Preferential amplification of CD8 effector-T cells after transcutaneous application of an inactivated influenza vaccine: a randomized phase I trial

Behazine Combadière, Annika Vogt, Brice Mahé, Dominique Costagliola, Sabrina Hadam, Olivia Bonduelle, Wolfram Sterry, Shlomo Staszewski, Hans Schaefer, Sylvie van der Werf, Christine Katlama, Brigitte Autran, Ulrike Blume-Peytavi, Behazine Combadière, Annika Vogt, Brice Mahé, Dominique Costagliola, Sabrina Hadam, Olivia Bonduelle, Wolfram Sterry, Shlomo Staszewski, Hans Schaefer, Sylvie van der Werf, Christine Katlama, Brigitte Autran, Ulrike Blume-Peytavi

Abstract

Background: Current conventional vaccination approaches do not induce potent CD8 T-cell responses for fighting mostly variable viral diseases such as influenza, avian influenza viruses or HIV. Following our recent study on vaccine penetration by targeting of vaccine to human hair follicular ducts surrounded by Langerhans cells, we tested in the first randomized Phase-Ia trial based on hair follicle penetration (namely transcutaneous route) the induction of virus-specific CD8 T cell responses.

Methods and findings: We chose the inactivated influenza vaccine - a conventional licensed tetanus/influenza (TETAGRIP) vaccine - to compare the safety and immunogenicity of transcutaneous (TC) versus IM immunization in two randomized controlled, multi-center Phase I trials including 24 healthy-volunteers and 12 HIV-infected patients. Vaccination was performed by application of inactivated influenza vaccine according to a standard protocol allowing the opening of the hair duct for the TC route or needle-injection for the IM route. We demonstrated that the safety of the two routes was similar. We showed the superiority of TC application, but not the IM route, to induce a significant increase in influenza-specific CD8 cytokine-producing cells in healthy-volunteers and in HIV-infected patients. However, these routes did not differ significantly for the induction of influenza-specific CD4 responses, and neutralizing antibodies were induced only by the IM route. The CD8 cell response is thus the major immune response observed after TC vaccination.

Conclusions: This Phase Ia clinical trial (Manon05) testing an anti-influenza vaccine demonstrated that vaccines designed for antibody induction by the IM route, generate vaccine-specific CD8 T cells when administered transcutaneously. These results underline the necessity of adapting vaccination strategies to control complex infectious diseases when CD8 cellular responses are crucial. Our work opens up a key area for the development of preventive and therapeutic vaccines for diseases in which CD8 cells play a crucial role.

Trial registration: Clinicaltrials.gov NCT00261001.

Conflict of interest statement

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

Figures

Figure 1. Differential induction of CD4 and…
Figure 1. Differential induction of CD4 and CD8 T cell responses after TC vaccine application compared to IM immunization in healthy individuals.
Intracytoplasmic cytokine staining (ICS) of influenza-specific effector CD4 and CD8 responses was performed on frozen PBMC samples from vaccinated individuals: 10/12 from the TC group and 7–9/12 from IM group with 90% cell viability after thawing. Three million cells were stimulated with the overlapping peptide covering H3, H1, and NP for 12 hours at 37°C. Brefeldin A was added 4 h before harvesting. ICS was performed by flow-cytometric assays on CD3+CD4+ (left panels) and CD3+CD8+ T cells (Right panels). At least 1,000,000 live events according to forward and side scatter parameters were accumulated and analyzed (M&M section). The expression of IFN-γ, TNF-α, and/or IL-2 (triple+double+single cytokine positive cells) by influenza-specific T cells was analyzed with the Boolean gating function of FlowJo software. Results are shown as percentages of cytokine-producing T cells (Δ Day 28- Day 0) after subtracting the unstimulated cell background. Mann-Whitney test was used to compare continuous variables between the groups. Significance was set at p0. The χ2 test was used to define categorical variables between TC and IM groups.
Figure 2. Flow cytometric representation of influenza-specific…
Figure 2. Flow cytometric representation of influenza-specific T cell response at day 28 post-vaccination by TC and IM routes.
A, B) Representative flow cytometric analysis of cytokine-producing influenza-specific effector CD4 and CD8 responses. Experiments were performed on frozen PBMCs from individuals vaccinated by TC and IM routes as described in figure 1. Results are shown for a representative healthy individual with a TC route (A) and an IM route (B) vaccination at day 28. C, D) Pie chart analyses of single (white), double (gray) and triple (black)-cytokine positive cells for CD4 (C) and CD8 (D) effector cells specific for the indicated influenza protein. The expression of IFN-γ, TNF-α, and/or IL-2 (triple+double+single cytokine positive cells) by influenza-specific T cells was analyzed with the Boolean gating function of FlowJo software. NA: not applicable.
Figure 3. Differential induction of CD4 and…
Figure 3. Differential induction of CD4 and CD8 T cell responses after TC and IM vaccine application in HIV-infected volunteers.
Intracytoplasmic cytokine staining (ICS) of influenza-specific effector CD4 and CD8 responses was performed on frozen PBMCs from vaccinated individuals. Experiments were performed on 12 HIV-infected individuals for whom cell sample viability was at least 90% after thawing. Three million cells were stimulated with the overlapping peptide covering H3, H1, and NP for 12 hours at 37°C. Brefeldin A (5 µg/ml) was added 4 h before harvesting. ICS was performed for IFN-γ by flow-cytometric assay on CD3+CD4+ (left panels) and CD3+CD8+ T cells (Right panels). At least 1,000,000 live events according to forward and side scatter parameters were accumulated and analyzed (M&M section). The expression of IFN-γ, TNF-α, and/or IL-2 (triple+double+single cytokine positive cells) by influenza-specific T cells was analyzed with the Boolean gating function of FlowJo software. Results are shown as percentages of cytokine-producing T cells (Δ Day 28-Day0) after subtracting the unstimulated cell background. Mann-Whitney tests were used to compare continuous variables between the groups. Significance was set at p

References

    1. Wharton M, Strikas RA, Harpaz R, Rotz LD, Schwartz B, et al. Recommendations for using smallpox vaccine in a pre-event vaccination program. Supplemental recommendations of the Advisory Committee on Immunization Practices (ACIP) and the Healthcare Infection Control Practices Advisory Committee (HICPAC). MMWR Recomm Rep. 2003;52:1–16.
    1. Patterson S, Papagatsias T, Benlahrech A. Use of adenovirus in vaccines for HIV. Handb Exp Pharmacol. 2009:275–293.
    1. Harro CD, Robertson MN, Lally MA, O'Neill LD, Edupuganti S, et al. Safety and immunogenicity of adenovirus-vectored near-consensus HIV type 1 clade B gag vaccines in healthy adults. AIDS Res Hum Retroviruses. 2009;25:103–114.
    1. White A. Why vaccines are not the answer - the failure of V520 and the importance of cell-mediated immunity in the fight against HIV. Med Hypotheses. 2008;71:909–913.
    1. Doherty PC, Hou S, Tripp RA. CD8+ T-cell memory to viruses. Curr Opin Immunol. 1994;6:545–552.
    1. Kaech SM, Wherry EJ, Ahmed R. Effector and memory T-cell differentiation: implications for vaccine development. Nat Rev Immunol. 2002;2:251–262.
    1. Robinson HL. T cells versus HIV-1: fighting exhaustion as well as escape. Nat Immunol. 2003;4:12–13.
    1. Post J, Ratnarajah S, Lloyd AR. Immunological determinants of the outcomes from primary hepatitis C infection. Cell Mol Life Sci. 2009;66:733–756.
    1. Brown LE, Kelso A. Prospects for an influenza vaccine that induces cross-protective cytotoxic T lymphocytes. Immunol Cell Biol 2009
    1. Seder RA, Darrah PA, Roederer M. T-cell quality in memory and protection: implications for vaccine design. Nat Rev Immunol. 2008;8:247–258.
    1. Miller JD, van der Most RG, Akondy RS, Glidewell JT, Albott S, et al. Human effector and memory CD8+ T cell responses to smallpox and yellow fever vaccines. Immunity. 2008;28:710–722.
    1. Godelaine D, Carrasco J, Lucas S, Karanikas V, Schuler-Thurner B, et al. Polyclonal CTL responses observed in melanoma patients vaccinated with dendritic cells pulsed with a MAGE-3.A1 peptide. J Immunol. 2003;171:4893–4897.
    1. Boon AC, de Mutsert G, Fouchier RA, Osterhaus AD, Rimmelzwaan GF. Functional profile of human influenza virus-specific cytotoxic T lymphocyte activity is influenced by interleukin-2 concentration and epitope specificity. Clin Exp Immunol. 2005;142:45–52.
    1. Boon AC, de Mutsert G, van Baarle D, Smith DJ, Lapedes AS, et al. Recognition of homo- and heterosubtypic variants of influenza A viruses by human CD8+ T lymphocytes. J Immunol. 2004;172:2453–2460.
    1. Germeau C, Ma W, Schiavetti F, Lurquin C, Henry E, et al. High frequency of antitumor T cells in the blood of melanoma patients before and after vaccination with tumor antigens. J Exp Med. 2005;201:241–248.
    1. Lau LL, Jamieson BD, Somasundaram T, Ahmed R. Cytotoxic T-cell memory without antigen. Nature. 1994;369:648–652.
    1. Hammarlund E, Lewis MW, Hansen SG, Strelow LI, Nelson JA, et al. Duration of antiviral immunity after smallpox vaccination. Nat Med 2003
    1. Combadiere B, Boissonnas A, Carcelain G, Lefranc E, Samri A, et al. Distinct time effects of vaccination on long-term proliferative and IFN-gamma-producing T cell memory to smallpox in humans. J Exp Med. 2004;199:1585–1593.
    1. Demkowicz WE, Jr, Littaua RA, Wang J, Ennis FA. Human cytotoxic T-cell memory: long-lived responses to vaccinia virus. J Virol. 1996;70:2627–2631.
    1. Banchereau J, Palucka AK. Dendritic cells as therapeutic vaccines against cancer. Nat Rev Immunol. 2005;5:296–306.
    1. Banchereau J, Steinman RM. Dendritic cells and the control of immunity. Nature. 1998;392:245–252.
    1. Kupper TS, Fuhlbrigge RC. Immune surveillance in the skin: mechanisms and clinical consequences. Nat Rev Immunol. 2004;4:211–222.
    1. Klechevsky E, Morita R, Liu M, Cao Y, Coquery S, et al. Functional specializations of human epidermal Langerhans cells and CD14+ dermal dendritic cells. Immunity. 2008;29:497–510.
    1. Combadiere B, Mahe B. Particle-based vaccines for transcutaneous vaccination. Comp Immunol Microbiol Infect Dis. 2008;31:293–315.
    1. Belyakov IM, Hammond SA, Ahlers JD, Glenn GM, Berzofsky JA. Transcutaneous immunization induces mucosal CTLs and protective immunity by migration of primed skin dendritic cells. J Clin Invest. 2004;113:998–1007.
    1. Berzofsky JA, Ahlers JD, Belyakov IM. Strategies for designing and optimizing new generation vaccines. Nat Rev Immunol. 2001;1:209–219.
    1. Le Borgne M, Etchart N, Goubier A, Lira SA, Sirard JC, et al. Dendritic cells rapidly recruited into epithelial tissues via CCR6/CCL20 are responsible for CD8+ T cell crosspriming in vivo. Immunity. 2006;24:191–201.
    1. Bos JD, Kapsenberg ML. The skin immune system: progress in cutaneous biology. Immunol Today. 1993;14:75–78.
    1. Glenn GM, Rao M, Matyas GR, Alving CR. Skin immunization made possible by cholera toxin. Nature. 1998;391:851.
    1. Glenn GM, Scharton-Kersten T, Vassell R, Mallett CP, Hale TL, et al. Transcutaneous immunization with cholera toxin protects mice against lethal mucosal toxin challenge. J Immunol. 1998;161:3211–3214.
    1. Glenn GM, Taylor DN, Li X, Frankel S, Montemarano A, et al. Transcutaneous immunization: a human vaccine delivery strategy using a patch. Nat Med. 2000;6:1403–1406.
    1. Hammond SA, Walwender D, Alving CR, Glenn GM. Transcutaneous immunization: T cell responses and boosting of existing immunity. Vaccine. 2001;19:2701–2707.
    1. Matyas GR, Friedlander AM, Glenn GM, Little S, Yu J, et al. Needle-free skin patch vaccination method for anthrax. Infect Immun. 2004;72:1181–1183.
    1. Glenn GM, Flyer DC, Ellingsworth LR, Frech SA, Frerichs DM, et al. Transcutaneous immunization with heat-labile enterotoxin: development of a needle-free vaccine patch. Expert Rev Vaccines. 2007;6:809–819.
    1. Frerichs DM, Ellingsworth LR, Frech SA, Flyer DC, Villar CP, et al. Controlled, single-step, stratum corneum disruption as a pretreatment for immunization via a patch. Vaccine. 2008;26:2782–2787.
    1. Vogt A, Combadiere B, Hadam S, Stieler KM, Lademann J, et al. 40 nm, but not 750 or 1,500 nm, nanoparticles enter epidermal CD1a+ cells after transcutaneous application on human skin. J Invest Dermatol. 2006;126:1316–1322.
    1. Vogt A, Mahe B, Costagliola D, Bonduelle O, Hadam S, et al. Transcutaneous anti-influenza vaccination promotes both CD4 and CD8 T cell immune responses in humans. J Immunol. 2008;180:1482–1489.
    1. Vogt A, Mandt N, Lademann J, Schaefer H, Blume-Peytavi U. Follicular targeting–a promising tool in selective dermatotherapy. J Investig Dermatol Symp Proc. 2005;10:252–255.
    1. Doherty PC, Turner SJ, Webby RG, Thomas PG. Influenza and the challenge for immunology. Nat Immunol. 2006;7:449–455.
    1. Greenbaum JA, Kotturi MF, Kim Y, Oseroff C, Vaughan K, et al. Pre-existing immunity against swine-origin H1N1 influenza viruses in the general human population. Proc Natl Acad Sci U S A. 2009;106:20365–20370.
    1. Kohlmeier JE, Woodland DL. Immunity to respiratory viruses. Annu Rev Immunol. 2009;27:61–82.
    1. Cerwenka A, Morgan TM, Dutton RW. Naive, effector, and memory CD8 T cells in protection against pulmonary influenza virus infection: homing properties rather than initial frequencies are crucial. J Immunol. 1999;163:5535–5543.
    1. Kohlmeier JE, Rumsey LM, Chan MA, Benedict SH. The outcome of T-cell costimulation through intercellular adhesion molecule-1 differs from costimulation through leucocyte function-associated antigen-1. Immunology. 2003;108:152–157.
    1. Walker BD, Burton DR. Toward an AIDS vaccine. Science. 2008;320:760–764.
    1. Goulder PJ, Watkins DI. Impact of MHC class I diversity on immune control of immunodeficiency virus replication. Nat Rev Immunol. 2008;8:619–630.
    1. Watkins DI, Burton DR, Kallas EG, Moore JP, Koff WC. Nonhuman primate models and the failure of the Merck HIV-1 vaccine in humans. Nat Med. 2008;14:617–621.
    1. Hansen SG, Vieville C, Whizin N, Coyne-Johnson L, Siess DC, et al. Effector memory T cell responses are associated with protection of rhesus monkeys from mucosal simian immunodeficiency virus challenge. Nat Med. 2009;15:293–299.
    1. Mahe B, Vogt A, Liard C, Duffy D, Abadie V, et al. Nanoparticle-Based Targeting of Vaccine Compounds to Skin Antigen-Presenting Cells By Hair Follicles and their Transport in Mice. J Invest Dermatol 2008
    1. Pulendran B, Smith JL, Caspary G, Brasel K, Pettit D, et al. Distinct dendritic cell subsets differentially regulate the class of immune response in vivo. Proc Natl Acad Sci U S A. 1999;96:1036–1041.
    1. Dudziak D, Kamphorst AO, Heidkamp GF, Buchholz VR, Trumpfheller C, et al. Differential antigen processing by dendritic cell subsets in vivo. Science. 2007;315:107–111.
    1. Stoitzner P, Tripp CH, Douillard P, Saeland S, Romani N. Migratory Langerhans cells in mouse lymph nodes in steady state and inflammation. J Invest Dermatol. 2005;125:116–125.
    1. Stoitzner P, Tripp CH, Eberhart A, Price KM, Jung JY, et al. Langerhans cells cross-present antigen derived from skin. Proc Natl Acad Sci U S A. 2006;103:7783–7788.
    1. Kissenpfennig A, Malissen B. Langerhans cells–revisiting the paradigm using genetically engineered mice. Trends Immunol. 2006;27:132–139.
    1. Matsuo M, Nagata Y, Sato E, Atanackovic D, Valmori D, et al. IFN-gamma enables cross-presentation of exogenous protein antigen in human Langerhans cells by potentiating maturation. Proc Natl Acad Sci U S A. 2004;101:14467–14472.
    1. Ratzinger G, Baggers J, de Cos MA, Yuan J, Dao T, et al. Mature human Langerhans cells derived from CD34+ hematopoietic progenitors stimulate greater cytolytic T lymphocyte activity in the absence of bioactive IL-12p70, by either single peptide presentation or cross-priming, than do dermal-interstitial or monocyte-derived dendritic cells. J Immunol. 2004;173:2780–2791.
    1. Belyakov IM, Ahlers JD, Berzofsky JA. Mucosal AIDS vaccines: current status and future directions. Expert Rev Vaccines. 2004;3:S65–73.
    1. Otberg N, Richter H, Schaefer H, Blume-Peytavi U, Sterry W, et al. Variations of hair follicle size and distribution in different body sites. J Invest Dermatol. 2004;122:14–19.
    1. Mangelsdorf S, Otberg N, Maibach HI, Sinkgraven R, Sterry W, et al. Ethnic variation in vellus hair follicle size and distribution. Skin Pharmacol Physiol. 2006;19:159–167.
    1. Etchart N, Hennino A, Friede M, Dahel K, Dupouy M, et al. Safety and efficacy of transcutaneous vaccination using a patch with the live-attenuated measles vaccine in humans. Vaccine. 2007;25:6891–6899.
    1. Yagi H, Hashizume H, Horibe T, Yoshinari Y, Hata M, et al. Induction of therapeutically relevant cytotoxic T lymphocytes in humans by percutaneous peptide immunization. Cancer Res. 2006;66:10136–10144.
    1. Doherty PC, Topham DJ, Tripp RA, Cardin RD, Brooks JW, et al. Effector CD4+ and CD8+ T-cell mechanisms in the control of respiratory virus infections. Immunol Rev. 1997;159:105–117.
    1. Hou S, Hyland L, Ryan KW, Portner A, Doherty PC. Virus-specific CD8+ T-cell memory determined by clonal burst size. Nature. 1994;369:652–654.
    1. Haanen JB, Wolkers MC, Kruisbeek AM, Schumacher TN. Selective expansion of cross-reactive CD8(+) memory T cells by viral variants. J Exp Med. 1999;190:1319–1328.
    1. Watabe S, Xin KQ, Ihata A, Liu LJ, Honsho A, et al. Protection against influenza virus challenge by topical application of influenza DNA vaccine. Vaccine. 2001;19:4434–4444.
    1. Graham MB, Braciale TJ. Resistance to and recovery from lethal influenza virus infection in B lymphocyte-deficient mice. J Exp Med. 1997;186:2063–2068.
    1. Epstein SL, Lo CY, Misplon JA, Bennink JR. Mechanism of protective immunity against influenza virus infection in mice without antibodies. J Immunol. 1998;160:322–327.
    1. Pala P, Townsend AR, Askonas BA. Viral recognition by influenza A virus cross-reactive cytotoxic T (Tc) cells: the proportion of Tc cells that recognize nucleoprotein varies between individual mice. Eur J Immunol. 1986;16:193–198.
    1. Townsend ARM, Rothbard J, Gotch FM, Bahadur G, Wraith D, et al. The epitopes of influenza nucleoprotein recognized by cytotoxic T lymphocytes can be defined with short synthetic peptides. Cell. 1986;44:959–968.
    1. Belz GT, Doherty PC. Virus-specific and bystander CD8+ T-cell proliferation in the acute and persistent phases of a gammaherpesvirus infection. J Virol. 2001;75:4435–4438.
    1. Belz GT, Xie W, Doherty PC. Diversity of epitope and cytokine profiles for primary and secondary influenza a virus-specific CD8+ T cell responses. J Immunol. 2001;166:4627–4633.
    1. Belz GT, Xie W, Altman JD, Doherty PC. A previously unrecognized H-2D(b)-restricted peptide prominent in the primary influenza A virus-specific CD8(+) T-cell response is much less apparent following secondary challenge. J Virol. 2000;74:3486–3493.
    1. Tamura M, Kuwano K, Kurane I, Ennis FA. Definition of amino acid residues on the epitope responsible for recognition by influenza A virus H1-specific, H2-specific, and H1- and H2-cross-reactive murine cytotoxic T-lymphocyte clones. J Virol. 1998;72:9404–9406.
    1. Kuwano K, Tamura M, Ennis FA. Cross-reactive protection against influenza A virus infections by an NS1-specific CTL clone. Virology. 1990;178:174–179.
    1. Kuwano K, Scott M, Young JF, Ennis FA. Active immunization against virus infections due to antigenic drift by induction of crossreactive cytotoxic T lymphocytes. J Exp Med. 1989;169:1361–1371.
    1. Kuwano K, Braciale TJ, Ennis FA. Cytotoxic T lymphocytes recognize a cross-reactive epitope on the transmembrane region of influenza H1 and H2 hemagglutinins. Viral Immunol. 1989;2:163–173.
    1. Kuwano K, Scott M, Young JF, Ennis FA. HA2 subunit of influenza A H1 and H2 subtype viruses induces a protective cross-reactive cytotoxic T lymphocyte response. J Immunol. 1988;140:1264–1268.
    1. Murasko DM, Bernstein ED, Gardner EM, Gross P, Munk G, et al. Role of humoral and cell-mediated immunity in protection from influenza disease after immunization of healthy elderly. Exp Gerontol. 2002;37:427–439.
    1. Bernstein E, Kaye D, Abrutyn E, Gross P, Dorfman M, et al. Immune response to influenza vaccination in a large healthy elderly population. Vaccine. 1999;17:82–94.
    1. Makedonas G, Betts MR. Polyfunctional analysis of human t cell responses: importance in vaccine immunogenicity and natural infection. Springer Semin Immunopathol. 2006;28:209–219.
    1. Harari A, Dutoit V, Cellerai C, Bart PA, Du Pasquier RA, et al. Functional signatures of protective antiviral T-cell immunity in human virus infections. Immunol Rev. 2006;211:236–254.
    1. Fan H, Lin Q, Morrissey GR, Khavari PA. Immunization via hair follicles by topical application of naked DNA to normal skin. Nat Biotechnol. 1999;17:870–872.
    1. Lademann J, Otberg N, Jacobi U, Hoffman RM, Blume-Peytavi U. Follicular penetration and targeting. J Investig Dermatol Symp Proc. 2005;10:301–303.
    1. Lademann J, Richter H, Schaefer UF, Blume-Peytavi U, Teichmann A, et al. Hair follicles - a long-term reservoir for drug delivery. Skin Pharmacol Physiol. 2006;19:232–236.
    1. Naito S, Maeyama J, Mizukami T, Takahashi M, Hamaguchi I, et al. Transcutaneous immunization by merely prolonging the duration of antigen presence on the skin of mice induces a potent antigen-specific antibody response even in the absence of an adjuvant. Vaccine. 2007;25:8762–8770.
    1. Glenn GM, Kenney RT. Mass vaccination: solutions in the skin. Curr Top Microbiol Immunol. 2006;304:247–268.
    1. Kenney RT, Frech SA, Muenz LR, Villar CP, Glenn GM. Dose sparing with intradermal injection of influenza vaccine. N Engl J Med. 2004;351:2295–2301.
    1. Halperin SA, Smith B, Clarke K, Treanor J, Mabrouk T, et al. Phase I, randomized, controlled trial to study the reactogenicity and immunogenicity of a nasal, inactivated trivalent influenza virus vaccine in healthy adults. Hum Vaccin. 2005;1:37–42.
    1. Draenert R, Altfeld M, Brander C, Basgoz N, Corcoran C, et al. Comparison of overlapping peptide sets for detection of antiviral CD8 and CD4 T cell responses. J Immunol Methods. 2003;275:19–29.
    1. Beavis AJ, Kalejta RF. Simultaneous analysis of the cyan, green, and yellow fluorescent proteins. Curr Protoc Cytom Chapter. 2001;1:Unit 1 16.
    1. Appay V, Rowland-Jones SL. The assessment of antigen-specific CD8+ T cells through the combination of MHC class I tetramer and intracellular staining. J Immunol Methods. 2002;268:9–19.
    1. Appay V, Nixon DF, Donahoe SM, Gillespie GM, Dong T, et al. HIV-specific CD8(+) T cells produce antiviral cytokines but are impaired in cytolytic function. J Exp Med. 2000;192:63–75.

Source: PubMed

3
Se inscrever