Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Immunol Invest ; 43(2): 160-9, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24303799

RESUMO

Interaction between antigen presenting cells and T lymphocytes, through receptors and co-stimulatory molecules present on the surface of these cells, is one of the key means to modulate the adaptive immune system. Tucaresol, a Schiff-base-forming chemical, can be used as a substitute for the physiological donor of carbonyl groups of antigen presenting cells, which can interact with the amine groups of T lymphocytes to modulate the adaptive immune system. This study was done to determine whether tucaresol can enhance killing of cancer cells in vitro as well as protect non-obese diabetic severe combined immunodeficient mice from tumor development by mucin 1 stimulated human mononuclear cells through the adaptive immune system. The expected hypothesis was not supported. Percent specific lysis of MCF-7 tumor cells by mucin 1 stimulated human mononuclear cells was reduced by tucaresol. Furthermore, tucaresol abolished the protective effect of mucin 1 stimulated human mononuclear cells against MCF-7 breast cancer cell growth in non-obese diabetic severe combined immunodeficient mice. This study implies that tucaresol may be of use as an immunosuppressive agent.


Assuntos
Adenocarcinoma/terapia , Células Apresentadoras de Antígenos/efeitos dos fármacos , Benzaldeídos/administração & dosagem , Benzoatos/administração & dosagem , Neoplasias da Mama/terapia , Leucócitos Mononucleares/imunologia , Linfócitos T/imunologia , Adenocarcinoma/imunologia , Animais , Células Apresentadoras de Antígenos/imunologia , Antígenos de Neoplasias/imunologia , Neoplasias da Mama/imunologia , Citotoxicidade Imunológica/efeitos dos fármacos , Humanos , Sinapses Imunológicas/metabolismo , Terapia de Imunossupressão , Ativação Linfocitária/efeitos dos fármacos , Células MCF-7 , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Mucina-1/imunologia , Carbonilação Proteica , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Cancer Immunol Immunother ; 61(6): 839-54, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22083345

RESUMO

Adoptive T cell therapy for cancer patients optimally requires participation of CD4 T cells. In this phase I/II study, we assessed the therapeutic effects of adoptively transferred IL-10- and IFN-γ-producing CD4 effector cells in patients with recurrent ovarian cancer. Using MUC1 peptide and IL-2 for ex vivo CD4 effector cell generation, we show that three monthly treatment cycles of autologous T cell restimulation and local intraperitoneal re-infusion-modulated T cell-mediated immune responses that were associated with enhanced patient survival. One patient remains disease-free, another patient experienced prolonged survival for nearly 16 months with recurrent disease, and two patients expired within 3-5 months following final infusion. Prolonged survivors showed elevated levels of systemic CD3(+)CD4(+)CD25(+) and CD3(+)CD4(+)CD25(-) T cells when compared to that of pre-treatment levels and similarly treated short-term survivors. Such cell populations among these patients contained variable levels of "Inducible" Tr1 (CD4(+)CD25(-)FoxP3(-)IL-10(+)) and "Natural" (CD4(+)CD25(+)CD45RO(+)FoxP3(+)) TReg cell numbers and ratios that were associated with prolonged and/or disease-free survival. Moreover, peptide-restimulated T cells from these patients showed an elevation in both IFN-γ production, memory cell phenotype, and select TNF family ligands associated with enhanced T cell survival and apoptosis-inducing activities. This suggests that intraperitoneally administered Th1-like cells, producing elevated levels of IL-10, may require and/or induce differential levels of distinct systemic TReg subpopulations that influence, in part, long-term tumor immunity and enhanced memory/effector CD4-mediated therapeutic potentials. Furthermore, treatment efficacy and enhanced memory cell phenotype did not appear to be dependent on TReg cell numbers but upon ratios of "Inducible" and "Natural" TReg subpopulations.


Assuntos
Imunoterapia , Interferon gama/imunologia , Interleucina-10/imunologia , Neoplasias Ovarianas/imunologia , Neoplasias Ovarianas/terapia , Linfócitos T Reguladores/imunologia , Idoso , Feminino , Humanos , Interferon gama/biossíntese , Pessoa de Meia-Idade , Mucina-1/imunologia , Neoplasias Ovarianas/genética , Fenótipo
3.
Immunol Invest ; 39(6): 570-86, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20653425

RESUMO

The influence of the number of apheresis-stimulation-infusion(s) cycles, and the time in culture before the infusion (one vs. two weeks), on the generation of tumor antigen-specific cytotoxic T-lymphocytes (CTL) was investigated in a phase I/II clinical adoptive immunotherapy trial. Two previously treated metastatic breast cancer patients with no evidence of disease, in complete remission (CR), were enrolled. Each apheretic peripheral blood mononuclear cell (PBMC) sample was stimulated twice with MUC-1 before infusion back into the patients. Killer T-cells responses against MUC-1-expressing MCF-7 (CTL), nonspecific natural killer (NK) and lymphokine-activated killer (LAK) target cell lines, as well as, cytokine production were measured before each infusion. Patients received 2 infusions per month for 4 months. There were no tumor recurrences or toxicity. CTL, NK and LAK cells, type 1 cytokine, gamma-interferon (G-INF), and CD4(+) and CD8(+) memory T-lymphocytes were initially generated, produced or induced, respectively, and then declined. The CTL, NK and LAK cells were only induced at the first infusion of the first month. Thus, maintaining PBMC in culture longer than the first infusion was of no benefit with regards to retaining functional killer T-cells. In conclusion, this study implies that one treatment is optimal.


Assuntos
Adenocarcinoma/imunologia , Protocolos Antineoplásicos , Neoplasias da Mama/imunologia , Citotoxicidade Imunológica/efeitos dos fármacos , Imunoterapia Adotiva , Linfócitos T Citotóxicos/metabolismo , Adenocarcinoma/patologia , Adenocarcinoma/fisiopatologia , Adenocarcinoma/terapia , Neoplasias da Mama/patologia , Neoplasias da Mama/fisiopatologia , Neoplasias da Mama/terapia , Linhagem Celular Tumoral , Feminino , Seguimentos , Humanos , Leucaférese , Mucina-1/imunologia , Metástase Neoplásica , Recidiva Local de Neoplasia , Indução de Remissão , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/patologia , Fatores de Tempo
4.
Immunopharmacol Immunotoxicol ; 32(4): 647-55, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20470225

RESUMO

Mucin1 (MUC1) with altered glycosylation behaves as an antigen unique to adenocarcinomas (ADCs). As a step toward DNA vaccines, the goal of this work was to determine whether MUC1 peptides substituted with an asparagine at O-linked glycosylation sites, might expose MUC1 peptide backbone to serve as immunogens to generate cytotoxic T lymphocytes (CTL) from peripheral blood mononuclear cells of patients with ADCs. Substitution of some or all tyrosine and serine residues by asparagine in MUC1 did not inhibit the generation of mucin-specific CTLs. This suggests that MUC1 tandem repeat altered sequences to prevent O-linked glycosylation may be useful as DNA vaccines with tumor specificity.


Assuntos
Substituição de Aminoácidos/imunologia , Citotoxicidade Imunológica/imunologia , Leucócitos Mononucleares/efeitos dos fármacos , Ativação Linfocitária/efeitos dos fármacos , Mucina-1/imunologia , Fragmentos de Peptídeos/imunologia , Neoplasias da Mama/imunologia , Linhagem Celular Tumoral , Citocinas/metabolismo , Testes Imunológicos de Citotoxicidade , Feminino , Glicosilação , Proteína gp120 do Envelope de HIV/imunologia , Humanos , Interleucina-2/farmacologia , Células Matadoras Ativadas por Linfocina/imunologia , Células Matadoras Naturais/imunologia , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/metabolismo , Ativação Linfocitária/imunologia , Repetições Minissatélites/genética , Repetições Minissatélites/imunologia , Mucina-1/genética , Fragmentos de Peptídeos/genética , Linfócitos T Citotóxicos/imunologia
5.
Clin Immunol ; 133(3): 333-52, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19762283

RESUMO

Adoptive T cell immunotherapy using autologous lymphocytes is a viable treatment for patients with cancer and requires participation of Ag-specific CD4 and CD8 T cells. Here, we assessed the immunotherapeutic effects of autologous MUC1 peptide-stimulated CD4(+) effector cells following adoptive transfer in patients with ovarian cancer. Using MUC1 peptide and IL-2 for ex vivo CD4(+)/Th1 effector cell generation, we show that three monthly treatment cycles of peripheral blood T cell restimulation and intraperitoneal re-infusion selectively modulated endogenous T cell-mediated immune responses that correlated with diminished serum CA125 tumor marker levels and enhanced patient survival. One patient remains disease-free, another patient survived long-term for nearly 16 months with recurrent disease and two patients expired within 3-5 months following final infusion. Although PBL from all patients showed elevated MUC1 cytolytic activity following therapy, such responses did not correlate with therapeutic efficacy. Long-term survivors showed elevated levels of systemic memory (CD45RO) and naïve (CD45RA) CD3/CD4/CD25(+) T cells when compared to that of pre-treatment levels and similarly treated short-term survivors. Such cells co-expressed different levels of Foxp3 and CTLA-4 that resulted in progressively lower systemic Foxp3/CTLA-4 memory T cell ratios that further correlated with disease-free survival. Lastly, these patients showed elevated levels of MUC1-specific T cells expressing the CCR5 and CCR1 chemokine receptors and the chemokine CCL4 associated with Th1 cell differentiation/memory. We suggest that effective immunotherapy with autologous MUC1-stimulated CD4(+) effector cells induces differential levels of systemic "Ag-experienced" and "Ag-inexperienced" CD4/CD25(+) TReg cell subpopulations that influence long-term tumor immunity in ovarian cancer patients.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Imunoterapia Adotiva/métodos , Mucina-1/imunologia , Neoplasias Ovarianas/terapia , Linfócitos T Reguladores/imunologia , Células Th1/imunologia , Idoso , Sequência de Aminoácidos , Antígenos CD/sangue , Antígenos CD/imunologia , Antígeno Ca-125/sangue , Antígeno Ca-125/imunologia , Linfócitos T CD4-Positivos/citologia , Antígeno CTLA-4 , Citocinas/genética , Citocinas/imunologia , Feminino , Citometria de Fluxo , Fatores de Transcrição Forkhead/sangue , Fatores de Transcrição Forkhead/imunologia , Humanos , Antígenos Comuns de Leucócito/sangue , Antígenos Comuns de Leucócito/imunologia , Pessoa de Meia-Idade , Dados de Sequência Molecular , Neoplasias Ovarianas/imunologia , Subpopulações de Linfócitos T/imunologia , Linfócitos T Reguladores/citologia , Células Th1/citologia
6.
Immunol Invest ; 38(8): 820-38, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19860591

RESUMO

The influence of tumor burden on the generation of tumor antigen-specific cytotoxic T-lymphocytes (CTL) was investigated in a phase I/II clinical adoptive immunotherapy trial. Four previously treated metastatic breast cancer patients, two with macroscopic disease and two with no evidence of disease, in complete remission (CR), were enrolled. Each apheretic peripheral blood mononuclear cell (PBMC) sample was stimulated twice with MUC-1 before infusion back into the patients. CTL responses against MCF-7 cell line and cytokine production were measured before infusion. Patients received two monthly CTL infusions and were monitored for toxicity, tumor response as well as tumor marker levels. The CTL generated from patients with high tumor burdens had less cytokine production and lower cytotoxicity of MCF-7 than the CTL of patients in CR. The differences between the two groups were observed after the two MUC1 in vitro stimulations of the cells obtained in first apheresis. This difference increased after the two MUC1 stimulations of the cells obtained in the second apheresis. The cytotoxicity function was sustained from the first infusion to the second apheresis only for the patients in CR. This suggests that tumor burden had an inverse effect on the function of the generated CTL.


Assuntos
Neoplasias da Mama/imunologia , Neoplasias da Mama/patologia , Imunoterapia Adotiva , Transfusão de Leucócitos , Mucina-1/imunologia , Linfócitos T Citotóxicos/metabolismo , Carga Tumoral/imunologia , Biomarcadores Tumorais/metabolismo , Transfusão de Sangue Autóloga , Neoplasias da Mama/terapia , Linhagem Celular Tumoral , Citocinas/biossíntese , Citotoxicidade Imunológica , Feminino , Humanos , Leucócitos Mononucleares , Pessoa de Meia-Idade , Metástase Neoplásica , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/patologia
7.
Clin Immunol ; 128(2): 205-18, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18511346

RESUMO

The chemotherapeutic agent methotrexate is widely used in the treatment of breast cancer. Although its mechanism-of-action has been defined, less is known about its interaction with T cell-mediated antitumor responses. Type 1 CD8 T cell-mediated immune responses (Tc1) are cytolytic, produce IFN-gamma and are associated with effective antitumor responses. Using a murine transgenic TCR tumor model, we show that single-dose treatment with methotrexate enhanced CD8-mediated type 1 antitumor responses when administered 3 days prior to Tc1 effector cell transfer. Co-treatment with methotrexate not only enhanced donor Tc1 cell accumulation and persistence at sites of primary tumor growth, but also promoted elevated levels of activated donor TIL cells. This markedly enhanced the appearance of endogenous differentiated (CD44(High)) CD8 tumor-infiltrating cells when compared to that of corresponding groups receiving either MTX or Tc1 cell transfer alone. Such cells were acutely activated as defined by co-expression of surface markers associated with TCR engagement (CD69) and T cell activation (CD25) at both early (days 1-8) and late (days 12-20) stages following treatment. Conversely, such animals showed an early decrease in CD4(+)/CD44(High)/CD25(+)/CD69(+) T cells that correlated with delays in tumor growth in vivo. Moreover, cellular response kinetics appeared to further correlate with the up-regulation of endogenous T cells producing the chemokine IP-10 in vivo. This suggested that Tc1 cell transfer, in combination with chemotherapy, can enhance antitumor responses by modulating immunoregulatory T cells involved in homeostasis and immune tolerance within the tumor environment. These studies offer insight into mechanisms that enhance T cell-based immunotherapy in cancer.


Assuntos
Adenocarcinoma/terapia , Antimetabólitos Antineoplásicos/uso terapêutico , Imunoterapia Adotiva , Neoplasias Mamárias Experimentais/terapia , Metotrexato/uso terapêutico , Linfócitos T Citotóxicos/imunologia , Adenocarcinoma/imunologia , Animais , Antígenos CD/metabolismo , Antígenos de Diferenciação de Linfócitos T/metabolismo , Antimetabólitos Antineoplásicos/administração & dosagem , Quimiocinas/biossíntese , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Feminino , Injeções Intraperitoneais , Injeções Intravenosas , Subunidade alfa de Receptor de Interleucina-2/metabolismo , Lectinas Tipo C , Ativação Linfocitária , Neoplasias Mamárias Experimentais/imunologia , Metotrexato/administração & dosagem , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Especificidade do Receptor de Antígeno de Linfócitos T , Linfócitos T Citotóxicos/metabolismo , Antígenos Thy-1/genética , Antígenos Thy-1/imunologia
8.
Immunol Invest ; 37(4): 315-38, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18569073

RESUMO

The chemotherapeutic agent methotrexate is widely used in the treatment of breast cancer. Although its mechanism-of-action has been defined, less is known about its interaction with Ag-specific T cell-mediated antitumor responses. Type 1 CD8 T cell-mediated immune responses (Tc1) are cytolytic, produce IFN-gamma and are associated with effective antitumor responses. Using a murine transgenic TCR tumor model, we show that single-dose-treatment with methotrexate enhanced CD8-mediated type 1 antitumor responses when administered three days prior to Tc1 effector cell transfer. Co-treatment with methotrexate not only enhanced donor Tc1 cell accumulation and persistence at sites of primary tumor growth, but also promoted elevated levels of activated CD25(+) expressing donor TIL cells. This correlated with a marked decrease in the appearance of endogenous differentiated (CD44(High)) CD3/CD8/CD49b and CD3/CD4/CD49b tumor-infiltrating effector T cells at both early (Days 1-8) and late (Days 12-20) stages following treatment when compared to that of corresponding groups receiving either MTX or Tc1 cell transfer alone. Moreover, such cellular response kinetics appeared to further correlate with the down-regulation of endogenous CD4/CD44(High)/CD49b effector T cells producing IL-10 and delays in tumor growth in vivo. This suggested that Ag-specific Tc1 cell transfer, in combination with chemotherapy, can enhance antitumor responses by modulating select CD49b-expressing T effector/memory cell subpopulations involved in homeostasis and immune tolerance within the tumor environment. These studies offer insight into mechanisms that enhance T cell-based immunotherapy in cancer. Supplementary materials are available for this article. Go to the publisher's online edition of Immunological Investigations for the following free supplemental resource(s): Addendum 1.


Assuntos
Adenocarcinoma/terapia , Integrina alfa2/imunologia , Interleucina-10/metabolismo , Neoplasias Mamárias Animais/terapia , Metotrexato/farmacologia , Subpopulações de Linfócitos T/imunologia , Linfócitos T Citotóxicos/imunologia , Adenocarcinoma/imunologia , Transferência Adotiva , Animais , Antimetabólitos Antineoplásicos/administração & dosagem , Antimetabólitos Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Terapia Combinada , Feminino , Integrina alfa2/metabolismo , Interleucina-10/imunologia , Neoplasias Mamárias Animais/imunologia , Metotrexato/administração & dosagem , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Subpopulações de Linfócitos T/efeitos dos fármacos , Linfócitos T Citotóxicos/efeitos dos fármacos
9.
Immunol Invest ; 37(3): 215-25, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18389441

RESUMO

Many human adenocarcinomas can be killed in vitro by targeted cytotoxic T-lymphocytes (CTL); however, major histocompatibility complex (MHC)-restrictions are typically required. The MUC1 antigen is common in many human adenocarcinomas, and is associated with a variable number of tandem repeats. It has been proposed that antigens with such repeated epitopes may be vulnerable to cytotoxic T-lymphocyte killing without MHC-restriction. Therefore, it is possible that MUC1-expressing malignant cells may be killed by targeted cytotoxic T-lymphocyte in the absence of MHC-restriction. In this study, a human MUC1-expressing murine mammary carcinoma cell line was used to determine if cytotoxic T-lymphocyte killing of MUC1-expressing adenocarcinoma cells requires MHC-restriction. Specifically, MUC1-stimulated human mononuclear cells (M1SMC) were observed to kill human MUC1-transfected, MUC1-expressing murine mammary carcinoma cells, but not the mock-transfected, non-MUC1-expressing murine mammary carcinoma cells. Furthermore, the killing was blocked by antibody to MUC1, indicating MUC1-specific killing. In conclusion, cytotoxic T-lymphocyte killing of MUC1-expressing adenocarcinoma cells can be MHC-unrestricted.


Assuntos
Adenocarcinoma/imunologia , Citotoxicidade Imunológica , Leucócitos Mononucleares/imunologia , Complexo Principal de Histocompatibilidade/imunologia , Neoplasias Mamárias Animais/imunologia , Mucina-1/imunologia , Adenocarcinoma/metabolismo , Animais , Anticorpos Antineoplásicos/imunologia , Anticorpos Antineoplásicos/metabolismo , Células Apresentadoras de Antígenos/imunologia , Antígenos de Neoplasias/biossíntese , Antígenos de Neoplasias/imunologia , Antígenos de Neoplasias/metabolismo , Linfócitos T CD4-Positivos/imunologia , Epitopos , Feminino , Humanos , Epitopos Imunodominantes/imunologia , Ativação Linfocitária , Neoplasias Mamárias Animais/metabolismo , Camundongos , Mucina-1/química , Transgenes , Células Tumorais Cultivadas
10.
Oncol Lett ; 11(1): 461-465, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26870234

RESUMO

Cytotoxic T-lymphocyte activation and extension of the cell life span is necessary in order to enable immunotherapy to perform effectively against cancer. In the present study, mucin 1 (MUC1)-stimulated human mononuclear cells (M1SHMCs) were costimulated with bead-attached monoclonal antibodies specific for cluster of differentiation (CD)3 and CD28 receptors. The study was undertaken to determine whether costimulation was capable of enhancing the killing of cancer cells in vitro and of protecting non-obese diabetic severe combined immunodeficient mice from tumor development. Lysis of MCF-7 tumor cells by M1SHMCs was reduced following costimulation with anti-CD3 and anti-CD28. Furthermore, costimulation with anti-CD3 and anti-CD28 eliminated the protective effects of M1SHMCs on MCF-7 breast cancer cell growth in the non-obese diabetic severe combined immunodeficient mice. The present study suggested that costimulation with anti-CD3 and anti-CD28 is not advisable following antigen activation of lymphocytes under the conditions used here. Using a lower anti-CD3/CD28 bead to T-cell ratio may prevent immune suppression, however, further studies are required to support this hypothesis.

11.
Int J Mol Med ; 9(4): 401-4, 2002 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11891536

RESUMO

Mucin1 stimulated hematopoietic mononuclear cells (M1SHMC) from patients with breast cancer, adoptively transferred to non-obese diabetic, severe combined immunodeficient (NOD SCID) mice, extended survival in a therapy model of gross adenocarcinoma and prevented tumor growth in a model of minimal disease. M1SHMC exhibited specific lysis of a human breast adenocarcinoma cell line expressing mucin1, MCF-7 and produced interferon gamma. M1SHMC were injected intraperitoneally (IP) in NOD SCID mice after gross, palpable tumors appeared after MCF-7 were injected subcutaneously (SC). Survival was increased as compared to no M1SHMC controls. However tumors eventually regrew in all mice. To determine whether minimal disease (MD) could be controlled, NOD SCID were injected with MCF-7 cells, and on the same day, injected IP with M1SHMC. The M1SHMC injected mice were protected from tumor growth. These results imply that M1SHMC can prolong survival, but not cure NOD SCID mice bearing gross palpable adenocarcinomas. However in a minimal disease model tumor growth was prevented.


Assuntos
Adenocarcinoma/terapia , Neoplasias da Mama/terapia , Imunoterapia Adotiva , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/transplante , Mucina-1/imunologia , Adenocarcinoma/imunologia , Adenocarcinoma/patologia , Animais , Neoplasias da Mama/imunologia , Neoplasias da Mama/patologia , Citocinas/metabolismo , Feminino , Humanos , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Mucina-1/uso terapêutico , Transplante de Neoplasias , Neoplasia Residual/imunologia , Neoplasia Residual/terapia
12.
Oncol Lett ; 7(6): 2113-2117, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24932299

RESUMO

The aim of the present study was to determine whether the combination of two modalities of immunotherapy, targeting two different tumor antigens, may be feasible and non-toxic, yet enhance the killing of a human breast cancer cell line. The first modality was tumor growth factor α-Pseudomonas exotoxin 38 (TGFα-PE38), which specifically targets and kills tumor cells that express the epidermal growth factor receptor. The second modality was mucin-1 (MUC1)-specific cytotoxic T lymphocytes (CTLs), generated by MUC1 stimulation of peripheral blood mononuclear cells, to target the human breast cancer cell line, MCF7. TGFα-PE38 exhibited specific lysis of the MCF7 cells in a concentration- and time-dependent manner. TGFα-PE38 did not kill the normal hematopoietic stem cells or CTLs. Furthermore, TGFα-PE38 was not inhibitory for the growth or differentiation of the normal human hematopoietic stem cells into erythroid and myeloid colonies. In addition, TGFα-PE38 did not inhibit the killing function of CTLs, either when preincubated or co-incubated with CTLs. Finally, therapeutic enhancement was observed, in that TGFα-PE38 and CTLs were additive in the specific lysis of the MCF7 cells. These two modalities of immunotherapy may be beneficial for humans with breast cancer with or without other therapies, including autologous hematopoietic stem cell transplantation, specifically for purging cancer cells from hematopoietic stem cells prior to transplantation.

13.
Oncoimmunology ; 2(2): e23335, 2013 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-23526065

RESUMO

Dendritic cells (DCs) are among the most potent antigen-presenting cells (APCs), stimulating peripheral blood mononuclear cells (PBMCs) to generate antigen-specific cytotoxic T lymphocytes (CTLs). The objectives of this study were to determine if interleukin (IL)-4 is beneficial or detrimental for the generation of human DCs in vitro and to understand whether DCs generated in vitro in the presence or absence of IL-4 stimulate the killing of adenocarcinoma cells by CTLs in vivo. Mucin 1 (MUC1), a glycoprotein found on the surface of adenocarcinoma cells was used to load DCs. MUC1-loaded DCs generated in the absence of IL-4 were superior to their counterparts produced with IL-4 in stimulating PBMCs to kill human breast cancer MCF-7 cells in vitro. A corollary in vivo protection experiment was performed by injecting immunodeficient NOD-SCID mice with MCF-7 cells s.c. and MUC1-loaded CTLs, PBMCs, or DCs generated in the absence of IL-4, i.p. Mice that received CTLs and MUC1-loaded DCs on days 0, 2, 4, 9, 14 and 19 were completely protected against the development of MCF-7-derived tumors, while other schedules conferred lower protection. Therefore, tumor antigen-loaded DCs enhance the efficacy of adoptive CTL transfer, and should thus be considered for combinatorial immunotherapeutic regimens.

14.
Oncol Lett ; 4(5): 1061-1063, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23162651

RESUMO

CpG-ODNs activate various immune cell subsets and induce the production of numerous cytokines. To determine whether a CpG-ODN-activated innate immune system, without the adaptive immune system, was capable of protecting against cancer cell growth, NOD/SCID mice, which do not have T or B cell function but have a functional innate immune system, were used as a model system. NOD/SCID mice were injected subcutaneously with human prostate cancer cells followed by subcutaneous injection of incremental doses of CpG-ODNs. CpG-ODNs displayed a dose-related antitumoral effect leading to the prevention of tumor growth. These results indicate that ODNs are capable of activating the innate immune system and destroying human cancer cells in the absence of the adaptive immune system.

15.
J Immunother ; 35(2): 196-204, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22306908

RESUMO

The objective was to evaluate the toxicity and feasibility of intraperitoneal infusion of tumor-specific cytotoxic T lymphocytes (CTL) as therapy for recurrent ovarian cancer, and to determine if repetitive cycles of CTL generation and infusion measurably increases the host's ovarian cancer immune response. In this study, 7 subjects with recurrent ovarian cancer confined to the peritoneal cavity underwent up to 4 cycles, each cycle beginning with a leukapheresis for collection of precursor lymphocytes, which were stimulated in vitro with mucin 1, a tumor-specific antigen found commonly in ovarian cancer cells. The resulting new CTL for each cycle were reintroduced into the host by intraperitoneal infusion. Immunologic parameters (killer cells, cytokine production, memory T lymphocytes, and natural killer cells) were studied. Toxicity, CA-125, and survival data were also evaluated. The tumor marker CA-125 was nonstatistically significantly reduced after the first month of immunotherapy. However, after that it rose. Killer cells, cytokine production, and memory T lymphocytes increased after the first cycle of stimulation, but plateaued or reduced thereafter. The percent of natural killer cells inversely correlated with other immune parameters. Median survival was 11.5 months. One subject is free of disease since December, 2000. Multiple cycles, beyond 1 cycle, of T-cell stimulation followed by adoptive T-cell infusion, may not enhance the in vivo immune response.


Assuntos
Imunoterapia Adotiva/métodos , Neoplasias Epiteliais e Glandulares/terapia , Neoplasias Ovarianas/terapia , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/transplante , Antígeno Ca-125/sangue , Carcinoma Epitelial do Ovário , Feminino , Humanos , Imunofenotipagem , Mucina-1/imunologia , Neoplasias Epiteliais e Glandulares/imunologia , Neoplasias Ovarianas/imunologia , Projetos Piloto
16.
Mol Cancer Ther ; 8(8): 2412-23, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19671748

RESUMO

Expression of Migration inducting gene-7 (Mig-7) is limited to tumor cells and to date not found in normal tissues. Multiple tumor microenvironment factors, such as epidermal and hepatocyte growth factors, in concert with alphavbeta5 integrin ligation, induce Mig-7 mRNA expression. Gain or loss of Mig-7 protein studies shows that Mig-7 promotes invasion of colon and endometrial carcinoma cells. These data led us to hypothesize that targeting Mig-7 through various methods could decrease invasion, enhance monocyte cell killing of tumor cells, and inhibit disease progression. To begin testing this hypothesis, an in vitro chemoinvasion assay of endometrial carcinoma cells treated with Mig-7-specific or control antibodies was used. Mig-7 antibody significantly reduced invasion by >60% compared with controls. In another approach to test this hypothesis, an in vitro analysis of peptide-stimulated human peripheral blood monocyte cells and their killing of MCF-7 breast carcinoma cells was used. Mig-7 peptide treatment increased monocyte cell tumor necrosis factor expression and killing of MCF-7 cells 30-fold over no peptide stimulation and 3-fold over MUC-1 or control peptide treatments. Furthermore, stably expressing Mig-7-specific short hairpin RNA resulted in significantly reduced Mig-7 protein levels and early primary tumor growth in a xenograft nude mouse model. Reduced phosphorylation of ERK1/2, Akt, and S6 kinase as well as decreased membrane-type 1 matrix metalloproteinase activity were mechanisms through which Mig-7 protein caused these effects. Based on these collective data, Mig-7 expression could be a potential candidate for future targeted cancer therapies.


Assuntos
Carcinoma/patologia , Carcinoma/terapia , Monócitos/patologia , Invasividade Neoplásica/patologia , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/genética , Animais , Linhagem Celular Tumoral , Humanos , Metaloproteinase 14 da Matriz/genética , Metaloproteinase 14 da Matriz/metabolismo , Camundongos , Invasividade Neoplásica/genética , Proteínas de Neoplasias/metabolismo , Fosforilação
17.
J Immunol ; 177(11): 8191-201, 2006 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-17114496

RESUMO

CD8 T cell-mediated immune responses fall into two distinct types based on effector cell-derived cytokine production. Type I CD8 T cells (Tc1) produce IFN-gamma, whereas type 2 cells (Tc2) secrete IL-4, IL-5, IL-10, and GM-CSF. Using a murine TCR transgenic T cell/breast tumor model, we show that adoptively transferred Ag-specific Tc1 cells are more effective in delaying mammary tumor growth and progression than that of functionally distinct Tc2 cells. Donor Tc1 cells administered 7 days posttumor challenge localized and persisted at sites of primary tumor growth with antitumor responses that were dependent, in part, on effector cell-derived IFN-gamma. Tc1-mediated responses markedly enhanced the appearance and local accumulation of highly differentiated (CD44(high)) CD4 and CD8 endogenous tumor-infiltrating T cells when compared with that of untreated tumor-bearing mice. Conversely, Tc1 cell transfer markedly delayed the appearance of corresponding nondifferentiated (CD44(low)) endogenous T cells. Such cells were acutely activated as defined by coexpression of surface markers associated with TCR engagement (CD69) and early T cell activation (CD25). Moreover, cellular response kinetics appeared to further correlate with the up-regulation of endogenous T cells producing the chemokine IFN-gamma-inducible protein-10 in vivo. This suggested that CD8-mediated type 1 antitumor responses cannot only promote accumulation of distinct endogenous CD4 and CD8 T cell subpopulations, but also facilitate and preferentially modulate their localization kinetics, persistence, states of activation/differentiation, and function within the primary tumor environment at various stages of tumor progression. These studies offer insight into potential mechanisms for enhancing T cell-based immunotherapy in breast cancer.


Assuntos
Neoplasias da Mama/imunologia , Linfócitos T CD8-Positivos/imunologia , Ativação Linfocitária/imunologia , Linfócitos do Interstício Tumoral/imunologia , Subpopulações de Linfócitos T/imunologia , Transferência Adotiva , Animais , Linfócitos T CD4-Positivos/imunologia , Linhagem Celular Tumoral , Quimiocinas/imunologia , Quimiocinas/metabolismo , Feminino , Citometria de Fluxo , Expressão Gênica , Camundongos , Camundongos Transgênicos , Receptores de Antígenos de Linfócitos T/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA