4
Please cite this article in press as: Evans TG, et al. Status of vaccine research and development of vaccines for tuberculosis. Vaccine (2016), http://dx.doi.org/10.1016/j.vaccine.2016.02.079 ARTICLE IN PRESS G Model JVAC-17430; No. of Pages 4 Vaccine xxx (2016) xxx–xxx Contents lists available at ScienceDirect Vaccine j our na l ho me page: www.elsevier.com/locate/vaccine Status of vaccine research and development of vaccines for tuberculosis Thomas G. Evans a,, Lew Schrager a , Jelle Thole b a Aeras, 1405 Research Blvd, Rockville, MD 20850, United States b Tuberculosis Vaccine Initiative, Lelystad, Netherlands a r t i c l e i n f o Article history: Received 17 August 2015 Accepted 10 February 2016 Available online xxx Keywords: Vaccine Tuberculosis Latency BCG a b s t r a c t TB is now the single pathogen that causes the greatest mortality in the world, at over 1.6 million deaths each year. The widely used the 90 year old BCG vaccine appears to have minimal impact on the world- wide incidence despite some efficacy in infants. Novel vaccine development has accelerated in the past 15 years, with 15 candidates entering human trials; two vaccines are now in large-scale efficacy stud- ies. Modeling by three groups has consistently shown that mass vaccination that includes activity in the latently infected population, especially adolescents and young adults, will likely have the largest impact on new disease transmission. At present the field requires better validated animal models, better understanding of a correlate of immunity, new cost-effective approaches to Proof of Concept trials, and increased appreciation by the public health and scientific community for the size of the problem and the need for a vaccine. Such a vaccine is likely to also play a role in the era of increasing antibiotic resistance. Ongoing efforts and studies are working to implement these needs over the next 5 years, which will lead to an understanding that will increase the likelihood of a successful TB vaccine. © 2016 The Authors. Published by Elsevier Ltd. This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/). 1. The disease and pathogen In March 1993, the World Health Organization (WHO) des- ignated tuberculosis (TB), a disease caused by Mycobacterium tuberculosis (Mtb), a global public health emergency. Since 1993 there are more than 1.5 million deaths from TB each year, making it the number nine overall cause of mortality worldwide. Nearly 1 billion people have died of TB over the past few centuries, an astounding number, and TB is a leading cause of mortality in HIV- infected individuals and in women of childbearing age. Ninety-nine per cent of the TB deaths and 95% of the over 9.5 million new cases each year occur in the low and middle-income countries that comprise 85% of the world’s population. One-third of the world’s population is estimated to be latently infected and 5–10% of these people may go on to develop active TB disease; however, the risk This is an Open Access article published under the CC BY 3.0 IGO license which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. In any use of this article, there should be no suggestion that WHO endorses any specific organisation, products or services. The use of the WHO logo is not permitted. This notice should be preserved along with the article’s original URL. Corresponding author. Tel.: +1 3015472934. E-mail addresses: [email protected] (T.G. Evans), [email protected] (L. Schrager), [email protected] (J. Thole). is considerably higher in the presence of predisposing factors. As such, in part the epidemic of TB in sub-Saharan Africa has been fueled by HIV disease, and the increasing incidence of diabetes in Asia further threatens attempts at control. One of the highest priorities of TB research is to develop vaccines that are more efficacious for preventing TB than Mycobacterium bovis bacillus Calmette–Guérin (BCG), the only vaccine available to (partially) protect against TB disease [1]. Research and devel- opment is one of the three pillars of the WHO TB strategy, and will play a crucial role in accelerating the reductions in TB inci- dence and mortality required to reach global TB targets to reduce TB deaths by 95% and to reduce new infections by 90% between 2015 and 2035. Better control of TB than that provided by BCG could be achieved by vaccines that protect individuals from initial infection with M. tuberculosis (Mtb), prevent those infected from progressing to active disease, or decrease the capacity for trans- mission by those with active disease. Different vaccines may be required to induce immune responses in diverse populations, such as infants, young adults, those already latently infected with Mtb, and those co-infected with Mtb and HIV. Experts in TB prevention and control mostly agree that the largest vaccine impact would result from mass vaccination of all adolescents/young adults in high burden countries, regardless of their infection status, even with a vaccine that is only 50–60% efficacious over a 10 year duration. Such an initial vaccine strategy could prevent up to 50 million cases of http://dx.doi.org/10.1016/j.vaccine.2016.02.079 0264-410X/© 2016 The Authors. Published by Elsevier Ltd. This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).

2016 Status of Vaccine Research and Development of Vaccines for Tuberculosis. VACCINE

Embed Size (px)

DESCRIPTION

Estado vacunal contra la TBC

Citation preview

Page 1: 2016 Status of Vaccine Research and Development of Vaccines for Tuberculosis. VACCINE

J

Sf

Ta

b

a

ARAA

KVTLB

1

itti1aipccpp

ptsut

(

h0

ARTICLE IN PRESSG ModelVAC-17430; No. of Pages 4

Vaccine xxx (2016) xxx–xxx

Contents lists available at ScienceDirect

Vaccine

j our na l ho me page: www.elsev ier .com/ locate /vacc ine

tatus of vaccine research and development of vaccinesor tuberculosis�

homas G. Evansa,∗, Lew Schragera, Jelle Tholeb

Aeras, 1405 Research Blvd, Rockville, MD 20850, United StatesTuberculosis Vaccine Initiative, Lelystad, Netherlands

r t i c l e i n f o

rticle history:eceived 17 August 2015ccepted 10 February 2016vailable online xxx

eywords:accineuberculosis

a b s t r a c t

TB is now the single pathogen that causes the greatest mortality in the world, at over 1.6 million deathseach year. The widely used the 90 year old BCG vaccine appears to have minimal impact on the world-wide incidence despite some efficacy in infants. Novel vaccine development has accelerated in the past15 years, with 15 candidates entering human trials; two vaccines are now in large-scale efficacy stud-ies. Modeling by three groups has consistently shown that mass vaccination that includes activity inthe latently infected population, especially adolescents and young adults, will likely have the largestimpact on new disease transmission. At present the field requires better validated animal models, better

atencyCG

understanding of a correlate of immunity, new cost-effective approaches to Proof of Concept trials, andincreased appreciation by the public health and scientific community for the size of the problem and theneed for a vaccine. Such a vaccine is likely to also play a role in the era of increasing antibiotic resistance.Ongoing efforts and studies are working to implement these needs over the next 5 years, which will leadto an understanding that will increase the likelihood of a successful TB vaccine.

© 2016 The Authors. Published by Elsevier Ltd. This is an open access article under the CC BY license

. The disease and pathogen

In March 1993, the World Health Organization (WHO) des-gnated tuberculosis (TB), a disease caused by Mycobacteriumuberculosis (Mtb), a global public health emergency. Since 1993here are more than 1.5 million deaths from TB each year, makingt the number nine overall cause of mortality worldwide. Nearly

billion people have died of TB over the past few centuries, anstounding number, and TB is a leading cause of mortality in HIV-nfected individuals and in women of childbearing age. Ninety-nineer cent of the TB deaths and 95% of the over 9.5 million newases each year occur in the low and middle-income countries that

Please cite this article in press as: Evans TG, et al. Status of vaccine

(2016), http://dx.doi.org/10.1016/j.vaccine.2016.02.079

omprise 85% of the world’s population. One-third of the world’sopulation is estimated to be latently infected and 5–10% of theseeople may go on to develop active TB disease; however, the risk

� This is an Open Access article published under the CC BY 3.0 IGO license whichermits unrestricted use, distribution, and reproduction in any medium, providedhe original work is properly cited. In any use of this article, there should be nouggestion that WHO endorses any specific organisation, products or services. These of the WHO logo is not permitted. This notice should be preserved along withhe article’s original URL.∗ Corresponding author. Tel.: +1 3015472934.

E-mail addresses: [email protected] (T.G. Evans), [email protected]. Schrager), [email protected] (J. Thole).

ttp://dx.doi.org/10.1016/j.vaccine.2016.02.079264-410X/© 2016 The Authors. Published by Elsevier Ltd. This is an open access article u

(http://creativecommons.org/licenses/by/4.0/).

is considerably higher in the presence of predisposing factors. Assuch, in part the epidemic of TB in sub-Saharan Africa has beenfueled by HIV disease, and the increasing incidence of diabetes inAsia further threatens attempts at control.

One of the highest priorities of TB research is to develop vaccinesthat are more efficacious for preventing TB than Mycobacteriumbovis bacillus Calmette–Guérin (BCG), the only vaccine availableto (partially) protect against TB disease [1]. Research and devel-opment is one of the three pillars of the WHO TB strategy, andwill play a crucial role in accelerating the reductions in TB inci-dence and mortality required to reach global TB targets to reduceTB deaths by 95% and to reduce new infections by 90% between2015 and 2035. Better control of TB than that provided by BCGcould be achieved by vaccines that protect individuals from initialinfection with M. tuberculosis (Mtb), prevent those infected fromprogressing to active disease, or decrease the capacity for trans-mission by those with active disease. Different vaccines may berequired to induce immune responses in diverse populations, suchas infants, young adults, those already latently infected with Mtb,and those co-infected with Mtb and HIV. Experts in TB preventionand control mostly agree that the largest vaccine impact would

research and development of vaccines for tuberculosis. Vaccine

result from mass vaccination of all adolescents/young adults in highburden countries, regardless of their infection status, even with avaccine that is only 50–60% efficacious over a 10 year duration. Suchan initial vaccine strategy could prevent up to 50 million cases of

nder the CC BY license (http://creativecommons.org/licenses/by/4.0/).

Page 2: 2016 Status of Vaccine Research and Development of Vaccines for Tuberculosis. VACCINE

ING ModelJ

2 ccine

iaois(mara

2

2

itpohatpsd

tbtmcianpa

2f

thiaTaT

a((vhp1l4detan

ARTICLEVAC-17430; No. of Pages 4

T.G. Evans et al. / Va

ncident TB cases in high-burden settings during the first 35 yearsfter its introduction [2]. This impact would likely save millionsf lives and billions of dollars in treatment and control costs. Its also important to note that strains of Mtb that are resistant totandard anti-tuberculous drugs, including multiply drug resistantMDR) and extensively drug-resistant (XDR) strains are becoming

ore common. While Mtb is likely to continue to evolve resistancegainst drugs develop to combat it, it is highly unlikely that suchesistance would result in a concomitant development of resistancegainst otherwise effective TB vaccines.

. Overview of current efforts

.1. Vaccines currently available and their limitations

The human immune system can contain tuberculosis infectionn the majority of cases following infection, and a partially effec-ive vaccine for infants exists. These data, as well as evidence thatrior Mtb infection may protect against later disease [3], paint anptimistic picture for the development of a vaccine. On the otherand, because Mtb has co-evolved with humans over many yearsnd may use the human immune system to maintain its propaga-ion [4], and because prior active pulmonary tuberculosis does notrotect and may actually be a risk factor for reinfection and disease,ome scientific skepticism has arisen concerning the prospect foreveloping an effective vaccine.

BCG is a live, attenuated vaccine that is widely administeredo infants in most areas endemic for TB. BCG has been shown toe effective for the prevention of more serious extrapulmonaryuberculosis in young children, such as tuberculous meningitis and

iliary tuberculosis [5]. A meta-analysis of prospective trials andase-control studies concluded efficacy against pulmonary TB innfants and adolescents to be approximately 50%, with a range from

low of zero to a high of eighty per cent [6]. When delivered toewborns, however, BCG is not fully effective in preventing adultulmonary TB, which constitutes the bulk of the global morbiditynd mortality disease burden.

.2. General approaches to vaccine development for this diseaseor low and middle income country markets

Most successful, licensed vaccines available today induce neu-ralizing antibodies that provide protective immunity. Animal anduman studies of TB, however, suggest that a robust cellular

mmune response is required for protection against Mtb infectionnd disease [7,8]. For this reason, the majority of current clinicalB vaccine candidates are based on a variety of vectors, adjuvantsnd antigens that induce classical TH1 cytokines such as IFN-� orNF-� from either CD4+ or CD8+ T cells.

These clinical candidates are based on a variety of vaccinepproaches, such as inactivated whole cell or whole cell extractsMycobacterium indicus pranii, Mycobacterium vaccae, DAR-901Mycobacterium obuense), RUTI and Mycobacterium smegmatis),iral-vectored candidates (vaccinia based MVA85A, influenza, anduman adenovirus 5 and 35 and chimpanzee adenovirus), fusionrotein subunits with TH1-inducing adjuvants (M72/AS01, Hybrid/CAF01, Hybrid 1/IC31, H4/IC31, H56/IC31 and ID93/GLA-SE), and

ive recombinant BCG or attenuated TB vaccines (VPM 1002, Aeras22, rBCG30, and MTBVAC). DNA vaccines are being developed inifferent countries, notably emerging economies, but have not yet

Please cite this article in press as: Evans TG, et al. Status of vaccine

(2016), http://dx.doi.org/10.1016/j.vaccine.2016.02.079

ntered into human clinical trials. To date, clinical trials charac-erizing these candidate vaccines have included studies of safetynd immunogenicity in diverse populations, including healthy, TB-aïve adults and infants, latently infected adolescents and adults,

PRESSxxx (2016) xxx–xxx

HIV-infected adults, as well as patients undergoing drug treatmentfor TB.

An initial Phase IIB proof-of-concept (PoC) efficacy trial in 2797BCG-vaccinated infants boosted at 4–6 months of age with a viral-vectored vaccine containing one antigen (MVA85A) showed noefficacy against TB disease or infection [9]. Whether this disap-pointing outcome was due to the magnitude of the immunologicresponse, the single dose of Ag85A antigen studied, the populationvaccinated, or an incorrect immunologic hypothesis is not clear. Alarge, Phase IIB trial in 3600 HIV-uninfected, adults latently infectedwith Mtb is under way in three African countries (South Africa,Kenya, Zambia) using the GSK M72 adjuvanted fusion proteinvaccine. This study should further our understanding of poten-tial correlates of protection, as well as the role of TH-1 inducedimmunological responses in preventing the development of diseasein latently infected individuals. Data are expected in 2018.

In addition to these large-scale PoC trials, a new set of humanstudies are under way, based on the use of innovative trials designsintended to show the biologic activity of vaccine candidates usingmore focused populations specifically selected to reduce samplesize. The first of these new trial designs is testing whether a novelvaccine (H4/IC31) or the use of BCG re-vaccination can preventsustained infection by Mtb (as opposed to disease). The trial usesnovel blood tests in which BCG vaccination does not interfere withthe test result – a common obstacle with the longtime, standarddiagnostic, the tuberculin skin test. The study is enrolling adoles-cents in South Africa with a high rate of incident Mtb infection,thereby requiring only 330 subjects per arm rather than the twothousand or more needed in the classic PoC trials. The second inno-vative trial design is to study the ability of a vaccine to preventthe 4–6% relapse and/or reinfection rate typically observed follow-ing successful treatment of active TB. A “Prevention of Recurrence”trial using the ID93 candidate will likely begin shortly and requireapproximately 450 subjects per arm. All of these trials representan attempt to make decisions earlier in development (a “shift tothe left”), including the initial attempts to coordinate a more glob-ally focused preclinical consortium effort, given the 20–50 milliondollar cost needed to conduct classical efficacy disease endpointstudies. Of note, a 10,000-person Phase 3 study is also underwayin Guanxi province, China, to test the efficacy of a killed M. vaccaelysate vaccine to prevent TB in adults with latent disease.

3. Technical and regulatory assessment

At present there are no accepted correlates of protection that,unto themselves, could support a decision to license a TB vaccine.It is presumed that prevention of actual disease will continue to bethe primary endpoint of Phase 2b or 3 trials. However, whether reg-ulators will accept stringent definitions based on clinical outcomesand supporting evidence for a licensure decision without a directculture of Mtb is not yet clear. Whether regulators would acceptdata from the nucleic acid-detecting GeneXpert, or infection-basedendpoints (such as IGRA tests) rather than endpoints based on overtclinical or culture-positive disease remain open issues. Similarly,discussions concerning the use of population or community ran-domized study designs rather than large double-blind individualrandomized studies are ongoing. There is clearly a need to engagethe TB community, along with the regulatory experts, to begin dis-cussions concerning these and other licensure issues [10].

We have no clear preclinical models upon which to identify the“best” vaccine Mtb antigens, as many TB vaccine animal challenge

research and development of vaccines for tuberculosis. Vaccine

models have a narrow dynamic range of responses that do not allowfor easy differentiation among candidates. This limitation is beingaddressed by refinement of the mouse, guinea pig and macaquemodels to better approximate natural infection by Mtb and better

Page 3: 2016 Status of Vaccine Research and Development of Vaccines for Tuberculosis. VACCINE

ARTICLE IN PRESSG ModelJVAC-17430; No. of Pages 4

T.G. Evans et al. / Vaccine xxx (2016) xxx–xxx 3

Table 1Development status of current vaccine candidates.

Candidate name/identifier Developer Type Phase I Phase IIa Phase IIb Phase III

Ad5 Ag85A [McMaster University, CanSino] Human adenovirus 5 1 antigen XTB/Flu-04L [RIBSP] Attenuated influenza XDAR-901 [Dartmouth, Aeras] Heat-killed NTM XChAdOx1.85A/MVA85A [Oxford] Chimp adenovirus/modified vaccinia XMTBVAC [TBVI, Zaragoza, Biofabri] Live attenuated TB XID93 + GLA-SE [IDRI, Aeras] 4 Ag adjuvanted fusion protein XVPM 1002 [Max Planck, VPM, TBVI, Serum Institute of India] Modified recombinant BCG XH1 + IC31 [Statens Serum Institut (SSI), TBVI, EDCTP] 2 Ag adjuvanted fusion protein XRUTI [Archivel Farma, S.L] Lysate of Mtb XH4/Aeras-404 + IC31 [SSI, Sanofi-Pasteur, Aeras, Intercell] 2 Ag adjuvanted fusion protein XH56/Aeras-456 + IC31 [SSI, Aeras, Intercell] 3 Ag adjuvanted fusion protein X

Ag adsate o

mPanmp

ipbvivecco

4

sascIDaapeNcAopio

ldtppoaws

t

M72 + AS01E [GlaxoSmithKline, Aeras] 2M. Vaccae [Anhui ZhifeiLongcon, China] Ly

imic human disease. The use of low dose challenge, sophisticatedET/CT imaging techniques, and novel vaccine candidates (suchs H56, Cytomegalovirus (CMV) approaches, and aerosolized ade-ovirus vaccines) have recently shown that the macaque modelay potentially be useful to delineate correlates of vaccine-induced

rotection.In human biomarker studies, gene expression patterns of

nflammatory biosignatures have correlated with risk for TB diseaserogression, and with the extent of radiographic involvement inoth active and latent TB cases [11,12]. In response to these data, TBaccine developers are pursuing a systems immunology approachn which gene expression signatures are compared in samples fromarious time points [13]. These signatures are then correlated toither specific measures of immunogenicity, or to protection in effi-acy studies. This method allows for a broader unbiased net to beast in assessing immune responses, and in searching for a correlatef vaccine-induce protection.

. Status of vaccine R&D activities

Sixteen candidate vaccines have moved forward into clinicaltudies in the last 10 years. Over a dozen candidates currentlyre under active clinical study, summarized in Table 1 [14]. Thesetudies are supported by a combination of multinational pharma-eutical companies (GSK, Crucell, Sanofi-Pasteur, Serum Institute ofndia), smaller vaccine developers (Staten Serum Institut, Infectiousisease Research Institute, Vakzine Projekt Management, BioFabri,nd others), academics (MPIIIB, US and European Universities),nd TB vaccine-focused non-profits (Aeras and TBVI). Support hasrimarily been through the Bill &Melinda Gates Foundation, gov-rnmental organizations such as the EC, DGIS, DIFD, NORAD and theIH, and through direct investments of companies. The businessase for TB vaccines, as recently developed by a working group oferas, TBVI, EC, EIB, and BMGF, is compelling, with potential returnsn investment that range into the billions of dollars. Due to the higherceived scientific risks, however, it has been difficult to engage

ndustry, whether large or small, from entering into this field with-ut substantial early subsidization from granting organizations.

Early signals of potential efficacy may be obtained from the fourarge trials mentioned above over the next 3 years: prevention ofisease by vaccination with the GSK or M. vaccae product; preven-ion of infection through use of the Sanofi candidate; and possiblyrevention of recurrence using the IDRI candidate. If any of theserove promising, there will be considerable momentum to carryne or more of these into larger efficacy studies. The use of a live,ttenuated TB vaccine, and the exploration of whole cell vaccines,

Please cite this article in press as: Evans TG, et al. Status of vaccine

(2016), http://dx.doi.org/10.1016/j.vaccine.2016.02.079

ill continue with a focus on better defining an immune responseurrogate.

Other innovative leads that will be aggressively pursued overhe next 5 years will include the use of aerosolized adenoviral

juvanted fusion protein Xf NTM X

and other candidates, either alone or in combination. The com-bination of aerosolized adenoviral vectored vaccines followed bymodified vaccinia Ankara has been especially promising in bothpreclinical models and in early human trials. CMV candidates willlikely move forward in both the TB and HIV area, as they induceprolonged and high levels of effector T cells in the mucosal loca-tion where the pathogen first encounters the human host. Otherpromising leads include intranasal attenuated parainfluenza orinfluenza-vectored viruses for induction of mucosal immunity, self-replicating RNA candidates, and electroporated DNA vaccines. Thevalue of a number of novel BCG replacement strategies will alsobecome clearer. Systematic study of heterologous platform combi-nations using common antigen sets is now underway for many ofthese approaches.

A variety of highly novel vaccine candidates are being devel-oped by academia, industry, and expert consortia, utilizing anumber of innovative and diverse approaches. There is renewedemphasis on prevention of infection through antibody-mediatedor other immunologic mechanisms; in addition, optimal glycolipidconstructs and adjuvants that induce responses via the CD1 sys-tem, as well as antigens for induction of gamma delta T cells ormucosal invariant T cells (MAITS), are being explored. All of theseapproaches are high risk/high reward investigations that expandthe immunologic response space being probed by TB vaccine can-didates. Testing of these novel candidates is being facilitated by theconcurrent development of new animal models of natural trans-mission. The models include human- or primate-to-guinea pigtransmission, as well as novel macaque-to-macaque transmissionin an environmentally controlled setting. While these programsprogress, significant attempts will be undertaken to build a safemycobacterial construct that can be used in a human TB challengemodel, which would open wide the field of early clinical vaccineassessment. The strain of Mtb used will need to exhibit some degreeof low level replication, a failsafe kill switch and a second attenua-tion mechanism to help ensure safety. The strain must be modifiedsuch that the bacterial burden can be easily measured in the chal-lenged subject through the imaging of a luminescent marker or bymeasuring a soluble, secreted marker in blood, exhaled air or urine.

There is also an important need to use a more rational approachfor selection of TB vaccine candidates for future studies [15]. First,there is a need to ensure that each vaccine carried forward intoefficacy studies addresses a new hypothesis, rather than pursuinga vaccine approach that has already been moved forward into effi-cacy trials. It will be critical to then choose only the best vaccinecandidate among those that are likely to induce a similar magni-tude and phenotype of immune responses. From candidates that

research and development of vaccines for tuberculosis. Vaccine

have similar target profiles, head to head comparisons of candidatesin animal and early human studies would be optimal, and mech-anisms and incentives (such as support from funding agencies) todo such comparisons are needed. This approach also implies the

Page 4: 2016 Status of Vaccine Research and Development of Vaccines for Tuberculosis. VACCINE

ING ModelJ

4 ccine

nnt

5

aTtanamGt

12

3

C

tOtatc

[

[

[

[

[14] Kaufmann SH, Lange C, Rao M, Balaji KN, Lotze M, Schito M, et al. Progress in

ARTICLEVAC-17430; No. of Pages 4

T.G. Evans et al. / Va

eed for a diverse and robust pipeline of candidates representingot just a set of minor improvements but truly novel approacheshat test different immunologic hypotheses.

. Likelihood for financing

TB continues to be a massive problem in both infants and youngdults. Accordingly, it is highly likely that GAVI would support aB vaccine that could save hundreds of thousands of lives. TB ishe 9th leading cause of death in the world, and it is inconceiv-ble that an effective intervention for the poorest countries wouldot be supported by the global health community. The TB researchnd funder community is attempting to organize a global portfolioanagement and information secretariat, known by the acronymTBVP (Global TB Vaccine Partnership), which may take shape in

he coming year. This group aims to have the following functions:

. Attract additional funders for TB vaccine R&D.

. Put TB vaccine R&D higher on the global health and researchagenda AND on global political agenda.

. Develop a consensus on TB vaccine priorities to guide fundingallocation decisions (a. portfolio management recommendationson clinical trials and its funding allocation; b. overall scientificstrategy TB vaccine development).

onflict of interest statement

Since August 2015 Thomas G. Evans has served as a consultanto TomegaVax, a small biotechnology company located in Portland,regon, which owns the rights to the CMV vaccine that is men-

Please cite this article in press as: Evans TG, et al. Status of vaccine

(2016), http://dx.doi.org/10.1016/j.vaccine.2016.02.079

ioned in the article. As of yet, he has not received compensation,nd that work was performed entirely by academic colleagues athe Oregon Health Sciences University. Other authors declare noonflict of interest.

[

PRESSxxx (2016) xxx–xxx

References

[1] Barker LF, Brennan MJ, Rosenstein PK, Sadoff JC. Tuberculosis vaccine research:the impact of immunology. Curr Opin Immunol 2009;21:331–8.

[2] Abu-Raddad LJ, Sabatelli L, Achterberg JT, Sugimoto JD, Longini Jr IM, Dye C,et al. Epidemiological benefits of more-effective tuberculosis vaccines, drugs,and diagnostics. Proc Natl Acad Sci U S A 2009;106:13980–5.

[3] Andrews JR, Noubary F, Walensky RP, Cerda R, Losina E, Horsburgh CR. Riskof progression to active tuberculosis following reinfection with Mycobacteriumtuberculosis. Clin Infect Dis 2012;54:784–91.

[4] Comas I, Chakravartti J, Small PM, Galagan J, Niemann S, Kremer K, et al.Human T cell epitopes of Mycobacterium tuberculosis are evolutionarily hyper-conserved. Nat Genet 2010;42:498–503.

[5] Rodrigues LC, Diwan VK, Wheeler JG. Protective effect of BCG against tuber-culous meningitis and miliary tuberculosis: a meta-analysis. Int J Epidemiol1993;22:1154–8.

[6] Colditz GA, Brewer TF, Berkey CS, Wilson ME, Burdick E, Fineberg HV, et al.Efficacy of BCG vaccine in the prevention of tuberculosis. Meta-analysis of thepublished literature. J Am Med Assoc 1994;271:698–702.

[7] Kaufmann SH, Hussey G, Lambert PH. New vaccines for tuberculosis. Lancet2010;375:2110–9.

[8] Baldwin SL, D’Souza C, Roberts AD, Kelly BP, Frank AA, Lui MA, et al. Evaluationof new vaccines in the mouse and guinea pig model of tuberculosis. InfectImmun 1998;66:2951–9.

[9] Tameris MD, Hatherill M, Landry BS, Scriba TJ, Snowden MA, Lockhart S, et al.Safety and efficacy of MVA85A, a new tuberculosis vaccine, in infants previouslyvaccinated with BCG: a randomized, placebo-controlled phase 2b trial. Lancet2013;381(9871):1021–8.

10] Hatherill M, Verver S, Mahomed H. The taskforce on clinical research issues STP-WGoTV. Consensus statement on diagnostic end points for infant tuberculosisvaccine trials. Clin Infect Dis 2011.

11] Berry MP, Graham CM, McNab FW, Xu Z, Bloch SA, Oni T, et al. Aninterferon-inducible neutrophil-driven blood transcriptional signature inhuman tuberculosis. Nature 2010;466:973–7.

12] Ottenhoff THM, Ellner JJ, Kaufmann SHE. Ten challenges for TB biomarkers.Tuberculosis 2012;92:S17–20.

13] Pulendran B. Learning immunology from the yellow fever vaccine: innateimmunity to systems vaccinology. Nat Rev Immunol 2009;9:741–7.

research and development of vaccines for tuberculosis. Vaccine

tuberculosis vaccine development and host-directed therapies: a state of theart review. Lancet Respir. Med 2014;2:301–20.

15] Barker L, Hessel L, Walker B. Rational approach to selection and clinical devel-opment of TB vaccine candidates. Tuberculosis 2012;92:S25–9.