Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Cancer Immunol Immunother ; 64(8): 941-9, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25904200

RESUMO

Human T cells expressing γδ T cell receptor have a potential to show antigen-presenting cell-like phenotype and function upon their activation. However, the mechanisms that underlie the alterations in human γδ T cells remain largely unclear. In this study, we have investigated the molecular characteristics of human γδ T cells related to their acquisition of antigen-presenting capacity in comparison with activated αß T cells. We found that activated γδ but not αß T cells upregulated cell surface expression of a scavenger receptor, CD36, which seemed to be mediated by signaling through mitogen-activated protein kinase and/or NF-κB pathways. Confocal microscopical analysis revealed that activated γδ T cells can phagocytose protein antigens. Activated γδ T cells could induce tumor antigen-specific CD8(+) T cells using both apoptotic and live tumor cells as antigen resources. Furthermore, we detected that C/EBPα, a critical transcription factor for the development of myeloid-lineage cells, is expressed much higher in γδ T cells than in αß T cells. These results unveiled the molecular mechanisms for the elicitation of antigen-presenting functions in γδ T cells and would also help designing new approaches for γδ T cell-mediated human cancer immunotherapy.


Assuntos
Apresentação de Antígeno , Antígenos de Neoplasias/imunologia , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Células Mieloides/imunologia , Receptores de Antígenos de Linfócitos T gama-delta/imunologia , Linfócitos T/imunologia , Antígenos CD36/genética , Antígenos CD36/metabolismo , Linfócitos T CD8-Positivos/imunologia , Diferenciação Celular , Linhagem Celular Tumoral , Linhagem da Célula , Citotoxicidade Imunológica , Compostos Heterocíclicos com 3 Anéis/farmacologia , Humanos , Imidazóis/farmacologia , Imunoterapia/métodos , Ativação Linfocitária , Antígeno MART-1/imunologia , NF-kappa B/antagonistas & inibidores , Neoplasias/imunologia , Neoplasias/terapia , Fagocitose , Piridinas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores
2.
Int J Cancer ; 134(8): 1810-22, 2014 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-24150772

RESUMO

Complex interactions among multiple cell types contribute to the immunosuppressive milieu of the tumor microenvironment. Using a murine model of adoptive T-cell immunotherapy (ACT) for B16 melanoma, we investigated the impact of tumor infiltrating cells on this complex regulatory network in the tumor. Transgenic pmel-1-specific cytotoxic T lymphocytes (CTLs) were injected intravenously into tumor-bearing mice and could be detected in the tumor as early as on day 1, peaking on day 3. They produced IFN-γ, exerted anti-tumor activity and inhibited tumor growth. However, CTL infiltration into the tumor was accompanied by the accumulation of large numbers of cells, the majority of which were CD11b(+) Gr1(+) myeloid-derived suppressor cells (MDSCs). Notably, CD11b(+) Gr1(int) Ly6G(-) Ly6C(+) monocytic MDSCs outnumbered the CTLs by day 5. They produced nitric oxide, arginase I and reactive oxygen species, and inhibited the proliferation of antigen-specific CD8(+) T cells. The anti-tumor activity of the adoptively-transferred CTLs and the accumulation of MDSCs both depended on IFN-γ production on recognition of tumor antigens by the former. In CCR2(-/-) mice, monocytic MDSCs did not accumulate in the tumor, and inhibition of tumor growth by ACT was improved. Thus, ACT triggered counter-regulatory immunosuppressive mechanism via recruitment of MDSCs. Our results suggest that strategies to regulate the treatment-induced recruitment of these MDSCs would improve the efficacy of immunotherapy.


Assuntos
Imunoterapia Adotiva/métodos , Interferon gama/metabolismo , Melanoma Experimental/imunologia , Células-Tronco Neoplásicas/imunologia , Linfócitos T Citotóxicos/imunologia , Animais , Arginase/biossíntese , Antígeno CD11b/metabolismo , Proliferação de Células , Células Cultivadas , Células Dendríticas/imunologia , Terapia de Imunossupressão , Interferon gama/biossíntese , Masculino , Melanoma Experimental/terapia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Mieloides/imunologia , Células Mieloides/metabolismo , Óxido Nítrico/biossíntese , Espécies Reativas de Oxigênio/metabolismo , Receptores CCR2/deficiência , Receptores CCR2/genética , Linfócitos T Citotóxicos/metabolismo , Microambiente Tumoral
3.
Biochem Biophys Res Commun ; 371(2): 242-6, 2008 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-18423376

RESUMO

Dendritic cells (DCs) transfected with mRNA encoding tumor-associated antigens (TAAs) can induce tumor-specific T-cell responses. To potentiate this, we transfected mature DCs (mDCs) with mRNA encoding TAA targeted to the proteasome. DCs were generated from bone marrow cells by culture with 20 ng/ml GM-CSF and maturation with 1 microg/ml LPS. These mDCs were then electroporated with 10 microg of mRNA. Antigen presentation after electroporation with in vitro transcribed mRNA was compared with mRNA from a construct of the TAA preceded by ubiquitin. Proteasomal targeting of mRNA encoding cotranslationally ubiquitinated antigen was found to enhance intracellular degradation of target protein, and result in more efficient priming and expansion of TAA-specific CD8(+) T-cells. We therefore suggest that RNA-transfected DC vaccine efficacy could be improved by the use of mRNA targeted to the proteasome.


Assuntos
Apresentação de Antígeno , Antígenos de Neoplasias/metabolismo , Vacinas Anticâncer/imunologia , Células Dendríticas/imunologia , Linfócitos T Citotóxicos/imunologia , Ubiquitinação , Animais , Antígenos de Neoplasias/análise , Antígenos de Neoplasias/genética , Técnicas de Cultura de Células , Células Dendríticas/efeitos dos fármacos , Eletroporação , Antígeno MART-1 , Camundongos , Camundongos Endogâmicos , Proteínas de Neoplasias/análise , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transfecção , Ubiquitinação/genética
5.
Cancer Immunol Res ; 3(1): 26-36, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25127875

RESUMO

To understand global effector mechanisms of CTL therapy, we performed microarray gene expression analysis in a murine model using pmel-1 T-cell receptor (TCR) transgenic T cells as effectors and B16 melanoma cells as targets. In addition to upregulation of genes related to antigen presentation and the MHC class I pathway, and cytotoxic effector molecules, cell-cycle-promoting genes were downregulated in the tumor on days 3 and 5 after CTL transfer. To investigate the impact of CTL therapy on the cell cycle of tumor cells in situ, we generated B16 cells expressing a fluorescent ubiquitination-based cell-cycle indicator (B16-fucci) and performed CTL therapy in mice bearing B16-fucci tumors. Three days after CTL transfer, we observed diffuse infiltration of CTLs into the tumor with a large number of tumor cells arrested at the G1 phase of the cell cycle, and the presence of spotty apoptotic or necrotic areas. Thus, tumor growth suppression was largely dependent on G1 cell-cycle arrest rather than killing by CTLs. Neutralizing antibody to IFNγ prevented both tumor growth inhibition and G1 arrest. The mechanism of G1 arrest involved the downregulation of S-phase kinase-associated protein 2 (Skp2) and the accumulation of its target cyclin-dependent kinase inhibitor p27 in the B16-fucci tumor cells. Because tumor-infiltrating CTLs are far fewer in number than the tumor cells, we propose that CTLs predominantly regulate tumor growth via IFNγ-mediated profound cytostatic effects rather than via cytotoxicity. This dominance of G1 arrest over other mechanisms may be widespread but not universal because IFNγ sensitivity varied among tumors.


Assuntos
Pontos de Checagem do Ciclo Celular , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Interferon gama/imunologia , Proteínas Quinases Associadas a Fase S/metabolismo , Linfócitos T Citotóxicos/imunologia , Transferência Adotiva , Animais , Linhagem Celular Tumoral , Fase G1 , Expressão Gênica , Humanos , Masculino , Melanoma Experimental/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
6.
Clin Exp Metastasis ; 19(6): 519-26, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12405289

RESUMO

The anti-metastatic efficacy of MMI-166, which is a selective matrix metalloproteinase (MMP) inhibitor, in combination with CPT-11 was examined using two metastasis models of human gastrointestinal cancer cells. In the liver metastasis model, C-IH human colon cancer cells were injected into the spleen of athymic BALB/c nude mice. Daily oral (p.o.) dosing of MMI-166 at 200 mg/kg starting 1 day after tumor inoculation led to a significantly prolonged survival effect by inhibiting liver metastasis of C-1H tumor cells. CPT-11 (5 or 20 mg/kg) was administered intraperitoneally (i.p.) three times on day 3, day 7 and day 11 and also improved the survival of tumor-inoculated mice compared with the vehicle control. When MMI-166 was combined with CPT-11, the anti-metastatic efficacy was significantly augmented. Moreover, long tumor-free survival was noted in two of eight mice that were given the combination therapy but not either MMI-166 or CPT-11 monotherapy. In the peritoneal dissemination model, TMK-1 human gastric cancer cells were injected i.p. into nude mice. While both MMI-166, administered daily p.o. from day 1 at 200 mg/kg, and CPT-11, administered intravenously (i.v.) three times, inhibited the tumor dissemination and growth, the combination therapy of MMI-166 plus CPT-11 showed a greater inhibitory effect than each monotherapy. A hematotoxicity study demonstrated that CPT-11 alone significantly decreased the number of white blood cells (WBC) and bone marrow cells (BMC) in the mice during treatment, while the daily administration of MMI-166 alone had no such effect. More importantly, the combination therapy of MMI-166 with CPT-11 did not augment the hematotoxicity caused by CPT-11. An in vitro cytotoxicity study showed that MMI-166 itself neither has direct cytotoxicity in C-1H and TMK-1 tumor cells, nor does it augment the cytotoxicity of SN-38, an active form of CPT-11. The findings indicate that the augmented anti-metastatic efficacy in combination treatment was not simply due to the augmentation of direct cytotoxic activity, but was rather an additive or synergistic effect of anti-metastatic activities with different mechanisms. In conclusion, we demonstrated that the anti-metastatic efficacy against C-1H colon cancer and TMK-1 gastric cancer were augmented by the combination therapy of MMI-166, an orally active MMP inhibitor, with CPT-11. However, the hematotoxicity caused by CPT-11 was not augmented in the combination with MMI-166. Thus, the combination therapy of MMI-166 and CPT-11 exhibited potent anti-metastatic efficacy without increased hematotoxicity. These results point to the clinical advantage of using MMI-166 in combination with CPT-11.


Assuntos
Camptotecina/análogos & derivados , Camptotecina/uso terapêutico , Neoplasias do Colo/patologia , Neoplasias Hepáticas/secundário , Inibidores de Metaloproteinases de Matriz , Metástase Neoplásica/prevenção & controle , Sulfonamidas/uso terapêutico , Animais , Antineoplásicos/uso terapêutico , Neoplasias do Colo/tratamento farmacológico , Modelos Animais de Doenças , Humanos , Irinotecano , Neoplasias Hepáticas/prevenção & controle , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Metástase Neoplásica/patologia , Transplante Heterólogo
7.
Neuromuscul Disord ; 13(10): 796-803, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14678802

RESUMO

Alpha-dystroglycan is a cell surface peripheral membrane protein which binds to the extracellular matrix, while beta-dystroglycan is a type I integral membrane protein which anchors alpha-dystroglycan to the cell membrane via the N-terminal extracellular domain. The complex composed of alpha- and beta-dystroglycan is called the dystroglycan complex. Although defects of the dystroglycan gene have not been identified as the primary causes of hereditary diseases in humans, secondary but significant abnormalities of the dystroglycan complex have been revealed in severe muscular dystrophies, including sarcoglycanopathy (LGMD2C, D, E and F). In this study, we investigated proteolytic processing of beta-dystroglycan and its effect on the extracellular matrix-cell membrane linkage in cardiomyopathic hamsters, the model animals of LGMD2F. Compared to normal controls, proteolytic processing of beta-dystroglycan was activated in the skeletal, cardiac and smooth muscles of cardiomyopathic hamsters and this resulted in the partial disruption of the dystroglycan complex in these tissues. These phenomena were observed from the early phase of muscle degeneration process. Our results suggest that proteolytic processing of beta-dystroglycan disrupts the extracellular matrix-cell membrane linkage via the dystroglycan complex and this may play a role in the molecular pathogenesis of muscle degeneration in cardiomyopathic hamsters.


Assuntos
Cardiomiopatias/metabolismo , Proteínas do Citoesqueleto/metabolismo , Glicoproteínas de Membrana/metabolismo , Músculos/metabolismo , Animais , Encéfalo/metabolismo , Cardiomiopatias/patologia , Cardiomiopatias/fisiopatologia , Linhagem Celular , Membrana Celular/metabolismo , Cricetinae , Dimerização , Modelos Animais de Doenças , Distroglicanas , Matriz Extracelular/metabolismo , Substâncias Macromoleculares , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Músculo Liso/metabolismo , Músculo Liso/patologia , Músculos/patologia , Músculos/fisiopatologia , Miocárdio/metabolismo , Miocárdio/patologia , Peptídeo Hidrolases/metabolismo , Ratos
8.
J Neuroimmunol ; 127(1-2): 134-8, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12044984

RESUMO

Matrix metalloproteinases (MMPs) have been reported to be involved in various inflammatory disorders. Previous studies revealed that MMP-2 and MMP-9 might play important roles in the breakdown of the blood-brain barrier (BBB) in the central nervous system (CNS) of patients with HTLV-I-associated myelopathy (HAM)/tropical spastic paraparesis (TSP). N-Biphenyl sulfonyl-phenylalanine hydroxamic acid (BPHA) selectively inhibits MMP-2, -9 and -14, but not MMP-1, -3 and -7. In the present study, we examined whether or not the selective MMP inhibitor BPHA could inhibit the heightened migrating activity of CD4+ T cells in HAM/TSP patients. The migration assay using an invasion chamber showed that migration of CD4+ T cells in HAM/TSP patients was inhibited by 25 microM BPHA. In addition, the inhibitory ratio of migrating CD4+ lymphocytes was higher in HAM patients compared to normal controls. These results suggest that the selective MMP inhibitor BPHA has therapeutic potential for HAM/TSP.


Assuntos
Linfócitos T CD4-Positivos/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Ácidos Hidroxâmicos/farmacologia , Inibidores de Metaloproteinases de Matriz , Paraparesia Espástica Tropical/imunologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Materiais Biocompatíveis , Linfócitos T CD4-Positivos/citologia , Movimento Celular/efeitos dos fármacos , Movimento Celular/imunologia , Colágeno , Combinação de Medicamentos , Feminino , Humanos , Laminina , Masculino , Metaloproteinase 9 da Matriz/metabolismo , Pessoa de Meia-Idade , Paraparesia Espástica Tropical/tratamento farmacológico , Paraparesia Espástica Tropical/enzimologia , Proteoglicanas
9.
Cancer Lett ; 182(1): 61-8, 2002 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-12175524

RESUMO

The antitumor efficacy of the combination of nedaplatin (NDP) with gemcitabine (GEM) was evaluated against Ma44/GEM, a GEM-refractory subline of Ma44 human lung cancer, which was established by serial in vitro passage of Ma44 cells in the presence of GEM.Ma44/GEM showed less sensitivity to GEM and cytosine arabinoside with resistance factors of 7.7 and 8.3, respectively, but not to Taxol, Irinotecan, Mitomycin C and NDP. Flow cytometry analysis demonstrated that membrane transporter molecules such as multidrug-resistant, multidrug-resistant related protein or lung resistant protein were not induced in Ma44/GEM cells. In vivo experiments confirmed the less sensitivity of Ma44/GEM to GEM. The resistant factor of Ma44/GEM to GEM in vivo was estimated to be 6.7 in terms of ED(50).MA44/GEM-implanted athymic mice were treated with GEM i.v. once followed by i.v. injection of NDP at an interval of approximately 30 min. The mice were treated again with GEM after 3 or 4 days. The combined dosing of NDP with GEM resulted in synergistically enhanced inhibition of tumor growth against Ma44/GEM. The antitumor efficacy of the combination of NDP and GEM was superior to the best effect of either monotherapy. These results demonstrate the effectiveness of the combination of NDP with GEM against the GEM-refractory human lung cancer model.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Desoxicitidina/análogos & derivados , Desoxicitidina/administração & dosagem , Neoplasias Pulmonares/tratamento farmacológico , Compostos Organoplatínicos/administração & dosagem , Partículas de Ribonucleoproteínas em Forma de Abóbada , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/análise , Transportadores de Cassetes de Ligação de ATP/análise , Animais , Citarabina/farmacologia , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Transplante de Neoplasias , Transplante Heterólogo , Células Tumorais Cultivadas , Gencitabina
10.
Cancer Lett ; 178(2): 151-9, 2002 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-11867199

RESUMO

Matrix metalloproteinases (MMPs) have been implicated in tumor invasion, metastasis, and angiogenesis. We have recently shown that MMI-166, a new orally active MMP inhibitor specific for MMP-2 and -9, suppressed experimental metastasis of Lewis lung cancer, C-H1 human colon cancer, and pancreatic cancer without affecting tumor growth in vitro. In the present study, we determined whether oral administration of MMI-166 reduces tumor growth not only in such tumors but also in squamous cell carcinoma of head and neck (SCCHN). MMI-166 inhibited both activity of MMP-2 and -9 without affecting steady state levels of their mRNAs in SCCHN. Interestingly, protein levels of MMP-2 and -9 from the cultures were drastically diminished by culturing with MMI-166. This was also observed in xenografts of MMI-166-administered mice. In addition, daily oral administration of MMI-166 (100mg/kg) inhibited local tumor growth accompanied by the reduction of blood vessel density and Ki-67-positivity and increase in terminal deoxynucleotidyl transferase-mediated cUDP nick-end labeling (TUNEL)-positivity. These results suggested that orally administered MMI-166 reduced in vivo tumor growth of SCCHN through inhibition of angiogenesis and induction of apoptosis accompanied by the reduction of MMP productions and activities. Therefore, MMI-166 seems to be useful for tumor dormancy therapy of SCCHN.


Assuntos
Carcinoma de Células Escamosas/irrigação sanguínea , Carcinoma de Células Escamosas/tratamento farmacológico , Inibidores Enzimáticos/farmacologia , Neoplasias de Cabeça e Pescoço/irrigação sanguínea , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Neovascularização Patológica/tratamento farmacológico , Sulfonamidas/farmacologia , Animais , Carcinoma de Células Escamosas/enzimologia , Carcinoma de Células Escamosas/patologia , Inibidores Enzimáticos/uso terapêutico , Feminino , Neoplasias de Cabeça e Pescoço/enzimologia , Neoplasias de Cabeça e Pescoço/patologia , Humanos , Inibidores de Metaloproteinases de Matriz , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Transplante de Neoplasias , Sulfonamidas/uso terapêutico , Transplante Heterólogo , Células Tumorais Cultivadas
11.
J Control Release ; 82(2-3): 183-7, 2002 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-12175736

RESUMO

Recombinant interleukin-2 (IL-2) was strongly and almost completely adsorbed onto small hydrophobic liposomes under optimal conditions (liposome: DSPC-DSPG; molar ratio, 10:1; 30-50 nm in size, ratio of IL-2 to liposome: 4.0 JRU/nmol lipid). This liposomal IL-2 improved the distribution of IL-2 after intravenous administration as reported, previously. Liposomal IL-2 (300-10000 JRU/mouse per day) was significantly more effective than free IL-2 alone for inhibiting against the experimental metastases of M5076 in mice. The inhibitory effect of liposomal IL-2 was greatest in the liver. The ED(50) of liposomal IL-2 and that of free IL-2 in the liver were 1640 and 12500 JRU/mouse per day, respectively. This simple preparation (mixture) using IL-2 and liposome suspension is expected to have potential for increasing therapeutic efficacy against hepatic metastases.


Assuntos
Antineoplásicos/administração & dosagem , Interleucina-2/administração & dosagem , Neoplasias Hepáticas Experimentais/tratamento farmacológico , Neoplasias Hepáticas Experimentais/secundário , Linfoma Difuso de Grandes Células B/tratamento farmacológico , Proteínas Recombinantes/administração & dosagem , Animais , Antineoplásicos/uso terapêutico , Modelos Animais de Doenças , Composição de Medicamentos , Feminino , Injeções Intravenosas , Interleucina-2/uso terapêutico , Lipossomos , Linfoma Difuso de Grandes Células B/patologia , Camundongos , Camundongos Endogâmicos , Proteínas Recombinantes/uso terapêutico
12.
Anticancer Res ; 22(6A): 3253-9, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12530072

RESUMO

BACKGROUND: MMI-166 is a selective matrix metalloproteinase (MMP) inhibitor. The purpose of this study was to evaluate the antitumor efficacy of the combined treatment of MMI-166 with paclitaxel or carboplatin. MATERIALS AND METHODS: Mice bearing B16-BL6 melanoma were treated p.o. with MMI-166 from 1 day after tumor inoculation. The mice were administered i.v. with either paclitaxel or carboplatin at the maximum tolerated dose (MTD). RESULTS: MMI-166 monotherapy inhibited in vivo growth of the B16-BL6 tumor to an extent similar to that of paclitaxel or carboplatin monotherapy. When MMI-166 was combined with paclitaxel or carboplatin, the antitumor efficacy was significantly (p < 0.01) augmented in comparison with either MMI-166 or each cytotoxic agent alone. The hematotoxicity study demonstrated that daily treatment with MMI-166 did not affect the blood cell number in the mice and more importantly the combination of MMI-166 with paclitaxel did not augment the hematotoxicity caused by paclitaxel. An in vitro cytotoxicity study showed that MMI-166 itself has neither direct cytotoxicity against B16-BL6 tumor cells nor does it augment the cytotoxicity of paclitaxel or carboplatin. CONCLUSION: These results indicate that augmented antitumor activity of the combination treatment was not simply due to the augmentation of direct cytotoxic activity, but was rather an additive effect of the antitumor activities of different mechanisms. They suggest the effectiveness of a combination therapy of MMI-166 with paclitaxel or carboplatin in clinical therapy.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Inibidores Enzimáticos/farmacologia , Inibidores de Metaloproteinases de Matriz , Melanoma Experimental/tratamento farmacológico , Sulfonamidas/farmacologia , Animais , Carboplatina/administração & dosagem , Carboplatina/farmacologia , Divisão Celular/efeitos dos fármacos , Sinergismo Farmacológico , Inibidores Enzimáticos/administração & dosagem , Feminino , Melanoma Experimental/enzimologia , Melanoma Experimental/patologia , Camundongos , Paclitaxel/administração & dosagem , Paclitaxel/farmacologia , Sulfonamidas/administração & dosagem
13.
Gan To Kagaku Ryoho ; 29(11): 1943-9, 2002 Nov.
Artigo em Japonês | MEDLINE | ID: mdl-12465394

RESUMO

The antitumor activity of a combination of paclitaxel (TXL) followed by nedaplatin (NDP) against SK-OV-3 human ovarian cancer was evaluated. We also compared the antitumor activity of TXL plus NDP with that of TXL plus carboplatin (CBDCA) or TXL plus cisplatin (CDDP). TXL was injected i.v. daily for four days and either NDP, CBDCA or CDDP was injected i.v. once after the TXL treatment, into tumor-bearing mice. The sequential administration of TXL prior to NDP resulted in enhanced inhibition of tumor growth in comparison with either TXL or NDP monotherapy. The combination in TXL plus NDP was synergistic and superior to that of TXL plus CDDP or TXL plus CBDCA therapy. Histological tests demonstrated that the fraction of BrdU-incorporated cells in tumor tissue was significantly inhibited by the combination of TXL with NDP. These results demonstrated the antitumor efficacy of TXL with NDP against human ovarian cancer and suggest the clinical effectiveness of combination of TXL with NDP.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias Ovarianas/tratamento farmacológico , Animais , Apoptose/efeitos dos fármacos , Relação Dose-Resposta a Droga , Esquema de Medicação , Sinergismo Farmacológico , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Nus , Transplante de Neoplasias , Compostos Organoplatínicos/administração & dosagem , Neoplasias Ovarianas/patologia , Paclitaxel/administração & dosagem
14.
Int J Oncol ; 45(5): 1847-56, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25189159

RESUMO

Specific cellular immunotherapy for cancer requires efficient generation and expansion of cytotoxic T lymphocytes (CTLs) that recognize tumor-associated antigens. However, it is difficult to isolate and expand functionally active T-cells ex vivo. In this study, we investigated the efficacy of a new method to induce expansion of antigen-specific CTLs for adoptive immunotherapy. We used tumor-associated antigen glypican-3 (GPC3)-derived peptide and cytomegalovirus (CMV)-derived peptide as antigens. Treatment of human peripheral blood mononuclear cells (PBMCs) with zoledronate is a method that enables large-scale γδ T-cell expansion. To induce expansion of γδ T cells and antigen-specific CTLs, the PBMCs of healthy volunteers or patients vaccinated with GPC3 peptide were cultured with both peptide and zoledronate for 14 days. The expansion of γδ T cells and peptide-specific CTLs from a few PBMCs using zoledronate yields cell numbers sufficient for adoptive transfer. The rate of increase of GPC3­specific CTLs was approximately 24- to 170,000-fold. These CD8(+) cells, including CTLs, showed GPC3-specific cytotoxicity against SK-Hep-1/hGPC3 and T2 pulsed with GPC3 peptide, but not against SK-Hep-1/vec and T2 pulsed with human immunodeficiency virus peptide. On the other hand, CD8(-) cells, including γδ T cells, showed cytotoxicity against SK-Hep-1/hGPC3 and SK-Hep-1/vec, but did not show GPC3 specificity. Furthermore, adoptive cell transfer of CD8(+) cells, CD8(-) cells, and total cells after expansion significantly inhibited tumor growth in an NOD/SCID mouse model. This study indicates that simultaneous expansion of γδ T cells and peptide-specific CTLs using zoledronate is useful for adoptive immunotherapy.


Assuntos
Glipicanas/imunologia , Imunoterapia Adotiva/métodos , Neoplasias/imunologia , Linfócitos T Citotóxicos/imunologia , Animais , Antígenos de Neoplasias/imunologia , Moléculas de Adesão Celular/imunologia , Citomegalovirus/imunologia , Difosfonatos/administração & dosagem , Humanos , Imidazóis/administração & dosagem , Leucócitos Mononucleares/imunologia , Camundongos , Proteínas de Neoplasias/imunologia , Neoplasias/patologia , Neoplasias/terapia , Peptídeos/imunologia , Ácido Zoledrônico
15.
Int Immunopharmacol ; 18(1): 90-7, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24269583

RESUMO

Recent progress has been made in understanding the mechanisms of antitumor immune responses, which may further clarify the immune status of cancer patients. In this study, we performed a detailed evaluation of the immunological status of 47 patients with advanced solid cancer, who had received no immunosuppressive treatment, and compared the results with 32 healthy subjects. Flow-cytometry data for peripheral blood were obtained using 19 monoclonal antibodies against various cell surface and intracellular molecules. Absolute numbers of T cells, several T cell subsets, B cells, and NK cells were significantly decreased in patients compared with healthy subjects. The percentage of CD27(+)CD45RA(+) T cells was lower and that of CD27(-)CD45RA(-) T cells was higher in patients compared with controls. Regulatory and type 2 helper T cells were elevated in patients relative to healthy subjects. The percentage of perforin(+) NK cells was significantly lower in patients than in controls. These results suggest a dysfunctional anti-tumor immune response in cancer patients. Furthermore, peripheral blood from 26 of 47 cancer patients was analyzed after adoptive T cell immunotherapy (ATI). ATI increased the number of T cell subsets, but not B and NK cells. The number and percentage of regulatory T cells decreased significantly. These results suggest that ATI can restore impaired and imbalanced T cell immune status.


Assuntos
Linfócitos B/imunologia , Imunoterapia Adotiva/métodos , Células Matadoras Naturais/imunologia , Subpopulações de Linfócitos/imunologia , Neoplasias/imunologia , Linfócitos T Reguladores/imunologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Contagem de Células , Células Cultivadas , Citocinas/metabolismo , Feminino , Nível de Saúde , Homeostase , Humanos , Imunidade Celular , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Neoplasias/diagnóstico , Resultado do Tratamento
16.
Anticancer Res ; 34(8): 4601-7, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25075106

RESUMO

Several types of immune-cell therapies, such as αß T-cell, γδ T-cell, and dendritic cell (DC) vaccine therapies, are clinically employed for cancer treatment. The safety of immune-cell therapy for the treatment of patients with malignancies should be maintained by continuous assessment of adverse events. In the present study, we surveyed the adverse events associated with immune-cell therapy using large-scale prospective data and analyzed the side-effect profiles. For the assessment of adverse events associated with immune-cell therapy, we evaluated 771 treatment profiles (484 for αß T-cell therapy, 58 for γδ T-cell therapy, 206 for DC vaccine therapy, and 23 for concurrent therapy with αß T-cells and DC vaccines) from 144 patients with various malignancies. For the assessment of fever, fatigue, and itching, each of these adverse events was found to be grade 1 or 2 in most of the treated patients, except for one patient who had grade 3 itching. It was suggested that αß T-cell therapy could elicit a more rapid and direct immune reaction in patients than DC vaccine therapy, as shown by the earlier development of fever and higher incidence rate of fatigue. It was found that grade 1 or 2 reaction at the injection site developed in 10.2% of the patients injected with DC vaccines. Most of the grade 3 non-hematological and hematological adverse events were associated with disease progression or side-effects of chemotherapy, and were not considered to be related to immune-cell therapy. In conclusion, immune-cell therapy, such as αß T-cell, γδ T-cell, or DC vaccine therapy, was well-tolerated for cancer treatment.


Assuntos
Vacinas Anticâncer/efeitos adversos , Células Dendríticas/imunologia , Imunoterapia Adotiva/efeitos adversos , Neoplasias/terapia , Linfócitos T/imunologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Receptores de Antígenos de Linfócitos T alfa-beta , Receptores de Antígenos de Linfócitos T gama-delta
17.
Int Immunopharmacol ; 23(2): 499-504, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25284343

RESUMO

Dendritic cell (DC) vaccine is a promising immunotherapy for cancer due to its ability to induce antigen-specific CTLs efficiently. However, a number of clinical studies have implied insufficient therapeutic benefits with the use of MHC class 1 restricted peptide-pulsed DC vaccine. To enhance the clinical efficacy, we examined combination therapy of DC vaccine pulsed with OVA peptide and intravenous low dose unmodified IL-2 (IL-2 solution) administration against EG7 tumor-bearing mice. Unexpectedly, no additional effects of IL-2 solution were observed on CTL induction and the therapeutic effects of DC vaccine, possibly because of the short half-life of IL-2 in plasma. Therefore, we generated IL-2-encapsulating polymeric micelles (IL-2 micelle), which showed prolonged IL-2 retention in the circulation after intravenous administration compared with IL-2 solution. When mice were treated with OVA peptide-pulsed DCs in combination with IL-2 micelle, OVA-specific CTLs were efficiently induced in the spleen in comparison with DC vaccine combined with IL-2 solution or DC vaccine alone. In addition, combination therapy of DC vaccine and IL-2 micelle against EG7 tumor-bearing mice induced the efficient accumulation of antigen-specific CTLs into the tumor and marked anti-tumor effects. Thus, the administration of IL-2 micelle can significantly enhance DC vaccine efficacy against tumors.


Assuntos
Antígenos/metabolismo , Vacinas Anticâncer/imunologia , Células Dendríticas/imunologia , Interleucina-2/farmacologia , Neoplasias Experimentais/terapia , Linfócitos T Citotóxicos/fisiologia , Animais , Quimioterapia Combinada , Feminino , Regulação Neoplásica da Expressão Gênica , Imunoterapia , Interleucina-2/administração & dosagem , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Micelas , Ovalbumina/genética , Ovalbumina/metabolismo , Fragmentos de Peptídeos , Polímeros , Ratos , Ratos Wistar
18.
Oncoimmunology ; 2(9): e25636, 2013 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-24319636

RESUMO

The addition of zoledronate to tumor-associated antigen (TAA)-loaded dendritic cells (DCs) promotes the activation of interferon γ-secreting Vγ9 γδ T cells, in turn eliciting TAA-specific CD8+ T-cell responses. Immunological responses induced by zoledronate-pulsed DC-based vaccines have been associated with therapeutic effects in clinical trials.

19.
Int Immunopharmacol ; 15(3): 488-97, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23474736

RESUMO

Recent commercial approval of cancer vaccine, demonstrating statistically significant improvement in overall survival of prostate cancer patients has spurred renewed interest in active immunotherapies; specifically, strategies that lead to enhanced biological activity and robust efficacy for dendritic cell vaccines. A simple, widely used approach to generating multivalent cancer vaccines is to load tumor whole cell lysates into dendritic cells (DCs). Current DC vaccine manufacturing processes require co-incubation of tumor lysate antigens with immature DCs and their subsequent maturation. We compared electroloading of tumor cell lysates directly into mature DCs with the traditional method of lysate co-incubation with immature DCs. Electroloaded mature DCs were more potent in vitro, as judged by their ability to elicit significantly (p < 0.05) greater expansion of peptide antigen-specific CD8(+) T cells, than either lysate-electroloaded immature DCs or lysate-co-incubated immature DCs, both of which must be subsequently matured. Expanded CD8(+) T cells were functional as judged by their ability to produce IFN-γ upon antigen-specific re-stimulation. The electroloading technology used herein is an automated, scalable, functionally closed cGMP-compliant manufacturing technology supported by a Master File at CBER, FDA and represents an opportunity for translation of enhanced potency DC vaccines at clinical/commercial scale.


Assuntos
Vacinas Anticâncer/imunologia , Células Dendríticas/metabolismo , Eletroporação/métodos , Imunoterapia Adotiva/métodos , Melanoma/imunologia , Linfócitos T Citotóxicos/imunologia , Apresentação de Antígeno , Antígenos de Neoplasias/imunologia , Antígenos CD8/metabolismo , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Técnicas de Cocultura , Células Dendríticas/citologia , Estudos de Viabilidade , Humanos , Interferon gama/metabolismo , Ativação Linfocitária , Melanoma/terapia
20.
Anticancer Res ; 33(7): 2971-6, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23780988

RESUMO

Dendritic cell (DC)-based vaccines with the use of various antigen loading methods have been developed for cancer immunotherapy. Electroporation (EP) of a whole tumor cell lysate into DCs was previously found to be more potent for eliciting antigen-specific CD8 + T-cells compared to co-incubation of tumor cell lysates with DCs in vitro. In the present report, we studied the feasibility, safety and antitumor effect in the clinical use of an EP-DC vaccine for the immunotherapy of various types of human solid tumors. We successfully prepared an autologous tumor lysate-loaded EP-DC vaccine with high cell viability by the closed-flow electroporation system. In the phase I clinical trial, mild adverse events associated with the EP-DC vaccine were found during the treatment of advanced or recurrent cancer, or during the adjuvant therapy of some types of cancer; no autoimmune responses were observed after treatment with the autologous tumor lysate-loaded EP-DC vaccines. For the antitumor effect of the EP-DC vaccine against the 41 various types of solid tumor, the overall response rate [complete remission (CR) + partial response (PR)] was 4.9% (2/41) and the clinical benefit rate [CR+ PR + long stable disease (SD)] was 31.7% (13/41). Furthermore, the delayed-type hypersensitivity (DTH) reactivity was positive in most cases of long SD and the positive rate of DTH was 91.7% (11/12) for the patients with clinical benefit. In conclusion, the safety and feasibility of the EP-DC vaccine with autologous tumor lysates were confirmed, and it was found that the antitumor effect might be associated with the immunological response induced by the EP-DC vaccine for cancer immunotherapy.


Assuntos
Antígenos de Neoplasias/imunologia , Vacinas Anticâncer/uso terapêutico , Células Dendríticas/imunologia , Eletroporação , Hipersensibilidade Tardia/etiologia , Imunoterapia , Neoplasias/terapia , Adulto , Idoso , Antígenos de Neoplasias/metabolismo , Células Dendríticas/metabolismo , Estudos de Viabilidade , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Neoplasias/imunologia , Neoplasias/metabolismo , Prognóstico , Linfócitos T Citotóxicos/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA