1
|
Kumar D, Gupta S, Gupta V, Tanwar R, Chandel A. Engineering the Future of Regenerative Medicines in Gut Health with Stem Cell-Derived Intestinal Organoids. Stem Cell Rev Rep 2025:10.1007/s12015-025-10893-w. [PMID: 40380985 DOI: 10.1007/s12015-025-10893-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/05/2025] [Indexed: 05/19/2025]
Abstract
The advent of intestinal organoids, three-dimensional structures derived from stem cells, has significantly advanced the field of biology by providing robust in vitro models that closely mimic the architecture and functionality of the human intestine. These organoids, generated from induced pluripotent stem cells (iPSCs), embryonic stem cells (ESCs), or adult stem cells, possess remarkable capabilities for self-renewal, differentiation into diverse intestinal cell types, and functional recapitulation of physiological processes, including nutrient absorption, epithelial barrier integrity, and host-microbe interactions. The utility of intestinal organoids has been extensively demonstrated in disease modeling, drug screening, and personalized medicine. Notable examples include iPSC-derived organoids, which have been effectively employed to model enteric infections, and ESC-derived organoids, which have provided critical insights into fetal intestinal development. Patient-derived organoids have emerged as powerful tools for investigating personalized therapeutics and regenerative interventions for conditions such as inflammatory bowel disease (IBD), cystic fibrosis, and colorectal cancer. Preclinical studies involving transplantation of human intestinal organoids into murine models have shown promising outcomes, including functional integration, epithelial restoration, and immune system interactions. Despite these advancements, several challenges persist, particularly in achieving reproducibility, scalability, and maturation of organoids, which hinder their widespread clinical translation. Addressing these limitations requires the establishment of standardized protocols for organoid generation, culture, storage, and analysis to ensure reproducibility and comparability of findings across studies. Nevertheless, intestinal organoids hold immense promise for transforming our understanding of gastrointestinal pathophysiology, enhancing drug development pipelines, and advancing personalized medicine. By bridging the gap between preclinical research and clinical applications, these organoids represent a paradigm shift in the exploration of novel therapeutic strategies and the investigation of gut-associated diseases.
Collapse
Affiliation(s)
- Dinesh Kumar
- School of Pharmacy, Desh Bhagat University, Mandi Gobindgarh, Punjab, India.
| | - Sonia Gupta
- Swami Devi Dyal Group of Professional Institute, Panchkula, India
| | - Vrinda Gupta
- School of Pharmacy, Desh Bhagat University, Mandi Gobindgarh, Punjab, India
| | - Rajni Tanwar
- School of Pharmacy, Desh Bhagat University, Mandi Gobindgarh, Punjab, India
| | - Anchal Chandel
- School of Pharmacy, Desh Bhagat University, Mandi Gobindgarh, Punjab, India
| |
Collapse
|
2
|
Sugihara HY, Okamoto R, Mizutani T. Intestinal organoids: The path towards clinical application. Eur J Cell Biol 2025; 104:151474. [PMID: 39740324 DOI: 10.1016/j.ejcb.2024.151474] [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: 08/30/2024] [Revised: 11/14/2024] [Accepted: 11/17/2024] [Indexed: 01/02/2025] Open
Abstract
Organoids have revolutionized the whole field of biology with their ability to model complex three-dimensional human organs in vitro. Intestinal organoids were especially consequential as the first successful long-term culture of intestinal stem cells, which raised hopes for translational medical applications. Despite significant contributions to basic research, challenges remain to develop intestinal organoids into clinical tools for diagnosis, prognosis, and therapy. In this review, we outline the current state of translational research involving adult stem cell and pluripotent stem cell derived intestinal organoids, highlighting the advances and limitations in disease modeling, drug-screening, personalized medicine, and stem cell therapy. Preclinical studies have demonstrated a remarkable functional recapitulation of infectious and genetic diseases, and there is mounting evidence for the reliability of intestinal organoids as a patient-specific avatar. Breakthroughs now allow the generation of structurally and cellularly complex intestinal models to better capture a wider range of intestinal pathophysiology. As the field develops and evolves, there is a need for standardized frameworks for generation, culture, storage, and analysis of intestinal organoids to ensure reproducibility, comparability, and interpretability of these preclinical and clinical studies to ultimately enable clinical translation.
Collapse
Affiliation(s)
- Hady Yuki Sugihara
- Department of Gastroenterology and Hepatology, Institute of Science Tokyo, 1-5-45, Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Ryuichi Okamoto
- Department of Gastroenterology and Hepatology, Institute of Science Tokyo, 1-5-45, Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Tomohiro Mizutani
- Department of Gastroenterology and Hepatology, Institute of Science Tokyo, 1-5-45, Yushima, Bunkyo-ku, Tokyo 113-8510, Japan.
| |
Collapse
|
3
|
Ramsey ML, Gokun Y, Sobotka LA, Wellner MR, Porter K, Kirkby SE, Li SS, Papachristou GI, Krishna SG, Stanich PP, Hart PA, Conwell DL, Lara LF. Cystic Fibrosis Transmembrane Conductance Regulator Modulator Use Is Associated With Reduced Pancreatitis Hospitalizations in Patients With Cystic Fibrosis. Am J Gastroenterol 2021; 116:2446-2454. [PMID: 34665155 PMCID: PMC8900539 DOI: 10.14309/ajg.0000000000001527] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 09/09/2021] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Acute pancreatitis (AP) occurs among patients with pancreas-sufficient cystic fibrosis (PS-CF) but is reportedly less common among patients with pancreas-insufficient cystic fibrosis (PI-CF). The incidence of AP may be influenced by cystic fibrosis transmembrane conductance regulator (CFTR) modulator use. We hypothesized that CFTR modulators would reduce AP hospitalizations, with the greatest benefit in PS-CF. METHODS MarketScan (2012-2018) was queried for AP hospitalizations and CFTR modulator use among patients with CF. Multivariable Poisson models that enabled crossover between CFTR modulator treatment groups were used to analyze the rate of AP hospitalizations on and off therapy. Pancreas insufficiency was defined by the use of pancreas enzyme replacement therapy. RESULTS A total of 10,417 patients with CF were identified, including 1,795 who received a CFTR modulator. AP was more common in PS-CF than PI-CF (2.9% vs 0.9%, P = 0.007). Overall, the observed rate ratio of AP during CFTR modulator use was 0.33 (95% confidence interval [CI] 0.10, 1.11, P = 0.07) for PS-CF and 0.38 (95% CI 0.16, 0.89, P = 0.03) for PI-CF, indicating a 67% and 62% relative reduction in AP hospitalizations, respectively. In a subset analysis of 1,795 patients who all had some CFTR modulator use, the rate ratio of AP during CFTR modulator use was 0.36 (95% CI 0.13, 1.01, P = 0.05) for PS-CF and 0.53 (95% CI 0.18, 1.58, P = 0.26) for PI-CF. DISCUSSION CFTR modulator use is associated with a reduction in AP hospitalizations among patients with CF. These observational data support the prospective study of CFTR modulators to reduce AP hospitalizations among patients with CF.
Collapse
Affiliation(s)
- Mitchell L Ramsey
- Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center
| | - Yevgeniya Gokun
- Center for Biostatistics, Department of Biomedical Informatics, The Ohio State University College of Medicine
| | - Lindsay A Sobotka
- Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center
| | - Michael R Wellner
- Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center
| | - Kyle Porter
- Center for Biostatistics, Department of Biomedical Informatics, The Ohio State University College of Medicine
| | - Stephen E Kirkby
- Division of Pulmonary and Critical Care Medicine, The Ohio State University Wexner Medical Center
| | - Susan S Li
- Division of General Internal Medicine, The Ohio State University Wexner Medical Center
| | - Georgios I Papachristou
- Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center
| | - Somashekar G Krishna
- Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center
| | - Peter P Stanich
- Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center
| | - Phil A Hart
- Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center
| | - Darwin L Conwell
- Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center
| | - Luis F Lara
- Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center
| |
Collapse
|
4
|
Obafemi-Ajayi T, Perkins A, Nanduri B, Wunsch II DC, Foster JA, Peckham J. No-boundary thinking: a viable solution to ethical data-driven AI in precision medicine. AI AND ETHICS 2021; 2:635-643. [PMID: 34870283 PMCID: PMC8628283 DOI: 10.1007/s43681-021-00118-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 11/02/2021] [Indexed: 10/28/2022]
Abstract
Today Artificial Intelligence (AI) supports difficult decisions about policy, health, and our personal lives. The AI algorithms we develop and deploy to make sense of information, are informed by data, and based on models that capture and use pertinent details of the population or phenomenon being analyzed. For any application area, more importantly in precision medicine which directly impacts human lives, the data upon which algorithms are run must be procured, cleaned, and organized well to assure reliable and interpretable results, and to assure that they do not perpetrate or amplify human prejudices. This must be done without violating basic assumptions of the algorithms in use. Algorithmic results need to be clearly communicated to stakeholders and domain experts to enable sound conclusions. Our position is that AI holds great promise for supporting precision medicine, but we need to move forward with great care, with consideration for possible ethical implications. We make the case that a no-boundary or convergent approach is essential to support sound and ethical decisions. No-boundary thinking supports problem definition and solving with teams of experts possessing diverse perspectives. When dealing with AI and the data needed to use AI, there is a spectrum of activities that needs the attention of a no-boundary team. This is necessary if we are to draw viable conclusions and develop actions and policies based on the AI, the data, and the scientific foundations of the domain in question.
Collapse
Affiliation(s)
| | - Andy Perkins
- Department of Computer Science and Engineering, Mississippi State University, Starkville, MS USA
| | - Bindu Nanduri
- Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, Starkville, MS USA
| | - Donald C. Wunsch II
- Electrical & Computer Engineering Department, Missouri University of Science and Technology, Rolla, MO USA
| | - James A. Foster
- Biological Sciences Department, University of Idaho, Moscow, ID USA
| | - Joan Peckham
- Computer Science & Statistics Department, University of Rhode Island, Kingston, RI USA
| |
Collapse
|
5
|
Potential of helper-dependent Adenoviral vectors in CRISPR-cas9-mediated lung gene therapy. Cell Biosci 2021; 11:145. [PMID: 34301308 PMCID: PMC8305863 DOI: 10.1186/s13578-021-00662-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 07/19/2021] [Indexed: 12/20/2022] Open
Abstract
Since CRISPR/Cas9 was harnessed to edit DNA, the field of gene therapy has witnessed great advances in gene editing. New avenues were created for the treatment of diseases such as Cystic Fibrosis (CF). CF is caused by mutations in the Cystic Fibrosis Transmembrane Conductance Regulator (CFTR) gene. Despite the success of gene editing with the CRISPR/Cas9 in vitro, challenges still exist when using CRISPR/Cas9 in vivo to cure CF lung disease. The delivery of CRISPR/Cas9 into lungs, as well as the difficulty to achieve the efficiency required for clinical efficacy, has brought forth new challenges. Viral and non-viral vectors have been shown to deliver DNA successfully in vivo, but the sustained expression of CFTR was not adequate. Before the introduction of Helper-Dependent Adenoviral vectors (HD-Ad), clinical trials of treating pulmonary genetic diseases with first-generation viral vectors have shown limited efficacy. With the advantages of larger capacity and lower immunogenicity of HD-Ad, together with the versatility of the CRISPR/Cas9 system, delivering CRISPR/Cas9 to the airway with HD-Ad for lung gene therapy shows great potential. In this review, we discuss the status of the application of CRISPR/Cas9 in CF gene therapy, the existing challenges in the field, as well as new hurdles introduced by the presence of CRISPR/Cas9 in the lungs. Through the analysis of these challenges, we present the potential of CRISPR/Cas9-mediated lung gene therapy using HD-Ad vectors with Cystic Fibrosis lung disease as a model of therapy.
Collapse
|
6
|
Prasad R. New Perspectives in Underlying Molecular Defects Based Cystic Fibrosis Therapeutics. Indian J Clin Biochem 2021; 36:255-256. [PMID: 34220000 DOI: 10.1007/s12291-021-00988-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Rajendra Prasad
- Department of Biochemistry, MM Institute of Medical Science and Research, MM (Deemed To Be University), Mullana, Ambala, Haryana 133207 India
| |
Collapse
|
7
|
F1099L-CFTR (c.3297C>G) has Impaired Channel Function and Associates with Mild Disease Phenotypes in Two Pediatric Patients. Life (Basel) 2021; 11:life11020131. [PMID: 33567498 PMCID: PMC7915716 DOI: 10.3390/life11020131] [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: 12/24/2020] [Revised: 02/03/2021] [Accepted: 02/05/2021] [Indexed: 11/18/2022] Open
Abstract
(1) Background: many rare cystic fibrosistransmembrane conductance regulator (CFTR) mutations remain poorly characterized with regard to functional consequences of the mutation. We present the clinical features of two pediatric cystic fibrosis (CF) subjects who are heterozygous for F1099L (c.3297C>G), one with G551D (a class III mutation) and one with 3849 + 10kbC->T (a class V mutation). We also identified the molecular defect(s) that are associated with F1099L mutation to correlate with the clinical features that we observed; (2) Methods: clinical findings and history were extracted from the electronic medical record and de-identified. F1099L-CFTR protein expression level and maturation status, channel function, and the effects of CFTR modulation on these characteristics were investigated using western blotting and iodide efflux assay; (3) Results: these two subjects have mild CF phenotypes when F1099L is combined with two known disease-causing mutations. F1099L-CFTR has a moderate defect in processing and maturation, causing fewer CFTR channels at the cell surface and, therefore, impaired channel activities. These defects could be effectively corrected using VX-809 (lumacaftor); and, (4) Conclusions: our biochemical data correlate with the disease manifestations and suggest that F1099L is potentially a CF-causing mutation. The study expands our knowledge of rare CFTR mutations and may help in developing effective therapies for subjects with F1099L mutation.
Collapse
|
8
|
Next-Generation Sequencing for Molecular Diagnosis of Cystic Fibrosis in a Brazilian Cohort. DISEASE MARKERS 2021; 2021:9812074. [PMID: 33613790 PMCID: PMC7878085 DOI: 10.1155/2021/9812074] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 12/23/2020] [Accepted: 01/27/2021] [Indexed: 12/05/2022]
Abstract
Cystic fibrosis (CF), an autosomal recessive genetic disease, is recognized as one of the most prevalent diseases in Caucasian populations. Epidemiological data show that the incidence of CF varies between countries and ethnic groups in the same region. CF occurs due to pathogenic variants in the gene encoding cystic fibrosis transmembrane conductance regulator (CFTR), located on chromosome 7q31.2. To date, more than 2,000 variants have been registered in the CFTR database. The study of these variants leads to the diagnosis and the possibility of a specific treatment for each patient through precision medicine. In this study, complete screening of CFTR was performed through next-generation sequencing (NGS) to gain insight into the variants circulating in the population of Rio de Janeiro and to provide patient access to treatment through genotype-specific therapies. Samples from 93 patients with an inconclusive molecular diagnosis were subjected to full-length screening of CFTR using an Illumina NGS HiSeq platform. Among these patients, 46 had two pathogenic variants, whereas 12 had only one CFTR variant. Twenty-four variants were not part of our routine screening. Of these 24 variants, V938Gfs∗37 had not been described in the CF databases previously. This research achieved a molecular diagnosis of the patients with CF and identification of possible molecular candidates for genotype-specific treatments.
Collapse
|
9
|
Johnson B, Lee S, Ezmigna D. Palliative care and advances in cystic fibrosis: where now? BMJ Support Palliat Care 2020; 11:122-123. [PMID: 32718954 DOI: 10.1136/bmjspcare-2020-002292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 06/29/2020] [Indexed: 11/04/2022]
Affiliation(s)
- Brandy Johnson
- Department of Pediatrics, University of Florida, Gainesville, Florida, USA
| | - Seon Lee
- Department of Pediatrics, University of Florida, Gainesville, Florida, USA
| | - Dima Ezmigna
- Department of Pediatrics, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
10
|
Houston CJ, Taggart CC, Downey DG. The role of inflammation in cystic fibrosis pulmonary exacerbations. Expert Rev Respir Med 2020; 14:889-903. [PMID: 32544353 DOI: 10.1080/17476348.2020.1778469] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
INTRODUCTION Cystic Fibrosis pulmonary exacerbations are critical events in the lives of people with CF that have deleterious effects on lung function, quality of life, and life expectancy. There are significant unmet needs in the management of exacerbations. We review here the associated inflammatory changes that underlie these events and are of interest for the development of biomarkers of exacerbation. AREAS COVERED Inflammatory responses in CF are abnormal and contribute to a sustained proinflammatory lung microenvironment, abundant in proinflammatory mediators and deficient in counter-regulatory mediators that terminate and resolve inflammation. There is increasing interest in these inflammatory pathways to discover novel biomarkers for pulmonary exacerbation management. In this review, we explore the inflammatory changes occurring during intravenous antibiotic therapy for exacerbation and how they may be applied as biomarkers to guide exacerbation therapy. A literature search was conducted using the PubMed database in February 2020. EXPERT OPINION Heterogeneity in inflammatory responses to treatment of a pulmonary exacerbation, a disease process with complex pathophysiology, limits the clinical utility of individual biomarkers. Biomarker panels may be a more successful strategy to capture informative changes within the CF population to improve pulmonary exacerbation management and outcomes.
Collapse
Affiliation(s)
- Claire J Houston
- Airway Innate Immunity Group (Aiir), Wellcome Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast , Northern Ireland
| | - Clifford C Taggart
- Airway Innate Immunity Group (Aiir), Wellcome Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast , Northern Ireland
| | - Damian G Downey
- Wellcome Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast , Northern Ireland.,Northern Ireland Regional Adult CF Centre, Belfast Health and Social Care Trust , Belfast, UK
| |
Collapse
|
11
|
Johnson BJ, Choby GW, O'Brien EK. Chronic rhinosinusitis in patients with cystic fibrosis-Current management and new treatments. Laryngoscope Investig Otolaryngol 2020; 5:368-374. [PMID: 32596478 PMCID: PMC7314487 DOI: 10.1002/lio2.401] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 04/27/2020] [Accepted: 05/02/2020] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVES The purpose of this article is to provide a state-of-the-art review of the literature and summarize the latest publications on medical and surgical treatment of cystic fibrosis (CF) chronic rhinosinusitis (CRS), with an emphasis on describing recent advancements in cystic fibrosis transmembrane conductance regulator (CFTR) modulator therapies. METHODS A comprehensive literature review was conducted utilizing the PubMed database with search phrases detailed within the body of the article. Abstracts were reviewed to include publications detailing medical, surgical, and CFTR modulating therapies for CF CRS. Findings from studies not previously reviewed and publications regarding CFTR modulators were emphasized. RESULTS No clear guidelines for treatment of CF CRS are available. Nasal saline rinses, topical steroids, topical antibiotics, and topical dornase alfa are reported medical therapies. Ivacaftor has some sinonasal symptomatic benefit. A new triple therapy CFTR modulator, elexacaftor/tezacaftor/ivacaftor, is capable of treating 90% of patients with CF and has shown the greatest FEV1 improvement of any CFTR modulator yet. There is no clear consensus on surgical indications or technique, though aggressive surgery in recalcitrant disease has shown some symptomatic benefit. Endoscopic sinus surgery after lung transplantation may benefit some patients in whom the sinuses serve as a reservoir for recurrent pulmonary infections by decreasing rates of bronchiolitis obliterans syndrome and improving 5 year survival. CONCLUSION As lifespan increases for patients with CF, further investigation into medical therapy, CFTR modulator sinonasal outcomes, and surgical technique and outcomes for CF CRS is needed. LEVEL OF EVIDENCE 5.
Collapse
Affiliation(s)
| | - Garret W. Choby
- Department of OtorhinolaryngologyMayo ClinicRochesterMinnesotaUSA
| | - Erin K. O'Brien
- Department of OtorhinolaryngologyMayo ClinicRochesterMinnesotaUSA
| |
Collapse
|
12
|
Guerra L, Favia M, Di Gioia S, Laselva O, Bisogno A, Casavola V, Colombo C, Conese M. The preclinical discovery and development of the combination of ivacaftor + tezacaftor used to treat cystic fibrosis. Expert Opin Drug Discov 2020; 15:873-891. [PMID: 32290721 DOI: 10.1080/17460441.2020.1750592] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
INTRODUCTION Cystic Fibrosis (CF) is caused by mutations in the Cystic Fibrosis Transmembrane Conductance Regulator (CFTR) gene. The most common mutation, F508del, induces protein misprocessing and loss of CFTR function. The discovery through in vitro studies of the CFTR correctors (i.e. lumacaftor, tezacaftor) that partially rescue the misprocessing of F508del-CFTR with the potentiator ivacaftor is promising in giving an unprecedented clinical benefit in affected patients. AREAS COVERED Online databases were searched using key phrases for CF and CFTR modulators. Tezacaftor-ivacaftor treatment has proved to be safer than lumacaftor-ivacaftor, although clinical efficacy is similar. Further clinical efficacy has ensued with the introduction of triple therapy, i.e. applying second-generation correctors, such as VX-569 and VX-445 (elexacaftor) to tezacaftor-ivacaftor. The triple combinations will herald the availability of etiologic therapies for patients for whom no CFTR modulators are currently applied (i.e. F508del/minimal function mutations) and enhance CFTR modulator therapy for patients homozygous for F508del. EXPERT OPINION CF patient-derived tissue models are being explored to determine donor-specific response to current approved and future novel CFTR modulators for F508del and other rare mutations. The discovery and validation of biomarkers of CFTR modulation will complement these studies in the long term and in real-life world.
Collapse
Affiliation(s)
- Lorenzo Guerra
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari , Bari, Italy
| | - Maria Favia
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari , Bari, Italy
| | - Sante Di Gioia
- Department of Medical and Surgical Sciences, University of Foggia , Foggia, Italy
| | - Onofrio Laselva
- Programme in Molecular Medicine, Research Institute, Hospital for Sick Children , Toronto, Ontario, Canada.,Department of Physiology, University of Toronto , Toronto, Ontario, Canada
| | - Arianna Bisogno
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Centro Regionale di Riferimento per la Fibrosi Cistica, Università degli Studi di Milano , Milan, Italy
| | - Valeria Casavola
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari , Bari, Italy
| | - Carla Colombo
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Centro Regionale di Riferimento per la Fibrosi Cistica, Università degli Studi di Milano , Milan, Italy
| | - Massimo Conese
- Department of Medical and Surgical Sciences, University of Foggia , Foggia, Italy
| |
Collapse
|
13
|
Zeggini E, Gloyn AL, Barton AC, Wain LV. Translational genomics and precision medicine: Moving from the lab to the clinic. Science 2019; 365:1409-1413. [PMID: 31604268 DOI: 10.1126/science.aax4588] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Translational genomics aims to improve human health by building on discoveries made through genetics research and applying them in the clinical setting. This progress has been made possible by technological advances in genomics and analytics and by the digital revolution. Such advances should enable the development of prognostic markers, tailored interventions, and the design of prophylactic preventive approaches. We are at the cusp of predicting disease risk for some disorders by means of polygenic risk scores integrated with classical epidemiological risk factors. This should lead to better risk stratification and clinical decision-making. A deeper understanding of the link between genome-wide sequence and association with well-characterized phenotypes will empower the development of biomarkers to aid diagnosis, inform disease progression trajectories, and allow better targeting of treatments to those patients most likely to respond.
Collapse
Affiliation(s)
- Eleftheria Zeggini
- Institute of Translational Genomics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany.
| | - Anna L Gloyn
- Oxford Centre for Diabetes Endocrinology and Metabolism, Oxford University, Oxford, UK.,Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK.,Oxford NIHR Biomedical Research Centre, Churchill Hospital, Oxford, UK
| | - Anne C Barton
- Versus Arthritis Centre for Genetics and Genomics, Centre for Musculoskeletal Research, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK.,NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester, UK
| | - Louise V Wain
- Department of Health Sciences, University of Leicester, Leicester, UK.,National Institute for Health Research, Leicester Respiratory Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| |
Collapse
|
14
|
Lommatzsch ST, Taylor-Cousar JL. The combination of tezacaftor and ivacaftor in the treatment of patients with cystic fibrosis: clinical evidence and future prospects in cystic fibrosis therapy. Ther Adv Respir Dis 2019; 13:1753466619844424. [PMID: 31027466 PMCID: PMC6487765 DOI: 10.1177/1753466619844424] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 12/19/2018] [Indexed: 12/26/2022] Open
Abstract
Years of tremendous study have dawned a new era for the treatment of cystic fibrosis (CF). For years CF care was rooted in the management of organ dysfunction resulting from the mal-effects of absent anion transport through the CF transmembrane regulator (CFTR) protein. CFTR, an adenosine triphosphate binding anion channel, has multiple functions, but primarily regulates the movement of chloride anions, thiocyanate and bicarbonate across luminal cell membranes. Additional roles include effects on other electrolyte channels such as the epithelial sodium channel (ENaC) and on pulmonary innate immunity. Inappropriate luminal anion movement leads to elevated sweat chloride concentrations, dehydrated airway surface liquid, overall viscous mucous production, and inspissated bile and pancreatic secretions. As a result, patients develop the well-known CF symptoms and disease-defining complications such as chronic cough, oily stools, recurrent pulmonary infections, bronchiectasis, chronic sinusitis and malnutrition. Traditionally, CF has been symptomatically managed, but over the past 6 years those with CF have been offered a new mode of therapy; CFTR protein modulation. These medications affect the basic defect in CF: abnormal CFTR function. Ivacaftor, approved for use in the United States in 2012, is the first medication in CF history to improve CFTR function at the molecular level. Its study and approval were followed by two additional CFTR modulators, lumacaftor/ivacaftor and tezacaftor/ivacaftor. To effectively use currently available CF therapies, clinicians should be familiar with the side effects of the drugs and their impacts on patient outcomes. As many new modulators are on the horizon, this information will equip providers to discuss the benefits and shortcomings of modulator therapy especially in the context of limited healthcare resources.
Collapse
|
15
|
Hamilton CM, Hung M, Chen G, Qureshi Z, Thompson JR, Sun B, Bear CE, Young RN. Synthesis and characterization of a photoaffinity labelling probe based on the structure of the cystic fibrosis drug ivacaftor. Tetrahedron 2018. [DOI: 10.1016/j.tet.2018.06.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
16
|
Santos-Carballal B, Fernández Fernández E, Goycoolea FM. Chitosan in Non-Viral Gene Delivery: Role of Structure, Characterization Methods, and Insights in Cancer and Rare Diseases Therapies. Polymers (Basel) 2018; 10:E444. [PMID: 30966479 PMCID: PMC6415274 DOI: 10.3390/polym10040444] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 04/04/2018] [Accepted: 04/11/2018] [Indexed: 12/23/2022] Open
Abstract
Non-viral gene delivery vectors have lagged far behind viral ones in the current pipeline of clinical trials of gene therapy nanomedicines. Even when non-viral nanovectors pose less safety risks than do viruses, their efficacy is much lower. Since the early studies to deliver pDNA, chitosan has been regarded as a highly attractive biopolymer to deliver nucleic acids intracellularly and induce a transgenic response resulting in either upregulation of protein expression (for pDNA, mRNA) or its downregulation (for siRNA or microRNA). This is explained as the consequence of a multi-step process involving condensation of nucleic acids, protection against degradation, stabilization in physiological conditions, cellular internalization, release from the endolysosome ("proton sponge" effect), unpacking and enabling the trafficking of pDNA to the nucleus or the siRNA to the RNA interference silencing complex (RISC). Given the multiple steps and complexity involved in the gene transfection process, there is a dearth of understanding of the role of chitosan's structural features (Mw and degree of acetylation, DA%) on each step that dictates the net transfection efficiency and its kinetics. The use of fully characterized chitosan samples along with the utilization of complementary biophysical and biological techniques is key to bridging this gap of knowledge and identifying the optimal chitosans for delivering a specific gene. Other aspects such as cell type and administration route are also at play. At the same time, the role of chitosan structural features on the morphology, size and surface composition of synthetic virus-like particles has barely been addressed. The ongoing revolution brought about by the recent discovery of CRISPR-Cas9 technology will undoubtedly be a game changer in this field in the short term. In the field of rare diseases, gene therapy is perhaps where the greatest potential lies and we anticipate that chitosans will be key players in the translation of research to the clinic.
Collapse
Affiliation(s)
| | - Elena Fernández Fernández
- Lung Biology Group, Department Clinical Microbiology, RCSI, Education and Research Centre, Beaumont Hospital, Dublin 9, Ireland.
| | | |
Collapse
|
17
|
Eyns H, De Wachter E, Malfroot A, Vaes P. Acute Pain Perception During Different Sampling Methods for Respiratory Culture in Cystic Fibrosis Patients. J Pain Symptom Manage 2018; 55:872-880. [PMID: 29154891 DOI: 10.1016/j.jpainsymman.2017.11.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 11/01/2017] [Accepted: 11/05/2017] [Indexed: 11/18/2022]
Abstract
CONTEXT Reliable identification of lower respiratory tract pathogens is crucial in the management of cystic fibrosis (CF). The multitude of treatments and clinical procedures are a considerable burden and are potentially provoking pain. OBJECTIVES As part of another study (NCT02363764), investigating the bacterial yield of three sampling methods, nasal swabs (NSs), cough swabs (CSs), and (induced) sputum samples ([I]SSs), in both expectorating patients (EPs) and non-expectorating patients (NEPs) with CF, the present study aimed to explore the prevalence of respiratory culture sampling-related pain as assessed by self-report within a cohort of children and adults. METHODS Literate patients with CF (aged six years or older) completed a questionnaire on pain perception related to the three aforementioned sampling methods (No/Yes; visual analogue scale for pain [VAS-Pain] [0-10 cm]). In addition, patients were asked to rank these methods by their own preference without taking into account the presumed bacterial yield. RESULTS In total, 119 questionnaires were returned. In the EPs-group, CS was most frequently (n%; mean VAS-Pain if pain [range]) reported as painful method: overall (n = 101; 12.9%; 1.8 [0.2-4.8]), children (n = 41; 22.0%; 1.4 [0.2-2.7]), and adults (n = 60; 6.7%; 2.5 [0.5-4.8]). Highest pain intensity scores were observed with NS overall (3.0%; 2.4 [0.3-6.2]) and in children (4.9%; 3.3 [0.3-6.2]), but not in adults (1.7%; 0.6 [-]).NEPs-children (n = 17) reported ISS most frequently and as most painful sampling method (17.6%; 2.0 [1.0-4.0]). The only NEP-adult did not perceive pain. NEPs preferred NS > CS > ISS (61.1%, 33.3%, 5.6%, respectively [P = 0.001]) as primary sampling method, whereas EPs preferred SS > NS > CS (65.7%, 26.3%, 8.1%, respectively [P < 0.0001]). Patients' preference for a specific method inversely correlated to pain perception and intensity in EPs (φ = -0.155 [P = 0.007] and ρ = -0.926 [P = 0.008], respectively), but not in NEPs (φ = -0.226 [P = 0.097] and ρ = -0.135 [P = 0.798], respectively). CONCLUSION A relatively large range of pain experiences was observed in patients with CF during respiratory culture sampling, which underlines the importance of individual pain assessment. Nevertheless, clinicians can confidently choose the sampling method based on validity over patients' preference.
Collapse
Affiliation(s)
- Hanneke Eyns
- Department of Paediatrics, Paediatric Pulmonology and Paediatric Infectious Diseases, Cystic Fibrosis Centre, Universitair Ziekenhuis Brussel (UZ Brussel), Vrije Universiteit Brussel (VUB), Brussels; Department of Physical Medicine and Physiotherapy, Universitair Ziekenhuis Brussel (UZ Brussel), Vrije Universiteit Brussel (VUB), Brussels; Department of Human Physiology, Faculty of Physical Education and Physiotherapy, Vrije Universiteit Brussel (VUB), Brussels, Belgium.
| | - Elke De Wachter
- Department of Paediatrics, Paediatric Pulmonology and Paediatric Infectious Diseases, Cystic Fibrosis Centre, Universitair Ziekenhuis Brussel (UZ Brussel), Vrije Universiteit Brussel (VUB), Brussels
| | - Anne Malfroot
- Department of Paediatrics, Paediatric Pulmonology and Paediatric Infectious Diseases, Cystic Fibrosis Centre, Universitair Ziekenhuis Brussel (UZ Brussel), Vrije Universiteit Brussel (VUB), Brussels
| | - Peter Vaes
- Department of Physical Medicine and Physiotherapy, Universitair Ziekenhuis Brussel (UZ Brussel), Vrije Universiteit Brussel (VUB), Brussels; Department of Human Physiology, Faculty of Physical Education and Physiotherapy, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| |
Collapse
|
18
|
Eyns H, Piérard D, De Wachter E, Eeckhout L, Vaes P, Malfroot A. Respiratory Bacterial Culture Sampling in Expectorating and Non-expectorating Patients With Cystic Fibrosis. Front Pediatr 2018; 6:403. [PMID: 30619797 PMCID: PMC6305441 DOI: 10.3389/fped.2018.00403] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 12/04/2018] [Indexed: 01/07/2023] Open
Abstract
Purpose: Different respiratory sampling methods exist to identify lower airway pathogens in patients with cystic fibrosis (CF), of which bronchoalveolar lavage (BAL), and expectorated sputum are considered the "gold standard." Because BAL cannot be repeated limitless, the diagnosis of lower respiratory tract infections in non-expectorating patients is challenging. Other sampling techniques are nasal swab, cough swab, and induced sputum. The purpose of this study (NCT02363764) was to compare concordance between the microbiological yield of nasal swab, cough swab, and expectorated sputum in expectorating patients; nasal swab, cough swab, and induced sputum in non-expectorating patients; nasal swab, cough swab, induced sputum, and BAL in patients requiring bronchoscopy ("BAL-group"); and to determine the clinical value of cough swab in non-expectorating patients with CF. Methods: Microbiological yield detected by these different sampling techniques was compared between and within 105 expectorating patients, 30 non-expectorating patients and BAL-group (n = 39) in a single CF clinic. Specificity, sensitivity, positive (PPV), and negative (NPV) predictive values were calculated. Results: Overall low sensitivity (6.3-58.0%) and wide-ranging predictive values (0.0-100.0%) indicated that nasal swab was not appropriate to detect lower airway pathogens [Pseudomonas aeruginosa (Pa), Staphylococcus aureus (Sa), and Haemophilus influenzae (Hi)] in all three patient groups. Microbiological yield, specificity, sensitivity, PPV, and NPV of cough swab and induced sputum were largely similar in non-expectorating patients and in BAL-group (except sensitivity (0.0%) of induced sputum for Hi in BAL-group). Calculations for Pa and Hi could not be performed for non-expectorating patients because of low prevalence (n = 2 and n = 3, respectively). In expectorating patients, concordance was found between cough swab and expectorated sputum, except for Hi (sensitivity of 40.0%). Conclusion: Our findings suggest that cough swab might be helpful in detecting the presence of some typical CF pathogens in the lower airways of clinically stable patients with CF. However, in symptomatic patients, who are unable to expectorate and who have a negative cough swab and induced sample, BAL should be performed as it currently remains the "gold standard."
Collapse
Affiliation(s)
- Hanneke Eyns
- Department of Pediatrics, Pediatric Pulmonology and Pediatric Infectious Diseases, Cystic Fibrosis Clinic, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium.,Department of Physical Medicine and Physiotherapy, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium.,Department of Physiotherapy, Human Physiology and Anatomy, Faculty of Physical Education and Physiotherapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Denis Piérard
- Department of Microbiology and Infection Control, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Elke De Wachter
- Department of Pediatrics, Pediatric Pulmonology and Pediatric Infectious Diseases, Cystic Fibrosis Clinic, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Leo Eeckhout
- Department of Microbiology and Infection Control, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Peter Vaes
- Department of Physical Medicine and Physiotherapy, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium.,Department of Physiotherapy, Human Physiology and Anatomy, Faculty of Physical Education and Physiotherapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Anne Malfroot
- Department of Pediatrics, Pediatric Pulmonology and Pediatric Infectious Diseases, Cystic Fibrosis Clinic, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
19
|
Grulich-Henn J, Klose D. Understanding childhood diabetes mellitus: new pathophysiological aspects. J Inherit Metab Dis 2018; 41:19-27. [PMID: 29247329 DOI: 10.1007/s10545-017-0120-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 11/28/2017] [Indexed: 12/16/2022]
Abstract
Diabetes mellitus (DM) is not a single disease, but several pathophysiological conditions where synthesis, release, and/or action of insulin are disturbed. A progressive autoimmune/autoinflammatory destruction of islet cells is still considered the main pathophysiological event in the development of T1DM, but there is evidence that T1DM itself is a heterogeneous disease. More than 50 gene regions are closely associated with T1DM and a variety of epigenetic factors and metabolic patterns have been characterized, which may play a role in the development of T1DM. The pathogenesis and genetics of type 2 DM (T2DM) are distinct from T1DM. Genes associated with T2DM are distinct from those in T1DM. Characteristic metabolic patterns, different from those in T1DM were reported in T2DM, and some children with T2DM also express islet-antibodies. Huge progress has been made in the characterization of other specific types of DM, which had been considered very rare before. The molecular clarification of maturity-onset diabetes of the young (MODY) has greatly improved our understanding of the pathophysiology of DM. There are genetic overlaps between T2DM and monogenetic DM. Neonatal DM has been shown to be monogenetic in most cases, and genetic elucidation leads to more precise and individualized therapies. Cystic fibrosis related DM (CFRDM) should be considered a genuine part of cystic fibrosis, and not a complication, since pancreatic fibrosis does not sufficiently explain the pathophysiology of CFRDM. Disturbances of cystic fibrosis transmembrane conductance regulator (CFTR) as well as autoimmunity are involved in the pathogenesis of CFRDM.
Collapse
MESH Headings
- Adolescent
- Age of Onset
- Autoantibodies/immunology
- Autoimmunity
- Blood Glucose/metabolism
- Child
- Child, Preschool
- Cystic Fibrosis/epidemiology
- Cystic Fibrosis/genetics
- Cystic Fibrosis/metabolism
- Cystic Fibrosis/physiopathology
- Cystic Fibrosis Transmembrane Conductance Regulator/genetics
- Cystic Fibrosis Transmembrane Conductance Regulator/metabolism
- Diabetes Mellitus, Type 1/blood
- Diabetes Mellitus, Type 1/epidemiology
- Diabetes Mellitus, Type 1/genetics
- Diabetes Mellitus, Type 1/physiopathology
- Diabetes Mellitus, Type 2/blood
- Diabetes Mellitus, Type 2/epidemiology
- Diabetes Mellitus, Type 2/genetics
- Diabetes Mellitus, Type 2/physiopathology
- Energy Metabolism/genetics
- Genetic Predisposition to Disease
- Humans
- Infant
- Infant, Newborn
- Infant, Newborn, Diseases/blood
- Infant, Newborn, Diseases/epidemiology
- Infant, Newborn, Diseases/genetics
- Infant, Newborn, Diseases/physiopathology
- Insulin/blood
- Islets of Langerhans/immunology
- Islets of Langerhans/metabolism
- Islets of Langerhans/pathology
- Islets of Langerhans/physiopathology
- Risk Factors
Collapse
Affiliation(s)
- Juergen Grulich-Henn
- University Children´s Hospital, University of Heidelberg, Im Neuenheimer Feld 430, Heidelberg, D-69120, Germany.
| | - Daniela Klose
- University Children´s Hospital, University of Heidelberg, Im Neuenheimer Feld 430, Heidelberg, D-69120, Germany
| |
Collapse
|
20
|
Heltshe SL, Cogen J, Ramos KJ, Goss CH. Cystic Fibrosis: The Dawn of a New Therapeutic Era. Am J Respir Crit Care Med 2017; 195:979-984. [PMID: 27710011 DOI: 10.1164/rccm.201606-1250pp] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Sonya L Heltshe
- 1 Division of Pediatric Pulmonology, Department of Pediatrics, and.,2 Cystic Fibrosis Foundation Therapeutics Development Network Coordinating Center, Seattle Children's Research Institute, Seattle, Washington
| | - Jonathan Cogen
- 1 Division of Pediatric Pulmonology, Department of Pediatrics, and
| | - Kathleen J Ramos
- 3 Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Washington, Seattle, Washington; and
| | - Christopher H Goss
- 1 Division of Pediatric Pulmonology, Department of Pediatrics, and.,3 Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Washington, Seattle, Washington; and.,2 Cystic Fibrosis Foundation Therapeutics Development Network Coordinating Center, Seattle Children's Research Institute, Seattle, Washington
| |
Collapse
|
21
|
Espel JC, Palac HL, Bharat A, Cullina J, Prickett M, Sala M, McColley SA, Jain M. The relationship between sweat chloride levels and mortality in cystic fibrosis varies by individual genotype. J Cyst Fibros 2017; 17:34-42. [PMID: 29221674 DOI: 10.1016/j.jcf.2017.11.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 11/09/2017] [Accepted: 11/09/2017] [Indexed: 01/20/2023]
Abstract
RATIONALE The association between CFTR genotype, sweat chloride and mortality has been inconsistent, but no previous analyses have examined the association stratified by individual genotypes. OBJECTIVES To evaluate the genotype-specific association between sweat chloride and mortality. METHODS The CFF Patient Registry was assessed and included all patients in the registry between 1996 and 2012 with at least one F508del allele. We excluded patients without a documented genotype or plausible sweat chloride level. The primary outcome was time to mortality during the observation period. We examined 15 genotypes using the three most prevalent alleles in each of 5 classes. We compared subgroups of sweat chloride using Kaplan-Meier curves, log-rank tests, and multivariable Cox PH models. The overall predictive value of sweat chloride on mortality was assessed using area under the receiver operating characteristic curves. MEASUREMENTS AND MAIN RESULTS 18,893 subjects met inclusion criteria. Sweat chloride distribution was similar across genotypes in patients with class 1 mutations, but was significantly different across genotypes in mutation classes 2-5. The R117H/F508del genotype patients demonstrated an association between sweat chloride and mortality (HR: 1.32 for every 10mmol/L increase in sweat chloride [95% CI 1.12-1.54]. There were also significant associations in patients with F508del/F508del, I507del/F508del, G551D/F508del and 2789+5G→A/F508del genotypes, though the clinical relevance for these genotypes is unclear. CONCLUSIONS There is significant variability in sweat chloride distribution across CFTR class 2-5 genotypes. The relationship between sweat chloride and mortality varies by genotype with a relatively strong relationship in R117H/F508del patients.
Collapse
Affiliation(s)
- Julia C Espel
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Hannah L Palac
- Department of Preventive Medicine, Northwestern University, Chicago, IL, United States
| | - Ankit Bharat
- Division of Thoracic Surgery, Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | | | - Michelle Prickett
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Marc Sala
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | | | - Manu Jain
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States.
| |
Collapse
|
22
|
Rowe S, Daines C, Ringshausen F, Kerem E, Wilson J, Tullis E, Nair N, Simard C, Han L, Ingenito E, McKee C, Lekstrom-Himes J, Davies J. Tezacaftor-Ivacaftor in Residual-Function Heterozygotes with Cystic Fibrosis. N Engl J Med 2017; 377:2024-2035. [PMID: 29099333 PMCID: PMC6472479 DOI: 10.1056/nejmoa1709847] [Citation(s) in RCA: 372] [Impact Index Per Article: 46.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Cystic fibrosis is an autosomal recessive disease caused by mutations in the CFTR gene that lead to progressive respiratory decline. Some mutant CFTR proteins show residual function and respond to the CFTR potentiator ivacaftor in vitro, whereas ivacaftor alone does not restore activity to Phe508del mutant CFTR. METHODS We conducted a randomized, double-blind, placebo-controlled, phase 3, crossover trial to evaluate the efficacy and safety of ivacaftor alone or in combination with tezacaftor, a CFTR corrector, in 248 patients 12 years of age or older who had cystic fibrosis and were heterozygous for the Phe508del mutation and a CFTR mutation associated with residual CFTR function. Patients were randomly assigned to one of six sequences, each involving two 8-week intervention periods separated by an 8-week washout period. They received tezacaftor-ivacaftor, ivacaftor monotherapy, or placebo. The primary end point was the absolute change in the percentage of predicted forced expiratory volume in 1 second (FEV1) from the baseline value to the average of the week 4 and week 8 measurements in each intervention period. RESULTS The number of analyzed intervention periods was 162 for tezacaftor-ivacaftor, 157 for ivacaftor alone, and 162 for placebo. The least-squares mean difference versus placebo with respect to the absolute change in the percentage of predicted FEV1 was 6.8 percentage points for tezacaftor-ivacaftor and 4.7 percentage points for ivacaftor alone (P<0.001 for both comparisons). Scores on the respiratory domain of the Cystic Fibrosis Questionnaire-Revised, a quality-of-life measure, also significantly favored the active-treatment groups. The incidence of adverse events was similar across intervention groups; most events were mild or moderate in severity, with no discontinuations of the trial regimen due to adverse events for tezacaftor-ivacaftor and few for ivacaftor alone (1% of patients) and placebo (<1%). CONCLUSIONS CFTR modulator therapy with tezacaftor-ivacaftor or ivacaftor alone was efficacious in patients with cystic fibrosis who were heterozygous for the Phe508del deletion and a CFTR residual-function mutation. (Funded by Vertex Pharmaceuticals and others; EXPAND ClinicalTrials.gov number, NCT02392234 .).
Collapse
Affiliation(s)
- S.M. Rowe
- Division of Pulmonary, Allergy & Critical Care Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - C. Daines
- Arizona Respiratory Center, University of Arizona, Tucson, AZ, United States
| | - F.C. Ringshausen
- Department of Respiratory Medicine, Hannover Medical School, and German Center for Lung Research (DZL), Hannover, Germany
| | - E. Kerem
- Hadassah Hebrew University Medical Center, Israel
| | | | - E. Tullis
- St. Michael’s Hospital, Toronto, ON, Canada
| | - N. Nair
- Vertex Pharmaceuticals Incorporated, Boston, MA, United States
| | - C. Simard
- Vertex Pharmaceuticals Incorporated, Boston, MA, United States
| | - L. Han
- Vertex Pharmaceuticals Incorporated, Boston, MA, United States
| | - E.P. Ingenito
- Vertex Pharmaceuticals Incorporated, Boston, MA, United States
| | - C. McKee
- Vertex Pharmaceuticals Incorporated, Boston, MA, United States
| | | | - J.C. Davies
- Royal Brompton & Harefield NHS Foundation Trust, Royal Brompton Hospital, London, United Kingdom
- Imperial College London, London, United Kingdom
| |
Collapse
|
23
|
CFTR-NHERF2-LPA₂ Complex in the Airway and Gut Epithelia. Int J Mol Sci 2017; 18:ijms18091896. [PMID: 28869532 PMCID: PMC5618545 DOI: 10.3390/ijms18091896] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2017] [Revised: 08/25/2017] [Accepted: 08/25/2017] [Indexed: 01/02/2023] Open
Abstract
The cystic fibrosis transmembrane conductance regulator (CFTR) is a cAMP- and cGMP-regulated chloride (Cl−) and bicarbonate (HCO3−) channel localized primarily at the apical plasma membrane of epithelial cells lining the airway, gut and exocrine glands, where it is responsible for transepithelial salt and water transport. Several human diseases are associated with altered CFTR channel function. Cystic fibrosis (CF) is caused by the absence or dysfunction of CFTR channel activity, resulting from mutations in the gene. Secretory diarrhea is caused by the hyperactivation of CFTR channel activity in the gastrointestinal tract. CFTR is a validated target for drug development to treat CF, and extensive research has been conducted to develop CFTR inhibitors for therapeutic interventions of secretory diarrhea. The intracellular processing, trafficking, apical membrane localization, and channel function of CFTR are regulated by dynamic protein–protein interactions in a complex network. In this paper, we review the current knowledge of a macromolecular complex of CFTR, Na+/H+ exchanger regulatory factor 2 (NHERF2), and lysophosphatidic acids (LPA) receptor 2 (LPA2) at the apical plasma membrane of airway and gut epithelial cells, and discuss its relevance in human physiology and diseases. We also explore the possibilities of targeting this complex to fine tune CFTR channel activity, with a hope to open up new avenues to develop novel therapies for CF and secretory diarrhea.
Collapse
|
24
|
Donaldson SH, Solomon GM, Zeitlin PL, Flume PA, Casey A, McCoy K, Zemanick ET, Mandagere A, Troha JM, Shoemaker SA, Chmiel JF, Taylor-Cousar JL. Pharmacokinetics and safety of cavosonstat (N91115) in healthy and cystic fibrosis adults homozygous for F508DEL-CFTR. J Cyst Fibros 2017; 16:371-379. [PMID: 28209466 DOI: 10.1016/j.jcf.2017.01.009] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 01/18/2017] [Accepted: 01/23/2017] [Indexed: 11/16/2022]
Abstract
BACKGROUND Cavosonstat (N91115), an orally bioavailable inhibitor of S-nitrosoglutathione reductase, promotes cystic fibrosis transmembrane conductance regulator (CFTR) maturation and plasma membrane stability, with a mechanism of action complementary to CFTR correctors and potentiators. METHODS A Phase I program evaluated pharmacokinetics, drug-drug interactions and safety of cavosonstat in healthy and cystic fibrosis (CF) subjects homozygous for F508del-CFTR. Exploratory outcomes included changes in sweat chloride in CF subjects. RESULTS Cavosonstat was rapidly absorbed and demonstrated linear and predictable pharmacokinetics. Exposure was unaffected by a high-fat meal or rifampin-mediated effects on drug metabolism and transport. Cavosonstat was well tolerated, with no dose-limiting toxicities or significant safety findings. At the highest dose, significant reductions from baseline in sweat chloride were observed (-4.1mmol/L; P=0.032) at day 28. CONCLUSIONS The favorable safety and clinical profile warrant further study of cavosonstat in CF. ClinicalTrials.gov Numbers: NCT02275936, NCT02013388, NCT02500667.
Collapse
Affiliation(s)
- Scott H Donaldson
- Cystic Fibrosis Research and Treatment Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | - George M Solomon
- Department of Medicine and the Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, AL, USA
| | - Pamela L Zeitlin
- Eudowood Division of Pediatric Respiratory Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Patrick A Flume
- Department of Medicine, Medical University of South Carolina, Charleston, SC, USA; Department of Pediatrics, Medical University of South Carolina, Charleston, SC, USA
| | - Alicia Casey
- Department of Medicine, Division of Respiratory Diseases, Boston Children's Hospital, Boston, MA, USA
| | - Karen McCoy
- Ohio State University, Nationwide Children's Hospital, Columbus, OH, USA
| | - Edith T Zemanick
- Department of Pediatrics, University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, CO, USA
| | | | | | | | - James F Chmiel
- Department of Pediatrics, Case Western Reserve University School of Medicine and Rainbow Babies and Children's Hospital, Cleveland, OH, USA
| | - Jennifer L Taylor-Cousar
- Department of Internal Medicine, Pulmonary Division, National Jewish Health, University of Colorado Health Sciences Center, Denver, CO, USA; Department of Pediatrics, Pulmonary Division, National Jewish Health, University of Colorado Health Sciences Center, Denver, CO, USA
| |
Collapse
|
25
|
Michl RK, Tabori H, Hentschel J, Beck JF, Mainz JG. Clinical approach to the diagnosis and treatment of cystic fibrosis and CFTR-related disorders. Expert Rev Respir Med 2016; 10:1177-1186. [DOI: 10.1080/17476348.2016.1240032] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
26
|
Gentili S, Mortali C, Mastrobattista L, Berretta P, Zaami S. Determination of different recreational drugs in sweat by headspace solid-phase microextraction gas chromatography mass spectrometry (HS-SPME GC/MS): Application to drugged drivers. J Pharm Biomed Anal 2016; 129:282-287. [DOI: 10.1016/j.jpba.2016.07.018] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 06/22/2016] [Accepted: 07/12/2016] [Indexed: 10/21/2022]
|
27
|
Within and beyond the communal turn to informed consent in industry-sponsored pharmacogenetics research: merits and challenges of community advisory boards. J Community Genet 2016; 7:261-270. [PMID: 27492247 DOI: 10.1007/s12687-016-0274-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 07/27/2016] [Indexed: 10/21/2022] Open
Abstract
The one-size-fits-all paradigm of drug development fails to address inter-individual variability in drug response. Pharmacogenetics research aims at studying the role of genotypic differences in drug response. Recently, the pharmaceutical industry has shown interest to embed pharmacogenetics studies in the process of drug development. Nevertheless, population-based and commercial aspects of such future-oriented studies pose challenges for individually based informed consent (IC). As an exemplar of the communal turn to IC procedures, community advisory boards (CABs) have been integrated into different types of medical research. CABs hold the promise of organizing the relationship between participants and researchers in a more reciprocal and participatory way, offering possible means of overcoming the lapses of individualistic IC. However, the involvement of CABs with pharmacogenetics research might be rife with difficulties, uncertainties, and challenges. The current study first reviews the existing literature to discuss added values and challenges of relying on CABs as a supplement to individually based IC. Then, the particular moral and regulatory landscape of pharmacogenetics research will be delineated to argue that community engagement is both necessary and promising beyond the communal turn to IC processes. Three main features of the landscape include (1) new supportive stances that some regulatory bodies have adopted toward pharmacogenetics research, (2) the motivation of the industry to draw reception and trust from the subpopulations, and (3) the important role of the society in generating and embedding pharmacogenetics knowledge. Finally, some points to consider will be discussed to contextualize relying on CABs within this landscape.
Collapse
|
28
|
Abstract
Cystic fibrosis (CF) is genetic autosomal recessive disease caused by reduced or absent function of CFTR protein. Treatments for patients with CF have primarily focused on the downstream end-organ consequences of defective CFTR. Since the discovery of the CFTR gene that causes CF in 1989 there have been tremendous advances in our understanding of the genetics and pathophysiology of CF. This has recently led to the development of new CFTR mutation-specific targeted therapies for select patients with CF. This review will discuss the characteristics of the CFTR gene, the CFTR mutations that cause CF and the new mutation specific pharmacological treatments including gene therapy that are contributing to the dawning of a new era in cystic fibrosis care.
Collapse
Affiliation(s)
- Suzanne C Carter
- National Referral Centre for Adult Cystic Fibrosis, St. Vincent's University Hospital, Elm Park, Dublin 4, Ireland
| | - Edward F McKone
- National Referral Centre for Adult Cystic Fibrosis, St. Vincent's University Hospital, Elm Park, Dublin 4, Ireland
| |
Collapse
|
29
|
Zhang W, Zhang X, Zhang YH, Strokes DC, Naren AP. Lumacaftor/ivacaftor combination for cystic fibrosis patients homozygous for Phe508del-CFTR. DRUGS OF TODAY (BARCELONA, SPAIN : 1998) 2016; 52:229-37. [PMID: 27252987 DOI: 10.1358/dot.2016.52.4.2467205] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Cystic fibrosis (CF) is a life-shortening inherited disease caused by the loss or dysfunction of the CF transmembrane conductance regulator (CFTR) channel activity resulting from mutations in the CFTR gene. Phe508del is the most prevalent mutation, with approximately 90% of all CF patients carrying it on at least one allele. Over the past two or three decades, significant progress has been made in understanding the pathogenesis of CF, and in the development of effective CF therapies. The approval of Orkambi® (lumacaftor/ivacaftor) marks another milestone in CF therapeutics development, which, with the advent of personalized medicine, could potentially revolutionize CF care and management. This article reviews the rationale, progress and future direction in the development of lumacaftor/ivacaftor combination to treat CF patients homozygous for the Phe508del-CFTR mutation.
Collapse
Affiliation(s)
- W Zhang
- Department of Pediatrics, Physiology, University of Tennessee Health Science Center, and University of Tennessee Cystic Fibrosis Care and Research Center at Le Bonheur Children's Hospital-Methodist University Hospital, Memphis, Tennessee, USA
| | - X Zhang
- Department of Pediatrics, University of Tennessee Health Science Center, and University of Tennessee Cystic Fibrosis Care and Research Center at Le Bonheur Children's Hospital-Methodist University Hospital, Memphis, Tennessee, USA
| | - Y H Zhang
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - D C Strokes
- Department of Pediatrics, University of Tennessee Health Science Center, and University of Tennessee Cystic Fibrosis Care and Research Center at Le Bonheur Children's Hospital-Methodist University Hospital, Memphis, Tennessee, USA
| | - A P Naren
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.
| |
Collapse
|
30
|
Cook DP, Rector MV, Bouzek DC, Michalski AS, Gansemer ND, Reznikov LR, Li X, Stroik MR, Ostedgaard LS, Abou Alaiwa MH, Thompson MA, Prakash YS, Krishnan R, Meyerholz DK, Seow CY, Stoltz DA. Cystic Fibrosis Transmembrane Conductance Regulator in Sarcoplasmic Reticulum of Airway Smooth Muscle. Implications for Airway Contractility. Am J Respir Crit Care Med 2016; 193:417-26. [PMID: 26488271 DOI: 10.1164/rccm.201508-1562oc] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE An asthma-like airway phenotype has been described in people with cystic fibrosis (CF). Whether these findings are directly caused by loss of CF transmembrane conductance regulator (CFTR) function or secondary to chronic airway infection and/or inflammation has been difficult to determine. OBJECTIVES Airway contractility is primarily determined by airway smooth muscle. We tested the hypothesis that CFTR is expressed in airway smooth muscle and directly affects airway smooth muscle contractility. METHODS Newborn pigs, both wild type and with CF (before the onset of airway infection and inflammation), were used in this study. High-resolution immunofluorescence was used to identify the subcellular localization of CFTR in airway smooth muscle. Airway smooth muscle function was determined with tissue myography, intracellular calcium measurements, and regulatory myosin light chain phosphorylation status. Precision-cut lung slices were used to investigate the therapeutic potential of CFTR modulation on airway reactivity. MEASUREMENTS AND MAIN RESULTS We found that CFTR localizes to the sarcoplasmic reticulum compartment of airway smooth muscle and regulates airway smooth muscle tone. Loss of CFTR function led to delayed calcium reuptake following cholinergic stimulation and increased myosin light chain phosphorylation. CFTR potentiation with ivacaftor decreased airway reactivity in precision-cut lung slices following cholinergic stimulation. CONCLUSIONS Loss of CFTR alters porcine airway smooth muscle function and may contribute to the airflow obstruction phenotype observed in human CF. Airway smooth muscle CFTR may represent a therapeutic target in CF and other diseases of airway narrowing.
Collapse
Affiliation(s)
- Daniel P Cook
- 1 Department of Internal Medicine.,2 Department of Molecular Physiology and Biophysics
| | | | | | | | | | | | | | | | | | | | - Michael A Thompson
- 3 Department of Anesthesiology and.,4 Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Y S Prakash
- 3 Department of Anesthesiology and.,4 Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Ramaswamy Krishnan
- 5 Department of Emergency Medicine, Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Boston, Massachusetts; and
| | | | - Chun Y Seow
- 7 Department of Pathology and Laboratory Medicine, James Hogg Research Centre/St. Paul's Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - David A Stoltz
- 1 Department of Internal Medicine.,2 Department of Molecular Physiology and Biophysics.,8 Department of Biomedical Engineering, and.,9 Pappajohn Biomedical Institute, University of Iowa, Iowa City, Iowa
| |
Collapse
|
31
|
Green IW, Glausier JR. Different Paths to Core Pathology: The Equifinal Model of the Schizophrenia Syndrome. Schizophr Bull 2016; 42:542-9. [PMID: 26392629 PMCID: PMC4838077 DOI: 10.1093/schbul/sbv136] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Schizophrenia is a clinically heterogeneous disorder that is perhaps more accurately characterized as "the schizophrenia syndrome." This clinical heterogeneity is reflected in the heterogeneous neurobiological presentations associated with the illness. Moreover, even highly specific neural aberrations that are associated with distinct symptoms of schizophrenia are linked to a wide range of risk factors. As such, any individual with schizophrenia likely has a particular set of risk factors that interact and converge to cross the disease threshold, forming a particular etiology that ultimately generates a core pathophysiology. This core pathophysiology may then produce 1 or more symptoms of schizophrenia, leading to common symptoms across individuals in spite of disparate etiologies. As such, the schizophrenia syndrome can be considered as anequifinalentity: a state of dysfunction that can arise from different upstream etiologies. Moreover, schizophrenia etiologies are multifactorial and can involve the interactive effects of a broad range of genetic, environmental, and developmental risk factors. Through a consideration of how disparate etiologies, caused by different sets of risk factors, converge on the same net dysfunction, this paper aims to model the equifinal nature of schizophrenia symptoms. To demonstrate the equifinal model, we discuss how maternal infection and adolescent cannabis use, 2 recognized schizophrenia risk factors, may interact with other genetic, environmental, and/or developmental risk factors to cause the conserved clinical presentation of impaired working memory.
Collapse
Affiliation(s)
- Isobel W. Green
- Department of Psychology, Harvard College, Harvard University, Cambridge, MA
| | - Jill R. Glausier
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA,*To whom correspondence should be addressed; Department of Psychiatry, Western Psychiatric Institute and Clinic, University of Pittsburgh, Biomedical Science Tower W1654, 3811 O’Hara Street, Pittsburgh, PA 15213, US; tel: 412-624-7869, fax: 412-624-9910, e-mail:
| |
Collapse
|
32
|
Jacquot J, Delion M, Gangloff S, Braux J, Velard F. Bone disease in cystic fibrosis: new pathogenic insights opening novel therapies. Osteoporos Int 2016; 27:1401-1412. [PMID: 26431978 DOI: 10.1007/s00198-015-3343-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 09/24/2015] [Indexed: 01/17/2023]
Abstract
Mutations within the gene encoding for the chloride ion channel cystic fibrosis transmembrane conductance regulator (CFTR) results in cystic fibrosis (CF), the most common lethal autosomal recessive genetic disease that causes a number of long-term health problems, as the bone disease. Osteoporosis and increased vertebral fracture risk associated with CF disease are becoming more important as the life expectancy of patients continues to improve. The etiology of low bone density is multifactorial, most probably a combination of inadequate peak bone mass during puberty and increased bone losses in adults. Body mass index, male sex, advanced pulmonary disease, malnutrition and chronic therapies are established additional risk factors for CF-related bone disease (CFBD). Consistently, recent evidence has confirmed that CFTR plays a major role in the osteoprotegerin (OPG) and COX-2 metabolite prostaglandin E2 (PGE2) production, two key regulators in the bone formation and regeneration. Several others mechanisms were also recognized from animal and cell models contributing to malfunctions of osteoblast (cell that form bone) and indirectly of bone-resorpting osteoclasts. Understanding such mechanisms is crucial for the development of therapies in CFBD. Innovative therapeutic approaches using CFTR modulators such as C18 have recently shown in vitro capacity to enhance PGE2 production and normalized the RANKL-to-OPG ratio in human osteoblasts bearing the mutation F508del-CFTR and therefore potential clinical utility in CFBD. This review focuses on the recently identified pathogenic mechanisms leading to CFBD and potential future therapies for treating CFBD.
Collapse
Affiliation(s)
- J Jacquot
- EA 4691, Biomatériaux et Inflammation en Site Osseux (BIOS), SFR CAP-Santé (FED 4231), Université Reims Champagne Ardenne, 1, Avenue du Maréchal Juin, 51095, Reims, France.
| | | | | | | | | |
Collapse
|
33
|
Abstract
RATIONALE In cystic fibrosis (CF), pulmonary exacerbations present an opportunity to define the effect of antibiotic therapy on systemic measures of inflammation. OBJECTIVES Investigate whether plasma inflammatory proteins demonstrate and predict a clinical response to antibiotic therapy and determine which proteins are associated with measures of clinical improvement. METHODS In this multicenter study, a panel of 15 plasma proteins was measured at the onset and end of treatment for pulmonary exacerbation and at a clinically stable visit in patients with CF who were 10 years of age or older. MEASUREMENTS AND MAIN RESULTS Significant reductions in 10 plasma proteins were observed in 103 patients who had paired blood collections during antibiotic treatment for pulmonary exacerbations. Plasma C-reactive protein, serum amyloid A, calprotectin, and neutrophil elastase antiprotease complexes correlated most strongly with clinical measures at exacerbation onset. Reductions in C-reactive protein, serum amyloid A, IL-1ra, and haptoglobin were most associated with improvements in lung function with antibiotic therapy. Having higher IL-6, IL-8, and α1-antitrypsin (α1AT) levels at exacerbation onset were associated with an increased risk of being a nonresponder (i.e., failing to recover to baseline FEV1). Baseline IL-8, neutrophil elastase antiprotease complexes, and α1AT along with changes in several plasma proteins with antibiotic treatment, in combination with FEV1 at exacerbation onset, were predictive of being a treatment responder. CONCLUSIONS Circulating inflammatory proteins demonstrate and predict a response to treatment of CF pulmonary exacerbations. A systemic biomarker panel could speed up drug discovery, leading to a quicker, more efficient drug development process for the CF community.
Collapse
|
34
|
Durupt S, Mazur S, Reix P. [Therapeutic advances in cystic fibrosis in 2014]. REVUE DE PNEUMOLOGIE CLINIQUE 2016; 72:77-86. [PMID: 25727661 DOI: 10.1016/j.pneumo.2014.11.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Revised: 11/03/2014] [Accepted: 11/08/2014] [Indexed: 06/04/2023]
Abstract
Twenty-five years after the cystic fibrosis (CF) gene identification, this discovery actually begins to benefit to patients. Increasing our knowledge on CFTR biology, as well as technical progress made in order to screen for new drugs have made therapeutic strategies move an important step forward. It is likely that in the forthcoming years, the panel of molecules available for CF patients will be larger, with new activators and potentiators. The disease by itself may consequently change in its natural history. CF is an example of the so-called personalized medicine, aiming to fit treatment according to patient's genetic background. Ongoing clinical trials may enlarge the actually limited eligible number of CF patients for new drugs such as ivacaftor. Beyond this exciting and promising new therapeutic approach, one may not push symptomatic treatments on the side. Improvements have been made for inhaled antibiotics administration, aiming to simplify patient's life; clinical trials using new molecules able to liquefy mucus or with anti-inflammatory properties are actually underway. One important next step in the care for CF will be to design and conduct early intervention trials in CF infants. Newborn screening program have been widely implanted around the word, and cohorts studies have shown that both functional and structural abnormalities occurred very early, making the therapeutic window of opportunity tight.
Collapse
Affiliation(s)
- S Durupt
- Service de médecine interne, centre de référence de la mucoviscidose, centre adulte de ressource et de compétences de la mucoviscidose, centre hospitalier Lyon-Sud, 69495 Pierre-Bénite cedex, France
| | - S Mazur
- Équipe EMET, UMR 5558, service de pédiatrie, d'allergologie et de pneumologie, centre de référence de la mucoviscidose, centre pédiatrique de ressources et de compétences de la mucoviscidose, hôpital femme-mère-enfant, 59, boulevard Pinel, 69677 Bron cedex, France
| | - P Reix
- Équipe EMET, UMR 5558, service de pédiatrie, d'allergologie et de pneumologie, centre de référence de la mucoviscidose, centre pédiatrique de ressources et de compétences de la mucoviscidose, hôpital femme-mère-enfant, 59, boulevard Pinel, 69677 Bron cedex, France.
| |
Collapse
|
35
|
Zhang X, Hothi JS, Zhang YH, Srinivasan S, Stokes DC, Zhang W. c.3623G > A mutation encodes a CFTR protein with impaired channel function. Respir Res 2016; 17:8. [PMID: 26800689 PMCID: PMC4724100 DOI: 10.1186/s12931-016-0326-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 01/19/2016] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND The aims of this study were to characterize clinical features of a pediatric African-American cystic fibrosis (CF) patient heterozygous for F508del and a novel c.3623G > A mutation, and to identify the molecular defect(s) associated with c.3623G > A mutation. METHODS The medical record of this patient was analyzed retrospectively. Western blotting and iodide efflux assay were used to study mutant CFTR protein expression level, maturation status, channel function, and the effects of CFTR modulation on these characteristics. RESULTS The encoding protein of c.3623G > A mutation, G1208D-CFTR, has a moderate processing defect and exhibits impaired channel function, which were partially rescued by using VX-809 or exposed to low temperature (28 °C). The patient has mild CF disease manifestations. CONCLUSIONS Our biochemical findings correlate with the clinical phenotype and suggest that c.3623G > A is a CF-causing mutation. The study helps expand our knowledge of rare CFTR mutations in a minority population and may have important clinical implications for personalized therapeutic intervention.
Collapse
Affiliation(s)
- Xiaoying Zhang
- Department of Pediatrics, University of Tennessee Health Science Center, 50 North Dunlap Street, Memphis, TN, 38103, USA.,University of Tennessee Cystic Fibrosis Care and Research Center, Le Bonheur Children's Hospital-Methodist University Hospital, Memphis, TN, 38103, USA
| | - Jaspal S Hothi
- Department of Pediatrics, University of Tennessee Health Science Center, 50 North Dunlap Street, Memphis, TN, 38103, USA.,University of Tennessee Cystic Fibrosis Care and Research Center, Le Bonheur Children's Hospital-Methodist University Hospital, Memphis, TN, 38103, USA
| | - Yanhui H Zhang
- Department of Bioscience Research, University of Tennessee Health Science Center, 875 Union Avenue, Memphis, TN, 38163, USA
| | - Saumini Srinivasan
- Department of Pediatrics, University of Tennessee Health Science Center, 50 North Dunlap Street, Memphis, TN, 38103, USA.,University of Tennessee Cystic Fibrosis Care and Research Center, Le Bonheur Children's Hospital-Methodist University Hospital, Memphis, TN, 38103, USA
| | - Dennis C Stokes
- Department of Pediatrics, University of Tennessee Health Science Center, 50 North Dunlap Street, Memphis, TN, 38103, USA.,University of Tennessee Cystic Fibrosis Care and Research Center, Le Bonheur Children's Hospital-Methodist University Hospital, Memphis, TN, 38103, USA
| | - Weiqiang Zhang
- Department of Pediatrics, University of Tennessee Health Science Center, 50 North Dunlap Street, Memphis, TN, 38103, USA. .,Department of Physiology, University of Tennessee Health Science Center, 50 North Dunlap Street, Room 309R, Memphis, TN, 38103, USA. .,University of Tennessee Cystic Fibrosis Care and Research Center, Le Bonheur Children's Hospital-Methodist University Hospital, Memphis, TN, 38103, USA.
| |
Collapse
|
36
|
Martin C, Burgel PR, Lepage P, Andréjak C, de Blic J, Bourdin A, Brouard J, Chanez P, Dalphin JC, Deslée G, Deschildre A, Gosset P, Touqui L, Dusser D. Host-microbe interactions in distal airways: relevance to chronic airway diseases. Eur Respir Rev 2015; 24:78-91. [PMID: 25726559 PMCID: PMC9487770 DOI: 10.1183/09059180.00011614] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
This article is the summary of a workshop, which took place in November 2013, on the roles of microorganisms in chronic respiratory diseases. Until recently, it was assumed that lower airways were sterile in healthy individuals. However, it has long been acknowledged that microorganisms could be identified in distal airway secretions from patients with various respiratory diseases, including cystic fibrosis (CF) and non-CF bronchiectasis, chronic obstructive pulmonary disease, asthma and other chronic airway diseases (e.g. post-transplantation bronchiolitis obliterans). These microorganisms were sometimes considered as infectious agents that triggered host immune responses and contributed to disease onset and/or progression; alternatively, microorganisms were often considered as colonisers, which were considered unlikely to play roles in disease pathophysiology. These concepts were developed at a time when the identification of microorganisms relied on culture-based methods. Importantly, the majority of microorganisms cannot be cultured using conventional methods, and the use of novel culture-independent methods that rely on the identification of microorganism genomes has revealed that healthy distal airways display a complex flora called the airway microbiota. The present article reviews some aspects of current literature on host–microbe (mostly bacteria and viruses) interactions in healthy and diseased airways, with a special focus on distal airways. Understanding host–microbe interactions in distal airways may lead to novel therapies for chronic airway diseaseshttp://ow.ly/HfENz
Collapse
Affiliation(s)
- Clémence Martin
- Hôpital Cochin, AP-HP, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Pierre-Régis Burgel
- Hôpital Cochin, AP-HP, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Patricia Lepage
- UMR1913-Microbiologie de l'Alimentation au Service de la Santé, l'Institut National de la Recherche Agronomique, Jouy-en-Josas, France
| | - Claire Andréjak
- Respiratory Intensive Care Unit, Centre Hospitalier Universitaire Sud, Amiens, France
| | - Jacques de Blic
- Hôpital Necker-Enfants Malades, AP-HP, Université Paris Descartes, Paris, France
| | - Arnaud Bourdin
- Hôpital Arnaud de Villeneuve, CHU Montpellier, Montpellier, France
| | - Jacques Brouard
- Dept of Pediatrics, CHU de Caen, Research Unit EA 4655 U2RM, Caen, France
| | - Pascal Chanez
- Dépt des Maladies Respiratoires, AP-HM, Laboratoire d'immunologie INSERM CNRS U 1067, UMR 7733, Aix Marseille Université, Marseille, France
| | | | - Gaetan Deslée
- Dept of Pulmonary Medicine, University Hospital of Reims, Reims, France
| | | | - Philippe Gosset
- Unité de défense innée et inflammation, Institut Pasteur, Paris, France INSERM U874, Paris, France
| | - Lhousseine Touqui
- Institut Pasteur de Lille, Centre d'Infection et d'Immunité de Lille, Lille, France
| | - Daniel Dusser
- Hôpital Cochin, AP-HP, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
37
|
Brennan ML, Schrijver I. Cystic Fibrosis: A Review of Associated Phenotypes, Use of Molecular Diagnostic Approaches, Genetic Characteristics, Progress, and Dilemmas. J Mol Diagn 2015; 18:3-14. [PMID: 26631874 DOI: 10.1016/j.jmoldx.2015.06.010] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 06/11/2015] [Accepted: 06/22/2015] [Indexed: 02/07/2023] Open
Abstract
Cystic fibrosis (CF) is an autosomal recessive disease with significant associated morbidity and mortality. It is now appreciated that the broad phenotypic CF spectrum is not explained by obvious genotype-phenotype correlations, suggesting that CF transmembrane conductance regulator (CFTR)-related disease may occur because of multiple additive effects. These contributing effects include complex CFTR alleles, modifier genes, mutations in alternative genes that produce CF-like phenotypes, epigenetic factors, and environmental influences. Most patients in the United States are now diagnosed through newborn screening and use of molecular testing methods. We review the molecular testing approaches and laboratory guidelines for carrier screening, prenatal testing, newborn screening, and clinical diagnostic testing, as well as recent developments in CF treatment, and reasons for the lack of a molecular diagnosis in some patients.
Collapse
Affiliation(s)
- Marie-Luise Brennan
- Department of Pathology, Stanford University Medical Center, Stanford, California
| | - Iris Schrijver
- Department of Pathology, Stanford University Medical Center, Stanford, California; Department of Pediatrics, Stanford University Medical Center, Stanford, California.
| |
Collapse
|
38
|
Solomon GM, Marshall SG, Ramsey BW, Rowe SM. Breakthrough therapies: Cystic fibrosis (CF) potentiators and correctors. Pediatr Pulmonol 2015; 50 Suppl 40:S3-S13. [PMID: 26097168 PMCID: PMC4620567 DOI: 10.1002/ppul.23240] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Revised: 05/29/2015] [Accepted: 06/03/2015] [Indexed: 12/28/2022]
Abstract
Cystic Fibrosis is caused by mutations in the Cystic Fibrosis Transmembrane conductance Regulator (CFTR) gene resulting in abnormal protein function. Recent advances of targeted molecular therapies and high throughput screening have resulted in multiple drug therapies that target many important mutations in the CFTR protein. In this review, we provide the latest results and current progress of CFTR modulators for the treatment of cystic fibrosis, focusing on potentiators of CFTR channel gating and Phe508del processing correctors for the Phe508del CFTR mutation. Special emphasis is placed on the molecular basis underlying these new therapies and emerging results from the latest clinical trials. The future directions for augmenting the rescue of Phe508del with CFTR modulators are also emphasized.
Collapse
Affiliation(s)
- George M Solomon
- Department of Medicine and the Gregory Fleming James Cystic Fibrosis Research Center, Birmingham, Alabama
| | - Susan G Marshall
- Division of Pulmonary Medicine, Department of Pediatrics, University of Washington School of Medicine, Seattle, Washington
| | - Bonnie W Ramsey
- Division of Pulmonary Medicine, Department of Pediatrics, University of Washington School of Medicine, Seattle, Washington.,Center for Clinical and Translational Research, Seattle Children's Research Institute, Seattle, Washington
| | - Steven M Rowe
- Department of Medicine and the Gregory Fleming James Cystic Fibrosis Research Center, Birmingham, Alabama.,Departments of Medicine, Pediatrics, Cell Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
39
|
|
40
|
Martin C, Hamard C, Kanaan R, Boussaud V, Grenet D, Abély M, Hubert D, Munck A, Lemonnier L, Burgel PR. Causes of death in French cystic fibrosis patients: The need for improvement in transplantation referral strategies! J Cyst Fibros 2015; 15:204-12. [PMID: 26391389 DOI: 10.1016/j.jcf.2015.09.002] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Revised: 08/30/2015] [Accepted: 09/06/2015] [Indexed: 11/16/2022]
Abstract
BACKGROUND Little data exist on causes of death in cystic fibrosis (CF) patients in the era of lung transplantation. METHODS Deaths in CF patients in France (2007-2010) were identified using the French CF Registry and causes of deaths were determined based on medical files by a mortality adjudication committee. RESULTS Of 256 deaths, half occurred after lung transplantation and were related to early or late complications of transplantation, whereas half occurred in patients who did not receive lung transplantation and were primarily related to respiratory failure or massive hemoptysis. Among patients who did not receive lung transplantation, only 19% died while waiting on a lung transplantation list. Lack of listing for lung transplantation was primarily related to late, or to lack of transplantation referral, rather than to contraindication to transplantation. CONCLUSIONS These data suggest that improvement in transplantation referral strategies may result in transplantation-related survival benefits.
Collapse
Affiliation(s)
- Clémence Martin
- Department of Respiratory Medicine, Cochin Hospital, Assistance Publique Hôpitaux de Paris, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France.
| | - Cécile Hamard
- Department of Respiratory Medicine, Cochin Hospital, Assistance Publique Hôpitaux de Paris, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France.
| | - Reem Kanaan
- Department of Respiratory Medicine, Cochin Hospital, Assistance Publique Hôpitaux de Paris, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France.
| | - Véronique Boussaud
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Department of Cardiac Surgery, Hôpital Européen Georges Pompidou, Assistance Publique Hôpitaux de Paris, Paris, France.
| | - Dominique Grenet
- Department of Respiratory Medicine, Hôpital Foch, Suresnes, France.
| | - Michel Abély
- Department of Pediatrics A, American Memorial Hospital, Reims, France.
| | - Dominique Hubert
- Department of Respiratory Medicine, Cochin Hospital, Assistance Publique Hôpitaux de Paris, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France.
| | - Anne Munck
- Department of Pediatric Gastroenterology and Respiratory Medicine, Hôpital Robert Debré, Assistance Publique Hôpitaux de Paris, University Paris 7, Paris, France.
| | | | - Pierre-Régis Burgel
- Department of Respiratory Medicine, Cochin Hospital, Assistance Publique Hôpitaux de Paris, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France.
| |
Collapse
|
41
|
Stafler P, Mei-Zahav M, Wilschanski M, Mussaffi H, Efrati O, Lavie M, Shoseyov D, Cohen-Cymberknoh M, Gur M, Bentur L, Livnat G, Aviram M, Alkrinawi S, Picard E, Prais D, Steuer G, Inbar O, Kerem E, Blau H. The impact of a national population carrier screening program on cystic fibrosis birth rate and age at diagnosis: Implications for newborn screening. J Cyst Fibros 2015; 15:460-6. [PMID: 26386752 DOI: 10.1016/j.jcf.2015.08.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Revised: 08/24/2015] [Accepted: 08/24/2015] [Indexed: 10/25/2022]
Abstract
BACKGROUND Population carrier screening (PCS) has been available in Israel since 1999 and universally subsidized since 2008. We sought to evaluate its impact. METHODS A retrospective review of governmental databanks, the national CF registry and CF centers. RESULTS CF rate per 100,000 live births has decreased from 14.5 in 1990 to 6 in 2011. From 2004-2011 there were 95 CF births: 22 utilized PCS; 68 (72%) had 2 known CFTR mutations; 37% were pancreatic sufficient. At diagnosis, age was 6 (0-98) months; 53/95 had respiratory symptoms, 41/95 failure to thrive and 19/95 pseudomonas. Thirty-four (36%) were Arabs and 19 (20%) orthodox Jews, compared to 20% and 8% respectively, in the general population. CONCLUSIONS PCS markedly reduced CF birth rates with a shift towards milder mutations, but was often avoided for cultural reasons. As children regularly have significant disease at diagnosis, we suggest a balanced approach, utilizing both PCS and newborn screening.
Collapse
Affiliation(s)
- Patrick Stafler
- 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.
| | - 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
| | - Michael Wilschanski
- Cystic Fibrosis Center, Hadassah-Hebrew University Medical Centre, Jerusalem, Israel
| | - Huda Mussaffi
- 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
| | - Ori Efrati
- Cystic Fibrosis Center, Sheba Medical Center, Tel Aviv, Israel
| | - Moran Lavie
- Cystic Fibrosis Center, Sheba Medical Center, Tel Aviv, Israel
| | - David Shoseyov
- Cystic Fibrosis Center, Hadassah-Hebrew University Medical Centre, Jerusalem, Israel
| | | | - Michal Gur
- Cystic Fibrosis Center, Rambam Medical Center, Haifa, Israel
| | - Lea Bentur
- Cystic Fibrosis Center, Rambam Medical Center, Haifa, Israel
| | - Galit Livnat
- Cystic Fibrosis Center, Carmel Medical Center, Haifa, Israel
| | - Micha Aviram
- Cystic Fibrosis Center, Soroka Medical Center, Beersheva, Israel
| | | | - Elie Picard
- Cystic Fibrosis Center, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Dario Prais
- Kathy and Lee Graub Cystic Fibrosis Center, Schneider Children's Medical Center of Israel, Petach Tikva, Israel
| | - Guy Steuer
- Kathy and Lee Graub Cystic Fibrosis Center, Schneider Children's Medical Center of Israel, Petach Tikva, Israel
| | - Ori Inbar
- Cystic Fibrosis Foundation of Israel, Israel
| | - Eitan Kerem
- Cystic Fibrosis Center, Hadassah-Hebrew University Medical Centre, Jerusalem, Israel
| | - Hannah Blau
- Kathy and Lee Graub Cystic Fibrosis Center, Schneider Children's Medical Center of Israel, Petach Tikva, Israel
| |
Collapse
|
42
|
Ramsey KA, Schultz A, Stick SM. Biomarkers in Paediatric Cystic Fibrosis Lung Disease. Paediatr Respir Rev 2015; 16:213-8. [PMID: 26051089 DOI: 10.1016/j.prrv.2015.05.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 05/06/2015] [Indexed: 01/15/2023]
Abstract
Biomarkers in cystic fibrosis are used i. for the measurement of cystic fibrosis transmembrane regulator function in order to diagnose cystic fibrosis, and ii. to assess aspects of lung disease severity (e.g. inflammation, infection). Effective biomarkers can aid disease monitoring and contribute to the development of new therapies. The tests of cystic fibrosis transmembrane regulator function each have unique strengths and weaknesses, and biomarkers of inflammation, infection and tissue destruction have the potential to enhance the management of cystic fibrosis through the early detection of disease processes. The development of biomarkers of cystic fibrosis lung disease, in particular airway inflammation and infection, is influenced by the challenges of obtaining relevant samples from infants and children for whom early detection and treatment of disease might have the greatest long term benefits.
Collapse
Affiliation(s)
- Kathryn A Ramsey
- Telethon Kids Institute, University of Western Australia, Australia; Cystic Fibrosis/Pulmonary Research and Treatment Center, University of North Carolina at Chapel Hill, North Carolina, USA
| | - André Schultz
- Telethon Kids Institute, University of Western Australia, Australia; Princess Margaret Hospital for Children, Western Australia, Australia; School of Paediatric and Child Health, University of Western Australia, Australia
| | - Stephen M Stick
- Telethon Kids Institute, University of Western Australia, Australia; Princess Margaret Hospital for Children, Western Australia, Australia; School of Paediatric and Child Health, University of Western Australia, Australia.
| |
Collapse
|
43
|
Mall MA, Galietta LJV. Targeting ion channels in cystic fibrosis. J Cyst Fibros 2015; 14:561-70. [PMID: 26115565 DOI: 10.1016/j.jcf.2015.06.002] [Citation(s) in RCA: 116] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Revised: 05/28/2015] [Accepted: 06/01/2015] [Indexed: 12/12/2022]
Abstract
Mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene cause a characteristic defect in epithelial ion transport that plays a central role in the pathogenesis of cystic fibrosis (CF). Hence, pharmacological correction of this ion transport defect by targeting of mutant CFTR, or alternative ion channels that may compensate for CFTR dysfunction, has long been considered as an attractive approach to a causal therapy of this life-limiting disease. The recent introduction of the CFTR potentiator ivacaftor into the therapy of a subgroup of patients with specific CFTR mutations was a major milestone and enormous stimulus for seeking effective ion transport modulators for all patients with CF. In this review, we discuss recent breakthroughs and setbacks with CFTR modulators designed to rescue mutant CFTR including the common mutation F508del. Further, we examine the alternative chloride channels TMEM16A and SLC26A9, as well as the epithelial sodium channel ENaC as alternative targets in CF lung disease, which remains the major cause of morbidity and mortality in patients with CF. Finally, we will focus on the hurdles that still need to be overcome to make effective ion transport modulation therapies available for all patients with CF irrespective of their CFTR genotype.
Collapse
Affiliation(s)
- Marcus A Mall
- Department of Translational Pulmonology, Translational Lung Research Center Heidelberg (TLRC), Member of the German Center for Lung Research (DZL), University of Heidelberg, Heidelberg, Germany; Division of Pediatric Pulmonology & Allergy and Cystic Fibrosis Center, Department of Pediatrics, University of Heidelberg, Heidelberg, Germany.
| | | |
Collapse
|
44
|
Barrio R. Management of endocrine disease: Cystic fibrosis-related diabetes: novel pathogenic insights opening new therapeutic avenues. Eur J Endocrinol 2015; 172:R131-41. [PMID: 25336504 DOI: 10.1530/eje-14-0644] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Cystic fibrosis (CF) is a recessive genetic disease caused by mutations in the CF transmembrane conductance regulator (CFTR). CFTR is primarily present in epithelial cells of the airways, intestine and in cells with exocrine and endocrine functions. Mutations in the gene encoding the channel protein complex (CFTR) cause alterations in the ionic composition of secretions from the lung, gastrointestinal tract, liver, and also the pancreas. CF-related diabetes (CFRD), the most common complication of CF, has a major detrimental impact on pulmonary function, nutrition and survival. Glucose derangements in CF seem to start from early infancy and, even when the pathophysiology is multifactorial, insulin insufficiency is clearly a major component. Consistently, recent evidence has confirmed that CFTR is an important regulator of insulin secretion by islet β-cells. In addition, several other mechanisms were also recognized from cellular and animals models also contributing to either β-cell mass reduction or β-cell malfunction. Understanding such mechanisms is crucial for the development of the so-called 'transformational' therapies in CF, including the preservation of insulin secretion. Innovative therapeutic approaches aim to modify specific CFTR mutant proteins or positively modulate their function. CFTR modulators have recently shown in vitro capacity to enhance insulin secretion and thereby potential clinical utility in CFDR, including synergistic effects between corrector and potentiator drugs. The introduction of incretins and the optimization of exocrine pancreatic replacement complete the number of therapeutic options of CFRD besides early diagnosis and implementation of insulin therapy. This review focuses on the recently identified pathogenic mechanisms leading to CFRD relevant for the development of novel pharmacological avenues in CFRD therapy.
Collapse
Affiliation(s)
- Raquel Barrio
- Pediatric Diabetes UnitDepartment of Pediatrics, Ramón y Cajal University Hospital, Alcalá University, Crta. Colmenar Km 9.1, 28034 Madrid, Spain
| |
Collapse
|
45
|
Comparative ex vivo, in vitro and in silico analyses of a CFTR splicing mutation: Importance of functional studies to establish disease liability of mutations. J Cyst Fibros 2015; 15:21-33. [PMID: 25735457 DOI: 10.1016/j.jcf.2015.02.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Revised: 01/09/2015] [Accepted: 02/04/2015] [Indexed: 11/23/2022]
Abstract
The Cystic Fibrosis p.Ile1234Val missense mutation actually creates a new dual splicing site possibly used either as a new acceptor or donor. Here, we aimed to test the accuracy of in silico predictions by comparing them with in vitro and ex vivo functional analyses of this mutation for an accurate CF diagnosis/prognosis. To this end, we applied a new in vitro strategy using a CFTR mini-gene which includes the complete CFTR coding sequence plus intron 22 (short version) which allows the assessment of alternatively spliced mRNA levels as well as the properties of the resulting abnormal CFTR protein regarding processing, intracellular localization and function. Our data demonstrate that p.Ile1234Val leads to usage of the alternative splicing donor (but not acceptor) resulting in alternative CFTR transcripts lacking 18 nts of exon 22 which produce a truncated CFTR protein with residual Cl- channel function. These results recapitulate data from native tissues of a CF patient. In conclusion, the existing in silico prediction models have limited application and ex vivo functional assessment of mutation effects should be made. Alternatively the in vitro strategy adopted here can be applied to assess the disease liability of mutations for an accurate CF diagnosis/prognosis.
Collapse
|
46
|
Affiliation(s)
- David A Stoltz
- Department of Internal Medicine, Roy J and Lucille A Carver College of Medicine, University of Iowa, Iowa City, Iowa 52242
- Department of Molecular Physiology and Biophysics, Roy J and Lucille A Carver College of Medicine, University of Iowa, Iowa City, Iowa 52242
- Department of Biomedical Engineering, University of Iowa, Iowa City, Iowa 52242
| | - David K Meyerholz
- Department of Pathology, Roy J and Lucille A Carver College of Medicine, University of Iowa, Iowa City, Iowa 52242
| | - Michael J Welsh
- Department of Internal Medicine, Roy J and Lucille A Carver College of Medicine, University of Iowa, Iowa City, Iowa 52242
- Department of Molecular Physiology and Biophysics, Roy J and Lucille A Carver College of Medicine, University of Iowa, Iowa City, Iowa 52242
- Howard Hughes Medical Institute, Roy J and Lucille A Carver College of Medicine University of Iowa, Iowa City, Iowa 52242
| |
Collapse
|
47
|
Liu Y, Wang L, Tian X, Xu KF, Xu W, Li X, Yue C, Zhang P, Xiao Y, Zhang X. Characterization of gene mutations and phenotypes of cystic fibrosis in Chinese patients. Respirology 2015; 20:312-8. [PMID: 25580864 DOI: 10.1111/resp.12452] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Revised: 08/03/2014] [Accepted: 09/29/2014] [Indexed: 12/25/2022]
Abstract
BACKGROUND AND OBJECTIVE Cystic fibrosis (CF) is a relatively common autosomal recessive disorder in Caucasians. CF is considered a very rare disease in Asians, and fewer than 30 Chinese CF patients are reported in the literature. We enrolled seven patients of Chinese Han origin diagnosed with CF at the Peking Union Medical College Hospital, to characterize gene mutations and phenotypes of CF in Chinese patients. METHODS We analysed the clinical presentation and screened the coding region of the CFTR gene for each patient. RESULTS Patients were 0-6 years old at onset of symptoms and were 10-28 years old at the time of diagnosis with CF. None of the seven patients had a family history of CF, and only one patient had parents who were consanguineous. Two patients had gastrointestinal symptoms but stool Sudan III results were normal. Four of the seven CF patients also had allergic bronchopulmonary aspergillosis. The concentration of chloride in patients' sweat ranged from 66 mmol/l to 154 mmol/l. In total, we identified 11 different mutations in seven CF patients, including one novel mutation (△E7-E11). Only one of these mutations (R553X) is present in the Caucasian CFTR common mutation-screening panel; and none of the 11 mutations are common in Caucasian CF patients. CONCLUSIONS CF in China is difficult to diagnose because of a combination of low awareness, atypical clinical symptoms, and a lack of sweat and genetic testing facilities in most hospitals. The mutations identified in Chinese CF patients are different from the common Caucasian gene mutations.
Collapse
Affiliation(s)
- Yaping Liu
- Department of Medical Genetics, School of Basic Medicine Peking Union Medical College, Beijing, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Reverri EJ, Morrissey BM, Cross CE, Steinberg FM. Inflammation, oxidative stress, and cardiovascular disease risk factors in adults with cystic fibrosis. Free Radic Biol Med 2014; 76:261-77. [PMID: 25172163 DOI: 10.1016/j.freeradbiomed.2014.08.005] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Revised: 07/31/2014] [Accepted: 08/05/2014] [Indexed: 12/21/2022]
Abstract
Cystic fibrosis (CF) represents one of a number of localized lung and non-lung diseases with an intense chronic inflammatory component associated with evidence of systemic oxidative stress. Many of these chronic inflammatory diseases are accompanied by an array of atherosclerotic processes and cardiovascular disease (CVD), another condition strongly related to inflammation and oxidative stress. As a consequence of a dramatic increase in long-lived patients with CF in recent decades, the specter of CVD must be considered in these patients who are now reaching middle age and beyond. Buttressed by recent data documenting that CF patients exhibit evidence of endothelial dysfunction, a recognized precursor of atherosclerosis and CVD, the spectrum of risk factors for CVD in CF is reviewed here. Epidemiological data further characterizing the presence and extent of atherogenic processes in CF patients would seem important to obtain. Such studies should further inform and offer mechanistic insights into how other chronic inflammatory diseases potentiate the processes leading to CVDs.
Collapse
Affiliation(s)
- Elizabeth J Reverri
- Department of Nutrition, University of California Davis, One Shields Avenue, 3135 Meyer Hall, Davis, CA 95616, USA
| | - Brian M Morrissey
- Adult Cystic Fibrosis Clinic and Division of Pulmonary-Critical Care Medicine, University of California Davis Medical Center, 4150 V Street, Sacramento, CA 95817, USA
| | - Carroll E Cross
- Adult Cystic Fibrosis Clinic and Division of Pulmonary-Critical Care Medicine, University of California Davis Medical Center, 4150 V Street, Sacramento, CA 95817, USA.
| | - Francene M Steinberg
- Department of Nutrition, University of California Davis, One Shields Avenue, 3135 Meyer Hall, Davis, CA 95616, USA
| |
Collapse
|
49
|
Barry PJ, Plant BJ, Nair A, Bicknell S, Simmonds NJ, Bell NJ, Shafi NT, Daniels T, Shelmerdine S, Felton I, Gunaratnam C, Jones AM, Horsley AR. Effects of ivacaftor in patients with cystic fibrosis who carry the G551D mutation and have severe lung disease. Chest 2014; 146:152-158. [PMID: 24522694 DOI: 10.1378/chest.13-2397] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
BACKGROUND The development of ivacaftor represents a significant advance in therapeutics for patients with cystic fibrosis (CF) who carry the G551D mutation. Patients with an FEV1 < 40% predicted represent a considerable proportion of eligible patients but were excluded from phase 3 clinical trials, and the effectiveness of the drug in this population is, therefore, unknown. METHODS Data were collected from adult CF centers in the United Kingdom and Ireland with patients enrolled in an ivacaftor compassionate use program (FEV1 < 40% or on lung transplant waiting list). Clinically recorded data were collated from patient records for 1 year prior and for a period of 90 to 270 days following ivacaftor commencement. Each patient was matched to two control subjects who would have met the requirements for the compassionate use program with the exception of genotype. RESULTS Twenty-one patients received ivacaftor for a median of 237 days. Mean FEV1 improved from 26.5% to 30.7% predicted (P = .01), representing a 16.7% relative improvement. Median weight improved from 49.8 to 51.6 kg (P = .006). Median inpatient IV antibiotic days declined from 23 to 0 d/y (P = .001) and median total IV treatment days decreased from 74 to 38 d/y (P = .002) following ivacaftor. Changes in pulmonary function and IV antibiotic requirements were significant compared with control subjects. CONCLUSIONS Ivacaftor was clinically effective in patients with CF who carry the G551D mutation and have severe pulmonary disease. The reductions in treatment requirements were clinically and statistically significant and have not been described in less severe populations.
Collapse
Affiliation(s)
- Peter J Barry
- University Hospital of South Manchester NHS Foundation Trust, Manchester, England
| | - Barry J Plant
- University Hospital of South Manchester NHS Foundation Trust, Manchester, England; Royal College of Physicians of Ireland, Dublin, Ireland; Cork University Hospital, University College Cork, Cork, Ireland
| | - Arjun Nair
- Royal Brompton Hospital and Imperial College, London, England
| | | | | | | | | | | | | | - Imogen Felton
- Royal Brompton Hospital and Imperial College, London, England
| | | | - Andrew M Jones
- University Hospital of South Manchester NHS Foundation Trust, Manchester, England
| | - Alex R Horsley
- University Hospital of South Manchester NHS Foundation Trust, Manchester, England; University of Manchester, Manchester Academic Health Science Centre, University Hospital of South Manchester NHS Foundation Trust, Manchester, England.
| |
Collapse
|
50
|
Rowe SM, Heltshe SL, Gonska T, Donaldson SH, Borowitz D, Gelfond D, Sagel SD, Khan U, Mayer-Hamblett N, Van Dalfsen JM, Joseloff E, Ramsey BW. Clinical mechanism of the cystic fibrosis transmembrane conductance regulator potentiator ivacaftor in G551D-mediated cystic fibrosis. Am J Respir Crit Care Med 2014; 190:175-84. [PMID: 24927234 DOI: 10.1164/rccm.201404-0703oc] [Citation(s) in RCA: 415] [Impact Index Per Article: 37.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
RATIONALE Ivacaftor is a cystic fibrosis transmembrane conductance regulator (CFTR) potentiator recently approved for patients with CF age 6 and older with the G551D mutation. OBJECTIVES To evaluate ivacaftor in a postapproval setting and determine mechanism of action and response of clinically relevant markers. METHODS We conducted a longitudinal cohort study in 2012-2013 in G551D CF patients age 6 and older with no prior exposure to ivacaftor. Study assessments were performed at baseline, 1, 3, and 6 months after ivacaftor initiation. Substudies evaluated mucociliary clearance, β-adrenergic sweat secretion rate, gastrointestinal pH, and sputum inflammation and microbiology Measurements and Main Results: A total of 151 of 153 subjects were prescribed ivacaftor and 88% completed the study through 6 months. FEV1 % predicted improved from baseline to 6 months (mean absolute change, 6.7%; P < 0.001). Similarly, body mass index improved from baseline to 6 months (mean change, 0.8 kg/m(2); P < 0.001). Sweat chloride decreased from baseline to 6 months (mean change, -53.8 mmol/L; 95% confidence interval, -57.7 to -49.9; P < 0.001), reflecting augmented CFTR function. There was significant improvement in hospitalization rate (P < 0.001) and Pseudomonas aeruginosa burden (P < 0.01). Significant improvements in mucociliary clearance (P < 0.001), gastrointestinal pH (P = 0.001), and microbiome were also observed, providing clinical mechanisms underlying the therapeutic benefit of ivacaftor. CONCLUSIONS Significant clinical and physiologic improvements were observed on initiation of ivacaftor in a broad patient population, including reduced infection with P. aeruginosa. Biomarker studies substantially improve the understanding of the mechanistic consequences of CFTR modulation on pulmonary and gastrointestinal physiology.
Collapse
Affiliation(s)
- Steven M Rowe
- 1 Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|