Long-term follow up of human T-cell responses to conserved HIV-1 regions elicited by DNA/simian adenovirus/MVA vaccine regimens

Nathifa Moyo, Nicola J Borthwick, Edmund G Wee, Silvia Capucci, Alison Crook, Lucy Dorrell, Tomáš Hanke, Nathifa Moyo, Nicola J Borthwick, Edmund G Wee, Silvia Capucci, Alison Crook, Lucy Dorrell, Tomáš Hanke

Abstract

Background: Durability of vaccine-elicited immune responses is one of the key determinants for vaccine success. Our aim is to develop a vaccination strategy against the human immunodeficiency virus type 1 (HIV-1), which induces protective and durable CD8+ T-cell responses. The central theorem of our approach is to focus T cells on highly conserved regions of the HIV-1 proteome and this is achieved through the use of the first-generation conserved vaccine immunogen HIVconsv. This immunogen vectored by plasmid DNA, simian adenovirus and poxvirus MVA was tested in healthy, HIV-1-negative adults in UK and induced high magnitudes of HIVconsv-specific plurifunctional CD8+ T cells capable of in vitro HIV-1 inhibition. Here, we assessed the durability of these responses.

Methods: Vaccine recipients in trial HIV-CORE 002 were invited to provide a blood sample at 1 and 2 years after vaccination. Their PBMCs were tested in IFN-γ ELISPOT, 25-analyte Luminex, CFSE proliferation and intracellular cytokine staining assays, the last enhanced by HLA-peptide dextramer analysis.

Results: 12/12 (1 year) and 8/8 (2 years) returning subjects had median (range) of 990 (150-2495) and 763 (70-1745) IFN-γ SFU/106 PBMC specific for HIVconsv, respectively, and recognized 5 (1-6) out of 6 peptide pools at 2 years. Over one-half of the HIVconsv-specific cells expressed at least 3 functions IFN-γ, TNF-α and CD107a, and were capable of proliferation. Among dextramer-reactive cells, naïve, transitional, effector and terminally differentiated memory subsets were similarly represented.

Conclusions: First generation HIVconsv vaccine induced human T cells, which were plurifunctional and persisted for at least 2 years.

Trial registration: ClinicalTrials.gov NCT01151319.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Fig 1. HIVconsv vaccine-induced human T-cell responses.
Fig 1. HIVconsv vaccine-induced human T-cell responses.
(A) Schematic representations of the HIVconsv immunogen and six pools of a total of 199 overlapping peptides used for the IFN-γ ELISPOT assay. HIVconsv is a chimeric protein assembled from 14 highly conserved regions of the HIV-1 proteome, the HIV-1 protein origins of which are colour-coded below. Each of the regions uses a consensus amino acid sequence of the HIV-1 clade indicated above the schematics. The C-terminal epitopes include Mamu-A*01- and H-2d-restricted immunodominant CTL epitopes and a Pk tag recognized by a monoclonal antibody, which together facilitate the quality control of the vaccines. (B) Fresh ex vivo net total (sum of six pools) IFN-γ ELISPOT assay frequencies of HIVconsv-specific T cells of returning healthy HIV-1-negative volunteers of trial HIV-CORE 002, who received either the CM (top) and DDDCM (bottom) vaccine regimen, are shown separately. Time points ‘S’ (screen) and 0–28 weeks were previously published [68] and are shown for completeness. ‘1y’ and ‘2y’ indicate 1 and 2 years after the last M (MVA.HIVconsv) vaccine administration at weeks 8 (CM) and 20 (DDDCM) indicated below the graphs. Volunteers’ ID numbers are shown on the graph legend. (C) Fresh ex vivo net total IFN-γ ELISPOT assay frequencies of HIVconsv-specific T cells after 1 and 2 years. The horizontal bars represent median frequencies for each regimen separately. The two time points and regimens were not statistically separable.
Fig 2. Long-term broad functional capacity of…
Fig 2. Long-term broad functional capacity of HIVconsv vaccine-elicited human T cells.
Cryopreserved and thawed PBMC samples from pre-vaccination (Pre), and 1 (1y) and 2 (2y) years after the last vaccine administration were re-stimulated with pools of stimulatory 15-mer peptides assembled for each returning volunteer individually for 48 hours and the tissue culture supernatants were analyzed in a 25-analyte Luminex assay. Individual values are shown with indicated median and boxed interquartile range. Volunteer 416 is depicted in blue. Only cytokines and chemokines with positive responses are shown. Results were analyzed using the Mann-Whitney U test for comparison between the long-term samples vs the pre-vaccination sample. Significant P values are indicated by asterisks, whereby: *—less that 0.05; **—less than 0.01; ***—less than 0.001; ****—less than 0.0001.
Fig 3. Proliferative capacity of long-term vaccine-elicited…
Fig 3. Proliferative capacity of long-term vaccine-elicited human T cells.
Cryopreserved and thawed PBMCs from pre-vaccination (Pre), 6 months (6m), and 1 (1y) and 2 (2y) years after the last vaccine administration were labelled with CFSE, stimulated with pools of stimulatory 15-mer peptides assembled for each returning volunteer for 5 days and analyzed using flow cytometry for dividing (decreased CFSE) CD4+ and CD8+ T cells. The CM and DDDCM vaccination regimens are indicated above. The gating strategy is given in S1 Fig. Individual values are shown with median as a horizontal line.
Fig 4. Functionality and memory subtypes of…
Fig 4. Functionality and memory subtypes of vaccine-elicited human CD4+ and CD8+ T cells.
Frozen and thawed PBMCs from 6 months (6m), and 1 (1y) and 2 (2y) years after the last vaccine administration were stimulated with personalized 15-mer peptide pools and subjected to ICS assay. The pie charts refer to the plurifunctionality of the responses defined by Boolean gating. The results from six individuals are shown. For the best responder 416, memory phenotypes were investigated by flow cytometry using IFN-⅟ release to identify antigen-specific cells. Memory subsets are defined as TN−naïve T cells (CD45RAhiCCR7hiCD27hi). TCM—central memory (CD45RAloCCR7hiCD27hi), TTM—transitional memory (CD45RAloCCR7loCD27hi), TEM—effector memory (CD45RAloCCR7loCD27lo), and TTD—terminally differentiated (CD45RAhiCCR7loCD27lo).
Fig 5. Dextramer-aided analysis of human YV9-…
Fig 5. Dextramer-aided analysis of human YV9- and KV10-specific CD8+ T cells.
For HLA-A*02:01-positive vaccine recipients, frozen and thawed PBMCs from 6 months (6m), and 1 (1y) and 2 (2y) years after vaccination were incubated with HLA-A*02:01-YV9 (A and C) or HLA-A*02:01-KV10 (B and D) dextramers together with other cell-surface markers and analyzed using flow cytometry (S3 Fig). Alternatively, PBMCs of subject 416 were stimulated with personalized 15-mer peptide pools and reacted with a panel of functional mAbs together with HLA-A*02:01-YV9 (E and F) or HLA-A*02:01-KV10 (H and I) dextramers. The pie charts (F and I) show the plurifunctionality of dextramer positive CD8+ T cells. PBMCs from subject 416 reactive with the HLA-A*02:01-YV9 (G) or HLA-A*02:01-KV10 (J) dextramers were phenotyped for memory subsets defined as TN−naïve T cells (CD45RAhiCCR7hiCD27hi). TCM—central memory (CD45RAloCCR7hiCD27hi), TTM—transitional memory (CD45RAloCCR7loCD27hi), TEM—effector memory (CD45RAloCCR7loCD27lo), and TTD—terminally differentiated (CD45RAhiCCR7loCD27lo) (S3 Fig).

References

    1. Plotkin SA. Vaccines: correlates of vaccine-induced immunity. Clin Infect Dis. 2008;47(3):401–9. Epub 2008/06/19. doi: .
    1. Pulendran B, Ahmed R. Translating innate immunity into immunological memory: implications for vaccine development. Cell. 2006;124(4):849–63. doi: .
    1. Appay V, Nixon DF, Donahoe SM, Gillespie GM, Dong T, King A, et al. HIV-specific CD8+ T cells produce antiviral cytokines but are impaired in cytolytic function. The Journal of experimental medicine. 2000;192:63–75.
    1. Catalina MD, Sullivan JL, Brody RM, Luzuriaga K. Phenotypic and functional heterogeneity of EBV epitope-specific CD8+ T cells. Journal of immunology. 2002;168(8):4184–91. .
    1. Gillespie GM, Wills MR, Appay V, O'Callaghan C, Murphy M, Smith N, et al. Functional heterogeneity and high frequencies of cytomegalovirus-specific CD8(+) T lymphocytes in healthy seropositive donors. Journal of virology. 2000;74(17):8140–50. ;
    1. Hamann D, Baars PA, Rep MH, Hooibrink B, Kerkhof-Garde SR, Klein MR, et al. Phenotypic and functional separation of memory and effector human CD8+ T cells. J Exp Med. 1997;186:1407–18.
    1. Makedonas G, Hutnick N, Haney D, Amick AC, Gardner J, Cosma G, et al. Perforin and IL-2 upregulation define qualitative differences among highly functional virus-specific human CD8 T cells. PLoS pathogens. 2010;6(3):e1000798 doi: ;
    1. Hanke T. Conserved immunogens in prime-bost strategies for the next-generation HIV-1 vaccines. Expert Opin Biol Ther. 2014;14:601–16. doi: .
    1. Haynes BF, Shaw GM, Korber B, Kelsoe G, Sodroski J, Hahn BH, et al. HIV-Host Interactions: Implications for Vaccine Design. Cell host & microbe. 2016;19(3):292–303. doi: ;
    1. Borrow P, Lewicki H, Hahn BE, Shaw GM, Oldstone MB. Virus-specific CD8+ CTL activity associated with control of viremia in primary HIV-1 infection. J Virol. 1994;68:6103–10.
    1. Fellay J, Shianna KV, Ge D, Colombo S, Ledergerber B, Weale M, et al. A whole-genome association study of major determinants for host control of HIV-1. Science. 2007;317(5840):944–7. doi: .
    1. Goonetilleke N, Liu MK, Salazar-Gonzalez JF, Ferrari G, Giorgi E, Ganusov VV, et al. The first T cell response to transmitted/founder virus contributes to the control of acute viremia in HIV-1 infection. The Journal of experimental medicine. 2009;206(6):1253–72. Epub 2009/06/03. doi: .
    1. Goulder PJ, Brander C, Tang Y, Tremblay C, Colbert RA, Addo MM, et al. Evolution and transmission of stable CTL escape mutations in HIV infection. Nature. 2001;412(6844):334–8. doi: .
    1. Koup RA, Safrit JT, Cao Y, Andrews CA, McLoed G, Borkowsky W, et al. Temporal association of cellular immune responses with the initial control of viremia in primary human immunodeficiency virus type 1 syndrome. J Virol. 1994;68:4650–5.
    1. Ogg GS, Jin X, Bonhoeffer S, Dunbar PR, Nowak MA, Monard S, et al. Quantitation of HIV-1-specific cytotoxic T lymphocytes and plasma load of viral RNA. Science. 1998; 279:2103–6.
    1. Carrington M, O'Brien SJ. The influence of HLA genotype on AIDS. Annu Rev Med. 2003;54:535–51. doi: .
    1. Dalmasso C, Carpentier W, Meyer L, Rouzioux C, Goujard C, Chaix ML, et al. Distinct genetic loci control plasma HIV-RNA and cellular HIV-DNA levels in HIV-1 infection: the ANRS Genome Wide Association 01 study. PloS one. 2008;3(12):e3907 doi: ;
    1. Salazar-Gonzalez JF, Salazar MG, Keele BF, Learn GH, Giorgi EE, Li H, et al. Genetic identity, biological phenotype, and evolutionary pathways of transmitted/founder viruses in acute and early HIV-1 infection. The Journal of experimental medicine. 2009;206(6):1273–89. Epub 2009/06/03. doi: .
    1. Carrington M, Nelson GW, Martin MP, Kissner T, Vlahov D, Goedert JJ, et al. HLA and HIV-1: heterozygote advantage and B*35-Cw*04 disadvantage. Science. 1999;283:1748–52.
    1. Goulder PJ, Walker BD. HIV and HLA class I: an evolving relationship. Immunity. 2012;37(3):426–40. doi: ;
    1. Goulder PJR, Phillips RE, Colbert RA, McAdam S, Ogg G, Nowak MA, et al. Late escape from an immunodominant cytotoxic T-lymphocyte response associated with progression to AIDS. Nature Med. 1997;3:212–7.
    1. Mothe B, Llano A, Ibarrondo J, Daniels M, Miranda C, Zamarreno J, et al. Definition of the viral targets of protective HIV-1-specific T cell responses. J Transl Med. 2011;9:208 Epub 2011/12/14. doi: .
    1. Murakoshi H, Akahoshi T, Koyanagi M, Chikata T, Naruto T, Maruyama R, et al. Clinical Control of HIV-1 by Cytotoxic T Cells Specific for Multiple Conserved Epitopes. Journal of virology. 2015;89:5330–9. doi: .
    1. Ondondo B, Murakoshi H, Clutton G, Abdul-Jawad S, Wee EG, Gatanaga H, et al. Novel Conserved-region T-cell Mosaic Vaccine With High Global HIV-1 Coverage Is Recognized by Protective Responses in Untreated Infection. Mol Ther. 2016;24(4):832–42. doi: .
    1. Matano T, Shibata R, Siemon C, Connors M, Lane HC, Martin MA. Administration of an anti-CD8 monoclonal antibody interferes with the clearance of chimeric simian/human immunodeficiency virus during primary infections of rhesus macaques. Journal of virology. 1998;72(1):164–9.
    1. Schmitz JE, Kuroda MJ, Sasseville VG, Simon MA, Lifton MA, Racz P, et al. Control of viremia in simian immunodeficiency virus infection by CD8+ lymphocytes. Science. 1999;283:857–60.
    1. Hansen SG, Ford JC, Lewis MS, Ventura AB, Hughes CM, Coyne-Johnson L, et al. Profound early control of highly pathogenic SIV by an effector memory T-cell vaccine. Nature. 2011;473(7348):523–7. Epub 2011/05/13. doi: .
    1. Hansen SG, Sacha JB, Hughes CM, Ford JC, Burwitz BJ, Scholz I, et al. Cytomegalovirus vectors violate CD8+ T cell epitope recognition paradigms. Science. 2013;340(6135):1237874 Epub 2013/05/25. doi: .
    1. Hansen SG, Vieville C, Whizin N, Coyne-Johnson L, Siess DC, Drummond DD, et al. Effector memory T cell responses are associated with protection of rhesus monkeys from mucosal simian immunodeficiency virus challenge. Nature medicine. 2009;15(3):293–9. Epub 2009/02/17. doi: .
    1. Hansen SG, Wu HL, Burwitz BJ, Hughes CM, Hammond KB, Ventura AB, et al. Broadly targeted CD8(+) T cell responses restricted by major histocompatibility complex E. Science. 2016;351(6274):714–20. doi: ;
    1. Hansen SG, Piatak M Jr., Ventura AB, Hughes CM, Gilbride RM, Ford JC, et al. Immune clearance of highly pathogenic SIV infection. Nature. 2013;502(7469):100–4. Epub 2013/09/13. doi: ;
    1. Demers KR, Reuter MA, Betts MR. CD8(+) T-cell effector function and transcriptional regulation during HIV pathogenesis. Immunological reviews. 2013;254(1):190–206. doi: ;
    1. McMichael AJ, Haynes BF. Lessons learned from HIV-1 vaccine trials: new priorities and directions. Nat Immunol. 2012;13(5):423–7. Epub 2012/04/20. doi: .
    1. Kiepiela P, Ngumbela K, Thobakgale C, Ramduth D, Honeyborne I, Moodley E, et al. CD8+ T-cell responses to different HIV proteins have discordant associations with viral load. Nature medicine. 2007;13(1):46–53. doi: .
    1. Rolland M, Heckerman D, Deng W, Rousseau CM, Coovadia H, Bishop K, et al. Broad and Gag-Biased HIV-1 Epitope Repertoires Are Associated with Lower Viral Loads. PloS one. 2008;3(1):e1424 doi: .
    1. Lee JK, Stewart-Jones G, Dong T, Harlos K, Di Gleria K, Dorrell L, et al. T cell cross-reactivity and conformational changes during TCR engagement. The Journal of experimental medicine. 2004;200(11):1455–66. doi: .
    1. Almeida JR, Price DA, Papagno L, Arkoub ZA, Sauce D, Bornstein E, et al. Superior control of HIV-1 replication by CD8+ T cells is reflected by their avidity, polyfunctionality, and clonal turnover. The Journal of experimental medicine. 2007;204(10):2473–85. doi: .
    1. Migueles SA, Laborico AC, Shupert WL, Sabbaghian MS, Rabin R, Hallahan CW, et al. HIV-specific CD8+ T cell proliferation is coupled to perforin expression and is maintained in nonprogressors. Nat Immunol. 2002;3(11):1061–8. Epub 2002/10/09. doi: .
    1. Betts MR, Nason MC, West SM, De Rosa SC, Migueles SA, Abraham J, et al. HIV nonprogressors preferentially maintain highly functional HIV-specific CD8+ T cells. Blood. 2006;107(12):4781–9. doi: .
    1. Yang H, Wu H, Hancock G, Clutton G, Sande N, Xu X, et al. Antiviral inhibitory capacity of CD8+ T cells predicts the rate of CD4+ T-cell decline in HIV-1 infection. J Infect Dis. 2012;206(4):552–61. Epub 2012/06/20. doi: .
    1. Groettrup M, Soza A, Eggers M, Kuehn L, Dick TP, Schild H, et al. A role for the proteasome regulator PA28alpha in antigen presentation. Nature. 1996;381(6578):166–8. Epub 1996/05/09. doi: .
    1. Cramer LA, Nelson SL, Klemsz MJ. Synergistic induction of the Tap-1 gene by IFN-gamma and lipopolysaccharide in macrophages is regulated by STAT1. Journal of immunology. 2000;165(6):3190–7. .
    1. Epperson DE, Arnold D, Spies T, Cresswell P, Pober JS, Johnson DR. Cytokines increase transporter in antigen processing-1 expression more rapidly than HLA class I expression in endothelial cells. Journal of immunology. 1992;149(10):3297–301. .
    1. Johnson DR, Pober JS. Tumor necrosis factor and immune interferon synergistically increase transcription of HLA class I heavy- and light-chain genes in vascular endothelium. Proceedings of the National Academy of Sciences of the United States of America. 1990;87(13):5183–7. ;
    1. Wallach D, Fellous M, Revel M. Preferential effect of gamma interferon on the synthesis of HLA antigens and their mRNAs in human cells. Nature. 1982;299(5886):833–6. .
    1. Aggarwal BB. Signalling pathways of the TNF superfamily: a double-edged sword. Nat Rev Immunol. 2003;3(9):745–56. doi: .
    1. Kull FC Jr. The TNF receptor in TNF-mediated cytotoxicity. Nat Immun Cell Growth Regul. 1988;7(5–6):254–65. .
    1. Lazdins JK, Grell M, Walker MR, Woods-Cook K, Scheurich P, Pfizenmaier K. Membrane tumor necrosis factor (TNF) induced cooperative signaling of TNFR60 and TNFR80 favors induction of cell death rather than virus production in HIV-infected T cells. The Journal of experimental medicine. 1997;185(1):81–90. ;
    1. Dinarello CA, Mier JW. Interleukins. Annu Rev Med. 1986;37:173–8. doi: .
    1. Peters PJ, Borst J, Oorschot V, Fukuda M, Krahenbuhl O, Tschopp J, et al. Cytotoxic T lymphocyte granules are secretory lysosomes, containing both perforin and granzymes. The Journal of experimental medicine. 1991;173(5):1099–109. ;
    1. Shankar P, Xu Z, Lieberman J. Viral-specific cytotoxic T lymphocytes lyse human immunodeficiency virus-infected primary T lymphocytes by the granule exocytosis pathway. Blood. 1999;94(9):3084–93. .
    1. Betts MR, Brenchley JM, Price DA, De Rosa SC, Douek DC, Roederer M, et al. Sensitive and viable identification of antigen-specific CD8+ T cells by a flow cytometric assay for degranulation. J Immunol Methods. 2003;281(1–2):65–78. Epub 2003/10/29. .
    1. Rubio V, Stuge TB, Singh N, Betts MR, Weber JS, Roederer M, et al. Ex vivo identification, isolation and analysis of tumor-cytolytic T cells. Nature medicine. 2003;9(11):1377–82. doi: .
    1. Rouvier E, Luciani MF, Golstein P. Fas involvement in Ca(2+)-independent T cell-mediated cytotoxicity. The Journal of experimental medicine. 1993;177(1):195–200. ;
    1. Letourneau S, Im E-J, Mashishi T, Brereton C, Bridgeman A, Yang H, et al. Design and pre-clinical evaluation of a universal HIV-1 vaccine. PloS one. 2007;2:e984 doi: .
    1. Deng K, Pertea M, Rongvaux A, Wang L, Durand CM, Ghiaur G, et al. Broad CTL response is required to clear latent HIV-1 due to dominance of escape mutations. Nature. 2015;517(7534):381–5. doi: .
    1. Ferguson AL, Mann JK, Omarjee S, Ndung'u T, Walker BD, Chakraborty AK. Translating HIV Sequences into Quantitative Fitness Landscapes Predicts Viral Vulnerabilities for Rational Immunogen Design. Immunity. 2013;38(3):606–17. Epub 2013/03/26. doi: .
    1. Frahm N, Kiepiela P, Adams S, Linde CH, Hewitt HS, Sango K, et al. Control of human immunodeficiency virus replication by cytotoxic T lymphocytes targeting subdominant epitopes. Nat Immunol. 2006;7(2):173–8. doi: .
    1. Hancock G, Yang H, Yorke E, Wainwright E, Bourne V, Frisbee A, et al. Identification of Effective Subdominant Anti-HIV-1 CD8+ T Cells Within Entire Post-infection and Post-vaccination Immune Responses. PLoS pathogens. 2015;11(2):e1004658 doi: ;
    1. Im E-J, Hong JP, Roshorm Y, Bridgeman A, Létourneau S, Liljeström P, et al. Protective efficacy of serially up-ranked subdominant CD8+ T cell epitopes against virus challenges. PLoS pathogens. 2011;7:e1002041 doi:
    1. Ahmed T, Borthwick NJ, Gilmour J, Hayes P, Dorrell L, Hanke T. Control of HIV-1 replication in vitro by vaccine-induced human CD8 T cells through conserved subdominant Pol epitopes. Vaccine. 2016;34:1215–24. doi: .
    1. Borthwick N, Ahmed T, Ondondo B, Hayes P, Rose A, Ebrahimsa U, et al. Vaccine-elicited human T cells recognizing conserved protein regions inhibit HIV-1. Mol Ther. 2014;22(2):464–75. doi: ;
    1. Borthwick N, Lin Z, Akahoshi T, Llano A, Silva-Arrieta S, Ahmed T, et al. Novel, in-natural-infection subdominant HIV-1 CD8+ T-cell epitopes revealed in human recipients of conserved-region T-cell vaccines. PloS one. 2017;12(4):e0176418 doi: .
    1. Hancock G, Morón-López S, Puertas MC, Giannoulatou E, Rose A, Salgado M, et al. Immunogenicity and impact on the HIV reservoir of a conserved region vaccine, MVA.HIVcons, in antiretroviral therapy-treated subjects: a randomised trial. Journal of the International AIDS Society. 2017;20: 21171.
    1. Mothe B, Manzardo C, Snachez-Bernabeau A, Coll P, Moron S, Peurtas MC, et al. Therapeutic ChAdV63.HIVconsv-MVA.HIVconsv vaccination refocused T cells to conserved regions of HIV in early reated HIV-1 infected individuals (BCN 01 study). Submitted.
    1. Mothe B, Moltó J, Manzardo C, Coll J, Puertas MC, Martinez-Picado J, et al. Viral control induced by HIVconsv vaccines & Romidepsin in early treated individuals. The Conference on Retroviruses and Opportunistic Infections; Seattle, WA, USA2017.
    1. Mutua G, Farah B, Langat R, Indangasi J, Ogola S, Onsembe B, et al. Broad HIV-1 inhibition in vitro by vaccine-elicited CD8+ T cells in African adults. Mol Ther Methods Clin Dev. 2016;3:16061 doi:
    1. Borthwick N, Ahmed T, Ondondo B, Hayes P, Rose A, Ebrahimsa U, et al. Vaccine-elicited human T cells recognizing conserved protein regions inhibit HIV-1. Mol Ther. 2014;22:464–75. Epub 2013/10/30. doi: .
    1. Defawe OD, Fong Y, Vasilyeva E, Pickett M, Carter DK, Gabriel E, et al. Optimization and qualification of a multiplex bead array to assess cytokine and chemokine production by vaccine-specific cells. Journal of immunological methods. 2012;382(1–2):117–28. Epub 2012/05/26. doi: ;
    1. Hersperger AR, Pereyra F, Nason M, Demers K, Sheth P, Shin LY, et al. Perforin expression directly ex vivo by HIV-specific CD8 T-cells is a correlate of HIV elite control. PLoS pathogens. 2010;6(5):e1000917 doi: ;
    1. Hayton EJ, Rose A, Ibrahimsa U, Del Sorbo M, Capone S, Crook A, et al. Safety and tolerability of conserved region vaccines vectored by plasmid DNA, simian adenovirus and modified vaccinia virus ankara administered to human immunodeficiency virus type 1-uninfected adults in a randomized, single-blind phase I trial. PloS one. 2014;9(7):e101591 doi: ;
    1. McElrath MJ, De Rosa SC, Moodie Z, Dubey S, Kierstead L, Janes H, et al. HIV-1 vaccine-induced immunity in the test-of-concept Step Study: a case-cohort analysis. Lancet. 2008;372(9653):1894–905. doi: .
    1. Hammer SM, Sobieszczyk ME, Janes H, Karuna ST, Mulligan MJ, Grove D, et al. Efficacy Trial of a DNA/rAd5 HIV-1 Preventive Vaccine. N Engl J Med. 2013;369:2083–92. Epub 2013/10/09. doi: .
    1. Addo MM, Yu XG, Rathod A, Cohen D, Eldridge RL, Strick D, et al. Comprehensive epitope analysis of human immunodeficiency virus type 1 (HIV-1)-specific T-cell responses directed against the entire expressed HIV-1 genome demonstrate broadly directed responses, but no correlation to viral load. Journal of virology. 2003;77:2081–92. doi:
    1. Betts MR, Ambrozak DR, Douek DC, Bonhoeffer S, Brenchley JM, Casazza JP, et al. Analysis of total human immunodeficiency virus (HIV)-specific CD4(+) and CD8(+) T-cell responses: relationship to viral load in untreated HIV infection. Journal of virology. 2001;75(24):11983–91. doi: .
    1. Gea-Banacloche JC, Migueles SA, Martino L, Shupert WL, McNeil AC, Sabbaghian MS, et al. Maintenance of large numbers of virus-specific CD8+ T cells in HIV-infected progressors and long-term nonprogressors. Journal of immunology. 2000;165(2):1082–92. .
    1. Sandberg JK, Fast NM, Nixon DF. Functional heterogeneity of cytokines and cytolytic effector molecules in human CD8+ T lymphocytes. Journal of immunology. 2001;167(1):181–7. .
    1. Jameson SC, Masopust D. Diversity in T cell memory: an embarrassment of riches. Immunity. 2009;31(6):859–71. doi: ;
    1. Masopust D, Vezys V, Marzo AL, Lefrancois L. Preferential localization of effector memory cells in nonlymphoid tissue. Science. 2001;291(5512):2413–7. doi: .
    1. Sallusto F, Lenig D, Forster R, Lipp M, Lanzavecchia A. Two subsets of memory T lymphocytes with distinct homing potentials and effector functions. Nature. 1999;401(6754):708–12. doi: .
    1. Hikono H, Kohlmeier JE, Takamura S, Wittmer ST, Roberts AD, Woodland DL. Activation phenotype, rather than central- or effector-memory phenotype, predicts the recall efficacy of memory CD8+ T cells. The Journal of experimental medicine. 2007;204(7):1625–36. doi: ;
    1. Olson JA, McDonald-Hyman C, Jameson SC, Hamilton SE. Effector-like CD8(+) T cells in the memory population mediate potent protective immunity. Immunity. 2013;38(6):1250–60. doi: ;
    1. Abboud G, Desai P, Dastmalchi F, Stanfield J, Tahiliani V, Hutchinson TE, et al. Tissue-specific programming of memory CD8 T cell subsets impacts protection against lethal respiratory virus infection. The Journal of experimental medicine. 2016;213(13):2897–911. doi: ;
    1. Soghoian DZ, Jessen H, Flanders M, Sierra-Davidson K, Cutler S, Pertel T, et al. HIV-specific cytolytic CD4 T cell responses during acute HIV infection predict disease outcome. Sci Transl Med. 2012;4(123):123ra25 doi: ;
    1. Johnson S, Eller M, Teigler JE, Maloveste SM, Schultz BT, Soghoian DZ, et al. Cooperativity of HIV-Specific Cytolytic CD4 T Cells and CD8 T Cells in Control of HIV Viremia. Journal of virology. 2015;89(15):7494–505. doi: ;

Source: PubMed

3
Sottoscrivi