1
|
Webber T, Macdonald C, Tameris M, Tredoux N, Bierman A, Gutschmidt A, Tönsing S, Hiemstra A, Noor F, Snyders C, Richardson T, Fransman B, Allwood B, Hatherill M, Kleynhans L, Loxton AG, Walzl G, Chegou N, du Plessis N, Shaw JA, Malherbe ST. Immune, metabolic, anatomical, and functional features of people after successful tuberculosis treatment: an exploratory analysis. Sci Rep 2025; 15:18392. [PMID: 40419526 DOI: 10.1038/s41598-025-01656-1] [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: 02/07/2025] [Accepted: 05/07/2025] [Indexed: 05/28/2025] Open
Abstract
We explored the underlying mechanisms that may drive post-tuberculosis (TB) lung disease, a multifactorial, heterogenous, and prevalent disease. Extensive clinical phenotyping through fluorine-18 Fluorodeoxyglucose (FDG) positron emission tomography (PET)-computed tomography (CT) scans, pulmonary function testing, and symptom and quality of life questionnaires, was performed on a cohort of 48 adults who completed TB treatment within 6 months prior. Immunological characteristics of paired blood- and bronchoalveolar lavage fluid (BALF)-derived immune cells were assessed by multiplex bead-based immunoassay, ELISA and flow cytometry. There was agreement between measures of inflammation on PET, the severity of anatomical abnormalities on CT, and pulmonary function testing. However, of these, only PET was associated with exercise tolerance and symptom scores. Measures of radiologic extent (total glycolytic activity and SUVmax on PET, and segments involved on CT) also correlated with proteins detected in blood that implicate type 1 (IFN-γ, TNFα, IL-12) and type 2 (IL-4, IL-33) responses, ongoing remodelling of lung tissue (MMPs), airways and vasculature (VEGF), as well as subsets of activated CD8+ and CD4+ T-cells. The radiologic extent of structural post-TB lung involvement is associated with a range of impaired lung function measures and immunological dysregulation. Our findings suggest that obstructive and restrictive lung pathology due to pulmonary TB do not occur in opposition but rather point towards a mixed pathology in most TB survivors.
Collapse
Affiliation(s)
- Tariq Webber
- South African Medical Research Council Centre for Tuberculosis Research, Biomedical Research Institute, Division of Immunology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Candice Macdonald
- South African Medical Research Council Centre for Tuberculosis Research, Biomedical Research Institute, Division of Immunology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Michele Tameris
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Nicolette Tredoux
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Anandi Bierman
- South African Medical Research Council Centre for Tuberculosis Research, Biomedical Research Institute, Division of Immunology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Andrea Gutschmidt
- South African Medical Research Council Centre for Tuberculosis Research, Biomedical Research Institute, Division of Immunology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Susanne Tönsing
- South African Medical Research Council Centre for Tuberculosis Research, Biomedical Research Institute, Division of Immunology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Andriëtte Hiemstra
- South African Medical Research Council Centre for Tuberculosis Research, Biomedical Research Institute, Division of Immunology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Firdows Noor
- South African Medical Research Council Centre for Tuberculosis Research, Biomedical Research Institute, Division of Immunology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Candice Snyders
- South African Medical Research Council Centre for Tuberculosis Research, Biomedical Research Institute, Division of Immunology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Tracy Richardson
- South African Medical Research Council Centre for Tuberculosis Research, Biomedical Research Institute, Division of Immunology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Bernadine Fransman
- South African Medical Research Council Centre for Tuberculosis Research, Biomedical Research Institute, Division of Immunology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Brian Allwood
- Division of Pulmonology, Department of Medicine, Stellenbosch University and Tygerberg Hospital, Cape Town, South Africa
| | - Mark Hatherill
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Leanie Kleynhans
- South African Medical Research Council Centre for Tuberculosis Research, Biomedical Research Institute, Division of Immunology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
- Mater Research Institute, The University of Queensland, Translational Research Institute, Brisbane, QLD, Australia
| | - André G Loxton
- South African Medical Research Council Centre for Tuberculosis Research, Biomedical Research Institute, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Gerhard Walzl
- South African Medical Research Council Centre for Tuberculosis Research, Biomedical Research Institute, Division of Immunology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Novel Chegou
- South African Medical Research Council Centre for Tuberculosis Research, Biomedical Research Institute, Division of Immunology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Nelita du Plessis
- South African Medical Research Council Centre for Tuberculosis Research, Biomedical Research Institute, Division of Immunology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Jane A Shaw
- South African Medical Research Council Centre for Tuberculosis Research, Biomedical Research Institute, Division of Immunology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Stephanus T Malherbe
- South African Medical Research Council Centre for Tuberculosis Research, Biomedical Research Institute, Division of Immunology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa.
| |
Collapse
|
2
|
Li H, Li Z, Cui X, Liu X, Zhao J, Zhang Y, Wang C, Li B, Fan Y, Han J, Xia Y, Xiong Z, Zou X, Zhu Y, Li M, Lu B, Cao B. Mycobacterium tuberculosis Detection in Diverse Clinical Specimens by GeneXpert MTB/RIF: A Large-Scale Retrospective Study. Infect Drug Resist 2025; 18:2401-2413. [PMID: 40357422 PMCID: PMC12068387 DOI: 10.2147/idr.s514220] [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: 01/22/2025] [Accepted: 04/18/2025] [Indexed: 05/15/2025] Open
Abstract
Purpose Mycobacterium tuberculosis (MTB) infection poses a significant global health challenge, with conventional diagnostic methods like acid-fast smear and culture techniques exhibiting limitations in sensitivity and efficiency. This retrospective study examined 4098 clinical samples from China-Japan Friendship Hospital between March 2018 and March 2019, focusing on the diagnostic performance of GeneXpert MTB/RIF across different specimen types. Methods The study encompassed various sample types, including bronchoalveolar lavage fluid (BALF), sputum, lung tissues, hydrothorax, and others. All samples were performed via acid-fast-straining, GeneXpert MTB/RIF and culture. Results GeneXpert MTB/RIF demonstrated superior sensitivity (81.46%) and specificity (98.98%) in respiratory specimens compared to tissues (62.50%) and hydrothorax (46.15%). Notably, in acid-fast-straining-negative samples, GeneXpert MTB/RIF showed sensitivity and specificity of 73.43% and 98.8%, respectively, and reduced false negatives of acid-fast staining. Furthermore, the study explored Cycler Threshold (CT) values, revealing associations with bacterial load and sample types. Conclusion The findings highlight the importance of considering sample types in MTB diagnosis and underscore the potential of GeneXpert MTB/RIF as a valuable diagnostic tool, especially in respiratory specimens, contributing to improved tuberculosis management strategies. Additionally, the study recommended direct GeneXpert MTB/RIF testing of hydrothorax samples instead of acid-fast staining, further enhancing diagnostic accuracy and efficiency.
Collapse
Affiliation(s)
- Haibo Li
- Laboratory of Clinical Microbiology and Infectious Diseases, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, National Center for Respiratory Medicine, New Cornerstone Science Laboratory, China-Japan Friendship Hospital, Beijing, People’s Republic of China
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, People’s Republic of China
| | - Ziyao Li
- Laboratory of Clinical Microbiology and Infectious Diseases, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, National Center for Respiratory Medicine, New Cornerstone Science Laboratory, China-Japan Friendship Hospital, Beijing, People’s Republic of China
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, People’s Republic of China
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
- Changping Laboratory, Beijing, People’s Republic of China
| | - Xiaojing Cui
- Laboratory of Clinical Microbiology and Infectious Diseases, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, National Center for Respiratory Medicine, New Cornerstone Science Laboratory, China-Japan Friendship Hospital, Beijing, People’s Republic of China
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, People’s Republic of China
| | - Xinmeng Liu
- Laboratory of Clinical Microbiology and Infectious Diseases, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, National Center for Respiratory Medicine, New Cornerstone Science Laboratory, China-Japan Friendship Hospital, Beijing, People’s Republic of China
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, People’s Republic of China
- China-Japan Friendship School of Clinical Medicine, Peking University, Beijing, People’s Republic of China
| | - Jiankang Zhao
- Laboratory of Clinical Microbiology and Infectious Diseases, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, National Center for Respiratory Medicine, New Cornerstone Science Laboratory, China-Japan Friendship Hospital, Beijing, People’s Republic of China
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, People’s Republic of China
| | - Yulin Zhang
- Laboratory of Clinical Microbiology and Infectious Diseases, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, National Center for Respiratory Medicine, New Cornerstone Science Laboratory, China-Japan Friendship Hospital, Beijing, People’s Republic of China
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, People’s Republic of China
| | - Chunlei Wang
- Laboratory of Clinical Microbiology and Infectious Diseases, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, National Center for Respiratory Medicine, New Cornerstone Science Laboratory, China-Japan Friendship Hospital, Beijing, People’s Republic of China
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, People’s Republic of China
| | - Binbin Li
- Laboratory of Clinical Microbiology and Infectious Diseases, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, National Center for Respiratory Medicine, New Cornerstone Science Laboratory, China-Japan Friendship Hospital, Beijing, People’s Republic of China
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, People’s Republic of China
| | - Yanyan Fan
- Laboratory of Clinical Microbiology and Infectious Diseases, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, National Center for Respiratory Medicine, New Cornerstone Science Laboratory, China-Japan Friendship Hospital, Beijing, People’s Republic of China
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, People’s Republic of China
| | - Jiajing Han
- Laboratory of Clinical Microbiology and Infectious Diseases, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, National Center for Respiratory Medicine, New Cornerstone Science Laboratory, China-Japan Friendship Hospital, Beijing, People’s Republic of China
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, People’s Republic of China
- China-Japan Friendship School of Clinical Medicine, Peking University, Beijing, People’s Republic of China
| | - Yudi Xia
- Laboratory of Clinical Microbiology and Infectious Diseases, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, National Center for Respiratory Medicine, New Cornerstone Science Laboratory, China-Japan Friendship Hospital, Beijing, People’s Republic of China
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, People’s Republic of China
| | - Zhujia Xiong
- Laboratory of Clinical Microbiology and Infectious Diseases, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, National Center for Respiratory Medicine, New Cornerstone Science Laboratory, China-Japan Friendship Hospital, Beijing, People’s Republic of China
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, People’s Republic of China
| | - Xiaohui Zou
- Laboratory of Clinical Microbiology and Infectious Diseases, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, National Center for Respiratory Medicine, New Cornerstone Science Laboratory, China-Japan Friendship Hospital, Beijing, People’s Republic of China
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, People’s Republic of China
| | - Yue Zhu
- Laboratory of Clinical Microbiology and Infectious Diseases, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, National Center for Respiratory Medicine, New Cornerstone Science Laboratory, China-Japan Friendship Hospital, Beijing, People’s Republic of China
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, People’s Republic of China
| | - Mengxue Li
- Laboratory of Clinical Microbiology and Infectious Diseases, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, National Center for Respiratory Medicine, New Cornerstone Science Laboratory, China-Japan Friendship Hospital, Beijing, People’s Republic of China
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, People’s Republic of China
- China-Japan Friendship School of Clinical Medicine, Peking University, Beijing, People’s Republic of China
| | - Binghuai Lu
- Laboratory of Clinical Microbiology and Infectious Diseases, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, National Center for Respiratory Medicine, New Cornerstone Science Laboratory, China-Japan Friendship Hospital, Beijing, People’s Republic of China
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, People’s Republic of China
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
- China-Japan Friendship School of Clinical Medicine, Peking University, Beijing, People’s Republic of China
| | - Bin Cao
- Laboratory of Clinical Microbiology and Infectious Diseases, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, National Center for Respiratory Medicine, New Cornerstone Science Laboratory, China-Japan Friendship Hospital, Beijing, People’s Republic of China
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, People’s Republic of China
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
- Changping Laboratory, Beijing, People’s Republic of China
- China-Japan Friendship School of Clinical Medicine, Peking University, Beijing, People’s Republic of China
- Tsinghua University-Peking University Joint Center for Life Sciences, Tsinghua University, Beijing, People’s Republic of China
- Department of Respiratory Medicine, Capital Medical University, Beijing, People’s Republic of China
| |
Collapse
|
3
|
Meghji J, Auld SC, Bisson GP, Khosa C, Masekela R, Navuluri N, Rachow A. Post-tuberculosis lung disease: towards prevention, diagnosis, and care. THE LANCET. RESPIRATORY MEDICINE 2025; 13:460-472. [PMID: 40127662 DOI: 10.1016/s2213-2600(24)00429-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 12/08/2024] [Accepted: 12/18/2024] [Indexed: 03/26/2025]
Abstract
There is a growing body of data describing the high burden of respiratory sequelae seen among tuberculosis survivors, including children, adolescents, and adults. This group of sequelae are known as post-tuberculosis lung disease and include parenchymal damage, airway disease, and pulmonary vascular disease. It is thought that approximately half of pulmonary tuberculosis survivors have ongoing structural pathology, lung function impairment, or respiratory symptoms after the resolution of active disease. Post-tuberculosis lung disease has been associated with adverse patient outcomes, including persistent symptoms and functional impairment, ongoing health seeking, and impacts on income and employment. There is still much to understand about the epidemiology and nature of post-tuberculosis lung disease, but in this Review we focus on strategies for prevention, diagnosis, and care to inform the ongoing work of tuberculosis-affected communities, health-care providers, researchers, and policy makers. We summarise recent data, highlight evidence gaps, and suggest key research priorities for those working in the field.
Collapse
Affiliation(s)
- Jamilah Meghji
- National Heart & Lung Institute, Imperial College London, London, UK; Department of Respiratory Medicine, Imperial College Healthcare NHS Trust, London, UK.
| | - Sara C Auld
- Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA; Department of Epidemiology and Department of Global Health, Emory University Rollins School of Public Health, Atlanta, GA, USA
| | - Gregory P Bisson
- Department of Medicine, Division of Infectious Diseases, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Celso Khosa
- Instituto Nacional de Saúde, Marracuene, Mozambique; Department of Physiological Science, Clinical Pharmacology, Faculty of Medicine, Eduardo Mondlane University, Maputo, Mozambique
| | - Refiloe Masekela
- Department of Paediatrics and Child Health, College of Health Sciences, School of Clinical Medicine, University of KwaZulu Natal, Durban, South Africa; Africa Health Research Institute, Durban, South Africa
| | - Neelima Navuluri
- Department of Medicine, Division of Pulmonary and Critical Care, Duke University School of Medicine, Durham, NC, USA; Duke Global Health Institute, Duke University, Durham, NC, USA
| | - Andrea Rachow
- Institute of Infectious Diseases and Tropical Medicine, LMU University Hospital, LMU Munich, Germany; German Centre for Infection Research (DZIF), Munich, Germany; Unit of Global Health, Helmholtz Centre Munich, German Research Centre for Environmental Health (HMGU), Neuherberg, Germany
| |
Collapse
|
4
|
Xie YL, Modi N, Lopez K, Reiss R, Robledo J, Eichberg C, Hapeela N, Nakabugo E, Anyango I, Arora K, Odero R, Van Heerden J, Zemanay W, Kaipilyawar VS, Kennedy S, Banada P, Nakiyingi L, Joloba ML, Centner C, McCarthy K, Ellner J, Salgame P, Alland D, Dorman SE. Prominence of Mycobacterium tuberculosis biomarkers among sputum culture-negative clinic attendees, independent of Ultra status. J Infect Public Health 2025; 18:102791. [PMID: 40315556 DOI: 10.1016/j.jiph.2025.102791] [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: 08/21/2024] [Revised: 04/02/2025] [Accepted: 04/21/2025] [Indexed: 05/04/2025] Open
Abstract
BACKGROUND Highly-sensitive molecular tests like GeneXpert MTB/RIF Ultra improve detection of paucibacillary pulmonary tuberculosis (TB) but occasionally detect Mycobacterium tuberculosis (Mtb) DNA in sputum from culture-negative individuals, with unclear significance. We hypothesized that Ultra may be detecting culture-negative TB, and manifest in a higher prevalence of TB biomarkers compared to Ultra-negative/culture-negative ('sputum-negative') individuals. METHODS From 1200 symptomatic African adults undergoing evaluation for TB, we identified 66 with discordant results (Ultra-positive, culture-negative), and matched 52 sputum-negative (Ultra-negative, culture-negative) and 30 sputum-positive (Ultra-positive, culture-positive) participants. Over 12 months, participants were assessed for Mtb biomarkers (Mtb growth in augmented or follow-up sputum cultures, Mtb mRNA in baseline sputum, and symptomatic Ultra-positive after baseline) and TB-associated host transcriptional signatures. RESULTS At baseline, TB-associated biomarker(s) were detected in 51.5 % of sputum-discordant versus 59.6 % of sputum-negative participants (p = 0.46), with at least one Mtb biomarker in 16.7 % versus 26.9 % respectively (p = 0.26). Longitudinally, 26.5 % of untreated sputum-discordant versus 41.7 % of untreated sputum-negative participants had Mtb biomarkers (p = 0.17) despite most reporting symptom improvement. Notably, 30 % of untreated sputum-negative participants converted to Ultra-positive at month 2. One sputum-discordant and one sputum-negative participant developed culture-confirmed TB at follow-up. CONCLUSION TB bacterial and host biomarkers were prevalent and no different between sputum-discordant and sputum-negative participants, raising concern for a considerable population of undiagnosed culture-negative TB. These findings parallel new evidence of Mtb aerosolization from sputum-negative individuals and highlight a need for more comprehensive diagnostics that detect sputum culture-negative TB with respect to infectiousness, pathology, and risk of progression.
Collapse
Affiliation(s)
- Yingda L Xie
- Department of Medicine and Public Health Research Institute, Rutgers New Jersey Medical School, Newark, NJ, United States.
| | - Nisha Modi
- Department of Medicine and Public Health Research Institute, Rutgers New Jersey Medical School, Newark, NJ, United States
| | - Kattya Lopez
- Department of Medicine and Public Health Research Institute, Rutgers New Jersey Medical School, Newark, NJ, United States
| | - Robert Reiss
- Department of Medicine and Public Health Research Institute, Rutgers New Jersey Medical School, Newark, NJ, United States
| | - Jorge Robledo
- Department of Medicine and Public Health Research Institute, Rutgers New Jersey Medical School, Newark, NJ, United States
| | | | - Nchimunya Hapeela
- Division of Medical Microbiology, University of Cape Town Faculty of Health Sciences, Cape Town, South Africa
| | | | - Irene Anyango
- Kenya Medical Research Institute, Center for Global Health Research, Kisumu, Kenya
| | - Kiranjot Arora
- Department of Medicine and Public Health Research Institute, Rutgers New Jersey Medical School, Newark, NJ, United States
| | - Ronald Odero
- Kenya Medical Research Institute, Center for Global Health Research, Kisumu, Kenya
| | - Judi Van Heerden
- Division of Medical Microbiology, University of Cape Town Faculty of Health Sciences, Cape Town, South Africa
| | - Widaad Zemanay
- Division of Medical Microbiology, University of Cape Town Faculty of Health Sciences, Cape Town, South Africa
| | - Vaishnavi S Kaipilyawar
- Department of Medicine and Public Health Research Institute, Rutgers New Jersey Medical School, Newark, NJ, United States
| | - Samuel Kennedy
- Medical University of South Carolina, Charleston, SC, United States
| | - Padmapriya Banada
- Department of Medicine and Public Health Research Institute, Rutgers New Jersey Medical School, Newark, NJ, United States
| | - Lydia Nakiyingi
- Infectious Diseases Institute, Makerere University, Kampala, Uganda
| | - Moses L Joloba
- Infectious Diseases Institute, Makerere University, Kampala, Uganda
| | - Chad Centner
- Division of Medical Microbiology, University of Cape Town Faculty of Health Sciences, Cape Town, South Africa
| | | | - Jerrold Ellner
- Department of Medicine and Public Health Research Institute, Rutgers New Jersey Medical School, Newark, NJ, United States
| | - Padmini Salgame
- Department of Medicine and Public Health Research Institute, Rutgers New Jersey Medical School, Newark, NJ, United States
| | - David Alland
- Department of Medicine and Public Health Research Institute, Rutgers New Jersey Medical School, Newark, NJ, United States
| | - Susan E Dorman
- Medical University of South Carolina, Charleston, SC, United States
| |
Collapse
|
5
|
Ssengooba W, Musisi E, Semugenze D, Komakech K, Ndema M, Sekaggya CW, Adakun S, Sloan DJ, Katamba A, Lamorde M, Joloba M, Sabiiti W. Performance evaluation of alternative bacteriological measures of response to MDR-TB therapy during the initial 16 weeks of treatment. RESEARCH SQUARE 2025:rs.3.rs-5834681. [PMID: 40297689 PMCID: PMC12036452 DOI: 10.21203/rs.3.rs-5834681/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Background Monitoring response to Multi-Drug-Resistant Tuberculosis (MDR-TB) treatment is burdensome to TB programmes and may benefit from alternative effective tools. We evaluated the concordance of alternative bacteriological measures of response to therapy (AMRT) during the initial sixteen weeks of MDR-TB treatment. Methods In a prospective study of MDR/RR-TB among smear positive adults, aged 18 year and above. Pooled early morning- and spot sputa were obtained before treatment initiation (95% on Bdq, Lzd, Lfx, Cfz, Cs regimen) and at weeks 2, 4, 6, 8, 12, and 16 during treatment between 14/02/2020 and 09/02/2024. Samples were tested using Concentrated Fluorescent Microscopy (CFM), Fluorescein-diacetate (FDA)-Acid Fast Bacilli (AFB) vital smear microscopy, the tuberculosis-Molecular bacterial load assay (TB-MBLA), and Middle brook 7H11 selective (MB7H11S) colony-forming units as the AMRT. Concordance of the AMRT for sputum conversion was compared to Mycobacterial Growth Indicator Tube (MGIT) culture conversion at weeks 12 and 16 of treatment. Results A total of 101 MDR/RR-TB patients were screened of which 42 were smear negative. Fifty-nine participants were enrolled, of whom 58 (98%) provided baseline sputa and these were included in the analysis. The concordance, n/N (%) of each AMRT test with MGIT culture conversion at week 12 were: 31/35(88.6%) for CFM, 32/33 (97.0%) for FDA, and 25/26 (96.2%) for TB-MBLA, and 11/11 (100%) for MB7H11S. At week 16, concordance of eachAMRT were: 39/40 (97.5%) for CFM, 35/36 (97.2%) for FDA, 32/32 (100%) for TB-MBLA, and 15/15 (100%) for MB7H11S. Among people living with HIV,the concordances of AMRT with MGIT culture conversion varied at week 8 but was 100% for all tests at weeks 12 and 16. Baseline clinical and/or bacteriological factors did not influence the concordance of AMRT to MGIT culture conversion at weeks 12, and 16. Conclusion Our data show that concentrated Fluorescent smear, Fluorescein-di-acetate smear microscopy, and TB-MBLA are suitable alternative measures of response to TB therapy compared to MGIT culture among MDR-TB participants. Use of these alternative rapid methods may allow timely decision making as well as rapid evaluation of alternative MDR-TB treatment regimens.
Collapse
Affiliation(s)
- Willy Ssengooba
- Department of Medical Microbiology, and Makerere University Biomedical Research Center (MAKBRC), College of Health Sciences Makerere University
| | - Emmanuel Musisi
- Division of Infection and Global Health, School of Medicine, University of St Andrews, KY16 9TF St Andrews
| | - Derrick Semugenze
- Department of Medical Microbiology, and Makerere University Biomedical Research Center (MAKBRC), College of Health Sciences Makerere University
| | - Kevin Komakech
- Department of Medical Microbiology, and Makerere University Biomedical Research Center (MAKBRC), College of Health Sciences Makerere University
| | - Moses Ndema
- Adult Tuberculosis Unit, Mulago National Referral and Teaching Hospital
| | | | - Susan Adakun
- Adult Tuberculosis Unit, Mulago National Referral and Teaching Hospital
| | - Derek J Sloan
- Division of Infection and Global Health, School of Medicine, University of St Andrews, KY16 9TF St Andrews
| | | | - Mohammed Lamorde
- Infectious Diseases Institute, Makerere University College of Health Sciences
| | - Moses Joloba
- Department of Medical Microbiology, and Makerere University Biomedical Research Center (MAKBRC), College of Health Sciences Makerere University
| | - Wilber Sabiiti
- Division of Infection and Global Health, School of Medicine, University of St Andrews, KY16 9TF St Andrews
| |
Collapse
|
6
|
Maiello P, Diedrich C, Rutledge T, Rodgers M, Kracinovsky K, Borish HJ, White A, Hopkins F, Chao MC, Klein E, Fortune S, Flynn JL, Lin PL. Characterizing PET CT patterns and bacterial dissemination features of tuberculosis relapse in the macaque model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.31.646419. [PMID: 40236101 PMCID: PMC11996433 DOI: 10.1101/2025.03.31.646419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Tuberculosis (TB) relapse after appropriate drug treatment is poorly understood but critical to developing shorter treatment regimens. Using a cynomolgus macaque model of human TB, macaques with active TB disease were treated with a short course of isoniazid and rifampin and subsequently infected with SIV. Serial clinical, microbiologic, immunologic and position emission and computed tomography (PET CT) assessments were performed to identify risk factors of relapse. Of the 12 animals, eight developed radiologically defined relapse including four that had clinical and/or microbiologic signs. Greater gross pathology and bacterial burden were observed in relapse animals. PET CT characteristics before, during, and at the end of treatment were similar amongst relapse and non-relapse animals. We show that complete sterilization or very low Mtb burden is protective against SIV-induced TB relapse but cannot be predicted by PET CT. Using bar-coded M. tuberculosis , we found that Mtb dissemination during relapse originated from both lung and thoracic lymph nodes, underscoring the importance of lymph nodes as a reservoir. By matching bar-coded Mtb and serial PET CT, we also demonstrate that not every site of persistent Mtb growth after drug treatment is capable of dissemination and relapse, underscoring the complex nature of drug treatment and relapse.
Collapse
|
7
|
Modi NH, Dunkley ORS, Bell AG, Hennig E, Wats A, Huang Y, Daivaa N, Myhrvold C, Xie YL, Banada P. Simplified Co-extraction of total Nucleic Acids from Respiratory Samples for detection of Mycobacterium tuberculosis and SARS-CoV-2 optimized for compatibility across Diagnostic Platforms. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.02.27.25322880. [PMID: 40093238 PMCID: PMC11908299 DOI: 10.1101/2025.02.27.25322880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Tuberculosis (TB) and COVID-19 are leading infectious diseases with high mortality, caused by Mycobacterium tuberculosis (Mtb) and SARS-CoV-2 (SC2), respectively. Co-infection is common but is often undiagnosed as it is challenging to process both pathogens from a single sample. In this study, we present a simple and efficient method for co-extracting nucleic acids (NA) from these two distinct respiratory pathogens for downstream diagnostic testing. We evaluated three different nucleic acid amplification (NAA)-based platforms, LightCycler480 (LC480) qPCR, Qiacuity digital PCR (dPCR), and Cytation3 for CRISPR-Cas13a-based SHINE-TB/SC2 detection assays. Chelex-100 chelating resin-based boiling preparation method was optimized for Mtb NA extraction from saliva and sputum. Saliva showed compatibility with all three platforms, with sensitivity as low as 100 CFU/ml (or 2 genomic copies/μl). This method worked well for sputum using dPCR at 100% (21/21) positivity, though the CRISPR-based SHINE-TB assay showed more variability and sensitivity to sputum inhibitor carry-over, resulting in an 81% positive rate (17/21). Diluting sputum with TE buffer (1:1) improved the detection (2/4). Extraction efficiency of our method was 48%, 62.2%, 86.4% and 99.3% for concentrations 105, 104, 103 and 10 CFU/ml, respectively. The dynamic range for Mtb spiked in pooled sputum showed 100% detection (N=8) at ≥103 CFU/ml with all three methods. Dual-pathogen co-extraction and detection of SC2 (105 PFU/ml) and Mtb (105 CFU/ml) in salivary sputum was successful using CRISPR-Cas13a assays. We have developed a rapid and efficient co-extraction method for multi-pathogen testing across diagnostic platforms and believe this is the first protocol optimized to co-extract Mtb and SARS-CoV-2 from a single sample.
Collapse
Affiliation(s)
- Nisha H Modi
- Department of Medicine, International Center for Public Health, Rutgers University, Newark, NJ, 07103
| | - Owen R S Dunkley
- Department of Molecular Biology, Princeton University, Princeton, NJ, 08544
| | - Alexandra G Bell
- Department of Molecular Biology, Princeton University, Princeton, NJ, 08544
| | - Emily Hennig
- Department of Medicine, International Center for Public Health, Rutgers University, Newark, NJ, 07103
| | - Aanchal Wats
- Department of Medicine, International Center for Public Health, Rutgers University, Newark, NJ, 07103
| | - Yujia Huang
- Department of Molecular Biology, Princeton University, Princeton, NJ, 08544
| | - Naranjargal Daivaa
- Department of Medicine, International Center for Public Health, Rutgers University, Newark, NJ, 07103
| | - Cameron Myhrvold
- Department of Molecular Biology, Princeton University, Princeton, NJ, 08544
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ 08544
- Omenn-Darling Bioengineering Institute, Princeton University, Princeton, NJ 08544
- Department of Chemistry, Princeton University, Princeton, NJ 08544
| | - Yingda L Xie
- Department of Medicine, International Center for Public Health, Rutgers University, Newark, NJ, 07103
| | - Padmapriya Banada
- Department of Medicine, International Center for Public Health, Rutgers University, Newark, NJ, 07103
| |
Collapse
|
8
|
Lange C, Mandalakas AM, Lillebaek T, Chesov D, Dheda K, Saluzzo F. Revisiting diagnostics: high priority tuberculosis diagnostic tests that fill an unmet need: what we need and what we do not need. Clin Microbiol Infect 2025; 31:312-314. [PMID: 39732394 DOI: 10.1016/j.cmi.2024.12.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 12/12/2024] [Accepted: 12/18/2024] [Indexed: 12/30/2024]
Affiliation(s)
- Christoph Lange
- Division of Clinical Infectious Diseases, Research Center Borstel, Borstel, Germany; German Center for Infection Research (DZIF), TTU-TB, Borstel, Germany; Respiratory Medicine & International Health, University of Lübeck, Germany; Department of Pediatrics, Division of Global and Immigrant Health, Baylor College of Medicine and Texas Children's Hospital, Houston, TX, USA.
| | - Anna Maria Mandalakas
- Division of Clinical Infectious Diseases, Research Center Borstel, Borstel, Germany; German Center for Infection Research (DZIF), TTU-TB, Borstel, Germany; Department of Pediatrics, Division of Global and Immigrant Health, Baylor College of Medicine and Texas Children's Hospital, Houston, TX, USA
| | - Troels Lillebaek
- International Reference Laboratory of Mycobacteriology, Statens Serum Institut, Copenhagen, Denmark; Global Health Section, Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | - Dumitru Chesov
- Division of Clinical Infectious Diseases, Research Center Borstel, Borstel, Germany; German Center for Infection Research (DZIF), TTU-TB, Borstel, Germany; Discipline of Pneumology and Allergology, Nicolae Testemitanu State University of Medicine and Pharmacy, Chisinau, Republic of Moldova
| | - Keertan Dheda
- Centre for Lung Infection and Immunity, Division of Pulmonology, Department of Medicine and UCT Lung Institute & South African MRC/UCT Centre for the Study of Antimicrobial Resistance, University of Cape Town, Cape Town, South Africa; Faculty of Infectious and Tropical Diseases, Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, UK
| | - Francesca Saluzzo
- Department of Immunology, Transplantation and Infectious Diseases, Vita Salute San Raffaele University, Milan, Italy; Department of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
9
|
Sehgal IS, Arora K, Agarwal R, Kumar R, Rana N, Dhooria S, Muthu V, Prasad KT, Garg M, Rudramurthy SM, Aggarwal AN, Chakrabarti A. Serial Fluorodeoxyglucose Positron Emission Tomography-Computed Tomography (18FDG-PET-CT) in Assessing Treatment Response in Chronic Pulmonary Aspergillosis. J Infect Dis 2025; 231:532-539. [PMID: 39159179 DOI: 10.1093/infdis/jiae409] [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: 06/29/2024] [Revised: 08/09/2024] [Accepted: 08/15/2024] [Indexed: 08/21/2024] Open
Abstract
BACKGROUND The role of 2-deoxy-2-18(F) fluoro-D-glucose (FDG) positron emission tomography (PET)-computed tomography (CT) in assessing treatment response in chronic pulmonary aspergillosis (CPA) remains to be determined. The study objective was to compare FDG-PET/CT parameters in persons with CPA achieving treatment success or failure after 6 months of oral itraconazole. METHODS We performed PET-CT at baseline and after 6 months of oral itraconazole therapy. FDG uptake similar to the background uptake or ≥13 units decline in Z-score was considered a complete metabolic response (CMR). A >25%, >30%, and > 45% decline in standardised uptake value (SUVmax), SUVpeak, and total glycolytic activity (TLG) was labelled as a partial metabolic response (PMR). A >30%, >30%, or >75% increase in the SUVmax, SUVpeak, and TLG represented progressive metabolic disease. RESULTS We included 94 persons with CPA (63 male) with a mean age of 46.2 years. A follow-up PET-CT was performed on 77 participants. We recorded treatment success and failure in 43 and 34 patients. CMR was seen in 18.6% of those with treatment success and none with treatment failure. A higher proportion of patients with treatment success achieved PMR; 19% of the patients with treatment success had progressive metabolic disease. CONCLUSIONS Most PET-CT parameters improved with treatment; however, PET-CT misclassified one-fifth of the participants.
Collapse
Affiliation(s)
- Inderpaul Singh Sehgal
- Department of Pulmonary Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Kajal Arora
- Department of Pulmonary Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Ritesh Agarwal
- Department of Pulmonary Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Rajender Kumar
- Department of Nuclear Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Nivedita Rana
- Department of Nuclear Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Sahajal Dhooria
- Department of Pulmonary Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Valliappan Muthu
- Department of Pulmonary Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Kuruswamy Thurai Prasad
- Department of Pulmonary Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Mandeep Garg
- Department of Radiodiagnosis, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Shivaprakash M Rudramurthy
- Department of Medical Microbiology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Ashutosh Nath Aggarwal
- Department of Pulmonary Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | | |
Collapse
|
10
|
Neumann M, Reimann M, Chesov D, Popa C, Dragomir A, Popescu O, Munteanu R, Hölscher A, Honeyborne I, Heyckendorf J, Lange C, Hölscher C, Kalsdorf B. The molecular bacterial load assay predicts treatment responses in patients with pre-XDR/XDR-tuberculosis more accurately than GeneXpert Ultra MTB/Rif. J Infect 2025; 90:106399. [PMID: 39733827 DOI: 10.1016/j.jinf.2024.106399] [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: 07/05/2024] [Revised: 10/13/2024] [Accepted: 12/22/2024] [Indexed: 12/31/2024]
Abstract
OBJECTIVES Early detection of treatment failure is essential to improve the management of drug-resistant tuberculosis (DR-TB). We evaluated the molecular bacterial load assay (MBLA) in comparison to standard diagnostic tests for monitoring therapy of patients affected by drug-resistant TB. METHODS The performance of MBLA in tracking treatment response in a prospective cohort of patients with pulmonary MDR/RR- and pre-XDR/XDR-TB was compared with mycobacterial culture, mycobacterial DNA detection using GeneXpert (Xpert) and microscopy detection of sputum acid-fast-bacilli. RESULTS Mycobacterium tuberculosis culture conversion was used as the read-out for treatment responses. The MBLA was most concordant during the early phase of treatment, detecting changes in bacterial load with similar accuracy to microscopy and outperforming Xpert. When considering all timepoints, concordance with MGIT results was 72.1% for MBLA, 57.4% for Xpert and 76.7% for microscopy. The AUC for culture conversion was higher for MBLA (0.88, CI 0.84-0.95) than for Xpert (0.78, CI 0.72-0.85) and microscopy (0.77, CI 0.71-0.83). CONCLUSIONS MBLA was superior in the early identification of successful culture conversion compared to microscopy and Xpert and could be a useful biomarker to evaluate novel entities in Phase IIA early-bactericidal-activity drug trials regardless of the degree of M. tuberculosis drug resistance.
Collapse
Affiliation(s)
- Marit Neumann
- Division of Infection Immunology, Research Center Borstel, Parkallee 1-40, 23845 Borstel, Germany; German Center for Infection Research (DZIF), Partner site Hamburg-Lübeck-Borstel-Riems, Borstel, Germany
| | - Maja Reimann
- German Center for Infection Research (DZIF), Partner site Hamburg-Lübeck-Borstel-Riems, Borstel, Germany; Division of Clinical Infectious Diseases, Research Center Borstel, Parkallee 1-40, 23845 Borstel, Germany; Respiratory Medicine & International Health, University of Lübeck, Lübeck, Germany
| | - Dumitru Chesov
- German Center for Infection Research (DZIF), Partner site Hamburg-Lübeck-Borstel-Riems, Borstel, Germany; Division of Clinical Infectious Diseases, Research Center Borstel, Parkallee 1-40, 23845 Borstel, Germany; Nicolae Testemitanu State University of Medicine and Pharmacy, Stefan cel Mare si Sfant Boulevard 165, MD-2004 Chisinau, Republic of Moldova
| | - Cristina Popa
- National Institute of Pneumology Marius Nasta, Șoseaua Viilor 90, 050159 Bucharest, Romania
| | - Antonela Dragomir
- National Institute of Pneumology Marius Nasta, Șoseaua Viilor 90, 050159 Bucharest, Romania
| | - Oana Popescu
- National Institute of Pneumology Marius Nasta, Șoseaua Viilor 90, 050159 Bucharest, Romania
| | - Roxana Munteanu
- National Institute of Pneumology Marius Nasta, Șoseaua Viilor 90, 050159 Bucharest, Romania
| | - Alexandra Hölscher
- Division of Infection Immunology, Research Center Borstel, Parkallee 1-40, 23845 Borstel, Germany
| | - Isobella Honeyborne
- Centre for Clinical Microbiology, University College London, Gower Street, London WC1E 6BT, United Kingdom
| | - Jan Heyckendorf
- Division of Clinical Infectious Diseases, Research Center Borstel, Parkallee 1-40, 23845 Borstel, Germany; Internal Medicine I, University Hospitals Schleswig-Holstein, Kiel, Germany; Pulmonology and Inflammatory Medicine, Christian Albrechts-University, Kiel, Germany
| | - Christoph Lange
- German Center for Infection Research (DZIF), Partner site Hamburg-Lübeck-Borstel-Riems, Borstel, Germany; Division of Clinical Infectious Diseases, Research Center Borstel, Parkallee 1-40, 23845 Borstel, Germany; Respiratory Medicine & International Health, University of Lübeck, Lübeck, Germany; Baylor College of Medicine and Texas Children´s Hospital, Houston, TX, USA
| | - Christoph Hölscher
- Division of Infection Immunology, Research Center Borstel, Parkallee 1-40, 23845 Borstel, Germany; German Center for Infection Research (DZIF), Partner site Hamburg-Lübeck-Borstel-Riems, Borstel, Germany.
| | - Barbara Kalsdorf
- German Center for Infection Research (DZIF), Partner site Hamburg-Lübeck-Borstel-Riems, Borstel, Germany; Division of Clinical Infectious Diseases, Research Center Borstel, Parkallee 1-40, 23845 Borstel, Germany
| |
Collapse
|
11
|
Abikhzer G, Treglia G, Pelletier-Galarneau M, Buscombe J, Chiti A, Dibble EH, Glaudemans AWJM, Palestro CJ, Sathekge M, Signore A, Jamar F, Israel O, Gheysens O. EANM/SNMMI guideline/procedure standard for [ 18F]FDG hybrid PET use in infection and inflammation in adults v2.0. Eur J Nucl Med Mol Imaging 2025; 52:510-538. [PMID: 39387894 PMCID: PMC11732780 DOI: 10.1007/s00259-024-06915-3] [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: 07/18/2024] [Accepted: 09/02/2024] [Indexed: 10/15/2024]
Abstract
INTRODUCTION Hybrid [18F]FDG PET imaging is currently the method of choice for a wide variety of infectious and inflammatory disorders and was recently adopted in several clinical guidelines. A large amount of evidence-based articles, guidelines and appropriate use criteria have been published since the first version of this guideline in 2013. PURPOSE To provide updated evidence-based information to assist physicians in recommending, performing and interpreting hybrid [18F]FDG PET examinations for infectious and inflammatory disorders in the adult population. METHODS A systematic literature search of evidence-based articles using whole-body [18F]FDG hybrid imaging on the indications covered within this guideline was performed. All systematic reviews and meta-analyses published within the last 10 years until January 2023 were identified in PubMed/Medline or Cochrane. For each indication covered in this manuscript, diagnostic performance was provided based on meta-analyses or systematic reviews. If not available, results from prospective or retrospective studies were considered based on predefined selection criteria. RESULTS AND CONCLUSIONS: Hybrid [18F]FDG PET is extremely useful in the work-up and management of adults with infectious and inflammatory diseases, as supported by extensive and rapidly growing evidence-based literature and adoption in clinical guidelines. Practical recommendations are provided describing evidence-based indications as well as interpretation criteria and pitfalls. Monitoring treatment response is the most challenging but insufficiently studied potential application in infection and inflammation imaging.
Collapse
Affiliation(s)
- Gad Abikhzer
- Department of Medical Imaging, Faculty of Medicine and Health Sciences, Jewish General Hospital, McGill University, Montreal, Quebec, Canada
| | - Giorgio Treglia
- Nuclear Medicine, Imaging Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, 6900, Lugano, Switzerland
| | | | - John Buscombe
- Department of Nuclear Medicine, Cambridge University Hospitals, Cambridge, United Kingdom
| | - Arturo Chiti
- Department of Nuclear Medicine, IRCCS San Raffaele and Vita-Salute San Raffaele University, Milano, Italy
| | - Elizabeth H Dibble
- Department of Diagnostic Imaging, Warren Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI, USA
| | - Andor W J M Glaudemans
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.
| | | | - Mike Sathekge
- Nuclear Medicine Research Infrastructure (NuMeRI), University of Pretoria, Steve Biko Academic Hospital, Pretoria, South Africa
| | - Alberto Signore
- Nuclear Medicine Unit, Department of Medical-Surgical Sciences and of Translational Medicine, University Hospital S. Andrea, "Sapienza" University, Roma, Italy
| | - Francois Jamar
- Department of Nuclear Medicine, Cliniques Universitaires Saint-Luc and Institute of Clinical and Experimental Research (IREC), Université Catholique de Louvain, Brussels, Belgium
| | - Ora Israel
- Rappaport School of Medicine, Technion- Israel Institute of Technology, Haifa, Israel
| | - Olivier Gheysens
- Department of Nuclear Medicine, Cliniques Universitaires Saint-Luc and Institute of Clinical and Experimental Research (IREC), Université Catholique de Louvain, Brussels, Belgium
| |
Collapse
|
12
|
Scriba TJ, Maseeme M, Young C, Taylor L, Leslie AJ. Immunopathology in human tuberculosis. Sci Immunol 2024; 9:eado5951. [PMID: 39671470 DOI: 10.1126/sciimmunol.ado5951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 11/15/2024] [Indexed: 12/15/2024]
Abstract
Mycobacterium tuberculosis (M.tb) is a bacterial pathogen that has evolved in humans, and its interactions with the host are complex and best studied in humans. Myriad immune pathways are involved in infection control, granuloma formation, and progression to tuberculosis (TB) disease. Inflammatory cells, such as macrophages, neutrophils, conventional and unconventional T cells, B cells, NK cells, and innate lymphoid cells, interact via cytokines, cell-cell communication, and eicosanoid signaling to contain or eliminate infection but can alternatively mediate pathological changes required for pathogen transmission. Clinical manifestations include pulmonary and extrapulmonary TB, as well as post-TB lung disease. Risk factors for TB progression, in turn, largely relate to immune status and, apart from traditional chemotherapy, interventions primarily target immune mechanisms, highlighting the critical role of immunopathology in TB. Maintaining a balance between effector mechanisms to achieve protective immunity and avoid detrimental inflammation is central to the immunopathogenesis of TB. Many research gaps remain and deserve prioritization to improve our understanding of human TB immunopathogenesis.
Collapse
Affiliation(s)
- Thomas J Scriba
- South African Tuberculosis Vaccine Initiative, Division of Immunology, Department of Pathology and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Mahlatse Maseeme
- Africa Health Research Institute, Durban, South Africa
- College of Heath Sciences, School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Carly Young
- South African Tuberculosis Vaccine Initiative, Division of Immunology, Department of Pathology and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Laura Taylor
- Forensic Pathology Services, Western Cape Government/University of Cape Town, Cape Town, South Africa
| | - Alasdair J Leslie
- Africa Health Research Institute, Durban, South Africa
- University College London, London, UK
| |
Collapse
|
13
|
Lyu J, Narum DE, Baldwin SL, Larsen SE, Bai X, Griffith DE, Dartois V, Naidoo T, Steyn AJC, Coler RN, Chan ED. Understanding the development of tuberculous granulomas: insights into host protection and pathogenesis, a review in humans and animals. Front Immunol 2024; 15:1427559. [PMID: 39717773 PMCID: PMC11663721 DOI: 10.3389/fimmu.2024.1427559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Accepted: 11/18/2024] [Indexed: 12/25/2024] Open
Abstract
Granulomas, organized aggregates of immune cells which form in response to Mycobacterium tuberculosis (Mtb), are characteristic but not exclusive of tuberculosis (TB). Despite existing investigations on TB granulomas, the determinants that differentiate host-protective granulomas from granulomas that contribute to TB pathogenesis are often disputed. Thus, the goal of this narrative review is to help clarify the existing literature on such determinants. We adopt the a priori view that TB granulomas are host-protective organelles and discuss the molecular and cellular determinants that induce protective granulomas and those that promote their failure. While reports about protective TB granulomas and their failure may initially seem contradictory, it is increasingly recognized that either deficiencies or excesses of the molecular and cellular components in TB granuloma formation may be detrimental to the host. More specifically, insufficient or excessive expression/representation of the following components have been reported to skew granulomas toward the less protective phenotype: (i) epithelioid macrophages; (ii) type 1 adaptive immune response; (iii) type 2 adaptive immune response; (iv) tumor necrosis factor; (v) interleukin-12; (vi) interleukin-17; (vii) matrix metalloproteinases; (viii) hypoxia in the TB granulomas; (ix) hypoxia inducible factor-1 alpha; (x) aerobic glycolysis; (xi) indoleamine 2,3-dioxygenase activity; (xii) heme oxygenase-1 activity; (xiii) immune checkpoint; (xiv) leukotriene A4 hydrolase activity; (xv) nuclear-factor-kappa B; and (xvi) transforming growth factor-beta. Rather, more precise and timely coordinated immune responses appear essential for eradication or containment of Mtb infection. Since there are several animal models of infection with Mtb, other species within the Mtb complex, and the surrogate Mycobacterium marinum - whether natural (cattle, elephants) or experimental (zebrafish, mouse, guinea pig, rabbit, mini pig, goat, non-human primate) infections - we also compared the TB granulomatous response and other pathologic lung lesions in various animals infected with one of these mycobacteria with that of human pulmonary TB. Identifying components that dictate the formation of host-protective granulomas and the circumstances that result in their failure can enhance our understanding of the macrocosm of human TB and facilitate the development of novel remedies - whether they be direct therapeutics or indirect interventions - to efficiently eliminate Mtb infection and prevent its pathologic sequelae.
Collapse
Affiliation(s)
- Jiwon Lyu
- Division of Pulmonary and Critical Medicine, Soon Chun Hyang University Cheonan Hospital, Seoul, Republic of Korea
- Department of Academic Affairs, National Jewish Health, Denver, CO, United States
| | - Drew E. Narum
- Department of Academic Affairs, National Jewish Health, Denver, CO, United States
| | - Susan L. Baldwin
- Center for Global Infectious Diseases, Seattle Children’s Research Institute, Seattle, WA, United States
| | - Sasha E. Larsen
- Center for Global Infectious Diseases, Seattle Children’s Research Institute, Seattle, WA, United States
| | - Xiyuan Bai
- Department of Academic Affairs, National Jewish Health, Denver, CO, United States
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado School of Medicine, Aurora, CO, United States
| | - David E. Griffith
- Department of Medicine, National Jewish Health, Denver, CO, United States
| | - Véronique Dartois
- Center for Discovery and Innovation, Hackensack Meridian School of Medicine, Nutley, NJ, United States
| | - Threnesan Naidoo
- Departments of Forensic & Legal Medicine and Laboratory Medicine & Pathology, Faculty of Medicine & Health Sciences, Walter Sisulu University, Mthatha, South Africa
| | - Adrie J. C. Steyn
- Africa Health Research Institute, University of KwaZulu-Natal, Durban, South Africa
- Department of Microbiology and Centers for AIDS Research and Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Rhea N. Coler
- Center for Global Infectious Diseases, Seattle Children’s Research Institute, Seattle, WA, United States
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, United States
- Department of Global Health, University of Washington, Seattle, WA, United States
| | - Edward D. Chan
- Department of Academic Affairs, National Jewish Health, Denver, CO, United States
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado School of Medicine, Aurora, CO, United States
- Department of Medicine, Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO, United States
| |
Collapse
|
14
|
Scherer J, Mukasa SL, Wolmarans K, Guler R, Kotze T, Song T, Dunn R, Laubscher M, Pape HC, Held M, Thienemann F. Multi-level tuberculosis of the spine identified by 18 F-FDG-PET/CT and concomitant urogenital tuberculosis: a case report from the spinal TB X cohort. Infection 2024; 52:2507-2519. [PMID: 38896371 PMCID: PMC11621135 DOI: 10.1007/s15010-024-02327-5] [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: 03/30/2024] [Accepted: 06/15/2024] [Indexed: 06/21/2024]
Abstract
BACKGROUND Tuberculosis (TB) is caused by Mycobacterium tuberculosis (Mtb) and typically infects the lungs. However, extrapulmonary forms of TB can be found in approximately 20% of cases. It is suggested, that up to 10% of extrapulmonary TB affects the musculoskeletal system, in which spinal elements (spinal tuberculosis, STB) are involved in approximately 50% of the cases. STB is a debilitating disease with nonspecific symptoms and diagnosis is often delayed for months to years. In our Spinal TB X Cohort, we aim to describe the clinical phenotype of STB using whole-body 18 F-fluorodeoxyglucose positron emission tomography computed tomography (PET/CT) and to identify a specific gene expression profile for the different stages of dissemination on PET/CT. Here we report on the first patient recruited into our cohort who underwent PET/CT before treatment initiation, at 6-months and at 12-months - time of TB treatment completion. CASE PRESENTATION A 27-year-old immunocompetent male presented with severe thoracolumbar back pain for 9 months with severe antalgic gait and night sweats. Magnetic resonance imaging (MRI) of the whole spine revealed multilevel spinal disease (T5/6, T11/12, L3/4) in keeping with STB. After informed consent and recruitment into the Spinal TB X Cohort, the patient underwent PET/CT as per protocol, which revealed isolated multilevel STB (T4-7, T11/12, L3/4) with no concomitant lung or urogenital lesion. However, sputum and urine were Xpert MTB/RIF Ultra positive and Mtb was cultured from the urine sample. CT-guided biopsy of the T11/12 lesion confirmed drug-sensitive Mtb on Xpert MTB/RIF Ultra and the patient was started on TB treatment according to local guidelines for 12 months. The 6-month follow-up PET/CT revealed new and existing spinal lesions with increased FDG-uptake despite significant improvement of clinical features and laboratory markers. After 9 months of treatment, the patient developed an acute urethral stricture, most likely due to urogenital TB, and a suprapubic catheter was inserted. The 12-month PET/CT showed significantly decreased PET/CT values of all lesions, however, significant persistent spinal inflammation was present at the end of TB treatment. Clinically, the patient was considered cured by the TB control program and currently awaits urethroplasty. CONCLUSIONS In our case, PET/CT emerged as a valuable imaging modality for the initial assessment, surpassing MRI by revealing more comprehensive extensive disease. Subsequent PET/CT scans at 6-month uncovered new lesions and increased inflammation in existing ones, while by the end of TB treatment, all lesions exhibited improvement. However, the interpretation of FDG avidity remains ambiguous, whether it correlates with active infection and viable Mtb. or fibro- and osteoblast activity indicative of the healing process. Additionally, the absence of extraspinal TB lesions on PET/CT despite positive microbiology from sputum and urine maybe explained by paucibacillary, subclinical infection of extraspinal organs. The Spinal TB X Cohort endeavours to shed light on whole-body imaging patterns at diagnosis, their evolution midway through TB treatment, and upon treatment completion. Ultimately, this study aims to advance our understanding of the biology of this complex disease.
Collapse
Affiliation(s)
- Julian Scherer
- General Medicine & Global Health (GMGH), Department of Medicine and Orthopaedic Research Unit (ORU), Division of Orthopaedic Surgery, Faculty of Health Science, University of Cape Town, Cape Town, South Africa.
- Department of Traumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland.
| | - Sandra L Mukasa
- General Medicine & Global Health (GMGH), Department of Medicine, Faculty of Health Science, University of Cape Town, Cape Town, South Africa
| | - Karen Wolmarans
- General Medicine & Global Health (GMGH), Department of Medicine, Faculty of Health Science, University of Cape Town, Cape Town, South Africa
| | - Reto Guler
- Institute of Infectious Diseases and Molecular Medicine (IDM), Department of Pathology, Division of Immunology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, Cape Town, South Africa
| | - Tessa Kotze
- Department of Medicine, CUBIC, PETCT, University of Cape Town, Cape Town, South Africa
| | - Taeksun Song
- Institute of Infectious Diseases and Molecular Medicine (IDM), Department of Pathology, Division of Medical Microbiology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Robert Dunn
- Orthopaedic Research Unit (ORU), Division of Orthopaedic Surgery, Faculty of Health Science, University of Cape Town, Cape Town, South Africa
| | - Maritz Laubscher
- Orthopaedic Research Unit (ORU), Division of Orthopaedic Surgery, Faculty of Health Science, University of Cape Town, Cape Town, South Africa
| | - Hans-Christoph Pape
- Department of Traumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Michael Held
- Orthopaedic Research Unit (ORU), Division of Orthopaedic Surgery, Faculty of Health Science, University of Cape Town, Cape Town, South Africa
| | - Friedrich Thienemann
- General Medicine & Global Health (GMGH), Department of Medicine, Faculty of Health Science, University of Cape Town, Cape Town, South Africa
- Department of Internal Medicine, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
15
|
Coetzee JL, Kriel NL, Loubser J, Dippenaar A, Sampson SL, Malherbe ST, Mouton JM. Assessing the propensity of TB clinical isolates to form viable but non-replicating subpopulations. Sci Rep 2024; 14:27686. [PMID: 39532967 PMCID: PMC11557868 DOI: 10.1038/s41598-024-79389-w] [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: 03/14/2024] [Accepted: 11/08/2024] [Indexed: 11/16/2024] Open
Abstract
Current tuberculosis (TB) treatment is typically effective against drug-susceptible Mycobacterium tuberculosis, but can fail due to acquired drug resistance or phenotypic resistance. M. tuberculosis persisters, a subpopulation of viable but non-replicating (VBNR) antibiotic-tolerant bacteria, are thought to contribute to poor TB treatment outcomes. In this exploratory study, we investigated treatment-naïve drug-susceptible clinical isolates collected from people with TB, who subsequently had unsuccessful treatment outcomes. These were compared to isolates from cured individuals in terms of their ability to form VBNR subpopulations. Clinical isolates from individuals with unfavorable treatment outcomes form larger subpopulations of VBNR M. tuberculosis (2.67-13.71%) than clinical isolates from cured cases (0- 1.63%) following infection of THP-1 macrophages. All isolates were drug susceptible based on phenotypic and genotypic analysis. Whole genome sequencing identified 23 non-synonymous genomic variants shared by treatment failure clinical isolates, that were not present in isolates from cured cases. This exploratory study highlights the ability of treatment-naïve clinical isolates to form heterogeneous populations containing VBNR M. tuberculosis. We also demonstrate that clinical isolates from individuals with unsuccessful treatment outcomes form higher percentages of VBNR M. tuberculosis. The findings of this exploratory study suggest that an increased propensity to form VBNR subpopulations may impact TB treatment outcome.
Collapse
Affiliation(s)
- Julian L Coetzee
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research, SA MRC Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, PO Box 19063, Tygerberg, Cape Town, 7505, South Africa
| | - Nastassja L Kriel
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research, SA MRC Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, PO Box 19063, Tygerberg, Cape Town, 7505, South Africa.
| | - Johannes Loubser
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research, SA MRC Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, PO Box 19063, Tygerberg, Cape Town, 7505, South Africa
| | - Anzaan Dippenaar
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research, SA MRC Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, PO Box 19063, Tygerberg, Cape Town, 7505, South Africa
- Tuberculosis Omics Research Consortium, Family Medicine and Population Health, Institute of Global Health, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Samantha L Sampson
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research, SA MRC Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, PO Box 19063, Tygerberg, Cape Town, 7505, South Africa
| | - Stephanus T Malherbe
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research, SA MRC Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, PO Box 19063, Tygerberg, Cape Town, 7505, South Africa
| | - Jacoba M Mouton
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research, SA MRC Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, PO Box 19063, Tygerberg, Cape Town, 7505, South Africa
| |
Collapse
|
16
|
Calderwood CJ, Sanchez Martinez A, Greenan-Barrett J, Turner CT, Oguti B, Roe JK, Gupta R, Martineau AR, Noursadeghi M. Resolution of tuberculosis blood RNA signatures fails to discriminate persistent sputum culture positivity after 8 weeks of anti-tuberculous treatment. Eur Respir J 2024; 64:2400457. [PMID: 39190790 PMCID: PMC11579542 DOI: 10.1183/13993003.00457-2024] [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: 03/05/2024] [Accepted: 07/09/2024] [Indexed: 08/29/2024]
Abstract
BACKGROUND Concerted efforts aim to reduce the burden of 6 months of anti-tuberculous treatment for tuberculosis (TB). Treatment cessation at 8 weeks is effective for most but incurs increased risk of disease relapse. We tested the hypothesis that blood RNA signatures or C-reactive protein (CRP) measurements discriminate 8-week sputum culture status, as a prerequisite for a biomarker to stratify risk of relapse following treatment cessation at this time-point. METHODS We identified blood RNA signatures of TB disease or cure by systematic review. We evaluated these signatures and CRP measurements in a pulmonary TB cohort, pre-treatment, at 2 and 8 weeks of treatment, and sustained cure after treatment completion. We tested biomarker discrimination of 8-week sputum culture status using area under the receiver operating characteristic curve (AUROC) analysis and, secondarily, assessed correlation of biomarker scores with time to culture positivity at 8 weeks of treatment. RESULTS 12 blood RNA signatures were reproduced in the dataset from 44 individuals with sputum culture-positive pulmonary TB. These normalised over time from TB treatment initiation. 11 out of 44 cases with blood RNA, CRP and sputum culture results were sputum culture-positive at 8 weeks of treatment. None of the contemporary blood RNA signatures discriminated sputum culture status at this time-point or correlated with bacterial load. CRP achieved modest discrimination with AUROC 0.69 (95% CI 0.52-0.87). CONCLUSIONS Selected TB blood RNA signatures and CRP do not provide biomarkers of microbiological clearance to support TB treatment cessation at 8 weeks. Resolution of blood transcriptional host responses in sputum culture-positive individuals suggests Mycobacterium tuberculosis may colonise the respiratory tract without triggering a detectable immune response.
Collapse
Affiliation(s)
- Claire J Calderwood
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
- C.J. Calderwood and A. Sanchez Martinez contributed equally
| | - Alvaro Sanchez Martinez
- Division of Infection and Immunity, University College London, London, UK
- C.J. Calderwood and A. Sanchez Martinez contributed equally
| | - James Greenan-Barrett
- Department of Respiratory Medicine, University College London Hospitals NHS Foundation Trust, London, UK
| | - Carolin T Turner
- Division of Infection and Immunity, University College London, London, UK
| | - Blanché Oguti
- Division of Infection and Immunity, University College London, London, UK
| | - Jennifer K Roe
- Division of Infection and Immunity, University College London, London, UK
| | - Rishi Gupta
- Institute of Health Informatics, University College London, London, UK
| | - Adrian R Martineau
- Blizard Institute, Queen Mary University of London, London, UK
- A.R. Martineau and M. Noursadeghi are co-senior authors
| | - Mahdad Noursadeghi
- Division of Infection and Immunity, University College London, London, UK
- A.R. Martineau and M. Noursadeghi are co-senior authors
| |
Collapse
|
17
|
Malakar B, Barth VC, Puffal J, Woychik NA, Husson RN. Phosphorylation of VapB antitoxins affects intermolecular interactions to regulate VapC toxin activity in Mycobacterium tuberculosis. J Bacteriol 2024; 206:e0023324. [PMID: 39315797 PMCID: PMC11500542 DOI: 10.1128/jb.00233-24] [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: 06/05/2024] [Accepted: 08/15/2024] [Indexed: 09/25/2024] Open
Abstract
Toxin-antitoxin modules are present in many bacterial pathogens. The VapBC family is particularly abundant in members of the Mycobacterium tuberculosis complex, with 50 modules present in the M. tuberculosis genome. In type IIA modules, the VapB antitoxin protein binds to and inhibits the activity of the co-expressed cognate VapC toxin protein. VapB proteins may also bind to promoter region sequences and repress the expression of the vapB-vapC operon. Though VapB-VapC interactions can control the amount of free VapC toxin in the bacterial cell, the mechanisms that affect this interaction are poorly understood. Based on our recent finding of Ser/Thr phosphorylation of VapB proteins in M. tuberculosis, we substituted phosphomimetic or phosphoablative amino acids at the phosphorylation sites of two VapB proteins. We found that phosphomimetic substitution of VapB27 and VapB46 resulted in decreased interaction with their respective cognate VapC proteins, whereas phosphoablative substitution did not alter binding. Similarly, we determined that phosphomimetic substitution interfered with VapB binding to promoter region DNA sequences. Both decreased VapB-VapC interaction and decreased VapB repression of vapB-vapC operon transcription would result in increased free VapC in the M. tuberculosis cell. In growth inhibition experiments, M. tuberculosis strains expressing vapB46-vapC46 constructs containing a phosphoablative vapB mutation resulted in lower toxicity compared to a strain expressing native vapB46, whereas similar or greater toxicity was observed in the strain expressing the phosphomimetic vapB mutation. These results identify a novel mechanism by which VapC toxicity activity can be regulated by VapB phosphorylation.IMPORTANCEIntracellular bacterial toxins are present in many bacterial pathogens and have been linked to bacterial survival in response to stresses encountered during infection. The activity of many toxins is regulated by a co-expressed antitoxin protein that binds to and sequesters the toxin protein. The mechanisms by which an antitoxin may respond to stresses to alter toxin activity are poorly understood. Here, we show that antitoxin interactions with its cognate toxin and with promoter DNA required for antitoxin and toxin expression can be altered by Ser/Thr phosphorylation of the antitoxin and, thus, affect toxin activity. This reversible modification may play an important role in regulating toxin activity within the bacterial cell in response to signals generated during infection.
Collapse
Affiliation(s)
- Basanti Malakar
- Division of Infectious Diseases, Boston Children’s Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Valdir C. Barth
- Division of Infectious Diseases, Boston Children’s Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Julia Puffal
- Department of Biochemistry and Molecular Biology, Rutgers University, Robert Wood Johnson Medical School, Piscataway, New Jersey, USA
| | - Nancy A. Woychik
- Department of Biochemistry and Molecular Biology, Rutgers University, Robert Wood Johnson Medical School, Piscataway, New Jersey, USA
| | - Robert N. Husson
- Division of Infectious Diseases, Boston Children’s Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
18
|
Auld SC, Barczak AK, Bishai W, Coussens AK, Dewi IMW, Mitini-Nkhoma SC, Muefong C, Naidoo T, Pooran A, Stek C, Steyn AJC, Tezera L, Walker NF. Pathogenesis of Post-Tuberculosis Lung Disease: Defining Knowledge Gaps and Research Priorities at the Second International Post-Tuberculosis Symposium. Am J Respir Crit Care Med 2024; 210:979-993. [PMID: 39141569 PMCID: PMC11531093 DOI: 10.1164/rccm.202402-0374so] [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: 02/17/2024] [Accepted: 08/13/2024] [Indexed: 08/16/2024] Open
Abstract
Post-tuberculosis (post-TB) lung disease is increasingly recognized as a major contributor to the global burden of chronic lung disease, with recent estimates indicating that over half of TB survivors have impaired lung function after successful completion of TB treatment. However, the pathologic mechanisms that contribute to post-TB lung disease are not well understood, thus limiting the development of therapeutic interventions to improve long-term outcomes after TB. This report summarizes the work of the Pathogenesis and Risk Factors Committee for the Second International Post-Tuberculosis Symposium, which took place in Stellenbosch, South Africa, in April 2023. The committee first identified six areas with high translational potential: 1) tissue matrix destruction, including the role of matrix metalloproteinase dysregulation and neutrophil activity; 2) fibroblasts and profibrotic activity; 3) granuloma fate and cell death pathways; 4) mycobacterial factors, including pathogen burden; 5) animal models; and 6) the impact of key clinical risk factors, including HIV, diabetes, smoking, malnutrition, and alcohol. We share the key findings from a literature review of those areas, highlighting knowledge gaps and areas where further research is needed.
Collapse
Affiliation(s)
- Sara C. Auld
- Departments of Medicine, Epidemiology, and Global Health, Emory University School of Medicine and Rollins School of Public Health, Atlanta, Georgia
| | - Amy K. Barczak
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, Massachusetts
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard, Cambridge, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - William Bishai
- Division of Infectious Diseases, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Anna K. Coussens
- Infectious Diseases and Immune Defence Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Intan M. W. Dewi
- Microbiology Division, Department of Biomedical Sciences, Faculty of Medicine, and
- Research Center for Care and Control of Infectious Diseases, Universitas Padjadjaran, Bandung, Indonesia
| | | | - Caleb Muefong
- Department of Microbiology, University of Chicago, Chicago, Illinois
| | - Threnesan Naidoo
- Department of Forensic & Legal Medicine and
- Department of Laboratory Medicine & Pathology, Faculty of Medicine & Health Sciences, Walter Sisulu University, Eastern Cape, South Africa
- Africa Health Research Institute, University of KwaZulu-Natal, Durban, South Africa
| | - Anil Pooran
- Centre for Lung Infection and Immunity, Division of Pulmonology, Department of Medicine, and
- University of Cape Town Lung Institute and Medical Research Council/University of Cape Town Centre for the Study of Antimicrobial Resistance, Cape Town, South Africa
| | - Cari Stek
- Wellcome Center for Infectious Diseases Research in Africa, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Adrie J. C. Steyn
- Africa Health Research Institute, University of KwaZulu-Natal, Durban, South Africa
- Department of Microbiology and
- Centers for AIDS Research and Free Radical Biology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Liku Tezera
- National Institute for Health and Care Research Biomedical Research Centre, School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Naomi F. Walker
- Department of Clinical Sciences and Centre for Tuberculosis Research, Liverpool School of Tropical Medicine, Liverpool, United Kingdom; and
- Tropical and Infectious Diseases Unit, Liverpool University Hospitals NHS Foundation Trust, Liverpool, United Kingdom
| |
Collapse
|
19
|
Malherbe ST, Chen RY, Yu X, Smith B, Liu X, Gao J, Diacon AH, Dawson R, Tameris M, Zhu H, Qu Y, Jin H, Pan S, Dodd LE, Wang J, Goldfeder LC, Cai Y, Arora K, Vincent J, Narunsky K, Serole K, Goliath RT, Da Costa L, Taliep A, Aziz S, Daroowala R, Thienemann F, Mukasa S, Court R, Sossen B, Ahlers P, Mendelsohn SC, White L, Gouel A, Lau CY, Hassan S, Liang L, Duan H, Moghaddam GK, Paripati P, Lahouar S, Harris M, Wollenberg K, Jeffrey B, Tartakovsky M, Rosenthal A, Duvenhage M, Armstrong DT, Song T, Winter J, Gao Q, Via LE, Wilkinson RJ, Walzl G, Barry CE. PET/CT guided tuberculosis treatment shortening: a randomized trial. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.10.03.24314723. [PMID: 39802795 PMCID: PMC11722446 DOI: 10.1101/2024.10.03.24314723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/21/2025]
Abstract
Six months of chemotherapy using current agents is standard of care for pulmonary, drug-sensitive tuberculosis (TB), even though some are believed to be cured more rapidly and others require longer therapy. Understanding what factors determine the length of treatment required for durable cure in individual patients would allow individualization of treatment durations, provide better clinical tools to determine the of appropriate duration of new regimens, as well as reduce the cost of large Phase III studies to determine the optimal combinations to use in TB control programs. We conducted a randomized clinical trial in South Africa and China that recruited 704 participants with newly diagnosed, drug-sensitive pulmonary tuberculosis and stratified them based on radiographic disease characteristics as assessed by FDG PET/CT scan readers. Participants with less extensive disease (N=273) were randomly assigned to complete therapy after four months or continue receiving treatment for six months. Amongst participants who received four months of therapy, 17 of 141 (12.1%) experienced unfavorable outcomes compared to only 2 of 132 (1.5%) who completed six months of treatment (treatment success 98.4% in B, 86.7% in C (difference -11.7%, 95% CI, -18.2%, -5.3%)). In the non-randomized arm that included participants with more extensive disease, only 8 of 248 (3.2%) experienced unfavorable outcomes. Total cavity volume and total lesion glycolysis at week 16 were significantly associated with risk of unfavorable outcome in the randomized participants. Based on PET/CT scans at TB recurrence, bacteriological relapses (confirmed by whole genome sequencing) predominantly occurred in the same active cavities originally present at baseline. Automated segmentation of the serial PET/CT scans was later performed, and machine-learning was used to classify participants according to their likelihood of relapse, allowing the development of predictive models with good performance based on CT, PET, microbiological and clinical characteristics. These results open the possibility for more efficient studies of future TB treatment regimens.
Collapse
Affiliation(s)
- Stephanus T. Malherbe
- DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Immunology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Ray Y. Chen
- Tuberculosis Research Section, Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, USA
| | - Xiang Yu
- Tuberculosis Research Section, Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, USA
| | - Bronwyn Smith
- DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Immunology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Xin Liu
- Henan Provincial Chest Hospital, Zhengzhou, Henan, China
| | - Jingcai Gao
- Sino-US Tuberculosis Collaborative Research Program, Zhengzhou, Henan, China
| | | | - Rodney Dawson
- Division of Pulmonology, Department of Medicine, University of Cape Town Lung Institute, Centre for TB Research Innovation, Observatory 7923, Republic of South Africa
| | - Michele Tameris
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine, Department of Pathology, University of Cape Town, Observatory 7923, Republic of South Africa
| | - Hong Zhu
- Sino-US Tuberculosis Collaborative Research Program, Zhengzhou, Henan, China
| | - Yahong Qu
- Kaifeng City Institute of Tuberculosis Prevention and Control, Kaifeng, Henan, China
| | - Hongjian Jin
- Xinmi Center for Disease Control and Prevention, Xinmi, Henan, China
| | - Shouguo Pan
- Zhongmu County Health and Epidemic Prevention Station, Zhongmu, Henan, China
| | - Lori E. Dodd
- Clinical Clinical Trials Research and Statistics Branch, Office of Biostatistics Research, Division of Clinical Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, USA
| | - Jing Wang
- Clinical Monitoring Research Program Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Lisa C. Goldfeder
- Tuberculosis Research Section, Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, USA
| | - Ying Cai
- Tuberculosis Research Section, Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, USA
| | - Kriti Arora
- Tuberculosis Research Section, Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, USA
| | - Joel Vincent
- Tuberculosis Research Section, Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, USA
| | - Kim Narunsky
- University of Cape Town Lung Institute (Pty) Ltd, Mowbray, Cape Town, 7700, Republic of South Africa
| | - Keboile Serole
- Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory 7925, Republic of South Africa
| | - Rene T. Goliath
- Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory 7925, Republic of South Africa
| | - Laylah Da Costa
- Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory 7925, Republic of South Africa
| | - Arshad Taliep
- Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory 7925, Republic of South Africa
| | - Saalikha Aziz
- Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory 7925, Republic of South Africa
| | - Remy Daroowala
- Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory 7925, Republic of South Africa
| | - Friedrich Thienemann
- Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory 7925, Republic of South Africa
- Department of Internal Medicine, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Sandra Mukasa
- Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory 7925, Republic of South Africa
| | - Richard Court
- Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory 7925, Republic of South Africa
- Department of Medicine, University of Cape Town, Observatory 7923, Republic of South Africa
| | - Bianca Sossen
- Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory 7925, Republic of South Africa
- Department of Medicine, University of Cape Town, Observatory 7923, Republic of South Africa
| | - Petri Ahlers
- DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Immunology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Simon C. Mendelsohn
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine, Department of Pathology, University of Cape Town, Observatory 7923, Republic of South Africa
| | - Lisa White
- University of Cape Town Lung Institute (Pty) Ltd, Mowbray, Cape Town, 7700, Republic of South Africa
| | - Aurélie Gouel
- Clinical Clinical Trials Research and Statistics Branch, Office of Biostatistics Research, Division of Clinical Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, USA
| | - Chuen-Yen Lau
- HIV Dynamics and Replication Program, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Samy Hassan
- Department of Internal Medicine, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Lili Liang
- Henan Provincial Chest Hospital, Zhengzhou, Henan, China
| | | | | | - Praveen Paripati
- Office of Cyber Infrastructure and Computational Biology, NIAID, NIH, Bethesda, MD, USA
| | - Saher Lahouar
- Office of Cyber Infrastructure and Computational Biology, NIAID, NIH, Bethesda, MD, USA
| | - Michael Harris
- Office of Cyber Infrastructure and Computational Biology, NIAID, NIH, Bethesda, MD, USA
| | - Kurt Wollenberg
- Office of Cyber Infrastructure and Computational Biology, NIAID, NIH, Bethesda, MD, USA
| | - Brendan Jeffrey
- Office of Cyber Infrastructure and Computational Biology, NIAID, NIH, Bethesda, MD, USA
| | - Mike Tartakovsky
- Office of Cyber Infrastructure and Computational Biology, NIAID, NIH, Bethesda, MD, USA
| | - Alex Rosenthal
- Office of Cyber Infrastructure and Computational Biology, NIAID, NIH, Bethesda, MD, USA
| | - Michael Duvenhage
- Research Data and Communication Technologies, Inc., Garrett Park, MD, USA
| | | | - Taeksun Song
- Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory 7925, Republic of South Africa
| | - Jill Winter
- Catalysis Foundation for Health, San Ramon, CA, USA
| | - Qian Gao
- School of Basic Medical Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Laura E. Via
- Tuberculosis Research Section, Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, USA
- Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory 7925, Republic of South Africa
| | - Robert J. Wilkinson
- Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory 7925, Republic of South Africa
- Francis Crick Institute, London, NW1 1AT, UK
- Department of Infectious Diseases, Imperial College London, W12 0NN, UK
| | - Gerhard Walzl
- DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Immunology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Clifton E. Barry
- Tuberculosis Research Section, Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, USA
- Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory 7925, Republic of South Africa
| |
Collapse
|
20
|
Suliman S, Maison DP, Henrich TJ. The promise and reality of new immune profiling technologies. Nat Immunol 2024; 25:1765-1769. [PMID: 39242838 DOI: 10.1038/s41590-024-01948-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/09/2024]
Affiliation(s)
- Sara Suliman
- Division of Experimental Medicine, University of California San Francisco, San Francisco, CA, USA.
- Chan Zuckerberg Biohub, San Francisco, CA, USA.
| | - David P Maison
- Division of Experimental Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Timothy J Henrich
- Division of Experimental Medicine, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
21
|
Dinkele R, Gessner S, Patterson B, McKerry A, Hoosen Z, Vazi A, Seldon R, Koch A, Warner DF, Wood R. Persistent Mycobacterium tuberculosis bioaerosol release in a tuberculosis-endemic setting. iScience 2024; 27:110731. [PMID: 39310776 PMCID: PMC11414687 DOI: 10.1016/j.isci.2024.110731] [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: 05/21/2024] [Revised: 07/22/2024] [Accepted: 08/12/2024] [Indexed: 09/25/2024] Open
Abstract
Pioneering studies linking symptomatic disease and cough-mediated Mycobacterium tuberculosis (Mtb) release established the infectious origin of tuberculosis (TB), simultaneously informing the notion that pathology is a prerequisite for Mtb transmission. Our recent work has challenged this assumption: by sampling TB clinic attendees, we detected equivalent release of Mtb-containing bioaerosols by confirmed TB patients and individuals not receiving a TB diagnosis and observed time-dependent reduction in Mtb bioaerosol positivity during 6-month follow-up of both cohorts, irrespective of anti-TB chemotherapy. Now, we report widespread Mtb release in our TB-endemic setting: of 89 randomly recruited community members, 79.8% (71/89) produced Mtb-containing bioaerosols independently of QuantiFERON status, a standard test for Mtb exposure. Moreover, during 2-month longitudinal sampling, only 2% (1/50) were serially Mtb bioaerosol negative. These results necessitate a reframing of the prevailing paradigm of Mtb transmission and TB etiology, perhaps explaining the historical inability to elucidate Mtb transmission networks in TB-endemic regions.
Collapse
Affiliation(s)
- Ryan Dinkele
- UCT Molecular Mycobacteriology Research Unit, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa
| | - Sophia Gessner
- UCT Molecular Mycobacteriology Research Unit, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa
| | - Benjamin Patterson
- Amsterdam Institute for Global Health and Development, University of Amsterdam, Amsterdam 1105, the Netherlands
| | - Andrea McKerry
- Aerobiology and TB Research Unit, Desmond Tutu Health Foundation, Cape Town 7925, South Africa
| | - Zeenat Hoosen
- Aerobiology and TB Research Unit, Desmond Tutu Health Foundation, Cape Town 7925, South Africa
| | - Andiswa Vazi
- Aerobiology and TB Research Unit, Desmond Tutu Health Foundation, Cape Town 7925, South Africa
| | - Ronnett Seldon
- Aerobiology and TB Research Unit, Desmond Tutu Health Foundation, Cape Town 7925, South Africa
| | - Anastasia Koch
- UCT Molecular Mycobacteriology Research Unit, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa
| | - Digby F. Warner
- UCT Molecular Mycobacteriology Research Unit, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa
- Wellcome Centre for Infectious Diseases Research in Africa, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa
| | - Robin Wood
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa
- Aerobiology and TB Research Unit, Desmond Tutu Health Foundation, Cape Town 7925, South Africa
| |
Collapse
|
22
|
Hu S, Yu Q, Liu F, Gong F. A Novel Inflammatory Indicator for Tuberculosis-Associated Obstructive Pulmonary Disease (TOPD): The Systemic Inflammatory Response Index (SIRI). J Inflamm Res 2024; 17:4219-4228. [PMID: 38974002 PMCID: PMC11227324 DOI: 10.2147/jir.s468232] [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: 03/11/2024] [Accepted: 06/12/2024] [Indexed: 07/09/2024] Open
Abstract
Background The development of chronic obstructive pulmonary disease (COPD) following tuberculosis (TB) is known as tuberculosis-associated obstructive pulmonary disease (TOPD). This study aimed to explore the predictive value of inflammatory indicators for TOPD in TB patients. Methods Data for this cross-sectional study were collected between January 2014 and January 2022 at Wuhan Jinyintan Hospital. The ratio of inflammatory indicators, including Systemic Inflammatory Response Index (SIRI), C-reactive protein-to-lymphocyte ratio (CLR), eosinophil count-to-lymphocyte count ratio (ELR), were calculated. Univariate and multivariate logistic regression analyses were conducted to explore the association between the ratio of inflammatory indicators and TOPD. Furthermore, the relationship between the ratio of inflammatory indicators and TOPD was investigated using propensity score matching (PSM) and receiver operating characteristic (ROC) curve analysis was performed to evaluate their predictive value for TOPD. Results The present study included a total of 737 patients, of whom 83 participants (11.26%) had TOPD. Sixty-nine TOPD patients and 69 non-TOPD (NTOPD) patients were successfully matched. Univariate and multivariable logistics regression analysis, conducted before and after PSM, revealed that SIRI was independently significantly associated with an increased risk of TOPD. The area under curve (AUC) of SIRI were 0.702 and 0.668 before and after PSM, respectively. Additionally, patients were stratified into four different groups based on SIRI quartiles for further analysis. The prevalence of TOPD in TB patients showed an increase with higher SIRI values, both before and after PSM. Conclusion Levels of inflammatory indicators were higher in TOPD patients when compared to NTOPD patients. SIRI may be a simple and useful inflammatory index for assessing TOPD, and TB patients with higher values of SIRI are more likely to be high-risk group for TOPD.
Collapse
Affiliation(s)
- Shengling Hu
- Department of Infectious Diseases, Wuhan Jinyintan Hospital, Tongji Medical College of Huazhong University of Science and Technology; Hubei Clinical Research Center for Infectious Diseases; Wuhan Research Center for Communicable Disease Diagnosis and Treatment, Chinese Academy of Medical Sciences; Joint Laboratory of Infectious Diseases and Health, Wuhan Institute of Virology and Wuhan Jinyintan Hospital, Chinese Academy of Sciences, Wuhan, 430023, People’s Republic of China
| | - Qi Yu
- Department of Infectious Diseases, Wuhan Jinyintan Hospital, Tongji Medical College of Huazhong University of Science and Technology; Hubei Clinical Research Center for Infectious Diseases; Wuhan Research Center for Communicable Disease Diagnosis and Treatment, Chinese Academy of Medical Sciences; Joint Laboratory of Infectious Diseases and Health, Wuhan Institute of Virology and Wuhan Jinyintan Hospital, Chinese Academy of Sciences, Wuhan, 430023, People’s Republic of China
| | - Fenfang Liu
- Department of Infectious Diseases, Wuhan Jinyintan Hospital, Tongji Medical College of Huazhong University of Science and Technology; Hubei Clinical Research Center for Infectious Diseases; Wuhan Research Center for Communicable Disease Diagnosis and Treatment, Chinese Academy of Medical Sciences; Joint Laboratory of Infectious Diseases and Health, Wuhan Institute of Virology and Wuhan Jinyintan Hospital, Chinese Academy of Sciences, Wuhan, 430023, People’s Republic of China
| | - Fengyun Gong
- Department of Infectious Diseases, Wuhan Jinyintan Hospital, Tongji Medical College of Huazhong University of Science and Technology; Hubei Clinical Research Center for Infectious Diseases; Wuhan Research Center for Communicable Disease Diagnosis and Treatment, Chinese Academy of Medical Sciences; Joint Laboratory of Infectious Diseases and Health, Wuhan Institute of Virology and Wuhan Jinyintan Hospital, Chinese Academy of Sciences, Wuhan, 430023, People’s Republic of China
| |
Collapse
|
23
|
Choi H, Xu JF, Chotirmall SH, Chalmers JD, Morgan LC, Dhar R. Bronchiectasis in Asia: a review of current status and challenges. Eur Respir Rev 2024; 33:240096. [PMID: 39322263 PMCID: PMC11423131 DOI: 10.1183/16000617.0096-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 07/31/2024] [Indexed: 09/27/2024] Open
Abstract
Recent bronchiectasis studies from large-scale multinational, multicentre registries have demonstrated that the characteristics of the disease vary according to geographic region. However, most perspectives on bronchiectasis are dominated by data from Western countries. This review intends to provide an Asian perspective on the disease, focusing on the established registries in India, Korea and China. Asian patients with bronchiectasis are less likely to show female predominance and experience exacerbations, are more likely to be younger, have milder disease, and have fewer options for guideline-recommended treatment than those living in other global regions. Furthermore, Asian bronchiectasis patients demonstrate different comorbidities, microbiological profiles and unique endophenotypes, including post-tuberculosis and dry bronchiectasis. Notably, each Asian region reveals further geographic variations and inter-patient differences. Future studies are warranted to better characterise Asian patients with bronchiectasis.
Collapse
Affiliation(s)
- Hayoung Choi
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Internal Medicine, Hallym University Kangnam Sacred Heart Hospital, Hallym University College of Medicine, Seoul, Republic of Korea
| | - Jin-Fu Xu
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
- Institute of Respiratory Medicine, School of Medicine, Tongji University, Shanghai, China
| | - Sanjay H Chotirmall
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Department of Respiratory and Critical Care Medicine, Tan Tock Seng Hospital, Singapore, Singapore
| | - James D Chalmers
- Division of Molecular and Clinical Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, UK
| | - Lucy C Morgan
- Department of Respiratory Medicine Concord Hospital, Concord Clinical School University of Syndey, Sydney, Australia
| | - Raja Dhar
- Department of Pulmonology, CK Birla Group of Hospitals, Kolkata, India
| |
Collapse
|
24
|
Khan RMN, Ahn YM, Marriner GA, Via LE, D'Hooge F, Seo Lee S, Yang N, Basuli F, White AG, Tomko JA, Frye LJ, Scanga CA, Weiner DM, Sutphen ML, Schimel DM, Dayao E, Piazza MK, Gomez F, Dieckmann W, Herscovitch P, Mason NS, Swenson R, Kiesewetter DO, Backus KM, Geng Y, Raj R, Anthony DC, Flynn JL, Barry CE, Davis BG. Distributable, metabolic PET reporting of tuberculosis. Nat Commun 2024; 15:5239. [PMID: 38937448 PMCID: PMC11211441 DOI: 10.1038/s41467-024-48691-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 05/09/2024] [Indexed: 06/29/2024] Open
Abstract
Tuberculosis remains a large global disease burden for which treatment regimens are protracted and monitoring of disease activity difficult. Existing detection methods rely almost exclusively on bacterial culture from sputum which limits sampling to organisms on the pulmonary surface. Advances in monitoring tuberculous lesions have utilized the common glucoside [18F]FDG, yet lack specificity to the causative pathogen Mycobacterium tuberculosis (Mtb) and so do not directly correlate with pathogen viability. Here we show that a close mimic that is also positron-emitting of the non-mammalian Mtb disaccharide trehalose - 2-[18F]fluoro-2-deoxytrehalose ([18F]FDT) - is a mechanism-based reporter of Mycobacteria-selective enzyme activity in vivo. Use of [18F]FDT in the imaging of Mtb in diverse models of disease, including non-human primates, successfully co-opts Mtb-mediated processing of trehalose to allow the specific imaging of TB-associated lesions and to monitor the effects of treatment. A pyrogen-free, direct enzyme-catalyzed process for its radiochemical synthesis allows the ready production of [18F]FDT from the most globally-abundant organic 18F-containing molecule, [18F]FDG. The full, pre-clinical validation of both production method and [18F]FDT now creates a new, bacterium-selective candidate for clinical evaluation. We anticipate that this distributable technology to generate clinical-grade [18F]FDT directly from the widely-available clinical reagent [18F]FDG, without need for either custom-made radioisotope generation or specialist chemical methods and/or facilities, could now usher in global, democratized access to a TB-specific PET tracer.
Collapse
Affiliation(s)
- R M Naseer Khan
- Department of Chemistry, University of Oxford, Chemistry Research Laboratory, Oxford, UK
- Clinical Pharmacology Lab, Clinical Center, NIHBC, NIH, Bethesda, MD, USA
| | - Yong-Mo Ahn
- Tuberculosis Research Section, Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research (DIR), National Institute of Allergy and Infectious Disease (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Gwendolyn A Marriner
- Tuberculosis Research Section, Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research (DIR), National Institute of Allergy and Infectious Disease (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Laura E Via
- Tuberculosis Research Section, Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research (DIR), National Institute of Allergy and Infectious Disease (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
- Tuberculosis Imaging Program, DIR, NIAID, NIH, Bethesda, MD, USA
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Francois D'Hooge
- Department of Chemistry, University of Oxford, Chemistry Research Laboratory, Oxford, UK
| | - Seung Seo Lee
- Department of Chemistry, University of Oxford, Chemistry Research Laboratory, Oxford, UK
- School of Chemistry, University of Southampton, Southampton, UK
| | - Nan Yang
- Department of Chemistry, University of Oxford, Chemistry Research Laboratory, Oxford, UK
- The Rosalind Franklin Institute, Oxfordshire, UK
| | - Falguni Basuli
- Chemistry and Synthesis Center, NHLBI, NIH, Bethesda, MD, USA
| | - Alexander G White
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, USA
| | - Jaime A Tomko
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, USA
| | - L James Frye
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, USA
| | - Charles A Scanga
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, USA
| | - Danielle M Weiner
- Tuberculosis Research Section, Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research (DIR), National Institute of Allergy and Infectious Disease (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Michelle L Sutphen
- Tuberculosis Research Section, Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research (DIR), National Institute of Allergy and Infectious Disease (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Daniel M Schimel
- Tuberculosis Research Section, Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research (DIR), National Institute of Allergy and Infectious Disease (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Emmanuel Dayao
- Tuberculosis Research Section, Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research (DIR), National Institute of Allergy and Infectious Disease (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | | | - Felipe Gomez
- Tuberculosis Imaging Program, DIR, NIAID, NIH, Bethesda, MD, USA
| | - William Dieckmann
- Positron Emission Tomography Department, Clinical Center, NIH, Bethesda, MD, USA
| | - Peter Herscovitch
- Positron Emission Tomography Department, Clinical Center, NIH, Bethesda, MD, USA
| | - N Scott Mason
- Department of Radiology, University of Pittsburgh, Pittsburgh, USA
| | - Rolf Swenson
- Chemistry and Synthesis Center, NHLBI, NIH, Bethesda, MD, USA
| | - Dale O Kiesewetter
- Molecular Tracer and Imaging Core Facility, National Institute of Biomedical Imaging and Bioengineering, NIH, Bethesda, MD, USA
| | - Keriann M Backus
- Department of Chemistry, University of Oxford, Chemistry Research Laboratory, Oxford, UK
- Biological Chemistry Department, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Yiqun Geng
- Department of Chemistry, University of Oxford, Chemistry Research Laboratory, Oxford, UK
| | - Ritu Raj
- Department of Chemistry, University of Oxford, Chemistry Research Laboratory, Oxford, UK
| | | | - JoAnne L Flynn
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, USA
| | - Clifton E Barry
- Tuberculosis Research Section, Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research (DIR), National Institute of Allergy and Infectious Disease (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA.
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa.
| | - Benjamin G Davis
- Department of Chemistry, University of Oxford, Chemistry Research Laboratory, Oxford, UK.
- The Rosalind Franklin Institute, Oxfordshire, UK.
- Department of Pharmacology, University of Oxford, Oxford, UK.
| |
Collapse
|
25
|
Malakar B, Barth V, Puffal J, Woychik N, Husson RN. Phosphorylation of VapB antitoxins affects intermolecular interactions to regulate VapC toxin activity in Mycobacterium tuberculosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.30.596101. [PMID: 38853858 PMCID: PMC11160731 DOI: 10.1101/2024.05.30.596101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Toxin-antitoxin modules are present in many bacterial pathogens. The VapBC family is particularly abundant in members of the Mycobacterium tuberculosis complex, with 50 modules present in the M. tuberculosis genome. In type IIA modules the VapB antitoxin protein binds to and inhibits the activity of the co-expressed cognate VapC toxin protein. VapB proteins also bind to promoter region sequences and repress expression of the vapB-vapC operon. Though VapB-VapC interactions can control the amount of free VapC toxin in the bacterial cell, the mechanisms that affect this interaction are poorly understood. Based on our recent finding of Ser/Thr phosphorylation of VapB proteins in M. tuberculosis, we substituted phosphomimetic or phosphoablative amino acids at the phosphorylation sites of two VapB proteins. We found that phosphomimetic substitution of VapB27 and VapB46 resulted in decreased interaction with their respective cognate VapC proteins, whereas phosphoablative substitution did not alter binding. Similarly, we determined that phosphomimetic substitution interfered with VapB binding to promoter region DNA sequences. Both decreased VapB-VapC interaction and decreased VapB repression of vapB-vapC operon transcription would result in increased free VapC in the M. tuberculosis cell. M. tuberculosis strains expressing vapB46-vapC46 constructs containing a phosphoablative vapB mutation resulted in lower toxicity compared to a strain expressing native vapB46, whereas similar or greater toxicity was observed in the strain expressing the phosphomimetic vapB mutation. These results identify a novel mechanism by which VapC toxicity activity can be regulated by VapB phosphorylation, potentially in response to extracytoplasmic as well as intracellular signals.
Collapse
Affiliation(s)
- Basanti Malakar
- Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Valdir Barth
- Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Julia Puffal
- Department of Biochemistry and Molecular Biology, Rutgers University, Robert Wood Johnson Medical School, Piscataway, NJ, USA
| | - Nancy Woychik
- Department of Biochemistry and Molecular Biology, Rutgers University, Robert Wood Johnson Medical School, Piscataway, NJ, USA
| | - Robert N. Husson
- Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
26
|
Espinosa-Pereiro J, Alagna R, Saluzzo F, González-Moreno J, Heinrich N, Sánchez-Montalvá A, Cirillo DM. A Systematic Review of Potential Biomarkers for Bacterial Burden and Treatment Efficacy Assessment in Tuberculosis Platform-Based Clinical Trials. J Infect Dis 2024; 229:1584-1595. [PMID: 37956107 DOI: 10.1093/infdis/jiad482] [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: 06/05/2023] [Revised: 09/28/2023] [Accepted: 10/27/2023] [Indexed: 11/15/2023] Open
Abstract
Adaptive platform trials can be more efficient than classic trials for developing new treatments. Moving from culture-based to simpler- or faster-to-measure biomarkers as efficacy surrogates may enhance this advantage. We performed a systematic review of treatment efficacy biomarkers in adults with tuberculosis. Platform trials can span different development phases. We grouped biomarkers as: α, bacterial load estimates used in phase 2a trials; β, early and end-of treatment end points, phase 2b-c trials; γ, posttreatment or trial-level estimates, phase 2c-3 trials. We considered as analysis unit (biomarker entry) each combination of biomarker, predicted outcome, and their respective measurement times or intervals. Performance metrics included: sensitivity, specificity, area under the receiver-operator curve (AUC), and correlation measures, and classified as poor, promising, or good. Eighty-six studies included 22 864 participants. From 1356 biomarker entries, 318 were reported with the performance metrics of interest, with 103 promising and 41 good predictors. Group results were: α, mycobacterial RNA and lipoarabinomannan (LAM) in sputum, and host metabolites in urine; β, mycobacterial RNA and host transcriptomic or cytokine signatures for early treatment response; and γ, host transcriptomics for recurrence. A combination of biomarkers from different categories could help in designing more efficient platform trials. Efforts to develop efficacy surrogates should be better coordinated.
Collapse
Affiliation(s)
- Juan Espinosa-Pereiro
- Infectious Diseases Department, Vall d'Hebrón University Hospital, Universitat Autónoma de Barcelona, Barcelona, Spain
- International Health Program, Catalan Institute of Health, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infeccioass, Instituto de Salud Carlos III, Madrid, Spain
| | - Riccardo Alagna
- San Raffaele Scientific Institute, Milan, Italy
- Qiagen, Srl, Milan, Italy
| | | | | | - Norbert Heinrich
- Center for International Health, University Hospital, Ludwig Maximilian University Munich, Munich, Germany
- German Center for Infection Research, Munich, Germany
- Division of Infectious Diseases and Tropical Medicine, University Hospital, Ludwig Maximilian University Munich (DZIF), Partner Site Munich, Munich, Germany
| | - Adrián Sánchez-Montalvá
- Infectious Diseases Department, Vall d'Hebrón University Hospital, Universitat Autónoma de Barcelona, Barcelona, Spain
- International Health Program, Catalan Institute of Health, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infeccioass, Instituto de Salud Carlos III, Madrid, Spain
- Grupo de Estudio de Micobacterias, Sociedad Española de Enfermedades Infecciosas y Microbiología Clínica, Madrid, Spain
| | | |
Collapse
|
27
|
Choi H, Shin J, Jung JH, Han K, Choi W, Lee HR, Yoo JE, Yeo Y, Lee H, Shin DW. Tuberculosis and osteoporotic fracture risk: development of individualized fracture risk estimation prediction model using a nationwide cohort study. Front Public Health 2024; 12:1358010. [PMID: 38721534 PMCID: PMC11076769 DOI: 10.3389/fpubh.2024.1358010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 04/08/2024] [Indexed: 05/15/2024] Open
Abstract
Purpose Tuberculosis (TB) is linked to sustained inflammation even after treatment, and fracture risk is higher in TB survivors than in the general population. However, no individualized fracture risk prediction model exists for TB survivors. We aimed to estimate fracture risk, identify fracture-related factors, and develop an individualized risk prediction model for TB survivors. Methods TB survivors (n = 44,453) between 2010 and 2017 and 1:1 age- and sex-matched controls were enrolled. One year after TB diagnosis, the participants were followed-up until the date of fracture, death, or end of the study period (December 2018). Cox proportional hazard regression analyses were performed to compare the fracture risk between TB survivors and controls and to identify fracture-related factors among TB survivors. Results During median 3.4 (interquartile range, 1.6-5.3) follow-up years, the incident fracture rate was significantly higher in TB survivors than in the matched controls (19.3 vs. 14.6 per 1,000 person-years, p < 0.001). Even after adjusting for potential confounders, TB survivors had a higher risk for all fractures (adjusted hazard ratio 1.27 [95% confidence interval 1.20-1.34]), including hip (1.65 [1.39-1.96]) and vertebral (1.35 [1.25-1.46]) fractures, than matched controls. Fracture-related factors included pulmonary TB, female sex, older age, heavy alcohol consumption, reduced exercise, and a higher Charlson Comorbidity Index (p < 0.05). The individualized fracture risk model showed good discrimination (concordance statistic = 0.678). Conclusion TB survivors have a higher fracture risk than matched controls. An individualized prediction model may help prevent fractures in TB survivors, especially in high-risk groups.
Collapse
Affiliation(s)
- Hayoung Choi
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Internal Medicine, Hallym University Kangnam Sacred Heart Hospital, Seoul, Republic of Korea
| | - Jungeun Shin
- International Healthcare Center, Samsung Medical Center, Seoul, Republic of Korea
| | - Jin-Hyung Jung
- Department of Biostatistics, College of Medicine, Catholic University of Korea, Seoul, Republic of Korea
| | - Kyungdo Han
- Department of Statistics and Actuarial Science, Soongsil University, Seoul, Republic of Korea
| | - Wonsuk Choi
- Department of Internal Medicine, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Hwasun, Republic of Korea
| | - Han Rim Lee
- International Healthcare Center, Samsung Medical Center, Seoul, Republic of Korea
- Department of Family Medicine and Supportive Care Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jung Eun Yoo
- Department of Family Medicine, Healthcare System Gangnam Center, Seoul National University Hospital, Seoul, Republic of Korea
| | - Yohwan Yeo
- Department of Family Medicine, Hallym University Dongtan Sacred Heart Hospital, Hallym University College of Medicine, Hwaseong, Republic of Korea
| | - Hyun Lee
- Division of Pulmonary Medicine and Allergy, Department of Internal Medicine, Hanyang Medical Center, Hanyang University College of Medicine, Seoul, Republic of Korea
| | - Dong Wook Shin
- Department of Family Medicine, Supportive Care Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Department of Clinical Research Design and Evaluation, Samsung Advanced Institute for Health Science and Technology (SAIHST), Sungkyunkwan University, Seoul, Republic of Korea
| |
Collapse
|
28
|
Cross GB, O’ Doherty J, Chang CC, Kelleher AD, Paton NI. Does PET-CT Have a Role in the Evaluation of Tuberculosis Treatment in Phase 2 Clinical Trials? J Infect Dis 2024; 229:1229-1238. [PMID: 37788578 PMCID: PMC11011169 DOI: 10.1093/infdis/jiad425] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 09/10/2023] [Accepted: 10/01/2023] [Indexed: 10/05/2023] Open
Abstract
Positron emission tomography-computed tomography (PET-CT) has the potential to revolutionize research in infectious diseases, as it has done with cancer. There is growing interest in it as a biomarker in the setting of early-phase tuberculosis clinical trials, particularly given the limitations of current biomarkers as adequate predictors of sterilizing cure for tuberculosis. PET-CT is a real-time tool that provides a 3-dimensional view of the spatial distribution of tuberculosis within the lung parenchyma and the nature of lesions with uptake (ie, whether nodular, consolidative, or cavitary). Its ability to provide functional data on changes in metabolism, drug penetration, and immune control of tuberculous lesions has the potential to facilitate drug development and regimen selection for advancement to phase 3 trials in tuberculosis. In this narrative review, we discuss the role that PET-CT may have in evaluating responses to drug therapy in active tuberculosis treatment and the challenges in taking PET-CT forward as predictive biomarker of relapse-free cure in the setting of phase 2 clinical trials.
Collapse
Affiliation(s)
- Gail B Cross
- Immunovirology and Pathogenesis Program, The Kirby Institute, UNSW, Sydney
- Burnet Institute, Victoria, Australia
| | - Jim O’ Doherty
- Siemens Medical Solutions, Malvern, PA
- Department of Radiology and Radiological Science, Medical University of South Carolina, Charleston, SC, USA
- Radiography & Diagnostic Imaging, University College Dublin, Dublin, Ireland
| | - Christina C Chang
- Department of Infectious Diseases, Alfred Hospital and Central Clinical School, Monash University, Melbourne, Victoria, Australia
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa
| | - Anthony D Kelleher
- Immunovirology and Pathogenesis Program, The Kirby Institute, UNSW, Sydney
- St Vincent's Hospital, Sydney, Australia
| | - Nicholas I Paton
- Infectious Disease Translational Research Programme, National University of Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- London School of Hygiene and Tropical Medicine, London, UK
| |
Collapse
|
29
|
Dinkele R, Gessner S, Patterson B, McKerry A, Hoosen Z, Vazi A, Seldon R, Koch A, Warner DF, Wood R. Persistent Mycobacterium tuberculosis bioaerosol release in a tuberculosis-endemic setting. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.04.02.24305196. [PMID: 38633787 PMCID: PMC11023659 DOI: 10.1101/2024.04.02.24305196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/19/2024]
Abstract
Pioneering studies linking symptomatic disease and cough-mediated release of Mycobacterium tuberculosis (Mtb) established the infectious origin of tuberculosis (TB), simultaneously informing the pervasive notion that pathology is a prerequisite for Mtb transmission. Our prior work has challenged this assumption: by sampling TB clinic attendees, we detected equivalent release of Mtb-containing bioaerosols by confirmed TB patients and individuals not receiving a TB diagnosis, and we demonstrated a time-dependent reduction in Mtb bioaerosol positivity during six-months' follow-up, irrespective of anti-TB chemotherapy. Now, by extending bioaerosol sampling to a randomly selected community cohort, we show that Mtb release is common in a TB-endemic setting: of 89 participants, 79.8% (71/89) produced Mtb bioaerosols independently of QuantiFERON-TB Gold status, a standard test for Mtb infection; moreover, during two-months' longitudinal sampling, only 2% (1/50) were serially Mtb bioaerosol negative. These results necessitate a reframing of the prevailing paradigm of Mtb transmission and infection, and may explain the current inability to elucidate Mtb transmission networks in TB-endemic regions.
Collapse
Affiliation(s)
- Ryan Dinkele
- UCT Molecular Mycobacteriology Research Unit, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, 7925, South Africa
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, 7925, South Africa
| | - Sophia Gessner
- UCT Molecular Mycobacteriology Research Unit, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, 7925, South Africa
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, 7925, South Africa
| | - Benjamin Patterson
- Amsterdam Institute for Global Health and Development, University of Amsterdam, Amsterdam, 1105, The Netherlands
| | - Andrea McKerry
- Aerobiology and TB Research Unit, Desmond Tutu Health Foundation, Cape Town, 7925, South Africa
| | - Zeenat Hoosen
- Aerobiology and TB Research Unit, Desmond Tutu Health Foundation, Cape Town, 7925, South Africa
| | - Andiswa Vazi
- Aerobiology and TB Research Unit, Desmond Tutu Health Foundation, Cape Town, 7925, South Africa
| | - Ronnett Seldon
- Aerobiology and TB Research Unit, Desmond Tutu Health Foundation, Cape Town, 7925, South Africa
| | - Anastasia Koch
- UCT Molecular Mycobacteriology Research Unit, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, 7925, South Africa
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, 7925, South Africa
| | - Digby F. Warner
- UCT Molecular Mycobacteriology Research Unit, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, 7925, South Africa
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, 7925, South Africa
- Wellcome Centre for Infectious Diseases Research in Africa, Faculty of Health Sciences, University of Cape Town, Cape Town, 7925, South Africa
| | - Robin Wood
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, 7925, South Africa
- Aerobiology and TB Research Unit, Desmond Tutu Health Foundation, Cape Town, 7925, South Africa
| |
Collapse
|
30
|
Fortune SM. The Titanic question in TB control: Should we worry about the bummock? Proc Natl Acad Sci U S A 2024; 121:e2403321121. [PMID: 38527210 PMCID: PMC10998566 DOI: 10.1073/pnas.2403321121] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2024] Open
Affiliation(s)
- Sarah M. Fortune
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA02115
| |
Collapse
|
31
|
MacLean ELH, Zimmer AJ, den Boon S, Gupta-Wright A, Cirillo DM, Cobelens F, Gillespie SH, Nahid P, Phillips PP, Ruhwald M, Denkinger CM. Tuberculosis treatment monitoring tests during routine practice: study design guidance. Clin Microbiol Infect 2024; 30:481-488. [PMID: 38182047 DOI: 10.1016/j.cmi.2023.12.027] [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: 09/15/2023] [Revised: 12/10/2023] [Accepted: 12/25/2023] [Indexed: 01/07/2024]
Abstract
SCOPE The current tools for tuberculosis (TB) treatment monitoring, smear microscopy and culture, cannot accurately predict poor treatment outcomes. Research into new TB treatment monitoring tools (TMTs) is growing, but data are unreliable. In this article, we aim to provide guidance for studies investigating and evaluating TB TMT for use during routine clinical care. Here, a TB TMT would guide treatment during the course of therapy, rather than testing for a cure at the regimen's end. This article does not cover the use of TB TMTs as surrogate endpoints in the clinical trial context. METHODS Guidelines were initially informed by experiences during a systematic review of TB TMTs. Subsequently, a small content expert group was consulted for feedback on initial recommendations. After revision, feedback from substantive experts across sectors was sought. QUESTIONS ADDRESSED BY THE GUIDELINE AND RECOMMENDATIONS The proposed considerations and recommendations for studies evaluating TB TMTs for use during the treatment in routine clinical care fall into eight domains. We provide specific recommendations regarding study design and recruitment, outcome definitions, reference standards, participant follow-up, clinical setting, study population, treatment regimen reporting, and index tests and data presentation. Overall, TB TMTs should be evaluated in a manner similar to diagnostic tests, but TB TMT accuracy must be assessed at multiple timepoints throughout the treatment course, and TB TMTs should be evaluated in study populations who have already received a diagnosis of TB. Study design and outcome definitions must be aligned with the developmental phase of the TB TMT under evaluation. There is no reference standard for TB treatment response, so different reference standards and comparator tests have been proposed, the selection of which will vary depending on the developmental phase of the TMT under assessment. The use of comparator tests can assist in generating evidence. Clarity is required when reporting of timepoints, TMT read-outs, and analysis results. Implementing these recommendations will lead to higher quality TB TMT studies that will allow data to be meaningfully compared, thereby facilitating the development of novel tools to guide individual TB therapy and improve treatment outcomes.
Collapse
Affiliation(s)
- Emily Lai-Ho MacLean
- NHMRC Clinical Trials Centre, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Alexandra J Zimmer
- Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, QC, Canada
| | - Saskia den Boon
- Global Tuberculosis Programme, World Health Organization, Geneva, Switzerland
| | | | - Daniela M Cirillo
- Emerging Bacterial Pathogens Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Frank Cobelens
- Department of Global Health and Amsterdam Institute for Global Health and Development, Amsterdam University Medical Centers Location, University of Amsterdam, Amsterdam, The Netherlands
| | - Stephen H Gillespie
- Division of Infection and Global Health, School of Medicine, University of St Andrews, St Andrews, UK
| | - Payam Nahid
- Center for Tuberculosis, University of California San Francisco, San Francisco, CA, USA
| | - Patrick P Phillips
- Center for Tuberculosis, University of California San Francisco, San Francisco, CA, USA
| | | | - Claudia M Denkinger
- Division of Clinical Tropical Medicine, Center of Infectious Diseases, Heidelberg University Hospital, Heidelberg, Germany; Center of Infection Research (DZIF), Partners Site Heidelberg, Heidelberg, Germany.
| |
Collapse
|
32
|
Kim T, Choi H, Kim SH, Yang B, Han K, Jung JH, Kim BG, Park DW, Moon JY, Kim SH, Kim TH, Yoon HJ, Shin DW, Lee H. Increased Risk of Incident Chronic Obstructive Pulmonary Disease and Related Hospitalizations in Tuberculosis Survivors: A Population-Based Matched Cohort Study. J Korean Med Sci 2024; 39:e105. [PMID: 38529575 DOI: 10.3346/jkms.2024.39.e105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 01/29/2024] [Indexed: 03/27/2024] Open
Abstract
BACKGROUND Tuberculosis (TB) survivors have an increased risk of developing chronic obstructive pulmonary disease (COPD). This study assessed the risk of COPD development and COPD-related hospitalization in TB survivors compared to controls. METHODS We conducted a population-based cohort study of TB survivors and 1:1 age- and sex-matched controls using data from the Korean National Health Insurance Service database collected from 2010 to 2017. We compared the risk of COPD development and COPD-related hospitalization between TB survivors and controls. RESULTS Of the subjects, 9.6% developed COPD, and 2.8% experienced COPD-related hospitalization. TB survivors had significantly higher COPD incidence rates (36.7/1,000 vs. 18.8/1,000 person-years, P < 0.001) and COPD-related hospitalization (10.7/1,000 vs. 4.3/1,000 person-years, P < 0.001) than controls. Multivariable Cox regression analyses revealed higher risks of COPD development (adjusted hazard ratio [aHR], 1.63; 95% confidence interval [CI], 1.54-1.73) and COPD-related hospitalization (aHR, 2.03; 95% CI, 1.81-2.27) in TB survivors. Among those who developed COPD, the hospitalization rate was higher in individuals with post-TB COPD compared to those with non-TB COPD (10.7/1,000 vs. 4.9/1,000 person-years, P < 0.001), showing an increased risk of COPD-related hospitalization (aHR, 1.84; 95% CI, 1.17-2.92). CONCLUSION TB survivors had higher risks of incident COPD and COPD-related hospitalization compared to controls. These results suggest that previous TB is an important COPD etiology associated with COPD-related hospitalization.
Collapse
Affiliation(s)
- Taehee Kim
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Internal Medicine, Hallym University Kangnam Sacred Heart Hospital, Hallym University College of Medicine, Seoul, Korea
| | - Hayoung Choi
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Internal Medicine, Hallym University Kangnam Sacred Heart Hospital, Hallym University College of Medicine, Seoul, Korea
| | - Sang Hyuk Kim
- Deparment of Internal Medicine, Dongguk University Gyeongju Hospital, Dongguk Univiersity College of Medicine, Gyeongju, Korea
| | - Bumhee Yang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Chungbuk National University Hospital, Chungbuk National University College of Medicine, Cheongju, Korea
| | - Kyungdo Han
- Department of Statistics and Actuarial Science, Soongsil University, Seoul, Korea
| | - Jin-Hyung Jung
- Department of Biostatistics, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Bo-Guen Kim
- Division of Pulmonary Medicine and Allergy, Department of Internal Medicine, Hanyang University College of Medicine, Seoul, Korea
| | - Dong Won Park
- Division of Pulmonary Medicine and Allergy, Department of Internal Medicine, Hanyang University College of Medicine, Seoul, Korea
| | - Ji Yong Moon
- Division of Pulmonary Medicine and Allergy, Department of Internal Medicine, Hanyang University College of Medicine, Seoul, Korea
| | - Sang-Heon Kim
- Division of Pulmonary Medicine and Allergy, Department of Internal Medicine, Hanyang University College of Medicine, Seoul, Korea
| | - Tae-Hyung Kim
- Division of Pulmonary Medicine and Allergy, Department of Internal Medicine, Hanyang University College of Medicine, Seoul, Korea
| | - Ho Joo Yoon
- Division of Pulmonary Medicine and Allergy, Department of Internal Medicine, Hanyang University College of Medicine, Seoul, Korea
| | - Dong Wook Shin
- Department of Family Medicine/Supportive Care Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
- Department of Clinical Research Design & Evaluation, Samsung Advanced Institute for Health Science & Technology (SAIHST), Sungkyunkwan University, Seoul, Korea.
| | - Hyun Lee
- Division of Pulmonary Medicine and Allergy, Department of Internal Medicine, Hanyang University College of Medicine, Seoul, Korea.
| |
Collapse
|
33
|
Patterson B, Dinkele R, Gessner S, Koch A, Hoosen Z, January V, Leonard B, McKerry A, Seldon R, Vazi A, Hermans S, Cobelens F, Warner DF, Wood R. Aerosolization of viable Mycobacterium tuberculosis bacilli by tuberculosis clinic attendees independent of sputum-Xpert Ultra status. Proc Natl Acad Sci U S A 2024; 121:e2314813121. [PMID: 38470917 PMCID: PMC10962937 DOI: 10.1073/pnas.2314813121] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 02/01/2024] [Indexed: 03/14/2024] Open
Abstract
Potential Mycobacterium tuberculosis (Mtb) transmission during different pulmonary tuberculosis (TB) disease states is poorly understood. We quantified viable aerosolized Mtb from TB clinic attendees following diagnosis and through six months' follow-up thereafter. Presumptive TB patients (n=102) were classified by laboratory, radiological, and clinical features into Group A: Sputum-Xpert Ultra-positive TB (n=52), Group B: Sputum-Xpert Ultra-negative TB (n=20), or Group C: TB undiagnosed (n=30). All groups were assessed for Mtb bioaerosol release at baseline, and subsequently at 2 wk, 2 mo, and 6 mo. Groups A and B were notified to the national TB program and received standard anti-TB chemotherapy; Mtb was isolated from 92% and 90% at presentation, 87% and 74% at 2 wk, 54% and 44% at 2 mo and 32% and 20% at 6 mo, respectively. Surprisingly, similar numbers were detected in Group C not initiating TB treatment: 93%, 70%, 48% and 22% at the same timepoints. A temporal association was observed between Mtb bioaerosol release and TB symptoms in all three groups. Persistence of Mtb bioaerosol positivity was observed in ~30% of participants irrespective of TB chemotherapy. Captured Mtb bacilli were predominantly acid-fast stain-negative and poorly culturable; however, three bioaerosol samples yielded sufficient biomass following culture for whole-genome sequencing, revealing two different Mtb lineages. Detection of viable aerosolized Mtb in clinic attendees, independent of TB diagnosis, suggests that unidentified Mtb transmitters might contribute a significant attributable proportion of community exposure. Additional longitudinal studies with sputum culture-positive and -negative control participants are required to investigate this possibility.
Collapse
Affiliation(s)
- Benjamin Patterson
- Amsterdam Institute for Global Health and Development, University of Amsterdam, Amsterdam1105, The Netherlands
| | - Ryan Dinkele
- South African Medical Research Council, National Health Laboratory Service, University of Cape Town Molecular Mycobacteriology Research Unit & Department of Science and Technology/National Research Foundation Centre of Excellence for Biomedical TB Research, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town7925, South Africa
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town7925, South Africa
| | - Sophia Gessner
- South African Medical Research Council, National Health Laboratory Service, University of Cape Town Molecular Mycobacteriology Research Unit & Department of Science and Technology/National Research Foundation Centre of Excellence for Biomedical TB Research, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town7925, South Africa
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town7925, South Africa
| | - Anastasia Koch
- South African Medical Research Council, National Health Laboratory Service, University of Cape Town Molecular Mycobacteriology Research Unit & Department of Science and Technology/National Research Foundation Centre of Excellence for Biomedical TB Research, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town7925, South Africa
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town7925, South Africa
| | - Zeenat Hoosen
- Aerobiology and TB Research Unit, Desmond Tutu Health Foundation, Cape Town7975, South Africa
| | - Vanessa January
- Aerobiology and TB Research Unit, Desmond Tutu Health Foundation, Cape Town7975, South Africa
| | - Bryan Leonard
- Aerobiology and TB Research Unit, Desmond Tutu Health Foundation, Cape Town7975, South Africa
| | - Andrea McKerry
- Aerobiology and TB Research Unit, Desmond Tutu Health Foundation, Cape Town7975, South Africa
| | - Ronnett Seldon
- Aerobiology and TB Research Unit, Desmond Tutu Health Foundation, Cape Town7975, South Africa
| | - Andiswa Vazi
- Aerobiology and TB Research Unit, Desmond Tutu Health Foundation, Cape Town7975, South Africa
| | - Sabine Hermans
- Amsterdam Institute for Global Health and Development, University of Amsterdam, Amsterdam1105, The Netherlands
| | - Frank Cobelens
- Amsterdam Institute for Global Health and Development, University of Amsterdam, Amsterdam1105, The Netherlands
| | - Digby F. Warner
- South African Medical Research Council, National Health Laboratory Service, University of Cape Town Molecular Mycobacteriology Research Unit & Department of Science and Technology/National Research Foundation Centre of Excellence for Biomedical TB Research, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town7925, South Africa
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town7925, South Africa
- Wellcome Centre for Infectious Diseases Research in Africa, Faculty of Health Sciences, University of Cape Town, Cape Town7925, South Africa
| | - Robin Wood
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town7925, South Africa
- Aerobiology and TB Research Unit, Desmond Tutu Health Foundation, Cape Town7975, South Africa
| |
Collapse
|
34
|
Nieuwenhuizen NE, Nouailles G, Sutherland JS, Zyla J, Pasternack AH, Heyckendorf J, Frye BC, Höhne K, Zedler U, Bandermann S, Abu Abed U, Brinkmann V, Gutbier B, Witzenrath M, Suttorp N, Zissel G, Lange C, Ritvos O, Kaufmann SHE, the CAPNETZ Study group, the DZIF TB study group. Activin A levels are raised during human tuberculosis and blockade of the activin signaling axis influences murine responses to M. tuberculosis infection. mBio 2024; 15:e0340823. [PMID: 38376260 PMCID: PMC10936190 DOI: 10.1128/mbio.03408-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 01/26/2024] [Indexed: 02/21/2024] Open
Abstract
Activin A strongly influences immune responses; yet, few studies have examined its role in infectious diseases. We measured serum activin A levels in two independent tuberculosis (TB) patient cohorts and in patients with pneumonia and sarcoidosis. Serum activin A levels were increased in TB patients compared to healthy controls, including those with positive tuberculin skin tests, and paralleled severity of disease, assessed by X-ray scores. In pneumonia patients, serum activin A levels were also raised, but in sarcoidosis patients, levels were lower. To determine whether blockade of the activin A signaling axis could play a functional role in TB, we harnessed a soluble activin type IIB receptor fused to human IgG1 Fc, ActRIIB-Fc, as a ligand trap in a murine TB model. The administration of ActRIIB-Fc to Mycobacterium tuberculosis-infected mice resulted in decreased bacterial loads and increased numbers of CD4 effector T cells and tissue-resident memory T cells in the lung. Increased frequencies of tissue-resident memory T cells corresponded with downregulated T-bet expression in lung CD4 and CD8 T cells. Altogether, the results suggest a disease-exacerbating role of ActRIIB signaling pathways. Serum activin A may be useful as a biomarker for diagnostic triage of active TB or monitoring of anti-tuberculosis therapy. IMPORTANCE Tuberculosis remains the leading cause of death by a bacterial pathogen. The etiologic agent of tuberculosis, Mycobacterium tuberculosis, can remain dormant in the infected host for years before causing disease. Significant effort has been made to identify biomarkers that can discriminate between latently infected and actively diseased individuals. We found that serum levels of the cytokine activin A were associated with increased lung pathology and could discriminate between active tuberculosis and tuberculin skin-test-positive healthy controls. Activin A signals through the ActRIIB receptor, which can be blocked by administration of the ligand trap ActRIIB-Fc, a soluble activin type IIB receptor fused to human IgG1 Fc. In a murine model of tuberculosis, we found that ActRIIB-Fc treatment reduced mycobacterial loads. Strikingly, ActRIIB-Fc treatment significantly increased the number of tissue-resident memory T cells. These results suggest a role for ActRIIB signaling pathways in host responses to Mycobacterium tuberculosis and activin A as a biomarker of ongoing disease.
Collapse
Affiliation(s)
- Natalie E. Nieuwenhuizen
- Department of Immunology, Max Planck Institute for Infection Biology, Chariteplatz, Berlin, Germany
- Institute for Hygiene and Microbiology, Julius Maximilian University of Würzburg, Würzburg, Germany
| | - Geraldine Nouailles
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Jayne S. Sutherland
- Vaccines and Immunity Theme, Medical Research Council Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Joanna Zyla
- Department of Data Science and Engineering, Silesian University of Technology, Gliwice, Poland
| | - Arja H. Pasternack
- Department of Physiology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Jan Heyckendorf
- Department of Medicine I, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Björn C. Frye
- Department of Pneumology, Clinic, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Kerstin Höhne
- Department of Pneumology, Clinic, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ulrike Zedler
- Department of Immunology, Max Planck Institute for Infection Biology, Chariteplatz, Berlin, Germany
| | - Silke Bandermann
- Department of Immunology, Max Planck Institute for Infection Biology, Chariteplatz, Berlin, Germany
| | - Ulrike Abu Abed
- Microscopy Core Facility, Max Planck Institute for Infection Biology, Chariteplatz, Berlin, Germany
| | - Volker Brinkmann
- Microscopy Core Facility, Max Planck Institute for Infection Biology, Chariteplatz, Berlin, Germany
| | - Birgitt Gutbier
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Martin Witzenrath
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- CAPNETZ STIFTUNG, Hannover, Germany
- German Center for Lung Research (DZL), Berlin, Germany
| | - Norbert Suttorp
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- CAPNETZ STIFTUNG, Hannover, Germany
- German Center for Lung Research (DZL), Berlin, Germany
| | - Gernot Zissel
- Department of Pneumology, Clinic, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Christoph Lange
- Division of Clinical Infectious Diseases, Research Center Borstel, Borstel, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Borstel, Germany
- Respiratory Medicine and International Health, University of Lübeck, Lübeck, Germany
- Baylor College of Medicine and Texas Children´s Hospital, Global TB Program, Houston, Texas, USA
| | - Olli Ritvos
- Department of Physiology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Stefan H. E. Kaufmann
- Department of Immunology, Max Planck Institute for Infection Biology, Chariteplatz, Berlin, Germany
- Max Planck Institute for Multidisciplinary Sciences, Emeritus Group Systems Immunology, Göttingen, Germany
- Hagler Institute for Advanced Study, Texas A&M University, College Station, Texas, USA
| | - the CAPNETZ Study group
- Department of Immunology, Max Planck Institute for Infection Biology, Chariteplatz, Berlin, Germany
- Institute for Hygiene and Microbiology, Julius Maximilian University of Würzburg, Würzburg, Germany
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Vaccines and Immunity Theme, Medical Research Council Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
- Department of Data Science and Engineering, Silesian University of Technology, Gliwice, Poland
- Department of Physiology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Medicine I, University Hospital Schleswig-Holstein, Kiel, Germany
- Department of Pneumology, Clinic, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Microscopy Core Facility, Max Planck Institute for Infection Biology, Chariteplatz, Berlin, Germany
- CAPNETZ STIFTUNG, Hannover, Germany
- German Center for Lung Research (DZL), Berlin, Germany
- Division of Clinical Infectious Diseases, Research Center Borstel, Borstel, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Borstel, Germany
- Respiratory Medicine and International Health, University of Lübeck, Lübeck, Germany
- Baylor College of Medicine and Texas Children´s Hospital, Global TB Program, Houston, Texas, USA
- Max Planck Institute for Multidisciplinary Sciences, Emeritus Group Systems Immunology, Göttingen, Germany
- Hagler Institute for Advanced Study, Texas A&M University, College Station, Texas, USA
| | - the DZIF TB study group
- Department of Immunology, Max Planck Institute for Infection Biology, Chariteplatz, Berlin, Germany
- Institute for Hygiene and Microbiology, Julius Maximilian University of Würzburg, Würzburg, Germany
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Vaccines and Immunity Theme, Medical Research Council Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
- Department of Data Science and Engineering, Silesian University of Technology, Gliwice, Poland
- Department of Physiology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Medicine I, University Hospital Schleswig-Holstein, Kiel, Germany
- Department of Pneumology, Clinic, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Microscopy Core Facility, Max Planck Institute for Infection Biology, Chariteplatz, Berlin, Germany
- CAPNETZ STIFTUNG, Hannover, Germany
- German Center for Lung Research (DZL), Berlin, Germany
- Division of Clinical Infectious Diseases, Research Center Borstel, Borstel, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Borstel, Germany
- Respiratory Medicine and International Health, University of Lübeck, Lübeck, Germany
- Baylor College of Medicine and Texas Children´s Hospital, Global TB Program, Houston, Texas, USA
- Max Planck Institute for Multidisciplinary Sciences, Emeritus Group Systems Immunology, Göttingen, Germany
- Hagler Institute for Advanced Study, Texas A&M University, College Station, Texas, USA
| |
Collapse
|
35
|
Sehgal IS, Dhooria S, Muthu V, Salzer HJF, Agarwal R. Burden, clinical features, and outcomes of post-tuberculosis chronic obstructive lung diseases. Curr Opin Pulm Med 2024; 30:156-166. [PMID: 37902135 DOI: 10.1097/mcp.0000000000001026] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2023]
Abstract
PURPOSE OF REVIEW Post-tuberculosis lung disease (PTLD) is an increasingly recognized and debilitating consequence of pulmonary tuberculosis (PTB). In this review, we provide a comprehensive overview of PTLD with airflow obstruction (PTLD-AFO), focusing on its burden, pathophysiology, clinical manifestations, diagnostic methods, and management strategies. RECENT FINDINGS The relationship between PTLD and airflow obstruction is complex and multifactorial. Approximately 60% of the patients with PTLD have some spirometric abnormality. Obstruction is documented in 18-22% of PTLD patients. The host susceptibility and host response to mycobacterium drive the pathogenic mechanism of PTLD. A balance between inflammatory, anti-inflammatory, and fibrotic pathways decides whether an individual with PTB would have PTLD after microbiological cure. An obstructive abnormality in PTLD-AFO is primarily due to destruction of bronchial walls, aberrant healing, and reduction of mucosal glands. The most common finding on computed tomography (CT) of thorax in patients with PTLD-AFO is bronchiectasis and cavitation. Therefore, the 'Cole's vicious vortex' described in bronchiectasis applies to PTLD. A multidisciplinary approach is required for diagnosis and treatment. The disability-adjusted life-years (DALYs) attributed to PTLD represent about 50% of the total estimated burden of DALYs due to tuberculosis (TB). Patients with PTLD require comprehensive care that includes psychosocial support, pulmonary rehabilitation, and vaccination against respiratory pathogens. In the absence of trials evaluating different treatments for PTLD-AFO, therapy is primarily symptomatic. SUMMARY PTLD with airflow obstruction has considerable burden and causes a significant morbidity and mortality. However, many aspects of PTLD-AFO still need to be answered. Studies are required to evaluate different phenotypes, especially concerning Aspergillus -related complications. The treatment should be personalized based on the predominant phenotype of airflow obstruction. Extensive studies to understand the exact burden, pathogenesis, and treatment of PTBLD-AFO are needed.
Collapse
Affiliation(s)
- Inderpaul Singh Sehgal
- Department of Pulmonary Medicine, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, Punjab, India
| | - Sahajal Dhooria
- Department of Pulmonary Medicine, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, Punjab, India
| | - Valliappan Muthu
- Department of Pulmonary Medicine, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, Punjab, India
| | - Helmut J F Salzer
- Division of Infectious Diseases and Tropical Medicine, Department of Internal Medicine 4 - Pneumology, Kepler University Hospital
- Medical Faculty, Johannes Kepler University Linz, Linz
- Ignaz-Semmelweis-Institute, Interuniversity Institute for Infection Research, Vienna, Austria
| | - Ritesh Agarwal
- Department of Pulmonary Medicine, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, Punjab, India
| |
Collapse
|
36
|
Alonso-Rodríguez N, Vianello E, van Veen S, Jenum S, Tonby K, van Riessen R, Lai X, Mortensen R, Ottenhoff THM, Dyrhol-Riise AM. Whole blood RNA signatures in tuberculosis patients receiving H56:IC31 vaccine as adjunctive therapy. Front Immunol 2024; 15:1350593. [PMID: 38433842 PMCID: PMC10904528 DOI: 10.3389/fimmu.2024.1350593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 01/25/2024] [Indexed: 03/05/2024] Open
Abstract
Introduction Therapeutic vaccination in tuberculosis (TB) represents a Host Directed Therapy strategy which enhances immune responses in order to improve clinical outcomes and shorten TB treatment. Previously, we have shown that the subunit H56:IC31 vaccine induced both humoral and cellular immune responses when administered to TB patients adjunctive to standard TB treatment (TBCOX2 study, NCT02503839). Here we present the longitudinal whole blood gene expression patterns in H56:IC31 vaccinated TB patients compared to controls receiving standard TB treatment only. Methods The H56:IC31 group (N=11) and Control group (N=7) underwent first-line TB treatment for 182 days. The H56:IC31 group received 5 micrograms of the H56:IC31 vaccine (Statens Serum Institut; SSI, Valneva Austria GmbH) intramuscularly at day 84 and day 140. Total RNA was extracted from whole blood samples collected in PAXgene tubes on days 0, 84, 98, 140, 154, 182 and 238. The expression level of 183 immune-related genes was measured by high-throughput microfluidic qPCR (Biomark HD system, Standard BioTools). Results The targeted gene expression profiling unveiled the upregulation of modules such as interferon (IFN) signalling genes, pattern recognition receptors and small nucleotide guanosine triphosphate (GTP)-ases in the vaccinated group compared to controls two weeks after administration of the first H56:IC31 vaccine. Additionally, the longitudinal analysis of the Adolescent Cohort Study-Correlation of Risk (ACS-COR) signature showed a progressive downregulation in both study arms towards the end of TB treatment, in congruence with reported treatment responses and clinical improvements. Still, two months after the end of TB treatment, vaccinated patients, and especially those developing both cellular and humoral vaccine responses, showed a lower expression of the ACS-COR genes compared to controls. Discussion Our data report gene expression patterns following H56:IC31 vaccination which might be interpreted as a lower risk of relapse in therapeutically vaccinated patients. Further studies are needed to conclude if these gene expression patterns could be used as prognostic biosignatures for therapeutic TB vaccine responses.
Collapse
Affiliation(s)
| | - Eleonora Vianello
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Suzanne van Veen
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Synne Jenum
- Department of Infectious Diseases, Oslo University Hospital, Oslo, Norway
| | - Kristian Tonby
- Department of Infectious Diseases, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Rosalie van Riessen
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Xiaoran Lai
- Oslo Centre for Biostatistics and Epidemiology, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Rasmus Mortensen
- Deptartment of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Tom H. M. Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Anne Ma Dyrhol-Riise
- Department of Infectious Diseases, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
37
|
Neuböck MJ, Günther G, Barac A, Davidsen JR, Laursen CB, Agarwal R, Sehgal IS, Lange C, Salzer HJF. Chronic Pulmonary Aspergillosis as a Considerable Complication in Post-Tuberculosis Lung Disease. Semin Respir Crit Care Med 2024; 45:102-113. [PMID: 38196060 DOI: 10.1055/s-0043-1776913] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2024]
Abstract
Post-tuberculosis lung disease (PTLD) has only recently been put in the spotlight as a medical entity. Recent data suggest that up to 50% of tuberculosis (TB) patients are left with PTLD-related impairment after completion of TB treatment. The presence of residual cavities in the lung is the largest risk factor for the development of chronic pulmonary aspergillosis (CPA) globally. Diagnosis of CPA is based on four criteria including a typical radiological pattern, evidence of Aspergillus species, exclusion of alternative diagnosis, and a chronic course of disease. In this manuscript, we provide a narrative review on CPA as a serious complication for patients with PTLD.
Collapse
Affiliation(s)
- Matthias J Neuböck
- Division of Infectious Diseases and Tropical Medicine, Department of Internal Medicine 4 - Pneumology, Kepler University Hospital, Linz, Austria
- Medical Faculty, Johannes Kepler University Linz, Linz, Austria
| | - Gunar Günther
- Department of Pulmonology and Allergology, Inselspital, Bern University Hospital, Bern, Switzerland
- Department of Medical Sciences, Faculty of Health Sciences, University of Namibia, Windhoek, Namibia
| | - Aleksandra Barac
- Clinic for Infectious and Tropical Diseases, University Clinical Center of Serbia, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Jesper R Davidsen
- Department of Respiratory Medicine, Pulmonary Aspergillosis Centre Denmark, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, Odense Respiratory Research Unit, University of Southern Denmark, Odense, Denmark
| | - Christian B Laursen
- Department of Respiratory Medicine, Pulmonary Aspergillosis Centre Denmark, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, Odense Respiratory Research Unit, University of Southern Denmark, Odense, Denmark
| | - Ritesh Agarwal
- Department of Pulmonary Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Inderpaul S Sehgal
- Department of Pulmonary Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Christoph Lange
- Division of Clinical Infectious Diseases, Research Center Borstel, Borstel, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
- Respiratory Medicine and International Health, University of Lübeck, Lübeck, Germany
- Baylor College of Medicine and Texas Children's Hospital, Global Tuberculosis Program, Houston, Texas
| | - Helmut J F Salzer
- Division of Infectious Diseases and Tropical Medicine, Department of Internal Medicine 4 - Pneumology, Kepler University Hospital, Linz, Austria
- Medical Faculty, Johannes Kepler University Linz, Linz, Austria
- Ignaz-Semmelweis-Institute, Interuniversity Institute for Infection Research, Vienna, Austria
| |
Collapse
|
38
|
Malefane L, Maarman G. Post-tuberculosis lung disease and inflammatory role players: can we characterise the myriad inflammatory pathways involved to gain a better understanding? Chem Biol Interact 2024; 387:110817. [PMID: 38006959 DOI: 10.1016/j.cbi.2023.110817] [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: 08/21/2023] [Revised: 10/31/2023] [Accepted: 11/20/2023] [Indexed: 11/27/2023]
Abstract
Tuberculosis (TB) remains a global health threat, and even after successful TB treatment, a subset of patients develops serious long-term lung impairments, recently termed post-tuberculosis lung disease (PTLD). Much remains to be discovered, as PTLD as a post-TB disease is a developing field, still in its infancy. The pathogenesis of PTLD is not fully elucidated but has been linked to elevated inflammatory pathways. The complexity of PTLD makes it challenging to pinpoint the specific inflammatory pathways involved in its pathophysiology. Therefore, this paper provides a comprehensive review of inflammatory cytokines and their potential roles in PLTD, with a specific focus on interleukin 6 (IL-6), IL-1, IL-8, tumour necrosis factor-alpha (TNF-α), transforming growth factor beta (TGF-β) and C-Reactive Protein (CRP). We delve into PTLD pathology, discuss its impact on lung function and review risk factors for PTLD. In addition, we summarise the current gaps in knowledge, provide recommendations for measuring inflammatory biomarkers and propose potential directions for future studies. We propose that future studies measure a wide range of inflammatory markers in TB populations with and without PTLD. In addition, studies could isolate peripheral blood mononuclear cells from patient blood to try and identify possible impairments that could be correlated with a PTLD diagnosis. Given that the PTLD field is still in an early stage of development, a comprehensive inflammatory analysis may help to know which pathways are key in PTLD development, and this may ultimately help to predict patients who are at risk. More research is warranted.
Collapse
Affiliation(s)
- Lindiwe Malefane
- CARMA: Centre for Cardio-Metabolic Research in Africa, Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine & Health Sciences, Stellenbosch University, Cape Town, 8000, South Africa
| | - Gerald Maarman
- CARMA: Centre for Cardio-Metabolic Research in Africa, Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine & Health Sciences, Stellenbosch University, Cape Town, 8000, South Africa.
| |
Collapse
|
39
|
Cupido G, Günther G. Post tuberculosis lung disease and tuberculosis sequelae: A narrative review. Indian J Tuberc 2024; 71:64-72. [PMID: 38296392 DOI: 10.1016/j.ijtb.2023.04.001] [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: 07/11/2022] [Revised: 03/28/2023] [Accepted: 04/05/2023] [Indexed: 02/08/2024]
Abstract
Post Tuberculosis lung disease (PTLD) and post tuberculosis sequelae is a global and poorly recognized problem, amplified by social factors and immunocompromising conditions, inadequate treatment, lack of effective prevention of tuberculosis (TB) infection and disease. As a disease, it remained until recently poorly defined, with studies heterogenous with regards to regions, population demographics, risk factors, cohort sizes, and methods. Pathophysiologically, even successfully treated pulmonary TB disease has sequelae i.e. involving central and peripheral airways, lung parenchyma and pleura, resulting in airway narrowing and dilatation, fibrocavitation and emphysema, pulmonary vascular changes as well as pleural fibrosis. Functionally patients have airflow limitation, restrictive disease or a mixture of both not rarely associated with respiratory, or even ventilatory failure. Quality of life is often impaired through disability, TB relapse, superinfections and through increased susceptibility to reinfection and persistent inflammation, leading to progressive lung function decline and an increased risk of cardiovascular disease and cancer. Premature mortality due to PTLD is very likely, but poorly described.
Collapse
Affiliation(s)
- Gordon Cupido
- Department of Internal Medicine, Katutura State Hospital, Windhoek, Namibia.
| | - Gunar Günther
- Department of Pulmonology and Allergology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland; Department of Medical Sciences, University of Namibia, School of Medicine, Windhoek, Namibia
| |
Collapse
|
40
|
Krug S, Gupta M, Kumar P, Feller L, Ihms EA, Kang BG, Srikrishna G, Dawson TM, Dawson VL, Bishai WR. Inhibition of host PARP1 contributes to the anti-inflammatory and antitubercular activity of pyrazinamide. Nat Commun 2023; 14:8161. [PMID: 38071218 PMCID: PMC10710439 DOI: 10.1038/s41467-023-43937-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 11/24/2023] [Indexed: 12/18/2023] Open
Abstract
The antibiotic pyrazinamide (PZA) is a cornerstone of tuberculosis (TB) therapy that shortens treatment durations by several months despite being only weakly bactericidal. Intriguingly, PZA is also an anti-inflammatory molecule shown to specifically reduce inflammatory cytokine signaling and lesion activity in TB patients. However, the target and clinical importance of PZA's host-directed activity during TB therapy remain unclear. Here, we identify the host enzyme Poly(ADP-ribose) Polymerase 1 (PARP1), a pro-inflammatory master regulator strongly activated in TB, as a functionally relevant host target of PZA. We show that PZA inhibits PARP1 enzymatic activity in macrophages and in mice where it reverses TB-induced PARP1 activity in lungs to uninfected levels. Utilizing a PZA-resistant mutant, we demonstrate that PZA's immune-modulatory effects are PARP1-dependent but independent of its bactericidal activity. Importantly, PZA's bactericidal efficacy is impaired in PARP1-deficient mice, suggesting that immune modulation may be an integral component of PZA's antitubercular activity. In addition, adjunctive PARP1 inhibition dramatically reduces inflammation and lesion size in mice and may be a means to reduce lung damage and shorten TB treatment duration. Together, these findings provide insight into PZA's mechanism of action and the therapeutic potential of PARP1 inhibition in the treatment of TB.
Collapse
Affiliation(s)
- Stefanie Krug
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Manish Gupta
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Pankaj Kumar
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Laine Feller
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Elizabeth A Ihms
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Bong Gu Kang
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Geetha Srikrishna
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ted M Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Valina L Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - William R Bishai
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
41
|
Ahimbisibwe G, Nakibuule M, Martin Ssejoba M, Oyamo D, Mulwana R, Nabulime J, Babirye F, Kizito MA, Lekuya HM, Adakun AS, Nalumansi D, Muryasingura S, Lukande R, Kyazze A, Baluku JB, Biraro IA, Cose S. Feasibility and acceptability of undertaking postmortem studies for tuberculosis medical research in a low income country. Front Immunol 2023; 14:1264351. [PMID: 38130719 PMCID: PMC10734300 DOI: 10.3389/fimmu.2023.1264351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 11/01/2023] [Indexed: 12/23/2023] Open
Abstract
Introduction If we are to break new ground in difficult-to-treat or difficult-to-vaccinate diseases (such as HIV, malaria, or tuberculosis), we must have a better understanding of the immune system at the site of infection in humans. For tuberculosis (TB), the initial site of infection is the lungs, but obtaining lung tissues from subjects suffering from TB has been limited to bronchoalveolar lavage (BAL) or sputum sampling, or surgical resection of diseased lung tissue. Methods We examined the feasibility of undertaking a postmortem study for human tuberculosis research at Mulago National Referral Hospital in Kampala, Uganda. Results Postmortem studies give us an opportunity to compare TB-involved and -uninvolved sites, for both diseased and non-diseased individuals. We report good acceptability of the next-of-kin to consent for their relative's tissue to be used for medical research; that postmortem and tissue processing can be undertaken within 8 hours following death; and that immune cells remain viable and functional up to 14 hours after death. Discussion Postmortem procedures remain a valuable and essential tool both to establish cause of death, and to advance our medical and scientific understanding of infectious diseases.
Collapse
Affiliation(s)
- Gift Ahimbisibwe
- Medical Research Council (MRC)/Uganda Virus Research Institute (UVRI) and London School of Hygiene and Tropical Medicine (LSHTM) Uganda Research Unit, Entebbe, Uganda
| | - Marjorie Nakibuule
- Medical Research Council (MRC)/Uganda Virus Research Institute (UVRI) and London School of Hygiene and Tropical Medicine (LSHTM) Uganda Research Unit, Entebbe, Uganda
| | - Marvin Martin Ssejoba
- Medical Research Council (MRC)/Uganda Virus Research Institute (UVRI) and London School of Hygiene and Tropical Medicine (LSHTM) Uganda Research Unit, Entebbe, Uganda
| | - David Oyamo
- Mulago National Referral Hospital, Kampala, Uganda
| | - Rose Mulwana
- Mulago National Referral Hospital, Kampala, Uganda
| | | | | | | | | | | | | | | | - Robert Lukande
- Department of Pathology, Makerere University, Kampala, Uganda
| | - Andrew Kyazze
- Division of Pulmonology, Kiruddu National Referral Hospital, Kampala, Uganda
| | | | - Irene Andia Biraro
- Division of Pulmonology, Kiruddu National Referral Hospital, Kampala, Uganda
- Department of Internal Medicine, Makerere University, Kampala, Uganda
| | - Stephen Cose
- Medical Research Council (MRC)/Uganda Virus Research Institute (UVRI) and London School of Hygiene and Tropical Medicine (LSHTM) Uganda Research Unit, Entebbe, Uganda
- Clinical Research Department, London School of Hygiene and Tropical Medicine, London, United Kingdom
| |
Collapse
|
42
|
Wang L, Ma H, Wen Z, Niu L, Chen X, Liu H, Zhang S, Xu J, Zhu Y, Li H, Chen H, Shi L, Wan L, Li L, Li M, Wong KW, Song Y. Single-cell RNA-sequencing reveals heterogeneity and intercellular crosstalk in human tuberculosis lung. J Infect 2023; 87:373-384. [PMID: 37690670 DOI: 10.1016/j.jinf.2023.09.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 08/21/2023] [Accepted: 09/06/2023] [Indexed: 09/12/2023]
Abstract
Lung inflammation indicated by 18F-labeled fluorodeoxyglucose (FDG) in patients with tuberculosis is associated with disease severity and relapse risk upon treatment completion. We revealed the heterogeneity and intercellular crosstalk in lung tissues with 18F-FDG avidity and adjacent uninvolved tissues from 6 tuberculosis patients by single-cell RNA-sequencing. Tuberculous lungs had an influx of regulatory T cells (Treg), exhausted CD8 T cells, immunosuppressive myeloid cells, conventional DC, plasmacytoid DC, and neutrophils. Immune cells in inflamed lungs showed general up-regulation of ATP synthesis and interferon-mediated signaling. Immunosuppressive myeloid and Treg cells strongly displayed transcriptions of genes related to tuberculosis disease progression. Intensive crosstalk between IL4I1-expressing myeloid cells and Treg cells involving chemokines, costimulatory molecules, and immune checkpoints, some of which are specific in 18F-FDG-avid lungs, were found. Our analysis provides insights into the transcriptomic heterogeneity and cellular crosstalk in pulmonary tuberculosis and guides unveiling cellular and molecular targets for tuberculosis therapy.
Collapse
Affiliation(s)
- Lin Wang
- Department of Thoracic Surgery, Shanghai Public Health Clinical Center, Shanghai, China
| | - Hui Ma
- Department of Scientific Research, Shanghai Public Health Clinical Center, Shanghai, China
| | - Zilu Wen
- Department of Scientific Research, Shanghai Public Health Clinical Center, Shanghai, China
| | - Liangfei Niu
- Department of Scientific Research, Shanghai Public Health Clinical Center, Shanghai, China
| | - Xinchun Chen
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University School of Medicine, Shenzhen, China
| | - Haiying Liu
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, and Center for Tuberculosis Research, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shulin Zhang
- Department of Scientific Research, Shanghai Public Health Clinical Center, Shanghai, China
| | - Jianqing Xu
- Department of Scientific Research, Shanghai Public Health Clinical Center, Shanghai, China
| | - Yijun Zhu
- Department of Thoracic Surgery, Shanghai Public Health Clinical Center, Shanghai, China
| | - Hongwei Li
- Department of Thoracic Surgery, Shanghai Public Health Clinical Center, Shanghai, China
| | - Hui Chen
- Department of Thoracic Surgery, Shanghai Public Health Clinical Center, Shanghai, China
| | - Lei Shi
- Department of Thoracic Surgery, Shanghai Public Health Clinical Center, Shanghai, China
| | - Laiyi Wan
- Department of Thoracic Surgery, Shanghai Public Health Clinical Center, Shanghai, China
| | - Leilei Li
- Department of Thoracic Surgery, Shanghai Public Health Clinical Center, Shanghai, China
| | - Meiyi Li
- Fudan Zhangjiang Institute, Fudan University, Shanghai, China.
| | - Ka-Wing Wong
- Department of Scientific Research, Shanghai Public Health Clinical Center, Shanghai, China.
| | - Yanzheng Song
- Department of Thoracic Surgery, Shanghai Public Health Clinical Center, Shanghai, China.
| |
Collapse
|
43
|
Martinez-Martinez YB, Huante MB, Chauhan S, Naqvi KF, Bharaj P, Endsley JJ. Helper T cell bias following tuberculosis chemotherapy identifies opportunities for therapeutic vaccination to prevent relapse. NPJ Vaccines 2023; 8:165. [PMID: 37898618 PMCID: PMC10613213 DOI: 10.1038/s41541-023-00761-4] [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: 01/29/2023] [Accepted: 10/09/2023] [Indexed: 10/30/2023] Open
Abstract
Therapeutic vaccines have promise as adjunctive treatment for tuberculosis (TB) or as preventives against TB relapse. An important development challenge is the limited understanding of T helper (Th) cell roles during these stages of disease. A murine model of TB relapse was used to identify changes in Th populations and cytokine microenvironment. Active TB promoted expansion of Th1, Th2, Th17, and Th22 cells and cytokines in the lung. Following drug therapy, pulmonary Th17 and Th22 cells contracted, Th1 cells remained elevated, while Th cells producing IL-4 or IL-10 expanded. At relapse, Th22 cells failed to re-expand in the lung despite a moderate re-expansion of Th1 and Th17 cells and an increase in Th cytokine polyfunctionality. The dynamics of Th populations further differed by tissue compartment and disease presentation. These outcomes identify immune bias by Th subpopulations during TB relapse as candidate mechanisms for pathogenesis and targets for therapeutic vaccination.
Collapse
Affiliation(s)
- Yazmin B Martinez-Martinez
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Matthew B Huante
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Sadhana Chauhan
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Kubra F Naqvi
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, 77555, USA
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Preeti Bharaj
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, 77555, USA
- Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ, 07103, USA
| | - Janice J Endsley
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, 77555, USA.
| |
Collapse
|
44
|
Günther G, Abu- Hussain N, Keller PM, Guler R, Mukasa SL, Wolmarans K, Thienemann F. To treat or not to treat tuberculosis -clinical decision making in patients with previous pulmonary tuberculosis using 18F-FDG PET/CT. Respir Med Case Rep 2023; 46:101932. [PMID: 38025249 PMCID: PMC10630663 DOI: 10.1016/j.rmcr.2023.101932] [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: 12/07/2022] [Accepted: 10/14/2023] [Indexed: 12/01/2023] Open
Abstract
Post-tuberculosis (TB) radiological changes and symptoms can mimic TB. PCR-based diagnostic tests can show positive results, suggesting the presence of Mycobacterium tuberculosis DNA in the absence of viable bacteria. We present a case with two episodes of previous TB. Despite workup including trace to low positive PCR results, after performing sputum analysis, bronchoalveolar lavage analysis, cyto-brush and 18F-FDG PET/CT guided transthoracic biopsy, no culturable mycobacteria were detected. 18F-FDG PET/CT showed a high metabolic activity of the biopsied lesions. More accurate strategies and tools in patients with previous TB and positive PCR results are required to make appropriate treatment decisions.
Collapse
Affiliation(s)
- Gunar Günther
- Department of Pulmonology and Allergology, Inselspital Bern, Bern University Hospital, University of Bern, Switzerland
- Department of Medical Sciences, School of Medicine, University of Namibia, Windhoek, Namibia
| | - Nebal Abu- Hussain
- Department of Pulmonology and Allergology, Inselspital Bern, Bern University Hospital, University of Bern, Switzerland
| | - Peter M. Keller
- Institute for Infectious Diseases, University of Bern, Bern, Switzerland
| | - Reto Guler
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, Cape Town, South Africa
- Institute of Infectious Diseases and Molecular Medicine (IDM), Department of Pathology, Division of Immunology, Faculty of Health Sciences, University of Cape Town, South Africa
- Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease and Molecular Medicine (IDM), Faculty of Health Sciences, University of Cape Town, South Africa
| | - Sandra L. Mukasa
- General Medicine & Global Health (GMGH), Cape Heart Institute, Faculty of Health Science, University of Cape Town, South Africa
- Department of Medicine, Faculty of Health Science, University of Cape Town, South Africa
| | - Karen Wolmarans
- General Medicine & Global Health (GMGH), Cape Heart Institute, Faculty of Health Science, University of Cape Town, South Africa
- Department of Medicine, Faculty of Health Science, University of Cape Town, South Africa
| | - Friedrich Thienemann
- General Medicine & Global Health (GMGH), Cape Heart Institute, Faculty of Health Science, University of Cape Town, South Africa
- Department of Medicine, Faculty of Health Science, University of Cape Town, South Africa
- Department of Internal Medicine, University of Zürich, Switzerland
| |
Collapse
|
45
|
Vargas R, Abbott L, Bower D, Frahm N, Shaffer M, Yu WH. Gene signature discovery and systematic validation across diverse clinical cohorts for TB prognosis and response to treatment. PLoS Comput Biol 2023; 19:e1010770. [PMID: 37471455 PMCID: PMC10393163 DOI: 10.1371/journal.pcbi.1010770] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 06/15/2023] [Indexed: 07/22/2023] Open
Abstract
While blood gene signatures have shown promise in tuberculosis (TB) diagnosis and treatment monitoring, most signatures derived from a single cohort may be insufficient to capture TB heterogeneity in populations and individuals. Here we report a new generalized approach combining a network-based meta-analysis with machine-learning modeling to leverage the power of heterogeneity among studies. The transcriptome datasets from 57 studies (37 TB and 20 viral infections) across demographics and TB disease states were used for gene signature discovery and model training and validation. The network-based meta-analysis identified a common 45-gene signature specific to active TB disease across studies. Two optimized random forest regression models, using the full or partial 45-gene signature, were then established to model the continuum from Mycobacterium tuberculosis infection to disease and treatment response. In model validation, using pooled multi-cohort datasets to mimic the real-world setting, the model provides robust predictive performance for incipient to active TB risk over a 2.5-year period with an AUROC of 0.85, 74.2% sensitivity, and 78.3% specificity, which approximates the minimum criteria (>75% sensitivity and >75% specificity) within the WHO target product profile for prediction of progression to TB. Moreover, the model strongly discriminates active TB from viral infection (AUROC 0.93, 95% CI 0.91-0.94). For treatment monitoring, the TB scores generated by the model statistically correlate with treatment responses over time and were predictive, even before treatment initiation, of standard treatment clinical outcomes. We demonstrate an end-to-end gene signature model development scheme that considers heterogeneity for TB risk estimation and treatment monitoring.
Collapse
Affiliation(s)
- Roger Vargas
- Bill & Melinda Gates Medical Research Institute, Cambridge, Massachusetts, United States of America
- Harvard University, Cambridge, Massachusetts, United States of America
| | - Liam Abbott
- Bill & Melinda Gates Medical Research Institute, Cambridge, Massachusetts, United States of America
| | - Daniel Bower
- Bill & Melinda Gates Medical Research Institute, Cambridge, Massachusetts, United States of America
| | - Nicole Frahm
- Bill & Melinda Gates Medical Research Institute, Cambridge, Massachusetts, United States of America
| | - Mike Shaffer
- Bill & Melinda Gates Medical Research Institute, Cambridge, Massachusetts, United States of America
| | - Wen-Han Yu
- Bill & Melinda Gates Medical Research Institute, Cambridge, Massachusetts, United States of America
| |
Collapse
|
46
|
Kayongo A, Nyiro B, Siddharthan T, Kirenga B, Checkley W, Lutaakome Joloba M, Ellner J, Salgame P. Mechanisms of lung damage in tuberculosis: implications for chronic obstructive pulmonary disease. Front Cell Infect Microbiol 2023; 13:1146571. [PMID: 37415827 PMCID: PMC10320222 DOI: 10.3389/fcimb.2023.1146571] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 06/05/2023] [Indexed: 07/08/2023] Open
Abstract
Pulmonary tuberculosis is increasingly recognized as a risk factor for COPD. Severe lung function impairment has been reported in post-TB patients. Despite increasing evidence to support the association between TB and COPD, only a few studies describe the immunological basis of COPD among TB patients following successful treatment completion. In this review, we draw on well-elaborated Mycobacterium tuberculosis-induced immune mechanisms in the lungs to highlight shared mechanisms for COPD pathogenesis in the setting of tuberculosis disease. We further examine how such mechanisms could be exploited to guide COPD therapeutics.
Collapse
Affiliation(s)
- Alex Kayongo
- Department of Medicine, Center for Emerging Pathogens, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States
- Department of Immunology and Molecular Biology, College of Health Sciences, Makerere University, Kampala, Uganda
- Makerere University College of Health Sciences, Lung Institute, Makerere University, Kampala, Uganda
| | - Brian Nyiro
- Department of Medicine, Center for Emerging Pathogens, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States
- Department of Immunology and Molecular Biology, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Trishul Siddharthan
- Division of Pulmonary and Critical Care Medicine, University of Miami, Miami, FL, United States
| | - Bruce Kirenga
- Makerere University College of Health Sciences, Lung Institute, Makerere University, Kampala, Uganda
| | - William Checkley
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, United States
- Center for Global Non-Communicable Disease Research and Training, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Moses Lutaakome Joloba
- Makerere University College of Health Sciences, Lung Institute, Makerere University, Kampala, Uganda
| | - Jerrold Ellner
- Department of Medicine, Center for Emerging Pathogens, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States
| | - Padmini Salgame
- Department of Medicine, Center for Emerging Pathogens, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States
| |
Collapse
|
47
|
Bea S, Lee H, Choi WS, Huh K, Jung J, Shin JY. Risk of mortality and clinical outcomes associated with healthcare delay among patients with tuberculosis. J Infect Public Health 2023; 16:1313-1321. [PMID: 37339564 DOI: 10.1016/j.jiph.2023.05.038] [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: 12/07/2022] [Revised: 05/03/2023] [Accepted: 05/31/2023] [Indexed: 06/21/2023] Open
Abstract
BACKGROUND To eliminate tuberculosis (TB), World Health Organization (WHO) initiated "The End TB Strategy" with the goal of a 95% reduction in deaths. While many resources are contributed to eradicating TB, a substantial number of TB patients are still unlikely to receive timely treatment. Thus, we aimed to measure healthcare delay and its association with clinical outcomes from 2013 to 2018. METHODS We conducted a retrospective cohort study using linked data of the National Tuberculosis Surveillance Registry and the health insurance claims data of South Korea. We included incident TB patients, and healthcare delay was defined as the period between the first medical visit with TB-related symptoms and the initiation of an anti-TB regimen. We described the distribution of healthcare delay, and the study population was classified into two groups with mean as a cutoff. The association between healthcare delay and clinical outcomes (all-cause mortality, pneumonia, progression to multi/extensively drug-resistant, intensive care unit admission, and mechanical ventilation use) was evaluated using the Cox proportional hazard model. Several stratified and sensitivity analyses were also conducted. RESULTS Among 39,747 patients with pulmonary TB, mean healthcare delay was 42.3 days and delayed and non-delayed groups, classified by mean (or average), were 10,680 (26.9%) and 29,067 (73.1%), respectively. Healthcare delay was associated with an increased risk of all-cause mortality (HR 1.10, 95% CI 1.03-1.17), pneumonia (HR 1.13, 95% CI 1.09-1.18), and mechanical ventilation use (HR 1.15, 95% CI 1.01-1.32). We also observed the duration-response of healthcare delay. Stratified analyses showed patients with respiratory diseases were at higher risk, and consistent results were observed in sensitivity analyses. CONCLUSIONS We observed a substantial number of patients experiencing healthcare delays, and it was associated with the deterioration of clinical outcomes. Our findings suggest that attention from authorities and healthcare professionals is needed to attenuate the preventable burden caused by TB through timely treatment.
Collapse
Affiliation(s)
- Sungho Bea
- School of Pharmacy, Sungkyunkwan University, Suwon, South Korea
| | - Hyesung Lee
- School of Pharmacy, Sungkyunkwan University, Suwon, South Korea; Department of Biohealth Regulatory Science, Sungkyunkwan University, Suwon, South Korea
| | - Won Suk Choi
- Division of Infectious Diseases, Department of Internal Medicine, Ansan Hospital, Korea University College of Medicine, Ansan, South Korea
| | - Kyungmin Huh
- Division of Infectious Diseases, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Jaehun Jung
- Department of Preventive Medicine, Gachon University College of Medicine, Incheon, Korea; Artificial Intelligence and Big-Data Convergence Center, Gil Medical Center, Gachon University College of Medicine, Incheon, Korea
| | - Ju-Young Shin
- Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University, Seoul, South Korea.
| |
Collapse
|
48
|
Krause R, Warren CM, Simmons JD, Rebeiro PF, Maruri F, Karim F, Sterling TR, Koethe JR, Leslie A, van der Heijden YF. Failure to decrease HbA1c levels following TB treatment is associated with elevated Th1/Th17 CD4+ responses. Front Immunol 2023; 14:1151528. [PMID: 37313404 PMCID: PMC10258338 DOI: 10.3389/fimmu.2023.1151528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 05/17/2023] [Indexed: 06/15/2023] Open
Abstract
Introduction The rising global burden of metabolic disease impacts the control of endemic tuberculosis (TB) in many regions, as persons with diabetes mellitus (DM) are up to three times more likely to develop active TB than those without DM. Active TB can also promote glucose intolerance during both acute infection and over a longer term, potentially driven by aspects of the immune response. Identifying patients likely to have persistent hyperglycemia following TB treatment would enable closer monitoring and care, and an improved understanding of underlying immunometabolic dysregulation. Methods We measured the relationship of plasma cytokine levels, T cell phenotypes and functional responses with the change in hemoglobin A1c (HbA1c) before and after treatment of pulmonary TB in a prospective observational cohort in Durban, South Africa. Participants were stratified based on stable/increased HbA1c (n = 16) versus decreased HbA1c (n = 46) levels from treatment initiation to 12 month follow-up. Results CD62 P-selectin was up- (1.5-fold) and IL-10 downregulated (0.85-fold) in plasma among individuals whose HbA1c remained stable/increased during TB treatment. This was accompanied by increased pro-inflammatory TB-specific IL-17 production (Th17). In addition, Th1 responses were upregulated in this group, including TNF-α production and CX3CR1 expression, with decreased IL-4 and IL-13 production. Finally, the TNF-α+ IFNγ+ CD8+ T cells were associated with stable/increased HbA1c. These changes were all significantly different in the stable/increased HbA1c relative to the decreased HbA1c group. Discussion Overall, these data suggest that patients with stable/increased HbA1c had an increased pro-inflammatory state. Persistent inflammation and elevated T cell activity in individuals with unresolved dysglycemia following TB treatment may indicate failure to fully resolve infection or may promote persistent dysglycemia in these individuals, and further studies are needed to explore potential mechanisms.
Collapse
Affiliation(s)
- Robert Krause
- Africa Health Research Institute (AHRI), Durban, South Africa
- College of Health Sciences, School of Laboratory Medicine & Medical Sciences, University of KwaZulu Natal, Durban, South Africa
| | - Christian M. Warren
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Joshua D. Simmons
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Peter F. Rebeiro
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, United States
- Department of Biostatistics, Vanderbilt University School of Medicine, Nashville, TN, United States
- Vanderbilt Tuberculosis Center, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Fernanda Maruri
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, United States
- Vanderbilt Tuberculosis Center, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Farina Karim
- Africa Health Research Institute (AHRI), Durban, South Africa
- College of Health Sciences, School of Laboratory Medicine & Medical Sciences, University of KwaZulu Natal, Durban, South Africa
- Vanderbilt Tuberculosis Center, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Timothy R. Sterling
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, United States
- Vanderbilt Tuberculosis Center, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - John R. Koethe
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Al Leslie
- Africa Health Research Institute (AHRI), Durban, South Africa
- College of Health Sciences, School of Laboratory Medicine & Medical Sciences, University of KwaZulu Natal, Durban, South Africa
- Division of Infection and Immunity, University College London, London, United Kingdom
| | - Yuri F. van der Heijden
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, United States
- Vanderbilt Tuberculosis Center, Vanderbilt University School of Medicine, Nashville, TN, United States
- The Aurum Institute, Johannesburg, South Africa
| |
Collapse
|
49
|
Huang W, Lee MKT, Sin ATK, Nazari RS, Chua SY, Sng LH. Evaluation of Xpert MTB/RIF ultra assay for detection of Mycobacterium tuberculosis and rifampicin resistance. Pathology 2023:S0031-3025(23)00116-2. [PMID: 37268484 DOI: 10.1016/j.pathol.2023.03.002] [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: 09/13/2022] [Revised: 01/05/2023] [Accepted: 03/14/2023] [Indexed: 06/04/2023]
Abstract
Tuberculosis (TB) is a public health challenge globally, and molecular testing is recommended to expedite diagnosis. Concerns that Xpert MTB/RIF assay (Xpert) may be less sensitive when testing paucibacillary samples led to the development of the Xpert MTB/RIF Ultra assay (Ultra). We evaluated the performance of Ultra against Xpert on clinical samples sent to the national reference laboratory in Singapore. In total, 149 samples collected between January 2019 and November 2020 were analysed. Mycobacterium tuberculosis complex (MTBC) was isolated from 55 cultures. Using culture as the reference standard, Ultra demonstrated higher sensitivity (96.4% vs 85.5%) and marginally lower specificity (88.3% vs 89.4%) compared to Xpert in the full cohort. When considering only paucibacillary specimens such as extrapulmonary and smear-negative samples, similar results were obtained. Reclassifying Ultra trace results (low levels of MTB are detected but no rifampicin resistant result is detected) as negative in the full cohort led to a decrease in sensitivity by 10.9% and a marginal increase in specificity by 1.1%. In instances of low bacillary load, Ultra also identified rifampicin resistance more accurately than Xpert, when corroborated against other methods such as broth microdilution, line probe assay and whole genome sequencing (WGS). One isolate tested rifampicin-resistant using Xpert and Ultra, but was phenotypically susceptible and WGS demonstrated the presence of the silent mutation Thr444Thr. Ultra is more sensitive than Xpert in the detection of MTBC and rifampicin resistance in our local setting. Nevertheless, the results of molecular testing should still be correlated with phenotypic studies.
Collapse
Affiliation(s)
- Wenjie Huang
- Central Tuberculosis Laboratory, Department of Microbiology, Singapore General Hospital, Singapore.
| | - Melody Kee Tai Lee
- Central Tuberculosis Laboratory, Department of Microbiology, Singapore General Hospital, Singapore
| | - Amanda Teo Kai Sin
- Central Tuberculosis Laboratory, Department of Microbiology, Singapore General Hospital, Singapore
| | | | - Syn Yu Chua
- Central Tuberculosis Laboratory, Department of Microbiology, Singapore General Hospital, Singapore
| | - Li-Hwei Sng
- Central Tuberculosis Laboratory, Department of Microbiology, Singapore General Hospital, Singapore
| |
Collapse
|
50
|
Quan S, Zou T, Duan L, Tian X, Wang Y, Zhu Y, Fang M, Shi Y, Wan C, Sun L, Shen A. Clinical Characteristics of Pulmonary Tuberculosis in Children Tested by Xpert MTB/RIF Ultra. Pediatr Infect Dis J 2023; 42:389-395. [PMID: 36854100 DOI: 10.1097/inf.0000000000003866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/02/2023]
Abstract
BACKGROUND The Xpert MTB/rifampicin Ultra (Xpert Ultra) assay improves the early diagnosis of active tuberculosis (TB) in children. Clinical evaluation is paramount for the interpretation of any positive Xpert Ultra test, especially those with low quantities of DNA. METHODS In this study, 391 children with suspected TB who were tested with Xpert Ultra were enrolled. The clinical characteristics and Xpert Ultra results were further analyzed. RESULTS The sensitivity and specificity of Xpert Ultra were 45.0% (149/331) and 96.7% (58/60), respectively. Children with higher semiquantitative scales of Xpert Ultra showed higher percentages of a positive MTB culture, positive acid-fast bacilli staining, severe type of disease, fever, cough and expectoration, a higher white blood cell count and higher C-reactive protein concentrations (all P < 0.01). Among 44 children with an Xpert Ultra trace result, there were no differences in clinical characteristics between confirmed cases and unconfirmed TB cases. CONCLUSIONS The prevalence of trace is relatively high and can be considered positive in paucibacillary children. Clinical presentations are associated with bacterial load quantified by Xpert Ultra. The interpretation of Xpert Ultra trace results based on clinical information is important for the diagnosis of TB.
Collapse
Affiliation(s)
- Shuting Quan
- From the National Clinical Research Center for Respiratory Diseases, National Key Discipline of Pediatrics, Capital Medical University, Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Tingting Zou
- Department of Pediatrics Infectious Diseases, West China Second Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Chengdu, China
| | - Li Duan
- Department of Pediatrics Infectious Diseases, The No. 1 People's Hospital of Liangshan Yizu Autonomous Prefecture, Liangshan, China
| | - Xue Tian
- From the National Clinical Research Center for Respiratory Diseases, National Key Discipline of Pediatrics, Capital Medical University, Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Yacui Wang
- From the National Clinical Research Center for Respiratory Diseases, National Key Discipline of Pediatrics, Capital Medical University, Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Yu Zhu
- Department of Pediatrics Infectious Diseases, West China Second Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Chengdu, China
| | - Min Fang
- Department of Pediatrics Infectious Diseases, The No. 1 People's Hospital of Liangshan Yizu Autonomous Prefecture, Liangshan, China
| | - Yan Shi
- Department of Pediatrics Infectious Diseases, The No. 1 People's Hospital of Liangshan Yizu Autonomous Prefecture, Liangshan, China
| | - Chaomin Wan
- Department of Pediatrics Infectious Diseases, West China Second Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Chengdu, China
| | - Lin Sun
- From the National Clinical Research Center for Respiratory Diseases, National Key Discipline of Pediatrics, Capital Medical University, Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Adong Shen
- From the National Clinical Research Center for Respiratory Diseases, National Key Discipline of Pediatrics, Capital Medical University, Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
- Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, China
| |
Collapse
|