Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
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
2.
Endocr Relat Cancer ; 12(1): 1-17, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15788636

RESUMO

Optimizing standard treatment modalities for breast cancer has improved the outlook for women afflicted with it, but the fact that 40% still ultimately die from the disease highlights the need for new therapies. Remarkable advances in molecular immunology and biotechnology have created a unique opportunity for developing active vaccination strategies that engage the patient's own immune system in the fight against breast cancer. Early clinical trials have established the safety and bioactivity of some breast cancer vaccine approaches, with a hint of clinical response. They have also highlighted the importance of elucidating the pharmacodynamic interactions between established therapies for breast cancer, such as tamoxifen, aromatase inhibitors, chemotherapy, the HER-2/neu-specific monoclonal antibody trastuzumab (Herceptin), and breast cancer vaccines. Preclinical studies have simultaneously defined the importance of developing targeted approaches for circumventing established immune tolerance to breast cancer during the vaccination process. The first strategies targeting the negative influence of CD4(+)CD25(+)T regulatory cells and the CTLA-4 signaling pathway are just entering clinical testing in combination with tumor vaccines. Developing the most potent approach for activating antitumor immunity while maintaining the efficacy of standard approaches to breast cancer management will ensure that active immunotherapy is successfully integrated into the standard of care.


Assuntos
Neoplasias da Mama/imunologia , Neoplasias da Mama/terapia , Vacinas Anticâncer , Imunidade/fisiologia , Antineoplásicos/uso terapêutico , Feminino , Humanos , Imunoterapia
3.
Cancer J ; 7(4): 324-37, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11561608

RESUMO

Pancreatic cancer remains one of the most difficult cancers to treat; very few effective therapies are available, with surgery being the sole chance for cure-yet surgery is not a viable option for most pancreatic cancer patients. Immunotherapy has the potential to provide a non-cross-resistant mechanism of antitumor activity that can be integrated with surgery, radiation therapy, and chemotherapy. However, the inherent instability of the tumorgenome as well as tumor tolerance mechanisms are significant practical obstacles that must be overcome if immune-based approaches for pancreatic cancer can achieve its promise. Recent advances in both tumor immunology and vaccine design have already resulted in promising preliminary data from phase I studies, and additional trials are already in progress. This article summarizes some of the progress and challenges in immunotherapy research.


Assuntos
Imunoterapia/métodos , Neoplasias Pancreáticas/terapia , Antígeno B7-1 , Vacinas Anticâncer/imunologia , Vacinas Anticâncer/uso terapêutico , Previsões , Humanos , Tolerância Imunológica , Imunidade Celular , Imunoterapia/classificação , Pâncreas/imunologia , Neoplasias Pancreáticas/imunologia , Membro 7 da Superfamília de Receptores de Fatores de Necrose Tumoral
4.
Curr Opin Investig Drugs ; 2(1): 133-5, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11527005

RESUMO

Recent advances in our understanding of immune function with regard to the generation of a potent antitumor response have resulted in a renewed interest in cancer vaccines and point to a role for immunotherapy in the treatment of cancer. Currently, the majority of vaccine strategies for the treatment of solid malignancies focus on the generation of cytotoxic T lymphocytes (CTL) that destroy tumor cells that express a given target protein, or antigen. However, antibody therapies have already been successful against some cancers. Current humoral immunotherapy typically involves the passive infusion of monoclonal antibodies, which usually target a specific tumor-encoded antigen. However, vaccines can be engineered to induce humoral immunity. By focusing on the cellular arm of the immune response at the expense of humoral immunity (or the converse), we may have inadvertently limited the potential efficacy of our anticancer vaccines. This article seeks to explore the notion that a vaccine designed to optimally activate both arms of the immune system may well generate an antitumor immune response greater than the sum of the two individual effector mechanisms alone.


Assuntos
Neoplasias/imunologia , Anticorpos Monoclonais/uso terapêutico , Formação de Anticorpos , Apresentação de Antígeno , Linfócitos B/imunologia , Vacinas Anticâncer/imunologia , Humanos , Imunidade Celular , Neoplasias/terapia , Linfócitos T/imunologia
5.
Gene Ther ; 8(13): 969-76, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11438831

RESUMO

In this study we analyzed two ways of retargeting of Ad-vectors to human pancreatic carcinoma with the aim of enhancing the gene transfer efficiency. First, we analyzed the expression of the epidermal growth factor receptor (EGFR) on primary, as well as established pancreatic carcinoma cells by flow cytometry which revealed high expression levels of EGFR on the surface of these cells. We showed that EGFR-retargeted entry pathway using a bispecific fusion protein formed by a recombinant soluble form of truncated Coxsackie and Adenovirus Receptor (sCAR) genetically fused with human EGF (sCAR-EGF) redirects them to the EGFR leading to an enhanced gene transfer efficiency to pancreatic carcinoma cells. Since flow cytometry revealed absence of CAR expression, but the presence of at least one of both alphav integrins on the pancreatic carcinoma cells, a second way of targeting was investigated using a genetically modified Ad vector which has an RGD (Arg-Gly-Asp)-containing peptide inserted into the HI-loop of the fiber knob. This RGD targeted Ad (AdlucRGD) revealed efficient CAR-independent infection by allowing binding to cellular integrins resulting in a dramatic enhancement of gene transfer. These findings have direct relevance for Ad-vector based gene therapy strategies for pancreatic carcinoma.


Assuntos
Receptores ErbB/metabolismo , Marcação de Genes/métodos , Técnicas de Transferência de Genes , Terapia Genética/métodos , Neoplasias Pancreáticas/metabolismo , Adenoviridae/genética , Vetores Genéticos , Humanos , Integrinas/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias Pancreáticas/patologia , Neoplasias Pancreáticas/virologia , Receptores de Vitronectina/metabolismo , Células Tumorais Cultivadas
6.
Cancer Res ; 61(11): 4320-4, 2001 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-11389052

RESUMO

Serial analysis of gene expression (SAGE) can be used to quantify gene expression in human tissues. Comparison of gene expression levels in neoplastic tissues with those seen in nonneoplastic tissues can, in turn, identify novel tumor markers. Such markers are urgently needed for highly lethal cancers like pancreatic adenocarcinoma, which typically presents at an incurable, advanced stage. The results of SAGE analyses of a large number of neoplastic and nonneoplastic tissues are now available online, facilitating the rapid identification of novel tumor markers. We searched an online SAGE database to identify genes preferentially expressed in pancreatic cancers as compared with normal tissues. SAGE libraries derived from pancreatic adenocarcinomas were compared with SAGE libraries derived from nonneoplastic tissues. Three promising tags were identified. Two of these tags corresponded to genes (lipocalin and trefoil factor 2) previously shown to be overexpressed in pancreatic carcinoma, whereas the third tag corresponded to prostate stem cell antigen (PSCA), a recently discovered gene thought to be largely restricted to prostatic basal cells and prostatic adenocarcinomas. PSCA was expressed in four of the six pancreatic cancer SAGE libraries, but not in the libraries derived from normal pancreatic ductal cells. We confirmed the overexpression of the PSCA mRNA transcript in 14 of 19 pancreatic cancer cell lines by reverse transcription-PCR, and using immunohistochemistry, we demonstrated PSCA protein overexpression in 36 of 60 (60%) primary pancreatic adenocarcinomas. In 59 of 60 cases, the adjacent nonneoplastic pancreas did not label for PSCA. PSCA is a novel tumor marker for pancreatic carcinoma that has potential diagnostic and therapeutic implications. These results establish the validity of analyses of SAGE databases to identify novel tumor markers.


Assuntos
Biomarcadores Tumorais/biossíntese , Glicoproteínas de Membrana/biossíntese , Proteínas de Neoplasias/biossíntese , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/imunologia , Adenocarcinoma/genética , Adenocarcinoma/imunologia , Antígenos de Neoplasias , Biomarcadores Tumorais/genética , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/imunologia , Proteínas Ligadas por GPI , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Imuno-Histoquímica , Glicoproteínas de Membrana/genética , Proteínas de Neoplasias/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator Trefoil-2 , Células Tumorais Cultivadas
7.
Cancer Res ; 61(9): 3689-97, 2001 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-11325840

RESUMO

Tumor-specific immune tolerance limits the effectiveness of cancer vaccines. In addition, tumor vaccines alone have a limited potential for the treatment of measurable tumor burdens. This highlights the importance of identifying more potent cancer vaccine strategies for clinical testing. We tested immune-modulating doses of chemotherapy in combination with a granulocyte/macrophage-colony stimulating factor (GM-CSF)-secreting, HER-2/neu (neu)-expressing whole-cell vaccine as a means to treat existing mammary tumors in antigen-specific tolerized neu transgenic mice. Earlier studies have shown that neu transgenic mice exhibit immune tolerance to the neu-expressing tumors similar to what is observed in patients with cancer. We found that cyclophosphamide, paclitaxel, and doxorubicin, when given in a defined sequence with a GM-CSF-secreting, neu-expressing whole-cell vaccine, enhanced the vaccine's potential to delay tumor growth in neu transgenic mice. In addition, we showed that these drugs mediate their effects by enhancing the efficacy of the vaccine rather than via a direct cytolytic effect on cancer cells. Furthermore, paclitaxel and cyclophosphamide appear to amplify the T helper 1 neu-specific T-cell response. These findings suggest that the combined treatment with immune-modulating doses of chemotherapy and the GM-CSF-secreting neu vaccine can overcome immune tolerance and induce an antigen-specific antitumor immune response. These data provide the immunological rationale for testing immune-modulating doses of chemotherapy in combination with tumor vaccines in patients with cancer.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Vacinas Anticâncer/imunologia , Genes erbB-2/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Tolerância Imunológica/imunologia , Células 3T3 , Animais , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/genética , Terapia Combinada , Ciclofosfamida/administração & dosagem , Doxorrubicina/administração & dosagem , Esquema de Medicação , Sinergismo Farmacológico , Epitopos de Linfócito T/imunologia , Feminino , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Humanos , Tolerância Imunológica/genética , Ativação Linfocitária/imunologia , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/imunologia , Neoplasias Mamárias Experimentais/terapia , Camundongos , Camundongos Transgênicos , Paclitaxel/administração & dosagem , Ratos , Linfócitos T/imunologia , Células Th1/imunologia
8.
Curr Opin Mol Ther ; 3(1): 77-84, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11249735

RESUMO

Cancer vaccines are on the threshold of taking their place alongside the more traditional cancer treatment modalities of surgery, radiation therapy and chemotherapy. The toxicology and immunopharmacology of therapeutic cancer vaccines, particularly those that secrete granulocyte macrophage colony stimulating factor (GM-CSF), are currently under active clinical investigation. Interestingly, drugs traditionally used for tumor cytoreduction can have both positive and negative effects on host immunity. Exploration of the potential pharmacodynamic interactions of antineoplastic drugs with GM-CSF-secreting vaccines has revealed that low doses of some chemotherapeutics can augment the antitumor immunity induced by GM-CSF-secreting vaccines. These interactions will require thorough preclinical evaluation to maximize the clinical impact of this type of therapeutic cancer vaccine.


Assuntos
Antineoplásicos/administração & dosagem , Vacinas Anticâncer/administração & dosagem , Neoplasias/tratamento farmacológico , Neoplasias/terapia , Adjuvantes Imunológicos/administração & dosagem , Adjuvantes Imunológicos/efeitos adversos , Animais , Antineoplásicos/efeitos adversos , Vacinas Anticâncer/efeitos adversos , Terapia Combinada , Ciclofosfamida/administração & dosagem , Ciclofosfamida/efeitos adversos , Doxorrubicina/administração & dosagem , Doxorrubicina/efeitos adversos , Fator Estimulador de Colônias de Granulócitos e Macrófagos/efeitos adversos , Fator Estimulador de Colônias de Granulócitos e Macrófagos/uso terapêutico , Humanos , Tolerância Imunológica , Modelos Biológicos , Paclitaxel/administração & dosagem , Paclitaxel/efeitos adversos
9.
Cancer Res ; 61(3): 880-3, 2001 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-11221874

RESUMO

HER-2/neu (neu) transgenic mice (neu-N mice), which express the nontransforming rat proto-oncogene, demonstrate immunological tolerance to neu that is similar to what is encountered in patients with neu-expressing breast cancer. We have shown previously that a significant increase in neu-specific T cells, but no induction of neu-specific antibody, is seen after neu-specific vaccination in neu-N mice. In contrast, a significant induction of both neu-specific T-cell and antibody responses is found in nontoleragenic FVB/N mice after vaccination. These mice are fully protected from a s.c. challenge with NT cells, a mammary tumor cell line derived from a spontaneous tumor that arose in a neu-N mouse, whereas neu-N mice are not. In this study, we demonstrate that CD4+ T cell-depleted FVB/N mice show no induction of neu-specific IgG after vaccination and are unable to reject an NT challenge (0 of 10 mice were tumor free). Conversely, the depletion of natural killer cells has no effect on vaccine-mediated tumor rejection (100% of mice were tumor free). In CD8+ T cell-depleted animals, where vaccine-induced neu-specific IgG titers were normal, NT growth was delayed, but only 10% of mice remained tumor free, demonstrating that neu-specific IgG alone is insufficient for protection from NT challenge. To directly assess the necessity for the combination of neu-specific cellular and humoral immune responses, severe combined immunodeficient mice were given an adoptive transfer of CTLs plus IgG derived from FVB/N mice. Animals that were given CTLs that recognized an irrelevant antigen plus neu-specific IgG developed tumors at a rate similar to CD8+ T cell-depleted FVB/N mice. Animals receiving an adoptive transfer of neu-specific CTLs plus control IgG derived from naive FVB/N mice were only partially protected from NT challenge (50% of animals were tumor free). However, only animals receiving the combination of neu-specific CTLs and neu-specific IgG were fully protected from NT challenge (100% of animals were tumor free). These studies specifically define the immunological requirements for the eradication of neu-expressing tumors in this model system, demonstrating that both cellular and humoral neu-specific responses are necessary for protection from an NT challenge. These data suggest that vaccines optimized to induce maximal T- and B-cell immunity to neu, and possibly to similar putative tumor-rejection antigens, may lead to more potent in vivo antitumor immunity.


Assuntos
Linfócitos B/imunologia , Neoplasias Mamárias Experimentais/imunologia , Receptor ErbB-2/imunologia , Linfócitos T/imunologia , Vacinação , Células 3T3/imunologia , Células 3T3/metabolismo , Animais , Feminino , Tolerância Imunológica , Imunoglobulina G/sangue , Imunoglobulina G/imunologia , Imunoterapia Adotiva , Ativação Linfocitária/imunologia , Neoplasias Mamárias Experimentais/metabolismo , Neoplasias Mamárias Experimentais/prevenção & controle , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID , Transplante de Neoplasias , Receptor ErbB-2/biossíntese , Subpopulações de Linfócitos T/imunologia
10.
J Clin Oncol ; 19(1): 145-56, 2001 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-11134207

RESUMO

PURPOSE: Allogeneic granulocyte-macrophage colony-stimulating factor (GM-CSF)-secreting tumor vaccines can cure established tumors in the mouse, but their efficacy against human tumors is uncertain. We have developed a novel GM-CSF-secreting pancreatic tumor vaccine. To determine its safety and ability to induce antitumor immune responses, we conducted a phase I trial in patients with surgically resected adenocarcinoma of the pancreas. PATIENTS AND METHODS: Fourteen patients with stage 1, 2, or 3 pancreatic adenocarcinoma were enrolled. Eight weeks after pancreaticoduodenectomy, three patients received 1 x 10(7) vaccine cells, three patients received 5 x 10(7) vaccine cells, three patients received 10 x 10(7) vaccine cells, and five patients received 50 x 10(7) vaccine cells. Twelve of 14 patients then went on to receive a 6-month course of adjuvant radiation and chemotherapy. One month after completing adjuvant treatment, six patients still in remission received up to three additional monthly vaccinations with the same vaccine dose that they had received originally. RESULTS: No dose-limiting toxicities were encountered. Vaccination induced increased delayed-type hypersensitivity (DTH) responses to autologous tumor cells in three patients who had received >or= 10 x 10(7) vaccine cells. These three patients also seemed to have had an increased disease-free survival time, remaining disease-free at least 25 months after diagnosis. CONCLUSION: Allogeneic GM-CSF-secreting tumor vaccines are safe in patients with pancreatic adenocarcinoma. This vaccine approach seems to induce dose-dependent systemic antitumor immunity as measured by increased postvaccination DTH responses against autologous tumors. Further clinical evaluation of this approach in patients with pancreatic cancer is warranted.


Assuntos
Adenocarcinoma/terapia , Vacinas Anticâncer/uso terapêutico , Fator Estimulador de Colônias de Granulócitos e Macrófagos/uso terapêutico , Neoplasias Pancreáticas/terapia , Adenocarcinoma/imunologia , Adenocarcinoma/patologia , Idoso , Vacinas Anticâncer/efeitos adversos , Vacinas Anticâncer/farmacocinética , Terapia Combinada , Qualidade de Produtos para o Consumidor , Intervalo Livre de Doença , Relação Dose-Resposta Imunológica , Feminino , Fator Estimulador de Colônias de Granulócitos e Macrófagos/efeitos adversos , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacocinética , Humanos , Hipersensibilidade Tardia/patologia , Masculino , Pessoa de Meia-Idade , Neoplasias Pancreáticas/imunologia , Neoplasias Pancreáticas/patologia
11.
Curr Protoc Hum Genet ; Chapter 13: Unit 13.8, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-18428256

RESUMO

This unit describes the use of retroviral vectors that can be successfully employed for gene transfer into both primary tumor cultures and established cell lines. The unit includes procedures for assaying the stability of the vaccine following gene transfer. Techniques for maintaining the retroviral producer lines and titering the retroviral vectors are also described. A protocol for frozen storage of the vaccine (transduced tumor cells) is provided. In addition, methods are described for characterizing the vaccine cells following gene transfer. Directions for testing the expression of the transferred gene in the transfected tumor line are also given. The final protocol provides suggestions for designing in vivo animal experiments and discusses what has to be observed in a clinical setting.


Assuntos
Vacinas Anticâncer/genética , Neoplasias/terapia , Animais , Citocinas/biossíntese , Citocinas/genética , Técnicas de Transferência de Genes , Vetores Genéticos , Genética Médica , Humanos , Camundongos , Neoplasias/genética , Neoplasias/imunologia , Retroviridae/genética , Linfócitos T/imunologia , Células Tumorais Cultivadas
12.
Int J Radiat Oncol Biol Phys ; 48(4): 1089-96, 2000 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-11072167

RESUMO

PURPOSE: (1) To determine the toxicity of an intensified postoperative adjuvant regimen for periampullary adenocarcinoma (pancreatic and nonpancreatic) utilizing concurrent 5-fluorouracil (5-FU), leucovorin (LV), dipyridamole (DPM), and mitomycin-C (MMC) combined with split-course locoregional external beam radiotherapy (EBRT) to 50 Gy. This was followed by 4 cycles of the same chemotherapy as adjuvant therapy. (2) To determine preliminary estimates of the overall and disease-free survival associated with the use of this regimen. (3) To compare the toxicities and early survival results of patients treated with the current regimen to those of patients who completed our prior trial of concurrent chemoradiation infusion with 5-FU/LV chemotherapy and regional nodal and prophylactic hepatic irradiation. METHODS: Postpancreaticoduodenectomy, patients received every 4 weeks bolus administration of 5-FU, (400 mg/m(2)), and LV, (20 mg/m(2), Days l-3), DPM (75 mg p.o., 4 times per day, Days 0-3, and every 8 weeks), MMC, (10 mg/m(2); maximum of 20 mg, Day l during EBRT). This was followed by 4 months of the same chemotherapy, beginning 1 month following the completion of EBRT. EBRT consisted of split-course 5000 cGy/20 fractions with a 2-week planned rest after the first 10 fractions (2500 cGy). RESULTS: From 4/96 to 6/99, 45 patients were enrolled and treated. Their experience constitutes the basis of this analysis. There were 29 patients with pancreatic cancer and 16 with nonpancreatic periampullary cancer. Seventeen patients had tumors of 3 cm or more, and 39 patients had at least 1 histologically involved lymph node. Thirteen patients had a histologically positive margin of resection. The mean time to start of treatment was 63 days following surgery. During chemoradiation therapy there were no Grade 3 or worse nonhematologic toxicities and 47% Grade 3 or Grade 4 hematologic toxicities of short duration. Following chemoradiation, during chemotherapy treatment only, there was one Grade 3 hepatic and one Grade 3 pulmonary toxicity which was nondebilitating (2% each case) and 42% Grade 3 or 4 hematologic toxicity. There were 2 episodes of neutropenic fever requiring admission and no treatment-related mortalities. One patient developed a mild case of HUS, which responded to standard management. One patient developed persistent shortness of breath (nondebilitating), and another patient had occasional dyspnea on exertion, both occurring after all therapy. The majority of patients complained of increased fatigue (Grade 1-2), greatest during the combined therapy and improving post all treatment. As of 6/23/99, 20 of 45 patients have relapsed, 13 in the liver. Twelve patients have died. Median follow-up for surviving patients is 14.3 months. Disease-free survival at 12 months following surgery is 66% (as compared to 25% in our prior study), and the median disease-free survival is 17 months (as compared to 8. 3 months in our prior study). Median survival has not yet been reached, but will be greater than 17 months. CONCLUSION: With a 14.3-month median follow-up, acute toxicity has been acceptable and manageable. Observed relapses were seen 9-13 months following surgical resection. Early survival analysis suggests a trend toward increased median disease-free survival (8.3 vs. 17 months), especially for patients with nonpancreatic periampullary adenocarcinoma.


Assuntos
Adenocarcinoma/terapia , Neoplasias Pancreáticas/terapia , Adenocarcinoma/mortalidade , Adenocarcinoma/patologia , Adulto , Idoso , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Quimioterapia Adjuvante , Dipiridamol/administração & dosagem , Dipiridamol/efeitos adversos , Intervalo Livre de Doença , Neoplasias Duodenais/mortalidade , Neoplasias Duodenais/patologia , Neoplasias Duodenais/terapia , Feminino , Fluoruracila/administração & dosagem , Fluoruracila/efeitos adversos , Seguimentos , Humanos , Leucovorina/administração & dosagem , Leucovorina/efeitos adversos , Masculino , Pessoa de Meia-Idade , Mitomicina/administração & dosagem , Mitomicina/efeitos adversos , Recidiva Local de Neoplasia , Neoplasias Pancreáticas/mortalidade , Neoplasias Pancreáticas/patologia , Pancreaticoduodenectomia , Dosagem Radioterapêutica , Radioterapia Adjuvante , Fatores de Tempo
13.
Immunity ; 13(4): 529-38, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11070171

RESUMO

T cell responsiveness to an epitope is affected both by its affinity for the presenting MHC molecule and the affinity of the MHC-peptide complex for TCR. One limitation of cancer immunotherapy is that natural tumor antigens elicit relatively weak T cell responses, in part because high-affinity T cells are rendered tolerant to these antigens. We report here that amino acid substitutions in a natural MHC class I-restricted tumor antigen that increase the stability of the MHC-peptide-TCR complex are significantly more potent as tumor vaccines. The improved immunity results from enhanced in vivo expansion of T cells specific for the natural tumor epitope. These results indicate peptides that stabilize the MHC-peptide-TCR complex may provide superior antitumor immunity through enhanced stimulation of specific T cells.


Assuntos
Antígenos de Neoplasias/imunologia , Neoplasias Colorretais/imunologia , Epitopos de Linfócito T/imunologia , Antígenos H-2/metabolismo , Oligopeptídeos/imunologia , Oligopeptídeos/metabolismo , Complexo Receptor-CD3 de Antígeno de Linfócitos T/metabolismo , Alanina/metabolismo , Substituição de Aminoácidos , Animais , Antígenos de Neoplasias/metabolismo , Células Clonais , Citotoxicidade Imunológica/imunologia , Epitopos de Linfócito T/metabolismo , Feminino , Antígeno de Histocompatibilidade H-2D , Ligantes , Camundongos , Camundongos Endogâmicos BALB C , Oligopeptídeos/síntese química , Ressonância de Plasmônio de Superfície , Subpopulações de Linfócitos T/imunologia
14.
J Immunother ; 23(4): 438-48, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-10916753

RESUMO

To explore the potential efficacy of local cytokine delivery against tumors in the central nervous system (CNS), C57BL6 mice were simultaneously given intracranial injections of tumor challenge and of irradiated B16F10 melanoma cells transduced to secrete interleukin-2 (IL-2). Intracranial IL-2 therapy generated antitumor responses capable of extending the survival of animals that received simultaneous intracranial tumor challenge either locally or at distant sites in the brain. Nontransduced melanoma cells had little effect. Animals that survived intracranial IL-2 therapy and tumor challenge showed prolonged survival compared with controls when challenged with a second tumor dose 70 days after initial treatment. In addition, animals that rejected intracranial tumors were also protected from tumor growth upon rechallenge at sites outside the CNS (i.e., subcutaneous tumor challenge). Conversely, identical or 10-fold larger doses of IL-2-transduced cells administered by subcutaneous injection failed to generate protection against intracranial tumor challenges. Elimination of T-cell and natural killer (NK) subsets using gene knockout mice and antibody-depletion techniques demonstrated that NK cells were most important for the initial antitumor response, whereas CD4+ T-cells were not necessary. These studies demonstrate that local IL-2 therapy in the brain not only generates an immediate local antitumor immune response, but also establishes long-term immunologic memory capable of eliminating subsequent tumor challenges within and outside of the CNS. Furthermore, the antitumor response to paracrine IL-2 in the brain differed significantly from that in the flank, suggesting that the intrinsic CNS cells involved in initiating immunity within the brain have different cytokine requirements from their peripheral counterparts.


Assuntos
Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/terapia , Interleucina-2/administração & dosagem , Interleucina-2/imunologia , Melanoma Experimental/imunologia , Melanoma Experimental/terapia , Adjuvantes Imunológicos/administração & dosagem , Adjuvantes Imunológicos/genética , Animais , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Antígenos CD4/genética , Antígenos CD4/imunologia , Vacinas Anticâncer/imunologia , Vacinas Anticâncer/uso terapêutico , Feminino , Memória Imunológica , Injeções Intralesionais , Injeções Subcutâneas , Interleucina-2/genética , Células Matadoras Naturais/imunologia , Depleção Linfocítica , Melanoma Experimental/genética , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Comunicação Parácrina , Neoplasias Cutâneas/imunologia , Neoplasias Cutâneas/terapia , Subpopulações de Linfócitos T/imunologia , Transdução Genética
15.
Cancer Res ; 60(13): 3569-76, 2000 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-10910070

RESUMO

HER-2/neu (neu-N) transgenic mice, which express the nontransforming rat proto-oncogene, develop spontaneous focal mammary adenocarcinomas beginning at 5-6 months of age. The development and histology of these tumors bears a striking resemblance to what is seen in patients with breast cancer. We have characterized the immunological responses to HER-2/neu (neu) in this animal model. neu-positive tumor lines, which were derived from spontaneous tumors that formed in neu-N animals, are highly immunogenic in parental, FVB/N mice. In contrast, a 100-fold lower tumor challenge is sufficient for growth in 100% of transgenic animals. Despite significant tolerance to the transgene, neu-specific immune responses similar to those observed in breast cancer patients can be demonstrated in neu-N mice prior to vaccination. Both cellular and humoral neu-specific responses in transgenic mice can be boosted with neu-specific vaccination, although to a significantly lesser degree than what is observed in FVB/N mice, indicating that the T cells involved are less responsive than in the nontoleragenic parental strain. Using irradiated whole-cell and recombinant vaccinia virus vaccinations we are able to protect neu-N mice from a neu-expressing tumor challenge. T-cell depletion experiments demonstrated that the observed protection is T cell dependent. The vaccine-dependent neu-specific immune response is also sufficient to delay the onset of spontaneous tumor formation in these mice. These data suggest that, despite tolerance to neu in this transgenic model, it is possible to immunize neu-specific T cells to achieve neu-specific tumor rejection in vivo. These transgenic mice provide a spontaneous tumor model for identifying vaccine approaches potent enough to overcome mechanisms of immune tolerance that are likely to exist in patients with cancer.


Assuntos
Adenocarcinoma/genética , Adenocarcinoma/imunologia , Vacinas Anticâncer , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/imunologia , Receptor ErbB-2/imunologia , Linfócitos T/imunologia , Células 3T3 , Adenocarcinoma/terapia , Animais , Feminino , Genes erbB-2 , Tolerância Imunológica , Neoplasias Mamárias Experimentais/terapia , Camundongos , Camundongos Endogâmicos , Camundongos Transgênicos , Ratos , Receptor ErbB-2/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Timo/imunologia
16.
Oncology (Williston Park) ; 14(2): 245-56; discussion 259-60, 265, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10736811

RESUMO

Although surgery remains the only curative option for patients with gastrointestinal (GI) malignancies, the use of adjuvant chemotherapy and/or localized radiation is considered standard therapy for patients who present with locoregional disease. Even with adjuvant therapy, however, the 5-year survival rate for such patients ranges from 2% to 50%, depending on the specific cancer type and stage. As a result, more effective interventions are necessary for all but the earliest stages of GI malignancies. Colon cancer represents the paradigm for the management of GI malignancies, not only because it is, by far, the most common cancer in this group, but also because the biological progression to disease is well characterized. Immunotherapy is an alternative approach for treating GI malignancies that can either: (1) activate tumor-specific T-cells; or (2) use monoclonal antibodies derived from tumor-specific antigens. Monoclonal antibodies act by a mechanism that is distinct from that of chemotherapy and, thus, represent a non-cross-resistant treatment with an entirely different spectrum of toxicities. Thanks to an improved understanding of tumor immunology, as well as the events needed to generate an optimal immune response, the possibility of designing an effective colon cancer vaccine approach that induces both humoral and cellular responses has become even more realistic.


Assuntos
Vacinas Anticâncer/uso terapêutico , Neoplasias Gastrointestinais/terapia , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/uso terapêutico , Antígenos de Neoplasias/imunologia , Vacina BCG/uso terapêutico , Neoplasias Gastrointestinais/imunologia , Humanos , Imunoterapia , Linfócitos T/imunologia
19.
J Clin Oncol ; 17(3): 1047-60, 1999 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10071300

RESUMO

It has been more than 100 years since the first reported attempts to activate a patient's immune system to eradicate developing cancers. Although a few of the subsequent vaccine studies demonstrated clinically significant treatment effects, active immunotherapy has not yet become an established cancer treatment modality. Two recent advances have allowed the design of more specific cancer vaccine approaches: improved molecular biology techniques and a greater understanding of the mechanisms involved in the activation of T cells. These advances have resulted in improved systemic antitumor immune responses in animal models. Because most tumor antigens recognized by T cells are still not known, the tumor cell itself is the best source of immunizing antigens. For this reason, most vaccine approaches currently being tested in the clinics use whole cancer cells that have been genetically modified to express genes that are now known to be critical mediators of immune system activation. In the future, the molecular definition of tumor-specific antigens that are recognized by activated T cells will allow the development of targeted antigen-specific vaccines for the treatment of patients with cancer.


Assuntos
Antígenos de Neoplasias/imunologia , Vacinas Anticâncer/imunologia , Linfócitos T/imunologia , Animais , Células Apresentadoras de Antígenos/imunologia , Antígenos de Neoplasias/genética , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/genética , Ensaios Clínicos como Assunto , Antígenos de Histocompatibilidade Classe II/imunologia , Humanos , Ativação Linfocitária , Transfecção
20.
Ann N Y Acad Sci ; 886: 67-72, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10667204

RESUMO

The goal of cancer treatment is to develop modalities that specifically target tumor cells, thereby avoiding unnecessary side effects to normal tissue. Vaccine strategies that result in the activation of the immune system specifically against proteins expressed by a cancer have the potential to be effective treatment for this purpose. An early vaccine approach that was developed by our group involves the insertion of the granulocyte-macrophage colony stimulating factor (GM-CSF) gene into cancer cells that are then used to immunize patients. These genetically modified tumor cells produce the immune activating protein GM-CSF in the local environment of the tumor cells, specifically activating the patient's T cells to eradicate cancer at metastatic sites. We have performed many studies that demonstrate that this vaccine can cure mice of cancer. We recently demonstrated that this approach can activate an immune response in patients with renal cell carcinoma. We are currently testing a similar approach in patients with pancreatic cancer. Until recently, whole tumor cells were used to produce the vaccine because the proteins expressed by the tumor cells that can be recognized by the immune system were unknown. However, recent advances have allowed the identification of many of the proteins expressed by some cancers. In addition, significant attention has been focused on the mechanisms by which antitumor immunity can be modulated. These active areas of research will undoubtably lead to the development of more specific and more potent vaccine strategies in the near future. The first part of this paper focuses on data from two recent clinical trials that evaluated the whole tumor cell approach. The second part of this paper discusses some of the more exciting antigen-specific vaccine approaches that are under development for the treatment of cancer.


Assuntos
Imunoterapia , Neoplasias/terapia , Animais , Vacinas Anticâncer/imunologia , Vacinas Anticâncer/uso terapêutico , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Humanos , Camundongos , Linfócitos T/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...