231 results on '"Konstan MW"'
Search Results
2. Amikacin liposome inhalation suspension for chronic Pseudomonas aeruginosa infection in cystic fibrosis
- Author
-
Bilton D, Pressler T, Fajac I, Clancy JP, Sands D, Minic P, Cipolli M, Galeva I, Solé A, Quittner AL, Liu K, McGinnis JP, Eagle G, Gupta R, Konstan MW, and CLEAR-108 Study Group
- Subjects
ALIS, Amikacin liposome inhalation suspension, CFQ-R, Cystic fibrosis, LAI, Liposomal amikacin for inhalation, Pseudomonas aeruginosa ,respiratory tract diseases - Abstract
Shortcomings of inhaled antibiotic treatments for Pseudomonas aeruginosa infection in patients with cystic fibrosis (CF) include poor drug penetration, inactivation by sputum, poor efficiency due to protective biofilm, and short residence in the lung.
- Published
- 2020
3. P255 Rate of lung function decline in patients with cystic fibrosis (cf) having a residual function gene mutation
- Author
-
Sawicki, G, primary, Konstan, MW, additional, McKone, E, additional, Moss, RB, additional, Lubarsky, B, additional, Suthoff, E, additional, Millar, S, additional, Pasta, DJ, additional, Mayer-Hamblett, N, additional, Goss, CH, additional, and Morgan, W, additional
- Published
- 2017
- Full Text
- View/download PDF
4. Tobramycin Inhalation Powder Is Effective and Safe in the Treatment of Chronic PulmonaryPseudomonas aeruginosa (Pa)Infection in Patients with Cystic Fibrosis.
- Author
-
Konstan, MW, primary, Geller, DE, additional, Brockhaus, F, additional, Zhang, J, additional, and Angyalosi, G, additional
- Published
- 2009
- Full Text
- View/download PDF
5. Ibuprofen therapy for cystic fibrosis lung disease: revisited.
- Author
-
Konstan MW
- Published
- 2008
- Full Text
- View/download PDF
6. Clinical use of Ibuprofen is associated with slower FEV1 decline in children with cystic fibrosis.
- Author
-
Konstan MW, Schluchter MD, Xue W, Davis PB, Konstan, Michael W, Schluchter, Mark D, Xue, Wei, and Davis, Pamela B
- Abstract
Rationale: High-dose ibuprofen in a 4-year controlled trial slowed FEV(1) decline in young subjects with cystic fibrosis, but the effectiveness of ibuprofen has not been assessed in a large group of patients treated clinically with this therapy.Objectives: To assess the effect of ibuprofen therapy on FEV(1) decline in children and adolescents with cystic fibrosis, using observational data from the Cystic Fibrosis Foundation Patient Registry.Methods: The rate of decline in FEV(1) percent predicted over 2-7 years among patients age 6-17 years with FEV(1) > 60% predicted, and who were treated with ibuprofen (1,365), was compared with patients of similar age and disease severity who were not treated with this therapy (8,960). Multilevel repeated-measures mixed-regression models were used to estimate rates of decline, adjusting for characteristics and therapies that influenced FEV(1) decline. Adverse effects were compared among those treated versus not treated with ibuprofen.Measurements and Main Results: FEV(1) declined less rapidly among patients treated with ibuprofen (difference, 0.60% predicted per year; 95% confidence interval, 0.31 to 0.89; P < 0.0001); a 29% reduction in slope based on an average decline of 2.08% predicted per year for patients not treated. Those treated with ibuprofen were more likely to have an episode of gastrointestinal bleeding requiring hospitalization, but the occurrence was rare in both groups (annual incidence, 0.37 vs. 0.14%; relative risk, 2.72; P < 0.001).Conclusions: Slower rates of FEV(1) decline are seen in children and adolescents with cystic fibrosis who are treated with ibuprofen. The apparent benefits of ibuprofen therapy outweigh the small risk of gastrointestinal bleeding. [ABSTRACT FROM AUTHOR]- Published
- 2007
- Full Text
- View/download PDF
7. Classifying severity of cystic fibrosis lung disease using longitudinal pulmonary function data.
- Author
-
Schluchter MD, Konstan MW, Drumm ML, Yankaskas JR, Knowles MR, Schluchter, Mark D, Konstan, Michael W, Drumm, Mitchell L, Yankaskas, James R, and Knowles, Michael R
- Abstract
Rationale: The study of genetic modifiers in cystic fibrosis (CF) lung disease requires rigorous phenotyping. One type of genetic association study design compares polymorphisms in patients at extremes of phenotype, requiring accurate classification of pulmonary disease at varying ages.Objective: To evaluate approaches to quantify severity of pulmonary disease and their ability to discriminate between patients with CF at the extremes of phenotype.Methods: DeltaF508 homozygotes (n = 828) were initially classified as "severe" (approximate lowest quartile of FEV(1) (% pred) for age, 8-25 yr) or "mild" disease (highest quartile of FEV(1) for age, > or = 15 yr). FEV(1) measurements from the 5 yr before enrollment (total = 18,501 measurements; average 23 per subject) were analyzed with mixed models, and patient-specific estimates of FEV(1) (% pred) at ages 5, 10, 15, 20, and 25 yr and slope of FEV(1) versus age were examined for their ability to discriminate between groups using receiver operating characteristics (ROC) curve areas.Results: Logistic regression of severity group on mixed model (empirical Bayes) estimates of intercept and slope of FEV(1) (% pred) versus age discriminated better than did classification using FEV(1) slope alone (ROC area = 0.995 vs. 0.821) and was equivalent to using estimated FEV(1) at 20 yr of age as a single discriminator. The estimated survival percentile from a joint survival/longitudinal model provided equally good classification (ROC area = 0.994).Conclusions: In CF, estimated FEV(1) (% pred) at 20 yr of age and the estimated survival percentile are useful indices of pulmonary disease severity. [ABSTRACT FROM AUTHOR]- Published
- 2006
- Full Text
- View/download PDF
8. Genetic modifiers of lung disease in cystic fibrosis.
- Author
-
Drumm ML, Konstan MW, Schluchter MD, Handler A, Pace R, Zou F, Zariwala M, Fargo D, Xu A, Dunn JM, Darrah RJ, Dorfman R, Sandford AJ, Corey M, Zielenski J, Durie P, Goddard K, Yankaskas JR, Wright FA, and Knowles MR
- Published
- 2005
9. Significant microbiological effect of inhaled tobramycin in young children with cystic fibrosis.
- Author
-
Gibson RL, Emerson J, McNamara S, Burns JL, Rosenfeld M, Yunker A, Hamblett N, Accurso F, Dovey M, Hiatt P, Konstan MW, Moss R, Retsch-Bogart G, Wagener J, Waltz D, Wilmott R, Zeitlin PL, Ramsey B, and Cystic Fibrosis Therapeutics Development Network Study Group
- Abstract
We conducted a double-blind, placebo-controlled, multicenter, randomized trial to test the hypothesis that 300 mg of tobramycin solution for inhalation administered twice daily for 28 days would be safe and result in a profound decrease in Pseudomonas aeruginosa (Pa) density from the lower airway of young children with cystic fibrosis. Ninety-eight subjects were to be randomized; however, the trial was stopped early because of evidence of a significant microbiological treatment effect. Twenty-one children under age 6 years were randomized (8 active; 13 placebo) and underwent bronchoalveolar lavage at baseline and on Day 28. There was a significant difference between treatment groups in the reduction in Pa density; no Pa was detected on Day 28 in 8 of 8 active group patients compared with 1 of 13 placebo group patients. We observed no differences between treatment groups for clinical indices, markers of inflammation, or incidence of adverse events. No abnormalities in serum creatinine or audiometry and no episodes of significant bronchospasm were observed in association with active treatment. We conclude that 28 days of tobramycin solution for inhalation of 300 mg twice daily is safe and effective for significant reduction of lower airway Pa density in young children with cystic fibrosis. [ABSTRACT FROM AUTHOR]
- Published
- 2003
- Full Text
- View/download PDF
10. Factors influencing outcomes in cystic fibrosis: a center-based analysis.
- Author
-
Johnson C, Butler SM, Konstan MW, Morgan W, Wohl MEB, Scientific Advisory Group and the Investigators of the Epidemiologic Study of Cystic Fibrosis, Johnson, Charles, Butler, Steven M, Konstan, Michael W, Morgan, Wayne, and Wohl, Mary Ellen B
- Abstract
Context: Guidelines for managing cystic fibrosis (CF) patients have been widely circulated, but little is known about the variations in practice between sites and their association with outcomes.Objective: To determine whether differences in lung health existed between groups of patients attending different CF care sites and to determine whether these differences are associated with differences in monitoring and intervention.Design: The analysis was conducted using data from the Epidemiologic Study of Cystic Fibrosis from 1995 through 1996.Setting: This was an observational database collecting prospective information from a large number of CF patients undergoing routine care in North America.Participants: Participating sites that had at least 50 CF patients who had each made at least one visit to a center during the 2-year study period were ranked on the basis of median values for FEV(1) within each of three age groups (6 to 12 years, 13 to 17 years, and >or= 18 years).Interventions: There were no prespecified interventions in this observational study.Main Outcome Measures: The frequency of patient monitoring and the use of therapeutic interventions were compared between sites in the upper and lower quartiles after stratification within the site for disease severity.Results: Within-site rankings tended to be consistent across the three age groups. Patients who were treated at higher ranking sites had more frequent monitoring of their clinical status, measurements of lung function, and cultures for respiratory pathogens. These patients also received more interventions, particularly IV antibiotics for pulmonary exacerbations.Conclusion: We found substantial differences in lung health across different CF care sites. We found that frequent monitoring and increased use of appropriate medications in the management of CF are associated with improved outcomes. [ABSTRACT FROM AUTHOR]- Published
- 2003
- Full Text
- View/download PDF
11. Effect of high-dose ibuprofen in patients with cystic fibrosis.
- Author
-
Konstan MW, Byard PJ, Hoppel CL, and Davis PB
- Published
- 1995
12. Rapid intravenous desensitization to colistin.
- Author
-
Tosi MF, Konstan MW, and Paschall VL
- Published
- 2012
- Full Text
- View/download PDF
13. P255 Rate of lung function decline in patients with cystic fibrosis (cf) having a residual function gene mutation
- Author
-
Sawicki, G, Konstan, MW, McKone, E, Moss, RB, Lubarsky, B, Suthoff, E, Millar, S, Pasta, DJ, Mayer-Hamblett, N, Goss, CH, and Morgan, W
- Abstract
ObjectivePatients with cystic fibrosis (CF) and mutations associated with residual CFTR chloride transport have improved survival rates compared with those homozygous for the F508del-CFTRmutation. Since little is known about rate of lung function decline in patients with CF and residual function (RF) mutations, we evaluated differences in rates of percent predicted FEV1(ppFEV1) decline between patients with an RF mutation who were heterozygous for F508deland those who were homozygous for F508del, and whether rates of ppFEV1decline differed across age groups.MethodsPatients in the US CF Foundation Patient Registry from 2006 to 2014 with an RF mutation heterozygous for F508delwere compared with F508del-homozygous patients. Mutations were identified based on clinical or in vitroevidence of residual ion transport. Annual rates of ppFEV1decline were estimated for patients 6 to 45 years of age with ≥3 ppFEV1values spanning ≥0.5 years in a randomly chosen 2 year period that began at the first ppFEV1measurement in the calendar year.ResultsA total of 1242 RF and 11,916 F508del-homozygous patients were included. At the first visit, the RF cohort was older (mean [SD], 23.0 [12.1] vs 18.0 [9.6] years) and had better nutritional status (mean [SD] BMI zscore, 0.36 [1.09] vs −0.29 [1.08]). Mean (SD) ppFEV1differed at the first visit between cohorts (80.4 [24.8] vs 73.4 [26.5]; p<0.001). Annual rate of ppFEV1decline was estimated at −0.70 (SE, 0.20) in the RF cohort compared with −1.91 (0.05) in the F508del-homozygous cohort (p<0.001). After excluding patients with R117H(n=889), the rate of decline was −1.05 (0.39) ppFEV1per year (p<0.001 vs F508del). The rate of decline for RF patients was most rapid in the 18 to 24–year age group, −1.38 (0.39), but was still significantly less than the −2.52 (0.09) for F508del-homozygous young adults (p=0.004).ConclusionPatients with CF and an RF mutation have lower rates of lung function decline compared with F508del-homozygous patients. However, patients with an RF mutation still demonstrate progressive lung disease, particularly during young adulthood.Please refer to page A259 for declarations of interest in relation to abstract P255.
- Published
- 2017
- Full Text
- View/download PDF
14. Infant care patterns at epidemiologic study of cystic fibrosis sites that achieve superior childhood lung function.
- Author
-
Padman R, McColley SA, Miller DP, Konstan MW, Morgan WJ, Schechter MS, Ren CL, Wagener JS, and Investigators and Coordinators of the Epidemiologic Study of Cystic Fibrosis
- Published
- 2007
- Full Text
- View/download PDF
15. Case Report: A delicate equilibrium of exocrine pancreatic recovery and hepatotoxicity with elexacaftor/tezacaftor/ivacaftor therapy in a pediatric patient with cystic fibrosis.
- Author
-
Coughlin MP, Sankararaman S, Roesch EA, Certo ED, Brej BL, and Konstan MW
- Abstract
This case report presents a comprehensive evaluation of the complex balance of therapeutic benefits and potential risks associated with the cystic fibrosis transmembrane conductance regulator (CFTR) modulator elexacaftor/tezacaftor/ivacaftor (ETI) therapy in managing an eight-year-old male with cystic fibrosis (CF) and exocrine pancreatic insufficiency (EPI). While ETI therapy significantly enhanced exocrine pancreatic function, it led to hepatotoxicity, necessitating therapy discontinuation. Attempts to restart ETI at reduced doses were unsuccessful due to persistent hepatic dysfunction. Reduced ETI dosing frequency, implemented due to hepatic dysfunctions, did not result in substantial therapeutic benefits. Clinical markers showed a resurgence of severe EPI and sustained need for gastrostomy tube feeds, with only modest improvement in hepatic function compared to the period following ETI cessation or during prior use of CFTR modulator therapy with lumacaftor/ivacaftor. This case underscores the importance of personalized therapeutic approaches, biomarker-guided monitoring, and multidisciplinary insights to optimize CF management while also highlighting the ongoing need for research to mitigate hepatotoxicity risks and ensure long-term therapeutic efficacy., Competing Interests: MK and ER have served as consultants to Vertex Pharmaceuticals. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (© 2024 Coughlin, Sankararaman, Roesch, Certo, Brej and Konstan.)
- Published
- 2024
- Full Text
- View/download PDF
16. Equity-Focused Evaluation of a Medicaid-Funded Statewide Diabetes Quality Improvement Project Collaborative.
- Author
-
Joseph JJ, Perzynski AT, Dungan KM, Beverly EA, Einstadter D, Fiegl J, Love TE, Spence D, Jenkins K, Lorenz A, Uddin SJ, McCutcheon Adams K, Konstan MW, Applegate MS, and Bolen SD
- Abstract
Objective: To evaluate the Ohio Diabetes Quality Improvement Project (QIP) equity aim to reduce the percentage of Non-Hispanic Black (NHB) and Hispanic patients with A1C >9% by ≥20% over 2 years., Research Design and Methods: The Ohio Department of Medicaid, Ohio Colleges of Medicine Government Resource Center, Ohio Medicaid managed care plans, and seven medical schools in Ohio formed the Diabetes QIP collaborative using the collective impact model to improve diabetes outcomes and equity in 20 practices across 11 health systems. The quality improvement (QI) strategies included data audit and feedback, peer-to-peer learning, QI coaching/practice facilitation, and subject matter expert consultation through coaching calls, monthly webinars, and annual virtual learning sessions. Electronic health record data were collected for preintervention (2019-2020) and intervention (2020-2022) periods. Assessments of improvements in A1C were based on prevalence of A1C >9% from preintervention, year 1, and year 2 with stratification by race and ethnicity., Results: The Diabetes QIP included 7,689 (54% female) sociodemographically diverse patients, self-identifying as non-Hispanic White (NHW) (42%), NHB (43%), Hispanic (8%), non-Hispanic Asian (4%), or other (3%). In year 2 compared with baseline, there were decreases in the proportion of patients with A1C >9% among NHW, NHB, and Hispanic patients (NHW from 19% to 12% [37% reduction], NHB 23% to 18% [22% reduction], and Hispanic 29% to 23% [20% reduction])., Conclusions: The Ohio Diabetes QIP, focused on multisector collaborative approaches, reduced the percentage of patients with A1C >9% by ≥20% among NHW, NHB, and Hispanic populations. Given the persistence of disparities, further equity-focused refinements are warranted to address disparities in diabetes control., (© 2024 by the American Diabetes Association.)
- Published
- 2024
- Full Text
- View/download PDF
17. The continuing need for dornase alfa for extracellular airway DNA hydrolysis in the era of CFTR modulators.
- Author
-
Roesch EA, Rahmaoui A, Lazarus RA, and Konstan MW
- Subjects
- Humans, Recombinant Proteins therapeutic use, Recombinant Proteins administration & dosage, Hydrolysis, Sputum metabolism, Expectorants therapeutic use, Lung drug effects, Lung metabolism, Lung physiopathology, DNA, Deoxyribonuclease I therapeutic use, Cystic Fibrosis drug therapy, Cystic Fibrosis physiopathology, Cystic Fibrosis metabolism, Cystic Fibrosis Transmembrane Conductance Regulator metabolism, Cystic Fibrosis Transmembrane Conductance Regulator genetics, Extracellular Traps drug effects, Extracellular Traps metabolism
- Abstract
Introduction: The availability of cystic fibrosis transmembrane conductance regulator (CFTR) modulators opens the possibility of discontinuing some chronic pulmonary therapies to decrease cystic fibrosis (CF) treatment burden. However, CFTR modulators may not adequately address neutrophilic inflammation, which contributes to a self-perpetual cycle of viscous CF sputum, airway obstruction, inflammation, and lung function decline., Areas Covered: This review discusses the emerging role of neutrophil extracellular traps in CF and its role in CF sputum viscosity, airway obstruction, and inflammation, based on a literature search of PubMed (1990-present). We summarize clinical trials and real-world studies that support the efficacy of dornase alfa (Pulmozyme) in improving lung function and reducing pulmonary exacerbation in people with CF (PwCF), and we discuss the potential role of dornase alfa in reducing airway inflammation. We also examine the findings of short-term trials evaluating the discontinuation of mucoactive therapy in PwCF receiving CFTR modulators., Expert Opinion: Long-term studies are needed to assess the impact of discontinuing mucoactive therapy in PwCF who are clinically stable while receiving CFTR modulatory therapy. Treatment decisions should take into account the severity of underlying lung disease. People with advanced CF will likely require ongoing mucoactive therapy.
- Published
- 2024
- Full Text
- View/download PDF
18. Efficacy and safety of LAU-7b in a Phase 2 trial in adults with cystic fibrosis.
- Author
-
Konstan MW, Polineni D, Chmiel JF, Bilodeau L, Middleton PG, Matouk E, Houle JM, Pislariu R, Colin P, Kianicka I, Potvin D, Radzioch D, Kotsimbos T, Zuckerman JB, Nasr SZ, Liou TG, and Lands LC
- Abstract
Background: Lung inflammation is associated with tissue damage in cystic fibrosis (CF). LAU-7b, a novel oral drug candidate, was shown to control inflammation and stabilize CFTR protein in the epithelial membrane during inflammatory stress in preclinical models of CF., Methods: A double-blind, randomized, placebo-controlled Phase 2 study was conducted to evaluate efficacy and safety of LAU-7b in adults with CF. LAU-7b or placebo was administered over 24 weeks as six 21-day treatment cycles each separated by 7 days. The primary efficacy endpoint was the absolute change from baseline in percent predicted forced expiratory volume in 1 second (ppFEV
1 ) at 24 weeks., Results: A total of 166 subjects received at least one dose of study drug (Intent-To-Treat population, ITT), of which 122 received ≥5 treatment cycles (Per-Protocol population, PP). Both treatment arms showed a mean lung function loss at 24 weeks of 1.18 ppFEV1 points with LAU-7b and 1.95 ppFEV1 with placebo, a 0.77 ppFEV1 (40 s) difference, p=0.345, and a 0.95 ppFEV1 (49 %) difference in the same direction in PP population, p=0.263. Primary analysis of mean ppFEV1 through 24 weeks showed differences of 1.01 and 1.23 ppFEV1 , in the ITT (65 % less loss, p=0.067) and PP populations (78 % less loss, reaching statistical significance p=0.049), respectively. LAU-7b had an acceptable safety profile., Conclusion: Although the study did not meet its primary efficacy endpoint in the ITT population, LAU-7b was generally well tolerated and showed evidence of preservation of lung function to support further development., Competing Interests: Declaration of competing interest MW Konstan, LC Lands, D Potvin and D Radzioch report receiving consulting fees from Laurent Pharmaceuticals; JM Houle, R Pislariu, P Colin, I Kianicka are employees of Laurent Pharmaceuticals; D Polineni, J Chmiel, L Bilodeau, PG Middleton, E Matouk, T Kotsimbos, JB Zuckerman, SZ Nasr, TG Liou report no conflict of interest related to this study., (Copyright © 2024. Published by Elsevier B.V.)- Published
- 2024
- Full Text
- View/download PDF
19. Heterogeneity of CFTR modulator-induced sweat chloride concentrations in people with cystic fibrosis.
- Author
-
Zemanick ET, Emerman I, McCreary M, Mayer-Hamblett N, Warden MN, Odem-Davis K, VanDevanter DR, Ren CL, Young J, and Konstan MW
- Subjects
- Humans, Male, Female, Chloride Channel Agonists therapeutic use, Adult, Genotype, Adolescent, Child, Quinolines, Pyrazoles therapeutic use, Pyridines, Cystic Fibrosis drug therapy, Cystic Fibrosis genetics, Cystic Fibrosis metabolism, Sweat chemistry, Sweat metabolism, Chlorides analysis, Chlorides metabolism, Cystic Fibrosis Transmembrane Conductance Regulator genetics, Benzodioxoles therapeutic use, Aminophenols therapeutic use, Quinolones therapeutic use, Indoles therapeutic use, Drug Combinations
- Abstract
Background: Sweat chloride (SC) concentrations in people with cystic fibrosis (PwCF) reflect relative CF transmembrane conductance regulator (CFTR) protein function, the primary CF defect. Populations with greater SC concentrations tend to have lesser CFTR function and more severe disease courses. CFTR modulator treatment can improve CFTR function within specific CF genotypes and is commonly associated with reduced SC concentration. However, SC concentrations do not necessarily fall to concentrations seen in the unaffected population, suggesting potential for better CFTR treatment outcomes. We characterized post-modulator SC concentration variability among CHEC-SC study participants by genotype and modulator., Methods: PwCF receiving commercially approved modulators for ≥90 days were enrolled for a single SC measurement. Clinical data were obtained from chart review and the CF Foundation Patient Registry (CFFPR). Variability of post-modulator SC concentrations was assessed by cumulative SC concentration frequencies., Results: Post-modulator SC concentrations (n = 3787) were collected from 3131 PwCF; most (n = 1769, 47 %) were collected after elexacaftor/tezacaftor/ivacaftor (ETI) treatment. Modulator use was associated with lower SC distributions, with post-ETI concentrations the lowest on average. Most post-ETI SC concentrations were <60 mmol/L (79 %); 26 % were <30 mmol/L. Post-ETI distributions varied by genotype. All genotypes containing at least one F508del allele had individuals with post-ETI SC ≥60 mmol/L, with the largest proportion being F508del/minimal function (31 %)., Conclusions: Post-modulator SC concentration heterogeneity was observed among all genotypes and modulators, including ETI. The presence of PwCF with post-modulator SC concentrations within the CF diagnostic range suggests room for additional treatment-associated CFTR restoration in this population., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 European Cystic Fibrosis Society. Published by Elsevier B.V. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
20. Assessing Priorities in a Statewide Cardiovascular and Diabetes Health Collaborative Based on the Results of a Needs Assessment: Cross-Sectional Survey Study.
- Author
-
Beverly EA, Koopman-Gonzalez S, Wright J, Dungan K, Pallerla H, Gubitosi-Klug R, Baughman K, Konstan MW, and Bolen SD
- Abstract
Background: The Ohio Cardiovascular and Diabetes Health Collaborative (Cardi-OH) unites general and subspecialty medical staff at the 7 medical schools in Ohio with community and public health partnerships to improve cardiovascular and diabetes health outcomes and eliminate disparities in Ohio's Medicaid population. Although statewide collaboratives exist to address health improvements, few deploy needs assessments to inform their work., Objective: Cardi-OH conducts an annual needs assessment to identify high-priority clinical topics, screening practices, policy changes for home monitoring devices and referrals, and preferences for the dissemination and implementation of evidence-based best practices. The results of the statewide needs assessment could also be used by others interested in disseminating best practices to primary care teams., Methods: A cross-sectional survey was distributed electronically via REDCap (Research Electronic Data Capture; Vanderbilt University) to both Cardi-OH grant-funded and non-grant-funded members (ie, people who have engaged with Cardi-OH but are not funded by the grant)., Results: In total, 88% (103/117) of Cardi-OH grant-funded members and 8.14% (98/1204) of non-grant-funded members completed the needs assessment survey. Of these, 51.5% (53/103) of Cardi-OH grant-funded members and 47% (46/98) of non-grant-funded members provided direct clinical care. The top cardiovascular medicine and diabetes clinical topics for Cardi-OH grant-funded members (clinical and nonclinical) were lifestyle prescriptions (50/103, 48.5%), atypical diabetes (38/103, 36.9%), COVID-19 and cardiovascular disease (CVD; 38/103, 36.9%), and mental health and CVD (38/103, 36.9%). For non-grant-funded members, the top topics were lifestyle prescriptions (53/98, 54%), mental health and CVD (39/98, 40%), alcohol and CVD (27/98, 28%), and cardiovascular complications (27/98, 28%). Regarding social determinants of health, Cardi-OH grant-funded members prioritized 3 topics: weight bias and stigma (44/103, 42.7%), family-focused interventions (40/103, 38.8%), and adverse childhood events (37/103, 35.9%). Non-grant-funded members' choices were family-focused interventions (51/98, 52%), implicit bias (43/98, 44%), and adverse childhood events (39/98, 40%). Assessment of other risk factors for CVD and diabetes across grant- and non-grant-funded members revealed screening for social determinants of health in approximately 50% of patients in each practice, whereas some frequency of depression and substance abuse screening occurred in 80% to 90% of the patients. Access to best practice home monitoring devices was challenging, with 30% (16/53) and 41% (19/46) of clinical grant-funded and non-grant-funded members reporting challenges in obtaining home blood pressure monitoring devices and 68% (36/53) and 43% (20/46) reporting challenges with continuous glucose monitors., Conclusions: Cardi-OH grant- and non-grant-funded members shared the following high-priority topics: lifestyle prescriptions, CVD and mental health, family-focused interventions, alcohol and CVD, and adverse childhood experiences. Identifying high-priority educational topics and preferred delivery modalities for evidence-based materials is essential for ensuring that the dissemination of resources is practical and useful for providers., (©Elizabeth A Beverly, Sarah Koopman-Gonzalez, Jackson Wright, Kathleen Dungan, Harini Pallerla, Rose Gubitosi-Klug, Kristin Baughman, Michael W Konstan, Shari D Bolen. Originally published in JMIR Formative Research (https://formative.jmir.org), 12.04.2024.)
- Published
- 2024
- Full Text
- View/download PDF
21. Effects of the Communities that Heal (CTH) intervention on perceived opioid-related community stigma in the HEALing Communities Study: results of a multi-site, community-level, cluster-randomized trial.
- Author
-
Davis A, Knudsen HK, Walker DM, Chassler D, Lunze K, Westgate PM, Oga E, Rodriguez S, Tan S, Holloway J, Walsh SL, Oser CB, Lefebvre RC, Fanucchi LC, Glasgow L, McAlearney AS, Surratt HL, Konstan MW, Huang TT, LeBaron P, Nakayima J, Stein MD, Rudorf M, Nouvong M, Kinnard EN, El-Bassel N, Tilley J, Macoubray A, Savitzky C, Farmer A, Beers D, Salsberry P, and Huerta TR
- Abstract
Background: Community stigma against people with opioid use disorder (OUD) and intervention stigma (e.g., toward naloxone) exacerbate the opioid overdose crisis. We examined the effects of the Communities that HEAL (CTH) intervention on perceived opioid-related community stigma by stakeholders in the HEALing Communities Study (HCS)., Methods: We collected three surveys from community coalition members in 66 communities across four states participating in HCS. Communities were randomized into Intervention (Wave 1) or Wait-list Control (Wave 2) arms. We conducted multilevel linear mixed models to compare changes in primary outcomes of community stigma toward people treated for OUD, naloxone, and medication for opioid use disorder (MOUD) by arm from time 1 (before the start of the intervention) to time 3 (end of the intervention period in the Intervention arm)., Findings: Intervention stakeholders reported a larger decrease in perceived community stigma toward people treated for OUD (adjusted mean change (AMC) -3.20 [95% C.I. -4.43, -1.98]) and toward MOUD (AMC -0.33 [95% C.I. -0.56, -0.09]) than stakeholders in Wait-list Control communities (AMC -0.18 [95% C.I. -1.38, 1.02], p = 0.0007 and AMC 0.11 [95% C.I. -0.09, 0.31], p = 0.0066). The relationship between intervention status and change in stigma toward MOUD was moderated by rural-urban status (urban AMC -0.59 [95% CI, -0.87, -0.32], rural AMC not sig.) and state. The difference in stigma toward naloxone between Intervention and Wait-list Control stakeholders was not statistically significant ( p = 0.18)., Interpretation: The CTH intervention decreased stakeholder perceptions of community stigma toward people treated for OUD and stigma toward MOUD. Implementing the CTH intervention in other communities could decrease OUD stigma across diverse settings nationally., Funding: US National Institute on Drug Abuse., Competing Interests: All authors declare no conflicts of interest related to the topic of the manuscript., (© 2024 The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
22. Impact of the Communities That HEAL Intervention on Buprenorphine-Waivered Practitioners and Buprenorphine Prescribing: A Prespecified Secondary Analysis of the HCS Randomized Clinical Trial.
- Author
-
Stopka TJ, Babineau DC, Gibson EB, Knott CE, Cheng DM, Villani J, Wai JM, Blevins D, David JL, Goddard-Eckrich DA, Lofwall MR, Massatti R, DeFiore-Hyrmer J, Lyons MS, Fanucchi LC, Harris DR, Talbert J, Hammerslag L, Oller D, Balise RR, Feaster DJ, Soares W, Zarkin GA, Glasgow L, Oga E, McCarthy J, D'Costa L, Chahine R, Gomori S, Dalvi N, Shrestha S, Garner C, Shadwick A, Salsberry P, Konstan MW, Freisthler B, Winhusen J, El-Bassel N, Samet JH, and Walsh SL
- Subjects
- Adult, Humans, Data Analysis, Educational Status, Intention, Adolescent, Multicenter Studies as Topic, Randomized Controlled Trials as Topic, Buprenorphine therapeutic use, Opiate Overdose, Opioid-Related Disorders drug therapy
- Abstract
Importance: Buprenorphine significantly reduces opioid-related overdose mortality. From 2002 to 2022, the Drug Addiction Treatment Act of 2000 (DATA 2000) required qualified practitioners to receive a waiver from the Drug Enforcement Agency to prescribe buprenorphine for treatment of opioid use disorder. During this period, waiver uptake among practitioners was modest; subsequent changes need to be examined., Objective: To determine whether the Communities That HEAL (CTH) intervention increased the rate of practitioners with DATA 2000 waivers and buprenorphine prescribing., Design, Setting, and Participants: This prespecified secondary analysis of the HEALing Communities Study, a multisite, 2-arm, parallel, community-level, cluster randomized, open, wait-list-controlled comparison clinical trial was designed to assess the effectiveness of the CTH intervention and was conducted between January 1, 2020, to December 31, 2023, in 67 communities in Kentucky, Massachusetts, New York, and Ohio, accounting for approximately 8.2 million adults. The participants in this trial were communities consisting of counties (n = 48) and municipalities (n = 19). Trial arm randomization was conducted using a covariate constrained randomization procedure stratified by state. Each state was balanced by community characteristics including urban/rural classification, fatal opioid overdose rate, and community population. Thirty-four communities were randomized to the intervention and 33 to wait-list control arms. Data analysis was conducted between March 20 and September 29, 2023, with a focus on the comparison period from July 1, 2021, to June 30, 2022., Intervention: Waiver trainings and other educational trainings were offered or supported by the HEALing Communities Study research sites in each state to help build practitioner capacity., Main Outcomes and Measures: The rate of practitioners with a DATA 2000 waiver (overall, and stratified by 30-, 100-, and 275-patient limits) per 100 000 adult residents aged 18 years or older during July 1, 2021, to June 30, 2022, were compared between the intervention and wait-list control communities. The rate of buprenorphine prescribing among those waivered practitioners was also compared between the intervention and wait-list control communities. Intention-to-treat and per-protocol analyses were performed., Results: A total of 8 166 963 individuals aged 18 years or older were residents of the 67 communities studied. There was no evidence of an effect of the CTH intervention on the adjusted rate of practitioners with a DATA 2000 waiver (adjusted relative rate [ARR], 1.04; 95% CI, 0.94-1.14) or the adjusted rate of practitioners with a DATA 2000 waiver who actively prescribed buprenorphine (ARR, 0.97; 95% CI, 0.86-1.10)., Conclusions and Relevance: In this randomized clinical trial, the CTH intervention was not associated with increases in the rate of practitioners with a DATA 2000 waiver or buprenorphine prescribing among those waivered practitioners. Supporting practitioners to prescribe buprenorphine remains a critical yet challenging step in the continuum of care to treat opioid use disorder., Trial Registration: ClinicalTrials.gov Identifier: NCT04111939.
- Published
- 2024
- Full Text
- View/download PDF
23. Longitudinal bacterial prevalence in cystic fibrosis airways: Fact and artifact.
- Author
-
VanDevanter DR, LiPuma JJ, and Konstan MW
- Subjects
- Humans, Prevalence, Artifacts, Lung microbiology, Cystic Fibrosis Transmembrane Conductance Regulator, Bacteria, Cystic Fibrosis complications
- Abstract
Background: Opportunistic bacterial infection is a hallmark of cystic fibrosis (CF) lung disease and early mortality. Poorly characterized prevalence changes have accompanied two decades of health improvements, with CFTR modulators likely to further affect infection epidemiology., Methods: Bacterial prevalence change trends across birth cohorts were assessed with linear regression using 2001-2019 US CF Foundation Patient Registry data. Informative missingness was assessed, as was age-to-age infection status., Results: Bacterial prevalence constantly changed from 2001 to 2019, with changes differing across birth cohorts. Informative censoring affected prevalence change for some organisms. Age-to-age infection status changes were greater than net changes in bacterial prevalence and varied by age., Conclusions: CF infection epidemiology changed over two decades and will continue to do so. Understanding how modulators affect infection epidemiology will require creative designs for longitudinal prevalence change studies emphasizing prevalence changes independent of effects on lung biology., Competing Interests: Declaration of Competing Interest The authors have no competing interests relative to this content to report. MWK received salary support to his institution from the Cystic Fibrosis Foundation and the National Institutes of Health (UL1TR002548). JJL received salary support from the Cystic Fibrosis Foundation (CAVERL22AB0)., (Copyright © 2023 European Cystic Fibrosis Society. Published by Elsevier B.V. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
24. A Medicaid-Funded Statewide Diabetes Quality Improvement Collaborative: Ohio 2020‒2022.
- Author
-
Bolen SD, Joseph JJ, Dungan KM, Beverly EA, Perzynski AT, Einstadter D, Fiegl J, Love TE, Spence D, Jenkins K, Lorenz A, Uddin SJ, Adams KM, Konstan MW, and Applegate MS
- Subjects
- United States, Humans, Ohio, Quality Improvement, Medicaid, Diabetes Mellitus epidemiology, Diabetes Mellitus therapy
- Abstract
We used a collective impact model to form a statewide diabetes quality improvement collaborative to improve diabetes outcomes and advance diabetes health equity. Between 2020 and 2022, in collaboration with the Ohio Department of Medicaid, Medicaid Managed Care Plans, and Ohio's seven medical schools, we recruited 20 primary care practices across the state. The percentage of patients with hemoglobin A1c greater than 9% improved from 25% to 20% over two years. Applying our model more broadly could accelerate improvement in diabetes outcomes. ( Am J Public Health. 2023;113(12):1254-1257. https://doi.org/10.2105/AJPH.2023.307410).
- Published
- 2023
- Full Text
- View/download PDF
25. Advancing the pipeline of cystic fibrosis clinical trials: a new roadmap with a global trial network perspective.
- Author
-
Mayer-Hamblett N, Clancy JP, Jain R, Donaldson SH, Fajac I, Goss CH, Polineni D, Ratjen F, Quon BS, Zemanick ET, Bell SC, Davies JC, Jain M, Konstan MW, Kerper NR, LaRosa T, Mall MA, McKone E, Pearson K, Pilewski JM, Quittell L, Rayment JH, Rowe SM, Taylor-Cousar JL, Retsch-Bogart G, and Downey DG
- Subjects
- Humans, Cystic Fibrosis Transmembrane Conductance Regulator genetics, Genetic Therapy, Quality of Life, Mutation, Cystic Fibrosis drug therapy
- Abstract
The growing use of modulator therapies aimed at restoring cystic fibrosis transmembrane conductance regulator (CFTR) protein function in people with cystic fibrosis has fundamentally altered clinical trial strategies needed to advance new therapeutics across an orphan disease population that is now divided by CFTR modulator eligibility. The development of a robust pipeline of nucleic acid-based therapies (NABTs)-initially directed towards the estimated 10% of the cystic fibrosis population who are genetically ineligible for, or intolerant of, CFTR modulators-is dependent on the optimisation of restricted trial participant resources across multiple development programmes, a challenge that will preclude the use of gold standard placebo-controlled trials. Advancement of a full pipeline of symptomatic therapies across the entire cystic fibrosis population will be challenged by smaller effect sizes and uncertainty regarding their clinical importance in a growing modulator-treated population with more mild and stable pulmonary disease. In this Series paper, we aim to lay the foundation for clinical trial strategy and community partnership that must deviate from established and familiar precedent to advance the future pipeline of cystic fibrosis therapeutics., Competing Interests: Declaration of interests NM-H reports grants from the Cystic Fibrosis Foundation (CFF), the National Institutes of Health (NIH; P30 DK 089507 and UL1 TR002319), and the Food and Drug Administration (FDA); consulting fees from Enterprise Therapeutics; and data safety monitoring board (DSMB) membership for the NIH. JPC is an employee of CFF. RJ reports grants from CFF; consulting fees from Boehringer Ingelheim and Recode Therapeutics; honoraria from Vertex Pharmaceuticals; and travel support from CFF. SHD reports contracts from Calithera, CFF, NIH (P30 DK065988), Vertex Pharmaceuticals, 4D Molecular Therapeutics, and Chiesi USA; consulting fees from Polarean, 501 Ventures, Enterprise Therapeutics, and Boehringer Ingelheim; fees for participation on advisory boards for Innova Healthcare and Boehringer Ingleheim; travel fees from Enterprise Therapeutics and CFF; and participation on a board for Abbvie. IF reports grants from AbbVie, Bayer, Boehringer Ingelheim, Insmed, GSK, and Vertex Pharmaceuticals; honoraria from Vertex Pharmaceuticals; board participation for AbbVie, Boehringer Ingelheim, Kither Biotech, and Vertex Pharmaceuticals; and support to her institution from the European Cystic Fibrosis Society (ECFS). CHG reports grants and contracts from CFF, NIH (P30 DK 089507 and UL1 TR000423), and FDA; consulting fees from Enterprise Therapeutics; honoraria from Gilead Sciences and Vertex Pharmaceuticals; travel support from Vertex Pharmaceuticals and Enterprise Therapeutics; participation on the board for Novartis; stock options for Air Therapeutics; and leadership roles for the American Thoracic Society (ATS). DP reports grants from CFF (002805121), NIH, and Aclaris Pharmaceuticals; travel support from CFF; and board participation for Vertex Pharmaceuticals and Translate Bio. FR reports grants from Vertex Pharmaceuticals and consulting fees from Vertex Pharmaceuticals and Calithera. BSQ reports grants from CFF, Cystic Fibrosis Canada, Vertex Pharmaceuticals, and Gilead Sciences; honoraria from Vertex Pharmaceuticals; and travel support from CFF. ETZ reports grants from NIH, Vertex Pharmaceuticals, and CFF (002884121); consulting fees from CFF; travel support from CFF, Vertex Pharmaceuticals, and ECFS; and participation on boards for CFF and Vertex Pharmaceuticals. SCB reports grants from the National Health and Medical Research Council Australia (APP1102494), the Medical Research Futures Fund Australia, and CFF (BELL1480 and BELL19A0); and honoraria from Vertex Pharmaceuticals. JCD reports grants from the UK Cystic Fibrosis Trust (as part of their Clinical Trials Accelerator Platform), CFF, Cystic Fibrosis Ireland, the Engineering and Physical Sciences Research Council, and the National Institute for Health and Care Research; and honoraria from Vertex Pharmaceuticals, Boehringer Ingelheim, Eloxx, Algipharma, Abbvie, Arcturus, Enterprise Therapeutics, Recode, LifeArc, Genentech, and Tavanta. JCD serves as Deputy Editor for the Journal of Cystic Fibrosis. MWK reports grants from NIH (P01HL128192 and UL1TR002548) and CFF; consulting fees from AbbVie, AzurRx, Cystetic Medicines, EnBiotix, First Wave Biopharma, Insmed, Laurent Pharmaceuticals, Mylan, and PBM BC Holdings; board participation for AbbVie, CFF, First Wave Biopharma, Insmed, Laurent Pharmaceuticals, Sionna, and Vertex; and committee membership for CFF. MAM reports grants from the German Research Foundation (CRC 1449 project #431232613), the German Ministry for Education and Research (82DZL009B1), the German Innovation Fund, and Vertex Pharmaceuticals; consulting fees from Abbvie, Antabio, Arrowhead, Boehringer Ingelheim, Enterprise Therapeutics, Kither Biotec, Prieris, Recode, Santhera, Splisense, and Vertex Pharmaceuticals; honoraria from Vertex Pharmaceuticals; travel support from Boehringer Ingelheim and Vertex Pharmaceuticals; and participation on boards for Abbvie, Antabio, Arrowhead, Boehringer Ingelheim, Enterprise Therapeutics, Kither Biotec, Pari, and Vertex Pharmaceuticals. EM reports grants from Vertex Pharmaceuticals; honoraria from Vertex Phamaceuticals; travel support from Menarini; and board participation for the Cystic Fibrosis Storm Clinical Trial, Vertex Pharmaceuticals, Janssen, Abbvie, and Insmed. JHR reports grants from Cystic Fibrosis Canada, CFF, Vertex Phamaceuticals, the Canada Foundation for Innovation, and the Canadian Institutes of Health Research; consulting fees from Sanofi; and travel support from Vertex Pharmaceuticals. SMR reports grant funding from CFF (P30DK072482), NIH (UL1TR003096), Vertex Pharmaceuticals, Galapagos/Abbvie, Eloxx, Synspira, Translate Bio, Arcturus, Astra-Zenica, and Ionis; and consulting fees from Vertex Pharmaceuticals, Synspira (including stock options), Renovion (including stock options), Cystetic Medicines, and Arcturus. SMR's potential competing interests were resolved or ended in 2022 or earlier. JLT-C reports grants and contracts from CFF, Vertex Pharmaceutics, Eloxx, and 4DMT; consulting fees from Vertex Phamaceuticals, Insmed, and 4DMT; participation on a DSMB for Abbvie; and participation on advisory boards for CFF, ATS, Journal of Cystic Fibrosis, The Lancet Respiratory Medicine, and Emily's Entourage. GR-B reports grants and contracts from Vertex Pharmaceuticals and CFF. DGD reports grants from Chiesi Farmaceutici and CFF; consulting fees from Vertex and Insmed; honoraria from Chiesi and Gilead; travel support from ECFS and CFF; board participation for Momab and CSL Behring; and support from ECFS as director of the Clinical Trials Network. MJ, NRK, TL, KP, JMP, and LQ declare no competing interests., (Copyright © 2023 Elsevier Ltd. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
26. Willingness of people with cystic fibrosis receiving elexacaftor/tezacaftor/ivacaftor (ETI) to participate in randomized modulator and inhaled antimicrobial clinical trials.
- Author
-
VanDevanter DR, Zemanick ET, Konstan MW, Ren CL, Odem-Davis K, Emerman I, Young J, and Mayer-Hamblett N
- Subjects
- Humans, Cystic Fibrosis Transmembrane Conductance Regulator genetics, Benzodioxoles adverse effects, Aminophenols adverse effects, Mutation, Cystic Fibrosis drug therapy, Cystic Fibrosis genetics, Anti-Infective Agents
- Abstract
Objective: To assess the feasibility of enrolling people with CF (pwCF) taking the CFTR modulator elexacaftor/tezacaftor/ivacaftor (ETI) in clinical trials of a new modulator., Methods: PwCF receiving ETI at CHEC-SC study (NCT03350828) enrollment were surveyed for interest in 2-week to 6-month placebo- (PC) and active-comparator (AC) modulator studies. Those taking inhaled antimicrobials (inhABX) were surveyed for interest in PC inhABX studies., Results: Of 1791 respondents, 75% [95% CI 73, 77] would enroll in a 2-week PC modulator study versus 51% [49, 54] for a 6-month study; 82% [81, 84] and 63% [61, 65] would enroll in 2-week and 6 month AC studies; 77% [74, 80] of 551 taking inhABX would enroll in a 2-week PC inhABX study versus 59% [55, 63] for a 6-month study. Previous clinical trial experience increased willingness., Conclusions: Study designs will affect feasibility of future clinical trials of new modulators and inhABX in people receiving ETI., Competing Interests: Declaration of Competing Interest The authors have no competing or conflicting interests to report., (Copyright © 2023. Published by Elsevier B.V.)
- Published
- 2023
- Full Text
- View/download PDF
27. Corrigendum to FEV1 Variability Helps Identify Patients with Cystic Fibrosis at Risk of Greater Loss of Lung Function [The Journal of Pediatrics (2016) 116-121].
- Author
-
Morgan WJ, VanDevanter DR, Pasta DJ, Foreman AJ, Wagener JS, and Konstan MW
- Published
- 2023
- Full Text
- View/download PDF
28. Rate of Lung Function Decline in People with Cystic Fibrosis Having a Residual Function Gene Mutation.
- Author
-
Sawicki GS, Konstan MW, McKone EF, Moss RB, Lubarsky B, Suthoff E, Millar SJ, Pasta DJ, Mayer-Hamblett N, Goss CH, Morgan WJ, Duncan ME, and Yang Y
- Abstract
Introduction: Cystic fibrosis (CF) is an autosomal recessive disease caused by mutations in the CF transmembrane conductance regulator (CFTR) gene. Approximately 5% of people with CF have residual function (RF) CFTR mutations that result in partially retained CFTR activity. Published literature on disease trajectory among those with RF mutations is limited. In this retrospective study, we characterized lung function decline across different age groups in CFTR modulator-untreated people with CF heterozygous for F508del and an RF mutation (F/RF)., Methods: Rate of decline in percent predicted forced expiratory volume in 1 s (ppFEV
1 ) was analyzed using data from the US CF Foundation Patient Registry (2006-2014) in F/RF (all), F/RF (excluding R117H), and F508del homozygous (F/F) cohorts. Annual rates of ppFEV1 decline were estimated over 2-year periods based on calendar year. Subgroup analyses by age [6-12 (children), 13-17 (adolescents), 18-24 (young adults), and ≥ 25 years (adults)] were performed., Results: The estimated annualized rate of ppFEV1 decline was - 0.70 percentage points per year (95% CI -1.09, -0.30) in the F/RF (all) cohort (N = 1242) versus -1.91 percentage points per year (95% CI -2.01, -1.80) in the F/F cohort (N = 11,916) [difference, 1.29 percentage points per year (95% CI 0.88, 1.70); P < 0.001]. In the F/RF (all) cohort, all age groups demonstrated lung function decline ranging from -0.30 to -1.38. In the F/RF (excluding R117H) cohort, the rate of decline was -1.05 percentage points per year (95% CI -1.51, -0.60) [difference versus F/F cohort, 0.95 percentage points per year (95% CI 0.48, 1.41; P < 0.001); not statistically significant in children and young adults]., Conclusion: Progressive lung function decline was observed in people with F/RF genotypes across all assessed age groups, reinforcing the importance of early intervention and clinical monitoring to preserve lung function in all people with CF., (© 2022. The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
29. Forming Cardi-OH: A Statewide Collaborative to Improve Cardiovascular Health in Ohio.
- Author
-
Bolen SD, Beverly EA, Khoury S, Regan S, Wright JT Jr, Koroukian S, Wexler R, Rao G, Hargraves D, Bricker D, Solomon GD, Holliday M, Gardner-Buckshaw S, Dworkin L, Perzynski AT, Littman E, Nevar A, Swiatkowski SM, Applegate M, and Konstan MW
- Abstract
Background Cardiovascular risk factor control is challenging, especially in disadvantaged populations. However, few statewide efforts exist to tackle this challenge. Therefore, our objective is to describe the formation of a unique statewide cardiovascular health collaborative so others may learn from this approach. Methodology With funding from the Ohio Department of Medicaid's Ohio Medicaid Technical Assistance and Policy Program, we used a collective impact model to link the seven medical schools in Ohio, primary care clinics across the state, the Ohio Department of Medicaid, and Ohio's Medicaid Managed Care Plans in a statewide health improvement collaborative for expanding primary care capacity to improve cardiovascular health in Ohio. Results Initial dissemination activities for primary care teams included a virtual case-based learning series focused on hypertension and social determinants of health, website resources, a monthly newsletter with clinical tips, webinars, and in-person conferences. The collaborative is aligned with a separately funded hypertension quality improvement project for paired implementation. Conclusions The collective impact model is a useful framework for developing a statewide collaborative focused on the dissemination and implementation of evidence-based best practices for cardiovascular health improvement and disparity reduction. Statewide collaboratives bringing payers, clinicians, and academic partners together have the potential to substantially impact cardiovascular health., Competing Interests: The authors have declared that no competing interests exist., (Copyright © 2022, Bolen et al.)
- Published
- 2022
- Full Text
- View/download PDF
30. Genomic heterogeneity underlies multidrug resistance in Pseudomonas aeruginosa: A population-level analysis beyond susceptibility testing.
- Author
-
Rojas LJ, Yasmin M, Benjamino J, Marshall SM, DeRonde KJ, Krishnan NP, Perez F, Colin AA, Cardenas M, Martinez O, Pérez-Cardona A, Rhoads DD, Jacobs MR, LiPuma JJ, Konstan MW, Vila AJ, Smania A, Mack AR, Scott JG, Adams MD, Abbo LM, and Bonomo RA
- Subjects
- Anti-Bacterial Agents pharmacology, Anti-Bacterial Agents therapeutic use, Drug Resistance, Multiple, Female, Humans, Microbial Sensitivity Tests, Phylogeny, Pseudomonas aeruginosa, Cystic Fibrosis complications, Cystic Fibrosis drug therapy, Cystic Fibrosis genetics, Pseudomonas Infections microbiology
- Abstract
Background: Pseudomonas aeruginosa is a persistent and difficult-to-treat pathogen in many patients, especially those with Cystic Fibrosis (CF). Herein, we describe a longitudinal analysis of a series of multidrug resistant (MDR) P. aeruginosa isolates recovered in a 17-month period, from a young female CF patient who underwent double lung transplantation. Our goal was to understand the genetic basis of the observed resistance phenotypes, establish the genomic population diversity, and define the nature of sequence evolution over time., Methods: Twenty-two sequential P. aeruginosa isolates were obtained within a 17-month period, before and after a double-lung transplant. At the end of the study period, antimicrobial susceptibility testing, whole genome sequencing (WGS), phylogenetic analyses and RNAseq were performed in order to understand the genetic basis of the observed resistance phenotypes, establish the genomic population diversity, and define the nature of sequence changes over time., Results: The majority of isolates were resistant to almost all tested antibiotics. A phylogenetic reconstruction revealed 3 major clades representing a genotypically and phenotypically heterogeneous population. The pattern of mutation accumulation and variation of gene expression suggested that a group of closely related strains was present in the patient prior to transplantation and continued to change throughout the course of treatment. A trend toward accumulation of mutations over time was observed. Different mutations in the DNA mismatch repair gene mutL consistent with a hypermutator phenotype were observed in two clades. RNAseq performed on 12 representative isolates revealed substantial differences in the expression of genes associated with antibiotic resistance and virulence traits., Conclusions: The overwhelming current practice in the clinical laboratories setting relies on obtaining a pure culture and reporting the antibiogram from a few isolated colonies to inform therapy decisions. Our analyses revealed significant underlying genomic heterogeneity and unpredictable evolutionary patterns that were independent of prior antibiotic treatment, highlighting the need for comprehensive sampling and population-level analysis when gathering microbiological data in the context of CF P. aeruginosa chronic infection. Our findings challenge the applicability of antimicrobial stewardship programs based on single-isolate resistance profiles for the selection of antibiotic regimens in chronic infections such as CF., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2022
- Full Text
- View/download PDF
31. Increasing life expectancy in cystic fibrosis: Advances and challenges.
- Author
-
McBennett KA, Davis PB, and Konstan MW
- Subjects
- Aminophenols, Cystic Fibrosis Transmembrane Conductance Regulator genetics, Humans, Life Expectancy, Mutation, Quality of Life, Cystic Fibrosis
- Abstract
Since the first description of cystic fibrosis in 1938, there have been significant advances in both quality of life and longevity for people living with this disease. In this article we describe the milestones of the last 80 years and what we perceive to be the remaining barriers to normalcy for this population., (© 2021 Wiley Periodicals LLC.)
- Published
- 2022
- Full Text
- View/download PDF
32. What Is Cystic Fibrosis?
- Author
-
Endres TM and Konstan MW
- Subjects
- Female, Humans, Male, Sex Factors, Cystic Fibrosis complications, Cystic Fibrosis diagnosis, Cystic Fibrosis genetics, Cystic Fibrosis therapy, Cystic Fibrosis Transmembrane Conductance Regulator genetics
- Published
- 2022
- Full Text
- View/download PDF
33. Disease burden in people with cystic fibrosis heterozygous for F508del and a minimal function mutation.
- Author
-
Sawicki GS, Van Brunt K, Booth J, Bailey E, Millar SJ, Konstan MW, and Flume PA
- Subjects
- Adolescent, Adult, Child, Child, Preschool, Cross-Sectional Studies, Female, Genotype, Humans, Male, Mutation, Registries, Retrospective Studies, Young Adult, Cost of Illness, Cystic Fibrosis genetics, Cystic Fibrosis therapy, Cystic Fibrosis Transmembrane Conductance Regulator genetics, Disease Progression
- Abstract
Background: People with cystic fibrosis (CF) heterozygous for F508del-CFTR and a minimal function CFTR mutation (F/MF) that results in no CFTR protein or results in CFTR protein that is not responsive to tezacaftor, ivacaftor, and tezacaftor/ivacaftor in vitro comprise a sizeable percentage of the US CF population. This retrospective, cross-sectional, observational study aimed to characterize CF burden in this subpopulation., Methods: People ≥2 years of age in the US CF Foundation Patient Registry with a CF diagnosis, F/MF genotype, and ≥1 encounters in 2017 were included. Descriptive analyses assessed lung function, nutritional parameters, microbiology, hospitalization and pulmonary exacerbation rates, and CF-related complications. Results were stratified by age group; select characteristics were summarized by percent predicted FEV
1 (ppFEV1 ) and ethnicity., Results: 5348 people met inclusion criteria. Rates of positive bacterial cultures, pulmonary exacerbations, and hospitalizations were generally higher in older age groups. Prevalence of prescribed symptomatic CF therapies was substantial and also generally higher in older age groups. ppFEV1 was lower in older age groups. A greater percentage of adolescents and adults reported complications, including cirrhosis, osteoporosis, osteopenia, and sinus disease, than younger age groups. Increased prevalence of cultured Pseudomonas aeruginosa and prescribed chronic therapy was seen with decreasing ppFEV1 . In each age group, ppFEV1 was slightly higher in the non-Hispanic cohort than in the Hispanic cohort., Conclusions: People with F/MF genotypes have substantial disease burden that worsened in older age groups consistent with the progressive nature of CF, indicating need for additional treatment options in this subpopulation., Competing Interests: Declaration of Competing Interest All authors received nonfinancial assistance (assistance with manuscript preparation) from ArticulateScience LLC, which received funding from Vertex Pharmaceuticals Incorporated. Additional disclosures are as follows: GSS reports personal fees from Vertex; KVB and JB are employees of Vertex and may own stock or stock options in Vertex; EB is a former employee of Vertex and may own stock or stock options in the company; SJM is an employee of ICON Clinical Research, which received funding from various pharmaceutical, biotechnology, and device companies for providing clinical research services and received funding from Vertex for analytical services during the conduct of this study; MWK reports grants and personal fees from Vertex; PAF reports grants and personal fees from Vertex., (Copyright © 2021. Published by Elsevier B.V.)- Published
- 2022
- Full Text
- View/download PDF
34. Measuring the impact of CFTR modulation on sweat chloride in cystic fibrosis: Rationale and design of the CHEC-SC study.
- Author
-
Zemanick ET, Konstan MW, VanDevanter DR, Rowe SM, Clancy JP, Odem-Davis K, Skalland M, and Mayer-Hamblett N
- Subjects
- Adolescent, Adult, Aged, Aminophenols, Aminopyridines, Benzodioxoles, Child, Drug Combinations, Female, Humans, Male, Middle Aged, Prospective Studies, Quinolones, Chloride Channel Agonists therapeutic use, Chlorides metabolism, Cystic Fibrosis diagnosis, Cystic Fibrosis Transmembrane Conductance Regulator drug effects, Sweat chemistry
- Abstract
Background: The Characterizing CFTR Modulated Changes in Sweat Chloride and their Association with Clinical Outcomes (CHEC-SC) study is a large epidemiologic study designed to determine the relationship between sweat chloride response and clinical outcomes in people with cystic fibrosis (CF) on commercially approved CFTR modulators. A challenge to study feasibility was capturing sweat chloride measurements before modulator initiation. We tested the hypothesis that historic sweat chloride approximated contemporary pre-modulator values to estimate CFTR modulator-induced changes, allowing a single-visit study design., Methods: GOAL and PROSPECT were multi-center prospective studies of individuals initiating ivacaftor or lumacaftor-ivacaftor. At enrollment, pre-modulator sweat chloride was measured and historic results recorded. Post-modulator sweat chloride was measured at 1, 3 and 6 months. For this analysis, differences between historic and pre-modulator sweat chloride were estimated. CFTR modulator-induced sweat chloride mean changes were compared using historic and pre-modulator sweat chloride., Results: Paired historic and pre-modulator sweat chloride (n=406 participants) revealed a non-significant mean change of -1.0 mmol/L (95% CI: -2.71, 0.66) over an average of 17.2 years. Calculating sweat response to ivacaftor or lumacaftor-ivacaftor using historic or pre-modulator values resulted in similar estimates of modulator response. Based on these results, the CHEC-SC study was designed with a single, post-modulator sweat chloride measurement., Conclusions: Historic sweat chloride values provide a reliable estimate of pre-modulator sweat chloride for people starting on modulator therapy. The CHEC-SC study anticipates capturing approximately 5,000 sweat chloride values, providing an unprecedented understanding of sweat chloride across the CF population in the era of CFTR modulators., Competing Interests: Declaration of Competing Interest ETZ, NMH, DRV, SMR, MWK: grants or consulting CFF; DRV consults for aMoon, Arrevus, Eloxx, Enbiotix, Felix, Ionis, Matinas, Merck, Polyphor, Respirion, Savara; SMR consults for Vertex; MWK consults for Anthera, AzurRx, Celtaxsys, Chiesi, Ionis, Kala, Laurent, Merck, Paranta, pH Pharma, Santhera, Vertex; JPC, KOD and MS: none., (Copyright © 2021. Published by Elsevier B.V.)
- Published
- 2021
- Full Text
- View/download PDF
35. Long-term amikacin liposome inhalation suspension in cystic fibrosis patients with chronic P. aeruginosa infection.
- Author
-
Bilton D, Fajac I, Pressler T, Clancy JP, Sands D, Minic P, Cipolli M, Galeva I, Solé A, Quittner AL, Jumadilova Z, Ciesielska M, and Konstan MW
- Subjects
- Administration, Inhalation, Adolescent, Adult, Child, Chronic Disease, Female, Forced Expiratory Volume, Humans, Liposomes, Male, Middle Aged, Suspensions, Amikacin administration & dosage, Anti-Bacterial Agents administration & dosage, Cystic Fibrosis drug therapy, Cystic Fibrosis microbiology, Pseudomonas Infections drug therapy
- Abstract
Background: . In CLEAR-108-a phase 3, randomised, open-label study-once-daily amikacin liposome inhalation suspension (ALIS) was noninferior to twice-daily tobramycin inhalation solution (TIS) in improving lung function in patients with cystic fibrosis (CF) and chronic Pseudomonas aeruginosa infection after 3 treatment cycles (28 days on/28 days off). The CLEAR-110 extension study (ClinicalTrials.gov: NCT01316276; EudraCT: 2011-000443-24) assessed long-term safety, tolerability, and efficacy of ALIS in eligible patients who completed CLEAR-108., Methods: . Patients received once-daily ALIS 590 mg for 12 treatment cycles (96 weeks). Patients were grouped by prior treatment: the "prior-ALIS" cohort received ALIS in CLEAR-108, and the "ALIS-naive" cohort received TIS in CLEAR-108., Results: . Overall, 206 patients (prior-ALIS, n=92; ALIS-naive, n=114) entered CLEAR-110 and received ≥1 dose of ALIS. Most patients (88.8%) experienced ≥1 treatment-emergent adverse event (TEAE) through day 672 (end of year 2). Most TEAEs (72.3%) were mild or moderate in severity. Severe TEAEs were reported in 31 patients (15.0%). Two life-threatening TEAEs (haemoptysis; intestinal obstruction) and 1 death (cardiac failure) were reported. Twenty-one patients (10.2%) discontinued treatment due to a TEAE (mostly infective pulmonary exacerbation of CF). Mean change from baseline in forced expiratory volume in 1 second percent predicted at day 672 was -3.1% (prior-ALIS, -4.0%; ALIS-naive, -2.3%). Mean change from baseline in sputum density of P. aeruginosa at day 672 was 0.02 (prior-ALIS, -0.16; ALIS-naive, 0.19) log CFU/g., Conclusions: . Long-term treatment with ALIS was well tolerated with a favourable adverse event profile and demonstrated continued antibacterial activity in CF patients with chronic P. aeruginosa infection., Competing Interests: Declaration of Competing Interest D. Bilton: National Institute for Health Research funding support through the Imperial College, Royal Brompton Hospital, Specialist Respiratory Bio-Medical Research Unit; has received research contract support from Insmed Incorporated. I. Fajac: Received research contract support from Insmed Incorporated, has received a research grant from Actelion, and has served on advisory boards for Gilead Sciences and Vertex. T. Pressler: Received research contract support from Insmed Incorporated. J. P. Clancy: Serves on the Scientific Advisory Board and has received clinical trial support for phase II studies of ALIS in CF for Insmed Incorporated. D. Sands: Received research contract support from Insmed Incorporated. P. Minic: Received research contract support from Insmed Incorporated. M. Cipolli: Received research contract support from Insmed Incorporated, a grant from Vertex Pharmaceuticals, and served on advisory boards for Chiesi and Vertex. I. Galeva: Received research contract support from Insmed Incorporated. A. Solé: Has served on the advisory board for Gilead Sciences and Vertex. A. L. Quittner: Consulting for AbbVie, Aradigm, Insmed Incorporated, and Vertex; investigator-initiated studies for the Cystic Fibrosis Foundation. Z. Jumadilova: Employee of Insmed Incorporated. M. Ciesielska: Employee of Insmed Incorporated. M. Konstan: Has served on the Arikayce Clinical Program Steering Committee; has received grants and personal fees from Insmed Incorporated, Laurent Pharmaceuticals, Anthera, the Cystic Fibrosis Foundation, and AzurRX; has received personal fees from Chiesi, Celtazsys, Ionis Pharmaceuticals, Merck, Paranta Biosciences, Santhera, Ph Pharma, and Kala Pharmaceuticals; and has received nonfinancial support from Insmed Incorportated., (Copyright © 2021. Published by Elsevier B.V.)
- Published
- 2021
- Full Text
- View/download PDF
36. Empire-CF study: A phase 2 clinical trial of leukotriene A4 hydrolase inhibitor acebilustat in adult subjects with cystic fibrosis.
- Author
-
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, and Rowe SM
- Subjects
- Adolescent, Adult, Double-Blind Method, Female, Humans, Male, Respiratory Function Tests, Severity of Illness Index, Azabicyclo Compounds therapeutic use, Benzoates therapeutic use, Cystic Fibrosis drug therapy, Epoxide Hydrolases therapeutic use
- Abstract
Background: Cystic fibrosis (CF) is characterized by neutrophilic inflammation in the airways. Leukotriene B4 (LTB
4 ) 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., Competing Interests: Declaration of Competing Interest Dr. Aaron has nothing to disclose. Dr. Ahuja reports personal fees from Celtaxsys, Inc., during the conduct of the study; In addition, Dr. Ahuja has a patent PCT/US19/034810 pending, and a patent US16/427,571 pending. Dr. Daines has nothing to disclose. Dr. Downey reports and Honoraria from Vertex, Proteostasis and Chiesi. Dr. Elborn reports other from Celtaxsys, during the conduct of the study; grants and other from Corbus, grants from Novartis, grants from polyphor, outside the submitted work. Dr. Fajac reports grants from Celtaxsys, during the conduct of the study; grants and personal fees from Boehringer Ingelheim, grants and personal fees from Proteostasis Therapeutics, grants and personal fees from Vertex Pharmaceuticals, outside the submitted work. Mr. Grosswald reports personal fees from Celtaxsys, Inc., during the conduct of the study; In addition, Dr. Grosswald has a patent PCT/US19/034810 pending, and a patent US16/427,571 pending. Dr. Horsley reports other from Celtaxys pharmaceuticals, during the conduct of the study; grants from NIHR, grants from CF Trust, grants from CF Foundation, personal fees from Vertex pharmaceuticals, personal fees from Mylan, outside the submitted work. Dr. Konstan reports personal fees from Celtaxsys, during the conduct of the study; grants and personal fees from Vertex Pharmaceuticals, personal fees from Chiesi, personal fees from Ionis Pharmaceuticals, grants and personal fees from Laurent Pharmaceuticals, grants and personal fees from Anthera, personal fees from Merck, personal fees from Paranta Biosciences, personal fees from Santhera, personal fees from pH Pharma, grants and personal fees from Cystic Fibrosis Foundation, grants from National Institutes of Health, grants and personal fees from AzurRx, personal fees from Kala Pharmaceuticals, outside the submitted work. John Mershon has nothing to disclose. Dr. Rowe reports grants from Celtaxsys, during the conduct of the study; grants and personal fees from Novartis, grants and personal fees from Bayer, grants from Translate Bio, non-financial support from Proteostasis, grants, personal fees and non-financial support from Galapagos/Abbvie, grants, personal fees and other from Synedgen/Synspira, grants from Eloxx, grants from Celtaxsys, grants, personal fees, non-financial support and other from Vertex Pharmaceuticals Inc, personal fees from Renovion, grants and personal fees from Arrowhead, grants and other from Ionis, grants from Astra Zenica, outside the submitted work. Dr. Springman reports personal fees from Celtaxsys, Inc., during the conduct of the study; In addition, Dr. Springman has a patent PCT/US19/034810 pending, and a patent US16/427,571 pending. Dr. Sutharsan reports personal fees personal Proteostasis Therapeutics, personal fees from Novartis Pharma GmbH, personal fees from Vertex Pharmaceuticals Incorporated, personal fees from Teva GmbH, personal fees from Chiesi GmbH, outside the submitted work; . SS has served as an investigator in clinical trials for Galapagos NV, Proteostasis, Celtaxsys, Flatley Discovery Lab, Novartis and Vertex. Dr. Taylor-Cousar reports grants and personal fees from Celtaxys, during the conduct of the study; grants and personal fees from Gilead, grants from N30, grants and personal fees from Vertex, grants and personal fees from Proteostasis, grants from Bayer, personal fees from Novartis, personal fees from Genentech, personal fees from Protalix, personal fees from Santhera, personal fees from 4DMT, personal fees from Polarean Imaging, personal fees from Insmed, personal fees from Abbvie, outside the submitted work; and Service on the CF TDN Clinical Research Executive CommitteeÐervice as the Chair of the ATS Clinical Problems Assembly Program Committee. Dr. Uluer has served in an advisory capacity with Vertex and Eloxx but these relationships are in no way associated with the work submitted in this manuscript, (Crown Copyright © 2021. Published by Elsevier B.V. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
37. Healthcare resource utilization and costs among children with cystic fibrosis in the United States.
- Author
-
Thorat T, McGarry LJ, Bonafede MM, Limone BL, Rubin JL, Jariwala-Parikh K, and Konstan MW
- Subjects
- Adolescent, Child, Child, Preschool, Cross-Sectional Studies, Health Care Costs, Hospitalization, Humans, Patient Acceptance of Health Care, Retrospective Studies, United States epidemiology, Cystic Fibrosis epidemiology, Cystic Fibrosis therapy
- Abstract
Background: Adverse health impacts of cystic fibrosis (CF) can be present in children before respiratory complications are observed. Children with CF show progressive health decline, with increasing lung function decline in adolescence. This study aims to quantify the healthcare resource utilization (HCRU) and costs attributable to CF by comparing children with CF with the general pediatric population., Methods: This retrospective, cross-sectional, observational study compared HCRU and costs among children with CF in the US with demographically similar children without CF (comparison group) over a 12-month period using administrative claims data spanning 2010-2017. Analyses were conducted by insurance type (commercially insured [COM] and Medicaid insured [MED]) and stratified by age (<2 years, 2 to <6 years, 6 to <12 years, and 12-17 years)., Results: Children with CF (2831 COM and 1896 MED) were matched to children in the comparison group (8493 COM and 5688 MED). Higher prevalence of comorbidities was seen in children with CF versus the comparison group across all ages. Across all ages, HCRU attributable to CF was substantial (higher hospitalization rates, more outpatient and emergency room visits, and greater use of prescription medications), and there were higher associated costs (all p values < .05), in COM and MED populations. HCRU and costs attributable to CF were highest for children aged 12-17 years., Conclusions: Substantial HCRU and costs are evident among children with CF across all ages, starting as young as infancy, with highest HCRU and costs among adolescents. Effective treatments from an early age are needed for children with CF., (© 2021 The Authors. Genetic Epidemiology Published by Wiley Periodicals LLC.)
- Published
- 2021
- Full Text
- View/download PDF
38. Evaluating the Impact of Stopping Chronic Therapies after Modulator Drug Therapy in Cystic Fibrosis: The SIMPLIFY Clinical Trial Study Design.
- Author
-
Mayer-Hamblett N, Nichols DP, Odem-Davis K, Riekert KA, Sawicki GS, Donaldson SH, Ratjen F, Konstan MW, Simon N, Rosenbluth DB, Retsch-Bogart G, Clancy JP, VanDalfsen JM, Buckingham R, and Gifford AH
- Subjects
- Aminophenols therapeutic use, Benzodioxoles therapeutic use, Cystic Fibrosis Transmembrane Conductance Regulator genetics, Humans, Cystic Fibrosis drug therapy, Quinolones
- Abstract
The care for individuals with cystic fibrosis (CF) with at least one F508del mutation will greatly change as a result of the unparalleled clinical benefits observed with the new triple-combination CFTR (CF transmembrane regulator)-modulator therapy elexacaftor/tezacaftor/ivacaftor (ETI). Incorporating ETI into the standard of care creates new motivation and opportunity to consider reductions in overall treatment burden and evaluate whether other chronic medications can now be safely discontinued without loss of clinical benefit. SIMPLIFY is a master protocol poised to test the impact of discontinuing versus continuing two commonly used chronic therapies in people with CF who are at least 12 years of age or older and stable on ETI therapy. The protocol is composed of two concurrent randomized controlled trials designed to evaluate the independent short-term effects of discontinuing hypertonic saline or dornase alfa, enabling individuals on both therapies to participate in one or both trials. The primary objective for each trial is to determine whether discontinuing treatment is noninferior to continuing treatment after establishment of ETI, as measured by the 6-week absolute change in the percent-predicted forced expiratory volume in 1 second. Developing this study required a balance between ideal study-design principles and feasibility. SIMPLIFY will be the largest multicenter, randomized, controlled medication-withdrawal study in CF. This study is uniquely positioned to provide timely evidence on whether the daily treatment burden can be reduced among individuals on CFTR-modulator therapy. Clinical trial registered with www.clinicaltrials.gov (NCT04378153).
- Published
- 2021
- Full Text
- View/download PDF
39. Long-Term Impact of Ivacaftor on Healthcare Resource Utilization Among People with Cystic Fibrosis in the United States.
- Author
-
Thorat T, McGarry LJ, Jariwala-Parikh K, Limone B, Bonafede M, Chandarana K, and Konstan MW
- 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.
- Published
- 2021
- Full Text
- View/download PDF
40. Epidemiologic Study of Cystic Fibrosis: 25 years of observational research.
- Author
-
Konstan MW, Pasta DJ, VanDevanter DR, Wagener JS, and Morgan WJ
- Subjects
- Administration, Intravenous, Deoxyribonuclease I therapeutic use, Humans, Lung, Observational Studies as Topic, Prospective Studies, Respiratory Function Tests, United States epidemiology, Cystic Fibrosis drug therapy, Cystic Fibrosis epidemiology
- Abstract
The Epidemiologic Study of Cystic Fibrosis (ESCF) was a prospective observational study of over 32,000 people with cystic fibrosis (CF) from 250 clinical care sites in North America from 1994 to 2005. Begun as a pharmacovigilance study in connection with the approval of dornase alfa in 1993, ESCF was open to all people with CF treated at any participating site in the United States or Canada. In addition to obtaining safety and effectiveness data on dornase alfa, ESCF collected encounter-based data to characterize the natural history and management of CF with a special focus on lung disease. During the study, 32,178 patients reported at least one encounter, contributing 869,136 encounters, 622,592 pulmonary function tests, 432,896 cultures, and 118,563 pulmonary exacerbations treated with intravenous antibiotics. Although ESCF data collection concluded in 2005, through a collaboration with the U.S. Cystic Fibrosis Foundation Patient Registry, additional follow-up data through 2017 was available for two-thirds of patients. This allowed for updating of CF genotype and survival information. Fifty-six peer-reviewed publications (cited over 3600 times) resulted from this study. In this manuscript we summarize the published ESCF manuscripts in thematic groups with key study findings and brief comments, and speculate on how ESCF findings will inform future data registries and patient care practices., (© 2021 Wiley Periodicals LLC.)
- Published
- 2021
- Full Text
- View/download PDF
41. Pseudomonas aeruginosa antimicrobial susceptibility test (AST) results and pulmonary exacerbation treatment responses in cystic fibrosis.
- Author
-
VanDevanter DR, Heltshe SL, Hilliard JB, and Konstan MW
- Subjects
- Adolescent, Adult, Cystic Fibrosis physiopathology, Female, Humans, Male, Microbial Sensitivity Tests, Pseudomonas aeruginosa, Respiratory Function Tests, Symptom Flare Up, Anti-Bacterial Agents therapeutic use, Cystic Fibrosis complications, Pseudomonas Infections drug therapy
- Abstract
Background Antimicrobial susceptibility testing (AST) of bacterial isolates is a time- and resource-intensive procedure recommended by cystic fibrosis (CF) treatment guidelines for antimicrobial selection for pulmonary exacerbation (PEx) treatment. Methods We studied relationships between Pseudomonas aeruginosa (Pa) isolate AST results, antipseudomonal PEx treatments, and treatment responses as change in weight and percent predicted forced expiratory volume in 1 s (ppFEV
1 ) as well as future antimicrobial treatment hazard for PEx occurring at a CF care center from 1999 through 2018. Treatments were categorized by "Pa coverage" as complete (all Pa isolates susceptible by AST to at least one administered agent), none (no isolates susceptible), incomplete (some, but not all isolates susceptible), and indeterminant (administered antipseudomonals not evaluated by AST). Weight and ppFEV1 responses were compared across Pa coverage categories using unadjusted and adjusted general estimating equations; hazard of future treatment was assessed by Cox and logistic regression. Results Among 3820 antimicrobial PEx treatment events in 413 patients with Pa, 62.6% (2390) had complete Pa coverage; 8.9% (340), 2.4% (99), and 26.2% (1000), had no, incomplete, and indeterminant Pa coverage, respectively. Mean baseline to follow-up weight change was +0.74 kg [95% CI 0.63, 0.86]; ppFEV1 change was +1.60 [1.29, 1.90]. Pa coverage category was not associated with significant differences in weight or ppFEV1 change or with future antimicrobial treatment hazard. Conclusions We did not observe superior responses for AST-defined complete Pa coverage treatments versus lesser coverage treatments, suggesting that AST may be of little utility in choosing antimicrobials for CF PEx treatment., Competing Interests: Declaration of Competing Interest None., (Copyright © 2020 European Cystic Fibrosis Society. Published by Elsevier B.V. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
42. Evaluating assumptions of definition-based pulmonary exacerbation endpoints in cystic fibrosis clinical trials.
- Author
-
VanDevanter DR, Hamblett NM, Simon N, McIntosh J, and Konstan MW
- Subjects
- Adolescent, Adult, Anti-Bacterial Agents therapeutic use, Child, Clinical Trials as Topic, Cystic Fibrosis therapy, Female, Humans, Male, Middle Aged, Retrospective Studies, Young Adult, Cystic Fibrosis diagnosis, Disease Progression
- Abstract
Background: Cystic fibrosis (CF) pulmonary exacerbations can be serious respiratory events and reduction in exacerbation rate or risk are important efficacy endpoints for CF therapeutic trials. Variability in exacerbation diagnoses and treatment have led drug developers to employ "objective" exacerbation definitions combining antimicrobial treatment (AT) and the presence of ≥4 of 12 respiratory criteria (first published by Fuchs et al. [NEJM 1994;331(10):637-42]). Assumptions underlying this approach have yet to be formally evaluated., Methods: Respiratory events (RE) observed during a 48-week trial of ataluren (NCT02139306), a read-through agent for premature nonsense codons, were compared across six exacerbation definitions: any AT, intravenous AT (IVAT), ≥4 Fuchs criteria present, AT plus ≥4 Fuchs criteria, IVAT plus ≥4 Fuchs criteria, and investigator assessment. Fuchs definitions were evaluated by assessing missingness of individual criteria and associations between criteria presence and clinician exacerbation assessment., Results: Among 751 RE, more than one third had ≥4 Fuchs criteria present but were not assessed as exacerbations by investigators. Data for ≥1 and for 4 Fuchs criteria, respectively, were missing for ~ 90% and >30% of RE. Only 6/12 Fuchs criteria were present more often when investigators assessed RE as exacerbations than when they did not., Conclusions: "Objective" definitions have shortcomings inconsistent with their purpose of optimizing exacerbation capture in clinical trials : 1) they capture events clinicians do not consider exacerbations, 2) are prone to data missingness which can bias the likelihood of meeting the definition, and 3) employ criteria that are not associated with investigator assessment of exacerbation., Competing Interests: Declaration of Competing Interest DRV reports personal fees from AbbVie, Aradigm, Arrevus, Calithera, Cystic Fibrosis Foundation, Chiesi, Eloxx, Enbiotix, Galephar, Matinas, Armata, Horizon, IBF, Ionis, Kala, Microbion, Recida, Respirion, Vast, and aMoon, outside the submitted work. NMH reports personal fees from Calithera and Kala outside the submitted work. NS has nothing to disclose. JM was an employee of PTC Therapeutics during the conduct of the study. MWK reports grants and personal fees from Anthera, AzurRx, Corbus Pharmaceuticals, Laurent Pharmaceuticals, PTC Therapeutics, Savara, and Vertex Pharmaceuticals, as well as personal fees from Albumedix, Celtaxsys, Chiesi, Genentech, Kala, Novartis, Pharmaceuticals, Merck, Paranta Biosciences, pH Pharma, Protalix Biotherapeutics, and Santhera and grants from the Cystic Fibrosis Foundation and the National Institutes of Health outside the submitted work., (Copyright © 2020. Published by Elsevier B.V.)
- Published
- 2021
- Full Text
- View/download PDF
43. Building global development strategies for cf therapeutics during a transitional cftr modulator era.
- Author
-
Mayer-Hamblett N, van Koningsbruggen-Rietschel S, Nichols DP, VanDevanter DR, Davies JC, Lee T, Durmowicz AG, Ratjen F, Konstan MW, Pearson K, Bell SC, Clancy JP, Taylor-Cousar JL, De Boeck K, Donaldson SH, Downey DG, Flume PA, Drevinek P, Goss CH, Fajac I, Magaret AS, Quon BS, Singleton SM, VanDalfsen JM, and Retsch-Bogart GZ
- Subjects
- Cystic Fibrosis genetics, Humans, Cystic Fibrosis drug therapy, Cystic Fibrosis Transmembrane Conductance Regulator drug effects, Drug Development organization & administration, International Cooperation
- Abstract
As CFTR modulator therapy transforms the landscape of cystic fibrosis (CF) care, its lack of uniform access across the globe combined with the shift towards a new standard of care creates unique challenges for the development of future CF therapies. The advancement of a full and promising CF therapeutics pipeline remains a necessary priority to ensure maximal clinical benefits for all people with CF. It is through collaboration across the global CF community that we can optimize the evaluation and approval process of new therapies. To this end, we must identify areas for which harmonization is lacking and for which efficiencies can be gained to promote ethical, feasible, and credible study designs amidst the changing CF care landscape. This article summarizes the counsel from core advisors across multiple international regions and clinical trial networks, developed during a one-day workshop in October 2019. The goal of the workshop was to identify, in consideration of the highly transitional era of CFTR modulator availability, the drug development areas for which global alignment is currently uncertain, and paths forward that will enable advancement of CF therapeutic development., Competing Interests: Declaration of Competing Interest NMH serves as a consultant through her institution in her role as Executive Director of the CF Therapeutics Development Network Coordinating Center (CF TDNCC) and has received personal consulting fees from Kala Pharmaceuticals and Calithera. She has received grant funding from the Cystic Fibrosis Foundation (CFF) and National Institutes of Health (NIH). SvKR has received personal consulting fees from Antabio, Proteostasis Therapeutics (PTI), and Vertex Pharmaceuticals (VRTX). She has received grant support to her institution for her participation in the European Union HORIZON 2020 work. DPN serves as a consultant through his institution in his role as Medical Director of CF TDNCC. He has received grant support to his Institution from the CFF and Gilead Sciences. DRV has received personal consulting fees from AbbVie, Albumedix, AN2, Aradigm, Armata, Arrevus, Calithera, Chiesi USA, Cipla, Corbus, CFF, Eloxx, Enbiotix, Eveo, Galephar, Horizon, IBF, ICON clinical sciences, Ionis, Kala, Merck, Microbion, NDA, Protalix, PTC, Pulmocide, Recida, Savara, Vast, and VRTX. JCD has received other support from Aligipharma AS, Bayer AG, BI, Galapagos NV, ImevaX GmbH, Nivalis Therapeutics, ProQR Therapeutics, PTI, Raptor Pharamceuticals, VRTX, Enterprise, Novartis, Pulmocide, Flately and Teva for advisory board and educational activities. She has received grant support from the CF Trust. TL has received personal fees from Alan Boyd Consultants, Ltd for his participation in DSMB activities. FAR has received personal consulting fees from Novartis, Bayer, Roche, and Genentech for participation in CF related consulting activities. He has received grant support to his institution from VRTX for his participation as site PI in multicenter trials which they have funded as well as personal consulting fees. MWK has received personal consulting fees for advisory board participation, grant support to his institution for clinical trial participation, and non-financial support from VRTX, Savara, Laurent, Corbus Pharmaceuticals, PTC, and AzurRx. He has received personal consulting fees and non-financial support from Chiesi, Celtaxsys, Merck, and Kala. Personal consulting fees were received from Albumedix, Paranta, Protalix, Santhera, pH Pharma, Novartis, Ionis, the Italian Cystic Fibrosis Foundation, and the Food and Drug Administration. Grant support was provided to his Institution by NIH. Grant support was provided to his Institution by Anthera as well as personal consulting fees. SCB has received support from VRTX, Galapagos, and AbbVie for his participation in advisory boards and as site PI in multicenter trials which they have funded. JLTC has received personal consulting fees from Gilead Sciences, Protalix, and Santhera for participation in advisory board activities. She has received grant support to her institution from PTI, Celtaxsys, and VRTX as well as personal consulting fees for advisory activities. Grant support to her institution for her participation as site PI in a multicenter trial which they have funded from Eloxx. KDB has received consulting fees from Boehringer-Ingelheim (BI), Protalix Biotherapeutics, Raptor Pharmaceuticals, Novabiotics, Eloxx Pharmaceutics, Galapagos, and Chiesi. She has received speaker fees from Teva Pharmaceutical Industries and serves on the Steering Committee and Advisory Board for VRTX. DGD has received consulting fees from VRTX and consulting fees as well as grant support from PTI and Chiesi. PAF has received personal consulting fees from the Food and Drug Administration, Polyphor, and Santhera. He has received personal consulting fees and grant support from CFF, PTI, Savara, and VRTX. He has received grant support from NIH, Novartis, Novoteris, and Sound Pharmaceuticals. PD has received personal consulting fees from VRTX, PTI, and Actelion Pharmaceuticals. He has also received support for his participation as site PI in multicenter trials funded by Corbus Pharmaceuticals and VRTX. CHG has received grant funding from CFF, NIH, the European Commission and the Food and Drug Administration. He has also received honoraria from Gilead Sciences for grant reviews, honoraria from VRTX and Mylan for invited talks, and BI for participation as a PI in a multicenter trial in CF that they have funded. IF has received consulting fees and support from PTI, VRTX and BI for her participation in advisory boards as well as site PI in multicenter trials which they have funded. She has received support from Corbus Pharmaceuticals for participation as site PI in multicenter trials which they have funded ASM has received grant support to her Institution from CFF. BSQ has received grant support to his institution from the Cystic Fibrosis Canada, CFF, Michael Smith Foundation for Health Research, BC Lung Association, and Gilead Sciences. GZRB's institution has received support from the CFF, NIH and VRTX. for his participation as site PI in multicenter trials which they have funded. AGD, KP, JPC, SHD, SMS, and JMVD have nothing to disclose., (Copyright © 2020. Published by Elsevier B.V.)
- Published
- 2020
- Full Text
- View/download PDF
44. Reopening Schools Safely: The Case for Collaboration, Constructive Disruption of Pre-Coronavirus 2019 Expectations, and Creative Solutions.
- Author
-
Cooper DM, Guay-Woodford L, Blazar BR, Bowman S, Byington CL, Dome J, Forthal D, Konstan MW, Kuppermann N, Liem RI, Ochoa ER Jr, Pollock BH, Price OA, Ramsey BW, Ross LF, Sokol RJ, and Wright RJ
- Subjects
- Adolescent, COVID-19, Child, Coronavirus Infections diagnosis, Humans, Pneumonia, Viral diagnosis, SARS-CoV-2, United States, Betacoronavirus, Child Welfare, Coronavirus Infections prevention & control, Creativity, Intersectoral Collaboration, Pandemics prevention & control, Pneumonia, Viral prevention & control, Safety, Schools
- Published
- 2020
- Full Text
- View/download PDF
45. Efficacy and safety of ataluren in patients with nonsense-mutation cystic fibrosis not receiving chronic inhaled aminoglycosides: The international, randomized, double-blind, placebo-controlled Ataluren Confirmatory Trial in Cystic Fibrosis (ACT CF).
- Author
-
Konstan MW, VanDevanter DR, Rowe SM, Wilschanski M, Kerem E, Sermet-Gaudelus I, DiMango E, Melotti P, McIntosh J, and De Boeck K
- Subjects
- Administration, Oral, Double-Blind Method, Female, Forced Expiratory Volume, Humans, Male, Respiratory Function Tests methods, Symptom Flare Up, Treatment Outcome, Codon, Nonsense, Cystic Fibrosis drug therapy, Cystic Fibrosis genetics, Cystic Fibrosis physiopathology, Cystic Fibrosis Transmembrane Conductance Regulator genetics, Drug Monitoring methods, Oxadiazoles administration & dosage, Oxadiazoles adverse effects
- Abstract
Background: Ataluren was developed for potential treatment of nonsense-mutation cystic fibrosis (CF). A previous phase 3 ataluren study failed to meet its primary efficacy endpoint, but post-hoc analyses suggested that aminoglycosides may have interfered with ataluren's action. Thus, this subsequent trial (NCT02139306) was designed to assess the efficacy and safety of ataluren in patients with nonsense-mutation CF not receiving aminoglycosides., Methods: Eligible subjects with nonsense-mutation CF (aged ≥6 years; percent predicted (pp) FEV
1 ≥40 and ≤90) from 75 sites in 16 countries were randomly assigned in double-blinded fashion to receive oral ataluren or matching placebo thrice daily for 48 weeks. The primary endpoint was absolute change in average ppFEV1 from baseline to the average of Weeks 40 and 48., Findings: 279 subjects were enrolled; 138 subjects in the ataluren arm and 136 in the placebo arm were evaluable for efficacy. Absolute ppFEV1 change from baseline did not differ significantly between the ataluren and placebo groups at Week 40 (-0.8 vs -1.8) or Week 48 (-1.7 vs -2.4). Average ppFEV1 treatment difference from baseline to Weeks 40 and 48 was 0.6 (95% CI -1.3, 2.5; p = 0.54). Pulmonary exacerbation rate per 48 weeks was not significantly different (ataluren 0.95 vs placebo 1.13; rate ratio p = 0.40). Safety was similar between groups. No life-threatening adverse events or deaths were reported., Interpretation: Neither ppFEV1 change nor pulmonary exacerbation rate over 48 weeks were statistically different between ataluren and placebo groups. Development of a nonsense-mutation CF therapy remains elusive., Competing Interests: Declaration of Competing Interest JM is an employee of PTC Therapeutics, the funder of this clinical trial, and holds financial interests in the company. MWK, SMR, and DRV received compensation for consultant services from PTC Therapeutics prior to and/or during this study. EK, MW, IS-G, and KDB received compensation for travel expenses for meetings related to the study. All other authors declare no competing interests., (Copyright © 2020. Published by Elsevier B.V.)- Published
- 2020
- Full Text
- View/download PDF
46. Lung function changes before and after pulmonary exacerbation antimicrobial treatment in cystic fibrosis.
- Author
-
Wagener JS, VanDevanter DR, Konstan MW, Pasta DJ, Millar SJ, and Morgan WJ
- Subjects
- Adolescent, Adult, Child, Disease Progression, Female, Forced Expiratory Volume, Humans, Lung physiopathology, Male, Young Adult, Anti-Infective Agents therapeutic use, Cystic Fibrosis drug therapy, Cystic Fibrosis physiopathology
- Abstract
Background: In cystic fibrosis, observation of a lung function drop (as percent predicted forced expiratory volume in 1 s [FEV
1 ]; ppFEV1 ) frequently precedes pulmonary exacerbation (PEx) diagnosis. Recovery of ppFEV1 to a previous "baseline" is commonly used to assess antimicrobial treatment response. However, not all diagnosed PEx are associated with a ppFEV1 drop, and it is unclear whether these are a different type of PEx from those associated with a ppFEV1 drop., Methods: We analyzed pre- and posttreatment ppFEV1 for PEx recorded in the Epidemiologic Study of Cystic Fibrosis from 2003 through 2005. Baseline, pretreatment, and follow-up ppFEV1 were the best recorded within 12-months pre-PEx, the lowest recorded -30 to +3 days of treatment, and the best recorded during 6-month follow-up, respectively. Logistic regression models for return of ppFEV1 to baseline during follow-up were developed separately for PEx with ≥10%, <10%, and no ppFEV1 drop before treatment., Results: Of 15 147 PEx, 10 166 (67.1%), 3479 (23.0%), and 1502 (9.9%) presented with a ≥10%, <10%, or no ppFEV1 drop at diagnosis, respectively. 19.5%, 35.2%, and 65.6% of PEx, respectively, had follow-up ppFEV1 equal to or exceeding baseline; overall 27.7% of all PEx treatments resulted in complete recovery of baseline ppFEV1 . Significant predictors of ppFEV1 recovery at follow-up were younger patient age, absence of Aspergillus, lower baseline ppFEV1 , fewer visits during the baseline, lower frequency of prior-year PEx, shorter elapsed time from baseline measure to treatment, smaller relative ppFEV1 drop before treatment, and non intravenous (ie, oral or inhaled antibiotic) treatment. PEx with ≥10%, <10%, and no ppFEV1 drop before treatment had only modest differences in covariate odds ratios associated with complete ppFEV1 recovery., Conclusions: Among the 10% of PEx presenting with no apparent ppFEV1 drop, more than one-third resulted in a decreased ppFEV1 during follow-up. Risk factors for this outcome were the same as those associated with lack of ppFEV1 recovery among PEx with pretreatment ppFEV1 drops. These results suggest that inherent FEV1 variability, baseline and follow-up sampling methodologies, ppFEV1 regression to the mean, and underlying lung disease progression complicate this approach for assessing effects of PEx and treatment response., (© 2019 Wiley Periodicals, Inc.)- Published
- 2020
- Full Text
- View/download PDF
47. Disease progression in patients with cystic fibrosis treated with ivacaftor: Data from national US and UK registries.
- Author
-
Volkova N, Moy K, Evans J, Campbell D, Tian S, Simard C, Higgins M, Konstan MW, Sawicki GS, Elbert A, Charman SC, Marshall BC, and Bilton D
- Subjects
- Adult, Chloride Channel Agonists therapeutic use, Cystic Fibrosis Transmembrane Conductance Regulator genetics, Female, Hospitalization statistics & numerical data, Humans, Longitudinal Studies, Male, Registries statistics & numerical data, United Kingdom epidemiology, United States epidemiology, Aminophenols therapeutic use, Cystic Fibrosis drug therapy, Cystic Fibrosis epidemiology, Cystic Fibrosis microbiology, Cystic Fibrosis physiopathology, Disease Progression, Pseudomonas aeruginosa isolation & purification, Quinolones therapeutic use, Respiratory Function Tests methods, Respiratory Function Tests statistics & numerical data
- Abstract
Background: Ivacaftor is the first in a class of drugs, CFTR modulators, that target the underlying defect in cystic fibrosis (CF). This long-term observational safety study evaluated CF disease progression in patients treated with ivacaftor in a real-world setting for up to 5 years., Methods: Data from existing US and UK CF patient registries were used to assess longitudinal patterns in lung function, nutritional status, pulmonary exacerbations and hospitalizations, CF-related diabetes (CFRD), and Pseudomonas aeruginosa in ivacaftor-treated vs untreated comparator cohorts matched by age, sex, and disease severity., Results: US analyses included 635 ivacaftor-treated patients and 1874 comparators followed for 5 years from year 1 of market availability (2012-2016). Evaluation of outcome patterns from pretreatment baseline (2011) through year 5 (2016), showed that relative to comparators, ivacaftor-treated patients had better preserved lung function (mean change in percent predicted FEV
1 , -0.7 percentage points with ivacaftor vs -8.3 percentage points in comparators) and improved nutritional status (mean body mass index change +2.4 kg/m2 with ivacaftor vs +1.6 kg/m2 in comparators). US patients treated with ivacaftor had significantly lower frequencies of exacerbations and hospitalizations in each of the 5 years of follow-up relative to pretreatment baseline and comparators. Favorable trends in CFRD and P. aeruginosa prevalence were also observed. Findings from the smaller UK registry were directionally similar to and consistent with US findings., Conclusions: This observational study represents the largest longitudinal analysis of patients treated with ivacaftor in a real-world setting. The findings support disease modification by CFTR modulation with ivacaftor., Competing Interests: Conflict of interest statement All authors received nonfinancial support (assistance with manuscript preparation) from ArticulateScience LLC, which received funding from Vertex Pharmaceuticals. Additional disclosures are as follows: NV, KM, JE, DC, ST, CS, and MH: employee of Vertex Pharmaceuticals and may own stock or stock options in Vertex Pharmaceuticals. MWK: grants, personal fees, and nonfinancial support from Vertex Pharmaceuticals, during the course of the study; grants and nonfinancial support from the Cystic Fibrosis Foundation (CFF); grants, personal fees, and nonfinancial support from Corbus, Laurent, PTC, and Savara; personal fees and nonfinancial support from Chiesi, Celtaxsys, Genentech, and Merck; personal fees from Albumedix, Anthera, Ionis, Paranta, Protalix, and Santhera outside the submitted work. GSS: grants and personal fees from Vertex Pharmaceuticals outside the submitted work. AE and BCM: other support from several pharmaceutical companies during the conduct of the study (The CFF has entered into therapeutic development award agreements and licensing agreements to assist with the development of CFTR modulators that may result in intellectual property rights, royalties and other fees provided to CFF by various pharmaceutical companies). SCC: service agreement between Vertex Pharmaceuticals and Cystic Fibrosis Services Limited for statistical analysis. DB: member of the Steering Committee of the UK CF Registry, which provided data for this study., (Copyright © 2019 The Authors. Published by Elsevier B.V. All rights reserved.)- Published
- 2020
- Full Text
- View/download PDF
48. Clinical care for cystic fibrosis: preparing for the future now.
- Author
-
Konstan MW and Flume PA
- Subjects
- Humans, Cystic Fibrosis
- Published
- 2020
- Full Text
- View/download PDF
49. Lumacaftor/Ivacaftor reduces pulmonary exacerbations in patients irrespective of initial changes in FEV 1 .
- Author
-
McColley SA, Konstan MW, Ramsey BW, Stuart Elborn J, Boyle MP, Wainwright CE, Waltz D, Vera-Llonch M, Marigowda G, Jiang JG, and Rubin JL
- Subjects
- Adolescent, Adult, Aged, Child, Cystic Fibrosis physiopathology, Disease Progression, Double-Blind Method, Drug Combinations, Female, Follow-Up Studies, Forced Expiratory Volume drug effects, Humans, Lung drug effects, Male, Middle Aged, Respiratory Function Tests, Retrospective Studies, Treatment Outcome, Young Adult, Aminophenols therapeutic use, Aminopyridines therapeutic use, Benzodioxoles therapeutic use, Cystic Fibrosis drug therapy, Forced Expiratory Volume physiology, Lung physiopathology, Quinolones therapeutic use
- Abstract
Background: Improved lung function and fewer pulmonary exacerbations (PEx) were observed with lumacaftor/ivacaftor (LUM/IVA) in patients with cystic fibrosis homozygous for F508del. It is unknown whether PEx reduction extends to patients without early lung function improvement., Methods: Post hoc analyses of pooled phase 3 data (NCT01807923, NCT01807949) categorized LUM/IVA-treated patients by percent predicted forced expiratory volume in 1 s (ppFEV
1 ) change from baseline to day 15 into threshold categories (absolute change ≤0 vs >0; relative change <5% vs ≥5%) and compared PEx rates vs placebo., Results: LUM (400 mg q12h)/IVA (250 mg q12h)-treated patients (n = 369) experienced significantly fewer PEx vs placebo, regardless of threshold category. With LUM/IVA, PEx rate per patient per year was 0.60 for those with absolute change in ppFEV1 > 0 and 0.85 for those with absolute change ≤0 (respective rate ratios vs placebo [95% CI]: 0.53 [0.40-0.69; P < .0001], 0.74 [0.55-0.99; P = .04])., Conclusions: LUM/IVA significantly reduced PEx, even in patients without early lung function improvement., (Copyright © 2018 The Author(s). Published by Elsevier B.V. All rights reserved.)- Published
- 2019
- Full Text
- View/download PDF
50. Modeling long-term health outcomes of patients with cystic fibrosis homozygous for F508del-CFTR treated with lumacaftor/ivacaftor.
- Author
-
Rubin JL, O'Callaghan L, Pelligra C, Konstan MW, Ward A, Ishak JK, Chandler C, and Liou TG
- Subjects
- Adolescent, Adult, Child, Cystic Fibrosis genetics, Cystic Fibrosis Transmembrane Conductance Regulator genetics, Drug Combinations, Female, Humans, Male, Mutation, Randomized Controlled Trials as Topic, Registries, Survival Rate, Time Factors, Treatment Outcome, Young Adult, Aminophenols therapeutic use, Aminopyridines therapeutic use, Benzodioxoles therapeutic use, Computer Simulation, Cystic Fibrosis drug therapy, Models, Statistical, Quinolones therapeutic use
- Abstract
Background: Lumacaftor/ivacaftor combination therapy is efficacious and generally safe for patients with cystic fibrosis (CF) homozygous for the F508del-CF transmembrane conductance regulator (CFTR) mutation. However, long-term survival benefits of lumacaftor/ivacaftor (LUM/IVA) cannot yet be quantified. Simulation models can provide predictions about long-term health outcomes. In this study, we aimed to project long-term health outcomes of LUM/IVA plus standard care (SC) in patients with CF homozygous for F508del-CFTR., Methods: This modeling study was an individual patient simulation in US patients aged ⩾6 years with CF, homozygous for F508del-CFTR. The primary outcome was projected survival among (a) a cohort of patients who ever initiated LUM/IVA, accounting for treatment discontinuations, and (b) a cohort of patients who remain on continuous LUM/IVA. Patient characteristics and model parameters were derived from clinical trials: VX14-809-109, VX13-809-011B, TRAFFIC/TRANSPORT, and PROGRESS; published literature; and the US CF Foundation Patient Registry., Results: Lumacaftor/ivacaftor + SC is expected to increase median survival by 6.1 years versus SC alone, accounting for treatment discontinuations. The incremental median predicted survival versus SC assuming initiation of LUM/IVA at ages 6, 12, 18, and 25 years was 17.7, 12.6, 8.0, and 3.8 years, respectively. Assuming lifetime treatment with LUM/IVA, incremental median survival was predicted to be 7.8 years longer in the LUM/IVA + SC cohort. Initiating LUM/IVA at ages 6, 12, 18, and 25 years and assuming lifetime treatment resulted in incremental median predicted survival of 23.4, 18.2, 11.0, and 4.8 years, respectively., Conclusions: Lumacaftor/ivacaftor is projected to increase survival for patients with CF. Initiation at an early age and treatment persistence result in further increments in projected survival.
- Published
- 2019
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.