Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Cancer Immunol Immunother ; 62(1): 39-49, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22733396

RESUMO

Lenalidomide is an effective therapeutic agent for multiple myeloma that exhibits immunomodulatory properties including the activation of T and NK cells. The use of lenalidomide to reverse tumor-mediated immune suppression and amplify myeloma-specific immunity is currently being explored. In the present study, we examined the effect of lenalidomide on T-cell activation and its ability to amplify responses to a dendritic cell-based myeloma vaccine. We demonstrate that exposure to lenalidomide in the context of T-cell expansion with direct ligation of CD3/CD28 complex results in polarization toward a Th1 phenotype characterized by increased IFN-γ, but not IL-10 expression. In vitro exposure to lenalidomide resulted in decreased levels of regulatory T cells and a decrease in T-cell expression of the inhibitory marker, PD-1. Lenalidomide also enhanced T-cell proliferative responses to allogeneic DCs. Most significantly, lenalidomide treatment potentiated responses to the dendritic cell/myeloma fusion vaccine, which were characterized by increased production of inflammatory cytokines and increased cytotoxic lymphocyte-mediated lysis of autologous myeloma targets. These findings indicate that lenalidomide enhances the immunologic milieu in patients with myeloma by promoting T-cell proliferation and suppressing inhibitory factors, and thereby augmenting responses to a myeloma-specific tumor vaccine.


Assuntos
Antineoplásicos/farmacologia , Vacinas Anticâncer/imunologia , Células Dendríticas/efeitos dos fármacos , Imunidade Celular/efeitos dos fármacos , Mieloma Múltiplo/imunologia , Linfócitos T/efeitos dos fármacos , Talidomida/análogos & derivados , Proliferação de Células/efeitos dos fármacos , Células Dendríticas/imunologia , Humanos , Immunoblotting , Lenalidomida , Ativação Linfocitária/efeitos dos fármacos , Subpopulações de Linfócitos T/efeitos dos fármacos , Subpopulações de Linfócitos T/imunologia , Linfócitos T/imunologia , Talidomida/farmacologia
2.
Blood ; 117(2): 393-402, 2011 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-21030562

RESUMO

We have developed a tumor vaccine in which patient-derived myeloma cells are chemically fused with autologous dendritic cells (DCs) such that a broad spectrum of myeloma-associated antigens are presented in the context of DC-mediated costimulation. We have completed a phase 1 study in which patients with multiple myeloma underwent serial vaccination with the DC/multiple myeloma fusions in conjunction with granulocyte-macrophage colony-stimulating factor. DCs were generated from adherent mononuclear cells cultured with granulocyte-macrophage colony-stimulating factor, interleukin-4, and tumor necrosis factor-α and fused with myeloma cells obtained from marrow aspirates. Vaccine generation was successful in 17 of 18 patients. Successive cohorts were treated with 1 × 10(6), 2 × 10(6), and 4 × 10(6) fusion cells, respectively, with 10 patients treated at the highest dose level. Vaccination was well tolerated, without evidence of dose-limiting toxicity. Vaccination resulted in the expansion of circulating CD4 and CD8 lymphocytes reactive with autologous myeloma cells in 11 of 15 evaluable patients. Humoral responses were documented by SEREX (Serologic Analysis of Recombinant cDNA Expression Libraries) analysis. A majority of patients with advanced disease demonstrated disease stabilization, with 3 patients showing ongoing stable disease at 12, 25, and 41 months, respectively. Vaccination with DC/multiple myeloma fusions was feasible and well tolerated and resulted in antitumor immune responses and disease stabilization in a majority of patients.


Assuntos
Vacinas Anticâncer/uso terapêutico , Células Dendríticas/imunologia , Imunoterapia/métodos , Mieloma Múltiplo/terapia , Adulto , Idoso , Anticorpos Antiprotozoários/imunologia , Antígenos de Protozoários/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/imunologia , Fusão Celular , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Mieloma Múltiplo/imunologia
3.
Mol Pharmacol ; 78(2): 166-74, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20444960

RESUMO

The MUC1 C-terminal transmembrane subunit (MUC1-C) oncoprotein is a direct activator of the canonical nuclear factor-kappaB (NF-kappaB) RelA/p65 pathway and is aberrantly expressed in human multiple myeloma cells. However, it is not known whether multiple myeloma cells are sensitive to the disruption of MUC1-C function for survival. The present studies demonstrate that peptide inhibitors of MUC1-C oligomerization block growth of human multiple myeloma cells in vitro. Inhibition of MUC1-C function also blocked the interaction between MUC1-C and NF-kappaB p65 and activation of the NF-kappaB pathway. In addition, inhibition of MUC1-C in multiple myeloma cells was associated with activation of the intrinsic apoptotic pathway and induction of late apoptosis/necrosis. Primary multiple myeloma cells, but not normal B-cells, were also sensitive to MUC1-C inhibition. Significantly, treatment of established U266 multiple myeloma xenografts growing in nude mice with a lead candidate MUC1-C inhibitor resulted in complete tumor regression and lack of recurrence. These findings indicate that multiple myeloma cells are dependent on intact MUC1-C function for constitutive activation of the canonical NF-kappaB pathway and for their growth and survival.


Assuntos
Sobrevivência Celular/fisiologia , Mucina-1/fisiologia , Mieloma Múltiplo/patologia , Sequência de Aminoácidos , Animais , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Dados de Sequência Molecular , Mucina-1/química , Mieloma Múltiplo/metabolismo , Mieloma Múltiplo/fisiopatologia , NF-kappa B/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transplante Heterólogo
4.
Clin Cancer Res ; 10(14): 4699-708, 2004 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-15269142

RESUMO

PURPOSE: Dendritic cells (DCs) are potent antigen-presenting cells that are uniquely capable of inducing tumor-specific immune responses. We have conducted a Phase I trial in which patients with metastatic breast and renal cancer were treated with a vaccine prepared by fusing autologous tumor and DCs. EXPERIMENTAL DESIGN: Accessible tumor tissue was disrupted into single cell suspensions. Autologous DCs were prepared from adherent peripheral blood mononuclear cells that were obtained by leukapheresis and cultured in granulocyte macrophage colony-stimulating factor, interleukin 4, and autologous plasma. Tumor cells and DCs were cocultured in the presence of polyethylene glycol to generate the fusions. Fusion cells were quantified by determining the percentage of cells that coexpress tumor and DC markers. Patients were vaccinated with fusion cells at 3-week intervals and assessed weekly for toxicity, and tumor response was assessed at 1, 3, and 6 months after completion of vaccination. RESULTS: The vaccine was generated for 32 patients. Twenty-three patients were vaccinated with 1 x 10(5) to 4 x 10(6) fusion cells. Fusion cells coexpressed tumor and DC antigens and stimulated allogeneic T-cell proliferation. There was no significant treatment-related toxicity and no clinical evidence of autoimmunity. In a subset of patients, vaccination resulted in an increased percentage of CD4 and CD8+ T cells expressing intracellular IFN-gamma in response to in vitro exposure to tumor lysate. Two patients with breast cancer exhibited disease regressions, including a near complete response of a large chest wall mass. Five patients with renal carcinoma and one patient with breast cancer had disease stabilization. CONCLUSIONS: Our findings demonstrate that fusion cell vaccination of patients with metastatic breast and renal cancer is a feasible, nontoxic approach associated with the induction of immunological and clinical antitumor responses.


Assuntos
Neoplasias da Mama/imunologia , Neoplasias Renais/imunologia , Neoplasias da Mama/patologia , Neoplasias da Mama/terapia , Proliferação de Células/efeitos dos fármacos , Separação Celular , Técnicas de Cocultura , Células Dendríticas/citologia , Células Dendríticas/imunologia , Feminino , Citometria de Fluxo , Hemocianinas/imunologia , Hemocianinas/farmacologia , Humanos , Células Híbridas/citologia , Células Híbridas/imunologia , Imuno-Histoquímica , Interferon gama/biossíntese , Neoplasias Renais/patologia , Neoplasias Renais/terapia , Masculino , Pessoa de Meia-Idade , Linfócitos T/citologia , Linfócitos T/imunologia , Linfócitos T/metabolismo , Resultado do Tratamento , Células Tumorais Cultivadas , Vacinação/métodos
5.
Cancer Res ; 73(17): 5569-79, 2013 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-23867470

RESUMO

Acute myeloid leukemia (AML) is a malignancy of stem cells with an unlimited capacity for self-renewal. MUC1 is a secreted, oncogenic mucin that is expressed aberrantly in AML blasts, but its potential uses to target AML stem cells have not been explored. Here, we report that MUC1 is highly expressed on AML CD34(+)/lineage(-)/CD38(-) cells as compared with their normal stem cell counterparts. MUC1 expression was not restricted to AML CD34(+) populations as similar results were obtained with leukemic cells from patients with CD34(-) disease. Engraftment of AML stem cell populations that highly express MUC1 (MUC1(high)) led to development of leukemia in NOD-SCID IL2Rgamma(null) (NSG) immunodeficient mice. In contrast, MUC1(low) cell populations established normal hematopoiesis in the NSG model. Functional blockade of the oncogenic MUC1-C subunit with the peptide inhibitor GO-203 depleted established AML in vivo, but did not affect engraftment of normal hematopoietic cells. Our results establish that MUC1 is highly expressed in AML stem cells and they define the MUC1-C subunit as a valid target for their therapeutic eradication.


Assuntos
Leucemia Mieloide Aguda/prevenção & controle , Mucina-1/metabolismo , Células-Tronco Neoplásicas/efeitos dos fármacos , Peptídeos/farmacologia , ADP-Ribosil Ciclase 1/genética , ADP-Ribosil Ciclase 1/metabolismo , Animais , Antígenos CD34/genética , Antígenos CD34/metabolismo , Apoptose/efeitos dos fármacos , Western Blotting , Diferenciação Celular , Proliferação de Células/efeitos dos fármacos , Feminino , Citometria de Fluxo , Humanos , Técnicas Imunoenzimáticas , Imunofenotipagem , Hibridização in Situ Fluorescente , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/patologia , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Mucina-1/química , Mucina-1/genética , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
6.
Clin Cancer Res ; 19(13): 3640-8, 2013 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-23685836

RESUMO

PURPOSE: A multiple myeloma vaccine has been developed whereby patient-derived tumor cells are fused with autologous dendritic cells, creating a hybridoma that stimulates a broad antitumor response. We report on the results of a phase II trial in which patients underwent vaccination following autologous stem cell transplantation (ASCT) to target minimal residual disease. EXPERIMENTAL DESIGN: Twenty-four patients received serial vaccinations with dendritic cell/myeloma fusion cells following posttransplant hematopoietic recovery. A second cohort of 12 patients received a pretransplant vaccine followed by posttransplant vaccinations. Dendritic cells generated from adherent mononuclear cells cultured with granulocyte macrophage colony-stimulating factor, interleukin-4, and TNF-α were fused with autologous bone marrow-derived myeloma fusion cells using polyethylene glycol. Fusion cells were quantified by determining the percentage of cells that coexpress dendritic cell and myeloma fusion antigens. RESULTS: The posttransplant period was associated with reduction in general measures of cellular immunity; however, an increase in CD4 and CD8(+) myeloma-specific T cells was observed after ASCT that was significantly expanded following posttransplant vaccination. Seventy-eight percent of patients achieved a best response of complete response (CR)+very good partial response (VGPR) and 47% achieved a CR/near CR (nCR). Remarkably, 24% of patients who achieved a partial response following transplant were converted to CR/nCR after vaccination and at more than 3 months posttransplant, consistent with a vaccine-mediated effect on residual disease. CONCLUSIONS: The posttransplant period for patients with multiple myeloma provides a unique platform for cellular immunotherapy in which vaccination with dendritic cell/myeloma fusion fusions resulted in the marked expansion of myeloma-specific T cells and cytoreduction of minimal residual disease.


Assuntos
Vacinas Anticâncer/imunologia , Células Dendríticas/imunologia , Transplante de Células-Tronco Hematopoéticas , Mieloma Múltiplo/imunologia , Mieloma Múltiplo/terapia , Adulto , Idoso , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/efeitos adversos , Feminino , Humanos , Imunidade Celular , Imunoterapia , Masculino , Pessoa de Meia-Idade , Indução de Remissão , Transplante Autólogo , Resultado do Tratamento , Vacinação
7.
J Immunother ; 35(7): 555-69, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22892452

RESUMO

The fowl pox vector expressing the tumor-associated antigens, mucin-1 and carcinoembryonic antigen in the context of costimulatory molecules (rF-PANVAC) has shown promise as a tumor vaccine. However, vaccine-mediated expansion of suppressor T-cell populations may blunt clinical efficacy. We characterized the cellular immune response induced by ex vivo dendritic cells (DCs) transduced with (rF)-PANVAC. Consistent with the functional characteristics of potent antigen-presenting cells, rF-PANVAC-DCs demonstrated strong expression of mucin-1 and carcinoembryonic antigen and costimulatory molecules, CD80, CD86, and CD83; decreased levels of phosphorylated STAT3, and increased levels of Tyk2, Janus kinase 2, and STAT1. rF-PANVAC-DCs stimulated expansion of tumor antigen-specific T cells with potent cytolytic capacity. However, rF-PANVAC-transduced DCs also induced the concurrent expansion of FOXP3 expressing CD4CD25 regulatory T cells (Tregs) that inhibited T-cell activation. Moreover, Tregs expressed high levels of Th2 cytokines [interleukin (IL)-10, IL-4, IL-5, and IL-13] together with phosphorylated STAT3 and STAT6. In contrast, the vaccine-expanded Treg population expressed high levels of Th1 cytokines IL-2 and interferon-γ and the proinflammatory receptor-related orphan receptor γt (RORγt) and IL-17A suggesting that these cells may share effector functions with conventional TH17 T cells. These data suggest that Tregs expanded by rF-PANVAC-DCs, exhibit immunosuppressive properties potentially mediated by Th2 cytokines, but simultaneous expression of Th1 and Th17-associated factors suggests a high degree of plasticity.


Assuntos
Vacinas Anticâncer/metabolismo , Antígeno Carcinoembrionário/metabolismo , Células Dendríticas/metabolismo , Glicoproteínas de Membrana/metabolismo , Mucina-1/metabolismo , Linfócitos T Citotóxicos/imunologia , Linfócitos T Reguladores/imunologia , Células Th17/imunologia , Antígenos CD4/metabolismo , Vacinas Anticâncer/genética , Antígeno Carcinoembrionário/genética , Células Cultivadas , Citocinas/metabolismo , Células Dendríticas/imunologia , Células Dendríticas/virologia , Fatores de Transcrição Forkhead/metabolismo , Vírus da Varíola das Aves Domésticas , Vetores Genéticos , Humanos , Subunidade alfa de Receptor de Interleucina-2/metabolismo , Ativação Linfocitária , Glicoproteínas de Membrana/genética , Mucina-1/genética , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/genética , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/metabolismo , Fatores de Transcrição STAT/genética , Fatores de Transcrição STAT/metabolismo , Equilíbrio Th1-Th2 , Transdução Genética
8.
Sci Signal ; 4(160): ra9, 2011 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-21325207

RESUMO

Signal transducer and activator of transcription 3 (STAT3) is activated in human breast cancer and other malignancies. Mucin 1 (MUC1) is a heterodimeric cell surface glycoprotein that is overexpressed in human carcinomas and, like STAT3, promotes cell survival and induces transformation. We found that in breast cancer cells, the MUC1 carboxyl-terminal receptor subunit (MUC1-C) associates with the gp130-Janus-activated kinase 1 (JAK1)-STAT3 complex. The MUC1-C cytoplasmic domain interacted directly with JAK1 and STAT3, and MUC1-C was necessary for JAK1-mediated STAT3 activation. In turn, MUC1-C and activated STAT3 occupied the promoter of MUC1, and MUC1-C contributed to STAT3-mediated activation of MUC1 transcription. The MUC1-C inhibitor GO-201 blocked the MUC1-C interaction with STAT3, thereby decreasing MUC1-C and STAT3 occupancy on the MUC1 and STAT3 promoters and activation of STAT3 target genes, including MUC1 itself. These findings indicate that MUC1-C promotes STAT3 activation and that MUC1-C and STAT3 function in an autoinductive loop that may play a role in cancer cell survival.


Assuntos
Janus Quinase 1/metabolismo , Mucina-1/metabolismo , Fator de Transcrição STAT3/metabolismo , Sítios de Ligação/genética , Linhagem Celular , Linhagem Celular Tumoral , Células HEK293 , Homeostase/efeitos dos fármacos , Humanos , Immunoblotting , Imunoprecipitação , Interleucina-6/farmacologia , Janus Quinase 1/genética , Luciferases/genética , Luciferases/metabolismo , Microscopia Confocal , Mucina-1/genética , Complexos Multiproteicos/metabolismo , Peptídeos/farmacologia , Fosforilação/efeitos dos fármacos , Regiões Promotoras Genéticas/genética , Ligação Proteica/efeitos dos fármacos , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Interferência de RNA , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Fator de Transcrição STAT3/genética
9.
J Immunother ; 34(5): 409-18, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21577144

RESUMO

We have developed a cancer vaccine in which autologous tumor is fused with dendritic cells (DCs) resulting in the presentation of tumor antigens in the context of DC-mediated costimulation. In clinical trials, immunologic responses have been observed, however responses may be muted by inhibitory pathways. The PD1/PDL1 pathway is an important element contributing to tumor-mediated immune suppression. In this study, we demonstrate that myeloma cells and DC/tumor fusions strongly express PD-L1. Compared with a control population of normal volunteers, increased PD-1 expression was observed on T cells isolated from patients with myeloma. It is interesting to note that after autologous transplantation, T-cell expression of PD-1 returned to levels seen in normal controls. We examined the effect of PD-1 blockade on T-cell response to DC/tumor fusions ex vivo. Presence of CT-011, an anti-PD1 antibody, promoted the vaccine-induced T-cell polarization towards an activated phenotype expressing Th1 compared with Th2 cytokines. A concomitant decrease in regulatory T cells and enhanced killing in a cytotoxicity assay was observed. In summary, we demonstrate that PD-1 expression is increased in T cells of patients with active myeloma, and that CT-011 enhances activated T-cell responses after DC/tumor fusion stimulation.


Assuntos
Antígenos CD/imunologia , Proteínas Reguladoras de Apoptose/imunologia , Vacinas Anticâncer , Células Dendríticas/imunologia , Leucócitos Mononucleares/imunologia , Mieloma Múltiplo/imunologia , Mieloma Múltiplo/patologia , Anticorpos Anti-Idiotípicos/imunologia , Antígenos CD/genética , Antígenos CD/metabolismo , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Antígeno B7-H1 , Vacinas Anticâncer/imunologia , Vacinas Anticâncer/farmacologia , Fusão Celular , Proliferação de Células/efeitos dos fármacos , Citotoxicidade Imunológica , Células Dendríticas/química , Células Dendríticas/metabolismo , Citometria de Fluxo , Humanos , Técnicas In Vitro , Leucócitos Mononucleares/química , Leucócitos Mononucleares/metabolismo , Ativação Linfocitária/imunologia , Mieloma Múltiplo/metabolismo , Mieloma Múltiplo/terapia , Receptor de Morte Celular Programada 1 , Linfócitos T Citotóxicos/imunologia , Linfócitos T Reguladores/imunologia , Equilíbrio Th1-Th2/efeitos dos fármacos , Transplante Autólogo
10.
J Immunother ; 33(2): 155-66, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20145548

RESUMO

Adoptive immunotherapy with tumor-specific T cells represents a promising treatment strategy for patients with malignancy. However, the efficacy of T-cell therapy has been limited by the ability to expand tumor-reactive cells with an activated phenotype that effectively target malignant cells. We have developed an anticancer vaccine in which patient-derived tumor cells are fused with autologous dendritic cells (DCs), such that a wide array of tumor antigens are presented in the context of DC-mediated costimulation. In this study, we demonstrate that DC/tumor fusions induce T cells that react with tumor and are dramatically expanded by subsequent ligation of the CD3/CD28 costimulatory complex. These T cells exhibit a predominantly activated phenotype as manifested by an increase in the percentage of cells expressing CD69 and interferon gamma. In addition, the T cells upregulate granzyme B expression and are highly effective in lysing autologous tumor targets. Targeting of tumor-specific antigen was demonstrated by the expansion of T cells with specificity for the MUC1 tetramer. Stimulation with anti-CD3/CD28 followed by DC/tumor fusions or either agent alone failed to result in a similar expansion of tumor-reactive T cells. Consistent with these findings, spectratyping analysis demonstrates selective expansion of T-cell clones as manifested by considerable skewing of the Vbeta repertoire following sequential stimulation with DC/tumor fusions and anti-CD3/CD28. Gene expression analysis was notable for the upregulation of inflammatory pathways. These findings indicate that stimulation with DC/tumor fusions provides a unique platform for subsequent expansion with anti-CD3/CD28 in adoptive T-cell therapy of cancer.


Assuntos
Antígenos CD/biossíntese , Antígenos de Diferenciação/biossíntese , Células Dendríticas/metabolismo , Granzimas/biossíntese , Linfócitos T/metabolismo , Apresentação de Antígeno/imunologia , Antígenos CD/genética , Antígenos CD/imunologia , Antígenos de Diferenciação/genética , Antígenos de Diferenciação/imunologia , Vacinas Anticâncer , Fusão Celular , Proliferação de Células , Células Clonais , Citotoxicidade Imunológica , Células Dendríticas/imunologia , Células Dendríticas/patologia , Genes Codificadores da Cadeia beta de Receptores de Linfócitos T , Granzimas/genética , Humanos , Interferon gama/metabolismo , Ativação Linfocitária , Mucina-1/imunologia , Mucina-1/metabolismo , Linfócitos T/imunologia , Linfócitos T/patologia
11.
Cancer Res ; 69(12): 5133-41, 2009 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-19491255

RESUMO

The mucin 1 (MUC1) oncoprotein is aberrantly overexpressed by approximately 90% of human breast cancers. However, there are no effective agents that directly inhibit MUC1 and induce death of breast cancer cells. We have synthesized a MUC1 inhibitor (called GO-201) that binds to the MUC1 cytoplasmic domain and blocks the formation of MUC1 oligomers in cells. GO-201, and not an altered version, attenuates targeting of MUC1 to the nucleus of human breast cancer cells, disrupts redox balance, and activates the DNA damage response. GO-201 also arrests growth and induces necrotic death. By contrast, the MUC1 inhibitor has no effect on cells null for MUC1 expression or nonmalignant mammary epithelial cells. Administration of GO-201 to nude mice bearing human breast tumor xenografts was associated with loss of tumorigenicity and extensive necrosis, which results in prolonged regression of tumor growth. These findings show that targeting the MUC1 oncoprotein is effective in inducing death of human breast cancer cells in vitro and in tumor models.


Assuntos
Antineoplásicos/farmacologia , Neoplasias da Mama/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Mucina-1/efeitos dos fármacos , Sequência de Aminoácidos , Animais , Biopolímeros , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Humanos , Camundongos , Camundongos Nus , Dados de Sequência Molecular , Mucina-1/química , Mucina-1/metabolismo
12.
J Immunol ; 181(1): 808-21, 2008 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-18566447

RESUMO

Vaccination of patients with dendritic cell (DC)/breast carcinoma fusions stimulated antitumor immune responses in a majority of patients with metastatic disease but only a subset demonstrate evidence of tumor regression. To define the factors that limit vaccine efficacy, we examined the biological characteristics of DC/breast carcinoma fusions as APCs and the nature of the vaccine-mediated T cell response. We demonstrate that fusion of DCs with breast carcinoma cells up-regulates expression of costimulatory and maturation markers and results in high levels of expression of IL-12 consistent with their role as activated APCs. Fusion cells also express the chemokine receptor CCR7, consistent with their ability to migrate to the draining lymph node. However, DC/breast cancer fusions stimulate a mixed T cell response characterized by the expansion of both activated and regulatory T cell populations, the latter of which is characterized by expression of CTLA-4, FOXP3, IL-10, and the suppression of T cell responses. Our results demonstrate that IL-12, IL-18, and TLR 9 agonist CpG oligodeoxynucleotides reduce the level of fusion-mediated regulatory T cell expansion. Our results also demonstrate that sequential stimulation with DC/breast carcinoma fusions and anti-CD3/CD28 results in the marked expansion of activated tumor-specific T cells. These findings suggest that DC/breast carcinoma fusions are effective APCs, but stimulate inhibitory T cells that limit vaccine efficacy. In contrast, exposure to TLR agonists, stimulatory cytokines, and anti-CD3/CD28 enhances vaccine efficacy by limiting the regulatory T cell response and promoting expansion of activated effector cells.


Assuntos
Neoplasias da Mama/imunologia , Antígenos CD28/imunologia , Complexo CD3/imunologia , Células Dendríticas/imunologia , Interleucina-12/farmacologia , Oligonucleotídeos/farmacologia , Linfócitos T Reguladores/imunologia , Anticorpos/imunologia , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Antígenos CD28/metabolismo , Complexo CD3/metabolismo , Diferenciação Celular , Fusão Celular , Proliferação de Células , Células Cultivadas , Células Dendríticas/citologia , Células Dendríticas/efeitos dos fármacos , Humanos , Interleucina-10/metabolismo , Interleucina-12/metabolismo , Ativação Linfocitária/imunologia , Mucina-1/metabolismo , Fenótipo , Ligação Proteica , Receptores CCR7/metabolismo , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/efeitos dos fármacos
13.
J Immunother ; 30(7): 749-61, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17893567

RESUMO

In the present study, we assessed the feasibility, toxicity, immunologic response, and clinical efficacy of vaccination with allogeneic dendritic cell (DC)/tumor fusions in patients with metastatic renal cell carcinoma (RCC). Patients with stage IV RCC with accessible tumor lesions or independent therapeutic indications for nephrectomy were eligible for enrollment. Tumors were processed into single cell suspensions and cryopreserved. DCs were generated from adherent peripheral blood mononuclear cells isolated from normal volunteers and cultured with granulocyte macrophage colony-stimulating factor, interleukin-4, and tumor necrosis factor-alpha. DCs were fused to patient derived RCC with serial electrical pulses. Patients received up to 3 vaccinations at a fixed dose of 4x10(7) to 1x10(8) cells administered at 6-week intervals. Twenty-four patients underwent vaccination. Twenty-one and 20 patients were evaluable for immunologic and clinical response, respectively. DCs demonstrated a characteristic phenotype with prominent expression of HLA class II and costimulatory molecules. A mean fusion efficiency of 20% was observed, determined by the percent of cells coexpressing DC and tumor antigens. No evidence of significant treatment related toxicity or auto-immunity was observed. Vaccination resulted in antitumor immune responses in 10/21 evaluable patients as manifested by an increase in CD4 and/or CD8 T-cell expression of interferon-gamma after ex vivo exposure to tumor lysate. Two patients demonstrated a partial clinical response by Response Evaluation Criteria in Solid Tumors criteria and 8 patients had stabilization of their disease. Vaccination of patients with RCC with allogeneic DC/tumor fusions was feasible, well tolerated, and resulted in immunologic and clinical responses in a subset of patients.


Assuntos
Vacinas Anticâncer/uso terapêutico , Carcinoma de Células Renais/terapia , Células Dendríticas/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Imunoterapia Adotiva , Neoplasias Renais/terapia , Adulto , Idoso , Idoso de 80 Anos ou mais , Apresentação de Antígeno , Carcinoma de Células Renais/imunologia , Feminino , Humanos , Imunoterapia Adotiva/efeitos adversos , Interferon gama/imunologia , Interferon gama/metabolismo , Estimativa de Kaplan-Meier , Neoplasias Renais/imunologia , Masculino , Pessoa de Meia-Idade , Linfócitos T/imunologia , Vacinação
14.
J Immunol ; 174(4): 2376-86, 2005 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-15699174

RESUMO

The MUC1 transmembrane mucin is expressed on the surface of activated human T cells; however, the physiologic signals responsible for the regulation of MUC1 in T cells are not known. The present studies demonstrate that IL-7, but not IL-2 or IL-4, markedly induces MUC1 expression on CD3+ T cells. MUC1 was also up-regulated by IL-15, but to a lesser extent than that found with IL-7. The results show that IL-7 up-regulates MUC1 on CD4+, CD8+, CD25+, CD69+, naive CD45RA+, and memory CD45RO+ T cells. In concert with induction of MUC1 expression by IL-7, activated dendritic cells (DC) that produce IL-7 up-regulate MUC1 on allogeneic CD3+ T cells. DC also induce MUC1 expression on autologous CD3+ T cells in the presence of recall Ag. Moreover, DC-induced MUC1 expression on T cells is blocked by a neutralizing anti-IL-7 Ab. The results also demonstrate that DC induce polarization of MUC1 on T cells at sites opposing the DC-T cell synapse. These findings indicate that DC-mediated activation of Ag-specific T cells is associated with induction and polarization of MUC1 expression by an IL-7-dependent mechanism.


Assuntos
Células Dendríticas/imunologia , Interleucina-7/fisiologia , Mucina-1/biossíntese , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Apresentação de Antígeno , Comunicação Celular/imunologia , Células Cultivadas , Técnicas de Cocultura , Células Dendríticas/citologia , Células Dendríticas/metabolismo , Humanos , Memória Imunológica , Antígenos Comuns de Leucócito/biossíntese , Ativação Linfocitária/imunologia , Mucina-1/metabolismo , Fase de Repouso do Ciclo Celular/imunologia , Subpopulações de Linfócitos T/citologia
15.
Br J Haematol ; 129(5): 687-700, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15916692

RESUMO

Dendritic cells (DCs) are potent antigen-presenting cells that are uniquely capable of inducing primary immune responses. Although tumour cells may directly inhibit DC maturation, exposure to tumour products may also result in their activation. Fusions of cancer cells and DCs are being explored as cancer vaccines. The effect of tumour cell fusion on DC maturation and their functional characteristics has not been defined. In the present study, immature and mature DC generated from human CD34+ and peripheral blood precursors were fused to multiple myeloma cells in the presence of polyethylene glycol. Fusion of both immature and mature DCs with tumour cells resulted in an activated phenotype. In this regard, fusion cells expressed interleukin-12, a cytokine essential for the induction of T-helper cell type 1 immunity. In contrast to immature DCs, fusion cells also strongly expressed CC-chemokine receptor R7, which is responsible for DC migration to draining lymph nodes. Fusions generated with both immature and mature DCs also potently stimulated T-cell expression of gamma-interferon and cytotoxic T lymphocyte killing of tumour targets. These findings demonstrate that tumour cell fusion induces DC maturation and the development of an activated phenotype necessary for their effectiveness as cancer vaccines.


Assuntos
Apresentação de Antígeno , Vacinas Anticâncer , Células Dendríticas/citologia , Mieloma Múltiplo/imunologia , Mieloma Múltiplo/patologia , Antígenos CD34/imunologia , Fusão Celular , Testes Imunológicos de Citotoxicidade , Citometria de Fluxo , Humanos , Células Híbridas , Interferon gama/imunologia , Interleucina-10/imunologia , Interleucina-12/imunologia , Ativação Linfocitária , Teste de Cultura Mista de Linfócitos , Receptores CCR7 , Receptores de Quimiocinas/imunologia , Linfócitos T/imunologia
16.
Blood ; 102(5): 1745-52, 2003 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-12714509

RESUMO

We have shown previously that primary dendritic cells and monocytes express equal levels of CD14 but are distinguishable by the presence of CD2 on dendritic cells. CD2 is known to mediate the activation of T and natural killer (NK) cells through its interaction with CD58. CD2 epitopes recognized by anti-T111, -T112, and -T113 monoclonal antibodies (mAbs) are present on dendritic cells. Here we show that CD2 engagement significantly increases class II, costimulatory (CD40, CD80, CD86), adhesion (CD54, CD58), and CCR7 molecule expression on primary dendritic cells. Conversely, minimal or no change in the expression of the above antigens occurs on monocyte-derived dendritic cells, because these molecules are already maximally expressed. However, both kinds of dendritic cells release interleukin-1beta (IL-1beta) and IL-12 after CD2 engagement. Lastly, interference with dendritic cell CD2-T-cell CD58 engagement decreases naive CD4+CD45RA+ T-cell proliferation. Collectively, our results suggest another role of the CD2-CD58 pathway that allows nonimmune and immune cells to interact directly with dendritic cells and initiate innate and adaptive immune responses.


Assuntos
Antígenos CD2/metabolismo , Linfócitos T CD4-Positivos/imunologia , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Animais , Antígenos de Superfície/imunologia , Antígenos de Superfície/metabolismo , Antígenos CD2/genética , Linfócitos T CD4-Positivos/citologia , Antígenos CD58/metabolismo , Células CHO , Comunicação Celular/imunologia , Cricetinae , Citocinas/biossíntese , Células Dendríticas/citologia , Endocitose/imunologia , Epitopos/metabolismo , Humanos , Imunofenotipagem , Antígenos Comuns de Leucócito/metabolismo , Ativação Linfocitária/imunologia , Teste de Cultura Mista de Linfócitos , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA