Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Proc Natl Acad Sci U S A ; 108(9): 3677-82, 2011 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-21321220

RESUMO

Regulatory T cells (Treg) play critical roles in the modulation of immune responses to infectious agents. Further understanding of the factors that control Treg activation and expansion in response to pathogens is needed to manipulate Treg function in acute and chronic infections. Here we show that chronic, but not acute, infection of mice with lymphocytic choriomeningitis virus results in a marked expansion of Foxp3(+) Treg that is dependent on retroviral superantigen (sag) genes encoded in the mouse genome. Sag-dependent Treg expansion was MHC class II dependent, CD4 independent, and required dendritic cells. Thus, one unique mechanism by which certain infectious agents evade host immune responses may be mediated by endogenous Sag-dependent activation and expansion of Treg.


Assuntos
Retrovirus Endógenos/imunologia , Coriomeningite Linfocítica/imunologia , Coriomeningite Linfocítica/virologia , Vírus da Coriomeningite Linfocítica/imunologia , Superantígenos/imunologia , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/imunologia , Animais , Proliferação de Células , Doença Crônica , Células Dendríticas/citologia , Células Dendríticas/imunologia , Fatores de Transcrição Forkhead/metabolismo , Antígenos de Histocompatibilidade Classe II/imunologia , Vírus do Tumor Mamário do Camundongo/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL
2.
J Virol ; 86(23): 12605-15, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22973033

RESUMO

Modified vaccinia virus Ankara (MVA) is a safe, attenuated orthopoxvirus that is being developed as a vaccine vector but has demonstrated limited immunogenicity in several early-phase clinical trials. Our objective was to rationally improve the immunogenicity of MVA-based HIV/AIDS vaccines via the targeted deletion of specific poxvirus immune-modulatory genes. Vaccines expressing codon-optimized HIV subtype C consensus Env and Gag antigens were generated from MVA vector backbones that (i) harbor simultaneous deletions of four viral immune-modulatory genes, encoding an interleukin-18 (IL-18) binding protein, an IL-1ß receptor, a dominant negative Toll/IL-1 signaling adapter, and CC-chemokine binding protein (MVAΔ4-HIV); (ii) harbor a deletion of an additional (fifth) viral gene, encoding uracil-DNA glycosylase (MVAΔ5-HIV); or (iii) represent the parental MVA backbone as a control (MVA-HIV). We performed head-to-head comparisons of the cellular and humoral immune responses that were elicited by these vectors during homologous prime-boost immunization regimens utilizing either high-dose (2 × 10(8) PFU) or low-dose (1 × 10(7) PFU) intramuscular immunization of rhesus macaques. At all time points, a majority of the HIV-specific T cell responses, elicited by all vectors, were directed against Env, rather than Gag, determinants, as previously observed with other vector systems. Both modified vectors elicited up to 6-fold-higher frequencies of HIV-specific CD8 and CD4 T cell responses and up to 25-fold-higher titers of Env (gp120)-specific binding (nonneutralizing) antibody responses that were relatively transient in nature. While the correlates of protection against HIV infection remain incompletely defined, our results indicate that the rational deletion of specific genes from MVA vectors can positively alter their cellular and humoral immunogenicity profiles in nonhuman primates.


Assuntos
Vacinas contra a AIDS/imunologia , Formação de Anticorpos/imunologia , Vetores Genéticos/imunologia , Linfócitos T/imunologia , Vacinas Virais/genética , Produtos do Gene env do Vírus da Imunodeficiência Humana/imunologia , Produtos do Gene gag do Vírus da Imunodeficiência Humana/imunologia , Vacinas contra a AIDS/administração & dosagem , Vacinas contra a AIDS/genética , Animais , Relação Dose-Resposta a Droga , Feminino , Deleção de Genes , Injeções Intramusculares , Peptídeos e Proteínas de Sinalização Intercelular/genética , Macaca mulatta , Receptores de Quimiocinas/genética , Receptores de Interleucina-1/genética , Vacinas de DNA , Vacinas Virais/imunologia , Produtos do Gene env do Vírus da Imunodeficiência Humana/genética , Produtos do Gene gag do Vírus da Imunodeficiência Humana/genética
3.
J Exp Med ; 203(10): 2263-9, 2006 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-16966427

RESUMO

Numerous microbes establish persistent infections, accompanied by antigen-specific CD8 T cell activation. Pathogen-specific T cells in chronically infected hosts are often phenotypically and functionally variable, as well as distinct from T cells responding to nonpersistent infections; this phenotypic heterogeneity has been attributed to an ongoing reencounter with antigen. Paradoxically, maintenance of memory CD8 T cells to acutely resolved infections is antigen independent, whereas there is a dependence on antigen for T cell survival in chronically infected hosts. Using two chronic viral infections, we demonstrate that new naive antigen-specific CD8 T cells are primed after the acute phase of infection. These newly recruited T cells are phenotypically distinct from those primed earlier. Long-lived antiviral CD8 T cells are defective in self-renewal, and lack of thymic output results in the decline of virus-specific CD8 T cells, indicating that newly generated T cells preserve antiviral CD8 T cell populations during chronic infection. These findings reveal a novel role for antigen in maintaining virus-specific CD8 T cells during persistent infection and provide insight toward understanding T cell differentiation in chronic infection.


Assuntos
Antígenos Virais/imunologia , Linfócitos T CD8-Positivos/citologia , Diferenciação Celular/imunologia , Vírus da Coriomeningite Linfocítica/imunologia , Polyomavirus/imunologia , Viroses/imunologia , Animais , Linfócitos T CD8-Positivos/imunologia , Feminino , Citometria de Fluxo , Camundongos , Camundongos Endogâmicos C57BL
4.
J Exp Med ; 211(9): 1905-18, 2014 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-25113973

RESUMO

Regulatory T (T reg) cells are critical for preventing autoimmunity mediated by self-reactive T cells, but their role in modulating immune responses during chronic viral infection is not well defined. To address this question and to investigate a role for T reg cells in exhaustion of virus-specific CD8 T cells, we depleted T reg cells in mice chronically infected with lymphocytic choriomeningitis virus (LCMV). T reg cell ablation resulted in 10-100-fold expansion of functional LCMV-specific CD8 T cells. Rescue of exhausted CD8 T cells was dependent on cognate antigen, B7 costimulation, and conventional CD4 T cells. Despite the striking recovery of LCMV-specific CD8 T cell responses, T reg cell depletion failed to diminish viral load. Interestingly, T reg cell ablation triggered up-regulation of the molecule programmed cell death ligand-1 (PD-L1), which upon binding PD-1 on T cells delivers inhibitory signals. Increased PD-L1 expression was observed especially on LCMV-infected cells, and combining T reg cell depletion with PD-L1 blockade resulted in a significant reduction in viral titers, which was more pronounced than that upon PD-L1 blockade alone. These results suggest that T reg cells effectively maintain CD8 T cell exhaustion, but blockade of the PD-1 inhibitory pathway is critical for elimination of infected cells.


Assuntos
Coriomeningite Linfocítica/imunologia , Coriomeningite Linfocítica/virologia , Receptor de Morte Celular Programada 1/imunologia , Linfócitos T Reguladores/imunologia , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Doença Crônica , Células Dendríticas/imunologia , Ativação Linfocitária , Depleção Linfocítica , Vírus da Coriomeningite Linfocítica/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Regulação para Cima
5.
PLoS One ; 4(5): e5445, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19421328

RESUMO

BACKGROUND: Modified Vaccinia virus Ankara (MVA) is a safe, highly attenuated orthopoxvirus that is being developed as a recombinant vaccine vector for immunization against a number of infectious diseases and cancers. However, the expression by MVA vectors of large numbers of poxvirus antigens, which display immunodominance over vectored antigens-of-interest for the priming of T cell responses, and the induction of vector-neutralizing antibodies, which curtail the efficacy of subsequent booster immunizations, remain as significant impediments to the overall utility of such vaccines. Thus, genetic approaches that enable the derivation of MVA vectors that are antigenically less complex may allow for rational improvement of MVA-based vaccines. PRINCIPAL FINDINGS: We have developed a genetic complementation system that enables the deletion of essential viral genes from the MVA genome, thereby allowing us to generate MVA vaccine vectors that are antigenically less complex. Using this system, we deleted the essential uracil-DNA-glycosylase (udg) gene from MVA and propagated this otherwise replication-defective variant on a complementing cell line that constitutively expresses the poxvirus udg gene and that was derived from a newly identified continuous cell line that is permissive for growth of wild type MVA. The resulting virus, MVADeltaudg, does not replicate its DNA genome or express late viral gene products during infection of non-complementing cells in culture. As proof-of-concept for immunological 'focusing', we demonstrate that immunization of mice with MVADeltaudg elicits CD8+ T cell responses that are directed against a restricted repertoire of vector antigens, as compared to immunization with parental MVA. Immunization of rhesus macaques with MVADeltaudg-gag, a udg(-) recombinant virus that expresses an HIV subtype-B consensus gag transgene, elicited significantly higher frequencies of Gag-specific CD8 and CD4 T cells following both primary (2-4-fold) and booster (2-fold) immunizations as compared to the udg(+) control virus MVA-gag, as determined by intracellular cytokine assay. In contrast, levels of HIV Gag-specific antibodies were elicited similarly in macaques following immunization with MVADeltaudg-gag and MVA-gag. Furthermore, both udg(-) and udg(+) MVA vectors induced comparatively similar titers of MVA-specific neutralizing antibody responses following immunization of mice (over a 4-log range: 10(4)-10(8) PFU) and rhesus macaques. These results suggest that the generation of MVA-specific neutralizing antibody responses are largely driven by input MVA antigens, rather than those that are synthesized de novo during infection, and that the processes governing the generation of antiviral antibody responses are more readily saturated by viral antigen than are those that elicit CD8+ T cell responses. SIGNIFICANCE: Our identification of a spontaneously-immortalized (but not transformed) chicken embryo fibroblast cell line (DF-1) that is fully permissive for MVA growth and that can be engineered to stably express MVA genes provides the basis for a genetic system for MVA. DF-1 cells (and derivatives thereof) constitute viable alternatives, for the manufacture of MVA-based vaccines, to primary CEFs -- the conventional cell substrate for MVA vaccines that is not amenable to genetic complementation strategies due to these cells' finite lifespan in culture. The establishment of a genetic system for MVA, as illustrated here to allow udg deletion, enables the generation of novel replication-defective MVA mutants and expands the repertoire of genetic viral variants that can now be explored as improved vaccine vectors.


Assuntos
Anticorpos Antivirais/imunologia , Teste de Complementação Genética , Vetores Genéticos/imunologia , Anticorpos Anti-HIV/imunologia , Vaccinia virus/imunologia , Vacinas Virais/imunologia , Animais , Southern Blotting , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Embrião de Galinha/citologia , Embrião de Galinha/metabolismo , Galinhas , Replicação do DNA , Feminino , Fibroblastos/citologia , Citometria de Fluxo , Imunização Secundária , Macaca mulatta , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Testes de Neutralização , Transgenes/fisiologia , Uracila-DNA Glicosidase/genética , Uracila-DNA Glicosidase/imunologia , Vaccinia virus/genética , Vacinas Virais/genética , Replicação Viral , Produtos do Gene gag do Vírus da Imunodeficiência Humana/imunologia , Produtos do Gene gag do Vírus da Imunodeficiência Humana/metabolismo
6.
Cancer Res ; 69(7): 3086-94, 2009 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-19293186

RESUMO

Dendritic cells (DC) have a critical effect on the outcome of adaptive immune responses against growing tumors. Whereas it is generally assumed that the presence of phenotypically mature DCs should promote protective antitumor immunity, evidence to the contrary does exist. We describe here a novel mechanism by which tumor-infiltrating dendritic cells (TIDC) actively contribute to the suppression of protective CD8(+) T-cell-based antitumor immunity. Using the BALB/NeuT model of spontaneously arising mammary carcinoma, we found that canonical MHC II(+)/CD11b(+)/CD11c(high) TIDCs act as regulatory DCs to suppress CD8(+) T-cell function, resulting in diminished T-cell-based antitumor immunity in vivo. Stimulation of naive T cells with regulatory TIDCs resulted in an altered cell fate program characterized by minimal T-cell expansion, impaired IFNgamma production, and anergy. Suppression by regulatory TIDCs overcame stimulatory signals provided by standard DCs, occurred in the absence of cognate interactions with T cells, and was mediated primarily by arginase metabolism of l-arginine. Immunosuppressive TIDCs were found in every murine tumor type examined and were phenotypically distinct from tumor-infiltrating CD11c(int-low)/CD11b(+)/Gr-1(+) myeloid-derived suppressor cells. Thus, within the tumor microenvironment, MHC II(+) TIDCs can function as potent suppressors of CD8(+) T-cell immunity.


Assuntos
Arginina/metabolismo , Linfócitos T CD8-Positivos/imunologia , Células Dendríticas/imunologia , Neoplasias Mamárias Experimentais/imunologia , Neoplasias Mamárias Experimentais/metabolismo , Animais , Antígeno CD11c/imunologia , Linfócitos T CD8-Positivos/metabolismo , Processos de Crescimento Celular/fisiologia , Anergia Clonal/imunologia , Células Dendríticas/metabolismo , Citometria de Fluxo , Ativação Linfocitária , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos
7.
Science ; 323(5922): 1726-9, 2009 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-19325114

RESUMO

In the early stages of viral infection, outcomes depend on a race between expansion of infection and the immune response generated to contain it. We combined in situ tetramer staining with in situ hybridization to visualize, map, and quantify relationships between immune effector cells and their targets in tissues. In simian immunodeficiency virus infections in macaques and lymphocytic choriomeningitis virus infections in mice, the magnitude and timing of the establishment of an excess of effector cells versus targets were found to correlate with the extent of control and the infection outcome (i.e., control and clearance versus partial or poor control and persistent infection). This method highlights the importance of the location, timing, and magnitude of the immune response needed for a vaccine to be effective against agents of persistent infection, such as HIV-1.


Assuntos
Infecções por Arenaviridae/imunologia , Vírus da Coriomeningite Linfocítica/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Vírus da Imunodeficiência Símia/imunologia , Linfócitos T Citotóxicos/imunologia , Animais , Infecções por Arenaviridae/virologia , Contagem de Células , Colo do Útero/imunologia , Colo do Útero/virologia , Feminino , Hibridização In Situ , Linfonodos/imunologia , Linfonodos/virologia , Tecido Linfoide/imunologia , Tecido Linfoide/virologia , Macaca mulatta , Camundongos , RNA Viral/análise , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Vírus da Imunodeficiência Símia/fisiologia , Baço/imunologia , Baço/virologia , Coloração e Rotulagem , Fatores de Tempo , Vagina/imunologia , Vagina/virologia , Replicação Viral
8.
J Immunol ; 174(8): 4662-9, 2005 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-15814690

RESUMO

Peripheral tolerance to shared Ags expressed on both tumors and normal self-tissues presents a major barrier to T cell-based immunotherapy as a treatment for cancer. To assess the activity of tumor-specific T cells against spontaneously arising carcinomas in the context of shared Ag expression, we developed a model system whereby an identified tumor Ag, tumor ERK (tERK), is expressed transgenically on both normal mammary tissue and spontaneous mammary carcinomas. Transfer of in vitro-activated, tERK-specific DUC18 T cells delayed spontaneous tumor development in tERK-expressing mice when T cells were given before the development of palpable carcinomas. However, antitumor activity mediated by in vitro-activated DUC18 T cells, as measured by responsiveness against a transplanted tERK-expressing fibrosarcoma challenge, was lost within days of transfer. This loss was due to expression of tERK as a self-Ag on normal tissues and was independent of the presence of mammary tumors. In contrast, transferred naive DUC18 T cells maintained a long-term protective function in tERK-expressing mice. Ten-fold fewer naive T cells activated in vivo were able to replicate the delay in spontaneous tumor development achieved by in vitro-activated T cells. These results are in contrast to our earlier studies using transplanted tumors alone, in which in vitro-activated DUC18 T cells were more efficacious than naive DUC18 T cells and highlight the need to perform tumor studies in the presence of tumor Ag expression on normal self-tissue.


Assuntos
Neoplasias Mamárias Experimentais/etiologia , Neoplasias Mamárias Experimentais/imunologia , Linfócitos T/imunologia , Animais , Antígenos de Neoplasias , Autoantígenos , Sequência de Bases , DNA Recombinante/genética , Feminino , Tolerância Imunológica , Imunoterapia Adotiva , Técnicas In Vitro , Ativação Linfocitária , Neoplasias Mamárias Experimentais/terapia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos
9.
Int Immunol ; 15(7): 797-805, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12807818

RESUMO

Utilization of ex vivo-expanded epitope-specific cytotoxic T lymphocytes has become a clinical standard in the adoptive immunotherapy of tumors. One of the obstacles faced by T cell-based immunotherapy is the development of tumor immune-escape variants. Using our previously reported CMS5 tumor/DUC18 CD8(+) TCR transgenic system, we sought to investigate whether large established tumors can be successfully eliminated before the development of escape variants. Using BALB/c mice that were s.c. transplanted with two tumors that had been growing for 8 days (double 8-day tumors), we assessed the in vivo anti-tumor activity of in vitro peptide-stimulated DUC18 T cells. A single infusion of activated DUC18 T cells showed a modest effect against the double 8-day tumors, whereas two and three administrations led to regression of both tumors within 10 days. However, in some mice, the tumors re-grew approximately 10 days after the regression. We found these tumors to be antigen-loss variants. These relapsed tumor cells progressively grew in DUC18 transgenic mice and did not express tERK-specific message. When four doses of activated DUC18 T cells were infused, the double 8-day tumors were successfully eliminated and the tumors did not grow out in any mice. Our results demonstrate that mono-specific CD8(+) T cells can effectively eliminate large established tumors before the development of antigen-loss variants when a high number of T cells is rapidly administered.


Assuntos
Antígenos de Neoplasias/imunologia , Epitopos de Linfócito T/imunologia , Imunoterapia Adotiva , Neoplasias Experimentais/terapia , Linfócitos T Citotóxicos/transplante , Animais , Variação Antigênica , Antígenos de Neoplasias/análise , Antígenos de Neoplasias/genética , Apoptose , Linhagem Celular Tumoral , Epitopos , Epitopos de Linfócito T/química , Epitopos de Linfócito T/metabolismo , Variação Genética , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos BALB C , Neoplasias Experimentais/imunologia , Peptídeos/genética , Peptídeos/imunologia , Linfócitos T Citotóxicos/imunologia
10.
J Immunol ; 172(1): 310-7, 2004 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-14688339

RESUMO

The lung is a common site of metastatic and primary tumor growth, and has been shown to be an immunosuppressive environment. We tested the impact of the lung environment on the development of tumor-specific T cell responses against the CMS5 fibrosarcoma, and found a deficit in the efficacy of naive tumor-specific DUC18 T cells against tumors established in the lung. One hundred-fold more naive tumor-specific T cells were required to protect against tumor development or reject established tumors in the lung than an identical tumor challenge delivered s.c. in the flank. Importantly, CMS5 growing in the flank facilitated the rejection of tumors present in the lungs. In the presence of flank tumors, transferred T cells were not phenotypically altered but were present in much greater numbers in the parabronchial lymph nodes, bronchoalveolar lavage, and lung parenchyma than in mice bearing lung tumors alone. We hypothesized that APC present in the lung and skin draining lymph nodes were differentially initiating T cell proliferation, leading to differences in the size of the final effector populations. A direct comparison of DUC18 T cell proliferation against APC from flank or lung draining lymph nodes showed profoundly greater proliferation to flank draining lymph node APC. The impaired stimulation of naive T cell proliferation by lung draining APC provides one mechanistic explanation for the lower overall immune response, and inability to effectively reject tumors, in the lung.


Assuntos
Antígenos de Neoplasias/imunologia , Epitopos de Linfócito T/imunologia , Neoplasias Pulmonares/imunologia , Subpopulações de Linfócitos T/imunologia , Animais , Divisão Celular/imunologia , Linhagem Celular Tumoral , Fibrossarcoma/imunologia , Fibrossarcoma/patologia , Fibrossarcoma/prevenção & controle , Rejeição de Enxerto/imunologia , Membro Posterior , Injeções Intravenosas , Injeções Subcutâneas , Interfase/imunologia , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/prevenção & controle , Linfonodos/imunologia , Linfonodos/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Transplante de Neoplasias , Neoplasias Cutâneas/imunologia , Neoplasias Cutâneas/patologia , Subpopulações de Linfócitos T/patologia , Regulação para Cima/imunologia
11.
J Immunol ; 172(7): 4215-24, 2004 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-15034034

RESUMO

Peptide vaccination is an immunotherapeutic strategy being pursued as a method of enhancing Ag-specific antitumor responses. To date, most studies have focused on the use of MHC class I-restricted peptides, and have not shown a correlation between Ag-specific CD8(+) T cell expansion and the generation of protective immune responses. We investigated the effects of CD4-directed peptide vaccination on the ability of CD8(+) T cells to mount protective antitumor responses in the DUC18/CMS5 tumor model system. To accomplish this, we extended the amino acid sequence of the known MHC class I-restricted DUC18 rejection epitope from CMS5 to allow binding to MHC class II molecules. Immunization with this peptide (tumor-derived extracellular signal-regulated kinase-II (tERK-II)) induced Ag-specific CD4(+) T cell effector function, but did not directly prime CD8(+) T cells. Approximately 31% of BALB/c mice immunized with tERK-II were protected from subsequent tumor challenge in a CD40-dependent manner. Priming of endogenous CD8(+) T cells in immunized mice was detected only after CMS5 challenge. Heightened CD4(+) Th cell function in response to tERK II vaccination allowed a 12-fold reduction in the number of adoptively transferred CD8(+) DUC18 T cells needed to protect recipients against tumor challenge as compared with previous studies using unimmunized mice. Furthermore, tERK-II immunization led to a more rapid and transient expansion of transferred DUC18 T cells than was seen in unimmunized mice. These findings illustrate that CD4-directed peptide vaccination augments antitumor immunity, but that the number of tumor-specific precursor CD8(+) T cells will ultimately dictate the success of immunotherapy.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Antineoplásicos/imunologia , Antígenos CD4/imunologia , Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/imunologia , Fibrossarcoma/imunologia , Fibrossarcoma/prevenção & controle , Células-Tronco/imunologia , Sequência de Aminoácidos , Animais , Antineoplásicos/administração & dosagem , Antígenos CD4/administração & dosagem , Antígenos CD40/fisiologia , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/transplante , Vacinas Anticâncer/administração & dosagem , Divisão Celular/genética , Divisão Celular/imunologia , Linhagem Celular Tumoral , Epitopos de Linfócito T/administração & dosagem , Epitopos de Linfócito T/imunologia , Feminino , Fibrossarcoma/patologia , Injeções Subcutâneas , Interfase/genética , Interfase/imunologia , Ativação Linfocitária/imunologia , Contagem de Linfócitos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Camundongos SCID , Camundongos Transgênicos , Dados de Sequência Molecular , Transplante de Neoplasias , Células-Tronco/citologia , Linfócitos T Reguladores/imunologia , Vacinas de Subunidades Antigênicas/administração & dosagem , Vacinas de Subunidades Antigênicas/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA