SUMMARY
Despite the advent of new diagnostics, drugs and regimens, tuberculosis (TB) remains a global public health threat. A significant challenge for TB control efforts has been the monitoring of TB therapy and determination of TB treatment success. Current recommendations for TB treatment monitoring rely on sputum and culture conversion, which have low sensitivity and long turnaround times, present biohazard risk, and are prone to contamination, undermining their usefulness as clinical treatment monitoring tools and for drug development. We review the pipeline of molecular technologies and assays that serve as suitable substitutes for current culture-based readouts for treatment response and outcome with the potential to change TB therapy monitoring and accelerate drug development.
KEYWORDS: tuberculosis, treatment monitoring, biomarkers, outcome
INTRODUCTION
Despite significant progress made in the diagnosis and management of patients in the recent past, tuberculosis (TB) remains a global public health threat with 10 million new cases and 1.5 million deaths annually (1). Recommended TB treatment regimens are long and often associated with severe adverse events, which impact both adherence and therapy outcome (2–5). To monitor treatment progress, clinicians traditionally apply a number of clinical, radiological, and laboratory biomarkers. The World Health Organization (WHO) recommends use of regular sputum microscopy and culture for monitoring of treatment and molecular and phenotypic drug susceptibility testing for identifying the Mycobacterium tuberculosis complex (MTBC) drug resistance profile and adjusting the individualized treatment regimen accordingly (3, 6). Although sputum culture conversion (i.e., at month 2 for drug-susceptible TB and at month 6 for drug-resistant TB) has been proposed as a surrogate for treatment outcome (7–9), its use is limited by a long turnaround time, relatively low sensitivity, and high risk of contamination.
Fourteen-day early bactericidal activity (EBA) studies, based on the quantification of TB bacilli in sputum samples and their decline or rise after initiation of treatment, may also be conducted to assess bactericidal activity of antitubercular drugs and their combinations. EBA studies have traditionally relied on the well-established methodology of counting CFU. This approach has several limitations, being both labor-intensive and requiring quality sputum samples with high bacterial burden to provide positive cultures throughout the period of study (10). As an alternative, bactericidal activity may be determined in liquid culture by noting the prolongation of time to positivity (TTP) from baseline (10). This approach gives a more accurate estimate of bacterial burden, but the long time to result limits its use in guiding adaptive trial designs and for clinical management.
TB treatment success is ultimately determined using standardized treatment outcome definitions that are also critically important for the development and testing of novel drugs. The WHO recommends defining TB treatment outcome based on sputum culture positivity and the need for treatment termination or permanent regimen change of at least two anti-TB drugs due to culture reversion, development of drug resistance, or adverse drug reactions (11). However, these definitions do not account for relapse-free survival, which is a more clinically relevant assessment of treatment efficacy (9). To address this, simplified treatment outcomes based on culture conversion and reversion and/or TB relapse within 1 year after treatment completion have been proposed by the TB Network European Trials Group (TBnet) (9). Recently, the WHO proposed updated TB outcome definitions, including an optional definition of sustained treatment success based on the absence of TB relapse within 6 months in case of drug-susceptible and 12 months in case of drug-resistant TB; however, this definition has been designated for operational research use only (12). While all current definitions continue to rely on sputum and culture conversion as monitoring tools, the drawbacks of these methods are recognized, and the need for new treatment monitoring tools is highlighted (12).
Diagnostic technology advances have revolutionized the management of TB. As the most important example, sputum-based, molecular assays, including GeneXpert (Cepheid, Inc., USA), TrueNat (Molbio Diagnostics Private Limited, India), and the line-probe assays (e.g., Hain Lifescience GmbH, Germany), have led to the early detection of drug resistance against important anti-TB drugs (13). Consequently, there is a pipeline of molecular technologies and assays with the potential to change TB therapy monitoring concepts. Such technologies are also potential substitutes for current culture-based readouts for treatment response and outcome. Since phase IIA/IIB clinical studies rely on bacteriological measurements, novel biomarkers may also be potential substitutes for MTBC culture in novel trial designs, accelerating drug development (14). Simpler tools to assess TB therapy response should also be able to signal TB treatment termination (i.e., according to patient response, as opposed to current standard regimen duration). The tests should be both simple and sensitive enough to replace smear microscopy in low-resource settings (15), ideally generating test results within few hours, and have very high diagnostic performance to predict relapse compared to clinical and microbiological follow-up of patients. Ideally, the test should use nonsputum samples to better ensure operator safety, and result output should preferably be quantitative to allow for better assessment of treatment response over time. The test should be suitable for both TB and extrapulmonary TB patients, including children and those with HIV coinfection.
The aim of this review is to highlight several biomarker candidates with the potential to improve treatment monitoring, accelerate drug development, and ultimately redefine treatment outcome definitions, enabling personalized medicine for TB.
HOST CHARACTERISTICS ASSAYS
Assays measuring the host response to TB infection as a correlate of disease severity or measure of treatment response and cure represent one option to simplify TB treatment monitoring. These approaches include tracking host immune responses through transcriptomic profiling, metabolomic profiling, and monitoring cytokine levels, but also can involve the use of imaging technologies as well as the evaluation of clinical signs and symptoms (Table 1) (16–18). Such technologies are attractive options for treatment monitoring given that they can be performed on readily accessible, nonsputum patient samples (e.g., blood or urine) obtainable in primary health care clinics (PHC) where TB patients often first enter the health care cascade. However, the development of these technologies has generally been challenged given the large range of host variabilities, including infection pathology, host drug metabolism, disease endpoints, coinfections, and even environmental factors (19, 20), as well as the practicalities of translating biomarkers into simple, rapid tests more suitable for PHC. The state and potential of current assays based upon the identification of host biomarkers or characteristics for treatment monitoring are detailed below.
TABLE 1.
Name/type | Host response cartridge | RISK6 score | Transcriptomics panel | TB22 | Other RNA signatures | TAM-TB assay | IGRAs | Cytokines | CRP assay | Metabolomics (e.g., tryptophan catabolism) | Deep learning imaging (CAD, CXR) | CT scan | 18F-FDG PET-CT | TB scores |
---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|
Developer | Cepheid | QuantumDX | Biomerieux | Research Center Borstel | Various | Ludwig-Maximilian University of Munich | Qiagen, Oxford Immunotec | Various | Various | Various (LC-MS/MS) | Qure.ai and others | Various | Various | BANDIM |
Concept | 3-Marker mRNA signature capturing an inflammatory response associated with TB | 6-Marker mRNA signature capturing an inflammatory response associated with TB | 30-Marker mRNA signature capturing an inflammatory response associated with TB | 22-Marker mRNA signature associated with relapse-free cure | RNA signatures with different no. of gene targets involved | Detection of upregulated activation markers on MTB antigen-specific T cells associated with active TB disease | IFN-γ release upon specific stimulation | Release of cytokines upon specific stimulation | Quantitative measurement of CRP | Measurement of tryptophan catabolism | AI-based abnormality scoring of paired digital CXR: change in abnormality score is associated with Rx response | Extent of TB lesions on CT scan and change over time | Change of tracer uptake in TB lesions (PET) and lesion change over time (CT) | Baseline and on-Rx clinical markers of risk can predict individual patient Rx success |
Use-case | TB detection, prediction, Rx monitoring | TB detection, prediction, Rx monitoring | TB detection, Rx monitoring | TB diagnosis, Rx monitoring | TB diagnosis, Rx monitoring | TB detection in children, Rx monitoring | Diagnosis of latent TB infection | Diagnosis of latent TB infection, Rx monitoring | Rx monitoring | Rx monitoring | TB detection, Rx monitoring | TB detection, Rx monitoring | TB detection, Rx monitoring | TB detection, Rx monitoring |
Development stage | RUO, design locked | RUO, design locked | RUO, design locked | In-house | In-house | Design locked | RUO, design locked | In-house | RUO, design locked | Experimental, in-house | On market | RUO, design locked | RUO, design locked; additional radiotracers being evaluated | NA |
Sample | 50 μL of capillary blood | 50 μL of capillary blood | Tempus tube | PaxGene tube | PaxGene tube | Whole blood, PBMCs | Whole blood, PBMCs | Whole blood, PBMCs | Serum/plasma | Serum/plasma | CXR images and CAD scoring | CT images, trained evaluation | PET-CT images, trained evaluation | None |
Instrument | GeneXpert 6- or 10-color instrument | Q-POC platform with cassette | BioFire FilmArray platform | Array or RNA-seq | Array or RNA-seq | Fluorescence-activated cell sorting | ELISA, ELISPOT | ELISA | Various | Various | Digital CXR, CAD/AI software | Various | Various | None |
Time to result | 45 min | 30 min | 120 min | 8 h | 1–2 days | 10 h | 1 day | 1 day | 2 h | Days | <1 min | 5 min | 1 h | Dependent on contributing variables |
Relevant clinical data | 3-Gene signature expression level changed significantly with respect to baseline for 31 pulmonary TB patients who were microbiologically cured by end of Rx. In first month, a median score of 1.97 (IQR = 1.03–2.33) suggested a promising Rx response (150). | Study in patients with and without HIV infection in Africa, Peru and Brazil. RISK6 differentiated between patients with cure and Rx failure (AUC 77.1, 95% CI = 52.9–100) (29). | This assay is currently being validated for diagnosis of active TB in conjunction with risk assessment, clinical context and diagnostic information in South Africa (ClinicalTrialsgov identifier NCT0499540) The Rx monitoring potential of the assay remains unclear. | TB22 model predicted clinical outcomes for TB patients without HIV infection with an AUC of up to 94%, (95% CI = 0.9–0.98) in patients mainly from Europe and Africa (21). | Large list of RNA signatures potentially serving as Rx monitoring tools based upon early exptl data showing Rx monitoring potential (24, 25, 27, 151–153). Certain signatures were identified in TB patients without HIV infection at risk of Rx failure 1–4 wks after start of Rx in South Africa (25, 151). | Frequencies of TB-specific cells from peripheral blood significantly declined over the course of Rx in a cohort without HIV from sub-Saharan Africa (n = 39) with active TB and a 16-yr-old HIV-negative patient with extrapulmonary TB from Afghanistan (33, 154). | A systematic review of 30 studies (24 QFT, 3 T-SPOT.TB, and 3 used both) noted a general decline of IFN-γ over time but with a large variation, concluding that the markers are of no use for Rx monitoring (41). | Several studies have described IFN-γ, TNF-α, IL-4, IL-6, and IP-10 as potential markers for Rx monitoring, though the significantly up-regulated data obtained to date is too heterogeneous to draw a conclusion that these cytokines are useful for Rx monitoring (42–45). | This assay has shown potential for Rx monitoring in active TB patients (with and without HIV infection). One study of HIV-positive and -negative patients in South Africa showed CRP decline to ≤55% of the baseline value by wk 2, a prediction of hospitalization or death with 99% negative predictive value was described (44–46). | A study in samples from 48 TB patients, 20 TB-DM patients, and 48 healthy controls without HIV infection from Indonesia revealed a three-target model to distinguish between groups (AUC = 0.91–0.97) and identified metabolites that lowered during Rx. Another study conducted in samples from HIV-negative, pulmonary TB patients from Georgia focused on decreased tryptophan/kynurenine ratios, which changed under Rx, and identified indoleamine 2,3-dioxygenase-1 as a potential target for host-directed therapy (52, 53). | Deep learning software has mainly been applied to CXRs from active TB patients at baseline for TB detection (64, 155, 156). Its value as Rx motoring tool is not yet well described. | CT findings correlate with Rx outcome, with larger cavities identified during Rx having a high predictive value for Rx failure in an HIV-uninfected South African drug-susceptible TB cohort. Cavity wall thickness and the total volume of intraparenchymal radio-dense lesions at the end of Rx were also found to have high predictive value of Rx failure in this cohort (65–67). | The potential of PET-CT scans to serve as markers for Rx shortening is currently under evaluation in a randomized control trial. PET-CT has correlated with clinical and sputum markers in HIV-negative TB patients in an EBA trial in South Africa (NCT02821832) (79, 80). | Several Rx response scores have been presented involving clinical, microbiological and radiological data. A risk stratification tool using patient demographics and clinical parameters (from phase III trials) in drug-susceptible, HIV-negative TB patients identified groups at low, moderate, and high risk for poor outcome in Germany (67, 85–88, 157). |
AI, artificial intelligence; CAD, computer-aided detection; CRP, C-reactive protein; CT, chest tomography; CXR, chest X-ray; DM, diabetes mellitus; ELISA, enzyme-linked immunosorbent assay; IGRA, interferon gamma release assay; LC, liquid chromatography; MS, mass spectrometry; MTB, Mycobacterium tuberculosis; NA, not applicable; PBMC, peripheral blood mononuclear cell; PET, positron emission tomography; QFT, QuantiFERON-TB; RUO, research use only; Rx, treatment; TAM, T cell activation marker; TB, tuberculosis.
Transcriptomic Profiling
Transcriptomic analyses from whole blood have yielded RNA signatures that correlate with future onset of disease, active TB, and cure (21–24). These signatures often feature genes related to type I interferon signaling (25, 26). Tracking RNA changes in whole blood appears to be a suitable approach for TB therapy monitoring since changes can be found both very early and at later stages of therapy in cohorts, including patients from Africa and Europe with or without HIV coinfection (21, 27–29). However, the potential of these approaches for TB treatment monitoring has yet to be confirmed in larger clinical trials. Still, whole-blood RNA assessment may be used as an alternative surrogate marker for EBA trials since changes appear rapidly following therapy initiation (27). In addition, since RNA alterations can be detected even following culture conversion, RNA may also be a suitable signal for phase IIB study endpoints and ultimately to indicate cure (21, 28). Recently, it was shown that positron emission tomography-computed tomography (PET/CT) changes over the course of TB therapy could be correlated with whole-blood RNA changes (30), though, to date, these changes have not been correlated with relapse in large-scale clinical trials. Furthermore, the potential for secure usage of transcriptomic data involving swarm intelligence to differentiate diseases such as COVID-19 or TB has been presented, supporting this approach (31). Currently, assays for TB treatment monitoring based upon transcriptomic signatures are still in early to middle stages of development, though the pipeline includes blood-based assays suitable for use at various levels of the health care system, from community clinics to district laboratories (32).
Host Adaptive Responses
Cellular immunological markers have promise as candidate nonsputum diagnostics for TB suitable for both adults and children (33). Stimulation of T cells with TB antigens and intracellular cytokine staining specifically measures these markers on TB-specific T cells. While processing of peripheral blood mononuclear cells for this purpose requires specialized capacity and has posed a significant obstacle to the application of this method, a new format for the T cell activation (TAM)-TB assay allows the use of whole blood (34). Notably, based on this method, activation markers CD38, HLA-DR, and Ki-67 on TB-specific T cells have been demonstrated to increase at the time of TB diagnosis and to decline with treatment in 10 patients with active, drug-susceptible TB over a 9-month period (35). Ahmed et al. have also noted a slower decline of those markers in HIV-negative patients with active, smear positive pulmonary TB who had a longer time to sustained sputum culture conversion over a 26-week treatment period (with liquid cultures performed at successive time points up to week 26), the primary endpoint used in the PanACEA MAMS TB-01 treatment trial (clinicaltrials.gov NCT01785186), with strong correlation (33). However, these validations of cellular immunological markers were made only against surrogate culture-based endpoints, not against the clinical endpoint of patient cure versus unfavorable outcome. In addition, it was demonstrated that CD27, which appeared to hold promise as a diagnostic marker, did not change with successful treatment in this small (39 patient) cohort. A principal limitation will be that the assay will only work in individuals with a measurable cellular immune response, i.e., a detectable IFN-γ response. However, certain cellular markers have been identified that demonstrate some correlation with baseline disease severity (36, 37), raising the prospect of sputum-independent diagnostic tests that can also inform of disease severity and prognosis, hence aiding treatment decisions and providing a measurement of treatment response in comparison to baseline. These tests, however, currently require facilities for incubation and flow cytometry, limiting their use to research laboratories or other facilities with similar equipment (38). If these assays could be established on the same cytometry platforms used for CD4 testing for HIV care, a wider availability could potentially be achieved. Additional evaluations in trial settings will be needed to confirm the clinical relevance of these markers for TB treatment monitoring.
Cytokines and Other Biomarkers
Proinflammatory cytokines produced by TB-specific T cells are well established as significant contributors to TB immunity (39, 40), as demonstrated by the inability of the host to control infection in their absence. The importance of IFN-γ expression in TB-infected subjects is reflected in the assay used to determine infection in the first place: commercially available IFN-γ release assays (IGRAs) such as QuantiFERON TB Gold (Qiagen, Germany) and T-SPOT-TB (Oxford Immunotec, Ltd., UK) assays frequently replace tuberculin skin tests (TSTs) to identify patients infected with TB. While levels of IFN-γ production in IGRAs have been suggested as a potential biomarker for treatment monitoring, a systematic review of 30 studies did not find uniform patterns of IGRA levels following treatment due to the high degree of variation between participants, undermining the usefulness of these tests for treatment monitoring (41). A more promising approach may be to directly measure cytokines circulating in plasma or serum samples, removing the added variability inherent to in vitro stimulation. Several technologies are available for this purpose, including simple enzyme-linked immunosorbent assays (ELISAs), as well as multiplexed bead-based and plate-based approaches (42–44). Using these approaches, it was demonstrated that several cytokines, including IFN-γ (42), TNF-α (42), IL-4 (42), IL-6 (44), and IP-10 (45), were increased in active TB disease compared to healthy or latently infected individuals, confirming their association with disease severity, and declined with TB treatment. These studies included evaluations of protein levels in blood samples from 319 active TB, HIV-positive and -negative patients at baseline versus following 8 weeks of combination therapy in TBTC Study 29 (44), plasma samples from 42 HIV-positive and -negative individuals with active pulmonary TB undergoing 26 weeks of treatment followed over 18 months in Durban, South Africa (45), and 67 patients with drug-susceptible TB disease followed over 6 months of treatment in Beijing, China (42). Unfortunately, even though multiple studies have measured the same cytokines, the evidence base is heterogeneous, suggesting poor sensitivity and specificity of these markers for the prediction of treatment progress. To date, no cytokine-based assay for treatment monitoring has progressed from early development stages, and currently used technologies are limited to central laboratories.
Beside cytokines, new or updated proteomic approaches have identified additional serum proteins as potential biomarkers for anti-TB treatment monitoring. Commercially available, C-reactive protein assays based upon finger-prick blood testing have already undergone WHO review and recommendation for community triage in people living with HIV (32) and the C-reactive protein has additionally been shown to consistently decline during treatment (44, 46, 47). In an assessment of 33 patients in a high-TB and -HIV setting with serial changes in C-reactive protein evaluated over 8 weeks (46), the failure of C-reactive protein to reduce to ≤55% of the baseline value by week 2 predicted hospitalization or death with 99% negative predictive value for patients with confirmed or presumed TB. Apolipoproteins and members of the complement cascade are also modulated by TB treatment and have been suggested as components of predictive proteomic signatures (48–50).
In addition, one study with access to patients who relapsed following treatment completion assessed whether a proteomic signature would be able to predict treatment outcome (51). Ronacher et al. showed distinct patterns of changes in immune markers (including cytokines, chemokines, soluble receptors and acute phase proteins) following treatment initiation in 78 cured and 12 relapsed HIV-negative patients with drug-susceptible TB from the Action TB Study who were used as a discovery cohort. A combination of four immune markers (TNF-β, sIL-6R, IL-12p40, and IP-10), in addition to TTP and body mass index at diagnosis, was able to discriminate relapsed from cured patients with an Area Under the Receiver Operating Characteristics (AUROC) curve of 0.819 (95% confidence interval [CI] = 0.679 to 0.942) in this discovery cohort, and this was validated in a second cohort of 23 cured and 17 relapse patients from Uganda and Brazil with an AUC of 0.718 (95% CI = 0.509 to 0.903). Interestingly, the predictive potential of this biosignature was most pronounced when measured at baseline since immune markers tended to normalize during treatment (51). As multiplex assays, such as those of SomaLogic, Inc. (USA), and O-link Holdings AB (Sweden), are becoming more widely available, confirmatory studies will allow for a targeted assessment of some of these markers as potential predictors of treatment outcome.
Metabolomic approaches are similarly being assessed for their potential for anti-TB treatment monitoring. Multiple metabolic pathways are affected by TB infection, with tryptophan catabolism in particular being heavily influenced by TB and its treatment (52, 53). Of note, this metabolic pathway is also being explored as a target of host-directed therapies, with blockade of indoleamine 2,3-dioxygenase (IDO; the rate-limiting host enzyme for catabolism of tryptophan to kynurenine) activity showing promising results in reducing clinical manifestations and pathological correlates of TB in a macaque model (54). Further studies will be necessary to determine whether these early findings translate clinically for TB patient treatment monitoring.
Imaging
Chest X-rays (CXRs) are a fast and inexpensive way to screen for active TB with many portable devices now available for testing in community settings. However, the process is subject to interpretation and thus the accuracy of this method largely depends on the experience of the reader. The use of computer-aided detection (CAD) technologies for TB detection in digital CXRs has the potential to standardize interpretation and improve the feasibility of CXRs for widespread TB screening and diagnosis (55). CAD software generates a score from 0 to 100 with some products, such as qXR from Qure.ai (India), further allowing comparisons of images obtained during treatment to provide a quantitative measure of treatment success (56). Recently, the WHO recommended three specific commercially available CAD software packages after finding the diagnostic accuracy and the overall performance of the software to be similar to the interpretation of digital CXR by a human reader (57). Many studies have additionally demonstrated the potential for CXR to be used for monitoring bacterial load and estimating TB disease severity alone and in combination with smear microscopy (58–61), with evidence of the association between extensive parenchymal involvement and 2-month culture conversion, as well as the number of cavities with relapse in non-HIV-infected patients (62). However, despite strong associations with patient-important treatment outcomes (58), studies have had conflicting results, as an assessment of the potential for treatment shortening in patients with noncavitary TB (with culture and CXR) found that month 2 sputum culture conversion and the absence of cavitation was insufficient to support treatment shortening (63). Ultimately, this use case has not been well evaluated for CAD software to date, with most studies using human readers (62, 63), and the potential of this technology for treatment monitoring remains questionable (64).
Chest computed tomography (CT) is more sensitive and specific for active TB diagnosis than CXR given cross-sectional imaging and higher resolution, and changes in CT findings over time have been shown to correlate with treatment outcome (65). Notation of greater volume of cavities on chest CT performed at baseline, month 1, and month 6 of treatment had a high predictive value for treatment failure in an HIV-uninfected South African drug-susceptible TB cohort (66), consistent with evidence that the presence and extent of cavities at initiation and conclusion of TB treatment is a risk factor for poor treatment outcome or delayed culture conversion (67–69). CT-detected cavity wall thickness and the total volume of intraparenchymal radiodense lesions at the end of treatment were also found to have high predictive value of treatment failure in the same cohort (66). However, only eight patients had treatment failure in this specific cohort, and it is notable that a significant proportion of individuals continue to have residual chest CT abnormalities even after apparent successful completion of TB treatment. It can be difficult to determine the presence of live TB bacteria in these lesions and the risk for subsequent disease recurrence with CT alone (70, 71). Combining 18F-fluorodeoxyglucose (18F-FDG) positron emission tomography (PET) with CT imaging (PET/CT) is an area of great interest for TB treatment monitoring to address this limitation. 18F-FDG accumulates in metabolically active cells, including inflammatory cells typically involved in active TB lesions (i.e., neutrophils, macrophages, and lymphocytes) (72). PET/CT thus overlays functional information about metabolic activity onto the pathological lesions identified on CT. Data from multiple cohorts have demonstrated that PET glycolytic activity decreases in response to effective TB treatment and can predict treatment outcome (31, 66, 73–78), though persistent lesions with ongoing inflammation, as well as new inflammatory lesions after completion of successful TB treatment, have also been noted (75, 76). The significance of these heterogenous patterns is unclear. The ability of PET/CT, along with other clinical and microbiologic parameters, to identify individuals with drug-susceptible TB for whom first-line anti-TB treatment can be shortened to 4 months is currently being tested in a randomized trial (NCT02821832) (79). PET/CT is also being explored for its potential role in identifying new drugs and regimens in EBA trials through the information it provides on lesion volume and PET glycolytic activity (80). The high cost and logistical complexity of performing PET/CT limit its broader implementation as a treatment monitoring tool, but this biomarker nonetheless holds promise as a potentially useful translational tool for confirming preclinical results of promising new drugs and regimens in earlier stage phase I/II trials before undertaking large expensive phase III trials.
Clinical Score, Signs, and Lung Function
A range of risk factors have been shown to be associated with poor TB patient outcomes, including demographic variables such as age, sex, ethnic background, and occupation, and clinical variables such as clinical form of TB, history of TB treatment, disease severity (e.g., smear grade and cavitary disease), comorbidities (e.g., HIV coinfection and malnutrition), and clinical signs and symptoms (e.g., weight, cough, lung function, and chest pain) (67, 81–84). Although current practice guidelines suggest that certain risk factors may indicate the need for adjustment of treatment regimens (e.g., treatment duration) (82), clinical risk scores and risk stratification algorithms using a suite of readily available predictors can be useful to further differentiate individuals to monitor treatment response, in both drug-susceptible and drug-resistant TB populations (67, 85–88). For example, a risk stratification tool has been developed using patient demographics and clinical parameters from pooled data obtained in contemporary phase III trials in drug-susceptible TB-treated patients, leading to identifying groups of low, moderate, and high risk for poor outcome (67, 89). In a patient population where the high-risk group had unfavorable outcome rates of roughly 30%, the tool provided support for the use of clinical scoring systems and algorithms to triage patients at baseline or during treatment to improve the probability of treatment success. Although clinical scoring systems and algorithms may provide a simple approach to evaluate response to treatment, careful consideration is required on the mechanisms for handling of missing data, on the cost, resources, and devices/tools necessary to measure variables, and on the high interobserver variability for subjective measurements (e.g., clinical symptoms). Importantly, more research is needed to assess the utility of clinical scoring systems to predict treatment outcomes and monitor treatment response across regimens of various compositions and dosing, as well as by demographics (e.g., ethnicity and sex) and clinical factors (e.g., weight and HIV status) that have also been associated with poor outcomes (68, 90–92). The overall moderate performance of clinical scoring systems may benefit from combination with drug exposure variables, along with other more quantitative and sensitive bacterial or host-derived markers, to serve as integrated tools to maximize precision in predicting treatment outcomes.
PATHOGEN BURDEN AND FITNESS ASSAYS
Treatment monitoring tools based upon the quantification of viable bacteria allow for better precision in TB clinical care by providing a specific metric for TB treatment efficacy and outcome prediction (93). Although TB burden may be inferred by growth-based MGIT culture, this method is time-consuming, requires sophisticated laboratory infrastructure, and is prone to contamination. While molecular methods to estimate TB burden such as Xpert MTB/RIF (Cepheid, Inc.) can overcome these limitations, the presence of DNA from dead or killed bacilli, which can remain detectable for months and even years after treatment, limits the use and interpretation of findings (94, 95). In this context, rapid and simple molecular assays to quantify both MTBC burden and viability in a patient sample present useful options for TB treatment monitoring (Table 2).
TABLE 2.
Name/type | Solid and liquid culture | Xpert MTB/RIF ultra | PMA/eMA pCR | TB LAM assays | TB-MBLA | Culture-free TB test | rRNA synthesis ratio | tNGS (e.g., Deeplex Myc-TB) | WGS |
---|---|---|---|---|---|---|---|---|---|
Developer | BD and others | Cepheid | Various | Otsuka, LSI Medience Corporation, FujiFilm Corp., SD Biosensor, Inc. | LifeArc | Tauns | University of Colorado | Genoscreen | Illumina and others |
Concept | Time to positivity in liquid culture/count of CFU on solid culture | Semiquantitative detection of mycobacterial DNA. Use of Xpert cycle threshold for estimate of bacterial burden and treatment monitoring | Membrane-permeable dyes modify and prevent amplification of dead cell-derived DNA, allowing quantification of viable bacteria via PCR or RT-PCR | Quantification of LAM in sputum as a correlate for MTBC burden | Quantification of rRNA as a correlate with the burden of live bacilli | Quantification of MPT64 released from live MTBC following 1 h of heat treatment of the sample correlating with MTBC burden | Quantification of the ratio of rRNA and liable splice as a correlate of bacterial burden and fitness | tNGS targets genes associated with TB drug resistance and identifies lineage | Genome sequencing provides complete mutation information for MTBC, including identification of TB drug resistance mutation and MTBC lineage |
Use-case | EBA, Rx monitoring | Diagnosis of TB, Rx monitoring | EBA, Rx monitoring | EBA, Rx monitoring | EBA, Rx monitoring | EBA, Rx monitoring | EBA, Rx monitoring | Differentiating new infection/relapse; identification of resistance and resistant populations | Differentiating new infection/relapse, Identification of resistance and resistant populations |
Development stage | On market | On market | In-house | RUO | RUO, on market | CE-MARK | In-house | CE-MARK | In-house |
Sample | Sputum | Sputum | Sputum | Sputum and urine | Sputum | Sputum or other | Sputum | Sputum | Sputum |
Instrument | MGIT | 6- and 10-color GeneXpert System | In-house PCR | Various ELISA and CLEIA and associated platforms (e.g., PATHFAST) | PCR | Automated ELISA | PCR | Illumina, Nanopore | Illumina, Nanopore |
Time to result | Up to 8 wks | 2 h | 4–6 h | Unknown | <4 h | 2.5–3 h | Unknown | 48 h | 48 h |
Relevant clinical data | Time to culture conversion, as well as 2- and 6-month culture conversion status, are currently used as surrogates for Rx response and outcome. The number of CFU on solid media or the time to positivity are also used to quantify bacteria in EBA studies (8, 9, 158–160). | Xpert Ultra appeared to be of no use as Rx monitoring tool in HIV-positive and -negative TB patients from the REMOX trial (n = 221, Cape Town, South Africa, and Mbeya, Tanzania). Furthermore, Xpert results had a high sensitivity but very low specificity when smear and culture were used as reference standard (sensitivity (97.0%, 95% CI = 95.8−97.9), specificity (48.6%, 95% CI = 45.0−52.2), though results are conflicting and the use of cycle threshold for treatment monitoring is still being explored (161–163). | The method seems to be promising to specifically detect viable MTBC bacilli, with a specificity of 84.6% and a sensitivity of 84.6% compared to counting CFU. No clinical evaluation has been performed to evaluate the potential of this technology for Rx monitoring to date (107–109). | A study involving 57 TB patients showed that HIV-positive and -negative participants with positive LAM test at month 2 had a 5.6-fold (95% CI = 1.2 to 25.2) greater risk of mortality. Another study involving 40 drug-susceptible pulmonary TB patients demonstrated that decline of sputum LAM concentrations during the first 56 days of therapy correlated with increases in time to culture positivity, with notable changes during the first 14 days (120, 121). | The MBLA corresponded well with culture in several early-phase clinical studies. In a study involving samples from 178 HIV-positive and -negative patients from Southeast Africa, it was shown that individuals with high pretreatment bacillary burdens were less likely to convert to negative by wk 8 of Rx than those with a low burden (96, 97, 100). | A study conducted in Taiwan involving 1102 patients with suspected TB infection revealed a sensitivity of 86.9% and a specificity of 92.0% with culture as reference standard. A follow-up study with HIV-negative TB patients from Japan showed a specificity of 89.5% to predict negative culture results on day 14 (114, 115). | The rRNA synthesis ratio was quantified for sputa from 17 Ugandan, 28 Vietnamese, and 19 Beninese HIV-positive and -negative patients treated with HRZE for drug-susceptible pulmonary TB. Rx was demonstrated to lead to a rapid decline of the rRNA synthesis ratio (106). | tNGS has been demonstrated to rapidly provide clinical data to guide personalized Rx and Rx management, with the potential for detection of acquired drug resistance, including low-level resistant populations, during Rx (140, 164). | WGS is the method currently used to fully, genotypically characterize MTB, allowing for differentiation between reinfection and relapse in principle. WGS, like tNGS, can also guide personalized Rx and Rx management with the potential for detection of acquired drug resistance, including heteroresistance (137, 165). |
CLEIA, chemiluminescence enzyme immunoassay; EBA, early bactericidal activity; ELISA, enzyme-linked immunosorbent assay; EMA, ethidium monoazide; LAM, lipoarabinomannan; MBLA, molecular bacterial load assay; MPT64, M. tuberculosis complex protein 64; MTBC, Mycobacterium tuberculosis complex; PMA, propidium monoazide; RT-PCR, real-time PCR; RUO, research use only; Rx, treatment; TB, tuberculosis; tNGS, targeted next-generation sequencing; WGS, whole-genome sequencing.
Sputum-Based Molecular Load Assays
The TB molecular bacterial load assay (TB-MBLA) is a culture-free assay that specifically detects and quantifies the viable MTBC bacillary load from a patient’s sample via reverse transcriptase quantitative PCR (RT-qPCR) in <4 h (96). By detecting 16S rRNA (rRNA), a component of the multiple ribosomes in a viable cell, the test can detect as few as 10 to 100 bacteria in a milliliter of sample (97–101). Targeting RNA makes TB-MBLA an accurate measure of live bacteria and distinguishes it from molecular tests that detect DNA (99, 100) and differentiates the assay from culture-based methods of monitoring the response to anti-TB therapy, where the TTP increases as patients clear their bacterial load (100). As early as 3 days after the initiation of therapy, TB-MBLA effectively detects the bactericidal effect of anti-TB therapy and is able to provide long-term assessment of slow treatment responders (100, 102). The assay has the added advantage of detecting viable, nonculturable MTBC bacilli, obviating complicating factors of media variability and dormant cell states (103, 104), as well as the need to decontaminate sputum, removing a significant variable in laboratory processing (99). It is also possible to perform the assay following heat inactivation of sputum samples with a small, but predictable, loss in measured viable count (105), reducing the need for expensive, high-containment facilities. A design locked TB-MBLA is entering clinical evaluation with the hope that, if successful, it can be introduced into the clinic as a suitable substitute for culture conversion for monitoring treatment response.
The mycobacterial precursor rRNA synthesis ratio assay has additionally shown promise in quantifying the ability of various anti-TB treatment regimens to potentially shorten treatment (106). This assay operates on the principle that rRNA synthesis in MTBC is distinctly impacted by sterilizing versus nonsterilizing drugs. Sterilizing drugs and highly effective drug regimens profoundly suppress M. tuberculosis rRNA synthesis, whereas nonsterilizing drugs and weaker regimens do not. Thus, the rRNA synthesis ratio provides a metric for drug effect and bactericidal activity that may help to differentiate sterilizing drugs and regimens that shorten treatment and promote cure (i.e., suppressing MTBC rRNA synthesis), as opposed to drugs and regimens that allow infecting MTBC to maintain rRNA synthesis. Although this technology is only recently being developed for TB treatment monitoring, assays including quantification of rRNA synthesis to this end may serve as a marker of the ability of a drug or drug regimen to shorten TB treatment.
Another sputum-based option to discriminate viable bacilli is to use propidium monoazide (PMA)- or ethidium monoazide (EMA)-PCR or real-time PCR. The PMA and EMA dyes can penetrate the bacterial membrane, where they modify DNA derived from dead cells, preventing PCR amplification. These assays have been demonstrated to rapidly discriminate dead from viable M. tuberculosis cells in early studies (107–109), avoiding the limitations of PCR-based assays such as Xpert MTB/RIF and Xpert MTB/RIF Ultra (Cepheid, Inc.), which are capable of detecting DNA from dead bacteria (94, 95), though further clinical evaluations will be necessary to determine the potential of PMA/EMA assays for anti-TB treatment monitoring and outcome.
The shorter time to result and the absence of the need to decontaminate samples mean that RNA-based assays like the TB-MBLA and rRNA synthesis ratio assays and DNA based assays such as PMA/EMA have the potential to inform clinical decision-making in real time (100). Furthermore, the decline in bacillary load correlates with patient resolution of clinical signs, particularly cough, and differentiates the patient response to different regimens (100, 110), which widens the application of these approaches to clinical trial monitoring.
M. tuberculosis Complex Protein 64 Release Assays
Studies suggest that the load and fitness of bacteria in a sample can be quantified by providing nutrients to it and measuring the induced protein expression response (111), by incorporating fluorescent trehalose in the M. tuberculosis membrane (112), or by conducting short-term culture with resuscitation promoting factors (113). Another recently described approach is to briefly expose the patient sample to 46°C heat to specifically induce the release of M. tuberculosis complex protein 64 (MPT64) from live bacilli (114). The MPT64 release assay has demonstrated 88% sensitivity and 97% specificity for TB detection, with MPT64 release strongly correlating with both sputum smear grade and MGIT TTP (115). When measured in sequentially collected sputum samples (days 0, 14, and 28) from 50 active pulmonary TB patients undergoing treatment, MPT64 release was strongly correlated with treatment response, with a sensitivity of 81% (95% CI = 58.1 to 94.6) for predicting positive day 28 cultures (116). The MPT64 release assay is currently under early development by Tauns Laboratories, Inc. (Japan), and in particular, given the simplicity and speed of the test, this approach could provide a useful alternative to the time- and labor-demanding phenotypic methods currently used for treatment monitoring, though it will likely still be limited to central laboratories, and it has been demonstrated to have some lineage-specific limitations (i.e., MPT64 is a poor predictor for L5 MTBC strains) (116). In addition, there are some concerns that MTBC genetic polymorphisms in the mpt64 gene change the MPT64 antigen sufficiently to result in false-negative results for MPT64 detection assays (117).
Lipoarabinomannan
Lipoarabinomannan (LAM) is a prominent glycolipid in the cell wall of M. tuberculosis with utility as a pathogen biomarker to assess patient therapeutic response (118, 119). As demonstrated by Drain et al., LAM decreases during anti-TB therapy, and patients with detectable LAM after an intensive phase of therapy appear to have greater mortality risk (120). As a marker of active TB, LAM is an attractive target for TB diagnostics and treatment monitoring, including one assay measuring LAM in sputum developed by LSI Medience Corporation (Japan) for EBA studies and treatment monitoring (121), and a number of urine-based LAM diagnostics, e.g., the SILVAMP TB test by FujiFilm Corporation (Japan) and the TB LAM assay by SD Biosensor, Inc. (Republic of Korea) (122, 123). Compared to sputum, urine samples are easily obtained from patients, have shown no evidence of TB transmission, and provide an opportunity to better diagnose TB in patients who have trouble producing sputum, such as HIV patients, children, and patients with extrapulmonary TB (120). Urine-based lateral-flow tests are also rapid, and there is no need for sophisticated laboratory infrastructure or highly trained technicians, making these technologies appropriate for use in community settings. However, there is generally less LAM present in urine compared to sputum (124), and the performance of LAM-based assays can be highly variable (125, 126), although their general performance appears to be at least as good as smear (121). In addition to urine-based LAM, serum-based LAM detection presents another option for treatment monitoring. However, even less LAM is present in serum compared to urine, and therefore lower detection limits of serum-based LAM diagnostics must be achieved to ensure these technologies are sufficiently sensitive to be useful as treatment monitoring tools. A number of LAM-based assays are at various stages of development, though to date only the urine-based LAM assays have demonstrated adequate diagnostic accuracy while maintaining ease of use and rapid time to result (121). Generally, these assays have shown higher sensitivity for HIV-positive compared to HIV-negative subgroups, with diagnostic performance correlating with immunosuppression (127). Clinical studies will be key to determine the potential of these assays for anti-TB treatment monitoring in all patient populations.
Whole-Genome Sequencing, Strains, and Fitness
MTBC has been considered to be of clonal origin due to the low level of overall genomic variation (128). However, very little consideration has been given to the fact that lineages could influence the clinical picture of the disease as well as drug response. Emerging data now associate specific lineages with clinical manifestations, including increased virulence of infections and the capacity to acquire drug resistance (129–133), directly impacting treatment response. In addition, some lineages may have an increased MIC to new anti-TB drugs and may require both an adjustment of the dosage and the critical concentration to be tested “in vitro” to inform treatment regimen selection (134, 135). For these reasons, it is crucial that any new trial for drugs and diagnostics includes settings where different lineages are sufficiently represented and drug effects are considered in light of MTBC lineage and sublineage.
Whole-genome sequencing (WGS) not only provides important information regarding MTBC lineage but also serves to screen for drug resistance development during treatment (136). Although WGS can be performed directly from smear-positive samples (137), it is mainly performed on isolates grown in culture, even though early liquid cultures are suitable specimens to lower the time to result. Targeted next-generation sequencing (tNGS), which detects known and novel variants in specific gene regions as opposed to the entire genome, is another attractive option for use in primary samples for similar purposes, with the advantage of detecting the presence of multiple strains in a primary specimen (i.e., mixed infection) and allowing the detection of minority variants bearing mutations associated with drug resistance (138). Since there is evidence that even slightly elevated MICs to first-line drug compounds can impact treatment outcome (139), tNGS presents a distinct advantage to identify resistance markers even when present in minority populations. In addition, the use of tNGS/WGS on smear-positive samples during therapy is the most efficient tool to promptly identify emergence of resistance. In addition, sequencing is the only tool currently capable of providing genomic information for gene regions associated with resistance to new and repurposed drugs.
Most importantly, the information provided by tNGS can be crucial to differentiate relapse from new infection, especially in case of strain selection during culture. Therefore, it is necessary to collect samples throughout therapy to enable comparative analysis in case of relapse or reinfection after therapy end. Although WGS and tNGS technologies are mostly limited to high-level, high-throughput testing centers with sufficient infrastructure and well-trained staff to perform these assays, they are both powerful tools for treatment response monitoring and provide key information for any anti-TB treatment response trial. New user-friendly and low-cost platforms such as the Oxford Nanopore Technologies Limited (Oxford, UK) MinION sequencer have the potential to move sequencing closer to point of need, supporting the management of people affected by TB and drug-resistant TB (140). The COVID-19 pandemic has increased the capacity to perform sequencing in a wide variety of settings, which also benefits the TB field. Currently, an array of sequencing solutions suitable for district labs are commercialized and on the pathway to WHO review (32).
MISCELLANEOUS
In addition to the previously referenced technologies, there is a variety of TB diagnostic innovations with TB treatment monitoring potential. For example, certain breath and cough tests operate under the principle that expelled air from TB patients harbors unique pathogen biomarkers or volatile organic compounds that can be used to diagnose infection, thereby providing a quick and potentially easy method for patient screening and detection. Although there are a number of developers investigating the potential of this type of technology (141–147), early clinical studies have shown variable results (sensitivity, 74 to 100%; specificity, 11 to 93%). The treatment monitoring and programmatic potential of this type of technology still remains unclear. Several manufacturers are also developing artificial intelligence (AI) algorithms to interpret and quantify images obtained by point-of-care ultrasound (148), sounds recorded with digital stethoscopes, and cough analyzers, and with the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic these tools have been fast tracked with promising results (149).
CONCLUSIONS AND FUTURE DIRECTIONS
TB biomarkers for therapy monitoring have the potential to serve as future surrogates for anti-TB treatment outcome in clinical trials and in routine clinical settings. However, few markers have been prospectively evaluated for this purpose and studies proving their applicability are desperately needed. In particular, studies with rigorous methodology to specifically evaluate treatment response are necessary, since the vast majority of clinical studies of biomarkers have only provided preliminary data, being underpowered, using only surrogate outcomes for treatment response, or failing to include sufficient time points or comparators. Although many markers appear promising for therapy monitoring or as markers for outcome prediction, most studies are not designed to prove the markers’ performance. Clinical evaluation in designated biomarker studies is especially important for markers that indicate cure during treatment, which may allow for personalized therapy durations for TB patients and may even identify those at risk for relapse after therapy end. Biomarkers signaling relapse risk would need to clearly identify relapse patients immediately following completion of standard or experimental therapies. It is important to note that most studies reporting clinical outcome as endpoint do not include relapse but rather indirect surrogates for therapy failure. This is especially important for studies evaluating biomarker guided individualized therapy durations, where studies would need to compare standard versus biomarker-guided therapy durations with relapse as most important endpoint. Regardless, long follow-up periods to exclude relapse will be necessary to underscore the potential of any biomarker to serve as a future outcome-defining surrogate.
Despite the promise of biomarkers for TB treatment and outcome monitoring, research in this area has not yet yielded a single marker that can sufficiently or completely substitute for established culture-based markers at any level of the health care system (32). It is very likely that no single marker can be used to identify all endpoints of interest (e.g., incipient disease, active disease, or cure versus relapse), and it may therefore be more promising to identify a marker’s disease stage-specific potential. For example, assays detecting sputum-based markers such as the MBLA are likely suitable indicators of bactericidal activity (i.e., in EBA trials) but might not indicate risk of relapse at the termination of therapy in phase III trials. In addition, a combined marker approach would likely improve the accuracy of individual markers for certain endpoints. In this context, a combination of host, pathogen, and imaging markers may eventually lead to suitable and rapid individual risk assessments. To accelerate the discovery of promising marker combinations, machine learning and artificial intelligence could be applied to large data sets to identify clinically interpretable marker sets or may even be included in individual assessments in the future.
In conclusion, there are a wide variety of TB treatment monitoring and outcome marker candidates currently under development or already on the market. The combination of these markers may be key to the comprehensive assessment of individual risks for various endpoints, and modern computational approaches are very suitable to accelerate marker identification and interpretation.
ACKNOWLEDGMENTS
This publication was written on behalf of all project partners of the UNITE4TB consortium (see www.unite4tb.org).
The UNITE4TB project has received funding from the Innovative Medicines Initiative 2 Joint Undertaking (JU) under grant agreement 101007873. The JU receives support from the European Union’s Horizon 2020 research and innovation program and EFPIA, Deutsches Zentrum für Infektionsforschung e. V. (DZIF), and Ludwig-Maximilians-Universität München (LMU). EFPIA/AP contribute to 50% of funding, whereas the contribution of DZIF and the LMU University Hospital Munich has been granted by the German Federal Ministry of Education and Research. See https://www.imi.europa.eu/.
The funders had no role in study design, data collection and interpretation, or the decision to submit the work for publication.
This communication reflects the views of members of the UNITE4TB consortium and neither IMI nor the European Union and EFPIA are liable for any use that may be made of the information contained here.
S.B.G. and M.R. declare that they are employed by FIND, the global alliance for diagnostics. FIND is a not-for-profit foundation that supports the evaluation of publicly prioritized tuberculosis assays and the implementation of WHO-approved (guidance and prequalification) assays using donor grants. FIND has product evaluation agreements with several private sector companies that design diagnostics for tuberculosis and other diseases. These agreements strictly define FIND’s independence and neutrality with regard to these private sector companies. N.H. reports receiving a product evaluation grant from a company developing TAM TB, and grants from EDCTP and German Center for Infection Research for product evaluations. S.H.G. reports receiving research grants to develop MBLA and also provides pro bono advice to LifeArc, which is developing the assay commercially.
Biographies
Jan Heyckendorf is a professor for respiratory care with focus on chronic inflammatory diseases at the Christian Albrechts University of Kiel and the University Hospital of Schleswig-Holstein in Kiel, Germany. Besides being a respiratory specialist, he is an ID and intensive care specialist. He has been working in the field of personalized medicine specially for tuberculosis patients. One of his major goals are the identification and validation of biomarkers for treatment response for tuberculosis patients, which was funded by the German Center for Infection Research (DZIF). He is the co-leader of the UNITE4TB work package biomarkers.
Sophia B. Georghiou, Ph.D., is a molecular epidemiologist with postgraduate training in molecular biology (M.S.) and global health (Ph.D.) from the University of California San Diego. She has more than a decade of infectious disease research experience, working with many different academic institutions, clinical laboratories and industry partners, and has contributed to a diversity of NTD, HIV and TB research projects. Her doctoral research focused upon the development and evaluation of next generation sequencing technologies for drug-resistant TB diagnosis. Sophia joined the TB program at FIND in August of 2016. Her work has informed WHO review and guideline development group meetings as well as technical documents for the use and implementation of TB diagnostics.
Nicole Frahm, Ph.D., is the Exploratory Biomarker Leader at the Bill and Melinda Gates Medical Research Institute and holds an affiliate Associate Professor appointment at the Fred Hutchinson Cancer Research Center. She received her Ph.D. in Immunology at the University of Hamburg, Germany, and completed her postgraduate work with a joint appointment at Massachusetts General Hospital and Harvard Medical School. During her career, Dr. Frahm studied the influence of HIV sequence diversity on its recognition by cytotoxic T lymphocytes, as well as the factors governing the recognition of sequence variants both in HIV-infected subjects and in vaccine trial participants. In her role as the Exploratory Biomarker Leader, she oversees the evaluation, prioritization, and implementation of cutting-edge biomarker technologies, with a particular focus on systems biology tools, across small molecule, biologics, vaccine and diagnostics programs across the portfolio of the Gates MRI. Dr. Frahm has published more than 100 peer-reviewed manuscripts.
Norbert Heinrich, M.D., is a unit head at University of Munich (LMU). He oversees the Division of Infectious Diseases and Tropical Medicine’s trials for development of new anti-TB drugs and diagnostics, coordinating the PanACEA trials MAMS TB 01, SUDOCU, and DECODE, and is leading the RaPaed TB and ERASE-TB diagnostic trials. Inbuilt into these trials, he is pursuing the validation of new markers for TB treatment response and co-leads the Unite4TB biomarker work package. He holds a degree as a pediatric specialist.
Irina Kontsevaya obtained her Ph.D. in Genetics of Mycobacterium tuberculosis at Samara State University, Russia. She worked as a Molecular Biologist and Research Project Coordinator at the N.V. Postnikov Samara Region Clinical Tuberculosis Dispensary, Samara, Russia, then as a Postdoctoral Researcher at Imperial College London, UK, and the Institute of Organic Chemistry and Biochemistry, Prague, Czech Republic. She is now a Postdoctoral Researcher at the Research Center Borstel, Germany and an Honorary Research Fellow at Imperial College London, UK. Being in the tuberculosis field for almost 14 years, Irina has particular interest in tuberculosis diagnostics, novel biomarkers of disease and cure and treatment monitoring tools as they are an essential part of successful management of people infected with tuberculosis, especially its drug-resistant forms.
Maja Reimann completed her Master’s degree in epidemiology at the University of Bremen, Germany, in 2017 and has since been working as an epidemiologist and statistician in the Clinical Infectious Diseases research group at the Research Center Borstel, Germany. She submitted her doctoral thesis to the University of Luebeck, Germany, in October 2021. In the course of her work, she became increasingly involved with bioinformatics aspects of research into personalized medicine in tuberculosis. She finds the possibilities offered by these approaches and by bringing together different expertise, such as biology and medicine, through good collaboration with colleagues interesting.
David Holtzman, M.D., is an infectious diseases physician based in Boston, MA. He received his undergraduate and medical degrees at the University of Pennsylvania in Philadelphia and completed his specialty training in internal medicine, pediatrics, and infectious diseases in Philadelphia. He also completed a Masters degree in Control of Infectious Diseases at the London School of Hygiene and Tropical Medicine. David has worked in Lesotho for Baylor International Pediatric AIDS Initiative, Elizabeth Glaser Pediatric AIDS Foundation, and most recently with Partners In Health as a Senior MDR TB Clinician. He is currently the Clinical Development Leader for TB Therapeutics at the Gates Medical Research Institute. David has over a decade of experience working in eastern and southern Africa focused on TB and HIV. He is committed to improving the lives of people with TB through improved TB treatment options and models of care.
Marjorie Imperial, Ph.D., is a postdoctoral scholar in the Department of Bioengineering and Therapeutic Sciences at the University of California, San Francisco. She received her BS in chemical engineering from the University of Washington in 2014 and her PhD in pharmaceutical sciences and pharmacogenomics from the University of California—San Francisco in 2020. Her research is in quantitative clinical pharmacology and involves development of evidence-based tools to gain comprehensive understanding in the interplay between disease dynamics, optimal regimens, and drug and biomarker response, with a focus on infectious diseases. During her Ph.D. and postdoctoral training, she applied model-based principles to characterize treatment response in patients with tuberculosis.
Daniela M. Cirillo, M.D., is a board-certified clinical microbiologist, Head of the Emerging Bacterial Pathogens Unit at the San Raffaele Scientific Institute (HSR) in Milan, Italy, and director of the WHO/Union TB Supranational Reference laboratory and WHO Collaborating Centre for “integrated laboratory strengthening on tuberculosis and other emerging infections.” She and her collaborators provide technical support to several countries where TB is endemic. She participates in international and national working groups on tuberculosis, including the WHO ACSM Working Group, the national subgroup of mycobacteriology, working group for national TB recommendations, writing committee for the national mycobacteriology manual. She is a member of the TBnet steering committee, the executive secretary of Stop TB Italia and of the Infection Control Committee of the HSR. She is the coordinator of one of the FP7 large collaborative projects approved by the European Union for study and clinical management of TB drug resistance.
Stephen H. Gillespie, M.D., is Professor of Medicine at the University of St Andrews. He has been involved in various aspects of tuberculosis diagnosis and drug development, including the evaluation of new candidate agents in vitro and has led trials exploring moxifloxacin with the TB Alliance in REMoxTB, STAND and SimpliciTB. He is one of the Chief Investigators of the PanACEA consortium. He has developed SLIC (Scattered Light Integrating Collector), performing antibiotic susceptibility testing in <30 minutes. This innovation won a Longitude Prize Discovery Award the Scottish Life Science Alliance Innovation of the Year 2017. He has also developed the Molecular Bacterial Load Assay for tuberculosis, nontuberculosis mycobacteria and the organisms of COPD. This is an innovative way of detecting, quantifying and determining the viability of bacteria in a single assay to monitor treatment response. Both innovations are in late-stage clinical development.
Morten Ruhwald, M.D., joined FIND in 2019 as Head of the TB programme. He is a medical doctor with over 15 years of professional experience, including 4 in clinical medicine and 12 in research and development in the area of vaccines and diagnostics for TB. Morten has been project lead on several diagnostic tests for latent M. tuberculosis infection, including specific skin tests and new simpler in-vitro diagnostics in the IGRA family. He has worked extensively with international stakeholders in translational medicine and product development for poverty-related diseases. Prior to joining FIND, he was the Chief Medical Officer and Head of Human Immunology at the Center for Vaccine Research at Statens Serum Institute, Denmark, and before that he led the TB immunology group at the Copenhagen University Hospital. Morten obtained his medical degree and Ph.D. from Copenhagen University, Denmark. and has published more than 80 academic papers.
REFERENCES
- 1.WHO. 2020. Global tuberculosis report. World Health Organization, Geneva, Switzerland. [Google Scholar]
- 2.WHO. 2019. WHO consolidated guidelines on drug-resistant tuberculosis treatment. WHO consolidated guidelines on drug-resistant tuberculosis treatment. World Health Organization, Geneva, Switzerland. [Google Scholar]
- 3.WHO. 2017. Guidelines for treatment of drug-susceptible tuberculosis and patient care (2017 update). World Health Organization, Geneva, Switzerland. [Google Scholar]
- 4.Dorman SE, Nahid P, Kurbatova EV, Phillips PPJ, Bryant K, Dooley KE, Engle M, Goldberg SV, Phan HTT, Hakim J, Johnson JL, Lourens M, Martinson NA, Muzanyi G, Narunsky K, Nerette S, Nguyen NV, Pham TH, Pierre S, Purfield AE, Samaneka W, Savic RM, Sanne I, Scott NA, Shenje J, Sizemore E, Vernon A, Waja Z, Weiner M, Swindells S, Chaisson RE, Tuberculosis Trials Consortium. 2021. Four-month rifapentine regimens with or without moxifloxacin for tuberculosis. N Engl J Med 384:1705–1718. doi: 10.1056/NEJMoa2033400. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Munro SA, Lewin SA, Smith HJ, Engel ME, Fretheim A, Volmink J. 2007. Patient adherence to tuberculosis treatment: a systematic review of qualitative research. PLoS Med 4:e238–1245. doi: 10.1371/journal.pmed.0040238. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.WHO. 2018. Technical manual for drug susceptibility testing of medicines used in the treatment of tuberculosis. World Health Organization, Geneva, Switzerland. [Google Scholar]
- 7.Wallis RS, Maeurer M, Mwaba P, Chakaya J, Rustomjee R, Migliori GB, Marais B, Schito M, Churchyard G, Swaminathan S, Hoelscher M, Zumla A. 2016. Tuberculosis: advances in development of new drugs, treatment regimens, host-directed therapies, and biomarkers. Lancet Infect Dis 16:E34–E46. doi: 10.1016/S1473-3099(16)00070-0. [DOI] [PubMed] [Google Scholar]
- 8.Kurbatova EV, Cegielski JP, Lienhardt C, Akksilp R, Bayona J, Becerra MC, Caoili J, Contreras C, Dalton T, Danilovits M, Demikhova OV, Ershova J, Gammino VM, Gelmanova I, Heilig CM, Jou R, Kazennyy B, Keshavjee S, Kim HJ, Kliiman K, Kvasnovsky C, Leimane V, Mitnick CD, Quelapio I, Riekstina V, Smith SE, Tupasi T, van der Walt M, Vasilyeva IA, Via LE, Viiklepp P, Volchenkov G, Walker AT, Wolfgang M, Yagui M, Zignol M. 2015. Sputum culture conversion as a prognostic marker for end-of-treatment outcome in patients with multidrug-resistant tuberculosis: a secondary analysis of data from two observational cohort studies. Lancet Respir Med 3:201–209. doi: 10.1016/S2213-2600(15)00036-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Günther G, Lange C, Alexandru S, Altet N, Avsar K, Bang D, Barbuta R, Bothamley G, Ciobanu A, Crudu V, Danilovits M, Dedicoat M, Duarte R, Gualano G, Kunst H, de Lange W, Leimane V, Magis-Escurra C, McLaughlin A-M, Muylle I, Polcová V, Popa C, Rumetshofer R, Skrahina A, Solodovnikova V, Spinu V, Tiberi S, Viiklepp P, van Leth F, for TBNET. 2016. Treatment outcomes in multidrug-resistant tuberculosis. N Engl J Med 375:1103–1105. doi: 10.1056/NEJMc1603274. [DOI] [PubMed] [Google Scholar]
- 10.Diacon AH, Donald PR. 2014. The early bactericidal activity of antituberculosis drugs. Expert Rev Anti Infect Ther 12:223–237. doi: 10.1586/14787210.2014.870884. [DOI] [PubMed] [Google Scholar]
- 11.WHO. 2020. Definitions and reporting framework for tuberculosis: 2013 revision (updated December 2014 and January 2020) (WHO/HTM/TB/2013.2). European Surveillance: Bulletin Européen Sur Les Maladies Transmissibles (European Communicable Disease Bulletin). World Health Organization, Geneva, Switzerland. [Google Scholar]
- 12.WHO. 2021. Meeting report of the WHO expert consultation on drug-resistant tuberculosis treatment outcome definitions, 17–19 November 2020. World Health Organization, Geneva, Switzerland. [Google Scholar]
- 13.World Health Organization. 2017. Algorithm for laboratory diagnosis and treatment-monitoring of pulmonary tuberculosis and drug-resistant tuberculosis using state-of-the-art rapid molecular diagnostic technologies. World Health Organization, Geneva, Switzerland. [Google Scholar]
- 14.Phillips PPJ, Gillespie SH, Boeree M, Heinrich N, Aarnoutse R, McHugh T, Pletschette M, Lienhardt C, Hafner R, Mgone C, Zumla A, Nunn AJ, Hoelscher M. 2012. Innovative trial designs are practical solutions for improving the treatment of tuberculosis. J Infect Dis 205:S250–S257. doi: 10.1093/infdis/jis041. [DOI] [PubMed] [Google Scholar]
- 15.Denkinger CM, Dolinger D, Schito M, Wells W, Cobelens F, Pai M, Zignol M, Cirillo DM, Alland D, et al. 2015. Target product profile of a molecular drug-susceptibility test for use in microscopy centers. J Infect Dis 211(Suppl 2):S39–S49. doi: 10.1093/infdis/jiu682. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Lange C, Aarnoutse R, Chesov D, van Crevel R, Gillespie SH, Grobbel H-P, Kalsdorf B, Kontsevaya I, van Laarhoven A, Nishiguchi T, Mandalakas A, Merker M, Niemann S, Köhler N, Heyckendorf J, Reimann M, Ruhwald M, Sanchez-Carballo P, Schwudke D, Waldow F, DiNardo AR. 2020. Perspective for precision medicine for tuberculosis. Front Immunol 11:566608. doi: 10.3389/fimmu.2020.566608. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Goletti D, Lindestam Arlehamn CS, Scriba TJ, Anthony R, Cirillo DM, Alonzi T, Denkinger CM, Cobelens F. 2018. Can we predict tuberculosis cure? Current tools available. Eur Respir J 52:1801089. doi: 10.1183/13993003.01089-2018. [DOI] [PubMed] [Google Scholar]
- 18.Heyckendorf J, Olaru ID, Ruhwald M, Lange C. 2014. Getting personal perspectives on individualized treatment duration in multidrug-resistant and extensively drug-resistant tuberculosis. Am J Respir Crit Care Med 190:374–383. doi: 10.1164/rccm.201402-0363PP. [DOI] [PubMed] [Google Scholar]
- 19.Seddon JA, Hesseling AC, Marais BJ, Jordaan A, Victor T, Schaaf HS. 2012. The evolving epidemic of drug-resistant tuberculosis among children in Cape Town, South Africa. Int J Tuberc Lung Dis 16:928–933. doi: 10.5588/ijtld.11.0679. [DOI] [PubMed] [Google Scholar]
- 20.Yong YK, Tan HY, Saeidi A, Wong WF, Vignesh R, Velu V, Eri R, Larsson M, Shankar EM. 2019. Immune biomarkers for diagnosis and treatment monitoring of tuberculosis: current developments and future prospects. Front Microbiol 10:2789. doi: 10.3389/fmicb.2019.02789. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Heyckendorf J, Marwitz S, Reimann M, Avsar K, DiNardo AR, Günther G, Hoelscher M, Ibraim E, Kalsdorf B, Kaufmann SHE, Kontsevaya I, van Leth F, Mandalakas AM, Maurer FP, Müller M, Nitschkowski D, Olaru ID, Popa C, Rachow A, Rolling T, Rybniker J, Salzer HJF, Sanchez-Carballo P, Schuhmann M, Schaub D, Spinu V, Suárez I, Terhalle E, Unnewehr M, Weiner J, Goldmann T, Lange C. 2021. Prediction of anti-tuberculosis treatment duration based on a 22-gene transcriptomic model. Eur Respir J 58:2003492. doi: 10.1183/13993003.03492-2020. [DOI] [PubMed] [Google Scholar]
- 22.Scriba TJ, Fiore-Gartland A, Penn-Nicholson A, Mulenga H, Kimbung Mbandi S, Borate B, Mendelsohn SC, Hadley K, Hikuam C, Kaskar M, Musvosvi M, Bilek N, Self S, Sumner T, White RG, Erasmus M, Jaxa L, Raphela R, Innes C, Brumskine W, Hiemstra A, Malherbe ST, Hassan-Moosa R, Tameris M, Walzl G, Naidoo K, Churchyard G, Hatherill M, Baepanye K, Baepanye T, Clarke K, Collignon M, Dlamini A, Eyre C, Feni T, Fikizolo M, Galane P, Goliath T, Gangat A, Malefo-Grootboom S, Janse van Rensburg E, Janse van Rensburg B, Kekana S, Zietsman M, Kock A, Kunene I, Lakhi A, Langa N, Ledwaba H, Luphoko M, et al. 2021. Biomarker-guided tuberculosis preventive therapy (CORTIS): a randomised controlled trial. Lancet Infect Dis 21:354–365. doi: 10.1016/S1473-3099(20)30914-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Hoang LT, Jain P, Pillay TD, Tolosa-Wright M, Niazi U, Takwoingi Y, Halliday A, Berrocal-Almanza LC, Deeks JJ, Beverley P, Kon OM, Lalvani A. 2021. Transcriptomic signatures for diagnosing tuberculosis in clinical practice: a prospective, multicentre cohort study. Lancet Infect Dis 21:366–375. doi: 10.1016/S1473-3099(20)30928-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Zak DE, Penn-Nicholson A, Scriba TJ, Thompson E, Suliman S, Amon LM, Mahomed H, Erasmus M, Whatney W, Hussey GD, Abrahams D, Kafaar F, Hawkridge T, Verver S, Hughes EJ, Ota M, Sutherland J, Howe R, Dockrell HM, Boom WH, Thiel B, Ottenhoff THM, Mayanja-Kizza H, Crampin AC, Downing K, Hatherill M, Valvo J, Shankar S, Parida SK, Kaufmann SHE, Walzl G, Aderem A, Hanekom WA. 2016. A blood RNA signature for tuberculosis disease risk: a prospective cohort study. Lancet 387:2312–2322. doi: 10.1016/S0140-6736(15)01316-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Berry MPR, Graham CM, McNab FW, Xu Z, Bloch SAA, Oni T, Wilkinson KA, Banchereau R, Skinner J, Wilkinson RJ, Quinn C, Blankenship D, Dhawan R, Cush JJ, Mejias A, Ramilo O, Kon OM, Pascual V, Banchereau J, Chaussabel D, O’Garra A. 2010. An interferon-inducible neutrophil-driven blood transcriptional signature in human tuberculosis. Nature 466:973–977. doi: 10.1038/nature09247. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Blankley S, Graham CM, Levin J, Turner J, Berry MPR, Bloom CI, Xu Z, Pascual V, Banchereau J, Chaussabel D, Breen R, Santis G, Blankenship DM, Lipman M, O’Garra A. 2016. A 380-gene meta-signature of active tuberculosis compared with healthy controls. Eur Respir J 47:1873–1876. doi: 10.1183/13993003.02121-2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Bloom CI, Graham CM, Berry MPR, Wilkinson KA, Oni T, Rozakeas F, Xu Z, Rossello-Urgell J, Chaussabel D, Banchereau J, Pascual V, Lipman M, Wilkinson RJ, O’Garra A. 2012. Detectable changes in the blood transcriptome are present after two weeks of antituberculosis therapy. PLoS One 7:e46191–13. doi: 10.1371/journal.pone.0046191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Darboe F, Mbandi SK, Naidoo K, Yende-Zuma N, Lewis L, Thompson EG, Duffy FJ, Fisher M, Filander E, van Rooyen M, Bilek N, Mabwe S, McKinnon LR, Chegou N, Loxton A, Walzl G, Tromp G, Padayatchi N, Govender D, Hatherill M, Karim SA, Zak DE, Penn-Nicholson A, Scriba TJ, SATVI Clinical Immunology Team. 2019. Detection of tuberculosis recurrence, diagnosis and treatment response by a blood transcriptomic risk signature in HIV-infected persons on antiretroviral therapy. Front Microbiol 10:1441. doi: 10.3389/fmicb.2019.01441. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Penn-Nicholson A, Mbandi SK, Thompson E, Mendelsohn SC, Suliman S, Chegou NN, Malherbe ST, Darboe F, Erasmus M, Hanekom WA, Bilek N, Fisher M, Kaufmann SHE, Winter J, Murphy M, Wood R, Morrow C, Van Rhijn I, Moody B, Murray M, Andrade BB, Sterling TR, Sutherland J, Naidoo K, Padayatchi N, Walzl G, Hatherill M, Zak D, Scriba TJ, CAPRISA IMPRESS Team. 2020. RISK6, a 6-gene transcriptomic signature of TB disease risk, diagnosis and treatment response. Sci Rep 10:8629. doi: 10.1038/s41598-020-65043-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Odia T, Malherbe ST, Meier S, Maasdorp E, Kleynhans L, Du Plessis N, Loxton AG, Zak DE, Thompson E, Duffy FJ, Kuivaniemi H, Ronacher K, Winter J, Walzl G, Tromp G, Catalysis TB-Biomarker Consortium. 2020. The peripheral blood transcriptome is correlated with PET measures of lung inflammation during successful tuberculosis treatment. Front Immunol 11:596173. doi: 10.3389/fimmu.2020.596173. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Warnat-Herresthal S, Schultze H, Shastry KL, Manamohan S, Mukherjee S, Garg V, Sarveswara R, Händler K, Pickkers P, Aziz NA, Ktena S, Tran F, Bitzer M, Ossowski S, Casadei N, Herr C, Petersheim D, Behrends U, Kern F, Fehlmann T, Schommers P, Lehmann C, Augustin M, Rybniker J, Altmüller J, Mishra N, Bernardes JP, Krämer B, Bonaguro L, Schulte-Schrepping J, De Domenico E, Siever C, Kraut M, Desai M, Monnet B, Saridaki M, Siegel CM, Drews A, Nuesch-Germano M, Theis H, Heyckendorf J, Schreiber S, Kim-Hellmuth S, Balfanz P, Eggermann T, Boor P, Hausmann R, Kuhn H, Isfort S, Stingl JC, COVID-19 Aachen Study (COVAS), et al. 2021. Swarm Learning for decentralized and confidential clinical machine learning. Nature 594:265–270. doi: 10.1038/s41586-021-03583-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Branigan D. 2021. Treatment Action Group (TAG) pipeline report: tuberculosis diagnostics. https://www.treatmentactiongroup.org/wp-content/uploads/2021/11/pipeline_TB_diagnostics_2021_final.pdf.
- 33.Ahmed MIM, Ntinginya NE, Kibiki G, Mtafya BA, Semvua H, Mpagama S, Mtabho C, Saathoff E, Held K, Loose R, Kroidl I, Chachage M, Both UV, Haule A, Mekota A-M, Boeree MJ, Gillespie SH, Hoelscher M, Heinrich N, Geldmacher C. 2018. Phenotypic changes on Mycobacterium tuberculosis-specific CD4 T cells as surrogate markers for tuberculosis treatment efficacy. Front Immunol 9:2247. doi: 10.3389/fimmu.2018.02247. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Musvosvi M, Duffy D, Filander E, Africa H, Mabwe S, Jaxa L, Bilek N, Llibre A, Rouilly V, Hatherill M, Albert M, Scriba TJ, Nemes E. 2018. T-cell biomarkers for diagnosis of tuberculosis: candidate evaluation by a simple whole blood assay for clinical translation. Eur Respir J 51:1800153. doi: 10.1183/13993003.00153-2018. [DOI] [PubMed] [Google Scholar]
- 35.Adekambi T, Ibegbu CC, Cagle S, Kalokhe AS, Wang YF, Hu Y, Day CL, Ray SM, Rengarajan J. 2015. Biomarkers on patient T cells diagnose active tuberculosis and monitor treatment response. J Clin Invest 125:1827–1838. doi: 10.1172/JCI77990. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Estévez O, Anibarro L, Garet E, Martínez A, Pena A, Barcia L, Peleteiro M, González-Fernández Á. 2020. Multi-parameter flow cytometry immunophenotyping distinguishes different stages of tuberculosis infection. J Infect 81:57–71. doi: 10.1016/j.jinf.2020.03.064. [DOI] [PubMed] [Google Scholar]
- 37.Du Bruyn E, Ruzive S, Lindestam Arlehamn CS, Sette A, Sher A, Barber DL, Wilkinson RJ, Riou C. 2021. Mycobacterium tuberculosis-specific CD4 T cells expressing CD153 inversely associate with bacterial load and disease severity in human tuberculosis. Mucosal Immunol 14:491–499. doi: 10.1038/s41385-020-0322-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Parbhoo T, Sampson SL, Mouton JM. 2020. Recent developments in the application of flow cytometry to advance our understanding of Mycobacterium tuberculosis physiology and pathogenesis. Cytometry A 97:683–693. doi: 10.1002/cyto.a.24030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Casanova J-L, Abel L. 2002. Genetic dissection of immunity to mycobacteria: the human model. Annu Rev Immunol 20:581–620. doi: 10.1146/annurev.immunol.20.081501.125851. [DOI] [PubMed] [Google Scholar]
- 40.Flynn JL, Chan J, Triebold KJ, Dalton DK, Stewart TA, Bloom BR. 1993. An essential role for interferon gamma in resistance to Mycobacterium tuberculosis infection. J Exp Med 178:2249–2254. doi: 10.1084/jem.178.6.2249. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Clifford V, He Y, Zufferey C, Connell T, Curtis N. 2015. Interferon gamma release assays for monitoring the response to treatment for tuberculosis: a systematic review. Tuberculosis (Edinb) 95:639–650. doi: 10.1016/j.tube.2015.07.002. [DOI] [PubMed] [Google Scholar]
- 42.Nie W, Wang J, Jing W, Shi W, Wang Q, Huang X, Cai B, Ge Q, Nie L, Han X, Du Y, Wang J, Guo R, Chu N. 2020. Value of serum cytokine biomarkers TNF-α, IL-4, sIL-2R, and IFN-γ for use in monitoring bacterial load and anti-tuberculosis treatment progress. Cytokine X 2:100028. doi: 10.1016/j.cytox.2020.100028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Bai R, Tao L, Li B, Liu A, Dai X, Ji Z, Jian M, Ding Z, Luo L, Chen T, Ma M, Peng Y, Bao F. 2019. Using cytometric bead arrays to detect cytokines in the serum of patients with different types of pulmonary tuberculosis. Int J Immunopathol Pharmacol 33:2058738419845176. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Sigal GB, Segal MR, Mathew A, Jarlsberg L, Wang M, Barbero S, Small N, Haynesworth K, Davis JL, Weiner M, Whitworth WC, Jacobs J, Schorey J, Lewinsohn DM, Nahid P. 2017. Biomarkers of tuberculosis severity and treatment effect: a directed screen of 70 host markers in a randomized clinical trial. EBioMedicine 25:112–121. doi: 10.1016/j.ebiom.2017.10.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Riou C, Perez Peixoto B, Roberts L, Ronacher K, Walzl G, Manca C, Rustomjee R, Mthiyane T, Fallows D, Gray CM, Kaplan G. 2012. Effect of standard tuberculosis treatment on plasma cytokine levels in patients with active pulmonary tuberculosis. PLoS One 7:e36886. doi: 10.1371/journal.pone.0036886. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Wilson D, Mahomed-Yunus SM, Cohen C, Cudahy P, Aldous C, Maartens G. 2018. Evaluation of tuberculosis treatment response with serial C-reactive protein measurements. Open Forum Infect Dis 5:ofy253. doi: 10.1093/ofid/ofy253. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Soedarsono S, Subiantoro MC. 2019. Changes of CRP serum levels in pulmonary TB patients with AFB smear-positive sputum before and two months after receiving anti-tuberculosis drug treatment. Indian J Tuberc 66:134–138. doi: 10.1016/j.ijtb.2018.07.007. [DOI] [PubMed] [Google Scholar]
- 48.Jiang T-T, Shi L-Y, Chen J, Wei L-L, Li M, Hu Y-T, Gan L, Liu C-M, Tu H-H, Li Z-B, Yi W-J, Li J-C. 2018. Screening and identification of potential protein biomarkers for evaluating the efficacy of intensive therapy in pulmonary tuberculosis. Biochem Biophys Res Commun 503:2263–2270. doi: 10.1016/j.bbrc.2018.06.147. [DOI] [PubMed] [Google Scholar]
- 49.Wang C, Wei L-L, Shi L-Y, Pan Z-F, Yu X-M, Li T-Y, Liu C-M, Peng Z-P, Jiang T-T, Chen Z-L, Mao L-G, Li Z-J, Li J-C. 2015. Screening and identification of five serum proteins as novel potential biomarkers for cured pulmonary tuberculosis. Sci Rep 5:15615. doi: 10.1038/srep15615. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Cai Y, Yang Q, Tang Y, Zhang M, Liu H, Zhang G, Deng Q, Huang J, Gao Z, Zhou B, Feng CG, Chen X. 2014. Increased complement C1q level marks active disease in human tuberculosis. PLoS One 9:e92340. doi: 10.1371/journal.pone.0092340. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Ronacher K, Chegou NN, Kleynhans L, Djoba Siawaya JF, Du Plessis N, Loxton AG, Maasdorp E, Tromp G, Kidd M, Stanley K, Kriel M, Menezes A, Gutschmidt A, van der Spuy GD, Warren RM, Dietze R, Okwera A, Thiel B, Belisle JT, Cliff JM, Boom WH, Johnson JL, van Helden PD, Dockrell HM, Walzl G. 2019. Distinct serum biosignatures are associated with different tuberculosis treatment outcomes. Tuberculosis (Edinb) 118:101859. doi: 10.1016/j.tube.2019.101859. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Vrieling F, Alisjahbana B, Sahiratmadja E, van Crevel R, Harms AC, Hankemeier T, Ottenhoff THM, Joosten SA. 2019. Plasma metabolomics in tuberculosis patients with and without concurrent type 2 diabetes at diagnosis and during antibiotic treatment. Sci Rep 9:18669. doi: 10.1038/s41598-019-54983-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Collins JM, Siddiqa A, Jones DP, Liu K, Kempker RR, Nizam A, Shah NS, Ismail N, Ouma SG, Tukvadze N, Li S, Day CL, Rengarajan J, Brust JCM, Gandhi NR, Ernst JD, Blumberg HM, Ziegler TR. 2020. Tryptophan catabolism reflects disease activity in human tuberculosis. JCI Insight 5. doi: 10.1172/jci.insight.137131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Gautam US, Foreman TW, Bucsan AN, Veatch AV, Alvarez X, Adekambi T, Golden NA, Gentry KM, Doyle-Meyers LA, Russell-Lodrigue KE, Didier PJ, Blanchard JL, Kousoulas KG, Lackner AA, Kalman D, Rengarajan J, Khader SA, Kaushal D, Mehra S. 2018. In vivo inhibition of tryptophan catabolism reorganizes the tuberculoma and augments immune-mediated control of Mycobacterium tuberculosis. Proc Natl Acad Sci USA 115:E62–E71. doi: 10.1073/pnas.1711373114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Jaeger S, Karargyris A, Candemir S, Siegelman J, Folio L, Antani S, Thoma G. 2013. Automatic screening for tuberculosis in chest radiographs: a survey. Quant Imaging Med Surg 3:89–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.qure.ai. 2021. Introducing our products. https://qure.ai/. Accessed 24 June 2021.
- 57.WHO. 2021. WHO consolidated guidelines on tuberculosis—module 2: screening. World Health Organization, Geneva, Switzerland. [PubMed] [Google Scholar]
- 58.Murthy SE, Chatterjee F, Crook A, Dawson R, Mendel C, Murphy ME, Murray SR, Nunn AJ, Phillips PPJ, Singh KP, McHugh TD, Gillespie SH, REMoxTB Consortium. 2018. Pretreatment chest x-ray severity and its relation to bacterial burden in smear positive pulmonary tuberculosis. BMC Med 16:73. doi: 10.1186/s12916-018-1053-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Dholakia YN, D’souza DTB, Tolani MP, Chatterjee A, Mistry NF. 2012. Chest X-rays and associated clinical parameters in pulmonary tuberculosis cases from the National Tuberculosis Programme, Mumbai. Infect Dis Rep 4:e10. doi: 10.4081/idr.2012.3438. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Te Riele JB, Buser V, Calligaro G, Esmail A, Theron G, Lesosky M, Dheda K. 2019. Relationship between chest radiographic characteristics, sputum bacterial load, and treatment outcomes in patients with extensively drug-resistant tuberculosis. Int J Infect Dis 79:65–71. doi: 10.1016/j.ijid.2018.10.026. [DOI] [PubMed] [Google Scholar]
- 61.Ralph AP. 2010. A simple, valid, numerical score for grading chest X-ray severity in adult smear-positive pulmonary tuberculosis. Thorax 65:863–869. LP doi: 10.1136/thx.2010.136242. [DOI] [PubMed] [Google Scholar]
- 62.Hesseling AC, Walzl G, Enarson DA, Carroll NM, Duncan K, Lukey PT, Lombard C, Donald PR, Lawrence KA, Gie RP, van Helden PD, Beyers N. 2010. Baseline sputum time to detection predicts month two culture conversion and relapse in non-HIV-infected patients. Int J Tuberc Lung Dis 14:560–570. [PubMed] [Google Scholar]
- 63.Johnson JL, Hadad DJ, Dietze R, Noia Maciel EL, Sewali B, Gitta P, Okwera A, Mugerwa RD, Alcaneses MR, Quelapio MI, Tupasi TE, Horter L, Debanne SM, Eisenach KD, Boom DH. 2009. Shortening treatment in adults with noncavitary tuberculosis and 2-month culture conversion. Am J Respir Crit Care Med 180:558–563. doi: 10.1164/rccm.200904-0536OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Lee JH, Kim O-H, Kim YJ, Shim TS, Jo K-W. 2020. Changes in chest X-ray findings in 1- and 2-month group after treatment initiation for suspected pulmonary tuberculosis. Korean J Intern Med 35:1145–1153. doi: 10.3904/kjim.2019.036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Im JG, Itoh H, Shim YS, Lee JH, Ahn J, Han MC, Noma S. 1993. Pulmonary tuberculosis: CT findings—early active disease and sequential change with antituberculous therapy. Radiology 186:653–660. doi: 10.1148/radiology.186.3.8430169. [DOI] [PubMed] [Google Scholar]
- 66.Malherbe ST, Chen RY, Dupont P, Kant I, Kriel M, Loxton AG, Smith B, Beltran CGG, van Zyl S, McAnda S, Abrahams C, Maasdorp E, Doruyter A, Via LE, Barry CE, Alland D, Richards SG-, Ellman A, Peppard T, Belisle J, Tromp G, Ronacher K, Warwick JM, Winter J, Walzl G. 2020. Quantitative 18F-FDG PET-CT scan characteristics correlate with tuberculosis treatment response. EJNMMI Res 10:8. doi: 10.1186/s13550-020-0591-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Imperial MZ, Nahid P, Phillips PPJ, Davies GR, Fielding K, Hanna D, Hermann D, Wallis RS, Johnson JL, Lienhardt C, Savic RM. 2018. A patient-level pooled analysis of treatment-shortening regimens for drug-susceptible pulmonary tuberculosis. Nat Med 24:1708–1715. doi: 10.1038/s41591-018-0224-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Savic RM, Weiner M, MacKenzie WR, Engle M, Whitworth WC, Johnson JL, Nsubuga P, Nahid P, Nguyen NV, Peloquin CA, Dooley KE, Dorman SE, Tuberculosis Trials Consortium of the Centers for Disease Control and Prevention. 2017. Defining the optimal dose of rifapentine for pulmonary tuberculosis: exposure–response relations from two phase II clinical trials. Clin Pharmacol Ther 102:321–331. doi: 10.1002/cpt.634. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Chang KC, Leung CC, Yew WW, Ho SC, Tam CM. 2004. A nested case–control study on treatment-related risk factors for early relapse of tuberculosis. Am J Respir Crit Care Med 170:1124–1130. doi: 10.1164/rccm.200407-905OC. [DOI] [PubMed] [Google Scholar]
- 70.Skoura E, Zumla A, Bomanji J. 2015. Imaging in tuberculosis. Int J Infect Dis 32:87–93. doi: 10.1016/j.ijid.2014.12.007. [DOI] [PubMed] [Google Scholar]
- 71.Seon HJ, Kim YI, Lim SC, Kim YH, Kwon YS. 2014. Clinical significance of residual lesions in chest computed tomography after anti-tuberculosis treatment. Int J Tuberc Lung Dis 18:341–346. doi: 10.5588/ijtld.13.0565. [DOI] [PubMed] [Google Scholar]
- 72.Eum S-Y, Kong J-H, Hong M-S, Lee Y-J, Kim J-H, Hwang S-H, Cho S-N, Via LE, Barry CE. 2010. Neutrophils are the predominant infected phagocytic cells in the airways of patients with active pulmonary TB. Chest 137:122–128. doi: 10.1378/chest.09-0903. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Demura Y, Tsuchida T, Uesaka D, Umeda Y, Morikawa M, Ameshima S, Ishizaki T, Fujibayashi Y, Okazawa H. 2009. Usefulness of 18F-fluorodeoxyglucose positron emission tomography for diagnosing disease activity and monitoring therapeutic response in patients with pulmonary mycobacteriosis. Eur J Nucl Med Mol Imaging 36:632–639. doi: 10.1007/s00259-008-1009-5. [DOI] [PubMed] [Google Scholar]
- 74.Martinez V, Castilla-Lievre MA, Guillet-Caruba C, Grenier G, Fior R, Desarnaud S, Doucet-Populaire F, Boué F. 2012. 18F-FDG PET/CT in tuberculosis: an early noninvasive marker of therapeutic response. Int J Tuberc Lung Dis 16:1180–1185. doi: 10.5588/ijtld.12.0010. [DOI] [PubMed] [Google Scholar]
- 75.Stelzmueller I, Huber H, Wunn R, Hodolic M, Mandl M, Lamprecht B, Schinko H, Fellner F, Skanjeti A, Giammarile F, Colletti PM, Rubello D, Gabriel M. 2016. 18F-FDG PET/CT in the initial assessment and for follow-up in patients with tuberculosis. Clin Nucl Med 41:e187-94–e194. doi: 10.1097/RLU.0000000000001102. [DOI] [PubMed] [Google Scholar]
- 76.Malherbe ST, Shenai S, Ronacher K, Loxton AG, Dolganov G, Kriel M, Van T, Chen RY, Warwick J, Via LE, Song T, Lee M, Schoolnik G, Tromp G, Alland D, Barry CE, Winter J, Walzl G, Lucas L, Spuy GVD, Stanley K, Thiart L, Smith B, Du Plessis N, Beltran CGG, Maasdorp E, Ellmann A, Choi H, Joh J, Dodd LE, Allwood B, Koegelenberg C, Vorster M, Griffith-Richards S, Catalysis TB–Biomarker Consortium. 2016. Persisting positron emission tomography lesion activity and Mycobacterium tuberculosis mRNA after tuberculosis cure. Nat Med 22:1094–1100. doi: 10.1038/nm.4177. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Choi JY, Jhun BW, Hyun SH, Chung MJ, Koh W-J. 2018. 18F-fluorodeoxyglucose positron emission tomography/computed tomography for assessing treatment response of pulmonary multidrug-resistant tuberculosis. J Clin Med 7:559. doi: 10.3390/jcm7120559. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Chen RY, Dodd LE, Lee M, Paripati P, Hammoud DA, Mountz JM, Jeon D, Zia N, Zahiri H, Coleman MT, Carroll MW, Lee JD, Jeong YJ, Herscovitch P, Lahouar S, Tartakovsky M, Rosenthal A, Somaiyya S, Lee S, Goldfeder LC, Cai Y, Via LE, Park S-K, Cho S-N, Barry CE. 2014. PET/CT imaging correlates with treatment outcome in patients with multidrug-resistant tuberculosis. Sci Transl Med 6:265ra166. doi: 10.1126/scitranslmed.3009501. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Chen RY, Via LE, Dodd LE, Walzl G, Malherbe ST, Loxton AG, Dawson R, Wilkinson RJ, Thienemann F, Tameris M, Hatherill M, Diacon AH, Liu X, Xing J, Jin X, Ma Z, Pan S, Zhang G, Gao Q, Jiang Q, Zhu H, Liang L, Duan H, Song T, Alland D, Tartakovsky M, Rosenthal A, Whalen C, Duvenhage M, Cai Y, Goldfeder LC, Arora K, Smith B, Winter J, Barry Iii CE, Predict TB Study Group. 2017. Using biomarkers to predict TB treatment duration (Predict TB): a prospective, randomized, noninferiority, treatment shortening clinical trial. Gates Open Res 1:9–23. doi: 10.12688/gatesopenres.12750.1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Xie YL, de Jager VR, Chen RY, Dodd LE, Paripati P, Via LE, Follmann D, Wang J, Lumbard K, Lahouar S, Malherbe ST, Andrews J, Yu X, Goldfeder LC, Cai Y, Arora K, Loxton AG, Vanker N, Duvenhage M, Winter J, Song T, Walzl G, Diacon AH, Barry CE. 2021. Fourteen-day PET/CT imaging to monitor drug combination activity in treated individuals with tuberculosis. Sci Transl Med 13:eabd7618. doi: 10.1126/scitranslmed.abd7618. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.WHO. 1996. TB/HIV: a clinical manual/writing team. World Health Organization, Geneva, Switzerland. [Google Scholar]
- 82.Nahid P, Dorman SE, Alipanah N, Barry PM, Brozek JL, Cattamanchi A, Chaisson LH, Chaisson RE, Daley CL, Grzemska M, Higashi JM, Ho CS, Hopewell PC, Keshavjee SA, Lienhardt C, Menzies R, Merrifield C, Narita M, O’Brien R, Peloquin CA, Raftery A, Saukkonen J, Schaaf HS, Sotgiu G, Starke JR, Migliori GB, Vernon A. 2016. Official American Thoracic Society/Centers for Disease Control and Prevention/Infectious Diseases Society of America Clinical Practice Guidelines: treatment of drug-susceptible tuberculosis. Clin Infect Dis 63:e147–e195. doi: 10.1093/cid/ciw376. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Ditah IC, Reacher M, Palmer C, Watson JM, Innes J, Kruijshaar ME, Luma HN, Abubakar I. 2008. Monitoring tuberculosis treatment outcome: analysis of national surveillance data from a clinical perspective. Thorax 63:440–446. doi: 10.1136/thx.2006.073916. [DOI] [PubMed] [Google Scholar]
- 84.Waitt CJ, Squire SB. 2011. A systematic review of risk factors for death in adults during and after tuberculosis treatment. Int J Tuberc Lung Dis 15:871–885. doi: 10.5588/ijtld.10.0352. [DOI] [PubMed] [Google Scholar]
- 85.Wejse C, Gustafson P, Nielsen J, Gomes VF, Aaby P, Andersen PL, Sodemann M. 2008. TBscore: signs and symptoms from tuberculosis patients in a low-resource setting have predictive value and may be used to assess clinical course. Scand J Infect Dis 40:111–120. doi: 10.1080/00365540701558698. [DOI] [PubMed] [Google Scholar]
- 86.Rudolf F, Lemvik G, Abate E, Verkuilen J, Schön T, Gomes VF, Eugen-Olsen J, Østergaard L, Wejse C. 2013. TBscore II: refining and validating a simple clinical score for treatment monitoring of patients with pulmonary tuberculosis. Scand J Infect Dis 45:825–836. doi: 10.3109/00365548.2013.826876. [DOI] [PubMed] [Google Scholar]
- 87.Pefura-Yone EW, Balkissou AD, Poka-Mayap V, Fatime-Abaicho HK, Enono-Edende PT, Kengne AP. 2017. Development and validation of a prognostic score during tuberculosis treatment. BMC Infect Dis 17:251. doi: 10.1186/s12879-017-2309-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Alene KA, Viney K, Gray DJ, McBryde ES, Xu Z, Clements ACA. 2020. Development of a risk score for prediction of poor treatment outcomes among patients with multidrug-resistant tuberculosis. PLoS One 15:e0227100. doi: 10.1371/journal.pone.0227100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Imperial MZ, Phillips PPJ, Nahid P, Savic RM. 2021. Precision-enhancing risk stratification tools for selecting optimal treatment durations in tuberculosis clinical trials. Am J Respir Crit Care Med 204:1086–1096. doi: 10.1164/rccm.202101-0117OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Kloprogge F, Mwandumba HC, Banda G, Kamdolozi M, Shani D, Corbett EL, Kontogianni N, Ward S, Khoo SH, Davies GR, Sloan DJ. 2020. Longitudinal pharmacokinetic-pharmacodynamic biomarkers correlate with treatment outcome in drug-sensitive pulmonary tuberculosis: a population pharmacokinetic-pharmacodynamic analysis. Open Forum Infect Dis 7:ofaa218. doi: 10.1093/ofid/ofaa218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Zheng X, Bao Z, Forsman LD, Hu Y, Ren W, Gao Y, Li X, Hoffner S, Bruchfeld J, Alffenar J-W. 2020. Drug exposure and minimum inhibitory concentration predict pulmonary tuberculosis treatment response. Clin Infect Dis 73:e3520–e3528. doi: 10.1093/cid/ciaa1569. [DOI] [PubMed] [Google Scholar]
- 92.Otalvaro JD, Hernandez AM, Rodriguez CA, Zuluaga AF. 2021. Population pharmacokinetic models of antituberculosis drugs in patients: a systematic critical review. Ther Drug Monit 43:108–115. doi: 10.1097/FTD.0000000000000803. [DOI] [PubMed] [Google Scholar]
- 93.Rockwood N, Du Bruyn E, Morris T, Wilkinson RJ. 2016. Assessment of treatment response in tuberculosis. Expert Rev Respir Med 10:643–654. doi: 10.1586/17476348.2016.1166960. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Theron G, Venter R, Calligaro G, Smith L, Limberis J, Meldau R, Chanda D, Esmail A, Peter J, Dheda K. 2016. Xpert MTB/RIF results in patients with previous tuberculosis: can we distinguish true from false-positive results? Clin Infect Dis 62:995–1001. doi: 10.1093/cid/civ1223. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Nicol MP. 2013. Xpert MTB/RIF: monitoring response to tuberculosis treatment. Lancet Respir Med 1:427–428. doi: 10.1016/S2213-2600(13)70133-4. [DOI] [PubMed] [Google Scholar]
- 96.Honeyborne I, McHugh TD, Phillips PPJ, Bannoo S, Bateson A, Carroll N, Perrin FM, Ronacher K, Wright L, van Helden PD, Walzl G, Gillespie SH. 2011. Molecular bacterial load assay, a culture-free biomarker for rapid and accurate quantification of sputum Mycobacterium tuberculosis bacillary load during treatment. J Clin Microbiol 49:3905–3911. doi: 10.1128/JCM.00547-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Honeyborne I, Mtafya B, Phillips PPJ, Hoelscher M, Ntinginya EN, Kohlenberg A, Rachow A, Rojas-Ponce G, McHugh TD, Heinrich N, Pan African Consortium for the Evaluation of Anti-tuberculosis Antibiotics. 2014. The molecular bacterial load assay replaces solid culture for measuring early bactericidal response to antituberculosis treatment. J Clin Microbiol 52:3064–3067. doi: 10.1128/JCM.01128-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Gillespie SH, Sabiiti W, Oravcova K. 2017. Mycobacterial load assay, p 89–105. In Bishop-Lilly KA (ed), Diagnostic bacteriology: methods and protocols. Springer, New York, NY. doi: 10.1007/978-1-4939-7037-7_5. [DOI] [PubMed] [Google Scholar]
- 99.Mtafya B, Sabiiti W, Sabi I, John J, Sichone E, Ntinginya NE, Gillespie SH. 2019. Molecular bacterial load assay concurs with culture on NaOH-induced loss of Mycobacterium tuberculosis viability. J Clin Microbiol 57:e01992-18. doi: 10.1128/JCM.01992-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Sabiiti W, Azam K, Farmer ECW, Kuchaka D, Mtafya B, Bowness R, Oravcova K, Honeyborne I, Evangelopoulos D, McHugh TD, Khosa C, Rachow A, Heinrich N, Kampira E, Davies G, Bhatt N, Ntinginya EN, Viegas S, Jani I, Kamdolozi M, Mdolo A, Khonga M, Boeree MJ, Phillips PPJ, Sloan D, Hoelscher M, Kibiki G, Gillespie SH. 2020. Tuberculosis bacillary load, an early marker of disease severity: the utility of tuberculosis molecular bacterial load assay. Thorax 75:606–608. doi: 10.1136/thoraxjnl-2019-214238. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Mbelele PM, Mohamed SY, Sauli E, Mpolya EA, Mfinanga SG, Addo KK, Heysell SK, Mpagama SG. 2018. Meta-narrative review of molecular methods for diagnosis and monitoring of multidrug-resistant tuberculosis treatment in adults. Int J Mycobacteriol 7:299–309. doi: 10.4103/ijmy.ijmy_135_18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Hai HT, Vinh DN, Thu DDA, Hanh NT, Phu NH, Srinivasan V, Thwaites GE, Tt Thuong N. 2019. Comparison of the Mycobacterium tuberculosis molecular bacterial load assay, microscopy and GeneXpert versus liquid culture for viable bacterial load quantification before and after starting pulmonary tuberculosis treatment. Tuberculosis (Edinb) 119:101864. doi: 10.1016/j.tube.2019.101864. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Hammond RJH, Baron VO, Oravcova K, Lipworth S, Gillespie SH. 2015. Phenotypic resistance in mycobacteria: is it because I am old or fat that I resist you? J Antimicrob Chemother 70:2823–2827. doi: 10.1093/jac/dkv178. [DOI] [PubMed] [Google Scholar]
- 104.Lipworth S, Hammond RJH, Baron VO, Hu Y, Coates A, Gillespie SH. 2016. Defining dormancy in mycobacterial disease. Tuberculosis (Edinb) 99:131–142. doi: 10.1016/j.tube.2016.05.006. [DOI] [PubMed] [Google Scholar]
- 105.Sabiiti W, Azam K, Esmeraldo E, Bhatt N, Rachow A, Gillespie SH. 2019. Heat inactivation renders sputum safe and preserves Mycobacterium tuberculosis RNA for downstream molecular tests. J Clin Microbiol 57:e01778-18. doi: 10.1128/JCM.01778-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Walter ND, Born SEM, Robertson GT, Reichlen M, Dide-Agossou C, Ektnitphong VA, Rossmassler K, Ramey ME, Bauman AA, Ozols V, Bearrows SC, Schoolnik G, Dolganov G, Garcia B, Musisi E, Worodria W, Huang L, Davis JL, Nguyen NV, Nguyen HV, Nguyen ATV, Phan H, Wilusz C, Podell BK, Sanoussi ND, de Jong BC, Merle CS, Affolabi D, McIlleron H, Garcia-Cremades M, Maidji E, Eshun-Wilson F, Aguilar-Rodriguez B, Karthikeyan D, Mdluli K, Bansbach C, Lenaerts AJ, Savic RM, Nahid P, Vásquez JJ, Voskuil MI. 2021. Mycobacterium tuberculosis precursor rRNA as a measure of treatment-shortening activity of drugs and regimens. Nat Commun 12:2899. doi: 10.1038/s41467-021-22833-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Fukuzawa S, Shiho H, Fujita T. 2019. Selective detection of DNA from viable Mycobacterium tuberculosis complex strains using the EMA-PCR method. Jpn J Infect Dis 72:19–22. doi: 10.7883/yoken.JJID.2018.111. [DOI] [PubMed] [Google Scholar]
- 108.Kim YJ, Lee SM, Park BK, Kim SS, Yi J, Kim HH, Lee EY, Chang CL. 2014. Evaluation of propidium monoazide real-time PCR for early detection of viable Mycobacterium tuberculosis in clinical respiratory specimens. Ann Lab Med 34:203–209. doi: 10.3343/alm.2014.34.3.203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.de Assunção TM, Batista EL, Deves C, Villela AD, Pagnussatti VE, de Oliveira Dias AC, Kritski A, Rodrigues-Junior V, Basso LA, Santos DS. 2014. Real time PCR quantification of viable Mycobacterium tuberculosis from sputum samples treated with propidium monoazide. Tuberculosis (Edinb) 94:421–427. doi: 10.1016/j.tube.2014.04.008. [DOI] [PubMed] [Google Scholar]
- 110.Mbelele PM, Mpolya EA, Sauli E, Mtafya B, Ntingya NE, Addo KK, Kreppel K, Mfinanga S, Phillips PPJ, Gillespie SH, Heysell SK, Sabiti W, Mpagama SG. 2021. Mycobactericidal effects of different regimens measured by molecular bacterial load assay among people treated for multidrug-resistant tuberculosis in Tanzania. J Clin Microbiol 59:e02927-20. doi: 10.1128/JCM.02927-20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Cangelosi GA, Meschke JS. 2014. Dead or alive: molecular assessment of microbial viability. Appl Environ Microbiol 80:5884–5891. doi: 10.1128/AEM.01763-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Kamariza M, Keyser SGL, Utz A, Knapp BD, Ahn G, Cambier CJ, Chen T, Huang KC, Bertozzi CR. 2020. Towards Mycobacterium tuberculosis detection at the point-of-care: a brighter solvatochromic probe permits the detection of mycobacteria within minutes. bioRxiv. https://www.biorxiv.org/content/10.1101/2020.05.29.124008v2. [DOI] [PMC free article] [PubMed]
- 113.Chengalroyen MD, Beukes GM, Gordhan BG, Streicher EM, Churchyard G, Hafner R, Warren R, Otwombe K, Martinson N, Kana BD. 2016. Detection and quantification of differentially culturable tubercle bacteria in sputum from patients with tuberculosis. Am J Respir Crit Care Med 194:1532–1540. doi: 10.1164/rccm.201604-0769OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Wang W-H, Takeuchi R, Jain S-H, Jiang Y-H, Watanuki S, Ohtaki Y, Nakaishi K, Watabe S, Lu P-L, Ito E. 2020. A novel, rapid (within hours) culture-free diagnostic method for detecting live Mycobacterium tuberculosis with high sensitivity. EBioMedicine 60:103007. doi: 10.1016/j.ebiom.2020.103007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Sakashita K, Takeuchi R, Takeda K, Takamori M, Ito K, et al. 2020. Ultrasensitive enzyme-linked immunosorbent assay for the detection of MPT64 secretory antigen to evaluate Mycobacterium tuberculosis viability in sputum. Int J Infect Dis 96:244–253. doi: 10.1016/j.ijid.2020.04.059. [DOI] [PubMed] [Google Scholar]
- 116.Sanoussi CN, de Jong BC, Odoun M, Arekpa K, Ali Ligali M, Bodi O, Harris S, Ofori-Anyinam B, Yeboah-Manu D, Otchere ID, Asante-Poku A, Anagonou S, Gagneux S, Coscolla M, Rigouts L, Affolabi D. 2018. Low sensitivity of the MPT64 identification test to detect lineage 5 of the Mycobacterium tuberculosis complex. J Med Microbiol 67:1718–1727. doi: 10.1099/jmm.0.000846. [DOI] [PubMed] [Google Scholar]
- 117.Singh K, Kumari R, Tripathi R, Gupta A, Anapurba S. 2019. Mutation in MPT64 gene influencing diagnostic accuracy of SD Bioline assay (capilia). BMC Infect Dis 19:1048. doi: 10.1186/s12879-019-4671-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Hamasur B, Bruchfeld J, Haile M, Pawlowski A, Bjorvatn B, Källenius G, Svenson SB. 2001. Rapid diagnosis of tuberculosis by detection of mycobacterial lipoarabinomannan in urine. J Microbiol Methods 45:41–52. doi: 10.1016/S0167-7012(01)00239-1. [DOI] [PubMed] [Google Scholar]
- 119.Sigal GB, Pinter A, Lowary TL, Kawasaki M, Li A, et al. 2018. A novel sensitive immunoassay targeting the 5-methylthio-d-xylofuranose-lipoarabinomannan epitope meets the WHO’s performance target for tuberculosis diagnosis. J Clin Microbiol 56:e01338-18. doi: 10.1128/JCM.01338-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Drain PK, Gounder L, Grobler A, Sahid F, Bassett IV, Moosa M-YS. 2015. Urine lipoarabinomannan to monitor antituberculosis therapy response and predict mortality in an HIV-endemic region: a prospective cohort study. BMJ Open 5:e006833. doi: 10.1136/bmjopen-2014-006833. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Kawasaki M, Echiverri C, Raymond L, Cadena E, Reside E, Gler MT, Oda T, Ito R, Higashiyama R, Katsuragi K, Liu Y. 2019. Lipoarabinomannan in sputum to detect bacterial load and treatment response in patients with pulmonary tuberculosis: analytic validation and evaluation in two cohorts. PLoS Med 16:e1002780–20. doi: 10.1371/journal.pmed.1002780. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Bulterys MA, Wagner B, Redard-Jacot M, Suresh A, Moreau E, Denkinger CM, Drain PK, Broger T. 2019. Point-of-care urine LAM tests for tuberculosis diagnosis: a status update. J Clin Med 9:111. doi: 10.3390/jcm9010111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Broger T, Nicol MP, Sigal GB, Gotuzzo E, Zimmer AJ, Surtie S, Caceres-Nakiche T, Mantsoki A, Reipold EI, Székely R, Tsionsky M, van Heerden J, Plisova T, Chikamatsu K, Lowary TL, Pinter A, Mitarai S, Moreau E, Schumacher SG, Denkinger CM. 2020. Diagnostic accuracy of 3 urine lipoarabinomannan tuberculosis assays in HIV-negative outpatients. J Clin Invest 130:5756–5764. doi: 10.1172/JCI140461. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Broger T, Nicol MP, Székely R, Bjerrum S, Sossen B, Schutz C, Opintan JA, Johansen IS, Mitarai S, Chikamatsu K, Kerkhoff AD, Macé A, Ongarello S, Meintjes G, Denkinger CM, Schumacher SG. 2020. Diagnostic accuracy of a novel tuberculosis point-of-care urine lipoarabinomannan assay for people living with HIV: a meta-analysis of individual in- and outpatient data. PLoS Med 17:e1003113-16. doi: 10.1371/journal.pmed.1003113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Peter JG, Theron G, van Zyl-Smit R, Haripersad A, Mottay L, Kraus S, Binder A, Meldau R, Hardy A, Dheda K. 2012. Diagnostic accuracy of a urine lipoarabinomannan strip-test for TB detection in HIV-infected hospitalized patients. Eur Respir J 40:1211–1220. doi: 10.1183/09031936.00201711. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Shah M, Hanrahan C, Wang ZY, Dendukuri N, Lawn SD, Denkinger CM, Steingart KR. 2016. Lateral flow urine lipoarabinomannan assay for detecting active tuberculosis in HIV-positive adults. Cochrane Database Syst Rev 2016:CD011420. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Minion J, Leung E, Talbot E, Dheda K, Pai M, Menzies D. 2011. Diagnosing tuberculosis with urine lipoarabinomannan: systematic review and meta-analysis. Eur Respir J 38:1398–1405. doi: 10.1183/09031936.00025711. [DOI] [PubMed] [Google Scholar]
- 128.Supply P, Warren RM, Bañuls A-L, Lesjean S, Van Der Spuy GD, Lewis L-A, Tibayrenc M, Van Helden PD, Locht C. 2003. Linkage disequilibrium between minisatellite loci supports clonal evolution of Mycobacterium tuberculosis in a high tuberculosis incidence area. Mol Microbiol 47:529–538. doi: 10.1046/j.1365-2958.2003.03315.x. [DOI] [PubMed] [Google Scholar]
- 129.Varghese B, Enani M, Alrajhi A, Al Johani S, Albarak A, Althawadi S, Elkhizzi N, et al. 2018. Impact of Mycobacterium tuberculosis complex lineages as a determinant of disease phenotypes from an immigrant rich moderate tuberculosis burden country. Respir Res 19:259. doi: 10.1186/s12931-018-0966-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Yimer SA, Kalayou S, Homberset H, Birhanu AG, Riaz T, Zegeye ED, Lutter T, Abebe M, Holm-Hansen C, Aseffa A, Tønjum T. 2020. Lineage-specific proteomic signatures in the Mycobacterium tuberculosis complex reveal differential abundance of proteins involved in virulence, DNA repair, CRISPR-Cas, bioenergetics and lipid metabolism. Front Microbiol 11:550760. doi: 10.3389/fmicb.2020.550760. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Nguyen VAT, Bañuls A-L, Tran THT, Pham KLT, Nguyen TS, Nguyen HV, Nguyen NLT, Nguyen NLT, Dang DA, Marks GB, Choisy M. 2016. Mycobacterium tuberculosis lineages and anti-tuberculosis drug resistance in reference hospitals across Viet Nam. BMC Microbiol 16:167. doi: 10.1186/s12866-016-0784-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Tientcheu LD, Haks MC, Agbla SC, Sutherland JS, Adetifa IM, Donkor S, Quinten E, Daramy M, Antonio M, Kampmann B, Ottenhoff THM, Dockrell HM, Ota MO. 2016. Host immune responses differ between M. africanum- and M. tuberculosis-infected patients following standard anti-tuberculosis treatment. PLoS Negl Trop Dis 10:e0004701. doi: 10.1371/journal.pntd.0004701. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Yimer SA, Norheim G, Namouchi A, Zegeye ED, Kinander W, Tønjum T, Bekele S, Mannsåker T, Bjune G, Aseffa A, Holm-Hansen C. 2015. Mycobacterium tuberculosis lineage 7 strains are associated with prolonged patient delay in seeking treatment for pulmonary tuberculosis in Amhara Region, Ethiopia. J Clin Microbiol 53:1301–1309. doi: 10.1128/JCM.03566-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Feuerriegel S, Schleusener V, Beckert P, Kohl TA, Miotto P, Cirillo DM, Cabibbe AM, Niemann S, Fellenberg K. 2015. PhyResSE: a web tool delineating Mycobacterium tuberculosis antibiotic resistance and lineage from whole-genome sequencing data. J Clin Microbiol 53:1908–1914. doi: 10.1128/JCM.00025-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Schön T, Werngren J, Machado D, Borroni E, Wikjander M, et al. 2020. Antimicrobial susceptibility testing of Mycobacterium tuberculosis complex isolates: the EUCAST broth microdilution reference method for MIC determination. Clin Microbiol Infect 26:1488–1492. doi: 10.1016/j.cmi.2020.07.036. [DOI] [PubMed] [Google Scholar]
- 136.Tagliani E, Anthony R, Kohl TA, de Neeling A, Nikolayevskyy V, Kodmon C, et al. 2021. Use of a whole-genome sequencing-based approach for Mycobacterium tuberculosis surveillance in Europe in 2017–2019: an ECDC pilot study. Eur Respir J 57:2002272. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Soundararajan L, Kambli P, Priyadarshini S, Let B, Murugan S, Iravatham C, Tornheim JA, Rodrigues C, Gupta R, Ramprasad VL. 2020. Whole genome enrichment approach for rapid detection of Mycobacterium tuberculosis and drug resistance-associated mutations from direct sputum sequencing. Tuberculosis (Edinb) 121:101915. doi: 10.1016/j.tube.2020.101915. [DOI] [PubMed] [Google Scholar]
- 138.Colman RE, Anderson J, Lemmer D, Lehmkuhl E, Georghiou SB, Heaton H, Wiggins K, Gillece JD, Schupp JM, Catanzaro DG, Crudu V, Cohen T, Rodwell TC, Engelthaler DM. 2016. Rapid drug susceptibility testing of drug-resistant Mycobacterium tuberculosis isolates directly from clinical samples by use of amplicon sequencing: a proof-of-concept study. J Clin Microbiol 54:2058–2067. doi: 10.1128/JCM.00535-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Colangeli R, Jedrey H, Kim S, Connell R, Ma S, Chippada Venkata UD, Chakravorty S, Gupta A, Sizemore EE, Diem L, Sherman DR, Okwera A, Dietze R, Boom WH, Johnson JL, Mac Kenzie WR, Alland D. 2018. Bacterial factors that predict relapse after tuberculosis therapy. N Engl J Med 379:823–833. doi: 10.1056/NEJMoa1715849. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Cabibbe AM, Spitaleri A, Battaglia S, Colman RE, Suresh A, Uplekar S, Rodwell TC, Cirillo DM. 2020. Application of targeted next-generation sequencing assay on a portable sequencing platform for culture-free detection of drug-resistant tuberculosis from clinical samples. J Clin Microbiol 58:e00632-20. doi: 10.1128/JCM.00632-20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Chen D, Bryden WA, Wood R. 2020. Detection of tuberculosis by the analysis of exhaled breath particles with high resolution. Sci Rep 10:7647. doi: 10.1038/s41598-020-64637-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Coronel Teixeira R, Rodríguez M, Jiménez de Romero N, Bruins M, Gómez R, Yntema JB, Chaparro Abente G, Gerritsen JW, Wiegerinck W, Pérez Bejerano D, Magis-Escurra C. 2017. The potential of a portable, point-of-care electronic nose to diagnose tuberculosis. J Infect 75:441–447. doi: 10.1016/j.jinf.2017.08.003. [DOI] [PubMed] [Google Scholar]
- 143.McNerney R, Wondafrash BA, Amena K, Tesfaye A, McCash EM, Murray NJ. 2010. Field test of a novel detection device for Mycobacterium tuberculosis antigen in cough. BMC Infect Dis 10:161. doi: 10.1186/1471-2334-10-161. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Sahota AS, Gowda R, Arasaradnam RP, Daulton E, Savage RS, Skinner JR, Adams E, Ward SA, Covington JA. 2016. A simple breath test for tuberculosis using ion mobility: a pilot study. Tuberculosis (Edinb) 99:143–146. doi: 10.1016/j.tube.2016.05.005. [DOI] [PubMed] [Google Scholar]
- 145.Zetola NM, Modongo C, Matsiri O, Tamuhla T, Mbongwe B, Matlhagela K, Sepako E, Catini A, Sirugo G, Martinelli E, Paolesse R, Di Natale C. 2017. Diagnosis of pulmonary tuberculosis and assessment of treatment response through analyses of volatile compound patterns in exhaled breath samples. J Infect 74:367–376. doi: 10.1016/j.jinf.2016.12.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Saktiawati AMI, Putera DD, Setyawan A, Mahendradhata Y, van der Werf TS. 2019. Diagnosis of tuberculosis through breath test: a systematic review. EBioMedicine 46:202–214. doi: 10.1016/j.ebiom.2019.07.056. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Saktiawati AMI, et al. 2019. Sensitivity and specificity of an electronic nose in diagnosing pulmonary tuberculosis among patients with suspected tuberculosis. PLoS One 14:e0217963. doi: 10.1371/journal.pone.0217963. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Lakhani P, Sundaram B. 2017. Deep learning at chest radiography: automated classification of pulmonary tuberculosis by using convolutional neural networks. Radiology 284:574–582. doi: 10.1148/radiol.2017162326. [DOI] [PubMed] [Google Scholar]
- 149.Ruhwald M, Carmona S, Pai M. 2021. Learning from COVID-19 to reimagine tuberculosis diagnosis. Lancet Microbe 2:e169–e170. doi: 10.1016/S2666-5247(21)00057-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Zimmer AJ, Schumacher SG, Södersten E, Mantsoki A, Wyss R, Persing DH, Banderby S, Strömqvist Meuzelaar L, Prieto J, Gnanashanmugam D, Khatri P, Ongarello S, Ruhwald M, Denkinger CM. 2021. A novel blood-based assay for treatment monitoring of tuberculosis. BMC Res Notes 14:247. doi: 10.1186/s13104-021-05663-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Thompson EG, Du Y, Malherbe ST, Shankar S, Braun J, Valvo J, Ronacher K, Tromp G, Tabb DL, Alland D, Shenai S, Via LE, Warwick J, Aderem A, Scriba TJ, Winter J, Walzl G, Zak DE, Catalysis TB–Biomarker Consortium. 2017. Host blood RNA signatures predict the outcome of tuberculosis treatment. Tuberculosis (Edinb) 107:48–58. doi: 10.1016/j.tube.2017.08.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Sweeney TE, Braviak L, Tato CM, Khatri P. 2016. Genome-wide expression for diagnosis of pulmonary tuberculosis: a multicohort analysis. Lancet Respir Med 4:213–224. doi: 10.1016/S2213-2600(16)00048-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Cliff JM, Lee J-S, Constantinou N, Cho J-E, Clark TG, Ronacher K, King EC, Lukey PT, Duncan K, Van Helden PD, Walzl G, Dockrell HM. 2013. Distinct phases of blood gene expression pattern through tuberculosis treatment reflect modulation of the humoral immune response. J Infect Dis 207:18–29. doi: 10.1093/infdis/jis499. [DOI] [PubMed] [Google Scholar]
- 154.Ahmed MIM, Ziegler C, Held K, Dubinski I, Ley-Zaporozhan J, Geldmacher C, von Both U. 2019. The TAM-TB Assay: a promising TB immune-diagnostic test with a potential for treatment monitoring. Front Pediatr 7:27. doi: 10.3389/fped.2019.00027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Qin ZZ, Sander MS, Rai B, Titahong CN, Sudrungrot S, Laah SN, Adhikari LM, Carter EJ, Puri L, Codlin AJ, Creswell J. 2019. Using artificial intelligence to read chest radiographs for tuberculosis detection: a multi-site evaluation of the diagnostic accuracy of three deep learning systems. Sci Rep 9:15000. doi: 10.1038/s41598-019-51503-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Engle E, Gabrielian A, Long A, Hurt DE, Rosenthal A. 2020. Performance of Qure.ai automatic classifiers against a large annotated database of patients with diverse forms of tuberculosis. PLoS One 15:e0224445. doi: 10.1371/journal.pone.0224445. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Heyckendorf J, van Leth F, Avsar K, Glattki G, Günther G, Kalsdorf B, Müller M, Olaru ID, Rolling T, Salzer HJF, Schuhmann M, Terhalle E, Lange C. 2018. Treatment responses in multidrug-resistant tuberculosis in Germany. Int J Tuberc Lung Dis 22:399–406. doi: 10.5588/ijtld.17.0741. [DOI] [PubMed] [Google Scholar]
- 158.Ahmad N, Ahuja SD, Akkerman OW, Alffenaar J-WC, Anderson LF, Baghaei P, Bang D, Barry PM, Bastos ML, Behera D, Benedetti A, Bisson GP, Boeree MJ, Bonnet M, Brode SK, Brust JCM, Cai Y, Caumes E, Cegielski JP, Centis R, Chan P-C, Chan ED, Chang K-C, Charles M, Cirule A, Dalcolmo MP, D’Ambrosio L, de Vries G, Dheda K, Esmail A, Flood J, Fox GJ, Fréchet-Jachym M, Fregona G, Gayoso R, Gegia M, Gler MT, Gu S, Guglielmetti L, Holtz TH, Hughes J, Isaakidis P, Jarlsberg L, Kempker RR, Keshavjee S, Khan FA, Kipiani M, Koenig SP, Koh W-J, Kritski A, Collaborative Group for the Meta-Analysis of Individual Patient Data in MDR-TB treatment–2017, et al. 2018. Treatment correlates of successful outcomes in pulmonary multidrug-resistant tuberculosis: an individual patient data meta-analysis. Lancet 392:821–834. doi: 10.1016/S0140-6736(18)31644-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Kurbatova EV, Gammino VM, Bayona J, Becerra MC, Danilovitz M, Falzon D, Gelmanova I, Keshavjee S, Leimane V, Mitnick CD, Quelapio MI, Riekstina V, Taylor A, Viiklepp P, Zignol M, Cegielski JP. 2012. Predictors of sputum culture conversion among patients treated for multidrug-resistant tuberculosis. Int J Tuberc Lung Dis 16:1335–1343. doi: 10.5588/ijtld.11.0811. [DOI] [PubMed] [Google Scholar]
- 160.Diacon AH, Maritz JS, Venter A, van Helden PD, Andries K, McNeeley DF, Donald PR. 2010. Time to detection of the growth of Mycobacterium tuberculosis in MGIT 960 for determining the early bactericidal activity of antituberculosis agents. Eur J Clin Microbiol Infect Dis 29:1561–1565. doi: 10.1007/s10096-010-1043-7. [DOI] [PubMed] [Google Scholar]
- 161.Friedrich SO, Rachow A, Saathoff E, Singh K, Mangu CD, Dawson R, Phillips PP, Venter A, Bateson A, Boehme CC, Heinrich N, Hunt RD, Boeree MJ, Zumla A, McHugh TD, Gillespie SH, Diacon AH, Hoelscher M. 2013. Assessment of the sensitivity and specificity of Xpert MTB/RIF assay as an early sputum biomarker of response to tuberculosis treatment. Lancet Respir Med 1:462–470. doi: 10.1016/S2213-2600(13)70119-X. [DOI] [PubMed] [Google Scholar]
- 162.Jayakumar A, et al. 2016. Xpert MTB/RIF Assay shows faster clearance of Mycobacterium tuberculosis DNA with higher levels of rifapentine exposure. J Clin Microbiol 54:12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Shenai S, Ronacher K, Malherbe S, Stanley K, Kriel M, Winter J, Peppard T, Barry CE, Wang J, Dodd LE, Via LE, Barry CE, Walzl G, Alland D. 2016. Bacterial loads measured by the Xpert MTB/RIF Assay as markers of culture conversion and bacteriological cure in pulmonary TB. PLoS One 11:e0160062. doi: 10.1371/journal.pone.0160062. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Feuerriegel S, Kohl TA, Utpatel C, Andres S, Maurer FP, Heyckendorf J, Jouet A, Badalato N, Foray L, Fouad Kamara R, Conteh OS, Supply P, Niemann S. 2021. Rapid genomic first- and second-line drug resistance prediction from clinical Mycobacterium tuberculosis specimens using Deeplex-MycTB. Eur Respir J 57:2001796. doi: 10.1183/13993003.01796-2020. [DOI] [PubMed] [Google Scholar]
- 165.Cohen KA, Manson AL, Desjardins CA, Abeel T, Earl AM. 2019. Deciphering drug resistance in Mycobacterium tuberculosis using whole-genome sequencing: progress, promise, and challenges. Genome Med 11:45. doi: 10.1186/s13073-019-0660-8. [DOI] [PMC free article] [PubMed] [Google Scholar]