibrosis Centre Innsbruck, Department of Paediatrics III, Medical University of Innsbruck, Innsbruck, Austria
| | | | - Sarah Sieber
- STAT-UP Statistical Consulting & Data Science GmbH, Munich, Germany
| | - Helmut Ellemunter
- Cystic Fibrosis Centre Innsbruck, Department of Paediatrics III, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
26
|
Athanazio RA, Tanni SE, Ferreira J, Dalcin PDTR, Fuccio MBD, Esposito C, Canan MGM, Coelho LS, Firmida MDC, Almeida MBD, Marostica PJC, Monte LDFV, Souza EL, Pinto LA, Rached SZ, Oliveira VSBD, Riedi CA, Silva Filho LVRFD. Brazilian guidelines for the pharmacological treatment of the pulmonary symptoms of cystic fibrosis. Official document of the Sociedade Brasileira de Pneumologia e Tisiologia (SBPT, Brazilian Thoracic Association). J Bras Pneumol 2023; 49:e20230040. [PMID: 37194817 DOI: 10.36416/1806-3756/e20230040] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Accepted: 03/31/2023] [Indexed: 05/18/2023] Open
Abstract
Cystic fibrosis (CF) is a genetic disease that results in dysfunction of the CF transmembrane conductance regulator (CFTR) protein, which is a chloride and bicarbonate channel expressed in the apical portion of epithelial cells of various organs. Dysfunction of that protein results in diverse clinical manifestations, primarily involving the respiratory and gastrointestinal systems, impairing quality of life and reducing life expectancy. Although CF is still an incurable pathology, the therapeutic and prognostic perspectives are now totally different and much more favorable. The purpose of these guidelines is to define evidence-based recommendations regarding the use of pharmacological agents in the treatment of the pulmonary symptoms of CF in Brazil. Questions in the Patients of interest, Intervention to be studied, Comparison of interventions, and Outcome of interest (PICO) format were employed to address aspects related to the use of modulators of this protein (ivacaftor, lumacaftor+ivacaftor, and tezacaftor+ivacaftor), use of dornase alfa, eradication therapy and chronic suppression of Pseudomonas aeruginosa, and eradication of methicillin-resistant Staphylococcus aureus and Burkholderia cepacia complex. To formulate the PICO questions, a group of Brazilian specialists was assembled and a systematic review was carried out on the themes, with meta-analysis when applicable. The results obtained were analyzed in terms of the strength of the evidence compiled, the recommendations being devised by employing the GRADE approach. We believe that these guidelines represent a major advance to be incorporated into the approach to patients with CF, mainly aiming to favor the management of the disease, and could become an auxiliary tool in the definition of public policies related to CF.
Collapse
Affiliation(s)
- Rodrigo Abensur Athanazio
- . Divisão de Pneumologia, Instituto do Coração, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo (SP) Brasil
| | - Suzana Erico Tanni
- . Faculdade de Medicina de Botucatu, Universidade Estadual Paulista Julio de Mesquita Filho - UNESP - Botucatu (SP) Brasil
| | - Juliana Ferreira
- . Divisão de Pneumologia, Instituto do Coração, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo (SP) Brasil
| | - Paulo de Tarso Roth Dalcin
- . Programa de Pós-Graduação em Ciências Pneumológicas, Universidade Federal do Rio Grande do Sul - UFRGS - Porto Alegre (RS) Brasil
- . Serviço de Pneumologia, Hospital de Clínicas de Porto Alegre - HCPA - Universidade Federal do Rio Grande do Sul - UFRGS - Porto Alegre (RS) Brasil
| | - Marcelo B de Fuccio
- . Hospital Júlia Kubitschek, Fundação Hospitalar do Estado de Minas Gerais - FHEMIG - Belo Horizonte (MG) Brasil
| | | | | | - Liana Sousa Coelho
- . Faculdade de Medicina de Botucatu, Universidade Estadual Paulista Julio de Mesquita Filho - UNESP - Botucatu (SP) Brasil
| | | | - Marina Buarque de Almeida
- . Unidade de Pneumologia, Instituto da Criança, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo (SP) Brasil
| | - Paulo José Cauduro Marostica
- . Unidade de Pneumologia Infantil, Hospital de Clínicas de Porto Alegre - HCPA - Universidade Federal do Rio Grande do Sul - UFRGS - Porto Alegre (RS) Brasil
| | | | - Edna Lúcia Souza
- . Faculdade de Medicina da Bahia, Universidade Federal da Bahia, Salvador (BA) Brasil
| | | | - Samia Zahi Rached
- . Divisão de Pneumologia, Instituto do Coração, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo (SP) Brasil
| | - Verônica Stasiak Bednarczuk de Oliveira
- . Hospital de Clínicas, Universidade Federal do Paraná, Curitiba (PR) Brasil
- . Unidos Pela Vida - Instituto Brasileiro de Atenção à Fibrose Cística, Curitiba (PR) Brasil
| | | | | |
Collapse
|
27
|
Gambazza S, Orenti A, Pizzamiglio G, Zolin A, Colombo C, Laquintana D, Ambrogi F, On behalf of ECFSPR. Association of Oxygen Therapy with the Natural Disease Progression of Cystic Fibrosis: A Multi-State Model of the European Cystic Fibrosis Society Patient Registry. Ther Clin Risk Manag 2023; 19:255-267. [PMID: 36935771 PMCID: PMC10022450 DOI: 10.2147/tcrm.s391476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 02/12/2023] [Indexed: 03/15/2023] Open
Abstract
Background Association between dependence on oxygen therapy (OT) and natural disease progression in people with cystic fibrosis (pwCF) has not been estimated yet. The aim of this study is to understand the prognosis for pwCF on OT, evaluating how the transition probabilities from being alive without lung transplantation (LTx) to LTx and to death, and from being alive after LTx to death change in pwCF with and without OT. Methods We used 2008-2017 data from the 35-country European CF Society Patient Registry. A multi-state model was fitted to assess the effects of individual risk factors on transition probabilities. Results We considered 48,343 pwCF aged from 6 to 50 years. OT (HR 5.78, 95% CI: 5.32-6.29) and abnormal FEV1 (HR 6.41, 95% CI: 5.28-7.79) were strongly associated with the probability of having LTx; chronic infection with Burkholderia cepacia complex (HR 3.19, 95% CI: 2.78-3.67), abnormal FEV1 (HR 5.00, 95% CI: 4.11-6.08) and the need for OT (HR 4.32, 95% CI: 3.93-4.76) showed the greatest association with the probability of dying without LTx. Once pwCF received LTx, OT (HR 1.75, 95% CI: 1.41-2.16) and abnormal FEV1 (HR 1.63, 95% CI: 1.18-2.25) were the main factors associated with the probability of dying. An association of gross national income with the probability of receiving LTx and with the probability of dying without LTx was also found. Conclusion Oxygen therapy is associated with poor survival in pwCF with and without LTx; harmonization of CF care throughout European countries and minimization of the onset of pulmonary gas exchange abnormalities using all available means remains of paramount importance.
Collapse
Affiliation(s)
- Simone Gambazza
- Healthcare Professions Department, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Clinical Sciences and Community Health, Laboratory of Medical Statistics, Biometry and Epidemiology “G. A. Maccacaro”, Università degli Studi di Milano, Milan, Italy
- Correspondence: Simone Gambazza, Healthcare Professions Department, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 35, Milan, 20122, Italy, Email
| | - Annalisa Orenti
- Department of Clinical Sciences and Community Health, Laboratory of Medical Statistics, Biometry and Epidemiology “G. A. Maccacaro”, Università degli Studi di Milano, Milan, Italy
| | - Giovanna Pizzamiglio
- Cystic Fibrosis Center – Adult Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Anna Zolin
- Department of Clinical Sciences and Community Health, Laboratory of Medical Statistics, Biometry and Epidemiology “G. A. Maccacaro”, Università degli Studi di Milano, Milan, Italy
| | - Carla Colombo
- Cystic Fibrosis Center, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milano, Italy
| | - Dario Laquintana
- Healthcare Professions Department, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Federico Ambrogi
- Department of Clinical Sciences and Community Health, Laboratory of Medical Statistics, Biometry and Epidemiology “G. A. Maccacaro”, Università degli Studi di Milano, Milan, Italy
- Scientific Directorate, IRCCS Policlinico San Donato, San Donato Milanese, MI, Italy
| | | |
Collapse
|
28
|
Foucaud P, Mercier JC. CFTR pharmacological modulators: A great advance in cystic fibrosis management. Arch Pediatr 2023; 30:1-9. [PMID: 36509624 DOI: 10.1016/j.arcped.2022.11.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 09/16/2022] [Accepted: 11/11/2022] [Indexed: 12/13/2022]
Abstract
Cystic fibrosis is a severe monogenic disease that affects around 7400 patients in France. More than 2100 mutations in the cystic fibrosis conductance transmembrane regulator (CFTR), the gene encoding for an epithelial ion channel that normally transports chloride and bicarbonate, lead to mucus dehydration and impaired bronchial clearance. Systematic neonatal screening in France since 2002 has enabled early diagnosis of cystic fibrosis. Although highly demanding, supportive treatments including daily chest physiotherapy, inhaled aerosol therapy, frequent antibiotic courses, nutritional and pancreatic extracts have improved the prognosis. Median age at death is now beyond 30 years. Ivacaftor was the first CFTR modulator found to both reduce sweat chloride concentration and improve pulmonary function in the rare CFTR gating mutations. Combinations of modulators such as lumacaftor + ivacaftor or tezacaftor + ivacaftor were found to improve pulmonary function both in patients homozygous for the F508del mutation characterized by the lack of CFTR protein and those heterozygous for F508del with minimal CFTR activity. The triple combination of ivacaftor + tezacaftor + elexacaftor was recently shown to significantly improve pulmonary function and quality of life, to normalize sweat chloride concentration, and to reduce the need for antibiotic therapy in patients with at least one F508del mutation (83% in France). These impressive data, however, need to be confirmed in the long term. Nevertheless, it is encouraging to hear treated patients testify about their markedly improved quality of life and to observe that the number of lung transplants for cystic fibrosis decreased dramatically in France after 2020, despite the COVID pandemic, with no increase in deaths without lung transplant.
Collapse
Affiliation(s)
- P Foucaud
- Vice-Président de l'Association Vaincre la Mucoviscidose, 181 Rue de Tolbiac, Paris 75013, France.
| | - J C Mercier
- Membre de la Commission de Transparence, Haute Autorité de Santé, 5 avenue du Stade de France, Saint Denis 93210, France
| |
Collapse
|
29
|
Organoid Technology and Its Role for Theratyping Applications in Cystic Fibrosis. CHILDREN (BASEL, SWITZERLAND) 2022; 10:children10010004. [PMID: 36670555 PMCID: PMC9856584 DOI: 10.3390/children10010004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/14/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022]
Abstract
Cystic fibrosis (CF) is a autosomal recessive, multisystemic disease caused by different mutations in the CFTR gene encoding CF transmembrane conductance regulator. Although symptom management is important to avoid complications, the approval of CFTR modulator drugs in the clinic has demonstrated significant improvements by targeting the primary molecular defect of CF and thereby preventing problems related to CFTR deficiency or dysfunction. CFTR modulator therapies have positively changed the patients' quality of life, especially for those who start their use at the onset of the disease. Due to early diagnosis with the implementation of newborn screening programs and considerable progress in the treatment options, nowadays pediatric mortality was dramatically reduced. In any case, the main obstacle to treat CF is to predict the drug response of patients due to genetic complexity and heterogeneity. Advances in 3D culture systems have led to the extrapolation of disease modeling and individual drug response in vitro by producing mini organs called "organoids" easily obtained from nasal and rectal mucosa biopsies. In this review, we focus primarily on patient-derived intestinal organoids used as in vitro model for CF disease. Organoids combine high-validity of outcomes with a high throughput, thus enabling CF disease classification, drug development and treatment optimization in a personalized manner.
Collapse
|
30
|
Mall MA, Brugha R, Gartner S, Legg J, Moeller A, Mondejar-Lopez P, Prais D, Pressler T, Ratjen F, Reix P, Robinson PD, Selvadurai H, Stehling F, Ahluwalia N, Arteaga-Solis E, Bruinsma BG, Jennings M, Moskowitz SM, Noel S, Tian S, Weinstock TG, Wu P, Wainwright CE, Davies JC. Efficacy and Safety of Elexacaftor/Tezacaftor/Ivacaftor in Children 6 Through 11 Years of Age with Cystic Fibrosis Heterozygous for F508del and a Minimal Function Mutation: A Phase 3b, Randomized, Placebo-controlled Study. Am J Respir Crit Care Med 2022; 206:1361-1369. [PMID: 35816621 PMCID: PMC9746869 DOI: 10.1164/rccm.202202-0392oc] [Citation(s) in RCA: 81] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Rationale: The triple-combination regimen elexacaftor/tezacaftor/ivacaftor (ELX/TEZ/IVA) was shown to be safe and efficacious in children aged 6 through 11 years with cystic fibrosis and at least one F508del-CFTR allele in a phase 3, open-label, single-arm study. Objectives: To further evaluate the efficacy and safety of ELX/TEZ/IVA in children 6 through 11 years of age with cystic fibrosis heterozygous for F508del and a minimal function CFTR mutation (F/MF genotypes) in a randomized, double-blind, placebo-controlled phase 3b trial. Methods: Children were randomized to receive either ELX/TEZ/IVA (n = 60) or placebo (n = 61) during a 24-week treatment period. The dose of ELX/TEZ/IVA administered was based on weight at screening, with children <30 kg receiving ELX 100 mg once daily, TEZ 50 mg once daily, and IVA 75 mg every 12 hours, and children ⩾30 kg receiving ELX 200 mg once daily, TEZ 100 mg once daily, and IVA 150 mg every 12 hours (adult dose). Measurements and Main Results: The primary endpoint was absolute change in lung clearance index2.5 from baseline through Week 24. Children given ELX/TEZ/IVA had a mean decrease in lung clearance index2.5 of 2.29 units (95% confidence interval [CI], 1.97-2.60) compared with 0.02 units (95% CI, -0.29 to 0.34) in children given placebo (between-group treatment difference, -2.26 units; 95% CI, -2.71 to -1.81; P < 0.0001). ELX/TEZ/IVA treatment also led to improvements in the secondary endpoint of sweat chloride concentration (between-group treatment difference, -51.2 mmol/L; 95% CI, -55.3 to -47.1) and in the other endpoints of percent predicted FEV1 (between-group treatment difference, 11.0 percentage points; 95% CI, 6.9-15.1) and Cystic Fibrosis Questionnaire-Revised Respiratory domain score (between-group treatment difference, 5.5 points; 95% CI, 1.0-10.0) compared with placebo from baseline through Week 24. The most common adverse events in children receiving ELX/TEZ/IVA were headache and cough (30.0% and 23.3%, respectively); most adverse events were mild or moderate in severity. Conclusions: In this first randomized, controlled study of a cystic fibrosis transmembrane conductance regulator modulator conducted in children 6 through 11 years of age with F/MF genotypes, ELX/TEZ/IVA treatment led to significant improvements in lung function, as well as robust improvements in respiratory symptoms and cystic fibrosis transmembrane conductance regulator function. ELX/TEZ/IVA was generally safe and well tolerated in this pediatric population with no new safety findings.
Collapse
Affiliation(s)
- Marcus A. Mall
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin and,Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany;,German Center for Lung Research, Associated Partner, Berlin, Germany
| | - Rossa Brugha
- Great Ormond Street Hospital for Children, London, United Kingdom
| | | | - Julian Legg
- Southampton Children's Hospital, Hampshire, United Kingdom
| | | | | | - Dario Prais
- Schneider Children’s Medical Center of Israel, Petah Tikva, Israel;,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | | - Felix Ratjen
- The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Philippe Reix
- Hôpital Femme Mère-Enfant, Hospices Civils de Lyon, Bron, France
| | - Paul D. Robinson
- The Children’s Hospital at Westmead, Sydney Children’s Hospital Network, Sydney, Australia
| | - Hiran Selvadurai
- The Children’s Hospital at Westmead, Sydney Children’s Hospital Network, Sydney, Australia
| | - Florian Stehling
- Universitätsklinikum Essen, Klinik für Kinderheilkunde III, Essen, Germany
| | - Neil Ahluwalia
- Vertex Pharmaceuticals Incorporated, Boston, Massachussetts
| | | | | | - Mark Jennings
- Vertex Pharmaceuticals Incorporated, Boston, Massachussetts
| | | | - Sabrina Noel
- Vertex Pharmaceuticals Incorporated, Boston, Massachussetts
| | - Simon Tian
- Vertex Pharmaceuticals Incorporated, Boston, Massachussetts
| | | | - Pan Wu
- Vertex Pharmaceuticals Incorporated, Boston, Massachussetts
| | | | - Jane C. Davies
- National Heart and Lung Institute, Imperial College London, London, United Kingdom;,Royal Brompton and Harefield Hospitals, part of Guy’s and St Thomas’ NHS Trust, London, United Kingdom; and,European Cystic Fibrosis Society Lung Clearance Index Core Facility, London, United Kingdom
| | | |
Collapse
|
31
|
Cornet M, Robin G, Ciciriello F, Bihouee T, Marguet C, Roy V, Lebourgeois M, Chedevergne F, Bonnel AS, Kelly M, Reix P, Lucidi V, Stoven V, Sermet-Gaudelus I. Profiling the response to lumacaftor-ivacaftor in children with cystic between fibrosis and new insight from a French-Italian real-life cohort. Pediatr Pulmonol 2022; 57:2992-2999. [PMID: 35996214 PMCID: PMC9826158 DOI: 10.1002/ppul.26123] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 08/11/2022] [Accepted: 08/21/2022] [Indexed: 01/11/2023]
Abstract
INTRODUCTION Clinical trials for CFTR modulators consider mean changes of clinical status at the cohort level, and thus fail to assess the heterogeneity of the response. We aimed to study the different response profiles to lumacaftor-ivacaftor according to age in children with cystic fibrosis (CF). METHODS A mathematical framework, including principal component analysis, data clustering, and data completion, was applied to a multicenter cohort of 112 children aged 6-18 years, treated with lumacaftor-ivacaftor. Studied parameters at baseline and 6 months included body mass index (BMI), number of days of antibiotics (ATB), Sweat test (ST), forced expiratory volume in 1 s expressed in percentage predicted (ppFEV1 ), forced vital capacity (ppFVC), and forced expiratory flow at 25%-75% of FVC (ppFEF25-75 ). RESULTS Change in ppFEV1 was the most significant parameter in characterizing response heterogeneity among the 12-18-year-old patients. Patients with minimal changes in ppFEV1 were further separated by change in BMI and ATB course. In the 6-12-year-old children both BMI and ppFEV1 evolution were the most relevant. ST change was not associated with a clinical response. CONCLUSIONS Change in ppFEV1 , BMI, and ATB course are the most relevant outcomes to discriminate clinical response profiles in children treated with lumacaftor-ivacaftor. Prepubertal and pubertal children display different response profiles.
Collapse
Affiliation(s)
| | - Geneviève Robin
- Laboratoire de Mathematiques et Modelisation d'Evry, Evry, France
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Ramsey KA, Schultz A. Monitoring disease progression in childhood bronchiectasis. Front Pediatr 2022; 10:1010016. [PMID: 36186641 PMCID: PMC9523123 DOI: 10.3389/fped.2022.1010016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 08/29/2022] [Indexed: 11/24/2022] Open
Abstract
Bronchiectasis (not related to cystic fibrosis) is a chronic lung disease caused by a range of etiologies but characterized by abnormal airway dilatation, recurrent respiratory symptoms, impaired quality of life and reduced life expectancy. Patients typically experience episodes of chronic wet cough and recurrent pulmonary exacerbations requiring hospitalization. Early diagnosis and management of childhood bronchiectasis are essential to prevent respiratory decline, optimize quality of life, minimize pulmonary exacerbations, and potentially reverse bronchial disease. Disease monitoring potentially allows for (1) the early detection of acute exacerbations, facilitating timely intervention, (2) tracking the rate of disease progression for prognostic purposes, and (3) quantifying the response to therapies. This narrative review article will discuss methods for monitoring disease progression in children with bronchiectasis, including lung imaging, respiratory function, patient-reported outcomes, respiratory exacerbations, sputum biomarkers, and nutritional outcomes.
Collapse
Affiliation(s)
- Kathryn A. Ramsey
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, University of Western Australia, Perth, WA, Australia
| | - André Schultz
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, University of Western Australia, Perth, WA, Australia
- Respiratory Medicine, Perth Children's Hospital, Perth, WA, Australia
| |
Collapse
|
33
|
Bhaskaran D, Kathryn B. A case of Elexacaftor-Tezacaftor-Ivacaftor induced rash resolving without interruption of treatment. J Cyst Fibros 2022; 21:1077-1079. [PMID: 35840534 DOI: 10.1016/j.jcf.2022.06.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 06/23/2022] [Accepted: 06/25/2022] [Indexed: 10/17/2022]
Abstract
The triple combination of Elexacaftor-Tezacaftor-Ivacaftor (ELX-TEZ-IVA) has been shown to markedly improve lung function in persons with cystic fibrosis (pwCF). An important adverse effect of the drug is rash, which was reported in clinical trials and highlighted in case reports. Our report demonstrates a similar adverse event with the drug in one of our patients with spontaneous resolution of the rash not necessitating cessation of treatment or desensitization to the drug as were done in other cases. We highlight through our report the heterogeneity of the clinical presentation of the rash when on triple therapy and the need for further studies to understand the immunological mechanism of this adverse event.
Collapse
Affiliation(s)
- Divyalakshmi Bhaskaran
- NIHR Academic Clinical Fellow, Leeds teaching Hospitals NHS Foundation Trust, United Kingdom.
| | - Bateman Kathryn
- Cystic Fibrosis/Respiratory Medicine Consultant, Bristol Adult Cystic Fibrosis Centre, United Kingdom
| |
Collapse
|
34
|
Abstract
Over the past decade there have been significant developments in the field of Cystic Fibrosis Transmembrane Regulator modulator drugs. Following treatment in patients with cystic fibrosis with common gating mutations using the potentiator drug ivacaftor, successive development of corrector drugs used in combination has led to highly effective modulator therapy being available to more than 85% of the cystic fibrosis population over 12 years of age in the form of elexacaftor/tezacaftor/ivacaftor. In this article, we review the evidence from clinical trials and mounting real-world observational and registry data that demonstrates the impact highly effective modulators have on both pulmonary and extra-pulmonary manifestations of cystic fibrosis. As clinical trials progress to younger patient groups, we discuss the challenges to demonstrating drug efficacy in early life, and also consider practicalities of drug development in an ever-shrinking modulator-naïve population. Drug-drug interactions are an important consideration in people with cystic fibrosis, where polypharmacy is commonplace, but also as the modulated population look to remain healthier for longer, we identify trials that aim to address treatment burden too. Inequity of care, through drug cost or ineligibility for modulators by genotype, is widening without apparent strategies to address this; however, we present evidence of hopeful early-stage drug development for non-modulatable genes and summarise the current state of gene-therapy development.
Collapse
|
35
|
Targeting fibrosis, mechanisms and cilinical trials. Signal Transduct Target Ther 2022; 7:206. [PMID: 35773269 PMCID: PMC9247101 DOI: 10.1038/s41392-022-01070-3] [Citation(s) in RCA: 222] [Impact Index Per Article: 74.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 06/17/2022] [Accepted: 06/20/2022] [Indexed: 02/05/2023] Open
Abstract
Fibrosis is characterized by the excessive extracellular matrix deposition due to dysregulated wound and connective tissue repair response. Multiple organs can develop fibrosis, including the liver, kidney, heart, and lung. Fibrosis such as liver cirrhosis, idiopathic pulmonary fibrosis, and cystic fibrosis caused substantial disease burden. Persistent abnormal activation of myofibroblasts mediated by various signals, such as transforming growth factor, platelet-derived growth factor, and fibroblast growh factor, has been recongized as a major event in the occurrence and progression of fibrosis. Although the mechanisms driving organ-specific fibrosis have not been fully elucidated, drugs targeting these identified aberrant signals have achieved potent anti-fibrotic efficacy in clinical trials. In this review, we briefly introduce the aetiology and epidemiology of several fibrosis diseases, including liver fibrosis, kidney fibrosis, cardiac fibrosis, and pulmonary fibrosis. Then, we summarise the abnormal cells (epithelial cells, endothelial cells, immune cells, and fibroblasts) and their interactions in fibrosis. In addition, we also focus on the aberrant signaling pathways and therapeutic targets that regulate myofibroblast activation, extracellular matrix cross-linking, metabolism, and inflammation in fibrosis. Finally, we discuss the anti-fibrotic drugs based on their targets and clinical trials. This review provides reference for further research on fibrosis mechanism, drug development, and clinical trials.
Collapse
|
36
|
Regard L, Martin C, Burnet E, Da Silva J, Burgel PR. CFTR Modulators in People with Cystic Fibrosis: Real-World Evidence in France. Cells 2022; 11:cells11111769. [PMID: 35681464 PMCID: PMC9179538 DOI: 10.3390/cells11111769] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 05/15/2022] [Accepted: 05/23/2022] [Indexed: 01/18/2023] Open
Abstract
Cystic fibrosis (CF) is a rare genetic multisystemic disease, the manifestations of which are due to mutations in the gene encoding the CF transmembrane conductance regulator (CFTR) protein and can lead to respiratory insufficiency and premature death. CFTR modulators, which were developed in the past decade, partially restore CFTR protein function. Their clinical efficacy has been demonstrated in phase 3 clinical trials, particularly in terms of lung function and pulmonary exacerbations, nutritional status, and quality of life in people with gating mutations (ivacaftor), homozygous for the F508del mutation (lumacaftor/ivacaftor and tezacaftor/ivacaftor), and in those with at least one F508del mutation (elexacaftor/tezacaftor/ivacaftor). However, many questions remain regarding their long-term safety and effectiveness, particularly in patients with advanced lung disease, liver disease, renal insufficiency, or problematic bacterial colonization. The impact of CFTR modulators on other important outcomes such as concurrent treatments, lung transplantation, chest imaging, or pregnancies also warrants further investigation. The French CF Reference Network includes 47 CF centers that contribute patient data to the comprehensive French CF Registry and have conducted nationwide real-world studies on CFTR modulators. This review seeks to summarize the results of these real-world studies and examine their findings against those of randomized control trials.
Collapse
Affiliation(s)
- Lucile Regard
- French Cystic Fibrosis National Reference Center, Department of Respiratory Medicine, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, 75014 Paris, France; (L.R.); (C.M.); (E.B.); (J.D.S.)
- Institut Cochin, Université de Paris Cité, INSERM U1016, 75014 Paris, France
- ERN Lung Cystic Fibrosis Network, Frankfurt, Germany
| | - Clémence Martin
- French Cystic Fibrosis National Reference Center, Department of Respiratory Medicine, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, 75014 Paris, France; (L.R.); (C.M.); (E.B.); (J.D.S.)
- Institut Cochin, Université de Paris Cité, INSERM U1016, 75014 Paris, France
- ERN Lung Cystic Fibrosis Network, Frankfurt, Germany
| | - Espérie Burnet
- French Cystic Fibrosis National Reference Center, Department of Respiratory Medicine, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, 75014 Paris, France; (L.R.); (C.M.); (E.B.); (J.D.S.)
- Institut Cochin, Université de Paris Cité, INSERM U1016, 75014 Paris, France
- ERN Lung Cystic Fibrosis Network, Frankfurt, Germany
| | - Jennifer Da Silva
- French Cystic Fibrosis National Reference Center, Department of Respiratory Medicine, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, 75014 Paris, France; (L.R.); (C.M.); (E.B.); (J.D.S.)
- ERN Lung Cystic Fibrosis Network, Frankfurt, Germany
| | - Pierre-Régis Burgel
- French Cystic Fibrosis National Reference Center, Department of Respiratory Medicine, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, 75014 Paris, France; (L.R.); (C.M.); (E.B.); (J.D.S.)
- Institut Cochin, Université de Paris Cité, INSERM U1016, 75014 Paris, France
- ERN Lung Cystic Fibrosis Network, Frankfurt, Germany
- Correspondence: ; Tel.: +33-1-58-41-23-67; Fax: +33-1-46-33-82-53
| |
Collapse
|
37
|
Roda J, Pinto-Silva C, Silva IA, Maia C, Almeida S, Ferreira R, Oliveira G. New drugs in cystic fibrosis: what has changed in the last decade? Ther Adv Chronic Dis 2022; 13:20406223221098136. [PMID: 35620188 PMCID: PMC9128052 DOI: 10.1177/20406223221098136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 04/13/2022] [Indexed: 11/16/2022] Open
Abstract
Cystic fibrosis (CF), a life-limiting chronic disease caused by mutations in the cystic fibrosis transmembrane regulator (CFTR) gene, affects more than 90,000 people worldwide. Until recently, the only available treatments were directed to symptom control, but they failed to change the course of the disease. New drugs developed in the last decade have the potential to change the expression, function, and stability of CFTR protein, targeting the basic molecular defect. The authors seek to provide an update on the new drugs, with a special focus on the most promising clinical trials that have been carried out to date. These newly approved drugs that target specific CFTR mutations are mainly divided into two main groups of CFTR modulators: potentiators and correctors. New therapies have opened the door for potentially disease-modifying, personalized treatments for patients with CF.
Collapse
Affiliation(s)
- Juliana Roda
- Pediatric Gastroenterology and Nutrition Unit, Centro Hospitalar e Universitario de Coimbra EPE Hospital Pediátrico de Coimbra, Avenida Afonso Romão 3000-602 Coimbra, Portugal
| | - Catarina Pinto-Silva
- Pediatric Gastroenterology and Nutrition Unit, Centro Hospitalar e Universitário de Coimbra EPE, Hospital Pediátrico de Coimbra, Coimbra, Portugal
| | - Iris A.I. Silva
- BioISI – Biosystems and Integrative Sciences Institute, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
| | - Carla Maia
- Pediatric Gastroenterology and Nutrition Unit, Centro Hospitalar e Universitário de Coimbra EPE, Hospital Pediátrico de Coimbra, Coimbra, Portugal
| | - Susana Almeida
- Pediatric Gastroenterology and Nutrition Unit, Centro Hospitalar e Universitário de Coimbra EPE, Hospital Pediátrico de Coimbra, Coimbra, Portugal
| | - Ricardo Ferreira
- Pediatric Gastroenterology and Nutrition Unit, Centro Hospitalar e Universitário de Coimbra EPE, Hospital Pediátrico de Coimbra, Coimbra, Portugal
| | - Guiomar Oliveira
- Centro de Desenvolvimento da Criança e Centro de Investigação e Formação Clínica, Centro Hospitalar e Universitario de Coimbra EPE, Hospital Pediátrico de Coimbra, Coimbra, Portugal
| |
Collapse
|
38
|
Graeber SY, Renz DM, Stahl M, Pallenberg ST, Sommerburg O, Naehrlich L, Berges J, Dohna M, Ringshausen FC, Doellinger F, Vitzthum C, Röhmel J, Allomba C, Hämmerling S, Barth S, Rückes-Nilges C, Wielpütz MO, Hansen G, Vogel-Claussen J, Tümmler B, Mall MA, Dittrich AM. Effects of Elexacaftor/Tezacaftor/Ivacaftor Therapy on Lung Clearance Index and Magnetic Resonance Imaging in Patients with Cystic Fibrosis and One or Two F508del Alleles. Am J Respir Crit Care Med 2022; 206:311-320. [PMID: 35536314 DOI: 10.1164/rccm.202201-0219oc] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE We recently demonstrated that triple combination CFTR modulator therapy with elexacaftor/tezacaftor/ivacaftor (ELX/TEZ/IVA) improves CFTR function in airway and intestinal epithelia to 40 to 50% of normal in patients with cystic fibrosis (CF) with one or two F508del alleles. In previous studies, this improvement of CFTR function was shown to improve clinical outcomes, however, effects on the lung clearance index (LCI) determined by multiple breath washout and abnormalities in lung morphology and perfusion detected by magnetic resonance imaging (MRI) have not been studied. OBJECTIVES To examine the effect of ELX/TEZ/IVA on LCI and lung MRI scores in patients with CF and one or two F508del alleles aged 12 years and older. METHODS This prospective, observational, multicenter, post-approval study assessed LCI and lung MRI scores before and 8-16 weeks after initiation of ELX/TEZ/IVA. MEASUREMENTS AND MAIN RESULTS A total of 91 patients with CF including 45 heterozygous for F508del and a minimal function mutation (MF) and 46 homozygous for F508del were enrolled in this study. Treatment with ELX/TEZ/IVA improved LCI in F508del/MF (-2.4;IQR, -3.7 - -1.1;P<0.001) and F508del homozygous (-1.4;IQR, -2.4 - -0.4;P<0.001) patients. Further, ELX/TEZ/IVA improved the MRI global score in F508del/MF (-6.0;IQR, -11.0 - -1.3;P<0.001) and F508del homozygous (-6.5;IQR, -11.0 - -1.3;P<0.001) patients. CONCLUSIONS Our data demonstrate that improvement of CFTR function by ELX/TEZ/IVA improves lung ventilation and abnormalities in lung morphology including airway mucus plugging and wall thickening in adolescent and adult patients with CF and one or two F508del alleles in a real-world, post-approval setting.
Collapse
Affiliation(s)
- Simon Y Graeber
- Charité Universitätsmedizin Berlin, 14903, Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine and Cystic Fibrosis Center, Berlin, Germany.,Berlin Institute of Health at Charité, 522475, Berlin, Germany.,German Center for Lung Research, 542891, associated partner site, Berlin, Germany
| | - Diane M Renz
- Hannover Medical School, 9177, Department for Radiology, Hannover, Germany
| | - Mirjam Stahl
- Charité Universitätsmedizin Berlin, 14903, Department of Pediatric Pulmonology, Immunology and Critical Care Medicine and Cystic Fibrosis Center, Berlin, Germany.,Berlin Institute of Health at Charité, 522475, Berlin, Germany.,German Center for Lung Research, 542891, associated partner site, Berlin, Germany
| | - Sophia T Pallenberg
- Hannover Medical School, 9177, Department of Pediatric Pneumology, Allergology and Neonatology, Hannover, Germany.,German Center for Lung Research, 542891, Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Hannover, Germany
| | - Olaf Sommerburg
- Heidelberg University, 9144, Division of Pediatric Pulmonology & Allergy and Cystic Fibrosis Center, Department of Pediatrics, Heidelberg, Germany.,German Center for Lung Research, 542891, Department of Translational Pulmonology, Translational Lung Research Center Heidelberg (TLRC), Heidelberg, Germany
| | - Lutz Naehrlich
- Justus Liebig Universitat Giessen, 9175, Department of Pediatrics, Giessen, Germany.,German Center for Lung Research, 542891, Universities Giessen and Marburg Lung Center (UGMLC), Giessen, Germany
| | - Julian Berges
- Heidelberg University, 9144, Division of Pediatric Pulmonology & Allergy and Cystic Fibrosis Center, Department of Pediatrics, Heidelberg, Germany.,German Center for Lung Research, 542891, Department of Translational Pulmonology, Translational Lung Research Center Heidelberg (TLRC), Heidelberg, Germany
| | - Martha Dohna
- Hannover Medical School, 9177, Department for Radiology, Hannover, Germany
| | - Felix C Ringshausen
- Hannover Medical School, 9177, Department for Pneumology, Hannover, Germany.,German Center for Lung Research, 542891, Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Hannover, Germany
| | - Felix Doellinger
- Charité Universitätsmedizin Berlin, 14903, Department of Radiology, Berlin, Germany
| | - Constanze Vitzthum
- Charité Universitätsmedizin Berlin, 14903, Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine and Cystic Fibrosis Center, Berlin, Germany.,German Center for Lung Research, 542891, associated partner site, Berlin, Germany
| | - Jobst Röhmel
- Charité Universitätsmedizin Berlin, 14903, Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine and Cystic Fibrosis Center, Berlin, Germany.,German Center for Lung Research, 542891, associated partner site, Berlin, Germany
| | - Christine Allomba
- Charité Universitätsmedizin Berlin, 14903, Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine and Cystic Fibrosis Center, Berlin, Germany.,German Center for Lung Research, 542891, associated partner site, Giessen, Germany
| | - Susanne Hämmerling
- University of Heidelberg, 9144, Department of Pediatrics, Division of Pediatric Pulmonology and Allergy and Cystic Fibrosis Center, Heidelberg, Germany
| | - Sandra Barth
- Justus Liebig Universitat Giessen, 9175, Department of Pediatrics, Giessen, Germany.,German Center for Lung Research, 542891, Universities Giessen and Marburg Lung Center (UGMLC), Giessen, Germany
| | | | - Mark O Wielpütz
- Heidelberg University, 9144, Department of Diagnostic and Interventional Radiology, Heidelberg, Germany.,German Center for Lung Research, 542891, Department of Translational Pulmonology, Translational Lung Research Center Heidelberg (TLRC), Heidelberg, Germany
| | - Gesine Hansen
- Hannover Medical School, 9177, Department for Pediatric Pneumology, Allergology and Neonatology, Hannover, Germany.,German Center for Lung Research, 542891, German Center for Lung Research, Biomedical Research in Endstage and Obstructive Lung Disease (BREATH), Hannover, Germany
| | - Jens Vogel-Claussen
- Hannover Medical School, 9177, Department for Radiology, Hannover, Germany.,Hannover Medical School, 9177, Department for Pediatric Pneumology, Hannover, Germany
| | - Burkhard Tümmler
- Hannover Medical School, 9177, Department for Pediatric Pneumology, Hannover, Germany.,German Center for Lung Research, 542891, Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Hannover, Germany
| | - Marcus A Mall
- Charité Universitätsmedizin Berlin, 14903, Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine and Cystic Fibrosis Center, Berlin, Germany.,Berlin Institute of Health at Charité, 522475, Berlin, Germany.,German Center for Lung Research, 542891, associated partner site, Berlin, Germany;
| | - Anna-Maria Dittrich
- Hannover Medical School, 9177, Department for Pediatric Pneumology, Allergology and Neonatology, Hannover, Germany.,German Center for Lung Research, 542891, Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Hannover, Germany
| |
Collapse
|
39
|
Brusa I, Sondo E, Falchi F, Pedemonte N, Roberti M, Cavalli A. Proteostasis Regulators in Cystic Fibrosis: Current Development and Future Perspectives. J Med Chem 2022; 65:5212-5243. [PMID: 35377645 PMCID: PMC9014417 DOI: 10.1021/acs.jmedchem.1c01897] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In cystic fibrosis (CF), the deletion of phenylalanine 508 (F508del) in the CF transmembrane conductance regulator (CFTR) leads to misfolding and premature degradation of the mutant protein. These defects can be targeted with pharmacological agents named potentiators and correctors. During the past years, several efforts have been devoted to develop and approve new effective molecules. However, their clinical use remains limited, as they fail to fully restore F508del-CFTR biological function. Indeed, the search for CFTR correctors with different and additive mechanisms has recently increased. Among them, drugs that modulate the CFTR proteostasis environment are particularly attractive to enhance therapy effectiveness further. This Perspective focuses on reviewing the recent progress in discovering CFTR proteostasis regulators, mainly describing the design, chemical structure, and structure-activity relationships. The opportunities, challenges, and future directions in this emerging and promising field of research are discussed, as well.
Collapse
Affiliation(s)
- Irene Brusa
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy.,Computational & Chemical Biology, Istituto Italiano di Tecnologia, 16163 Genova, Italy
| | - Elvira Sondo
- UOC Genetica Medica, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy
| | | | | | - Marinella Roberti
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy
| | - Andrea Cavalli
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy.,Computational & Chemical Biology, Istituto Italiano di Tecnologia, 16163 Genova, Italy
| |
Collapse
|
40
|
Gambazza S, Ambrogi F, Carta F, Moroni L, Russo M, Brivio A, Colombo C. Lung clearance index to characterize clinical phenotypes of children and adolescents with cystic fibrosis. BMC Pulm Med 2022; 22:122. [PMID: 35365111 PMCID: PMC8976307 DOI: 10.1186/s12890-022-01903-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 03/11/2022] [Indexed: 12/31/2022] Open
Abstract
Background Lung clearance index (LCI) is accepted as an early marker of lung disease in cystic fibrosis (CF), however the utility of LCI to identify subgroups of CF disease in the paediatric age group has never been explored. The aim of the study was to characterize phenotypes of children with CF using LCI as a marker of ventilation inhomogeneity and to investigate whether these phenotypes distinguished patients based on time to pulmonary exacerbation (PE).
Methods Data were collected on patients with CF aged < 18 years old, attending the CF Center of Milan during outpatient follow-up visits between October 2014 and September 2019. Cluster analysis using agglomerative nesting hierarchical method was performed to generate distinct phenotypes. Time-to-recurrent event analysis investigated association of phenotypes with PE. Results We collected 313 multiple breath washout tests on 125 children aged 5.5–16.8 years. Cluster analysis identified two divergent phenotypes in children and adolescents of same age, presenting with almost normal FEV1 but with substantial difference in markers of ventilation inhomogeneity (mean LCI difference of 3.4, 95% Confidence Interval [CI] 2.6–4.2). A less severe phenotype was associated with a lower risk of PE relapse (Hazard Ratio 0.45, 95% CI 0.34–0.62). Conclusions LCI is useful in clinical practice to characterize distinct phenotypes of children and adolescents with mild/normal FEV1. A less severe phenotype translates into a lower risk of PE relapse. Supplementary Information The online version contains supplementary material available at 10.1186/s12890-022-01903-5.
Collapse
Affiliation(s)
- Simone Gambazza
- Department of Clinical Sciences and Community Health, Laboratory of Medical Statistics, Biometry and Epidemiology "G. A. Maccacaro", University of Milan, Milan, Italy. .,Healthcare Professions Department, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.
| | - Federico Ambrogi
- Department of Clinical Sciences and Community Health, Laboratory of Medical Statistics, Biometry and Epidemiology "G. A. Maccacaro", University of Milan, Milan, Italy
| | - Federica Carta
- Healthcare Professions Department, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Cystic Fibrosis Centre, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Laura Moroni
- Cystic Fibrosis Centre, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Maria Russo
- Cystic Fibrosis Centre, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Anna Brivio
- Healthcare Professions Department, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Cystic Fibrosis Centre, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Carla Colombo
- Cystic Fibrosis Centre, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| |
Collapse
|
41
|
Barr HL, Bihouee T, Zwitserloot AM. A year in review: Real world evidence, functional monitoring and emerging therapeutics in 2021. J Cyst Fibros 2022; 21:191-196. [PMID: 35272931 PMCID: PMC8900606 DOI: 10.1016/j.jcf.2022.02.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 02/23/2022] [Indexed: 12/20/2022]
Affiliation(s)
- H L Barr
- Nottingham University Hospitals NHS Trust, Nottingham, United Kingdom; Nottingham Respiratory Biomedical Research Centre, University of Nottingham, Nottingham, United Kingdom.
| | - T Bihouee
- Chronic Childhood Diseases unit, Pediatric Department, Nantes University Hospital, Nantes, France
| | - A M Zwitserloot
- University of Groningen, Department of Pediatric Pulmonology and Pediatric Allergy, University Medical Center Groningen, Beatrix Children's Hospital, Groningen, The Netherlands; University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, The Netherlands
| |
Collapse
|
42
|
Gabel ME, Fox CK, Grimes RA, Lowman JD, McDonald CM, Stallings VA, Michel SH. Overweight and cystic fibrosis: An unexpected challenge. Pediatr Pulmonol 2022; 57 Suppl 1:S40-S49. [PMID: 34738328 DOI: 10.1002/ppul.25748] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 10/06/2021] [Accepted: 10/18/2021] [Indexed: 02/06/2023]
Abstract
Achieving a healthy weight balance has been a central focus of care for people who have cystic fibrosis (CF). Over the years, the emphasis has primarily been on promoting weight gain to optimize pulmonary outcomes. With continued improvements in CF care, including highly effective CF modulators available for many people, the CF community is now experiencing a new challenge: addressing the concern that some people are gaining weight excessively. While at this time, we do not know to what extent overweight and obesity will affect health outcomes for people with CF, it is likely that excessive weight gain may have negative health impacts similar to those seen in the general population. In this paper, we review the history of nutritional guidelines for people with CF, as well as more recent trends toward overweight and obesity for some. A multidisciplinary approach is needed to collaboratively start the oftentimes difficult conversation regarding excessive weight gain, and to identify resources to help people achieve and maintain a healthy weight through diet, exercise, and behavioral modification.
Collapse
Affiliation(s)
- Megan E Gabel
- Department of Pediatrics, University of Rochester Medical Center, Rochester, New York, USA
| | - Claudia K Fox
- Department of Pediatrics, Center for Pediatric Obesity Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Rachel A Grimes
- Department of Psychiatry, Cook Children's Medical Center, Fort Worth, Texas, USA
| | - John D Lowman
- Department of Physical Therapy, Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Catherine M McDonald
- Department of Clinical Nutrition, Primary Children's Hospital, Salt Lake City, Utah, USA
| | - Virginia A Stallings
- Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Suzanne H Michel
- Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| |
Collapse
|
43
|
Goss CH, Fajac I, Jain R, Seibold W, Gupta A, Hsu MC, Sutharsan S, Davies JC, Mall MA. Efficacy and safety of inhaled ENaC inhibitor BI 1265162 in patients with cystic fibrosis: BALANCE-CF 1, a randomised, phase II study. Eur Respir J 2022; 59:2100746. [PMID: 34385272 PMCID: PMC8850685 DOI: 10.1183/13993003.00746-2021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 06/19/2021] [Indexed: 12/05/2022]
Abstract
BACKGROUND Inhibition of the epithelial sodium channel (ENaC) in cystic fibrosis (CF) airways provides a mutation-agnostic approach that could improve mucociliary clearance in all CF patients. BI 1265162 is an ENaC inhibitor with demonstrated pre-clinical efficacy and safety already demonstrated in humans. OBJECTIVE We present results from BALANCE-CFTM 1, a phase II, placebo-controlled, randomised, double-blind study of four dose levels of BI 1265162 versus placebo for 4 weeks on top of standard of care in adults and adolescents with CF. RESULTS Initially, 28 randomised subjects (BI 1265162 200 µg twice daily n=14, placebo twice daily n=14) were assessed at an interim futility analysis. Compared with placebo, numerical changes of -0.8% (95% CI -6.6 to 4.9%) in percentage predicted forced expiratory volume in 1s (ppFEV1) and +2.1 units (95% CI -2.4 to 6.5 units) in lung clearance index (LCI) were observed in the active group, meeting a pre-defined stopping rule; accordingly, the study was terminated. Recruitment had continued during the interim analysis and pending results; 24 patients were added across three dose levels and placebo. The final results including these patients (+1.5% ppFEV1, 200 µg twice-daily dose versus placebo) were not supportive of relevant clinical effect. Furthermore, LCI change was not supportive, although interpretation was limited due to insufficient traces meeting quality criteria. A 9.4-point improvement in the Cystic Fibrosis Questionnaire - Revised Respiratory Domain was observed in the 200 µg twice daily dose group versus placebo. BI 1265162 up to 200 µg twice daily was safe and well-tolerated. Pharmacokinetics were similar to those in healthy volunteers. CONCLUSION BI 1265162 was safe, but did not demonstrate a potential for clinical benefit. Development has been terminated.
Collapse
Affiliation(s)
- Christopher H Goss
- Dept of Medicine, Dept of Pediatrics, University of Washington, Seattle Children's Hospital and Research Institute, Seattle, WA, USA
| | | | - Raksha Jain
- Dept of Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | | | | - Ming-Chi Hsu
- Boehringer Ingelheim, Shanghai, China
- Shanghai Junshi Biosciences Co. Ltd, Shanghai, China
| | - Sivagurunathan Sutharsan
- Division for Cystic Fibrosis, Dept of Pulmonary Medicine, University Medicine Essen - Ruhrlandklinik, Essen, Germany
| | - Jane C Davies
- National Heart and Lung Institute, Imperial College London, London, UK
- Paediatric Respiratory Medicine, Royal Brompton and Harefield Hospitals, London, UK
| | - Marcus A Mall
- Dept of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
- German Center for Lung Research (DZL), associated partner site, Berlin, Germany
| |
Collapse
|
44
|
Li Q, Liu S, Ma X, Yu J. Effectiveness and Safety of Cystic Fibrosis Transmembrane Conductance Regulator Modulators in Children With Cystic Fibrosis: A Meta-Analysis. Front Pediatr 2022; 10:937250. [PMID: 35844763 PMCID: PMC9276987 DOI: 10.3389/fped.2022.937250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 05/27/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND AND AIM Cystic fibrosis (CF) is a genetic disease that is difficult to treat and caused by dysfunction of the cystic fibrosis transmembrane conductance regulator (CFTR) protein. Small molecules have been used to treat the symptom caused by CFTR mutations by restoring CFTR protein function. However, the data on children with CF are scarce. This meta-analysis aimed to evaluate the effectiveness and safety of this therapy in children diagnosed with CF. MATERIALS AND METHODS Relevant studies were identified through searching medical databases before April 1, 2022. The primary outcomes of ppFEV1, lung clearance index2.5 (LCI2.5), sweat chloride concentration (SwCI), and Cystic Fibrosis Questionnaire-Revised (CFQ-R) score were pooled and analyzed. The secondary outcomes were nutritional status (weight, BMI, stature, and their z-score) and adverse events under therapy. RESULTS A total of twelve studies were included. Compared with the placebo group, the pooled outcome of the ppFEV1, LCI2.5, SwCI, and CFQ-R score were improved by 7.91 {[95% confidence interval (CI), 3.71-12.12], -1.00 (95% CI, -1.38 to -0.63), -35.22 (95% CI, -55.51 to -14.92), and 4.45 (95% CI, 2.31-6.59), respectively}. Compared with the placebo group, the pooled result of the change in weight was improved by 1.53 (95% CI, 0.42-2.63). All the aforementioned results were also improved in single-arm studies. No clear differences in adverse events were found between CFTR modulator therapy and the placebo group. CONCLUSION CFTR modulators could improve multiaspect function in children with CF and result in comparable adverse events.
Collapse
Affiliation(s)
- Qiyu Li
- Department of Pediatrics, General Hospital of Northern Theater Command, Shenyang, China
| | - Siyuan Liu
- Department of Pediatrics, General Hospital of Northern Theater Command, Shenyang, China
| | - Xuemei Ma
- Department of Pediatrics, General Hospital of Northern Theater Command, Shenyang, China
| | - Jiaping Yu
- Department of Pediatrics, General Hospital of Northern Theater Command, Shenyang, China
| |
Collapse
|
45
|
Caterini JE, Ratjen F, Barker AR, Williams CA, Rendall K, Schneiderman JE, Wells GD. Exercise intolerance in cystic fibrosis-the role of CFTR modulator therapies. J Cyst Fibros 2021; 21:282-292. [PMID: 34955387 DOI: 10.1016/j.jcf.2021.11.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 10/18/2021] [Accepted: 11/20/2021] [Indexed: 12/11/2022]
Abstract
Exercise intolerance is common in people with CF (pwCF), but not universal among all individuals. While associated with disease prognosis, exercise intolerance is not simply a reflection of the degree of lung disease. In people with severe CF, respiratory limitations may contribute more significantly to impaired exercise capacity than in those with mild-moderate CF. At all levels of disease severity, there are peripheral factors e.g., abnormal macro- and micro-vascular function that impair blood flow and reduce oxygen extraction, and mitochondrial defects that diminish metabolic efficiency. We discuss advances in understanding the central and peripheral mechanisms underlying exercise intolerance in pwCF. Exploring both the central and peripheral factors that contribute to exercise intolerance in CF can help inform the development of new therapeutic targets, as well as help define prognostic criteria.
Collapse
Affiliation(s)
- Jessica E Caterini
- Translational Medicine Program, SickKids Research Institute, Toronto, ON M5G 0A4, Canada; Queen's Medical School, Kingston, ON K7L 3N6, Canada
| | - Felix Ratjen
- Translational Medicine Program, SickKids Research Institute, Toronto, ON M5G 0A4, Canada; Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, ON M5G 1X8, Canada; Division of Respiratory Medicine, The Hospital for Sick Children, University of Toronto, Toronto, ON M5G 1X8, Canada
| | - Alan R Barker
- Children's Health and Exercise Research Centre, Sport and Health Sciences, University of Exeter, Exeter EX1 2LU, UK
| | - Craig A Williams
- Children's Health and Exercise Research Centre, Sport and Health Sciences, University of Exeter, Exeter EX1 2LU, UK
| | - Kate Rendall
- Translational Medicine Program, SickKids Research Institute, Toronto, ON M5G 0A4, Canada
| | - Jane E Schneiderman
- Division of Respiratory Medicine, The Hospital for Sick Children, University of Toronto, Toronto, ON M5G 1X8, Canada; Faculty of Kinesiology & Physical Education, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Greg D Wells
- Translational Medicine Program, SickKids Research Institute, Toronto, ON M5G 0A4, Canada.
| |
Collapse
|
46
|
Triple combination cystic fibrosis transmembrane conductance regulator modulator therapy in the real world - opportunities and challenges. Curr Opin Pulm Med 2021; 27:554-566. [PMID: 34420018 DOI: 10.1097/mcp.0000000000000819] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
PURPOSE OF REVIEW The purpose of this review is to describe on-going and upcoming real-world studies that will aid the cystic fibrosis (CF) community in understanding the long-term efficacy, safety and challenges in utilizing this therapy and managing care. RECENT FINDINGS The triple combination of elexacaftor, tezacaftor and ivacaftor (ETI) has been demonstrated to improve lung function, weight and quality of life in children and adults with CF with at least one copy of Phe508del. Treatment with ETI will potentially be available for approximately 90% of the CF population and change the face of CF. SUMMARY In spite of early demonstrations of short-term efficacy and safety, for medications that may be given for much of someone's life, continued assessment of these outcomes is necessary. Furthermore, the CF community must evaluate and address the issues that arise with increased longevity including parenthood, preventive care management and the potential comorbidities of aging.
Collapse
|
47
|
Meoli A, Fainardi V, Deolmi M, Chiopris G, Marinelli F, Caminiti C, Esposito S, Pisi G. State of the Art on Approved Cystic Fibrosis Transmembrane Conductance Regulator ( CFTR) Modulators and Triple-Combination Therapy. Pharmaceuticals (Basel) 2021; 14:ph14090928. [PMID: 34577628 PMCID: PMC8471029 DOI: 10.3390/ph14090928] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 09/09/2021] [Accepted: 09/10/2021] [Indexed: 11/23/2022] Open
Abstract
Cystic fibrosis (CF) is the most common life-limiting inherited disease in Caucasian populations, affecting approximately 80,000 people worldwide. CF is a complex multi-organ monogenic autosomal recessive disorder caused by a mutation in cystic fibrosis transmembrane conductance regulator (CFTR) gene. Since the discovery of the CFTR gene in 1989, more than 2000 mutations have been identified so far and about 240 can cause CF. Until recently, the treatment for CF was aimed to prevent and manage the manifestations of CFTR dysfunction, primarily recurrent pulmonary infections and pancreatic exocrine failure. Over the past few decades, the therapeutic approach to CF has been revolutionized by the development of a new class of small molecules called CFTR modulators that target specific defects caused by mutations in the CFTR gene. CFTR modulators have been shown to change profoundly the clinical course of the CF, leading to meaningful improvements in the lives of a large proportion of people of CF heterozygous for F508del, especially if started in young children. Further studies are needed to extend the use of triple CFTR modulation therapy also for young children in order to prevent the irreversible effects of the disease and for patients with very rare mutations with a personalized approach to treatment.
Collapse
Affiliation(s)
- Aniello Meoli
- Paediatric Clinic, Department of Medicine and Surgery, University Hospital of Parma, Via Gramsci 14, 43126 Parma, Italy; (A.M.); (V.F.); (M.D.); (G.C.); (F.M.); (G.P.)
| | - Valentina Fainardi
- Paediatric Clinic, Department of Medicine and Surgery, University Hospital of Parma, Via Gramsci 14, 43126 Parma, Italy; (A.M.); (V.F.); (M.D.); (G.C.); (F.M.); (G.P.)
| | - Michela Deolmi
- Paediatric Clinic, Department of Medicine and Surgery, University Hospital of Parma, Via Gramsci 14, 43126 Parma, Italy; (A.M.); (V.F.); (M.D.); (G.C.); (F.M.); (G.P.)
| | - Giulia Chiopris
- Paediatric Clinic, Department of Medicine and Surgery, University Hospital of Parma, Via Gramsci 14, 43126 Parma, Italy; (A.M.); (V.F.); (M.D.); (G.C.); (F.M.); (G.P.)
| | - Francesca Marinelli
- Paediatric Clinic, Department of Medicine and Surgery, University Hospital of Parma, Via Gramsci 14, 43126 Parma, Italy; (A.M.); (V.F.); (M.D.); (G.C.); (F.M.); (G.P.)
| | - Caterina Caminiti
- Research and Innovation Unit, University Hospital of Parma, Via Gramsci 14, 43126 Parma, Italy;
| | - Susanna Esposito
- Paediatric Clinic, Department of Medicine and Surgery, University Hospital of Parma, Via Gramsci 14, 43126 Parma, Italy; (A.M.); (V.F.); (M.D.); (G.C.); (F.M.); (G.P.)
- Correspondence:
| | - Giovanna Pisi
- Paediatric Clinic, Department of Medicine and Surgery, University Hospital of Parma, Via Gramsci 14, 43126 Parma, Italy; (A.M.); (V.F.); (M.D.); (G.C.); (F.M.); (G.P.)
| |
Collapse
|
48
|
Abstract
Rationale: Ivacaftor’s clinical effects in the residual function mutations 3849 + 10kb C→T and D1152H warrant further characterization. Objectives: To evaluate ivacaftor’s effect in people with cystic fibrosis aged ≥6 years with 3849 + 10kb C→T or D1152H residual function mutations and to explore the correlation between ivacaftor-induced organoid-based cystic fibrosis transmembrane conductance regulator function measurements and clinical response to ivacaftor. Methods: Participants were randomized (1:1) in this placebo-controlled crossover study; each treatment sequence included two 8-week treatments with an 8-week washout period. The primary endpoint was absolute change in lung clearance index2.5 from baseline through Week 8. Additional endpoints included lung function, patient-reported outcomes, and in vitro intestinal organoid–based measurements of ivacaftor-induced cystic fibrosis transmembrane conductance regulator function. Results: Of 38 participants, 37 completed the study. The primary endpoint was met; the Bayesian posterior probability of improvement in lung clearance index2.5 with ivacaftor versus placebo was >99%. Additional endpoints improved with ivacaftor. Safety findings were consistent with ivacaftor’s known safety profile. Dose-dependent swelling was observed in 23 of 25 viable organoid cultures with ivacaftor treatment. Correlations between ivacaftor-induced organoid swelling and clinical endpoints were negligible to low. Conclusions: In people with cystic fibrosis aged ≥6 years with a 3849 + 10kb C→T or D1152H mutation, ivacaftor treatment improved clinical endpoints compared with placebo; however, there was no correlation between organoid swelling and change in clinical endpoints. The organoid assay may assist in identification of ivacaftor-responsive mutations but in this study did not predict magnitude of clinical benefit for individual people with cystic fibrosis with these two mutations. Clinical trial registered with ClinicalTrials.gov (NCT03068312).
Collapse
|
49
|
Effects of Lumacaftor-Ivacaftor on Lung Clearance Index, Magnetic Resonance Imaging, and Airway Microbiome in Phe508del Homozygous Patients with Cystic Fibrosis. Ann Am Thorac Soc 2021; 18:971-980. [PMID: 33600745 DOI: 10.1513/annalsats.202008-1054oc] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Rationale: Previous studies showed that lumacaftor-ivacaftor therapy results in partial rescue of CFTR (cystic fibrosis [CF] transmembrane conductance regulator) activity and a moderate improvement of spirometry in Phe508del homozygous patients with CF. However, the effects of lumacaftor-ivacaftor on lung clearance index (LCI), lung morphology and perfusion detected by chest magnetic resonance imaging (MRI), and effects on the airway microbiome and inflammation remain unknown. Objectives: To investigate the effects of lumacaftor-ivacaftor on LCI, lung MRI scores, and airway microbiome and inflammation. Methods: In this prospective observational study we assessed clinical outcomes including spirometry and body mass index, LCI, lung MRI scores, sputum microbiome, and proinflammatory cytokines in 30 Phe508del homozygous patients with CF 12 years and older before and 8-16 weeks after initiation of lumacaftor-ivacaftor therapy. Results: Lumacaftor-ivacaftor had no effects on forced expiratory volume in 1 second (FEV1% predicted) (1.7%; 95% confidence interval [CI], -1.0% to 4.3%; P = 0.211) but improved LCI (-1.6; 95% CI, -2.6 to -0.5; P < 0.01) and MRI morphology (-1.3; 95% CI, -2.3 to -0.3; P < 0.05) and perfusion score (-1.2; 95% CI, -2.3 to -0.2; P < 0.05) in our study cohort. Furthermore, lumacaftor-ivacaftor decreased the total bacterial load (-1.8; 95% CI, -3.3 to -0.34; P < 0.05) and increased the Shannon diversity of the airway microbiome (0.4; 95% CI, 0.1 to 0.8; P < 0.05), and reduced IL-1β (interleukin-1β) concentration (median change, -324.2 pg/ml; 95% CI, -938.7 to 290.4 pg/ml; P < 0.05) in sputum of Phe508del homozygous patients. Conclusions: This study shows that lumacaftor-ivacaftor has beneficial effects on lung ventilation, morphology, and perfusion, as well as on the airway microbiome and inflammation in Phe508del homozygous patients. Our results suggest that LCI and MRI may be more sensitive than FEV1% predicted to detect response to CFTR modulator therapy in patients with chronic CF lung disease. Clinical trial registered with ClinicalTrials.gov (NCT02807415).
Collapse
|
50
|
Zemanick ET, Taylor-Cousar JL, Davies J, Gibson RL, Mall MA, McKone EF, McNally P, Ramsey BW, Rayment JH, Rowe SM, Tullis E, Ahluwalia N, Chu C, Ho T, Moskowitz SM, Noel S, Tian S, Waltz D, Weinstock TG, Xuan F, Wainwright CE, McColley SA. 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. Am J Respir Crit Care Med 2021; 203:1522-1532. [PMID: 33734030 PMCID: PMC8483230 DOI: 10.1164/rccm.202102-0509oc] [Citation(s) in RCA: 174] [Impact Index Per Article: 43.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
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).
Collapse
Affiliation(s)
- Edith T Zemanick
- Department of Pediatrics, University of Colorado Anschutz Medical Campus and Children's Hospital Colorado, Aurora, Colorado
| | | | - Jane Davies
- National Heart and Lung Institute, Imperial College London, National Institute for Health Research Imperial Biomedical Research Centre and Royal Brompton and Harefield National Health Service Foundation Trust, London, United Kingdom
| | - Ronald L Gibson
- University of Washington/Seattle Children's Hospital, Seattle, Washington
| | - Marcus A Mall
- Department of Pediatric Pulmonology, Immunology and Intensive Care Medicine, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health, Berlin, Germany.,German Center for Lung Research, Berlin, Germany
| | - Edward F McKone
- St. Vincent's University Hospital and University College, Dublin, Ireland
| | - Paul McNally
- Children's Health Ireland and Royal College of Surgeons in Ireland University of Medicine and Health Sciences, Dublin, Ireland
| | - Bonnie W Ramsey
- University of Washington/Seattle Children's Hospital, Seattle, Washington
| | - Jonathan H Rayment
- British Columbia Children's Hospital, Vancouver, British Columbia, Canada
| | - Steven M Rowe
- University of Alabama at Birmingham, Birmingham, Alabama
| | | | - Neil Ahluwalia
- Vertex Pharmaceuticals Incorporated, Boston, Massachusetts
| | - Chenghao Chu
- Vertex Pharmaceuticals Incorporated, Boston, Massachusetts
| | - Thang Ho
- Vertex Pharmaceuticals Incorporated, Boston, Massachusetts
| | | | - Sabrina Noel
- Vertex Pharmaceuticals Incorporated, Boston, Massachusetts
| | - Simon Tian
- Vertex Pharmaceuticals Incorporated, Boston, Massachusetts
| | - David Waltz
- Vertex Pharmaceuticals Incorporated, Boston, Massachusetts
| | | | - Fengjuan Xuan
- Vertex Pharmaceuticals Incorporated, Boston, Massachusetts
| | | | - Susanna A McColley
- Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois; and.,Northwestern University Feinberg School of Medicine, Chicago, Illinois
| |
Collapse
|