101
|
Trimble A, Zeman K, Wu J, Ceppe A, Bennett W, Donaldson S. Effect of airway clearance therapies on mucociliary clearance in adults with cystic fibrosis: A randomized controlled trial. PLoS One 2022; 17:e0268622. [PMID: 35594286 PMCID: PMC9122229 DOI: 10.1371/journal.pone.0268622] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 04/04/2022] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND Cystic fibrosis (CF) is an inherited disorder causing impaired mucociliary clearance within the respiratory tract, and is associated with bronchiectasis, chronic respiratory infections, and early death. Airway clearance therapies have long been a cornerstone of management of individuals with CF, although evidence supporting their use is lacking. We designed a randomized controlled trial to quantitatively compare the effects of different forms of airway clearance on mucociliary clearance. METHODS Three different physiotherapy methods to augment cough-clearance were studied in addition to cough-clearance alone: high-frequency chest-wall oscillating vest, oscillatory positive expiratory pressure, and whole-body vibration. We used gamma scintigraphy after inhalation of radiolabeled particles to quantify mucus clearance before, during, and after physiotherapy. As secondary endpoints, we measured concentrations of small molecules in exhaled breath that may impact mucus clearance. RESULTS Ten subjects were enrolled and completed study procedures. No differences were identified between any method of airway clearance, including cough clearance alone. We did identify changes in certain small molecule concentrations in exhaled breath following airway clearance. CONCLUSIONS Due to the limitations of this study, we do not believe the negative results suggest a change in clinical practice with regard to airway clearance. Findings pertaining to small molecules in exhaled breath may serve as future opportunities for study.
Collapse
Affiliation(s)
- Aaron Trimble
- Department of Medicine, Oregon Health and Science University (OHSU), Portland, Oregon, United States of America
- Department of Medicine, University of North Carolina (UNC), Chapel Hill, North Carolina, United States of America
| | - Kirby Zeman
- Department of Medicine, University of North Carolina (UNC), Chapel Hill, North Carolina, United States of America
| | - Jihong Wu
- Department of Medicine, University of North Carolina (UNC), Chapel Hill, North Carolina, United States of America
| | - Agathe Ceppe
- Department of Medicine, University of North Carolina (UNC), Chapel Hill, North Carolina, United States of America
| | - William Bennett
- Department of Medicine, University of North Carolina (UNC), Chapel Hill, North Carolina, United States of America
| | - Scott Donaldson
- Department of Medicine, University of North Carolina (UNC), Chapel Hill, North Carolina, United States of America
| |
Collapse
|
102
|
Keith JD, Henderson AG, Fernandez-Petty CM, Davis JM, Oden AM, Birket SE. Muc5b Contributes to Mucus Abnormality in Rat Models of Cystic Fibrosis. Front Physiol 2022; 13:884166. [PMID: 35574458 PMCID: PMC9096080 DOI: 10.3389/fphys.2022.884166] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 04/14/2022] [Indexed: 11/13/2022] Open
Abstract
Cystic fibrosis (CF) airway disease is characterized by excessive and accumulative mucus in the airways. Mucociliary clearance becomes defective as mucus secretions become hyperconcentrated and viscosity increases. The CFTR-knockout (KO) rat has been previously shown to progressively develop delayed mucociliary transport, secondary to increased viscoelasticity of airway secretions. The humanized-G551D CFTR rat model has demonstrated that abnormal mucociliary clearance and hyperviscosity is reversed by ivacaftor treatment. In this study, we sought to identify the components of mucus that changes as the rat ages to contribute to these abnormalities. We found that Muc5b concentrations, and to a lesser extent Muc5ac, in the airway were increased in the KO rat compared to WT, and that Muc5b concentration was directly related to the viscosity of the mucus. Additionally, we found that methacholine administration to the airway exacerbates these characteristics of disease in the KO, but not WT rat trachea. Lastly we determined that at 6 months of age, CF rats had mucus that was adherent to the airway epithelium, a process that is reversed by ivacaftor therapy in the hG551D rat. Overall, these data indicate that accumulation of Muc5b initiates the muco-obstructive process in the CF lung prior to infection.
Collapse
Affiliation(s)
- Johnathan D Keith
- Department of Medicine, Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Alexander G Henderson
- Department of Medicine, Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Courtney M Fernandez-Petty
- Department of Medicine, Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Joy M Davis
- Department of Medicine, Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Ashley M Oden
- Department of Medicine, Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Susan E Birket
- Department of Medicine, Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
103
|
Greaney C, Bohan K, Tecklenborg S, Casserly B, Green J, Van de Ven P, Robinson K, Tierney A. Diet quality in cystic fibrosis - associations with patient reported outcome measures and enablers and barriers to eating a healthy diet: A protocol paper for a mixed methods study. HRB Open Res 2022; 5:33. [PMID: 36091185 PMCID: PMC9428499 DOI: 10.12688/hrbopenres.13533.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/29/2022] [Indexed: 11/20/2022] Open
Abstract
Background: People with cystic fibrosis (PWCF) have increased energy requirements. However, in recent years concerns have emerged regarding the 'cystic fibrosis (CF) diet' in terms of reliance on energy-dense, nutrient poor foods, which tend to be higher in saturated fat, sugar, and salt. These foods lack essential nutrients and are aetiologically linked with diet-related chronic diseases. The aim is to explore habitual dietary intakes in PWCF and (i) assess adherence to CF dietary guidelines and population specific healthy eating guidelines; (ii) derive a diet quality score and the inflammatory potential for the average diet consumed by PWCF and assess associations with patient reported outcome measures; (iii) assess drivers for current consumption patterns and enablers and barriers to eating a healthy diet. Methods: The aim is to recruit between 100-180 PWCF. A mixed methods study will be performed. Using three-day food diaries and food frequency questionnaires, aims (i) and (ii) will be addressed. The Dietary Approaches to Stop Hypertension (DASH) score and Healthy Eating Index-International (HEI-I) will derive diet quality scores. The Dietary Inflammatory Index (DII®) will ascertain inflammatory potential of the diet. Validated questionnaires will be used to report health related quality of life measures. Online focus groups and semi-structured interview with PWCF will address aim (iii). Conclusions: It is timely to revise dietary priorities and targets for CF. However, a greater understanding of what adults with CF currently consume and what they require in terms of nutrition and dietary guidance into the future is needed. In doing so, this research will help to clarify nutrition priorities and simplify the dietary aspects of CF treatment, thereby supporting adherence.
Collapse
Affiliation(s)
- Cian Greaney
- School of Allied Health, University of Limerick, Limerick, V94 T9PX, Ireland
| | - Katie Bohan
- School of Allied Health, University of Limerick, Limerick, V94 T9PX, Ireland
| | - Sarah Tecklenborg
- Cystic Fibrosis Ireland, 24 Lower Rathmines Rd, Rathmines, Dublin, Ireland
| | - Brian Casserly
- Department of Respiratory Medicine, University Hospital Limerick, Dooradoyle, Limerick, V94 F858, Ireland
| | - James Green
- School of Allied Health, University of Limerick, Limerick, V94 T9PX, Ireland
- Health Research Institute, University of Limerick, Limerick, V94 T9PX, Ireland
| | - Pepijn Van de Ven
- Department of Electronic and Computer Engineering, University of Limerick, Limerick, V94 T9PX, Ireland
- Health Implementation Science and Technology Cluster, Health Research Institute, University of Limerick, Limerick, V94 T9PX, Ireland
| | - Katie Robinson
- School of Allied Health, University of Limerick, Limerick, V94 T9PX, Ireland
- Health Research Institute, University of Limerick, Limerick, V94 T9PX, Ireland
| | - Audrey Tierney
- School of Allied Health, University of Limerick, Limerick, V94 T9PX, Ireland
- Health Implementation Science and Technology Cluster, Health Research Institute, University of Limerick, Limerick, V94 T9PX, Ireland
- Department of Dietetics, Human Nutrition and Sport, La Trobe University, Melbourne, Victoria, 3086, Australia
| |
Collapse
|
104
|
Saluzzo F, Riberi L, Messore B, Loré NI, Esposito I, Bignamini E, De Rose V. CFTR Modulator Therapies: Potential Impact on Airway Infections in Cystic Fibrosis. Cells 2022; 11:cells11071243. [PMID: 35406809 PMCID: PMC8998122 DOI: 10.3390/cells11071243] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 03/27/2022] [Accepted: 03/31/2022] [Indexed: 11/16/2022] Open
Abstract
Cystic Fibrosis (CF) is an autosomal recessive disease caused by mutations in the gene encoding for the Cystic Fibrosis Transmembrane conductance Regulator (CFTR) protein, expressed on the apical surface of epithelial cells. CFTR absence/dysfunction results in ion imbalance and airway surface dehydration that severely compromise the CF airway microenvironment, increasing infection susceptibility. Recently, novel therapies aimed at correcting the basic CFTR defect have become available, leading to substantial clinical improvement of CF patients. The restoration or increase of CFTR function affects the airway microenvironment, improving local defence mechanisms. CFTR modulator drugs might therefore affect the development of chronic airway infections and/or improve the status of existing infections in CF. Thus far, however, the full extent of these effects of CFTR-modulators, especially in the long-term remains still unknown. This review aims to provide an overview of current evidence on the potential impact of CFTR modulators on airway infections in CF. Their role in affecting CF microbiology, the susceptibility to infections as well as the potential efficacy of their use in preventing/decreasing the development of chronic lung infections and the recurrent acute exacerbations in CF will be critically analysed.
Collapse
Affiliation(s)
- Francesca Saluzzo
- Emerging Bacterial Pathogens Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy;
| | - Luca Riberi
- Postgraduate School in Respiratory Medicine, University of Torino, 10124 Torino, Italy;
| | - Barbara Messore
- Adult Cystic Fibrosis Centre, Azienda Ospedaliero-Universitaria San Luigi Gonzaga, 10043 Orbassano, Italy;
| | - Nicola Ivan Loré
- WHO Collaborating Centre and TB Supranational Reference Laboratory, Emerging Bacterial Pathogens Unit, IRCCS Ospedale San Raffaele, 20132 Milan, Italy;
| | - Irene Esposito
- Paediatric Pulmonology Unit, Regina Margherita Hospital AOU Città della Salute e della Scienza, 10126 Torino, Italy; (I.E.); (E.B.)
| | - Elisabetta Bignamini
- Paediatric Pulmonology Unit, Regina Margherita Hospital AOU Città della Salute e della Scienza, 10126 Torino, Italy; (I.E.); (E.B.)
| | - Virginia De Rose
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy
- Correspondence:
| |
Collapse
|
105
|
Campbell CD, Barnett C, Sulaiman I. A clinicians’ review of the respiratory microbiome. Breathe (Sheff) 2022; 18:210161. [PMID: 36338247 PMCID: PMC9584600 DOI: 10.1183/20734735.0161-2021] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 03/02/2022] [Indexed: 11/25/2022] Open
Abstract
The respiratory microbiome and its impact in health and disease is now well characterised. With the development of next-generation sequencing and the use of other techniques such as metabolomics, the functional impact of microorganisms in different host environments can be elucidated. It is now clear that the respiratory microbiome plays an important role in respiratory disease. In some diseases, such as bronchiectasis, examination of the microbiome can even be used to identify patients at higher risk of poor outcomes. Furthermore, the microbiome can aid in phenotyping. Finally, development of multi-omic analysis has revealed interactions between the host and microbiome in some conditions. This review, although not exhaustive, aims to outline how the microbiome is investigated, the healthy respiratory microbiome and its role in respiratory disease. The respiratory microbiome encompasses bacterial, fungal and viral communities. In health, it is a dynamic structure and dysbiotic in disease. Dysbiosis can be related to disease severity and may be utilised to predict patients at clinical risk.https://bit.ly/3pNSgnA
Collapse
|
106
|
Luettich K, Sharma M, Yepiskoposyan H, Breheny D, Lowe FJ. An Adverse Outcome Pathway for Decreased Lung Function Focusing on Mechanisms of Impaired Mucociliary Clearance Following Inhalation Exposure. FRONTIERS IN TOXICOLOGY 2022; 3:750254. [PMID: 35295103 PMCID: PMC8915806 DOI: 10.3389/ftox.2021.750254] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 11/11/2021] [Indexed: 01/23/2023] Open
Abstract
Adverse outcome pathways (AOPs) help to organize available mechanistic information related to an adverse outcome into key events (KEs) spanning all organizational levels of a biological system(s). AOPs, therefore, aid in the biological understanding of a particular pathogenesis and also help with linking exposures to eventual toxic effects. In the regulatory context, knowledge of disease mechanisms can help design testing strategies using in vitro methods that can measure or predict KEs relevant to the biological effect of interest. The AOP described here evaluates the major processes known to be involved in regulating efficient mucociliary clearance (MCC) following exposures causing oxidative stress. MCC is a key aspect of the innate immune defense against airborne pathogens and inhaled chemicals and is governed by the concerted action of its functional components, the cilia and airway surface liquid (ASL). The AOP network described here consists of sequences of KEs that culminate in the modulation of ciliary beat frequency and ASL height as well as mucus viscosity and hence, impairment of MCC, which in turn leads to decreased lung function.
Collapse
Affiliation(s)
- Karsta Luettich
- Philip Morris International R&D, Philip Morris Products S.A., Neuchatel, Switzerland
| | - Monita Sharma
- PETA Science Consortium International e.V., Stuttgart, Germany
| | - Hasmik Yepiskoposyan
- Philip Morris International R&D, Philip Morris Products S.A., Neuchatel, Switzerland
| | - Damien Breheny
- British American Tobacco (Investments) Ltd., Group Research and Development, Southampton, United Kingdom
| | - Frazer J Lowe
- Broughton Nicotine Services, Earby, Lancashire, United Kingdom
| |
Collapse
|
107
|
O’Connor JB, Mottlowitz M, Kruk ME, Mickelson A, Wagner BD, Harris JK, Wendt CH, Laguna TA. Network Analysis to Identify Multi-Omic Correlations in the Lower Airways of Children With Cystic Fibrosis. Front Cell Infect Microbiol 2022; 12:805170. [PMID: 35360097 PMCID: PMC8960254 DOI: 10.3389/fcimb.2022.805170] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 02/16/2022] [Indexed: 11/13/2022] Open
Abstract
The leading cause of morbidity and mortality in cystic fibrosis (CF) is progressive lung disease secondary to chronic airway infection and inflammation; however, what drives CF airway infection and inflammation is not well understood. By providing a physiological snapshot of the airway, metabolomics can provide insight into these processes. Linking metabolomic data with microbiome data and phenotypic measures can reveal complex relationships between metabolites, lower airway bacterial communities, and disease outcomes. In this study, we characterize the airway metabolome in bronchoalveolar lavage fluid (BALF) samples from persons with CF (PWCF) and disease control (DC) subjects and use multi-omic network analysis to identify correlations with the airway microbiome. The Biocrates targeted liquid chromatography mass spectrometry (LC-MS) platform was used to measure 409 metabolomic features in BALF obtained during clinically indicated bronchoscopy. Total bacterial load (TBL) was measured using quantitative polymerase chain reaction (qPCR). The Qiagen EZ1 Advanced automated extraction platform was used to extract DNA, and bacterial profiling was performed using 16S sequencing. Differences in metabolomic features across disease groups were assessed univariately using Wilcoxon rank sum tests, and Random forest (RF) was used to identify features that discriminated across the groups. Features were compared to TBL and markers of inflammation, including white blood cell count (WBC) and percent neutrophils. Sparse supervised canonical correlation network analysis (SsCCNet) was used to assess multi-omic correlations. The CF metabolome was characterized by increased amino acids and decreased acylcarnitines. Amino acids and acylcarnitines were also among the features most strongly correlated with inflammation and bacterial burden. RF identified strong metabolomic predictors of CF status, including L-methionine-S-oxide. SsCCNet identified correlations between the metabolome and the microbiome, including correlations between a traditional CF pathogen, Staphylococcus, a group of nontraditional taxa, including Prevotella, and a subnetwork of specific metabolomic markers. In conclusion, our work identified metabolomic characteristics unique to the CF airway and uncovered multi-omic correlations that merit additional study.
Collapse
Affiliation(s)
- John B. O’Connor
- Department of Pediatrics, Division of Pulmonary and Sleep Medicine, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL, United States
- *Correspondence: John B. O’Connor,
| | - Madison Mottlowitz
- Department of Pediatrics, Division of Pulmonary and Sleep Medicine, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL, United States
| | - Monica E. Kruk
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, United States
| | - Alan Mickelson
- Department of Medicine, University of Minnesota, Minneapolis VA Medical Center, Minneapolis, MN, United States
| | - Brandie D. Wagner
- School of Medicine, University of Colorado, Aurora, CO, United States
- Colorado School of Public Health, University of Colorado Denver, Aurora, CO, United States
| | | | - Christine H. Wendt
- Department of Medicine, University of Minnesota, Minneapolis VA Medical Center, Minneapolis, MN, United States
| | - Theresa A. Laguna
- Department of Pediatrics, Division of Pulmonary and Sleep Medicine, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL, United States
- Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| |
Collapse
|
108
|
Petersen MC, Begnel L, Wallendorf M, Litvin M. Effect of elexacaftor-tezacaftor-ivacaftor on body weight and metabolic parameters in adults with cystic fibrosis. J Cyst Fibros 2022; 21:265-271. [PMID: 34862121 PMCID: PMC9999463 DOI: 10.1016/j.jcf.2021.11.012] [Citation(s) in RCA: 103] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 11/20/2021] [Accepted: 11/21/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND Though weight gain has been reported in some clinical trials of CFTR modulators, the effect of elexacaftor-tezacaftor-ivacaftor on body weight, body mass index (BMI), blood pressure, lipids and glycemic control in the real-world setting remains incompletely described. METHODS We performed a single-center, retrospective, observational analysis of the effect of elexacaftor-tezacaftor-ivacaftor on body weight and cardiometabolic parameters in 134 adult CF patients of the Washington University Adult Cystic Fibrosis Center. Body weight, BMI, and blood pressure were extracted from outpatient clinic visits for the year preceding and the period following the initiation of elexacaftor-tezacaftor-ivacaftor. Other metabolic parameters were extracted at baseline and at latest available follow-up. RESULTS A mean of 12.2 months of follow-up data was available for analysis. The mean rate of change in BMI was 1.47 kg/m2/yr (95% CI, 1.08 to 1.87) greater after initiation of elexacaftor-tezacaftor-ivacaftor. Significant increases in blood pressure were observed. In those without CFRD, random blood glucose and hemoglobin A1c were decreased after elexacaftor-tezacaftor-ivacaftor initiation. In those with CFRD, elexacaftor-tezacaftor-ivacaftor increased serum total cholesterol, HDL-cholesterol, and LDL-cholesterol. CONCLUSIONS In this single-center, retrospective, observational study of 134 adults with CF, initiation of elexacaftor-tezacaftor-ivacaftor was associated with increases in BMI at a mean follow up of 12.2 months. Changes in other cardiometabolic risk factors were also observed. Widespread use of elexacaftor-tezacaftor-ivacaftor may be expected to increase the incidence of overnutrition in the CF population.
Collapse
Affiliation(s)
- Max C Petersen
- Division of Endocrinology, Metabolism, & Lipid Research, Department of Medicine, Washington University, St. Louis, Missouri, USA
| | - Lauren Begnel
- Nutrition Support Service, Barnes-Jewish Hospital, St. Louis, Missouri, USA
| | - Michael Wallendorf
- Division of Biostatistics, Washington University, St. Louis, Missouri, USA
| | - Marina Litvin
- Division of Endocrinology, Metabolism, & Lipid Research, Department of Medicine, Washington University, St. Louis, Missouri, USA.
| |
Collapse
|
109
|
Ramsey BW, Bell SC. Cystic Fibrosis: A Disease in Transformation, yet More Work to Be Done! Am J Respir Crit Care Med 2022; 205:487-489. [PMID: 35073504 PMCID: PMC8906488 DOI: 10.1164/rccm.202112-2782ed] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Bonnie W Ramsey
- Department of Pediatrics University of Washington School of Medicine Seattle, Washington
- Seattle Children's Research Institute Seattle, Washington
| | - Scott C Bell
- Adult Cystic Fibrosis Centre The Prince Charles Hospital Brisbane, Australia
- Children's Health Research Centre The University of Queensland Brisbane, Australia
- Translational Research Institute Brisbane, Australia
| |
Collapse
|
110
|
Gabel ME, Fox CK, Grimes RA, Lowman JD, McDonald CM, Stallings VA, Michel SH. Overweight and cystic fibrosis: An unexpected challenge. Pediatr Pulmonol 2022; 57 Suppl 1:S40-S49. [PMID: 34738328 DOI: 10.1002/ppul.25748] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 10/06/2021] [Accepted: 10/18/2021] [Indexed: 02/06/2023]
Abstract
Achieving a healthy weight balance has been a central focus of care for people who have cystic fibrosis (CF). Over the years, the emphasis has primarily been on promoting weight gain to optimize pulmonary outcomes. With continued improvements in CF care, including highly effective CF modulators available for many people, the CF community is now experiencing a new challenge: addressing the concern that some people are gaining weight excessively. While at this time, we do not know to what extent overweight and obesity will affect health outcomes for people with CF, it is likely that excessive weight gain may have negative health impacts similar to those seen in the general population. In this paper, we review the history of nutritional guidelines for people with CF, as well as more recent trends toward overweight and obesity for some. A multidisciplinary approach is needed to collaboratively start the oftentimes difficult conversation regarding excessive weight gain, and to identify resources to help people achieve and maintain a healthy weight through diet, exercise, and behavioral modification.
Collapse
Affiliation(s)
- Megan E Gabel
- Department of Pediatrics, University of Rochester Medical Center, Rochester, New York, USA
| | - Claudia K Fox
- Department of Pediatrics, Center for Pediatric Obesity Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Rachel A Grimes
- Department of Psychiatry, Cook Children's Medical Center, Fort Worth, Texas, USA
| | - John D Lowman
- Department of Physical Therapy, Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Catherine M McDonald
- Department of Clinical Nutrition, Primary Children's Hospital, Salt Lake City, Utah, USA
| | - Virginia A Stallings
- Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Suzanne H Michel
- Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| |
Collapse
|
111
|
Tam RY, van Dorst JM, McKay I, Coffey M, Ooi CY. Intestinal Inflammation and Alterations in the Gut Microbiota in Cystic Fibrosis: A Review of the Current Evidence, Pathophysiology and Future Directions. J Clin Med 2022; 11:649. [PMID: 35160099 PMCID: PMC8836727 DOI: 10.3390/jcm11030649] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/22/2022] [Accepted: 01/25/2022] [Indexed: 12/12/2022] Open
Abstract
Cystic fibrosis (CF) is a life-limiting autosomal recessive multisystem disease. While its burden of morbidity and mortality is classically associated with pulmonary disease, CF also profoundly affects the gastrointestinal (GI) tract. Chronic low-grade inflammation and alterations to the gut microbiota are hallmarks of the CF intestine. The etiology of these manifestations is likely multifactorial, resulting from cystic fibrosis transmembrane conductance regulator (CFTR) dysfunction, a high-fat CF diet, and the use of antibiotics. There may also be a bidirectional pathophysiological link between intestinal inflammation and changes to the gut microbiome. Additionally, a growing body of evidence suggests that these GI manifestations may have significant clinical associations with growth and nutrition, quality of life, and respiratory function in CF. As such, the potential utility of GI therapies and long-term GI outcomes are areas of interest in CF. Further research involving microbial modulation and multi-omics techniques may reveal novel insights. This article provides an overview of the current evidence, pathophysiology, and future research and therapeutic considerations pertaining to intestinal inflammation and alterations in the gut microbiota in CF.
Collapse
Affiliation(s)
- Rachel Y. Tam
- Discipline of Paediatrics & Child Health, Randwick Clinical Campus, School of Clinical Medicine, UNSW Medicine & Health, University of New South Wales, Sydney, NSW 2031, Australia; (R.Y.T.); (J.M.v.D.); (M.C.)
| | - Josie M. van Dorst
- Discipline of Paediatrics & Child Health, Randwick Clinical Campus, School of Clinical Medicine, UNSW Medicine & Health, University of New South Wales, Sydney, NSW 2031, Australia; (R.Y.T.); (J.M.v.D.); (M.C.)
| | - Isabelle McKay
- Wagga Wagga Base Hospital, Wagga Wagga, NSW 2650, Australia;
| | - Michael Coffey
- Discipline of Paediatrics & Child Health, Randwick Clinical Campus, School of Clinical Medicine, UNSW Medicine & Health, University of New South Wales, Sydney, NSW 2031, Australia; (R.Y.T.); (J.M.v.D.); (M.C.)
- Department of Gastroenterology, Sydney Children’s Hospital Randwick, Sydney, NSW 2031, Australia
| | - Chee Y. Ooi
- Discipline of Paediatrics & Child Health, Randwick Clinical Campus, School of Clinical Medicine, UNSW Medicine & Health, University of New South Wales, Sydney, NSW 2031, Australia; (R.Y.T.); (J.M.v.D.); (M.C.)
- Department of Gastroenterology, Sydney Children’s Hospital Randwick, Sydney, NSW 2031, Australia
| |
Collapse
|
112
|
van Dorst JM, Tam RY, Ooi CY. What Do We Know about the Microbiome in Cystic Fibrosis? Is There a Role for Probiotics and Prebiotics? Nutrients 2022; 14:480. [PMID: 35276841 PMCID: PMC8840103 DOI: 10.3390/nu14030480] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 01/13/2022] [Accepted: 01/17/2022] [Indexed: 12/12/2022] Open
Abstract
Cystic fibrosis (CF) is a life-shortening genetic disorder that affects the cystic fibrosis transmembrane conductance regulator (CFTR) protein. In the gastrointestinal (GI) tract, CFTR dysfunction results in low intestinal pH, thick and inspissated mucus, a lack of endogenous pancreatic enzymes, and reduced motility. These mechanisms, combined with antibiotic therapies, drive GI inflammation and significant alteration of the GI microbiota (dysbiosis). Dysbiosis and inflammation are key factors in systemic inflammation and GI complications including malignancy. The following review examines the potential for probiotic and prebiotic therapies to provide clinical benefits through modulation of the microbiome. Evidence from randomised control trials suggest probiotics are likely to improve GI inflammation and reduce the incidence of CF pulmonary exacerbations. However, the highly variable, low-quality data is a barrier to the implementation of probiotics into routine CF care. Epidemiological studies and clinical trials support the potential of dietary fibre and prebiotic supplements to beneficially modulate the microbiome in gastrointestinal conditions. To date, limited evidence is available on their safety and efficacy in CF. Variable responses to probiotics and prebiotics highlight the need for personalised approaches that consider an individual's underlying microbiota, diet, and existing medications against the backdrop of the complex nutritional needs in CF.
Collapse
Affiliation(s)
- Josie M. van Dorst
- Discipline of Paediatrics & Child Health, Randwick Clinical Campus, School of Clinical Medicine, UNSW Medicine & Health, UNSW, Sydney 2031, Australia; (J.M.v.D.); (R.Y.T.)
| | - Rachel Y. Tam
- Discipline of Paediatrics & Child Health, Randwick Clinical Campus, School of Clinical Medicine, UNSW Medicine & Health, UNSW, Sydney 2031, Australia; (J.M.v.D.); (R.Y.T.)
| | - Chee Y. Ooi
- Discipline of Paediatrics & Child Health, Randwick Clinical Campus, School of Clinical Medicine, UNSW Medicine & Health, UNSW, Sydney 2031, Australia; (J.M.v.D.); (R.Y.T.)
- Molecular and Integrative Cystic Fibrosis (miCF) Research Centre, Sydney 2031, Australia
- Department of Gastroenterology, Sydney Children’s Hospital Randwick, Sydney 2031, Australia
| |
Collapse
|
113
|
Donaldson SH, Laube BL, Mogayzel P, Corcoran TE, Pilewski JM, Ceppe A, Wu J, Bhambhvani PG, Ratjen F, Sagel SD, Clancy J, Rowe SM, Bennett WD. Effect of lumacaftor-ivacaftor on mucociliary clearance and clinical outcomes in cystic fibrosis: Results from the PROSPECT MCC sub-study. J Cyst Fibros 2022; 21:143-145. [PMID: 34083123 PMCID: PMC8630086 DOI: 10.1016/j.jcf.2021.05.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 05/03/2021] [Accepted: 05/04/2021] [Indexed: 01/03/2023]
Abstract
CFTR function is required for normal mucociliary clearance (MCC) and cough-assisted clearance (CC). Lumacaftor-ivacaftor is approved for use in people with cystic fibrosis (CF) carrying two copies of F508del-CFTR. In this observational study performed at four study sites, we characterized the effect of lumacaftor-ivacaftor on mucociliary and cough clearance and related this to other clinical and research endpoints after one month of treatment. Twenty-five adolescents and adults were enrolled. No effect on whole lung MCC was observed, but CC was significantly increased. Sweat chloride improved by 18 mEq/L in this group, indicating a modest restoration of CFTR activity, but no demonstrable change in FEV1 or lung clearance index was observed. We speculate that the modest effect of lumacaftor-ivacaftor on CFTR function was insufficient to yield an improvement in MCC.
Collapse
Affiliation(s)
- Scott H. Donaldson
- Department of Medicine and the Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Beth L. Laube
- Department of Pediatrics, Johns Hopkins University, Baltimore, MD
| | - Peter Mogayzel
- Department of Pediatrics, Johns Hopkins University, Baltimore, MD
| | | | | | - Agathe Ceppe
- Department of Medicine and the Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Jihong Wu
- Department of Medicine and the Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Pradeep G. Bhambhvani
- Department of Medicine and the Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, AL
| | - Felix Ratjen
- Department of Pediatrics, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Scott D. Sagel
- Department of Pediatrics, Children’s Hospital Colorado and University of Colorado Anschutz Medical Campus, Aurora, CO
| | | | - Steven M. Rowe
- Department of Medicine and the Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, AL
| | - William D. Bennett
- Department of Medicine and the Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC
| |
Collapse
|
114
|
Medicina de precisión en fibrosis quística. REVISTA MÉDICA CLÍNICA LAS CONDES 2022. [DOI: 10.1016/j.rmclc.2021.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
115
|
Pseudomonas aeruginosa in the Cystic Fibrosis Lung. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1386:347-369. [DOI: 10.1007/978-3-031-08491-1_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
116
|
Novel Immunomodulatory Therapies for Respiratory Pathologies. COMPREHENSIVE PHARMACOLOGY 2022. [PMCID: PMC8238403 DOI: 10.1016/b978-0-12-820472-6.00073-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
117
|
Dowd C, Van Citters AD, Dieni O, Willis A, Powell L, Sabadosa KA. Design and methods for understanding the state of cystic fibrosis care amid the COVID-19 pandemic. J Cyst Fibros 2021; 20 Suppl 3:3-8. [PMID: 34930539 PMCID: PMC8683121 DOI: 10.1016/j.jcf.2021.08.028] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 08/12/2021] [Accepted: 08/24/2021] [Indexed: 02/06/2023]
Abstract
Background Novel therapies have dramatically changed cystic fibrosis (CF) and innovative care delivery systems are needed to meet future patient needs. Telehealth has been shown to be an efficient and desirable form of care delivery. The COVID-19 pandemic caused a rapid shift to telehealth, and this presented a unique opportunity to study facilitators, barriers, and satisfaction with this mode of care delivery. We aim to report survey methods, demographics and telehealth use among CF care programs, patients, and families during the pandemic. Methods CF programs completed two surveys between July 29 and September 18, 2020, and between April 19 and May 19, 2021. Patients and families completed a similar survey between August 31 and October 30, 2020. The surveys addressed topics assessing the pandemic's financial impact, telehealth modes and experiences, licensure and reimbursement issues, health screening, and remote monitoring. Quantitative data were analyzed with descriptive statistics and were compared to the CF Foundation Patient Registry. Results Most programs (278 at timepoint one and 274 at timepoint two) provided telehealth during the pandemic. The percent of visits containing either telephone or video components changed from 45% to 25% over the time periods. Additionally, 424 patients and families from various ages and backgrounds responded to the survey and 81% reported having a telehealth visit. Conclusions The pandemic accelerated telehealth adoption and these datasets are a valuable source for exploring telehealth barriers and facilitators, the quality-of-care experience, financial and workforce implications, the impact on underrepresented populations, and implications for coverage and reimbursement.
Collapse
Affiliation(s)
- Christopher Dowd
- Cystic Fibrosis Foundation, 4550 Montgomery Avenue, Suite 1100N, Bethesda, MD 20814, USA.
| | - Aricca D Van Citters
- The Dartmouth Institute for Health Policy & Clinical Practice, Geisel School of Medicine at Dartmouth, Williamson Translational Research Building, Level 5, One Medical Center Drive, Lebanon, NH 03766 United States
| | - Olivia Dieni
- Cystic Fibrosis Foundation, 4550 Montgomery Avenue, Suite 1100N, Bethesda, MD 20814, USA
| | - Anne Willis
- Cystic Fibrosis Foundation, 4550 Montgomery Avenue, Suite 1100N, Bethesda, MD 20814, USA
| | - Leslie Powell
- Cystic Fibrosis Foundation, 4550 Montgomery Avenue, Suite 1100N, Bethesda, MD 20814, USA
| | - Kathryn A Sabadosa
- Cystic Fibrosis Foundation, 4550 Montgomery Avenue, Suite 1100N, Bethesda, MD 20814, USA
| |
Collapse
|
118
|
Graeber SY, Vitzthum C, Pallenberg ST, Naehrlich L, Stahl M, Rohrbach A, Drescher M, Minso R, Ringshausen FC, Rueckes-Nilges C, Klajda J, Berges J, Yu Y, Scheuermann H, Hirtz S, Sommerburg O, Dittrich AM, Tümmler B, Mall MA. Effects of Elexacaftor/Tezacaftor/Ivacaftor Therapy on CFTR Function in Patients with Cystic Fibrosis and One or Two F508del Alleles. Am J Respir Crit Care Med 2021; 205:540-549. [PMID: 34936849 DOI: 10.1164/rccm.202110-2249oc] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE The CFTR modulator combination elexacaftor/tezacaftor/ivacaftor (ELX/TEZ/IVA) was shown to improve clinical outcomes and sweat chloride concentration (SCC) in patients with cystic fibrosis (CF) and one or two F508del alleles. However, the effect of ELX/TEZ/IVA on CFTR function in the airways and intestine has not been studied. OBJECTIVES To assess the effect of ELX/TEZ/IVA on CFTR function in airway and intestinal epithelia in patients with CF and one or two F508del alleles aged 12 years and older. METHODS This prospective observational multicenter study assessed clinical outcomes including FEV1 %predicted and body mass index, and the CFTR biomarkers SCC, nasal potential difference (NPD) and intestinal current measurement (ICM) before and 8-16 weeks after initiation of ELX/TEZ/IVA. MEASUREMENTS AND MAIN RESULTS A total of 107 patients with CF including 55 patients with one F508del and a minimal function mutation and 52 F508del homozygous patients were enrolled in this study. In patients with one F508del allele, NPD and ICM showed that ELX/TEZ/IVA improved CFTR function in nasal epithelia to a level of 46.5% (IQR, 27.5-72.4; P<0.001) and in intestinal epithelia to 41.8% of normal (IQR, 25.1-57.6; P<0.001). In F508del homozygous patients, ELX/TEZ/IVA exceeded improvement of CFTR function observed with TEZ/IVA and increased CFTR-mediated Cl- secretion to a level of 47.4% of normal (IQR, 19.3-69.2; P<0.001) in nasal and to 45.9% (IQR, 19.7-66.6; P<0.001) in intestinal epithelia. CONCLUSIONS Treatment with ELX/TEZ/IVA results in effective improvement of CFTR function in airway and intestinal epithelia in patients with CF and one or two F508del alleles.
Collapse
Affiliation(s)
- Simon Y Graeber
- Charité Universitätsmedizin Berlin, 14903, Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine and Cystic Fibrosis Center, Berlin, Germany.,Berlin Institute of Health, 522475, Berlin, Germany.,German Center for Lung Research, 542891, associated partner, Berlin, Germany
| | - Constanze Vitzthum
- Charité Universitätsmedizin Berlin, 14903, Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine and Cystic Fibrosis Center, Berlin, Germany.,German Center for Lung Research, 542891, associated partner, Berlin, Germany
| | - Sophia T Pallenberg
- Hannover Medical School, 9177, Department of Pediatric Pneumology, Allergology and Neonatology, Hannover, Germany.,German Center for Lung Research, 542891, Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Hannover, Germany
| | - Lutz Naehrlich
- Justus-Liebig-University Giessen, Department of Pediatrics, Giessen, Germany.,German Center for Lung Research, 542891, Universities of Giessen and Marburg Lung Center (UGMLC), Giessen, Germany
| | - Mirjam Stahl
- Charité Universitätsmedizin Berlin, 14903, Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine and Cystic Fibrosis Center, Berlin, Germany.,Berlin Institute of Health, 522475, Berlin, Germany.,German Center for Lung Research, 542891, associated partner, Berlin, Germany
| | - Alexander Rohrbach
- Charité Universitätsmedizin Berlin, 14903, Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine and Cystic Fibrosis Center, Berlin, Germany
| | - Marika Drescher
- Charité Universitätsmedizin Berlin, 14903, Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine and Cystic Fibrosis Center, Berlin, Germany.,German Center for Lung Research, 542891, associated partner, Berlin, Germany
| | - Rebecca Minso
- Hannover Medical School, 9177, Department of Pediatric Pneumology, Allergology and Neonatology, Hannover, Germany
| | - Felix C Ringshausen
- Hannover Medical School, 9177, Department of Respiratory Medicine, Hannover, Germany.,German Center for Lung Research, 542891, Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Hannover, Germany
| | | | - Jan Klajda
- Justus-Liebig-University Giessen, Department of Pediatrics, Giessen, Germany
| | - Julian Berges
- University of Heidelberg, 9144, Department of Pediatrics, Division of Pediatric Pulmonology and Allergy and Cystic Fibrosis Center, Heidelberg, Germany.,University of Heidelberg, 9144, Department of Translational Pulmonology, Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany
| | - Yin Yu
- University of Heidelberg, 9144, Department of Pediatrics, Division of Pediatric Pulmonology and Allergy and Cystic Fibrosis Center, Heidelberg, Germany.,University of Heidelberg, 9144, German Centre for Lung Research (DZL), Translational Lung Research Center Heidelberg (TLRC), Department of Translational Pulmonology, Heidelberg, Germany
| | - Heike Scheuermann
- University of Heidelberg, 9144, Department of Pediatrics, Division of Pediatric Pulmonology and Allergy and Cystic Fibrosis Center, Heidelberg, Germany.,University of Heidelberg, 9144, Department of Translational Pulmonology, Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany
| | - Stephanie Hirtz
- University of Heidelberg, 9144, Department of Pediatrics, Division of Pediatric Pulmonology and Allergy and Cystic Fibrosis Center, Heidelberg, Germany.,University of Heidelberg, 9144, Department of Translational Pulmonology, Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany
| | - Olaf Sommerburg
- University of Heidelberg, 9144, Department of Pediatrics, Division of Pediatric Pulmonology and Allergy and Cystic Fibrosis Center, Heidelberg, Germany.,University of Heidelberg, 9144, Department of Translational Pulmonology, Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany
| | - Anna-Maria Dittrich
- Hannover Medical School, 9177, Department of Pediatric Pneumology, Allergology and Neonatology, Hannover, Germany.,German Center for Lung Research, 542891, Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Hannover, Germany
| | - Burkhard Tümmler
- Hannover Medical School, 9177, Department of Pediatric Pneumology, Allergology and Neonatology, Hannover, Germany.,German Center for Lung Research, 542891, Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Hannover, Germany
| | - Marcus A Mall
- Charité Universitätsmedizin Berlin, 14903, Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine and Cystic Fibrosis Center, Berlin, Germany.,Berlin Institute of Health, 522475, Berlin, Germany.,German Center for Lung Research, 542891, associated partner, Berlin, Germany;
| |
Collapse
|
119
|
Abstract
Drugs called CFTR modulators improve the physiologic defect underlying cystic fibrosis (CF) and alleviate many disease manifestations. However, studies to date indicate that chronic lung infections that are responsible for most disease-related mortality generally persist. Here, we investigated whether combining the CFTR modulator ivacaftor with an intensive 3.5-month antibiotic course could clear chronic Pseudomonas aeruginosa or Staphylococcus aureus lung infections in subjects with R117H-CFTR, who are highly ivacaftor-responsive. Ivacaftor alone improved CFTR activity, and lung function and inflammation within 48 h, and reduced P. aeruginosa and S. aureus pathogen density by ∼10-fold within a week. Antibiotics produced an additional ∼10-fold reduction in pathogen density, but this reduction was transient in subjects who remained infected. Only 1/5 P. aeruginosa-infected and 1/7 S. aureus-infected subjects became persistently culture-negative after the combined treatment. Subjects appearing to clear infection did not have particularly favorable baseline lung function or inflammation, pathogen density or antibiotic susceptibility, or bronchiectasis scores on CT scans, but they did have remarkably low sweat chloride values before and after ivacaftor. All persistently P. aeruginosa-positive subjects remained infected by their pretreatment strain, whereas subjects persistently S. aureus-positive frequently lost and gained strains. This work suggests chronic CF infections may resist eradication despite marked and rapid modulator-induced improvements in lung infection and inflammation parameters and aggressive antibiotic treatment.
Collapse
|
120
|
Lin Q, Pilewski JM, Di YP. Acidic Microenvironment Determines Antibiotic Susceptibility and Biofilm Formation of Pseudomonas aeruginosa. Front Microbiol 2021; 12:747834. [PMID: 34867864 PMCID: PMC8640179 DOI: 10.3389/fmicb.2021.747834] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 10/20/2021] [Indexed: 01/02/2023] Open
Abstract
Pseudomonas aeruginosa is the most prevalent bacterial species that contribute to cystic fibrosis (CF) respiratory failure. The impaired function of CF transmembrane conductance regulator leads to abnormal epithelial Cl-/HCO3 - transport and acidification of airway surface liquid. However, it remains unclear why the CF lung is most commonly infected by Pseudomonas aeruginosa versus other pathogens. We carried out studies to investigate if lower pH helps Pseudomonas aeruginosa adapt and thrive in the CF-like acidic lung environment. Our results revealed that Pseudomonas aeruginosa generally forms more biofilm, induces antibiotic resistance faster in acidic conditions, and can be reversed by returning the acidic environment to physiologically neutral conditions. Pseudomonas aeruginosa appears to be highly adaptive to the CF-like acidic pH environment. By studying the effects of an acidic environment on bacterial response, we may provide a new therapeutic option in preventing chronic Pseudomonas aeruginosa infection and colonization.
Collapse
Affiliation(s)
- Qiao Lin
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA, United States
| | - Joseph M Pilewski
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Y Peter Di
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
121
|
Infield DT, Strickland KM, Gaggar A, McCarty NA. The molecular evolution of function in the CFTR chloride channel. J Gen Physiol 2021; 153:212705. [PMID: 34647973 PMCID: PMC8640958 DOI: 10.1085/jgp.202012625] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 08/11/2021] [Accepted: 09/09/2021] [Indexed: 12/13/2022] Open
Abstract
The ATP-binding cassette (ABC) transporter superfamily includes many proteins of clinical relevance, with genes expressed in all domains of life. Although most members use the energy of ATP binding and hydrolysis to accomplish the active import or export of various substrates across membranes, the cystic fibrosis transmembrane conductance regulator (CFTR) is the only known animal ABC transporter that functions primarily as an ion channel. Defects in CFTR, which is closely related to ABCC subfamily members that bear function as bona fide transporters, underlie the lethal genetic disease cystic fibrosis. This article seeks to integrate structural, functional, and genomic data to begin to answer the critical question of how the function of CFTR evolved to exhibit regulated channel activity. We highlight several examples wherein preexisting features in ABCC transporters were functionally leveraged as is, or altered by molecular evolution, to ultimately support channel function. This includes features that may underlie (1) construction of an anionic channel pore from an anionic substrate transport pathway, (2) establishment and tuning of phosphoregulation, and (3) optimization of channel function by specialized ligand–channel interactions. We also discuss how divergence and conservation may help elucidate the pharmacology of important CFTR modulators.
Collapse
Affiliation(s)
- Daniel T Infield
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA
| | | | - Amit Gaggar
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL.,Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, AL.,Program in Protease and Matrix Biology, University of Alabama at Birmingham, Birmingham, AL.,Birmingham Veterans Administration Medical Center, Birmingham, AL
| | - Nael A McCarty
- Department of Pediatrics, Emory University, Atlanta, GA.,Children's Healthcare of Atlanta Center for Cystic Fibrosis and Airways Disease Research, Emory University, Atlanta, GA
| |
Collapse
|
122
|
Averna M, Melotti P, Sorio C. Revisiting the Role of Leukocytes in Cystic Fibrosis. Cells 2021; 10:cells10123380. [PMID: 34943888 PMCID: PMC8699441 DOI: 10.3390/cells10123380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/17/2021] [Accepted: 11/29/2021] [Indexed: 11/30/2022] Open
Abstract
Cystic fibrosis in characterized by pulmonary bacterial colonization and hyperinflammation. Lymphocytes, monocytes/macrophages, neutrophils, and dendritic cells of patients with CF express functional CFTR and are directly affected by altered CFTR expression/function, impairing their ability to resolve infections and inflammation. However, the mechanism behind and the contribution of leukocytes in the pathogenesis of CF are still poorly characterized. The recent clinical introduction of specific CFTR modulators added an important tool not only for the clinical management of the disease but also to the investigation of the pathophysiological mechanisms related to CFTR dysfunction and dysregulated immunity. These drugs treat the basic defect in cystic fibrosis (CF) by increasing CFTR function with improvement of lung function and quality of life, and may improve clinical outcomes also by correcting the dysregulated immune function that characterizes CF. Measure of CFTR function, protein expression profiling and several omics methods were used to identify molecular changes in freshly isolated leukocytes of CF patients, highlighting two roles of leukocytes in CF: one more generally related to the mechanism(s) causing immune dysregulation in CF and unresolved inflammation, and another more applicative role, which identifies in myeloid cells, an important tool predictive of the therapeutic response of CF patients. In this review we will summarize available data on CFTR expression and function in leukocyte populations and will discuss potential clinical applications based on available data.
Collapse
Affiliation(s)
- Monica Averna
- Department of Experimental Medicine (DIMES), University of Genova, 16132 Genova, Italy;
| | - Paola Melotti
- Cystic Fibrosis Centre, Azienda Ospedaliera Universitaria Integrata Verona, 37126 Verona, Italy;
| | - Claudio Sorio
- Department of Medicine, General Pathology Division, University of Verona, 37134 Verona, Italy
- Correspondence: ; Tel.: +39-045-802-7688
| |
Collapse
|
123
|
Williamson M, Casey M, Gabillard-Lefort C, Alharbi A, Teo YQJ, McElvaney NG, Reeves EP. Current evidence on the effect of highly effective CFTR modulation on interleukin-8 in cystic fibrosis. Expert Rev Respir Med 2021; 16:43-56. [PMID: 34726115 DOI: 10.1080/17476348.2021.2001333] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
INTRODUCTION Cystic fibrosis (CF) is a genetically inherited disease, with mortality and morbidity associated with respiratory disease. The inflammatory response in CF is characterized by excessive neutrophil influx to the airways, mainly due to the increased local production and retention of interleukin-8 (IL-8), a potent neutrophil chemoattractant. AREAS COVERED We discuss how the chemokine IL-8 dominates the inflammatory profile of the airways in CF lung disease. Cystic fibrosis transmembrane conductance regulator (CFTR) modulator therapies are designed to correct the malfunctioning protein resulting from specific CFTR mutations. This review covers current evidence on the impact of CFTR impairment on levels of IL-8 and outlines the influence of effective CFTR modulation on inflammation in CF with a focus on cytokine production. Review of the literature was carried out using the PUBMED database, Google Scholar, and The Cochrane Library databases, using several appropriate generic terms. EXPERT OPINION Therapeutic interventions specifically targeting the defective CFTR protein have improved the outlook for CF. Accumulating studies on the effect of highly effective CFTR modulation on inflammation indicate an impact on IL-8 levels. Further studies are required to increase our knowledge of early onset innate inflammatory dysregulation and on anti-inflammatory mechanisms of CFTR modulators.
Collapse
Affiliation(s)
- Michael Williamson
- Royal College of Surgeons in Ireland, Irish Centre for Genetic Lung Disease, Department of Medicine, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland
| | - Michelle Casey
- Royal College of Surgeons in Ireland, Irish Centre for Genetic Lung Disease, Department of Medicine, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland
| | - Claudie Gabillard-Lefort
- Royal College of Surgeons in Ireland, Irish Centre for Genetic Lung Disease, Department of Medicine, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland
| | - Aram Alharbi
- Royal College of Surgeons in Ireland, Irish Centre for Genetic Lung Disease, Department of Medicine, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland
| | - Yu Qing Jolene Teo
- Royal College of Surgeons in Ireland, Irish Centre for Genetic Lung Disease, Department of Medicine, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland
| | - Noel G McElvaney
- Royal College of Surgeons in Ireland, Irish Centre for Genetic Lung Disease, Department of Medicine, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland
| | - Emer P Reeves
- Royal College of Surgeons in Ireland, Irish Centre for Genetic Lung Disease, Department of Medicine, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland
| |
Collapse
|
124
|
Nichols DP, Paynter AC, Heltshe SL, Donaldson SH, Frederick CA, Freedman SD, Gelfond D, Hoffman LR, Kelly A, Narkewicz MR, Pittman JE, Ratjen F, Rosenfeld M, Sagel SD, Schwarzenberg SJ, Singh PK, Solomon GM, Stalvey MS, Clancy JP, Kirby S, Van Dalfsen JM, Kloster MH, Rowe SM. Clinical Effectiveness of Elexacaftor/Tezacftor/Ivacaftor in People with Cystic Fibrosis. Am J Respir Crit Care Med 2021; 205:529-539. [PMID: 34784492 PMCID: PMC8906485 DOI: 10.1164/rccm.202108-1986oc] [Citation(s) in RCA: 214] [Impact Index Per Article: 53.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Rationale The cystic fibrosis (CF) modulator drug, elexacaftor/tezacaftor/ivacaftor (ETI), proved highly effective in controlled clinical trials for individuals with at least one F508del allele, which occurs in at least 85% of people with CF. Objectives PROMISE is a postapproval study to understand the broad effects of ETI through 30 months’ clinical use in a more diverse U.S. patient population with planned analyses after 6 months. Methods Prospective, observational study in 487 people with CF age 12 years or older with at least one F508del allele starting ETI for the first time. Assessments occurred before and 1, 3, and 6 months into ETI therapy. Outcomes included change in percent predicted FEV1 (ppFEV1), sweat chloride concentration, body mass index (BMI), and self-reported respiratory symptoms. Measurements and Main Results Average age was 25.1 years, and 44.1% entered the study using tezacaftor/ivacaftor or lumacaftor/ivacaftor, whereas 6.7% were using ivacaftor, consistent with F508del homozygosity and G551D allele, respectively. At 6 months into ETI therapy, ppFEV1 improved 9.76 percentage points (95% confidence interval [CI], 8.76 to 10.76) from baseline, cystic fibrosis questionnaire–revised respiratory domain score improved 20.4 points (95% CI, 18.3 to 22.5), and sweat chloride decreased −41.7 mmol/L (95% CI, −43.8 to −39.6). BMI also significantly increased. Changes were larger in those naive to modulators but substantial in all groups, including those treated with ivacaftor at baseline. Conclusions ETI by clinical prescription provided large improvements in lung function, respiratory symptoms, and BMI in a diverse population naive to modulator drug therapy, using existing two-drug combinations, or using ivacaftor alone. Each group also experienced significant reductions in sweat chloride concentration, which correlated with improved ppFEV1 in the overall study population. Clinical trial registered with www.clinicaltrials.gov (NCT NCT04038047).
Collapse
Affiliation(s)
- David P Nichols
- University of Washington School of Medicine, 12353, Pediatrics, Seattle, Washington, United States.,Seattle Children's Hospital, 7274, Pediatric Pulmonology, Seattle, Washington, United States;
| | - Alex C Paynter
- Seattle Children's Research Institute, 145793, Seattle, Washington, United States
| | - Sonya L Heltshe
- Seattle Children's Research Institute, Cystic Fibrosis Therapeutics Development Network Coordinating Center, Seattle, Washington, United States.,University of Washington School of Medicine, Pediatrics, Seattle, Washington, United States
| | | | - Carla A Frederick
- University at Buffalo Jacobs School of Medicine and Biomedical Sciences, 12291, Buffalo, New York, United States
| | - Steven D Freedman
- Beth Israel Deaconess Medical Center, Gastroenterology, Boston, Massachusetts, United States
| | - Daniel Gelfond
- Western New York Pediatric Gastroenterology, Batavia, New York, United States
| | - Lucas R Hoffman
- University of Washington School of Medicine, 12353, Seattle, Washington, United States.,Seattle Children's Hospital, 7274, Seattle, Washington, United States
| | - Andrea Kelly
- The Children's Hospital of Philadelphia, 6567, Pediatrics, Division of Endocrinology & Diabetes, Philadelphia, Pennsylvania, United States
| | - Michael R Narkewicz
- Children's Hospital Colorado and University of Colorado School of Medicine, Digestive Health Institute and Pediatrics, Aurora, Colorado, United States
| | - Jessica E Pittman
- Washington University in Saint Louis School of Medicine, 12275, Department of Pediatrics, Saint Louis, Missouri, United States
| | - Felix Ratjen
- University of Toronto HSC, Division of Respiratory Medicine, Toronto, Ontario, Canada
| | - Margaret Rosenfeld
- Seattle Children's, Pediatrics / Pulmonary, Seattle, Washington, United States
| | - Scott D Sagel
- University of Colorado School of Medicine, Pediatrics, Aurora, Colorado, United States
| | - Sarah Jane Schwarzenberg
- University of Minnesota Masonic Children's Hospital, 501779, Pediatrics, Minneapolis, Minnesota, United States
| | - Pradeep K Singh
- University of Washington, 7284, Department of Microbiology and Medicine, Seattle, Washington, United States
| | - George M Solomon
- University of Alabama at Birmingham, 9968, Medicine, Gregory Fleming James Cystic Fibrosis Research Center, Birmingham, Alabama, United States
| | - Michael S Stalvey
- The University of Alabama at Birmingham School of Medicine, 9967, Birmingham, Alabama, United States
| | - John P Clancy
- Cincinnati Children's Hospital Medical Center, 2518, Department of Pediatrics, Cincinnati, Ohio, United States.,Cincinnati Children's Hospital Medical Center, 2518, Division of Pulmonary Medicine, Cincinnati, Ohio, United States
| | - Shannon Kirby
- Seattle Children's Research Institute, 145793, Seattle, Washington, United States
| | - Jill M Van Dalfsen
- Cystic Fibrosis Foundation Therapeutics Development Network Coordinating Center, Seattle Children's Research Institute, Seattle, Washington, United States
| | - Margaret H Kloster
- Seattle Children's Research Institute, Cystic Fibrosis Therapeutics Development Network Coordinating Center, Seattle, Washington, United States
| | - Steven M Rowe
- University of Alabama at Birmingham, Medicine, Cystic Fibrosis Center, Birmingham, Alabama, United States
| | | |
Collapse
|
125
|
Dumas MP, Xia S, Bear CE, Ratjen F. Perspectives on the translation of in-vitro studies to precision medicine in Cystic Fibrosis. EBioMedicine 2021; 73:103660. [PMID: 34740114 PMCID: PMC8577330 DOI: 10.1016/j.ebiom.2021.103660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 10/04/2021] [Accepted: 10/15/2021] [Indexed: 11/22/2022] Open
Abstract
Recent strides towards precision medicine in Cystic Fibrosis (CF) have been made possible by patient-derived in-vitro assays with the potential to predict clinical response to small molecule-based therapies. Here, we discuss the status of primary and stem-cell derived tissues used to evaluate the preclinical efficacy of CFTR modulators highlighting both their potential and limitations. Validation of these assays requires correlation of in-vitro responses to in-vivo measures of clinical biomarkers of disease outcomes. While initial efforts have shown some success, this translation requires methodologies that are sensitive enough to capture treatment responses in a CF population that now predominantly has mild lung disease. Future development of in-vitro and in-vivo biomarkers will facilitate the generation of new therapeutics particularly for those patients with rare mutations where clinical trials are not feasible so that in the future every CF patient will have access to effective targeted therapies.
Collapse
Affiliation(s)
- Marie-Pier Dumas
- Respiratory Medicine, Hospital for Sick Children, Toronto, Canada; Translational Medicine, Hospital for Sick Children, Toronto, Canada
| | - Sunny Xia
- Molecular Medicine, Hospital for Sick Children, Toronto, Canada.; Department of Physiology, University of Toronto, Toronto, Canada
| | - Christine E Bear
- Molecular Medicine, Hospital for Sick Children, Toronto, Canada.; Department of Physiology, University of Toronto, Toronto, Canada; Department of Biochemistry University of Toronto, Toronto, Canada
| | - Felix Ratjen
- Respiratory Medicine, Hospital for Sick Children, Toronto, Canada; Translational Medicine, Hospital for Sick Children, Toronto, Canada
| |
Collapse
|
126
|
Pallenberg ST, Junge S, Ringshausen FC, Sauer-Heilborn A, Hansen G, Dittrich AM, Tümmler B, Nietert M. CFTR modulation with elexacaftor-tezacaftor-ivacaftor in people with cystic fibrosis assessed by the β-adrenergic sweat rate assay. J Cyst Fibros 2021; 21:442-447. [PMID: 34756683 DOI: 10.1016/j.jcf.2021.10.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 09/22/2021] [Accepted: 10/14/2021] [Indexed: 12/22/2022]
Abstract
BACKGROUND The cystic fibrosis (CF) sweat gland is defective in β-adrenergically-stimulated sweat secretion in the coil and chloride reabsorption in the duct. Whereas chloride reabsorption is regularly assessed by quantitative pilocarpine iontophoresis (QPIT), the measurement of β-adrenergic sweat secretion is not yet established in clinical practice. METHODS A novel sweat bubble imaging protocol was developed that determines sweat secretion rates by automatic recording, processing and quality control of the kinetics of sweat droplet formation. RESULTS Treatment of CF patients with the CFTR modulators elexacaftor, tezacaftor and ivacaftor reduced the sweat chloride concentration measured in QPIT in the majority of patients to values in the intermediate or normal range. In contrast, the β-adrenergically-stimulated sweat secretion rate assayed by the automated bubble sweat test was normalized in only 3 patients, slightly increased in 12 patients and remained undetectable in 8 patients. CONCLUSIONS β-adrenergic sweat stimulation in the coil is apparently rather stringent in its requirements for a wild type CFTR conformation whereas chloride reabsorption in the duct tolerates residual structural and functional deficits of native or pharmacologically rescued mutant CFTR in the apical membrane.
Collapse
Affiliation(s)
- Sophia Theres Pallenberg
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
| | - Sibylle Junge
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
| | - Felix C Ringshausen
- Department of Respiratory Medicine, Hannover Medical School, Hannover, Germany; Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research, Hannover Medical School, Hannover, Germany
| | | | - Gesine Hansen
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany; Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research, Hannover Medical School, Hannover, Germany
| | - Anna Maria Dittrich
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany; Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research, Hannover Medical School, Hannover, Germany
| | - Burkhard Tümmler
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany; Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research, Hannover Medical School, Hannover, Germany
| | - Manuel Nietert
- Department of Medical Bioinformatics, University Medical Center Göttingen, Göttingen, Germany.
| |
Collapse
|
127
|
O’Connor JB, Mottlowitz MM, Wagner BD, Boyne KL, Stevens MJ, Robertson CE, Harris JK, Laguna TA. Divergence of bacterial communities in the lower airways of CF patients in early childhood. PLoS One 2021; 16:e0257838. [PMID: 34613995 PMCID: PMC8494354 DOI: 10.1371/journal.pone.0257838] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 09/10/2021] [Indexed: 11/18/2022] Open
Abstract
Rationale Chronic airway infection and inflammation resulting in progressive, obstructive lung disease is the leading cause of morbidity and mortality in cystic fibrosis. Understanding the lower airway microbiota across the ages can provide valuable insight and potential therapeutic targets. Objectives To characterize and compare the lower airway microbiota in cystic fibrosis and disease control subjects across the pediatric age spectrum. Methods Bronchoalveolar lavage fluid samples from 191 subjects (63 with cystic fibrosis) aged 0 to 21 years were collected along with relevant clinical data. We measured total bacterial load using quantitative polymerase chain reaction and performed 16S rRNA gene sequencing to characterize bacterial communities with species-level sensitivity for select genera. Clinical comparisons were investigated. Measurements and main results Cystic fibrosis samples had higher total bacterial load and lower microbial diversity, with a divergence from disease controls around 2–5 years of age, as well as higher neutrophilic inflammation relative to bacterial burden. Cystic fibrosis samples had increased abundance of traditional cystic fibrosis pathogens and decreased abundance of the Streptococcus mitis species group in older subjects. Interestingly, increased diversity in the heterogeneous disease controls was independent of diagnosis and indication. Sequencing was more sensitive than culture, and antibiotic exposure was more common in disease controls, which showed a negative relationship with load and neutrophilic inflammation. Conclusions Analysis of lower airway samples from people with cystic fibrosis and disease controls across the ages revealed key differences in airway microbiota and inflammation. The divergence in subjects during early childhood may represent a window of opportunity for intervention and additional study.
Collapse
Affiliation(s)
- John B. O’Connor
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, Illinois, United States of America
- * E-mail:
| | - Madison M. Mottlowitz
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, Illinois, United States of America
| | - Brandie D. Wagner
- Department of Pediatrics, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, United States of America
- Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Kathleen L. Boyne
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, Illinois, United States of America
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
- Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Mark J. Stevens
- Department of Pediatrics, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Charles E. Robertson
- Department of Pediatrics, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Jonathan K. Harris
- Department of Pediatrics, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Theresa A. Laguna
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, Illinois, United States of America
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
- Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| |
Collapse
|
128
|
Abstract
Cystic fibrosis (CF) is a heritable, multiorgan disease that impacts all tissues that normally express cystic fibrosis transmembrane conductance regulator (CFTR) protein. While the importance of the airway microbiota has long been recognized, the intestinal microbiota has only recently been recognized as an important player in both intestinal and lung health outcomes for persons with CF (pwCF). Here, we summarize current literature related to the gut-lung axis in CF, with a particular focus on three key ideas: (i) mechanisms through which microbes influence the gut-lung axis, (ii) drivers of microbiota alterations, and (iii) the potential for intestinal microbiota remediation.
Collapse
Affiliation(s)
- Courtney E. Price
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover New Hampshire, USA
| | - George A. O’Toole
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover New Hampshire, USA
| |
Collapse
|
129
|
Elborn JS, Konstan MW, Taylor-Cousar JL, Fajac I, Horsley A, Sutharsan S, Aaron SD, Daines CL, Uluer A, Downey DG, Lucidi VV, Ahuja S, Springman E, Mershon J, Grosswald R, Rowe SM. Empire-CF study: A phase 2 clinical trial of leukotriene A4 hydrolase inhibitor acebilustat in adult subjects with cystic fibrosis. J Cyst Fibros 2021; 20:1026-1034. [PMID: 34538755 DOI: 10.1016/j.jcf.2021.08.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 08/15/2021] [Accepted: 08/16/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND Cystic fibrosis (CF) is characterized by neutrophilic inflammation in the airways. Leukotriene B4 (LTB4) is a neutrophil chemoattractant and has been implicated in CF pathogenesis. Acebilustat, a novel, synthetic, small-molecule leukotriene A4 hydrolase inhibitor, reduces LTB4 production. We report findings from a randomized placebo-controlled trial of acebilustat in adult subjects with mild-to-moderate lung disease. METHODS Subjects were randomized (1:1:1) to once-daily acebilustat 50 mg, 100 mg or placebo for 48 weeks, concomitantly with their current therapeutic regimen. Subjects were stratified by use of concomitant CF transmembrane conductance regulator (CFTR) modulators, baseline percent predicted forced expiratory volume in 1 second (ppFEV1) 50-75 and >75, and number of pulmonary exacerbations in the past year (1 or >1). Primary endpoints were the change from baseline in ppFEV1 and safety. Secondary endpoints included the rate of pulmonary exacerbations. RESULTS Overall, 199 subjects were randomized and dosed (acebilustat 50 mg, n=67; acebilustat 100 mg, n=66; placebo, n=66). Baseline demographics and disease profile were well balanced among treatment groups. Acebilustat had no statistically significant effect on the primary endpoint of change in ppFEV1 at week 48 or the secondary endpoint pulmonary exacerbations. There was a trend towards reduced pulmonary exacerbations in subjects receiving acebilustat in pre-specified populations with ppFEV1>75 (35% rate reduction) and those on concomitant CFTR modulator therapy (20% rate reduction). Acebilustat was well tolerated. CONCLUSIONS Acebilustat did not improve lung function. A trend towards reduced pulmonary exacerbations in subjects with an earlier stage of lung disease suggests a potential effect in this population.
Collapse
Affiliation(s)
- J Stuart Elborn
- Faculty of Medicine Health and Life sciences Queens University School of Medicine, Belfast, UK
| | - Michael W Konstan
- Department of Pediatrics, Case Western Reserve University School of Medicine and Rainbow Babies & Children's Hospital, Cleveland, OH, USA
| | | | - Isabelle Fajac
- Physiology Department, Cochin Hospital, APHP Centre, Paris, France; Université de Paris, Paris, France
| | - Alexander Horsley
- Division of Infection, Immunity and Respiratory Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Sivagurunathan Sutharsan
- Division of Cystic Fibrosis, Department of Pulmonary Medicine, University Medicine EssenRuhrlandklinik, University of DuisburgEssen, Essen, Germany
| | - Shawn D Aaron
- Ottawa Health Research Institute, Ottawa, Ontario, Canada
| | - Cori L Daines
- Department of Pediatrics, University of Arizona, Tucson, AZ, USA
| | - Ahmet Uluer
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Damian G Downey
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, UK
| | - Vincenzina V Lucidi
- Cystic Fibrosis Center, Children's Hospital and Research Institute, Bambino Gesu, Rome, Italy
| | | | | | | | | | - Steven M Rowe
- Gregory Fleming James Cystic Fibrosis Research Center and Department of Medicine, University of Alabama at Birmingham, 1918 University Blvd, MCLM 804, Birmingham 35294, AL, USA.
| | | |
Collapse
|
130
|
Impact of CFTR Therapy on Chronic Rhinosinusitis and Health Status: Deep Learning CT Analysis and Patient Reported Outcomes. Ann Am Thorac Soc 2021; 19:12-19. [PMID: 34436985 PMCID: PMC8787790 DOI: 10.1513/annalsats.202101-057oc] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
RATIONALE Elexacaftor/tezacaftor/ivacaftor (ETI) in triple combination improves pulmonary health for people with cystic fibrosis (PwCF) however its impact on objective measures of sinus disease and health utility is unestablished. OBJECTIVES To evaluate the impact of ETI on chronic rhinosinusitis (CRS) and general health status incorporating computed tomography (CT), quality-of-life (QOL) and productivity loss. METHODS Adult PwCF+CRS with CF transmembrane conductance regulator genotype F508del/F508del or F508del/minimal function who clinically initiated ETI participated in a prospective, observational study. The primary endpoint was change in percent sinus CT opacification (%SO) after 6 months of ETI assessed via deep learning-based methods. Secondary endpoints included changes in sinonasal QOL, health utility value and productivity loss, which were evaluated monthly via validated metrics. RESULTS 30 PwCF provided baseline data; 25 completed the study. At baseline, the cohort had substantial CRS, with mean 22-question SinoNasal Outcome Test (SNOT-22) score 33.1 and mean sinus CT %SO 63.7%. At 6-month follow-up, %SO improved by mean 22.9% (p<0.001). %SO improvement trended toward greater magnitude for those naïve to prior modulator therapy (p=0.09). Mean SNOT-22 scores and health utility improved by 15.3 and 0.068 [6.8%] (all p<0.007). Presenteeism, activity impairment and overall productivity loss improved (all p<0.049). Improvements in SNOT-22 scores and health utility occurred by one month and remained improved over the study. CONCLUSIONS ETI is associated with substantial improvements in sinus CT opacification and productivity loss, and clinically meaningful improvements in sinonasal QOL and health utility. Most improvements were rapid, robust and durable over the study.
Collapse
|
131
|
Bass R, Brownell JN, Stallings VA. The Impact of Highly Effective CFTR Modulators on Growth and Nutrition Status. Nutrients 2021; 13:2907. [PMID: 34578785 PMCID: PMC8470943 DOI: 10.3390/nu13092907] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 08/04/2021] [Accepted: 08/18/2021] [Indexed: 02/07/2023] Open
Abstract
Patients with cystic fibrosis (CF) are at increased risk of malnutrition and growth failure due to multiple factors as a result of suboptimal or absent function of the CFTR chloride channel protein. Dysfunctional CFTR contributes to increased energy expenditure, exocrine pancreatic insufficiency causing impaired dietary macronutrient digestion and absorption, intestinal dysbiosis, and impaired bile acid homeostasis. Poor nutritional status as a result of these mechanisms is associated with decreased lung function, worse clinical outcomes, and ultimately, increased mortality. Nutritional interventions addressing these mechanisms, such as pancreatic enzyme-replacement therapy and enteral caloric supplementation, have improved nutritional status and, by association, clinical outcomes. In the last decade, the advent of medications targeting defective CFTR proteins has revolutionized the care of patients with CF by reducing the overall impact of CFTR dysfunction. Below, we summarize the effects of highly effective CFTR modulators on nutritional status overall as well as specific factors including bile acid metabolism, pancreatic function, energy expenditure, and intestinal dysbiosis. The future of CF nutrition care will require a paradigm shift away from focusing on methods addressing CFTR dysfunction such as excess calorie provision and toward an individualized, holistic approach in the context of specific mutations and CFTR-directed therapy.
Collapse
Affiliation(s)
- Rosara Bass
- Children’s Hospital of Philadelphia, 3401 Civic Center Blvd, Philadelphia, PA 19104, USA
| | - Jefferson N. Brownell
- School of Medicine, University of Pennsylvania Perelman, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; (J.N.B.); (V.A.S.)
| | - Virginia A. Stallings
- School of Medicine, University of Pennsylvania Perelman, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; (J.N.B.); (V.A.S.)
| |
Collapse
|
132
|
Effects of Lumacaftor-Ivacaftor on Lung Clearance Index, Magnetic Resonance Imaging, and Airway Microbiome in Phe508del Homozygous Patients with Cystic Fibrosis. Ann Am Thorac Soc 2021; 18:971-980. [PMID: 33600745 DOI: 10.1513/annalsats.202008-1054oc] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Rationale: Previous studies showed that lumacaftor-ivacaftor therapy results in partial rescue of CFTR (cystic fibrosis [CF] transmembrane conductance regulator) activity and a moderate improvement of spirometry in Phe508del homozygous patients with CF. However, the effects of lumacaftor-ivacaftor on lung clearance index (LCI), lung morphology and perfusion detected by chest magnetic resonance imaging (MRI), and effects on the airway microbiome and inflammation remain unknown. Objectives: To investigate the effects of lumacaftor-ivacaftor on LCI, lung MRI scores, and airway microbiome and inflammation. Methods: In this prospective observational study we assessed clinical outcomes including spirometry and body mass index, LCI, lung MRI scores, sputum microbiome, and proinflammatory cytokines in 30 Phe508del homozygous patients with CF 12 years and older before and 8-16 weeks after initiation of lumacaftor-ivacaftor therapy. Results: Lumacaftor-ivacaftor had no effects on forced expiratory volume in 1 second (FEV1% predicted) (1.7%; 95% confidence interval [CI], -1.0% to 4.3%; P = 0.211) but improved LCI (-1.6; 95% CI, -2.6 to -0.5; P < 0.01) and MRI morphology (-1.3; 95% CI, -2.3 to -0.3; P < 0.05) and perfusion score (-1.2; 95% CI, -2.3 to -0.2; P < 0.05) in our study cohort. Furthermore, lumacaftor-ivacaftor decreased the total bacterial load (-1.8; 95% CI, -3.3 to -0.34; P < 0.05) and increased the Shannon diversity of the airway microbiome (0.4; 95% CI, 0.1 to 0.8; P < 0.05), and reduced IL-1β (interleukin-1β) concentration (median change, -324.2 pg/ml; 95% CI, -938.7 to 290.4 pg/ml; P < 0.05) in sputum of Phe508del homozygous patients. Conclusions: This study shows that lumacaftor-ivacaftor has beneficial effects on lung ventilation, morphology, and perfusion, as well as on the airway microbiome and inflammation in Phe508del homozygous patients. Our results suggest that LCI and MRI may be more sensitive than FEV1% predicted to detect response to CFTR modulator therapy in patients with chronic CF lung disease. Clinical trial registered with ClinicalTrials.gov (NCT02807415).
Collapse
|
133
|
Rehman T, Karp PH, Tan P, Goodell BJ, Pezzulo AA, Thurman AL, Thornell IM, Durfey SL, Duffey ME, Stoltz DA, McKone EF, Singh PK, Welsh MJ. Inflammatory cytokines TNF-α and IL-17 enhance the efficacy of cystic fibrosis transmembrane conductance regulator modulators. J Clin Invest 2021; 131:e150398. [PMID: 34166230 DOI: 10.1172/jci150398] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 06/22/2021] [Indexed: 12/12/2022] Open
Abstract
Without cystic fibrosis transmembrane conductance regulator-mediated (CFTR-mediated) HCO3- secretion, airway epithelia of newborns with cystic fibrosis (CF) produce an abnormally acidic airway surface liquid (ASL), and the decreased pH impairs respiratory host defenses. However, within a few months of birth, ASL pH increases to match that in non-CF airways. Although the physiological basis for the increase is unknown, this time course matches the development of inflammation in CF airways. To learn whether inflammation alters CF ASL pH, we treated CF epithelia with TNF-α and IL-17 (TNF-α+IL-17), 2 inflammatory cytokines that are elevated in CF airways. TNF-α+IL-17 markedly increased ASL pH by upregulating pendrin, an apical Cl-/HCO3- exchanger. Moreover, when CF epithelia were exposed to TNF-α+IL-17, clinically approved CFTR modulators further alkalinized ASL pH. As predicted by these results, in vivo data revealed a positive correlation between airway inflammation and CFTR modulator-induced improvement in lung function. These findings suggest that inflammation is a key regulator of HCO3- secretion in CF airways. Thus, they explain earlier observations that ASL pH increases after birth and indicate that, for similar levels of inflammation, the pH of CF ASL is abnormally acidic. These results also suggest that a non-cell-autonomous mechanism, airway inflammation, is an important determinant of the response to CFTR modulators.
Collapse
Affiliation(s)
- Tayyab Rehman
- Department of Internal Medicine and Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine and
| | - Philip H Karp
- Department of Internal Medicine and Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine and.,Howard Hughes Medical Institute, University of Iowa, Iowa City, Iowa, USA
| | - Ping Tan
- Department of Internal Medicine and Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine and
| | - Brian J Goodell
- Department of Internal Medicine and Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine and
| | - Alejandro A Pezzulo
- Department of Internal Medicine and Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine and
| | - Andrew L Thurman
- Department of Internal Medicine and Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine and
| | - Ian M Thornell
- Department of Internal Medicine and Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine and
| | - Samantha L Durfey
- Departments of Medicine and Microbiology, University of Washington, Seattle, Washington, USA
| | - Michael E Duffey
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York, USA
| | - David A Stoltz
- Department of Internal Medicine and Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine and.,Department of Molecular Physiology and Biophysics, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Edward F McKone
- National Referral Centre for Adult Cystic Fibrosis, St. Vincent's University Hospital and University College Dublin School of Medicine, Dublin, Ireland
| | - Pradeep K Singh
- Departments of Medicine and Microbiology, University of Washington, Seattle, Washington, USA
| | - Michael J Welsh
- Department of Internal Medicine and Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine and.,Howard Hughes Medical Institute, University of Iowa, Iowa City, Iowa, USA.,Department of Molecular Physiology and Biophysics, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
134
|
Clinical Effectiveness of Lumacaftor/Ivacaftor in Patients with Cystic Fibrosis Homozygous for F508del-CFTR. A Clinical Trial. Ann Am Thorac Soc 2021; 18:75-83. [PMID: 32644818 DOI: 10.1513/annalsats.202002-144oc] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Rationale: The combination of lumacaftor (LUM) and ivacaftor (IVA) is an approved CFTR (cystic fibrosis [CF] transmembrane conductance regulator) modulator treatment for homozygous F508del patients with CF.Objectives: To evaluate the effectiveness of LUM/IVA in children (6 yr or more) and adults (more than 18 yr) in a postapproval setting.Methods: This longitudinal cohort study, performed at 38 centers in the U.S. CF Therapeutics Development Network, enrolled homozygous F508del patients with CF ages 6 years old and older with no prior exposure to LUM/IVA. Study assessments were performed at baseline and at 1, 3, 6, and 12 months after LUM/IVA initiation.Results: A total of 193 patients initiated LUM/IVA, and 85% completed the study through 1 year. Baseline mean percent-predicted forced expiratory volume in 1 second (ppFEV1) was 85 (standard deviation, 22.4) in this cohort. No statistically significant change in ppFEV1 was observed from baseline to any of the follow-up time points, with a mean absolute change at 12 months of -0.3 (95% confidence interval [CI], -1.8 to 1.2). Body mass index improved from baseline to 12 months (mean change, 0.8 kg/m2; P < 0.001). Sweat chloride decreased from baseline to 1 month (mean change, -18.5 mmol/L; 95% CI, -20.7 to -16.3; P < 0.001), and these reductions were sustained through the study period. There were no significant changes in hospitalization rate for pulmonary exacerbations and Pseudomonas aeruginosa infection status with treatment.Conclusions: In this real-world multicenter cohort of children and adults, LUM/IVA treatment was associated with significant improvements in growth and reductions in sweat chloride without statistically significant or clinically meaningful changes in lung function, hospitalization rates, or P. aeruginosa infection.Clinical trial registered with www.clinicaltrials.gov (NCT02477319).
Collapse
|
135
|
Harwood KH, McQuade RM, Jarnicki A, Schneider-Futschik EK. Anti-Inflammatory Influences of Cystic Fibrosis Transmembrane Conductance Regulator Drugs on Lung Inflammation in Cystic Fibrosis. Int J Mol Sci 2021; 22:7606. [PMID: 34299226 PMCID: PMC8306345 DOI: 10.3390/ijms22147606] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/04/2021] [Accepted: 07/13/2021] [Indexed: 12/24/2022] Open
Abstract
Cystic fibrosis (CF) is caused by a defect in the cystic fibrosis transmembrane conductance regulator protein (CFTR) which instigates a myriad of respiratory complications including increased vulnerability to lung infections and lung inflammation. The extensive influx of pro-inflammatory cells and production of mediators into the CF lung leading to lung tissue damage and increased susceptibility to microbial infections, creates a highly inflammatory environment. The CF inflammation is particularly driven by neutrophil infiltration, through the IL-23/17 pathway, and function, through NE, NETosis, and NLRP3-inflammasome formation. Better understanding of these pathways may uncover untapped therapeutic targets, potentially reducing disease burden experienced by CF patients. This review outlines the dysregulated lung inflammatory response in CF, explores the current understanding of CFTR modulators on lung inflammation, and provides context for their potential use as therapeutics for CF. Finally, we discuss the determinants that need to be taken into consideration to understand the exaggerated inflammatory response in the CF lung.
Collapse
Affiliation(s)
- Kiera H. Harwood
- Department of Biochemistry & Pharmacology, Faculty of Medicine, Dentistry and Health Sciences, School of Biomedical Sciences, The University of Melbourne, Parkville, VIC 3010, Australia;
| | - Rachel M. McQuade
- Gut-Axis Injury and Repair Laboratory, Department of Medicine Western Health, Melbourne University, Melbourne, VIC 3021, Australia;
- The Florey Institute of Neuroscience and Mental Health, Parkville, VIC 3010, Australia
| | - Andrew Jarnicki
- Lung Disease Research Laboratory, Department of Biochemistry & Pharmacology, Melbourne University, Melbourne, VIC 3021, Australia
| | - Elena K. Schneider-Futschik
- Department of Biochemistry & Pharmacology, Faculty of Medicine, Dentistry and Health Sciences, School of Biomedical Sciences, The University of Melbourne, Parkville, VIC 3010, Australia;
| |
Collapse
|
136
|
Henen S, Denton C, Teckman J, Borowitz D, Patel D. Review of Gastrointestinal Motility in Cystic Fibrosis. J Cyst Fibros 2021; 20:578-585. [PMID: 34147362 DOI: 10.1016/j.jcf.2021.05.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 05/24/2021] [Accepted: 05/25/2021] [Indexed: 02/07/2023]
Abstract
Gastrointestinal manifestations in patients with cystic fibrosis (CF) are extremely common and have recently become a research focus. Gastrointestinal (GI) dysfunction is poorly understood in the CF population, despite many speculations including the role of luminal pH, bacterial overgrowth, and abnormal microbiome. Nevertheless, dysmotility is emerging as a possible key player in CF intestinal symptoms. Our review article aims to explore the sequelae of defective cystic fibrosis transmembrane conductance regulator (CFTR) genes on the GI tract as studied in both animals and humans, describe various presentations of intestinal dysmotility in CF, review newer diagnostic motility techniques including intraluminal manometry, and review the current literature regarding the potential role of dysmotility in CF-related intestinal pathologies.
Collapse
Affiliation(s)
- Sara Henen
- Saint Louis University School of Medicine, SSM Health Cardinal Glennon Children's Hospital, 1465 S Grand Blvd, St. Louis, MO 63104.
| | - Christine Denton
- Saint Louis University School of Medicine, SSM Health Cardinal Glennon Children's Hospital, 1465 S Grand Blvd, St. Louis, MO 63104
| | - Jeff Teckman
- Interim Chair, Department of Pediatrics, Professor of Pediatrics and Biochemistry, Saint Louis University School of Medicine, SSM Health Cardinal Glennon Children's Hospital, 1465 S Grand BLVD, St. Louis, MO 63104.
| | - Drucy Borowitz
- Emeritus Professor of Clinical Pediatrics, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, NY, 1001 Main Street, Buffalo, NY, 14203.
| | - Dhiren Patel
- Associate Professor and Medical Director, Neurogastroenterology and Motility Program, Department of Pediatrics, Saint Louis University School of Medicine, SSM Health Cardinal Glennon Children's Hospital, 1465 S Grand Blvd, St. Louis, MO 63104.
| |
Collapse
|
137
|
Systems Biology and Bile Acid Signalling in Microbiome-Host Interactions in the Cystic Fibrosis Lung. Antibiotics (Basel) 2021; 10:antibiotics10070766. [PMID: 34202495 PMCID: PMC8300688 DOI: 10.3390/antibiotics10070766] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 06/14/2021] [Accepted: 06/21/2021] [Indexed: 12/16/2022] Open
Abstract
The study of the respiratory microbiota has revealed that the lungs of healthy and diseased individuals harbour distinct microbial communities. Imbalances in these communities can contribute to the pathogenesis of lung disease. How these imbalances occur and establish is largely unknown. This review is focused on the genetically inherited condition of Cystic Fibrosis (CF). Understanding the microbial and host-related factors that govern the establishment of chronic CF lung inflammation and pathogen colonisation is essential. Specifically, dissecting the interplay in the inflammation–pathogen–host axis. Bile acids are important host derived and microbially modified signal molecules that have been detected in CF lungs. These bile acids are associated with inflammation and restructuring of the lung microbiota linked to chronicity. This community remodelling involves a switch in the lung microbiota from a high biodiversity/low pathogen state to a low biodiversity/pathogen-dominated state. Bile acids are particularly associated with the dominance of Proteobacterial pathogens. The ability of bile acids to impact directly on both the lung microbiota and the host response offers a unifying principle underpinning the pathogenesis of CF. The modulating role of bile acids in lung microbiota dysbiosis and inflammation could offer new potential targets for designing innovative therapeutic approaches for respiratory disease.
Collapse
|
138
|
Hubert D, Marguet C, Benichou J, DeSouza C, Payen-Champenois C, Kinnman N, Chandarana K, Munck A, Fajac I. Real-World Long-Term Ivacaftor for Cystic Fibrosis in France: Clinical Effectiveness and Healthcare Resource Utilization. Pulm Ther 2021; 7:455-468. [PMID: 34101145 PMCID: PMC8589905 DOI: 10.1007/s41030-021-00158-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 04/22/2021] [Indexed: 11/29/2022] Open
Abstract
Introduction Ivacaftor is a cystic fibrosis transmembrane conductance regulator (CFTR) potentiator that has demonstrated clinical benefits in phase 3 trials. We report results from a real-world study (BRIO) to assess the effectiveness of ivacaftor in people with cystic fibrosis (pwCF) in France. Methods BRIO was an observational study conducted at 35 centers in France. Both pwCF initiating ivacaftor treatment and those already taking ivacaftor were included and prospectively followed for 24 months. The primary objective was to evaluate the effect of ivacaftor on percent predicted forced expiratory volume in 1 s (ppFEV1); secondary objectives were evaluating the effect of ivacaftor on clinical effectiveness, healthcare resource utilization (HCRU), and safety. Results A total of 129 pwCF were enrolled; 58.9% were aged < 18 years; 64.3% had a G551D-CFTR allele. Mean age at ivacaftor initiation was 19.1 years (range, 2–64 years); ppFEV1 increased by a least squares mean of 8.49 percentage points in the first 6 months and was sustained through 36 months of ivacaftor use. Growth metrics increased during the first 12 months post-ivacaftor and remained stable. The rate of pulmonary exacerbations (PEx) decreased during the 12 months post-ivacaftor compared with the 12 months pre-ivacaftor; estimated rate ratios (95% CI) were 0.57 (0.43–0.75) for PEx events and 0.25 (0.13–0.48) for PEx requiring hospitalization. No new safety concerns were identified; no deaths occurred. Conclusions The results from this real-world study of ivacaftor usage in France were consistent with prior clinical trial outcomes, confirming the clinical effectiveness of ivacaftor, as well as an associated reduction in HCRU. Supplementary Information The online version contains supplementary material available at 10.1007/s41030-021-00158-5.
Collapse
Affiliation(s)
- Dominique Hubert
- Respiratory Medicine and National Cystic Fibrosis Reference Center, Cochin Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France.
| | - Christophe Marguet
- Pediatric Respiratory Diseases and Cystic Fibrosis Center, Rouen University Hospital, INSERM CIC1404, EA 2656 UNI ROUEN, Normandy University, Rouen, France
| | - Jacques Benichou
- Department of Biostatistics and Clinical Research, Rouen University Hospital and INSERM U 1018, University of Rouen, Rouen, France
| | | | | | - Nils Kinnman
- Vertex Pharmaceuticals (Europe) Limited, London, UK
| | | | - Anne Munck
- Cystic Fibrosis Center, Hôpital Robert Debré, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Isabelle Fajac
- Respiratory Medicine and National Cystic Fibrosis Reference Center, Cochin Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France.,Physiology Department, AP-HP Centre-Université de Paris, Hôpital Cochin, Paris, France.,Université de Paris, Paris, France
| | | |
Collapse
|
139
|
Bergeron C, Cantin AM. New Therapies to Correct the Cystic Fibrosis Basic Defect. Int J Mol Sci 2021; 22:ijms22126193. [PMID: 34201249 PMCID: PMC8227161 DOI: 10.3390/ijms22126193] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/31/2021] [Accepted: 06/04/2021] [Indexed: 12/24/2022] Open
Abstract
Rare diseases affect 400 million individuals worldwide and cause significant morbidity and mortality. Finding solutions for rare diseases can be very challenging for physicians and researchers. Cystic fibrosis (CF), a genetic, autosomal recessive, multisystemic, life-limiting disease does not escape this sad reality. Despite phenomenal progress in our understanding of this disease, treatment remains difficult. Until recently, therapies for CF individuals were focused on symptom management. The discovery of the cystic fibrosis transmembrane conductance regulator (CFTR) gene and its product, a protein present at the apical surface of epithelial cells regulating ion transport, allowed the scientific community to learn about the basic defect in CF and to study potential therapies targeting the dysfunctional protein. In the past few years, promising therapies with the goal to restore CFTR function became available and changed the lives of several CF patients. These medications, called CFTR modulators, aim to correct, potentialize, stabilize or amplify CFTR function. Furthermore, research is ongoing to develop other targeted therapies that could be more efficient and benefit a larger proportion of the CF community. The purpose of this review is to summarize our current knowledge of CF genetics and therapies restoring CFTR function, particularly CFTR modulators and gene therapy.
Collapse
Affiliation(s)
- Christelle Bergeron
- Department of Medicine, Respiratory Division, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, QC J1H 5N4, Canada;
| | - André M. Cantin
- Department of Medicine, Respiratory Division, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, QC J1H 5N4, Canada;
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Respiratory Division, Faculty of Medicine, University of Sherbrooke, 3001, 12e Avenue Nord, Sherbrooke, QC J1H 5N4, Canada
- Correspondence: ; Tel.: +1-819-346-1110 (ext. 14893); Fax: +1-819-564-5377
| |
Collapse
|
140
|
Shteinberg M, Haq IJ, Polineni D, Davies JC. Cystic fibrosis. Lancet 2021; 397:2195-2211. [PMID: 34090606 DOI: 10.1016/s0140-6736(20)32542-3] [Citation(s) in RCA: 392] [Impact Index Per Article: 98.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 10/03/2020] [Accepted: 11/20/2020] [Indexed: 12/11/2022]
Abstract
Cystic fibrosis is a monogenic disease considered to affect at least 100 000 people worldwide. Mutations in CFTR, the gene encoding the epithelial ion channel that normally transports chloride and bicarbonate, lead to impaired mucus hydration and clearance. Classical cystic fibrosis is thus characterised by chronic pulmonary infection and inflammation, pancreatic exocrine insufficiency, male infertility, and might include several comorbidities such as cystic fibrosis-related diabetes or cystic fibrosis liver disease. This autosomal recessive disease is diagnosed in many regions following newborn screening, whereas in other regions, diagnosis is based on a group of recognised multiorgan clinical manifestations, raised sweat chloride concentrations, or CFTR mutations. Disease that is less easily diagnosed, and in some cases affecting only one organ, can be seen in the context of gene variants leading to residual protein function. Management strategies, including augmenting mucociliary clearance and aggressively treating infections, have gradually improved life expectancy for people with cystic fibrosis. However, restoration of CFTR function via new small molecule modulator drugs is transforming the disease for many patients. Clinical trial pipelines are actively exploring many other approaches, which will be increasingly needed as survival improves and as the population of adults with cystic fibrosis increases. Here, we present the current understanding of CFTR mutations, protein function, and disease pathophysiology, consider strengths and limitations of current management strategies, and look to the future of multidisciplinary care for those with cystic fibrosis.
Collapse
Affiliation(s)
- Michal Shteinberg
- Pulmonology Institute and CF Center, Carmel Medical Center, Haifa, Israel; Rappaport Faculty of Medicine, The Technion-Israel Institute of Technology, Haifa, Israel
| | - Iram J Haq
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK; Great North Children's Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | | | - Jane C Davies
- National Heart and Lung Institute, Imperial College London, London, UK; Royal Brompton and Harefield, Guy's and St Thomas' NHS Foundation Trust, London, UK.
| |
Collapse
|
141
|
McNally P, Butler D, Karpievitch YV, Linnane B, Ranganathan S, Stick SM, Hall GL, Schultz A. Ivacaftor and Airway Inflammation in Preschool Children with Cystic Fibrosis. Am J Respir Crit Care Med 2021; 204:605-608. [PMID: 34077699 DOI: 10.1164/rccm.202012-4332le] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Paul McNally
- RCSI, 8863, Paediatrics, Dublin, Ireland.,Children's Health Ireland, 575376, Respiratory Medicine, Dublin, Ireland;
| | - Daryl Butler
- RCSI, 8863, Paediatrics, Dublin, Ireland.,Children's Health Ireland, 575376, Respiratory Medicine, Dublin, Ireland.,National Children's Research Centre, 549923, Dublin, Ireland
| | - Yuliya V Karpievitch
- Telethon Kids Institute, 117610, Respiratory Research Centre, West Perth, Western Australia, Australia
| | - Barry Linnane
- University of Limerick, 8808, School of Medicine and Centre for Interventions in Infection, Inflammation and Immunity (4i), Limerick, Ireland.,National Children's Research Centre, 549923, Dublin, Ireland
| | - Sarath Ranganathan
- The Royal Children's Hospital Melbourne, 6453, Department of Respiratory Medicine, Parkville, Victoria, Australia.,University of Melbourne, Department of Paediatrics, Parkville, Victoria, Australia.,Murdoch Childrens Research Institute, 34361, Parkville, Victoria, Australia
| | - Stephen M Stick
- Telethon Kids Institute, 117610, Wal-Yan Respiratory Research Centre, Nedlands, Australia.,Telethon Kids Institute, 117610, Northern Star Professor of Children's Respiratory Health Research, Nedlands, Australia.,Perth Children's Hospital, 60081, Department of Respiratory and Sleep Medicine, Nedlands, Australia
| | - Graham L Hall
- Telethon Kids Institute, 117610, Children's Lung Health, West Perth, Western Australia, Australia
| | - Andre Schultz
- Telethon Kids Institute, 117610, Wal-Yan Respiratory Research Centre, Perth, Western Australia, Australia.,Perth Children's Hospital, 60081, Department of Respiratory and Sleep Medicine, Nedlands, Western Australia, Australia.,The University of Western Australia, 2720, Division of Paediatrics, School of Medicine, Perth, Western Australia, Australia
| | | |
Collapse
|
142
|
Green M, Lindgren N, Henderson A, Keith JD, Oden AM, Birket SE. Ivacaftor partially corrects airway inflammation in a humanized G551D rat. Am J Physiol Lung Cell Mol Physiol 2021; 320:L1093-L1100. [PMID: 33825507 PMCID: PMC8285630 DOI: 10.1152/ajplung.00082.2021] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 03/22/2021] [Accepted: 04/04/2021] [Indexed: 01/09/2023] Open
Abstract
Animal models have been highly informative for understanding the pathogenesis and progression of cystic fibrosis (CF) lung disease. In particular, the CF rat models recently developed have addressed mechanistic causes of the airway mucus defect characteristic of CF, and how these may change when cystic fibrosis transmembrane conductance regulator (CFTR) activity is restored using new modulator therapies. We hypothesized that inflammatory changes to the airway would develop spontaneously and progressively, and that these changes would be resolved with modulator therapy. To test this, we used a humanized-CFTR rat expressing the G551D variant that responds to the CFTR modulator ivacaftor. Markers typically found in the CF lung were assessed, including neutrophil influx, small airway histopathology, and inflammatory cytokine concentration. Young hG551D rats did not express inflammatory cytokines at baseline but did upregulate these in response to inflammatory trigger. As the hG551D rats aged, histopathology worsened, accompanied by neutrophil influx into the airway and increasing concentrations of TNF-α, IL-1α, and IL-6 in the airways. Ivacaftor administration reduced concentrations of these cytokines when administered to the rats at baseline but was less effective in the rats that had also received inflammatory stimulus. Therefore, we conclude that administration of ivacaftor resulted in an incomplete resolution of inflammation when rats received an external trigger, suggesting that CFTR activation may not be enough to resolve inflammation in the lungs of patients with CF.
Collapse
Affiliation(s)
- Morgan Green
- Department of Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Natalie Lindgren
- Department of Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Alexander Henderson
- Department of Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Johnathan D Keith
- Department of Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Ashley M Oden
- Department of Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Susan E Birket
- Department of Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
143
|
Bene Z, Fejes Z, Szanto TG, Fenyvesi F, Váradi J, Clarke LA, Panyi G, Macek M, Amaral MD, Balogh I, Nagy B. Enhanced Expression of Human Epididymis Protein 4 (HE4) Reflecting Pro-Inflammatory Status Is Regulated by CFTR in Cystic Fibrosis Bronchial Epithelial Cells. Front Pharmacol 2021; 12:592184. [PMID: 34054511 PMCID: PMC8160512 DOI: 10.3389/fphar.2021.592184] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 04/16/2021] [Indexed: 12/18/2022] Open
Abstract
Decreased human epididymis protein 4 (HE4) plasma levels were reported in cystic fibrosis (CF) patients under CFTR potentiator ivacaftor therapy, which inversely correlated with lung function improvement. In this study, we investigated whether HE4 expression was affected via modulation of CFTR function in CF bronchial epithelial (CFBE) cells in vitro. HE4 protein levels were measured in the supernatants of CFBE 41o− cells expressing F508del-CFTR or wild-type CFTR (wt-CFTR) after administration of lumacaftor/ivacaftor or tezacaftor/ivacaftor, while HE4 expression in CFBE 41o− cells were also analyzed following application of adenylate cyclase activators Forskolin/IBMX or CFTRinh172. The effect of all of these compounds on CFTR function was monitored by the whole-cell patch-clamp technique. Induced HE4 expression was studied with interleukin-6 (IL-6) in F508del-CFTR CFBE 41o− cells under TNF-α stimulation for 1 h up to 1 week in duration. In parallel, plasma HE4 was determined in CF subjects homozygous for p.Phe508del-CFTR mutation receiving lumacaftor/ivacaftor (Orkambi®) therapy. NF-κB-mediated signaling was observed via the nuclear translocation of p65 subunit by fluorescence microscopy together with the analysis of IL-6 expression by an immunoassay. In addition, HE4 expression was examined after NF-κB pathway inhibitor BAY 11-7082 treatment with or without CFTR modulators. CFTR modulators partially restored the activity of F508del-CFTR and reduced HE4 concentration was found in F508del-CFTR CFBE 41o− cells that was close to what we observed in CFBE 41o− cells with wt-CFTR. These data were in agreement with decreased plasma HE4 concentrations in CF patients treated with Orkambi®. Furthermore, CFTR inhibitor induced elevated HE4 levels, while CFTR activator Forskolin/IBMX downregulated HE4 in the cell cultures and these effects were more pronounced in the presence of CFTR modulators. Higher activation level of baseline and TNF-α stimulated NF-κB pathway was detected in F508del-CFTR vs. wt-CFTR CFBE 41o− cells that was substantially reduced by CFTR modulators based on lower p65 nuclear positivity and IL-6 levels. Finally, HE4 expression was upregulated by TNF-α with elevated IL-6, and both protein levels were suppressed by combined administration of NF-κB pathway inhibitor and CFTR modulators in CFBE 41o− cells. In conclusion, CFTR dysfunction contributes to abnormal HE4 expression via NF-κB in CF.
Collapse
Affiliation(s)
- Zsolt Bene
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,Department of Pediatrics, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,Kálmán Laki Doctoral School of Biomedical and Clinical Sciences, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zsolt Fejes
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,Kálmán Laki Doctoral School of Biomedical and Clinical Sciences, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Tibor Gabor Szanto
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Ferenc Fenyvesi
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary
| | - Judit Váradi
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary
| | - Luka A Clarke
- Faculty of Sciences, BioISI-Biosystems and Integrative Sciences Institute, University of Lisboa, Lisboa, Portugal
| | - Gyorgy Panyi
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Milan Macek
- Department of Biology and Medical Genetics, Charles University-2nd Faculty of Medicine and Motol University Hospital, Prague, Czech
| | - Margarida D Amaral
- Faculty of Sciences, BioISI-Biosystems and Integrative Sciences Institute, University of Lisboa, Lisboa, Portugal
| | - István Balogh
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,Division of Clinical Genetics, Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Béla Nagy
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,Kálmán Laki Doctoral School of Biomedical and Clinical Sciences, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
144
|
Mainz JG, Arnold C, Wittstock K, Hipler UC, Lehmann T, Zagoya C, Duckstein F, Ellemunter H, Hentschel J. Ivacaftor Reduces Inflammatory Mediators in Upper Airway Lining Fluid From Cystic Fibrosis Patients With a G551D Mutation: Serial Non-Invasive Home-Based Collection of Upper Airway Lining Fluid. Front Immunol 2021; 12:642180. [PMID: 34025651 PMCID: PMC8131546 DOI: 10.3389/fimmu.2021.642180] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 04/19/2021] [Indexed: 11/24/2022] Open
Abstract
In cystic fibrosis (CF) therapy, the recent approval of CF-transmembrane conductance regulator (CFTR) channel modulators is considered to be the major breakthrough. However, the current first-line approach based mainly on pulmonary function to measure effects of the novel therapy, tested by forced expiratory volumes in one second (FEV1), provides restricted sensitivity to detect early structural damages. Accordingly, there is a need for new sensitive surrogate parameters. Most interestingly, these should quantify inflammation that precedes a decline of pulmonary function. We present a novel method assessing inflammatory markers in the upper airways’ epithelial lining fluid (ELF) obtained by nasal lavage (NL). In contrast to broncho-alveolar lavage, ELF sampling by NL is an attractive method due to its limited invasiveness which allows repeated analyses, even performed in a home-based setting. In a longitudinal cohort study (ClinicalTrials.gov, Identifier: NCT02311140), we assessed changes of inflammatory mediators in 259 serially obtained nasal lavages taken up to every second day before and during therapy with ivacaftor from ten CF patients carrying a G551D mutation. Patients were trained to sample NL-fluid at home, to immediately freeze and transfer chilled secretions to centers. Neutrophil Elastase, Interleukins IL-1β, IL-6 and IL-8 in NL were quantified. During 8-12 weeks of ivacaftor-treatment, median values of IL-1β and IL-6 significantly declined 2.29-fold (2.97→1.30 pg/mL), and 1.13-fold (6.48→5.72 pg/mL), respectively. In parallel, sweat tests and pulmonary function improved considerably. This is the first study assessing changes of airway inflammation on a day-to-day basis in CF patients receiving a newly administered CFTR-modulator therapy. It proves a decline of airway inflammation during ivacaftor-therapy.
Collapse
Affiliation(s)
- Jochen G Mainz
- Cystic Fibrosis Center, Brandenburg Medical School (MHB) University, Klinikum Westbrandenburg, Brandenburg an der Havel, Germany.,CF-Center, Jena University Hospital, Jena, Germany.,Faculty of Health Sciences, Joint Faculty of the Brandenburg University of Technology Cottbus -Senftenberg, The Brandenburg Medical School Theodor Fontane and the University of Potsdam, Cottbus, Brandenburg an der Havel and Potsdam, Germany
| | | | | | | | - Thomas Lehmann
- Jena University Hospital, Centre for Clinical Studies (Biometrics), Jena, Germany
| | - Carlos Zagoya
- Cystic Fibrosis Center, Brandenburg Medical School (MHB) University, Klinikum Westbrandenburg, Brandenburg an der Havel, Germany
| | - Franziska Duckstein
- Cystic Fibrosis Center, Brandenburg Medical School (MHB) University, Klinikum Westbrandenburg, Brandenburg an der Havel, Germany
| | | | - Julia Hentschel
- Institute of Human Genetics, Leipzig University Hospital, Leipzig, Germany
| |
Collapse
|
145
|
Long-Term Impact of Ivacaftor on Healthcare Resource Utilization Among People with Cystic Fibrosis in the United States. Pulm Ther 2021; 7:281-293. [PMID: 33913076 PMCID: PMC8137794 DOI: 10.1007/s41030-021-00154-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 03/19/2021] [Indexed: 12/18/2022] Open
Abstract
INTRODUCTION Ivacaftor was first approved in 2012 for the treatment of a select population of individuals with cystic fibrosis (CF), a rare, life-shortening genetic disease. Reductions in healthcare resource utilization (HCRU) associated with ivacaftor have been observed during limited follow-up and for selected outcomes in real-world studies. This study aimed to further describe the long-term impact of ivacaftor treatment on multiple measures of HCRU among people with CF (pwCF). METHODS This retrospective study used US commercial and Medicaid claims data from 2011-2018. We included pwCF ≥ 6 years of age with ≥ 1 claim for ivacaftor and 12 months of continuous health plan enrollment before ivacaftor initiation ("pre-ivacaftor" period) who also had 36 months of continuous enrollment and persistent ivacaftor use (i.e., no gap ≥ 90 days between refills) following initiation ("post-ivacaftor" period). We compared comorbidities occurring pre-ivacaftor versus the last 12 months post-ivacaftor. HCRU outcomes included medication use, inpatient admissions, and outpatient office visits. We compared medication use pre-ivacaftor versus the last 12 months post-ivacaftor and inpatient admissions and outpatient office visits pre-ivacaftor versus the post-ivacaftor period annualized across 36 months. RESULTS Seventy-nine pwCF met all criteria, including persistent ivacaftor use during the post-ivacaftor period. Ivacaftor treatment was associated with a significant reduction in pneumonia prevalence (10.1% vs. 26.6%; p < 0.001) and significantly fewer mean [SD] antibiotics claims (8.0 [7.3] vs. 12.3 [11.1]; p < 0.001) in the last 12 months post-ivacaftor versus pre-ivacaftor. In comparing the 36-month post-ivacaftor period to the pre-ivacaftor period, we also observed fewer mean [SD] annual inpatient admissions (0.2 [0.4] vs. 0.4 [0.7]), CF-related inpatient admissions (0.1 [0.2] vs. 0.2 [0.5]), and outpatient office visits (8.8 [4.9] vs. 9.9 [5.4]) (all, p < 0.05). CONCLUSION Long-term ivacaftor treatment reduced HCRU, consistent with trends observed in prior real-world studies. Our results support the sustained, long-term value of ivacaftor treatment in reducing CF burden.
Collapse
|
146
|
Kristensen MI, de Winter-de Groot KM, Berkers G, Chu MLJN, Arp K, Ghijsen S, Heijerman HGM, Arets HGM, Majoor CJ, Janssens HM, van der Meer R, Bogaert D, van der Ent CK. Individual and Group Response of Treatment with Ivacaftor on Airway and Gut Microbiota in People with CF and a S1251N Mutation. J Pers Med 2021; 11:jpm11050350. [PMID: 33925519 PMCID: PMC8146888 DOI: 10.3390/jpm11050350] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/15/2021] [Accepted: 04/25/2021] [Indexed: 12/19/2022] Open
Abstract
Ivacaftor has been shown to restore the functionality of the S1251N (also known as c.3752G>A) mutated CFTR, which may cause alterations in both airway and gut physiology and micro-environment, resulting in a change of microbiota in these organs. The aim of the present study was to analyze the effects of ivacaftor on the microbial community composition of both airway and gut in subjects with CF carrying one S1251N mutation, using a 16S rRNA gene-based sequencing approach. In 16 subjects with CF, repetitive samples from airways and gut were collected just before, and 2 months after, and, for 8 patients, also 9 and 12 months after, start of ivacaftor. 16S rRNA based sequencing identified 344 operational taxonomical units (OTUs) in a total of 139 samples (35 nasopharyngeal, 39 oropharyngeal, 29 sputum, and 36 fecal samples). Ivacaftor significantly enhanced bacterial diversity and overall microbiota composition in the gut (p < 0.01). There were no significant changes in the overall microbial composition and alpha diversity in upper and lower airways of these patients after ivacaftor treatment. Treatment with ivacaftor induces changes in gut microbiota whereas airway microbiota do not change significantly over time.
Collapse
Affiliation(s)
- Maartje I. Kristensen
- Department of Pediatric Pulmonology and Allergology, Wilhelmina Children’s Hospital—University Medical Center, Utrecht University, P.O. Box 85090, 3508 AB Utrecht, The Netherlands; (M.I.K.); (K.M.d.W.-d.G.); (G.B.); (S.G.); (H.G.M.A.); (C.K.v.d.E.)
| | - Karin M. de Winter-de Groot
- Department of Pediatric Pulmonology and Allergology, Wilhelmina Children’s Hospital—University Medical Center, Utrecht University, P.O. Box 85090, 3508 AB Utrecht, The Netherlands; (M.I.K.); (K.M.d.W.-d.G.); (G.B.); (S.G.); (H.G.M.A.); (C.K.v.d.E.)
| | - Gitte Berkers
- Department of Pediatric Pulmonology and Allergology, Wilhelmina Children’s Hospital—University Medical Center, Utrecht University, P.O. Box 85090, 3508 AB Utrecht, The Netherlands; (M.I.K.); (K.M.d.W.-d.G.); (G.B.); (S.G.); (H.G.M.A.); (C.K.v.d.E.)
| | - Mei Ling J. N. Chu
- Department of Pediatric Immunology and Infectious Diseases, Wilhelmina Children’s Hospital—University Medical Center, Utrecht University, P.O. Box 85090, 3508 AB Utrecht, The Netherlands; (M.L.J.N.C.); (K.A.)
| | - Kayleigh Arp
- Department of Pediatric Immunology and Infectious Diseases, Wilhelmina Children’s Hospital—University Medical Center, Utrecht University, P.O. Box 85090, 3508 AB Utrecht, The Netherlands; (M.L.J.N.C.); (K.A.)
| | - Sophie Ghijsen
- Department of Pediatric Pulmonology and Allergology, Wilhelmina Children’s Hospital—University Medical Center, Utrecht University, P.O. Box 85090, 3508 AB Utrecht, The Netherlands; (M.I.K.); (K.M.d.W.-d.G.); (G.B.); (S.G.); (H.G.M.A.); (C.K.v.d.E.)
| | - Harry G. M. Heijerman
- Department of Pulmonology, University Medical Center, Utrecht University, P.O. Box 85500, 3508 GA Utrecht, The Netherlands;
| | - Hubertus G. M. Arets
- Department of Pediatric Pulmonology and Allergology, Wilhelmina Children’s Hospital—University Medical Center, Utrecht University, P.O. Box 85090, 3508 AB Utrecht, The Netherlands; (M.I.K.); (K.M.d.W.-d.G.); (G.B.); (S.G.); (H.G.M.A.); (C.K.v.d.E.)
| | - Christof J. Majoor
- Department of Respiratory Medicine, Amsterdam University Medical Center, P.O. Box 22660, 1100 DD Amsterdam, The Netherlands;
| | - Hettie M. Janssens
- Department of Pediatric Pulmonology, Erasmus Medical Center/Sophia Children’s Hospital, 3015 GD Rotterdam, The Netherlands;
| | - Renske van der Meer
- Department of Pulmonology, Haga Teaching Hospital, 2545 AA The Hague, The Netherlands;
| | - Debby Bogaert
- Department of Pediatric Immunology and Infectious Diseases, Wilhelmina Children’s Hospital—University Medical Center, Utrecht University, P.O. Box 85090, 3508 AB Utrecht, The Netherlands; (M.L.J.N.C.); (K.A.)
- The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
- Correspondence:
| | - Cornelis K. van der Ent
- Department of Pediatric Pulmonology and Allergology, Wilhelmina Children’s Hospital—University Medical Center, Utrecht University, P.O. Box 85090, 3508 AB Utrecht, The Netherlands; (M.I.K.); (K.M.d.W.-d.G.); (G.B.); (S.G.); (H.G.M.A.); (C.K.v.d.E.)
| |
Collapse
|
147
|
Girón Moreno RM, García-Clemente M, Diab-Cáceres L, Martínez-Vergara A, Martínez-García MÁ, Gómez-Punter RM. Treatment of Pulmonary Disease of Cystic Fibrosis: A Comprehensive Review. Antibiotics (Basel) 2021; 10:486. [PMID: 33922413 PMCID: PMC8144952 DOI: 10.3390/antibiotics10050486] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 04/13/2021] [Accepted: 04/17/2021] [Indexed: 01/08/2023] Open
Abstract
Cystic fibrosis (CF) is a genetic disease that causes absence or dysfunction of a protein named transmembrane conductance regulatory protein (CFTR) that works as an anion channel. As a result, the secretions of the organs where CFTR is expressed are very viscous, so their functionality is altered. The main cause of morbidity is due to the involvement of the respiratory system as a result of recurrent respiratory infections by different pathogens. In recent decades, survival has been increasing, rising by around age 50. This is due to the monitoring of patients in multidisciplinary units, early diagnosis with neonatal screening, and advances in treatments. In this chapter, we will approach the different therapies used in CF for the treatment of symptoms, obstruction, inflammation, and infection. Moreover, we will discuss specific and personalized treatments to correct the defective gene and repair the altered protein CFTR. The obstacle for personalized CF treatment is to predict the drug response of patients due to genetic complexity and heterogeneity of uncommon mutations.
Collapse
Affiliation(s)
- Rosa María Girón Moreno
- Servicio de Neumología, Instituto de Investigación Sanitaria La Princesa, 28006 Madrid, Spain; (R.M.G.M.); (R.M.G.-P.)
| | - Marta García-Clemente
- Servicio de Neumología, Hospital Universitario Central de Asturias, C/Avenida de Roma S/n, 33011 Oviedo, Spain
| | - Layla Diab-Cáceres
- Servicio de Neumología, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain;
| | | | | | - Rosa Mar Gómez-Punter
- Servicio de Neumología, Instituto de Investigación Sanitaria La Princesa, 28006 Madrid, Spain; (R.M.G.M.); (R.M.G.-P.)
| |
Collapse
|
148
|
Künzi L, Easter M, Hirsch MJ, Krick S. Cystic Fibrosis Lung Disease in the Aging Population. Front Pharmacol 2021; 12:601438. [PMID: 33935699 PMCID: PMC8082404 DOI: 10.3389/fphar.2021.601438] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 03/15/2021] [Indexed: 01/02/2023] Open
Abstract
The demographics of the population with cystic fibrosis (CF) is continuously changing, with nowadays adults outnumbering children and a median predicted survival of over 40 years. This leads to the challenge of treating an aging CF population, while previous research has largely focused on pediatric and adolescent patients. Chronic inflammation is not only a hallmark of CF lung disease, but also of the aging process. However, very little is known about the effects of an accelerated aging pathology in CF lungs. Several chronic lung disease pathologies show signs of chronic inflammation with accelerated aging, also termed “inflammaging”; the most notable being chronic obstructive pulmonary disease (COPD) and idiopathic pulmonary fibrosis (IPF). In these disease entities, accelerated aging has been implicated in the pathogenesis via interference with tissue repair mechanisms, alterations of the immune system leading to impaired defense against pulmonary infections and induction of a chronic pro-inflammatory state. In addition, CF lungs have been shown to exhibit increased expression of senescence markers. Sustained airway inflammation also leads to the degradation and increased turnover of cystic fibrosis transmembrane regulator (CFTR). This further reduces CFTR function and may prevent the novel CFTR modulator therapies from developing their full efficacy. Therefore, novel therapies targeting aging processes in CF lungs could be promising. This review summarizes the current research on CF in an aging population focusing on accelerated aging in the context of chronic airway inflammation and therapy implications.
Collapse
Affiliation(s)
- Lisa Künzi
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL, United States.,Epidemiology, Biostatistics and Prevention Institute, Department of Public and Global Health, University of Zürich, Zürich, Switzerland
| | - Molly Easter
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Meghan June Hirsch
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Stefanie Krick
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL, United States.,Gregory Fleming Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, AL, United States.,Comprehensive Center for Healthy Aging, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
149
|
Duckers J, Lesher B, Thorat T, Lucas E, McGarry LJ, Chandarana K, De Iorio F. Real-World Outcomes of Ivacaftor Treatment in People with Cystic Fibrosis: A Systematic Review. J Clin Med 2021; 10:1527. [PMID: 33917386 PMCID: PMC8038673 DOI: 10.3390/jcm10071527] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/24/2021] [Accepted: 03/26/2021] [Indexed: 12/20/2022] Open
Abstract
Cystic fibrosis (CF) is a rare, progressive, multi-organ genetic disease. Ivacaftor, a small-molecule CF transmembrane conductance regulator modulator, was the first medication to treat the underlying cause of CF. Since its approval, real-world clinical experience on the use of ivacaftor has been documented in large registries and smaller studies. Here, we systematically review data from real-world observational studies of ivacaftor treatment in people with CF (pwCF). Searches of MEDLINE and Embase identified 368 publications reporting real-world studies that enrolled six or more pwCF treated with ivacaftor published between January 2012 and September 2019. Overall, 75 publications providing data from 57 unique studies met inclusion criteria and were reviewed. Studies reporting within-group change for pwCF treated with ivacaftor consistently showed improvements in lung function, nutritional parameters, and patient-reported respiratory and sino-nasal symptoms. Benefits were evident as early as 1 month following ivacaftor initiation and were sustained over long-term follow-up. Decreases in pulmonary exacerbations, Pseudomonas aeruginosa prevalence, and healthcare resource utilization also were reported for up to 66 months following ivacaftor initiation. In studies comparing ivacaftor treatment to modulator untreated comparator groups, clinical benefits similarly were reported as were decreases in mortality, organ-transplantation, and CF-related complications. The safety profile of ivacaftor observed in these real-world studies was consistent with the well-established safety profile based on clinical trial data. Our systematic review of real-world studies shows ivacaftor treatment in pwCF results in highly consistent and sustained clinical benefit in both pulmonary and non-pulmonary outcomes across various geographies, study designs, patient characteristics, and follow-up durations, confirming and expanding upon evidence from clinical trials.
Collapse
Affiliation(s)
- Jamie Duckers
- All Wales Adult Cystic Fibrosis Centre, University Hospital Llandough, Cardiff CF64 2XX, UK
| | - Beth Lesher
- Pharmerit—An OPEN Health Company, 4350 East-West Highway, Suite 1100, Bethesda, MD 20814, USA; (B.L.); (E.L.)
| | - Teja Thorat
- Vertex Pharmaceuticals Incorporated, Boston, MA 02210, USA; (T.T.); (L.J.M.); (K.C.)
| | - Eleanor Lucas
- Pharmerit—An OPEN Health Company, 4350 East-West Highway, Suite 1100, Bethesda, MD 20814, USA; (B.L.); (E.L.)
| | - Lisa J. McGarry
- Vertex Pharmaceuticals Incorporated, Boston, MA 02210, USA; (T.T.); (L.J.M.); (K.C.)
| | - Keval Chandarana
- Vertex Pharmaceuticals Incorporated, Boston, MA 02210, USA; (T.T.); (L.J.M.); (K.C.)
| | - Fosca De Iorio
- Vertex Pharmaceuticals (Europe) Limited, London W2 6BD, UK;
| |
Collapse
|
150
|
Treggiari D, Kleinfelder K, Bertini M, Tridello G, Fedrigo A, Pintani E, Iansa P, Casiraghi A, Minghetti P, Cipolli M, Sorio C, Melotti P. Optical Measurements of Sweat for in Vivo Quantification of CFTR Function in Individual Sweat Glands. J Cyst Fibros 2021; 20:824-827. [PMID: 33814321 DOI: 10.1016/j.jcf.2021.03.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 02/01/2021] [Accepted: 03/06/2021] [Indexed: 11/25/2022]
Abstract
Optical measurement of CFTR-dependent sweat secretion stimulated by a beta-adrenergic cocktail (C-phase) vs. CFTR-independent sweat secretion induced by methacholine (M-phase) can discriminate cystic fibrosis (CF) patientts from controls and healthy carriers by the ratio of sweat rate in the C-phase vs. the M-phase (C/M ratio). However, image analysis is experimentally demanding and time-consuming. Here, sweat droplet number (SDN) in the C-phase, corresponding to the number of sweat-secreting glands, was a statistically significant predictor for detecting the effects of CFTR-targeted therapy. We show that in 44 non-CF subjects and 110 CF patients, SDN in the C-phase provides a linear readout of CFTR function that is more sensitive than that using the C/M ratio. In CF patients, increased SDN in the C-phase during treatment with (LUMA/IVA) was associated with a trend toward improved lung function (FEV1). Our method is suitable for multicenter monitoring of the effects of CFTR modulators.
Collapse
Affiliation(s)
- Davide Treggiari
- Cystic Fibrosis Centre, Azienda Ospedaliera Universitaria Integrata Verona, piazzale Stefani, 1, 37126 Verona, Italy.
| | - Karina Kleinfelder
- Department of Medicine, University of Verona, Division of General Pathology, Strada Le Grazie 8, 37134 Verona, Italy.
| | - Marina Bertini
- Cystic Fibrosis Centre, Azienda Ospedaliera Universitaria Integrata Verona, piazzale Stefani, 1, 37126 Verona, Italy.
| | - Gloria Tridello
- Cystic Fibrosis Centre, Azienda Ospedaliera Universitaria Integrata Verona, piazzale Stefani, 1, 37126 Verona, Italy.
| | - Arianna Fedrigo
- Department of Medicine, University of Verona, Division of General Pathology, Strada Le Grazie 8, 37134 Verona, Italy.
| | - Emily Pintani
- Cystic Fibrosis Centre, Azienda Ospedaliera Universitaria Integrata Verona, piazzale Stefani, 1, 37126 Verona, Italy.
| | - Patrizia Iansa
- Cystic Fibrosis Centre, Azienda Ospedaliera Universitaria Integrata Verona, piazzale Stefani, 1, 37126 Verona, Italy.
| | - Antonella Casiraghi
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, via G. Colombo 71, Milan, 20133, Italy.
| | - Paola Minghetti
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, via G. Colombo 71, Milan, 20133, Italy.
| | - Marco Cipolli
- Cystic Fibrosis Centre, Azienda Ospedaliera Universitaria Integrata Verona, piazzale Stefani, 1, 37126 Verona, Italy.
| | - Claudio Sorio
- Department of Medicine, University of Verona, Division of General Pathology, Strada Le Grazie 8, 37134 Verona, Italy.
| | - Paola Melotti
- Cystic Fibrosis Centre, Azienda Ospedaliera Universitaria Integrata Verona, piazzale Stefani, 1, 37126 Verona, Italy.
| |
Collapse
|