Abstract

Infection with Mycobacterium tuberculosis predominantly establishes subclinical latent infection over the lifetime of an individual, with a fraction of infected individuals rapidly progressing to active disease. The immune control in latent infection can be perturbed by comorbidities such as diabetes mellitus, obesity, smoking, and coinfection with helminthes or HIV. Modeling the varying aspects of natural infection remains incomplete when using zebrafish and mice. However, the nonhuman primate model of tuberculosis offers a unique and accurate model to investigate host responses to infection, test novel therapeutics, and thoroughly assess preclinical vaccine candidates. Rhesus macaques and cynomolgus macaques manifest the full gamut of clinical and pathological findings in human Mycobacterium tuberculosis infection, including the ability to co-infect macaques with Simian Immunodeficiency Virus to model HIV co-infection. Here we discuss advanced techniques to assay various clinical outcomes of the natural progression of infection as well as therapeutics in development and novel preclinical vaccines. Finally, we survey the translational aspects of nonhuman primate research and argue the urgent need to thoroughly examine preclinical therapeutics and vaccines using this model prior to clinical implementation.

Introduction

The nonhuman primate (NHP) model of tuberculosis remains the gold standard for modeling Mycobacterium tuberculosis (Mtb) infection. Rhesus macaques, Macaca mulatta, and Cynomolgus macaques, Macaca fascicularis, have long been used in experimental tuberculosis research. Macaques were first used in the late 1960s when scientists hypothesized that macaques increased susceptibility to natural tuberculosis infection could be utilized to rapidly test therapeutics and to study natural and vaccine-induced immunity to infection (Clarke 1968; Good 1968; Schmidt 1966). The first vaccine study occurred in the early 1970s and sought to test the efficacy of the already 50-year-old Bacille Calmette-Guerin vaccine through intravenous administration followed by aerosol infection with virulent Mtb (Barclay et al. 1970, 1973). This study, among others, brought the NHP model to the forefront of tuberculosis research, and has since been ever-growing in importance for clinical application of novel therapeutics and preclinical testing of vaccines (Pena and Ho 2016). This review focuses on the strides NHP research has brought to the field of tuberculosis research, the adaptation of the model to advanced technologies, and the clinical implementation of current ongoing NHP research.

The Development of the NHP Model and its Use

Rationale for Use of NHPs

While humans have been coevolving with Mtb throughout our long history, mice are not natural hosts of Mtb. Mice have not experienced the genetic evolutionary pressure that would have selected for increased control of Mtb infection, and are thus permissively susceptible to infection characterized by severe dissemination of bacteria (Kramnik and Beamer 2016). However, due to their high genetic similarity to humans, the macaque response to Mtb accurately models all aspects of tuberculosis disease. Critical amongst these are the ability to develop long-term subclinical latent disease due to local immune control and to progress to clinically apparent active disease (Darrah et al. 2014; Dutta et al. 2010; Foreman et al. 2016; Kaushal et al. 2015; Mehra et al. 2012, 2013, 2015; Phillips et al. 2015; Slight et al. 2013). In comparison to human tuberculosis and in contrast to mouse models, macaques form highly structured granulomas with readily-apparent lymphocytic and macrophage tropic layers, central caseation, and severe neutrophilic immunopathology seen in active disease (Mehra et al. 2013). The clinical manifestations mimic those seen in human tuberculosis infection, including dyspnea, pyrexia, cachexia, positive PPD and IGRA responses, detectible bacilli in sputum, and pneumatic inflammation upon x-ray imaging (Capuano et al. 2003; Dutta et al. 2010; Gormus et al. 2004; Kaushal et al. 2015; Mehra et al. 2012). Furthermore, while the immune mediators required for the control of pulmonary bacterial infection are present in both mice and primates, subtle differences exist. These include differences in specific macrophage killing mechanisms and disparate utilization of several immunosuppressive pathways (Su et al. 2014). This has led to debate about the specific mechanisms of control of Mtb infection and their relative importance in mice vs humans (Liu et al. 2006; Nathan 2006; Thoma-Uszynski et al. 2001). The progression of infection in the wake of immune response optimal for a specific model can thus generate diverse endpoints. This discourse led to the refrain that some of the most important experiments can be performed only in mice and not humans, reflecting a key gap that precluded progress in understanding both the pathogenesis of and immunity to tuberculosis. Immunological studies have, however, highlighted the overlapping immune responses to Mtb infection both in peripheral blood and cells isolated from bronchoalveolar lavage, thus demonstrating the immune response in the macaque model parallels that in humans (Ernst et al. 2012; Mothe et al. 2015). In addition, Mtb-infected macaques demonstrate remarkable heterogeneity in both disease outcomes (Capuano et al. 2003; Mehra et al. 2015) as well as granuloma pathology (Kaushal et al. 2012). Hence, studies in macaques can fill an important gap between the mouse model of tuberculosis and humans.

Numerous reasons, however, historically restricted the use of macaques more widely to study the progression of infection and to understand correlates of disease. These included expense, requirement of specialized containment facilities and husbandry staff, and lack of availability. The AIDS epidemic resulted in the development of the macaque models of simian immunodeficiency virus (SIV) infection to primarily study HIV infection and this was conducted mostly by one of the several NIH-supported National Primate Centers. However, biosafety concerns initially kept these centers from developing tuberculosis research programs. UCLA (Walsh et al. 1996) and the University of Pittsburgh (Capuano et al. 2003) were amongst the first facilities in the modern era to study tuberculosis in macaques. While the use of primates can often be limiting due to capabilities of facilities and monetary resources, throughout this review the authors suggest that all novel therapeutics and vaccines ought to be tested in the NHP model as a checkpoint prior to clinical implementation. Though the immune response and subsequent clinical manifestation to infection mimics that of humans, there are subtle differences between rhesus and cynomolgus macaques, including strengths and weaknesses for each species that must be considered when designing NHP studies.

Different Species of Macaques and their Use in the Tuberculosis Model

Rhesus macaques were initially the dominant species used to establish the NHP model of tuberculosis (Barclay et al. 1970, 1973; Good 1968; Janicki et al. 1973), largely due to their relative ease of maintaining and breeding in captivity and use in alternative modeling including polio (Sabin 1957). In the first studies, rhesus macaques were experimentally infected with the H37Rv strain of Mtb, causing the majority of animals to progress to active disease within 50 to 60 days postinfection. Since then, with lower doses and differing strains of Mtb, rhesus macaques have been shown to establish latent, asymptomatic infection determined by tuberculin skin test conversion and assessment of lungs for persistent bacteria months after infection (Foreman et al. 2016; Mehra et al. 2011). However, when challenged with more virulent strains such as Erdman K01, rhesus macaques not only develop higher bacterial burdens but increased pathology over time (Sharpe et al. 2016, 2009). Vaccination with BCG provides increased protection in cynomolgus macaques when challenged with a high dose of Erdman. These animals in general exhibit lower bacterial burdens relative to BCG-vaccinated rhesus macaques (Langermans et al. 2001). This increased susceptibility to highly virulent strains is not detrimental to the rhesus macaque model, however, as it provides an accurate model for rapid testing of vaccine candidates wherein protection in a more susceptible species may provide greater justification for clinical testing of the preclinical vaccine.

The first studies utilizing cynomolgus macaques established a chronic tuberculosis model when using very low doses (10 colony forming units) of the virulent Erdman strain (Walsh et al. 1996). This model was further tested up to 20 months post-low-dose infection with Erdman, where researchers described latent, active, and spontaneously reactive (latently infected progressing to active disease) as synonymous to human tuberculosis disease (Capuano et al. 2003; Lin et al. 2009). The cynomolgus model has since been rapidly exploited for vaccine studies, which used moderate doses of Erdman (Mehra et al. 2013; Okada et al. 2007; Reed et al. 2009). Similarly, the rhesus macaque model, using both high doses of low-virulence strains like CDC1551 or low doses of high-virulence strains like Erdman, has been used in vaccine development efforts (Darrah et al. 2014; Kaushal et al. 2015). As studies delineate the subtle differences in the progression of Mtb infection in these two species, it is becoming increasingly clear that both provide insights into the different aspects of human disease syndrome that must be protected against.

Routes of Vaccination and Infection

Throughout the NHP model there have been two primary routes of challenge or vaccination. The vast majority of studies have used intra-tracheal or intra-bronchial instillation of bacteria, which allows for tighter control of the presented dose but only allows deposition of bacteria in isolated pulmonary lobes. However, recent studies have established an aerosol infection route, which provides inherent variables but is more representative of the natural route of infection. These variables include the rate and volume at which the animal is breathing; however, this route exposes bacteria to all lobes of the lung. Controlling for the time (and thus the volume of infectious air the animal breathes) allows for increased control of low infectious doses such as 10 to 20 CFU (Foreman et al. 2016).

It has been shown in lower animal models, as well as in rhesus macaques more recently, that aerosol-delivered BCG provides increased protection when compared to intradermal delivery (Kaushal et al. 2015; Sharpe et al. 2016; White et al. 2015). This may point to the value in eliciting localized adaptive responses. As clinicians continue to advocate for mucosal-delivered vaccines, the NHP model must continue to also test, and even compare, route of vaccination to hone in on optimal dose and route of protective vaccines. It is accepted that aerosol delivery of vaccines contains inherent difficulties in logistics and implementation, especially in the regions of the world where vaccination is most urgently needed. Therefore, the NHP model serves as an optimal model for testing alternative vaccine strategies such as intranasal, sublingual, oral, or adjuvanted intramuscular/intradermal routes. Alternative vaccine delivery routes have already been tested in rhesus macaques albeit to induce immunity to SIV (Veazey et al. 2015).

The Use of Different Strains of Mtb in the NHP Model

Due to genetic differences between mice and macaques, at least some immune responses to Mtb are divergent in these model systems. Recent work in the NHP model has not only demonstrated that macaques recapitulate the human tuberculosis syndrome (Flynn et al. 2015; Kaushal and Mehra 2012; Kaushal et al. 2012; Lin et al. 2006, 2009; Pena and Ho 2016), but also established key differences in the pathogenesis of disease with rodent models. This could be due to differential induction of key virulence and adaptation programs in the bacilli such as hypoxia, which is found in well-organized necrotic granulomas. As previously described, no major pathological differences were observed when BALB/c and C57Bl/6 mice (Rustad et al. 2008) were infected with Mtb strains deficient in hypoxia response genes (dosR, dosS, dosT). However, when these same strains were used to infect rhesus macaques, there were significant differences in pathology, bacterial burden, as well as the immune response (Mehra et al. 2015). Furthermore, a mutant of Mtb deficient in its ability to scavenge oxidative stress (MtbΔsigH) was not restricted in bacterial replication in the lungs of C57Bl/6 mice (Kaushal et al. 2002), but failed to grow in rhesus macaque lungs (Mehra et al. 2012). These results strongly suggest that Mtb bacilli experience greater stress in primate tissues relative to their murine counterparts and demonstrate that the NHP model can additionally be used to screen mutant strains of Mtb, particularly in genes that may not be particularly necessary in the mouse model. The NHP model can further be utilized to assay multiple gene knockouts via infection with transposon mutant libraries (Dutta et al. 2010). When such infections were compared between macaques and mice, it was found that the growth of a significantly larger frequency of Tn-mutants was interrupted in the former, relative to the latter model system, again underscoring greater and/or wider variety of stress in macaques. There are some advantages of the mouse model, however, especially in reagent optimization, which may not be possible to perform using macaques. One such contrasting attribute that mice permit is the ability to test novel PET tracers more effectively in mice (Jain 2017; Weinstein et al. 2014). Such screening experiments are impractical to perform in NHPs, because these molecules are synthesized in minute amounts and leave the animal radioactive (generally requiring sacrifice and radioactive waste disposal).

With each bacillus capable of establishing a single granuloma, the macaque lung can be assayed for bacterial persistence, pathogenicity and attenuation, and novel immune phenotypes such as development of inducible bronchus associated lymphoid tissue (Gopal et al. 2013; Kaushal et al. 2015; Slight et al. 2013). Using barcoded strains of Mtb and low-dose, intrabronchial infection, the NHP model was used to track the intralobar, intrapulmonary, and extrapulmonary migration of individual bacilli. This study was the first to show that phagocytic cells traffic from the individual granulomas to the draining lymph nodes where multiple barcodes can often be found within a single bronchial lymph node (Lin et al. 2014). Complementing these approaches, this model has recently been leveraged to study the transcriptome of the pathogen in its intra-granulomatous niches (Hudock et al. 2017) using a microdissection coupled with microarray analysis approach. This result suggests that the genes of the DosR regulon are expressed in virtually all lesion types. These would include both early lesions as well as lesions during the acute infection of lungs. However, the extent of DosR-regulon expression was higher in discrete, caseating lesions from animals with asymptomatic, latent infection. The results parallel with both the recently uncovered phenotype of this regulon in models that generate human-like lung pathology (Gautam et al. 2014; Malhotra et al. 2004; Mehra et al. 2015), but also recent knowledge of Mtb gene expression in humans (Commandeur et al. 2013; Walter et al. 2016). These studies open the possibility that in the near future, the field may be able to leverage recent progress in bar coding, high-throughput sequencing, and RNAseq to better understand single cell Mtb transcriptomics within individual compartments of the various pathological niches. These fundamental experiments should extend our understanding of the physiology of the pathogen in its natural environment, eventually leading to proteome- and lipidome-wide studies.

Modeling Comorbidities

One highly unique aspect of the NHP model of tuberculosis is the ability to co-infect macaques with SIV, as a model of human HIV co-infection (Diedrich et al. 2010; Foreman et al. 2016). The macaque model of SIV infection has been well established since the emergence of AIDS in the 1980s. Much of the initial work in this direction was performed in the “pathogenic” model involving either rhesus or pigtailed macaques (Macaca nemestrina) infected with strains of SIV that resulted in rapid immunodeficiency. The rhesus and cynomolgus model clearly showed that SIV infection results in a significant depletion of CD4+ T cells from the blood, but even more profoundly impacts lymphoid tissues, specifically the gut. This work had a significant impact on the field and has since led to the testing of remarkable therapeutic and vaccination approaches (Apetrei et al. 2012; Haase 2011; McNicholl 2016; Shedlock et al. 2009). There has now emerged a specific interest in the nonpathogenic model of SIV co-infection, using sooty mangabeys (Cercocebus atys), African green monkeys (Cercopithecus aethiops), or other such species that do not exhibit signs of rapid pathogenic infection; however, co-infection with Mtb has not been modeled using the species less susceptible to SIV infection. Mtb/SIV co-infection studies have primarily used rhesus (Foreman et al. 2016; Mehra et al. 2011) or cynomolgus (Diedrich et al. 2010; Mattila et al. 2011) macaques, where results have highlighted that while CD4+ T-cells play a critical role in mediating protection from SIV-induced reactivation, especially by regulating the timing of cytokine signaling (Coleman et al. 2014a), other CD4+ independent mechanisms such CD8+ T-cells and B-cells may compensate for the loss of CD4+ T-cells (Foreman et al. 2016). Work in this area also suggests that treatment of mycobacteria/SIV co-infection by antiretroviral therapy can control reactivation by restoring Mtb-specific T-cells (Shen et al. 2001), although it is possible that all T-cells with all antigen-specificities may not be completely restored. It is expected that in near future, this model will be extended to include mechanistic studies on viral-mediated dissemination to extrathoracic regions, to model immune reconstitution inflammatory syndrome, and to further understand the role of CD4+-independent mechanisms of protection from tuberculosis.

In addition, the NHP model of tuberculosis lends itself well to the study of various other co-infections and comorbidities. In preliminary studies, macaques that received long-term alcohol exposure exhibited reduced levels of key correlates of immunity to tuberculosis and progressed more rapidly to acute disease (Kaushal D, unpublished observations). Alcoholism has long been associated with the progression of human tuberculosis (Lonnroth et al. 2008). It is possible that the impact of chronic cigarette smoke exposure on Mtb infection can also be studied in this model. Recent data indicate that cigarette smoke exposure has a profound effect on macrophage function in the context of Mtb infection (O'Leary et al. 2014), and long-term exposure to cigarette smoke can be modeled in macaques (Polverino et al. 2015). Moreover, it has recently been demonstrated that lysosomal storage impairment impairs macrophage migration causing granuloma breakdown and disease expansion (Berg et al. 2016). It is tempting to speculate that that comparable phenomenon may be at play in smokers. Finally, it is possible that the control of SIV (HIV)-mediated reactivation of LTBI may also similarly depend on macrophage dysfunction (D. Kaushal, M. Kuroda, unpublished observations).

Our understanding of the role played by central metabolic processes in regulating immunity in general and particularly Mtb-specific immune responses has increased. Thus, we understand that diabetes mellitus can impact the progression of tuberculosis via different mechanisms. These data have prompted the use of Metformin, a key DM drug, as an adjunctive therapy for tuberculosis (Singhal et al. 2014). Similarly, the role of key cellular metabolic processes and pathways like sirtuins (Singhal et al., unpublished data), mTOR, poly (ADP-ribose) polymerases, DAMP inhibitors, etc. is now being studied in this model. In this vein, in vivo inhibition of indoleamine dioxygenase (IDO1) activity in rhesus macaques infected with Mtb, using a safe and approved compound, resulted in reduction of disease, pathology, and bacterial levels, concomitant with enhanced Th1 responses (S Mehra et al, unpublished data). IDO1 encodes a tryptophan catabolizing activity as an innate strategy to starve pathogens of this essential amino acid. IDO1 is also a powerful immunosuppressor of T-cell activity, since Trp depletion can impact the development of rapidly proliferating Th1 effectors (Munn et al. 1999). Mtb can, however, synthesize its own Trp (Zhang and Rubin 2013; Zhang et al. 2013), and thus the induction of IDO1 in Mtb-infected lesions is unable to contain bacillary replication; rather it dampens productive immune responses. A IDO1-/- knockout strain in B6 mice did not exhibit any differences in terms of Mtb replication and pathology relative to parental mice. These results again underscore the value of studying Mtb-host interplay in the setting of human-like lung granulomas.

Technological Advances

Recently, technologies have emerged that permit big data acquisition. Similarly, multiple platforms for high-throughput data analysis have become available. NHPs are a valuable model for vetting determining the value of these technologies for diagnostics, therapeutics, and preventative measures. One such technology, fluorine-18 fluorodeoxyglucose (18F-FDG) positron emission tomography with computer tomography (PET/CT), has emerged as a pertinent tool for measuring granuloma dynamics, determining study data output, and consistent multisite comparison of data. Tubercular lesions were originally misdiagnosed as small cell carcinomas or primary lung cancer in human patients being screened by 18F-FDG PET/CT (Hofmeyr et al. 2007). This finding suggested an exciting potential to monitor the progression of tuberculosis disease. 18F-FDG incorporates into cells with increased glycolysis such as inflammatory macrophages and lymphocytes, and this finding led to the first Phase I trial to test the efficacy of PET/CT as a noninvasive biomarker of therapeutic responses (Martinez et al. 2012) and was rapidly followed up with a confirmatory study using the cynomolgus model and a four-drug regimen where the animals were serially imaged using PET/CT to track both disease progression and chemotherapeutic responses (Lin et al. 2013). PET/CT was further established to accurately measure pathology findings when the marmoset, Callithrix jacchus, model of tuberculosis was developed (Via et al. 2013). These initial studies spurred a movement to incorporate PET/CT analysis into all preclinical trials using the NHP model. However, this imaging platform comes with inherent challenges such as establishing facilities compliant for both radiographic material (e.g., 18F-FDG) and biosafety level 3 animal housing (Scanga et al. 2014). Furthermore, data analysis of PET/CT imaging from multiple studies at multiple locations must be standardized to conform to human clinical trial standards. Once implemented, PET/CT imaging can be used as a worldwide standard readout of prophylactic and therapeutic efficacy in vaccine and drug trials using the NHP model.

Due to the integrity of the NHP model, PET/CT imaging can also be utilized to track natural disease progression. Using the cynomolgus model of SIV co-infection, researchers demonstrated that early changes in PET/CT-imaged granulomas predict the long-term clinical outcomes (Coleman et al. 2014b) and established a correlative readout for biomarker discovery (Gideon et al. 2016). The dose and route of infection can further be tailored to address specific questions PET/CT imaging can analyze such as intrapulmonary spread of bacilli, individual granuloma plasticity, and perturbation of granuloma integrity by comorbidities. As more radioactive reagents are made with differing specificities (e.g., 18F-FDG for glycolysis, α-CD3 radioactive-labeled antibody for T-cells), PET/CT imaging can be implemented not only as a standard readout for large-scale drug or vaccine studies, but also to produce data for hypothesis generating, exploratory, or mechanistic NHP studies.

Translating from the NHP Model to Clinical Vaccine Efficacy Trials

The NHP model has served to advance our understanding about the many key aspects of tuberculosis disease and infection, and through the established integrity of the model will continue to serve as a prominent checkpoint of preclinical candidates. While BCG was already 50 years old, the first NHP vaccine study tested intravenous delivery of BCG and monitored the primates after aerosol challenge with the virulent strain H37Rv. Since then macaques have served as the premier model system to study immune responses and efficacy of vaccine candidates. Several of these candidates then moved to human clinical trials (Table 1). The H56 vaccine, a subunit fusion protein of Ag85B, ESAT-6, and Rv2660c delivered with IC31 adjuvant, is one such vaccine candidate that progressed through mouse models into the NHP model. Animals were primed with BCG and subsequently boosted with H56:IC31 and compared to unvaccinated and unboosted animals. H56:IC31-vaccinated animals survived longer and had reduced pathology and bacterial burden after challenge (Lin et al. 2012). Subsequent studies further examined the stimulation of adjuvant by priming with BCG and boosting with H56 mixed with four different adjuvants (Billeskov et al. 2016). These studies provided significant justification for progression into clinical trials where the findings of these NHP studies could be validated. H56:IC31 has now progressed through Phase I clinical trials (Luabeya et al. 2015) and continuing into Phase II prevention of tuberculosis infection trials with extremely promising, albeit preliminary, results. Additionally, ID93, a subunit vaccine comprised of Mtb proteins Rv3619, Rv1813, Rv3620, and Rv2608, adjuvanted with GLA-SE, a synthetic TLR4 agonist, has also been tested in multiple preclinical animal models including macaques (Bertholet et al. 2010). Instead of utilizing the macaque model to test the prevention of tuberculosis disease, the study was designed to use the vaccine therapeutically after antibacterial chemotherapy drug use. After high-dose infection, macaques were treated with a suboptimal chemotherapy regimen for 4 weeks and subsequently vaccinated with ID93:GLA-SE. Animals that were vaccinated therapeutically displayed significantly less bacterial burden and radiological evidence of disease compared to treated but unvaccinated animals (Coler et al. 2013). Subsequently this vaccine candidate has progressed into the first human trial to therapeutically vaccinate infected humans (Clinical trials ID NCT02465216) (Table 1). Another vaccine currently being considered is MTBVAC (Martin et al. 2006; Verreck et al. 2009), which is based on the deletion of the phoP allele (Perez et al. 2001) and fadD26 (Arbues et al. 2013). This strain has proven to be safe in a Phase I trial (Spertini et al. 2015) and is currently being further evaluated in Phase II (Table 1).

Table 1

Translation use of the NHP model

Vaccines in clinical trialsTypePhaseNHP safety studyNHP efficacy studyChallenge
M. vaccae (SRL 172)Whole-cell M. vaccaeIII
M. indicus praniiWhole-cell M. indicus praniiIII
M72/AS01Protein/adjuvantIIb++500 CFU Erdman
Aeras-402Viral vectorIIb++275 CFU Erdman
VPM1002Live-recombinant BCGIIa+
H4:IC31Protein/adjuvantIIa
H1/H56:IC31Protein/adjuvantIIa++500/25 CFU Erdman
ID93 + GLA-SEProtein/adjuvantIIa+
MTBVACLive-attenuated M. tuberculosisIIa++1000 CFU Erdman
RUTIWhole-cell fractionated M. tuberculosisIIa
Dar-901Whole-cell M. obuenseI
Ad5 Ag85AViral vectorI
Crucell Ad35 + MVA85AViral vectorI
ChAdOc1.85A + MVA85AViral vectorI
MVA85A-AerosolViral vectorI+
MVA85A-IMX313Viral vector/adjuvantI+
TB/FLU-04LViral vectorI
Vaccines previously in clinical trials
BCG-MVA85A boostViral vector boost++1000 CFU Erdman
Vaccines in clinical trialsTypePhaseNHP safety studyNHP efficacy studyChallenge
M. vaccae (SRL 172)Whole-cell M. vaccaeIII
M. indicus praniiWhole-cell M. indicus praniiIII
M72/AS01Protein/adjuvantIIb++500 CFU Erdman
Aeras-402Viral vectorIIb++275 CFU Erdman
VPM1002Live-recombinant BCGIIa+
H4:IC31Protein/adjuvantIIa
H1/H56:IC31Protein/adjuvantIIa++500/25 CFU Erdman
ID93 + GLA-SEProtein/adjuvantIIa+
MTBVACLive-attenuated M. tuberculosisIIa++1000 CFU Erdman
RUTIWhole-cell fractionated M. tuberculosisIIa
Dar-901Whole-cell M. obuenseI
Ad5 Ag85AViral vectorI
Crucell Ad35 + MVA85AViral vectorI
ChAdOc1.85A + MVA85AViral vectorI
MVA85A-AerosolViral vectorI+
MVA85A-IMX313Viral vector/adjuvantI+
TB/FLU-04LViral vectorI
Vaccines previously in clinical trials
BCG-MVA85A boostViral vector boost++1000 CFU Erdman
Table 1

Translation use of the NHP model

Vaccines in clinical trialsTypePhaseNHP safety studyNHP efficacy studyChallenge
M. vaccae (SRL 172)Whole-cell M. vaccaeIII
M. indicus praniiWhole-cell M. indicus praniiIII
M72/AS01Protein/adjuvantIIb++500 CFU Erdman
Aeras-402Viral vectorIIb++275 CFU Erdman
VPM1002Live-recombinant BCGIIa+
H4:IC31Protein/adjuvantIIa
H1/H56:IC31Protein/adjuvantIIa++500/25 CFU Erdman
ID93 + GLA-SEProtein/adjuvantIIa+
MTBVACLive-attenuated M. tuberculosisIIa++1000 CFU Erdman
RUTIWhole-cell fractionated M. tuberculosisIIa
Dar-901Whole-cell M. obuenseI
Ad5 Ag85AViral vectorI
Crucell Ad35 + MVA85AViral vectorI
ChAdOc1.85A + MVA85AViral vectorI
MVA85A-AerosolViral vectorI+
MVA85A-IMX313Viral vector/adjuvantI+
TB/FLU-04LViral vectorI
Vaccines previously in clinical trials
BCG-MVA85A boostViral vector boost++1000 CFU Erdman
Vaccines in clinical trialsTypePhaseNHP safety studyNHP efficacy studyChallenge
M. vaccae (SRL 172)Whole-cell M. vaccaeIII
M. indicus praniiWhole-cell M. indicus praniiIII
M72/AS01Protein/adjuvantIIb++500 CFU Erdman
Aeras-402Viral vectorIIb++275 CFU Erdman
VPM1002Live-recombinant BCGIIa+
H4:IC31Protein/adjuvantIIa
H1/H56:IC31Protein/adjuvantIIa++500/25 CFU Erdman
ID93 + GLA-SEProtein/adjuvantIIa+
MTBVACLive-attenuated M. tuberculosisIIa++1000 CFU Erdman
RUTIWhole-cell fractionated M. tuberculosisIIa
Dar-901Whole-cell M. obuenseI
Ad5 Ag85AViral vectorI
Crucell Ad35 + MVA85AViral vectorI
ChAdOc1.85A + MVA85AViral vectorI
MVA85A-AerosolViral vectorI+
MVA85A-IMX313Viral vector/adjuvantI+
TB/FLU-04LViral vectorI
Vaccines previously in clinical trials
BCG-MVA85A boostViral vector boost++1000 CFU Erdman

While the NHP model of tuberculosis has served a positive predictor and provides clear justification for progression into clinical trials, the model has also served as a checkpoint, limiting unjustified vaccine candidates from progression. Thus, Aeras-402 could not protect rhesus macaques from a lethal 200-cfu Mtb Erdman challenge (Darrah et al. 2014) despite the generation of robust and sustained T cell responses in the lung (Darrah et al. 2014; Hokey et al. 2014). Limiting progression from preclinical status into clinical trials can save significant financial and material resources to be directed to more promising vaccine candidates. MVA85A, a virus vector vaccine, is a vaccine candidate that progressed into Phase I/II clinical trials in the absence of NHP efficacy data. The lack of efficacy of MVA85A in clinical trials (Sharpe et al. 2010; Verreck et al. 2009) has highlighted the value in using animal models to appropriately model clinical trials, and based on the data currently available, NHPs are the most appropriate such system. Such studies in the past could have resulted in a better use of clinical trial resources. While macaque experiments are themselves expensive, these have the potential to result in significant downstream fiscal savings. The authors would like to suggest that efficacy must be demonstrated in the NHP model of tuberculosis before vaccine candidates are approved for clinical trials.

Conclusions

The NHP model of tuberculosis has evolved into an accurate preclinical model of Mtb infection. Similarities in genetics and immune responses allow the prognosis of disease to parallel human infection. There remain subtle differences between the differing species of NHPs, strains of Mtb, and routes of infection/vaccination that all serve a purpose to allow the model to be optimized for testing the hypothesis of the study. While resources, the need for specialty facilities, and financial costs often impede on the widespread use of the NHP model, the accuracy yet plasticity of the model justifies the necessary use for many aspects of tuberculosis research. The NHP model further serves to perform preclinical tests on the advent and repurposing of novel technologies. These technologies can and have already been used to increase our clinical analyses of NHP studies, and may even shorten the time needed to demonstrate efficacy in vaccine or drug trials. Utilization of the NHP model as a checkpoint prior to clinical implementation of vaccines and therapeutics may also serve to save significant financial resources and time invested necessary for clinical trials.

Acknowledgments

The authors would like to acknowledge funding support from PHS (OD011104, RR026006, GM110760, AI089323, AI111943, AI127160, AI127222 and AI058609). This article is dedicated to the memory of Andrew A. Lackner, DVM, PhD, DACVP, long-serving Director of the TNPRC, who passed away on April 2, 2017, after revisions to this article were complete.

References

Apetrei
C
,
Pandrea
I
,
Mellors
JW
.
2012
.
Nonhuman primate models for HIV cure research
.
PLoS Pathog
8
(
8
):
e1002892
.

Arbues
A
,
Aguilo
JI
,
Gonzalo-Asensio
J
,
Marinova
D
,
Uranga
S
,
Puentes
E
,
Fernandez
C
,
Parra
A
,
Cardona
PJ
,
Vilaplana
C
,
Ausina
V
,
Williams
A
,
Clark
S
,
Malaga
W
,
Guilhot
C
,
Gicquel
B
,
Martin
C
.
2013
.
Construction, characterization and preclinical evaluation of MTBVAC, the first live-attenuated M. tuberculosis-based vaccine to enter clinical trials
.
Vaccine
31
(
42
):
4867
4873
.

Barclay
WR
,
Anacker
RL
,
Brehmer
W
,
Leif
W
,
Ribi
E
.
1970
.
Aerosol-induced tuberculosis in subhuman primates and the course of the disease after intravenous BCG vaccination
.
Infect Immun
2
(
5
):
574
582
.

Barclay
WR
,
Busey
WM
,
Dalgard
DW
,
Good
RC
,
Janicki
BW
,
Kasik
JE
,
Ribi
E
,
Ulrich
CE
,
Wolinsky
E
.
1973
.
Protection of monkeys against airborne tuberculosis by aerosol vaccination with bacillus Calmette-Guerin
.
Am Rev Respir Dis
107
(
3
):
351
358
.

Berg
RD
,
Levitte
S
,
O'Sullivan
MP
,
O'Leary
SM
,
Cambier
CJ
,
Cameron
J
,
Takaki
KK
,
Moens
CB
,
Tobin
DM
,
Keane
J
,
Ramakrishnan
L
.
2016
.
Lysosomal disorders drive susceptibility to tuberculosis by compromising macrophage migration
.
Cell
165
(
1
):
139
152
.

Bertholet
S
,
Ireton
GC
,
Ordway
DJ
,
Windish
HP
,
Pine
SO
,
Kahn
M
,
Phan
T
,
Orme
IM
,
Vedvick
TS
,
Baldwin
SL
,
Coler
RN
,
Reed
SG
.
2010
.
A defined tuberculosis vaccine candidate boosts BCG and protects against multidrug-resistant Mycobacterium tuberculosis
.
Sci Transl Med
2
(
53
):
53ra74
.

Billeskov
R
,
Tan
EV
,
Cang
M
,
Abalos
RM
,
Burgos
J
,
Pedersen
BV
,
Christensen
D
,
Agger
EM
,
Andersen
P
.
2016
.
Testing the H56 vaccine delivered in 4 different adjuvants as a BCG-booster in a non-human primate model of tuberculosis
.
PLoS One
11
(
8
):
e0161217
.

Capuano
SV
3rd,
Croix
DA
,
Pawar
S
,
Zinovik
A
,
Myers
A
,
Lin
PL
,
Bissel
S
,
Fuhrman
C
,
Klein
E
,
Flynn
JL
.
2003
.
Experimental Mycobacterium tuberculosis infection of cynomolgus macaques closely resembles the various manifestations of human M. tuberculosis infection
.
Infect Immun
71
(
10
):
5831
5844
.

Clarke
GL
.
1968
.
The relationship of hypersensitivity to shedding of Mycobacterium tuberculosis in experimentally infected Macaca mulatta
.
Am Rev Respir Dis
98
(
3
):
416
423
.

Coleman
MT
,
Chen
RY
,
Lee
M
,
Lin
PL
,
Dodd
LE
,
Maiello
P
,
Via
LE
,
Kim
Y
,
Marriner
G
,
Dartois
V
,
Scanga
C
,
Janssen
C
,
Wang
J
,
Klein
E
,
Cho
SN
,
Barry
CE
,
Flynn
JL
.
2014
a.
PET/CT imaging reveals a therapeutic response to oxazolidinones in macaques and humans with tuberculosis
.
Sci Transl Med
6
(
265
):
265ra167
.

Coleman
MT
,
Maiello
P
,
Tomko
J
,
Frye
LJ
,
Fillmore
D
,
Janssen
C
,
Klein
E
,
Lin
PL
.
2014
b.
Early changes by (18)fluorodeoxyglucose positron emission tomography coregistered with computed tomography predict outcome after Mycobacterium tuberculosis infection in cynomolgus macaques
.
Infect Immun
82
(
6
):
2400
2404
.

Coler
RN
,
Bertholet
S
,
Pine
SO
,
Orr
MT
,
Reese
V
,
Windish
HP
,
Davis
C
,
Kahn
M
,
Baldwin
SL
,
Reed
SG
.
2013
.
Therapeutic immunization against Mycobacterium tuberculosis is an effective adjunct to antibiotic treatment
.
J Infect Dis
207
(
8
):
1242
1252
.

Commandeur
S
,
van Meijgaarden
KE
,
Prins
C
,
Pichugin
AV
,
Dijkman
K
,
van den Eeden
SJ
,
Friggen
AH
,
Franken
KL
,
Dolganov
G
,
Kramnik
I
,
Schoolnik
GK
,
Oftung
F
,
Korsvold
GE
,
Geluk
A
,
Ottenhoff
TH
.
2013
.
An unbiased genome-wide Mycobacterium tuberculosis gene expression approach to discover antigens targeted by human T cells expressed during pulmonary infection
.
J Immunol
190
(
4
):
1659
1671
.

Darrah
PA
,
Bolton
DL
,
Lackner
AA
,
Kaushal
D
,
Aye
PP
,
Mehra
S
,
Blanchard
JL
,
Didier
PJ
,
Roy
CJ
,
Rao
SS
,
Hokey
DA
,
Scanga
CA
,
Sizemore
DR
,
Sadoff
JC
,
Roederer
M
,
Seder
RA
.
2014
.
Aerosol vaccination with AERAS-402 elicits robust cellular immune responses in the lungs of rhesus macaques but fails to protect against high-dose mycobacterium tuberculosis challenge
.
J Immunol
193
:
1799
1811
.

Diedrich
CR
,
Mattila
JT
,
Klein
E
,
Janssen
C
,
Phuah
J
,
Sturgeon
TJ
,
Montelaro
RC
,
Lin
PL
,
Flynn
JL
.
2010
.
Reactivation of latent tuberculosis in cynomolgus macaques infected with SIV is associated with early peripheral T cell depletion and not virus load
.
PLoS One
5
(
3
):
e9611
.

Dutta
NK
,
Mehra
S
,
Didier
PJ
,
Roy
CJ
,
Doyle
LA
,
Alvarez
X
,
Ratterree
M
,
Be
NA
,
Lamichhane
G
,
Jain
SK
,
Lacey
MR
,
Lackner
AA
,
Kaushal
D
.
2010
.
Genetic requirements for the survival of tubercle bacilli in primates
.
J Infect Dis
201
(
11
):
1743
1752
.

Ernst
JD
,
Hanekom
W
,
Hawn
T
,
Kampmann
B
,
Rengarajan
J
2012
.
Meeting report: The International Conference on Human Immunity to Tuberculosis
.
Tuberculosis (Edinb)
92
(
5
):
440
444
.

Flynn
JL
,
Gideon
HP
,
Mattila
JT
,
Lin
PL
.
2015
.
Immunology studies in non-human primate models of tuberculosis
.
Immunol Rev
264
(
1
):
60
73
.

Foreman
TW
,
Mehra
S
,
LoBato
DN
,
Malek
A
,
Alvarez
X
,
Golden
NA
,
Bucsan
AN
,
Didier
PJ
,
Doyle-Meyers
LA
,
Russell-Lodrigue
KE
,
Roy
CJ
,
Blanchard
J
,
Kuroda
MJ
,
Lackner
AA
,
Chan
J
,
Khader
SA
,
Jacobs
WR
Jr
,
Kaushal
D
.
2016
.
CD4+ T-cell-independent mechanisms suppress reactivation of latent tuberculosis in a macaque model of HIV coinfection
.
Proc Natl Acad Sci USA
113
(
38
):
E5636
5644
.

Gautam
US
,
McGillivray
A
,
Mehra
S
,
Didier
PJ
,
Midkiff
CC
,
Kissee
RS
,
Golden
NA
,
Alvarez
X
,
Niu
T
,
Rengarajan
J
,
Sherman
DR
,
Kaushal
D
.
2014
.
DosS is required for the complete virulence of mycobacterium tuberculosis in mice with classical granulomatous lesions
.
Am J Respir Cell Mol Biol
52
:
708
716
.

Gideon
HP
,
Skinner
JA
,
Baldwin
N
,
Flynn
JL
,
Lin
PL
.
2016
.
Early whole blood transcriptional signatures are associated with severity of lung inflammation in cynomolgus macaques with Mycobacterium tuberculosis infection
.
J Immunol
197
(
12
):
4817
4828
.

Good
RC
.
1968
.
Biology of the mycobacterioses. Simian tuberculosis: immunologic aspects
.
Ann NY Acad Sci
154
(
1
):
200
213
.

Gopal
R
,
Monin
L
,
Torres
D
,
Slight
S
,
Mehra
S
,
McKenna
K
,
Fallert Junecko
BA
,
Reinhart
TA
,
Kolls
J
,
Báez-Saldaña
R
,
Cruz-Lagunas
A
,
Rodríguez-Reyna
TS
,
Kumar
NP
,
Tessier
P
,
Roth
J
,
Selman
M
,
Becerril-Villanueva
E
,
Baquera-Heredia
J
,
Cumming
B
,
Kasprowicz
VO
,
Steyn
AJ
,
Babu
S
,
Kaushal
D
,
Zúñiga
J
,
Vogl
T
,
Rangel-Moreno
J
,
Khader
SA
.
2013
.
S100A8/A9 proteins mediate neutrophilic inflammation and lung pathology during tuberculosis
.
Am J Respir Crit Care Med
188
:
1137
1146
.

Gormus
BJ
,
Blanchard
JL
,
Alvarez
XH
,
Didier
PJ
.
2004
.
Evidence for a rhesus monkey model of asymptomatic tuberculosis
.
J Med Primatol
33
(
3
):
134
145
.

Haase
AT
.
2011
.
Early events in sexual transmission of HIV and SIV and opportunities for interventions
.
Annu Rev Med
62
:
127
139
.

Hofmeyr
A
,
Lau
WF
,
Slavin
MA
.
2007
.
Mycobacterium tuberculosis infection in patients with cancer, the role of 18-fluorodeoxyglucose positron emission tomography for diagnosis and monitoring treatment response
.
Tuberculosis (Edinb)
87
(
5
):
459
463
.

Hokey
DA
,
Wachholder
R
,
Darrah
PA
,
Bolton
DL
,
Barouch
DH
,
Hill
K
,
Dheenadhayalan
V
,
Schwander
S
,
Godin
CS
,
Douoguih
M
,
Pau
MG
,
Seder
RA
,
Roederer
M
,
Sadoff
JC
,
Sizemore
D
.
2014
.
A nonhuman primate toxicology and immunogenicity study evaluating aerosol delivery of AERAS-402/Ad35 vaccine: Evidence for transient t cell responses in peripheral blood and robust sustained responses in the lungs
.
Hum Vaccin Immunother
10
(
8
):
2199
2210
.

Hudock
TA
,
Foreman
TW
,
Bandyopadhyay
N
,
Gautam
US
,
Veatch
AV
,
LoBato
DN
,
Gentry
KM
,
Golden
NA
,
Cavigli
A
,
Mueller
M
,
Hwang
SA
,
Hunter
RL
,
Alvarez
X
,
Lackner
AA
,
Bader
JS
,
Mehra
S
,
Kaushal
D
.
2017
.
Hypoxia sensing and persistence genes are expressed during the intra-granulomatous survival of M. tuberculosis
.
Am J Respir Cell Mol Biol
56
(
5
):
637
647
.

Jain
SK
2017
.
The promise of molecular imaging in the study and treatment of infectious diseases
.
Mol Imaging Biol
19
(
3
):
341
347
.

Janicki
BW
,
Good
RC
,
Minden
P
,
Affronti
LF
,
Hymes
WF
.
1973
.
Immune responses in rhesus monkeys after bacillus Calmette-Guerin vaccination and aerosol challenge with Mycobacterium tuberculosis
.
Am Rev Respir Dis
107
(
3
):
359
366
.

Kaushal
D
,
Foreman
TW
,
Gautam
US
,
Alvarez
X
,
Adekambi
T
,
Rangel-Moreno
J
,
Golden
NA
,
Johnson
AM
,
Phillips
BL
,
Ahsan
MH
,
Russell-Lodrigue
KE
,
Doyle
LA
,
Roy
CJ
,
Didier
PJ
,
Blanchard
JL
,
Rengarajan
J
,
Lackner
AA
,
Khader
SA
,
Mehra
S
.
2015
.
Mucosal vaccination with attenuated Mycobacterium tuberculosis induces strong central memory responses and protects against tuberculosis
.
Nat Commun
6
:
8533
.

Kaushal
D
,
Mehra
S
.
2012
.
Faithful experimental models of human infection
.
Mycobact Dis
2
:
e108
.

Kaushal
D
,
Mehra
S
,
Didier
PJ
,
Lackner
AA
.
2012
.
The non-human primate model of tuberculosis
.
J Med Primatol
41
(
3
):
191
201
.

Kaushal
D
,
Schroeder
BG
,
Tyagi
S
,
Yoshimatsu
T
,
Scott
C
,
Ko
C
,
Carpenter
L
,
Mehrotra
J
,
Manabe
YC
,
Fleischmann
RD
,
Bishai
WR
.
2002
.
Reduced immunopathology and mortality despite tissue persistence in a Mycobacterium tuberculosis mutant lacking alternative sigma factor, SigH
.
Proc Natl Acad Sci USA
99
(
12
):
8330
8335
.

Kramnik
I
,
Beamer
G
.
2016
.
Mouse models of human TB pathology: Roles in the analysis of necrosis and the development of host-directed therapies
.
Semin Immunopathol
38
(
2
):
221
237
.

Langermans
JA
,
Andersen
P
,
van Soolingen
D
,
Vervenne
RA
,
Frost
PA
,
van der Laan
T
,
van Pinxteren
LA
,
van den Hombergh
J
,
Kroon
S
,
Peekel
I
,
Florquin
S
,
Thomas
AW
.
2001
.
Divergent effect of bacillus Calmette-Guerin (BCG) vaccination on Mycobacterium tuberculosis infection in highly related macaque species: implications for primate models in tuberculosis vaccine research
.
Proc Natl Acad Sci USA
98
(
20
):
11497
11502
.

Lin
PL
,
Coleman
T
,
Carney
JP
,
Lopresti
BJ
,
Tomko
J
,
Fillmore
D
,
Dartois
V
,
Scanga
C
,
Frye
LJ
,
Janssen
C
,
Klein
E
,
Barry
CE
III
,
Flynn
JL
.
2013
.
Radiologic responses in cynomolgous macaques for assessing tuberculosis chemotherapy regimens
.
Antimicrob Agents Chemother
57
:
4237
4244
.

Lin
PL
,
Dietrich
J
,
Tan
E
,
Abalos
RM
,
Burgos
J
,
Bigbee
C
,
Bigbee
M
,
Milk
L
,
Gideon
HP
,
Rodgers
M
,
Cochran
C
,
Guinn
KM
,
Sherman
DR
,
Klein
E
,
Janssen
C
,
Flynn
JL
,
Andersen
P
.
2012
.
The multistage vaccine H56 boosts the effects of BCG to protect cynomolgus macaques against active tuberculosis and reactivation of latent Mycobacterium tuberculosis infection
.
J Clin Invest
122
(
1
):
303
314
.

Lin
PL
,
Ford
CB
,
Coleman
MT
,
Myers
AJ
,
Gawande
R
,
Ioerger
T
,
Sacchettini
J
,
Fortune
SM
,
Flynn
JL
.
2014
.
Sterilization of granulomas is common in active and latent tuberculosis despite within-host variability in bacterial killing
.
Nat Med
20
(
1
):
75
79
.

Lin
PL
,
Pawar
S
,
Myers
A
,
Pegu
A
,
Fuhrman
C
,
Reinhart
TA
,
Capuano
SV
,
Klein
E
,
Flynn
JL
.
2006
.
Early events in Mycobacterium tuberculosis infection in cynomolgus macaques
.
Infect Immun
74
(
7
):
3790
3803
.

Lin
PL
,
Rodgers
M
,
Smith
L
,
Bigbee
M
,
Myers
A
,
Bigbee
C
,
Chiosea
I
,
Capuano
SV
,
Fuhrman
C
,
Klein
E
,
Flynn
JL
.
2009
.
Quantitative comparison of active and latent tuberculosis in the cynomolgus macaque model
.
Infect Immun
77
(
10
):
4631
4642
.

Liu
PT
,
Stenger
S
,
Li
H
,
Wenzel
L
,
Tan
BH
,
Krutzik
SR
,
Ochoa
MT
,
Schauber
J
,
Wu
K
,
Meinken
C
,
Kamen
DL
,
Wagner
M
,
Bals
R
,
Steinmeyer
A
,
Zügel
U
,
Gallo
RL
,
Eisenberg
D
,
Hewison
M
,
Hollis
BW
,
Adams
JS
,
Bloom
BR
,
Modlin
RL
.
2006
.
Toll-like receptor triggering of a vitamin D-mediated human antimicrobial response
.
Science
311
(
5768
):
1770
1773
.

Lonnroth
K
,
Williams
BG
,
Stadlin
S
,
Jaramillo
E
,
Dye
C
.
2008
.
Alcohol use as a risk factor for tuberculosis—a systematic review
.
BMC Public Health
8
:
289
.

Luabeya
AK
,
Kagina
BM
,
Tameris
MD
,
Geldenhuys
H
,
Hoff
ST
,
Shi
Z
,
Kromann
I
,
Hatherill
M
,
Mahomed
H
,
Hanekom
WA
,
Andersen
P
,
Scriba
TJ
; H56-032 Trial Study Group,
Schoeman
E
,
Krohn
C
,
Day
CL
,
Africa
H
,
Makhethe
L
,
Smit
E
,
Brown
Y
,
Suliman
S
,
Hughes
EJ
,
Bang
P
,
Snowden
MA
,
McClain
B
,
Hussey
GD
.
2015
.
First-in-human trial of the post-exposure tuberculosis vaccine H56:IC31 in Mycobacterium tuberculosis infected and non-infected healthy adults
.
Vaccine
33
(
33
):
4130
4140
.

Malhotra
V
,
Sharma
D
,
Ramanathan
VD
,
Shakila
H
,
Saini
DK
,
Chakravorty
S
,
Das
TK
,
Li
Q
,
Silver
RF
,
Narayanan
PR
,
Tyagi
JS
.
2004
.
Disruption of response regulator gene, devR, leads to attenuation in virulence of Mycobacterium tuberculosis
.
FEMS Microbiol Lett
231
(
2
):
237
245
.

Martin
C
,
Williams
A
,
Hernandez-Pando
R
,
Cardona
PJ
,
Gormley
E
,
Bordat
Y
,
Soto
CY
,
Clark
SO
,
Hatch
GJ
,
Aguilar
D
,
Ausina
V
,
Gicquel
B
.
2006
.
The live Mycobacterium tuberculosis phoP mutant strain is more attenuated than BCG and confers protective immunity against tuberculosis in mice and guinea pigs
.
Vaccine
24
(
17
):
3408
3419
.

Martinez
V
,
Castilla-Lievre
MA
,
Guillet-Caruba
C
,
Grenier
G
,
Fior
R
,
Desarnaud
S
,
Doucet-Populaire
F
,
Boue
F
.
2012
.
(18)F-FDG PET/CT in tuberculosis: An early non-invasive marker of therapeutic response
.
Int J Tuberc Lung Dis
16
(
9
):
1180
1185
.

Mattila
JT
,
Diedrich
CR
,
Lin
PL
,
Phuah
J
,
Flynn
JL
.
2011
.
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
186
(
6
):
3527
3537
.

McNicholl
JM
.
2016
.
Combining biomedical preventions for HIV: Vaccines with pre-exposure prophylaxis, microbicides or other HIV preventions
.
Hum Vaccin Immunother
12
(
12
):
3202
3211
.

Mehra
S
,
Alvarez
X
,
Didier
PJ
,
Doyle
LA
,
Blanchard
JL
,
Lackner
AA
,
Kaushal
D
.
2013
.
Granuloma correlates of protection against tuberculosis and mechanisms of immune modulation by Mycobacterium tuberculosis
.
J Infect Dis
207
(
7
):
1115
1127
.

Mehra
S
,
Foreman
TW
,
Didier
PJ
,
Ahsan
MH
,
Hudock
TA
,
Kissee
R
,
Golden
NA
,
Gautam
US
,
Johnson
AM
,
Alvarez
X
,
Russell-Lodrigue
KE
,
Doyle
LA
,
Roy
CJ
,
Niu
T
,
Blanchard
JL
,
Khader
SA
,
Lackner
AA
,
Sherman
DR
,
Kaushal
D
.
2015
.
The DosR regulon modulates adaptive immunity and is essential for M. tuberculosis persistence
.
Am J Respir Crit Care Med
191
:
1185
1196
.

Mehra
S
,
Golden
NA
,
Dutta
NK
,
Midkiff
CC
,
Alvarez
X
,
Doyle
LA
,
Asher
M
,
Russell-Lodrigue
K
,
Monjure
C
,
Roy
CJ
,
Blanchard
JL
,
Didier
PJ
,
Veazey
RS
,
Lackner
AA
,
Kaushal
D
.
2011
.
Reactivation of latent tuberculosis in rhesus macaques by coinfection with simian immunodeficiency virus
.
J Med Primatol
40
(
4
):
233
243
.

Mehra
S
,
Golden
NA
,
Stuckey
K
,
Didier
PJ
,
Doyle
LA
,
Russell-Lodrigue
KE
,
Sugimoto
C
,
Hasegawa
A
,
Sivasubramani
SK
,
Roy
CJ
,
Alvarez
X
,
Kuroda
MJ
,
Blanchard
JL
,
Lackner
AA
,
Kaushal
D
.
2012
.
The Mycobacterium tuberculosis stress response factor SigH is required for bacterial burden as well as immunopathology in primate lungs
.
J Infect Dis
205
(
8
):
1203
1213
.

Mothe
BR
,
Lindestam Arlehamn
CS
,
Dow
C
,
Dillon
MB
,
Wiseman
RW
,
Bohn
P
,
Karl
J
,
Golden
NA
,
Gilpin
T
,
Foreman
TW
,
Rodgers
MA
,
Mehra
S
,
Scriba
TJ
,
Flynn
JL
,
Kaushal
D
,
O'Connor
DH
,
Sette
A
.
2015
.
The TB-specific CD4(+) T cell immune repertoire in both cynomolgus and rhesus macaques largely overlap with humans
.
Tuberculosis (Edinb)
95
(
6
):
722
735
.

Munn
DH
,
Shafizadeh
E
,
Attwood
JT
,
Bondarev
I
,
Pashine
A
,
Mellor
AL
.
1999
.
Inhibition of T cell proliferation by macrophage tryptophan catabolism
.
J Exp Med
189
(
9
):
1363
1372
.

Nathan
C
.
2006
.
Role of iNOS in human host defense
.
Science
312
(
5782
):
1874
5
; author reply 1874–1875.

O'Leary
SM
,
Coleman
MM
,
Chew
WM
,
Morrow
C
,
McLaughlin
AM
,
Gleeson
LE
,
O'Sullivan
MP
,
Keane
J
.
2014
.
Cigarette smoking impairs human pulmonary immunity to Mycobacterium tuberculosis
.
Am J Respir Crit Care Med
190
(
12
):
1430
1436
.

Okada
M
,
Kita
Y
,
Nakajima
T
,
Kanamaru
N
,
Hashimoto
S
,
Nagasawa
T
,
Kaneda
Y
,
Yoshida
S
,
Nishida
Y
,
Fukamizu
R
,
Tsunai
Y
,
Inoue
R
,
Nakatani
H
,
Namie
Y
,
Yamada
J
,
Takao
K
,
Asai
R
,
Asaki
R
,
Matsumoto
M
,
McMurray
DN
,
Dela Cruz
EC
,
Tan
EV
,
Abalos
RM
,
Burgos
JA
,
Gelber
R
,
Sakatani
M
.
2007
.
Evaluation of a novel vaccine (HVJ-liposome/HSP65 DNA+IL-12 DNA) against tuberculosis using the cynomolgus monkey model of TB
.
Vaccine
25
(
16
):
2990
2993
.

Pena
JC
,
Ho
WZ
.
2016
.
Non-human primate models of tuberculosis
.
Microbiol Spectr
4
(
4
):
1
13
.

Perez
E
,
Samper
S
,
Bordas
Y
,
Guilhot
C
,
Gicquel
B
,
Martin
C
.
2001
.
An essential role for phoP in Mycobacterium tuberculosis virulence
.
Mol Microbiol
41
(
1
):
179
187
.

Phillips
BL
,
Mehra
S
,
Ahsan
MH
,
Selman
M
,
Khader
SA
,
Kaushal
D
.
2015
.
LAG3 expression in active Mycobacterium tuberculosis infections
.
Am J Pathol
185
(
3
):
820
833
.

Polverino
F
,
Doyle-Eisele
M
,
McDonald
J
,
Wilder
JA
,
Royer
C
,
Laucho-Contreras
M
,
Kelly
EM
,
Divo
M
,
Pinto-Plata
V
,
Mauderly
J
,
Celli
BR
,
Tesfaigzi
Y
,
Owen
CA
.
2015
.
A novel nonhuman primate model of cigarette smoke-induced airway disease
.
Am J Pathol
185
(
3
):
741
755
.

Reed
SG
,
Coler
RN
,
Dalemans
W
,
Tan
EV
,
DeLa Cruz
EC
,
Basaraba
RJ
,
Orme
IM
,
Skeiky
YA
,
Alderson
MR
,
Cowgill
KD
,
Prieels
JP
,
Abalos
RM
,
Dubois
MC
,
Cohen
J
,
Mettens
P
,
Lobet
Y
.
2009
.
Defined tuberculosis vaccine, Mtb72F/AS02A, evidence of protection in cynomolgus monkeys
.
Proc Natl Acad Sci USA
106
(
7
):
2301
2306
.

Rustad
TR
,
Harrell
MI
,
Liao
R
,
Sherman
DR
.
2008
.
The enduring hypoxic response of Mycobacterium tuberculosis
.
PLoS One
3
(
1
):
e1502
.

Sabin
AB
1957
.
Present status of attenuated live virus poliomyelitis vaccine
.
Bull NY Acad Med
33
(
1
):
17
39
.

Scanga
CA
,
Lopresti
BJ
,
Tomko
J
,
Frye
LJ
,
Coleman
TM
,
Fillmore
D
,
Carney
JP
,
Lin
PL
,
Flynn
JL
,
Gardner
CL
,
Sun
C
,
Klimstra
WB
,
Ryman
KD
,
Reed
DS
,
Fisher
DJ
,
Cole
KS
.
2014
.
In vivo imaging in an ABSL-3 regional biocontainment laboratory
.
Pathog Dis
71
(
2
):
205
210
.

Schmidt
LH
.
1966
.
Studies on the antituberculous activity of ethambutol in monkeys
.
Ann NY Acad Sci
135
(
2
):
747
758
.

Sharpe
S
,
White
A
,
Sarfas
C
,
Sibley
L
,
Gleeson
F
,
McIntyre
A
,
Basaraba
R
,
Clark
S
,
Hall
G
,
Rayner
E
,
Williams
A
,
Marsh
PD
,
Dennis
M
.
2016
.
Alternative BCG delivery strategies improve protection against Mycobacterium tuberculosis in non-human primates: Protection associated with mycobacterial antigen-specific CD4 effector memory T-cell populations
.
Tuberculosis (Edinb)
101
:
174
190
.

Sharpe
SA
,
Eschelbach
E
,
Basaraba
RJ
,
Gleeson
F
,
Hall
GA
,
McIntyre
A
,
Williams
A
,
Kraft
SL
,
Clark
S
,
Gooch
K
,
Hatch
G
,
Orme
IM
,
Marsh
PD
,
Dennis
MJ
.
2009
.
Determination of lesion volume by MRI and stereology in a macaque model of tuberculosis
.
Tuberculosis (Edinb)
89
(
6
):
405
416
.

Sharpe
SA
,
McShane
H
,
Dennis
MJ
,
Basaraba
RJ
,
Gleeson
F
,
Hall
G
,
McIntyre
A
,
Gooch
K
,
Clark
S
,
Beveridge
NE
,
Nuth
E
,
White
A
,
Marriott
A
,
Dowall
S
,
Hill
AV
,
Williams
A
,
Marsh
PD
.
2010
.
Establishment of an aerosol challenge model of tuberculosis in rhesus macaques and an evaluation of endpoints for vaccine testing
.
Clin Vaccine Immunol
17
(
8
):
1170
1182
.

Shedlock
DJ
,
Silvestri
G
,
Weiner
DB
.
2009
.
Monkeying around with HIV vaccines: Using rhesus macaques to define ‘gatekeepers’ for clinical trials
.
Nat Rev Immunol
9
(
10
):
717
728
.

Shen
Y
,
Shen
L
,
Sehgal
P
,
Zhou
D
,
Simon
M
,
Miller
M
,
Enimi
EA
,
Henckler
B
,
Chalifoux
L
,
Sehgal
N
,
Gastron
M
,
Letvin
NL
,
Chen
ZW
.
2001
.
Antiretroviral agents restore Mycobacterium-specific T-cell immune responses and facilitate controlling a fatal tuberculosis-like disease in Macaques coinfected with simian immunodeficiency virus and Mycobacterium bovis BCG
.
J Virol
75
(
18
):
8690
8696
.

Singhal
A
,
Jie
L
,
Kumar
P
,
Hong
GS
,
Leow
MK
,
Paleja
B
,
Tsenova
L
,
Kurepina
N
,
Chen
J
,
Zolezzi
F
,
Kreiswirth
B
,
Poidinger
M
,
Chee
C
,
Kaplan
G
,
Wang
YT
,
De Libero
G
.
2014
.
Metformin as adjunct antituberculosis therapy
.
Sci Transl Med
6
(
263
):
263ra159
.

Slight
SR
,
Rangel-Moreno
J
,
Gopal
R
,
Lin
Y
,
Fallert Junecko
BA
,
Mehra
S
,
Selman
M
,
Becerril-Villanueva
E
,
Baquera-Heredia
J
,
Pavon
L
,
Kaushal
D
,
Reinhart
TA
,
Randall
TD
,
Khader
SA
.
2013
.
CXCR5(+) T helper cells mediate protective immunity against tuberculosis
.
J Clin Invest
123
(
2
):
712
726
.

Spertini
F
,
Audran
R
,
Chakour
R
,
Karoui
O
,
Steiner-Monard
V
,
Thierry
AC
,
Mayor
CE
,
Rettby
N
,
Jaton
K
,
Vallotton
L
,
Lazor-Blanchet
C
,
Doce
J
,
Puentes
E
,
Marinova
D
,
Aguilo
N
,
Martin
C
.
2015
.
Safety of human immunisation with a live-attenuated Mycobacterium tuberculosis vaccine: A randomised, double-blind, controlled phase I trial
.
Lancet Respir Med
3
(
12
):
953
962
.

Su
J
,
Chen
X
,
Huang
Y
,
Li
W
,
Li
J
,
Cao
K
,
Cao
G
,
Zhang
L
,
Li
F
,
Roberts
AI
,
Kang
H
,
Yu
P
,
Ren
G
,
Ji
W
,
Wang
Y
,
Shi
Y
.
2014
.
Phylogenetic distinction of iNOS and IDO function in mesenchymal stem cell-mediated immunosuppression in mammalian species
.
Cell Death Differ
21
(
3
):
388
396
.

Thoma-Uszynski
S
,
Stenger
S
,
Takeuchi
O
,
Ochoa
MT
,
Engele
M
,
Sieling
PA
,
Barnes
PF
,
Rollinghoff
M
,
Bolcskei
PL
,
Wagner
M
,
Akira
S
,
Norgard
MV
,
Belisle
JT
,
Godowski
PJ
,
Bloom
BR
,
Modlin
RL
.
2001
.
Induction of direct antimicrobial activity through mammalian toll-like receptors
.
Science
291
(
5508
):
1544
1547
.

Veazey
RS
,
Chatterji
U
,
Bobardt
M
,
Russell-Lodrigue
KE
,
Li
J
,
Wang
X
,
Gallay
PA
.
2015
.
C5A protects macaques from vaginal simian-human immunodeficiency virus challenge
.
Antimicrob Agents Chemother
60
(
1
):
693
698
.

Verreck
FA
,
Vervenne
RA
,
Kondova
I
,
van Kralingen
KW
,
Remarque
EJ
,
Braskamp
G
,
van der Werff
NM
,
Kersbergen
A
,
Ottenhoff
TH
,
Heidt
PJ
,
Gilbert
SC
,
Gicquel
B
,
Hill
AV
,
Martin
C
,
McShane
H
,
Thomas
AW
.
2009
.
MVA.85A boosting of BCG and an attenuated, phoP deficient M. tuberculosis vaccine both show protective efficacy against tuberculosis in rhesus macaques
.
PLoS One
4
(
4
):
e5264
.

Via
LE
,
Weiner
DM
,
Schimel
D
,
Lin
PL
,
Dayao
E
,
Tankersley
SL
,
Cai
Y
,
Coleman
MT
,
Tomko
J
,
Paripati
P
,
Orandle
M
,
Kastenmayer
RJ
,
Tartakovsky
M
,
Rosenthal
A
,
Portevin
D
,
Eum
SY
,
Lahouar
S
,
Gagneux
S
,
Young
DB
,
Flynn
JL
,
Barry
CE
III.
2013
.
Differential virulence and disease progression following Mycobacterium tuberculosis complex infection of the common marmoset (Callithrix jacchus)
.
Infect Immun
81
(
8
):
2909
2919
.

Walsh
GP
,
Tan
EV
,
dela Cruz
EC
,
Abalos
RM
,
Villahermosa
LG
,
Young
LJ
,
Cellona
RV
,
Nazareno
JB
,
Horwitz
MA
.
1996
.
The Philippine cynomolgus monkey (Macaca fasicularis) provides a new nonhuman primate model of tuberculosis that resembles human disease
.
Nat Med
2
(
4
):
430
436
.

Walter
ND
,
de Jong
BC
,
Garcia
BJ
,
Dolganov
GM
,
Worodria
W
,
Byanyima
P
,
Musisi
E
,
Huang
L
,
Chan
ED
,
Van
TT
,
Antonio
M
,
Ayorinde
A
,
Kato-Maeda
M
,
Nahid
P
,
Leung
AM
,
Yen
A
,
Fingerlin
TE
,
Kechris
K
,
Strong
M
,
Voskuil
MI
,
Davis
JL
,
Schoolnik
GK
.
2016
.
Adaptation of Mycobacterium tuberculosis to impaired host immunity in HIV-infected patients
.
J Infect Dis
214
(
8
):
1205
1211
.

Weinstein
EA
,
Ordonez
AA
,
DeMarco
VP
,
Murawski
AM
,
Pokkali
S
,
MacDonald
EM
,
Klunk
M
,
Mease
RC
,
Pomper
MG
,
Jain
SK
.
2014
.
Imaging Enterobacteriaceae infection in vivo with 18F-fluorodeoxysorbitol positron emission tomography
.
Sci Transl Med
6
(
259
):
259ra146
.

White
AD
,
Sarfas
C
,
West
K
,
Sibley
LS
,
Wareham
AS
,
Clark
S
,
Dennis
MJ
,
Williams
A
,
Marsh
PD
,
Sharpe
SA
.
2015
.
Evaluation of the immunogenicity of Mycobacterium bovis BCG delivered by aerosol to the lungs of macaques
.
Clin Vaccine Immunol
22
(
9
):
992
1003
.

Zhang
YJ
,
Reddy
MC
,
Ioerger
TR
,
Rothchild
AC
,
Dartois
V
,
Schuster
BM
,
Trauner
A
,
Wallis
D
,
Galaviz
S
,
Huttenhower
C
,
Sacchettini
JC
,
Behar
SM
,
Rubin
EJ
.
2013
.
Tryptophan biosynthesis protects mycobacteria from CD4 T-cell-mediated killing
.
Cell
155
(
6
):
1296
1308
.

Zhang
YJ
,
Rubin
EJ
.
2013
.
Feast or famine: The host-pathogen battle over amino acids
.
Cell Microbiol
15
(
7
):
1079
1087
.