Sustained viremia suppression by SHIVSF162P3CN-recalled effector-memory CD8+ T cells after PD1-based vaccination

Yik Chun Wong, Wan Liu, Lok Yan Yim, Xin Li, Hui Wang, Ming Yue, Mengyue Niu, Lin Cheng, Lijun Ling, Yanhua Du, Samantha M Y Chen, Ka-Wai Cheung, Haibo Wang, Xian Tang, Jiansong Tang, Haoji Zhang, Youqiang Song, Lisa A Chakrabarti, Zhiwei Chen, Yik Chun Wong, Wan Liu, Lok Yan Yim, Xin Li, Hui Wang, Ming Yue, Mengyue Niu, Lin Cheng, Lijun Ling, Yanhua Du, Samantha M Y Chen, Ka-Wai Cheung, Haibo Wang, Xian Tang, Jiansong Tang, Haoji Zhang, Youqiang Song, Lisa A Chakrabarti, Zhiwei Chen

Abstract

HIV-1 functional cure requires sustained viral suppression without antiretroviral therapy. While effector-memory CD8+ T lymphocytes are essential for viremia control, few vaccines elicit such cellular immunity that could be potently recalled upon viral infection. Here, we investigated a program death-1 (PD1)-based vaccine by fusion of simian immunodeficiency virus capsid antigen to soluble PD1. Homologous vaccinations suppressed setpoint viremia to undetectable levels in vaccinated macaques following a high-dose intravenous challenge by the pathogenic SHIVSF162P3CN. Poly-functional effector-memory CD8+ T cells were not only induced after vaccination, but were also recalled upon viral challenge for viremia control as determined by CD8 depletion. Vaccine-induced effector memory CD8+ subsets displayed high cytotoxicity-related genes by single-cell analysis. Vaccinees with sustained viremia suppression for over two years responded to boost vaccination without viral rebound. These results demonstrated that PD1-based vaccine-induced effector-memory CD8+ T cells were recalled by AIDS virus infection, providing a potential immunotherapy for functional cure.

Conflict of interest statement

I have read the journal’s policy and the authors of this manuscript have the following competing interests: Z.C. is a co-inventor on the PD1-based vaccine patent. However, ZC and all other authors do not have any competing interests or additional financial interests.

Figures

Fig 1. Immunogenicity of RhPD1-based vaccines in…
Fig 1. Immunogenicity of RhPD1-based vaccines in mice.
(A) Schematic of DNA vaccination in mice. C57BL/6 mice were immunized with the empty pVAX vector, pGag, pRhPD1-Gag or pRhPD1-p27 via intramuscular injection with 3 consecutive electroporation in 3-week intervals. Immunological analysis was performed at 2 weeks or 6 months after the last immunization. (B)—(E) Acute humoral and cellular immunity induced by sPD1-based vaccines 2 weeks post-last immunization. (B) Anti-Gag antibody titers in sera. (C) Gag-specific cellular immune responses in spleens evaluated by IFN-γ ELISpot assays. (D) Representative FACS plots showing the presence of H-2Db/AL11-specific CD8+ T cells in the spleens of vaccinated mice by tetramer staining analysis. (E) Frequencies of H-2Db/AL11-specific CD8+ T cells in vaccinated mice. (F) The efficacy of the sPD1-based vaccines against recombinant vaccinia virus challenge. Two weeks after last immunization, mice were challenged with recombinant vaccinia virus MVTT-SIVgpe via intranasal route. Viral loads in the lungs were determined two days after challenge. (G)—(I) Memory humoral and cellular immune responses elicited by the sPD1-based vaccines at 6 months post last immunization. (G) Anti-Gag IgG titers in sera. (H) Gag-specific IFN-γ ELISpot responses and (I) the frequencies of H-2Db/AL11-specific CD8+ T cells in spleens.
Fig 2. Viral suppression in rhesus macaques…
Fig 2. Viral suppression in rhesus macaques immunised with the PD1-based pRhPD1-p27 DNA vaccine after high dose pathogenic SHIVSF162P3CN challenge.
(A) Schematic of DNA vaccination in Chinese-origin rhesus macaques. Two studies with different vaccination durations were conducted. In both studies, Chinese-origin rhesus macaques were immunised with the PD1-based pRhPD1-p27 DNA vaccine via intramuscular injection with electroporation 4 times in 6- to 13-week intervals, followed by intravenous challenge with 5000 TCID50 of SHIVSF162P3CN at 23 or 36 weeks after last immunization. Unvaccinated animals were included as controls. (B) Plasma viral loads of the vaccinated (top) and unvaccinated macaques (below) after SHIV challenge. Animals that were euthanised were marked with †. Anti-CD8β antibody (clone CD8b255R1) was infused into the vaccinated macaques of Group A intravenously at 17 weeks post-challenge, after confirming pVL were below detection levels. Data of this treatment is presented in Fig 5D. (C) Comparison of the peak viremia and (D) geometric means of setpoint viremia determined from 6–20 weeks post-infection (wpi). (E) Proviral DNA loads in PBMC in vaccinated and unvaccinated macaques. Geometric means ± geometric SD are shown in (C)–(E). Significances of differences were determined by the Kruskal-Wallis test followed by the Dunn’s multiple comparisons test (C), or the two-tailed Wilcoxon rank sum test (D and E). (F) The CD4+/CD8+ T cell ratios in vaccinated rhesus macaques from Group A and B (left) or unvaccinated macaques (right) after SHIVSF162P3CN challenge. (G) Changes of body weight of the vaccinated (left) and unvaccinated macaques (right). Animals that were euthanised over the measurement period were marked with †. (H) Survival of vaccinated and unvaccinated macaques after SHIV challenge. The significance of difference was determined by log-rank test.
Fig 3. T cell immunogenicity of the…
Fig 3. T cell immunogenicity of the pRhPD1-p27 vaccine in rhesus macaques.
(A) Kinetics of p27-specific cellular immune responses in peripheral blood measured by IFN-γ ELISpot. (B) Comparison of ELISpot-detected p27-specific T cell responses induced in Groups A and B by the pRhPD1-p27 vaccine with historical data of Gag-specific T cell responses induced by MVTTSIVgpe/AD5SIVgpe vaccination in Chinese-origin rhesus macaques 12. (C) Representative FACS plots demonstrating the production of IFN-γ, TNF-α and IL-2 by CD8+ (left) and CD4+ T cells (right) at 2 weeks post last immunization, as detected by ICS assays after ex vivo stimulation with the overlapping SIV Gag-p27 peptide pool. (D) Kinetics of p27-specific CD8+ (top) and CD4+ T cells (bottom) in PBMCs after last immunization, as detected by ICS assays. (E) Poly-functionality analysis of p27-specific CD8+ (left) and CD4+ T cell responses (right) at 2 weeks after last immunisation. (F) Poly-functionality analysis of CD8+ (left) and CD4+ T cell responses (right) during memory phase at 22 weeks after last immunisation. For (E) and (F), means ± SEM are shown. (G) Phenotypes of p27-specific CD8+ and (H) CD4+ T cells induced by pRhPD1-p27 vaccination in macaques. PBMC isolated at 8 weeks post last immunisation were stimulated with the overlapping Gag-p27 peptide pool, followed by surface and ICS analysis. Representative FACS plots (left) show the expression of CD95, CD28, and CCR7 on the overall T cell populations (grey) and IFN-γ-producing T cells (red). Frequencies of TEM (CD95+ CD28+or—CCR7-) were compared between the overall and IFN-γ-producing T cells (right). Statistical significance was determined by the Wilcoxon matched-pairs signed rank test.
Fig 4. Specificities of the T cell…
Fig 4. Specificities of the T cell responses induced by the pRhPD1-p27 vaccine.
Representative results of T cell epitope mapping based on the IFN-γ ELISpot and ICS assays on macaque A04 from Group A. (A) PBMC isolated after third/fourth vaccination were stimulated with individual overlapping 15mer peptides spanning the Gag-p27 antigen in IFN-γ ELISpot assays. The dotted line represents the cut-off value. (B) Peptides that successfully induced responses above cut-off values (2x medium alone values) in IFN-γ ELISpot assays were then tested for their ability to induce IFN-γ production in T cells by ICS assays. (C) Epitope mapping analysis of the pRhPD1-p27-vaccinated macaques. CD8+ and CD4+ T cell epitopes are represented by orange and green boxes respectively. (D) Representative ICS FACS plots showing IFN-γ expression in CD8+ (upper) and CD4+ T cells (lower) from pRhPD1-p27-vaccinated or unvaccinated macaques 4 weeks after SHIVSF162P3CN challenge, after ex vivo stimulation with SIV p27 or Nef peptide pools. (E) Frequencies of CD8+ and (F) CD4+ T cells specific to Gag-p27 and Nef antigens at 4 or 17 weeks after SHIVSF162P3CN challenge, as determined by ICS analysis. Means ± SEM are shown. Significance of difference was determined by the two-tailed Wilcoxon rank sum test.
Fig 5. Reactivity of T cell epitopes…
Fig 5. Reactivity of T cell epitopes induced by the pRhPD1-p27 vaccine during SHIVSF162P3CN challenge.
(A) T cell responses against the PD1-based vaccine-induced CD8+ and (B) CD4+ T cell epitopes at 17 weeks after SHIVSF162P3CN challenge. (C) T cell epitope profiles on Gag-p27 of vaccinated macaques in Group B after SHIVSF162P3CN challenge. (D) Long-term pVL monitoring in the vaccinated/challenged macaques from Group A. Anti-CD8β antibody (clone CD8b255R1) was infused into these vaccinated macaques intravenously at 17 weeks post-challenge, after confirming pVL had reduced to undetectable levels. At 103 weeks post-SHIVSF162P3CN challenge, these macaques also received a boost immunisation of the pRhPD1-p27 vaccine via i.m./EP. (E) Frequencies of CD8+ and (F) CD4+ T cells specific to Gag-p27 and Nef antigens 3 weeks after pRhPD1-p27 re-immunization. Means ± SEM are shown.
Fig 6. Single-cell RNA sequencing analysis of…
Fig 6. Single-cell RNA sequencing analysis of CD8+ T cells induced by pRhPD1-p27 immunisation.
CD8+ T cell from macaques B01, B02, and B03 in Study B were purified at before, 4 weeks post, and 12 weeks post last immunisation followed by scRNAseq using the 10X scRNAseq platform. (A) UMAP clustering analysis revealed 8 major CD8+ T cell clusters. (B) Expression of different marker genes of the indicated CD8+ T cell clusters. (C) Fractions of different CD8+ T cell clusters at different time points after vaccination (top) and trajectory inference (below) showing the dynamic of cell progression after receiving vaccination of individual macaques. (D) GO analysis of differentially expressed genes of different pathways related to biological process. GeneRatio represents the ratio of the number of genes related to the GO term to the total number of significant genes.

References

    1. Katlama C, Deeks SG, Autran B, Martinez-Picado J, van Lunzen J, Rouzioux C, et al.. Barriers to a cure for HIV: new ways to target and eradicate HIV-1 reservoirs. The Lancet. 2013;381(9883):2109–17. doi: 10.1016/S0140-6736(13)60104-X
    1. Sáez-Cirión A, Lacabaratz C, Lambotte O, Versmisse P, Urrutia A, Boufassa F, et al.. HIV controllers exhibit potent CD8 T cell capacity to suppress HIV infection ex vivo and peculiar cytotoxic T lymphocyte activation phenotype. Proc Natl Acad Sci U S A. 2007;104(16):6776–81. doi: 10.1073/pnas.0611244104 Proceedings of the National Academy of Sciences.
    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: 10.1182/blood-2005-12-4818
    1. Koup RA, Safrit JT, Cao Y, Andrews CA, McLeod 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. Journal of Virology. 1994;68(7):4650–5. doi: 10.1128/JVI.68.7.4650-4655.1994
    1. Borrow P, Lewicki H, Hahn BH, Shaw GM, Oldstone MB. Virus-specific CD8+ cytotoxic T-lymphocyte activity associated with control of viremia in primary human immunodeficiency virus type 1 infection. Journal of Virology. 1994;68(9):6103–10. doi: 10.1128/JVI.68.9.6103-6110.1994
    1. Jin X, Bauer DE, Tuttleton SE, Lewin S, Gettie A, Blanchard J, et al.. Dramatic Rise in Plasma Viremia after CD8+ T Cell Depletion in Simian Immunodeficiency Virus–infected Macaques. The Journal of Experimental Medicine. 1999;189(6):991–8. doi: 10.1084/jem.189.6.991
    1. Schmitz JE, Kuroda MJ, Santra S, Sasseville VG, Simon MA, Lifton MA, et al.. Control of Viremia in Simian Immunodeficiency Virus Infection by CD8+ Lymphocytes. Science. 1999;283(5403):857–60. doi: 10.1126/science.283.5403.857 Science.
    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;15:293–9. doi: 10.1038/nm.1935
    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. doi: 10.1038/nature10003
    1. Barouch DH, Liu J, Li H, Maxfield LF, Abbink P, Lynch DM, et al.. Vaccine protection against acquisition of neutralization-resistant SIV challenges in rhesus monkeys. Nature. 2012;482:89–93. doi: 10.1038/nature10766
    1. Hansen SG, Jr MP, Ventura AB, Hughes CM, Gilbride RM, Ford JC, et al.. Immune clearance of highly pathogenic SIV infection. Nature. 2013;502:100–4. doi: 10.1038/nature12519
    1. Sun C, Chen Z, Tang X, Zhang Y, Feng L, Du Y, et al.. Mucosal priming with a replicating-vaccinia virus-based vaccine elicits protective immunity to simian immunodeficiency virus challenge in rhesus monkeys. Journal of Virology. 2013;87:5669–77. doi: 10.1128/JVI.03247-12
    1. Chen Z, Huang Y, Zhao X, Ba L, Zhang W, Ho DD. Design, Construction, and Characterization of a Multigenic Modified Vaccinia Ankara Candidate Vaccine Against Human Immunodeficiency Virus Type 1 Subtype C/B′. JAIDS Journal of Acquired Immune Deficiency Syndromes. 2008;47(4):412–21. doi: 10.1097/QAI.0b013e3181651bb2 00126334-200804010-00002.
    1. Liu J, O’Brien KL, Lynch DM, Simmons NL, La Porte A, Riggs AM, et al.. Immune control of an SIV challenge by a T-cell-based vaccine in rhesus monkeys. Nature. 2008;457:87–91. doi: 10.1038/nature07469
    1. Fischer W, Perkins S, Theiler J, Bhattacharya T, Yusim K, Funkhouser R, et al.. Polyvalent vaccines for optimal coverage of potential T-cell epitopes in global HIV-1 variants. Nature Medicine. 2007;13(1):100–6. doi: 10.1038/nm1461 .
    1. Barouch DH, O’Brien KL, Simmons NL, King SL, Abbink P, Maxfield LF, et al.. Mosaic HIV-1 vaccines expand the breadth and depth of cellular immune responses in rhesus monkeys. Nature Medicine. 2010;16:319–23. doi: 10.1038/nm.2089
    1. Santra S, Liao HX, Zhang R, Muldoon M, Watson S, Fischer W, et al.. Mosaic vaccines elicit CD8+ T lymphocyte responses that confer enhanced immune coverage of diverse HIV strains in monkeys. Nature Medicine. 2010;16(3):324–8. Epub 2010/02/23. doi: 10.1038/nm.2108 ; PubMed Central PMCID: PMC2834806.
    1. Barouch DH, Stephenson KE, Borducchi EN, Smith K, Stanley K, McNally AG, et al.. Protective efficacy of a global HIV-1 mosaic vaccine against heterologous SHIV challenges in rhesus monkeys. Cell. 2013;155(3):531–9. Epub 2013/11/19. doi: 10.1016/j.cell.2013.09.061 ; PubMed Central PMCID: PMC3846288.
    1. Barouch DH, Tomaka FL, Wegmann F, Stieh DJ, Alter G, Robb ML, et al.. Evaluation of a mosaic HIV-1 vaccine in a multicentre, randomised, double-blind, placebo-controlled, phase 1/2a clinical trial (APPROACH) and in rhesus monkeys (NHP 13–19). The Lancet. 2018;392(10143):232–43.
    1. Flynn BJ, Kastenmüller K, Wille-Reece U, Tomaras GD, Alam M, Lindsay RW, et al.. Immunization with HIV Gag targeted to dendritic cells followed by recombinant New York vaccinia virus induces robust T-cell immunity in nonhuman primates. Proc Natl Acad Sci U S A. 2011;108(17):7131–6. doi: 10.1073/pnas.1103869108 Proceedings of the National Academy of Sciences.
    1. Flamar A-L, Bonnabau H, Zurawski S, Lacabaratz C, Montes M, Richert L, et al.. HIV-1 T cell epitopes targeted to Rhesus macaque CD40 and DCIR: A comparative study of prototype dendritic cell targeting therapeutic vaccine candidates. PLOS ONE. 2018;13(11):e0207794. doi: 10.1371/journal.pone.0207794
    1. Zurawski G, Shen X, Zurawski S, Tomaras GD, Montefiori DC, Roederer M, et al.. Superiority in Rhesus Macaques of Targeting HIV-1 Env gp140 to CD40 versus LOX-1 in Combination with Replication-Competent NYVAC-KC for Induction of Env-Specific Antibody and T Cell Responses. Journal of Virology. 2017;91(9):e01596–16. doi: 10.1128/JVI.01596-16 Journal of Virology.
    1. Nchinda G, Kuroiwa J, Oks M, Trumpfheller C, Park CG, Huang Y, et al.. The efficacy of DNA vaccination is enhanced in mice by targeting the encoded protein to dendritic cells. J Clin Invest. 2008;118(4):1427–36. Epub 2008/03/08. doi: 10.1172/JCI34224 ; PubMed Central PMCID: PMC2263146.
    1. Zhou J, Cheung AK, Tan Z, Wang H, Yu W, Du Y, et al.. PD1-based DNA vaccine amplifies HIV-1 GAG-specific CD8+ T cells in mice. The Journal of clinical investigation. 2013;123(6):2629–42. Epub 2013/05/03. doi: 10.1172/JCI64704 ; PubMed Central PMCID: PMC3668817.
    1. Tan Z, Zhou J, Cheung AKL, Yu Z, Cheung K-W, Liang J, et al.. Vaccine-elicited CD8+ T cells cure mesothelioma by overcoming tumor-induced immunosuppressive environment. Cancer Research. 2014;74:6010–21. doi: 10.1158/0008-5472.CAN-14-0473
    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;13:46–53. doi: 10.1038/nm1520
    1. Zuñiga R, Lucchetti A, Galvan P, Sanchez S, Sanchez C, Hernandez A, et al.. Relative Dominance of Gag p24-Specific Cytotoxic T Lymphocytes Is Associated with Human Immunodeficiency Virus Control. Journal of Virology. 2006;80(6):3122–5. doi: 10.1128/JVI.80.6.3122-3125.2006 Journal of Virology.
    1. Harouse JM, Gettie A, Eshetu T, Tan RCH, Bohm R, Blanchard J, et al.. Mucosal Transmission and Induction of Simian AIDS by CCR5-Specific Simian/Human Immunodeficiency Virus SHIVSF162P3. Journal of Virology. 2001;75(4):1990–5. doi: 10.1128/JVI.75.4.1990-1995.2001 Journal of Virology.
    1. Harouse JM, Gettie A, Tan RCH, Blanchard J, Cheng-Mayer C. Distinct Pathogenic Sequela in Rhesus Macaques Infected with CCR5 or CXCR4 Utilizing SHIVs. Science. 1999;284(5415):816–9. doi: 10.1126/science.284.5415.816
    1. Chen Z. Monkey Models and HIV Vaccine Research. Adv Exp Med Biol. 2018;1075:97–124. Epub 2018/07/22. doi: 10.1007/978-981-13-0484-2_5 .
    1. Picker LJ, Reed-Inderbitzin EF, Hagen SI, Edgar JB, Hansen SG, Legasse A, et al.. IL-15 induces CD4+ effector memory T cell production and tissue emigration in nonhuman primates. The Journal of clinical investigation. 2006;116(6):1514–24. doi: 10.1172/JCI27564
    1. Mahnke YD, Brodie TM, Sallusto F, Roederer M, Lugli E. The who’s who of T-cell differentiation: Human memory T-cell subsets. European Journal of Immunology. 2013;43(11):2797–809. doi: 10.1002/eji.201343751
    1. Geldmacher C, Currier JR, Herrmann E, Haule A, Kuta E, McCutchan F, et al.. CD8 T-Cell Recognition of Multiple Epitopes within Specific Gag Regions Is Associated with Maintenance of a Low Steady-State Viremia in Human Immunodeficiency Virus Type 1-Seropositive Patients. Journal of Virology. 2007;81(5):2440–8. doi: 10.1128/JVI.01847-06 Journal of Virology.
    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;351:714–20. doi: 10.1126/science.aac9475
    1. Mühl T, Krawczak M, ten Haaft P, Hunsmann G, Sauermann U. MHC Class I Alleles Influence Set-Point Viral Load and Survival Time in Simian Immunodeficiency Virus-Infected Rhesus Monkeys. Journal of Immunology. 2002;169(6):3438–46. doi: 10.4049/jimmunol.169.6.3438 The Journal of Immunology.
    1. Loffredo JT, Maxwell J, Qi Y, Glidden CE, Borchardt GJ, Soma T, et al.. Mamu-B*08-Positive Macaques Control Simian Immunodeficiency Virus Replication. Journal of Virology. 2007;81(16):8827–32. doi: 10.1128/JVI.00895-07
    1. Cao Y, Wang X, Peng G. SCSA: A Cell Type Annotation Tool for Single-Cell RNA-seq Data. Front Genet. 2020;11:490. Epub 2020/06/02. doi: 10.3389/fgene.2020.00490 ; PubMed Central PMCID: PMC7235421.
    1. Zhang X, Lan Y, Xu J, Quan F, Zhao E, Deng C, et al.. CellMarker: a manually curated resource of cell markers in human and mouse. Nucleic Acids Res. 2019;47(D1):D721–D8. Epub 2018/10/06. doi: 10.1093/nar/gky900 ; PubMed Central PMCID: PMC6323899.
    1. Vasan S, Schlesinger SJ, Huang Y, Hurley A, Lombardo A, Chen Z, et al.. Phase 1 Safety and Immunogenicity Evaluation of ADVAX, a Multigenic, DNA-Based Clade C/B’ HIV-1 Candidate Vaccine. PLOS ONE. 2010;5(1):e8617. doi: 10.1371/journal.pone.0008617
    1. Lambricht L, Lopes A, Kos S, Sersa G, Préat V, Vandermeulen G. Clinical potential of electroporation for gene therapy and DNA vaccine delivery. Expert Opinion on Drug Delivery. 2016;13(2):295–310. doi: 10.1517/17425247.2016.1121990
    1. Tewari K, Flynn BJ, Boscardin SB, Kastenmueller K, Salazar AM, Anderson CA, et al.. Poly(I:C) is an effective adjuvant for antibody and multi-functional CD4+ T cell responses to Plasmodium falciparum circumsporozoite protein (CSP) and αDEC-CSP in non human primates. Vaccine. 2010;28(45):7256–66. doi: 10.1016/j.vaccine.2010.08.098
    1. Tenbusch M, Ignatius R, Nchinda G, Trumpfheller C, Salazar AM, Töpfer K, et al.. Immunogenicity of DNA Vaccines Encoding Simian Immunodeficiency Virus Antigen Targeted to Dendritic Cells in Rhesus Macaques. PLOS ONE. 2012;7(6):e39038. doi: 10.1371/journal.pone.0039038
    1. Tenbusch M, Ignatius R, Temchura V, Nabi G, Tippler B, Stewart-Jones G, et al.. Risk of Immunodeficiency Virus Infection May Increase with Vaccine-Induced Immune Response. Journal of Virology. 2012;86(19):10533–9. doi: 10.1128/JVI.00796-12 Journal of Virology.
    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;340:1237874. doi: 10.1126/science.1237874
    1. Mudd PA, Martins MA, Ericsen AJ, Tully DC, Power KA, Bean AT, et al.. Vaccine-induced CD8+ T cells control AIDS virus replication. Nature. 2012;491:129–33. doi: 10.1038/nature11443
    1. Barouch DH, Santra S, Schmitz JE, Kuroda MJ, Fu T-M, Wagner W, et al.. Control of Viremia and Prevention of Clinical AIDS in Rhesus Monkeys by Cytokine-Augmented DNA Vaccination. Science. 2000;290(5491):486–92. doi: 10.1126/science.290.5491.486 Science.
    1. Yant LJ, Friedrich TC, Johnson RC, May GE, Maness NJ, Enz AM, et al.. The High-Frequency Major Histocompatibility Complex Class I Allele Mamu-B*17 Is Associated with Control of Simian Immunodeficiency Virus SIVmac239 Replication. Journal of Virology. 2006;80(10):5074–7. doi: 10.1128/JVI.80.10.5074-5077.2006 Journal of Virology.
    1. Wojcechowskyj JA, Yant LJ, Wiseman RW, O’Connor SL, O’Connor DH. Control of Simian Immunodeficiency Virus SIVmac239 Is Not Predicted by Inheritance of Mamu-B*17-Containing Haplotypes. Journal of Virology. 2007;81(1):406–10. doi: 10.1128/JVI.01636-06 Journal of Virology.
    1. Maness NJ, Yant LJ, Chung C, Loffredo JT, Friedrich TC, Piaskowski SM, et al.. Comprehensive Immunological Evaluation Reveals Surprisingly Few Differences between Elite Controller and Progressor Mamu-B*17-Positive Simian Immunodeficiency Virus-Infected Rhesus Macaques. Journal of Virology. 2008;82(11):5245–54. doi: 10.1128/JVI.00292-08 Journal of Virology.
    1. Karimi M, Cao TM, Baker JA, Verneris MR, Soares L, Negrin RS. Silencing human NKG2D, DAP10, and DAP12 reduces cytotoxicity of activated CD8+ T cells and NK cells. J Immunol. 2005;175(12):7819–28. Epub 2005/12/13. doi: 10.4049/jimmunol.175.12.7819 .
    1. Pena SV, Hanson DA, Carr BA, Goralski TJ, Krensky AM. Processing, subcellular localization, and function of 519 (granulysin), a human late T cell activation molecule with homology to small, lytic, granule proteins. J Immunol. 1997;158(6):2680–8. Epub 1997/03/15. .
    1. Voskoboinik I, Whisstock JC, Trapani JA. Perforin and granzymes: function, dysfunction and human pathology. Nat Rev Immunol. 2015;15(6):388–400. Epub 2015/05/23. doi: 10.1038/nri3839 .
    1. Patel NP, Vukmanovic-Stejic M, Suarez- Farinas M, Chambers ES, Sandhu D, Fuentes-Duculan J, et al.. Impact of Zostavax Vaccination on T-Cell Accumulation and Cutaneous Gene Expression in the Skin of Older Humans After Varicella Zoster Virus Antigen–Specific Challenge. The Journal of Infectious Diseases. 2018;218(suppl_2):S88–S98. doi: 10.1093/infdis/jiy420
    1. Minervina AA, Pogorelyy MV, Komech EA, Karnaukhov VK, Bacher P, Rosati E, et al.. Primary and secondary anti-viral response captured by the dynamics and phenotype of individual T cell clones. eLife. 2020;9:e53704. doi: 10.7554/eLife.53704
    1. Wiseman RW, Karl JA, Bohn PS, Nimityongskul FA, Starrett GJ, O’Connor DH. Haplessly hoping: macaque major histocompatibility complex made easy. ILAR journal. 2013;54(2):196–210. doi: 10.1093/ilar/ilt036 .
    1. McWilliam H, Robinson J, Halliwell JA, Lopez R, Marsh SGE. IPD—the Immuno Polymorphism Database. Nucleic Acids Research. 2012;41(D1):D1234–D40. doi: 10.1093/nar/gks1140%J Nucleic Acids Research.
    1. Montefiori DC. Evaluating Neutralizing Antibodies Against HIV, SIV, and SHIV in Luciferase Reporter Gene Assays. Current Protocols in Immunology. 2004;64(1):12.1.1–.1.7. doi: 10.1002/0471142735.im1211s64
    1. Lu X, Liu L, Zhang X, Lau TCK, Tsui SKW, Kang Y, et al.. F18, a Novel Small-Molecule Nonnucleoside Reverse Transcriptase Inhibitor, Inhibits HIV-1 Replication Using Distinct Binding Motifs as Demonstrated by Resistance Selection and Docking Analysis. Antimicrobial Agents and Chemotherapy. 2012;56(1):341–51. doi: 10.1128/AAC.05537-11 Antimicrobial Agents and Chemotherapy.
    1. Nichole Cline A, Bess JW, Piatak M Jr, Lifson JD. Highly sensitive SIV plasma viral load assay: practical considerations, realistic performance expectations, and application to reverse engineering of vaccines for AIDS. Journal of Medical Primatology. 2005;34(5–6):303–12. doi: 10.1111/j.1600-0684.2005.00128.x
    1. Chung H-k, Unangst T, Treece J, Weiss D, Markham P. Development of real-time PCR assays for quantitation of simian betaretrovirus serotype-1, -2, -3, and -5 viral DNA in Asian monkeys. Journal of Virological Methods. 2008;152(1):91–7.
    1. Liu W, Wong YC, Chen SMY, Tang J, Wang H, Cheung AKL, et al.. DNA prime/MVTT boost regimen with HIV-1 mosaic Gag enhances the potency of antigen-specific immune responses. Vaccine. 2018;36(31):4621–32. doi: 10.1016/j.vaccine.2018.06.047
    1. Satija R, Farrell JA, Gennert D, Schier AF, Regev A. Spatial reconstruction of single-cell gene expression data. Nat Biotechnol. 2015;33(5):495–502. Epub 2015/04/14. doi: 10.1038/nbt.3192 ; PubMed Central PMCID: PMC4430369.
    1. McGinnis CS, Murrow LM, Gartner ZJ. DoubletFinder: Doublet Detection in Single-Cell RNA Sequencing Data Using Artificial Nearest Neighbors. Cell Syst. 2019;8(4):329–37 e4. Epub 2019/04/08. doi: 10.1016/j.cels.2019.03.003 ; PubMed Central PMCID: PMC6853612.
    1. Haghverdi L, Lun ATL, Morgan MD, Marioni JC. Batch effects in single-cell RNA-sequencing data are corrected by matching mutual nearest neighbors. Nat Biotechnol. 2018;36(5):421–7. Epub 2018/04/03. doi: 10.1038/nbt.4091 ; PubMed Central PMCID: PMC6152897.
    1. Yu G, Wang LG, Han Y, He QY. clusterProfiler: an R package for comparing biological themes among gene clusters. OMICS. 2012;16(5):284–7. Epub 2012/03/30. doi: 10.1089/omi.2011.0118 ; PubMed Central PMCID: PMC3339379.

Source: PubMed

3
購読する