1
|
Kim HW, Lee JW, Yoon HS, Park HW, Lee YI, Lee SK, Whang J, Kim JS. Restriction of mitochondrial oxidation of glutamine or fatty acids enhances intracellular growth of Mycobacterium abscessus in macrophages. Virulence 2025; 16:2454323. [PMID: 39828906 PMCID: PMC11749347 DOI: 10.1080/21505594.2025.2454323] [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: 08/18/2024] [Revised: 11/28/2024] [Accepted: 01/12/2025] [Indexed: 01/22/2025] Open
Abstract
Mycobacterium abscessus (Mab), a nontuberculous mycobacterium, is increasing in prevalence worldwide and causes treatment-refractory pulmonary diseases. However, how Mab rewires macrophage energy metabolism to facilitate its survival is poorly understood. We compared the metabolic profiles of murine bone marrow-derived macrophages (BMDMs) infected with smooth (S)- and rough (R)-type Mab using extracellular flux technology. Mab infection shifted BMDMs towards a more energetic phenotype, marked by increased oxidative phosphorylation (OXPHOS) and glycolysis, with a significantly greater enhancement in OXPHOS. This metabolic adaptation was characterized by enhanced ATP production rates, particularly in cells infected with S-type Mab, highlighting OXPHOS as a key energy source. Notably, Mab infection also modulated mitochondrial substrate preferences, increasing fatty acid oxidation capabilities while revealing significant changes in glutamine dependency and flexibility. R-type Mab infections exhibited a marked decrease in glutamine reliance but enhanced metabolic flexibility and capacity. Furthermore, targeting metabolic pathways related to glutamine and fatty acid oxidation exacerbated Mab growth within macrophages, suggesting these pathways play a protective role against infection. These insights advance our understanding of Mab's impact on host cell metabolism and propose a novel avenue for therapeutic intervention. By manipulating host mitochondrial metabolism, we identify a potential host-directed therapeutic strategy against Mab, offering a promising alternative to conventional treatments beleaguered by drug resistance. This study underscores the importance of exploring metabolic interventions to combat Mab infection, paving the way for innovative approaches in the fight against this formidable pathogen.
Collapse
Affiliation(s)
- Ho Won Kim
- Myunggok Medical Research Institute, College of Medicine, Konyang University, Daejeon, South Korea
| | - Ji Won Lee
- Myunggok Medical Research Institute, College of Medicine, Konyang University, Daejeon, South Korea
| | - Hoe Sun Yoon
- Myunggok Medical Research Institute, College of Medicine, Konyang University, Daejeon, South Korea
| | - Hwan-Woo Park
- Department of Cell Biology, Konyang University Hospital and College of Medicine, Daejeon, South Korea
| | | | - Sung Ki Lee
- Department of Obstetrics and Gynecology, Konyang University Hospital, Daejeon, South Korea
| | - Jake Whang
- Korea Mycobacterium Resource Center (KMRC), Department of Research and Development, The Korean Institute of Tuberculosis, Osong, South Korea
| | - Jong-Seok Kim
- Myunggok Medical Research Institute, College of Medicine, Konyang University, Daejeon, South Korea
- Department of Cell Biology, Konyang University Hospital and College of Medicine, Daejeon, South Korea
| |
Collapse
|
2
|
Murgia C, Yazdi Z, Soto E. Susceptibility of Non-Tuberculous Mycobacteria Biofilm to Common Disinfectants in Aquaculture Systems. JOURNAL OF FISH DISEASES 2025; 48:e14091. [PMID: 39920900 DOI: 10.1111/jfd.14091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 01/09/2025] [Accepted: 01/20/2025] [Indexed: 02/10/2025]
Abstract
Mycobacteriosis is a common and persistent bacterial disease affecting cultured, wild and pet fish. The disease can be caused by various Mycobacterium spp. Currently, depopulation and disinfection are the main recommended measures for containing disease outbreaks, as no vaccines are commercially available, and only a few reports of successful antimicrobial therapies have been made. While disinfectant susceptibility studies have been conducted on planktonic forms of some non-tuberculous mycobacteria (NTM) affecting fish, biofilm-related research remains limited. In this study, biofilm formation of Mycobacterium chelonae, Mycobacterium salmoniphilum, Mycobacterium arcueilense and Mycobacterium marinum isolates recovered from diseased fish were initially evaluated using the minimal biofilm eradication concentration (MBEC) assay system. All Mycobacterium spp. were able to form biofilms within a 2-week period when incubated at 25°C, but M. chelonae, M. salmoniphilum and M. arcueilense produced a faster and greater biofilm than M. marinum. To investigate the susceptibility of mycobacterial biofilms to common disinfectants, mature biofilms were divided into six treatment groups based on disinfectant type and exposure time: povidone-iodine (50 mg L-1 free iodine) and bleach (200 mg L-1 free chlorine) were applied for 30 min each; hydrogen peroxide (3% H2O2) and Virkon Aquatic (10 g L-1) were applied for 15 min each; and 70% ethanol was tested at both 15 and 30 min. Results demonstrated variable susceptibility among species. M. marinum was susceptible to all disinfectants, while M. arcueilense was susceptible to bleach, povidone-iodine and 70% ethanol (30-min exposure). None of the tested disinfectants at recommended doses effectively eradicated M. chelonae or M. salmoniphilum biofilms. These findings underscore species-specific differences in biofilm formation and resistance, emphasising the critical need for robust disinfection protocols in aquaculture settings to prevent mycobacteriosis transmission. Further research is essential to understand the mechanisms underlying disinfectant resistance among NTMs, optimise disinfection strategies and ensure the health and biosecurity of aquaculture facilities.
Collapse
Affiliation(s)
- Claudio Murgia
- Department of Veterinary Medicine, University of Sassari, Sassari, Italy
| | - Zeinab Yazdi
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California, Davis, California, USA
| | - Esteban Soto
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California, Davis, California, USA
| |
Collapse
|
3
|
Cocorullo M, Stamilla A, Recchia D, Marturano MC, Maci L, Stelitano G. Mycobacterium abscessus Virulence Factors: An Overview of Un-Explored Therapeutic Options. Int J Mol Sci 2025; 26:3247. [PMID: 40244091 PMCID: PMC11990050 DOI: 10.3390/ijms26073247] [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: 03/06/2025] [Revised: 03/28/2025] [Accepted: 03/29/2025] [Indexed: 04/18/2025] Open
Abstract
Mycobacterium abscessus (Mab) is an opportunistic pathogen gaining increased importance due to its capacity to colonize the respiratory tract of patients with chronic lung diseases such as individuals with Cystic Fibrosis. The actual therapeutic regimen to treat Mab infections is based on repurposed drugs from therapies against Mycobacterium tuberculosis and avium. In addition to the need for new specific drugs against this bacterium, a possible strategy for shortening the therapeutic time and improving the success rate could be targeting Mab virulence factors. These drugs could become an important integration to the actual therapeutic regimen, helping the immune system to fight the infection. Moreover, this strategy applies a low selective pressure on the bacteria, since these elements are not essential for Mab survival but crucial for establishing the infection. This review aims to provide an overview of the Mab's virulence factors that are poorly studied and mostly unknown, suggesting some interesting alternatives to classical drug development.
Collapse
Affiliation(s)
| | | | | | | | | | - Giovanni Stelitano
- Department of Biology and Biotechnology “Lazzaro Spallanzani”, University of Pavia, 27100 Pavia, Italy; (M.C.); (A.S.); (D.R.); (M.C.M.); (L.M.)
| |
Collapse
|
4
|
Isozaki Y, Makikawa T, Kimura K, Nishihara D, Fujino M, Tanaka Y, Hayashi C, Ishizaki Y, Igarashi M, Yokoyama T, Toshima K, Takahashi D. Creation of a macrolide antibiotic against non-tuberculous Mycobacterium using late-stage boron-mediated aglycon delivery. SCIENCE ADVANCES 2025; 11:eadt2352. [PMID: 40043128 PMCID: PMC11881915 DOI: 10.1126/sciadv.adt2352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 01/30/2025] [Indexed: 05/13/2025]
Abstract
Non-tuberculous mycobacteria (NTM) is gaining clinical recognition as a recently emerging pulmonary pathogen. Mycobacterium avium complex (MAC), the most common NTM, is the cause of pulmonary MAC disease. Currently, the macrolide azithromycin (AZM) is the standard first-line antibiotic for treatment of the disease. However, the rise of drug-resistant MAC necessitates the development of alternative therapeutics. Here, we present a late-stage boron-mediated aglycon delivery strategy for selective modification of AZM, generating a library of potential anti-MAC drugs designated KU01 to KU13. Screening of KU01 to KU13 revealed that KU13 exhibited enhanced antimicrobial activity against wild-type and macrolide-resistant MAC compared to AZM. Cryo-electron microscopy analysis indicated that the inserted tercyclic moiety of KU13 formed a robust anchor on the bacterial ribosome, creating a binding pocket with base flipping of U2847, potentially bypassing the standard mechanism of macrolide resistance. These results position KU13 as a promising lead for therapeutics against macrolide-resistant MAC.
Collapse
Affiliation(s)
- Yuka Isozaki
- Department of Applied Chemistry, Faculty of Science and Technology, Keio University, 3-14-1 Hiyoshi, Kohoku-ku, Yokohama 223-8522, Japan
| | - Takumi Makikawa
- Department of Applied Chemistry, Faculty of Science and Technology, Keio University, 3-14-1 Hiyoshi, Kohoku-ku, Yokohama 223-8522, Japan
| | - Kosuke Kimura
- Department of Applied Chemistry, Faculty of Science and Technology, Keio University, 3-14-1 Hiyoshi, Kohoku-ku, Yokohama 223-8522, Japan
| | - Daiki Nishihara
- Graduate School of Life Sciences, Tohoku University, 2-1-1 Katahira, Aoba-ku, Sendai, Miyagi 980-8577, Japan
| | - Maho Fujino
- Graduate School of Life Sciences, Tohoku University, 2-1-1 Katahira, Aoba-ku, Sendai, Miyagi 980-8577, Japan
| | - Yoshikazu Tanaka
- Graduate School of Life Sciences, Tohoku University, 2-1-1 Katahira, Aoba-ku, Sendai, Miyagi 980-8577, Japan
- The Advanced Center for Innovations in Next-Generation Medicine (INGEM), Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8573, Japan
| | - Chigusa Hayashi
- Institute of Microbial Chemistry (BIKAKEN), 3-14-23 Kamiosaki, Shinagawa-ku, Tokyo 141-0021, Japan
| | - Yoshimasa Ishizaki
- Institute of Microbial Chemistry (BIKAKEN), 3-14-23 Kamiosaki, Shinagawa-ku, Tokyo 141-0021, Japan
| | - Masayuki Igarashi
- Institute of Microbial Chemistry (BIKAKEN), 3-14-23 Kamiosaki, Shinagawa-ku, Tokyo 141-0021, Japan
| | - Takeshi Yokoyama
- Graduate School of Life Sciences, Tohoku University, 2-1-1 Katahira, Aoba-ku, Sendai, Miyagi 980-8577, Japan
- The Advanced Center for Innovations in Next-Generation Medicine (INGEM), Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8573, Japan
| | - Kazunobu Toshima
- Department of Applied Chemistry, Faculty of Science and Technology, Keio University, 3-14-1 Hiyoshi, Kohoku-ku, Yokohama 223-8522, Japan
| | - Daisuke Takahashi
- Department of Applied Chemistry, Faculty of Science and Technology, Keio University, 3-14-1 Hiyoshi, Kohoku-ku, Yokohama 223-8522, Japan
| |
Collapse
|
5
|
Ning H, He G, Mei Y, Yu J, Shi J, Cui X, Qiu C, Jiang X. Clinical correlation study of non-tuberculous mycobacterial isolates from bronchoalveolar lavage fluid. AMB Express 2025; 15:40. [PMID: 40045108 PMCID: PMC11883075 DOI: 10.1186/s13568-025-01842-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Accepted: 02/06/2025] [Indexed: 03/09/2025] Open
Abstract
Non-tuberculous mycobacterial (NTM) infections have emerged as a significant public health concern, posing a threat to human health. This study aims to identify various NTM strains from bronchoalveolar lavage fluid, assess their drug resistance profiles, and investigate the risk factors associated with NTM disease. Gene chip technology was used to identify NTM strains. The broth microdilution method assessed the drug sensitivity of isolated NTM pathogenic bacteria, determining their minimum inhibitory concentrations (MICs). Logistic regression analysis identified potential risk factors for NTM disease. Results showed the slow-growing NTM strains isolated from bronchoalveolar lavage fluid to be predominantly Mycobacterium avium and Mycobacterium intracellulare, accounting for 32.05% and 29.49% of the isolates, respectively. The rapidly growing NTM strains were mainly Mycobacterium chelonae and Mycobacterium abscessus, each constituting 25.64% of the isolates. Mycobacterium avium was found to be sensitive to clarithromycin, while linezolid demonstrated high antibacterial efficacy against Mycobacterium intracellulare. In drug susceptibility testing of Mycobacterium chelonae and Mycobacterium abscessus, amikacin exhibited the highest sensitivity, followed by clarithromycin. For patients with NTM-positive cultures, the risk factors for NTM lung disease included age (45-60 years, > 60 years), a smoking history exceeding 10 years, chronic obstructive pulmonary disease (COPD), bronchiectasis, immunocompromised status, and the presence of thin-walled pulmonary cavities. Collectively, this study elucidates the distribution of NTM strains, their drug susceptibility profiles, and key risk factors for NTM lung disease, highlighting the need for proactive screening, early intervention, and targeted preventive strategies to improve diagnosis and optimize treatment outcomes.
Collapse
Affiliation(s)
- Hongye Ning
- Department of Infection, Wenzhou Central Hospital, Dingli Clinical College of Wenzhou Medical University, No.252, Baili East Road, Lucheng District, Wenzhou, Zhejiang, China
| | - Guiqing He
- Department of Infection, Wenzhou Central Hospital, Dingli Clinical College of Wenzhou Medical University, No.252, Baili East Road, Lucheng District, Wenzhou, Zhejiang, China
| | - Yanhong Mei
- Department of Infection, Wenzhou Central Hospital, Dingli Clinical College of Wenzhou Medical University, No.252, Baili East Road, Lucheng District, Wenzhou, Zhejiang, China
| | - Jiandan Yu
- Department of Infection, Wenzhou Central Hospital, Dingli Clinical College of Wenzhou Medical University, No.252, Baili East Road, Lucheng District, Wenzhou, Zhejiang, China
| | - Jichan Shi
- Department of Infection, Wenzhou Central Hospital, Dingli Clinical College of Wenzhou Medical University, No.252, Baili East Road, Lucheng District, Wenzhou, Zhejiang, China
| | - Xiaoya Cui
- Department of Infection, Wenzhou Central Hospital, Dingli Clinical College of Wenzhou Medical University, No.252, Baili East Road, Lucheng District, Wenzhou, Zhejiang, China
| | - Chaochao Qiu
- Department of Infection, Wenzhou Central Hospital, Dingli Clinical College of Wenzhou Medical University, No.252, Baili East Road, Lucheng District, Wenzhou, Zhejiang, China
| | - Xiangao Jiang
- Department of Infection, Wenzhou Central Hospital, Dingli Clinical College of Wenzhou Medical University, No.252, Baili East Road, Lucheng District, Wenzhou, Zhejiang, China.
| |
Collapse
|
6
|
Diricks M, Maurer FP, Dreyer V, Barilar I, Utpatel C, Merker M, Wetzstein N, Niemann S. Genomic insights into the plasmidome of non-tuberculous mycobacteria. Genome Med 2025; 17:19. [PMID: 40038805 PMCID: PMC11877719 DOI: 10.1186/s13073-025-01443-7] [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: 10/22/2024] [Accepted: 02/21/2025] [Indexed: 03/06/2025] Open
Abstract
BACKGROUND Non-tuberculous mycobacteria (NTM) are a diverse group of environmental bacteria that are increasingly associated with human infections and difficult to treat. Plasmids, which might carry resistance and virulence factors, remain largely unexplored in NTM. METHODS We used publicly available complete genome sequence data of 328 NTM isolates belonging to 125 species to study gene content, genomic diversity, and clusters of 196 annotated NTM plasmids. Furthermore, we analyzed 3755 draft genome assemblies from over 200 NTM species and 5415 short-read sequence datasets from six clinically relevant NTM species or complexes including M. abscessus, M. avium complex, M. ulcerans complex and M. kansasii complex, for the presence of these plasmids. RESULTS Between one and five plasmids were present in approximately one-third of the complete NTM genomes. The annotated plasmids varied widely in length (most between 10 and 400 kbp) and gene content, with many genes having an unknown function. Predicted gene functions primarily involved plasmid replication, segregation, maintenance, and mobility. Only a few plasmids contained predicted genes that are known to confer resistance to antibiotics commonly used to treat NTM infections. Out of 196 annotated plasmid sequences, 116 could be grouped into 31 clusters of closely related sequences, and about one-third were found across multiple NTM species. Among clinically relevant NTM, the presence of NTM plasmids showed significant variation between species, within (sub)species, and even among strains within (sub)lineages, such as dominant circulating clones of Mycobacterium abscessus. CONCLUSIONS Our analysis demonstrates that plasmids are a diverse and heterogeneously distributed feature in NTM bacteria. The frequent occurrence of closely related putative plasmid sequences across different NTM species suggests they may play a significant role in NTM evolution through horizontal gene transfer at least in some groups of NTM. However, further in vitro investigations and access to more complete genomes are necessary to validate our findings, elucidate gene functions, identify novel plasmids, and comprehensively assess the role of plasmids in NTM.
Collapse
Affiliation(s)
- Margo Diricks
- Molecular and Experimental Mycobacteriology, Research Center Borstel, Borstel, Germany.
- German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Borstel, Germany.
- National and WHO Supranational Reference Laboratory for Mycobacteria, Research Center Borstel, Leibniz Lung Center, Borstel, Germany.
| | - Florian P Maurer
- National and WHO Supranational Reference Laboratory for Mycobacteria, Research Center Borstel, Leibniz Lung Center, Borstel, Germany
- Institute of Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Viola Dreyer
- Molecular and Experimental Mycobacteriology, Research Center Borstel, Borstel, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Borstel, Germany
- National and WHO Supranational Reference Laboratory for Mycobacteria, Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | - Ivan Barilar
- Molecular and Experimental Mycobacteriology, Research Center Borstel, Borstel, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Borstel, Germany
- National and WHO Supranational Reference Laboratory for Mycobacteria, Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | - Christian Utpatel
- Molecular and Experimental Mycobacteriology, Research Center Borstel, Borstel, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Borstel, Germany
- National and WHO Supranational Reference Laboratory for Mycobacteria, Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | - Matthias Merker
- German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Borstel, Germany
- Evolution of the Resistome, Research Center Borstel, Borstel, Germany
| | - Nils Wetzstein
- Molecular and Experimental Mycobacteriology, Research Center Borstel, Borstel, Germany
- Department of Internal Medicine, Infectious Diseases, Goethe University Frankfurt, University Hospital, Frankfurt Am Main, Germany
- Mycobacterial Infection Research Unit (MIRU), Goethe University Frankfurt, Frankfurt Am Main, Germany
| | - Stefan Niemann
- Molecular and Experimental Mycobacteriology, Research Center Borstel, Borstel, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Borstel, Germany
- National and WHO Supranational Reference Laboratory for Mycobacteria, Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| |
Collapse
|
7
|
Clemente-Hernández B, Álvarez-Salafranca M, Gracia-Cazaña T, Muelas-Rives I, Ferrando-Lamana L, Viñuelas-Bayón J, Gilaberte Y. Combination of Methyl Aminolevulinate-Based Photodynamic Therapy and Antibiotics in the Treatment of Mycobacterium gordonae Scalp Infection: A Case Report. PHOTODERMATOLOGY, PHOTOIMMUNOLOGY & PHOTOMEDICINE 2025; 41:e70012. [PMID: 40098269 DOI: 10.1111/phpp.70012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 03/01/2025] [Accepted: 03/05/2025] [Indexed: 03/19/2025]
Affiliation(s)
| | | | - Tamara Gracia-Cazaña
- Department of Dermatology, Hospital Universitario Miguel Servet, Zaragoza, Spain
| | - Itziar Muelas-Rives
- Department of Dermatology, Hospital Universitario Miguel Servet, Zaragoza, Spain
| | | | - Jesús Viñuelas-Bayón
- Department of Microbiology, Hospital Universitario Miguel Servet, Zaragoza, Spain
| | - Yolanda Gilaberte
- Department of Dermatology, Hospital Universitario Miguel Servet, Zaragoza, Spain
| |
Collapse
|
8
|
Zheng J, Liu Q, Wang H, Huo F, Jia J, Huang H, Chen S. Efficacy of carbonyl cyanide-3-chlorophenylhydrazone in combination with antibiotics against Mycobacterium abscessus. Microbiol Spectr 2025; 13:e0177724. [PMID: 39727410 PMCID: PMC11792458 DOI: 10.1128/spectrum.01777-24] [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: 07/16/2024] [Accepted: 11/26/2024] [Indexed: 12/28/2024] Open
Abstract
Given the intrinsic resistance of Mycobacterium abscessus to a wide range of conventional antibiotics, it is urgent to explore new therapeutic approaches to manage this infection effectively. Carbonyl cyanide 3-chlorophenylhydrazone (CCCP), a proton pump inhibitor, has shown good bacteriostatic activity against M. abscessus. This study aimed to determine its synergistic antimicrobial effects when combined with commonly used antibiotics. The M. abscessus reference strain and 39 clinical isolates were collected. The minimum inhibitory concentration and fractional inhibitory concentration index were determined for the combined treatment using CCCP with various antibiotics including clarithromycin, amikacin, linezolid, bedaquiline, and clofazimine. A time-killing assay was used to measure the effect of the combined drug regimens quantitatively. The simultaneous use of CCCP with traditional antibiotics shows a synergistic effect across a wide range, significantly boosting the ability to inhibit the growth of M. abscessus throughout its growth phases. When CCCP is used in combination with clarithromycin, amikacin, and linezolid, it produces a synergistic effect on both the standard strain and most clinical isolates. When CCCP is paired with bedaquiline and clofazimine, it exhibits additive effects. Moreover, high levels of CCCP in combination with other antibiotics were found to rapidly eradicate the bacteria. The use of CCCP as a potential treatment for M. abscessus infections shows promising results, especially when combined with other antibiotics to achieve a potent bactericidal effect.IMPORTANCEMycobacterium abscessus poses a significant public health threat due to its intrinsic resistance to a broad spectrum of conventional antibiotics. This resistance necessitates urgently exploring novel therapeutic strategies to effectively combat infections caused by this pathogen. Our previous research has identified carbonyl cyanide-3-chlorophenylhydrazone (CCCP) as a potent direct antimicrobial agent against M. abscessus and as an enhancer of clarithromycin activity. Our results demonstrate that the concurrent administration of CCCP with traditional antibiotics exhibits a synergistic effect across a wide range, which could be crucial for overcoming the challenges posed by M. abscessus infections. Furthermore, the use of high concentrations of CCCP in combination with other antibiotics was found to rapidly eliminate M. abscessus, suggesting a potential therapeutic advantage. These insights not only advance our understanding of antimicrobial synergy but also hold promise for the development of more effective treatment regimens against drug-resistant M. abscessus infections.
Collapse
Affiliation(s)
- Jifang Zheng
- National Clinical Laboratory on Tuberculosis, Beijing Key Laboratory for Drug-Resistant Tuberculosis Research, Beijing Chest Hospital, Beijing Tuberculosis and Thoracic Tumor Institute, Capital Medical University, Beijing, China
| | - Qiang Liu
- National Clinical Laboratory on Tuberculosis, Beijing Key Laboratory for Drug-Resistant Tuberculosis Research, Beijing Chest Hospital, Beijing Tuberculosis and Thoracic Tumor Institute, Capital Medical University, Beijing, China
| | - Haoran Wang
- National Clinical Laboratory on Tuberculosis, Beijing Key Laboratory for Drug-Resistant Tuberculosis Research, Beijing Chest Hospital, Beijing Tuberculosis and Thoracic Tumor Institute, Capital Medical University, Beijing, China
| | - Fengmin Huo
- National Clinical Laboratory on Tuberculosis, Beijing Key Laboratory for Drug-Resistant Tuberculosis Research, Beijing Chest Hospital, Beijing Tuberculosis and Thoracic Tumor Institute, Capital Medical University, Beijing, China
| | - Junnan Jia
- National Clinical Laboratory on Tuberculosis, Beijing Key Laboratory for Drug-Resistant Tuberculosis Research, Beijing Chest Hospital, Beijing Tuberculosis and Thoracic Tumor Institute, Capital Medical University, Beijing, China
| | - Hairong Huang
- National Clinical Laboratory on Tuberculosis, Beijing Key Laboratory for Drug-Resistant Tuberculosis Research, Beijing Chest Hospital, Beijing Tuberculosis and Thoracic Tumor Institute, Capital Medical University, Beijing, China
| | - Suting Chen
- National Clinical Laboratory on Tuberculosis, Beijing Key Laboratory for Drug-Resistant Tuberculosis Research, Beijing Chest Hospital, Beijing Tuberculosis and Thoracic Tumor Institute, Capital Medical University, Beijing, China
| |
Collapse
|
9
|
Dhasmana DJ, Whitaker P, van der Laan R, Frost F. A practical guide to the diagnosis and management of suspected Non-tuberculous Mycobacterial Pulmonary Disease (NTM-PD) in the United Kingdom. NPJ Prim Care Respir Med 2024; 34:45. [PMID: 39709516 PMCID: PMC11663218 DOI: 10.1038/s41533-024-00403-9] [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: 03/12/2024] [Accepted: 11/27/2024] [Indexed: 12/23/2024] Open
Abstract
Non-tuberculous Mycobacterial Pulmonary Disease (NTM-PD) is a chronic disease characterised by progressive inflammatory lung damage due to infection by non-tuberculous mycobacteria (NTM). Global prevalence of NTM-PD is generally low but is rising, likely due to a combination of increased surveillance, increasing multimorbidity and improved diagnostic techniques. Most disease is caused by Mycobacterium avium complex species. NTM-PD can be challenging to both diagnose and manage but given the risk of untreated disease and the challenges around drug treatments, it is vital that all healthcare professionals involved in primary care consider NTM-PD at the earliest opportunity. In particular, NTM-PD should be considered where there are respiratory symptoms in the setting of pre-existing chronic lung disease such as chronic obstructive pulmonary disease (COPD) and bronchiectasis. Early suspicion should lead to appropriate primary screening measures. This article discusses the relevance of NTM-PD today, risk factors for developing disease, pathways from clinical presentation to referral to specialist care, and discusses management and drug treatments. A flow diagram of a screening process is presented as a guideline for best practice from a United Kingdom perspective.
Collapse
Grants
- Medical writing support, under the direction of the authors, was provided by Ashfield MedComms GmbH (Mannheim, Germany), an Inizio company, and funded by Insmed Incorporated.
- Medical writing support, under the direction of the authors, was provided by Ashfield MedComms GmbH (Mannheim, Germany), an Inizio company, and funded by Insmed Incorporated. Payment on honoraria for lectures, presentations, speaker bureaus, manuscript writing or educational events from Insmed Incorporated.
- Medical writing support, under the direction of the authors, was provided by Ashfield MedComms GmbH (Mannheim, Germany), an Inizio company, and funded by Insmed Incorporated. Employee of Insmed Incorporated.
- Medical writing support, under the direction of the authors, was provided by Ashfield MedComms GmbH (Mannheim, Germany), an Inizio company, and funded by Insmed Incorporated. Support for attending meetings and/or travel from Chiesi Ltd.
Collapse
Affiliation(s)
- D J Dhasmana
- Victoria Hospital, Kirkcaldy, NHS Fife, Kirkcaldy, UK.
- Infection and Global Health, School of Medicine, University of St Andrews, St Andrews, UK.
| | - P Whitaker
- Bradford Teaching Hospitals, Bradford, UK
| | | | - F Frost
- Liverpool Heart & Chest Hospital NHS Foundation Trust, Liverpool, UK
- Institute of Infection, Veterinary and Ecology Sciences, University of Liverpool, Liverpool, UK
| |
Collapse
|
10
|
Dartois V, Dick T. Toward better cures for Mycobacterium abscessus lung disease. Clin Microbiol Rev 2024; 37:e0008023. [PMID: 39360834 PMCID: PMC11629636 DOI: 10.1128/cmr.00080-23] [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] [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
|
11
|
Andrade Meirelles M, Almeida VM, Sullivan JR, de Toledo I, Dos Reis CV, Cunha MR, Zigweid R, Shim A, Sankaran B, Woodward EL, Seibold S, Liu L, Mian MR, Battaile KP, Riley J, Duncan C, Simeons FRC, Ferguson L, Joji H, Read KD, Lovell S, Staker BL, Behr MA, Pilli RA, Couñago RM. Rational Exploration of 2,4-Diaminopyrimidines as DHFR Inhibitors Active against Mycobacterium abscessus and Mycobacterium avium, Two Emerging Human Pathogens. J Med Chem 2024; 67:19143-19164. [PMID: 39468773 DOI: 10.1021/acs.jmedchem.4c01594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/30/2024]
Abstract
Nontuberculous mycobacteria (NTM) are emerging human pathogens linked to severe pulmonary diseases. Current treatments involve the prolonged use of multiple drugs and are often ineffective. Bacterial dihydrofolate reductase (DHFR) is a key enzyme targeted by antibiotics in Gram-negative bacterial infections. However, existing DHFR inhibitors designed for Gram-negative bacteria often fail against mycobacterial DHFRs. Here, we detail the rational design of NTM DHFR inhibitors based on P218, a malarial DHFR inhibitor. We identified compound 8, a 2,4-diaminopyrimidine exhibiting improved pharmacological properties and activity against purified DHFR, and whole cell cultures of two predominant NTM species: Mycobacterium avium and Mycobacterium abscessus. This study underscores the potential of compound 8 as a promising candidate for the in vivo validation of DHFR as an effective treatment against NTM infections.
Collapse
Affiliation(s)
- Matheus Andrade Meirelles
- Department of Organic Chemistry, Institute of Chemistry, University of Campinas, UNICAMP, 13083-970-Campinas, SP, Brazil
| | - Vitor M Almeida
- Center of Medicinal Chemistry (CQMED), Center for Molecular Biology and Genetic Engineering (CBMEG), University of Campinas, UNICAMP, 13083-886-Campinas, SP, Brazil
| | - Jaryd R Sullivan
- Department of Microbiology & Immunology, McGill University, Montréal H3A 2B4, Canada
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montréal H4A 3J1, Canada
- McGill International TB Centre, Montréal H4A 3S5, Canada
| | - Ian de Toledo
- Department of Organic Chemistry, Institute of Chemistry, University of Campinas, UNICAMP, 13083-970-Campinas, SP, Brazil
| | - Caio Vinicius Dos Reis
- Center of Medicinal Chemistry (CQMED), Center for Molecular Biology and Genetic Engineering (CBMEG), University of Campinas, UNICAMP, 13083-886-Campinas, SP, Brazil
| | - Micael Rodrigues Cunha
- Center of Medicinal Chemistry (CQMED), Center for Molecular Biology and Genetic Engineering (CBMEG), University of Campinas, UNICAMP, 13083-886-Campinas, SP, Brazil
| | - Rachel Zigweid
- Center for Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington 98109, United States
- Seattle Structural Genomics Center for Infectious Disease (SSGCID), Seattle, Washington 98109, United States
| | - Abraham Shim
- Center for Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington 98109, United States
- Seattle Structural Genomics Center for Infectious Disease (SSGCID), Seattle, Washington 98109, United States
| | - Banumathi Sankaran
- Molecular Biophysics and Integrated Bioimaging, Berkeley Center for Structural Biology, Lawrence Berkeley National Laboratory, Berkeley, California 94720, United States
| | - Elijah L Woodward
- Seattle Structural Genomics Center for Infectious Disease (SSGCID), Seattle, Washington 98109, United States
- Protein Structure and X-ray Crystallography Laboratory, Del Shankel Structural Biology Center, University of Kansas, Lawrence, Kansas 66047, United States
| | - Steve Seibold
- Seattle Structural Genomics Center for Infectious Disease (SSGCID), Seattle, Washington 98109, United States
- Protein Structure and X-ray Crystallography Laboratory, Del Shankel Structural Biology Center, University of Kansas, Lawrence, Kansas 66047, United States
| | - Lijun Liu
- Seattle Structural Genomics Center for Infectious Disease (SSGCID), Seattle, Washington 98109, United States
- Protein Structure and X-ray Crystallography Laboratory, Del Shankel Structural Biology Center, University of Kansas, Lawrence, Kansas 66047, United States
| | - Mohammad Rasel Mian
- Seattle Structural Genomics Center for Infectious Disease (SSGCID), Seattle, Washington 98109, United States
- Protein Structure and X-ray Crystallography Laboratory, Del Shankel Structural Biology Center, University of Kansas, Lawrence, Kansas 66047, United States
| | - Kevin P Battaile
- New York Structural Biology Center, Upton, New York 11973, United States
| | - Jennifer Riley
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry, University of Dundee, Dundee DD1 5EH, U.K
| | - Christina Duncan
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry, University of Dundee, Dundee DD1 5EH, U.K
| | - Frederick R C Simeons
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry, University of Dundee, Dundee DD1 5EH, U.K
| | - Liam Ferguson
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry, University of Dundee, Dundee DD1 5EH, U.K
| | - Halimatu Joji
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry, University of Dundee, Dundee DD1 5EH, U.K
| | - Kevin D Read
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry, University of Dundee, Dundee DD1 5EH, U.K
| | - Scott Lovell
- Seattle Structural Genomics Center for Infectious Disease (SSGCID), Seattle, Washington 98109, United States
- Protein Structure and X-ray Crystallography Laboratory, Del Shankel Structural Biology Center, University of Kansas, Lawrence, Kansas 66047, United States
| | - Bart L Staker
- Center for Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington 98109, United States
- Seattle Structural Genomics Center for Infectious Disease (SSGCID), Seattle, Washington 98109, United States
| | - Marcel A Behr
- Department of Microbiology & Immunology, McGill University, Montréal H3A 2B4, Canada
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montréal H4A 3J1, Canada
- McGill International TB Centre, Montréal H4A 3S5, Canada
- Department of Medicine, McGill University Health Centre, Montréal H4A 3J1, Canada
| | - Ronaldo A Pilli
- Department of Organic Chemistry, Institute of Chemistry, University of Campinas, UNICAMP, 13083-970-Campinas, SP, Brazil
| | - Rafael M Couñago
- Center of Medicinal Chemistry (CQMED), Center for Molecular Biology and Genetic Engineering (CBMEG), University of Campinas, UNICAMP, 13083-886-Campinas, SP, Brazil
- Structural Genomics Consortium and Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
12
|
Tan SYL, Tan JBX, Ng DHL. Stable antimicrobial resistance patterns of Mycobacterium abscessus complex antibiograms in Singapore from 2013 to 2022: a retrospective review. Singapore Med J 2024:00077293-990000000-00166. [PMID: 39498525 DOI: 10.4103/singaporemedj.smj-2024-075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 07/09/2024] [Indexed: 01/11/2025]
Affiliation(s)
- Shireen Yan Ling Tan
- Department of Microbiology, Singapore General Hospital, Singapore
- Pathology Academic Clinical Programme, SingHealth Duke-NUS, Duke-NUS Medical School, Singapore
| | - Jamie Bee Xian Tan
- Department of Microbiology, Singapore General Hospital, Singapore
- Pathology Academic Clinical Programme, SingHealth Duke-NUS, Duke-NUS Medical School, Singapore
| | - Dorothy Hui Lin Ng
- Department of Infectious Diseases, Singapore General Hospital, Singapore
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| |
Collapse
|
13
|
Longo BM, Trunfio M, Calcagno A. Dual β-lactams for the treatment of Mycobacterium abscessus: a review of the evidence and a call to act against an antibiotic nightmare. J Antimicrob Chemother 2024; 79:2731-2741. [PMID: 39150384 PMCID: PMC11932079 DOI: 10.1093/jac/dkae288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/17/2024] Open
Abstract
Mycobacterium abscessus complex is a group of rapidly growing non-tuberculous mycobacteria (NTM), increasingly emerging as opportunistic pathogens. Current treatment options for these microorganisms are limited and associated with a high rate of treatment failure, toxicity and recurrence. In search of new therapeutic strategies, interest has grown in dual β-lactam (DBL) therapy, as research recently discovered that M. abscessus cell wall synthesis is mainly regulated by two types of enzymes (d,d-transpeptidases and l,d-transpeptidases) differently susceptible to inhibition by distinct β-lactams. In vitro studies testing several DBL combinations have shown synergy in extracellular broth cultures as well as in the intracellular setting: cefoxitin/imipenem, ceftaroline/imipenem, ceftazidime/ceftaroline and ceftazidime/imipenem. The addition of specific β-lactamase inhibitors (BLIs) targeting M. abscessus β-lactamase did not significantly enhance the activity of DBL combinations. However, in vivo data are lacking. We reviewed the literature on DBL/DBL-BLI-based therapies for M. abscessus infections to raise greater attention on this promising yet overlooked treatment option and to guide future preclinical and clinical studies.
Collapse
Affiliation(s)
- Bianca Maria Longo
- Department of Medical Sciences, Unit of Infectious Diseases, Amedeo di Savoia Hospital, University of Turin, 10149 Turin, Italy
| | - Mattia Trunfio
- Department of Medical Sciences, Unit of Infectious Diseases, Amedeo di Savoia Hospital, University of Turin, 10149 Turin, Italy
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California San Diego, La Jolla, CA 92037, USA
| | - Andrea Calcagno
- Department of Medical Sciences, Unit of Infectious Diseases, Amedeo di Savoia Hospital, University of Turin, 10149 Turin, Italy
| |
Collapse
|
14
|
Chu Y, Wang X, Dou M, Wang J, Wang B, Wang H, Lv S, Lu S, Li T. Clinical Characteristics, Species Distribution, and Drug Resistance of Non-Tuberculous Mycobacteria Lung Disease in Qingdao, China. Infect Drug Resist 2024; 17:4807-4814. [PMID: 39502134 PMCID: PMC11537159 DOI: 10.2147/idr.s475015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 10/18/2024] [Indexed: 11/08/2024] Open
Abstract
Objective To analyze the clinical characteristics, species distribution and drug resistance of patients with non-tuberculous mycobacteria (NTM) lung disease in Qingdao, China. Methods Clinical data of patients with NTM lung disease and pulmonary tuberculosis (TB) treated at Qingdao Chest Hospital from July 2021 to July 2023 were retrospectively analyzed. Results The prevalence of NTM lung disease was 8.03%, with a high rate of drug resistance during the study period. Patients with NTM lung disease had higher rates of older age, bronchiectasis, malignancy, HIV infection and bronchial dilatation shadow and lower rates of hollow shadow compared to patients with pulmonary TB. Conclusion Comprehensive understanding of NTM lung disease, improved laboratory testing techniques and appropriate treatment regimens are essential for the management of NTM lung disease.
Collapse
Affiliation(s)
- Yan Chu
- Qingdao Chest Hospital, Qingdao, Shandong, People’s Republic of China
| | - Xiaomin Wang
- National Clinical Research Center for Infectious Diseases, Shenzhen Third People’s Hospital, Shenzhen, Guangdong, People’s Republic of China
| | - Min Dou
- Qingdao Chest Hospital, Qingdao, Shandong, People’s Republic of China
| | - Jin Wang
- National Clinical Research Center for Infectious Diseases, Shenzhen Third People’s Hospital, Shenzhen, Guangdong, People’s Republic of China
| | - Baoqian Wang
- Qingdao Chest Hospital, Qingdao, Shandong, People’s Republic of China
| | - Hairong Wang
- Qingdao Chest Hospital, Qingdao, Shandong, People’s Republic of China
| | - Shasha Lv
- Qingdao Chest Hospital, Qingdao, Shandong, People’s Republic of China
| | - Shuihua Lu
- National Clinical Research Center for Infectious Diseases, Shenzhen Third People’s Hospital, Shenzhen, Guangdong, People’s Republic of China
| | - Tongxia Li
- Qingdao Chest Hospital, Qingdao, Shandong, People’s Republic of China
| |
Collapse
|
15
|
Le Run E, Tettelin H, Holland SM, Zelazny AM. Evolution toward extremely high imipenem resistance in Mycobacterium abscessus outbreak strains. Antimicrob Agents Chemother 2024; 68:e0067324. [PMID: 39254295 PMCID: PMC11459939 DOI: 10.1128/aac.00673-24] [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: 05/06/2024] [Accepted: 08/15/2024] [Indexed: 09/11/2024] Open
Abstract
Treatment of Mycobacterium abscessus pulmonary disease requires multiple antibiotics including intravenous β-lactams (e.g., imipenem). M. abscessus produces a β-lactamase (BlaMab) that inactivates β-lactam drugs but less efficiently carbapenems. Due to intrinsic and acquired resistance in M. abscessus and poor clinical outcomes, it is critical to understand the development of antibiotic resistance both within the host and in the setting of outbreaks. We compared serial longitudinally collected M. abscessus subsp. massiliense isolates from the index case of a cystic fibrosis center outbreak and four outbreak-related strains. We found strikingly high imipenem resistance in the later patient isolates, including the outbreak strain (MIC > 512 µg/mL). The phenomenon was recapitulated upon exposure of intracellular bacteria to imipenem. Addition of the β-lactamase inhibitor avibactam abrogated the resistant phenotype. Imipenem resistance was caused by an increase in β-lactamase activity and increased blaMab mRNA level. Concurrent increase in transcription of the preceding ppiA gene indicated upregulation of the entire operon in the resistant strains. Deletion of the porin mspA coincided with the first increase in MIC (from 8 to 32 µg/mL). A frameshift mutation in msp2 responsible for the rough colony morphology and a SNP in ATP-dependent helicase hrpA cooccurred with the second increase in MIC (from 32 to 256 µg/mL). Increased BlaMab expression and enzymatic activity may have been due to altered regulation of the ppiA-blaMab operon by the mutated HrpA alone or in combination with other genes described above. This work supports using carbapenem/β-lactamase inhibitor combinations for treating M. abscessus, particularly imipenem-resistant strains.
Collapse
Affiliation(s)
- Eva Le Run
- Laboratory of Clinical Immunology and Microbiology (LCIM), Immunopathogenesis Section, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, Maryland, USA
| | - Hervé Tettelin
- Department of Microbiology and Immunology, Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Steven M. Holland
- Laboratory of Clinical Immunology and Microbiology (LCIM), Immunopathogenesis Section, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, Maryland, USA
| | - Adrian M. Zelazny
- Department of Laboratory Medicine (DLM), Microbiology Service, Clinical Center, NIH, Bethesda, Maryland, USA
| |
Collapse
|
16
|
Sreekumar A, Kumar A, Biswas R, Biswas L. Emerging and alternative strategies for the treatment of nontuberculous mycobacterial infections. Expert Rev Anti Infect Ther 2024; 22:835-853. [PMID: 39161153 DOI: 10.1080/14787210.2024.2395003] [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: 07/13/2024] [Accepted: 08/18/2024] [Indexed: 08/21/2024]
Abstract
INTRODUCTION Nontuberculous mycobacteria (NTM) infections have emerged as a significant clinical challenge due to their intrinsic multidrug resistance and the limited efficacy of existing treatments. These infections are becoming increasingly prevalent, with a need for new and effective therapeutic strategies. AREAS COVERED This review addresses several key aspects of NTM infections: i) pathogenesis and epidemiology; ii) the limitations and challenges of current treatment options; iii) emerging and alternative therapeutic strategies; iv) advanced drug delivery systems such as nanoparticles and efflux pump inhibitors; v) innovative antibacterial alternatives like antimicrobial peptides, bacteriophage therapy, and phytochemicals; and vi) other potential treatment modalities such as inhaled nitric oxide, small molecules, surgical debridement, phototherapy, and immunomodulatory therapy. EXPERT OPINION Personalized medicine, advanced drug delivery systems, and alternative therapies hold promise for the future of NTM treatment. Early and accurate identification of NTM species, enabled by improved diagnostic methods, is critical for tailoring treatment regimens. Emerging therapies show promise against drug-resistant NTM strains, but overcoming barriers like clinical trials, regulatory hurdles, and high production costs is crucial. Continued research and innovation are essential to improve treatment efficacy and patient outcomes.
Collapse
Affiliation(s)
| | - Anil Kumar
- Department of Microbiology, Amrita Institute of Medical Sciences & Research Centre, Amrita Vishwa Vidyapeetham, Kochi, India
| | - Raja Biswas
- Amrita Center for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, India
| | - Lalitha Biswas
- Amrita Center for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, India
| |
Collapse
|
17
|
Datta D, Jamwal S, Jyoti N, Patnaik S, Kumar D. Actionable mechanisms of drug tolerance and resistance in Mycobacterium tuberculosis. FEBS J 2024; 291:4433-4452. [PMID: 38676952 DOI: 10.1111/febs.17142] [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: 09/11/2023] [Revised: 02/23/2024] [Accepted: 04/10/2024] [Indexed: 04/29/2024]
Abstract
The emergence of antimicrobial resistance (AMR) across bacterial pathogens presents a serious threat to global health. This threat is further exacerbated in tuberculosis (TB), mainly due to a protracted treatment regimen involving a combination of drugs. A diversity of factors contributes to the emergence of drug resistance in TB, which is caused by the pathogen Mycobacterium tuberculosis (Mtb). While the traditional genetic mutation-driven drug resistance mechanisms operate in Mtb, there are also several additional unique features of drug resistance in this pathogen. Research in the past decade has enriched our understanding of such unconventional factors as efflux pumps, bacterial heterogeneity, metabolic states, and host microenvironment. Given that the discovery of new antibiotics is outpaced by the emergence of drug resistance patterns displayed by the pathogen, newer strategies for combating drug resistance are desperately needed. In the context of TB, such approaches include targeting the efflux capability of the pathogen, modulating the host environment to prevent bacterial drug tolerance, and activating the host anti-mycobacterial pathways. In this review, we discuss the traditional mechanisms of drug resistance in Mtb, newer understandings and the shaping of a set of unconventional approaches to target both the emergence and treatment of drug resistance in TB.
Collapse
Affiliation(s)
- Dipanwita Datta
- Cellular Immunology Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT), Bhubaneswar, India
| | - Shaina Jamwal
- Cellular Immunology Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Nishant Jyoti
- Cellular Immunology Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Srinivas Patnaik
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT), Bhubaneswar, India
| | - Dhiraj Kumar
- Cellular Immunology Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| |
Collapse
|
18
|
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
|
19
|
Reil I, Duvnjak S, Špičić S, Kompes G, Bagarić A, Đuras M, Gudan Kurilj A, Lukač M, Jelić M, Zdelar-Tuk M. Isolation of Multidrug-Resistant Mycobacterium Avium Subsp. Avium from a Wild Eurasian Otter (Lutra Lutra). Antibiotics (Basel) 2024; 13:591. [PMID: 39061273 PMCID: PMC11273438 DOI: 10.3390/antibiotics13070591] [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: 05/20/2024] [Revised: 06/21/2024] [Accepted: 06/24/2024] [Indexed: 07/28/2024] Open
Abstract
Mycobacterium avium subsp. avium is pathogenic mainly to birds, although cases of mycobacteriosis caused by these bacteria have also been reported in other animals and humans. Not much is known about the effects of this pathogen on otters. The aim of this study was to report for the first time the isolation of M. avium subsp. avium in wild otter and to describe its multidrug resistance profile. A female otter injured in a car accident was found dead and subjected to postmortem examination. Apart from the trauma changes, no other macroscopic pathological changes were detected. Bacteriologic examination revealed the presence of acid-fast bacilli in the lymph nodes, which were confirmed by molecular methods as M. avium subsp. avium. Antimicrobial susceptibility testing revealed susceptibility to clarithromycin and amikacin, but resistance to linezolid, moxifloxacin, streptomycin, isoniazid, trimethoprim/sulfamethoxazole, ciprofloxacin, doxycycline, and ethionamide. This is unusual for wild species, which generally should not come into contact with antimicrobials, and may suggest that multidrug-resistant MAC strains are circulating between wild and domestic animals. These results emphasise the need for additional epidemiological studies on non-tuberculous mycobacteria in wildlife and their implications for one health.
Collapse
Affiliation(s)
- Irena Reil
- Croatian Veterinary Institute, Savska Cesta 143, 10000 Zagreb, Croatia; (I.R.); (S.Š.); (G.K.); (A.B.); (M.Z.-T.)
| | - Sanja Duvnjak
- Croatian Veterinary Institute, Savska Cesta 143, 10000 Zagreb, Croatia; (I.R.); (S.Š.); (G.K.); (A.B.); (M.Z.-T.)
| | - Silvio Špičić
- Croatian Veterinary Institute, Savska Cesta 143, 10000 Zagreb, Croatia; (I.R.); (S.Š.); (G.K.); (A.B.); (M.Z.-T.)
| | - Gordan Kompes
- Croatian Veterinary Institute, Savska Cesta 143, 10000 Zagreb, Croatia; (I.R.); (S.Š.); (G.K.); (A.B.); (M.Z.-T.)
| | - Antonela Bagarić
- Croatian Veterinary Institute, Savska Cesta 143, 10000 Zagreb, Croatia; (I.R.); (S.Š.); (G.K.); (A.B.); (M.Z.-T.)
| | - Martina Đuras
- Faculty of Veterinary Medicine, University of Zagreb, Heinzelova 55, 10000 Zagreb, Croatia; (M.Đ.); (A.G.K.); (M.L.)
| | - Andrea Gudan Kurilj
- Faculty of Veterinary Medicine, University of Zagreb, Heinzelova 55, 10000 Zagreb, Croatia; (M.Đ.); (A.G.K.); (M.L.)
| | - Maja Lukač
- Faculty of Veterinary Medicine, University of Zagreb, Heinzelova 55, 10000 Zagreb, Croatia; (M.Đ.); (A.G.K.); (M.L.)
| | - Mišel Jelić
- Varaždin City Museum, Department of Natural Sciences, Šetalište Josipa Jurja Strossmayera 1, 42000 Varaždin, Croatia;
| | - Maja Zdelar-Tuk
- Croatian Veterinary Institute, Savska Cesta 143, 10000 Zagreb, Croatia; (I.R.); (S.Š.); (G.K.); (A.B.); (M.Z.-T.)
| |
Collapse
|
20
|
Vadankula GR, Nilkanth VV, Rizvi A, Yandrapally S, Agarwal A, Chirra H, Biswas R, Arifuddin M, Nema V, Mallika A, Mande SC, Banerjee S. Confronting Tuberculosis: A Synthetic Quinoline-Isonicotinic Acid Hydrazide Hybrid Compound as a Potent Lead Molecule Against Mycobacterium tuberculosis. ACS Infect Dis 2024; 10:2288-2302. [PMID: 38717380 DOI: 10.1021/acsinfecdis.4c00277] [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] [Indexed: 06/15/2024]
Abstract
The current tuberculosis (TB) treatment is challenged by a complex first-line treatment for drug-sensitive (DS) TB. Additionally, the prevalence of multidrug (MDR)- and extensively drug (XDR)-resistant TB necessitates the search for new drug prototypes. We synthesized and screened 30 hybrid compounds containing aminopyridine and 2-chloro-3-formyl quinoline to arrive at a compound with potent antimycobacterial activity, UH-NIP-16. Subsequently, antimycobacterial activity against DS and MDR Mycobacterium tuberculosis (M.tb) strains were performed. It demonstrated an MIC50 value of 1.86 ± 0.21 μM for laboratory pathogenic M.tb strain H37Rv and 3.045 ± 0.813 μM for a clinical M.tb strain CDC1551. UH-NIP-16 also decreased the MIC50 values of streptomycin, isoniazid, ethambutol, and bedaquiline to about 45, 55, 68, and 76%, respectively, when used in combination, potentiating their activities. The molecule was active against a clinical MDR M.tb strain. Cytotoxicity on PBMCs from healthy donors and on human cell lines was found to be negligible. Further, blind docking of UH-NIP-16 using Auto Dock Vina and MGL tools onto diverse M.tb proteins showed high binding affinities with multiple M.tb proteins, the top five targets being metabolically critical proteins CelA1, DevS, MmaA4, lysine acetyltransferase, and immunity factor for tuberculosis necrotizing toxin. These bindings were confirmed by fluorescence spectroscopy using a representative protein, MmaA4. Envisaging that a pathogen will have a lower probability of developing resistance to a hybrid molecule with multiple targets, we propose that UH-NIP-16 can be further developed as a lead molecule with the bacteriostatic potential against M.tb, both alone and in combination with first-line drugs.
Collapse
Affiliation(s)
- Govinda Raju Vadankula
- Laboratory of Molecular Pathogenesis, Department of Biochemistry, School of Life Sciences, University of Hyderabad (UoH), Hyderabad 500046, India
| | - Vipul V Nilkanth
- Laboratory of Molecular Pathogenesis, Department of Biochemistry, School of Life Sciences, University of Hyderabad (UoH), Hyderabad 500046, India
- Bioinformatics Centre, Savitribai Phule Pune University, Pune 411007, India
| | - Arshad Rizvi
- Laboratory of Molecular Pathogenesis, Department of Biochemistry, School of Life Sciences, University of Hyderabad (UoH), Hyderabad 500046, India
| | - Sriram Yandrapally
- Laboratory of Molecular Pathogenesis, Department of Biochemistry, School of Life Sciences, University of Hyderabad (UoH), Hyderabad 500046, India
| | - Anushka Agarwal
- Laboratory of Molecular Pathogenesis, Department of Biochemistry, School of Life Sciences, University of Hyderabad (UoH), Hyderabad 500046, India
| | - Hepshibha Chirra
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Rashmita Biswas
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Mohammed Arifuddin
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Vijay Nema
- Molecular Biology Division, ICMR-National Institute for Translational Virology and AIDS Research, Pune 411026, India
| | - Alvala Mallika
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Shekhar C Mande
- National Centre for Cell Science, Pune 411007, India
- Bioinformatics Centre, Savitribai Phule Pune University, Pune 411007, India
| | - Sharmistha Banerjee
- Laboratory of Molecular Pathogenesis, Department of Biochemistry, School of Life Sciences, University of Hyderabad (UoH), Hyderabad 500046, India
| |
Collapse
|
21
|
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
|
22
|
Kobayashi T, Ford B, Diekema DJ, Sekar P. Disseminated Mycobacterium chimaera infection successfully treated with a clofazimine-containing regimen and long-term follow-up after discontinuing treatment. IDCases 2024; 36:e01991. [PMID: 38846029 PMCID: PMC11154623 DOI: 10.1016/j.idcr.2024.e01991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Accepted: 05/21/2024] [Indexed: 06/09/2024] Open
Abstract
Mycobacterium chimaera is a slow-growing member of the Mycobacterium avium complex. It can contaminate tap water and has been detected in water tanks of heater-cooler devices used during open-heart surgery. Herein we report a case of a 67-year-old- male with disseminated M. chimaera infection who presented with fevers, significant weight loss and was found to have bacteremia with prosthetic valve endocarditis, chorioretinitis, bone marrow and splenic granulomas two years after an open-heart surgery. He developed multiple drug adverse events over the course of treatment but was successfully treated using a clofazimine containing regimen along with aortic valve replacement. He has remained symptom-free with no signs of recurrence three years after completion of antimicrobials. Clofazimine is an effective alternative that can be used as part of a multi-drug regimen in M. chimaera infection when there is resistance to first-line drugs or when adverse drug reactions occur.
Collapse
Affiliation(s)
- Takaaki Kobayashi
- Department of Internal Medicine, Division of Infectious Diseases, University of Iowa Hospitals and Clinics, Iowa City, IA, United States
| | - Bradley Ford
- Department of Pathology, University of Iowa Hospitals and Clinics, Iowa City, IA, United States
| | - Daniel J. Diekema
- Department of Internal Medicine, Division of Infectious Diseases, University of Iowa Hospitals and Clinics, Iowa City, IA, United States
- Department of Medicine, Division of Infectious Diseases, Maine Medical Center, Portland, ME, United States
| | - Poorani Sekar
- Department of Internal Medicine, Division of Infectious Diseases, University of Iowa Hospitals and Clinics, Iowa City, IA, United States
| |
Collapse
|
23
|
Dartois V, Dick T. Therapeutic developments for tuberculosis and nontuberculous mycobacterial lung disease. Nat Rev Drug Discov 2024; 23:381-403. [PMID: 38418662 PMCID: PMC11078618 DOI: 10.1038/s41573-024-00897-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/24/2024] [Indexed: 03/02/2024]
Abstract
Tuberculosis (TB) drug discovery and development has undergone nothing short of a revolution over the past 20 years. Successful public-private partnerships and sustained funding have delivered a much-improved understanding of mycobacterial disease biology and pharmacology and a healthy pipeline that can tolerate inevitable attrition. Preclinical and clinical development has evolved from decade-old concepts to adaptive designs that permit rapid evaluation of regimens that might greatly shorten treatment duration over the next decade. But the past 20 years also saw the rise of a fatal and difficult-to-cure lung disease caused by nontuberculous mycobacteria (NTM), for which the drug development pipeline is nearly empty. Here, we discuss the similarities and differences between TB and NTM lung diseases, compare the preclinical and clinical advances, and identify major knowledge gaps and areas of cross-fertilization. We argue that applying paradigms and networks that have proved successful for TB, from basic research to clinical trials, will help to populate the pipeline and accelerate curative regimen development for NTM disease.
Collapse
Affiliation(s)
- Véronique Dartois
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, USA.
- Department of Medical Sciences, Hackensack Meridian School of Medicine, Nutley, NJ, USA.
| | - Thomas Dick
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, USA
- Department of Medical Sciences, Hackensack Meridian School of Medicine, Nutley, NJ, USA
- Department of Microbiology and Immunology, Georgetown University, Washington, DC, USA
| |
Collapse
|
24
|
Bell KJ, Adams DJ, Brietzke S, Spencer SE. Successful Salvage Therapy Including Tigecycline for Pediatric Mycobacterium abscessus Mastoiditis. Clin Pediatr (Phila) 2024; 63:300-303. [PMID: 37246637 DOI: 10.1177/00099228231176244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Affiliation(s)
- Katherine J Bell
- Department of Pediatrics, Naval Medical Center Portsmouth, Portsmouth, VA, USA
| | - Daniel J Adams
- Department of Pediatrics, Naval Medical Center Portsmouth, Portsmouth, VA, USA
| | | | - Steven E Spencer
- Department of Pediatrics, Brooke Army Medical Center, San Antonio, TX, USA
| |
Collapse
|
25
|
Bao S, Chen S, Zheng J, Ma J, Yang J, Huang H, Duan H. Efficacy of clofazimine-containing regimens for treatment of Mycobacterium avium complex-pulmonary disease in patients unsuitable for standard treatment regimen. Int J Antimicrob Agents 2024; 63:107061. [PMID: 38103753 DOI: 10.1016/j.ijantimicag.2023.107061] [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: 09/04/2023] [Revised: 11/28/2023] [Accepted: 12/08/2023] [Indexed: 12/19/2023]
Abstract
OBJECTIVES Patients with Mycobacterium avium complex-pulmonary disease (MAC-PD) can exhibit contraindications in applying the recommended treatment regimens by the guidelines. Clofazimine (CFZ) is considered a promising drug for MAC-PD treatment and is frequently included in alternative regimens; however, its efficacy remains unclear. METHODS MAC-PD patients, unsuitable for standard regimens, were enrolled continuously in a prospective study at Beijing Chest Hospital. The treatment response of the CFZ-containing regimen was monitored. RESULTS Fifty patients were enrolled in the initial treatment, and 25 patients had a history of anti-TB treatment. Nodular bronchiectasis was observed in 34 patients, while 8 patients exhibited fibrocavitary changes. Additionally, eight patients displayed a combination of both patterns. In a multivariate analysis, MAC-PD patients with CFZ MIC < 0.25 mg/L were significantly associated with culture conversion [OR 8.415, 95% CI (1.983-35.705); P = 0.004]. Among patients who had previous TB treatment history, patients with CFZ MIC < 0.25 mg/L had a higher chance of acquiring culture conversion outcomes [(OR 7.737, 95% CI 1.032-57.989); P = 0.046]. In contrast, among patients with no previous TB treatment history, the RIF-containing regimen had a higher chance of acquiring culture conversion outcomes [(OR 11.038, 95%CI 1.008-120.888); P = 0.049]. CONCLUSION MAC-PD patients unsuitable for standard regimens could benefit from a CFZ-containing regimen, especially for patients with previous TB treatment history and baseline CFZ MIC values lower than 0.25 mg/L.
Collapse
Affiliation(s)
- Shengjuan Bao
- Tuberculosis Department, Beijing Chest Hospital, Capital Medical University, Beijing, China; 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
| | - Suting Chen
- 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
| | - Jifang Zheng
- 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
| | - Junke Ma
- The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China
| | - Jiali Yang
- Puer People's Hospital, Puer, Yunnan, 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.
| | - Hongfei Duan
- Tuberculosis Department, Beijing Chest Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
26
|
Phelps GA, Cheramie MN, Fernando DM, Selchow P, Meyer CJ, Waidyarachchi SL, Dharuman S, Liu J, Meuli M, Molin MD, Killam BY, Murphy PA, Reeve SM, Wilt LA, Anderson SM, Yang L, Lee RB, Temrikar ZH, Lukka PB, Meibohm B, Polikanov YS, Hobbie SN, Böttger EC, Sander P, Lee RE. Development of 2nd generation aminomethyl spectinomycins that overcome native efflux in Mycobacterium abscessus. Proc Natl Acad Sci U S A 2024; 121:e2314101120. [PMID: 38165935 PMCID: PMC10786304 DOI: 10.1073/pnas.2314101120] [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: 08/17/2023] [Accepted: 11/11/2023] [Indexed: 01/04/2024] Open
Abstract
Mycobacterium abscessus (Mab), a nontuberculous mycobacterial (NTM) species, is an emerging pathogen with high intrinsic drug resistance. Current standard-of-care therapy results in poor outcomes, demonstrating the urgent need to develop effective antimycobacterial regimens. Through synthetic modification of spectinomycin (SPC), we have identified a distinct structural subclass of N-ethylene linked aminomethyl SPCs (eAmSPCs) that are up to 64-fold more potent against Mab over the parent SPC. Mechanism of action and crystallography studies demonstrate that the eAmSPCs display a mode of ribosomal inhibition consistent with SPC. However, they exert their increased antimicrobial activity through enhanced accumulation, largely by circumventing efflux mechanisms. The N-ethylene linkage within this series plays a critical role in avoiding TetV-mediated efflux, as lead eAmSPC 2593 displays a mere fourfold susceptibility improvement against Mab ΔtetV, in contrast to the 64-fold increase for SPC. Even a minor shortening of the linkage by a single carbon, akin to 1st generation AmSPC 1950, results in a substantial increase in MICs and a 16-fold rise in susceptibility against Mab ΔtetV. These shifts suggest that longer linkages might modify the kinetics of drug expulsion by TetV, ultimately shifting the equilibrium towards heightened intracellular concentrations and enhanced antimicrobial efficacy. Furthermore, lead eAmSPCs were also shown to synergize with various classes of anti-Mab antibiotics and retain activity against clinical isolates and other mycobacterial strains. Encouraging pharmacokinetic profiles coupled with robust efficacy in Mab murine infection models suggest that eAmSPCs hold the potential to be developed into treatments for Mab and other NTM infections.
Collapse
Affiliation(s)
- Gregory A. Phelps
- Department of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, Memphis, TN38105
- Graduate School of Biomedical Sciences, St. Jude Children’s Research Hospital, Memphis, TN38103
| | - Martin N. Cheramie
- Department of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, Memphis, TN38105
| | - Dinesh M. Fernando
- Department of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, Memphis, TN38105
| | - Petra Selchow
- Institute of Medical Microbiology, University of Zurich, ZurichCH-8006, Switzerland
| | - Christopher J. Meyer
- Department of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, Memphis, TN38105
| | - Samanthi L. Waidyarachchi
- Department of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, Memphis, TN38105
| | - Suresh Dharuman
- Department of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, Memphis, TN38105
| | - Jiuyu Liu
- Department of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, Memphis, TN38105
| | - Michael Meuli
- Institute of Medical Microbiology, University of Zurich, ZurichCH-8006, Switzerland
- National Reference Center for Mycobacteria, ZurichCH-8006, Switzerland
| | - Michael Dal Molin
- Institute of Medical Microbiology, University of Zurich, ZurichCH-8006, Switzerland
| | - Benjamin Y. Killam
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL60607
| | - Patricia A. Murphy
- Department of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, Memphis, TN38105
| | - Stephanie M. Reeve
- Department of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, Memphis, TN38105
| | - Laura A. Wilt
- Department of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, Memphis, TN38105
| | - Shelby M. Anderson
- Department of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, Memphis, TN38105
| | - Lei Yang
- Department of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, Memphis, TN38105
| | - Robin B. Lee
- Department of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, Memphis, TN38105
| | - Zaid H. Temrikar
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN38163
| | - Pradeep B. Lukka
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN38163
| | - Bernd Meibohm
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN38163
| | - Yury S. Polikanov
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL60607
- Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL60607
- Center for Biomolecular Sciences, University of Illinois at Chicago, Chicago, IL60607
| | - Sven N. Hobbie
- Institute of Medical Microbiology, University of Zurich, ZurichCH-8006, Switzerland
| | - Erik C. Böttger
- Institute of Medical Microbiology, University of Zurich, ZurichCH-8006, Switzerland
- National Reference Center for Mycobacteria, ZurichCH-8006, Switzerland
| | - Peter Sander
- Institute of Medical Microbiology, University of Zurich, ZurichCH-8006, Switzerland
- National Reference Center for Mycobacteria, ZurichCH-8006, Switzerland
| | - Richard E. Lee
- Department of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, Memphis, TN38105
| |
Collapse
|
27
|
Dreyer V, Sonnenkalb L, Diricks M, Utpatel C, Barilar I, Mohr V, Niemann S, Kohl TA, Merker M. Use of Whole Genome Sequencing for Mycobacterium tuberculosis Complex Antimicrobial Susceptibility Testing: From Sequence Data to Resistance Profiles. Methods Mol Biol 2024; 2833:195-210. [PMID: 38949712 DOI: 10.1007/978-1-0716-3981-8_18] [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] [Indexed: 07/02/2024]
Abstract
Whole genome sequencing of Mycobacterium tuberculosis complex (MTBC) isolates has been shown to provide accurate predictions for resistance and susceptibility for many first- and second-line anti-tuberculosis drugs. However, bioinformatic pipelines and mutation catalogs to predict antimicrobial resistances in MTBC isolates are often customized and detailed protocols are difficult to access. Here, we provide a step-by-step workflow for the processing and interpretation of short-read sequencing data and give an overview of available analysis pipelines.
Collapse
Affiliation(s)
- Viola Dreyer
- Molecular and Experimental Mycobacteriology, Research Center Borstel - Leibniz Lung Center, Borstel, Germany
| | - Lindsay Sonnenkalb
- Molecular and Experimental Mycobacteriology, Research Center Borstel - Leibniz Lung Center, Borstel, Germany
| | - Margo Diricks
- Molecular and Experimental Mycobacteriology, Research Center Borstel - Leibniz Lung Center, Borstel, Germany
| | - Christian Utpatel
- Molecular and Experimental Mycobacteriology, Research Center Borstel - Leibniz Lung Center, Borstel, Germany
| | - Ivan Barilar
- Molecular and Experimental Mycobacteriology, Research Center Borstel - Leibniz Lung Center, Borstel, Germany
| | - Vanessa Mohr
- Molecular and Experimental Mycobacteriology, Research Center Borstel - Leibniz Lung Center, Borstel, Germany
| | - Stefan Niemann
- Molecular and Experimental Mycobacteriology, Research Center Borstel - Leibniz Lung Center, Borstel, Germany
| | - Thomas A Kohl
- Molecular and Experimental Mycobacteriology, Research Center Borstel - Leibniz Lung Center, Borstel, Germany
| | - Matthias Merker
- Evolution of the Resistome, Research Center Borstel - Leibniz Lung Center, Borstel, Germany.
| |
Collapse
|
28
|
Reil I, Barbić L, Kompes G, Tuk MZ, Duvnjak S, Cvetnić Ž, Habrun B, Arapović J, Špičić S. Risk of zoonoses involving slow-growing non-tuberculous mycobacteria: Survey of antimicrobial resistance among strains from domestic and wild animals. J Glob Antimicrob Resist 2023; 35:6-10. [PMID: 37573944 DOI: 10.1016/j.jgar.2023.08.002] [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: 03/03/2023] [Revised: 07/10/2023] [Accepted: 08/01/2023] [Indexed: 08/15/2023] Open
Abstract
OBJECTIVES Non-tuberculous mycobacteria are opportunistic pathogens that cause disease mainly in immunocompromised hosts. The present study assessed the prevalence of antibiotic resistance among such mycobacteria from domestic and wild animals in Croatia sampled during several years within a national surveillance program. METHODS A total of 44 isolates belonging to nine slow-growing species were genotyped and analyzed for susceptibility to 13 antimicrobials often used to treat non-tuberculous mycobacterial infections in humans. RESULTS Most prevalent resistance was to moxifloxacin (77.3%), doxycycline (76.9%), and rifampicin (76.9%), followed by ciprofloxacin (65.4%), trimethoprim-sulfamethoxazole (65.4%), and linezolid (61.4%). Few isolates were resistant to rifabutin (7.7%) or amikacin (6.8%). None of the isolates was resistant to clarithromycin. Nearly all isolates (86.4%) were resistant to multiple antibiotics. CONCLUSION Our findings suggest substantial risk that human populations may experience zoonotic infections with non-tuberculous mycobacteria that will be difficult to treat using the current generation of antibiotics. Future work should clarify how resistance emerges in wild populations of non-tuberculous mycobacteria.
Collapse
Affiliation(s)
- Irena Reil
- Croatian Veterinary Institute, Zagreb, Croatia
| | - Ljubo Barbić
- Faculty of Veterinary Medicine, University of Zagreb, Zagreb, Croatia
| | | | | | | | | | | | - Jurica Arapović
- University Clinical Hospital Mostar, Department of Infectious Diseases, Mostar, Bosnia and Herzegovina; School of Medicine, University of Mostar, Mostar, Bosnia and Herzegovina
| | | |
Collapse
|
29
|
Sirichoat A, Kaewprasert O, Hinwan Y, Faksri K. Phenotypic drug-susceptibility profiles and genetic analysis based on whole-genome sequencing of Mycobacterium avium complex isolates in Thailand. PLoS One 2023; 18:e0294677. [PMID: 37992075 PMCID: PMC10664917 DOI: 10.1371/journal.pone.0294677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 10/31/2023] [Indexed: 11/24/2023] Open
Abstract
Mycobacterium avium complex (MAC) infections are a significant clinical challenge. Determining drug-susceptibility profiles and the genetic basis of drug resistance is crucial for guiding effective treatment strategies. This study aimed to determine the drug-susceptibility profiles of MAC clinical isolates and to investigate the genetic basis conferring drug resistance using whole-genome sequencing (WGS) analysis. Drug-susceptibility profiles based on minimum inhibitory concentration (MIC) assays were determined for 38 MAC clinical isolates (12 Mycobacterium avium and 26 Mycobacterium intracellulare). Mutations associated with drug resistance were identified through genome analysis of these isolates, and their phylogenetic relationships were also examined. Drug resistance, based on MIC values, was most commonly observed for moxifloxacin (81.6%), followed by linezolid (78.9%), clarithromycin (44.7%) and amikacin (36.8%). We identified specific mutations associated with resistance to amikacin. These include the rrs mutation at C464T in amikacin intermediate-resistance M. avium, and two mutations at T250A and G1453T in amikacin non-susceptible M. intracellulare. Mutations in rrl at A2058G, A2059C and A2059G were potentially linked to clarithromycin resistance. MAC clinical isolates not susceptible to linezolid exhibited mutations in rplC at G237C and C459T, as well as two rplD mutations at G443A and A489G. GyrB substitution Thr521Ala (T521A) was identified in moxifloxacin non-susceptible isolates, which may contribute to this resistance. A phylogeny of our MAC isolates revealed high levels of genetic diversity. Our findings suggest that the standard treatment regimen for MAC infections using moxifloxacin, linezolid, clarithromycin and amikacin may be driving development of resistance, potentially due to specific mutations. The combination of phenotypic and genotypic susceptibility testing can be valuable in guiding the clinical use of drugs for the treatment of MAC infections.
Collapse
Affiliation(s)
- Auttawit Sirichoat
- Department of Microbiology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Research and Diagnostic Center for Emerging Infectious Diseases (RCEID), Khon Kaen University, Khon Kaen, Thailand
| | - Orawee Kaewprasert
- Department of Microbiology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Research and Diagnostic Center for Emerging Infectious Diseases (RCEID), Khon Kaen University, Khon Kaen, Thailand
| | - Yothin Hinwan
- Department of Microbiology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Research and Diagnostic Center for Emerging Infectious Diseases (RCEID), Khon Kaen University, Khon Kaen, Thailand
| | - Kiatichai Faksri
- Department of Microbiology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Research and Diagnostic Center for Emerging Infectious Diseases (RCEID), Khon Kaen University, Khon Kaen, Thailand
| |
Collapse
|
30
|
Reil I, Špičić S, Barbić L, Duvnjak S, Kompes G, Benić M, Stojević D, Cvetnić Ž, Arapović J, Zdelar-Tuk M. Antimicrobial Resistance in Rapidly Growing Nontuberculous Mycobacteria among Domestic and Wild Animals Emphasizing the Zoonotic Potential. Microorganisms 2023; 11:2520. [PMID: 37894179 PMCID: PMC10609087 DOI: 10.3390/microorganisms11102520] [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: 08/30/2023] [Revised: 09/29/2023] [Accepted: 10/08/2023] [Indexed: 10/29/2023] Open
Abstract
Non-tuberculous mycobacteria (NTM) are opportunistic pathogens capable of causing infections in humans and animals. The aim of this study was to demonstrate the potential role of domestic and wild animals as a reservoir of multiple resistant, rapidly growing NTM strains representing a potential zoonotic threat to humans. A total of 87 animal isolates belonging to 11 rapidly growing species (visible colonies appear within three to seven days) were genotyped and tested for susceptibility to the 15 most commonly used antibiotics in the treatment of such infections in a human clinic. By determining the antimicrobial susceptibility, the most prevalent resistance was found to cephalosporins (>50%), followed by amoxicillin-clavulanate (31.0%), clarithromycin (23.0%), tobramycin (14.9%) and doxycycline (10.3%). Resistance to imipenem, ciprofloxacin, minocycline and linezolid was notably lower (<7.0%). All tested isolates were susceptible to amikacin and moxifloxacin. The most frequent resistance was proved in the most pathogenic species: M. fortuitum, M. neoaurum, M. vaccae and M. porcinum. Meanwhile, other species displayed a higher sensitivity rate. No significant resistance differences between domestic and wild animals were found. The established significant frequency of resistance highlights the significant zoonotic potential posed by circulating rapidly growing NTM strains, which could lead to challenges in the treatment of these infections.
Collapse
Affiliation(s)
- Irena Reil
- Croatian Veterinary Institute, 10000 Zagreb, Croatia; (I.R.); (S.Š.); (M.B.); (D.S.); (Ž.C.); (M.Z.-T.)
| | - Silvio Špičić
- Croatian Veterinary Institute, 10000 Zagreb, Croatia; (I.R.); (S.Š.); (M.B.); (D.S.); (Ž.C.); (M.Z.-T.)
| | - Ljubo Barbić
- The Faculty of Veterinary Medicine, University of Zagreb, 10000 Zagreb, Croatia;
| | - Sanja Duvnjak
- Croatian Veterinary Institute, 10000 Zagreb, Croatia; (I.R.); (S.Š.); (M.B.); (D.S.); (Ž.C.); (M.Z.-T.)
| | - Gordan Kompes
- Croatian Veterinary Institute, 10000 Zagreb, Croatia; (I.R.); (S.Š.); (M.B.); (D.S.); (Ž.C.); (M.Z.-T.)
| | - Miroslav Benić
- Croatian Veterinary Institute, 10000 Zagreb, Croatia; (I.R.); (S.Š.); (M.B.); (D.S.); (Ž.C.); (M.Z.-T.)
| | - Dora Stojević
- Croatian Veterinary Institute, 10000 Zagreb, Croatia; (I.R.); (S.Š.); (M.B.); (D.S.); (Ž.C.); (M.Z.-T.)
| | - Željko Cvetnić
- Croatian Veterinary Institute, 10000 Zagreb, Croatia; (I.R.); (S.Š.); (M.B.); (D.S.); (Ž.C.); (M.Z.-T.)
| | - Jurica Arapović
- Department of Infectious Diseases, University Clinical Hospital Mostar, 88000 Mostar, Bosnia and Herzegovina;
- School of Medicine, University of Mostar, 88000 Mostar, Bosnia and Herzegovina
| | - Maja Zdelar-Tuk
- Croatian Veterinary Institute, 10000 Zagreb, Croatia; (I.R.); (S.Š.); (M.B.); (D.S.); (Ž.C.); (M.Z.-T.)
| |
Collapse
|
31
|
Batista S, Fernandez-Pittol M, Nicolás LS, Martínez D, Rubio M, Garrigo M, Vila J, Tudó G, González-Martin J. In Vitro Effect of Three-Antibiotic Combinations plus Potential Antibiofilm Agents against Biofilm-Producing Mycobacterium avium and Mycobacterium intracellulare Clinical Isolates. Antibiotics (Basel) 2023; 12:1409. [PMID: 37760706 PMCID: PMC10526108 DOI: 10.3390/antibiotics12091409] [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: 08/04/2023] [Revised: 09/01/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
Patients with chronic pulmonary diseases infected by Mycobacterium avium complex (MAC) often develop complications and suffer from treatment failure due to biofilm formation. There is a lack of correlation between in vitro susceptibility tests and the treatment of clinical isolates producing biofilm. We performed susceptibility tests of 10 different three-drug combinations, including two recommended in the guidelines, in biofilm forms of eight MAC clinical isolates. Biofilm developed in the eight isolates following incubation of the inoculum for 3 weeks. Then, the biofilm was treated with three-drug combinations with and without the addition of potential antibiofilm agents (PAAs). Biofilm bactericidal concentrations (BBCs) were determined using the Vizion lector system. All selected drug combinations showed synergistic activity, reducing BBC values compared to those treated with single drugs, but BBC values remained high enough to treat patients. However, with the addition of PAAs, the BBCs steadily decreased, achieving similar values to the combinations in planktonic forms and showing synergistic activity in all the combinations and in both species. In conclusion, three-drug combinations with PAAs showed synergistic activity in biofilm forms of MAC isolates. Our results suggest the need for clinical studies introducing PAAs combined with antibiotics for the treatment of patients with pulmonary diseases infected by MAC.
Collapse
Affiliation(s)
- Sara Batista
- Unitat de Microbiologia, Department de Fonaments Clínics, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, c/Casanova 143, 08036 Barcelona, Spain; (S.B.); (M.F.-P.); (J.V.)
- ISGlobal Barcelona, Institute for Global Health, c/Rosselló 132, 08036 Barcelona, Spain
- Servei de Microbiologia, CDB, Hospital Clínic de Barcelona, c/Villarroel 170, 08036 Barcelona, Spain; (L.S.N.); (D.M.)
| | - Mariana Fernandez-Pittol
- Unitat de Microbiologia, Department de Fonaments Clínics, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, c/Casanova 143, 08036 Barcelona, Spain; (S.B.); (M.F.-P.); (J.V.)
- ISGlobal Barcelona, Institute for Global Health, c/Rosselló 132, 08036 Barcelona, Spain
- Servei de Microbiologia, CDB, Hospital Clínic de Barcelona, c/Villarroel 170, 08036 Barcelona, Spain; (L.S.N.); (D.M.)
| | - Lorena San Nicolás
- Servei de Microbiologia, CDB, Hospital Clínic de Barcelona, c/Villarroel 170, 08036 Barcelona, Spain; (L.S.N.); (D.M.)
| | - Diego Martínez
- Servei de Microbiologia, CDB, Hospital Clínic de Barcelona, c/Villarroel 170, 08036 Barcelona, Spain; (L.S.N.); (D.M.)
| | - Marc Rubio
- Servei de Microbiologia, Fundació de Gestió de l’Hospital de la Santa Creu i Sant Pau, c/Sant Quintí 89, 08026 Barcelona, Spain; (M.R.); (M.G.)
- Institut d’Investigació Biomèdica Sant Pau (IIB Sant Pau), c/Sant Quintí, 89, 08026 Barcelona, Spain
| | - Montserrat Garrigo
- Servei de Microbiologia, Fundació de Gestió de l’Hospital de la Santa Creu i Sant Pau, c/Sant Quintí 89, 08026 Barcelona, Spain; (M.R.); (M.G.)
- Institut d’Investigació Biomèdica Sant Pau (IIB Sant Pau), c/Sant Quintí, 89, 08026 Barcelona, Spain
| | - Jordi Vila
- Unitat de Microbiologia, Department de Fonaments Clínics, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, c/Casanova 143, 08036 Barcelona, Spain; (S.B.); (M.F.-P.); (J.V.)
- ISGlobal Barcelona, Institute for Global Health, c/Rosselló 132, 08036 Barcelona, Spain
- Servei de Microbiologia, CDB, Hospital Clínic de Barcelona, c/Villarroel 170, 08036 Barcelona, Spain; (L.S.N.); (D.M.)
- CIBER of Infectious Diseases (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Griselda Tudó
- Unitat de Microbiologia, Department de Fonaments Clínics, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, c/Casanova 143, 08036 Barcelona, Spain; (S.B.); (M.F.-P.); (J.V.)
- ISGlobal Barcelona, Institute for Global Health, c/Rosselló 132, 08036 Barcelona, Spain
- Servei de Microbiologia, CDB, Hospital Clínic de Barcelona, c/Villarroel 170, 08036 Barcelona, Spain; (L.S.N.); (D.M.)
| | - Julian González-Martin
- Unitat de Microbiologia, Department de Fonaments Clínics, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, c/Casanova 143, 08036 Barcelona, Spain; (S.B.); (M.F.-P.); (J.V.)
- ISGlobal Barcelona, Institute for Global Health, c/Rosselló 132, 08036 Barcelona, Spain
- Servei de Microbiologia, CDB, Hospital Clínic de Barcelona, c/Villarroel 170, 08036 Barcelona, Spain; (L.S.N.); (D.M.)
- CIBER of Infectious Diseases (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
32
|
Wang XY, Jia QN, Li J. Treatment of non-tuberculosis mycobacteria skin infections. Front Pharmacol 2023; 14:1242156. [PMID: 37731736 PMCID: PMC10508292 DOI: 10.3389/fphar.2023.1242156] [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: 06/18/2023] [Accepted: 08/25/2023] [Indexed: 09/22/2023] Open
Abstract
Non-tuberculosis mycobacteria (NTM) skin infections have become increasingly prevalent in recent years, presenting a unique challenge in clinical management. This review explored the complexities of NTM infections localized to the superficial tissues and provided valuable insights into the optimal therapeutic strategies. The antibiotic selection should base on NTM species and their susceptibility profiles. It is recommended to adopt a comprehensive approach that considers the unique characteristics of superficial tissues to improve treatment effectiveness and reduce the incidence of adverse reactions, infection recurrence, and treatment failure. Infection control measures, patient education, and close monitoring should complement the treatment strategies to achieve favorable outcomes in managing NTM skin infections. Further efforts are warranted to elucidate factors and mechanisms contributing to treatment resistance and relapse. Future research should focus on exploring novel treatment options, innovative drug development/delivery platforms, and precise methodologies for determining therapeutic duration. Longitudinal studies are also needed to assess the long-term safety profiles of the integrated approaches.
Collapse
Affiliation(s)
| | | | - Jun Li
- Department of Dermatology and Venereology, Peking Union Medical College Hospital (Dongdan Campus), Beijing, China
| |
Collapse
|
33
|
Levendosky K, Janisch N, Quadri LEN. Comprehensive essentiality analysis of the Mycobacterium kansasii genome by saturation transposon mutagenesis and deep sequencing. mBio 2023; 14:e0057323. [PMID: 37350613 PMCID: PMC10470612 DOI: 10.1128/mbio.00573-23] [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: 03/07/2023] [Accepted: 05/01/2023] [Indexed: 06/24/2023] Open
Abstract
Mycobacterium kansasii (Mk) is an opportunistic pathogen that is frequently isolated from urban water systems, posing a health risk to susceptible individuals. Despite its ability to cause tuberculosis-like pulmonary disease, very few studies have probed the genetics of this opportunistic pathogen. Here, we report a comprehensive essentiality analysis of the Mk genome. Deep sequencing of a high-density library of Mk Himar1 transposon mutants revealed that 86.8% of the chromosomal thymine-adenine (TA) dinucleotide target sites were permissive to insertion, leaving 13.2% TA sites unoccupied. Our analysis identified 394 of the 5,350 annotated open reading frames (ORFs) as essential. The majority of these essential ORFs (84.8%) share essential mutual orthologs with Mycobacterium tuberculosis (Mtb). A comparative genomics analysis identified 139 Mk essential ORFs that share essential orthologs in four other species of mycobacteria. Thirteen Mk essential ORFs share orthologs in all four species that were identified as being not essential, while only two Mk essential ORFs are absent in all species compared. We used the essentiality data and a comparative genomics analysis reported here to highlight differences in essentiality between candidate Mtb drug targets and the corresponding Mk orthologs. Our findings suggest that the Mk genome encodes redundant or additional pathways that may confound validation of potential Mtb drugs and drug target candidates against the opportunistic pathogen. Additionally, we identified 57 intergenic regions containing four or more consecutive unoccupied TA sites. A disproportionally large number of these regions were located upstream of pe/ppe genes. Finally, we present an essentiality and orthology analysis of the Mk pRAW-like plasmid, pMK1248. IMPORTANCE Mk is one of the most common nontuberculous mycobacterial pathogens associated with tuberculosis-like pulmonary disease. Drug resistance emergence is a threat to the control of Mk infections, which already requires long-term, multidrug courses. A comprehensive understanding of Mk biology is critical to facilitate the development of new and more efficacious therapeutics against Mk. We combined transposon-based mutagenesis with analysis of insertion site identification data to uncover genes and other genomic regions required for Mk growth. We also compared the gene essentiality data set of Mk to those available for several other mycobacteria. This analysis highlighted key similarities and differences in the biology of Mk compared to these other species. Altogether, the genome-wide essentiality information generated and the results of the cross-species comparative genomics analysis represent valuable resources to assist the process of identifying and prioritizing potential Mk drug target candidates and to guide future studies on Mk biology.
Collapse
Affiliation(s)
- Keith Levendosky
- Department of Biology, Brooklyn College, City University of New York, Brooklyn, New York, USA
- Biology Program, Graduate Center, Biology Program, Graduate Center, City University of New York, New York, New York, USA
| | - Niklas Janisch
- Department of Biology, Brooklyn College, City University of New York, Brooklyn, New York, USA
- Biology Program, Graduate Center, Biology Program, Graduate Center, City University of New York, New York, New York, USA
| | - Luis E. N. Quadri
- Department of Biology, Brooklyn College, City University of New York, Brooklyn, New York, USA
- Biology Program, Graduate Center, Biology Program, Graduate Center, City University of New York, New York, New York, USA
- Biochemistry Program, Graduate Center, City University of New York, New York, New York, USA
| |
Collapse
|
34
|
Brčić J, Tong A, Wender PA, Cegelski L. Conjugation of Vancomycin with a Single Arginine Improves Efficacy against Mycobacteria by More Effective Peptidoglycan Targeting. J Med Chem 2023; 66:10226-10237. [PMID: 37477249 PMCID: PMC10783851 DOI: 10.1021/acs.jmedchem.3c00565] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/22/2023]
Abstract
Drug resistant bacterial infections have emerged as one of the greatest threats to public health. The discovery and development of new antimicrobials and anti-infective strategies are urgently needed to address this challenge. Vancomycin is one of the most important antibiotics for the treatment of Gram-positive infections. Here, we introduce the vancomycin-arginine conjugate (V-R) as a highly effective antimicrobial against actively growing mycobacteria and difficult-to-treat mycobacterial biofilm populations. Further improvement in efficacy through combination treatment of V-R to inhibit peptidoglycan synthesis and ethambutol to inhibit arabinogalactan synthesis underscores the ability to identify compound synergies to more effectively target the Achilles heel of the cell-wall assembly. Moreover, we introduce mechanistic activity data and a molecular model derived from a d-Ala-d-Ala-bound vancomycin structure that we hypothesize underlies the molecular basis for the antibacterial improvement attributed to the arginine modification that is specific to peptidoglycan chemistry employed by mycobacteria and distinct from Gram-positive pathogens.
Collapse
Affiliation(s)
- Jasna Brčić
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| | - Alan Tong
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| | - Paul A. Wender
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA 94305, USA
| | - Lynette Cegelski
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
35
|
Alcaraz M, Edwards TE, Kremer L. New therapeutic strategies for Mycobacterium abscessus pulmonary diseases - untapping the mycolic acid pathway. Expert Rev Anti Infect Ther 2023; 21:813-829. [PMID: 37314394 PMCID: PMC10529309 DOI: 10.1080/14787210.2023.2224563] [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: 02/28/2023] [Accepted: 06/08/2023] [Indexed: 06/15/2023]
Abstract
INTRODUCTION Treatment options against Mycobacterium abscessus infections are very limited. New compounds are needed to cure M. abscessus pulmonary diseases. While the mycolic acid biosynthetic pathway has been largely exploited for the treatment of tuberculosis, this metabolic process has been overlooked in M. abscessus, although it offers many potential drug targets for the treatment of this opportunistic pathogen. AREAS COVERED Herein, the authors review the role of the MmpL3 membrane protein and the enoyl-ACP reductase InhA involved in the transport and synthesis of mycolic acids, respectively. They discuss their importance as two major vulnerable drug targets in M. abscessus and report the activity of MmpL3 and InhA inhibitors. In particular, they focus on NITD-916, a direct InhA inhibitor against M. abscessus, particularly warranted in the context of multidrug resistance. EXPERT OPINION There is an increasing body of evidence validating the mycolic acid pathway as an attractive drug target to be further exploited for M. abscessus lung disease treatments. The NITD-916 studies provide a proof-of-concept that direct inhibitors of InhA are efficient in vitro, in macrophages and in zebrafish. Future work is now required to improve the activity and pharmacological properties of these inhibitors and their evaluation in pre-clinical models.
Collapse
Affiliation(s)
- Matthéo Alcaraz
- Centre National de la Recherche Scientifique UMR 9004, Institut de Recherche en Infectiologie de Montpellier (IRIM), Université de Montpellier, 1919 route de Mende, 34293, Montpellier, France
| | - Thomas E. Edwards
- UCB BioSciences, Bainbridge Island, WA 98109 USA
- Seattle Structural Genomics Center for Infectious Disease (SSGCID), Seattle, WA 98109 USA
| | - Laurent Kremer
- Centre National de la Recherche Scientifique UMR 9004, Institut de Recherche en Infectiologie de Montpellier (IRIM), Université de Montpellier, 1919 route de Mende, 34293, Montpellier, France
- INSERM, IRIM, 34293 Montpellier, France
| |
Collapse
|
36
|
Thapa J, Chizimu JY, Kitamura S, Akapelwa ML, Suwanthada P, Miura N, Toyting J, Nishimura T, Hasegawa N, Nishiuchi Y, Gordon SV, Nakajima C, Suzuki Y. Characterization of DNA Gyrase Activity and Elucidation of the Impact of Amino Acid Substitution in GyrA on Fluoroquinolone Resistance in Mycobacterium avium. Microbiol Spectr 2023; 11:e0508822. [PMID: 37067420 PMCID: PMC10269562 DOI: 10.1128/spectrum.05088-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 03/28/2023] [Indexed: 04/18/2023] Open
Abstract
Mycobacterium avium, a member of the M. avium complex (MAC), is the major pathogen contributing to nontuberculous mycobacteria (NTM) infections worldwide. Fluoroquinolones (FQs) are recommended for the treatment of macrolide-resistant MACs. The association of FQ resistance and mutations in the quinolone resistance-determining region (QRDR) of gyrA of M. avium is not yet clearly understood, as many FQ-resistant clinical M. avium isolates do not have such mutations. This study aimed to elucidate the role of amino acid substitution in the QRDR of M. avium GyrA in the development of FQ resistance. We found four clinical M. avium subsp. hominissuis isolates with Asp-to-Gly change at position 95 (Asp95Gly) and Asp95Tyr mutations in gyrA that were highly resistant to FQs and had 2- to 32-fold-higher MICs than the wild-type (WT) isolates. To clarify the contribution of amino acid substitutions to FQ resistance, we produced recombinant WT GyrA, GyrB, and four GyrA mutant proteins (Ala91Val, Asp95Ala, Asp95Gly, and Asp95Tyr) to elucidate their potential role in FQ resistance, using them to perform FQ-inhibited DNA supercoiling assays. While all the mutant GyrAs contributed to the higher (1.3- to 35.6-fold) FQ 50% inhibitory concentration (IC50) than the WT, Asp95Tyr was the most resistant mutant, with an IC50 15- to 35.6-higher than that of the WT, followed by the Asp95Gly mutant, with an IC50 12.5- to 17.6-fold higher than that of the WT, indicating that these amino acid substitutions significantly reduced the inhibitory activity of FQs. Our results showed that amino acid substitutions in the gyrA of M. avium contribute to FQ resistance. IMPORTANCE The emergence of fluoroquinolone (FQ) resistance has further compounded the control of emerging Mycobacterium avium-associated nontuberculous mycobacteria infections worldwide. For M. avium, the association of FQ resistance and mutations in the quinolone resistance-determining region (QRDR) of gyrA is not yet clearly understood. Here, we report that four clinical M. avium isolates with a mutation in the QRDR of gyrA were highly resistant to FQs. We further clarified the impact of mutations in the QRDR of GyrA proteins by performing in vitro FQ-inhibited DNA supercoiling assays. These results confirmed that, like in Mycobacterium tuberculosis, mutations in the QRDR of gyrA also strongly contribute to FQ resistance in M. avium. Since many FQ-resistant M. avium isolates do have these mutations, the detailed molecular mechanism of FQ resistance in M. avium needs further exploration.
Collapse
Affiliation(s)
- Jeewan Thapa
- Division of Bioresources, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Joseph Yamweka Chizimu
- Division of Bioresources, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
- Zambian National Public Health Institute, Ministry of Health, Lusaka, Zambia
| | - Soyoka Kitamura
- Department of Medical Laboratory Science, Faculty of Health Sciences, Hokkaido University, Sapporo, Japan
| | - Mwangala Lonah Akapelwa
- Division of Bioresources, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Pondpan Suwanthada
- Division of Bioresources, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Nami Miura
- Division of Bioresources, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Jirachaya Toyting
- Division of Bioresources, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | | | - Naoki Hasegawa
- Department of Infectious Diseases, Keio University School of Medicine, Tokyo, Japan
| | - Yukiko Nishiuchi
- Toneyama Institute for Tuberculosis Research, Osaka City University Medical School, Osaka, Japan
- Office of Academic Research and Industry-Government Collaboration, Section of Microbial Genomics and Ecology, Hiroshima University, Higashi-Hiroshima, Japan
| | - Stephen V. Gordon
- School of Veterinary Medicine, University College Dublin, Dublin, Ireland
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Chie Nakajima
- Division of Bioresources, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
- Institute for Vaccine Research and Development, Hokkaido University, Sapporo, Japan
| | - Yasuhiko Suzuki
- Division of Bioresources, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
- Institute for Vaccine Research and Development, Hokkaido University, Sapporo, Japan
| |
Collapse
|
37
|
Roy CF, Balakrishnan K, Boudewyns A, Cheng A, Chun RH, Daniel SJ, Fayoux P, Hart C, Hemansson A, Hewitt R, Hsu WC, Kuo M, Liu C, Maddalozzo J, Messner AH, Pransky S, Rahbar R, Rickert S, Roy S, Russell J, Rutter MJ, Sie KCY, Sidell D, Smith R, Soma M, Spratley J, Watters K, White DR, Wolter N, Zalzal G, Yeung JC. International Pediatric Otolaryngology Group: Consensus guidelines on the diagnosis and management of non-tuberculous mycobacterial cervicofacial lymphadenitis. Int J Pediatr Otorhinolaryngol 2023; 166:111469. [PMID: 36764081 DOI: 10.1016/j.ijporl.2023.111469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 01/23/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023]
Abstract
INTRODUCTION Non-tuberculous mycobacterial (NTM) infection commonly manifests as subacute or chronic cervicofacial lymphadenitis in immunocompetent children. The optimal management of this pathology remains controversial. OBJECTIVES This international consensus guideline aims to understand the practice patterns for NTM cervicofacial lymphadenitis and to address the primary diagnostic and management challenges. METHODS A modified three-iterative Delphi method was used to establish expert recommendations on the diagnostic considerations, expectant or medical management, and operative considerations. The recommendations herein are derived from current expert consensus and critical review of the literature. SETTING Multinational, multi-institutional, tertiary pediatric hospitals. RESULTS Consensus recommendations include diagnostic work-up, goals of treatment and management options including surgery, prolonged antibiotic therapy and observation. CONCLUSION The recommendations formulated in this International Pediatric Otolaryngology Group (IPOG) consensus statement on the diagnosis and management of patients with NTM lymphadenitis are aimed at improving patient care and promoting future hypothesis generation.
Collapse
Affiliation(s)
- Catherine F Roy
- Department of Otolaryngology Head & Neck Surgery, McGill University Health Center, Montreal, QC, Canada
| | - Karthik Balakrishnan
- Department of Otolaryngology-Head & Neck Surgery, Lucile Packard Children's Hospital, Stanford University, Palo Alto, CA, USA
| | - An Boudewyns
- Department of Otorhinolaryngology, Head and Neck Surgery, Antwerp University Hospital, University of Antwerp, Belgium
| | - Alan Cheng
- Department of Pediatric Otolaryngology, The Sydney Children's Hospital Network-Westmead Campus, The University of Sydney, Sydney, NSW, Australia
| | - Robert H Chun
- Department of Otolaryngology, Children's Hospital of Wisconsin, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Sam J Daniel
- Department of Otolaryngology Head & Neck Surgery, McGill University Health Center, Montreal, QC, Canada
| | - Pierre Fayoux
- Department of Pediatric Otolaryngology-Head and Neck Surgery, CHU Lille, F-59000, Lille, France
| | - Catherine Hart
- Division of Pediatric Otolaryngology-Head and Neck Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Ann Hemansson
- Department of Otorhinolaryngology, Head and Neck Surgery, Lund University Hospital, Lund, Sweden
| | - Richard Hewitt
- Department of Ear, Nose and Throat Surgery, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Wei-Chung Hsu
- Division of Pediatric Otolaryngology, National Taiwan University College of Medicine and Children's Hospital, Taipei, Taiwan
| | - Michael Kuo
- Department of Pediatric Otolaryngology, Birmingham Children's Hospital, Birmingham, United Kingdom
| | - Christopher Liu
- Division of Pediatric Otolaryngology, UT Southwestern Medical Center, Dallas, TX, USA
| | - John Maddalozzo
- Department of Otolaryngology/Head and Neck Surgery, Lurie Children's Hospital, Chicago, IL, USA
| | - Anna H Messner
- Department of Otolaryngology/Head & Neck Surgery, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, USA
| | - Seth Pransky
- Department of Pediatric Otolaryngology, Rady Children's Hospital, San Diego, CA, USA
| | - Reza Rahbar
- Department of Pediatric Otolaryngology and Communication Enhancement, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Scott Rickert
- Division of Pediatric Otolaryngology, Hassenfeld Children's Hospital, New York University Langone Health, New York, NY, USA
| | - Soham Roy
- Department of Otolaryngology-Head and Neck Surgery, Children's Hospital Colorado, Aurora, CO, USA
| | - John Russell
- Department of Pediatric Otorhinolaryngology, Children's Health Ireland (Crumlin), Dublin, Ireland
| | - Michael J Rutter
- Division of Pediatric Otolaryngology-Head and Neck Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Kathleen C Y Sie
- Department of Otolaryngology Head and Neck Surgery, Seattle Children's Hospital, University of Washington, Seattle, USA
| | - Douglas Sidell
- Department of Otolaryngology-Head & Neck Surgery, Lucile Packard Children's Hospital, Stanford University, Palo Alto, CA, USA
| | - Richard Smith
- Department of Otolaryngology-Head & Neck Surgery, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| | - Marlene Soma
- Department of Paediatric Otolaryngology, Sydney Children's Hospital, Randwick, NSW, Australia
| | - Jorge Spratley
- Pediatric Otorhinolaryngology, University of Porto Faculty of Medicine, S. Joao Hospital Center, CINTESIS, Porto, Portugal
| | - Karen Watters
- Department of Pediatric Otolaryngology, Rady Children's Hospital, San Diego, CA, USA
| | - David R White
- Division of Pediatric Otolaryngology, MUSC Shawn Jenkins Children's Hospital, Charleston, SC, USA
| | - Nikolaus Wolter
- Department of Otolaryngology - Head and Neck Surgery, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - George Zalzal
- Division of Otolaryngology, Children's National Medical Center, George Washington University, Washington, DC, USA
| | - Jeffrey C Yeung
- Department of Otolaryngology Head & Neck Surgery, McGill University Health Center, Montreal, QC, Canada.
| |
Collapse
|
38
|
Janisch N, Levendosky K, Budell WC, Quadri LEN. Genetic Underpinnings of Carotenogenesis and Light-Induced Transcriptome Remodeling in the Opportunistic Pathogen Mycobacterium kansasii. Pathogens 2023; 12:86. [PMID: 36678434 PMCID: PMC9861118 DOI: 10.3390/pathogens12010086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 12/24/2022] [Accepted: 12/28/2022] [Indexed: 01/06/2023] Open
Abstract
Mycobacterium kansasii (Mk) causes opportunistic pulmonary infections with tuberculosis-like features. The bacterium is well known for its photochromogenicity, i.e., the production of carotenoid pigments in response to light. The genetics defining the photochromogenic phenotype of Mk has not been investigated and defined pigmentation mutants to facilitate studies on the role of carotenes in the bacterium's biology are not available thus far. In this study, we set out to identify genetic determinants involved in Mk photochromogenicity. We screened a library of ~150,000 transposon mutants for colonies with pigmentation abnormalities. The screen rendered a collection of ~200 mutants. Each of these mutants could be assigned to one of four distinct phenotypic groups. The insertion sites in the mutant collection clustered in three chromosomal regions. A combination of phenotypic analysis, sequence bioinformatics, and gene expression studies linked these regions to carotene biosynthesis, carotene degradation, and monounsaturated fatty acid biosynthesis. Furthermore, introduction of the identified carotenoid biosynthetic gene cluster into non-pigmented Mycobacterium smegmatis endowed the bacterium with photochromogenicity. The studies also led to identification of MarR-type and TetR/AcrR-type regulators controlling photochromogenicity and carotenoid breakdown, respectively. Lastly, the work presented also provides a first insight into the Mk transcriptome changes in response to light.
Collapse
Affiliation(s)
- Niklas Janisch
- Department of Biology, Brooklyn College, City University of New York, 2900 Bedford Avenue, Brooklyn, NY 11210, USA
- Biology Program, Graduate Center, City University of New York, 365 Fifth Avenue, New York, NY 10016, USA
| | - Keith Levendosky
- Department of Biology, Brooklyn College, City University of New York, 2900 Bedford Avenue, Brooklyn, NY 11210, USA
- Biology Program, Graduate Center, City University of New York, 365 Fifth Avenue, New York, NY 10016, USA
| | - William C. Budell
- Department of Biology, Brooklyn College, City University of New York, 2900 Bedford Avenue, Brooklyn, NY 11210, USA
- Biology Program, Graduate Center, City University of New York, 365 Fifth Avenue, New York, NY 10016, USA
| | - Luis E. N. Quadri
- Department of Biology, Brooklyn College, City University of New York, 2900 Bedford Avenue, Brooklyn, NY 11210, USA
- Biology Program, Graduate Center, City University of New York, 365 Fifth Avenue, New York, NY 10016, USA
- Biochemistry Program, Graduate Center, City University of New York, 365 Fifth Avenue, New York, NY 10016, USA
| |
Collapse
|
39
|
Renau-Mínguez C, Herrero-Abadía P, Ruiz-Rodriguez P, Sentandreu V, Torrents E, Chiner-Oms Á, Torres-Puente M, Comas I, Julián E, Coscolla M. Genomic analysis of Mycobacterium brumae sustains its nonpathogenic and immunogenic phenotype. Front Microbiol 2023; 13:982679. [PMID: 36687580 PMCID: PMC9850167 DOI: 10.3389/fmicb.2022.982679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 12/06/2022] [Indexed: 01/07/2023] Open
Abstract
Mycobacterium brumae is a rapid-growing, non-pathogenic Mycobacterium species, originally isolated from environmental and human samples in Barcelona, Spain. Mycobacterium brumae is not pathogenic and it's in vitro phenotype and immunogenic properties have been well characterized. However, the knowledge of its underlying genetic composition is still incomplete. In this study, we first describe the 4 Mb genome of the M. brumae type strain ATCC 51384T assembling PacBio reads, and second, we assess the low intraspecies variability by comparing the type strain with Illumina reads from three additional strains. Mycobacterium brumae genome is composed of a circular chromosome with a high GC content of 69.2% and containing 3,791 CDSs, 97 pseudogenes, one prophage and no CRISPR loci. Mycobacterium brumae has shown no pathogenic potential in in vivo experiments, and our genomic analysis confirms its phylogenetic position with other non-pathogenic and rapid growing mycobacteria. Accordingly, we determined the absence of virulence-related genes, such as ESX-1 locus and most PE/PPE genes, among others. Although the immunogenic potential of M. brumae was proved to be as high as Mycobacterium bovis BCG, the only mycobacteria licensed to treat cancer, the genomic content of M. tuberculosis T cell and B cell antigens in M. brumae genome is considerably lower than those antigens present in M. bovis BCG genome. Overall, this work provides relevant genomic data on one of the species of the mycobacterial genus with high therapeutic potential.
Collapse
Affiliation(s)
| | - Paula Herrero-Abadía
- Genetics and Microbiology Department, Faculty of Biosciences, Autonomous University of Barcelona, Barcelona, Spain
| | | | - Vicente Sentandreu
- Genomics Unit, Central Service for Experimental Research (SCSIE), University of Valencia, Burjassot, Spain
| | - Eduard Torrents
- Bacterial Infections and Antimicrobial Therapies Group, Institute for Bioengineering of Catalonia (IBEC), Barcelona, Spain
- Microbiology Section, Department of Genetics, Microbiology, and Statistics, Biology Faculty, Universitat de Barcelona, Barcelona, Spain
| | | | | | - Iñaki Comas
- Instituto de Biomedicina de Valencia (IBV), CSIC, Valencia, Spain
| | - Esther Julián
- Genetics and Microbiology Department, Faculty of Biosciences, Autonomous University of Barcelona, Barcelona, Spain
| | - Mireia Coscolla
- I2SysBio, University of Valencia-FISABIO Joint Unit, Paterna, Spain
| |
Collapse
|
40
|
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: 1.0] [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
|
41
|
Kim KJ, Oh SH, Jeon D, Chang CL. Isolation and Antimicrobial Susceptibility of Nontuberculous Mycobacteria in a Tertiary Hospital in Korea, 2016 to 2020. Tuberc Respir Dis (Seoul) 2023; 86:47-56. [PMID: 36480820 PMCID: PMC9816495 DOI: 10.4046/trd.2022.0115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 12/08/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND There is a global increase in isolation of nontuberculous mycobacteria (NTM). The aim of the study was to analyze longitudinal trends of NTM identification and pattern of antimicrobial susceptibility testing. METHODS NTM recovery rates, distribution of NTM species identification, and antimicrobial susceptibility pattern of NTM at Pusan National University Yangsan Hospital between January 2016 and December 2020 were retrospectively analyzed. RESULTS A total of 52,456 specimens from 21,264 patients were submitted for mycobacterial culture, of which 2,521 from 1,410 patients were NTM positive over five years (January 2016 to December 2020). NTM isolation showed an increasing trend from 2016 to 2020 (p<0.001, test for trend) mainly caused by Mycobacterium avium complex. The vast majority of M. avium complex were susceptible to key agents clarithromycin and amikacin. For Mycobacterium kansasii, resistance to rifampin and clarithromycin is rare. Amikacin was the most effective drug against Mycobacterium abscessus subspecies abscessus and Mycobacterium subspecies massiliense. Most of M. subspecies massiliense were susceptible to clarithromycin, while the majority of M. abscessus subspecies abscessus were resistant to clarithromycin (p<0.001). CONCLUSION There was an increasing trend of NTM isolation in our hospital. Resistance to key drugs was uncommon for most NTM species except for M. abscessus subspecies abscessus against clarithromycin.
Collapse
Affiliation(s)
- Keun Ju Kim
- Department of Laboratory Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea,Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| | - Seung-Hwan Oh
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea,Department of Laboratory Medicine, Pusan National University School of Medicine, Yangsan, Republic of Korea,Medical Research Institute, Pusan National University, Yangsan, Republic of Korea
| | - Doosoo Jeon
- Department of Internal Medicine, Pusan National University Yangsan Hospital, Pusan National University School of Medicin, Yangsan, Republic of Korea
| | - Chulhun L. Chang
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea,Department of Laboratory Medicine, Pusan National University School of Medicine, Yangsan, Republic of Korea,Medical Research Institute, Pusan National University, Yangsan, Republic of Korea,Address for correspondence Chulhun L. Chang, M.D., Ph.D. Department of Laboratory Medicine, Pusan National University Yangsan Hospital, 20 Geumo-ro, Mulgeum-eup, Yangsan 50612, Republic of Korea Phone 82-55-360-1877 Fax 82-55-360-1880 E-mail
| |
Collapse
|
42
|
Gupta R, Rohde KH. Implementation of a mycobacterial CRISPRi platform in Mycobacterium abscessus and demonstration of the essentiality of ftsZ Mab. Tuberculosis (Edinb) 2023; 138:102292. [PMID: 36495774 PMCID: PMC11552333 DOI: 10.1016/j.tube.2022.102292] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 11/22/2022] [Accepted: 11/27/2022] [Indexed: 12/04/2022]
Abstract
Mycobacterium abscessus (Mab) is a highly drug-resistant non-tuberculous mycobacterial species that causes debilitating TB-like pulmonary infections. The lack of genetic tools has hampered characterization of its extensive repertoire of virulence factors, antimicrobial resistance mechanisms, and drug targets. In this study, we evaluated the performance of a mycobacterial single plasmid CRISPRi-dCas9 system optimized for M. tuberculosis and M. smegmatis for inducible gene silencing in Mab. The efficacy of CRISPRi-mediated repression of two antibiotic resistance genes (blaMab, whiB7Mab) and two putative essential genes (ftsZMab,topAMab) was determined by measuring mRNA transcript levels and phenotypic outcomes. While our results support the utility of this mycobacterial CRISPRi dCas9Sth1 single-plasmid platform for inducible silencing of specific target genes in Mab, they also highlighted several caveats and nuances that may warrant species-specific optimization for Mab. We observed overall lower levels of gene repression in Mab including variable silencing of different target genes despite use of PAMs of similar predicted strength. In addition, leaky gene repression in the absence of inducer was noted for some genes but not others. Nonetheless, using CRISPRi we demonstrated the silencing of multiple target genes and validated ftsZMab as an essential gene and promising drug target for the first time.
Collapse
Affiliation(s)
- Rashmi Gupta
- Division of Immunity and Pathogenesis, College of Medicine, Burnett School of Biomedical Sciences, University of Central Florida, 6900 Lake Nona Blvd, FL, 32827, USA.
| | - Kyle H Rohde
- Division of Immunity and Pathogenesis, College of Medicine, Burnett School of Biomedical Sciences, University of Central Florida, 6900 Lake Nona Blvd, FL, 32827, USA.
| |
Collapse
|
43
|
Rais M, Abdelaal H, Reese VA, Ferede D, Larsen SE, Pecor T, Erasmus JH, Archer J, Khandhar AP, Cooper SK, Podell BK, Reed SG, Coler RN, Baldwin SL. Immunogenicity and protection against Mycobacterium avium with a heterologous RNA prime and protein boost vaccine regimen. Tuberculosis (Edinb) 2023; 138:102302. [PMID: 36586154 PMCID: PMC10361416 DOI: 10.1016/j.tube.2022.102302] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 12/16/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022]
Abstract
Prophylactic efficacy of two different delivery platforms for vaccination against Mycobacterium avium (M. avium) were tested in this study; a subunit and an RNA-based vaccine. The vaccine antigen, ID91, includes four mycobacterial antigens: Rv3619, Rv2389, Rv3478, and Rv1886. We have shown that ID91+GLA-SE is effective against a clinical NTM isolate, M. avium 2-151 smt. Here, we extend these results and show that a heterologous prime/boost strategy with a repRNA-ID91 (replicon RNA) followed by protein ID91+GLA-SE boost is superior to the subunit protein vaccine given as a homologous prime/boost regimen. The repRNA-ID91/ID91+GLA-SE heterologous regimen elicited a higher polyfunctional CD4+ TH1 immune response when compared to the homologous protein prime/boost regimen. More significantly, among all the vaccine regimens tested only repRNA-ID91/ID91+GLA-SE induced IFN-γ and TNF-secreting CD8+ T cells. Furthermore, the repRNA-ID91/ID91+GLA-SE vaccine strategy elicited high systemic proinflammatory cytokine responses and induced strong ID91 and an Ag85B-specific humoral antibody response a pre- and post-challenge with M. avium 2-151 smt. Finally, while all prophylactic prime/boost vaccine regimens elicited a degree of protection in beige mice, the heterologous repRNA-ID91/ID91+GLA-SE vaccine regimen provided greater pulmonary protection than the homologous protein prime/boost regimen. These data indicate that a prophylactic heterologous repRNA-ID91/ID91+GLA-SE vaccine regimen augments immunogenicity and confers protection against M. avium.
Collapse
Affiliation(s)
- Maham Rais
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, 98145, USA
| | - Hazem Abdelaal
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, 98145, USA
| | - Valerie A Reese
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, 98145, USA
| | - Debora Ferede
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, 98145, USA
| | - Sasha E Larsen
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, 98145, USA
| | - Tiffany Pecor
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, 98145, USA
| | | | | | | | - Sarah K Cooper
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, 98145, USA
| | - Brendan K Podell
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, 98145, USA
| | | | - Rhea N Coler
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, 98145, USA; Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, 98195, USA; Department of Global Health, University of Washington, Seattle, WA, 98195, USA
| | - Susan L Baldwin
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, 98145, USA.
| |
Collapse
|
44
|
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: 12] [Impact Index Per Article: 6.0] [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
|
45
|
Phage Therapy for Nontuberculous Mycobacteria: Challenges and Opportunities. Pulm Ther 2022; 9:91-107. [PMID: 36583829 PMCID: PMC9931961 DOI: 10.1007/s41030-022-00210-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 11/24/2022] [Indexed: 12/31/2022] Open
Abstract
Non-tuberculous mycobacterium (NTM) infections are often clinically challenging, with lengthy antibiotic regimens that fail to resolve the infections with few good outcomes remaining. Mycobacteriophages-viruses that infect Mycobacterium hosts-show promise as therapeutic agents for NTM infections and have been used in 20 compassionate use cases. Favorable outcomes were observed in many but not all cases, although the phages show exceptional safety profiles and no evidence of phage resistance was observed, even when only a single phage was administered. Phage-specific antibodies are commonly present following intravenous administration and are often neutralizing for the phage in vitro. However, phage neutralization does not consistently correlate with poor treatment outcomes and may not be a therapeutic limitation in all patients, even when immunocompetent. Currently, the therapeutic potential of phages is substantially limited by the great variation in phage susceptibility and a relatively small repertoire of therapeutically useful phages. As many as 45% of clinical isolates can have a smooth colony morphotype, and phages that both efficiently infect and kill these strains have yet to be described. In contrast, ~ 75% of rough strains are susceptible to and killed by one or more phages and therapeutic options can be considered on a compassionate use basis. Although therapies must currently be personalized, elucidating the determinants of phage host specificity, expanding the useful phage repertoire, and identifying the key determinants of clinical outcomes will reveal their full therapeutic potential.
Collapse
|
46
|
Liu Y, Ma X, Chen J, Wang H, Yu Z. Nontuberculous mycobacteria by metagenomic next-generation sequencing: Three cases reports and literature review. Front Public Health 2022; 10:972280. [PMID: 36452947 PMCID: PMC9702513 DOI: 10.3389/fpubh.2022.972280] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 10/18/2022] [Indexed: 11/16/2022] Open
Abstract
Background The increasing worldwide incidence of nontuberculous mycobacterial lung disease (NTM-LD) and the similarity of its manifestations to those of tuberculosis (TB) pose huge challenges in the diagnosis and treatment of NTM-LD, which is commonly misdiagnosed and mistreated as TB. Proper diagnosis and treatment at an early stage can greatly improve patient outcomes. Case presentation Mycobacterium avium was identified by mNGS in lung tissue of case 1 and bronchioalveolar fluid from case 2 that was not identified using conventional microbiological methods. Multiple NTM species were detected in the blood mNGS samples from case 3 who had disseminated NTM infection. Although NTM was isolated from blood culture, conventional methods failed to identify the organisms to the level of species. All three patients were suffering from and being treated for myelodysplastic syndrome, rheumatoid arthritis, systemic lupus erythematosus, or acute lymphoblastic leukemia, making them immunosuppressed and susceptible to NTM infections. Case 1 and Case 2 significantly improved after anti-NTM treatment, but case 3 succumbed to the infection due to her underlying medical illness despite aggressive treatment. Conclusions The cases in this study demonstrate the effectiveness of mNGS in facilitating and improving the clinical diagnosis of NTM infections. We propose combining mNGS with traditional diagnostic methods to identify pathogens at the early stages of the disease so that targeted treatment can be implemented.
Collapse
Affiliation(s)
- Ying Liu
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaoxu Ma
- Department of Respiratory Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jiajun Chen
- School of Public Health, Zhengzhou University, Zhengzhou, China
| | - Huifen Wang
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zujiang Yu
- Gene Hospital of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China,*Correspondence: Zujiang Yu
| |
Collapse
|
47
|
Alcaraz M, Roquet-Banères F, Leon-Icaza SA, Abendroth J, Boudehen YM, Cougoule C, Edwards TE, Kremer L. Efficacy and Mode of Action of a Direct Inhibitor of Mycobacterium abscessus InhA. ACS Infect Dis 2022; 8:2171-2186. [PMID: 36107992 DOI: 10.1021/acsinfecdis.2c00314] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
There is an unmet medical need for effective treatments against Mycobacterium abscessus pulmonary infections, to which cystic fibrosis (CF) patients are particularly vulnerable. Recent studies showed that the antitubercular drug isoniazid is inactive against M. abscessus due to the incapacity of the catalase-peroxidase to convert the pro-drug into a reactive metabolite that inhibits the enoyl-ACP reductase InhA. To validate InhAMAB as a druggable target in M. abscessus, we assayed the activity of NITD-916, a 4-hydroxy-2-pyridone lead candidate initially described as a direct inhibitor of InhA that bypasses KatG bioactivation in Mycobacterium tuberculosis. The compound displayed low MIC values against rough and smooth clinical isolates in vitro and significantly reduced the bacterial burden inside human macrophages. Moreover, treatment with NITD-916 reduced the number and size of intracellular mycobacterial cords, regarded as markers of the severity of the infection. Importantly, NITD-916 significantly lowered the M. abscessus burden in CF-derived lung airway organoids. From a mechanistic perspective, NITD-916 abrogated de novo synthesis of mycolic acids and NITD-916-resistant spontaneous mutants harbored point mutations in InhAMAB at residue 96. That NITD-916 targets InhAMAB directly without activation requirements was confirmed genetically and by resolving the crystal structure of the protein in complex with NADH and NITD-916. These findings collectively indicate that InhAMAB is an attractive target to be exploited for future chemotherapeutic developments against this difficult-to-treat mycobacterium and highlight the potential of NITD-916 derivatives for further evaluation in preclinical settings.
Collapse
Affiliation(s)
- Matthéo Alcaraz
- Centre National de la Recherche Scientifique UMR 9004, Institut de Recherche en Infectiologie de Montpellier (IRIM), Université de Montpellier, 1919 route de Mende, 34293 Montpellier, France
| | - Françoise Roquet-Banères
- Centre National de la Recherche Scientifique UMR 9004, Institut de Recherche en Infectiologie de Montpellier (IRIM), Université de Montpellier, 1919 route de Mende, 34293 Montpellier, France
| | - Stephen Adonai Leon-Icaza
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, 31400 Toulouse, France
| | - Jan Abendroth
- UCB BioSciences, Bainbridge Island, Washington 98109, United States.,Seattle Structural Genomics Center for Infectious Disease (SSGCID), Seattle, Washington 98109, United States
| | - Yves-Marie Boudehen
- Centre National de la Recherche Scientifique UMR 9004, Institut de Recherche en Infectiologie de Montpellier (IRIM), Université de Montpellier, 1919 route de Mende, 34293 Montpellier, France
| | - Céline Cougoule
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, 31400 Toulouse, France
| | - Thomas E Edwards
- UCB BioSciences, Bainbridge Island, Washington 98109, United States.,Seattle Structural Genomics Center for Infectious Disease (SSGCID), Seattle, Washington 98109, United States
| | - Laurent Kremer
- Centre National de la Recherche Scientifique UMR 9004, Institut de Recherche en Infectiologie de Montpellier (IRIM), Université de Montpellier, 1919 route de Mende, 34293 Montpellier, France.,INSERM, IRIM, 34293 Montpellier, France
| |
Collapse
|
48
|
Musser E, Smith C, Halse TA, Kohlerschmidt D, Rourke A, Fiero A, Musser KA, Escuyer V, Lapierre P. Characterization of Mycobacterium salfingeri sp. nov.: A novel nontuberculous mycobacteria isolated from a human wound infection. Front Microbiol 2022; 13:992610. [PMID: 36299734 PMCID: PMC9589434 DOI: 10.3389/fmicb.2022.992610] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 09/21/2022] [Indexed: 11/25/2022] Open
Abstract
Nontuberculous mycobacteria (NTM) are environmental bacteria commonly found in soil and water in almost every part of the world. While usually non-pathogenic, they can cause acute respiratory and cutaneous infections under certain circumstances or in patients with underlying medical conditions. Contrary to members of the Mycobacterium tuberculosis complex, documented human-to-human transmissions of NTM have been rarely reported and most cases result from direct environmental exposure. Here we describe the identification of a new NTM species isolated from a hand laceration of a New York State patient after a fall. This new NTM forms rough, orange pigmented colonies and is naturally resistant to doxycycline and tobramycin. Whole genome analysis reveal no close relatives present in public databases, and our findings are in accordance with the recognition of a new taxonomic species of NTM. We propose the name Mycobacterium salfingeri sp. nov. for this new NTM representative. The type strain is 20-157661T (DSM = 113368T, BCCM = ITM 501207T).
Collapse
|
49
|
Drevinek P, Canton R, Johansen HK, Hoffman L, Coenye T, Burgel PR, Davies JC. New concepts in antimicrobial resistance in cystic fibrosis respiratory infections. J Cyst Fibros 2022; 21:937-945. [DOI: 10.1016/j.jcf.2022.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 10/09/2022] [Accepted: 10/12/2022] [Indexed: 11/22/2022]
|
50
|
In Vitro Susceptibility Testing of Eravacycline against Nontuberculous Mycobacteria. Antimicrob Agents Chemother 2022; 66:e0068922. [PMID: 35943269 PMCID: PMC9487454 DOI: 10.1128/aac.00689-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Nontuberculous mycobacteria (NTM) infections are increasing worldwide. Mycobacterium avium complex (MAC) and the M. abscessus species are the most commonly cultured NTM and treatment options are limited, especially for the M. abscessus species. In this study, the in vitro activity of eravacycline, a new tetracycline derivative, was tested against 110 clinical isolates of NTM. MIC testing was performed as recommended by the Clinical and Laboratory Standards Institute against 60 isolates of rapidly growing mycobacteria (RGM), of which ~70% were tetracycline resistant. These included M. abscessus subsp. abscessus (8 isolates), M. abscessus subsp. massiliense (5), M. chelonae (10), M. immunogenum (3), M. fortuitum group (20) including 12 doxycycline-resistant isolates, and M. mucogenicum group (10) including three doxycycline-resistant isolates. Due to trailing, eravacycline MICs were read at 80% and 100% inhibition. Eravacycline was active against all RGM species, with MIC50 ranges of ≤0.015 to 0.5 and ≤0.015 to 0.12 μg/mL for 100% and 80% inhibition, respectively. For M. abscessus subsp. abscessus, MIC50 values were 0.12 and 0.03 μg/mL with 100% and 80% inhibition, respectively. MICs for tigecycline were generally within 1 to 2 dilutions of the 100%-inhibition eravacycline MIC values. Fifty isolates of slowly growing mycobacteria (SGM) species, including 16 isolates of MAC, were also tested. While there was no trailing observed in most SGM, the eravacycline MICs were higher (MIC range of >8 μg/mL), except for M. kansasii and M. marinum which had MIC50 values of 1 μg/mL. This study supports further evaluation of eravacycline, including clinical trials for the development of RGM treatment regimens, especially for M. abscessus.
Collapse
|