Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
J Immunol ; 205(5): 1449-1460, 2020 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-32737148

RESUMO

Optimal ex vivo expansion protocols of tumor-specific T cells followed by adoptive cell therapy must yield T cells able to home to tumors and effectively kill them. Our previous study demonstrated ex vivo activation in the presence of IL-12-induced optimal CD8+ T cell expansion and melanoma regression; however, adverse side effects, including autoimmunity, can occur. This may be due to transfer of high-avidity self-specific T cells. In this study, we compared mouse low- and high-avidity T cells targeting the tumor Ag tyrosinase-related protein 2 (TRP2). Not surprisingly, high-avidity T cells provide superior tumor control, yet low-avidity T cells can promote tumor regression. The addition of IL-12 during in vitro expansion boosts low-avidity T cell responsiveness, tumor regression, and prevents T cell exhaustion. In this study, we demonstrate that IL-12-primed T cells are resistant to PD-1/PD-L1-mediated suppression and retain effector function. Importantly, IL-12 preconditioning prevented exhaustion as LAG-3, PD-1, and TOX were decreased while simultaneously increasing KLRG1. Using intravital imaging, we also determined that high-avidity T cells have sustained contacts with intratumoral dendritic cells and tumor targets compared with low-avidity T cells. However, with Ag overexpression, this defect is overcome, and low-avidity T cells control tumor growth. Taken together, these data illustrate that low-avidity T cells can be therapeutically beneficial if cocultured with IL-12 cytokine during in vitro expansion and highly effective in vivo if Ag is not limiting. Clinically, low-avidity T cells provide a safer alternative to high-avidity, TCR-engineered T cells, as IL-12-primed, low-avidity T cells cause less autoimmune vitiligo.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Interleucina-12/imunologia , Ativação Linfocitária/imunologia , Melanoma Experimental/imunologia , Melanoma Experimental/terapia , Animais , Antígenos de Neoplasias/imunologia , Autoimunidade/imunologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Terapia Baseada em Transplante de Células e Tecidos/métodos , Imunoterapia Adotiva/métodos , Proteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Fragmentos de Peptídeos/imunologia , Receptores de Antígenos de Linfócitos T/imunologia
2.
Melanoma Res ; 30(2): 147-158, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31205227

RESUMO

Therapeutic activation of macrophage phagocytosis has the ability to restrain tumour growth through phagocytic clearance of tumour cells and activation of the adaptive immune response. Our objective for this study was to evaluate the effects of modulating pro- and anti-phagocytic pathways in malignant melanoma. In order to identify evolutionarily conserved mechanisms of resistance that may be important for melanoma cell survival, we utilized a multi-species approach and examined the phagocytosis of human, mouse, and dog melanoma cells. We observed that melanoma cells from all three species displayed unexpected resistance to phagocytosis that could not be fully mitigated by blockade of the 'don't eat me' signal CD47 or by chemotherapeutic enhancement of known 'eat me' signals. Additionally, CD47 blockade failed to promote anti-melanoma immune responses or tumour regression in vivo. This melanoma resistance to phagocytosis was not mediated by soluble factors, and it was unaffected by siRNA-mediated knockdown of 47 prospective 'don't eat me' signals or by CRISPR-Cas-mediated CD47 knockout. Unexpectedly, CD47 knockout also did not enhance phagocytosis of lymphoma cells, but it eliminated the pro-phagocytic effect of CD47 blockade, suggesting that the pro-phagocytic effects of CD47 blockade are due in part to Fc receptor engagement. From this study, we conclude that melanoma cells possess an evolutionarily conserved resistance to macrophage phagocytosis. Further investigation will be needed to overcome the mechanisms that mediate melanoma cell resistance to innate immunity.


Assuntos
Antígeno CD47/metabolismo , Melanoma/genética , Fagocitose/fisiologia , Animais , Linhagem Celular Tumoral , Humanos , Camundongos , Transdução de Sinais , Transfecção , Regulação para Cima
3.
J Immunol ; 191(7): 3681-93, 2013 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-23997225

RESUMO

A hallmark of T cell activation in vitro and in vivo is the clustering of T cells with each other via interaction of the LFA-1 integrin with ICAM-1. The functional significance of these homotypic aggregates in regulating T cell function remains unknown. We used an APC-free in vitro activation system to demonstrate that stimulation of purified naive CD8 T cells results in enhanced expression of ICAM-1 on T cells that is sustained by the inflammatory cytokine IL-12 and associated with robust T cell aggregates. ICAM-1-deficient CD8 T cells proliferate normally but demonstrate a striking failure to aggregate. Interestingly, loss of ICAM-1 expression results in elevated levels of IFN-γ and granzyme B, as well as enhanced cytotoxicity. Similar results were obtained when anti-LFA-1 Ab was used to block the clustering of wild-type T cells. ICAM-1 ligation is not required for IFN-γ regulation, as clustering of ICAM-1-deficient CD8 T cells with wild-type T cells reduces IFN-γ expression. Analysis using a fluorescent reporter that monitors TCR signal strength indicates that T cell clustering limits T cell exposure to Ag during activation. Furthermore, T cell clustering promotes the upregulation of the CTLA-4 inhibitory receptor and the downregulation of eomesodermin, which controls effector molecule expression. Activation of ICAM-1-deficient CD8 T cells in vivo results in an enhanced percentage of KLRG-1(+) T cells indicative of short-lived effectors. These results suggest that T cell clustering represents a mechanism that allows continued proliferation but regulates T cell effector function and differentiation.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Diferenciação Celular/imunologia , Molécula 1 de Adesão Intercelular/metabolismo , Ativação Linfocitária/imunologia , Animais , Antígenos/imunologia , Linfócitos T CD8-Positivos/citologia , Antígeno CTLA-4/genética , Antígeno CTLA-4/metabolismo , Comunicação Celular/imunologia , Diferenciação Celular/genética , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Regulação da Expressão Gênica , Molécula 1 de Adesão Intercelular/genética , Interferon gama/biossíntese , Interleucina-12/farmacologia , Ativação Linfocitária/efeitos dos fármacos , Ativação Linfocitária/genética , Camundongos , Subpopulações de Linfócitos T/citologia , Subpopulações de Linfócitos T/efeitos dos fármacos , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo
4.
Immunity ; 39(1): 111-22, 2013 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-23871208

RESUMO

Immune cells sense microbial products through Toll-like receptors (TLR), which trigger host defense responses including type 1 interferons (IFNs) secretion. A coding polymorphism in the protein tyrosine phosphatase nonreceptor type 22 (PTPN22) gene is a susceptibility allele for human autoimmune and infectious disease. We report that Ptpn22 selectively regulated type 1 IFN production after TLR engagement in myeloid cells. Ptpn22 promoted host antiviral responses and was critical for TLR agonist-induced, type 1 IFN-dependent suppression of inflammation in colitis and arthritis. PTPN22 directly associated with TNF receptor-associated factor 3 (TRAF3) and promotes TRAF3 lysine 63-linked ubiquitination. The disease-associated PTPN22W variant failed to promote TRAF3 ubiquitination, type 1 IFN upregulation, and type 1 IFN-dependent suppression of arthritis. The findings establish a candidate innate immune mechanism of action for a human autoimmunity "risk" gene in the regulation of host defense and inflammation.


Assuntos
Autoimunidade/imunologia , Imunidade/imunologia , Interferon Tipo I/imunologia , Proteína Tirosina Fosfatase não Receptora Tipo 22/imunologia , Receptores Toll-Like/imunologia , Animais , Artrite/genética , Artrite/imunologia , Autoimunidade/genética , Linhagem Celular , Células Cultivadas , Colite/induzido quimicamente , Colite/genética , Colite/imunologia , Sulfato de Dextrana/imunologia , Células HEK293 , Interações Hospedeiro-Patógeno/imunologia , Humanos , Imunidade/genética , Immunoblotting , Interferon Tipo I/genética , Interferon Tipo I/metabolismo , Coriomeningite Linfocítica/genética , Coriomeningite Linfocítica/imunologia , Coriomeningite Linfocítica/virologia , Vírus da Coriomeningite Linfocítica/imunologia , Vírus da Coriomeningite Linfocítica/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Células Mieloides/imunologia , Células Mieloides/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 22/genética , Proteína Tirosina Fosfatase não Receptora Tipo 22/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator 3 Associado a Receptor de TNF/genética , Fator 3 Associado a Receptor de TNF/imunologia , Fator 3 Associado a Receptor de TNF/metabolismo , Receptores Toll-Like/genética , Receptores Toll-Like/metabolismo , Ubiquitinação/imunologia
5.
J Immunol ; 191(3): 1011-5, 2013 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-23804712

RESUMO

Naive CD8 T cells proliferate in response to TCR and CD28 signals, but require IL-12 or type I IFN to survive and develop optimal effector functions. Although murine CTL generated in vitro in response to IL-12 or IFN-α had comparable effector functions, IL-12-stimulated cells were significantly more effective in controlling tumor in an adoptive immunotherapy model. They maintained high numbers and function, whereas IFN-α-stimulated cells declined in number and became exhausted. Consistent with this, IFN-α-stimulated cells in the tumor expressed higher levels of programmed death 1 (PD-1) inhibitory receptor than did IL-12-stimulated cells. When blocking Ab specific for the PD-L1 ligand of PD-1 was administered, the efficacy of IFN-α-stimulated CTL became comparable with that of IL-12-stimulated cells. Thus, IL-12 and IFN-α differentially program CD8 T cells to re-express distinct levels of PD-1 upon re-encountering Ag, resulting in IL-12-stimulated cells being less susceptible to exhaustion in the face of sustained tumor Ag.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Interferon Tipo I/metabolismo , Interferon-alfa/metabolismo , Interleucina-12/metabolismo , Receptor de Morte Celular Programada 1/metabolismo , Animais , Anticorpos Monoclonais/imunologia , Antígeno B7-H1/imunologia , Antígeno B7-H1/metabolismo , Proliferação de Células , Ativação Linfocitária/imunologia , Camundongos , Neoplasias/imunologia , Receptor de Morte Celular Programada 1/biossíntese , Transdução de Sinais/imunologia , Linfócitos T Citotóxicos/imunologia
6.
J Immunol ; 189(2): 659-68, 2012 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-22706089

RESUMO

Autocrine IFN-γ signaling is important for CD4 differentiation to Th1 effector cells, but it has been unclear whether it contributes to CD8 T cell differentiation. We show in this paper that naive murine CD8 T cells rapidly and transiently produce low levels of IFN-γ upon stimulation with Ag and B7-1, with production peaking at ∼8 h and declining by 24 h. The autocrine IFN-γ signals for upregulation of expression of T-bet and granzyme B and induces weak cytolytic activity and effector IFN-γ production. IFN-α acts synergistically with IFN-γ to support development of strong effector functions, whereas IL-12 induces high T-bet expression and strong function in the absence of IFN-γ signaling. Thus, IFN-γ is not only an important CD8 T cell effector cytokine, it is an autocrine/paracrine factor whose contributions to differentiation vary depending on whether the response is supported by IL-12 or type I IFN.


Assuntos
Comunicação Autócrina/imunologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Diferenciação Celular/imunologia , Interferon-alfa/fisiologia , Interferon gama/fisiologia , Transferência Adotiva/métodos , Animais , Comunicação Autócrina/genética , Linfócitos T CD8-Positivos/citologia , Diferenciação Celular/genética , Linhagem Celular Tumoral , Células Cultivadas , Interferon gama/deficiência , Interferon gama/genética , Interleucina-12/fisiologia , Melanoma Experimental/imunologia , Melanoma Experimental/metabolismo , Melanoma Experimental/patologia , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Comunicação Parácrina/genética , Comunicação Parácrina/imunologia , Proteínas com Domínio T/biossíntese , Proteínas com Domínio T/fisiologia , Regulação para Cima/genética , Regulação para Cima/imunologia
7.
Clin Cancer Res ; 17(10): 3134-45, 2011 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-21252159

RESUMO

PURPOSE: Tumor-released proangiogenic factors suppress endothelial adhesion molecule (EAM) expression and prevent leukocyte extravasation into the tumor. This is one reason why immunotherapy has met with limited success in the clinic. We hypothesized that overcoming EAM suppression with angiogenesis inhibitors would increase leukocyte extravasation and subsequently enhance the effectiveness of cellular immunotherapy. EXPERIMENTAL DESIGN: Intravital microscopy, multiple color flow cytometry, immunohistochemistry, and various tumor mouse (normal and T-cell deficient) models were used to investigate the temporal dynamics of cellular and molecular events that occur in the tumor microenvironment during tumor progression and angiostatic intervention. RESULTS: We report that while EAM levels and T-cell infiltration are highly attenuated early on in tumor growth, angiostatic therapy modulates these effects. In tumor models with normal and T-cell-deficient mice, we show the active involvement of the adaptive immune system in cancer and differentiate antiangiogenic effects from antiangiogenic mediated enhancement of immunoextravasation. Our results indicate that a compromised immune response in tumors can be obviated by the use of antiangiogenic agents. Finally, with adoptive transfer studies in mice, we show that a phased combination of angiostatic therapy and T-cell transfer significantly (P < 0.0013) improves tumor growth inhibition. CONCLUSIONS: This research contributes to understand the cellular mechanism of action of angiostatic agents and the immune response within the tumor microenvironment, in particular as a consequence of the temporal dynamics of EAM levels. Moreover, our results suggest that adjuvant therapy with angiogenesis inhibitors holds promise for cellular immunotherapy in the clinic.


Assuntos
Proteínas Angiostáticas/uso terapêutico , Imunidade Celular/efeitos dos fármacos , Imunoterapia Adotiva/métodos , Neoplasias/terapia , Linfócitos T/efeitos dos fármacos , Adjuvantes Imunológicos/uso terapêutico , Inibidores da Angiogênese/uso terapêutico , Animais , Células Cultivadas , Quimioterapia Adjuvante , Terapia Combinada , Feminino , Humanos , Imunidade Celular/fisiologia , Melanoma Experimental/tratamento farmacológico , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Nus , Neoplasias/tratamento farmacológico , Neoplasias/imunologia , Linfócitos T/imunologia , Linfócitos T/fisiologia , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/imunologia
8.
Curr Opin Immunol ; 22(3): 333-40, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20363604

RESUMO

CD8 T cells require a third signal, along with Ag and costimulation, to make a productive response and avoid death and/or tolerance induction. Recent studies indicate that IL-12 and Type I IFN (IFNalpha/beta) are the major sources of signal 3 in a variety of responses, and that the two cytokines stimulate a common regulatory program involving altered expression of about 350 genes. Signal 3-driven chromatin remodeling is likely to play a major role in this regulation. Although less well studied, there is emerging evidence that CD4 T cells may also require a 'third signal' for a productive response and that IL-1 can provide this signal. Signal 3 cytokines can replace adjuvants in supporting in vivo T cell responses to peptide and protein antigens, and a better understanding of their activities and mechanisms should contribute to more rational design of vaccines.


Assuntos
Citocinas/imunologia , Regulação da Expressão Gênica/imunologia , Inflamação/metabolismo , Ativação Linfocitária/imunologia , Transdução de Sinais , Linfócitos T/imunologia , Animais , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/imunologia , Citocinas/biossíntese , Humanos , Inflamação/imunologia , Camundongos , Linfócitos T/citologia
9.
J Immunol ; 183(3): 1695-704, 2009 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-19592655

RESUMO

A third signal that can be provided by IL-12 or type I IFN is required for differentiation of naive CD8 T cells responding to Ag and costimulation. The cytokines program development of function and memory within 3 days of initial stimulation, and we show here that programming involves regulation of a common set of approximately 355 genes including T-bet and eomesodermin. Much of the gene regulation program is initiated in response to Ag and costimulation within 24 h but is then extinguished unless a cytokine signal is available. Histone deacetylase inhibitors mimic the effects of IL-12 or type I IFN signaling, indicating that the cytokines relieve repression and allow continued gene expression by promoting increased histone acetylation. In support of this, increased association of acetylated histones with the promoter loci of granzyme B and eomesodermin is shown to occur in response to IL-12, IFN-alpha, or histone deacetylase inhibitors. Thus, IL-12 and IFN-alpha/beta enforce in common a complex gene regulation program that involves, at least in part, chromatin remodeling to allow sustained expression of a large number of genes critical for CD8 T cell function and memory.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Montagem e Desmontagem da Cromatina/imunologia , Regulação da Expressão Gênica/imunologia , Memória Imunológica/genética , Interferon Tipo I/fisiologia , Interleucina-12/fisiologia , Acetilação , Animais , Apresentação de Antígeno , Diferenciação Celular , Histonas/metabolismo , Camundongos , Proteínas com Domínio T/genética
10.
J Immunol ; 182(5): 2726-37, 2009 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-19234167

RESUMO

MHC-II presentation by dendritic cells (DC) is necessary both for initial priming of CD4 T cells and for induction of peripheral effector function. Although CD4 T cells can be critical for competent immunization-mediated cancer immunosurveillance, unmanipulated CD4 T cell responses to poorly immunogenic tumors result in either complete ignorance or tolerance induction, suggesting inadequate DC function. In this study, we investigated the phenotype, Ag uptake, and MHC-II presentation capacity of normal dermal DC and tumor-infiltrating DC (TIDC) in both lymphoid and peripheral sites. We found that murine tumors were extensively infiltrated by partially activated TIDC that closely resembled dermal DC by surface marker expression. However, in contrast to dermal DC, TIDC were inefficient at MHC-II presentation due to poor intrinsic protein uptake capability. This resulted in both inferior initiation of T cell responses in the draining lymph node and poor peripheral effector cell accumulation. In addition, TLR stimulation selectively enhanced MHC-II presentation of Ag by dermal DC, but not TIDC in the draining lymph node, and did not affect overall peripheral Ag uptake of either. These results show that TIDC are functionally distinct from normal interstitial DC, thus indicating that neoplastic tissues can evade effector CD4 T cells through modification of DC competence.


Assuntos
Apresentação de Antígeno/imunologia , Movimento Celular/imunologia , Células Dendríticas/imunologia , Células Dendríticas/patologia , Antígenos de Histocompatibilidade Classe II/imunologia , Antígenos de Histocompatibilidade Classe II/metabolismo , Melanoma Experimental/imunologia , Melanoma Experimental/patologia , Transferência Adotiva , Animais , Apresentação de Antígeno/genética , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Comunicação Celular/imunologia , Linhagem Celular Tumoral , Movimento Celular/genética , Células Dendríticas/transplante , Feminino , Antígenos de Histocompatibilidade Classe II/genética , Células de Langerhans/imunologia , Células de Langerhans/patologia , Células de Langerhans/transplante , Linfonodos/imunologia , Linfonodos/metabolismo , Linfonodos/patologia , Masculino , Melanoma Experimental/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Timoma/imunologia , Timoma/metabolismo , Timoma/patologia
11.
J Immunol ; 182(5): 2786-94, 2009 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-19234173

RESUMO

Inflammation can have both positive and negative effects on development of CD8 T cell memory, but the relative contributions and cellular targets of the cytokines involved are unclear. Using CD8 T cells lacking receptors for IL-12, type I IFN, or both, we show that these cytokines act directly on CD8 T cells to support memory formation in response to vaccinia virus and Listeria monocytogenes infections. Development of memory to vaccinia is supported predominantly by IL-12, whereas both IL-12 and type I IFN contribute to memory formation in response to Listeria. In contrast to memory formation, the inability to respond to IL-12 or type I IFN had a relatively small impact on the level of primary expansion, with at most a 3-fold reduction in the case of responses to Listeria. We further show that programming for memory development by IL-12 is complete within 3 days of the initial naive CD8 T cell response to Ag. This programming does not result in formation of a population that expresses killer cell lectin-like receptor G1, and the majority of the resulting memory cells have a CD62L(high) phenotype characteristic of central memory cells. Consistent with this, the cells undergo strong expansion upon rechallenge and provide protective immunity. These data demonstrate that IL-12 and type I IFN play an essential early role in determining whether Ag encounter by naive CD8 T cells results in formation of a protective memory population.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Memória Imunológica , Interferon Tipo I/fisiologia , Interleucina-12/fisiologia , Animais , Linfócitos T CD8-Positivos/transplante , Linhagem Celular , Células Cultivadas , Memória Imunológica/genética , Interferon Tipo I/metabolismo , Interleucina-12/metabolismo , Listeria monocytogenes/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Receptor de Interferon alfa e beta/deficiência , Receptor de Interferon alfa e beta/genética , Receptores de Interleucina-12/deficiência , Receptores de Interleucina-12/genética , Vaccinia virus/imunologia
12.
J Immunol ; 181(1): 155-64, 2008 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-18566380

RESUMO

Cancer immunosurveillance failure is largely attributed to insufficient activation signals and dominant inhibitory stimuli for tumor Ag (TAg)-specific CD8 T cells. CD4 T cells have been shown to license dendritic cells (DC), thereby having the potential for converting CD8 T cell responses from tolerance to activation. To understand the potential cooperation of TAg-specific CD4 and CD8 T cells, we have characterized the responses of naive TCR transgenic CD8 and CD4 T cells to poorly immunogenic murine tumors. We found that whereas CD8 T cells sensed TAg and were tolerized, the CD4 T cells remained ignorant throughout tumor growth and did not provide help. This disparity in responses was due to normal TAg MHC class I cross-presentation by immature CD8alpha+ DC in the draining lymph node, but poor MHC class II presentation on all DC subsets due to selective inhibition by the tumor microenvironment. Thus, these results reveal a novel mechanism of cancer immunosubversion, in which inhibition of MHC-II TAg presentation on DC prevents CD4 T cell priming, thereby blocking any potential for licensing CD8alpha+ DC and helping tolerized CD8 T cells.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Células Dendríticas/imunologia , Antígenos de Histocompatibilidade Classe II/imunologia , Neoplasias/imunologia , Animais , Antígenos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Linhagem Celular Tumoral , Citoplasma/imunologia , Linfonodos/imunologia , Camundongos , Camundongos Transgênicos , Transplante de Neoplasias , Neoplasias/patologia , Ovalbumina/imunologia , Ovalbumina/metabolismo
13.
Am J Clin Oncol ; 31(2): 173-81, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18391603

RESUMO

OBJECTIVE: To evaluate the safety and activity of large multivalent immunogen (LMI), prepared by immobilizing autologous tumor cell plasma membrane on 5-microm diameter silica beads, in patients with melanoma and renal cell carcinoma (RCC). METHODS: Thirty patients with stage IV metastatic melanoma and 31 patients with stage IV RCC were randomly assigned to 1 of 3 trial arms and received monthly treatment with (1) LMI alone, (2) cyclophosphamide followed 8 days later with LMI, or (3) the same treatment as in arm 2 with IL-2 given for 5 days beginning 1 week after LMI administration. RESULTS: No grade 4 toxicities were observed. For patients with melanoma, median overall survival time for all 30 patients was 20.4 months [95% confidence interval (CI): 8.0-not assessable], and median progression-free survival was 2.8 months (95% CI: 1.9-6.3). For patients with RCC, median overall survival exceeded 46.2 months (95% CI: 30.3-not assessable), and median progression-free survival was 12.2 months (95% CI: 4.6-not assessable). Two patients had a partial response to LMI treatment. CONCLUSIONS: Based on our results that demonstrate the safety and tolerability of LMI vaccine, further development of this therapy is warranted to evaluate its clinical efficacy.


Assuntos
Antígenos de Neoplasias/uso terapêutico , Vacinas Anticâncer/uso terapêutico , Carcinoma de Células Renais/terapia , Neoplasias Renais/terapia , Melanoma/terapia , Neoplasias Cutâneas/terapia , Adulto , Idoso , Idoso de 80 Anos ou mais , Antígenos de Neoplasias/efeitos adversos , Antígenos de Neoplasias/imunologia , Antineoplásicos/administração & dosagem , Vacinas Anticâncer/efeitos adversos , Vacinas Anticâncer/imunologia , Carcinoma de Células Renais/patologia , Terapia Combinada , Ciclofosfamida/administração & dosagem , Feminino , Humanos , Interleucina-2/administração & dosagem , Neoplasias Renais/patologia , Masculino , Melanoma/patologia , Pessoa de Meia-Idade , Análise Multivariada , Metástase Neoplásica , Neoplasias Cutâneas/patologia , Análise de Sobrevida
14.
J Immunol ; 180(4): 2107-16, 2008 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-18250416

RESUMO

The high-affinity chain of the IL-7 receptor, IL-7Ralpha (CD127), is expressed by effector CD8 T cells that have the capacity to become memory cells. IL-7Ralpha expression is uniformly high on naive CD8 T cells, and the majority of these cells down-regulate expression upon antigenic challenge. At the peak of expansion, the fraction of effectors expressing high IL-7Ralpha varies depending on the response examined. The signals that a CD8 T cell receives during a response to Ag that lead to altered expression of IL-7Ralpha have not been fully defined. In vitro experiments demonstrated that Ag alone is sufficient to down-regulate IL-7Ralpha on all cells and most of the cells rapidly re-express the receptor upon removal from Ag. Expression was not altered by the B7.1 costimulatory ligand or when IL-12 was present to provide the signal needed for development of effector functions, indicating that TCR engagement is sufficient to regulate IL-7Ralpha expression. Consistent with this, in vivo priming with peptide Ag resulted in IL-7Ralpha expression that inversely correlated with Ag levels, and expression levels were not changed when IL-12 or adjuvant were administered with Ag. A large fraction of the cells present at the peak of expansion had re-expressed IL-7Ralpha, but most of these cells failed to survive; those that did survive expressed high IL-7Ralpha levels. Thus, Ag-dependent signals regulate IL-7Ralpha levels on responding CD8 T cells, and this occurs whether the responding cells become fully activated or are rendered tolerant by administration of peptide Ag alone.


Assuntos
Antígenos/fisiologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Tolerância Imunológica , Ativação Linfocitária/imunologia , Receptores de Interleucina-7/biossíntese , Receptores de Interleucina-7/genética , Animais , Linfócitos T CD8-Positivos/microbiologia , Células Cultivadas , Células Clonais , Regulação para Baixo/genética , Regulação para Baixo/imunologia , Proteínas do Ovo/fisiologia , Tolerância Imunológica/genética , Memória Imunológica/genética , Listeria monocytogenes/imunologia , Ativação Linfocitária/genética , Camundongos , Camundongos Congênicos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Ovalbumina/fisiologia , Fragmentos de Peptídeos , Receptores de Interleucina-7/antagonistas & inibidores , Reprodutibilidade dos Testes , Fase de Repouso do Ciclo Celular/genética , Fase de Repouso do Ciclo Celular/imunologia
15.
J Exp Med ; 204(11): 2667-77, 2007 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-17954566

RESUMO

CD8 is critical for T cell recognition of peptide/class I major histocompatability complex ligands, yet is down-regulated during activation of CD8 T cells. We report that loss of CD8 expression early during in vivo responses to vaccinia virus or Listeria monocytogenes (LM) correlates with decreased T cell staining with specific class I/peptide tetramers and reduced CD8 T cell sensitivity for antigen. Loss of CD8 cell surface expression occurs despite sustained mRNA expression, and CD8 levels return to normal levels during differentiation of memory cells, indicating a transient effect. We determined that during response to LM, CD8 down-regulation is regulated by T cell reactivity to type I interferon (IFN-I) because CD8 loss was averted on IFN-I receptor-deficient T cells. IFN-I alone was not sufficient to drive CD8 down-regulation, however, as antigen was also required for CD8 loss. These results suggest that CD8 effector T cell differentiation involves a transient down-regulation of antigen sensitivity (CTL "detuning"), via reduced CD8 expression, a feature that may focus the effector response on target cells expressing high levels of antigen (e.g., infected cells), while limiting collateral damage to bystander cells.


Assuntos
Antígenos CD8/genética , Linfócitos T CD8-Positivos/imunologia , Ativação Linfocitária , Animais , Regulação para Baixo , Regulação da Expressão Gênica/imunologia , Humanos , Listeria monocytogenes/imunologia , Camundongos , Camundongos Transgênicos , Ovalbumina/genética , Vaccinia virus/imunologia
16.
Semin Cancer Biol ; 17(4): 299-308, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17656106

RESUMO

Naïve CD8 T cells respond to signals provided by Ag, costimulation and cytokines by proliferating and differentiating to develop effector functions. Following initial clonal expansion, however, the cells develop activation-induced non-responsiveness (AINR), a form of anergy characterized by an inability to produce IL-2. Cells in the AINR state can carry out effector functions (cytolysis, IFN-gamma production) but cannot continue to proliferate and expand in the face of persisting Ag. AINR limits the ability of activated CTL to control tumor growth but can be reversed by IL-2, provided either therapeutically or by activated CD4 T helper cells, to allow continued expansion.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Anergia Clonal/imunologia , Ativação Linfocitária/imunologia , Neoplasias/imunologia , Animais , Humanos , Tolerância Imunológica , Linfócitos T Citotóxicos
17.
Int Immunol ; 19(6): 733-43, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17545279

RESUMO

CD8 T cell responses to vaccinia virus (VV) and a virus-encoded ovalbumin peptide (OVAP) epitope were examined using adoptively transferred OT-I T cells. The results demonstrate that upon intra-peritoneal challenge with ovalbumin-expressing VV (VV-OVAP), OT-I T cell proliferation occurs initially in lymph nodes and spleens followed by migration of the divided cells to the peritoneal cavity. Massive clonal expansion occurs in response to both the virus and the virus-encoded ovalbumin (OVA) epitope, as demonstrated using low numbers of adoptively transferred cells, and the responding OT-I cells display marked site-dependent functional heterogeneity with respect to IFN-gamma and tumor necrosis factor-alpha (TNF-alpha) production and granzyme B expression. OT-I cells responding to VV-OVAP develop the capacity to produce IFN-gamma in response to antigen as they proliferate and differentiate. In marked contrast, naive OT-I cells rapidly produce TNF-alpha upon antigen recognition, and this capacity declines as the cells proliferate in response to the virus, suggesting that this potent inflammatory cytokine may be important primarily during initiation of the response. At the peak of clonal expansion, a large fraction (30-60%) of the OT-I cells responding to the virus express high IL-7Ralpha levels, and the majority of these cells is subsequently lost. While high IL-7Ralpha expression may be necessary for a CD8 T cell to transition to memory, it is clearly not sufficient. Thus, OT-I cells responding to VV infection exhibit a high degree of heterogeneity within the responding population that differs depending on their anatomical location, despite the specificity and affinity of the TCR being identical on all of the cells.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Linfonodos/imunologia , Baço/imunologia , Vaccinia virus/imunologia , Transferência Adotiva , Animais , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/transplante , Contagem de Células , Proliferação de Células , Proteínas do Ovo/genética , Proteínas do Ovo/imunologia , Granzimas/metabolismo , Interferon gama/metabolismo , Linfonodos/citologia , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Ovalbumina/genética , Ovalbumina/imunologia , Fragmentos de Peptídeos , Cavidade Peritoneal/citologia , Receptores de Antígenos de Linfócitos T/genética , Receptores de Interleucina-7/metabolismo , Baço/citologia , Antígenos Thy-1/genética , Fatores de Tempo , Fator de Necrose Tumoral alfa/metabolismo , Vacínia/imunologia , Vaccinia virus/genética
18.
J Immunol ; 178(12): 7640-8, 2007 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-17548600

RESUMO

IL-21, the most recently described member of the common gamma-chain cytokine family, is produced by activated CD4 T cells, whereas CD8 T cells express the IL-21 receptor. To investigate a possible role for IL-21 in the priming of naive CD8 T cells, we examined responses of highly purified naive OT-I CD8 T cells to artificial APCs displaying Ag and B7-1 on their surface. We found that IL-21 enhanced OT-I clonal expansion and supported development of cytotoxic effector function. High levels of IL-2 did not support development of effector functions, but IL-2 was required for optimal responses in the presence of IL-21. IL-12 and IFN-alpha have previously been shown to support naive CD8 T cell differentiation and acquisition of effector functions through a STAT4-dependent mechanism. Here, we show that IL-21 does not require STAT4 to stimulate development of cytolytic activity. Furthermore, IL-21 fails to induce IFN-gamma or IL-4 production and can partially block IL-12 induction of IFN-gamma production. CD8 T cells that differentiate in response to IL-21 have a distinct surface marker expression pattern and are characterized as CD44(high), PD-1(low), CD25(low), CD134(low), and CD137(low). Thus, IL-21 can provide a signal required by naive CD8 T cells to differentiate in response to Ag and costimulation, and the resulting effector cells represent a unique effector phenotype with highly effective cytolytic activity, but deficient capacity to secrete IFN-gamma.


Assuntos
Apresentação de Antígeno , Linfócitos T CD8-Positivos/imunologia , Citotoxicidade Imunológica , Interleucinas/fisiologia , Ativação Linfocitária , Animais , Células Apresentadoras de Antígenos/imunologia , Antígenos de Superfície/análise , Antígenos de Superfície/metabolismo , Antígeno B7-1/imunologia , Antígeno B7-1/farmacologia , Linfócitos T CD8-Positivos/efeitos dos fármacos , Diferenciação Celular , Membrana Celular , Interferon-alfa , Interferon gama/antagonistas & inibidores , Interferon gama/metabolismo , Interleucina-12/antagonistas & inibidores , Interleucina-2/metabolismo , Interleucinas/farmacologia , Camundongos , Camundongos Transgênicos , Fenótipo , Fator de Transcrição STAT4/metabolismo , Transdução de Sinais
19.
J Immunol ; 178(11): 6752-60, 2007 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-17513722

RESUMO

CD8 T cells need a third signal, along with Ag and costimulation, for effective survival and development of effector functions, and this can be provided by IL-12 or type I IFN. Adoptively transferred OT-I T cells, specific for H-2K(b) and OVA, encounter Ag in the draining lymph nodes of mice with the OVA-expressing E.G7 tumor growing at a s.c. site. The OT-I cells respond by undergoing limited clonal expansion and development of effector functions (granzyme B expression and IFN-gamma production), and they migrate to the tumor where they persist but fail to control tumor growth. In contrast, OT-I T cells deficient for both the IL-12 and type I IFN receptors expand only transiently and rapidly disappear. These results suggested that some signal 3 cytokine is available, but that it is insufficient to support a CTL response that can control tumor growth. Consistent with this, administration of IL-12 at day 10 of tumor growth resulted in a large and sustained expansion of wild-type OT-I cells with enhanced effector functions, and tumor growth was controlled. This did not occur when the OT-I cells lacked the IL-12 and type I IFN receptors, demonstrating that the therapeutic effect of IL-12 results from direct delivery of signal 3 to the CD8 T cells responding to tumor Ag in the signal 3-deficient environment of the tumor.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Citocinas/fisiologia , Timoma/imunologia , Transferência Adotiva , Animais , Apresentação de Antígeno/genética , Linfócitos T CD8-Positivos/patologia , Linhagem Celular Tumoral , Movimento Celular/genética , Movimento Celular/imunologia , Proliferação de Células , Citocinas/biossíntese , Imunofenotipagem , Interferon Tipo I/fisiologia , Interleucina-12/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neoplasias Peritoneais/imunologia , Neoplasias Peritoneais/patologia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/transplante , Timoma/patologia
20.
Semin Immunol ; 19(3): 153-61, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17382557

RESUMO

CD8 T cells specific for self-antigens are present in the peripheral lymphoid system and can contribute to autoimmunity or transplant rejection. Whether recognition of Ag leads to full activation, or to induction of tolerance, depends upon availability of cytokine at critical stages of the response. Signals provided by IL-12 and/or IFN-alpha/beta are required for activation of naïve CD8 T cells, and IL-2 is needed to sustain and further expand the effector cells if Ag persists. These critical signaling requirements provide new insights into the factors that regulate the CD8 T cell contributions to development of autoimmunity or rejection of transplants.


Assuntos
Antígenos CD28/fisiologia , Tolerância Imunológica , Imunidade Celular/imunologia , Ativação Linfocitária , Subpopulações de Linfócitos T/imunologia , Animais , Antígenos CD28/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA