1
|
Zemanick ET, Ramsey B, Sands D, McKone EF, Fajac I, Taylor-Cousar JL, Mall MA, Konstan MW, Nair N, Zhu J, Arteaga-Solis E, Van Goor F, McGarry L, Prieto-Centurion V, Sosnay PR, Bozic C, Waltz D, Mayer-Hamblett N. Sweat chloride reflects CFTR function and correlates with clinical outcomes following CFTR modulator treatment. J Cyst Fibros 2025:S1569-1993(24)01854-X. [PMID: 39755444 DOI: 10.1016/j.jcf.2024.12.006] [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: 08/11/2024] [Revised: 12/06/2024] [Accepted: 12/18/2024] [Indexed: 01/06/2025]
Abstract
BACKGROUND Highly effective CFTR modulators improve CFTR function and lead to dramatic improvements in health outcomes in many people with cystic fibrosis (pwCF). The relationship between measures of CFTR function, such as sweat chloride concentration, and clinical outcomes in pwCF treated with CFTR modulators is poorly defined. We conducted analyses to better understand the relationships between sweat chloride and CFTR function in vitro, and between sweat chloride and clinical outcomes following CFTR modulator treatment. METHODS Mean sweat chloride values in healthy people, CF carriers, and pwCF treated with CFTR modulators at different doses were compared to chloride transport in corresponding human bronchial epithelial (HBE) cells. A pooled analysis of phase 3 CFTR modulator studies was performed to evaluate the relationship between attained values of sweat chloride and improvements in lung function, body mass index (BMI), patient reported outcomes, pulmonary exacerbations, and lung function change over time. RESULTS Sweat chloride concentrations in vivo correlated strongly with CFTR-dependent chloride current in HBE cells in vitro. Sweat chloride values of <30 mmol/L and ≥30 to <60 mmol/L in pwCF following CFTR modulator treatment were associated with better clinical outcomes than sweat chloride ≥60 to <80 mmol/L and ≥80 mmol/L. CONCLUSIONS In pwCF treated with CFTR modulators, lower sweat chloride levels (reflecting greater CFTR function) are associated with better clinical outcomes. These results support the therapeutic strategy of further restoring CFTR function towards normal, as reflected in lowering sweat chloride to below the diagnostic threshold for CF (<60 mmol/L) and to normal (<30 mmol/L), with CFTR modulators.
Collapse
Affiliation(s)
- Edith T Zemanick
- Department of Pediatrics, University of Colorado Anschutz Medical Campus and Children's Hospital Colorado, Aurora, CO, USA.
| | - Bonnie Ramsey
- Cystic Fibrosis Therapeutics Development Network Coordinating Center, Seattle Children's Hospital, Seattle, WA, USA; Department of Pediatrics, University of Washington, Seattle, WA, USA
| | - Dorota Sands
- Cystic Fibrosis Department, Institute of Mother and Child, Warsaw, Poland
| | - Edward F McKone
- St. Vincent's University Hospital and University College, Dublin, Ireland
| | | | - Jennifer L Taylor-Cousar
- Departments of Internal Medicine and Pediatrics, Pulmonary Divisions National Jewish Health, Denver, CO, USA
| | - 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 German Center for Lung Research, Associated Partner, Berlin, Germany and German Center for Child and Adolescent Health (DZKJ), partner site Berlin, Berlin, Germany
| | - Michael W Konstan
- Department of Pediatrics, Case Western Reserve University School of Medicine and Rainbow Babies and Children's Hospital, Cleveland, OH, USA
| | - Nitin Nair
- Vertex Pharmaceuticals Incorporated, Boston, MA, USA
| | - Jiaqiang Zhu
- Vertex Pharmaceuticals Incorporated, Boston, MA, USA
| | | | | | - Lisa McGarry
- Vertex Pharmaceuticals Incorporated, Boston, MA, USA
| | | | | | - Carmen Bozic
- Vertex Pharmaceuticals Incorporated, Boston, MA, USA
| | - David Waltz
- Vertex Pharmaceuticals Incorporated, Boston, MA, USA
| | - Nicole Mayer-Hamblett
- Cystic Fibrosis Therapeutics Development Network Coordinating Center, Seattle Children's Hospital, Seattle, WA, USA; Department of Pediatrics, University of Washington, Seattle, WA, USA; Department of Biostatistics, University of Washington, Seattle, WA, USA
| |
Collapse
|
2
|
Stahl M, Roehmel J, Eichinger M, Doellinger F, Naehrlich L, Kopp MV, Dittrich AM, Sommerburg O, Ray P, Maniktala A, Xu T, Conner S, Joshi A, Mascia M, Wielpütz MO, Mall MA. Long-Term Impact of Lumacaftor/Ivacaftor Treatment on Cystic Fibrosis Disease Progression in Children 2-5 Years of Age Homozygous for F508del-CFTR: A Phase 2, Open-Label Clinical Trial. Ann Am Thorac Soc 2024; 21:1550-1559. [PMID: 39173175 DOI: 10.1513/annalsats.202402-201oc] [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: 02/26/2024] [Accepted: 08/22/2024] [Indexed: 08/24/2024] Open
Abstract
Rationale: Clinical trials show that lumacaftor/ivacaftor (LUM/IVA) treatment has the potential to modify early cystic fibrosis (CF) disease progression in children as young as 2 years of age. Objectives: To assess the long-term impact of LUM/IVA treatment on CF disease progression in children aged 2-5 years. Methods: This phase 2 trial had two parts: part 1, a 48-week, randomized, double-blind, placebo-controlled study of LUM/IVA in children aged 2-5 years (previously reported) was followed by a 48-week open-label treatment period in which all children received LUM/IVA (part 2; reported here). Endpoints assessed in part 2 included absolute changes from baseline in chest magnetic resonance imaging (MRI) global score at Week 96; weight-for-age, stature-for-age, and body mass index (BMI)-for-age z-scores at Week 96; lung clearance index based on lung volume turnover required to reach 2.5% of starting N2 concentration (LCI2.5) through Week 96; chest MRI morphological score, chest MRI perfusion score, weight, stature, BMI, and microbiology cultures (oropharyngeal swabs) at Week 96; sweat chloride, amount of immunoreactive trypsinogen, fecal elastase-1 concentration, and fecal calprotectin through Week 96; and number of pulmonary exacerbations, time to first pulmonary exacerbation, and number of CF-related hospitalizations. Results: Forty-nine children received one or more doses of LUM/IVA in the open-label period (33 in the LUM/IVA to LUM/IVA group and 16 in the placebo to LUM/IVA group), with a mean exposure of 47.1 (standard deviation [SD], 5.2) weeks. The mean absolute change in MRI global score (negative value indicates improvement) from baseline at Week 96 was -2.7 (SD, 7.0; 95% confidence interval [CI], -5.2 to -0.1) in the LUM/IVA to LUM/IVA group and -5.6 (SD, 6.9; 95% CI, -9.2 to -1.9) in the placebo to LUM/IVA group. Improvements in LCI2.5, sweat chloride concentration, and markers of pancreatic function and intestinal inflammation were also observed in both groups. Growth parameters remained stable in both groups. The majority of children had adverse events considered mild (38.8%) or moderate (40.8%). Two (4.1%) children discontinued LUM/IVA treatment because of adverse events (distal intestinal obstruction syndrome [n = 1] and alanine aminotransferase increase [n = 1]). Conclusions: These findings confirm the potential for early LUM/IVA treatment to alter the trajectory of CF disease progression, including CF lung disease, in children as young as 2 years of age. Clinical trial registered with ClinicalTrials.gov (NCT03625466).
Collapse
Affiliation(s)
- Mirjam Stahl
- Department of Pediatric Respiratory Medicine, Immunology, and Critical Care Medicine and
- German Center for Lung Research (DZL), Associated Partner Site, Berlin, Germany
- Berlin Institute of Health at Charité-Universitätsmedizin, Berlin, Germany
| | - Jobst Roehmel
- Department of Pediatric Respiratory Medicine, Immunology, and Critical Care Medicine and
- German Center for Lung Research (DZL), Associated Partner Site, Berlin, Germany
- Berlin Institute of Health at Charité-Universitätsmedizin, Berlin, Germany
| | - Monika Eichinger
- Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany
- Department of Diagnostic and Interventional Radiology and
- Department of Diagnostic and Interventional Radiology with Nuclear Medicine, Thoraxklinik at University Hospital Heidelberg, Heidelberg, Germany
| | - Felix Doellinger
- Department of Radiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, 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
| | - Matthias V Kopp
- Pediatric Respiratory Medicine, Department of Pediatrics, Inselspital, University of Bern, Bern, Switzerland
- Airway Research Center North (ARCN), German Center for Lung Research (DZL), Grosshansdorf, Germany
| | - Anna-Maria Dittrich
- Department for Pediatric Pulmonology, Allergology, and Neonatology, and
- BREATH, German Center for Lung Research (DZL), Hannover Medical School, Hannover, Germany
| | - Olaf Sommerburg
- Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany
- Division of Pediatric Pulmonology and Allergy and Cystic Fibrosis Center, Department of Pediatrics, Heidelberg University Hospital, Heidelberg, Germany
| | - Partha Ray
- Vertex Pharmaceuticals Incorporated, Boston, Massachusetts; and
| | - Anita Maniktala
- Vertex Pharmaceuticals Incorporated, Boston, Massachusetts; and
- ICON plc, Global Strategic Solutions, Raleigh, North Carolina
| | - Tu Xu
- Vertex Pharmaceuticals Incorporated, Boston, Massachusetts; and
| | - Sarah Conner
- Vertex Pharmaceuticals Incorporated, Boston, Massachusetts; and
| | - Aniket Joshi
- Vertex Pharmaceuticals Incorporated, Boston, Massachusetts; and
| | - Molly Mascia
- Vertex Pharmaceuticals Incorporated, Boston, Massachusetts; and
| | - Mark O Wielpütz
- Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany
- Department of Diagnostic and Interventional Radiology and
- Department of Diagnostic and Interventional Radiology with Nuclear Medicine, Thoraxklinik at University Hospital Heidelberg, Heidelberg, Germany
| | - Marcus A Mall
- Department of Pediatric Respiratory Medicine, Immunology, and Critical Care Medicine and
- German Center for Lung Research (DZL), Associated Partner Site, Berlin, Germany
- Berlin Institute of Health at Charité-Universitätsmedizin, Berlin, Germany
| |
Collapse
|
3
|
Baroni D, Scarano N, Ludovico A, Brandas C, Parodi A, Lunaccio D, Fossa P, Moran O, Cichero E, Millo E. In Silico and In Vitro Evaluation of the Mechanism of Action of Three VX809-Based Hybrid Derivatives as Correctors of the F508del CFTR Protein. Pharmaceuticals (Basel) 2023; 16:1702. [PMID: 38139828 PMCID: PMC10748060 DOI: 10.3390/ph16121702] [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: 11/11/2023] [Revised: 12/05/2023] [Accepted: 12/06/2023] [Indexed: 12/24/2023] Open
Abstract
Cystic fibrosis (CF), the most common autosomal recessive fatal genetic disease in the Caucasian population, is caused by mutations in the gene encoding the cystic fibrosis transmembrane conductance regulator (CFTR), an anion channel that regulates salt and water transport across a variety of secretory epithelia. Deletion of phenylalanine at position 508, F508del, the most common CF-causing mutation, destabilises the CFTR protein, causing folding and trafficking defects that lead to a dramatic reduction in its functional expression. Small molecules called correctors have been developed to rescue processing-defective F508del CFTR. We have combined in silico and in vitro approaches to investigate the mechanism of action and potential as CFTR correctors of three hybrid derivatives (2a, 7a, and 7m) obtained by merging the amino-arylthiazole core with the benzodioxole carboxamide moiety characterising the corrector lumacaftor. Molecular modelling analyses suggested that the three hybrids interact with a putative region located at the MSD1/NBD1 interface. Biochemical analyses confirmed these results, showing that the three molecules affect the expression and stability of the F508del NBD1. Finally, the YFP assay was used to evaluate the influence of the three hybrid derivatives on F508del CFTR function, assessing that their effect is additive to that of the correctors VX661 and VX445. Our study shows that the development and testing of optimised compounds targeting different structural and functional defects of mutant CFTR is the best strategy to provide more effective correctors that could be used alone or in combination as a valuable therapeutic option to treat an even larger cohort of people affected by CF.
Collapse
Affiliation(s)
- Debora Baroni
- Istituto di Biofisica, Consiglio Nazionale delle Ricerche (CNR), Via De Marini, 6, 16149 Genova, Italy; (A.L.); (O.M.)
| | - Naomi Scarano
- Department of Pharmacy, Section of Medicinal Chemistry, School of Medical and Pharmaceutical Sciences, University of Genova, Viale Benedetto XV, 3, 16132 Genoa, Italy; (N.S.); (P.F.)
| | - Alessandra Ludovico
- Istituto di Biofisica, Consiglio Nazionale delle Ricerche (CNR), Via De Marini, 6, 16149 Genova, Italy; (A.L.); (O.M.)
| | - Chiara Brandas
- Istituto di Biofisica, Consiglio Nazionale delle Ricerche (CNR), Via De Marini, 6, 16149 Genova, Italy; (A.L.); (O.M.)
| | - Alice Parodi
- Department of Experimental Medicine, Section of Biochemistry, University of Genoa, Viale Benedetto XV 1, 16132 Genova, Italy; (A.P.); (D.L.); (E.M.)
| | - Dario Lunaccio
- Department of Experimental Medicine, Section of Biochemistry, University of Genoa, Viale Benedetto XV 1, 16132 Genova, Italy; (A.P.); (D.L.); (E.M.)
| | - Paola Fossa
- Department of Pharmacy, Section of Medicinal Chemistry, School of Medical and Pharmaceutical Sciences, University of Genova, Viale Benedetto XV, 3, 16132 Genoa, Italy; (N.S.); (P.F.)
| | - Oscar Moran
- Istituto di Biofisica, Consiglio Nazionale delle Ricerche (CNR), Via De Marini, 6, 16149 Genova, Italy; (A.L.); (O.M.)
| | - Elena Cichero
- Department of Pharmacy, Section of Medicinal Chemistry, School of Medical and Pharmaceutical Sciences, University of Genova, Viale Benedetto XV, 3, 16132 Genoa, Italy; (N.S.); (P.F.)
| | - Enrico Millo
- Department of Experimental Medicine, Section of Biochemistry, University of Genoa, Viale Benedetto XV 1, 16132 Genova, Italy; (A.P.); (D.L.); (E.M.)
| |
Collapse
|
4
|
Flores J, Ziegler B, Silvello D, Dalcin P. Effects of an early rehabilitation program for adult cystic fibrosis patients during hospitalization: a randomized clinical trial. Braz J Med Biol Res 2023; 56:e12752. [PMID: 37585917 PMCID: PMC10427163 DOI: 10.1590/1414-431x2023e12752] [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: 02/24/2023] [Accepted: 06/14/2023] [Indexed: 08/18/2023] Open
Abstract
There is little information on pulmonary rehabilitation in patients with cystic fibrosis (CF) with pulmonary exacerbation. This study aimed to evaluate the effects of an early rehabilitation program on lung function, muscle strength, inflammatory markers, and quality of life in adults with CF hospitalized for pulmonary exacerbation. In this randomized controlled trial, 19 patients were included in the intervention group and 15 in the control group. The intervention group underwent an early rehabilitation program for 14 days after admission. All patients underwent spirometry, one-repetition maximum tests (1RM), and the 6-min walk test, and answered the Revised Cystic Fibrosis Questionnaire (CFQ-R) for quality of life and the International Physical Activity Questionnaire. Serum levels of interleukin and tumor necrosis factor alpha (TNF-α) were measured. In the intervention group, there were increases in 1RM biceps (P=0.009), triceps (P=0.005), shoulder abductors (P=0.002), shoulder flexors (P=0.004), hamstrings (P<0.001), and quadriceps values (P<0.001). In addition, there were improvements in CFQ-R-emotion (P=0.002), treatment burden (P=0.002), vitality (P=0.011), and physical scores (P=0.026), and a reduction in the Borg resting fatigue score (P=0.037). The interleukins levels did not change after the intervention. In adult CF patients with pulmonary exacerbation, early hospital rehabilitation had a significant impact on improving resting fatigue, muscle strength, and quality of life.
Collapse
Affiliation(s)
- J. Flores
- Programa de Pós-Graduação em Ciências Pneumológicas, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brasil
| | - B. Ziegler
- Hospital de Clínicas de Porto Alegre, Serviço de Pneumologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brasil
| | - D. Silvello
- Programa de Pós-Graduação em Ciências Pneumológicas, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brasil
| | - P.T.R. Dalcin
- Programa de Pós-Graduação em Ciências Pneumológicas, Hospital de Clínicas de Porto Alegre, Serviço de Pneumologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brasil
| |
Collapse
|
5
|
Abdelhamid AG, Yousef AE. Combating Bacterial Biofilms: Current and Emerging Antibiofilm Strategies for Treating Persistent Infections. Antibiotics (Basel) 2023; 12:1005. [PMID: 37370324 DOI: 10.3390/antibiotics12061005] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 05/29/2023] [Accepted: 06/01/2023] [Indexed: 06/29/2023] Open
Abstract
Biofilms are intricate multicellular structures created by microorganisms on living (biotic) or nonliving (abiotic) surfaces. Medically, biofilms often lead to persistent infections, increased antibiotic resistance, and recurrence of infections. In this review, we highlighted the clinical problem associated with biofilm infections and focused on current and emerging antibiofilm strategies. These strategies are often directed at disrupting quorum sensing, which is crucial for biofilm formation, preventing bacterial adhesion to surfaces, impeding bacterial aggregation in viscous mucus layers, degrading the extracellular polymeric matrix, and developing nanoparticle-based antimicrobial drug complexes which target persistent cells within the biofilm core. It is important to acknowledge, however, that the use of antibiofilm agents faces obstacles, such as limited effectiveness in vivo, potential cytotoxicity to host cells, and propensity to elicit resistance in targeted biofilm-forming microbes. Emerging next generation antibiofilm strategies, which rely on multipronged approaches, were highlighted, and these benefit from current advances in nanotechnology, synthetic biology, and antimicrobial drug discovery. The assessment of current antibiofilm mitigation approaches, as presented here, could guide future initiatives toward innovative antibiofilm therapeutic strategies. Enhancing the efficacy and specificity of some emerging antibiofilm strategies via careful investigations, under conditions that closely mimic biofilm characteristics within the human body, could bridge the gap between laboratory research and practical application.
Collapse
Affiliation(s)
- Ahmed G Abdelhamid
- Department of Food Science and Technology, The Ohio State University, 2015 Fyffe Court, Columbus, OH 43210, USA
- Botany and Microbiology Department, Faculty of Science, Benha University, Benha 13518, Egypt
| | - Ahmed E Yousef
- Department of Food Science and Technology, The Ohio State University, 2015 Fyffe Court, Columbus, OH 43210, USA
- Department of Microbiology, The Ohio State University, 105 Biological Sciences Building, 484 West 12th Avenue, Columbus, OH 43210, USA
| |
Collapse
|
6
|
Occhigrossi L, Rossin F, Villella VR, Esposito S, Abbate C, D'Eletto M, Farrace MG, Tosco A, Nardacci R, Fimia GM, Raia V, Piacentini M. The STING/TBK1/IRF3/IFN type I pathway is defective in cystic fibrosis. Front Immunol 2023; 14:1093212. [PMID: 36923406 PMCID: PMC10008931 DOI: 10.3389/fimmu.2023.1093212] [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: 11/08/2022] [Accepted: 02/14/2023] [Indexed: 03/02/2023] Open
Abstract
Cystic fibrosis (CF) is a rare autosomal recessive disease caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene. The most common mutation is F508del-CFTR (ΔF) which leads the encoded ion channel towards misfolding and premature degradation. The disease is characterized by chronic bronchopulmonary obstruction, inflammation and airways colonization by bacteria, which are the major cause of morbidity and mortality. The STING pathway is the main signaling route activated in the presence of both self and pathogen DNA, leading to Type I Interferon (IFN I) production and the innate immune response. In this study, we show for the first time the relationship existing in CF between resistant and recurrent opportunistic infections by Pseudomonas aeruginosa and the innate immunity impairment. We demonstrate through ex vivo and in vivo experiments that the pathway is inadequately activated in ΔF condition and the use of direct STING agonists, as 2',3'-cyclic GMP-AMP (2', 3' cGAMP), is able to restore the immune response against bacterial colonization. Indeed, upon treatment with the STING pathway agonists, we found a reduction of colony forming units (CFUs) consequent to IFN-β enhanced production in Pseudomonas aeruginosa infected bone marrow derived macrophages and lung tissues from mice affected by Cystic Fibrosis. Importantly, we also verified that the impairment detected in the primary PBMCs obtained from ΔF patients can be corrected by 2', 3' cGAMP. Our work indicates that the cGAS/STING pathway integrity is crucial in the Cystic Fibrosis response against pathogens and that the restoration of the pathway by 2', 3' cGAMP could be exploited as a possible new target for the symptomatic treatment of the disease.
Collapse
Affiliation(s)
- Luca Occhigrossi
- Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases IRCCS 'L, Spallanzani', Rome, Italy
| | - Federica Rossin
- Department of Biology, University of Rome 'Tor Vergata', Rome, Italy
| | - Valeria Rachela Villella
- European Institute for Research in Cystic Fibrosis, at National Institute for Infectious Diseases IRCCS 'L, Spallanzani', Rome, Italy
| | - Speranza Esposito
- European Institute for Research in Cystic Fibrosis, at National Institute for Infectious Diseases IRCCS 'L, Spallanzani', Rome, Italy.,Department of Molecular Medicine, University of Rome "La Sapienza", Rome, Italy
| | - Carlo Abbate
- Department of Biology, University of Rome 'Tor Vergata', Rome, Italy
| | - Manuela D'Eletto
- Department of Biology, University of Rome 'Tor Vergata', Rome, Italy
| | | | - Antonella Tosco
- Pediatric Unit, Department of Translational Medical Sciences, Regional Cystic Fibrosis Center, Federico II University Naples, Naples, Italy
| | - Roberta Nardacci
- Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases IRCCS 'L, Spallanzani', Rome, Italy.,Departmental Faculty of Medicine and Surgery, UniCamillus-Saint Camillus International University of Health and Medical Sciences, Rome, Italy
| | - Gian Maria Fimia
- Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases IRCCS 'L, Spallanzani', Rome, Italy.,Department of Molecular Medicine, University of Rome "La Sapienza", Rome, Italy
| | - Valeria Raia
- European Institute for Research in Cystic Fibrosis, at National Institute for Infectious Diseases IRCCS 'L, Spallanzani', Rome, Italy.,Pediatric Unit, Department of Translational Medical Sciences, Regional Cystic Fibrosis Center, Federico II University Naples, Naples, Italy
| | - Mauro Piacentini
- Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases IRCCS 'L, Spallanzani', Rome, Italy.,Department of Biology, University of Rome 'Tor Vergata', Rome, Italy
| |
Collapse
|
7
|
Montemayor K, Jain R. Cystic Fibrosis: Highly Effective Targeted Therapeutics and the Impact on Sex and Racial Disparities. Med Clin North Am 2022; 106:1001-1012. [PMID: 36280328 DOI: 10.1016/j.mcna.2022.07.005] [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] [Indexed: 11/21/2022]
Abstract
People with Cystic Fibrosis (CF) are living longer and healthier lives due in part to new therapies, called Cystic Fibrosis Transmembrane Conductance Regulator (CFTR) modulators. However, disparities in outcomes still exist, with females demonstrating a shorter life expectancy than males; this is opposed to the typical female versus male life expectancy in the general United States population. In addition, minority populations such as those of Hispanic ethnicity and African Americans are less frequently eligible for these new CFTR modulators. The mechanisms driving this difference and the relative contribution of CFTR to the etiology are not yet elucidated.
Collapse
Affiliation(s)
- Kristina Montemayor
- Department of Medicine, Johns Hopkins University, 1830 E. Monument Street 5th Floor, Baltimore, MD 21205, USA
| | - Raksha Jain
- Department of Medicine, University of Texas Southwestern, 5323 Harry Hines Boulevard, Dallas, TX 75390-8558, USA.
| |
Collapse
|
8
|
Van der Plas SE, Kelgtermans H, Mammoliti O, Menet C, Tricarico G, De Blieck A, Joannesse C, De Munck T, Lambin D, Cowart M, Dropsit S, Martina SLX, Gees M, Wesse AS, Conrath K, Andrews M. Discovery of GLPG2451, a Novel Once Daily Potentiator for the Treatment of Cystic Fibrosis. J Med Chem 2021; 64:343-353. [PMID: 33399458 DOI: 10.1021/acs.jmedchem.0c01796] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cystic fibrosis (CF) is a life-threatening recessive genetic disease caused by mutations in the gene encoding for the cystic fibrosis transmembrane conductance regulator (CFTR). With the discovery of Ivacaftor and Lumacaftor, it has been shown that administration of one or more small molecules can partially restore the CFTR function. Correctors are small molecules that enhance the amount of CFTR on the cell surface, while potentiators improve the gating function of the CFTR channel. Herein, we describe the discovery and optimization of a novel potentiator series. Scaffold hopping, focusing on retaining the different intramolecular contacts, was crucial in the whole discovery process to identify a novel series devoid of genotoxic liabilities. From this series, the clinical candidate GLPG2451 was selected based on its pharmacokinetic properties, allowing QD dosing and based on its low CYP induction potential.
Collapse
Affiliation(s)
| | - Hans Kelgtermans
- Galapagos NV, Generaal De Wittelaan L11 A3, 2800 Mechelen, Belgium
| | - Oscar Mammoliti
- Galapagos NV, Generaal De Wittelaan L11 A3, 2800 Mechelen, Belgium
| | - Christel Menet
- Galapagos NV, Generaal De Wittelaan L11 A3, 2800 Mechelen, Belgium
| | | | - Ann De Blieck
- Galapagos NV, Generaal De Wittelaan L11 A3, 2800 Mechelen, Belgium
| | | | - Tom De Munck
- Galapagos NV, Generaal De Wittelaan L11 A3, 2800 Mechelen, Belgium
| | - Dominique Lambin
- Galapagos NV, Generaal De Wittelaan L11 A3, 2800 Mechelen, Belgium
| | - Marlon Cowart
- Abbvie, Discovery Chemistry and Technology, North Chicago, Illinois 60064, United States
| | | | | | | | | | - Katja Conrath
- Galapagos NV, Generaal De Wittelaan L11 A3, 2800 Mechelen, Belgium
| | - Martin Andrews
- Galapagos NV, Generaal De Wittelaan L11 A3, 2800 Mechelen, Belgium
| |
Collapse
|
9
|
Senior BA. Cystic fibrosis: On the cusp of cure? Int Forum Allergy Rhinol 2020; 11:745-746. [PMID: 33070464 DOI: 10.1002/alr.22714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Accepted: 10/05/2020] [Indexed: 11/10/2022]
Affiliation(s)
- Brent A Senior
- Department of Otolaryngology/Head and Neck Surgery, University of North Carolina at Chapel Hill, Chapel Hill, NC
| |
Collapse
|
10
|
Jayawardena ADL, Fracchia MS, Bartley BL, Yonker LM, Lapey A, Virgin F, Hartnick CJ. Working towards consensus in the management of pediatric chronic rhinosinusitis in cystic fibrosis. Int J Pediatr Otorhinolaryngol 2020; 135:110047. [PMID: 32446041 DOI: 10.1016/j.ijporl.2020.110047] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 04/07/2020] [Accepted: 04/09/2020] [Indexed: 11/27/2022]
Abstract
OBJECTIVE The prevalence of chronic rhinosinusitis (CRS), defined by mucosal thickening on imaging, approaches 100% in the cystic fibrosis (CF) population. CRS is associated with significant morbidity in CF, including its ability to trigger pulmonary exacerbations. CRS in CF is typically managed by pediatricians, otolaryngologists and pulmonologists. This survey evaluates the variance in practice patterns of CRS in CF amongst specialists. METHODS This is a cross-sectional, electronic survey in which maximum variation purposive sampling was used by a multi-disciplinary group of pediatric, otolaryngology and pulmonology providers in order to select a survey population with expertise in CRS in CF patients. The survey was distributed to 381 practitioners from September to October 2019. RESULTS 175 participants responded (45% response rate). Ten (of 54) statements achieved 75% consensus agreement. Consensus statements included: The decision to pursue surgical intervention for CRS in CF is a multi-disciplinary approach (94%; n = 146); maximal medical management should include nasal saline irrigation (93%; n = 142), topical steroids (75%; n = 117), maximal medical management should not include intravenous steroids (79%; n = 122); image guidance in surgery is necessary for all surgery involving the frontal sinuses (77%; n = 43), and all revision surgery(80%, n = 45); the appropriate setting for sinus surgery in a CF patient varies depending on patient presentation (89%; n = 133); post-operative regimen should include nasal saline (93%; n = 137); but does depend on the severity of disease discovered intra-operatively (84%; n = 124); post-operative antibiotics should be guided by intra-operative culture data (82%; n = 121). CONCLUSIONS There is a great deal of variation amongst specialists in the treatment of CRS in CF, however 10 statements met consensus criteria and should be considered when forming clinical care guidelines in this population.
Collapse
Affiliation(s)
- Asitha D L Jayawardena
- Massachusetts Eye and Ear Infirmary, Department of Otolaryngology Head and Neck Surgery, Boston, MA, USA
| | - M Shannon Fracchia
- Massachusetts General Hospital, Department of Pediatric Pulmonology, Boston, MA, USA
| | - Bethany L Bartley
- Massachusetts General Hospital, Department of Pediatric Pulmonology, Boston, MA, USA
| | - Lael M Yonker
- Massachusetts General Hospital, Department of Pediatric Pulmonology, Boston, MA, USA
| | - Allen Lapey
- Massachusetts General Hospital, Department of Pediatric Pulmonology, Boston, MA, USA
| | - Frank Virgin
- Vanderbilt University Medical Center, Nashville, TN, USA
| | - Christopher J Hartnick
- Massachusetts Eye and Ear Infirmary, Department of Otolaryngology Head and Neck Surgery, Boston, MA, USA.
| |
Collapse
|
11
|
Conway RF, Frum T, Conchola AS, Spence JR. Understanding Human Lung Development through In Vitro Model Systems. Bioessays 2020; 42:e2000006. [PMID: 32310312 PMCID: PMC7433239 DOI: 10.1002/bies.202000006] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 02/28/2020] [Indexed: 12/19/2022]
Abstract
An abundance of information about lung development in animal models exists; however, comparatively little is known about lung development in humans. Recent advances using primary human lung tissue combined with the use of human in vitro model systems, such as human pluripotent stem cell-derived tissue, have led to a growing understanding of the mechanisms governing human lung development. They have illuminated key differences between animal models and humans, underscoring the need for continued advancements in modeling human lung development and utilizing human tissue. This review discusses the use of human tissue and the use of human in vitro model systems that have been leveraged to better understand key regulators of human lung development and that have identified uniquely human features of development. This review also examines the implementation and challenges of human model systems and discusses how they can be applied to address knowledge gaps.
Collapse
Affiliation(s)
- Renee F Conway
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, 48104, USA
| | - Tristan Frum
- Department of Internal Medicine, Gastroenterology, University of Michigan Medical School, Ann Arbor, MI, 48104, USA
| | - Ansley S Conchola
- Cell and Molecular Biology (CMB) Training Program, University of Michigan Medical School, Ann Arbor, MI, 48104, USA
| | - Jason R Spence
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, 48104, USA
- Department of Internal Medicine, Gastroenterology, University of Michigan Medical School, Ann Arbor, MI, 48104, USA
- Cell and Molecular Biology (CMB) Training Program, University of Michigan Medical School, Ann Arbor, MI, 48104, USA
- Department of Biomedical Engineering, University of Michigan College of Engineering, Ann Arbor, MI, 48104, USA
| |
Collapse
|
12
|
Metwally AA, Ascoli C, Turturice B, Rani A, Ranjan R, Chen Y, Schott C, Faro A, Ferkol TW, Finn PW, Perkins DL. Pediatric lung transplantation: Dynamics of the microbiome and bronchiolitis obliterans in cystic fibrosis. J Heart Lung Transplant 2020; 39:824-834. [PMID: 32580896 DOI: 10.1016/j.healun.2020.04.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 04/02/2020] [Accepted: 04/17/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Compositional changes in the microbiome are associated with the development of bronchiolitis obliterans (BO) after lung transplantation (LTx) in adults with cystic fibrosis (CF). The association between the lower airway bacterial community and BO after LTx in children with CF remains largely unexplored and is possibly influenced by frequent antibiotic therapy. The objectives of this study were to examine the relationship between bacterial community dynamics and the development of BO and analyze antibiotic resistance trends in children after LTx for CF. METHODS For 3 years from the time of transplant, 12 LTx recipients were followed longitudinally, with 5 subjects developing BO during the study period. A total of 82 longitudinal bronchoalveolar lavage samples were collected during standard of care bronchoscopies. Metagenomic shotgun sequencing was performed on the extracted microbial DNA from bronchoalveolar lavage specimens. Taxonomic profiling was constructed using WEVOTE pipeline. The longitudinal association between development of BO and temporal changes in bacterial diversity and abundance were evaluated with MetaLonDA. The analysis of antibiotic resistance genes was performed with the ARGs-OAP v2.0 pipeline. RESULTS All recipients demonstrated a Proteobacteria-predominant lower airways community. Temporal reduction in bacterial diversity was significantly associated with the development of BO and associated with neutrophilia and antibiotic therapy. Conversely, an increasing abundance of the phylum Actinobacteria and the orders Neisseriales and Pseudonocardiales in the lower airways was significantly associated with resilience to BO. A more diverse bacterial community was related to a higher expression of multidrug resistance genes and increased proteobacterial abundance. CONCLUSIONS Decreased diversity within bacterial communities may suggest a contribution to pediatric lung allograft rejection in CF.
Collapse
Affiliation(s)
- Ahmed A Metwally
- Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois; Division of Pulmonary, Critical Care, Sleep, and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois; Department of Genetics, Stanford University, Stanford, California
| | - Christian Ascoli
- Division of Pulmonary, Critical Care, Sleep, and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Benjamin Turturice
- Division of Pulmonary, Critical Care, Sleep, and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois; Departments of Microbiology and Immunology
| | - Asha Rani
- Division of Pulmonary, Critical Care, Sleep, and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Ravi Ranjan
- Division of Pulmonary, Critical Care, Sleep, and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Yang Chen
- Division of Pulmonary, Critical Care, Sleep, and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois; Biological Sciences, University of Illinois at Chicago, Chicago, Illinois
| | - Cody Schott
- Division of Pulmonary, Critical Care, Sleep, and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois; Departments of Microbiology and Immunology
| | - Albert Faro
- Cystic Fibrosis Foundation, Bethesda, Maryland; Division of Pediatric Allergy, Immunology, and Pulmonary Medicine, Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri
| | - Thomas W Ferkol
- Division of Pediatric Allergy, Immunology, and Pulmonary Medicine, Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri; Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri
| | - Patricia W Finn
- Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois; Division of Pulmonary, Critical Care, Sleep, and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois; Departments of Microbiology and Immunology.
| | - David L Perkins
- Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois; Division of Nephrology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois; Department of Surgery, University of Illinois at Chicago, Chicago, Illinois.
| |
Collapse
|
13
|
|
14
|
Deep learning to automate Brasfield chest radiographic scoring for cystic fibrosis. J Cyst Fibros 2020; 19:131-138. [DOI: 10.1016/j.jcf.2019.04.016] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 03/27/2019] [Accepted: 04/21/2019] [Indexed: 12/15/2022]
|
15
|
Ferkol TW. Prevention of cystic fibrosis: The beginning of the end? Sci Transl Med 2019; 11:11/485/eaax2361. [PMID: 30918110 DOI: 10.1126/scitranslmed.aax2361] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 03/08/2019] [Indexed: 01/04/2023]
Abstract
Prenatal and postnatal treatment with a CFTR modifier attenuates pathological changes in a ferret model of cystic fibrosis (Sun et al., this issue).
Collapse
Affiliation(s)
- Thomas W Ferkol
- Department of Pediatrics and Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
16
|
Southern KW, Clancy JP, Ranganathan S. Aerosolized agents for airway clearance in cystic fibrosis. Pediatr Pulmonol 2019; 54:858-864. [PMID: 30884217 DOI: 10.1002/ppul.24306] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 01/22/2019] [Accepted: 01/24/2019] [Indexed: 11/11/2022]
Abstract
The outlook for people with cystic fibrosis (CF) has improved considerably as a result of conventional therapies including aerosolized agents for airway clearance. These will continue to play a significant role in maintaining well-being and improving survival, even as newer agents emerge that correct the underlying CF defect. In this review, we explore the evidence supporting the use of dornase alfa, hypertonic saline, and mannitol in improving mucus clearance in patients with CF from different age groups with differing disease severity. We also discuss the clinical use of these agents in the context of available international guidelines as well as practical considerations in the clinic, highlighting the importance of a multidisciplinary approach and shared decision-making. Unanswered questions regarding the optimal use of these agents are highlighted.
Collapse
Affiliation(s)
- Kevin W Southern
- Department of Women's and Children's Health, University of Liverpool, Liverpool, UK
| | - John P Clancy
- Division of Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Sarath Ranganathan
- Department of Respiratory and Sleep Medicine, Royal Children's Hospital, Melbourne, Victoria, Australia
| |
Collapse
|
17
|
de Wilde G, Gees M, Musch S, Verdonck K, Jans M, Wesse AS, Singh AK, Hwang TC, Christophe T, Pizzonero M, Van der Plas S, Desroy N, Cowart M, Stouten P, Nelles L, Conrath K. Identification of GLPG/ABBV-2737, a Novel Class of Corrector, Which Exerts Functional Synergy With Other CFTR Modulators. Front Pharmacol 2019; 10:514. [PMID: 31143125 PMCID: PMC6521598 DOI: 10.3389/fphar.2019.00514] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 04/24/2019] [Indexed: 01/28/2023] Open
Abstract
The deletion of phenylalanine at position 508 (F508del) in cystic fibrosis transmembrane conductance regulator (CFTR) causes a severe defect in folding and trafficking of the chloride channel resulting in its absence at the plasma membrane of epithelial cells leading to cystic fibrosis. Progress in the understanding of the disease increased over the past decades and led to the awareness that combinations of mechanistically different CFTR modulators are required to obtain meaningful clinical benefit. Today, there remains an unmet need for identification and development of more effective CFTR modulator combinations to improve existing therapies for patients carrying the F508del mutation. Here, we describe the identification of a novel F508del corrector using functional assays. We provide experimental evidence that the clinical candidate GLPG/ABBV-2737 represents a novel class of corrector exerting activity both on its own and in combination with VX809 or GLPG/ABBV-2222.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Tzyh-Chang Hwang
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, United States
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Marinko J, Huang H, Penn WD, Capra JA, Schlebach JP, Sanders CR. Folding and Misfolding of Human Membrane Proteins in Health and Disease: From Single Molecules to Cellular Proteostasis. Chem Rev 2019; 119:5537-5606. [PMID: 30608666 PMCID: PMC6506414 DOI: 10.1021/acs.chemrev.8b00532] [Citation(s) in RCA: 167] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Indexed: 12/13/2022]
Abstract
Advances over the past 25 years have revealed much about how the structural properties of membranes and associated proteins are linked to the thermodynamics and kinetics of membrane protein (MP) folding. At the same time biochemical progress has outlined how cellular proteostasis networks mediate MP folding and manage misfolding in the cell. When combined with results from genomic sequencing, these studies have established paradigms for how MP folding and misfolding are linked to the molecular etiologies of a variety of diseases. This emerging framework has paved the way for the development of a new class of small molecule "pharmacological chaperones" that bind to and stabilize misfolded MP variants, some of which are now in clinical use. In this review, we comprehensively outline current perspectives on the folding and misfolding of integral MPs as well as the mechanisms of cellular MP quality control. Based on these perspectives, we highlight new opportunities for innovations that bridge our molecular understanding of the energetics of MP folding with the nuanced complexity of biological systems. Given the many linkages between MP misfolding and human disease, we also examine some of the exciting opportunities to leverage these advances to address emerging challenges in the development of therapeutics and precision medicine.
Collapse
Affiliation(s)
- Justin
T. Marinko
- Department
of Biochemistry, Vanderbilt University, Nashville, Tennessee 37240, United States
- Center
for Structural Biology, Vanderbilt University, Nashville, Tennessee 37240, United States
| | - Hui Huang
- Department
of Biochemistry, Vanderbilt University, Nashville, Tennessee 37240, United States
- Center
for Structural Biology, Vanderbilt University, Nashville, Tennessee 37240, United States
| | - Wesley D. Penn
- Department
of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
| | - John A. Capra
- Center
for Structural Biology, Vanderbilt University, Nashville, Tennessee 37240, United States
- Department
of Biological Sciences, Vanderbilt University, Nashville, Tennessee 37245, United States
| | - Jonathan P. Schlebach
- Department
of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
| | - Charles R. Sanders
- Department
of Biochemistry, Vanderbilt University, Nashville, Tennessee 37240, United States
| |
Collapse
|
19
|
Hamilos DL. Biofilm Formations in Pediatric Respiratory Tract Infection Part 2: Mucosal Biofilm Formation by Respiratory Pathogens and Current and Future Therapeutic Strategies to Inhibit Biofilm Formation or Eradicate Established Biofilm. Curr Infect Dis Rep 2019; 21:8. [DOI: 10.1007/s11908-019-0657-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
20
|
Tchoukaev A, Taytard J, Rousselet N, Rebeyrol C, Debray D, Blouquit-Laye S, Moisan MP, Foury A, Guillot L, Corvol H, Tabary O, Le Rouzic P. Opposite Expression of Hepatic and Pulmonary Corticosteroid-Binding Globulin in Cystic Fibrosis Patients. Front Pharmacol 2018; 9:545. [PMID: 29922157 PMCID: PMC5996105 DOI: 10.3389/fphar.2018.00545] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 05/08/2018] [Indexed: 01/02/2023] Open
Abstract
Cystic fibrosis (CF) is characterized by a chronic pulmonary inflammation. In CF, glucocorticoids (GC) are widely used, but their efficacy and benefit/risk ratio are still debated. In plasma, corticosteroid-binding globulin (CBG) binds 90% of GC and delivers them to the inflammatory site. The main goal of this work was to study CBG expression in CF patients in order to determine whether CBG could be used to optimize GC treatment. The expression of CBG was measured in liver samples from CF cirrhotic and non-CF cirrhotic patients by qPCR and Western blot and in lung samples from non-CF and CF patients by qPCR. CBG binding assays with 3H-cortisol and the measurement of the elastase/α1-antitrypsin complex were performed using the plasmas. CBG expression increased in the liver at the transcript and protein level but not in the plasma of CF patients. This is possibly due to an increase of plasmatic elastase. We demonstrated that pulmonary CBG was expressed in the bronchi and bronchioles and its expression decreased in the CF lungs, at both levels studied. Despite the opposite expression of hepatic and pulmonary CBG in CF patients, the concentration of CBG in the plasma was normal. Thus, CBG might be useful to deliver an optimized synthetic GC displaying high affinity for CBG to the main inflammatory site in the context of CF, e.g., the lung.
Collapse
Affiliation(s)
- Anastasia Tchoukaev
- INSERM, Centre de Recherche Saint-Antoine, Sorbonne Université, Paris, France
| | - Jessica Taytard
- INSERM, Centre de Recherche Saint-Antoine, Sorbonne Université, Paris, France.,Pediatric Respiratory Department, Trousseau Hospital, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Nathalie Rousselet
- INSERM, Centre de Recherche Saint-Antoine, Sorbonne Université, Paris, France
| | - Carine Rebeyrol
- INSERM, Centre de Recherche Saint-Antoine, Sorbonne Université, Paris, France
| | - Dominique Debray
- INSERM, Centre de Recherche Saint-Antoine, Sorbonne Université, Paris, France.,Pediatric Hepatology Unit, Necker Enfants Malades Hospital, Paris, France
| | - Sabine Blouquit-Laye
- INSERM U1173, UFR des Sciences de la Santé Simone Veil, Université de Versailles Saint-Quentin-en-Yvelines, Versailles, France
| | - Marie-Pierre Moisan
- INRA, Laboratoire NutriNeurO, UMR 1286, Université de Bordeaux, Bordeaux, France
| | - Aline Foury
- INRA, Laboratoire NutriNeurO, UMR 1286, Université de Bordeaux, Bordeaux, France
| | - Loic Guillot
- INSERM, Centre de Recherche Saint-Antoine, Sorbonne Université, Paris, France
| | - Harriet Corvol
- INSERM, Centre de Recherche Saint-Antoine, Sorbonne Université, Paris, France.,Pediatric Respiratory Department, Trousseau Hospital, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Olivier Tabary
- INSERM, Centre de Recherche Saint-Antoine, Sorbonne Université, Paris, France
| | - Philippe Le Rouzic
- INSERM, Centre de Recherche Saint-Antoine, Sorbonne Université, Paris, France
| |
Collapse
|
21
|
Association of Antibiotics, Airway Microbiome, and Inflammation in Infants with Cystic Fibrosis. Ann Am Thorac Soc 2018; 14:1548-1555. [PMID: 28708417 DOI: 10.1513/annalsats.201702-121oc] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
RATIONALE The underlying defect in the cystic fibrosis (CF) airway leads to defective mucociliary clearance and impaired bacterial killing, resulting in endobronchial infection and inflammation that contributes to progressive lung disease. Little is known about the respiratory microbiota in the early CF airway and its relationship to inflammation. OBJECTIVES To examine the bacterial microbiota and inflammatory profiles in bronchoalveolar lavage fluid and oropharyngeal secretions in infants with CF. METHODS Infants with CF from U.S. and Australian centers were enrolled in a prospective, observational study examining the bacterial microbiota and inflammatory profiles of the respiratory tract. Bacterial diversity and density (load) were measured. Lavage samples were analyzed for inflammatory markers (interleukin 8, unbound neutrophil elastase, and absolute neutrophil count) in the epithelial lining fluid. RESULTS Thirty-two infants (mean age, 4.7 months) underwent bronchoalveolar lavage and oropharyngeal sampling. Shannon diversity strongly correlated between upper and lower airway samples from a given subject, although community compositions differed. Microbial diversity was lower in younger subjects and in those receiving daily antistaphylococcal antibiotic prophylaxis. In lavage samples, reduced diversity correlated with lower interleukin 8 concentration and absolute neutrophil count. CONCLUSIONS In infants with CF, reduced bacterial diversity in the upper and lower airways was strongly associated with the use of prophylactic antibiotics and younger age at the time of sampling; less diversity in the lower airway correlated with lower inflammation on bronchoalveolar lavage. Our findings suggest modification of the respiratory microbiome in infants with CF may influence airway inflammation.
Collapse
|
22
|
van Horck M, van de Kant K, Winkens B, Wesseling G, Gulmans V, Hendriks H, van der Grinten C, Jöbsis Q, Dompeling E. Risk factors for lung disease progression in children with cystic fibrosis. Eur Respir J 2018; 51:13993003.02509-2017. [PMID: 29773689 DOI: 10.1183/13993003.02509-2017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 04/16/2018] [Indexed: 12/15/2022]
Abstract
To identify potential risk factors for lung disease progression in children with cystic fibrosis (CF), we studied the longitudinal data of all children with CF (aged ≥5 years) registered in the Dutch CF Registry (2009-2014).Lung disease progression was expressed as a decline in lung function (forced expiratory volume in 1 s (FEV1) % pred) and pulmonary exacerbation rate. Potential risk factors at baseline included sex, age, best FEV1 % pred, best forced vital capacity % pred, genotype, body mass index z-score, pancreatic insufficiency, medication use (proton pump inhibitors (PPIs), prophylactic antibiotics and inhaled corticosteroids), CF-related diabetes, allergic bronchopulmonary aspergillosis and colonisation with Pseudomonas aeruginosaThe data of 545 children were analysed. PPI use was associated with both annual decline of FEV1 % pred (p=0.017) and future pulmonary exacerbation rate (p=0.006). Moreover, lower FEV1 % pred at baseline (p=0.007), prophylactic inhaled antibiotic use (p=0.006) and pulmonary exacerbations in the baseline year (p=0.002) were related to pulmonary exacerbations in subsequent years.In a cohort of Dutch children with CF followed for 5 years, we were able to identify several risk factors for future exacerbations. In particular, the association between PPI use and lung disease progression definitely requires further investigation.
Collapse
Affiliation(s)
- Marieke van Horck
- Dept of Paediatric Respiratory Medicine, Care and Public Health Research Institute (CAPHRI), Maastricht University Medical Centre (MUMC+), Maastricht, The Netherlands.,Dept of Paediatrics, Zuyderland Medical Centre, Sittard-Geleen, The Netherlands
| | - Kim van de Kant
- Dept of Paediatric Respiratory Medicine, Care and Public Health Research Institute (CAPHRI), Maastricht University Medical Centre (MUMC+), Maastricht, The Netherlands.,Dept of Family Medicine, Care and Public Health Research Institute (CAPHRI), Maastricht University Medical Centre (MUMC+), Maastricht, The Netherlands
| | - Bjorn Winkens
- Dept of Methodology and Statistics, Care and Public Health Research Institute (CAPHRI), Maastricht University Medical Centre (MUMC+), Maastricht, The Netherlands
| | - Geertjan Wesseling
- Dept of Respiratory Medicine, Care and Public Health Research Institute (CAPHRI), Maastricht University Medical Centre (MUMC+), Maastricht, The Netherlands
| | | | - Han Hendriks
- Dept of Paediatric Respiratory Medicine, Care and Public Health Research Institute (CAPHRI), Maastricht University Medical Centre (MUMC+), Maastricht, The Netherlands.,Dept of Paediatrics, Zuyderland Medical Centre, Sittard-Geleen, The Netherlands
| | - Chris van der Grinten
- Dept of Respiratory Medicine, Care and Public Health Research Institute (CAPHRI), Maastricht University Medical Centre (MUMC+), Maastricht, The Netherlands
| | - Quirijn Jöbsis
- Dept of Paediatric Respiratory Medicine, Care and Public Health Research Institute (CAPHRI), Maastricht University Medical Centre (MUMC+), Maastricht, The Netherlands
| | - Edward Dompeling
- Dept of Paediatric Respiratory Medicine, Care and Public Health Research Institute (CAPHRI), Maastricht University Medical Centre (MUMC+), Maastricht, The Netherlands
| |
Collapse
|
23
|
Saldana PS, Pomeranz JL, Young ME. More than a job: Career development of individuals with cystic fibrosis. Work 2018; 59:425-437. [PMID: 29630585 DOI: 10.3233/wor-182694] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Cystic fibrosis and its employment corollaries have received little attention despite the fact that complications of CF represent numerous theorized barriers to optimal career outcomes. OBJECTIVES The objective of the study was to conduct grounded theory research that results in an understanding of the employment experiences of people with CF and ultimately a substantive grounded theory of career development applicable to individuals with this disease. METHODS This study utilized the grounded theory method of qualitative inquiry. A purposive sample of ten young adults with CF participated in in-depth semi-structured qualitative interviews. Analysis of the interview transcripts followed the constant comparative approach to coding, which identified core themes and sub-themes and culminated in a conceptual framework of variables influencing employment and career development. RESULTS An ecologically-based grounded theory of career development was developed. Major themes that influenced employment and career development were illness appraisal, occupational compromise, persistence, and altruism. Two patterns of career development outcomes emerged - the uninterrupted and the interrupted. CONCLUSIONS A complicated interaction of numerous ecological variables (individual characteristics, personal contextual factors, mediating factors, and the environment) collectively influenced career development. The presence of CF alone did not guarantee negative effects on career development.
Collapse
Affiliation(s)
- Pablo S Saldana
- Nemours Children's Clinic, Cystic Fibrosis Center, Jacksonville, FL, USA
| | - Jamie L Pomeranz
- University of Florida, Behavioral Science and Community Health, Gainesville, FL, USA
| | - Mary Ellen Young
- University of Florida, Behavioral Science and Community Health, Gainesville, FL, USA
| |
Collapse
|
24
|
MicroRNA-9 downregulates the ANO1 chloride channel and contributes to cystic fibrosis lung pathology. Nat Commun 2017; 8:710. [PMID: 28955034 PMCID: PMC5617894 DOI: 10.1038/s41467-017-00813-z] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 07/30/2017] [Indexed: 01/06/2023] Open
Abstract
Cystic fibrosis results from reduced cystic fibrosis transmembrane conductance regulator protein activity leading to defective epithelial ion transport. Ca2+-activated Cl− channels mediate physiological functions independently of cystic fibrosis transmembrane conductance regulator. Anoctamin 1 (ANO1/TMEM16A) was identified as the major Ca2+-activated Cl− channel in airway epithelial cells, and we recently demonstrated that downregulation of the anoctamin 1 channel in cystic fibrosis patients contributes to disease severity via an unknown mechanism. Here we show that microRNA-9 (miR-9) contributes to cystic fibrosis and downregulates anoctamin 1 by directly targeting its 3′UTR. We present a potential therapy based on blockage of miR-9 binding to the 3′UTR by using a microRNA target site blocker to increase anoctamin 1 activity and thus compensate for the cystic fibrosis transmembrane conductance regulator deficiency. The target site blocker is tested in in vitro and in mouse models of cystic fibrosis, and could be considered as an alternative strategy to treat cystic fibrosis. Downregulation of the anoctamin 1 calcium channel in airway epithelial cells contributes to pathology in cystic fibrosis. Here the authors show that microRNA-9 targets anoctamin 1 and that inhibiting this interaction improves mucus dynamics in mouse models.
Collapse
|
25
|
Progress in understanding mucus abnormalities in cystic fibrosis airways. J Cyst Fibros 2017; 17:S35-S39. [PMID: 28951068 DOI: 10.1016/j.jcf.2017.09.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 09/13/2017] [Accepted: 09/14/2017] [Indexed: 12/18/2022]
Abstract
Normal airways below the carina maintain an essentially sterile environment via a multi-pronged innate defence system that includes mucus clearance via mucociliary clearance and cough, multiple antimicrobials and cellular components including macrophages and neutrophils. In cystic fibrosis (CF), loss of CFTR function compromises these defences, and with present standard of care virtually all people with CF eventually develop mucus accumulation, plugging and chronic infections. This review focuses on how mucus is affected by CFTR loss.
Collapse
|
26
|
Ferrari E, Monzani R, Villella VR, Esposito S, Saluzzo F, Rossin F, D'Eletto M, Tosco A, De Gregorio F, Izzo V, Maiuri MC, Kroemer G, Raia V, Maiuri L. Cysteamine re-establishes the clearance of Pseudomonas aeruginosa by macrophages bearing the cystic fibrosis-relevant F508del-CFTR mutation. Cell Death Dis 2017; 8:e2544. [PMID: 28079883 PMCID: PMC5386380 DOI: 10.1038/cddis.2016.476] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 11/17/2016] [Accepted: 12/16/2016] [Indexed: 12/19/2022]
Abstract
Cystic fibrosis (CF), the most common lethal monogenic disease in Caucasians, is characterized by recurrent bacterial infections and colonization, mainly by Pseudomonas aeruginosa, resulting in unresolved airway inflammation. CF is caused by mutations in the gene coding for the cystic fibrosis transmembrane conductance regulator (CFTR) protein, which functions as a chloride channel in epithelial cells, macrophages, and other cell types. Impaired bacterial handling by macrophages is a feature of CF airways, although it is still debated how defective CFTR impairs bacterial killing. Recent evidence indicates that a defective autophagy in CF macrophages leads to alterations of bacterial clearance upon infection. Here we use bone marrow-derived macrophages from transgenic mice to provide the genetic proof that defective CFTR compromises both uptake and clearance of internalized Pseudomonas aeruginosa. We demonstrate that the proteostasis regulator cysteamine, which rescues the function of the most common F508del-CFTR mutant and hence reduces lung inflammation in CF patients, can also repair the defects of CF macrophages, thus restoring both bacterial internalization and clearance through a process that involves upregulation of the pro-autophagic protein Beclin 1 and re-establishment of the autophagic pathway. Altogether these results indicate that cysteamine restores the function of several distinct cell types, including that of macrophages, which might contribute to its beneficial effects on CF.
Collapse
Affiliation(s)
- Eleonora Ferrari
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, European Institute for Research in Cystic Fibrosis, Milan 20132, Italy
| | - Romina Monzani
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, European Institute for Research in Cystic Fibrosis, Milan 20132, Italy
| | - Valeria R Villella
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, European Institute for Research in Cystic Fibrosis, Milan 20132, Italy
| | - Speranza Esposito
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, European Institute for Research in Cystic Fibrosis, Milan 20132, Italy
| | - Francesca Saluzzo
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, European Institute for Research in Cystic Fibrosis, Milan 20132, Italy
| | - Federica Rossin
- Department of Biology, University of Rome 'Tor Vergata', Rome, Italy
| | - Manuela D'Eletto
- Department of Biology, University of Rome 'Tor Vergata', Rome, Italy
| | - Antonella Tosco
- Regional Cystic Fibrosis Center, Pediatric Unit, Department of Translational Medical Sciences, FedericoII University Naples 80131, Italy
| | - Fabiola De Gregorio
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, European Institute for Research in Cystic Fibrosis, Milan 20132, Italy.,Regional Cystic Fibrosis Center, Pediatric Unit, Department of Translational Medical Sciences, FedericoII University Naples 80131, Italy
| | - Valentina Izzo
- Equipe11 labellisée Ligue Nationale contrele Cancer, Centre de Recherche des Cordeliers, Paris, France
| | - Maria C Maiuri
- Equipe11 labellisée Ligue Nationale contrele Cancer, Centre de Recherche des Cordeliers, Paris, France
| | - Guido Kroemer
- Equipe11 labellisée Ligue Nationale contrele Cancer, Centre de Recherche des Cordeliers, Paris, France.,INSERM U1138, Centre de Recherche des Cordeliers, Paris, France.,Université Paris Descartes, Paris, France.,Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France.,Pôlede Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, Franceand.,Karolinska Institute, Department of Women's and Children's Health, Karolinska University Hospital, Stockholm 17176, Sweden
| | - Valeria Raia
- Regional Cystic Fibrosis Center, Pediatric Unit, Department of Translational Medical Sciences, FedericoII University Naples 80131, Italy
| | - Luigi Maiuri
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, European Institute for Research in Cystic Fibrosis, Milan 20132, Italy.,SCDU of Pediatrics, Department of Health Sciences, University of Piemonte Orientale, Novara 28100, Italy
| |
Collapse
|
27
|
Belanger AR, Nguyen K, Osman U, Gilbert CR, Allen K, Al Rais AF, Yarmus L, Akulian JA. Pleural effusions in non-transplanted cystic fibrosis patients. J Cyst Fibros 2016; 16:499-502. [PMID: 27979723 DOI: 10.1016/j.jcf.2016.11.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 11/17/2016] [Accepted: 11/18/2016] [Indexed: 11/25/2022]
Abstract
BACKGROUND Pleural effusions are considered rare in cystic fibrosis (CF) patients. There is a paucity of available information in the literature concerning the nature and significance of pleural effusions in non-transplanted CF patients. METHODS We conducted a multicenter retrospective evaluation of non-transplanted adult CF patients. Given the small sample size, only descriptive statistics were performed. RESULTS A total of 17 CF patients with pleural effusion were identified, of whom 9 patients underwent thoracentesis. The crude incidence of pleural effusion was 43 per 10,000 person-years in hospitalized CF patients at large CF centers. All sampled effusions were inflammatory in nature. All samples submitted for culture grew at least one organism. CONCLUSION Pleural effusions are rare in adult non-transplanted CF patients. These fluid collections appear to be quite inflammatory with a higher rate of empyema than in the general population.
Collapse
Affiliation(s)
- Adam R Belanger
- Division of Pulmonary and Critical Care Medicine, University of North Carolina, 8007 Burnett Womack Building, CB 7219, Chapel Hill, NC 27713, United States.
| | - Kimtuyen Nguyen
- Division of Pulmonary and Critical Care Medicine, University of North Carolina, 8007 Burnett Womack Building, CB 7219, Chapel Hill, NC 27713, United States.
| | - Umar Osman
- Division of Pulmonary, Allergy, and Critical Care Medicine, Penn State College of Medicine, 500 University Drive, Mail Stop H041, Hersey, PA 17033, United States.
| | - Christopher R Gilbert
- Swedish Cancer Institute, Swedish Thoracic Surgery, 1101 Madison Street, Suite 900, Seattle, WA 98104, United States.
| | - Katie Allen
- Division of Pulmonary and Critical Care, Johns Hopkins Hospital, Sheikh Zayed Tower, Suite 7-125, 1800 Orleans Street, Baltimore, MD 21287, United States.
| | - Ahmad Farid Al Rais
- Division of Pulmonary and Critical Care, Johns Hopkins Hospital, Sheikh Zayed Tower, Suite 7-125, 1800 Orleans Street, Baltimore, MD 21287, United States.
| | - Lonny Yarmus
- Division of Pulmonary and Critical Care, Johns Hopkins Hospital, Sheikh Zayed Tower, Suite 7-125, 1800 Orleans Street, Baltimore, MD 21287, United States.
| | - Jason A Akulian
- Division of Pulmonary and Critical Care Medicine, University of North Carolina, 8007 Burnett Womack Building, CB 7219, Chapel Hill, NC 27713, United States.
| |
Collapse
|
28
|
Abstract
Respiratory system involvement in cystic fibrosis is the leading cause of morbidity and mortality. Defects in the cystic fibrosis transmembrane regulator (CFTR) gene throughout the sinopulmonary tract result in recurrent infections with a variety of organisms including Pseudomonas aeruginosa, methicillin-resistant Staphylococcus aureus, and nontuberculous mycobacteria. Lung disease occurs earlier in life than once thought and ideal methods of monitoring lung function, decline, or improvement with therapy are debated. Treatment of sinopulmonary disease may include physiotherapy, mucus-modifying and antiinflammatory agents, antimicrobials, and surgery. In the new era of personalized medicine, CFTR correctors and potentiators may change the course of disease.
Collapse
Affiliation(s)
- Danielle M Goetz
- Pediatric Pulmonology, Jacobs School of Medicine, Women & Children's Hospital of Buffalo, State University of New York, 219 Bryant Street, Buffalo, NY 14222, USA.
| | - Shipra Singh
- Pediatric Pulmonology, Jacobs School of Medicine, Women & Children's Hospital of Buffalo, State University of New York, 219 Bryant Street, Buffalo, NY 14222, USA
| |
Collapse
|
29
|
Hamilos DL. Chronic Rhinosinusitis in Patients with Cystic Fibrosis. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY-IN PRACTICE 2016; 4:605-12. [DOI: 10.1016/j.jaip.2016.04.013] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2016] [Revised: 04/26/2016] [Accepted: 04/27/2016] [Indexed: 12/01/2022]
|
30
|
Esposito S, Tosco A, Villella VR, Raia V, Kroemer G, Maiuri L. Manipulating proteostasis to repair the F508del-CFTR defect in cystic fibrosis. Mol Cell Pediatr 2016; 3:13. [PMID: 26976279 PMCID: PMC4791443 DOI: 10.1186/s40348-016-0040-z] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Accepted: 03/07/2016] [Indexed: 12/31/2022] Open
Abstract
Cystic fibrosis (CF) is a lethal monogenic disease caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene that entails the (diagnostic) increase in sweat electrolyte concentrations, progressive lung disease with chronic inflammation and recurrent bacterial infections, pancreatic insufficiency, and male infertility. Therapies aimed at restoring the CFTR defect have emerged. Thus, a small molecule which facilitates chloride channel opening, the potentiator Ivacaftor, has been approved for the treatment of CF patients bearing a particular class of rare CFTR mutations. However, small molecules that directly target the most common misfolded CFTR mutant, F508del, and improve its intracellular trafficking in vitro, have been less effective than expected when tested in CF patients, even in combination with Ivacaftor. Thus, new strategies are required to circumvent the F508del-CFTR defect. Airway and intestinal epithelial cells from CF patients bearing the F508del-CFTR mutation exhibit an impressive derangement of cellular proteostasis, with oxidative stress, overactivation of the tissue transglutaminase (TG2), and disabled autophagy. Proteostasis regulators such as cysteamine can rescue and stabilize a functional F508del-CFTR protein through suppressing TG2 activation and restoring autophagy in vivo in F508del-CFTR homozygous mice, in vitro in CF patient-derived cell lines, ex vivo in freshly collected primary patient’s nasal cells, as well as in a pilot clinical trial involving homozygous F508del-CFTR patients. Here, we discuss how the therapeutic normalization of defective proteostasis can be harnessed for the treatment of CF patients with the F508del-CFTR mutation.
Collapse
Affiliation(s)
- Speranza Esposito
- European Institute for Research in Cystic Fibrosis, Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, 20132, Italy
| | - Antonella Tosco
- Regional Cystic Fibrosis Center, Pediatric Unit, Department of Translational Medical Sciences, Federico II University, Naples, 80131, Italy
| | - Valeria R Villella
- European Institute for Research in Cystic Fibrosis, Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, 20132, Italy
| | - Valeria Raia
- Regional Cystic Fibrosis Center, Pediatric Unit, Department of Translational Medical Sciences, Federico II University, Naples, 80131, Italy.
| | - Guido Kroemer
- Equipe 11 labellisée par la Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France. .,Cell Biology and Metabolomics Platforms, GustaveRoussy Comprehensive Cancer Center, Villejuif, France. .,INSERM, U1138, Paris, France. .,Université Paris Descartes, Sorbonne Paris Cité, Paris, France. .,Université Pierre et Marie Curie, Paris, France. .,Pôle de Biologie, HôpitalEuropéen Georges Pompidou, AP-HP, Paris, France. .,Karolinska Institute, Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden.
| | - Luigi Maiuri
- European Institute for Research in Cystic Fibrosis, Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, 20132, Italy. .,SCDU of Pediatrics, Department of Health Sciences, University of Piemonte Orientale, Novara, 28100, Italy.
| |
Collapse
|
31
|
Hoo ZH, Curley R, Campbell MJ, Walters SJ, Hind D, Wildman MJ. Accurate reporting of adherence to inhaled therapies in adults with cystic fibrosis: methods to calculate "normative adherence". Patient Prefer Adherence 2016; 10:887-900. [PMID: 27284242 PMCID: PMC4883819 DOI: 10.2147/ppa.s105530] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Preventative inhaled treatments in cystic fibrosis will only be effective in maintaining lung health if used appropriately. An accurate adherence index should therefore reflect treatment effectiveness, but the standard method of reporting adherence, that is, as a percentage of the agreed regimen between clinicians and people with cystic fibrosis, does not account for the appropriateness of the treatment regimen. We describe two different indices of inhaled therapy adherence for adults with cystic fibrosis which take into account effectiveness, that is, "simple" and "sophisticated" normative adherence. METHODS TO CALCULATE NORMATIVE ADHERENCE Denominator adjustment involves fixing a minimum appropriate value based on the recommended therapy given a person's characteristics. For simple normative adherence, the denominator is determined by the person's Pseudomonas status. For sophisticated normative adherence, the denominator is determined by the person's Pseudomonas status and history of pulmonary exacerbations over the previous year. Numerator adjustment involves capping the daily maximum inhaled therapy use at 100% so that medication overuse does not artificially inflate the adherence level. THREE ILLUSTRATIVE CASES Case A is an example of inhaled therapy under prescription based on Pseudomonas status resulting in lower simple normative adherence compared to unadjusted adherence. Case B is an example of inhaled therapy under-prescription based on previous exacerbation history resulting in lower sophisticated normative adherence compared to unadjusted adherence and simple normative adherence. Case C is an example of nebulizer overuse exaggerating the magnitude of unadjusted adherence. CONCLUSION Different methods of reporting adherence can result in different magnitudes of adherence. We have proposed two methods of standardizing the calculation of adherence which should better reflect treatment effectiveness. The value of these indices can be tested empirically in clinical trials in which there is careful definition of treatment regimens related to key patient characteristics, alongside accurate measurement of health outcomes.
Collapse
Affiliation(s)
- Zhe Hui Hoo
- School of Health and Related Research (ScHARR), University of Sheffield, Sheffield, UK
- Sheffield Adult Cystic Fibrosis Centre, Northern General Hospital, University of Sheffield, Sheffield, UK
| | - Rachael Curley
- School of Health and Related Research (ScHARR), University of Sheffield, Sheffield, UK
- Sheffield Adult Cystic Fibrosis Centre, Northern General Hospital, University of Sheffield, Sheffield, UK
| | - Michael J Campbell
- School of Health and Related Research (ScHARR), University of Sheffield, Sheffield, UK
| | - Stephen J Walters
- School of Health and Related Research (ScHARR), University of Sheffield, Sheffield, UK
| | - Daniel Hind
- Sheffield Clinical Trials Research Unit, University of Sheffield, Sheffield, UK
| | - Martin J Wildman
- School of Health and Related Research (ScHARR), University of Sheffield, Sheffield, UK
- Sheffield Adult Cystic Fibrosis Centre, Northern General Hospital, University of Sheffield, Sheffield, UK
- Correspondence: Martin J Wildman, Sheffield Adult Cystic Fibrosis Centre, Brearley Outpatient, Northern General Hospital, Herries Road, Sheffield S5 7AU, UK, Tel +44 114 271 5212, Fax +44 114 226 6280, Email
| |
Collapse
|
32
|
Pittman JE. Assessment and Detection of Early Lung Disease in Cystic Fibrosis. PEDIATRIC ALLERGY IMMUNOLOGY AND PULMONOLOGY 2015; 28:212-219. [DOI: 10.1089/ped.2015.0568] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Jessica E. Pittman
- Division of Pediatric Allergy, Immunology, and Pulmonary Medicine, Washington University School of Medicine, Saint Louis, Missouri
| |
Collapse
|
33
|
|