Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 10.676
2.
Eur Respir J ; 63(6)2024 Jun.
Article En | MEDLINE | ID: mdl-38697648

BACKGROUND: Elevated markers of systemic and pulmonary inflammation are associated with failure to recover lung function following pulmonary exacerbations in people with cystic fibrosis (pwCF). Our aim was to determine whether adjuvant oral prednisone treatment would improve recovery of forced expiratory volume in 1 s (FEV1) % pred in CF pulmonary exacerbations not responding to antibiotic therapy. METHODS: This was a randomised, double-blind, placebo-controlled trial in pwCF treated with intravenous antibiotics for a pulmonary exacerbation. At day 7, those who had not returned to >90% baseline FEV1 % pred were randomised to adjuvant prednisone 1 mg·kg-1 twice daily (maximum 60 mg·day-1) or placebo for 7 days. The primary outcome was the difference in proportion of subjects who recovered >90% baseline FEV1 % pred at day 14 of i.v. antibiotic therapy. RESULTS: 173 subjects were enrolled, with 76 randomised. 50% of subjects in the prednisone group recovered baseline FEV1 on day 14 compared with 39% of subjects in the placebo group (difference of 11%, 95% CI -11-34%; p=0.34). The mean±sd change in FEV1 % pred from day 7 to day 14 was 6.8±8.8% predicted in the prednisone group and 4.6±6.9% predicted in the placebo group (mean difference 2.2% predicted, 95% CI -1.5-5.9%; p=0.24). Time to subsequent exacerbation was not prolonged in prednisone-treated subjects (hazard ratio 0.83, 95% CI 0.45-1.53; p=0.54). CONCLUSIONS: This study failed to detect a difference in FEV1 % pred recovery between adjuvant oral prednisone and placebo treatment in pwCF not responding at day 7 of i.v. antibiotic therapy for pulmonary exacerbations.


Anti-Bacterial Agents , Cystic Fibrosis , Prednisone , Humans , Cystic Fibrosis/drug therapy , Cystic Fibrosis/physiopathology , Cystic Fibrosis/complications , Male , Female , Prednisone/administration & dosage , Prednisone/therapeutic use , Double-Blind Method , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/therapeutic use , Forced Expiratory Volume , Administration, Oral , Adult , Young Adult , Adolescent , Disease Progression , Treatment Outcome , Lung/physiopathology , Lung/drug effects
3.
Turk J Pediatr ; 66(2): 257-264, 2024 May 23.
Article En | MEDLINE | ID: mdl-38814300

BACKGROUND: Acute respiratory failure is a prevalent condition in childhood with a high rate of mortality. Invasive mechanical ventilation support may be required for the management of these patients. Extracorporeal membrane oxygenation (ECMO) is a method used when ventilation support is insufficient. However, the less invasive extracorporeal carbon dioxide removal method can be used as an alternative in cases of hypercapnic respiratory failure. CASE: A 9-year-old patient with cystic fibrosis presented to the hospital with acute respiratory failure due to pneumonia. Bilateral patchy areas of consolidation were evident in the chest x-ray. Invasive mechanical ventilation support was consequently provided to treat severe hypercapnia. Although peak and plateau pressure levels exceeded 32 cmH2O (49 cmH2O) and 28 cmH2O (35 cmH2O), respectively, the patient continued to have severe respiratory acidosis. Therefore extracorporeal carbon dioxide removal support was initiated to provide lung-protective ventilation. By Day 10, venovenous ECMO support was initiated due to deteriorating oxygenation. CONCLUSION: In cases where conventional invasive mechanical ventilation support is insufficient due to acute hypercapnic respiratory failure, extracorporeal carbon dioxide removal support, which is less invasive compared to ECMO, should be considered as an effective and reliable alternative method. However, it should be noted that extracorporeal carbon dioxide removal support does not affect oxygenation; it functions solely as a carbon dioxide removal system.


Carbon Dioxide , Cystic Fibrosis , Extracorporeal Membrane Oxygenation , Hypercapnia , Respiratory Insufficiency , Humans , Cystic Fibrosis/complications , Cystic Fibrosis/therapy , Respiratory Insufficiency/therapy , Respiratory Insufficiency/etiology , Extracorporeal Membrane Oxygenation/methods , Hypercapnia/therapy , Hypercapnia/etiology , Child , Male , Acute Disease , Respiration, Artificial/methods
4.
Zhongguo Dang Dai Er Ke Za Zhi ; 26(5): 506-511, 2024 May 15.
Article Zh | MEDLINE | ID: mdl-38802912

OBJECTIVES: To summarize the clinical characteristics and genetic variations in children with cystic fibrosis (CF) primarily presenting with pseudo-Bartter syndrome (CF-PBS), with the aim to enhance understanding of this disorder. METHODS: A retrospective analysis was performed on the clinical data of three children who were diagnosed with CF-PBS in Hunan Children's Hospital from January 2018 to August 2023, and a literature review was performed. RESULTS: All three children had the onset of the disease in infancy. Tests after admission showed hyponatremia, hypokalemia, hypochloremia, and metabolic alkalosis, and genetic testing showed the presence of compound heterozygous mutation in the CFTR gene. All three children were diagnosed with CF. Literature review obtained 33 Chinese children with CF-PBS, with an age of onset of 1-36 months and an age of diagnosis of 3-144 months. Among these children, there were 29 children with recurrent respiratory infection or persistent pneumonia (88%), 26 with malnutrition (79%), 23 with developmental retardation (70%), and 18 with pancreatitis or extrapancreatic insufficiency (55%). Genetic testing showed that c.2909G>A was the most common mutation site of the CFTR gene, with a frequency of allelic variation of 23% (15/66). CONCLUSIONS: CF may have no typical respiratory symptoms in the early stage. The possibility of CF-PBS should be considered for infants with recurrent hyponatremia, hypokalemia, hypochloremia, and metabolic alkalosis, especially those with malnutrition and developmental retardation. CFTR genetic testing should be performed as soon as possible to help with the diagnosis of CF.


Bartter Syndrome , Cystic Fibrosis Transmembrane Conductance Regulator , Cystic Fibrosis , Mutation , Humans , Cystic Fibrosis/genetics , Cystic Fibrosis/complications , Male , Female , Infant , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Bartter Syndrome/genetics , Bartter Syndrome/diagnosis , Bartter Syndrome/complications , Child, Preschool , Child , Retrospective Studies
5.
BMJ Open Respir Res ; 11(1)2024 May 03.
Article En | MEDLINE | ID: mdl-38702073

The major cause of mortality in people with cystic fibrosis (pwCF) is progressive lung disease characterised by acute and chronic infections, the accumulation of mucus, airway inflammation, structural damage and pulmonary exacerbations. The prevalence of Pseudomonas aeruginosa rises rapidly in the teenage years, and this organism is the most common cause of chronic lung infection in adults with cystic fibrosis (CF). It is associated with an accelerated decline in lung function and premature death. New P. aeruginosa infections are treated with antibiotics to eradicate the organism, while chronic infections require long-term inhaled antibiotic therapy. The prevalence of P. aeruginosa infections has decreased in CF registries since the introduction of CF transmembrane conductance regulator modulators (CFTRm), but clinical observations suggest that chronic P. aeruginosa infections usually persist in patients receiving CFTRm. This indicates that pwCF may still need inhaled antibiotics in the CFTRm era to maintain long-term control of P. aeruginosa infections. Here, we provide an overview of the changing perceptions of P. aeruginosa infection management, including considerations on detection and treatment, the therapy burden associated with inhaled antibiotics and the potential effects of CFTRm on the lung microbiome. We conclude that updated guidance is required on the diagnosis and management of P. aeruginosa infection. In particular, we highlight a need for prospective studies to evaluate the consequences of stopping inhaled antibiotic therapy in pwCF who have chronic P. aeruginosa infection and are receiving CFTRm. This will help inform new guidelines on the use of antibiotics alongside CFTRm.


Anti-Bacterial Agents , Cystic Fibrosis Transmembrane Conductance Regulator , Cystic Fibrosis , Pseudomonas Infections , Pseudomonas aeruginosa , Humans , Administration, Inhalation , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/therapeutic use , Cystic Fibrosis/complications , Cystic Fibrosis/microbiology , Cystic Fibrosis/drug therapy , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/isolation & purification , Pseudomonas Infections/drug therapy
7.
J Antimicrob Chemother ; 79(6): 1432-1440, 2024 Jun 03.
Article En | MEDLINE | ID: mdl-38708553

OBJECTIVES: Despite the introduction of cystic fibrosis transmembrane conductance regulator (CFTR) modulators, Pseudomonas aeruginosa is still a major pathogen in people with cystic fibrosis (pwCF). We determine the activity of cefiderocol and comparators in a collection of 154 P. aeruginosa isolates recovered from pwCF during three multicentre studies performed in 17 Spanish hospitals in 2013, 2017 and 2021. METHODS: ISO broth microdilution was performed and MICs were interpreted with CLSI and EUCAST criteria. Mutation frequency and WGS were also performed. RESULTS: Overall, 21.4% were MDR, 20.8% XDR and 1.3% pandrug-resistant (PDR). Up to 17% of the isolates showed a hypermutator phenotype. Cefiderocol demonstrated excellent activity; only 13 isolates (8.4%) were cefiderocol resistant by EUCAST (none using CLSI). A high proportion of the isolates resistant to ceftolozane/tazobactam (71.4%), meropenem/vaborbactam (70.0%), imipenem/relebactam (68.0%) and ceftazidime/avibactam (55.6%) were susceptible to cefiderocol. Nine out of 13 cefiderocol-resistant isolates were hypermutators (P < 0.001). Eighty-three STs were detected, with ST98 being the most frequent. Only one isolate belonging to the ST175 high-risk clone carried blaVIM-2. Exclusive mutations affecting genes involved in membrane permeability, AmpC overexpression (L320P-AmpC) and efflux pump up-regulation were found in cefiderocol-resistant isolates (MIC = 4-8 mg/L). Cefiderocol resistance could also be associated with mutations in genes related to iron uptake (tonB-dependent receptors and pyochelin/pyoverdine biosynthesis). CONCLUSIONS: Our results position cefiderocol as a therapeutic option in pwCF infected with P. aeruginosa resistant to most recent ß-lactam/ß-lactamase inhibitor combinations.


Anti-Bacterial Agents , Cefiderocol , Cephalosporins , Cystic Fibrosis , Microbial Sensitivity Tests , Pseudomonas Infections , Pseudomonas aeruginosa , Humans , Cystic Fibrosis/microbiology , Cystic Fibrosis/complications , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/isolation & purification , Pseudomonas Infections/microbiology , Spain/epidemiology , Anti-Bacterial Agents/pharmacology , Cephalosporins/pharmacology , Drug Resistance, Multiple, Bacterial/genetics , Adolescent , Adult , Child , Mutation , Tazobactam/pharmacology , Female , Male
8.
Respir Med ; 227: 107661, 2024 Jun.
Article En | MEDLINE | ID: mdl-38729529

Antibiotic-resistant bacteria associated with LRTIs are frequently associated with inefficient treatment outcomes. Antibiotic-resistant Streptococcus pneumoniae, Haemophilus influenzae, Pseudomonas aeruginosa, and Staphylococcus aureus, infections are strongly associated with pulmonary exacerbations and require frequent hospital admissions, usually following failed management in the community. These bacteria are difficult to treat as they demonstrate multiple adaptational mechanisms including biofilm formation to resist antibiotic threats. Currently, many patients with the genetic disease cystic fibrosis (CF), non-CF bronchiectasis (NCFB) and chronic obstructive pulmonary disease (COPD) experience exacerbations of their lung disease and require high doses of systemically administered antibiotics to achieve meaningful clinical effects, but even with high systemic doses penetration of antibiotic into the site of infection within the lung is suboptimal. Pulmonary drug delivery technology that reliably deliver antibacterials directly into the infected cells of the lungs and penetrate bacterial biofilms to provide therapeutic doses with a greatly reduced risk of systemic adverse effects. Inhaled liposomal-packaged antibiotic with biofilm-dissolving drugs offer the opportunity for targeted, and highly effective antibacterial therapeutics in the lungs. Although the challenges with development of some inhaled antibiotics and their clinicals trials have been studied; however, only few inhaled products are available on market. This review addresses the current treatment challenges of antibiotic-resistant bacteria in the lung with some clinical outcomes and provides future directions with innovative ideas on new inhaled formulations and delivery technology that promise enhanced killing of antibiotic-resistant biofilm-dwelling bacteria.


Anti-Bacterial Agents , Biofilms , Drug Delivery Systems , Respiratory Tract Infections , Humans , Biofilms/drug effects , Administration, Inhalation , Anti-Bacterial Agents/administration & dosage , Respiratory Tract Infections/drug therapy , Respiratory Tract Infections/microbiology , Drug Resistance, Bacterial , Streptococcus pneumoniae/drug effects , Liposomes , Bronchiectasis/drug therapy , Bronchiectasis/microbiology , Haemophilus influenzae/drug effects , Pulmonary Disease, Chronic Obstructive/drug therapy , Pseudomonas aeruginosa/drug effects , Staphylococcus aureus/drug effects , Cystic Fibrosis/microbiology , Cystic Fibrosis/drug therapy , Cystic Fibrosis/complications
10.
Respir Med ; 228: 107654, 2024 Jul.
Article En | MEDLINE | ID: mdl-38735372

BACKGROUND: Quality of life and survival in Cystic Fibrosis (CF) have improved dramatically, making family planning a feasible option. Maternal and perinatal outcomes in women with CF (wwCF) are similar to those seen in the general population. However, the effect of undergoing multiple pregnancies is unknown. METHODS: A multinational-multicenter retrospective cohort study. Data was obtained from 18 centers worldwide, anonymously, on wwCF 18-45 years old, including disease severity and outcome, as well as obstetric and newborn complications. Data were analyzed, within each individual patient to compare the outcomes of an initial pregnancy (1st or 2nd) with a multigravid pregnancy (≥3) as well as secondary analysis of grouped data to identify risk factors for disease progression or adverse neonatal outcomes. Three time periods were assessed - before, during, and after pregnancy. RESULTS: The study population included 141 wwCF of whom 41 (29%) had ≥3 pregnancies, "multiparous". Data were collected on 246 pregnancies, between 1973 and 2020, 69 (28%) were multiparous. A greater decline in ppFEV1 was seen in multiparous women, primarily in pancreatic insufficient (PI) wwCF and those with two severe (class I-III) mutations. Multigravid pregnancies were shorter, especially in wwCF over 30 years old, who had high rates of prematurity and newborn complications. There was no effect on pulmonary exacerbations or disease-related complications. CONCLUSIONS: Multiple pregnancies in wwCF are associated with accelerated respiratory deterioration and higher rates of preterm births. Therefore, strict follow-up by a multidisciplinary CF and obstetric team is needed in women who desire to carry multiple pregnancies.


Cystic Fibrosis , Pregnancy Outcome , Humans , Cystic Fibrosis/complications , Female , Pregnancy , Adult , Retrospective Studies , Young Adult , Infant, Newborn , Adolescent , Parity , Middle Aged , Pregnancy Complications/epidemiology , Disease Progression , Premature Birth/epidemiology , Pregnancy, Multiple , Severity of Illness Index , Risk Factors
11.
Respir Med ; 228: 107664, 2024 Jul.
Article En | MEDLINE | ID: mdl-38759874

BACKGROUND: Impaired glycemic control and the subsequent development of Cystic fibrosis Related Diabetes (CFRD) are prevalent complications, affecting up to 50 % of adults with cystic fibrosis (CF). CFTR modulator (CFTRm) therapies improve pulmonary functions, reduce exacerbation rates, increase survival in people with CF (pwCF) and appear to have a positive effect on extrapulmonary manifestations, such as nutritional state, improvements in upper respiratory symptoms, and quality of life. Initial findings indicate that CFTRm may have a positive impact on short-term glycemic control; however, long-term effects remain uncertain at present. METHODS: In this retrospective study, data were collected and analyzed on 15 pwCF, ages 13-37 years, started on CFTRm therapy. Oral Glucose Tolerance Test (OGTT) results were compared pre- and post-CFTRm therapy. RESULTS: The 120-min OGTT value decreased from 159.7 mg/dL to 130.4 mg/dL post-CFTRm (p = 0.047). The average time elapsed between the two OGTTs was 49.87 months (ranging 9-157 months, median 38 months). Glycemic status improved in six pwCF (two CFRD to normal (NGT)/indeterminate (INDET) glucose tolerance; two impaired glucose tolerance (IGT) to INDET; two INDET to NGT) and worsened in one (IGT to CFRD). Six pwCF and NGT remained stable with no changes in glycemic status throughout the follow-up period. CONCLUSIONS: CFTRm therapy may decelerate the glycemic control deterioration in pwCF over an extended period. These findings indicate the need for periodic OGTTs following the initiation of CFTRm therapy to appropriately adjust insulin requirements and prevent hypoglycemia. Further larger cohorts are required to authenticate and substantiate these findings.


Blood Glucose , Cystic Fibrosis Transmembrane Conductance Regulator , Cystic Fibrosis , Glucose Tolerance Test , Humans , Cystic Fibrosis/drug therapy , Cystic Fibrosis/physiopathology , Cystic Fibrosis/complications , Adolescent , Adult , Retrospective Studies , Male , Female , Young Adult , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Blood Glucose/metabolism , Blood Glucose/drug effects , Aminophenols/therapeutic use , Quinolones/therapeutic use , Aminopyridines/therapeutic use , Benzodioxoles/therapeutic use , Diabetes Mellitus/drug therapy , Diabetes Mellitus/metabolism , Glycemic Control/methods , Time Factors , Glucose/metabolism , Glucose Intolerance/drug therapy , Glucose Intolerance/metabolism
12.
BMC Oral Health ; 24(1): 446, 2024 Apr 12.
Article En | MEDLINE | ID: mdl-38609911

BACKGROUND: Oral health impacts systemic health, individual well-being, and quality of life. It is important to identify conditions that may exacerbate oral disease to aid public health and policy development and promote targeted patient treatment strategies. Developmental defects can increase an individual's risk of dental caries, hypersensitivity, premature tooth wear, erosion, and poor aesthetics. As part of an ongoing study assessing oral health in adults with cystic fibrosis at Cork University Dental School and Hospital, a systematic review of available literature was conducted to assess the prevalence of enamel defects in people with cystic fibrosis. AIMS: To critically evaluate the literature to determine if the prevalence of developmental defects of enamel is higher in people with cystic fibrosis (PwCF). METHODS: Data Sources: Three online databases were searched Embase, Scopus, and Web of Science Core Collection. Studies that examined an association between cystic fibrosis and developmental defects of enamel were included in this systematic review. RESULTS: The initial search identified 116 publications from the following databases Embase, Web of Science Core Collection, and Scopus. Eleven studies were included for qualitative analysis. Nine studies concluded that PwCF had a higher prevalence of enamel defects than control people and one study found no difference in cystic fibrosis (CF) status. All studies had a risk of bias that may influence study results and their interpretation. CONCLUSIONS: The results of the systematic review show a consistent pattern that PwCF have a higher prevalence of DDE than people without CF. Genetic dysfunction, chronic systemic infections, and long-term antibiotic use are possible aetiological causes. This review highlights the need for future studies to investigate if DDEs are caused by the underlying CFTR mutation or as a consequence of disease manifestations and/or management.


Cystic Fibrosis , Dental Caries , Developmental Defects of Enamel , Adult , Humans , Prevalence , Cystic Fibrosis/complications , Cystic Fibrosis/epidemiology , Quality of Life , Dental Enamel
13.
Sci Rep ; 14(1): 9056, 2024 04 20.
Article En | MEDLINE | ID: mdl-38643191

The impact of evolving treatment regimens, airway clearance strategies, and antibiotic combinations on the incidence and prevalence of respiratory infection in cystic fibrosis (CF) in children and adolescents remains unclear. The incidence, prevalence, and prescription trends from 2002 to 2019 with 18,339 airway samples were analysed. Staphylococcus aureus [- 3.86% (95% CI - 5.28-2.43)] showed the largest annual decline in incidence, followed by Haemophilus influenzae [- 3.46% (95% CI - 4.95-1.96)] and Pseudomonas aeruginosa [- 2.80%95% CI (- 4.26-1.34)]. Non-tuberculous mycobacteria and Burkholderia cepacia showed a non-significant increase in incidence. A similar pattern of change in prevalence was observed. No change in trend was observed in infants < 2 years of age. The mean age of the first isolation of S. aureus (p < 0.001), P. aeruginosa (p < 0.001), H. influenza (p < 0.001), Serratia marcescens (p = 0.006) and Aspergillus fumigatus (p = 0.02) have increased. Nebulised amikacin (+ 3.09 ± 2.24 prescription/year, p = 0.003) and colistin (+ 1.95 ± 0.3 prescriptions/year, p = 0.032) were increasingly prescribed, while tobramycin (- 8.46 ± 4.7 prescriptions/year, p < 0.001) showed a decrease in prescription. Dornase alfa and hypertonic saline nebulisation prescription increased by 16.74 ± 4.1 prescriptions/year and 24 ± 4.6 prescriptions/year (p < 0.001). There is a shift in CF among respiratory pathogens and prescriptions which reflects the evolution of cystic fibrosis treatment strategies over time.


Cystic Fibrosis , Pneumonia , Pseudomonas Infections , Child , Infant , Humans , Adolescent , Cystic Fibrosis/complications , Cystic Fibrosis/epidemiology , Cystic Fibrosis/microbiology , Staphylococcus aureus , Respiratory System/microbiology , Anti-Bacterial Agents/therapeutic use , Pseudomonas Infections/drug therapy , Pneumonia/drug therapy , Pseudomonas aeruginosa
14.
Int J Mol Sci ; 25(7)2024 Mar 27.
Article En | MEDLINE | ID: mdl-38612524

The interaction between extracellular vesicles (EVs) and SARS-CoV-2, the virus causing COVID-19, especially in people with cystic fibrosis (PwCF) is insufficiently studied. EVs are small membrane-bound particles involved in cell-cell communications in different physiological and pathological conditions, including inflammation and infection. The CF airway cells release EVs that differ from those released by healthy cells and may play an intriguing role in regulating the inflammatory response to SARS-CoV-2. On the one hand, EVs may activate neutrophils and exacerbate inflammation. On the other hand, EVs may block IL-6, a pro-inflammatory cytokine associated with severe COVID-19, and protect PwCF from adverse outcomes. EVs are regulated by TGF-ß signaling, essential in different disease states, including COVID-19. Here, we review the knowledge, identify the gaps in understanding, and suggest future research directions to elucidate the role of EVs in PwCF during COVID-19.


COVID-19 , Cystic Fibrosis , Extracellular Vesicles , Humans , Cystic Fibrosis/complications , SARS-CoV-2 , Inflammation
15.
Respir Res ; 25(1): 190, 2024 Apr 29.
Article En | MEDLINE | ID: mdl-38685088

BACKGROUND: Children with advanced pulmonary disease due to cystic fibrosis (CF) are at risk of acute respiratory failure due to pulmonary exacerbations leading to their admission to pediatric intensive care units (PICU). The objectives of this study were to determine short and medium-term outcomes of children with CF admitted to PICU for acute respiratory failure due to pulmonary exacerbation and to identify prognosis factors. METHODS: This retrospective monocentric study included patients less than 18 years old admitted to the PICU of a French university hospital between 2000 and 2020. Cox proportional hazard regression methods were used to determine prognosis factors of mortality or lung transplant. RESULTS: Prior to PICU admission, the 29 patients included (median age 13.5 years) had a severe lung disease (median Forced Expiratory Volume in 1 s percentage predicted at 29%). Mortality rates were respectively 17%, 31%, 34%, 41% at discharge and at 3, 12 and 36 months post-discharge. Survival rates free of lung transplant were 34%, 32%, 24% and 17% respectively. Risk factors associated with mortality or lung transplant using the univariate analysis were female sex and higher pCO2 and chloride levels at PICU admission, and following pre admission characteristics: home respiratory and nutritional support, registration on lung transplant list and Stenotrophomonas Maltophilia bronchial colonization. CONCLUSION: Children with CF admitted to PICU for acute respiratory failure secondary to pulmonary exacerbations are at high risk of death, both in the short and medium terms. Lung transplant is their main chance of survival and should be considered early.


Cystic Fibrosis , Intensive Care Units, Pediatric , Respiratory Insufficiency , Humans , Cystic Fibrosis/mortality , Cystic Fibrosis/complications , Cystic Fibrosis/diagnosis , Female , Male , Retrospective Studies , Child , Adolescent , Respiratory Insufficiency/mortality , Respiratory Insufficiency/diagnosis , Respiratory Insufficiency/etiology , Intensive Care Units, Pediatric/statistics & numerical data , Risk Factors , Disease Progression , France/epidemiology , Child, Preschool , Treatment Outcome
16.
Int J Antimicrob Agents ; 63(6): 107161, 2024 Jun.
Article En | MEDLINE | ID: mdl-38561094

OBJECTIVE: Hypermutable Pseudomonas aeruginosa strains are highly prevalent in chronic lung infections of patients with cystic fibrosis (CF). Acute exacerbations of these infections have limited treatment options. This study aimed to investigate inhaled aztreonam and tobramycin against clinical hypermutable P. aeruginosa strains using the CDC dynamic in vitro biofilm reactor (CBR), mechanism-based mathematical modelling (MBM) and genomic studies. METHODS: Two CF multidrug-resistant strains were investigated in a 168 h CBR (n = 2 biological replicates). Regimens were inhaled aztreonam (75 mg 8-hourly) and tobramycin (300 mg 12-hourly) in monotherapies and combination. The simulated pharmacokinetic profiles of aztreonam and tobramycin (t1/2 = 3 h) were based on published lung fluid concentrations in patients with CF. Total viable and resistant counts were determined for planktonic and biofilm bacteria. MBM of total and resistant bacterial counts and whole genome sequencing were completed. RESULTS: Both isolates showed reproducible bacterial regrowth and resistance amplification for the monotherapies by 168 h. The combination performed synergistically, with minimal resistant subpopulations compared to the respective monotherapies at 168 h. Mechanistic synergy appropriately described the antibacterial effects of the combination regimen in the MBM. Genomic analysis of colonies recovered from monotherapy regimens indicated noncanonical resistance mechanisms were likely responsible for treatment failure. CONCLUSION: The combination of aztreonam and tobramycin was required to suppress the regrowth and resistance of planktonic and biofilm bacteria in all biological replicates of both hypermutable multidrug-resistant P. aeruginosa CF isolates. The developed MBM could be utilised for future investigations of this promising inhaled combination.


Anti-Bacterial Agents , Aztreonam , Biofilms , Cystic Fibrosis , Drug Synergism , Pseudomonas Infections , Pseudomonas aeruginosa , Tobramycin , Whole Genome Sequencing , Tobramycin/administration & dosage , Tobramycin/pharmacology , Aztreonam/pharmacology , Aztreonam/administration & dosage , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/genetics , Biofilms/drug effects , Cystic Fibrosis/microbiology , Cystic Fibrosis/complications , Humans , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/pharmacokinetics , Anti-Bacterial Agents/therapeutic use , Administration, Inhalation , Pseudomonas Infections/drug therapy , Pseudomonas Infections/microbiology , Microbial Sensitivity Tests , Drug Resistance, Multiple, Bacterial/genetics , Models, Theoretical , Drug Therapy, Combination
17.
Diagn Microbiol Infect Dis ; 109(3): 116321, 2024 Jul.
Article En | MEDLINE | ID: mdl-38677054

Cystic fibrosis patients' lungs are chronically colonized by multiple microbial species capable of forming biofilms. This study aimed to characterize the polymicrobial biofilm formed by Candida spp. and S. aureus, co-isolated from sputum samples of cystic fibrosis patients regarding microbial density, metabolic activity, and structure. 67 samples from 28 patients were collected with a 96% alteration rate. 34% showed alterations by both Candida spp. and Gram-positive bacteria, predominantly Candida spp. and S. aureus in 77% of cases, accounting for 6 associations. Biofilm biomass was quantified using the crystal violet assay, and metabolic activity was assessed using the MTT reduction assay. Scanning electron microscopy analyzed the C. tropicalis/S. aureus24 biofilm architecture. Candida spp. isolates demonstrated the ability to form mixed biofilms with S. aureus. The C. tropicalis/S. aureus24 association exhibited the highest production of biofilm and metabolic activity, along with the C. albicans17/C. rugosa/S. aureus7 in both single and mixed biofilms.


Biofilms , Candida , Cystic Fibrosis , Sputum , Staphylococcus aureus , Biofilms/growth & development , Humans , Cystic Fibrosis/microbiology , Cystic Fibrosis/complications , Staphylococcus aureus/isolation & purification , Staphylococcus aureus/physiology , Algeria , Candida/isolation & purification , Candida/classification , Candida/physiology , Sputum/microbiology , Staphylococcal Infections/microbiology , Coinfection/microbiology , Female , Male , Adult , Candidiasis/microbiology , Microscopy, Electron, Scanning , Young Adult , Adolescent , Child
18.
Diagn Microbiol Infect Dis ; 109(3): 116294, 2024 Jul.
Article En | MEDLINE | ID: mdl-38678689

Cystic fibrosis (CF) is a progressive and inherited disease that affects approximately 70000 individuals all over the world annually. A mutation in the cystic fibrosis transmembrane conductance regulator (CFTR) gene serves as its defining feature. Bacterial infections have a significant impact on the occurrence and development of CF. In this manuscript, we discuss the role and virulence factors of Staphylococcus aureus as an important human pathogen with the ability to induce respiratory tract infections. Recent studies have reported S. aureus as the first isolated bacteria in CF patients. Methicillin-resistant Staphylococcus aureus (MRSA) pathogens are approximately resistant to all ß-lactams. CF patients are colonized by MRSA expressing various virulence factors including toxins, and Staphylococcal Cassette Chromosome mec (SCCmec) types, and have the potential for biofilm formation. Therefore, variations in clinical outcomes will be manifested. SCCmec type II has been reported in CF patients more than in other SCCmec types from different countries. The small-colony variants (SCVs) as specific morphologic subtypes of S. aureus with slow growth and unusual properties can also contribute to persistent and difficult-to-treat infections in CF patients. The pathophysiology of SCVs is complicated and not fully understood. Patients with cystic fibrosis should be aware of the intrinsic risk factors for complex S. aureus infections, including recurring infections, physiological issues, or coinfection with P. aeruginosa.


Cystic Fibrosis , Staphylococcal Infections , Staphylococcus aureus , Virulence Factors , Cystic Fibrosis/microbiology , Cystic Fibrosis/complications , Humans , Staphylococcal Infections/microbiology , Staphylococcus aureus/genetics , Staphylococcus aureus/pathogenicity , Virulence Factors/genetics , Respiratory Tract Infections/microbiology , Biofilms/growth & development , Methicillin-Resistant Staphylococcus aureus/genetics , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Microbial Interactions , Cystic Fibrosis Transmembrane Conductance Regulator/genetics
19.
mBio ; 15(5): e0051924, 2024 May 08.
Article En | MEDLINE | ID: mdl-38564694

Today, more than 90% of people with cystic fibrosis (pwCF) are eligible for the highly effective cystic fibrosis transmembrane conductance regulator (CFTR) modulator therapy called elexacaftor/tezacaftor/ivacaftor (ETI) and its use is widespread. Given the drastic respiratory symptom improvement experienced by many post-ETI, clinical studies are already underway to reduce the number of respiratory therapies, including antibiotic regimens, that pwCF historically relied on to combat lung disease progression. Early studies suggest that bacterial burden in the lungs is reduced post-ETI, yet it is unknown how chronic Pseudomonas aeruginosa populations are impacted by ETI. We found that pwCF remain infected throughout their upper and lower respiratory tract with their same strain of P. aeruginosa post-ETI, and these strains continue to evolve in response to the newly CFTR-corrected airway. Our work underscores the continued importance of CF airway microbiology in the new era of highly effective CFTR modulator therapy. IMPORTANCE: The highly effective cystic fibrosis transmembrane conductance regulator modulator therapy Elexakaftor/Tezacaftor/Ivacaftor (ETI) has changed cystic fibrosis (CF) disease for many people with cystic fibrosis. While respiratory symptoms are improved by ETI, we found that people with CF remain infected with Pseudomonas aeruginosa. How these persistent and evolving bacterial populations will impact the clinical manifestations of CF in the coming years remains to be seen, but the role and potentially changing face of infection in CF should not be discounted in the era of highly effective modulator therapy.


Aminophenols , Benzodioxoles , Cystic Fibrosis Transmembrane Conductance Regulator , Cystic Fibrosis , Drug Combinations , Indoles , Pseudomonas Infections , Pseudomonas aeruginosa , Quinolones , Cystic Fibrosis/microbiology , Cystic Fibrosis/drug therapy , Cystic Fibrosis/complications , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/genetics , Humans , Pseudomonas Infections/drug therapy , Pseudomonas Infections/microbiology , Aminophenols/therapeutic use , Quinolones/therapeutic use , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Benzodioxoles/therapeutic use , Indoles/therapeutic use , Pyrazoles/therapeutic use , Pyrroles/therapeutic use , Pyridines/therapeutic use , Thiophenes/therapeutic use , Thiophenes/pharmacology , Female , Quinolines
20.
J Hum Nutr Diet ; 37(3): 804-814, 2024 Jun.
Article En | MEDLINE | ID: mdl-38664916

Nutrition has played a central role in the management and outcomes of people with cystic fibrosis (pwCF) since the 1970s. Advances in therapies and practices in recent decades have led to a significant change in the patient landscape with dramatic improvements in life expectancy, as well as quality of life, bringing with it new issues. Historically, cystic fibrosis was a condition associated with childhood and malnutrition; however, changes in patient demographics, nutritional assessment and fundamental nutritional management have evolved, and it has become an increasingly prevalent adult disease with new nutritional challenges, including obesity. This paper aims to describe these changes and the impact and challenges they bring for those working in this field. Nutritional professionals will need to evolve, adapt and remain agile to the wider range of situations and support required for a new generation of pwCF. Specialised nutrition support will continue to be required, and it will be additionally important to improve and optimise quality of life and long-term health.


Cystic Fibrosis , Quality of Life , Humans , Cystic Fibrosis/complications , Cystic Fibrosis/diet therapy , Cystic Fibrosis/therapy , Child , Nutritional Status , Malnutrition/etiology , Malnutrition/prevention & control , Malnutrition/therapy , Nutrition Assessment , Nutritional Support/methods , Nutrition Therapy/methods , Adolescent
...