Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Immunotherapy ; 7(5): 545-61, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26065479

RESUMO

Chimeric antigen receptor (CAR) T-cell therapy has recently come into the spotlight due to impressive results in patients with B-cell acute lymphoblastic leukemia. By targeting CD19, a marker expressed most B-cell tumors, as well as normal B cells, CAR T-cell therapy has been investigated as a treatment strategy for B-cell leukemia and lymphoma. This review will discuss the successes of this therapy for the treatment of B-cell acute lymphoblastic leukemia and the challenges to this therapeutic strategy. We will also discuss application of CAR T-cell therapy to chronic lymphocytic leukemia and other B-cell malignancies including a follicular lymphoma, diffuse large B-cell lymphoma, as well as acute and plasma cell malignancies.


Assuntos
Neoplasias Hematológicas/terapia , Imunoterapia Adotiva/métodos , Neoplasias de Plasmócitos/terapia , Leucemia-Linfoma Linfoblástico de Células Precursoras B/terapia , Receptores de Antígenos de Linfócitos T/imunologia , Proteínas Recombinantes de Fusão/imunologia , Linfócitos T/transplante , Animais , Neoplasias Hematológicas/imunologia , Neoplasias Hematológicas/patologia , Humanos , Neoplasias de Plasmócitos/imunologia , Neoplasias de Plasmócitos/patologia , Leucemia-Linfoma Linfoblástico de Células Precursoras B/imunologia , Leucemia-Linfoma Linfoblástico de Células Precursoras B/patologia , Receptores de Antígenos de Linfócitos T/genética , Proteínas Recombinantes de Fusão/genética , Linfócitos T/imunologia , Linfócitos T/patologia
2.
Mol Ther ; 23(4): 769-78, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25582824

RESUMO

Adoptive cell therapy with genetically modified T cells expressing a chimeric antigen receptor (CAR) is a promising therapy for patients with B-cell acute lymphoblastic leukemia. However, CAR-modified T cells (CAR T cells) have mostly failed in patients with solid tumors or low-grade B-cell malignancies including chronic lymphocytic leukemia with bulky lymph node involvement. Herein, we enhance the antitumor efficacy of CAR T cells through the constitutive expression of CD40 ligand (CD40L, CD154). T cells genetically modified to constitutively express CD40L (CD40L-modified T cells) demonstrated increased proliferation and secretion of proinflammatory TH1 cytokines. Further, CD40L-modified T cells augmented the immunogenicity of CD40(+) tumor cells by the upregulated surface expression of costimulatory molecules (CD80 and CD86), adhesion molecules (CD54, CD58, and CD70), human leukocyte antigen (HLA) molecules (Class I and HLA-DR), and the Fas-death receptor (CD95). Additionally, CD40L-modified T cells induced maturation and secretion of the proinflammatory cytokine interleukin-12 by monocyte-derived dendritic cells. Finally, tumor-targeted CD19-specific CAR/CD40L T cells exhibited increased cytotoxicity against CD40(+) tumors and extended the survival of tumor-bearing mice in a xenotransplant model of CD19(+) systemic lymphoma. This preclinical data supports the clinical application of CAR T cells additionally modified to constitutively express CD40L with anticipated enhanced antitumor efficacy.


Assuntos
Ligante de CD40/metabolismo , Imunoterapia , Linfoma Folicular/terapia , Proteínas Recombinantes de Fusão/metabolismo , Linfócitos T/imunologia , Animais , Linhagem Celular Tumoral , Xenoenxertos , Humanos , Imunofenotipagem , Linfoma Folicular/imunologia , Camundongos
3.
Leuk Res ; 38(8): 948-54, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24934848

RESUMO

In order to stimulate antigen presentation and T cell activity against cancer, we treated three different tumor models in mice with the monoclonal antibodies anti-CD40 plus anti-CD137 (BiMab). In a subcutaneous transplantable MC38 colon cancer model, there was significant enhancement in the survival of mice following BiMab treatment. Anti-CD40 has shown considerable success against lymphoma in previous studies by other investigators, and we also showed in this study that, in a model of Eµ-Myc lymphoma, there was a statistically significant enhancement of survival of mice following BiMab treatment. Following the success of the BiMab treatment in the previous two models, we wished to determine if it would be successful in a mouse model of multiple myeloma. Firstly, we tested a transplantable model of disease in which multiple myeloma cells derived from Vk*MYC mice were injected intravenously. A minor proportion of anti-CD137 and BiMab treated mice experienced prolongation of life beyond 250 days. Then we tested the therapy in a spontaneously occurring multiple myeloma model, in Vk*MYC transgenic mice. The majority of mice treated survived longer than control mice, although statistical significance was not demonstrated.


Assuntos
Anticorpos/uso terapêutico , Antígenos CD40/antagonistas & inibidores , Linfoma/terapia , Mieloma Múltiplo/terapia , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/antagonistas & inibidores , Animais , Antígenos CD40/imunologia , Transformação Celular Neoplásica/genética , Modelos Animais de Doenças , Feminino , Genes myc , Imunoterapia/métodos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Células Tumorais Cultivadas , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/imunologia
4.
PLoS One ; 9(5): e95847, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24788789

RESUMO

We have found previously that the tumor cell lines, Renca (a renal cancer) and MC38 (a colon tumor) which had been injected subcutaneously in mice, could be successfully treated with a combination therapy of an oligodeoxynucleotide (CpG1826) (injected intratumorally) and anti-CD137 antibody (injected intraperitoneally). Thus the combination treatment was expected to initiate a "danger" signal via TLR9 on immune cells, and the anti-CD137 was expected to further activate T cells. In the present study, we found that several other tumor types injected subcutaneously could also be successfully treated with this combination therapy. In addition, we wished to determine if the treatment could work as effectively in an orthotopic metastatic model, which is more physiologically relevant to cancer in humans. Renca was selected as we were familiar with injecting this orthotopically into the outer cortex of the kidney in mice, and it spontaneously metastasizes to lung and abdominal sites. We tested various routes of delivery of CpG combined with intraperitoneal delivery of anti-CD137. Orthotopic tumors were injected with CpG intratumorally, using ultrasound-guided delivery on multiple occasions, combined with anti-CD137 intraperitoneally. A reduction in primary tumor size was observed following intratumoral injection of CpG compared to other treatments. We found that there was a statistically significant increase in survival of mice with orthotopic Renca tumor following intratumoral injection of CpG. However, we determined that the most effective route of delivery of CpG was intravenous, which led to further significantly enhanced survival of mice when combined with anti-CD137 intraperitoneally, likely due to inhibition of metastatic disease. Our data supports future development of this combination therapy for cancer.


Assuntos
Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/farmacologia , Neoplasias Renais/terapia , Oligodesoxirribonucleotídeos/administração & dosagem , Oligodesoxirribonucleotídeos/farmacologia , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/imunologia , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/uso terapêutico , Linhagem Celular Tumoral , Vias de Administração de Medicamentos , Interações Medicamentosas , Feminino , Humanos , Neoplasias Renais/patologia , Camundongos , Oligodesoxirribonucleotídeos/uso terapêutico , Análise de Sobrevida , Carga Tumoral/efeitos dos fármacos
5.
Cancer J ; 20(2): 127-33, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24667958

RESUMO

CD19-targeted chimeric antigen receptor (CAR) T cells are currently being tested in the clinic with very promising outcomes. However, limitations to CAR T cell therapy exist. These include lack of efficacy against some tumors, specific targeting of tumor cells without affecting normal tissue and retaining activity within the suppressive tumor microenvironment. Whereas promising clinical trials are in progress, preclinical development is focused on optimizing CAR design, to generate "armored CAR T cells," which are protected from the inhibitory tumor microenvironment. Studies investigating the expression of cytokine transgenes, combination therapy with small molecule inhibitors, or monoclonal antibodies, are aimed at improving the antitumor efficacy of CAR T cell therapy. Other strategies aimed at improving CAR T cell therapy include using dual CARs and chemokine receptors to more specifically target tumor cells. This review will describe the current clinical data and some novel armored CAR T cell approaches for improving antitumor efficacy therapy.


Assuntos
Antígenos CD28/uso terapêutico , Imunoterapia Adotiva , Receptores de Antígenos de Linfócitos T/uso terapêutico , Linfócitos T/imunologia , Antígenos CD28/imunologia , Humanos , Receptores de Antígenos de Linfócitos T/imunologia
6.
Int J Hematol ; 99(4): 361-71, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24311149

RESUMO

The genetic modification of autologous T cells with chimeric antigen receptors (CARs) represents a breakthrough for gene engineering as a cancer therapy for hematologic malignancies. By targeting the CD19 antigen, we have demonstrated robust and rapid anti-leukemia activity in patients with heavily pre-treated and chemotherapy-refractory B cell acute lymphoblastic leukemia (B-ALL). We demonstrated rapid induction of deep molecular remissions in adults, which has been recently confirmed in a case report involving a child with B-ALL. In contrast to the results when treating B-ALL, outcomes have been more modest in patients with chronic lymphocytic leukemia (CLL) or other non-hodgkin's lymphoma (NHL). We review the clinical trial experience targeting B-ALL and CLL and speculate on the possible reasons for the different outcomes and propose potential optimization to CAR T cell therapy when targeting CLL or other indolent NHL. Lastly, we discuss the pre-clinical development and potential for clinical translation for using CAR T cells against multiple myeloma and acute myeloid leukemia. We highlight the potential risks and benefits by targeting these poor outcome hematologic malignancies.


Assuntos
Neoplasias Hematológicas/imunologia , Neoplasias Hematológicas/terapia , Imunoterapia Adotiva , Receptores de Antígenos/imunologia , Proteínas Recombinantes de Fusão/imunologia , Linfócitos T/imunologia , Animais , Antígenos CD19/imunologia , Antígenos CD19/metabolismo , Terapia Genética , Neoplasias Hematológicas/genética , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/imunologia , Leucemia Mieloide Aguda/terapia , Mieloma Múltiplo/genética , Mieloma Múltiplo/imunologia , Mieloma Múltiplo/terapia , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras B/imunologia , Leucemia-Linfoma Linfoblástico de Células Precursoras B/terapia , Receptores de Antígenos/genética , Receptores de Antígenos/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Linfócitos T/metabolismo
7.
Mol Ther ; 22(1): 18-27, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24048441

RESUMO

The tumor microenvironment can promote tumor growth and reduce treatment efficacy. Tumors can occur in many sites in the body, but how surrounding normal tissues at different anatomical sites affect tumor microenvironments and their subsequent response to therapy is not known.We demonstrated that tumors from renal, colon, or prostate cell lines in orthotopic locations responded to immunotherapy consisting of three agonist antibodies, termed Tri-mAb, to a much lesser extent than the same tumor type located subcutaneously. A tissue-specific response to Tri-mAb was confirmed by ex vivo separation of subcutaneous (SC) or orthotopic tumor cells from stromal cells, followed by reinjection of tumor cells into the opposite site. Compared with SC tumors, orthotopic tumors had a microenvironment associated with a type 2 immune response, related to immunosuppression, and an involvement of alternatively activated macrophages in the kidney model. Orthotopic kidney tumors were more highly vascularized than SC tumors. Neutralizing the macrophage- and Th2-associated molecules chemokine (C-C motif) ligand 2 or interleukin-13 led to a significantly improved therapeutic effect. This study highlights the importance of the tissue of implantation in sculpting the tumor microenvironment. These are important fundamental issues in tumor biology and crucial factors to consider in the design of experimental models and treatment strategies.


Assuntos
Imunoterapia , Neoplasias/imunologia , Neoplasias/patologia , Microambiente Tumoral/imunologia , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacologia , Antígenos CD40/antagonistas & inibidores , Antígenos CD40/imunologia , Linhagem Celular Tumoral , Quimiocina CCL2/imunologia , Neoplasias do Colo/imunologia , Modelos Animais de Doenças , Expressão Gênica , Interleucina-13/imunologia , Neoplasias Renais/imunologia , Neoplasias Renais/metabolismo , Neoplasias Renais/patologia , Macrófagos/imunologia , Macrófagos/metabolismo , Masculino , Camundongos , Neoplasias/mortalidade , Neoplasias/terapia , Neovascularização Patológica/imunologia , Especificidade de Órgãos/imunologia , Próstata/imunologia , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/antagonistas & inibidores , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/imunologia , Resultado do Tratamento , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/antagonistas & inibidores , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/imunologia
8.
Blood ; 119(18): 4133-41, 2012 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-22354001

RESUMO

Adoptive cell therapy with tumor-targeted T cells is a promising approach to cancer therapy. Enhanced clinical outcome using this approach requires conditioning regimens with total body irradiation, lymphodepleting chemotherapy, and/or additional cytokine support. However, the need for prior conditioning precludes optimal application of this approach to a significant number of cancer patients intolerant to these regimens. Herein, we present preclinical studies demonstrating that treatment with CD19-specific, chimeric antigen receptor (CAR)-modified T cells that are further modified to constitutively secrete IL-12 are able to safely eradicate established disease in the absence of prior conditioning. We demonstrate in a novel syngeneic tumor model that tumor elimination requires both CD4(+) and CD8(+) T-cell subsets, autocrine IL-12 stimulation, and subsequent IFNγ secretion by the CAR(+) T cells. Importantly, IL-12-secreting, tumor-targeted T cells acquire intrinsic resistance to T regulatory cell-mediated inhibition. Based on these preclinical data, we anticipate that adoptive therapy using CAR-targeted T cells modified to secrete IL-12 will obviate or reduce the need for potentially hazardous conditioning regimens to achieve optimal antitumor responses in cancer patients.


Assuntos
Imunoterapia Adotiva , Interleucina-12/metabolismo , Subpopulações de Linfócitos T/transplante , Timoma/terapia , Neoplasias do Timo/terapia , Condicionamento Pré-Transplante , Animais , Antígenos CD19/genética , Antígenos CD19/imunologia , Antineoplásicos Alquilantes/uso terapêutico , Linfócitos B/efeitos dos fármacos , Antígeno B7-1/genética , Terapia Combinada , Ciclofosfamida/uso terapêutico , Citotoxicidade Imunológica , Humanos , Interferon gama/sangue , Interleucina-12/sangue , Interleucina-12/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Proteínas Recombinantes de Fusão/genética , Especificidade do Receptor de Antígeno de Linfócitos T , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/imunologia , Timoma/tratamento farmacológico , Timoma/imunologia , Neoplasias do Timo/tratamento farmacológico , Neoplasias do Timo/imunologia , Transplante Isogênico
9.
J Gene Med ; 14(6): 405-15, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22262649

RESUMO

BACKGROUND: The genetic engineering of T cells through the introduction of a chimeric antigen receptor (CAR) allows for generation of tumor-targeted T cells. Once expressed by T cells, CARs combine antigen-specificity with T cell activation in a single fusion molecule. Most CARs are comprised of an antigen-binding domain, an extracellular spacer/hinge region, a trans-membrane domain and an intracellular signaling domain resulting in T cell activation after antigen binding. METHODS: We performed a search of the literature regarding tumor immunotherapy using CAR-modified T cells to provide a concise review of this topic. RESULTS: This review aims to focus on the elements of CAR design required for successful application of this technology in cancer immunotherapy. Most notably, proper target antigen selection, co-stimulatory signaling, and the ability of CAR-modified T cells to traffic, persist and retain function after adoptive transfer are required for optimal tumor eradication. Furthermore, recent clinical trials have demonstrated tumor burden and chemotherapy conditioning before adoptive transfer as being critically important for this therapy. Future research into counteracting the suppressive tumor microenvironment and the ability to activate an endogenous anti-tumor response by CAR-modified T cells may enhance the therapeutic potential of this treatment. CONCLUSIONS: In conclusion, CAR-modified T cell therapy is a highly promising treatment for cancer, having already demonstrated both promising preclinical and clinical results. However, further modification and additional clinical trials will need to be conducted to ultimately optimize the anti-tumor efficacy of this approach.


Assuntos
Antígenos de Neoplasias/genética , Neoplasias/terapia , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/imunologia , Linfócitos T/imunologia , Transferência Adotiva , Técnicas de Transferência de Genes , Genes Codificadores dos Receptores de Linfócitos T , Engenharia Genética , Terapia Genética , Humanos , Imunoterapia Adotiva , Ativação Linfocitária , Neoplasias/imunologia , Receptores de Antígenos de Linfócitos T/biossíntese , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Transdução de Sinais
10.
Cancer Res ; 71(8): 2871-81, 2011 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-21487038

RESUMO

Human T cells genetically modified to express chimeric antigen receptors (CAR) specific to the B cell tumor antigen CD19 can successfully eradicate systemic human CD19(+) tumors in immunocompromised SCID (severe combined immunodeficient)-Beige mice. However, in the clinical setting, CD4(+) CD25(hi) T regulatory cells (Treg) present within the tumor microenvironment may be potent suppressors of tumor-targeted effector T cells. In order to assess the impact of Tregs on CAR-modified T cells in the SCID-Beige xenotransplant model, we isolated, genetically targeted and expanded natural T regulatory cells (nTreg). In vitro nTregs modified to express CD19-targeted CARs efficiently inhibited the proliferation of activated human T cells, as well as the capacity of CD19-targeted 19-28z(+) effector T cells to lyse CD19(+) Raji tumor cells. Intravenous infusion of CD19-targeted nTregs into SCID-Beige mice with systemic Raji tumors traffic to sites of tumor and recapitulate a clinically relevant hostile tumor microenvironment. Antitumor efficacy of subsequently infused 19-28z(+) effector T cells was fully abrogated as assessed by long-term survival of treated mice. Optimal suppression by genetically targeted nTregs was dependent on nTreg to effector T-cell ratios and in vivo nTreg activation. Prior infusion of cyclophosphamide in the setting of this nTreg-mediated hostile microenvironment was able to restore the antitumor activity of subsequently infused 19-28z(+) effector T cells through the eradication of tumor-targeted nTregs. These findings have significant implications for the design of future clinical trials utilizing CAR-based adoptive T-cell therapies of cancer.


Assuntos
Antígenos CD19/imunologia , Linfoma de Burkitt/terapia , Linfócitos T CD8-Positivos/imunologia , Imunoterapia Adotiva/métodos , Receptores de Antígenos/imunologia , Linfócitos T Reguladores/imunologia , Animais , Linfoma de Burkitt/tratamento farmacológico , Linfoma de Burkitt/imunologia , Linhagem Celular Tumoral , Terapia Combinada , Ciclofosfamida/farmacologia , Citotoxicidade Imunológica , Humanos , Ativação Linfocitária , Linfócitos do Interstício Tumoral/imunologia , Camundongos , Camundongos SCID , Células NIH 3T3 , Receptores de Antígenos/biossíntese , Receptores de Antígenos/genética , Retroviridae/genética , Transdução Genética , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Cancer Immunol Immunother ; 60(5): 671-83, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21327636

RESUMO

Toll-like receptor (TLR) agonists can trigger broad inflammatory responses that elicit rapid innate immunity and promote the activities of lymphocytes, which can potentially enhance adoptive immunotherapy in the tumor-bearing setting. In the present study, we found that Polyinosinic:Polycytidylic Acid [Poly(I:C)] and CpG oligodeoxynucleotide 1826 [CpG], agonists for TLR 3 and 9, respectively, potently activated adoptively transferred T cells against a murine model of established melanoma. Intratumoral injection of Poly(I:C) and CpG, combined with systemic transfer of activated pmel-1 T cells, specific for gp100(25-33), led to enhanced survival and eradication of 9-day established subcutaneous B16F10 melanomas in a proportion of mice. A series of survival studies in knockout mice supported a key mechanistic pathway, whereby TLR agonists acted via host cells to enhance IFN-γ production by adoptively transferred T cells. IFN-γ, in turn, enhanced the immunogenicity of the B16F10 melanoma line, leading to increased killing by adoptively transferred T cells. Thus, this combination approach counteracted tumor escape from immunotherapy via downregulation of immunogenicity. In conclusion, TLR agonists may represent advanced adjuvants within the setting of adoptive T-cell immunotherapy of cancer and hold promise as a safe means of enhancing this approach within the clinic.


Assuntos
Imunoterapia Adotiva , Melanoma Experimental/terapia , Oligodesoxirribonucleotídeos/uso terapêutico , Poli I-C/uso terapêutico , Linfócitos T/imunologia , Receptores Toll-Like/agonistas , Adjuvantes Imunológicos/administração & dosagem , Animais , Linhagem Celular Tumoral , Células Dendríticas/imunologia , Citometria de Fluxo , Inflamação , Interferon gama/biossíntese , Ativação Linfocitária , Melanoma Experimental/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores Toll-Like/imunologia
12.
Leuk Res ; 35(1): 14-21, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20719383

RESUMO

Allogeneic hematopoietic stem cell transplantation (HSCT) for leukemia can play a major role in reducing the risk of relapse by inducing a graft versus leukemia (GVL) effect. Here, we review the effectiveness of mismatching inhibitory killer-cell-immunoglobulin-like receptors (KIR) on donor natural killer (NK) cells as a mechanism for GVL. We review the range of KIR and the importance of T cell and NK cell content of the graft, together with considerations of the graft source. Further understanding of conditioning and mechanisms to reduce graft versus host disease (GVHD) will improve our ability to manipulate NK cells in HSCT.


Assuntos
Transplante de Células-Tronco Hematopoéticas , Células Matadoras Naturais/imunologia , Doença Enxerto-Hospedeiro/prevenção & controle , Humanos , Receptores KIR/imunologia
13.
Immunol Cell Biol ; 89(2): 216-24, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20567250

RESUMO

Natural killer (NK) cells are potent immune effector cells that can respond to infection and cancer, as well as allowing maternal adaptation to pregnancy. In response to malignant transformation or pathogenic invasion, NK cells can secrete cytokine and may be directly cytolytic, as well as exerting effects indirectly through other cells of the immune system. To recognize and respond to inflamed or infected tissues, NK cells express a variety of activating and inhibitory receptors including NKG2D, Ly49 or KIR, CD94-NKG2 heterodimers and natural cytotoxicity receptors, as well as co-stimulatory receptors. These receptors recognize cellular stress ligands as well as major histocompatibility complex class I and related molecules, which can lead to NK cell responses. Importantly, NK cells must remain tolerant of healthy tissue, and some of these receptors can also prevent activation of NK cells. In this review, we describe the expression of prominent NK cell receptors, as well as expression of their ligands and their role in immune responses. In addition, we describe the main signaling pathways used by NK cell receptors. Although we now appreciate that NK cell biology is more complicated than first thought, there are still facets of their biology that remain unclear. These will be highlighted and discussed in this review.


Assuntos
Células Matadoras Naturais/imunologia , Ativação Linfocitária/imunologia , Receptores de Células Matadoras Naturais/imunologia , Animais , Citotoxicidade Imunológica , Humanos , Multimerização Proteica
14.
Cancer Immunol Immunother ; 59(8): 1235-46, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20376439

RESUMO

Natural killer (NK) cells represent a promising cell type to utilize for effective adoptive immunotherapy. However, little is known about the important cytolytic molecules and signaling pathways used by NK cells in the adoptive transfer setting. To address this issue, we developed a novel mouse model to investigate the trafficking and mechanism of action of these cells. We demonstrate that methylcholanthrene-induced RKIK sarcoma cells were susceptible to NK cell-mediated lysis in vitro and in vivo following adoptive transfer of NK cells in C57BL/6 RAG-2(-/-)gammac(-/-) mice. Cytotoxic molecules perforin, granzymes B and M as well as the death ligand TRAIL and pro-inflammatory cytokine IFN-gamma were found to be important in the anti-tumor effect mediated by adoptively transferred NK cells. Importantly, we demonstrate that adoptively transferred NK cells could traffic to the tumor site and persisted in vivo which correlated with the anti-tumor effect observed. Overall, the results of this study have important implications for enhancing NK cell-based immunotherapies.


Assuntos
Citotoxicidade Imunológica , Imunoterapia Adotiva , Células Matadoras Naturais/imunologia , Ativação Linfocitária , Sarcoma Experimental/imunologia , Sarcoma Experimental/terapia , Animais , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/genética , Interleucina-2/imunologia , Interleucina-2/metabolismo , Células Matadoras Naturais/metabolismo , Células Matadoras Naturais/patologia , Metilcolantreno , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Mutantes , Sarcoma Experimental/patologia
15.
J Transl Med ; 8: 42, 2010 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-20426873

RESUMO

BACKGROUND: Combination immunotherapies can be effective against subcutaneous tumors in mice but the effect against orthotopic malignant disease is less well characterized. In particular, a combination of three agonist antibodies, termed Tri-mAb, consisting of anti-DR5, anti-CD40 and anti-CD137 has previously been demonstrated to eradicate a large proportion of subcutaneous renal cell carcinoma (Renca) tumors (75% long-term survival), but the effect against orthotopic disease is not known. PURPOSE: To determine the relative response of orthotopic tumors, we inoculated Renca into the kidney followed by treatment with Tri-mAb. RESULTS: We found that orthotopic tumors responded much less to treatment (approximately 13% survival), but a significant improvement in survival was achieved through the addition of IL-2 to the treatment regimen (55% survival). All three agonist antibodies and high dose IL-2, 100,000 IU for up to six doses, were required. CD8+ T cells were also required for optimal anti-tumor responses. Coadministration of IL-2 led to enhanced T cell activity as demonstrated by an increased frequency of IFN-gamma-producing T cells in tumor-draining lymph nodes, which may have contributed to the observed improvement of therapy against kidney tumors. IMPLICATIONS: Responses of subcutaneous tumors to immunotherapy do not necessarily reflect how orthotopic tumors respond. The use of combination immunotherapy stimulating multiple facets of immunity and including cytokine support for T cells can induce effective anti-tumor responses against orthotopic and metastatic tumors.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Anticorpos Antineoplásicos/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Interleucina-2/administração & dosagem , Interleucina-2/uso terapêutico , Neoplasias Renais/tratamento farmacológico , Animais , Anticorpos Monoclonais/farmacologia , Anticorpos Antineoplásicos/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/imunologia , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Memória Imunológica/efeitos dos fármacos , Memória Imunológica/imunologia , Interferon gama/biossíntese , Neoplasias Renais/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Análise de Sobrevida
16.
Expert Opin Biol Ther ; 10(4): 531-45, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20132063

RESUMO

IMPORTANCE OF THE FIELD: Conventional therapies, including surgery, chemotherapy and radiotherapy have contributed much to cancer treatment. However, these treatment modalities fail in a large proportion of patients, and there is a great need for effective alternate therapies. Adoptive immunotherapy can be effective against some cancers that have failed all other treatment options, even when disease burdens are massive. AREAS COVERED IN THIS REVIEW: This review gives a brief introduction of the historical origins of adoptive immunotherapy and then provides details of strategies for increasing the potency of cell transfer. Approaches for enhancing adoptive immunotherapy include: selecting the right type of cell; providing cytokine support; preconditioning patients and tuning the tumor microenvironment. The review also provides insights into the safety, feasibility and costs of this form of therapy. WHAT THE READER WILL GAIN: This article will give the reader an appreciation of the potential of adoptive immunotherapy, as well as an understanding of some limitations and current approaches for optimizing the effectiveness of this approach. TAKE HOME MESSAGE: With recent developments in knowledge of the interactions between the immune system and tumors, the field of adoptive immunotherapy is now poised to make dramatic contributions to cancer therapy.


Assuntos
Imunoterapia Adotiva , Neoplasias/terapia , Antineoplásicos/uso terapêutico , Terapia Combinada , Citocinas/administração & dosagem , Humanos , Neoplasias/tratamento farmacológico , Condicionamento Pré-Transplante
17.
Clin Cancer Res ; 15(24): 7624-7633, 2009 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-19996209

RESUMO

PURPOSE: To determine the antitumor activity of a novel combination of two immunomodulatory agents that simultaneously direct multiple components of immunity against cancer. EXPERIMENTAL DESIGN: We combined the Toll-like receptor agonist CpG 1826 with a T-cell costimulatory antibody specific for CD137 in an optimal treatment route and dosing schedule against established tumors in two mouse models. Mechanistic insight was gained using gene-deficient mice and cell-depleting antibodies. RESULTS: The combination was shown to eradicate tumors in a large proportion of mice. Crucial roles for CD8(+) T cells, natural killer cells, and IFNs were shown. CpG and anti-CD137 injection led to activation of dendritic cells and optimal expansion of activated T cells in the blood. Macrophages were not necessary for therapeutic effect, and indeed depletion of macrophages in vivo enhanced therapy leading to tumor rejection in 100% of mice, which has not been previously reported in the immunotherapeutic setting. Long-term surviving mice were resistant to tumor rechallenge, demonstrating immunologic memory. In addition, we show, for the first time, that mice lacking B cells have a total loss of a recall response against tumor, suggesting a role for B cells in the induction of antitumor immunologic memory. CONCLUSION: This study provides support for the use of a novel combination of immunomodulatory agents stimulating multiple facets of immunity for the effective immunotherapy of cancer. (Clin Cancer Res 2009;15(24):7624-33).

18.
Immunotherapy ; 1(4): 623-30, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20635990

RESUMO

Immunotherapy of cancer is a rapidly developing field; one such development is the manipulation and use of natural killer (NK) cells. These cells with 'killer instincts' are an attractive cell to utilize, as they are directly reactive toward tumor and could potentially activate the endogenous adaptive immune system. Their employment in adoptive cell transfer treatments has yielded important results and discoveries, although effective antitumor responses are limited. To address these limitations, NK cells are the target of a new generation of immunotherapy involving gene transfer. The gene modification of immune cells is a relatively recent technique and some groups have targeted NK cells for gene modification to improve their antitumor efficacy. This review will investigate studies describing the gene modification of NK cells and their encouraging antitumor effects.


Assuntos
Citocinas/metabolismo , Imunoterapia Adotiva , Células Matadoras Naturais/metabolismo , Receptores de Fatores de Crescimento/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Animais , Antígenos CD/genética , Antígenos CD/imunologia , Antígenos CD/metabolismo , Citocinas/genética , Citocinas/imunologia , Citotoxicidade Imunológica/genética , Técnicas de Transferência de Genes , Humanos , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/patologia , Ativação Linfocitária/genética , Engenharia de Proteínas , Receptores de Fatores de Crescimento/genética , Receptores de Fatores de Crescimento/imunologia , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia
19.
J Immunol ; 181(5): 3449-55, 2008 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-18714017

RESUMO

NK cells hold great potential for improving the immunotherapy of cancer. Nevertheless, tumor cells can effectively escape NK cell-mediated apoptosis through interaction of MHC molecules with NK cell inhibitory receptors. Thus, to harness NK cell effector function against tumors, we used Amaxa gene transfer technology to gene-modify primary mouse NK cells with a chimeric single-chain variable fragment (scFv) receptor specific for the human erbB2 tumor-associated Ag. The chimeric receptor was composed of the extracellular scFv anti-erbB2 Ab linked to the transmembrane and cytoplasmic CD28 and TCR-zeta signaling domains (scFv-CD28-zeta). In this study we demonstrated that mouse NK cells gene-modified with this chimera could specifically mediate enhanced killing of an erbB2(+) MHC class I(+) lymphoma in a perforin-dependent manner. Expression of the chimera did not interfere with NK cell-mediated cytotoxicity mediated by endogenous NK receptors. Furthermore, adoptive transfer of gene-modified NK cells significantly enhanced the survival of RAG mice bearing established i.p. RMA-erbB2(+) lymphoma. In summary, these data suggest that use of genetically modified NK cells could broaden the scope of cancer immunotherapy for patients.


Assuntos
Imunoterapia Adotiva/métodos , Células Matadoras Naturais/transplante , Linfoma/terapia , Proteínas Recombinantes de Fusão/uso terapêutico , Animais , Anticorpos/genética , Antígenos CD28/genética , Citotoxicidade Imunológica , Progressão da Doença , Técnicas de Transferência de Genes , Humanos , Região Variável de Imunoglobulina , Células Matadoras Naturais/metabolismo , Linfoma/mortalidade , Camundongos , Camundongos Endogâmicos C57BL , Receptor ErbB-2/imunologia , Receptores de Antígenos de Linfócitos T/genética , Proteínas Recombinantes de Fusão/genética , Taxa de Sobrevida
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...