1
|
Sweetser B, Nkereuwem E, Nakafeero J, Gomez M, Wambi P, Nsereko M, Andama A, Ernst JD, Cattamanchi A, Kampmann B, Jaganath D, Wobudeya E. A Prospective Evaluation of a Three-Gene Host Response Signature to Classify Tuberculosis Severity in Children. J Pediatric Infect Dis Soc 2025; 14:piaf041. [PMID: 40319382 DOI: 10.1093/jpids/piaf041] [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: 02/18/2025] [Accepted: 04/29/2025] [Indexed: 05/07/2025]
Abstract
BACKGROUND Children with non-severe TB may benefit from short-course treatment, but point-of-care tools are needed to stratify disease severity. We prospectively evaluated the Cepheid Xpert MTB-Host Response (HR) prototype cartridge for distinguishing TB severity in children with pulmonary TB (PTB) in The Gambia and Uganda. METHODS We included children <15 with microbiologically confirmed or clinically diagnosed unconfirmed PTB. Severity was defined using the World Health Organization (WHO) guidelines for a four-month, drug-susceptible regimen. Capillary or venous blood was tested with the HR cartridge for PCR-based detection of 3 mRNA genes and calculation of a TB score from cycle thresholds. We generated receiver operating characteristic curves with the TB score to classify severe TB and assessed if Xpert-HR could achieve the WHO target accuracy for treatment optimization (≥90% sensitivity, ≥70% specificity). RESULTS Among 106 children, the median age was 4 years (IQR 1-7), 56.6% were female, and 13.2% were living with HIV. In all children with PTB, Xpert-HR achieved an AUC of 0.67 (95% CI 0.55-0.78), with 89.3% sensitivity (95% CI 71.8-97.7) and 29.5% specificity (95% CI 19.7-40.9, cutoff ≤ -0.60). By confirmation status, Xpert-HR approached the target accuracy in children with Confirmed TB, with 62.5% specificity (95% CI 24.5-91.5) at 91.7% sensitivity (95% CI 61.5-99.8, cut-off ≤ -1.349). Among children with Unconfirmed TB, specificity was lower (24.3%, 95% CI 14.8-36.0) at 93.8% sensitivity (95% CI 69.8-99.8, cutoff ≤ -0.450). Target accuracy was almost achieved in children 5-9 regardless of confirmation status (100% sensitivity [95% CI 71.5-100], 66.7% specificity [95% CI 43.0-85.4], cutoff ≤ -1.35), but specificity (28.2%, 95% CI 18.6-39.5) was lower for children < 5 (92.9% sensitivity, 95% CI 76.5-99.1, cutoff ≤ -0.550). CONCLUSIONS Xpert-HR approached the target accuracy to stratify PTB severity in older children and those with Confirmed TB but had lower specificity in children with Unconfirmed TB. Child-specific signatures may be needed to improve performance in younger children with paucibacillary disease.
Collapse
Affiliation(s)
- Brittney Sweetser
- Division of Pulmonary Diseases & Critical Care Medicine, University of California, Irvine, Orange, United States
- Center for Tuberculosis, Institute for Global Health Sciences, University of California, San Francisco, San Francisco, United States
| | - Esin Nkereuwem
- Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Fajara, The Gambia
| | | | - Marie Gomez
- Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Fajara, The Gambia
| | | | | | | | - Joel D Ernst
- Division of Experimental Medicine, University of California, San Francisco, San Francisco, United States
- Center for Tuberculosis, Institute for Global Health Sciences, University of California, San Francisco, San Francisco, United States
| | - Adithya Cattamanchi
- Division of Pulmonary Diseases & Critical Care Medicine, University of California, Irvine, Orange, United States
- Center for Tuberculosis, Institute for Global Health Sciences, University of California, San Francisco, San Francisco, United States
| | - Beate Kampmann
- Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Fajara, The Gambia
- Charité Centre for Global Health, Institute of International Health, Berlin, Germany
| | - Devan Jaganath
- Center for Tuberculosis, Institute for Global Health Sciences, University of California, San Francisco, San Francisco, United States
- Division of Pediatric Infectious Diseases, University of California, San Francisco, San Francisco, United States
| | | |
Collapse
|
2
|
Wang X, Harper K, Sinha P, Johnson WE, Patil P. Analysis of the cross-study replicability of tuberculosis gene signatures using 49 curated human transcriptomic datasets. Tuberculosis (Edinb) 2025; 153:102649. [PMID: 40359654 DOI: 10.1016/j.tube.2025.102649] [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: 11/21/2024] [Revised: 02/20/2025] [Accepted: 05/06/2025] [Indexed: 05/15/2025]
Abstract
BACKGROUND Tuberculosis (TB) is the leading cause of infectious disease mortality worldwide. Numerous host blood-based gene expression signatures have been proposed in the literature as alternative tools for diagnosing TB infection. However, the generalizability of these signatures to different patient contexts is not well-characterized. There is a pressing need for a well-curated database of TB gene expression studies for the systematic assessment of existing and newly developed TB gene signatures. RESULTS We built curatedTBData, a manually-curated database of 49 human TB transcriptomic studies. This data resource is freely available through GitHub and as an R Bioconductor package that allows users to validate new and existing biomarkers without the challenges of harmonizing heterogeneous studies. We demonstrate the use of this data resource with cross-study comparisons for 72 human host blood-based TB gene signatures. For the comparison of subjects with active TB from healthy controls, 19 gene signatures had weighted mean AUC of 0.90 or greater, with the highest result of 0.94. In active TB disease versus latent TB infection, 7 gene signatures had weighted mean AUC of 0.90 or greater, with a maximum of 0.93. CONCLUSIONS The curatedTBData data package offers a comprehensive resource of curated human blood-based gene expression and clinically annotated data. This resource will facilitate the development of new signatures that are generalizable across cohorts or more applicable to specific subsets of patients.
Collapse
Affiliation(s)
- Xutao Wang
- Department of Biostatistics, Boston University School of Public Health, Boston, USA
| | - Katie Harper
- Division of Computational Biomedicine, Boston University School of Medicine, Boston, USA
| | - Pranay Sinha
- Section of Infectious Diseases, Department of Medicine, Boston University School of Medicine, Boston, USA
| | - W Evan Johnson
- Division of Infectious Disease, Center for Data Science, Rutgers New Jersey Medical School, New Jersey, USA
| | - Prasad Patil
- Department of Biostatistics, Boston University School of Public Health, Boston, USA.
| |
Collapse
|
3
|
Sutherland JS, van der Spuy GD, Shaw JA, Richardson T, Tjon Kon Fat EM, Gindeh A, Owolabi O, Thuong NTT, Van LH, Van NH, Thao DTT, Mayanja-Kizza H, Nsereko M, Namuganga A, Nalukwago S, Belisle J, Moreau E, Penn-Nicholson A, Thwaites G, Winter J, Dockrell HM, Scriba TJ, Stanley K, Smith B, Chegou NN, Malherbe ST, Geluk A, Corstjens P, Walzl G. Performance of 2 Finger-Stick Blood Tests to Triage Adults With Symptoms of Pulmonary Tuberculosis: A Prospective Multisite Diagnostic Accuracy Study. Clin Infect Dis 2025:ciaf105. [PMID: 40237453 DOI: 10.1093/cid/ciaf105] [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: 12/09/2024] [Indexed: 04/18/2025] Open
Abstract
BACKGROUND Non-sputum-based, point-of-care triage tests for pulmonary tuberculosis could enhance tuberculosis diagnostic programs. We assessed the diagnostic accuracy of 2 finger-stick blood tests: the Cepheid 3 gene host-response cartridge (Xpert-HR), which measures 3 host messenger RNA transcripts, and the 3-host protein multibiomarker test (MBT). METHODS We performed a prospective diagnostic accuracy study of consecutive participants with symptoms compatible with pulmonary tuberculosis in The Gambia, South Africa, Uganda, and Vietnam. A composite reference standard for active pulmonary tuberculosis incorporated chest radiography, symptom resolution, and sputum microbiological test results. A training-test set approach was used to evaluate test cutoff specificities at 90% sensitivity. RESULTS Between 1 November 2020 and 1 May 2023, we screened 1262 participants aged 12-70 years with cough lasting >2 weeks and another symptom suggestive of tuberculosis. Of those who were classifiable by reference tests, 1154 participants had evaluable Xpert-HR results and 961 had evaluable MBT results. Xpert-HR had an area under the receiver operating characteristic (AUROC) curve of 0.92 at a cutoff of -1.275 or below, with a sensitivity of 92.8%, specificity of 62.5%, positive predictive value of 47.9%, and negative predictive value of 95.9%. The MBT had an AUROC of 0.91 at a cutoff of ≥0.42, with a sensitivity of 91.4%, specificity of 73.2%, positive predictive value of 52.0%, and negative predictive value of 96.4%. CONCLUSIONS Our results show that both Xpert-HR and the MBT are promising non-sputum-based point-of-care tests. The MBT met the World Health Organization target product profile for a triage test, which suggests it should be further developed.
Collapse
Affiliation(s)
- Jayne S Sutherland
- Vaccines and Immunity Theme, MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, Banjul, The Gambia
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Gian D van der Spuy
- DSI-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
| | - Jane A Shaw
- DSI-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
| | - Tracy Richardson
- DSI-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
| | - Elisa M Tjon Kon Fat
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Awa Gindeh
- Vaccines and Immunity Theme, MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, Banjul, The Gambia
| | - Olumuyiwa Owolabi
- Vaccines and Immunity Theme, MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, Banjul, The Gambia
| | - Nguyen Thuy Thuong Thuong
- Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Le Hong Van
- Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Nguyen Hoang Van
- Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Dang Thi Thanh Thao
- Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | | | - Mary Nsereko
- Department of Medicine, Makerere University, Kampala, Uganda
| | | | | | - John Belisle
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, USA
| | | | | | - Guy Thwaites
- Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Jill Winter
- Catalysis Foundation, Berkeley, California, USA
| | - Hazel M Dockrell
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Thomas J Scriba
- Division of Immunology, Department of Pathology, South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Kim Stanley
- DSI-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
| | - Bronwyn Smith
- DSI-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
| | - Novel N Chegou
- DSI-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
| | - Stephanus T Malherbe
- DSI-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
| | - Annemieke Geluk
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Paul Corstjens
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Gerhard Walzl
- DSI-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
| |
Collapse
|
4
|
Wu C, Xie X, Yang X, Du M, Lin H, Huang J. Applications of gene pair methods in clinical research: advancing precision medicine. MOLECULAR BIOMEDICINE 2025; 6:22. [PMID: 40202606 PMCID: PMC11982013 DOI: 10.1186/s43556-025-00263-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: 09/06/2024] [Revised: 03/18/2025] [Accepted: 03/21/2025] [Indexed: 04/10/2025] Open
Abstract
The rapid evolution of high-throughput sequencing technologies has revolutionized biomedical research, producing vast amounts of gene expression data that hold immense potential for biological discovery and clinical applications. Effectively mining these large-scale, high-dimensional data is crucial for facilitating disease detection, subtype differentiation, and understanding the molecular mechanisms underlying disease progression. However, the conventional paradigm of single-gene profiling, measuring absolute expression levels of individual genes, faces critical limitations in clinical implementation. These include vulnerability to batch effects and platform-dependent normalization requirements. In contrast, emerging approaches analyzing relative expression relationships between gene pairs demonstrate unique advantages. By focusing on binary comparisons of two genes' expression magnitudes, these methods inherently normalize experimental variations while capturing biologically stable interaction patterns. In this review, we systematically evaluate gene pair-based analytical frameworks. We classify eleven computational approaches into two fundamental categories: expression value-based methods quantifying differential expression patterns, and rank-based methods exploiting transcriptional ordering relationships. To bridge methodological development with practical implementation, we establish a reproducible analytical pipeline incorporating feature selection, classifier construction, and model evaluation modules using real-world benchmark datasets from pulmonary tuberculosis studies. These findings position gene pair analysis as a transformative paradigm for mining high-dimensional omics data, with direct implications for precision biomarker discovery and mechanistic studies of disease progression.
Collapse
Affiliation(s)
- Changchun Wu
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Xueqin Xie
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Xin Yang
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Mengze Du
- School of Healthcare Technology, Chengdu Neusoft University, Chengdu, 611844, China
| | - Hao Lin
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 611731, China.
| | - Jian Huang
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 611731, China.
| |
Collapse
|
5
|
Zhang S, Bei C, Li M, Zeng J, Yang L, Ren T, Deng G, Hong R, Cai J, Li D, Wang C, Xu P, Takiff H, Lu S, Zhang P, Gao Q. Identification and evaluation of blood transcriptional biomarker for tuberculosis screening. Int J Infect Dis 2025; 153:107838. [PMID: 39922484 DOI: 10.1016/j.ijid.2025.107838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 02/01/2025] [Accepted: 02/04/2025] [Indexed: 02/10/2025] Open
Abstract
OBJECTIVES Non-sputum-based screening methods for active case finding are a priority for ending tuberculosis. We sought to identify and evaluate blood transcriptional biomarkers suitable for tuberculosis screening. METHODS We integrated five blood RNA-seq datasets from global tuberculosis patients and identified genes that are differentially expressed between tuberculosis patients and healthy controls, using resampling and exhaustive testing. Three candidate biomarker combinations were identified from seven microarray datasets and small-scale clinical samples. The performance of these combinations for screening was evaluated in a cohort of close contacts of pulmonary tuberculosis (PTB) patients, and the results compared with Xpert HR. RESULTS We identified three 3-gene biomarker combinations, each containing two upregulated genes (FCGR1A, BATF2, or GBP5) and one downregulated gene (KLF2), and used these combinations to screen 352 close contacts of PTB. The biomarker combinations distinguished confirmed PTB patients from other participants with AUCs ranging from 0.848 to 0.870. With specificity fixed at 70%, all three combinations showed sensitivities of 87.5%. In a cohort of 205 presumptive pulmonary tuberculosis patients, the AUCs for distinguishing confirmed tuberculosis patients from other diseases ranged from 0.784 to 0.806. At 70% specificity, sensitivities were 75.9-81.5%, and were significantly higher with larger sputum bacterial loads. The performances of the three combinations for tuberculosis screening or diagnosis were comparable to Xpert HR. CONCLUSION The three transcriptomic biomarkers identified in this study performed well for tuberculosis screening, nearly meeting the minimum WHO benchmarks for a triage test and showed potential utility in the development of new screening tools.
Collapse
Affiliation(s)
- Siqi Zhang
- National Clinical Research Center for Infectious Diseases, Shenzhen Clinical Research Center for Tuberculosis, Shenzhen Third People's Hospital, Shenzhen, Guangdong, China
| | - Cheng Bei
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Science, Shanghai Medical College, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
| | - Meng Li
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Science, Shanghai Medical College, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
| | - Jianfeng Zeng
- Department of Pulmonary Medicine and Tuberculosis, Third People's Hospital of Shenzhen, Shenzhen, Guangdong, China
| | - Liangzi Yang
- Department of Pulmonary Medicine and Tuberculosis, Third People's Hospital of Shenzhen, Shenzhen, Guangdong, China
| | - Tantan Ren
- Department of Pulmonary Medicine and Tuberculosis, Third People's Hospital of Shenzhen, Shenzhen, Guangdong, China
| | - Guofang Deng
- Department of Pulmonary Medicine and Tuberculosis, Third People's Hospital of Shenzhen, Shenzhen, Guangdong, China
| | - Ruimin Hong
- National Clinical Research Center for Infectious Diseases, Shenzhen Clinical Research Center for Tuberculosis, Shenzhen Third People's Hospital, Shenzhen, Guangdong, China
| | - Juanjia Cai
- National Clinical Research Center for Infectious Diseases, Shenzhen Clinical Research Center for Tuberculosis, Shenzhen Third People's Hospital, Shenzhen, Guangdong, China
| | - Dan Li
- National Clinical Research Center for Infectious Diseases, Shenzhen Clinical Research Center for Tuberculosis, Shenzhen Third People's Hospital, Shenzhen, Guangdong, China
| | - Chuan Wang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Science, Shanghai Medical College, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
| | - Peng Xu
- National Clinical Research Center for Infectious Diseases, Shenzhen Clinical Research Center for Tuberculosis, Shenzhen Third People's Hospital, Shenzhen, Guangdong, China
| | - Howard Takiff
- Laboratorio de Genética Molecular, CMBC, IVIC, Caracas, Venezuela
| | - Shuihua Lu
- National Clinical Research Center for Infectious Diseases, Shenzhen Clinical Research Center for Tuberculosis, Shenzhen Third People's Hospital, Shenzhen, Guangdong, China; Department of Pulmonary Medicine and Tuberculosis, Third People's Hospital of Shenzhen, Shenzhen, Guangdong, China
| | - Peize Zhang
- Department of Pulmonary Medicine and Tuberculosis, Third People's Hospital of Shenzhen, Shenzhen, Guangdong, China
| | - Qian Gao
- National Clinical Research Center for Infectious Diseases, Shenzhen Clinical Research Center for Tuberculosis, Shenzhen Third People's Hospital, Shenzhen, Guangdong, China; Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Science, Shanghai Medical College, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China.
| |
Collapse
|
6
|
Li Z, Hu Y, Zou F, Gao W, Feng S, Chen G, Yang J, Wang W, Shi C, Cai Y, Deng G, Chen X. Assessing the risk of TB progression: Advances in blood-based biomarker research. Microbiol Res 2025; 292:128038. [PMID: 39752806 DOI: 10.1016/j.micres.2024.128038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 12/18/2024] [Accepted: 12/19/2024] [Indexed: 01/19/2025]
Abstract
This review addresses the significant advancements in the identification of blood-based prognostic biomarkers for tuberculosis (TB), highlighting the importance of early detection to prevent disease progression. The manuscript discusses various biomarker categories, including transcriptomic, proteomic, metabolomic, immune cell-based, cytokine-based, and antibody response-based markers, emphasizing their potential in predicting TB incidence. Despite promising results, practical application is hindered by high costs, technical complexities, and the need for extensive validation across diverse populations. Transcriptomic biomarkers, such as the Risk16 signature, show high sensitivity and specificity, while proteomic and metabolic markers provide insights into protein-level changes and biochemical alterations linked to TB. Immune cell and cytokine markers offer real-time data on the body's response to infection. The manuscript also explores the role of single-nucleotide polymorphisms in TB susceptibility and the challenges of implementing novel RNA signatures as point-of-care tests in low-resource settings. The review concludes that, while significant progress has been made, further research and development are necessary to refine these biomarkers, improve their practical application, and achieve the World Health Organization's TB elimination goals.
Collapse
Affiliation(s)
- Zhaodong Li
- Guangdong Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University School of Medicine, Shenzhen 518000, China; Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen 518060, China
| | - Yunlong Hu
- Guangdong Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University School of Medicine, Shenzhen 518000, China
| | - Fa Zou
- Guangdong Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University School of Medicine, Shenzhen 518000, China
| | - Wei Gao
- Guangdong Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University School of Medicine, Shenzhen 518000, China
| | - SiWan Feng
- Guangdong Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University School of Medicine, Shenzhen 518000, China
| | - Guanghuan Chen
- Guangdong Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University School of Medicine, Shenzhen 518000, China
| | - Jing Yang
- Guangdong Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University School of Medicine, Shenzhen 518000, China
| | - Wenfei Wang
- National Clinical Research Center for Infectious Disease, The Third People's Hospital of Shenzhen, Southern University of Science and Technology, Shenzhen 518112, China
| | - Chenyan Shi
- Department of Preventive Medicine, School of Public Health, Shenzhen University, Shenzhen 518000, China
| | - Yi Cai
- Guangdong Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University School of Medicine, Shenzhen 518000, China
| | - Guofang Deng
- Guangdong Key Lab for Diagnosis & Treatment of Emerging Infectious Diseases, Shenzhen Third People's Hospital, Shenzhen, China.
| | - Xinchun Chen
- Guangdong Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University School of Medicine, Shenzhen 518000, China.
| |
Collapse
|
7
|
Viz-Lasheras S, Gómez-Carballa A, Pardo-Seco J, Bello X, Rivero-Calle I, Dacosta AI, Kaforou M, Habgood-Coote D, Cunnington AJ, Emonts M, Herberg JA, Wright VJ, Carrol ED, Paulus SC, Zenz W, Kohlfürst DS, Van der Flier M, de Groot R, Schlapbach LJ, Agyeman P, Pollard AJ, Fink C, Kuijpers TT, Anderson S, Calvo C, Martínez-Padilla MDC, Pérez-Aragón A, Gómez-Sánchez E, Valencia-Ramos J, Giménez-Sánchez F, Alonso-Quintela P, Moreno-Galarraga L, von Both U, Pokorn M, Zavadska D, Tsolia M, Vermont CL, Moll HA, Levin M, Martinón-Torres F, Salas A, on behalf of EUCLIDS, DIAMONDS, GENDRES and, PERFORM consortia. A 5-transcript signature for discriminating viral and bacterial etiology in pediatric pneumonia. iScience 2025; 28:111747. [PMID: 39906557 PMCID: PMC11791257 DOI: 10.1016/j.isci.2025.111747] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 10/24/2024] [Accepted: 01/02/2025] [Indexed: 02/06/2025] Open
Abstract
Pneumonia stands as the primary cause of death among children under five, yet current diagnosis methods often result in inadequate or unnecessary treatments. Our research seeks to address this gap by identifying host transcriptomic biomarkers in the blood of children with definitive viral and bacterial pneumonia. We performed RNA sequencing on 192 prospectively collected whole blood samples, including 38 controls and 154 pneumonia cases, uncovering a 5-transcript signature (genes FAM20A, BAG3, TDRD9, MXRA7, and KLF14) that effectively distinguishes bacterial from viral pneumonia (area under the curve (AUC): 0.95 [0.88-1.00]). Initial validation using combined definitive and probable cases yielded an AUC of 0.87 [0.77-0.97], while full validation in a new prospective cohort of 32 patients achieved an AUC of 0.92 [0.83-1.00]. This robust signature holds significant potential to enhance diagnostics accuracy for pediatric pneumonia, reducing diagnostic delays and unnecessary treatments and potentially transforming clinical practice.
Collapse
Affiliation(s)
- Sandra Viz-Lasheras
- Unidade de Xenética, Instituto de Ciencias Forenses, Facultade de Medicina, Universidade de Santiago de Compostela, and Genética de Poblaciones en Biomedicina (GenPoB) Research Group, Instituto de Investigación Sanitaria (IDIS), 15706 Hospital Clínico Universitario de Santiago (SERGAS), Galicia, Spain
- Genetics, Vaccines and Infections Research Group (GenViP), Instituto de Investigación Sanitaria de Santiago, 15706 Universidade de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-ES), Madrid, Spain
| | - Alberto Gómez-Carballa
- Unidade de Xenética, Instituto de Ciencias Forenses, Facultade de Medicina, Universidade de Santiago de Compostela, and Genética de Poblaciones en Biomedicina (GenPoB) Research Group, Instituto de Investigación Sanitaria (IDIS), 15706 Hospital Clínico Universitario de Santiago (SERGAS), Galicia, Spain
- Genetics, Vaccines and Infections Research Group (GenViP), Instituto de Investigación Sanitaria de Santiago, 15706 Universidade de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-ES), Madrid, Spain
| | - Jacobo Pardo-Seco
- Unidade de Xenética, Instituto de Ciencias Forenses, Facultade de Medicina, Universidade de Santiago de Compostela, and Genética de Poblaciones en Biomedicina (GenPoB) Research Group, Instituto de Investigación Sanitaria (IDIS), 15706 Hospital Clínico Universitario de Santiago (SERGAS), Galicia, Spain
- Genetics, Vaccines and Infections Research Group (GenViP), Instituto de Investigación Sanitaria de Santiago, 15706 Universidade de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-ES), Madrid, Spain
| | - Xabier Bello
- Unidade de Xenética, Instituto de Ciencias Forenses, Facultade de Medicina, Universidade de Santiago de Compostela, and Genética de Poblaciones en Biomedicina (GenPoB) Research Group, Instituto de Investigación Sanitaria (IDIS), 15706 Hospital Clínico Universitario de Santiago (SERGAS), Galicia, Spain
- Genetics, Vaccines and Infections Research Group (GenViP), Instituto de Investigación Sanitaria de Santiago, 15706 Universidade de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-ES), Madrid, Spain
| | - Irene Rivero-Calle
- Genetics, Vaccines and Infections Research Group (GenViP), Instituto de Investigación Sanitaria de Santiago, 15706 Universidade de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-ES), Madrid, Spain
- Translational Pediatrics and Infectious Diseases, Department of Pediatrics, 15706 Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
| | - Ana Isabel Dacosta
- Genetics, Vaccines and Infections Research Group (GenViP), Instituto de Investigación Sanitaria de Santiago, 15706 Universidade de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-ES), Madrid, Spain
- Translational Pediatrics and Infectious Diseases, Department of Pediatrics, 15706 Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
| | - Myrsini Kaforou
- Department of Infectious Disease, Imperial College London, London W2 1PG, UK
| | | | | | - Marieke Emonts
- Great North Children’s Hospital, Paediatric Immunology, Infectious Diseases & Allergy, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne NE1 4LP, UK
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
- NIHR Newcastle Biomedical Research Centre based at Newcastle upon Tyne Hospitals NHS Trust and Newcastle University, Newcastle upon Tyne NE4 5PL, UK
| | - Jethro A. Herberg
- Department of Infectious Disease, Imperial College London, London W2 1PG, UK
| | - Victoria J. Wright
- Department of Infectious Disease, Imperial College London, London W2 1PG, UK
| | - Enitan D. Carrol
- Department of Infectious Diseases, Alder Hey Children’s NHS Foundation Trust, Liverpool L12 2AP, UK
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L69 7BE, UK
| | - Stephane C. Paulus
- Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford OX3 9DU, UK
| | - Werner Zenz
- Department of General Paediatrics, Medical University of Graz, Graz, Auenbruggerplatz 34/2 8036, Graz, Austria
| | - Daniela S. Kohlfürst
- Department of General Paediatrics, Medical University of Graz, Graz, Auenbruggerplatz 34/2 8036, Graz, Austria
| | - Michiel Van der Flier
- Pediatric Infectious Diseases and Immunology, Wilhelmina Children’s Hospital, University Medical Center Utrecht, Utrecht 3508 AB, the Netherlands
- Pediatric Infectious Diseases and Immunology, Amalia Children’s Hospital, and Section Pediatric Infectious Diseases, Laboratory of Medical Immunology, Department of Laboratory Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen 6500 HB, the Netherlands
| | - Ronald de Groot
- Pediatric Infectious Diseases and Immunology, Amalia Children’s Hospital, and Section Pediatric Infectious Diseases, Laboratory of Medical Immunology, Department of Laboratory Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen 6500 HB, the Netherlands
| | - Luregn J. Schlapbach
- Department of Intensive Care and Neonatology, and Children’s Research Center, University Children’s Hospital Zürich, University of Zürich, Zürich, Switzerland
| | - Philipp Agyeman
- Department of Pediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Andrew J. Pollard
- Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford OX3 9DU, UK
| | - Colin Fink
- Micropathology Ltd, University of Warwick, Warwick CV4 7EZ, UK
| | - Taco T. Kuijpers
- Division of Pediatric Immunology, Rheumatology and Infectious diseases, Emma Children’s Hospital, Amsterdam Univiersyt Medical Center (Amsterdam UMC), Amsterdam 1105 AZ, the Netherlands
| | - Suzanne Anderson
- Medical Research Council Unit at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia
| | - Cristina Calvo
- General Pediatrics, Infectious and Tropical Diseases Department, Hospital La Paz, 28046 Madrid, Spain
- La Paz Research Institute (IdiPAZ), 28029 Madrid, Spain
- Faculty of Medicine, Universidad Autónoma de Madrid (UAM), 28049 Madrid, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
- Red de Investigación Traslacional en Infectología Pediátrica (RITIP), Madrid, Spain
| | | | - Ana Pérez-Aragón
- Hospital Universitario Virgen de las Nieves, Servicio de Pediatría, Granada, Spain
| | - Esteban Gómez-Sánchez
- Department of Pediatric Intensive Care Unit, Hospital Universitario de Burgos, Burgos, Spain
| | - Juan Valencia-Ramos
- Department of Pediatric Intensive Care Unit, Hospital Universitario de Burgos, Burgos, Spain
| | | | - Paula Alonso-Quintela
- Neonatal Intensive Care Unit, Complejo Asistencial Universitario de León, León, Spain
| | - Laura Moreno-Galarraga
- Department of Pediatrics, Complejo Hospitalario de Navarra, Servicio Navarro de Salud, Pamplona, Spain
- IdiSNA (Instituto de Investigación Sanitaria de Navarra), Navarra Institute for Health Research, Pamplona, Spain
| | - Ulrich von Both
- Infectious Diseases, Department of Pediatrics, Dr von Hauner Children’s Hospital, University Hospital, LMU Munich, Munich, Germany
| | - Marko Pokorn
- Division of Paediatrics, University Medical Centre Ljubljana and Medical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Dace Zavadska
- Children’s Clinical University Hospital, Rīga Stradins University, Rïga, Latvia
| | - María Tsolia
- Second Department of Paediatrics, National and Kapodistrian University of Athens (NKUA), School of Medicine, Panagiotis & Aglaia, Kyriakou Children’s Hospital, Athens, Greece
| | | | | | - Michael Levin
- Department of Infectious Disease, Imperial College London, London W2 1PG, UK
| | - Federico Martinón-Torres
- Genetics, Vaccines and Infections Research Group (GenViP), Instituto de Investigación Sanitaria de Santiago, 15706 Universidade de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-ES), Madrid, Spain
- Translational Pediatrics and Infectious Diseases, Department of Pediatrics, 15706 Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
| | - Antonio Salas
- Unidade de Xenética, Instituto de Ciencias Forenses, Facultade de Medicina, Universidade de Santiago de Compostela, and Genética de Poblaciones en Biomedicina (GenPoB) Research Group, Instituto de Investigación Sanitaria (IDIS), 15706 Hospital Clínico Universitario de Santiago (SERGAS), Galicia, Spain
- Genetics, Vaccines and Infections Research Group (GenViP), Instituto de Investigación Sanitaria de Santiago, 15706 Universidade de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-ES), Madrid, Spain
| | - on behalf of EUCLIDS
- Unidade de Xenética, Instituto de Ciencias Forenses, Facultade de Medicina, Universidade de Santiago de Compostela, and Genética de Poblaciones en Biomedicina (GenPoB) Research Group, Instituto de Investigación Sanitaria (IDIS), 15706 Hospital Clínico Universitario de Santiago (SERGAS), Galicia, Spain
- Genetics, Vaccines and Infections Research Group (GenViP), Instituto de Investigación Sanitaria de Santiago, 15706 Universidade de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-ES), Madrid, Spain
- Translational Pediatrics and Infectious Diseases, Department of Pediatrics, 15706 Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
- Department of Infectious Disease, Imperial College London, London W2 1PG, UK
- Great North Children’s Hospital, Paediatric Immunology, Infectious Diseases & Allergy, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne NE1 4LP, UK
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
- NIHR Newcastle Biomedical Research Centre based at Newcastle upon Tyne Hospitals NHS Trust and Newcastle University, Newcastle upon Tyne NE4 5PL, UK
- Department of Infectious Diseases, Alder Hey Children’s NHS Foundation Trust, Liverpool L12 2AP, UK
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L69 7BE, UK
- Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford OX3 9DU, UK
- Department of General Paediatrics, Medical University of Graz, Graz, Auenbruggerplatz 34/2 8036, Graz, Austria
- Pediatric Infectious Diseases and Immunology, Wilhelmina Children’s Hospital, University Medical Center Utrecht, Utrecht 3508 AB, the Netherlands
- Pediatric Infectious Diseases and Immunology, Amalia Children’s Hospital, and Section Pediatric Infectious Diseases, Laboratory of Medical Immunology, Department of Laboratory Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen 6500 HB, the Netherlands
- Department of Intensive Care and Neonatology, and Children’s Research Center, University Children’s Hospital Zürich, University of Zürich, Zürich, Switzerland
- Department of Pediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Micropathology Ltd, University of Warwick, Warwick CV4 7EZ, UK
- Division of Pediatric Immunology, Rheumatology and Infectious diseases, Emma Children’s Hospital, Amsterdam Univiersyt Medical Center (Amsterdam UMC), Amsterdam 1105 AZ, the Netherlands
- Medical Research Council Unit at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia
- General Pediatrics, Infectious and Tropical Diseases Department, Hospital La Paz, 28046 Madrid, Spain
- La Paz Research Institute (IdiPAZ), 28029 Madrid, Spain
- Faculty of Medicine, Universidad Autónoma de Madrid (UAM), 28049 Madrid, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
- Red de Investigación Traslacional en Infectología Pediátrica (RITIP), Madrid, Spain
- Unidad de Cuidados Intensivos Pediátricos, Complejo Hospitalario de Jaen, Jaen, Spain
- Hospital Universitario Virgen de las Nieves, Servicio de Pediatría, Granada, Spain
- Department of Pediatric Intensive Care Unit, Hospital Universitario de Burgos, Burgos, Spain
- Instituto Hispalense de Pediatría, Instituto Balmis de Vacunas, Almería, Spain
- Neonatal Intensive Care Unit, Complejo Asistencial Universitario de León, León, Spain
- Department of Pediatrics, Complejo Hospitalario de Navarra, Servicio Navarro de Salud, Pamplona, Spain
- IdiSNA (Instituto de Investigación Sanitaria de Navarra), Navarra Institute for Health Research, Pamplona, Spain
- Infectious Diseases, Department of Pediatrics, Dr von Hauner Children’s Hospital, University Hospital, LMU Munich, Munich, Germany
- Division of Paediatrics, University Medical Centre Ljubljana and Medical Faculty, University of Ljubljana, Ljubljana, Slovenia
- Children’s Clinical University Hospital, Rīga Stradins University, Rïga, Latvia
- Second Department of Paediatrics, National and Kapodistrian University of Athens (NKUA), School of Medicine, Panagiotis & Aglaia, Kyriakou Children’s Hospital, Athens, Greece
- Department of Pediatrics, Erasmus MC, Rotterdam, the Netherlands
| | - DIAMONDS
- Unidade de Xenética, Instituto de Ciencias Forenses, Facultade de Medicina, Universidade de Santiago de Compostela, and Genética de Poblaciones en Biomedicina (GenPoB) Research Group, Instituto de Investigación Sanitaria (IDIS), 15706 Hospital Clínico Universitario de Santiago (SERGAS), Galicia, Spain
- Genetics, Vaccines and Infections Research Group (GenViP), Instituto de Investigación Sanitaria de Santiago, 15706 Universidade de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-ES), Madrid, Spain
- Translational Pediatrics and Infectious Diseases, Department of Pediatrics, 15706 Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
- Department of Infectious Disease, Imperial College London, London W2 1PG, UK
- Great North Children’s Hospital, Paediatric Immunology, Infectious Diseases & Allergy, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne NE1 4LP, UK
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
- NIHR Newcastle Biomedical Research Centre based at Newcastle upon Tyne Hospitals NHS Trust and Newcastle University, Newcastle upon Tyne NE4 5PL, UK
- Department of Infectious Diseases, Alder Hey Children’s NHS Foundation Trust, Liverpool L12 2AP, UK
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L69 7BE, UK
- Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford OX3 9DU, UK
- Department of General Paediatrics, Medical University of Graz, Graz, Auenbruggerplatz 34/2 8036, Graz, Austria
- Pediatric Infectious Diseases and Immunology, Wilhelmina Children’s Hospital, University Medical Center Utrecht, Utrecht 3508 AB, the Netherlands
- Pediatric Infectious Diseases and Immunology, Amalia Children’s Hospital, and Section Pediatric Infectious Diseases, Laboratory of Medical Immunology, Department of Laboratory Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen 6500 HB, the Netherlands
- Department of Intensive Care and Neonatology, and Children’s Research Center, University Children’s Hospital Zürich, University of Zürich, Zürich, Switzerland
- Department of Pediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Micropathology Ltd, University of Warwick, Warwick CV4 7EZ, UK
- Division of Pediatric Immunology, Rheumatology and Infectious diseases, Emma Children’s Hospital, Amsterdam Univiersyt Medical Center (Amsterdam UMC), Amsterdam 1105 AZ, the Netherlands
- Medical Research Council Unit at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia
- General Pediatrics, Infectious and Tropical Diseases Department, Hospital La Paz, 28046 Madrid, Spain
- La Paz Research Institute (IdiPAZ), 28029 Madrid, Spain
- Faculty of Medicine, Universidad Autónoma de Madrid (UAM), 28049 Madrid, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
- Red de Investigación Traslacional en Infectología Pediátrica (RITIP), Madrid, Spain
- Unidad de Cuidados Intensivos Pediátricos, Complejo Hospitalario de Jaen, Jaen, Spain
- Hospital Universitario Virgen de las Nieves, Servicio de Pediatría, Granada, Spain
- Department of Pediatric Intensive Care Unit, Hospital Universitario de Burgos, Burgos, Spain
- Instituto Hispalense de Pediatría, Instituto Balmis de Vacunas, Almería, Spain
- Neonatal Intensive Care Unit, Complejo Asistencial Universitario de León, León, Spain
- Department of Pediatrics, Complejo Hospitalario de Navarra, Servicio Navarro de Salud, Pamplona, Spain
- IdiSNA (Instituto de Investigación Sanitaria de Navarra), Navarra Institute for Health Research, Pamplona, Spain
- Infectious Diseases, Department of Pediatrics, Dr von Hauner Children’s Hospital, University Hospital, LMU Munich, Munich, Germany
- Division of Paediatrics, University Medical Centre Ljubljana and Medical Faculty, University of Ljubljana, Ljubljana, Slovenia
- Children’s Clinical University Hospital, Rīga Stradins University, Rïga, Latvia
- Second Department of Paediatrics, National and Kapodistrian University of Athens (NKUA), School of Medicine, Panagiotis & Aglaia, Kyriakou Children’s Hospital, Athens, Greece
- Department of Pediatrics, Erasmus MC, Rotterdam, the Netherlands
| | - GENDRES and
- Unidade de Xenética, Instituto de Ciencias Forenses, Facultade de Medicina, Universidade de Santiago de Compostela, and Genética de Poblaciones en Biomedicina (GenPoB) Research Group, Instituto de Investigación Sanitaria (IDIS), 15706 Hospital Clínico Universitario de Santiago (SERGAS), Galicia, Spain
- Genetics, Vaccines and Infections Research Group (GenViP), Instituto de Investigación Sanitaria de Santiago, 15706 Universidade de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-ES), Madrid, Spain
- Translational Pediatrics and Infectious Diseases, Department of Pediatrics, 15706 Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
- Department of Infectious Disease, Imperial College London, London W2 1PG, UK
- Great North Children’s Hospital, Paediatric Immunology, Infectious Diseases & Allergy, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne NE1 4LP, UK
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
- NIHR Newcastle Biomedical Research Centre based at Newcastle upon Tyne Hospitals NHS Trust and Newcastle University, Newcastle upon Tyne NE4 5PL, UK
- Department of Infectious Diseases, Alder Hey Children’s NHS Foundation Trust, Liverpool L12 2AP, UK
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L69 7BE, UK
- Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford OX3 9DU, UK
- Department of General Paediatrics, Medical University of Graz, Graz, Auenbruggerplatz 34/2 8036, Graz, Austria
- Pediatric Infectious Diseases and Immunology, Wilhelmina Children’s Hospital, University Medical Center Utrecht, Utrecht 3508 AB, the Netherlands
- Pediatric Infectious Diseases and Immunology, Amalia Children’s Hospital, and Section Pediatric Infectious Diseases, Laboratory of Medical Immunology, Department of Laboratory Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen 6500 HB, the Netherlands
- Department of Intensive Care and Neonatology, and Children’s Research Center, University Children’s Hospital Zürich, University of Zürich, Zürich, Switzerland
- Department of Pediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Micropathology Ltd, University of Warwick, Warwick CV4 7EZ, UK
- Division of Pediatric Immunology, Rheumatology and Infectious diseases, Emma Children’s Hospital, Amsterdam Univiersyt Medical Center (Amsterdam UMC), Amsterdam 1105 AZ, the Netherlands
- Medical Research Council Unit at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia
- General Pediatrics, Infectious and Tropical Diseases Department, Hospital La Paz, 28046 Madrid, Spain
- La Paz Research Institute (IdiPAZ), 28029 Madrid, Spain
- Faculty of Medicine, Universidad Autónoma de Madrid (UAM), 28049 Madrid, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
- Red de Investigación Traslacional en Infectología Pediátrica (RITIP), Madrid, Spain
- Unidad de Cuidados Intensivos Pediátricos, Complejo Hospitalario de Jaen, Jaen, Spain
- Hospital Universitario Virgen de las Nieves, Servicio de Pediatría, Granada, Spain
- Department of Pediatric Intensive Care Unit, Hospital Universitario de Burgos, Burgos, Spain
- Instituto Hispalense de Pediatría, Instituto Balmis de Vacunas, Almería, Spain
- Neonatal Intensive Care Unit, Complejo Asistencial Universitario de León, León, Spain
- Department of Pediatrics, Complejo Hospitalario de Navarra, Servicio Navarro de Salud, Pamplona, Spain
- IdiSNA (Instituto de Investigación Sanitaria de Navarra), Navarra Institute for Health Research, Pamplona, Spain
- Infectious Diseases, Department of Pediatrics, Dr von Hauner Children’s Hospital, University Hospital, LMU Munich, Munich, Germany
- Division of Paediatrics, University Medical Centre Ljubljana and Medical Faculty, University of Ljubljana, Ljubljana, Slovenia
- Children’s Clinical University Hospital, Rīga Stradins University, Rïga, Latvia
- Second Department of Paediatrics, National and Kapodistrian University of Athens (NKUA), School of Medicine, Panagiotis & Aglaia, Kyriakou Children’s Hospital, Athens, Greece
- Department of Pediatrics, Erasmus MC, Rotterdam, the Netherlands
| | - PERFORM consortia
- Unidade de Xenética, Instituto de Ciencias Forenses, Facultade de Medicina, Universidade de Santiago de Compostela, and Genética de Poblaciones en Biomedicina (GenPoB) Research Group, Instituto de Investigación Sanitaria (IDIS), 15706 Hospital Clínico Universitario de Santiago (SERGAS), Galicia, Spain
- Genetics, Vaccines and Infections Research Group (GenViP), Instituto de Investigación Sanitaria de Santiago, 15706 Universidade de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-ES), Madrid, Spain
- Translational Pediatrics and Infectious Diseases, Department of Pediatrics, 15706 Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
- Department of Infectious Disease, Imperial College London, London W2 1PG, UK
- Great North Children’s Hospital, Paediatric Immunology, Infectious Diseases & Allergy, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne NE1 4LP, UK
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
- NIHR Newcastle Biomedical Research Centre based at Newcastle upon Tyne Hospitals NHS Trust and Newcastle University, Newcastle upon Tyne NE4 5PL, UK
- Department of Infectious Diseases, Alder Hey Children’s NHS Foundation Trust, Liverpool L12 2AP, UK
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L69 7BE, UK
- Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford OX3 9DU, UK
- Department of General Paediatrics, Medical University of Graz, Graz, Auenbruggerplatz 34/2 8036, Graz, Austria
- Pediatric Infectious Diseases and Immunology, Wilhelmina Children’s Hospital, University Medical Center Utrecht, Utrecht 3508 AB, the Netherlands
- Pediatric Infectious Diseases and Immunology, Amalia Children’s Hospital, and Section Pediatric Infectious Diseases, Laboratory of Medical Immunology, Department of Laboratory Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen 6500 HB, the Netherlands
- Department of Intensive Care and Neonatology, and Children’s Research Center, University Children’s Hospital Zürich, University of Zürich, Zürich, Switzerland
- Department of Pediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Micropathology Ltd, University of Warwick, Warwick CV4 7EZ, UK
- Division of Pediatric Immunology, Rheumatology and Infectious diseases, Emma Children’s Hospital, Amsterdam Univiersyt Medical Center (Amsterdam UMC), Amsterdam 1105 AZ, the Netherlands
- Medical Research Council Unit at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia
- General Pediatrics, Infectious and Tropical Diseases Department, Hospital La Paz, 28046 Madrid, Spain
- La Paz Research Institute (IdiPAZ), 28029 Madrid, Spain
- Faculty of Medicine, Universidad Autónoma de Madrid (UAM), 28049 Madrid, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
- Red de Investigación Traslacional en Infectología Pediátrica (RITIP), Madrid, Spain
- Unidad de Cuidados Intensivos Pediátricos, Complejo Hospitalario de Jaen, Jaen, Spain
- Hospital Universitario Virgen de las Nieves, Servicio de Pediatría, Granada, Spain
- Department of Pediatric Intensive Care Unit, Hospital Universitario de Burgos, Burgos, Spain
- Instituto Hispalense de Pediatría, Instituto Balmis de Vacunas, Almería, Spain
- Neonatal Intensive Care Unit, Complejo Asistencial Universitario de León, León, Spain
- Department of Pediatrics, Complejo Hospitalario de Navarra, Servicio Navarro de Salud, Pamplona, Spain
- IdiSNA (Instituto de Investigación Sanitaria de Navarra), Navarra Institute for Health Research, Pamplona, Spain
- Infectious Diseases, Department of Pediatrics, Dr von Hauner Children’s Hospital, University Hospital, LMU Munich, Munich, Germany
- Division of Paediatrics, University Medical Centre Ljubljana and Medical Faculty, University of Ljubljana, Ljubljana, Slovenia
- Children’s Clinical University Hospital, Rīga Stradins University, Rïga, Latvia
- Second Department of Paediatrics, National and Kapodistrian University of Athens (NKUA), School of Medicine, Panagiotis & Aglaia, Kyriakou Children’s Hospital, Athens, Greece
- Department of Pediatrics, Erasmus MC, Rotterdam, the Netherlands
| |
Collapse
|
8
|
Seifert M, Catanzaro DG, Gracia M, Hillery N, Tahseen S, Masood F, Hussain A, Majeed U, Colman RE, Syed RR, Catanzaro A, Rodwell T. Prospective Exploratory Evaluation of Cepheid Xpert Mycobacterium tuberculosis Host Response Cartridge: A Focus on Adolescents and Young Adults. Clin Infect Dis 2025; 80:180-188. [PMID: 39233548 PMCID: PMC11797382 DOI: 10.1093/cid/ciae461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 08/05/2024] [Accepted: 09/04/2024] [Indexed: 09/06/2024] Open
Abstract
BACKGROUND An accurate, rapid, non-sputum-based triage test for diagnosing tuberculosis (TB) is needed. METHODS A prospective evaluation of the Cepheid GeneXpert Mycobacterium tuberculosis Host Response cartridge (Xpert-MTB-HR), a prototype blood-based host response mRNA signature assay, among individuals presenting with TB-like symptoms was performed in Pakistan and results were compared to 3 reference standards: Xpert MTB/RIF Ultra, bacteriological confirmation (Xpert MTB/RIF Ultra and/or culture positivity), and composite clinical diagnosis (clinician diagnosis, treatment initiation, Xpert MTB/RIF Ultra, and/or culture positivity). Analyses were conducted both for the entire study cohort and separately in the adolescent and young adult cohort (aged 10-24 years). RESULTS A total of 497 participants, aged 6-83 years, returned valid Xpert-MTB-HR results. When a diagnostic threshold was set for a sensitivity of >90%, specificity was 32% (95% confidence interval [CI], 28%-37%) compared to Xpert MTB/RIF Ultra, 29% (95% CI, 25%-34%) compared to a bacteriological confirmation, and 22% (95% CI, 18%-26%) compared to a composite clinical diagnosis. However, when evaluating only the adolescent and young adult cohort with a diagnostic threshold set for sensitivity of >90%, specificity was 82% (95% CI, 74%-89%) compared to Xpert MTB/RIF Ultra, 84% (95% CI, 75%-90%) compared to a bacteriological confirmation, and 54% (95% CI, 44%-64%) compared to a composite clinical diagnosis. CONCLUSIONS While the Xpert-MTB-HR does not meet World Health Organization minimum criteria in the general population, in our study it does meet the minimum sensitivity and specificity requirements for a non-sputum-based triage test among adolescents and young adults when compared to Xpert MTB/RIF Ultra or bacteriological confirmation.
Collapse
Affiliation(s)
- Marva Seifert
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, University of California San Diego, La Jolla, USA
| | - Donald G Catanzaro
- Department of Biological Sciences, University of Arkansas, Fayetteville, USA
| | - Michael Gracia
- Department of Pediatrics, University of California San Diego, La Jolla, USA
| | - Naomi Hillery
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, University of California San Diego, La Jolla, USA
| | - Sabira Tahseen
- National Tuberculosis Control Program, National Tuberculosis Reference Laboratory, Islamabad, Pakistan
| | - Faisal Masood
- National Tuberculosis Control Program, National Tuberculosis Reference Laboratory, Islamabad, Pakistan
| | - Alamdar Hussain
- National Tuberculosis Control Program, National Tuberculosis Reference Laboratory, Islamabad, Pakistan
| | - Uzma Majeed
- National Tuberculosis Control Program, National Tuberculosis Reference Laboratory, Islamabad, Pakistan
| | - Rebecca E Colman
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, University of California San Diego, La Jolla, USA
| | - Rehan R Syed
- Division of Infectious Diseases and Global Public Health, Department of Medicine, University of California, San Diego, La Jolla, USA
| | - Antonino Catanzaro
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, University of California San Diego, La Jolla, USA
| | - Timothy Rodwell
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, University of California San Diego, La Jolla, USA
| |
Collapse
|
9
|
Painter H, Larsen SE, Williams BD, Abdelaal HFM, Baldwin SL, Fletcher HA, Fiore-Gartland A, Coler RN. Backtranslation of human RNA biosignatures of tuberculosis disease risk into the preclinical pipeline is condition dependent. mSphere 2025; 10:e0086424. [PMID: 39651886 PMCID: PMC11774039 DOI: 10.1128/msphere.00864-24] [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: 10/11/2024] [Accepted: 11/03/2024] [Indexed: 12/18/2024] Open
Abstract
It is unclear whether human progression to active tuberculosis disease (TB) risk signatures are viable endpoint criteria for evaluations of treatments in development. TB is the deadliest infectious disease globally and more efficacious vaccines are needed to reduce this mortality. However, the immune correlates of protection for either preventing infection with Mycobacterium tuberculosis or preventing TB disease have yet to be completely defined, making the advancement of candidate vaccines through the pipeline slow, costly, and fraught with risk. Human-derived correlate of risk (COR) gene signatures, which identify an individual's risk of progressing to active TB disease, provide an opportunity for evaluating new therapies for TB with clear and defined endpoints. Though prospective clinical trials with longitudinal sampling are prohibitively expensive, the characterization of COR gene signatures is practical with preclinical models. Using a 3Rs (replacement, reduction, and refinement) approach we reanalyzed heterogeneous publicly available transcriptional data sets to determine whether a specific set of COR signatures are viable endpoints in the preclinical pipeline. We selected RISK6, Sweeney3, and BATF2 human-derived blood-based RNA biosignatures because they require relatively few genes and have been carefully evaluated across several clinical cohorts. These data suggest that in certain experimental designs and in several tissue types, human COR signatures correlate with disease progression as measured by the bacterial burden in the preclinical TB model pipeline. We observed the best performance when the model most closely reflected human infection or disease conditions. Human-derived COR signatures offer an opportunity for high-throughput preclinical endpoint criteria of vaccine and drug therapy evaluations. IMPORTANCE Understanding the strengths or limitations of back-translating human-derived correlate of risk (COR) RNA signatures into the preclinical pipeline may help streamline down-selection of therapeutic vaccine and drug candidates and better align preclinical models with proposed clinical trial efficacy endpoints.
Collapse
Affiliation(s)
- Hannah Painter
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Sasha E. Larsen
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Brittany D. Williams
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, USA
- Department of Global Health, University of Washington, Seattle, Washington, USA
| | - Hazem F. M. Abdelaal
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Susan L. Baldwin
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Helen A. Fletcher
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Andrew Fiore-Gartland
- Biostatistics, Bioinformatics and Epidemiology Program, Fred Hutch Cancer Center, Seattle, Washington, USA
| | - Rhea N. Coler
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, USA
- Department of Global Health, University of Washington, Seattle, Washington, USA
- Department of Pediatrics, University of Washington School of Medicine, Seattle, Washington, USA
| |
Collapse
|
10
|
Rosenheim J, Abebe M, Belay M, Tulu B, Tayachew D, Tegegn M, Younis S, Jolliffe DA, Aseffa A, Ameni G, Reece ST, Noursadeghi M, Martineau AR. Detection of M. tuberculosis DNA in peripheral blood mononuclear cells of tuberculosis contacts does not associate with blood RNA signatures for incipient tuberculosis. Eur Respir J 2024; 64:2400479. [PMID: 39227070 PMCID: PMC11635380 DOI: 10.1183/13993003.00479-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 08/05/2024] [Indexed: 09/05/2024]
Abstract
Human exposure to Mycobacterium tuberculosis (Mtb) is thought to result in a spectrum of outcomes, including bacillary clearance, quiescent Mtb infection, incipient tuberculosis (TB), subclinical TB and active TB [1]. Incipient TB – defined as a prolonged asymptomatic phase of early disease preceding clinical presentation as active disease [2] – may be distinguished from quiescent Mtb infection by detection of host gene expression signatures in blood, whose presence associates with increased risk of progression to active TB [3]. Detection of M. tuberculosis DNA in TB contacts’ PBMCs does not associate with blood RNA signatures for incipient tuberculosis https://bit.ly/4dliODg
Collapse
Affiliation(s)
- Joshua Rosenheim
- Division of Infection and Immunity, University College London, London, UK
| | - Markos Abebe
- Armauer Hansen Research Institute, Addis Ababa, Ethiopia
| | - Mulugeta Belay
- Armauer Hansen Research Institute, Addis Ababa, Ethiopia
- Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Begna Tulu
- Research Center Borstel, Leibniz Lung Center, Borstel, Germany
- Bahir Dar University, Bahir Dar, Ethiopia
| | - Dawit Tayachew
- Armauer Hansen Research Institute, Addis Ababa, Ethiopia
| | | | - Sidra Younis
- Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- National University of Medical Sciences, Rawalpindi, Pakistan
| | - David A Jolliffe
- Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Abraham Aseffa
- Armauer Hansen Research Institute, Addis Ababa, Ethiopia
| | - Gobena Ameni
- Aklilu Lemma Institute of Pathobiology, Addis Ababa University, Addis Ababa, Ethiopia
- Department of Veterinary Medicine, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Stephen T Reece
- Infectious Diseases and Vaccines, Kymab, Babraham Research Campus, Cambridge, UK
| | - Mahdad Noursadeghi
- Division of Infection and Immunity, University College London, London, UK
- These authors contributed equally
| | - Adrian R Martineau
- Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- These authors contributed equally
| |
Collapse
|
11
|
Sybertz NM, Al Jubaer S, Larsen MH, Alexander KA. Assessment of transcriptional markers for the differentiation of Mycobacterium mungi infection status in free-ranging banded mongoose (Mungos mungo). Tuberculosis (Edinb) 2024; 149:102565. [PMID: 39293135 DOI: 10.1016/j.tube.2024.102565] [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: 06/01/2024] [Revised: 08/14/2024] [Accepted: 09/04/2024] [Indexed: 09/20/2024]
Abstract
There is an increasingly urgent need to improve our ability to accurately forecast and control zoonotic diseases in wildlife reservoirs. We are confronted, however, with the continued challenge of accurately determining host infection status across space and time. This dilemma is epitomized with the Mycobacterium tuberculosis Complex (MTBC) pathogens and particularly in free-ranging wildlife, a critical global challenge for both human and animal health. In humans, transcriptional markers have been increasingly identified as a robust tool for diagnosing Mycobacterium tuberculosis (MTB) infection status but have rarely been utilized for diagnosing TB in free-ranging wildlife populations. Here, we report the first use of transcriptional markers to evaluate TB infection status in a free-ranging wildlife species, banded mongoose (Mungos mungo), infected with the MTBC pathogen, Mycobacterium mungi. In this study, we found that GBP5 and DUSP3 were significantly upregulated in free-ranging banded mongoose infected with M. mungi. These results provide the first step in developing an antemortem diagnostic tool for use in free-ranging wildlife species. Our results highlight the potential of transcriptional marker-based assays to advance our ability to detect and manage TB in free-ranging wildlife, especially in field studies and other scenarios when conventional diagnostics are not feasible.
Collapse
Affiliation(s)
- Nicholas M Sybertz
- Department of Fish and Wildlife Conservation, Virginia Tech, 310 West Campus Drive, Blacksburg, VA, USA.
| | - Shamim Al Jubaer
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY, USA.
| | - Michelle H Larsen
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY, USA.
| | - Kathleen A Alexander
- Department of Fish and Wildlife Conservation, Virginia Tech, 310 West Campus Drive, Blacksburg, VA, USA; Chobe Research Institute, Center for African Resources: Communities, Animals, and Land Use (CARACAL), Plot 3102 Airport Road, Kasane, Botswana.
| |
Collapse
|
12
|
Shan Q, Li Y, Yuan K, Yang X, Yang L, He JQ. Distinguish active tuberculosis with an immune-related signature and molecule subtypes: a multi-cohort analysis. Sci Rep 2024; 14:29564. [PMID: 39609541 PMCID: PMC11605007 DOI: 10.1038/s41598-024-80072-3] [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: 06/03/2024] [Accepted: 11/14/2024] [Indexed: 11/30/2024] Open
Abstract
BACKGROUND Distinguishing latent tuberculosis infection (LTBI) from active tuberculosis (ATB) is very important. This study aims to analyze cases from multiple cohorts and get the signature that can distinguish LTBI from ATB. METHODS Thirteen datasets were downloaded from the gene expression omnibus (GEO) database. Three datasets were selected as discovery datasets, and the hub genes were discovered through WGCNA. In the training cohort, we use machine learning to establish the signature, verify the authentication ability of the signature in the remaining datasets, and compare it with other signatures. Cluster analysis was carried out on ATB cases, immune cell infiltration analysis, GSVA analysis, and drug sensitivity analysis were carried out on different clusters. RESULTS In the discovery datasets, we discovered five hub genes. A signature (SLC26A8, ANKRD22, and FCGR1B) is obtained in the training cohort. In the total cohort, the three-gene signature can separate LTBI from ATB (the total area under ROC curve (AUC) is 0.801, 95% CI 0.771-0.830). Compared with other author's signatures, our signature shows good identification ability. Immunological analysis showed that SLC26A8, ANKRD22, and FCGR1B were closely related to the infiltration of immune cells. According to the expression of the three genes, ATB can be divided into two clusters, which are different in immune cell infiltration analysis, gene set variation, and drug sensitivity. CONCLUSION Our study produced an immune-related three-gene signature to distinguish LTBI from ATB, which may help us to manage and treat tuberculosis patients.
Collapse
Affiliation(s)
- Qingqing Shan
- Department of Respiration, West China Hospital of Sichuan University, 37# Guo Xue Xiang, Chengdu, 610041, Sichuan Province, China
- Department of Respiration, Chengdu First People's Hospital, Chengdu, 610095, China
| | - Yangke Li
- Department of Respiration, Chengdu First People's Hospital, Chengdu, 610095, China
| | - Kun Yuan
- Department of Respiration, Chengdu First People's Hospital, Chengdu, 610095, China
| | - Xiao Yang
- Department of Respiration, Chengdu First People's Hospital, Chengdu, 610095, China
| | - Li Yang
- Xiaojiahe Community Health Service Center, Chengdu, 610094, China
| | - Jian-Qing He
- Department of Respiration, West China Hospital of Sichuan University, 37# Guo Xue Xiang, Chengdu, 610041, Sichuan Province, China.
| |
Collapse
|
13
|
Öhrnberg I, Karlsson L, Sayyab S, Paues J, Martínez-Enguita D, Gustafsson M, Espinoza-Lopez P, Méndez-Aranda M, Meza E, Ugarte-Gil C, Kiprotich N, Diero L, Tonui R, Lerm M. A DNA methylation signature identified in the buccal mucosa reflecting active tuberculosis is changing during tuberculosis treatment. Sci Rep 2024; 14:29552. [PMID: 39609478 PMCID: PMC11604703 DOI: 10.1038/s41598-024-80570-4] [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: 07/31/2024] [Accepted: 11/19/2024] [Indexed: 11/30/2024] Open
Abstract
Tuberculosis (TB) poses a significant global health threat, with high mortality rates if left untreated. Current sputum-based TB treatment monitoring methods face numerous challenges, particularly in relation to sample collection and analysis. This pilot study explores the potential of TB status assessment using DNA methylation (DNAm) signatures, which are gaining recognition as diagnostic and predictive tools for various diseases. We collected buccal swab samples from pulmonary TB patients at the commencement of TB treatment (n = 10), and at one, two, and six-month follow-up intervals. We also collected samples from healthy controls (n = 10) and individuals exposed to TB (n = 10). DNAm patterns were mapped using the Illumina Infinium Methylation EPIC 850 K platform. A DNAm profile distinct from controls was discovered in the oral mucosa of TB patients at the start of treatment, and this profile changed throughout the course of TB treatment. These findings were corroborated in a separate validation cohort of TB patients (n = 41), monitored at two and six months into their TB treatment. We developed a machine learning model to predict symptom scores using the identified DNAm TB profile. The model was trained and evaluated on the pilot, validation, and two additional independent cohorts, achieving an R2 of 0.80, Pearson correlation of 0.90, and mean absolute error of 0.13. While validation is needed in larger cohorts, the result opens the possibility of employing DNAm-based diagnostic and prognostic tools for TB in future clinical practice.
Collapse
Affiliation(s)
- Isabelle Öhrnberg
- Division of Inflammation and Infection, Lab 1, Floor 12, Linköping University, 58185, Linköping, Sweden
| | - Lovisa Karlsson
- Division of Inflammation and Infection, Lab 1, Floor 12, Linköping University, 58185, Linköping, Sweden
| | - Shumaila Sayyab
- Division of Inflammation and Infection, Lab 1, Floor 12, Linköping University, 58185, Linköping, Sweden
| | - Jakob Paues
- Division of Inflammation and Infection, Lab 1, Floor 12, Linköping University, 58185, Linköping, Sweden
- Division of Infectious Diseases, Department of Biomedical and Clinical Sciences, Faculty of Medicine and Health Sciences, Linköping University, Linköping, Sweden
| | | | - Mika Gustafsson
- Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden
| | - Patricia Espinoza-Lopez
- Facultad de Medicina, Universidad Peruana Cayetano Heredia, Lima, Peru
- Instituto de Medicina Tropical Alexander Von Humboldt, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Melissa Méndez-Aranda
- Laboratorios de Investigación y Desarrollo, Facultad de Ciencias e Ingeniería, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Ericka Meza
- Instituto de Medicina Tropical Alexander Von Humboldt, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Cesar Ugarte-Gil
- Facultad de Medicina, Universidad Peruana Cayetano Heredia, Lima, Peru
- Instituto de Medicina Tropical Alexander Von Humboldt, Universidad Peruana Cayetano Heredia, Lima, Peru
- Department of Epidemiology, School of Public and Population Health, University of Texas Medical Branch, Galveston, TX, USA
| | - Nicholas Kiprotich
- Biochemistry and Clinical Chemistry, Moi University, Eldoret, Kenya
- AMPATH Kenya, Moi University, Eldoret, Kenya
| | - Lameck Diero
- AMPATH Kenya, Moi University, Eldoret, Kenya
- Department of Medicine, Moi University, Eldoret, Kenya
| | - Ronald Tonui
- AMPATH Kenya, Moi University, Eldoret, Kenya
- Department of Pathology, Moi University, Eldoret, Kenya
| | - Maria Lerm
- Division of Inflammation and Infection, Lab 1, Floor 12, Linköping University, 58185, Linköping, Sweden.
| |
Collapse
|
14
|
Tepekule B, Jörimann L, Schenkel CD, Opitz L, Tschumi J, Wolfensberger R, Neumann K, Kusejko K, Zeeb M, Boeck L, Kälin M, Notter J, Furrer H, Hoffmann M, Hirsch HH, Calmy A, Cavassini M, Labhardt ND, Bernasconi E, Oesch G, Metzner KJ, Braun DL, Günthard HF, Kouyos RD, Duffy F, Nemeth J, the Swiss HIV Cohort Study. Transcriptional profile of Mycobacterium tuberculosis infection in people living with HIV. iScience 2024; 27:111228. [PMID: 39555417 PMCID: PMC11565417 DOI: 10.1016/j.isci.2024.111228] [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: 05/05/2024] [Revised: 07/04/2024] [Accepted: 10/18/2024] [Indexed: 11/19/2024] Open
Abstract
In people with HIV-1 (PWH), Mycobacterium tuberculosis (MTB) infection poses a significant threat. While active tuberculosis (TB) accelerates immunodeficiency, the interaction between MTB and HIV-1 during asymptomatic phases remains unclear. Analysis of peripheral blood mononuclear cells (PBMC) transcriptomic profiles in PWH, with and without controlled viral loads, revealed distinct clustering in MTB-infected individuals. Functional annotation identified alterations in IL-6, TNF, and KRAS pathways. Notably, MTB-related genes displayed an inverse correlation with HIV-1 viremia, at both individual and signature score levels. These findings suggest that MTB infection in PWH induces a shift in immune system activation, inversely related to HIV-1 viral load. These results may explain the observed enhanced antiretroviral control in MTB-infected PWH. This study highlights the complex interplay between MTB and HIV-1, emphasizing the importance of understanding their interaction for managing co-infections in this population.
Collapse
Affiliation(s)
- Burcu Tepekule
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, Zurich, Switzerland
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, NJ, USA
| | - Lisa Jörimann
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, Zurich, Switzerland
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Corinne D. Schenkel
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, Zurich, Switzerland
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Lennart Opitz
- Functional Genomics Center Zurich, Swiss Federal Institute of Technology and University of Zurich, Zurich, Switzerland
| | - Jasmin Tschumi
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, Zurich, Switzerland
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Rebekka Wolfensberger
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, Zurich, Switzerland
| | - Kathrin Neumann
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, Zurich, Switzerland
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Katharina Kusejko
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, Zurich, Switzerland
| | - Marius Zeeb
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, Zurich, Switzerland
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Lucas Boeck
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Marisa Kälin
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, Zurich, Switzerland
| | - Julia Notter
- Division of Infectious Diseases and Hospital Epidemiology, Cantonal Hospital St Gallen, St. Gallen, Switzerland
| | - Hansjakob Furrer
- Department of Infectious Diseases, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Matthias Hoffmann
- Division of Infectious Diseases and Hospital Epidemiology, Cantonal Hospital Olten, Olten, Switzerland
| | - Hans H. Hirsch
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Basel, Basel, Switzerland
- Clinical Virology, Laboratory Medicine, University Hospital Basel, Basel, Switzerland
- Department Biomedicine, Transplantation and Clinical Virology, University of Basel, Basel, Switzerland
| | - Alexandra Calmy
- Division of Infectious Diseases, University Hospital Geneva, University of Geneva, Geneva, Switzerland
| | - Matthias Cavassini
- Division of Infectious Diseases, University Hospital Lausanne, University of Lausanne, Lausanne, Switzerland
| | - Niklaus D. Labhardt
- Division Clinical Epidemiology, Department of Clinical Research, University Hospital Basel, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Enos Bernasconi
- Division of Infectious Diseases, University Hospital Geneva, University of Geneva, Geneva, Switzerland
- Division of Infectious Diseases, Ente Ospedaliero Cantonale, Lugano, Switzerland
- University of Geneva and University of Southern Switzerland, Lugano, Switzerland
| | - Gabriela Oesch
- Department of Child Neurology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Karin J. Metzner
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, Zurich, Switzerland
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Dominique L. Braun
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, Zurich, Switzerland
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Huldrych F. Günthard
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, Zurich, Switzerland
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Roger D. Kouyos
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, Zurich, Switzerland
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Fergal Duffy
- Seattle Children’s Research Institute, Seattle, WA, USA
| | - Johannes Nemeth
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, Zurich, Switzerland
| | - the Swiss HIV Cohort Study
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, Zurich, Switzerland
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, NJ, USA
- Functional Genomics Center Zurich, Swiss Federal Institute of Technology and University of Zurich, Zurich, Switzerland
- Department of Biomedicine, University of Basel, Basel, Switzerland
- Division of Infectious Diseases and Hospital Epidemiology, Cantonal Hospital St Gallen, St. Gallen, Switzerland
- Department of Infectious Diseases, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Division of Infectious Diseases and Hospital Epidemiology, Cantonal Hospital Olten, Olten, Switzerland
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Basel, Basel, Switzerland
- Clinical Virology, Laboratory Medicine, University Hospital Basel, Basel, Switzerland
- Department Biomedicine, Transplantation and Clinical Virology, University of Basel, Basel, Switzerland
- Division of Infectious Diseases, University Hospital Geneva, University of Geneva, Geneva, Switzerland
- Division of Infectious Diseases, University Hospital Lausanne, University of Lausanne, Lausanne, Switzerland
- Division Clinical Epidemiology, Department of Clinical Research, University Hospital Basel, Basel, Switzerland
- University of Basel, Basel, Switzerland
- Division of Infectious Diseases, Ente Ospedaliero Cantonale, Lugano, Switzerland
- University of Geneva and University of Southern Switzerland, Lugano, Switzerland
- Department of Child Neurology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Seattle Children’s Research Institute, Seattle, WA, USA
| |
Collapse
|
15
|
Nakiboneka R, Walbaum N, Musisi E, Nevels M, Nyirenda T, Nliwasa M, Msefula CL, Sloan D, Sabiiti W. Specific human gene expression in response to infection is an effective marker for diagnosis of latent and active tuberculosis. Sci Rep 2024; 14:26884. [PMID: 39505948 PMCID: PMC11541504 DOI: 10.1038/s41598-024-77164-5] [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: 08/12/2024] [Accepted: 10/21/2024] [Indexed: 11/08/2024] Open
Abstract
RNA sequencing and microarray analysis revealed transcriptional markers expressed in whole blood can differentiate active pulmonary TB (ATB) from other respiratory diseases (ORDs), and latent TB infection (LTBI) from healthy controls (HC). Here we describe a streamlined reverse transcriptase quantitative polymerase chain reaction (RT-qPCR) assay that could be applied at near point-of-care for diagnosing and distinguishing ATB from ORDs and LTBI from HC. A literature review was undertaken to identify the most plausible host-gene markers (HGM) of TB infection. Primers, and dual labelled hydrolysis probes were designed and analytically evaluated for accuracy in an in-vitro model of infection using a lung fibroblast cell-line. Best performing genes were multiplexed into panels of 2-4 targets and taken forward for clinical evaluation. Mycobacteria Growth Indicator Tube and QuantiFERON-TB Gold Plus were used as reference tests for ATB and LTBI respectively. A total of 16 HGM were selected and incorporated into five multiplex RT-qPCR panels. PCR assay efficiency of all evaluated targets was ≥ 90% with a median analytical sensitivity of 292 copies/µl [IQR: 215.0-358.3 copies/µl], and a median limit of quantification of 61.7 copies/µl [IQR: 29.4-176.3 copies/µl]. Clinically, ATB was characterised by higher gene expression than ORDs, while LTBI was associated with lower gene expression than HC, Kruskal-Wallis p < 0.0001. Crucially, BATF2, CD64, GBP5, C1QB, GBP6, DUSP3, and GAS6 exhibited high differentiative ability for ATB from ORDs, LTBI or HC while KLF2, PTPRC, NEMF, ASUN, and ZNF296 independently discriminated LTBI from HC. Our results show that different HGM maybe required for ATB and LTBI differentiation from ORDs or HC respectively and demonstrate the feasibility of host gene-based RT-qPCR to diagnose ATB and LTBI at near point-of-care.
Collapse
Affiliation(s)
- Ritah Nakiboneka
- Division of Infection and Global Health, School of Medicine, University of St Andrews, St Andrews, KY16 9TF, UK
- Department of Pathology, Kamuzu University of Health Sciences, Blantyre, Malawi
- Helse Nord Tuberculosis Initiative (HNTI), Pathology Department, Kamuzu University of Health Sciences, Blantyre, Malawi
- Africa Centre for Public Health and Herbal Medicines (ACEPHEM), Kamuzu University of Health Sciences, Blantyre, Malawi
| | - Natasha Walbaum
- Division of Infection and Global Health, School of Medicine, University of St Andrews, St Andrews, KY16 9TF, UK
| | - Emmanuel Musisi
- Division of Infection and Global Health, School of Medicine, University of St Andrews, St Andrews, KY16 9TF, UK
- Adroit Biomedical and Bio-entrepreneurship Research Services (ABBRS), Kampala, Uganda
| | - Michael Nevels
- Biomedical Sciences Research Complex (BSRC), School of Biology, University of St Andrews, St Andrews, UK
| | - Tonney Nyirenda
- Department of Pathology, Kamuzu University of Health Sciences, Blantyre, Malawi
| | - Marriott Nliwasa
- Department of Pathology, Kamuzu University of Health Sciences, Blantyre, Malawi
- Helse Nord Tuberculosis Initiative (HNTI), Pathology Department, Kamuzu University of Health Sciences, Blantyre, Malawi
| | - Chisomo L Msefula
- Department of Pathology, Kamuzu University of Health Sciences, Blantyre, Malawi
- Helse Nord Tuberculosis Initiative (HNTI), Pathology Department, Kamuzu University of Health Sciences, Blantyre, Malawi
- Africa Centre for Public Health and Herbal Medicines (ACEPHEM), Kamuzu University of Health Sciences, Blantyre, Malawi
| | - Derek Sloan
- Division of Infection and Global Health, School of Medicine, University of St Andrews, St Andrews, KY16 9TF, UK
| | - Wilber Sabiiti
- Division of Infection and Global Health, School of Medicine, University of St Andrews, St Andrews, KY16 9TF, UK.
| |
Collapse
|
16
|
Hai HT, Thanh Hoang Nhat L, Tram TTB, Vinh DD, Nath AP, Donovan J, Thu NTA, Van Thanh D, Bang ND, Ha DTM, Phu NH, Nghia HDT, Van LH, Inouye M, Thwaites GE, Thuong Thuong NT. Whole blood transcriptional profiles and the pathogenesis of tuberculous meningitis. eLife 2024; 13:RP92344. [PMID: 39475467 PMCID: PMC11524586 DOI: 10.7554/elife.92344] [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] [Indexed: 11/02/2024] Open
Abstract
Mortality and morbidity from tuberculous meningitis (TBM) are common, primarily due to inflammatory response to Mycobacterium tuberculosis infection, yet the underlying mechanisms remain poorly understood. We aimed to uncover genes and pathways associated with TBM pathogenesis and mortality, and determine the best predictors of death, utilizing whole-blood RNA sequencing from 281 Vietnamese adults with TBM, 295 pulmonary tuberculosis (PTB), and 30 healthy controls. Through weighted gene co-expression network analysis, we identified hub genes and pathways linked to TBM severity and mortality, with a consensus analysis revealing distinct patterns between HIV-positive and HIV-negative individuals. We employed multivariate elastic-net Cox regression to select candidate predictors of death, then logistic regression and internal bootstrap validation to choose best predictors. Increased neutrophil activation and decreased T and B cell activation pathways were associated with TBM mortality. Among HIV-positive individuals, mortality associated with increased angiogenesis, while HIV-negative individuals exhibited elevated TNF signaling and impaired extracellular matrix organization. Four hub genes-MCEMP1, NELL2, ZNF354C, and CD4-were strong TBM mortality predictors. These findings indicate that TBM induces a systemic inflammatory response similar to PTB, highlighting critical genes and pathways related to death, offering insights for potential therapeutic targets alongside a novel four-gene biomarker for predicting outcomes.
Collapse
Affiliation(s)
- Hoang Thanh Hai
- Oxford University Clinical Research UnitHo Chi Minh CityViet Nam
| | | | | | - Do Dinh Vinh
- Oxford University Clinical Research UnitHo Chi Minh CityViet Nam
| | - Artika P Nath
- Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes InstituteMelbourneAustralia
| | - Joseph Donovan
- Oxford University Clinical Research UnitHo Chi Minh CityViet Nam
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of OxfordOxfordUnited Kingdom
| | | | - Dang Van Thanh
- Oxford University Clinical Research UnitHo Chi Minh CityViet Nam
| | | | | | - Nguyen Hoan Phu
- Oxford University Clinical Research UnitHo Chi Minh CityViet Nam
- Hospital for Tropical DiseasesHo Chi Minh CityViet Nam
| | - Ho Dang Trung Nghia
- Oxford University Clinical Research UnitHo Chi Minh CityViet Nam
- Hospital for Tropical DiseasesHo Chi Minh CityViet Nam
- Pham Ngoc Thach University of MedicineHo Chi Minh CityViet Nam
| | - Le Hong Van
- Oxford University Clinical Research UnitHo Chi Minh CityViet Nam
| | - Michael Inouye
- Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes InstituteMelbourneAustralia
- Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of CambridgeCambridgeUnited Kingdom
| | - Guy E Thwaites
- Oxford University Clinical Research UnitHo Chi Minh CityViet Nam
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of OxfordOxfordUnited Kingdom
| | - Nguyen Thuy Thuong Thuong
- Oxford University Clinical Research UnitHo Chi Minh CityViet Nam
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of OxfordOxfordUnited Kingdom
| |
Collapse
|
17
|
Ji Z, Bi S, Lu B, Zheng L, Jin X, Huang S, Jiang L, Wang Y, Ding C, Xu K. Diagnostic Efficiency of the Blood-Based Cepheid 3-Gene Host Response Test and Urine-Based Lipoarabinomannan for Active Tuberculosis Case Detection at a General Hospital in China. Infect Drug Resist 2024; 17:4467-4475. [PMID: 39435459 PMCID: PMC11492898 DOI: 10.2147/idr.s484123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 10/01/2024] [Indexed: 10/23/2024] Open
Abstract
Objective To assess the diagnostic performance of the blood-based Cepheid 3-gene Host Response test (MTB-HR), urine-based Lipoarabinomannan (LAM), and a combination of MTB-HR and LAM (MTB-HR & LAM) for detecting active tuberculosis (ATB). Methods All participants were recruited from the First Affiliated Hospital, Zhejiang University School of Medicine, between June 8, 2023 and September 13, 2023. Subsequently, the participants were classified into the ATB group or non-active tuberculosis (non-ATB) group based on microbiological evidence. MTB-HR and LAM tests were performed using fingerstick blood and urine samples from each participant, respectively. The diagnostic performance of the tests was evaluated based on the sensitivity, specificity, Youden index, and Kappa value. Pairwise comparisons of the areas under the receiver operating characteristic curves (AUROCs) between different tests were conducted using nonparametric methods. Results A total of 297 participants were included. The MTB-HR test demonstrated diagnostic efficacy with a sensitivity of 77.37% (95% CI: 70.37-84.38) and a specificity of 85.63% (95% CI: 80.19-91.06). The LAM test demonstrated a high specificity of 97.50% (95% CI: 95.08-99.92), albeit with a lower sensitivity of 54.74% (95% CI: 46.41-63.082). The sensitivity and specificity of the MTB-HR & LAM were 83.21% (95% CI: 76.95-89.47) and 83.13% (95% CI: 77.32-88.93), respectively. Only MTB-HR & LAM exhibited higher values of area under the receiver operating characteristic curve than the LAM test (MTB-HR & LAM vs LAM: 0.83 vs 0.76, P=0.0031). Conclusion In this study, although both non-sputum-based triage MTB-HR and LAM do not meet the WHO diagnostic target currently, they show possible values for triage and diagnosis in ATB. Compared to single MTB-HR or LAM test, the combined MTB-HR & LAM does not demonstrate advantages.
Collapse
Affiliation(s)
- Zhongkang Ji
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, People’s Republic of China
| | - Sheng Bi
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, People’s Republic of China
| | - Bin Lu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, People’s Republic of China
- Department of Infectious Diseases, Affiliated Dongyang Hospital of Wenzhou Medical University, Dongyang, Zhejiang, People’s Republic of China
| | - Lin Zheng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, People’s Republic of China
| | - Xiuyuan Jin
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, People’s Republic of China
| | - Shujuan Huang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, People’s Republic of China
| | - Liangxiu Jiang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, People’s Republic of China
| | - Yuping Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, People’s Republic of China
| | - Cheng Ding
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, People’s Republic of China
| | - Kaijin Xu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, People’s Republic of China
| |
Collapse
|
18
|
Schildkraut JA, Köhler N, Lange C, Duarte R, Gillespie SH. Advances in tuberculosis biomarkers: unravelling risk factors, active disease and treatment success. Breathe (Sheff) 2024; 20:240003. [PMID: 39660087 PMCID: PMC11629168 DOI: 10.1183/20734735.0003-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 08/23/2024] [Indexed: 12/12/2024] Open
Abstract
Tuberculosis (TB) is a major global health threat and demands improved diagnostic and treatment monitoring methods. Conventional diagnostics, such as sputum smear microscopy and culture, are limited by slow results and low sensitivity, particularly in certain patient groups. Recent advances in biomarker research offer promising solutions in three key areas: risk of disease, diagnosis of active disease and monitoring of treatment response. For risk assessment, novel genetic signatures and metabolites show potential in predicting the progression from TB infection to active TB. A 16-gene signature, for example, predicts this progression with significant accuracy. In diagnosing active TB, RNA-based transcriptomic signatures provide higher diagnostic accuracy than traditional methods. These signatures, such as a three-gene RNA sequence, effectively differentiate active TB from other diseases and infections, addressing issues of specificity and sensitivity. Monitoring treatment response is crucial, given the varying response rates in treating TB. Emerging biomarkers focus on bacterial burden and host response. They offer more precise and timely assessments of treatment efficacy, enhance personalised treatment approaches and potentially improve patient outcomes. These advancements in biomarkers for TB risk, diagnosis and treatment response represent significant steps towards more effective TB management and control, aligning with global efforts to decrease the burden of TB. Here we aim to highlight several promising biomarkers used to predict risk of disease progression, active TB disease and treatment success.
Collapse
Affiliation(s)
- Jodie A. Schildkraut
- Department of Pulmonary Disease, Radboudumc Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
- J.A. Schildkraut and N. Köhler contributed equally as first authors
| | - Niklas Köhler
- Division of Clinical Infectious Diseases, Research Center Borstel, Borstel, Germany
- German Center for Infection Research (DZIF), Partner Site Borstel-Hamburg-Lübeck-Riems, Borstel, Germany
- Respiratory Medicine and International Health, University of Lübeck, Lübeck, Germany
- J.A. Schildkraut and N. Köhler contributed equally as first authors
| | - Christoph Lange
- Division of Clinical Infectious Diseases, Research Center Borstel, Borstel, Germany
- German Center for Infection Research (DZIF), Partner Site Borstel-Hamburg-Lübeck-Riems, Borstel, Germany
- Respiratory Medicine and International Health, University of Lübeck, Lübeck, Germany
- Department of Pediatrics, Global and Immigrant Health, Global Tuberculosis Program, Baylor College of Medicine and Texas Children's Hospital, Houston, TX, USA
| | - Raquel Duarte
- EPI Unit, Instituto de Saúde Pública da Universidade do Porto, Porto, Portugal
- Laboratório associado para a Investigação Integrativa e Translacional em Saúde Populacional (ITR), Porto, Portugal
- Departamento de Estudos das Populações, Instituto de Ciências Biomédicas Abel Salazar, Porto, Portugal
- INSA, Instituto de Saúde Pública Doutor Ricardo Jorge, INSA Porto, Portugal
| | - Stephen H. Gillespie
- Division of Infection and Global Health, School of Medicine, University of St Andrews, St Andrews, UK
| |
Collapse
|
19
|
Wei Z, Chen Y, Dong P, Liu Z, Lai X, Wang N, Li H, Wang Q, Tao L, Su N, Yang Y, Meng F. CXCL9/CXCL10 as biomarkers the monitoring of treatment responses in Pulmonary TB patients: a systematic review and meta-analysis. BMC Infect Dis 2024; 24:1037. [PMID: 39333908 PMCID: PMC11428339 DOI: 10.1186/s12879-024-09939-0] [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: 03/05/2024] [Accepted: 09/16/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Tuberculosis (TB) remains a persistent threat to global public health and traditional treatment monitoring approaches are limited by their potential for contamination and need for timely evaluation. Therefore, new biomarkers are urgently required for monitoring the treatment efficacy of TB. METHODS This study aimed to elucidate the levels of CXCL10 and CXCL9 in pulmonary TB patients who underwent anti-TB treatment. The data was acquired from five databases, including PubMed, Ovid, Web of Science, Embase, and the Cochrane Library. A meta-analysis of CXCL10 data from all time points was conducted. Furthermore, a trend meta-analysis of temporal data of CXCL10 and CXCL9 from multiple time points was also performed. RESULTS It was revealed that patients who responded poorly to anti-TB treatment had higher serum levels relative to those who responded well (SMD: 1.23, 95% CI: -0.37-2.84) at the end of intensive treatment (2 months). Furthermore, heterogeneity was observed in these results, which might be because patients with a prior history of TB and different treatment monitoring methods than those selected in this study were also included. The analysis of alterations in CXCL10 and CXCL9 levels since the last collection time points indicated that their levels reduced with time. CONCLUSION In summary, the study revealed that reductions in CXCL10 levels during the first two months of anti-TB treatment are correlated with treatment responses. Furthermore, decreasing levels of CXCL9 during the treatment suggest that it may also serve as a biomarker with a similar value to CXCL10. Future in-depth studies are thus warranted to further probe the relevance of CXCL10 and CXCL9 in monitoring the treatment efficacy of TB.
Collapse
Grants
- 2022YFC2304800 the National Key Research and Development Program of China
- 2022YFC2304800 the National Key Research and Development Program of China
- 2022YFC2304800 the National Key Research and Development Program of China
- 2022YFC2304800 the National Key Research and Development Program of China
- 2022YFC2304800 the National Key Research and Development Program of China
- 2022YFC2304800 the National Key Research and Development Program of China
- 2022YFC2304800 the National Key Research and Development Program of China
- 2022YFC2304800 the National Key Research and Development Program of China
- 2022YFC2304800 the National Key Research and Development Program of China
- 2022YFC2304800 the National Key Research and Development Program of China
- 2022YFC2304800 the National Key Research and Development Program of China
- 2022YFC2304800 the National Key Research and Development Program of China
- 202002030152, 202102020910, 202206010134, 202201010697, 2023A03J0539, 2023A03J0992 Guangzhou Science and Technology Planning Project
- 202002030152, 202102020910, 202206010134, 202201010697, 2023A03J0539, 2023A03J0992 Guangzhou Science and Technology Planning Project
- 202002030152, 202102020910, 202206010134, 202201010697, 2023A03J0539, 2023A03J0992 Guangzhou Science and Technology Planning Project
- 202002030152, 202102020910, 202206010134, 202201010697, 2023A03J0539, 2023A03J0992 Guangzhou Science and Technology Planning Project
- 202002030152, 202102020910, 202206010134, 202201010697, 2023A03J0539, 2023A03J0992 Guangzhou Science and Technology Planning Project
- 202002030152, 202102020910, 202206010134, 202201010697, 2023A03J0539, 2023A03J0992 Guangzhou Science and Technology Planning Project
- 202002030152, 202102020910, 202206010134, 202201010697, 2023A03J0539, 2023A03J0992 Guangzhou Science and Technology Planning Project
- 202002030152, 202102020910, 202206010134, 202201010697, 2023A03J0539, 2023A03J0992 Guangzhou Science and Technology Planning Project
- 202002030152, 202102020910, 202206010134, 202201010697, 2023A03J0539, 2023A03J0992 Guangzhou Science and Technology Planning Project
- 202002030152, 202102020910, 202206010134, 202201010697, 2023A03J0539, 2023A03J0992 Guangzhou Science and Technology Planning Project
- 202002030152, 202102020910, 202206010134, 202201010697, 2023A03J0539, 2023A03J0992 Guangzhou Science and Technology Planning Project
- 202002030152, 202102020910, 202206010134, 202201010697, 2023A03J0539, 2023A03J0992 Guangzhou Science and Technology Planning Project
- 2018A030313550, 2023A1515010461 Guangdong Natural Science Foundation Project
- 2018A030313550, 2023A1515010461 Guangdong Natural Science Foundation Project
- 2018A030313550, 2023A1515010461 Guangdong Natural Science Foundation Project
- 2018A030313550, 2023A1515010461 Guangdong Natural Science Foundation Project
- 2018A030313550, 2023A1515010461 Guangdong Natural Science Foundation Project
- 2018A030313550, 2023A1515010461 Guangdong Natural Science Foundation Project
- 2018A030313550, 2023A1515010461 Guangdong Natural Science Foundation Project
- 2018A030313550, 2023A1515010461 Guangdong Natural Science Foundation Project
- 2018A030313550, 2023A1515010461 Guangdong Natural Science Foundation Project
- 2018A030313550, 2023A1515010461 Guangdong Natural Science Foundation Project
- 2018A030313550, 2023A1515010461 Guangdong Natural Science Foundation Project
- 2018A030313550, 2023A1515010461 Guangdong Natural Science Foundation Project
- 20231A011051, 20241A011049 Guangzhou Health Science and Technology Project
- 20231A011051, 20241A011049 Guangzhou Health Science and Technology Project
- 20231A011051, 20241A011049 Guangzhou Health Science and Technology Project
- 20231A011051, 20241A011049 Guangzhou Health Science and Technology Project
- 20231A011051, 20241A011049 Guangzhou Health Science and Technology Project
- 20231A011051, 20241A011049 Guangzhou Health Science and Technology Project
- 20231A011051, 20241A011049 Guangzhou Health Science and Technology Project
- 20231A011051, 20241A011049 Guangzhou Health Science and Technology Project
- 20231A011051, 20241A011049 Guangzhou Health Science and Technology Project
- 20231A011051, 20241A011049 Guangzhou Health Science and Technology Project
- 20231A011051, 20241A011049 Guangzhou Health Science and Technology Project
- 20231A011051, 20241A011049 Guangzhou Health Science and Technology Project
- 20231251 Guangdong Bureau of Traditional Chinese Medicine Scientific Research Project
- 20231251 Guangdong Bureau of Traditional Chinese Medicine Scientific Research Project
- 20231251 Guangdong Bureau of Traditional Chinese Medicine Scientific Research Project
- 20231251 Guangdong Bureau of Traditional Chinese Medicine Scientific Research Project
- 20231251 Guangdong Bureau of Traditional Chinese Medicine Scientific Research Project
- 20231251 Guangdong Bureau of Traditional Chinese Medicine Scientific Research Project
- 20231251 Guangdong Bureau of Traditional Chinese Medicine Scientific Research Project
- 20231251 Guangdong Bureau of Traditional Chinese Medicine Scientific Research Project
- 20231251 Guangdong Bureau of Traditional Chinese Medicine Scientific Research Project
- 20231251 Guangdong Bureau of Traditional Chinese Medicine Scientific Research Project
- 20231251 Guangdong Bureau of Traditional Chinese Medicine Scientific Research Project
- 20231251 Guangdong Bureau of Traditional Chinese Medicine Scientific Research Project
- A2023284 Guangdong Medical Science and Technology Research Fund Project
- A2023284 Guangdong Medical Science and Technology Research Fund Project
- A2023284 Guangdong Medical Science and Technology Research Fund Project
- A2023284 Guangdong Medical Science and Technology Research Fund Project
- A2023284 Guangdong Medical Science and Technology Research Fund Project
- A2023284 Guangdong Medical Science and Technology Research Fund Project
- A2023284 Guangdong Medical Science and Technology Research Fund Project
- A2023284 Guangdong Medical Science and Technology Research Fund Project
- A2023284 Guangdong Medical Science and Technology Research Fund Project
- A2023284 Guangdong Medical Science and Technology Research Fund Project
- A2023284 Guangdong Medical Science and Technology Research Fund Project
Collapse
Affiliation(s)
- Zeyou Wei
- State Key Laboratory of Respiratory Disease, Guangzhou Key Laboratory of Tuberculosis Research, Institute of Pulmonary Diseases, Guangzhou Chest Hospital, Institute of Tuberculosis, Guangzhou Medical University, 62 Hengzhigang Rd, Yuexiu District, Guangzhou, 510095, People's Republic of China
| | - Yuanjin Chen
- State Key Laboratory of Respiratory Disease, Guangzhou Key Laboratory of Tuberculosis Research, Institute of Pulmonary Diseases, Guangzhou Chest Hospital, Institute of Tuberculosis, Guangzhou Medical University, 62 Hengzhigang Rd, Yuexiu District, Guangzhou, 510095, People's Republic of China
| | - Pengyan Dong
- State Key Laboratory of Respiratory Disease, Guangzhou Key Laboratory of Tuberculosis Research, Institute of Pulmonary Diseases, Guangzhou Chest Hospital, Institute of Tuberculosis, Guangzhou Medical University, 62 Hengzhigang Rd, Yuexiu District, Guangzhou, 510095, People's Republic of China
| | - Zhihui Liu
- State Key Laboratory of Respiratory Disease, Guangzhou Key Laboratory of Tuberculosis Research, Institute of Pulmonary Diseases, Guangzhou Chest Hospital, Institute of Tuberculosis, Guangzhou Medical University, 62 Hengzhigang Rd, Yuexiu District, Guangzhou, 510095, People's Republic of China
| | - Xiaomin Lai
- State Key Laboratory of Respiratory Disease, Guangzhou Key Laboratory of Tuberculosis Research, Institute of Pulmonary Diseases, Guangzhou Chest Hospital, Institute of Tuberculosis, Guangzhou Medical University, 62 Hengzhigang Rd, Yuexiu District, Guangzhou, 510095, People's Republic of China
- School of Public Health, Sun Yat-sen University, Shen Zhen, China
| | - Nan Wang
- State Key Laboratory of Respiratory Disease, Guangzhou Key Laboratory of Tuberculosis Research, Institute of Pulmonary Diseases, Guangzhou Chest Hospital, Institute of Tuberculosis, Guangzhou Medical University, 62 Hengzhigang Rd, Yuexiu District, Guangzhou, 510095, People's Republic of China
| | - Hua Li
- State Key Laboratory of Respiratory Disease, Guangzhou Key Laboratory of Tuberculosis Research, Institute of Pulmonary Diseases, Guangzhou Chest Hospital, Institute of Tuberculosis, Guangzhou Medical University, 62 Hengzhigang Rd, Yuexiu District, Guangzhou, 510095, People's Republic of China
| | - Qi Wang
- State Key Laboratory of Respiratory Disease, Guangzhou Key Laboratory of Tuberculosis Research, Institute of Pulmonary Diseases, Guangzhou Chest Hospital, Institute of Tuberculosis, Guangzhou Medical University, 62 Hengzhigang Rd, Yuexiu District, Guangzhou, 510095, People's Republic of China
| | - Lan Tao
- State Key Laboratory of Respiratory Disease, Guangzhou Key Laboratory of Tuberculosis Research, Department of Tuberculosis, Guangzhou Chest Hospital, Institute of Tuberculosis, Guangzhou Medical University, Guangzhou, P.R. China
| | - Ning Su
- State Key Laboratory of Respiratory Disease, Guangzhou Key Laboratory of Tuberculosis Research, Department of Oncology, Guangzhou Chest Hospital, Institute of Tuberculosis, Guangzhou Medical University, Guangzhou, P.R. China
| | - Yu Yang
- State Key Laboratory of Respiratory Disease, Guangzhou Key Laboratory of Tuberculosis Research, Institute of Pulmonary Diseases, Guangzhou Chest Hospital, Institute of Tuberculosis, Guangzhou Medical University, 62 Hengzhigang Rd, Yuexiu District, Guangzhou, 510095, People's Republic of China.
| | - Fanrong Meng
- State Key Laboratory of Respiratory Disease, Guangzhou Key Laboratory of Tuberculosis Research, Institute of Pulmonary Diseases, Guangzhou Chest Hospital, Institute of Tuberculosis, Guangzhou Medical University, 62 Hengzhigang Rd, Yuexiu District, Guangzhou, 510095, People's Republic of China.
| |
Collapse
|
20
|
Vito O, Psarras S, Syggelou A, Wright VJ, Amanatidou V, Newton SM, Shailes H, Trochoutsou K, Tsagaraki M, Levin M, Kaforou M, Tsolia M. Novel RNA biomarkers improve discrimination of children with tuberculosis disease from those with non-TB pneumonia after in vitro stimulation. Front Immunol 2024; 15:1401647. [PMID: 39391304 PMCID: PMC11464340 DOI: 10.3389/fimmu.2024.1401647] [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: 03/15/2024] [Accepted: 09/04/2024] [Indexed: 10/12/2024] Open
Abstract
The diagnosis of pediatric tuberculosis (TB) poses a challenge for clinical teams worldwide. TB-mediated changes in the expression of host genes in the peripheral blood can serve as diagnostic biomarkers and can provide better insights into the host immune mechanisms of childhood TB. Peripheral blood mononuclear cells (PBMCs) from children (n=102) with microbiologically confirmed TB disease, TB infection (TBI), pneumonia, and healthy controls (HC) were stimulated with either the Purified Protein Derivative (PPD) or the Early Secretory Antigen 6kDa-Culture Filtrate Protein 10 (ESAT6-CFP10) complex of Mycobacterium tuberculosis (Mtb). RNA was extracted and quantified using gene expression microarrays. Differential expression analysis was performed comparing microbiologically confirmed TB to the other diagnostic groups for the stimulated and unstimulated samples. Using variable selection, we identified sparse diagnostic gene signatures; one gene (PID1) was able to distinguish TB from pneumonia after ESAT6-CFP10 stimulation with an AUC of 100% in the test set, while a combination of two genes (STAT1 and IFI44) achieved an AUC of 91.7% (CI95% 75.0%-100%) in the test set after PPD stimulation. The number of significantly differentially expressed (SDE) genes was higher when contrasting TB to pneumonia or HC in stimulated samples, compared to unstimulated ones, leading to a larger pool of candidate diagnostic biomarkers. Our approach provides enlightened aspects of peripheral TB-specific responses and can form the basis for a point of care test meeting the World Health Organization (WHO) Target Product Profile (TPP) for pediatric TB.
Collapse
Affiliation(s)
- Ortensia Vito
- Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, United Kingdom
- Centre for Pediatrics and Child Health, Imperial College London, London, United Kingdom
| | - Stelios Psarras
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens , Athens, Greece
| | - Angeliki Syggelou
- Second Department of Pediatrics, National and Kapodistrian University of Athens (NKUA), School of Medicine, P. and A. Kyriakou Children’s Hospital, Athens, Greece
| | - Victoria J. Wright
- Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, United Kingdom
- Centre for Pediatrics and Child Health, Imperial College London, London, United Kingdom
| | - Virginia Amanatidou
- Second Department of Pediatrics, National and Kapodistrian University of Athens (NKUA), School of Medicine, P. and A. Kyriakou Children’s Hospital, Athens, Greece
| | - Sandra M. Newton
- Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, United Kingdom
- Centre for Pediatrics and Child Health, Imperial College London, London, United Kingdom
| | - Hannah Shailes
- Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, United Kingdom
- Centre for Pediatrics and Child Health, Imperial College London, London, United Kingdom
| | - Katerina Trochoutsou
- Second Department of Pediatrics, National and Kapodistrian University of Athens (NKUA), School of Medicine, P. and A. Kyriakou Children’s Hospital, Athens, Greece
| | - Maria Tsagaraki
- Second Department of Pediatrics, National and Kapodistrian University of Athens (NKUA), School of Medicine, P. and A. Kyriakou Children’s Hospital, Athens, Greece
| | - Michael Levin
- Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, United Kingdom
- Centre for Pediatrics and Child Health, Imperial College London, London, United Kingdom
| | - Myrsini Kaforou
- Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, United Kingdom
- Centre for Pediatrics and Child Health, Imperial College London, London, United Kingdom
| | - Maria Tsolia
- Second Department of Pediatrics, National and Kapodistrian University of Athens (NKUA), School of Medicine, P. and A. Kyriakou Children’s Hospital, Athens, Greece
| |
Collapse
|
21
|
Fiore-Gartland A, Srivastava H, Seese A, Day T, Penn-Nicholson A, Luabeya AKK, Du Plessis N, Loxton AG, Bekker LG, Diacon A, Walzl G, Sagawa ZK, Reed SG, Scriba TJ, Hatherill M, Coler R. Co-regulation of innate and adaptive immune responses induced by ID93+GLA-SE vaccination in humans. Front Immunol 2024; 15:1441944. [PMID: 39381003 PMCID: PMC11458388 DOI: 10.3389/fimmu.2024.1441944] [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: 05/31/2024] [Accepted: 09/02/2024] [Indexed: 10/10/2024] Open
Abstract
Introduction Development of an effective vaccine against tuberculosis is a critical step towards reducing the global burden of disease. A therapeutic vaccine might also reduce the high rate of TB recurrence and help address the challenges of drug-resistant strains. ID93+GLA-SE is a candidate subunit vaccine that will soon be evaluated in a phase 2b efficacy trial for prevention of recurrent TB among patients undergoing TB treatment. ID93+GLA-SE vaccination was shown to elicit robust CD4+ T cell and IgG antibody responses among recently treated TB patients in the TBVPX-203 Phase 2a study (NCT02465216), but the mechanisms underlying these responses are not well understood. Methods In this study we used specimens from TBVPX-203 participants to describe the changes in peripheral blood gene expression that occur after ID93+GLA-SE vaccination. Results Analyses revealed several distinct modules of co-varying genes that were either up- or down-regulated after vaccination, including genes associated with innate immune pathways at 3 days post-vaccination and genes associated with lymphocyte expansion and B cell activation at 7 days post-vaccination. Notably, the regulation of these gene modules was affected by the dose schedule and by participant sex, and early innate gene signatures were correlated with the ID93-specific CD4+ T cell response. Discussion The results provide insight into the complex interplay of the innate and adaptive arms of the immune system in developing responses to vaccination with ID93+GLA-SE and demonstrate how dosing and schedule can affect vaccine responses.
Collapse
Affiliation(s)
- Andrew Fiore-Gartland
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - Himangi Srivastava
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - Aaron Seese
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - Tracey Day
- Infectious Diseases and Vaccines, Innovative Medicine, Johnson & Johnson, Leiden, Netherlands
| | | | - Angelique Kany Kany Luabeya
- South African Tuberculosis Vaccine Initiative (SATVI), Institute of Infectious Disease & Molecular Medicine and Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Nelita Du Plessis
- Department of Science and Technology/National Research Foundation (DST-NRF) Centre of Excellence for Biomedical Tuberculosis Research, 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
| | - Andre G. Loxton
- Department of Science and Technology/National Research Foundation (DST-NRF) Centre of Excellence for Biomedical Tuberculosis Research, 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
| | - Linda-Gail Bekker
- The Desmond Tutu Human Immunodeficiency Virus (HIV) Centre, University of Cape Town, Cape Town, South Africa
| | | | - Gerhard Walzl
- Department of Science and Technology/National Research Foundation (DST-NRF) Centre of Excellence for Biomedical Tuberculosis Research, 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
| | | | | | - Thomas J. Scriba
- South African Tuberculosis Vaccine Initiative (SATVI), Institute of Infectious Disease & Molecular Medicine and Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Mark Hatherill
- South African Tuberculosis Vaccine Initiative (SATVI), Institute of Infectious Disease & Molecular Medicine and Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Rhea Coler
- Seattle Children’s Research Institute, Center for Global Infectious Disease Research, Seattle Children’s, 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
| |
Collapse
|
22
|
Xu W, Yang J, Yu H, Li S. Diagnostic value of lncRNAs LINC00152 and LARS2-AS1 and their regulatory roles in macrophage immune response in tuberculosis. Tuberculosis (Edinb) 2024; 148:102530. [PMID: 38857553 DOI: 10.1016/j.tube.2024.102530] [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: 12/28/2023] [Revised: 06/04/2024] [Accepted: 06/05/2024] [Indexed: 06/12/2024]
Abstract
OBJECTIVES To determine the usefulness of LINC00152 and LARS2-AS1 as potential biomarkers for latent tuberculosis (LTB) and active tuberculosis (ATB), as well as their effect on Mycobacterium (Mtb) infection. METHODS The expression levels of LINC00152 and LARS2-AS1 in the health, patients with LTB and ATB were detected by qRT-PCR. The ROC curves were constructed to show their potential as biomarkers. The intracellular survival assays for Mtb and the levels of immune-related cytokines were determined to discover the effect of LINC00152 and LARS2-AS1 on Mtb infection. The relationships of miR-485-5p with LINC00152 and LARS2-AS1 were explored. RESULTS LINC00152 and LARS2-AS1 levels were significantly elevated in patients with ATB and LTB, and Mtb-infected macrophages. LINC00152 and LARS2-AS1 can distinguish the LTB from the health and ATB from LTB. LARS2-AS1 and LINC00152 knock-down reduced the intracellular Mtb survival and induced cellular immune response after Mtb challenge. miR-485-5p was a targeting miRNA for LINC00152 and LARS2-AS1. CONCLUSIONS LINC00152 and LARS2-AS1 can be considered as potential biomarkers for tuberculosis disease. LINC00152 and LARS2-AS1 have anti-Mtb effects.
Collapse
Affiliation(s)
- Wenlong Xu
- Department of Clinical Laboratory, Shanghai Yangsi Hospital, Shanghai, 200126, China
| | - Jihua Yang
- Department of Ultrasound, Central Hospital Affiliated to Shandong First Medical University, Jinan, 250013, China
| | - Haizhen Yu
- Department of Clinical Laboratory, Zhucheng People's Hospital, Zhucheng, 262299, China
| | - Shizhen Li
- Department of Clinical Laboratory, Zhucheng People's Hospital, Zhucheng, 262299, China.
| |
Collapse
|
23
|
Huynh J, Nhat LHT, Bao NLH, Hai HT, Thu DDA, Tram TTB, Dung VTM, Vinh DD, Ngoc NM, Donovan J, Phu NH, Van Thanh D, Thu NTA, Bang ND, Ha DTM, Nghia HDT, Van Tan L, Van LH, Thwaites G, Thuong NTT. The Ability of a 3-Gene Host Signature in Blood to Distinguish Tuberculous Meningitis From Other Brain Infections. J Infect Dis 2024; 230:e268-e278. [PMID: 38169323 PMCID: PMC11326836 DOI: 10.1093/infdis/jiad606] [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: 08/07/2023] [Revised: 12/10/2023] [Accepted: 12/29/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Tuberculous meningitis (TBM) is difficult to diagnose. We investigated whether a 3-gene host response signature in blood can distinguish TBM from other brain infections. METHODS The expression of 3 genes (dual specificity phosphatase 3 [DUSP3], guanylate-binding protein [GBP5], krupple-like factor 2 [KLF2]) was analyzed by RNA sequencing of archived whole blood from 4 cohorts of Vietnamese adults: 281 with TBM, 279 with pulmonary tuberculosis, 50 with other brain infections, and 30 healthy controls. Tuberculosis scores (combined 3-gene expression) were calculated following published methodology and discriminatory performance compared using area under a receiver operator characteristic curve (AUC). RESULTS GBP5 was upregulated in TBM compared to other brain infections (P < .001), with no difference in DUSP3 and KLF2 expression. The diagnostic performance of GBP5 alone (AUC, 0.74; 95% confidence interval [CI], .67-.81) was slightly better than the 3-gene tuberculosis score (AUC, 0.66; 95% CI, .58-.73) in TBM. Both GBP5 expression and tuberculosis score were higher in participants with human immunodeficiency virus (HIV; P < .001), with good diagnostic performance of GBP5 alone (AUC, 0.86; 95% CI, .80-.93). CONCLUSIONS The 3-gene host signature in whole blood has the ability to discriminate TBM from other brain infections, including in individuals with HIV. Validation in large prospective diagnostic study is now required.
Collapse
Affiliation(s)
- Julie Huynh
- Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, Oxford University, OxfordUnited Kingdom
| | | | | | - Hoang Thanh Hai
- Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam
| | - Do Dang Anh Thu
- Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam
| | | | - Vu Thi Mong Dung
- Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam
| | - Do Dinh Vinh
- Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam
| | - Nghiem My Ngoc
- Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam
| | - Joseph Donovan
- Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, Oxford University, OxfordUnited Kingdom
| | - Nguyen Hoan Phu
- Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam
- School of Medicine, Vietnam National University of Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Dang Van Thanh
- Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam
| | | | - Nguyen Duc Bang
- Pham Ngoc Thach Hospital for Tuberculosis and Lung Disease, Ho Chi Minh City, Vietnam
| | - Dang Thi Minh Ha
- Pham Ngoc Thach Hospital for Tuberculosis and Lung Disease, Ho Chi Minh City, Vietnam
| | - Ho Dang Trung Nghia
- Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam
- Pham Ngoc Thach University of Medicine, Ho Chi Minh City, Vietnam
- The Hospital for Tropical Diseases, Ho Chi Minh City, Vietnam
| | - Le Van Tan
- Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, Oxford University, OxfordUnited Kingdom
| | - Le Hong Van
- Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam
| | - Guy Thwaites
- Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, Oxford University, OxfordUnited Kingdom
| | - Nguyen Thuy Thuong Thuong
- Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, Oxford University, OxfordUnited Kingdom
| |
Collapse
|
24
|
Li Z, Hu Y, Wang W, Zou F, Yang J, Gao W, Feng S, Chen G, Shi C, Cai Y, Deng G, Chen X. Integrating pathogen- and host-derived blood biomarkers for enhanced tuberculosis diagnosis: a comprehensive review. Front Immunol 2024; 15:1438989. [PMID: 39185416 PMCID: PMC11341448 DOI: 10.3389/fimmu.2024.1438989] [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: 05/27/2024] [Accepted: 07/24/2024] [Indexed: 08/27/2024] Open
Abstract
This review explores the evolving landscape of blood biomarkers in the diagnosis of tuberculosis (TB), focusing on biomarkers derived both from the pathogen and the host. These biomarkers provide critical insights that can improve diagnostic accuracy and timeliness, essential for effective TB management. The document highlights recent advancements in molecular techniques that have enhanced the detection and characterization of specific biomarkers. It also discusses the integration of these biomarkers into clinical practice, emphasizing their potential to revolutionize TB diagnostics by enabling more precise detection and monitoring of the disease progression. Challenges such as variability in biomarker expression and the need for standardized validation processes are addressed to ensure reliability across different populations and settings. The review calls for further research to refine these biomarkers and fully harness their potential in the fight against TB, suggesting a multidisciplinary approach to overcome existing barriers and optimize diagnostic strategies. This comprehensive analysis underscores the significance of blood biomarkers as invaluable tools in the global effort to control and eliminate TB.
Collapse
Affiliation(s)
- Zhaodong Li
- Guangdong Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University School of Medicine, Shenzhen, China
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, China
| | - Yunlong Hu
- Guangdong Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University School of Medicine, Shenzhen, China
| | - Wenfei Wang
- National Clinical Research Center for Infectious Disease, The Third People's Hospital of Shenzhen, Southern University of Science and Technology, Shenzhen, China
| | - Fa Zou
- Guangdong Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University School of Medicine, Shenzhen, China
| | - Jing Yang
- Guangdong Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University School of Medicine, Shenzhen, China
| | - Wei Gao
- Guangdong Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University School of Medicine, Shenzhen, China
| | - SiWan Feng
- Guangdong Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University School of Medicine, Shenzhen, China
| | - Guanghuan Chen
- Guangdong Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University School of Medicine, Shenzhen, China
| | - Chenyan Shi
- Department of Preventive Medicine, School of Public Health, Shenzhen University, Shenzhen, China
| | - Yi Cai
- Guangdong Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University School of Medicine, Shenzhen, China
| | - Guofang Deng
- Guangdong Key Lab for Diagnosis & Treatment of Emerging Infectious Diseases, Shenzhen Third People's Hospital, Shenzhen, China
| | - Xinchun Chen
- Guangdong Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University School of Medicine, Shenzhen, China
| |
Collapse
|
25
|
Muwanga VM, Mendelsohn SC, Leukes V, Stanley K, Mbandi SK, Erasmus M, Flinn M, Fisher TL, Raphela R, Bilek N, Malherbe ST, Tromp G, Van Der Spuy G, Walzl G, Chegou NN, Scriba TJ. Blood transcriptomic signatures for symptomatic tuberculosis in an African multicohort study. Eur Respir J 2024; 64:2400153. [PMID: 38964778 PMCID: PMC11325265 DOI: 10.1183/13993003.00153-2024] [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: 01/22/2024] [Accepted: 06/12/2024] [Indexed: 07/06/2024]
Abstract
BACKGROUND Multiple host blood transcriptional signatures have been developed as non-sputum triage tests for tuberculosis (TB). We aimed to compare the diagnostic performance of 20 blood transcriptomic TB signatures for differentiating between symptomatic patients who have TB versus other respiratory diseases (ORD). METHODS As part of a nested case-control study, individuals presenting with respiratory symptoms at primary healthcare clinics in Ethiopia, Malawi, Namibia, Uganda, South Africa and The Gambia were enrolled. TB was diagnosed based on clinical, microbiological and radiological findings. Transcriptomic signatures were measured in whole blood using microfluidic real-time quantitative PCR. Diagnostic performance was benchmarked against the World Health Organization Target Product Profile (TPP) for a non-sputum TB triage test. RESULTS Among 579 participants, 158 had definite, microbiologically confirmed TB, 32 had probable TB, while 389 participants had ORD. Nine signatures differentiated between ORD and TB with equivalent performance (Satproedprai7: area under the curve 0.83 (95% CI 0.79-0.87); Jacobsen3: 0.83 (95% CI 0.79-0.86); Suliman2: 0.82 (95% CI 0.78-0.86); Roe1: 0.82 (95% CI 0.78-0.86); Kaforou22: 0.82 (95% CI 0.78-0.86); Sambarey10: 0.81 (95% CI 0.77-0.85); Duffy9: 0.81 (95% CI 0.76-0.86); Gliddon3: 0.8 (95% CI 0.75-0.85); Suliman4 0.79 (95% CI 0.75-0.84)). Benchmarked against a 90% sensitivity, these signatures achieved specificities between 44% (95% CI 38-49%) and 54% (95% CI 49-59%), not meeting the TPP criteria. Signature scores significantly varied by HIV status and country. In country-specific analyses, several signatures, such as Satproedprai7 and Penn-Nicholson6, met the minimal TPP criteria for a triage test in Ethiopia, Malawi and South Africa. CONCLUSION No signatures met the TPP criteria in a pooled analysis of all countries, but several signatures met the minimum criteria for a non-sputum TB triage test in some countries.
Collapse
Affiliation(s)
- Vanessa Mwebaza Muwanga
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine, Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Simon C Mendelsohn
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine, Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Vinzeigh Leukes
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Immunology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Kim Stanley
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Immunology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Stanley Kimbung Mbandi
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine, Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Mzwandile Erasmus
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine, Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Marika Flinn
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Immunology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Tarryn-Lee Fisher
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Immunology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Rodney Raphela
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine, Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Nicole Bilek
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine, Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Stephanus T Malherbe
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Immunology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Gerard Tromp
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Immunology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Gian Van Der Spuy
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Gerhard Walzl
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Immunology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Novel N Chegou
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Immunology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Thomas J Scriba
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine, Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
26
|
Krishnan P, Bobak CA, Hill JE. Sex-specific blood-derived RNA biomarkers for childhood tuberculosis. Sci Rep 2024; 14:16859. [PMID: 39039071 PMCID: PMC11263679 DOI: 10.1038/s41598-024-66946-6] [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: 04/07/2024] [Accepted: 07/05/2024] [Indexed: 07/24/2024] Open
Abstract
Confirmatory diagnosis of childhood tuberculosis (TB) remains a challenge mainly due to its dependence on sputum samples and the paucibacillary nature of the disease. Thus, only ~ 30% of suspected cases in children are diagnosed and the need for minimally invasive, non-sputum-based biomarkers remains unmet. Understanding host molecular changes by measuring blood-based transcriptomic markers has shown promise as a diagnostic tool for TB. However, the implication of sex contributing to disease heterogeneity and therefore diagnosis remains to be understood. Using publicly available gene expression data (GSE39939, GSE39940; n = 370), we report a sex-specific RNA biomarker signature that could improve the diagnosis of TB disease in children. We found four gene biomarker signatures for male (SLAMF8, GBP2, WARS, and FCGR1C) and female pediatric patients (GBP6, CELSR3, ALDH1A1, and GBP4) from Kenya, South Africa, and Malawi. Both signatures achieved a sensitivity of 85% and a specificity of 70%, which approaches the WHO-recommended target product profile for a triage test. Our gene signatures outperform most other gene signatures reported previously for childhood TB diagnosis.
Collapse
Affiliation(s)
- Preethi Krishnan
- Department of Chemical and Biological Engineering, University of British Columbia, Vancouver, V6T 1Z3, Canada
| | - Carly A Bobak
- Department of Biomedical Data Science, Dartmouth College, Hanover, NH, 03755, USA
| | - Jane E Hill
- Department of Chemical and Biological Engineering, University of British Columbia, Vancouver, V6T 1Z3, Canada.
| |
Collapse
|
27
|
Mann T, Minnies S, Gupta RK, Reeve BWP, Nyawo G, Palmer Z, Naidoo C, Doubell A, Pecararo A, John TJ, Schubert P, Calderwood CJ, Chandran A, Theron G, Noursadeghi M. Blood RNA signatures outperform CRP triage of tuberculosis lymphadenitis and pericarditis. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.06.21.24309099. [PMID: 38946942 PMCID: PMC11213046 DOI: 10.1101/2024.06.21.24309099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Background Limited data are available on the diagnostic accuracy of blood RNA biomarker signatures for extrapulmonary TB (EPTB). We addressed this question among people investigated for TB lymphadenitis and TB pericarditis, in Cape Town, South Africa. Methods We enrolled 440 consecutive adults referred to a hospital for invasive sampling for presumptive TB lymphadenitis (n=300) or presumptive TB pericarditis (n=140). Samples from the site of disease underwent culture and/or molecular testing for Mycobacterium tuberculosis complex (Mtb). Discrimination of patients with and without TB defined by microbiology or cytology reference standards was evaluated using seven previously reported blood RNA signatures by area under the receiver-operating characteristic curve (AUROC) and sensitivity/specificity at predefined thresholds, benchmarked against blood C-reactive protein (CRP) and the World Health Organization (WHO) target product profile (TPP) for a TB triage test. Decision curve analysis (DCA) was used to evaluate the clinical utility of the best performing blood RNA signature and CRP. Results Data from 374 patients for whom results were available from at least one microbiological test from the site of disease, and blood CRP and RNA measurements, were included. Using microbiological results as the reference standard in the primary analysis (N=204 with TB), performance was similar across lymphadenitis and pericarditis patients. In the pooled analysis of both cohorts, all RNA signatures had comparable discrimination with AUROC point estimates ranging 0.77-0.82, superior to that of CRP (0.61, 95% confidence interval 0.56-0.67). The best performing signature (Roe3) achieved an AUROC of 0.82 (0.77-0.86). At a predefined threshold of 2 standard deviations (Z2) above the mean of a healthy reference control group, this signature achieved 78% (72-83%) sensitivity and 69% (62-75%) specificity. In this setting, DCA revealed that Roe3 offered greater net benefit than other approaches for services aiming to reduce the number needed to investigate with confirmatory testing to <4 to identify each case of TB. Interpretation RNA biomarkers show better accuracy and clinical utility than CRP to trigger confirmatory TB testing in patients with TB lymphadenitis and TB pericarditis, but still fall short of the WHO TPP for TB triage tests. Funding South African MRC, EDCTP2, NIH/NIAID, Wellcome Trust, NIHR, Royal College of Physicians London. Research in context Evidence before this study: Blood RNA biomarker signatures and CRP measurements have emerged as potential triage tests for TB, but evidence is mostly limited to their performance in pulmonary TB. Microbiological diagnosis of extrapulmonary TB (EPTB) is made challenging by the need for invasive sampling to obtain tissue from the site of disease. This is compounded by lower sensitivity of confirmatory molecular tests for EPTB compared to their performance in pulmonary disease. We performed a systematic review of diagnostic accuracy studies of blood RNA biomarkers or CRP measurements for EPTB, which could mitigate the need for site-of-disease sampling for the diagnosis of TB. We searched PubMed up to 1 st August 2023, using the following criteria: "extrapulmonary [title/abstract] AND tuberculosis [title/abstract] AND biomarker [title/abstract]". Although extrapulmonary TB was included in several studies, none focused specifically on EPTB or included an adequate number of EPTB cases to provide precise estimates of test accuracy. Added value of this study: To the best of our knowledge, we report the first diagnostic accuracy study of blood RNA biomarkers and CRP for TB among people with EPTB syndromes. We examined the performance of seven previously identified blood RNA biomarkers as triage tests for TB lymphadenitis and TB pericarditis compared to a microbiology reference standard among people referred to hospital for invasive sampling in a high TB and HIV prevalence setting. Multiple blood RNA biomarkers showed comparable diagnostic accuracy to that previously reported for pulmonary TB in both EPTB disease cohorts, irrespective of HIV status. All seven blood RNA biomarkers showed superior diagnostic accuracy to CRP for both lymphadenitis and pericarditis, but failed to meet the combined >90% sensitivity and >70% specificity recommended for a blood-based diagnostic triage test by WHO. Nonetheless, in decision curve analysis, an approach of using the best performing blood RNA biomarker to trigger confirmatory microbiological testing showed superior clinical utility in clinical services seeking to reduce the number needed to test (using invasive confirmatory testing) to less than 4 for each EPTB case detected. If acceptable to undertake invasive testing in more than 4 people for each true case detected, then a test-all approach will provide greater net benefit in this TB/HIV hyperendemic setting.Implications of all the available evidence: Blood RNA biomarkers show some potential as diagnostic triage tests for TB lymphadenitis and TB pericarditis, but do not provide the level of accuracy for blood-based triage tests recommended by WHO for community-based tests. CRP has inferior diagnostic accuracy to blood RNA biomarkers and cannot be recommended for diagnostic triage among people with EPTB syndromes referred for invasive sampling.
Collapse
Affiliation(s)
- Tiffeney Mann
- Division of Infection and Immunity, University College London, London, UK
| | - Stephanie Minnies
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research; South African Medical Research Council Centre for Tuberculosis Research; Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town
| | - Rishi K Gupta
- UCL Respiratory, Division of Medicine, University College London, London, UK
| | - Byron WP Reeve
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research; South African Medical Research Council Centre for Tuberculosis Research; Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town
| | - Georgina Nyawo
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research; South African Medical Research Council Centre for Tuberculosis Research; Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town
| | - Zaida Palmer
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research; South African Medical Research Council Centre for Tuberculosis Research; Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town
| | - Charissa Naidoo
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research; South African Medical Research Council Centre for Tuberculosis Research; Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town
| | - Anton Doubell
- Department of Medicine, Division of Cardiology, Stellenbosch University & Tygerberg Academic Hospital, South Africa
| | - Alfonso Pecararo
- Department of Medicine, Division of Cardiology, Stellenbosch University & Tygerberg Academic Hospital, South Africa
| | - Thadathilankal-Jess John
- Department of Medicine, Division of Cardiology, Stellenbosch University & Tygerberg Academic Hospital, South Africa
| | - Pawel Schubert
- National Health Laboratory Service, Tygerberg Hospital, Cape Town, Western Cape, South Africa
- Division Anatomical Pathology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, Western Cape, South Africa
| | - Claire J Calderwood
- Department of Clinical Research, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, UK
| | - Aneesh Chandran
- Division of Infection and Immunity, University College London, London, UK
| | - Grant Theron
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research; South African Medical Research Council Centre for Tuberculosis Research; Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town
| | - Mahdad Noursadeghi
- Division of Infection and Immunity, University College London, London, UK
| |
Collapse
|
28
|
Anh NK, Phat NK, Thu NQ, Tien NTN, Eunsu C, Kim HS, Nguyen DN, Kim DH, Long NP, Oh JY. Discovery of urinary biosignatures for tuberculosis and nontuberculous mycobacteria classification using metabolomics and machine learning. Sci Rep 2024; 14:15312. [PMID: 38961191 PMCID: PMC11222504 DOI: 10.1038/s41598-024-66113-x] [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: 04/16/2024] [Accepted: 06/27/2024] [Indexed: 07/05/2024] Open
Abstract
Nontuberculous mycobacteria (NTM) infection diagnosis remains a challenge due to its overlapping clinical symptoms with tuberculosis (TB), leading to inappropriate treatment. Herein, we employed noninvasive metabolic phenotyping coupled with comprehensive statistical modeling to discover potential biomarkers for the differential diagnosis of NTM infection versus TB. Urine samples from 19 NTM and 35 TB patients were collected, and untargeted metabolomics was performed using rapid liquid chromatography-mass spectrometry. The urine metabolome was analyzed using a combination of univariate and multivariate statistical approaches, incorporating machine learning. Univariate analysis revealed significant alterations in amino acids, especially tryptophan metabolism, in NTM infection compared to TB. Specifically, NTM infection was associated with upregulated levels of methionine but downregulated levels of glutarate, valine, 3-hydroxyanthranilate, and tryptophan. Five machine learning models were used to classify NTM and TB. Notably, the random forest model demonstrated excellent performance [area under the receiver operating characteristic (ROC) curve greater than 0.8] in distinguishing NTM from TB. Six potential biomarkers for NTM infection diagnosis, including methionine, valine, glutarate, 3-hydroxyanthranilate, corticosterone, and indole-3-carboxyaldehyde, were revealed from univariate ROC analysis and machine learning models. Altogether, our study suggested new noninvasive biomarkers and laid a foundation for applying machine learning to NTM differential diagnosis.
Collapse
Affiliation(s)
- Nguyen Ky Anh
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan, 47392, Republic of Korea
- Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City, Vietnam
| | - Nguyen Ky Phat
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan, 47392, Republic of Korea
| | - Nguyen Quang Thu
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan, 47392, Republic of Korea
| | - Nguyen Tran Nam Tien
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan, 47392, Republic of Korea
| | - Cho Eunsu
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan, 47392, Republic of Korea
| | - Ho-Sook Kim
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan, 47392, Republic of Korea
| | - Duc Ninh Nguyen
- Section for Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of Copenhagen, 1870, Frederiksberg, Denmark
| | - Dong Hyun Kim
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan, 47392, Republic of Korea
| | - Nguyen Phuoc Long
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan, 47392, Republic of Korea.
| | - Jee Youn Oh
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, Korea University Guro Hospital, Seoul, 08308, Republic of Korea.
| |
Collapse
|
29
|
Kreitmann L, D'Souza G, Miglietta L, Vito O, Jackson HR, Habgood-Coote D, Levin M, Holmes A, Kaforou M, Rodriguez-Manzano J. A computational framework to improve cross-platform implementation of transcriptomics signatures. EBioMedicine 2024; 105:105204. [PMID: 38901146 PMCID: PMC11245942 DOI: 10.1016/j.ebiom.2024.105204] [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: 01/10/2024] [Revised: 05/29/2024] [Accepted: 06/02/2024] [Indexed: 06/22/2024] Open
Abstract
The emergence of next-generation sequencing technologies and computational advances have expanded our understanding of gene expression regulation (i.e., the transcriptome). This has also led to an increased interest in using transcriptomic biomarkers to improve disease diagnosis and stratification, to assess prognosis and predict the response to treatment. Significant progress in identifying transcriptomic signatures for various clinical needs has been made, with large discovery studies accounting for challenges such as patient variability, unwanted batch effects, and data complexities; however, obstacles related to the technical aspects of cross-platform implementation still hinder the successful integration of transcriptomic technologies into standard diagnostic workflows. In this article, we discuss the challenges associated with integrating transcriptomic signatures derived using high-throughput technologies (such as RNA-sequencing) into clinical diagnostic tools using nucleic acid amplification (NAA) techniques. The novelty of the proposed approach lies in our aim to embed constraints related to cross-platform implementation in the process of signature discovery. These constraints could include technical limitations of amplification platform and chemistry, the maximal number of targets imposed by the chosen multiplexing strategy, and the genomic context of identified RNA biomarkers. Finally, we propose to build a computational framework that would integrate these constraints in combination with existing statistical and machine learning models used for signature identification. We envision that this could accelerate the integration of RNA signatures discovered by high-throughput technologies into NAA-based approaches suitable for clinical applications.
Collapse
Affiliation(s)
- Louis Kreitmann
- Section of Adult Infectious Disease, Faculty of Medicine, Imperial College London, London, W12 0NN, United Kingdom; Centre for Antimicrobial Optimisation, Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, W12 0NN, United Kingdom
| | - Giselle D'Souza
- Section of Adult Infectious Disease, Faculty of Medicine, Imperial College London, London, W12 0NN, United Kingdom; Centre for Antimicrobial Optimisation, Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, W12 0NN, United Kingdom; Section of Paediatric Infectious Disease, Faculty of Medicine, Imperial College London, London, W2 1NY, United Kingdom; Centre for Paediatrics and Child Health, Imperial College London, London, W2 1NY, United Kingdom
| | - Luca Miglietta
- Section of Adult Infectious Disease, Faculty of Medicine, Imperial College London, London, W12 0NN, United Kingdom; Centre for Antimicrobial Optimisation, Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, W12 0NN, United Kingdom
| | - Ortensia Vito
- Section of Paediatric Infectious Disease, Faculty of Medicine, Imperial College London, London, W2 1NY, United Kingdom; Centre for Paediatrics and Child Health, Imperial College London, London, W2 1NY, United Kingdom
| | - Heather R Jackson
- Section of Paediatric Infectious Disease, Faculty of Medicine, Imperial College London, London, W2 1NY, United Kingdom; Centre for Paediatrics and Child Health, Imperial College London, London, W2 1NY, United Kingdom
| | - Dominic Habgood-Coote
- Section of Paediatric Infectious Disease, Faculty of Medicine, Imperial College London, London, W2 1NY, United Kingdom; Centre for Paediatrics and Child Health, Imperial College London, London, W2 1NY, United Kingdom
| | - Michael Levin
- Section of Paediatric Infectious Disease, Faculty of Medicine, Imperial College London, London, W2 1NY, United Kingdom; Centre for Paediatrics and Child Health, Imperial College London, London, W2 1NY, United Kingdom
| | - Alison Holmes
- Section of Adult Infectious Disease, Faculty of Medicine, Imperial College London, London, W12 0NN, United Kingdom; Centre for Antimicrobial Optimisation, Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, W12 0NN, United Kingdom
| | - Myrsini Kaforou
- Section of Paediatric Infectious Disease, Faculty of Medicine, Imperial College London, London, W2 1NY, United Kingdom; Centre for Paediatrics and Child Health, Imperial College London, London, W2 1NY, United Kingdom
| | - Jesus Rodriguez-Manzano
- Section of Adult Infectious Disease, Faculty of Medicine, Imperial College London, London, W12 0NN, United Kingdom; Centre for Antimicrobial Optimisation, Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, W12 0NN, United Kingdom.
| |
Collapse
|
30
|
Zhang P, Zheng J, Han T, Ma J, Gnanashanmugam D, Li M, Tang YW, Deng G. A blood-based 3-gene signature score for therapeutic monitoring in patients with pulmonary tuberculosis. Tuberculosis (Edinb) 2024; 147:102521. [PMID: 38801793 DOI: 10.1016/j.tube.2024.102521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 05/12/2024] [Accepted: 05/20/2024] [Indexed: 05/29/2024]
Abstract
OBJECTIVE To assess the validity of Xpert Tuberculosis Fingerstick score for monitoring treatment response and analyze factors influencing its performance. METHODS 122 adults with pulmonary tuberculosis were recruited and stratified into three cohorts: Diabetic-drug-susceptible-TB (DM-TB), Non-diabetic-drug-susceptible-TB (NDM-TB) and Non-diabetic Multidrug-resistant TB (MDR-TB). Fingerstick blood specimens were tested at treatment initiation (M0) and the end of the first (M1), second (M2), and sixth month (M6) to generate a TB-score. RESULTS The TB-score in all participants yielded an AUC of 0.707 (95% CI: 0.579-0.834) at M2 when its performance was evaluated against sputum culture conversion. In all non-diabetes patients, the AUC reached 0.88 (95% CI: 0.756-1.000) with an optimal cut-off value of 1.95 at which sensitivity was 90.0% (95% CI: 59.6-98.2%) and specificity was 81.3% (95% CI: 70.0-88.9%). The mean TB score was higher in patients with low bacterial loads (n = 31) than those with high bacterial loads (n = 91) at M0, M1, M2, and M6, and was higher in non-cavitary patients (n = 71) than those with cavitary lesions (n = 51) at M0, M1, and M2. CONCLUSION Xpert TB-score shows promising predictive value for culture conversion in non-diabetic TB patients. Sputum bacterial load and lung cavitation status have an influence on the value of TB score.
Collapse
Affiliation(s)
- Peize Zhang
- Department of Pulmonary Medicine and Tuberculosis, The Third People's Hospital of Shenzhen, China.
| | - Junfeng Zheng
- Department of Pulmonary Medicine and Tuberculosis, The Third People's Hospital of Shenzhen, China.
| | - Tingting Han
- Guangdong Medical University, The First Clinical Medical College, Zhanjiang, Guangdong, China.
| | - Jian Ma
- Medical Affairs, Danaher Corporation/Cepheid (China), Shanghai, China.
| | | | - Mengran Li
- Department of Biostatistics & Data Management, Beckman Coulter, Shanghai, China.
| | - Yi-Wei Tang
- Medical Affairs, Danaher Corporation/Cepheid (China), Shanghai, China.
| | - Guofang Deng
- Department of Pulmonary Medicine and Tuberculosis, The Third People's Hospital of Shenzhen, China.
| |
Collapse
|
31
|
Arya R, Shakya H, Chaurasia R, Kumar S, Vinetz JM, Kim JJ. Computational reassessment of RNA-seq data reveals key genes in active tuberculosis. PLoS One 2024; 19:e0305582. [PMID: 38935691 PMCID: PMC11210783 DOI: 10.1371/journal.pone.0305582] [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: 02/27/2024] [Accepted: 05/31/2024] [Indexed: 06/29/2024] Open
Abstract
BACKGROUND Tuberculosis is a serious life-threatening disease among the top global health challenges and rapid and effective diagnostic biomarkers are vital for early diagnosis especially given the increasing prevalence of multidrug resistance. METHODS Two human whole blood microarray datasets, GSE42826 and GSE42830 were retrieved from publicly available gene expression omnibus (GEO) database. Deregulated genes (DEGs) were identified using GEO2R online tool and Gene Ontology (GO), protein-protein interaction (PPI) network analysis was performed using Metascape and STRING databases. Significant genes (n = 8) were identified using T-test/ANOVA and Molecular Complex Detection (MCODE) score ≥10, which was validated in GSE34608 dataset. The diagnostic potential of three biomarkers was assessed using Area Under Curve (AUC) of Receiver Operating Characteristic (ROC) plot. The transcriptional levels of these genes were also examined in a separate dataset GSE31348, to monitor the patterns of variation during tuberculosis treatment. RESULTS A total of 62 common DEGs (57 upregulated, 7 downregulated genes) were identified in two discovery datasets. GO functions and pathway enrichment analysis shed light on the functional roles of these DEGs in immune response and type-II interferon signaling. The genes in Module-1 (n = 18) were linked to innate immune response, interferon-gamma signaling. The common genes (n = 8) were validated in GSE34608 dataset, that corroborates the results obtained from discovery sets. The gene expression levels demonstrated responsiveness to Mtb infection during anti-TB therapy in GSE31348 dataset. In GSE34608 dataset, the expression levels of three specific genes, GBP5, IFITM3, and EPSTI1, emerged as potential diagnostic makers. In combination, these genes scored remarkable diagnostic performance with 100% sensitivity and 89% specificity, resulting in an impressive Area Under Curve (AUC) of 0.958. However, GBP5 alone showed the highest AUC of 0.986 with 100% sensitivity and 89% specificity. CONCLUSIONS The study presents valuable insights into the critical gene network perturbed during tuberculosis. These genes are determinants for assessing the effectiveness of an anti-TB response and distinguishing between active TB and healthy individuals. GBP5, IFITM3 and EPSTI1 emerged as candidate core genes in TB and holds potential as novel molecular targets for the development of interventions in the treatment of TB.
Collapse
Affiliation(s)
- Rakesh Arya
- Department of Biotechnology, Yeungnam University, Gyeongsan, Gyeongbuk, South Korea
| | - Hemlata Shakya
- Department of Biomedical Engineering, Shri G. S. Institute of Technology and Science, Indore, Madhya Pradesh, India
| | - Reetika Chaurasia
- Department of Internal Medicine, Section of Infectious Diseases, Yale University School of Medicine, New Haven, CT, United States of America
| | - Surendra Kumar
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Joseph M. Vinetz
- Department of Internal Medicine, Section of Infectious Diseases, Yale University School of Medicine, New Haven, CT, United States of America
| | - Jong Joo Kim
- Department of Biotechnology, Yeungnam University, Gyeongsan, Gyeongbuk, South Korea
| |
Collapse
|
32
|
Painter H, Larsen SE, Williams BD, Abdelaal HFM, Baldwin SL, Fletcher HA, Fiore-Gartland A, Coler RN. Backtranslation of human RNA biosignatures of tuberculosis disease risk into the preclinical pipeline is condition dependent. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.21.600067. [PMID: 38948876 PMCID: PMC11212953 DOI: 10.1101/2024.06.21.600067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
It is not clear whether human progression to active tuberculosis disease (TB) risk signatures are viable endpoint criteria for evaluations of treatments in clinical or preclinical development. TB is the deadliest infectious disease globally and more efficacious vaccines are needed to reduce this mortality. However, the immune correlates of protection for either preventing infection with Mycobacterium tuberculosis or preventing TB disease have yet to be completely defined, making the advancement of candidate vaccines through the pipeline slow, costly, and fraught with risk. Human-derived correlate of risk (COR) gene signatures, which identify an individual's risk to progressing to active TB disease, provide an opportunity for evaluating new therapies for TB with clear and defined endpoints. Though prospective clinical trials with longitudinal sampling are prohibitively expensive, characterization of COR gene signatures is practical with preclinical models. Using a 3Rs (Replacement, Reduction and Refinement) approach we reanalyzed heterogeneous publicly available transcriptional datasets to determine whether a specific set of COR signatures are viable endpoints in the preclinical pipeline. We selected RISK6, Sweeney3 and BATF2 human-derived blood-based RNA biosignatures because they require relatively few genes to assign a score and have been carefully evaluated across several clinical cohorts. Excitingly, these data provide proof-of-concept that human COR signatures seem to have high fidelity across several tissue types in the preclinical TB model pipeline and show best performance when the model most closely reflected human infection or disease conditions. Human-derived COR signatures offer an opportunity for high-throughput preclinical endpoint criteria of vaccine and drug therapy evaluations. One Sentence Summary Human-derived biosignatures of tuberculosis disease progression were evaluated for their predictive fidelity across preclinical species and derived tissues using available public data sets.
Collapse
|
33
|
Wang X, VanValkenberg A, Odom AR, Ellner JJ, Hochberg NS, Salgame P, Patil P, Johnson WE. Comparison of gene set scoring methods for reproducible evaluation of tuberculosis gene signatures. BMC Infect Dis 2024; 24:610. [PMID: 38902649 PMCID: PMC11191245 DOI: 10.1186/s12879-024-09457-z] [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: 07/29/2023] [Accepted: 05/31/2024] [Indexed: 06/22/2024] Open
Abstract
BACKGROUND Blood-based transcriptional gene signatures for tuberculosis (TB) have been developed with potential use to diagnose disease. However, an unresolved issue is whether gene set enrichment analysis of the signature transcripts alone is sufficient for prediction and differentiation or whether it is necessary to use the original model created when the signature was derived. Intra-method comparison is complicated by the unavailability of original training data and missing details about the original trained model. To facilitate the utilization of these signatures in TB research, comparisons between gene set scoring methods cross-data validation of original model implementations are needed. METHODS We compared the performance of 19 TB gene signatures across 24 transcriptomic datasets using both rrebuilt original models and gene set scoring methods. Existing gene set scoring methods, including ssGSEA, GSVA, PLAGE, Singscore, and Zscore, were used as alternative approaches to obtain the profile scores. The area under the ROC curve (AUC) value was computed to measure performance. Correlation analysis and Wilcoxon paired tests were used to compare the performance of enrichment methods with the original models. RESULTS For many signatures, the predictions from gene set scoring methods were highly correlated and statistically equivalent to the results given by the original models. In some cases, PLAGE outperformed the original models when considering signatures' weighted mean AUC values and the AUC results within individual studies. CONCLUSION Gene set enrichment scoring of existing gene sets can distinguish patients with active TB disease from other clinical conditions with equivalent or improved accuracy compared to the original methods and models. These data justify using gene set scoring methods of published TB gene signatures for predicting TB risk and treatment outcomes, especially when original models are difficult to apply or implement.
Collapse
Affiliation(s)
- Xutao Wang
- Department of Biostatistics, Boston University, Boston, MA, USA
- Division of Computational Biomedicine and Bioinformatics Program, Boston University, Boston, MA, USA
| | - Arthur VanValkenberg
- Division of Infectious Disease, Center for Data Science, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Aubrey R Odom
- Division of Computational Biomedicine and Bioinformatics Program, Boston University, Boston, MA, USA
| | - Jerrold J Ellner
- Department of Medicine, Center for Emerging Pathogens, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Natasha S Hochberg
- Boston Medical Center, Boston, MA, USA
- Section of Infectious Diseases, Boston University School of Medicine, Boston, MA, USA
| | - Padmini Salgame
- Department of Medicine, Center for Emerging Pathogens, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Prasad Patil
- Department of Biostatistics, Boston University, Boston, MA, USA
| | - W Evan Johnson
- Division of Infectious Disease, Center for Data Science, Rutgers New Jersey Medical School, Newark, NJ, USA.
- Department of Medicine, Center for Emerging Pathogens, Rutgers New Jersey Medical School, Newark, NJ, USA.
| |
Collapse
|
34
|
Sun Z, He B, Yang Z, Huang Y, Duan Z, Yu C, Dan Z, Paek C, Chen P, Zhou J, Lei J, Wang F, Liu B, Yin L. Cost-Effective Whole Transcriptome Sequencing Landscape and Diagnostic Potential Biomarkers in Active Tuberculosis. ACS Infect Dis 2024; 10:2318-2332. [PMID: 38832694 DOI: 10.1021/acsinfecdis.4c00374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2024]
Abstract
Tuberculosis (TB) is a prevalent and severe infectious disease that poses a significant threat to human health. However, it is frequently disregarded as there are not enough quick and accurate ways to diagnose tuberculosis. Here, we develop a strategy for tuberculosis detection to address the challenges, including an experimental strategy, namely, Double Adapter Directional Capture sequencing (DADCSeq), an easily operated and low-cost whole transcriptome sequencing method, and a computational method to identify hub differentially expressed genes as well as the diagnosis of TB based on whole transcriptome data using DADCSeq on peripheral blood mononuclear cells (PBMCs) from active TB and latent TB or healthy control. Applying our approach to create a robust and stable TB multi-mRNA risk probability model (TBMMRP) that can accurately distinguish active and latent TB patients, including active TB and healthy controls in clinical cohorts, this diagnostic biomarker was successfully validated by several independent cross-platform cohorts with favorable performance in differentiating active TB from latent TB or active TB from healthy controls and further demonstrated superior or similar diagnostic accuracy compared to previous diagnostic markers. Overall, we develop a low-cost and effective strategy for tuberculosis diagnosis; as the clinical cohort increases, we can expand to different disease kinds and learn new features through our disease diagnosis strategy.
Collapse
Affiliation(s)
- Zaiqiao Sun
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Department of Clinical Oncology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, Hubei Province 430072, China
| | - Boxiao He
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Department of Clinical Oncology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, Hubei Province 430072, China
| | - Zhifeng Yang
- Department of Chest Surgery, Wuhan Jinyintan Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei Province 430040, China
| | - Yi Huang
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei Province 430030, China
| | - Zhaoyu Duan
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Department of Clinical Oncology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, Hubei Province 430072, China
| | - Chengyi Yu
- Department of Active and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province 430071, China
| | - Zhaokui Dan
- Clinical Medicine School of Hubei University of Science and Technology, Xianning, Hubei Province 437100, China
| | - Chonil Paek
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Department of Clinical Oncology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, Hubei Province 430072, China
| | - Peng Chen
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Department of Clinical Oncology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, Hubei Province 430072, China
| | - Jin Zhou
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Department of Clinical Oncology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, Hubei Province 430072, China
| | - Jun Lei
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Department of Clinical Oncology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, Hubei Province 430072, China
| | - Feng Wang
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei Province 430030, China
| | - Bing Liu
- Department of Active and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province 430071, China
- Wuhan Research Center for Infectious Diseases and Cancer, Chinese Academy of Medical Sciences, Wuhan, Hubei Province 100730, China
| | - Lei Yin
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Department of Clinical Oncology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, Hubei Province 430072, China
| |
Collapse
|
35
|
Chang A, Loy CJ, Eweis-LaBolle D, Lenz JS, Steadman A, Andgrama A, Nhung NV, Yu C, Worodria W, Denkinger CM, Nahid P, Cattamanchi A, De Vlaminck I. Circulating cell-free RNA in blood as a host response biomarker for detection of tuberculosis. Nat Commun 2024; 15:4949. [PMID: 38858368 PMCID: PMC11164910 DOI: 10.1038/s41467-024-49245-6] [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: 03/08/2023] [Accepted: 05/29/2024] [Indexed: 06/12/2024] Open
Abstract
Tuberculosis (TB) remains a leading cause of death from an infectious disease worldwide, partly due to a lack of effective strategies to screen and triage individuals with potential TB. Whole blood RNA signatures have been tested as biomarkers for TB, but have failed to meet the World Health Organization's (WHO) optimal target product profiles (TPP). Here, we use RNA sequencing and machine-learning to investigate the utility of plasma cell-free RNA (cfRNA) as a host-response biomarker for TB in cohorts from Uganda, Vietnam and Philippines. We report a 6-gene cfRNA signature, which differentiates TB-positive and TB-negative individuals with AUC = 0.95, 0.92, and 0.95 in test, training and validation, respectively. This signature meets WHO TPPs (sensitivity: 97.1% [95% CI: 80.9-100%], specificity: 85.2% [95% CI: 72.4-100%]) regardless of geographic location, sample collection method and HIV status. Overall, our results identify plasma cfRNA as a promising host response biomarker to diagnose TB.
Collapse
Affiliation(s)
- Adrienne Chang
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Conor J Loy
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | | | - Joan S Lenz
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | | | - Alfred Andgrama
- World Alliance for Lung and Intensive Care Medicine in Uganda, Kampala, Uganda
| | | | - Charles Yu
- De La Salle Medical and Health Sciences Institute, Dasmarinas, Philippines
| | - William Worodria
- World Alliance for Lung and Intensive Care Medicine in Uganda, Kampala, Uganda
| | - Claudia M Denkinger
- University Hospital Heidelberg & German Center of Infection Research, Heidelberg, Germany
| | - Payam Nahid
- UCSF Center for Tuberculosis, University of California San Francisco, San Francisco, CA, USA
| | - Adithya Cattamanchi
- UCSF Center for Tuberculosis, University of California San Francisco, San Francisco, CA, USA
- Division of Pulmonary and Critical Care Medicine, University of California Irvine, Orange, CA, USA
| | - Iwijn De Vlaminck
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA.
| |
Collapse
|
36
|
Sun J, Han X, Yan H, Zhang X, Jiang T, Zhang T, Wu H, Kaminskiy G, Ma Y, Karamov E, Su B. Advances in technology for the laboratory diagnosis of individuals with HIV/AIDS coinfected with Mycobacterium tuberculosis. BIOSAFETY AND HEALTH 2024; 6:133-142. [PMID: 40078723 PMCID: PMC11895006 DOI: 10.1016/j.bsheal.2024.04.003] [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: 12/04/2023] [Revised: 04/22/2024] [Accepted: 04/25/2024] [Indexed: 03/14/2025] Open
Abstract
The high morbidity and mortality rate of individuals with human immunodeficiency virus (HIV) / acquired immunodeficiency syndrome (AIDS) coinfected with Mycobacterium tuberculosis (MTB) is a tough challenge for current global tuberculosis prevention and control efforts. HIV/MTB coinfection is more complex than a single infection, and the interaction between the two diseases aggravates the deterioration caused by the disease, resulting in increased hospitalizations and deaths. Rapid screening and early diagnosis facilitate the timely initiation of anti-tuberculosis treatment in HIV/MTB coinfected individuals, thereby reducing transmission and the incidence of adverse prognoses. To date, pathogenic detection has remained the gold standard for diagnosing tuberculosis, but its sensitivity and specificity are greatly affected by the body's immune status, which limits its application in the diagnosis of HIV/MTB coinfection. Recently, immunology and molecular detection technology has developed rapidly. New detection technologies, such as interferon-γ release assays, interferon-gamma inducible protein 10, and GeneXpert MTB/RIF assay have overcome the limitations of traditional detection methods, significantly improved the sensitivity and specificity of tuberculosis diagnosis, and brought new hope to the detection of HIV/MTB coinfection. In this article, the principle, scope of application, and latest research progress of relevant detection methods are reviewed to provide a reference for the early diagnosis of HIV/MTB coinfection.
Collapse
Affiliation(s)
- Jin Sun
- Beijing Key Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| | - Xiaoxu Han
- Beijing Key Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| | - Hongxia Yan
- Beijing Key Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| | - Xin Zhang
- Beijing Key Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| | - Taiyi Jiang
- Beijing Key Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| | - Tong Zhang
- Beijing Key Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| | - Hao Wu
- Beijing Key Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| | - Grigory Kaminskiy
- State Health Organization Tula Regional Center for Control and Prevention of AIDS and Infectious Diseases, Tula 300002, Russia
| | - Yingmin Ma
- Department of Respiratory and Critical Care Medicine, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| | - Eduard Karamov
- Gamaleya National Research Centre for Epidemiology and Microbiology of the Ministry of Health of the Russian Federation, Moscow 123098, Russia
| | - Bin Su
- Beijing Key Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| |
Collapse
|
37
|
Davies LRL, Wang C, Steigler P, Bowman KA, Fischinger S, Hatherill M, Fisher M, Mbandi SK, Rodo M, Ottenhoff THM, Dockrell HM, Sutherland JS, Mayanja-Kizza H, Boom WH, Walzl G, Kaufmann SHE, Nemes E, Scriba TJ, Lauffenburger D, Alter G, Fortune SM. Age and sex influence antibody profiles associated with tuberculosis progression. Nat Microbiol 2024; 9:1513-1525. [PMID: 38658786 PMCID: PMC11153143 DOI: 10.1038/s41564-024-01678-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 03/20/2024] [Indexed: 04/26/2024]
Abstract
Antibody features vary with tuberculosis (TB) disease state. Whether clinical variables, such as age or sex, influence associations between Mycobacterium tuberculosis-specific antibody responses and disease state is not well explored. Here we profiled Mycobacterium tuberculosis-specific antibody responses in 140 TB-exposed South African individuals from the Adolescent Cohort Study. We identified distinct response features in individuals progressing to active TB from non-progressing, matched controls. A multivariate antibody score differentially associated with progression (SeroScore) identified progressors up to 2 years before TB diagnosis, earlier than that achieved with the RISK6 transcriptional signature of progression. We validated these antibody response features in the Grand Challenges 6-74 cohort. Both the SeroScore and RISK6 correlated better with risk of TB progression in adolescents compared with adults, and in males compared with females. This suggests that age and sex are important, underappreciated modifiers of antibody responses associated with TB progression.
Collapse
Affiliation(s)
- Leela R L Davies
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- Brigham and Women's Hospital, Boston, MA, USA
| | - Chuangqi Wang
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Pia Steigler
- South African Tuberculosis Vaccine Initiative and Institute of Infectious Disease and Molecular Medicine, Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
- Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease and Molecular Medicine and Division of Immunology, Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Kathryn A Bowman
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- Brigham and Women's Hospital, Boston, MA, USA
| | | | - Mark Hatherill
- South African Tuberculosis Vaccine Initiative and Institute of Infectious Disease and Molecular Medicine, Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Michelle Fisher
- South African Tuberculosis Vaccine Initiative and Institute of Infectious Disease and Molecular Medicine, Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Stanley Kimbung Mbandi
- South African Tuberculosis Vaccine Initiative and Institute of Infectious Disease and Molecular Medicine, Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Miguel Rodo
- South African Tuberculosis Vaccine Initiative and Institute of Infectious Disease and Molecular Medicine, Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Tom H M Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, the Netherlands
| | - Hazel M Dockrell
- Vaccines and Immunity Theme, Medical Research Council Unit The Gambia at the London School of Hygiene and Tropical Medicine, Banjul, The Gambia
| | - Jayne S Sutherland
- Vaccines and Immunity Theme, Medical Research Council Unit The Gambia at the London School of Hygiene and Tropical Medicine, Banjul, The Gambia
| | - Harriet Mayanja-Kizza
- Department of Medicine and Department of Microbiology, Makerere University, Kampala, Uganda
| | - W Henry Boom
- Tuberculosis Research Unit, Case Western Reserve University, Cleveland, OH, USA
| | - Gerhard Walzl
- Department of Science and Technology National Research Foundation Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Stefan H E Kaufmann
- Max Planck Institute for Infection Biology, Berlin, Germany
- Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Hagler Institute for Advanced Study, Texas A&M University, College Station, TX, USA
| | - Elisa Nemes
- South African Tuberculosis Vaccine Initiative and Institute of Infectious Disease and Molecular Medicine, Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Thomas J Scriba
- South African Tuberculosis Vaccine Initiative and Institute of Infectious Disease and Molecular Medicine, Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | | | - Galit Alter
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA.
- Moderna Therapeutics, Cambridge, MA, USA.
| | - Sarah M Fortune
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| |
Collapse
|
38
|
Zhang L, Liu Q, Guo Y, Tian L, Chen K, Bai D, Yu H, Han X, Luo W, Feng T, Deng S, Xie G. DNA-based molecular classifiers for the profiling of gene expression signatures. J Nanobiotechnology 2024; 22:189. [PMID: 38632615 PMCID: PMC11025223 DOI: 10.1186/s12951-024-02445-0] [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: 09/28/2023] [Accepted: 03/28/2024] [Indexed: 04/19/2024] Open
Abstract
Although gene expression signatures offer tremendous potential in diseases diagnostic and prognostic, but massive gene expression signatures caused challenges for experimental detection and computational analysis in clinical setting. Here, we introduce a universal DNA-based molecular classifier for profiling gene expression signatures and generating immediate diagnostic outcomes. The molecular classifier begins with feature transformation, a modular and programmable strategy was used to capture relative relationships of low-concentration RNAs and convert them to general coding inputs. Then, competitive inhibition of the DNA catalytic reaction enables strict weight assignment for different inputs according to their importance, followed by summation, annihilation and reporting to accurately implement the mathematical model of the classifier. We validated the entire workflow by utilizing miRNA expression levels for the diagnosis of hepatocellular carcinoma (HCC) in clinical samples with an accuracy 85.7%. The results demonstrate the molecular classifier provides a universal solution to explore the correlation between gene expression patterns and disease diagnostics, monitoring, and prognosis, and supports personalized healthcare in primary care.
Collapse
Affiliation(s)
- Li Zhang
- Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
- Department of Forensic Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Qian Liu
- Nuclear Medicine Department, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Yongcan Guo
- Clinical Laboratory, Traditional Chinese Medicine Hospital Affiliated to Southwest Medical University, Luzhou, 646000, China
| | - Luyao Tian
- Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Kena Chen
- Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Dan Bai
- Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Hongyan Yu
- Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Xiaole Han
- Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Wang Luo
- Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Tong Feng
- Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Shixiong Deng
- Department of Forensic Medicine, Chongqing Medical University, Chongqing, 400016, China.
| | - Guoming Xie
- Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
39
|
Washington P. A Perspective on Crowdsourcing and Human-in-the-Loop Workflows in Precision Health. J Med Internet Res 2024; 26:e51138. [PMID: 38602750 PMCID: PMC11046386 DOI: 10.2196/51138] [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: 07/22/2023] [Revised: 11/15/2023] [Accepted: 01/30/2024] [Indexed: 04/12/2024] Open
Abstract
Modern machine learning approaches have led to performant diagnostic models for a variety of health conditions. Several machine learning approaches, such as decision trees and deep neural networks, can, in principle, approximate any function. However, this power can be considered to be both a gift and a curse, as the propensity toward overfitting is magnified when the input data are heterogeneous and high dimensional and the output class is highly nonlinear. This issue can especially plague diagnostic systems that predict behavioral and psychiatric conditions that are diagnosed with subjective criteria. An emerging solution to this issue is crowdsourcing, where crowd workers are paid to annotate complex behavioral features in return for monetary compensation or a gamified experience. These labels can then be used to derive a diagnosis, either directly or by using the labels as inputs to a diagnostic machine learning model. This viewpoint describes existing work in this emerging field and discusses ongoing challenges and opportunities with crowd-powered diagnostic systems, a nascent field of study. With the correct considerations, the addition of crowdsourcing to human-in-the-loop machine learning workflows for the prediction of complex and nuanced health conditions can accelerate screening, diagnostics, and ultimately access to care.
Collapse
Affiliation(s)
- Peter Washington
- Information and Computer Sciences, University of Hawaii at Manoa, Honolulu, HI, United States
| |
Collapse
|
40
|
Arya R, Shakya H, Chaurasia R, Haque MA, Kim JJ. Exploring the Role of Extracellular Vesicles in the Pathogenesis of Tuberculosis. Genes (Basel) 2024; 15:434. [PMID: 38674369 PMCID: PMC11049626 DOI: 10.3390/genes15040434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 03/27/2024] [Accepted: 03/28/2024] [Indexed: 04/28/2024] Open
Abstract
Tuberculosis (TB) remains a significant global health concern, necessitating accurate diagnosis and treatment monitoring. Extracellular vesicles (EVs), including exosomes, play crucial roles in disease progression, with their associated genes serving as potential biomarkers and therapeutic targets. Leveraging publicly available RNA-Seq datasets of TB patients and healthy controls (HCs), to identify differentially expressed genes (DEGs) and their associated protein-protein interaction networks and immune cell profiles, the common EV-related DEGs were identified and validated in the GSE42830 and GSE40553 datasets. We have identified nine common EV-related DEGs (SERPINA1, TNFAIP6, MAPK14, STAT1, ITGA2B, VAMP5, CTSL, CEACAM1, and PLAUR) upregulated in TB patients. Immune cell infiltration analysis revealed significant differences between TB patients and HCs, highlighting increased proportions of various immune cells in TB patients. These DEGs are involved in crucial cellular processes and pathways related to exocytosis and immune response regulation. Notably, VAMP5 exhibited excellent diagnostic performance (AUC-0.993, sensitivity-93.8%, specificity-100%), with potential as a novel biomarker for TB. The EV-related genes can serve as novel potential biomarkers that can distinguish between TB and HCs. VAMP5, which functions in exosome biogenesis and showed significant upregulation in TB, can be targeted for therapeutic interventions and treatment outcomes.
Collapse
Affiliation(s)
- Rakesh Arya
- Department of Biotechnology, Yeungnam University, Gyeongsan 38541, Republic of Korea; (R.A.); (M.A.H.)
| | - Hemlata Shakya
- Department of Biomedical Engineering, Shri G. S. Institute of Technology and Science, Indore 452003, Madhya Pradesh, India;
| | - Reetika Chaurasia
- Department of Internal Medicine, Section of Infectious Diseases, Yale University School of Medicine, New Haven, CT 06519, USA
| | - Md Azizul Haque
- Department of Biotechnology, Yeungnam University, Gyeongsan 38541, Republic of Korea; (R.A.); (M.A.H.)
| | - Jong-Joo Kim
- Department of Biotechnology, Yeungnam University, Gyeongsan 38541, Republic of Korea; (R.A.); (M.A.H.)
| |
Collapse
|
41
|
Russomando G, Sanabria D, Díaz Acosta CC, Rojas L, Franco L, Arenas R, Delogu G, Ndiaye MDB, Bayaa R, Rakotosamimanana N, Goletti D, Hoffmann J. C1q and HBHA-specific IL-13 levels as surrogate plasma biomarkers for monitoring tuberculosis treatment efficacy: a cross-sectional cohort study in Paraguay. Front Immunol 2024; 15:1308015. [PMID: 38545118 PMCID: PMC10967656 DOI: 10.3389/fimmu.2024.1308015] [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: 10/05/2023] [Accepted: 02/28/2024] [Indexed: 04/04/2024] Open
Abstract
Introduction New diagnostic tools are needed to rapidly assess the efficacy of pulmonary tuberculosis (PTB) treatment. The aim of this study was to evaluate several immune biomarkers in an observational and cross-sectional cohort study conducted in Paraguay. Methods Thirty-two patients with clinically and microbiologically confirmed PTB were evaluated before starting treatment (T0), after 2 months of treatment (T1) and at the end of treatment (T2). At each timepoint plasma levels of IFN-y, 17 pro- and anti-inflammatory cytokines/chemokines and complement factors C1q, C3 and C4 were assessed in unstimulated and Mtb-specific stimulated whole blood samples using QuantiFERON-TB gold plus and recombinant Mycobacterium smegmatis heparin binding hemagglutinin (rmsHBHA) as stimulation antigen. Complete blood counts and liver enzyme assays were also evaluated and correlated with biomarker levels in plasma. Results In unstimulated plasma, C1q (P<0.001), C4 (P<0.001), hemoglobin (P<0.001), lymphocyte proportion (P<0.001) and absolute white blood cell count (P=0.01) were significantly higher in PTB patients at baseline than in cured patients. C1q and C4 levels were found to be related to Mycobacterium tuberculosis load in sputum. Finally, a combinatorial analysis identified a plasma host signature comprising the detection of C1q and IL-13 levels in response to rmsHBHA as a tool differentiating PTB patients from cured TB profiles, with an AUC of 0.92 (sensitivity 94% and specificity 79%). Conclusion This observational study provides new insights on host immune responses throughout anti-TB treatment and emphasizes the role of host C1q and HBHA-specific IL-13 response as surrogate plasma biomarkers for monitoring TB treatment efficacy.
Collapse
Affiliation(s)
- Graciela Russomando
- Instituto de Investigaciones en Ciencias de la Salud, National University of Asunción, Asunción, Paraguay
| | - Diana Sanabria
- Instituto de Investigaciones en Ciencias de la Salud, National University of Asunción, Asunción, Paraguay
| | | | - Leticia Rojas
- Instituto de Investigaciones en Ciencias de la Salud, National University of Asunción, Asunción, Paraguay
| | - Laura Franco
- Instituto de Investigaciones en Ciencias de la Salud, National University of Asunción, Asunción, Paraguay
| | - Rossana Arenas
- Hospital General de San Lorenzo, Ministerio de Salud Pública y Bienestar Social (MSPyBS), Asunción, Paraguay
| | - Giovanni Delogu
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie – Sezione di Microbiologia, Università Cattolica del Sacro Cuore, Rome, Italy
| | | | - Rim Bayaa
- Medical and Scientific Department, Fondation Mérieux, Lyon, France
| | | | - Delia Goletti
- Translational Research Unit, Department of Epidemiology and Preclinical Research, “L. Spallanzani” National Institute for Infectious Diseases (INMI), IRCCS, Rome, Italy
| | | |
Collapse
|
42
|
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
|
43
|
Drain PK, Dalmat RR. The elusive allure of a rapid host blood signature for tuberculosis disease. J Clin Microbiol 2024; 62:e0128923. [PMID: 38270458 PMCID: PMC10865849 DOI: 10.1128/jcm.01289-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2024] Open
Abstract
A rapid host transcriptional signature cartridge could be a major advancement for tuberculosis diagnosis and treatment monitoring. In a recent study, M. Li, Y. Qiu, M. Guo, R. Qu, et al. (J Clin Microbiol 61:e00911-23, 2023, https://doi.org/10.1128/jcm.00911-23) conducted an evaluation of the Cepheid 3-gene assay (Xpert-MTB-HR) within a diagnostic case-control study in China. While the study provides a strong contribution for determining the value of the Xpert-MTB-HR assay for diagnostic accuracy and treatment response, further assay optimization and more prospective studies are necessary before adaptation into clinical practice.
Collapse
Affiliation(s)
- Paul K. Drain
- Department of Global Health, University of Washington, Seattle, Washington, USA
| | - Ronit R. Dalmat
- Department of Global Health, University of Washington, Seattle, Washington, USA
| |
Collapse
|
44
|
Olbrich L, Verghese VP, Franckling-Smith Z, Sabi I, Ntinginya NE, Mfinanga A, Banze D, Viegas S, Khosa C, Semphere R, Nliwasa M, McHugh TD, Larsson L, Razid A, Song R, Corbett EL, Nabeta P, Trollip A, Graham SM, Hoelscher M, Geldmacher C, Zar HJ, Michael JS, Heinrich N. Diagnostic accuracy of a three-gene Mycobacterium tuberculosis host response cartridge using fingerstick blood for childhood tuberculosis: a multicentre prospective study in low-income and middle-income countries. THE LANCET. INFECTIOUS DISEASES 2024; 24:140-149. [PMID: 37918414 PMCID: PMC10808504 DOI: 10.1016/s1473-3099(23)00491-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 07/22/2023] [Accepted: 07/25/2023] [Indexed: 11/04/2023]
Abstract
BACKGROUND Childhood tuberculosis remains a major cause of morbidity and mortality in part due to missed diagnosis. Diagnostic methods with enhanced sensitivity using easy-to-obtain specimens are needed. We aimed to assess the diagnostic accuracy of the Cepheid Mycobacterium tuberculosis Host Response prototype cartridge (MTB-HR), a candidate test measuring a three-gene transcriptomic signature from fingerstick blood, in children with presumptive tuberculosis disease. METHODS RaPaed-TB was a prospective diagnostic accuracy study conducted at four sites in African countries (Malawi, Mozambique, South Africa, and Tanzania) and one site in India. Children younger than 15 years with presumptive pulmonary or extrapulmonary tuberculosis were enrolled between Jan 21, 2019, and June 30, 2021. MTB-HR was performed at baseline and at 1 month in all children and was repeated at 3 months and 6 months in children on tuberculosis treatment. Accuracy was compared with tuberculosis status based on standardised microbiological, radiological, and clinical data. FINDINGS 5313 potentially eligible children were screened, of whom 975 were eligible. 784 children had MTB-HR test results, of whom 639 had a diagnostic classification and were included in the analysis. MTB-HR differentiated children with culture-confirmed tuberculosis from those with unlikely tuberculosis with a sensitivity of 59·8% (95% CI 50·8-68·4). Using any microbiological confirmation (culture, Xpert MTB/RIF Ultra, or both), sensitivity was 41·6% (34·7-48·7), and using a composite clinical reference standard, sensitivity was 29·6% (25·4-34·2). Specificity for all three reference standards was 90·3% (95% CI 85·5-94·0). Performance was similar in different age groups and by malnutrition status. Among children living with HIV, accuracy against the strict reference standard tended to be lower (sensitivity 50·0%, 15·7-84·3) compared with those without HIV (61·0%, 51·6-69·9), although the difference did not reach statistical significance. Combining baseline MTB-HR result with one Ultra result identified 71·2% of children with microbiologically confirmed tuberculosis. INTERPRETATION MTB-HR showed promising diagnostic accuracy for culture-confirmed tuberculosis in this large, geographically diverse, paediatric cohort and hard-to-diagnose subgroups. FUNDING European and Developing Countries Clinical Trials Partnership, UK Medical Research Council, Swedish International Development Cooperation Agency, Bundesministerium für Bildung und Forschung; German Center for Infection Research (DZIF).
Collapse
Affiliation(s)
- Laura Olbrich
- Division of Infectious Diseases and Tropical Medicine, LMU University Hospital, LMU Munich, Munich, Germany; German Centre for Infection Research (DZIF), Partner Site Munich, Munich, Germany; Fraunhofer Institute ITMP, Immunology, Infection and Pandemic Research, Munich, Germany; Oxford Vaccine Group, Department of Paediatrics and the NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Valsan P Verghese
- Pediatric Infectious Diseases, Department of Pediatrics, Christian Medical College, Vellore, India
| | - Zoe Franckling-Smith
- Department of Paediatrics and Child Health, SA-MRC Unit on Child and Adolescent Health, University of Cape Town, Cape Town, South Africa
| | - Issa Sabi
- Mbeya Medical Research Centre, National Institute for Medical Research, Mbeya, Tanzania
| | - Nyanda E Ntinginya
- Mbeya Medical Research Centre, National Institute for Medical Research, Mbeya, Tanzania
| | - Alfred Mfinanga
- Mbeya Medical Research Centre, National Institute for Medical Research, Mbeya, Tanzania
| | - Denise Banze
- Instituto Nacional de Saúde, Marracuene, Mozambique
| | - Sofia Viegas
- Instituto Nacional de Saúde, Marracuene, Mozambique
| | - Celso Khosa
- Instituto Nacional de Saúde, Marracuene, Mozambique
| | - Robina Semphere
- Helse Nord Tuberculosis Initiative, Department of Pathology, Kamuzu University of Health Sciences, Blantyre, Malawi
| | - Marriott Nliwasa
- Helse Nord Tuberculosis Initiative, Department of Pathology, Kamuzu University of Health Sciences, Blantyre, Malawi
| | - Timothy D McHugh
- Centre for Clinical Microbiology, University College London, London, UK
| | - Leyla Larsson
- Division of Infectious Diseases and Tropical Medicine, LMU University Hospital, LMU Munich, Munich, Germany; German Centre for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - Alia Razid
- Division of Infectious Diseases and Tropical Medicine, LMU University Hospital, LMU Munich, Munich, Germany; German Centre for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - Rinn Song
- Oxford Vaccine Group, Department of Paediatrics and the NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Elizabeth L Corbett
- Helse Nord Tuberculosis Initiative, Department of Pathology, Kamuzu University of Health Sciences, Blantyre, Malawi; Clinical Research Department, London School of Hygiene & Tropical Medicine, London, UK
| | - Pamela Nabeta
- Foundation for Innovative New Diagnostics (FIND), Geneva, Switzerland
| | - Andre Trollip
- Foundation for Innovative New Diagnostics (FIND), Geneva, Switzerland
| | - Stephen M Graham
- Department of Paediatrics, University of Melbourne and Murdoch Children's Research Institute, Melbourne, VIC, Australia
| | - Michael Hoelscher
- Division of Infectious Diseases and Tropical Medicine, LMU University Hospital, LMU Munich, Munich, Germany; CIHLMU Center for International Health, LMU University Hospital, LMU Munich, Munich, Germany; German Centre for Infection Research (DZIF), Partner Site Munich, Munich, Germany; Fraunhofer Institute ITMP, Immunology, Infection and Pandemic Research, Munich, Germany; Unit Global Health, Helmholtz Zentrum München, German Research Center for Environmental Health (HMGU), Neuherberg, Germany
| | - Christof Geldmacher
- Division of Infectious Diseases and Tropical Medicine, LMU University Hospital, LMU Munich, Munich, Germany; German Centre for Infection Research (DZIF), Partner Site Munich, Munich, Germany; Fraunhofer Institute ITMP, Immunology, Infection and Pandemic Research, Munich, Germany
| | - Heather J Zar
- Department of Paediatrics and Child Health, SA-MRC Unit on Child and Adolescent Health, University of Cape Town, Cape Town, South Africa
| | | | - Norbert Heinrich
- Division of Infectious Diseases and Tropical Medicine, LMU University Hospital, LMU Munich, Munich, Germany; CIHLMU Center for International Health, LMU University Hospital, LMU Munich, Munich, Germany; German Centre for Infection Research (DZIF), Partner Site Munich, Munich, Germany; Fraunhofer Institute ITMP, Immunology, Infection and Pandemic Research, Munich, Germany.
| |
Collapse
|
45
|
Verma R, Ellappan K, Kempsell KE, Joseph NM. Triage test to diagnose presumptive pulmonary tuberculosis. Lancet Glob Health 2024; 12:e175-e176. [PMID: 38245104 DOI: 10.1016/s2214-109x(23)00604-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 12/18/2023] [Indexed: 01/22/2024]
Affiliation(s)
- Renu Verma
- Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, India; Institute of Bioinformatics, International Tech Park, Bangalore, India
| | - Kalaiarasan Ellappan
- Department of Microbiology, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry 605006, India
| | - Karen E Kempsell
- Science Group: Research and Evaluation, UK Health Security Agency, Salisbury, UK
| | - Noyal Mariya Joseph
- Department of Microbiology, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry 605006, India.
| |
Collapse
|
46
|
Gupta-Wright A, Ha H, Abdulgadar S, Crowder R, Emmanuel J, Mukwatamundu J, Marcelo D, Phillips PPJ, Christopher DJ, Nhung NV, Theron G, Yu C, Nahid P, Cattamanchi A, Worodria W, Denkinger CM. Evaluation of the Xpert MTB Host Response assay for the triage of patients with presumed pulmonary tuberculosis: a prospective diagnostic accuracy study in Viet Nam, India, the Philippines, Uganda, and South Africa. Lancet Glob Health 2024; 12:e226-e234. [PMID: 38245113 PMCID: PMC11046618 DOI: 10.1016/s2214-109x(23)00541-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 11/06/2023] [Accepted: 11/09/2023] [Indexed: 01/22/2024]
Abstract
BACKGROUND Non-sputum-based triage tests for tuberculosis are a priority for ending tuberculosis. We aimed to evaluate the diagnostic accuracy of the late-prototype Xpert MTB Host Response (Xpert HR) blood-based assay. METHODS We conducted a prospective diagnostic accuracy study among outpatients with presumed tuberculosis in outpatient clinics in Viet Nam, India, the Philippines, Uganda, and South Africa. Eligible participants were aged 18 years or older and reported cough lasting at least 2 weeks. We excluded those receiving tuberculosis treatment in the preceding 12 months and those who were unwilling to consent. Xpert HR was performed on capillary or venous blood. Reference standard testing included sputum Xpert MTB/RIF Ultra and mycobacterial culture. We performed receiver operating characteristic (ROC) analysis to identify the optimal cutoff value for the Xpert HR to achieve the target sensitivity of 90% or more while maximising specificity, then calculated diagnostic accuracy using this cutoff value. This study was prospectively registered with ClinicalTrials.gov, NCT04923958. FINDINGS Between July 13, 2021, and Aug 15, 2022, 2046 adults with at least 2 weeks of cough were identified, of whom 1499 adults (686 [45·8%] females and 813 [54·2%] males) had valid Xpert HR and reference standard results. 329 (21·9%) had microbiologically confirmed tuberculosis. Xpert HR had an area under the ROC curve of 0·89 (95% CI 0·86-0·91). The optimal cutoff value was less than or equal to -1·25, giving a sensitivity of 90·3% (95% CI 86·5-93·3; 297 of 329) and a specificity of 62·6% (95% CI 59·7-65·3; 732 of 1170). Sensitivity was similar across countries, by sex, and by subgroups, although specificity was lower in people living with HIV (45·1%, 95% CI 37·8-52·6) than in those not living with HIV (65·9%, 62·8-68·8; difference of 20·8%, 95% CI 13·0-28·6; p<0·0001). Xpert HR had high negative predictive value (95·8%, 95% CI 94·1-97·1), but positive predictive value was only 40·1% (95% CI 36·8-44·1). Using the Xpert HR as a triage test would have reduced confirmatory sputum testing by 57·3% (95% CI 54·2-60·4). INTERPRETATION Xpert HR did not meet WHO minimum specificity targets for a non-sputum-based triage test for pulmonary tuberculosis. Despite promise as a rule-out test that could reduce confirmatory sputum testing, further cost-effectiveness modelling and data on acceptability and usability are needed to inform policy recommendations. FUNDING National Institute of Allergy and Infectious Diseases of the US National Institutes of Health. TRANSLATIONS For the Vietnamese and Tagalog translations of the abstract see Supplementary Materials section.
Collapse
Affiliation(s)
- Ankur Gupta-Wright
- Division of Infectious Disease and Tropical Medicine and German Centre for Infection Research, Heidelberg University Hospital, Heidelberg, Germany; Institute for Global Health, University College London, London, UK.
| | - Huy Ha
- Hanoi Lung Hospital, Hanoi, Viet Nam
| | - Shima Abdulgadar
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Rebecca Crowder
- UCSF Center for Tuberculosis, San Francisco General Hospital, University of California San Francisco, San Francisco, CA, USA
| | - Jerusha Emmanuel
- Department of Pulmonary Medicine, Christian Medical College, Vellore, India
| | - Job Mukwatamundu
- World Alliance for Lung and Intensive Care Medicine in Uganda, Kampala, Uganda
| | - Danaida Marcelo
- De La Salle Medical Health Sciences Institute, Dasmariñas City, Cavite, Philippines
| | - Patrick P J Phillips
- UCSF Center for Tuberculosis, San Francisco General Hospital, University of California San Francisco, San Francisco, CA, USA; Division of Pulmonary and Critical Care Medicine, University of California San Francisco, San Francisco, CA, USA
| | | | | | - Grant Theron
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Charles Yu
- De La Salle Medical Health Sciences Institute, Dasmariñas City, Cavite, Philippines
| | - Payam Nahid
- UCSF Center for Tuberculosis, San Francisco General Hospital, University of California San Francisco, San Francisco, CA, USA; Division of Pulmonary and Critical Care Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Adithya Cattamanchi
- UCSF Center for Tuberculosis, San Francisco General Hospital, University of California San Francisco, San Francisco, CA, USA; Division of Pulmonary Diseases and Critical Care Medicine, University of California Irvine, Irvine, CA, USA
| | - William Worodria
- World Alliance for Lung and Intensive Care Medicine in Uganda, Kampala, Uganda; Division of Pulmonology, Mulago National Referral Hospital, Kampala, Uganda
| | - Claudia M Denkinger
- Division of Infectious Disease and Tropical Medicine and German Centre for Infection Research, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
47
|
Choe C, Andreasson JOL, Melaine F, Kladwang W, Wu MJ, Portela F, Wellington-Oguri R, Nicol JJ, Wayment-Steele HK, Gotrik M, Participants E, Khatri P, Greenleaf WJ, Das R. Compact RNA sensors for increasingly complex functions of multiple inputs. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.04.572289. [PMID: 38260323 PMCID: PMC10802310 DOI: 10.1101/2024.01.04.572289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Designing single molecules that compute general functions of input molecular partners represents a major unsolved challenge in molecular design. Here, we demonstrate that high-throughput, iterative experimental testing of diverse RNA designs crowdsourced from Eterna yields sensors of increasingly complex functions of input oligonucleotide concentrations. After designing single-input RNA sensors with activation ratios beyond our detection limits, we created logic gates, including challenging XOR and XNOR gates, and sensors that respond to the ratio of two inputs. Finally, we describe the OpenTB challenge, which elicited 85-nucleotide sensors that compute a score for diagnosing active tuberculosis, based on the ratio of products of three gene segments. Building on OpenTB design strategies, we created an algorithm Nucleologic that produces similarly compact sensors for the three-gene score based on RNA and DNA. These results open new avenues for diverse applications of compact, single molecule sensors previously limited by design complexity.
Collapse
Affiliation(s)
- Christian Choe
- Department of Bioengineering, Stanford University School of Medicine, Stanford, CA, USA
| | - Johan O. L. Andreasson
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Current address: Airity Technologies, Redwood City, CA, USA
| | - Feriel Melaine
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
| | - Wipapat Kladwang
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
- Current address: Inceptive, Palo Alto, CA, USA
| | - Michelle J. Wu
- Program in Biomedical Informatics, Stanford University School of Medicine, Stanford, CA, USA
- Current address: Verily Life Sciences, South San Francisco, CA, USA
| | - Fernando Portela
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
- Eterna Massive Open Laboratory
| | - Roger Wellington-Oguri
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
- Eterna Massive Open Laboratory
| | - John J. Nicol
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
- Eterna Massive Open Laboratory
| | | | - Michael Gotrik
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
- Current address: Protillion Biosciences, Burlingame, CA, USA
| | | | - Purvesh Khatri
- Stanford Center for Biomedical Informatics Research, Stanford University, Stanford, CA, USA
- Stanford Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA
| | - William J. Greenleaf
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Rhiju Das
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
- Program in Biomedical Informatics, Stanford University School of Medicine, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA
| |
Collapse
|
48
|
Xin R, Cheng Q, Chi X, Feng X, Zhang H, Wang Y, Duan M, Xie T, Song X, Yu Q, Fan Y, Huang L, Zhou F. Computational Characterization of Undifferentially Expressed Genes with Altered Transcription Regulation in Lung Cancer. Genes (Basel) 2023; 14:2169. [PMID: 38136991 PMCID: PMC10742656 DOI: 10.3390/genes14122169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 11/19/2023] [Accepted: 11/27/2023] [Indexed: 12/24/2023] Open
Abstract
A transcriptome profiles the expression levels of genes in cells and has accumulated a huge amount of public data. Most of the existing biomarker-related studies investigated the differential expression of individual transcriptomic features under the assumption of inter-feature independence. Many transcriptomic features without differential expression were ignored from the biomarker lists. This study proposed a computational analysis protocol (mqTrans) to analyze transcriptomes from the view of high-dimensional inter-feature correlations. The mqTrans protocol trained a regression model to predict the expression of an mRNA feature from those of the transcription factors (TFs). The difference between the predicted and real expression of an mRNA feature in a query sample was defined as the mqTrans feature. The new mqTrans view facilitated the detection of thirteen transcriptomic features with differentially expressed mqTrans features, but without differential expression in the original transcriptomic values in three independent datasets of lung cancer. These features were called dark biomarkers because they would have been ignored in a conventional differential analysis. The detailed discussion of one dark biomarker, GBP5, and additional validation experiments suggested that the overlapping long non-coding RNAs might have contributed to this interesting phenomenon. In summary, this study aimed to find undifferentially expressed genes with significantly changed mqTrans values in lung cancer. These genes were usually ignored in most biomarker detection studies of undifferential expression. However, their differentially expressed mqTrans values in three independent datasets suggested their strong associations with lung cancer.
Collapse
Affiliation(s)
- Ruihao Xin
- Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, College of Computer Science and Technology, Jilin University, Changchun 130012, China; (R.X.); (Y.W.); (M.D.); (L.H.)
- Jilin Institute of Chemical Technology, College of Information and Control Engineering, Jilin 132000, China; (Q.C.); (X.C.); (H.Z.)
| | - Qian Cheng
- Jilin Institute of Chemical Technology, College of Information and Control Engineering, Jilin 132000, China; (Q.C.); (X.C.); (H.Z.)
| | - Xiaohang Chi
- Jilin Institute of Chemical Technology, College of Information and Control Engineering, Jilin 132000, China; (Q.C.); (X.C.); (H.Z.)
| | - Xin Feng
- School of Science, Jilin Institute of Chemical Technology, Jilin 132000, China;
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun 130012, China;
| | - Hang Zhang
- Jilin Institute of Chemical Technology, College of Information and Control Engineering, Jilin 132000, China; (Q.C.); (X.C.); (H.Z.)
| | - Yueying Wang
- Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, College of Computer Science and Technology, Jilin University, Changchun 130012, China; (R.X.); (Y.W.); (M.D.); (L.H.)
| | - Meiyu Duan
- Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, College of Computer Science and Technology, Jilin University, Changchun 130012, China; (R.X.); (Y.W.); (M.D.); (L.H.)
| | - Tunyang Xie
- Centre for Mathematical Sciences, University of Cambridge, Wilberforce Road, Cambridge CB3 0WA, UK;
| | - Xiaonan Song
- Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, College of Software, Jilin University, Changchun 130012, China;
| | - Qiong Yu
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun 130012, China;
| | - Yusi Fan
- Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, College of Software, Jilin University, Changchun 130012, China;
| | - Lan Huang
- Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, College of Computer Science and Technology, Jilin University, Changchun 130012, China; (R.X.); (Y.W.); (M.D.); (L.H.)
| | - Fengfeng Zhou
- Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, College of Computer Science and Technology, Jilin University, Changchun 130012, China; (R.X.); (Y.W.); (M.D.); (L.H.)
- School of Biology and Engineering, Guizhou Medical University, Guiyang 550025, China
| |
Collapse
|
49
|
Wu X, Tan G, Ma J, Yang J, Guo Y, Lu H, Ke H, Li M, Tang YW, Sha W, Yu F. Assessment of the Cepheid 3-gene Host Response Fingerstick Blood Test (MTB-HR) on rapid diagnosis of tuberculosis. Emerg Microbes Infect 2023; 12:2261561. [PMID: 37848021 PMCID: PMC10583623 DOI: 10.1080/22221751.2023.2261561] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 09/17/2023] [Indexed: 10/19/2023]
Abstract
ABSTRACTThe World Health Organization has identified high-priority target product profiles for new TB diagnostics which include rapid biomarker-based, non-sputum-based diagnostic testing, using an easily accessible sample. The Cepheid 3-gene Host Response Fingerstick Blood Prototype Test (MTB-HR) quantifies relative mRNA levels of a 3-gene signature (GBP5, DUSP3, and KLF2) from a whole-blood sample on the GeneXpert platform. The objective of the present study was to evaluate the performance of the MTB-HR to distinguish between active tuberculosis (ATB), latent Mycobacterium tuberculosis infection (LTBI), other pulmonary diseases, and healthy volunteers at a tertiary care centre. Among 653 participants enrolled in this study, 192 were diagnosed as having ATB, and the remaining 461 were classified as non-ATB, including 137 cases of LTBI, 224 cases of other pulmonary diseases, and 100 healthy volunteers. The corresponding AUCs of the MTB-HR in distinguishing untreated ATB from non-ATB, LTBI, other pulmonary diseases, and healthy volunteers were 0.814 (95% CI, 0.760-0.868, sensitivity 76.1%, specificity 71.6%), 0.739 (95% CI, 0.667-0.812, sensitivity 59.7%, specificity 78.1%), 0.825 (95% CI, 0.770-0.880, sensitivity 82.1%, specificity 65.6%), 0.892 (95% CI, 0.839-0.945, sensitivity 76.1%, specificity 88.0%), respectively. When only samples with TAT of less than 1 h were included, the AUC of the MTB-HR in distinguishing untreated ATB from non-ATB was largest, 0.920 (95% CI, 0.822-1.000, sensitivity 81.3%, specificity 87.7%). In conclusion, the MTB-HR assay shows potential as a rapid, blood-based screening and triage test for ATB, especially for untreated ATB, with the advantage of increased diagnostic yield since blood is more readily available.
Collapse
Affiliation(s)
- Xiaocui Wu
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, People’s Republic of China
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, People’s Republic of China
| | - Guangkun Tan
- Department of Clinical Laboratory, Shanghai University of Traditional Chinese Medical Attached Shuguang Hospital, Shanghai, People’s Republic of China
| | - Jian Ma
- Medical Affairs, Danaher Diagnostic Platform People’s Republic of China/Cepheid, Shanghai, People’s Republic of China
| | - Juan Yang
- Department of Tuberculosis, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, People’s Republic of China
| | - Yinjuan Guo
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, People’s Republic of China
| | - Haiwen Lu
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, People’s Republic of China
| | - Hui Ke
- Department of Tuberculosis, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, People’s Republic of China
| | - Mengran Li
- Department of Biostatistics & Data Management, Beckman Coulter People’s Republic of China, Danaher, Shanghai, People’s Republic of China
| | - Yi-Wei Tang
- Medical Affairs, Danaher Diagnostic Platform People’s Republic of China/Cepheid, Shanghai, People’s Republic of China
| | - Wei Sha
- Department of Tuberculosis, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, People’s Republic of China
| | - Fangyou Yu
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, People’s Republic of China
- Department of Laboratory Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People’s Republic of China
| |
Collapse
|
50
|
Li M, Qiu Y, Guo M, Qu R, Tian F, Wang G, Wang Y, Ma J, Liu S, Takiff H, Tang YW, Gao Q. Evaluation of the Cepheid 3-gene host response blood test for tuberculosis diagnosis and treatment response monitoring in a primary-level clinic in rural China. J Clin Microbiol 2023; 61:e0091123. [PMID: 37902328 PMCID: PMC10662368 DOI: 10.1128/jcm.00911-23] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Accepted: 09/20/2023] [Indexed: 10/31/2023] Open
Abstract
A rapid, accurate, non-sputum-based triage test for diagnosing tuberculosis (TB) is a high-priority need. Cepheid developed a novel prototype blood test, Xpert Mycobacterium tuberculosis Host Response (Xpert-MTB-HR), which generates a TB score based on the mRNA expression of three genes. We conducted a case-control study with prospective recruitment to evaluate its accuracy in the clinic of the Wusheng County Centers for Disease Prevention and Control in China. We enrolled 149 TB patients, 248 other respiratory diseases (ORD) patients, and 193 healthy controls. In addition, whole-blood samples taken from TB patients after 2, 5, and 6 months of treatment were tested with Xpert-MTB-HR to evaluate its ability to monitor treatment response. Xpert-MTB-HR discriminated between TB and healthy controls with an area under the curve (AUC) of 0.912 (95% CI, 0.878-0.945). With the specificity of 70% envisioned for a triage test, its sensitivity was 90.1% (84.9%-94.6%). Xpert-MTB-HR discriminated between TB and ORD with an AUC of 0.798 (0.750-0.847), and at specificity of 70%, the sensitivity was only 75.8% (68.5%-82.8%). In patients determined by Ultra to have medium or high sputum bacillary loads, with specificity of 70%, the sensitivity for discriminating patients with TB from healthy controls was 100.0% (100.0-100.0) and from patients with ORD, 95.1% (89.8-100.0). The TB scores generally increased by 2 months of treatment and then remained stable. Xpert-MTB-HR met the criteria for a triage test to discriminate between TB and healthy controls, but not between TB and ORD, except when limited to patients with high sputum bacillary loads. Xpert-MTB-HR showed promise for monitoring response to treatment but needs to be further evaluated in larger prospective studies.
Collapse
Affiliation(s)
- Meng Li
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Science, Shanghai Medical College, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
| | - Yong Qiu
- Wusheng County Center for Disease Control and Prevention, Guang’an, China
| | - Mingcheng Guo
- Wusheng County Center for Disease Control and Prevention, Guang’an, China
| | - Rong Qu
- Wusheng County Center for Disease Control and Prevention, Guang’an, China
| | - Fajun Tian
- Wusheng County Center for Disease Control and Prevention, Guang’an, China
| | - Gengsheng Wang
- Wusheng County Center for Disease Control and Prevention, Guang’an, China
| | - Ya Wang
- Wusheng County Center for Disease Control and Prevention, Guang’an, China
| | - Jian Ma
- Medical Affairs, Danaher Diagnostic Platform/Cepheid, Shanghai, China
| | - Siyuan Liu
- Medical Affairs, Danaher Diagnostic Platform/Cepheid, Shanghai, China
| | - Howard Takiff
- Laboratorio de Genética Molecular, CMBC, Instituto Venezolano de Investigaciones Científicas, IVIC, Caracas, Venezuela
| | - Yi-Wei Tang
- Medical Affairs, Danaher Diagnostic Platform/Cepheid, Shanghai, China
| | - Qian Gao
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Science, Shanghai Medical College, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
| |
Collapse
|