The TLR-4 agonist adjuvant, GLA-SE, improves magnitude and quality of immune responses elicited by the ID93 tuberculosis vaccine: first-in-human trial

Rhea N Coler, Tracey A Day, Ruth Ellis, Franco M Piazza, Anna Marie Beckmann, Julie Vergara, Tom Rolf, Lenette Lu, Galit Alter, David Hokey, Lakshmi Jayashankar, Robert Walker, Margaret Ann Snowden, Tom Evans, Ann Ginsberg, Steven G Reed, TBVPX-113 Study Team, Jill Ashman, Zachary K Sagawa, D Tait, Sadritdin Ishmukhamedov, Gretta Blatner, Sharon Sutton, Barbara Shepherd, Casey Johnson, Rhea N Coler, Tracey A Day, Ruth Ellis, Franco M Piazza, Anna Marie Beckmann, Julie Vergara, Tom Rolf, Lenette Lu, Galit Alter, David Hokey, Lakshmi Jayashankar, Robert Walker, Margaret Ann Snowden, Tom Evans, Ann Ginsberg, Steven G Reed, TBVPX-113 Study Team, Jill Ashman, Zachary K Sagawa, D Tait, Sadritdin Ishmukhamedov, Gretta Blatner, Sharon Sutton, Barbara Shepherd, Casey Johnson

Abstract

Tuberculosis (TB) is the leading cause of infectious death worldwide. Development of improved TB vaccines that boost or replace BCG is a major global health goal. ID93 + GLA-SE is a fusion protein TB vaccine candidate combined with the Toll-like Receptor 4 agonist adjuvant, GLA-SE. We conducted a phase 1, randomized, double-blind, dose-escalation clinical trial to evaluate two dose levels of the ID93 antigen, administered intramuscularly alone or in combination with two dose levels of the GLA-SE adjuvant, in 60 BCG-naive, QuantiFERON-negative, healthy adults in the US (ClinicalTrials.gov identifier: NCT01599897). When administered as 3 injections, 28 days apart, all dose levels of ID93 alone and ID93 + GLA-SE demonstrated an acceptable safety profile. All regimens elicited vaccine-specific humoral and cellular responses. Compared with ID93 alone, vaccination with ID93 + GLA-SE elicited higher titers of ID93-specific antibodies, a preferential increase in IgG1 and IgG3 subclasses, and a multifaceted Fc-mediated effector function response. The addition of GLA-SE also enhanced the magnitude and polyfunctional cytokine profile of CD4+ T cells. The data demonstrate an acceptable safety profile and indicate that the GLA-SE adjuvant drives a functional humoral and T-helper 1 type cellular response.

Conflict of interest statement

S.G.R. is a founder of, and holds an equity interest in, Immune Design Corp., a licensee of certain rights associated with GLA. The remaining authors declare no competing interests.

Figures

Fig. 1
Fig. 1
ID93 + GLA-SE elicited significantly higher ID93-specific total IgG antibody responses than those vaccinated with ID93 protein alone. a Magnitude of ID93-specific antibody responses for differing doses of antigen and/or adjuvant. Geometric mean titer and 95% confidence interval as determined by ELISA using serum collected on the days shown. Vaccinations were administered on Days 0, 28, and 56 (arrows) after blood collection. ID93 + GLA-SE elicited significantly higher ID93-specific total IgG antibody responses than those vaccinated with ID93 protein alone. Statistical significance between adjuvant containing regimens and protein alone regimens at each time point was evaluated by two-way ANOVA and Tukey’s multiple comparisons test with p-values indicated as: <0.05 (*), <0.01 (**), <0.0001 (***). b IgG subclass analysis of ID93-specific antibody responses for different antigen and adjuvant doses. Geometric mean titer and 95% confidence interval as determined by ELISA using serum collected at the days shown. Vaccinations were administered on Days 0, 28, and 56. Serum samples were collected on those days prior to study injections. Inclusion of the GLA-SE adjuvant elicited a preferential induction of IgG1 and IgG3 subclasses. c Component antigen specificity of antibody responses for individuals administered different antigen and adjuvant doses. Bar represents group geometric mean titer as determined by ELISA using serum collected at the days shown. Vaccinations were administered on Days 0, 28, and 56. Serum samples were collected on those days prior to study injections. Total IgG responses were measured against all four component antigens with Rv1813 being the highest
Fig. 2
Fig. 2
Magnitude and specificity of antigen-specific T cell response. a Kinetics of ID93 antigen-specific CD4+ T cells measured from cryopreserved PBMC at baseline and 2 weeks after each vaccination. Median frequencies of CD4+ T cells positive for any antigen-specific marker (IFNγ, TNF, IL-2, CD154, IL-4/IL-22, and/or IL-17) as measured by intracellular cytokine staining of antigen (peptide pools)-stimulated PBMCs with unstimulated values subtracted. Error bars show 95% confidence intervals. Vaccinations were administered on Days 0, 28, and 56. b Frequencies of ID93-specific (defined as positive for CD154, IFNγ, TNF, and/or IL-2) for CD4+ and CD8+ T cell responses in individual subjects as measured by intracellular cytokine staining of ID93 protein-stimulated whole blood with unstimulated values subtracted. Vaccinations were administered on Days 0, 28, and 56
Fig. 3
Fig. 3
T cell cytokine profiles from whole blood ICS for Cohorts 1 and 2. Frequencies of ID93-specific CD4 T cells co-expressing different cytokines are shown for Cohorts 1 and 2 as measured by intracellular cytokine staining of ID93-stimulated whole blood with unstimulated values subtracted. The bar shows the median frequency, the box shows the interquartile range, and the whiskers show the maximum and minimum values. Vaccinations were given on Days 0, 28, and 56. a Combined data for 2 μg and 10 μg ID93 recipients, b 2 μg ID93 + 2 μg GLA-SE, c 10 μg ID93 + 2 μg GLA-SE. Wilcoxon matched pairs signed rank test to compare frequencies of ID93-specific cytokine response between Day 0 and Day 42 timepoints. d Proportions of ID93-specific CD4 T cells co-expressing different cytokines are shown for Cohort 1 and 2 subjects administered 2 μg ID93, 10 μg ID93, 2 μg ID93 + 2 μg GLA-SE, or 10 μg ID93 + 2 μg GLA-SE as measured by intracellular cytokine staining of ID93-stimulated whole blood with unstimulated values subtracted. Results are shown for Day 70 (2 weeks post third vaccination). Due to low subject numbers, results for 2 μg ID93 and 10 μg ID93 were combined
Fig. 4
Fig. 4
Vaccine induced antibody response profiles. a Heatmap shows changes in vaccine antigen-specific and control influenza hemagglutinin-specific antibody isotype titers after vaccination. Each row represents an individual. Each column represents an antibody isotype. The vaccine regimen is specified on the left with legend showing that red represents the top 5th percentile in the amount of change (large changes) and blue represents the 95th percentile in the amount of change (small changes). b Changes in ID93-specific antibody-dependent cellular cytotoxicity (ADCC) NK cell production of IFNγ, MIP1β, and CD107a, and antibody-dependent cellular phagocytosis (ADCP) are plotted. Each line represents each individual before (Day 0) and after (Day 84) vaccination and individuals are grouped by vaccine regimen. Statistical significance calculated by Wilcoxon matched pairs signed rank test is indicated (*p < 0.05, ***p < 0.001). c Spearman correlation coefficients between ID93 antibody-specific isotypes and ID93-specific antibody effector functions are depicted by bars with red denoting positive and blue denoting negative values. d Polyfunctionality as defined by total number of ID93-specific antibody effector functions (NK cell-mediated IFNγ, MIP1β, and CD107a, and antibody-dependent cellular phagocytosis) are graphed on a per individual basis. Each individual is represented by a dot, and individuals are grouped into vaccine regimens as noted (purple = ID93, green = ID93 + GLA-SE)
Fig. 5
Fig. 5
Clinical trial CONSORT diagram for TBVPX-113. Flow chart shows the number of subjects entering the study from enrollment, allocation, and follow-up (FU). Subjects missing the second and third injection dose either withdrew consent or were lost to FU. Subjects were in FU for 1 year after the third injection. The Safety Population is defined as all subjects who received at least one study injection; the Per Protocol Population comprises subjects who received all three study injections and completed Day 84. All subjects received at least one study injection and were included in the analyses of safety and immunology

References

    1. World Health Organization. Global Tuberculosis Report 2017. (2017)..
    1. Colditz GA, et al. The efficacy of bacillus Calmette-Guerin vaccination of newborns and infants in the prevention of tuberculosis: meta-analyses of the published literature. Pediatrics. 1995;96:29–35.
    1. Colditz GA, et al. Efficacy of BCG vaccine in the prevention of tuberculosis. Meta-analysis of the published literature. J. Am. Med. Assoc. 1994;271:698–702. doi: 10.1001/jama.1994.03510330076038.
    1. Hawn TR, et al. Tuberculosis vaccines and prevention of infection. Microbiol. Mol. Biol. Rev. 2014;78:650–671. doi: 10.1128/MMBR.00021-14.
    1. Karp CL, Wilson CB, Stuart LM. Tuberculosis vaccines: barriers and prospects on the quest for a transformative tool. Immunol. Rev. 2015;264:363–381. doi: 10.1111/imr.12270.
    1. Cooper AM, Flynn JL. The protective immune response to Mycobacterium tuberculosis. Curr. Opin. Immunol. 1995;7:512–516. doi: 10.1016/0952-7915(95)80096-4.
    1. Flynn JL, Bloom BR. Role of T1 and T2 cytokines in the response to Mycobacterium tuberculosis. Ann. N. Y. Acad. Sci. 1996;795:137–146. doi: 10.1111/j.1749-6632.1996.tb52662.x.
    1. Flynn JL, Ernst JD. Immune responses in tuberculosis. Curr. Opin. Immunol. 2000;12:432–436. doi: 10.1016/S0952-7915(00)00116-3.
    1. Flynn JL, Chan J. Immunology of tuberculosis. Annu. Rev. Immunol. 2001;19:93–129. doi: 10.1146/annurev.immunol.19.1.93.
    1. Flynn JL. Immunology of tuberculosis and implications in vaccine development. Tuberculosis. 2004;84:93–101. doi: 10.1016/j.tube.2003.08.010.
    1. Lu LL, et al. A functional role for antibodies in tuberculosis. Cell. 2016;167:433–443 e414. doi: 10.1016/j.cell.2016.08.072.
    1. Lu, L.L., Suscovich, T.J., Fortune, S.M. & Alter, G. Beyond binding: antibody effector functions in infectious diseases. Nat. Rev. Immunol.18, 46–61 (2017).
    1. Sherman DR, et al. Regulation of the Mycobacterium tuberculosis hypoxic response gene encoding alpha -crystallin. Proc. Natl Acad. Sci. USA. 2001;98:7534–7539. doi: 10.1073/pnas.121172498.
    1. Brennan, M. J. The enigmatic PE/PPE multigene family of mycobacteria and tuberculosis vaccination. Infect. Immun. 85, e00969-16 (2017).
    1. Gey Van Pittius, N. C. et al. The ESAT-6 gene cluster of Mycobacterium tuberculosis and other high G+C Gram-positive bacteria. Genome Biol. 2, RESEARCH0044 (2001).
    1. Bertholet S, et al. Identification of human T cell antigens for the development of vaccines against Mycobacterium tuberculosis. J. Immunol. 2008;181:7948–7957. doi: 10.4049/jimmunol.181.11.7948.
    1. Bertholet S, et al. A defined tuberculosis vaccine candidate boosts BCG and protects against multidrug-resistant Mycobacterium tuberculosis. Sci. Transl. Med. 2010;2:53ra74. doi: 10.1126/scitranslmed.3001094.
    1. Coler RN, et al. Development and characterization of synthetic glucopyranosyl lipid adjuvant system as a vaccine adjuvant. PLoS ONE. 2011;6:e16333. doi: 10.1371/journal.pone.0016333.
    1. Baldwin SL, et al. The importance of adjuvant formulation in the development of a tuberculosis vaccine. J. Immunol. 2012;188:2189–2197. doi: 10.4049/jimmunol.1102696.
    1. Baldwin, S. L. et al. Protection and long-lived immunity induced by the ID93/GLA-SE vaccine candidate against a clinical Mycobacterium tuberculosis isolate. Clin. Vaccine Immunol. (2015).
    1. Cha SB, et al. Pulmonary immunity and durable protection induced by the ID93/GLA-SE vaccine candidate against the hyper-virulent Korean Beijing Mycobacterium tuberculosis strain K. Vaccine. 2016;34:2179–2187. doi: 10.1016/j.vaccine.2016.03.029.
    1. Coler RN, et al. Therapeutic immunization against Mycobacterium tuberculosis is an effective adjunct to antibiotic treatment. J. Infect. Dis. 2013;207:1242–1252. doi: 10.1093/infdis/jis425.
    1. Vidarsson G, Dekkers G, Rispens T. IgG subclasses and allotypes: from structure to effector functions. Front. Immunol. 2014;5:520. doi: 10.3389/fimmu.2014.00520.
    1. Jegaskanda S, Weinfurter JT, Friedrich TC, Kent SJ. Antibody-dependent cellular cytotoxicity is associated with control of pandemic H1N1 influenza virus infection of macaques. J. Virol. 2013;87:5512–5522. doi: 10.1128/JVI.03030-12.
    1. Ackerman ME, et al. A robust, high-throughput assay to determine the phagocytic activity of clinical antibody samples. J. Immunol. Methods. 2011;366:8–19. doi: 10.1016/j.jim.2010.12.016.
    1. Coler RN, et al. From mouse to man: safety, immunogenicity and efficacy of a candidate leishmaniasis vaccine LEISH-F3+GLA-SE. Clin. Transl. Immunol. 2015;4:e35. doi: 10.1038/cti.2015.6.
    1. Tendler M, Almeida M, Simpson A. Development of the Brazilian anti schistosomiasis vaccine based on the recombinant fatty acid binding protein Sm14 plus GLA-SE adjuvant. Front. Immunol. 2015;6:218. doi: 10.3389/fimmu.2015.00218.
    1. Sirima SB, et al. Safety and immunogenicity of a recombinant Plasmodium falciparum AMA1-DiCo malaria vaccine adjuvanted with GLA-SE or Alhydrogel(R) in European and African adults: a phase 1a/1b, randomized, double-blind multi-centre trial. Vaccine. 2017;35:6218–6227. doi: 10.1016/j.vaccine.2017.09.027.
    1. Velez ID, et al. Safety and immunogenicity of a defined vaccine for the prevention of cutaneous leishmaniasis. Vaccine. 2009;28:329–337. doi: 10.1016/j.vaccine.2009.10.045.
    1. Chakravarty J, et al. A clinical trial to evaluate the safety and immunogenicity of the LEISH-F1+MPL-SE vaccine for use in the prevention of visceral leishmaniasis. Vaccine. 2011;29:3531–3537. doi: 10.1016/j.vaccine.2011.02.096.
    1. Aagaard C, Dietrich J, Doherty M, Andersen P. TB vaccines: current status and future perspectives. Immunol. Cell Biol. 2009;87:279–286. doi: 10.1038/icb.2009.14.
    1. Aagaard CS, Hoang TT, Vingsbo-Lundberg C, Dietrich J, Andersen P. Quality and vaccine efficacy of CD4+ T cell responses directed to dominant and subdominant epitopes in ESAT-6 from Mycobacterium tuberculosis. J. Immunol. 2009;183:2659–2668. doi: 10.4049/jimmunol.0900947.
    1. Lindenstrom T, et al. Tuberculosis subunit vaccination provides long-term protective immunity characterized by multifunctional CD4 memory T cells. J. Immunol. 2009;182:8047–8055. doi: 10.4049/jimmunol.0801592.
    1. Geldenhuys H, et al. The tuberculosis vaccine H4:IC31 is safe and induces a persistent polyfunctional CD4 T cell response in South African adults: a randomized controlled trial. Vaccine. 2015;33:3592–3599. doi: 10.1016/j.vaccine.2015.05.036.
    1. Leroux-Roels I, et al. Improved CD4(+) T cell responses to Mycobacterium tuberculosis in PPD-negative adults by M72/AS01 as compared to the M72/AS02 and Mtb72F/AS02 tuberculosis candidate vaccine formulations: a randomized trial. Vaccine. 2013;31:2196–2206. doi: 10.1016/j.vaccine.2012.05.035.
    1. Orr MT, et al. Adjuvant formulation structure and composition are critical for the development of an effective vaccine against tuberculosis. J. Control. Release. 2013;172:190–200. doi: 10.1016/j.jconrel.2013.07.030.
    1. Rook GA. Th2 cytokines in susceptibility to tuberculosis. Curr. Mol. Med. 2007;7:327–337. doi: 10.2174/156652407780598557.
    1. Moguche AO, et al. Antigen availability shapes T cell differentiation and function during tuberculosis. Cell Host Microbe. 2017;21:695–706 e695. doi: 10.1016/j.chom.2017.05.012.
    1. Woodworth JS, et al. Subunit vaccine H56/CAF01 induces a population of circulating CD4 T cells that traffic into the Mycobacterium tuberculosis-infected lung. Mucosal Immunol. 2017;10:555–564. doi: 10.1038/mi.2016.70.
    1. Pethe K, et al. The heparin-binding haemagglutinin of M. tuberculosis is required for extrapulmonary dissemination. Nature. 2001;412:190–194. doi: 10.1038/35084083.
    1. Abebe F, Bjune G. The protective role of antibody responses during Mycobacterium tuberculosis infection. Clin. Exp. Immunol. 2009;157:235–243. doi: 10.1111/j.1365-2249.2009.03967.x.
    1. Alter, G. in Host Response in Tuberculosis, Keystone, Santa Fe, January 22–27, 2015 (Santa Fe; 2015).
    1. Lu LL, Suscovich TJ, Fortune SM, Alter G. Beyond binding: antibody effector functions in infectious diseases. Nat. Rev. Immunol. 2018;18:46–61. doi: 10.1038/nri.2017.106.
    1. Montoya J, et al. A randomized, controlled dose-finding phase II study of the M72/AS01 candidate tuberculosis vaccine in healthy PPD-positive adults. J. Clin. Immunol. 2013;33:1360–1375. doi: 10.1007/s10875-013-9949-3.
    1. Penn-Nicholson A, et al. Safety and immunogenicity of candidate vaccine M72/AS01E in adolescents in a TB endemic setting. Vaccine. 2015;33:4025–4034. doi: 10.1016/j.vaccine.2015.05.088.
    1. Thacher EG, et al. Safety and immunogenicity of the M72/AS01 candidate tuberculosis vaccine in HIV-infected adults on combination antiretroviral therapy: a phase I/II, randomized trial. AIDS. 2014;28:1769–1781. doi: 10.1097/QAD.0000000000000343.
    1. Zvi A, Ariel N, Fulkerson J, Sadoff JC, Shafferman A. Whole genome identification of Mycobacterium tuberculosis vaccine candidates by comprehensive data mining and bioinformatic analyses. BMC Med. Genomics. 2008;1:18. doi: 10.1186/1755-8794-1-18.
    1. He H, Hovey R, Kane J, Singh V, Zahrt TC. MprAB is a stress-responsive two-component system that directly regulates expression of sigma factors SigB and SigE in Mycobacterium tuberculosis. J. Bacteriol. 2006;188:2134–2143. doi: 10.1128/JB.188.6.2134-2143.2006.
    1. Griffin JE, et al. High-resolution phenotypic profiling defines genes essential for mycobacterial growth and cholesterol catabolism. PLoS Pathog. 2011;7:e1002251. doi: 10.1371/journal.ppat.1002251.
    1. Marmiesse M, et al. Macro-array and bioinformatic analyses reveal mycobacterial ‘core’ genes, variation in the ESAT-6 gene family and new phylogenetic markers for the Mycobacterium tuberculosis complex. Microbiology. 2004;150:483–496. doi: 10.1099/mic.0.26662-0.
    1. Graves AJ, Padilla MG, Hokey DA. OMIP-022: comprehensive assessment of antigen-specific human T-cell functionality and memory. Cytom. A. 2014;85:576–579. doi: 10.1002/cyto.a.22478.
    1. Kagina BM, et al. Qualification of a whole blood intracellular cytokine staining assay to measure mycobacteria-specific CD4 and CD8 T cell immunity by flow cytometry. J. Immunol. Methods. 2015;417:22–33. doi: 10.1016/j.jim.2014.12.003.
    1. Brown EP, et al. High-throughput, multiplexed IgG subclassing of antigen-specific antibodies from clinical samples. J. Immunol. Methods. 2012;386:117–123. doi: 10.1016/j.jim.2012.09.007.
    1. Darrah PA, et al. Multifunctional TH1 cells define a correlate of vaccine-mediated protection against Leishmania major. Nat. Med. 2007;13:843–850. doi: 10.1038/nm1592.

Source: PubMed

3
Abonner