Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Cancer Res Clin Oncol ; 140(11): 1815-23, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24927808

RESUMO

PURPOSE: Lipopolysaccharide (LPS) is a major component of the outer surface membrane of Gram-negative bacteria which has been proved an effective immune enhancer. Here, we investigated the anti-tumor effect of irradiated tumor cells that stimulated by LPS in mouse xenografts models. METHODS: Tumor cells were irradiated after stimulation with 1 µg/mL LPS for 48 h. The C57BL/6 mice were immunized subcutaneously with irradiated tumor cells. The anti-tumor effect of lymphocytes of immunized mice was investigated. The cytotoxicity of spleen lymphocytes from immunized mice was determined by a standard (51)Cr-release assay. The roles of immune cell subsets in anti-tumor activity were assessed by injected intraperitoneally with monoclonal antibodies. RESULTS: We observed that the vaccine of irradiated tumor cell with LPS-stimulated elicited a stronger protective anti-tumor immunity than other controls. Adoptive transfer of lymphocytes of immunized mice showed that the cellular immune response was involved in the anti-tumor effect. And this effect was achieved by activation of antigen-specific CD8(+) T cell response and reduction of myeloid-derived suppressor cells (MDSCs, Gr1(+) CD11b (+) ), which were confirmed by depletion of immune cell subsets and flow cytometry analysis. CONCLUSIONS: In summary, our study showed that stimulation of LPS was able to enhance anti-tumor immunity of vaccination with tumor cells after irradiation treatment, which might be a new strategy for cancer therapy.


Assuntos
Vacinas Anticâncer/imunologia , Lipopolissacarídeos/farmacologia , Melanoma Experimental/terapia , Animais , Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/efeitos da radiação , Linhagem Celular Tumoral , Feminino , Hibridomas , Imunoterapia Adotiva , Melanoma Experimental/imunologia , Melanoma Experimental/patologia , Camundongos Endogâmicos C57BL , Baço/imunologia , Carga Tumoral/imunologia , Vacinação , Ensaios Antitumorais Modelo de Xenoenxerto
2.
BMC Cancer ; 14: 48, 2014 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-24475975

RESUMO

BACKGROUND: Although the whole tumor cell vaccine can provide the best source of immunizing antigens, there is still a limitation that most tumors are not naturally immunogenic. Tumor cells genetically modified to secrete immune activating cytokines have been proved to be more immunogenic. IL-18 could augment proliferation of T cells and cytotoxicity of NK cells. GM-CSF could stimulate dendritic cells, macrophages and enhance presentation of tumor antigens. In our study, we used mouse GM-CSF combined with IL-18 to modify Lewis lung cancer LL/2, then investigated whether vaccination could suppress tumor growth and promote survival. METHODS: The Lewis lung cancer LL/2 was transfected with co-expressing mouse GM-CSF and IL-18 plasmid by cationic liposome, then irradiated with a sublethal dose X ray (100 Gy) to prepare vaccines. Mice were subcutaneously immunized with this inactivated vaccine and then inoculated with autologous LL/2 to estimate the antitumor efficacy. RESULTS: The studies reported here showed that LL/2 tumor cell vaccine modified by a co-expressing mouse GM-CSF and IL-18 plasmid could significantly inhibit tumor growth and increased survival of the mice bearing LL/2 tumor whether prophylactic or adoptive immunotherapy in vivo. A significant reduction of proliferation and increase of apoptosis were also observed in the tumor treated with vaccine of co-expressing GM-CSF and IL-18. The potent antitumor effect correlated with higher secretion levels of pro-inflammatory cytokines such as IL-18, GM-CSF, interferon-γ in serum, the proliferation of CD4+ IFN-γ+, CD8+ IFN-γ+ T lymphocytes in spleen and the infiltration of CD4+, CD8+ T in tumor. Furthermore, the mechanism of tumor-specific immune response was further proved by 51Cr cytotoxicity assay in vitro and depletion of CD4, CD8, NK immune cell subsets in vivo. The results suggested that the antitumor mechanism was mainly depended on CD4+, CD8+ T lymphocytes. CONCLUSIONS: These results provide a new insight into therapeutic mechanisms of IL-18 plus GM-CSF modified tumor cell vaccine and provide a potential clinical cancer immunotherapeutic agent for improved antitumor immunity.


Assuntos
Vacinas Anticâncer/biossíntese , Carcinoma Pulmonar de Lewis/terapia , Terapia Genética/métodos , Fator Estimulador de Colônias de Granulócitos e Macrófagos/biossíntese , Interleucina-18/biossíntese , Animais , Apoptose , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/genética , Vacinas Anticâncer/efeitos da radiação , Carcinoma Pulmonar de Lewis/genética , Carcinoma Pulmonar de Lewis/imunologia , Carcinoma Pulmonar de Lewis/metabolismo , Carcinoma Pulmonar de Lewis/patologia , Linhagem Celular Tumoral , Proliferação de Células , Feminino , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/efeitos da radiação , Imunização , Imunoterapia Adotiva , Interleucina-18/genética , Interleucina-18/efeitos da radiação , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Baço/imunologia , Baço/metabolismo , Fatores de Tempo , Transfecção , Carga Tumoral
3.
Ann Surg ; 253(2): 328-35, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21217520

RESUMO

PURPOSE: Surgical resection provides the only possibility of cure for pancreas cancer. A standard adjuvant approach has not been established. We tested the safety and efficacy of a granulocyte-macrophage colony-stimulating factor (GM-CSF)-based immunotherapy administered in patients with resected pancreatic adenocarcinoma. PATIENTS AND METHODS: A single institution phase II study of 60 patients with resected pancreatic adenocarcinoma was performed. Each immunotherapy treatment consisted of a total of 5 × 108 GM-CSF-secreting cells distributed equally among 3 lymph node regions. The first immunotherapy treatment was administered 8 to 10 weeks after surgical resection. Subsequently, patients received 5-FU based chemoradiation. Patients who remained disease-free after completion of chemoradiotherapy received treatments 2 to 4, each 1 month apart. A fifth and final booster was administered 6 months after the fourth immunotherapy. The primary endpoint was disease free survival and secondary endpoints were overall survival and toxicity, and the induction of mesothelin specific T cell responses. RESULTS: The median disease-free survival is 17.3 months (95% CI, 14.6-22.8) with median survival of 24.8 months (95% CI, 21.2-31.6). The administration of immunotherapy was well tolerated. In addition, the post-immunotherapy induction of mesothelin-specific CD8+ T cells in HLA-A1+ and HLA-A2+patients correlates with disease-free survival. CONCLUSIONS: An immunotherapy approach integrated with chemoradiation is safe and demonstrates an overall survival that compares favorably with published data for resected pancreas cancer. These data suggest additional boost immunotherapies given at regular intervals beyond 1 year postsurgery should be tested in future studies, and provide the rationale for conducting a multicenter phase II study.


Assuntos
Vacinas Anticâncer/uso terapêutico , Carcinoma Ductal Pancreático/terapia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/uso terapêutico , Imunoterapia , Neoplasias Pancreáticas/terapia , Adulto , Idoso , Idoso de 80 Anos ou mais , Antígenos de Neoplasias/imunologia , Vacinas Anticâncer/efeitos adversos , Vacinas Anticâncer/imunologia , Vacinas Anticâncer/efeitos da radiação , Carcinoma Ductal Pancreático/imunologia , Carcinoma Ductal Pancreático/mortalidade , Linhagem Celular Tumoral/metabolismo , Linhagem Celular Tumoral/efeitos da radiação , Terapia Combinada , Intervalo Livre de Doença , Feminino , Proteínas Ligadas por GPI/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Humanos , Hipersensibilidade Tardia/imunologia , Masculino , Mesotelina , Pessoa de Meia-Idade , Neoplasias Pancreáticas/imunologia , Neoplasias Pancreáticas/mortalidade , Taxa de Sobrevida , Linfócitos T/imunologia , Transfecção
4.
Biol Blood Marrow Transplant ; 17(3): 330-40, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20946965

RESUMO

Vaccination with irradiated autologous tumor cells, engineered to secrete granulocyte macrophage-colony stimulating factor (GM-CSF) (GM tumor), can generate potent antitumor effects when combined with autologous bone marrow transplantation (BMT). That notwithstanding, the post-BMT milieu, characterized by marked cytopenia, can pose a challenge to the implementation of vaccine immunotherapies. To bypass this problem, partial post-BMT immune reconstitution has been allowed to develop prior to vaccination. However, delaying vaccination can also potentially allow the expansion of residual tumor cells. Other approaches have used reinfusion of "primed" autologous lymphocytes and multiple administrations of GM tumor cells, which required the processing of large amounts of tumor. Utilizing the MMB3.19 murine myeloid leukemia model, we tested whether a single dose of GM tumor cells, 7 days prior to syngeneic BMT, could be a curative treatment in MMB3.19-challenged recipient mice. This vaccination protocol significantly improved survival of mice by eliciting long-lasting host immune responses that survived lethal irradiation, and were even protective against post-BMT tumor rechallenge. Furthermore, we demonstrated that mature donor lymphocytes can also play a limited role in mounting the antitumor response, but our pre-BMT vaccination strategy obviated the need for either established de novo immune reconstitution or the use of multiple post-BMT immunizations.


Assuntos
Imunidade Adaptativa , Transplante de Medula Óssea/imunologia , Vacinas Anticâncer/uso terapêutico , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Leucemia Mieloide/metabolismo , Leucemia Mieloide/prevenção & controle , Imunidade Adaptativa/efeitos da radiação , Animais , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/efeitos da radiação , Linhagem Celular Tumoral , Genes Reporter , Efeito Enxerto vs Leucemia/imunologia , Efeito Enxerto vs Leucemia/efeitos da radiação , Imunidade Celular/efeitos da radiação , Injeções Intraperitoneais , Leucemia Mieloide/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Transplante de Neoplasias , Análise de Sobrevida , Linfócitos T/imunologia , Linfócitos T/efeitos da radiação , Linfócitos T/transplante , Transplante Autólogo , Irradiação Corporal Total
5.
Vaccine ; 26(46): 5855-63, 2008 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-18771701

RESUMO

Ovarian cancer is one of the leading causes of death from gynecological cancers in the United States. Conventional therapies are unlikely to control advanced stage ovarian cancers, thus requiring innovative alternative therapies. In the current study, we characterized the therapeutic effect of tumor cell-based vaccines combined with the adjuvant, alpha-galactosylceramide (alpha-GalCer) using two different mouse models. Our data suggests that treatment with alpha-GalCer led to an increase in the IFN-gamma serum levels in the presence or absence of irradiated mouse ovarian surface epithelial tumor cells (MOSEC). Furthermore, administration of irradiated MOSEC tumor cells with adjuvant alpha-GalCer generated significant protective and therapeutic antitumor effects against MOSEC tumors in vaccinated C57BL/6 mice. In addition, immune cells expressing CD4, CD8 or NK1.1 markers were found to be important for the protective antitumor effects generated by irradiated tumor cell-based vaccines combined with adjuvant alpha-GalCer. We also found that treatment of a spontaneous ovarian cancer murine model, the Müllerian inhibiting substance type II receptor T antigen (TgMISIIR-TAg) transgenic mice with ovarian tumor cell-based vaccines combined with adjuvant alpha-GalCer led to prolonged survival as well as increased numbers of tumor-specific CD8+ T cells. Therefore, irradiated tumor cell-based vaccines in combination with alpha-GalCer are capable of breaking immune tolerance and generating significant antitumor effects in two different mouse tumor models. Our study serves as a foundation for future clinical translation.


Assuntos
Adjuvantes Imunológicos/farmacologia , Vacinas Anticâncer/imunologia , Vacinas Anticâncer/uso terapêutico , Galactosilceramidas/farmacologia , Neoplasias Ovarianas/imunologia , Neoplasias Ovarianas/prevenção & controle , Animais , Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/efeitos da radiação , Linhagem Celular Tumoral , Sobrevivência Celular , Citocinas/biossíntese , Feminino , Citometria de Fluxo , Interferon gama/sangue , Subpopulações de Linfócitos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Transplante de Neoplasias/imunologia , Análise de Sobrevida , Linfócitos T Citotóxicos/imunologia , Vacinação
6.
J Immunother ; 31(4): 345-58, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18391761

RESUMO

We have previously reported that local tumor irradiation, without inducing cell death, can augment the therapeutic efficacy of intratumoral (IT) dendritic cell (DC) vaccination. This study examined potential mechanisms underlying radiation enhancement of IT DC therapy in this setting. Even though ionizing radiation did not mediate tumor cell killing, bone marrow-derived DCs acquired in vitro tumor antigens from irradiated D5 murine melanoma cells more efficiently than from untreated cells. This radiation-enhanced loading of DCs did not induce DC maturation, but was associated with improved cross-priming of T cells both in vitro and in vivo. Furthermore, in vivo pulsing of DCs with irradiated versus untreated tumor cells resulted in superior presentation of tumor antigens to T cells. In addition, tumor irradiation facilitated homing of IT administered DCs to the draining lymph node, possibly by down-regulating CCL21 expression within the tumor mass. Studies of the tumor microenvironment in irradiated versus untreated tumors did not reveal significant inflammatory changes. Moreover, radiation did not promote accumulation of CD4 or CD8 effector T cells within solid tumors. Our results indicate that, without inducing cytotoxicity, tumor irradiation can enhance the ability of DCs to capture tumor antigens, migrate to the draining lymph node, and present processed antigens to T cells. These findings may prove useful in designing future strategies for human cancer immunotherapy.


Assuntos
Vacinas Anticâncer/efeitos da radiação , Movimento Celular/efeitos da radiação , Células Dendríticas/imunologia , Células Dendríticas/efeitos da radiação , Animais , Apresentação de Antígeno/efeitos da radiação , Antígenos de Neoplasias/imunologia , Antígenos de Neoplasias/metabolismo , Vacinas Anticâncer/uso terapêutico , Quimiocina CCL21/imunologia , Quimiocina CCL21/metabolismo , Apresentação Cruzada/efeitos da radiação , Células Dendríticas/metabolismo , Fluoresceínas , Humanos , Imunoterapia Adotiva , Melanoma Experimental/imunologia , Melanoma Experimental/patologia , Melanoma Experimental/radioterapia , Camundongos , Camundongos Transgênicos , Radiação Ionizante , Succinimidas , Linfócitos T/citologia , Linfócitos T/imunologia , Linfócitos T/metabolismo
7.
Cancer Immunol Immunother ; 55(3): 277-88, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16158275

RESUMO

Allogeneic whole tumour cell vaccines are inherently practical compared with autologous vaccines. Cell lines are derived from allogeneic tumour, grown in bulk and then administered as a vaccine to the patient, following irradiation, which not only prevents any replication but also enhances antigen presentation. Protection is believed to occur through the presentation of antigens shared between the syngeneic and allogeneic tumours. Although cytokine-transfected tumour whole cell vaccines have been used clinically, little data is available comparing the effects of immunomodulatory cytokine-transfection directly on the same cells when used as both an allogeneic and autologous vaccine. To address this, weakly immunogenic B16-F10 (H-2b) murine melanoma was transfected to secrete either GM-CSF, IL-4 or IL-7. Prophylactic vaccination of both syngeneic C57/BL6 (H-2b) (B6) and allogeneic C3H/Hej (H-2k) (C3H) mice showed the effects of transfected cytokine varied between models. Both GM-CSF and IL-7 significantly (P<0.05) increased the levels of protection within syngeneic B6 mice, but had a diminished effect (P>0.05) within C3H allogeneic mice. Allogeneic B16-F10 cells and syngeneic K1735 cells generated CTL against K1735 suggesting cross-reactive immunity. Using cells labeled with fluorescent dye we demonstrate that irradiated vaccines, of either syngeneic or allogeneic origin, appear to generate potent immune responses and fragments of either vaccine remain at the injection site for up to 9 days. This study shows that protection can be enhanced in vivo by using transfected cytokine, but suggests that irradiated whole cell vaccines, of either tissue-type, are rapidly processed. This leads to the conclusion that the cytokine effects are transient and thus transfection with cytokine may be of limited long-term use in situ.


Assuntos
Vacinas Anticâncer/efeitos da radiação , Citocinas/metabolismo , Melanoma Experimental/prevenção & controle , Transplante Homólogo/imunologia , Transplante Isogênico/imunologia , Animais , Transplante de Células , Citotoxicidade Imunológica , Feminino , Citometria de Fluxo , Camundongos , Transplante de Neoplasias , Transfecção , Células Tumorais Cultivadas
8.
Cancer Immunol Immunother ; 55(7): 873-83, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16133106

RESUMO

PURPOSE: We investigated granulocyte-macrophage colony-stimulating factor (GM-CSF) and interleukin-12 (IL-12) infused into the injection site of irradiated tumor vaccine (TV) as therapy for gliomas. METHODS: Rats with subcutaneous RT-2 gliomas were treated with irradiated TV and/or subcutaneous infusion of GM-CSF and/or IL-12 via osmotic minipump 5 days after tumor-cell inoculation. Cytotoxic T lymphocyte (CTL) and natural killer (NK) cell activity were analyzed to investigate immune responses. Rats with intracerebral gliomas were treated with irradiated TV and infused GM-CSF/IL-12 3 days after tumor-cell inoculation. Tumor growth rates and animal survival were followed. Survivors were re-challenged with wild-type RT-2 cells subcutaneously or intracerebrally to study long-term anti-tumor immunity. RESULTS: Rats with subcutaneous gliomas treated with GM-CSF and IL-12 or TV plus GM-CSF or IL-12 did not have increased survival rate (P>0.2), but did have prolonged survival time (P<0.05); in contrast, rats treated with TV plus GM-CSF/IL-12 had increased survival rate (P<0.05) and prolonged survival time (P<0.05) compared with controls. These treatment strategies showed enhanced CTL and NK cell activities. Rats with intra-cerebral gliomas treated with TV plus GM-CSF/IL-12 did not have increased survival rate (P=0.11), but did have prolonged survival time (P<0.0001). Survivors in each group were re-challenged with wild-type RT-2 cells, and all had long-term survival. CONCLUSIONS: Irradiated TV plus continuous localized infusion of GM-CSF/IL-12 may induce a tumor-specific anti-tumor immune response on established subcutaneous or intra-cerebral gliomas, and such a treatment strategy deserves consideration as adjuvant treatment for glioma.


Assuntos
Vacinas Anticâncer/efeitos da radiação , Glioma/terapia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/uso terapêutico , Interleucina-12/uso terapêutico , Animais , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/terapia , Vacinas Anticâncer/uso terapêutico , Ensaios de Seleção de Medicamentos Antitumorais , Glioma/tratamento farmacológico , Fator Estimulador de Colônias de Granulócitos e Macrófagos/administração & dosagem , Bombas de Infusão Implantáveis , Infusões Parenterais , Injeções Intralesionais , Interleucina-12/administração & dosagem , Células Matadoras Naturais/imunologia , Transplante de Neoplasias , Ratos , Ratos Endogâmicos F344 , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/uso terapêutico , Tela Subcutânea , Linfócitos T Citotóxicos/imunologia
9.
J Exp Med ; 202(11): 1507-16, 2005 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-16330814

RESUMO

If irradiated tumor cells could be rendered immunogenic, they would provide a safe, broad, and patient-specific array of antigens for immunotherapies. Prior approaches have emphasized genetic transduction of live tumor cells to express cytokines, costimulators, and surrogate foreign antigens. We asked if immunity could be achieved by delivering irradiated, major histocompatibility complex-negative plasmacytoma cells to maturing mouse dendritic cells (DCs) within lymphoid organs. Tumor cells injected intravenously (i.v.) were captured by splenic DCs, whereas subcutaneous (s.c.) injection led only to weak uptake in lymph node or spleen. The natural killer T (NKT) cells mobilizing glycolipid alpha-galactosyl ceramide, used to mature splenic DCs, served as an effective adjuvant to induce protective immunity. This adjuvant function was mimicked by a combination of poly IC and agonistic alphaCD40 antibody. The adjuvant glycolipid had to be coadministered with tumor cells i.v. rather than s.c. Specific resistance was generated both to a plasmacytoma and lymphoma. The resistance afforded by a single vaccination lasted >2 mo and required both CD4+ and CD8+ T cells. Mature tumor capturing DCs stimulated the differentiation of P1A tumor antigen-specific, CD8+ T cells and uniquely transferred tumor resistance to naive mice. Therefore, the access of dying tumor cells to DCs that are maturing to activated NKT cells efficiently induces long-lived adaptive resistance.


Assuntos
Apresentação de Antígeno/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Células Dendríticas/imunologia , Células Matadoras Naturais/imunologia , Plasmocitoma/imunologia , Adjuvantes Imunológicos/administração & dosagem , Animais , Apresentação de Antígeno/efeitos dos fármacos , Apresentação de Antígeno/efeitos da radiação , Antígenos de Neoplasias/imunologia , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/imunologia , Vacinas Anticâncer/efeitos da radiação , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/imunologia , Feminino , Raios gama , Imunoterapia Ativa/métodos , Linfonodos/imunologia , Ativação Linfocitária/imunologia , Linfoma/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Transplante de Neoplasias , Neoplasias Experimentais , Plasmocitoma/terapia , Baço/imunologia
10.
Cytotherapy ; 7(1): 46-56, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16040383

RESUMO

BACKGROUND: GM-CSF-secreting, allogeneic cell-based cancer vaccines have shown promise for the treatment of a variety of solid tumors. We have now applied this approach to breast cancer. The aim of these studies was to optimize expansion parameters, qualify the manufacturing process, and establish expected outcomes for cGMP-compliant manufacturing of two GM-CSF-secreting breast tumor cell lines. METHODS: The variables affecting the efficiency of expanding and formulating two allogeneic GM-CSF-secreting cell lines, 2T47D-V and 3SKBR3-7, were systematically evaluated. Production criteria investigated included alternative cell culture vessels (flasks vs. cell factories), centrifugation time and speed variables for large volume cell concentration, cell seeding density, the minimal concentration of FBS required for maximal cell expansion, and the dose and timing of irradiation in relation to cryopreservation. RESULTS: These studies demonstrate that, in comparison with standard 150-cm2 tissue culture flasks, Nunc 10-Stack Cell Factories are a more efficient and practical cell culture vessel for vaccine cell line manufacture. Centrifugation optimization studies using the COBE 2991 Cell Processor established that a speed of 2000 r.p.m. (450 g) for 2 min reliably concentrated the cells while maintaining acceptable viability and bioactivity. Radiation studies established that lethal irradiation prior to cryopreservation does not compromise the quality of the product, as measured by post-thaw cell viability and GM-CSF cell line-specific secretion levels. Finally, studies aimed at optimizing the production of one vaccine cell line, 3SKBR3-7, demonstrated that seeding the cells at a higher density and maintaining them in half the initial concentration of FBS maximized the yield of bioactive cells, resulting in significant cost savings. DISCUSSION: A manufacturing process that simultaneously maximizes cell yield, minimizes cell manipulation and maintains vaccine cell potency is critical for producing cell-based cancer vaccines in an academic setting. These studies define a feasible, reproducible and cost-effective methodology for production of a GM-CSF-secreting breast cancer vaccine that is cGMP compliant.


Assuntos
Neoplasias da Mama/terapia , Vacinas Anticâncer/síntese química , Carcinoma/terapia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/síntese química , Centros Médicos Acadêmicos/economia , Centros Médicos Acadêmicos/métodos , Centros Médicos Acadêmicos/normas , Neoplasias da Mama/imunologia , Vacinas Anticâncer/economia , Vacinas Anticâncer/efeitos da radiação , Carcinoma/imunologia , Técnicas de Cultura de Células/economia , Técnicas de Cultura de Células/métodos , Linhagem Celular Tumoral , Análise Custo-Benefício , Criopreservação/métodos , Feminino , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Fator Estimulador de Colônias de Granulócitos e Macrófagos/uso terapêutico , Fidelidade a Diretrizes , Humanos , Laboratórios/economia , Laboratórios/normas , Doses de Radiação , Transplante Homólogo/economia , Transplante Homólogo/imunologia , Transplante Homólogo/métodos
11.
Int J Cancer ; 116(1): 78-86, 2005 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-15756675

RESUMO

Tumor vaccination by the use of gene-modified cancer cells that provide costimulatory signals has been successfully applied in preclinical animal models and is currently evaluated in a variety of clinical settings. In previous work, we demonstrated the efficacy of B7.1/CD80 to promote tumor immunity in syngeneic murine models and to prevent deletion of activated T cells by activation-induced cell death (AICD). In clinical trials, tumor cell vaccines are generally inactivated to avoid transfer of live tumor cells, i.e., additional tumor burden. Previous data indicated, however, that inactivation of tumor cells by lethal ionizing irradiation abrogates tumor vaccination by CD80-expressing cells. Here, we compare living and irradiated allogeneic tumor cells regarding their capacity to induce T-cell effector functions and their propensity to interfere with T-cell deletion by apoptosis. Both lethally irradiated and nonirradiated tumor cells facilitated T-cell proliferation, tumor cell lysis, and interfered with T-cell AICD to a similar extent. In contrast, necrotic tumor cells failed to costimulate T-cell effector functions. Thus, irradiation does not seem to hamper tumor cell-mediated costimulation of T-cell effector functions. In contrast, necrosis of gene-modified tumor cells abrogates costimulation of T cells by CD80-expressing cells.


Assuntos
Antígeno B7-1/metabolismo , Morte Celular , Citotoxicidade Imunológica , Ativação Linfocitária , Linfócitos T/imunologia , Antígeno B7-1/genética , Vacinas Anticâncer/efeitos da radiação , Morte Celular/imunologia , Técnicas de Cocultura , Citotoxicidade Imunológica/efeitos da radiação , Humanos , Ativação Linfocitária/efeitos da radiação , Rabdomiossarcoma/imunologia , Rabdomiossarcoma/metabolismo , Rabdomiossarcoma/patologia , Linfócitos T/efeitos da radiação , Transfecção , Células Tumorais Cultivadas
12.
J Immunol ; 173(10): 6025-32, 2004 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-15528337

RESUMO

Tumor cells engineered to secrete cytokines, referred to as tumor cell vaccines, can often generate systemic antitumor immunity and, in many cases, cause tumor regression. We compared the efficacy of s.c. immunization or intrahepatic immunization of GM-CSF-expressing tumor cell vaccines on the growth of s.c. or orthotopic liver tumors. A chemically transformed hepatic epithelial cell line, GP7TB, derived from Fischer 344 rats, was used to generate tumor models and tumor cell vaccines. Our results demonstrated that two s.c. injections of an irradiated tumor cell vaccine significantly controlled the growth of s.c. tumors, but was completely ineffective against orthotopic liver tumors. Effector cell infiltration in liver tumors was markedly reduced compared with s.c. tumors. Enhanced apoptosis of some effector cells was observed in the liver tumors compared with the s.c. tumors. Furthermore, the T cells induced by s.c. immunization preferentially migrated to s.c. tumor sites, as demonstrated by adoptive transfer experiments. In contrast, intrahepatic immunization, using parental tumor cells admixed with adenoviruses carrying the GM-CSF gene, yielded significantly better therapeutic effects on the liver tumors than on the s.c. tumors. Adoptive transfer experiments further confirmed that the T cells induced by liver immunization preferentially migrated to the liver tumor sites. Our results demonstrate that distinct T cell populations are induced by different immunization routes. Thus, the homing behavior of T cells depends on the route of immunization and is an important factor determining the efficacy of immunotherapy for regional tumors.


Assuntos
Vacinas Anticâncer/administração & dosagem , Citocinas/metabolismo , Inibidores do Crescimento/administração & dosagem , Imunoterapia Adotiva/métodos , Neoplasias Hepáticas Experimentais/metabolismo , Neoplasias Hepáticas Experimentais/patologia , Linfócitos do Interstício Tumoral/imunologia , Adenoviridae/genética , Animais , Apoptose/imunologia , Vacinas Anticâncer/imunologia , Vacinas Anticâncer/efeitos da radiação , Vacinas Anticâncer/uso terapêutico , Contagem de Células , Linhagem Celular Tumoral , Proliferação de Células/efeitos da radiação , Fator Estimulador de Colônias de Granulócitos e Macrófagos/administração & dosagem , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Inibidores do Crescimento/imunologia , Inibidores do Crescimento/efeitos da radiação , Inibidores do Crescimento/uso terapêutico , Injeções Subcutâneas , Fígado/imunologia , Fígado/patologia , Neoplasias Hepáticas Experimentais/imunologia , Neoplasias Hepáticas Experimentais/prevenção & controle , Linfócitos do Interstício Tumoral/patologia , Masculino , Transplante de Neoplasias , Ratos , Ratos Endogâmicos F344 , Pele/imunologia , Pele/patologia , Neoplasias Esplênicas/imunologia , Neoplasias Esplênicas/patologia , Neoplasias Esplênicas/prevenção & controle
13.
J Biomed Sci ; 11(2): 228-38, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-14966373

RESUMO

In preclinical studies, tumor cells genetically engineered to secrete cytokines, hereafter referred to as tumor cell vaccines, can often generate systemic antitumor immunity. This study investigated the therapeutic effects of live or irradiated tumor cell vaccines that secrete granulocyte-macrophage colony-stimulating factor (GM-CSF) on established orthotopic liver tumors. Experimental results indicated that two doses (3 x 10(7) cells per dose) of irradiated tumor cell vaccines were therapeutically ineffective, whereas one dose (3 x 10(6) cells) of live tumor cell vaccines caused complete tumor regression. In vivo depletion of CD8+ T cells, but not natural killer cells, restored tumor formation in the live vaccine-treated animals. Additionally, the treatment of cells with live vaccine induced markedly higher levels of cytotoxic T lymphocyte activity than the irradiated vaccines in the draining lymph nodes. The higher levels of cytokine and antigen loads could partly explain the superior antitumor activity of live tumor cell vaccines, but other unidentified mechanisms could also play a role in the early T cell activation in the lymph nodes. A protocol using multiple and higher dosages of irradiated tumor cell vaccines also caused significant regression of liver tumors. These results suggest that the GM-CSF-secreting tumor cell vaccines are highly promising for orthotopic liver tumors if higher levels of immune responses are elicited during early tumor development.


Assuntos
Vacinas Anticâncer/uso terapêutico , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Neoplasias Hepáticas Experimentais/terapia , Animais , Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/imunologia , Vacinas Anticâncer/efeitos da radiação , Linhagem Celular Transformada , Transplante de Células , Testes Imunológicos de Citotoxicidade , Células Matadoras Naturais/imunologia , Ratos , Ratos Endogâmicos F344 , Linfócitos T Citotóxicos/imunologia , Resultado do Tratamento
14.
J Immunother ; 24(3): 232-6, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11394500

RESUMO

Multiple clinically applicable methods have been used to induce dendritic cells (DCs) to express whole cell tumor antigens, including pulsing DCs with tumor lysate, and mixing DCs with apoptotic or live tumor cells. Herein we demonstrate, using two different tumor systems, that these methods are equipotent inducers of systemic antitumor immunity. Furthermore, tumor lysate pulsed DC vaccines generate more potent antitumor immunity than immunization with irradiated tumor cells plus the classic adjuvant, Corynebacterium parvum.


Assuntos
Antígenos de Neoplasias/administração & dosagem , Vacinas Anticâncer/administração & dosagem , Células Dendríticas/imunologia , Animais , Antígenos de Neoplasias/efeitos da radiação , Vacinas Anticâncer/efeitos da radiação , Feminino , Neoplasias Mamárias Experimentais/imunologia , Neoplasias Mamárias Experimentais/terapia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Propionibacterium acnes/imunologia , Sarcoma Experimental/imunologia , Sarcoma Experimental/terapia
15.
Immunol Lett ; 74(1): 81-6, 2000 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-10996632

RESUMO

Since melanoma is a model immunogenic malignancy incurable in the disseminated phase of its natural course different immunotherapeutic approaches are tested in clinical trials. A number of tumour vaccines genetically modified (GMTV), with various immunostimulatory factors, are tested in phase I/II clinical trials. These factors include cytokines, tumour antigens (TA), costimulatory molecules or HLA antigens. We have designed a novel, mixed auto/allogeneic cellular melanoma vaccine modified with the IL-6 and the sIL-6R genes. Preclinical studies in a mouse model demonstrated that the IL-6/sIL-6R based vaccine is able to elicit efficient anti-tumour responses, mediated by CD8+ and NK cells, which resulted in inhibition of the tumour growth, metastases formation and prolonged survival of the animals treated. Irradiation of vaccine cells does not only lead to their sterilisation but also causes increased secretion of exogenous IL-6 and sIL-6R. Since January 1996 we have vaccinated more than one hundred metastatic melanoma patients. Promising clinical results (22% CR+PR, 32% SD) and the evidence of immune responses in the vaccinated patients have prompted us to design a phase III clinical trial which is to be open in 2000.


Assuntos
Vacinas Anticâncer/uso terapêutico , Ensaios Clínicos como Assunto , Engenharia Genética , Imunoterapia Ativa , Melanoma/terapia , Vacinas Sintéticas/uso terapêutico , Animais , Vacinas Anticâncer/genética , Vacinas Anticâncer/imunologia , Vacinas Anticâncer/efeitos da radiação , Modelos Animais de Doenças , Humanos , Imunoterapia Ativa/métodos , Interleucina-6/genética , Interleucina-6/imunologia , Melanoma/imunologia , Receptores de Interleucina-6/genética , Receptores de Interleucina-6/imunologia , Solubilidade , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Vacinas Sintéticas/efeitos da radiação
16.
Cancer Res ; 60(9): 2444-8, 2000 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-10811122

RESUMO

We have previously shown that antibodies to CTLA-4, an inhibitory receptor on T cells, can be effective at inducing regression of transplantable murine tumors. In this study, we demonstrate that an effective immune response against primary prostate tumors in transgenic (TRAMP) mice can be elicited using a strategy that combines CTLA-4 blockade and an irradiated tumor cell vaccine. Treatment of TRAMP mice at 14 weeks of age resulted in a significant reduction in tumor incidence (15% versus control, 75%), as assessed 2 months after treatment. Histopathological analysis revealed that treated mice had a lower tumor grade with significant accumulation of inflammatory cells in interductal spaces when treated with anti-CTLA-4 and a granulocyte-macrophage colony-stimulating factor-expressing vaccine. Vaccination of nontransgenic mice with this regimen resulted in marked prostatitis accompanied by destruction of epithelium, indicating that the immune response was, at least in part, directed against normal prostate antigens. These findings demonstrate that this combinatorial treatment can elicit a potent antiprostate response and suggest potential of this approach for treatment of prostate cancer.


Assuntos
Adenocarcinoma/terapia , Antígenos de Diferenciação/imunologia , Vacinas Anticâncer/uso terapêutico , Imunização Passiva , Imunoconjugados , Neoplasias da Próstata/terapia , Abatacepte , Animais , Anticorpos Antineoplásicos/farmacologia , Antígenos CD , Antígeno CTLA-4 , Vacinas Anticâncer/efeitos da radiação , Imuno-Histoquímica , Inflamação , Masculino , Camundongos , Camundongos Transgênicos , Neoplasias da Próstata/imunologia
17.
Folia Biol (Praha) ; 46(1): 11-5, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-10730877

RESUMO

It has been found previously that irradiated, IL-2 gene-modified plasmacytoma (X63-m-IL-2) vaccines are more efficient in the therapy of the parental (X63-Ag8.653) plasmacytoma than live plasmacytoma vaccines. In this communication, we have demonstrated that irradiation of murine IL-2-producing plasmacytoma vaccines resulted in upregulation of CD80 molecule expression and IL-2 production. The expression of MHC class I antigens was not altered. The upregulation of the CD80 membrane molecule expression in X63-m-IL-2 cells was higher after irradiation with 150 Gy than after irradiation with 50 Gy. Comparable upregulation of the CD80 molecule expression has also been demonstrated after irradiation of the parental murine X63-Ag8.653 plasmacytoma cells. The results indicate that upregulation of the CD80 molecule expression and enhanced IL-2 production in irradiated X63-m-IL-2 cells was responsible for the higher therapeutic effectiveness of the irradiated plasmacytoma vaccine.


Assuntos
Antígeno B7-1/metabolismo , Vacinas Anticâncer/genética , Vacinas Anticâncer/efeitos da radiação , Interleucina-2/biossíntese , Interleucina-2/genética , Plasmocitoma/terapia , Animais , Vacinas Anticâncer/imunologia , Antígenos de Histocompatibilidade Classe I/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Plasmocitoma/genética , Plasmocitoma/imunologia , Células Tumorais Cultivadas , Regulação para Cima/efeitos da radiação
18.
Hum Gene Ther ; 10(13): 2109-22, 1999 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-10498243

RESUMO

Philadelphia chromosome-positive acute lymphoblastic leukemia (Ph+ ALL) is a highly aggressive malignancy caused by the bcr-abl translocation oncogene. To explore alternative treatments for Ph+ ALL we tested gene-modified cell vaccines in the BALB/c-derived BM185 leukemia model. We compared the efficacy of BM185 cell vaccine expressing CD80 alone or in combination with IL-2 or GM-CSF. Mice injected with viable BM185 leukemia cells modified to express CD80 and GM-CSF (BM185/CD80+GM-CSF) showed the highest leukemia rejection rates. Cell vaccines consisting of irradiated BM185/CD80+GM-CSF cells administered subcutaneously stimulated a potent cytotoxic T lymphocyte (CTL) response against parental BM185. Histological examination of the vaccination site showed a large concentration of immune cells. Administration of the BM185/CD80+GM-CSF cell vaccine before intravenous challenge with parental cells caused strong inhibition of leukemia development. Vaccination after subcutaneous challenge with BM185 cells caused efficient elimination of leukemia promoting 40-60% long-term survival rates. The immunization efficacy of the BM185/CD80+ GM-CSF cell vaccine was directly correlated with the percentage of cells expressing the transgenes. In all, this preclinical study shows that leukemia cell vaccines coexpressing CD80 and GM-CSF can potentially be explored for immunotherapy in Ph+ ALL patients.


Assuntos
Antígeno B7-1/metabolismo , Vacinas Anticâncer/uso terapêutico , Terapia Genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Animais , Células Apresentadoras de Antígenos/imunologia , Vacinas Anticâncer/metabolismo , Vacinas Anticâncer/efeitos da radiação , Linhagem Celular , Citotoxicidade Imunológica , Técnicas de Transferência de Genes , Humanos , Imuno-Histoquímica , Imunoterapia , Interleucina-2/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Microscopia Eletrônica , Transplante de Neoplasias , Leucemia-Linfoma Linfoblástico de Células Precursoras/imunologia , Linfócitos T Citotóxicos/imunologia
19.
Clin Exp Metastasis ; 17(1): 63-70, 1999 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10390149

RESUMO

Interleukin (IL)-12 can activate cytotoxic lymphocytes, stimulate natural killer cell activity, induce the production of INF-gamma and inhibit the development of various experimental tumors. We previously demonstrated that immunotherapy of melanoma bearing mice with an irradiated melanoma vaccine (IMV) coupled with IL-2 or GM-CSF had beneficial effects against primary melanoma growth and against subsequent spontaneous metastasis. We also had found that treatment of melanoma bearing mice with IL-12 (300 ng/day) for 4 weeks inhibited the development of primary melanoma tumors in 40% of mice. The purpose of this study was to investigate the efficacy of combined therapy of experimental melanoma with an IMV prepared from B16F10 melanoma cells coupled with IL-12 treatment. C57BL/6 mice were challenged subcutaneously in the tail with B16F10 melanoma cells and by the 45th day, more than 50% of the mice had developed visible primary melanoma tumors at the injection site. Subsequent immunotherapy of mice with IMV, when coupled with IL-12, provided partial inhibition of primary melanoma tumor growth. Optimal results against primary tumor growth were observed when IMV therapy was coupled with IL-12 at a dose of 50 ng/day. Combination of IMV with IL-12 at a dose of 100 ng/day significantly reduced melanoma metastasis to the lungs compared with control mice, and an improvement in mean survival time was observed in mice treated with a combination of IMV with IL-12 (300 ng/day).


Assuntos
Vacinas Anticâncer/uso terapêutico , Imunoterapia , Interleucina-12/uso terapêutico , Melanoma Experimental/terapia , Animais , Vacinas Anticâncer/efeitos da radiação , Neoplasias Pulmonares/prevenção & controle , Neoplasias Pulmonares/secundário , Camundongos , Camundongos Endogâmicos C57BL , Transplante de Neoplasias , Taxa de Sobrevida
20.
Folia Biol (Praha) ; 45(1): 7-11, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10732712

RESUMO

IL-2 kinetics was assessed in mice vaccinated with irradiated syngeneic tumour vaccines carrying an inserted IL-2 gene and producing constitutively IL-2. For comparison, the kinetics of i.v. administered recombinant IL-2 was also examined. During regular time intervals after the vaccination or administration of recombinant IL-2, samples of serum and peritoneal fluid were collected and examined, using CTLL bioassay or its MTT modification. After i.p. administration of irradiated IL-2-producing plasmacytoma (X63-m-IL-2) vaccine, the levels of IL-2 were substantially higher in the peritoneal fluid than in the serum. Both in the peritoneal fluid and in the serum, the IL-2 level was increasing up to 60 min after administration and then it gradually decreased. The last time point when IL-2 was still detectable both in the peritoneal fluid and in the serum was 30 h. Almost identical results were obtained when the IL-2 levels were detected by the conventional CTLL assay, in which DNA synthesis was monitored by 3H-thymidine labeling, and by the isotope-free MTT modification of the CTLL assay, in which the DNA synthesis was monitored by staining. The MTT modification has the advantage of an isotope-free method. Comparison of two different IL-2-producing vaccines, a murine plasmacytoma X63-m-IL-2, with high IL-2 production, and murine sarcoma MC12-IL-2, with low IL-2 production, revealed that whereas after i.p. administration of the high producers, the peak of IL-2 was reached both in the peritoneal fluid and in the serum after 1 h, the administration of low producers gave the peak level of IL-2 later, 5 h after i.p. administration. Comparison of IL-2 levels obtained after i.p. administration of live and irradiated X63-m-IL-2 vaccine revealed that the irradiated vaccine produced both in vitro and in vivo higher amounts of IL-2. As compared to i.p. administration, the kinetics after i.v. administration of the X63-m-IL-2 vaccine was different. The maximum level of recombinant IL-2 was reached 10 min after administration and IL-2 was undetectable after 5 h. When the injections of recombinant IL-2 were repeated, the elimination of IL-2 from the circulation was substantially faster.


Assuntos
Vacinas Anticâncer , Interleucina-2/genética , Vacinas Sintéticas , Animais , Vacinas Anticâncer/efeitos da radiação , Fibrossarcoma/patologia , Interleucina-2/análise , Interleucina-2/farmacocinética , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Transplante de Neoplasias , Cavidade Peritoneal , Plasmocitoma/patologia , Proteínas Recombinantes de Fusão/farmacocinética , Células Tumorais Cultivadas/efeitos da radiação , Vacinação , Vacinas Sintéticas/efeitos da radiação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...