1
|
Hu Y, Tuo B. The function of chloride channels in digestive system disease (Review). Int J Mol Med 2025; 55:99. [PMID: 40314091 PMCID: PMC12045473 DOI: 10.3892/ijmm.2025.5540] [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: 12/16/2024] [Accepted: 03/11/2025] [Indexed: 05/03/2025] Open
Abstract
Cation channels have been extensively studied in the context of digestive disorders, but comparatively little attention has been given to anions and their associated channels. Chloride ions, the most abundant anions in the human body, act as signaling molecules, modulating cellular behavior and playing a key role in regulating multiorgan physiological and pathophysiological mechanisms. The intra‑ and extracellular distributions of chloride ions are primarily controlled by various chloride channels and transporters. Currently, these chloride channels are classified into several groups: The chloride channels family, cystic fibrosis transmembrane conductance regulator, calcium‑activated chloride channels, volume‑regulated anion channels, proton‑activated chloride channels and ligand‑gated anion channels. This review aims to summarize the roles of chloride ion channels and transporter proteins in digestive system diseases, providing a theoretical basis for future research and offering potential new strategies for disease treatment.
Collapse
Affiliation(s)
- Yanxia Hu
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Biguang Tuo
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| |
Collapse
|
2
|
West NE, Konstan MW, Flume PA, VanDevanter DR, Magaret A, Mazurek H, Amelina EL, Laki I, Chiron R, Sutharsan S, Columbo C, Dorkin HL, Downey D, Sole A, Hjelte L, Tullis E, Dgetluck N, Dinh Q, Constantine S, White B, Elborn JS, Chmiel JF, JBT101-CF-002 Study Group. Cannabinoid receptor 2 agonist, lenabasum, for the treatment of pulmonary exacerbations in cystic fibrosis. J Cyst Fibros 2025:S1569-1993(25)00111-0. [PMID: 40425421 DOI: 10.1016/j.jcf.2025.03.015] [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: 04/10/2024] [Revised: 02/07/2025] [Accepted: 03/24/2025] [Indexed: 05/29/2025]
Abstract
BACKGROUND Lenabasum is a cannabinoid receptor 2 (CB2) agonist under development for cystic fibrosis (CF), targeting inflammation. We evaluated the efficacy and safety of lenabasum in people with CF (pwCF). METHODS We conducted a global, 28-week, randomized, double-blind, placebo-controlled Phase 2b trial. PwCF were ≥12 years old with 2-3 pulmonary exacerbations (PEx) treated with intravenous (IV) antibiotics (or 1 PEx treated with IV and ≥1 PEx treated with oral antibiotics) in the past year. Subjects were randomized 2:1:2 to lenabasum 20 mg BID, lenabasum 5 mg BID, or placebo BID. Primary endpoint was rate of PEx, comparing lenabasum 20 mg BID to placebo. RESULTS Among 447 subjects from 21 countries, mean age was 26.9 (10.3 SD) years, 53.6% were female, 45.2% homozygous for F508del, and 24.9% received CFTR modulators. Highest ppFEV1 in the previous year was 69.2% with the majority having 1-2 PEx treated with IV antibiotics (2-7 PEx treated with either IV or oral antibiotics). PEx incidence over 28 weeks was 0.84 for placebo, 0.75 for lenabasum 5 mg BID, and 0.91 for lenabasum 20 mg BID; rates were not lower relative to placebo in the 5 mg (incidence rate ratio (IRR)=0.89, 95% CI 0.66 to 1.19, p = 0.44) or the 20 mg group (IRR 1.08, 95% CI 0.86 to 1.37, p = 0.51). PEx occurred less frequently in participants from Eastern Europe, but there was no evidence of regional variation in treatment efficacy. Lenabasum was well tolerated, without safety signals. CONCLUSION Lenabasum did not improve key clinical outcomes in this Phase 2b study in pwCF.
Collapse
Affiliation(s)
- Natalie E West
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Michael W Konstan
- Rainbow Babies and Children's Hospital and Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | | | | | | | - Henryk Mazurek
- Department of Pneumonology and Cystic Fibrosis, National Institute of Tuberculosis and Lung Disorders, Rabka-Zdrój, Poland
| | - Elena L Amelina
- Pulmonary Research Institute, Federal Medical-Biological Agency, Moscow, Russia
| | - István Laki
- Third Department of Medicine, Semmelweis University, Budapest, Hungary
| | - Raphael Chiron
- Cystic Fibrosis Center, Hôpital Arnaud de Villeneuve, Centre Hospitalier Universitaire de Montpellier, Univ Montpellier, France
| | - Sivagurunathan Sutharsan
- Department of Pulmonary Medicine, University Hospital Essen - Ruhrlandklinik, Adult Cystic Fibrosis Center, University of Duisburg-Essen, Essen, Germany
| | | | - Henry L Dorkin
- Department of Pediatrics, Division of Pulmonology, Boston Children's Hospital, Boston, MA, USA
| | - DamianG Downey
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, UK
| | - Amparo Sole
- Unidad de Trasplante Pulmonary Fibrosis Quística, Hospital Universitari la Fe, Universitat de Valencia, Valencia, Spain
| | - Lena Hjelte
- Stockholm CF Center, Karolinska University Hospital Huddinge, Division of Pediatrics, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Elizabeth Tullis
- Adult Cystic Fibrosis Clinic, St Michael's Hospital and University of Toronto, Toronto, Canada
| | | | - Quinn Dinh
- Corbus Pharmaceuticals, Inc., Norwood, MA, USA
| | | | | | - J Stuart Elborn
- Imperial College and Royal Brompton Hospital, London, UK; Queen's University, Belfast, United Kingdom
| | - James F Chmiel
- Riley Hospital for Children at IU Health, Indiana University School of Medicine, Indianapolis, IN, USA.
| | | |
Collapse
|
3
|
Murphree-Terry M, Keith JD, Oden AM, Birket SE. Spontaneous lung colonization in the cystic fibrosis rat model is linked to gastrointestinal obstruction. mBio 2025; 16:e0388324. [PMID: 40042272 PMCID: PMC11980572 DOI: 10.1128/mbio.03883-24] [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: 12/13/2024] [Accepted: 01/24/2025] [Indexed: 03/29/2025] Open
Abstract
Cystic fibrosis (CF) is a genetic disease caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene, resulting in CFTR protein dysfunction. CFTR dysfunction has multi-organ consequences, leading to dehydrated mucus that is adherent to epithelia. In the lungs, this leads to recalcitrant infections with bacteria such as Pseudomonas aeruginosa. In the gut, mucus-laden feces can adhere to the intestines, resulting in distal intestinal obstruction syndrome (DIOS). There is limited information on how lung colonization and DIOS are correlated in people with CF (pwCF). In this novel work, we describe the development of spontaneous lung colonization of CF pathogens in young (<3 months old) CF rats, preceding the development of DIOS. Once DIOS is established, the lung microbiome becomes predominated by taxa also observed in the feces. Induced infection with P. aeruginosa in the CF rats reflects data found in pwCF, as once CF rats are infected, they retain a higher relative abundance of P. aeruginosa than their healthy agemates. Finally, we found that ivacaftor treatment favors a healthier gut microbiome in CF rats, decreasing the relative abundance of Escherichia coli. These results indicate that the CF rat model is recapitulative of human CF disease with the spontaneous lung colonization of traditional CF pathogens and maintenance of P. aeruginosa after induced infection. Furthermore, these results indicate a possible role for the gut-lung axis in lung colonization and DIOS in CF.IMPORTANCEThese data describe for the first time the development of spontaneous lung colonization in the cystic fibrosis (CF) rat model, a hallmark aspect of human CF disease. We also find that CF rats infected with Pseudomonas aeruginosa maintain higher relative abundance following chronic infection as compared to healthy rats, similar to those is seen in people with CF. Additionally, we describe the possible contribution of the gut-lung axis linking lung health with distal intestinal obstruction syndrome, a relationship largely unexplored in the context of CF.
Collapse
Affiliation(s)
- Mikayla Murphree-Terry
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Johnathan D. Keith
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Ashley M. Oden
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Susan E. Birket
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
4
|
Iftikhar IH, Rao ST, Nadama R, Janahi I, BaHammam AS. Comparative Efficacy of CFTR Modulators: A Network Meta-analysis. Lung 2025; 203:49. [PMID: 40102290 DOI: 10.1007/s00408-025-00802-w] [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: 12/02/2024] [Accepted: 03/05/2025] [Indexed: 03/20/2025]
Abstract
PURPOSE The objective was to study comparative efficacies of cystic fibrosis transmembrane conductance regulator (CFTR) modulators, vanzacaftor-tezacaftor-deutivacaftor (VTD), elexacaftor-tezacaftor-ivacaftor (ETI), tezacaftor-ivacaftor (Tez-Iva), and lumacaftor-ivacaftor (Lum-Iva) in people with cystic fibrosis (pwCF), aged ≥ 12 years, carrying at least one F508del-CFTR-allele. METHODS Data from randomized controlled or randomized active comparator trials were included in this network meta-analysis which used frequentist approach for comparing the efficacy of drugs and ranking based on P-scores. Outcomes of interest were mean differences in percentage-predicted forced expiratory volume in one second (ppFEV1), CF questionnaire-revised respiratory domain (CFQ-R) scores, sweat chloride (SwCl) levels, and odds ratios (OR) for serious adverse events (SAE). RESULTS Data from 13 studies were analyzed. Compared to placebo, the effects of VTD and ETI on ppFEV1 were almost quadruple of Tez-Iva and Lum-Iva (VTD: 12.78 [95% confidence intervals: 6.41; 19.15] and ETI: 11.95 [7.40; 16.50]) and almost seven times of Tez-Iva and Lum-Iva for CFQ-R (VTD: 21.23 [- 28.72; 71.18] and ETI: 19.27 [10.56; 27.98]). A statistically significant difference was noted between VTD and ETI in SwCl reduction (mean difference: - 8.59 [- 15.53; - 1.65]). There were no statistically significant ORs for SAEs for any CFTR modulators but VTD, ETI, and Tez-Iva were least associated with SAEs (ORs were 0.15 [0.01; 1.79], 0.49 [0.31; 0.78], and 0.74 [0.50; 1.09], respectively, as compared to placebo). Overall, P-score ranking ranked VTD as first and ETI as second, followed by others. CONCLUSION VTD and ETI were more efficacious than Tez-Iva and Lum-Iva in pwCF with at least one F508del-CFTR-allele.
Collapse
Affiliation(s)
- Imran Hasan Iftikhar
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care & Sleep Medicine, Emory University School of Medicine, 613 Michael St, NE, Atlanta, GA, USA.
- Atlanta Veterans Affairs Medical Center, Decatur, USA.
| | - Saad T Rao
- Shifa College of Medicine, Shifa Tameer-e-Millat University, Islamabad, Pakistan
| | - Rufai Nadama
- Pulmonary Division, King Khalid University Hospital, Riyadh, Kingdom of Saudi Arabia
| | - Ibrahim Janahi
- Pediatric Pulmonology, Pediatric Medicine, Sidra Medicine, Doha, Qatar
- Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Ahmed S BaHammam
- Department of Medicine, University Sleep Disorders Center, and Pulmonary Service, King Saud University, Riyadh, Kingdom of Saudi Arabia
- Strategic Technologies Program of the National Plan for Sciences and Technology and Innovation in the Kingdom of Saudi Arabia, Riyadh, Saudi Arabia
| |
Collapse
|
5
|
Hynes J, Taggart CC, Tirouvanziam R, Coppinger JA. Innate Immunity in Cystic Fibrosis: Varied Effects of CFTR Modulator Therapy on Cell-to-Cell Communication. Int J Mol Sci 2025; 26:2636. [PMID: 40141278 PMCID: PMC11942055 DOI: 10.3390/ijms26062636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 02/26/2025] [Accepted: 03/06/2025] [Indexed: 03/28/2025] Open
Abstract
Cystic Fibrosis (CF) is a life-shortening, multi-organ disease caused by mutations in the CF transmembrane conductance regulator (CFTR) gene. Prominent clinical features of CF take place in the lung, hallmarked by cycles of bacterial infection and a dysfunctional inflammatory airway response, leading to eventual respiratory failure. Bidirectional crosstalk between epithelial cells, leukocytes (e.g., neutrophils, macrophages) and bacteria via release of intra-cellular mediators is key to driving inflammation in CF airways. In recent years, a highly effective combination of therapeutics targeting the CFTR defect have revolutionized treatment in CF. Despite these advancements and due to the complexity of the immune response in the CF airway, the full impact of highly effective modulator therapy (HEMT) on airway inflammation is not fully determined. This review provides the evidence to date on crosstalk mechanisms between host epithelium, leukocytes and bacteria and examines the effect of HEMT on both soluble and membrane-derived immune mediators in clinical samples. The varied effects of HEMT on expression of key proteases, cytokines and extracellular vesicles (EVs) in relation to clinical parameters is assessed. Advances in treatment with HEMT have shown potential in dampening the chronic inflammatory response in CF airways. However, to fully quell inflammation and maximize lung tissue resilience, further interventions may be necessary. Exploring the effects of HEMT on key immune mediators paves the way for identifying new anti-inflammatory approaches targeting host immune cell interactions, such as EV-directed lung therapies.
Collapse
Affiliation(s)
- Jennifer Hynes
- School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, D02 YN77 Dublin, Ireland;
- Children’s Health Ireland Translational Research Centre, Children’s Health Ireland at Crumlin, D12 N512 Dublin, Ireland
| | - Clifford C. Taggart
- Airway Innate Immunity Research (AiiR) Group, Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, Belfast BT9 7BL, UK;
| | - Rabindra Tirouvanziam
- Department of Pediatrics, Emory University, Atlanta, GA 30322, USA;
- Center for CF and Airways Disease Research, Children's Healthcare of Atlanta, Atlanta, GA 30322, USA
| | - Judith A. Coppinger
- School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, D02 YN77 Dublin, Ireland;
- Children’s Health Ireland Translational Research Centre, Children’s Health Ireland at Crumlin, D12 N512 Dublin, Ireland
| |
Collapse
|
6
|
Chen CB, Granneman JA, Yadav SR. Challenges to Optimizing Nutrition in Children With Cystic Fibrosis. Curr Gastroenterol Rep 2025; 27:20. [PMID: 40053205 DOI: 10.1007/s11894-025-00969-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/25/2025] [Indexed: 05/13/2025]
Abstract
PURPOSE OF REVIEW Cystic fibrosis is a chronic condition that has significant effects on the nutritional status of pediatric patients. Malnutrition is frequently encountered in this population and has been shown to contribute to poor pulmonary and overall disease outcomes. This article will provide an overview of the physiologic and psychosocial challenges toward attaining optimal nutrition in pediatric cystic fibrosis patients. RECENT FINDINGS Newer therapies such as CFTR modulators have played significant roles in improving the nutritional status of patients with cystic fibrosis. There is also a greater focus on becoming more aware of psychosocial and cultural barriers in the care of cystic fibrosis patients. Many challenges exist in optimizing nutritional support including but not limited to the patient's clinical manifestations and disease severity, caregiver ability, and access to care. Both gastrointestinal and non-gastrointestinal disorders lead to insufficient caloric intake, increased loss and metabolic needs, and micronutrient and macronutrient deficiency. Social factors including stressful patient and caregiver relationships and altered body image also contribute to poor nutritional status.
Collapse
|
7
|
Bonfield TL, Lazarus HM. Human mesenchymal stem cell therapy: Potential advances for reducing cystic fibrosis infection and organ inflammation. Best Pract Res Clin Haematol 2025; 38:101602. [PMID: 40274338 DOI: 10.1016/j.beha.2025.101602] [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: 11/20/2024] [Revised: 02/11/2025] [Accepted: 03/04/2025] [Indexed: 04/26/2025]
Abstract
Innovation in cystic fibrosis (CF) supportive care, including implementing new antimicrobial agents, improved physiotherapy, and highly effective modulators therapy, has advanced patient survival into the 4th and 5th decades of life. However, even with these remarkable improvements in therapy, CF patients continue to suffer from pulmonary infection and other visceral organ complications associated with long-term deficient cystic fibrosis transmembrane conductance regulator (CFTR) expression. Human mesenchymal stem cells (MSCs) have been utilized in tissue engineering based upon their capacity to provide structural components of mesenchymal tissues. An alternative role of MSCs, however is their versatile utilization as cell-based infusion powerhouses due to the unique capacity to deliver milieu specific soluble biologic factors, promoting immune supportive antimicrobial and anti-inflammatory potency. MSCs derived from umbilical cord blood, bone marrow, adipose and other tissues can be expanded in ex vivo using good manufacturing procedure facilities for a safe, unique therapeutic to reduce and limit CF infection and facilitate the resolution of multi-organ inflammation. In our efforts, we conducted extensive preclinical development and validation of an allogeneic derived bone marrow derived MSC product in preparation for a clinical trial in CF. In this process, potency models were developed to ensure the functional capacity of the MSC product to provide clinical benefit. In vitro, murine in vivo and patient tissue ex vivo potency models were utilized to follow MSC anti-infective and anti-inflammatory potency associated with the CFTR deficient environment. We showed in our "First in CF" clinical trial that the allogeneic MSCs obtained from healthy volunteer bone marrow samples were safe. The advent of improved CF care measures and exciting new small molecules has changed the survival and morbidity phenotype of patients with CF, however, there are CF patients who cannot tolerate or have genotypes that are non-responsive to modulators. Additionally, even with the small molecule therapy, CF patients are living longer, but without genetic correction, with the CF disease manifestation aggravated by the continuance of pre-existing CFTR-associated clinical issues such as ongoing inflammation. MSCs secrete bio-active factors that enhance and protect tissue function and can promote "self-immune" regulation. These properties can provide therapeutic support for the traditional and changing face of CF disease clinical complications. Further, MSC-derived bio-active factors can directly mitigate colonizing pathogens' survival by producing antimicrobial peptides (AMPs) which change the pathogen surface and increase host recognition, elimination, and sensitivity to antibiotics. Herein, we review the potential of MSC therapeutics for treating many facets of CF, emphasizing the potential for providing great additive therapeutics for managing morbidity and quality of life.
Collapse
Affiliation(s)
- Tracey L Bonfield
- Genetics and Genome Sciences, National Center for Regenerative Medicine, Pediatrics and Pathology, Case Western Reserve University, Cleveland, Ohio, 44106, USA.
| | - Hillard M Lazarus
- Department of Medicine, Hematology and National Center for Regenerative Medicine, Case Western Reserve University, Cleveland, Ohio, 44106, USA
| |
Collapse
|
8
|
Jaudas F, Bartenschlager F, Shashikadze B, Santamaria G, Reichart D, Schnell A, Stöckl JB, Degroote RL, Cambra JM, Graeber SY, Bähr A, Kartmann H, Stefanska M, Liu H, Naumann-Bartsch N, Bruns H, Berges J, Hanselmann L, Stirm M, Krebs S, Deeg CA, Blum H, Schulz C, Zawada D, Janda M, Caballero-Posadas I, Kunzelmann K, Moretti A, Laugwitz KL, Kupatt C, Saalmüller A, Fröhlich T, Wolf E, Mall MA, Mundhenk L, Gerner W, Klymiuk N. Perinatal dysfunction of innate immunity in cystic fibrosis. Sci Transl Med 2025; 17:eadk9145. [PMID: 39841805 DOI: 10.1126/scitranslmed.adk9145] [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: 09/19/2023] [Revised: 07/13/2024] [Accepted: 12/22/2024] [Indexed: 01/24/2025]
Abstract
In patients with cystic fibrosis (CF), repeated cycles of infection and inflammation eventually lead to fatal lung damage. Although diminished mucus clearance can be restored by highly effective CFTR modulator therapy, inflammation and infection often persist. To elucidate the role of the innate immune system in CF etiology, we investigated a CF pig model and compared these results with those for preschool children with CF. In newborn CF pigs, we observed changes in lung immune cell composition before the onset of infection that were dominated by increased monocyte infiltration, whereas neutrophil numbers remained constant. Flow cytometric and transcriptomic profiling revealed that the infiltrating myeloid cells displayed a more immature status. Cells with comparably immature transcriptomic profiles were enriched in the blood of CF pigs at birth as well as in preschool children with CF. This pattern coincided with decreased CD16 expression in the myeloid cells of both pigs and humans, which translated into lower phagocytic activity and reduced production of reactive oxygen species in both species. These results were indicative of a congenital, translationally conserved, and functionally relevant aberration of the immune system in CF. In newborn wild-type pigs, CFTR transcription in immune cells, including lung-derived and circulating monocytes, isolated from the bone marrow, thymus, spleen, and blood was below the detection limits of highly sensitive assays, suggesting an indirect etiology of the observed effects. Our findings highlight the need for additional immunological treatments to target innate immune deficits in patients with CF.
Collapse
Affiliation(s)
- Florian Jaudas
- First Department of Medicine, Cardiology, TUM University Hospital, Technical University of Munich, School of Medicine and Health, Munich 81675, Germany
- Chair of Molecular Animal Breeding and Biotechnology, Gene Center, LMU Munich, Munich 81377, Germany
- Center of Innovative Medical Models (CiMM), LMU Munich, Oberschleissheim 85764, Germany
- Medizinische Klinik und Poliklinik I, LMU Munich, Munich 81377, Germany
| | | | - Bachuki Shashikadze
- Laboratory for Functional Genome Analysis, Gene Center, LMU Munich, Munich 81377, Germany
| | - Gianluca Santamaria
- First Department of Medicine, Cardiology, TUM University Hospital, Technical University of Munich, School of Medicine and Health, Munich 81675, Germany
- Department of Experimental and Clinical Medicine, University "Magna Graecia" of Catanzaro, Catanzaro 88100, Italy
| | - Daniel Reichart
- Medizinische Klinik und Poliklinik I, LMU Munich, Munich 81377, Germany
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich 80802, Germany
- Gene Center Munich, LMU Munich, Munich 81377, Germany
| | - Alexander Schnell
- Department for Pediatric and Adolescent Medicine, University Hospital Erlangen, Erlangen 91054, Germany
| | - Jan Bernd Stöckl
- Laboratory for Functional Genome Analysis, Gene Center, LMU Munich, Munich 81377, Germany
| | - Roxane L Degroote
- Chair of Physiology, Department of Veterinary Sciences, LMU Munich, Planegg 82152, Germany
| | - Josep M Cambra
- First Department of Medicine, Cardiology, TUM University Hospital, Technical University of Munich, School of Medicine and Health, Munich 81675, Germany
- Center of Innovative Medical Models (CiMM), LMU Munich, Oberschleissheim 85764, 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 13353, Germany
- German Center for Lung Research (DZL), associated partner site, Berlin 13353, Germany
- German Center for Child and Adolescent Health (DZKJ), partner site, Berlin 13353, Germany
| | - Andrea Bähr
- First Department of Medicine, Cardiology, TUM University Hospital, Technical University of Munich, School of Medicine and Health, Munich 81675, Germany
- Center of Innovative Medical Models (CiMM), LMU Munich, Oberschleissheim 85764, Germany
| | - Heike Kartmann
- Medizinische Klinik und Poliklinik I, LMU Munich, Munich 81377, Germany
- Gene Center Munich, LMU Munich, Munich 81377, Germany
| | - Monika Stefanska
- Medizinische Klinik und Poliklinik I, LMU Munich, Munich 81377, Germany
| | - Huan Liu
- Medizinische Klinik und Poliklinik I, LMU Munich, Munich 81377, Germany
| | - Nora Naumann-Bartsch
- Department for Pediatric and Adolescent Medicine, University Hospital Erlangen, Erlangen 91054, Germany
| | - Heiko Bruns
- Department of Pediatrics and Adolescent Medicine, FAU Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen 91054, Germany
| | - Johannes Berges
- Department of Pediatrics and Adolescent Medicine, FAU Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen 91054, Germany
| | - Lea Hanselmann
- Institute of Veterinary Pathology, Freie Universität Berlin, Berlin 14163, Germany
| | - Michael Stirm
- Chair of Molecular Animal Breeding and Biotechnology, Gene Center, LMU Munich, Munich 81377, Germany
- Center of Innovative Medical Models (CiMM), LMU Munich, Oberschleissheim 85764, Germany
| | - Stefan Krebs
- Laboratory for Functional Genome Analysis, Gene Center, LMU Munich, Munich 81377, Germany
| | - Cornelia A Deeg
- Chair of Physiology, Department of Veterinary Sciences, LMU Munich, Planegg 82152, Germany
| | - Helmut Blum
- Laboratory for Functional Genome Analysis, Gene Center, LMU Munich, Munich 81377, Germany
| | - Christian Schulz
- Medizinische Klinik und Poliklinik I, LMU Munich, Munich 81377, Germany
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich 80802, Germany
- Department of Immunopharmacology, Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, Heidelberg University, Mannheim 68167, Germany
| | - Dorota Zawada
- First Department of Medicine, Cardiology, TUM University Hospital, Technical University of Munich, School of Medicine and Health, Munich 81675, Germany
| | - Melanie Janda
- Chair of Molecular Animal Breeding and Biotechnology, Gene Center, LMU Munich, Munich 81377, Germany
| | | | - Karl Kunzelmann
- Institut für Physiologie, Universität Regensburg, Regensburg 93053, Germany
| | - Alessandra Moretti
- First Department of Medicine, Cardiology, TUM University Hospital, Technical University of Munich, School of Medicine and Health, Munich 81675, Germany
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich 80802, Germany
- Chair of Regenerative Medicine in Cardiovascular Disease, TUM University Hospital, Technical University of Munich, School of Medicine and Health, Munich 81675, Germany
| | - Karl-Ludwig Laugwitz
- First Department of Medicine, Cardiology, TUM University Hospital, Technical University of Munich, School of Medicine and Health, Munich 81675, Germany
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich 80802, Germany
| | - Christian Kupatt
- First Department of Medicine, Cardiology, TUM University Hospital, Technical University of Munich, School of Medicine and Health, Munich 81675, Germany
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich 80802, Germany
| | - Armin Saalmüller
- Institute of Immunology, University of Veterinary Medicine, Vienna 1210, Austria
| | - Thomas Fröhlich
- Laboratory for Functional Genome Analysis, Gene Center, LMU Munich, Munich 81377, Germany
| | - Eckhard Wolf
- Chair of Molecular Animal Breeding and Biotechnology, Gene Center, LMU Munich, Munich 81377, Germany
- Center of Innovative Medical Models (CiMM), LMU Munich, Oberschleissheim 85764, Germany
- Gene Center Munich, LMU Munich, Munich 81377, 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 13353, Germany
- German Center for Lung Research (DZL), associated partner site, Berlin 13353, Germany
- German Center for Child and Adolescent Health (DZKJ), partner site, Berlin 13353, Germany
| | - Lars Mundhenk
- Institute of Veterinary Pathology, Freie Universität Berlin, Berlin 14163, Germany
| | - Wilhelm Gerner
- Institute of Immunology, University of Veterinary Medicine, Vienna 1210, Austria
| | - Nikolai Klymiuk
- First Department of Medicine, Cardiology, TUM University Hospital, Technical University of Munich, School of Medicine and Health, Munich 81675, Germany
- Center of Innovative Medical Models (CiMM), LMU Munich, Oberschleissheim 85764, Germany
| |
Collapse
|
9
|
Liu CM, Fischer JL, Zemanick ET, Woods JC, Markarian KK, Fain SB, Froh D, Heltshe SL, Hoffman LR, Humphries SM, Kramer EL, Ode KL, Lewis M, Li DA, Mata J, Milla SS, Niedbalski PJ, Sawatzky BD, Sim MS, Sullivan JS, Trout AT, Goss CH, Taylor-Cousar JL, Beswick DM. The impact of highly effective modulator therapy on sinusitis and dysosmia in young children with cystic fibrosis: a prospective study protocol. ERJ Open Res 2025; 11:00137-2024. [PMID: 39811548 PMCID: PMC11726580 DOI: 10.1183/23120541.00137-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 07/10/2024] [Indexed: 01/16/2025] Open
Abstract
Background Chronic rhinosinusitis (CRS) and olfactory dysfunction (OD) are prevalent disease complications in people with cystic fibrosis. These understudied comorbidities significantly impact quality of life. The impact of highly effective modulator therapy (HEMT) in young children with cystic fibrosis (YCwCF) on these disease complications is unknown. This proposed study aims to characterise CRS and OD in YCwCF and assess the efficacy of HEMT in improving sinus and olfactory health in this young age group. Methods This six-centre, prospective, observational study will enrol 80 YCwCF aged 2-8 years. Patients are divided into two groups: those receiving HEMT and those not on HEMT based on clinical indication. Both groups undergo sinus magnetic resonance imaging, psychophysical olfactory tests, and complete patient- or parent-reported quality of life surveys over 2 years. Outcomes will be compared before and after initiation of HEMT and between groups. Ethical approval has been obtained for all sites, and this study has been registered on ClinicalTrials.gov (NCT06191640). Results Enrolment began in April 2023. 21 participants have been enrolled as of October 2023 with ongoing enrolment at all sites. Conclusion This investigation is expected to provide critical insights into the potential benefits of early HEMT initiation in managing CRS and OD in YCwCF. It will assist in developing targeted interventions and contribute to the understanding of HEMT's role in altering the disease course in this demographic.
Collapse
Affiliation(s)
- Christine M. Liu
- Department of Otolaryngology-Head and Neck Surgery, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jakob L. Fischer
- Department of Otolaryngology-Head and Neck Surgery, University of California, Los Angeles, Los Angeles, CA, USA
| | - Edith T. Zemanick
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, USA
- Children's Hospital Colorado, Breathing Institute, Aurora, CO, USA
| | - Jason C. Woods
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Karolin K. Markarian
- University of California, Los Angeles, CTSI, David Geffen School of Medicine, Los Angeles, CA, USA
| | - Sean B. Fain
- Department of Radiology, University of Iowa, Iowa City, IA, USA
| | - Deborah Froh
- Department of Pediatrics, University of Virginia, Charlottesville, VA, USA
| | - Sonya L. Heltshe
- Department of Pediatrics, University of Washington and Seattle Children's Research Institute, Seattle, WA, USA
| | - Lucas R. Hoffman
- Department of Pediatrics, University of Washington and Seattle Children's Research Institute, Seattle, WA, USA
- Department of Medicine and Pediatrics, University of Washington, Seattle, WA, USA
| | | | - Elizabeth L. Kramer
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Katie Larson Ode
- Department of Pediatrics, University of Iowa, Iowa City, IA, USA
| | - Michael Lewis
- Department of Pediatrics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Douglas A. Li
- Department of Pediatrics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jaime Mata
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, VA, USA
| | - Sarah S. Milla
- Department of Radiology, Children's Hospital Colorado, Aurora, CO, USA
- Department of Radiology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Peter J. Niedbalski
- Department of Pulmonary, Critical Care, and Sleep Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | | | - Myung-Shin Sim
- Department of Pediatrics, University of California, Los Angeles, Los Angeles, CA, USA
| | | | - Andrew T. Trout
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Radiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Christopher H. Goss
- Department of Medicine and Pediatrics, University of Washington, Seattle, WA, USA
| | - Jennifer L. Taylor-Cousar
- Department of Medicine, National Jewish Health, Aurora, CO, USA
- Department of Pediatrics, National Jewish Health, Aurora, CO, USA
| | - Daniel M. Beswick
- Department of Otolaryngology-Head and Neck Surgery, University of California, Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
10
|
Despotes KA, Ceppe AS, Goralski JL, Donaldson SH. New era, new GOALs: cardiovascular screening and lipid management in cystic fibrosis. Ther Adv Respir Dis 2025; 19:17534666251317200. [PMID: 39891563 PMCID: PMC11786282 DOI: 10.1177/17534666251317200] [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: 08/16/2024] [Accepted: 01/15/2025] [Indexed: 02/03/2025] Open
Abstract
BACKGROUND Cardiovascular disease (CVD) risks are increasing in people with cystic fibrosis (pwCF). While cholesterol levels were historically low in pwCF, higher levels after initiating highly effective modulator therapy (HEMT) have been reported. Mechanisms are unclear and there is little guidance on screening. OBJECTIVES To evaluate serum lipid changes at multiple timepoints after ivacaftor initiation, and to assess current screening practices for CVD risk factors among CF providers. DESIGN This was a post-hoc correlative analysis of prospectively collected clinical data and serum samples from the GOAL cohort study. Cross-sectional survey methodology was also employed. METHODS We evaluated serum lipids (total cholesterol (TC), low-density lipoprotein (LDL), and high-density lipoprotein (HDL)) at baseline, 3- and 18 months after ivacaftor initiation using samples from the GOAL study biorepository. We also surveyed CF providers across the United States on their CVD risk screening practices. RESULTS Fifty GOAL participants' samples were analyzed. Using the repeated measures model, TC significantly varied by visit (p = 0.004), driven by a significant increase from baseline at 3 months (mean difference 9.4 mg/dL). This difference diminished by 18 months. BMI was a significant covariate for TC. No significant differences by visit were detected in LDL or HDL. Seventy-five respondents participated in the survey (response rate 5.6%; 41 adult providers, 18 pediatric providers, and 10 providers caring for both) with 67% reporting no lipid screening policy existed in their center. In the past year, 29% of adult providers prescribed lipid-lowering therapy, 54% started anti-hypertensive medications, and 48% initiated ischemic cardiac evaluations for pwCF. CONCLUSION TC significantly increased within 3 months of initiating ivacaftor, but subsequently diminished toward baseline by 18 months. Lipid screening practices among CF providers were variable and providers are increasingly being confronted with managing CVD risk factors. Partnering with primary care providers is likely to become increasingly important in CF care models.
Collapse
Affiliation(s)
- Katherine A. Despotes
- Department of Pediatrics, School of Medicine, The University of North Carolina at Chapel Hill, 450 MacNider Building, Campus box #7217, 333 S. Columbia Street, Chapel Hill, NC 27599, USA
- Department of Medicine, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27514-4309, USA
| | - Agathe S. Ceppe
- Department of Medicine, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jennifer L. Goralski
- Department of Pediatrics, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Medicine, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Scott H. Donaldson
- Department of Medicine, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
11
|
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
|
12
|
Glaenzel U, Huth F, Eggimann F, Hackling M, Leuthold LA, Meissner A, Bebrevska L. Absorption, Distribution, Metabolism, and Excretion of Icenticaftor (QBW251) in Healthy Male Volunteers at Steady State and In Vitro Phenotyping of Major Metabolites. Drug Metab Dispos 2024; 52:1379-1387. [PMID: 39313328 DOI: 10.1124/dmd.124.001751] [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: 04/09/2024] [Revised: 09/03/2024] [Accepted: 09/09/2024] [Indexed: 09/25/2024] Open
Abstract
Icenticaftor (QBW251) is a potentiator of the cystic fibrosis transmembrane conductance regulator protein and is currently in clinical development for the treatment of chronic obstructive pulmonary disease and chronic bronchitis. An absorption, distribution, metabolism, and excretion study was performed at steady state to determine the pharmacokinetics, mass balance, and metabolite profiles of icenticaftor in humans. In this open-label study, six healthy men were treated with unlabeled oral icenticaftor (400 mg b.i.d.) for 4 days. A single oral dose of [14C]icenticaftor was administered on Day 5, and unlabeled icenticaftor was administered twice daily from the evening of Day 5 to Day 12. Unchanged icenticaftor accounted for 18.5% of plasma radioactivity. Moderate to rapid absorption of icenticaftor was observed (median time to reach peak or maximum concentration: 4 hours), with 93.4% of the dose absorbed. It exhibited moderate distribution (Vz/F: 335 L) and was extensively metabolized, principally through N-glucuronidation, O-glucuronidation, and/or O-demethylation. The metabolites M8 and M9, formed by N-glucuronidation and O-glucuronidation of icenticaftor, respectively, represented the main entities detected in plasma (35.3% and 14.5%, respectively) in addition to unchanged icenticaftor (18.5%). The apparent mean terminal half-life of icenticaftor was 15.4 hours in blood and 20.6 hours in plasma. Icenticaftor was eliminated from the body mainly through metabolism followed by renal excretion, and excretion of radioactivity was complete after 9 days. In vitro phenotyping of icenticaftor showed that cytochrome P450 and uridine diphosphate glucuronosyltransferase were responsible for 31% and 69% of the total icenticaftor metabolism in human liver microsomes, respectively. This study provided invaluable insights into the disposition of icenticaftor. SIGNIFICANCE STATEMENT: The absorption, distribution, metabolism, and excretion of a single radioactive oral dose of icenticaftor was evaluated at steady state to investigate the nonlinear pharmacokinetics observed previously with icenticaftor. [14C]Icenticaftor demonstrated good systemic availability after oral administration and was extensively metabolized and moderately distributed to peripheral tissues. The most abundant metabolites, M8 and M9, were formed by N-glucuronidation and O-glucuronidation of icenticaftor, respectively. Phenotyping demonstrated that [14C]icenticaftor was metabolized predominantly by UGT1A9 with a remarkably low Km value.
Collapse
Affiliation(s)
- Ulrike Glaenzel
- Novartis Pharma AG, Basel, Switzerland (U.G., F.H., F.E., L.A.L., A.M., L.B.) and Novartis Pharmaceuticals Corporation, East Hanover, New Jersey (M.H.)
| | - Felix Huth
- Novartis Pharma AG, Basel, Switzerland (U.G., F.H., F.E., L.A.L., A.M., L.B.) and Novartis Pharmaceuticals Corporation, East Hanover, New Jersey (M.H.)
| | - Fabian Eggimann
- Novartis Pharma AG, Basel, Switzerland (U.G., F.H., F.E., L.A.L., A.M., L.B.) and Novartis Pharmaceuticals Corporation, East Hanover, New Jersey (M.H.)
| | - Melissa Hackling
- Novartis Pharma AG, Basel, Switzerland (U.G., F.H., F.E., L.A.L., A.M., L.B.) and Novartis Pharmaceuticals Corporation, East Hanover, New Jersey (M.H.)
| | - Luc Alexis Leuthold
- Novartis Pharma AG, Basel, Switzerland (U.G., F.H., F.E., L.A.L., A.M., L.B.) and Novartis Pharmaceuticals Corporation, East Hanover, New Jersey (M.H.)
| | - Axel Meissner
- Novartis Pharma AG, Basel, Switzerland (U.G., F.H., F.E., L.A.L., A.M., L.B.) and Novartis Pharmaceuticals Corporation, East Hanover, New Jersey (M.H.)
| | - Lidiya Bebrevska
- Novartis Pharma AG, Basel, Switzerland (U.G., F.H., F.E., L.A.L., A.M., L.B.) and Novartis Pharmaceuticals Corporation, East Hanover, New Jersey (M.H.)
| |
Collapse
|
13
|
Keith JD, Murphree-Terry M, Bollar G, Oden AM, Doty IH, Birket SE. Ivacaftor ameliorates mucus burden, bacterial load, and inflammation in acute but not chronic P. aeruginosa infection in hG551D rats. Respir Res 2024; 25:397. [PMID: 39497082 PMCID: PMC11536857 DOI: 10.1186/s12931-024-03029-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 10/28/2024] [Indexed: 11/06/2024] Open
Abstract
BACKGROUND Newly approved highly effective modulation therapies (HEMT) have been life-changing for people with CF. Although these drugs have resulted in significant improvements in lung function and exacerbation rate, bacterial populations in the lung have not been eradicated. The mechanisms behind the continued colonization are not completely clear. METHODS We used a humanized rat to assess the effects of ivacaftor therapy on infection outcomes. Rats harbor an insert expressing humanized CFTR cDNA, including the G551D mutation. hG551D rats were treated with ivacaftor either during or before infection with P. aeruginosa. The response to infection was assessed by bacterial burden in the lung and mucus burden in the lung. RESULTS We found that hG551D rats treated with ivacaftor had reduced bacteria present in the lung in the acute phase of the infection but were not different than vehicle control in the chronic phase of the infection. Similarly, the percentage of neutrophils in the airways were reduced at the acute, but not chronic, timepoints. Overall weight data indicated that the hG551D rats had significantly better weight recovery during the course of infection when treated with ivacaftor. Potentiation of the G551D mutation with ivacaftor resultant in short-circuit current measurements equal to WT, even during the chronic phase of the infection. Despite the persistent infection, hG551D rats treated with ivacaftor had fewer airways with mucus plugs during the chronic infection. CONCLUSIONS The data indicate that the hG551D rats have better outcomes during infection when treated with ivacaftor compared to the vehicle group. Rats have increased weight gain, increased CFTR protein function, and decreased mucus accumulation, despite the persistence of infection and inflammation. These data suggest that ivacaftor improves tolerance of infection, rather than eradication, in this rat model.
Collapse
Affiliation(s)
- Johnathan D Keith
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, 1720 2nd Ave S, Birmingham, AL, 35294, USA
| | - Mikayla Murphree-Terry
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, 1720 2nd Ave S, Birmingham, AL, 35294, USA
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Gretchen Bollar
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, 1720 2nd Ave S, Birmingham, AL, 35294, USA
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ashley M Oden
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ian H Doty
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Susan E Birket
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, 1720 2nd Ave S, Birmingham, AL, 35294, USA.
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
14
|
Murphree-Terry M, Keith JD, Oden AM, Birket SE. Normalization of Muc5b ameliorates airway mucus plugging during persistent Pseudomonas aeruginosa infection in the CFTR -/- rat. Am J Physiol Lung Cell Mol Physiol 2024; 327:L672-L683. [PMID: 39316674 PMCID: PMC11563644 DOI: 10.1152/ajplung.00381.2023] [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: 12/04/2023] [Revised: 07/19/2024] [Accepted: 09/02/2024] [Indexed: 09/26/2024] Open
Abstract
In cystic fibrosis, the airway gel-forming mucin MUC5B accumulates in the airways, preventing clearance of pathogens like Pseudomonas aeruginosa (PA). The cystic fibrosis transmembrane conductance regulator (CFTR)-/- (KO) rat model exhibits a similar accumulation of Muc5b. Our lab has shown that increased Muc5b precipitates the development of chronic PA infection. We hypothesized that reducing Muc5b in the KO rat airway would prevent occlusive mucus plugs and development of persistent PA infection. Six-month-old KO rats received Muc5b or scramble siRNA via intratracheal instillation. Rats were then inoculated with 106 colony-forming units of mucoid P. aeruginosa isolate PAM57-15 and euthanized at 3- or 14-days post infection (dpi) to assess acute and persistent infection. At 14 dpi, Muc5b siRNA-treated KO rats had increased weight, decreased neutrophilic inflammation, and reduced mucus plugging in the small airways compared with scramble-treated KO and WT rats. These results indicate that pharmacological intervention of Muc5b reduces mucus plugging during persistent PA infection.NEW & NOTEWORTHY Although highly effective modulator therapies for cystic fibrosis (CF) have improved mucus-related outcomes of disease for people with CF, eradication of Pseudomonas aeruginosa (PA) infection has not been achieved in this population. In addition, current therapies for CF do not target mucin hypersecretion directly. Here, we show that a novel approach of normalizing airway Muc5b hypersecretion ameliorates infection-induced mucus plugging and neutrophilic inflammation during persistent PA infection in CFTR-/- rats.
Collapse
Affiliation(s)
- Mikayla Murphree-Terry
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Johnathan D Keith
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Ashley M Oden
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Susan E Birket
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, Alabama, United States
| |
Collapse
|
15
|
Milczewska J, Syunyaeva Z, Żabińska-Jaroń A, Sands D, Thee S. Changing profile of bacterial infection and microbiome in cystic fibrosis: when to use antibiotics in the era of CFTR-modulator therapy. Eur Respir Rev 2024; 33:240068. [PMID: 39631927 PMCID: PMC11615665 DOI: 10.1183/16000617.0068-2024] [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: 04/07/2024] [Accepted: 10/03/2024] [Indexed: 12/07/2024] Open
Abstract
The advent of cystic fibrosis transmembrane conductance regulator (CFTR) modulator therapy, especially the triple therapy combining the drugs elexacaftor, tezacaftor, ivacaftor (ETI), has significantly changed the course of the disease in people with cystic fibrosis (pwCF). ETI, which is approved for the majority (80-90%) of pwCF, partially restores CFTR channel function, resulting in improved mucociliary clearance and, consequently, improved lung function, respiratory symptoms and pulmonary exacerbations. The bacterial burden of classical CF pathogens such as Pseudomonas aeruginosa and Staphylococcus aureus is reduced without reaching eradication in the majority of infected patients. Limited data is available on less common or emerging bacterial pathogens. ETI has a positive effect on the lung microbiome but does not fully restore it to a healthy state. Due to the significant reduction in sputum production under ETI, respiratory samples such as deep-throat swabs are commonly taken, despite their inadequate representation of lower respiratory tract pathogens. Currently, there are still unanswered questions related to this new therapy, such as the clinical impact of infection with cystic fibrosis (CF) pathogens, the value of molecular diagnostic tests, the durability of the effects on respiratory infection and the role of fungal and viral infections. This article reviews the changes in bacterial lung infections and the microbiome in CF to provide evidence for the use of antibiotics in the era of ETI.
Collapse
Affiliation(s)
- Justyna Milczewska
- Cystic Fibrosis Department, Institute of Mother and Child, Warsaw, Poland
- Cystic Fibrosis Centre, Pediatric Hospital, Dziekanow Lesny, Poland
- Joint first authors
| | - Zulfiya Syunyaeva
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Joint first authors
| | | | - Dorota Sands
- Cystic Fibrosis Department, Institute of Mother and Child, Warsaw, Poland
- Cystic Fibrosis Centre, Pediatric Hospital, Dziekanow Lesny, Poland
| | - Stephanie Thee
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, 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
| |
Collapse
|
16
|
Vijaykumar K, Solomon GM, Guimbellot J, Acosta EP, Bhambhavni PG, White S, Kim H, Raju SV, Rasmussen LW, Harris N, Liu B, Hathorne H, Rowe SM, Dransfield MT. Ivacaftor for Chronic Obstructive Pulmonary Disease - Results from a Phase 2, Randomized Controlled Trial. Am J Respir Crit Care Med 2024; 211:823-831. [PMID: 39316773 DOI: 10.1164/rccm.202407-1302oc] [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: 07/03/2024] [Accepted: 09/23/2024] [Indexed: 09/26/2024] Open
Abstract
RATIONALE Patients with chronic obstructive pulmonary disease (COPD) exhibit acquired cystic fibrosis transmembrane conductance regulator (CFTR) dysfunction. CFTR modulators may improve outcomes in patients with COPD, although recent data regarding magnitude of benefit have been inconclusive and effects on mucociliary clearance are unknown. We conducted a phase 2, randomized, double blind placebo control trial to determine safety and tolerability, and explore the potential mechanism of ivacaftor for the treatment of COPD. METHODS We randomized 40 patients with moderate to severe COPD and symptoms of chronic bronchitis to ivacaftor (N=30) or placebo (N=10) 150mg BID for 12 weeks. Primary endpoints included evaluation of safety of ivacaftor and pharmacokinetics (PK). Secondary endpoints included measures of CFTR activity and clinical outcomes. RESULTS Ivacaftor was safe and tolerable with similar rates of adverse events rates between groups. Most common adverse event was diarrhea in the ivacaftor group and acute COPD exacerbation in the placebo group. PK analysis found the mean area under the curve over 12 hours (AUC12) to be 72% of the previously reported AUC12 in cystic fibrosis (CF). Treatment with ivacaftor did not improve sweat chloride, whole lung mucociliary clearance, lung function or respiratory symptoms. CONCLUSION Ivacaftor was safe and well tolerated, but did not improve measures of CFTR activity or mucus clearance. As serum concentrations achieved were lower than observed in CF at the same dose, and modulation of wild type CFTR differs from G551D, further dose determination studies are needed to better understand treatment efficacy of CFTR potentiators in COPD. Clinical trial registration available at www. CLINICALTRIALS gov, ID: NCT03085485.
Collapse
Affiliation(s)
| | - George M Solomon
- University of Alabama at Birmingham, Medicine, Gregory Fleming James Cystic Fibrosis Research Center, Birmingham, Alabama, United States
| | - Jennifer Guimbellot
- University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States
| | - Edward P Acosta
- The University of Alabama at Birmingham, Pharmacology and Toxicology, Birmingham, Alabama, United States
| | | | - Sharon White
- The University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Harrison Kim
- The University of Alabama at Birmingham, Department of Radiology, Birmingham, Alabama, United States
| | - S Vamsee Raju
- The University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Lawrence W Rasmussen
- The University of Alabama at Birmingham, Division of Pulmonary, Allergy, and Critical Care Medicine and the Gregory Fleming James Cystic Fibrosis Research Center, Birmingham, Alabama, United States
| | - Necole Harris
- The University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Bo Liu
- University of Alabama at Birmingham, Medicine, Gregory Fleming James Cystic Fibrosis Research Center, Birmingham, Alabama, United States
| | - Heather Hathorne
- The University of Alabama at Birmingham Heersink School of Medicine, Gregory Fleming James Cystic Fibrosis Research Center, Birmingham, Alabama, United States
| | - Steven M Rowe
- The University of Alabama at Birmingham, Medicine, Birmingham, Alabama, United States;
| | - Mark T Dransfield
- The University of Alabama at Birmingham, Medicine/Pulmonary, Allergy and Critical Care, Birmingham, Alabama, United States
| |
Collapse
|
17
|
Merlo CA, Thorat T, DerSarkissian M, McGarry LJ, Nguyen C, Gu YM, Healy J, Rubin JL, Brookhart MA. Long-term impact of ivacaftor on mortality rate and health outcomes in people with cystic fibrosis. Thorax 2024; 79:925-933. [PMID: 38937105 PMCID: PMC11503052 DOI: 10.1136/thorax-2023-220558] [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: 06/06/2023] [Accepted: 04/21/2024] [Indexed: 06/29/2024]
Abstract
BACKGROUND Ivacaftor (IVA) has been shown to improve lung function and other clinical outcomes in people with cystic fibrosis (CF). A decade of real-world IVA availability has enabled the examination of long-term outcomes with this treatment. This retrospective, longitudinal cohort study investigated the impact of IVA on mortality rate and health outcomes among people with CF in the US. METHODS Data from the US CF Foundation Patient Registry from January 2010 to December 2019 were analysed. The IVA-treated cohort included people with a CF transmembrane conductance regulator (CFTR) gating mutation (excluding R117H); age-matched comparator cohort included people with a F508del and a minimal function CFTR mutation who had no prior CFTR modulator treatment. Baseline characteristics were balanced between cohorts using standardised mortality ratio weighting generated from propensity scores. Outcomes of interest were overall survival, lung transplant, percent predicted forced expiratory volume in 1 s (ppFEV1), body mass index (BMI), pulmonary exacerbations (PEx), outpatient visits and hospitalisations. FINDINGS Over a maximum follow-up of 7.9 years, the IVA-treated cohort (N=736) had lower rates of mortality (hazard ratio [HR] (95% CI): 0.22 (0.09 to 0.45)), lung transplant (HR: 0.11 (95% CI 0.02 to 0.28)), PEx (rate ratio: 0.49 (95% CI 0.42 to 0.55)) and all-cause hospitalisations (rate ratio: 0.50 (95% CI 0.43 to 0.56)) as well as better lung function (mean difference in ppFEV1: 8.46 (95% CI 7.34 to 9.75)) and higher BMI/BMI z-scores (mean difference 1.20 (95% CI 0.92 to 1.71) kg/m2 and 0.27 (95% CI 0.25 to 0.40), respectively) than the comparator cohort (N=733). INTERPRETATION Our analysis suggests that IVA provides sustained clinical benefits in people with CF over a follow-up period of approximately 8 years. These findings reinforce the existing real-world evidence that IVA can slow disease progression and decrease the healthcare burden of CF over the long term.
Collapse
Affiliation(s)
| | - Teja Thorat
- Vertex Pharmaceuticals Incorporated, Boston, Massachusetts, USA
| | | | - Lisa J McGarry
- Vertex Pharmaceuticals Incorporated, Boston, Massachusetts, USA
| | | | - Yuqian M Gu
- Analysis Group Inc, Los Angeles, California, USA
| | - Joe Healy
- Vertex Pharmaceuticals Incorporated, Boston, Massachusetts, USA
| | - Jaime L Rubin
- Vertex Pharmaceuticals Incorporated, Boston, Massachusetts, USA
| | | |
Collapse
|
18
|
Roesch EA, Rahmaoui A, Lazarus RA, Konstan MW. The continuing need for dornase alfa for extracellular airway DNA hydrolysis in the era of CFTR modulators. Expert Rev Respir Med 2024; 18:677-691. [PMID: 39176450 DOI: 10.1080/17476348.2024.2394694] [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: 04/26/2024] [Revised: 08/11/2024] [Accepted: 08/16/2024] [Indexed: 08/24/2024]
Abstract
INTRODUCTION The availability of cystic fibrosis transmembrane conductance regulator (CFTR) modulators opens the possibility of discontinuing some chronic pulmonary therapies to decrease cystic fibrosis (CF) treatment burden. However, CFTR modulators may not adequately address neutrophilic inflammation, which contributes to a self-perpetual cycle of viscous CF sputum, airway obstruction, inflammation, and lung function decline. AREAS COVERED This review discusses the emerging role of neutrophil extracellular traps in CF and its role in CF sputum viscosity, airway obstruction, and inflammation, based on a literature search of PubMed (1990-present). We summarize clinical trials and real-world studies that support the efficacy of dornase alfa (Pulmozyme) in improving lung function and reducing pulmonary exacerbation in people with CF (PwCF), and we discuss the potential role of dornase alfa in reducing airway inflammation. We also examine the findings of short-term trials evaluating the discontinuation of mucoactive therapy in PwCF receiving CFTR modulators. EXPERT OPINION Long-term studies are needed to assess the impact of discontinuing mucoactive therapy in PwCF who are clinically stable while receiving CFTR modulatory therapy. Treatment decisions should take into account the severity of underlying lung disease. People with advanced CF will likely require ongoing mucoactive therapy.
Collapse
Affiliation(s)
- Erica A Roesch
- Department of Pediatrics, Rainbow Babies and Children's Hospital and Case Western Reserve University, Cleveland, OH, USA
| | | | - Robert A Lazarus
- Departments of Biological Chemistry and Early Discovery Biochemistry, Genentech, Inc., South San Francisco, CA, USA
| | - Michael W Konstan
- Department of Pediatrics, Rainbow Babies and Children's Hospital and Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
19
|
Baker E, Harris WT, Guimbellot JS, Bliton K, Rowe SM, Raju SV, Oates GR. Association between biomarkers of tobacco smoke exposure and clinical efficacy of ivacaftor in the G551D observational trial (GOAL). J Cyst Fibros 2024; 23:959-966. [PMID: 39033068 PMCID: PMC11410542 DOI: 10.1016/j.jcf.2024.07.010] [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: 04/20/2024] [Revised: 06/15/2024] [Accepted: 07/15/2024] [Indexed: 07/23/2024]
Abstract
BACKGROUND Acrolein, an aldehyde in smoke from tobacco products, inhibits CFTR function in vitro. Ivacaftor is an FDA-approved potentiator that improves mutant CFTR function. This human clinical study investigated the relationship between two urinary markers of tobacco smoke exposure - the acrolein metabolite 3-HPMA and the nicotine metabolite NNAL - and sweat chloride response to ivacaftor in the G551D Observational Trial (GOAL). METHODS 3-HPMA (low: <50th centile; moderate: 50-75th centile; high: >75th centile) and NNAL (detectable/undetectable) in GOAL samples was quantified with LC-MS/MS. Self-report of tobacco smoke exposure (Y/N) served as a subjective measure. Change in sweat chloride from pre- to 6 months post-ivacaftor treatment (ΔSC) was the primary CFTR-dependent readout. RESULTS The sample included 151 individuals, mean age 20.7 (SD 11.4) years, range 6-59 years. Smoke exposure prevalence was 15 % per self-reports but 27 % based on detectable NNAL. 3-HPMA was increased in those reporting tobacco smoke exposure (607 vs 354 ng/ml, p = 0.008), with a higher proportion of smoke-exposed in the high- vs low-acrolein group (31 % vs 9 %, p=0.040). Compared to low-acrolein counterparts, high-acrolein participants experienced less decrease in sweat chloride (-35.2 vs -48.2 mmol/L; p = 0.020) and had higher sweat chloride values (50.6 vs 37.6 mmol/L; p = 0.020) 6 months post-ivacaftor. The odds of ivacaftor-mediated potentiation to near normative CFTR function (defined as SC6mo <40 mmol/L) was more than twice as high in the low-acrolein cohort (OR: 2.51, p = 0.026). CONCLUSIONS Increased urinary 3-HPMA, an acrolein metabolite of tobacco smoke, is associated with a diminished sweat chloride response to ivacaftor potentiation of CFTR function.
Collapse
Affiliation(s)
- Elizabeth Baker
- Medicine University of Alabama at Birmingham 1808 7th Ave S, BDB 853 Birmingham, AL 35233 United States
| | - William T Harris
- Medicine University of Alabama at Birmingham 1808 7th Ave S, BDB 853 Birmingham, AL 35233 United States
| | - Jennifer S Guimbellot
- Medicine University of Alabama at Birmingham 1808 7th Ave S, BDB 853 Birmingham, AL 35233 United States; The University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Kyle Bliton
- Medicine University of Alabama at Birmingham 1808 7th Ave S, BDB 853 Birmingham, AL 35233 United States
| | - Steven M Rowe
- Medicine University of Alabama at Birmingham 1808 7th Ave S, BDB 853 Birmingham, AL 35233 United States
| | - S Vamsee Raju
- Medicine University of Alabama at Birmingham 1808 7th Ave S, BDB 853 Birmingham, AL 35233 United States
| | - Gabriela R Oates
- Medicine University of Alabama at Birmingham 1808 7th Ave S, BDB 853 Birmingham, AL 35233 United States.
| |
Collapse
|
20
|
Green N, Chan C, Ooi CY. The gastrointestinal microbiome, small bowel bacterial overgrowth, and microbiome modulators in cystic fibrosis. Pediatr Pulmonol 2024; 59 Suppl 1:S70-S80. [PMID: 39105345 DOI: 10.1002/ppul.26913] [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: 08/27/2023] [Revised: 01/12/2024] [Accepted: 01/31/2024] [Indexed: 08/07/2024]
Abstract
People with cystic fibrosis (pwCF) have an altered gastrointestinal microbiome. These individuals also demonstrate propensity toward developing small intestinal bacterial overgrowth (SIBO). The dysbiosis present has intestinal and extraintestinal implications, including potential links with the higher rates of gastrointestinal malignancies described in CF. Given these implications, there is growing interest in therapeutic options for microbiome modulation. Alternative therapies, including probiotics and prebiotics, and current CF transmembrane conductance regulator gene modulators are promising interventions for ameliorating gut microbiome dysfunction in pwCF. This article will characterize and discuss the current state of knowledge and expert opinions on gut dysbiosis and SIBO in the context of CF, before reviewing the current evidence supporting gut microbial modulating therapies in CF.
Collapse
Affiliation(s)
- Nicole Green
- Department of Pediatrics, Division of Gastroenterology and Hepatology, Seattle Children's Hospital, University of Washington, Seattle, Washington, USA
| | - Christopher Chan
- Discipline of Paediatrics & Child Health, Randwick Clinical Campus, School of Clinical Medicine, UNSW Medicine & Health, University of New South Wales, Sydney, New South Wales, Australia
| | - Chee Y Ooi
- Discipline of Paediatrics & Child Health, Randwick Clinical Campus, School of Clinical Medicine, UNSW Medicine & Health, University of New South Wales, Sydney, New South Wales, Australia
- Department of Gastroenterology, Sydney Children's Hospital, Randwick, New South Wales, Australia
| |
Collapse
|
21
|
Magaret AS, Graham E, Caverly LJ, Cromwell EA, Paynter A, Rosenfeld M, Thornton CS, Goss CH. Impact of day-to-day variation in FEV1 on measures of change: A conceptual description. J Cyst Fibros 2024; 23:943-946. [PMID: 39147620 PMCID: PMC11410506 DOI: 10.1016/j.jcf.2024.07.005] [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: 04/06/2024] [Revised: 06/20/2024] [Accepted: 07/09/2024] [Indexed: 08/17/2024]
Abstract
Clinical trials often demonstrate treatment efficacy through change in forced expiratory volume in one second (FEV1), comparing single FEV1 measurements from post- versus pre-treatment timepoints. Day-to-day variation in measured FEV1 is common for reasons such as diurnal variation and intermittent health changes, relative to a stable, monthly average. This variation can alter estimation of associations between change in FEV1 and baseline in predictable ways, through a phenomenon called regression to the mean. We quantify and explain day-to-day variation in percent-predicted FEV1 (ppFEV1) from 4 previous trials, and we present a statistical, data-driven explanation for potential bias in ceiling and floor effects due to commonly observed amounts of variation. We recommend accounting for variation when assessing associations between baseline value and change in CF outcomes in single-arm trials, and we consider possible impact of variation on conventional standards for study eligibility.
Collapse
Affiliation(s)
- Amalia S Magaret
- Depts. of Biostatistics, at the University of Washington, Seattle, WA, USA; Pediatrics, at the University of Washington, Seattle, WA, USA; Seattle Children's Research Institute, Seattle, WA, USA.
| | - Ellen Graham
- Depts. of Biostatistics, at the University of Washington, Seattle, WA, USA
| | - Lindsay J Caverly
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI, USA
| | | | - Alex Paynter
- Seattle Children's Research Institute, Seattle, WA, USA
| | - Margaret Rosenfeld
- Pediatrics, at the University of Washington, Seattle, WA, USA; Seattle Children's Research Institute, Seattle, WA, USA
| | - Christina S Thornton
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI, USA; Department of Medicine, University of Calgary, Calgary, AB, CAN
| | - Christopher H Goss
- Medicine, at the University of Washington, Seattle, WA, USA; Seattle Children's Research Institute, Seattle, WA, USA
| |
Collapse
|
22
|
Cordery C, Craddock J, Malý M, Basavaraja K, Webb JS, Walsh MA, Tews I. Control of phosphodiesterase activity in the regulator of biofilm dispersal RbdA from Pseudomonas aeruginosa. RSC Chem Biol 2024:d4cb00113c. [PMID: 39247681 PMCID: PMC11372557 DOI: 10.1039/d4cb00113c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 08/26/2024] [Indexed: 09/10/2024] Open
Abstract
The switch between planktonic and biofilm lifestyle correlates with intracellular concentration of the second messenger bis-(3'-5')-cyclic dimeric guanosine monophosphate (c-di-GMP). While bacteria possess cyclase and phosphodiesterase enzymes to catalyse formation or hydrolysis of c-di-GMP, both enzymatic domains often occur in a single protein. It is tacitly assumed that one of the two enzymatic activities is dominant, and that additional domains and protein interactions enable responses to environmental conditions and control activity. Here we report the structure of the phosphodiesterase domain of the membrane protein RbdA (regulator of biofilm dispersal) in a dimeric, activated state and show that phosphodiesterase activity is controlled by the linked cyclase. The phosphodiesterase region around helices α5/α6 forms the dimer interface, providing a rationale for activation, as this region was seen in contact with the cyclase domain in an auto-inhibited structure previously described. Kinetic analysis supports this model, as the activity of the phosphodiesterase alone is lower when linked to the cyclase. Analysis of a computed model of the RbdA periplasmatic domain reveals an all-helical architecture with a large binding pocket that could accommodate putative ligands. Unravelling the regulatory circuits in multi-domain phosphodiesterases like RbdA is important to develop strategies to manipulate or disperse bacterial biofilms.
Collapse
Affiliation(s)
- Charlotte Cordery
- Biological Sciences, Institute for Life Sciences, University of Southampton Southampton SO17 1BJ UK
- National Biofilms Innovation Centre, University of Southampton Southampton SO17 1BJ UK
- Diamond Light Source, Harwell Science and Innovation Campus Didcot Oxfordshire OX11 0DE UK
- Research Complex at Harwell, Harwell Science and Innovation Campus Didcot Oxfordshire OX11 0FA UK
| | - Jack Craddock
- Biological Sciences, Institute for Life Sciences, University of Southampton Southampton SO17 1BJ UK
- National Biofilms Innovation Centre, University of Southampton Southampton SO17 1BJ UK
| | - Martin Malý
- Biological Sciences, Institute for Life Sciences, University of Southampton Southampton SO17 1BJ UK
| | - Kieran Basavaraja
- Biological Sciences, Institute for Life Sciences, University of Southampton Southampton SO17 1BJ UK
- National Biofilms Innovation Centre, University of Southampton Southampton SO17 1BJ UK
- Diamond Light Source, Harwell Science and Innovation Campus Didcot Oxfordshire OX11 0DE UK
- Research Complex at Harwell, Harwell Science and Innovation Campus Didcot Oxfordshire OX11 0FA UK
| | - Jeremy S Webb
- Biological Sciences, Institute for Life Sciences, University of Southampton Southampton SO17 1BJ UK
- National Biofilms Innovation Centre, University of Southampton Southampton SO17 1BJ UK
| | - Martin A Walsh
- Diamond Light Source, Harwell Science and Innovation Campus Didcot Oxfordshire OX11 0DE UK
- Research Complex at Harwell, Harwell Science and Innovation Campus Didcot Oxfordshire OX11 0FA UK
| | - Ivo Tews
- Biological Sciences, Institute for Life Sciences, University of Southampton Southampton SO17 1BJ UK
- National Biofilms Innovation Centre, University of Southampton Southampton SO17 1BJ UK
| |
Collapse
|
23
|
Fuchs T, Zlamy M, Zöggeler T, Appelt D, Niedermayr K, Siedl A, Gasser V, Eder J, Ellemunter H. Detection of cytokines in nasal lavage samples of patients with cystic fibrosis: comparison of two different cytokine detection assays. BMC Pulm Med 2024; 24:286. [PMID: 38890643 PMCID: PMC11186286 DOI: 10.1186/s12890-024-03103-9] [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: 11/20/2023] [Accepted: 06/14/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND Cystic fibrosis (CF) is a genetic multisystem disorder. Inflammatory processes, which presumably begin early in infancy, play a crucial role in the progression of the disease. The detection of inflammatory biomarkers, especially in the airways, has therefore gained increasing attention. Due to improved treatment options, patients with CF produce less sputum. Nasal lavage samples therefore represent a promising alternative to induced sputum or bronchoalveolar lavage specimens. However, methodology of cytokine measurements is not standardised and comparisons of results are therefore often difficult. The aim of this study was to identify suitable detection methods of cytokines in nasal lavage samples by comparison of two different assays. METHODS Nasal lavage samples were obtained from the same patient at the same time by trained respiratory physiotherapists using a disposable syringe and 10 ml of 0.9% sodium chloride per nostril during outpatient visits. The cytokines IL-17 A, IL-2, IL-6 and IL-10 were measured using two different assays (BD™ and Milliplex®), which have already been applied in sputum and nasal lavage samples, despite different lower detection limits. RESULTS 22 participants were included in the study. In 95.5% of measurements, values were below the limit of detection with respect to the BD™ assay. Only IL-6 could be detected in approximately half of the patients. Individual cytokine levels were considerably higher when measured with Milliplex®, which is also reflected in a statistically significant manner (p = < 0.01). CONCLUSION The right choice of analysis method is crucial for measuring inflammatory markers in nasal lavage samples. Compared to the literature, Milliplex® showed higher detection rates and similar concentrations to other studies. TRIAL REGISTRATION Ethics approval was obtained from the ethics committee at Medical University of Innsbruck (EK Nr: 1055/2022).
Collapse
Affiliation(s)
- Teresa Fuchs
- Department of Pediatrics III, Cystic Fibrosis Centre Innsbruck, Medical University of Innsbruck, Innsbruck, Austria.
| | - Manuela Zlamy
- Tiroler Gesundheitsfond, Amt der Tiroler Landesregierung, Innsbruck, Austria
| | - Thomas Zöggeler
- Department of Pediatrics I, Medical University of Innsbruck, Innsbruck, Austria
| | - Dorothea Appelt
- Department of Pediatrics III, Cystic Fibrosis Centre Innsbruck, Medical University of Innsbruck, Innsbruck, Austria
| | - Katharina Niedermayr
- Department of Pediatrics I, Cystic Fibrosis Centre Innsbruck, Tirol Kliniken, Innsbruck, Austria
| | - Anja Siedl
- Department of Pediatrics I, Cystic Fibrosis Centre Innsbruck, Tirol Kliniken, Innsbruck, Austria
| | - Verena Gasser
- Department of Pediatrics I, Cystic Fibrosis Centre Innsbruck, Tirol Kliniken, Innsbruck, Austria
| | - Johannes Eder
- Department of Pediatrics I, Cystic Fibrosis Centre Innsbruck, Tirol Kliniken, Innsbruck, Austria
| | - Helmut Ellemunter
- Department of Pediatrics III, Cystic Fibrosis Centre Innsbruck, Medical University of Innsbruck, Innsbruck, Austria
- Medical University of Innsbruck, Austria, Medical Research Affiliate, Innsbruck, Austria
| |
Collapse
|
24
|
Rehman T, Pezzulo AA, Thurman AL, Zemans RL, Welsh MJ. Epithelial responses to CFTR modulators are improved by inflammatory cytokines and impaired by antiinflammatory drugs. JCI Insight 2024; 9:e181836. [PMID: 38888974 PMCID: PMC11383177 DOI: 10.1172/jci.insight.181836] [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: 04/08/2024] [Accepted: 06/12/2024] [Indexed: 06/20/2024] Open
Abstract
Cystic fibrosis (CF) is a genetic disorder that disrupts CF transmembrane conductance regulator (CFTR) anion channels and impairs airway host defenses. Airway inflammation is ubiquitous in CF, and suppressing it has generally been considered to improve outcomes. However, the role of inflammation in people taking CFTR modulators, small-molecule drugs that restore CFTR function, is not well understood. We previously showed that inflammation enhances the efficacy of CFTR modulators. To further elucidate this relationship, we treated human ΔF508-CF epithelia with TNF-α and IL-17, two inflammatory cytokines that are elevated in CF airways. TNF-α+IL-17 enhanced CFTR modulator-evoked anion secretion through mechanisms that raise intracellular Cl- (Na+/K+/2Cl- cotransport) and HCO3- (carbonic anhydrases and Na+/HCO3- cotransport). This enhancement required p38 MAPK signaling. Importantly, CFTR modulators did not affect CF airway surface liquid viscosity under control conditions but prevented the rise in viscosity in epithelia treated with TNF-α+IL-17. Finally, antiinflammatory drugs limited CFTR modulator responses in TNF-α+IL-17-treated epithelia. These results provide critical insights into mechanisms by which inflammation increases responses to CFTR modulators. They also suggest an equipoise between potential benefits and limitations of suppressing inflammation in people taking modulators, call into question current treatment approaches, and highlight a need for additional studies.
Collapse
Affiliation(s)
- Tayyab Rehman
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
- Department of Internal Medicine, Pappajohn Biomedical Institute, University of Iowa, Iowa City, Iowa, USA
| | - Alejandro A. Pezzulo
- Department of Internal Medicine, Pappajohn Biomedical Institute, University of Iowa, Iowa City, Iowa, USA
| | - Andrew L. Thurman
- Department of Internal Medicine, Pappajohn Biomedical Institute, University of Iowa, Iowa City, Iowa, USA
| | - Rachel L. Zemans
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Michael J. Welsh
- Department of Internal Medicine, Pappajohn Biomedical Institute, University of Iowa, Iowa City, Iowa, USA
- Howard Hughes Medical Institute, University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
25
|
Bass R, Tanes C, Bittinger K, Li Y, Lee H, Friedman ES, Koo I, Patterson AD, Liu Q, Wu GD, Stallings VA. Changes in fecal lipidome after treatment with ivacaftor without changes in microbiome or bile acids. J Cyst Fibros 2024; 23:481-489. [PMID: 37813785 PMCID: PMC10998923 DOI: 10.1016/j.jcf.2023.09.010] [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: 03/13/2023] [Revised: 08/28/2023] [Accepted: 09/20/2023] [Indexed: 10/11/2023]
Abstract
BACKGROUND Alterations in gastrointestinal health are prominent manifestations of cystic fibrosis (CF) and can independently impact pulmonary function. Ivacaftor has been associated with robust improvements in pulmonary function and weight gain, but less is known about the impact of ivacaftor on the fecal microbiome, lipidome, and bile acids. METHODS Stool samples from 18 patients with CF and gating mutations (ages 6-61 years, 13 pancreatic insufficient) were analyzed for fecal microbiome and lipidome composition as well as bile acid concentrations at baseline and after 3 months of treatment with ivacaftor. Microbiome composition was also assessed in a healthy reference cohort. RESULTS Alpha and beta diversity of the microbiome were different between CF and reference cohort at baseline, but no treatment effect was seen in the CF cohort between baseline and 3 months. Seven lipids increased with treatment. No differences were seen in bile acid concentrations after treatment in CF. At baseline, 403 lipids and unconjugated bile acids were different between pancreatic insufficient (PI-CF) and sufficient (PS-CF) groups and 107 lipids were different between PI-CF and PS-CF after 3 months of treatment. CONCLUSIONS The composition and diversity of the fecal microbiome were different in CF as compared to a healthy reference, and did not change after 3 months of ivacaftor. We detected modest differences in the fecal lipidome with treatment. Differences in lipid and bile acid profiles between PS-CF and PI-CF were attenuated after 3 months of treatment.
Collapse
Affiliation(s)
- Rosara Bass
- Department of Pediatrics, Division of Gastroenterology, Hepatology and Nutrition, Children's Hospital of Philadelphia, 3401 Civic Center Blvd, Philadelphia, PA, 19104, USA; Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, Philadelphia, PA, 19104, USA.
| | - Ceylan Tanes
- Department of Pediatrics, Division of Gastroenterology, Hepatology and Nutrition, Children's Hospital of Philadelphia, 3401 Civic Center Blvd, Philadelphia, PA, 19104, USA
| | - Kyle Bittinger
- Department of Pediatrics, Division of Gastroenterology, Hepatology and Nutrition, Children's Hospital of Philadelphia, 3401 Civic Center Blvd, Philadelphia, PA, 19104, USA
| | - Yun Li
- Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, 423 Guardian Dr., Philadelphia, PA 19104, USA
| | - Hongzhe Lee
- Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, 423 Guardian Dr., Philadelphia, PA 19104, USA
| | - Elliot S Friedman
- Division of Gastroenterology and Hepatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 3400 Civic Center Blvd, Philadelphia, PA, 19104, USA
| | - Imhoi Koo
- Department of Veterinary and Biomedical Sciences, Pennsylvania State University, 322 Life Sciences Building, University Park, PA 16802, USA
| | - Andrew D Patterson
- Department of Veterinary and Biomedical Sciences, Pennsylvania State University, 322 Life Sciences Building, University Park, PA 16802, USA
| | - Qing Liu
- Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Gary D Wu
- Division of Gastroenterology and Hepatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 3400 Civic Center Blvd, Philadelphia, PA, 19104, USA
| | - Virginia A Stallings
- Department of Pediatrics, Division of Gastroenterology, Hepatology and Nutrition, Children's Hospital of Philadelphia, 3401 Civic Center Blvd, Philadelphia, PA, 19104, USA; Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, Philadelphia, PA, 19104, USA
| |
Collapse
|
26
|
Horati H, Margaroli C, Chandler JD, Kilgore MB, Manai B, Andrinopoulou ER, Peng L, Guglani L, Tiddens HAMW, Caudri D, Scholte BJ, Tirouvanziam R, Janssens HM. Key inflammatory markers in bronchoalveolar lavage predict bronchiectasis progression in young children with CF. J Cyst Fibros 2024; 23:450-456. [PMID: 38246828 DOI: 10.1016/j.jcf.2024.01.002] [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: 09/14/2023] [Revised: 01/02/2024] [Accepted: 01/05/2024] [Indexed: 01/23/2024]
Abstract
INTRODUCTION Inflammation appears early in cystic fibrosis (CF) pathogenesis, with specific elevated inflammatory markers in bronchoalveolar lavage fluid (BALF) correlating with structural lung disease. Our aim was to identify markers of airway inflammation able to predict bronchiectasis progression over two years with high sensitivity and specificity. METHODS Children with CF with two chest computed tomography (CT) scans and bronchoscopies at a two-year interval were included (n= 10 at 1 and 3 years and n= 27 at 3 and 5 years). Chest CTs were scored for increase in bronchiectasis (Δ%Bx), using the PRAGMA-CF score. BALF collected with the first CT scan were analyzed for neutrophil% (n= 36), myeloperoxidase (MPO) (n= 25), neutrophil elastase (NE) (n= 26), and with a protein array for inflammatory and fibrotic markers (n= 26). RESULTS MPO, neutrophil%, and inducible T-cell costimulator ligand (ICOSLG), but not clinical characteristics, correlated significantly with Δ%Bx. Evaluation of neutrophil%, NE, MPO, interleukin-8 (IL-8), ICOSLG, and hepatocyte growth factor (HGF), for predicting an increase of > 0.5% of Δ%Bx in two years, showed that IL-8 had the best sensitivity (82%) and specificity (73%). Neutrophil%, ICOSLG and HGF had sensitivities of 85, 82, and 82% and specificities of 59, 67 and 60%, respectively. The odds ratio for risk of >0.5% Δ%Bx was higher for IL-8 (12.4) than for neutrophil%, ICOSLG, and HGF (5.9, 5.3, and 6.7, respectively). Sensitivity and specificity were lower for NE and MPO). CONCLUSIONS High levels of IL-8, neutrophil%, ICOSGL and HGF in BALF may be good predictors for progression of bronchiectasis in young children with CF.
Collapse
Affiliation(s)
- Hamed Horati
- Department of Pediatrics, Division of Respiratory Medicine and Allergology, Erasmus MC-Sophia Children's Hospital, University Hospital Rotterdam, I-BALL program, office Sp3456 Dr. Molewaterplein 40, 3015 GD Rotterdam, Postal address: Box 2060, Rotterdam 3000 CB, The Netherlands
| | - Camilla Margaroli
- Department of Pediatrics, Emory University School of Medicine & Center for CF and Airways Disease Research, Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Joshua D Chandler
- Department of Pediatrics, Emory University School of Medicine & Center for CF and Airways Disease Research, Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Matthew B Kilgore
- Department of Pediatrics, Emory University School of Medicine & Center for CF and Airways Disease Research, Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Badies Manai
- Department of Pediatrics, Division of Respiratory Medicine and Allergology, Erasmus MC-Sophia Children's Hospital, University Hospital Rotterdam, I-BALL program, office Sp3456 Dr. Molewaterplein 40, 3015 GD Rotterdam, Postal address: Box 2060, Rotterdam 3000 CB, The Netherlands
| | - Eleni-Rosalina Andrinopoulou
- Department of Biostatistics and Bioinformatics, Erasmus MC, University Hospital Rotterdam, Rotterdam, The Netherlands
| | - Limin Peng
- Department of Biostatistics and Bioinformatics, Emory University School of Public Health, Atlanta, GA, USA
| | - Lokesh Guglani
- Department of Pediatrics, Emory University School of Medicine & Center for CF and Airways Disease Research, Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Harm A M W Tiddens
- Department of Pediatrics, Division of Respiratory Medicine and Allergology, Erasmus MC-Sophia Children's Hospital, University Hospital Rotterdam, I-BALL program, office Sp3456 Dr. Molewaterplein 40, 3015 GD Rotterdam, Postal address: Box 2060, Rotterdam 3000 CB, The Netherlands; Department of radiology, Erasmus MC, University Hospital Rotterdam, Rotterdam, The Netherlands; Thirona, Nijmegen, The Netherlands
| | - Daan Caudri
- Department of Pediatrics, Division of Respiratory Medicine and Allergology, Erasmus MC-Sophia Children's Hospital, University Hospital Rotterdam, I-BALL program, office Sp3456 Dr. Molewaterplein 40, 3015 GD Rotterdam, Postal address: Box 2060, Rotterdam 3000 CB, The Netherlands
| | - Bob J Scholte
- Department of Pediatrics, Division of Respiratory Medicine and Allergology, Erasmus MC-Sophia Children's Hospital, University Hospital Rotterdam, I-BALL program, office Sp3456 Dr. Molewaterplein 40, 3015 GD Rotterdam, Postal address: Box 2060, Rotterdam 3000 CB, The Netherlands; Department of Cell Biology, Erasmus MC, University Hospital Rotterdam, Rotterdam, The Netherlands
| | - Rabindra Tirouvanziam
- Department of Pediatrics, Emory University School of Medicine & Center for CF and Airways Disease Research, Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Hettie M Janssens
- Department of Pediatrics, Division of Respiratory Medicine and Allergology, Erasmus MC-Sophia Children's Hospital, University Hospital Rotterdam, I-BALL program, office Sp3456 Dr. Molewaterplein 40, 3015 GD Rotterdam, Postal address: Box 2060, Rotterdam 3000 CB, The Netherlands.
| |
Collapse
|
27
|
Han X, Li D, Zhu Y, Schneider-Futschik EK. Recommended Tool Compounds for Modifying the Cystic Fibrosis Transmembrane Conductance Regulator Channel Variants. ACS Pharmacol Transl Sci 2024; 7:933-950. [PMID: 38633590 PMCID: PMC11019735 DOI: 10.1021/acsptsci.3c00362] [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: 12/14/2023] [Revised: 02/20/2024] [Accepted: 02/23/2024] [Indexed: 04/19/2024]
Abstract
Cystic fibrosis (CF) is a genetic disorder arising from variations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene, leading to multiple organ system defects. CFTR tool compounds are molecules that can modify the activity of the CFTR channel. Especially, patients that are currently not able to benefit from approved CFTR modulators, such as patients with rare CFTR variants, benefit from further research in discovering novel tools to modulate CFTR. This Review explores the development and classification of CFTR tool compounds, including CFTR blockers (CFTRinh-172, GlyH-101), potentiators (VRT-532, Genistein), correctors (VRT-325, Corr-4a), and other approved and unapproved modulators, with detailed descriptions and discussions for each compound. The challenges and future directions in targeting rare variants and optimizing drug delivery, and the potential synergistic effects in combination therapies are outlined. CFTR modulation holds promise not only for CF treatment but also for generating CF models that contribute to CF research and potentially treating other diseases such as secretory diarrhea. Therefore, continued research on CFTR tool compounds is critical.
Collapse
Affiliation(s)
- XiaoXuan Han
- Department of Biochemistry & Pharmacology,
School of Biomedical Sciences, Faculty of Medicine, Dentistry and
Health Sciences, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Danni Li
- Department of Biochemistry & Pharmacology,
School of Biomedical Sciences, Faculty of Medicine, Dentistry and
Health Sciences, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Yimin Zhu
- Department of Biochemistry & Pharmacology,
School of Biomedical Sciences, Faculty of Medicine, Dentistry and
Health Sciences, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Elena K. Schneider-Futschik
- Department of Biochemistry & Pharmacology,
School of Biomedical Sciences, Faculty of Medicine, Dentistry and
Health Sciences, The University of Melbourne, Parkville, VIC 3010, Australia
| |
Collapse
|
28
|
Duehlmeyer S, Elson EC, Oermann CM. Effect of Elexacaftor/Tezacaftor/Ivacaftor on Pseudomonas aeruginosa Acquisition and Chronic Infection at a Single Pediatric Cystic Fibrosis Care Center. J Pediatr Pharmacol Ther 2024; 29:135-139. [PMID: 38596420 PMCID: PMC11001205 DOI: 10.5863/1551-6776-29.2.135] [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: 10/25/2022] [Accepted: 03/29/2023] [Indexed: 04/11/2024]
Abstract
OBJECTIVES As cystic fibrosis (CF) lung disease progresses, the airways become infected with opportunistic pathogens, such as Pseudomonas aeruginosa (PA). In October 2019, the US Food and Drug Administration approved elexacaftor/tezacaftor/ivacaftor (ETI), a highly effective modulator therapy (HEMT), for individuals 12 years and older with 1 copy of the F508del cystic fibrosis transmembrane conductance regulator (CFTR) mutation. ETI increases the amount of and function of CFTR in the respiratory epithelium, improving mucociliary clearance and reducing static airway mucus, a major trigger for chronic infection and inflammation. METHODS A retrospective analysis of inhaled tobramycin (iTOB) prescriptions between January 1, 2016, and December 31, 2021, was performed. This captured data before and after ETI approval at Children's Mercy Kansas City (CMKC). The number of individuals with new PA acquisition and individuals considered -chronically infected was analyzed. RESULTS The number of eradication prescriptions declined in 2020 and 2021, with 15 (7%) and 12 (5%) -individuals prescribed therapy for those years, respectively. A similar pattern was observed for -prescriptions for chronic infection. A reduction was seen in 2020 and 2021, with 28 (13%) and 20 (9%) individuals -prescribed therapy for the respective years. CONCLUSIONS The CMKC experienced a decrease in the number of courses of iTOB prescribed during the last 6 years. The reasons for this are likely multifactorial and may include the implementation of standardized PA surveillance and eradication protocols, the effect of HEMT on mucociliary clearance and airway microbiology, and the poorly understood effects of the SARS-CoV-2 pandemic on the epidemiology of respiratory infections.
Collapse
Affiliation(s)
- Stephanie Duehlmeyer
- Departments of Pharmacy (SRD, ECE), Children's Mercy- Kansas City, Kansas City, MO
| | | | | |
Collapse
|
29
|
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
|
30
|
Rano S, Bhaduri A, Singh M. Nanoparticle-based platforms for targeted drug delivery to the pulmonary system as therapeutics to curb cystic fibrosis: A review. J Microbiol Methods 2024; 217-218:106876. [PMID: 38135160 DOI: 10.1016/j.mimet.2023.106876] [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: 10/17/2021] [Revised: 12/15/2023] [Accepted: 12/16/2023] [Indexed: 12/24/2023]
Abstract
Cystic fibrosis (CF) is a genetic disorder of the respiratory system caused by mutation of the Cystic Fibrosis Trans-Membrane Conductance Regulator (CFTR) gene that affects a huge number of people worldwide. It results in difficulty breathing due to a large accumulation of mucus in the respiratory tract, resulting in serious bacterial infections, and subsequent death. Traditional drug-based treatments face hindered penetration at the site of action due to the thick mucus layer. Nanotechnology offers possibilities for developing advanced and effective treatment platforms by focusing on drugs that can penetrate the dense mucus layer, fighting against the underlying bacterial infections, and targeting the genetic cause of the disease. In this review, current nanoparticle-mediated drug delivery platforms for CF, challenges in therapeutics, and future prospects have been highlighted. The effectiveness of the different types of nano-based systems conjugated with various drugs to combat the symptoms and the challenges of treating CF are brought into focus. The toxic effects of these nano-medicines and the various factors that are responsible for their effectiveness are also highlighted.
Collapse
Affiliation(s)
- Sujoy Rano
- Department of Biotechnology, Haldia Institute of Technology, HIT Campus, Purba Medinipur, Haldia 721657, West Bengal, India; In-vitro Biology, Aragen Life Sciences, Hyderabad 500076, Telangana, India
| | - Ahana Bhaduri
- Department of Biotechnology, Haldia Institute of Technology, HIT Campus, Purba Medinipur, Haldia 721657, West Bengal, India
| | - Mukesh Singh
- Department of Biotechnology, Haldia Institute of Technology, HIT Campus, Purba Medinipur, Haldia 721657, West Bengal, India; Department of Botany, Kabi Nazrul College, Murarai, Birbhum 731219 (West Bengal), India.
| |
Collapse
|
31
|
Duckworth LA, Sutton KA, Shaikh N, Wang J, Hall-Moore C, Holtz LR, Tarr PI, Rubenstein RC. Quantification of Enteric Dysfunction in Cystic Fibrosis: Inter- and Intraindividual Variability. J Pediatr 2024; 265:113800. [PMID: 37866678 PMCID: PMC10869934 DOI: 10.1016/j.jpeds.2023.113800] [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: 06/29/2023] [Revised: 09/18/2023] [Accepted: 10/17/2023] [Indexed: 10/24/2023]
Abstract
OBJECTIVES To test the utility of various biomarkers as indicators of gut dysfunction in cystic fibrosis (CF) and determine whether intraindividual variations in these measures are repeatable over short intervals and whether interindividual variations correlate with clinical outcomes. STUDY DESIGN We performed a cross-sectional, limited longitudinal study of children with CF aged 1-21 years who provided blood and stool samples at 2 or 3 visits, 2 weeks and 3 months apart, which were assayed for markers of intestinal inflammation (fecal calprotectin [fCal], lipocalin-2 [fLcn2], neopterin), and permeability (plasma lipopolysaccharide [LPS] antibodies, LPS-binding protein) by enzyme immunoassays. Control specimens were obtained from children without CF who had undergone esophagogastroduodenoscopy and had no evidence of gut inflammation. RESULTS Twenty-six of 29 participants with CF completed the study. Sixty-nine stools (57 case/12 control) and 76 plasmas (60 case/16 control) were analyzed. LPS antibody had reliable intraindividual stability. fCal, fLcn2, and neopterin were significantly greater in CF than in control samples. fCal was negatively correlated with 3-month interval change (Δ) in weight-for-age z-score, body mass index/weight-for-length z-score, and forced expiratory volume in 1 second. fLcn2 was negatively correlated with FEV1 but not with anthropometrics. No marker correlated with Δbody mass index/weight-for-length z-score or ΔFEV1. CONCLUSIONS fLcn2 is elevated in people with CF and might predict worse interval pulmonary function. Expanded studies are warranted to test if fLcn2 correlates with changes in additional outcomes.
Collapse
Affiliation(s)
- Laura A Duckworth
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Washington University in St Louis, St Louis, MO.
| | - Kimberly A Sutton
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Washington University in St Louis, St Louis, MO
| | - Nurmohammad Shaikh
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Washington University in St Louis, St Louis, MO
| | - Jinli Wang
- Center for Biostatistics and Data Science, Washington University in St Louis, St Louis, MO
| | - Carla Hall-Moore
- Center for Biostatistics and Data Science, Washington University in St Louis, St Louis, MO
| | - Lori R Holtz
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Washington University in St Louis, St Louis, MO
| | - Phillip I Tarr
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Washington University in St Louis, St Louis, MO
| | - Ronald C Rubenstein
- Division of Allergy and Pulmonary Medicine, Department of Pediatrics, Washington University in St Louis, St Louis, MO
| |
Collapse
|
32
|
Anton-Păduraru DT, Murgu AM, Bozomitu LI, Mîndru DE, Iliescu Halițchi CO, Trofin F, Ciongradi CI, Sârbu I, Eṣanu IM, Azoicăi AN. Diagnosis and Management of Gastrointestinal Manifestations in Children with Cystic Fibrosis. Diagnostics (Basel) 2024; 14:228. [PMID: 38275475 PMCID: PMC10814426 DOI: 10.3390/diagnostics14020228] [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: 12/15/2023] [Revised: 01/16/2024] [Accepted: 01/20/2024] [Indexed: 01/27/2024] Open
Abstract
Cystic fibrosis (CF) is primarily known for its pulmonary consequences, which are extensively explored in the existing literature. However, it is noteworthy that individuals with CF commonly display gastrointestinal (G-I) manifestations due to the substantial presence of the cystic fibrosis transmembrane conductance regulator (CFTR) protein in the intestinal tract. Recognized as pivotal nonpulmonary aspects of CF, G-I manifestations exhibit a diverse spectrum. Identifying and effectively managing these manifestations are crucial for sustaining health and influencing the overall quality of life for CF patients. This review aims to synthesize existing knowledge, providing a comprehensive overview of the G-I manifestations associated with CF. Each specific G-I manifestation, along with the diagnostic methodologies and therapeutic approaches, is delineated, encompassing the impact of innovative treatments targeting the fundamental effects of CF on the G-I tract. The findings underscore the imperative for prompt diagnosis and meticulous management of G-I manifestations, necessitating a multidisciplinary team approach for optimal care and enhancement of the quality of life for affected individuals. In conclusion, the authors emphasize the urgency for further clinical studies to establish a more robust evidence base for managing G-I symptoms within the context of this chronic disease. Such endeavors are deemed essential for advancing understanding and refining the clinical care of CF patients with G-I manifestations.
Collapse
Affiliation(s)
- Dana-Teodora Anton-Păduraru
- Department of Mother and Child Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iaṣi, Romania; (D.-T.A.-P.); (L.I.B.); (D.E.M.); (C.O.I.H.); (A.N.A.)
- “Sf. Maria” Children Emergency Hospital, 700309 Iasi, Romania; (C.I.C.); (I.S.)
| | - Alina Mariela Murgu
- Department of Mother and Child Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iaṣi, Romania; (D.-T.A.-P.); (L.I.B.); (D.E.M.); (C.O.I.H.); (A.N.A.)
- “Sf. Maria” Children Emergency Hospital, 700309 Iasi, Romania; (C.I.C.); (I.S.)
| | - Laura Iulia Bozomitu
- Department of Mother and Child Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iaṣi, Romania; (D.-T.A.-P.); (L.I.B.); (D.E.M.); (C.O.I.H.); (A.N.A.)
- “Sf. Maria” Children Emergency Hospital, 700309 Iasi, Romania; (C.I.C.); (I.S.)
| | - Dana Elena Mîndru
- Department of Mother and Child Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iaṣi, Romania; (D.-T.A.-P.); (L.I.B.); (D.E.M.); (C.O.I.H.); (A.N.A.)
- “Sf. Maria” Children Emergency Hospital, 700309 Iasi, Romania; (C.I.C.); (I.S.)
| | - Codruța Olimpiada Iliescu Halițchi
- Department of Mother and Child Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iaṣi, Romania; (D.-T.A.-P.); (L.I.B.); (D.E.M.); (C.O.I.H.); (A.N.A.)
| | - Felicia Trofin
- Department of Preventive Medicine and Interdisciplinarity–Microbiology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iaṣi, Romania;
| | - Carmen Iulia Ciongradi
- “Sf. Maria” Children Emergency Hospital, 700309 Iasi, Romania; (C.I.C.); (I.S.)
- 2nd Department of Surgery, Pediatric Surgery and Orthopedics, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iaṣi, Romania
| | - Ioan Sârbu
- “Sf. Maria” Children Emergency Hospital, 700309 Iasi, Romania; (C.I.C.); (I.S.)
- 2nd Department of Surgery, Pediatric Surgery and Orthopedics, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iaṣi, Romania
| | - Irina Mihaela Eṣanu
- Medical Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iaṣi, Romania;
| | - Alice Nicoleta Azoicăi
- Department of Mother and Child Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iaṣi, Romania; (D.-T.A.-P.); (L.I.B.); (D.E.M.); (C.O.I.H.); (A.N.A.)
- “Sf. Maria” Children Emergency Hospital, 700309 Iasi, Romania; (C.I.C.); (I.S.)
| |
Collapse
|
33
|
Bacalhau M, Camargo M, Lopes-Pacheco M. Laboratory Tools to Predict CFTR Modulator Therapy Effectiveness and to Monitor Disease Severity in Cystic Fibrosis. J Pers Med 2024; 14:93. [PMID: 38248793 PMCID: PMC10820563 DOI: 10.3390/jpm14010093] [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: 12/11/2023] [Revised: 12/28/2023] [Accepted: 01/11/2024] [Indexed: 01/23/2024] Open
Abstract
The implementation of cystic fibrosis (CF) transmembrane conductance regulator (CFTR) modulator drugs into clinical practice has been attaining remarkable therapeutic outcomes for CF, a life-threatening autosomal recessive genetic disease. However, there is elevated CFTR allelic heterogeneity, and various individuals carrying (ultra)rare CF genotypes remain without any approved modulator therapy. Novel translational model systems based on individuals' own cells/tissue are now available and can be used to interrogate in vitro CFTR modulator responses and establish correlations of these assessments with clinical features, aiming to provide prediction of therapeutic effectiveness. Furthermore, because CF is a progressive disease, assessment of biomarkers in routine care is fundamental in monitoring treatment effectiveness and disease severity. In the first part of this review, we aimed to focus on the utility of individual-derived in vitro models (such as bronchial/nasal epithelial cells and airway/intestinal organoids) to identify potential responders and expand personalized CF care. Thereafter, we discussed the usage of CF inflammatory biomarkers derived from blood, bronchoalveolar lavage fluid, and sputum to routinely monitor treatment effectiveness and disease progression. Finally, we summarized the progress in investigating extracellular vesicles as a robust and reliable source of biomarkers and the identification of microRNAs related to CFTR regulation and CF inflammation as novel biomarkers, which may provide valuable information for disease prognosis.
Collapse
Affiliation(s)
- Mafalda Bacalhau
- Biosystems & Integrative Sciences Institute (BioISI), Faculty of Sciences, University of Lisbon, 1749-016 Lisbon, Portugal;
| | - Mariana Camargo
- Department of Surgery, Division of Urology, Sao Paulo Federal University, Sao Paulo 04039-060, SP, Brazil
| | - Miquéias Lopes-Pacheco
- Biosystems & Integrative Sciences Institute (BioISI), Faculty of Sciences, University of Lisbon, 1749-016 Lisbon, Portugal;
| |
Collapse
|
34
|
da Silva LVRF, Athanazio RA, Tonon CR, Ferreira JC, Tanni SE. Use of elexacaftor+tezacaftor+ivacaftor in individuals with cystic fibrosis and at least one F508del allele: a systematic review and meta-analysis. J Bras Pneumol 2024; 49:e20230187. [PMID: 38198345 PMCID: PMC10760416 DOI: 10.36416/1806-3756/e20230187] [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: 05/27/2023] [Accepted: 08/11/2023] [Indexed: 01/12/2024] Open
Abstract
OBJECTIVE To evaluate the effect of treatment with the combination of three cystic fibrosis transmembrane conductance regulator (CFTR) modulators-elexacaftor+tezacaftor+ivacaftor (ETI)-on important clinical endpoints in individuals with cystic fibrosis. METHODS This was a systematic review and meta-analysis of randomized clinical trials that compared the use of ETI in individuals with CF and at least one F508del allele with that of placebo or with an active comparator such as other combinations of CFTR modulators, following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) recommendations and the Patients of interest, Intervention to be studied, Comparison of interventions, and Outcome of interest (PICO) methodology. We searched the following databases: MEDLINE, EMBASE, Cochrane Central Register of Controlled Trials, and ClinicalTrials.gov from their inception to December 26th, 2022. The risk of bias was assessed using the Cochrane risk-of-bias tool, and the quality of evidence was based on the Grading of Recommendations Assessment, Development and Evaluation (GRADE). RESULTS We retrieved 54 studies in the primary search. Of these, 6 met the inclusion criteria and were analyzed (1,127 patients; 577 and 550 in the intervention and control groups, respectively). The meta-analysis revealed that the use of ETI increased FEV1% [risk difference (RD), +10.47%; 95% CI, 6.88-14.06], reduced the number of acute pulmonary exacerbations (RD, -0.16; 95% CI, -0.28 to -0.04), and improved quality of life (RD, +14.93; 95% CI, 9.98-19.89) and BMI (RD, +1.07 kg/m2; 95% CI, 0.90-1.25). Adverse events did not differ between groups (RD, -0.03; 95% CI, -0.08 to 0.01), and none of the studies reported deaths. CONCLUSIONS Our findings demonstrate that ETI treatment substantially improves clinically significant, patient-centered outcomes.
Collapse
Affiliation(s)
- Luiz Vicente Ribeiro Ferreira da Silva
- . Unidade de Pneumologia Pediátrica, Instituto da Criança e do Adolescente, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo (SP) Brasil
| | - Rodrigo Abensur Athanazio
- . Divisão de Pneumologia, Instituto do Coração - InCor - Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo (SP) Brasil
| | | | - Juliana Carvalho Ferreira
- . Divisão de Pneumologia, Instituto do Coração - InCor - Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo (SP) Brasil
| | - Suzana Erico Tanni
- . Departamento de Clínica Médica, Faculdade de Medicina de Botucatu, Botucatu, (SP) Brasil
| |
Collapse
|
35
|
Wiesel V, Aviram M, Mei-Zahav M, Dotan M, Prais D, Cohen-Cymberknoh M, Gur M, Bar-Yoseph R, Livnat G, Goldbart A, Hazan G, Hazan I, Golan-Tripto I. Eradication of Nontuberculous Mycobacteria in People with Cystic Fibrosis Treated with Elexacaftor/Tezacaftor/Ivacaftor: A Multicenter Cohort Study. J Cyst Fibros 2024; 23:41-49. [PMID: 37173154 DOI: 10.1016/j.jcf.2023.05.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 05/01/2023] [Accepted: 05/03/2023] [Indexed: 05/15/2023]
Abstract
BACKGROUND The prevalence of nontuberculous mycobacteria (NTM) infections is rising in people with cystic fibrosis (pwCF). NTM infection, especially infection with Mycobacterium abscessus complex (MABC), is commonly associated with severe lung deterioration. The current treatment modalities, including multiple intravenous antibiotics, frequently fail to achieve airway eradication. Although treatment with elexacaftor/tezacaftor/ivacaftor (ETI) has been shown to modulate the lung microbiome, data regarding its role in eradicating NTM in pwCF is lacking. Our aim was to evaluate the impact of ETI on the rate of NTM eradication in pwCF. METHODS This retrospective multicenter cohort study included pwCF from five CF centers in Israel. PwCF aged older than 6 who had at least one positive NTM airway culture in the past two years and were treated with ETI for at least one year were included. The annual NTM and bacterial isolations, pulmonary function tests, and body mass index were analyzed before and after ETI treatment. RESULTS Fifteen pwCF were included (median age 20.9 years, 73.3% females, 80% pancreatic insufficient). In nine patients (66%) NTM isolations were eradicated following treatment with ETI. Seven of them had MABC. The median time between the first NTM isolation and treatment with ETI was 2.71 years (0.27-10.35 years). Eradication of NTM was associated with improved pulmonary function tests (p<0.05). CONCLUSIONS For the first time, we report successful eradication of NTM, including MABC, following treatment with ETI in pwCF. Additional studies are needed to assess whether treatment with ETI can result in the long-term eradication of NTM.
Collapse
Affiliation(s)
- Vered Wiesel
- Medical School for International Health, Ben Gurion University, Beer Sheva, Israel; Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Micha Aviram
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel; Pediatric Pulmonary Unit, Soroka University Medical Center, Beer Sheva, Israel
| | - Meir Mei-Zahav
- Kathy and Lee Graub Cystic Fibrosis Center, Schneider Children's Medical Center of Israel, Petach Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Miri Dotan
- Kathy and Lee Graub Cystic Fibrosis Center, Schneider Children's Medical Center of Israel, Petach Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Dario Prais
- Kathy and Lee Graub Cystic Fibrosis Center, Schneider Children's Medical Center of Israel, Petach Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Malena Cohen-Cymberknoh
- Pediatric Pulmonary Unit and Cystic Fibrosis Center, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Michal Gur
- Pediatric Pulmonary Institute and CF Center, Rappaport Children's Hospital, Rambam Health Care Campus, Haifa, Israel; Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Ronen Bar-Yoseph
- Pediatric Pulmonary Institute and CF Center, Rappaport Children's Hospital, Rambam Health Care Campus, Haifa, Israel; Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Galit Livnat
- Pediatric Pulmonology Unit and CF center, Carmel Medical Center, Haifa, Israel
| | - Aviv Goldbart
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel; Pediatric Pulmonary Unit, Soroka University Medical Center, Beer Sheva, Israel
| | - Guy Hazan
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel; Pediatric Pulmonary Unit, Soroka University Medical Center, Beer Sheva, Israel
| | - Itai Hazan
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel; Clinical Research Center, Soroka University Medical Center and Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Inbal Golan-Tripto
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel; Pediatric Pulmonary Unit, Soroka University Medical Center, Beer Sheva, Israel.
| |
Collapse
|
36
|
Donaldson SH, Corcoran TE, Pilewski JM, Mogayzel P, Laube BL, Boitet ER, Harris ES, Ceppe A, Edwards LJ, Zeman K, Wu J, Esther CR, Nichols DP, Bennett WD, Rowe SM. Effect of elexacaftor/tezacaftor/ivacaftor on mucus and mucociliary clearance in cystic fibrosis. J Cyst Fibros 2024; 23:155-160. [PMID: 37845149 PMCID: PMC10948316 DOI: 10.1016/j.jcf.2023.10.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 09/15/2023] [Accepted: 10/12/2023] [Indexed: 10/18/2023]
Abstract
BACKGROUND The cystic fibrosis transmembrane conductance regulator (CFTR) modulator elexacaftor/tezacaftor/ivacaftor (E/T/I) is highly effective clinically for those with at least one F508del-CFTR allele. The effects of E/T/I on mucociliary clearance (MCC) and sputum properties are unknown. We, therefore, sought to characterize the effects of E/T/I on in vivo MCC and sputum characteristics hypothesized to impact mucus transport. METHODS Forty-four participants ≥12 years of age were enrolled into this prospective, observational trial prior to initiation of E/T/I and had baseline measurement of MCC and characterization of induced sputum and exhaled breath condensate (EBC) samples. Study procedures were repeated after 1 month of E/T/I treatment. RESULTS Average age was 27.7 years with baseline forced expiratory volume in 1 second (FEV1) of 78.2 % predicted. 52 % of subjects had previously been treated with a 2-drug CFTR modulator combination. The average whole lung MCC rate measured over 60 min (WLAveClr60) significantly improved from baseline to post-E/T/I (14.8 vs. 22.8 %; p = 0.0002), as did other MCC indices. Sputum% solids also improved (modeled mean 3.4 vs. 2.2 %; p<0.0001), whereas non-significant reductions in sputum macrorheology (G', G") were observed. No meaningful changes in exhaled breath condensate endpoints (sialic acid:urea ratio, pH) were observed. CONCLUSIONS E/T/I improved the hydration of respiratory secretions (% solids) and markedly accelerated MCC. These data confirm the link between CFTR function, mucus solid content, and MCC and help to define the utility of MCC and mucus-related bioassays in future efforts to restore CFTR function in all people with CF.
Collapse
Affiliation(s)
| | - Timothy E Corcoran
- Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, USA
| | - Joseph M Pilewski
- Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, USA
| | - Peter Mogayzel
- Pediatric Pulmonary Medicine, Johns Hopkins University, USA
| | - Beth L Laube
- Pediatric Pulmonary Medicine, Johns Hopkins University, USA
| | - Evan R Boitet
- Department of Medicine and the Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, USA
| | - Elex S Harris
- Department of Medicine and the Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, USA
| | - Agathe Ceppe
- Department of Medicine, Univ. North Carolina at Chapel Hill, USA
| | - Lloyd J Edwards
- Department of Biostatistics, University of Alabama at Birmingham, USA
| | - Kirby Zeman
- Department of Medicine, Univ. North Carolina at Chapel Hill, USA
| | - Jihong Wu
- Department of Medicine, Univ. North Carolina at Chapel Hill, USA
| | - Charles R Esther
- Pediatric Pulmonology, Department of Pediatrics, Univ. North Carolina at Chapel Hill, USA
| | - David P Nichols
- Department of Pediatrics, University of Washington School of Medicine, USA
| | | | - Steven M Rowe
- Department of Medicine and the Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, USA
| |
Collapse
|
37
|
Miravitlles M, Criner GJ, Mall MA, Rowe SM, Vogelmeier CF, Hederer B, Schoenberger M, Altman P. Potential systemic effects of acquired CFTR dysfunction in COPD. Respir Med 2024; 221:107499. [PMID: 38104786 DOI: 10.1016/j.rmed.2023.107499] [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: 09/26/2023] [Revised: 11/25/2023] [Accepted: 12/10/2023] [Indexed: 12/19/2023]
Abstract
Chronic obstructive pulmonary disease (COPD) is characterized by airflow limitation, respiratory symptoms, inflammation of the airways, and systemic manifestations of the disease. Genetic susceptibility and environmental factors are important in the development of the disease, particularly exposure to cigarette smoke which is the most notable risk factor. Mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene are the cause of cystic fibrosis (CF), which shares several pathophysiological pulmonary features with COPD, including airway obstruction, chronic airway inflammation and bacterial colonization; in addition, both diseases also present systemic defects leading to comorbidities such as pancreatic, gastrointestinal, and bone-related diseases. In patients with COPD, systemic CFTR dysfunction can be acquired by cigarette smoking, inflammation, and infection. This dysfunction is, on average, about half of that found in CF. Herein we review the literature focusing on acquired CFTR dysfunction and the potential role in the pathogenesis of comorbidities associated with COPD and chronic bronchitis.
Collapse
Affiliation(s)
- Marc Miravitlles
- Pneumology Department Hospital Universitari Vall d'Hebron, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Campus, Barcelona, Spain.
| | - Gerard J Criner
- Department of Thoracic Medicine and Surgery, Lewis Katz School of Medicine at Temple University, Philadelphia, USA
| | - Marcus A Mall
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité-Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; Berlin Institute of Health at the Charité - Universitätsmedizin Berlin, Berlin, Germany; German Centre for Lung Research, Berlin, Germany
| | - Steven M Rowe
- Univeristy of Alabama at Birmingham, Birmingham, USA
| | - Claus F Vogelmeier
- Department of Medicine, Pulmonary and Critical Care Medicine, University Hospital Marburg UKGM, German Centre for Lung Research (DZL), Marburg, Germany
| | | | | | - Pablo Altman
- Novartis Pharmaceuticals Corporation, East Hanover, New Jersey, USA
| |
Collapse
|
38
|
Baxter C, Crary I, Coler B, Marcell L, Huebner EM, Rutz S, Adams Waldorf KM. Addressing a broken drug pipeline for preterm birth: why early preterm birth is an orphan disease. Am J Obstet Gynecol 2023; 229:647-655. [PMID: 37516401 PMCID: PMC10818026 DOI: 10.1016/j.ajog.2023.07.042] [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: 04/11/2023] [Revised: 07/19/2023] [Accepted: 07/23/2023] [Indexed: 07/31/2023]
Abstract
Preterm birth remains one of the most urgent unresolved medical problems in obstetrics, yet only 2 therapeutics for preventing preterm birth have ever been approved by the United States Food and Drug Administration, and neither remains on the market. The recent withdrawal of 17-hydroxyprogesterone caproate (17-OHPC, Makena) marks a new but familiar era for obstetrics with no Food and Drug Administration-approved pharmaceuticals to address preterm birth. The lack of pharmaceuticals reflects a broad and ineffective pipeline hindered by extensive regulatory hurdles, soaring costs of performing drug research, and concerns regarding adverse effects among a particularly vulnerable population. The pharmaceutical industry has historically limited investments in research for diseases with similarly small markets, such as cystic fibrosis, given their rarity and diminished projected financial return. The Orphan Drug Act, however, incentivizes drug development for "orphan diseases", defined as affecting <200,000 people in the United States annually. Although the total number of preterm births in the United States exceeds this threshold annually, the early subset of preterm birth (<34 weeks' gestation) would qualify, which is predominantly caused by inflammation and infection. The scientific rationale for classifying preterm birth into early and late subsets is strong given that their etiologies differ, and therapeutics that may be efficacious for one subset may not work for the other. For example, antiinflammatory therapeutics would be expected to be highly effective for early but not late preterm birth. A robust therapeutic pipeline of antiinflammatory drugs already exists, which could be used to target spontaneous early preterm birth, in combination with antibiotics shown to sterilize the amniotic cavity. New applications for therapeutics targeting spontaneous early preterm birth could categorize as orphan disease drugs, which could revitalize the preterm birth therapeutic pipeline. Herein, we describe why drugs targeting early preterm birth should qualify for orphan status, which may increase pharmaceutical interest for this vitally important obstetrical condition.
Collapse
Affiliation(s)
- Carly Baxter
- School of Medicine, University of Washington, Seattle, WA
| | - Isabelle Crary
- School of Medicine, University of Washington, Seattle, WA
| | - Brahm Coler
- Elson S. Floyd College of Medicine, Washington State University, Spokane, WA
| | - Lauren Marcell
- School of Medicine, University of Washington, Seattle, WA
| | | | - Sara Rutz
- School of Medicine, University of Washington, Seattle, WA
| | - Kristina M Adams Waldorf
- Departments of Obstetrics and Gynecology and Global Health, University of Washington, Seattle, WA.
| |
Collapse
|
39
|
Lieu N, Prentice BJ, Field P, Fitzgerald DA. Trials and tribulations of highly effective modulator therapies in cystic fibrosis. Paediatr Respir Rev 2023; 48:10-19. [PMID: 37914566 DOI: 10.1016/j.prrv.2023.09.001] [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: 09/13/2023] [Accepted: 09/13/2023] [Indexed: 11/03/2023]
Abstract
Highly effective modulator therapies (HEMTs) have revolutionised the management approach of most patients living with cystic fibrosis (CF) who have access to these therapies. Clinical trials have reported significant improvements across multiorgan systems, with patients surviving longer. However, there are accumulating case reports and observational data describing various adverse events following initiation of HEMTs including drug-to-drug interactions, drug induced liver injury, Stevens-Johnson syndrome, and neurocognitive symptoms including psychosis and depression, which have required discontinuation of therapy. Current clinical trials are assessing efficacy in younger patients with CF, yet long-term studies are also required to better understand the safety profile in the real-world setting across all ages and the impact of HEMT dose alteration or discontinuation.
Collapse
Affiliation(s)
- Nathan Lieu
- Department of Respiratory Medicine, The Children's Hospital at Westmead, Sydney, New South Wales, Australia, 2145; Discipline of Paediatrics and Child Health, University of Sydney, Sydney, New South Wales, Australia.
| | - Bernadette J Prentice
- Department of Respiratory Medicine, Sydney Children's Hospital, Randwick, Sydney, New South Wales, Australia, 2031; Discipline of Paediatrics and Child Health, UNSW, Sydney, New South Wales, Australia
| | - Penelope Field
- Department of Respiratory Medicine, The Children's Hospital at Westmead, Sydney, New South Wales, Australia, 2145; Department of Respiratory Medicine, Sydney Children's Hospital, Randwick, Sydney, New South Wales, Australia, 2031; Discipline of Paediatrics and Child Health, UNSW, Sydney, New South Wales, Australia
| | - Dominic A Fitzgerald
- Department of Respiratory Medicine, The Children's Hospital at Westmead, Sydney, New South Wales, Australia, 2145; Discipline of Paediatrics and Child Health, University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
40
|
Cholon DM, Greenwald MA, Higgs MG, Quinney NL, Boyles SE, Meinig SL, Minges JT, Chaubal A, Tarran R, Ribeiro CMP, Wolfgang MC, Gentzsch M. A Novel Co-Culture Model Reveals Enhanced CFTR Rescue in Primary Cystic Fibrosis Airway Epithelial Cultures with Persistent Pseudomonas aeruginosa Infection. Cells 2023; 12:2618. [PMID: 37998353 PMCID: PMC10670530 DOI: 10.3390/cells12222618] [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: 10/03/2023] [Revised: 10/31/2023] [Accepted: 11/06/2023] [Indexed: 11/25/2023] Open
Abstract
People with cystic fibrosis (pwCF) suffer from chronic and recurring bacterial lung infections that begin very early in life and contribute to progressive lung failure. CF is caused by mutations in the CF transmembrane conductance regulator (CFTR) gene, which encodes an ion channel important for maintaining the proper hydration of pulmonary surfaces. When CFTR function is ablated or impaired, airways develop thickened, adherent mucus that contributes to a vicious cycle of infection and inflammation. Therapeutics for pwCF, called CFTR modulators, target the CFTR defect directly, restoring airway surface hydration and mucociliary clearance. However, even with CFTR modulator therapy, bacterial infections persist. To develop a relevant model of diseased airway epithelium, we established a primary human airway epithelium culture system with persistent Pseudomonas aeruginosa infection. We used this model to examine the effects of CFTR modulators on CFTR maturation, CFTR function, and bacterial persistence. We found that the presence of P. aeruginosa increased CFTR mRNA, protein, and function. We also found that CFTR modulators caused a decrease in P. aeruginosa burden. These results demonstrate the importance of including live bacteria to accurately model the CF lung, and that understanding the effects of infection on CFTR rescue by CFTR modulators is critical to evaluating and optimizing drug therapies for all pwCF.
Collapse
Affiliation(s)
- Deborah M. Cholon
- Marsico Lung Institute and Cystic Fibrosis Research Center, School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA; (D.M.C.); (M.A.G.)
| | - Matthew A. Greenwald
- Marsico Lung Institute and Cystic Fibrosis Research Center, School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA; (D.M.C.); (M.A.G.)
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Matthew G. Higgs
- Marsico Lung Institute and Cystic Fibrosis Research Center, School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA; (D.M.C.); (M.A.G.)
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Nancy L. Quinney
- Marsico Lung Institute and Cystic Fibrosis Research Center, School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA; (D.M.C.); (M.A.G.)
| | - Susan E. Boyles
- Marsico Lung Institute and Cystic Fibrosis Research Center, School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA; (D.M.C.); (M.A.G.)
| | - Suzanne L. Meinig
- Marsico Lung Institute and Cystic Fibrosis Research Center, School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA; (D.M.C.); (M.A.G.)
- Pharmaceutical Product Development (PPD), Thermo Fisher Scientific, Morrisville, NC 27560, USA
| | - John T. Minges
- Marsico Lung Institute and Cystic Fibrosis Research Center, School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA; (D.M.C.); (M.A.G.)
| | - Ashlesha Chaubal
- Marsico Lung Institute and Cystic Fibrosis Research Center, School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA; (D.M.C.); (M.A.G.)
| | - Robert Tarran
- Marsico Lung Institute and Cystic Fibrosis Research Center, School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA; (D.M.C.); (M.A.G.)
- Division of Genetic, Department of Internal Medicine, Environmental and Inhalational Disease, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Carla M. P. Ribeiro
- Marsico Lung Institute and Cystic Fibrosis Research Center, School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA; (D.M.C.); (M.A.G.)
- Division of Pulmonary Diseases, Department of Medicine, School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- Department of Cell Biology and Physiology, School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Matthew C. Wolfgang
- Marsico Lung Institute and Cystic Fibrosis Research Center, School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA; (D.M.C.); (M.A.G.)
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Martina Gentzsch
- Marsico Lung Institute and Cystic Fibrosis Research Center, School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA; (D.M.C.); (M.A.G.)
- Department of Cell Biology and Physiology, School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- Division of Pediatric Pulmonology, Department of Pediatrics, School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
| |
Collapse
|
41
|
Wang S, Niroula S, Hoffman A, Khorrami M, Khorrami M, Yuan F, Gasser GN, Choi S, Liu B, Li J, Metersky ML, Vincent M, Crum CP, Boucher RC, Karmouty-Quintana H, Huang HJ, Sheshadri A, Dickey BF, Parekh KR, Engelhardt JF, McKeon FD, Xian W. Inflammatory Activity of Epithelial Stem Cell Variants from Cystic Fibrosis Lungs Is Not Resolved by CFTR Modulators. Am J Respir Crit Care Med 2023; 208:930-943. [PMID: 37695863 PMCID: PMC10870857 DOI: 10.1164/rccm.202305-0818oc] [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: 05/06/2023] [Accepted: 09/11/2023] [Indexed: 09/13/2023] Open
Abstract
Rationale: CFTR (cystic fibrosis transmembrane conductance regulator) modulator drugs restore function to mutant channels in patients with cystic fibrosis (CF) and lead to improvements in body mass index and lung function. Although it is anticipated that early childhood treatment with CFTR modulators will significantly delay or even prevent the onset of advanced lung disease, lung neutrophils and inflammatory cytokines remain high in patients with CF with established lung disease despite modulator therapy, underscoring the need to identify and ultimately target the sources of this inflammation in CF lungs. Objectives: To determine whether CF lungs, like chronic obstructive pulmonary disease (COPD) lungs, harbor potentially pathogenic stem cell "variants" distinct from the normal p63/Krt5 lung stem cells devoted to alveolar fates, to identify specific variants that might contribute to the inflammatory state of CF lungs, and to assess the impact of CFTR genetic complementation or CFTR modulators on the inflammatory variants identified herein. Methods: Stem cell cloning technology developed to resolve pathogenic stem cell heterogeneity in COPD and idiopathic pulmonary fibrosis lungs was applied to end-stage lungs of patients with CF (three homozygous CFTR:F508D, one CFTR F508D/L1254X; FEV1, 14-30%) undergoing therapeutic lung transplantation. Single-cell-derived clones corresponding to the six stem cell clusters resolved by single-cell RNA sequencing of these libraries were assessed by RNA sequencing and xenografting to monitor inflammation, fibrosis, and mucin secretion. The impact of CFTR activity on these variants after CFTR gene complementation or exposure to CFTR modulators was assessed by molecular and functional studies. Measurements and Main Results: End-stage CF lungs display a stem cell heterogeneity marked by five predominant variants in addition to the normal lung stem cell, of which three are proinflammatory both at the level of gene expression and their ability to drive neutrophilic inflammation in xenografts in immunodeficient mice. The proinflammatory functions of these three variants were unallayed by genetic or pharmacological restoration of CFTR activity. Conclusions: The emergence of three proinflammatory stem cell variants in CF lungs may contribute to the persistence of lung inflammation in patients with CF with advanced disease undergoing CFTR modulator therapy.
Collapse
Affiliation(s)
- Shan Wang
- Department of Biology and Biochemistry, University of Houston, Houston, Texas
| | - Suchan Niroula
- Department of Biology and Biochemistry, University of Houston, Houston, Texas
| | - Ashley Hoffman
- Department of Biology and Biochemistry, University of Houston, Houston, Texas
| | - Melika Khorrami
- Department of Biology and Biochemistry, University of Houston, Houston, Texas
| | - Melina Khorrami
- Department of Biology and Biochemistry, University of Houston, Houston, Texas
| | - Feng Yuan
- Department of Anatomy and Cell Biology
- Gene Therapy Center for Cystic Fibrosis and Other Genetic Diseases, and
| | - Grace N. Gasser
- Department of Anatomy and Cell Biology
- Gene Therapy Center for Cystic Fibrosis and Other Genetic Diseases, and
| | - Soon Choi
- Department of Anatomy and Cell Biology
- Gene Therapy Center for Cystic Fibrosis and Other Genetic Diseases, and
| | - Bovey Liu
- Department of Biology and Biochemistry, University of Houston, Houston, Texas
| | | | - Mark L. Metersky
- Center for Bronchiectasis Care, Pulmonary, Critical Care, and Sleep Medicine, University of Connecticut Health Center, Farmington, Connecticut
| | | | - Christopher P. Crum
- Women’s and Perinatal Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| | - Richard C. Boucher
- Cystic Fibrosis and Pulmonary Research and Treatment Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Harry Karmouty-Quintana
- Department of Biochemistry and Molecular Biology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas
| | - Howard J. Huang
- Department of Medicine, Houston Methodist Hospital, Houston, Texas; and
| | - Ajay Sheshadri
- Department of Pulmonary Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Burton F. Dickey
- Department of Pulmonary Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Kalpaj R. Parekh
- Department of Anatomy and Cell Biology
- Gene Therapy Center for Cystic Fibrosis and Other Genetic Diseases, and
- Division of Thoracic Surgery, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - John F. Engelhardt
- Department of Anatomy and Cell Biology
- Gene Therapy Center for Cystic Fibrosis and Other Genetic Diseases, and
| | - Frank D. McKeon
- Department of Biology and Biochemistry, University of Houston, Houston, Texas
| | - Wa Xian
- Department of Biology and Biochemistry, University of Houston, Houston, Texas
| |
Collapse
|
42
|
Greaney C, Doyle A, Drummond N, King S, Hollander-Kraaijeveld F, Robinson K, Tierney A. What do people with cystic fibrosis eat? Diet quality, macronutrient and micronutrient intakes (compared to recommended guidelines) in adults with cystic fibrosis-A systematic review. J Cyst Fibros 2023; 22:1036-1047. [PMID: 37648586 DOI: 10.1016/j.jcf.2023.08.004] [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: 04/06/2023] [Revised: 07/18/2023] [Accepted: 08/13/2023] [Indexed: 09/01/2023]
Abstract
BACKGROUND Treatment advancements have improved life expectancy and nutritional status of people with cystic fibrosis (CF). Alongside reductions in malnutrition, incidences of overweight, obesity and risk factors for diet-related chronic diseases have increased in recent years. This study aimed to synthesise the available literature on diet quality, macronutrient and micronutrient intakes compared to the recommended guidelines in adults with CF, an essential step in deducing the optimal dietary pattern and intakes for CF adults. METHODS A systematic search of five electronic databases from inception until April 2023 was conducted using keywords related to CF, diet quality and nutrient intakes. RESULTS Twenty-one studies were included comprising 18 cross-sectional, one cohort and two case control studies, reporting data from 724 adults with CF. Energy and / or macronutrient intake data was reported across 17 cohorts, eight studies provided micronutrients data, and diet quality was determined for four CF cohorts by using a diet quality score, and / or categorising food intake into servings per day for food groups and comparing findings to national dietary guidelines. Although energy intake recommendations were met, and most micronutrient requirements were achieved through supplementation, total energy intake from fat was above recommendations and diet quality was poor. CONCLUSION This is the first systematic review comprehensively evaluating literature on dietary intakes of adults with CF. Energy-dense, nutrient-poor foods contribute to intakes which pose risk in developing diet-related chronic diseases. Revision of dietary guidelines and practice change in CF nutritional therapy is warranted to optimise nutrition and health outcomes.
Collapse
Affiliation(s)
- Cian Greaney
- School of Allied Health, University of Limerick, Limerick, Ireland; Health Implementation Science and Technology Research Group, Health Research Institute, University of Limerick, Limerick, Ireland..
| | - Ailish Doyle
- School of Allied Health, University of Limerick, Limerick, Ireland
| | - Nicola Drummond
- School of Allied Health, University of Limerick, Limerick, Ireland
| | - Susannah King
- Nutrition Department, Alfred Health, Melbourne, Victoria, 2004, Australia
| | | | - Katie Robinson
- School of Allied Health, University of Limerick, Limerick, Ireland; Aging Research Centre, Health Research Institute, University of Limerick, Limerick, Ireland
| | - Audrey Tierney
- School of Allied Health, University of Limerick, Limerick, Ireland; Health Implementation Science and Technology Research Group, Health Research Institute, University of Limerick, Limerick, Ireland.; Discipline of Food, Nutrition and Dietetics, La Trobe University, Melbourne, Victoria, 3086, Australia
| |
Collapse
|
43
|
Wagner BD, Zemanick ET, Sagel SD, Robertson CE, Stevens MJ, Mayer-Hamblett N, Retsch-Bogart G, Ramsey BW, Harris JK. Limited effects of azithromycin on the oropharyngeal microbiome in children with CF and early pseudomonas infection. BMC Microbiol 2023; 23:312. [PMID: 37891457 PMCID: PMC10612347 DOI: 10.1186/s12866-023-03073-8] [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: 08/23/2023] [Accepted: 10/17/2023] [Indexed: 10/29/2023] Open
Abstract
BACKGROUND Tobramycin inhalation solution (TIS) and chronic azithromycin (AZ) have known clinical benefits for children with CF, likely due to antimicrobial and anti-inflammatory activity. The effects of chronic AZ in combination with TIS on the airway microbiome have not been extensively investigated. Oropharyngeal swab samples were collected in the OPTIMIZE multicenter, randomized, placebo-controlled trial examining the addition of AZ to TIS in 198 children with CF and early P. aeruginosa infection. Bacterial small subunit rRNA gene community profiles were determined. The effects of TIS and AZ were assessed on oropharyngeal microbial diversity and composition to uncover whether effects on the bacterial community may be a mechanism of action related to the observed changes in clinical outcomes. RESULTS Substantial changes in bacterial communities (total bacterial load, diversity and relative abundance of specific taxa) were observed by week 3 of TIS treatment for both the AZ and placebo groups. On average, these shifts were due to changes in non-traditional CF taxa that were not sustained at the later study visits (weeks 13 and 26). Bacterial community measures did not differ between the AZ and placebo groups. CONCLUSIONS This study provides further evidence that the mechanism for AZ's effect on clinical outcomes is not due solely to action on airway microbial composition.
Collapse
Affiliation(s)
- Brandie D Wagner
- Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado, Aurora, CO, USA.
- Children's Hospital Colorado, Aurora, CO, USA.
| | - Edith T Zemanick
- Children's Hospital Colorado, Aurora, CO, USA
- Department of Pediatrics, University of Colorado, Aurora, CO, USA
| | - Scott D Sagel
- Children's Hospital Colorado, Aurora, CO, USA
- Department of Pediatrics, University of Colorado, Aurora, CO, USA
| | | | - Mark J Stevens
- Children's Hospital Colorado, Aurora, CO, USA
- Department of Pediatrics, University of Colorado, Aurora, CO, USA
| | - Nicole Mayer-Hamblett
- Department of Pediatrics, University of Washington, Seattle, WA, USA
- Department of Biostatistics, University of Washington, Seattle, WA, USA
- Seattle Children's Hospital, Seattle, WA, USA
| | | | - Bonnie W Ramsey
- Department of Pediatrics, University of Washington, Seattle, WA, USA
- Seattle Children's Hospital, Seattle, WA, USA
| | - J Kirk Harris
- Children's Hospital Colorado, Aurora, CO, USA
- Department of Pediatrics, University of Colorado, Aurora, CO, USA
| |
Collapse
|
44
|
Carbone A, Vitullo P, Di Gioia S, Conese M. Lung Inflammatory Genes in Cystic Fibrosis and Their Relevance to Cystic Fibrosis Transmembrane Conductance Regulator Modulator Therapies. Genes (Basel) 2023; 14:1966. [PMID: 37895314 PMCID: PMC10606852 DOI: 10.3390/genes14101966] [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: 09/30/2023] [Revised: 10/16/2023] [Accepted: 10/17/2023] [Indexed: 10/29/2023] Open
Abstract
Cystic fibrosis (CF) is a monogenic syndrome determined by over 2000 mutations in the CF Transmembrane Conductance Regulator (CFTR) gene harbored on chromosome 7. In people with CF (PWCF), lung disease is the major determinant of morbidity and mortality and is characterized by a clinical phenotype which differs in the presence of equal mutational assets, indicating that genetic and environmental modifiers play an important role in this variability. Airway inflammation determines the pathophysiology of CF lung disease (CFLD) both at its onset and progression. In this narrative review, we aim to depict the inflammatory process in CF lung, with a particular emphasis on those genetic polymorphisms that could modify the clinical outcome of the respiratory disease in PWCF. The natural history of CF has been changed since the introduction of CFTR modulator therapies in the clinical arena. However, also in this case, there is a patient-to-patient variable response. We provide an overview on inflammatory/immunity gene variants that affect CFLD severity and an appraisal of the effects of CFTR modulator therapies on the inflammatory process in lung disease and how this knowledge may advance the optimization of the management of PWCF.
Collapse
Affiliation(s)
- Annalucia Carbone
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy; (A.C.); (S.D.G.)
| | - Pamela Vitullo
- Cystic Fibrosis Support Center, Ospedale “G. Tatarella”, 71042 Cerignola, Italy;
| | - Sante Di Gioia
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy; (A.C.); (S.D.G.)
| | - Massimo Conese
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy; (A.C.); (S.D.G.)
| |
Collapse
|
45
|
Hisert KB, Birket SE, Clancy JP, Downey DG, Engelhardt JF, Fajac I, Gray RD, Lachowicz-Scroggins ME, Mayer-Hamblett N, Thibodeau P, Tuggle KL, Wainwright CE, De Boeck K. Understanding and addressing the needs of people with cystic fibrosis in the era of CFTR modulator therapy. THE LANCET. RESPIRATORY MEDICINE 2023; 11:916-931. [PMID: 37699420 DOI: 10.1016/s2213-2600(23)00324-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 08/07/2023] [Accepted: 08/20/2023] [Indexed: 09/14/2023]
Abstract
Cystic fibrosis is a multiorgan disease caused by impaired function of the cystic fibrosis transmembrane conductance regulator (CFTR). Since the introduction of the CFTR modulator combination elexacaftor-tezacaftor-ivacaftor (ETI), which acts directly on mutant CFTR to enhance its activity, most people with cystic fibrosis (pwCF) have seen pronounced reductions in symptoms, and studies project marked increases in life expectancy for pwCF who are eligible for ETI. However, modulator therapy has not cured cystic fibrosis and the success of CFTR modulators has resulted in immediate questions about the new state of cystic fibrosis disease and clinical challenges in the care of pwCF. In this Series paper, we summarise key questions about cystic fibrosis disease in the era of modulator therapy, highlighting state-of-the-art research and clinical practices, knowledge gaps, new challenges faced by pwCF and the potential for future health-care challenges, and the pressing need for additional therapies to treat the underlying genetic or molecular causes of cystic fibrosis.
Collapse
Affiliation(s)
| | - Susan E Birket
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | - Damian G Downey
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, Northern Ireland
| | - John F Engelhardt
- Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Isabelle Fajac
- Assistance Publique-Hôpitaux de Paris, Université Paris Cité, Paris, France
| | - Robert D Gray
- Institution of Regeneration and Repair, Centre for Inflammation Research, The University of Edinburgh, Edinburgh, UK
| | | | - Nicole Mayer-Hamblett
- Department of Pediatrics, Department of Biostatistics, Seattle Children's Research Institute, University of Washington, Seattle, WA, USA
| | | | | | | | | |
Collapse
|
46
|
Taylor-Cousar JL, Robinson PD, Shteinberg M, Downey DG. CFTR modulator therapy: transforming the landscape of clinical care in cystic fibrosis. Lancet 2023; 402:1171-1184. [PMID: 37699418 DOI: 10.1016/s0140-6736(23)01609-4] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 07/17/2023] [Accepted: 07/31/2023] [Indexed: 09/14/2023]
Abstract
Following discovery of the cystic fibrosis transmembrane conductance regulator (CFTR) gene in 1989 and subsequent elucidation of the varied CFTR protein abnormalities that result, a new era of cystic fibrosis management has emerged-one in which scientific principles translated from the bench to the bedside have enabled us to potentially treat the basic defect in the majority of children and adults with cystic fibrosis, with a resultant burgeoning adult cystic fibrosis population. However, the long-term effects of these therapies on the multiple manifestations of cystic fibrosis are still under investigation. Understanding the effects of modulators in populations excluded from clinical trials is also crucial. Furthermore, establishing appropriate disease measures to assess efficacy in the youngest potential trial participants and in those whose post-modulator lung function is in the typical range for people without chronic lung disease is essential for continued drug development. Finally, recognising that a health outcome gap has been created for some people and widened for others who are not eligible for, cannot tolerate, or do not have access to modulators is important.
Collapse
Affiliation(s)
- Jennifer L Taylor-Cousar
- Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, CO, USA; Division of Pediatric Pulmonary Medicine, National Jewish Health, Denver, CO, USA; Division of Pulmonary Sciences and Critical Care Medicine, Anschutz Medical Campus, University of Colorado, Aurora, CO, USA.
| | - Paul D Robinson
- Department of Respiratory Medicine, Queensland Children's Hospital, Brisbane, QLD, Australia; Children's Health and Environment Program, Child Health Research Centre, University of Queensland, Brisbane, QLD, Australia
| | - Michal Shteinberg
- Pulmonology Institute and CF Center, Carmel Medical Center, Haifa, Israel; B Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Damian G Downey
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, UK
| |
Collapse
|
47
|
Lussac-Sorton F, Charpentier É, Imbert S, Lefranc M, Bui S, Fayon M, Berger P, Enaud R, Delhaes L. The gut-lung axis in the CFTR modulator era. Front Cell Infect Microbiol 2023; 13:1271117. [PMID: 37780857 PMCID: PMC10540301 DOI: 10.3389/fcimb.2023.1271117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 08/30/2023] [Indexed: 10/03/2023] Open
Abstract
The advent of CFTR modulators represents a turning point in the history of cystic fibrosis (CF) management, changing profoundly the disease's clinical course by improving mucosal hydration. Assessing changes in airway and digestive tract microbiomes is of great interest to better understand the mechanisms and to predict disease evolution. Bacterial and fungal dysbiosis have been well documented in patients with CF; yet the impact of CFTR modulators on microbial communities has only been partially deciphered to date. In this review, we aim to summarize the current state of knowledge regarding the impact of CFTR modulators on both pulmonary and digestive microbiomes. Our analysis also covers the inter-organ connections between lung and gut communities, in order to highlight the gut-lung axis involvement in CF pathophysiology and its evolution in the era of novel modulators therapies.
Collapse
Affiliation(s)
- Florian Lussac-Sorton
- Univ. Bordeaux, Centre de Recherche Cardio-Thoracique de Bordeaux, INSERM U1045, Pessac, France
- INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, Pessac, France
| | - Éléna Charpentier
- Univ. Bordeaux, Centre de Recherche Cardio-Thoracique de Bordeaux, INSERM U1045, Pessac, France
- INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, Pessac, France
| | - Sébastien Imbert
- Univ. Bordeaux, Centre de Recherche Cardio-Thoracique de Bordeaux, INSERM U1045, Pessac, France
- INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, Pessac, France
- CHU Bordeaux, Service de Parasitologie et Mycologie, Centre de Ressources et de Compétences de la Mucoviscidose (CRCM), Service de Pédiatrie, Service d’Exploration Fonctionnelle Respiratoire, CIC, Bordeaux, France
| | - Maxime Lefranc
- CHU Bordeaux, Service de Parasitologie et Mycologie, Centre de Ressources et de Compétences de la Mucoviscidose (CRCM), Service de Pédiatrie, Service d’Exploration Fonctionnelle Respiratoire, CIC, Bordeaux, France
| | - Stéphanie Bui
- Univ. Bordeaux, Centre de Recherche Cardio-Thoracique de Bordeaux, INSERM U1045, Pessac, France
- INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, Pessac, France
- CHU Bordeaux, Service de Parasitologie et Mycologie, Centre de Ressources et de Compétences de la Mucoviscidose (CRCM), Service de Pédiatrie, Service d’Exploration Fonctionnelle Respiratoire, CIC, Bordeaux, France
| | - Michael Fayon
- Univ. Bordeaux, Centre de Recherche Cardio-Thoracique de Bordeaux, INSERM U1045, Pessac, France
- INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, Pessac, France
- CHU Bordeaux, Service de Parasitologie et Mycologie, Centre de Ressources et de Compétences de la Mucoviscidose (CRCM), Service de Pédiatrie, Service d’Exploration Fonctionnelle Respiratoire, CIC, Bordeaux, France
| | - Patrick Berger
- Univ. Bordeaux, Centre de Recherche Cardio-Thoracique de Bordeaux, INSERM U1045, Pessac, France
- INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, Pessac, France
- CHU Bordeaux, Service de Parasitologie et Mycologie, Centre de Ressources et de Compétences de la Mucoviscidose (CRCM), Service de Pédiatrie, Service d’Exploration Fonctionnelle Respiratoire, CIC, Bordeaux, France
| | - Raphaël Enaud
- Univ. Bordeaux, Centre de Recherche Cardio-Thoracique de Bordeaux, INSERM U1045, Pessac, France
- INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, Pessac, France
- CHU Bordeaux, Service de Parasitologie et Mycologie, Centre de Ressources et de Compétences de la Mucoviscidose (CRCM), Service de Pédiatrie, Service d’Exploration Fonctionnelle Respiratoire, CIC, Bordeaux, France
| | - Laurence Delhaes
- Univ. Bordeaux, Centre de Recherche Cardio-Thoracique de Bordeaux, INSERM U1045, Pessac, France
- INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, Pessac, France
- CHU Bordeaux, Service de Parasitologie et Mycologie, Centre de Ressources et de Compétences de la Mucoviscidose (CRCM), Service de Pédiatrie, Service d’Exploration Fonctionnelle Respiratoire, CIC, Bordeaux, France
| |
Collapse
|
48
|
Mokra D, Mokry J, Barosova R, Hanusrichterova J. Advances in the Use of N-Acetylcysteine in Chronic Respiratory Diseases. Antioxidants (Basel) 2023; 12:1713. [PMID: 37760016 PMCID: PMC10526097 DOI: 10.3390/antiox12091713] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 08/23/2023] [Accepted: 08/30/2023] [Indexed: 09/29/2023] Open
Abstract
N-acetylcysteine (NAC) is widely used because of its mucolytic effects, taking part in the therapeutic protocols of cystic fibrosis. NAC is also administered as an antidote in acetaminophen (paracetamol) overdosing. Thanks to its wide antioxidative and anti-inflammatory effects, NAC may also be of benefit in other chronic inflammatory and fibrotizing respiratory diseases, such as chronic obstructive pulmonary disease, bronchial asthma, idiopathic lung fibrosis, or lung silicosis. In addition, NAC exerts low toxicity and rare adverse effects even in combination with other treatments, and it is cheap and easily accessible. This article brings a review of information on the mechanisms of inflammation and oxidative stress in selected chronic respiratory diseases and discusses the use of NAC in these disorders.
Collapse
Affiliation(s)
- Daniela Mokra
- Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, SK-03601 Martin, Slovakia; (R.B.); (J.H.)
| | - Juraj Mokry
- Department of Pharmacology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, SK-03601 Martin, Slovakia;
| | - Romana Barosova
- Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, SK-03601 Martin, Slovakia; (R.B.); (J.H.)
| | - Juliana Hanusrichterova
- Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, SK-03601 Martin, Slovakia; (R.B.); (J.H.)
| |
Collapse
|
49
|
McGarry ME, Huang CY, Ly NP. Ethnic differences in staphylococcus aureus acquisition in cystic fibrosis. J Cyst Fibros 2023; 22:909-915. [PMID: 37460380 PMCID: PMC10802839 DOI: 10.1016/j.jcf.2023.07.004] [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: 10/17/2022] [Revised: 07/06/2023] [Accepted: 07/08/2023] [Indexed: 08/31/2023]
Abstract
BACKGROUND Hispanic people with CF (pwCF) have increased morbidity than non-Hispanic White pwCF, including increased risk of Pseudomonas aeruginosa. We aimed to determine if Staphylococcus aureus (S. aureus) acquisition varies between Hispanic and non-Hispanic White pwCF. METHODS This longitudinal cohort study of pwCF ages 0-25 years in the CF Foundation Patient Registry compared acquisition of methicillin-sensitive S. aureus (MSSA), methicillin-resistant S. aureus (MRSA), persistent MRSA between Hispanic and non-Hispanic White pwCF. Risk of acquisition was assessed by Kaplan-Meier survival curves and its association with ethnicity was evaluated using Cox regressions. Adjusted associations were evaluated using multivariate Cox models adjusting for sex, age of entry into CFFPR, CFTR variant severity, pancreatic insufficiency, CF-related diabetes, maternal education, insurance status. RESULTS Of 10,640 pwCF, 7.5% were Hispanic and 92.5% were non-Hispanic White. Hispanic pwCF had a 19% higher risk of acquiring MSSA (HR 1.19, 95% CI 1.10-1.28, p<0.001) and 13% higher risk of acquiring MRSA (HR 1.13, 95% CI 1.02-1.26, p = 0.02) than non-Hispanic White pwCF. The difference in persistent MRSA between ethnicities did not reach statistical significance. After adjusting for confounding variables, only the risk of MSSA was significantly associated with ethnicity. Compared to non-Hispanic White pwCF, Hispanic pwCF acquired MSSA and MRSA at younger median ages (4.9 vs. 3.8 years (p<0.001), 22.4 vs. 20.8 years (p = 0.02). CONCLUSION Hispanic pwCF <25 years of age have an increased risk of acquiring MSSA and acquired MSSA and MRSA at an earlier age. Differences in S. aureus acquisition may contribute to increased morbidity in Hispanic pwCF.
Collapse
Affiliation(s)
- Meghan E McGarry
- Division of Pediatric Pulmonology, Department of Pediatrics, University of California, 550 16th Ave, Box 0632, San Francisco, CA 94158, United States.
| | - Chiung-Yu Huang
- Department of Epidemiology and Biostatistics, University of California, San Francisco, United States
| | - Ngoc P Ly
- Division of Pediatric Pulmonology, Department of Pediatrics, University of California, 550 16th Ave, Box 0632, San Francisco, CA 94158, United States
| |
Collapse
|
50
|
Marsh R, Dos Santos C, Hanson L, Ng C, Major G, Smyth AR, Rivett D, van der Gast C. Tezacaftor/Ivacaftor therapy has negligible effects on the cystic fibrosis gut microbiome. Microbiol Spectr 2023; 11:e0117523. [PMID: 37607068 PMCID: PMC10581179 DOI: 10.1128/spectrum.01175-23] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 06/28/2023] [Indexed: 08/24/2023] Open
Abstract
People with cystic fibrosis (pwCF) experience a range of persistent gastrointestinal symptoms throughout life. There is evidence indicating interaction between the microbiota and gut pathophysiology in CF. However, there is a paucity of knowledge on the potential effects of CF transmembrane conductance regulator (CFTR) modulator therapies on the gut microbiome. In a pilot study, we investigated the impact of Tezacaftor/Ivacaftor dual combination CFTR modulator therapy on the gut microbiota and metabolomic functioning in pwCF. Fecal samples from 12 pwCF taken at baseline and following placebo or Tezacaftor/Ivacaftor administration were subjected to microbiota sequencing and to targeted metabolomics to assess the short-chain fatty acid (SCFA) composition. Ten healthy matched controls were included as a comparison. Inflammatory calprotectin levels and patient symptoms were also investigated. No significant differences were observed in overall gut microbiota characteristics between any of the study stages, extended also across intestinal inflammation, gut symptoms, and SCFA-targeted metabolomics. However, microbiota and SCFA metabolomic compositions, in pwCF, were significantly different from controls in all study treatment stages. CFTR modulator therapy with Tezacaftor/Ivacaftor had negligible effects on both the gut microbiota and SCFA composition across the course of the study and did not alter toward compositions observed in healthy controls. Future longitudinal CFTR modulator studies will investigate more effective CFTR modulators and should use prolonged sampling periods, to determine whether longer-term changes occur in the CF gut microbiome. IMPORTANCE People with cystic fibrosis (pwCF) experience persistent gastrointestinal (GI) symptoms throughout life. The research question "how can we relieve gastrointestinal symptoms, such as stomach pain, bloating, and nausea?" remains a top priority for clinical research in CF. While CF transmembrane conductance regulator (CFTR) modulator therapies are understood to correct underlying issues of CF disease and increasing the numbers of pwCF are now receiving some form of CFTR modulator treatment. It is not known how these therapies affect the gut microbiome or GI system. In this pilot study, we investigated, for the first time, effects of the dual combination CFTR modulator medicine, Tezacaftor/Ivacaftor. We found it had negligible effects on patient GI symptoms, intestinal inflammation, or gut microbiome composition and functioning. Our findings are important as they fill important knowledge gaps on the relative effectiveness of these widely used treatments. We are now investigating triple combination CFTR modulators with prolonged sampling periods.
Collapse
Affiliation(s)
- Ryan Marsh
- Department of Applied Sciences, Northumbria University, Newcastle, United Kingdom
| | - Claudio Dos Santos
- Department of Natural Sciences, Manchester Metropolitan University, Manchester, United Kingdom
| | - Liam Hanson
- Department of Natural Sciences, Manchester Metropolitan University, Manchester, United Kingdom
- Department of Life Sciences, Manchester Metropolitan University, Manchester, United Kingdom
| | - Christabella Ng
- School of Medicine, University of Nottingham, Nottingham, United Kingdom
- NIHR Nottingham Biomedical Research Centre, Nottingham, United Kingdom
| | - Giles Major
- School of Medicine, University of Nottingham, Nottingham, United Kingdom
- Nestlé Institute of Health Sciences, Société des Produits Nestlé, Lausanne, Switzerland
| | - Alan R. Smyth
- School of Medicine, University of Nottingham, Nottingham, United Kingdom
- NIHR Nottingham Biomedical Research Centre, Nottingham, United Kingdom
| | - Damian Rivett
- Department of Natural Sciences, Manchester Metropolitan University, Manchester, United Kingdom
| | - Christopher van der Gast
- Department of Applied Sciences, Northumbria University, Newcastle, United Kingdom
- Department of Respiratory Medicine, Salford Royal NHS Foundation Trust, Salford, United Kingdom
| |
Collapse
|