Luke D. Jasenosky Thomas J. Scriba Willem A. Hanekom Anne E. Goldfeld

T cells and adaptive immunity to Mycobacterium tuberculosis in humans

Authors’ addresses Luke D. Jasenosky1,*, Thomas J. Scriba2,*, Willem A. Hanekom2,3, Anne E. Goldfeld1 1 Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA. 2 South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine & School of Child and Adolescent Health, University of Cape Town, Cape Town, South Africa. 3 Bill and Melinda Gates Foundation, Seattle, WA, USA.

Summary: The adaptive immune response mediated by T cells is critical for control of Mycobacterium tuberculosis (M. tuberculosis) infection in humans. However, the M. tuberculosis antigens and host T-cell responses that are required for an effective adaptive immune response to M. tuberculosis infection are yet to be defined. Here, we review recent findings on CD4+ and CD8+ T-cell responses to M. tuberculosis infection and examine the roles of distinct M. tuberculosis-specific T-cell subsets in control of de novo and latent M. tuberculosis infection, and in the evolution of T-cell immunity to M. tuberculosis in response to tuberculosis treatment. In addition, we discuss recent studies that elucidate aspects of M. tuberculosis-specific adaptive immunity during human immunodeficiency virus co-infection and summarize recent findings from vaccine trials that provide insight into effective adaptive immune responses to M. tuberculosis infection.

*These authors contributed equally to the writing of this article. Correspondence to: Anne E. Goldfeld Program in Cellular and Molecular Medicine Children’s Hospital Boston 200 Longwood Ave. Boston, MA 02115, USA Tel.: +1 617 713 8778 e-mail: [email protected] Acknowledgements The authors have no conflicts of interest to declare.

This article is part of a series of reviews covering Tuberculosis appearing in Volume 264 of Immunological Reviews. Video podcast available Go to www.immunologicalreviews.com to watch an interview with Guest Editor Carl Nathan.

Immunological Reviews 2015 Vol. 264: 74–87

© 2015 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd

Immunological Reviews

Keywords: TB, CD4+, CD8+, IL-10-producing immunosuppressive and Treg cells, Th1 and Th17 cells, HIV co-infection

T-cell responses and detection of M. tuberculosis infection In humans, the adaptive immune responses to Mycobacterium tuberculosis (M. tuberculosis), which become detectable 3–8 weeks after infection and play a critical role in the ultimate outcome of M. tuberculosis infection, are primarily comprised of antigenspecific CD4+ T-cell responses. In the majority of M. tuberculosis-infected persons, a CD4+ T-cell-driven type IV delayed hypersensitivity reaction that is characterized by visible skin induration can be detected 24–72 h after intradermal administration of M. tuberculosis purified protein derivative (PPD), or tuberculin [tuberculin skin test (TST)] (reviewed in 1). Careful clinical observation of individuals exposed to persons with infectious TB suggests that this immunological sensitivity to PPD frequently coincides with fever and erythema nodosum (2). Through measurement of IFN-c release assay (IGRA) positivity, which is currently used as a proxy for latent M. tuberculosis infection, and based on the prevalence of persons with immunological sensitization to PPD, it has been estimated that more than 2 billion persons globally may be infected with

0105-2896

74

© 2015 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd Immunological Reviews 264/2015

Jasenosky et al  T cells and the human immune response to TB

M. tuberculosis (3). Although one can argue that sensitization is dependent on some level of exposure to M. tuberculosis or, as is more likely, infection with M. tuberculosis, the nature of immunological memory suggests that persons with self-cured disease, aborted infection, or sterilization may also retain antigen-specific immune responses (4) (see below). It thus has been argued that the real prevalence of persons with persistent M. tuberculosis infection is likely lower than typically reported (5). On the other hand, antigen-specific anergy as measured by lack of TST responses in otherwise immunocompetent TB patients has been well-documented in several populations of distinct ethnicity and can confound estimates of TB prevalence and TB diagnosis itself. For example, the Yanomami indigenous population of Brazil was shown to have a high rate of anergy (approximately 46%) to PPD by TST skin response (75% of the total CD4+ T-cell recall response in patients with latent M. tuberculosis infection, >80% of the circulating CD4+ T cells that recognized these epitopes were CXCR3+ CCR6+ CD4+ T cells (87). Furthermore, isolated CXCR3+ CCR6+ CD4+ T cells from patients with latent M. tuberculosis infection proliferated strongly upon stimulation with M. tuberculosis antigens when compared to CXCR3 CCR6+ and CCR6 CD4+ T cells that were stimulated with the same antigens. Notably, nearly half of these epitopes were derived from M. tuberculosis proteins that had not previously been identified as antigens (87). These findings indicate that a distinct Th1/Th17-like subset that can respond potently to a relatively restricted assortment of M. tuberculosis antigens dominates the CD4+ Tcell memory compartment in latently infected individuals. Future studies are necessary to validate these findings in different geographic areas, evaluate their association with M. tuberculosis disease risk patterns, and determine their role in limiting breakthrough of M. tuberculosis from latency. M. tuberculosis-specific CD8+ T cells during natural infection Although less studied, changes in CD8+ T-cell function during active TB have also been described. Both circulating M. tuberculosis-specific CD8+ T cells (88) and CD8+ T cells located at sites of active TB disease (89), show reduced cytotoxic activity compared to those found in healthy, latently infected patients. Although a recent report found enrichment of IFN-c+ TNF+ IL-2+ trifunctional M. tuberculosis-specific CD69+ CD8+ T cells in pleural fluid spaces of TB patients (90), an earlier study reported that these cells were reduced in patients with active TB versus persons with latent infection and that high M. tuberculosis antigen burden appeared to exacerbate CD8+ T-cell dysfunction (65). Terminally differentiated CD45RA+ CCR7 CD8+ T cells with potent antimycobacterial capacity have also been

77

Jasenosky et al  T cells and the human immune response to TB

described and appear to be reduced in active TB patients compared to persons with latent M. tuberculosis infection, and this phenotype corresponded with diminished IFN-c+ IL2+ CD8+ T-cell percentages (91). In addition, M. tuberculosisspecific CD8+ T cells from South African patients with active TB were primarily CD45RA+ CCR7+ (92). In this latter study, senescent and pro-apoptotic markers were upregulated on M. tuberculosis-specific CD8+ T cells to a greater extent in patients with active TB as compared to those who had latent infection. Furthermore, in another report proliferative deficiencies persisted in M. tuberculosis-specific CD8+ T cells after TB therapy (93). These data thus suggest that, during active TB disease, the M. tuberculosis-specific CD8+ T-cell population is arrested at an intermediate differentiation point characterized by both reduced proliferative potential and reduced cytoxic activity. Notably, infliximab anti-TNF treatment in patients with autoimmune disorders who also had latent TB infection led to a reduction in, the numbers of terminally differentiated, TNF-dependent, CD45RA+ CD8+ T cells with potent antimycobacterial capacity (94). It is thus intriguing to speculate that prophylactic and/or early TB treatment may limit CD8+ T-cell effector dysfunction, and promote both improved control of latent infection and reduce the risk of TB re-infection. The role of IL-17-producing cells in natural M. tuberculosis infection Over the past decade, the role of IL-17-producing (Th17) cells in M. tuberculosis infection has been described in several studies in the murine TB model. IL-23 drives differentiation and functionality of Th17 cells (95), and an early report in mice found that knockout of the IL-23 subunit p19, which promotes the production of IL-17, resulted in almost complete loss of M. tuberculosis-specific Th17 cells, but intriguingly had little effect on infection control, which was driven by Th1-derived IFN-c (96). Later murine studies found that, early after infection, IL-17 is primarily produced by cd T cells (97, 98), and implicated as a positive player in granuloma formation in response to high dose intratracheal infection (97, 99). Dysregulated IL-17 production in the murine lung led to immunopathology driven by excess neutrophil recruitment (100, 101); however, IL-17 signaling appears to be critical for proper neutrophil targeting to the infection site early after infection for control of M. tuberculosis in murine models (102). Recently, it has been demonstrated that IL-17 / mice are unable to control infection by the hypervirulent M. tuberculosis strain HN878 but survive infection by less pathogenic

78

strains. This outcome was associated with reduced T-cell recruitment to lymphoid follicles in the lung in the absence of IL-17 (103). In humans, most studies have found that ex vivo stimulation of whole blood or PBMC from individuals with latent TB infection as compared to active TB patients leads to greater IL17 production and/or increased frequencies of IL-17+ CD4+ T cells (104, 105, 106, 107, 108, 109, 110). One report, however, found higher IL-17 release in response to PPD stimulation in whole blood of active TB patients versus those with latent infection (111). Longer durations of ex vivo stimulation appear to enhance the ability to detect IL-17 synthesis (104, 111, 112), and it is has been reported recently that M. tuberculosis-induced expansion of PBMC from latent TB patients leads to the gradual accumulation of IFN-c+/IL-17+ CD4+ T cells, and the surface expression of CXCR3 and CCR6 (104). This observation suggested that the capacity to produce IL-17 is delayed in comparison to IFN-c in M. tuberculosis-specific memory CD4+ T cells that are circulating in the periphery of latent TB patients. Intriguingly, increased proportions of IFN-c+ IL-17+ CD4+ T cells were found after ex vivo stimulation with M. tuberculosis lysate in patients with more severe TB disease (113). The latter finding suggested that greater bacterial burden and/or extrapulmonary M. tuberculosis growth may lead to increased recirculation of IL-17+ CD4+ T cells, which are normally sequestered at the infection site, leading to reduced presence in the peripheral blood (107). As has been reported in mice, IL-17+ c-d T cells have also been found to be increased in human TB patients (114). Notably, the M. tuberculosis antigen HBHA can also activate Th17 cells; HBHA stimulation ex vivo induced activation of a greater frequency of CD4+ T cells co-expressing IFN-c, IL-2, and IL-17 in household contacts as compared to active TB patients (115). In summary, Th17 and other IL-17-producing cells appear to contribute to M. tuberculosis control, most likely at an early stage of infection, and differences in pathogenic M. tuberculosis strains may play a role in differential Th17 responses observed in humans. The role of immunosuppressive IL-10-producing CD4+ T cells and FoxP3+ CD4+ Tregs during natural infection As described above, a link between immunosuppressive CD4+ T-regulatory cells (Tregs) and reduced immunity against M. tuberculosis was first described through the identification of IL-10-producing Tregs in otherwise immunocompetent Cambodian patients with active TB who showed anergy to PPD by skin test (7). T cells from these patients produced high levels of IL-10 in response to stimulation by © 2015 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd Immunological Reviews 264/2015

Jasenosky et al  T cells and the human immune response to TB

autologous PPD-loaded APCs, which inhibited T-cell proliferation in a contact-dependent and -independent manner. By contrast, T cells from PPD-responsive TB patients produced both IFN-c and IL-10 and proliferated. Blocking IL-10 resulted in restoration of proliferation of T cells from anergic patients (7). Anergy to PPD in a portion of the anergic patients was transient. PPD responsiveness was regained in this group of patients and was reflected in regaining a T cell proliferative response to PPD in in vitro studies. Strikingly, a subset of patients remained persistently anergic to PPD by TST but did respond with a positive skin test to Candida antigen. Furthermore, PBMC from these persistently PPD-anergic patients in vitro retained their inability to respond to Candida antigen stimulation with IFN-c production and proliferation (8). The subsequent identification of natural CD4+ regulatory Tregs, which express FoxP3 and can suppress effector responses in a contact-dependent and IL-10 contact-independent manners (116–118), led to further work aimed at finding additional immunosuppressive T-cell populations in TB patients. CD4+ Treg frequencies were shown to be increased in the peripheral blood and at disease sites of patients with active TB and these cells could suppress M. tuberculosis-specific IFN-c production by Th1 cells ex vivo (112, 119–123). A study of granulomatous lesions in children with TB found that FoxP3+ CD4+ Treg cells were present in high numbers and that their quantity correlated inversely with CD8+ T-cell numbers at infection sites (124). At the same time, studies in mice found that CD4+ Tregs prevented M. tuberculosis bacterial control (125–127), and M. tuberculosis-specific murine CD4+ Tregs were shown to expand alongside the expansion of M. tuberculosis-specific Th1 cells in pulmonary lymph nodes, leading to reduced effector CD4+ and CD8+ T-cell recruitment to the lung during early M. tuberculosis infection (128). In macaques, redistribution of CD4+ Tregs from the peripheral blood to the lung early after M. tuberculosis infection was shown to be associated with subsequent elevation of CD4+ Tregs in the circulation (129). In a second macaque study, simultaneous IL-2-mediated expansion of M. tuberculosis-specific Th1 cells and CD4+ Tregs was detected during early TB infection (130). A population of FoxP3+ Tregs in humans co-expresses the surface ectonucleosidase CD39, which metabolizes pro-inflammatory extracellular ATP (131). These cells are CCR6+ CD45RO+, indicating that they are antigen-experienced (131). CD39+ Tregs were found to expand disproportionately in response to M. tuberculosis-specific stimulation of PBMCs from active TB patients and to © 2015 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd Immunological Reviews 264/2015

potently suppress M. tuberculosis-specific IFN-c production (132). Furthermore, it was recently demonstrated that CD39+ Tregs are enriched in the M. tuberculosis-specific memory CD4+ T-cell compartment of active TB patients compared to persons with latent M. tuberculosis, and these cells promoted IL-10 production in PBMC cultures stimulated with M. tuberculosis antigens (133). Intriguingly, a direct correlation between CD39+ CD4+ Tregs and M. tuberculosis-specific Th1 and Th17 responses was demonstrated in humans immunized with the experimental TB vaccine MVA-85A, as a proportional decrease in this cellular subpopulation within PBMCs coincided with a peak in antigen-specific IL-17 and IFN-c production by CD4+ T cells (134, 135). A major takeaway from these findings is that CD4+ Tregs contribute to impaired M. tuberculosis control/clearance as well as reduced inflammation at infection sites. Moreover, IL-10 derived from Tregs or other cell types (see section about type I interferon responses below) is linked to poor immunity to M. tuberculosis. As a potent suppressor of macrophage activation, especially activation by IFN-c, excess IL-10 production by T-cell subsets may impair pulmonary macrophage killing of engulfed M. tuberculosis and lead to reduced antigen-presenting capacity and diminished production of IL-1a/b, IL-12, and TNF by myeloid cells, with concomitant impairment of M. tuberculosis-specific CD4+ and CD8+ T-cell activation and proliferation (136–141). Thus, limited expansion of CD4+ Tregs and other immunosuppressive T-cell subsets may contribute to an optimal response to M. tuberculosis characterized by modulated inflammation in the first weeks after infection is established (142, 143), but too much Treg expansion leads to impaired TB immunity. Inflammation and TB Active TB is characterized by inflammation and the upregulation of acute phase proteins, cytokines, chemokines, and matrix metalloproteinases (MMPs) (144–146), which in turn can affect T-cell function and development in the blood, sputum, and at sites of disease. Cavitation, which is a hallmark of adult pulmonary TB disease, is driven by elevated expression of inflammatory matrix metalloproteinase-1 (MMP-1) and MMP-3, which are found at greater levels in sputum and bronchoalveolar lavage fluid from patients with active TB (147). Whole blood transcriptomic analyses have also been applied to investigate human inflammatory responses in M. tuberculosis infection and TB disease. Several reports have described strong upregulation of genes

79

Jasenosky et al  T cells and the human immune response to TB

associated with type I/II interferon responses, inflammation, and myeloid cell activation, and the downregulation of lymphoid cell genes, in blood from TB patients compared with latently M. tuberculosis-infected controls (148–153). An inflammatory signature appears to also occur in children with TB, and a transcriptomic signature consisting largely of type I/II interferon genes has recently been applied as a diagnostic test for TB in children (154). Strong type I IFN production during M. tuberculosis infection is detrimental in mice. For example, mice lacking the common type I IFN receptor exhibit better control of M. tuberculosis infection as compared to wild type controls (155–158). Furthermore, agonistic activation of type I IFN synthesis in the lung impaired M. tuberculosis immunity and control of mycobacterial burden (159). It has been reported that type I IFNs drive enhanced neutrophil recruitment to the lung in hypersusceptible 129S2 mice, leading to local tissue damage and increased M. tuberculosis dissemination (160). Type I IFN responses that were experimentally induced by influenza infection also resulted in greater susceptibility to subsequent M. tuberculosis infection in mice (161). Another recent murine study established proof of concept that augmentation of prostaglandin E2 levels, which were shown to promote IL-1 production and restrict excessive type I interferon production, led to containment of M. tuberculosis and prevented mortality (162). IL-10 has also been implicated in the negative repercussions of type I IFN signaling in murine models, as type I IFN induction of IL-10 in myeloid subsets in the lung during M. tuberculosis infection was found to repress IL-1 production (142). Furthermore, M. tuberculosis-infected murine macrophages were shown to synthesize IL-10 after type I IFN treatment, leading to reduced mycobacterial killing in these cells in response to lymphoid-derived IFN-c (143). It should be noted, however, that type I IFNs may also serve a protective role once infection is established in mice by reducing the number of M. tuberculosis-susceptible target cells in the lung, thereby limiting disease progression at the site of infection (163). In humans, a recent study of the effects of type I interferons on human macrophages in vitro showed that IFN-b upregulated IL-10 expression, which in turn inhibited IFN-cmediated macrophage activation and killing of intracellular mycobacteria (164). Combined with several reports that have found augmented type I IFN production to correspond with active TB disease development and/or severity (149, 165, 166), these findings provide further support for a neg-

80

ative impact of type I IFNs on host control of M. tuberculosis. Indeed, although more research is necessary, it is likely that the adaptive T-cell response is inhibited by type I IFNs, as reduced production of critical T cell-activating factors like IL-12, TNF, and IL-1b due to type I IFN-mediated IL-10 upregulation would be expected to drive a markedly diminished M. tuberculosis-specific Th1 response. T-cell responses and M. tuberculosis/HIV co-infection Recent studies on TB/HIV co-infection have elucidated new insights into adaptive T-cell responses to TB. M. tuberculosisspecific CD4+ T cells are depleted from the periphery early after HIV infection in humans, and evidence suggests that this is partially due to preferential targeting of M. tuberculosisspecific memory CD4+ T cells by HIV (167, 168). A number of reports have found that TB disease enhances CCR5+ expression and/or the frequency of CCR5+ CD4+ T cells in the absence (169–172) and presence of HIV co-infection (169, 173, 174, Haridas et al., manuscript submitted), indicating that the chronic immune activation that accompanies active TB leads to a larger CCR5+ CD4+ T-cell population that is presumably susceptible to highly efficient infection by HIV. Furthermore, a strong CD4+ T-cell response to M. tuberculosis might simultaneously lead to enhanced HIV replication, as demonstrated in in vitro studies where a primary subtype AE HIV-1 isolate replicated to a greater extent in CD4+ T cells from HIV-negative PPD-responsive donors compared to CD4+ T cells from HIV-negative PPD-anergic donors after PPD stimulation (175). Notably, this process was partly mediated by PPD-induced IL-10 suppression of viral replication in the anergic donor cells (175). Furthermore, distinct M. tuberculosis strains can drive differential HIV replication in ex vivo PBMCs, as less virulent strains that drive strong proinflammatory cytokine production were found to promote enhanced replication of primary subtype B, C, and AE viruses, while more virulent M. tuberculosis strains that are less immunogenic drove lower levels of viral replication (176). These data indicate that antigen-specific responses to M. tuberculosis would be expected to enhance HIV replication, consistent with the higher viral loads that are detected in TB/HIV co-infected patients versus patients singly infected with HIV who exhibit similar levels of immunosuppression (177, 178, Haridas et al., manuscript submitted). Examination of polyfunctional T-cell responses in M. tuberculosis/HIV co-infection found that viral load in co-infected TB patients with latent TB inversely correlated with M. tuberculosis-specific IL-2-expressing CD4+ T cells that were positive for TNF or IFN-c, while the M. tuberculosis-specific © 2015 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd Immunological Reviews 264/2015

Jasenosky et al  T cells and the human immune response to TB

CD8+ T-cell population was highly enriched in IL-2 cells (179). A later report confirmed that increased immunosuppression in HIV+ patients with latent TB infection was associated with a loss of polyfunctional M. tuberculosis-specific CD4+ T-cell responses, which were restored following ART (180). Bronchoalveolar lavage (BAL) fluid from HIV+ patients with latent M. tuberculosis infection also exhibited reduced M. tuberculosis-specific polyfunctional as well as bi- and mono-functional CD4+ T-cell frequencies compared to those from patients free of HIV (181, 182). Another report found that ART did not fully restore M. tuberculosis-specific polyfunctional CD4+ T-cell responses in bronchoalveolar cells (183). Notably, however, BCG-vaccinated HIV+ patients who maintained M. tuberculosis-specific IFN-c-secreting T cells had significantly reduced risk of future active TB disease (184). Recapitulating what has been established in human ex vivo studies, the cynomolgus macaque model of latent M. tuberculosis and SIV infection has linked depletion of peripheral CD4+ T cells to TB reactivation (185). Furthermore, it was reported that animals who experience early TB reactivation relative to those who reactivate at later time points exhibit an elevation in M. tuberculosis-specific trifunctional IFNc+ TNF+ IL-2+ CD4+ T-cell frequencies (186). Again, this latter finding is consistent with the reports described above that demonstrated that elevated human M. tuberculosis-specific trifunctional IFN-c+ TNF+ IL-2+ CD4+ T cells are a consequence of high M. tuberculosis burden and not necessarily a sign of effective M. tuberculosis-restrictive immunity.

ciated with risk of TB-IRIS, further supporting a role for CD4+ T-cell activation in the development of TB-IRIS (193). In a prospective and large immunological study of T-cell evolution in TB-IRIS patients, which was nested within the Cambodian Early versus Late Introduction of Antiretrovirals (CAMELIA) randomized clinical trial in Cambodia (194), whole blood immunophenotypes were analyzed prior to ART initiation and at several time points following ART initiation in severely immunocompromised (median CD4+ Tcell count of 25/mm3) patients with documented TB who had initiated TB therapy (195). Patients who developed TBIRIS began ART with significantly higher frequencies of activated (HLA-DR+ CD45RO+) CD4+ T-cells and CCR5+ CD4+ T-cells than patients who avoided TB-IRIS (195). In addition, elevated pre-ART OX40+ CD4+ T-cell proportions were a specific risk factor for TB-IRIS. Furthermore, after ART initiation, patients who experienced TB-IRIS exhibited a significantly greater expansion in effector + memory (CD62L CD45RA ) CD4 T-cell frequencies and a reciprocal contraction in central memory (CD62L+ CD45RA ) T-cell frequencies (195). These latter findings indicate that TB-IRIS drives the formation of a CD4+ T-cell memory compartment that is distinct from that seen in similarly immune compromised patients who avoid TB-IRIS. In summary, TB-IRIS appears to represent the far end of a spectrum of potent MTb-specific CD4+ T-cell responses that may provide superior long-term protection from TB reactivation and/or reinfection (194). T-cell responses and vaccination against TB

Lessons from TB-associated immune reconstitution syndrome M. tuberculosis-specific T-cell responses also play a critical role in TB-associated immune reconstitution inflammatory syndrome (TB-IRIS), a condition that commonly occurs in HIV+ patients who are undergoing simultaneous TB therapy and ART or during the unmasking of occult TB in HIV patients who are initiating ART (187). Several studies have demonstrated that ex vivo M. tuberculosis antigen stimulation of blood or PBMCs from TB-IRIS patients collected prior to, at the time of, or soon after the TB-IRIS event results in strong M. tuberculosis-specific Th1 responses (188–191). In one investigation, ex vivo stimulation of PBMCs from TB-IRIS patients with M. tuberculosis antigens led to a strong enrichment in IFN-c+ TNF+ IL-2 CD4+ T cells (192). In a recent report, higher soluble IL-2 receptor plasma levels were asso-

© 2015 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd Immunological Reviews 264/2015

The only current vaccine against TB is Bacillus Calmette Guerin (BCG), a live attenuated strain of Mycobacterium bovis. This vaccine is given to the majority of the world’s newborns soon after birth, who are then protected against disseminated forms of tuberculosis, such as meningitis or miliary disease. BCG induces a Th1-like CD4+ T-cell response (196) and following vaccination multiple patterns of CD4+ T-cell cytokine production are common. Combined expression of IFN-c, TNF, and IL-2 and single expression of IFN-c have been described most frequently (73, 196, 197). In addition, IL-17 expression, mostly distinct from Th1 cytokine production, is detectable (73). BCG also activates CD8+ T cells, but at quantitatively much lower levels than CD4+ T cells, with IFN-c production dominating in these cells (73, 196, 197). Vaccine-induced CD4+ and CD8+ T cells may also have cytotoxic potential, a function that appears distinct from cytokine producing capacity, and

81

Jasenosky et al  T cells and the human immune response to TB

induced cells have proliferative potential (197, 198). The peak response to newborn BCG vaccination appears to be around 6 weeks of age, after which the response declines but remains detectable until at least 12 months of age (197). Delaying BCG vaccination from the first day of life to later in infancy, and administration of the vaccine to adults may affect quantitative, qualitative, and kinetic characteristics of the immune response to the vaccine (199–201). The relative contribution of BCG-induced T-cell immunity to protection against childhood TB disease is unclear. A large, prospective study of BCG-vaccinated infants showed that, 10 weeks after newborn vaccination, there was no difference in any measure of BCG-specific T-cell immunity, including markers of Th1 and Th17 activation, between infants who subsequently developed TB disease in their first 2 years of life and those who did not (73). The IFN-c/IL12 axis is clearly essential for protection against mycobacteria, as shown by congenital deficiencies in humans and knockouts of components of this pathway in animals; however, as this study implies, it is unclear whether this immunity is sufficient for protection (202). Alternatively, the correlates of risk of TB disease might have been measured at a suboptimal time point in this particular infant study. The same dilemma complicates elucidation of the factors that contribute to an effective T-cell response after immunization with novel vaccines formulations, and we do not know whether, and which, T-cell responses correlate with protection against TB disease. Most novel vaccines in the current clinical portfolio, including whole mycobacterial candidates, adjuvanted proteins, and viral vectored vaccines, induce type I CD4+ T-cell responses comprised mainly of IFN-c+ TNF+ IL-2+ or at least Th1 responses (203–206). In some cases, CD8+ T-cell immunity is also elicited. One viral vectored candidate, MVA85A, induced easily detectable CD4+ T-cell responses in a randomized, placebo-controlled phase IIb trial of infants who received the vaccine or placebo about 4 months after newborn BCG; regardless, there was no difference in protection against TB disease between the two groups (207). This result again questions the role of these cells in protection against TB disease, and challenges the common practice of measuring conventional

CD4+ and CD8+ T-cell responses as immunogenicity outcomes in studies of novel TB vaccines. Similarly, although the hypothesis is attractive, there is no evidence that qualitative and quantitative differences in CD4+ or CD8+ T-cell immunity induced by current novel vaccines are important. Until we have correlates of protection against TB disease, which is possible only following a successful placebo-controlled efficacy trial of a novel TB vaccine, we will have to devise alternate strategies to gain insights into immune responses that are necessary for effective immunity. First, in every clinical study, much more detailed assessment of induced immunity is important for iterative learning and effort should be made to measure global immunological signatures characteristic of an effective adaptive immune response, thereby reducing the risk that preconceived experimental biases will obscure or preclude unanticipated observations. Further, results from prospective studies of human populations at elevated risk of TB disease might guide the search of correlates of protection. At the very least, results could be used to learn whether individual candidates induce immunity that might be detrimental rather than protective, such as induction of lung inflammation leading to increased recruitment of target cells for M. tuberculosis infection. Finally, as we do not know whether conventional CD4+ and CD8+ T-cell immunity is sufficient for protection, nor to which antigens these responses should be directed, an attractive alternate strategy in vaccination against TB could involve eliciting ‘uncommon’ or non-conventional T-cell responses, particularly if antigen presentation involved is less polymorphic. Examples include induction of CD1 and HLA-E-mediated T-cell responses, cd T-cell responses, or populations of T cells that primarily reside in the lung, such as mucosal-associated T cells (208–210). The development of improved vaccination approaches will require greater characterization of the antigens involved in the early phases of M. tuberculosis infection and that play a part in the early stages of latent mycobacterial reactivation combined with. They will also require a deeper understanding of the memory characteristics of induced T-cell populations necessary for the induction of an effective and long-lasting adaptive immune response in human TB disease.

References 1. Yang H, Kruh-Garcia NA, Dobos KM. Purified protein derivatives of tuberculin–past, present, and future. FEMS Immunol Med Microbiol 2012;66:273–280. 2. Wallgren A. The time-table of tuberculosis. Tubercle 1948;29:245–251.

82

3. DyeC,ScheeleS,DolinP,PathaniaV,RaviglioneMC. Consensusstatement.Globalburdenoftuberculosis: estimatedincidence,prevalence,andmortalityby country.WHOGlobalSurveillanceandMonitoring Project.JAMA1999;282:677–686.

4. Millington KA, Gooding S, Hinks TS, Reynolds DJ, Lalvani A. Mycobacterium tuberculosisspecific cellular immune profiles suggest bacillary persistence decades after spontaneous cure in untreated tuberculosis. J Infect Dis 2010;202:1685–1689.

© 2015 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd Immunological Reviews 264/2015

Jasenosky et al  T cells and the human immune response to TB

5. 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. 6. Sousa AO, et al. An epidemic of tuberculosis with a high rate of tuberculin anergy among a population previously unexposed to tuberculosis, the Yanomami Indians of the Brazilian Amazon. Proc Natl Acad Sci USA 1997;94:13227–13232. 7. Boussiotis VA, et al. IL-10-producing T cells suppress immune responses in anergic tuberculosis patients. J Clin Invest 2000;105:1317–1325. 8. Delgado JC, et al. Antigen-specific and persistent tuberculin anergy in a cohort of pulmonary tuberculosis patients from rural Cambodia. Proc Natl Acad Sci USA 2002;99:7576–7581. 9. Cobat A, et al. Two loci control tuberculin skin test reactivity in an area hyperendemic for tuberculosis. J Exp Med 2009;206:2583–2591. 10. Delgado JC, Quinones-Berrocal J, Thim S, Miranda LF, Goldfeld AE. Diagnostic and clinical implications of response to tuberculin in two ethnically distinct populations from Peru and Cambodia. Int J Tuberc Lung Dis 2004;8:982–987. 11. Baliko Z, Szereday L, Szekeres-Bartho J. Th2 biased immune response in cases with active Mycobacterium tuberculosis infection and tuberculin anergy. FEMS Immunol Med Microbiol 1998;22:199–204. 12. Thye T, et al. IL10 haplotype associated with tuberculin skin test response but not with pulmonary TB. PLoS ONE 2009;4:e5420. 13. Yan L, Cui H, Xiao H, Zhang Q. Anergic pulmonary tuberculosis is associated with contraction of the Vd2+ T cell population, apoptosis and enhanced inhibitory cytokine production. PLoS ONE 2013;8:e71245. 14. Szereday L, Baliko Z, Szekeres-Bartho J. Gamma/ delta T cell subsets in patients with active Mycobacterium tuberculosis infection and tuberculin anergy. Clin Exp Immunol 2003;131:287–291. 15. Zembrzuski VM, et al. Cytokine genes are associated with tuberculin skin test response in a native Brazilian population. Tuberculosis 2010;90:44–49. 16. Lindenau JD, et al. Cytokine gene polymorphisms are associated with susceptibility to tuberculosis in an Amerindian population. Int J Tuberc Lung Dis 2014;18:952–957. 17. Lindenau JD, et al. Association between HLADR4 haplotypes and tuberculin skin test response in the Ache population. Tissue Antigens 2014;84:479–483. 18. Delgado JC, Baena A, Thim S, Goldfeld AE. Aspartic acid homozygosity at codon 57 of HLADQ beta is associated with susceptibility to pulmonary tuberculosis in Cambodia. J Immunol 2006;176:1090–1097. 19. Opie EL, Aronson JD. Tubercle bacilli in latent tuberculosis lesions and in lung tissue without tuberculous lesions. Arch Path & Lab Med 1927;4:1–21. 20. Keane J, et al. Tuberculosis associated with infliximab, a tumor necrosis factor alpha-

21.

22.

23. 24. 25.

26.

27.

28.

29.

30.

31.

32.

33.

34.

35.

36.

37.

38.

neutralizing agent. N Engl J Med 2001;345:1098–1104. Singh JA, et al. Adverse effects of biologics: a network meta-analysis and Cochrane overview. Cochrane Database Syst Rev 2011;2:CD008794. Kaufmann SH, McMichael AJ. Annulling a dangerous liaison: vaccination strategies against AIDS and tuberculosis. Nat Med 2005;11: S33–S44. Philips JA, Ernst JD. Tuberculosis pathogenesis and immunity. Ann Rev Pathol 2012;7:353–384. Flynn JL, Chan J. Immunology of tuberculosis. Annu Rev Immunol 2001;19:93–129. Ernst JD. The immunological life cycle of tuberculosis. Nat Rev Immunol 2012;12: 581–591. Gupta A, Kaul A, Tsolaki AG, Kishore U, Bhakta S. Mycobacterium tuberculosis: immune evasion, latency and reactivation. Immunobiology 2012;217:363–374. Wiker HG, Mustafa T, Bjune GA, Harboe M. Evidence for waning of latency in a cohort study of tuberculosis. BMC Infect Dis 2010;10:37. Wood R, Lawn SD, Caldwell J, Kaplan R, Middelkoop K, Bekker LG. Burden of new and recurrent tuberculosis in a major South African city stratified by age and HIV-status. PLoS ONE 2011;6:e25098. Charalambous S, et al. Contribution of reinfection to recurrent tuberculosis in South African gold miners. Int J Tuberc Lung Dis 2008;12:942–948. Crampin AC, et al. Recurrent TB: relapse or reinfection? The effect of HIV in a general population cohort in Malawi. AIDS 2010;24:417–426. van Rie A, et al. Exogenous reinfection as a cause of recurrent tuberculosis after curative treatment. N Engl J Med 1999;341:1174–1179. Sonnenberg P, Murray J, Glynn JR, Shearer S, Kambashi B, Godfrey-Faussett P. HIV-1 and recurrence, relapse, and reinfection of tuberculosis after cure: a cohort study in South African mineworkers. Lancet 2001;358: 1687–1693. Hawn TR, et al. TB vaccines and prevention of infection. Microb Mol Biol Rev 2014;78:650– 671. Fine PE, Bruce J, Ponnighaus JM, Nkhosa P, Harawa A, Vynnycky E. Tuberculin sensitivity: conversions and reversions in a rural African population. Int J Tuberc Lung Dis 1999;3: 962–975. Adams JM, Kalajan VA, Mork BO, Rosenblatt M, Rothrock WJ, O’Loughlin BJ. Reversal of tuberculin reaction in early tuberculosis. Dis Chest 1959;35:348–356. Hill PC, et al. Longitudinal assessment of an ELISPOT test for Mycobacterium tuberculosis infection. PLoS Med 2007;4:e192. Machingaidze S, et al. Predictive value of recent QuantiFERON conversion for tuberculosis disease in adolescents. Am J Respir Crit Care Med 2012;186:1051–1056. Dahlstrom AW. The instability of the tuberculin reaction. Am Rev Turberc 1940;42:471–487.

© 2015 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd Immunological Reviews 264/2015

39. Andrews JR, et al. The dynamics of QuantiFERON-TB Gold In-Tube convesion and reversion in a cohort of South African adolescents. Am J Respir Crit Care Med 2015, in press. 40. Khader SA, et al. Interleukin 12p40 is required for dendritic cell migration and T cell priming after Mycobacterium tuberculosis infection. J Exp Med 2006;203:1805–1815. 41. Wolf AJ, et al. Initiation of the adaptive immune response to Mycobacterium tuberculosis depends on antigen production in the local lymph node, not the lungs. J Exp Med 2008;205:105–115. 42. Chackerian AA, Alt JM, Perera TV, Dascher CC, Behar SM. Dissemination of Mycobacterium tuberculosis is influenced by host factors and precedes the initiation of T-cell immunity. Infect Immun 2002;70:4501–4509. 43. Wolf AJ, et al. Mycobacterium tuberculosis infects dendritic cells with high frequency and impairs their function in vivo. J Immunol 2007;179:2509–2519. 44. Srivastava S, Ernst JD. Cell-to-cell transfer of M. tuberculosis antigens optimizes CD4 T cell priming. Cell Host Microbe 2014;15:741–752. 45. Rajalingam R, Mehra NK, Jain RC, Myneedu VP, Pande JN. Polymerase chain reaction–based sequence-specific oligonucleotide hybridization analysis of HLA class II antigens in pulmonary tuberculosis: relevance to chemotherapy and disease severity. J Infect Dis 1996;173:669–676. 46. Pospelov LE, et al. Association of various genetic markers with tuberculosis and other lung diseases in Tuvinian children. Tuber Lung Dis 1996;77:77–80. 47. Brahmajothi V, et al. Association of pulmonary tuberculosis and HLA in south India. Tubercle 1991;72:123–132. 48. Khomenko AG, Litvinov VI, Chukanova VP, Pospelov LE. Tuberculosis in patients with various HLA phenotypes. Tubercle 1990;71: 187–192. 49. Bothamley GH, et al. Association of tuberculosis and M. tuberculosis-specific antibody levels with HLA. J Infect Dis 1989;159:549–555. 50. Singh SP, Mehra NK, Dingley HB, Pande JN, Vaidya MC. Human leukocyte antigen (HLA)linked control of susceptibility to pulmonary tuberculosis and association with HLA-DR types. J Infect Dis 1983;148:676–681. 51. Kettaneh A, Seng L, Tiev KP, Toledano C, Fabre B, Cabane J. Human leukocyte antigens and susceptibility to tuberculosis: a meta-analysis of case-control studies. Int J Tuberc Lung Dis 2006;10:717–725. 52. Singh M, Balamurugan A, Katoch K, Sharma SK, Mehra NK. Immunogenetics of mycobacterial infections in the North Indian population. Tissue Antigens 2007;69(Suppl 1):228–230. 53. Ravikumar M, et al. Associations of HLA-DRB1, DQB1 and DPB1 alleles with pulmonary tuberculosis in south India. Tuber Lung Dis 1999;79:309–317. 54. Goldfeld AE, et al. Association of an HLA-DQ allele with clinical tuberculosis. J Am Med Assoc 1998;279:226–228.

83

Jasenosky et al  T cells and the human immune response to TB

55. Mogues T, Goodrich ME, Ryan L, LaCourse R, North RJ. The relative importance of T cell subsets in immunity and immunopathology of airborne Mycobacterium tuberculosis infection in mice. J Exp Med 2001;193:271–280. 56. Yao S, Huang D, Chen CY, Halliday L, Wang RC, Chen ZW. CD4+ T cells contain early extrapulmonary tuberculosis (TB) dissemination and rapid TB progression and sustain multieffector functions of CD8+ T and CD3lymphocytes: mechanisms of CD4+ T cell immunity. J Immunol 2014;192:2120–2132. 57. Lin PL, et al. CD4 T cell depletion exacerbates acute Mycobacterium tuberculosis while reactivation of latent infection is dependent on severity of tissue depletion in cynomolgus macaques. AIDS Res Hum Retroviruses 2012;28:1693–1702. 58. Chen CY, et al. A critical role for CD8 T cells in a nonhuman primate model of tuberculosis. PLoS Pathog 2009;5:e1000392. 59. Rossouw M, Nel HJ, Cooke GS, van Helden PD, Hoal EG. Association between tuberculosis and a polymorphic NFkappaB binding site in the interferon gamma gene. Lancet 2003;361: 1871–1872. 60. Seder RA, Darrah PA, Roederer M. T-cell quality in memory and protection: implications for vaccine design. Nat Rev Immunol 2008;8: 247–258. 61. Jeong YH, et al. Differentiation of antigenspecific T cells with limited functional capacity during Mycobacterium tuberculosis infection. Infect Immun 2014;82:132–139. 62. Chiacchio T, et al. Higher frequency of T-cell response to M. tuberculosis latency antigen Rv2628 at the site of active tuberculosis disease than in peripheral blood. PLoS ONE 2011;6: e27539. 63. Pollock KM, et al. T-cell immunophenotyping distinguishes active from latent tuberculosis. J Infect Dis 2013;208:952–968. 64. Harari A, et al. Dominant TNF-alpha+ Mycobacterium tuberculosis-specific CD4+ T cell responses discriminate between latent infection and active disease. Nat Med 2011;17:372–376. 65. Day CL, et al. Functional capacity of Mycobacterium tuberculosis-specific T cell responses in humans is associated with mycobacterial load. J Immunol 2011;187: 2222–2232. 66. Petruccioli E, et al. IFNgamma/TNFalpha specific-cells and effector memory phenotype associate with active tuberculosis. J Infect 2013;66:475–486. 67. Riou C, et al. A subset of circulating blood mycobacteria-specific CD4 T cells can predict the time to Mycobacterium tuberculosis sputum culture conversion. PLoS ONE 2014;9:e102178. 68. Sutherland JS, Adetifa IM, Hill PC, Adegbola RA, Ota MO. Pattern and diversity of cytokine production differentiates between Mycobacterium tuberculosis infection and disease. Eur J Immunol 2009;39:723–729. 69. Qiu Z, et al. Multifunctional CD4 T cell responses in patients with active tuberculosis. Sci Rep 2012;2:216.

84

70. Mueller H, et al. Mycobacterium tuberculosisspecific CD4+, IFNgamma+, and TNFalpha+ multifunctional memory T cells coexpress GMCSF. Cytokine 2008;43:143–148. 71. Caccamo N, et al. Multifunctional CD4(+) T cells correlate with active Mycobacterium tuberculosis infection. Eur J Immunol 2010;40:2211–2220. 72. Young JM, Adetifa IM, Ota MO, Sutherland JS. Expanded polyfunctional T cell response to mycobacterial antigens in TB disease and contraction post-treatment. PLoS ONE 2010;5: e11237. 73. Kagina BM, 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;182:1073–1079. 74. Kumar NP, et al. Expansion of pathogen-specific mono- and multifunctional Th1 and Th17 cells in multi-focal tuberculous lymphadenitis. PLoS ONE 2013;8:e57123. 75. Jenum S, et al. The frequencies of IFNgamma+IL2+TNFalpha+ PPD-specific CD4+CD45RO+ T-cells correlate with the magnitude of the QuantiFERON(R) gold in-tube response in a prospective study of healthy indian adolescents. PLoS ONE 2014;9:e101224. 76. Streitz M, et al. Loss of receptor on tuberculinreactive T-cells marks active pulmonary tuberculosis. PLoS ONE 2007;2:e735. 77. Commandeur S, et al. Double- and monofunctional CD4(+) and CD8(+) T-cell responses to Mycobacterium tuberculosis DosR antigens and peptides in long-term latently infected individuals. Eur J Immunol 2011;41:2925–2936. 78. Li L, Qiao D, Fu X, Lao S, Zhang X, Wu C. Identification of Mycobacterium tuberculosisspecific Th1, Th17 and Th22 cells using the expression of CD40L in tuberculous pleurisy. PLoS ONE 2011;6:e20165. 79. Wang X, et al. Association of mycobacterial antigen-specific CD4(+) memory T cell subsets with outcome of pulmonary tuberculosis. J Infect 2010;60:133–139. 80. El Fenniri L, et al. Polyfunctional Mycobacterium tuberculosis-specific effector memory CD4+ T cells at sites of pleural TB. Tuberculosis 2011;91:224–230. 81. Adekambi T, Ibegbu CC, Kalokhe AS, Yu T, Ray SM, Rengarajan J. Distinct effector memory CD4+ T cell signatures in latent Mycobacterium tuberculosis infection, BCG vaccination and clinically resolved tuberculosis. PLoS ONE 2012;7:e36046. 82. Arlehamn CS, et al. Dissecting mechanisms of immunodominance to the common tuberculosis antigens ESAT-6, CFP10, Rv2031c (hspX), Rv2654c (TB7.7), and Rv1038c (EsxJ). J Immunol 2012;188:5020–5031. 83. Goletti D, Butera O, Bizzoni F, Casetti R, Girardi E, Poccia F. Region of difference 1 antigenspecific CD4+ memory T cells correlate with a favorable outcome of tuberculosis. J Infect Dis 2006;194:984–992. 84. Han Q, Bagheri N, Bradshaw EM, Hafler DA, Lauffenburger DA, Love JC. Polyfunctional

85.

86.

87.

88.

89.

90.

91.

92.

93.

94.

95.

96.

97.

responses by human T cells result from sequential release of cytokines. Proc Natl Acad Sci USA 2012;109:1607–1612. Kaveh DA, Whelan AO, Hogarth PJ. The duration of antigen-stimulation significantly alters the diversity of multifunctional CD4 T cells measured by intracellular cytokine staining. PLoS ONE 2012;7:e38926. Acosta-Rodriguez EV, et al. Surface phenotype and antigenic specificity of human interleukin 17-producing T helper memory cells. Nat Immunol 2007;8:639–646. Lindestam Arlehamn CS, et al. Memory T cells in latent Mycobacterium tuberculosis infection are directed against three antigenic islands and largely contained in a CXCR3+CCR6+ Th1 subset. PLoS Pathog 2013;9:e1003130. Smith SM, et al. Human CD8(+) T cells specific for Mycobacterium tuberculosis secreted antigens in tuberculosis patients and healthy BCGvaccinated controls in The Gambia. Infect Immun 2000;68:7144–7148. Andersson J, Samarina A, Fink J, Rahman S, Grundstrom S. Impaired expression of perforin and granulysin in CD8+ T cells at the site of infection in human chronic pulmonary tuberculosis. Infect Immun 2007;75: 5210–5222. Li L, Yang B, Zhang X, Lao S, Changyou W. Mycobacterium tuberculosis-specific polyfunctional cytotoxic CD8+ T cells express CD69. Tuberculosis 2014;94:219–225. Caccamo N, et al. Analysis of Mycobacterium tuberculosis-specific CD8 T-cells in patients with active tuberculosis and in individuals with latent infection. PLoS ONE 2009;4:e5528. Axelsson-Robertson R, et al. A broad profile of co-dominant epitopes shapes the peripheral Mycobacterium tuberculosis specific CD8+ T-cell immune response in South African patients with active tuberculosis. PLoS ONE 2013;8: e58309. Day CL, et al. Patients with tuberculosis disease have Mycobacterium tuberculosis-specific CD8 T cells with a pro-apoptotic phenotype and impaired proliferative capacity, which is not restored following treatment. PLoS ONE 2014;9: e94949. Bruns H, et al. Anti-TNF immunotherapy reduces CD8+ T cell-mediated antimicrobial activity against Mycobacterium tuberculosis in humans. J Clin Invest 2009;119:1167–1177. Gaffen SL, et al. The IL-23-IL-17 immune axis: from mechanisms to therapeutic testing. Nat Rev Immunol 2014;14:585–600. Khader SA, et al. IL-23 compensates for the absence of IL-12p70 and is essential for the IL17 response during tuberculosis but is dispensable for protection and antigen-specific IFN-gamma responses if IL-12p70 is available. J Immunol 2005;175:788–795. Umemura M, et al. IL-17-mediated regulation of innate and acquired immune response against pulmonary Mycobacterium bovis bacille Calmette-Guerin infection. J Immunol 2007;178:3786–3796.

© 2015 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd Immunological Reviews 264/2015

Jasenosky et al  T cells and the human immune response to TB

98. Lockhart E, Green AM, Flynn JL. IL-17 production is dominated by gammadelta T cells rather than CD4 T cells during Mycobacterium tuberculosis infection. J Immunol 2006;177:4662–4669. 99. Okamoto Yoshida Y, et al. Essential role of IL17A in the formation of a mycobacterial infection-induced granuloma in the lung. J Immunol 2010;184:4414–4422. 100. Cruz A, et al. Cutting edge: IFN-gamma regulates the induction and expansion of IL-17-producing CD4 T cells during mycobacterial infection. J Immunol 2006;177:1416–1420. 101. Desvignes L, Ernst JD. Interferon-gammaresponsive nonhematopoietic cells regulate the immune response to Mycobacterium tuberculosis. Immunity 2009;31:974–985. 102. Freches D, Korf H, Denis O, Havaux X, Huygen K, Romano M. Mice genetically inactivated in interleukin-17A receptor are defective in longterm control of Mycobacterium tuberculosis infection. Immunology 2013;140: 220–231. 103. Gopal R, et al. Unexpected role for IL-17 in protective immunity against hypervirulent Mycobacterium tuberculosis HN878 infection. PLoS Pathog 2014;10:e1004099. 104. Perreau M, et al. Lack of Mycobacterium tuberculosis-specific interleukin-17A-producing CD4+ T cells in active disease. Eur J Immunol 2013;43:939–948. 105. Chen X, et al. Reduced Th17 response in patients with tuberculosis correlates with IL-6R expression on CD4+ T Cells. Am J Respir Crit Care Med 2010;181:734–742. 106. Hur YG, et al. Combination of cytokine responses indicative of latent TB and active TB in Malawian adults. PLoS ONE 2013;8:e79742. 107. Marin ND, Paris SC, Rojas M, Garcia LF. Functional profile of CD4+ and CD8+ T cells in latently infected individuals and patients with active TB. Tuberculosis 2013;93:155–166. 108. Cowan J, Pandey S, Filion LG, Angel JB, Kumar A, Cameron DW. Comparison of interferongamma-, interleukin (IL)-17- and IL-22expressing CD4 T cells, IL-22-expressing granulocytes and proinflammatory cytokines during latent and active tuberculosis infection. Clin Exp Immunol 2012;167:317–329. 109. Bandaru A, et al. Phosphorylated STAT3 and PD1 regulate IL-17 production and IL-23 receptor expression in Mycobacterium tuberculosis infection. Eur J Immunol 2014;44:2013–2024. 110. Peng MY, et al. Interleukin 17-producing gamma delta T cells increased in patients with active pulmonary tuberculosis. Cell Mol Immunol 2008;5:203–208. 111. Sutherland JS, de Jong BC, Jeffries DJ, Adetifa IM, Ota MO. Production of TNF-alpha, IL-12(p40) and IL-17 can discriminate between active TB disease and latent infection in a West African cohort. PLoS ONE 2010;5:e12365. 112. Marin ND, Paris SC, Velez VM, Rojas CA, Rojas M, Garcia LF. Regulatory T cell frequency and modulation of IFN-gamma and IL-17 in active and latent tuberculosis. Tuberculosis 2010;90:252–261.

113. Scriba TJ, et al. Distinct, specific IL-17- and IL22-producing CD4+ T cell subsets contribute to the human anti-mycobacterial immune response. J Immunol 2008;180:1962–1970. 114. Jurado JO, et al. IL-17 and IFN-gamma expression in lymphocytes from patients with active tuberculosis correlates with the severity of the disease. J Leukoc Biol 2012;91:991– 1002. 115. Loxton AG, Black GF, Stanley K, Walzl G. Heparin-binding hemagglutinin induces IFNgamma(+) IL-2(+) IL-17(+) multifunctional CD4 (+) T cells during latent but not active tuberculosis disease. Clin Vaccine Immunol 2012;19:746–751. 116. Belkaid Y, Piccirillo CA, Mendez S, Shevach EM, Sacks DL. CD4+CD25+ regulatory T cells control Leishmania major persistence and immunity. Nature 2002;420:502–507. 117. Fontenot JD, Gavin MA, Rudensky AY. Foxp3 programs the development and function of CD4+CD25+ regulatory T cells. Nat Immunol 2003;4:330–336. 118. Hori S, Nomura T, Sakaguchi S. Control of regulatory T cell development by the transcription factor Foxp3. Science 2003;299:1057–1061. 119. Pang H, et al. Frequency of regulatory T-cells in the peripheral blood of patients with pulmonary tuberculosis from shanxi province, china. PLoS ONE 2013;8:e65496. 120. Guyot-Revol V, Innes JA, Hackforth S, Hinks T, Lalvani A. Regulatory T cells are expanded in blood and disease sites in patients with tuberculosis. Am J Respir Crit Care Med 2006;173:803–810. 121. Ribeiro-Rodrigues R, et al. A role for CD4+CD25+ T cells in regulation of the immune response during human tuberculosis. Clin Exp Immunol 2006;144:25–34. 122. Chen X, et al. CD4(+)CD25(+)FoxP3(+) regulatory T cells suppress Mycobacterium tuberculosis immunity in patients with active disease. Clin Immunol 2007;123:50–59. 123. He XY, et al. T regulatory cells and Th1/Th2 cytokines in peripheral blood from tuberculosis patients. Eur J Clin Microbiol Infect Dis 2010;29:643–650. 124. Rahman S, et al. Compartmentalization of immune responses in human tuberculosis: few CD8+ effector T cells but elevated levels of FoxP3+ regulatory t cells in the granulomatous lesions. Am J Pathol 2009;174:2211–2224. 125. Kursar M, et al. Cutting edge: regulatory T cells prevent efficient clearance of Mycobacterium tuberculosis. J Immunol 2007;178:2661–2665. 126. Mason CM, Porretta E, Zhang P, Nelson S. CD4+ CD25+ transforming growth factor-betaproducing T cells are present in the lung in murine tuberculosis and may regulate the host inflammatory response. Clin Exp Immunol 2007;148:537–545. 127. Scott-Browne JP, et al. Expansion and function of Foxp3-expressing T regulatory cells during tuberculosis. J Exp Med 2007;204:2159–2169.

© 2015 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd Immunological Reviews 264/2015

128. Shafiani S, Tucker-Heard G, Kariyone A, Takatsu K, Urdahl KB. Pathogen-specific regulatory T cells delay the arrival of effector T cells in the lung during early tuberculosis. J Exp Med 2010;207:1409–1420. 129. Green AM, Mattila JT, Bigbee CL, Bongers KS, Lin PL, Flynn JL. CD4(+) regulatory T cells in a cynomolgus macaque model of Mycobacterium tuberculosis infection. J Infect Dis 2010;202:533–541. 130. Chen CY, et al. IL-2 simultaneously expands Foxp3+ T regulatory and T effector cells and confers resistance to severe tuberculosis (TB): implicative Treg-T effector cooperation in immunity to TB. J Immunol 2012;188: 4278–4288. 131. Borsellino G, et al. Expression of ectonucleotidase CD39 by Foxp3+ Treg cells: hydrolysis of extracellular ATP and immune suppression. Blood 2007;110:1225–1232. 132. Chiacchio T, et al. Characterization of regulatory T cells identified as CD4(+)CD25(high)CD39(+) in patients with active tuberculosis. Clin Exp Immunol 2009;156:463–470. 133. Kim K, et al. Circulating mycobacterial-reactive CD4+ T cells with an immunosuppressive phenotype are higher in active tuberculosis than latent tuberculosis infection. Tuberculosis 2014;94:494–501. 134. de Cassan SC, et al. Investigating the induction of vaccine-induced Th17 and regulatory T cells in healthy, Mycobacterium bovis BCG-immunized adults vaccinated with a new tuberculosis vaccine, MVA85A. Clin Vaccine Immunol 2010;17:1066–1073. 135. Griffiths KL, et al. Th1/Th17 cell induction and corresponding reduction in ATP consumption following vaccination with the novel Mycobacterium tuberculosis vaccine MVA85A. PLoS ONE 2011;6:e23463. 136. de Waal Malefyt R, Abrams J, Bennett B, Figdor C, Vries J. IL-10 inhibits cytokine synthesis by human monocytes: an autoregulatory role of IL10 produced by monocytes. J Exp Med 1991;174:1209–1220. 137. de Waal Malefyt R, et al. IL-10 and v-IL-10 strongly reduce antigen specific human T cell responses by diminishing the antigen presenting capacity of monocytes via down-regulation of class II MHC expression. J Exp Med 1991;174:915–924. 138. Ding L, Shevach EM. IL-10 inhibits mitogeninduced T cell proliferation by selectively inhibiting macrophage costimulatory function. J Immunol 1992;148:3133–3139. 139. Ding L, Linsley PS, Huang LY, Germain RN, Shevach EM. IL-10 inhibits macrophage costimulatory activity by selectively inhibiting the upregulation of B7 expression. J Immunol 1993;151:1224–1234. 140. Fiorentino DF, Zlotnik A, Mosmann TR, Howard M, O’Garra A. IL-10 inhibits cytokine production by activated macrophages. J Immunol 1991;147:3815–3822. 141. Fiorentino DF, et al. IL-10 acts on the antigenpresenting cell to inhibit cytokine production by Th1 cells. J Immunol 1991;146:3444–3451.

85

Jasenosky et al  T cells and the human immune response to TB

142. Mayer-Barber KD, et al. Innate and adaptive interferons suppress IL-1alpha and IL-1beta production by distinct pulmonary myeloid subsets during Mycobacterium tuberculosis infection. Immunity 2011;35:1023–1034. 143. McNab FW, et al. Type I IFN induces IL-10 production in an IL-27-independent manner and blocks responsiveness to IFN-gamma for production of IL-12 and bacterial killing in Mycobacterium tuberculosis-infected macrophages. J Immunol 2014;193:3600–3612. 144. Chegou NN, et al. Utility of host markers detected in Quantiferon supernatants for the diagnosis of tuberculosis in children in a highburden setting. PLoS ONE 2013;8:e64226. 145. Phalane KG, et al. Differential expression of host biomarkers in saliva and serum samples from individuals with suspected pulmonary tuberculosis. Mediators Inflamm 2013;2013:981984. 146. Djoba Siawaya JF, et al. Differential cytokine/ chemokines and KL-6 profiles in patients with different forms of tuberculosis. Cytokine 2009;47:132–136. 147. Elkington P, et al. MMP-1 drives immunopathology in human tuberculosis and transgenic mice. J Clin Invest 2011;121:1827– 1833. 148. Jacobsen M, et al. Candidate biomarkers for discrimination between infection and disease caused by Mycobacterium tuberculosis. J Mol Med 2007;85:613–621. 149. Berry MP, et al. An interferon-inducible neutrophil-driven blood transcriptional signature in human tuberculosis. Nature 2010;466:973– 977. 150. Maertzdorf J, et al. Functional correlations of pathogenesis-driven gene expression signatures in tuberculosis. PLoS ONE 2011;6:e26938. 151. Maertzdorf J, et al. Human gene expression profiles of susceptibility and resistance in tuberculosis. Genes Immun 2011;12:15–22. 152. Mistry R, et al. Gene-expression patterns in whole blood identify subjects at risk for recurrent tuberculosis. J Infect Dis 2007;195:357–365. 153. Bloom CI, et al. Transcriptional blood signatures distinguish pulmonary tuberculosis, pulmonary sarcoidosis, pneumonias and lung cancers. PLoS ONE 2013;8:e70630. 154. Anderson ST, et al. Diagnosis of childhood tuberculosis and host RNA expression in Africa. N Engl J Med 2014;370:1712–1723. 155. Stanley SA, Johndrow JE, Manzanillo P, Cox JS. The Type I IFN response to infection with Mycobacterium tuberculosis requires ESX-1mediated secretion and contributes to pathogenesis. J Immunol 2007;178: 3143–3152. 156. Ordway D, et al. The hypervirulent Mycobacterium tuberculosis strain HN878 induces a potent TH1 response followed by rapid down-regulation. J Immunol 2007;179:522–531. 157. Manca C, et al. Hypervirulent M. tuberculosis W/Beijing strains upregulate type I IFNs and increase expression of negative regulators of the

86

158.

159.

160.

161.

162.

163.

164.

165.

166.

167.

168.

169.

170.

171.

172.

Jak-Stat pathway. J Interferon Cytokine Res 2005;25:694–701. Manca C, et al. Virulence of a Mycobacterium tuberculosis clinical isolate in mice is determined by failure to induce Th1 type immunity and is associated with induction of IFN-alpha/beta. Proc Natl Acad Sci USA 2001;98:5752–5757. Antonelli LR, et al. Intranasal Poly-IC treatment exacerbates tuberculosis in mice through the pulmonary recruitment of a pathogen-permissive monocyte/macrophage population. J Clin Invest 2010;120:1674–1682. Dorhoi A, et al. Type I IFN signaling triggers immunopathology in tuberculosis-susceptible mice by modulating lung phagocyte dynamics. Eur J Immunol 2014;44:2380–2393. Redford PS, et al. Influenza A virus impairs control of Mycobacterium tuberculosis coinfection through a type I interferon receptordependent pathway. J Infect Dis 2014;209:270– 274. Mayer-Barber KD, et al. Host-directed therapy of tuberculosis based on interleukin-1 and type I interferon crosstalk. Nature 2014;511:99–103. Desvignes L, Wolf AJ, Ernst JD. Dynamic roles of type I and type II IFNs in early infection with Mycobacterium tuberculosis. J Immunol 2012;188:6205–6215. Teles RM, et al. Type I interferon suppresses type II interferon-triggered human anti-mycobacterial responses. Science 2013;339:1448–1453. Maertzdorf J, et al. Common patterns and disease-related signatures in tuberculosis and sarcoidosis. Proc Natl Acad Sci USA 2012;109:7853–7858. Ottenhoff TH, et al. Genome-wide expression profiling identifies type 1 interferon response pathways in active tuberculosis. PLoS ONE 2012;7:e45839. Geldmacher C, et al. Preferential infection and depletion of Mycobacterium tuberculosis-specific CD4 T cells after HIV-1 infection. J Exp Med 2010;207:2869–2881. Geldmacher C, et al. Early depletion of Mycobacterium tuberculosis-specific T helper 1 cell responses after HIV-1 infection. J Infect Dis 2008;198:1590–1598. Juffermans NP, et al. Patients with active tuberculosis have increased expression of HIV coreceptors CXCR4 and CCR5 on CD4(+) T cells. Clin Infect Dis 2001;32:650–652. Shalekoff S, Pendle S, Johnson D, Martin DJ, Tiemessen CT. Distribution of the human immunodeficiency virus coreceptors CXCR4 and CCR5 on leukocytes of persons with human immunodeficiency virus type 1 infection and pulmonary tuberculosis: implications for pathogenesis. J Clin Immunol 2001;21:390–401. Santucci MB, et al. Expansion of CCR5+ CD4+ Tlymphocytes in the course of active pulmonary tuberculosis. Eur Respir J 2004;24:638–643. Rosas-Taraco AG, Arce-Mendoza AY, CaballeroOlin G, Salinas-Carmona MC. Mycobacterium tuberculosis upregulates coreceptors CCR5 and CXCR4 while HIV modulates CD14 favoring concurrent infection. AIDS Res Hum Retroviruses 2006;22:45–51.

173. Wolday D, et al. Expression of chemokine receptors CCR5 and CXCR4 on CD4+ T cells and plasma chemokine levels during treatment of active tuberculosis in HIV-1-coinfected patients. J Acquir Immune Defic Syndr 2005;39:265–271. 174. Kabue JP, de Swardt D, de Beer C, Glashoff RH. Short-term antiretroviral therapy fails to reduce the expanded activated CCR5-expressing CD4+ T lymphocyte population or to restore the depleted naive population in chronically HIV-infected individuals with active pulmonary tuberculosis. AIDS Res Hum Retroviruses 2013;29:769–777. 175. Ranjbar S, Ly N, Thim S, Reynes JM, Goldfeld AE. Mycobacterium tuberculosis recall antigens suppress HIV-1 replication in anergic donor cells via CD8+ T cell expansion and increased IL-10 levels. J Immunol 2004;172:1953–1959. 176. Ranjbar S, Boshoff HI, Mulder A, Siddiqi N, Rubin EJ, Goldfeld AE. HIV-1 replication is differentially regulated by distinct clinical strains of Mycobacterium tuberculosis. PLoS ONE 2009;4:e6116. 177. Day JH, et al. Does tuberculosis increase HIV load? J Infect Dis 2004;190:1677–1684. 178. Barnabas RV, Webb EL, Weiss HA, Wasserheit JN. The role of coinfections in HIV epidemic trajectory and positive prevention: a systematic review and meta-analysis. AIDS 2011;25: 1559–1573. 179. Day CL, et al. Detection of polyfunctional Mycobacterium tuberculosis-specific T cells and association with viral load in HIV-1-infected persons. J Infect Dis 2008;197:990–999. 180. Sutherland JS, et al. Polyfunctional CD4(+) and CD8(+) T cell responses to tuberculosis antigens in HIV-1-infected patients before and after antiretroviral treatment. J Immunol 2010;184:6537– 6544. 181. Kalsdorf B, et al. HIV-1 infection impairs the bronchoalveolar T-cell response to mycobacteria. Am J Respir Crit Care Med 2009;180: 1262–1270. 182. Jambo KC, et al. Bronchoalveolar CD4+ T cell responses to respiratory antigens are impaired in HIV-infected adults. Thorax 2011;66:375–382. 183. Jambo KC, et al. Asymptomatic HIV-infected Individuals on Antiretroviral Therapy Exhibit Impaired Lung CD4(+) T-Cell Responses to Mycobacteria. Am J Respir Crit Care Med 2014;190:938–947. 184. Lahey T, et al. Interferon gamma responses to mycobacterial antigens protect against subsequent HIV-associated tuberculosis. J Infect Dis 2010;202:1265–1272. 185. Diedrich CR, et al. Reactivation of latent tuberculosis in cynomolgus macaques infected with SIV is associated with early peripheral T cell depletion and not virus load. PLoS ONE 2010;5: e9611. 186. Mattila JT, Diedrich CR, Lin PL, Phuah J, Flynn JL. Simian immunodeficiency virus-induced changes in T cell cytokine responses in cynomolgus macaques with latent Mycobacterium tuberculosis infection are associated with timing of reactivation. J Immunol 2011;186:3527–3537.

© 2015 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd Immunological Reviews 264/2015

Jasenosky et al  T cells and the human immune response to TB

187. Lai RP, Nakiwala JK, Meintjes G, Wilkinson RJ. The immunopathogenesis of the HIV tuberculosis immune reconstitution inflammatory syndrome. Eur J Immunol 2013;43:1995–2002. 188. Bourgarit A, et al. Explosion of tuberculinspecific Th1-responses induces immune restoration syndrome in tuberculosis and HIV coinfected patients. Aids 2006;20: F1–F7. 189. Meintjes G, Rabie H, Wilkinson RJ, Cotton MF. Tuberculosis-associated immune reconstitution inflammatory syndrome and unmasking of tuberculosis by antiretroviral therapy. Clin Chest Med 2009;30:797–810. 190. Elliott JH, et al. Immunopathogenesis and diagnosis of tuberculosis and tuberculosisassociated immune reconstitution inflammatory syndrome during early antiretroviral therapy. J Infect Dis 2009;200:1736–1745. 191. Vignesh R, et al. TB-IRIS after initiation of antiretroviral therapy is associated with expansion of preexistent Th1 responses against Mycobacterium tuberculosis antigens. J Acquir Immune Defic Syndr 2013;64:241–248. 192. Bourgarit A, et al. Tuberculosis-associated immune restoration syndrome in HIV-1-infected patients involves tuberculin-specific CD4 Th1 cells and KIR-negative gammadelta T cells. J Immunol 2009;183:3915–3923. 193. Chakrabarti LA, et al. Biomarkers of CD4+ T cell activation as risk factors for tuberculosisassociated immune reconstitution inflammatory syndrome. AIDS 2014;28:1593–1602. 194. Blanc FX, et al. Earlier versus later start of antiretroviral therapy in HIV-infected adults with

195.

196.

197.

198.

199.

200.

201.

202.

203.

tuberculosis. N Engl J Med 2011;365:1471– 1481. Haridas V, et al. TB-IRIS, T-cell activation, and remodeling of the T-cell compartment in highly immunosuppressed HIV-infected patients with TB. AIDS 2015;29:263–273. Soares AP, et al. Bacillus Calmette-Guerin vaccination of human newborns induces T cells with complex cytokine and phenotypic profiles. J Immunol 2008;180:3569–3577. Soares AP, et al. Longitudinal changes in CD4(+) T-cell memory responses induced by BCG vaccination of newborns. J Infect Dis 2013;207:1084–1094. Murray RA, et al. Bacillus Calmette Guerin vaccination of human newborns induces a specific, functional CD8+ T cell response. J Immunol 2006;177:5647–5651. Lutwama F, et al. Distinct T-cell responses when BCG vaccination is delayed from birth to 6 weeks of age in Ugandan infants. J Infect Dis 2014;209:887–897. Kagina BM, et al. Delaying BCG vaccination from birth to 10 weeks of age may result in an enhanced memory CD4 T cell response. Vaccine 2009;27:5488–5495. Pathan AA, et al. Boosting BCG with recombinant modified vaccinia ankara expressing antigen 85A: different boosting intervals and implications for efficacy trials. PLoS ONE 2007;2:e1052. O’Garra A, et al. The immune response in tuberculosis. Annu Rev Immunol 2013;31:475– 527. Abel B, et al. The novel tuberculosis vaccine, AERAS-402, induces robust and polyfunctional

© 2015 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd Immunological Reviews 264/2015

204.

205.

206.

207.

208.

209.

210.

CD4+ and CD8+ T cells in adults. Am J Respir Crit Care Med 2010;181:1407–1417. Day CL, et al. Induction and regulation of T-cell immunity by the novel tuberculosis vaccine M72/AS01 in South African adults. Am J Respir Crit Care Med 2013;188:492–502. Scriba TJ, et al. Modified vaccinia Ankaraexpressing Ag85A, a novel tuberculosis vaccine, is safe in adolescents and children, and induces polyfunctional CD4+ T cells. Eur J Immunol 2010;40:279–290. Grode L, Ganoza CA, Brohm C, Weiner J 3rd, Eisele B, Kaufmann SH. Safety and immunogenicity of the recombinant BCG vaccine VPM1002 in a phase 1 open-label randomized clinical trial. Vaccine 2013;31:1340–1348. Tameris MD, 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;381:1021–1028. Gold MC, et al. MR1-restricted MAIT cells display ligand discrimination and pathogen selectivity through distinct T cell receptor usage. J Exp Med 2014;211:1601–1610. Joosten SA, et al. Mycobacterium tuberculosis peptides presented by HLA-E molecules are targets for human CD8 T-cells with cytotoxic as well as regulatory activity. PLoS Pathog 2010;6: e1000782. Spencer CT, et al. Granzyme A produced by gamma(9)delta(2) T cells induces human macrophages to inhibit growth of an intracellular pathogen. PLoS Pathog 2013;9: e1003119.

87

T cells and adaptive immunity to Mycobacterium tuberculosis in humans.

The adaptive immune response mediated by T cells is critical for control of Mycobacterium tuberculosis (M. tuberculosis) infection in humans. However,...
175KB Sizes 0 Downloads 9 Views