Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cancer Immunol Immunother ; 70(12): 3525-3540, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33903974

RESUMO

Immune checkpoint inhibitors (ICIs) have emerged as promising therapies for the treatment of cancer. However, existing ICIs, namely PD-(L)1 and CTLA-4 inhibitors, generate durable responses only in a subset of patients. TIGIT is a co-inhibitory receptor and member of the DNAM-1 family of immune modulating proteins. We evaluated the prevalence of TIGIT and its cognate ligand, PVR (CD155), in human cancers by assessing their expression in a large set of solid tumors. TIGIT is expressed on CD4+ and CD8+ TILs and is upregulated in tumors compared to normal tissues. PVR is expressed on tumor cells and tumor-associated macrophages from multiple solid tumors. We explored the therapeutic potential of targeting TIGIT by generating COM902, a fully human anti-TIGIT hinge-stabilized IgG4 monoclonal antibody that binds specifically to human, cynomolgus monkey, and mouse TIGIT, and disrupts the binding of TIGIT with PVR. COM902, either alone or in combination with a PVRIG (COM701) or PD-1 inhibitor, enhances antigen-specific human T cell responses in-vitro. In-vivo, a mouse chimeric version of COM902 in combination with an anti-PVRIG or anti-PD-L1 antibody inhibited tumor growth and increased survival in two syngeneic mouse tumor models. In summary, COM902 enhances anti-tumor immune responses and is a promising candidate for the treatment of advanced malignancies.


Assuntos
Anticorpos Monoclonais/imunologia , Antígeno B7-H1/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Receptores de Superfície Celular/imunologia , Receptores Imunológicos/imunologia , Transdução de Sinais/imunologia , Animais , Linhagem Celular Tumoral , Proliferação de Células/fisiologia , Feminino , Humanos , Imunoglobulina G/imunologia , Imunoterapia/métodos , Células Jurkat , Macaca fascicularis , Camundongos , Camundongos Endogâmicos BALB C
2.
Biochim Biophys Acta ; 1865(1): 72-82, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26241169

RESUMO

Immunotherapy is emerging as a promising anti-cancer curative modality. However, in contrast to recent advances obtained employing checkpoint blockade agents and T cell therapies, clinical efficacy of therapeutic cancer vaccines is still limited. Most vaccination attempts in the clinic represent "off-the shelf" approaches since they target common "self" tumor antigens, shared among different patients. In contrast, personalized approaches of vaccination are tailor-made for each patient and in spite being laborious, hold great potential. Recent technical advancement enabled the first steps in the clinic of personalized vaccines that target patient-specific mutated neo-antigens. Such vaccines could induce enhanced tumor-specific immune response since neo-antigens are mutation-derived antigens that can be recognized by high affinity T cells, not limited by central tolerance. Alternatively, the use of personalized vaccines based on whole autologous tumor cells, overcome the need for the identification of specific tumor antigens. Whole autologous tumor cells could be administered alone, pulsed on dendritic cells as lysate, DNA, RNA or delivered to dendritic cells in-vivo through encapsulation in nanoparticle vehicles. Such vaccines may provide a source for the full repertoire of the patient-specific tumor antigens, including its private neo-antigens. Furthermore, combining next-generation personalized vaccination with other immunotherapy modalities might be the key for achieving significant therapeutic outcome.


Assuntos
Imunoterapia Ativa , Neoplasias/terapia , Medicina de Precisão , Antígenos de Neoplasias/imunologia , Autoantígenos/imunologia , Vacinas Anticâncer/imunologia , Células Dendríticas/imunologia , Humanos , Nanopartículas
3.
Blood ; 121(15): 3033-40, 2013 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-23446736

RESUMO

Generation of T cells endowed with graft-versus-leukemia (GVL) and depleted of graft-versus-host (GVH) activity represents a highly desirable goal in bone marrow transplantation (BMT). Here, we demonstrate that donor anti-third-party CD8 T cells with central memory phenotype (Tcm) exhibit marked GVL reactivity through a unique T-cell receptor-independent mechanism. Thus, in a residual disease mouse model, Tcm therapy following autologous BMT led to significant survival prolongation, with 30% to 40% of the treated mice displaying long-term tumor-free survival. A more impressive finding was that infusion of donor Tcm in an allogeneic model rapidly eliminated residual lymphoma cells and led to long-term survival of 100% in the absence of GVH disease. Collectively, the strong GVL reactivity of anti-third-party Tcm, coupled with their demonstrated enhancement of bone marrow allografting, suggests that the use of Tcm therapy in conjunction with allogeneic T-cell-depleted BMT could be of particular benefit in patients with B-cell malignancies who cannot tolerate intensive myeloablative conditioning.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Memória Imunológica/imunologia , Linfoma/imunologia , Neoplasia Residual/imunologia , Animais , Apoptose/imunologia , Linfócitos B/imunologia , Linfócitos B/metabolismo , Transplante de Medula Óssea/métodos , Linfócitos T CD8-Positivos/metabolismo , Linhagem Celular Tumoral , Citotoxicidade Imunológica/imunologia , Feminino , Citometria de Fluxo , Doença Enxerto-Hospedeiro/imunologia , Efeito Enxerto vs Leucemia/imunologia , Molécula 1 de Adesão Intercelular/imunologia , Molécula 1 de Adesão Intercelular/metabolismo , Antígeno-1 Associado à Função Linfocitária/imunologia , Antígeno-1 Associado à Função Linfocitária/metabolismo , Linfoma/metabolismo , Linfoma/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Nus , Camundongos Transgênicos , Fatores de Tempo , Transplante Homólogo
4.
Blood ; 121(7): 1220-8, 2013 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-23223359

RESUMO

Transplantation of T cell-depleted BM (TDBM) under mild conditioning, associated with minimal toxicity and reduced risk of GVHD, offers an attractive therapeutic option for patients with nonmalignant hematologic disorders and can mediate immune tolerance to subsequent organ transplantation. However, overcoming TDBM rejection after reduced conditioning remains a challenge. Here, we address this barrier using donorderived central memory CD8(+) T cells (Tcms), directed against third-party antigens. Our results show that fully allogeneic or (hostXdonor)F1-Tcm, support donor chimerism (> 6 months) in sublethally irradiated (5.5Gy) mice, without GVHD symptoms. Chimerism under yet lower irradiation (4.5Gy) was achieved by combining Tcm with short-term administration of low-dose Rapamycin. Importantly, this chimerism resulted in successful donor skin acceptance, whereas third-party skin was rejected. Tracking of host anti-donor T cells (HADTCs), that mediate TDBMT rejection, in a novel bioluminescence-imaging model revealed that Tcms both induce accumulation and eradicate HADTCs in the LNs,concomitant with their elimination from other organs, including the BM. Further analysis with 2-photon microcopy revealed that Tcms form conjugates with HADTCs, resulting in decelerated and confined movement of HADTCs within the LNs in an antigen-specific manner. Thus, anti-third-party Tcms support TDBMT engraftment under reduced-conditioning through lymph-node sequestration and deletion of HADTCs, offering a novel and potentially safe approach for attaining stable hematopoietic chimerism.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Quimeras de Transplante/imunologia , Condicionamento Pré-Transplante/métodos , Animais , Transplante de Medula Óssea/imunologia , Feminino , Doença Enxerto-Hospedeiro/imunologia , Doença Enxerto-Hospedeiro/prevenção & controle , Doenças Hematológicas/imunologia , Doenças Hematológicas/terapia , Humanos , Memória Imunológica , Imunossupressores/administração & dosagem , Isoantígenos , Linfonodos/imunologia , Depleção Linfocítica , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Nus , Camundongos Transgênicos , Sirolimo/administração & dosagem , Transplante de Pele/imunologia , Linfócitos T/imunologia , Doadores de Tecidos
5.
Cancer Immunol Res ; 12(7): 876-890, 2024 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-38752503

RESUMO

Cancers that are poorly immune infiltrated pose a substantial challenge, with current immunotherapies yielding limited clinical success. Stem-like memory T cells (TSCM) have been identified as a subgroup of T cells that possess strong proliferative capacity and that can expand and differentiate following interactions with dendritic cells (DCs). In this study, we explored the pattern of expression of a recently discovered inhibitory receptor poliovirus receptor-related immunoglobulin domain protein (PVRIG) and its ligand, poliovirus receptor-related ligand 2 (PVRL2), in the human tumor microenvironment. Using spatial and single-cell RNA transcriptomics data across diverse cancer indications, we found that among the T-cell checkpoints, PVRIG is uniquely expressed on TSCM and PVRL2 is expressed on DCs in immune aggregate niches in tumors. PVRIG blockade could therefore enhance TSCM-DC interactions and efficiently drive T-cell infiltration to tumors. Consistent with these data, following PVRIG blockade in patients with poorly infiltrated tumors, we observed immune modulation including increased tumor T-cell infiltration, T-cell receptor (TCR) clonality, and intratumoral T-cell expansion, all of which were associated with clinical benefit. These data suggest PVRIG blockade as a promising strategy to induce potent antitumor T-cell responses, providing a novel approach to overcome resistance to immunotherapy in immune-excluded tumors.


Assuntos
Células Dendríticas , Neoplasias , Microambiente Tumoral , Humanos , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Microambiente Tumoral/imunologia , Neoplasias/imunologia , Neoplasias/terapia , Neoplasias/metabolismo , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/metabolismo , Células T de Memória/imunologia , Células T de Memória/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo
6.
Cancer Immunol Res ; 12(6): 687-703, 2024 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-38592331

RESUMO

Recombinant cytokines have limited anticancer efficacy mostly due to a narrow therapeutic window and systemic adverse effects. IL18 is an inflammasome-induced proinflammatory cytokine, which enhances T- and NK-cell activity and stimulates IFNγ production. The activity of IL18 is naturally blocked by a high-affinity endogenous binding protein (IL18BP). IL18BP is induced in the tumor microenvironment (TME) in response to IFNγ upregulation in a negative feedback mechanism. In this study, we found that IL18 is upregulated in the TME compared with the periphery across multiple human tumors and most of it is bound to IL18BP. Bound IL18 levels were largely above the amount required for T-cell activation in vitro, implying that releasing IL18 in the TME could lead to potent T-cell activation. To restore the activity of endogenous IL18, we generated COM503, a high-affinity anti-IL18BP that blocks the IL18BP:IL18 interaction and displaces precomplexed IL18, thereby enhancing T- and NK-cell activation. In vivo, administration of a surrogate anti-IL18BP, either alone or in combination with anti-PD-L1, resulted in significant tumor growth inhibition and increased survival across multiple mouse tumor models. Moreover, the anti-IL18BP induced pronounced TME-localized immune modulation including an increase in polyfunctional nonexhausted T- and NK-cell numbers and activation. In contrast, no increase in inflammatory cytokines and lymphocyte numbers or activation state was observed in serum and spleen. Taken together, blocking IL18BP using an Ab is a promising approach to harness cytokine biology for the treatment of cancer.


Assuntos
Interleucina-18 , Microambiente Tumoral , Animais , Humanos , Interleucina-18/metabolismo , Camundongos , Microambiente Tumoral/imunologia , Microambiente Tumoral/efeitos dos fármacos , Linhagem Celular Tumoral , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/metabolismo , Neoplasias/imunologia , Neoplasias/tratamento farmacológico , Ativação Linfocitária/imunologia , Ativação Linfocitária/efeitos dos fármacos , Feminino , Camundongos Endogâmicos C57BL , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto , Linfócitos T/imunologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/metabolismo
7.
Blood ; 117(3): 1042-52, 2011 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-21045195

RESUMO

Cytotoxic T lymphocytes (CTLs) suppress T cell responses directed against their antigens regardless of their own T cell receptor (TCR) specificity. This makes the use of CTLs promising for tolerance induction in autoimmunity and transplantation. It has been established that binding of the CTL CD8 molecule to the major histocompatibility complex (MHC) class I α3 domain of the recognizing T cell must be permitted for death of the latter cell to ensue. However, the signaling events triggered in the CTL by this molecular interaction in the absence of TCR recognition have never been clarified. Here we use single-cell imaging to study the events occurring in CTLs serving as targets for recognition by specific T cells. We demonstrate that CTLs actively respond to recognition by polarizing their cytotoxic granules to the contact area, releasing their lethal cargo, and vigorously proliferating. Using CTLs from perforin knockout (KO) mice and lymphocyte specific kinase (Lck) knockdown with specific small interfering RNA (siRNA), we show that the killing of the recognizing CD8 T cell is perforin dependent and is initiated by Lck signaling in the CTL. Collectively, these data suggest a novel mechanism in which the entire cascade generally triggered by TCR engagement is "hijacked" in CTLs serving as targets for T cell recognition without TCR ligation.


Assuntos
Grânulos Citoplasmáticos/imunologia , Ativação Linfocitária/imunologia , Linfócitos T Citotóxicos/imunologia , Linfócitos T/imunologia , Sequência de Aminoácidos , Animais , Grânulos Citoplasmáticos/metabolismo , Citotoxicidade Imunológica/efeitos dos fármacos , Citotoxicidade Imunológica/imunologia , Ácido Egtázico/análogos & derivados , Ácido Egtázico/farmacologia , Citometria de Fluxo , Proteína Tirosina Quinase p56(lck) Linfócito-Específica/genética , Proteína Tirosina Quinase p56(lck) Linfócito-Específica/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos DBA , Camundongos Endogâmicos , Camundongos Knockout , Modelos Imunológicos , Ovalbumina/farmacologia , Fragmentos de Peptídeos/farmacologia , Perforina/genética , Perforina/metabolismo , Interferência de RNA , Linfócitos T/metabolismo , Linfócitos T Citotóxicos/metabolismo
8.
J Immunol ; 187(4): 2006-14, 2011 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-21753148

RESUMO

We previously demonstrated that anti-third-party CTLs (stimulated under IL-2 deprivation against cells with an MHC class I [MHC-I] background different from that of the host and the donor) are depleted of graft-versus-host reactivity and can eradicate B cell chronic lymphocytic leukemia cells in vitro or in an HU/SCID mouse model. We demonstrated in the current study that human allogeneic or autologous anti-third-party CTLs can also efficiently eradicate primary non-Hodgkin B cell lymphoma by inducing slow apoptosis of the pathological cells. Using MHC-I mutant cell line as target cells, which are unrecognizable by the CTL TCR, we demonstrated directly that this killing is TCR independent. Strikingly, this unique TCR-independent killing is induced through lymphoma MHC-I engagement. We further showed that this killing mechanism begins with durable conjugate formation between the CTLs and the tumor cells, through rapid binding of tumor ICAM-1 to the CTL LFA-1 molecule. This conjugation is followed by a slower second step of MHC-I-dependent apoptosis, requiring the binding of the MHC-I α2/3 C region on tumor cells to the CTL CD8 molecule for killing to ensue. By comparing CTL-mediated killing of Daudi lymphoma cells (lacking surface MHC-I expression) to Daudi cells with reconstituted surface MHC-I, we demonstrated directly for the first time to our knowledge, in vitro and in vivo, a novel role for MHC-I in the induction of lymphoma cell apoptosis by CTLs. Additionally, by using different knockout and transgenic strains, we further showed that mouse anti-third-party CTLs also kill lymphoma cells using similar unique TCR-independence mechanism as human CTLs, while sparing normal naive B cells.


Assuntos
Apoptose/imunologia , Antígenos CD8/imunologia , Antígenos de Histocompatibilidade Classe I/imunologia , Imunidade Celular , Linfoma de Células B/imunologia , Linfócitos T Citotóxicos/imunologia , Animais , Apoptose/genética , Linfócitos B/imunologia , Linfócitos B/patologia , Antígenos CD8/genética , Linhagem Celular Tumoral , Feminino , Antígenos de Histocompatibilidade Classe I/genética , Humanos , Molécula 1 de Adesão Intercelular/genética , Molécula 1 de Adesão Intercelular/imunologia , Antígeno-1 Associado à Função Linfocitária/genética , Antígeno-1 Associado à Função Linfocitária/imunologia , Linfoma de Células B/genética , Linfoma de Células B/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Camundongos SCID , Mutação , Linfócitos T Citotóxicos/patologia
9.
Blood ; 115(10): 2095-104, 2010 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-20042725

RESUMO

Enabling engraftment of allogeneic T cell-depleted bone marrow (TDBM) under reduced-intensity conditioning represents a major challenge in bone marrow transplantation (BMT). Anti-third-party cytotoxic T lymphocytes (CTLs) were previously shown to be endowed with marked ability to delete host antidonor T cells in vitro, but were found to be less effective in vivo. This could result from diminished lymph node (LN) homing caused by the prolonged activation, which induces a CD44(+)CD62L(-) effector phenotype, and thereby prevents effective colocalization with, and neutralization of, alloreactive host T cells (HTCs). In the present study, LN homing, determined by imaging, was enhanced upon culture conditions that favor the acquisition of CD44(+)CD62L(+) central memory cell (Tcm) phenotype by anti-third-party CD8(+) cells. These Tcm-like cells displayed strong proliferation and prolonged persistence in BM transplant recipients. Importantly, adoptively transferred HTCs bearing a transgenic T-cell receptor (TCR) with antidonor specificity were efficiently deleted only by donor-type Tcms. All these attributes were found to be associated with improved efficacy in overcoming T cell-mediated rejection of TDBM, thereby enabling high survival rate and long-term donor chimerism, without causing graft-versus-host disease. In conclusion, anti-third-party Tcms, which home to recipient LNs and effectively delete antidonor T cells, could provide an effective and novel tool for overcoming rejection of BM allografts.


Assuntos
Transplante de Medula Óssea/métodos , Linfócitos T CD8-Positivos/transplante , Doença Enxerto-Hospedeiro/prevenção & controle , Memória Imunológica/fisiologia , Tolerância ao Transplante/imunologia , Animais , Transplante de Medula Óssea/imunologia , Linfócitos T CD8-Positivos/imunologia , Células Cultivadas , Quimiotaxia de Leucócito/fisiologia , Feminino , Doença Enxerto-Hospedeiro/imunologia , Memória Imunológica/imunologia , Linfonodos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Nus , Doadores de Tecidos , Condicionamento Pré-Transplante/métodos , Transplante Homólogo , Regulação para Cima/imunologia
10.
Cancer Discov ; 11(5): 1040-1051, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33687987

RESUMO

Therapeutic antibodies targeting the CTLA4/PD-1 pathways have revolutionized cancer immunotherapy by eliciting durable remission in patients with cancer. However, relapse following early response, attributable to primary and adaptive resistance, is frequently observed. Additional immunomodulatory pathways are being studied in patients with primary or acquired resistance to CTLA4 or PD-1 blockade. The DNAM1 axis is a potent coregulator of innate and adaptive immunity whose other components include the immunoglobulin receptors TIGIT, PVRIG, and CD96, and their nectin and nectin-like ligands. We review the basic biology and therapeutic relevance of this family, which has begun to show promise in cancer clinical trials. SIGNIFICANCE: Recent studies have outlined the immuno-oncologic ascendancy of coinhibitory receptors in the DNAM1 axis such as TIGIT and PVRIG and, to a lesser extent, CD96. Biological elucidation backed by ongoing clinical trials of single-agent therapy directed against TIGIT or PVRIG is beginning to provide the rationale for testing combination regimens of DNAM1 axis blockers in conjunction with anti-PD-1/PD-L1 agents.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Antígenos de Diferenciação de Linfócitos T/metabolismo , Neoplasias/tratamento farmacológico , Receptores Imunológicos/metabolismo , Humanos , Imunoterapia
11.
Cancer Immunol Res ; 7(2): 244-256, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30659055

RESUMO

A limitation to antitumor immunity is the dysfunction of T cells in the tumor microenvironment, in part due to upregulation of coinhibitory receptors such as PD-1. Here, we describe that poliovirus receptor-related immunoglobulin domain protein (PVRIG) acts as a coinhibitory receptor in mice. Murine PVRIG interacted weakly with poliovirus receptor (PVR) but bound poliovirus receptor-like 2 (PVRL2) strongly, making the latter its principal ligand. As in humans, murine NK and NKT cells constitutively expressed PVRIG. However, when compared with humans, less PVRIG transcript and surface protein was detected in murine CD8+ T cells ex vivo However, activated CD8+ T cells upregulated PVRIG expression. In the mouse tumor microenvironment, infiltrating CD8+ T cells expressed PVRIG whereas its ligand, PVRL2, was detected predominantly on myeloid cells and tumor cells, mirroring the expression pattern in human tumors. PVRIG-deficient mouse CD8+ T cells mounted a stronger antigen-specific effector response compared with wild-type CD8+ T cells during acute Listeria monocytogenes infection. Furthermore, enhanced CD8+ T-cell effector function inhibited tumor growth in PVRIG-/- mice compared with wild-type mice and PD-L1 blockade conferred a synergistic antitumor response in PVRIG-/- mice. Therapeutic intervention with antagonistic anti-PVRIG in combination with anti-PD-L1 reduced tumor growth. Taken together, our results suggest PVRIG is an inducible checkpoint receptor and that targeting PVRIG-PVRL2 interactions results in increased CD8+ T-cell function and reduced tumor growth.See related article on p. 257.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Neoplasias/imunologia , Neoplasias/metabolismo , Receptores de Superfície Celular/metabolismo , Animais , Antígeno B7-H1/antagonistas & inibidores , Biomarcadores , Linhagem Celular Tumoral , Modelos Animais de Doenças , Feminino , Humanos , Ativação Linfocitária/genética , Ativação Linfocitária/imunologia , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/metabolismo , Camundongos , Camundongos Knockout , Neoplasias/patologia , Interferência de RNA , Especificidade do Receptor de Antígeno de Linfócitos T/imunologia , Carga Tumoral , Microambiente Tumoral/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto
12.
Cancer Immunol Res ; 7(2): 257-268, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30659054

RESUMO

Although checkpoint inhibitors that block CTLA-4 and PD-1 have improved cancer immunotherapies, targeting additional checkpoint receptors may be required to broaden patient response to immunotherapy. PVRIG is a coinhibitory receptor of the DNAM/TIGIT/CD96 nectin family that binds to PVRL2. We report that antagonism of PVRIG and TIGIT, but not CD96, increased CD8+ T-cell cytokine production and cytotoxic activity. The inhibitory effect of PVRL2 was mediated by PVRIG and not TIGIT, demonstrating that the PVRIG-PVRL2 pathway is a nonredundant signaling node. A combination of PVRIG blockade with TIGIT or PD-1 blockade further increased T-cell activation. In human tumors, PVRIG expression on T cells was increased relative to normal tissue and trended with TIGIT and PD-1 expression. Tumor cells coexpressing PVR and PVRL2 were observed in multiple tumor types, with highest coexpression in endometrial cancers. Tumor cells expressing either PVR or PVRL2 were also present in numbers that varied with the cancer type, with ovarian cancers having the highest percentage of PVR-PVRL2+ tumor cells and colorectal cancers having the highest percentage of PVR+PVRL2- cells. To demonstrate a role of PVRIG and TIGIT on tumor-derived T cells, we examined the effect of PVRIG and TIGIT blockade on human tumor-infiltrating lymphocytes. For some donors, blockade of PVRIG increased T-cell function, an effect enhanced by combination with TIGIT or PD-1 blockade. In summary, we demonstrate that PVRIG and PVRL2 are expressed in human cancers and the PVRIG-PVRL2 and TIGIT-PVR pathways are nonredundant inhibitory signaling pathways.See related article on p. 244.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Nectinas/metabolismo , Neoplasias/imunologia , Neoplasias/metabolismo , Receptores de Superfície Celular/metabolismo , Animais , Regulação Neoplásica da Expressão Gênica , Humanos , Ativação Linfocitária/imunologia , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/metabolismo , Linfócitos do Interstício Tumoral/patologia , Camundongos , Neoplasias/genética , Neoplasias/patologia , Receptor de Morte Celular Programada 1/genética , Receptor de Morte Celular Programada 1/metabolismo , Ligação Proteica , Receptores Imunológicos/genética , Receptores Imunológicos/metabolismo , Transdução de Sinais
13.
Sci Transl Med ; 10(436)2018 04 11.
Artigo em Inglês | MEDLINE | ID: mdl-29643231

RESUMO

We conducted a pilot clinical trial testing a personalized vaccine generated by autologous dendritic cells (DCs) pulsed with oxidized autologous whole-tumor cell lysate (OCDC), which was injected intranodally in platinum-treated, immunotherapy-naïve, recurrent ovarian cancer patients. OCDC was administered alone (cohort 1, n = 5), in combination with bevacizumab (cohort 2, n = 10), or bevacizumab plus low-dose intravenous cyclophosphamide (cohort 3, n = 10) until disease progression or vaccine exhaustion. A total of 392 vaccine doses were administered without serious adverse events. Vaccination induced T cell responses to autologous tumor antigen, which were associated with significantly prolonged survival. Vaccination also amplified T cell responses against mutated neoepitopes derived from nonsynonymous somatic tumor mutations, and this included priming of T cells against previously unrecognized neoepitopes, as well as novel T cell clones of markedly higher avidity against previously recognized neoepitopes. We conclude that the use of oxidized whole-tumor lysate DC vaccine is safe and effective in eliciting a broad antitumor immunity, including private neoantigens, and warrants further clinical testing.


Assuntos
Vacinas Anticâncer/uso terapêutico , Imunoterapia/métodos , Neoplasias Ovarianas/terapia , Antígenos de Neoplasias/imunologia , Bevacizumab/uso terapêutico , Ciclofosfamida/uso terapêutico , Células Dendríticas/metabolismo , Feminino , Humanos , Mutação/genética , Neoplasias Ovarianas/tratamento farmacológico , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/metabolismo
14.
Neuron ; 81(1): 179-94, 2014 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-24361077

RESUMO

Perceptual decisions involve distributed cortical activity. Does information flow sequentially from one cortical area to another, or do networks of interconnected areas contribute at the same time? Here we delineate when and how activity in specific areas drives a whisker-based decision in mice. A short-term memory component temporally separated tactile "sensation" and "action" (licking). Using optogenetic inhibition (spatial resolution, 2 mm; temporal resolution, 100 ms), we surveyed the neocortex for regions driving behavior during specific behavioral epochs. Barrel cortex was critical for sensation. During the short-term memory, unilateral inhibition of anterior lateral motor cortex biased responses to the ipsilateral side. Consistently, barrel cortex showed stimulus-specific activity during sensation, whereas motor cortex showed choice-specific preparatory activity and movement-related activity, consistent with roles in motor planning and movement. These results suggest serial information flow from sensory to motor areas during perceptual decision making.


Assuntos
Vias Aferentes/fisiologia , Córtex Cerebral/fisiologia , Tomada de Decisões/fisiologia , Tato/fisiologia , Potenciais de Ação/fisiologia , Animais , Córtex Cerebral/citologia , Channelrhodopsins , Condicionamento Operante/fisiologia , Discriminação Psicológica/fisiologia , Feminino , Proteínas de Fluorescência Verde/genética , Masculino , Camundongos , Camundongos Transgênicos , Percepção de Movimento , Rede Nervosa/fisiologia , Neurônios/fisiologia , Estimulação Luminosa , Proteínas Vesiculares de Transporte de Aminoácidos Inibidores/genética , Vibrissas/inervação
15.
PLoS One ; 9(2): e88678, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24520413

RESUMO

The mouse is an increasingly prominent model for the analysis of mammalian neuronal circuits. Neural circuits ultimately have to be probed during behaviors that engage the circuits. Linking circuit dynamics to behavior requires precise control of sensory stimuli and measurement of body movements. Head-fixation has been used for behavioral research, particularly in non-human primates, to facilitate precise stimulus control, behavioral monitoring and neural recording. However, choice-based, perceptual decision tasks by head-fixed mice have only recently been introduced. Training mice relies on motivating mice using water restriction. Here we describe procedures for head-fixation, water restriction and behavioral training for head-fixed mice, with a focus on active, whisker-based tactile behaviors. In these experiments mice had restricted access to water (typically 1 ml/day). After ten days of water restriction, body weight stabilized at approximately 80% of initial weight. At that point mice were trained to discriminate sensory stimuli using operant conditioning. Head-fixed mice reported stimuli by licking in go/no-go tasks and also using a forced choice paradigm using a dual lickport. In some cases mice learned to discriminate sensory stimuli in a few trials within the first behavioral session. Delay epochs lasting a second or more were used to separate sensation (e.g. tactile exploration) and action (i.e. licking). Mice performed a variety of perceptual decision tasks with high performance for hundreds of trials per behavioral session. Up to four months of continuous water restriction showed no adverse health effects. Behavioral performance correlated with the degree of water restriction, supporting the importance of controlling access to water. These behavioral paradigms can be combined with cellular resolution imaging, random access photostimulation, and whole cell recordings.


Assuntos
Comportamento Animal/fisiologia , Etologia/métodos , Animais , Peso Corporal , Discriminação Psicológica , Cabeça , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Recompensa , Sacarose , Análise e Desempenho de Tarefas , Água , Privação de Água
16.
Front Immunol ; 3: 93, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22566971

RESUMO

The induction of immune tolerance by specific agents, as opposed to general immune suppression, is a most desirable goal in transplantation biology. One approach to attain this goal is afforded by the use of donor-derived cells endowed with veto activity, which is the ability of a cell to specifically suppress only T cells directed against its antigens. A megadose of purified veto CD34(+) hematopoietic stem cells is already used in patients to allow hematopoietic stem cells transplantation (HSCT) across major genetic barriers, while avoiding severe graft versus host disease (GVHD). However, allowing engraftment of such T cell-depleted HSCT under safer reduced intensity conditioning (RIC) protocols still remains a challenge. Therefore, combining megadose of CD34(+) HSCT with other GVHD-depleted veto cells could enable facilitation of engraftment of HSCT under RIC without the adverse complication of GVHD. This approach might provide a safer modality for enabling engraftment of HSCT, enabling its application in elderly patients who cannot tolerate intensive protocols and to a variety of patients with non-malignant disorders, associated with longer life expectancy, in whom the use of a high risk conditioning cannot be considered.

17.
Best Pract Res Clin Haematol ; 24(3): 393-401, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21925092

RESUMO

Haploidentical hematopoietic stem cell transplantation (HSCT) offers the advantage of readily available family member donors for nearly all patients. A 'megadose' of purified CD34+ hematopoietic stem cells is used to overcome the host's residual immunity surviving the myeloablative conditioning, while avoiding severe GVHD. However, the number of CD34+ cells that can be harvested is insufficient for overcoming the large numbers of host T cells remaining after reduced intensity conditioning (RIC). Therefore, combining a 'megadose' of CD34+ HSCT with other tolerizing cells could potentially support and promote successful engraftment of haploidentical purified stem cell transplantation under a safer RIC. One approach to address this challenge could be afforded by using Donor CD8 T cells directed against 3rd-party stimulators, bearing an ex-vivo induced central memory phenotype (Tcm). These Tcm cells,depleted of GVH reactivity, were shown to be highly efficient in overcoming host T cells mediated rejection and in promoting fully mismatched bone-marrow (BM) engraftment, in HSCT murine models. This is likely due to the marked lymph node homing of the Tcm, their strong proliferative capacity and prolonged persistence in BM transplant recipients. Thus, combining anti 3rd-party Tcm cell therapy with a 'megadose' of purified CD34+ stem cells, could offer a safer RIC protocol for attaining hematopoietic chimerism in patients with hematological diseases and as a platform for organ transplantation or cell therapy in cancer patients.


Assuntos
Linfócitos T CD8-Positivos/transplante , Transplante de Células-Tronco Hematopoéticas , Memória Imunológica , Imunoterapia Adotiva/métodos , Condicionamento Pré-Transplante/métodos , Tolerância ao Transplante/imunologia , Animais , Linfócitos T CD8-Positivos/imunologia , Modelos Animais de Doenças , Feminino , Doença Enxerto-Hospedeiro/imunologia , Doença Enxerto-Hospedeiro/prevenção & controle , Humanos , Masculino , Camundongos , Doadores de Tecidos , Transplante Homólogo
18.
Int Immunopharmacol ; 9(6): 694-700, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19150657

RESUMO

The use of hematopoietic stem cell transplantation (HSCT) for the establishment of mixed chimerism represents a viable and attractive approach for generating tolerance in transplantation biology, as it generally leads to durable immune tolerance, enabling the subsequent engraftment of organ transplants without the need for a deleterious continuous immunosuppressive therapy. However, in order to apply HSCT to patients in a manner that enables long term survival, transplant-related mortality must be minimized by eliminating the risk for graft-versus-host-disease (GVHD) and by reducing the toxicity of the conditioning protocol. T-cell depleted bone marrow transplants (TDBMT) have been shown to adequately eliminate GVHD. However, even in leukemia patients undergoing supralethal conditioning, mismatched TDBMT are vigorously rejected. This barrier can be overcome through the modulatory activity of CD34 cells, which are endowed with veto activity, by the use of megadose stem cell transplants. In mice, megadoses of Sca+lin-hematopoietic stem cells can induce mixed chimerism following sub-lethal conditioning. Nevertheless, the number of human CD34 cells that can be harvested is not likely to be sufficient to overcome rejection under reduced intensity conditioning (RIC), which might be acceptable in recipients of organ transplantation. To address this challenge, we investigated a novel source of veto cells, namely anti 3rd-party cytotoxic T cells (CTLs) which are depleted of GVH reactivity, combined with megadoses of purified stem cells and a RIC protocol. This approach might provide a safer modality for the induction of durable chimerism.


Assuntos
Doença Enxerto-Hospedeiro/imunologia , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/imunologia , Condicionamento Pré-Transplante/métodos , Tolerância ao Transplante , Animais , Doença Enxerto-Hospedeiro/prevenção & controle , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Linfócitos T Citotóxicos/imunologia
19.
J Biol Chem ; 281(39): 28958-63, 2006 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-16864573

RESUMO

Gap junctions form intercellular channels that mediate metabolic and electrical signaling between neighboring cells in a tissue. Lack of an atomic resolution structure of the gap junction has made it difficult to identify interactions that stabilize its transmembrane domain. Using a recently computed model of this domain, which specifies the locations of each amino acid, we postulated the existence of several interactions and tested them experimentally. We introduced mutations within the transmembrane domain of the gap junction-forming protein connexin that were previously implicated in genetic diseases and that apparently destabilized the gap junction, as evidenced here by the absence of the protein from the sites of cell-cell apposition. The model structure helped identify positions on adjacent helices where second-site mutations restored membrane localization, revealing possible interactions between residue pairs. We thus identified two putative salt bridges and one pair involved in packing interactions in which one disease-causing mutation suppressed the effects of another. These results seem to reveal some of the physical forces that underlie the structural stability of the gap junction transmembrane domain and suggest that abrogation of such interactions bring about some of the effects of disease-causing mutations.


Assuntos
Conexinas/genética , Junções Comunicantes , Mutação , Comunicação Celular , Conexina 26 , Células HeLa , Humanos , Modelos Moleculares , Conformação Molecular , Conformação Proteica , Estrutura Terciária de Proteína , Transdução de Sinais , Transfecção , Proteína beta-1 de Junções Comunicantes
20.
Proc Natl Acad Sci U S A ; 103(31): 11784-9, 2006 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-16864778

RESUMO

Alzheimer's disease (AD) is characterized by plaque formation, neuronal loss, and cognitive decline. The functions of the local and systemic immune response in this disease are still controversial. Using AD double-transgenic (APP/PS1) mice, we show that a T cell-based vaccination with glatiramer acetate, given according to a specific regimen, resulted in decreased plaque formation and induction of neurogenesis. It also reduced cognitive decline, assessed by performance in a Morris water maze. The vaccination apparently exerted its effect by causing a phenotype switch in brain microglia to dendritic-like (CD11c) cells producing insulin-like growth factor 1. In vitro findings showed that microglia activated by aggregated beta-amyloid, and characterized as CD11b(+)/CD11c(-)/MHC class II(-)/TNF-alpha(+) cells, impeded neurogenesis from adult neural stem/progenitor cells, whereas CD11b(+)/CD11c(+)/MHC class II(+)/TNF-alpha(-) microglia, a phenotype induced by IL-4, counteracted the adverse beta-amyloid-induced effect. These results suggest that dendritic-like microglia, by facilitating the necessary adjustment, might contribute significantly to the brain's resistance to AD and argue against the use of antiinflammatory drugs.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/imunologia , Diferenciação Celular/fisiologia , Imunossupressores/uso terapêutico , Fator de Crescimento Insulin-Like I/metabolismo , Microglia , Peptídeos/uso terapêutico , Doença de Alzheimer/patologia , Doença de Alzheimer/fisiopatologia , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Antígeno CD11b/metabolismo , Antígeno CD11c/metabolismo , Genes MHC da Classe II , Acetato de Glatiramer , Hipocampo/citologia , Hipocampo/metabolismo , Imunossupressores/administração & dosagem , Interleucina-4/metabolismo , Aprendizagem em Labirinto , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Transgênicos , Microglia/citologia , Microglia/fisiologia , Peptídeos/administração & dosagem , Fenótipo , Placa Amiloide/metabolismo , Placa Amiloide/patologia , Presenilina-1 , Linfócitos T/fisiologia , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA