Abstract
Tuberculosis is once again the most fatal global infectious disease and has killed many more humans than any other pathogen. Despite the identification of Mycobacterium tuberculosis (Mtb) over 140 years ago, we have yet to control the epidemic. A central issue is the complexity of the host-pathogen interaction, with diverse immunological routes leading to tuberculosis disease. This intricate relationship results from the prolonged co-evolution of the pathogen with humans, potentially due to Mtb exposure giving a survival advantage. Emerging methodologies, such as single cell and spatial transcriptomics, offer a golden opportunity to understand the immunology unpinning this host-pathogen interaction at unprecedented resolution. However, these analyses will be fundamentally flawed if they do not consider the intricacies of human Mtb infection. Here, we propose that attempts to find single unifying immunological mechanisms of tuberculosis are hindering progress, and we must embrace the complexity of multiple paths to disease to allow the systems biology era to deliver transformative solutions.
Introduction
The study of tuberculosis (TB) pioneered infectious disease research in the modern scientific era, leading to the formulation of Koch’s postulates demonstrating that an illness can have an infectious origin (1). Mycobacterium tuberculosis (Mtb) infects and survives within macrophages, subverting the host immune response by multiple mechanisms including inhibition of phagosome maturation and downregulation of antigen presenting molecules, leading to the formation of complex immune aggregates, known as granulomas (2). Even though TB was the first definitively identified infection, it remains the world’s deadliest infectious disease despite well over 100 years of research (3). This contrast raises a critical question: why is Mtb proving so resistant to human efforts to control it?
Mtb has eluded attempts to develop a fully protective vaccine, despite a partially effective vaccine being available since the 1920’s. Mycobacterium bovis BCG was developed by sequential culture of M bovis and protects children against disseminated TB but has limited protection against adult disease (4, 5). Although exciting progress has been made with a vaccine that reduces TB disease by approximately 50% in a phase II study (6), currently the immune mechanisms underpinning protection have not been identified. Subsequently, a major trial of an alternative vaccine showed no efficacy in preventing recurrence. Indeed, despite being highly immunogenic, the relapse rate tended to be higher in the vaccine group (7). Similarly, repeat BCG vaccination does not increase protection, despite inducing a strong Mtb-specific CD4 T cell response (8).
These difficulties highlight the urgent need to understand the host-pathogen interaction more fully. We have insufficient knowledge of key steps in disease progression to develop transformative interventions. Currently, almost all investigations into the immune response to Mtb are based on the premise that a unifying disease mechanism exists, and this can be understood through reductionist scientific approaches. However, clinical observations demonstrate that there are multiple paths to TB, and so seeking to define a single mechanism is likely to be flawed.
Immunological insights from historic and recent clinical observations
TB has co-evolved with humans for millennia, with some estimates suggesting up to 70,000 years (9). The field of paleoarchaeology, for example, provides extensive evidence of TB from the early neolithic period in the Middle East, with approximately 5% of skeletons from a 10,000-year-old village showing signs of TB (10). This was just before animal domestication and pottery, in hunter-gatherers who built stone houses, and so Mtb was already successfully transmitting in humans before the subsequent population growth that occurred with farming (10). Potentially, to survive in relatively small hunter-gatherer communities Mtb may have needed to have reduced virulence to avoid excessive deaths and a latent period to permit sustainable transmission in low population numbers (11).
Mtb successfully persisted over the ages and then flourished in the more dense populations that occurred with the industrial revolution (12), giving rise to the modern TB era of high transmission approximately 250 years ago. The fundamental cause of TB remained unknown until Koch’s seminal work (1). Early investigations demonstrated that the first Mtb infection point was the lung base, while Mtb exits from the apices of the lungs (13). This life cycle must involve several distinct host/pathogen interactions, as initially immune evasion is required for Mtb to survive, but then later immune engagement is necessary to cause the inflammation and lung destruction needed to optimise transmission (14). Notably, most people (approximately 90%) initially infected with Mtb never progress to active, clinical disease (3). In addition, in the pre-antibiotic era, the progression and regression of different lesions in the same individual were observed on chest radiographs, and one third of patients with active TB disease self-healed, showing that the host-pathogen interaction is finely balanced at all stages of infection (12).
Modern immunological techniques and the development of biologic therapies that target specific immune processes have provided extensive insight into TB disease mechanisms. The greatly increased occurrence of TB in the context of HIV co-infection, for example, highlighted immunodeficiency as a major driver of disease (15). Similarly, the occurrence of TB after anti-TNF-α therapy for autoimmune disease confirmed the importance of TNF-α in control of latent infection (16). Furthermore, genetic investigations have identified numerous immunodeficiencies via studies of Mendelian Susceptibility to Mycobacterial Diseases (MSMD), with mutations typically along the IL-12/IFN-γ/STAT signalling pathway (17–19). With less clearly defined immunologic mechanisms, malnutrition is a significant risk factor for TB (20), and food supplementation reduces TB incidence in contacts (21). Therefore, diverse immune deficiencies can lead to active TB.
The vast majority of patients who develop TB, however, have no clear identifiable immunodeficiency. Indeed, Comstock’s seminal study from the 1970s showed that children from Haiti with the strongest recall responses to Mtb antigens actually had the greatest subsequent risk of developing TB (22). These observations have been replicated in more recent studies using IFN-γ release assays (IGRAs) in response to TB antigens, where higher IFN-γ production associates with increased risk of progressing to disease in both children and adults (23, 24). TB is commonest in young adults in their immunological prime, and more frequent in males than females, characterised by an excessive inflammatory response (25). The implication that TB can also be caused by immune excess is now supported by recently introduced cancer immunotherapies (26). Anti-PD-1 treatment, which activates the immune response and represents the immunological opposite to anti-TNF therapy, should control TB if immunodeficiency were the critical component. However, anti-PD-1 treatment can lead to rapid reactivation of latent TB infection, first identified in case reports (27). This finding is supported by studies in mice (28, 29), the non-human primate (30) and 3D cellular models (31), and ultimately has been validated by patient registry studies (32, 33). Similarly, type II diabetes is associated with an increased risk of TB (20), characterized by a hyper-inflammatory immune response (34). Therefore, diverse clinical evidence demonstrates that there are multiple immunological routes to TB disease (35) (Figure 1).

Many routes can lead to TB via opposing immunological extremes.
Generally, these can be viewed as immune deficiencies or immune excess, but the majority of patients who develop TB are relatively young with a competent immune response. Though not quantitative, font size relates to contribution to global incidence. Left arrow: Ghon focus at lung base; Right arrow: cavity at lung apex.
Even when TB develops in the face of a “normal” immune system, there are likely to be many different subgroups that have not yet been identified due to limitations in standard immunological profiling. A quarter of the world’s population are thought to be exposed to Mtb (36), and so the vast majority of those infected with Mtb remain healthy lifelong (3), while those who progress to active TB likely represent distinct outlier populations. The diverse causes of active TB, such as anti-TNF, MSMD, HIV, diabetes and anti-PD1, demonstrate that patients do not reach TB by a single route, but instead can lose the immunological balance that controls Mtb by multiple paths. This may explain why genetic studies have generally failed to find consistent predispositions. Evidence of heritability can be demonstrated but is often hard to validate in different populations (37). Mutations in the macrophage endosomal protein NRAMP1, for example, were shown to associate with TB in an early seminal study (38), but since then few consistent traits of genetic susceptibility to TB have been identified (39). Similarly, studies of individuals who resist Mtb infection despite recurrent exposure generates a diverse list of potentially protective features, including T cell subsets or activation (40–42), TNF-α responses (43), antibody activity (44) and innate immune training (45). These human observations are supported by murine genetic experiments, which show that increased susceptibility to Mtb can result from a wide range of immunological alterations (46). However, despite this evidence of diversity, the TB field continues to seek a single unifying explanation for disease progression, which is incompatible with clinical observations.
Ultimately, to transmit efficiently, Mtb needs to cause pulmonary disease to then spread by airborne droplets (47), and for Mtb it does not matter the route taken, as long as it ends at pulmonary TB. Experimentally, TB is a fundamentally challenging disease to model, as the interaction is prolonged and Mtb is an obligate human pathogen (48). The primary driver of advances in immunology in the last decades has been transgenic mice (49), but the mouse model of TB does not accurately reflect human disease (50). Disease heterogeneity has been highlighted in describing clinical TB endotypes observed during active disease (51), but less attention has been given to the different immunological pathways that can lead to the same disease phenotype.
Mtb’s single successful colonisation of humans
Just as new tools are providing insights into the complexity of human TB progression, advances in mycobacterial genomics are highlighting the unique nature of the human-Mtb relationship (52). Mtb is a near clonal organism, with evidence suggesting that there has been only one successful and sustained penetration into the human population (52, 53). The entire spectrum of Mtb strains globally only differs by a total of approximately 2,000 SNPs (52). This genetic conservation has persisted even though Mtb has been flourishing in human populations since the industrial revolution, suggesting that Mtb was already close to being optimised for human hosts. Further evolution may have been to increase transmission within specific populations as humans diverged genetically (53).
A very similar organism, M canetti, can cause disease but cannot transmit from human to human (54). Similarly, M bovis is 99.9% identical to Mtb (55), but has never achieved sustained human-to-human transmission, despite many millions of human exposures during the pre-pasteurisation era and common lymph node infections (53). Therefore, human TB is caused exclusively by Mtb, unlike other very closely related mycobacteria, which maintain infection cycles in other higher mammals but not humans (53). Clues to the key pathogenic mechanisms may lie in the differences between mycobacterial species, but ultimately similarities between Mtb strains must be critical to their ongoing success. For example, the hyper conservation of T cell epitopes may be evidence that Mtb manipulates the host immune response to favour disease and transmission (56). Given the size of the Mtb genome, identifying the critical conserved features will be challenging, especially as half of its genes still lack a known function 25 years after the genome was first sequenced (57).
One highly intriguing proposition is that latent TB may itself give humans an evolutionary advantage (57). Exposure to Mtb modifies innate immune training (58), and so a mechanism whereby Mtb could protect from other fatal infections is plausible. Similarly, M bovis BCG, a live-attenuated strain of M bovis, causes innate immune training (59), suggesting this effect is common to Mtb and BCG. BCG vaccination reduces mortality much more significantly than can be explained by the effect on TB incidence alone (60), with a protective effect confirmed in diverse studies (61). For example, BCG reduces viral infections in infants in Africa (62) and experimentally challenged adults (63), and associates with improves survival in Europe (64). Together, these observations strongly imply that mycobacterial infection protects from other infectious causes of death.
Mtb almost certainly first colonised humans in East Africa (53), potentially about 70,000 years ago (9). Several human migrations out of Africa are thought to have preceded this date, but all ultimately became extinct, with the first sustained human dispersal 60 – 70,000 years ago (65, 66). The primary selective pressure in these early communities would have been infectious disease (67). This raises a novel hypothesis that a survival advantage for the first successful human migrants out of Africa was Mtb circulating in the community, reducing mortality from other infectious diseases and thereby enabling sustainable population growth.
Mtb transmission may have benefitted humans by increasing innate immune resistance to infection at the cost of 10% disease penetrance that permits Mtb propagation. This selective pressure over many millennia would progressively remove genotypes that lead to high susceptibility to TB, but equally tend to select against individuals with complete resistance to Mtb colonisation (Figure 2). This could explain why consistent genetic traits for susceptibility or resistance to TB have been hard to identify (39). More recent mass infection events, such as the smallpox epidemics that killed approximately 25% of the population (67), may have further favoured individuals immunologically trained by Mtb. Likewise, successive waves of plague killed approximately 25% of the population, at the end of the Roman empire and then in the European middle ages, adding to selective pressure from endemic infections (68, 69). If humans have been selected to be permissive to Mtb colonisation but broadly resistant to disease, it suggests the development of active disease must be a relatively unusual event in a subset of outlier individuals. In some sense, we could be regarded as having a symbiotic relationship with Mtb, with disease representing a necessary evil, caused by an imbalance in the predominantly stable host pathogen interaction (70, 71).

Selection pressure of prolonged co-evolution favours individuals permissive to colonisation but resistant to active disease.
Over millennia, Mtb circulation in society will remove genetic traits resulting in high susceptibility to active TB infection. Perhaps less intuitively, if Mtb generates trained immunity that protects against other fatal infections, individuals highly resistant to Mtb colonisation will also be selected against. The resulting population would then reflect modern humans; susceptible to initial Mtb infection but generally resistant to TB disease.
The host-pathogen interaction at a cellular level
The early histological era described the wide range of human TB lesions and granuloma types, and identified TB as a disease characterised by spatial organisation (72). Classically, the granuloma has been proposed to be where the outcome of the host-pathogen interaction is determined (73). Recent methodological advances are permitting much greater dissection of events and further highlight the importance of spatial organisation within the granuloma (74–76). However, just as the early X-ray era showed some lesions progressing and some regressing, these studies demonstrate the great heterogeneity between granuloma types. Studies in the non-human primate have allowed investigation into features of progressing and controlling granulomas, identifying potential correlates of immune control (77), but even this model only partially recapitulates human disease.
Furthermore, the recent spatial studies highlight the complexity of cellular players, including the established fulcrum of macrophages and T cells, but additionally the importance of B cells, neutrophils and fibroblasts. For example, fibroblasts are emerging as important immune regulators in other lung diseases (78), and so it seems highly likely that they play an active role in TB-related inflammation. Fibroblast zonation can lead to feed-forward inflammatory loops and so may propagate disease (79). Consequently, the full spectrum of cell types in Mtb-infected lesions needs to be considered. Given that multiple immunological routes can lead to active TB, it seems unlikely that a single cellular component will fully explain the balance between Mtb containment and progression to active disease. However, the majority of studies continue to look for a single protective immune mechanism (41, 77, 80–82).
Considering the clinical and experimental evidence, immunological failure is likely to be a multistep process, whereby either one large deficit, or numerous small imbalances, can lead to progression and disease (Figure 3). Mtb and humans interact over many years, as the pathogen is difficult to eradicate due to its highly evolved survival mechanisms (83), and therefore in those individuals in whom it survives, there is a long period where it can escape host control. Potentially these different paths may ultimately cross or converge; if so, understanding the key nodes will allow more broadly effective treatments to emerge. For example, lung extracellular matrix degradation could be regarded as a final common pathway (84), but, again, this may result from different collagenases including macrophage-derived MMP-1 or neutrophil-derived MMP-8 (85, 86).

Escaping the human immune response from the pathogen’s perspective.
As control of Mtb requires a co-ordinated host response, there are multiple routes that Mtb can escape by. A major immune disturbance, such as TNF-α or PD-1 inhibition, gives a relatively direct route of exit. However, most individuals develop TB due to a series of less apparent immune events and no obvious global immune disturbance.
Emerging methodologies and the challenge of data analysis
The recent adoption of “omic” methodologies, including single cell transcriptomics, spatial transcriptomics, and proteomics, offers unprecedented opportunities to dissect the mechanisms of human TB pathogenesis and accelerate the development of effective interventions. These approaches generate large-scale, complex datasets that capture the heterogeneity of TB lesions. However, this complexity also presents significant analytical challenges. Standard computational pipelines, if not adapted to account for the biological and technical variability in these data, are unlikely to deliver robust or reproducible insights. A major obstacle is the integration of data from multiple studies and platforms, which can differ both within a single omic layer (horizontal integration) and across multiple omic layers (vertical integration) (87). Such integration risks data loss and inconsistent results, especially when data are not harmonised.
The prolonged co-evolution between host and pathogen has resulted in multiple routes to active TB, significantly adding to the biological heterogeneity that complicates the analysis and interpretation of multi-omic data. Addressing this complexity requires well-annotated clinical cohorts that capture the full spectrum of TB heterogeneity, ideally with longitudinal outcome data rather than single time-point snapshots. To accommodate the diversity of disease pathways, novel bioinformatic approaches are needed that move beyond the assumption of a single, unifying route to active TB.
The power of multi-omic approaches to reveal complex molecular relationships depends on both the quality of the omic data and the fit between experimental design and data integration strategies. For example, correlation-based integration requires matched samples across omics, sufficient sample numbers, and comparable variance structures. These requirements are often overlooked, leading to insufficient power, noisy data, and unrealistic integration (88). Each omic technology brings its own challenges. Signal-to-noise ratios differ across modalities, and appropriate algorithms are needed to estimate sample size and power for each. Notably, using the same sample numbers across modalities does not ensure comparable statistical power; achieving equal power can require unbalanced sample sizes (89). Other recognised challenges include the interpretation and validation of multi-omic models, standardised annotation, and data sharing (89). Recent developments include webbased, user-friendly tools that enable both knowledge- and data-driven multi-omic integration (90). Importantly, a wider use of artificial intelligence (AI) is transforming omics data analysis by providing robust methods to interpret complex biological datasets (91). Machine learning and deep learning techniques are now routinely applied to DNA, transcriptomic, proteomic, and metabolomic data, enabling more integrated and comprehensive analyses (91). In proteomics, for example, AI-driven approaches have enhanced peptide measurement predictions and accelerated biomarker discovery, often outperforming conventional assays (92). To improve interpretability, explainable AI (XAI) methods are increasingly employed, with feature relevance mapping and visual explanations emerging as preferred post hoc strategies (93). However, despite these developments, significant challenges remain in implementing XAI. Further research is needed to overcome these barriers and unlock the full translational potential of AI in omics analysis (92, 93). Ultimately, the utility of these innovations will depend on the functional validation of the resulting models.
Given the complexity of human TB, careful study design and unbiased integration methods that accommodate data limitations are essential. Spatial context is particularly important, as TB pathology involves three-dimensional immune responses. Extracellular matrix remodelling is a hallmark of TB granulomas [81], and the matrix regulates host cell biology [84]. Consequently, multiple data inputs such as matrix composition and organisation may be needed alongside host and Mtb transcriptomic data. Ultimately, local cellular events must be modelled at the tissue level, providing a second layer of computational complexity [85].
Addressing these challenges will require substantial investment in analytical capacity. Multi-omics is already transforming our understanding of disease heterogeneity, facilitating the identification of previously unrecognized subgroups, refining prognostic and therapeutic approaches, and providing deeper mechanistic insights. This strategy has successfully stratified rare tumours (94), profiled healthy populations (95), and enabled health screening to reveal previously hidden disease and risk subgroups (96), supporting the shift toward precision medicine. As human disease processes are rarely uniform, advances in multi-omic study design and analysis for TB will likely benefit a broad range of conditions.
Implications of diverse disease pathways for new TB interventions
Ultimately, the complexity of human TB underlies our inability to deploy a transformative intervention, and so global mortality remains depressingly high. The human-Mtb interaction is so closely co-evolved that experimental findings need to be interpreted in light of human disease phenomena. Multiomic studies in TB are currently being undertaken on small numbers due to cost and challenges in obtaining appropriate clinical samples, in specific regions, and so are unlikely to reflect the global heterogeneity. Therefore, results need to be interpreted with caution and wider studies will be needed to confirm the generalisability of findings. A critical aspect will be carefully curated and fully accessible metadata, so that as the body of omic datasets on human TB increase, they can be accurately integrated into wider analyses. Recurrent mining of these datasets is likely to be fundamental to understanding the breadth of TB pathogenesis. Missing metadata makes interpretation difficult, and in worst cases misleading. In addition, innovative computational approaches will be required whereby the analysis specifically accommodates multiple immune pathways to disease.
Given the toll that TB takes in the poorest parts of the world, we have a moral imperative to end the epidemic (97). To achieve this, we must acknowledge the complexity of human TB that has resulted from our prolonged co-evolution with the pathogen and the selective pressure of persistent Mtb exposure over millennia. Success depends on integrating the full spectrum of TB disease into our bioinformatic analyses, and ultimately understanding TB’s complexity can then inform logical interventions. If we seek a single unified explanation of TB disease, this seems to be unlikely to be successful.
Acknowledgements
PE was supported by the MRC (MR/W025728/1), MR by the Rosetrees Trust (CF-2021-2\126), SM by the MRC (MR/S024220/1) and AL by the Wellcome Trust (210662/Z/18/Z) and the BMGF (OPP1137006).
References
- 1.A short history of Robert Koch’s fight against tuberculosis: Those who do not remember the past are condemned to repeat itTuberculosis (Edinb) 83:86–90PubMedGoogle Scholar
- 2.The immune response in tuberculosisAnnu Rev Immunol 31:475–527https://doi.org/10.1146/annurev-immunol-032712-095939PubMedGoogle Scholar
- 3.TuberculosisLancet 405:850–66https://doi.org/10.1016/S0140-6736(24)02479-6PubMedGoogle Scholar
- 4.Protection by BCG Vaccine Against Tuberculosis: A Systematic Review of Randomized Controlled TrialsClin Infect Dis 58:470–80https://doi.org/10.1093/cid/cit790PubMedGoogle Scholar
- 5.Effect of BCG vaccination on childhood tuberculous meningitis and miliary tuberculosis worldwide: a meta-analysis and assessment of cost-effectivenessLancet 367:1173–80https://doi.org/10.1016/S0140-6736(06)68507-3PubMedGoogle Scholar
- 6.Final Analysis of a Trial of M72/AS01E Vaccine to Prevent TuberculosisN Engl J Med 381:2429–39https://doi.org/10.1056/NEJMoa1909953PubMedGoogle Scholar
- 7.Immunogenicity, safety, and efficacy of the vaccine H56:IC31 in reducing the rate of tuberculosis disease recurrence in HIV-negative adults successfully treated for drug-susceptible pulmonary tuberculosis: a double-blind, randomised, placebo-controlled, phase 2b trialLancet Infect Dis https://doi.org/10.1016/S1473-3099(24)00814-4PubMedGoogle Scholar
- 8.BCG Revaccination for the Prevention of Mycobacterium tuberculosis InfectionN Engl J Med 392:1789–800https://doi.org/10.1056/NEJMoa2412381PubMedGoogle Scholar
- 9.Co-evolution of Mycobacterium tuberculosis and Homo sapiensImmunol Rev 264:6–24https://doi.org/10.1111/imr.12264PubMedGoogle Scholar
- 10.The paleopathology and paleoepidemiology of Upper paleolithic tuberculosis: Review of evidence and hypothesesTuberculosis (Edinb) 143S:102348https://doi.org/10.1016/j.tube.2023.102348PubMedGoogle Scholar
- 11.Host-pathogen coevolution in human tuberculosisPhilos Trans R Soc Lond B Biol Sci 367:850–9https://doi.org/10.1098/rstb.2011.0316PubMedGoogle Scholar
- 12.The white plague : tuberculosis, man, and societyNew Brunswick: London: Rutgers University Press Google Scholar
- 13.The Primary Lung Focus of Tuberculosis in ChildrenLondon: J. & A. Churchill Google Scholar
- 14.Permutations of time and place in tuberculosisLancet Infect Dis 15:1357–60https://doi.org/10.1016/S1473-3099(15)00135-8PubMedGoogle Scholar
- 15.HIV and tuberculosis: a deadly human syndemicClin Microbiol Rev 24:351–76https://doi.org/10.1128/CMR.00042-10PubMedGoogle Scholar
- 16.Tuberculosis associated with infliximab, a tumor necrosis factor alpha-neutralizing agentN Engl J Med 345:1098–104https://doi.org/10.1056/NEJMoa011110PubMedGoogle Scholar
- 17.Interferon-gamma-receptor deficiency in an infant with fatal bacille Calmette-Guerin infectionN Engl J Med 335:1956–61PubMedGoogle Scholar
- 18.Impairment of mycobacterial but not viral immunity by a germline human STAT1 mutationScience 293:300–3https://doi.org/10.1126/science.1061154PubMedGoogle Scholar
- 19.Tuberculosis in otherwise healthy adults with inherited TNF deficiencyNature 633:417–25https://doi.org/10.1038/s41586-024-07866-3PubMedGoogle Scholar
- 20.TuberculosisLancet 387:1211–26https://doi.org/10.1016/S0140-6736(15)00151-8PubMedGoogle Scholar
- 21.Nutritional supplementation to prevent tuberculosis incidence in household contacts of patients with pulmonary tuberculosis in India (RATIONS): a field-based, open-label, cluster-randomised, controlled trialLancet 402:627–40https://doi.org/10.1016/S0140-6736(23)01231-XPubMedGoogle Scholar
- 22.The prognosis of a positive tuberculin reaction in childhood and adolescenceAmerican journal of epidemiology 99:131–8PubMedGoogle Scholar
- 23.Serial QuantiFERON testing and tuberculosis disease risk among young children: an observational cohort studyThe lancet Respiratory medicine 5:282–90https://doi.org/10.1016/S2213-2600(17)30060-7PubMedGoogle Scholar
- 24.Interferon-gamma release assay levels and risk of progression to active tuberculosis: a systematic review and dose-response meta-regression analysisBMC infectious diseases 21:467https://doi.org/10.1186/s12879-021-06141-4PubMedGoogle Scholar
- 25.Sex Differences in Tuberculosis Burden and Notifications in Low-and Middle-Income Countries: A Systematic Review and Meta-analysisPLoS Med 13:e1002119https://doi.org/10.1371/journal.pmed.1002119PubMedGoogle Scholar
- 26.Reconsidering the Optimal Immune Response to Mycobacterium tuberculosisAm J Respir Crit Care Med 201:407–13https://doi.org/10.1164/rccm.201908-1506PPPubMedGoogle Scholar
- 27.Implications of Tuberculosis Reactivation after Immune Checkpoint InhibitionAm J Respir Crit Care Med 198:1451–3https://doi.org/10.1164/rccm.201807-1250LEPubMedGoogle Scholar
- 28.Programmed death-1 (PD-1)-deficient mice are extraordinarily sensitive to tuberculosisProc Natl Acad Sci U S A 107:13402–7https://doi.org/10.1073/pnas.1007394107PubMedGoogle Scholar
- 29.CD4 T cells promote rather than control tuberculosis in the absence of PD-1-mediated inhibitionJ Immunol 186:1598–607https://doi.org/10.4049/jimmunol.1003304PubMedGoogle Scholar
- 30.PD-1 blockade exacerbates Mycobacterium tuberculosis infection in rhesus macaquesSci Immunol 6https://doi.org/10.1126/sciimmunol.abf3861PubMedGoogle Scholar
- 31.Anti-PD-1 immunotherapy leads to tuberculosis reactivation via dysregulation of TNF-αlphaeLife :9https://doi.org/10.7554/eLife.52668PubMedGoogle Scholar
- 32.Increased Tuberculosis Incidence Due to Immunotherapy Based on PD-1 and PD-L1 Blockade: A Systematic Review and Meta-AnalysisFrontiers in immunology 13:727220https://doi.org/10.3389/fimmu.2022.727220PubMedGoogle Scholar
- 33.Pulmonary tuberculosis associated with immune checkpoint inhibitors: a pharmacovigilance studyThorax https://doi.org/10.1136/thoraxjnl-2021-217575PubMedGoogle Scholar
- 34.Impact of Intermediate Hyperglycemia and Diabetes on Immune Dysfunction in TuberculosisClin Infect Dis 72:69–78https://doi.org/10.1093/cid/ciaa751PubMedGoogle Scholar
- 35.Rethinking the burden of latent tuberculosis to reprioritize researchNat Microbiol 9:1157–8https://doi.org/10.1038/s41564-024-01683-0PubMedGoogle Scholar
- 36.The Global Burden of Latent Tuberculosis Infection: A Re-estimation Using Mathematical ModellingPLoS Med 13:e1002152https://doi.org/10.1371/journal.pmed.1002152PubMedGoogle Scholar
- 37.Multi-ancestry meta-analysis of host genetic susceptibility to tuberculosis identifies shared genetic architectureeLife :13https://doi.org/10.7554/eLife.84394PubMedGoogle Scholar
- 38.Variations in the NRAMP1 gene and susceptibility to tuberculosis in West AfricansN Engl J Med 338:640–4https://doi.org/10.1056/NEJM199803053381002PubMedGoogle Scholar
- 39.Human genetics of tuberculosis: a long and winding roadPhilos Trans R Soc Lond B Biol Sci 369:20130428https://doi.org/10.1098/rstb.2013.0428PubMedGoogle Scholar
- 40.MR1-restricted T cell clonotypes are associated with “resistance” to Mycobacterium tuberculosis infectionJCI Insight 9https://doi.org/10.1172/jci.insight.166505PubMedGoogle Scholar
- 41.Specific CD4(+) T cell phenotypes associate with bacterial control in people who ‘resist’ infection with Mycobacterium tuberculosisNat Immunol 25:1411–21https://doi.org/10.1038/s41590-024-01897-8PubMedGoogle Scholar
- 42.Mycobacterium tuberculosis resisters despite HIV exhibit activated T cells and macrophages in their pulmonary alveoliJ Clin Invest https://doi.org/10.1172/JCI188016PubMedGoogle Scholar
- 43.Monocyte Transcriptional Responses to Mycobacterium tuberculosis Associate with Resistance to Tuberculin Skin Test and Interferon Gamma Release Assay ConversionmSphere :e0015922https://doi.org/10.1128/msphere.00159-22PubMedGoogle Scholar
- 44.IFN-gamma-independent immune markers of Mycobacterium tuberculosis exposureNat Med 25:977–87https://doi.org/10.1038/s41591-019-0441-3PubMedGoogle Scholar
- 45.Early Clearance of Mycobacterium tuberculosis Is Associated With Increased Innate Immune ResponsesJ Infect Dis 221:1342–50https://doi.org/10.1093/infdis/jiz147PubMedGoogle Scholar
- 46.Host pathogen genetic interactions underlie tuberculosis susceptibility in genetically diverse miceeLife :11https://doi.org/10.7554/eLife.74419PubMedGoogle Scholar
- 47.Cavitary pulmonary tuberculosis: The Holey Grail of disease transmissionCurrent Sci 86:74–81Google Scholar
- 48.In Vitro Granuloma Models of Tuberculosis: Potential and ChallengesJ Infect Dis 219:1858–66https://doi.org/10.1093/infdis/jiz020PubMedGoogle Scholar
- 49.Reconciling Mouse and Human Immunology at the Altar of GeneticsAnnu Rev Immunol 41:39–71https://doi.org/10.1146/annurev-immunol-101721-065201PubMedGoogle Scholar
- 50.Animal models of tuberculosisEur J Immunol 39:2011–4https://doi.org/10.1002/eji.200939542PubMedGoogle Scholar
- 51.Tuberculosis endotypes to guide stratified host-directed therapyMed (N Y) 2:217–32https://doi.org/10.1016/j.medj.2020.11.003PubMedGoogle Scholar
- 52.The Mycobacterium tuberculosis genome at 25 years: lessons and lingering questionsJ Clin Invest 133https://doi.org/10.1172/JCI173156PubMedGoogle Scholar
- 53.Ecology, global diversity and evolutionary mechanisms in the Mycobacterium tuberculosis complexNat Rev Microbiol https://doi.org/10.1038/s41579-025-01159-wPubMedGoogle Scholar
- 54.A smooth tubercle bacillus from Ethiopia phylogenetically close to the Mycobacterium tuberculosis complexNature communications 14:7519https://doi.org/10.1038/s41467-023-42755-9PubMedGoogle Scholar
- 55.The complete genome sequence of Mycobacterium bovisProc Natl Acad Sci U S A 100:7877–82https://doi.org/10.1073/pnas.1130426100PubMedGoogle Scholar
- 56.Human T cell epitopes of Mycobacterium tuberculosis are evolutionarily hyperconservedNat Genet 42:498–503https://doi.org/10.1038/ng.590PubMedGoogle Scholar
- 57.Mycobacterium tuberculosis as teacherNat Microbiol https://doi.org/10.1038/s41564-023-01454-3PubMedGoogle Scholar
- 58.M. tuberculosis Reprograms Hematopoietic Stem Cells to Limit Myelopoiesis and Impair Trained ImmunityCell 183:752–70 e22https://doi.org/10.1016/j.cell.2020.09.062PubMedGoogle Scholar
- 59.BCG Educates Hematopoietic Stem Cells to Generate Protective Innate Immunity against TuberculosisCell 172:176–90 e19https://doi.org/10.1016/j.cell.2017.12.031PubMedGoogle Scholar
- 60.Beyond Tuberculosis: The Surprising Immunological Benefits of the Bacillus Calmette-Guerin (BCG) Vaccine in Infectious, Auto-Immune, and Inflammatory DiseasesPathogens 14https://doi.org/10.3390/pathogens14020196PubMedGoogle Scholar
- 61.Non-specific effects of BCG vaccine on viral infectionsClin Microbiol Infect 25:1473–8https://doi.org/10.1016/j.cmi.2019.04.020PubMedGoogle Scholar
- 62.Acute lower respiratory tract infections and respiratory syncytial virus in infants in Guinea-Bissau: a beneficial effect of BCG vaccination for girls community based case-control studyVaccine 23:1251–7https://doi.org/10.1016/j.vaccine.2004.09.006PubMedGoogle Scholar
- 63.BCG Vaccination Protects against Experimental Viral Infection in Humans through the Induction of Cytokines Associated with Trained ImmunityCell Host Microbe 23:89–100 e5https://doi.org/10.1016/j.chom.2017.12.010PubMedGoogle Scholar
- 64.Vaccinations against smallpox and tuberculosis are associated with better long-term survival: a Danish case-cohort study 1971-2010International journal of epidemiology 46:695–705https://doi.org/10.1093/ije/dyw120PubMedGoogle Scholar
- 65.The Persian plateau served as hub for Homo sapiens after the main out of Africa dispersalNature communications 15:1882https://doi.org/10.1038/s41467-024-46161-7PubMedGoogle Scholar
- 66.Origins of modern human ancestryNature 590:229–37https://doi.org/10.1038/s41586-021-03244-5PubMedGoogle Scholar
- 67.Infectious diseases and human population historyBioscience 46:115–26Google Scholar
- 68.Plague and the end of antiquity : the pandemic of 541-750New York: Cambridge University Press :360Google Scholar
- 69.The Black Death, 1346-1353 : the complete historyWoodbridge, Suffolk, UK ; Rochester, N.Y., USA: Boydell Press :433Google Scholar
- 70.Beyond Killing Mycobacterium tuberculosis: Disease ToleranceFrontiers in immunology 9:2976https://doi.org/10.3389/fimmu.2018.02976PubMedGoogle Scholar
- 71.Tolerating the Unwelcome Guest; How the Host Withstands Persistent Mycobacterium tuberculosisFrontiers in immunology 9:2094https://doi.org/10.3389/fimmu.2018.02094PubMedGoogle Scholar
- 72.Tuberculosis as a three-act play: A new paradigm for the pathogenesis of pulmonary tuberculosisTuberculosis (Edinb) 97:8–17https://doi.org/10.1016/j.tube.2015.11.010PubMedGoogle Scholar
- 73.The Formation and Function of GranulomasAnnu Rev Immunol 36:639–65https://doi.org/10.1146/annurev-immunol-032712-100022PubMedGoogle Scholar
- 74.Spatial mapping reveals granuloma diversity and histopathological superstructure in human tuberculosisJ Exp Med 220https://doi.org/10.1084/jem.20221392PubMedGoogle Scholar
- 75.The immunoregulatory landscape of human tuberculosis granulomasNat Immunol https://doi.org/10.1038/s41590-021-01121-xPubMedGoogle Scholar
- 76.Inflammatory signaling in human tuberculosis granulomas is spatially organizedNat Med 22:531–8https://doi.org/10.1038/nm.4073PubMedGoogle Scholar
- 77.Multimodal profiling of lung granulomas in macaques reveals cellular correlates of tuberculosis controlImmunity 55:827–46 e10https://doi.org/10.1016/j.immuni.2022.04.004PubMedGoogle Scholar
- 78.Pulmonary inflammation and fibroblast immunoregulation: from bench to bedsideJ Clin Invest 133https://doi.org/10.1172/JCI170499PubMedGoogle Scholar
- 79.Fibroblasts as immune regulators in infection, inflammation and cancerNat Rev Immunol 21:704–17https://doi.org/10.1038/s41577-021-00540-zPubMedGoogle Scholar
- 80.CD8+ lymphocytes are critical for early control of tuberculosis in macaquesJ Exp Med 220https://doi.org/10.1084/jem.20230707PubMedGoogle Scholar
- 81.Antigen-specific B cells direct T follicular-like helper cells into lymphoid follicles to mediate Mycobacterium tuberculosis controlNat Immunol https://doi.org/10.1038/s41590-023-01476-3PubMedGoogle Scholar
- 82.Noncanonical T cell responses are associated with protection from tuberculosis in mice and humansJ Exp Med 222https://doi.org/10.1084/jem.20241760PubMedGoogle Scholar
- 83.Mycobacterium tuberculosis and the intimate discourse of a chronic infectionImmunol Rev 240:252–68https://doi.org/10.1111/j.1600-065X.2010.00984.xPubMedGoogle Scholar
- 84.Understanding the tuberculosis granuloma: the matrix revolutionsTrends Mol Med 28:143–54https://doi.org/10.1016/j.molmed.2021.11.004PubMedGoogle Scholar
- 85.MMP-1 drives immunopathology in human tuberculosis and transgenic miceJ Clin Invest 121:1827–33https://doi.org/10.1172/JCI45666PubMedGoogle Scholar
- 86.Neutrophil-Derived MMP-8 Drives AMPK-Dependent Matrix Destruction in Human Pulmonary TuberculosisPLoS Pathog 11:e1004917https://doi.org/10.1371/journal.ppat.1004917PubMedGoogle Scholar
- 87.Multi-omics data integration using ratio-based quantitative profiling with Quartet reference materialsNat Biotechnol 42:1133–49https://doi.org/10.1038/s41587-023-01934-1PubMedGoogle Scholar
- 88.Harmonization of quality metrics and power calculation in multi-omic studiesNature communications 11:3092https://doi.org/10.1038/s41467-020-16937-8PubMedGoogle Scholar
- 89.Undisclosed, unmet and neglected challenges in multi-omics studiesNat Comput Sci 1:395–402https://doi.org/10.1038/s43588-021-00086-zPubMedGoogle Scholar
- 90.Web-based multi-omics integration using the Analyst software suiteNat Protoc 19:1467–97https://doi.org/10.1038/s41596-023-00950-4PubMedGoogle Scholar
- 91.Artificial intelligence for omics data analysisBMC Methods 1:4https://doi.org/10.1186/s44330-024-00004-5Google Scholar
- 92.Artificial intelligence for proteomics and biomarker discoveryCell Syst 12:759–70https://doi.org/10.1016/j.cels.2021.06.006PubMedGoogle Scholar
- 93.Explainable artificial intelligence for omics data: a systematic mapping studyBrief Bioinform 25https://doi.org/10.1093/bib/bbad453PubMedGoogle Scholar
- 94.Molecular landscape and multi-omic measurements of heterogeneity in fetal adenocarcinoma of the lungNPJ Precis Oncol 8:99https://doi.org/10.1038/s41698-024-00569-yPubMedGoogle Scholar
- 95.A roadmap to the molecular human linking multiomics with population traits and diabetes subtypesNature communications 15:7111https://doi.org/10.1038/s41467-024-51134-xPubMedGoogle Scholar
- 96.Disease prediction with multi-omics and biomarkers empowers case-control genetic discoveries in the UK BiobankNat Genet 56:1821–31https://doi.org/10.1038/s41588-024-01898-1PubMedGoogle Scholar
- 97.Scientific advances and the end of tuberculosis: a report from the Lancet Commission on TuberculosisLancet 402:1473–98https://doi.org/10.1016/S0140-6736(23)01379-XPubMedGoogle Scholar
Article and author information
Author information
Version history
- Sent for peer review:
- Preprint posted:
- Reviewed Preprint version 1:
Cite all versions
You can cite all versions using the DOI https://doi.org/10.7554/eLife.108175. This DOI represents all versions, and will always resolve to the latest one.
Copyright
© 2025, Reichmann et al.
This article is distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use and redistribution provided that the original author and source are credited.
Metrics
- views
- 8
- downloads
- 0
- citations
- 0
Views, downloads and citations are aggregated across all versions of this paper published by eLife.