1
|
Dartois V, Dick T. Toward better cures for Mycobacterium abscessus lung disease. Clin Microbiol Rev 2024:e0008023. [PMID: 39360834 DOI: 10.1128/cmr.00080-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/30/2024] Open
Abstract
SUMMARYThe opportunistic pathogen Mycobacterium abscessus (Mab) causes fatal lung infections that bear similarities-and notable differences-with tuberculosis (TB) pulmonary disease. In contrast to TB, no antibiotic is formally approved to treat Mab disease, there is no reliable cure, and the discovery and development pipeline is incredibly thin. Here, we discuss the factors behind the unsatisfactory cure rates of Mab disease, namely intrinsic resistance and persistence of the pathogen, and the use of underperforming, often parenteral and toxic, repurposed drugs. We propose preclinical strategies to build injectable-free sterilizing and safe regimens: (i) prioritize oral bactericidal antibiotic classes, with an initial focus on approved agents or advanced clinical candidates to provide immediate options for desperate patients, (ii) test drug combinations early, (iii) optimize novel leads specifically for M. abscessus, and (iv) consider pharmacokinetic-pharmacodynamic targets at the site of disease, the lung lesions in which drug tolerant bacterial populations reside. Knowledge and tool gaps in the preclinical drug discovery process are identified, including validated mouse models and computational platforms to enable in vitro mouse-human translation. We briefly discuss recent advances in clinical development, the need for readouts and biomarkers that correlate with cure, and clinical trial concepts adapted to the uniqueness of Mab patient populations for new regimen development. In an era when most pharmaceutical firms have withdrawn from antimicrobial drug discovery, the breakthroughs needed to fill the regimen development pipeline will likely come from partnerships between academia, biotech, pharma, non-profit organizations, and governments, with incentives that reward cooperation.
Collapse
Affiliation(s)
- Véronique Dartois
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, New Jersey, USA
- Department of Medical Sciences, Hackensack Meridian School of Medicine, Nutley, New Jersey, USA
| | - Thomas Dick
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, New Jersey, USA
- Department of Medical Sciences, Hackensack Meridian School of Medicine, Nutley, New Jersey, USA
- Department of Microbiology and Immunology, Georgetown University, Washington, DC, USA
| |
Collapse
|
2
|
Heine HS, Drusano G, Purcell BK, Anastasiou D, Tanaka SK, Serio AW. Omadacycline is active in vitro and in vivo against ciprofloxacin-resistant Bacillus anthracis. Antimicrob Agents Chemother 2024; 68:e0059524. [PMID: 39133023 PMCID: PMC11373220 DOI: 10.1128/aac.00595-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 07/13/2024] [Indexed: 08/13/2024] Open
Abstract
Bacillus anthracis, the causative agent of anthrax, is among the most likely bacterial pathogens to be used in a biological attack. Inhalation anthrax is a serious, life-threatening form of infection, and the mortality from acute inhaled anthrax can approach 100% if not treated early and aggressively. Food and Drug Administration-approved antibiotics indicated for post-exposure prophylaxis (PEP) or treatment of anthrax are limited. This study assessed the in vitro activity and in vivo efficacy of omadacycline and comparators against clinical isolates of B. anthracis, including a ciprofloxacin-resistant isolate. Minimum inhibitory concentrations (MICs) of omadacycline, ciprofloxacin, and doxycycline were determined against animal and human clinical isolates of B. anthracis, including the ciprofloxacin-resistant Ames strain BACr4-2. Mice were challenged with aerosolized BACr4-2 spores, and survival was monitored for 28 days post-challenge. Treatment was initiated 24 h after aerosol challenge and administered for 14 days. Omadacycline demonstrated in vitro activity against 53 B. anthracis isolates with an MIC range of ≤0.008-0.25 µg/mL, and an MIC50/MIC90 of 0.015/0.03 µg/mL. Consistent with this, omadacycline demonstrated in vivo efficacy in a PEP mouse model of inhalation anthrax caused by the Ames BACr4-2 ciprofloxacin-resistant B. anthracis isolate. Omadacycline treatment significantly increased survival compared with the vehicle control group and the ciprofloxacin treatment group. As antibiotic resistance rates continue to rise worldwide, omadacycline may offer an alternative PEP or treatment option against inhalation anthrax, including anthrax caused by antibiotic-resistant B. anthracis.
Collapse
Affiliation(s)
| | | | | | - Diane Anastasiou
- Paratek Pharmaceuticals, Inc., King of Prussia, Pennsylvania, USA
| | - S Ken Tanaka
- Paratek Pharmaceuticals, Inc., King of Prussia, Pennsylvania, USA
| | - Alisa W Serio
- Paratek Pharmaceuticals, Inc., King of Prussia, Pennsylvania, USA
| |
Collapse
|
3
|
Rimal B, Lippincott CK, Panthi CM, Xie Y, Keepers TR, Alley MRK, Lamichhane G. Efficacy of epetraborole against Mycobacteroides abscessus in a mouse model of lung infection. Antimicrob Agents Chemother 2024; 68:e0064824. [PMID: 39016592 PMCID: PMC11323969 DOI: 10.1128/aac.00648-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 06/25/2024] [Indexed: 07/18/2024] Open
Abstract
Mycobacteroides abscessus (Mab or Mycobacterium abscessus) is a fast-growing mycobacterium that is ubiquitous in the environment and can cause opportunistic disease in people with lung comorbidity and immunodeficiency. There are no Food and Drug Administration-approved drugs for this disease, and repurposed antibiotics have a poor microbiological response. To address the need for effective new antibiotics, we determined the efficacy of epetraborole (EBO) against three Mab clinical isolates in a mouse model of lung Mab infection. Reduction in lung Mab burden over 4 weeks of treatment was the study end point. EBO was administered orally once daily at doses of 25 and 50 mg/kg, which achieved exposures approximating the once-daily dosing of 250 mg and 500 mg, respectively, in humans. EBO administration led to a gradual reduction in the lung Mab burden. After 4 weeks of treatment, the efficacies of 25 and 50 mg/kg EBO against isolates ATCC 19977 and M9501 were comparable. However, against isolate M9530, 50 mg/kg EBO was more efficacious than 25 mg/kg and comparable with parenteral imipenem, one of the most efficacious antibiotics against Mab. We also undertook a dose-ranging study by evaluating the efficacies of once-daily oral administration of 0.5, 5, 10, 25, and 100 mg/kg EBO against M9501 over 4 weeks. Once-daily oral 100 mg/kg EBO was as effective as twice-daily 100 mg/kg imipenem injection. Our study suggests that EBO could address the unmet need for effective oral treatment options for Mab lung disease, given the high rates of Mab drug resistance and limited tolerable intravenous options.
Collapse
Affiliation(s)
- Binayak Rimal
- Division of Infectious Diseases, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Christopher K. Lippincott
- Division of Infectious Diseases, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
- Center for Nontuberculous Mycobacteria and Bronchiectasis, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Chandra M. Panthi
- Division of Infectious Diseases, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Yi Xie
- Division of Infectious Diseases, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | | | - MRK Alley
- AN2 Therapeutics, Inc., Menlo Park, California, USA
| | - Gyanu Lamichhane
- Division of Infectious Diseases, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
- Center for Nontuberculous Mycobacteria and Bronchiectasis, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
4
|
Ignatius EH, Rimal B, Panthi CM, Belz DC, Lippincott CK, Deck DH, Serio AW, Lamichhane G. Efficacies of omadacycline + amikacin + imipenem and an all-oral regimen omadacycline + clofazimine + linezolid in a mouse model of M. abscessus lung disease. mSphere 2024; 9:e0038124. [PMID: 38980071 PMCID: PMC11288010 DOI: 10.1128/msphere.00381-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 06/26/2024] [Indexed: 07/10/2024] Open
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.
Collapse
Affiliation(s)
- Elisa H. Ignatius
- Division of Infectious Diseases, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
- Center for Nontuberculous Mycobacteria and Bronchiectasis, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
- Division of Clinical Pharmacology, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Binayak Rimal
- Division of Infectious Diseases, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Chandra M. Panthi
- Division of Infectious Diseases, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Daniel C. Belz
- Center for Nontuberculous Mycobacteria and Bronchiectasis, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Christopher K. Lippincott
- Division of Infectious Diseases, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
- Center for Nontuberculous Mycobacteria and Bronchiectasis, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Daniel H. Deck
- Paratek Pharmaceuticals Inc., King of Prussia, Pennsylvania, USA
| | - Alisa W. Serio
- Paratek Pharmaceuticals Inc., King of Prussia, Pennsylvania, USA
| | - Gyanu Lamichhane
- Division of Infectious Diseases, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
- Center for Nontuberculous Mycobacteria and Bronchiectasis, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
5
|
Dartois V, Bonfield TL, Boyce JP, Daley CL, Dick T, Gonzalez-Juarrero M, Gupta S, Kramnik I, Lamichhane G, Laughon BE, Lorè NI, Malcolm KC, Olivier KN, Tuggle KL, Jackson M. Preclinical murine models for the testing of antimicrobials against Mycobacterium abscessus pulmonary infections: Current practices and recommendations. Tuberculosis (Edinb) 2024; 147:102503. [PMID: 38729070 PMCID: PMC11168888 DOI: 10.1016/j.tube.2024.102503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 03/08/2024] [Accepted: 03/17/2024] [Indexed: 05/12/2024]
Abstract
Mycobacterium abscessus, a rapidly growing nontuberculous mycobacterium, is increasingly recognized as an important pathogen of the human lung, disproportionally affecting people with cystic fibrosis (CF) and other susceptible individuals with non-CF bronchiectasis and compromised immune functions. M. abscessus infections are extremely difficult to treat due to intrinsic resistance to many antibiotics, including most anti-tuberculous drugs. Current standard-of-care chemotherapy is long, includes multiple oral and parenteral repurposed drugs, and is associated with significant toxicity. The development of more effective oral antibiotics to treat M. abscessus infections has thus emerged as a high priority. While murine models have proven instrumental in predicting the efficacy of therapeutic treatments for M. tuberculosis infections, the preclinical evaluation of drugs against M. abscessus infections has proven more challenging due to the difficulty of establishing a progressive, sustained, pulmonary infection with this pathogen in mice. To address this issue, a series of three workshops were hosted in 2023 by the Cystic Fibrosis Foundation (CFF) and the National Institute of Allergy and Infectious Diseases (NIAID) to review the current murine models of M. abscessus infections, discuss current challenges and identify priorities toward establishing validated and globally harmonized preclinical models. This paper summarizes the key points from these workshops. The hope is that the recommendations that emerged from this exercise will facilitate the implementation of informative murine models of therapeutic efficacy testing across laboratories, improve reproducibility from lab-to-lab and accelerate preclinical-to-clinical translation.
Collapse
Affiliation(s)
- Véronique Dartois
- Center for Discovery and Innovation & Department of Medical Sciences, Hackensack Meridian School of Medicine, Hackensack Meridian Health, Nutley, NJ, USA.
| | - Tracey L Bonfield
- Genetics and Genome Sciences and National Center for Regenerative Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Jim P Boyce
- Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Charles L Daley
- Department of Medicine, National Jewish Health, Denver, CO, USA; Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| | - Thomas Dick
- Center for Discovery and Innovation & Department of Medical Sciences, Hackensack Meridian School of Medicine, Hackensack Meridian Health, Nutley, NJ, USA; Department of Microbiology and Immunology, Georgetown University, Washington, DC, USA
| | - Mercedes Gonzalez-Juarrero
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, 80523-1682, USA
| | - Shashank Gupta
- Laboratory of Chronic Airway Infection, Pulmonary Branch, National Heart, Lung, and Blood Institute, Bethesda, MD, USA; Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Igor Kramnik
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, 02215, USA; Department of Medicine, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Gyanu Lamichhane
- Division of Infectious Diseases, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Barbara E Laughon
- Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Nicola I Lorè
- Emerging Bacterial Pathogens Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Kenneth C Malcolm
- Department of Medicine, National Jewish Health, Denver, CO, USA; Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| | - Kenneth N Olivier
- Department of Medicine, Division of Pulmonary Diseases and Critical Care Medicine, University of North Carolina, USA; Marsico Lung Institute, Chapel Hill, 27599-7248, NC, USA
| | | | - Mary Jackson
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, 80523-1682, USA.
| |
Collapse
|
6
|
Boorgula GD, Gumbo T, Singh S, McShane PJ, Philley JV, Srivastava S. Omadacycline drug susceptibility testing for non-tuberculous mycobacteria using oxyrase to overcome challenges with drug degradation. Tuberculosis (Edinb) 2024; 147:102519. [PMID: 38754247 PMCID: PMC11345947 DOI: 10.1016/j.tube.2024.102519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 05/01/2024] [Accepted: 05/12/2024] [Indexed: 05/18/2024]
Abstract
BACKGROUND Drug susceptibility testing (DST) protocol of omadacycline against non-tuberculous mycobacteria has not yet been established. We developed a method to accurately determine MIC omadacycline MIC against Mycobacterium abscessus (Mab), Mycobacterium avium-complex (MAC), and Mycobacterium kansasii (Mkn). METHODS First, we identified the oxyrase concentration not affecting Mab, MAC, and Mkn growth followed by omadacycline MIC experiments with and without oxyrase using reference and clinical strains. RESULTS Oxyrase 0.5 % (v/v) stabilized omadacycline in the culture medium. The median omadacycline MIC was 1 mg/L for Mab and 8 mg/L for Mkn. For MAC, the median omadacycline MIC was 2 mg/L for M. avium, 256 mg/L for M. intracellulare, and 4 mg/L for M. chimaera (p < 0.0001). Wilcoxon matched-pairs signed rank test revealed statistically lower MICs with oxyrase for all MAC subspecies (p < 0.0001), all Mab subspecies (p < 0.0001), and Mkn (p = 0.0002). The decrease in MICs with oxyrase was 17/18 of Mab, 14/19 of Mkn, 8/8 of M. avium, 4/5 M. chimera, but only 11/18 of M. intracellulare (p < 0.013). CONCLUSION Use of 0.5 % oxyrase could be a potential solution to reliable and reproducible omadacycline MIC of Mab. However, oxyrase demonstrated a variable effect in reducing MICs against MAC and Mkn.
Collapse
Affiliation(s)
- Gunavanthi D Boorgula
- Department of Medicine, University of Texas at Tyler School of Medicine, Tyler, TX, USA
| | - Tawanda Gumbo
- Mathematical Modeling and AI Department, Praedicare Inc., Dallas, TX, USA; Hollow Fiber System & Experimental Therapeutics Laboratories, Wet Lab Systems, Praedicare Inc., Dallas, TX, USA.
| | - Sanjay Singh
- Department of Medicine, University of Texas at Tyler School of Medicine, Tyler, TX, USA
| | - Pamela J McShane
- Department of Medicine, Section of Pulmonary and Critical Care, University of Texas at Tyler School of Medicine, Tyler, TX, USA
| | - Julie V Philley
- Department of Medicine, Section of Pulmonary and Critical Care, University of Texas at Tyler School of Medicine, Tyler, TX, USA
| | - Shashikant Srivastava
- Department of Medicine, University of Texas at Tyler School of Medicine, Tyler, TX, USA; Department of Cellular and Molecular Biology, University of Texas Health Science Centre at Tyler, Tyler, TX, USA.
| |
Collapse
|
7
|
Wang M, Men P, Zhang W, Wu J, Gu Y, Wang F, Huang H, Yu X, Duan H. Bedaquiline susceptibility testing of Mycobacterium abscessus complex and Mycobacterium avium complex: A meta-analysis study. J Glob Antimicrob Resist 2024; 37:135-140. [PMID: 38561143 DOI: 10.1016/j.jgar.2024.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 03/01/2024] [Accepted: 03/17/2024] [Indexed: 04/04/2024] Open
Abstract
OBJECTIVE This study aims to estimate the overall in vitro activity of bedaquiline (BDQ) against clinical isolates of Mycobacterium abscessus complex (MABS) and M. avium complex (MAC), considering BDQ as a repurposed drug for non-tuberculous mycobacteria (NTM) infections. METHODS We conducted a systematic review of publications in PubMed/ MEDLINE, Web of Science, and Embase up to 15 April 2023. Studies were included if they followed the Clinical and Laboratory Standards Institute (CLSI) criteria for drug susceptibility testing (DST). Using a random effects model, we assessed the overall in vitro BDQ resistance rate in clinical isolates of MABS and MAC. Sources of heterogeneity were analysed using Cochran's Q and the I2 statistic. All analyses were performed using CMA V3.0. RESULTS A total of 24 publications (19 reports for MABS and 11 for MAC) were included. Using 1 µg/mL and 2 µg/mL as the breakpoint for BDQ resistance, the pooled rates of in vitro BDQ resistance in clinical isolates of MABS were found to be 1.8% (95% confidence interval [CI], 0.7-4.6%) and 1.7% (95% CI, 0.6-4.4%), respectively. In the case of MAC, the pooled rates were 1.7% (95% CI, 0.4-6.9%) and 1.6% (95% CI, 0.4-6.8%) for 1 µg/mL and 2 µg/mL, respectively. CONCLUSION This study reports the prevalence of BDQ resistance in clinical isolates of MABS and MAC. The findings suggest that BDQ holds potential as a repurposed drug for treating MABS and MAC infections.
Collapse
Affiliation(s)
- Ming Wang
- Department of Tuberculosis, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Peixuan Men
- Institute of Medical Information/Medical Library, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Weihe Zhang
- National Clinical Laboratory on Tuberculosis, Beijing Key Laboratory for Drug-Resistant Tuberculosis Research, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Institute, Beijing, China
| | - Jing Wu
- National Clinical Laboratory on Tuberculosis, Beijing Key Laboratory for Drug-Resistant Tuberculosis Research, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Institute, Beijing, China
| | - Yuzhen Gu
- National Clinical Laboratory on Tuberculosis, Beijing Key Laboratory for Drug-Resistant Tuberculosis Research, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Institute, Beijing, China
| | - Fen Wang
- National Clinical Laboratory on Tuberculosis, Beijing Key Laboratory for Drug-Resistant Tuberculosis Research, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Institute, Beijing, China
| | - Hairong Huang
- National Clinical Laboratory on Tuberculosis, Beijing Key Laboratory for Drug-Resistant Tuberculosis Research, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Institute, Beijing, China
| | - Xia Yu
- National Clinical Laboratory on Tuberculosis, Beijing Key Laboratory for Drug-Resistant Tuberculosis Research, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Institute, Beijing, China
| | - Hongfei Duan
- Department of Tuberculosis, Beijing Chest Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
8
|
Tunesi S, Zelazny A, Awad Z, Mougari F, Buyck JM, Cambau E. Antimicrobial susceptibility of Mycobacterium abscessus and treatment of pulmonary and extra-pulmonary infections. Clin Microbiol Infect 2024; 30:718-725. [PMID: 37797824 DOI: 10.1016/j.cmi.2023.09.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 09/17/2023] [Accepted: 09/27/2023] [Indexed: 10/07/2023]
Abstract
BACKGROUND Mycobacterium abscessus (MAB) is the mycobacterial species least susceptible to antimicrobials. Infections are difficult to treat, and cure rates are below 50% even after a combination of 4-5 drugs for many months. OBJECTIVES To examine antimicrobial susceptibilities and treatment recommendations in light of what is known about mechanisms of resistance and pharmacodynamics/pharmacokinetics (PK/PD) interactions. SOURCES Original papers on the topics of 'antimicrobials', 'susceptibility', 'treatment', and 'outcome' from 2019 onwards, in the context of the evidence brought by the guidelines published in 2020 for pulmonary infections. CONTENT MAB is susceptible in vitro to only a few antimicrobials. Breakpoints were set by the Clinical and Laboratory Standards Institute and are revised by the European Committee on Antimicrobial Susceptibility Testing for epidemiological cut-off values. Innate resistance is due to multiple resistance mechanisms involving efflux pumps, inactivating enzymes, and low drug-target affinity. In addition, MAB may display acquired resistance to macrolides and amikacin through mutations in drug binding sites. Treatment outcomes are better for macrolide-based combinations and MAB subspecies massiliense. New compounds in the family of cyclines, oxazolidinones, and penem-β-lactamase inhibitor combinations (described in another paper), as well as bedaquiline, a new antituberculous agent, are promising, but their efficacy remains to be proven. PK/PD studies, which are critical for establishing optimal dosing regimens, were mainly done for monotherapy and healthy individuals. IMPLICATIONS Medical evidence is poor, and randomized clinical trials or standardized cohorts are needed to compare outcomes of patients with similar underlying disease, clinical characteristics, and identified MAB subspecies/sequevar. Microbiological diagnosis and susceptibility testing need to be harmonized to enable the comparison of agents and the testing of new compounds. Testing antimicrobial combinations requires new methods, especially for PK/PD parameters. Molecular testing may help in assessing MAB resistance prior to treatment. New antimicrobials need to be systematically tested against MAB to find an effective antimicrobial regimen.
Collapse
Affiliation(s)
- Simone Tunesi
- UOC Malattie infettive, Azienda Ospedaliera SS Antonio e Biagio e Cesare Arrigo, Alessandria, Italy
| | - Adrian Zelazny
- Microbiology Service, Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Zeina Awad
- Service de mycobactériologie spécialisée et de référence, Laboratoire associé du CNR des mycobactéries et de la résistance des mycobactéries aux antituberculeux (CNR-MyRMA) APHP GHU Paris Nord, Hôpital Bichat, Paris, France
| | - Faiza Mougari
- Service de mycobactériologie spécialisée et de référence, Laboratoire associé du CNR des mycobactéries et de la résistance des mycobactéries aux antituberculeux (CNR-MyRMA) APHP GHU Paris Nord, Hôpital Bichat, Paris, France
| | - Julien M Buyck
- Université de Poitiers, PHAR2, Inserm UMR 1070, Poitiers, France
| | - Emmanuelle Cambau
- Service de mycobactériologie spécialisée et de référence, Laboratoire associé du CNR des mycobactéries et de la résistance des mycobactéries aux antituberculeux (CNR-MyRMA) APHP GHU Paris Nord, Hôpital Bichat, Paris, France; Université Paris Cité, IAME, Inserm UMR 1137, Paris, France.
| |
Collapse
|
9
|
Calcagno A, Coppola N, Sarmati L, Tadolini M, Parrella R, Matteelli A, Riccardi N, Trezzi M, Di Biagio A, Pirriatore V, Russo A, Gualano G, Pontali E, Surace L, Falbo E, Mencarini J, Palmieri F, Gori A, Schiuma M, Lapadula G, Goletti D. Drugs for treating infections caused by non-tubercular mycobacteria: a narrative review from the study group on mycobacteria of the Italian Society of Infectious Diseases and Tropical Medicine. Infection 2024; 52:737-765. [PMID: 38329686 PMCID: PMC11142973 DOI: 10.1007/s15010-024-02183-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 01/12/2024] [Indexed: 02/09/2024]
Abstract
BACKGROUND Non-tuberculous mycobacteria (NTM) are generally free-living organism, widely distributed in the environment, with sporadic potential to infect. In recent years, there has been a significant increase in the global incidence of NTM-related disease, spanning across all continents and an increased mortality after the diagnosis has been reported. The decisions on whether to treat or not and which drugs to use are complex and require a multidisciplinary approach as well as patients' involvement in the decision process. METHODS AND RESULTS This review aims at describing the drugs used for treating NTM-associated diseases emphasizing the efficacy, tolerability, optimization strategies as well as possible drugs that might be used in case of intolerance or resistance. We also reviewed data on newer compounds highlighting the lack of randomised clinical trials for many drugs but also encouraging preliminary data for others. We also focused on non-pharmacological interventions that need to be adopted during care of individuals with NTM-associated diseases CONCLUSIONS: Despite insufficient efficacy and poor tolerability this review emphasizes the improvement in patients' care and the needs for future studies in the field of anti-NTM treatments.
Collapse
Affiliation(s)
- A Calcagno
- Unit of Infectious Diseases, Department of Medical Sciences, University of Turin, Turin, Italy.
- Stop TB Italy, Milan, Italy.
| | - N Coppola
- Infectious Diseases Unit, Section of Infectious Diseases, Department of Mental Health and Public Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - L Sarmati
- Department of System Medicine, Tor Vergata University and Infectious Disease Clinic, Policlinico Tor Vergata, Rome, Italy
| | - M Tadolini
- Stop TB Italy, Milan, Italy
- Infectious Diseases Unit, IRCCS Azienda Ospedaliero-Universitaria Di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - R Parrella
- Stop TB Italy, Milan, Italy
- Respiratory Infectious Diseases Unit, Cotugno Hospital, A. O. R. N. dei Colli, Naples, Italy
| | - A Matteelli
- Institute of Infectious and Tropical Diseases, WHO Collaborating Centre for TB Prevention, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - N Riccardi
- Stop TB Italy, Milan, Italy
- Infectious Diseases Unit, Department of Clinical and Experimental Medicine, Azienda Ospedaliero Universitaria Pisana, University of Pisa, Pisa, Italy
| | - M Trezzi
- Stop TB Italy, Milan, Italy
- Infectious and Tropical Diseases Unit, Department of Medical Sciences, Azienda Ospedaliero-Universitaria Senese, Siena, Italy
| | - A Di Biagio
- Infectious Diseases Unit, San Martino Policlinico Hospital-IRCCS for Oncology and Neurosciences, Genoa, Italy
- Department of Health Sciences (DISSAL), University of Genoa, Genoa, Italy
| | - V Pirriatore
- Stop TB Italy, Milan, Italy
- Unit of Infectious Diseases, "DivisioneA", Ospedale Amedeo di Savoia, ASL CIttà di Torino, Turin, Italy
| | - A Russo
- Infectious Diseases Unit, Section of Infectious Diseases, Department of Mental Health and Public Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - G Gualano
- Stop TB Italy, Milan, Italy
- Respiratory Infectious Diseases Unit, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - E Pontali
- Department of Infectious Diseases, Galliera Hospital, Genoa, Italy
| | - L Surace
- Stop TB Italy, Milan, Italy
- Dipartimento Di Prevenzione, Azienda Sanitaria Provinciale di Catanzaro, Centro di Medicina del Viaggiatore e delle Migrazioni, P. O. Giovanni Paolo II, Lamezia Terme, CZ, Italy
| | - E Falbo
- Stop TB Italy, Milan, Italy
- Dipartimento Di Prevenzione, Azienda Sanitaria Provinciale di Catanzaro, Centro di Medicina del Viaggiatore e delle Migrazioni, P. O. Giovanni Paolo II, Lamezia Terme, CZ, Italy
| | - J Mencarini
- Infectious and Tropical Diseases Unit, Careggi University Hospital, Florence, Italy
| | - F Palmieri
- Respiratory Infectious Diseases Unit, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - A Gori
- Dipartimento di Fisiopatologia Medico-Chirurgica e dei Trapianti, ASST Fatebenefratelli Sacco-Ospedale Luigi Sacco-Polo Universitario and Università Degli Studi di Milano, Milano, Italy
| | - M Schiuma
- Dipartimento di Fisiopatologia Medico-Chirurgica e dei Trapianti, ASST Fatebenefratelli Sacco-Ospedale Luigi Sacco-Polo Universitario and Università Degli Studi di Milano, Milano, Italy
| | - G Lapadula
- Infectious Diseases Unit, Fondazione IRCCS San Gerardo dei Tintori, University of Milano-Bicocca, Monza, Italy
| | - D Goletti
- Stop TB Italy, Milan, Italy
- Translational Research Unit, Epidemiology Department, National Institute for Infectious Diseases-IRCCS L. Spallanzani, Rome, Italy
| |
Collapse
|
10
|
Rimal B, Panthi CM, Xie Y, Belz DC, Ignatius EH, Lippincott CK, Deck DH, Serio AW, Lamichhane G. Efficacies of three drug regimens containing omadacycline to treat Mycobacteroides abscessus disease. Tuberculosis (Edinb) 2024; 146:102482. [PMID: 38364332 DOI: 10.1016/j.tube.2024.102482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 01/11/2024] [Accepted: 01/28/2024] [Indexed: 02/18/2024]
Abstract
Mycobacteroides abscessus (Mab, also known as Mycobacterium abscessus) causes opportunistic pulmonary and soft tissue infections that are difficult to cure with existing treatments. Omadacycline, a new tetracycline antibiotic, exhibits potent in vitro and in vivo activity against Mab. As regimens containing multiple antibiotics are required to produce a durable cure for Mab disease, we assessed efficacies of three three-drug combinations in a pre-clinical mouse model of pulmonary Mab disease to identify companion drugs with which omadacycline exhibits the highest efficacy. Additionally, we assessed the susceptibility of Mab recovered from mouse lungs after four weeks of exposure to the three triple-drug regimens. Among the three-drug regimens, omadacycline + imipenem + amikacin produced the largest reduction in Mab burden, whereas omadacycline + imipenem + linezolid exhibited the most effective early bactericidal activity. Omadacycline + linezolid + clofazimine, a regimen that can be administered orally, lacked early bactericidal activity but produced a gradual reduction in the lung Mab burden over time. The robust efficacy exhibited by these three regimens in the mouse model supports their further evaluation in patients with Mab lung disease. As we were unable to isolate drug-resistant Mab mutants at the completion of four weeks of treatment, these triple-drug combinations show promise of producing durable cure and minimizing selection of resistant mutants.
Collapse
Affiliation(s)
- Binayak Rimal
- Division of Infectious Diseases, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, 21287, USA
| | - Chandra M Panthi
- Division of Infectious Diseases, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, 21287, USA
| | - Yi Xie
- Division of Infectious Diseases, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, 21287, USA
| | - Daniel C Belz
- Center for Nontuberculous Mycobacteria and Bronchiectasis, School of Medicine, Johns Hopkins University, Baltimore, MD, 21287, USA; Division of Pulmonary and Critical Care Medicine, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, 21287, USA
| | - Elisa H Ignatius
- Division of Infectious Diseases, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, 21287, USA; Center for Nontuberculous Mycobacteria and Bronchiectasis, School of Medicine, Johns Hopkins University, Baltimore, MD, 21287, USA; Division of Clinical Pharmacology, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, 21287, USA
| | - Christopher K Lippincott
- Division of Infectious Diseases, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, 21287, USA; Center for Nontuberculous Mycobacteria and Bronchiectasis, School of Medicine, Johns Hopkins University, Baltimore, MD, 21287, USA
| | - Daniel H Deck
- Paratek Pharmaceuticals Inc, King of Prussia, PA, 19406, USA
| | - Alisa W Serio
- Paratek Pharmaceuticals Inc, King of Prussia, PA, 19406, USA
| | - Gyanu Lamichhane
- Division of Infectious Diseases, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, 21287, USA; Center for Nontuberculous Mycobacteria and Bronchiectasis, School of Medicine, Johns Hopkins University, Baltimore, MD, 21287, USA.
| |
Collapse
|
11
|
Nguyen TQ, Heo BE, Jeon S, Ash A, Lee H, Moon C, Jang J. Exploring antibiotic resistance mechanisms in Mycobacterium abscessus for enhanced therapeutic approaches. Front Microbiol 2024; 15:1331508. [PMID: 38380095 PMCID: PMC10877060 DOI: 10.3389/fmicb.2024.1331508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 01/17/2024] [Indexed: 02/22/2024] Open
Abstract
Mycobacterium abscessus, a leading cause of severe lung infections in immunocompromised individuals, poses significant challenges for current therapeutic strategies due to resistance mechanisms. Therefore, understanding the intrinsic and acquired antibiotic resistance of M. abscessus is crucial for effective treatment. This review highlights the mechanisms employed by M. abscessus to sustain antibiotic resistance, encompassing not only conventional drugs but also newly discovered drug candidates. This comprehensive analysis aims to identify novel entities capable of overcoming the notorious resistance exhibited by M. abscessus, providing insights for the development of more effective therapeutic interventions.
Collapse
Affiliation(s)
- Thanh Quang Nguyen
- Division of Life Science, Department of Bio & Medical Big Data (BK21 Four Program), Research Institute of Life Science, Gyeongsang National University, Jinju, Republic of Korea
| | - Bo Eun Heo
- Division of Life Science, Department of Bio & Medical Big Data (BK21 Four Program), Research Institute of Life Science, Gyeongsang National University, Jinju, Republic of Korea
| | - Seunghyeon Jeon
- Division of Life Science, Department of Bio & Medical Big Data (BK21 Four Program), Research Institute of Life Science, Gyeongsang National University, Jinju, Republic of Korea
| | - Anwesha Ash
- Division of Life Science, Department of Bio & Medical Big Data (BK21 Four Program), Research Institute of Life Science, Gyeongsang National University, Jinju, Republic of Korea
| | - Heehyun Lee
- Division of Life Science, Department of Bio & Medical Big Data (BK21 Four Program), Research Institute of Life Science, Gyeongsang National University, Jinju, Republic of Korea
| | - Cheol Moon
- Department of Clinical Laboratory Science, Semyung University, Jecheon, Republic of Korea
| | - Jichan Jang
- Division of Life Science, Department of Bio & Medical Big Data (BK21 Four Program), Research Institute of Life Science, Gyeongsang National University, Jinju, Republic of Korea
| |
Collapse
|
12
|
Adhikrao PA, Motiram GM, Kumar G. Tackling Nontuberculous Mycobacteria by Repurposable Drugs and Potential Leads from Natural Products. Curr Top Med Chem 2024; 24:1291-1326. [PMID: 38288807 DOI: 10.2174/0115680266276938240108060247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 12/12/2023] [Accepted: 12/19/2023] [Indexed: 07/25/2024]
Abstract
Nontuberculous Mycobacteria (NTM) refer to bacteria other than all Mycobacterium species that do not cause tuberculosis or leprosy, excluding the species of the Mycobacterium tuberculosis complex, M. leprae and M. lepromatosis. NTM are ubiquitous and present in soils and natural waters. NTM can survive in a wide range of environmental conditions. The direct inoculum of the NTM from water or other materials is most likely a source of infections. NTMs are responsible for several illnesses, including pulmonary alveolar proteinosis, cystic fibrosis, bronchiectasis, chronic obstructive pneumoconiosis, and pulmonary disease. Recent reports suggest that NTM species have become insensitive to sterilizing agents, antiseptics, and disinfectants. The efficacy of existing anti-NTM regimens is diminishing and has been compromised due to drug resistance. New and recurring cases of multidrug-resistant NTM strains are increasing. Thus, there is an urgent need for ant-NTM regimens with novel modes of action. This review sheds light on the mode of antimicrobial resistance in the NTM species. Then, we discussed the repurposable drugs (antibiotics) that have shown new indications (activity against NTM strains) that could be developed for treating NTM infections. Also, we have summarised recently identified natural leads acting against NTM, which have the potential for treating NTM-associated infections.
Collapse
Affiliation(s)
- Patil Amruta Adhikrao
- Department of Natural Products, Chemical Sciences, National Institute of Pharmaceutical Education and Research-Hyderabad, Hyderabad, Balanagar, 500037, India
| | - Gudle Mayuri Motiram
- Department of Natural Products, Chemical Sciences, National Institute of Pharmaceutical Education and Research-Hyderabad, Hyderabad, Balanagar, 500037, India
| | - Gautam Kumar
- Department of Natural Products, Chemical Sciences, National Institute of Pharmaceutical Education and Research-Hyderabad, Hyderabad, Balanagar, 500037, India
| |
Collapse
|
13
|
Kaur P, Krishnamurthy RV, Shandil RK, Mohan R, Narayanan S. A Novel Inhibitor against the Biofilms of Non-Tuberculous Mycobacteria. Pathogens 2023; 13:40. [PMID: 38251347 PMCID: PMC10819454 DOI: 10.3390/pathogens13010040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 12/16/2023] [Accepted: 12/18/2023] [Indexed: 01/23/2024] Open
Abstract
Non-tuberculous Mycobacteria (NTM), previously classified as environmental microbes, have emerged as opportunistic pathogens causing pulmonary infections in immunocompromised hosts. The formation of the biofilm empowers NTM pathogens to escape from the immune response and antibiotic action, leading to treatment failures. NF1001 is a novel thiopeptide antibiotic first-in-class compound with potent activity against planktonic/replicating and biofilm forms of various NTM species. It is potent against both drug-sensitive and -resistant NTM. It has demonstrated a concentration-dependent killing of replicating and intracellularly growing NTM, and has inhibited and reduced the viability of NTM in biofilms. Combination studies using standard-of-care (SoC) drugs for NTM exhibited synergetic/additive effects, but no antagonism against both planktonic and biofilm populations of Mycobacterium abscessus and Mycobacterium avium. In summary, the activity of NF1001 alone or in combination with SoC drugs projects NF1001 as a promising candidate for the treatment of difficult-to-treat NTM pulmonary diseases (NTM-PD) and cystic fibrosis (CF) in patients.
Collapse
Affiliation(s)
- Parvinder Kaur
- Foundation for Neglected Disease Research (FNDR), Doddaballapur, Bengaluru 561203, Karnataka, India; (R.V.K.); (R.K.S.); (S.N.)
| | - Ramya Vadageri Krishnamurthy
- Foundation for Neglected Disease Research (FNDR), Doddaballapur, Bengaluru 561203, Karnataka, India; (R.V.K.); (R.K.S.); (S.N.)
| | - Radha Krishan Shandil
- Foundation for Neglected Disease Research (FNDR), Doddaballapur, Bengaluru 561203, Karnataka, India; (R.V.K.); (R.K.S.); (S.N.)
| | - Rahul Mohan
- National Center for Polar & Ocean Research (NCPOR), Headland Sada, Vasco da Gama 403802, Goa, India;
| | - Shridhar Narayanan
- Foundation for Neglected Disease Research (FNDR), Doddaballapur, Bengaluru 561203, Karnataka, India; (R.V.K.); (R.K.S.); (S.N.)
| |
Collapse
|
14
|
Ingram PR, Jones EE, Allen B, Murray RJ, Keehner TJ, Whitmore TJ. Omadacycline therapy for Mycobacterium abscessus species infections. Intern Med J 2023; 53:2257-2263. [PMID: 36917124 DOI: 10.1111/imj.16071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 03/10/2023] [Indexed: 03/16/2023]
Abstract
BACKGROUND Antimicrobial resistance and therapy-related adverse effects make Mycobacterium abscessus treatment challenging. Omadacycline is a novel, bioavailable aminomethylcycline with favourable in vitro activity against M. abscessus. AIMS To describe a case report and review the published literature describing outcomes for M. abscessus infections treated with omadacycline. METHODS Systematic literature review. RESULTS We identified three articles that, in addition to our case report, describe 18 patients. Pulmonary infections were most frequent. Minimum inhibitory concentrations were reported for two isolates (0.25 and 0.5 mg/L). Despite half the patients starting omadacycline because of failure of prior therapy, 15 (83%) had a favourable outcome, defined as 'cure', 'improvement' or 'clinical success' as determined by the primary study authors. One patient (6%) discontinued omadacycline because of gastrointestinal intolerance. CONCLUSIONS Although the limited observational data and in vitro susceptibility results are encouraging, randomised control trials are required to determine the role of omadacycline as part of combination therapy for this most difficult-to-treat pathogen.
Collapse
Affiliation(s)
- Paul R Ingram
- Royal Perth Hospital, Perth, Western Australia, Australia
- PathWest Laboratory Medicine, Perth, Western Australia, Australia
- University of Western Australia, Perth, Western Australia, Australia
| | - Eva E Jones
- Royal Perth Hospital, Perth, Western Australia, Australia
| | - Bethwyn Allen
- Royal Perth Hospital, Perth, Western Australia, Australia
| | - Ronan J Murray
- PathWest Laboratory Medicine, Perth, Western Australia, Australia
- University of Western Australia, Perth, Western Australia, Australia
- Sir Charles Gairdner Hospital, Perth, Western Australia, Australia
| | | | - Timothy J Whitmore
- Royal Perth Hospital, Perth, Western Australia, Australia
- Anita Clayton Centre, Perth, Western Australia, Australia
| |
Collapse
|
15
|
El Ghali A, Morrisette T, Alosaimy S, Lucas K, Tupayachi-Ortiz MG, Vemula R, Wadle C, Philley JV, Mejia-Chew C, Hamad Y, Stevens RW, Zeuli JD, Webb AJ, Fiske CT, Simonyan A, Cimino CL, Mammadova M, Umana VE, Hasbun R, Butt S, Molina KC, Thomas M, Kaip EA, Bouchard J, Gore TW, Howard C, Cabanilla MG, Holger DJ, Frens JJ, Barger M, Ong A, Cohen KA, Rybak MJ. Long-term evaluation of clinical success and safety of omadacycline in nontuberculous mycobacteria infections: a retrospective, multicenter cohort of real-world health outcomes. Antimicrob Agents Chemother 2023; 67:e0082423. [PMID: 37768312 PMCID: PMC10583686 DOI: 10.1128/aac.00824-23] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 07/13/2023] [Indexed: 09/29/2023] Open
Abstract
Infections due to nontuberculous mycobacteria (NTM) continue to increase in prevalence, leading to problematic clinical outcomes. Omadacycline (OMC) is an aminomethylcycline antibiotic with FDA orphan drug and fast-track designations for pulmonary NTM infections, including Mycobacteroides abscessus (MAB). This multicenter retrospective study across 16 U.S. medical institutions from January 2020 to March 2023 examined the long-term clinical success, safety, and tolerability of OMC for NTM infections. The cohort included patients aged ≥18 yr, who were clinically evaluable, and` had been treated with OMC for ≥3 mo without a previous diagnosis of cystic fibrosis. The primary outcome was 3 mo clinical success, with secondary outcomes including clinical improvement and mortality at 6- and 12 mo, persistence or reemergence of infection, adverse effects, and reasons for OMC utilization. Seventy-five patients were included in this analysis. Most patients were female (48/75, 64.0%) or Caucasian (58/75, 77.3%), with a median (IQR) age of 59 yr (49-67). Most had NTM pulmonary disease (33/75, 44.0%), skin and soft tissue disease (19/75, 25.3%), or osteomyelitis (10/75, 13.3%), and Mycobacterium abscessus (60/75, 80%) was the most commonly isolated NTM pathogen. The median (IQR) treatment duration was 6 mo (4 - 14), and the most commonly co-administered antibiotic was azithromycin (33/70, 47.1%). Three-month clinical success was observed in 80.0% (60/75) of patients, and AEs attributable to OMC occurred in 32.0% (24/75) of patients, leading to drug discontinuation in 9.3% (7/75).
Collapse
Affiliation(s)
- Amer El Ghali
- Anti-Infective Research Laboratory, Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan, USA
| | - Taylor Morrisette
- Department of Clinical Pharmacy & Outcomes Sciences, Medical University of South Carolina College of Pharmacy, Charleston, South Carolina, USA
- Department of Pharmacy Services, Medical University of South Carolina (MUSC) Health, Charleston, South Carolina, USA
| | - Sara Alosaimy
- Anti-Infective Research Laboratory, Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan, USA
| | - Kristen Lucas
- Anti-Infective Research Laboratory, Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan, USA
| | - Maria G. Tupayachi-Ortiz
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Raaga Vemula
- University of Texas Health Science Center, University of Texas, Tyler, Texas, USA
| | - Carly Wadle
- University of Texas Health Science Center, University of Texas, Tyler, Texas, USA
| | - Julie V. Philley
- University of Texas Health Science Center, University of Texas, Tyler, Texas, USA
| | - Carlos Mejia-Chew
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Yasir Hamad
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Ryan W. Stevens
- Department of Pharmacy, Mayo Clinic, Rochester, Minnesota, USA
| | - John D. Zeuli
- Department of Pharmacy, Mayo Clinic, Rochester, Minnesota, USA
| | - Andrew J. Webb
- Department of Pharmacy, Mayo Clinic, Rochester, Minnesota, USA
| | - Christina T. Fiske
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Anahit Simonyan
- Department of Pharmaceutical Services, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Christo L. Cimino
- Department of Pharmaceutical Services, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Mehriban Mammadova
- Division of Infectious Diseases, Department of Medicine, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Virginia E. Umana
- Division of Infectious Diseases, Department of Medicine, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Rodrigo Hasbun
- Division of Infectious Diseases, Department of Medicine, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Saira Butt
- Division of Infectious Diseases, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Kyle C. Molina
- Department of Emergency Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Michael Thomas
- Department of Emergency Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Emily A. Kaip
- Department of Pharmaceutical Services, University of California, San Francisco Medical Center, San Francisco, North Carolina, USA
| | - Jeannette Bouchard
- College of Pharmacy, University of South Carolina, Columbia, South Carolina, USA
| | - Tristan W. Gore
- College of Pharmacy, University of South Carolina, Columbia, South Carolina, USA
| | - Catessa Howard
- Department of Pharmacy, West Virginia University Medicine, Morgantown, West Virginia, USA
| | - M. Gabriela Cabanilla
- Division of Infectious Diseases, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA
| | - Dana J. Holger
- Department of Pharmacy Practice, Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, Florida, USA
| | - Jeremy J. Frens
- Department of Pharmacy Services, Cone Health, Greensboro, North Carolina, USA
| | - Melissa Barger
- Department of Medicine, Ventura County Medical Center, Ventura, California, USA
| | - Aaron Ong
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Marlyand, USA
| | - Keira A. Cohen
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Marlyand, USA
| | - Michael J. Rybak
- Anti-Infective Research Laboratory, Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan, USA
- Department of Medicine, Division of Infectious Diseases, School of Medicine, Wayne State University, Detroit, Michigan, USA
- Department of Pharmacy Services, Detroit Receiving Hospital, Detroit Medical Center, Detroit, Michigan, USA
| |
Collapse
|
16
|
Jahanbakhsh S, Howland J, Ndayishimiye Uwineza MO, Thwaites MT, Pillar CM, Serio AW, Anastasiou DM, Hufnagel DA. Evaluation of omadacycline against intracellular Mycobacterium abscessus in an infection model in human macrophages. JAC Antimicrob Resist 2023; 5:dlad104. [PMID: 37720564 PMCID: PMC10502775 DOI: 10.1093/jacamr/dlad104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 09/05/2023] [Indexed: 09/19/2023] Open
Abstract
Background Omadacycline is an aminomethylcycline antibiotic in the tetracycline class that was approved by the US FDA in 2018 for the treatment of community-acquired bacterial pneumonia and acute bacterial skin and skin structure infections. It is available in both IV and oral formulations. Omadacycline has broad-spectrum in vitro activity and clinical efficacy against infections caused by Gram-positive and Gram-negative pathogens. Omadacycline is being evaluated in a 3 month placebo-controlled Phase 2 clinical trial of oral omadacycline versus placebo in adults with non-tuberculous mycobacteria (NTM) pulmonary disease caused by Mycobacterium abscessus (NCT04922554). Objectives To determine if omadacycline has intracellular antimicrobial activity against NTM, bacteria that can cause chronic lung disease, in an ex vivo model of intracellular infection. Methods Two strains of M. abscessus were used to infect THP-1 macrophages. Intracellular M. abscessus was then challenged with omadacycline and control antibiotics at multiples of the MIC over time to evaluate intracellular killing. Results At 16 × the MIC at 72 h, omadacycline treatment of intracellular NTM yielded a log10 reduction in cfu of 1.1 (91.74% reduction in cfu) and 1.6 (97.65% reduction in cfu) consistent with killing observed with tigecycline, whereas amikacin and clarithromycin at 16 × the MIC did not show any reduction in cfu against the intracellular M. abscessus. Conclusions Omadacycline displayed intracellular activity against M. abscessus within macrophages. The activity was similar to that of tigecycline; as expected, intracellular killing was not observed with clarithromycin and amikacin.
Collapse
Affiliation(s)
- S Jahanbakhsh
- Microbiologics Antibiotic and Microbiome Research Center, Kalamazoo, MI, USA
| | - J Howland
- Microbiologics Antibiotic and Microbiome Research Center, Kalamazoo, MI, USA
| | | | - M T Thwaites
- Microbiologics Antibiotic and Microbiome Research Center, Kalamazoo, MI, USA
| | - C M Pillar
- Microbiologics Antibiotic and Microbiome Research Center, Kalamazoo, MI, USA
| | - A W Serio
- Paratek Pharmaceuticals, Inc., King of Prussia, PA, USA
| | | | - D A Hufnagel
- Microbiologics Antibiotic and Microbiome Research Center, Kalamazoo, MI, USA
| |
Collapse
|
17
|
Srivastava S, Gumbo T. Comment on "Long-term Safety and Tolerability of Omadacycline for the Treatment of Mycobacterium abscessus Infections". Open Forum Infect Dis 2023; 10:ofad423. [PMID: 37577111 PMCID: PMC10416810 DOI: 10.1093/ofid/ofad423] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 08/04/2023] [Indexed: 08/15/2023] Open
Affiliation(s)
- Shashikant Srivastava
- Department of Medicine, University of Texas School of Medicine, Tyler, Texas, USA
- Department of Cellular and Molecular Biology, University of Texas Health Science Center at Tyler, Tyler, Texas, USA
| | - Tawanda Gumbo
- Quantitative Preclinical and Clinical Sciences Department, Praedicare Inc, Dallas, Texas, USA
- Hollow Fiber System and Experimental Therapeutics Laboratories, Praedicare Inc, Dallas, Texas, USA
| |
Collapse
|
18
|
Srivastava S. Safety and tolerability of long-term use of omadacycline in the treatment of Mycobacterium abscessus infections. IDCases 2023; 33:e01843. [PMID: 37457813 PMCID: PMC10344830 DOI: 10.1016/j.idcr.2023.e01843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 07/04/2023] [Indexed: 07/18/2023] Open
Affiliation(s)
- Shashikant Srivastava
- Correspondence to: Department of Medicine, UT Tyler School of Medicine, 11937 US Highway 271, Tyler, TX 75708, USA.
| |
Collapse
|
19
|
Mingora CM, Bullington W, Faasuamalie PE, Levin A, Porter G, Stadnik R, Varley CD, Addrizzo-Harris D, Daley CL, Olivier KN, Winthrop KL, Dorman SE, Flume PA. Long-term Safety and Tolerability of Omadacycline for the Treatment of Mycobacterium abscessus Infections. Open Forum Infect Dis 2023; 10:ofad335. [PMID: 37476076 PMCID: PMC10354853 DOI: 10.1093/ofid/ofad335] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 06/29/2023] [Indexed: 07/22/2023] Open
Abstract
Background Mycobacterium abscessus is a virulent human pathogen. Treatment is complex and often poorly tolerated with suboptimal rates of eradication, highlighting the need for improved therapeutics. This study reports clinical experience with omadacycline for treatment of M abscessus infections at five large nontuberculous mycobacterial (NTM) disease clinics across the United States to better understand long-term safety and tolerability. Methods We conducted a multicenter retrospective chart review of adults with M abscessus infections. All patients treated with omadacycline as part of a multidrug therapeutic regimen through December 2021 were included. Clinical data from time of omadacycline initiation and up to 12 months of follow-up were collected. Descriptive statistics were performed. Results Analysis included 117 patients. Among patients with M abscessus isolate subspeciation, 58 of 71 (81.7%) were M abscessus spp abscessus. In isolates with reported drug susceptibility testing, 15 of 70 (21.4%) had confirmed susceptibility to macrolides. The most common site of infection was lungs. Median duration omadacycline treatment was 8 months (range, 0.25-33 months; interquartile range, 4-15 months). Omadacycline was discontinued in 60 patients (51.3%); 20 completed planned treatment course, 23 experienced intolerance or adverse event leading to drug cessation, and 17 stopped due to cost, death (unrelated to NTM infection or therapy), or another reason. In those with pulmonary disease, 44 of 95 (46%) had 1 or more negative cultures at time of final microbiological assessment, with 17 of 95 (18%) achieving culture conversion. Conclusions This study reports data supporting long-term safety and tolerability of omadacycline along with signal of effectiveness in treatment of M abscessus infections.
Collapse
Affiliation(s)
| | - Wendy Bullington
- Medical University of South Carolina, Charleston, South Carolina, USA
| | - Paige E Faasuamalie
- Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Adrah Levin
- National Jewish Health and University of Colorado School of Medicine,Denver, Colorado, USA
| | - Gabriella Porter
- New York University Grossman School of Medicine, New York, New York, USA
| | - Ryan Stadnik
- Oregon Health & Science University, Portland, Oregon, USA
| | - Cara D Varley
- Oregon Health & Science University, Portland, Oregon, USA
| | | | - Charles L Daley
- National Jewish Health and University of Colorado School of Medicine,Denver, Colorado, USA
| | - Kenneth N Olivier
- Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | | | - Susan E Dorman
- Medical University of South Carolina, Charleston, South Carolina, USA
| | - Patrick A Flume
- Medical University of South Carolina, Charleston, South Carolina, USA
| |
Collapse
|
20
|
Singh S, Wang JY, Heysell SK, McShane PJ, Wadle C, Shankar P, Huang HL, Pasipanodya J, Boorgula GD, Philley JV, Gumbo T, Srivastava S. Omadacycline pharmacokinetics/pharmacodynamics in the hollow fiber model and clinical validation of efficacy to treat pulmonary Mycobacterium abscessus disease. Int J Antimicrob Agents 2023; 62:106847. [PMID: 37187338 PMCID: PMC10330927 DOI: 10.1016/j.ijantimicag.2023.106847] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 05/03/2023] [Accepted: 05/09/2023] [Indexed: 05/17/2023]
Abstract
BACKGROUND Guideline-based therapy (GBT) for pulmonary Mycobacterium abscessus (Mab) disease achieves sustained sputum culture conversion (SSCC) rates of 30%; this is reflected by poor efficacy of GBT in the hollow fiber system model of Mab (HFS-Mab), which killed ∼1.22 log10 CFU/mL. This study was performed to determine which clinical dose of omadacycline, a tetracycline antibiotic, should be used in combination therapy to treat pulmonary Mab disease for relapse-free cure. METHODS First, omadacycline intrapulmonary concentration-time profiles of seven daily doses were mimicked in the HFS-Mab model and exposures associated with optimal efficacy were identified. Second, 10,000 subject Monte-Carlo simulations were performed to determine whether oral omadacycline 300 mg/day achieved these optimal exposures. Third, a retrospective clinical study on omadacycline vs. primarily tigecycline-based salvage therapy was conducted to assess rates of SSCC and toxicity. Fourth, a single patient was recruited to validate the findings. RESULTS Omadacycline efficacy in the HFS-Mab was 2.09 log10 CFU/mL at exposures achieved in >99% of patients on 300 mg/day omadacycline. In the retrospective study of omadacycline 300 mg/day-based combinations vs. comparators, SSCC was achieved in 8/10 vs. 1/9 (P=0.006), symptom improvement in 8/8 vs. 5/9 (P=0.033), toxicity in 0 vs. 9/9 (P<0.001), and therapy discontinuation due to toxicity in 0 vs. 3/9 (P<0.001) cases, respectively. In one prospectively recruited patient, omadacycline 300 mg/day salvage therapy achieved SSCC and symptom-resolution in 3 months. CONCLUSION Based on the preclinical and clinical data, omadacycline 300 mg/day in combination regimens could be appropriate for testing in Phase III trials in patients with Mab pulmonary disease.
Collapse
Affiliation(s)
- Sanjay Singh
- Department of Medicine, University of Texas School of Medicine, Tyler, Texas, USA
| | - Jann-Yuan Wang
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Scott K Heysell
- Division of Infectious Diseases and International Health, University of Virginia, USA
| | - Pamela J McShane
- Section of Pulmonary and Critical Care, University of Texas at Tyler, Tyler, Texas, USA
| | - Carly Wadle
- Section of Pulmonary and Critical Care, University of Texas at Tyler, Tyler, Texas, USA
| | - Prem Shankar
- Department of Medicine, University of Texas School of Medicine, Tyler, Texas, USA
| | - Hung-Ling Huang
- Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan; Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jotam Pasipanodya
- Quantitative Preclinical & Clinical Sciences Department, Praedicare Inc., Dallas, Texas
| | | | - Julie V Philley
- Section of Pulmonary and Critical Care, University of Texas at Tyler, Tyler, Texas, USA
| | - Tawanda Gumbo
- Quantitative Preclinical & Clinical Sciences Department, Praedicare Inc., Dallas, Texas; Hollow Fiber System & Experimental Therapeutics Laboratories, Praedicare Inc, Dallas, TX
| | - Shashikant Srivastava
- Department of Medicine, University of Texas School of Medicine, Tyler, Texas, USA; Department of Cellular and Molecular Biology, University of Texas Health Science Center at Tyler, Tyler, Texas, USA; Center for Biomedical Research, University of Texas Health Science Centre at Tyler, Tyler, Texas, USA.
| |
Collapse
|
21
|
Li A, He S, Li J, Zhang Z, Li B, Chu H. Omadacycline, Eravacycline, and Tigecycline Express Anti-Mycobacterium abscessus Activity In Vitro. Microbiol Spectr 2023; 11:e0071823. [PMID: 37140428 PMCID: PMC10269442 DOI: 10.1128/spectrum.00718-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 04/17/2023] [Indexed: 05/05/2023] Open
Abstract
Mycobacterium abscessus infections are increasing worldwide necessitating the development of new antibiotics and treatment regimens. The utility of third-generation tetracycline antibiotics was reestablished; their anti-M. abscessus activity needs further study. The activities of omadacycline (OMC), eravacycline (ERC), tigecycline (TGC), and sarecycline (SAC) were tested against two reference strains and 193 clinical M. abscessus isolates at different temperatures (30°C and 37°C). The minimum bactericidal concentrations (MBCs) of the four drugs were determined to distinguish between their bactericidal and bacteriostatic activities. The MICs of OMC, ERC, and TGC for the reference strains and clinical isolates were summarized and compared. OMC, ERC, and TGC exhibited a high level of bacteriostatic activity against M. abscessus. The MICs of OMC and ERC for M. abscess remained stable, while the MICs of TGC for the isolates/strains increased with increasing temperature. Notably, the MICs of OMC for M. abscessus isolates obtained in the United States are lower than for those obtained in China. IMPORTANCE The antimicrobial activities of four third-generation tetracycline-class drugs, omadacycline (OMC), eravacycline (ERC), tigecycline (TGC), and sarecycline (SAC), were determined for 193 M. abscessus isolates. The activities of the four drugs at two different temperatures (30°C and 37°C) were also tested. OMC, ERC, and TGC exhibited significant activity against M. abscessus. The anti-M. abscessus activity of TGC increased when the temperature was increased from 30°C to 37°C; the activities of OMC and ERC, on the other hand, remained the same. We found that in vitro MICs of OMC against Chinese and American isolates were distinct. Evaluations in in vivo models of M. abscessus disease or in the clinical setting will provide more accurate insight into potency of OMC against distinct isolates.
Collapse
Affiliation(s)
- Anqi Li
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
- School of Medicine, Tongji University, Shanghai, China
| | - Siyuan He
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
- School of Medicine, Tongji University, Shanghai, China
| | - Jingren Li
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
- School of Medicine, Tongji University, Shanghai, China
| | - Zhemin Zhang
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
- School of Medicine, Tongji University, Shanghai, China
| | - Bing Li
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
- School of Medicine, Tongji University, Shanghai, China
| | - Haiqing Chu
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
- School of Medicine, Tongji University, Shanghai, China
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
22
|
Fujiwara K, Aono A, Asami T, Morimoto K, Kamada K, Morishige Y, Igarashi Y, Chikamatsu K, Murase Y, Yamada H, Takaki A, Mitarai S. In Vitro Synergistic Effects of Omadacycline with Other Antimicrobial Agents against Mycobacterium abscessus. Antimicrob Agents Chemother 2023; 67:e0157922. [PMID: 37154742 PMCID: PMC10269086 DOI: 10.1128/aac.01579-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 04/17/2023] [Indexed: 05/10/2023] Open
Abstract
The clinical importance of Mycobacterium abscessus species (MABS) infections has been increasing. However, the standard treatment regimens recommended in the current guidelines often result in unfavorable outcomes. Therefore, we investigated the in vitro activity of omadacycline (OMC), a novel tetracycline, against MABS to explore its potential as a novel therapeutic option. The drug susceptibilities of 40 Mycobacterium abscessus subsp. abscessus (Mab) clinical strains obtained from the sputum of 40 patients from January 2005 to May 2014 were investigated. The MIC results for OMC, amikacin (AMK), clarithromycin (CLR), clofazimine (CLO), imipenem (IPM), rifabutin (RFB), and tedizolid (TZD) alone and their combined effects (with OMC) were examined using the checkerboard method. Additionally, we studied the differences in the effectiveness of the antibiotic combinations based on the colony morphotype of Mab. The MIC50 and MIC90 of OMC alone were 2 and 4 μg/mL, respectively. The combinations of OMC with AMK, CLR, CLO, IPM, RFB, and TZD showed synergy against 17.5%, 75.8%, 25.0%, 21.1%, 76.9%, and 34.4% of the strains, respectively. Additionally, OMC combined with CLO (47.1% versus 9.5%, P = 0.023) or TZD (60.0% versus 12.5%, P = 0.009) showed significantly higher synergy against strains with rough morphotypes than those with smooth morphotypes. In conclusion, the checkerboard analyses revealed that the synergistic effects of OMC were observed most frequently with RFB, followed by CLR, TZD, CLO, IPM, and AMK. Furthermore, OMC tended to be more effective against rough-morphotype Mab strains.
Collapse
Affiliation(s)
- Keiji Fujiwara
- Department of Mycobacterium Reference and Research, Research Institute of Tuberculosis, Japan Anti-Tuberculosis Association, Tokyo, Japan
- Respiratory Disease Center, Fukujuji Hospital, Japan Anti-Tuberculosis Association, Tokyo, Japan
- Department of Basic Mycobacteriosis, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Akio Aono
- Department of Mycobacterium Reference and Research, Research Institute of Tuberculosis, Japan Anti-Tuberculosis Association, Tokyo, Japan
| | - Takahiro Asami
- Department of Mycobacterium Reference and Research, Research Institute of Tuberculosis, Japan Anti-Tuberculosis Association, Tokyo, Japan
| | - Kozo Morimoto
- Respiratory Disease Center, Fukujuji Hospital, Japan Anti-Tuberculosis Association, Tokyo, Japan
- Department of Clinical Mycobacteriosis, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Division of Clinical Research, Fukujuji Hospital, Japan Anti-Tuberculosis Association, Tokyo, Japan
| | - Keisuke Kamada
- Department of Mycobacterium Reference and Research, Research Institute of Tuberculosis, Japan Anti-Tuberculosis Association, Tokyo, Japan
| | - Yuta Morishige
- Department of Mycobacterium Reference and Research, Research Institute of Tuberculosis, Japan Anti-Tuberculosis Association, Tokyo, Japan
| | - Yuriko Igarashi
- Department of Mycobacterium Reference and Research, Research Institute of Tuberculosis, Japan Anti-Tuberculosis Association, Tokyo, Japan
| | - Kinuyo Chikamatsu
- Department of Mycobacterium Reference and Research, Research Institute of Tuberculosis, Japan Anti-Tuberculosis Association, Tokyo, Japan
| | - Yoshiro Murase
- Department of Mycobacterium Reference and Research, Research Institute of Tuberculosis, Japan Anti-Tuberculosis Association, Tokyo, Japan
| | - Hiroyuki Yamada
- Department of Mycobacterium Reference and Research, Research Institute of Tuberculosis, Japan Anti-Tuberculosis Association, Tokyo, Japan
| | - Akiko Takaki
- Department of Mycobacterium Reference and Research, Research Institute of Tuberculosis, Japan Anti-Tuberculosis Association, Tokyo, Japan
| | - Satoshi Mitarai
- Department of Mycobacterium Reference and Research, Research Institute of Tuberculosis, Japan Anti-Tuberculosis Association, Tokyo, Japan
- Department of Basic Mycobacteriosis, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| |
Collapse
|
23
|
Rimal B, Nicklas DA, Panthi CM, Lippincott CK, Belz DC, Ignatius EH, Deck DH, Serio AW, Lamichhane G. Efficacy of Omadacycline-Containing Regimen in a Mouse Model of Pulmonary Mycobacteroides abscessus Disease. mSphere 2023; 8:e0066522. [PMID: 36912629 PMCID: PMC10117123 DOI: 10.1128/msphere.00665-22] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 02/21/2023] [Indexed: 03/14/2023] Open
Abstract
Mycobacteroides abscessus is an opportunistic pathogen in people with structural lung conditions such as bronchiectasis, chronic obstructive pulmonary disease, and cystic fibrosis. Pulmonary M. abscessus infection causes progressive symptomatic and functional decline as well as diminished lung function and is often incurable with existing antibiotics. We investigated the efficacy of a new tetracycline, omadacycline, in combination with existing antibiotics recommended to treat this indication, in a mouse model of M. abscessus lung disease. Amikacin, azithromycin, bedaquiline, biapenem, cefoxitin, clofazimine, imipenem, linezolid, and rifabutin were selected as companions to omadacycline. M. abscessus burden in the lungs of mice over a 4-week treatment duration was considered the endpoint. Omadacycline in combination with linezolid, imipenem, cefoxitin, biapenem, or rifabutin exhibited early bactericidal activity compared to any single drug. Using three M. abscessus isolates, we also determined the in vitro frequency of spontaneous resistance against omadacycline to be between 1.9 × 10-10 and 6.2 × 10-10 and the frequency of persistence against omadacycline to be between 5.3 × 10-6 and 1.3 × 10-5. Based on these findings, the combination of omadacycline and select drugs that are included in the recent treatment guidelines may exhibit improved potency to treat M. abscessus lung disease. IMPORTANCE M. abscessus disease incidence is increasing in the United States. This disease is difficult to cure with existing antibiotics. In this study, we describe the efficacy of a new tetracycline antibiotic, omadacycline, in combination with an existing antibiotic to treat this disease. A mouse model of M. abscessus lung disease was used to assess the efficacies of these experimental treatment regimens. Omadacycline in combination with select existing antibiotics exhibited bactericidal activity during the early phase of treatment.
Collapse
Affiliation(s)
- Binayak Rimal
- Division of Infectious Diseases, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Danielle A. Nicklas
- Division of Infectious Diseases, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Chandra M. Panthi
- Division of Infectious Diseases, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Christopher K. Lippincott
- Division of Infectious Diseases, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
- Center for Nontuberculous Mycobacteria and Bronchiectasis, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Daniel C. Belz
- Center for Nontuberculous Mycobacteria and Bronchiectasis, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Elisa H. Ignatius
- Division of Infectious Diseases, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
- Center for Nontuberculous Mycobacteria and Bronchiectasis, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
- Division of Clinical Pharmacology, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Daniel H. Deck
- Paratek Pharmaceuticals, Inc., King of Prussia, Pennsylvania, USA
| | - Alisa W. Serio
- Paratek Pharmaceuticals, Inc., King of Prussia, Pennsylvania, USA
| | - Gyanu Lamichhane
- Division of Infectious Diseases, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
- Center for Nontuberculous Mycobacteria and Bronchiectasis, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
24
|
In Vitro Antimicrobial Activities of Tigecycline, Eravacycline, Omadacycline, and Sarecycline against Rapidly Growing Mycobacteria. Microbiol Spectr 2023; 11:e0323822. [PMID: 36475850 PMCID: PMC9927410 DOI: 10.1128/spectrum.03238-22] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Infections caused by rapidly growing mycobacteria (RGM) have increased globally. Chemotherapy against these infections is challenging due to the minimal antimicrobial choices available. The main aim of this study was to evaluate the in vitro susceptibilities of four tetracyclines against different RGM species. The MICs of eravacycline (ERC), omadacycline (OMC), sarecycline (SAC), and tigecycline (TGC) against the reference strains of 27 RGM species and 121 RGM clinical isolates were determined by microtiter plate assay. The minimum bactericidal concentrations (MBCs) and cytotoxicities of these antibiotics were also tested. Except for SAC, the other three tetracyclines had MICs of ≤0.5 μg/mL against all 27 RGM reference strains. ERC generally presented the lowest MICs, with MIC90s against the clinical isolates of Mycobacterium abscessus subsp. abscessus, Mycobacterium abscessus subsp. massiliense, and Mycobacterium fortuitum of 0.25 μg/mL, 0.25 μg/mL, and 0.06 μg/mL, respectively. TGC and OMC also showed equivalent in vitro inhibitory activities against the isolates, while the TGC MIC90s for M. abscessus subsp. abscessus, M. abscessus subsp. massiliense, and M. fortuitum were lower than or equal to the OMC MIC90s (1, 1, and 0.25 μg/mL versus 1, 2, and 2 μg/mL). In addition, the MIC50s of three of the antibiotics for each species were always 2-fold lower than the corresponding MIC90s. MBC and cytotoxicity assays indicated that all four tetracycline antibiotics tested were bacteriostatic agents with low toxicity to the THP-1 cell line. Tetracycline antibiotics are efficacious in RGM infection treatment, with omadacycline showing the best promise for clinical application due to its potent antimicrobial activity, safety, and convenient administration route. IMPORTANCE The global rise in antibiotic-resistant nontuberculous mycobacteria has prompted the urgent need for new antimicrobials, especially oral antibiotics. Currently, adverse effects have limited the use of tetracycline-class antibiotics, particularly tigecycline (TGC), in the treatment of rapidly growing mycobacteria (RGM). However, several new tetracycline-class antibiotics might overcome the limitations of TGC. We assessed the in vitro antibiotic susceptibilities of four tetracyclines (eravacycline, omadacycline, sarecycline, and tigecycline) against reference RGM strains and clinical isolates of different RGM species. We showed that three of these antibiotics (tigecycline, eravacycline, and omadacycline) might be efficacious in M. abscessus subsp. abscessus, M. abscessus subsp. massiliense, and M. fortuitum treatment. Furthermore, omadacycline was more promising for clinical application for M. abscessus infections as an oral drug, whereas sarecycline, which had the best safety parameters, should be considered a potential antibiotic for M. abscessus infections caused by susceptible strains. Our work underscores the possible clinical applications of tetracycline-class antibiotics in the treatment of RGM infections.
Collapse
|
25
|
Ochsner UA, De Groote MA, Jarvis TC, Liu H, Youmans T, Hoang T, Ribble W, Day J, Li W, Pearce C, Walz A, Panthi CM, Rimal B, Stevens CM, Zgurskaya HI, Jackson M, Ordway D, Gonzalez-Juarrero M, Sun X, Lamichhane G, Mason C. Microbiological profile, preclinical pharmacokinetics and efficacy of CRS0393, a novel antimycobacterial agent targeting MmpL3. Tuberculosis (Edinb) 2023; 138:102288. [PMID: 36470124 PMCID: PMC9892229 DOI: 10.1016/j.tube.2022.102288] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/11/2022] [Accepted: 11/21/2022] [Indexed: 11/30/2022]
Abstract
The benzothiazole amide CRS0393 demonstrated excellent in vitro activity against nontuberculous mycobacteria (NTM), including M. abscessus isolates from cystic fibrosis (CF) patients, with minimum inhibitory concentrations (MICs) of ≤0.03-0.5 μg/mL. The essential transport protein MmpL3 was confirmed as the target via analysis of spontaneous resistant mutants and further biological profiling. In mouse pharmacokinetic studies, intratracheal instillation of a single dose of CRS0393 resulted in high concentrations of drug in epithelial lining fluid (ELF) and lung tissue, which remained above the M. abscessus MIC for at least 9 hours post-dose. This exposure resulted in a penetration ratio of 261 for ELF and 54 for lung tissue relative to plasma. CRS0393 showed good oral bioavailability, particularly when formulated in kolliphor oil, with a lung-to-plasma penetration ratio ranging from 0.5 to 4. CRS0393 demonstrated concentration-dependent reduction of intracellular M. abscessus in a THP-1 macrophage infection model. CRS0393 was well tolerated following intranasal administration (8 mg/kg) or oral dosing (25 mg/kg) once daily for 28 days in dexamethasone-treated C3HeB/FeJ mice. Efficacy against M. abscessus strain 103 was achieved via the intranasal route, while oral dosing will need further optimization. CRS0393 holds promise for development as a novel agent with broad antimycobacterial activity.
Collapse
Affiliation(s)
| | | | | | - Hang Liu
- Crestone, Inc., 6075 Longbow Dr, Boulder, CO, USA
| | | | - Teresa Hoang
- Crestone, Inc., 6075 Longbow Dr, Boulder, CO, USA
| | - Wendy Ribble
- Crestone, Inc., 6075 Longbow Dr, Boulder, CO, USA
| | - Joshua Day
- Crestone, Inc., 6075 Longbow Dr, Boulder, CO, USA
| | - Wei Li
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, 200 West Lake Street, Colorado State University, Fort Collins, CO, USA
| | - Camron Pearce
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, 200 West Lake Street, Colorado State University, Fort Collins, CO, USA
| | - Amanda Walz
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, 200 West Lake Street, Colorado State University, Fort Collins, CO, USA
| | - Chandra M Panthi
- Johns Hopkins University, 1550 Orleans Street, Baltimore, MD, USA
| | - Binayak Rimal
- Johns Hopkins University, 1550 Orleans Street, Baltimore, MD, USA
| | - Casey M Stevens
- University of Oklahoma, Department of Chemistry and Biochemistry, 101 Stephenson Parkway, Norman, OK, USA
| | - Helen I Zgurskaya
- University of Oklahoma, Department of Chemistry and Biochemistry, 101 Stephenson Parkway, Norman, OK, USA
| | - Mary Jackson
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, 200 West Lake Street, Colorado State University, Fort Collins, CO, USA
| | - Diane Ordway
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, 200 West Lake Street, Colorado State University, Fort Collins, CO, USA
| | - Mercedes Gonzalez-Juarrero
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, 200 West Lake Street, Colorado State University, Fort Collins, CO, USA
| | - Xicheng Sun
- Crestone, Inc., 6075 Longbow Dr, Boulder, CO, USA
| | - Gyanu Lamichhane
- Johns Hopkins University, 1550 Orleans Street, Baltimore, MD, USA
| | | |
Collapse
|
26
|
Nicola F, Cirillo DM, Lorè NI. Preclinical murine models to study lung infection with Mycobacterium abscessus complex. Tuberculosis (Edinb) 2023; 138:102301. [PMID: 36603391 DOI: 10.1016/j.tube.2022.102301] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 12/15/2022] [Accepted: 12/22/2022] [Indexed: 12/31/2022]
Abstract
Mycobacterium abscessus is a non-tuberculous mycobacterium (NTM) able to cause invasive pulmonary infections, named NTM pulmonary disease. The therapeutic approaches are limited, and infections are difficult to treat due to antibiotic resistance conferred by an impermeable cell wall, drug efflux pumps, or drug-modifying enzymes. The development of new therapeutics, intended as antimicrobials or drug limiting immunopathology, is urgently necessary. In this context, the preclinical murine models of M. abscessus represent a useful tool to validate and translate in vitro-proofed concepts. These in vivo models are essential for developing new targets and drugs, ameliorating our knowledge in combinatorial regimens of current existing antibiotic treatments, and repurposing existing drugs for new therapeutic options against M. abscessus infection. Thus, this review aims at providing an overview of the current state of the art of preclinical murine models to study M. abscessus lung infection and its exploitation for new therapeutic approaches. This review discusses the murine models available focusing on the different bacterial challenges (aerosol, intranasal, intratracheal, and intravenous administrations), murine genetic background, and additional bacterial related factors. Then, we discuss the successful preclinical models for M. abscessus respiratory infection exploited to study the efficacy and safety of new antimicrobials or to determine the best dosage and route of administration of existing drugs. Finally, we present the current murine models exploited to develop new therapeutic approaches to modulate the host immune response and limit immunopathological damage during M. abscessus lung disease. In conclusion, our review article provides an overview of current and available murine models to characterize acute or chronic infections and to study the outcome of new therapeutic strategies against M. abscessus lung infection.
Collapse
Affiliation(s)
- Francesca Nicola
- Emerging Bacterial Pathogens Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Daniela M Cirillo
- Emerging Bacterial Pathogens Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Nicola I Lorè
- Emerging Bacterial Pathogens Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|
27
|
Shankar P, Singh S, Boorgula GD, Gumbo T, Heysell SK, Srivastava S. Challenges and a potential solution to perform drug susceptibility testing of omadacycline against nontuberculous mycobacteria. Tuberculosis (Edinb) 2022; 137:102269. [PMID: 36209660 PMCID: PMC10626481 DOI: 10.1016/j.tube.2022.102269] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 09/27/2022] [Accepted: 09/30/2022] [Indexed: 11/06/2022]
Abstract
BACKGROUND Minimum inhibitory concentration (MIC) of slow growing mycobacteria (SGM) often do not correlate with the treatment response. Among the challenges is the identification of MIC of drugs that degrade in solution faster than the doubling time of the SGM. METHODS First, we identified the rate of omadacycline degradation in solution, and its effect on the rapidly growing methicillin resistant Staphylococcus aureus (MRSA). We then identified doubling times versus MICs for Mycobacterium abscessus, M. avium, and M. kansasii, with and without supplementation for degraded drug. RESULTS Omadacycline concentration in solution declined ∼50% over 24hr. In the MRSA experiments, omadacycline demonstrated 66.48 ± 19.38% loss in potency over 24hr, confirming the degradation rate in solution. M. abscessus had a doubling time of 8.75 ± 1.23hr and the omadacycline MIC after 24hr of incubation was 2mg/L with and without 50% daily drug supplementation, indicating that drug degradation had no effect on this rapid grower. The doubling time for M. avium was 29.52hr (95% confidence interval (CI): 23.18-33.89hr) and 31.15hr (95%CI: 19.45-38.49 hr) for M. kansasii. The M. avium MICs ±50% daily omadacycline supplementation were 1mg/L and 0.5mg/L on day 7, whereas the M. kansasii MICs ±50% daily supplementation were >128mg/L and 32mg/L on day 7. CONCLUSION Omadacycline degradation in solution leads to falsely high MICs when SGM doubling time exceed the drug degradation rates in solution. The challenge could be overcome by daily drug supplementation to account for the loss of potency, which is laborious, or perhaps stabilizing the drug from degradation.
Collapse
Affiliation(s)
- Prem Shankar
- Department of Pulmonary Immunology, University of Texas Health Science Centre at Tyler, Tyler, TX, USA
| | - Sanjay Singh
- Department of Pulmonary Immunology, University of Texas Health Science Centre at Tyler, Tyler, TX, USA
| | - Gunavanthi D Boorgula
- Department of Pulmonary Immunology, University of Texas Health Science Centre at Tyler, Tyler, TX, USA
| | - Tawanda Gumbo
- Quantitative Preclinical & Clinical Sciences Department, Praedicare Inc., Dallas, TX, USA; Hollow Fiber System & Experimental Therapeutics Laboratories, Praedicare Inc, Dallas, TX, USA
| | - Scott K Heysell
- Division of Infectious Diseases and International Health, University of Virginia, USA
| | - Shashikant Srivastava
- Department of Pulmonary Immunology, University of Texas Health Science Centre at Tyler, Tyler, TX, USA.
| |
Collapse
|
28
|
Sriram D, Wahi R, Maggioncalda EC, Panthi CM, Lamichhane G. Clofazimine as a comparator for preclinical efficacy evaluations of experimental therapeutics against pulmonary M. abscessus infection in mice. Tuberculosis (Edinb) 2022; 137:102268. [PMID: 36228452 PMCID: PMC10739713 DOI: 10.1016/j.tube.2022.102268] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 09/22/2022] [Accepted: 09/23/2022] [Indexed: 11/06/2022]
Abstract
Mycobacteroides abscessus (Mab, also known as Mycobacterium abscessus) can cause chronic pulmonary disease in the setting of structural lung conditions. Current treatment recommendations require at least one year of daily therapy with repurposed antibiotics. Yet these therapies are often ineffective and associated with significant adverse events. To address this challenge, research efforts are underway to develop new antibiotics and regimens. During the preclinical phase of treatment development, experimental agents require testing and comparison alongside positive controls that are known agents with clinical history. As there are no FDA approved treatments for this indication, here, we have considered repurposed antibiotics currently included in the recommendation for treating Mab disease as candidates for selection of an ideal standard comparator that can serve as a positive control in preclinical studies. Clofazimine meets the criteria for an ideal positive control as it can be administered via the least invasive route, requires only once-daily dosing, is well tolerated, and is widely available in high purity from independent sources. Using a mouse model of pulmonary Mab disease, we assessed for ideal dosages of clofazimine in C3HeB/FeJ and BALB/c mice in a six-week treatment window. Clofazimine, 25 mg/kg, once daily, produced desired reduction in Mab burden in the lungs of C3HeB/FeJ and BALB/c mice. Based on these findings, we conclude that clofazimine meets the criteria for a positive control comparator in mice for use in preclinical efficacy assessments of agents for treatment of Mab pulmonary disease. Although not included in the current standard-of-care for treating Mab disease, rifabutin, 20 mg/kg, also produced desired reduction in Mab lung burden in C3HeB/FeJ mice but not in BALB/c mice. IMPORTANCE: Mycobacteroides abscessus can cause life-threatening infections in patients with chronic lung conditions. New treatments are needed as cure rate using existing drugs is low. During pre-clinical phase of treatment development, it is important to compare the efficacy of the experimental drug against existing ones with known history. Here, we demonstrate that clofazimine, one of the antibiotics repurposed for treating Mab disease, can serve as a positive control comparator for efficacy assessments of experimental drugs and regimens to treat M. abscessus disease in mice.
Collapse
Affiliation(s)
- Divya Sriram
- Division of Infectious Diseases, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, 21287, USA
| | - Rishi Wahi
- Division of Infectious Diseases, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, 21287, USA
| | - Emily C Maggioncalda
- Division of Infectious Diseases, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, 21287, USA
| | - Chandra M Panthi
- Division of Infectious Diseases, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, 21287, USA
| | - Gyanu Lamichhane
- Division of Infectious Diseases, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, 21287, USA.
| |
Collapse
|
29
|
Solanki P, Lipman M, McHugh TD, Satta G. Whole genome sequencing and prediction of antimicrobial susceptibilities in non-tuberculous mycobacteria. Front Microbiol 2022; 13:1044515. [PMID: 36523832 PMCID: PMC9745125 DOI: 10.3389/fmicb.2022.1044515] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 11/14/2022] [Indexed: 12/02/2022] Open
Abstract
Non-tuberculous mycobacteria (NTM) are opportunistic pathogens commonly causing chronic, pulmonary disease which is notoriously hard to treat. Current treatment for NTM infections involves at least three active drugs (including one macrolide: clarithromycin or azithromycin) over 12 months or longer. At present there are limited phenotypic in vitro drug susceptibility testing options for NTM which are standardised globally. As seen with tuberculosis, whole genome sequencing has the potential to transform drug susceptibility testing in NTM, by utilising a genotypic approach. The Comprehensive Resistance Prediction for Tuberculosis is a database used to predict Mycobacterium tuberculosis resistance: at present there are no similar databases available to accurately predict NTM resistance. Recent studies have shown concordance between phenotypic and genotypic NTM resistance results. To benefit from the advantages of whole genome sequencing, further advances in resistance prediction need to take place, as well as there being better information on novel drug mutations and an understanding of the impact of whole genome sequencing on NTM treatment outcomes.
Collapse
Affiliation(s)
- Priya Solanki
- UCL-TB and UCL Centre for Clinical Microbiology, University College London, London, United Kingdom
| | - Marc Lipman
- UCL-TB and UCL Respiratory, University College London, London, United Kingdom
- Royal Free London NHS Foundation Trust, London, United Kingdom
| | - Timothy D. McHugh
- UCL-TB and UCL Centre for Clinical Microbiology, University College London, London, United Kingdom
| | - Giovanni Satta
- UCL-TB and UCL Centre for Clinical Microbiology, University College London, London, United Kingdom
| |
Collapse
|
30
|
Omadacycline for management of Mycobacterium abscessus infections: a review of its effectiveness, place in therapy, and considerations for use. BMC Infect Dis 2022; 22:874. [PMID: 36419143 PMCID: PMC9682665 DOI: 10.1186/s12879-022-07857-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 11/08/2022] [Indexed: 11/24/2022] Open
Abstract
The Mycobacterium abscessus complex (MABC) is a group of acid-fast, rapidly dividing non-tuberculous mycobacteria (NTM) that include a number of clinically important subspecies, including M. abscessus, M. bolletii, and M. massiliense. These organisms are prevalent in the environment and are primarily associated with human pulmonary or skin and skin structure infections (SSSI) but may cause more deep-seeded disseminated infections and bacteremia in the immunocompromised. Importantly, these NTM are resistant to most first-line anti-tuberculous agents and, due to intrinsic or acquired resistance, exhibit exceedingly low, variable, and geographically distinct susceptibilities to commonly used antibacterial agents including older tetracyclines, macrolides, aminoglycosides, cephalosporins, carbapenems, and sulfamethoxazole-trimethoprim. Omadacycline is a novel third-generation member of the tetracycline family of antibacterials that has recently been demonstrated to have potent anti-NTM effects and clinical efficacy against MABC, including M. abscessus. The purpose of this review is to present a comprehensive and up-to-date assessment on the body of literature on the role of omadacycline for M. abscessus infections. Specifically, the in vitro and in vivo microbiology, mechanisms of action, mechanisms of resistance, clinical pharmacokinetics, clinical efficacy, adverse effects, dosage and administration, and place in therapy of omadacycline in management of M. abscessus infections will be detailed.
Collapse
|
31
|
Outcomes of Short-Term Tigecycline-Containing Regimens for Mycobacterium abscessus Pulmonary Disease. Antimicrob Agents Chemother 2022; 66:e0077422. [PMID: 36165626 PMCID: PMC9578423 DOI: 10.1128/aac.00774-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Short-term intravenous tigecycline therapy during a 1-month initial phase may improve early microbiological response in patients with Mycobacterium abscessus pulmonary disease (PD). However, short-term use of tigecycline did not improve the long-term culture conversion rate of M. abscessus PD. Further studies on the efficacy of prolonged intravenous tigecycline-containing regimens are needed.
Collapse
|
32
|
Omadacycline Pharmacokinetics/Pharmacodynamics in the Hollow Fiber System Model and Potential Combination Regimen for Short Course Treatment of Mycobacterium kansasii Pulmonary Disease. Antimicrob Agents Chemother 2022; 66:e0068722. [PMID: 35976006 PMCID: PMC9487483 DOI: 10.1128/aac.00687-22] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The 12-month therapy duration for the treatment of Mycobacterium kansasii pulmonary disease calls for more efficacious drugs for better treatment outcomes and to shorten the therapy duration. We performed (i) omadacycline MIC with M. kansasii ATCC 12478 strain and 21 clinical isolates, (ii) dose-response study in the hollow fiber system model of M. kansasii (HFS-Mkn) with six human equivalent omadacycline daily doses to determine the optimal drug exposure for the maximal kill, and (iii) a second HFS-Mkn study to determine the efficacy of omadacycline (300 mg/day) plus moxifloxacin (600 mg/day) plus tedizolid (200 mg/day) combination regimen with standard regimen as comparator. GraphPad Prism was used for data analysis and graphing. MIC of the reference strain was 4 mg/L but ranged from 8 to 32 mg/L among the 21 clinical isolates. In the HFS-Mkn, the exposure required for 50% of the maximal effect (EC50) was an omadacycline area under the concentration-time curve to MIC (AUC0-24/MIC) ratio of 1.95. The optimal exposure was an AUC0-24/MIC of 3.05, which could be achieved with 300 mg/day clinical dose. The omadacycline-moxifloxacin-tedizolid combination sterilized the HFS-Mkn in 14 days with a linear-regression based kill rate of -0.309 ± 0.044 log10 CFU/mL/day compared to the kill rate of -0.084 ± 0.036log10 CFU/mL/day with the standard regimen or 3.7-times faster. Omadacycline has efficacy against M. kansasii and could be used at 300 mg/day in combination with moxifloxacin and tedizolid for the treatment of M. kansasii pulmonary diseases with the potential to shorten the currently recommended 12-month therapy duration.
Collapse
|
33
|
Duah M, Beshay M. Omadacycline in first-line combination therapy for pulmonary Mycobacterium abscessus infection: A case series. Int J Infect Dis 2022; 122:953-956. [DOI: 10.1016/j.ijid.2022.06.061] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 06/22/2022] [Accepted: 06/30/2022] [Indexed: 11/26/2022] Open
|
34
|
T405, a New Penem, Exhibits In Vivo Efficacy against M. abscessus and Synergy with β-Lactams Imipenem and Cefditoren. Antimicrob Agents Chemother 2022; 66:e0053622. [PMID: 35638855 PMCID: PMC9211421 DOI: 10.1128/aac.00536-22] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Mycobacteroides abscessus (Mab) is an emerging environmental microbe that causes chronic lung disease in patients with compromised lung function such as cystic fibrosis and bronchiectasis. It is intrinsically resistant to most antibiotics, therefore there are only few antibiotics that can be repurposed to treat Mab disease. Although current recommendations require daily intake of multiple antibiotics for more than a year, cure rate is low and often associated with significant adverse events. Here, we describe in vivo efficacy of T405, a recently discovered β-lactam antibiotic of the penem subclass, in a mouse model of pulmonary Mab infection. Imipenem, one of the standard-of-care drugs to treat Mab disease, and also a β-lactam antibiotic from a chemical class similar to T405, was included as a comparator. Probenecid was included with both T405 and imipenem to reduce the rate of their renal clearance. T405 exhibited bactericidal activity against Mab from the onset of treatment and reduced Mab lung burden at a rate similar to that exhibited by imipenem. The MIC of T405 against Mab was unaltered after 4 weeks of exposure to T405 in the lungs of mice. Using an in vitro assay, we also demonstrate that T405 in combination with imipenem, cefditoren or avibactam exhibits synergism against Mab. Additionally, we describe a scheme for synthesis and purification of T405 on an industrial scale. These attributes make T405 a promising candidate for further preclinical assessment to treat Mab disease.
Collapse
|
35
|
Galanis C, Maggioncalda EC, Kumar P, Lamichhane G. Glby, Encoded by MAB_3167c, Is Required for In Vivo Growth of Mycobacteroides abscessus and Exhibits Mild β-Lactamase Activity. J Bacteriol 2022; 204:e0004622. [PMID: 35380462 PMCID: PMC9112878 DOI: 10.1128/jb.00046-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 03/10/2022] [Indexed: 11/24/2022] Open
Abstract
Mycobacteroides abscessus (Mab; also known as Mycobacterium abscessus) is an emerging opportunistic pathogen. Patients with structural lung conditions such as bronchiectasis, cystic fibrosis, and chronic obstructive pulmonary disease are at high risk of developing pulmonary Mab disease. This disease is often chronic as the current treatment regimens are sub-efficacious. Here, we characterize the phenotype of a Mab strain lacking the MAB_3167c locus, which encodes a protein hereafter referred to as Glby. We demonstrate that the loss of Glby impairs normal planktonic growth in liquid broth, results in longer average cell length, and a melding of surfaces between cells. Glby also exhibits a mild β-lactamase activity. We also present evidence that amino acid substitutions that potentially alter Glby function are not favored. Lastly, we demonstrate that, in a mouse model of pulmonary Mab infection, the mutant lacking Glby was unable to proliferate, gradually cleared, and was undetectable after 3 weeks. These data suggest that an agent that inhibits Glby in vivo may be an efficacious treatment against Mab disease. IMPORTANCE Mycobacteroides abscessus can cause chronic pulmonary infections requiring administration of multiple antibiotics, still resulting in a low cure rate. The incidence of M. abscessus disease is increasing in the United States and the developed regions of the world. We show for the first time that a protein, Glby, affects growth of this bacterium. Using a mouse model of lung M. abscessus disease, we demonstrate that Glby is required for this bacterium to cause disease.
Collapse
Affiliation(s)
- Christos Galanis
- Center for Tuberculosis Research, Department of Medicine, School of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Emily C. Maggioncalda
- Center for Tuberculosis Research, Department of Medicine, School of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Pankaj Kumar
- Center for Tuberculosis Research, Department of Medicine, School of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Gyanu Lamichhane
- Center for Tuberculosis Research, Department of Medicine, School of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
36
|
Parmanik A, Das S, Kar B, Bose A, Dwivedi GR, Pandey MM. Current Treatment Strategies Against Multidrug-Resistant Bacteria: A Review. Curr Microbiol 2022; 79:388. [PMID: 36329256 PMCID: PMC9633024 DOI: 10.1007/s00284-022-03061-7] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 09/26/2022] [Indexed: 11/06/2022]
Abstract
There are several bacteria called superbugs that are resistant to multiple antibiotics which can be life threatening specially for critically ill and hospitalized patients. This article provides up-to-date treatment strategies employed against some major superbugs, like methicillin-resistant Staphylococcus aureus, carbapenem-resistant Enterobacteriaceae, vancomycin-resistant Enterococcus, multidrug-resistant Pseudomonas aeruginosa, and multidrug-resistant Escherichia coli. The pathogen-directed therapeutics decrease the toxicity of bacteria by altering their virulence factors by specific processes. On the other hand, the host-directed therapeutics limits these superbugs by modulating immune cells, enhancing host cell functions, and modifying disease pathology. Several new antibiotics against the global priority superbugs are coming to the market or are in the clinical development phase. Medicinal plants possessing potent secondary metabolites can play a key role in the treatment against these superbugs. Nanotechnology has also emerged as a promising option for combatting them. There is urgent need to continuously figure out the best possible treatment strategy against these superbugs as resistance can also be developed against the new and upcoming antibiotics in future. Rational use of antibiotics and maintenance of proper hygiene must be practiced among patients.
Collapse
Affiliation(s)
- Ankita Parmanik
- grid.412612.20000 0004 1760 9349School of Pharmaceutical Sciences, Siksha O Anusandhan (Deemed to be University), Bhubaneswar, Odisha 751003 India
| | - Soumyajit Das
- grid.412612.20000 0004 1760 9349School of Pharmaceutical Sciences, Siksha O Anusandhan (Deemed to be University), Bhubaneswar, Odisha 751003 India
| | - Biswakanth Kar
- grid.412612.20000 0004 1760 9349School of Pharmaceutical Sciences, Siksha O Anusandhan (Deemed to be University), Bhubaneswar, Odisha 751003 India
| | - Anindya Bose
- grid.412612.20000 0004 1760 9349School of Pharmaceutical Sciences, Siksha O Anusandhan (Deemed to be University), Bhubaneswar, Odisha 751003 India
| | - Gaurav Raj Dwivedi
- grid.464904.b0000 0004 0506 3705ICMR-Regional Medical Research Centre, Gorakhpur, Uttar Pradesh 273013 India
| | - Murali Monohar Pandey
- grid.418391.60000 0001 1015 3164Birla Institute of Technology and Science (BITS), Pilani, Rajasthan 333031 India
| |
Collapse
|
37
|
Bich Hanh BT, Quang NT, Park Y, Heo BE, Jeon S, Park JW, Jang J. Omadacycline Potentiates Clarithromycin Activity Against Mycobacterium abscessus. Front Pharmacol 2021; 12:790767. [PMID: 34955859 PMCID: PMC8693020 DOI: 10.3389/fphar.2021.790767] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 11/15/2021] [Indexed: 11/29/2022] Open
Abstract
Mycobacterium abscessus is a difficult respiratory pathogen to treat, when compared to other nontuberculus mycobacteria (NTM), due to its drug resistance. In this study, we aimed to find a new clarithromycin partner that potentiated strong, positive, synergy against M. abscessus among current anti-M. abscessus drugs, including omadacycline, amikacin, rifabutin, bedaquiline, and cefoxitine. First, we determined the minimum inhibitory concentrations required of all the drugs tested for M. abscessus subsp. abscessus CIP104536T treatment using a resazurin microplate assay. Next, the best synergistic partner for clarithromycin against M. abscessus was determined using an in vitro checkerboard combination assay. Among the drug combinations evaluated, omadacycline showed the best synergistic effect with clarithromycin, with a fractional inhibitory concentration index of 0.4. This positive effect was also observed against M. abscessus clinical isolates and anti-M. abscessus drug resistant strains. Lastly, this combination was further validated using a M. abscessus infected zebrafish model. In this model, the clarithromycin-omadacyline regimen was found to inhibit the dissemination of M. abscessus, and it significantly extended the lifespan of the M. abscessus infected zebrafish. In summation, the synergy between two anti-M. abscessus compounds, clarithromycin and omadacycline, provides an attractive foundation for a new M. abscessus treatment regimen.
Collapse
Affiliation(s)
- Bui Thi Bich Hanh
- Division of Applied Life Science (BK21 Four Program), Research Institute of Life Science, Gyeongsang National University, Jinju, South Korea
| | - Nguyen Thanh Quang
- Division of Life Science, Department of Bio & Medical Big Data (BK21 Four Program), Research Institute of Life Science, Gyeongsang National University, Jinju, South Korea
| | - Yujin Park
- Division of Life Science, Department of Bio & Medical Big Data (BK21 Four Program), Research Institute of Life Science, Gyeongsang National University, Jinju, South Korea
| | - Bo Eun Heo
- Division of Life Science, Department of Bio & Medical Big Data (BK21 Four Program), Research Institute of Life Science, Gyeongsang National University, Jinju, South Korea
| | - Seunghyeon Jeon
- Division of Life Science, Gyeongsang National University, Jinju, South Korea
| | - June-Woo Park
- Department of Environmental Toxicology and Chemistry, Korea Institute of Toxicology, Korea & Human and Environmental Toxicology Program, Korea University of Science and Technology (UST), Daejeon, South Korea
| | - Jichan Jang
- Division of Life Science, Department of Bio & Medical Big Data (BK21 Four Program), Research Institute of Life Science, Gyeongsang National University, Jinju, South Korea.,Division of Life Science, Gyeongsang National University, Jinju, South Korea
| |
Collapse
|