1,955
Views
16
CrossRef citations to date
0
Altmetric
Review

A review of clinical models for the evaluation of human TB vaccines

&
Pages 1177-1187 | Received 31 Aug 2015, Accepted 17 Dec 2015, Published online: 08 Apr 2016

ABSTRACT

While much progress has been made in the fight against the scourge of tuberculosis (TB), we are still some way from reaching the ambitious targets of eliminating it as a global public health problem by the mid twenty-first century. A new and effective vaccine that protects against pulmonary TB disease will be an essential element of any control strategy. Over a dozen vaccines are currently in development, but recent efficacy trial data from one of the most advanced candidates have been disappointing. Limitations of current preclinical animal models exist, together with a lack of a complete understanding of host immunity to TB or robust correlates of disease risk and protection. Therefore, in the context of such obstacles, we discuss the lessons identified from recent efficacy trials, current concepts of biomarkers and correlates of protection, the potential of innovative clinical models such as human challenge and conducting trials in high-incidence settings to evaluate TB vaccines in humans, and the use of systems vaccinology and novel technologies including transcriptomics and metabolomics, that may facilitate their utility.

Introduction

In the 1990s the World Health Organization (WHO) set targets to identify 70% and cure 85% of TB cases, which were reached in many countries by 2005.Citation1,2 Subsequently, targets to achieve a 50% reduction in the global mortality and prevalence of TB disease by 2015 were established by the Stop TB Partnership in 2000.Citation3 It has been estimated that in 2013 there were 9 million new cases and 1.5 million deaths attributed to TB, representing reductions in TB mortality and prevalence of 45% and 41%, respectively.Citation4

While this progress is significant, it is insufficient. It represents a reduction in the global TB incidence of approximately 1.5% per year over recent years.Citation4 However, to achieve the WHO target to eliminate TB as a global public health problem by 2050 (<1 case per million per year), a 1,000-fold reduction in global TB incidence over the next 35 years is required, corresponding to an unprecedented 20% reduction per year.Citation3,5

Until recently, the main focus of TB control and elimination strategies has been the prompt diagnosis and effective treatment of individuals with active disease to interrupt transmission. This approach is important, however even if transmission were interrupted completely and instantly in 2015, reactivation of established, and relapse of persisting, Mycobacterium tuberculosis (M. tb) infection would still cause an estimated >100 cases per million population in 2050.Citation5 An effective pre-exposure vaccine to protect against M. tb infection would not only be the most cost effective approach of control, but would also be a crucial component of any strategy to eliminate the global burden of TB.Citation6,7 In addition, there is an increasing realization that the management of latently infected individuals, which represents a huge reservoir of potential new disease and thus infectiousness, will also be required to reduce TB disease. Modeling suggests that the effective treatment of latent infection with a drug or a vaccine (and likely both) would reduce TB incidence significantly.Citation8

TB vaccines – the old and the new

Bacille Calmette-Guerin (BCG), an attenuated strain of M. bovis, is the only licensed vaccine for TB.Citation9 It has been part of the WHO Expanded Programme on Immunisation (EPI) since the early 1970s and is the most widely used vaccine in history, with over 4 billion doses administered to date. The WHO currently recommends that a single dose of BCG be given to neonates or as soon as possible after birth in countries with a high prevalence of TB.Citation10 Low-burden countries may choose to limit BCG vaccination to neonates and infants of recognized high-risk groups for TB or to tuberculin-negative older children. BCG vaccination is also recommended for unvaccinated, tuberculin-negative persons in non-endemic areas who are exposed to multi-drug resistant M. tb.

BCG has been shown to be effective at preventing disseminated TB disease, such as miliary and meningeal TB, in children.Citation11 In addition, BCG vaccination is thought to have non-specific effects and there is evidence that it correlates with a reduction in general infant mortality.Citation12,13 While generally safe, it is not recommended for use in immunocompromised individuals due to concerns over the possible development of disseminated BCG disease.Citation14,15

However, it is considered that BCG vaccination has had little impact on the overall incidence of TB.Citation16 The efficacy of BCG is highly variable, ranging from 0–80% in different settings, vaccine-induced immunity may wane with time, and it fails to confer adequate protection against pulmonary disease, particularly among adolescents and young adults in high-endemic regions.Citation17-21 This is a significant problem as this population continues to propagate the TB epidemic; the incidence of latent TB infection (LTBI) reaches 60–70% in adults aged over 25 years in most affected areas.Citation22 There is therefore an urgent need to develop novel, effective, TB vaccines.

Vaccines against TB may be either prophylactic and/or therapeutic and have the potential to be directed against several stages of M. tb infection and disease. Prophylactic vaccines may be administered either pre- or post-exposure to M. tb and mathematical modeling suggests that such deployment would most rapidly achieve global control of the TB epidemic.Citation8 A pre-exposure vaccine would prevent primary acquisition of M. tb infection and would ideally be administered in infancy, prior to infection. A post-exposure vaccine would be administered to adolescents and young adults following infection with M. tb to prevent post-primary disease and/or reactivation of latent infection. Finally, a therapeutic vaccine would target individuals with active TB disease as adjunctive therapy to simplify, enhance the efficacy of, and shorten drug treatment. Such vaccines should be effective against both drug-sensitive and resistant strains of M. tb.Citation16,23 In addition, the ideal vaccine would be safe in all age groups, in patients with HIV infection and would induce long-term and effective immunological memory, abrogating the need for repeated vaccination or boosting.Citation23

Current strategies to develop an improved TB vaccination regimen have focused on improving BCG, boosting it, or replacing it with a different vaccine altogether.Citation24-26 The fact that many of these strategies attempt to exploit the immunity induced by priming with BCG is logistically pragmatic as the majority of the target population has been vaccinated with BCG in childhood as part of EPI. TB vaccines can be broadly classified as either subunit or whole, live-attenuated, mycobacterial vaccines, and at present there are 16 candidate vaccines in active clinical evaluation ().Citation27 A detailed description of these has recently been undertaken and is beyond the scope of this article, however valuable lessons may be drawn from reviewing one of the most advanced of the candidate TB vaccines, MVA85A.Citation28,29

Table 1. Summary of TB vaccines currently under clinical assessment (adapted fromCitation27).

Lessons from recent efficacy trials

MVA85A is a recombinant strain of Modified Vaccinia virus Ankara expressing the conserved M. tb antigen 85A.Citation30 While still important in terms of immunogenicity, recent evidence suggests that antigen 85A is less immunodominant than previously thought.Citation31 MVA85A was developed for a heterologous prime-boost strategy, to be administrated following BCG vaccination to augment antigen-specific T cells. It has recently completed the largest infant phase IIb efficacy trial since the introduction of BCG over 90 years ago in a BCG-prime, MVA85A-boost regimen, and the disappointing results have highlighted the significant challenges in the field of TB vaccine development and testing.Citation32

Extensive preclinical studies in animal models (mice, guinea pigs and non-human primates) demonstrated that boosting of BCG with MVA85A could improve protection against mycobacterial challenge, although not consistently in every challenge experiment.Citation33-36 There are several limitations to these models which are discussed below and extensively reviewed elsewhere.Citation37

Several human phase I/IIa studies in both high and low disease burden settings among adults, adolescents, children and infants showed MVA85A to be safe and immunogenic (the two endpoints tested in such trials).Citation38-48 Among healthy, M. tb-infected or HIV-infected individuals, MVA85A induced antigen-specific Th1 and Th17 cells, which are both considered important in protection against M. tb.Citation30,43,46,47

Recently, almost 2800 healthy, HIV-negative, BCG-vaccinated South African infants (4–6 months old) were randomized to receive either MVA85A or placebo. MVA85A was well tolerated but induced only modest antigen-specific T cell responses (several-fold lower than those seen in UK adults) and did not confer any additional protection over BCG alone to M. tb infection (vaccine efficacy was −3.8% and 17.3% against M. tb infection and disease, respectively).Citation32 These outcomes were clearly in contrast to the earlier encouraging preclinical results.

A further phase IIb trial assessing the efficacy of MVA85A in over 650 healthy adults infected with HIV in South Africa and Senegal has recently been reported.Citation49 MVA85A was safe and immunogenic, inducing significant increases in antigen-specific T cell responses which were primarily monofunctional interferon gamma (IFN-γ) and tumor necrosis factor α (TNF-α)-producing CD4+ T cells. However, there was no efficacy against M. tb infection or disease in the MVA85A group when compared to placebo (vaccine efficacy was 11.7% and 32.8% against M. tb infection and disease, respectively).

Despite these outcomes, the advanced MVA85A trials have been extremely important. Firstly, they demonstrate that it is feasible to conduct large-scale clinical efficacy trials of vaccines against TB in high-burden settings and in the target population. Secondly, the results raise several fundamental questions relevant to the whole field, which has stimulated debate and generated innovative proposals for the evaluation of TB vaccine candidates in the future.

It is now evident that the variable and modest efficacy seen in preclinical animal models was not able to predict protection in BCG-vaccinated infants or HIV-infected adults. Several reasons have been suggested to account for the disparity between the animal data and the outcomes of human efficacy trials.Citation37

Species differences that may influence the predictive ability of animal models exist. The manifestations of M. tb infection and disease are different between species and immune responses to vaccination are more variable in humans.Citation19,50 For example, the structure and heterogeneity of murine granulomas do not mimic those seen in humans infected with M. tb and there are no simple animal models of latent M. tb infection that easily represent the human situation except potentially a non-human primate model.Citation51 There is also genetic immunological variation between species (e.g. the absence of most CD1 subtypes in mice), with implications on correlating immune responses with those seen in humans.Citation52 Furthermore, experimental animal models generally use adult animals and while the target population for some candidate vaccines is human adults, many are designed for use in infants and adolescents. Immunogenicity studies are needed to assess different immune responses at various ages.

Secondly, there are significant differences in the nature of exposure between animal challenge experiments and natural infection in humans. In the former, laboratory strains of M. tb are used in a single exposure via a variety of challenge routes and at much higher inocula than that seen in natural infection. However, humans are likely to experience multiple low dose exposures of clinical strains, with the establishment of infection following one or several of these exposures (or, indeed, not at all).Citation53 Recent evidence from a study assessing the protection of BCG and a novel candidate vaccine against newly emerging, mostly highly virulent, strains of M. tb has highlighted the importance of the fitness of prevalent strains of M. tb at clinical trial sites when trying to show vaccine efficacy.Citation54

There is, therefore, a need to develop experimental models of infection more comparable to natural challenge.

Thirdly, there are fundamental differences in study power, definitions and endpoints. The MVA85A efficacy trial was powered to detect a 60% improvement over BCG, however the magnitude of candidate vaccine efficacy in animal models was much lower than this and would not have been detected in clinical trials.Citation32,37 Also, the definition of protection varies between human and animal models. In animals, protective efficacy of a candidate vaccine is assessed in terms of improvement in the extent of disease using markers such as organ bacillary load, severity of pathology or time to death. In contrast, human trials define efficacy as the prevention of disease. Clearly, there is a fundamental difference between these endpoints, which lack intuitive correlation. It has been suggested that changing animal trial endpoints from TB disease reduction to disease prevention should be considered and, still further, possibly establishing animal challenge models to show prevention of M. tb infection rather than protection from disease, which may be a more feasible endpoint in human efficacy trials.Citation37 However, limitations of this approach include the need to use much larger numbers of animals, and the lack of validated biomarkers of M. tb infection to correlate parameters in animal models with human efficacy data. As discussed later, while to date the focus of human efficacy trials has been prevention of disease, the low number of endpoints requires large clinical trials which are both expensive and time consuming. Recently there has been renewed interest in using prevention of infection as a measure of candidate vaccine efficacy which would provide more endpoints but presupposes that the underlying immune mechanisms of M. tb infection and disease are similar and highlights the limitations of diagnostics and the definition of M. tb infection.Citation55

Finally, there are several other aspects of variation in human clinical trial settings that contrast with the laboratory, such as nutritional status and diet, exposure to non-tuberculous mycobacteria (NTM), helminth infection and the effect of host genetic heterogeneity on susceptibility. For example, several studies have shown poor BCG vaccine efficacy in human populations with high levels of prior exposure to environmental (NTM).Citation56-59 A recent systematic review of randomized controlled trials found that the absence of sensitization with NTM was associated with higher efficacy of BCG against pulmonary (and also possibly miliary and meningeal) TB.Citation60 It has been suggested that pre-existing immunity to NTM results in either ‘blocking’ the effects of BCG vaccination by inhibiting the replication of BCG and preventing the induction of protective immune responses or ‘masking’ the effects as BCG is unable to further boost background immunity induced by NTM. In addition, chronic helminth infections may also have an modulatory effect on vaccine efficacy, causing a shift toward Th2-type immunity, impaired antigen-specific and Th1-type responses and the induction of regulatory T cells producing transforming growth factor-β and other inhibitory cytokines which suppress pro-inflammatory cytokines. These responses have been associated with reduced efficacy of BCG in endemic settings.Citation61-64

Fundamental questions that remain are what is the magnitude of improvement in animal challenge models needed to predict the significant improvement in protection required in humans and which immunological parameters have the greatest predictive power for vaccine efficacy? Recent efficacy trial data have highlighted the need to establish biomarkers that correlate with vaccine-induced protection against TB disease as, without such biomarkers, the only way to safely assess vaccine efficacy is by large trials with the diagnosis of TB disease as the endpoint.

Biomarkers and correlates of protection – what should we look for, how and in whom?

Current concepts

Immune correlates of vaccine-induced protection are biomarkers that reliably predict the level of protective efficacy induced by a vaccine. A biomarker is a unique indicator of a biological process and a biosignature is a combination of independent biomarkers which markedly increases the power of an individual marker.Citation65,66 Biomarkers for vaccines are typically identified by assessing differences in immune parameters between vaccinated individuals who are protected and unvaccinated, unprotected, control groups.Citation67 At present there are no validated correlates to reliably assess the efficacy of candidate TB vaccines. Moreover, host biomarkers are urgently needed to improve TB diagnostics and for the development of more effective and shorter treatment regimens. The perfect biomarker or biosignature for TB would differentiate between patients with active disease and latent infection, return to baseline levels following successful treatment, reproducibly predict clinical outcomes, predict vaccine efficacy and provide endpoints for clinical trials.Citation67 As such, robust biomarkers would have significant utility as surrogate endpoints rather than relying on clinical endpoints. However, based on our current understanding and knowledge of the biological and immunological responses that underlie discrete states of TB immunopathogenesis, it is unlikely that such a perfect biomarker exists.

The lack of a correlate of protection is a significant obstacle in TB vaccine development and persists due to our incomplete understanding of the natural infection and mechanisms contributing to host immunity to TB. To date, the most common parameters measured to assess TB vaccine immunogenicity are those considered important for protection against infection or disease. These have been determined through observational studies of mycobacterial susceptibility in humans and experimental animal models. The importance of IFN-γ production by T cells is demonstrated by the significant increases in rates of M. tb in conditions of immune deficiency. These may be primary (e.g., the syndromes of Mendelian susceptibility to mycobacterial diseases, MSMD) or acquired (e.g. HIV infection) and reduce the number and/or function of CD4+ T cells or impair IFN-γ signaling.Citation68-70 As such, the primary immunological readout of TB vaccine studies is antigen-specific IFN-γ production by T cells, typically using ELISpot assays.Citation67 In addition, other Th1 cytokines such as TNF-α and frequencies of polyfunctional CD4+ T cells, determined by multi-parametric flow cytometry, are also thought to be important.Citation67,71 However, there is clearly a disparity between those immunological responses stimulated by vaccination (correlates of immunogenicity) and those associated with protection from TB disease (correlates of protection).Citation72 Several studies have shown that while significant increases in antigen-specific IFN-γ secretion and changes in polyfunctional T cell profiles may be induced by vaccination, these responses do not correlate with protection against M. tb.Citation32,40,73-75

Thus far, the immune responses considered essential for protection may well be necessary but they are not sufficient and as such these parameters do not have utility as correlates of risk or protection in TB vaccine efficacy trials. Other elements thought to have a role in protection that warrant further investigation include IL17-producing Th-17 cells, regulatory CD4+ T cells, CD8+ T cells, γδ cells, natural killer cells and components of innate immunity. The role of B cells also remains to be defined.Citation67,71 It is likely that rather than a simple effector or memory output (such as that for the serogroup C meningococcal vaccine), multiple factors deployed in a coordinated and balanced immune response will be crucial for effective protection and therefore should be assessed in concert.Citation76

Characterizing naturally induced protection in M. tb infection among household contacts exposed to patients with pulmonary disease could be used to identify biomarkers that correlate with protective immunity.Citation71 Such contacts exhibit diverse immune responses that would be suitable to study using systems vaccinology and would improve our understanding of the spectrum of M. tb immunopathology. For example, it is hypothesized that, despite significant exposure to M. tb, some individuals have no evidence of immune sensitization which is likely due to inherent resistance or the elimination of infection through an effective innate immune response or non-primed adaptive immunity. In contrast, latently infected, asymptomatic individuals, probably exist on a spectrum of M. tb infection and exhibit immunological evidence of T cell priming and persisting quiescent infection which is controlled by the acquired immune response.Citation77,78 Within this heterogeneous group the host-pathogen relationship is highly dynamic and some individuals may eventually effectively eliminate the infection, others will maintain persistent, life-long infection, while another group will develop subclinical disease and progress to primary active disease or reactivation TB.Citation77,79,80 Household contacts therefore define a range of immune phenotypes that could help in characterizing correlates of risk and protection with application to TB vaccine development. Studies of this group are also relevant as individuals with LTBI are a potential target population for vaccination.

The potential and pitfalls of systems vaccinology

One potential method to identify biosignatures of protective immunity to TB is to use high-throughput ‘omics’ technologies in a ‘systems vaccinology’ approach.Citation72,81–84 This approach can be used to study the mechanisms of vaccine-induced immunity by assessing the dynamics and interactions of multiple components of the immune system through iterative cycles of perturbations and high-throughput biology. It is exemplified by early studies with the yellow fever vaccine, YF-17D, in which systems biology was used to identify the mediators and predictors of the immune response following vaccination.Citation85,86 Similar approaches have now been applied to several other vaccines.Citation72,87

Systems vaccinology is based on the same principles as systems biology. Following vaccination, perturbations of the immune system are profiled by using high-throughput techniques on biological samples (e.g., DNA and RNA sequencing, transcriptomic microarrays, proteomics and metabolomics). Data derived from these multiple platforms are then integrated with those obtained from assays routinely used in vaccinology (e.g. ELISpot and flow cytometry). Mathematical models are subsequently created from a variety of modeling frameworks to describe and/or predict the vaccine-induced immune responses observed.Citation88 Such biosignatures must then be validated using independent sets of samples.Citation72,89

A significant benefit to using ‘omic’ technologies is that they offer an unbiased, hypothesis-generating approach from which findings may be subsequently investigated by more targeted experiments. As described, we still do not fully understand the basis of host immunity to M. tb or the protection afforded by vaccination, and immune responses are heterogeneous and controlled by several levels of regulation. While targeted approaches assessing specific biomarkers such as cytokine production or the characterization of immune cell phenotypes in antigen-stimulated samples (e.g., whole blood or peripheral blood mononuclear cells) have advanced our knowledge, they are insufficient. Moreover, peripheral blood samples, though easily obtainable, may not represent the complete immunological milieu at the principal interface between host and mycobacteria - the pulmonary granuloma. This structure provides the morphological architecture for regional immune processes which are central to outcome in TB.Citation90 Untargeted approaches using more appropriate material, such as bronchoalveolar lavage samples and lung tissue, may yield greater insights and contribute to filling some of the gaps in our knowledge.

Genome-wide differential gene expression studies, typically microarray transcriptomics, represent the archetypal ‘omic’ approach. Several studies have identified gene signatures that discriminate individuals with TB from healthy controls. Increased IFN-α/β signaling, pro-inflammatory signaling through the Janus kinase pathway, and differential expression of Fc-γ receptors, innate immune-related genes and gene clusters involved in apoptosis and natural killer cell activity have all been described.Citation91-101 Changes in the transcriptome during treatment, indicative of modulation of humoral responses, have also been reported.Citation102 Interestingly, a recent combined ‘meta-like’ analysis of all transcriptomic data reported in human TB pathogenesis showed a myeloid-derived inflammatory signature to be of particular importance.Citation103 There are undoubtedly unresolved issues in this area, such as the need to define disease-specific profiles and difficulties in comparing data from studies using different cohorts, experimental techniques and approaches to data analysis.Citation104 However, there are also several exciting developments including the increasing availability of affordable RNA sequencing and growing interest in the role of microRNAs (miRNAs) in TB and their potential utility as biomarkers and even as targets for therapeutic intervention.Citation105-111

Proteomic profiling of serum samples from TB patients using high-resolution mass spectrometry or protein microarrays has identified several peptides and antibodies that may have diagnostic potential.Citation112-116 Most recently, mass spectrometry has also been used to isolate a novel antigen from human TB granulomas.Citation117

Finally, metabolic profiling is the identification of small molecular metabolites (e.g. amino acids, lipids, fatty acid, sugars and nucleotides) in clinical samples using high-throughput methods. This approach has shown distinct metabolic biosignatures associated with different TB disease states and responses to treatment in serum, urine, and breath from patients and uninfected controls.Citation118-121 Differential profiles have also been reported between different lineages of infecting mycobacteria following anti-TB chemotherapy.Citation122

However, there are several challenges to developing systems vaccinology. Integrating large datasets from different techniques is computationally complex and advances in bioinformatics are needed to obtain the greatest value from these high-throughput data.Citation123 Recent progress in this area includes the rationalisation of large numbers of genes into smaller, distinct immune modules in which the constituents are highly related and typically expressed in a coordinated way.Citation124 This approach is logical as biological processes occur in a modular manner and it affords simpler data analysis and more intuitive interpretation. It has been applied successfully to several diseases and infections (including M. tb) and recently to vaccination studies.Citation91,125–127

Systems vaccinology is an exciting development and potentially an extremely powerful tool for understanding vaccine-induced immunity and predicting vaccine efficacy. However, it will need further refinement and close collaboration between vaccinologists, immunologists and bioinformaticians if it is to yield truly valuable outputs.

Alternative approaches to clinical efficacy trials

An alternative, but complementary approach, to testing vaccine efficacy in clinical trials is to identify correlates of protection using human pathogen challenge models. Such models have shown great utility in malaria, influenza, dengue, cholera and typhoid vaccine development.Citation128-135 They also have the potential advantage of gating promising vaccine candidates at an early stage, with significant implications on cost and time.

Clearly, it is not possible to challenge volunteers with strains of virulent M. tb, however there is increasing interest in using BCG derived from M. bovis as a surrogate for M. tb infection in a human mycobacterial challenge model. The basis of this model in the hypothesis that an effective TB vaccine, which reduces or prevents M. tb replication, should have a similar effect on BCG replication. The benefits of using BCG in this model include that it has significant genetic sequence homology to live M. bovis (and therefore M. tb), it is a functional, replicating organism that results in limited infection in the immunocompetent host and, crucially, it is safe and licensed for use in humans by intradermal administration.Citation136 Optimisation of a BCG human challenge model could be used to establish the clinical parameters for subsequent challenge models using attenuated M. tb strains.

A murine model has shown that live BCG can persist in skin for at least 4 weeks and in draining lymph nodes for up to 12 weeks, and that intradermal BCG vaccination consistently protects against a BCG challenge, independent of vaccine dose, route of challenge (intradermal or intranasal) or the interval between vaccination and challenge.Citation137 Similar findings have been seen using a novel BCG intranodal challenge model in cattle.Citation138 Other murine studies have demonstrated the efficacy of BCG vaccination against M. tb aerosol challenge.Citation139-141 Taken together, these data suggest that intradermal BCG challenge may reflect a pulmonary vaccine effect, supporting the relevance of a mycobacterial skin challenge in predicting vaccine efficacy against M. tb. This approach has most recently been used in a human model in which healthy BCG-naïve and BCG-vaccinated volunteers were challenged with intradermal BCG and BCG load was quantified from skin biopsy specimens. In previously BCG-vaccinated individuals, quantitative PCR analysis of biopsies reflected a degree of mycobacterial immunity to challenge.Citation142 These data were supported by subsequent transcriptomic analysis showing that immune signatures, particularly IFN-γ and IL-17 pathways, were strongly induced in previously BCG-vaccinated volunteers and correlated with reduced mycobacterial growth following BCG challenge.Citation143,144

However, in early studies culture data were not supportive of the PCR findings in detecting a difference between naïve and vaccinated groups, which may be due to an overestimation of the protective effect of BCG by PCR, or less contemporaneous and reliably comparable results obtained by culture.Citation142 In addition, a major limitation to date has been low mycobacterial recovery which has reduced the sensitivity of the model and its ability to discriminate between individuals with differing levels of vaccine-induced anti-mycobacterial immunity. Optimisation studies addressing this issue by evaluating the effect of BCG strain and dose on mycobacterial recovery are in progress. Furthermore, confirmation that BCG challenge reflects pulmonary vaccine effect may ultimately require parallel intradermal and pulmonary challenge trials and comparison of validated immune correlates of protection. Interestingly, a novel intranodal BCG challenge model in cattle has previously shown that BCG vaccine effect is similar to that seen following aerosol challenge with M. bovis.Citation138 While it is recognized that further optimisation and development of human BCG challenge models is needed, together with consideration of the target population and type of vaccine candidate that the model has greatest relevance to, the concept represents a promising development that warrants further investigation.

There is increasing interest in matching the route of vaccination to the route of natural infection and the first phase I trial assessing a candidate vaccine administered to humans by aerosol has been completed.Citation145 The rationale for this approach is that local protective immune responses may be enhanced following delivery of a TB vaccine directly to the respiratory mucosa, thus optimising protection against pulmonary disease. Similarly, the development of a human aerosol BCG challenge model, in a carefully controlled setting, would be more representative of natural infection and could validate an intradermal BCG challenge model.

Finally, in the future there will undoubtedly still be a need to conduct clinical efficacy trials of vaccines that have been highly selected and show greatest potential. Undertaking trials in high TB incidence settings may be of particular value. A study among adolescents in South Africa showed that 50% of the cohort had evidence of LTBI, as demonstrated by a positive IFN-γ release assay (IGRA).Citation146,147 It has been estimated that with a 10% annual conversion rate and 0.05% error, a vaccine trial powered to show a 60% effect against infection would need 1000 participants and one year of follow-up.Citation55 In contrast to regions with low rates of M. tb infection where IGRA serial testing shows frequent conversions and reversions, among adolescents in a high incidence setting there is less IGRA variability, conversion rates are similar to those seen with TST and predict the development of active TB disease in subsequent years.Citation148-150 Therefore, IGRAs may have utility as a biomarker for M. tb infection in such settings and offer an endpoint for vaccine trials.Citation55

However, several of the issues previously discussed that may impact vaccine efficacy in human clinical trials will be prevalent in these settings, such as exposure to NTM and helminth infection.

Conclusions

TB remains one of the greatest burdens on global health and the importance of an effective vaccine in controlling the epidemic is undisputed. Over the past 20 years, much progress in TB vaccine development has been made resulting in several candidates in clinical assessment. While the results of recent efficacy trials have been disappointing, they have highlighted the need to overcome important obstacles which will be essential if we are to succeed in developing a vaccine against M. tb, such as the need to define and validate biosignatures of immune correlates of vaccine-induced protection. This has forced the field to re-evaluate its approach to TB vaccine design and evaluation which has been a positive outcome, complemented by the emergence of exciting and potentially very powerful technologies such as those of systems vaccinology. Despite the many challenges ahead, with continued coordination and collaboration within the TB vaccine community, iterative progress will be made.

Disclosure of potential conflicts of interest

HMcS was previously a shareholder in the Oxford-Emergent Tuberculosis Consortium (OETC), a joint venture established for the development of MVA85A (OETC no longer exists). Both authors declare no competing interests.

Funding

HMcS holds a Wellcome Trust Senior Clinical Research Fellowship and is a Jenner Investigator. MKO is a Wellcome Trust Research Training Fellow.

References

  • Dye C, Garnett GP, Sleeman K, Williams BG. Prospects for worldwide tuberculosis control under the WHO DOTS strategy. Directly observed short-course therapy. Lancet 1998; 352:1886-91; PMID:9863786; http://dx.doi.org/10.1016/S0140-6736(98)03199-7
  • Dye C, Hosseini M, Watt C. Did we reach the 2005 targets for tuberculosis control? Bull World Health Organ 2007; 85:364-9; PMID:17639221; http://dx.doi.org/10.2471/BLT.06.037580
  • Partnership ST. The Global Plan to Stop TB 2011–2015. Geneva, Switzerland: World Health Organization, 2010
  • WHO. Global tuberculosis report 2014. Geneva, Switzerland: World Health Organization, 2014
  • Dye C, Glaziou P, Floyd K, Raviglione M. Prospects for tuberculosis elimination. Annu Rev Public Health 2013; 34:271-86; PMID:23244049; http://dx.doi.org/10.1146/annurev-publhealth-031912-114431
  • Dye C, Williams BG. Eliminating human tuberculosis in the twenty-first century. J R Soc Interface 2008; 5:653-62; PMID:17690054; http://dx.doi.org/10.1098/rsif.2007.1138
  • Tseng CL, Oxlade O, Menzies D, Aspler A, Schwartzman K. Cost-effectiveness of novel vaccines for tuberculosis control: a decision analysis study. BMC Public Health 2011; 11:55; PMID:21269503; http://dx.doi.org/10.1186/1471-2458-11-55
  • Abu-Raddad LJ, Sabatelli L, Achterberg JT, Sugimoto JD, Longini IM, Jr., Dye C, Halloran ME. Epidemiological benefits of more-effective tuberculosis vaccines, drugs, and diagnostics. Proc Natl Acad Sci U S A 2009; 106:13980-5.; PMID:19666590; http://dx.doi.org/10.1073/pnas.0901720106
  • Calmette A. Preventive Vaccination Against Tuberculosis with BCG. Proc R Soc Med 1931; 24:1481-90; PMID:19988326
  • BCG vaccine. WHO position paper. Wkly Epidemiol Rec 2004; 79:27-38; PMID:14768305
  • Trunz BB, Fine P, Dye C. Effect of BCG vaccination on childhood tuberculous meningitis and miliary tuberculosis worldwide: a meta-analysis and assessment of cost-effectiveness. Lancet 2006; 367:1173-80; PMID:16616560; http://dx.doi.org/10.1016/S0140-6736(06)68507-3
  • Blok BA, Arts RJ, van Crevel R, Benn CS, Netea MG. Trained innate immunity as underlying mechanism for the long-term, nonspecific effects of vaccines. J Leukoc Biol 2015; 98(3):347-56
  • Roth A, Garly ML, Jensen H, Nielsen J, Aaby P. Bacillus Calmette-Guerin vaccination and infant mortality. Expert Rev Vaccines 2006; 5:277-93; PMID:16608427; http://dx.doi.org/10.1586/14760584.5.2.277
  • Hesseling AC, Marais BJ, Gie RP, Schaaf HS, Fine PE, Godfrey-Faussett P, Beyers N. The risk of disseminated Bacille Calmette-Guerin (BCG) disease in HIV-infected children. Vaccine 2007; 25:14-8; PMID:16959383; http://dx.doi.org/10.1016/j.vaccine.2006.07.020
  • Revised BCG vaccination guidelines for infants at risk for HIV infection. Wkly Epidemiol Rec 2007; 82:193-6
  • Andersen P, Kaufmann SH. Novel vaccination strategies against tuberculosis. Cold Spring Harb Perspect Med 2014; 4; pii: a018523; PMID:24890836.
  • Abubakar I, Pimpin L, Ariti C, Beynon R, Mangtani P, Sterne JA, Fine PE, Smith PG, Lipman M, Elliman D, et al. Systematic review and meta-analysis of the current evidence on the duration of protection by bacillus Calmette-Guerin vaccination against tuberculosis. Health Technol Assess 2013; 17:1-372, v-vi; PMID:24021245
  • Andersen P, Doherty TM. The success and failure of BCG - implications for a novel tuberculosis vaccine. Nat Rev Microbiol 2005; 3:656-62; PMID:16012514; http://dx.doi.org/10.1038/nrmicro1211
  • Colditz GA, Brewer TF, Berkey CS, Wilson ME, Burdick E, Fineberg HV, Mosteller F. Efficacy of BCG vaccine in the prevention of tuberculosis. Meta-analysis of the published literature. Jama 1994; 271:698-702; PMID:8309034; http://dx.doi.org/10.1001/jama.1994.03510330076038
  • Fine PE. Variation in protection by BCG: implications of and for heterologous immunity. Lancet 1995; 346:1339-45; PMID:7475776; http://dx.doi.org/10.1016/S0140-6736(95)92348-9
  • Colditz GA, Berkey CS, Mosteller F, Brewer TF, Wilson ME, Burdick E, Fineberg HV. The efficacy of bacillus Calmette-Guerin vaccination of newborns and infants in the prevention of tuberculosis: meta-analyses of the published literature. Pediatrics 1995; 96:29-35; PMID:7596718
  • Gallant CJ, Cobat A, Simkin L, Black GF, Stanley K, Hughes J, Doherty TM, Hanekom WA, Eley B, Beyers N, et al. Impact of age and sex on mycobacterial immunity in an area of high tuberculosis incidence. Int J Tuberc Lung Dis 2010; 14:952-9; PMID:20626938
  • Checkley AM, McShane H. Tuberculosis vaccines: progress and challenges. Trends Pharmacol Sci 2011; 32:601-6; PMID:21803435; http://dx.doi.org/10.1016/j.tips.2011.06.003
  • Kaufmann SH, Lange C, Rao M, Balaji KN, Lotze M, Schito M, Zumla AI, Maeurer M. Progress in tuberculosis vaccine development and host-directed therapies–a state of the art review. Lancet Respir Med 2014; 2:301-20; PMID:24717627; http://dx.doi.org/10.1016/S2213-2600(14)70033-5
  • Orme IM. Vaccine development for tuberculosis: current progress. Drugs 2013; 73:1015-24; PMID:23794129; http://dx.doi.org/10.1007/s40265-013-0081-8
  • Orme IM. Tuberculosis vaccine types and timings. Clin Vaccine Immunol 2015; 22:249-57; PMID:25540272; http://dx.doi.org/10.1128/CVI.00718-14
  • Frick M. The Tuberculosis Vaccines Pipeline: A New Path to the Same Destination? London, UK HIV i-Base/Treatment Action Group, 2015
  • da Costa C, Walker B, Bonavia A. Tuberculosis vaccines–state of the art, and novel approaches to vaccine development. Int J Infect Dis 2015; 32:5-12; PMID:25809749; http://dx.doi.org/10.1016/j.ijid.2014.11.026
  • Groschel MI, Prabowo SA, Cardona PJ, Stanford JL, van der Werf TS. Therapeutic vaccines for tuberculosis–a systematic review. Vaccine 2014; 32:3162-8; PMID:24726245; http://dx.doi.org/10.1016/j.vaccine.2014.03.047
  • McShane H, Pathan AA, Sander CR, Keating SM, Gilbert SC, Huygen K, Fletcher HA, Hill AV. Recombinant modified vaccinia virus Ankara expressing antigen 85A boosts BCG-primed and naturally acquired antimycobacterial immunity in humans. Nat Med 2004; 10:1240-4; PMID:15502839; http://dx.doi.org/10.1038/nm1128
  • Carpenter C, Sidney J, Kolla R, Nayak K, Tomiyama H, Tomiyama C, Padilla OA, Rozot V, Ahamed SF, Ponte C, et al. A side-by-side comparison of T cell reactivity to fifty-nine Mycobacterium tuberculosis antigens in diverse populations from five continents. Tuberculosis (Edinb) 2015; 95(6):713-21; PMID:26277695
  • Tameris MD, Hatherill M, Landry BS, Scriba TJ, Snowden MA, Lockhart S, Shea JE, McClain JB, Hussey GD, Hanekom WA, et al. Safety and efficacy of MVA85A, a new tuberculosis vaccine, in infants previously vaccinated with BCG: a randomised, placebo-controlled phase 2b trial. Lancet 2013; 381:1021-8; PMID:23391465; http://dx.doi.org/10.1016/S0140-6736(13)60177-4
  • Goonetilleke NP, McShane H, Hannan CM, Anderson RJ, Brookes RH, Hill AV. Enhanced immunogenicity and protective efficacy against Mycobacterium tuberculosis of bacille Calmette-Guerin vaccine using mucosal administration and boosting with a recombinant modified vaccinia virus Ankara. J Immunol 2003; 171:1602-9; PMID:12874255; http://dx.doi.org/10.4049/jimmunol.171.3.1602
  • Verreck FA, Vervenne RA, Kondova I, van Kralingen KW, Remarque EJ, Braskamp G, van der Werff NM, Kersbergen A, Ottenhoff TH, Heidt PJ, et al. MVA.85A boosting of BCG and an attenuated, phoP deficient M. tuberculosis vaccine both show protective efficacy against tuberculosis in rhesus macaques. PloS One 2009; 4:e5264; PMID:19367339; http://dx.doi.org/10.1371/journal.pone.0005264
  • Vordermeier HM, Villarreal-Ramos B, Cockle PJ, McAulay M, Rhodes SG, Thacker T, Gilbert SC, McShane H, Hill AV, Xing Z, et al. Viral booster vaccines improve Mycobacterium bovis BCG-induced protection against bovine tuberculosis. Infect Immun 2009; 77:3364-73; PMID:19487476; http://dx.doi.org/10.1128/IAI.00287-09
  • Williams A, Goonetilleke NP, McShane H, Clark SO, Hatch G, Gilbert SC, Hill AV. Boosting with poxviruses enhances Mycobacterium bovis BCG efficacy against tuberculosis in guinea pigs. Infect Immun 2005; 73:3814-6; PMID:15908420; http://dx.doi.org/10.1128/IAI.73.6.3814-3816.2005
  • McShane H, Williams A. A review of preclinical animal models utilised for TB vaccine evaluation in the context of recent human efficacy data. Tuberculosis (Edinb) 2014; 94:105-10; PMID:24369986; http://dx.doi.org/10.1016/j.tube.2013.11.003
  • Beveridge NE, Price DA, Casazza JP, Pathan AA, Sander CR, Asher TE, Ambrozak DR, Precopio ML, Scheinberg P, Alder NC, et al. Immunisation with BCG and recombinant MVA85A induces long-lasting, polyfunctional Mycobacterium tuberculosis-specific CD4+ memory T lymphocyte populations. Eur J Immunol 2007; 37:3089-100; PMID:17948267; http://dx.doi.org/10.1002/eji.200737504
  • Brookes RH, Hill PC, Owiafe PK, Ibanga HB, Jeffries DJ, Donkor SA, Fletcher HA, Hammond AS, Lienhardt C, Adegbola RA, et al. Safety and immunogenicity of the candidate tuberculosis vaccine MVA85A in West Africa. PloS One 2008; 3:e2921; PMID:18698342; http://dx.doi.org/10.1371/journal.pone.0002921
  • Hawkridge T, Scriba TJ, Gelderbloem S, Smit E, Tameris M, Moyo S, Lang T, Veldsman A, Hatherill M, Merwe L, et al. Safety and immunogenicity of a new tuberculosis vaccine, MVA85A, in healthy adults in South Africa. J Infect Dis 2008; 198:544-52; PMID:18582195; http://dx.doi.org/10.1086/590185
  • Minassian AM, Rowland R, Beveridge NE, Poulton ID, Satti I, Harris S, Poyntz H, Hamill M, Griffiths K, Sander CR, et al. A Phase I study evaluating the safety and immunogenicity of MVA85A, a candidate TB vaccine, in HIV-infected adults. BMJ Open 2011; 1:e000223; PMID:22102640; http://dx.doi.org/10.1136/bmjopen-2011-000223
  • Odutola AA, Owolabi OA, Owiafe PK, McShane H, Ota MO. A new TB vaccine, MVA85A, induces durable antigen-specific responses 14 months after vaccination in African infants. Vaccine 2012; 30:5591-4; PMID:22749600; http://dx.doi.org/10.1016/j.vaccine.2012.06.054
  • Ota MO, Odutola AA, Owiafe PK, Donkor S, Owolabi OA, Brittain NJ, Williams N, Rowland-Jones S, Hill AV, Adegbola RA, et al. Immunogenicity of the tuberculosis vaccine MVA85A is reduced by coadministration with EPI vaccines in a randomized controlled trial in Gambian infants. Sci Transl Med 2011; 3:88ra56; PMID:21697532; http://dx.doi.org/10.1126/scitranslmed.3002461
  • Pathan AA, Minassian AM, Sander CR, Rowland R, Porter DW, Poulton ID, Hill AV, Fletcher HA, McShane H. Effect of vaccine dose on the safety and immunogenicity of a candidate TB vaccine, MVA85A, in BCG vaccinated UK adults. Vaccine 2012; 30:5616-24; PMID:22789508; http://dx.doi.org/10.1016/j.vaccine.2012.06.084
  • Sander CR, Pathan AA, Beveridge NE, Poulton I, Minassian A, Alder N, Van Wijgerden J, Hill AV, Gleeson FV, Davies RJ, et al. Safety and immunogenicity of a new tuberculosis vaccine, MVA85A, in Mycobacterium tuberculosis-infected individuals. Am J Respir Crit Care Med 2009; 179:724-33; PMID:19151191; http://dx.doi.org/10.1164/rccm.200809-1486OC
  • Scriba TJ, Tameris M, Mansoor N, Smit E, van der Merwe L, Isaacs F, Keyser A, Moyo S, Brittain N, Lawrie A, et al. Modified vaccinia Ankara-expressing Ag85A, a novel tuberculosis vaccine, is safe in adolescents and children, and induces polyfunctional CD4+ T cells. Eur J Immunol 2010; 40:279-90; PMID:20017188; http://dx.doi.org/10.1002/eji.200939754
  • Scriba TJ, Tameris M, Mansoor N, Smit E, van der Merwe L, Mauff K, Hughes EJ, Moyo S, Brittain N, Lawrie A, et al. Dose-finding study of the novel tuberculosis vaccine, MVA85A, in healthy BCG-vaccinated infants. J Infect Dis 2011; 203:1832-43; PMID:21606542; http://dx.doi.org/10.1093/infdis/jir195
  • Scriba TJ, Tameris M, Smit E, van der Merwe L, Hughes EJ, Kadira B, Mauff K, Moyo S, Brittain N, Lawrie A, et al. A phase IIa trial of the new tuberculosis vaccine, MVA85A, in HIV- and/or Mycobacterium tuberculosis-infected adults. Am J Respir Crit Care Med 2012; 185:769-78; PMID:22281831; http://dx.doi.org/10.1164/rccm.201108-1548OC
  • Ndiaye BP, Thienemann F, Ota M, Landry BS, Camara M, Dieye S, Dieye TN, Esmail H, Goliath R, Huygen K, et al. Safety, immunogenicity, and efficacy of the candidate tuberculosis vaccine MVA85A in healthy adults infected with HIV-1: a randomised, placebo-controlled, phase 2 trial. Lancet Respir Med 2015; 3:190-200; PMID:25726088; http://dx.doi.org/10.1016/S2213-2600(15)00037-5
  • McMurray DN. Disease model: pulmonary tuberculosis. Trends Mol Med 2001; 7:135-7; PMID:11286786; http://dx.doi.org/10.1016/S1471-4914(00)01901-8
  • Flynn JL, Gideon HP, Mattila JT, Lin PL. Immunology studies in non-human primate models of tuberculosis. Immunol Rev 2015; 264:60-73; PMID:25703552; http://dx.doi.org/10.1111/imr.12258
  • Graves AJ, Hokey DA. Tuberculosis vaccine development: Shifting focus amid increasing development challenges. Hum Vaccin Immunother 2015; 11:1910-6; PMID:26125249; http://dx.doi.org/10.1080/21645515.2015.1040955
  • Fennelly KP, Jones-Lopez EC, Ayakaka I, Kim S, Menyha H, Kirenga B, Muchwa C, Joloba M, Dryden-Peterson S, Reilly N, et al. Variability of infectious aerosols produced during coughing by patients with pulmonary tuberculosis. Am J Respir Crit Care Med 2012; 186:450-7; PMID:22798319; http://dx.doi.org/10.1164/rccm.201203-0444OC
  • Henao-Tamayo M, Shanley CA, Verma D, Zilavy A, Stapleton MC, Furney SK, Podell B, Orme IM. The Efficacy of the BCG Vaccine against Newly Emerging Clinical Strains of Mycobacterium tuberculosis. PloS One 2015; 10:e0136500; PMID:26368806; http://dx.doi.org/10.1371/journal.pone.0136500
  • Ellis RD, Hatherill M, Tait D, Snowden M, Churchyard G, Hanekom W, Evans T, Ginsberg AM. Innovative clinical trial designs to rationalize TB vaccine development. Tuberculosis (Edinb) 2015; 95:352-7; PMID:25802031; http://dx.doi.org/10.1016/j.tube.2015.02.036
  • Bennett AR, Gorak-Stolinska P, Ben-Smith A, Floyd S, de Lara CM, Weir RE, Lalor MK, Makamo K, Msiska GK, Crampin AC, et al. The PPD-specific T-cell clonal response in UK and Malawian subjects following BCG vaccination: a new repertoire evolves over 12 months. Vaccine 2006; 24:2617-26; PMID:16414159; http://dx.doi.org/10.1016/j.vaccine.2005.12.011
  • Black GF, Fine PEM, Warndorff DK, Floyd S, Weir RE, Blackwell JM, Bliss L, Sichali L, Mwaungulu L, Chaguluka S, et al. Relationship between IFN-gamma and skin test responsiveness to Mycobacterium tuberculosis PPD in healthy, non-BCG-vaccinated young adults in Northern Malawi. Int J Tuberc Lung Dis 2001; 5:664-72; PMID:11467373
  • Black GF, Weir RE, Floyd S, Bliss L, Warndorff DK, Crampin AC, Ngwira B, Sichali L, Nazareth B, Blackwell JM, et al. BCG-induced increase in interferon-gamma response to mycobacterial antigens and efficacy of BCG vaccination in Malawi and the UK: two randomised controlled studies. Lancet 2002; 359:1393-401; PMID:11978337; http://dx.doi.org/10.1016/S0140-6736(02)08353-8
  • Weir RE, Black GF, Nazareth B, Floyd S, Stenson S, Stanley C, Branson K, Sichali L, Chaguluka SD, Donovan L, et al. The influence of previous exposure to environmental mycobacteria on the interferon-gamma response to bacille Calmette-Guerin vaccination in southern England and northern Malawi. Clin Exp Immunol 2006; 146:390-9; PMID:17100757; http://dx.doi.org/10.1111/j.1365-2249.2006.03222.x
  • Mangtani P, Abubakar I, Ariti C, Beynon R, Pimpin L, Fine PE, Rodrigues LC, Smith PG, Lipman M, Whiting PF, et al. Protection by BCG vaccine against tuberculosis: a systematic review of randomized controlled trials. Clin Infect Dis 2014; 58:470-80; PMID:24336911; http://dx.doi.org/10.1093/cid/cit790
  • Elias D, Akuffo H, Britton S. Helminthes could influence the outcome of vaccines against TB in the tropics. Parasite Immunol 2006; 28:507-13; PMID:16965286; http://dx.doi.org/10.1111/j.1365-3024.2006.00854.x
  • Elias D, Akuffo H, Pawlowski A, Haile M, Schon T, Britton S. Schistosoma mansoni infection reduces the protective efficacy of BCG vaccination against virulent Mycobacterium tuberculosis. Vaccine 2005; 23:1326-34; PMID:15661380; http://dx.doi.org/10.1016/j.vaccine.2004.09.038
  • Elias D, Britton S, Aseffa A, Engers H, Akuffo H. Poor immunogenicity of BCG in helminth infected population is associated with increased in vitro TGF-β production. Vaccine 2008; 26:3897-902; PMID:18554755; http://dx.doi.org/10.1016/j.vaccine.2008.04.083
  • Elias D, Wolday D, Akuffo H, Petros B, Bronner U, Britton S. Effect of deworming on human T cell responses to mycobacterial antigens in helminth-exposed individuals before and after bacille Calmette-Guerin (BCG) vaccination. Clin Exp Immunol 2001; 123:219-25; PMID:11207651; http://dx.doi.org/10.1046/j.1365-2249.2001.01446.x
  • Biomarkers and surrogate endpoints: preferred definitions and conceptual framework. Clin Pharmacol Ther 2001; 69:89-95; PMID:11240971; http://dx.doi.org/10.1067/mcp.2001.113989
  • Weiner J, Maertzdorf J, Kaufmann SH. The dual role of biomarkers for understanding basic principles and devising novel intervention strategies in tuberculosis. Ann N Y Acad Sci 2013; 1283:22-9; PMID:23181737; http://dx.doi.org/10.1111/j.1749-6632.2012.06802.x
  • Walzl G, Ronacher K, Hanekom W, Scriba TJ, Zumla A. Immunological biomarkers of tuberculosis. Nat Rev Immunol 2011; 11:343-54; PMID:21475309; http://dx.doi.org/10.1038/nri2960
  • Al-Muhsen S, Casanova JL. The genetic heterogeneity of mendelian susceptibility to mycobacterial diseases. J Allergy Clin Immunol 2008; 122:1043-51; quiz 52-3; PMID:19084105; http://dx.doi.org/10.1016/j.jaci.2008.10.037
  • Diedrich CR, Flynn JL. HIV-1/mycobacterium tuberculosis coinfection immunology: how does HIV-1 exacerbate tuberculosis? Infect Immun 2011; 79:1407-17; PMID:21245275; http://dx.doi.org/10.1128/IAI.01126-10
  • Flynn JL, Chan J, Triebold KJ, Dalton DK, Stewart TA, Bloom BR. An essential role for interferon gamma in resistance to Mycobacterium tuberculosis infection. J Exp Med 1993; 178:2249-54; PMID:7504064; http://dx.doi.org/10.1084/jem.178.6.2249
  • Bhatt K, Verma S, Ellner JJ, Salgame P. Quest for correlates of protection against tuberculosis. Clin Vaccine Immunol 2015; 22:258-66; PMID:25589549; http://dx.doi.org/10.1128/CVI.00721-14
  • Nakaya HI, Pulendran B. Vaccinology in the era of high-throughput biology. Philos Trans R Soc Lond B Biol Sci 2015; 370; pii: 20140146; PMID:25964458; http://dx.doi.org/10.1098/rstb.2014.0146
  • Elias D, Akuffo H, Britton S. PPD induced in vitro interferon gamma production is not a reliable correlate of protection against Mycobacterium tuberculosis. Trans R Soc Trop Med Hyg 2005; 99:363-8; PMID:15780343; http://dx.doi.org/10.1016/j.trstmh.2004.08.006
  • Kagina BM, Abel B, Scriba TJ, Hughes EJ, Keyser A, Soares A, Gamieldien H, Sidibana M, Hatherill M, Gelderbloem S, et al. Specific T cell frequency and cytokine expression profile do not correlate with protection against tuberculosis after bacillus Calmette-Guerin vaccination of newborns. Am J Respir Crit Care Med 2010; 182:1073-9; PMID:20558627; http://dx.doi.org/10.1164/rccm.201003-0334OC
  • Mittrucker HW, Steinhoff U, Kohler A, Krause M, Lazar D, Mex P, Miekley D, Kaufmann SH. Poor correlation between BCG vaccination-induced T cell responses and protection against tuberculosis. Proc Natl Acad Sci U S A 2007; 104:12434-9; PMID:17640915; http://dx.doi.org/10.1073/pnas.0703510104
  • Miller E, Salisbury D, Ramsay M. Planning, registration, and implementation of an immunisation campaign against meningococcal serogroup C disease in the UK: a success story. Vaccine 2001; 20 Suppl 1:S58-67; PMID:11587814; http://dx.doi.org/10.1016/S0264-410X(01)00299-7
  • Barry CE, 3rd, Boshoff HI, Dartois V, Dick T, Ehrt S, Flynn J, Schnappinger D, Wilkinson RJ, Young D. The spectrum of latent tuberculosis: rethinking the biology and intervention strategies. Nat Rev Microbiol 2009; 7:845-55; PMID:19855401
  • O'Garra A, Redford PS, McNab FW, Bloom CI, Wilkinson RJ, Berry MP. The immune response in tuberculosis. Annu Rev Immunol 2013; 31:475-527; PMID:23516984; http://dx.doi.org/10.1146/annurev-immunol-032712-095939
  • Esmail H, Barry CE, 3rd, Wilkinson RJ. Understanding latent tuberculosis: the key to improved diagnostic and novel treatment strategies. Drug Discov Today 2012; 17:514-21; PMID:22198298; http://dx.doi.org/10.1016/j.drudis.2011.12.013
  • Esmail H, Barry CE, 3rd, Young DB, Wilkinson RJ. The ongoing challenge of latent tuberculosis. Philos Trans R Soc Lond B Biol Sci 2014; 369:20130437; PMID:24821923; http://dx.doi.org/10.1098/rstb.2013.0437
  • Mooney M, McWeeney S, Sekaly RP. Systems immunogenetics of vaccines. Semin Immunol 2013; 25:124-9; PMID:23886894; http://dx.doi.org/10.1016/j.smim.2013.06.003
  • Pulendran B, Ahmed R. Immunological mechanisms of vaccination. Nat Immunol 2011; 12:509-17; PMID:21739679; http://dx.doi.org/10.1038/ni.2039
  • Pulendran B, Li S, Nakaya HI. Systems vaccinology. Immunity 2010; 33:516-29; PMID:21029962; http://dx.doi.org/10.1016/j.immuni.2010.10.006
  • Zak DE, Aderem A. Systems biology of innate immunity. Immunol Rev 2009; 227:264-82; PMID:19120490; http://dx.doi.org/10.1111/j.1600-065X.2008.00721.x
  • Gaucher D, Therrien R, Kettaf N, Angermann BR, Boucher G, Filali-Mouhim A, Moser JM, Mehta RS, Drake DR, 3rd, Castro E, et al. Yellow fever vaccine induces integrated multilineage and polyfunctional immune responses. J Exp Med 2008; 205:3119-31; PMID:19047440; http://dx.doi.org/10.1084/jem.20082292
  • Querec TD, Akondy RS, Lee EK, Cao W, Nakaya HI, Teuwen D, Pirani A, Gernert K, Deng J, Marzolf B, et al. Systems biology approach predicts immunogenicity of the yellow fever vaccine in humans. Nat Immunol 2009; 10:116-25; PMID:19029902; http://dx.doi.org/10.1038/ni.1688
  • Maertzdorf J, Kaufmann SH, Weiner J, 3rd. Molecular signatures for vaccine development. Vaccine 2015; 33(40):5256-61
  • Nakaya HI, Li S, Pulendran B. Systems vaccinology: learning to compute the behavior of vaccine induced immunity. Wiley Interdiscip Rev Syst Biol Med 2012; 4:193-205; PMID:22012654; http://dx.doi.org/10.1002/wsbm.163
  • Nakaya HI, Wrammert J, Lee EK, Racioppi L, Marie-Kunze S, Haining WN, Means AR, Kasturi SP, Khan N, Li GM, et al. Systems biology of vaccination for seasonal influenza in humans. Nat Immunol 2011; 12:786-95; PMID:21743478; http://dx.doi.org/10.1038/ni.2067
  • Ulrichs T, Kaufmann SH. New insights into the function of granulomas in human tuberculosis. J Pathol 2006; 208:261-9; PMID:16362982; http://dx.doi.org/10.1002/path.1906
  • Berry MP, Graham CM, McNab FW, Xu Z, Bloch SA, Oni T, Wilkinson KA, Banchereau R, Skinner J, Wilkinson RJ, et al. An interferon-inducible neutrophil-driven blood transcriptional signature in human tuberculosis. Nature 2010; 466:973-7; PMID:20725040; http://dx.doi.org/10.1038/nature09247
  • Jacobsen M, Mattow J, Repsilber D, Kaufmann SH. Novel strategies to identify biomarkers in tuberculosis. Biol Chem 2008; 389:487-95; PMID:18953715; http://dx.doi.org/10.1515/BC.2008.053
  • Jacobsen M, Repsilber D, Gutschmidt A, Neher A, Feldmann K, Mollenkopf HJ, Ziegler A, Kaufmann SH. Candidate biomarkers for discrimination between infection and disease caused by Mycobacterium tuberculosis. J Mol Med (Berl) 2007; 85:613-21; PMID:17318616; http://dx.doi.org/10.1007/s00109-007-0157-6
  • Lesho E, Forestiero FJ, Hirata MH, Hirata RD, Cecon L, Melo FF, Paik SH, Murata Y, Ferguson EW, Wang Z, et al. Transcriptional responses of host peripheral blood cells to tuberculosis infection. Tuberculosis (Edinb) 2011; 91:390-9; PMID:21835698; http://dx.doi.org/10.1016/j.tube.2011.07.002
  • Lu C, Wu J, Wang H, Wang S, Diao N, Wang F, Gao Y, Chen J, Shao L, Weng X, et al. Novel biomarkers distinguishing active tuberculosis from latent infection identified by gene expression profile of peripheral blood mononuclear cells. PloS One 2011; 6:e24290; PMID:21904626; http://dx.doi.org/10.1371/journal.pone.0024290
  • Maertzdorf J, Ota M, Repsilber D, Mollenkopf HJ, Weiner J, Hill PC, Kaufmann SH. Functional correlations of pathogenesis-driven gene expression signatures in tuberculosis. PloS One 2011; 6:e26938; PMID:22046420; http://dx.doi.org/10.1371/journal.pone.0026938
  • Maertzdorf J, Repsilber D, Parida SK, Stanley K, Roberts T, Black G, Walzl G, Kaufmann SH. Human gene expression profiles of susceptibility and resistance in tuberculosis. Genes Immun 2011; 12:15-22; PMID:20861863; http://dx.doi.org/10.1038/gene.2010.51
  • Maertzdorf J, Weiner J, 3rd, Mollenkopf HJ, Bauer T, Prasse A, Muller-Quernheim J, Kaufmann SH. Common patterns and disease-related signatures in tuberculosis and sarcoidosis. Proc Natl Acad Sci U S A 2012; 109:7853-8; PMID:22547807; http://dx.doi.org/10.1073/pnas.1121072109
  • Mistry R, Cliff JM, Clayton CL, Beyers N, Mohamed YS, Wilson PA, Dockrell HM, Wallace DM, van Helden PD, Duncan K, et al. Gene-expression patterns in whole blood identify subjects at risk for recurrent tuberculosis. J Infect Dis 2007; 195:357-65; PMID:17205474; http://dx.doi.org/10.1086/510397
  • Sutherland JS, Loxton AG, Haks MC, Kassa D, Ambrose L, Lee JS, Ran L, van Baarle D, Maertzdorf J, Howe R, et al. Differential gene expression of activating Fcgamma receptor classifies active tuberculosis regardless of human immunodeficiency virus status or ethnicity. Clin Microbiol Infect 2014; 20:O230-8; PMID:24205913; http://dx.doi.org/10.1111/1469-0691.12383
  • Kaforou M, Wright VJ, Oni T, French N, Anderson ST, Bangani N, Banwell CM, Brent AJ, Crampin AC, Dockrell HM, et al. Detection of tuberculosis in HIV-infected and -uninfected African adults using whole blood RNA expression signatures: a case-control study. PLoS Med 2013; 10:e1001538; PMID:24167453; http://dx.doi.org/10.1371/journal.pmed.1001538
  • Cliff JM, Lee JS, Constantinou N, Cho JE, Clark TG, Ronacher K, King EC, Lukey PT, Duncan K, Van Helden PD, et al. Distinct phases of blood gene expression pattern through tuberculosis treatment reflect modulation of the humoral immune response. J Infect Dis 2013; 207:18-29; PMID:22872737; http://dx.doi.org/10.1093/infdis/jis499
  • Joosten SA, Fletcher HA, Ottenhoff TH. A helicopter perspective on TB biomarkers: pathway and process based analysis of gene expression data provides new insight into TB pathogenesis. PloS One 2013; 8:e73230; PMID:24066041; http://dx.doi.org/10.1371/journal.pone.0073230
  • Maertzdorf J, Weiner J, 3rd, Kaufmann SH. Enabling biomarkers for tuberculosis control. Int J Tuberc Lung Dis 2012; 16:1140-8; PMID:22871324; http://dx.doi.org/10.5588/ijtld.12.0246
  • Fu Y, Yi Z, Wu X, Li J, Xu F. Circulating microRNAs in patients with active pulmonary tuberculosis. J Clin Microbiol 2011; 49:4246-51; PMID:21998423; http://dx.doi.org/10.1128/JCM.05459-11
  • Iannaccone M, Dorhoi A, Kaufmann SH. Host-directed therapy of tuberculosis: what is in it for microRNA? Expert Opin Ther Targets 2014; 18:491-4; PMID:24641181; http://dx.doi.org/10.1517/14728222.2014.897696
  • Liu Y, Wang X, Jiang J, Cao Z, Yang B, Cheng X. Modulation of T cell cytokine production by miR-144* with elevated expression in patients with pulmonary tuberculosis. Mol Immunol 2011; 48:1084-90; PMID:21367459; http://dx.doi.org/10.1016/j.molimm.2011.02.001
  • Wang C, Yang S, Sun G, Tang X, Lu S, Neyrolles O, Gao Q. Comparative miRNA expression profiles in individuals with latent and active tuberculosis. PloS One 2011; 6:e25832; PMID:22003408; http://dx.doi.org/10.1371/journal.pone.0025832
  • Wang Z, Gerstein M, Snyder M. RNA-Seq: a revolutionary tool for transcriptomics. Nat Rev Genet 2009; 10:57-63; PMID:19015660; http://dx.doi.org/10.1038/nrg2484
  • Wu J, Lu C, Diao N, Zhang S, Wang S, Wang F, Gao Y, Chen J, Shao L, Lu J, et al. Analysis of microRNA expression profiling identifies miR-155 and miR-155* as potential diagnostic markers for active tuberculosis: a preliminary study. Hum Immunol 2012; 73:31-7; PMID:22037148; http://dx.doi.org/10.1016/j.humimm.2011.10.003
  • Wu LS, Lee SW, Huang KY, Lee TY, Hsu PW, Weng JT. Systematic expression profiling analysis identifies specific microRNA-gene interactions that may differentiate between active and latent tuberculosis infection. Biomed Res Int 2014; 2014:895179; PMID:25276827
  • Agranoff D, Fernandez-Reyes D, Papadopoulos MC, Rojas SA, Herbster M, Loosemore A, Tarelli E, Sheldon J, Schwenk A, Pollok R, et al. Identification of diagnostic markers for tuberculosis by proteomic fingerprinting of serum. Lancet 2006; 368:1012-21; PMID:16980117; http://dx.doi.org/10.1016/S0140-6736(06)69342-2
  • Deng C, Lin M, Hu C, Li Y, Gao Y, Cheng X, Zhang F, Dong M, Li Y. Establishing a serologic decision tree model of extrapulmonary tuberculosis by MALDI-TOF MS analysis. Diagn Microbiol Infect Dis 2011; 71:144-50; PMID:21855247; http://dx.doi.org/10.1016/j.diagmicrobio.2011.06.021
  • Kunnath-Velayudhan S, Salamon H, Wang HY, Davidow AL, Molina DM, Huynh VT, Cirillo DM, Michel G, Talbot EA, Perkins MD, et al. Dynamic antibody responses to the Mycobacterium tuberculosis proteome. Proc Natl Acad Sci U S A 2010; 107:14703-8; PMID:20668240; http://dx.doi.org/10.1073/pnas.1009080107
  • Liu Q, Chen X, Hu C, Zhang R, Yue J, Wu G, Li X, Wu Y, Wen F. Serum protein profiling of smear-positive and smear-negative pulmonary tuberculosis using SELDI-TOF mass spectrometry. Lung 2010; 188:15-23; PMID:20012079; http://dx.doi.org/10.1007/s00408-009-9199-6
  • Sartain MJ, Slayden RA, Singh KK, Laal S, Belisle JT. Disease state differentiation and identification of tuberculosis biomarkers via native antigen array profiling. Mol Cell Proteomics 2006; 5:2102-13; http://dx.doi.org/10.1074/mcp.M600089-MCP200
  • Yu Y, Jin D, Hu S, Zhang Y, Zheng X, Zheng J, Liao M, Chen X, Graner M, Liu H, et al. A novel tuberculosis antigen identified from human tuberculosis granulomas. Mol Cell Proteomics 2015; 14:1093-103; http://dx.doi.org/10.1074/mcp.M114.045237
  • Banday KM, Pasikanti KK, Chan EC, Singla R, Rao KV, Chauhan VS, Nanda RK. Use of urine volatile organic compounds to discriminate tuberculosis patients from healthy subjects. Anal Chem 2011; 83:5526-34; PMID:21619052; http://dx.doi.org/10.1021/ac200265g
  • Mahapatra S, Hess AM, Johnson JL, Eisenach KD, DeGroote MA, Gitta P, Joloba ML, Kaplan G, Walzl G, Boom WH, et al. A metabolic biosignature of early response to anti-tuberculosis treatment. BMC Infect Dis 2014; 14:53; PMID:24484441; http://dx.doi.org/10.1186/1471-2334-14-53
  • Phillips M, Basa-Dalay V, Bothamley G, Cataneo RN, Lam PK, Natividad MP, Schmitt P, Wai J. Breath biomarkers of active pulmonary tuberculosis. Tuberculosis (Edinb) 2010; 90:145-51; PMID:20189456; http://dx.doi.org/10.1016/j.tube.2010.01.003
  • Weiner J, 3rd, Parida SK, Maertzdorf J, Black GF, Repsilber D, Telaar A, Mohney RP, Arndt-Sullivan C, Ganoza CA, Fae KC, et al. Biomarkers of inflammation, immunosuppression and stress with active disease are revealed by metabolomic profiling of tuberculosis patients. PloS one 2012; 7:e40221; PMID:22844400; http://dx.doi.org/10.1371/journal.pone.0040221
  • Tientcheu LD, Maertzdorf J, Weiner J, Adetifa IM, Mollenkopf HJ, Sutherland JS, Donkor S, Kampmann B, Kaufmann SH, Dockrell HM, et al. Differential transcriptomic and metabolic profiles of M. africanum- and M. tuberculosis-infected patients after, but not before, drug treatment. Genes Immu 2015; 16:347-55; PMID:26043170; http://dx.doi.org/10.1038/gene.2015.21
  • Weiner J, 3rd, Kaufmann SH, Maertzdorf J. High-throughput data analysis and data integration for vaccine trials. Vaccine 2015; 33(40):5249-55
  • Chaussabel D, Quinn C, Shen J, Patel P, Glaser C, Baldwin N, Stichweh D, Blankenship D, Li L, Munagala I, et al. A modular analysis framework for blood genomics studies: application to systemic lupus erythematosus. Immunity 2008; 29:150-64; PMID:18631455; http://dx.doi.org/10.1016/j.immuni.2008.05.012
  • Banchereau R, Jordan-Villegas A, Ardura M, Mejias A, Baldwin N, Xu H, Saye E, Rossello-Urgell J, Nguyen P, Blankenship D, et al. Host immune transcriptional profiles reflect the variability in clinical disease manifestations in patients with Staphylococcus aureus infections. PloS One 2012; 7:e34390; PMID:22496797; http://dx.doi.org/10.1371/journal.pone.0034390
  • Chiche L, Jourde-Chiche N, Whalen E, Presnell S, Gersuk V, Dang K, Anguiano E, Quinn C, Burtey S, Berland Y, et al. Modular transcriptional repertoire analyses of adults with systemic lupus erythematosus reveal distinct type I and type II interferon signatures. Arthritis Rheumatol 2014; 66:1583-95; http://dx.doi.org/10.1002/art.38628
  • Obermoser G, Presnell S, Domico K, Xu H, Wang Y, Anguiano E, Thompson-Snipes L, Ranganathan R, Zeitner B, Bjork A, et al. Systems scale interactive exploration reveals quantitative and qualitative differences in response to influenza and pneumococcal vaccines. Immunity 2013; 38:831-44; PMID:23601689; http://dx.doi.org/10.1016/j.immuni.2012.12.008
  • Dunachie S, Hill AV, Fletcher HA. Profiling the host response to malaria vaccination and malaria challenge. Vaccine 2015; 33(40):5316-20
  • Huang KY, Li CK, Clutterbuck E, Chui C, Wilkinson T, Gilbert A, Oxford J, Lambkin-Williams R, Lin TY, McMichael AJ, et al. Virus-specific antibody secreting cell, memory B-cell, and sero-antibody responses in the human influenza challenge model. J Infect Dis 2014; 209:1354-61; PMID:24415790; http://dx.doi.org/10.1093/infdis/jit650
  • Jones C, Darton TC, Pollard AJ. Why the development of effective typhoid control measures requires the use of human challenge studies. Front Microbiol 2014; 5:707; PMID:25566221; http://dx.doi.org/10.3389/fmicb.2014.00707
  • McArthur MA, Fresnay S, Magder LS, Darton TC, Jones C, Waddington CS, Blohmke CJ, Dougan G, Angus B, Levine MM, et al. Activation of Salmonella Typhi-specific regulatory T cells in typhoid disease in a wild-type S. Typhi challenge model. PLoS Pathog 2015; 11:e1004914; PMID:26001081; http://dx.doi.org/10.1371/journal.ppat.1004914
  • Memoli MJ, Czajkowski L, Reed S, Athota R, Bristol T, Proudfoot K, Fargis S, Stein M, Dunfee RL, Shaw PA, et al. Validation of the wild-type influenza A human challenge model H1N1pdMIST: an A(H1N1)pdm09 dose-finding investigational new drug study. Clin Infect Dis 2015; 60:693-702; PMID:25416753; http://dx.doi.org/10.1093/cid/ciu924
  • Shirley DA, McArthur MA. The utility of human challenge studies in vaccine development: lessons learned from cholera. Vaccine 2011; 2011:3-13; PMID:24482781
  • Spring M, Polhemus M, Ockenhouse C. Controlled human malaria infection. J Infect Dis 2014; 209 Suppl 2:S40-5; PMID:24872394; http://dx.doi.org/10.1093/infdis/jiu063
  • Gunther VJ, Putnak R, Eckels KH, Mammen MP, Scherer JM, Lyons A, Sztein MB, Sun W. A human challenge model for dengue infection reveals a possible protective role for sustained interferon gamma levels during the acute phase of illness. Vaccine 2011; 29:3895-904; PMID:21443963; http://dx.doi.org/10.1016/j.vaccine.2011.03.038
  • Garnier T, Eiglmeier K, Camus JC, Medina N, Mansoor H, Pryor M, Duthoy S, Grondin S, Lacroix C, Monsempe C, et al. The complete genome sequence of Mycobacterium bovis. Proc Natl Acad Sci U S A 2003; 100:7877-82; PMID:12788972; http://dx.doi.org/10.1073/pnas.1130426100
  • Minassian AM, Ronan EO, Poyntz H, Hill AV, McShane H. Preclinical development of an in vivo BCG challenge model for testing candidate TB vaccine efficacy. PloS One 2011; 6:e19840; PMID:21629699; http://dx.doi.org/10.1371/journal.pone.0019840
  • Villarreal-Ramos B, Berg S, Chamberlain L, McShane H, Hewinson RG, Clifford D, Vordermeier M. Development of a BCG challenge model for the testing of vaccine candidates against tuberculosis in cattle. Vaccine 2014; 32:5645-9; PMID:25138291; http://dx.doi.org/10.1016/j.vaccine.2014.08.009
  • Chen L, Wang J, Zganiacz A, Xing Z. Single intranasal mucosal Mycobacterium bovis BCG vaccination confers improved protection compared to subcutaneous vaccination against pulmonary tuberculosis. Infect Immun 2004; 72:238-46; PMID:14688101; http://dx.doi.org/10.1128/IAI.72.1.238-246.2004
  • Jeon BY, Derrick SC, Lim J, Kolibab K, Dheenadhayalan V, Yang AL, Kreiswirth B, Morris SL. Mycobacterium bovis BCG immunization induces protective immunity against nine different Mycobacterium tuberculosis strains in mice. Infect Immun 2008; 76:5173-80; PMID:18710860; http://dx.doi.org/10.1128/IAI.00019-08
  • Santosuosso M, McCormick S, Zhang X, Zganiacz A, Xing Z. Intranasal boosting with an adenovirus-vectored vaccine markedly enhances protection by parenteral Mycobacterium bovis BCG immunization against pulmonary tuberculosis. Infect Immun 2006; 74:4634-43; PMID:16861651; http://dx.doi.org/10.1128/IAI.00517-06
  • Minassian AM, Satti I, Poulton ID, Meyer J, Hill AV, McShane H. A human challenge model for Mycobacterium tuberculosis using Mycobacterium bovis bacille Calmette-Guerin. J Infect Dis 2012; 205:1035-42; PMID:22396610; http://dx.doi.org/10.1093/infdis/jis012
  • Harris SA, Meyer J, Satti I, Marsay L, Poulton ID, Tanner R, Minassian AM, Fletcher HA, McShane H. Evaluation of a human BCG challenge model to assess antimycobacterial immunity induced by BCG and a candidate tuberculosis vaccine, MVA85A, alone and in combination. J Infect Dis 2014; 209:1259-68; PMID:24273174; http://dx.doi.org/10.1093/infdis/jit647
  • Matsumiya M, Satti I, Chomka A, Harris SA, Stockdale L, Meyer J, Fletcher HA, McShane H. Gene expression and cytokine profile correlate with mycobacterial growth in a human BCG challenge model. J Infect Dis 2015; 211:1499-509; PMID:25381367; http://dx.doi.org/10.1093/infdis/jiu615
  • Satti I, Meyer J, Harris SA, Manjaly Thomas ZR, Griffiths K, Antrobus RD, Rowland R, Ramon RL, Smith M, Sheehan S, et al. Safety and immunogenicity of a candidate tuberculosis vaccine MVA85A delivered by aerosol in BCG-vaccinated healthy adults: a phase 1, double-blind, randomised controlled trial. Lancet Infect Dis 2014; 14:939-46; PMID:25151225; http://dx.doi.org/10.1016/S1473-3099(14)70845-X
  • Mahomed H, Ehrlich R, Hawkridge T, Hatherill M, Geiter L, Kafaar F, Abrahams DA, Mulenga H, Tameris M, Geldenhuys H, et al. TB incidence in an adolescent cohort in South Africa. PloS One 2013; 8:e59652; PMID:23533639; http://dx.doi.org/10.1371/journal.pone.0059652
  • Mahomed H, Hawkridge T, Verver S, Geiter L, Hatherill M, Abrahams DA, Ehrlich R, Hanekom WA, Hussey GD. Predictive factors for latent tuberculosis infection among adolescents in a high-burden area in South Africa. Int J Tuberc Lung Dis 2011; 15:331-6; PMID:21333099
  • Andrews JR, Hatherill M, Mahomed H, Hanekom WA, Campo M, Hawn TR, Wood R, Scriba TJ. The dynamics of QuantiFERON-TB gold in-tube conversion and reversion in a cohort of South African adolescents. Am J Respir Crit Care Med 2015; 191:584-91; PMID:25562578; http://dx.doi.org/10.1164/rccm.201409-1704OC
  • Machingaidze S, Verver S, Mulenga H, Abrahams DA, Hatherill M, Hanekom W, Hussey GD, Mahomed H. Predictive value of recent QuantiFERON conversion for tuberculosis disease in adolescents. Am J Respir Crit Care Med 2012; 186:1051-6; PMID:22955316; http://dx.doi.org/10.1164/rccm.201206-1134OC
  • Mahomed H, Hawkridge T, Verver S, Abrahams D, Geiter L, Hatherill M, Ehrlich R, Hanekom WA, Hussey GD. The tuberculin skin test versus QuantiFERON TB Gold(R) in predicting tuberculosis disease in an adolescent cohort study in South Africa. PloS One 2011; 6:e17984; PMID:21479236; http://dx.doi.org/10.1371/journal.pone.0017984