Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.760
Filter
1.
Stud Health Technol Inform ; 316: 212-213, 2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39176710

ABSTRACT

Respiratory tract infections are a serious threat to health, especially in the presence of antimicrobial resistance (AMR). Existing AMR detection methods are limited by slow turnaround times and low accuracy due to the presence of false positives and negatives. In this study, we simulate 1,116 clinical metagenomics samples on both Illumina and Nanopore sequencing from curated, real-world sequencing of A. baumannii respiratory infections and build AI models to predict resistance to amikacin. The best performance is achieved by XGBoost on Illumina sequencing (area under the ROC curve = 0.7993 on 5-fold cross-validation).


Subject(s)
Acinetobacter baumannii , Amikacin , Drug Resistance, Bacterial , Metagenomics , Amikacin/pharmacology , Amikacin/therapeutic use , Acinetobacter baumannii/drug effects , Acinetobacter baumannii/genetics , Humans , Drug Resistance, Bacterial/genetics , Respiratory Tract Infections/drug therapy , Respiratory Tract Infections/microbiology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Acinetobacter Infections/drug therapy , Acinetobacter Infections/microbiology
2.
J Infect Dev Ctries ; 18(6): 978-981, 2024 Jun 30.
Article in English | MEDLINE | ID: mdl-38990993

ABSTRACT

INTRODUCTION: Mycetoma is a chronic granulomatous inflammatory disease of the subcutaneous tissue, which affects deep structures and bone. Most cases of actinomycetoma are caused by members of the genus Nocardia. CASE PRESENTATION: Here we report the case of a 43-year-old male who presented a disseminated mycetoma on the forearm, chest and neck, characterized by enlarged and erythematous lesions through which seropurulent material drains, and numerous atrophic scars. Molecular identification was performed by 16S gene amplification and sequencing. Nocardia mexicana was identified with 100% identity. Trimethoprim-sulfamethoxazole, diaminodiphenyl sulfone and amikacin was a successful treatment after 6 months. CONCLUSIONS: Nocardia mexicana is a rare organism that causes mycetoma. We report a case of extensive mycetoma on the forearm with spread to the neck and thorax associated with manipulation of the mouth of a calf.


Subject(s)
Anti-Bacterial Agents , Forearm , Mycetoma , Neck , Nocardia Infections , Nocardia , RNA, Ribosomal, 16S , Thorax , Humans , Male , Adult , Nocardia/isolation & purification , Nocardia/genetics , Mycetoma/microbiology , Mycetoma/drug therapy , Mycetoma/diagnosis , Nocardia Infections/microbiology , Nocardia Infections/drug therapy , Nocardia Infections/diagnosis , Forearm/microbiology , Forearm/pathology , Thorax/diagnostic imaging , Thorax/microbiology , Neck/pathology , Anti-Bacterial Agents/therapeutic use , RNA, Ribosomal, 16S/genetics , Sequence Analysis, DNA , DNA, Bacterial/genetics , Treatment Outcome , Trimethoprim, Sulfamethoxazole Drug Combination/therapeutic use , Amikacin/therapeutic use , DNA, Ribosomal/genetics , DNA, Ribosomal/chemistry
3.
mSphere ; 9(7): e0038124, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-38980071

ABSTRACT

Treatment outcomes for Mycobacteroides abscessus (Mab, also known as Mycobacterium abscessus) disease are still unsatisfactory, mainly due to issues with drug toxicity, tolerability, and efficacy. Treating Mab disease is challenging due to its high baseline antibiotic resistance, initial requirement for intravenous therapy, and poor medication tolerance. Omadacycline, a new tetracycline, is active against Mab. Since any new antibiotic effective against Mab is expected to be used in combination with other antibiotics, we evaluated the efficacy of two triple-drug combinations comprising omadacycline, omadacycline + amikacin + imipenem, and omadacycline + clofazimine + linezolid against two contemporary Mab clinical isolates in a mouse model of Mab lung disease. Antibiotic administration was initiated 1-week post-infection and was given daily, with Mab burden in the lungs at treatment completion serving as the endpoint. Omadacycline alone moderately reduced Mab levels and maintained better health in mice compared to untreated ones, which typically suffered from the infection. The omadacycline + clofazimine + linezolid combination showed immediate bactericidal activity and enhanced efficacy over 6 weeks, particularly against the more resistant strain (M9507). However, the clofazimine + linezolid combination lacked early bactericidal activity. When combined with amikacin and imipenem, omadacycline did not improve the regimen's effectiveness over 4 weeks of treatment. Our study showed that omadacycline + clofazimine + linezolid exhibited significant bactericidal activity over an extended treatment duration. However, adding omadacycline to amikacin and imipenem did not improve regimen effectiveness against the evaluated clinical isolates within 4 weeks. Further research in Mab disease patients is needed to determine the most effective omadacycline-containing regimen.IMPORTANCEMycobacteroides abscessus is a common environmental bacterium that causes infections in people with compromised lung function, including those with bronchiectasis, cystic fibrosis, chronic obstructive pulmonary disease, and weakened immune systems, especially among older individuals. Treating M. abscessus disease is challenging due to the limited effectiveness and toxicity of current antibiotics, which often require prolonged use. Omadacycline, a new antibiotic, shows promise against M. abscessus. Using a mouse model that mimics M. abscessus disease in humans, we studied the effectiveness of including omadacycline with recommended antibiotics. Adding omadacycline to clofazimine and linezolid significantly improved treatment outcomes, rapidly clearing the bacteria from the lungs and maintaining effectiveness throughout. This oral combination is convenient for patients. However, adding omadacycline to amikacin and imipenem did not improve treatment effectiveness within 4 weeks. Further study with M. abscessus patients is necessary to optimize omadacycline-based treatment strategies for this disease.


Subject(s)
Amikacin , Anti-Bacterial Agents , Clofazimine , Disease Models, Animal , Drug Therapy, Combination , Imipenem , Linezolid , Mycobacterium Infections, Nontuberculous , Mycobacterium abscessus , Tetracyclines , Animals , Clofazimine/administration & dosage , Clofazimine/therapeutic use , Linezolid/administration & dosage , Linezolid/therapeutic use , Mice , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/therapeutic use , Amikacin/administration & dosage , Amikacin/therapeutic use , Tetracyclines/administration & dosage , Tetracyclines/therapeutic use , Tetracyclines/pharmacology , Mycobacterium abscessus/drug effects , Imipenem/administration & dosage , Imipenem/therapeutic use , Imipenem/pharmacology , Mycobacterium Infections, Nontuberculous/drug therapy , Mycobacterium Infections, Nontuberculous/microbiology , Female , Treatment Outcome , Microbial Sensitivity Tests , Lung Diseases/drug therapy , Lung Diseases/microbiology , Administration, Oral , Lung/microbiology
4.
Antimicrob Agents Chemother ; 68(8): e0152023, 2024 Aug 07.
Article in English | MEDLINE | ID: mdl-38990014

ABSTRACT

Mycobacterium abscessus pulmonary infections are increasingly problematic, especially for immunocompromised individuals and those with underlying lung conditions. Currently, there is no reliable standardized treatment, underscoring the need for improved preclinical drug testing. We present a simplified immunosuppressed mouse model using only four injections of cyclophosphamide, which allows for sustained M. abscessus lung burden for up to 16 days. This model proved effective for antibiotic efficacy evaluation, as demonstrated with imipenem or amikacin.


Subject(s)
Amikacin , Anti-Bacterial Agents , Cyclophosphamide , Disease Models, Animal , Mycobacterium Infections, Nontuberculous , Mycobacterium abscessus , Animals , Cyclophosphamide/pharmacology , Mycobacterium abscessus/drug effects , Mice , Mycobacterium Infections, Nontuberculous/drug therapy , Mycobacterium Infections, Nontuberculous/microbiology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Amikacin/pharmacology , Amikacin/therapeutic use , Imipenem/pharmacology , Imipenem/therapeutic use , Lung/microbiology , Lung/drug effects , Immunocompromised Host , Female
5.
BMJ Open ; 14(7): e078379, 2024 Jul 24.
Article in English | MEDLINE | ID: mdl-39053960

ABSTRACT

INTRODUCTION: An effective rifampicin-resistant tuberculosis (RR-TB) treatment regimen should include prevention of resistance amplification. While bedaquiline (BDQ) has been recommended in all-oral RR-TB treatment regimen since 2019, resistance is rising at alarming rates. This may be due to BDQ's delayed bactericidal effect, which increases the risk of selecting for resistance to fluoroquinolones and/or BDQ in the first week of treatment when the bacterial load is highest. We aim to strengthen the first week of treatment with the injectable drug amikacin (AMK). To limit the ototoxicity risk while maximising the bactericidal effect, we will evaluate the safety of adding a 30 mg/kg AMK injection on the first and fourth day of treatment. METHODS AND ANALYSIS: We will conduct a single-arm clinical trial on 20 RR-TB patients nested within an operational study called ShoRRT (All oral Shorter Treatment Regimen for Drug resistant Tuberculosis). In addition to all-oral RR-TB treatment, patients will receive two doses of AMK. The primary safety endpoint is any grade 3-4 adverse event during the first 2 weeks of treatment related to the use of AMK. With a sample size of 20 patients, we will have at least 80% statistical power to support the alternative hypothesis, indicating that less than 14% of patients treated with AMK experience a grade 3-4 adverse event related to its use. Safety data obtained from this study will inform a larger multicountry study on using two high doses of AMK to prevent acquired resistance. ETHICS AND DISSEMINATION: Approval was obtained from the ethics committee of Rwanda, Rwanda Food and Drug Authority, Universitair Ziekenhuis, the Institute of Tropical Medicine ethics review board. All participants will provide informed consent. Study results will be disseminated through peer-reviewed journals and conferences. TRIAL REGISTRATION NUMBER: NCT05555303.


Subject(s)
Amikacin , Rifampin , Tuberculosis, Multidrug-Resistant , Humans , Amikacin/administration & dosage , Amikacin/adverse effects , Amikacin/therapeutic use , Tuberculosis, Multidrug-Resistant/drug therapy , Rifampin/administration & dosage , Rifampin/therapeutic use , Antitubercular Agents/administration & dosage , Antitubercular Agents/adverse effects , Antitubercular Agents/therapeutic use , Administration, Oral , Adult , Mycobacterium tuberculosis/drug effects , Male , Female , Drug Administration Schedule
6.
J Infect Dev Ctries ; 18(5): 826-828, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38865393

ABSTRACT

INTRODUCTION: Mycobacterium canariasense is a relatively rare and rapidly growing nontuberculous mycobacterium (NTM) infection. CASE REPORT: This case report describes a 36-year-old man with a Canariasense infection in the lung with solitary cavitation nodules located subpleural on CT scan, for which the final diagnosis was made by metagenomic next-generation sequencing (mNGS) of bronchoalveolar lavage fluid (BALF-mNGS). It was successfully treated with levofloxacin and amikacin. CONCLUSIONS: This experience is instructive because clinical diagnostic and CT imaging characteristics and treatment strategy guidelines for pulmonary infections caused by M. canariasense have not yet been established.


Subject(s)
Anti-Bacterial Agents , Mycobacterium Infections, Nontuberculous , Tomography, X-Ray Computed , Humans , Male , Adult , Mycobacterium Infections, Nontuberculous/diagnosis , Mycobacterium Infections, Nontuberculous/microbiology , Mycobacterium Infections, Nontuberculous/drug therapy , Mycobacterium Infections, Nontuberculous/diagnostic imaging , Anti-Bacterial Agents/therapeutic use , Bronchoalveolar Lavage Fluid/microbiology , Lung/microbiology , Lung/pathology , Lung/diagnostic imaging , Levofloxacin/therapeutic use , Amikacin/therapeutic use , Nontuberculous Mycobacteria/isolation & purification , Nontuberculous Mycobacteria/genetics , High-Throughput Nucleotide Sequencing , Treatment Outcome
7.
Microbiol Spectr ; 12(6): e0009624, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38695572

ABSTRACT

Pseudomonas can lead to peritoneal dialysis-associated peritonitis, which is characterized by a poor prognosis, such as a substantial failure rate and a high death rate. This study aimed to provide an overview of Pseudomonas peritonitis's clinical features, the regimens of antibiotic, antibiotic resistance, and outcomes in peritoneal dialysis (PD) patients. This study observed patients with Pseudomonas peritonitis in two large PD centers in South China from January 2008 to December 2022. The demographics, symptomatology, antibiotics regimens, resistance to common antibiotics, and clinical outcomes of all included patients were reviewed. A total of 3,459 PD patients were included, among them 57 cases of peritonitis caused by Pseudomonas, including 48 cases (84.2%) of Pseudomonas aeruginosa. The incidence rate of Pseudomonas peritonitis was 0.0041 episode per patient-year. Of them, 28.1% (16 cases) of the patients were accompanied by exit site infection (ESI), and all had abdominal pain and turbid ascites at the time of onset. The most commonly used antibiotic combination was ceftazidime combined with amikacin. Approximately 89% of Pseudomonas species were sensitive to ceftazidime, and 88% were sensitive to amikacin. The overall primary response rate was 28.1% (16 patients), and the complete cure rate was 40.4% (23 patients). There was no significant difference in the complete cure rate of peritonitis using three and other antibiotic treatment regimens (44.8% vs 46.4%; P = 0.9). The successful treatment group had higher baseline albumin level (35.9 ± 6.2; P = 0.008) and residual urine volume (650.7 ± 375.5; P = 0.04). Although the incidence of peritonitis caused by Pseudomonas was low, the symptoms were serious, and prognosis was very poor. Pseudomonas was still highly susceptible to first-line antibiotics currently in use against Gram-negative bacteria. Patients with successful treatment had higher albumin levels and higher urine output. IMPORTANCE: Although the incidence of peritoneal dialysis-associated peritonitis caused by Pseudomonas is very low, it seriously affects the technique survival of peritoneal dialysis patients. However, there are few studies and reports on Pseudomonas peritonitis in the Chinese mainland area. Therefore, the purpose of this study is to describe the clinical characteristics, the regimens of antibiotic, drug resistance, and outcome of peritoneal dialysis patients in southern China in the past 15 years and summarize the clinical experience in the treatment of Pseudomonas peritonitis.


Subject(s)
Anti-Bacterial Agents , Peritoneal Dialysis , Peritonitis , Pseudomonas Infections , Pseudomonas , Humans , Peritonitis/drug therapy , Peritonitis/microbiology , Peritonitis/epidemiology , Anti-Bacterial Agents/therapeutic use , China/epidemiology , Pseudomonas Infections/drug therapy , Pseudomonas Infections/microbiology , Pseudomonas Infections/epidemiology , Peritoneal Dialysis/adverse effects , Male , Female , Middle Aged , Aged , Pseudomonas/drug effects , Pseudomonas/isolation & purification , Adult , Pseudomonas aeruginosa/drug effects , Retrospective Studies , Ceftazidime/therapeutic use , Microbial Sensitivity Tests , Amikacin/therapeutic use
8.
PLoS One ; 19(5): e0304265, 2024.
Article in English | MEDLINE | ID: mdl-38809914

ABSTRACT

To analyze the results of proficiency testing for anti-tuberculosis drug susceptibility testing (DST) in China. Number of laboratory participating the proficiency testing performed DST, and the sensitivity, specificity, reproducibility, and accordance rate were calculated from data of 13 rounds proficiency testing results for DST from 2008 to 2021. A total of 30 and 20 strains of Mycobacterium tuberculosis with known susceptibility results were sent to each laboratory in 2008 to 2019, 2020 and 2021, respectively. The number of participating laboratories ranged from 30 in 2009 to 546 in 2021. L-J DST was the predominant method. The specificity presented relatively higher than sensitivity. Improvement of specificity were observed for all drugs through the years, while sensitivity did not show improvement for amikacin and capreomycin. Accordance rate of pyrazinamide and kanamycin and reproducibility of capreomycin and pyrazinamide were not significantly improved through the years. Most of the participating laboratories significantly improved the quality of their DST through the consecutive rounds of proficiency testing except for second-line injectable drugs and pyrazinamide. The results highlight the importance of developing novel and/or improving existing methods for phenotypic DST for certain drugs.


Subject(s)
Antitubercular Agents , Microbial Sensitivity Tests , Mycobacterium tuberculosis , Mycobacterium tuberculosis/drug effects , China , Antitubercular Agents/pharmacology , Antitubercular Agents/therapeutic use , Humans , Laboratory Proficiency Testing , Reproducibility of Results , Phenotype , Amikacin/pharmacology , Amikacin/therapeutic use , Pyrazinamide/therapeutic use
9.
J Am Vet Med Assoc ; 262(9): 1-7, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38810661

ABSTRACT

OBJECTIVE: To report the clinical use, adverse events, and outcomes after using amikacin in 30% poloxamer 407 (amikacin-P407) during open wound management or in a closed wound application in dogs. ANIMALS: 29 client-owned dogs. METHODS: Medical records from January 2017 to August 2023 from a single hospital were reviewed for dogs that received amikacin-P407 in an open or closed wound application. Information reviewed included signalment, nature of wound and/or surgical site infection (SSI), bacterial cultures, amikacin dose, gel volume, route of administration, estimated wound surface area, biochemistry parameters, urine casts, wound progression, and general clinical outcome. RESULTS: Amikacin-P407 was applied during open wound care (10 dogs), via injection (5 dogs), and at time of wound closure (13 dogs) and was used both in open and closed wound management (1 dog). Wounds were associated with SSIs in 18 of 30 sites. Multidrug resistance was noted in 21 of 30 preapplication cultures. Median amikacin dose was 14.5 mg/kg (range, 3 to 59.5 mg/kg), median total volume was 5.0 mL (range, 1 to 12 mL), and median tissue surface area was 6.6 cm2 (range, 1.6 to 36 cm2), for a local wound dose of 62.5 mg/cm2 (range, 6.9 to 214.3 mg/cm2). No short-term adverse local or systemic effects were noted in any wounds or dogs. No dehiscence was seen in 17 of 19 closed sites. CLINICAL RELEVANCE: The results of this case series suggested that Amikacin-P407 can be applied in a variety of ways with no adverse effects. Amikacin-P407 may be considered in open wound management or in a closed setting for infected wounds and SSIs.


Subject(s)
Amikacin , Anti-Bacterial Agents , Dog Diseases , Poloxamer , Animals , Dogs , Amikacin/administration & dosage , Amikacin/therapeutic use , Anti-Bacterial Agents/therapeutic use , Anti-Bacterial Agents/administration & dosage , Female , Dog Diseases/drug therapy , Male , Retrospective Studies , Treatment Outcome
10.
Nat Commun ; 15(1): 3947, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38729951

ABSTRACT

Gram-negative bacteria (GNB) are a major cause of neonatal sepsis in low- and middle-income countries (LMICs). Although the World Health Organization (WHO) reports that over 80% of these sepsis deaths could be prevented through improved treatment, the efficacy of the currently recommended first- and second-line treatment regimens for this condition is increasingly affected by high rates of drug resistance. Here we assess three well known antibiotics, fosfomycin, flomoxef and amikacin, in combination as potential antibiotic treatment regimens by investigating the drug resistance and genetic profiles of commonly isolated GNB causing neonatal sepsis in LMICs. The five most prevalent bacterial isolates in the NeoOBS study (NCT03721302) are Klebsiella pneumoniae, Acinetobacter baumannii, E. coli, Serratia marcescens and Enterobacter cloacae complex. Among these isolates, high levels of ESBL and carbapenemase encoding genes are detected along with resistance to ampicillin, gentamicin and cefotaxime, the current WHO recommended empiric regimens. The three new combinations show excellent in vitro activity against ESBL-producing K. pneumoniae and E. coli isolates. Our data should further inform and support the clinical evaluation of these three antibiotic combinations for the treatment of neonatal sepsis in areas with high rates of multidrug-resistant Gram-negative bacteria.


Subject(s)
Acinetobacter baumannii , Anti-Bacterial Agents , Gram-Negative Bacteria , Gram-Negative Bacterial Infections , Klebsiella pneumoniae , Microbial Sensitivity Tests , Neonatal Sepsis , Humans , Anti-Bacterial Agents/therapeutic use , Anti-Bacterial Agents/pharmacology , Neonatal Sepsis/microbiology , Neonatal Sepsis/drug therapy , Infant, Newborn , Gram-Negative Bacteria/drug effects , Gram-Negative Bacteria/genetics , Gram-Negative Bacteria/isolation & purification , Gram-Negative Bacterial Infections/drug therapy , Gram-Negative Bacterial Infections/microbiology , Acinetobacter baumannii/drug effects , Acinetobacter baumannii/isolation & purification , Acinetobacter baumannii/genetics , Klebsiella pneumoniae/drug effects , Klebsiella pneumoniae/isolation & purification , Klebsiella pneumoniae/genetics , Amikacin/pharmacology , Amikacin/therapeutic use , Fosfomycin/pharmacology , Fosfomycin/therapeutic use , beta-Lactamases/genetics , beta-Lactamases/metabolism , Escherichia coli/drug effects , Escherichia coli/genetics , Escherichia coli/isolation & purification , Developing Countries , Drug Resistance, Multiple, Bacterial/genetics , Drug Therapy, Combination , Serratia marcescens/drug effects , Serratia marcescens/genetics , Serratia marcescens/isolation & purification , Enterobacter cloacae/drug effects , Enterobacter cloacae/genetics , Enterobacter cloacae/isolation & purification , Bacterial Proteins/genetics , Bacterial Proteins/metabolism
11.
BMJ Case Rep ; 17(5)2024 May 15.
Article in English | MEDLINE | ID: mdl-38749526

ABSTRACT

We report the case of a female patient in her late 20s who visited the clinic with concerns about poor vision, redness, watering and a burning sensation in her left eye 2 weeks after undergoing a small incision lenticule extraction. She had no history of systemic illness or immunosuppressed status. On slit lamp examination, she was found to have corneal stromal infiltrates in the interface at multiple locations. Given the clinical diagnosis of microbial keratitis, corneal scraping of the interface infiltrate was performed and sent for microbiological examination revealing gram-positive, thin, beaded filaments that were acid-fast positive and later identified by growth in culture media as Nocardia species. This case was managed successfully with the use of topical amikacin and systemic trimethoprim-sulfamethoxazole with complete resolution of infection.


Subject(s)
Anti-Bacterial Agents , Eye Infections, Bacterial , Keratitis , Nocardia Infections , Humans , Female , Nocardia Infections/diagnosis , Nocardia Infections/drug therapy , Keratitis/microbiology , Keratitis/drug therapy , Keratitis/diagnosis , Keratitis/surgery , Anti-Bacterial Agents/therapeutic use , Eye Infections, Bacterial/diagnosis , Eye Infections, Bacterial/microbiology , Eye Infections, Bacterial/drug therapy , Amikacin/therapeutic use , Amikacin/administration & dosage , Adult , Trimethoprim, Sulfamethoxazole Drug Combination/therapeutic use , Nocardia/isolation & purification , Surgical Wound Infection/microbiology , Surgical Wound Infection/drug therapy , Surgical Wound Infection/diagnosis
13.
BMJ Case Rep ; 17(4)2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38688568

ABSTRACT

Catheter-related bloodstream infection (CRBSI) is one of the common healthcare-acquired infections imposing a high burden of morbidity and mortality on the patients. Non-tuberculous mycobacterium is a rare aetiology for CRBSI and poses challenges in laboratory diagnosis and clinical management. This is a case of a woman in her early 60s with underlying end-stage renal failure, diabetes mellitus and hypertension presented with a 2-week history of high-grade fever postregular haemodialysis, vomiting, lethargy and altered mental status.Blood cultures from a permanent catheter and peripheral taken concurrently yielded Mycobacterium senegalense, identified by matrix-assisted laser desorption/ionisation time-of-flight mass spectrometry, which established the diagnosis of CRBSI atypically presented with concurrent acute intracranial bleeding and cerebrovascular infarction at initial presentation. She was started on a combination of oral azithromycin, oral amikacin and intravenous imipenem, and the permanent catheter was removed. Despite the treatments instituted, she developed septicaemia, acute myocardial infarction and macrophage activation-like syndrome, causing the patient's death.


Subject(s)
Anti-Bacterial Agents , Catheter-Related Infections , Mycobacterium Infections, Nontuberculous , Humans , Female , Catheter-Related Infections/microbiology , Catheter-Related Infections/diagnosis , Catheter-Related Infections/drug therapy , Mycobacterium Infections, Nontuberculous/diagnosis , Mycobacterium Infections, Nontuberculous/drug therapy , Middle Aged , Anti-Bacterial Agents/therapeutic use , Anti-Bacterial Agents/administration & dosage , Fatal Outcome , Bacteremia/drug therapy , Bacteremia/diagnosis , Bacteremia/microbiology , Renal Dialysis , Kidney Failure, Chronic/therapy , Kidney Failure, Chronic/complications , Amikacin/therapeutic use , Amikacin/administration & dosage
14.
Diagn Microbiol Infect Dis ; 109(3): 116296, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38640607

ABSTRACT

Pulmonary infection due to Mycobacterium abscessus complex (MABC) usually occurs in children with underlying risk factors including cystic fibrosis (CF), chronic lung disease, and immunocompromised status, but rarely in immunocompetent children without underlying lung disease, especially in infants. We present a case of MABC pulmonary disease (MABC-PD) in an otherwise healthy 53-day-old male infant with one week of cough and respiratory distress. Computed tomography showed multiple masses across both lungs. Isolated mycobacteria from his bronchoalveolar lavage fluid were identified as MABC. We describe our complete evaluation, including immunodeficiency evaluation incorporating whole exome sequencing and our therapeutic process given complicated susceptibility pattern of the M. abscessus isolate, and review literature for MABC-PD in immunocompetent children. The infant was successfully treated through prolonged treatment with parenteral Amikacin, Cefoxitin, Linezolid, and Clarithromycin, combined with inhaled Amikacin.


Subject(s)
Anti-Bacterial Agents , Mycobacterium Infections, Nontuberculous , Mycobacterium abscessus , Humans , Male , Mycobacterium abscessus/drug effects , Mycobacterium abscessus/isolation & purification , Mycobacterium Infections, Nontuberculous/drug therapy , Mycobacterium Infections, Nontuberculous/microbiology , Mycobacterium Infections, Nontuberculous/diagnosis , Anti-Bacterial Agents/therapeutic use , Infant , Bronchoalveolar Lavage Fluid/microbiology , Amikacin/therapeutic use , Pneumonia, Bacterial/drug therapy , Pneumonia, Bacterial/microbiology , Pneumonia, Bacterial/diagnosis , Treatment Outcome , Tomography, X-Ray Computed , Clarithromycin/therapeutic use , Linezolid/therapeutic use
15.
Arch Microbiol ; 206(4): 148, 2024 Mar 11.
Article in English | MEDLINE | ID: mdl-38462558

ABSTRACT

Pseudomonas aeruginosa is an opportunistic gram-negative pathogenic microorganism that poses a significant challenge in clinical treatment. Antibiotics exhibit limited efficacy against mature biofilm, culminating in an increase in the number of antibiotic-resistant strains. Therefore, novel strategies are essential to enhance the effectiveness of antibiotics against Pseudomonas aeruginosa biofilms. D-histidine has been previously identified as a prospective anti-biofilm agent. However, limited attention has been directed towards its impact on Pseudomonas aeruginosa. Therefore, this study was undertaken to explore the effect of D-histidine on Pseudomonas aeruginosa in vitro. Our results demonstrated that D-histidine downregulated the mRNA expression of virulence and quorum sensing (QS)-associated genes in Pseudomonas aeruginosa PAO1 without affecting bacterial growth. Swarming and swimming motility tests revealed that D-histidine significantly reduced the motility and pathogenicity of PAO1. Moreover, crystal violet staining and confocal laser scanning microscopy demonstrated that D-histidine inhibited biofilm formation and triggered the disassembly of mature biofilms. Notably, D-histidine increased the susceptibility of PAO1 to amikacin compared to that in the amikacin-alone group. These findings underscore the efficacy of D-histidine in combating Pseudomonas aeruginosa by reducing biofilm formation and increasing biofilm disassembly. Moreover, the combination of amikacin and D-histidine induced a synergistic effect against Pseudomonas aeruginosa biofilms, suggesting the potential utility of D-histidine as a preventive strategy against biofilm-associated infections caused by Pseudomonas aeruginosa.


Subject(s)
Amikacin , Pseudomonas Infections , Humans , Amikacin/pharmacology , Amikacin/metabolism , Amikacin/therapeutic use , Pseudomonas aeruginosa , Histidine/pharmacology , Histidine/metabolism , Histidine/therapeutic use , Biofilms , Quorum Sensing , Anti-Bacterial Agents/chemistry , Pseudomonas Infections/microbiology , Virulence Factors/metabolism
19.
mBio ; 15(2): e0316523, 2024 Feb 14.
Article in English | MEDLINE | ID: mdl-38236031

ABSTRACT

Hospital-acquired pneumonia (HAP) is a leading cause of morbidity and mortality, commonly caused by Pseudomonas aeruginosa. Meropenem is a commonly used therapeutic agent, although emergent resistance occurs during treatment. We used a rabbit HAP infection model to assess the bacterial kill and resistance pharmacodynamics of meropenem. Meropenem 5 mg/kg administered subcutaneously (s.c.) q8h (±amikacin 3.33-5 mg/kg q8h administered intravenously[i.v.]) or meropenem 30 mg/kg s.c. q8h regimens were assessed in a rabbit lung infection model infected with P. aeruginosa, with bacterial quantification and phenotypic/genotypic characterization of emergent resistant isolates. The pharmacokinetic/pharmacodynamic output was fitted to a mathematical model, and human-like regimens were simulated to predict outcomes in a clinical context. Increasing meropenem monotherapy demonstrated a dose-response effect to bacterial kill and an inverted U relationship with emergent resistance. The addition of amikacin to meropenem suppressed the emergence of resistance. A network of porin loss, efflux upregulation, and increased expression of AmpC was identified as the mechanism of this emergent resistance. A bridging simulation using human pharmacokinetics identified meropenem 2 g i.v. q8h as the licensed clinical regimen most likely to suppress resistance. We demonstrate an innovative experimental platform to phenotypically and genotypically characterize bacterial emergent resistance pharmacodynamics in HAP. For meropenem, we have demonstrated the risk of resistance emergence during therapy and identified two mitigating strategies: (i) regimen intensification and (ii) use of combination therapy. This platform will allow pre-clinical assessment of emergent resistance risk during treatment of HAP for other antimicrobials, to allow construction of clinical regimens that mitigate this risk.IMPORTANCEThe emergence of antimicrobial resistance (AMR) during antimicrobial treatment for hospital-acquired pneumonia (HAP) is a well-documented problem (particularly in pneumonia caused by Pseudomonas aeruginosa) that contributes to the wider global antimicrobial resistance crisis. During drug development, regimens are typically determined by their sufficiency to achieve bactericidal effect. Prevention of the emergence of resistance pharmacodynamics is usually not characterized or used to determine the regimen. The innovative experimental platform described here allows characterization of the emergence of AMR during the treatment of HAP and the development of strategies to mitigate this. We have demonstrated this specifically for meropenem-a broad-spectrum antibiotic commonly used to treat HAP. We have characterized the antimicrobial resistance pharmacodynamics of meropenem when used to treat HAP, caused by initially meropenem-susceptible P. aeruginosa, phenotypically and genotypically. We have also shown that intensifying the regimen and using combination therapy are both strategies that can both treat HAP and suppress the emergence of resistance.


Subject(s)
Cross Infection , Healthcare-Associated Pneumonia , Pseudomonas Infections , Animals , Humans , Rabbits , Meropenem/pharmacology , Pseudomonas aeruginosa , Amikacin/pharmacology , Amikacin/therapeutic use , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Pseudomonas Infections/drug therapy , Pseudomonas Infections/microbiology , Healthcare-Associated Pneumonia/drug therapy , Microbial Sensitivity Tests
SELECTION OF CITATIONS
SEARCH DETAIL